WO2004000997A2 - Therapeutic polypeptides, nucleic acids encoding same, and methods of use - Google Patents

Therapeutic polypeptides, nucleic acids encoding same, and methods of use Download PDF

Info

Publication number
WO2004000997A2
WO2004000997A2 PCT/US2003/017512 US0317512W WO2004000997A2 WO 2004000997 A2 WO2004000997 A2 WO 2004000997A2 US 0317512 W US0317512 W US 0317512W WO 2004000997 A2 WO2004000997 A2 WO 2004000997A2
Authority
WO
WIPO (PCT)
Prior art keywords
nucleic acid
polypeptide
novx
seq
amino acid
Prior art date
Application number
PCT/US2003/017512
Other languages
French (fr)
Other versions
WO2004000997A3 (en
Inventor
David W. Anderson
Ferenc L. Boldog
Catherine E. Burgess
Stacie J. Casman
Shlomit R. Edinger
Andrew Eisen
Karen Ellerman
Valerie L. Gerlach
Linda Gorman
Xiaojia Guo
Vladimir Y. Gusev
Weizhen Ji
Li Li
John R. Macdougall
Uriel M. Malyankar
Isabelle Millet
Tatiana Ort
Muralidhara Padigaru
Sudhirdas K. Prayaga
Meera Patturajan
Carol E. A. Pena
John A. Peyman
Daniel K. Rieger
Mark E. Rothenberg
Paul Sciore
Suresh G. Shenoy
Glennda Smithson
Kimberly A. Spytek
David J. Stone
Raymond J. Taupier, Jr.
Velizar T. Tchernev
Corine A. M. Vernet
Edward Z. Voss
Bryan D. Zerhusen
Mei Zhong
Original Assignee
Curagen Corporation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US10/454,246 external-priority patent/US20050053930A1/en
Application filed by Curagen Corporation filed Critical Curagen Corporation
Priority to AU2003272200A priority Critical patent/AU2003272200A1/en
Priority to EP03754372A priority patent/EP1572948A2/en
Priority to CA002486490A priority patent/CA2486490A1/en
Publication of WO2004000997A2 publication Critical patent/WO2004000997A2/en
Publication of WO2004000997A3 publication Critical patent/WO2004000997A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals

Definitions

  • the present invention relates to novel polypeptides, and the nucleic acids encoding them, having properties related to stimulation of biochemical or physiological responses in a cell, a tissue, an organ or an organism. More particularly, the novel polypeptides are gene products of novel genes, or are specified biologically active fragments or derivatives thereof. Methods of use encompass diagnostic and prognostic assay procedures as well as methods of treating diverse pathological conditions.
  • Eukaryotic cells are characterized by biochemical and physiological processes, which under normal conditions are extremely balanced to achieve the preservation and propagation of the cells.
  • the regulation of the biochemical and physiological processes involves intricate signaling pathways.
  • signaling pathways involve extracellular signaling proteins, cellular receptors that bind the signaling proteins and signal transducing components located within the cells.
  • Signaling proteins may be classified as endocrine effectors, paracrine effectors or autocrine effectors.
  • Endocrine effectors are signaling molecules secreted by a given organ into the circulatory system, which are then transported to a distant target organ or tissue.
  • the target cells include the receptors for the endocrine effector, and when the endocrine effector binds, a signaling cascade is induced.
  • Paracrine effectors involve secreting cells and receptor cells in close proximity to each other, for example, two different classes of cells in the same tissue or organ.
  • One class of cells secretes the paracrine effector, which then reaches the second class of cells, for example by diffusion through the extracellular fluid.
  • the second class of cells contains the receptors for the paracrine effector; binding of the effector results in induction of the signaling cascade that elicits the corresponding biochemical or physiological effect.
  • Autocrine effectors are highly analogous to paracrine effectors, except that the same cell type that secretes the autocrine effector also contains the receptor. Thus the autocrine effector binds to receptors on the same cell, or on identical neighboring cells. The binding process then elicits the characteristic biochemical or physiological effect. Signaling processes may elicit a variety of effects on cells and tissues including, by way of nonlimiting example, induction of cell or tissue proliferation, suppression of growth or proliferation, induction of differentiation or maturation of a cell or tissue, and suppression of differentiation or maturation of a cell or tissue.
  • pathological conditions involve dysregulation of expression of important effector proteins.
  • the dysregulation is manifested as diminished or suppressed level of synthesis and secretion of protein effectors.
  • the dysregulation is manifested as increased or up-regulated level of synthesis and secretion of protein effectors.
  • a subject may be suspected of suffering from a condition brought on by altered or mis-regulated levels of a protein effector of interest. Therefore there is a need to assay for the level of the protein effector of interest in a biological sample from such a subject, and to compare the level with that characteristic of a nonpathological condition. There also is a need to provide the protein effector as a product of manufacture.
  • Administration of the effector to a subject in need thereof is useful in treatment of the pathological condition. Accordingly, there is a need for a method of treatment of a pathological condition brought on by a diminished or suppressed levels of the protein effector of interest. In addition, there is a need for a method of treatment of a pathological condition brought on by a increased or up-regulated levels of the protein effector of interest.
  • Antibodies are multichain proteins that bind specifically to a given antigen, and bind poorly, or not at all, to substances deemed not to be cognate antigens.
  • Antibodies are comprised of two short chains termed light chains and two long chains termed heavy chains. These chains are constituted of immunoglobulin domains, of which generally there are two classes: one variable domain per chain, one constant domain in light chains, and three or more constant domains in heavy chains.
  • the antigen-specific portion of the immunoglobulin molecules resides in the variable domains; the variable domains of one light chain and one heavy chain associate with each other to generate the antigen-binding moiety.
  • Antibodies that bind immunospecifically to a cognate or target antigen bind with high affinities. Accordingly, they are useful in assaying specifically for the presence of the antigen in a sample. In addition, they have the potential of inactivating the activity of the antigen.
  • the invention is based in part upon the discovery of isolated polypeptides including amino acid sequences selected from mature forms of the amino acid sequences selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141.
  • novel nucleic acids and polypeptides are referred to herein as NOVX, or NOV1, NOV2, NOV3, etc., nucleic acids and polypeptides.
  • NOVX nucleic acid or polypeptide sequences.
  • the invention also is based in part upon variants of a mature form of the amino acid sequence selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141, wherein any amino acid in the mature form is changed to a different amino acid, provided that no more than 15% of the amino acid residues in the sequence of the mature form are so changed.
  • the invention includes the amino acid sequences selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141.
  • the invention also comprises variants of the amino acid sequence selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141, wherein any amino acid specified in the chosen sequence is changed to a different amino acid, provided that no more than 15% of the amino acid residues in the sequence are so changed.
  • the invention also involves fragments of any of the mature forms of the amino acid sequences selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141, or any other amino acid sequence selected from this group.
  • the invention also comprises fragments from these groups in which up to 15% of the residues are changed.
  • the invention encompasses polypeptides that are naturally occurring allelic variants of the sequence selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141.
  • allelic variants include amino acid sequences that are the translations of nucleic acid sequences differing by a single nucleotide from nucleic acid sequences selected from the group consisting of SEQ ID NOS: 2n-l, wherein n is an integer between 1 and 141.
  • the variant polypeptide where any amino acid changed in the chosen sequence is changed to provide a conservative substitution.
  • the invention comprises a pharmaceutical composition involving a polypeptide with an amino acid sequence selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141, and a pharmaceutically acceptable carrier.
  • the invention involves a kit, including, in one or more containers, this pharmaceutical composition.
  • the invention includes the use of a therapeutic in the manufacture of a medicament for treating a syndrome associated with a human disease, the disease being selected from a pathology associated with a polypeptide with an amino acid sequence selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141, wherein said therapeutic is the polypeptide selected from this group.
  • the invention comprises a method for determining the presence or amount of a polypeptide with an amino acid sequence selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141, in a sample, the method involving providing the sample; introducing the sample to an antibody that binds immunospecifically to the polypeptide; and determining the presence or amount of antibody bound to the polypeptide, thereby determining the presence or amount of polypeptide in the sample.
  • the invention includes a method for determining the presence of or predisposition to a disease associated with altered levels of a polypeptide with an amino acid sequence selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141, in a first mammalian subject, the method involving measuring the level of expression of the polypeptide in a sample from the first mammalian subject; and comparing the amount of the polypeptide in this sample to the amount of the polypeptide present in a control sample from a second mammalian subject known not to have, or not to be predisposed to, the disease, wherein an alteration in the expression level of the polypeptide in the first subject as compared to the control sample indicates the presence of or predisposition to the disease.
  • the invention involves a method of identifying an agent that binds to a polypeptide with an amino acid sequence selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141 , the method including introducing the polypeptide to the agent; and determining whether the agent binds to the polypeptide.
  • the agent could be a cellular receptor or a downstream effector.
  • the invention involves a method for identifying a potential therapeutic agent for use in treatment of a pathology, wherein the pathology is related to aberrant expression or aberrant physiological interactions of a polypeptide with an amino acid sequence selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141 , the method including providing a cell expressing the polypeptide of the invention and having a property or function ascribable to the polypeptide; contacting the cell with a composition comprising a candidate substance; and determining whether the substance alters the property or function ascribable to the polypeptide; whereby, if an alteration observed in the presence of the substance is not observed when the cell is contacted with a composition devoid of the substance, the substance is identified as a potential therapeutic agent.
  • the invention involves a method for screening for a modulator of activity or of latency or predisposition to a pathology associated with a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141 , the method including administering a test compound to a test animal at increased risk for a pathology associated with the polypeptide of the invention, wherein the test animal recombinantly expresses the polypeptide of the invention; measuring the activity of the polypeptide in the test animal after administering the test compound; and comparing the activity of the protein in the test animal with the activity of the polypeptide in a control animal not administered the polypeptide, wherein a change in the activity of the polypeptide in the test animal relative to the control animal indicates the test compound is a modulator of latency of, or predisposition to, a pathology associated with the polypeptide of the invention.
  • the recombinant test animal could express a test protein transgene or express the transgene under the control of a promoter at an increased level relative to a wild-type test animal
  • the promoter may or may not b the native gene promoter of the transgene.
  • the invention involves a method for modulating the activity of a polypeptide with an amino acid sequence selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141, the method including introducing a cell sample expressing the polypeptide with a compound that binds to the polypeptide in an amount sufficient to modulate the activity of the polypeptide.
  • the invention involves a method of treating or preventing a pathology associated with a polypeptide with an amino acid sequence selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141, the method including administering the polypeptide to a subject in which such treatment or prevention is desired in an amount sufficient to treat or prevent the pathology in the subject.
  • the subject could be human.
  • the invention involves a method of treating a pathological state in a mammal, the method including administering to the mammal a polypeptide in an amount that is sufficient to alleviate the pathological state, wherein the polypeptide is a polypeptide having an amino acid sequence at least 95% identical to a polypeptide having the amino acid sequence selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141, or a biologically active fragment thereof.
  • the invention involves an isolated nucleic acid molecule comprising a nucleic acid sequence encoding a polypeptide having an amino acid sequence selected from the group consisting of a mature form of the amino acid sequence given SEQ ID NO:2n, wherein n is an integer between 1 and 141, a variant of a mature form of the amino acid sequence selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141, wherein any amino acid in the mature form of the chosen sequence is changed to a different amino acid, provided that no more than 15% of the amino acid residues in the sequence of the mature form are so changed; the amino acid sequence selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141, a variant of the amino acid sequence selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141, in which any amino acid specified in the chosen sequence is changed to a different amino acid, provided that no more than 15% of
  • the invention comprises an isolated nucleic acid molecule having a nucleic acid sequence encoding a polypeptide comprising an amino acid sequence selected from the group consisting of a mature form of the amino acid sequence given SEQ ID NO:2n, wherein n is an integer between 1 and 141, wherein the nucleic acid molecule comprises the nucleotide sequence of a naturally occurring allelic nucleic acid variant.
  • the invention involves an isolated nucleic acid molecule including a nucleic acid sequence encoding a polypeptide having an amino acid sequence selected from the group consisting of a mature form of the amino acid sequence given SEQ ID NO:2n, wherein n is an integer between 1 and 141, that encodes a variant polypeptide, wherein the variant polypeptide has the polypeptide sequence of a naturally occurring polypeptide variant.
  • the invention comprises an isolated nucleic acid molecule having a nucleic acid sequence encoding a polypeptide comprising an amino acid sequence selected from the group consisting of a mature form of the amino acid sequence given SEQ ID NO:2n, wherein n is an integer between 1 and 141 , wherein the nucleic acid molecule differs by a single nucleotide from a nucleic acid sequence selected from the group consisting of SEQ ID NOS: 2n-l, wherein n is an integer between 1 and 141.
  • the invention includes an isolated nucleic acid molecule having a nucleic acid sequence encoding a polypeptide including an amino acid sequence selected from the group consisting of a mature form of the amino acid sequence given SEQ ID NO:2n, wherein n is an integer between 1 and 141, wherein the nucleic acid molecule comprises a nucleotide sequence selected from the group consisting of the nucleotide sequence selected from the group consisting of SEQ ID NO:2n-l , wherein n is an integer between 1 and 141, a nucleotide sequence wherein one or more nucleotides in the nucleotide sequence selected from the group consisting of SEQ ID NO:2n-l, wherein n is an integer between 1 and 141, is changed from that selected from the group consisting of the chosen sequence to a different nucleotide provided that no more than 15% of the nucleotides are so changed; a nucleic acid fragment of the sequence selected from the group consisting of SEQ ID NO:
  • the invention includes an isolated nucleic acid molecule having a nucleic acid sequence encoding a polypeptide including an amino acid sequence selected from the group consisting of a mature form of the amino acid sequence given SEQ ID NO:2n, wherein n is an integer between 1 and 141, wherein the nucleic acid molecule hybridizes under stringent conditions to the nucleotide sequence selected from the group consisting of SEQ ID NO:2n-l, wherein n is an integer between 1 and 141, or a complement of the nucleotide sequence.
  • the invention includes an isolated nucleic acid molecule having a nucleic acid sequence encoding a polypeptide including an amino acid sequence selected from the group consisting of a mature form of the amino acid sequence given SEQ ID NO:2n, wherein n is an integer between 1 and 141, wherein the nucleic acid molecule has a nucleotide sequence in which any nucleotide specified in the coding sequence of the chosen nucleotide sequence is changed from that selected from the group consisting of the chosen sequence to a different nucleotide provided that no more than 15% of the nucleotides in the chosen coding sequence are so changed, an isolated second polynucleotide that is a complement of the first polynucleotide, or a fragment of any of them.
  • the invention includes a vector involving the nucleic acid molecule having a nucleic acid sequence encoding a polypeptide including an amino acid sequence selected from the group consisting of a mature form of the amino acid sequence given SEQ ID NO:2n, wherein n is an integer between 1 and 141.
  • This vector can have a promoter operably linked to the nucleic acid molecule. This vector can be located within a cell.
  • the invention involves a method for determining the presence or amount of a nucleic acid molecule having a nucleic acid sequence encoding a polypeptide including an amino acid sequence selected from the group consisting of a mature form of the amino acid sequence given SEQ ID NO:2n, wherein n is an integer between 1 and 141, in a sample, the method including providing the sample; introducing the sample to a probe that binds to the nucleic acid molecule; and determining the presence or amount of the probe bound to the nucleic acid molecule, thereby determining the presence or amount of the nucleic acid molecule in the sample.
  • the presence or amount of the nucleic acid molecule is used as a marker for cell or tissue type.
  • the cell type can be cancerous.
  • the invention involves a method for determining the presence of or predisposition for a disease associated with altered levels of a nucleic acid molecule having a nucleic acid sequence encoding a polypeptide including an amino acid sequence selected from the group consisting of a mature form of the amino acid sequence given SEQ ID NO:2n, wherein n is an integer between 1 and 141, in a first mammalian subject, the method including measuring the amount of the nucleic acid in a sample from the first mammalian subject; and comparing the amount of the nucleic acid in the sample of step (a) to the amount of the nucleic acid present in a control sample from a second mammalian subject known not to have or not be predisposed to, the disease; wherein an alteration in the level of the nucleic acid in the first subject as compared to the control sample indicates the presence of or predisposition to the disease.
  • the present invention provides novel nucleotides and polypeptides encoded thereby. Included in the invention are the novel nucleic acid sequences, their encoded polypeptides, antibodies, and other related compounds.
  • the sequences are collectively referred to herein as "NOVX nucleic acids” or “NOVX polynucleotides” and the corresponding encoded polypeptides are referred to as “NOVX polypeptides” or “NOVX proteins.” Unless indicated otherwise, “NOVX” is meant to refer to any of the novel sequences disclosed herein.
  • Table A provides a summary of the NOVX nucleic acids and their encoded polypeptides. TABLE A. Sequences and Corresponding SEQ TD Numbers
  • Table A indicates the homology of NOVX polypeptides to known protein families.
  • nucleic acids and polypeptides, antibodies and related compounds according to the invention corresponding to a NOVX as identified in column 1 of Table A will be useful in therapeutic and diagnostic applications implicated in, for example, pathologies and disorders associated with the known protein families identified in column 5 of Table A.
  • Pathologies, diseases, disorders, conditions and the like that are associated with NOVX sequences include, but are not limited to, e.g., cardiomyopathy, atherosclerosis, hypertension, congenital heart defects, aortic stenosis, atrial septal defect (ASD), atrioventricular (A-V) canal defect, ductus arteriosus, pulmonary stenosis, subaortic stenosis, ventricular septal defect (VSD), valve diseases, tuberous sclerosis, scleroderma, obesity, metabolic disturbances associated with obesity, transplantation, adrenoleukodystrophy, congenital adrenal hyperplasia, prostate cancer, diabetes, metabolic disorders, neoplasm; adenocarcinoma, lymphoma, uterus cancer, fertility, hemophilia, hypercoagulation, idiopathic thrombocytopenic purpura, immunodeficiencies, graft versus host disease, AIDS, bronchial asthma, Crohn'
  • NOVX nucleic acids and their encoded polypeptides are useful in a variety of applications and contexts.
  • the various NOVX nucleic acids and polypeptides according to the invention are useful as novel members of the protein families according to the presence of domains and sequence relatedness to previously described proteins. Additionally, NOVX nucleic acids and polypeptides can also be used to identify proteins that are members of the family to which the NOVX polypeptides belong.
  • the NOVX polypeptides of the present invention show homology to, and contain domains that are characteristic of, other members of such protein families. Details of the sequence relatedness and domain analysis for each NOVX are presented in Example A.
  • the NOVX nucleic acids and polypeptides can also be used to screen for molecules, which inhibit or enhance NOVX activity or function.
  • the nucleic acids and polypeptides according to the invention may be used as targets for the identification of small molecules that modulate or inhibit diseases associated with the protein families listed in Table A.
  • NOVX nucleic acids and polypeptides are also useful for detecting specific cell types. Details of the expression analysis for each NOVX are presented in Example C. Accordingly, the NOVX nucleic acids, polypeptides, antibodies and related compounds according to the invention will have diagnostic and therapeutic applications in the detection of a variety of diseases with differential expression in normal vs. diseased tissues, e.g., detection of a variety of cancers.
  • NOVX clones NOVX nucleic acids and their encoded polypeptides are useful in a variety of applications and contexts.
  • the various NOVX nucleic acids and polypeptides according to the invention are useful as novel members of the protein families according to the presence of domains and sequence relatedness to previously described proteins. Additionally, NOVX nucleic acids and polypeptides can also be used to identify proteins that are members of the family to which the NOVX polypeptides belong.
  • the NOVX genes and their corresponding encoded proteins are useful for preventing, treating or ameliorating medical conditions, e.g., by protein or gene therapy.
  • Pathological conditions can be diagnosed by determining the amount of the new protein in a sample or by determining the presence of mutations in the new genes.
  • Specific uses are described for each of the NOVX genes, based on the tissues in which they are most highly expressed. Uses include developing products for the diagnosis or treatment of a variety of diseases and disorders.
  • the NOVX nucleic acids and proteins of the invention are useful in potential diagnostic and therapeutic applications and as a research tool. These include serving as a specific or selective nucleic acid or protein diagnostic and/or prognostic marker, wherein the presence or amount of the nucleic acid or the protein are to be assessed, as well as potential therapeutic applications such as the following: (i) a protein therapeutic, (ii) a small molecule drug target, (iii) an antibody target (therapeutic, diagnostic, drug targeting/cytotoxic antibody), (iv) a nucleic acid useful in gene therapy (gene delivery/gene ablation), and (v) a composition promoting tissue regeneration in vitro and in vivo (vi) a biological defense weapon.
  • the invention includes an isolated polypeptide comprising an amino acid sequence selected from the group consisting of: (a) a mature form of the amino acid sequence selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141, (b) a variant of a mature form of the amino acid sequence selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141, wherein any amino acid in the mature form is changed to a different amino acid, provided that no more than 15% of the amino acid residues in the sequence of the mature form are so changed; (c) an amino acid sequence selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141, (d) a variant of the amino acid sequence selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141, wherein any amino acid specified in the chosen sequence is changed to a different amino acid, provided that no more than 15% of the amino acid
  • the invention includes an isolated nucleic acid molecule comprising a nucleic acid sequence encoding a polypeptide comprising an amino acid sequence selected from the group consisting of: (a) a mature form of the amino acid sequence given SEQ ID NO:2n, wherein n is an integer between 1 and 141; (b) a variant of a mature form of the amino acid sequence selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141, wherein any amino acid in the mature form of the chosen sequence is changed to a different amino acid, provided that no more than 15% of the amino acid residues in the sequence of the mature form are so changed; (c) the amino acid sequence selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141 ; (d) a variant of the amino acid sequence selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141 , in which any amino acid specified in the chosen
  • the invention includes an isolated nucleic acid molecule, wherein said nucleic acid molecule comprises a nucleotide sequence selected from the group consisting of: (a) the nucleotide sequence selected from the group consisting of SEQ ID NO:2n-l, wherein n is an integer between 1 and 141; (b) a nucleotide sequence wherein one or more nucleotides in the nucleotide sequence selected from the group consisting of SEQ ID NO:2n-l, wherein n is an integer between 1 and 141, is changed from that selected from the group consisting of the chosen sequence to a different nucleotide provided that no more than 15% of the nucleotides are so changed; (c) a nucleic acid fragment of the sequence selected from the group consisting of SEQ ID NO:2n-l, wherein n is an integer between 1 and 141; and (d) a nucleic acid fragment wherein one or more nucleotides in the nucleotide sequence
  • NOVX Nucleic Acids and Polypeptides
  • One aspect of the invention pertains to isolated nucleic acid molecules that encode
  • nucleic acid fragments sufficient for use as hybridization probes to identify NOVX-encoding nucleic acids (e.g., NOVX mRNAs) and fragments for use as PCR primers for the amplification and/or mutation of NOVX nucleic acid molecules.
  • nucleic acid molecule is intended to include DNA molecules (e.g., cDNA or genomic DNA), RNA molecules (e.g., mRNA), analogs of the DNA or RNA generated using nucleotide analogs, and derivatives, fragments and homologs thereof.
  • the nucleic acid molecule may be single-stranded or double-stranded, but preferably is comprised double-stranded DNA.
  • a NOVX nucleic acid can encode a mature NOVX polypeptide.
  • a NOVX nucleic acid can encode a mature NOVX polypeptide.
  • a NOVX nucleic acid can encode a mature NOVX polypeptide.
  • a NOVX nucleic acid
  • mature form of a polypeptide or protein disclosed in the present invention is the product of a naturally occurring polypeptide, precursor form, or proprotein.
  • the naturally occurring polypeptide, precursor or proprotein includes, by way of nonlimiting example, the full-length gene product encoded by the corresponding gene. Alternatively, it may be defined as the polypeptide, precursor or proprotein encoded by an ORF described herein.
  • the product "mature" form arises, by way of nonlimiting example, as a result of one or more naturally occurring processing steps that may take place within the cell (e.g., host cell) in which the gene product arises.
  • Examples of such processing steps leading to a "mature" form of a polypeptide or protein include the cleavage of the N-terminal methionine residue encoded by the initiation codon of an ORF or the proteolytic cleavage of a signal peptide or leader sequence.
  • a mature form arising from a precursor polypeptide or protein that has residues 1 to N, where residue 1 is the N-terminal methionine would have residues 2 through N remaining after removal of the N-terminal methionine.
  • a mature form arising from a precursor polypeptide or protein having residues 1 to N, in which an N-terminal signal sequence from residue 1 to residue M is cleaved would have the residues from residue M+l to residue N remaining.
  • a "mature" form of a polypeptide or protein may arise from a post-translational modification step other than a proteolytic cleavage event.
  • additional processes include, by way of non-limiting example, glycosylation, myristylation or phosphorylation.
  • a mature polypeptide or protein may result from the operation of only one of these processes, or a combination of any of them.
  • probe refers to nucleic acid sequences of variable length, preferably between at least about 10 nucleotides (nt), about 100 nt, or as many as approximately, e.g., 6,000 nt, depending upon the specific use.
  • Probes are used in the detection of identical, similar, or complementary nucleic acid sequences. Longer length probes are generally obtained from a natural or recombinant source, are highly specific, and much slower to hybridize than shorter-length oligomer probes. Probes may be single- or double-stranded and designed to have specificity in PCR, membrane-based hybridization technologies, or ELISA-like technologies.
  • isolated nucleic acid molecule is a nucleic acid that is separated from other nucleic acid molecules which are present in the natural source of the nucleic acid.
  • an “isolated” nucleic acid is free of sequences which naturally flank the nucleic acid (i.e., sequences located at the 5'- and 3 '-termini of the nucleic acid) in the genomic DNA of the organism from which the nucleic acid is derived.
  • the isolated NOVX nucleic acid molecules can contain less than about 5 kb, about 4 kb, about 3 kb, about 2 kb, about 1 kb, about 0.5 kb, or about 0.1 kb, of nucleotide sequences which naturally flank the nucleic acid molecule in genomic DNA of the cell/tissue from which the nucleic acid is derived (e.g., brain, heart, liver, spleen, etc.).
  • an "isolated" nucleic acid molecule such as a cDNA molecule, can be substantially free of other cellular material, or culture medium, or of chemical precursors or other chemicals.
  • a nucleic acid molecule of the invention e.g., a nucleic acid molecule having the nucleotide sequence of SEQ ID NOS: 2n-l, wherein n is an integer between 1 and 141, or a complement of this nucleotide sequence, can be isolated using standard molecular biology techniques and the sequence information provided herein.
  • NOVX molecules can be isolated using standard hybridization and cloning techniques (e.g., as described in Sambrook, et al., (eds.), MOLECULAR CLONING: A LABORATORY MANUAL 2 nd Ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, 1989; and Ausubel, ⁇ t al, (eds.), CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, John Wiley & Sons, New York, NY, 1993).
  • standard hybridization and cloning techniques e.g., as described in Sambrook, et al., (eds.), MOLECULAR CLONING: A LABORATORY MANUAL 2 nd Ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, 1989; and Ausubel, ⁇ t al, (eds.), CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, John Wiley & Sons, New York, NY, 1993).
  • a nucleic acid of the invention can be amplified using cDNA, mRNA or, alternatively, genomic DNA as a template with appropriate oligonucleotide primers according to standard PCR amplification techniques.
  • the nucleic acid so amplified can be cloned into an appropriate vector and characterized by DNA sequence analysis.
  • oligonucleotides conesponding to NOVX nucleotide sequences can be prepared by standard synthetic techniques, e.g., using an automated DNA synthesizer.
  • oligonucleotide refers to a series of linked nucleotide residues.
  • a short oligonucleotide sequence may be based on, or designed from, a genomic or cDNA sequence and is used to amplify, confirm, or reveal the presence of an identical, similar or complementary DNA or RNA in a particular cell or tissue.
  • Oligonucleotides comprise a nucleic acid sequence having about 10 nt, 50 nt, or 100 nt in length, preferably about 15 nt to 30 nt in length.
  • an oligonucleotide comprising a nucleic acid molecule less than 100 nt in length would further comprise at least 6 contiguous nucleotides of SEQ ID NOS:2n-l , wherein n is an integer between 1 and 141 , or a complement thereof. Oligonucleotides may be chemically synthesized and may also be used as probes.
  • an isolated nucleic acid molecule of the invention comprises a nucleic acid molecule that is a complement of the nucleotide sequence shown in SEQ ID NOS:2n-l, wherein n is an integer between 1 and 141, or a portion of this nucleotide sequence (e.g., a fragment that can be used as a probe or primer or a fragment encoding a biologically-active portion of a NOVX polypeptide).
  • a nucleic acid molecule that is complementary to the nucleotide sequence of SEQ ID NOS:2n-l, wherein n is an integer between 1 and 141, is one that is sufficiently complementary to the nucleotide sequence of SEQ ID NOS:2n-l, wherein n is an integer between 1 and 141, that it can hydrogen bond with few or no mismatches to a nucleotide sequence of SEQ ID NOS:2n-l, wherein n is an integer between 1 and 141 , thereby forming a stable duplex.
  • binding means the physical or chemical interaction between two polypeptides or compounds or associated polypeptides or compounds or combinations thereof. Binding includes ionic, non-ionic, van der Waals, hydrophobic interactions, and the like.
  • a physical interaction can be either direct or indirect. Indirect interactions may be through or due to the effects of another polypeptide or compound. Direct binding refers to interactions that do not take place through, or due to, the effect of another polypeptide or compound, but instead are without other substantial chemical intermediates.
  • a “fragment” provided herein is defined as a sequence of at least 6 (contiguous) nucleic acids or at least 4 (contiguous) amino acids, a length sufficient to allow for specific hybridization in the case of nucleic acids or for specific recognition of an epitope in the case of amino acids, and is at most some portion less than a full length sequence. Fragments may be derived from any contiguous portion of a nucleic acid or amino acid sequence of choice.
  • a full-length NOVX clone is identified as containing an ATG translation start codon and an in-frame stop codon. Any disclosed NOVX nucleotide sequence lacking an ATG start codon therefore encodes a truncated C-terminal fragment of the respective NOVX polypeptide, and requires that the corresponding full-length cDNA extend in the 5' direction of the disclosed sequence. Any disclosed NOVX nucleotide sequence lacking an in-frame stop codon similarly encodes a truncated N-terminal fragment of the respective NOVX polypeptide, and requires that the conesponding full-length cDNA extend in the 3' direction of the disclosed sequence.
  • a “derivative” is a nucleic acid sequence or amino acid sequence formed from the native compounds either directly, by modification or partial substitution.
  • An “analog” is a nucleic acid sequence or amino acid sequence that has a structure similar to, but not identical to, the native compound, e.g. they differs from it in respect to certain components or side chains. Analogs may be synthetic or derived from a different evolutionary origin and may have a similar or opposite metabolic activity compared to wild type.
  • a “homolog” is a nucleic acid sequence or amino acid sequence of a particular gene that is derived from different species.
  • Derivatives and analogs may be full length or other than full length.
  • Derivatives or analogs of the nucleic acids or proteins of the invention include, but are not limited to, molecules comprising regions that are substantially homologous to the nucleic acids or proteins of the invention, in various embodiments, by at least about 70%, 80%, or 95% identity (with a preferred identity of 80-95%) over a nucleic acid or amino acid sequence of identical size or when compared to an aligned sequence in which the alignment is done by a computer homology program known in the art, or whose encoding nucleic acid is capable of hybridizing to the complement of a sequence encoding the proteins under stringent, moderately stringent, or low stringent conditions. See e.g.
  • homologous nucleic acid sequence or “homologous amino acid sequence,” or variations thereof, refer to sequences characterized by a homology at the nucleotide level or amino acid level as discussed above.
  • homologous nucleotide sequences include those sequences coding for isoforms of NOVX polypeptides. Isoforms can be expressed in different tissues of the same organism as a result of, for example, alternative splicing of RNA. Alternatively, isoforms can be encoded by different genes.
  • homologous nucleotide sequences include nucleotide sequences encoding for a NOVX polypeptide of species other than humans, including, but not limited to: vertebrates, and thus can include, e.g., frog, mouse, rat, rabbit, dog, cat cow, horse, and other organisms.
  • homologous nucleotide sequences also include, but are not limited to, naturally occurring allelic variations and mutations of the nucleotide sequences set forth herein.
  • a homologous nucleotide sequence does not, however, include the exact nucleotide sequence encoding human NOVX protein.
  • Homologous nucleic acid sequences include those nucleic acid sequences that encode conservative amino acid substitutions (see below) in SEQ ID NO:2 «-l, wherein n is an integer between 1 and 141, as well as a polypeptide possessing NOVX biological activity. Various biological activities of the NOVX proteins are described below.
  • a NOVX polypeptide is encoded by the open reading frame ("ORF") of a NOVX nucleic acid.
  • An ORF corresponds to a nucleotide sequence that could potentially be translated into a polypeptide.
  • a stretch of nucleic acids comprising an ORF is uninterrupted by a stop codon.
  • An ORF that represents the coding sequence for a full protein begins with an ATG "start” codon and terminates with one of the three “stop” codons, namely, TAA, TAG, or TGA.
  • an ORF may be any part of a coding sequence, with or without a start codon, a stop codon, or both.
  • a minimum size requirement is often set, e.g., a stretch of DNA that would encode a protem of 50 amino acids or more.
  • the nucleotide sequences determined from the cloning of the human NOVX genes allows for the generation of probes and primers designed for use in identifying and or cloning NOVX homologues in other cell types, e.g. from other tissues, as well as NOVX homologues from other vertebrates.
  • the probe/primer typically comprises substantially purified oligonucleotide.
  • the oligonucleotide typically comprises a region of nucleotide sequence that hybridizes under stringent conditions to at least about 12, 25, 50, 100, 150, 200, 250, 300, 350 or 400 consecutive sense strand nucleotide sequence of SEQ ID
  • Probes based on the human NOVX nucleotide sequences can be used to detect transcripts or genomic sequences encoding the same or homologous proteins.
  • the probe has a detectable label attached, e.g. the label can be a radioisotope, a fluorescent compound, an enzyme, or an enzyme co-factor.
  • Such probes can be used as a part of a diagnostic test kit for identifying cells or tissues which mis-express a NOVX protein, such as by measuring a level of a NOVX-encoding nucleic acid in a sample of cells from a subject e.g., detecting NOVX mRNA levels or determining whether a genomic NOVX gene has been mutated or deleted.
  • a polypeptide having a biologically-active portion of a NOVX polypeptide refers to polypeptides exhibiting activity similar, but not necessarily identical to, an activity of a polypeptide of the invention, including mature forms, as measured in a particular biological assay, with or without dose dependency.
  • a nucleic acid fragment encoding a "biologically-active portion of NOVX” can be prepared by isolating a portion of SEQ ID NO:2 «-l, wherein n is an integer between 1 and 141, that encodes a polypeptide having a NOVX biological activity (the biological activities of the NOVX proteins are described below), expressing the encoded portion of NOVX protein (e.g., by recombinant expression in vitro) and assessing the activity of the encoded portion of NOVX.
  • the invention further encompasses nucleic acid molecules that differ from the nucleotide sequences of SEQ ID NO:2 «-l, wherein n is an integer between 1 and 141 , due to degeneracy of the genetic code and thus encode the same NOVX proteins as that encoded by the nucleotide sequences of SEQ ID NO:2 «-l , wherein n is an integer between 1 and 141.
  • an isolated nucleic acid molecule of the . invention has a nucleotide sequence encoding a protein having an amino acid sequence of SEQ ID NO:27z, wherein n is an integer between 1 and 141.
  • NOVX nucleotide sequences of SEQ ID NO:2 «-l, wherein n is an integer between 1 and 141 .
  • DNA sequence polymorphisms that lead to changes in the amino acid sequences of the NOVX polypeptides may exist within a population (e.g., the human population).
  • Such genetic polymorphism in the NOVX genes may exist among individuals within a population due to natural allelic variation.
  • the terms "gene” and “recombinant gene” refer to nucleic acid molecules comprising an open reading frame (ORF) encoding a NOVX protein, preferably a vertebrate NOVX protein.
  • Such natural allelic variations can typically result in 1-5% variance in the nucleotide sequence of the NOVX genes. Any and all such nucleotide variations and resulting amino acid polymorphisms in the NOVX polypeptides, which are the result of natural allelic variation and that do not alter the functional activity of the NOVX polypeptides, are intended to be within the scope of the invention. Moreover, nucleic acid molecules encoding NOVX proteins from other species, and thus that have a nucleotide sequence that differs from a human SEQ ID NO:2 «-l, wherein n is an integer between 1 and 141, are intended to be within the scope of the invention.
  • Nucleic acid molecules corresponding to natural allelic variants and homologues of the NOVX cDNAs of the invention can be isolated based on their homology to the human NOVX nucleic acids disclosed herein using the human cDNAs, or a portion thereof, as a hybridization probe according to standard hybridization techniques under stringent hybridization conditions.
  • an isolated nucleic acid molecule of the invention is at least 6 nucleotides in length and hybridizes under stringent conditions to the nucleic acid molecule comprising the nucleotide sequence of SEQ ID NO:2?z-l, wherein n is an integer between 1 and 141.
  • the nucleic acid is at least 10, 25, 50, 100, 250, 500, 750, 1000, 1500, or 2000 or more nucleotides in length.
  • an isolated nucleic acid molecule of the invention hybridizes to the coding region.
  • the term "hybridizes under stringent conditions" is intended to describe conditions for hybridization and washing under which nucleotide sequences at least about 65% homologous to each other typically remain hybridized to each other.
  • Homologs i.e., nucleic acids encoding NOVX proteins derived from species other than human
  • other related sequences e.g., paralogs
  • stringent hybridization conditions refers to conditions under which a probe, primer or oligonucleotide will hybridize to its target sequence, but to no other sequences. Stringent conditions are sequence-dependent and will be different in different circumstances. Longer sequences hybridize specifically at higher temperatures than shorter sequences. Generally, stringent conditions are selected to be about 5 °C lower than the thermal melting point (Tm) for the specific sequence at a defined ionic strength and pH. The Tm is the temperature (under defined ionic strength, pH and nucleic acid concentration) at which 50% of the probes complementary to the target sequence hybridize to the target sequence at equilibrium. Since the target sequences are generally present at excess, at Tm, 50% of the probes are occupied at equilibrium.
  • Tm thermal melting point
  • stringent conditions will be those in which the salt concentration is less than about 1.0 M sodium ion, typically about 0.01 to 1.0 M sodium ion (or other salts) at pH 7.0 to 8.3 and the temperature is at least about 30 °C for short probes, primers or oligonucleotides (e.g., 10 nt to 50 nt) and at least about 60 °C for longer probes, primers and oligonucleotides.
  • Stringent conditions may also be achieved with the addition of destabilizing agents, such as formamide.
  • Stringent conditions are known to those skilled in the art and can be found in Ausubel, et al., (eds.), CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, John Wiley & Sons, N.Y. (1989), 6.3.1-6.3.6.
  • the conditions are such that sequences at least about 65%, 70%, 75%, 85%, 90%, 95%, 98%, or 99% homologous to each other typically remain hybridized to each other.
  • a non-limiting example of stringent hybridization conditions are hybridization in a high salt buffer comprising 6X SSC, 50 mM Tris-HCl (pH 7.5), 1 mM EDTA, 0.02% PVP, 0.02% Ficoll, 0.02% BSA, and 500 mg/ml denatured salmon sperm DNA at 65°C, followed by one or more washes in 0.2X SSC, 0.01%o BSA at 50°C.
  • An isolated nucleic acid molecule of the invention that hybridizes under stringent conditions to a sequence of SEQ ID NO:2n-l , wherein n is an integer between 1 and 141, corcesponds to a naturally-occurring nucleic acid molecule.
  • a "naturally-occurring" nucleic acid molecule refers to an RNA or DNA molecule having a nucleotide sequence that occurs in nature (e.g., encodes a natural protein).
  • a nucleic acid sequence that is hybridizable to the nucleic acid molecule comprising the nucleotide sequence of SEQ ID NO:2 «-l , wherein n is an integer between 1 and 141, or fragments, analogs or derivatives thereof, under conditions of moderate stringency is provided.
  • moderate stringency hybridization conditions are hybridization in 6X SSC, 5X Reinhardt's solution, 0.5% SDS and 100 mg/ml denatured salmon sperm DNA at 55 °C, followed by one or more washes in IX SSC, 0.1% SDS at 37 °C.
  • Other conditions of moderate stringency that may be used are well-known within the art. See, e.g., Ausubel, et al. (eds.), 1993, CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, John Wiley & Sons, NY, and Krieger, 1990; GENE TRANSFER AND EXPRESSION, A LABORATORY MANUAL, Stockton Press, NY.
  • a nucleic acid that is hybridizable to the nucleic acid molecule comprising the nucleotide sequences of SEQ ID NO:2w-l, wherein n is an integer between 1 and 141, or fragments, analogs or derivatives thereof, under conditions of low stringency, is provided.
  • low stringency hybridization conditions are hybridization in 35% formamide, 5X SSC, 50 mM Tris-HCl (pH 7.5), 5 mM EDTA, 0.02% PVP, 0.02% Ficoll, 0.2% BSA, 100 mg/ml denatured salmon sperm DNA, 10% (wt/vol) dextran sulfate at 40°C, followed by one or more washes in 2X SSC, 25 mM Tris-HCl (pH 7.4), 5 M EDTA, and 0.1% SDS at 50 °C.
  • Other conditions of low stringency that may be used are well known in the art (e.g., as employed for cross-species hybridizations).
  • nucleotide sequences of SEQ ID NO:2>J-1 wherein n is an integer between l and 141, thereby leading to changes in the amino acid sequences of the encoded NOVX protein, without altering the functional ability of that NOVX protein.
  • nucleotide substitutions leading to amino acid substitutions at "non-essential" amino acid residues can be made in the sequence of SEQ ID NO:2w, wherein n is an integer between 1 and 141.
  • non-essential amino acid residue is a residue that can be altered from the wild-type sequences of the NOVX proteins without altering their biological activity, whereas an "essential" amino acid residue is required for such biological activity.
  • amino acid residues that are conserved among the NOVX proteins of the invention are predicted to be particularly non-amenable to alteration. Amino acids for which conservative substitutions can be made are well-known within the art.
  • Another aspect of the invention pertains to nucleic acid molecules encoding
  • NOVX proteins that contain changes in amino acid residues that are not essential for activity. Such NOVX proteins differ in amino acid sequence from SEQ ID NO:2 «-l, wherein n is an integer between 1 and 141, yet retain biological activity.
  • the isolated nucleic acid molecule comprises a nucleotide sequence encoding a protein, wherein the protem comprises an amino acid sequence at least about 40% homologous to the amino acid sequences of SEQ ID NO:2/2, wherein n is an integer between 1 and 141.
  • the protein encoded by the nucleic acid molecule is at least about 60% homologous to SEQ ID NO:2«, wherein n is an integer between 1 and 141; more preferably at least about 70% homologous to SEQ ID NO:2n, wherein n is an integer between 1 and 141; still more preferably at least about 80% homologous to SEQ ID NO:2«, wherein n is an integer between 1 and 141; even more preferably at least about 90% homologous to SEQ ID NO:2n, wherein n is an integer between 1 and 141 ; and most preferably at least about 95% homologous to SEQ ID NO:2«, wherein n is an integer between 1 and 141.
  • An isolated nucleic acid molecule encoding a NOVX protein homologous to the protein of SEQ ID NO:2 «, wherein n is an integer between 1 and 141, can be created by introducing one or more nucleotide substitutions, additions or deletions into the nucleotide sequence of SEQ ID NO:2 «-l, wherein n is an integer between 1 and 141, such that one or more amino acid substitutions, additions or deletions are introduced into the encoded protein.
  • Mutations can be introduced any one of SEQ ID NO:2/z-l, wherein n is an integer between 1 and 141, by standard techniques, such as site-directed mutagenesis and
  • conservative amino acid substitutions are made at one or more predicted, non-essential amino acid residues.
  • a "conservative amino acid substitution” is one in which the amino acid residue is replaced with an amino acid residue having a similar side chain. Families of amino acid residues having similar side chains have been defined within the art.
  • amino acids with basic side chains e.g., lysine, arginine, histidine
  • acidic side chains e.g., aspartic acid, glutamic acid
  • uncharged polar side chains e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine
  • nonpolar side chains e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan
  • beta-branched side chains e-g; threonine, valine, isoleucine
  • aromatic side chains e.g., tyrosine, phenylalanine, tryptophan, histidine
  • a predicted non-essential amino acid residue in the NOVX protein is replaced with another amino acid residue from the same side chain family.
  • mutations can be introduced randomly along all or part of a NOVX coding sequence, such as by saturation mutagenesis, and the resultant mutants can be screened for NOVX biological activity to identify mutants that retain activity.
  • mutagenesis of a nucleic acid of SEQ ID NO:2 «-l, wherein n is an integer between 1 and 141 the encoded protein can be expressed by any recombinant technology known in the art and the activity of the protein can be determined.
  • amino acid families may also be determined based on side chain interactions.
  • Substituted amino acids may be fully conserved "strong” residues or fully conserved “weak” residues.
  • the "strong” group of conserved amino acid residues may be any one of the following groups: STA, NEQK, NHQK, NDEQ, QHRK, MILV, MILF, HY, FYW, wherein the single letter amino acid codes are grouped by those amino acids that may be substituted for each other.
  • the "weak" group of conserved residues may be any one of the following: CSA, ATV, SAG, STNK, STPA, SGND, SNDEQK, NDEQHK, NEQHRK, HFY, wherein the letters within each group represent the single letter amino acid code.
  • a mutant NOVX protein can be assayed for (i) the ability to form proteimprotein interactions with other NOVX proteins, other cell-surface proteins, or biologically-active portions thereof, (ii) complex formation between a mutant NOVX protein and a NOVX ligand; or (iii) the ability of a mutant NOVX protein to bind to an intracellular target protein or biologically-active portion thereof; (e.g. avidin proteins).
  • a mutant NOVX protein can be assayed for the ability to regulate a specific biological function (e.g., regulation of insulin release).
  • NOVX gene expression can be attenuated by RNA interference.
  • RNA interference One approach well-known in the art is short interfering RNA (siRNA) mediated gene silencing where expression products of a NOVX gene are targeted by specific double stranded NOVX derived siRNA nucleotide sequences that are complementary to at least a 19-25 nt long segment of the NOVX gene transcript, including the 5' untranslated (UT) region, the ORF, or the 3' UT region.
  • siRNA short interfering RNA
  • Targeted genes can be a NOVX gene, or an upstream or downstream modulator of the NOVX gene.
  • upstream or downstream modulators of a NOVX gene include, e.g., a transcription factor that binds the NOVX gene promoter, a kinase or phosphatase that interacts with a NOVX polypeptide, and polypeptides involved in a NOVX regulatory pathway.
  • NOVX gene expression is silenced using short interfering RNA.
  • a NOVX polynucleotide according to the invention includes a siRNA polynucleotide.
  • a NOVX siRNA can be obtained using a NOVX polynucleotide sequence, for example, by processing the NOVX ribopolynucleotide sequence in a cell-free system, such as but not limited to a Drosophila extract, or by transcription of recombinant double stranded NOVX RNA or by chemical synthesis of nucleotide sequences homologous to a NOVX sequence.
  • RNA synthesis provides about 1 milligram of siRNA, which is sufficient for 1000 transfection experiments using a 24-well tissue culture plate format.
  • siRNA duplexes composed of a 21-nt sense strand and a 21-nt antisense strand, paired in a manner to have a 2-nt 3' overhang.
  • the sequence of the 2-nt 3' overhang makes an additional small contribution to the specificity of siRNA target recognition.
  • the contribution to specificity is localized to the unpaired nucleotide adjacent to the first paired bases.
  • the nucleotides in the 3' overhang are ribonucleotides.
  • the nucleotides in the 3' overhang are deoxyribonucleotides.
  • a contemplated recombinant expression vector of the invention comprises a NOVX DNA molecule cloned into an expression vector comprising operatively-linked regulatory sequences flanking the NOVX sequence in a manner that allows for expression (by transcription of the DNA molecule) of both strands.
  • An RNA molecule that is antisense to NOVX mRNA is transcribed by a first promoter (e.g., a promoter sequence 3 ' of the cloned DNA) and an RNA molecule that is the sense strand for the NOVX mRNA is transcribed by a second promoter (e.g., a promoter sequence 5' of the cloned DNA).
  • the sense and antisense strands may hybridize in vivo to generate siRNA constructs for silencing of the NOVX gene.
  • two constructs can be utilized to create the sense and anti-sense strands of a siRNA construct.
  • cloned DNA can encode a construct having secondary structure, wherein a single transcript has both the sense and complementary antisense sequences from the target gene or genes.
  • a hairpin RNAi product is homologous to all or a portion of the target gene.
  • a hairpin RNAi product is a siRNA.
  • the regulatory sequences flanking the NOVX sequence may be identical or may be different, such that their expression may be modulated independently, or in a temporal or spatial manner.
  • siRNAs are transcribed intracellularly by cloning the NOVX gene templates into a vector containing, e.g., a RNA pol III transcription unit from the smaller nuclear RNA (snRNA) U6 or the human RNase P RNA HI .
  • a vector system is the GeneSuppressorTM RNA Interference kit (commercially available from Imgenex).
  • the U6 and HI promoters are members of the type III class of Pol III promoters.
  • the +1 nucleotide of the U6-like promoters is always guanosine, whereas the +1 for HI promoters is adenosine.
  • the termination signal for these promoters is defined by five consecutive thymidines.
  • the transcript is typically cleaved after the second uridine. Cleavage at this position generates a 3' UU overhang in the expressed siRNA, which is similar to the 3' overhangs of synthetic siRNAs. Any sequence less than 400 nucleotides in length can be transcribed by these promoter, therefore they are ideally suited for the expression of around 21 -nucleotide siRNAs in, e.g., an approximately 50-nucleotide RNA stem-loop transcript.
  • siRNA vector appears to have an advantage over synthetic siRNAs where long term knock-down of expression is desired.
  • Cells transfected with a siRNA expression vector would experience steady, long-term mRNA inhibition.
  • cells transfected with exogenous synthetic siRNAs typically recover from mRNA suppression within seven days or ten rounds of cell division.
  • the long-term gene silencing ability of siRNA expression vectors may provide for applications in gene therapy.
  • siRNAs are chopped from longer dsRNA by an ATP-dependent ribonuclease called DICER.
  • DICER is a member of the RNase III family of double-stranded RNA-specific endonucleases. The siRNAs assemble with cellular proteins into an endonuclease complex.
  • siRNAs/protein complex siRNP
  • RISC RNA-induced silencing complex
  • the siRNA thus acts as a guide, restricting the ribonuclease to cleave only mRNAs complementary to one of the two siRNA strands.
  • a NOVX mRNA region to be targeted by siRNA is generally selected from a desired NOVX sequence beginning 50 to 100 nt downstream of the start codon. Alternatively, 5' or 3' UTRs and regions nearby the start codon can be used but are generally avoided, as these may be richer in regulatory protein binding sites. UTR-binding proteins and/or translation initiation complexes may interfere with binding of the siRNP or RISC endonuclease complex. An initial BLAST homology search for the selected siRNA sequence is done against an available nucleotide sequence library to ensure that only one gene is targeted.
  • siRNA duplexes Specificity of target recognition by siRNA duplexes indicate that a single point mutation located in the paired region of an siRNA duplex is sufficient to abolish target mRNA degradation. See, Elbashir et al. 2001 EMBO J. 20(23):6877-88. Hence, consideration should be taken to accommodate SNPs, polymorphisms, allelic variants or species-specific variations when targeting a desired gene.
  • a complete NOVX siRNA experiment includes the proper negative control.
  • a negative control siRNA generally has the same nucleotide composition as the NOVX siRNA but lack significant sequence homology to the genome. Typically, one would scramble the nucleotide sequence of the NOVX siRNA and do a homology search to make sure it lacks homology to any other gene.
  • Two independent NOVX siRNA duplexes can be used to knock-down a target NOVX gene. This helps to control for specificity of the silencing effect.
  • expression of two independent genes can be simultaneously knocked down by using equal concentrations of different NOVX siRNA duplexes, e.g., a NOVX siRNA and an siRNA for a regulator of a NOVX gene or polypeptide.
  • NOVX siRNA duplexes e.g., a NOVX siRNA and an siRNA for a regulator of a NOVX gene or polypeptide.
  • Availability of siRNA-associating proteins is believed to be more limiting than target mRNA accessibility.
  • a targeted NOVX region is typically a sequence of two adenines (AA) and two thymidines (TT) divided by a spacer region of nineteen (NI 9) residues (e.g.,
  • a desirable spacer region has a G/C-content of approximately 30% to 70%, and more preferably of about 50%. If the sequence AA(N19)TT is not present in the target sequence, an alternative target region would be AA(N21).
  • Symmetric 3' overhangs may help to ensure that the siRNPs are formed with approximately equal ratios of sense and antisense target RNA-cleaving siRNPs. See, e.g., Elbashir, Lendeckel and Tuschl (2001). Genes & Dev. 15: 188-200, incorporated by reference herein in its entirely.
  • the modification of the overhang of the sense sequence of the siRNA duplex is not expected to affect targeted mRNA recognition, as the antisense siRNA strand guides target recognition.
  • the NOVX target mRNA does not contain a suitable AA(N21) sequence
  • the sequence of the sense strand and antisense strand may still be synthesized as 5' (N19)TT, as it is believed that the sequence of the 3'-most nucleotide of the antisense siRNA does not contribute to specificity.
  • the secondary structure of the target mRNA does not appear to have a strong effect on silencing. See, Harborth, et al. (2001) J. Cell Science 114: 4557-4565, incorporated by reference in its entirety.
  • Transfection of NOVX siRNA duplexes can be achieved using standard nucleic acid transfection methods, for example, OLIGOFECTAMINE Reagent (commercially available from Invitrogen).
  • An assay for NOVX gene silencing is generally performed approximately 2 days after transfection. No NOVX gene silencing has been observed in the absence of transfection reagent, allowing for a comparative analysis of the wild-type and silenced NOVX phenotypes.
  • approximately 0.84 ⁇ g of the siRNA duplex is generally sufficient. Cells are typically seeded the previous day, and are transfected at about 50% confluence.
  • the choice of cell culture media and conditions are routine to those of skill in the art, and will vary with the choice of cell type.
  • the efficiency of transfection may depend on the cell type, but also on the passage number and the confluency of the cells.
  • the time and the manner of formation of siRNA-liposome complexes are also critical. Low transfection efficiencies are the most frequent cause of unsuccessful NOVX silencing.
  • the efficiency of transfection needs to be carefully examined for each new cell line to be used.
  • Prefened cell are derived from a mammal, more preferably from a rodent such as a rat or mouse, and most preferably from a human. Where used for therapeutic treatment, the cells are preferentially autologous, although non-autologous cell sources are also contemplated as within the scope of the present invention.
  • transfection of 0.84 ⁇ g single-stranded sense NOVX siRNA will have no effect on NOVX silencing, and 0.84 ⁇ g antisense siRNA has a weak silencing effect when compared to 0.84 ⁇ g of duplex siRNAs.
  • Control experiments again allow for a comparative analysis of the wild-type and silenced NOVX phenotypes.
  • targeting of common proteins is typically performed, for example targeting of lamin A/C or transfection of a CMV-driven EGFP-expression plasmid (e.g. commercially available from Clontech).
  • a determination of the fraction of lamin A C knockdown in cells is determined the next day by such techniques as immunofluorescence, Western blot, Northern blot or other similar assays for protein expression or gene expression.
  • Lamin A/C monoclonal antibodies may be obtained from Santa Cruz Biotechnology.
  • a knock-down phenotype may become apparent after 1 to 3 days, or even later.
  • depletion of the NOVX polynucleotide may be observed by immunofluorescence or Western blotting. If the NOVX polynucleotide is still abundant after 3 days, cells need to be split and transferred to a fresh 24-well plate for re-transfection. If no knock-down of the targeted protein is observed, it may be desirable to analyze whether the target mRNA (NOVX or a NOVX upstream or downstream gene) was effectively destroyed by the transfected siRNA duplex.
  • RNA is prepared, reverse transcribed using a target-specific primer, and PCR-amplified with a primer pair covering at least one exon-exon junction in order to control for amplification of pre-mRNAs.
  • RT/PCR of a non-targeted mRNA is also needed as control. Effective depletion of the mRNA yet undetectable reduction of target protein may indicate that a large reservoir of stable NOVX protein may exist in the cell. Multiple transfection in sufficiently long intervals may be necessary until the target protein is finally depleted to a point where a phenotype may become apparent. If multiple transfection steps are required, cells are split 2 to 3 days after transfection. The cells may be transfected immediately after splitting.
  • An inventive therapeutic method of the invention contemplates administering a NOVX siRNA construct as therapy to compensate for increased or aberrant NOVX expression or activity.
  • the NOVX ribopolynucleotide is obtained and processed into siRNA fragments, or a NOVX siRNA is synthesized, as described above.
  • the NOVX siRNA is administered to cells or tissues using known nucleic acid transfection techniques, as described above.
  • a NOVX siRNA specific for a NOVX gene will decrease or knockdown NOVX transcription products, which will lead to reduced NOVX polypeptide production, resulting in reduced NOVX polypeptide activity in the cells or tissues.
  • the present invention also encompasses a method of treating a disease or condition associated with the presence of a NOVX protein in an individual comprising administering to the individual an RNAi construct that targets the mRNA of the protein (the mRNA that encodes the protein) for degradation.
  • a specific RNAi construct includes a siRNA or a double stranded gene transcript that is processed into siRNAs. Upon treatment, the target protein is not produced or is not produced to the extent it would be in the absence of the treatment.
  • a control sample of cells or tissues from healthy individuals provides a reference standard for determining NOVX expression levels. Expression levels are detected using the assays described, e.g., RT-PCR, Northern blotting, Western blotting, ELISA, and the like.
  • a subject sample of cells or tissues is taken from a mammal, preferably a human subject, suffering from a disease state.
  • the NOVX ribopolynucleotide is used to produce siRNA constructs, that are specific for the NOVX gene product.
  • NOVX siRNA' s are treated by administering NOVX siRNA' s to the cells or tissues by methods described for the transfection of nucleic acids into a cell or tissue, and a change in NOVX polypeptide or polynucleotide expression is observed in the subject sample relative to the control sample, using the assays described.
  • This NOVX gene knockdown approach provides a rapid method for determination of a NOVX minus (NOVX " ) phenotype in the treated subject sample.
  • NOVX " phenotype observed in the treated subject sample thus serves as a marker for monitoring the course of a disease state during treatment.
  • a NOVX siRNA is used in therapy.
  • Methods for the generation and use of a NOVX siRNA are known to those skilled in the art. Example techniques are provided below. Production of RNAs
  • Sense RNA (ssRNA) and antisense RNA (asRNA) of NOVX are produced using known methods such as transcription in RNA expression vectors.
  • the sense and antisense RNA are about 500 bases in length each.
  • the produced ssRNA and asRNA (0.5 ⁇ M) in 10 mM Tris-HCl (pH 7.5) with 20 mM NaCl were heated to 95° C for 1 min then cooled and annealed at room temperature for 12 to 16 h.
  • the RNAs are precipitated and resuspended in lysis buffer (below).
  • RNAs are electrophoresed in a 2% agarose gel in TBE buffer and stained with ethidium bromide. See, e.g., Sambrook et al., Molecular Cloning. Cold Spring Harbor Laboratory Press, Plainview, N.Y. (1989).
  • Untreated rabbit reticulocyte lysate (Ambion) are assembled according to the manufacturer's directions. dsRNA is incubated in the lysate at 30° C for 10 min prior to the addition of mRNAs. Then NOVX mRNAs are added and the incubation continued for an additional 60 min. The molar ratio of double stranded RNA and mRNA is about 200: 1. The NOVX mRNA is radiolabeled (using known techniques) and its stability is monitored by gel electrophoresis. In a parallel experiment made with the same conditions, the double stranded RNA is internally radiolabeled with a 32 P-ATP.
  • the band of double stranded RNA about 21-23 bps, is eluded.
  • the efficacy of these 21-23 mers for suppressing NOVX transcription is assayed in vitro using the same rabbit reticulocyte assay described above using 50 nanomolar of double stranded 21-23 mer for each assay.
  • the sequence of these 21-23 mers is then determined using standard nucleic acid sequencing techniques.
  • RNAs are chemically synthesized using Expedite RNA phosphoramidites and thymidine phosphoramidite (Proligo, Germany). Synthetic oligonucleotides are deprotected and gel-purified (Elbashir, Lendeckel, & Tuschl, Genes & Dev. 15, 188-200 (2001)), followed by Sep-Pak C18 cartridge (Waters, Milford, Mass., USA) purification (Tuschl, et al., Biochemistry, 32:11658-11668 (1993)).
  • RNAs (20 ⁇ M) single strands are incubated in annealing buffer (100 mM potassium acetate, 30 mM HEPES-KOH at pH 7.4, 2 mM magnesium acetate) for 1 min at 90° C followed by 1 h at 37° C.
  • annealing buffer 100 mM potassium acetate, 30 mM HEPES-KOH at pH 7.4, 2 mM magnesium acetate
  • a cell culture known in the art to regularly express NOVX is propagated using standard conditions. 24 hours before transfection, at approx. 80% confluency, the cells are trypsinized and diluted 1 :5 with fresh medium without antibiotics (1-3 X 105 cells/ml) and transferred to 24-well plates (500 ml/well). Transfection is performed using a commercially available lipofection kit and NOVX expression is monitored using standard techniques with positive and negative control. A positive control is cells that naturally express NOVX while a negative control is cells that do not express NOVX. Base-paired 21 and 22 nt siRNAs with overhanging 3' ends mediate efficient sequence-specific mRNA degradation in lysates and in cell culture. Different concentrations of siRNAs are used.
  • An efficient concentration for suppression in vitro in mammalian culture is between 25 nM to 100 nM final concentration. This indicates that siRNAs are effective at concentrations that are several orders of magnitude below the concentrations applied in conventional antisense or ribozyme gene targeting experiments.
  • the above method provides a way both for the deduction of NOVX siRNA sequence and the use of such siRNA for in vitro suppression. In vivo suppression may be performed using the same siRNA using well known in vivo transfection or gene therapy transfection techniques.
  • Antisense Nucleic Acids Another aspect of the invention pertains to isolated antisense nucleic acid molecules that are hybridizable to or complementary to the nucleic acid molecule comprising the nucleotide sequence of SEQ ID NO:2 «-l , wherein n is an integer between 1 and 141, or fragments, analogs or derivatives thereof.
  • An "antisense" nucleic acid comprises a nucleotide sequence that is complementary to a "sense" nucleic acid encoding a protein (e.g., complementary to the coding strand of a double-stranded cDNA molecule or complementary to an mRNA sequence).
  • antisense nucleic acid molecules comprise a sequence complementary to at least about 10, 25, 50, 100, 250 or 500 nucleotides or an entire NOVX coding strand, or to only a portion thereof.
  • Nucleic acid molecules encoding fragments, homologs, derivatives and analogs of a NOVX protein of SEQ ID NO:2?j, wherein n is an integer between 1 and 141, or antisense nucleic acids complementary to a NOVX nucleic acid sequence of SEQ ID NO:2n-l, wherein n is an integer between 1 and 141, are additionally provided.
  • an antisense nucleic acid molecule is antisense to a "coding region" of the coding strand of a nucleotide sequence encoding a NOVX protein.
  • coding region refers to the region of the nucleotide sequence comprising codons which are translated into amino acid residues.
  • the antisense nucleic acid molecule is antisense to a "noncoding region" of the coding strand of a nucleotide sequence encoding the NOVX protein.
  • noncoding region refers to 5' and 3' sequences which flank the coding region that are not translated into amino acids (i. e. , also referred to as 5' and 3' untranslated regions).
  • antisense nucleic acids of the invention can be designed according to the rules of Watson and Crick or Hoogsteen base pairing.
  • the antisense nucleic acid molecule can be complementary to the entire coding region of NOVX mRNA, but more preferably is an oligonucleotide that is antisense to only a portion of the coding or noncoding region of NOVX mRNA.
  • the antisense oligonucleotide can be complementary to the region smrounding the translation start site of NOVX mRNA.
  • An antisense oligonucleotide can be, for example, about 5, 10, 15, 20, 25, 30, 35, 40, 45 or 50 nucleotides in length.
  • An antisense nucleic acid of the invention can be constructed using chemical synthesis or enzymatic ligation reactions using procedures known in the art.
  • an antisense nucleic acid e.g., an antisense oligonucleotide
  • an antisense nucleic acid can be chemically synthesized using naturally-occurring nucleotides or variously modified nucleotides designed to increase the biological stability of the molecules or to increase the physical stability of the duplex formed between the antisense and sense nucleic acids (e.g., phosphorothioate derivatives and acridine substituted nucleotides can be used).
  • modified nucleotides that can be used to generate the antisense nucleic acid include: 5-fluorouracil, 5-bromouracil, 5-chlorouracil, 5-iodouracil, hypoxanthine, xanthine, 4-acetylcytosine, 5-carboxymethylaminomethyl-2-thiouridine, 5-(carboxyhydroxylmethyl) uracil, 5-carboxymethylaminomethyluracil, dihydrouracil, beta-D-galactosylqueosine, inosine, N6-isopentenyladenine, 1-methylguanine, 1-methylinosine, 2,2-dimethylguanine, 2-methyladenine, 2-methylguanine, 5-methoxyuracil, 3-methylcytosine, 5-methylcytosine, N6-adenine, 7-methylguanine, 5-methylaminomethyluracil, 5-methoxyaminomethyl-2-thiouracil, 2-thiouracil, 4-thiouracil
  • the antisense nucleic acid can be produced biologically using an expression vector into which a nucleic acid has been subcloned in an antisense orientation (i.e., RNA transcribed from the inserted nucleic acid will be of an antisense orientation to a target nucleic acid of interest, described further in the following subsection).
  • the antisense nucleic acid molecules of the invention are typically administered to a subject or generated in situ such that they hybridize with or bind to cellular mRNA and/or genomic DNA encoding a NOVX protein to thereby inhibit expression of the protein (e.g., by inhibiting transcription and/or translation).
  • the hybridization can be by conventional nucleotide complementarity to form a stable duplex, or, for example, in the case of an antisense nucleic acid molecule that binds to DNA duplexes, through specific interactions in the major groove of the double helix.
  • An example of a route of administration of antisense nucleic acid molecules of the invention includes direct injection at a tissue site.
  • antisense nucleic acid molecules can be modified to target selected cells and then administered systemically.
  • antisense molecules can be modified such that they specifically bind to receptors or antigens expressed on a selected cell surface (e.g., by linking the antisense nucleic acid molecules to peptides or antibodies that bind to cell surface receptors or antigens).
  • the antisense nucleic acid molecules can also be delivered to cells using the vectors described herein.
  • vector constructs in which the antisense nucleic acid molecule is placed under the control of a strong pol II or pol III promoter are preferred.
  • the antisense nucleic acid molecule of the invention is an ⁇ -anomeric nucleic acid molecule.
  • An ⁇ -anomeric nucleic acid molecule forms specific double-stranded hybrids with complementary RNA in which, contrary to the usual ⁇ -units, the strands run parallel to each other. See, e.g., Gaultier, et al, 1987. Nucl. Acids Res. 15: 6625-6641.
  • the antisense nucleic acid molecule can also comprise a 2'-o-methylribonucleotide (See, e.g., Inoue, et al. 1987. Nucl. Acids Res. 15: 6131-6148) or a chimeric RNA-DNA analogue (See, e.g, Inoue, et al, 1987. FEBS Lett. 215: 327-330.
  • Nucleic acid modifications include, by way of non-limiting example, modified bases, and nucleic acids whose sugar phosphate backbones are modified or derivatized. These modifications are carried out at least in part to enhance the chemical stability of the modified nucleic acid, such that they may be used, for example, as antisense binding nucleic acids in therapeutic applications in a subject.
  • an antisense nucleic acid of the invention is a ribozyme.
  • Ribozymes are catalytic RNA molecules with ribonuclease activity that are capable of cleaving a single-stranded nucleic acid, such as an mRNA, to which they have a complementary region.
  • ribozymes e.g., hammerhead ribozymes as described in Haselhoff and Gerlach 1988. Nature 334: 585-591
  • a ribozyme having specificity for a NOVX-encoding nucleic acid can be designed based upon the nucleotide sequence of a NOVX cDNA disclosed herein (i.e., SEQ ID NO:2 «-l, wherein n is an integer between 1 and 141).
  • a derivative of a Tetrahymena L-19 IVS RNA can be constructed in which the nucleotide sequence of the active site is complementary to the nucleotide sequence to be cleaved in a NOVX-encoding mRNA. See, e.g., U.S. Patent 4,987,071 to Cech, et al. and U.S. Patent 5,116,742 to Cech, et al.
  • NOVX mRNA can also be used to select a catalytic RNA having a specific ribonuclease activity from a pool of RNA molecules. See, e.g., Bartel et al., (1993) Science 261:1411-1418.
  • NOVX gene expression can be inhibited by targeting nucleotide sequences complementary to the regulatory region of the NOVX nucleic acid (e.g., the NOVX promoter and/or enhancers) to form triple helical structures that prevent transcription of the NOVX gene in target cells.
  • nucleotide sequences complementary to the regulatory region of the NOVX nucleic acid e.g., the NOVX promoter and/or enhancers
  • the NOVX nucleic acids can be modified at the base moiety, sugar moiety or phosphate backbone to improve, e.g., the stability, hybridization, or solubility of the molecule.
  • the deoxyribose phosphate backbone of the nucleic acids can be modified to generate peptide nucleic acids. See, e.g., Hyrup, et al., 1996. BioorgMed Chem 4: 5-23.
  • peptide nucleic acids refer to nucleic acid mimics (e.g., DNA mimics) in which the deoxyribose phosphate backbone is replaced by a pseudopeptide backbone and only the four natural nucleotide bases are retained.
  • the neutral backbone of PNAs has been shown to allow for specific hybridization to DNA and RNA under conditions of low ionic strength.
  • the synthesis of PNA oligomer can be performed using standard solid phase peptide synthesis protocols as described in Hyrup, et al., 1996. supra; Perry-O'Keefe, et al., 1996. Proc. Natl. Acad. Sci. USA 93: 14670-14675.
  • PNAs of NOVX can be used in therapeutic and diagnostic applications.
  • PNAs can be used as antisense or antigene agents for sequence-specific modulation of gene expression by, e.g., inducing transcription or translation arrest or inhibiting replication.
  • PNAs of NOVX can also be used, for example, in the analysis of single base pair mutations in a gene (e.g., PNA directed PCR clamping; as artificial restriction enzymes when used in combination with other enzymes, e.g., Si nucleases (See, Hyrup, et ah, 1996.supra); or as probes or primers for DNA sequence and hybridization (See, Hyrup, et al., 1996, supra; Perry-O'Keefe, et al., 1996. supra).
  • PNAs of NOVX can be modified, e.g., to enhance their stability or cellular uptake, by attaching lipophilic or other helper groups to PNA, by the formation of PNA-DNA chimeras, or by the use of liposomes or other techniques of drug delivery known in the art.
  • PNA-DNA chimeras of NOVX can be generated that may combine the advantageous properties of PNA and DNA.
  • Such chimeras allow DNA recognition enzymes (e.g., RNase H and DNA polymerases) to interact with the DNA portion while the PNA portion would provide high binding affinity and specificity.
  • PNA-DNA chimeras can be linked using linkers of appropriate lengths selected in terms of base stacking, number of bonds between the nucleotide bases, and orientation (see, Hyrup, et al., 1996. supra).
  • the synthesis of PNA-DNA chimeras can be performed as described in Hyrup, et ah, 1996. supra and Finn, et ah, 1996. Nucl Acids Res 24: 3357-3363.
  • a DNA chain can be synthesized on a solid support using standard phosphoramidite coupling chemistry, and modified nucleoside analogs, e.g.,
  • 5'-(4-methoxytrityl)amino-5'-deoxy-thymidine phosphoramidite can be used between the PNA and the 5' end of DNA. See, e.g., Mag, et al, 1989. Nucl Acid Res 17: 5973-5988. PNA monomers are then coupled in a stepwise manner to produce a chimeric molecule with a 5' PNA segment and a 3' DNA segment. See, e.g., Finn, et al., 1996. supra. Alternatively, chimeric molecules can be synthesized with a 5' DNA segment and a 3' PNA segment. See, e.g., Petersen, et al., 1975. Bioorg. Med.
  • the oligonucleotide may include other appended groups such as peptides (e.g., for targeting host cell receptors in vivo), or agents facilitating transport across the cell membrane (see, e.g., Letsinger, et al., 1989. Proc. Natl. Acad. Sci. U.S.A. 86: 6553-6556; Lemairre, et al, ⁇ 9 7. Proc. Natl. Acad. Sci. 84: 648-652; PCT Publication No. WO88/09810) or the blood-brain barrier (see, e.g., PCT Publication No. WO 89/10134).
  • other appended groups such as peptides (e.g., for targeting host cell receptors in vivo), or agents facilitating transport across the cell membrane (see, e.g., Letsinger, et al., 1989. Proc. Natl. Acad. Sci. U.S.A. 86: 6553-
  • oligonucleotides can be modified with hybridization triggered cleavage agents (see, e.g., Krol, et al, 1988. BioTechniques 6:958-976) or intercalating agents (see, e.g., Zon, 1988. Pharm. Res. 5: 539-549).
  • the oligonucleotide may be conjugated to another molecule, e.g., a peptide, a hybridization triggered cross-linking agent, a transport agent, a hybridization-triggered cleavage agent, and the like.
  • a polypeptide according to the invention includes a polypeptide including the amino acid sequence of NOVX polypeptides whose sequences are provided in any one of SEQ ID NO:272, wherein n is an integer between 1 and 141.
  • the invention also includes a mutant or variant protein any of whose residues may be changed from the conesponding residues shown in any one of SEQ ID NO:2 «, wherein n is an integer between 1 and 141, while still encoding a protein that maintains its NOVX activities and physiological functions, or a functional fragment thereof.
  • a NOVX variant that preserves NOVX-like function includes any variant in which residues at a particular position in the sequence have been substituted by other amino acids, and further include the possibility of inserting an additional residue or residues between two residues of the parent protein as well as the possibility of deleting one or more residues from the parent sequence. Any amino acid substitution, insertion, or deletion is encompassed by the invention. In favorable circumstances, the substitution is a conservative substitution as defined above.
  • One aspect of the invention pertains to isolated NOVX proteins, and biologically-active portions thereof, or derivatives, fragments, analogs or homologs thereof. Also provided are polypeptide fragments suitable for use as immunogens to raise anti-NOVX antibodies.
  • native NOVX proteins can be isolated from cells or tissue sources by an appropriate purification scheme using standard protein purification techniques.
  • NOVX proteins are produced by recombinant DNA techniques.
  • a NOVX protein or polypeptide can be synthesized chemically using standard peptide synthesis techniques.
  • an “isolated” or “purified” polypeptide or protein or biologically-active portion thereof is substantially free of cellular material or other contaminating proteins from the cell or tissue source from which the NOVX protein is derived, or substantially free from chemical precursors or other chemicals when chemically synthesized.
  • substantially free of cellular material includes preparations of NOVX proteins in which the protein is separated from cellular components of the cells from which it is isolated or recombinantly-produced.
  • the language “substantially free of cellular material” includes preparations of NOVX proteins having less than about 30% (by dry weight) of non-NOVX proteins (also referred to herein as a "contaminating protein”), more preferably less than about 20% of non-NOVX proteins, still more preferably less than about 10% of non-NOVX proteins, and most preferably less than about 5% of non-NOVX proteins.
  • NOVX protein or biologically-active portion thereof When the NOVX protein or biologically-active portion thereof is recombinantly-produced, it is also preferably substantially free of culture medium, i.e., culture medium represents less than about 20%, more preferably less than about 10%, and most preferably less than about 5% of the volume of the NOVX protein preparation.
  • the language “substantially free of chemical precursors or other chemicals” includes preparations of NOVX proteins in which the protein is separated from chemical precursors or other chemicals that are involved in the synthesis of the protein.
  • the language “substantially free of chemical precursors or other chemicals” includes preparations of NOVX proteins having less than about 30% (by dry weight) of chemical precursors or non-NOVX chemicals, more preferably less than about 20% chemical precursors or non-NOVX chemicals, still more preferably less than about 10% chemical precursors or non-NOVX chemicals, and most preferably less than about 5% chemical precursors or non-NOVX chemicals.
  • Biologically-active portions of NOVX proteins include peptides comprising amino acid sequences sufficiently homologous to or derived from the amino acid sequences of the NOVX proteins (e.g., the amino acid sequence of SEQ ID NO:2 «, wherein n is an integer between 1 and 141) that include fewer amino acids than the full-length NOVX proteins, and exhibit at least one activity of a NOVX protein.
  • biologically-active portions comprise a domain or motif with at least one activity of the NOVX protein.
  • a biologically-active portion of a NOVX protein can be a polypeptide which is, for example, 10, 25, 50, 100 or more amino acid residues in length.
  • biologically-active portions in which other regions of the protein are deleted, can be prepared by recombinant techniques and evaluated for one or more of the functional activities of a native NOVX protein.
  • the NOVX protein has an amino acid sequence of SEQ ID NO:2«, wherein n is an integer between 1 and 141.
  • the NOVX protein is substantially homologous to SEQ ID NO:2«, wherein n is an integer between 1 and 141, and retains the functional activity of the protein of SEQ ID NO:2 «, wherein n is an integer between 1 and 141, yet differs in amino acid sequence due to natural allelic variation or mutagenesis, as described in detail, below.
  • the NOVX protein is a protein that comprises an amino acid sequence at least about 45% homologous to the amino acid sequence of SEQ ID NO:2 «, wherein n is an integer between 1 and 141, and retains the functional activity of the NOVX proteins of SEQ ID NO:2n, wherein n is an integer between 1 and 141.
  • sequences are aligned for optimal comparison purposes (e.g., gaps can be introduced in the sequence of a first amino acid or nucleic acid sequence for optimal alignment with a second amino or nucleic acid sequence).
  • the amino acid residues or nucleotides at corresponding amino acid positions or nucleotide positions are then compared.
  • amino acid or nucleic acid “homology” is equivalent to amino acid or nucleic acid “identity”
  • the nucleic acid sequence homology may be determined as the degree of identity between two sequences.
  • the homology may be determined using computer programs known in the art, such as GAP software provided in the GCG program package. See, Needleman and Wunsch, 1970. J Mol Biol 48: 443-453.
  • the coding region of the analogous nucleic acid sequences refened to above exhibits a degree of identity preferably of at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, or 99%, with the CDS (encoding) part of the DNA sequence of SEQ ID NO:2 «-l , wherein n is an integer between 1 and 141.
  • sequence identity refers to the degree to which two polynucleotide or polypeptide sequences are identical on a residue-by-residue basis over a particular region of comparison.
  • percentage of sequence identity is calculated by comparing two optimally aligned sequences over that region of comparison, determining the number of positions at which the identical nucleic acid base (e.g., A, T, C, G, U, or I, in the case of nucleic acids) occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the region of comparison (i.e., the window size), and multiplying the result by 100 to yield the percentage of sequence identity.
  • substantially identical denotes a characteristic of a polynucleotide sequence, wherein the polynucleotide comprises a sequence that has at least 80 percent sequence identity, preferably at least 85 percent identity and often 90 to 95 percent sequence identity, more usually at least 99 percent sequence identity as compared to a reference sequence over a comparison region.
  • the invention also provides NOVX chimeric or fusion proteins.
  • NOVX chimeric or fusion proteins.
  • NOVX "chimeric protein” or “fusion protein” comprises a NOVX polypeptide operatively-linked to a non-NOVX polypeptide.
  • An "NOVX polypeptide” refers to a polypeptide having an amino acid sequence corresponding to a NOVX protein of SEQ ID NO:2 «, wherein n is an integer between 1 and 141, whereas a “non-NOVX polypeptide” refers to a polypeptide having an amino acid sequence conesponding to a protein that is not substantially homologous to the NOVX protein, e.g., a protein that is different from the NOVX protein and that is derived from the same or a different organism.
  • a NOVX fusion protein comprises at least one biologically-active portion of a NOVX protein.
  • a NOVX fusion protein comprises at least two biologically-active portions of a NOVX protein.
  • a NOVX fusion protein comprises at least three biologically-active portions of a NOVX protein.
  • the term "operatively-linked" is intended to indicate that the NOVX polypeptide and the non-NOVX polypeptide are fused in-frame with one another.
  • the non-NOVX polypeptide can be fused to the N-terminus or C-terminus of the NOVX polypeptide.
  • the fusion protein is a GST-NO VX fusion protein in which the
  • NOVX sequences are fused to the C-terminus of the GST (glutathione S-transferase) sequences.
  • GST glutthione S-transferase
  • Such fusion proteins can facilitate the purification of recombinant NOVX polypeptides.
  • the fusion protein is a NOVX protein containing a heterologous signal sequence at its N-terminus.
  • NOVX a heterologous signal sequence at its N-terminus.
  • expression and or secretion of NOVX can be increased through use of a heterologous signal sequence.
  • the fusion protein is a NOVX-immunoglobulin fusion protein in which the NOVX sequences are fused to sequences derived from a member of the immunoglobulin protein family.
  • the NOVX-immunoglobulin fusion proteins of the invention can be inco ⁇ orated into pharmaceutical compositions and administered to a subject to inhibit an interaction between a NOVX ligand and a NOVX protein on the surface of a cell, to thereby suppress NOVX-mediated signal transduction in vivo.
  • the NOVX-immunoglobulin fusion proteins can be used to affect the bioavailability of a NOVX cognate ligand.
  • NOVX-immunoglobulin fusion proteins of the invention can be used as immunogens to produce anti-NOVX antibodies in a subject, to purify NOVX ligands, and in screening assays to identify molecules that inhibit the interaction of NOVX with a NOVX ligand.
  • a NOVX chimeric or fusion protein of the invention can be produced by standard recombinant DNA techniques.
  • DNA fragments coding for the different polypeptide sequences are ligated together in-frame in accordance with conventional techniques, e.g., by employing blunt-ended or stagger-ended termini for ligation, restriction enzyme digestion to provide for appropriate termini, filling-in of cohesive ends as appropriate, alkaline phosphatase treatment to avoid undesirable joining, and enzymatic ligation.
  • the fusion gene can be synthesized by conventional techniques including automated DNA synthesizers.
  • PCR amplification of gene fragments can be carried out using anchor primers that give rise to complementary overhangs between two consecutive gene fragments that can subsequently be annealed and reamplified to generate a chimeric gene sequence (see, e.g., Ausubel, et al. (eds.) CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, John Wiley & Sons, 1992).
  • anchor primers that give rise to complementary overhangs between two consecutive gene fragments that can subsequently be annealed and reamplified to generate a chimeric gene sequence
  • expression vectors are commercially available that already encode a fusion moiety (e.g., a GST polypeptide).
  • a NOVX-encoding nucleic acid can be cloned into such an expression vector such that the fusion moiety is linked in-frame to the NOVX protein.
  • NOVX Agonists and Antagonists The invention also pertains to variants of the NOVX proteins that function as either NOVX agonists (i.e., mimetics) or as NOVX antagonists.
  • Variants of the NOVX protein can be generated by mutagenesis (e.g., discrete point mutation or truncation of the NOVX protein).
  • An agonist of the NOVX protein can retain substantially the same, or a subset of, the biological activities of the naturally occurring form of the NOVX protein.
  • An antagonist of the NOVX protein can inhibit one or more of the activities of the naturally occurring form of the NOVX protein by, for example, competitively binding to a downstream or upstream member of a cellular signaling cascade which includes the NOVX protein.
  • treatment of a subject with a variant having a subset of the biological activities of the naturally occurring form of the protein has fewer side effects in a subject relative to treatment with the naturally occurring form of the NOVX proteins.
  • Variants of the NOVX proteins that function as either NOVX agonists (i.e., mimetics) or as NOVX antagonists can be identified by screening combinatorial libraries of mutants (e.g., truncation mutants) of the NOVX proteins for NOVX protein agonist or antagonist activity.
  • a variegated library of NOVX variants is generated by combinatorial mutagenesis at the nucleic acid level and is encoded by a variegated gene library.
  • a variegated library of NOVX variants can be produced by, for example, enzymatically ligating a mixture of synthetic oligonucleotides into gene sequences such that a degenerate set of potential NOVX sequences is expressible as individual polypeptides, or alternatively, as a set of larger fusion proteins (e.g., for phage display) containing the set of NOVX sequences therein.
  • a degenerate set of potential NOVX sequences is expressible as individual polypeptides, or alternatively, as a set of larger fusion proteins (e.g., for phage display) containing the set of NOVX sequences therein.
  • methods which can be used to produce libraries of potential NOVX variants from a degenerate oligonucleotide sequence. Chemical synthesis of a degenerate gene sequence can be performed in an automatic DNA synthesizer, and the synthetic gene then ligated into an appropriate expression vector.
  • degenerate set of genes allows for the provision, in one mixture, of all of the sequences encoding the desired set of potential NOVX sequences.
  • Methods for synthesizing degenerate oligonucleotides are well-known within the art. See, e.g., Narang, 1983. Tetrahedron 39: 3; Itakura, et al, 1984. Annu. Rev. Biochem. 53: 323; Itakura, et al, 1984. Science 198: 1056; Ike, et al, 1983. Nucl. Acids Res. 11: 477. Polypeptide Libraries
  • libraries of fragments of the NOVX protein coding sequences can be used to generate a variegated population of NOVX fragments for screening and subsequent selection of variants of a NOVX protein.
  • a library of coding sequence fragments can be generated by treating a double stranded PCR fragment of a NOVX coding sequence with a nuclease under conditions wherein nicking occurs only about once per molecule, denaturing the double sfranded DNA, renaturing the DNA to form double-stranded DNA that can include sense/antisense pairs from different nicked products, removing single stranded portions from reformed duplexes by treatment with Si nuclease, and ligating the resulting fragment library into an expression vector.
  • expression libraries can be derived which encodes N-terminal and internal fragments of various sizes of the NOVX proteins.
  • Recursive ensemble mutagenesis (REM), a new technique that enhances the frequency of functional mutants in the libraries, can be used in combination with the screening assays to identify NOVX variants. See, e.g., Arkin and Yourvan, 1992. Proc. Natl. Acad. Sci. USA 89: 7811-7815; Delgrave, et al, 1993. Protein Engineering 6:327-331.
  • antibody refers to immunoglobulin molecules and immunologically active portions of immunoglobulin (Ig) molecules, i.e., molecules that contain an antigen binding site that specifically binds (immunoreacts with) an antigen.
  • Ig immunoglobulin
  • Such antibodies include, but are not limited to, polyclonal, monoclonal, chimeric, single chain, F a , F a t > ' and F b- ⁇ fragments, and an F a expression library.
  • antibody molecules obtained from humans relates to any of the classes IgG, IgM, IgA, IgE and IgD, which differ from one another by the nature of the heavy chain present in the molecule.
  • the light chain may be a kappa chain or a lambda chain.
  • Reference herein to antibodies includes a reference to all such classes, subclasses and types of human antibody species .
  • An isolated protein of the invention intended to serve as an antigen, or a portion or fragment thereof, can be used as an immunogen to generate antibodies that immunospecifically bind the antigen, using standard techniques for polyclonal and monoclonal antibody preparation.
  • the full-length protein can be used or, alternatively, the invention provides antigenic peptide fragments of the antigen for use as immunogens.
  • An antigenic peptide fragment comprises at least 6 amino acid residues of the amino acid sequence of the full length protein, such as an amino acid sequence of SEQ ID NO:2 «, wherein n is an integer between 1 and 141, and encompasses an epitope thereof such that an antibody raised against the peptide forms a specific immune complex with the full length protein or with any fragment that contains the epitope.
  • the antigenic peptide comprises at least 10 amino acid residues, or at least 15 amino acid residues, or at least 20 amino acid residues, or at least 30 amino acid residues.
  • Prefened epitopes encompassed by the antigenic peptide are regions of the protein that are located on its surface; commonly these are hydrophilic regions.
  • at least one epitope encompassed by the antigenic peptide is a region of NOVX that is located on the surface of the protein, e.g., a hydrophilic region.
  • a hydrophobicity analysis of the human NOVX protein sequence will indicate which regions of a NOVX polypeptide are particularly hydrophilic and, therefore, are likely to encode surface residues useful for targeting antibody production.
  • hydropathy plots showing regions of hydrophilicity and hydrophobicity may be generated by any method well known in the art, including, for example, the Kyte Doolittle or the Hopp Woods methods, either with or without Fourier transformation. See, e.g., Hopp and Woods, 1981, Proc. Nat. Acad. Sci. USA 78: 3824-3828; Kyte and Doolittle 1982, J. Mol. Biol. 157: 105-142, each inco ⁇ orated herein by reference in their entirety.
  • Antibodies that are specific for one or more domains within an antigenic protein, or derivatives, fragments, analogs or homologs thereof, are also provided herein.
  • epitope includes any protein determinant capable of specific binding to an immunoglobulin or T-cell receptor.
  • Epitopic determinants usually consist of chemically active surface groupings of molecules such as amino acids or sugar side chains and usually have specific three dimensional structural characteristics, as well as specific charge characteristics.
  • a NOVX polypeptide or a fragment thereof comprises at least one antigenic epitope.
  • An anti-NOVX antibody of the present invention is said to specifically bind to antigen NOVX when the equilibrium binding constant (KQ) is ⁇ 1 ⁇ M, preferably ⁇ 100 nM, more preferably ⁇ 10 nM, and most preferably ⁇ 100 pM to about 1 pM, as measured by assays such as radioligand binding assays or similar assays known to those skilled in the art.
  • KQ equilibrium binding constant
  • a protein of the mvention, or a derivative, fragment, analog, homolog or ortholog thereof, may be utilized as an immunogen in the generation of antibodies that immunospecifically bind these protein components.
  • irnmunogenic preparation can contain, for example, the naturally occurring immunogenic protein, a chemically synthesized polypeptide representing the irnmunogenic protein, or a recombinantly expressed immunogenic protein.
  • the protein may be conjugated to a second protein known to be immunogenic in the mammal being immunized.
  • immunogenic proteins include but are not limited to keyhole limpet hemocyanin, serum albumin, bovine thyroglobulin, and soybean trypsin inhibitor.
  • the preparation can further include an adjuvant.
  • adjuvants used to increase the immunological response include, but are not limited to, Freund's (complete and incomplete), mineral gels (e.g., aluminum hydroxide), surface active substances (e.g., lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, dinifrophenol, etc.), adjuvants usable in humans such as Bacille Calmette-Guerin and Corynebacterium parvum, or similar immunostimulatory agents.
  • Additional examples of adjuvants which can be employed include MPL-TDM adjuvant (monophosphoryl Lipid A, synthetic trehalose dicorynomycolate).
  • the polyclonal antibody molecules directed against the immunogenic protein can be isolated from the mammal (e.g., from the blood) and further purified by well known techniques, such as affinity chromatography using protem A or protein G, which provide primarily the IgG fraction of immune serum. Subsequently, or alternatively, the specific antigen which is the target of the immunoglobulin sought, or an epitope thereof, may be immobilized on a column to purify the immune specific antibody by immunoaffinity chromatography. Purification of immunoglobulins is discussed, for example, by D. Wilkinson (The Engineer, published by The Engineer, Inc., Philadelphia PA, Vol. 14, No. 8 (April 17, 2000), pp.25-28). Monoclonal Antibodies
  • MAb monoclonal antibody
  • CDRs complementarity determining regions
  • MAbs thus contain an antigen binding site capable of immunoreacting with a particular epitope of the antigen characterized by a unique binding affinity for it.
  • Monoclonal antibodies can be prepared using hybridoma methods, such as those described by Kohler and Milstein, Nature, 256:495 (1975).
  • a mouse, hamster, or other appropriate host animal is typically immunized with an immunizing agent to elicit lymphocytes that produce or are capable of producing antibodies that will specifically bind to the immunizing agent.
  • the lymphocytes can be immunized in vitro.
  • the immunizing agent will typically include the protein antigen, a fragment thereof or a fusion protein thereof.
  • peripheral blood lymphocytes are used if cells of human origin are desired, or spleen cells or lymph node cells are used if non-human mammalian sources are desired.
  • the lymphocytes are then fused with an immortalized cell line using a suitable fusing agent, such as polyethylene glycol, to form a hybridoma cell (Goding, Monoclonal Antibodies: Principles and Practice, Academic Press, (1986) pp. 59-103).
  • Immortalized cell lines are usually transformed mammalian cells, particularly myeloma cells of rodent, bovine and human origin.
  • rat or mouse myeloma cell lines are employed.
  • the hybridoma cells can be cultured in a suitable culture medium that preferably contains one or more substances that inhibit the growth or survival of the unfused, immortalized cells.
  • a suitable culture medium that preferably contains one or more substances that inhibit the growth or survival of the unfused, immortalized cells.
  • the culture medium for the hybridomas typically will include hypoxanthine, aminopterin, and thymidine (“HAT medium”), which substances prevent the growth of HGPRT-deficient cells.
  • Preferred immortalized cell lines are those that fuse efficiently, support stable high level expression of antibody by the selected antibody-producing cells, and are sensitive to a medium such as HAT medium. More preferred immortalized cell lines are murine myeloma lines, which can be obtained, for instance, from the Salk Institute Cell
  • the culture medium in which the hybridoma cells are cultured can then be assayed for the presence of monoclonal antibodies directed against the antigen.
  • the binding specificity of monoclonal antibodies produced by the hybridoma cells is determined by immunoprecipitation or by an in vitro binding assay, such as radioimmunoassay (RIA) or enzyme-linked immunoabsorbent assay (ELISA).
  • RIA radioimmunoassay
  • ELISA enzyme-linked immunoabsorbent assay
  • the binding affinity of the monoclonal antibody can, for example, be determined by the Scatchard analysis of Munson and Pollard, Anal. Biochem., 107:220 (1980). It is an objective, especially important in therapeutic applications of monoclonal antibodies, to identify antibodies having a high degree of specificity and a high binding affinity for the target antigen.
  • the clones can be subcloned by limiting dilution procedures and grown by standard methods (Goding, 1986). Suitable culture media for this pu ⁇ ose include, for example, Dulbecco's Modified Eagle's Medium and RPMI-1640 medium. Alternatively, the hybridoma cells can be grown in vivo as ascites in a mammal.
  • the monoclonal antibodies secreted by the subclones can be isolated or purified from the culture medium or ascites fluid by conventional immunoglobulin purification procedures such as, for example, protein A-Sepharose, hydroxylapatite chromatography, gel electrophoresis, dialysis, or affinity chromatography.
  • the monoclonal antibodies can also be made by recombinant DNA methods, such as those described in U.S. Patent No.4,816,567.
  • DNA encoding the monoclonal antibodies of the invention can be readily isolated and sequenced using conventional procedures (e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of murine antibodies).
  • the hybridoma cells of the invention serve as a prefened source of such DNA.
  • the DNA can be placed into expression vectors, which are then transfected into host cells such as simian COS cells, Chinese hamster ovary (CHO) cells, or myeloma cells that do not otherwise produce immunoglobulin protein, to obtain the synthesis of monoclonal antibodies in the recombinant host cells.
  • host cells such as simian COS cells, Chinese hamster ovary (CHO) cells, or myeloma cells that do not otherwise produce immunoglobulin protein, to obtain the synthesis of monoclonal antibodies in the recombinant host cells.
  • the DNA also can be modified, for example, by substituting the coding sequence for human heavy and light chain constant domains in place of the homologous murine sequences (U.S.
  • a non-immunoglobulin polypeptide can be substituted for the constant domains of an antibody of the invention, or can be substituted for the variable domains of one antigen-combining site of an antibody of the invention to create a chimeric bivalent antibody.
  • the antibodies directed against the protein antigens of the invention can further comprise humanized antibodies or human antibodies. These antibodies are suitable for administration to humans without engendering an immune response by the human against the administered immunoglobulin.
  • Humanized forms of antibodies are chimeric immunoglobulins, immunoglobulin chains or fragments thereof (such as Fv, Fab, Fab', F(ab') 2 or other antigen-binding subsequences of antibodies) that are principally comprised of the sequence of a human immunoglobulin, and contain minimal sequence derived from a non-human immunoglobulin.
  • Humanization can be performed following the method of Winter and co-workers (Jones et al., Nature, 321:522-525 (1986); Riechmann et al., Nature, 332:323-327 (1988); Verhoeyen et al., Science, 239:1534-1536 (1988)), by substituting rodent CDRs or CDR sequences for the corresponding sequences of a human antibody. (See also U.S. Patent No. 5,225,539.) In some instances, Fv framework residues of the human immunoglobulin are replaced by conesponding non-human residues. Humanized antibodies can also comprise residues which are found neither in the recipient antibody nor in the imported CDR or framework sequences.
  • the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin and all or substantially all of the framework regions are those of a human immunoglobulin consensus sequence.
  • the humanized antibody optimally also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin (Jones et al., 1986; Riechmann et al., 1988; and Presta, Curr. Op. Struct. Biol., 2:593-596 (1992)).
  • Fc immunoglobulin constant region
  • Fully human antibodies essentially relate to antibody molecules in which the entire sequence of both the light chain and the heavy chain, including the CDRs, arise from human genes. Such antibodies are termed "human antibodies", or “fully human antibodies” herein.
  • Human monoclonal antibodies can be prepared by the trioma technique; the human B-cell hybridoma technique (see Kozbor, et al., 1983 Immunol Today 4: 72) and the EBV hybridoma technique to produce human monoclonal antibodies (see Cole, et al., 1985 In: MONOCLONAL ANTIBODIES AND CANCER THERAPY, Alan R. Liss, Inc., pp. 77-96).
  • Human monoclonal antibodies may be utilized in the practice of the present invention and may be produced by using human hybridomas (see Cote, et al., 1983. Proc Natl Acad Sci USA 80: 2026-2030) or by transforming human B-cells with Epstein Barr Virus in vitro (see Cole, et al., 1985 In: MONOCLONAL ANTIBODIES AND CANCER THERAPY, Alan R. Liss, Inc., pp. 77-96).
  • human antibodies can also be produced using additional techniques, including phage display libraries (Hoogenboom and Winter, J. Mol. Biol., 227:381 (1991); Marks et al., J. Mol.
  • human antibodies can be made by introducing human immunoglobulin loci into transgenic animals, e.g., mice in which the endogenous immunoglobulin genes have been partially or completely inactivated. Upon challenge, human antibody production is observed, which closely resembles that seen in humans in all respects, including gene rearrangement, assembly, and antibody repertoire.
  • transgenic animals e.g., mice in which the endogenous immunoglobulin genes have been partially or completely inactivated.
  • human antibody production is observed, which closely resembles that seen in humans in all respects, including gene rearrangement, assembly, and antibody repertoire.
  • This approach is described, for example, in U.S. Patent Nos. 5,545,807; 5,545,806; 5,569,825; 5,625,126; 5,633,425; 5,661,016, and in Marks et al. (Bio/Technology 10. 779-783 (1992)); Lonberg et al.
  • Human antibodies may additionally be produced using transgenic nonhuman animals which are modified so as to produce fully human antibodies rather than the animal's endogenous antibodies in response to challenge by an antigen.
  • transgenic nonhuman animals which are modified so as to produce fully human antibodies rather than the animal's endogenous antibodies in response to challenge by an antigen.
  • the endogenous genes encoding the heavy and light immunoglobulin chains in the nonhuman host have been incapacitated, and active loci encoding human heavy and light chain immunoglobulins are inserted into the host's genome.
  • the human genes are inco ⁇ orated, for example, using yeast artificial chromosomes containing the requisite human DNA segments. An animal which provides all the desired modifications is then obtained as progeny by crossbreeding intermediate transgenic animals containing fewer than the full complement of the modifications.
  • nonhuman animal is a mouse, and is termed the XenomouseTM as disclosed in PCT publications WO 96/33735 and WO 96/34096.
  • This animal produces B cells which secrete fully human immunoglobulins.
  • the antibodies can be obtained directly from the animal after immunization with an immunogen of interest, as, for example, a preparation of a polyclonal antibody, or alternatively from immortalized B cells derived from the animal, such as hybridomas producing monoclonal antibodies.
  • the genes encoding the immunoglobulins with human variable regions can be recovered and expressed to obtain the antibodies directly, or can be further modified to obtain analogs of antibodies such as, for example, single chain Fv molecules.
  • U.S. Patent No. 5,939,598 An example of a method of producing a nonhuman host, exemplified as a mouse, lacking expression of an endogenous immunoglobulin heavy chain is disclosed in U.S. Patent No. 5,939,598. It can be obtained by a method including deleting the J segment genes from at least one endogenous heavy chain locus in an embryonic stem cell to prevent rea ⁇ angement of the locus and to prevent formation of a transcript of a rearranged immunoglobulin heavy chain locus, the deletion being effected by a targeting vector containing a gene encoding a selectable marker; and producing from the embryonic stem cell a transgenic mouse whose somatic and germ cells contain the gene encoding the selectable marker.
  • a method for producing an antibody of interest such as a human antibody, is disclosed in U.S. Patent No. 5,916,771. It includes introducing an expression vector that contains a nucleotide sequence encoding a heavy chain into one mammalian host cell in culture, introducing an expression vector containing a nucleotide sequence encoding a light chain into another mammalian host cell, and fusing the two cells to form a hybrid cell.
  • the hybrid cell expresses an antibody containing the heavy chain and the light chain.
  • techniques can be adapted for the production of single-chain antibodies specific to an antigenic protein of the invention (see e.g., U.S. Patent No. 4,946,778).
  • methods can be adapted for the construction of F a expression libraries (see e.g., Huse, et al., 1989 Science 246: 1275-1281) to allow rapid and effective identification of monoclonal F ab fragments with the desired specificity for a protein or derivatives, fragments, analogs or homologs thereof.
  • Antibody fragments that contain the idiotypes to a protein antigen may be produced by techniques known in the art including, but not limited to: (i) an F( a y )2 fragment produced by pepsin digestion of an antibody molecule; (ii) an F ab fragment generated by reducing the disulfide bridges of an F( a ' ) 2 fragment; (iii) an F a b fragment generated by the treatment of the antibody molecule with papain and a reducing agent and (iv) F v fragments.
  • Bispecific antibodies are monoclonal, preferably human or humanized, antibodies that have binding specificities for at least two different antigens.
  • one of the binding specificities is for an antigenic protein of the invention.
  • the second binding target is any other antigen, and advantageously is a cell-surface protein or receptor or receptor subunit.
  • bispecific antibodies are known in the art. Traditionally, the recombinant production of bispecific antibodies is based on the co-expression of two immunoglobulin heavy-chain/light-chain pairs, where the two heavy chains have different specificities (Milstein and Cuello, Nature, 305:537-539 (1983)). Because of the random assortment of immunoglobulin heavy and light chains, these hybridomas (quadromas) produce a potential mixture often different antibody molecules, of which only one has the conect bispecific structure. The purification of the conect molecule is usually accomplished by affinity chromatography steps. Similar procedures are disclosed in WO 93/08829, published 13 May 1993, and in Traunecker et al., EMBO J., 10:3655-3659 (1991).
  • Antibody variable domains with the desired binding specificities can be fused to immunoglobulin constant domain sequences.
  • the fusion preferably is with an immunoglobulin heavy-chain constant domain, comprising at least part of the hinge, CH2, and CH3 regions. It is prefened,to have the first heavy-chain constant region (CHI) containing the site necessary for light-chain binding present in at least one of the fusions.
  • CHI first heavy-chain constant region
  • the interface between a pair of antibody molecules can be engineered to maximize the percentage of heterodimers which are recovered from recombinant cell culture.
  • the prefened interface comprises at least a part of the CH3 region of an antibody constant domain.
  • one or more small amino acid side chains from the interface of the first antibody molecule are replaced with larger side chains (e.g. tyrosine or tryptophan).
  • Compensatory "cavities" of identical or similar size to the large side chain(s) are created on the interface of the second antibody molecule by replacing large amino acid side chains with smaller ones (e.g. alanine or threonine). This provides a mechanism for increasing the yield of the heterodimer over other unwanted end-products such as homodimers.
  • Bispecific antibodies can be prepared as full length antibodies or antibody fragments (e.g. F(ab') 2 bispecific antibodies). Techniques for generating bispecific antibodies from antibody fragments have been described in the literature. For example, bispecific antibodies can be prepared using chemical linkage. Brennan et al., Science
  • Fab' fragments can be directly recovered from E. coli and chemically coupled to form bispecific antibodies.
  • Shalaby et al., J. Exp. Med. 175:217-225 (1992) describe the production of a fully humanized bispecific antibody F(ab') 2 molecule.
  • Each Fab' fragment was separately secreted from E. coli and subjected to directed chemical coupling in vitro to form the bispecific antibody.
  • the bispecific antibody thus formed was able to bind to cells overexpressing the ErbB2 receptor and normal human T cells, as well as trigger the lytic activity of human cytotoxic lymphocytes against human breast tumor targets.
  • bispecific antibodies have been produced using leucine zippers.
  • the leucine zipper peptides from the Fos and Jun proteins were linked to the Fab' portions of two different antibodies by gene fusion.
  • the antibody homodimers were reduced at the hinge region to form monomers and then re-oxidized to form the antibody heterodimers. This method can also be utilized for the production of antibody homodimers.
  • the fragments comprise a heavy-chain variable domain (V H ) connected to a light-chain variable domain (V L ) by a linker which is too short to allow pairing between the two domains on the same chain. Accordingly, the V H and V L domains of one fragment are forced to pair with the complementary V and V H domains of another fragment, thereby forming two antigen-binding sites.
  • V H and V L domains of one fragment are forced to pair with the complementary V and V H domains of another fragment, thereby forming two antigen-binding sites.
  • sFv single-chain Fv
  • Another strategy for making bispecific antibody fragments by the use of single-chain Fv (sFv) dimers has also been reported. See, Gruber et al., J. Immunol. 152:5368 (1994). Antibodies with more than two valencies are contemplated. For example, trispecific antibodies can be prepared. Tutt et al., J. Immunol. 147:60 (1991).
  • bispecific antibodies can bind to two different epitopes, at least one of which originates in the protein antigen of the invention.
  • an anti-antigenic arm of an immunoglobulin molecule can be combined with an arm which binds to a triggering molecule on a leukocyte such as a T-cell receptor molecule (e.g. CD2, CD3, CD28, or B7), or Fc receptors for IgG (Fc ⁇ R), such as Fc ⁇ RI (CD64), Fc ⁇ RII (CD32) and Fc ⁇ RIII (CD 16) so as to focus cellular defense mechanisms to the cell expressing the particular antigen.
  • Bispecific antibodies can also be used to direct cytotoxic agents to cells which express a particular antigen.
  • antibodies possess an antigen-binding arm and an arm which binds a cytotoxic agent or a radionuclide chelator, such as EOTUBE, DPTA, DOTA, or TETA.
  • a cytotoxic agent or a radionuclide chelator such as EOTUBE, DPTA, DOTA, or TETA.
  • Another bispecific antibody of interest binds the protein antigen described herein and further binds tissue factor (TF).
  • Heteroconjugate antibodies are also within the scope of the present invention.
  • Heteroconjugate antibodies are composed of two covalently joined antibodies. Such antibodies have, for example, been proposed to target immune system cells to unwanted cells (U.S. Patent No. 4,676,980), and for treatment of HTV infection (WO 91/00360; WO 92/200373; EP 03089).
  • the antibodies can be prepared in vitro using known methods in synthetic protein chemistry, including those involving crosslinking agents.
  • immunotoxins can be constructed using a disulfide exchange reaction or by forming a thioether bond. Examples of suitable reagents for this pu ⁇ ose include iminothiolate and methyl-4-mercaptobutyrimidate and those disclosed, for example, in U.S. Patent No. 4,676,980.
  • cysteine residue(s) can be introduced into the Fc region, thereby allowing interchain disulfide bond formation in this region.
  • the homodimeric antibody thus generated can have improved internalization capability and/or increased complement-mediated cell killing and antibody-dependent cellular cytotoxicity (ADCC). See Caron et al., J. Exp Med share 176: 1191-1195 (1992) and Shopes, J. Immunol., 148:
  • Homodimeric antibodies with enhanced anti-tumor activity can also be prepared using heterobifunctional cross-linkers as described in Wolff et al. Cancer Research, 53: 2560-2565 (1993).
  • an antibody can be engineered that has dual Fc regions and can thereby have enhanced complement lysis and ADCC capabilities. See Stevenson et al., Anti-Cancer Drug Design, 3: 219-230 (1989).
  • the invention also pertains to immunoconjugates comprising an antibody conjugated to a cytotoxic agent such as a chemotherapeutic agent, toxin (e.g., an enzymatically active toxin of bacterial, fungal, plant, or animal origin, or fragments thereof), or a radioactive isotope (i.e., a radioconjugate).
  • a cytotoxic agent such as a chemotherapeutic agent, toxin (e.g., an enzymatically active toxin of bacterial, fungal, plant, or animal origin, or fragments thereof), or a radioactive isotope (i.e., a radioconjugate).
  • Enzymatically active toxins and fragments thereof that can be used include diphtheria A chain, nonbinding active fragments of diphtheria toxin, exotoxin A chain (from Pseudomonas aeruginosa), ricin A chain, abrin A chain, modeccin A chain, alpha-sarcin, Aleurites fordii proteins, dianthin proteins, Phytolaca americana proteins (PAPI, PAPII, and PAP-S), momordica charantia inhibitor, curcin, crotin, sapaonaria officinalis inhibitor, gelonin, mitogellin, restrictocin, phenomycin, enomycin, and the tricothecenes.
  • radionuclides are available for the production of radioconjugated antibodies. Examples include 212 Bi, 131 1, 131 In, 90 Y, and 186 Re. Conjugates of the antibody and cytotoxic agent are made using a variety of bifunctional protein-coupling agents such as N-succinimidyl-3-(2-pyridyldithiol) propionate (SPDP), iminothiolane (IT), bifunctional derivatives of imidoesters (such as dimethyl adipimidate HCL), active esters (such as disuccinimidyl suberate), aldehydes (such as glutareldehyde), bis-azido compounds (such as bis (p-azidobenzoyl) hexanediamine), bis-diazonium derivatives (such as bis-(p-diazoniumbenzoyl)-ethylenediamine), diisocyanates (such as tolyene 2,6-d ⁇ socyanate), and bis--coup
  • a ricin immunotoxin can be prepared as described in Vitetta et al., Science, 238: 1098 (1987).
  • Carbon-14-labeled l-isothiocyanatobenzyl-3-methyldiethylene triaminepentaacetic acid (MX-DTPA) is an exemplary chelating agent for conjugation of radionucleotide to the antibody. See WO94/11026.
  • the antibody in another embodiment, can be conjugated to a "receptor" (such streptavidin) for utilization in tumor pretargeting wherein the antibody-receptor conjugate is administered to the patient, followed by removal of unbound conjugate from the circulation using a clearing agent and then administration of a "ligand” (e.g., avidin) that is in turn conjugated to a cytotoxic agent.
  • a "receptor” such streptavidin
  • a "ligand” e.g., avidin
  • the antibodies disclosed herein can also be formulated as immunoliposomes.
  • Liposomes containing the antibody are prepared by methods known in the art, such as described in Epstein et al., Proc. Natl. Acad. Sci. USA. 82: 3688 (1985); Hwang et al., Proc. Natl Acad. Sci. USA. 77: 4030 (1980); and U.S. Pat. Nos. 4,485,045 and 4,544,545. Liposomes with enhanced circulation time are disclosed in U.S. Patent No. 5,013,556.
  • Particularly useful liposomes can be generated by the reverse-phase evaporation method with a lipid composition comprising phosphatidylcholine, cholesterol, and PEG-derivatized phosphatidylethanolamine (PEG-PE). Liposomes are extruded through filters of defined pore size to yield liposomes with the desired diameter.
  • Fab' fragments of the antibody of the present invention can be conjugated to the liposomes as described in Martin et al ., J. Biol. Chem., 257: 286-288 (1982) via a disulfide-interchange reaction.
  • a chemotherapeutic agent such as Doxorubicin
  • Antibodies directed against a protein of the invention may be used in methods known within the art relating to the localization and/or quantitation of the protein (e.g., for use in measuring levels of the protein within appropriate physiological samples, for use in diagnostic methods, for use in imaging the protein, and the like).
  • antibodies against the proteins, or derivatives, fragments, analogs or homologs thereof, that contain the antigen binding domain are utilized as pharmacologically-active compounds (see below).
  • An antibody specific for a protein of the invention can be used to isolate the protein by standard techniques, such as immunoaffinity chromatography or immunoprecipitation. Such an antibody can facilitate the purification of the natural protein antigen from cells and of recombinantly produced antigen expressed in host cells. Moreover, such an antibody can be used to detect the antigenic protein (e.g., in a cellular lysate or cell supernatant) in order to evaluate the abundance and pattern of expression of the antigenic protein. Antibodies directed against the protein can be used diagnostically to monitor protein levels in tissue as part of a clinical testing procedure, e.g., to, for example, determine the efficacy of a given treatment regimen.
  • Detection can be facilitated by coupling (i.e., physically linking) the antibody to a detectable substance.
  • detectable substances include various enzymes, prosthetic groups, fluorescent materials, luminescent materials, bioluminescent materials, and radioactive materials.
  • suitable enzymes include horseradish peroxidase, alkaline phosphatase, ⁇ -galactosidase, or acetylcholinesterase;
  • suitable prosthetic group complexes include sfreptavidin/biotin and avidin/biotin;
  • suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride orphycoerythrin;
  • an example of a luminescent material includes luminol;
  • bioluminescent materials include luciferase, luciferin, and aequorin, and examples of suitable radioactive material include 125
  • Antibodies of the invention may be used as therapeutic agents. Such agents will generally be employed to treat or prevent a disease or pathology in a subject.
  • An antibody preparation preferably one having high specificity and high affinity for its target antigen, is administered to the subject and will generally have an effect due to its binding with the target.
  • Such an effect may be one of two kinds, depending on the specific nature of the interaction between the given antibody molecule and the target antigen in question.
  • administration of the antibody may abrogate or inhibit the binding of the target with an endogenous ligand to which it naturally binds.
  • the antibody binds to the target and masks a binding site of the naturally occurring ligand, wherein the ligand serves as an effector molecule.
  • the receptor mediates a signal transduction pathway for which ligand is responsible.
  • the effect may be one in which the antibody elicits a physiological result by virtue of binding to an effector binding site on the target molecule.
  • the target a receptor having an endogenous ligand which may be absent or defective in the disease or pathology, binds the antibody as a sunogate effector ligand, initiating a receptor-based signal transduction event by the receptor.
  • a therapeutically effective amount of an antibody of the invention relates generally to the amount needed to achieve a therapeutic objective. As noted above, this may be a binding interaction between the antibody and its target antigen that, in certain cases, interferes with the functioning of the target, and in other cases, promotes a physiological response.
  • the amount required to be administered will furthermore depend on the binding affinity of the antibody for its specific antigen, and will also depend on the rate at which an administered antibody is depleted from the free volume other subject to which it is administered.
  • Common ranges for therapeutically effective dosing of an antibody or antibody fragment of the invention may be, by way of nonlimiting example, from about 0.1 mg/kg body weight to about 50 mg/kg body weight. Common dosing frequencies may range, for example, from twice daily to once a week.
  • Antibodies specifically binding a protein of the invention, as well as other molecules identified by the screening assays disclosed herein, can be administered for the treatment of various disorders in the form of pharmaceutical compositions.
  • Principles and considerations involved in preparing such compositions, as well as guidance in the choice of components are provided, for example, in Remington : The Science And Practice Of Pharmacy 19th ed. (Alfonso R. Gennaro, et al., editors) Mack Pub. Co., Easton, Pa. : 1995; Drug Abso ⁇ tion Enhancement : Concepts, Possibilities, Limitations, And Trends, Harwood Academic Publishers, Langhorne, Pa., 1994; and Peptide And Protein Drug Delivery (Advances In Parenteral Sciences, Vol.4), 1991, M.
  • antigenic protein is intracellular and whole antibodies are used as inhibitors, internalizing antibodies are prefened.
  • liposomes can also be used to deliver the antibody, or an antibody fragment, into cells. Where antibody fragments are used, the smallest inhibitory fragment that specifically binds to the binding domain of the target protein is prefened.
  • peptide molecules can be designed that retain the ability to bind the target protein sequence. Such peptides can be synthesized chemically and/or produced by recombinant DNA technology. See, e.g., Marasco et al., Proc. Natl. Acad. Sci. USA, 90: 7889-7893 (1993).
  • the formulation herein can also contain more than one active compound as necessary for the particular indication being freated, preferably those with complementary activities that do not adversely affect each other.
  • the composition can comprise an agent that enhances its function, such as, for example, a cytotoxic agent, cytokine, chemotherapeutic agent, or growth-inhibitory agent.
  • cytotoxic agent such as, for example, a cytotoxic agent, cytokine, chemotherapeutic agent, or growth-inhibitory agent.
  • Such molecules are suitably present in combination in amounts that are effective for the pu ⁇ ose intended.
  • the active ingredients can also be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly-(methylmethacrylate) microcapsules, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles, and nanocapsules) or in macroemulsions.
  • colloidal drug delivery systems for example, liposomes, albumin microspheres, microemulsions, nano-particles, and nanocapsules
  • the formulations to be used for in vivo administration must be sterile. This is readily accomplished by filtration through sterile filtration membranes.
  • sustained-release preparations can be prepared. Suitable examples of sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing the antibody, which matrices are in the form of shaped articles, e.g., films, or microcapsules. Examples of sustained-release matrices include polyesters, hydrogels (for example, poly(2-hydroxyethyl-methacrylate), or poly(vinylalcohol)), polylactides (U.S. Pat. No.
  • copolymers of L-glutamic acid and ⁇ ethyl-L-glutamate copolymers of L-glutamic acid and ⁇ ethyl-L-glutamate, non-degradable ethylene-vinyl acetate, degradable lactic acid-glycolic acid copolymers such as the LUPRON DEPOT TM (injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate), and poly-D-(-)-3-hydroxybutyric acid. While polymers such as ethylene-vinyl acetate and lactic acid-glycolic acid enable release of molecules for over 100 days, certain hydrogels release proteins for shorter time periods.
  • An agent for detecting an analyte protein is an antibody capable of binding to an analyte protein, preferably an antibody with a detectable label.
  • Antibodies can be polyclonal, or more preferably, monoclonal. An intact antibody, or a fragment thereof (e.g., F ab or F( ab ) 2 ) can be used.
  • the term "labeled", with regard to the probe or antibody, is intended to encompass direct labeling of the probe or antibody by coupling (i.e., physically linking) a detectable substance to the probe or antibody, as well as indirect labeling of the probe or antibody by reactivity with another reagent that is directly labeled.
  • biological sample is intended to include tissues, cells and biological fluids isolated from a subject, as well as tissues, cells and fluids present within a subject. Included within the usage of the term "biological sample”, therefore, is blood and a fraction or component of blood including blood serum, blood plasma, or lymph. That is, the detection method of the invention can be used to detect an analyte mRNA, protein, or genomic DNA in a biological sample in vitro as well as in vivo.
  • in vitro techniques for detection of an analyte mRNA include Northern hybridizations and in situ hybridizations.
  • In vitro techniques for detection of an analyte protein include enzyme linked immunosorbent assays (ELISAs), Western blots, immunoprecipitations, and immunofluorescence.
  • In vitro techniques for detection of an analyte genomic DNA include Southern hybridizations. Procedures for conducting immunoassays are described, for example in "ELISA: Theory and Practice: Methods in Molecular Biology", Vol. 42, J. R. Crowther (Ed.) Human Press, Totowa, NJ, 1995; "Immunoassay", E. Diamandis and T.
  • analyte protein in vivo techniques for detection of an analyte protein include introducing into a subject a labeled anti-an analyte protein antibody.
  • the antibody can be labeled with a radioactive marker whose presence and location in a subject can be detected by standard imaging techniques.
  • vectors preferably expression vectors, containing a nucleic acid encoding a NOVX protein, or derivatives, fragments, analogs or homologs thereof.
  • vector refers to a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked.
  • plasmid refers to a circular double stranded DNA loop into which additional DNA segments can be ligated.
  • viral vector is another type of vector, wherein additional DNA segments can be ligated into the viral genome.
  • vectors are capable of autonomous replication in a host cell into which they are introduced (e.g., bacterial vectors having a bacterial origin of replication and episomal mammalian vectors).
  • Other vectors e.g., non-episomal mammalian vectors
  • certain vectors are capable of directing the expression of genes to which they are operatively-linked. Such vectors are refened to herein as "expression vectors".
  • useful expression vectors in recombinant DNA techniques are often in the form of plasmids.
  • plasmid and "vector” can be used interchangeably as the plasmid is the most commonly used form of vector.
  • the invention is intended to include such other forms of expression vectors, such as viral vectors (e.g., replication defective refroviruses, adenoviruses and adeno-associated viruses), which serve equivalent functions.
  • viral vectors e.g., replication defective refroviruses, adenoviruses and adeno-associated viruses
  • the recombinant expression vectors of the invention comprise a nucleic acid of the invention in a form suitable for expression of the nucleic acid in a host cell, which means that the recombinant expression vectors include one or more regulatory sequences, selected on the basis of the host cells to be used for expression, that is operatively-linked to the nucleic acid sequence to be expressed.
  • "operably-linked" is intended to mean that the nucleotide sequence of interest is linked to the regulatory sequence(s) in a manner that allows for expression of the nucleotide sequence (e.g., in an in vitro transcription/translation system or in a host cell when the vector is introduced into the host cell).
  • regulatory sequence is intended to includes promoters, enhancers and other expression control elements (e.g., polyadenylation signals). Such regulatory sequences are described, for example, in Goeddel, GENE EXPRESSION TECHNOLOGY: METHODS IN ENZYMOLOGY 185, Academic Press, San Diego, Calif. (1990). Regulatory sequences include those that direct constitutive expression of a nucleotide sequence in many types of host cell and those that direct expression of the nucleotide sequence only in certain host cells (e.g., tissue-specific regulatory sequences). It will be appreciated by those skilled in the art that the design of the expression vector can depend on such factors as the choice of the host cell to be transformed, the level of expression of protein desired, etc.
  • the expression vectors of the invention can be introduced into host cells to thereby produce proteins or peptides, including fusion proteins or peptides, encoded by nucleic acids as described herein (e.g., NOVX proteins, mutant forms of NOVX proteins, fusion proteins, etc.).
  • the recombinant expression vectors of the invention can be designed for expression of NOVX proteins in prokaryotic or eukaryotic cells.
  • NOVX proteins can be expressed in bacterial cells such as Escherichia coli, insect cells (using baculovirus expression vectors) yeast cells or mammalian cells. Suitable host cells are discussed further in Goeddel, GENE EXPRESSION TECHNOLOGY: METHODS IN
  • the recombinant expression vector can be transcribed and translated in vitro, for example using T7 promoter regulatory sequences and T7 polymerase.
  • Fusion vectors add a number of amino acids to a protein encoded therein, usually to the amino terminus of the recombinant protein.
  • Such fusion vectors typically serve three pu ⁇ oses: (i) to increase expression of recombinant protein; (ii) to increase the solubility of the recombinant protein; and (iii) to aid in the purification of the recombinant protein by acting as a ligand in affinity purification.
  • a proteolytic cleavage site is introduced at the junction of the fusion moiety and the recombinant protein to enable separation of the recombinant protein from the fusion moiety subsequent to purification of the fusion protein.
  • enzymes, and their cognate recognition sequences include Factor Xa, thrombin and enterokinase.
  • Typical fusion expression vectors include pGEX (Pharmacia Biotech Inc; Smith and Johnson, 1988. Ge «e 67: 31-40), pMAL (New England Biolabs, Beverly, Mass.) and pRIT5 (Pharmacia, Piscataway, N. J.) that fuse glutathione S-fransferase (GST), maltose E binding protein, or protein A, respectively, to the target recombinant protein.
  • E. coli expression vectors examples include pTrc (Amrann et al, (1988) Gene 69:301-315) and pET lid (Studier et al, GENE EXPRESSION TECHNOLOGY: METHODS IN ENZYMOLOGY 185, Academic Press, San Diego, Calif. (1990) 60-89).
  • One strategy to maximize recombinant protem expression in E. coli is to express the protein in a host bacteria with an impaired capacity to proteolytically cleave the recombinant protein. See, e.g., Gottesman, GENE EXPRESSION TECHNOLOGY: METHODS IN ENZYMOLOGY 185, Academic Press, San Diego, Calif. (1990) 119-128.
  • Another strategy is to alter the nucleic acid sequence of the nucleic acid to be inserted into an expression vector so that the individual codons for each amino acid are those preferentially utilized in E. coli (see, e.g., Wada, et al, 1992. Nucl. Acids Res. 20: 2111-2118). Such alteration of nucleic acid sequences of the invention can be carried out by standard DNA synthesis techniques.
  • the NOVX expression vector is a yeast expression vector.
  • yeast expression vectors for expression in yeast Saccharomyces cerivisae include pYepSecl (Baldari, et al, 1987. EMBOJ. 6: 229-234), pMFa (Kurjan and Herskowitz, 1982. Cell 30: 933-943), pJRY88 (Schultz et al, 1987. Gene 54: 113-123), pYES2 (Invitrogen Co ⁇ oration, San Diego, Calif.), and picZ (InVitrogen Co ⁇ , San Diego, Calif.).
  • NOVX can be expressed in insect cells using baculovirus expression vectors.
  • Baculovirus vectors available for expression of proteins in cultured insect cells include the pAc series (Smith, et al, 1983. Mol. Cell. Biol. 3: 2156-2165) and the pVL series (Lucklow and Summers, 1989. Virology 170: 31-39).
  • a nucleic acid of the invention is expressed in mammalian cells using a mammalian expression vector. Examples of mammalian expression vectors include pCDM8 (Seed, 1987. Nature 329: 840) and ⁇ MT2PC (Kaufman, et al, 1987. EMBO J.
  • the expression vector's control functions are often provided by viral regulatory elements.
  • promoters are derived from polyoma, adenovirus 2, cytomegalovirus, and simian virus 40.
  • suitable expression systems for both prokaryotic and eukaryotic cells see, e.g., Chapters 16 and 17 of Sambrook, et al, MOLECULAR CLONING: A LABORATORY MANUAL. 2nd ed., Cold Spring Harbor Laboratory, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1989.
  • the recombinant mammalian expression vector is capable of directing expression of the nucleic acid preferentially in a particular cell type (e.g., tissue-specific regulatory elements are used to express the nucleic acid).
  • tissue-specific regulatory elements are known in the art.
  • suitable tissue-specific promoters include the albumin promoter (liver-specific; Pinkert, et al, 1987. Genes Dev. 1: 268-277), lymphoid-specific promoters (Calame and Eaton, 1988. Adv. Immunol. 43: 235-275), in particular promoters of T cell receptors (Winoto and Baltimore, 1989. EMBO J.
  • promoters are also encompassed, e.g., the murine hox promoters (Kessel and Grass, 1990. Science 249: 374-379) and the -fetoprotein promoter (Ca pes and Tilghman, 1989. Genes Dev. 3: 537-546).
  • the invention further provides a recombinant expression vector comprising a DNA molecule of the invention cloned into the expression vector in an antisense orientation. That is, the DNA molecule is operatively-linked to a regulatory sequence in a manner that allows for expression (by transcription of the DNA molecule) of an RNA molecule that is antisense to NOVX mRNA.
  • Regulatory sequences operatively linked to a nucleic acid cloned in the antisense orientation can be chosen that direct the continuous expression of the antisense RNA molecule in a variety of cell types, for instance viral promoters and/or enhancers, or regulatory sequences can be chosen that direct constitutive, tissue specific or cell type specific expression of antisense RNA.
  • the antisense expression vector can be in the form of a recombinant plasmid, phagemid or attenuated virus in which antisense nucleic acids are produced under the control of a high efficiency regulatory region, the activity of which can be determined by the cell type into which the vector is introduced.
  • a high efficiency regulatory region the activity of which can be determined by the cell type into which the vector is introduced.
  • a host cell can be any prokaryotic or eukaryotic cell.
  • NOVX protein can be expressed in bacterial cells such as E. coli, insect cells, yeast or mammalian cells (such as Chinese hamster ovary cells (CHO) or COS cells).
  • bacterial cells such as E. coli, insect cells, yeast or mammalian cells (such as Chinese hamster ovary cells (CHO) or COS cells).
  • mammalian cells such as Chinese hamster ovary cells (CHO) or COS cells.
  • Other suitable host cells are known to those skilled in the art.
  • Vector DNA can be introduced into prokaryotic or eukaryotic cells via conventional transformation or transfection techniques.
  • transformation and “transfection” are intended to refer to a variety of art-recognized techniques for introducing foreign nucleic acid (e.g. , DNA) into a host cell, including calcium phosphate or calcium chloride co-precipitation, DEAE-dexfran-mediated transfection, lipofection, or electroporation. Suitable methods for transforming or transfecting host cells can be found in Sambrook, et al. (MOLECULAR CLONING: A LABORATORY MANUAL. 2nd ed., Cold Spring Harbor Laboratory, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1989), and other laboratory manuals.
  • a gene that encodes a selectable marker (e.g., resistance to antibiotics) is generally introduced into the host cells along with the gene of interest.
  • selectable markers include those that confer resistance to drugs, such as G418, hygromycin and methotrexate.
  • Nucleic acid encoding a selectable marker can be introduced into a host cell on the same vector as that encoding NOVX or can be introduced on a separate vector. Cells stably transfected with the introduced nucleic acid can be identified by drug selection (e.g., cells that have inco ⁇ orated the selectable marker gene will survive, while the other cells die).
  • a host cell of the invention such as a prokaryotic or eukaryotic host cell in culture, can be used to produce (i.e., express) NOVX protein.
  • the invention further provides methods for producing NOVX protein using the host cells of the invention.
  • the method comprises culturing the host cell of invention (into which a recombinant expression vector encoding NOVX protein has been introduced) in a suitable medium such that NOVX protein is produced.
  • the method further comprises isolating NOVX protein from the medium or the host cell.
  • the host cells of the invention can also be used to produce non-human transgenic animals.
  • a host cell of the invention is a fertilized oocyte or an embryonic stem cell into which NOVX protein-coding sequences have been infroduced.
  • Such host cells can then be used to create non-human transgenic animals in which exogenous NOVX sequences have been introduced into their genome or homologous recombinant animals in which endogenous NOVX sequences have been altered.
  • Such animals are useful for studying the function and/or activity of NOVX protein and for identifying and/or evaluating modulators of NOVX protein activity.
  • a "transgenic animal” is a non-human animal, preferably a mammal, more preferably a rodent such as a rat or mouse, in which one or more of the cells of the animal includes a transgene.
  • Other examples of transgenic animals include non-human primates, sheep, dogs, cows, goats, chickens, amphibians, etc.
  • a transgene is exogenous DNA that is integrated into the genome of a cell from which a transgenic animal develops and that remains in the genome of the mature animal, thereby directing the expression of an encoded gene product in one or more cell types or tissues of the transgenic animal.
  • a "homologous recombinant animal” is a non-human animal, preferably a mammal, more preferably a mouse, in which an endogenous NOVX gene has been altered by homologous recombination between the endogenous gene and an exogenous DNA molecule introduced into a cell of the animal, e.g., an embryonic cell of the animal, prior to development of the animal.
  • a transgenic animal of the invention can be created by introducing NOVX-encoding nucleic acid into the male pronuclei of a fertilized oocyte (e.g., by microinjection, retroviral infection) and allowing the oocyte to develop in a pseudopregnant female foster animal.
  • the human NOVX cDNA sequences i.e., any one of SEQ ID NOS:2n-l, wherein n is an integer between 1 and 141, can be introduced as a transgene into the genome of a non-human animal.
  • a non-human homologue of the human NOVX gene such as a mouse NOVX gene
  • a non-human homologue of the human NOVX gene can be isolated based on hybridization to the human NOVX cDNA (described further supra) and used as a transgene.
  • Intronic sequences and polyadenylation signals can also be included in the transgene to increase the efficiency of expression of the fransgene.
  • a tissue-specific regulatory sequence(s) can be operably-linked to the NOVX transgene to direct expression of NOVX protein to particular cells.
  • transgenic founder animal can be identified based upon the presence of the NOVX fransgene in its genome and/or expression of NOVX mRNA in tissues or cells of the animals. A transgenic founder animal can then be used to breed additional animals carrying the transgene. Moreover, transgenic animals carrying a transgene-encoding NOVX protein can further be bred to other transgenic animals carrying other transgenes.
  • a vector which contains at least a portion of a NOVX gene into which a deletion, addition or substitution has been infroduced to thereby alter, e.g., functionally disrupt, the NOVX gene.
  • the NOVX gene can be a human gene (e.g., the cDNA of any one of SEQ ID NOS:2n-l , wherein n is an integer between 1 and 141), but more preferably, is a non-human homologue of a human NOVX gene.
  • a mouse homologue of human NOVX gene of SEQ ID NOS:2n-l can be used to construct a homologous recombination vector suitable for altering an endogenous NOVX gene in the mouse genome.
  • the vector is designed such that, upon homologous recombination, the endogenous NOVX gene is functionally disrupted (i.e., no longer encodes a functional protein; also referred to as a "knock out" vector).
  • the vector can be designed such that, upon homologous recombination, the endogenous NOVX gene is mutated or otherwise altered but still encodes functional protein (e.g., the upstream regulatory region can be altered to thereby alter the expression of the endogenous NOVX protein).
  • the altered portion of the NOVX gene is flanked at its 5'- and 3'-termini by additional nucleic acid of the NOVX gene to allow for homologous recombination to occur between the exogenous NOVX gene carried by the vector and an endogenous NOVX gene in an embryonic stem cell.
  • flanking NOVX nucleic acid is of sufficient length for successful homologous recombination with the endogenous gene.
  • flanking DNA both at the 5'- and 3'-termini
  • the vector is ten introduced into an embryonic stem cell line (e.g., by elecfroporation) and cells in which the infroduced NOVX gene has homologously-recombined with the endogenous NOVX gene are selected. See, e.g., Li, et al, 1992. Cell 69: 915.
  • an animal e.g., a mouse
  • a chimeric embryo can then be implanted into a suitable pseudopregnant female foster animal and the embryo brought to term.
  • Progeny harboring the homologously-recombined DNA in their germ cells can be used to breed animals in which all cells of the animal contain the homologously-recombined DNA by germline transmission of the transgene.
  • transgenic non-humans animals can be produced that contain selected systems that allow for regulated expression of the transgene.
  • a system is the cre/loxP recombinase system of bacteriophage PI.
  • cre/loxP recombinase system See, e.g, Lakso, et al, 1992. Proc. Natl. Acad. Sci. USA 89: 6232-6236.
  • FLP recombinase system of Saccharomyces cerevisiae. See, O'Gorman, et al, 1991. Science 251:1351-1355.
  • mice containing transgenes encoding both the Cre recombinase and a selected protein are required.
  • Such animals can be provided through the construction of "double" transgenic animals, e.g., by mating two transgenic animals, one containing a transgene lencoding a selected protein and the other containing a transgene encoding a recombinase.
  • Clones of the non-human transgenic animals described herein can also be produced according to the methods described in Wilmut, et al, 1997. Nature 385: 810-813.
  • a cell e.g., a somatic cell
  • the quiescent cell can then be fused, e.g., through the use of electrical pulses, to an enucleated oocyte from an animal of the same species from which the quiescent cell is isolated.
  • the reconstructed oocyte is then cultured such that it develops to morula or blastocyte and then transferred to pseudopregnant female foster animal.
  • the offspring borne of this female foster animal will be a clone of the animal from which the cell (e.g., the somatic cell) is isolated.
  • compositions suitable for administration can be inco ⁇ orated into pharmaceutical compositions suitable for administration.
  • compositions typically comprise the nucleic acid molecule, protein, or antibody and a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable carrier is intended to include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and abso ⁇ tion delaying agents, and the like, compatible with pharmaceutical administration.
  • Suitable caniers are described in the most recent edition of Remington's Pharmaceutical Sciences, a standard reference text in the field, which is inco ⁇ orated herein by reference.
  • Prefened examples of such carriers or diluents include, but are not limited to, water, saline, finger's solutions, dextrose solution, and 5% human serum albumin. Liposomes and non-aqueous vehicles such as fixed oils may also be used.
  • the use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active compound, use thereof in the compositions is contemplated. Supplementary active compounds can also be inco ⁇ orated into the compositions.
  • a pharmaceutical composition of the invention is formulated to be compatible with its intended route of administration.
  • routes of administration include parenteral, e.g., intravenous, intradermal, subcutaneous, oral (e.g., inhalation), transdermal (i.e., topical), transmucosal, and rectal administration.
  • Solutions or suspensions used for parenteral, intradermal, or subcutaneous application can include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetefraacetic acid (EDTA); buffers such as acetates, citrates or phosphates, and agents for the adjustment of tonicity such as sodium chloride or dextrose.
  • the pH can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide.
  • the parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic.
  • compositions suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion.
  • suitable carriers include physiological saline, bacteriostatic water, Cremophor EL (BASF, Parsippany, N. J.) or phosphate buffered saline (PBS).
  • the composition must be sterile and should be fluid to the extent that easy syringeability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi.
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), and suitable mixtures thereof.
  • the proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • Prevention of the action of microorganisms can be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like.
  • isotonic agents for example, sugars, polyalcohols such as manitol, sorbitol, sodium chloride in the composition.
  • Prolonged abso ⁇ tion of the injectable compositions can be brought about by including in the composition an agent which delays abso ⁇ tion, for example, aluminum monostearate and gelatin.
  • Sterile injectable solutions can be prepared by inco ⁇ orating the active compound (e.g., a NOVX protein or anti-NOVX antibody) in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization.
  • the active compound e.g., a NOVX protein or anti-NOVX antibody
  • dispersions are prepared by inco ⁇ orating the active compound into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from thoseenumerated above.
  • methods of preparation are vacuum drying and freeze-drying that yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • Oral compositions generally include an inert diluent or an edible carrier. They can be enclosed in gelatin capsules or compressed into tablets. For the pu ⁇ ose of oral therapeutic administration, the active compound can be inco ⁇ orated with excipients and used in the form of tablets, troches, or capsules. Oral compositions can also be prepared using a fluid carrier for use as a mouthwash, wherein the compound in the fluid carrier is applied orally and swished and expectorated or swallowed. Pharmaceutically compatible binding agents, and/or adjuvant materials can be included as part of the composition.
  • the tablets, pills, capsules, troches and the like can contain any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum teagacanth or gelatin; an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or corn starch; a lubricant such as magnesium stearate or Sterotes; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring.
  • a binder such as microcrystalline cellulose, gum teagacanth or gelatin
  • an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or corn starch
  • a lubricant such as magnesium stearate or Sterotes
  • a glidant such as colloidal silicon dioxide
  • the compounds are delivered in the form of an aerosol spray from pressured container or dispenser which contains a suitable propellant, e.g., a gas such as carbon dioxide, or a nebulizer.
  • a suitable propellant e.g., a gas such as carbon dioxide, or a nebulizer.
  • Systemic administration can also be by transmucosal or teansdermal means.
  • penetrants appropriate to the barrier to be permeated are used in the formulation.
  • penetrants are generally known in the art, and include, for example, for transmucosal administration, detergents, bile salts, and fusidic acid derivatives.
  • Transmucosal administration can be accomplished through the use of nasal sprays or suppositories.
  • the active compounds are formulated into ointments, salves, gels, or creams as generally known in the art.
  • the compounds can also be prepared in the form of suppositories (e.g., with conventional suppository bases such as cocoa butter and other glycerides) or retention enemas for rectal delivery.
  • suppositories e.g., with conventional suppository bases such as cocoa butter and other glycerides
  • retention enemas for rectal delivery.
  • the active compounds are prepared with carriers that will protect the compound against rapid elimination from the body, such as a controlled release formulation, including implants and microencapsulated delivery systems.
  • a controlled release formulation including implants and microencapsulated delivery systems.
  • Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Methods for preparation of such formulations will be apparent to those skilled in the art.
  • the materials can also be obtained commercially from Alza Co ⁇ oration and Nova Pharmaceuticals, Inc.
  • Liposomal suspensions (including liposomes targeted to infected cells with monoclonal antibodies to viral antigens) can also be used as pharmaceutically acceptable carriers. These can be prepared according to methods known to those skilled in the art, for example, as described in U.S. Patent No. 4,522,811.
  • Dosage unit form refers to physically discrete units suited as unitary dosages for the subject to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • the specification for the dosage unit forms of the invention are dictated by and directly dependent on the unique characteristics of the active compound and the particular therapeutic effect to be achieved, and the limitations inherent in the art of compounding such an active compound for the treatment of individuals.
  • the nucleic acid molecules of the invention can be inserted into vectors and used as gene therapy vectors.
  • Gene therapy vectors can be delivered to a subject by, for example, intravenous injection, local administration (see, e.g., U.S. Patent No. 5,328,470) or by stereotactic injection (see, e.g., Chen, et al, 1994. Proc. Natl. Acad. Sci. USA 91: 3054-3057).
  • the pharmaceutical preparation of the gene therapy vector can include the gene therapy vector in an acceptable diluent, or can comprise a slow release matrix in which the gene delivery vehicle is imbedded.
  • the pharmaceutical preparation can include one or more cells that produce the gene delivery system.
  • compositions can be included in a container, pack, or dispenser together with instructions for administration. Screening and Detection Methods
  • the isolated nucleic acid molecules of the invention can be used to express NOVX protein (e.g., via a recombinant expression vector in a host cell in gene therapy applications), to detect NOVX mRNA (e.g., in a biological sample) or a genetic lesion in a NOVX gene, and to modulate NOVX activity, as described further, below.
  • the NOVX proteins can be used to screen drags or compounds that modulate the NOVX protein activity or expression as well as to treat disorders characterized by insufficient or excessive production of NOVX protein or production of NOVX protein forms that have decreased or abenant activity compared to NOVX wild-type protein (e.g.; diabetes (regulates insulin release); obesity (binds and transport lipids); metabolic disturbances associated with obesity, the metabolic syndrome X as well as anorexia and wasting disorders associated with chronic diseases and various cancers, and infectious disease(possesses anti-microbial activity) and the various dyslipidemias.
  • the anti-NOVX antibodies of the invention can be used to detect and isolate NOVX proteins and modulate NOVX activity.
  • the invention can be used in methods to influence appetite, abso ⁇ tion of nutrients and the disposition of metabolic substrates in both a positive and negative fashion.
  • the invention further pertains to novel agents identified by the screening assays described herein and uses thereof for treatments as described, supra.
  • the invention provides a method (also refened to herein as a "screening assay") for identifying modulators, i.e., candidate or test compounds or agents (e.g., peptides, peptidomimetics, small molecules or other drags) that bind to NOVX proteins or have a stimulatory or inhibitory effect on, e.g., NOVX protein expression or NOVX protein activity.
  • modulators i.e., candidate or test compounds or agents (e.g., peptides, peptidomimetics, small molecules or other drags) that bind to NOVX proteins or have a stimulatory or inhibitory effect on, e.g., NOVX protein expression or NOVX protein activity.
  • modulators i.e., candidate or test compounds or agents (e.g., peptides, peptidomimetics, small molecules or other drags) that bind to NOVX proteins or have a stimulatory or inhibitory effect on, e.g., NOVX protein expression
  • the invention provides assays for screening candidate or test compounds which bind to or modulate the activity of the membrane-bound form of a
  • test compounds of the invention can be obtained using any of the numerous approaches in combinatorial library methods known in the art, including: biological libraries; spatially addressable parallel solid phase or solution phase libraries; synthetic library methods requiring deconvolution; the "one-bead one-compound” library method; and synthetic library methods using affinity chromatography selection.
  • biological libraries are limited to peptide libraries, while the other four approaches are applicable to peptide, non-peptide oligomer or small molecule libraries of compounds. See, e.g., Lam, 1997. Anticancer Drug Design 12: 145.
  • a "small molecule” as used herein, is meant to refer to a composition that has a molecular weight of less than about 5 kD and most preferably less than about 4 kD.
  • Small molecules can be, e.g., nucleic acids, peptides, polypeptides, peptidomimetics, carbohydrates, lipids or other organic or inorganic molecules.
  • Libraries of chemical and/or biological mixtures, such as fungal, bacterial, or algal extracts, are known in the art and can be screened with any of the assays of the invention.
  • Biotechniques 13: 412-421 or on beads (Lam, 1991. Nature 354: 82-84), on chips (Fodor, 1993. Nature 364: 555-556), bacteria (Ladner, U.S. Patent No. 5,223,409), spores (Ladner, U.S. Patent 5,233,409), plasmids (Cull, et al, 1992. Proc. Natl. Acad. Sci. USA 89: 1865-1869) or on phage (Scott and Smith, 1990. Science 249: 386-390; Devlin, 1990. Science 249: 404-406; Cwirla, et al, 1990. Proc. Natl. Acad. Sci. U.S.A. 87: 6378-6382; Felici, 1991. J. Mol. Biol 222: 301-310; Ladner, U.S. Patent No. 5,233,409.).
  • an assay is a cell-based assay in which a cell which expresses a membrane-bound form of NOVX protein, or a biologically-active portion thereof, on the cell surface is contacted with a test compound and the ability of the test compound to bind to a NOVX protein determined.
  • the cell for example, can of mammalian origin or a yeast cell. Determining the ability of the test compound to bind to the NOVX protein can be accomplished, for example, by coupling the test compound with a radioisotope or enzymatic label such that binding of the test compound to the NOVX protein or biologically-active portion thereof can be determined by detecting the labeled compound in a complex.
  • test compounds can be labeled with 125 1, 35 S, 14 C, or 3 H, either directly or indirectly, and the radioisotope detected by direct counting of radioemission or by scintillation counting.
  • test compounds can be enzymatically-labeled with, for example, horseradish peroxidase, alkaline phosphatase, or luciferase, and the enzymatic label detected by determination of conversion of an appropriate substrate to product.
  • the assay comprises contacting a cell which expresses a membrane-bound form of NOVX protein, or a biologically-active portion thereof, on the cell surface with a known compound which binds NOVX to form an assay mixture, contacting the assay mixture with a test compound, and determining the ability of the test compound to interact with a NOVX protein, wherein determining the ability of the test compound to interact with a NOVX protein comprises determining the ability of the test compound to preferentially bind to NOVX protein or a biologically-active portion thereof as compared to the known compound.
  • an assay is a cell-based assay comprising contacting a cell expressing a membrane-bound form of NOVX protein, or a biologically-active portion thereof, on the cell surface with a test compound and determining the ability of the test compound to modulate (e.g., stimulate or inhibit) the activity of the NOVX protein or biologically-active portion thereof. Determining the ability of the test compound to modulate the activity of NOVX or a biologically-active portion thereof can be accomplished, for example, by determining the ability of the NOVX protein to bind to or interact with a NOVX target molecule.
  • a "target molecule” is a molecule with which a NOVX protein binds or interacts in nature, for example, a molecule on the surface of a cell which expresses a NOVX interacting protein, a molecule on the surface of a second cell, a molecule in the extracellular milieu, a molecule associated with the internal surface of a cell membrane or a cytoplasmic molecule, a NOVX target molecule can be a non-NOVX molecule or a NOVX protein or polypeptide of the invention.
  • a NOVX target molecule is a component of a signal transduction pathway that facilitates transduction of an extracellular signal (e.g.
  • the target for example, can be a second intercellular protein that has catalytic activity or a protein that facilitates the association of downstream signaling molecules with NOVX.
  • Determining the ability of the NOVX protein to bind to or interact with a NOVX target molecule can be accomplished by one of the methods described above for determining direct binding. In one embodiment, determining the ability of the NOVX protein to bind to or interact with a NOVX target molecule can be accomplished by determiningJhe activity of the target molecule. For example, the activity of the target molecule can be determined by detecting induction of a cellular second messenger of the target (i.e.
  • a reporter gene comprising a NOVX-responsive regulatory element operatively linked to a nucleic acid encoding a detectable marker, e.g., luciferase
  • a cellular response for example, cell survival, cellular differentiation, or cell proliferation.
  • an assay of the invention is a cell-free assay comprising contacting a NOVX protein or biologically-active portion thereof with a test compound and determining the ability of the test compound to bind to the NOVX protein or biologically-active portion thereof. Binding of the test compound to the NOVX protein can be determined either directly or indirectly as described above.
  • the assay comprises contacting the NOVX protein or biologically-active portion thereof with a known compound which binds NOVX to form an assay mixture, contacting the assay mixture with a test compound, and determining the ability of the test compound to interact with a NOVX protein, wherein determining the ability of the test compound to interact with a NOVX protein comprises determining the ability of the test compound to preferentially bind to NOVX or biologically-active portion thereof as compared to the known compound.
  • an assay is a cell-free assay comprising contacting NOVX protein or biologically-active portion thereof with a test compound and determining the ability of the test compound to modulate (e.g. stimulate or inhibit) the activity of the NOVX protein or biologically-active portion thereof. Determining the ability of the test compound to modulate the activity of NOVX can be accomplished, for example, by determining the ability of the NOVX protein to bind to a NOVX target molecule by one of the methods described above for determining direct binding. In an alternative embodiment, determining the ability of the test compound to modulate the activity of NOVX protein can be accomplished by determining the ability of the NOVX protein further modulate a NOVX target molecule. For example, the catalytic/enzymatic activity of the target molecule on an appropriate substrate can be determined as described, supra.
  • the cell-free assay comprises contacting the NOVX protein or biologically-active portion thereof with a known compound which binds NOVX protein to form an assay mixture, contacting the assay mixture with a test compound, and determining the ability of the test compound to interact with a NOVX protein, wherein determining the ability of the test compound to interact with a NOVX protein comprises determining the ability of the NOVX protein to preferentially bind to or modulate the activity of a NOVX target molecule.
  • the cell-free assays of the invention are amenable to use of both the soluble form or the membrane-bound form of NOVX protein.
  • solubilizing agent such that the membrane-bound form of NOVX protein is maintained in solution.
  • solubilizing agents include non-ionic detergents such as n-octylglucoside, n-dodecylglucoside, n-dodecylmaltoside, octanoyl-N-methylglucamide, decanoyl-N-methylglucamide, Triton ® X-100, Triton ® X-l 14, Thesit ® , Isotridecypoly(ethylene glycol ether) n , N-dodecyl ⁇ N,N-dimethyl-3-ammonio-l -propane sulfonate, 3-(3-cholamidopropyl) dimethylamminiol-1 -propane sulfonate (CHAPS), or 3-
  • binding of a test compound to NOVX protein, or interaction of NOVX protein with a target molecule in the presence and absence of a candidate compound can be accomplished in any vessel suitable for containing the reactants. Examples of such vessels include microtiter plates, test tubes, and micro-centrifuge tubes.
  • a fusion protein can be provided that adds a domain that allows one or both of the proteins to be bound to a matrix.
  • GST-NO VX fusion proteins or GST-target fusion proteins can be adsorbed onto glutathione sepharose beads (Sigma Chemical, St. Louis, MO) or glutathione derivatized microtiter plates, that are then combined with the test compound or the test compound and either the non-adsorbed target protein or NOVX protein, and the mixture is incubated under conditions conducive to complex formation (e.g., at physiological conditions for salt and pH). Following incubation, the beads or microtiter plate wells are washed to remove any unbound components, the matrix immobilized in the case of beads, complex determined either directly or indirectly, for example, as described, supra.
  • glutathione sepharose beads Sigma Chemical, St. Louis, MO
  • glutathione derivatized microtiter plates glutathione derivatized microtiter plates
  • the complexes can be dissociated from the matrix, and the level of NOVX protein binding or activity determined using standard techniques.
  • Other techniques for immobilizing proteins on matrices can also be used in the screening assays of the invention.
  • either the NOVX protein or its target molecule can be immobilized utilizing conjugation of biotin and streptavidin.
  • Biotinylated NOVX protein or target molecules can be prepared from biotin-NHS (N-hydroxy-succinimide) using techniques well-known within the art (e.g., biotinylation kit, Pierce Chemicals, Rockford, 111.), and immobilized in the wells of streptavidin-coated 96 well plates (Pierce Chemical).
  • antibodies reactive with NOVX protein or target molecules can be derivatized to the wells of the plate, and unbound target or NOVX protein trapped in the wells by antibody conjugation.
  • Methods for detecting such complexes include immunodetection of complexes using antibodies reactive with the NOVX protein or target molecule, as well as enzyme-linked assays that rely on detecting an enzymatic activity associated with the NOVX protein or target molecule.
  • modulators of NOVX protein expression are identified in a method wherein a cell is contacted with a candidate compound and the expression of NOVX mRNA or protein in the cell is determined. The level of expression of NOVX mRNA or protein in the presence of the candidate compound is compared to the level of expression of NOVX mRNA or protein in the absence of the candidate compound. The candidate compound can then be identified as a modulator of NOVX mRNA or protein expression based upon this comparison. For example, when expression of NOVX mRNA or protein is greater (i.e., statistically significantly greater) in the presence of the candidate compound than in its absence, the candidate compound is identified as a stimulator of NOVX mRNA or protein expression.
  • the candidate compound when expression of NOVX mRNA or protein is less (statistically significantly less) in the presence of the candidate compound than in its absence, the candidate compound is identified as an inhibitor of NOVX mRNA or protein expression.
  • the level of NOVX mRNA or protein expression in the cells can be determined by methods described herein for detecting NOVX mRNA or protein.
  • the NOVX proteins can be used as "bait proteins" in a two-hybrid assay or three hybrid assay (see, e.g., U.S. Patent No. 5,283,317; Zervos, et al, 1993. Cell 72: 223-232; Madura, et al, 1993. J. Biol. Chem. 268: 12046-12054; Bartel, etal, 1993. Biotechniques 14: 920-924; Iwabuchi, etal, 1993.
  • NOVX-binding proteins proteins that bind to or interact with NOVX
  • NOVX-binding proteins proteins that bind to or interact with NOVX
  • NOVX-binding proteins are also likely to be involved in the propagation of signals by the NOVX proteins as, for example, upstream or downstream elements of the NOVX pathway.
  • the two-hybrid system is based on the modular nature of most transcription factors, which consist of separable DNA-binding and activation domains. Briefly, the assay utilizes two different DNA constructs.
  • the gene that codes for NOVX is fused to a gene encoding the DNA binding domain of a known transcription factor (e.g., GAL-4).
  • a DNA sequence, from a library of DNA sequences, that encodes an unidentified protein (“prey” or “sample”) is fused to a gene that codes for the activation domain of the known transcription factor. If the "bait" and the "prey” proteins are able to interact, in vivo, forming a NOVX-dependent complex, the DNA-binding and activation domains of the transcription factor are brought into close proximity. This proximity allows transcription of a reporter gene (e.g., LacZ) that is operably linked to a transcriptional regulatory site responsive to the transcription factor. Expression of the reporter gene can be detected and cell colonies containing the functional transcription factor can be isolated and used to obtain the cloned gene that encodes the protein which interacts with NOVX.
  • a reporter gene e.g., LacZ
  • the invention further pertains to novel agents identified by the aforementioned screening assays and uses thereof for treatments as described herein.
  • cDNA sequences identified herein can be used in numerous ways as polynucleotide reagents.
  • these sequences can be used to: (i) map their respective genes on a chromosome; and, thus, locate gene regions associated with genetic disease; (ii) identify an individual from a minute biological sample (tissue typing); and (iii) aid in forensic identification of a biological sample.
  • chromosome mapping Once the sequence (or a portion of the sequence) of a gene has been isolated, this sequence can be used to map the location of the gene on a chromosome. This process is called chromosome mapping. Accordingly, portions or fragments of a NOVX sequence, i.e., of SEQ ID NOS :2n-l, wherein n is an integer between 1 and 141, or fragments or derivatives thereof, can be used to map the location of the NOVX genes, respectively, on a chromosome. The mapping of the NOVX sequences to chromosomes is an important first step in conelating these sequences with genes associated with disease. Briefly, NOVX genes can be mapped to chromosomes by preparing PCR primers
  • sequences can be used to rapidly select primers that do not span more than one exon in the genomic DNA, thus complicating the amplification process. These primers can then be used for PCR screening of somatic cell hybrids containing individual human chromosomes. Only those hybrids containing the human gene conesponding to the NOVX sequences will yield an amplified fragment.
  • Somatic cell hybrids are prepared by fusing somatic cells from different mammals (e.g., human and mouse cells). As hybrids of human and mouse cells grow and divide, they gradually lose human chromosomes in random order, but retain the mouse chromosomes. By using media in which mouse cells cannot grow, because they lack a particular enzyme, but in which human cells can, the one human chromosome that contains the gene encoding the needed enzyme will be retained. By using various media, panels of hybrid cell lines can be established. Each cell line in a panel contains either a single human chromosome or a small number of human chromosomes, and a full set of mouse chromosomes, allowing easy mapping of individual genes to specific human chromosomes.
  • mammals e.g., human and mouse cells.
  • Somatic cell hybrids containing only fragments of human chromosomes can also be produced by using human chromosomes with translocations and deletions.
  • PCR mapping of somatic cell hybrids is a rapid procedure for assigning a particular sequence to a particular chromosome. Three or more sequences can be assigned per day using a single thermal cycler. Using the NOVX sequences to design oligonucleotide primers, sub-localization can be achieved with panels of fragments from specific chromosomes.
  • Fluorescence in situ hybridization (FISH) of a DNA sequence to a metaphase chromosomal spread can further be used to provide a precise chromosomal location in one step.
  • Chromosome spreads can be made using cells whose division has been blocked in metaphase by a chemical like colcemid that disrupts the mitotic spindle.
  • the chromosomes can be treated briefly with trypsin, and then stained with Giemsa. A pattern of light and dark bands develops on each chromosome, so that the chromosomes can be identified individually.
  • the FISH technique can be used with a DNA sequence as short as 500 or 600 bases.
  • clones larger than 1,000 bases have a higher likelihood of binding to a unique chromosomal location with sufficient signal intensity for simple detection.
  • Reagents for chromosome mapping can be used individually to mark a single chromosome or a single site on that chromosome, or panels of reagents can be used for marking multiple sites and/or multiple chromosomes. Reagents conesponding to noncoding regions of the genes actually are prefened for mapping pu ⁇ oses. Coding sequences are more likely to be conserved within gene families, thus increasing the chance of cross hybridizations during chromosomal mapping. Once a sequence has been mapped to a precise chromosomal location, the physical position of the sequence on the chromosome can be conelated with genetic map data.
  • differences in the DNA sequences between individuals affected and unaffected with a disease associated with the NOVX gene can be determined. If a mutation is observed in some or all of the affected individuals but not in any unaffected individuals, then the mutation is likely to be the causative agent of the particular disease. Comparison of affected and unaffected individuals generally involves first looking for structural alterations in the chromosomes, such as deletions or translocations that are visible from chromosome spreads or detectable using PCR based on that DNA sequence. Ultimately, complete sequencing of genes from several individuals can be performed to confirm the presence of a mutation and to distinguish mutations from polymo ⁇ hisms. Tissue Typing
  • the NOVX sequences of the invention can also be used to identify individuals from minute biological samples.
  • an individual's genomic DNA is digested with one or more restriction enzymes, and probed on a Southern blot to yield unique bands for identification.
  • the sequences of the invention are useful as additional DNA markers for RFLP ("restriction fragment length polymo ⁇ hisms," described in U.S. Patent No. 5,272,057).
  • sequences of the invention can be used to provide an alternative technique that determines the actual base-by-base DNA sequence of selected portions of an individual's genome.
  • NOVX sequences described herein can be used to prepare two PCR primers from the 5'- and 3'-termini of the sequences. These primers can then be used to amplify an individual's DNA and subsequently sequence it.
  • Panels of conesponding DNA sequences from individuals, prepared in this manner, can provide unique individual identifications, as each individual will have a unique set of such DNA sequences due to allelic differences.
  • the sequences of the invention can be used to obtain such identification sequences from individuals and from tissue.
  • the NOVX sequences of the invention uniquely represent portions of the human genome. Allelic variation occurs to some degree in the coding regions of these sequences, and to a greater degree in the noncoding regions. It is estimated that allelic variation between individual humans occurs with a frequency of about once per each 500 bases. Much of the allelic variation is due to single nucleotide polymo ⁇ hisms (SNPs), which include restriction fragment length polymo ⁇ hisms (RFLPs).
  • SNPs single nucleotide polymo ⁇ hisms
  • RFLPs restriction fragment length polymo ⁇ hisms
  • each of the sequences described herein can, to some degree, be used as a standard against which DNA from an individual can be compared for identification pu ⁇ oses. Because greater numbers of polymo ⁇ hisms occur in the noncoding regions, fewer sequences are necessary to differentiate individuals.
  • the noncoding sequences can comfortably provide positive individual identification with a panel of perhaps 10 to 1,000 primers that each yield a noncoding amplified sequence of 100 bases. If coding sequences, such as those of SEQ ID NOS:2n-l, wherein n is an integer between 1 and 141, are used, a more appropriate number of primers for positive individual identification would be 500-2,000.
  • the invention also pertains to the field of predictive medicine in which diagnostic assays, prognostic assays, pharmacogenomics, and monitoring clinical trials are used for prognostic (predictive) pu ⁇ oses to thereby treat an individual prophylactically. Accordingly, one aspect of the invention relates to diagnostic assays for determining
  • NOVX protein and/or nucleic acid expression as well as NOVX activity in the context of a biological sample (e.g., blood, serum, cells, tissue) to thereby determine whether an individual is afflicted with a disease or disorder, or is at risk of developing a disorder, associated with abenant NOVX expression or activity.
  • the disorders include metabolic disorders, diabetes, obesity, infectious disease, anorexia, cancer-associated cachexia, cancer, neurodegenerative disorders, Alzheimer's Disease, Parkinson's Disorder, immune disorders, and hematopoietic disorders, and the various dyslipidemias, metabolic disturbances associated with obesity, the metabolic syndrome X and wasting disorders associated with chronic diseases and various cancers.
  • the invention also provides for prognostic (or predictive) assays for determining whether an individual is at risk of developing a disorder associated with NOVX protein, nucleic acid expression or activity. For example, mutations in a NOVX gene can be assayed in a biological sample. Such assays can be used for prognostic or predictive pu ⁇ ose to thereby prophylactically treat an individual prior to the onset of a disorder characterized by or associated with NOVX protein, nucleic acid expression, or biological activity.
  • Another aspect of the invention provides methods for determining NOVX protein, nucleic acid expression or activity in an individual to thereby select appropriate therapeutic or prophylactic agents for that individual (refened to herein as "pharmacogenomics").
  • Pharmacogenomics allows for the selection of agents (e.g., drugs) for therapeutic or prophylactic freatment of an individual based on the genotype of the individual (e.g., the genotype of the individual examined to determine the ability of the individual to respond to a particular agent.)
  • Yet another aspect of the invention pertains to monitoring the influence of agents (e.g., drugs, compounds) on the expression or activity of NOVX in clinical trials.
  • agents e.g., drugs, compounds
  • An exemplary method for detecting the presence or absence of NOVX in a biological sample involves obtaining a biological sample from a test subject and contacting the biological sample with a compound or an agent capable of detecting NOVX protein or nucleic acid (e.g., mRNA, genomic DNA) that encodes NOVX protein such that the presence of NOVX is detected in the biological sample.
  • a compound or an agent capable of detecting NOVX protein or nucleic acid e.g., mRNA, genomic DNA
  • An agent for detecting NOVX mRNA or genomic DNA is a labeled nucleic acid probe capable of hybridizing to NOVX mRNA or genomic DNA.
  • the nucleic acid probe can be, for example, a full-length NOVX nucleic acid, such as the nucleic acid of SEQ ID NOS:2n-l, wherein n is an integer between 1 and 141, or a portion thereof, such as an oligonucleotide of at least 15, 30, 50, 100, 250 or 500 nucleotides in length and sufficient to specifically hybridize under stringent conditions to NOVX mRNA or genomic DNA.
  • n is an integer between 1 and 141
  • a portion thereof such as an oligonucleotide of at least 15, 30, 50, 100, 250 or 500 nucleotides in length and sufficient to specifically hybridize under stringent conditions to NOVX mRNA or genomic DNA.
  • Other suitable probes for use in the diagnostic assays of the invention are described herein.
  • An agent for detecting NOVX protein is an antibody capable of binding to NOVX protein, preferably an antibody with a detectable label.
  • Antibodies can be polyclonal, or more preferably, monoclonal. An intact antibody, or a fragment thereof (e.g., Fab or F(ab') 2 ) can be used.
  • the term "labeled", with regard to the probe or antibody, is intended to encompass direct labeling of the probe or antibody by coupling (i.e., physically linking) a detectable substance to the probe or antibody, as well as indirect labeling of the probe or antibody by reactivity with another reagent that is directly labeled.
  • Examples of indirect labeling include detection of a primary antibody using a fluorescently-labeled secondary antibody and end-labeling of a DNA probe with biotin such that it can be detected with fluorescently-labeled streptavidin.
  • biological sample is intended to include tissues, cells and biological fluids isolated from a subject, as well as tissues, cells and fluids present within a subject.. That is, the detection method of the invention can be used to detect NOVX mRNA, protein, or genomic DNA in a biological sample in vitro as well as in vivo.
  • in vitro techniques for detection of NOVX mRNA include Northern hybridizations and in situ hybridizations.
  • In vitro techniques for detection of NOVX protein include enzyme linked immunosorbent assays (ELISAs), Western blots, immunoprecipitations, and immunofluorescence.
  • In vitro techniques for detection of NOVX genomic DNA include Southern hybridizations.
  • in vivo techniques for detection of NOVX protein include introducing into a subject a labeled anti-NOVX antibody.
  • the antibody can be labeled with a radioactive marker whose presence and location in a subject can be detected by standard imaging techniques.
  • the biological sample contains protein molecules from the test subject.
  • the biological sample can contain mRNA molecules from the test subject or genomic DNA molecules from the test subject.
  • a prefened biological sample is a peripheral blood leukocyte sample isolated by conventional means from a subject.
  • the methods further involve obtaining a control biological sample from a confrol subject, contacting the control sample with a compound or agent capable of detecting NOVX protein, mRNA, or genomic DNA, such that the presence of NOVX protein, mRNA or genomic DNA is detected in the biological sample, and comparing the presence of NOVX protein, mRNA or genomic DNA in the control sample with the presence of NOVX protein, mRNA or genomic DNA in the test sample.
  • kits for detecting the presence of NOVX in a biological sample can comprise: a labeled compound or agent capable of detecting NOVX protein or mRNA in a biological sample; means for determining the amount of NOVX in the sample; and means for comparing the amount of NOVX in the sample with a standard.
  • the compound or agent can be packaged in a suitable container.
  • the kit can further comprise instructions for using the kit to detect NOVX protein or nucleic acid.
  • the diagnostic methods described herein can furthermore be utilized to identify subjects having or at risk of developing a disease or disorder associated with abenant NOVX expression or activity.
  • the assays described herein such as the preceding diagnostic assays or the following assays, can be utilized to identify a subject having or at risk of developing a disorder associated with NOVX protein, nucleic acid expression or activity.
  • the prognostic assays can be utilized to identify a subject having or at risk for developing a disease or disorder.
  • the invention provides a method for identifying a disease or disorder associated with abenant NOVX expression or activity in which a test sample is obtained from a subject and NOVX protein or nucleic acid (e.g., mRNA, genomic DNA) is detected, wherein the presence of NOVX protein or nucleic acid is diagnostic for a subject having or at risk of developing a disease or disorder associated with abenant NOVX expression or activity.
  • a test sample refers to a biological sample obtained from a subject of interest.
  • a test sample can be a biological fluid (e.g., serum), cell sample, or tissue.
  • the prognostic assays described herein can be used to determine whether a subject can be administered an agent (e.g., an agonist, antagonist, peptidomimetic, protein, peptide, nucleic acid, small molecule, or other drag candidate) to treat a disease or disorder associated with abenant NOVX expression or activity.
  • an agent e.g., an agonist, antagonist, peptidomimetic, protein, peptide, nucleic acid, small molecule, or other drag candidate
  • such methods can be used to determine whether a subject can be effectively treated with an agent for a disorder.
  • the invention provides methods for determining whether a subject can be effectively treated with an agent for a disorder associated with abenant NOVX expression or activity in which a test sample is obtained and NOVX protein or nucleic acid is detected (e.g., wherein the presence of NOVX protein or nucleic acid is diagnostic for a subject that can be administered the agent to teeat a disorder associated with abenant NOVX expression or activity).
  • the methods of the invention can also be used to detect genetic lesions in a NOVX gene, thereby determining if a subject with the lesioned gene is at risk for a disorder characterized by abenant cell proliferation and/or differentiation.
  • the methods include detecting, in a sample of cells from the subject, the presence or absence of a genetic lesion characterized by at least one of an alteration affecting the integrity of a gene encoding a NOVX-protein, or the misexpression of the NOVX gene.
  • such genetic lesions can be detected by ascertaining the existence of at least one of: (i) a deletion of one or more nucleotides from a NOVX gene; (ii) an addition of one or more nucleotides to a NOVX gene; (iii) a substitution of one or more nucleotides of a NOVX gene, (fv) a chromosomal reanangement of a NOVX gene; (v) an alteration in the level of a messenger RNA transcript of a NOVX gene, (vi) abenant modification of a NOVX gene, such as of the methylation pattern of the genomic DNA, (vii) the presence of a non- wild-type splicing pattern of a messenger RNA transcript of a NOVX gene, (viii) a non-wild-type level of a NOVX protein, (ix) allelic loss of a NOVX gene, and (x) inappropriate post-translational modification of a NOVX protein.
  • a prefened biological sample is a peripheral blood leukocyte sample isolated by conventional means from a subject.
  • any biological sample containing nucleated cells may be used, including, for example, buccal mucosal cells.
  • detection of the lesion involves the use of a probe/primer in a polymerase chain reaction (PCR) (see, e.g., U.S. Patent Nos.
  • PCR polymerase chain reaction
  • This method can include the steps of collecting a sample of cells from a patient, isolating nucleic acid (e.g., genomic, mRNA or both) from the cells of the sample, contacting the nucleic acid sample with one or more primers that specifically hybridize to a NOVX gene under conditions such that hybridization and amplification of the NOVX gene (if present) occurs, and detecting the presence or absence of an amplification product, or detecting the size of the amplification product and comparing the length to a control sample. It is anticipated that PCR and/or LCR may be desirable to use as a preliminary amplification step in conjunction with any of the techniques used for detecting mutations described herein.
  • nucleic acid e.g., genomic, mRNA or both
  • Alternative amplification methods include: self sustained sequence replication (see, Guatelli, et al, 1990. Proc. Natl. Acad. Sci. USA 87: 1874-1878), transcriptional amplification system (see, Kwoh, et al, 1989. Proc. Natl. Acad. Sci. USA 86: 1173-1177); Q ⁇ Replicase (see, Lizardi, et al, 1988. BioTechnology 6: 1197), or any other nucleic acid amplification method, followed by the detection of the amplified molecules using techniques well known to those of skill in the art. These detection schemes are especially useful for the detection of nucleic acid molecules if such molecules are present in very low numbers.
  • mutations in a NOVX gene from a sample cell can be identified by alterations in restriction enzyme cleavage patterns.
  • sample and control DNA is isolated, amplified (optionally), digested with one or more restriction endonucleases, and fragment length sizes are determined by gel electrophoresis and compared. Differences in fragment length sizes between sample and control DNA indicates mutations in the sample DNA.
  • sequence specific ribozymes see, e.g., U.S. Patent No. 5,493,531 can be used to score for the presence of specific mutations by development or loss of a ribozyme cleavage site.
  • genetic mutations in NOVX can be identified by hybridizing sample and control nucleic acids, e.g., DNA or RNA to high-density anays containing hundreds or thousands of oligonucleotide probes. See, e.g., Cronin, et al, 1996. Human Mutation 7: 244-255; Kozal, et al, 1996. Nat. Med. 2: 753-759.
  • genetic mutations in NOVX can be identified in two-dimensional anays containing light-generated DNA probes as described in Cronin, et al, supra.
  • a first hybridization anay of probes can be used to scan through long sfretches of DNA in a sample and confrol to identify base changes between the sequences by making linear anays of sequential overlapping probes. This step allows the identification of point mutations.
  • a second hybridization anay that allows the characterization of specific mutations by using smaller, specialized probe anays complementary to all variants or mutations detected.
  • Each mutation anay is composed of parallel probe sets, one complementary to the wild-type gene and the other complementary to the mutant gene.
  • any of a variety of sequencing reactions known in the art can be used to directly sequence the NOVX gene and detect mutations by comparing the sequence of the sample NOVX with the conesponding wild-type (control) sequence.
  • sequencing reactions include those based on techniques developed by Maxim and Gilbert, 1977. Proc. Natl. Acad. Sci. USA 74: 560 or Sanger, 1977. Proc. Natl. Acad. Sci. USA 74: 5463. It is also contemplated that any of a variety of automated sequencing procedures can be utilized when performing the diagnostic assays (see, e.g., Naeve, et al, 1995.
  • Biotechniques 19: 448 including sequencing by mass specfrometry (see, e.g., PCT International Publication No. WO 94/16101; Cohen, et al, 1996. Adv. Chromatography 36: 127-162; and Griffin, et al, 1993. Appl. Biochem. Biotechnol. 38: 147-159).
  • RNA/RNA or RNA/DNA heteroduplexes Other methods for detecting mutations in the NOVX gene include methods in which protection from cleavage agents is used to detect mismatched bases in RNA/RNA or RNA/DNA heteroduplexes. See, e.g., Myers, et al, 1985. Science 230: 1242.
  • the art technique of "mismatch cleavage" starts by providing heteroduplexes of formed by hybridizing (labeled) RNA or DNA containing the wild-type NOVX sequence with potentially mutant RNA or DNA obtained from a tissue sample.
  • the double-stranded duplexes are treated with an agent that cleaves single-stranded regions of the duplex such as which will exist due to basepair mismatches between the control and sample strands.
  • RNA/DNA duplexes can be treated with RNase and DNA DNA hybrids freated with Si nuclease to enzymatically digesting the mismatched regions.
  • either DNA/DNA or RNA/DNA duplexes can be freated with hydroxylamine or osmium tefroxide and with piperidine in order to digest mismatched regions. After digestion of the mismatched regions, the resulting material is then separated by size on denaturing polyacrylamide gels to determine the site of mutation. See, e.g., Cotton, et al, 1988. Proc. Natl. Acad. Sci. USA 85: 4397; Saleeba, et al, 1992. Methods Enzymol. 217: 286-295.
  • the confrol DNA or RNA can be labeled for detection.
  • the mismatch cleavage reaction employs one or more proteins that recognize mismatched base pairs in double-stranded DNA (so called "DNA mismatch repair" enzymes) in defined systems for detecting and mapping point mutations in NOVX cDNAs obtained from samples of cells.
  • DNA mismatch repair enzymes
  • the mutY enzyme of E. coli cleaves A at G/A mismatches and the thymidine DNA glycosylase from HeLa cells cleaves T at G/T mismatches. See, e.g., Hsu, et al, 1994. Carcinogenesis 15: 1657-1662.
  • a probe based on a NOVX sequence e.g., a wild-type NOVX sequence
  • a cDNA or other DNA product from a test cell(s).
  • the duplex is treated with a DNA mismatch repair enzyme, and the cleavage products, if any, can be detected from electrophoresis protocols or the like. See, e.g., U.S. Patent No. 5,459,039.
  • alterations in electrophoretic mobility will be used to identify mutations in NOVX genes.
  • single strand conformation polymo ⁇ hism may be used to detect differences in electrophoretic mobility between mutant and wild type nucleic acids. See, e.g., Orita, et al, 1989. Proc. Natl. Acad. Sci. USA: 86: 2766; Cotton, 1993. Mutat. Res. 285: 125-144; Hayashi, 1992. Genet. Anal. Tech. Appl 9: 73-79. Single-stranded DNA fragments of sample and control NOVX nucleic acids will be denatured and allowed to renature.
  • the secondary structure of single-stranded nucleic acids varies according to sequence, the resulting alteration in electrophoretic mobility enables the detection of even a single base change.
  • the DNA fragments may be labeled or detected with labeled probes.
  • the sensitivity of the assay may be enhanced by using RNA (rather than DNA), in which the secondary structure is more sensitive to a change in sequence.
  • the subject method utilizes heteroduplex analysis to separate double sfranded heteroduplex molecules on the basis of changes in electrophoretic mobility. See, e.g., Keen, et al, 1991. Trends Genet. 7: 5.
  • the movement of mutant or wild-type fragments in polyacrylamide gels containing a gradient of denaturant is assayed using denaturing gradient gel electrophoresis (DGGE).
  • DGGE denaturing gradient gel electrophoresis
  • DNA will be modified to insure that it does not completely denature, for example by adding a GC clamp of approximately 40 bp of high-melting GC-rich DNA by PCR.
  • a temperature gradient is used in place of a denaturing gradient to identify differences in the mobility of control and sample DNA. See, e.g., Rosenbaum andReissner, 1987 ' . Biophys. Chem. 265: 12753.
  • oligonucleotide primers may be prepared in which the known mutation is placed centrally and then hybridized to target DNA under conditions that permit hybridization only if a perfect match is found. See, e.g., Saiki, et al, 1986. Nature 324: 163; Saiki, et al, 1989. Proc. Natl. Acad. Sci. USA 86: 6230.
  • Such allele specific oligonucleotides are hybridized to PCR amplified target DNA or a number of different mutations when the oligonucleotides are attached to the hybridizing membrane and hybridized with labeled target DNA.
  • Oligonucleotides used as primers for specific amplification may carry the mutation of interest in the center of the molecule (so that amplification depends on differential hybridization; see, e.g., Gibbs, et al, 1989. Nucl. Acids Res. 17: 2437-2448) or at the extreme 3 '-terminus of one primer where, under appropriate conditions, mismatch can prevent, or reduce polymerase extension (see, e.g., Prossner, 1993. T ⁇ btech. 11: 238).
  • amplification may also be performed using Taq ligase for amplification. See, e.g., Barany, 1991. Proc. Natl. Acad. Sci. USA 88: 189. In such cases, ligation will ocpur only if there is a perfect match at the 3'-terminus of the 5' sequence, making it possible to detect the presence of a known mutation at a specific site by looking for the presence or absence of amplification.
  • the methods described herein may be performed, for example, by utilizing pre-packaged diagnostic kits comprising at least one probe nucleic acid or antibody reagent described herein, which may be conveniently used, e.g., in clinical settings to diagnose patients exhibiting symptoms or family history of a disease or illness involving a NOVX gene.
  • any cell type or tissue preferably peripheral blood leukocytes, in which NOVX is expressed maybe utilized in the prognostic assays described herein.
  • any biological sample containing nucleated cells may be used, including, for example, buccal mucosal cells.
  • Agents, or modulators that have a stimulatory or inhibitory effect on NOVX activity can be administered to individuals to treat (prophylactically or therapeutically) disorders
  • disorders include metabolic disorders, diabetes, obesity, infectious disease, anorexia, cancer-associated cachexia, cancer, neurodegenerative disorders, Alzheimer's Disease, Parkinson's Disorder, immune disorders, and hematopoietic disorders, and the various dyslipidemias, metabolic disturbances associated with obesity, the metabolic syndrome X and wasting disorders associated with chronic diseases and various cancers.
  • the pharmacogenomics i.e., the study of the relationship between an individual's genotype and that individual's response to a foreign compound or drag
  • the individual may be considered.
  • the pharmacogenomics of the individual permits the selection of effective agents (e.g., drugs) for prophylactic or therapeutic treatments based on a consideration of the individual's genotype. Such pharmacogenomics can further be used to determine appropriate dosages and therapeutic regimens. Accordingly, the activity of NOVX protein, expression of NOVX nucleic acid, or mutation content of NOVX genes in an individual can be determined to thereby select appropriate agent(s) for therapeutic or prophylactic treatment of the individual.
  • Pharmacogenomics deals with clinically significant hereditary variations in the response to drugs due to altered drag disposition and abnormal action in affected persons. See e.g., Eichelbaum, 1996. Clin. Exp. Pharmacol. Physiol, 23: 983-985; Linder, 1997. Clin. 'Chem., 43: 254-266.
  • two types of pharmacogenetic conditions can be differentiated. Genetic conditions transmitted as a single factor altering the way drags act on the body (altered drag action) or genetic conditions transmitted as single factors altering the way the body acts on drugs (altered drag metabolism). These pharmacogenetic conditions can occur either as rare defects or as polymo ⁇ hisms.
  • G6PD glucose-6-phosphate dehydrogenase
  • the activity of drug metabolizing enzymes is a major determinant of both the intensity and duration of drug action.
  • drag metabolizing enzymes e.g., N-acetylteansferase 2 (NAT 2) and cytochrome Pregnancy Zone Protein Precursor enzymes CYP2D6 and C YP2C 19
  • NAT 2 N-acetylteansferase 2
  • CYP2D6 and C YP2C 19 cytochrome Pregnancy Zone Protein Precursor enzymes
  • These polymo ⁇ hisms are expressed in two phenotypes in the population, the extensive metabolizer (EM) and poor metabolizer (PM). The prevalence of PM is different among different populations.
  • EM extensive metabolizer
  • PM poor metabolizer
  • CYP2D6 is highly polymo ⁇ hic and several mutations have been identified in PM, which all lead to the absence of functional CYP2D6. Poor metabolizers of CYP2D6 and CYP2C19 quite frequently experience exaggerated drug response and side effects when they receive standard doses. If a metabolite is the active therapeutic moiety, PM show no therapeutic response, as demonstrated for the analgesic effect of codeine mediated by its CYP2D6-formed metabolite mo ⁇ hine. At the other extreme are the so called ultra-rapid metabolizers who do not respond to standard doses. Recently, the molecular basis of ultra-rapid metabolism has been identified to be due to CYP2D6 gene amplification.
  • the activity of NOVX protein, expression of NOVX nucleic acid, or mutation content of NOVX genes in an individual can be determined to thereby select appropriate agent(s) for therapeutic or prophylactic treatment of the individual.
  • pharmacogenetic studies can be used to apply genotyping of polymo ⁇ hic alleles encoding drug-metabolizing enzymes to the identification of an individual's drag responsiveness phenotype. This knowledge, when applied to dosing or drag selection, can avoid adverse reactions or therapeutic failure and thus enhance therapeutic or prophylactic efficiency when treating a subject with a NOVX modulator, such as a modulator identified by one of the exemplary screening assays described herein. Monitoring of Effects During Clinical Trials
  • Monitoring the influence of agents (e.g., drugs, compounds) on the expression or activity of NOVX can be applied not only in basic drug screening, but also in clinical trials.
  • agents e.g., drugs, compounds
  • the effectiveness of an agent determined by a screening assay as described herein to increase NOVX gene expression, protein levels, or upregulate NOVX activity can be monitored in clinical trails of subjects exhibiting decreased NOVX gene expression, protein levels, or downregulated NOVX activity.
  • the effectiveness of an agent determined by a screening assay to decrease NOVX gene expression, protein levels, or downregulate NOVX activity can be monitored in clinical trails of subjects exhibiting increased NOVX gene expression, protein levels, or upregulated NOVX activity.
  • the expression or activity of NOVX and, preferably, other genes that have been implicated in, for example, a cellular proliferation or immune disorder can be used as a "read out" or markers of the immune responsiveness of a particular cell .
  • genes including NOVX, that are modulated in cells by freatment with an agent (e.g., compound, drug or small molecule) that modulates NOVX activity (e.g., identified in a screening assay as described herein) can be identified.
  • an agent e.g., compound, drug or small molecule
  • NOVX activity e.g., identified in a screening assay as described herein
  • cells can be isolated and RNA prepared and analyzed for the levels of expression of NOVX and other genes implicated in the disorder.
  • the levels of gene expression can be quantified by Northern blot analysis or RT-PCR, as described herein, or alternatively by measuring the amount of protein produced, by one of the methods as described herein, or by measuring the levels of activity of NOVX or other genes.
  • the gene expression pattern can serve as a marker, indicative of the physiological response of the cells to the agent. Accordingly, this response state may be determined before, and at various points during, treatment of the individual with the agent.
  • the invention provides a method for monitoring the effectiveness of treatment of a subject with an agent (e.g., an agonist, antagonist, protein, peptide, peptidomimetic, nucleic acid, small molecule, or other drug candidate identified by the screening assays described herein) comprising the steps of (i) obtaining a pre-administeation sample from a subject prior to administration of the agent; (ii) detecting the level of expression of a NOVX protein, mRNA, or genomic DNA in the preadminisfration sample; (iii) obtaining one or more post-administration samples from the subject; (zv) detecting the level of expression or activity of the NOVX protein, mRNA, or genomic DNA in the post-administration samples; (v) comparing the level of expression or activity of the NOVX protein, mRNA, or genomic DNA in the pre-administration sample with the NOVX protein, mRNA, or genomic DNA in the post administration sample or samples; and (vi) altering the administration of the agent to the subject accordingly
  • increased administration of the agent may be desirable to increase the expression or activity of NOVX to higher levels than detected, i.e., to increase the effectiveness of the agent.
  • decreased administration of the agent may be desirable to decrease expression or activity of NOVX to lower levels than detected, i.e., to decrease the effectiveness of the agent.
  • the invention provides for both prophylactic and therapeutic methods of treating a subject at risk of (or susceptible to) a disorder or having a disorder associated with abenant NOVX expression or activity.
  • the disorders include cardiomyopathy, atherosclerosis, hypertension, congenital heart defects, aortic stenosis, ateial septal defect (ASD), atrioventricular (A-V) canal defect, ductus arteriosus, pulmonary stenosis, subaortic stenosis, ventricular septal defect (VSD), valve diseases, tuberous sclerosis, scleroderma, obesity, transplantation, adrenoleukodystrophy, congenital adrenal hype ⁇ lasia, prostate cancer, neoplasm; adenocarcinoma, lymphoma, uterus cancer, fertility, hemophilia, hypercoagulation, idiopathic thrombocytopenic pu ⁇ ura, immunodeficiencies, graft versus host disease, AIDS, bronchial
  • Therapeutics that antagonize i.e., reduce or inhibit activity.
  • Therapeutics that antagonize activity maybe administered in a therapeutic or prophylactic manner.
  • Therapeutics that may be utilized include, but are not limited to: (i) an aforementioned peptide, or analogs, derivatives, fragments or homologs thereof; (ii) antibodies to an aforementioned peptide; (iii) nucleic acids encoding an aforementioned peptide; (iv) administration of antisense nucleic acid and nucleic acids that are "dysfunctional" (i.e., due to a heterologous insertion within the coding sequences of coding sequences to an aforementioned peptide) that are utilized to "knockout" endogenous function of an aforementioned peptide by homologous recombination (see, e.g., Capecchi, 1989.
  • modulators i.e., inhibitors, agonists and antagonists, including additional peptide mimetic of the invention or antibodies specific to a peptide of the invention
  • modulators i.e., inhibitors, agonists and antagonists, including additional peptide mimetic of the invention or antibodies specific to a peptide of the invention
  • Therapeutics that increase (i.e., are agonists to) activity may be administered in a therapeutic or prophylactic manner.
  • Therapeutics that may be utilized include, but are not limited to, an aforementioned peptide, or analogs, derivatives, fragments or homologs thereof; or an agonist that increases bioavailability.
  • Increased or decreased levels can be readily detected by quantifying peptide and/or RNA, by obtaining a patient tissue sample (e.g., from biopsy tissue) and assaying it in vitro for RNA or peptide levels, structure and/or activity of the expressed peptides (or mRNAs of an aforementioned peptide).
  • tissue sample e.g., from biopsy tissue
  • assaying it in vitro for RNA or peptide levels, structure and/or activity of the expressed peptides (or mRNAs of an aforementioned peptide).
  • Methods that are well-known within the art include, but are not limited to, immunoassays (e.g., by Western blot analysis, immunoprecipitation followed by sodium dodecyl sulfate (SDS) polyacrylamide gel electrophoresis, immunocytochemistry, etc.) and/or hybridization assays to detect expression of mRNAs (e.g., Northern assays, dot blots, in situ hybridization, and the like).
  • immunoassays e.g., by Western blot analysis, immunoprecipitation followed by sodium dodecyl sulfate (SDS) polyacrylamide gel electrophoresis, immunocytochemistry, etc.
  • hybridization assays to detect expression of mRNAs e.g., Northern assays, dot blots, in situ hybridization, and the like.
  • the invention provides a method for preventing, in a subject, a disease or condition associated with an abenant NOVX expression or activity, by administering to the subject an agent that modulates NOVX expression or at least one NOVX activity.
  • Subjects at risk for a disease that is caused or contributed to by abenant NOVX expression or activity can be identified by, for example, any or a combination of diagnostic or prognostic assays as described herein.
  • Administration of a prophylactic agent can occur prior to the manifestation of symptoms characteristic of the NOVX abenancy, such that a disease or disorder is prevented or, alternatively, delayed in its progression.
  • a NOVX agonist or NOVX antagonist agent can be used for treating the subject.
  • the appropriate agent can be determined based on screening assays described herein. The prophylactic methods of the invention are further discussed in the following subsections. Therapeutic Methods
  • the modulatory method of the invention involves contacting a cell with an agent that modulates one or more of the activities of NOVX protein activity associated with the cell.
  • An agent that modulates NOVX protein activity can be an agent as described herein, such as a nucleic acid or a protein, a naturally-occurring cognate ligand of a NOVX protein, a peptide, a NOVX peptidomimetic, or other small molecule.
  • the agent stimulates one or more NOVX protein activity. Examples of such stimulatory agents include active NOVX protein and a nucleic acid molecule encoding NOVX that has been introduced into the cell.
  • the agent inhibits one or more NOVX protein activity.
  • inhibitory agents include antisense NOVX nucleic acid molecules and anti-NOVX antibodies. These modulatory methods can be performed in vitro (e.g., by culturing the cell with the agent) or, alternatively, in vivo (e.g., by administering the agent to a subject).
  • the invention provides methods of teeating an individual afflicted with a disease or disorder characterized by abenant expression or activity of a NOVX protein or nucleic acid molecule.
  • the method involves administering an agent (e.g., an agent identified by a screening assay described herein), or combination of agents that modulates (e.g., up-regulates or down-regulates) NOVX expression or activity.
  • an agent e.g., an agent identified by a screening assay described herein
  • the method involves administering a NOVX protein or nucleic acid molecule as therapy to compensate for reduced or abenant NOVX expression or activity.
  • Stimulation of NOVX activity is desirable in situations in which NOVX is abnormally downregulated and/or in which increased NOVX activity is likely to have a beneficial effect.
  • a subject has a disorder characterized by abenant cell proliferation and/or differentiation (e.g., cancer or immune associated disorders).
  • a gestational disease e.g., preclampsia.
  • suitable in vitro or in vivo assays are performed to determine the effect of a specific Therapeutic and whether its administration is indicated for treatment of the affected tissue.
  • in vitro assays may be performed with representative cells of the type(s) involved in the patient's disorder, to determine if a given Therapeutic exerts the desired effect upon the cell type(s).
  • Compounds for use in therapy may be tested in suitable animal model systems including, but not limited to rats, mice, chicken, cows, monkeys, rabbits, and the like, prior to testing in human subjects. Similarly, for in vivo testing, any of the animal model system known in the art may be used prior to administration to human subjects.
  • the NOVX nucleic acids and proteins of the invention are useful in potential prophylactic and therapeutic applications implicated in a variety of disorders including, but not limited to: metabolic disorders, diabetes, obesity, infectious disease, anorexia, cancer-associated cancer, neurodegenerative disorders, Alzheimer's Disease, Parkinson's Disorder, immune disorders, hematopoietic disorders, and the various dyslipidemias, metabolic disturbances associated with obesity, the metabolic syndrome X and wasting disorders associated with chronic diseases and various cancers.
  • a cDNA encoding the NOVX protein of the invention may be useful in gene therapy, and the protein may be useful when administered to a subject in need thereof.
  • compositions of the invention will have efficacy for treatment of patients suffering from: metabolic disorders, diabetes, obesity, infectious disease, anorexia, cancer-associated cachexia, cancer, neurodegenerative disorders, Alzheimer's Disease, Parkinson's Disorder, immune disorders, hematopoietic disorders, and the various dyslipidemias.
  • Both the novel nucleic acid encoding the NOVX protein, and the NOVX protein of the invention, or fragments thereof, may also be useful in diagnostic applications, wherein the presence or amount of the nucleic acid or the protein are to be assessed.
  • a further use could be as an anti-bacterial molecule (i.e., some peptides have been found to possess anti-bacterial properties).
  • These materials are further useful in the generation of antibodies, which immunospecifically-bind to the novel substances of the invention for use in therapeutic or diagnostic methods.
  • Example A Polynucleotide And Polypeptide Sequences, And Homology Data Example 1.
  • the NOV1 clone was analyzed, and the nucleotide and encoded polypeptide sequences are shown in Table 1 A.
  • MAPSA AIC L GG X.LHGGSSGPSPGPSVPR RLSYRGAWRKPSSTMMETFSRY LSANRSAIF GPQGS NLQAMY DEYRDRLF GG DALYSLR DQAWPDPREVLWPPQPGQREECVRKGRDP TECANFVRVLQPHNR TH LACGTGAFQPTCALITVGHRGEHVLHLEPGSVESGRGRCPHEPSRPFASTFIDGELYTGLTADF GREAM IFRSGGPRPALRSDSDQSL HDPRFVMAARIPENSDQDNDKVYFFFSETVPSPDGGSNHV VSRVGRVCVNDA GGQRVLVNK STFLKAR VCSVPGPGGAETHFDQLEDVFL WPKA.GKS EVYALFSTVSAVFQGFAVCVYHMA DI EVFNGPFAHRDGPQHQWGPYGGKVPFPRPGVCPSKMTAQPGRPFGSTKDYPDEVLQFARAHPLMFWPVRP RHG
  • NOVla APSA AIC L GGL LHGGSSGPSPGPSVPR R SYRGAWRKPSSTMWMETFSRYL S
  • NOVla SEQ ID NO: 2
  • NOVlb SEQ ID NO: 4
  • PSG a new signal peptide prediction method
  • N-region length 0; pos.chg 0; neg.chg 0 H-region: length 31; peak value 9.35 PSG score : 4.95
  • GvH von Heijne's method for signal seq. recognition
  • GvH score (threshold: -2.1): 1.50 possible cleavage site: between 22 and 23
  • Gavel prediction of cleavage sites for mitochondrial preseq R-2 motif at 73 RS
  • NUCDISC discrimination of nuclear localization signals pat : none pat7: PSLGKRR (3) at 570 bipartite : none content of basic residues: 11.4% NLS Score: -0.22
  • KDEL ER retention motif in the C-terminus : none
  • SKL peroxisomal targeting signal in the C-terminus: none
  • VAC possible vacuolar targeting motif
  • Actinia-type actin-binding motif type 1: none type 2 : none NMYR: N-myristoylation pattern : none
  • Prenylation motif none me YQRL: transport motif from cell surface to Golgi: none
  • NNCN Reinhardt's method for Cytoplasmic/Nuclear discrimination Prediction: cytoplasmic Reliability: 89
  • COIL Lupas's algorithm to detect coiled-coil regions total: 0 residues
  • NOVla protein was found to have homology to the proteins shown in the BLASTP data in Table IE.
  • NOV2h NOV2a GHLHPPLGVSGSS CLACVSWMPCGFSPSPVAHHLVPGPPDXPAQQLRCGWXVGG LLSL.
  • NOV2a NGPFRYQENPRAAWLPIANPIPNFQCGXLPEIGPNENLXERSLQDAQRLFLMSEAVQPVX
  • PSG a new signal peptide prediction method
  • N-region length 11; pos.chg 1; neg.chg 1 H-region: length 5; peak value -8.91 PSG score: -13.31
  • GvH von Heijne's method for signal seq. recognition
  • GvH score (threshold: -2.1): -7.G5 possible cleavage site: between 53 and 5
  • Gavel prediction of cleavage sites for mitochondrial preseq R-2 motif at 104 LRAlPG
  • NUCDISC discrimination of nuclear localization signals pat4 : none pat7 : none bipartite: none N content of basic residues: 9.3% NLS Score: -0.47
  • KDEL ER retention motif in the C-terminus : none
  • SKL peroxisomal targeting signal in the C-terminus: none
  • VAC possible vacuolar targeting motif
  • Actinin-type actin-binding motif type 1 : none type 2 : none
  • NMYR N-myristoylation pattern : none
  • Prenylation motif none memYQRL: transport motif from cell surface to Golgi: none
  • COIL Lupas's algorithm to detect coiled-coil regions total: 0 residues
  • NOV2a protein was found to have homology to the proteins shown in the BLASTP data in Table 2E.
  • the NOV3 clone was analyzed, and the nucleotide and encoded polypeptide sequences are shown in Table 3 A.
  • VAC possible vacuolar targeting motif
  • Actinin-type actin-binding motif type 1 : none type 2 : none
  • NMYR N-myristoylation pattern : none
  • Prenylation motif none memYQRL: transport motif from cell surface to Golgi: none
  • COIL Lupas's algorithm to detect coiled-coil regions total : 0 residues
  • NOV3a protein was found to have homology to the proteins shown in the BLASTP data in Table 3D.
  • the NOV4 clone was analyzed, and the nucleotide and encoded polypeptide sequences are shown in Table 4A.
  • NOV4a SEQ ID NO: 26
  • NOV4b SEQ ID NO: 28
  • PSG a new signal peptide prediction method
  • N-region length 4; pos.chg 1; neg.chg 0 H-region: length 18; peak value 11.91 PSG score: 7.51
  • GvH von Heijne's method for signal seq. recognition
  • GvH score (threshold: -2.1): 4.21 possible cleavage site: between 27 and 28
  • NUCDISC discrimination of nuclear localization signals pat4 : none pat7: none bipartite: none content of basic residues: 9.5% NLS Score: -0.47
  • KDEL ER retention motif in the C-terminus : none
  • VAC possible vacuolar targeting motif
  • Actinin-type actin-binding motif type 1: none type 2 : none
  • NMYR N-myristoylation pattern : none
  • Prenylation motif none memYQRL: transport motif from cell surface to Golgi: none
  • NNCN Reinhardt's method for Cytoplasmic/Nuclear discrimination Prediction: cytoplasmic Reliability: 76.7
  • COIL Lupas's algorithm to detect coiled-coil regions total : 0 residues
  • NOV4a protein was found to have homology to the proteins shown in the BLASTP data in Table 4E.
  • NOV5 clone was analyzed, and the nucleotide and encoded polypeptide sequences are shown in Table 5A. Table 5A. NOV5 Sequence Analysis

Abstract

Disclosed herein are nucleic acid sequences that encode novel polypeptides. Also disclosed are polypeptides encoded by these nucleic acid sequences, and antibodies that immunospecifically bind to the polypeptide, as well as derivatives, variants, mutants, or fragments of the novel polypeptide, polynucleotide, or antibody specific to the polypeptide. The invention further discloses therapeutic, diagnostic and research methods for diagnosis, treatment, and prevention of disorders involving any one of these novel human nucleic acids and proteins.

Description

THERAPEUTIC POLYPEPTIDES, NUCLEIC ACIDS ENCODING SAME, AND METHODS OF USE
FIELD OF THE INVENTION
The present invention relates to novel polypeptides, and the nucleic acids encoding them, having properties related to stimulation of biochemical or physiological responses in a cell, a tissue, an organ or an organism. More particularly, the novel polypeptides are gene products of novel genes, or are specified biologically active fragments or derivatives thereof. Methods of use encompass diagnostic and prognostic assay procedures as well as methods of treating diverse pathological conditions.
BACKGROUND OF THE INVENTION
Eukaryotic cells are characterized by biochemical and physiological processes, which under normal conditions are exquisitely balanced to achieve the preservation and propagation of the cells. When such cells are components of multicellular organisms such as vertebrates or, more particularly, organisms such as mammals, the regulation of the biochemical and physiological processes involves intricate signaling pathways. Frequently, such signaling pathways involve extracellular signaling proteins, cellular receptors that bind the signaling proteins and signal transducing components located within the cells. Signaling proteins may be classified as endocrine effectors, paracrine effectors or autocrine effectors. Endocrine effectors are signaling molecules secreted by a given organ into the circulatory system, which are then transported to a distant target organ or tissue. The target cells include the receptors for the endocrine effector, and when the endocrine effector binds, a signaling cascade is induced. Paracrine effectors involve secreting cells and receptor cells in close proximity to each other, for example, two different classes of cells in the same tissue or organ. One class of cells secretes the paracrine effector, which then reaches the second class of cells, for example by diffusion through the extracellular fluid. The second class of cells contains the receptors for the paracrine effector; binding of the effector results in induction of the signaling cascade that elicits the corresponding biochemical or physiological effect. Autocrine effectors are highly analogous to paracrine effectors, except that the same cell type that secretes the autocrine effector also contains the receptor. Thus the autocrine effector binds to receptors on the same cell, or on identical neighboring cells. The binding process then elicits the characteristic biochemical or physiological effect. Signaling processes may elicit a variety of effects on cells and tissues including, by way of nonlimiting example, induction of cell or tissue proliferation, suppression of growth or proliferation, induction of differentiation or maturation of a cell or tissue, and suppression of differentiation or maturation of a cell or tissue.
Many pathological conditions involve dysregulation of expression of important effector proteins. In certain classes of pathologies the dysregulation is manifested as diminished or suppressed level of synthesis and secretion of protein effectors. In other classes of pathologies the dysregulation is manifested as increased or up-regulated level of synthesis and secretion of protein effectors. In a clinical setting a subject may be suspected of suffering from a condition brought on by altered or mis-regulated levels of a protein effector of interest. Therefore there is a need to assay for the level of the protein effector of interest in a biological sample from such a subject, and to compare the level with that characteristic of a nonpathological condition. There also is a need to provide the protein effector as a product of manufacture. Administration of the effector to a subject in need thereof is useful in treatment of the pathological condition. Accordingly, there is a need for a method of treatment of a pathological condition brought on by a diminished or suppressed levels of the protein effector of interest. In addition, there is a need for a method of treatment of a pathological condition brought on by a increased or up-regulated levels of the protein effector of interest.
Antibodies are multichain proteins that bind specifically to a given antigen, and bind poorly, or not at all, to substances deemed not to be cognate antigens. Antibodies are comprised of two short chains termed light chains and two long chains termed heavy chains. These chains are constituted of immunoglobulin domains, of which generally there are two classes: one variable domain per chain, one constant domain in light chains, and three or more constant domains in heavy chains. The antigen-specific portion of the immunoglobulin molecules resides in the variable domains; the variable domains of one light chain and one heavy chain associate with each other to generate the antigen-binding moiety. Antibodies that bind immunospecifically to a cognate or target antigen bind with high affinities. Accordingly, they are useful in assaying specifically for the presence of the antigen in a sample. In addition, they have the potential of inactivating the activity of the antigen.
Therefore there is a need to assay for the level of a protein effector of interest in a biological sample from such a subject, and to compare this level with that characteristic of a nonpathological condition. In particular, there is a need for such an assay based on the use of an antibody that binds immunospecifically to the antigen. There further is a need to inhibit the activity of the protein effector in cases where a pathological condition arises from elevated or excessive levels of the effector based on the use of an antibody that binds immunospecifically to the effector. Thus, there is a need for the antibody as a product of manufacture. There further is a need for a method of treatment of a pathological condition brought on by an elevated or excessive level of the protein effector of interest based on administering the antibody to the subject. SUMMARY OF THE INVENTION
The invention is based in part upon the discovery of isolated polypeptides including amino acid sequences selected from mature forms of the amino acid sequences selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141. The novel nucleic acids and polypeptides are referred to herein as NOVX, or NOV1, NOV2, NOV3, etc., nucleic acids and polypeptides. These nucleic acids and polypeptides, as well as derivatives, homologs, analogs and fragments thereof, will hereinafter be collectively designated as "NOVX" nucleic acid or polypeptide sequences. The invention also is based in part upon variants of a mature form of the amino acid sequence selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141, wherein any amino acid in the mature form is changed to a different amino acid, provided that no more than 15% of the amino acid residues in the sequence of the mature form are so changed. In another embodiment, the invention includes the amino acid sequences selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141. In another embodiment, the invention also comprises variants of the amino acid sequence selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141, wherein any amino acid specified in the chosen sequence is changed to a different amino acid, provided that no more than 15% of the amino acid residues in the sequence are so changed. The invention also involves fragments of any of the mature forms of the amino acid sequences selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141, or any other amino acid sequence selected from this group. The invention also comprises fragments from these groups in which up to 15% of the residues are changed.
In another embodiment, the invention encompasses polypeptides that are naturally occurring allelic variants of the sequence selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141. These allelic variants include amino acid sequences that are the translations of nucleic acid sequences differing by a single nucleotide from nucleic acid sequences selected from the group consisting of SEQ ID NOS: 2n-l, wherein n is an integer between 1 and 141. The variant polypeptide where any amino acid changed in the chosen sequence is changed to provide a conservative substitution. In another embodiment, the invention comprises a pharmaceutical composition involving a polypeptide with an amino acid sequence selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141, and a pharmaceutically acceptable carrier. In another embodiment, the invention involves a kit, including, in one or more containers, this pharmaceutical composition.
In another embodiment, the invention includes the use of a therapeutic in the manufacture of a medicament for treating a syndrome associated with a human disease, the disease being selected from a pathology associated with a polypeptide with an amino acid sequence selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141, wherein said therapeutic is the polypeptide selected from this group.
In another embodiment, the invention comprises a method for determining the presence or amount of a polypeptide with an amino acid sequence selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141, in a sample, the method involving providing the sample; introducing the sample to an antibody that binds immunospecifically to the polypeptide; and determining the presence or amount of antibody bound to the polypeptide, thereby determining the presence or amount of polypeptide in the sample.
In another embodiment, the invention includes a method for determining the presence of or predisposition to a disease associated with altered levels of a polypeptide with an amino acid sequence selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141, in a first mammalian subject, the method involving measuring the level of expression of the polypeptide in a sample from the first mammalian subject; and comparing the amount of the polypeptide in this sample to the amount of the polypeptide present in a control sample from a second mammalian subject known not to have, or not to be predisposed to, the disease, wherein an alteration in the expression level of the polypeptide in the first subject as compared to the control sample indicates the presence of or predisposition to the disease.
In another embodiment, the invention involves a method of identifying an agent that binds to a polypeptide with an amino acid sequence selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141 , the method including introducing the polypeptide to the agent; and determining whether the agent binds to the polypeptide. The agent could be a cellular receptor or a downstream effector. In another embodiment, the invention involves a method for identifying a potential therapeutic agent for use in treatment of a pathology, wherein the pathology is related to aberrant expression or aberrant physiological interactions of a polypeptide with an amino acid sequence selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141 , the method including providing a cell expressing the polypeptide of the invention and having a property or function ascribable to the polypeptide; contacting the cell with a composition comprising a candidate substance; and determining whether the substance alters the property or function ascribable to the polypeptide; whereby, if an alteration observed in the presence of the substance is not observed when the cell is contacted with a composition devoid of the substance, the substance is identified as a potential therapeutic agent.
In another embodiment, the invention involves a method for screening for a modulator of activity or of latency or predisposition to a pathology associated with a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141 , the method including administering a test compound to a test animal at increased risk for a pathology associated with the polypeptide of the invention, wherein the test animal recombinantly expresses the polypeptide of the invention; measuring the activity of the polypeptide in the test animal after administering the test compound; and comparing the activity of the protein in the test animal with the activity of the polypeptide in a control animal not administered the polypeptide, wherein a change in the activity of the polypeptide in the test animal relative to the control animal indicates the test compound is a modulator of latency of, or predisposition to, a pathology associated with the polypeptide of the invention. The recombinant test animal could express a test protein transgene or express the transgene under the control of a promoter at an increased level relative to a wild-type test animal The promoter may or may not b the native gene promoter of the transgene.
In another embodiment, the invention involves a method for modulating the activity of a polypeptide with an amino acid sequence selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141, the method including introducing a cell sample expressing the polypeptide with a compound that binds to the polypeptide in an amount sufficient to modulate the activity of the polypeptide.
In another embodiment, the invention involves a method of treating or preventing a pathology associated with a polypeptide with an amino acid sequence selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141, the method including administering the polypeptide to a subject in which such treatment or prevention is desired in an amount sufficient to treat or prevent the pathology in the subject. The subject could be human. In another embodiment, the invention involves a method of treating a pathological state in a mammal, the method including administering to the mammal a polypeptide in an amount that is sufficient to alleviate the pathological state, wherein the polypeptide is a polypeptide having an amino acid sequence at least 95% identical to a polypeptide having the amino acid sequence selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141, or a biologically active fragment thereof.
In another embodiment, the invention involves an isolated nucleic acid molecule comprising a nucleic acid sequence encoding a polypeptide having an amino acid sequence selected from the group consisting of a mature form of the amino acid sequence given SEQ ID NO:2n, wherein n is an integer between 1 and 141, a variant of a mature form of the amino acid sequence selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141, wherein any amino acid in the mature form of the chosen sequence is changed to a different amino acid, provided that no more than 15% of the amino acid residues in the sequence of the mature form are so changed; the amino acid sequence selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141, a variant of the amino acid sequence selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141, in which any amino acid specified in the chosen sequence is changed to a different amino acid, provided that no more than 15% of the amino acid residues in the sequence are so changed; a nucleic acid fragment encoding at least a portion of a polypeptide comprising the amino acid sequence selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141, or any variant of the polypeptide wherein any amino acid of the chosen sequence is changed to a different amino acid, provided that no more than 10% of the amino acid residues in the sequence are so changed; and the complement of any of the nucleic acid molecules. In another embodiment, the invention comprises an isolated nucleic acid molecule having a nucleic acid sequence encoding a polypeptide comprising an amino acid sequence selected from the group consisting of a mature form of the amino acid sequence given SEQ ID NO:2n, wherein n is an integer between 1 and 141, wherein the nucleic acid molecule comprises the nucleotide sequence of a naturally occurring allelic nucleic acid variant.
In another embodiment, the invention involves an isolated nucleic acid molecule including a nucleic acid sequence encoding a polypeptide having an amino acid sequence selected from the group consisting of a mature form of the amino acid sequence given SEQ ID NO:2n, wherein n is an integer between 1 and 141, that encodes a variant polypeptide, wherein the variant polypeptide has the polypeptide sequence of a naturally occurring polypeptide variant.
In another embodiment, the invention comprises an isolated nucleic acid molecule having a nucleic acid sequence encoding a polypeptide comprising an amino acid sequence selected from the group consisting of a mature form of the amino acid sequence given SEQ ID NO:2n, wherein n is an integer between 1 and 141 , wherein the nucleic acid molecule differs by a single nucleotide from a nucleic acid sequence selected from the group consisting of SEQ ID NOS: 2n-l, wherein n is an integer between 1 and 141. In another embodiment, the invention includes an isolated nucleic acid molecule having a nucleic acid sequence encoding a polypeptide including an amino acid sequence selected from the group consisting of a mature form of the amino acid sequence given SEQ ID NO:2n, wherein n is an integer between 1 and 141, wherein the nucleic acid molecule comprises a nucleotide sequence selected from the group consisting of the nucleotide sequence selected from the group consisting of SEQ ID NO:2n-l , wherein n is an integer between 1 and 141, a nucleotide sequence wherein one or more nucleotides in the nucleotide sequence selected from the group consisting of SEQ ID NO:2n-l, wherein n is an integer between 1 and 141, is changed from that selected from the group consisting of the chosen sequence to a different nucleotide provided that no more than 15% of the nucleotides are so changed; a nucleic acid fragment of the sequence selected from the group consisting of SEQ ID NO:2n-l, wherein n is an integer between 1 and 141, and a nucleic acid fragment wherein one or more nucleotides in the nucleotide sequence selected from the group consisting of SEQ ID NO:2n-l, wherein n is an integer between 1 and 141, is changed from that selected from the group consisting of the chosen sequence to a different nucleotide provided that no more than 15% of the nucleotides are so changed. In another embodiment, the invention includes an isolated nucleic acid molecule having a nucleic acid sequence encoding a polypeptide including an amino acid sequence selected from the group consisting of a mature form of the amino acid sequence given SEQ ID NO:2n, wherein n is an integer between 1 and 141, wherein the nucleic acid molecule hybridizes under stringent conditions to the nucleotide sequence selected from the group consisting of SEQ ID NO:2n-l, wherein n is an integer between 1 and 141, or a complement of the nucleotide sequence. In another embodiment, the invention includes an isolated nucleic acid molecule having a nucleic acid sequence encoding a polypeptide including an amino acid sequence selected from the group consisting of a mature form of the amino acid sequence given SEQ ID NO:2n, wherein n is an integer between 1 and 141, wherein the nucleic acid molecule has a nucleotide sequence in which any nucleotide specified in the coding sequence of the chosen nucleotide sequence is changed from that selected from the group consisting of the chosen sequence to a different nucleotide provided that no more than 15% of the nucleotides in the chosen coding sequence are so changed, an isolated second polynucleotide that is a complement of the first polynucleotide, or a fragment of any of them. In another embodiment, the invention includes a vector involving the nucleic acid molecule having a nucleic acid sequence encoding a polypeptide including an amino acid sequence selected from the group consisting of a mature form of the amino acid sequence given SEQ ID NO:2n, wherein n is an integer between 1 and 141. This vector can have a promoter operably linked to the nucleic acid molecule. This vector can be located within a cell.
In another embodiment, the invention involves a method for determining the presence or amount of a nucleic acid molecule having a nucleic acid sequence encoding a polypeptide including an amino acid sequence selected from the group consisting of a mature form of the amino acid sequence given SEQ ID NO:2n, wherein n is an integer between 1 and 141, in a sample, the method including providing the sample; introducing the sample to a probe that binds to the nucleic acid molecule; and determining the presence or amount of the probe bound to the nucleic acid molecule, thereby determining the presence or amount of the nucleic acid molecule in the sample. The presence or amount of the nucleic acid molecule is used as a marker for cell or tissue type. The cell type can be cancerous.
In another embodiment, the invention involves a method for determining the presence of or predisposition for a disease associated with altered levels of a nucleic acid molecule having a nucleic acid sequence encoding a polypeptide including an amino acid sequence selected from the group consisting of a mature form of the amino acid sequence given SEQ ID NO:2n, wherein n is an integer between 1 and 141, in a first mammalian subject, the method including measuring the amount of the nucleic acid in a sample from the first mammalian subject; and comparing the amount of the nucleic acid in the sample of step (a) to the amount of the nucleic acid present in a control sample from a second mammalian subject known not to have or not be predisposed to, the disease; wherein an alteration in the level of the nucleic acid in the first subject as compared to the control sample indicates the presence of or predisposition to the disease.
Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, suitable methods and materials are described below. All publications, patent applications, patents, and other references mentioned herein are incoφorated by reference in their entirety. In the case of conflict, the present specification, including definitions, will control. In addition, the materials, methods, and examples are illustrative only and are not intended to be limiting.
Other features and advantages of the invention will be apparent from the following detailed description and claims.
DETAILED DESCRIPTION OF THE INVENTION
The present invention provides novel nucleotides and polypeptides encoded thereby. Included in the invention are the novel nucleic acid sequences, their encoded polypeptides, antibodies, and other related compounds. The sequences are collectively referred to herein as "NOVX nucleic acids" or "NOVX polynucleotides" and the corresponding encoded polypeptides are referred to as "NOVX polypeptides" or "NOVX proteins." Unless indicated otherwise, "NOVX" is meant to refer to any of the novel sequences disclosed herein. Table A provides a summary of the NOVX nucleic acids and their encoded polypeptides. TABLE A. Sequences and Corresponding SEQ TD Numbers
Figure imgf000013_0001
Figure imgf000014_0001
Figure imgf000015_0001
Figure imgf000016_0001
Figure imgf000017_0001
Figure imgf000018_0001
Table A indicates the homology of NOVX polypeptides to known protein families. Thus, the nucleic acids and polypeptides, antibodies and related compounds according to the invention corresponding to a NOVX as identified in column 1 of Table A will be useful in therapeutic and diagnostic applications implicated in, for example, pathologies and disorders associated with the known protein families identified in column 5 of Table A.
Pathologies, diseases, disorders, conditions and the like that are associated with NOVX sequences include, but are not limited to, e.g., cardiomyopathy, atherosclerosis, hypertension, congenital heart defects, aortic stenosis, atrial septal defect (ASD), atrioventricular (A-V) canal defect, ductus arteriosus, pulmonary stenosis, subaortic stenosis, ventricular septal defect (VSD), valve diseases, tuberous sclerosis, scleroderma, obesity, metabolic disturbances associated with obesity, transplantation, adrenoleukodystrophy, congenital adrenal hyperplasia, prostate cancer, diabetes, metabolic disorders, neoplasm; adenocarcinoma, lymphoma, uterus cancer, fertility, hemophilia, hypercoagulation, idiopathic thrombocytopenic purpura, immunodeficiencies, graft versus host disease, AIDS, bronchial asthma, Crohn's disease; multiple sclerosis, treatment of Albright Hereditary Ostoeodystrophy, infectious disease, anorexia, cancer-associated cachexia, cancer, neurodegenerative disorders, Alzheimer's Disease, Parkinson's Disorder, immune disorders, hematopoietic disorders, and the various dyslipidemias, the metabolic syndrome X and wasting disorders associated with chronic diseases and various cancers, as well as conditions such as transplantation and fertility.
NOVX nucleic acids and their encoded polypeptides are useful in a variety of applications and contexts. The various NOVX nucleic acids and polypeptides according to the invention are useful as novel members of the protein families according to the presence of domains and sequence relatedness to previously described proteins. Additionally, NOVX nucleic acids and polypeptides can also be used to identify proteins that are members of the family to which the NOVX polypeptides belong.
Consistent with other known members of the family of proteins, identified in column 5 of Table A, the NOVX polypeptides of the present invention show homology to, and contain domains that are characteristic of, other members of such protein families. Details of the sequence relatedness and domain analysis for each NOVX are presented in Example A. The NOVX nucleic acids and polypeptides can also be used to screen for molecules, which inhibit or enhance NOVX activity or function. Specifically, the nucleic acids and polypeptides according to the invention may be used as targets for the identification of small molecules that modulate or inhibit diseases associated with the protein families listed in Table A.
The NOVX nucleic acids and polypeptides are also useful for detecting specific cell types. Details of the expression analysis for each NOVX are presented in Example C. Accordingly, the NOVX nucleic acids, polypeptides, antibodies and related compounds according to the invention will have diagnostic and therapeutic applications in the detection of a variety of diseases with differential expression in normal vs. diseased tissues, e.g., detection of a variety of cancers.
Additional utilities for NOVX nucleic acids and polypeptides according to the invention are disclosed herein.
NOVX clones NOVX nucleic acids and their encoded polypeptides are useful in a variety of applications and contexts. The various NOVX nucleic acids and polypeptides according to the invention are useful as novel members of the protein families according to the presence of domains and sequence relatedness to previously described proteins. Additionally, NOVX nucleic acids and polypeptides can also be used to identify proteins that are members of the family to which the NOVX polypeptides belong.
The NOVX genes and their corresponding encoded proteins are useful for preventing, treating or ameliorating medical conditions, e.g., by protein or gene therapy. Pathological conditions can be diagnosed by determining the amount of the new protein in a sample or by determining the presence of mutations in the new genes. Specific uses are described for each of the NOVX genes, based on the tissues in which they are most highly expressed. Uses include developing products for the diagnosis or treatment of a variety of diseases and disorders.
The NOVX nucleic acids and proteins of the invention are useful in potential diagnostic and therapeutic applications and as a research tool. These include serving as a specific or selective nucleic acid or protein diagnostic and/or prognostic marker, wherein the presence or amount of the nucleic acid or the protein are to be assessed, as well as potential therapeutic applications such as the following: (i) a protein therapeutic, (ii) a small molecule drug target, (iii) an antibody target (therapeutic, diagnostic, drug targeting/cytotoxic antibody), (iv) a nucleic acid useful in gene therapy (gene delivery/gene ablation), and (v) a composition promoting tissue regeneration in vitro and in vivo (vi) a biological defense weapon. In one specific embodiment, the invention includes an isolated polypeptide comprising an amino acid sequence selected from the group consisting of: (a) a mature form of the amino acid sequence selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141, (b) a variant of a mature form of the amino acid sequence selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141, wherein any amino acid in the mature form is changed to a different amino acid, provided that no more than 15% of the amino acid residues in the sequence of the mature form are so changed; (c) an amino acid sequence selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141, (d) a variant of the amino acid sequence selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141, wherein any amino acid specified in the chosen sequence is changed to a different amino acid, provided that no more than 15% of the amino acid residues in the sequence are so changed; and (e) a fragment of any of (a) through (d).
In another specific embodiment, the invention includes an isolated nucleic acid molecule comprising a nucleic acid sequence encoding a polypeptide comprising an amino acid sequence selected from the group consisting of: (a) a mature form of the amino acid sequence given SEQ ID NO:2n, wherein n is an integer between 1 and 141; (b) a variant of a mature form of the amino acid sequence selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141, wherein any amino acid in the mature form of the chosen sequence is changed to a different amino acid, provided that no more than 15% of the amino acid residues in the sequence of the mature form are so changed; (c) the amino acid sequence selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141 ; (d) a variant of the amino acid sequence selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141 , in which any amino acid specified in the chosen sequence is changed to a different amino acid, provided that no more than 15% of the amino acid residues in the sequence are so changed; (e) a nucleic acid fragment encoding at least a portion of a polypeptide comprising the amino acid sequence selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141, or any variant of said polypeptide wherein any amino acid of the chosen sequence is changed to a different amino acid, provided that no more than 10% of the amino acid residues in the sequence are so changed; and (f) the complement of any of said nucleic acid molecules. In yet another specific embodiment, the invention includes an isolated nucleic acid molecule, wherein said nucleic acid molecule comprises a nucleotide sequence selected from the group consisting of: (a) the nucleotide sequence selected from the group consisting of SEQ ID NO:2n-l, wherein n is an integer between 1 and 141; (b) a nucleotide sequence wherein one or more nucleotides in the nucleotide sequence selected from the group consisting of SEQ ID NO:2n-l, wherein n is an integer between 1 and 141, is changed from that selected from the group consisting of the chosen sequence to a different nucleotide provided that no more than 15% of the nucleotides are so changed; (c) a nucleic acid fragment of the sequence selected from the group consisting of SEQ ID NO:2n-l, wherein n is an integer between 1 and 141; and (d) a nucleic acid fragment wherein one or more nucleotides in the nucleotide sequence selected from the group consisting of SEQ ID NO:2n-l, wherein n is an integer between 1 and 141, is changed from that selected from the group consisting of the chosen sequence to a different nucleotide provided that no more than 15% of the nucleotides are so changed.
NOVX Nucleic Acids and Polypeptides One aspect of the invention pertains to isolated nucleic acid molecules that encode
NOVX polypeptides or biologically active portions thereof. Also included in the invention are nucleic acid fragments sufficient for use as hybridization probes to identify NOVX-encoding nucleic acids (e.g., NOVX mRNAs) and fragments for use as PCR primers for the amplification and/or mutation of NOVX nucleic acid molecules. As used herein, the term "nucleic acid molecule" is intended to include DNA molecules (e.g., cDNA or genomic DNA), RNA molecules (e.g., mRNA), analogs of the DNA or RNA generated using nucleotide analogs, and derivatives, fragments and homologs thereof. The nucleic acid molecule may be single-stranded or double-stranded, but preferably is comprised double-stranded DNA. A NOVX nucleic acid can encode a mature NOVX polypeptide. As used herein, a
"mature" form of a polypeptide or protein disclosed in the present invention is the product of a naturally occurring polypeptide, precursor form, or proprotein. The naturally occurring polypeptide, precursor or proprotein includes, by way of nonlimiting example, the full-length gene product encoded by the corresponding gene. Alternatively, it may be defined as the polypeptide, precursor or proprotein encoded by an ORF described herein. The product "mature" form arises, by way of nonlimiting example, as a result of one or more naturally occurring processing steps that may take place within the cell (e.g., host cell) in which the gene product arises. Examples of such processing steps leading to a "mature" form of a polypeptide or protein include the cleavage of the N-terminal methionine residue encoded by the initiation codon of an ORF or the proteolytic cleavage of a signal peptide or leader sequence. Thus a mature form arising from a precursor polypeptide or protein that has residues 1 to N, where residue 1 is the N-terminal methionine, would have residues 2 through N remaining after removal of the N-terminal methionine. Alternatively, a mature form arising from a precursor polypeptide or protein having residues 1 to N, in which an N-terminal signal sequence from residue 1 to residue M is cleaved, would have the residues from residue M+l to residue N remaining. Further as used herein, a "mature" form of a polypeptide or protein may arise from a post-translational modification step other than a proteolytic cleavage event. Such additional processes include, by way of non-limiting example, glycosylation, myristylation or phosphorylation. In general, a mature polypeptide or protein may result from the operation of only one of these processes, or a combination of any of them. The term "probe", as utilized herein, refers to nucleic acid sequences of variable length, preferably between at least about 10 nucleotides (nt), about 100 nt, or as many as approximately, e.g., 6,000 nt, depending upon the specific use. Probes are used in the detection of identical, similar, or complementary nucleic acid sequences. Longer length probes are generally obtained from a natural or recombinant source, are highly specific, and much slower to hybridize than shorter-length oligomer probes. Probes may be single- or double-stranded and designed to have specificity in PCR, membrane-based hybridization technologies, or ELISA-like technologies.
The term "isolated" nucleic acid molecule, as used herein, is a nucleic acid that is separated from other nucleic acid molecules which are present in the natural source of the nucleic acid. Preferably, an "isolated" nucleic acid is free of sequences which naturally flank the nucleic acid (i.e., sequences located at the 5'- and 3 '-termini of the nucleic acid) in the genomic DNA of the organism from which the nucleic acid is derived. For example, in various embodiments, the isolated NOVX nucleic acid molecules can contain less than about 5 kb, about 4 kb, about 3 kb, about 2 kb, about 1 kb, about 0.5 kb, or about 0.1 kb, of nucleotide sequences which naturally flank the nucleic acid molecule in genomic DNA of the cell/tissue from which the nucleic acid is derived (e.g., brain, heart, liver, spleen, etc.). Moreover, an "isolated" nucleic acid molecule, such as a cDNA molecule, can be substantially free of other cellular material, or culture medium, or of chemical precursors or other chemicals.
A nucleic acid molecule of the invention, e.g., a nucleic acid molecule having the nucleotide sequence of SEQ ID NOS: 2n-l, wherein n is an integer between 1 and 141, or a complement of this nucleotide sequence, can be isolated using standard molecular biology techniques and the sequence information provided herein. Using all or a portion of the nucleic acid sequence of SEQ ID NOS:2n-l , wherein n is an integer between 1 and 141, as a hybridization probe, NOVX molecules can be isolated using standard hybridization and cloning techniques (e.g., as described in Sambrook, et al., (eds.), MOLECULAR CLONING: A LABORATORY MANUAL 2nd Ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, 1989; and Ausubel, βt al, (eds.), CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, John Wiley & Sons, New York, NY, 1993).
A nucleic acid of the invention can be amplified using cDNA, mRNA or, alternatively, genomic DNA as a template with appropriate oligonucleotide primers according to standard PCR amplification techniques. The nucleic acid so amplified can be cloned into an appropriate vector and characterized by DNA sequence analysis.
Furthermore, oligonucleotides conesponding to NOVX nucleotide sequences can be prepared by standard synthetic techniques, e.g., using an automated DNA synthesizer.
As used herein, the term "oligonucleotide" refers to a series of linked nucleotide residues. A short oligonucleotide sequence may be based on, or designed from, a genomic or cDNA sequence and is used to amplify, confirm, or reveal the presence of an identical, similar or complementary DNA or RNA in a particular cell or tissue. Oligonucleotides comprise a nucleic acid sequence having about 10 nt, 50 nt, or 100 nt in length, preferably about 15 nt to 30 nt in length. In one embodiment of the invention, an oligonucleotide comprising a nucleic acid molecule less than 100 nt in length would further comprise at least 6 contiguous nucleotides of SEQ ID NOS:2n-l , wherein n is an integer between 1 and 141 , or a complement thereof. Oligonucleotides may be chemically synthesized and may also be used as probes. In another embodiment, an isolated nucleic acid molecule of the invention comprises a nucleic acid molecule that is a complement of the nucleotide sequence shown in SEQ ID NOS:2n-l, wherein n is an integer between 1 and 141, or a portion of this nucleotide sequence (e.g., a fragment that can be used as a probe or primer or a fragment encoding a biologically-active portion of a NOVX polypeptide). A nucleic acid molecule that is complementary to the nucleotide sequence of SEQ ID NOS:2n-l, wherein n is an integer between 1 and 141, is one that is sufficiently complementary to the nucleotide sequence of SEQ ID NOS:2n-l, wherein n is an integer between 1 and 141, that it can hydrogen bond with few or no mismatches to a nucleotide sequence of SEQ ID NOS:2n-l, wherein n is an integer between 1 and 141 , thereby forming a stable duplex.
As used herein, the term "complementary" refers to Watson-Crick or Hoogsteen base pairing between nucleotides units of a nucleic acid molecule, and the term "binding" means the physical or chemical interaction between two polypeptides or compounds or associated polypeptides or compounds or combinations thereof. Binding includes ionic, non-ionic, van der Waals, hydrophobic interactions, and the like. A physical interaction can be either direct or indirect. Indirect interactions may be through or due to the effects of another polypeptide or compound. Direct binding refers to interactions that do not take place through, or due to, the effect of another polypeptide or compound, but instead are without other substantial chemical intermediates. A "fragment" provided herein is defined as a sequence of at least 6 (contiguous) nucleic acids or at least 4 (contiguous) amino acids, a length sufficient to allow for specific hybridization in the case of nucleic acids or for specific recognition of an epitope in the case of amino acids, and is at most some portion less than a full length sequence. Fragments may be derived from any contiguous portion of a nucleic acid or amino acid sequence of choice.
A full-length NOVX clone is identified as containing an ATG translation start codon and an in-frame stop codon. Any disclosed NOVX nucleotide sequence lacking an ATG start codon therefore encodes a truncated C-terminal fragment of the respective NOVX polypeptide, and requires that the corresponding full-length cDNA extend in the 5' direction of the disclosed sequence. Any disclosed NOVX nucleotide sequence lacking an in-frame stop codon similarly encodes a truncated N-terminal fragment of the respective NOVX polypeptide, and requires that the conesponding full-length cDNA extend in the 3' direction of the disclosed sequence. A "derivative" is a nucleic acid sequence or amino acid sequence formed from the native compounds either directly, by modification or partial substitution. An "analog" is a nucleic acid sequence or amino acid sequence that has a structure similar to, but not identical to, the native compound, e.g. they differs from it in respect to certain components or side chains. Analogs may be synthetic or derived from a different evolutionary origin and may have a similar or opposite metabolic activity compared to wild type. A "homolog" is a nucleic acid sequence or amino acid sequence of a particular gene that is derived from different species.
Derivatives and analogs may be full length or other than full length. Derivatives or analogs of the nucleic acids or proteins of the invention include, but are not limited to, molecules comprising regions that are substantially homologous to the nucleic acids or proteins of the invention, in various embodiments, by at least about 70%, 80%, or 95% identity (with a preferred identity of 80-95%) over a nucleic acid or amino acid sequence of identical size or when compared to an aligned sequence in which the alignment is done by a computer homology program known in the art, or whose encoding nucleic acid is capable of hybridizing to the complement of a sequence encoding the proteins under stringent, moderately stringent, or low stringent conditions. See e.g. Ausubel, et al., CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, John Wiley & Sons, New York, NY, 1993, and below. A "homologous nucleic acid sequence" or "homologous amino acid sequence," or variations thereof, refer to sequences characterized by a homology at the nucleotide level or amino acid level as discussed above. Homologous nucleotide sequences include those sequences coding for isoforms of NOVX polypeptides. Isoforms can be expressed in different tissues of the same organism as a result of, for example, alternative splicing of RNA. Alternatively, isoforms can be encoded by different genes. In the invention, homologous nucleotide sequences include nucleotide sequences encoding for a NOVX polypeptide of species other than humans, including, but not limited to: vertebrates, and thus can include, e.g., frog, mouse, rat, rabbit, dog, cat cow, horse, and other organisms. Homologous nucleotide sequences also include, but are not limited to, naturally occurring allelic variations and mutations of the nucleotide sequences set forth herein. A homologous nucleotide sequence does not, however, include the exact nucleotide sequence encoding human NOVX protein. Homologous nucleic acid sequences include those nucleic acid sequences that encode conservative amino acid substitutions (see below) in SEQ ID NO:2«-l, wherein n is an integer between 1 and 141, as well as a polypeptide possessing NOVX biological activity. Various biological activities of the NOVX proteins are described below.
A NOVX polypeptide is encoded by the open reading frame ("ORF") of a NOVX nucleic acid. An ORF corresponds to a nucleotide sequence that could potentially be translated into a polypeptide. A stretch of nucleic acids comprising an ORF is uninterrupted by a stop codon. An ORF that represents the coding sequence for a full protein begins with an ATG "start" codon and terminates with one of the three "stop" codons, namely, TAA, TAG, or TGA. For the purposes of this invention, an ORF may be any part of a coding sequence, with or without a start codon, a stop codon, or both. For an ORF to be considered as a good candidate for coding for a honafide cellular protein, a minimum size requirement is often set, e.g., a stretch of DNA that would encode a protem of 50 amino acids or more.
The nucleotide sequences determined from the cloning of the human NOVX genes allows for the generation of probes and primers designed for use in identifying and or cloning NOVX homologues in other cell types, e.g. from other tissues, as well as NOVX homologues from other vertebrates. The probe/primer typically comprises substantially purified oligonucleotide. The oligonucleotide typically comprises a region of nucleotide sequence that hybridizes under stringent conditions to at least about 12, 25, 50, 100, 150, 200, 250, 300, 350 or 400 consecutive sense strand nucleotide sequence of SEQ ID
NO :2n- 1 , wherein n is an integer between 1 and 141 ; or an anti-sense strand nucleotide sequence of SEQ ID NO:2n-l, wherein n is an integer between 1 and 141; or ofa naturally occurring mutant of SEQ ID NO:2n-l, wherein n is an integer between 1 and 141. Probes based on the human NOVX nucleotide sequences can be used to detect transcripts or genomic sequences encoding the same or homologous proteins. In various embodiments, the probe has a detectable label attached, e.g. the label can be a radioisotope, a fluorescent compound, an enzyme, or an enzyme co-factor. Such probes can be used as a part of a diagnostic test kit for identifying cells or tissues which mis-express a NOVX protein, such as by measuring a level of a NOVX-encoding nucleic acid in a sample of cells from a subject e.g., detecting NOVX mRNA levels or determining whether a genomic NOVX gene has been mutated or deleted. "A polypeptide having a biologically-active portion of a NOVX polypeptide" refers to polypeptides exhibiting activity similar, but not necessarily identical to, an activity of a polypeptide of the invention, including mature forms, as measured in a particular biological assay, with or without dose dependency. A nucleic acid fragment encoding a "biologically-active portion of NOVX" can be prepared by isolating a portion of SEQ ID NO:2«-l, wherein n is an integer between 1 and 141, that encodes a polypeptide having a NOVX biological activity (the biological activities of the NOVX proteins are described below), expressing the encoded portion of NOVX protein (e.g., by recombinant expression in vitro) and assessing the activity of the encoded portion of NOVX.
NOVX Nucleic Acid and Polypeptide Variants
The invention further encompasses nucleic acid molecules that differ from the nucleotide sequences of SEQ ID NO:2«-l, wherein n is an integer between 1 and 141 , due to degeneracy of the genetic code and thus encode the same NOVX proteins as that encoded by the nucleotide sequences of SEQ ID NO:2«-l , wherein n is an integer between 1 and 141. In another embodiment, an isolated nucleic acid molecule of the . invention has a nucleotide sequence encoding a protein having an amino acid sequence of SEQ ID NO:27z, wherein n is an integer between 1 and 141.
In addition to the human NOVX nucleotide sequences of SEQ ID NO:2«-l, wherein n is an integer between 1 and 141 , it will be appreciated by those skilled in the art that DNA sequence polymorphisms that lead to changes in the amino acid sequences of the NOVX polypeptides may exist within a population (e.g., the human population). Such genetic polymorphism in the NOVX genes may exist among individuals within a population due to natural allelic variation. As used herein, the terms "gene" and "recombinant gene" refer to nucleic acid molecules comprising an open reading frame (ORF) encoding a NOVX protein, preferably a vertebrate NOVX protein. Such natural allelic variations can typically result in 1-5% variance in the nucleotide sequence of the NOVX genes. Any and all such nucleotide variations and resulting amino acid polymorphisms in the NOVX polypeptides, which are the result of natural allelic variation and that do not alter the functional activity of the NOVX polypeptides, are intended to be within the scope of the invention. Moreover, nucleic acid molecules encoding NOVX proteins from other species, and thus that have a nucleotide sequence that differs from a human SEQ ID NO:2«-l, wherein n is an integer between 1 and 141, are intended to be within the scope of the invention. Nucleic acid molecules corresponding to natural allelic variants and homologues of the NOVX cDNAs of the invention can be isolated based on their homology to the human NOVX nucleic acids disclosed herein using the human cDNAs, or a portion thereof, as a hybridization probe according to standard hybridization techniques under stringent hybridization conditions.
Accordingly, in another embodiment, an isolated nucleic acid molecule of the invention is at least 6 nucleotides in length and hybridizes under stringent conditions to the nucleic acid molecule comprising the nucleotide sequence of SEQ ID NO:2?z-l, wherein n is an integer between 1 and 141. In another embodiment, the nucleic acid is at least 10, 25, 50, 100, 250, 500, 750, 1000, 1500, or 2000 or more nucleotides in length. In yet another embodiment, an isolated nucleic acid molecule of the invention hybridizes to the coding region. As used herein, the term "hybridizes under stringent conditions" is intended to describe conditions for hybridization and washing under which nucleotide sequences at least about 65% homologous to each other typically remain hybridized to each other.
Homologs (i.e., nucleic acids encoding NOVX proteins derived from species other than human) or other related sequences (e.g., paralogs) can be obtained by low, moderate or high stringency hybridization with all or a portion of the particular human sequence as a probe using methods well known in the art for nucleic acid hybridization and cloning.
As used herein, the phrase "stringent hybridization conditions" refers to conditions under which a probe, primer or oligonucleotide will hybridize to its target sequence, but to no other sequences. Stringent conditions are sequence-dependent and will be different in different circumstances. Longer sequences hybridize specifically at higher temperatures than shorter sequences. Generally, stringent conditions are selected to be about 5 °C lower than the thermal melting point (Tm) for the specific sequence at a defined ionic strength and pH. The Tm is the temperature (under defined ionic strength, pH and nucleic acid concentration) at which 50% of the probes complementary to the target sequence hybridize to the target sequence at equilibrium. Since the target sequences are generally present at excess, at Tm, 50% of the probes are occupied at equilibrium. Typically, stringent conditions will be those in which the salt concentration is less than about 1.0 M sodium ion, typically about 0.01 to 1.0 M sodium ion (or other salts) at pH 7.0 to 8.3 and the temperature is at least about 30 °C for short probes, primers or oligonucleotides (e.g., 10 nt to 50 nt) and at least about 60 °C for longer probes, primers and oligonucleotides. Stringent conditions may also be achieved with the addition of destabilizing agents, such as formamide.
Stringent conditions are known to those skilled in the art and can be found in Ausubel, et al., (eds.), CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, John Wiley & Sons, N.Y. (1989), 6.3.1-6.3.6. Preferably, the conditions are such that sequences at least about 65%, 70%, 75%, 85%, 90%, 95%, 98%, or 99% homologous to each other typically remain hybridized to each other. A non-limiting example of stringent hybridization conditions are hybridization in a high salt buffer comprising 6X SSC, 50 mM Tris-HCl (pH 7.5), 1 mM EDTA, 0.02% PVP, 0.02% Ficoll, 0.02% BSA, and 500 mg/ml denatured salmon sperm DNA at 65°C, followed by one or more washes in 0.2X SSC, 0.01%o BSA at 50°C. An isolated nucleic acid molecule of the invention that hybridizes under stringent conditions to a sequence of SEQ ID NO:2n-l , wherein n is an integer between 1 and 141, corcesponds to a naturally-occurring nucleic acid molecule. As used herein, a "naturally-occurring" nucleic acid molecule refers to an RNA or DNA molecule having a nucleotide sequence that occurs in nature (e.g., encodes a natural protein). In a second embodiment, a nucleic acid sequence that is hybridizable to the nucleic acid molecule comprising the nucleotide sequence of SEQ ID NO:2«-l , wherein n is an integer between 1 and 141, or fragments, analogs or derivatives thereof, under conditions of moderate stringency is provided. A non-limiting example of moderate stringency hybridization conditions are hybridization in 6X SSC, 5X Reinhardt's solution, 0.5% SDS and 100 mg/ml denatured salmon sperm DNA at 55 °C, followed by one or more washes in IX SSC, 0.1% SDS at 37 °C. Other conditions of moderate stringency that may be used are well-known within the art. See, e.g., Ausubel, et al. (eds.), 1993, CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, John Wiley & Sons, NY, and Krieger, 1990; GENE TRANSFER AND EXPRESSION, A LABORATORY MANUAL, Stockton Press, NY. In a third embodiment, a nucleic acid that is hybridizable to the nucleic acid molecule comprising the nucleotide sequences of SEQ ID NO:2w-l, wherein n is an integer between 1 and 141, or fragments, analogs or derivatives thereof, under conditions of low stringency, is provided. A non-limiting example of low stringency hybridization conditions are hybridization in 35% formamide, 5X SSC, 50 mM Tris-HCl (pH 7.5), 5 mM EDTA, 0.02% PVP, 0.02% Ficoll, 0.2% BSA, 100 mg/ml denatured salmon sperm DNA, 10% (wt/vol) dextran sulfate at 40°C, followed by one or more washes in 2X SSC, 25 mM Tris-HCl (pH 7.4), 5 M EDTA, and 0.1% SDS at 50 °C. Other conditions of low stringency that may be used are well known in the art (e.g., as employed for cross-species hybridizations). See, e.g., Ausubel, et al. (eds.), 1993, CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, John Wiley & Sons, NY, and Kriegler, 1990, GENE TRANSFER AND EXPRESSION, A LABORATORY MANUAL, Stockton Press, NY; Shilo and Weinberg, 1981. Proc Natl Acad Sci USA 78: 6789-6792. Conservative Mutations
In addition to naturally-occurring allelic variants of NOVX sequences that may exist in the population, the skilled artisan will further appreciate that changes can be introduced by mutation into the nucleotide sequences of SEQ ID NO:2>J-1, wherein n is an integer between l and 141, thereby leading to changes in the amino acid sequences of the encoded NOVX protein, without altering the functional ability of that NOVX protein. For example, nucleotide substitutions leading to amino acid substitutions at "non-essential" amino acid residues can be made in the sequence of SEQ ID NO:2w, wherein n is an integer between 1 and 141. A "non-essential" amino acid residue is a residue that can be altered from the wild-type sequences of the NOVX proteins without altering their biological activity, whereas an "essential" amino acid residue is required for such biological activity. For example, amino acid residues that are conserved among the NOVX proteins of the invention are predicted to be particularly non-amenable to alteration. Amino acids for which conservative substitutions can be made are well-known within the art. Another aspect of the invention pertains to nucleic acid molecules encoding
NOVX proteins that contain changes in amino acid residues that are not essential for activity. Such NOVX proteins differ in amino acid sequence from SEQ ID NO:2«-l, wherein n is an integer between 1 and 141, yet retain biological activity. In one embodiment, the isolated nucleic acid molecule comprises a nucleotide sequence encoding a protein, wherein the protem comprises an amino acid sequence at least about 40% homologous to the amino acid sequences of SEQ ID NO:2/2, wherein n is an integer between 1 and 141. Preferably, the protein encoded by the nucleic acid molecule is at least about 60% homologous to SEQ ID NO:2«, wherein n is an integer between 1 and 141; more preferably at least about 70% homologous to SEQ ID NO:2n, wherein n is an integer between 1 and 141; still more preferably at least about 80% homologous to SEQ ID NO:2«, wherein n is an integer between 1 and 141; even more preferably at least about 90% homologous to SEQ ID NO:2n, wherein n is an integer between 1 and 141 ; and most preferably at least about 95% homologous to SEQ ID NO:2«, wherein n is an integer between 1 and 141.
An isolated nucleic acid molecule encoding a NOVX protein homologous to the protein of SEQ ID NO:2«, wherein n is an integer between 1 and 141, can be created by introducing one or more nucleotide substitutions, additions or deletions into the nucleotide sequence of SEQ ID NO:2«-l, wherein n is an integer between 1 and 141, such that one or more amino acid substitutions, additions or deletions are introduced into the encoded protein.
Mutations can be introduced any one of SEQ ID NO:2/z-l, wherein n is an integer between 1 and 141, by standard techniques, such as site-directed mutagenesis and
PCR-mediated mutagenesis. Preferably, conservative amino acid substitutions are made at one or more predicted, non-essential amino acid residues. A "conservative amino acid substitution" is one in which the amino acid residue is replaced with an amino acid residue having a similar side chain. Families of amino acid residues having similar side chains have been defined within the art. These families include amino acids with basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine), nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), beta-branched side chains (e-g; threonine, valine, isoleucine) and aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan, histidine). Thus, a predicted non-essential amino acid residue in the NOVX protein is replaced with another amino acid residue from the same side chain family. Alternatively, in another embodiment, mutations can be introduced randomly along all or part of a NOVX coding sequence, such as by saturation mutagenesis, and the resultant mutants can be screened for NOVX biological activity to identify mutants that retain activity. Following mutagenesis of a nucleic acid of SEQ ID NO:2«-l, wherein n is an integer between 1 and 141, the encoded protein can be expressed by any recombinant technology known in the art and the activity of the protein can be determined.
The relatedness of amino acid families may also be determined based on side chain interactions. Substituted amino acids may be fully conserved "strong" residues or fully conserved "weak" residues. The "strong" group of conserved amino acid residues may be any one of the following groups: STA, NEQK, NHQK, NDEQ, QHRK, MILV, MILF, HY, FYW, wherein the single letter amino acid codes are grouped by those amino acids that may be substituted for each other. Likewise, the "weak" group of conserved residues may be any one of the following: CSA, ATV, SAG, STNK, STPA, SGND, SNDEQK, NDEQHK, NEQHRK, HFY, wherein the letters within each group represent the single letter amino acid code.
In one embodiment, a mutant NOVX protein can be assayed for (i) the ability to form proteimprotein interactions with other NOVX proteins, other cell-surface proteins, or biologically-active portions thereof, (ii) complex formation between a mutant NOVX protein and a NOVX ligand; or (iii) the ability of a mutant NOVX protein to bind to an intracellular target protein or biologically-active portion thereof; (e.g. avidin proteins).
In yet another embodiment, a mutant NOVX protein can be assayed for the ability to regulate a specific biological function (e.g., regulation of insulin release).
Interfering RNA In one aspect of the invention, NOVX gene expression can be attenuated by RNA interference. One approach well-known in the art is short interfering RNA (siRNA) mediated gene silencing where expression products of a NOVX gene are targeted by specific double stranded NOVX derived siRNA nucleotide sequences that are complementary to at least a 19-25 nt long segment of the NOVX gene transcript, including the 5' untranslated (UT) region, the ORF, or the 3' UT region. See, e.g., PCT applications WO00/44895, WO99/32619, WO01/75164, WO01/92513, WO01/29058, WO01/89304, WO02/16620, and WO02/29858, each incorporated by reference herein in their entirety. Targeted genes can be a NOVX gene, or an upstream or downstream modulator of the NOVX gene. Nonlimiting examples of upstream or downstream modulators of a NOVX gene include, e.g., a transcription factor that binds the NOVX gene promoter, a kinase or phosphatase that interacts with a NOVX polypeptide, and polypeptides involved in a NOVX regulatory pathway. According to the methods of the present invention, NOVX gene expression is silenced using short interfering RNA. A NOVX polynucleotide according to the invention includes a siRNA polynucleotide. Such a NOVX siRNA can be obtained using a NOVX polynucleotide sequence, for example, by processing the NOVX ribopolynucleotide sequence in a cell-free system, such as but not limited to a Drosophila extract, or by transcription of recombinant double stranded NOVX RNA or by chemical synthesis of nucleotide sequences homologous to a NOVX sequence. See, e.g., Tuschl, Zamore, Lehmann, Bartel and Shaφ (1999), Genes & Dev. 13: 3191-3197, incorporated herein by reference in its entirety. When synthesized, a typical 0.2 micromolar-scale RNA synthesis provides about 1 milligram of siRNA, which is sufficient for 1000 transfection experiments using a 24-well tissue culture plate format.
The most efficient silencing is generally observed with siRNA duplexes composed of a 21-nt sense strand and a 21-nt antisense strand, paired in a manner to have a 2-nt 3' overhang. The sequence of the 2-nt 3' overhang makes an additional small contribution to the specificity of siRNA target recognition. The contribution to specificity is localized to the unpaired nucleotide adjacent to the first paired bases. In one embodiment, the nucleotides in the 3' overhang are ribonucleotides. In an alternative embodiment, the nucleotides in the 3' overhang are deoxyribonucleotides. Using 2'-deoxyribonucleotides in the 3' overhangs is as efficient as using ribonucleotides, but deoxyribonucleotides are often cheaper to synthesize and are most likely more nuclease resistant.
A contemplated recombinant expression vector of the invention comprises a NOVX DNA molecule cloned into an expression vector comprising operatively-linked regulatory sequences flanking the NOVX sequence in a manner that allows for expression (by transcription of the DNA molecule) of both strands. An RNA molecule that is antisense to NOVX mRNA is transcribed by a first promoter (e.g., a promoter sequence 3 ' of the cloned DNA) and an RNA molecule that is the sense strand for the NOVX mRNA is transcribed by a second promoter (e.g., a promoter sequence 5' of the cloned DNA). The sense and antisense strands may hybridize in vivo to generate siRNA constructs for silencing of the NOVX gene. Alternatively, two constructs can be utilized to create the sense and anti-sense strands of a siRNA construct. Finally, cloned DNA can encode a construct having secondary structure, wherein a single transcript has both the sense and complementary antisense sequences from the target gene or genes. In an example of this embodiment, a hairpin RNAi product is homologous to all or a portion of the target gene. In another example, a hairpin RNAi product is a siRNA. The regulatory sequences flanking the NOVX sequence may be identical or may be different, such that their expression may be modulated independently, or in a temporal or spatial manner.
In a specific embodiment, siRNAs are transcribed intracellularly by cloning the NOVX gene templates into a vector containing, e.g., a RNA pol III transcription unit from the smaller nuclear RNA (snRNA) U6 or the human RNase P RNA HI . One example of a vector system is the GeneSuppressor™ RNA Interference kit (commercially available from Imgenex). The U6 and HI promoters are members of the type III class of Pol III promoters. The +1 nucleotide of the U6-like promoters is always guanosine, whereas the +1 for HI promoters is adenosine. The termination signal for these promoters is defined by five consecutive thymidines. The transcript is typically cleaved after the second uridine. Cleavage at this position generates a 3' UU overhang in the expressed siRNA, which is similar to the 3' overhangs of synthetic siRNAs. Any sequence less than 400 nucleotides in length can be transcribed by these promoter, therefore they are ideally suited for the expression of around 21 -nucleotide siRNAs in, e.g., an approximately 50-nucleotide RNA stem-loop transcript.
A siRNA vector appears to have an advantage over synthetic siRNAs where long term knock-down of expression is desired. Cells transfected with a siRNA expression vector would experience steady, long-term mRNA inhibition. In contrast, cells transfected with exogenous synthetic siRNAs typically recover from mRNA suppression within seven days or ten rounds of cell division. The long-term gene silencing ability of siRNA expression vectors may provide for applications in gene therapy.
In general, siRNAs are chopped from longer dsRNA by an ATP-dependent ribonuclease called DICER. DICER is a member of the RNase III family of double-stranded RNA-specific endonucleases. The siRNAs assemble with cellular proteins into an endonuclease complex. In vitro studies in Drosophila suggest that the siRNAs/protein complex (siRNP) is then transferred to a second enzyme complex, called an RNA-induced silencing complex (RISC), which contains an endoribonuclease that is distinct from DICER. RISC uses the sequence encoded by the antisense siRNA strand to find and destroy mRNAs of complementary sequence. The siRNA thus acts as a guide, restricting the ribonuclease to cleave only mRNAs complementary to one of the two siRNA strands. A NOVX mRNA region to be targeted by siRNA is generally selected from a desired NOVX sequence beginning 50 to 100 nt downstream of the start codon. Alternatively, 5' or 3' UTRs and regions nearby the start codon can be used but are generally avoided, as these may be richer in regulatory protein binding sites. UTR-binding proteins and/or translation initiation complexes may interfere with binding of the siRNP or RISC endonuclease complex. An initial BLAST homology search for the selected siRNA sequence is done against an available nucleotide sequence library to ensure that only one gene is targeted. Specificity of target recognition by siRNA duplexes indicate that a single point mutation located in the paired region of an siRNA duplex is sufficient to abolish target mRNA degradation. See, Elbashir et al. 2001 EMBO J. 20(23):6877-88. Hence, consideration should be taken to accommodate SNPs, polymorphisms, allelic variants or species-specific variations when targeting a desired gene.
In one embodiment, a complete NOVX siRNA experiment includes the proper negative control. A negative control siRNA generally has the same nucleotide composition as the NOVX siRNA but lack significant sequence homology to the genome. Typically, one would scramble the nucleotide sequence of the NOVX siRNA and do a homology search to make sure it lacks homology to any other gene.
Two independent NOVX siRNA duplexes can be used to knock-down a target NOVX gene. This helps to control for specificity of the silencing effect. In addition, expression of two independent genes can be simultaneously knocked down by using equal concentrations of different NOVX siRNA duplexes, e.g., a NOVX siRNA and an siRNA for a regulator of a NOVX gene or polypeptide. Availability of siRNA-associating proteins is believed to be more limiting than target mRNA accessibility.
A targeted NOVX region is typically a sequence of two adenines (AA) and two thymidines (TT) divided by a spacer region of nineteen (NI 9) residues (e.g.,
AA(N19)TT). A desirable spacer region has a G/C-content of approximately 30% to 70%, and more preferably of about 50%. If the sequence AA(N19)TT is not present in the target sequence, an alternative target region would be AA(N21). The sequence of the NOVX sense siRNA conesponds to (N19)TT or N21, respectively. In the latter case, conversion of the 3' end of the sense siRNA to TT can be performed if such a sequence does not naturally occur in the NOVX polynucleotide. The rationale for this sequence conversion is to generate a symmetric duplex with respect to the sequence composition of the sense and antisense 3' overhangs. Symmetric 3' overhangs may help to ensure that the siRNPs are formed with approximately equal ratios of sense and antisense target RNA-cleaving siRNPs. See, e.g., Elbashir, Lendeckel and Tuschl (2001). Genes & Dev. 15: 188-200, incorporated by reference herein in its entirely. The modification of the overhang of the sense sequence of the siRNA duplex is not expected to affect targeted mRNA recognition, as the antisense siRNA strand guides target recognition.
Alternatively, if the NOVX target mRNA does not contain a suitable AA(N21) sequence, one may search for the sequence NA(N21). Further, the sequence of the sense strand and antisense strand may still be synthesized as 5' (N19)TT, as it is believed that the sequence of the 3'-most nucleotide of the antisense siRNA does not contribute to specificity. Unlike antisense or ribozyme technology, the secondary structure of the target mRNA does not appear to have a strong effect on silencing. See, Harborth, et al. (2001) J. Cell Science 114: 4557-4565, incorporated by reference in its entirety.
Transfection of NOVX siRNA duplexes can be achieved using standard nucleic acid transfection methods, for example, OLIGOFECTAMINE Reagent (commercially available from Invitrogen). An assay for NOVX gene silencing is generally performed approximately 2 days after transfection. No NOVX gene silencing has been observed in the absence of transfection reagent, allowing for a comparative analysis of the wild-type and silenced NOVX phenotypes. In a specific embodiment, for one well of a 24-well plate, approximately 0.84 μg of the siRNA duplex is generally sufficient. Cells are typically seeded the previous day, and are transfected at about 50% confluence. The choice of cell culture media and conditions are routine to those of skill in the art, and will vary with the choice of cell type. The efficiency of transfection may depend on the cell type, but also on the passage number and the confluency of the cells. The time and the manner of formation of siRNA-liposome complexes (e.g. inversion versus vortexing) are also critical. Low transfection efficiencies are the most frequent cause of unsuccessful NOVX silencing. The efficiency of transfection needs to be carefully examined for each new cell line to be used. Prefened cell are derived from a mammal, more preferably from a rodent such as a rat or mouse, and most preferably from a human. Where used for therapeutic treatment, the cells are preferentially autologous, although non-autologous cell sources are also contemplated as within the scope of the present invention.
For a control experiment, transfection of 0.84 μg single-stranded sense NOVX siRNA will have no effect on NOVX silencing, and 0.84 μg antisense siRNA has a weak silencing effect when compared to 0.84 μg of duplex siRNAs. Control experiments again allow for a comparative analysis of the wild-type and silenced NOVX phenotypes. To control for transfection efficiency, targeting of common proteins is typically performed, for example targeting of lamin A/C or transfection of a CMV-driven EGFP-expression plasmid (e.g. commercially available from Clontech). In the above example, a determination of the fraction of lamin A C knockdown in cells is determined the next day by such techniques as immunofluorescence, Western blot, Northern blot or other similar assays for protein expression or gene expression. Lamin A/C monoclonal antibodies may be obtained from Santa Cruz Biotechnology.
Depending on the abundance and the half life (or turnover) of the targeted NOVX polynucleotide in a cell, a knock-down phenotype may become apparent after 1 to 3 days, or even later. In cases where no NOVX knock-down phenotype is observed, depletion of the NOVX polynucleotide may be observed by immunofluorescence or Western blotting. If the NOVX polynucleotide is still abundant after 3 days, cells need to be split and transferred to a fresh 24-well plate for re-transfection. If no knock-down of the targeted protein is observed, it may be desirable to analyze whether the target mRNA (NOVX or a NOVX upstream or downstream gene) was effectively destroyed by the transfected siRNA duplex. Two days after transfection, total RNA is prepared, reverse transcribed using a target-specific primer, and PCR-amplified with a primer pair covering at least one exon-exon junction in order to control for amplification of pre-mRNAs. RT/PCR of a non-targeted mRNA is also needed as control. Effective depletion of the mRNA yet undetectable reduction of target protein may indicate that a large reservoir of stable NOVX protein may exist in the cell. Multiple transfection in sufficiently long intervals may be necessary until the target protein is finally depleted to a point where a phenotype may become apparent. If multiple transfection steps are required, cells are split 2 to 3 days after transfection. The cells may be transfected immediately after splitting.
An inventive therapeutic method of the invention contemplates administering a NOVX siRNA construct as therapy to compensate for increased or aberrant NOVX expression or activity. The NOVX ribopolynucleotide is obtained and processed into siRNA fragments, or a NOVX siRNA is synthesized, as described above. The NOVX siRNA is administered to cells or tissues using known nucleic acid transfection techniques, as described above. A NOVX siRNA specific for a NOVX gene will decrease or knockdown NOVX transcription products, which will lead to reduced NOVX polypeptide production, resulting in reduced NOVX polypeptide activity in the cells or tissues. The present invention also encompasses a method of treating a disease or condition associated with the presence of a NOVX protein in an individual comprising administering to the individual an RNAi construct that targets the mRNA of the protein (the mRNA that encodes the protein) for degradation. A specific RNAi construct includes a siRNA or a double stranded gene transcript that is processed into siRNAs. Upon treatment, the target protein is not produced or is not produced to the extent it would be in the absence of the treatment.
Where the NOVX gene function is not correlated with a known phenotype, a control sample of cells or tissues from healthy individuals provides a reference standard for determining NOVX expression levels. Expression levels are detected using the assays described, e.g., RT-PCR, Northern blotting, Western blotting, ELISA, and the like. A subject sample of cells or tissues is taken from a mammal, preferably a human subject, suffering from a disease state. The NOVX ribopolynucleotide is used to produce siRNA constructs, that are specific for the NOVX gene product. These cells or tissues are treated by administering NOVX siRNA' s to the cells or tissues by methods described for the transfection of nucleic acids into a cell or tissue, and a change in NOVX polypeptide or polynucleotide expression is observed in the subject sample relative to the control sample, using the assays described. This NOVX gene knockdown approach provides a rapid method for determination of a NOVX minus (NOVX") phenotype in the treated subject sample. The NOVX" phenotype observed in the treated subject sample thus serves as a marker for monitoring the course of a disease state during treatment.
In specific embodiments, a NOVX siRNA is used in therapy. Methods for the generation and use of a NOVX siRNA are known to those skilled in the art. Example techniques are provided below. Production of RNAs
Sense RNA (ssRNA) and antisense RNA (asRNA) of NOVX are produced using known methods such as transcription in RNA expression vectors. In the initial experiments, the sense and antisense RNA are about 500 bases in length each. The produced ssRNA and asRNA (0.5 μM) in 10 mM Tris-HCl (pH 7.5) with 20 mM NaCl were heated to 95° C for 1 min then cooled and annealed at room temperature for 12 to 16 h. The RNAs are precipitated and resuspended in lysis buffer (below). To monitor annealing, RNAs are electrophoresed in a 2% agarose gel in TBE buffer and stained with ethidium bromide. See, e.g., Sambrook et al., Molecular Cloning. Cold Spring Harbor Laboratory Press, Plainview, N.Y. (1989).
Lysate Preparation
Untreated rabbit reticulocyte lysate (Ambion) are assembled according to the manufacturer's directions. dsRNA is incubated in the lysate at 30° C for 10 min prior to the addition of mRNAs. Then NOVX mRNAs are added and the incubation continued for an additional 60 min. The molar ratio of double stranded RNA and mRNA is about 200: 1. The NOVX mRNA is radiolabeled (using known techniques) and its stability is monitored by gel electrophoresis. In a parallel experiment made with the same conditions, the double stranded RNA is internally radiolabeled with a 32P-ATP. Reactions are stopped by the addition of 2 X proteinase K buffer and deproteinized as described previously (Tuschl et al., Genes Dev., 13:3191-3197 (1999)). Products are analyzed by electrophoresis in 15% or 18% polyacrylamide sequencing gels using appropriate RNA standards. By monitoring the gels for radioactivity, the natural production of 10 to 25 nt RNAs from the double stranded RNA can be determined.
The band of double stranded RNA, about 21-23 bps, is eluded. The efficacy of these 21-23 mers for suppressing NOVX transcription is assayed in vitro using the same rabbit reticulocyte assay described above using 50 nanomolar of double stranded 21-23 mer for each assay. The sequence of these 21-23 mers is then determined using standard nucleic acid sequencing techniques.
RNA Preparation
21 nt RNAs, based on the sequence determined above, are chemically synthesized using Expedite RNA phosphoramidites and thymidine phosphoramidite (Proligo, Germany). Synthetic oligonucleotides are deprotected and gel-purified (Elbashir, Lendeckel, & Tuschl, Genes & Dev. 15, 188-200 (2001)), followed by Sep-Pak C18 cartridge (Waters, Milford, Mass., USA) purification (Tuschl, et al., Biochemistry, 32:11658-11668 (1993)).
These RNAs (20 μM) single strands are incubated in annealing buffer (100 mM potassium acetate, 30 mM HEPES-KOH at pH 7.4, 2 mM magnesium acetate) for 1 min at 90° C followed by 1 h at 37° C. Cell Culture
A cell culture known in the art to regularly express NOVX is propagated using standard conditions. 24 hours before transfection, at approx. 80% confluency, the cells are trypsinized and diluted 1 :5 with fresh medium without antibiotics (1-3 X 105 cells/ml) and transferred to 24-well plates (500 ml/well). Transfection is performed using a commercially available lipofection kit and NOVX expression is monitored using standard techniques with positive and negative control. A positive control is cells that naturally express NOVX while a negative control is cells that do not express NOVX. Base-paired 21 and 22 nt siRNAs with overhanging 3' ends mediate efficient sequence-specific mRNA degradation in lysates and in cell culture. Different concentrations of siRNAs are used. An efficient concentration for suppression in vitro in mammalian culture is between 25 nM to 100 nM final concentration. This indicates that siRNAs are effective at concentrations that are several orders of magnitude below the concentrations applied in conventional antisense or ribozyme gene targeting experiments. The above method provides a way both for the deduction of NOVX siRNA sequence and the use of such siRNA for in vitro suppression. In vivo suppression may be performed using the same siRNA using well known in vivo transfection or gene therapy transfection techniques.
Antisense Nucleic Acids Another aspect of the invention pertains to isolated antisense nucleic acid molecules that are hybridizable to or complementary to the nucleic acid molecule comprising the nucleotide sequence of SEQ ID NO:2«-l , wherein n is an integer between 1 and 141, or fragments, analogs or derivatives thereof. An "antisense" nucleic acid comprises a nucleotide sequence that is complementary to a "sense" nucleic acid encoding a protein (e.g., complementary to the coding strand of a double-stranded cDNA molecule or complementary to an mRNA sequence). In specific aspects, antisense nucleic acid molecules are provided that comprise a sequence complementary to at least about 10, 25, 50, 100, 250 or 500 nucleotides or an entire NOVX coding strand, or to only a portion thereof. Nucleic acid molecules encoding fragments, homologs, derivatives and analogs of a NOVX protein of SEQ ID NO:2?j, wherein n is an integer between 1 and 141, or antisense nucleic acids complementary to a NOVX nucleic acid sequence of SEQ ID NO:2n-l, wherein n is an integer between 1 and 141, are additionally provided.
In one embodiment, an antisense nucleic acid molecule is antisense to a "coding region" of the coding strand of a nucleotide sequence encoding a NOVX protein. The term "coding region" refers to the region of the nucleotide sequence comprising codons which are translated into amino acid residues. In another embodiment, the antisense nucleic acid molecule is antisense to a "noncoding region" of the coding strand of a nucleotide sequence encoding the NOVX protein. The term "noncoding region" refers to 5' and 3' sequences which flank the coding region that are not translated into amino acids (i. e. , also referred to as 5' and 3' untranslated regions).
Given the coding strand sequences encoding the NOVX protein disclosed herein, antisense nucleic acids of the invention can be designed according to the rules of Watson and Crick or Hoogsteen base pairing. The antisense nucleic acid molecule can be complementary to the entire coding region of NOVX mRNA, but more preferably is an oligonucleotide that is antisense to only a portion of the coding or noncoding region of NOVX mRNA. For example, the antisense oligonucleotide can be complementary to the region smrounding the translation start site of NOVX mRNA. An antisense oligonucleotide can be, for example, about 5, 10, 15, 20, 25, 30, 35, 40, 45 or 50 nucleotides in length. An antisense nucleic acid of the invention can be constructed using chemical synthesis or enzymatic ligation reactions using procedures known in the art. For example, an antisense nucleic acid (e.g., an antisense oligonucleotide) can be chemically synthesized using naturally-occurring nucleotides or variously modified nucleotides designed to increase the biological stability of the molecules or to increase the physical stability of the duplex formed between the antisense and sense nucleic acids (e.g., phosphorothioate derivatives and acridine substituted nucleotides can be used). Examples of modified nucleotides that can be used to generate the antisense nucleic acid include: 5-fluorouracil, 5-bromouracil, 5-chlorouracil, 5-iodouracil, hypoxanthine, xanthine, 4-acetylcytosine, 5-carboxymethylaminomethyl-2-thiouridine, 5-(carboxyhydroxylmethyl) uracil, 5-carboxymethylaminomethyluracil, dihydrouracil, beta-D-galactosylqueosine, inosine, N6-isopentenyladenine, 1-methylguanine, 1-methylinosine, 2,2-dimethylguanine, 2-methyladenine, 2-methylguanine, 5-methoxyuracil, 3-methylcytosine, 5-methylcytosine, N6-adenine, 7-methylguanine, 5-methylaminomethyluracil, 5-methoxyaminomethyl-2-thiouracil, 2-thiouracil, 4-thiouracil, beta-D-mannosylqueosine, 5'-methoxycarboxymethyluracil, 2-methylthio-N6-isopentenyladenine, uracil-5-oxyacetic acid (v), wybutoxosine, pseudouracil, queosine, 2-thiocytosine, 5-methyl-2-thiouracil, 5-methyluracil, uracil-5-oxyacetic acid methylester, uracil-5-oxyacetic acid (v), 5-methyl-2-thiouracil, 3-(3-amino-3-N-2-carboxypropyl) uracil, (acp3)w, and 2,6-diaminopurine. Alternatively, the antisense nucleic acid can be produced biologically using an expression vector into which a nucleic acid has been subcloned in an antisense orientation (i.e., RNA transcribed from the inserted nucleic acid will be of an antisense orientation to a target nucleic acid of interest, described further in the following subsection). The antisense nucleic acid molecules of the invention are typically administered to a subject or generated in situ such that they hybridize with or bind to cellular mRNA and/or genomic DNA encoding a NOVX protein to thereby inhibit expression of the protein (e.g., by inhibiting transcription and/or translation). The hybridization can be by conventional nucleotide complementarity to form a stable duplex, or, for example, in the case of an antisense nucleic acid molecule that binds to DNA duplexes, through specific interactions in the major groove of the double helix. An example of a route of administration of antisense nucleic acid molecules of the invention includes direct injection at a tissue site. Alternatively, antisense nucleic acid molecules can be modified to target selected cells and then administered systemically. For example, for systemic administration, antisense molecules can be modified such that they specifically bind to receptors or antigens expressed on a selected cell surface (e.g., by linking the antisense nucleic acid molecules to peptides or antibodies that bind to cell surface receptors or antigens). The antisense nucleic acid molecules can also be delivered to cells using the vectors described herein. To achieve sufficient nucleic acid molecules, vector constructs in which the antisense nucleic acid molecule is placed under the control of a strong pol II or pol III promoter are preferred.
In yet another embodiment, the antisense nucleic acid molecule of the invention is an α-anomeric nucleic acid molecule. An α-anomeric nucleic acid molecule forms specific double-stranded hybrids with complementary RNA in which, contrary to the usual β-units, the strands run parallel to each other. See, e.g., Gaultier, et al, 1987. Nucl. Acids Res. 15: 6625-6641. The antisense nucleic acid molecule can also comprise a 2'-o-methylribonucleotide (See, e.g., Inoue, et al. 1987. Nucl. Acids Res. 15: 6131-6148) or a chimeric RNA-DNA analogue (See, e.g, Inoue, et al, 1987. FEBS Lett. 215: 327-330.
Ribozymes and PNA Moieties
Nucleic acid modifications include, by way of non-limiting example, modified bases, and nucleic acids whose sugar phosphate backbones are modified or derivatized. These modifications are carried out at least in part to enhance the chemical stability of the modified nucleic acid, such that they may be used, for example, as antisense binding nucleic acids in therapeutic applications in a subject.
In one embodiment, an antisense nucleic acid of the invention is a ribozyme. Ribozymes are catalytic RNA molecules with ribonuclease activity that are capable of cleaving a single-stranded nucleic acid, such as an mRNA, to which they have a complementary region. Thus, ribozymes (e.g., hammerhead ribozymes as described in Haselhoff and Gerlach 1988. Nature 334: 585-591) can be used to catalytically cleave NOVX mRNA transcripts to thereby inhibit translation of NOVX mRNA. A ribozyme having specificity for a NOVX-encoding nucleic acid can be designed based upon the nucleotide sequence of a NOVX cDNA disclosed herein (i.e., SEQ ID NO:2«-l, wherein n is an integer between 1 and 141). For example, a derivative of a Tetrahymena L-19 IVS RNA can be constructed in which the nucleotide sequence of the active site is complementary to the nucleotide sequence to be cleaved in a NOVX-encoding mRNA. See, e.g., U.S. Patent 4,987,071 to Cech, et al. and U.S. Patent 5,116,742 to Cech, et al. NOVX mRNA can also be used to select a catalytic RNA having a specific ribonuclease activity from a pool of RNA molecules. See, e.g., Bartel et al., (1993) Science 261:1411-1418.
Alternatively, NOVX gene expression can be inhibited by targeting nucleotide sequences complementary to the regulatory region of the NOVX nucleic acid (e.g., the NOVX promoter and/or enhancers) to form triple helical structures that prevent transcription of the NOVX gene in target cells. See, e.g., Helene, 1991. Anticancer Drug Des. 6: 569-84; Helene, et al. 1992. Ann. N.Y. Acad. Sci. 660: 27-36; Maher, 1992. Bioassays 14: 807-15. In various embodiments, the NOVX nucleic acids can be modified at the base moiety, sugar moiety or phosphate backbone to improve, e.g., the stability, hybridization, or solubility of the molecule. For example, the deoxyribose phosphate backbone of the nucleic acids can be modified to generate peptide nucleic acids. See, e.g., Hyrup, et al., 1996. BioorgMed Chem 4: 5-23. As used herein, the terms "peptide nucleic acids" or "PNAs" refer to nucleic acid mimics (e.g., DNA mimics) in which the deoxyribose phosphate backbone is replaced by a pseudopeptide backbone and only the four natural nucleotide bases are retained. The neutral backbone of PNAs has been shown to allow for specific hybridization to DNA and RNA under conditions of low ionic strength. The synthesis of PNA oligomer can be performed using standard solid phase peptide synthesis protocols as described in Hyrup, et al., 1996. supra; Perry-O'Keefe, et al., 1996. Proc. Natl. Acad. Sci. USA 93: 14670-14675. PNAs of NOVX can be used in therapeutic and diagnostic applications. For example, PNAs can be used as antisense or antigene agents for sequence-specific modulation of gene expression by, e.g., inducing transcription or translation arrest or inhibiting replication. PNAs of NOVX can also be used, for example, in the analysis of single base pair mutations in a gene (e.g., PNA directed PCR clamping; as artificial restriction enzymes when used in combination with other enzymes, e.g., Si nucleases (See, Hyrup, et ah, 1996.supra); or as probes or primers for DNA sequence and hybridization (See, Hyrup, et al., 1996, supra; Perry-O'Keefe, et al., 1996. supra).
In another embodiment, PNAs of NOVX can be modified, e.g., to enhance their stability or cellular uptake, by attaching lipophilic or other helper groups to PNA, by the formation of PNA-DNA chimeras, or by the use of liposomes or other techniques of drug delivery known in the art. For example, PNA-DNA chimeras of NOVX can be generated that may combine the advantageous properties of PNA and DNA. Such chimeras allow DNA recognition enzymes (e.g., RNase H and DNA polymerases) to interact with the DNA portion while the PNA portion would provide high binding affinity and specificity. PNA-DNA chimeras can be linked using linkers of appropriate lengths selected in terms of base stacking, number of bonds between the nucleotide bases, and orientation (see, Hyrup, et al., 1996. supra). The synthesis of PNA-DNA chimeras can be performed as described in Hyrup, et ah, 1996. supra and Finn, et ah, 1996. Nucl Acids Res 24: 3357-3363. For example, a DNA chain can be synthesized on a solid support using standard phosphoramidite coupling chemistry, and modified nucleoside analogs, e.g.,
5'-(4-methoxytrityl)amino-5'-deoxy-thymidine phosphoramidite, can be used between the PNA and the 5' end of DNA. See, e.g., Mag, et al, 1989. Nucl Acid Res 17: 5973-5988. PNA monomers are then coupled in a stepwise manner to produce a chimeric molecule with a 5' PNA segment and a 3' DNA segment. See, e.g., Finn, et al., 1996. supra. Alternatively, chimeric molecules can be synthesized with a 5' DNA segment and a 3' PNA segment. See, e.g., Petersen, et al., 1975. Bioorg. Med. Chem. Lett. 5: 1119-11124. In other embodiments, the oligonucleotide may include other appended groups such as peptides (e.g., for targeting host cell receptors in vivo), or agents facilitating transport across the cell membrane (see, e.g., Letsinger, et al., 1989. Proc. Natl. Acad. Sci. U.S.A. 86: 6553-6556; Lemairre, et al, \9 7. Proc. Natl. Acad. Sci. 84: 648-652; PCT Publication No. WO88/09810) or the blood-brain barrier (see, e.g., PCT Publication No. WO 89/10134). In addition, oligonucleotides can be modified with hybridization triggered cleavage agents (see, e.g., Krol, et al, 1988. BioTechniques 6:958-976) or intercalating agents (see, e.g., Zon, 1988. Pharm. Res. 5: 539-549). To this end, the oligonucleotide may be conjugated to another molecule, e.g., a peptide, a hybridization triggered cross-linking agent, a transport agent, a hybridization-triggered cleavage agent, and the like. NOVX Polypeptides
A polypeptide according to the invention includes a polypeptide including the amino acid sequence of NOVX polypeptides whose sequences are provided in any one of SEQ ID NO:272, wherein n is an integer between 1 and 141. The invention also includes a mutant or variant protein any of whose residues may be changed from the conesponding residues shown in any one of SEQ ID NO:2«, wherein n is an integer between 1 and 141, while still encoding a protein that maintains its NOVX activities and physiological functions, or a functional fragment thereof.
In general, a NOVX variant that preserves NOVX-like function includes any variant in which residues at a particular position in the sequence have been substituted by other amino acids, and further include the possibility of inserting an additional residue or residues between two residues of the parent protein as well as the possibility of deleting one or more residues from the parent sequence. Any amino acid substitution, insertion, or deletion is encompassed by the invention. In favorable circumstances, the substitution is a conservative substitution as defined above. One aspect of the invention pertains to isolated NOVX proteins, and biologically-active portions thereof, or derivatives, fragments, analogs or homologs thereof. Also provided are polypeptide fragments suitable for use as immunogens to raise anti-NOVX antibodies. In one embodiment, native NOVX proteins can be isolated from cells or tissue sources by an appropriate purification scheme using standard protein purification techniques. In another embodiment, NOVX proteins are produced by recombinant DNA techniques. Alternative to recombinant expression, a NOVX protein or polypeptide can be synthesized chemically using standard peptide synthesis techniques.
An "isolated" or "purified" polypeptide or protein or biologically-active portion thereof is substantially free of cellular material or other contaminating proteins from the cell or tissue source from which the NOVX protein is derived, or substantially free from chemical precursors or other chemicals when chemically synthesized. The language
"substantially free of cellular material" includes preparations of NOVX proteins in which the protein is separated from cellular components of the cells from which it is isolated or recombinantly-produced. In one embodiment, the language "substantially free of cellular material" includes preparations of NOVX proteins having less than about 30% (by dry weight) of non-NOVX proteins (also referred to herein as a "contaminating protein"), more preferably less than about 20% of non-NOVX proteins, still more preferably less than about 10% of non-NOVX proteins, and most preferably less than about 5% of non-NOVX proteins. When the NOVX protein or biologically-active portion thereof is recombinantly-produced, it is also preferably substantially free of culture medium, i.e., culture medium represents less than about 20%, more preferably less than about 10%, and most preferably less than about 5% of the volume of the NOVX protein preparation.
The language "substantially free of chemical precursors or other chemicals" includes preparations of NOVX proteins in which the protein is separated from chemical precursors or other chemicals that are involved in the synthesis of the protein. In one embodiment, the language "substantially free of chemical precursors or other chemicals" includes preparations of NOVX proteins having less than about 30% (by dry weight) of chemical precursors or non-NOVX chemicals, more preferably less than about 20% chemical precursors or non-NOVX chemicals, still more preferably less than about 10% chemical precursors or non-NOVX chemicals, and most preferably less than about 5% chemical precursors or non-NOVX chemicals.
Biologically-active portions of NOVX proteins include peptides comprising amino acid sequences sufficiently homologous to or derived from the amino acid sequences of the NOVX proteins (e.g., the amino acid sequence of SEQ ID NO:2«, wherein n is an integer between 1 and 141) that include fewer amino acids than the full-length NOVX proteins, and exhibit at least one activity of a NOVX protein. Typically, biologically-active portions comprise a domain or motif with at least one activity of the NOVX protein. A biologically-active portion of a NOVX protein can be a polypeptide which is, for example, 10, 25, 50, 100 or more amino acid residues in length.
Moreover, other biologically-active portions, in which other regions of the protein are deleted, can be prepared by recombinant techniques and evaluated for one or more of the functional activities of a native NOVX protein.
In an embodiment, the NOVX protein has an amino acid sequence of SEQ ID NO:2«, wherein n is an integer between 1 and 141. In other embodiments, the NOVX protein is substantially homologous to SEQ ID NO:2«, wherein n is an integer between 1 and 141, and retains the functional activity of the protein of SEQ ID NO:2«, wherein n is an integer between 1 and 141, yet differs in amino acid sequence due to natural allelic variation or mutagenesis, as described in detail, below. Accordingly, in another embodiment, the NOVX protein is a protein that comprises an amino acid sequence at least about 45% homologous to the amino acid sequence of SEQ ID NO:2«, wherein n is an integer between 1 and 141, and retains the functional activity of the NOVX proteins of SEQ ID NO:2n, wherein n is an integer between 1 and 141.
Determining Homology Between Two or More Sequences To determine the percent homology of two amino acid sequences or of two nucleic acids, the sequences are aligned for optimal comparison purposes (e.g., gaps can be introduced in the sequence of a first amino acid or nucleic acid sequence for optimal alignment with a second amino or nucleic acid sequence). The amino acid residues or nucleotides at corresponding amino acid positions or nucleotide positions are then compared. When a position in the first sequence is occupied by the same amino acid residue or nucleotide as the corcesponding position in the second sequence, then the molecules are homologous at that position (i.e., as used herein amino acid or nucleic acid "homology" is equivalent to amino acid or nucleic acid "identity").
The nucleic acid sequence homology may be determined as the degree of identity between two sequences. The homology may be determined using computer programs known in the art, such as GAP software provided in the GCG program package. See, Needleman and Wunsch, 1970. J Mol Biol 48: 443-453. Using GCG GAP software with the following settings for nucleic acid sequence comparison: GAP creation penalty of 5.0 and GAP extension penalty of 0.3, the coding region of the analogous nucleic acid sequences refened to above exhibits a degree of identity preferably of at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, or 99%, with the CDS (encoding) part of the DNA sequence of SEQ ID NO:2«-l , wherein n is an integer between 1 and 141.
The term "sequence identity" refers to the degree to which two polynucleotide or polypeptide sequences are identical on a residue-by-residue basis over a particular region of comparison. The term "percentage of sequence identity" is calculated by comparing two optimally aligned sequences over that region of comparison, determining the number of positions at which the identical nucleic acid base (e.g., A, T, C, G, U, or I, in the case of nucleic acids) occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the region of comparison (i.e., the window size), and multiplying the result by 100 to yield the percentage of sequence identity. The term "substantial identity" as used herein denotes a characteristic of a polynucleotide sequence, wherein the polynucleotide comprises a sequence that has at least 80 percent sequence identity, preferably at least 85 percent identity and often 90 to 95 percent sequence identity, more usually at least 99 percent sequence identity as compared to a reference sequence over a comparison region.
Chimeric and Fusion Proteins The invention also provides NOVX chimeric or fusion proteins. As used herein, a
NOVX "chimeric protein" or "fusion protein" comprises a NOVX polypeptide operatively-linked to a non-NOVX polypeptide. An "NOVX polypeptide" refers to a polypeptide having an amino acid sequence corresponding to a NOVX protein of SEQ ID NO:2«, wherein n is an integer between 1 and 141, whereas a "non-NOVX polypeptide" refers to a polypeptide having an amino acid sequence conesponding to a protein that is not substantially homologous to the NOVX protein, e.g., a protein that is different from the NOVX protein and that is derived from the same or a different organism. Within a NOVX fusion protein the NOVX polypeptide can correspond to all or a portion of a NOVX protein. In one embodiment, a NOVX fusion protein comprises at least one biologically-active portion of a NOVX protein. In another embodiment, a NOVX fusion protein comprises at least two biologically-active portions of a NOVX protein. In yet another embodiment, a NOVX fusion protein comprises at least three biologically-active portions of a NOVX protein. Within the fusion protein, the term "operatively-linked" is intended to indicate that the NOVX polypeptide and the non-NOVX polypeptide are fused in-frame with one another. The non-NOVX polypeptide can be fused to the N-terminus or C-terminus of the NOVX polypeptide. In one embodiment, the fusion protein is a GST-NO VX fusion protein in which the
NOVX sequences are fused to the C-terminus of the GST (glutathione S-transferase) sequences. Such fusion proteins can facilitate the purification of recombinant NOVX polypeptides.
In another embodiment, the fusion protein is a NOVX protein containing a heterologous signal sequence at its N-terminus. In certain host cells (e.g, mammalian host cells), expression and or secretion of NOVX can be increased through use of a heterologous signal sequence.
In yet another embodiment, the fusion protein is a NOVX-immunoglobulin fusion protein in which the NOVX sequences are fused to sequences derived from a member of the immunoglobulin protein family. The NOVX-immunoglobulin fusion proteins of the invention can be incoφorated into pharmaceutical compositions and administered to a subject to inhibit an interaction between a NOVX ligand and a NOVX protein on the surface of a cell, to thereby suppress NOVX-mediated signal transduction in vivo. The NOVX-immunoglobulin fusion proteins can be used to affect the bioavailability of a NOVX cognate ligand. Inhibition of the NOVX ligand/NOVX interaction may be useful therapeutically for both the treatment of proliferative and differentiative disorders, as well as modulating (e.g. promoting or inhibiting) cell survival. Moreover, the NOVX-immunoglobulin fusion proteins of the invention can be used as immunogens to produce anti-NOVX antibodies in a subject, to purify NOVX ligands, and in screening assays to identify molecules that inhibit the interaction of NOVX with a NOVX ligand. A NOVX chimeric or fusion protein of the invention can be produced by standard recombinant DNA techniques. For example, DNA fragments coding for the different polypeptide sequences are ligated together in-frame in accordance with conventional techniques, e.g., by employing blunt-ended or stagger-ended termini for ligation, restriction enzyme digestion to provide for appropriate termini, filling-in of cohesive ends as appropriate, alkaline phosphatase treatment to avoid undesirable joining, and enzymatic ligation. In another embodiment, the fusion gene can be synthesized by conventional techniques including automated DNA synthesizers. Alternatively, PCR amplification of gene fragments can be carried out using anchor primers that give rise to complementary overhangs between two consecutive gene fragments that can subsequently be annealed and reamplified to generate a chimeric gene sequence (see, e.g., Ausubel, et al. (eds.) CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, John Wiley & Sons, 1992). Moreover, many expression vectors are commercially available that already encode a fusion moiety (e.g., a GST polypeptide). A NOVX-encoding nucleic acid can be cloned into such an expression vector such that the fusion moiety is linked in-frame to the NOVX protein.
NOVX Agonists and Antagonists The invention also pertains to variants of the NOVX proteins that function as either NOVX agonists (i.e., mimetics) or as NOVX antagonists. Variants of the NOVX protein can be generated by mutagenesis (e.g., discrete point mutation or truncation of the NOVX protein). An agonist of the NOVX protein can retain substantially the same, or a subset of, the biological activities of the naturally occurring form of the NOVX protein. An antagonist of the NOVX protein can inhibit one or more of the activities of the naturally occurring form of the NOVX protein by, for example, competitively binding to a downstream or upstream member of a cellular signaling cascade which includes the NOVX protein. Thus, specific biological effects can be elicited by treatment with a variant of limited function. In one embodiment, treatment of a subject with a variant having a subset of the biological activities of the naturally occurring form of the protein has fewer side effects in a subject relative to treatment with the naturally occurring form of the NOVX proteins.
Variants of the NOVX proteins that function as either NOVX agonists (i.e., mimetics) or as NOVX antagonists can be identified by screening combinatorial libraries of mutants (e.g., truncation mutants) of the NOVX proteins for NOVX protein agonist or antagonist activity. In one embodiment, a variegated library of NOVX variants is generated by combinatorial mutagenesis at the nucleic acid level and is encoded by a variegated gene library. A variegated library of NOVX variants can be produced by, for example, enzymatically ligating a mixture of synthetic oligonucleotides into gene sequences such that a degenerate set of potential NOVX sequences is expressible as individual polypeptides, or alternatively, as a set of larger fusion proteins (e.g., for phage display) containing the set of NOVX sequences therein. There are a variety of methods which can be used to produce libraries of potential NOVX variants from a degenerate oligonucleotide sequence. Chemical synthesis of a degenerate gene sequence can be performed in an automatic DNA synthesizer, and the synthetic gene then ligated into an appropriate expression vector. Use of a degenerate set of genes allows for the provision, in one mixture, of all of the sequences encoding the desired set of potential NOVX sequences. Methods for synthesizing degenerate oligonucleotides are well-known within the art. See, e.g., Narang, 1983. Tetrahedron 39: 3; Itakura, et al, 1984. Annu. Rev. Biochem. 53: 323; Itakura, et al, 1984. Science 198: 1056; Ike, et al, 1983. Nucl. Acids Res. 11: 477. Polypeptide Libraries
In addition, libraries of fragments of the NOVX protein coding sequences can be used to generate a variegated population of NOVX fragments for screening and subsequent selection of variants of a NOVX protein. In one embodiment, a library of coding sequence fragments can be generated by treating a double stranded PCR fragment of a NOVX coding sequence with a nuclease under conditions wherein nicking occurs only about once per molecule, denaturing the double sfranded DNA, renaturing the DNA to form double-stranded DNA that can include sense/antisense pairs from different nicked products, removing single stranded portions from reformed duplexes by treatment with Si nuclease, and ligating the resulting fragment library into an expression vector. By this method, expression libraries can be derived which encodes N-terminal and internal fragments of various sizes of the NOVX proteins.
Various techniques are known in the art for screening gene products of combinatorial libraries made by point mutations or truncation, and for screening cDNA libraries for gene products having a selected property. Such techniques are adaptable for rapid screening of the gene libraries generated by the combinatorial mutagenesis of NOVX proteins. The most widely used techniques, which are amenable to high throughput analysis, for screening large gene libraries typically include cloning the gene library into replicable expression vectors, transforming appropriate cells with the resulting library of vectors, and expressing the combinatorial genes under conditions in which detection of a desired activity facilitates isolation of the vector encoding the gene whose product was detected. Recursive ensemble mutagenesis (REM), a new technique that enhances the frequency of functional mutants in the libraries, can be used in combination with the screening assays to identify NOVX variants. See, e.g., Arkin and Yourvan, 1992. Proc. Natl. Acad. Sci. USA 89: 7811-7815; Delgrave, et al, 1993. Protein Engineering 6:327-331.
NOVX Antibodies The term "antibody" as used herein refers to immunoglobulin molecules and immunologically active portions of immunoglobulin (Ig) molecules, i.e., molecules that contain an antigen binding site that specifically binds (immunoreacts with) an antigen. Such antibodies include, but are not limited to, polyclonal, monoclonal, chimeric, single chain, Fa , Fat>' and F b-μ fragments, and an Fa expression library. In general, antibody molecules obtained from humans relates to any of the classes IgG, IgM, IgA, IgE and IgD, which differ from one another by the nature of the heavy chain present in the molecule. Certain classes have subclasses as well, such as IgGi, IgG2, and others. Furthermore, in humans, the light chain may be a kappa chain or a lambda chain. Reference herein to antibodies includes a reference to all such classes, subclasses and types of human antibody species .
An isolated protein of the invention intended to serve as an antigen, or a portion or fragment thereof, can be used as an immunogen to generate antibodies that immunospecifically bind the antigen, using standard techniques for polyclonal and monoclonal antibody preparation. The full-length protein can be used or, alternatively, the invention provides antigenic peptide fragments of the antigen for use as immunogens. An antigenic peptide fragment comprises at least 6 amino acid residues of the amino acid sequence of the full length protein, such as an amino acid sequence of SEQ ID NO:2«, wherein n is an integer between 1 and 141, and encompasses an epitope thereof such that an antibody raised against the peptide forms a specific immune complex with the full length protein or with any fragment that contains the epitope. Preferably, the antigenic peptide comprises at least 10 amino acid residues, or at least 15 amino acid residues, or at least 20 amino acid residues, or at least 30 amino acid residues. Prefened epitopes encompassed by the antigenic peptide are regions of the protein that are located on its surface; commonly these are hydrophilic regions. In certain embodiments of the invention, at least one epitope encompassed by the antigenic peptide is a region of NOVX that is located on the surface of the protein, e.g., a hydrophilic region. A hydrophobicity analysis of the human NOVX protein sequence will indicate which regions of a NOVX polypeptide are particularly hydrophilic and, therefore, are likely to encode surface residues useful for targeting antibody production. As a means for targeting antibody production, hydropathy plots showing regions of hydrophilicity and hydrophobicity may be generated by any method well known in the art, including, for example, the Kyte Doolittle or the Hopp Woods methods, either with or without Fourier transformation. See, e.g., Hopp and Woods, 1981, Proc. Nat. Acad. Sci. USA 78: 3824-3828; Kyte and Doolittle 1982, J. Mol. Biol. 157: 105-142, each incoφorated herein by reference in their entirety. Antibodies that are specific for one or more domains within an antigenic protein, or derivatives, fragments, analogs or homologs thereof, are also provided herein.
The term "epitope" includes any protein determinant capable of specific binding to an immunoglobulin or T-cell receptor. Epitopic determinants usually consist of chemically active surface groupings of molecules such as amino acids or sugar side chains and usually have specific three dimensional structural characteristics, as well as specific charge characteristics. A NOVX polypeptide or a fragment thereof comprises at least one antigenic epitope. An anti-NOVX antibody of the present invention is said to specifically bind to antigen NOVX when the equilibrium binding constant (KQ) is <1 μM, preferably < 100 nM, more preferably < 10 nM, and most preferably < 100 pM to about 1 pM, as measured by assays such as radioligand binding assays or similar assays known to those skilled in the art.
A protein of the mvention, or a derivative, fragment, analog, homolog or ortholog thereof, may be utilized as an immunogen in the generation of antibodies that immunospecifically bind these protein components.
Various procedures known within the art may be used for the production of polyclonal or monoclonal antibodies directed against a protein of the invention, or against derivatives, fragments, analogs homologs or orthologs thereof (see, for example, Antibodies: A Laboratory Manual, Harlow E, and Lane D, 1988, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, incoφorated herein by reference). Some of these antibodies are discussed below. Polyclonal Antibodies
For the production of polyclonal antibodies, various suitable host animals (e.g., rabbit, goat, mouse or other mammal) may be immunized by one or more injections with the native protein, a synthetic variant thereof, or a derivative of the foregoing. An appropriate irnmunogenic preparation can contain, for example, the naturally occurring immunogenic protein, a chemically synthesized polypeptide representing the irnmunogenic protein, or a recombinantly expressed immunogenic protein. Furthermore, the protein may be conjugated to a second protein known to be immunogenic in the mammal being immunized. Examples of such immunogenic proteins include but are not limited to keyhole limpet hemocyanin, serum albumin, bovine thyroglobulin, and soybean trypsin inhibitor. The preparation can further include an adjuvant. Various adjuvants used to increase the immunological response include, but are not limited to, Freund's (complete and incomplete), mineral gels (e.g., aluminum hydroxide), surface active substances (e.g., lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, dinifrophenol, etc.), adjuvants usable in humans such as Bacille Calmette-Guerin and Corynebacterium parvum, or similar immunostimulatory agents. Additional examples of adjuvants which can be employed include MPL-TDM adjuvant (monophosphoryl Lipid A, synthetic trehalose dicorynomycolate).
The polyclonal antibody molecules directed against the immunogenic protein can be isolated from the mammal (e.g., from the blood) and further purified by well known techniques, such as affinity chromatography using protem A or protein G, which provide primarily the IgG fraction of immune serum. Subsequently, or alternatively, the specific antigen which is the target of the immunoglobulin sought, or an epitope thereof, may be immobilized on a column to purify the immune specific antibody by immunoaffinity chromatography. Purification of immunoglobulins is discussed, for example, by D. Wilkinson (The Scientist, published by The Scientist, Inc., Philadelphia PA, Vol. 14, No. 8 (April 17, 2000), pp.25-28). Monoclonal Antibodies
The term "monoclonal antibody" (MAb) or "monoclonal antibody composition", as used herein, refers to a population of antibody molecules that contain only one molecular species of antibody molecule consisting of a unique light chain gene product and a unique heavy chain gene product. In particular, the complementarity determining regions (CDRs) of the monoclonal antibody are identical in all the molecules of the population. MAbs thus contain an antigen binding site capable of immunoreacting with a particular epitope of the antigen characterized by a unique binding affinity for it. Monoclonal antibodies can be prepared using hybridoma methods, such as those described by Kohler and Milstein, Nature, 256:495 (1975). In a hybridoma method, a mouse, hamster, or other appropriate host animal, is typically immunized with an immunizing agent to elicit lymphocytes that produce or are capable of producing antibodies that will specifically bind to the immunizing agent. Alternatively, the lymphocytes can be immunized in vitro.
The immunizing agent will typically include the protein antigen, a fragment thereof or a fusion protein thereof. Generally, either peripheral blood lymphocytes are used if cells of human origin are desired, or spleen cells or lymph node cells are used if non-human mammalian sources are desired. The lymphocytes are then fused with an immortalized cell line using a suitable fusing agent, such as polyethylene glycol, to form a hybridoma cell (Goding, Monoclonal Antibodies: Principles and Practice, Academic Press, (1986) pp. 59-103). Immortalized cell lines are usually transformed mammalian cells, particularly myeloma cells of rodent, bovine and human origin. Usually, rat or mouse myeloma cell lines are employed. The hybridoma cells can be cultured in a suitable culture medium that preferably contains one or more substances that inhibit the growth or survival of the unfused, immortalized cells. For example, if the parental cells lack the enzyme hypoxanthine guanine phosphoribosyl transferase (HGPRT or HPRT), the culture medium for the hybridomas typically will include hypoxanthine, aminopterin, and thymidine ("HAT medium"), which substances prevent the growth of HGPRT-deficient cells.
Preferred immortalized cell lines are those that fuse efficiently, support stable high level expression of antibody by the selected antibody-producing cells, and are sensitive to a medium such as HAT medium. More preferred immortalized cell lines are murine myeloma lines, which can be obtained, for instance, from the Salk Institute Cell
Distribution Center, San Diego, California and the American Type Culture Collection, Manassas, Virginia. Human myeloma and mouse-human heteromyeloma cell lines also have been described for the production of human monoclonal antibodies (Kozbor, J. Immunol., 133:3001 (1984); Brodeur et al., Monoclonal Antibody Production Techniques and Applications, Marcel Dekker, Inc., New York, ( 1987) pp. 51 -63).
The culture medium in which the hybridoma cells are cultured can then be assayed for the presence of monoclonal antibodies directed against the antigen. Preferably, the binding specificity of monoclonal antibodies produced by the hybridoma cells is determined by immunoprecipitation or by an in vitro binding assay, such as radioimmunoassay (RIA) or enzyme-linked immunoabsorbent assay (ELISA). Such techniques and assays are known in the art. The binding affinity of the monoclonal antibody can, for example, be determined by the Scatchard analysis of Munson and Pollard, Anal. Biochem., 107:220 (1980). It is an objective, especially important in therapeutic applications of monoclonal antibodies, to identify antibodies having a high degree of specificity and a high binding affinity for the target antigen.
After the desired hybridoma cells are identified, the clones can be subcloned by limiting dilution procedures and grown by standard methods (Goding, 1986). Suitable culture media for this puφose include, for example, Dulbecco's Modified Eagle's Medium and RPMI-1640 medium. Alternatively, the hybridoma cells can be grown in vivo as ascites in a mammal.
The monoclonal antibodies secreted by the subclones can be isolated or purified from the culture medium or ascites fluid by conventional immunoglobulin purification procedures such as, for example, protein A-Sepharose, hydroxylapatite chromatography, gel electrophoresis, dialysis, or affinity chromatography.
The monoclonal antibodies can also be made by recombinant DNA methods, such as those described in U.S. Patent No.4,816,567. DNA encoding the monoclonal antibodies of the invention can be readily isolated and sequenced using conventional procedures (e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of murine antibodies). The hybridoma cells of the invention serve as a prefened source of such DNA. Once isolated, the DNA can be placed into expression vectors, which are then transfected into host cells such as simian COS cells, Chinese hamster ovary (CHO) cells, or myeloma cells that do not otherwise produce immunoglobulin protein, to obtain the synthesis of monoclonal antibodies in the recombinant host cells. The DNA also can be modified, for example, by substituting the coding sequence for human heavy and light chain constant domains in place of the homologous murine sequences (U.S. Patent No.4,816,567; Morrison, Nature 368, 812-13 ( 1994)) or by covalently j oining to the immunoglobulin coding sequence all or part of the coding sequence for a non-immunoglobulin polypeptide. Such a non-immunoglobulin polypeptide can be substituted for the constant domains of an antibody of the invention, or can be substituted for the variable domains of one antigen-combining site of an antibody of the invention to create a chimeric bivalent antibody. Humanized Antibodies
The antibodies directed against the protein antigens of the invention can further comprise humanized antibodies or human antibodies. These antibodies are suitable for administration to humans without engendering an immune response by the human against the administered immunoglobulin. Humanized forms of antibodies are chimeric immunoglobulins, immunoglobulin chains or fragments thereof (such as Fv, Fab, Fab', F(ab')2 or other antigen-binding subsequences of antibodies) that are principally comprised of the sequence of a human immunoglobulin, and contain minimal sequence derived from a non-human immunoglobulin. Humanization can be performed following the method of Winter and co-workers (Jones et al., Nature, 321:522-525 (1986); Riechmann et al., Nature, 332:323-327 (1988); Verhoeyen et al., Science, 239:1534-1536 (1988)), by substituting rodent CDRs or CDR sequences for the corresponding sequences of a human antibody. (See also U.S. Patent No. 5,225,539.) In some instances, Fv framework residues of the human immunoglobulin are replaced by conesponding non-human residues. Humanized antibodies can also comprise residues which are found neither in the recipient antibody nor in the imported CDR or framework sequences. In general, the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin and all or substantially all of the framework regions are those of a human immunoglobulin consensus sequence. The humanized antibody optimally also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin (Jones et al., 1986; Riechmann et al., 1988; and Presta, Curr. Op. Struct. Biol., 2:593-596 (1992)).
Human Antibodies Fully human antibodies essentially relate to antibody molecules in which the entire sequence of both the light chain and the heavy chain, including the CDRs, arise from human genes. Such antibodies are termed "human antibodies", or "fully human antibodies" herein. Human monoclonal antibodies can be prepared by the trioma technique; the human B-cell hybridoma technique (see Kozbor, et al., 1983 Immunol Today 4: 72) and the EBV hybridoma technique to produce human monoclonal antibodies (see Cole, et al., 1985 In: MONOCLONAL ANTIBODIES AND CANCER THERAPY, Alan R. Liss, Inc., pp. 77-96). Human monoclonal antibodies may be utilized in the practice of the present invention and may be produced by using human hybridomas (see Cote, et al., 1983. Proc Natl Acad Sci USA 80: 2026-2030) or by transforming human B-cells with Epstein Barr Virus in vitro (see Cole, et al., 1985 In: MONOCLONAL ANTIBODIES AND CANCER THERAPY, Alan R. Liss, Inc., pp. 77-96). In addition, human antibodies can also be produced using additional techniques, including phage display libraries (Hoogenboom and Winter, J. Mol. Biol., 227:381 (1991); Marks et al., J. Mol. Biol., 222:581 (1991)). Similarly, human antibodies can be made by introducing human immunoglobulin loci into transgenic animals, e.g., mice in which the endogenous immunoglobulin genes have been partially or completely inactivated. Upon challenge, human antibody production is observed, which closely resembles that seen in humans in all respects, including gene rearrangement, assembly, and antibody repertoire. This approach is described, for example, in U.S. Patent Nos. 5,545,807; 5,545,806; 5,569,825; 5,625,126; 5,633,425; 5,661,016, and in Marks et al. (Bio/Technology 10. 779-783 (1992)); Lonberg et al. (Nature 368 856-859 (1994)); Morrison ( Nature 368, 812-13 (1994)); Fishwild et al,( Nature Biotechnology 14, 845-51 (1996)); Neuberger (Nature Biotechnology 14, 826 (1996)); and Lonberg and Huszar (Intern. Rev. Immunol. 13 65-93 (1995)).
Human antibodies may additionally be produced using transgenic nonhuman animals which are modified so as to produce fully human antibodies rather than the animal's endogenous antibodies in response to challenge by an antigen. (See PCT publication WO94/02602). The endogenous genes encoding the heavy and light immunoglobulin chains in the nonhuman host have been incapacitated, and active loci encoding human heavy and light chain immunoglobulins are inserted into the host's genome. The human genes are incoφorated, for example, using yeast artificial chromosomes containing the requisite human DNA segments. An animal which provides all the desired modifications is then obtained as progeny by crossbreeding intermediate transgenic animals containing fewer than the full complement of the modifications. The preferred embodiment of such a nonhuman animal is a mouse, and is termed the Xenomouse™ as disclosed in PCT publications WO 96/33735 and WO 96/34096. This animal produces B cells which secrete fully human immunoglobulins. The antibodies can be obtained directly from the animal after immunization with an immunogen of interest, as, for example, a preparation of a polyclonal antibody, or alternatively from immortalized B cells derived from the animal, such as hybridomas producing monoclonal antibodies. Additionally, the genes encoding the immunoglobulins with human variable regions can be recovered and expressed to obtain the antibodies directly, or can be further modified to obtain analogs of antibodies such as, for example, single chain Fv molecules.
An example of a method of producing a nonhuman host, exemplified as a mouse, lacking expression of an endogenous immunoglobulin heavy chain is disclosed in U.S. Patent No. 5,939,598. It can be obtained by a method including deleting the J segment genes from at least one endogenous heavy chain locus in an embryonic stem cell to prevent reaπangement of the locus and to prevent formation of a transcript of a rearranged immunoglobulin heavy chain locus, the deletion being effected by a targeting vector containing a gene encoding a selectable marker; and producing from the embryonic stem cell a transgenic mouse whose somatic and germ cells contain the gene encoding the selectable marker.
A method for producing an antibody of interest, such as a human antibody, is disclosed in U.S. Patent No. 5,916,771. It includes introducing an expression vector that contains a nucleotide sequence encoding a heavy chain into one mammalian host cell in culture, introducing an expression vector containing a nucleotide sequence encoding a light chain into another mammalian host cell, and fusing the two cells to form a hybrid cell. The hybrid cell expresses an antibody containing the heavy chain and the light chain. In a further improvement on this procedure, a method for identifying a clinically relevant epitope on an immunogen, and a conelative method for selecting an antibody that binds immunospecifically to the relevant epitope with high affinity, are disclosed in PCT publication WO 99/53049.
Fab Fragments and Single Chain Antibodies
According to the invention, techniques can be adapted for the production of single-chain antibodies specific to an antigenic protein of the invention (see e.g., U.S. Patent No. 4,946,778). In addition, methods can be adapted for the construction of Fa expression libraries (see e.g., Huse, et al., 1989 Science 246: 1275-1281) to allow rapid and effective identification of monoclonal Fab fragments with the desired specificity for a protein or derivatives, fragments, analogs or homologs thereof. Antibody fragments that contain the idiotypes to a protein antigen may be produced by techniques known in the art including, but not limited to: (i) an F(ay)2 fragment produced by pepsin digestion of an antibody molecule; (ii) an Fab fragment generated by reducing the disulfide bridges of an F(a ')2 fragment; (iii) an Fab fragment generated by the treatment of the antibody molecule with papain and a reducing agent and (iv) Fv fragments.
Bispecific Antibodies
Bispecific antibodies are monoclonal, preferably human or humanized, antibodies that have binding specificities for at least two different antigens. In the present case, one of the binding specificities is for an antigenic protein of the invention. The second binding target is any other antigen, and advantageously is a cell-surface protein or receptor or receptor subunit.
Methods for making bispecific antibodies are known in the art. Traditionally, the recombinant production of bispecific antibodies is based on the co-expression of two immunoglobulin heavy-chain/light-chain pairs, where the two heavy chains have different specificities (Milstein and Cuello, Nature, 305:537-539 (1983)). Because of the random assortment of immunoglobulin heavy and light chains, these hybridomas (quadromas) produce a potential mixture often different antibody molecules, of which only one has the conect bispecific structure. The purification of the conect molecule is usually accomplished by affinity chromatography steps. Similar procedures are disclosed in WO 93/08829, published 13 May 1993, and in Traunecker et al., EMBO J., 10:3655-3659 (1991).
Antibody variable domains with the desired binding specificities (antibody-antigen combining sites) can be fused to immunoglobulin constant domain sequences. The fusion preferably is with an immunoglobulin heavy-chain constant domain, comprising at least part of the hinge, CH2, and CH3 regions. It is prefened,to have the first heavy-chain constant region (CHI) containing the site necessary for light-chain binding present in at least one of the fusions. DNAs encoding the immunoglobulin heavy-chain fusions and, if desired, the immunoglobulin light chain, are inserted into separate expression vectors, and are co-transfected into a suitable host organism. For further details of generating bispecific antibodies see, for example, Suresh et al., Methods in Enzymology, 121:210 (1986).
According to another approach described in WO 96/27011, the interface between a pair of antibody molecules can be engineered to maximize the percentage of heterodimers which are recovered from recombinant cell culture. The prefened interface comprises at least a part of the CH3 region of an antibody constant domain. In this method, one or more small amino acid side chains from the interface of the first antibody molecule are replaced with larger side chains (e.g. tyrosine or tryptophan). Compensatory "cavities" of identical or similar size to the large side chain(s) are created on the interface of the second antibody molecule by replacing large amino acid side chains with smaller ones (e.g. alanine or threonine). This provides a mechanism for increasing the yield of the heterodimer over other unwanted end-products such as homodimers.
Bispecific antibodies can be prepared as full length antibodies or antibody fragments (e.g. F(ab')2 bispecific antibodies). Techniques for generating bispecific antibodies from antibody fragments have been described in the literature. For example, bispecific antibodies can be prepared using chemical linkage. Brennan et al., Science
229:81 (1985) describe a procedure wherein intact antibodies are proteolytically cleaved to generate F(ab')2 fragments. These fragments are reduced in the presence of the dithiol complexing agent sodium arsenite to stabilize vicinal dithiols and prevent intermolecular disulfide formation. The Fab' fragments generated are then converted to thionitrobenzoate (TNB) derivatives. One of the Fab'-TNB derivatives is then reconverted to the Fab'-thiol by reduction with mercaptoethylamine and is mixed with an equimolar amount of the other Fab'-TNB derivative to form the bispecific antibody. The bispecific antibodies produced can be used as agents for the selective immobilization of enzymes.
Additionally, Fab' fragments can be directly recovered from E. coli and chemically coupled to form bispecific antibodies. Shalaby et al., J. Exp. Med. 175:217-225 (1992) describe the production of a fully humanized bispecific antibody F(ab')2 molecule. Each Fab' fragment was separately secreted from E. coli and subjected to directed chemical coupling in vitro to form the bispecific antibody. The bispecific antibody thus formed was able to bind to cells overexpressing the ErbB2 receptor and normal human T cells, as well as trigger the lytic activity of human cytotoxic lymphocytes against human breast tumor targets.
Various techniques for making and isolating bispecific antibody fragments directly from recombinant cell culture have also been described. For example, bispecific antibodies have been produced using leucine zippers. Kostelny et al., J. Immunol. 148(5): 1547-1553 (1992). The leucine zipper peptides from the Fos and Jun proteins were linked to the Fab' portions of two different antibodies by gene fusion. The antibody homodimers were reduced at the hinge region to form monomers and then re-oxidized to form the antibody heterodimers. This method can also be utilized for the production of antibody homodimers. The "diabody" technology described by Hollinger et al., Proc. Natl. Acad. Sci. USA 90:6444-6448 (1993) has provided an alternative mechanism for making bispecific antibody fragments. The fragments comprise a heavy-chain variable domain (VH) connected to a light-chain variable domain (VL) by a linker which is too short to allow pairing between the two domains on the same chain. Accordingly, the VH and VL domains of one fragment are forced to pair with the complementary V and VH domains of another fragment, thereby forming two antigen-binding sites. Another strategy for making bispecific antibody fragments by the use of single-chain Fv (sFv) dimers has also been reported. See, Gruber et al., J. Immunol. 152:5368 (1994). Antibodies with more than two valencies are contemplated. For example, trispecific antibodies can be prepared. Tutt et al., J. Immunol. 147:60 (1991).
Exemplary bispecific antibodies can bind to two different epitopes, at least one of which originates in the protein antigen of the invention. Alternatively, an anti-antigenic arm of an immunoglobulin molecule can be combined with an arm which binds to a triggering molecule on a leukocyte such as a T-cell receptor molecule (e.g. CD2, CD3, CD28, or B7), or Fc receptors for IgG (FcγR), such as FcγRI (CD64), FcγRII (CD32) and FcγRIII (CD 16) so as to focus cellular defense mechanisms to the cell expressing the particular antigen. Bispecific antibodies can also be used to direct cytotoxic agents to cells which express a particular antigen. These antibodies possess an antigen-binding arm and an arm which binds a cytotoxic agent or a radionuclide chelator, such as EOTUBE, DPTA, DOTA, or TETA. Another bispecific antibody of interest binds the protein antigen described herein and further binds tissue factor (TF).
Heteroconjugate Antibodies
Heteroconjugate antibodies are also within the scope of the present invention. Heteroconjugate antibodies are composed of two covalently joined antibodies. Such antibodies have, for example, been proposed to target immune system cells to unwanted cells (U.S. Patent No. 4,676,980), and for treatment of HTV infection (WO 91/00360; WO 92/200373; EP 03089). It is contemplated that the antibodies can be prepared in vitro using known methods in synthetic protein chemistry, including those involving crosslinking agents. For example, immunotoxins can be constructed using a disulfide exchange reaction or by forming a thioether bond. Examples of suitable reagents for this puφose include iminothiolate and methyl-4-mercaptobutyrimidate and those disclosed, for example, in U.S. Patent No. 4,676,980.
Effector Function Engineering
It can be desirable to modify the antibody of the invention with respect to effector function, so as to enhance, e.g., the effectiveness of the antibody in treating cancer. For example, cysteine residue(s) can be introduced into the Fc region, thereby allowing interchain disulfide bond formation in this region. The homodimeric antibody thus generated can have improved internalization capability and/or increased complement-mediated cell killing and antibody-dependent cellular cytotoxicity (ADCC). See Caron et al., J. Exp Med„ 176: 1191-1195 (1992) and Shopes, J. Immunol., 148:
2918-2922 (1992). Homodimeric antibodies with enhanced anti-tumor activity can also be prepared using heterobifunctional cross-linkers as described in Wolff et al. Cancer Research, 53: 2560-2565 (1993). Alternatively, an antibody can be engineered that has dual Fc regions and can thereby have enhanced complement lysis and ADCC capabilities. See Stevenson et al., Anti-Cancer Drug Design, 3: 219-230 (1989).
Immunoconjugates
The invention also pertains to immunoconjugates comprising an antibody conjugated to a cytotoxic agent such as a chemotherapeutic agent, toxin (e.g., an enzymatically active toxin of bacterial, fungal, plant, or animal origin, or fragments thereof), or a radioactive isotope (i.e., a radioconjugate).
Chemotherapeutic agents useful in the generation of such immunoconjugates have been described above. Enzymatically active toxins and fragments thereof that can be used include diphtheria A chain, nonbinding active fragments of diphtheria toxin, exotoxin A chain (from Pseudomonas aeruginosa), ricin A chain, abrin A chain, modeccin A chain, alpha-sarcin, Aleurites fordii proteins, dianthin proteins, Phytolaca americana proteins (PAPI, PAPII, and PAP-S), momordica charantia inhibitor, curcin, crotin, sapaonaria officinalis inhibitor, gelonin, mitogellin, restrictocin, phenomycin, enomycin, and the tricothecenes. A variety of radionuclides are available for the production of radioconjugated antibodies. Examples include 212Bi, 1311, 131In, 90Y, and 186Re. Conjugates of the antibody and cytotoxic agent are made using a variety of bifunctional protein-coupling agents such as N-succinimidyl-3-(2-pyridyldithiol) propionate (SPDP), iminothiolane (IT), bifunctional derivatives of imidoesters (such as dimethyl adipimidate HCL), active esters (such as disuccinimidyl suberate), aldehydes (such as glutareldehyde), bis-azido compounds (such as bis (p-azidobenzoyl) hexanediamine), bis-diazonium derivatives (such as bis-(p-diazoniumbenzoyl)-ethylenediamine), diisocyanates (such as tolyene 2,6-dϋsocyanate), and bis-active fluorine compounds (such as l,5-difluoro-2,4-dinitrobenzene). For example, a ricin immunotoxin can be prepared as described in Vitetta et al., Science, 238: 1098 (1987). Carbon-14-labeled l-isothiocyanatobenzyl-3-methyldiethylene triaminepentaacetic acid (MX-DTPA) is an exemplary chelating agent for conjugation of radionucleotide to the antibody. See WO94/11026.
In another embodiment, the antibody can be conjugated to a "receptor" (such streptavidin) for utilization in tumor pretargeting wherein the antibody-receptor conjugate is administered to the patient, followed by removal of unbound conjugate from the circulation using a clearing agent and then administration of a "ligand" (e.g., avidin) that is in turn conjugated to a cytotoxic agent.
Immunoliposomes
The antibodies disclosed herein can also be formulated as immunoliposomes. Liposomes containing the antibody are prepared by methods known in the art, such as described in Epstein et al., Proc. Natl. Acad. Sci. USA. 82: 3688 (1985); Hwang et al., Proc. Natl Acad. Sci. USA. 77: 4030 (1980); and U.S. Pat. Nos. 4,485,045 and 4,544,545. Liposomes with enhanced circulation time are disclosed in U.S. Patent No. 5,013,556.
Particularly useful liposomes can be generated by the reverse-phase evaporation method with a lipid composition comprising phosphatidylcholine, cholesterol, and PEG-derivatized phosphatidylethanolamine (PEG-PE). Liposomes are extruded through filters of defined pore size to yield liposomes with the desired diameter. Fab' fragments of the antibody of the present invention can be conjugated to the liposomes as described in Martin et al ., J. Biol. Chem., 257: 286-288 (1982) via a disulfide-interchange reaction. A chemotherapeutic agent (such as Doxorubicin) is optionally contained within the liposome. See Gabizon et al., J. National Cancer Inst.. 81(19): 1484 (1989). Diagnostic Applications of Antibodies Directed Against the Proteins of the Invention
Antibodies directed against a protein of the invention may be used in methods known within the art relating to the localization and/or quantitation of the protein (e.g., for use in measuring levels of the protein within appropriate physiological samples, for use in diagnostic methods, for use in imaging the protein, and the like). In a given embodiment, antibodies against the proteins, or derivatives, fragments, analogs or homologs thereof, that contain the antigen binding domain, are utilized as pharmacologically-active compounds (see below).
An antibody specific for a protein of the invention can be used to isolate the protein by standard techniques, such as immunoaffinity chromatography or immunoprecipitation. Such an antibody can facilitate the purification of the natural protein antigen from cells and of recombinantly produced antigen expressed in host cells. Moreover, such an antibody can be used to detect the antigenic protein (e.g., in a cellular lysate or cell supernatant) in order to evaluate the abundance and pattern of expression of the antigenic protein. Antibodies directed against the protein can be used diagnostically to monitor protein levels in tissue as part of a clinical testing procedure, e.g., to, for example, determine the efficacy of a given treatment regimen. Detection can be facilitated by coupling (i.e., physically linking) the antibody to a detectable substance. Examples of detectable substances include various enzymes, prosthetic groups, fluorescent materials, luminescent materials, bioluminescent materials, and radioactive materials. Examples of suitable enzymes include horseradish peroxidase, alkaline phosphatase, β-galactosidase, or acetylcholinesterase; examples of suitable prosthetic group complexes include sfreptavidin/biotin and avidin/biotin; examples of suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride orphycoerythrin; an example of a luminescent material includes luminol; examples of bioluminescent materials include luciferase, luciferin, and aequorin, and examples of suitable radioactive material include 1251, 1311, 35S or 3H. Antibody Therapeutics
Antibodies of the invention, including polyclonal, monoclonal, humanized and fully human antibodies, may used as therapeutic agents. Such agents will generally be employed to treat or prevent a disease or pathology in a subject. An antibody preparation, preferably one having high specificity and high affinity for its target antigen, is administered to the subject and will generally have an effect due to its binding with the target. Such an effect may be one of two kinds, depending on the specific nature of the interaction between the given antibody molecule and the target antigen in question. In the first instance, administration of the antibody may abrogate or inhibit the binding of the target with an endogenous ligand to which it naturally binds. In this case, the antibody binds to the target and masks a binding site of the naturally occurring ligand, wherein the ligand serves as an effector molecule. Thus the receptor mediates a signal transduction pathway for which ligand is responsible.
Alternatively, the effect may be one in which the antibody elicits a physiological result by virtue of binding to an effector binding site on the target molecule. In this case the target, a receptor having an endogenous ligand which may be absent or defective in the disease or pathology, binds the antibody as a sunogate effector ligand, initiating a receptor-based signal transduction event by the receptor.
A therapeutically effective amount of an antibody of the invention relates generally to the amount needed to achieve a therapeutic objective. As noted above, this may be a binding interaction between the antibody and its target antigen that, in certain cases, interferes with the functioning of the target, and in other cases, promotes a physiological response. The amount required to be administered will furthermore depend on the binding affinity of the antibody for its specific antigen, and will also depend on the rate at which an administered antibody is depleted from the free volume other subject to which it is administered. Common ranges for therapeutically effective dosing of an antibody or antibody fragment of the invention may be, by way of nonlimiting example, from about 0.1 mg/kg body weight to about 50 mg/kg body weight. Common dosing frequencies may range, for example, from twice daily to once a week.
Pharmaceutical Compositions of Antibodies
Antibodies specifically binding a protein of the invention, as well as other molecules identified by the screening assays disclosed herein, can be administered for the treatment of various disorders in the form of pharmaceutical compositions. Principles and considerations involved in preparing such compositions, as well as guidance in the choice of components are provided, for example, in Remington : The Science And Practice Of Pharmacy 19th ed. (Alfonso R. Gennaro, et al., editors) Mack Pub. Co., Easton, Pa. : 1995; Drug Absoφtion Enhancement : Concepts, Possibilities, Limitations, And Trends, Harwood Academic Publishers, Langhorne, Pa., 1994; and Peptide And Protein Drug Delivery (Advances In Parenteral Sciences, Vol.4), 1991, M. Dekker, New York. If the antigenic protein is intracellular and whole antibodies are used as inhibitors, internalizing antibodies are prefened. However, liposomes can also be used to deliver the antibody, or an antibody fragment, into cells. Where antibody fragments are used, the smallest inhibitory fragment that specifically binds to the binding domain of the target protein is prefened. For example, based upon the variable-region sequences of an antibody, peptide molecules can be designed that retain the ability to bind the target protein sequence. Such peptides can be synthesized chemically and/or produced by recombinant DNA technology. See, e.g., Marasco et al., Proc. Natl. Acad. Sci. USA, 90: 7889-7893 (1993). The formulation herein can also contain more than one active compound as necessary for the particular indication being freated, preferably those with complementary activities that do not adversely affect each other. Alternatively, or in addition, the composition can comprise an agent that enhances its function, such as, for example, a cytotoxic agent, cytokine, chemotherapeutic agent, or growth-inhibitory agent. Such molecules are suitably present in combination in amounts that are effective for the puφose intended.
The active ingredients can also be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly-(methylmethacrylate) microcapsules, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles, and nanocapsules) or in macroemulsions.
The formulations to be used for in vivo administration must be sterile. This is readily accomplished by filtration through sterile filtration membranes.
Sustained-release preparations can be prepared. Suitable examples of sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing the antibody, which matrices are in the form of shaped articles, e.g., films, or microcapsules. Examples of sustained-release matrices include polyesters, hydrogels (for example, poly(2-hydroxyethyl-methacrylate), or poly(vinylalcohol)), polylactides (U.S. Pat. No. 3,773,919), copolymers of L-glutamic acid and γ ethyl-L-glutamate, non-degradable ethylene-vinyl acetate, degradable lactic acid-glycolic acid copolymers such as the LUPRON DEPOT ™ (injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate), and poly-D-(-)-3-hydroxybutyric acid. While polymers such as ethylene-vinyl acetate and lactic acid-glycolic acid enable release of molecules for over 100 days, certain hydrogels release proteins for shorter time periods.
ELISA Assay
An agent for detecting an analyte protein is an antibody capable of binding to an analyte protein, preferably an antibody with a detectable label. Antibodies can be polyclonal, or more preferably, monoclonal. An intact antibody, or a fragment thereof (e.g., Fab or F(ab)2) can be used. The term "labeled", with regard to the probe or antibody, is intended to encompass direct labeling of the probe or antibody by coupling (i.e., physically linking) a detectable substance to the probe or antibody, as well as indirect labeling of the probe or antibody by reactivity with another reagent that is directly labeled. Examples of indirect labeling include detection of a primary antibody using a fluorescently-labeled secondary antibody and end-labeling of a DNA probe with biotin such that it can be detected with fluorescently-labeled sfreptavidin. The term "biological sample" is intended to include tissues, cells and biological fluids isolated from a subject, as well as tissues, cells and fluids present within a subject. Included within the usage of the term "biological sample", therefore, is blood and a fraction or component of blood including blood serum, blood plasma, or lymph. That is, the detection method of the invention can be used to detect an analyte mRNA, protein, or genomic DNA in a biological sample in vitro as well as in vivo. For example, in vitro techniques for detection of an analyte mRNA include Northern hybridizations and in situ hybridizations. In vitro techniques for detection of an analyte protein include enzyme linked immunosorbent assays (ELISAs), Western blots, immunoprecipitations, and immunofluorescence. In vitro techniques for detection of an analyte genomic DNA include Southern hybridizations. Procedures for conducting immunoassays are described, for example in "ELISA: Theory and Practice: Methods in Molecular Biology", Vol. 42, J. R. Crowther (Ed.) Human Press, Totowa, NJ, 1995; "Immunoassay", E. Diamandis and T. Christopoulus, Academic Press, Inc., San Diego, CA, 1996; and "Practice and Theory of Enzyme Immunoassays", P. Tijssen, Elsevier Science Publishers, Amsterdam, 1985. Furthermore, in vivo techniques for detection of an analyte protein include introducing into a subject a labeled anti-an analyte protein antibody. For example, the antibody can be labeled with a radioactive marker whose presence and location in a subject can be detected by standard imaging techniques. NOVX Recombinant Expression Vectors and Host Cells
Another aspect of the invention pertains to vectors, preferably expression vectors, containing a nucleic acid encoding a NOVX protein, or derivatives, fragments, analogs or homologs thereof. As used herein, the term "vector" refers to a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked. One type of vector is a "plasmid", which refers to a circular double stranded DNA loop into which additional DNA segments can be ligated. Another type of vector is a viral vector, wherein additional DNA segments can be ligated into the viral genome. Certain vectors are capable of autonomous replication in a host cell into which they are introduced (e.g., bacterial vectors having a bacterial origin of replication and episomal mammalian vectors). Other vectors (e.g., non-episomal mammalian vectors) are integrated into the genome of a host cell upon introduction into the host cell, and thereby are replicated along with the host genome. Moreover, certain vectors are capable of directing the expression of genes to which they are operatively-linked. Such vectors are refened to herein as "expression vectors". In general, useful expression vectors in recombinant DNA techniques are often in the form of plasmids. In the present specification, "plasmid" and "vector" can be used interchangeably as the plasmid is the most commonly used form of vector. However, the invention is intended to include such other forms of expression vectors, such as viral vectors (e.g., replication defective refroviruses, adenoviruses and adeno-associated viruses), which serve equivalent functions.
The recombinant expression vectors of the invention comprise a nucleic acid of the invention in a form suitable for expression of the nucleic acid in a host cell, which means that the recombinant expression vectors include one or more regulatory sequences, selected on the basis of the host cells to be used for expression, that is operatively-linked to the nucleic acid sequence to be expressed. Within a recombinant expression vector, "operably-linked" is intended to mean that the nucleotide sequence of interest is linked to the regulatory sequence(s) in a manner that allows for expression of the nucleotide sequence (e.g., in an in vitro transcription/translation system or in a host cell when the vector is introduced into the host cell). The term "regulatory sequence" is intended to includes promoters, enhancers and other expression control elements (e.g., polyadenylation signals). Such regulatory sequences are described, for example, in Goeddel, GENE EXPRESSION TECHNOLOGY: METHODS IN ENZYMOLOGY 185, Academic Press, San Diego, Calif. (1990). Regulatory sequences include those that direct constitutive expression of a nucleotide sequence in many types of host cell and those that direct expression of the nucleotide sequence only in certain host cells (e.g., tissue-specific regulatory sequences). It will be appreciated by those skilled in the art that the design of the expression vector can depend on such factors as the choice of the host cell to be transformed, the level of expression of protein desired, etc. The expression vectors of the invention can be introduced into host cells to thereby produce proteins or peptides, including fusion proteins or peptides, encoded by nucleic acids as described herein (e.g., NOVX proteins, mutant forms of NOVX proteins, fusion proteins, etc.).
The recombinant expression vectors of the invention can be designed for expression of NOVX proteins in prokaryotic or eukaryotic cells. For example, NOVX proteins can be expressed in bacterial cells such as Escherichia coli, insect cells (using baculovirus expression vectors) yeast cells or mammalian cells. Suitable host cells are discussed further in Goeddel, GENE EXPRESSION TECHNOLOGY: METHODS IN
ENZYMOLOGY 185, Academic Press, San Diego, Calif. (1990). Alternatively, the recombinant expression vector can be transcribed and translated in vitro, for example using T7 promoter regulatory sequences and T7 polymerase.
Expression of proteins in prokaryotes is most often carried out in Escherichia coli with vectors containing constitutive or inducible promoters directing the expression of either fusion or non-fusion proteins. Fusion vectors add a number of amino acids to a protein encoded therein, usually to the amino terminus of the recombinant protein. Such fusion vectors typically serve three puφoses: (i) to increase expression of recombinant protein; (ii) to increase the solubility of the recombinant protein; and (iii) to aid in the purification of the recombinant protein by acting as a ligand in affinity purification.
Often, in fusion expression vectors, a proteolytic cleavage site is introduced at the junction of the fusion moiety and the recombinant protein to enable separation of the recombinant protein from the fusion moiety subsequent to purification of the fusion protein. Such enzymes, and their cognate recognition sequences, include Factor Xa, thrombin and enterokinase. Typical fusion expression vectors include pGEX (Pharmacia Biotech Inc; Smith and Johnson, 1988. Ge«e 67: 31-40), pMAL (New England Biolabs, Beverly, Mass.) and pRIT5 (Pharmacia, Piscataway, N. J.) that fuse glutathione S-fransferase (GST), maltose E binding protein, or protein A, respectively, to the target recombinant protein.
Examples of suitable inducible non-fusion E. coli expression vectors include pTrc (Amrann et al, (1988) Gene 69:301-315) and pET lid (Studier et al, GENE EXPRESSION TECHNOLOGY: METHODS IN ENZYMOLOGY 185, Academic Press, San Diego, Calif. (1990) 60-89).
One strategy to maximize recombinant protem expression in E. coli is to express the protein in a host bacteria with an impaired capacity to proteolytically cleave the recombinant protein. See, e.g., Gottesman, GENE EXPRESSION TECHNOLOGY: METHODS IN ENZYMOLOGY 185, Academic Press, San Diego, Calif. (1990) 119-128. Another strategy is to alter the nucleic acid sequence of the nucleic acid to be inserted into an expression vector so that the individual codons for each amino acid are those preferentially utilized in E. coli (see, e.g., Wada, et al, 1992. Nucl. Acids Res. 20: 2111-2118). Such alteration of nucleic acid sequences of the invention can be carried out by standard DNA synthesis techniques.
In another embodiment, the NOVX expression vector is a yeast expression vector. Examples of vectors for expression in yeast Saccharomyces cerivisae include pYepSecl (Baldari, et al, 1987. EMBOJ. 6: 229-234), pMFa (Kurjan and Herskowitz, 1982. Cell 30: 933-943), pJRY88 (Schultz et al, 1987. Gene 54: 113-123), pYES2 (Invitrogen Coφoration, San Diego, Calif.), and picZ (InVitrogen Coφ, San Diego, Calif.).
Alternatively, NOVX can be expressed in insect cells using baculovirus expression vectors. Baculovirus vectors available for expression of proteins in cultured insect cells (e.g., SF9 cells) include the pAc series (Smith, et al, 1983. Mol. Cell. Biol. 3: 2156-2165) and the pVL series (Lucklow and Summers, 1989. Virology 170: 31-39). In yet another embodiment, a nucleic acid of the invention is expressed in mammalian cells using a mammalian expression vector. Examples of mammalian expression vectors include pCDM8 (Seed, 1987. Nature 329: 840) and ρMT2PC (Kaufman, et al, 1987. EMBO J. 6: 187-195). When used in mammalian cells, the expression vector's control functions are often provided by viral regulatory elements. For example, commonly used promoters are derived from polyoma, adenovirus 2, cytomegalovirus, and simian virus 40. For other suitable expression systems for both prokaryotic and eukaryotic cells see, e.g., Chapters 16 and 17 of Sambrook, et al, MOLECULAR CLONING: A LABORATORY MANUAL. 2nd ed., Cold Spring Harbor Laboratory, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1989.
In another embodiment, the recombinant mammalian expression vector is capable of directing expression of the nucleic acid preferentially in a particular cell type (e.g., tissue-specific regulatory elements are used to express the nucleic acid). Tissue-specific regulatory elements are known in the art. Non-limiting examples of suitable tissue-specific promoters include the albumin promoter (liver-specific; Pinkert, et al, 1987. Genes Dev. 1: 268-277), lymphoid-specific promoters (Calame and Eaton, 1988. Adv. Immunol. 43: 235-275), in particular promoters of T cell receptors (Winoto and Baltimore, 1989. EMBO J. 8: 729-733) and immunoglobulins (Banerji, et al, 1983. Cell 33: 729-740; Queen and Baltimore, 1983. Cell 33: 741-748), neuron-specific promoters (e.g., the neurofϊlament promoter; Byrne and Ruddle, 1989. Proc. Natl. Acad. Sci. USA 86: 5473-5477), pancreas-specific promoters (Edlund, et al, 1985. Science 230: 912-916), and mammary gland-specific promoters (e.g., milk whey promoter; U.S. Pat. No. 4,873,316 and European Application Publication No. 264, 166). Developmentally-regulated promoters are also encompassed, e.g., the murine hox promoters (Kessel and Grass, 1990. Science 249: 374-379) and the -fetoprotein promoter (Ca pes and Tilghman, 1989. Genes Dev. 3: 537-546).
The invention further provides a recombinant expression vector comprising a DNA molecule of the invention cloned into the expression vector in an antisense orientation. That is, the DNA molecule is operatively-linked to a regulatory sequence in a manner that allows for expression (by transcription of the DNA molecule) of an RNA molecule that is antisense to NOVX mRNA. Regulatory sequences operatively linked to a nucleic acid cloned in the antisense orientation can be chosen that direct the continuous expression of the antisense RNA molecule in a variety of cell types, for instance viral promoters and/or enhancers, or regulatory sequences can be chosen that direct constitutive, tissue specific or cell type specific expression of antisense RNA. The antisense expression vector can be in the form of a recombinant plasmid, phagemid or attenuated virus in which antisense nucleic acids are produced under the control of a high efficiency regulatory region, the activity of which can be determined by the cell type into which the vector is introduced. For a discussion of the regulation of gene expression using antisense genes see, e.g., Weintraub, et al, "Antisense RNA as a molecular tool for genetic analysis," Reviews-Trends in Genetics, Vol. 1(1) 1986. Another aspect of the invention pertains to host cells into which a recombinant expression vector of the invention has been introduced. The terms "host cell" and "recombinant host cell" are used interchangeably herein. It is understood that such terms refer not only to the particular subject cell but also to the progeny or potential progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term as used herein.
A host cell can be any prokaryotic or eukaryotic cell. For example, NOVX protein can be expressed in bacterial cells such as E. coli, insect cells, yeast or mammalian cells (such as Chinese hamster ovary cells (CHO) or COS cells). Other suitable host cells are known to those skilled in the art.
Vector DNA can be introduced into prokaryotic or eukaryotic cells via conventional transformation or transfection techniques. As used herein, the terms "transformation" and "transfection" are intended to refer to a variety of art-recognized techniques for introducing foreign nucleic acid (e.g. , DNA) into a host cell, including calcium phosphate or calcium chloride co-precipitation, DEAE-dexfran-mediated transfection, lipofection, or electroporation. Suitable methods for transforming or transfecting host cells can be found in Sambrook, et al. (MOLECULAR CLONING: A LABORATORY MANUAL. 2nd ed., Cold Spring Harbor Laboratory, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1989), and other laboratory manuals.
For stable transfection of mammalian cells, it is known that, depending upon the expression vector and transfection technique used, only a small fraction of cells may integrate the foreign DNA into their genome. In order to identify and select these integrants, a gene that encodes a selectable marker (e.g., resistance to antibiotics) is generally introduced into the host cells along with the gene of interest. Various selectable markers include those that confer resistance to drugs, such as G418, hygromycin and methotrexate. Nucleic acid encoding a selectable marker can be introduced into a host cell on the same vector as that encoding NOVX or can be introduced on a separate vector. Cells stably transfected with the introduced nucleic acid can be identified by drug selection (e.g., cells that have incoφorated the selectable marker gene will survive, while the other cells die).
A host cell of the invention, such as a prokaryotic or eukaryotic host cell in culture, can be used to produce (i.e., express) NOVX protein. Accordingly, the invention further provides methods for producing NOVX protein using the host cells of the invention. In one embodiment, the method comprises culturing the host cell of invention (into which a recombinant expression vector encoding NOVX protein has been introduced) in a suitable medium such that NOVX protein is produced. In another embodiment, the method further comprises isolating NOVX protein from the medium or the host cell.
Transgenic NOVX Animals
The host cells of the invention can also be used to produce non-human transgenic animals. For example, in one embodiment, a host cell of the invention is a fertilized oocyte or an embryonic stem cell into which NOVX protein-coding sequences have been infroduced. Such host cells can then be used to create non-human transgenic animals in which exogenous NOVX sequences have been introduced into their genome or homologous recombinant animals in which endogenous NOVX sequences have been altered. Such animals are useful for studying the function and/or activity of NOVX protein and for identifying and/or evaluating modulators of NOVX protein activity. As used herein, a "transgenic animal" is a non-human animal, preferably a mammal, more preferably a rodent such as a rat or mouse, in which one or more of the cells of the animal includes a transgene. Other examples of transgenic animals include non-human primates, sheep, dogs, cows, goats, chickens, amphibians, etc. A transgene is exogenous DNA that is integrated into the genome of a cell from which a transgenic animal develops and that remains in the genome of the mature animal, thereby directing the expression of an encoded gene product in one or more cell types or tissues of the transgenic animal. As used herein, a "homologous recombinant animal" is a non-human animal, preferably a mammal, more preferably a mouse, in which an endogenous NOVX gene has been altered by homologous recombination between the endogenous gene and an exogenous DNA molecule introduced into a cell of the animal, e.g., an embryonic cell of the animal, prior to development of the animal.
A transgenic animal of the invention can be created by introducing NOVX-encoding nucleic acid into the male pronuclei of a fertilized oocyte (e.g., by microinjection, retroviral infection) and allowing the oocyte to develop in a pseudopregnant female foster animal. The human NOVX cDNA sequences, i.e., any one of SEQ ID NOS:2n-l, wherein n is an integer between 1 and 141, can be introduced as a transgene into the genome of a non-human animal. Alternatively, a non-human homologue of the human NOVX gene, such as a mouse NOVX gene, can be isolated based on hybridization to the human NOVX cDNA (described further supra) and used as a transgene. Intronic sequences and polyadenylation signals can also be included in the transgene to increase the efficiency of expression of the fransgene. A tissue-specific regulatory sequence(s) can be operably-linked to the NOVX transgene to direct expression of NOVX protein to particular cells. Methods for generating transgenic animals via embryo manipulation and microinjection, particularly animals such as mice, have become conventional in the art and are described, for example, in U.S. Patent Nos. 4,736,866; 4,870,009; and 4,873,191; and Hogan, 1986. In: MANIPULATING THE MOUSE EMBRYO, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. Similar methods are used for production of other transgenic animals. A transgenic founder animal can be identified based upon the presence of the NOVX fransgene in its genome and/or expression of NOVX mRNA in tissues or cells of the animals. A transgenic founder animal can then be used to breed additional animals carrying the transgene. Moreover, transgenic animals carrying a transgene-encoding NOVX protein can further be bred to other transgenic animals carrying other transgenes.
To create a homologous recombinant animal, a vector is prepared which contains at least a portion of a NOVX gene into which a deletion, addition or substitution has been infroduced to thereby alter, e.g., functionally disrupt, the NOVX gene. The NOVX gene can be a human gene (e.g., the cDNA of any one of SEQ ID NOS:2n-l , wherein n is an integer between 1 and 141), but more preferably, is a non-human homologue of a human NOVX gene. For example, a mouse homologue of human NOVX gene of SEQ ID NOS:2n-l , wherein n is an integer between 1 and 141 , can be used to construct a homologous recombination vector suitable for altering an endogenous NOVX gene in the mouse genome. In one embodiment, the vector is designed such that, upon homologous recombination, the endogenous NOVX gene is functionally disrupted (i.e., no longer encodes a functional protein; also referred to as a "knock out" vector).
Alternatively, the vector can be designed such that, upon homologous recombination, the endogenous NOVX gene is mutated or otherwise altered but still encodes functional protein (e.g., the upstream regulatory region can be altered to thereby alter the expression of the endogenous NOVX protein). In the homologous recombination vector, the altered portion of the NOVX gene is flanked at its 5'- and 3'-termini by additional nucleic acid of the NOVX gene to allow for homologous recombination to occur between the exogenous NOVX gene carried by the vector and an endogenous NOVX gene in an embryonic stem cell. The additional flanking NOVX nucleic acid is of sufficient length for successful homologous recombination with the endogenous gene. Typically, several kilobases of flanking DNA (both at the 5'- and 3'-termini) are included in the vector. See, e.g., Thomas, et al, 1987. Cell 51 : 503 for a description of homologous recombination vectors. The vector is ten introduced into an embryonic stem cell line (e.g., by elecfroporation) and cells in which the infroduced NOVX gene has homologously-recombined with the endogenous NOVX gene are selected. See, e.g., Li, et al, 1992. Cell 69: 915. The selected cells_are then injected into a blastocyst of an animal (e.g., a mouse) to form aggregation chimeras. See, e.g., Bradley, 1987. In: TERATOCARCINOMAS AND EMBRYONIC STEM CELLS: A PRACTICAL APPROACH, Robertson, ed. IRL, Oxford, pp. 113-152. A chimeric embryo can then be implanted into a suitable pseudopregnant female foster animal and the embryo brought to term. Progeny harboring the homologously-recombined DNA in their germ cells can be used to breed animals in which all cells of the animal contain the homologously-recombined DNA by germline transmission of the transgene. Methods for constructing homologous recombination vectors and homologous recombinant animals are described further in Bradley, 1991. Curr. Opin. Biotechnol. 2: 823-829; PCT International Publication Nos.: WO 90/11354; WO 91/01140; WO 92/0968; and WO 93/04169.
In another embodiment, transgenic non-humans animals can be produced that contain selected systems that allow for regulated expression of the transgene. One example of such a system is the cre/loxP recombinase system of bacteriophage PI. For a description of the cre/loxP recombinase system, See, e.g, Lakso, et al, 1992. Proc. Natl. Acad. Sci. USA 89: 6232-6236. Another example of a recombinase system is the FLP recombinase system of Saccharomyces cerevisiae. See, O'Gorman, et al, 1991. Science 251:1351-1355. If a cre/loxP recombinase system is used to regulate expression of the transgene, animals containing transgenes encoding both the Cre recombinase and a selected protein are required. Such animals can be provided through the construction of "double" transgenic animals, e.g., by mating two transgenic animals, one containing a transgene lencoding a selected protein and the other containing a transgene encoding a recombinase. Clones of the non-human transgenic animals described herein can also be produced according to the methods described in Wilmut, et al, 1997. Nature 385: 810-813. In brief, a cell (e.g., a somatic cell) from the transgenic animal can be isolated and induced to exit the growth cycle and enter Go phase. The quiescent cell can then be fused, e.g., through the use of electrical pulses, to an enucleated oocyte from an animal of the same species from which the quiescent cell is isolated. The reconstructed oocyte is then cultured such that it develops to morula or blastocyte and then transferred to pseudopregnant female foster animal. The offspring borne of this female foster animal will be a clone of the animal from which the cell (e.g., the somatic cell) is isolated. Pharmaceutical Compositions
The NOVX nucleic acid molecules, NOVX proteins, and anti-NOVX antibodies (also refened to herein as "active compounds") of the invention, and derivatives, fragments, analogs and homologs thereof, can be incoφorated into pharmaceutical compositions suitable for administration. Such compositions typically comprise the nucleic acid molecule, protein, or antibody and a pharmaceutically acceptable carrier. As used herein, "pharmaceutically acceptable carrier" is intended to include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absoφtion delaying agents, and the like, compatible with pharmaceutical administration. Suitable caniers are described in the most recent edition of Remington's Pharmaceutical Sciences, a standard reference text in the field, which is incoφorated herein by reference. Prefened examples of such carriers or diluents include, but are not limited to, water, saline, finger's solutions, dextrose solution, and 5% human serum albumin. Liposomes and non-aqueous vehicles such as fixed oils may also be used. The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active compound, use thereof in the compositions is contemplated. Supplementary active compounds can also be incoφorated into the compositions.
A pharmaceutical composition of the invention is formulated to be compatible with its intended route of administration. Examples of routes of administration include parenteral, e.g., intravenous, intradermal, subcutaneous, oral (e.g., inhalation), transdermal (i.e., topical), transmucosal, and rectal administration. Solutions or suspensions used for parenteral, intradermal, or subcutaneous application can include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetefraacetic acid (EDTA); buffers such as acetates, citrates or phosphates, and agents for the adjustment of tonicity such as sodium chloride or dextrose. The pH can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide. The parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic.
Pharmaceutical compositions suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion. For intravenous administration, suitable carriers include physiological saline, bacteriostatic water, Cremophor EL (BASF, Parsippany, N. J.) or phosphate buffered saline (PBS). In all cases, the composition must be sterile and should be fluid to the extent that easy syringeability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), and suitable mixtures thereof. The proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. Prevention of the action of microorganisms can be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars, polyalcohols such as manitol, sorbitol, sodium chloride in the composition. Prolonged absoφtion of the injectable compositions can be brought about by including in the composition an agent which delays absoφtion, for example, aluminum monostearate and gelatin.
Sterile injectable solutions can be prepared by incoφorating the active compound (e.g., a NOVX protein or anti-NOVX antibody) in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incoφorating the active compound into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from thoseenumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, methods of preparation are vacuum drying and freeze-drying that yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
Oral compositions generally include an inert diluent or an edible carrier. They can be enclosed in gelatin capsules or compressed into tablets. For the puφose of oral therapeutic administration, the active compound can be incoφorated with excipients and used in the form of tablets, troches, or capsules. Oral compositions can also be prepared using a fluid carrier for use as a mouthwash, wherein the compound in the fluid carrier is applied orally and swished and expectorated or swallowed. Pharmaceutically compatible binding agents, and/or adjuvant materials can be included as part of the composition. The tablets, pills, capsules, troches and the like can contain any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum teagacanth or gelatin; an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or corn starch; a lubricant such as magnesium stearate or Sterotes; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring.
For administration by inhalation, the compounds are delivered in the form of an aerosol spray from pressured container or dispenser which contains a suitable propellant, e.g., a gas such as carbon dioxide, or a nebulizer. Systemic administration can also be by transmucosal or teansdermal means. For transmucosal or fransdermal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art, and include, for example, for transmucosal administration, detergents, bile salts, and fusidic acid derivatives. Transmucosal administration can be accomplished through the use of nasal sprays or suppositories. For fransdermal administration, the active compounds are formulated into ointments, salves, gels, or creams as generally known in the art.
The compounds can also be prepared in the form of suppositories (e.g., with conventional suppository bases such as cocoa butter and other glycerides) or retention enemas for rectal delivery.
In one embodiment, the active compounds are prepared with carriers that will protect the compound against rapid elimination from the body, such as a controlled release formulation, including implants and microencapsulated delivery systems. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Methods for preparation of such formulations will be apparent to those skilled in the art. The materials can also be obtained commercially from Alza Coφoration and Nova Pharmaceuticals, Inc. Liposomal suspensions (including liposomes targeted to infected cells with monoclonal antibodies to viral antigens) can also be used as pharmaceutically acceptable carriers. These can be prepared according to methods known to those skilled in the art, for example, as described in U.S. Patent No. 4,522,811.
It is especially advantageous to formulate oral or parenteral compositions in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the subject to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The specification for the dosage unit forms of the invention are dictated by and directly dependent on the unique characteristics of the active compound and the particular therapeutic effect to be achieved, and the limitations inherent in the art of compounding such an active compound for the treatment of individuals.
The nucleic acid molecules of the invention can be inserted into vectors and used as gene therapy vectors. Gene therapy vectors can be delivered to a subject by, for example, intravenous injection, local administration (see, e.g., U.S. Patent No. 5,328,470) or by stereotactic injection (see, e.g., Chen, et al, 1994. Proc. Natl. Acad. Sci. USA 91: 3054-3057). The pharmaceutical preparation of the gene therapy vector can include the gene therapy vector in an acceptable diluent, or can comprise a slow release matrix in which the gene delivery vehicle is imbedded. Alternatively, where the complete gene delivery vector can be produced intact from recombinant cells, e.g., retroviral vectors, the pharmaceutical preparation can include one or more cells that produce the gene delivery system.
The pharmaceutical compositions can be included in a container, pack, or dispenser together with instructions for administration. Screening and Detection Methods
The isolated nucleic acid molecules of the invention can be used to express NOVX protein (e.g., via a recombinant expression vector in a host cell in gene therapy applications), to detect NOVX mRNA (e.g., in a biological sample) or a genetic lesion in a NOVX gene, and to modulate NOVX activity, as described further, below. In addition, the NOVX proteins can be used to screen drags or compounds that modulate the NOVX protein activity or expression as well as to treat disorders characterized by insufficient or excessive production of NOVX protein or production of NOVX protein forms that have decreased or abenant activity compared to NOVX wild-type protein (e.g.; diabetes (regulates insulin release); obesity (binds and transport lipids); metabolic disturbances associated with obesity, the metabolic syndrome X as well as anorexia and wasting disorders associated with chronic diseases and various cancers, and infectious disease(possesses anti-microbial activity) and the various dyslipidemias. In addition, the anti-NOVX antibodies of the invention can be used to detect and isolate NOVX proteins and modulate NOVX activity. In yet a further aspect, the invention can be used in methods to influence appetite, absoφtion of nutrients and the disposition of metabolic substrates in both a positive and negative fashion. The invention further pertains to novel agents identified by the screening assays described herein and uses thereof for treatments as described, supra.
Screening Assays
The invention provides a method (also refened to herein as a "screening assay") for identifying modulators, i.e., candidate or test compounds or agents (e.g., peptides, peptidomimetics, small molecules or other drags) that bind to NOVX proteins or have a stimulatory or inhibitory effect on, e.g., NOVX protein expression or NOVX protein activity. The invention also includes compounds identified in the screening assays described herein.
In one embodiment, the invention provides assays for screening candidate or test compounds which bind to or modulate the activity of the membrane-bound form of a
NOVX protein or polypeptide or biologically-active portion thereof. The test compounds of the invention can be obtained using any of the numerous approaches in combinatorial library methods known in the art, including: biological libraries; spatially addressable parallel solid phase or solution phase libraries; synthetic library methods requiring deconvolution; the "one-bead one-compound" library method; and synthetic library methods using affinity chromatography selection. The biological library approach is limited to peptide libraries, while the other four approaches are applicable to peptide, non-peptide oligomer or small molecule libraries of compounds. See, e.g., Lam, 1997. Anticancer Drug Design 12: 145.
A "small molecule" as used herein, is meant to refer to a composition that has a molecular weight of less than about 5 kD and most preferably less than about 4 kD. Small molecules can be, e.g., nucleic acids, peptides, polypeptides, peptidomimetics, carbohydrates, lipids or other organic or inorganic molecules. Libraries of chemical and/or biological mixtures, such as fungal, bacterial, or algal extracts, are known in the art and can be screened with any of the assays of the invention.
Examples of methods for the synthesis of molecular libraries can be found in the art, for example in: DeWitt, et al, 1993. Proc. Natl. Acad. Sci. U.S.A. 90: 6909; Erb, et al, 1994. Proc. Natl. Acad. Sci. U.S.A. 91 : 11422; Zuckermann, et al, 1994. J. Med. Chem. 37: 2678; Cho, et al, 1993. Science 261: 1303; Canell, et al, 1994. Angew. Chem. Int. Ed. Engl. 33: 2059; Carell, et al, 1994. Angew. Chem. Int. Ed. Engl 33: 2061; and Gallop, et al, 1994. J. Med. Chem. 37: 1233. Libraries of compounds may be presented in solution (e.g. , Houghten, 1992.
Biotechniques 13: 412-421), or on beads (Lam, 1991. Nature 354: 82-84), on chips (Fodor, 1993. Nature 364: 555-556), bacteria (Ladner, U.S. Patent No. 5,223,409), spores (Ladner, U.S. Patent 5,233,409), plasmids (Cull, et al, 1992. Proc. Natl. Acad. Sci. USA 89: 1865-1869) or on phage (Scott and Smith, 1990. Science 249: 386-390; Devlin, 1990. Science 249: 404-406; Cwirla, et al, 1990. Proc. Natl. Acad. Sci. U.S.A. 87: 6378-6382; Felici, 1991. J. Mol. Biol 222: 301-310; Ladner, U.S. Patent No. 5,233,409.).
In one embodiment, an assay is a cell-based assay in which a cell which expresses a membrane-bound form of NOVX protein, or a biologically-active portion thereof, on the cell surface is contacted with a test compound and the ability of the test compound to bind to a NOVX protein determined. The cell, for example, can of mammalian origin or a yeast cell. Determining the ability of the test compound to bind to the NOVX protein can be accomplished, for example, by coupling the test compound with a radioisotope or enzymatic label such that binding of the test compound to the NOVX protein or biologically-active portion thereof can be determined by detecting the labeled compound in a complex. For example, test compounds can be labeled with 1251, 35S, 14C, or 3H, either directly or indirectly, and the radioisotope detected by direct counting of radioemission or by scintillation counting. Alternatively, test compounds can be enzymatically-labeled with, for example, horseradish peroxidase, alkaline phosphatase, or luciferase, and the enzymatic label detected by determination of conversion of an appropriate substrate to product. In one embodiment, the assay comprises contacting a cell which expresses a membrane-bound form of NOVX protein, or a biologically-active portion thereof, on the cell surface with a known compound which binds NOVX to form an assay mixture, contacting the assay mixture with a test compound, and determining the ability of the test compound to interact with a NOVX protein, wherein determining the ability of the test compound to interact with a NOVX protein comprises determining the ability of the test compound to preferentially bind to NOVX protein or a biologically-active portion thereof as compared to the known compound. In another embodiment, an assay is a cell-based assay comprising contacting a cell expressing a membrane-bound form of NOVX protein, or a biologically-active portion thereof, on the cell surface with a test compound and determining the ability of the test compound to modulate (e.g., stimulate or inhibit) the activity of the NOVX protein or biologically-active portion thereof. Determining the ability of the test compound to modulate the activity of NOVX or a biologically-active portion thereof can be accomplished, for example, by determining the ability of the NOVX protein to bind to or interact with a NOVX target molecule. As used herein, a "target molecule" is a molecule with which a NOVX protein binds or interacts in nature, for example, a molecule on the surface of a cell which expresses a NOVX interacting protein, a molecule on the surface of a second cell, a molecule in the extracellular milieu, a molecule associated with the internal surface of a cell membrane or a cytoplasmic molecule, a NOVX target molecule can be a non-NOVX molecule or a NOVX protein or polypeptide of the invention. In one embodiment, a NOVX target molecule is a component of a signal transduction pathway that facilitates transduction of an extracellular signal (e.g. a signal generated by binding of a compound to a membrane-bound NOVX molecule) through the cell membrane and into the cell. The target, for example, can be a second intercellular protein that has catalytic activity or a protein that facilitates the association of downstream signaling molecules with NOVX.
Determining the ability of the NOVX protein to bind to or interact with a NOVX target molecule can be accomplished by one of the methods described above for determining direct binding. In one embodiment, determining the ability of the NOVX protein to bind to or interact with a NOVX target molecule can be accomplished by determiningJhe activity of the target molecule. For example, the activity of the target molecule can be determined by detecting induction of a cellular second messenger of the target (i.e. intracellular Ca2+, diacylglycerol, IP3, etc.), detecting catalytic/enzymatic activity of the target an appropriate substrate, detecting the induction of a reporter gene (comprising a NOVX-responsive regulatory element operatively linked to a nucleic acid encoding a detectable marker, e.g., luciferase), or detecting a cellular response, for example, cell survival, cellular differentiation, or cell proliferation.
In yet another embodiment, an assay of the invention is a cell-free assay comprising contacting a NOVX protein or biologically-active portion thereof with a test compound and determining the ability of the test compound to bind to the NOVX protein or biologically-active portion thereof. Binding of the test compound to the NOVX protein can be determined either directly or indirectly as described above. In one such embodiment, the assay comprises contacting the NOVX protein or biologically-active portion thereof with a known compound which binds NOVX to form an assay mixture, contacting the assay mixture with a test compound, and determining the ability of the test compound to interact with a NOVX protein, wherein determining the ability of the test compound to interact with a NOVX protein comprises determining the ability of the test compound to preferentially bind to NOVX or biologically-active portion thereof as compared to the known compound.
In still another embodiment, an assay is a cell-free assay comprising contacting NOVX protein or biologically-active portion thereof with a test compound and determining the ability of the test compound to modulate (e.g. stimulate or inhibit) the activity of the NOVX protein or biologically-active portion thereof. Determining the ability of the test compound to modulate the activity of NOVX can be accomplished, for example, by determining the ability of the NOVX protein to bind to a NOVX target molecule by one of the methods described above for determining direct binding. In an alternative embodiment, determining the ability of the test compound to modulate the activity of NOVX protein can be accomplished by determining the ability of the NOVX protein further modulate a NOVX target molecule. For example, the catalytic/enzymatic activity of the target molecule on an appropriate substrate can be determined as described, supra.
In yet another embodiment, the cell-free assay comprises contacting the NOVX protein or biologically-active portion thereof with a known compound which binds NOVX protein to form an assay mixture, contacting the assay mixture with a test compound, and determining the ability of the test compound to interact with a NOVX protein, wherein determining the ability of the test compound to interact with a NOVX protein comprises determining the ability of the NOVX protein to preferentially bind to or modulate the activity of a NOVX target molecule. The cell-free assays of the invention are amenable to use of both the soluble form or the membrane-bound form of NOVX protein. In the case of cell-free assays comprising the membrane-bound form of NOVX protein, it may be desirable to utilize a solubilizing agent such that the membrane-bound form of NOVX protein is maintained in solution. Examples of such solubilizing agents include non-ionic detergents such as n-octylglucoside, n-dodecylglucoside, n-dodecylmaltoside, octanoyl-N-methylglucamide, decanoyl-N-methylglucamide, Triton® X-100, Triton® X-l 14, Thesit®, Isotridecypoly(ethylene glycol ether)n, N-dodecyl~N,N-dimethyl-3-ammonio-l -propane sulfonate, 3-(3-cholamidopropyl) dimethylamminiol-1 -propane sulfonate (CHAPS), or 3-(3-cholamidopropyl)dimethylammimol-2-hydroxy-l-propane sulfonate (CHAPSO). In more than one embodiment of the above assay methods of the invention, it may be desirable to immobilize either NOVX protein or its target molecule to facilitate separation of complexed from uncomplexed forms of one or both of the proteins, as well as to accommodate automation of the assay. Binding of a test compound to NOVX protein, or interaction of NOVX protein with a target molecule in the presence and absence of a candidate compound, can be accomplished in any vessel suitable for containing the reactants. Examples of such vessels include microtiter plates, test tubes, and micro-centrifuge tubes. In one embodiment, a fusion protein can be provided that adds a domain that allows one or both of the proteins to be bound to a matrix. For example, GST-NO VX fusion proteins or GST-target fusion proteins can be adsorbed onto glutathione sepharose beads (Sigma Chemical, St. Louis, MO) or glutathione derivatized microtiter plates, that are then combined with the test compound or the test compound and either the non-adsorbed target protein or NOVX protein, and the mixture is incubated under conditions conducive to complex formation (e.g., at physiological conditions for salt and pH). Following incubation, the beads or microtiter plate wells are washed to remove any unbound components, the matrix immobilized in the case of beads, complex determined either directly or indirectly, for example, as described, supra. Alternatively, the complexes can be dissociated from the matrix, and the level of NOVX protein binding or activity determined using standard techniques. Other techniques for immobilizing proteins on matrices can also be used in the screening assays of the invention. For example, either the NOVX protein or its target molecule can be immobilized utilizing conjugation of biotin and streptavidin. Biotinylated NOVX protein or target molecules can be prepared from biotin-NHS (N-hydroxy-succinimide) using techniques well-known within the art (e.g., biotinylation kit, Pierce Chemicals, Rockford, 111.), and immobilized in the wells of streptavidin-coated 96 well plates (Pierce Chemical). Alternatively, antibodies reactive with NOVX protein or target molecules, but which do not interfere with binding of the NOVX protein to its target molecule, can be derivatized to the wells of the plate, and unbound target or NOVX protein trapped in the wells by antibody conjugation. Methods for detecting such complexes, in addition to those described above for the GST-immobilized complexes, include immunodetection of complexes using antibodies reactive with the NOVX protein or target molecule, as well as enzyme-linked assays that rely on detecting an enzymatic activity associated with the NOVX protein or target molecule. In another embodiment, modulators of NOVX protein expression are identified in a method wherein a cell is contacted with a candidate compound and the expression of NOVX mRNA or protein in the cell is determined. The level of expression of NOVX mRNA or protein in the presence of the candidate compound is compared to the level of expression of NOVX mRNA or protein in the absence of the candidate compound. The candidate compound can then be identified as a modulator of NOVX mRNA or protein expression based upon this comparison. For example, when expression of NOVX mRNA or protein is greater (i.e., statistically significantly greater) in the presence of the candidate compound than in its absence, the candidate compound is identified as a stimulator of NOVX mRNA or protein expression. Alternatively, when expression of NOVX mRNA or protein is less (statistically significantly less) in the presence of the candidate compound than in its absence, the candidate compound is identified as an inhibitor of NOVX mRNA or protein expression. The level of NOVX mRNA or protein expression in the cells can be determined by methods described herein for detecting NOVX mRNA or protein.
In yet another aspect of the invention, the NOVX proteins can be used as "bait proteins" in a two-hybrid assay or three hybrid assay (see, e.g., U.S. Patent No. 5,283,317; Zervos, et al, 1993. Cell 72: 223-232; Madura, et al, 1993. J. Biol. Chem. 268: 12046-12054; Bartel, etal, 1993. Biotechniques 14: 920-924; Iwabuchi, etal, 1993. Oncogene 8: 1693-1696; and Brent WO 94/10300), to identify other proteins that bind to or interact with NOVX ("NOVX-binding proteins" or "NOVX-bp") and modulate NOVX activity. Such NOVX-binding proteins are also likely to be involved in the propagation of signals by the NOVX proteins as, for example, upstream or downstream elements of the NOVX pathway. The two-hybrid system is based on the modular nature of most transcription factors, which consist of separable DNA-binding and activation domains. Briefly, the assay utilizes two different DNA constructs. In one construct, the gene that codes for NOVX is fused to a gene encoding the DNA binding domain of a known transcription factor (e.g., GAL-4). In the other construct, a DNA sequence, from a library of DNA sequences, that encodes an unidentified protein ("prey" or "sample") is fused to a gene that codes for the activation domain of the known transcription factor. If the "bait" and the "prey" proteins are able to interact, in vivo, forming a NOVX-dependent complex, the DNA-binding and activation domains of the transcription factor are brought into close proximity. This proximity allows transcription of a reporter gene (e.g., LacZ) that is operably linked to a transcriptional regulatory site responsive to the transcription factor. Expression of the reporter gene can be detected and cell colonies containing the functional transcription factor can be isolated and used to obtain the cloned gene that encodes the protein which interacts with NOVX.
The invention further pertains to novel agents identified by the aforementioned screening assays and uses thereof for treatments as described herein.
Detection Assays
Portions or fragments of the cDNA sequences identified herein (and the corresponding complete gene sequences) can be used in numerous ways as polynucleotide reagents. By way of example, and not of limitation, these sequences can be used to: (i) map their respective genes on a chromosome; and, thus, locate gene regions associated with genetic disease; (ii) identify an individual from a minute biological sample (tissue typing); and (iii) aid in forensic identification of a biological sample. Some of these applications are described in the subsections, below.
Chromosome Mapping Once the sequence (or a portion of the sequence) of a gene has been isolated, this sequence can be used to map the location of the gene on a chromosome. This process is called chromosome mapping. Accordingly, portions or fragments of a NOVX sequence, i.e., of SEQ ID NOS :2n-l, wherein n is an integer between 1 and 141, or fragments or derivatives thereof, can be used to map the location of the NOVX genes, respectively, on a chromosome. The mapping of the NOVX sequences to chromosomes is an important first step in conelating these sequences with genes associated with disease. Briefly, NOVX genes can be mapped to chromosomes by preparing PCR primers
(preferably 15-25 bp in length) from the NOVX sequences. Computer analysis of the NOVX, sequences can be used to rapidly select primers that do not span more than one exon in the genomic DNA, thus complicating the amplification process. These primers can then be used for PCR screening of somatic cell hybrids containing individual human chromosomes. Only those hybrids containing the human gene conesponding to the NOVX sequences will yield an amplified fragment.
Somatic cell hybrids are prepared by fusing somatic cells from different mammals (e.g., human and mouse cells). As hybrids of human and mouse cells grow and divide, they gradually lose human chromosomes in random order, but retain the mouse chromosomes. By using media in which mouse cells cannot grow, because they lack a particular enzyme, but in which human cells can, the one human chromosome that contains the gene encoding the needed enzyme will be retained. By using various media, panels of hybrid cell lines can be established. Each cell line in a panel contains either a single human chromosome or a small number of human chromosomes, and a full set of mouse chromosomes, allowing easy mapping of individual genes to specific human chromosomes. See, e.g., D'Eustachio, et al, 1983. Science 220: 919-924. Somatic cell hybrids containing only fragments of human chromosomes can also be produced by using human chromosomes with translocations and deletions.
PCR mapping of somatic cell hybrids is a rapid procedure for assigning a particular sequence to a particular chromosome. Three or more sequences can be assigned per day using a single thermal cycler. Using the NOVX sequences to design oligonucleotide primers, sub-localization can be achieved with panels of fragments from specific chromosomes.
Fluorescence in situ hybridization (FISH) of a DNA sequence to a metaphase chromosomal spread can further be used to provide a precise chromosomal location in one step. Chromosome spreads can be made using cells whose division has been blocked in metaphase by a chemical like colcemid that disrupts the mitotic spindle. The chromosomes can be treated briefly with trypsin, and then stained with Giemsa. A pattern of light and dark bands develops on each chromosome, so that the chromosomes can be identified individually. The FISH technique can be used with a DNA sequence as short as 500 or 600 bases. However, clones larger than 1,000 bases have a higher likelihood of binding to a unique chromosomal location with sufficient signal intensity for simple detection. Preferably 1 ,000 bases, and more preferably 2,000 bases, will suffice to get good results at a reasonable amount of time. For a review of this technique, see, Verma, et al, HUMAN CHROMOSOMES: A MANUAL OF BASIC TECHNIQUES (Pergamon Press, New York 1988).
Reagents for chromosome mapping can be used individually to mark a single chromosome or a single site on that chromosome, or panels of reagents can be used for marking multiple sites and/or multiple chromosomes. Reagents conesponding to noncoding regions of the genes actually are prefened for mapping puφoses. Coding sequences are more likely to be conserved within gene families, thus increasing the chance of cross hybridizations during chromosomal mapping. Once a sequence has been mapped to a precise chromosomal location, the physical position of the sequence on the chromosome can be conelated with genetic map data. Such data are found, e.g., in McKusick, MENDELIAN INHERITANCE IN MAN, available on-line through Johns Hopkins University Welch Medical Library). The relationship between genes and disease, mapped to the same chromosomal region, can then be identified through linkage analysis (co-inheritance of physically adjacent genes), described in, e.g., Egeland, et al, 1987. Nature, 325: 783-787.
Moreover, differences in the DNA sequences between individuals affected and unaffected with a disease associated with the NOVX gene, can be determined. If a mutation is observed in some or all of the affected individuals but not in any unaffected individuals, then the mutation is likely to be the causative agent of the particular disease. Comparison of affected and unaffected individuals generally involves first looking for structural alterations in the chromosomes, such as deletions or translocations that are visible from chromosome spreads or detectable using PCR based on that DNA sequence. Ultimately, complete sequencing of genes from several individuals can be performed to confirm the presence of a mutation and to distinguish mutations from polymoφhisms. Tissue Typing
The NOVX sequences of the invention can also be used to identify individuals from minute biological samples. In this technique, an individual's genomic DNA is digested with one or more restriction enzymes, and probed on a Southern blot to yield unique bands for identification. The sequences of the invention are useful as additional DNA markers for RFLP ("restriction fragment length polymoφhisms," described in U.S. Patent No. 5,272,057).
Furthermore, the sequences of the invention can be used to provide an alternative technique that determines the actual base-by-base DNA sequence of selected portions of an individual's genome. Thus, the NOVX sequences described herein can be used to prepare two PCR primers from the 5'- and 3'-termini of the sequences. These primers can then be used to amplify an individual's DNA and subsequently sequence it.
Panels of conesponding DNA sequences from individuals, prepared in this manner, can provide unique individual identifications, as each individual will have a unique set of such DNA sequences due to allelic differences. The sequences of the invention can be used to obtain such identification sequences from individuals and from tissue. The NOVX sequences of the invention uniquely represent portions of the human genome. Allelic variation occurs to some degree in the coding regions of these sequences, and to a greater degree in the noncoding regions. It is estimated that allelic variation between individual humans occurs with a frequency of about once per each 500 bases. Much of the allelic variation is due to single nucleotide polymoφhisms (SNPs), which include restriction fragment length polymoφhisms (RFLPs).
Each of the sequences described herein can, to some degree, be used as a standard against which DNA from an individual can be compared for identification puφoses. Because greater numbers of polymoφhisms occur in the noncoding regions, fewer sequences are necessary to differentiate individuals. The noncoding sequences can comfortably provide positive individual identification with a panel of perhaps 10 to 1,000 primers that each yield a noncoding amplified sequence of 100 bases. If coding sequences, such as those of SEQ ID NOS:2n-l, wherein n is an integer between 1 and 141, are used, a more appropriate number of primers for positive individual identification would be 500-2,000. Predictive Medicine
The invention also pertains to the field of predictive medicine in which diagnostic assays, prognostic assays, pharmacogenomics, and monitoring clinical trials are used for prognostic (predictive) puφoses to thereby treat an individual prophylactically. Accordingly, one aspect of the invention relates to diagnostic assays for determining
NOVX protein and/or nucleic acid expression as well as NOVX activity, in the context of a biological sample (e.g., blood, serum, cells, tissue) to thereby determine whether an individual is afflicted with a disease or disorder, or is at risk of developing a disorder, associated with abenant NOVX expression or activity. The disorders include metabolic disorders, diabetes, obesity, infectious disease, anorexia, cancer-associated cachexia, cancer, neurodegenerative disorders, Alzheimer's Disease, Parkinson's Disorder, immune disorders, and hematopoietic disorders, and the various dyslipidemias, metabolic disturbances associated with obesity, the metabolic syndrome X and wasting disorders associated with chronic diseases and various cancers. The invention also provides for prognostic (or predictive) assays for determining whether an individual is at risk of developing a disorder associated with NOVX protein, nucleic acid expression or activity. For example, mutations in a NOVX gene can be assayed in a biological sample. Such assays can be used for prognostic or predictive puφose to thereby prophylactically treat an individual prior to the onset of a disorder characterized by or associated with NOVX protein, nucleic acid expression, or biological activity.
Another aspect of the invention provides methods for determining NOVX protein, nucleic acid expression or activity in an individual to thereby select appropriate therapeutic or prophylactic agents for that individual (refened to herein as "pharmacogenomics"). Pharmacogenomics allows for the selection of agents (e.g., drugs) for therapeutic or prophylactic freatment of an individual based on the genotype of the individual (e.g., the genotype of the individual examined to determine the ability of the individual to respond to a particular agent.)
Yet another aspect of the invention pertains to monitoring the influence of agents (e.g., drugs, compounds) on the expression or activity of NOVX in clinical trials. These and other agents are described in further detail in the following sections. Diagnostic Assays
An exemplary method for detecting the presence or absence of NOVX in a biological sample involves obtaining a biological sample from a test subject and contacting the biological sample with a compound or an agent capable of detecting NOVX protein or nucleic acid (e.g., mRNA, genomic DNA) that encodes NOVX protein such that the presence of NOVX is detected in the biological sample. An agent for detecting NOVX mRNA or genomic DNA is a labeled nucleic acid probe capable of hybridizing to NOVX mRNA or genomic DNA. The nucleic acid probe can be, for example, a full-length NOVX nucleic acid, such as the nucleic acid of SEQ ID NOS:2n-l, wherein n is an integer between 1 and 141, or a portion thereof, such as an oligonucleotide of at least 15, 30, 50, 100, 250 or 500 nucleotides in length and sufficient to specifically hybridize under stringent conditions to NOVX mRNA or genomic DNA. Other suitable probes for use in the diagnostic assays of the invention are described herein.
An agent for detecting NOVX protein is an antibody capable of binding to NOVX protein, preferably an antibody with a detectable label. Antibodies can be polyclonal, or more preferably, monoclonal. An intact antibody, or a fragment thereof (e.g., Fab or F(ab')2) can be used. The term "labeled", with regard to the probe or antibody, is intended to encompass direct labeling of the probe or antibody by coupling (i.e., physically linking) a detectable substance to the probe or antibody, as well as indirect labeling of the probe or antibody by reactivity with another reagent that is directly labeled. Examples of indirect labeling include detection of a primary antibody using a fluorescently-labeled secondary antibody and end-labeling of a DNA probe with biotin such that it can be detected with fluorescently-labeled streptavidin. The term "biological sample" is intended to include tissues, cells and biological fluids isolated from a subject, as well as tissues, cells and fluids present within a subject.. That is, the detection method of the invention can be used to detect NOVX mRNA, protein, or genomic DNA in a biological sample in vitro as well as in vivo. For example, in vitro techniques for detection of NOVX mRNA include Northern hybridizations and in situ hybridizations. In vitro techniques for detection of NOVX protein include enzyme linked immunosorbent assays (ELISAs), Western blots, immunoprecipitations, and immunofluorescence. In vitro techniques for detection of NOVX genomic DNA include Southern hybridizations. Furthermore, in vivo techniques for detection of NOVX protein include introducing into a subject a labeled anti-NOVX antibody. For example, the antibody can be labeled with a radioactive marker whose presence and location in a subject can be detected by standard imaging techniques.
In one embodiment, the biological sample contains protein molecules from the test subject. Alternatively, the biological sample can contain mRNA molecules from the test subject or genomic DNA molecules from the test subject. A prefened biological sample is a peripheral blood leukocyte sample isolated by conventional means from a subject.
In another embodiment, the methods further involve obtaining a control biological sample from a confrol subject, contacting the control sample with a compound or agent capable of detecting NOVX protein, mRNA, or genomic DNA, such that the presence of NOVX protein, mRNA or genomic DNA is detected in the biological sample, and comparing the presence of NOVX protein, mRNA or genomic DNA in the control sample with the presence of NOVX protein, mRNA or genomic DNA in the test sample.
The invention also encompasses kits for detecting the presence of NOVX in a biological sample. For example, the kit can comprise: a labeled compound or agent capable of detecting NOVX protein or mRNA in a biological sample; means for determining the amount of NOVX in the sample; and means for comparing the amount of NOVX in the sample with a standard. The compound or agent can be packaged in a suitable container. The kit can further comprise instructions for using the kit to detect NOVX protein or nucleic acid. Prognostic Assays
The diagnostic methods described herein can furthermore be utilized to identify subjects having or at risk of developing a disease or disorder associated with abenant NOVX expression or activity. For example, the assays described herein, such as the preceding diagnostic assays or the following assays, can be utilized to identify a subject having or at risk of developing a disorder associated with NOVX protein, nucleic acid expression or activity. Alternatively, the prognostic assays can be utilized to identify a subject having or at risk for developing a disease or disorder. Thus, the invention provides a method for identifying a disease or disorder associated with abenant NOVX expression or activity in which a test sample is obtained from a subject and NOVX protein or nucleic acid (e.g., mRNA, genomic DNA) is detected, wherein the presence of NOVX protein or nucleic acid is diagnostic for a subject having or at risk of developing a disease or disorder associated with abenant NOVX expression or activity. As used herein, a "test sample" refers to a biological sample obtained from a subject of interest. For example, a test sample can be a biological fluid (e.g., serum), cell sample, or tissue.
Furthermore, the prognostic assays described herein can be used to determine whether a subject can be administered an agent (e.g., an agonist, antagonist, peptidomimetic, protein, peptide, nucleic acid, small molecule, or other drag candidate) to treat a disease or disorder associated with abenant NOVX expression or activity. For example, such methods can be used to determine whether a subject can be effectively treated with an agent for a disorder. Thus, the invention provides methods for determining whether a subject can be effectively treated with an agent for a disorder associated with abenant NOVX expression or activity in which a test sample is obtained and NOVX protein or nucleic acid is detected (e.g., wherein the presence of NOVX protein or nucleic acid is diagnostic for a subject that can be administered the agent to teeat a disorder associated with abenant NOVX expression or activity).
The methods of the invention can also be used to detect genetic lesions in a NOVX gene, thereby determining if a subject with the lesioned gene is at risk for a disorder characterized by abenant cell proliferation and/or differentiation. In various embodiments, the methods include detecting, in a sample of cells from the subject, the presence or absence of a genetic lesion characterized by at least one of an alteration affecting the integrity of a gene encoding a NOVX-protein, or the misexpression of the NOVX gene. For example, such genetic lesions can be detected by ascertaining the existence of at least one of: (i) a deletion of one or more nucleotides from a NOVX gene; (ii) an addition of one or more nucleotides to a NOVX gene; (iii) a substitution of one or more nucleotides of a NOVX gene, (fv) a chromosomal reanangement of a NOVX gene; (v) an alteration in the level of a messenger RNA transcript of a NOVX gene, (vi) abenant modification of a NOVX gene, such as of the methylation pattern of the genomic DNA, (vii) the presence of a non- wild-type splicing pattern of a messenger RNA transcript of a NOVX gene, (viii) a non-wild-type level of a NOVX protein, (ix) allelic loss of a NOVX gene, and (x) inappropriate post-translational modification of a NOVX protein. As described herein, there are a large number of assay techniques known in the art which can be used for detecting lesions in a NOVX gene. A prefened biological sample is a peripheral blood leukocyte sample isolated by conventional means from a subject. However, any biological sample containing nucleated cells may be used, including, for example, buccal mucosal cells. In certain embodiments, detection of the lesion involves the use of a probe/primer in a polymerase chain reaction (PCR) (see, e.g., U.S. Patent Nos. 4,683,195 and 4,683,202), such as anchor PCR or RACE PCR, or, alternatively, in a ligation chain reaction (LCR) (see, e.g., Landegran, et al, 1988. Science 241: 1077-1080; and Nakazawa, et al, 1994. Proc. Natl. Acad. Sci. USA 91 : 360-364), the latter of which can be particularly useful for detecting point mutations in the NOVX-gene (see, Abravaya, et al, 1995. Nucl. Acids Res. 23: 675-682). This method can include the steps of collecting a sample of cells from a patient, isolating nucleic acid (e.g., genomic, mRNA or both) from the cells of the sample, contacting the nucleic acid sample with one or more primers that specifically hybridize to a NOVX gene under conditions such that hybridization and amplification of the NOVX gene (if present) occurs, and detecting the presence or absence of an amplification product, or detecting the size of the amplification product and comparing the length to a control sample. It is anticipated that PCR and/or LCR may be desirable to use as a preliminary amplification step in conjunction with any of the techniques used for detecting mutations described herein.
Alternative amplification methods include: self sustained sequence replication (see, Guatelli, et al, 1990. Proc. Natl. Acad. Sci. USA 87: 1874-1878), transcriptional amplification system (see, Kwoh, et al, 1989. Proc. Natl. Acad. Sci. USA 86: 1173-1177); Qβ Replicase (see, Lizardi, et al, 1988. BioTechnology 6: 1197), or any other nucleic acid amplification method, followed by the detection of the amplified molecules using techniques well known to those of skill in the art. These detection schemes are especially useful for the detection of nucleic acid molecules if such molecules are present in very low numbers.
In an alternative embodiment, mutations in a NOVX gene from a sample cell can be identified by alterations in restriction enzyme cleavage patterns. For example, sample and control DNA is isolated, amplified (optionally), digested with one or more restriction endonucleases, and fragment length sizes are determined by gel electrophoresis and compared. Differences in fragment length sizes between sample and control DNA indicates mutations in the sample DNA. Moreover, the use of sequence specific ribozymes (see, e.g., U.S. Patent No. 5,493,531) can be used to score for the presence of specific mutations by development or loss of a ribozyme cleavage site.
In other embodiments, genetic mutations in NOVX can be identified by hybridizing sample and control nucleic acids, e.g., DNA or RNA to high-density anays containing hundreds or thousands of oligonucleotide probes. See, e.g., Cronin, et al, 1996. Human Mutation 7: 244-255; Kozal, et al, 1996. Nat. Med. 2: 753-759. For example, genetic mutations in NOVX can be identified in two-dimensional anays containing light-generated DNA probes as described in Cronin, et al, supra. Briefly, a first hybridization anay of probes can be used to scan through long sfretches of DNA in a sample and confrol to identify base changes between the sequences by making linear anays of sequential overlapping probes. This step allows the identification of point mutations. This is followed by a second hybridization anay that allows the characterization of specific mutations by using smaller, specialized probe anays complementary to all variants or mutations detected. Each mutation anay is composed of parallel probe sets, one complementary to the wild-type gene and the other complementary to the mutant gene.
In yet another embodiment, any of a variety of sequencing reactions known in the art can be used to directly sequence the NOVX gene and detect mutations by comparing the sequence of the sample NOVX with the conesponding wild-type (control) sequence. Examples of sequencing reactions include those based on techniques developed by Maxim and Gilbert, 1977. Proc. Natl. Acad. Sci. USA 74: 560 or Sanger, 1977. Proc. Natl. Acad. Sci. USA 74: 5463. It is also contemplated that any of a variety of automated sequencing procedures can be utilized when performing the diagnostic assays (see, e.g., Naeve, et al, 1995. Biotechniques 19: 448), including sequencing by mass specfrometry (see, e.g., PCT International Publication No. WO 94/16101; Cohen, et al, 1996. Adv. Chromatography 36: 127-162; and Griffin, et al, 1993. Appl. Biochem. Biotechnol. 38: 147-159).
Other methods for detecting mutations in the NOVX gene include methods in which protection from cleavage agents is used to detect mismatched bases in RNA/RNA or RNA/DNA heteroduplexes. See, e.g., Myers, et al, 1985. Science 230: 1242. In general, the art technique of "mismatch cleavage" starts by providing heteroduplexes of formed by hybridizing (labeled) RNA or DNA containing the wild-type NOVX sequence with potentially mutant RNA or DNA obtained from a tissue sample. The double-stranded duplexes are treated with an agent that cleaves single-stranded regions of the duplex such as which will exist due to basepair mismatches between the control and sample strands. For instance, RNA/DNA duplexes can be treated with RNase and DNA DNA hybrids freated with Si nuclease to enzymatically digesting the mismatched regions. In other embodiments, either DNA/DNA or RNA/DNA duplexes can be freated with hydroxylamine or osmium tefroxide and with piperidine in order to digest mismatched regions. After digestion of the mismatched regions, the resulting material is then separated by size on denaturing polyacrylamide gels to determine the site of mutation. See, e.g., Cotton, et al, 1988. Proc. Natl. Acad. Sci. USA 85: 4397; Saleeba, et al, 1992. Methods Enzymol. 217: 286-295. In an embodiment, the confrol DNA or RNA can be labeled for detection.
In still another embodiment, the mismatch cleavage reaction employs one or more proteins that recognize mismatched base pairs in double-stranded DNA (so called "DNA mismatch repair" enzymes) in defined systems for detecting and mapping point mutations in NOVX cDNAs obtained from samples of cells. For example, the mutY enzyme of E. coli cleaves A at G/A mismatches and the thymidine DNA glycosylase from HeLa cells cleaves T at G/T mismatches. See, e.g., Hsu, et al, 1994. Carcinogenesis 15: 1657-1662. According to an exemplary embodiment, a probe based on a NOVX sequence, e.g., a wild-type NOVX sequence, is hybridized to a cDNA or other DNA product from a test cell(s). The duplex is treated with a DNA mismatch repair enzyme, and the cleavage products, if any, can be detected from electrophoresis protocols or the like. See, e.g., U.S. Patent No. 5,459,039.
In other embodiments, alterations in electrophoretic mobility will be used to identify mutations in NOVX genes. For example, single strand conformation polymoφhism (SSCP) may be used to detect differences in electrophoretic mobility between mutant and wild type nucleic acids. See, e.g., Orita, et al, 1989. Proc. Natl. Acad. Sci. USA: 86: 2766; Cotton, 1993. Mutat. Res. 285: 125-144; Hayashi, 1992. Genet. Anal. Tech. Appl 9: 73-79. Single-stranded DNA fragments of sample and control NOVX nucleic acids will be denatured and allowed to renature. The secondary structure of single-stranded nucleic acids varies according to sequence, the resulting alteration in electrophoretic mobility enables the detection of even a single base change. The DNA fragments may be labeled or detected with labeled probes. The sensitivity of the assay may be enhanced by using RNA (rather than DNA), in which the secondary structure is more sensitive to a change in sequence. In one embodiment, the subject method utilizes heteroduplex analysis to separate double sfranded heteroduplex molecules on the basis of changes in electrophoretic mobility. See, e.g., Keen, et al, 1991. Trends Genet. 7: 5.
In yet another embodiment, the movement of mutant or wild-type fragments in polyacrylamide gels containing a gradient of denaturant is assayed using denaturing gradient gel electrophoresis (DGGE). See, e.g., Myers, et al, 1985. Nature 313: 495. When DGGE is used as the method of analysis, DNA will be modified to insure that it does not completely denature, for example by adding a GC clamp of approximately 40 bp of high-melting GC-rich DNA by PCR. In a further embodiment, a temperature gradient is used in place of a denaturing gradient to identify differences in the mobility of control and sample DNA. See, e.g., Rosenbaum andReissner, 1987 '. Biophys. Chem. 265: 12753.
Examples of other techniques for detecting point mutations include, but are not limited to, selective oligonucleotide hybridization, selective amplification, or selective primer extension. For example, oligonucleotide primers may be prepared in which the known mutation is placed centrally and then hybridized to target DNA under conditions that permit hybridization only if a perfect match is found. See, e.g., Saiki, et al, 1986. Nature 324: 163; Saiki, et al, 1989. Proc. Natl. Acad. Sci. USA 86: 6230. Such allele specific oligonucleotides are hybridized to PCR amplified target DNA or a number of different mutations when the oligonucleotides are attached to the hybridizing membrane and hybridized with labeled target DNA.
Alternatively, allele specific amplification technology that depends on selective PCR amplification may be used in conjunction with the instant invention. Oligonucleotides used as primers for specific amplification may carry the mutation of interest in the center of the molecule (so that amplification depends on differential hybridization; see, e.g., Gibbs, et al, 1989. Nucl. Acids Res. 17: 2437-2448) or at the extreme 3 '-terminus of one primer where, under appropriate conditions, mismatch can prevent, or reduce polymerase extension (see, e.g., Prossner, 1993. Tϊbtech. 11: 238). In addition it may be desirable to introduce a novel restriction site in the region of the mutation to create cleavage-based detection. See, e.g., Gasparini, et al, 1992. Mol. Cell Probes 6: 1. It is anticipated that in certain embodiments amplification may also be performed using Taq ligase for amplification. See, e.g., Barany, 1991. Proc. Natl. Acad. Sci. USA 88: 189. In such cases, ligation will ocpur only if there is a perfect match at the 3'-terminus of the 5' sequence, making it possible to detect the presence of a known mutation at a specific site by looking for the presence or absence of amplification. The methods described herein may be performed, for example, by utilizing pre-packaged diagnostic kits comprising at least one probe nucleic acid or antibody reagent described herein, which may be conveniently used, e.g., in clinical settings to diagnose patients exhibiting symptoms or family history of a disease or illness involving a NOVX gene.
Furthermore, any cell type or tissue, preferably peripheral blood leukocytes, in which NOVX is expressed maybe utilized in the prognostic assays described herein. However, any biological sample containing nucleated cells may be used, including, for example, buccal mucosal cells.
Pharmacogenomics
Agents, or modulators that have a stimulatory or inhibitory effect on NOVX activity (e.g., NOVX gene expression), as identified by a screening assay described herein can be administered to individuals to treat (prophylactically or therapeutically) disorders (The disorders include metabolic disorders, diabetes, obesity, infectious disease, anorexia, cancer-associated cachexia, cancer, neurodegenerative disorders, Alzheimer's Disease, Parkinson's Disorder, immune disorders, and hematopoietic disorders, and the various dyslipidemias, metabolic disturbances associated with obesity, the metabolic syndrome X and wasting disorders associated with chronic diseases and various cancers.) In conjunction with such treatment, the pharmacogenomics (i.e., the study of the relationship between an individual's genotype and that individual's response to a foreign compound or drag) of the individual may be considered. Differences in metabolism of therapeutics can lead to severe toxicity or therapeutic failure by altering the relation between dose and blood concenfration of the pharmacologically active drug. Thus, the pharmacogenomics of the individual permits the selection of effective agents (e.g., drugs) for prophylactic or therapeutic treatments based on a consideration of the individual's genotype. Such pharmacogenomics can further be used to determine appropriate dosages and therapeutic regimens. Accordingly, the activity of NOVX protein, expression of NOVX nucleic acid, or mutation content of NOVX genes in an individual can be determined to thereby select appropriate agent(s) for therapeutic or prophylactic treatment of the individual.
Pharmacogenomics deals with clinically significant hereditary variations in the response to drugs due to altered drag disposition and abnormal action in affected persons. See e.g., Eichelbaum, 1996. Clin. Exp. Pharmacol. Physiol, 23: 983-985; Linder, 1997. Clin. 'Chem., 43: 254-266. In general, two types of pharmacogenetic conditions can be differentiated. Genetic conditions transmitted as a single factor altering the way drags act on the body (altered drag action) or genetic conditions transmitted as single factors altering the way the body acts on drugs (altered drag metabolism). These pharmacogenetic conditions can occur either as rare defects or as polymoφhisms. For example, glucose-6-phosphate dehydrogenase (G6PD) deficiency is a common inherited enzymopathy in which the main clinical complication is hemolysis after ingestion of oxidant drags (anti-malarials, sulfonamides, analgesics, nitrofurans) and consumption of fava beans.
As an illustrative embodiment, the activity of drug metabolizing enzymes is a major determinant of both the intensity and duration of drug action. The discovery of genetic polymoφhisms of drag metabolizing enzymes (e.g., N-acetylteansferase 2 (NAT 2) and cytochrome Pregnancy Zone Protein Precursor enzymes CYP2D6 and C YP2C 19) has provided an explanation as to why some patients do not obtain the expected drag effects or show exaggerated drag response and serious toxicity after taking the standard and safe dose of a drug. These polymoφhisms are expressed in two phenotypes in the population, the extensive metabolizer (EM) and poor metabolizer (PM). The prevalence of PM is different among different populations. For example, the gene coding for
CYP2D6 is highly polymoφhic and several mutations have been identified in PM, which all lead to the absence of functional CYP2D6. Poor metabolizers of CYP2D6 and CYP2C19 quite frequently experience exaggerated drug response and side effects when they receive standard doses. If a metabolite is the active therapeutic moiety, PM show no therapeutic response, as demonstrated for the analgesic effect of codeine mediated by its CYP2D6-formed metabolite moφhine. At the other extreme are the so called ultra-rapid metabolizers who do not respond to standard doses. Recently, the molecular basis of ultra-rapid metabolism has been identified to be due to CYP2D6 gene amplification. Thus, the activity of NOVX protein, expression of NOVX nucleic acid, or mutation content of NOVX genes in an individual can be determined to thereby select appropriate agent(s) for therapeutic or prophylactic treatment of the individual. In addition, pharmacogenetic studies can be used to apply genotyping of polymoφhic alleles encoding drug-metabolizing enzymes to the identification of an individual's drag responsiveness phenotype. This knowledge, when applied to dosing or drag selection, can avoid adverse reactions or therapeutic failure and thus enhance therapeutic or prophylactic efficiency when treating a subject with a NOVX modulator, such as a modulator identified by one of the exemplary screening assays described herein. Monitoring of Effects During Clinical Trials
Monitoring the influence of agents (e.g., drugs, compounds) on the expression or activity of NOVX (e.g., the ability to modulate abenant cell proliferation and/or differentiation) can be applied not only in basic drug screening, but also in clinical trials. For example, the effectiveness of an agent determined by a screening assay as described herein to increase NOVX gene expression, protein levels, or upregulate NOVX activity, can be monitored in clinical trails of subjects exhibiting decreased NOVX gene expression, protein levels, or downregulated NOVX activity. Alternatively, the effectiveness of an agent determined by a screening assay to decrease NOVX gene expression, protein levels, or downregulate NOVX activity, can be monitored in clinical trails of subjects exhibiting increased NOVX gene expression, protein levels, or upregulated NOVX activity. In such clinical trials, the expression or activity of NOVX and, preferably, other genes that have been implicated in, for example, a cellular proliferation or immune disorder can be used as a "read out" or markers of the immune responsiveness of a particular cell .
By way of example, and not of limitation, genes, including NOVX, that are modulated in cells by freatment with an agent (e.g., compound, drug or small molecule) that modulates NOVX activity (e.g., identified in a screening assay as described herein) can be identified. Thus, to study the effect of agents on cellular proliferation disorders, for example, in a clinical trial, cells can be isolated and RNA prepared and analyzed for the levels of expression of NOVX and other genes implicated in the disorder. The levels of gene expression (i.e., a gene expression pattern) can be quantified by Northern blot analysis or RT-PCR, as described herein, or alternatively by measuring the amount of protein produced, by one of the methods as described herein, or by measuring the levels of activity of NOVX or other genes. In this manner, the gene expression pattern can serve as a marker, indicative of the physiological response of the cells to the agent. Accordingly, this response state may be determined before, and at various points during, treatment of the individual with the agent.
In one embodiment, the invention provides a method for monitoring the effectiveness of treatment of a subject with an agent (e.g., an agonist, antagonist, protein, peptide, peptidomimetic, nucleic acid, small molecule, or other drug candidate identified by the screening assays described herein) comprising the steps of (i) obtaining a pre-administeation sample from a subject prior to administration of the agent; (ii) detecting the level of expression of a NOVX protein, mRNA, or genomic DNA in the preadminisfration sample; (iii) obtaining one or more post-administration samples from the subject; (zv) detecting the level of expression or activity of the NOVX protein, mRNA, or genomic DNA in the post-administration samples; (v) comparing the level of expression or activity of the NOVX protein, mRNA, or genomic DNA in the pre-administration sample with the NOVX protein, mRNA, or genomic DNA in the post administration sample or samples; and (vi) altering the administration of the agent to the subject accordingly. For example, increased administration of the agent may be desirable to increase the expression or activity of NOVX to higher levels than detected, i.e., to increase the effectiveness of the agent. Alternatively, decreased administration of the agent may be desirable to decrease expression or activity of NOVX to lower levels than detected, i.e., to decrease the effectiveness of the agent.
Methods of Treatment The invention provides for both prophylactic and therapeutic methods of treating a subject at risk of (or susceptible to) a disorder or having a disorder associated with abenant NOVX expression or activity. The disorders include cardiomyopathy, atherosclerosis, hypertension, congenital heart defects, aortic stenosis, ateial septal defect (ASD), atrioventricular (A-V) canal defect, ductus arteriosus, pulmonary stenosis, subaortic stenosis, ventricular septal defect (VSD), valve diseases, tuberous sclerosis, scleroderma, obesity, transplantation, adrenoleukodystrophy, congenital adrenal hypeφlasia, prostate cancer, neoplasm; adenocarcinoma, lymphoma, uterus cancer, fertility, hemophilia, hypercoagulation, idiopathic thrombocytopenic puφura, immunodeficiencies, graft versus host disease, AIDS, bronchial asthma, Crohn's disease; multiple sclerosis, freatment of Albright Hereditary Ostoeodysfrophy, and other diseases, disorders and conditions of the like.
These methods of freatment will be discussed more fully, below.
Diseases and Disorders
Diseases and disorders that are characterized by increased (relative to a subject not suffering from the disease or disorder) levels or biological activity may be freated with
Therapeutics that antagonize (i.e., reduce or inhibit) activity. Therapeutics that antagonize activity maybe administered in a therapeutic or prophylactic manner. Therapeutics that may be utilized include, but are not limited to: (i) an aforementioned peptide, or analogs, derivatives, fragments or homologs thereof; (ii) antibodies to an aforementioned peptide; (iii) nucleic acids encoding an aforementioned peptide; (iv) administration of antisense nucleic acid and nucleic acids that are "dysfunctional" (i.e., due to a heterologous insertion within the coding sequences of coding sequences to an aforementioned peptide) that are utilized to "knockout" endogenous function of an aforementioned peptide by homologous recombination (see, e.g., Capecchi, 1989. Science 244: 1288-1292); or (v) modulators ( i.e., inhibitors, agonists and antagonists, including additional peptide mimetic of the invention or antibodies specific to a peptide of the invention) that alter the interaction between an aforementioned peptide and its binding partner.
Diseases and disorders that are characterized by decreased (relative to a subject not suffering from the disease or disorder) levels or biological activity may be treated with Therapeutics that increase (i.e., are agonists to) activity. Therapeutics that upregulate activity may be administered in a therapeutic or prophylactic manner. Therapeutics that may be utilized include, but are not limited to, an aforementioned peptide, or analogs, derivatives, fragments or homologs thereof; or an agonist that increases bioavailability.
Increased or decreased levels can be readily detected by quantifying peptide and/or RNA, by obtaining a patient tissue sample (e.g., from biopsy tissue) and assaying it in vitro for RNA or peptide levels, structure and/or activity of the expressed peptides (or mRNAs of an aforementioned peptide). Methods that are well-known within the art include, but are not limited to, immunoassays (e.g., by Western blot analysis, immunoprecipitation followed by sodium dodecyl sulfate (SDS) polyacrylamide gel electrophoresis, immunocytochemistry, etc.) and/or hybridization assays to detect expression of mRNAs (e.g., Northern assays, dot blots, in situ hybridization, and the like). Prophylactic Methods
In one aspect, the invention provides a method for preventing, in a subject, a disease or condition associated with an abenant NOVX expression or activity, by administering to the subject an agent that modulates NOVX expression or at least one NOVX activity. Subjects at risk for a disease that is caused or contributed to by abenant NOVX expression or activity can be identified by, for example, any or a combination of diagnostic or prognostic assays as described herein. Administration of a prophylactic agent can occur prior to the manifestation of symptoms characteristic of the NOVX abenancy, such that a disease or disorder is prevented or, alternatively, delayed in its progression. Depending upon the type of NOVX abenancy, for example, a NOVX agonist or NOVX antagonist agent can be used for treating the subject. The appropriate agent can be determined based on screening assays described herein. The prophylactic methods of the invention are further discussed in the following subsections. Therapeutic Methods
Another aspect of the invention pertains to methods of modulating NOVX expression or activity for therapeutic puφoses. The modulatory method of the invention involves contacting a cell with an agent that modulates one or more of the activities of NOVX protein activity associated with the cell. An agent that modulates NOVX protein activity can be an agent as described herein, such as a nucleic acid or a protein, a naturally-occurring cognate ligand of a NOVX protein, a peptide, a NOVX peptidomimetic, or other small molecule. In one embodiment, the agent stimulates one or more NOVX protein activity. Examples of such stimulatory agents include active NOVX protein and a nucleic acid molecule encoding NOVX that has been introduced into the cell. In another embodiment, the agent inhibits one or more NOVX protein activity. Examples of such inhibitory agents include antisense NOVX nucleic acid molecules and anti-NOVX antibodies. These modulatory methods can be performed in vitro (e.g., by culturing the cell with the agent) or, alternatively, in vivo (e.g., by administering the agent to a subject). As such, the invention provides methods of teeating an individual afflicted with a disease or disorder characterized by abenant expression or activity of a NOVX protein or nucleic acid molecule. In one embodiment, the method involves administering an agent (e.g., an agent identified by a screening assay described herein), or combination of agents that modulates (e.g., up-regulates or down-regulates) NOVX expression or activity. In another embodiment, the method involves administering a NOVX protein or nucleic acid molecule as therapy to compensate for reduced or abenant NOVX expression or activity.
Stimulation of NOVX activity is desirable in situations in which NOVX is abnormally downregulated and/or in which increased NOVX activity is likely to have a beneficial effect. One example of such a situation is where a subject has a disorder characterized by abenant cell proliferation and/or differentiation (e.g., cancer or immune associated disorders). Another example of such a situation is where the subject has a gestational disease (e.g., preclampsia). Determination of the Biological Effect of the Therapeutic
In various embodiments of the invention, suitable in vitro or in vivo assays are performed to determine the effect of a specific Therapeutic and whether its administration is indicated for treatment of the affected tissue. In various specific embodiments, in vitro assays may be performed with representative cells of the type(s) involved in the patient's disorder, to determine if a given Therapeutic exerts the desired effect upon the cell type(s). Compounds for use in therapy may be tested in suitable animal model systems including, but not limited to rats, mice, chicken, cows, monkeys, rabbits, and the like, prior to testing in human subjects. Similarly, for in vivo testing, any of the animal model system known in the art may be used prior to administration to human subjects.
Prophylactic and Therapeutic Uses of the Compositions of the Invention
The NOVX nucleic acids and proteins of the invention are useful in potential prophylactic and therapeutic applications implicated in a variety of disorders including, but not limited to: metabolic disorders, diabetes, obesity, infectious disease, anorexia, cancer-associated cancer, neurodegenerative disorders, Alzheimer's Disease, Parkinson's Disorder, immune disorders, hematopoietic disorders, and the various dyslipidemias, metabolic disturbances associated with obesity, the metabolic syndrome X and wasting disorders associated with chronic diseases and various cancers. As an example, a cDNA encoding the NOVX protein of the invention may be useful in gene therapy, and the protein may be useful when administered to a subject in need thereof. By way of non-limiting example, the compositions of the invention will have efficacy for treatment of patients suffering from: metabolic disorders, diabetes, obesity, infectious disease, anorexia, cancer-associated cachexia, cancer, neurodegenerative disorders, Alzheimer's Disease, Parkinson's Disorder, immune disorders, hematopoietic disorders, and the various dyslipidemias.
Both the novel nucleic acid encoding the NOVX protein, and the NOVX protein of the invention, or fragments thereof, may also be useful in diagnostic applications, wherein the presence or amount of the nucleic acid or the protein are to be assessed. A further use could be as an anti-bacterial molecule (i.e., some peptides have been found to possess anti-bacterial properties). These materials are further useful in the generation of antibodies, which immunospecifically-bind to the novel substances of the invention for use in therapeutic or diagnostic methods.
The invention will be further described in the following examples, which do not limit the scope of the mvention described in the claims.
EXAMPLES
Example A: Polynucleotide And Polypeptide Sequences, And Homology Data Example 1.
The NOV1 clone was analyzed, and the nucleotide and encoded polypeptide sequences are shown in Table 1 A.
Table 1A. NO VI Sequence Analysis
NOVla, CGI 03945-02 KEQ ID NO: 1 2414 bp
DNA Sequence |0RF Start: ATG at 1 lORF Stop: TAG at 2401
ATGGCCCCCTCGGCCTGGGCCATTTGCTGGCTGCTAGGGGGCCTCCTGCTCCATGGGGGTAGCTCTGGCCCCA GCCCCGGCCCCAGTGTGCCCCGCCTGCGGCTCTCCTACCGAGGAGCCGTGGTCCGAAAGCCTTCCAGCACCAT GTGGATGGAAACATTTTCCAGATACCTCCTGTCTGCCAACCGCTCTGCCATCTTTCTGGGCCCCCAGGGCTCC CTGAACCTCCAGGCCATGTACCTAGATGAGTACCGAGACCGCCTCTTTCTGGGTGGCCTGGACGCCCTCTACT CTCTGCGGCTGGACCAGGCATGGCCAGATCCCCGGGAGGTCCTGTGGCCACCGCAGCCAGGACAGAGGGAGGA GTGTGTTCGAAAGGGAAGAGATCCTTTGACAGAGTGCGCCAACTTCGTGCGGGTGCTACAGCCTCACAACCGG ACCCACCTGCTAGCCTGTGGCACTGGGGCCTTCCAGCCCACCTGTGCCCTCATCACA.GTTGGCCACCGTGGGG AGCATGTGCTCCACCTGGAGCCTGGCAGTGTGGAAAGTGGCCGGGGGCGGTGCCCTCACGAGCCCAGCCGTCC CTTTGCCAGCACCTTCATAGACGGGGAGCTGTACACGGGTCTCACTGCTGACTTCCTGGGGCGAGAGGCCATG ATCTTCCGAAGTGGAGGTCCTCGGCCAGCTCTGCGTTCCGACTCTGACCAGAGTCTCTTGCACGACCCCCGGT TTGTGATGGCCGCCCGGATCCCTGAGAACTCTGACCAGGACAATGACAAGGTGTACTTCTTCTTCTCGGAGAC GGTCCCCTCGCCCGATGGTGGCTCGAACCATGTCACTGTCAGCCGCGTGGGCCGCGTCTGCGTGAATGATGCT GGGGGCCAGCGGGTGCTGGTGAACAAATGGAGCACTTTCCTCAAGGCCAGGCTGGTCTGCTCGGTGCCCGGCC CTGGTGGTGCCGAGACCCACTTTGACCAGCTAGAGGATGTGTTCCTGCTGTGGCCCAAGGCCGGGAAGAGCCT CGAGGTGTACGCGCTGTTCAGCACCGTCAGTGCCGTGTTCCAGGGCTTCGCCGTCTGTGTGTACCACATGGCA GACATCTGGGAGGTTTTCAACGGGCCCTTTGCCCACCGAGATGGGCCTCAGCACCAGTGGGGGCCCTATGGGG GCAAGGTGCCCTTCCCTCGCCCTGGCGTGTGCCCCAGCAAGATGACCGCACAGCCAGGACGGCCTTTTGGCAG CACCAAGGACTACCCAGATGAGGTGCTGCAGTTTGCCCGAGCCCACCCCCTCATGTTCTGGCCTGTGCGGCCT CGACATGGCCGCCCTGTCCTTGTCAAGACCCACCTGGCCCAGCAGCTACACCAGATCGTGGTGGACCGCGTGG AGGCAGAGGATGGGACCTACGATGTCATTTTCCTGGGGACTGACTCAGGGTCTGTGCTCAAAGTCATCGCTCT CCAGGCAGGGGGCTCAGCTGAACCTGAGGAAGTGGTTCTGGAGGAGCTCCAGGTGTTTAAGGTGCCAACACCT ATCACCGAAATGGAGATCTCTGTCAAAAGGCAAATGCTATACGTGGGCTCTCGGCTGGGTGTGGCCCAGCTGC GGCTGCACCAATGTGAGACTTACGGCACTGCCTGTGCAGAGTGCTGCCTGGCCCGGGACCCATACTGTGCCTG GGATGGTGCCTCCTGTACCCACTACCGCCCCAGCCTTGGCAAGCGCCGGTTCCGCCGGCAGGACATCCGGCAC GGCAACCCTGCCCTGCAGTGCCTGGGCCAGAGCCAGGAAGAAGAGGCAGTGGGACTTGTGGCAGCCACCATGG TCTACGGCACGGAGCACAATAGCACCTTCCTGGAGTGCCTGCCCAAGTCTCCCCAGGCTGCTGTGCGCTGGCT CTTGCAGAGGCCAGGGGATGAGGGGCCTGACCAGGTGAAGACGGACGAGCGAGTCTTGCACACGGAGCGGGGG CTGCTGTTCCGCAGGCTTAGCCGTTTCGATGCGGGCACCTACACCTGCACCACTCTGGAGCATGGCTTCTCCC AGACTGTGGTCCGCCTGGCTCTGGTGGTGATTGTGGCCTCACAGCTGGACAACCTGTTCCCTCCGGAGCCAAA GCCAGAGGAGCCCCCAGCCCGGGGAGGCCTGGCTTCCACCCCACCCAAGGCCTGGTACAAGGACATCCTGCAG CTCATTGGCTTCGCCAACCTGCCCCGGGTGGATGAGTACTGTGAGCGCGTGTGGTGCAGGGGCACCACGGAAT GCTCAGGCTGCTTCCGGAGCCGGAGCCGGGGCAAGCAGGCCAGGGGCAAGAGCTGGGCAGGGCTGGAGCTAGG CAAGAAGATGAAGAGCCGGGTGCATGCCGAGCACAATCGGACGCCCCGGGAGGTGGAGGCCACGTAGAAGGGG GCAGA NOVla, CG103945-02 SEQ ID NO: 2 800 aa MW at 88800.3kD Protein Sequence
MAPSA AIC L GG X.LHGGSSGPSPGPSVPR RLSYRGAWRKPSSTMMETFSRY LSANRSAIF GPQGS NLQAMY DEYRDRLF GG DALYSLR DQAWPDPREVLWPPQPGQREECVRKGRDP TECANFVRVLQPHNR TH LACGTGAFQPTCALITVGHRGEHVLHLEPGSVESGRGRCPHEPSRPFASTFIDGELYTGLTADF GREAM IFRSGGPRPALRSDSDQSL HDPRFVMAARIPENSDQDNDKVYFFFSETVPSPDGGSNHV VSRVGRVCVNDA GGQRVLVNK STFLKAR VCSVPGPGGAETHFDQLEDVFL WPKA.GKS EVYALFSTVSAVFQGFAVCVYHMA DI EVFNGPFAHRDGPQHQWGPYGGKVPFPRPGVCPSKMTAQPGRPFGSTKDYPDEVLQFARAHPLMFWPVRP RHGRPVLVKTHLAQQLHQIVVDRVEAEDGTYDVIF GTDSGSVLKVIALQAGGSAEPEEVV EE QVFKVPTP ITEMEISVKRQM YVGSR GVAQLR HQCETYGTACAECCLARDPYCA DGASCTHYRPSLGKRRFRRQDIRH GNPALQCLGQSQEEEAVGLVAATMVYGTEHNSTFLECLPKSPQAAVR QRPGDEGPDQVKTDERVLHTERG LLFRR SRFDAGTYTCTTLEHGFSQTWRLALWIVASQ DNLFPPEPKPEEPPARGG ASTPPKA YKDILQ LIGFANLPRVDEYCERVWCRGTTECSGCFRSRSRGKQARGKS AGLELGKKMKSRVHAEHNRTPREVEAT
NOVlb, CG103945-01 jSEQ ID NO: 3 4700 bp
DNA Sequence ORF Start: ATG at 1 ORF Stop: TAG at 2347
ATGGCCCCCTCGGCCTGGGCCATTTGCTGGCTGCTAGGGGGCCTCCTGCTCCATGGGGGTAGCTCTGGCCCCA GCCCCGGCCCCAGTGTGCCCCGCCTGCGGCTCTCCTACCGAGACCTCCTGTCTGCCAACCGCTCTGCCATCTT TCTGGGCCCCCAGGGCTCCCTGAACCTCCAGGCCATGTACCTAGATGAGTACCGAGACCGCCTCTTTCTGGGT GGCCTGGACGCCCTCTACTCTCTGCGGCTGGACCAGGCATGGCCAGATCCCCGGGAGGTCCTGTGGCCACCGC AGCCAGGACAGAGGGAGGAGTGTGTTCGAAAGGGAAGAGATCCTTTGACAGAGTGCGCCAACTTCGTGCGGGT GCTACAGCCTCACAACCGGACCCACCTGCTAGCCTGTGGCACTGGGGCCTTCCAGCCCACCTGTGCCCTCATC ACAGTTGGCCACCGTGGGGAGCATGTGCTCCACCTGGAGCCTGGCAGTGTGGAAAGTGGCCGGGGGCGGTGCC CTCACGAGCCCAGCCGTCCCTTTGCCAGCACCTTCATAGACGGGGAGCTGTACACGGGTCTCACTGCTGACTT CCTGGGGCGAGAGGCCATGATCTTCCGAAGTGGAGGTCCTCGGCCAGCTCTGCGTTCCGACTCTGACCAGAGT CTCTTGCACGACCCCCGGTTTGTGATGGCCGCCCGGATCCCTGAGAACTCTGACCAGGACAATGACAAGGTGT ACTTCTTCTTCTCGGAGACGGTCCCCTCGCCCGATGGTGGCTCGAACCATGTCACTGTCAGCCGCGTGGGCCG CGTCTGCGTGAATGATGCTGGGGGCCAGCGGGTGCTGGTGAACAAATGGAGCACTTTCCTCAAGGCCAGGCTG GTCTGCTCGGTGCCCGGCCCTGGTGGTGCCGAGACCCACTTTGACCAGCTAGAGGATGTGTTCCTGCTGTGGC CCAAGGCCGGGAAGAGCCTCGAGGTGTACGCGCTGTTCAGCACCGTCAGTGCCGTGTTCCAGGGCTTCGCCGT CTGTGTGTACCACATGGCAGACATCTGGGAGGTTTTCAACGGGCCCTTTGCCCACCGAGATGGGCCTCAGCAC CAGTGGGGGCCCTATGGGGGCAAGGTGCCCTTCCCTCGCCCTGGCGTGTGCCCCAGCAAGATGACCGCACAGC CAGGACGGCCTTTTGGCAGCACCAAGGACTACCCAGATGAGGTGCTGCAGTTTGCCCGAGCCCACCCCCTCAT GTTCTGGCCTGTGCGGCCTCGACATGGCCGCCCTGTCCTTGTCAAGACCCACCTGGCCCAGCAGCTACACCAG ATCGTGGTGGACCGCGTGGAGGCAGAGGATGGGACCTACGATGTCATTTTCCTGGGGACTGACTCAGGGTCTG TGCTCAAAGTCATCGCTCTCCAGGCAGGGGGCTCAGCTGAACCTGAGGAAGTGGTTCTGGAGGAGCTCCAGGT GTTTAAGGTGCCAACACCTATCACCGAAATGGAGATCTCTGTCAAAAGGCAAATGCTATACGTGGGCTCTCGG CTGGGTGTGGCCCAGCTGCGGCTGCACCAATGTGAGACTTACGGCACTGCCTGTGCAGAGTGCTGCCTGGCCC GGGACCCATACTGTGCCTGGGATGGTGCCTCCTGTACCCACTACCGCCCCAGCCTTGGCAAGCGCCGGTTCCG CCGGCAGGACATCCGGCACGGCAACCCTGCCCTGCAGTGCCTGGGCCAGAGCCAGGAAGAAGAGGCAGTGGGA CTTGTGGCAGCCACCATGGTCTACGGCACGGAGCACAATAGCACCTTCCTGGAGTGCCTGCCCAAGTCTCCCC AGGCTGCTGTGCGCTGGCTCTTGCAGAGGCCAGGGGATGAGGGGCCTGACCAGGTGAAGACGGACGAGCGAGT CTTGCACACGGAGCGGGGGCTGCTGTTCCGCAGGCTTAGCCGTTTCGATGCGGGCACCTACACCTGCACCACT CTGGAGCATGGCTTCTCCCAGACTGTGGTCCGCCTGGCTCTGGTGGTGATTGTGGCCTCACAGCTGGACAACC TGTTCCCTCCGGAGCCAAAGCCAGAGGAGCCCCCAGCCCGGGGAGGCCTGGCTTCCACCCCACCCAAGGCCTG GTACAAGGACATCCTGCAGCTCATTGGCTTCGCCAACCTGCCCCGGGTGGATGAGTACTGTGAGCGCGTGTGG TGCAGGGGCACCACGGAATGCTCAGGCTGCTTCCGGAGCCGGAGCCGGGGCAAGCAGGCCAGGGGCAAGAGCT GGGCAGGGCTGGAGCTAGGCAAGAAGATGAAGAGCCGGGTGCATGCCGAGCACAATCGGACGCCCCGGGAGGT GGAGGCCACGTAGAAGGGGGCAGAGGAGGGGTGGTCAGGATGGGCTGGGGGGCCCACTAGCAGCCCCCAGCAT CTCCCACCCACCCAGCTAGGGCAGAGGGGTCAGGATGTCTGTTTGCCTCTTAGAGACAGGTGTCTCTGCCCCC ACACCGCTACTGGGGTCTAATGGAGGGGCTGGGTTCTTGAAGCCTGTTCCCTGCCCTTCTCTGTGCTCTTAGA CCCAGCTGGAGCCAGCACCCTCTGGCTGCTGGCAGCCCCAAGGGATCTGCCATTTGTTCTCAGAGATGGCCTG GCTTCCGCAACACATTTCCGGGTGTGCCCAGAGGCAAGAGGGTTGGGTGGTTCTTTCCCAGCCTACAGAACAA TGGCCATTCTGAGTGACCCTCAGAGTGGGTGTGTGGGTGCGTCTAGGGGGTATCCCGGTAGGGGGCCTGCAGG GAGCCAGAGGGTGGAAATGGCCTCTAAGCTAGCACCCCGTAAGAAGAGCCTACCTGACCGACTTGGGGAGGGA ACACAGAGGTGTTGGGAAGGTGGAGCAACAATGCACCTCCCCTCCTGTCGCGCCGTGATATCTTGGTGGCTCC
CTGCCACTGCCCACCGCCTCTTCTCCATCTGAGAATCACGGAGAGGTGTAGATAATCTAGAGGCATAGACTGC
ITAGAGCCCCCAGGGATCTGGGGTGGTCAGGGCTCAGGCTTCACTTTGTAAACCAGGTGGGGGCATCTCACAGC
CTGACTTCCCTTCCCCAGGCCAGGGTTGCTGGGATGCCTGCCCCTCCTGAGAGGACCCCCTCCCCATTGTCAG
GCTCTCCATGTCCACGAGCGGGGAGGGGTGGGTTCTGGGGCATTGTTGTCCCTTGTGTCTGTGGACTAGAGAT
AGGGTGGGGGAGCTGGGGAAGGGTGCAGGCGGGAAGAGTGGGCTGTCTTTCCCAGGGTGATGCAAGCATGCCG
CAGCCCTGGAGGCTGGGAATGTGGAGGCTCTGTGAGCCCTGCAGCCC
TCAGAATCAGGGCCAGGGATGCAGAAGATTGAGAGGATATGGAGATGGATAGAGGGCAGGAGACCCTTAGGAT
AGATTGTGGGACCCAGGCAGGAACAGGTGTCCACAAGAACTCAGGATGGCATCAGTTAGCTCAGAAGCCACCT
GGAAGACCCAGTGTTTCCATCTCTGGAATCTCTGTTTTATGCTAAATGGATTTAGGAAGACTGTTTTTCTTTT AGGGGGAAACAAGGTAGAGAAAAGGACGAAGAAGTGTAAGTCCCGCTGATTCTCGGGGGTAAGGCTCGGATG
GCAAGGACGCGTTCTGCCTGGGCATGTAGGGGAGGTGTTTTTGCCATCACCAGTTTCTCAGGCTGGGGAGCAC
AGAGGGGAGGAGGAGGACTAAATGAAAAGTTGTTCCCAGCCTGCACATGAACACATTCATGACACACAAAACT
GGCTGGAAGGAGATAAGAGCACTGGGTTTGAGATTCCCTCCATTAAAACAACCAAGACAAAGAAAGGAGGGGA iAAAAAAGATAAAAAGCAAGCCAGGGTTCCCTGCCCTATTGAAACTCAAACCCAGACTGCCTTGGGTTTTATCT
TTCCCTTACCCCTGGCACCTCCAGAGAACTGGGACCTGAAATAGTCCCTCCGTTCTCCCCTTTGACCATGTAA
ITAAATGAACCAGAAGCACTGAGATTAACCTATCAACGCCCTGAGAAGCCTTCCAGCCTGCGGTGCTGTCTGCT iGGGAGGTCAGCTGGTCAAGGCAGAGGAGGAGAGGAGGAAAGGATGGGGGCTGAAGAGCAGAAGGGAGGGGAGA
CAGAGGGGATTAAAGAGGGGAGGAGAGAGTGCAGAGCTCCAGGAAAGGGTATCAGAGCTGCAGCCAGCTCTGC
CCTCTACCCTAGGGAGGCCAGAAAGACACAAACAGCCCTCCGGGCCTTTACGCTGGACTCTGGCTTGGCAGGC
TCCAGGCAGGGTCCTCTGGGAAGTTACTCTAGAAAACGAAGGGAGGAGGAGCACAAGATCCTCAGCAACGAAC
ACCTGCACTTAGAAAAAGTGGACAGCTTCTGCCAACCACACCCTACCCATGGTACTGTATGCTATTAACTCCT iGGAAACGCCCCGTAAATGCGAGTTGTTTTTGTATTTGTGTGTTGAGATGGGCCTTGTGGTTTCTCTGTACTCA
GAGCACATTTCTTGTAATTACTATTGTTATTTTTATTGTCATGACTGCCCCTGAGCTCTGGTGAGAAAAGCTG lAATTTACAAGGAAAGGGATGAAGTTAATATTTGCATCACATAATTATATCATTACTGTGTATCTGTGTATTGT
ACTAAATGGACTGATGCTGCGCACATGAGCTGAAAATGAAGAGCCCTCCCATCC
NOVlb, CG103945-01 SEQ ID NO: 4 782 aa MW at 86699.9kD
Protein Sequence I
MAPSAWAIC LLGG bHGGSSGPSPGPSVPRLRLSYRDLLSANRSAIF GPQGSLNLQAMYLDEYRDRLFLG GLDA YS RLDQA PDPREV PPQPGQREECVRKGRDP TECANFVRVLQPHNRTH ACGTGAFQPTCA I TVGHRGEHVLHLEPGSVESGRGRCPHEPSRPFASTFIDGELYTG TADF GREAMIFRSGGPRPA RSDSDQS LHDPRFVMAARIPENSDQDNDKVYFFFSETVPSPDGGSNHVTVSRVGRVCVNDAGGQRVVNKWSTF KARL VCSVPGPGGAETHFDQ EDVFL PKAGKSLEVYALFSTVSAVFQGFAVCVYH ADIWEVFNGPFAHRDGPQH Q GPYGGKVPFPRPGVCPSKMTAQPGRPFGSTKDYPDEVLQFARAHPLMF PVRPRHGRPVLVKTH AQQ HQ IWDRVEAEDGTYDVIF GTDSGSV KVIALQAGGSAEPEEWLEE QVFKVPTPITEMEISVKRQMLYVGSR LGVAQ R HQCETYGTACAECCLARDPYCAWDGASCTHYRPS GKRRFRRQDIRHGNPA QCLGQSQEEEAVG VAATMVYGTEHNSTFLECLPKSPQAAVRWL QRPGDEGPDQVKTDERV HTERGLLFRR SRFDAGTYTCTT LEHGFSQTWR AWIVASQLDN FPPEPKPEEPPARGGLASTPPKAWYKDIL.QLIGFANLPRVDEYCERVW CRGTTECSGCFRSRSRGKQARGKSWAGLELGKKMKSRVHAEHNRTPREVEAT
A ClustalW comparison of the above protein sequences yields the following sequence alignment shown in Table IB. Table IB. Comparison of the NOVl protein sequences.
NOVla APSA AIC L GGL LHGGSSGPSPGPSVPR R SYRGAWRKPSSTMWMETFSRYL S
NOVlb MAPSAWAICWLLGG LLHGGSSGPSPGPSVPR R SYR D LLS
NOVla ANRSAIF GPQGSLNLQAMYLDEYRDRLFLGG DALYSLRLDQA PDPREVIi PPQPGQR
NOVlb ANRSAIF GPQGSLNLQAMYLDEYRDR F GG DAYSLRLDQAWPDPREVLWPPQPGQR
NOVla EECVRKGRDP TECANFVRVIiQPHNRTHLLACGTGAFQPTCA ITVGHRGEHVLH EPGS
NOVlb EECVRKGRDP TECANFVRVLQPHNRTHLLACGTGAFQPTCALITVGHRGEHVHLEPGS
NOVla VESGRGRCPHEPSRPFASTFIDGELYTG TADF GREAMIFRSGGPRPA RSDSDQSL H
NOVlb VESGRGRCPHEPSRPFASTFIDGE YTGLTADFLGREAMIFRSGGPRPALRSDSDQSLLH
NOVla DPRFVMAARIPENSDQDNDKVYFFFSETVPSPDGGSNHVTVSRVGRVCVNDAGGQRVLVN
NOVlb DPRFVMAARIPENSDQDNDKVYFFFSETVPSPDGGSNHVTVSRVGRVCVNDAGGQRVLVN
NOVla KWSTFLKARLVCSVPGPGGAETHFDQLEDVF WPKAGKSLΞVYA FSTVSAVFQGFAVC
NOVlb KWSTF KARLVCSVPGPGGAETHFDQLEDVFLLWPKAGKSLEVYALFSTVSAVFQGFAVC
NOVla VYH ADI EVFNGPFAHRDGPQHQ GPYGGKVPFPRPGVCPSKMTAQPGRPFGST DYPD
NOVlb VYHMADIWEVFNGPFAHRDGPQHQ GPYGGKVPFPRPGVCPSKMTAQPGRPFGSTKDYPD
NOVla EVLQFARAHPLMFWP PRHGRPV VKTH AQQLHQIVVDRVEAEDGTYDVIFLGTDSGS
NOVlb EVLQFARAHPLMF PVRPRHGRPVIiVKTH AQQ HQIWDRVEAEDGTYDVIFLGTDSGS
NOVla V KVI LQAGGSAEPEEWLEE QVFKVPTPITEMEISVKRQMLYVGSRLGVAQLRLHQC
NOVlb V KVIA QAGGSAEPEEVVLEELQVFKVPTPITEMEISVKRQMLYVGSR GVAQLRLHQC
NOVla ETYGTACAECCLARDPYCA DGASCTHYRPSLG RRFRRQDIRHGNPALQCLGQSQEEEA
NOVlb ETYGTACAECCLARDPYCA DGASCTHYRPS GKRRFRRQDIRHGNPALQCLGQSQEEEA
NOVla VGLVAATMVYGTEHNSTFLECLPKSPQAAVRWLLQRPGDEGPDQVKTDERVLHTERGL F
NOVlb VGLVAATMVYGTEHNSTF ECLPKSPQAAVRWL QRPGDEGPDQVKTDERVLHTERG LF
NOVla RR SRFDAGTYTCTTLEHGFSQTWRLAWIVASQLDNLFPPEPKPEEPPARGGLASTP
NOVlb RRLSRFDAGTYTCTTLEHGFSQTWRLAWIVASQLDNLFPPEPKPEEPPARGGLASTP
NOVla PKAYKDILQLIGFAN PRVDEYCERV CRGTTECSGCFRSRSRGKQARGKS AGLELGK
NOVlb PKAWYKDILQLIGFAN PRVDEYCERV CRGTTECSGCFRSRSRGKQARGKS AGLE GK
NOVla KMKSRVHAEHNRTPREVEAT
NOVlb KMKSRVHAEHNRTPREVEAT
NOVla (SEQ ID NO: 2) NOVlb (SEQ ID NO: 4)
Further analysis of the NOVla protein yielded the following properties shown in Table IC. Table IC. Protein Sequence Properties NOVla
SignalP analysis: j Cleavage site between residues 23 and 24
PSORT II analysis:
PSG: a new signal peptide prediction method
N-region: length 0; pos.chg 0; neg.chg 0 H-region: length 31; peak value 9.35 PSG score : 4.95
GvH: von Heijne's method for signal seq. recognition GvH score (threshold: -2.1): 1.50 possible cleavage site: between 22 and 23
>>> Seems to have a cleavable signal peptide (1 to 22)
ALOM: Klein et al s method for TM region allocation Init position for calculation: 23
Tentative number of TMS(s) for the threshold 0.5: 3 Number of TMS(s) for threshold 0.5: 1
INTEGRAL Likelihood = -2.02 Transmembrane 345 - 361 PERIPHERAL Likelihood = 2.86 (at 150) ALOM score: -2.02 (number of TMSs : 1)
MTOP: Prediction of membrane topology (Hartmann et al.) Center position for calculation: 11 Charge difference: 0.5 C( 1.5) - N( 1.0) C > N: C-terminal side will be inside
»>Caution: Inconsistent top result with signal peptide
>>> membrane topology: type la (cytoplasmic tail 362 to 800)
MITDISC: discrimination of mitochondrial targeting seq R content: 4 Hyd Moment (75): 2.13 Hyd Moment (95): 2.46 G content: 7 D/E content: 1 S/T content: 9 Score : -2.94
Gavel : prediction of cleavage sites for mitochondrial preseq R-2 motif at 73 RS|AI
NUCDISC: discrimination of nuclear localization signals pat : none pat7: PSLGKRR (3) at 570 bipartite : none content of basic residues: 11.4% NLS Score: -0.22
KDEL: ER retention motif in the C-terminus : none
ER Membrane Retention Signals: none
SKL: peroxisomal targeting signal in the C-terminus: none
PTS2: 2nd peroxisomal targeting signal: none
VAC: possible vacuolar targeting motif: none
RNA-binding motif : none
Actinia-type actin-binding motif: type 1: none type 2 : none NMYR: N-myristoylation pattern : none
Prenylation motif: none me YQRL: transport motif from cell surface to Golgi: none
Tyrosines in the tail : too long tail
Dileucine motif in the tail : found LL at 633 LL at 658 checking 63 PROSITE DNA binding motifs: none checking 71 PROSITE ribosomal protein motifs: none checking 33 PROSITE prokaryotic DNA binding motifs: none
NNCN: Reinhardt's method for Cytoplasmic/Nuclear discrimination Prediction: cytoplasmic Reliability: 89
COIL: Lupas's algorithm to detect coiled-coil regions total: 0 residues
Final Results (k = 9/23)
44.4 endoplasmic reticulum 22.2 Golgi 22.2 extracellular, including cell wall 11.1 plasma membrane
» prediction for CG103945-02 is end (k=9)
A search of the NOVla protein against the Geneseq database, a proprietary database that contains sequences published in patents and patent publication, yielded several homologous proteins shown in Table ID.
Figure imgf000113_0001
In a BLAST search of public sequence databases, the NOVla protein was found to have homology to the proteins shown in the BLASTP data in Table IE.
Il l
Figure imgf000114_0001
PFam analysis predicts that the NOVla protein contains the domains shown in the Table IF.
Figure imgf000114_0002
Example 2.
The NOV2 clone was analyzed, and the nucleotide and encoded polypeptide sequences are shown in Table 2A.
Figure imgf000115_0001
GGGCCTTGACTGCCTGGGGCCAGCCATCCACATCGCCAACTGCTCCAGGAATGGGGCGTGGACCCCGTGGTCA TCGTGGGCGCTGTGCAGCACGTCCTGTGGCATCGGCTTCCAGGTCCGCCAGCGAAGTTGCAGCAACCCTGCTC CCCGCCACGGGGGCCGCATCTGCGTGGGCAAGAGCCGGGAGGAACGGTTCTGTAATGAGAACACGCCTTGCCC GGTGCCCATCTTCTGGGCTTCCTGGGGCTCCTGGAGCAAGTGCAGCAGCAACTGTGGAGGGGGCATGCAGTCG CGGCGTCGGGCCTGCGAGAACGGCAACTCCTGCCTGGGCTGCGGCGTGGAGTTCAAGACGTGCAACCCCGAGG GCTGCCCCGAAGTGCGGCGCAACACCCCCTGGACGCCGTGGCTGCCCGTGAACGTGACGCAGGGCGGGGCACG GCAGGAGCAGCGGTTCCGCTTCACCTGCCGCGCGCCCCTTGCAGACCCGCACGGCCTGCAGTTCGGCAGGAGA AGGACCGAGACGAGGACCTGTCCCGCGGACGGCTCCGGCTCCTGCGACACCGACGCCCTGGTGGAGGACCTCC TGCGCAGCGGGAGCACCTCCCCGCACACGGTGAGCGGGGGCTGGGCCGCCTGGGGCCCGTGGTCGTCCTGCTC CCGGGACTGCGAGCTGGGCTTCCGCGTCCGCAAGAGAACGTGCACTAACCCGGAGCCCCGCAACGGGGGCCTG CCCTGCGTGGGCGATGCTGCCGAGTACCAGGACTGCAACCCCCAGGCTTGCCCAGTTCGGGGTGCTTGGTCCT GCTGGACCTCATGGTCTCCATGCTCAGCTTCCTGTGGTGGGGGTCACTATCAACGCACCCGTTCCTGCACCAG CCCCGCACCCTCCCCAGGTGAGGACATCTGTCTCGGGCTGCACACGGAGGAGGCACTATGTGCCACACAGGCC TGCCCAGAAGGCTGGTCGCCCTGGTCTGAGTGGAGTAAGTGCACTGACGACGGAGCCCAGAGCCGAAGCCGGC ACTGTGAGGAGCTCCTCCCAGGGTCCAGCGCCTGTGCTGGAAACAGCAGCCAGAGCCGCCCCTGCCCCTACAG CGAGATTCCCGTCATCCTGCCAGCCTCCAGCATGGAGGAGGCCACCGGCTGTGCAGGGTTCAATCTCATCCAC TTGGTGGCCACGGGCATCTCCTGCTTCTTGGGCTCTGGGCTCCTGACCCTAGCAGTGTACCTGTCTTGCCAGC ACTGCCAGCGTCAGTCCCAGGAGTCCACACTGGTCCATCCTGCCACCCCCAACCATTTGCACTACAAGGGCGG AGGCACCCCGAAGAATGAAAAGTACACACCCATGGAATTCAAGACCCTGAACAAGAATAACTTGATCCCTGAT GACAGAGCCAACTTCTACCCATTGCAGCAGACCAAΪGTGTACACGACTACTTACTACCCAAGCCCCCTGAACA AACACAGCTTCCGGCCCGAGGCCTCACCTGGACAACGGTGCTTCCCCAACAGCTGATACCGCCGTCCTGGGGA CTTGGGCTTCTTGCCTTCATAAGGCACAGAGCAGATGGAGATGGGACAGTGGAGCCAGTTTGGTTTTCTCCCT
CTGCACTAGGCCAAGAACTTGCTGCCTTGCCTGTGGGGGGTCCCATCCGGCTTCAGAGAGCTCTGGCTGGCAT
TGACCATGGGGGAAAGGGCTGGTTTCAGGCTGACATATGGCCGCAGGTCCAGTTCAGCCCAGGTCTCTCATGG
TTATCTTCCAACCCACTGTCACGCTGACACTATGCTΘCCATGCCTGGGCTGTGGACCTACTGGGCATTTGAGG lAACTGGAGAATGGAGATGGCAAGAGGGCAGGCTTTTAAGTTTGGGTTGGAGACAACTTCCTGTGGCCCCCACA lAGCTGAGTCTGGCCTTCTCCAGCTGGCCCCAAAAAAGGCCTTTGCTACATCCTGATTATCTCTGAAAGTAATC lAATCAAGTGGCTCCAGTAGCTCTGGATTTTCTGCCAGGGCTGGGCCATTGTGGTGCTGCCCCAGTATGACATG
GGACCAAGGCCAGCGCAGGTTATCCACCTCTGCCTGGAAGTCTATACTCTACCCAGGGCATCCCTCTGGTCAG lAGGCAGTGAGTACTGGGAACTGGAGGCTGACCTGTGCTTAGAAGTCCTTTAATCTGGGCTGGTACAGGCCTCA
GCCTTGCCCTCAATGCACGAAAGGTGGCCCAGGAGAGAGGATCAATGCCACAGGAGGCAGAAGTCTGGCCTCT
GTGCCTCTATGGAGACTATCTTCCAGTTGCTGCTCAACAGAGTTGTTGGCTGAGACCTGCTTGGGAGTCTCTG
CTGGCCCTTCATCTGTTCAGGAACACACACACACACACACTCACACACGCACACACAATCACAATTTGCTACA
GCAACAAAAAAGACATTGGGCTGTGGCATTATTAATTAAAGATGATATCCAGTCTCC
NOV2a, CG106951-01 SEQ ID NO: 6 1352 aa MW at 145674.1kD Protein Sequence
MPAGEGASAHRAGHHTRQARGGSRPSSRG QGAPSRSSARLEAGGCSARRGRSAPAPSSFSLPLPSFSPFACN SSPTAPSLLLLPRSPPPCSLRAPGRELVGARG VPEPSSAEPGGSAAHPAAAGSPSAAGAGPGGDCTGALRAG GRSCAAAPFPDRPPAHLVSSRRSAPPGSREPRGTGH HPPLGVSGSS CLACVS PCGFSPSPVAHHLVPGP PDTPAQQLRCGWTVGG LLSLVRGLLPCLPPGARTAEGPIMVLAGPLAVSLLLPSLTLLVSHLSSSQDVSSEP SSEQQLCALSKHPTVAFEDLQP VSNFTYPGARDFSQLALDPSGNQLIVGARNYLFRLSLAVSLLQATEWAS SEDTRRSCQSKG TEEECQNYVRVLIVAGRKVFMCGTNAFSPMCTSRQVGNLSRTTEKINGVARCPYDPRHNS TAVISSQGELYAATVIDFSGRDPAIYRSLGSGPPLRTAQYNSKWLNEPNFVAAYDIGLFAYFFLRENAVEHDC GRTVYSRVARVCK DVGGRFLLEDTWTTFMKAR NCSRPGEVPFYYNELQSAFHLPEQDLIYGVFTTNVNSIA ASAVCAFNLSAISQAFNGPFRYQENPRAAWLPIANP1PNFQCGTLPETGPNENLTERSLQDAQRLFLMSEAVQ PVTPEPCV QDSVRFSHLVVDLVQAKDT YH'VYIGTESGTILKALSTASRSLHGCYLEΞ HVLPPGRREPLR SLRI HSARALFVGLRDGVLRVPLERCAAYRSQGACLGARDPYCG DGKQQRCSTLEDSS MSLWTQNITACP VRNVTRDGGFGPWSP QPCEHLDGDNSGSCLCRARSCDSPRPRCGGLDCLGPAIHIANCSRNGAWTP SSWA CSTSCGIGFQVRQRSCSNPAPRHGGRICVGKSREERFCNENTPCPVPIF ASWGS SKCSSNCGGGMQSRRRA CENGNSCLGCGVEFKTCNPEGCPEVRR TPWTP LPVNV QGGARQEQRFRFTCRAPLADPHGLQFGRRRTET RTCPADGSGSCDTDALVEDLLRSGSTSPH VSGGWAAWGPWSSCSRDCELGFRVRKRTCTNPEPRNGG PCVG DAAEYQDCNPQACPVRGA SCWTSWSPCSASCGGGHYQRTRSCTSPAPSPGEDICLGLHTEEALCATQACPEG WSPWSEWSKCTDDGAQSRSRHCEELLPGSSACAGNSSQSRPCPYSEIPVILPASS EEATGCAGFNLIHLVAT GISCFLGSG IiTLAVYLSCQHCQRQSQESTLVHPATPNHLHYKGGGTPKNEKYTPMEFKTLNKN BIPDDRAN FYPLQQTNVYTTTYYPSPLNKHSFRPEASPGQRCFPNS |NOV2b, CGI 06951-04 fSEQ IDNO: 7 |3631 bp
DNA Sequence
GCGGCCGCCCCATTCCCAGACCGGCCGCCAGCCCATCTGGTTAGCTCCCGCCGCTCCGCGCCGCCCGGGAGTC
GGGAGCCGCGGGGAACCGGGCACCTGCACCCGCCTCTGGGAGTGAGTGGTTCCAGCTGGTGCCTGGCCTGTGT
CTCTTGGATGCCCTGTGGCTTCAGTCCGTCTCCTGTTGCCCACCACCTCGTCCCTGGGCCGCCTGATACCCCA
GCCCAACAGCTAAGGTGTGGATGGACAGTAGGGGGCTGGCTTCTCTCACTGGTCAGGGGTCTTCTCCCCTGTC TGCCTCCCGGAGCTAGGACTGCAGAGGGGCCTATCATGGTGCTTGCAGGCCCCCTGGCTGTCTCGCTGTTGCT GCCCAGCCTCACACTGCTGGTGTCCCACCTCTCCAGCTCCCAGGATGTCTCCAGTGAGCCCAGCAGTGAGCAG CAGCTGTGCGCCCTTAGCAAGCACCCCACCGTGGCCTTTGAAGACCTGCAGCCGTGGGTCTCTAACTTCACCT ACCCTGGAGCCCGGGATTTCTCCCAGCTGGCTTTGGACCCCXCCGGGAACCAGCTCATCGTGGGAGCCAGGAA CTACCTCTTCAGACTCAGCCTTGCCAATGTCTCTCTTCTTCAGGCCACAGAGTGGGCCTCCAGTGAGGACACG CGCCGCTCCTGCCTSLAAGCAAAGGGAAGACTGAGGAGGAGTGTCAGAACTACGTGCGAGTCCTGATCGTCGCCG GCCGGAAGGTGTTCATGTGTGGAACCAATGCCTTTTCCCCCATGTGCACCAGCAGACAGGTGGGGAACCTCAG CCGGACTACTGAGAAGATCAATGGTGTGGCCCGCTGCCCCTATGACCCACGCCACAACTCCACAGCTGTCATC TCCTCCCAGGGGGAGCTCTATGCAGCCACGGTCATCGACTTCTCAGGTCGGGACCCTGCCATCTACCGCAGCC TGGGCAGTGGGCCACCGCTTCGCACTGCCCAATATAACTCCAAGTGGCTTAATGAGCCAAACTTCGTGGCAGC CTATGATATTGGGCTGTTTGCATACTTCTTCCTGCGGGAGAACGCAGTGGAGCACGACTGTGGACGCACCGTG TACTCTCGCGTGGCCCGCGTGTGCAAGAATGACGTGGGGGGCCGATTCCTGCTGGAGGACACATGGACCACAT TCATGAAGGCCCGGCTCAACTGCTCCCGCCCGGGCGAGGTCCCCTTCTACTATAACGAGCTGCAGAGTGCCTT CCACTTGCCAGAGCAGGACCTCATCTATGGAGTTTTCACAACCAACGTAAACAGCATCGCGGCTTCTGCTGTC TGCGCCTTCAACCTCAGTGCTATCTCCCAGGCTTTCAATGGCCCATTTCGCTACCAGGAGAACCCCAGGGCTG CCTGGCTCCCCATAGCCAACCCCATCCCCAATTTCCAGTGTGGCACCCTGCCTGAGACCGGTCCCAACGAGAA CCTGACGGAGCGCAGCCTGCAGGACGCGCAGCGCCTCTTCCTGATGAGCGAGGCCGTGCAGCCGGTGACACCC GAGCCCTGTGTCACCCAGGACAGCGTGCGCTTCTCACACCTCGTGGTGGACCTGGTGCAGGCTAAAGACACGC TCTACCATGTACTCTACATTGGCACCGAGTCGGGCACCATCCTGAAGGCGCTGTCCACGGCGAGCCGCAGCCT CCACGGCTGCTACCTGGAGGAGCTGCACGTGCTGCCCCCCGGGCGCCGCGAGCCCCTGCGCAGCCTGCGCATC CTGCACAGCGCCCGCGCGCTCTTCGTGGGGCTGAGAGACGGCGTCCTGCGGGTCCCACTGGAGAGGTGCGCCG CCTACCGCAGCCAGGGGGCATGCCTGGGGGCCCGGGACCCGTACTGTGGCTGGGACGGGAAGCAGCAACGTTG CAGCACACTCGAGGACAGCTCCAACATGAGCCTCTGGACCCAGAACATCACCGCCTGTCCTGTGCGGAATGTG ACACGGGATGGGGGCTTCGGCCCATGGTCACCATGGCAACCATGTGAGCACTTGGATGGGGACAACTCAGGCT CTTGCCTGTGTCGAGCTCGATCCTGTGATTCCCCTCGACCCCGCTGTGGGGGCCTTGACTGCCTGGGGCCAGC CATCCACATCGCCAACTGCTCCAGGAATGGGGCGTGGACCCCGTGGTCATCGTGGGCGCTGTGCAGCACGTCC TGTGGCATCGGCTTCCAGGTCCGCCAGCGAAGTTGCAGCAACCCTGCTCCCCGCCACGGGGGCCGCATCTGCG TGGGCAAGAGCCGGGAGGAACGGTTCTGTAATGAGAACACGCCTTGCCCGGTGCCCATCTTCTGGGCTTCCTG GGGCTCCTGGAGCAAGTGCAGCAGCAACTGTGGAGGGGGCATGCAGTCGCGGCGTCGGGCCTGCGAGAACGGC AACTCCTGCCTGGGCTGCGGCGTGGAGTTCAAGACGTGCAACCCCGAGGGCTGCCCCGAAGTGCGGCGCAACA CCCCCTGGACGCCGTGGCTGCCCGTGAACGTGACGCAGGGCGGGGCACGGCAGGAGCAGCGGTTCCGCTTCAC CTGCCGCGCGCCCCTTGCAGACCCGCACGGCCTGCAGTTCGGCAGGAGAAGGACCGAGACGAGGACCTGTCCC GCGGACGGCTCCGGCTCCTGCGACACCGACGCCCTGGTGGAGGACCTCCTGCGCAGCGGGAGCACCTCCCCGC ACACGGTGAGCGGGGGCTGGGCCGCCTGGGGCCCGTGGTCGTCCTGCTCCCGGGACTGCGAGCTGGGCTTCCG CGTCCGCAAGAGAACGTGCACTAACCCGGAGCCCCGCAACGGGGGCCTGCCCTGCGTGGGCGATGCTGCCGAG TACCAGGACTGCAACCCCCAGGCTTGCCCAGTTCGGGGTGCTTGGTCCTGCTGGACCTCATGGTCTCCATGCT CAGCTTCCTGTGGTGGGGGTCACTATCAACGCACCCGTTCCTGCACCAGCCCCGCACCCTCCCCAGAAGGCTG GTCGCCCTGGTCTGAGTGGAGTAAGTGCACTGACGACGGAGCCCAGAGCCGAAGCCGGCACTGTGAGGAGCTC CTCCCAGGGTCCAGCGCCTGTGCTGGAAACAGCAGCCAGAGCCGCCCCTGCCCCTACAGCGAGATTCCCGTCA TCCTGCCAGCCTCCAGCATGGAGGAGGCCACCGGCTGTGCAGGGTTCAATCTCATCCACTTGGTGGCCACGGG CATCTCCTGCTTCTTGGGCTCTGGGCTCCTGACCCTAGCAGTGTACCTGTCTTGCCAGCACTGCCAGCGTCAG TCCCAGGAGTCCACACTGGTCCATCCTGCCACCCCCAACCATTTGCACTACAAGGGCGGAGGCACCCCGAAGA ATGAAAAGTACACACCCATGGAATTCAAGACCCTGAACAAGAATAACTTGATCCCTGATGACAGAGCCAACTT CTACCCATTGCAGCAGACCAATGTGTACACGACTACTTACTACCCAAGCCCCCTGAACAAACACAGCTTCCGG CCCGAGGCCTCACCTGGACAACGGTGCTTCCCCAACAGCTGATACCGCCGTCCTGGGGACTTGGGCTTCTTGC CTTCATAAGGCACAGAGCAGATGGAGATGGGACAGTGGAGCCAGTTTGGTTTCT
Figure imgf000118_0001
Figure imgf000119_0001
Figure imgf000119_0002
NOV2d, 209829553 SEQ ID NO: 12 401 aa MW at43246.4kD Protein Sequence
GSGPWSPWQPCEHLDGDNSGSCLCRARSCDSPRPRCGGLDCLGPAIHIANCSRNGAWXPWSSWALCSXSCGIG FQVRQRSCSNPAPRHGGRICVGKSREERFCNENXPCPVPIF ASWGSWSKCSSNCGGGMQSRRRACENGNSC GCGVEF XCNPEGCPEVRRNXP XP LPVNVXQGGARQEQRFRFXCRAPLADPHGLQFGRRRXEXRTCPADGS GSCDXDALVEDLLRSGSXSPHXVSGGWAAWGP SSCSRDCELGFRVRKRXCXNPESRNGGLPCVGDAAEYQDC NPQACPVRGA SC XSWSPCSASCGGGHYQRXRSCXSPAPSPGEDICLGLHXEEALCAXQACPEGWSP SE S KCXDDGAQSRSRHCEELLPGSSACAGNSSQSRPCVD
Figure imgf000120_0002
Figure imgf000120_0001
|NOV2f, 209829670 SEQ ID NO: 15 1203 bp |DNA Sequence ORF Start: at 1 JORF Stop: end of sequence
GGAXCCGGCCCAXGGXCACCAXGGCAACCAXGXGAGCACXXGGAXGGGGACAACXCAGGCXCXXGCCXGXGXC GAGCXCGAXCCXGXGAXXCCCCXCGACCCCGCXGTGGGGGCCXTGACXGCCXGGGGCCAACCAXCCACAXCGC CAACXGCXCCAGGAAXGGGGCGXGGACCCCGXGGXCAXCGXGGGCGCXGXGCAGCACGXCCXGXGGCAXCGGC XXCCAGGXCCGCCAGCGAAGXXGCAGCAACCCXGCXCCCCGCCACGGGGGCCGCAXCXGCGXGGGCAAGAGCC GGGAGGAACGGXXCXGXAAIGAGAACACGCCXXGCCCGGXGCCCAXCXXCXGGGCXXCCXGGGGCXCCXGGAG CAAGXGCGGCAGCAACXGXGGAGGGGGCAXGCAGICGCGGCGXCGGGCCXGCGAGAACGGCAACXCCXGCCXG GGCXGCGGCGTGGAGXXCAAGACGIGCAACCCCGAGGGCXGCCCCGAAGXGCGGCGCAACACCCCCXGGACGC CGXGGCXGCCCGXGAACGXGACGCAGGGCGGGGCACGGCAGGAGCAGCGGXXCCGCXXCACCTGCCGCGCGCC CCIXGCAGACCCGCACGGCCXGCAGXXCGGCAGGAGAAGGACCGAGACGAGGACCXGXCCCGCGGACGGCXCC GGCXCCXGCGACACCGACGCCCXGGXGGAGGXCCXCCXGCGCAGCGGGAGCACCXCCCCGCACACGGXGAGCG GGGGCXGGGCCGCCXGGGGCCCGXGGXCGXCCXGCXCCCGGGACXGCGAGCXGGGCXTCCGCGXCCGCAAGAG AACGXGCACXAACCCGGAGCCCCGCAACGGGGGCCTGCCCXGCGTGGGCGAXGCXGCCGAGTACCAGGACXGC AACCCCCAGGCTXGCCCAGTXCGGGGXGCXXGGTCCXGCXGGACCXCAIGGXCXCCAXGCXCAGCXXCCXGXG GXGGGGGXCACXAXCAACGCACCCGXXCCTGCACCAGCCCCGCACCCTCCCCAGGXGAGGACAXCXGXCXCGG GCXGCACACGGAGGAGGCACXAXGTGCCACACAGGCCTGCCCAGAAGGCXGGXCGCCCXGGTCXGAGXGGAGX AAGXGCACXGACGACGGAGCCCAGAGCCGAAGCCGGCACXGXGAGGAGCXCCXCCCAGGGXCCAGCGCCXGXG CXGGAAACAGCAGCCAGAGCCGCCCCXGCGTCGAC
Figure imgf000121_0001
|NOV2g7cG106951-02 SEQ ID NO: 17 Ϊ4233 bp
DNA Sequence j ORF Start: ATG at 2 pRF Stop: TGA at 3281
CATGGXGCXTGCAGGCCCCCXGGCXGXCXCGCXGTXGCXGCCCAGCCXCACACXGCXGGXGTCCCACCXCXCC AGCXCCCAGGATGXCXCCAGXGAGCCCAGCAGXGAGCAGCAGCXGXGCGCCCTXAGCAAGCACCCCACCGXGG CCXXXGAAGACCXGCAGCCGXGGGXCXCXAACXXCACCXACCCXGGAGCCCGGGAXXXCXCCCAGCXGGCXXX GGACCCCXCCGGGAACCAGCXCAXCGXGGGAGCCAGGAACXACCXCXXCAGACXCAGCCXXGCCAAXGXCTCX CXXCXXCAGGCCACAGAGXGGGCCXCCAGXGAGGACACGCGCCGCXCCXGCCAAAGCAAAGGGAAGACXGAGG AGGAGXGXCAGAACXACGXGCGAGXCCXGAXCGXCGCCGGCCGGAAGGXGXXCAXGXGXGGAACCAAXGCCXI XXCCCCCATGTGCACCAGCAGACAGGXGGGGAACCICAGCCGGACXACXGAGAAGATCAAXGGTGXGGCCCGC XGCCCCXAXGACCCACGCCACAACXCCACAGCXGTCAXCXCCXCCCAGGGGGAGCXCXAXGCAGCCACGGXCA XCGACXXCTCAGGXCGGGACCCXGCCAXCXACCGCAGCCXGGGCAGXGGGCCACCGCXXCGCACXGCCCAAXA XAACXCCAAGXGGCXXAAXGAGCCAAACXXCGXGGCAGCCXAXGAXAXXGGGCXGXXXGCAXACXXCXXCCXG CGGGAGAACGCAGXGGAGCACGACXGXGGACGCACCGXGXACXCXCGCGXGGCCCGCGXGXGCAAGAAXGACG XGGGGGGCCGAXXCCXGCXGGAGGACACAXGGACCACAXXCAXGAAGGCCCGGCXCAACXGCXCCCGCCCGGG CGAGGXCCCCXXCXACXAXAACGAGCXGCAGAGXGCCXXCCACTXGCCAGAGCAGGACCXCAXCXAXGGAGIX XXCACAACCAACGXAAACAGCAXCGCGGCXXCXGCXGXCXGCGCCXXCAACCTCAGXGCXAXCXCCCAGGCXX XCAAXGGCCCAXXXCGCXACCAGGAGAACCCCAGGGCXGCCXGGCXCCCCAXAGCCAACCCCAXCCCCAATXX CCAGTGXGGCACCCXGCCTGAGACCGGXCCCAACGAGAACCTGACGGAGCGCAGCCXGCAGGACGCGCAGCGC CXCXXCCXGATGAGCGAGGCCGXGCAGCCGGXGACACCCGAGCCCXGXGXCACCCAGGACAGCGXGCGCTXCX CACACCXCGTGGXGGACCXGGXGCAGGCXAAAGACACGCXCXACCAXGXACXCXACAXXGGCACCGAGXCGGG CACCAXCCTGAAGGCGCXGXCCACGGCGAGCCGCAGCCXCCACGGCXGCXACCXGGAGGAGCXGCACGXGCXG CCCCCCGGGCGCCGCGAGCCCCXGCGCAGCCXGCGCATCCXGCACAGCGCCCGCGCGCXCTTCGXGGGGCXGA GAGACGGCGTCCXGCGGGXCCCACXGGAGAGGIGCGCCGCCXACCGCAGCCAGGGGGCAXGCCXGGGGGCCCG GGACCCGXACXGXGGCXGGGACGGGAAGCAGCAACGXXGCAGCACACICGAGGACAGCXCCAACAXGAGCCXC XGGACCCAGAACAXCACCGCCXGXCCXGXGCGGAAXGXGACACGGGAXGGGGGCXXCGGCCCATGGTCACCAX GGCAACCATGTGAGCACXXGGAXGGGGACAACXCAGGCXCXTGCCXGXGXCGAGCXCGAXCCXGTGAXXCCCC XCGACCCCGCTGTGGGGGCCXXGACTGCCXGGGGCCAGCCATCCACAXCGCCAACTGCXCCAGGAAXGGGGCG XGGACCCCGXGGXCAXCGXGGGCGCTGXGCAGCACGXCCXGXGGCAXCGGCXTCCAGGXCCGCCAGCGAAGTX GCAGCAACCCXGCTCCCCGCCACGGGGGCCGCAXCXGCGXGGGCAAGAGCCGGGAGGAACGGXXCXGXAAXGA GAACACGCCXTGCCCGGXGCCCAXCTXCXGGGCXTCCXGGGGCXCCXGGAGCAAGXGCAGCAGCAACXGXGGA GGGGGCAXGCAGXCGCGGCGXCGGGCCXGCGAGAACGGCAACXCCXGCCXGGGCXGCGGCGXGGAGTXCAAGA CGXGCAACCCCGAGGGCXGCCCCGAAGXGCGGCGCAACACCCCCXGGACGCCGXGGCXGCCCGXGAACGTGAC GCAGGGCGGGGCACGGCAGGAGCAGCGGXXCCGCTXCACCXGCCGCGCGCCCCXXGCAGACCCGCACGGCCXG CAGXXCGGCAGGAGAAGGACCGAGACGAGGACCXGXCCCGCGGACGGCXCCGGCXCCXGCGACACCGACGCCC XGGXGGAGGACCXCCIGCGCAGCGGGAGCACCXCCCCGCACACGGXGAGCGGGGGCXGGGCCGCCXGGGGCCC GXGGXCGXCCTGCXCCCGGGACXGCGAGCXGGGCTXCCGCGTCCGCAAGAGAACGTGCACXAACCCGGAGCCC CGCAACGGGGGCCXGCCCXGCGXGGGCGAXGCXGCCGAGXACCAGGACXGCAACCCCCAGGCXXGCCCAGXXC GGGGXGCXXGGXCCTGCXGGACCXCAXGGXCXCCAXGCXCAGCXXCCXGXGGTGGGGGXCACXAXCAACGCAC CCGXXCCXGCACCAGCCCCGCACCCTCCCCAGGXGAGGACATCIGXCXCGGGCXGCACACGGAGGAGGCACXA XGXGCCACACAGGCCXGCCCAGAAGGCXGGXCGCCCXGGXCXGAGXGGAGXAAGXGCACXGACGACGGAGCCC AGAGCCGAAGCCGGCACXGXGAGGAGCXCCXCCCAGGGXCCAGCGCCXGXGCTGGAAACAGCAGCCAGAGCCG CCCCIGCCCCTACAGCGAGAXXCCCGXCAXCCXGCCAGCCXCCAGCAXGGAGGAGGCCACCGGCXGXGCAGGG TXCAAXCXCATCCACXXGGXGGCCACGGGCAXCXCCXGCXXCXXGGGCXCXGGGCXCCXGACCCXAGCAGXGX ACCXGXCXXGCCAGCACXGCCAGCGTCAGXCCCAGGAGXCCACACXGGXCCATCCXGCCACCCCCAACCAXTX GCACXACAAGGGCGGAGGCACCCCGAAGAAXGAAAAGXACACACCCAXGGAATXCAAGACCCXGAACAAGAAX AACXXGAXCCCXGAXGACAGAGCCAACIXCTACCCAXXGCAGCAGACCAAXGTGIACACGACXACXXACTACC CAAGCCCCCXGAACAAACACAGCXXCCGGCCCGAGGCCXCACCXGGACAACGGXGCXXCCCCAACAGCTGAXA CCGCCGXCCXGGGGACXXGGGCXXCXXGCCXXCAXAAGGCACAGAGCAGAXGGAGAXGGGACAGXGGAGCCAG
IXTXGGXXXXCXCCCXCXGCACXAGGCCAAGAACXXGCXGCCXXGCCXGXGGGGGGXCCCAXCCGGCXXCAGAG lAGCXCXGGCTGGCAXXGACCAXGGGGGAAAGGGCXGGXXXCAGGCXGACAXAXGGCCGCAGGXCCAGXXCAGC
CCAGGXCXCXCAXGGXXAXCXXCCAACCCACXGXCACGCXGACACXAXGCXGCCAXGCCXGGGCXGXGGACCT
ACXGGGCAXTTGAGGAACXGGAGAAXGGAGAXGGCAAGAGGGCAGGCXXXXAAGXXXGGGXXGGAGACAACXX
CCXGXGGCCCCCACAAGCXGAGXCXGGCCXXCXCCAGCTGGCCCCAAAAAAGGCCXXXGCXACAXCCXGAXXA
TCXCXGAAAGXAAXCAAXCAAGXGGCXCCAGXAGCXCTGGAXXXXCXGCCAGGGCXGGGCCAXXGXGGXGCXG
CCCCAGXAXGACAXGGGACCAAGGCCAGCGCAGGXXAXCCACCXCXGCCXGGAAGXCXATACXCXACCCAGGG
CAXCCCXCXGGXCAGAGGCAGXGAGXACXGGGAACXGGAGGCXGACCXGXGCXXAGAAGTCCXXXAAXCXGGG
CXGGXACAGGCCXCAGCCXXGCCCXCAAXGCACGAAAGGXGGCCCAGGAGAGAGGAXCAAXGCCACAGGAGGC lAGAAGXCXGGCCXCXGXGCCXCXAXGGAGACXAXCXXCCAGXXGCXGCXCAACAGAGXXGXXGGCXGAGACCX
GCXXGGGAGTCTCXGCXGGCCCXXCAXCXGXXCAGGAACACACACACACACACACXCACACACGCACACACAA
XCACAAXXXGCXACAGCAACAAAAAAGACATXGGGCXGXGGCAXXAXXAAXXAAAGAXGAXAXCCAGXCXCC
NOV2g, CG106951-02 SEQ ID NO: 18 1093 aa MW at ll9865.3kD Protein Sequence
MVLAGPLAVSLLLPSLXLLVSHLSSSQDVSSEPSSEQQLCALSKHPXVAFEDLQPWVSNFXYPGARDFSQLAL DPSGNQLIVGARNYLFRLSLANVSLLQAXEWASSEDXRRSCQSKGKXEEECQNYVRVL.IVAGRKVFMCGXNAF SPMCXSRQVGNLSRXXEKINGVARCPYDPRHNSXAVISSQGELYAAXVIDFSGRDPAIYRSLGSGPP RXAQY NSKWLNEPNFVAAYDIGLFAYFFLRENAVEHDCGRXVYSRVARVCKNDVGGRFLLEDXWXXFMKARLNCSRPG EVPFYYNELQSAFH PEQDLIYGVFXXNV SIAASAVCAFNLSAISQAFNGPFRYQENPRAAWLPIANPIPNF QCGXLPEXGPNENLXERSLQDAQRLFL SEAVQPVXPEPCVXQDSVRFSHLWDLVQAKDXLYHVLYIGXESG XILKALSXASRSLHGCYLEELHVLPPGRREPLRSLRILHSARALFVGLRDGVLRVPLERCAAYRSQGACLGAR DPYCGWDGKQQRCSXLEDSSNMSLWXQNIXACPVRNVXRDGGFGP SPWQPCEHLDGDNSGSC CRARSCDSP RPRCGGLDC GPAIHIANCSRNGA XP SSWALCSXSCGIGFQVRQRSCSNPAPRHGGRICVGKSREERFCNE NXPCPVPIFWASWGS SKCSSNCGGGMQSRRRACENGNSCLGCGVEFKXCNPEGCPEVRRNXPWXPWLPVNVX QGGARQEQRFRFXCRAPLADPHGLQFGRRRXEXRTCPADGSGSCDXDALVEDLLRSGSXSPHXVSGGAA GP SSCSRDCELGFRVRKRXCXNPEPRNGG PCVGDAAEYQDCNPQACPVRGA SCWTS SPCSASCGGGHYQRX RSCXSPAPSPGEDIC GLHXEEALCAXQACPEG SPWSE SKCXDDGAQSRSRHCEELLPGSSACAGNSSQSR PCPYSEIPVILPASS EEAXGCAGFNLIHLVAXGISCFLGSGLLXLAVYLSCQHCQRQSQESXLVHPAXP HL HYKGGGXPKNEKYXPMEFKXLNKNNLIPDDRANFYPLQQXNVYXXXYYPSPLNKHSFRPEASPGQRCFPNS
|NOV2h, CG106951-03 [SEQ ID NO: 19 1203 bp
DNA Sequence ORF Start: at 7 ORF Stop: at 1198
GGAXCCGGCCCAXGGXCACCAXGGCAACCAXGXGAGCACXXGGAXGGGGACAACXCAGGCXCXXGCCXGXGXC
GAGCXCGAXCCXGXGAXXCCCCXCGACCCCGCXGXGGGGGCCXXGACXGCCXGGGGCCAACCAXCCACAXCGC CAACXGCXCCAGGAAXGGGGCGXGGACCCCGXGGXCAXCGXGGGCGCXGXGCAGCACGXCCXGXGGCAXCGGC XXCCAGGXCCGCCAGCGAAGXXGCAGCAACCCXGCXCCCCGCCACGGGGGCCGCAXCXGCGXGGGCAAGAGCC GGGAGGAACGGXXCXGXAAXGAGAACACGCCXXGCCCGGXGCCCAXCXXCXGGGCXXCCXGGGGCTCCXGGAG CAAGXGCGGCAGCAACXGXGGAGGGGGCAXGCAGTCGCGGCGXCGGGCCXGCGAGAACGGCAACXCCXGCCXG GGCXGCGGCGXGGAGXXCAAGACGXGCAACCCCGAGGGCXGCCCCGAAGXGCGGCGCAACACCCCCXGGACGC CGXGGCXGCCCGXGAACGXGACGCAGGGCGGGGCACGGCAGGAGCAGCGGXXCCGCXXCACCXGCCGCGCGCC CCXXGCAGACCCGCACGGCCXGCAGXXCGGCAGGAGAAGGACCGAGACGAGGACCXGXCCCGCGGACGGCXCC GGCXCCXGCGACACCGACGCCCXGGXGGAGGXCCTCCXGCGCAGCGGGAGCACCXCCCCGCACACGGXGAGCG GGGGCXGGGCCGCCXGGGGCCCGXGGXCGXCCXGCXCCCGGGACXGCGAGCXGGGCXXCCGCGXCCGCAAGAG AACGXGCACXAACCCGGAGCCCCGCAACGGGGGCCXGCCCXGCGXGGGCGAXGCXGCCGAGXACCAGGACXGC AACCCCCAGGCXXGCCCAGTXCGGGGXGCXXGGXCCXGCXGGACCXCAXGGXCXCCAXGCXCAGCXXCCXGXG GXGGGGGXCACTAXCAACGCACCCGXXCCXGCACCAGCCCCGCACCCXCCCCAGGXGAGGACAXCXGXCXCGG GCXGCACACGGAGGAGGCACXAXGXGCCACACAGGCCXGCCCAGAAGGCXGGXCGCCCXGGXCXGAGXGGAGX AAGXGCACXGACGACGGAGCCCAGAGCCGAAGCCGGCACXGXGAGGAGCXCCXCCCAGGGXCCAGCGCCXGIG CXGGAAACAGCAGCCAGAGCCGCCCCXGCGXCGAC
Figure imgf000123_0001
|NOV2i, SNP13382456 of |SEQ ID O: 21 ]6408 bp
CGI 06951-01, DNA Sequence ORF Start: ATG at 1400 ORF Stop: TGA at 5456
SNP Pos: 5770 SNP Change: C to T
CCXGGGACXCXGGGAGAAXGGXCCAGAGCXCAXXGXCCXXGXXGAXAAAAXGAXAGAXIXGGACXCAAXAXCC
CAXGCXGCCXCXXCCAACXXGAXXXXXACCCCAGACXGGGCXACCAGACXGGXAXGCCCACACAXGCCCGXXX
CCXXXCXXXXCXXCXCTGCAXCXCXGCCXXTGXGXCCAGAGCGXGXXXXCCCXXXGCAAGXTXCXCXCCAXXC iXGCACAXXAXGAGXXXCAGCAXXXCXGXXGCCCXAGAAAGXCXAXCXXXGAGAXCXXGCACTGXXXCXCXXXX
XACAGXGXCXCAXAAACXCCCXXCXXGGAXTCAGAACCACCCXXXCXXTCCCAXXAXCCXGTCAAACXGCXXC
XTGCCAXGGXCCAGGGGXAGGAGGAXGGCAGGCAGGAGGTGCXXCXCXGGGGCXCXXAGXGTCXCAAXXCXXC
IXGCXTXAXCXGGGXXXTCCXXXACCCAGAAXXXXAXXAXGXAAAAXGCXXCACXCAGACXXTGTXCXAAXXAX
CCAAXXXXXGGCAXACTCXAGAAAGXCXXXXGAXAXIXXCCXXCCXCCAACXXATCXAXXXTXAXTXCAXAGX
IXCXCXXXGGXXAXCXCTTAGAAXCACACXXTCCXGGXXXXAAXXXXXCAAAXCCTXXGXCXTXCXCACXCGXX
CITAGGXCACCXXXXXXXACAXXXXCAAAXAXAXXXXXXGXXCAGCAGAGGGCXCCCXXCCCAXCCCXCXXGC
AGCCCGGGCAGCXAGGAXXXGAAGCTXGCCCCXXGAAXCXXXCXCXCCCGCCXXCXAGCCATCAGAAACACXA
GAXCACXXAAACXXGXAAACAAXXCGGCCXCGCXCCXXGXGAXTGCGCXAAACCTXCCGXCCXCAGCXGAGAA
CGCXCCACCACCXCCCCGGAXCGCXCAXCXCTXGGCXGCCCXCCCACXGXTCCXGAXGXXATXIXACXCCCCG iXAXCCCCXACXCGXXCXXCACAAXXCXGXAGGGXGCGXATXACXAACCCCAGXXTACAGCXGAGGAAACXGAG
GCXXGGAGAGGXXCGCXCGGXAXCGXACAGXXXGCAAGGXXAACCCXAAXCCGGCCAGXXCTGGCXXXCCAGC
CCAGCCCAGCAGCCXAGCCXCCCXCTCXGCCGCXGCAGGIXAXAACGGCXCXCCCCCGXXXTACACGAGGXCC
CXXCCCCXXCAAAXCCACAGGCAGGAAGAXCGXXCCGAACXGACGGGGCXGGGGAAXGXGGGAGXCCGGAGXG iGGGXTXGGGGGAGCXXCCXCAGGCCCXGAGTGXXGGGGXGGGCAGGCCGCGCCGAXGGCCCTCGGGGAXGXCA
CAXXCGAGAXGGGGXGACCGAGAACGGCAAGGCGGGAXGXGGCAAACGGCGGCAAGXGCTCGGAGXCCXAGGX
CXXGCCGCCGGAATGCCGGCCGGGGAAGGGGCXXCGGCCCACCGGGCXGGXCACCACACXCGGCAGGCCCGGG
GCGGGAGXCGGCCGAGCAGCCGCGGGAXGCAGGGCGCCCCCXCGCGCXCCXCCGCGCGCCXCGAGGCXGGCGG GXGCAGCGCCCGCCGCGGCAGGXCXGCXCCAGCCCCCXCCXCXXXXTCGCXCCCGCXCCCCTCCXXCXCXCCC XXXGCXXGCAACXCCXCCCCCACCGCCCCCXCCCXCCXXCXGCXCCCGCGGICXCCXCCXCCCXGCXCXCXCC GAGCGCCGGGXCGGGAGCXAGXXGGAGCGCGGGGGXXGGXGCCAGAGCCCAGCXCCGCCGAGCCGGGCGGGXC GGCAGCGCAXCCAGCGGCXGCXGGGAGCCCGAGCGCAGCGGGCGCGGGCCCGGGTGGGGACTGCACCGGAGCG CXGAGAGCTGGAGGCCGXXCCXGCGCGGCCGCCCCAXXCCCAGACCGGCCGCCAGCCCAXCTGGXXAGCXCCC GCCGCXCCGCGCCGCCCGGGAGXCGGGAGCCGCGGGGAACCGGGCACCXGCACCCGCCXCXGGGAGXGAGXGG XXCCAGCXGGXGCCXGGCCXGXGXCXCXXGGAXGCCCXGXGGCXXCAGXCCGXCXCCXGXXGCCCACCACCXC GXCCCXGGGCCGCCXGAXACCCCAGCCCAACAGCXAAGGXGXGGAXGGACAGXAGGGGGCXGGCXXCXCXCAC XGGXCAGGGGTCXXCXCCCCXGXCXGCCXCCCGGAGCXAGGACXGCAGAGGGGCCXAXCAXGGXGCXXGCAGG CCCCCXGGCXGXCXCGCXGXXGCXGCCCAGCCXCACACXGCXGGXGXCCCACCXCXCCAGCTCCCAGGAXGXC XCCAGXGAGCCCAGCAGXGAGCAGCAGCXGXGCGCCCXXAGCAAGCACCCCACCGXGGCCXTXGAAGACCXGC AGCCGXGGGXCXCXAACXXCACCXACCCXGGAGCCCGGGAXXXCXCCCAGCXGGCXXXGGACCCCXCCGGGAA CCAGCXCATCGXGGGAGCCAGGAACXACCXCTXCAGACXCAGCCXXGCCAATGXCXCXCXXCXXCAGGCCACA GAGXGGGCCXCCAGXGAGGACACGCGCCGCXCCXGCCAAAGCAAAGGGAAGACXGAGGAGGAGXGXCAGAACX ACGXGCGAGXCCXGAXCGXCGCCGGCCGGAAGGXGXXCAXGXGXGGAACCAAXGCCXXXXCCCCCAXGXGCAC CAGCAGACAGGXGGGGAACCXCAGCCGGACXACXGAGAAGAXCAAXGGXGXGGCCCGCXGCCCCXAXGACCCA CGCCACAACXCCACAGCXGXCAXCXCCTCCCAGGGGGAGCXCXAXGCAGCCACGGXCAXCGACXXCXCAGGXC GGGACCCXGCCAXCXACCGCAGCCXGGGCAGTGGGCCACCGCXXCGCACXGCCCAAXAXAACXCCAAGXGGCX XAAXGAGCCAAACXXCGXGGCAGCCXAXGAXAXXGGGCXGXXXGCAXACXXCXXCCXGCGGGAGAACGCAGXG GAGCACGACXGXGGACGCACCGXGXACXexCGCGXGGCCCGCGXGXGCAAGAAXGACGXGGGGGGCCGAXXCC XGCXGGAGGACACAXGGACCACAXXCAXGAAGGCCCGGCXCAACXGCXCCCGCCCGGGCGAGGXCCCCXXCXA CXAXAACGAGCXGCAGAGXGCCTXCCACXXGCCAGAGCAGGACCXCAXCXAXGGAGXXXXCACAACCAACGXA AACAGCAXCGCGGCXXCXGCXGXCXGCGCCXXCAACCXCAGTGCXATCXCCCAGGCXXXCAAXGGCCCAXXTC GCXACCAGGAGAACCCCAGGGCXGCCXGGCXCCCCAXAGCCAACCCCAXCCCCAAXXTCCAGXGXGGCACCCX GCCXGAGACCGGXCCCAACGAGAACCXGACGGAGCGCAGCCXGCAGGACGCGCAGCGCCXCXXCCXGAXGAGC GAGGCCGXGCAGCCGGXGACACCCGAGCCCXGXGTCACCCAGGACAGCGXGCGCXXCTCACACCXCGXGGXGG ACCXGGIGCAGGCXAAAGACACGCXCXACCAXGXACXCXACAXXGGCACCGAGXCGGGCACCAXCCXGAAGGC GCXGXCCACGGCGAGCCGCAGCCXCCACGGCXGCTACCXGGAGGAGCTGCACGTGCXGCCCCCCGGGCGCCGC GAGCCCCXGCGCAGCCTGCGCAXCCXGCACAGCGCCCGCGCGCXCXXCGXGGGGCXGAGAGACGGCGXCCXGC GGGXCCCACXGGAGAGGXGCGCCGCCXACCGCAGCCAGGGGGCAXGCCIGGGGGCCCGGGACCCGXACXGXGG CXGGGACGGGAAGCAGCAACGXXGCAGCACACXCGAGGACAGCXCCAACAXGAGCCXCXGGACCCAGAACAXC ACCGCCXGTCCXGXGCGGAAXGXGACACGGGAXGGGGGCXXCGGCCCAXGGXCACCAXGGCAACCAXGXGAGC ACXXGGAXGGGGACAACXCAGGCXCXXGCCXGXGXCGAGCXCGAXCCXGXGATXCCCCXCGACCCCGCXGXGG GGGCCXXGACXGCCXGGGGCCAGCCAXCCACAXCGCCAACXGCXCCAGGAAXGGGGCGTGGACCCCGXGGXCA XCGXGGGCGCXGXGCAGCACGXCCXGXGGCAXCGGCXXCCAGGXCCGCCAGCGAAGXXGCAGCAACCCXGCXC CCCGCCACGGGGGCCGCAXCXGCGXGGGCAAGAGCCGGGAGGAACGGTXCXGTAAXGAGAACACGCCXTGCCC GGXGCCCAXCXXCXGGGCXXCCXGGGGCXCCXGGAGCAAGXGCAGCAGCAACXGXGGAGGGGGCAXGCAGXCG CGGCGXCGGGCCXGCGAGAACGGCAACXCCXGCCXGGGCXGCGGCGXGGAGXTCAAGACGXGCAACCCCGAGG GCXGCCCCGAAGIGCGGCGCAACACCCCCXGGACGCCGXGGCXGCCCGXGAACGXGACGCAGGGCGGGGCACG GCAGGAGCAGCGGTXCCGCXXCACCXGCCGCGCGCCCCXXGCAGACCCGCACGGCCXGCAGXXCGGCAGGAGA AGGACCGAGACGAGGACCXGXCCCGCGGACGGCXCCGGCXCCXGCGACACCGACGCCCTGGXGGAGGACCXCC XGCGCAGCGGGAGCACCXCCCCGCACACGGXGAGCGGGGGCXGGGCCGCCXGGGGCCCGXGGXCGXCCXGCXC CCGGGACXGCGAGCXGGGCXXCCGCGXCCGCAAGAGAACGXGCACXAACCCGGAGCCCCGCAACGGGGGCCXG CCCXGCGXGGGCGAXGCXGCCGAGXACCAGGACXGCAACCCCCAGGCXXGCCCAGXXCGGGGXGCXXGGXCCX GCXGGACCXCAXGGXCXCCAXGCICAGCXXCCXGXGGXGGGGGXCACXAXCAACGCACCCGXXCCXGCACCAG CCCCGCACCCXCCCCAGGXGAGGACAXCXGXCXCGGGCXGCACACGGAGGAGGCACXATGXGCCACACAGGCC XGCCCAGAAGGCXGGXCGCCCXGGXCIGAGXGGAGXAAGXGCACXGACGACGGAGCCCAGAGCCGAAGCCGGC ACXGXGAGGAGCXCCXCCCAGGGXCCAGCGCCXGXGCXGGAAACAGCAGCCAGAGCCGCCCCXGCCCCXACAG CGAGAXXCCCGXCAXCCXGCCAGCCXCCAGCAXGGAGGAGGCCACCGGCXGXGCAGGGTXCAAXCXCAXCCAC XXGGXGGCCACGGGCAXCXCCXGCXXCXXGGGCXCXGGGCXCCXGACCCXAGCAGXGXACCXGXCXXGCCAGC ACXGCCAGCGXCAGXCCCAGGAGXCCACACXGGXCCAXCCXGCCACCCCCAACCAXXXGCACXACAAGGGCGG AGGCACCCCGAAGAAXGAAAAGXACACACCCAXGGAAXTCAAGACCCXGAACAAGAATAACXXGAXCCCXGAX GACAGAGCCAACXXCXACCCAXXGCAGCAGACCAAXGXGXACACGACXACIXACXACCCAAGCCCCCXGAACA AACACAGCXTCCGGCCCGAGGCCXCACCXGGACAACGGXGCXXCCCCAACAGCTGAXACCGCCGXCCXGGGGA CXXGGGCXXCXXGCCXXCAXAAGGCACAGAGCAGATGGAGAXGGGACAGXGGAGCCAGTXXGGXXXXCXCCCX CXGCACXAGGCCAAGAACXXGCXGCCXXGCCXGXGGGGGGTCCCAXCCGGCXXCAGAGAGCXCXGGCXGGCAX XGACCAXGGGGGAAAGGGCXGGXXTCAGGCXGACAXAXGGCCGCAGGXCCAGTXCAGCCCAGGXCXCXCATGG XXATCXXCCAACCCACXGXCACGCXGACACXAXGCXGCCAXGCCXGGGCXGXGGACCXACXGGGCAXXXGAGG AATTGGAGAAXGGAGAXGGCAAGAGGGCAGGCXXXXAAGXXXGGGXXGGAGACAACXXCCXGXGGCCCCCACA AGCTGAGXCTGGCCXTCXCCAGCXGGCCCCAAAAAAGGCCXXXGCXACAXCCXGAXXATCXCXGAAAGXAAXC AAXCAAGXGGCXCCAGXAGCXCXGGAXXXXCXGCCAGGGCXGGGCCAXXGXGGXGCTGCCCCAGXAXGACAXG GGACCAAGGCCAGCGCAGGXXAXCCACCXCXGCCXGGAAGXCXAXACXCXACCCAGGGCAXCCCTCXGGXCAG AGGCAGXGAGXACXGGGAACXGGAGGCXGACCXGXGCXXAGAAGXCCXXXAAXCXGGGCXGGXACAGGCCTCA GCCXXGCCCTCAAXGCACGAAAGGXGGCCCAGGAGAGAGGAXCAAXGCCACAGGAGGCAGAAGXCXGGCCTCX GXGCCXCXATGGAGACXAXCXXCCAGXXGCXGCXCAACAGAGXXGXXGGCXGAGACCXGCXXGGGAGXCXCTG CXGGCCCXXCAXCXGTXCAGGAACACACACACACACACACXCACACACGCACACACAATCACAATXXGCXACA GCAACAAAAAAGACATXGGGCTGXGGCAXXAXXAAXXAAAGAXGAXAXCCAGXCXCC
Figure imgf000125_0001
A ClustalW comparison of the above protein sequences yields the following sequence alignment shown in Table 2B.
Table 2B. Comparison of the NOV2 protein sequences.
NOV2a MPAGEGASAHRAGHHXRQARGGSRPSSRGMQGAPSRSSARLEAGGCSARRGRSAPAPSSF
NOV2b
NOV2c
NOV2d
NOV2e
NOV2f
NOV2g
NOV2h
NOV2a SLPLPSFSPFACNSSPXAPSLLL PRSPPPCSLRAPGRELVGARGLVPEPSSAEPGGSAA
NOV2b
NOV2c
NOV2d
NOV2e
NOV2f
NOV2g
NOV2h
NOV2a HPAAAGSPSAAGAGPGGDCXGALRAGGRSCAAAPFPDRPPAHLVSSRRSAPPGSREPRGX
NOV2b
NOV2c
NOV2d
NOV2e
NOV2f
NOV2g
NOV2h NOV2a GHLHPPLGVSGSS CLACVSWMPCGFSPSPVAHHLVPGPPDXPAQQLRCGWXVGG LLSL.
NOV2b MPCGFSPSPVAHHLVPGPPDXPAQQLRCG XVGG LLSL.
NOV2c
NOV2d
NOV2e
NOV2f
NOV2g
NOV2h
NOV2a VRG LPCLPPGARXAEGPI VLAGPLAVSL LPSLXLLVSHLSSSQDVSSEPSSEQQLCA
NOV2b VRGLLPCLPPGARXAEGPIMV AGPLAVSL LPSLXLLVSHLSSSQDVSSEPSSEQQLCA
NOV2C
NOV2d
NOV2e
NOV2f
NOV2g MVLAGPLAVSLL PSLXLLVSHLSSSQDVSSEPSSEQQLCA
NOV2h
NOV2a LSKHPXVAFEDLQP VSNFXYPGARDFSQLALDPSGNQLIVGARNYLFRLSLANVSLLQA
NOV2b LSKHPXVAFEDLQP VSNFXYPGARDFSQLALDPSGNQLIVGARNYLFRLSLANVS LQA
NOV2C
NOV2d
NOV2e
NOV2f
NOV2g LSKHPXVAFEDLQPWVSNFXYPGARDFSQLALDPSGNQLIVGARNYLFRLSLANVSLLQA
NOV2h
NOV2a XEWASSEDXRRSCQSKGKXEEECQNYVRVLIVAGRKVFMCGXNAFSPMCXSRQVGNLSRX
NOV2b TE ASSEDXRRSCQSKGKXEEECQNYVRVLIVAGRKVFMCGXNAFSP CTSRQVGNLSRX
NOV2c
NOV2d
N0V2e
NOV2f
NOV2g TEWASSEDXRRSCQSKGKXEEECQNYVRVLIVAGR VFMCGXNAFSPMCTSRQVGNLSRX
NOV2h
NOV2a XEKINGVARCPYDPRHNSXAVISSQGELYAAXVIDFSGRDPAIYRSLGSGPPLRXAQYNS
NOV2b XEK1NGVARCPYDPRHNSXAVISSQGE YAAXVIDFSGRDPAIYRSLGSGPPLRXAQYNS
NOV2c
NOV2d
NOV2e
NOV2f
NOV2g TEKINGVARCPYDPRHNSXAVISSQGELYAAXVIDFSGRDPAIYRSLGSGPPLRXAQYNS
NOV2h
NOV2a KWLNEPNFVAAYDIGLFAYFFLRENAVEHDCGRXVYSRVARVCKNDVGGRFLLEDXWXXF
NOV2b KWLNEPNFVAAYDIGLFAYFFLRENAVEHDCGRXVYSRVARVCKNDVGGRFLLEDXWXXF
NOV2c
NOV2d
NOV2e
NOV2f
NOV2g K LNEPNFVAAYDIGLFAYFF RENAVEHDCGRTVYSRVARVCKNDVGGRFLLEDXWXTF
N0V2h
NOV2a MKARLNCSRPGEVPFYYNELQSAFHLPEQDLIYGVFTXNVNSIAASAVCAFNLSAISQAF
NOV2b MKARLNCSRPGEVPFYYNELQSAFHLPEQD IYGVFTX VNSIAASAVCAFNLSAISQAF
NOV2C
NOV2d
NOV2e NOV2f ;
NOV2g MKARLNCSRPGEVPFYYNELQSAFHLPEQDLIYGVFXX VNSIAASAVCAF LSAISQAF
NOV2h
NOV2a NGPFRYQENPRAAWLPIANPIPNFQCGXLPEIGPNENLXERSLQDAQRLFLMSEAVQPVX
NOV2b NGPFRYQENPRAAWLPIANPIPNFQCGTLPEXGPNENLXERSLQDAQRLFLMSEAV/QPVX
NOV2C
NOV2d
NOV2e
NOV2f
NOV2g NGPFRYQENPRAAWLPIANPIPNFQCGXLPEXGPNENLXERSLQDAQRLFL SEAVQPVX
NOV2h
NOV2a PEPCVTQDSWFSHLWDLVQAKDXLYHVLYIGXESGXILKALSXASRSLHGCYLEELHV
NOV2b PEPCV QDSVRFSHLWDLVQA DXLYHV YIGIESGTILKALSXASRSLHGCYLEELHV
NOV2c
NOV2d
NOV2e
NOV2f
NOV2g PEPCVXQDSVRFSHLWD VQAKDXLYHVLYIGXESGXILKALSXASRSLHGCYLEELHV
NOV2h
NOV2a LPPGRREPLRSLRILHSARALFVGLRDGVLRVPLERCAAYRSQGACLGARDPYCGWDGKQ
NOV2b LPPGRREPLRSLRILHSARALFVGLRDGVLRVPLERCAAYRSQGACLGARDPYCGWDGKQ
NOV2c
NOV2d
NOV2e
NOV2f
NOV2g LPPGRREPLRSLRILHSARALFVGLRDGVL.RVPLERCAAYRSQGACLGARDPYCG DGKQ
NOV2h
NOV2a QRCSXLEDSSNMSL XQNIXACPVRNVXRDGGFGPWSPWQPCEHLDGDNSGSCLCRARSC
NOV2b QRCSXLEDSSNMSL XQNIXACPVRNVXRDGGFGPWSP QPCEHLDGDNSGSCLCRARSC
NOV2c GSGP SP QPCEHLDGDNSGSCLCRARSC
NOV2d GSGPWSPWQPCEHLDGDNSGSCLCRARSC
NOV2e GSGPWSPWQPCEHLDGDNSGSCLCRARSC
NOV2f GSGPWSPWQPCEHLDGDNSGSCLCRARSC
NOV2g QRCSXLEDSSNMSLWXQNIXACPVRNVXRDGGFGPWSPWQPCEHLDGDNSGSCLCRARSC
NOV2h GPWSPWQPCEHLDGDNSGSCLCRARSC
NOV2a DSPRPRCGGLDCLGPAIHIANCSRNGAWXPWSSWALCSXSCGIGFQVRQRSCSNPAPRHG
NOV2b DSPRPRCGGLDCLGPAIHIANCSRNGAWIPWSSWALCSXSCGIGFQVRQRSCSNPAPRHG
NOV2c DSPRPRCGGLDCLGPAIHIANCSRNGAWXPWSSWALCSXSCGIGFQVRQRSCSNPAPRHG
NOV2d DSPRPRCGGLDCLGPAIHIANCSRNGAWIPWSSWALCSXSCGIGFQVRQRSCSNPAPRHG
NOV2e DSPRPRCGGLDCLGPAIHIANCSRNGAWXPWSSWALCSXSCGIGFQVRQRSCSNPAPRHG
NOV2f DSPRPRCGGLDCLGPXIHIANCSRNGAWXPWSSWALCSXSCGIGFQVRQRSCSNPAPRHG
NOV2g DSPRPRCGGLDCLGPAIHIANCSRNGAWXPWSSWAIiCSXSCGIGFQVRQRSCSNPAPRHG
NOV2h DSPRPRCGGLDCLGPXIHIANCSRNGAWXPWSSWALCSXSCGIGFQVRQRSCSNPAPRHG
NOV2a GRICVGKSREERFCNENXPCPVPIFWASWGSWSKCSSNCGGGMQSRRRACENGNSCLGCG
NOV2b GRICVGKSREERFCNENXPCPVPIFWASWGSWSKCSSNCGGGMQSRRRACENGNSCLGCG
NOV2c GRICVGKSREERFCNENXPCPVPIFWASWGSWSKCSSNCGGGMRSRRRACENGNSCLGCG
NOV2d GRICVGKSREERFCNENXPCPVPIFWASWGSWSKCSSNCGGG QSRRRACENGNSCLGCG
NOV2e GRICVGKSREERFCNENXPCPVPIFWASWGSWSKCSSNCGGGMQSRRRACENGNSCLGCG
NOV2f GRICVGKSREERFCNENTPCPVPIFWASWGSWSKCGSNCGGGMQSRRRACENGNSCLGCG
NOV2g GRICVGKSREERFCNENXPCPVPIFWASWGSWSKCSSNCGGGMQSRRRACENGNSCLGCG
NOV2h GRICVGKSREERFCNENXPCPVPIFWASWGSWSKCGSNCGGGMQSRRRACENGNSCLGCG
NOV2a VEFKXCNPEGCPEVRRNXPWXPWLPVNVXQGGARQEQRFRFXCRAPLADPHGLQFGRRRX NOV2b VEFKXCNPEGCPEVRRNXPWXPWLPVNVXQGGARQEQRFRFXCRAPLADPHGLQFGRRRX
NOV2c VEF XCNPEGCPEVRRNXPWXPWLPVNVXQGGARQEQRFRFXCRAPLADPHGLQFGRRRX
NOV2d VEFKXCNPEGCPEVRRNXPWXPWLPVNVXQGGARQEQRFRFXCRAPLADPHGLQFGRRRX
NOV2e VEFKXCNPEGCPEVRRNXPWXPWLPVNVXQGGARQEQRFRFXCRAPLADPHGLQFGRRRX
N0V2f VEFKXCNPEGCPEVRRNXPWXPWLPVNVXQGGARQEQRFRFXCRAPLADPHGLQFGRRRX
NOV2g VEFKXCNPEGCPEVRRNXPWXPWLPVNVXQGGARQEQRFRFXCRAPLADPHGLQFGRRRX
NOV2h VEFKXCNPEGCPEVRRNXPWXPWLPVNVXQGGARQEQRFRFXCRAPLADPHGLQFGRRRX
NOV2a EXRXCPADGSGSCDXDALVEDLLRSGSXSPHXVSGGWAAWGPWSSCSRDCELGFRVRKRX
NOV2b EXRXCPADGSGSCDXDALVEDLLRSGSXSPHXVSGGWAAWGPWSSCSRDCELGFRVRKRX
NOV2c EXRXCPADGSGSCDXDALVEDLLRSGSXSPHXVSGGWAAWGPWSSCSRDCELGFRVRKRX
NOV2d EXRXCPADGSGSCDXDALVEDLLRSGSXSPHXVSGGWAAWGPWSSCSRDCELGFRVRKRX
NOV2e EXRXCPADGSGSCDXDALVEDLLRSGSXSPHXVSGGWAAWGPWSSCSRDCELGFRVRKRX
NOV2f EXRXCPADGSGSCDXDALVEVLLRSGSXSPHXVSGGWAAWGPWSSCSRDCELGFRVRKRX
NOV2g EXRXCPADGSGSCDXDALVEDLLRSGSXSPHXVSGGWAAWGPWSSCSRDCELGFRVRKRX
NOV2h EXRXCPADGSGSCDXDALVEVLLRSGSXSPHXVSGGWAAWGPWSSCSRDCELGFRVRKRX
NOV2a CXNPEPRNGGLPCVGDAAEYQDCNPQACPVRGAWSCWXSWSPCSASCGGGHYQRXRSCXS
NOV2b CXNPEPRNGGLPCVGDAAEYQDCNPQACPVRGAWSCWXSWSPCSASCGGGHYQRXRSCXS
NOV2C CXNPEPRNGGLPCVGDAAEYQDCNPQACPVRGAWSCWXSWSPCSASCGGGHYQRXRSCXS
NOV2d CXNPESRNGGLPCVGDAAEYQDCNPQACPVRGAWSCWXSWSPCSASCGGGHYQRXRSCXS
NOV2e CXNPEPRNGGLPCVGDAAEYQDCNPQACPVRGAWSCWXSWSPCSASCGGGHYQRXRSCXS
NOV2f CXNPEPRNGGLPCVGDAAEYQDCNPQACPVRGAWSCWXSWSPCSASCGGGHYQRXRSCXS
NOV2g CXNPEPRNGGLPCVGDAAEYQDCNPQACPVRGAWSCWXSWSPCSASCGGGHYQRXRSCXS
NOV2h CXNPEPRNGGLPCVGDAAEYQDCNPQACPVRGAWSCWXSWSPCSASCGGGHYQRXRSCXS
NOV2a PAPSPGEDICLGLHXEEALCAXQACPEGWSPWSEWSKCXDDGAQSRSRHCEELLPGSSAC
NOV2b PAPSP EGWSPWSEWSKCXDDGAQSRSRHCEELLPGSSAC
NOV2c PAPSPGEDICLGLHXEEALCAXQACPEGWSPWSEWSKCXDDGAQSRSRHCEELLPGSSAC
NOV2d PAPSPGEDICLGLHXEEALCAXQACPEGWSPWSEWSKCXDDGAQSRSRHCEELLPGSSAC
NOV2e PAPSPGEDICLGLHXEEALCAXQACPEGWSPWSEWSKCXDDGAQSRSRHCEELLPGSSAC
NOV2f PAPSPGEDICLGLHXEEALCAXQACPEGWSPWSEWSKCXDDGAQSRSRHCEELLPGSSAC
NOV2g PAPSPGEDICLGLHXEEALCAXQACPEGWSPWSEWSKCXDDGAQSRSRHCEELLPGSSAC
NOV2h PAPSPGEDICLGLHXEEALCAXQACPEGWSPWSEWSKCXDDGAQSRSRHCEELLPGSSAC
NOV2a AGNSSQSRPCPYSEIPVILPASS EEAXGCAGFNL1HLVAXGISCFLGSGLLXLAVYLSC
NOV2b AGNSSQSRPCPYSEIPVILPASSMEEAXGCAGFNL1HLVAXGISCFLGSGLLXLAVYLSC
NOV2c AGNSSQSRPCVD
NOV2d AGNSSQSRPCVD
NOV2e AGNSSQSRPCVD
NOV2f AGNSSQSRPCVD
NOV2g AGNSSQSRPCPYSEIPVILPASSMEEAXGCAGFNLIHLVAXGISCFLGSGLLXLAVYLSC
NOV2h AGNSSQSRPC
NOV2a QHCQRQSQESTLVHPAXPNHLHYKGGGXPKNEKYXPMEFKXLNKNNLIPDDRANFYPLQQ
NOV2b QHCQRQSQESXLVHPAXPNHLHYKGGGXPKNEKYXPMEFKXLNKNNLIPDDRANFYPLQQ
NOV2c
NOV2d
NOV2e
NOV2f
NOV2g QHCQRQSQESTLVHPAXPNHLHYKGGGXPKNE YXPMEFKXLNKNNLIPDDRANFYPLQQ
NOV2h
NOV2a XNVYXXXYYPSPLNKHSFRPEASPGQRCFPNS
NOV2b XNVYXXXYYPSPLNKHSFRPEASPGQRCFPNS
NOV2c
NOV2d
NOV2e
NOV2f NOV2g XNVYXXXYYPSPLNKHSFRPEASPGQRCFPNS
NOV2h NOV2a (SEQ ID NO 6)
NOV2b (SEQ ID NO 8)
NOV2c (SEQ ID NO 10)
NOV2d (SEQ ID NO 12)
NOV2e (SEQ ID NO 14)
NOV2f (SEQ ID NO 16)
NOV2g (SEQ ID NO 18)
NOV2h (SEQ ID NO 20)
Further analysis of the NOV2a protein yielded the following properties shown in Table 2C.
Table 2C. Protein Sequence Properties NOV2a
SignalP analysis: No Known Signal Sequence Predicted
PSORT II analysis:
PSG: a new signal peptide prediction method
N-region: length 11; pos.chg 1; neg.chg 1 H-region: length 5; peak value -8.91 PSG score: -13.31
GvH: von Heijne's method for signal seq. recognition GvH score (threshold: -2.1): -7.G5 possible cleavage site: between 53 and 5
»> Seems to have no N-terminal signal peptide
ALOM: Klein et al ' s method for TM region allocation Iπit position for calculation: 1
Tentative number of TMS(s) for the threshold 0.5: Number of TMS(s) for threshold 0.5: 1 INTEGRAL Likelihood = -4.83 Transmembrane 259 275 PERIPHERAL Likelihood = 1.54 (at 232)
ALOM score: -4.83 (number of TMSs : 1)
MTOP: Prediction of membrane topology (Hartmann et al . ) Center position for calculation: 266 Charge difference: -3.5 C(-2.5) - N( 1.0) N >= C: N-terminal side will be inside
>>> membrane topology: type 2 (cytoplasmic tail 1 to 259)
MITDISC: discrimination of mitochondrial targeting seq R content: 7 Hyd Moment (75): 4.89 Hyd Moment (95): 4.42 G content: 7 D/E content: 2 S/T content: 8 Score: -1.94
Gavel: prediction of cleavage sites for mitochondrial preseq R-2 motif at 104 LRAlPG
NUCDISC: discrimination of nuclear localization signals pat4 : none pat7 : none bipartite: none N content of basic residues: 9.3% NLS Score: -0.47
KDEL: ER retention motif in the C-terminus : none
ER Membrane Retention Signals : none
SKL: peroxisomal targeting signal in the C-terminus: none
PTS2: 2nd peroxisomal targeting signal: none
VAC: possible vacuolar targeting motif: none
RNA-binding motif : none
Actinin-type actin-binding motif: type 1 : none type 2 : none
NMYR: N-myristoylation pattern : none
Prenylation motif : none memYQRL: transport motif from cell surface to Golgi: none
Tyrosines in the tail: too long tail
Dileucine motif in the tail : found LL at 81 LL at 82 LL at 83 LL at 237 checking 63 PROSITE DNA binding motifs: none checking 71 PROSITE ribosomal protein motifs: none checking 33 PROSITE prokaryotic DNA binding motifs: none
NNCN: Reinhardt's method for Cytoplasmic/Nuclear discrimination Prediction: nuclear Reliability: 94.1
COIL: Lupas's algorithm to detect coiled-coil regions total: 0 residues
Final Results (k = 9/23) :
47.8 %: nuclear
26.1 %: mitochondrial
8.7 % : cytoplasmic
4.3 : Golgi
4.3 % : plasma membrane
4.3 %: extracellular, including cell wall
4.3 % : peroxisomal
» prediction for CG106951-01 is nuc (k=23)
A search of the NOV2a protein against the Geneseq database, a proprietary database that contains sequences published in patents and patent publication, yielded several homologous proteins shown in Table 2D.
Figure imgf000131_0001
In a BLAST search of public sequence databases, the NOV2a protein was found to have homology to the proteins shown in the BLASTP data in Table 2E.
Figure imgf000132_0001
PFam analysis predicts that the NOV2a protein contains the domains shown in the Table 2F.
Figure imgf000133_0001
Example 3.
The NOV3 clone was analyzed, and the nucleotide and encoded polypeptide sequences are shown in Table 3 A.
Table 3A. NOV3 Sequence Analysis
NOV3a, CG121295-01 SEQ ID NO: 23 750 bp DNA Sequence ORF Start: ATG at 41 ORF Stop: TGA at 701
TTCAGTTTGAACGGGAGGTTTTTGATCCCTTTTTTTCAGAATGGATTATTTGCTCATGATTTTCTCTCTGCTG
TTTGTGGCTTGCCAAGGAGCTCCAGAAACAGCAGTCTTAGGCGCTGAGCTCAGCGCGGTGGGTGAGAACGGCG GGGAGAAACCCACTCCCAGTCCACCCTGGCGGCTCCGCCGGTCCAAGCGCTGCTCCTGCTCGTCCCTGATGGA TAAAGAGTGTGTCTACTTCTGCCACCTGGACATCATTTGGGTCAACACTCCCGATTTCTTTCTCTCTTTGGAT AATAGGCACGTTGTTCCGTATGGACTTGGAAGCCCTAGGTCCAAGAGAGCCTTGGAGAATTTACTTCCCACAA AGGCAACAGACCGTGAGAATAGATGCCAATGTGCTAGCCAAAAAGACAAGAAGTGCTGGAATTTTTGCCAAGC AGGAAAAGAACTCAGGGCTGAAGACATTATGGAGAAAGACTGGAATAATCATAAGAAAGGAAAAGACTGTTCC AAGCTTGGGAAAAAGTGTATTTATCAGCAGTTAGTGAGAGGAAGAAAAATCAGAAGAAGTTCAGAGGAACACC TAAGACAAACCAGGTCGGAGACCATGAGAAACAGCGTCAAATCATCTTTTCATGATCCCAAGCTGAAAGGCAA GCCCTCCAGAGAGCGTTATGTGACCCACAACCGAGCACATTGGTGACAGACCTTCGGGGCCTGTCTGAAGCCA TAGCCTCCACGGAGAGCCCT
Figure imgf000134_0001
Further analysis of the NOV3a protein yielded the following properties shown in Table 3B.
Figure imgf000134_0002
VAC: possible vacuolar targeting motif: none
RNA-binding motif: none
Actinin-type actin-binding motif: type 1 : none type 2 : none
NMYR: N-myristoylation pattern : none
Prenylation motif : none memYQRL: transport motif from cell surface to Golgi: none
Tyrosines in the tail : none
Dileucine motif in the tail: none checking 63 PROSITE DNA binding motifs: none checking 71 PROSITE riboso al protein motifs: none checking 33 PROSITE prokaryotic DNA binding motifs: none
NNCN: Reinhardt's method for Cytoplasmic/Nuclear discrimination Prediction: nuclear Reliability: 94.1
COIL: Lupas's algorithm to detect coiled-coil regions total : 0 residues
Final Results (k = 9/23) :
69.6 %: nuclear
13.0 % : mitochondrial
8.7 %: extracellular, including cell wall
8.7 % : cytoplasmic
» prediction for CG121295-01 is nuc (k=23)
A search of the NOV3a protein against the Geneseq database, a proprietary database that contains sequences published in patents and patent publication, yielded several homologous proteins shown in Table 3C.
Figure imgf000136_0001
In a BLAST search of public sequence databases, the NOV3a protein was found to have homology to the proteins shown in the BLASTP data in Table 3D.
Figure imgf000136_0002
PFam analysis predicts that the NOV3a protein contains the domains shown in the Table 3E.
Figure imgf000137_0001
Example 4.
The NOV4 clone was analyzed, and the nucleotide and encoded polypeptide sequences are shown in Table 4A.
Table 4A. NOV4 Sequence Analysis OV4a, CG124756-01 SEQ ID NO: 25 £1076 bp DNA Sequence ORF Start: ATG at 75 [ORF Stop: TGA at 834
GGCTCCTGGTCCCACTGCTGCTCAGCCCAGTGGCCTCACAGGACACCAGCTTCCCAGGAGGCGTCTGACACAG
TATGATGATGAAGATCCCATGGGGCAGCATCCCAGTACTGATGTTGCTCCTGCTCCTGGGCCTAATCGATATC TCCCAGGCCCAGCTCAGCTGCACCGGGCCCCCAGCCATCCCTGGCATCCCGGGTATCCCTGGGACACCTGGCC CCGATGGCCAACCTGGGACCCCAGGGATAAAAGGAGAGAAAGGGCTTCCAGGGCTGGCTGGAGACCATGGTGA GTTCGGAGAGAAGGGAGACCCAGGGATTCCTGGGAATCCAGGAAAAGTCGGCCCCAAGGGCCCCATGGGCCCT iAAAGGTGGCCCAGGGGCCCCTGGAGCCCCAGGCCCCAAAGGTGAATCGGGAGACTACAAGGCCACCCAGAAAA TCGCCTTCTCTGCCACAAGAACCATCAACGTCCCCCTGCGCCGGGACCAGACCATCCGCTTCGACCACGTGAT CACCAACATGAACAACAATTATGAGCCCCGCAGTGGCAAGTTCACCTGCAAGGTGCCCGGTCTCTACTACTTC ACCTACCACGCCAGCTCTCGAGGGAACCTGTGCGTGAACCTCATGCGTGGCCGGGAGCGTGCACAGAAGGTGG TCACCTTCTGTGACTATGCCTACAACACCTTCCAGGTCACCACCGGTGGCATGGTCCTCAAGCTGGAGCAGGG GGAGAACGTCTTCCTGCAGGCCACCGACAAGAACTCACTACTGGGCATGGAGGGTGCCAACAGCATCTTTTCC GGGTTCCTGCTCTTTCCAGATATGGAGGCCTGACCTGTGGGCTGCTTCACATCCACCCCGGCTCCCCCTGCCA GCAACGCTCACTCTACCCCCAACACCACCCCTTGCCCAGCCAATGCACACAGTAGGGCTTGGTGAATGCTGCT
GAGTGAATGAGTAAATAAACTCTTCAAGGCCAAGGGAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAA
AAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAA
|NOV4a, CG124756-01 |SEQ ID NO: 26 253 aa MW at 26721.5kD Protem Sequence
JMMMKIPWGSIPVLMLLLLLGLIDISQAQLSCTGPPAIPGIPGIPGTPGPDGQPGTPGIKGEKGLPGLAGDHGE FGEKGDPGIPGNPGKVGPKGPMGPKGGPGAPGAPGPKGESGDYKATQKIAFSATRTINVPLRRDQTIRFDHVI TNMNNNYEPRSGKFTCKVPGLYYFTYHASSRGNLCVNLMRGRERAQKVVTFCDYAYNTFQVTTGGMVLKLEQG ENVFLQATDKNSLLGMEGANS I FSGFLLFPD EA
Figure imgf000138_0001
NOV4b, CG124756-02 SEQ ID NO: 28 253 aa MW at 26721.5 D Protein Sequence
MM KIPWGSIPVLMLLLLLGLIDISQAQLSCTGPPAIPGIPGIPGTPGPDGQPGTPGIKGEKGLPGLAGDHGE FGEKGDPGIPGNPGKVGPKGPMGP GGPGAPGAPGPKGESGDYKATQKIAFSATRTINVPLRRDQTIRFDHVI TNMNNNYEPRSGKFTCKVPGLYYFTYHASSRGNLCVNLiiRGRERAQKVVTFCDYAYNTFQVTTGG VLKLEQG ENVFLQATDKNSLLGMEGANSIFSGFLLFPDMEA
Figure imgf000138_0002
NOV4c, SNP13382475 of SEQ ID NO: 30 253 aa MW at 26707.5kD CG124756-01, Protein Sequence SNP Pos: 76 jSNP Change: Glu to Asp
MMMKIP GSIPVLMLLLLLGLIDISQAQLSCTGPPAIPGIPGIPGTPGPDGQPGTPGIKGEKGLPGLAGDHGE FGDKGDPGIPGNPGKVGPKGPMGPKGGPGAPGAPGP GESGDYKATQKIAFSATRTINVPLRRDQTIRFDHVI TNMNNNYEPRSGKFTCKVPGLYYFTYHASSRGNLCVNLMRGRERAQKVVTFCDYAYNTFQVTTGGMVLKLEQG ENVFLQATDKNSLLG EGANSIFSGFLLFPDME
Figure imgf000139_0001
NOV4d, SNP13382476 of SEQ ID NO: 32 253 aa MW at 26749.6kD CGI 24756-01, Protein Sequence SNP Pos: 120 SNP Change: Gin to Arg
MMMKIPWGSIPVLMLLLLLGLIDISQAQLSCTGPPAIPGIPGIPGTPGPDGQPGTPGIKGEKGLPGLAGDHGE FGEKGDPGIPGNPG VGPKGPMGPKGGPGAPGAPGPKGESGDYKATRKIAFSATRTINVPLRRDQTIRFDHVI TNMNNNYEPRSGKFTCKVPGLYYFTYHASSRGNLCVNLMRGRERAQKVVTFCDYAYNTFQVTTGGMVLKLEQG ENVFLQATDKNSLLGMEGANSIFSGFLLFPDMEA
A ClustalW comparison of the above protein sequences yields the following sequence alignment shown in Table 4B. Table 4B. Comparison of the NOV4 protein sequences.
NOV4a MKIPWGSIPVLMLLLLLGLIDISQAQLSCTGPPAIPGIPGIPGTPGPDGQPGTPGIKG
NOV4b MMMKIPWGSIPVLMLLLLLGLIDISQAQLSCTGPPAIPGIPGIPGTPGPDGQPGTPGIKG
NOV4a EKGLPGLAGDHGEFGEKGDPGIPGNPGKVGPKGPMGPKGGPGAPGAPGPKGESGDYKATQ
NOV4b EKGLPGLAGDHGEFGEKGDPGIPGNPGKVGPKGPMGPKGGPGAPGAPGPKGESGDYKATQ
NOV4a KlAFSAT INVP RRDQ IRFDHVIT ^^N YEPRSGKFTCKVPG FTYHASS GNL
NOV4b KIAFSATRTINVPLRRDQTIRFDHVITNMNNNYEPRSGKFTCKVPGLYYFTYHASSRGNL
NOV4a CVNLMRGRERAQKVVTFCDYAYNTFQVTTGGIVrVLKLEQGENVFLQATDKNSLLGMEGANS
NOV4b CWLM G F!RAQKVVTFCDYAYNTFQVTTGG^W KLEQGENVF QATDKNSL GMEGANS
NOV4a IFSGFLLFPDMEA
NOV4b IFSGFLLFPDMEA
NOV4a (SEQ ID NO: 26) NOV4b (SEQ ID NO: 28)
Further analysis of the NOV4a protein yielded the following properties shown in Table 4C.
Table 4C. Protein Sequence Properties NOV4a
SignalP analysis: I Cleavage site between residues 28 and 29
PSORT II analysis:
PSG: a new signal peptide prediction method
N-region: length 4; pos.chg 1; neg.chg 0 H-region: length 18; peak value 11.91 PSG score: 7.51
GvH: von Heijne's method for signal seq. recognition GvH score (threshold: -2.1): 4.21 possible cleavage site: between 27 and 28
>>> Seems to have a cleavable signal peptide (1 to 27)
ALOM: Klein et al ' s method for TM region allocation Init position for calculation: 28
Tentative number of TMS(s) for the threshold 0.5: number of TMS(s) .. fixed PERIPHERAL Likelihood = 2.60 (at 232) ALOM score: 2.60 (number of TMSs: 0)
MTOP: Prediction of membrane topology (Hartmann et al.) Center position for calculation: 13 Charge difference: -3.0 C(-1.0) - N( 2.0) N >= C: N-terminal side will be inside
MITDISC: discrimination of mitochondrial targeting seq R content: 0 Hyd Moment (75) : 6.93 Hyd Moment (95): 5.45 G content: 2 D/E content: 1 S/T content: 1 Score: -5.61 Gavel : prediction of cleavage sites or mitochondrial preseq cleavage site motif not found
NUCDISC: discrimination of nuclear localization signals pat4 : none pat7: none bipartite: none content of basic residues: 9.5% NLS Score: -0.47
KDEL: ER retention motif in the C-terminus : none
ER Membrane Retention Signals : none
SKL: peroxisomal targeting signal in the C-terminus : none
PTS2 : 2nd peroxisomal targeting signal : none
VAC: possible vacuolar targeting motif: none
RNA-binding motif: none
Actinin-type actin-binding motif: type 1: none type 2 : none
NMYR: N-myristoylation pattern : none
Prenylation motif : none memYQRL: transport motif from cell surface to Golgi: none
Tyrosines in the tail : none
Dileucine motif in the tail : none checking 63 PROSITE DNA binding motifs: none checking 71 PROSITE ribosomal protein motifs: none checking 33 PROSITE prokaryotic DNA binding motifs: none
NNCN: Reinhardt's method for Cytoplasmic/Nuclear discrimination Prediction: cytoplasmic Reliability: 76.7
COIL: Lupas's algorithm to detect coiled-coil regions total : 0 residues
Final Results (k = 9/23) :
22.2 %: extracellular, including cell wall
22.2 %: vacuolar
22.2 %: mitochondrial
22.2 %: endoplasmic reticulum
11.1 %: Golgi
» prediction for CG124756-01 is exc (k=9) A search of the NOV4a protein against the Geneseq database, a proprietary database that contains sequences published in patents and patent publication, yielded several homologous proteins shown in Table 4D.
Figure imgf000142_0001
In a BLAST search of public sequence databases, the NOV4a protein was found to have homology to the proteins shown in the BLASTP data in Table 4E.
Figure imgf000143_0001
PFam analysis predicts that the NOV4a protein contains the domains shown in the Table 4F.
Figure imgf000143_0002
Example 5.
The NOV5 clone was analyzed, and the nucleotide and encoded polypeptide sequences are shown in Table 5A. Table 5A. NOV5 Sequence Analysis
NOV5a, CG50353-01 3EQ ID NO: 33 1628 bp
DNA Sequence JORF Start: ATG at 1 |ORF Stop: TGA at 1048
ATGAACCGGAAAGCGCGGCGCTGCCTGGGCCACCTCTTTCTCAGCCTGGGCATGGTCTGTCTCCTAGCATGTG GCTTCTCCTCAGTGGTAGCTCTGGGCGCAACGGTCATCTGTAACAAGATCCCAGGCCTGGCTCCCAGACAGCG GGCGATCTGCCAGAGCCGGCCCGACGCCATCATCGTCATAGGAGAAGGCTCACAAATGGGCCTGGACGAGTGT CAGTTTCAGTTCCGCAATGGCCGCTGGAACTGCTCTGCACTGGGAGAGCGCACCGTCTTCGGGAAGGAGCTCA AAGTGGGGAGCCGGGACGGTGCGTTCACCTACGCCATCATTGCCGCCGGCGTGGCCCACGCCATCACAGCTGC CTGTACCCATGGCAACCTGAGCGACTGTGGCTGCGACAAAGAGAAGCAAGGCCAGTACCACCGGGACGAGGGC TGGAAGTGGGGTGGCTGCTCTGCCGACATCCGCTACGGCATCGGCTTCGCCAAGGTCTTCGTGGACGCTCGGG AGATCATGAAGAACGCGCGGCGCCTCATGAACCTGCATAACAATGAGGCCGGCAGGAAGGTTCTAGAGGACCG GATGCAGCTGGAGTGCAAGTGCCACGGCGTGTCTGGCTCCTGCACCACCAAAACCTGCTGGACCACGCTGCCC AAGTTCCGAGAGGTGGGCCACCTGCTGAAGGAGAAGTACAACGCGGCCGTGCAGGTGGAGGTGGTGCGGGCCA GCCGTCTGCGGCAGCCCACCTTCCTGCGCATCAAACAGCTGCGCAGCTATCGCAAGCCCATGAAGACGGACCT GGTGTACATCGAGAAGTCGCCCAACTACTGCGAGGAGGACCCGGTGACCGGCAGTGTGGGCACGCAGGGCCGC GCCTGCAACAAGACGGCTCCCCAGGCCAGCGGCTGTGACCTCATGTGCTGTGGGCGTGGCTACAACACCCACC AGTACGCCCGCGTGTGGCAGTGCAACTGTAAGTTCCACTGGTGCTGCTATGTCAAGTGCAACACGTGCAGCGA GCGCACGGAGATGTACACGTGCAAGTGAGCCCCGTGTGCACACCACCCTCCCGCTGCAAGTCAGATTGCTGGG AGGACTGGACCGTTTCCAAGCTGCGGGCTCCCTGGCAGGATGCTGAGCTTGTCTTTTCTGCTGAGGAGGGTAC
TTTTCCTGGGTTTCCTGCAGGCATCCGTGGGGGAAAAAAAATCTCTCAGAGCCCTCAACTATTCTGTTCCACA
CCCAATGCTGCTCCACCCTCCCCCAGACACAGCCCAGGTCCCTCCGCGGCTGGAGCGAAGCCTTCTGCAGCAG
GAACTCTGGACCCCTGGGCCTCATCACAGCAATATTTAACAATTTATTCTGATAAAAATAATATTAATTTATT
TAATTAAAAAGAATTCTTCCACCTCGTCGGGATCCGTTTTCTGCAATCAAAGTGGACTGCTTGCTTTCCTAGC
JAGGATGATTTTGTTGCTAGGACAAGGAGCCGTGTAGAAGTGTACATAACTATTCTTTATGCAGATATTTCTAC
TAGCTGATTTTGCAGGTACCCACCTTGCAGCACTAGATGTTTAAGTACAAGAGGAGACATCTTTTATGCATAT
ATAGATATACACACACGAAAAA
NOV5a, CG50353-01 SEQ ID NO: 34 349 aa MW at 38980.7kD Protein Sequence j j
MNRKARRCLGHLFLSLGMVCLLACGFSSWALGATVICNKIPGLAPRQRAICQSRPDAIIVIGEGSQ GLDEC QFQFRNGRWNCSALGERTVFGKELKVGSRDGAFTYAIIAAGVAHAITAACTHGNLSDCGCDKEKQGQYHRDEG WKWGGCSADIRYGIGFAKVFVDAREIMKNARRLMNLHNNEAGRKVLEDRMQLECKCHGVSGSCTTKTC TTLP KFREVGHLLKEKYNAAVQVEWRASRLRQPTFLRIKQLRSYRKPMKTDLVYIEKSPNYCEEDPVTGSVGTQGR ACNKTAPQASGCDLMCCGRGYNTHQYARVWQCNCKFHWCCYVKCNTCSERTEMYTCK
Figure imgf000144_0001
Figure imgf000145_0001
Figure imgf000145_0003
NOV5c, 169475673 SEQ ID NO: 38 322 aa MW at 36054.9 D Protein Sequence
RSLGATVICNKIPGLAPRQRAICQSRPDAIIVIGEGSQMGLDECQFQFRNGRWNCSALGERTVFGKELKVGSR EAAFTYAIIAAGVAHAITAACTQGNLSDCGCDKEKQGQYHRDEG KWGGCSADIRYGIGFAKVFVDAREIKQN ARTLMNLHNNEAGRKILEENMKLECKCHGVSGSCTTKTCWTTLPQFRELGYVLKDKYNEAVHVEPVRASRNKR PTFLKIKKPLSYRKPMDTDLVYIEKSPNYCEEDPVTGSVGTQGRACNKTAPQASGCDLMCCGRGYNTHQYARV WQCNCKFHWCCYVKCNTCSERTEMYTCKLE
Figure imgf000145_0002
NOV5d, 228753459 (SEQ ID NO: 40 |322 aa M at 36054.9kD
Protein Sequence
RSLGATVICNKIPGLAPRQRAICQSRPDAIIVIGEGSQMGLDECQFQFRNGRWNCSALGERTVFGKELKVGSR EAAFTYAIIAAGVAHAITAACTQGNLSDCGCDKEKQGQYHRDEGWKWGGCSADIRYGIGFAKVFVDAREIKQN ARTIM^LHNNEAGR I EENMKLECKCHGVSGSCTTKTCWTTLPQFRELGYVLKDKYNEAVHVEPVRASRNKR PTFLKIKKPLSYRKPMDTDLVYIEKSPNYCEEDPVTGSVGTQGRACNKTAPQASGCDLMCCGRGYNTHQYARV WQCNCKFHWCCYVKCNTCSERTEMYTCKLE
NOV5e, 228753462 |SEQ ID NO: 41 966 bp DNA Sequence PJF Start atT lORF Stop: end of sequence
AGATCTCTGGGCGCAACGGTCATCTGTAACAAGATCCCAGGCCTGGCTCCCAGACAGCGGGCGATCTGCCAGA GCCGGCCCGACGCCATCATCGTCATAGGAGAAGGCTCACAAATGGGCCTGGACGAGTGTCAGTTTCAGTTCCG CAATGGCCGCTGGAACTGCTCTGCACTGGGAGAGCGCACCGTCTTCGGGAAGGAGCTCAAAGTGGGGAGCCGG GAGGCTGCATTCACCTACGCCATCATTGCCGCCGGCGTGGTCCACGCCATCACAGCTGCCTGTACCCAGGGCA ACCTGAGCGACTGTGGCTGCGACAAAGAGAAGCAAGGCCAGTACCACCGGGACGAGGGCTGGAAGTGGGGTGG CTGCTCCGCCGACATCCGCTACGGCATCGGCTTCGCCAAGGTCTTTGTGGATGCCCGGGAGATCAAGCAGAAT GCCCGGACTCTCATGAACTTGCACAACAACGAGGCAGGCCGAAAGATCCTGGAGGAGAACATGAAGCTGGAAT GTAAGTGCCACGGCGTGTCAGGCTCGTGCACCACCAAGACGTGCTGGACCACACTGCCACAGTTTCGGGAGCT GGGCTACGTGCTCAAGGACAAGTACAACGAGGCCGTTCACGTGGAGCCTGTGCGTGCCAGCCGCAACAAGCGG CCCACCTTCCTGAAGATCAAGAAGCCACTGTCGTACCGCAAGCCCATGGACACGGACCTGGTGTACATCGAGA AGTCGCCCAACTACTGCGAGGAGGACCCGGTGACCGGCAGTGTGGGCACCCAGGGCCGCGCCTGCAACAAGAC GGCTCCCCAGGCCAGCGGCTGTGACCTCATGTGCTGTGGGCGTGGCTACAACACCCACCAGTACGCCCGCGTG TGGCAGTGCAACTGTAAGTTCCACTGGTGCTGCTATGTCAAGTGCAACACGTGCAGCGAGCGCACGGAGATGT ACACGTGCAAGCTCGAG
NOV5e, 228753462 SEQ ID NO: 42 322 aa MW at 36083.0kD Protein Sequence
RSLGATVICNKIPGLAPRQRAICQSRPDAIIVIGEGSQMGLDECQFQFRNGRWNCSALGERTVFGKELKVGSR EAAFTYAIIAAGWHAITAACTQGNLSDCGCDKEKQGQYHRDEGWKWGGCSADIRYGIGFAKVFVDAREIKQN ARTL NLHNNEAGRKILEENMKLECKCHGVSGSCTTKTCWTTLPQFRELGYVLKDKYNEAVHVEPVRASRNKR PTFLKIKKPLSYRKPMDTDLVYIEKSPNYCEEDPVTGSVGTQGRACNKTAPQASGCDLMCCGRGYNTHQYARV WQCNCKFHWCCYVKCNTCSERTEMYTCKLE
Figure imgf000146_0001
Figure imgf000147_0001
Figure imgf000147_0002
NOV5g, 228753465 SEQ ID NO: 46 322 aa MW at 36173.0kD Protein Sequence ι
RSLGATVICNKIPGLAPRQRAICQSRPDAIIVIGEGSQMGLDECQFQFRNGRWNCSALGERTVFGKELKVGSR EAAFTYAIIAAGVAHAITAACTQGNLSDCDCDKEKQGQYHRDEGWKWGGCSADIRYGIGFAKVFVDAREIKQN ARTLMNLHNNEAGRKILEENMKLECKCHGVSGSCTTKTCWTTLPQFRELGYVLKDKYNEAVHVEPVRASRNKR PTFLKIKKPLSYRKPMDTDLVYIEKSPNYCΞEDPVTGSVGTQGRACNKTAPQASGCDLMCCGRGYNTHQYARV WQYNCKFHWCCYVKCNTCSERTEMYTCKLE
NOV5h, 228753438 SEQ ID NO: 47 966 bp DNA Sequence ORF Start: at 1 ORF Stop: end of sequence
AGATCTCTGGGCGCAACGGTCATCTGTAACAAGATCCCAGGCCTGGCTCCCAGACAGCGGGCGATCTGCCAGA GCCGGCCCGACGCCATCATCGTCATAGGAGAAGGCTCACAAATGGGCCTGGACGAGTGTCAGTTTCAGTTCCG CAATGGCCGCTGGAACTGCTCTGCACTGGGAGAGCGCACCGTCTTCGGGAAGGAGCTCAAAGTGGGGAGCCGG GAGGCTGCGTTCACCTACGCCATCATTGCCGCCGGCGTGGCCCACGCCATCACAGCTGCCTGTACCCAGGGCA ACCTGAGCGACTGTGGCTGCGACAAAGAGAAGCAAGGCCAGTACCACCGGGACGAGGGCTGGAAGTGGGGTGG CTGCTCTGCCGACATCCGCTACGGCATCGGCTTCGCCAAGGTCTTTGTGGATGCCCGGGAGATCAAGCAGAAT GCCCGGACTCTCATGAACTTGCACAACAACGAGGCAGGCCGAAAGATCCTGGAGGAGAACATGAAGCTGGAAT GTAAGTGCCACGGCGTGTCAGGCTCGTGCACCACCAAGACGTGCTGGACCACACTGCCACAGTTTCGGGAGCT GGGCTACGTGCTCAAGGACAAGTACAACGAGGCCGTTCACGTGGAGCCTGTGCGTGCCAGCCGCAACAAGCGG CCCACCTTCCTGAAGATCAAGAAGCCACTGTCGTACCGCAAGCCCATGGACACGGACCTGGTGTACATCGAGA AGTCGACCAACTGCTGCGAGGAGGACCCGGTGACCGGCAGTGTGGGCACCCAGGGCCGCGCCTGCAACAAGAC GGCTCCCCAGGCCAGCGGCTGTGACCTCATGTGCTGTGGGCGTGGCTACAACACCCACCAGTACGCCCGCGTG TGGCAGTGCAACTGTAAGTTCCACTGGTGCTGCTATGTCAAGTGCAACACGTGCAGCGAGCGCACGGAGATGT ACACGTGCAAGCTCGAG
Figure imgf000148_0001
Figure imgf000148_0002
NOV5i, 228753449 SEQ ID NO: 50 322 aa MW at 35926.8kD
Protein Sequence § j |
RSLGATVICNKIPGLAPRQRAICQSRPDAIIVIGEGSQMGLDECQFQFRNGRWNCSALGERTVFGKELKVGSR EAAFTYAIIAAGVAHAITAACTQGNLSDCGCDKEKQGQYHRDEGWKWGGCSADIRYGIGFAKVFVDAREIKQN ARTLMNLHNNEAGRKILEENMKLGCKCHGVSGSCTTKTCWTTLPQFRELGYVLKDKYNEAVHVEPVRASRNKR PTFLKIKKPLSYRKPMDTDLVYIEKSTNCCEEDPVTGSVGTQGRACNKTAPQASGCDLMCCGRGYNTHQYARV WQCNCKFHWCCYVKCNTCSERTEMYTCKLE
MOV5J, CG50353-02 iSEQ ID NO: 51 966 bp
DNA Sequence ORF Start: at 7 ORF Stop: at 961
AGATCTCTGGGCGCAACGGTCATCTGTAACAAGATCCCAGGCCTGGCTCCCAGACAGCGGGCGATCTGCCAGA
GCCGGCCCGACGCCATCATCGTCATAGGAGAAGGCTCACAAATGGGCCTGGACGAGTGTCAGTTTCAGTTCCG CAATGGCCGCTGGAACTGCTCTGCACTGGGAGAGCGCACCGTCTTCGGGAAGGAGCTCAAAGTGGGGAGCCGG GAGGCTGCGTTCACCTACGCCATCATTGCCGCCGGCGTGGCCCACGCCATCACAGCTGCCTGTACCCAGGGCA ACCTGAGCGACTGTGGCTGCGACAAAGAGAAGCAAGGCCAGTACCACCGGGACGAGGGCTGGAAGTGGGGTGG CTGCTCTGCCGACATCCGCTACGGCATCGGCTTCGCCAAGGTCTTTGTGGATGCCCGGGAGATCAAGCAGAAT GCCCGGACTCTCATGAACTTGCACAACAACGAGGCAGGCCGAAAGATCCTGGAGGAGAACATGAAGCTGGAAT GTAAGTGCCACGGCGTGTCAGGCTCGTGCACCACCAAGACGTGCTGGACCACACTGCCACAGTTTCGGGAGCT GGGCTACGTGCTCAAGGACAAGTACAACGAGGCCGTTCACGTGGAGCCTGTGCGTGCCAGCCGCAACAAGCGG CCCACCTTCCTGAAGATCAAGAAGCCACTGTCGTACCGCAAGCCCATGGACACGGACCTGGTGTACATCGAGA AGTCGCCCAACTACTGCGAGGAGGACCCGGTGACCGGCAGTGTGGGCACCCAGGGCCGCGCCTGCAACAAGAC GGCTCCCCAGGCCAGCGGCTGTGACCTCATGTGCTGTGGGCGTGGCTACAACACCCACCAGTACGCCCGCGTG TGGCAGTGCAACTGTAAGTTCCACTGGTGCTGCTATGTCAAGTGCAACACGTGCAGCGAGCGCACGGAGATGT ACACGTGCAAGCTCGAG
Figure imgf000149_0001
NOV5k, CG50353-03 SEQ ID NO: 53 |lQ57 bp DNA Sequence !ORF Start: ATG at 1 ORF Stop: TGA at 1048
ATGAACCGGAAAGCGCGGCGCTGCCTGGGCCACCTCTTTCTCAGCCTGGGCATGGTCTGTCTCCTAGCATGTG GCTTCTCCTCAGTGGTAGCTCTGGGCGCAACGGTCATCTGTAACAAGATCCCAGGCCTGGCTCCCAGACAGCG GGCGATCTGCCAGAGCCGGCCCGACGCCATCATCGTCATAGGAGAAGGCTCACAAATGGGCCTGGACGAGTGT CAGTTTCAGTTCCGCAATGGCCGCTGGAACTGCTCTGCACTGGGAGAGCGCACCGTCTTCGGGAAGGAGCTCA AAGTGGGGAGCCGGGACGGTGCGTTCACCTACGCCATCATTGCCGCCGGCGTGGCCCACGCCATCACAGCTGC CTGTACCCATGGCAACCTGAGCGACTGTGGCTGCGACAAAGAGAAGCAAGGCCAGTACCACCGGGACGAGGGC TGGAAGTGGGGTGGCTGCTCTGCCGACATCCGCTACGGCATCGGCTTCGCCAAGGTCTTCGTGGACGCTCGGG AGATCATGAAGAACGCGCGGCGCCTCATGAACCTGCATAACAATGAGGCCGGCAGGAAGGTTCTAGAGGACCG GATGCAGCTGGAGTGCAAGTGCCACGGCGTGTCTGGCTCCTGCACCACCAAAACCTGCTGGACCACGCTGCCC AAGTTCCGAGAGGTGGGCCACCTGCTGAAGGAGAAGTACAACGCGGCCGTGCAGGTGGAGGTGGTGCGGGCCA GCCGTCTGCGGCAGCCCACCTTCCTGCGCATCAAACAGCTGCGCAGCTATCGCAAGCCCATGAAGACGGACCT GGTGTACATCGAGAAGTCGCCCAACTACTGCGAGGAGGACCCGGTGACCGGCAGTGTGGGCACGCAGGGCCGC GCCTGCAACAAGACGGCTCCCCAGGCCAGCGGCTGTGACCTCATGTGCTGTGGGCGTGGCTACAACACCCACC AGTACGCCCGCGTGTGGCAGTGCAACTGTAAGTTCCACTGGTGCTGCTATGTCAAGTGCAACACGTGCAGCGA GCGCACGGAGATGTACACGTGCAAGTGAGCCCCGT
NOV5k, CG50353-03 SEQ ID NO: 54 349 aa MW at 38980.7kD Protein Sequence
MNRKARRCLGHLFLSLGMVCLLACGFSSWALGATVICNKIPGLAPRQRAICQSRPDAIIVIGEGSQMGLDEC QFQFRNGRWNCSALGERTVFGKELKVGSRDGAFTYAIIAAGVAHAITAACTHGNLSDCGCDKEKQGQYHRDEG WKWGGCSADIRYGIGFAKVFVDAREIMKNARRLMNLHNNEAGRKVLEDRMQLECKCHGVSGSCTTKTCWTTLP KFREVGHLLKEKYNAAVQVEWRASRLRQPTFLRIKQLRSYRKPMKTDLVYIEKSPNYCEEDPVTGSVGTQGR ACNKTAPQASGCDLMCCGRGYNTHQYARVWQCNCKFHWCCYVKCNTCSERTEMYTCK
Figure imgf000150_0001
A Clustal comparison of the above protein sequences yields the following sequence alignment shown in Table 5B. Table5B. Comparison ofthe NOV5protein sequences. ovs MNRKARRCLGHLFLSLGMVCLLACGFSSWALGATVICNKIPGLAPRQRAICQSRPDAII
NOV5b SLGATVICNKIPGLAPRQRAICQSRPDAII
NOV5c RSLGATVICNKIPGLAPRQRAICQSRPDAII
NOV5d RSLGATVICNKIPGLAPRQRAICQSRPDAII
N0V5e RSLGATVICNKIPGLAPRQRAICQSRPDAII
NOV5f SAEFALRSLGATVICNKIPGLAPRQRAICQSRPDAII
NOV5g RSLGATVICNKIPGLAPRQRAICQSRPDAII
NOV5h RSLGATVICNKIPGLAPRQRAICQSRPDAII
NOV5i RSLGATVICNKIPGLAPRQRAICQSRPDAII
NOV5j LGATVICNKIPGLAPRQRAICQSRPDAII
NOV5k MNRKARRCLGHLFLSLGMVCLLACGFSSWALGATVICNKIPGLAPRQRAICQSRPDAII
NOV5a VIGEGSQMGLDECQFQFRNGRWNCSALGERTVFGKELKVGSRDGAFTYAIIAAGVAHAIT
NOV5b VIGEGSQMGLDECQFQFRNGRWNCSALGERTVFGKELKVGSREAAFTYAIIAAGVAHAIT
NOV5c VIGEGSQMGLDECQFQFRNGRWNCSALGERTVFGKELKVGSREAAFTYAIIAAGVAHAIT
NOV5d VIGEGSQMGLDECQFQFRNGRWNCSALGERTVFGKELKVGSREAAFTYAIIAAGVAHAIT
NOV5e VIGEGSQMGLDECQFQFRNGRWNCSALGERTVFGKELKVGSREAAFTYAIIAAGWHAIT
NOV5f VIGEGSQMGLDECQFQFRNGRWNCSALGERTVFGKELKVGSREAAFTYAIIAAGVAHAIT
NOV5g VIGEGSQMGLDECQFQFRNGRWNCSALGERTVFGKELKVGSREAAFTYAIIAAGVAHAIT
NOV5 VIGEGSQMGLDECQFQFRNGRWNCSALGERTVFGKELKVGSREAAFTYAIIAAGVAHAIT
NOV5i VIGEGSQMGLDECQFQFRNGRWNCSALGERTVFGKELKVGSREAAFTYAIIAAGVAHAIT
NOV5J VIGEGSQMGLDECQFQFRNGRWNCSALGERTVFGKELKVGSREAAFTYAIIAAGVAHAIT
NOV5k VIGEGSQMGLDECQFQFRNGRWNCSALGERTVFGKELKVGSRDGAFTYAIIAAGVAHAIT
NOV5a AACTHGNLSDCGCDKEKQGQYHRDEGWKWGGCSADIRYGIGFAKVFVDAREIMKNARRLM
NOV5b AACTQGNLSDCGCDKEKQGQYHRDEGWKWGGCSADIRYGIGFAKVFVDAREIKQNARTLM
N0V5c AACTQGNLSDCGCDKEKQGQYHRDEGWKWGGCSADIRYGIGFAKVFVDAREIKQNARTLM
NOV5d AACTQGNLSDCGCDKEKQGQYHRDEGWKWGGCSADIRYGIGFAKVFVDAREIKQNARTLM
NOV5e AACTQGNLSDCGCDKEKQGQYHRDEGWKWGGCSADIRYGIGFAKVFVDAREIKQNARTLM
NOV5f AACTQGNLSDCGCDKEKQGQYHRDEGWKWGGCSADIRYGIGFAKVFVDAREIKQNARTLM
NOV5g AACTQGNLSDCDCDKEKQGQYHRDEGWKWGGCSADIRYGIGFAKVFVDAREIKQNARTLM
NOV5h AACTQGNLSDCGCDKEKQGQYHRDΞGWKWGGCSADIRYGIGFAKVFVDARΞIKQNARTLM
NOV5i AACTQGNLSDCGCDKEKQGQYHRDEGWKWGGCSADIRYGIGFAKVFVDAREIKQNARTLM
NOV5J AACTQGNLSDCGCDKEKQGQYHRDEGWKWGGCSADIRYGIGFAKVFVDAREIKQNARTLM
NOV5k AACTHGNLSDCGCDKEKQGQYHRDEGWKWGGCSADIRYGIGFAKVFVDAREIMKNARRLM
NOV5a NLHNNEAGRKVLEDRMQLECKCHGVSGSCTTKTCWTTLPKFREVGHLLKEKYNAAVQVEV
NOV5b NLHNNEAGRKILEENMKLECKCHGVSGSCTTKTCWTTLPQFRELGYVLKDKYNEAVHVEP
NOV5c NLHNNEAGRKILEENMKLECKCHGVSGSCTTKTCWTTLPQFRELGYVLKDKYNEAVHVEP
NOV5d NLHNNEAGRKILEENMKLECKCHGVSGSCTTKTCWTTLPQFRELGYVLKDKYNEAVHVEP
NOV5e NLHNNΞAGRKILEENMKLECKCHGVSGSCTTKTCWTTLPQFRELGYVLKDKYNEAVHVEP
NOV5f NLHNNEAGRKILEENMKLECKCHGVSGSCTTKTCWTTLPQFRELGYVLKDKYNEAVHVEP
NOV5g NLHKNEAGRKILEENMKLECKCHGVSGSCTTKTCWTTLPQFRELGYVLKDKYNEAVHVEP
NOV5h NLHNNEAGRKILEENMKLECKCHGVSGSCTTKTCWTTLPQFRELGYVLKDKYNEAVHVEP
NOV5i NLHNNEAGRKILEENMKLGCKCHGVSGSCTTKTCWTTLPQFRELGYVLKDKYNEAVHVEP
NOV5J NLHNNEAGRKILEENMKLECKCHGVSGSCTTKTCWTTLPQFRELGYVLKDKYNEAVHVEP
NOV5k NLHNNEAGRKVLEDRMQLECKCHGVSGSCTTKTCWTTLPKFREVGHLLKEKYNAAVQVEV
NOV5a VRASRLRQPTFLRIKQLRSYRKPMKTDLVYIEKSPNYCEEDPVTGSVGTQGRACNKTAPQ
NOV5b VRASRNKRPTFLKIKKPLSYRKPMDTDLVYIEKSPNYCEEDPVTGSVGTQGRACNKTAPQ
NOV5c VRASRNKRPTFLKIKKPLSYRKPMDTDLVYIEKSPNYCEEDPVTGSVGTQGRACNKTAPQ
NOV5d VRASRNKRPTFLKIKKPLSYRKPMDTDLVYIEKSPNYCEEDPVTGSVGTQGRACNKTAPQ
NOV5e VRASRNKRPTFLKIKKPLSYRKPMDTDLVYIEKSPNYCEEDPVTGSVGTQGRACNKTAPQ
NOV5f VRASRNKRPTFLKIKKPLSYRKPMDTDLVYIEKSPNYCEEDPVTGSVGTQGRACNKTAPQ
NOV5g VRASRNKRPTFLKIKKPLSYRKPMDTDLVYIEKSPNYCEEDPVTGSVGTQGRACNKTAPQ
NOV5h VRASRNKRPTFLKIKKPLSYRKPMDTDLVYIEKSTNCCEEDPVTGSVGTQGRACNKTAPQ NOV5i VRASRNKRPTFLKIKKPLSYRKPMDTDLVYIEKSTNCCEEDPVTGSVGTQGRACNKTAPQ NOV5J VRASRNKRPTFLKIKKPLSYRKPMDTDLVYIEKSPNYCEEDPVTGSVGTQGRACNKTAPQ NOV5k VRASRLRQPTFLRIKQLRSYRKPMKTDLVYIEKSPNYCEEDPVTGSVGTQGRACNKTAPQ
NOV5a ASGCDLMCCGRGYNTHQYARVWQCNCKFHWCCYVKCNTCSERTEMYTCK-- NOV5b ASGCDLMCCGRGYNTHQYARVWQCNCKFHWCCYVKCNTCSERTEMYTCKLE NOV5c ASGCDLMCCGRGYNTHQYARVWQCNCKFHWCCYVKCNTCSERTEMYTCKLE NOV5d ASGCDLMCCGRGYNTHQYARVWQCNCKFHWCCYVKCNTCSERTEMYTCKLE NOV5e ASGCDLMCCGRGYNTHQYARVWQCNCKFHWCCYVKCNTCSERTEMYTCKLE NOV5f ASGCDLMCCGRGYNTHQYARVWQCNCKFHWCYYVKCNTCSERTEMYTCKLE NOV5g ASGCDLMCCGRGYNTHQYARVWQYNCKFHWCCYVKCNTCSERTEMYTCKLE N0V5h ASGCDLMCCGRGYNTHQYARVWQCNCKFHWCCYVKCNTCSERTEMYTCKLE NOV5i ASGCDLMCCGRGYNTHQYARVWQCNCKFHWCCYVKCNTCSERTEMYTCKLE NOV5J ASGCDLMCCGRGYNTHQYARVWQCNCKFHWCCYVKCNTCSERTEMYTCK-- NOV5k ASGCDLMCCGRGYNTHQYARVWQCNCKFHWCCYVKCNTCSERTEMYTCK--
NOV5a (SEQ ID NO 34) NOV5b (SEQ ID NO 36) NOV5c (SEQ ID NO 38) NOV5d (SEQ ID NO 40) NOV5e (SEQ ID NO 42) NOV5f (SEQ ID NO 44) NO 5g (SEQ ID NO 46) NOV5h (SEQ ID NO 48) NOV5i (SEQ ID NO 50) NOV5J (SEQ ID NO 52) NOV5k (SEQ ID NO 54)
Further analysis of the NOV5a protein yielded the following properties shown in Table 5C.
Table 5C. Protein Sequence Properties NOV5a
SignalP analysis: Cleavage site between residues 32 and 33
PSORT H analysis:
PSG: a new signal peptide prediction method
N-region: length 7; pos.chg 4; neg.chg 0 H-region: length 32; peak value 10.30 PSG score: 5.90
GvH: von Heijne's method for signal seq. recognition GvH score (threshold: -2.1): -0.60 possible cleavage site : between 27 and 28
>>> Seems to have a cleavable signal peptide (1 to 27)
ALOM: Klein et al's method for TM region allocation Init position for calculation: 28
Tentative number of TMS(s) for the threshold 0.5 Number of TMS(s) for threshold 0.5: 0 PERIPHERAL Likelihood = 6.89 (at 151) ALOM score: 0.05 (number of TMSs: 0)
MTOP: Prediction of membrane topology (Hartmann et al.) Center position for calculation: 13 Charge difference: -2.5 C( 3.0) - N( 5.5) N >= C: N-terminal side will be inside
MITDISC: discrimination of mitochondrial targeting seq R content: 6 Hyd Moment (75) : 11.39 Hyd Moment (95): 16.83 G content: 5 D/E content: 1 S/T content: 5 Score: 1.59
Gavel: prediction of cleavage sites for mitochondrial preseq R-2 motif at 6j5 SRP|DA
NUCDISC: discrimination of nuclear localization signals pat : none pat7 : none bipartite : none content of basic residues: 14.6% NLS Score: -0.47
KDEL: ER retention motif in the C-terminus: none
ER Membrane Retention Signals :
XXRR-like motif in the N-terminus: NRKA
SKL: peroxisomal targeting signal in the C-terminus : none
PTS2: 2nd peroxisomal targeting signal: none
VAC: possible vacuolar targeting motif: found TLPK at 217
RNA-binding motif: none
Actinin-type actin-binding motif: type 1 : none type 2 : none
NMYR: N-myristoylation pattern : none
Prenylation motif: none memYQRL: transport motif from cell surface to Golgi: none
Tyrosines in the tail: none
Dileucine motif in the tail: none checking 63 PROSITE DNA binding motifs: none checking 71 PROSITE ribosomal protein motifs: none checking 33 PROSITE prokaryotic DNA binding motifs: none
NNCN: Reinhardt's method for Cytoplasmic/Nuclear discrimination Prediction: cytoplasmic Reliability: 55.5
COIL: Lupas's algorithm to detect coiled-coil regions total : 0 residues
Final Results (k = 9/23) :
65.2 %: mitochondrial 13.0 % : Golgi
8.7 %: extracellular, including cell wall
8.7 %: endoplasmic reticulum
4.3 % : cytoplasmic
» prediction for CG50353-01 is mit (k=23)
A search of the NOV5a protein against the Geneseq database, a proprietary database that contains sequences published in patents and patent publication, yielded several homologous proteins shown in Table 5D.
Figure imgf000154_0001
In a BLAST search of public sequence databases, the NOV5a protein was found to have homology to the proteins shown in the BLASTP data in Table 5E.
Figure imgf000155_0001
PFam analysis predicts that the NOV5a protein contains the domains shown in the Table
5F.
Figure imgf000155_0002
Example 6.
The NOV6 clone was analyzed, and the nucleotide and encoded polypeptide sequences are shown in Table 6A.
Figure imgf000156_0001
NOV6a, CG50709-03 SEQ ID NO: 58 331 aa MW at 36432.2kD
Protein Sequence J J
LTGREVLTPFPGLGTAAAPAQGGAHLKQCDLLKLSRRQKQLCRREPGLAETLRDAAHLGLLECQFQFRHER N CSLEGRTGLLKRGFKETAFLYAVSSAALTHTLARACSAGRMERCTCDDSPGLESRQAWQ GVCGDNLKYSTKF LSNFLGSKRGNKDLRARADAHNTHVGIKAVKSGLRTTCKCHGVSGSCAVRTC KQLSPFRETGQVLKLRYDSA VKVSSATNEALGRLELWAPARQGSLTKGLAPRSGDLVYMEDSPSFCRPSKYSPGTAGRVCSREASCSSLCCGR GYDTQSRLVAFSCHCQVQWCCYVECQQCVQEELVYTCKH
Figure imgf000156_0002
NOV6b, 282997951 SEQ ID NO: 60 (309 aa MW at 34226.6kD
Protein Sequence |
TGSQCDLLKLSRRQKQLCRREPGLAETLRDAAHLGLLECQFQFRHERNCSLEGRTGLLKRGFKETAFLYAVS SAALTHTLARACSAGRMERCTCDDSPGLESRQA Q GVCGDNLKYSTKFLSNFLGSKRGNKDLRARADAHNTH VGIKAVKSGLRTTCKCHGVSGSCAVRTC KQLSPFRETGQVLKLRYDSAVKVSSATNEALGRLEL APARQGS LTKGLAPRSGDLVYMEDSPSFCRPSKYSPGTAGRVCSREASCSSLCCGRGYDTQSRLVAFSCHCQVQ CCYVE CQQCVQEELVYTCKLEG
NOV6c, CG50709-05 SEQ ID NO: 61 1464 bp DNA Sequence ORF Start: ATG at 38 ORF Stop: TAG at 1109
GCGAGGAGATGCTAGAGGGCGCAGCGCCGCCAGCACCATGCGCCCCCCGCCCGCGCTGGCCCTGGCCGGGCTC
TGCCTGCTGGCGCTGCCCGCCGCCGCCGCCTCCTACTTCGGCCTGACCGGGCGGGAAGTCCTGACGCCCTTCC CAGGATTGGGCACTGCGGCAGCCCCGGCACAGGGCGGGGCCCACCTGAAGCAGTGTGACCTGCTGAAGCTGTC CCGGCGGCAGAAGCAGCTCTGCCGGAGGGAGCCCGGCCTGGCTGAGACCCTGAGGGATGCTGCGCACCTCGGC CTGCTTGAGTGCCAGTTTCAGTTCCGGCATGAGCGCTGGAACTGTAGCCTGGAGGGCAGGACGGGCCTGCTCA AGAGAGGCTTCAAAGAGACAGCTTTCCTGTACGCGGTGTCCTCTGCCGCCCTCACCCACACCCTGGCCCGGGC CTGCAGCGCTGGGCGCATGGAGCGCTGCACCTGTGATGACTCTCCGGGGCTGGAGAGCCGGCAGGCCTGGCAG TGGGGCGTGTGCGGTGACAACCTCAAGTACAGCACCAAGTTTCTGAGCAACTTCCTGGGGTCCAAGAGAGGAA ACAAGGACCTGCGGGCACGGGCAGACGCCCACAATACCCACGTGGGCATCAAGGCTGTGAAGAGTGGCCTCAG GACCACGTGTAAGTGCCATGGCGTATCAGGCTCCTGTGCCGTGCGCACCTGCTGGAAGCAGCTCTCCCCGTTC CGTGAGACGGGCCAGGTGCTGAAACTGCGCTATGACTCGGCTGTCAAGGTGTCCAGTGCCACCAATGAGGCCT TGGGCCGCCTAGAGCTGTGGGCCCCTGCCAGGCAGGGCAGCCTCACCAAAGGCCTGGCCCCAAGGTCTGGGGA CCTGGTGTACATGGAGGACTCACCCAGCTTCTGCCGGCCCAGCAAGTACTCACCTGGCACAGCAGGTAGGGTG TGCTCCCGGGAGGCCAGCTGCAGCAGCCTGTGCTGCGGGCGGGGCTATGACACCCAGAGCCGCCTGGTGGCCT TCTCCTGCCACTGCCAGGTGCAGTGGTGCTGCTACGTGGAGTGCCAGCAATGTGTGCAGGAGGAGCTTGTGTA CACCTGCAAGCACTAGGCCTACTGCCCAGCAAGCCAGTCTGGCACTGCCAGGACCTCCTGTGGCACCCTTCAA GCTGCCCAGCCGGCCCTCTGGGCAGACTGTCATCACATGCATGCATAAACCGGCATGTGTGCCAATGCACACG
AGTGTGCCACTCACCACCATTCCTTGGCCAGCCTTTTGCCTCCCTCGATACTCAACAAAGAGAAGCAAAGCCT
CCTCCCTTAACCCAAGCATCCCCAACCTTGTTGAGGACTTGGAGAGGAGGGCAGAGTGAGAAAGACATGGAGG
GAAATAAGGGAGACCAAGAGCACAGCAGGACTGAAATTTTGGACGGGAGAGAGGGGCTATTCCATCTTGCTTC
CTGG
NOV6c, CG50709-05 SEQ ID NO: 62 357 aa MW at 38970.2kD Protein Sequence
MRPPPALALAGLCLLALPAAAASYFGLTGREVLTPFPGLGTAAAPAQGGAHLKQCDLLKLSRRQKQLCRREPG LAETLRDAAHLGLLECQFQFRHERWNCSLEGRTGLLKRGFKETAFLYAVSSAALTHTLARACSAGRMERCTCD DSPGLESRQA QWGVCGDNLKYSTKFLSNFLGSKRGNKDLRARADAHNTHVGIKAVKSGLRTTCKCHGVSGSC AVRTC KQLSPFRETGQVLKLRYDSAVKVSSATNEALGRLEL APARQGSLTKGLAPRSGDLVYMEDSPSFCR PSKYSPGTAGRVCSREASCSSLCCGRGYDTQSRLVAFSCHCQVQ CCYVECQQCVQEELVYTCKH NOV6d, 277582109 SEQ ID NO: 63 [1093 bp
DNA Sequence ORF Start: at 2 jORF Stop: end of sequence
CACCGGATCCACCATGCGCCCCCCGCCCGCGCTGGCCCTGGCCGGGCTCTGCCTGCTGGCGCTGCCCGCCGCC GCCGCCTCCTACTTCGGCCTGACCGGGCGGGAAGTCCTGACGCCCTTCCCAGGATTGGGCACTGCGGCAGCCC CGGCACAGGGCGGGGCCCACCTGAAGCAGTGTGACCTGCTGAAGCTGTCCCGGCGGCAGAAGCAGCTCTGCCG GAGGGAGCCCGGCCTGGCTGAGACCCTGAGGGATGCTGCGCACCTCGGCCTGCTTGAGTGCCAGTTTCAGTTC CGGCATGAGCGCTGGAACTGTAGCCTGGAGGGCAGGACGGGCCTGCTCAAGAGAGGCTTCAAAGAGACAGCTT TCCTGTACGCGGTGTCCTCTGCCGCCCTCACCCACACCCTGGCCCGGGCCTGCAGCGCTGGGCGCATGGAGCG CTGCACCTGTGATGACTCTCCGGGGCTGGAGAGCCGGCAGGCCTGGCAGTGGGGCGTGTGCGGTGACAACCTC AAGTACAGCACCAAGTTTCTGAGCAACTTCCTGGGGTCCAAGAGAGGAAACAAGGACCTGCGGGCACGGGCAG ACGCCCACAATACCCACGTGGGCATCAAGGCTGTGAAGAGTGGCCTCAGGACCACGTGTAAGTGCCATGGCGT ATCAGGCTCCTGTGCCGTGCGCACCTGCTGGAAGCAGCTCTCCCCGTTCCGTGAGACGGGCCAGGTGCTGAAA CTGCGCTATGACTCGGCTGTCAAGGTGTCCAGTGCCACCAATGAGGCCTTGGGCCGCCTAGAGCTGTGGGCCC CTGCCAGGCAGGGCAGCCTCACCAAAGGCCTGGCCCCAAGGTCTGGGGACCTGGTGTACATGGAGGACTCACC CAGCTTCTGCCGGCCCAGCAAGTACTCACCTGGCACAGCAGGTAGGGTGTGCTCCCGGGAGGCCAGCTGCAGC AGCCTGTGCTGCGGGCGGGGCTATGACACCCAGAGCCGCCTGGTGGCCTTCTCCTGCCACTGCCAGGTGCAGT GGTGCTGCTACGTGGAGTGCCAGCAATGTGTGCAGGAGGAGCTTGTGTACACCTGCAAGCACCTCGAGGGC
NOV6d, 277582109 SEQ ID NO: 64 364 aa MW at 39615.9kD Protein Sequence
TGSTMRPPPALALAGLCLLALPAAAASYFGLTGREVLTPFPGLGTAAAPAQGGAHLKQCDLLKLSRRQKQLCR REPGLAETLRDAAHLGLLECQFQFRHERWNCSLEGRTGLL RGFKETAFLYAVSSAALTHTLARACSAGRMER CTCDDSPGLESRQA Q GVCGDNLKYSTKFLSNFLGSKRGNKDLRARADAHNTHVGIKAVKSGLRTTCKCHGV SGSCAVRTCWKQLSPFRETGQVLKLRYDSAVKVSSATNEALGRLEL APARQGSLTKGLAPRSGDLVYMEDSP SFCRPSKYSPGTAGRVCSREASCSSLCCGRGYDTQSRLVAFSCHCQVQ CCYVECQQCVQEELVYTCKHLEG
Figure imgf000158_0001
NOV6e, 277582117 SEQ ID NO: 66 341 aa MW at 37431.2kD Protein Sequence j
TGSSYFGLTGREVLTPFPGLGTAAAPAQGGAHLKQCDLLKLSRRQKQLCRREPGLAETLRDAAHLGLLECQFQ FRHERWNCSLEGRTGLLKRGFKETAFLYAVSSAALTHTLARACSAGRMERCTCDDSPGLESRQAWQWGVCGDN LKYSTKFLSNFLGSKRGNKDLRARADAHNTHVGIKAVKSGLRTTCKCHGVSGSCAVRTCW QLSPFRETGQVL KLRYDSAVKVSSATNEALGRLELAPARQGSLTKGLAPRSGDLVYMEDSPSFCRPSKYSPGTAGRVCSREASC SSLCCGRGYDTQSRLVAFSCHCQVQ CCYVECQQCVQEELVYTCKHLEG
Figure imgf000159_0001
NOV6f, CG50709-01 SEQ ID NO: 68 331 aa MW at 36462.3kD
[Protein Sequence j _ [ ] ^ __^ ^ m^m^m^^m^^ LTGREVLTPFPGLGTA^
CSLEGRMGLL RGFKETAFLYAVSSAALTHTLARACSAGRMERCTCDDSPGLESRQA QWGVCGDNLKYSTKF
LSNFLGSKRGNKDLRARADAHNTHVGIKAVKSGLRTTCKCHGVSGSCAVRTCWKQLSPFRETGQVLKLRYDSA
VKVSSATNEALGRLELWAPARQGSLTKGLAPRSGDLVYMEDSPSFCRPSKYSPGTAGRVCSREASCSSLCCGR
[GYDTQSRLVAFSCHCQVQWCCYVECQQCVQEELVYTCKH
ΪNOVόg,CG50709-02 SEQIDNO: 69 933bp
DNA Sequence IlδORF Start: ATG at274 JORF Stop: TAG at928
GGGGCCCACCTGAAGCAGTGTGACCTGCTGAAGCTGTCCCGGCGGCAGAAGCAGCTCTGCCGGAGGGAGCCCG
GCCTGGCTGAGACCCTGAGGGATGCTGCGCACCTCGGCCTGCTTGAGTGCCAGTTTCAGTTCCGGCATGAGCG
CTGGAACTGTAGCCTGGAGGGCAGGACGGGCCTGCTCAAGAGAGGCTTCAAAGAGACAGCTTTCCTGTACGCG
IGTGTCCTCTGCCGCCCTCACCCACACCCTGGCCCGGGCCTGCAGCGCTGGGCGCATGGAGCGCTGCACCTGTG
ATGACTCTCCGGGGCTGGAGAGCCGGCAGGCCTGGCAGTGGGGCGTGTGCGGTGACAACCTCAAGTACAGCAC CAAGTTTCTGAGCAACTTCCTGGGGTCCAAGAGAGGAAACAAGGACCTGCGGGCACGGGCAGACGCCCACAAT ACCCACGTGGGCATCAAGGCTGTGAAGAGTGGCCTCAGGACCACGTGTAAGTGCCATGGCGTATCAGGCTCCT GTGCCGTGCGCACCTGCTGGAAGCAGCTCTCCCCGTTCCGTGAGACGGGCCAGGTGCTGAAACTGCGCTATGA CTCGGCTGTCAAGGTGTCCAGTGCCACCAATGAGGCCTTGGGCCGCCTAGAGCTGTGGGCCCCTGCCAGGCAG GGCAGCCTCACCAAAGGCCTGGCCCCAAGGTCTGGGGACCTGGTGTACATGGAGGACTCACCCAGCTTCTGCC GGCCCAGCAAGTACTCACCTGGCACAGCAGGTAGGGTGTGCTCCCGGGAGGCCAGCTGCAGCAGCCTGTGCTG CGGGCGGGGCTATGACACCCAGAGCCGCCTGGTGGCCTTCTCCTGCCACTGCCAGGTGCAGTGGTGCTGCTAC GTGGAGTGCCAGCAATGTGTGCAGGAGGAGCTGGTGTACACCTGCAAGCACTAGGCC NOV6g, CG50709-02 SEQ ID NO: 70 218 aa MW at 24076.1kD Protein Sequence
MERCTCDDSPGLESRQAWQ GVCGDNLKYSTKFLSNFLGSKRGNKDLRARADAHNTHVGIKAVKSGLRTTCKC IHGVSGSCAVRTC KQLSPFRETGQVLKLRYDSAVKVSSATNEALGRLEL APARQGSLTKGLAPRSGDLVYME DSPSFCRPSKYSPGTAGRVCSREASCSSLCCGRGYDTQSRLVAFSCHCQVQ CCYVECQQCVQEELVYTCKH
Figure imgf000160_0001
NOV6h, CG50709-04 SEQ IDNO: 72 283 aa MWat 31272.4kD Protein Sequence
KQCDLLKLSRRQKQLCRREPGLAETLRDAAHLGLLECQFQFRHER NCSLEGRMGLLKRGFKETAFLYAVSSA ALTHTLARACSAGRMERCTCDDSPGLESRQA QWGVCGDNLKYSTKFLSNFLGSKRGN DLRARADAHNTHVG IKAVKSGLRTTCKCHGVSGSCAVRTCWKQLSPFRETGQVLKLRYDSAVKVSSATNEALGRLELWAPARQGSLT KGLAPRSGDLVYMEDSPSFCRPS YSPGTAGRVCSREASCSSLCCGRGYDTQSRLVAFSCHCQV
Figure imgf000160_0002
NOV6i, CG50709-06 SEQ IDNO: 74 360 aa MW at 39269.6kD Protein Sequence
MRPPPALALAGLCLLALPAAAASYFGLTGREVLTPFPGLGTAAAPAQGGAHLKQCDLLKLSRRQKQLCRREPG LAETLRDAAHLGLLECQFQFRHERWNCSLEGRTGLLKRGFKETAFLYAVSSAALTHTLARACSAGRMERCTCD DSPGLESRQAWQ GVCGDNLKYSTKFLSNFLGSKRGNKDLRARADAHNTHVGIKAVKSGLRTTCKCHGVSGSC AVRTCWKQLSPFRETGQVLKLRYDSAVKVSSATNEALGRLELWAPARQGSLTKGLAPRSGDLVYMEDSPSFCR PSKYSPGTAGRVCSREASCSSLCCGRGYDTQSRLVAFSCHCQVQ CCYVECQQCVQEELVYTCKHLEG
Figure imgf000161_0001
NOV6J, CG50709-07 SEQ IDNO: 76 341 aa MWat37431.2kD Protein Sequence
TGSSYFGLTGREVLTPFPGLGTAAAPAQGGAHLKQCDLLKLSRRQKQLCRREPGLAETLRDAAHLGLLECQFQ FRHERWNCSLEGRTGLLKRGFKETAFLYAVSSAALTHTLARACSAGRMERCTCDDSPGLESRQA Q GVCGDN LKYSTKFLSNFLGSKRGNKDLRARADAHNTHVGIKAV SGLRTTCKCHGVSGSCAVRTC KQLSPFRETGQVL KLRYDSAVKVSSATNEALGRLELWAPARQGSLTKGLAPRSGDLVYMEDSPSFCRPSKYSPGTAGRVCSREASC SSLCCGRGYDTQSRLVAFSCHCQVQ CCYVECQQCVQEELVYTCKHLEG
Figure imgf000162_0001
NOV6k, SNP13381605 of SEQ IDNO: 78 331 aa MW at 36458.3kD CG50709-03,Protein Sequence TGREVLTPFPGLGTAAAPAQGGAHLKQCDLLKLSRRQKQLCRREPGLAETLRDAAHLGLLECQFQFRHERWN CSLEGRTGLLKRGFKETAFLYAVSSAALTHTLARACSAGRMERCTCDDSPGLESRQAWQ GVCGDNLKYSTKF LSNFLGSKRGNKDLRARADAHNTHVGIKAVKSGLRTTCKCHGVSGSCAVRTC KQLSPFRETGQVLKLRYDLA VKVSSATNEALGRLELWAPARQGSLTKGLAPRSGDLVYMEDSPSFCRPSKYSPGTAGRVCSREASCSSLCCGR GYDTQSRLVAFSCHCQVQWCCYVECQQCVQEELVYTCKH
Figure imgf000162_0002
Figure imgf000163_0001
Figure imgf000163_0002
Figure imgf000164_0002
Figure imgf000164_0001
Figure imgf000164_0003
NOV60, SNP13378336 of SEQ ID NO: 86 331 aa MW at 36372.2kD CG50709-03, Protein Sequence SNP Pos: 294 |SNP Change: Tyr to Cys TGREVLTPFPGLGTAAAPAQGGAHLKQCDLLKLSRRQKQLCRREPGLAETLRDAAHLGLLECQFQFRHERWN CSLEGRTGLLKRGFKETAFLYAVSSAALTHTLARACSAGRMERCTCDDSPGLESRQAWQWGVCGDNLKYSTKF LSNFLGSKRGNKDLRARADAHNTHVGIKAVKSGLRTTCKCHGVSGSCAVRTC KQLSPFRETGQVLKLRYDSA [VKVSSATNΞALGRLELWAPARQGSLT GLAPRSGDLVYMEDSPSFCRPSKYSPGTAGRVCSREASCSSLCCGR GCDTQSRLVAFSCHCQVQ CCYVECQQCVQEELVYTCKH
Figure imgf000165_0001
Figure imgf000165_0002
A ClustalW comparison of the above protein sequences yields the following sequence alignment shown in Table 6B.
Table 6B. Comparison of the NOV6 protein sequences.
NOV6a TGREVLTPFPGLGTAAAPAQGGAHLKQCD
NOVδb TGSQCD
NOV6c MRPPPALALAGLCLLALPAAAASYFGLTGREVLTPFPGLGTAAAPAQGGAHLKQCD
NOV6d GSTMRPPPALALAGLCLLALPAAAASYFGLTGREVLTPFPGLGTAAAPAQGGAHLKQCD
NOV6e TGSSYFGLTGREVLTPFPGLGTAAAPAQGGAHLKQCD
NOV6f LTGREVLTPFPGLGTAAAPAQGGAHLKQCD
NOV6g
NOV6h KQCD
NOV6i MRPPPALALAGLCLLALPAAAASYFGLTGREVLTPFPGLGTAAAPAQGGAHLKQCD N0 6J -TGSSYFGLTGREVLTPFPGLGTAAAPAQGGAHLKQCD
NOVSa LLKLSRRQKQLCRREPGLAETLRDAAHLGLLECQFQFRHER NCSLEGRTGLLKRGFKET
NOV6b LLKLSRRQKQLCRREPGLAETLRDAAHLGLLECQFQFRHERNCSLEGRTGLLKRGFKET
N0V6c LLKLSRRQKQLCRREPGLAETLRDAAHLGLLECQFQFRHER NCSLEGRTGLLKRGFKET
NOV6d LLKLSRRQKQLCRREPGLAETLRDAAHLGLLECQFQFRHERWNCSLEGRTGLLKRGFKET
NOV6e LLKLSRRQKQLCRREPGLAETLRDAAHLGLLECQFQFRHERWNCSLEGRTGLLKRGFKET
NOV6f LLKLSRRQKQLCRREPGLAETLRDAAHLGLLECQFQFRHER NCSLEGRMGLLKRGFKET
N0V6g
N0V6h LLKLSRRQKQLCRREPGLAETLRDAAHLGLLECQFQFRHER NCSLEGRMGLLKRGFKET
NOV6i LLKLSRRQKQLCRREPGLAETLRDAAHLGLLECQFQFRHER NCSLEGRTGLLKRGFKET
N0V6j LLKLSRRQKQLCRREPGLAETLRDAAHLGLLECQFQFRHERNCSLEGRTGLLKRGFKET
NOV6a AFLYAVSSAALTHTLARACSAGRMERCTCDDSPGLESRQA QWGVCGDNLKYSTKFLSNF
N0V6b AFLYAVSSAALTHTLARACSAGRMERCTCDDSPGLESRQA Q GVCGDNLKYSTKFLSNF
N0V6c AFLYAVSSAALTHTLARACSAGRMERCTCDDSPGLESRQA Q GVCGDNLKYSTKFLSNF
N0V6d AFLYAVSSAALTHTLARACSAGRMERCTCDDSPGLESRQA Q GVCGDNLKYSTKFLSNF
N0V6e AFLYAVSSAALTHTLARACSAGRMERCTCDDSPGLESRQAWQWGVCGDNLKYSTKFLSNF
NOVSf AFLYAVSSAALTHTLARACSAGRMERCTCDDSPGLESRQA QWGVCGDNLKYSTKFLSNF
N0V6g MERCTCDDSPGLESRQA Q GVCGDNLKYSTKFLSNF
NOVGh AFLYAVSSAALTHTLARACSAGRMERCTCDDSPGLESRQA Q GVCGDNLKYSTKFLSNF
NOVSi AFLYAVSSAALTHTLARACSAGRMERCTCDDSPGLESRQA Q GVCGDNLKYSTKFLSNF
NOV6J AFLYAVSSAALTHTLARACSAGRMERCTCDDSPGLESRQAQ GVCGDNLKYSTKFLSNF
NOV6a LGSKRGNKDLRARADAHNTHVGIKAVKSGLRTTCKCHGVSGSCAVRTCWKQLSPFRETGQ
NOV6b LGSKRGNKDLRARADAHNTHVGIKAVKSGLRTTCKCHGVSGSCAVRTCWKQLSPFRETGQ
NOV6c LGSKRGNKDLRARADAHNTHVGIKAVKSGLRTTCKCHGVSGSCAVRTCWKQLSPFRETGQ
NOV6d LGSKRGNKDLRARADAHNTHVGIKAVKSGLRTTCKCHGVSGSCAVRTC KQLSPFRETGQ
NOV6e LGSKRGNKDLRARADAHNTHVGIKAVKSGLRTTCKCHGVSGSCAVRTCWKQLSPFRETGQ
NOV6f LGSKRGNKDLRARADAHNTHVGIKAVKSGLRTTCKCHGVSGSCAVRTC KQLSPFRETGQ
NOV6g LGSKRGNKDLRARADAHNTHVGIKAVKSGLRTTCKCHGVSGSCAVRTC KQLSPFRETGQ
NOVδh LGSKRGNKDLRARADAHNTHVGIKAVKSGLRTTCKCHGVSGSCAWTCWKQLSPFRETGQ
NOVSi LGSKRGNKDLRARADAHNTHVGIKAVKSGLRTTCKCHGVSGSCAVRTCWKQLSPFRETGQ
NOV6j LGSKRGNKDLRARADAHNTHVGIKAVKSGLRTTCKCHGVSGSCAVRTCWKQLSPFRETGQ
NOV6a VLKLRYDSAVKVSSATNEALGRLELWAPARQGSLTKGLAPRSGDLVYMEDSPSFCRPSKY
NOV6b VLKLRYDSAVKVSSATNEALGRLELWAPARQGSLTKGLAPRSGDLVYMEDSPSFCRPSKY
N0V6C VLKLRYDSAVKVSSATNEALGRLELWAPARQGSLTKGLAPRSGDLVYMEDSPSFCRPSKY
NOV6d VLKLRYDSAVKVSSATNEALGRLELWAPARQGSLTKGLAPRSGDLVYMEDSPSFCRPSKY
NOV6e VLKLRYDSAVKVSSATNEALGRLEL APARQGSLTKGLAPRSGDLVYMEDSPSFCRPSKY
NOV6f VLKLRYDSAVKVSSATNEALGRLELWAPARQGSLTKGLAPRSGDLVYMEDSPSFCRPSKY
NOV6g VLKLRYDSAVKVSSATNEALGRLEL APARQGSLTKGLAPRSGDLVYMEDSPSFCRPSKY
NOV6h VLKLRYDSAVKVSSATNEALGRLEL APARQGSLTKGLAPRSGDLVYMEDSPSFCRPSKY
NOV6i VLKLRYDSAVKVSSATNEALGRLEL APARQGSLTKGLAPRSGDLVYMEDSPSFCRPSKY
NOVβj VLKLRYDSAVKVSSATNEALGRLELWAPARQGSLTKGLAPRSGDLVYMEDSPSFCRPSKY
NOV6a SPGTAGRVCSREASCSSLCCGRGYDTQSRLVAFSCHCQVQ CCYVECQQCVQEELVYTCK
NOV6b SPGTAGRVCSREASCSSLCCGRGYDTQSRLVAFSCHCQVQWCCYVECQQCVQEELVYTCK
NOVSc SPGTAGRVCSREASCSSLCCGRGYDTQSRLVAFSCHCQVQWCCYVECQQCVQEELVYTCK
NOV6d SPGTAGRVCSREASCSSLCCGRGYDTQSRLVAFSCHCQVQ CCYVECQQCVQEELVYTCK
NOV6e SPGTAGRVCSREASCSSLCCGRGYDTQSRLVAFSCHCQVQWCCYVECQQCVQEELVYTCK
NOV6f SPGTAGRVCSREASCSSLCCGRGYDTQSRLVAFSCHCQVQWCCYVECQQCVQEELVYTCK
NOV6g SPGTAGRVCSREASCSSLCCGRGYDTQSRLVAFSCHCQVQWCCYVECQQCVQEELVYTCK
NOV6h SPGTAGRVCSREASCSSLCCGRGYDTQSRLVAFSCHCQV
NOV6i SPGTAGRVCSREASCSSLCCGRGYDTQSRLVAFSCHCQVQ CCYVECQQCVQEELVYTCK
NOV6J SPGTAGRVCSREASCSSLCCGRGYDTQSRLVAFSCHCQVQ CCYVECQQCVQEELVYTCK
NOV6a H NOV6b LEG- NOV6c H
NOV6d HLEG
NOV6e HLEG
NOV6f H
NOV6g H
NOVδh
NOV6i HLEG
NOV6J HLEG
NOV6a (SEQ ID NO 58)
N0V6b (SEQ ID NO 60)
N0V6C (SEQ ID NO 62)
NOVδd (SEQ ID NO 64)
N0V6e (SEQ ID NO 66)
NOVβf (SEQ ID NO 68)
NOV6g (SEQ ID NO 70)
N0V6h (SEQ ID NO 72)
N0V6i (SEQ ID NO 74)
N0V6J (SEQ ID NO 76)
Further analysis of the NOV6a protein yielded the following properties shown in Table 6C.
Table 6C. Protein Sequence Properties NOV6a
SignalP analysis: No Known Signal Sequence Predicted
PSORT II analysis:
PSG: a new signal peptide prediction method
N-region: length 5; pos.chg 1; neg.chg 1 H-region: length 21; peak value 7.18 PSG score: 2.78
GvH: von Heijne's method for signal seq. recognition GvH score (threshold: -2.1): -5.38 possible cleavage site: between 22 and 23
»> Seems to have no N-terminal signal peptide
ALOM: Klein et al ' s method for TM region allocation Init position for calculation: 1
Tentative number of TMS(s) for the threshold 0.5: number of TMS(s) .. fixed PERIPHERAL Likelihood = 4.08 (at 90) ALOM score: 4.08 (number of TMSs: 0)
MTOP: Prediction of membrane topology (Hartmann et al.) Center position for calculation: 6 Charge difference: 3.5 C( 4.5) - N( 1.0) C > N: C-terminal side will be inside
»>Caution: Inconsistent mtop result with signal peptide MITDISC: discrimination of mitochondrial targeting seq R content: 1 Hyd Moment (75): 11.91 Hyd Moment (95) : 8.21 G content: 5 D/E content: 2 S/T content: 3 Score: -6.56 Gavel : prediction of cleavage sites for mitochondrial preseq R-2 motif at 53 CRR|EP
NUCDISC: discrimination of nuclear localization signals pat4 : none pat7 : none bipartite: none content of basic residues: 14.2% NLS Score: -0.47
KDEL: ER retention motif in the C-terminus : none
ER Membrane Retention Signals :
XXRR-like motif in the N-terminus : TGRE KKXX-like motif in the C-terminus : YTCK
SKL: peroxisomal targeting signal in the C-terminus : none
PTS2 : 2nd peroxisomal targeting signal .- found KLSRRQKQL at 33
VAC: possible vacuolar targeting motif: none
RNA-binding motif: none
Actinin-type actin-binding motif: type 1 : none type 2 : none
NMYR: N-myristoylation pattern : none
Prenylation motif: none memYQRL: transport motif from cell surface to Golgi: none
Tyrosines in the tail : none
Dileucine motif in the tail : none checking 63 PROSITE DNA binding motifs: none checking 71 PROSITE ribosomal protein motifs: none checking 33 PROSITE prokaryotic DNA binding motifs: none
NNCN: Reinhardt's method for Cytoplasmic/Nuclear discrimination Prediction: nuclear Reliability: 76.7
COIL: Lupas ' s algorithm to detect coiled-coil regions total : 0 residues
Final Results (k = 9/23) :
78.3 %: nuclear 13.0 %: mitochondrial 8.7 %: cytoplasmic
>> prediction for CG50709-03 is nuc (k=23) A search of the NOV6a protein against the Geneseq database, a proprietary database that contains sequences published in patents and patent publication, yielded several homologous proteins shown in Table 6D.
Figure imgf000169_0001
In a BLAST search of public sequence databases, the NOV6a protein was found to have homology to the proteins shown in the BLASTP data in Table 6E.
Figure imgf000170_0001
PFam analysis predicts that the NOV6a protein contains the domains shown in the Table 6F.
Figure imgf000170_0002
Example 7.
The NOV7 clone was analyzed, and the nucleotide and encoded polypeptide sequences are shown in Table 7A. Table 7A. NOV7 Sequence Analysis
NOV7a, CG53054-02 SEQ ID NO: 89 jl 128 bp
DNA Sequence ORF Start: ATG at 31 ORF Stop: TGA at 1102
TCCCGGCCCTCCGCGCCCTCTCGCGCGGCGATGGCCCCACTCGGATACTTCTTACTCCTCTGCAGCCTGAAGC
AGGCTCTGGGCAGCTACCCGATCTGGTGGCTGACGGGCAGCGAGCCCCTGACCATCCTCCCGCTGACCCTGGA GCCAGAGGCGGCTGCCCAGGCGCACTACAAGGCCTGCGACCGGCTGAAGCTGGAGCGGAAGCAGCGGCGCATG TGCCGCCGGGACCCGGGCGTGGCAGAGACGCTGGTGGAGGCCGTGAGCATGAGTGCGCTCGAGTGCCAGTTCC AGTTCCGCTTTGAGCGCTGGAACTGCACGCTGGAGGGCCGCTACCGGGCCAGCCTGCTCAAGCGAGGTTTCAA GGAGACTGCCTTCCTCTATGCCATCTCCTCGGCTGGCCTGACGCACGCACTGGCCAAGGCGTGCAGCGCGGGC CGCATGGAGCGCTGTACCTGCGATGAGGCACCCGACCTGGAGAACCGTGAGGCCTGGCAGTGGGGGGGCTGCG GAGACAACCTTAAGTACAGCAGCAAGTTCGTCAAGGAATTCCTGGGCAGACGGTCAAGCAAGGATCTGCGAGC CCGTGTGGACTTCCACAACAACCTCGTGGGTGTGAAGGTGATCAAGGCTGGGGTGGAGACCACCTGCAAGTGC CACGGCGTGTCAGGCTCATGCACGGTGCGGACCTGCTGGCGGCAGTTGGCGCCTTTCCATGAGGTGGGCAAGC ATCTGAAGCACAAGTATGAGACGGCACTCAAGGTGGGCAGCACCACCAATGAAGCTGCCGGCGAGGCAGGTGC CATCTCCCCACCACGGGGCCGTGCCTCGGGGGCAGGTGGCAGCGACCCGCTGCCCCGCACTCCAGAGCTGGTG CACCTGGATGACTCGCCTAGCTTCTGCCTGGCTGGCCGCTTCTCCCCGGGCACCGCTGGCCGTAGGTGCCACC GTGAGAAGAACTGCGAGAGCATCTGCTGTGGCCGCGGCCATAACACACAGAGCCGGGTGGTGACAAGGCCCTG CCAGTGCCAGGTGCGTTGGTGCTGCTATGTGGAGTGCAGGCAGTGCACGCAGCGTGAGGAGGTCTACACCTGC AAGGGCTGAGTTCCCAGGCCCTGCCAGCCCTGC
NOV7a, CG53054-02 SEQ ID NO: 90 357 aa MW at 39756.1kD Protein Sequence
MAPLGYFLLLCSLKQALGSYPI WLTGSEPLTILPLTLEPEAAAQAHYKACDRLKLERKQRRMCRRDPGVAET LVEAVSMSAIiECQFQFRFER NCTLEGRYRASLLKRGPKETAFLYAISSAGLTHALAKACSAGRMERCTCDEA PDLENREA Q GGCGDNLKYSSKFVKEFLGRRSSKDLRARVDFHNNLVGVKVIKAGVETTCKCHGVSGSCTVR TCWRQLAPFHEVGKHLKHKYETALKVGSTTNEAAGEAGAISPPRGRASGAGGSDPLPRTPELVHLDDSPSFCL AGRFSPGTAGRRCHREKNCESICCGRGHNTQSRWTRPCQCQVR CCYVECRQCTQREEVYTCKG
Figure imgf000171_0001
NOV7b, 170251039 SEQ ID NO: 92 343 aa MW at 38208.1kD Protein Sequence
GSSYPI WLTGSEPLTILPLTLEPEAGAQAHYKACDRLKLERKQRRMCRRDPGWETLVEAVSMSALECQFQF RFERNCTLEGRYRASLLKRGFKETAFLYAISSAGLTHALAKACSAGRMERCTCDEAPDLENREA Q GGCGD NLKYSSKFVKEFLGRRSSKDLRARVDFHNNLVGVKVIKAGVETTCKCHGVSGSCTVRTC RQLAPFHEVGKHL KHKYETAL VGSTTNEAAGEAGAISPPRGRASGAGGSDPLPRTPELVHLDDSPSFCLAGRFSPGTAGRRCHRE KNCESICCGRGHNTQSRWTRPCQCQVR CCYVECRQCTQREEVYTCKGVD
pSfOV7c, 170251076 SEQ ID NO: 93 1029 bp
DNA Sequence ORF Start: at 1 [ORF Stop: end of sequence
GGATCCAGCTACCCGATCTGGTGGCTGACGGGCAGCGAGCCCCTGACCATCCTCCCGCTGACCCTGGAGCCAG AGGCGGCCGCCCAGGCGCACTACAAGGCCTGCGACCGGCTGAAGCTGGAGCGGAAGCAGCGGCGCATGTGCCG CCGGGACCCGGGCGTGGCAGAGACGCTGGTGGAGGCCGTGAGCATGAGTGCGCTCGAGTGCCAGTTCCAGTTC CGCTTTGAGCGCTGGAACTGCACGCTGGAGGGCCGCTACCGGGCCAGCCTGCTCAAGCGAGGCTTCAAGGAGA CTGCCTTCCTCTATGCCATCTCCTCGGCTGGCCTGACGCACGCACTGGCCAAGGCGTGCAGCGCGGGCCGCAT GGAGCGCTGTACCTGCGATGAGGCACCCGACCTGGAGAACCGTGAGGCCTGGCAGTGGGGGGGCTGCGGAGAC AACCTTAAGTACAGCAGCAAGTTCGTCAAGGAATTCCTGGGCAGACGGTCAAGCAAGGATCTGCGAGCCCGTG TGGACTTCCACAACAACCTCGTGGGTGTGAAGGTGATCAAGGCTGGGGTGGAGACCACCTGCAAGTGCCACGG CGTGTCAGGCTCATGCACGGTGCGGACCTGCTGGCGGCAGTTGGCGCCTTTCCATGAGGTGGGCAAGCATCTG AAGCACAAGTATGAGACGGCACTCAAGGTGGGCAGCACCACCAATGAAGCTGCCGGCGAGGCAGGTGCCATCT CCCCACCACGGGGCCGTGCCTCGGGGGCAGGTGGCAGCGACCCGCTGCCCCGCACTCCAGAGCTGGTGCACCT GGATGACTCGCCTAGCTTCTGCCTGGCTGGCCGCTTCTCCCCGGGCACCGCTGGCCGTAGGTGCCACCGTGAG AAGAACTGCGAGAGCATCTGCTGTGGCCGCGGCCATAACACACAGAGCCGGGTGGTGACAAGGCCCTGCCAGT GCCAGGTGCGTTGGTGCTGCTATGTGGAGTGCAGGCAGTGCACGCAGCGTGAGGAGGTCTACACCTGCAAGGG CGTCGAC
NOV7c, 170251076 SEQ ID NO: 94 343 aa MW at 38194.1kD Protein Sequence
GSSYPIWWLTGSEPLTILPLTLEPEAAAQAHYKACDRLKLERKQRRMCRRDPGVAETLVEAVSMSALECQFQF RFERVJNCTLEGRYRASLLKRGFKETAFLYAISSAGLTHALAKACSAGRMERCTCDEAPDLENREAWQ GGCGD NLKYSSKFVKEFLGRRSSKDLRARVDFHNNLVGVKVIKAGVETTCKCHGVSGSCTVRTC RQLAPFHEVGKHL KHKYETALKVGSTTNEAAGEAGAISPPRGRASGAGGSDPLPRTPELVHLDDSPSFCLAGRFSPGTAGRRCHRE KNCESICCGRGHNTQSRWTRPCQCQVR CCYVECRQCTQREEVYTCKGVD
NOV7d, CG53054-01 SEQ ID NO: 95 11085 bp
DNA Sequence R Start: ATG at 13 fORF Stop: TGA at 1078
TAGTGAGCCGAGATGGCACTACTATATTCCAGCTTGGGTGTGGTTGTGTGCACCTGTAGTCCTAGTTACTTTG
GACTGACGGGCAGCGAGCCCCTGACCATCCTCCCGCTGACCCTGGAGCCAGAGGCGGCTGCCCAGGCGCACTA CAAGGCCTGCGACCGGCTGAAGCTGGAGCGGAAGCAGCGGCGCATGTGCCGCCGGGACCCGGGCGTGGCAGAG ACGCTGGTGGAGGCCGTGAGCATGAGTGCGCTCGAGTGCCAGTTCCAGTTCCGCTTTGAGCGCTGGAACTGCA CGCTGGAGGGCCGCTACCGGGCCAGCCTGCTCAAGCGAGGTTTCAAGGAGACTGCCTTCCTCTATGCCATCTC CTCGGCTGGCCTGACGCACGCACTGGCCAAGGCGTGCAGCGCGGGCCGCATGGAGCGCTGTACCTGCGATGAG GCACCCGACCTGGAGAACCGTGAGGGCTGGAAGTGGGGTGGCTGTAGCGAGGACATCGAGTTTGGTGGGATGG TGTCTCGGGAGTTCGCCGACGCCCGGGAGAACCGGCCAGATGCCCGCTCAGCCATGAACCGCCACAACAACGA GGCTGGGCGCCAGGTGATCAAGGCTGGGGTGGAGACCACCTGCAAGTGCCACGGCGTGTCAGGCTCATGCACG GTGCGGACCTGCTGGCGGCAGTTGGCGCCTTTCCATGAGGTGGGCAAGCATCTGAAGCACAAGTATGAGTCGG CACTCAAGGTGGGCAGCACCACCAATGAAGCTGCCGGCGAGGCAGGTGCCATCTCCCCACCACGGGGCCGTGC CTCGGGGGCAGGTGGCAGCGACCCGCTGCCCCGCACTCCAGAGCTGGTGCACCTGGATGACTCGCCTAGCTTC TGCCTGGCTGGCCGCTTCTCCCCGGGCACCGCTGGCCGTAGGTGCCACCGTGAGAAGAACTGCGAGAGCATCT GCTGTGGCCGCGGCCATAACACACAGAGCCGGGTGGTGACAAGGCCCTGCCAGTGCCAGGTGCGTTGGTGCTG CTATGTGGAGTGCAGGCAGTGCACGCAGCGTGAGGAGGTCTACACCTGCAAGGGCTGAGTTCC
NOV7d, CG53054-01 SEQ ID NO: 96 355 aa MW at 39194.1kD Protein Sequence
MALLYSSLGVWCTCSPSYFGLTGSEPLTILPLTLEPEAAAQAHYKACDRLKLERKQRRMCRRDPGVAETLVE AVSMSALECQFQFRFERWNCTLEGRYRASLLKRGFKETAFLYAISSAGLTHALAKACSAGRMERCTCDEAPDL ENREG K GGCSEDIEFGGMVSREFADARENRPDARSAMNRHNNEAGRQVIKAGVETTCKCHGVSGSCTVRTC RQLAPFHEVGKHLKH YESALKVGSTTNEAAGEAGAISPPRGRASGAGGSDPLPRTPELVHLDDSPSFCLAG RFSPGTAGRRCHREKNCESICCGRGHNTQSRWTRPCQCQVRWCCYVECRQCTQREEVYTCKG
Figure imgf000173_0001
NOV7e, CG53054-03 SEQ ID NO: 98 339 aa MWat 37835.8kD Protein Sequence
SYPI WLTGSEPLTILPLTLEPEAAAQAHYKACDRLKLERKQRRMCRRDPGVAETLVEAVSMSALECQFQFRF ERWNCTLEGRYRASLLKRGFKETAFLYAISSAGLTHALAKACSAGRMERCTCDEAPDLENREA Q GGCGDNL KYSSKFVKEFLGRRSSKDLRARVDFHNNLVGVIVIKAGVETTCKCHGVSGSCTVRTCWRQLAPFHEVGKHLKH KYETALKVGSTTNEAAGEAGAISPPRGRASGAGGSDPLPRTPELVHLDDSPSFCLAGRFSPGTAGRRCHREKN CESICCGRGHNTQSRWTRPCQCQVRWCCYVECRQCTQREEVYTCKG
NOV7f, CG53054-04 SEQ ID NO: 99 1631 bp DNA Sequence ORF Start: ATG at 12 ORF Stop: TGA at 1107
GGCGCGGCAAGATGCTGGATGGGTCCCCGCTGGCGCGCTGGCTGGCCGCGGCCTTCGGGCTGACGCTGCTGCT
CGCCGCGCTGCGCCCTTCGGCCGCCTACTTCGGGCTGACGGGCAGCGAGCCCCTGACCATCCTCCCGCTGACC CTGGAGCCAGAGGCGGCCGCCCAGGCGCACTACAAGGCCTGCGACCGGCTGAAGCTGGAGCGGAAGCAGCGGC GCATGTGCCGCCGGGACCCGGGCGTGGCAGAGACGCTGGTGGAGGCCGTGAGCATGAGTGCGCTCGAGTGCCA GTTCCAGTTCCGCTTTGAGCGCTGGAACTGCACGCTGGAGGGCCGCTACCGGGCCAGCCTGCTCAAGCGAGGC TTCAAGGAGACTGCCTTCCTCTATGCCATCTCCTCGGCTGGCCTGACGCACGCACTGGCCAAGGCGTGCAGCG CGGGCCGCATGGAGCGCTGTACCTGCGATGAGGCACCCGACCTGGAGAACCGTGAGGCCTGGCAGTGGGGGGG CTGCGGAGACAACCTTAAGTACAGCAGCAAGTTCGTCAAGGAATTCCTGGGCAGACGGTCAAGCAAGGATCTG CGAGCCCGTGTGGACTTCCACAACAACCTCGTGGGTGTGAAGGTGATCAAGGCTGGGGTGGAGACCACCTGCA AGTGCCACGGCGTGTCAGGCTCATGCACGGTGCGGACCTGCTGGCGGCAGTTGGCGCCTTTCCATGAGGTGGG CAAGCATCTGAAGCACAAGTATGAGACGGCACTCAAGGTGGGCAGCACCACCAATGAAGCTGCCGGCGAGGCA GGTGCCATCTCCCCACCACGGGGCCGTGCCTCGGGGGCAGGTGGCAGCGACCCGCTGCCCCGCACTCCAGAGC TGGTGCACCTGGATGACTCGCCTAGCTTCTGCCTGGCTGGCCGCTTCTCCCCGGGCACCGCTGGCCGTAGGTG CCACCGTGAGAAGAACTGCGAGAGCATCTGCTGTGGCCGCGGCCATAACACACAGAGCCGGGTGGTGACAAGG CCCTGCCAGTGCCAGGTGCGTTGGTGCTGCTATGTGGAGTGCAGGCAGTGCACGCAGCGTGAGGAGGTCTACA CCTGCAAGGGCTGAGTTCCCAGGCCCTGCCAGCCCTGCTGCACAGGGTGCAGGCATTGCACACGGTGTGAAGG GTCTACACCTGCACAGGCTGAGTTCCTGGGCTCGACCAGCCCAGCTGCGTGGGGTACAGGCATTGCACACAGT
GTGAATGGGTCTACACCTGCATGGGCTGAGTCCCTGGGCTCAGACCTAGCAGCGTGGGGTAGTCCCTGGGCTC
AGTCCTAGCTGCATGGGGTGCAGGCATTGCACAGAGCATGAATGGGCCTACACCTGCCAAGGCTGAATCCCTG
GGCCCAGCCAGCCCTGCTGCACATGGCACAGGCATTGCACACGGTGTGAGGAGTGTACACCTGCAAGGGCTGA
GGCCCTGGGCCCAGTCAGCCCTGCTGCTCAGAGTGCAGGCATTGCACATGGTGTGAGAAGGTCTACACCTGCA
AGGGACGAGTCCCCGGGCCTGGCCAACCCTGCTGTGCAGGGTGAGGGCCATGCATGCTAGTATGAGGGGTCTA
CACCTGCAAGGACTGAGAGGCTTTT
Figure imgf000174_0001
A ClustalW comparison of the above protein sequences yields the following sequence alignment shown in Table 7B. Table 7B. Comparison of the NOV7 protein sequences.
NOV7a MAPLGYFLLLCSLKQALGSYPIWWLTGSEPLTILPLTLEPEAAAQAHYKACD
NOV7b GSSYPI LTGSEPLTILPLTLEPEAGAQAHYKACD
NOV7c GSSYPI WLTGSEPLTILPLTLEPEAAAQAHYKACD
NOV7d MALLYSSLGWVCTCSPSYFGLTGSEPLTILPLTLEPEAAAQAHYKACD
NOV7e SYPI LTGSEPLTILPLTLEPEAAAQAHYKACD
NOV7f MLDGSPLAR LAAAFGLTLLLAALRPSAAYFGLTGSEPLTILPLTLEPEAAAQAHYKACD
NOV7a RLKLERKQRRMCRRDPGVAETLVEAVSMSALECQFQFRFERWNCTLEGRYRASLLKRGFK NOV7b RL LERKQRRMCRRDPGWETLVEAVSMSALECQFQFRFERWNCTLEGRYRASLLKRGFK NOV7c RLKLERKQRRMCRRDPGVAETLVEAVSMSALECQFQFRFERWNCTLEGRYRASLLKRGFK NOV7d RLKLERKQRRMCRRDPGVAETLVEAVSMSALECQFQFRFERWNCTLEGRYRASLLKRGFK NOV7e RLKLERKQRRMCRRDPGVAETLVEAVSMSALECQFQFRFERWNCTLEGRYRASLLKRGFK NOV7f RLKLERKQRRMCRRDPGVAETLVEAVSMSALECQFQFRFERWNCTLEGRYRASLLKRGFK
NOV7a ETAFLYAISSAGLTHALAKACSAGRMERCTCDEAPDLENREAWQ GGCGDNLKYSSKFVK NOV7b ETAFLYAISSAGLTHALAKACSAGRME'RCTCDEAPDLENREA QWGGCGDNLKYSSKFVK NOV7c ETAFLYAISSAGLTHALAKACSAGRMERCTCDEAPDLENREAWQWGGCGDNLKYSSKFVK NOV7d ETAFLYAISSAGLTHALAKACSAGRMERCTCDEAPDLENREG K GGCSEDIEFGGMVSR NOV7e ETAFLYAISSAGLTHALAKACSAGRMERCTCDEAPDLENREA QWGGCGDNLKYSSKFVK NOV7f ETAFLYAISSAGLTHALAKACSAGRMERCTCDEAPDLENREAWQWGGCGDNLKYSSKFVK
NOV7a EFLGRRSSK-DLRARVDFHNNLVGVKVIKAGVETTCKCHGVSGSCTVRTC RQLAPFHEV NOV7b EFLGRRSSK-DLRARVDFHNNLVGVKVIKAGVETTCKCHGVSGSCTVRTCWRQLAPFHEV NOV7C EFLGRRSSK-DLRARVDFHNNLVGVKVIKAGVETTCKCHGVSGSCTVRTC RQLAPFHEV NOV7d EFADARENRPDARSAMNRHNNEAGRQVIKAGVEΪTCKCHGVSGSCTVRTC RQLAPFHEV NO 7e EFLGRRSSK-DLRARVDFHNNLVGVKVIKAGVETTCKCHGVSGSCTVRTCWRQLAPFHEV NOV7f EFLGRRSSK-DLRARVDFHNNLVGVKVIKAGVETTCKCHGVSGSCTVRTCWRQLAPFHEV
NOV7a GKHLKHKYETALKVGSTTNEAAGEAGAISPPRGRASGAGGSDPLPRTPELVHLDDSPSFC NOV7b GKHLKHKYETALKVGSTTNEAAGEAGAISPPRGRASGAGGSDPLPRTPELVHLDDSPSFC NOV7c GKHLKHKYETALKVGSTTNEAAGEAGAISPPRGRASGAGGSDPLPRTPELVHLDDSPSFC NOV7d GKHLKHKYESALKVGSTTNEAAGEAGAISPPRGRASGAGGSDPLPRTPELVHLDDSPSFC NOV7e GKHLKHKYETALKVGSTTNEAAGEAGAISPPRGRASGAGGSDPLPRTPELVHLDDSPSFC NOV7f GKHLKHKYETALKVGSTTNEAAGEAGAISPPRGRASGAGGSDPLPRTPELVHLDDSPSFC
NOV7a LAGRFSPGTAGRRCHREKNCESICCGRGHNTQSRWTRPCQCQVR CCYVECRQCTQREE NOV7b LAGRFSPGTAGRRCHREKNCESICCGRGHNTQSRWTRPCQCQVR CCYVECRQCTQREE NOV7C LAGRFSPGTAGRRCHREKNCESICCGRGHNTQSRWTRPCQCQVR CCYVECRQCTQREE NOV7d LAGRFSPGTAGRRCHREKNCESICCGRGHNTQSRWTRPCQCQVR CCYVECRQCTQREE NO 7e LAGRFSPGTAGRRCHREKNCESICCGRGHNTQSRWTRPCQCQVRWCCYVECRQCTQREE NOV7f LAGRFSPGTAGRRCHREKNCESICCGRGHNTQSRWTRPCQCQVRWCCYVECRQCTQREE
NOV7 VYTCKG- - NOV7b VYTCKGVD NOV7C VYTCKGVD NOV7d VYTCKG- - NOV7e VYTCKG- - NOV7f VYTCKG--
NO 7a (SEQ ID NO 90) NOV7b (SEQ ID NO 92) NOV7c (SEQ ID NO 94) NOV7d (SEQ ID NO 96) NOV7e (SEQ ID NO 98) NOV7f (SEQ ID NO 100) Further analysis of the NOV7a protein yielded the following properties shown in Table 7C.
Table 7C. Protein Sequence Properties NOV7a
SignalP analysis: Cleavage site between residues 19 and 20
PSORT H analysis:
PSG: a new signal peptide prediction method
N-region: length 0; pos.chg 0; neg.chg 0 H-region: length 13; peak value 9.00 PSG score: 4.60
GvH: von Heijne's method for signal seq. recognition GvH score (threshold: -2.1): 0.73 possible cleavage site: between 18 and 19
»> Seems to have a cleavable signal peptide (1 to 18)
ALOM: Klein et al ' s method for TM region allocation Init position for calculation: 19
Tentative number of TMS(s) for the threshold 0.5: 0 number of TMS(s) .. fixed PERIPHERAL Likelihood = 3.76 (at 114) ALOM score: 3.76 (number of TMSs: 0)
MTOP: Prediction of membrane topology (Hartmann et al . ) Center position for calculation: 9 Charge difference: 0.0 C( 1.0) - N( 1.0) N >= C: N-terminal side will be inside
MITDISC: discrimination of mitochondrial targeting seq R content: 0 Hyd Moment (75): 1.56 Hyd Moment (95) : 3.50 G content: 3 D/E content: 1 S/T content: 4 Score: -6.15
Gavel : prediction of cleavage sites for mitochondrial preseq cleavage site motif not found
NUCDISC: discrimination of nuclear localization signals pat4 : none pat7 : none bipartite: none content of basic residues: 14.8% NLS Score: -0.47
KDEL: ER retention motif in the C-terminus : none
ER Membrane Retention Signals :
KKXX-like motif in the C-terminus : YTCK
SKL: peroxisomal targeting signal in the C-terminus : none
PTS2 : 2nd peroxisomal targeting signal: none
VAC: possible vacuolar targeting motif: none
RNA-binding motif: none
Actinin-type actin-binding motif : type 1: none type : none
NMYR: N-myristoylation pattern : none
Prenylation motif: none memYQRL: transport motif from cell surface to Golgi: none
Tyrosines in the tail : none
Dileucine motif in the tail : none checking 63 PROSITE DNA binding motifs: none checking 71 PROSITE ribosomal protein motifs: none checking 33 PROSITE prokaryotic DNA binding motifs: none
NNCN: Reinhardt's method for Cytoplasmic/Nuclear discrimination Prediction: nuclear Reliability: 70.6
COIL-. Lupas's algorithm to detect coiled-coil regions total : 0 residues
Final Results (k = 9/23) :
55.6 %: extracellular, including cell wall
22.2 % : mitochondrial
11.1 %: vacuolar
11.1 %: nuclear
» prediction for CG53054-02 is exc (k=9)
A search of the NOV7a protein against the Geneseq database, a proprietary database that contains sequences published in patents and patent publication, yielded several homologous proteins shown in Table 7D.
Figure imgf000178_0001
In a BLAST search of public sequence databases, the NOV7a protein was found to have homology to the proteins shown in the BLASTP data in Table 7E.
Figure imgf000178_0002
PFam analysis predicts that the NOV7a protein contains the domains shown in the Table 7F.
Figure imgf000179_0002
Example 8.
The NOV8 clone was analyzed, and the nucleotide and encoded polypeptide sequences are shown in Table 8A.
Table 8A. NOV8 Sequence Analysis
NOV8a, CG53473-02 SEQ ID NO: 101 514 bp DNA Sequence ORF Start: ATG at 37 [ORF Stop: TGA at 400
CGCGCGCCCGAACGAAGCCGCGGCCCGGGCACAGCCATGGCCCGGCGGGCGGGGGGCGCTCGGATGTTCGGCA
GCCTCCTGCTCTTCGCCCTGCTCGCTGCCGGCGTCGCCCCGCTCAGCTGGGATCTCCCGGAGCCCCGCAGCCG AGCCAGCAAGATCCGAGTGCACTCGCGAGGCAACCTCTGGGCCACCGGTCACTTCATGGGCAAGAAGAGTCTG GAGCCTTCCAGCCCATCCCCATTGGGGACAGCTCCCCACACCTCCCTGAGGGACCAGCGACTGCAGCTGAGTC ATGATCTGCTCGGAATCCTCCTGCTAAAGAAGGCTCTGGGCGTGAGCCTCAGCCGCCCCGCACCCCAAATCCA GTACAGGAGGCTGCTGGTACAAATACTGCAGAAATGACACCAATAATGGGGCAGACACAACAGCGTGGCTTAG ATTGTGCCCACCCAGGGAAGGTGCTGAATGGGACCCTGTTGATGGCCATCAACAGGGTCCCATTCAGCACAGG
CTG
NOV8a, CG53473-02 SEQ ID NO: 102 121 aa MW at 13251.4kD Protein Sequence
MARRAGGARMFGSLLLFALLAAGVAPLSWDLPEPRSRASKIRVHSRGNL ATGHFMGKKSLEPSSPSPLGTAP HTSLRDQRLQLSHDLLGILLLKKALGVSLSRPAPQIQYRRLLVQILQK
Figure imgf000179_0001
NOV8b, CG53473-01 SEQ ID NO: 104 112 aa MW at 12402.5kD Protein Sequence
MFGSLLHFALLAAGWPLSWDLPEPRSRASKIRVHSRGKLWAIGHFMGKKSLEPSSPSPLGTAPHTSLRDQRL QLSHDLLGILLLKKALGVSLSRPAPQIQYRRLLVQILQK
NOV8c, CG53473-03 SEQ ID NO: 105 30 bp DNA Sequence ORF Start: at 1 ORF Stop: end of sequence
GGCAAGCTCTGGGCCATCGGTCACTTCATG
NOV8c, CG53473-03 SEQ ID NO: 106 10 aa MW at ll59.4kD Protein Sequence
GKLWAIGHFM
NOV8d, SNP13376396 of SEQ ID NO: 107 514 bp CG53473-02, DNA Sequence ORF Start: ATG at 37 ORF Stop: TGA at 400
SNP Pos: 190 SNP Change: A to G
CGCGCGCCCGAACGAAGCCGCGGCCCGGGCACAGCCATGGCCCGGCGGGCGGGGGGCGCTCGGATGTTCGGCA
GCCTCCTGCTCTTCGCCCTGCTCGCTGCCGGCGTCGCCCCGCTCAGCTGGGATCTCCCGGAGCCCCGCAGCCG AGCCAGCAAGATCCGAGTGCACTCGCGAGGCAACCTCTGGGCCGCCGGTCACTTCATGGGCAAGAAGAGTCTG GAGCCTTCCAGCCCATCCCCATTGGGGACAGCTCCCCACACCTCCCTGAGGGACCAGCGACTGCAGCTGAGTC ATGATCTGCTCGGAATCCTCCTGCTAAAGAAGGCTCTGGGCGTGAGCCTCAGCCGCCCCGCACCCCAAATCCA GTACAGGAGGCTGCTGGTACAAATACTGCAGAAATGACACCAATAATGGGGCAGACACAACAGCGTGGCTTAG ATTGTGCCCACCCAGGGAAGGTGCTGAATGGGACCCTGTTGATGGCCATCAACAGGGTCCCATTCAGCACAGG
CTG
NOV8d, SNP13376396 of CG53473-02, Protein Sequence g^ro^^θ ^"2Ta7~]MW^ 322'Ϊ^D ZI
MARRAGGARMFGSLLLFALLAAGVAPLSWDLPEPRSRASKIRVHSRGNLWAAGHFMGKKSLEPSSPSPLGTAP HTSLRDQRLQIiSHDLLGILLLKKALGVSLSRPAPQIQYRRLLVQILQK
NOV8e, SNP13376395 of SEQ ID NO: 109 514 bp CG53473-02, DNA Sequence ORF Start: ATG at 37 ORF Stop: TGA at 400
SNP Pos: 253 |SNP Change: C to A
CGCGCGCCCGAACGAAGCCGCGGCCCGGGCACAGCCATGGCCCGGCGGGCGGGGGGCGCTCGGATGTTCGGCA
GCCTCCTGCTCTTCGCCCTGCTCGCTGCCGGCGTCGCCCCGCTCAGCTGGGATCTCCCGGAGCCCCGCAGCCG AGCCAGCAAGATCCGAGTGCACTCGCGAGGCAACCTCTGGGCCACCGGTCACTTCATGGGCAAGAAGAGTCTG GAGCCTTCCAGCCCATCCCCATTGGGGACAGCTACCCACACCTCCCTGAGGGACCAGCGACTGCAGCTGAGTC ATGATCTGCTCGGAATCCTCCTGCTAAAGAAGGCTCTGGGCGTGAGCCTCAGCCGCCCCGCACCCCAAATCCA GTACAGGAGGCTGCTGGTACAAATACTGCAGAAATGACACCAATAATGGGGCAGACACAACAGCGTGGCTTAG ATTGTGCCCACCCAGGGAAGGTGCTGAATGGGACCCTGTTGATGGCCATCAACAGGGTCCCATTCAGCACAGG
CTG
!NOV8e, SNP13376395 of SEQ ID NO: 110 121 aa JMW "at 13255.4kD~" CG53473-02, Protein Sequence SNP Pos: 73 |SNP Change: Pro to Thr
MARRAGGARMFGSLLLFALLAAGVAPLSWDLPEPRSRASKIRVHSRGNLWATGHFMGKKSLEPSSPSPLGTAT HTSLRDQRLQLSHDLLGILLLKKALGVSLSRPAPQIQYRRLLVQILQK
NOV8f, SNP13376394 of SEQ ID NO: 111 1514 bp CG53473-02, DNA Sequence θRF Start: ATG at 37 JORF Stop: TAA at 400
1SNP Pos: 401 " " SNP Change: G to A
CGCGCGCCCGAACGAAGCCGCGGCCCGGGCACAGCCATGGCCCGGCGGGCGGGGGGCGCTCGGATGTTCGGCA
GCCTCCTGCTCTTCGCCCTGCTCGCTGCCGGCGTCGCCCCGCTCAGCTGGGATCTCCCGGAGCCCCGCAGCCG AGCCAGCAAGATCCGAGTGCACTCGCGAGGCAACCTCTGGGCCACCGGTCACTTCATGGGCAAGAAGAGTCTG GAGCCTTCCAGCCCATCCCCATTGGGGACAGCTCCCCACACCTCCCTGAGGGACCAGCGACTGCAGCTGAGTC ATGATCTGCTCGGAATCCTCCTGCTAAAGAAGGCTCTGGGCGTGAGCCTCAGCCGCCCCGCACCCCAAATCCA GTACAGGAGGCTGCTGGTACAAATACTGCAGAAATAACACCAATAATGGGGCAGACACAACAGCGTGGCTTAG ATTGTGCCCACCCAGGGAAGGTGCTGAATGGGACCCTGTTGATGGCCATCAACAGGGTCCCATTCAGCACAGG
CTG
|NOV8f, SNP13376394 of SEQ ID NO: 112 121 aa MW at 13251.4kD CG53473-02, Protein Sequence jSNP Change: no change
MARRAGGARMFGSLLLFALLAAGVAPLS DLPEPRSRASKIRVHSRGNL ATGHFMGKKSLEPSSPSPLGTAP HTSLRDQRLQLSHDLLGILLLKKALGVSLSRPAPQIQYRRLLVQILQK
A ClustalW comparison of the above protein sequences yields the following sequence alignment shown in Table 8B. Table 8B. Comparison of the NOV8 protein sequences.
NOV8a MARRAGGARMFGSLLLFALLAAGVAPLS DLPEPRSRASKIRVHSRGNL ATGHFMGKKS
NOV8b MFGSLLHFALLAAGWPLS DLPEPRSRASKIRVHSRGKL AIGHFMGKKS
NOVSc GKL AIGHFM
NOV8a LEPSSPSPLGTAPHTSLRDQRLQLSHDLLGILLLKKALGVSLSRPAPQIQYRRLLVQILQ
NOV8b LEPSSPSPLGTAPHTSLRDQRLQLSHDLLGILLLKKALGVSLSRPAPQIQYRRLLVQILQ
NOV8C
NOV8a K
NOV8b K
NOV8c -
NOV8a (SEQ ID NO: 102)
NOV8b (SEQ ID NO: 104)
NOV8c (SEQ ID NO: 106)
Further analysis of the NOV8a protein yielded the following properties shown in Table 8C.
Table 8C. Protein Sequence Properties NOV8a
SignalP analysis: Cleavage site between residues 27 and 28
PSORT II analysis:
PSG: a new signal peptide prediction method
N-region: length 9; pos.chg 3; neg.chg 0 H-region: length 20; peak value 10.93 PSG score: 6.53
GvH: von Heijne's method for signal seq. recognition GvH score (threshold: -2.1): 2.23 possible cleavage site: between 26 and 27
>>> Seems to have a cleavable signal peptide (1 to 26)
ALOM: Klein et al's method for TM region allocation Init position for calculation: 27
Tentative number of TMS(s) for the threshold 0.5: 0 number of TMS(s) .. fixed PERIPHERAL Likelihood = 1.85 (at 87) ALOM score: 1.85 (number of TMSs: 0)
MTOP: Prediction of membrane topology (Hartmann et al . ) Center position for calculation: 13 Charge difference: -1.5 C( 2.5) - N( 4.0) N >= C: N-terminal side will be inside
MITDISC: discrimination of mitochondrial targeting seq R content: 3 Hyd Moment (75): 12.45 Hyd Momen (95): 10.60 G content: 4 D/E content: 1 S/T content: 2 Score: -2.16
Gavel: prediction of cleavage sites for mitochondrial preseq R-2 motif at 19 ARM|FG NUCDISC: discrimination of nuclear localization signals pat4 : none pat7 : none bipartite : none content of basic residues: 14.9% NLS Score: -0.47
KDEL: ER retention motif in the C-terminus : none
ER Membrane Retention Signals :
XXRR-like motif in the N-terminus : ARRA
SKL: peroxisomal targeting signal in the C-terminus: none
PTS2 : 2nd peroxisomal targeting signal: none
VAC: possible vacuolar targeting motif: none
RNA-binding motif: none
Actinin-type actin-binding motif: type 1: none type 2 : none
NMYR: N-myristoylation pattern : none
Prenylation motif: none memYQRL: transport motif from cell surface to Golgi: none
Tyrosines in the tail : none
Dileucine motif in the tail: none checking 63 PROSITE DNA binding motifs: none checking 71 PROSITE ribosomal protein motifs: none checking 33 PROSITE prokaryotic DNA binding motifs: none
HNCN: Reinhardt's method for Cytoplasmic/Nuclear discrimination Prediction: nuclear Reliability: 89
COIL: Lupas's algorithm to detect colled-coil regions total: 0 residues
Final Results (k = 9/23) :
44.4 %: extracellular, including cell wall 33.3 %: mitochondrial 22.2 %: nuclear
» prediction for CG53473-02 is exc (k=9)
A search of the NOV8a protein against the Geneseq database, a proprietary database that contains sequences published in patents and patent publication, yielded several homologous proteins shown in Table 8D.
Figure imgf000184_0001
In a BLAST search of public sequence databases, the NOV8a protein was found to have homology to the proteins shown in the BLASTP data in Table 8E.
Figure imgf000185_0001
PFam analysis predicts that the NOVδa protein contains the domains shown in the Table 8F.
Figure imgf000185_0002
Example 9.
The NOV9 clone was analyzed, and the nucleotide and encoded polypeptide sequences are shown in Table 9A. Table 9A. NOV9 Sequence Analysis
NOV9a, CG55184-03 1SEQ ID NO: 113 614 bp
DNA Sequence |QKP Start: ATG at 4 {ORF Stop: TAG at 607
ACCATGGGCTCCGGGCGCCGGGCGCTGTCCGCGGTGCCGGCCGTGCTGCTGGTCCTCACGCTGCCGGGGCTGC
CCGTCTGGGCACAGAACGACACGGAGCCCATCGTGCTGGAGGGCAAGTGTCTGGTGGTGTGCGACTCGAACCC GGCCACGGACTCCAAGGGCTCCTCTTCCTCCCCGCTGGGGATATCGGTCCGGGCGGCCAACTCCAAGGTCGCC TTCTCGGCGGTGCGGAGCACCAACCACGAGCCATCCGAGATGAGCAACAAGACGCGCATCATTTACTTCGATC AGATCCTGGTGAATGTGGGTAATTTTTTCACATTGGAGTCTGTCTTTGTAGCACCAAGAAAAGGAATTTACAG TTTCAGTTTTCACGTGATTAAAGTCTACCAGAGCCAAACTATCCAGGTTAACTTGATGTTAAATGGAAAACCA GTAATATCTGCCTTTGCGGGGGACAAAGATGTTACTCGTGAAGCTGCCACGAATGGTGTCCTGCTCTACCTAG ATAAAGAGGATAAGGTTTACCTAAAACTGGAGAAAGGTAATTTGGTTGGAGGCTGGCAGTATTCCACGTTTTC TGGCTTTCTGGTGTTCCCCCTATAGGATTC
NOV9a, CG55184-03 SEQ ID NO: 114 201 aa MW at 21807.9 D Protein Sequence
MGSGRRALSAVPAVLLVLTLPGLPVWAQNDTEPIVLEGKCLWCDSNPATDSKGSSSSPLGISVRAANSKVAF SAVRSTNHEPSEMSNKTRIIYFDQILVNVGNFFTLESVFVAPRKGIYSFSFHVIKVYQSQTIQVNLMLNGKPV ISAFAGDKDVTREAATNGVLLYLDKEDKVYLKLEKGNLVGGWQYSTFSGFLVFPL
|NOV9b, CG55184-01 }SEQ IDNO: 115 |614 bp jDNA Sequence ORF Start: ATG at 4 iORF Stop: TAG at 607
ACCATGGGCTCCGGGCGCCGGGCGCTGTCCGCGGTGCCGGCCGTGCTGCTGGTCCTCACGCTGCCGGGGCTGC
CCGTCTGGGCACAGAACGACACGGAGCCCATCGTGCTGGAGGGCAAGTGTCTGGTGGTGTGCGACTCGAACCC GGCCACGGACTCCAAGGGCTCCTCTTCCTCCCCGCTGGGGATATCGGTCCGGGCGGCCAACTCCAAGGTCGCC TTCTCGGCGGTGCGGAGCACCAACCACGAGCCATCCGAGATGAGCAACAAGACGCGCATCATTTACTTCGATC AGATCCTGGTGAATGTGGGTAATTTTTTCACATTGGAGTCTGTCTTTGTAGCACCAAGAAAAGGAATTTACAG TTTCAGTTTTCACGTGATTAAAGTCTACCAGAGCCAAACTATCCAGGTTAACTTGATGTTAAATGGAAAACCA GTAATATCTGCCTTTGCGGGGGACAAAGATGTTACTCGTGAAGCTGCCACGAATGGTGTCCTGCTCTACCTAG ATAAAGAGGATAAGGTTTACCTAAAACTGGAGAAAGGTAATTTGGTTGGAGGCTGGCAGTATTCCACGTTTTC TGGCTTTCTGGTGTTCCCCCTATAGGATTC
NOV9b, CG55184-01 jSEQ ID NO: 116 J201 aa MWat21807.9kD
Protein Sequence
MGSGRRALSAVPAVLLVLTLPGLPV AQNDTEPIVLEGKCLWCDSNPATDSKGSSSSPLGISVRAANSKVAF SAVRSTNHEPSEMSNKTRIIYFDQILVNVGNFFTLESVFVAPRKGIYSFSFHVIKVYQSQTIQVNLMLNGKPV ISAFAGD DVTREAATNGVLLYLDKEDKVYLKLE GNLVGGWQYSTFSGFLVFPL
Figure imgf000187_0001
NOV9c, CG55184-02 SEQ ID NO: 118 174 aa MW at 19080.6kD Protein Sequence
QNDTEPIVLEGKCLWCDSNPATDSKGSSSSPLGISVRAANSKVAFSAVRSTNHEPSE SNKTRIIYFDQILV NVGNFFTLESVFVAPRKGIYSFSFHVIKVYQSQTIQVNLMLNGKPVISAFAGDKDVTREAATNGVLLYLDKED KVYLKLEKGNLVGG QYSTFSGFLVFPL "
ΪNOV9d, CG55184-04 SEQ ID NO: 119 48 bp DNA Sequence ORF Start: at 1 jORF Stop: end of sequence
GCGGCCAACTCCAAGGTCGCCTTCTCGGCGGTGCGGAGCACCAACCAC
Figure imgf000187_0002
|NOV9e, CG55184-05 SEQ ID NO: 122 15 aa MW at l588.7kD Protein Sequence
ANSKVAFSAVRSTNH
A ClustalW comparison of the above protein sequences yields the following sequence alignment shown in Table 9B. Table 9B. Comparison of the NOV9 protein sequences.
NOV9a MGSGRRALSAVPAVLLVX.TLPGLPV AQNDTEPIVLEGKCLWCDSNPATDSKGSSSSPL
NOV9b MGSGRRALSAVPAVLLVLTLPGLPV AQNDTEPIVLEG CLWCDSNPATDSKGSSSSPL
NOV9c QNDTEPIVLEGKCLWCDSNPATDSKGSSSSPL
NOV9d
NOV9e
NOV9a GISVRAANSKVAFSAVRSTNHEPSEMSNKTRIIYFDQILVNVGNFFTLESVFVAPRKGIY NOV9b GISVRAANSKVAFSAVRSTNHEPSEMSNKTRIIYFDQILVNVGNFFTLESVFVAPR GIY NOV9c GISVRAANSKVAFSAVRSTNHEPSEMSNKTRIIYFDQILVNVGNFFTLESVFVAPRKGIY NOV9d AANS VAFSAVRSTNH NOV9e ANSKVAFSAVRSTNH
NOV9 SFSFHVIKVYQSQTIQVNLMLNGKPVISAFAGDKDVTREAATNGVLLYLDKEDKVYLKLE NOV9b SFSFHVIKVYQSQTIQVNl-røjNGKPVISAFAGDKDVTREAATNGVLLYLDKEDKVYLKLE NOV9c SFSFHVIKVYQSQTIQVNLMLNGKPVISAFAGDKDVTREAATNGVLLYLDKED VYLKLE NOV9d NOV9e
NOV9a GNLVGGWQYSTFSGFLVFPL NOV9b KGNLVGGWQYSTFSGFLVFPL NOV9c KGNLVGGWQYSTFSGFLVFPL
NOV9e NOV9a (SEQ ID NO 114)
NOV9b (SEQ ID NO 116)
NOV9c (SEQ ID NO 118)
NOV9d (SEQ ID NO 120)
NOV9e (SEQ ID NO 122)
Further analysis of the NOV9a protein yielded the following properties shown in Table 9C.
Table 9C. Protein Sequence Properties NOV9a
SignalP analysis: Cleavage site between residues 28 and 29
PSORT II analysis:
PSG: a new signal peptide prediction method
N-region: length 6; pos . chg 2 ; neg. chg 0 H-region: length 23 ; peak value 10.04 PSG score: 5.64
GvH-. von Heijne ' s method for signal seq. recognition GvH score (threshold: -2.1) : 0.95 possible cleavage site : between 27 and 28
>>> Seems to have a cleavable signal peptide (1 to 27)
ALOM: Klein et al ' s method for TM region allocation Init position for calculation: 28 Tentative number of TMS (s) for the threshold 0.5 : Number of TMS(s) for threshold 0.5: 0 PERIPHERAL Likelihood = 5.67 (at 60) ALOM score: 0.10 (number of TMSs: 0)
MTOP: Prediction of membrane topology (Hartmann et al.) Center position for calculation: 13 Charge difference: -5.0 C(-2.0) - N( 3.0) N >= C: N-terminal side will be inside
MITDISC: discrimination of mitochondrial targeting seq R content: 2 Hyd Moment (75): 11.01 Hyd Moment (95) : 9.83 G content: 3 D/E content: 1 S/T content: 3 Score: -2.58
Gavel : prediction of cleavage sites for mitochondrial preseq R-2 motif at 16 RRA|LS
NUCDISC: discrimination of nuclear localization signals pat4: none pat7 : none bipartite: none content of basic residues: 9.5% NLS Score: -0.47
KDEL: ER retention motif in the C-terminus : none
ER Membrane Retention Signals :
XXRR-like motif in the N-terminus : GSGR none
SKL: peroxisomal targeting signal in the C-terminus: none
PTS2 : 2nd peroxisomal targeting signal: none
VAC: possible vacuolar targeting motif: none
RNA-binding motif: none
Actinin-type actin-binding motif: type 1 : none type 2 : none
NMYR: N-myristoylation pattern : none
Prenylation motif: none memYQRL: transport motif from cell surface to Golgi: none
Tyrosines in the tail : none
Dileucine motif in the tail : none checking 63 PROSITE DNA binding motifs: none checking 71 PROSITE ribosomal protein motifs: none checking 33 PROSITE prokaryotic DNA binding motifs: none
NNCN: Reinhardt's method for Cytoplasmic/Nuclear discrimination Prediction: cytoplasmic Reliability: 94.1
COIL: Lupas's algorithm to detect coiled-coil regions total : 0 residues Final Results (k = 9/23) :
33.3 % extracellular, including cell wall 33.3 % mitochondrial 22.2 % endoplasmic reticulum 11.1 % Golgi
» prediction for CG55184-03 is exc (k=9)
A search of the NOV9a protein against the Geneseq database, a proprietary database that contains sequences published in patents and patent publication, yielded several homologous proteins shown in Table 9D.
Figure imgf000190_0001
In a BLAST search of public sequence databases, the NOV9a protein was found to have homology to the proteins shown in the BLASTP data in Table 9E.
Figure imgf000191_0001
PFam analysis predicts that the NOV9a protein contains the domains shown in the Table 9F.
Figure imgf000191_0002
Example 10.
The NOV10 clone was analyzed, and the nucleotide and encoded polypeptide sequences are shown in Table 10A.
Figure imgf000191_0003
NOVlOa, CG55274-05 SEQ ID NO: 124 86 aa MW at 9590.0kD Protein Sequence ALQAEFDKAAEDVRKLPTRPADNKELKKLDGLYKQAIIGDINIEYLGMLDLKGKAKCAAWTLQKRLSKEDAT SVSISKAKEPIEK
Figure imgf000192_0001
NOVlOb, CG55274-01 SEQ ID NO: 126 86 aa MW at 9624.1kD Protein Sequence
MALQAEFDKAAEDVRKLPTRPADNKELKKLDGLYKQAIIGDINIEYLGMLDFKGKAKCAA TLQKRLSKEDAT SVSISKAKEPIEK
NOVlOc, CG55274-02 SEQ ID NO: 127 289 bp DNA Sequence ORF Start: ATG at 17 |ORF Stop: TAG at 272
ITGCGGCCGCCACCACCATGGCACTGCAGGCTGATCGAGACAAGGCTGCAGAAGACGTGAGGAAGCTGCCAACA
AGACCAGATGAGAAAGAACTGAAAAAACTCGATGGACTTTACAAACAAGCTATAATTGGAGACATTAATATTG AGTATCTGGGAATGCTGGATTTAAAGGGCAAGGCCAAATGCGCAGCATGGACCCTCCAAAAAAGGTTGTCAAA GGAAGATGCAACGAGTGTCTCTATTTCTAAGGCAAAAGAGCCGATAGAAAAATAGGACATTTAGAATACA
NOVlOc, CG55274-02 SEQ ID NO: 128 85 aa MW at 9528.9kD Protein Sequence
MALQADRDKAAEDVRKLPTRPDEKELKKLDGLYKQAIIGDINIEYLGMLDLKGKAKCAAWTLQKRLSKEDATS VSISKAKEPIEK
NOVlOd, CG55274-03 SEQ ID NO: 129 60 bp DNA Sequence ORF Start: at 1 ORF Stop: end of sequence
CAAGCTATAATTGGAGACATTAATATTGAGTATCTGGGAATGCTGGATTTAAAGGGCAAG
NOVlOd, CG55274-03 SEQ ID NO: 130 20 aa |MW at 2204.6kD Protein Sequence
QAI I GDINI EYLGMLDLKGK
Figure imgf000193_0001
pNTOVlOe, CG55274-04 SEQ ID NO: 132 18 aa MW at 2053.4kD jProtein Sequence
QAI IGDINIEYLGMLDFK
A ClustalW comparison of the above protein sequences yields the following sequence alignment shown in Table 10B.
Table 10B. Comparison of the NOV10 protein sequences.
NOVlOa MALQAEFDKAAEDVRKLPTRPADNKELKKLDGLYKQAIIGDINIEYLGMLDLKGKAKCAA
NOVl0b ALQAEFDKAAEDVRKLPTRPADNKELKKLDGLYKQAIIGDINIEYLGMLDFKGKAKCAA
NOVl0c MALQADRDKAAEDVRKLPTRPDE-KELKKLDGLYKQAIIGDINIEYLGMLDLKGKAKCAA
NOVlOd QAIIGDINIEYLGMLDLKGK
NOVlOe QAIIGDINIEYLGMLDFK
NOVlOa TLQKRLSKEDATSVSISKAKEPIEK
NOVlOb WTLQKRLSKEDATSVSISKAKEPIEK
NOVlOc WTLQKRLSKEDATSVSISKAKEPIEK
NOVlOd
NOVlOe
NOVlOa (SEQ ID NO 124) NOVlOb (SEQ ID NO 126) NOVlOc (SEQ ID NO 128) NOVlOd (SEQ ID NO 130) NOVlOe (SEQ ID NO 132)
Further analysis of the NOVlOa protein yielded the following properties shown in Table IOC.
Table IOC. Protein Sequence Properties NOVlOa
SignalP analysis: No Known Signal Sequence Predicted
PSORT II analysis:
PSG: a new signal peptide prediction method
N-region: length 9; pos.chg 1; neg.chg 2 H-region: length 2; peak value 0.00 PSG score: -4.40
GvH: von Heijne's method for signal seq. recognition GvH score (threshold: -2.1) : -10.68 possible cleavage site: between 58 and 59 >>> Seems to have no N-terminal signal peptide
ALOM: Klein et al ' s method for TM region allocation Init position for calculation: 1
Tentative number of TMS(s) for the threshold 0.5: 0 number of TMS(s) .. fixed PERIPHERAL Likelihood = 7.11 (at 36) ALOM score: 7.11 (number of TMSs: 0)
MITDISC: discrimination of mitochondrial targeting seq R content: 0 Hyd Moment (75) : 3.71 Hyd Moment (95): 2.95 G content: 0 D/E content: 2 s/T content: 0 Score: -7.75
Gavel: prediction of cleavage sites for mitochondrial preseq cleavage site motif not found
NUCDISC: discrimination of nuclear localization signals pat4 : none pat7: none bipartite : none content of basic residues: 19.8% NLS Score: -0.47
KDEL: ER retention motif in the C-terminus : none
ER Membrane Retention Signals : none
SKL: peroxisomal targeting signal in the C-terminus : none
PTS : 2nd peroxisomal targeting signal : none
VAC: possible vacuolar targeting motif: none
RNA-binding motif: none
Actinin-type actin-binding motif: type 1 : none type 2 : none
NMYR: N-myristoylation pattern : none
Prenylation motif: none memYQRL: transport motif from cell surface to Golgi: none
Tyrosines in the tail: none
Dileucine motif in the tail: none checking 63 PROSITE DNA binding motifs: none checking 71 PROSITE ribosomal protein motifs: none checking 33 PROSITE prokaryotic DNA binding motifs: none
NNCN: Reinhardt's method for Cytoplasmic/Nuclear discrimination Prediction: nuclear Reliability: 76.7
COIL: Lupas's algorithm to detect coiled-coil regions total : 0 residues Final Results (k = 9/23) :
82.6 % : nuclear
4.3 % : cytoskeletal
4.3 % : mitochondrial
4 .3 % : cytoplasmic
4.3 % : peroxisomal
>> prediction for CG55274-05 is nuc (k=23)
A search of the NOVlOa protein against the Geneseq database, a proprietary database that contains sequences published in patents and patent publication, yielded several homologous proteins shown in Table 10D.
Figure imgf000195_0001
In a BLAST search of public sequence databases, the NOVlOa protein was found to have homology to the proteins shown in the BLASTP data in Table 10E.
Figure imgf000196_0001
PFam analysis predicts that the NOVlOa protein contains the domains shown in the Table 10F.
Figure imgf000196_0002
Example 11.
The NOVll clone was analyzed, and the nucleotide and encoded polypeptide sequences are shown in Table 11 A. Table 11A. NOVll Sequence Analysis
NOVlla, CG55379-04 SEQ ID NO: 133 jDNA Sequence ORF Start: ATG at 1 ORF Stop: TAG at 3763
ATGGCGCGGGGGGACGCCGGCCGCGGCCGCGGGCTCCTCGCGTTGACCTTCTGCCTGTTGGCCGCGCGCGGGG
AGCTGCTGTTGCCCCAGGAGACGACTGTGGAGCTGAGCTGTGGAGTGGGGCCACTGCAAGTGATCCTGGGCCC
AGAGCAGGCTGCAGTGCTAAACTGTAGCCTGGGGGCTGCTGCCGCTGGACCCCCCACCAGGGTGACCTGGAGC
AAGGATGGGGACACCCTGCTGGAGCACGACCACTTACACCTGCTGCCCAATGGTTCCCTGTGGCTGTCCCAGC
CACTAGCACCCAATGGCAGTGACGAGTCAGTCCCTGAGGCTGTGGGGGTCATTGAAGGCAACTATTCGTGCCT
AGCCCACGGCCCCCTCGGAGTGCTGGCCAGCCAGACTGCTGTCGTCAAGCTTGCCAGTCTCGCAGACTTCTCT
CTGCACCCGGAGTCTCAGACGGTGGAGGAGAACGGGACAGCTCGCTTTGAGTGCCACATTGAAGGGCTGCCAG
CTCCCATCATTACTTGGGAGAAGGACCAGGTGACATTGCCTGAGGAGCCTCGGCTCATCGTGCTTCCCAACGG
CGTCCTTCAGATCCTGGATGTTCAGGAGAGTGATGCAGGCCCCTACCGCTGCGTGGCCACCAACTCAGCTCGC
CAGCACTTCAGCCAGGAGGCCCTACTCAGTGTGGCCCACAGAGGTTCCCTGGCGTCCACCAGGGGGCAGGACG
TGGTCATTGTGGCAGCCCCAGAGAACACCACAGTGGTGTCTGGCCAGAGTGTGGTGATGGAATGTGTGGCCTC
AGCTGACCCCACCCCTTTTGTGTCCTGGGTCCGAGACGGGAAGCCCATCTCCACAGATGTCATCGTCCTGGGC
CGCACCAACCTACTAATTGCCAACGCGCAGCCCTGGCACTCCGGCGTCTATGTCTGCCGCGCCAACAAGCCCC
GCACGCGCGACTTCGCCACTGCAGCCGCTGAGCTCCGTGTGCTGCTAGCGGCTCCCGCCATCACTCAGGCGCC
CGAGGCGCTGTCGCGGACGCGGGCGAGCACAGCGCGCTTCGTGTGCCGCGCGTCGGGGGAGCCGCGGCCAGCG
CTGCGCTGGCTGCACAACGGGGCGCCGCTGCGGCCCAACGGGCGCGTCAAGGTCCAGGGCGGCGGTGGCAGCC
TGGTCATCACACAGATCGGCCTGCAGGACGCCGGCTACTACCAGTGCGTGGCTGAGAACAGCGCGGGAATGGC
GTGCGCTGCCGCGTCGCTGGCCGTGGTGGTGCGCGAGGGGCTGCCCAGCGCCCCCACGCGGGTCACTGCTACG
CCACTGAGCAGCTCCGCTGTGTTGGTGGCCTGGGAGCGGCCCGAGATGCACAGCGAGCAGATCATCGGCTTCT
CTCTCCACTACCAGAAGGCACGGGGTATGGACAATGTGGAATACCAGTTTGCAGTGAACAACGACACCACAGA
ACTACAGGTTCGGGACCTGGAACCCAACACAGATTATGAGTTCTACGTGGTGGCCTACTCCCAGCTGGGAGCC
AGCCGCACCTCCACCCCAGCACTGGTGCACACACTGGATGATGTCCCCAGTGCAGCACCCCAGCTCTCCCTGT
CCAGCCCCAACCCTTCGGACATCAGGGTGGCGTGGCTGCCCCTGCCCCCCAGCCTGAGCAATGGGCAGGTGGT
GAAGTACAAGATAGAATACGGTTTGGGAAAGGAAGGTGAGTGGGGGGATCAGATTTTCTCTACTGAGGTGCGA
GGAAATGAGACACAGCTTATGCTGAACTCGCTTCAGCCAAACAAGGTGTATCGAGTACGGATTTCGGCTGGTA
CAGCAGCCGGCTTCGGGGCCCCCTCCCAGTGGATGCATCACAGGACGCCCAGTATGCACAACCAGAGCCATGT
CCCTTTTGCCCCTGCAGAGTTGAAGGTGCAGGCAAAGATGGAGTCCCTGGTCGTGTCATGGCAGCCACCCCCT
CACCCCACCCAGATCTCTGGCTACAAACTATATTGGCGGGAGGTGGGGGCTGAGGAGGAGGCCAATGGCGATC
GCCTGCCAGGGGGCCGTGGAGACCAGGCTTGGGATGTGGGGCCTGTCCGGCTCAAGAAGAAAGTGAAGCAGTA
TGAGCTGACCCAGCTAGTCCCTGGCCGGCTGTACGAGGTGAAGCTCGTGGCTTTCAACAAACATGAGGATGGC
TATGCAGCAGTGTGGAAGGGCAAGACGGAGAAGGCGCCGGCACCAGACATGCCTATCCAGAGGGGACCACCCC
TGCCTCCAGCCCACGTCCATGCGGAATCAAACAGCTCCACATCCATCTGGCTTCGGTGGAAAAAGCCAGATTT
CACCACAGTCAAGATTGTCAACTACACTGTGCGCTTCAGCCCCTGGGGGCTCAGGAATGCCTCCCTGGTCACC
TATTACACCAGTTCTGGAGAAGACATCCTCATTGGCGGCTTGAAGCCATTCACCAAATACGAGTTTGCAGTGC
AGTCTCACGGCGTGGACATGGATGGGCCTTTCGGCTCTGTGGTGGAGCGCTCCACCCTGCCTGACCGGCCCTC
CACACCCCCATCCGACCTGCGACTGAGCCCCCTGACACCGTCCACGGTTCGGCTGCACTGGTGCCCCCCCACA
GAGCCCAACGGGGAGATCGTGGAGTATCTGATCCTGTACAGCAGCAACCACACGCAGCCTGAGCACCAGTGGA
CCTTGCTCACCACGCAGGGAAACATCTTCAGTGCTGAGGTCCATGGCCTGGAGAGCGACACTCGGTACTTCTT
CAAGATGGGGGCGCGCACAGAGGTGGGACCTGGGCCTTTCTCCCGCCTGCAGGATGTGATCACGCTCCAGGAG
AAGCTGTCAGACTCGCTGGACATGCACTCAGTCACGGGCATCATCGTGGGTGTCTGCCTGGGCCTCCTCTGCC
TCCTGGCCTGCATGTGTGCTGGCCTGCGCCGCAGCCCCCACAGGGAATCCCTCCCAGGCCTGTCCTCCACCGC
CACCCCCGGGAATCCCGCGCTGTACTCCAGAGCTCGGCTTGGCCCCCCCAGCCCCCCAGCTGCCCATGAATTG
GAGTCCCTTGTGCACCCCCATCCCCAGGACTGGTCCCCGCCACCCTCAGACGTGGAGGACAGGGCTGAAGTGC
ACAGCCTTATGGGTGGCGGTGTTTCTGAAGGCCGGAGTCACTCCAAAAGAAAGATCTCCTGGGCTCAACCAAG
CGGGCTGAGCTGGGCTGGTTCCTGGGCAGGCTGTGAGCTGCCCCAGGCAGGCCCCCGGCCGGCTCTGACCCGG
GCCCTGCTGCCCC^TGCTGGAAC^GGCAGACGCTGTTGCTGCAGGC ._________
TCTGGTGTACGACGCCATAAAGGGCAATGGGAGGAAGAAGTCACCCCCAGCCTGCAGGAACCAGGTGGAGGCT
GAAGTCATTGTCCACTCTGACTTTAGTGCATCTAACGGGAACCCTGACCTCCATCTCCAAGACCTGGAGCCTG
AGGACCCCCTGCCTCCAGAGGCTCCTGATCTCATCTCGGGTGTTGGGGATCCAGGGCAGGGGGCAGCCTGGCT
GGACAGGGAGTTGGGAGGGTGTGAGCTGGCAGCCCCCGGGCCAGACAGACTTACCTGCTTGCCAGAGGCAGCC
AGTGCTTCCTGCTCCTACCCGGACCTCCAGCCAGGCGAGGTGCTAGAGGAGACCCCTGGAGATAGCTGCCAGC
TCAAATCCCCCTGCCCTCTAGGAGCCAGCCCAGGCCTGCCCAGATCCCCGGTCTCCTCCTCTGCCTAGCTCTT
CCCAGAGGATGTGGTTTGGGGCAGGCAGGTATGGATCACATAGGATGCGATACCTGTGGCCGTGTATGTCCAC
ATGTGTGCCTGTAGATACATCATCAAGCCCTTTGGAGCTTCCTAAGTTGCTTTGGCTGAGGGGAGAGGAAAAC ATGGATTATTCACTCCCCCCATACTCTTTGTGATACACATGTGACATGTGAAAGACATACGAGACATAGCTAC
ATGTGATGTGCACATGTGTGAAGTGCATGTATGCGTACTGGTTGTTGAGCTGGGAAACCGTGGCCCAGGCAGT
GGTCACTACAGCCTGATTGGTCCTCCAGGTCAGAACGGTGCCCCACAGTGGTCAGTCCCCAGCCCTGTGGGCC
CCCACCTCCATCGCCCAGCCTTTTATTACACACTCTGAGAGTGTCTCCAATGCCTGTCTGACAAAGACAGTCC
CAGCCCATTCTCCTGTCTGGCTGGGTTGGGTGCAAGCAGGCTCTGAATGCCTGGCATTTCAGCTGCATCACCT
CCCAGCTCCTTATTGCCCAAATAGAGAGGGTGGCCCTGGCTCCCCTCCGAGCAACTCTGCATTTAATTTTGTA
ATCTGGGAAGTGCCTGGTTTTGAAAATCCGCTTTCTCTCACTCTTCCCCTCCTTCCTTGCCCCTGGCTGCTCT
AGTGTTCTGTCTCCCAGTCACCTCGCTCTCCCAGCACCAGTGCCCTTCTCCTGCTCCCAGATACTCTTTCCTT
TCCTCTCTCCTGTTTTCCTTCCTCTGCTATCTCTCACACCTCTCCCAGACTATGTCATCTTGTTCTCCTGCCT
GGGTTCAAACTCTGCATCCTTCTCTAACAACGTGACTACCTCATGTCTGCTTCAAGGCCCCCGTGCCCTTCCT
GTATCCGCGGCTGCCGCGCACTCGCCTGCCATCCTCCTGCCTCCTCTTCACTCAGTGCTTCTGCTTGCCCTGC
CCCAGGCAGCCCACCCACGCCCAGTGCGGGTGTGGAGAAGATCTTCTGGCTTCCCTGCATCTTGCCTTTGGGA
TTGGGATCCAAGGGTTCTCCATGGATGGATCCAAGTCATAGAGGGGAATGTTTGAGACAGGGAAGGGGGCTGT
GATCCAGAGGCTCAGAATAAAAAGATGCCCTCCCTTCTATGCAGGGGGGCAAGTTTACTGGATGGAGATGATT
TGGGCCTCTCTTCCAGAAGAAGCTAAAGGAAGAGAAGGGGAGTGAGAGTTCAGGGAGGCCCTTCCCACCCTGT
GAGGCTTGACTTGATCTGGATTGGGGATGACAGGAATCTCACCCTCTGGGGTGCTGGCAAGGAGGTCTTTGCA
CAGGAAAAGGGGTAGCTCATTTCAGTTTGTTTTTTCTTTAAATTGAATCCTCAAGTCATTTTCTGTTCACCTG
CCGCACAGGGACAAGCTTGACTTCTATTTTCTGTGTAGTGAAAACAATGTCATTTATTTGGTTTTTCACCTCA iGCCCTCTCATAGGAGCATAGAATGTTAGGGTCTTTACTCCCTAATGATGTCTGATTGGCACATCAAGAGTTAA
CTCTGCCTTCTGGGCCAAATTCGAAATAACCAGTCCATTTTTCCTTTTTTTTTTTTTTTTTTTTTTTTAAATG
IGTGGAATGTCTCTCAGCACAGTTGCGGCTTCCTCAAACCCTGAAAGCATCTGTGTTTATTATACTCGGGTGTC
ACTCACTGTTGATGTCTGCACCTACGTTTCCACCTCCTCCCCCTCCTTCAGCCAGCCTATGATAACACTAAAG lATTATTAATGTTGGTTTTGTATCTCGTTAAAGACAGAATTGTCACTTGTAGTATTTCTGTAGCATTCAGCGCT
GCTGTGGCTAACACCACTGTGTATGTTTCATCATTGCTCTGAAGGTCAAAAGCCTCATTTTATTTTGCTGGTT
TGATTTTTTTTTTTTAAAGAAGAAAAAAAAACTGCCCTGAATTAAATGGCTGTTTTAACAGTAGGCTCTTAGC lATTATACCACATAGTCATTTTTCATGTTCTTGTTTAACAGGCACTGAGGTTCTGGTTTAAATTAAATAGCTGC AATGAGACAATTTATAACCCATTAGGTTGGGTGGAAAATTGTTTCTCAAAAGCAAATAAGTAATAAATCTGG
TATCTGCCTATAACTCACAGTTGATAAGAAAGTGGCCATTTCTCACTAGCACTATATATGATTTGGGCTCTGG
GTAATTTGGAAGTGTTAGGTTTGTGTCTTTGTAGCAGTATTTTTATTAGAAAAGAATCTATTGGCCTTTTACA
GGGTATTAATCCCTTTGTCACCTACCATTGATGCCTTAAGTTTTCTGAGTCTCAATTAAAAATCTTCCTTTTC
TTGATGCATGACAAGTGTAATCAGTACTTGCTCATTTATTTGTCTGTATTTAGTTTATGCTGTACTATTTAAT
TATCCTTCCAGCGTTTTTTTTTTCTCCTTACAAATATGATACTCTTTAGTGTTAAGCTAAGGCATTGATTCAT
GTATCTGTCCTTATAATGAATTAATAAACTATTTTCCAG
NOVlla, CG55379-04 SEQ ID NO: 134 1254 aa MW at 134608.7kD Protein Sequence ARGDAGRGRGLLALTFCLLAARGELLLPQETTVELSCGVGPLQVILGPEQAAVLNCSLGAAAAGPPTRVTWS JKDGDTLLEHDHLHLLPNGSLWLSQPLAPNGSDESVPEAVGVIEGNYSCLAHGPLGVLASQTAWKLASLADFS LHPESQTVEENGTARFECHIEGLPAPIITWEKDQVTLPEEPRLIVLPNGVLQILDVQESDAGPYRCVATNSAR QHFSQEALLSVAHRGSLASTRGQDWIVAAPENTTWSGQSWMECVASADPTPFVSWVRDGKPISTDVIVLG RTNLLIANAQPWHSGVYVCRANKPRTRDFATAAAELRVLLAAPAITQAPEALSRTRASTARFVCRASGEPRPA LRWLHNGAPLRPNGRVKVQGGGGSLVITQIGLQDAGYYQCVAENSAGMACAAASLAVWREGLPSAPTRVTAT PLSSSAVLVAWERPEMHSEQIIGFSLHYQKARGMDNVEYQFAVNNDTTELQVRDLEPNTDYEFYWAYSQLGA SRTSTPAI.VHTLDDVPSAAPQLSLSSPNPSDIRVAWLPLPPSLSNGQWKYKIEYGLGKEGEWGDQIFSTEVR GNETQLMLNSLQPNKVYRVRISAGTAAGFGAPSQWMHHRTPSMHNQSHVPFAPAELKVQAKMESLWSWQPPP HPTQISGYKLYWREVGAEEEANGDRLPGGRGDQAWDVGPVRLKKKVKQYELTQLVPGRLYEVKLVAFNKHEDG YAAVWKGKTEKAPAPDMPIQRGPPLPPAHVHAESNSSTSIWLRWKKPDFTTVKIVNYTVRFSPWGLRNASLVT YYTSSGEDILIGGLKPFTKYEFAVQSHGVDMDGPFGSWERSTLPDRPSTPPSDLRLSPLTPSTVRLHWCPPT EPNGEIVEYLILYSSNHTQPEHQWTLLTTQGNIFSAEVHGLESDTRYFFKMGARTEVGPGPFSRLQDVITLQE KLSDSLDMHSVTGIIVGVCLGLLCLLACMCAGLRRSPHRESLPGLSSTATPGNPALYSRARLGPPSPPAAHEL ESLVHPHPQDWSPPPSDVEDRAEVHSLMGGGVSEGRSHSKRKISWAQPSGLSWAGSWAGCELPQAGPRPALTR ALLPPAGTGQTLLLQALVYDAIKGNGRKKSPPACRNQVEAEVIVHSDFSASNGNPDLHLQDLEPEDPLPPEAP DLISGVGDPGQGAAWLDRELGGCELAAPGPDRLTCLPEAASASCSYPDLQPGEVLEETPGDSCQLKSPCPLGA SPGLPRSPVSSSA NOVllb, CG55379-01 ISEQ IDNO: 135 J3741~bp
DNA Sequence |ORF Start: ATG at 1 |θRF Stop: end of sequence
ATGGCGCGGGGGGACGCCGGCCGCGGCCGCGGGCTCCTCGCGTTGACCTTCTGCCTGTTGGCCGCGCGCGGGG AGCTGCTGTTGCCCCAGGAGACGACTGTGGAGCTGAGCTGTGGAGTGGGGCCACTGCAAGTGATCCTGGGCCC AGAGCAGGCTGCAGTGCTAAACTGTAGCCTGGGGGCTGCTGCCGCTGGACCCCCCACCAGGGTGACCTGGAGC AAGGATGGGGACACCCTGCTGGAGCACGACCACTTACACCTGCTGGCCAATGGTTCCCTGTGGCTGTCCCAGC CACTAGCACCCAATGGCAGTGACGAGTCAGTCCCTGAGGCTGTGGGGGTCATTGAAGGCAACTATTCGTGCCT AGCCCACGGNCCCCCTCGAGTGCTGGCCAGCCAGACTGCTGTCGTCAAGCTTGCCAGTCTCGCAGACTTCTCT CTGCACCCGGAGTCTCAGACGGTGGAGGAGAACGGGACAGCTCGCTTTGAGTGCCACATTGAAGGGCTGCCAG CTCCCATCATTACTTGGGAGAAGGACCAGGTGACATTGCCTGAGGAGCCTCGGCTCATCGTGCTTCCCAACGG CGTCCTTCAGATCCTGGATGTTCAGGAGAGTGATGCAGGCCCCTACCGCTGCGTGGCCACCAACTCAGCTCGC CAGCACTTCAGCCAGGAGGCCCTACTCAGTGTGGCCCACAGAGGTTCCCTGGCGTCCACCAGGGGGCAGGACG TGGTCATTGTGGCAGCCCCAGAGAACACCACAGTGGTGTCTGGCCAGAGTGTGGTGATGGAATGTGTGGCCTC AGCTGACCCCACCCCTTTTGTGTCCTGGGTCCGAGACGGGAAGCCCATCTCCACAGATGTCATCGTCCTGGGC CGCACCAACCTACTAATTGCCAACGCGCAGCCCTGGCACTCCGGCGTCTATGTCTGCCGCGCCAACAAGCCCC GCACGCGCGACTTCGCCACTGCAGCCGCTGAGCTCCGTGTGCTGCTAGCGGCTCCCGCCATCACTCAGGCGCC CGAGGCGCTGTCGCGGACGCGGGCGAGCACAGCGCGCTTCGTGTGCCGCGCGTCGGGGGAGCCGCGGCCAGCG CTGCGCTGGCTGCACAACGGGGCGCCGCTGCGGCCCAACGGGCGCGTCAAGGTCCAGGGCGGCGGTGGCAGCC TGGTCATCACACAGATCGGCCTGCAGGACGCCGGCTACTACCAGTGCGTGGCTGAGAACAGCGCGGGAATGGC GTGCGCTGCCGCGTCGCTGGCCGTGGTGGTGCGCGAGGGGCTGCCCAGCGCCCCCACGCGGGTCACTGCTACG CCACTGAGCAGCTCCGCTGTGTTGGTGGCCTGGGAGCGGCCCGAGATGCACAGCGAGCAGATCATCGGCTTCT CTCTCCACTACCAGAAGGCACGGGGTATGGACAATGTGGAATACCAGTTTGCAGTGAACAACGACACCACAGA ACTACAGGTTCGGGACCTGGAACCCAACACAGATTATGAGTTCTACGTGGTGGCCTACTCCCAGCTGGGAGCC AGCCGCACCTCCACCCCAGCACTGGTGCACACACTGGATGATGTCCCCAGTGCAGCACCCCAGCTCTCCCTGT CCAGCCCCAACCCTTCGGACATCAGGGTGGCGTGGCTGCCCCTGCCCCCCAGCCTGAGCAATGGGCAGGTGGT GAAGTACAAGATAGAATACGGTTTGGGAAAGGAAGATCAGATTTTCTCTACTGAGGTGCGAGGAAATGAGACA CAGCTTATGCTGAACTCGCTTCAGCCAAACAAGGTGTATCGAGTACGGATTTCGGCTGGTACAGCAGCCGGCT TCGGGGCCCCCTCCCAGTGGATGCATCACAGGACGCCCAGTATGCACAACCAGAGCCATGTCCCTTTTGCCCC TGCAGAGTTGAAGGTGCAGGCAAAGATGGAGTCCCTGGTCGTGTCATGGCAGCCACCCCCTCACCCCACCCAG ATCTCTGGCTACAAACTATATTGGCGGGAGGTGGGGGCTGAGGAGGAGGCCAATGGCGATCGCCTGCCAGGGG GCCGTGGAGACCAGGCTTGGGATGTGGGGCCTGTCCGGCTCAAGAAGAAAGTGAAGCAGTATGAGCTGACCCA GCTAGTCCCTGGCCGGCTGTACGAGGTGAAGCTCGTGGCTTTCAACAAACATGAGGATGGCTATGCAGCAGTG TGGAAGGGCAAGACGGAGAAGGCGCCGGCACCAGACATGCCTATCCAGAGGGGACCACCCCTGCCTCCAGCCC ACGTCCATGCGGAATCAAACAGCTCCACATCCATCTGGCTTCGGTGGAAAAAGCCAGATTTCACCACAGTCAA GATTGTCAACTACACTGTGCGCTTCAGCCCCTGGGGGCTCAGGAATGCCTCCCTGGTCACCTATTACAGTTCT GGAGAAGACATCCTCATTGGCGGCTTGAAGCCATTCACCAAATACGAGTTTGCAGTGCAGTCTCACGGCGTGG ACATGGATGGGCCTTTCGGCTCTGTGGTGGAGCGCTCCACCCTGCCTGACCGTCCCTCCACACCCCCATCCGA CCTGCGACTGAGCCCCCTGACACCGTCCACGGTTCGGCTGCACTGGTGCCCCCCCACAGAGCCCAACGGGGAG ATCGTGGAGTATCTGATCCTGTACAGCAGCAACCACACGCAGCCTGAGCACCAGTGGACCTTGCTCACCACGC AGGGTGAGGGAAACATCTTCAGTGCTGAGGTCCATGGCCTGGAGAGCGACACTCGGTACTTCTTCAAGATGGG GGCGCGCACAGAGGTGGGACCTGGGCCTTTCTCCCGCCTGCAGGATGTGATCACGCTCCAGGAGAAGCTGTCA GACTCGCTGGACATGCACTCAGTCACGGGCATCATCGTGGGTGTCTGCCTGGGCCTCCTCTGCCTCCTGGCCT GCATGTGTGCTGGCCTGCGCCGCAGCCCCCACAGGGAATCCCTCCCAGGCCTGTCCTCCACCGCCACCCCCGG GAATCCCGCGCTGTACTCCAGAGCTCGGCTTGGCCCCCCCAGCCCCCCAGCTGCCCATGAATTGGAGTCCCTT GTGCACCCCCATCCCCAGGACTGGTCCCCGCCACCCTCAGACGTGGAGGACAGGGCTGAAGTGCACAGCCTTA TGGGTGGCGGTGTTTCTGAAGGCCGGAGTCACTCCAAAAGAAAGGTAAGTGCTCAACCAAGCGGGCTGAGCTG GGCTGGTTCCTGGGCAGGCTGTGAGCTGCCCCAGGCAGGCCCCCGGCCGGCTCTGACCCGGGCCCTGCTGCCC CCTGCTGGAACTGGGCAGACGCTGTTGCTGCAGGTTCTCTGCTCTGA
TCAGGGCAATGGGAGGAAGAAGTCACCCCCAGCCTGCAGGAACCAGGTGGAGGCTGAAGTCATTGTCCACTCT GACTTTAGTGCATCTAACGGGAACCCTGACCTCCATCTCCAAGACCTGGAGCCTGAGGACCCCCTGCCTCCAG AGGCTCCTGATCTCATCTCGGGTGTTGGGGATCCAGGGCAGGGGGCAGCCTGGCTGGACAGGGAGTTGGGAGG GTGTGAGCTGGCAGCCCCCGGGCCAGACAGACTTACCTGCTTGCCAGAGGCAGCCAGTGCTTCCTGCTCCTAC CCGGACCTCCAGCCAGGCGAGGTGCTAGAGGAGACCCCTGGAGATAGCTGCCAGCTCAAATCCCCCTGCCCTC TAGGAGCCAGCCCAGGCCTGCCCAGATCCCCGGTCTCCTCCTCT
Figure imgf000200_0001
CNOVllc, 258065951 SEQ ID NO: 137 1609 bp
DNA Sequence ORF Start: at 1 ORF Stop: at 1609
GGTACCGCGTCGCTGGCCGTGGTGGTGCGCGAGGGGCTGCCCAGCGCCCCCACGCGGGTCACTGCTACGCCAC TGAGCAGCTCCGCTGTGTTGGTGGCCTGGGAGCGGCCCGAGATGCACAGCGAGCAGATCATCGGCTTCTCTCT CCACTACCAGAAGGCACGGGGCATGGACAATGTGGAATACCAGTTTGCAGTGAACAACGACACCACAGAACTA CAGGTTCGGGACCTGGAACCCAACACAGATTATGAGTTCTACGTGGTGGCCTACTCCCAGCTGGGAGCCAGCC GCACCTCCACCCCAGCACTGGTGCACACACTGGATGATGTCCCCAGTGCAGCACCCCAGCTCTCCCTGTCCAG CCCCAACCCTTCGGACATCAGGGTGGCGTGGCTGCCCCTGCCCCCCAGCCTGAGCAATGGGCAGGTGGTGAAG TACAAGATAGAATACGGTTTGGGAAAGGAAGATCAGATTTTCTCTACTGAGGTGCGAGGAAATGAGACACAGC TTATGCTGAACTCGCTTCAGCCAAACAAGGTGTATCGAGTACGGATTTCGGCTGGTACAGCAGCCGGCTTCGG GGCCCCCTCCCAGTGGATGCATCACAGGACGCCCAGTATGCACAACCAGAGCCATGTCCCTTTTGCCCCTGCA GAGTTGAAGGTGCAGGCAAAGATGGAGTCCCTGGTCGTGTCATGGCAGCCACCCCCTCACCCCACCCAGATCT CTGGCTACAAACTATATTGGCGGGAGGTGGGGGCTGAGGAGGAGGCCAATGGCGATCGCCTGCCAGGGGGCCG TGGAGACCAGGCTTGGGATGTGGGGCCTGTCCGGCTCAAGAAGAAAGTGAAGCAGTATGAGCTGACCCAGCTA GTCCCTGGCCGGCTGTACGAGGTGAAGCTCGTGGCTTTCAACAAACATGAGGATGGCTATGCAGCAGTGTGGA AGGGCAAGACGGAGAAGGCGCCGGCACCAGACATGCCTATCCAGAGGGGACCACCCCTGCCTCCAGCCCACGT CCATGCGGAATCAAACAGCTCCACATCCATCTGGCTTCGGTGGAAAAAGCCAGATTTCACCACAGTCAAGATT GTCAACTACACTGTGCGCTTCAGCCCCTGGGGGCTCAGGAATGCCTCCCTGGTCACCTATTACACCAGTTCTG GAGAAGACATCCTCATTGGCGGCTTGAAGCCATTCACCAAATACGAGTTTGCAGTGCAGTCTCACGGCGTGGA CATGGATGGGCCTTTCGGCTCTGTGGTGGAGCGCTCCACCCTGCCTGACCGGCCCTCCACACCCCCATCCGAC CTGCGACTGAGCCCCCTGACACCGTCCACGGTTCGGCTGCACTGGTGCCCCCCCACAGAGCCCAACGGGGAGA TCGTGGAGTATCTGATCCTGTACAGCAGCAACCACACGCAGCCTGAGCACCAGTGGACCTTGCTCACCACGCA GGGAAACATCTTCAGTGCTGAGGTCCATGGCCTGGAGAGCGACACTCGGTACTTCTTCAAGATGGGGGCGCGC ACAGAGGTGGGACCTGGGCCTTTCTCCCGCCTGCAGGATGTGATCACGCTCCAGGAGAAGCTGTCAGACTCGG TCG NOVl lc, 258065951 SEQ ID NO: 138 536 aa MW at 59532.7kD Protein Sequence
GTASLAVVVREGLPSAPTRVTATPLSSSAVLVAWERPEMHSEQIIGFSLHYQKARGMDNVEYQFAVNNDTTEL QVRDLEPNTDYEFYWAYSQLGASRTSTPALVHTLDDVPSAAPQLSLSSPNPSDIRVAWLPLPPSLSNGQWK YKIEYGLGKEDQIFSTEVRGNETQLMLNSLQPNKVYRVRISAGTAAGFGAPSQWMHHRTPS HNQSHVPFAPA ELKVQAKMESLWSWQPPPHPTQISGYKLYWREVGAEEEANGDRLPGGRGDQAWDVGPVRLKKKVKQYELTQL VPGRLYEVKLVAFNKHEDGYAAVWKGKTEKAPAPDMPIQRGPPLPPAHVHAESNSSTSIWLRWKKPDFTTVKI VNYTVRFSPWGLRNASLVTYYTSSGEDILIGGLKPFTKYEFAVQSHGVDMDGPFGSWERSTLPDRPSTPPSD LRLSPLTPSTVRLHWCPPTEPNGEIVEYLILYSSNHTQPEHQWTLLTTQGNIFSAEVHGLESDTRYFFKMGAR TEVGPGPFSRLQDVITLQEKLSDSV
Figure imgf000201_0001
NOVl Id, 209886264 SEQ ID NO: 140 537 aa MW at 59607.7kD Protein Sequence
GTASLAWVREGLPSAPTRVTATPLSSSAVLVAWERPEMPSEQIIGFSLHYQKARGMDNVEYQFAVNNDTTEL QVRDLEPNTDYEFYWAYSQLGASRTSTPALVHTLDDVPSAAPQLSLSSPNPSDIRVAWLPLPPSLSNGQWK YKIEYGLGKEDQIFSTEVRGNETQLMLNSLQPNKVYRVRISAGTAAGFGAPSQW HHRTPSMHNQSHVPFAPA ELKVQAKMESLVVSWQPPPHPTQISGYKLYWREVGAEEEANGDRLPGGRGDQAWDVGPVRLKKKVKQYELTQL VPGRLYEVKLVAFNKHEDGYAAVWKGKTEKAPAPDMPIQRGPPLPPAHVHAESNSSTSIWLRWKKPDFTTVKI VNYTVRFSPWGLRNASLVTYYTSSGEDILIGGLKPFTKYEFAVQSHGVDMDGPFGSWERSTLPDRPSTPPSD LRLSPLTPSTVRLHWCPPTEPNGEIVEYLILYSSNHTQPEHQWTLLTTQGNIFSAEVHGLESDTRYFFKMGAR TEVGPGPFSRLQDVITLQEKLSDSVD
Figure imgf000202_0001
NOVl le, 209886345 SEQ ID NO: 142 557 aa MW at 61878.3kD Protein Sequence _
GTASLAVWREGLPSAPTRVTATPLSSSAVLVAWERPEMPSEQIIGFSLHYQKARGMDNVEYQFAVNNDTTEL QVRDLEPNTDYEFYWAYSQLGASRTSTPALVHTLDDVPSAAPQLSLSSPNPSDIRVAWLPLPPSLSNGQWK YKIEYGLGKEDQIFSTEVRGNETQLMLNSLQPNKVYRVRISAGTAAGFGAPSQWMHHRTPSMHNQSHVPFAPA ELKVQARMESLWSWQPPPHPTQISGYKLYWREVGAEEEANGDRLPGGRGDQAWDVGPVRLKKKVKQYELTQL VPGRLYEVKLVAFNKHEDGYAAVWKGKTEKAPAPDMPIQRGPPLPPAHVHAESNSSTSIWLRWKKPDFTTVKI VNYTVRFSPWGLRNASLVTYYTSSGEDILIGGLKPFTKYEFAVQSHGVDMDGPFGSWERSTLPDRPSTPPSD LRLSPLTPSTVRLHWCPPTEPNGEIVEYLILYSSNHTQPEHQWTLLTTQGNIFSAEVHGLESDTRYFFK GAR TEVGPGPFSRLQDVITLQEKLSDSVDSFSWSVITAPRAPPRPATRY
Figure imgf000202_0002
CCATGCGGAATCAAACAGCTCCACATCCATCTGGCTTCGGTGGAAAAAGCCAGATTTCACCACAGTCAAGATT GTCAACTACACTGTGCGCTTCAGCCCCTGGGGGCTCAGGAATGCCTCCCTGGTCACCTATTACACCAGTTCTG GAGAAGACATCCTCATTGGCGGCTTGAAGCCATTCACCAAATACGAGTTTGCAGTGCAGTCTCACGGCGTGGA CATGGATGGGCCTTTCGGCTCTGTGGTGGAGCGCTCCACCCTGCCTGACCGGCCCTCCACACCCCCATCCGAC CTGCGACTGAGCCCCCTGACGCCGTCCACGGTTCGGCTGCACTGGTGCCCCCCCACAGAGCCCAACGGGGAGA TCGTGGAGTATCTGATCCTGTACAGCAGCAACCACACGCAGCCTGAGCACCAGTGGACCTTGCTCACCACGCA GGGAAACATCTTCAGTGCTGAGGTCCATGGCCTGGAGAGCGACACTCGGTACTTCTTCAAGATGGGGGCGCGC ACAGAGGTGGGACCTGGGCCTTTCTCCCGCCTGCAGGATGTGATCACGCTCCAGGAGAAGCTGTCAGACTCGG TCGAC
NOVl If, 209886357 SEQ ID NO: 144 537 aa MW at 59607.7kD Protein Sequence
GTASLAVWREGLPSAPTRVTATPLSSSAVLVAWERPEMPSEQIIGFSLHYQKARGMDNVEYQFAVNNDTTEL QVRDLEPNTDYEFYWAYSQLGASRTSTPALVHTLDDVPSAAPQLSLSSPNPSDIRVAWLPLPPSLSNGQWK ΫKIEYGLGKEDQIFSTEVRGNETQLMLNSLQPNKVYRVRISAGTAAGFGAPSQWMHHRTPSMHNQSHVPFAPA ELKVQAKMESLWSWQPPPHPTQISGYKLYWREVGAEEEANGDRLPGGRGDQAWDVGPVRLKKKVKQYELTQL VPGRLYEVKLVAFNKHEDGYAAVWKGKTEKAPAPDMPIQRGPPLPPAHVHAESNSSTSIWLRWKKPDFTTVKI VNYTVRFSPWGLRNASLVTYYTSSGEDILIGGLKPFTKYEFAVQSHGVDMDGPFGSWERSTLPDRPSTPPSD LRLSPLTPSTVRLHWCPPTEPNGEIVEYLILYSSNHTQPEHQWTLLTTQGNIFSAEVHGLESDTRYFFKMGAR TEVGPGPFSRLQDVITLQEKLSDSVD
Figure imgf000203_0001
Figure imgf000204_0001
NOVllh, CG55379-03 SEQ ID NO: 147 jl672 bp
DNA Sequence ORF Start: at 7 |ORF Stop: at 1606
GGTACCGCGTCGCTGGCCGTGGTGGTGCGCGAGGGGCTGCCCAGCGCCCCCACGCGGGTCACTGCTACGCCAC
TGAGCAGCTCCGCTGTGTTGGTGGCCTGGGAGCGGCCCGAGATGCCCAGCGAGCAGATCATCGGCTTCTCTCT CCACTACCAGAAGGCACGGGGCATGGACAATGTGGAATACCAGTTTGCAGTGAACAACGACACCACAGAACTA CAGGTTCGGGACCTGGAACCCAACACAGATTATGAGTTCTACGTGGTGGCCTACTCCCAGCTGGGAGCCAGCC GCACCTCCACCCCAGCACTGGTGCACACACTGGATGATGTCCCCAGTGCAGCACCCCAGCTCTCCCTGTCCAG CCCCAACCCTTCGGACATCAGGGTGGCGTGGCTGCCCCTGCCCCCCAGCCTGAGCAATGGGCAGGTGGTGAAG TACAAGATAGAATACGGTTTGGGAAAGGAAGATCAGATTTTCTCTACTGAGGTGCGAGGAAATGAGACACAGC TTATGTTGAACTCGCTTCAGCCAAACAAGGTGTATCGAGTACGGATTTCGGCTGGTACAGCAGCCGGCTTCGG GGCCCCCTCCCAGTGGATGCATCACAGGACGCCCAGTATGCACAACCAGAGCCATGTCCCTTTTGCCCCTGCA GAGTTGAAGGTGCAGGCAAGGATGGAGTCCCTGGTCGTGTCATGGCAGCCACCCCCTCACCCCACCCAGATCT CTGGCTACAAACTATATTGGCGGGAGGTGGGGGCTGAGGAGGAGGCCAATGGCGATCGCCTGCCAGGGGGCCG TGGAGACCAGGCTTGGGATGTGGGGCCTGTCCGGCTCAAGAAGAAAGTGAAGCAGTATGAGCTGACCCAGCTA GTCCCTGGCCGGCTGTACGAGGTGAAGCTCGTGGCTTTCAACAAACATGAGGATGGCTATGCAGCAGTGTGGA AGGGCAAGACGGAGAAGGCGCCGGCACCAGACATGCCTATCCAGAGGGGACCACCCCTGCCTCCAGCCCACGT CCATGCGGAATCAAACAGCTCCACATCCATCTGGCTTCGGTGGAAAAAGCCAGATTTCACCACAGTCAAGATT GTCAACTACACTGTGCGCTTCAGCCCCTGGGGGCTCAGGAATGCCTCCCTGGTCACCTATTACACCAGTTCTG GAGAAGACATCCTCATTGGCGGCTTGAAGCCATTCACCAAATACGAGTTTGCAGTGCAGTCTCACGGCGTGGA CATGGATGGGCCTTTCGGCTCTGTGGTGGAGCGCTCCACCCTGCCTGACCGGCCCTCCACACCCCCATCCGAC CTGCGACTGAGCCCCCTGACGCCGTCCACGGTTCGGCTGCACTGGTGCCCCCCCACAGAGCCCAACGGGGAGA TCGTGGAGTATCTGATCCTGTACAGCAGCAACCACACGCAGCCTGAGCACCAGTGGACCTTGCTCACCACGCA GGGAAACATCTTCAGTGCTGAGGTCCATGGCCTGGAGAGCGACACTCGGTACTTCTTCAAGATGGGGGCGCGC ACAGAGGTGGGACCTGGGCCTTTCTCCCGCCTGCAGGATGTGATCACGCTCCAGGAGAAGCTGTCAGACTCGG TCGACAGCTTCTCCTGGAGCGTGATCACAGCCCCTCGCGCACCACCACGGCCAGCGACGCGGTACC
NOVllh, CG55379-03 SEQ ID NO: 148 533 aa |MW at 59263.4kD Protein Sequence
ASLAVWREGLPSAPTRVTATPLSSSAVLVAWERPEMPSEQIIGFSLHYQKARGMDNVEYQFAVNNDTTELQV RDLEPNTDYEFYWAYSQLGASRTSTPALVHTLDDVPSAAPQLSLSSPNPSDIRVAWLPLPPSLSNGQWKYK IEYGLGKEDQIFSTEVRGNETQLMLNSLQPNKVΎRVRISAGTAAGFGAPSQWMHHRTPS HNQSHVPFAPAEL KVQARMESLWSWQPPPHPTQISGYKLYWREVGAEEEANGDRLPGGRGDQAWDVGPVRLKKKVKQYELTQLVP GRLYEVKLVAFNKHEDGYAA KGKTEKAPAPDMPIQRGPPLPPAHVHAESNSSTSIWLRWKKPDFTTVKIVN YTVRFSPWGLRNASLVTYYTSSGEDILIGGLKPFTKYEFAVQSHGVDMDGPFGSWERSTLPDRPSTPPSDLR LSPLTPSTVRLHWCPPTEPNGEIVEYLILYSSNHTQPEHQWTLLTTQGNIFSAEVHGLESDTRYFFKMGARTE VGPGPFSRLQDVITLQEKLSDS A ClustalW comparison of the above protein sequences yields the following sequence alignment shown in Table 1 IB.
Table 11B. Comparison of the NOVll protein sequences.
NOVlla MARGDAGRGRGLLALTFCLLAARGELLLPQETTVELSCGVGPLQVILGPEQAAVLNCSLG
NOVllb MARGDAGRGRGLLALTFCLLAARGELLLPQETTVELSCGVGPLQVILGPEQAAVLNCSLG
NOVllc
NOVlld
NOVlle
NOVlIf
NOVllg
NOVllh
NOVlla AAAAGPPTRVTWSKDGDTLLEHDHLHLLPNGSLWLSQPLAPNGSDESVPEAVGVIEGNYS
NOVllb AAAAGPPTRVTWSKDGDTLLEHDHLHLLANGSLWLSQPLAPNGSDESVPEAVGVIEGNYS
NOVllc
NOVlld
NOVlle
NOVlIf
NOVllg
NOVllh
NOVlla CLAHGPLGVLASQTAWKLASLADFSLHPESQTVEENGTARFECHIEGLPAPIITWEKDQ
NOVllb CLAHGPPRVLASQTAWKLASLADFSLHPESQTVEENGTARFECHIEGLPAPIITWEKDQ
NOVllc
NOVlld
NOVlle
NOVlIf
NOVllg
NOVllh
NOVlla VTLPEEPRLIVLPNGVLQILDVQESDAGPYRCVATNSARQHFSQEALLSVAHRGSLASTR
NOVllb VTLPEEPRLIVLPNGVLQILDVQESDAGPYRCVATNSARQHFSQEALLSVAHRGSLASTR
NOVllc
NOVlld
NOVlle
NOVlIf
NOVllg
NOVllh
NOVlla GQDWIVAAPENTTWSGQSWMECVASADPTPFVSWVRDGKPISTDVIVLGRTNLLIAN
NOVllb GQDWIVAAPENTTWSGQSWMECVASADPTPFVSWVRDGKPISTDVIVLGRTNLLIAN
NOVllc
NOVlld
NOVlle
NOVllf
NOVllg
NOVllh
NOVlla AQPWHSGVYVCRANKPRTRDFATAAAELRVLLAAPAITQAPEALSRTRASTARFVCRASG
NOVllb AQPWHSGVYVCRANKPRTRDFATAAAELRVLLAAPAITQAPEALSRTRASTARFVCRASG
NOVllc
NOVlld
NOVlle NOVllf
NOVllg
NOVllh
NOVlla EPRPALRWLHNGAPLRPNGRVKVQGGGGSLVITQIGLQDAGYYQCVAENSAGMACAAASL
NOVllb EPRPALRWLHNGAPLRPNGRVKVQGGGGSLVITQIGLQDAGYYQCVAENSAG ACAAASL
NOVllc GTASL
NOVlld GTASL
NOVlle GTASL
NOVllf GTASL
NOVllg ASL
NOVllh ASL
NOVlla AVWREGLPSAPTRVTATPLSSSAVLVAWERPEMHSEQIIGFSLHYQKARGMDNVΞYQFA
NOVllb AVWREGLPSAPTRVTATPLSSSAVLVAWERPEMHSEQIIGFSLHYQKARGMDNVEYQFA
NOVllc AVWREGLPSAPTRVTATPLSSSAVLVAWERPEMHSEQIIGFSLHYQKARGMDNVEYQFA
NOVlld AVWREGLPSAPTRVTATPLSSSAVLVAWERPEMPSEQIIGFSLHYQKARGMDNVEYQFA
NOVlle AVWREGLPSAPTRVTATPLSSSAVLVAWERPEMPSEQIIGFSLHYQKARGMDN/EYQFA
NOVllf AVWREGLPSAPTRVTATPLSSSAVLVAWERPEMPSEQIIGFSLHYQKARGMDNVEYQFA
NOVllg AVWREGLPSAPTRVTATPLSSSAVLVAWERPEMPSEQIIGFSLHYQKARGMDNVEYQFA
NOVllh AVWREGLPSAPTRVTATPLSSSAVLVAWERPEMPSEQIIGFSLHYQKARGMDNVEYQFA
NOVlla VNNDTTELQVRDLEPNTDYEFYWAYSQLGASRTSTPALVHTLDDVPSAAPQLSLSSPNP
NOVllb VNNDTTELQVRDLEPNTDYEFYWAYSQLGASRTSTPALVHTLDDVPSAAPQLSLSSPNP
NOVllc VNNDTTELQVRDLEPNTDYEFYWAYSQLGASRTSTPALVHTLDDVPSAAPQLSLSSPNP
NOVlId VNNDTTELQVRDLEPNTDYEFYWAYSQLGASRTSTPALVHTLDDVPSAAPQLSLSSPNP
NOVlle VNNDTTELQVRDLEPNTDYEFYWAYSQLGASRTSTPALVHTLDDVPSAAPQLSLSSPNP
NOVlIf VNNDTTELQVRDLEPNTDYEFYWAYSQLGASRTSTPALVHTLDDVPSAAPQLSLSSPNP
NOVllg VNNDTTELQVRDLEPNTDYEFYWAYSQLGASRTSTPALVHTLDDVPSAAPQLSLSSPNP
NOVllh VNNDTTELQVRDLEPNTDYEFYWAYSQLGASRTSTPALVHTLDDVPSAAPQLSLSSPNP
NOVlla SDIRVAWLPLPPSLSNGQWKYKIEYGLGKEGEWGDQIFSTEVRGNETQLMLNSLQPNKV
NOVllb SDIRVAWLPLPPSLSNGQWKYKIEYGLGKE DQIFSTEVRGNETQLMLNSLQPNKV
NOVllc SDIRVAWLPLPPSLSNGQWKYKIEYGLGKE DQIFSTEVRGNETQLMLNSLQPNKV
NOVlld SDIRVAWLPLPPSLSNGQWKYKIEYGLGKE DQIFSTEVRGNETQLMLNSLQPNKV
NOVlle SDIRVAWLPLPPSLSNGQWKYKIEYGLGKE DQIFSTEVRGNETQLMLNSLQPNKV
NOVl1f SDIRVAWLPLPPSLSNGQWKYKIEYGLGKE DQIFSTEVRGNETQLMLNSLQPNKV
NOVllg SDIRVAWLPLPPSLSNGQWKYKIEYGLGKE DQIFSTEVRGNETQLMLNSLQPNKV
NOVllh SDIRVAWLPLPPSLSNGQWKYKIEYGLGKE DQIFSTEVRGNETQLMLNSLQPNKV
NOVlla YRVRISAGTAAGFGAPSQWMHHRTPSMHNQSHVPFAPAELKVQAKMESLWSWQPPPHPT
NOVllb YRVRISAGTAAGFGAPSQWMHHRTPSMHNQSHVPFAPAELKVQAKMESLWSWQPPPHPT
NOVllc YRVRISAGTAAGFGAPSQWMHHRTPSMHNQSHVPFAPAELKVQAKMESLWSWQPPPHPT
NOVlld YRVRISAGTAAGFGAPSQWMHHRTPSMHNQSHVPFAPAELKVQAKMESLWSWQPPPHPT
NOVlle YRVRISAGTAAGFGAPSQWMHHRTPSMHNQSHVPFAPAELKVQARMESLWSWQPPPHPT
NOVlIf YRVRISAGTAAGFGAPSQWMHHRTPSMHNQSHVPFAPAELKVQAKMESLWSWQPPPHPT
NOVllg YRVRISAGTAAGFGAPSQWMHHRTPSMHNQSHVPFAPAELKVQAKMESLWSWQPPPHPT
NOVllh YRVRISAGTAAGFGAPSQWMHHRTPSMHNQSHVPFAPAELKVQARMESLWSWQPPPHPT
NOVlla QISGYKLYWREVGAEEEANGDRLPGGRGDQAWDVGPVRLKKKVKQYELTQLVPGRLYEVK
NOVllb QISGYKLYWREVGAEEEANGDRLPGGRGDQAWDVGPVRLKKKVKQYELTQLVPGRLYEVK
NOVl1c QISGYKLYWREVGAEEEANGDRLPGGRGDQAWDVGPVRLKKKVKQYELTQLVPGRLYEVK
NOVlId QISGYKLYWREVGAEEEANGDRLPGGRGDQAWDVGPVRLKKKVKQYELTQLVPGRLYEVK
NOVl1e QISGYKLYWREVGAEEEANGDRLPGGRGDQAWDVGPVRLKKKVKQYELTQLVPGRLYEVK
NOVllf QISGYKLYWREVGAEEEANGDRLPGGRGDQAWDVGPVRLKKKVKQYELTQLVPGRLYEVK
NOVllg QISGYKLYWREVGAEEEANGDRLPGGRGDQAWDVGPVRLKKKVKQYELTQLVPGRLYEVK
NOVllh QISGYKLYWREVGAEEEANGDRLPGGRGDQAWDVGPVRLKKKVKQYELTQLVPGRLYEVK
NOVlla LVAFNKHEDGYAAVWKGKTEKAPAPDMPIQRGPPLPPAHVHAESNSSTSIWLRWKKPDFT NOVllb LVAFNKHEDGYAAVWKGKTΞKAPAPDMPIQRGPPLPPAHVHAESNSSTSIWLRWKKPDFT
NOVllc LVAFNKHEDGYAAVWKGKTEKAPAPDMPIQRGPPLPPAHVHAESNSSTSIWLRWKKPDFT
NOVlld LVAFNKHEDGYAAVWKGKTEKAPAPDMPIQRGPPLPPAHVHAESNSSTSIWLRWKKPDFT
NOVlle LVAFNKHEDGYAAVWKGKTEKAPAPDMPIQRGPPLPPAHVHAESNSSTSIWLRWKKPDFT
NOVllf LVAFNKHEDGYAAVWKGKTEKAPAPDMPIQRGPPLPPAHVHAESNSSTSIWLRWKKPDFT
NOVllg LVAFNKHEDGYAAVWKGKTEKAPAPDMPIQRGPPLPPAHVHAESNSSTSIWLRWKKPDFT
NOVllh LVAFNKHEDGYAAVWKGKTEKAPAPDMPIQRGPPLPPAHVHAESNSSTSIWLRWKKPDFT
NOVlla TVKIVNYTVRFSPWGLRNASLVTYYTSSGEDILIGGLKPFTKYEFAVQSHGVDMDGPFGS
NOVllb TVKIVNYTVRFSPWGLRNASLVTYYSS-GEDILIGGLKPFTKYEFAVQSHGVDMDGPFGS
NOVllc TVKIVNYTVRFSPWGLRNASLVTYYTSSGEDILIGGLKPFTKYEFAVQSHGVDMDGPFGS
NOVlId TVKIVNYTVRFSPWGLRNASLVTYYTSSGEDILIGGLKPFTKYEFAVQSHGVDMDGPFGS
NOVlle TVKIVNYTVRFSPWGLRNASLVTYYTSSGEDILIGGLKPFTKYEFAVQSHGVDMDGPFGS
NOVllf TVKIVNYTVRFSPWGLRNASLVTYYTSSGEDILIGGLKPFTKYEFAVQSHGVDMDGPFGS
NOVllg TVKIVNYTVRFSPWGLRNASLVTYYTSSGEDILIGGLKPFTKYEFAVQSHGVDMDGPFGS
NOVllh TVKIVNYTVRFSPWGLRNASLVTYYTSSGEDILIGGLKPFTKYEFAVQSHGVDMDGPFGS
NOVlla WERSTLPDRPSTPPSDLRLSPLTPSTVRLHWCPPTEPNGEIVEYLILYSSNHTQPEHQW
NOVllb WERSTLPDRPSTPPSDLRLSPLTPSTVRLHWCPPTEPNGEIVEYLILYSSNHTQPEHQW
NOVllc WERSTLPDRPSTPPSDLRLSPLTPSTVRLHWCPPTEPNGEIVEYLILYSSNHTQPEHQW
NOVlld WERSTLPDRPSTPPSDLRLSPLTPSTVRLHWCPPTEPNGEIVEYLILYSSNHTQPEHQW
NOVlle WERSTLPDRPSTPPSDLRLSPLTPSTVRLHWCPPTEPNGEIVEYLILYSSNHTQPEHQW
NOVllf WERSTLPDRPSTPPSDLRLSPLTPSTVRLHWCPPTEPNGEIVEYLILYSSNHTQPEHQW
NOVllg WERSTLPDRPSTPPSDLRLSPLTPSTVRLHWCPPTEPNGEIVEYLILYSSNHTQPEHQW
NOVllh WERSTLPDRPSTPPSDLRLSPLTPSTVRLHWCPPTEPNGEIVEYLILYSSNHTQPEHQW
NOVlla TLLTTQG--NIFSAEVHGLESDTRYFFKMGARTEVGPGPFSRLQDVITLQEKLSDSLDMH
NOVllb TLLTTQGEGNIFSAEVHGLESDTRYFFKMGARTEVGPGPFSRLQDVITLQEKLSDSLDMH
NOVllc TLLTTQG--NIFSAEVHGLESDTRYFFKMGARTEVGPGPFSRLQDVITLQEKLSDSV
NOVlld TLLTTQG- -NIFSAEVHGLESDTRYFFKMGARTEVGPGPFSRLQDVITLQEKLSDSVD--
NOVlle TLLTTQG--NIFSAEVHGLESDTRYFFKMGARTEVGPGPFSRLQDVITLQEKLSDSVDSF
NOVllf TLLTTQG--NIFSAEVHGLESDTRYFFKMGARTEVGPGPFSRLQDVITLQEKLSDSVD--
NOVllg TLLTTQG- -NIFSAEVHGLESDTRYFFKMGARTEVGPGPFSRLQDVITLQEKLSDS
NOVllh TLLTTQG--NIFSAEVHGLESDTRYFFKMGARTEVGPGPFSRLQDVITLQEKLSDS
NOVlla SVTGIIVGVCLGLLCLLACMCAGLRRSPHRESLPGLSSTATPGNPALYSRARLGPPSPPA
NOVllb SVTGIIVGVCLGLLCLLACMCAGLRRSPHRESLPGLSSTATPGNPALYSRARLGPPSPPA
NOVllc
NOVlld
NOVlle SWSVITAPRAPPRPATRY
NOVllf
NOVllg
NOVllh
NOVlla AHELESLVHPHPQDWSPPPSDVEDRAEVHSLMGGGVSEGRSHSKRKISWAQPSGLSWAGS
NOVllb AHELESLVHPHPQDWSPPPSDVEDRAEVHSLMGGGVSEGRSHSKRK-VSAQPSGLSWAGS
NOVllc
NOVlld
NOVlle
NOVllf
NOVllg
NOVllh
NOVlla WAGCELPQAGPRPALTRALLPPAGTGQTLLLQALVYDAIKGNGRKKSPPACRNQVEAEVI
NOVllb WAGCELPQAGPRPALTRALLPPAGTGQTLLLQVLCSD--QGNGRKKSPPACRNQVEAEVI
NOVllc
NOVlld
NOVlle
NOVllf NOVllg
NOVllh
NOVlla VHSDFSASNGNPDLHLQDLEPEDPLPPEAPDLISGVGDPGQGAAWLDRELGGCELAAPGP
NOVllb VHSDFSASNGNPDLHLQDLEPEDPLPPEAPDLISGVGDPGQGAAWLDRELGGCELAAPGP
NOVllc
NOVlld
NOVlle
NOVllf
NOVllg
NOVllh
NOVlla DRLTCLPEAASASCSYPDLQPGEVLEETPGDSCQLKSPCPLGASPGLPRSPVSSSA
NOVllb DRLTCLPEAASASCSYPDLQPGEVLEETPGDSCQLKSPCPLGASPGLPRSPVSSS-
NOVllc
NOVlld
NOVlle
NOVllf
NOVllg
NOVllh
NOVlla (SEQ ID NO 134) NOVllb (SEQ ID NO 136) NOVllc (SEQ ID NO 138) NOVlld (SEQ ID NO 140) NOVlle (SEQ ID NO 142) NOVllf (SEQ ID NO 144) NOVllg (SEQ ID NO 146) NOVllh (SEQ ID NO 148)
Further analysis of the NOVl la protein yielded the following properties shown in Table llC.
Table 11C. Protein Sequence Properties NOVlla
SignalP analysis: Cleavage site between residues 25 and 26
PSORT π analysis:
PSG: a new signal peptide prediction method
N-region: length 10; pos.chg 3; neg.chg 1 H-region: length 12; peak value 10.30 PSG score: 5.90
GvH: von Heijne's method for signal seq. recognition GvH score (threshold: -2.1): 1.11 possible cleavage site: between 24 and 25
»> Seems to have a cleavable signal peptide (1 to 24)
ALOM: Klein et al • s method for TM region allocation Init position for calculation: 25
Tentative number of TMS(s) for the threshold 0.5: Number of TMS(s) for threshold 0.5: 1 INTEGRAL Likelihood =-11.89 Transmembrane 963 979 PERIPHERAL Likelihood = 0.90 (at 321) ALOM score: -11.89 (number of TMSs: 1) MTOP: Prediction of membrane topology (Hartmann et al . ) Center position for calculation: 12 Charge difference: -5.0 C(-2.0) - N( 3.0) N >= C: N-terminal side will be inside
>>> membrane topology: type la (cytoplasmic tail 980 to 1254)
MITDISC: discrimination of mitochondrial targeting seq R content: 4 Hyd Moment (75) : 11.83 Hyd Moment (95): 5.53 G content: 5 D/E content: 2 S/T content: 1 Score : -4.74
Gavel : prediction of cleavage sites or mitochondrial preseq R-2 motif at 33 ARG| EL
NUCDISC: discrimination of nuclear localization signals pat4 : none pat7: PVRLKKK (4) at 696 bipartite : none content of basic residues: 8.2% NLS Score: -0.13
KDEL: ER retention motif in the C-terminus : none
ER Membrane Retention Signals:
XXRR-like motif in the N-terminus -. ARGD none
SKL: peroxisomal targeting signal in the C-terminus : none
PTS2 : 2nd peroxisomal targeting signal : none
VAC: possible vacuolar targeting motif: none
RNA-binding motif: none
Actinin-type actin-binding motif: type 1 : none type 2 : none
NMYR: N-myristoylation pattern : none
Prenylation motif: none memYQRL: transport motif from cell surface to Golgi: none
Tyrosines in the tail : too long tail
Dileucine motif in the tail : found LL at 1097 LL at 1107 checking 63 PROSITE DNA binding motifs: none checking 71 PROSITE ribosomal protein motifs : none checking 33 PROSITE prokaryotic DNA binding motifs: none
NNCN: Reinhardt's method for Cytoplasmic/Nuclear discrimination Prediction: cytoplasmic Reliability: 55.5
COIL: Lupas's algorithm to detect coiled-coil regions total : 0 residues Final Results (k = 9/23) :
55.6 % endoplasmic reticulum 22.2 % Golgi 11.1 % plasma membrane 11.1 % extracellular, including cell wall
» prediction for CG55379-04 is end (k=9)
A search of the NOVl la protein against the Geneseq database, a proprietary database that contains sequences published in patents and patent publication, yielded several homologous proteins shown in Table 1 ID.
Figure imgf000210_0001
In a BLAST search of public sequence databases, the NOVl la protein was found to have homology to the proteins shown in the BLASTP data in Table 1 IE.
Figure imgf000211_0001
PFam analysis predicts that the NOVl la protein contains the domains shown in the Table 11F.
Figure imgf000211_0002
Example 12.
The NOVl 2 clone was analyzed, and the nucleotide and encoded polypeptide sequences are shown in Table 12 A.
Table 12A. NOV12 Sequence Analysis
NOV12a, CG55688-01 |SEQ IDNO: 149 18_87bp_ DNA Sequence _ {^"Start: ATCaTδT Joi stop: TAA at 1224
IGCGCACGGCCTGTCCGCTGCACACCAGCTTGTTGGCGTCTTCGTCGCCGCGCTCGCCCCGGGCTACTCCTGCG
CGCCACAATGAGCTCCCGCATCGCCAGGGCGCTCGCCTTAGTCGTCACCCTTCTCCACTTGACCAGGCTGGCG
CTCTCCACCTGCCCCGCTGCCTGCCACTGCCCCCTGGAGGCGCCCAAGTGCGCGCCGGGAGTCGGGCTGGTCC GGGACGGCTGCGGCTGCTGTAAGGTCTGCGCCAAGCAGCTCAACGAGGACTGCAGCAAAACGCAGCCCTGCGA CCACACCAAGGGGCTGGAATGCAACTTCGGCGCCAGCTCCACCGCTCTGAAGGGGATCTGCAGAGCTCAGTCA GAGGGCAGACCCTGTGAATATAACTCCAGAATCTACCAAAACGGGGAAAGTTTCCAGCCCAACTGTAAACATC AGTGCACATGTATTGATGGCGCCGTGGGCTGCATTCCTCTGTGTCCCCAAGAACTATCTCTCCCCAACTTGGG CTGTCCCAACCCTCGGCTGGTCAAAGTTACCGGGCAGTGCTGCGAGGAGTGGGTCTGTGACGAGGATAGTATC AAGGACCCCATGGAGGACCAGGACGGCCTCCTTGGTAAGGAGCTGGGATTCGATGCCTCCGAGGTGGAGTTGA CGAGAAACAATGAATTGATTGCAGTTGGAAAAGGCAGCTCACTGAAGCGGATCCCTGTTTTTGGAATGGAGCC TCGCATCCGATACAACCCTTTACAAGGCCAGAAATGTATTGTTCAAACAACTTCATGGTCCCAGTGCTCAAAG ACCTGTGGAACTGGTATCTCCACACGAGTTACCAATGACAACCCTGAGTGCCGCCTTGTGAAAGAAACCCGGA TTTGTGAGGTGCGGCCTTGTGGACAGCCAGTGTACAGCAGCCTGAAAAAGGGCAAGAAATGCAGCAAGACCAA GAAATCCCCCGAACCAGTCAGGTTTACTTACGCTGGATGTTTGAGTGTGAAGAAATACCGGCCCAAGTACTGC GGTTCCTGCGTGGACGGCCGATGCTGCACGCCCCAGCTGACCAGGACTGTGAAGATGCGGTTCCGCTGCGAAG ATGGGGAGACATTTTCCAAGAACGTCATGATGATCCAGTCCTGCAAATGCAACTACAACTGCCCGCATGCCAA TGAAGCAGCGTTTCCCTTCTACAGGCTGTTCAATGACATTCACAAATTTAGGGACTAAATGCTACCTGGGTTT CCAGGGCACACCTAGACAAACAAGGGAGAAGAGTGTCAGAATCAGAATCATGGAGAAAATGGGCGGGGGTGGT
GTGGGTGATGGGACTCATTGTAGAAAGGAAGCCTTGCTCATTCTTGAGGAGCATTAAGGTATTTCGAAACTGC
CAAGGGTGCTGGTGCGGATGGACACTAATGCAGCCACGATTGGAGAATACTTTGCTTCATAGTATTGGAGCAC lATGTTACTGCTTCATTTTGGAGCTTGTGGAGTTGATGACTTTCTGTTTTCTGTTTGTAAATTATTTGCTAAGC
ATATTTTCTCTAGGCTTTTTTCCTTTTGGGGTTCTACAGTCGTAAAAGAGATAATAAGATTAGTTGGACAGTT
TAAAGCTTTTATTCGTCCTTTGACAAAAGTAAATGGGAGGGCATTCCATCCCTTCCTGAAGGGGGACACTCCA iTGAGTGTCTGTGAGAGGCAGCTATCTGCACTCTAAACTGCAAACAGAAATCAGGTGTTTTAAGACTGAATGTT
TTATTTATCAAAATGTAGCTTTTGGGGAGGGAGGGGAAATGTAATACTGGAATAATTTGTAAATGATTTTAAT
TTTATATTCAGTGAAAAGATTTTATTTATGGAATTAACCATTTAATAAAGAAATATTTACCT
NOV12a, CG55688-01 SEQ ID NO: 150 381 aa !MW at 42069. lkD Protein Sequence
MSSRIARALALWTLLHLTRLALSTCPAACHCPLEAPKCAPGVGLVRDGCGCCKVCAKQLNEDCSKTQPCDHT KGLECNFGASSTALKGICRAQSEGRPCEYNSRIYQNGESFQPNCKHQCTCIDGAVGCIPLCPQELSLPNLGCP iNPRLVKVTGQCCEE VCDEDSIKDPMEDQDGLLGKELGFDASEVELTRNNELIAVGKGSSLKRIPVFGMEPRI RYNPLQGQKCIVQTTS SQCSKTCGTGISTRVTNDNPECRLVKETRICEVRPCGQPVYSSLKKGKKCSKTKKS PEPVRFTYAGCLSVKKΥRPKYCGSCVDGRCCTPQLTRTVKMRFRCEDGETFSKNVM IQSCKCNYNCPHANEA AFPFYRLFNDIHKFRD NOV12b, 254087906 SEQ ID NO: 151 1158 bp DNA Sequence ORF Start: at 1 jORF Stop: end of sequence
AGATCTACCATGAGCTCCCGCATCGCCAGGGCGCTCGCCTTAGTCGTCACCCTTCTCCACTTGACCAGGCTGG CGCTCTCCACCTGCCCCGCTGCCTGCCACTGCCCCCTGGAGGCGCCCAAGTGCGCGCCGGGAGTCGGGCTGGT CCGGGACGGCTGCGGCTGCTGTAAGGTCTGCGCCAAGCAGCTCAACGAGGACTGCAGCAAAACGCAGCCCTGC GACCACACCAAGGGGCTGGAATGCAACTTCGGCGCCAGCTCCACCGCTCTGAAGGGGATCTGCAGAGCTCAGT CAGAGGGCAGACCCTGTGAATATAACTCCAGAATCTACCAAAACGGGGAAAGTTTCCAGCCCAACTGTAAACA TCAGTGCACATGTATTGATGGCGCCGTGGGCTGCATTCCTCTGTGTCCCCAAGAACTATCTCTCCCCAACTTG GGCTGTCCCAACCCTCGGCTGGTCAAAGTTACCGGGCAGTGCTGCGAGGAGTGGGTCTGTGACGAGGATAGTA TCAAGGACCCCATGGAGGACCAGGACGGCCTCCTTGGCAAGGAGCTGGGATTCGATGCCTCCGAGGTGGAGTT GACGAGAAACAATGAATTGATTGCAGTTGGAAAAGGCAGCTCACTGAAGCGGCTCCCTGTTTTTGGAATGGAG CCTCGCATCCTATACAACCCTTTACAAGGCCAGAAATGTATTGTTCAAACAACTTCATGGTCCCAGTGCTCAA AGACCTGTGGAACTGGTATCTCCACACGAGTTACCAATGACAACCCTGAGTGCCGCCTTGTGAAAGAAACCCG GATTTGTGAGGTGCGGCCTTGTGGACAGCCAGTGTACAGCAGCCTGAAAAAGGGCAAGAAATGCAGCAAGACC AAGAAATCCCCCGAACCAGTCAGGTTTACTTACGCTGGATGTTTGAGTGTGAAGAAATACCGGCCCAAGTACT GCGGTTCCTGCGTGGACGGCCGATGCTGCACGCCCCAGCTGACCAGGACTGTGAAGATGCGGTTCCGCTGCGA AGATGGGGAGACATTTTCCAAGAACGTCATGATGATCCAGTCCTGCAAATGCAACTACAACTGCCCGCATGCC AATGAAGCAGCGTTTCCCTTCTACAGGCTGTTCAATGACATTCACAAATTTAGGGACCTCGAG
NOV12b, 254087906 SEQ ID NO: 152 386 aa MW at 42612.7kD Protein Sequence
RSTMSSRIARALALWTLLHLTRLALSTCPAACHCPLΞAPKCAPGVGLVRDGCGCCKVCAKQLNEDCSKTQPC DHTKGLECNFGASSTALKGICRAQSEGRPCEYNSRIYQNGESFQPNCKHQCTCIDGAVGCIPLCPQELSLPNL GCPNPRLVKVTGQCCEE VCDEDSIKDPMEDQDGLLGKELGFDASEVELTRNNELIAVGKGSSLKRLPVFGME PRILYNPLQGQKCIVQTTS SQCSKTCGTGISTRVTNDNPECRLVKETRICEVRPCGQPVYSSLKKGKKCSKT KKSPEPVRFTYAGCLSVKKYRPKYCGSCVDGRCCTPQLTRTVKMRFRCΞDGETFSKNVMMIQSCKCNYNCPHA NEAAFPFYRLFNDIHKFRDLE
NOV12c, 259278648 SEQ ID NO: 153 [204 bp DNA Sequence ORF Start: at 1 ORF Stop: end of sequence
[AGATCTTGTGAATATAACTCCAGAATCTACCAAAACGGGGAAAGTTTCCAGCCCAACTGTAAACATCAGTGCA CATGTATTGATGGCGCCGTGGGCTGCATTCCTCTGTGTCCCCAAGAACTATCTCTCCCCAACTTGGGCTGTCC CAACCCTCGGCTGGTCAAAGTTACCGGGCAGTGCTGCGAGGAGTGGGTCTGTCTCGAG
NOV12c, 259278648 SEQ ID NO: 154 68 aa MW at 7576.6 D
Protein Sequence
RSCEYNSRIYQNGESFQPNCKHQCTCIDGAVGCIPLCPQELSLPNLGCPNPRLVKVTGQCCEE VCLE NOV12d, 259280032 SEQ ED NO: 155 228 bp DNA Sequence ORF Start: at 1 fORF Stop: end of sequence
AGATCTTGCCCCGCTGCCTGCCACTGCCCCCTGGAGGCGCCCAAGTGCGCGCCGGGAGTCGGGCTGGTCCGGG ACGGCTGCGGCTGCTGTAAGGTCTGCGCCAAGCAGCTCAACGAGGACTGCAGCAAAACGCAGCCCTGCGACCA CACCAAGGGGCTGGAATGCAACTTCGGCGCCAGCTCCACCGCTCTGAAGGGGATCTGCAGAGCTCAGTCAGAG
GGCCTCGAG
Figure imgf000214_0001
NOV12e, 254756530 SEQ ID NO: 157 J228bp_ DNA Sequence JORF Start: at 1 JORF StopTend of sequence
AGATCTTGCCCCGCTGCCTGCCACTGCCCCCTGGAGGCGCCCAAGTGCGCGCCGGGAGTCGGGCTGGTCCGGG ACGGCTGCGGCTGCTGTAAGGTCTGCGCCAAGCAGCTCAACGAGGACTGCAGCAAAACGCAGCCCTGCGACCA CACCAAGGGGCTGGAATGCAACTTCGGCGCCAGCTCCACCGCTCTGAAGGGGATCTGCAGAGCTCAGTCAGAG GGCCTCGAG
Figure imgf000214_0002
Figure imgf000214_0003
NOV12f, 229509618 EQ ED NO: 160 408 aa MW at 45009.5kD Protein Sequence
YKKAGSARPPFTTMSSRIAR ZALALVI S VTLLHLTRLALSTCPAACHCPLEAPKCAPGVGLVRDGCGCCKVCAKQL NEDCSKTQPCDHTKGLECNFGASSTALKGICRAQSEGRPCEYNSRIYQNGESFQPNCKHQCTCIDGAVGCIPL CPQELSLPNLGCPNPRLVKVTGQCCEEWVCDEDSIKDPMEDQDGLLGKELGFDASEVELTRNNELIAVGKGSS LKRLPVFG EPRILYNPLQGQKCIVQTTSWSQCSKTCGTGISTRVTNDNPECRLVKETRICEVRPCGQPVYSS LKKGKKCSKTKKSPEPVRFTYAGCLSVKKYRPKYCGSCVDGRCCTPQLTRTVKMRFRCEDGETFSKNVMMIQS CKCNYNCPHANEAAFPFYRLFNDIHKFRDKGGRALSRVRRPRS
NOV12g, 229509658 SEQ ID NO: 161 1111 bp DNA Sequence ORF Start: at 2 JORF Stop: end of sequence
AGGCTCCGCGGCCGCCCCCTTCACCACCTGCCCCGCTGCCTGCCACTGCCCCCTGGAGGCGCCCAAGTGCGCG CCGGGAGTCGGGCTGGTCCGGGACGGCTGCGGCTGCTGTAAGGTCTGCGCCAAGCAGCTCAACGAGGACTGCA GCAAAACGCAGCCCTGCGACCACACCAAGGGGCTGGAATGCAACTTCGGCGCCAGCTCCACCGCTCTGAAGGG GATCTGCAGAGCTCAGTCAGAGGGCAGACCCTGTGAATATAACTCCAGAATCTACCAAAACGGGGAAAGTTTC CAGCCCAACTGTAAACATCAGTGCACATGTATTGATGGCGCCGTGGGCTGCATTCCTCTGTGTCCCCAAGAAC TATCTCTCCCCAACTTGGGCTGTCCCAACCCTCGGCTGGTCAAAGTTACCGGGCAGTGCTGCGAGGAGTGGGT CTGTGACGAGGATAGTATCAAGGACCCCATGGAGGACCAGGACGGCCTCCTTGGCAAGGAGCTGGGATTCGAT GCCTCCGAGGTGGAGTTAACGAGAAACAATGAATTGATTGCAGTTGGAAAAGGCAGCTCACTGAAGCGGCTCC CTGTTTTTGGAATGGAGCCTCGCATCCTATACAACCCTTTACAAGGCCAGAAATGTATTGTTCAAACAACTTC ATGGTCCCAGTGCTCAAAGACCTGTGGAACTGGTATCTCCACACGAGTTACCAATGACAACCCTGAGTGCCGC CTTGTGAAAGAAACCCGGATTTGTGAGGTGCGGCCTTGTGGACAGCCAGTGTACAGCAGCCTGAAAAAGGGCA AGAAATGCAGCAAGACCAAGAAATCCCCCGAACCAGTCAGGTTTACTTACGCTGGATGTTTGAGTGTGAAGAA ATACCGGCCCAAGTACTGCGGTTCCTGCGTGGACGGCCGATGCTGCACGCCCCAGCTGACCAGGACTGTGAAG ATGCGGTTCCGCTGCGAAGATGGGGAGACATTTTCCAAGAACGTCATGATGATCCAGTCCTGCAAATGCAACT ACAACTGCCCGCATGCCAATGAAGCAGCGTTTCCCTTCTACAGGCTGTTCAATGACATTCACAAATTTAGGGA CAAGGGTGGGCGCGCC
NOV12g, 229509658 SEQ ID NO: 162 370 aa MWat 40610.3kD Protein Sequence
GSAAAPFTTCPAACHCPLEAPKCAPGVGLVRDGCGCCKVCAKQLNEDCSKTQPCDHTKGLECNFGASSTALKG ICRAQSEGRPCEYNSRIYQNGESFQPNCKHQCTCIDGAVGCIPLCPQELSLPNLGCPNPRLVKVTGQCCEEWV CDEDSIKDP EDQDGLLGKELGFDASEVELTRNNELIAVGKGSSLKRLPVFGMEPRILYNPLQGQKCIVQTTS SQCSKTCGTGISTRVTNDNPECRLVKETRICEVRPCGQPVYSSLKKGKKCSKTKKSPEPVRFTYAGCLSVKK YRPKYCGSCVDGRCCTPQLTRTVKMRFRCEDGETFSKNVMMIQSCKCNYNCPHANEAAFPFYRLFNDIHKFRD KGGRA
NOV12h, CG55688- 2" |SEQ roNoTl63 }l068bp
DNA Sequence jORF Start: at 1 joRF Stop: end of sequence
ACCTGCCCCGCTGCCTGCCACTGCCCCCTGGAGGCGCCCAAGTGCGCGCCGGGAGTCGGGCTGGTCCGGGACG GCTGCGGCTGCTGTAAGGTCTGCGCCAAGCAGCTCAACGAGGACTGCAGCAAAACGCAGCCCTGCGACCACAC CAAGGGGCTGGAATGCAACTTCGGCGCCAGCTCCACCGCTCTGAAGGGGATCTGCAGAGCTCAGTCAGAGGGC AGACCCTGTGAATATAACTCCAGAATCTACCAAAACGGGGAAAGTTTCCAGCCCAACTGTAAACATCAGTGCA CATGTATTGATGGCGCCGTGGGCTGCATTCCTCTGTGTCCCCAAGAACTATCTCTCCCCAACTTGGGCTGTCC CAACCCTCGGCTGGTCAAAGTTACCGGGCAGTGCTGCGAGGAGTGGGTCTGTGACGAGGATAGTATCAAGGAC CCCATGGAGGACCAGGACGGCCTCCTTGGCAAGGAGCTGGGATTCGATGCCTCCGAGGTGGAGTTGACGAGAA ACAATGAATTGATTGCAGTTGGAAAAGGCAGCTCACTGAAGCGGCTCCCTGTTTTTGGAATGGAGCCTCGCAT CCTATACAACCCTTTACAAGGCCAGAAATGTATTGTTCAAACAACTTCATGGTCCCAGTGCTCAAAGACCTGT GGAACTGGTATCTCCACACGAGTTACCAATGACAACCCTGAGTGCCGCCTTGTGAAAGAAACCCGGATTTGTG AGGTGCGGCCTTGTGGACAGCCAGTGTACAGCAGCCTGAAAAAGGGCAAGAAATGCAGCAAGACCAAGAAATC CCCCGAACCAGTCAGGTTTACTTACGCTGGATGTTTGAGTGTGAAGAAATACCGGCCCAAGTACTGCGGTTCC TGCGTGGACGGCCGATGCTGCACGCCCCAGCTGACCAGGACTGTGAAGATGCGGTTCCGCTGCGAAGATGGGG AGACATTTTCCAAGAACGTCATGATGATCCAGTCCTGCAAATGCAACTACAACTGCCCGCACGCCAATGAAGC AGCGTTTCCCTTCTACAGGCTGTTCAATGACATTCACAAATTTAGG
NOV12h, CG55688-02 SEQ ID NO: 164 1356 aa MW at 39322.9kD Protein Sequence
TCPAACHCPLΞAPKCAPGVGLVRDGCGCCKVCAKQLNEDCSKTQPCDHTKGLECNFGASSTALKGICRAQSEG RPCEYNSRIYQNGESFQPNCKHQCTCIDGAVGCIPLCPQELSLPNLGCPNPRLVKVTGQCCEE VCDEDSIKD PMEDQDGLLGKELGFDASEVELTRNNELIAVGKGSSLKRLPVFGMEPRILYNPLQGQKCIVQTTSWSQCSKTC iGTGISTRVTNDNPECRLVKETRICEVRPCGQPVYSSLKKGKKCSKTKKSPEPVRFTYAGCLSVKKYRPKYCGS ICVDGRCCTPQLTRTVKMRFRCEDGETFSKNVM IQSCKCNYNCPHANEAAFPFYRLFNDIHKFR
NOV12i, CG55688-03 SEQ ID NO: 165 1198 bp
DNA Sequence (ORF StartTat 2 ORF Stop: end of sequence
GTACAAAAAAGCAGGCTCCGCGGCCGCCCCCTTCACCACCATGAGCTCCCGCATCGCCAGGGCGCTCGCCTTA GTCGTCACCCTTCTCCACTTGACCAGGCTGGCGCTCTCCACCTGCCCCGCTGCCTGCCACTGCCCCCTGGAGG CGCCCAAGTGCGCGCCGGGAGTCGGGCTGGTCCGGGACGGCTGCGGCTGCTGTAAGGTCTGCGCCAAGCAGCT CAACGAGGACTGCAGCAAAACGCAGCCCTGCGACCACACCAAGGGGCTGGAATGCAACTTCGGCGCCAGCTCC ACCGCTCTGAAGGGGATCTGCAGAGCTCAGTCAGAGGGCAGACCCTGTGAATATAACTCCAGAATCTACCAAA ACGGGGAAAGTTTCCAGCCCAACTGTAAACATCAGTGCACATGTATTGATGGCGCCGTGGGCTGCATTCCTCT GTGTCCCCAAGAACTATCTCTCCCCAACTTGGGCTGTCCCAACCCTCGGCTGGTCAAAGTTACCGGGCAGTGC TGCGAGGAGTGGGTCTGTGACGAGGATAGTATCAAGGACCCCATGGAGGACCAGGACGGCCTCCTTGGCAAGG AGCTGGGATTCGATGCCTCCGAGGTGGAGTTGACGAGAAACAATGAATTGATTGCAGTTGGAAAAGGCAGCTC ACTGAAGCGGCTCCCTGTTTTTGGAATGGAGCCTCGCATCCTATACAACCCTTTACAAGGCCAGAAATGTATT GTTCAAACAACTTCATGGTCCCAGTGCTCAAAGACCTGTGGAACTGGTATCTCCACACGAGTTACCAATGACA ACCCTGAGTGCCGCCTTGTGAAAGAAACCCGGATTTGTGAGGTGCGGCCTTGTGGACAGCCAGTGTACAGCAG CCTGAAAAAGGGCAAGAAATGCAGCAAGACCAAGAAATCCCCCGAACCAGTCAGGTTTACTTACGCTGGATGT TTGAGTGTGAAGAAATACCGGCCCAAGTACTGCGGTTCCTGCGTGGACGGCCGATGCTGCACGCCCCAGCTGA CCAGGACTGTGAAGATGCGGTTCCGCTGCGAAGATGGGGAGACATTTTCCAAGAACGTCATGATGATCCAGTC CTGCAAATGCAACTACAACTGCCCGCATGCCAATGAAGCAGCGTTTCCCTTCTACAGGCTGTTCAATGACATT CACAAATTTAGGGACAAGGGTGGGCGCGCC NOV12i, CG55688-03 SEQ ID NO: 166 399 aa MW at 43790.1kD Protein Sequence
YKKAGSAAAPFTTMSSRIARALALWTLLHLTRLALSTCPAACHCPLEAPKCAPGVGLVRDGCGCCKVCAKQL NEDCSKTQPCDHTKGLECNFGASSTALKGICRAQSEGRPCEYNSRIYQNGESFQPNCKHQCTCIDGAVGCIPL CPQELSLPNLGCPNPRLVKVTGQCCEEWVCDEDSIKDP EDQDGLLGKELGFDASEVELTRNNELIAVGKGSS LKRLPVFG EPRILYNPLQGQKCIVQTTS SQCSKTCGTGISTRVTNDNPECRLVKETRICEVRPCGQPVYSS LKKGKKCSKTKKSPEPVRFTYAGCLSVKIYRPKYCGSOraGRCCTPQLTRTVKMRFRCEDGETFSKNVMMIQS CKCNYNCPHANEAAFPFYRLFNDIHKFRDKGGRA
Figure imgf000217_0001
NOV12J, CG55688-04 SEQ ID NO: 168 370 aa MW at 40610.3kD Protein Sequence
GSAAAPFTTCPAACHCPLEAPKCAPGVGLVRDGCGCCKVCAKQLNEDCSKTQPCDHTKGLECNFGASSTALKG ICRAQSEGRPCEYNSRIYQNGESFQPNCKHQCTCIDGAVGCIPLCPQELSLPNLGCPNPRLVKVTGQCCEE V CDEDSIKDPMEDQDGLLGKELGFDASEVELTRNNELIAVGKGSSLKRLPVFGMEPRILYNPLQGQKCIVQTTS SQCSKTCGTGISTRVTNDNPECRLVKETRICEVRPCGQPVYSSLKKGKKCSKTKKSPEPVRFTYAGCLSVKK YRPKYCGSCVDGRCCTPQLTRTVK RFRCEDGETFSKNVMMIQSCKCNYNCPHANEAAFPFYRLFNDIHKFRD KGGRA
Figure imgf000218_0001
NOV12k, CG55688-05 SEQ ID NO: 170 381 aa MW at 42026.1kD
Protein Sequence
MSSRIARALALWTLLHLTRLALSTCPAACHCPLEAPKCAPGVGLVRDGCGCCKVCAKQLNEDCSKTQPCDHT KGLECNFGASSTALKGICRAQSEGRPCEYNSRIYQNGESFQPNCKHQCTCIDGAVGCIPLCPQELSLPNLGCP NPRLVKVTGQCCEE CDEDSIKDPMEDQDGLLGKELGFDASEVELTRNNELIAVGKGSSLKRLPVFGMEPRI LYNPLQGQKCIVQTTS SQCSKTCGTGISTRVTNDNPECRLVKETRICEVRPCGQPVYSSLKKGKKCSKTKKS PEPVRFTYAGCLSVKKYRPKYCGSCVDGRCCTPQLTRTVKMRFRCEDGETFSKNVMMIQSCKCNYNCPHANEA AFPFYRLFNDIHKFRD
NOV121, CG55688-06 SEQ ID NO: 171 1168 bp
DNA Sequence ORF Start: ATG at 14 |ORF Stop: TAG at 1157
CACCGGATCCACCATGAGCTCCCGCATCGCCAGGGCGCTCGCCTTAGTCGTCACCCTTCTCCACTTGACCAGG
CTGGCGCTCTCCACCTGCCCCGCTGCCTGCCACTGCCCCCTGGAGGCGCCCAAGTGCGCGCCGGGAGTCGGGC TGGTCCGGGACGGCTGCGGCTGCTGTAAGGTCTGCGCCAAGCAGCTCAACGAGGACTGCAGCAAAACGCAGCC CTGCGACCACACCAAGGGGCTGGAATGCAACTTCGGCGCCAGCTCCACCGCTCTGAAGGGGATCTGCAGAGCT CAGTCAGAGGGCAGACCCTGTGAATATAACTCCAGAATCTACCAAAACGGGGAAAGTTTCCAGCCCAACTGTA AACATCAGTGCACATGTATTGATGGCGCCGTGGGCTGCATTCCTCTGTGTCCCCAAGAACTATCTCTCCCCAA CTTGGGCTGTCCCAACCCTCGGCTGGTCAAAGTTACCGGGCAGTGCTGCGAGGAGTGGGTCTGTGACGAGGAT AGTATCAAGGACCCCATGGAGGACCAGGACGGCCTCCTTGGCAAGGAGCTGGGATTCGATGCCTCCGAGGTGG AGTTGACGAGAAACAATGAATTGATTGCAGTTGGAAAAGGCAGCTCACTGAAGCGGCTCCCTGTTTTTGGAAT GGAGCCTCGCATCCTATACAACCCTTTACAAGGCCAGAAATGTATTGTTCAAACAACTTCATGGTCCCAGTGC TCAAAGACCTGTGGAACTGGTATCTCCACACGAGTTACCAATGACAACCCTGAGTGCCGCCTTGTGAAAGAAA CCCGGATTTGTGAGGTGCGGCCTTGTGGACAGCCAGTGTACAGCAGCCTGAAAAAGGGCAAGAAATGCAGCAA GACCAAGAAATCCCCCGAACCAGTCAGGTTTACTTACGCTGGATGTTTGAGTGTGAAGAAATACCGGCCCAAG TACTGCGGTTCCTGCGTGGACGGCCGATGCTGCACGCCCCAGCTGACCAGGACTGTGAAGATGCGGTTCCGCT GCGAAGATGGGGAGACATTTTCCAAGAACGTCATGATGATCCAGTCCTGCAAATGCAACTACAACTGCCCGCA TGCCAATGAAGCAGCGTTTCCCTTCTACAGGCTGTTCAATGACATTCACAAATTTAGGGACTAGGTCGACGGC NOV121, CG55688-06 SEQ ID NO: 172 381 aa MW at 42026. lkD Protein Sequence SSRIARALALWTLLHLTRLALSTCPAACHCPLEAPKCAPGVGLVRDGCGCCKVCAKQLNEDCSKTQPCDHT KGLECNFGASSTALKGICRAQSEGRPCEYNSRIYQNGESFQPNCKHQCTCIDGAVGCIPLCPQELSLPNLGCP NPRLVKVTGQCCΞEWVCDEDSIKDPMEDQDGLLGKELGFDASEVELTRNNELIAVGKGSSLKRLPVFGMEPRI LYNPLQGQKCIVQTTS SQCSKTCGTGISTRVTNDNPECRLVKETRICEVRPCGQPVYSSLKKGKKCSKTKKS PEPVRFTYAGCLSVK YRPKYCGSCVDGRCCTPQLTRTVKMRFRCEDGETFSKlvrVMMIQSCKCNYNCPHANEA AFPFYRLFNDIHKFRD
NOV12m, SNP13376428 of SEQ ID NO: 173 1887 bp CG55688-01, DNA Sequence ORF Start: ATG at 81 JORF Stop: TAA at 1224
SNP Pos: 571 SNP Change: A to G
GCGCACGGCCTGTCCGCTGCACACCAGCTTGTTGGCGTCTTCGTCGCCGCGCTCGCCCCGGGCTACTCCTGCG
CGCCACAATGAGCTCCCGCATCGCCAGGGCGCTCGCCTTAGTCGTCACCCTTCTCCACTTGACCAGGCTGGCG
CTCTCCACCTGCCCCGCTGCCTGCCACTGCCCCCTGGAGGCGCCCAAGTGCGCGCCGGGAGTCGGGCTGGTCC GGGACGGCTGCGGCTGCTGTAAGGTCTGCGCCAAGCAGCTCAACGAGGACTGCAGCAAAACGCAGCCCTGCGA CCACACCAAGGGGCTGGAATGCAACTTCGGCGCCAGCTCCACCGCTCTGAAGGGGATCTGCAGAGCTCAGTCA GAGGGCAGACCCTGTGAATATAACTCCAGAATCTACCAAAACGGGGAAAGTTTCCAGCCCAACTGTAAACATC AGTGCACATGTATTGATGGCGCCGTGGGCTGCATTCCTCTGTGTCCCCAAGAACTATCTCTCCCCAACTTGGG CTGTCCCAACCCTCGGCTGGTCAAAGTTACCGGGCAGTGCTGCGAGGAGTGGGTCTGTGGCGAGGATAGTATC AAGGACCCCATGGAGGACCAGGACGGCCTCCTTGGTAAGGAGCTGGGATTCGATGCCTCCGAGGTGGAGTTGA CGAGAAACAATGAATTGATTGCAGTTGGAAAAGGCAGCTCACTGAAGCGGATCCCTGTTTTTGGAATGGAGCC TCGCATCCGATACAACCCTTTACAAGGCCAGAAATGTATTGTTCAAACAACTTCATGGTCCCAGTGCTCAAAG ACCTGTGGAACTGGTATCTCCACACGAGTTACCAATGACAACCCTGAGTGCCGCCTTGTGAAAGAAACCCGGA TTTGTGAGGTGCGGCCTTGTGGACAGCCAGTGTACAGCAGCCTGAAAAAGGGCAAGAAATGCAGCAAGACCAA GAAATCCCCCGAACCAGTCAGGTTTACTTACGCTGGATGTTTGAGTGTGAAGAAATACCGGCCCAAGTACTGC GGTTCCTGCGTGGACGGCCGATGCTGCACGCCCCAGCTGACCAGGACTGTGAAGATGCGGTTCCGCTGCGAAG ATGGGGAGACATTTTCCAAGAACGTCATGATGATCCAGTCCTGCAAATGCAACTACAACTGCCCGCATGCCAA TGAAGCAGCGTTTCCCTTCTACAGGCTGTTCAATGACATTCACAAATTTAGGGACTAAATGCTACCTGGGTTT CCAGGGCACACCTAGACAAACAAGGGAGAAGAGTGTCAGAATCAGAATCATGGAGAAAATGGGCGGGGGTGGT
GTGGGTGATGGGACTCATTGTAGAAAGGAAGCCTTGCTCATTCTTGAGGAGCATTAAGGTATTTCGAAACTGC
CAAGGGTGCTGGTGCGGATGGACACTAATGCAGCCACGATTGGAGAATACTTTGCTTCATAGTATTGGAGCAC iATGTTACTGCTTCATTTTGGAGCTTGTGGAGTTGATGACTTTCTGTTTTCTGTTTGTAAATTATTTGCTAAGC lATATTTTCTCTAGGCTTTTTTCCTTTTGGGGTTCTACAGTCGTAAAAGAGATAATAAGATTAGTTGGACAGTT iTAAAGCTTTTATTCGTCCTTTGACAAAAGTAAATGGGAGGGCATTCCATCCCTTCCTGAAGGGGGACACTCCA
TGAGTGTCTGTGAGAGGCAGCTATCTGCACTCTAAACTGCAAACAGAAATCAGGTGTTTTAAGACTGAATGTT
TTATTΓATCAAAATGTAGCTTTTGGGGAGGGAGGGGAAATGTAATACTGGAATAATTTGTAAATGATTTTAAT
TTTATATTCAGTGAAAAGATTTTATTTATGGAATTAACCATTTAATAAAGAAATATTTACCT
NOV12m, SNP13376428 of |SEQ IDNO: 174 381 aa MW at 42011.1kD CG55688-01, Protein Sequence SNP Pos: 164 SNP Change: Asp to Gly
MSSRIARALALWTLLHLTRLALSTCPAACHCPLEAPKCAPGVGLVRDGCGCCKVCAKQLNEDCSKTQPCDHT KGLECNFGASSTALKGICRAQSEGRPCEYNSRIYQNGESFQPNCKHQCTCIDGAVGCIPLCPQELSLPNLGCP NPRLVKVTGQCCEEWVCGEDSIKDPMEDQDGLLGKELGFDASEVELTRNNELIAVGKGSSLKRIPVFG EPRI RYNPLQGQKCIVQTTS SQCSKTCGTGISTRVTNDNPECRLVKETRICEVRPCGQPVYSSLKKGKKCSKTKKS PEPVRFTYAGCLSVKKyRPKYCGSCVDGRCCTPQLTRTVKMRFRCEDGETFSKNVMMIQSCKCNYNCPHANEA AFPFYRLFNDIHKFRD A ClustalW comparison of the above protein sequences yields the following sequence alignment shown in Table 12B.
Table 12B. Comparison of the NOV12 protein sequences.
NOV12a MSSRIARALALWTLLHLTRLALSTCPAACHCPLEAPKCAPGVGLVR
NOV12b RSTMSSRIARALALWTLLHLTRLALSTCPAACHCPLEAPKCAPGVGLVR
NOV12c RSCEYNSRIYQNGESFQPNCKHQC
NOV12d RSCPAACHCPLEAPKCAPGVGLVR
NOV12e RSCPAACHCPLEAPKCAPGVGLVR
NOV12f YKKAGSARPPFTTMSSRIARALALWTLLHLTRLALSTCPAACHCPLEAPKCAPGVGLVR
NOV12g GSAAAPFTTCPAACHCPLEAPKCAPGVGLVR
NOV12h TCPAACHCPLEAPKCAPGVGLVR
NOV12i YKKAGSAAAPFTTMSSRIARALALWTLLHLTRLALSTCPAACHCPLEAPKCAPGVGLVR
NOV12j GSAAAPFTTCPAACHCPLEAPKCAPGVGLVR
NOV12k -MSSRIARALALWTLLHLTRLALSTCPAACHCPLEAPKCAPGVGLVR
NOV121 MSSRIARALALWTLLHLTRLALSTCPAACHCPLEAPKCAPGVGLVR
NOV12a DGCGCCKVCAKQLNEDCSKTQPCDHTKGLECNFGASSTALKGICRAQSEGRPCEYNSRIY
NOV12b DGCGCCKVCAKQLNEDCSKTQPCDHTKGLECNFGASSTALKGICRAQSEGRPCEYNSRIY
NOV12C TCIDGAVGCIPLCPQELSLPNLGCPNPRLVKVTGQCCEEWVCLE
NOV12d DGCGCCKVCAKQLNEDCSKTQPCDHTKGLECNFGASSTALKGICRAQSEGLE
NOV12e, DGCGCCKVCAKQLNEDCSKTQPCDHTKGLECNFGASSTALKGICRAQSEGLE
NOV12f DGCGCCKVCAKQLNEDCSKTQPCDHTKGLECNFGASSTALKGICRAQSEGRPCEYNSRIY
NOV12g DGCGCCKVCAKQLNEDCSKTQPCDHTKGLECNFGASSTALKGICRAQSEGRPCEYNSRIY
NOV12h DGCGCCKVCAKQLNEDCSKTQPCDHTKGLECNFGASSTALKGICRAQSEGRPCEYNSRIY
NOV12i DGCGCCKVCAKQLNEDCSKTQPCDHTKGLECNFGASSTALKGICRAQSEGRPCEYNSRIY
NOV12J DGCGCCKVCAKQLNEDCSKTQPCDHTKGLECNFGASSTALKGICRAQSEGRPCEYNSRIY
NOV12k DGCGCCKVCAKQLNEDCSKTQPCDHTKGLECNFGASSTALKGICRAQSEGRPCEYNSRIY
NOV121 DGCGCCKVCAKQLNEDCSKTQPCDHTKGLECNFGASSTALKGICRAQSEGRPCEYNSRIY
NOV12a QNGESFQPNCKHQCTCIDGAVGCIPLCPQELSLPNLGCPNPRLVKVTGQCCEE VCDEDS
NOV12b QNGESFQPNCKHQCTCIDGAVGCIPLCPQELSLPNLGCPNPRLVKVTGQCCEE VCDEDS
NOV12C
NOV12d
NOV12e
NOV12f QNGESFQPNCKHQCTCIDGAVGCIPLCPQELSLPNLGCPNPRLVKVTGQCCEEWVCDEDS
NOV12g QNGESFQPNCKHQCTCIDGAVGCIPLCPQELSLPNLGCPNPRLVKVTGQCCEEWVCDEDS
NOV12h QNGESFQPNCKHQCTCIDGAVGCIPLCPQELSLPNLGCPNPRLVKVTGQCCEEWVCDEDS
NOV12i QNGESFQPNCKHQCTCIDGAVGCIPLCPQELSLPNLGCPNPRLVKVTGQCCEEWVCDEDS
NOV12J QNGESFQPNCKHQCTCIDGAVGCIPLCPQELSLPNLGCPNPRLVKVTGQCCEE VCDEDS
NOV12k QNGESFQPNCKHQCTCIDGAVGCIPLCPQELSLPNLGCPNPRLVKVTGQCCEE VCDEDS
NOV121 QNGESFQPNCKHQCTCIDGAVGCIPLCPQELSLPNLGCPNPRLVKVTGQCCEE VCDEDS
NOV12a IKDPMEDQDGLLGKELGEDASEVELTRNNELIAVGKGSSLKRIPVFGMEPRIRYNPLQGQ
NOV12b IKDPMEDQDGLLGKELGFDASEVELTRNNELIAVGKGSSLKRLPVFGMEPRILYNPLQGQ
NOV12C
NOV12d
NOV12e
NOV12f IKDPMEDQDGLLGKELGFDASEVELTRNNELIAVGKGSSLKRLPVFGMEPRILYNPLQGQ
NOV12g IKDPMEDQDGLLGKELGFDASEVELTRNNELIAVGKGSSLKRLPVFGMEPRILYNPLQGQ
NOV12h IKDPMEDQDGLLGKELGFDASEVELTRNNELIAVGKGSSLKRLPVFGMEPRILYNPLQGQ
NOV12i IKDPMEDQDGLLGKELGFDASEVELTRNNELIAVGKGSSLKRLPVFGMEPRILYNPLQGQ
NOV12J IKDPMEDQDGLLGKELGFDASEVELTRNNELIAVGKGSSLKRLPVFGMEPRILYNPLQGQ
NOV12k IKDPMEDQDGLLGKELGFDASEVELTRNNELIAVGKGSSLKRLPVFG EPRILYNPLQGQ
NOV121 IKDPMEDQDGLLGKELGFDASEVELTRNNELIAVGKGSSLKRLPVFGMEPRILYNPLQGQ NOV12a KCIVQTTS SQCSKTCGTGISTRVTNDNPECRLVKETRICEVRPCGQPVYSSLKKGKKCS
NOV12b KCIVQTTS SQCSKTCGTGISTRVTNDNPECRLVKETRICEVRPCGQPVYSSLKKGKKCS
NOV12c
NOV12d
NOV12e
NOV12f KCIVQTTSWSQCSKTCGTGISTRVTNDNPECRLVKETRICEVRPCGQPVYSSLKKGKKCS
NOV12g KCIVQTTS SQCSKTCGTGISTRVTNDNPECRLVKETRICEVRPCGQPVYSSLKKGKKCS
NOV12h KCIVQTTS SQCSKTCGTGISTRVTNDNPECRLVKETRICEVRPCGQPVYSSLKKGKKCS
NOV12i KCIVQTTSWSQCSKTCGTGISTRVTNDNPECRLVKETRICEVRPCGQPVYSSLKKGKKCS
NOVl2j KCIVQTTSWSQCSKTCGTGISTRVTNDNPECRLVKETRICEVRPCGQPVYSSLKKGKKCS
NOV12k KCIVQTTS SQCSKTCGTGISTRVTNDNPECRLVKETRICEVRPCGQPVYSSLKKGKKCS
NOV121 KCIVQTTS SQCSKTCGTGISTRVTNDNPECRLVKETRICEVRPCGQPVYSSLKKGKKCS
NOV12a KTKKSPEPVRFTYAGCLSVKKYRPKYCGSCVDGRCCTPQLTRTVKMRFRCEDGETFSKNV
NOVl2b KTKKSPEPVRFTYAGCLSVKKYRPKYCGSCVDGRCCTPQLTRTVKMRFRCEDGETFSKNV
NOV12c
NOV12d
NOV12e
NOVl2f KTKKSPEPVRFTYAGCLSVKKYRPKYCGSCVDGRCCTPQLTRTVKMRFRCEDGETFSKNV
NOV12g KTKKSPΞPVRFTYAGCLSVKKYRPKYCGSCVDGRCCTPQLTRTVKMRFRCEDGETFSKNV
NOV12h KTKKSPEPVRFTYAGCLSVKKYRPKYCGSCVDGRCCTPQLTRTVKMRFRCEDGETFSKNV
NOV12i KTKKSPEPVRFTYAGCLSVKKYRPKYCGSCVDGRCCTPQLTRTVKMRFRCEDGETFSKNV
NOV12J KTKKSPEPVRFTYAGCLSVKKYRPKYCGSCVDGRCCTPQLTRTVKMRFRCEDGETFSKNV
NOV12k KTKKSPEPVRFTYAGCLSVKKYRPKYCGSCVDGRCCTPQLTRTVKMRFRCEDGETFSKNV
NOV121 KTKKSPEPVRFTYAGCLSVKKYRPKYCGSCVDGRCCTPQLTRTVKMRFRCEDGETFSKNV
NOV12a MMIQSCKCNYNCPHANEAAFPFYRLFNDIHKFRD
NOV12b MMIQSCKCNYNCPHANEAAFPFYRLFNDIHKFRDLE
NOV12c
NOV12d
NOV12e
NOV12f MMIQSCKCNYNCPHANEAAFPFYRLFNDIHKFRDKGGRALSRVRRPRS
NOV12g MMIQSCKCNYNCPHANEAAFPFYRLFNDIHKFRDKGGRA
NOV12h MMIQSCKCNYNCPHANEAAFPFYRLFNDIHKFR
NOVl2i MMIQSCKCNYNCPHANEAAFPFYRLFNDIHKFRDKGGRA
NOV12j MMIQSCKCNYNCPHANEAAFPFYRLFNDIHKFRDKGGRA
NOV12k MMIQSCKCNYNCPHANEAAFPFYRLFNDIHKFRD
NOV121 MMIQSCKCNYNCPHANEAAFPFYRLFNDIHKFRD
NOVl a (SEQ ID NO 150)
NOV12b (SEQ ID NO 152)
NOVl c (SEQ ID NO 154)
NOV12d (SEQ ID NO 156)
NOVl2e (SEQ ID NO 158)
NOVl2f (SEQ ID NO 160)
NOVl2g (SEQ ID NO 162)
NOV12h (SEQ ID NO 164)
NOVl2i (SEQ ID NO 166)
NOVl2j (SEQ ID NO 168)
NOVl2k (SEQ ID NO 170)
NOVl 1 (SEQ ID NO 172)
Further analysis of the NOV12a protein yielded the following properties shown in Table 12C. Table 12C. Protein Sequence Properties NOV12a
SignalP analysis: Cleavage site betweenresidues 25 and 26
PSORT II analysis:
PSG: a new signal peptide prediction method
N-region: length 7; pos.chg 2; neg.chg 0 H-region: length 12; peak value 10.04 PSG score: 5.64
GvH: von Heijne's method for signal seq. recognition GvH score (threshold: -2.1): -1.17 possible cleavage site: between 21 and 22
>>> Seems to have a cleavable signal peptide (1 to 21)
ALOM: Klein et al ' s method for TM region allocation Init position for calculation: 22
Tentative number of TMS(s) for the threshold 0.5: 0 number of TMS(s) .. fixed PERIPHERAL Likelihood = 5.73 (at 124) ALOM score: 5.73 (number of TMSs : 0)
MTOP: Prediction of membrane topology (Hartmann et al.) Center position for calculation: 10 Charge difference: -1.0 C( 2.0) - N( 3.0) N >= C: N-terminal side will be inside
MITDISC: discrimination of mitochondrial targeting seq R content: 3 Hyd Moment (75) : 7.35 Hyd Moment (95) : 12.47 G content: 0 D/E content: 1 S/T content: 6 Score: 0.47
Gavel : prediction of cleavage sites for mitochondrial preseg R-2 motif at 57 VRD|GC
NUCDISC: discrimination of nuclear localization signals pat4 : none pat7 : none bipartite: none content of basic residues: 13.9% NLS Score: -0.47
KDEL: ER retention motif in the C-terminus : none
ER Membrane Retention Signals:
XXRR-like motif in the N-terminus : SSRI KKXX-like motif in the C-terminus : HKFR
SKL: peroxisomal targeting signal in the C-terminus : none
PTS2 : 2nd peroxisomal targeting signal : none
VAC: possible vacuolar targeting motif: none
RNA-binding motif: none
Actinin-type actin-binding motif: type 1 : none type 2 : none
NMYR: N-myristoylation pattern : none Prenyl tion motif: none memYQRL: transport motif from cell surface to Golgi: none
Tyrosines in the tail : none
Dileucine motif in the tail : none checking 63 PROSITE DNA binding motifs: none checking 71 PROSITE ribosomal protein motifs: none checking 33 PROSITE prokaryotic DNA binding motifs: none
NNCN: Reinhardt's method for Cytoplasmic/Nuclear discrimination Prediction: nuclear Reliability: 94.1
COIL: Lupas's algorithm to detect coiled-coil regions total : 0 residues
Final Results (k = 9/23) :
56.5 % mitochondrial
17.4 % extracellular, including cell wall
17.4 % nuclear
8.7 % cytoplasmic
» prediction for CG55688-01 is mit (k=23)
A search of the NOVl 2a protein against the Geneseq database, a proprietary database that contains sequences published in patents and patent publication, yielded several homologous proteins shown in Table 12D.
Figure imgf000224_0001
In a BLAST search of public sequence databases, the NOVl 2a protein was found to have homology to the proteins shown in the BLASTP data in Table 12E.
Figure imgf000225_0001
PFam analysis predicts that the NOVl 2a protein contains the domains shown in the Table 12F.
Figure imgf000225_0002
Example 13.
The NOVl 3 clone was analyzed, and the nucleotide and encoded polypeptide sequences are shown in Table 13 A.
Table 13A. NOV13 Sequence Analysis
[NOV13a, CG56768-01 SEQ ID NO: 175 11214 bp
DNA Sequence ORF Start: ATG at 60 ORF Stop: TAG at 1155
CTCCTTTCTTCCCTCTCCAGAAGTCCATTGGAATATTAAGCCCAGGAGTTGCTTTGGGGATGGCTGGAAGTGC
AATGTCTTCCAAGTTCTTCCTAGTGGCTTTGGCCATATTTTTCTCCTTCGCCCAGGTTGTAATTGAAGCCAAT TCTTGGTGGTCGCTAGGTATGAATAACCCTGTTCAGATGTCAGAAGTATATATTATAGGAGCACAGCCTCTCT GCAGCCAACTGGCAGGACTTTCTCAAGGACAGAAGAAACTGTGCCACTTGTATCAGGACCACATGCAGTACAT CGGAGAAGGCGCGAAGACAGGCATCAAAGAATGCCAGTATCAATTCCGACATCGAAGGTGGAACTGCAGCACT GTGGATAACACCTCTGTTTTTGGCAGGGTGATGCAGATAGGCAGCCGCGAGACGGCCTTCACATACGCGGTGA GCGCAGCAGGGGTGGTGAACGCCATGAGCCGGGCGTGCCGCGAGGGCGAGCTGTCCACCTGCGGCTGCAGCCG CGCCGCGCGCCCCAAGGACCTGCCGCGGGACTGGCTCTGGGGCGGCTGCGGCGACAACATCGACTATGGCTAC CGCTTTGCCAAGGAGTTCGTGGACGCCCGCGAGCGGGAGCGCATCCACGCCAAGGGCTCCTACGAGAGTGCTC GCATCCTCATGAACCTGCACAACAACGAGGCCGGCCGCAGGACGGTGTACAACCTGGCTGATGTGGCCTGCAA GTGCCATGGGGTGTCCGGCTCATGTAGCCTGAAGACATGCTGGCTGCAGCTGGCAGACTTCCGCAAGGTGGGT GATGCCCTGAAGGAGAAGTACGACAGCGCGGCGGCCATGCGGCTCAACAGCCGGGGCAAGTTGGTACAGGTCA ACAGCCGCTTCAACTCGCCCACCACACAAGACCTGGTCTACATCGACCCCAGCCCTGACTACTGCGTGCGCAA TGAGAGCACCGGCTCGCTGGGCACGCAGGGCCGCCTGTGCAACAAGACGTCGGAGGGCATGGATGGCTGCGAG CTCATGTGCTGCGGCCGTGGCTACGACCAGTTCAAGACCGTGCAGACGGAGCGCTGCCACTGCAAGTTCCACT GGTGCTGCTACGTCAAGTGCAAGAAGTGCACGGAGATCGTGGACCAGTTTGTGTGCAAGTAGTGGGTGCCACC CAGCACTCAGCCCCGCCCCCAGGACCCGCTTATTTATAGAAAGTAC
NOVl 3a, CG56768-01 SEQ ID NO: 176 365 aa MW at 40886.3kD Protein Sequence
MAGSAMSSKFFLVAIJAIFFSFAQVVIEANS SLG^1 NPVQMSEVYIIGAQP CSQL GLSQGQKKLCHLYQD HMQYIGEGAKTGIKECQYQFRHRRWNCSTVDNTSVFGRVMQIGSRETAFTYAVSAAGWNAMSRACREGELST CGCSRAARPKDLPRDWL GGCGDNIDYGYRFAKEFVDARERERIHAKGSYESARILMNLHNNEAGRRTVYNLA DVACKCHGVSGSCSLKTCWLQLADFRKVGDALKEKYDSAAAMRLNSRGKLVQVNSRFNSPTTQDLVYIDPSPD YCVRNESTGSLGTQGRLCNKTSEGMDGCELMCCGRGYDQFKTVQTERCHCKFH CCYVKCKKCTEIVDQFVCK
NOV13b, CG56768-02 SEQ ID NO: 177 1026 bp
DNA Sequence |θRF Start: at 7 JORF Stop: at 1021
GGATCCGCCAATTCTTGGTGGTCGCTAGGTATGAATAACCCTGTTCAGATGTCAGAAGTATATATTATAGGAG
CACAGCCTCTCTGCAGCCAACTGGCAGGACTTTCTCAAGGACAGAAGAAACTGTGCCACTTGTATCAGGACCA CATGCAGTACATCGGAGAAGGCGCGAAGACAGGCATCAAAGAATGCCAGTATCAATTCCGACATCGAAGGTGG AACTGCAGCACTGTGGATAACACCTCTGTTTTTGGCAGGGTGATGCAGATAGGCAGCCGCGAGACGGCCTTCA CATACGCGGTGAGCGCAGCAGGGGTGGTGAACGCCATGAGCCGGGCGTGCCGCGAGGGCGAGCTGTCCACCTG CGGCTGCAGCCGCGCCGCGCGCCCCAAGGACCTGCCGCGGGACTGGCTCTGGGGCGGCTGCGGCGACAACATC GACTATGGCTACCGCTTTGCCAAGGAGTTCGTGGACGCCCGCGAGCGGGAGCGCATCCACGCCAAGGGCTCCT ACGAGAGTGCTCGCATCCTCATGAACCTGCACAACAACGAGGCCGGCCGCAGGACGGTGTACAACCTGGCTGA TGTGGCCTGCAAGTGCCATGGGGTGTCCGGCTCATGTAGCCTGAAGACATGCTGGCTGCAGCTGGCAGACTTC CGCAAGGTGGGTGATGCCCTGAAGGAGAAGTACGACAGCGCGGCGGCCATGCGGCTCAACAGCCGGGGCAAGT TGGTACAGGTCAACAGCCGCTTCAACTCGCCCACCACACAAGACCTGGTCTACATCGACCCCAGCCCTGACTA CTGCGTGCGCAATGAGAGCACCGGCTCGCTGGGCACGCAGGGCCGCCTGTGCAACAAGACGTCGGAGGGCATG GATGGCTGCGAGCTCATGTGCTGCGGCCGTGGCTACGACCAGTTCAAGACCGTGCAGACGGAGCGCTGCCACT GCAAGTTCCACTGGTGCTGCTACGTCAAGTGCAAGAAGTGCACGGAGATCGTGGACCAGTTTGTGTGCAAGCT CGAG _____ NOVl 3b, CG56768-02 SEQ ID NO: 178 338 aa MW at 37991.8kD Protein Sequence
ANS SLGi πsTNPVQMSEVYIIGAQPLCSQLAGLSQGQKKLCHLYQDHMQYIGEGAKTGIKECQYQFRHRRWNC STVDNTSVFGRVMQIGSRETAFTYAVSAAGWNA SRACREGELSTCGCSRAARPKDLPRDWLWGGCGDNIDY GYRFAKEFVDARERERIHAKGSYESARILMNLHNNEAGRRTVYNLADVACKCHGVSGSCSLKTCWLQLADFRK VGDALKEKYDSAAAMRLNSRGKLVQVNSRFNSPTTQDLVYIDPSPDYCVRNESTGSLGTQGRLCNKTSEGMDG CELMCCGRGYDQFKTVQTERCHCKFHWCCYVKCKKCTEIVDQFVCK
Figure imgf000227_0001
NOV13c, CG56768-03 SEQ ID NO: 180 380 aa MW at42082.8kD Protein Sequence
LQKSIGILSPGVALG AGSA SSKFFLVALAIFFSFAQWIEANSW SLGMNNPVQMSEVYIIGAQPLCSQLA GLSQGQK LCHLYQDHMQYIGEGAKTGIKECQYQFRHRRWNCSTVDNTSVFGRVMQIGSRETAFTYAVSAAGV VNAMSRACREGELSTCGCSRAARPKDLPRDWLWGGSGATNKKGYRSAKEIVHARERGRIHAKGSYESARILMN LHNNEAGRRTλ ZNLADVACKCHGVSGSCSLKTC LQLADFRKVGDALKEKYDSAAAMRLNSRGKLVQVNSRFN SPTTQDLVYIDPSPDYCVRNESTGSLGTQGRLCNKTSEGMDGCELMCCGRGYDQFKTVQTERCHCKFH CCYV KCK CTE I VDQFVCK
A ClustalW comparison of the above protein sequences yields the following sequence alignment shown in Table 13B. Table 13B. Comparison of the NOV13 protein sequences.
NOVl3a AGSAMSSKFFLVALAIFFSFAQWIEANS SLGMNNPVQMSEV
NOV13b ANSWWSLGMNNPVQMSEV
NOV13c LQKSIGILSPGVALGMAGSAMSSKFFLVALAIFFSFAQWIEANSWWSLGMNNPVQMSEV
NOV13a YIIGAQPLCSQLAGLSQGQKKLCHLYQDH QYIGEGAKTGIKECQYQFRHRRWNCSTVDN
NOV13b YIIGAQPLCSQLAGLSQGQ KLCHLYQDHMQYIGEGAKTGIKECQYQFRHRRWNCSTVDN
NOV13C YIIGAQPLCSQLAGLSQGQKKLCHLYQDHMQYIGEGAKTGIKECQYQFRHRR NCSTVDN
NOVl3a TSVFGRVMQIGSRETAFTYAVSAAGVVNAMSRACREGELSTCGCSRAARPKDLPRDWLWG
NOV13b TSVFGRVMQIGSRETAFTYAVSAAGWNAMSRACREGELSTCGCSRAARPKDLPRDWL G
NOV13c TSVFGRVMQIGSRETAFTYAVSAAGWNAMSRACREGELSTCGCSRAARPKDLPRD L G
NOV13a GCGDNIDYGYRFAKEFVDARERERIHAKGSYESARILMNLHNNEAGRRTVYNLADVACKC
NOV13b GCGDNIDYGYRFAKEFVDARERERIHAKGSYESARILMNLHNNEAGRRTVYNLADVACKC
NOV13c GSGATNKKGYRSAKEIVHARERGRIHAKGSYESARILMNLHNNEAGRRTVYNLADVACKC
NOVl3a HGVSGSCSLKTCWLQLADFRKVGDALKEKYOSAAAMRLNSRGKLVQVNSRFNSPTTQDLV
NOV13b HGVSGSCSLKTCWLQLADFRKVGDALKEKYDSAAAMRLNSRGKLVQVNSRFNSPTTQDLV
NOVl3c HGVSGSCSLKTC LQLADFRKVGDALKEKYDSAAAMRLNSRGKLVQVNSRFNSPTTQDLV
NOV13a YIDPSPDYCVRNESTGSLGTQGRLCNKTSΞGMDGCELMCCGRGYDQFKTVQTERCHCKFH
NOV13b YIDPSPDYCVRNESTGSLGTQGRLCNKTSEGMDGCELMCCGRGYDQFKTVQTERCHCKFH
NOV13C YIDPSPDYCVRNESTGSLGTQGRLCNKTSEGMDGCELMCCGRGYDQFKTVQTERCHCKFH
NOV13a WCCYVKCKKCTEIVDQFVCK NOV13b WCCYVKCKKCTEIVDQFVCK NOV13C WCCYVKCKKCTEIVDQFVCK
NOV13 (SEQ ID NO 176) NOV13b (SEQ ID NO 178) NOV13C (SEQ ID NO 180)
Further analysis of the NOV13a protein yielded the following properties shown in Table 13C.
Table 13C. Protein Sequence Properties NOV13a
SignalP analysis: Cleavage site between residues 28 and 29
PSORT II analysis:
PSG: a new signal peptide prediction method
N-region: length 9; pos . chg 1 ; neg .chg 0 H-region: length 17 ; peak value 11.46 PSG score: 7.06
GvH: von Heijne ' s method for signal seq. recognition GvH score (threshold: -2.1) : 2.33 possible cleavage site: between 22 and 23
>>> Seems to have a cleavable signal peptide (1 to 22)
ALOM: Klein et al ' s method for TM region allocation Init position for calculation: 23 Tentative number of TMS(s) for the threshold 0.5: 0 number of TMS(s) .. fixed
PERIPHERAL Likelihood = 4.51 (at 45)
ALOM score: 4.51 (number of TMSs: 0)
MTOP: Prediction of membrane topology (Hartmann et al.) Center position for calculation: 11 Charge difference: -3.0 , C(-1.0) - N( 2.0) N >= C: N-terminal side will be inside
MITDISC: discrimination of mitochondrial targeting seq R content: 0 Hyd Moment (75) : 3.30 Hyd Moment (95): 2.27 G content: 1 D/E content : 1 S/T content : 4 Score: -5.10
Gavel : prediction of cleavage sites for mitochondrial preseq cleavage site motif not found
NUCDISC: discrimination of nuclear localization signals pat4: RHRR (3) at 94 pat7 : none bipartite: none content of basic residues : 12.6% NLS Score: -0.29
KDEL: ER retention motif in the C-terminus : none
ER Membrane Retention Signals: none
SKL: peroxisomal targeting signal in the C-terminus : none
PTS2: 2nd peroxisomal targeting signal: none
VAC: possible vacuolar targeting motif: none
RNA-binding motif: none
Actinin-type actin-binding motif: type 1 : none type 2 : none
NMYR: N-myristoylation pattern : none
Prenylation motif.- none memYQRL: transport motif from cell surface to Golgi: none
Tyrosines in the tail: none
Dileucine motif in the tail: none checking 63 PROSITE DNA binding motifs: none checking 71 PROSITE ribosomal protein motifs: none checking 33 PROSITE prokaryotic DNA binding motifs: none
NNCN: Reinhardt's method for Cytoplasmic/Nuclear discrimination Prediction: cytoplasmic Reliability: 55.5
COIL: Lupas's algorithm to detect coiled-coil regions total: 0 residues Final Results (k = 9/23 ) :
22 .2 % : extracellular, including cell wall
22 .2 % vacuolar
22.2 % mitochondrial
22 .2 % endoplas ic reticulum
11.1 % Golgi
» prediction for CG56768-01 is exc (k=9)
A search of the NOVl 3a protein against the Geneseq database, a proprietary database that contains sequences published in patents and patent publication, yielded several homologous proteins shown in Table 13D.
Figure imgf000230_0001
In a BLAST search of public sequence databases, the NOVl 3a protein was found to have homology to the proteins shown in the BLASTP data in Table 13E.
Figure imgf000231_0001
PFam analysis predicts that the NOVl 3a protein contains the domains shown in the Table 13F.
Figure imgf000231_0002
Example 14.
The NOVl 4 clone was analyzed, and the nucleotide and encoded polypeptide sequences are shown in Table 14A. Table 14A. NOV14 Sequence Analysis
NOV14a, CG57054-03 SEQ ID NO: 181 1215 bp DNA Sequence ORF Start: ATG at 55 jORF Stop: TGA at 1165
CCAAACCACTGGAGGTCCTGATCGATCTGCCCACCGGAGCCTCCGGGCTTCGACATGCTGGAGGAGCCCCGGC
CGCGGCCTCCGCCCTCGGGCCTCGCGGGTCTCCTGTTCCTGGCGTTGTGCAGTCGGGCTCTAAGCAATGAGAT TCTGGGCCTGAAGTTGCCTGGCGAGCCGCCGCTGACGGCCAACACCGTGTGCTTGGCGCTGTCCGGCCTGAGC AAGCGGCAGCTAGGCCTGTGCCTGCGCAACCCCGACGTGACGGCGTCCGCGCTTCAGGGTCTGCACATCGCGG TCCACGAGTGTCAGCACCAGCTGCGCGACCAGCGCTGGAACTGCTCCGCGCTTGAGGGCGGCGGCCGCCTGCC σCACCACAGCGCCATCCTCAAGCGGGCCTGGTGTAGGGGAAGGCTTGGACACCCAAATGGTTTCCGAGAAAGT GCTTTTTCCTTCTCCATGCTGGCTGCTGGGGTCATGCACGCAGTAGCCACGGCCTGCAGCCTGGGCAAGCTGG TGAGCTGTGGCTGTGGCTGGAAGGGCAGTGGTGAGCAGGATCGGCTGAGGGCCAAACTGCTGCAGCTGCAGGC ACTGTCCCGAGGCAAGAGTTTCCCCCACTCTCTGCCCAGCCCTGGCCCTGGCTCAAGCCCCAGCCCTGGCCCC CAGGACACATGGGAATGGGGTGGCTGTAACCATGACATGGACTTTGGAGAGAAGTTCTCTCGGGATTTCTTGG ATTCCAGGGAAGCTCCCCGGGACATCCAGGCACGAATGCGAATCCACAACAACAGGGTGGGGCGCCAGGTGGT AACTGAAAACCTGAAGCGGAAATGCAAGTGTCATGGCACATCTGGCAGCTGCCAGTTCAAGACAAATTCTGGA GCCTTCCAGCCCCGTCTGCGTCCCCGTCGCCTCTCAGGAGAGCTGGTCTACTTTGAGAAGTCTCCTGACTTCT GTGAGCGAGACCCCACTATGGGCTCCCCAGGGACAAGGGGCCGGGCCTGCAACAAGACCAGCCGCCTGTTGGA TGGCTGTGGCAGCCTGTGCTGTGGCCGTGGGCACAACGTGCTCCGGCAGACACGAGTTGAGCGCTGCCATTGC CGCTTCCACTGGTGCTGCTATGTGCTGTGTGATGAGTGCAAGGTTACAGAGTGGGTGAATGTGTGTAAGTGAG GGTCAGCCTTACCTTGGGGCTGGGGAAGAGGACTGTGTGAGAGGGGT
NOV14a, CG57054-03 |SEO IDNO: 182 !370 aa IMW at 40782.4kD
Protein Sequence LEEPRPRPPPSGLAGLLFLALCSRALSNEILGLKLPGEPPLTANTVCLALSGLSKRQLGLCLRNPDVTASAL QGLHIAVHECQHQ RDQRWNCS EGGGRLPHHSAILKRA CRGRLGHP GFRESAFSFSMA GV^fflAVATA CSLGKLVSCGCG KGSGEQDRLRAKLLQLQALSRGKSFPHSLPSPGPGSSPSPGPQDT EWGGCNHDMDFGEK FSRDFLDSREAPRDIQARMRIHNNRVGRQWTENLKRKCKCHGTSGSCQFKTNSGAFQPRLRPRRLSGELVYF EKSPDFCERDPTMGSPGTRGRACNKTSRLLDGCGSLCCGRGHNVLRQTRVERCHCRFH CCYVLCDEC VTE VNVCK
Figure imgf000232_0001
NOV14b, CG57054-01 SEQ ID NO: 184 345 aa MW at 38351.4 D Protein Sequence
NTVCLTLSGLSKRQLGLCLRNPDVTASALQGLHIAVHECQHQLRDQR NCSALEGGGRLPHHSAILKRGFRES AFSFSMLAAGVMHAVATACGLG LVSCGCGWKGSGEQDRLRAKLLQLQALSRGKSFPHSLPSPGPGSSPSPGP QDTWE GGCNHDMDFGEKFSRDFLDSREAPRDIQAR RIHNNRVGRQWTENLKRKCKCHGTSGSCQFKTC R AAPEFRAVGAALRERLGRAIFIDTHNRNSGAFQPRLRPRRLSGELVYFEKSPDFCERDPTMGSPGTRGRACNK TSRLLDGCGSLCCGRGHNVLRQTRVERCHCRFHWCCYVLCDECKVTE VNVCK
NOV14c, CG57054-02 SEQ ID NO: 185 J1317bp DNA Sequence ! ORF Start: ATG at 55 }ORF Stop: TGA at 1222
CCAAACCACTGGAGGTCCTGATCGATCTGCCCACCGGAGCCTCCGGGCTTCGACATGCTGGAGGAGCCCCGGC
CGCGGCCTCCGCCCTCGGGCCTCGCGGGTCTCCTGTTCCTGGCGTTGTGCAGTCGGGCTCTAAGCAATGAGAT TCTGGGCCTGAAGTTGCCTGGCGAGCCGCCGCTGACGGCCAACACCGTGTGCTTGACGCTGTCCGGCCTGAGC AAGCGGCAGCTAGGCCTGTGCCTGCGCAACCCCGACGTGACGGCGTCCGCGCTTCAGGGTCTGCACATCGCGG TCCACGAGTGTCAGCACCAGCTGCGCGACCAGCGCTGGAACTGCTCCGCGCTTGAGGGCGGCGGCCGCCTGCC GCACCACAGCGCCATCCTCAAGCGCGGTTTCCGAGAAAGTGCTTTTTCCTTCTCCATGCTGGCTGCTGGGGTC ATGCACGCAGTAGCCACGGCCTGCAGCCTGGGCAAGCTGGTGAGCTGTGGCTGTGGCTGGAAGGGCAGTGGTG AGCAGGATCGGCTGAGGGCCAAACTGCTGCAGCTGCAGGCACTGTCCCGAGGCAAGAGTTTCCCCCACTCTCT GCCCAGCCCTGGCCCTGGCTCAAGCCCCAGCCCTGGCCCCCAGGACACATGGGAATGGGGTGGCTGTAACCAT GACATGGACTTTGGAGAGAAGTTCTCTCGGGATTTCTTGGATTCCAGGGAAGCTCCCCGGGACATCCAGGCAC GAATGCGAATCCACAACAACAGGGTGGGGCGCCAGGTGGTAACTGAAAACCTGAAGCGGAAATGCAAGTGTCA TGGCACATCAGGCAGCTGCCAGTTCAAGACATGCTGGAGGGCGGCCCCAGAGTTCCGGGCAGTGGGGGCGGCG TTGAGGGAGCGGCTGGGCCGGGCCATCTTCATTGATACCCACAACCGCAATTCTGGAGCCTTCCAGCCCCGTC TGCGTCCCCGTCGCCTCTCAGGAGAGCTGGTCTACTTTGAGAAGTCTCCTGACTTCTGTGAGCGAGACCCCAC TATGGGCTCCCCAGGGACAAGGGGCCGGGCCTGCAACAAGACCAGCCGCCTGTTGGATGGCTGTGGCAGCCTG TGCTGTGGCCGTGGGCACAACGTGCTCCGGCAGACACGAGTTGAGCGCTGCCATTGCCGCTTCCACTGGTGCT GCTATGTGCTGTGTGATGAGTGCAAGGTTACAGAGTGGGTGAATGTGTGTAAGTGAGGGTCAGCCTTACCTTG GGGGCTGGGGAAGAGGACTGTGTGAGAGGGGCGCCTTTTCAGCCCTTTGCTCTGATTTCCTTCCAAGGTCACT
CTT
NOV14c, CG57054-02 !SEQ IDNO: 186 J389 aa jMW at 42999.9kD
Protein Sequence j
MLEEPRPRPPPSGLAGLLFLALCSRALSNEILGLKLPGEPPLTANTVCLTLSGLSKRQLGLCLRNPDVTASAL QGLHIAVHECQHQLRDQRWNCSALEGGGRLPHHSAILKRGFRESAFSFSπ^AAGVMHAVATACSLGKLVSCGC GWKGSGEQDRLRAKLLQLQALSRGKSFPHSLPSPGPGSSPSPGPQDT EWGGCNHDMDFGEKFSRDFLDSREA PRDIQARMRIHNNRVGRQWTENLKRKCKCHGTSGSCQFKTC RAAPEFRAVGAALRERLGRAIFIDTHNRNS GAFQPRLRPRRLSGELVYFEKSPDFCERDPTMGSPGTRGRACNKTSRLLDGCGSLCCGRGHNVLRQTRVERCH CRFHWCCYVLCDECKVTEWVNVCK
A ClustalW comparison of the above protein sequences yields the following sequence alignment shown in Table 14B. Table 14B. Comparison of the NOV14 protein sequences.
NOV14a LEEPRPRPPPSGLAGLLFLALCSRALSNEILGLKLPGEPPLTANTVCLALSGLSKRQLG
NOV14b NTVCLTLSGLSKRQLG
NOV14C LEEPRPRPPPSGLAGLLFLALCSRALSNEILGLKLPGEPPLTANTVCLTLSGLSRRQLG
NOV1 a LCLRNPDVTASALQGLHIAVHECQHQLRDQR NCSALEGGGRLPHHSAILKRAWCRGRLG NOVl4b LCLRNPDVTASALQGLHIAVHECQHQLRDQRWNCSALEGGGRLPHHSAILKR G NOV14C LCLRNPDVTASALQGLHIAVHECQHQLRDQRWNCSALEGGGRLPHHSAILKR G
NOVl4a HPNGFRESAFSFSMLAAGVMHAVATACSLGKtiVSCGCGWKGSGEQDRLRAKLLQLQALSR NOV14b FRESAFSFSMLAAGVMHAVATACGLGKLVSCGCGWKGSGEQDRLRAKLLQLQALSR NOVl4c FRESAFSFSMLAAGVMHAVATACSLGKLVSCGCGWKGSGEQDRLRAKLLQLQALSR
NOVl4 GKSFPHSLPSPGPGSSPSPGPQDTWEWGGCNHD DFGΞKFSRDFLDSREAPRDIQARMRI NOV14b GKSFPHSLPSPGPGSSPSPGPQDTWE GGCNHD DFGEKFSRDFLDSREAPRDIQARMRI NOV14C GKSFPHSLPSPGPGSSPSPGPQDTWEWGGCNHD DFGEKFSRDFLDSREAPRDIQARMRI
NOV14a HNNRVGRQWTENLKRKCKCHGTSGSCQFKT NOV14b HNNRVGRQWTΞNLKRKCKCHGTSGSCQF TCWRAAPEFRAVGAALRERLGRAIFIDTHN NOVl4c HNNRVGRQWTENLKR CKCHGTSGSCQFKTC RAAPEFRAVGAALRERLGRAIFIDTHN
NOV14a -NSGAFQPRLRPRRLSGELVYFEKSPDFCERDPTMGSPGTRGRACNKTSRLLDGCGSLCC NOV14b RNSGAFQPRLRPRRLSGELVYFE SPDFCERDPTMGSPGTRGRACN TSRLLDGCGSLCC NOV14C RNSGAFQPRLRPRRLSGELVYFEKSPDFCERDPTMGSPGTRGRACNKTSRLLDGCGSLCC
NOV1 GRGHNVLRQTRVERCHCRFH CCYVLCDECKVTEWVNVCK NOV14b GRGHNVLRQTRVERCHCRFH CCYVLCDECKVTEWVNVCK NOVI4c GRGHNVLRQTRVERCHCRFHWCCYVLCDECKVTE VNVCK
NOVl4a (SEQ ID NO 182) NOV14b (SEQ ID NO 184) NOV14C (SEQ ID NO 186)
Further analysis of the NOV14a protein yielded the following properties shown in Table 14C.
Table 14C. Protem Sequence Properties NOV14a
SignalP analysis: Cleavage site between residues 29 and 30
PSORTII analysis:
PSG: a new signal peptide prediction method
N-region: length 8; pos.chg 2; neg.chg 2 H-region: length 16; peak value 10.20 PSG score: 5.80
GvH: von Heijne's method for signal seq. recognition GvH score (threshold: -2.1): 2.10 possible cleavage site: between 28 and 29
»> Seems to have a cleavable signal peptide (1 to 28)
ALOM: Klein et al's method for TM region allocation Init position for calculation: 29 Tentative number of TMS(s) for the threshold 0.5: 0 number of TMS(s) .. fixed
PERIPHERAL Likelihood = 0.58 (at 134)
ALOM score: 0.58 (number of TMSs: 0)
MTOP: Prediction of membrane topology (Hartmann et al . ) Center position for calculation: 14 Charge difference: -1.0 C( 0.0) - N( 1.0) N >= C: N-terminal side will be inside
MITDISC: discrimination of mitochondrial targeting seq R content: 0 Hyd Moment (75) : 9.19 Hyd Moment (95): 7.93 G content: 0 D/E content: 2 S/T content: 0 Score: -6.41
Gavel : prediction of cleavage sites for mitochondrial preseq cleavage site motif not found
NUCDISC: discrimination of nuclear localization signals pat4: RPRR (4) at 281 pat7: PRLRPRR (3) at 278 bipartite : none content of basic residues: 14.1% NLS Score: 0.04
KDEL: ER retention motif in the C-terminus: none
ER Membrane Retention Signals : none
SKL: peroxisomal targeting signal in the C-terminus: none
PTS2 : 2nd peroxisomal targeting signal : found RLRAKLLQL at 167
VAC: possible vacuolar targeting motif: none
RNA-binding motif: none
Actinin-type actin-binding motif: type 1: none type 2 : none
NMYR: N-myristoylation pattern : none
Prenyl ion motif : none memYQRL: transport motif from cell surface to Golgi: none
Tyrosines in the tail: none
Dileucine motif in the tail: none checking 63 PROSITE DNA binding motifs: none checking 71 PROSITE ribosomal protein motifs: none checking 33 PROSITE prokaryotic DNA binding moti s: none
NNCN: Reinhardt's method for Cytoplasmic/Nuclear discrimination Prediction: nuclear Reliability: 89
COIL: Lupas's algorithm to detect coiled-coil regions total : 0 residues Final Results (k = 9/23) :
65.2 % nuclear
17.4 % mitochondrial
13.0 % extracellular, including cell wall
4.3 % cytoplasmic
» prediction for CG57054-03 is nuc (k=23)
A search of the NOVl 4a protein against the Geneseq database, a proprietary database that contains sequences published in patents and patent publication, yielded several homologous proteins shown in Table 14D.
Figure imgf000236_0001
In a BLAST search of public sequence databases, the NOV14a protem was found to have homology to the proteins shown in the BLASTP data in Table 14E.
Figure imgf000237_0001
PFam analysis predicts that the NOVl 4a protein contains the domains shown in the Table 14F.
Figure imgf000237_0002
Example 15.
The NOVl 5 clone was analyzed, and the nucleotide and encoded polypeptide sequences are shown in Table 15 A.
Figure imgf000238_0001
Figure imgf000238_0002
NOVl 5b, CG57431-02 SEQ ID NO: 190 178 aa MW at l9918.5kD Protein Sequence
MVSVPSTWCSVALALLVALHEGKGQAAATLEQPASSSHAQGTHLRLRRCSCSSWLDKECVYFCHLDIIWVNTP EQTAPYGLGNPPRRRRRSLPRRCQCSSARDPACATFCLRRPWTEAGAVPSRKSPADVFQTGKTGATTGELLQR LRDISTVKSLFAKRQQEAMREPRSTHSR KER
NOV15c, CG57431-01 SEQ ID NO: 191 668 bp DNA Sequence ORF Start: ATG at 40 ORF Stop: TAG at 574
CTCCCTGCTCCAGTCCAGCCTGCGCGCTCCACCGCCGCTATGGTTTCCGTGCCTAGCACCTGGTGCTCCGTTG
CGCTAGCCCTGCTCGTGGCCCTGCATGAAGGGAAGGGCCAGGCTGCTGCCACCCTGGAGCAGCCAGCGTCCTC ATCTCATGCCCAAGGCACCCACCTTCGGCTTCGCCGTTGCTCCTGCAGCTCCTGGCTCGACAAGGAGTGCGTC TACTTCTGCCACTTGGACATCATCTGGGTGAACACTCCTGAACAGACAGCTCCTTACGGCCTGGGAAACCCGC CAAGACGCCGGCGCCGCTCCCTGCCAAGGCGCTGTCAGTGCTCCAGTGCCAGGGACCCCGCCTGTGCCACCTT CTGCCTTCGAAGGCCCTGGACTGAAGCCGGGGCAGTCCCAAGCCGGAAGTCCCCTGCAGACGTGTTCCAGACT GGCAAGACAGGGGCCACTACAGGAGAGCTTCTCCAAAGGCTGAGGGACATTTCCACAGTCAAGAGCCTCTTTG CCAAGCGACAACAGGAGGCCATGCGGGAGCCTCGGTCCACACATTCCAGGTGGAGGAAGAGATAGTGTCGTGA GCTGGAGGAACATTGGGAAGGAAGCCCGCGGGGAGAGAGGAGGAGAGAAGTGGCCAGGGCTTGTGGACTCTCC
TGCTGCTTTCT
NOV15c, CG57431-01 lSEQ ID NO: 192 1178 aa |MW at 19945.6kD
Protein Sequence
MVSVPSTWCSVALALLVALHEGKGQAAATLEQPASSSHAQGTHLRLRRCSCSS LDKECVYFCHLDIIWVNTP EQTAPYGLGNPPRRRRRSLPRRCQCSSARDPACATFCLRRPWTEAGAVPSR SPADVFQTGKTGATTGELLQR LRDISTVKSLFAKRQQEAMREPRSTHSRWRKR
NOV15d, CG57431-04 SEQ ID NO: 194 104 aa MW at ll793.4 D Protein Sequence
MVSVPTT CSVALALLVALHEGKGQAAATLEQPASSSHAQGTHLRLRRCSCSS LDKECVYFCHLDIIWVNTP EDISTVKSLFAKRQQEAMREPRSTHSR RKR
A ClustalW comparison of the above protein sequences yields the following sequence alignment shown in Table 15B. Table 15B. Comparison of the NOV15 protein sequences.
NOV15a MVSVPTT CSVALALLVALHEGKGQAAATLEQPASSSHAQGTHLRLRRCSCSSWLDKECV
NOV15b MVSVPST CSVALALLVALHEGKGQAAATLEQPASSSHAQGTHLRLRRCSCSSWLDKECV
NOV15C MVSVPSTWCSVALALLVALHEGKGQAAATLEQPASSSHAQGTHLRLRRCSCSS LDKECV
NOV15d MVSVPTTWCSVALALLVALHEGKGQAAATLEQPASSSHAQGTHLRLRRCSCSS LDKECV
NOV15a YFCHLDII VNTPEQTAPYGLGNPPRRRRR
NOV15b YFCHLDIIWVNTPEQTAPYGLGNPPRRRRRSLPRRCQCSSARDPACATFCLRRPWTEAGA
NOV15c YFCHLDII VNTPEQTAPYGLGNPPRRRRRSLPRRCQCSSARDPACATFCLRRPWTEAGA
NOV15d YFCHLDIIWVNT
NOV15a SLPRRCQCSSARDPTCATFCLRRPWDISTVKSLFAKRQQEAMREPRSTHSR RKR
NOV15b VPSRKSPADVFQTGKTGATTGELLQRLRDISTVKSLFAKRQQEAMREPRSTHSR KER
NOV15C VPSRKSPADVFQTGKTGATTGELLQRLRDISTV SLFAKRQQEAMREPRSTHSR RKR
NOV15d ' PEDISTVKSLFAKRQQEAMREPRSTHSRWRKR
NOV15a (SEQ ID NO 188)
NOV15b (SEQ ID NO 190)
NOVl5C (SEQ ID NO 192)
NOV15d (SEQ ID NO 194)
Further analysis of the NOVl 5a protein yielded the following properties shown in Table 15C.
Table 15C. Protein Sequence Properties NOVl 5a
SignalP analysis: Cleavage site between residues 25 and 26
PSORTII analysis:
PSG: a new signal peptide prediction method
N-region: length 0; pos.chg 0; neg.chg 0 H-region: length 20; peak value 9.73 PSG score: 5.33
GvH: von Heijne's method for signal seq. recognition GvH score (threshold: -2.1): -1.24 possible cleavage site: between 24 and 25
»> Seems to have a cleavable signal peptide (1 to 24)
ALOM: Klein et al ' s method for TM region allocation Init position for calculation: 25 Tentative number of TMS(s) for the threshold 0.5: number of TMS(s) .. fixed PERIPHERAL Likelihood = 7.85 (at 54) ALOM score: 7.85 (number of T Ss: 0)
MTOP: Prediction of membrane topology (Hartmann et al.) Center position for calculation: 12 Charge difference: -1.5 C(-0.5) - N( 1.0) N >= C: N-terminal side will be inside
MITDISC: discrimination of mitochondrial targeting seq R content: 0 Hyd Moment (75): 1.71 Hyd Moment (95): 1.49 G content: 0 D/E content: 1 S/T content: 4 Score : -4.94
Gavel : prediction of cleavage sites for mitochondrial preseq cleavage site motif not found
NUCDISC: discrimination of nuclear localization signals pat4: PRRR (4) at 85 pat4: RRRR (5) at 86 pat4: RRRR (5) at 87 pat7: PPRRRRR (5) at 84 pat7: PRRRRRS (5) at 85 bipartite: none content of basic residues: 16.6% NLS Score: 1.27
KDEL: ER retention motif in the C-terminus: none
ER Membrane Retention Signals:
KKXX-like motif in the C-terminus: R RK
SKL: peroxisomal targeting signal in the C-terminus: none
PTS2 : 2nd peroxisomal targeting signal: none
VAC: possible vacuolar targeting motif: none
RNA-binding motif: none
Actinin-type actin-binding motif: type 1 : none type 2 : none
NMYR: N-myristoylation pattern : none
Prenylat on motif: none memYQRL: transport motif from cell surface to Golgi: none
Tyrosines in the tail : none
Dileucine motif in the tail .- none checking 63 PROSITE DNA binding motifs: none checking 71 PROSITE ribosomal protein motifs: none checking 33 PROSITE prokaryotic DNA binding motifs: none
NNCN: Reinhardt's method for Cytoplasmic/Nuclear discrimination Prediction: nuclear Reliability: 94.1
COIL: Lupas's algorithm to detect coiled-coil regions total : 0 residues
Final Results (k = 9/23):
55.6 %: extracellular, including cell wall 33.3 %: nuclear 11.1 %: cytoplasmic
>> prediction for CG57431-03 is exc (k=9) A search of the NOVl 5a protein against the Geneseq database, a proprietary database that contains sequences published in patents and patent publication, yielded several homologous proteins shown in Table 15D.
Figure imgf000242_0001
In a BLAST search of public sequence databases, the NOVl 5a protein was found to have homology to the proteins shown in the BLASTP data in Table 15E.
Figure imgf000243_0001
PFam analysis predicts that the NOVl 5a protein contains the domains shown in the Table 15F.
Figure imgf000243_0002
Example 16.
The NOVl 6 clone was analyzed, and the nucleotide and encoded polypeptide sequences are shown in Table 16A. Table 16A. NOV16 Sequence Analysis
NOV16a, CG59253-01 SEQ ID NO: 195 1894bp DNA Sequence ORF Start: ATG at 46 ORF Stop: TAG at 1474
TGGCATTTCTGAGCAGGGGCCACCCTGACTTCACCTTGGCCCACCATGAGGGTCTTCCTGCTTTGTGCCTACA
TACTGCTGCTGATGGTTTCCCAGTTGAGGGCAGTCAGCTTTCCTGAAGATGATGAACCCCTTAATACTGTCGA CTATCACTATTCAAGGCAATATCCGGTTTTTAGAGGACGCCCTTCAGGCAATGAATCGCAGCACAGGCTGGAC TTTCAGCTGATGTTGAAAATTCGAGACACACTTTATATTGCTGGCAGGGATCAAGTTTATACAGTAAACTTAA ATGAAATGCCCAAAACAGAAGTAATACCCAACAAGAAACTGACATGGCGATCAAGACAACAGGATCGAGAAAA CTGTGCTATGAAAGGCAAGCATAAAGATGAATGCCACAACTTTATCAAAGTATTTGTTCCAAGAAACGATGAG ATGGTTTTTGTTTGTGGTACCAATGCATTCAATCCCATGTGTAGATACTACAGGTTGAGTACCTTAGAATATG ATGGGGAAGAAATTAGTGGCCTGGCAAGATGCCCATTTGATGCCAGACAAACCAATGTTGCCCTCTTTGCTGA TGGGAAGCTGTATTCTGCCACAGTGGCTGACTTCTTGGCCAGCGATGCCGTTATTTATCGAAGCATGGGTGAT GGATCTGCCCTTCGCACAATAAAATATGATTCCAAATGGATAAAAGAGCCACACTTTCTTCATGCCATAGAAT ATGGAAACTATGTCTATTTCTTCTTTCGAGAAATCGCTGTCGAACATAATAATTTAGGCAAGGCTGTGTATTC CCGCGTGGCCCGCATATGTAAAAACGACATGGGTGGTTCCCAGCGGGTCCTGGAGAAACACTGGACTTCATTT CTAAAGGCTCGGCTGAACTGTTCTGTCCCTGGAGATTCGTTTTTCTACTTTGATGTTCTGCAGTCTATTACAG ACATAATACAAATCAATGGCATCCCCACTGTGGTCGGGGTGTTTACCACGCAGCTCAATAGCATCCCTGGTTC TGCTGTCTGTGCATTTAGCATGGATGACATTGAAAAAGTATTCAAAGGACGGTTTAAGGAACAGAAAACTCCA GATTCTGTTTGGACAGCAGTTCCCGAAGACAAAGTGCCAAAGCCAAGGCCTGGCTGTTGTGCAAAACACGGCC TTGCCGAAGCTTATAAAACCTCCATCGATTTCCCGGATGAAACTCTGTCATTCATCAAATCTCATCCCCTGAT GGACTCTGCCGTTCCACCCATTGCCGATGAGCCCTGGTTCACAAAGACTCGGGTCAGGTACAGACTGACGGCC ATCTCAGTGGACCATTCAGCCGGACCCTACCAGAACTACACAGTCATCTTTGTTGGCTCTGAAGCTGGCATGG TACTTAAAGTTCTGGCAAAGACCAGTCCTTTCTCTTTGAACGACAGCGTATTACTGGAAGAGATTGAAGCCTA CAACCATGCAAAGTAGGTATATGTTACGAGAACGCCCTTCAGCACTGCTCAAAAATTTTCGGCATGTATTTCA TCTAGTCATGTCCTTTTGGTCCTCTAAATTAGCAGTGGTTTGGCATAATAGTGTTTTGTGTTTTTTTTCTCAT
TGAAATAAATCTTGGGTTTGTTTTTTTCCCGAGCCTGCTAGGGCGAGGGGGGTGAATGGTTGATGAGTTTAAA lAATAATGCAGCCCTTGTTTTTCACCTGTAGAATATGAGAACATTTTAACAGCACCTCTCTTATCTTGCAGATA
TATTCCAAGATGCTACATGCAGCAGACAGCTGTGAGCTTGCATACACACACACACAAATATACATGCACATAC jATACACAGAATGTAGTACTAGTTAAGTATTTCCTTCCTATCTTTAATAAGTAAGAGAATATTTAGACCA
NOV16a, CG59253-01 SEQ ID NO: 196 476 aa MW at 54216.4kD Protein Sequence
MRVFLLCAYILLLMVSQLRAVSFPEDDEPLNTVDYHYSRQYPVFRGRPSGNESQHRLDFQLMLKIRDTLYIAG RDQVYTVNLNEMPKTEVIPNKKLTWRSRQQDRENCAM GKHKDECHNFIKVFVPRNDEMVFVCGTNAFNP CR YYRLSTLEYDGEEISGLARCPFDARQTNVALFADGKLYSATVADFLASDAVIYRSMGDGSALRTIKYDSKWIK EPHFLHAIEYGNYVYFFFREIAVEHNNLGKAVYSRVARICKND GGSQRVLEKHWTSFLKARLNCSVPGDSFF YFDVLQSITDIIQINGIPTWGVFTTQLNSIPGSAVCAFSMDDIEKVFKGRFKEQKTPDSVTAVPED VPKP RPGCCAKHGLAEAYKTSIDFPDETLSFIKSHPLMDSAVPPIADEP FTKTRVRYRLTAISVDHSAGPYQNYTV IFVGSEAGMVLKVLAKTSPFSLNDSVLLEEIEAYNHAK
NOV16b, 194877881 SEQ ID NO: 197 1383 bp DNA Sequence ORF Start: at 1 ORF Stop: end of sequence
GGATCCGTCAGCTTTCCTGAAGATGATGAACCCCTTAATACTGTCGACTATCACTATTCAAGGCAATATCCGG TTTTTAGAGGACGCCCTTCAGGCAATGAATCGCAGCACAGGCTGGACTTTCAGCTGATGTTGAAAATTCGAGA CACACTTTATATTGCTGGCAGGGATCAAGTTTATACAGTAAACTTAAATGAAATGCCCAAAACAGAAGTAATA CCCAACAAGAAACTGACATGGCGATCAAGACAACAGGATCGAGAAAACTGTGCTATGAAAGGCAAGCATAAAG ATGAATGCCACAACTTTATCAAAGTATTTGTTCCAAGAAACGATGAGATGGTTTTTGTTTGTGGTACCAATGC ATTCAATCCCATGTGTAGATACTACAGGTTGAGTACCTTAGAATATGATGGGGAAGAAATTAGTGGCCTGGCA AGATGCCCATTTGATGCCAGACAAACCAATGTTGCCCTCTTTGCTGATGGGAAGCTGTATTCTGCCACAGTGG CTGACTTCTTGGCCAGCGATGCCGTTATTTATCGAAGCATGGGTGATGGATCTGCCCTTCGCACAATAAAATA TGATTCCAAATGGATAAAAGAGCCACACTTTCTTCATGCCATAGAATATGGAAACTATGTCTATTTCTTCTTT CGAGAAATCGCTGTCGAACATAATAATTTAGGCAAGGCTGTGTATTCCCGCGTGGCCCGCATATGTAAAAACG ACATGGGTGGTTCCCAGCGGGTCCTGGAGAAACACTGGACTTCATTTCTAAAGGCTCGGCTGAACTGTTCTGT CCCTGGAGATTCGTTTTTCTACTTTGATGTTCTGCAGTCTATTACAGACATAATACAAATCAATGGCATCCCC ACTGTGGTCGGGGTGTTTACCACGCAGCTCAATAGCATCCCTGGTTCTGCTGTCTGTGCATTTAGCATGGATG ACATTGAAAAAGTATTCAAAGGACGGTTTAAGGAACAGAAAACTCCAGATTCTGTTTGGACAGCAGTTCCCGA AGACAAAGTGCCAAAGCCAAGGCCTGGCTGTTGTGCAAAACACGGCCTTGCCGAAGCTTATAAAACCTCCATC GATTTCCCGGATGAAACTCTGTCATTCATCAAATCTCATCCCCTGATGGACTCTGCCGTTCCACCCATTGCCG ATGAGCCCTGGTTCACAAAGACTCGGGTCAGGTACAGACTGACGGCCATCTCAGTGGACCATTCAGCCGGACC CTACCAGAACTACACAGTCATCTTTGTTGGCTCTGAAGCTGGCATGGTACTTAAAGTTCTGGCAAAGACCAGT CCTTTCTCTTTGAACGACAGCGTATTACTGGAAGAGATTGAAGCCTACAACCATGCAAAGTGCCTCGAG
NOVl 6b, 194877881 SEQ ID NO: 198 461 aa MW at 52370.0kD Protein Sequence
GSVSFPEDDEPLNTVDYHYSRQYPVFRGRPSGNESQHRLDFQLMLKIRDTLYIAGRDQVYTVNLNEMPKTEVI PNKKLTWRSRQQDRENCAMKGKH DECHNFIKVFVPRNDEMVFVCGTNAFNPMCRYYRLSTLEYDGEEISGLA RCPFDARQTNVALFADGKLYSATVADFLASDAVIYRSMGDGSALRTIKYDS WIKEPHFLHAIEYGNYVYFFF REIAVEHNNLGKAVYSRVARICKNDMGGSQRVLEKH TSFLKARLNCSVPGDSFFYFDVLQSITDIIQINGIP TWGVFTTQLNSIPGSAVCAFSMDDIEKVFKGRFKEQKTPDSV TAVPEDKVPKPRPGCCAKHGLAEAYKTSI DFPDETLSFIKSHPLMDSAVPPIADEPWFTKTRVRYRLTAISVDHSAGPYQNYTVIFVGSEAGMVLKVLAKTS PFSLNDSVLLEEIEAYNHAKCLE
NOV16c, CG59253-02 1SEQ ID NO: 199 J3205 bp
DNA Sequence
TGGCATTTCTGAGCAGGGGCCACCCTGACTTCACCTTGGCCCACCATGAGGGTCTTCCTGCTTTGTGCCTACA
TACTGCTGCTGATGGTTTCCCAGTTGAGGGCAGTCAGCTTTCCTGAAGATGATGAACCCCTTAATACTGTCGA CTATCACTGTAAGTCGTCTAGGCAATATCCGGTTTTTAGAGGACGCCCTTCAGGCAATGAATCGCAGCACAGG CTGGACTTTCAGCTGATGTTGAAAATTCGAGACACACTTTATATTGCTGGCAGGGATCAAGTTTATACAGTAA ACTTAAATGAAATGCCCAAAACAGAAGTAATATGGCAACAGAAACTGACATGGCGATCAAGACAACAGGATCG AGAAAACTGTGCTATGAAAGGCAAGCATAAAGATGAATGCCACAACTTTATCAAAGTATTTGTTCCAAGAAAC GATGAGATGGTTTTTGTTTGTGGTACCAATGCATTCAATCCCATGTGTAGATACTACAGGGTAAGTACCTTAG AATATGATGGGGAAGAAATTAGTGGCCTGGCAAGATGCCCATTTGATGCCAGACAAACCAATGTTGCCCTCTT TGCTGATGGGAAGCTGTATTCTGCCACAGTGGCTGACTTCTTGGCCAGCGATGCCGTTATTTATCGAAGCATG GGTGATGGATCTGCCCTTCGCACAATAAAATATGATTCCAAATGGATAAAAGAGCCACACTTTCTTCATGCCA TAGAATATGGAAACTATGTCTATTTCTTCTTTCGAGAAATCGCTGTCGAACATAATAATTTAGGCAAGGCTGT GTATTCCCGCGTGGCCCGCATATGTAAAAACGACATGGGTGGTTCCCAGCGGGTCCTGGAGAAACACTGGACT TCATTTCTAAAGGCTCGGCTGAACTGTTCTGTCCCTGGAGATTCGTTTTTCTACTTTGATGTTCTGCAGTCTA TTACAGACATAATACAAATCAATGGCATCCCCACTGTGGTCGGGGTGTTTACCACGCAGCTCAATAGCATCCC TGGTTCTGCTGTCTGTGCATTTAGCATGGATGACATTGAAAAAGTATTCAAAGGACGGTTTAAGGAACAGAAA ACTCCAGATTCTGTTTGGACAGCAGTTCCCGAAGACAAAGTGCCAAAGCCAAGGCCTGGCTGTTGTGCAAAAC ACGGCCTTGCCGAAGCTTATAAAACCTCCATCGATTTCCCGGATGAAACTCTGTCATTCATCAAATCTCATCC CCTGATGGACTCTGCCGTTCCACCCATTGCCGATGAGCCCTGGTTCACAAAGACTCGGGTCAGGTACAGACTG ACGGCCATCTCAGTGGACCATTCAGCCGGACCCTACCAGAACTACACAGTCATCTTTGTTGGCTCTGAAGCTG GCATGGTACTTAAAGTTCTGGCAAAGACCAGTCCTTTCTCTTTGAACGACAGCGTATTACTGGAAGAGATTGA AGCCTACAACCATGCAAAGTGCAGTGCTGAGAATGAGGAAGACAAAAAGGTCATCTCATTACAGTTGGATAAA GATCACCACGCTTTATATGTGGCGTTCTCTAGCTGCATTATCCGCATCCCCCTCAGTCGCTGTGAGCGTTATG GATCATGTAAAAAGTCTTGTATTGCATCTCGTGACCCGTATTGTGGCTGGTTAAGCCAGGGATCCTGTGGTAG AGTGACCCCAAACCACAGTGCTGAAGGATATGAACAAGACACAGAATTCGGCAACACAGCTCATCTAGGGGAC TGCCATGCATATGAACCATATGAAGGTCGTGTTGGCTCACTGAAAGCCATTTGCTATTTATTATTATTTTTAA AAAGCACCTTATTCACATTGTCCCATGTGTCTATTTCAGGTGTACGATGGGAAGTCCAGTCTGGAGAGTCCAA CCAGATGGTCCACATGAATGTCCTCATCACCTGTGTCTTTGCTGCTTTTGTTTTGGGGGCATTCATTGCAGGT GTGGCAGTATACTGCTATCGAGACATGTTTGTTCGGAAAAACAGAAAGATCCATAAAGATGCAGAGTCCGCCC AGTCATGCACAGACTCCAGTGGAAGTTTTGCCAAACTGAATGGTCTCTTTGACAGCCCTGTCAAGGAATACCA ACAGAATATTGATTCTCCTAAACTGTATAGTAACCTGCTAACCAGTCGGAAAGAGCTACCACCCAATGGAGAT ACTAAATCCATGGTAATGGACCATCGAGGGCAACCTCCAGAGTTGGCTGCTCTTCCTACTCCTGAGTCTACAC CCGTGCTTCACCAGAAGACCCTGCAGGCCATGAAGAGCCACTCAGAAAAGGCCCATGGCCATGGAGCTTCAAG GAAAGAAACCCCTCAGTTTTTTCCGTCTAGTCCGCCACCTCATTCCCCATTAAGTCATGGGCATATCCCCAGT GCCATTGTTCTTCCAAATGCTACCCATGACTACAACACGTCTTTCTCAAACTCCAATGCTCACAAAGCTGAAA AGAAGCTTCAAAACATTGATCACCCTCTCACAAAGTCATCCAGTAAGAGAGATCACCGGCGTTCTGTTGATTC CAGAAATACCCTCAATGATCTCCTGAAGCATCTGAATGACCCAAATAGTAACCCCAAAGCCATCATGGGAGAC ATCCAGATGGCACACCAGAACTTAATGCTGGATCCCATGGGATCGATGTCTGAGGTCCCACCTAAAGTCCCTA ACCGGGAGGCATCGCTATACTCCCCTCCTTCAACTCTCCCCAGAAATAGCCCAACCAAGCGAGTGGATGTCCC CACCACTCCTGGAGTCCCAATGACTTCTCTGGAAAGACAAAGAGGTTATCACAAAAATTCCTCCCAGAGGCAC TCTATATCTGCTATGCCTAAAAACTTAAACTCACCAAATGGTGTTTTGTTATCCAGACAGCCTAGTATGAACC GTGGAGGATATATGCCCACCCCCACTGGGGCGAAGGTGGACTATATTCAGGGAACACCAGTGAGTGTTCATCT GCAGCCTTCCCTCTCCAGACAGAGCAGCTACACCAGTAATGGCACTCTTCCTAGGACGGGACTAAAGAGGACG CCGTCCTTAAAACCTGACGTGCCACCAAAGCCTTCCTTTGTTCCTCAAACCCCATCTGTCAGACCACTGAACA AATACACATACTAGGCCTCAAGTGTGCTATTCCCATGTGGCTTTATCCTGTCCGTGTTGTTGAGAG
Figure imgf000247_0001
NOV16d, 191815765 SEQ ID NO: 202 571 aa MW at 64535.4kD Protein Sequence
GSVSFPEDDEPLNTTOYHYSRQYPVFRGRPSGNESQHRLDFQ
PNKKLTWRSRQQDRENCAMKGKHKOECHNFIKVFVPRNDEMVFVCGTNAFNP CRYYRLSTLEYDGEEISGLA
RCPFDARQTNVALFADGKLYSATVADFLASDAVIYRSMGDGSALRTIKYDSKWIKEPHFLHAIEYGNYVYFFF
REIAVEHNNLGKAVYSRVARICKNDMGGSQRVLEKH TSFLKARLNCSVPGDSFFYFDVLQSITDIIQINGIP
TWGVFTTQLNSIPGSAVCAFSMDDIEKVFKGRFKEQKTPDSVWTAVPEDKVPKPRPGCCAKHGLAEAYKTSI
DFPDETLSFI SHPLMDSAVPPIADEPWFTKTRVRYRLTAISVDHSAGPYQNYTVIFVGSEAGMVL VLAKTS
PFSLNDSVLLEEIEAYNHAKCNAENEEDKKVISLQLDKDHHALYVAFSSCIIRIPLSRCERYGSCK SCIASR
DPYCGWLSQGSCGRVTPGMLAEGYEQDTEFGNTAHLGDCHGVRWEVQSGESNQ VH NLE
Figure imgf000248_0001
|NOV16e, CG59253-03 SEQ ID NO: 204 456 aa MW at 51880.5kD
Protein Sequence
VSFPEDDEPLNTVDYHYSRQYPVFRGRPSGNESQHRLDFQLMLKIRDTLYIAGRDQVYTVNLNEMPKTEVIPN KKLTWRSRQQDRENCAMKGKHKDECHNFIKVFVPRNDEMVFVCGTNAFNPMCRYYRLSTLEYDGEEISGLARC PFDARQTNVALFADGKLYSATVADFLASDAVIYRSMGDGSALRTIKYDS IKEPHFLHAIEYGNYVYFFFRE IAVEHNNLGKAVYSRVARIC ND GGSQRVLEKHWTSFLKARLNCSVPGDSFFYFDVLQSITDIIQINGIPTV VGVFTTQLNSIPGSAVCAFSMDDIEKVFKGRFKEQKTPDSV TAVPEDKVPKPRPGCCAKHGLAEAYKTSIDF PDETLSFIKSHPLMDSAVPPIADEPWFTKTRVRYRLTAISVDHSAGPYQNYTVIFVGSEAGMVLKVLAKTSPF SLNDSVLLEEIEAYNHAK NOVlδf, CG59253-04 SEQ ID NO: 205 "Hτ713 bp DNA Sequence
GGATCCGTCAGCTTTCCTGAAGATGATGAACCCCTTAATACTGTCGACTATCACTATTCAAGGCAATATCCGG
TTTTTAGAGGACGCCCTTCAGGCAATGAATCGCAGCACAGGCTGGACTTTCAGCTGATGTTGAAAATTCGAGA CACACTTTATATTGCTGGCGGGGATCAAGTTTATACAGTAAACTTAAATGAAATGCCCAAAACAGAAGTAATA CCCAACAAGAAACTGACATGGCGATCAAGACAACAGGATCGAGAAAACTGTGCTATGAAAGGCAAGCATAAAG ATGAATGCCACAACTTTATCAAAGTATTTGTTCCAAGAAACGATGAGATGGTTTTTGTTTGTGGTACCAATGC ATTCAATCCCATGTGTAGATACTACAGGTTGAGTACCTTAGAATATGATGGGGAAGAAATTAGTGGCCTGGCA AGATGCCCATTTGATGCCAGACAAACCAATGTTGCCCTCTTTGCTGATGGGAAGCTGTATTCTGCCACAGTGG CTGACTTCTTGGCCAGCGATGCCGTTATTTATCGAAGCATGGGTGATGGATCTGCCCTTCGCACAATAAAATA TGATTCCAAATGGATAAAAGAGCCACACTTTCTTCATGCCATAGAATATGGAAACTATGTCTATTTCTTCTTT CGAGAAATCGCTGTCGAACATAATAATTTAGGCAAGGCTGTGTATTCCCGCGTGGCCCGCATATGTAAAAACG ACATGGGTGGTTCCCAGCGGGTCCTGGAGAAACACTGGACTTCATTTCTAAAGGCTCGGCTGAACTGTTCTGT CCCTGGAGATTCGTTTTTCTACTTTGATGTTCTGCAGTCTATTACAGACATAATACAAATCAATGGCATCCCC ACTGTGGTCGGGGTGTTTACCACGCAGCTCAATAGCATCCCTGGTTCTGCTGTCTGTGCATTTAGCATGGATG ACATTGAAAAAGTATTCAAAGGACGGTTTAAGGAACAGAAAACTCCAGATTCTGTTTGGACAGCAGTTCCCGA AGACAAAGTGCCAAAGCCAAGGCCTGGCTGTTGTGCAAAACACGGCCTTGCCGAAGCTTATAAAACCTCCATC GATTTCCCGGATGAAACTCTGTCGTTCATCAAATCTCATCCCCTGATGGACTCTGCCGTTCCACCCATTGCCG ATGAGCCCTGGTTCACAAAGACTCGGGTCAGGTACAGACTGACGGCCATCTCAGTGGACCATTCAGCCGGACC CTACCAGAACTACACAGTCATCTTTGTTGGCTCTGAAGCTGGCATGGTACTTAAAGTTCTGGCAAAGACCAGT CCTTTCTCTTTGAACGACAGCGTATTACTGGAAGAGATTGAAGCCTACAACCATGCAAAGTGCAATGCTGAGA ATGAGGAAGACAAAAAGGTCATCTCATTACAGTTGGATAAAGATCACCACGCTTTATATGTGGCGTTCTCTAG CTGCATTATCCGCATCCCCCTCAGTCGCTGTGAGCGTTATGGATCATGTAAAAAGTCTTGTATTGCATCTCGT GACCCGTATTGTGGCTGGTTAAGCCAGGGATCCTGTGGTAGAGTGACCCCAGGGATGCTTGCTGAAGGATATG AACAAGACACAGAATTCGGCAACACAGCTCATCTAGGGGACTGCCATGGTGTACGATGGGAAGTCCAGTCTGG AGAGTCCAACCAGATGGTCCACATGAATCTCGAG
NOV16f, CG59253-04 SEQ ID NO: 206 567 aa MW at 64149. lkD Protein Sequence
VSFPEDDEPLNTVDYHYSRQYPVFRGRPSGNESQHRLDFQLMLKIRDTLYIAGGDQVYTVNLNEMPKTEVIPN KKLTWRSRQQDRENCAMKGKHKDECHNFIKVFVPRNDEMVFVCGTNAFNPMCRYYRLSTLEYDGEEISGLARC PFDARQTNVALFADGKLYSATVADFLASDAVIYRS GDGSALRTIKYDSKWIKEPHFLHAIEYGNYVYFFFRE IAVEHNNLGKAVYSRVARICKNDMGGSQRVLEKH TSFLKARLNCSVPGDSFFYFDVLQSITDIIQINGIPTV VGVFTTQLNSIPGSAVCAFSMDDIEKVFKGRFKEQKTPDSV TAVPEDKVP PRPGCCAKHGLAEAYKTSIDF PDETLSFIKSHPLMDSAVPPIADEPWFT TRVRYRLTAISVDHSAGPYQNYTVIFVGSEAGMVLKVLAKTSPF SLNDSVLLEEIEAYNHAKCNAENEEDKKVISLQLDKDHHALYVAFSSCIIRIPLSRCERYGSCKKSCIASRDP YCGWLSQGSCGRVTPGMLAEGYEQDTEFGNTAHLGDCHGVR EVQSGESNQMVHMN
Figure imgf000249_0001
TTACAGACATAATACAAATCAATGGCATCCCCACTGTGGTCGGGGTGTTTACCACGCAGCTCAATAGCATCCC TGGTTCTGCTGTCTGTGCATTTAGCATGGATGACATTGAAAAAGTATTCAAAGGACGGTTTAAGGAACAGAAA ACTCCAGATTCTGTTTGGACAGCAGTTCCCGAAGACAAAGTGCCAAAGCCAAGGCCTGGCTGTTGTGCAAAAC ACGGCCTTGCCGAAGCTTATAAAACCTCCATCGATTTCCCGGATGAAACTCTGTCATTCATCAAATCTCATCC CCTGATGGACTCTGCCGTTCCACCCATTGCCGATGAGCCCTGGTTCACAAAGACTCGGGTCAGGTACAGACTG ACGGCCATCTCAGTGGACCATTCAGCCGGACCCTACCAGAACTACACAGTCATCTTTGTTGGCTCTGAAGCTG GCATGGTACTTAAAGTTCTGGCAAAGACCAGTCCTTTCTCTTTGAACGACAGCGTATTACTGGAAGAGATTGA AGCCTACAACCATGCAAAGTGCAGTGCTGAGAATGAGGAAGACAAAAAGGTCATCTCATTACAGTTGGATAAA GATCACCACGCTTTATATGTGGCGTTCTCTAGCTGCATTATCCGCATCCCCCTCAGTCGCTGTGAGCGTTATG GATCATGTAAAAAGTCTTGTATTGCATCTCGTGACCCGTATTGTGGCTGGTTAAGCCAGGGATCCTGTGGTAG AGTGACCCCAGGGATGCTGCTGTTAACCGAAGACTTCTTTGCTTTCCATAACCACAGTGCTGAAGGATATGAA CAAGACACAGAATTCGGCAACACAGCTCATCTAGGGGACTGCCATGAAATTTTGCCTACTTCAACTACACCAG ATTACAAAATATTTGGCGGTCCAACATCTGGTGTACGATGGGAAGTCCAGTCTGGAGAGTCCAACCAGATGGT CCACATGAATGTCCTCATCACCTGTGTCTTTGCTGCTTTTGTTTTGGGGGCATTCATTGCAGGTGTGGCAGTA TACTGCTATCGAGACATGTTTGTTCGGAAAAACAGAAAGATCCATAAAGATGCAGAGTCCGCCCAGTCATGCA CAGACTCCAGTGGAAGTTTTGCCAAACTGAATGGTCTCTTTGACAGCCCTGTCAAGGAATACCAACAGAATAT TGATTCTCCTAAACTGTATAGTAACCTGCTAACCAGTCGGAAAGAGCACGAATTCAGCGGCCGCTGAATTCTA G
NOV16g, CG59253-05 jSEQ ID NO: 208 |712 aa MW at 80536.8kD
Protein Sequence
MRVFLLCAYILLLMVSQLRAVSFPEDDEPLNTVDYHC SSRQYPVFRGRPSGNESQHRLDFQLMLKIRDTLYI AGRDQVYTVNLNEMPKTEVI QQKLT RSRQQDRENCAMKGKHKDECHNFIKVFVPRNDEMVFVCGTNAFNPM CRYYRVSTLEYDGEEISGLARCPFDARQTNVALFADGKLYSATVADFLASDAVIYRSMGDGSALRTI YDSKW IKEPHFLHAIEYGNYVYFFFREIAVEHNNLGKAVYSRVARICKNDMGGSQRVLE HWTSFLKARLNCSVPGDS FFYFDVLQSITDIIQINGIPTWGVFTTQLNSIPGSAVCAFSMDDIEKVFKGRFKEQKTPDSVWTAVPEDKVP KPRPGCCAKHGLAEAY TSIDFPDETLSFIKSHPL DSAVPPIADEPWFTKTRVRYRLTAISVDHSAGPYQNY TVIFVGSEAGMVLKVLAKTSPFSLNDSVLLEEIEAYNHAKCSAENEEDKKVISLQLDKDHHALYVAFSSCIIR IPLSRCERYGSCKKSCIASRDPYCGWLSQGSCGRVTPGMLLLTEDFFAFHNHSAEGYEQDTEFGNTAHLGDCH EILPTSTTPDYKIFGGPTSGVR EVQSGESNQMVHM LITCVFAAFVLGAFIAGVAVYCYRDMFVRKNRKIH KDAESAQSCTDSSGSFAKLNGLFDSPVKEYQQNIDSPKLYSNLLTSRKEHEFSGR
NOVlόh, CG59253-06 SEQ ID NO: 209~ 3196 bp
DNA Sequence |ORF Start: ATG at 46 ORF Stop: TAG at 3142
TGGCATTTCTGAGCAGGGGCCACCCTGACTTCACCTTGGCCCACCATGAGGGTCTTCCTGCTTTGTGCCTACA
TACTGCTGCTGATGGTTTCCCAGTTGAGGGCAGTCAGCTTTCCTGAAGATGATGAACCCCTTAATACTGTCGA CTATCACTGTAAGTCGTCTAGGCAATATCCGGTTTTTAGAGGACGCCCTTCAGGCAATGAATCGCAGCACAGG CTGGACTTTCAGCTGATGTTGAAAATTCGAGACACACTTTATATTGCTGGCAGGGATCAAGTTTATACAGTAA ACTTAAATGAAATGCCCAAAACAGAAGTAATATGGCAACAGAAACTGACATGGCGATCAAGACAACAGGATCG AGAAAACTGTGCTATGAAAGGCAAGCATAAAGATGAATGCCACAACTTTATCAAAGTATTTGTTCCAAGAAAC GATGAGATGGTTTTTGTTTGTGGTACCAATGCATTCAATCCCATGTGTAGATACTACAGGGTAAGTACCTTAG AATATGATGGGGAAGAAATTAGTGGCCTGGCAAGATGCCCATTTGATGCCAGACAAACCAATGTTGCCCTCTT TGCTGATGGGAAGCTGTATTCTGCCACAGTGGCTGACTTCTTGGCCAGCGATGCCGTTATTTATCGAAGCATG GGTGATGGATCTGCCCTTCGCACAATAAAATATGATTCCAAATGGATAAAAGAGCCACACTTTCTTCATGCCA TAGAATATGGAAACTATGTCTATTTCTTCTTTCGAGAAATCGCTGTCGAACATAATAATTTAGGCAAGGCTGT GTATTCCCGCGTGGCCCGCATATGTAAAAACGACATGGGTGGTTCCCAGCGGGTCCTGGAGAAACACTGGACT TCATTTCTAAAGGCTCGGCTGAACTGTTCTGTCCCTGGAGATTCGTTTTTCTACTTTGATGTTCTGCAGTCTA TTACAGACATAATACAAATCAATGGCATCCCCACTGTGGTCGGGGTGTTTACCACGCAGCTCAATAGCATCCC TGGTTCTGCTGTCTGTGCATTTAGCATGGATGACATTGAAAAAGTATTCAAAGGACGGTTTAAGGAACAGAAA ACTCCAGATTCTGTTTGGACAGCAGTTCCCGAAGACAAAGTGCCAAAGCCAAGGCCTGGCTGTTGTGCAAAAC ACGGCCTTGCCGAAGCTTATAAAACCTCCATCGATTTCCCGGATGAAACTCTGTCATTCATCAAATCTCATCC CCTGATGGACTCTGCCGTTCCACCCATTGCCGATGAGCCCTGGTTCACAAAGACTCGGGTCAGGTACAGACTG ACGGCCATCTCAGTGGACCATTCAGCCGGACCCTACCAGAACTACACAGTCATCTTTGTTGGCTCTGAAGCTG GCATGGTACTTAAAGTTCTGGCAAAGACCAGTCCTTTCTCTTTGAACGACAGCGTATTACTGGAAGAGATTGA AGCCTACAACCATGCAAAGTGCAGTGCTGAGAATGAGGAAGACAAAAAGGTCATCTCATTACAGTTGGATAAA GATCACCACGCTTTATATGTGGCGTTCTCTAGCTGCATTATCCGCATCCCCCTCAGTCGCTGTGAGCGTTATG GATCATGTAAAAAGTCTTGTATTGCATCTCGTGACCCGTATTGTGGCTGGTTAAGCCAGGGATCCTGTGGTAG AGTGACCCCAGGGATGCTGCTGTTAACCGAAGACTTCTTTGCTTTCCATAACCACAGTGCTGAAGGATATGAA CAAGACACAGAATTCGGCAACACAGCTCATCTAGGGGACTGCCATGAAATTTTGCCTACTTCAACTACACCAG ATTACAAAATATTTGGCGGTCCAACATCTGGTGTACGATGGGAAGTCCAGTCTGGAGAGTCCAACCAGATGGT CCACATGAATGTCCTCATCACCTGTGTCTTTGCTGCTTTTGTTTTGGGGGCATTCATTGCAGGTGTGGCAGTA TACTGCTATCGAGACATGTTTGTTCGGAAAAACAGAAAGATCCATAAAGATGCAGAGTCCGCCCAGTCATGCA CAGACTCCAGTGGAAGTTTTGCCAAACTGAATGGTCTCTTTGACAGCCCTGTCAAGGAATACCAACAGAATAT TGATTCTCCTAAACTGTATAGTAACCTGCTAACCAGTCGGAAAGAGCTACCACCCAATGGAGATTCTAAATCC ATGGTAATGGACCATCGAGGGCAACCTCCAGAGTTGGCTGCTCTTCCTACTCCTGAGTCTACACCCGTGCTTC ACCAGAAGACCCTGCAGGCCATGAAGAGCCACTCAGAAAAGGCCCATGGCCATGGAGCTTCAAGGAAAGAAAC CCCTCAGTTTTTTCCGTCTAGTCCGCCACCTCATTCCCCATTAAGTCATGGGCATATCCCCAGTGCCATTGTT CTTCCAAATGCTACCCATGACTACAACACGTCTTTCTCAAACTCCAATGCTCACAAAGCTGAAAAGAAGCTTC AAAACATTGATCACCCTCTCACAAAGTCATCCAGTAAGAGAGATCACCGGCGTTCTGTTGATTCCAGAAATAC CCTCAATGATCTCCTGAAGCATCTGAATGACCCAAATAGTAACCCCAAAGCCATCATGGGAGACATCCAGATG GCACACCAGAACTTAATGCTGGATCCCATGGGATCGATGTCTGAGGTCCCACCTAAAGTCCCTAACCGGGAGG CATCGCTATACTCCCCTCCTTCAACTCTCCCCAGAAATAGCCCAACCAAGCGAGTGGATGTCCCCACCACTCC TGGAGTCCCAATGACTTCTCTGGAAAGACAAAGAGGTTATCACAAAAATTCCTCCCAGAGGCACTCTATATCT GCTATGCCTAAAAACTTAAACTCACCAAATGGTGTTTTGTTATCCAGACAGCCTAGTATGAACCGTGGAGGAT ATATGCCCACCCCCACTGGGGCGAAGGTGGACTATATTCAGGGAACACCAGTGAGTGTTCATCTGCAGCCTTC CCTCTCCAGACAGAGCAGCTACACCAGTAATGGCACTCTTCCTAGGACGGGACTAAAGAGGACGCCGTCCTTA AAACCTGACGTGCCACCAAAGCCTTCCTTTGTTCCTCAAACCCCATCTGTCAGACCACTGAACAAATACACAT ACTAGGCCTCAAGTGTGCTATTCCCATGTGGCTTTATCCTGTCCGTGTTGTTGAGAG
NOV16h, CG59253-06 SEQ ED NO: 210 1032 aa MW at l l5525.0kD
Protein Sequence RVFLLCAYILLLMVSQLRAVSFPEDDEPLNTVDYHCKSSRQYPVFRGRPSGNESQHRLDFQLMLKIRDTLYI AGRDQVYTVNLNEMPKTEVIWQQKLTWRSRQQDRENCAM GKHKDECHNFI VFVPRNDEMVFVCGTNAFNPM CRYYRVSTLEYDGEEISGLARCPFDARQTNVALFADGKLYSATVADFLASDAVIYRSMGDGSALRTIKYDSK IKEPHFLHAIEYGNYVYFFFREIAVEHNNLGKAVYSRVARICKNDMGGSQRVLEKH TSFLKARLNCSVPGDS FFYFDVLQSITDIIQINGIPTWGVFTTQLNSIPGSAVCAFSMDDIEKVFKGRFKEQKTPDSVWTAVPEDKVP KPRPGCCAKHGLAEAYKTSIDFPDETLSFIKSHPLMDSAVPPIADEP FTKTRVRYRLTAISVDHSAGPYQNY TVIFVGSEAGIvn^KVLAKTSPFSLNDSVLLEEIEAYNHAKCSAENEEDKKVISLQLDKDHHALYVAFSSCIIR IPLSRCERYGSCKKSCIASRDPYCG LSQGSCGRVTPGMLLLTEDFFAFHNHSAEGYEQDTEFGNTAHLGDCH EILPTSTTPDYKIFGGPTSGVR EVQSGESNQMVHMNVLITCVFAAFVLGAFIAGVAVYCYRDMFVRKNRKIH KDAESAQSCTDSSGSFAKLNGLFDSPVKEYQQNIDSPKLYSNLLTSR ELPPNGDSKS VMDHRGQPPELAAL PTPESTPVLHQKTLQAM SHSEKAHGHGASRKETPQFFPSSPPPHSPLSHGHIPSAIVLPNATHDYNTSFSNS NAHKAEKKLQNIDHPLTKSSSKRDHRRSVDSRNTLNDLLKHLNDPNSNPKAIMGDIQMAHQNL LDPMGSMSE VPPKVPNREASLYSPPSTLPRNSPTKRVDVPTTPGVP TSLERQRGYHKNSSQRHSISAMPKNLNSPNGVLLS RQPSMNRGGYMPTPTGAKVDYIQGTPVSVHLQPSLSRQSSYTSNGTLPRTGLKRTPSLKPDVPPKPSFVPQTP SVRPLNKYTY
NOV16i, CG59253-07 SEQ ED NO: 211
DNA Sequence ORF Start: ATG at 46 lORF Stop: TGA at 2350
TGGCATTTCTGAGCAGGGGCCACCCTGACTTCACCTTGGCCCACCATGAGGGTCTTCCTGCTTTGTGCCTACA
TACTGCTGCTGATGGTTTCCCAGTTGAGGGCAGTCAGCTTTCCTGAAGATGATGAACCCCTTAATACTGTCGA CTATCACTGTAAGTCGTCTAGGCAATATCCGGTTTTTAGAGGACGCCCTTCAGGCAATGAATCGCAGCACAGG CTGGACTTTCAGCTGATGTTGAAAATTCGAGACACACTTTATATTGCTGGCAGGGATCAAGTTTATACAGTAA ACTTAAATGAAATGCCCAAAACAGAAGTAATATGGCAACAGAAACTGACATGGCGATCAAGACAACAGGATCG AGAAAACTGTGCTATGAAAGGCAAGCATAAAGATGAATGCCACAACTTTATCAAAGTATTTGTTCCAAGAAAC GATGAGATGGTTTTTGTTTGTGGTACCAATGCATTCAATCCCATGTGTAGATACTACAGGGTAAGTACCTTAG AATATGATGGGGAAGAAATTAGTGGCCTGGCAAGATGCCCATTTGATGCCAGACAAACCAATGTTGCCCTCTT TGCTGATGGGAAGCTGTATTCTGCCACAGTGGCTGACTTCTTGGCCAGCGATGCCGTTATTTATCGAAGCATG GGTGATGGATCTGCCCTTCGCACAATAAAATATGATTCCAAATGGATAAAAGAGCCACACTTTCTTCATGCCA TAGAATATGGAAACTATGTCTATTTCTTCTTTCGAGAAATCGCTGTCGAACATAATAATTTAGGCAAGGCTGT GTATTCCCGCGTGGCCCGCATATGTAAAAACGACATGGGTGGTTCCCAGCGGGTCCTGGAGAAACACTGGACT TCATTTCTAAAGGCTCGGCTGAACTGTTCTGTCCCTGGAGATTCGTTTTTCTACTTTGATGTTCTGCAGTCTA TTACAGACATAATACAAATCAATGGCATCCCCACTGTGGTCGGGGTGTTTACCACGCAGCTCAATAGCATCCC TGGTTCTGCTGTCTGTGCATTTAGCATGGATGACATTGAAAAAGTATTCAAAGGACGGTTTAAGGAACAGAAA ACTCCAGATTCTGTTTGGACAGCAGTTCCCGAAGACAAAGTGCCAAAGCCAAGGCCTGGCTGTTGTGCAAAAC ACGGCCTTGCCGAAGCTTATAAAACCTCCATCGATTTCCCGGATGAAACTCTGTCATTCATCAAATCTCATCC CCTGATGGACTCTGCCGTTCCACCCATTGCCGATGAGCCCTGGTTCACAAAGACTCGGGTCAGGTACAGACTG ACGGCCATCTCAGTGGACCATTCAGCCGGACCCTACCAGAACTACACAGTCATCTTTGTTGGCTCTGAAGCTG GCATGGTACTTAAAGTTCTGGCAAAGACCAGTCCTTTCTCTTTGAACGACAGCGTATTACTGGAAGAGATTGA AGCCTACAACCATGCAAAGTGCAGTGCTGAGAATGAGGAAGACAAAAAGGTCATCTCATTACAGTTGGATAAA GATCACCACGCTTTATATGTGGCGTTCTCTAGCTGCATTATCCGCATCCCCCTCAGTCGCTGTGAGCGTTATG GATCATGTAAAAAGTCTTGTATTGCATCTCGTGACCCGTATTGTGGCTGGTTAAGCCAGGGATCCTGTGGTAG AGTGACCCCAGGGATGCTGCTGTTAACCGAAGACTTCTTTGCTTTCCATAACCACAGTGCTGAAGGATATGAA CAAGACACAGAATTCGGCAACACAGCTCATCTAGGGGACTGCCATGAAATTTTGCCTACTTCAACTACACCAG ATTACAAAATATTTGGCGGTCCAACATCTGACATGGAGGTATCTTCATCTTCTGTTACCACAATGGCAAGTAT CCCAGAAATCACACCTAAAGTGATTGATACCTGGAGACCTAAACTGACAAGCTCTCGGAAATTTGTAGTTCAA GATGATCCAAACACTTCTGATTTTACTGATCCTTTATCGGGTATCCCAAAGGGTGTACGATGGGAAGTCCAGT CTGGAGAGTCCAACCAGATGGTCCACATGAATGTCCTCATCACCTGTGTCTTTGCTGCTTTTGTTTTGGGGGC ATTCATTGCAGGTGTGGCAGTATACTGCTATCGAGACATGTTTGTTCGGAAAAACAGAAAGATCCATAAAGAT GCAGAGTCCGCCCAGTCATGCACAGACTCCAGTGGAAGTTTTGCCAAACTGAATGGTCTCTTTGACAGCCCTG TCAAGGAATACCAACAGAATATTGATTCTCCTAAACTGTATAGTAACCTGCTAACCAGTCGGAAAGAGCACGA ATTCAGCGGCCGCTGAATTCTAG
Figure imgf000252_0001
Figure imgf000253_0001
Figure imgf000254_0001
|NOV16k, CG59253-09 SEQ ID NO: 215 |3231 bp
DNA Sequence ORF Start: ATG at 10 ORF Stop: TGA at 3229
CGCAGATCTATGAGGGTCTTCCTGCTTTGTGCCTACATACTGCTGCTGATGGTTTCCCAGTTGAGGGCAGTCA
GCTTTCCTGAAGATGATGAACCCCTTAATACTGTCGACTATCACTATTCAAGGCAATATCCGGTTTTTAGAGG ACGCCCTTCAGGCAATGAATCGCAGCACAGGCTGGACTTTCAGCTGATGTTGAAAATTCGAGACACACTTTAT ATTGCTGGCAGGGATCAAGTTTATACAGTAAACTTAAATGAAATGCCCAAAACAGAAGTAATACCAAACAAGA AACTGACATGGCGATCAAGACAACAGGATCGAGAAAACTGTGCTATGAAAGGCAAGCATAAAGATGAATGCCA CAACTTTATCAAAGTATTTGTTCCAAGAAACGATGAGATGGTTTTTGTTTGTGGTACCAATGCATTCAATCCC ATGTGTAGATACTACAGGTTGAGTACCTTAGAATATGATGGGGAAGAAATTAGTGGCCTGGCAAGATGCCCAT TTGATGCCAGACAAACCAATGTTGCCCTCTTTGCTGATGGGAAGCTGTATTCTGCCACAGTGGCTGACTTCTT GGCCAGCGATGCCGTTATTTATCGAAGCATGGGTGATGGATCTGCCCTTCGCACAATAAAATATGATTCCAAA TGGATAAAAGAGCCACACTTTCTTCATGCCATAGAATATGGAAACTATGTCTATTTCTTCTTTCGAGAAATCG CTGTCGAACATAATAATTTAGGCAAGGCTGTGTATTCCCGCGTGGCCCGCATATGTAAAAACGACATGGGTGG TTCCCAGCGGGTCCTGGAGAAACACTGGACTTCATTTCTAAAGGCTCGGCTGAACTGTTCTGTCCCTGGAGAT TCGTTTTTCTACTTTGATGTTCTGCAGTCTATTACAGACATAATACAAATCAATGGCATCCCCACTGTGGTCG GGGTGTTTACCACGCAGCTCAATAGCATCCCTGGTTCTGCTGTCTGTGCATTTAGCATGGATGACATTGAAAA AGTATTCAAAGGACGGTTTAAGGAACAGAAAACTCCAGATTCTGTTTGGACAGCAGTTCCCGAAGACAAAGTG CCAAAGCCAAGGCCTGGCTGTTGTGCAAAACACGGCCTTGCCGAAGCTTATAAAACCTCCATCGATTTCCCGG ATGAAACTCTGTCATTCATCAAATCTCATCCCCTGATGGACTCTGCCGTTCCACCCATTGCTGATGAGCCCTG GTTCACAAAGACTCGGGTCAGGTACAGACTGACGGCCATCTCAGTGGACCATTCAGCCGGACCCTACCAGAAC TACACAGTCATCTTTGTTGGCTCTGAAGCTGGCATGGTACTTAAAGTTCTGGCAAAGACCAGTCCTTTCTCTT TGAACGACAGCGTATTACTGGAAGAGATTGAAGCCTACAACCATGCAAAGTGCAGTGCTGAGAATGAGGAAGA CAAAAAGGTCATCTCATTACAGTTGGATAAAGATCACCACGCTTTATATGTGGCGTTCTCTAGCTGCATTATC CGCATCCCCCTCAGTCGCTGTGAGCGTTATGGATCATGTAAAAAGTCTTGTATTGCATCTCGTGACCCGTATT GTGGCTGGTTAAGCCAGGGATCCTGTGGTAGAGTGACCCCAGGGATGCTTGCTGAAGGATATGAACAAGACAC AGAATTCGGCAACACAGCTCATCTAGGGGACTGCCATGAAATTTTGCCTACTTCAACTACACCAGATTACAAA ATATTTGGCGGTCCAACATCTGACATGGAGGTATCTTCATCTTCTGTTACCACAATGGCAAGTATCCCAGAAA TCACACCTAAAGTGATTGATACCTGGAGACCTAAACTGACAAGCTCTCGGAAATTTGTAGTTCAAGATGATCC AAACACTTCTGATTTTACTGATCCTTTATCGGGTATCCCAAAGGGTGTACGATGGGAAGTCCAGTCTGGAGAG TCCAACCAGATGGTCCACATGAATGTCCTCATCACCTGTGTCTTTGCTGCTTTTGTTTTGGGGGCATTCATTG CAGGTGTGGCAGTATACTGCTATCGAGACATGTTTGTTCGGAAAAACAGAAAGATCCATAAAGATGCAGAGTC CGCCCAGTCATGCACAGACTCCAGTGGAAGTTTTGCCAAACTGAATGGTCTCTTTGACAGCCCTGTCAAGGAA TACCAACAGAATATTGATTCTCCTAAACTGTATAGTAACCTGCTAACCAGTCGGAAAGAGCTACCACCCAATG GAGATACTAAATCCATGGTAATGGACCATCGAGGGCAACCTCCAGAGTTGGCTGCTCTTCCTACTCCTGAGTC TACACCCGTGCTTCACCAGAAGACCCTGCAGGCCATGAAGAGCCACTCAGAAAAGGCCCATGGCCATGGAGCT TCAAGGAAAGAAACCCCTCAGTTTTTTCCGTCTAGTCCGCCACCTCATTCCCCATTAAGTCATGGGCATATCC CCAGTGCCATTGTTCTTCCAAATGCTACCCATGACTACAACACGTCTTTCTCAAACTCCAATGCTCACAAAGC TGAAAAGAAGCTTCAAAACATTGATCACCCTCTCACAAAGTCATCCAGTAAGAGAGATCACCGGCGTTCTGTT GATTCCAGAAATACCCTCAATGATCTCCTGAAGCATCTGAATGACCCAAATAGTAACCCCAAAGCCATCATGG GAGACATCCAGATGGCACACCAGAACTTAATGCTGGATCCCATGGGATCGATGTCTGAGGTCCCACCTAAAGT CCCTAACCGGGAGGCATCGCTATACTCCCCTCCTTCAACTCTCCCCAGAAATAGCCCAACCAAGCGAGTGGAT GTCCCCACCACTCCTGGAGTCCCAATGACTTCTCTGGAAAGACAAAGAGGTTATCACAAAAATTCCTCCCAGA GGCACTCTATATCTGCTATGCCTAAAAACTTAAACTCACCAAATGGTGTTTTGTTATCCAGACAGCCTAGTAT GAACCGTGGAGGATATATGCCCACCCCCACTGGGGCGAAGGTGGACTATATTCAGGGAACACCAGTGAGTGTT CATCTGCAGCCTTCCCTCTCCAGACAGAGCAGCTACACCAGTAATGGCACTCTTCCTAGGACGGGACTAAAGA GGACGCCGTCCTTAAAACCTGACGTGCCACCAAAGCCTTCCTTTGTTCCTCAAACCCCATCTGTCAGACCACT GAACAAATACACATACTGA
Figure imgf000256_0001
NOV161, CG59253-10 SEQ ID NO: 218 644 aa MW at 72707.5kD Protein Sequence
VSFPEDDEPLNTVDYHYSRQYPVFRGRPSGNESQHRLDFQLMLKIRDTLYIAGRDQVYTVNLNEMPKTEVIPN KKLT RSRQQDRENO^KGIHKDECHNFIKVFVPRNDEIWFVCGTNAFNPMCRYYRLSTLEYDGEEISGLARC PFDARQTNVALFADGKLYSATVADFLASDAVIYRS GDGSALRTIKYDSK IKEPHFLHAIEYGNYVYFFFRE IAVEHNNLGKAVYSRVARICKNDMGGSQRVLEKHWTSFLKARLNCSVPGDSFFYFDVLQSITDIIQINGIPTV VGVFTTQLNSIPGSAVCAFSMDDIEKVFKGRFKEQKTPDSVWTAVPEDKVPKPRPGCCAKHGLAEAYKTSIDF PDETLSFIKSHPL DSAVPPIADEPWFT TRVRYRLTAISVDHSAGPYQNYTVIFVGSEAGMVLKVLAKTSPF SLNDSVLLEEIEAYNHA CSAENEEDKKVISLQLDKDHHALYVAFSSCIIRIPLSRCERYGSCKKSCIASRDP YCGWLSQGSCGRVTPG LLLTEDFFAFHNHSAEGYEQDTEFGNTAHLGDCHEILPTSTTPDYKIFGGPTSDME VSSSSVTTMASIPEITPKVIDT RPKLTSSRKFWQDDPNTSDFTDPLSGIPKGVR EVQ
NOVl 6m, SNP13381547 of SEQ ID NO: 219 |1894bp CG59253-01, DNA Sequence
SNP Pos: 215 SNP Change: T to C
TGGCATTTCTGAGCAGGGGCCACCCTGACTTCACCTTGGCCCACCATGAGGGTCTTCCTGCTTTGTGCCTACA
TACTGCTGCTGATGGTTTCCCAGTTGAGGGCAGTCAGCTTTCCTGAAGATGATGAACCCCTTAATACTGTCGA CTATCACTATTCAAGGCAATATCCGGTTTTTAGAGGACGCCCTTCAGGCAATGAATCGCAGCACAGGCCGGAC TTTCAGCTGATGTTGAAAATTCGAGACACACTTTATATTGCTGGCAGGGATCAAGTTTATACAGTAAACTTAA ATGAAATGCCCAAAACAGAAGTAATACCCAACAAGAAACTGACATGGCGATCAAGACAACAGGATCGAGAAAA CTGTGCTATGAAAGGCAAGCATAAAGATGAATGCCACAACTTTATCAAAGTATTTGTTCCAAGAAACGATGAG ATGGTTTTTGTTTGTGGTACCAATGCATTCAATCCCATGTGTAGATACTACAGGTTGAGTACCTTAGAATATG ATGGGGAAGAAATTAGTGGCCTGGCAAGATGCCCATTTGATGCCAGACAAACCAATGTTGCCCTCTTTGCTGA TGGGAAGCTGTATTCTGCCACAGTGGCTGACTTCTTGGCCAGCGATGCCGTTATTTATCGAAGCATGGGTGAT GGATCTGCCCTTCGCACAATAAAATATGATTCCAAATGGATAAAAGAGCCACACTTTCTTCATGCCATAGAAT ATGGAAACTATGTCTATTTCTTCTTTCGAGAAATCGCTGTCGAACATAATAATTTAGGCAAGGCTGTGTATTC CCGCGTGGCCCGCATATGTAAAAACGACATGGGTGGTTCCCAGCGGGTCCTGGAGAAACACTGGACTTCATTT CTAAAGGCTCGGCTGAACTGTTCTGTCCCTGGAGATTCGTTTTTCTACTTTGATGTTCTGCAGTCTATTACAG ACATAATACAAATCAATGGCATCCCCACTGTGGTCGGGGTGTTTACCACGCAGCTCAATAGCATCCCTGGTTC TGCTGTCTGTGCATTTAGCATGGATGACATTGAAAAAGTATTCAAAGGACGGTTTAAGGAACAGAAAACTCCA GATTCTGTTTGGACAGCAGTTCCCGAAGACAAAGTGCCAAAGCCAAGGCCTGGCTGTTGTGCAAAACACGGCC TTGCCGAAGCTTATAAAACCTCCATCGATTTCCCGGATGAAACTCTGTCATTCATCAAATCTCATCCCCTGAT GGACTCTGCCGTTCCACCCATTGCCGATGAGCCCTGGTTCACAAAGACTCGGGTCAGGTACAGACTGACGGCC ATCTCAGTGGACCATTCAGCCGGACCCTACCAGAACTACACAGTCATCTTTGTTGGCTCTGAAGCTGGCATGG TACTTAAAGTTCTGGCAAAGACCAGTCCTTTCTCTTTGAACGACAGCGTATTACTGGAAGAGATTGAAGCCTA CAACCATGCAAAGTAGGTATATGTTACGAGAACGCCCTTCAGCACTGCTCAAAAATTTTCGGCATGTATTTCA TCTAGTCATGTCCTTTTGGTCCTCTAAATTAGCAGTGGTTTGGCATAATAGTGTTTTGTGTTTTTTTTCTCAT
TGAAATAAATCTTGGGTTTGTTTTTTTCCCGAGCCTGCTAGGGCGAGGGGGGTGAATGGTTGATGAGTTTAAA
AATAATGCAGCCCTTGTTTTTCACCTGTAGAATATGAGAACATTTTAACAGCACCTCTCTTATCTTGCAGATA
TATTCCAAGATGCTACATGCAGCAGACAGCTGTGAGCTTGCATACACACACACACAAATATACATGCACATAC
ATACACAGAATGTAGTACTAGTTAAGTATTTCCTTCCTATCTTTAATAAGTAAGAGAATATTTAGACCA
|NOV16m, SNP13381547 of SEQ ID NO: 220 1476 aa MW at 54200.4kD
CG59253-01, Protein Sequence SNP Pos: 57 ISNP Change: Leu to Pro
MRVFLLCAYILLLMVSQLRAVSFPΞDDEPLNTVDYHYSRQYPVFRGRPSGNESQHRPDFQLMLKIRDTLYIAG RDQVYTVNLNEMPKTEVIPNKKLT RSRQQDRENCAMKGKHKDECHNFIKVFVPRNDEMVFVCGTNAFNPMCR YYRLSTLEYDGEEISGLARCPFDARQTNVALFADGKLYSATVADFLASDAVIYRSMGDGSALRTIKYDSK IK EPHFLHAIEYGNYVYFFFREIAVEHNNLGKAVYSRVARICKND GGSQRVLEKH TSFLKARLNCSVPGDSFF YFDVLQSITDIIQINGIPTWGVFTTQLNSIPGSAVCAFSMDDIEKVFKGRFKEQKTPDSVTAVPEDKVPKP RPGCCAKHGLAEAYKTSIDFPDETLSFIKSHPLMDSAVPPIADEPWFTKTRVRYRLTAISVDHSAGPYQNYTV IFVGSEAGMVLKVLAKTSPFSLNDSVLLEEIEAYNHAK
Figure imgf000258_0001
NOVl 60, SNP13378935 of SEQ ID NO: 223 1894 bp CG59253-01, DNA Sequence ORF Start: ATG at 46 ORF Stop: TAG at 1474
SNP Pos: 965 JSNP Change: A to G
TGGCATTTCTGAGCAGGGGCCACCCTGACTTCACCTTGGCCCACCATGAGGGTCTTCCTGCTTTGTGCCTACA
TACTGCTGCTGATGGTTTCCCAGTTGAGGGCAGTCAGCTTTCCTGAAGATGATGAACCCCTTAATACTGTCGA CTATCACTATTCAAGGCAATATCCGGTTTTTAGAGGACGCCCTTCAGGCAATGAATCGCAGCACAGGCTGGAC TTTCAGCTGATGTTGAAAATTCGAGACACACTTTATATTGCTGGCAGGGATCAAGTTTATACAGTAAACTTAA ATGAAATGCCCAAAACAGAAGTAATACCCAACAAGAAACTGACATGGCGATCAAGACAACAGGATCGAGAAAA CTGTGCTATGAAAGGCAAGCATAAAGATGAATGCCACAACTTTATCAAAGTATTTGTTCCAAGAAACGATGAG ATGGTTTTTGTTTGTGGTACCAATGCATTCAATCCCATGTGTAGATACTACAGGTTGAGTACCTTAGAATATG ATGGGGAAGAAATTAGTGGCCTGGCAAGATGCCCATTTGATGCCAGACAAACCAATGTTGCCCTCTTTGCTGA TGGGAAGCTGTATTCTGCCACAGTGGCTGACTTCTTGGCCAGCGATGCCGTTATTTATCGAAGCATGGGTGAT GGATCTGCCCTTCGCACAATAAAATATGATTCCAAATGGATAAAAGAGCCACACTTTCTTCATGCCATAGAAT ATGGAAACTATGTCTATTTCTTCTTTCGAGAAATCGCTGTCGAACATAATAATTTAGGCAAGGCTGTGTATTC CCGCGTGGCCCGCATATGTAAAAACGACATGGGTGGTTCCCAGCGGGTCCTGGAGAAACACTGGACTTCATTT CTAAAGGCTCGGCTGAACTGTTCTGTCCCTGGAGATTCGTTTTTCTACTTTGATGTTCTGCAGTCTATTACAG ACATAATACAAATCAGTGGCATCCCCACTGTGGTCGGGGTGTTTACCACGCAGCTCAATAGCATCCCTGGTTC TGCTGTCTGTGCATTTAGCATGGATGACATTGAAAAAGTATTCAAAGGACGGTTTAAGGAACAGAAAACTCCA GATTCTGTTTGGACAGCAGTTCCCGAAGACAAAGTGCCAAAGCCAAGGCCTGGCTGTTGTGCAAAACACGGCC TTGCCGAAGCTTATAAAACCTCCATCGATTTCCCGGATGAAACTCTGTCATTCATCAAATCTCATCCCCTGAT GGACTCTGCCGTTCCACCCATTGCCGATGAGCCCTGGTTCACAAAGACTCGGGTCAGGTACAGACTGACGGCC ATCTCAGTGGACCATTCAGCCGGACCCTACCAGAACTACACAGTCATCTTTGTTGGCTCTGAAGCTGGCATGG TACTTAAAGTTCTGGCAAAGACCAGTCCTTTCTCTTTGAACGACAGCGTATTACTGGAAGAGATTGAAGCCTA CAACCATGCAAAGTAGGTATATGTTACGAGAACGCCCTTCAGCACTGCTCAAAAATTTTCGGCATGTATTTCA TCTAGTCATGTCCTTTTGGTCCTCTAAATTAGCAGTGGTTTGGCATAATAGTGTTTTGTGTTTTTTTTCTCAT
TGAAATAAATCTTGGGTTTGTTTTTTTCCCGAGCCTGCTAGGGCGAGGGGGGTGAATGGTTGATGAGTTTAAA lAATAATGCAGCCCTTGTTTTTCACCTGTAGAATATGAGAACATTTTAACAGCACCTCTCTTATCTTGCAGATA
TATTCCAAGATGCTACATGCAGCAGACAGCTGTGAGCTTGCATACACACACACACAAATATACATGCACATAC lATACACAGAATGTAGTACTAGTTAAGTATTTCCTTCCTATCTTTAATAAGTAAGAGAATATTTAGACCA
Figure imgf000259_0001
NOV16p, SNP13381569 of SEQ ID NO: 225 1894 bp CG59253-01, DNA Sequence ORF Start: ATG at 46 {ORF Stop: TAG at 1474
SNP Pos: 1351
TGGCATTTCTGAGCAGGGGCCACCCTGACTTCACCTTGGCCCACCATGAGGGTCTTCCTGCTTTGTGCCTACA
TACTGCTGCTGATGGTTTCCCAGTTGAGGGCAGTCAGCTTTCCTGAAGATGATGAACCCCTTAATACTGTCGA CTATCACTATTCAAGGCAATATCCGGTTTTTAGAGGACGCCCTTCAGGCAATGAATCGCAGCACAGGCTGGAC TTTCAGCTGATGTTGAAAATTCGAGACACACTTTATATTGCTGGCAGGGATCAAGTTTATACAGTAAACTTAA ATGAAATGCCCAAAACAGAAGTAATACCCAACAAGAAACTGACATGGCGATCAAGACAACAGGATCGAGAAAA CTGTGCTATGAAAGGCAAGCATAAAGATGAATGCCACAACTTTATCAAAGTATTTGTTCCAAGAAACGATGAG ATGGTTTTTGTTTGTGGTACCAATGCATTCAATCCCATGTGTAGATACTACAGGTTGAGTACCTTAGAATATG ATGGGGAAGAAATTAGTGGCCTGGCAAGATGCCCATTTGATGCCAGACAAACCAATGTTGCCCTCTTTGCTGA TGGGAAGCTGTATTCTGCCACAGTGGCTGACTTCTTGGCCAGCGATGCCGTTATTTATCGAAGCATGGGTGAT GGATCTGCCCTTCGCACAATAAAATATGATTCCAAATGGATAAAAGAGCCACACTTTCTTCATGCCATAGAAT ATGGAAACTATGTCTATTTCTTCTTTCGAGAAATCGCTGTCGAACATAATAATTTAGGCAAGGCTGTGTATTC CCGCGTGGCCCGCATATGTAAAAACGACATGGGTGGTTCCCAGCGGGTCCTGGAGAAACACTGGACTTCATTT CTAAAGGCTCGGCTGAACTGTTCTGTCCCTGGAGATTCGTTTTTCTACTTTGATGTTCTGCAGTCTATTACAG ACATAATACAAATCAATGGCATCCCCACTGTGGTCGGGGTGTTTACCACGCAGCTCAATAGCATCCCTGGTTC TGCTGTCTGTGCATTTAGCATGGATGACATTGAAAAAGTATTCAAAGGACGGTTTAAGGAACAGAAAACTCCA GATTCTGTTTGGACAGCAGTTCCCGAAGACAAAGTGCCAAAGCCAAGGCCTGGCTGTTGTGCAAAACACGGCC TTGCCGAAGCTTATAAAACCTCCATCGATTTCCCGGATGAAACTCTGTCATTCATCAAATCTCATCCCCTGAT GGACTCTGCCGTTCCACCCATTGCCGATGAGCCCTGGTTCACAAAGACTCGGGTCAGGTACAGACTGACGGCC ATCTCAGTGGACCATTCAGCCGGACCCTACCAGAACCACACAGTCATCTTTGTTGGCTCTGAAGCTGGCATGG TACTTAAAGTTCTGGCAAAGACCAGTCCTTTCTCTTTGAACGACAGCGTATTACTGGAAGAGATTGAAGCCTA CAACCATGCAAAGTAGGTATATGTTACGAGAACGCCCTTCAGCACTGCTCAAAAATTTTCGGCATGTATTTCA TCTAGTCATGTCCTTTTGGTCCTCTAAATTAGCAGTGGTTTGGCATAATAGTGTTTTGTGTTTTTTTTCTCAT
TGAAATAAATCTTGGGTTTGTTTTTTTCCCGAGCCTGCTAGGGCGAGGGGGGTGAATGGTTGATGAGTTTAAA
AATAATGCAGCCCTTGTTTTTCACCTGTAGAATATGAGAACATTTTAACAGCACCTCTCTTATCTTGCAGATA
ITATTCCAAGATGCTACATGCAGCAGACAGCTGTGAGCTTGCATACACACACACACAAATATACATGCACATAC
ATACACAGAATGTAGTACTAGTTAAGTATTTCCTTCCTATCTTTAATAAGTAAGAGAATATTTAGACCA
NOVlόp, SNP13381569 of SEQ ID NO: 226 476 aa MW at 54190.4kD CG59253-01, Protein Sequence SNP Pos: 436 SNP Change: Tyr to His
MRVFLLCAYILLLMVSQLRAVSFPEDDEPLNTVDYHYSRQYPVFRGRPSGNESQHRLDFQLMLKIRDTLYIAG RDQVYTVNLNEMPKTEVIPNKKLTWRSRQQDRENCAMKGKHKDECHNFIKVFVPRNDEMVFVCGTNAFNPMCR YYRLSTLEYDGEEISGLARCPFDARQTNVALFADG LYSATVADFLASDAVIYRSMGDGSALRTIKYDSK IK EPHFLHAIEYGNYVYFFFREIAVEHNNLGKAVYSRVARICKNDMGGSQRVLEKH TSFLKARLNCSVPGDSFF YFDVLQSITDIIQINGIPTWGVFTTQLNSIPGSAVCAFSMDDIEKVFKGRFKEQKTPDSVTAVPEDKVPKP RPGCCAKHGLAEAYKTSIDFPDETLSFIKSHPLMDSAVPPIADEPWFTKTRVRYRLTAISVDHSAGPYQNHTV IFVGSEAGMVLKVLAKTSPFSLNDSVLLEEIEAYNHAK
NOV16q, SNP13382528 of SEQ ID NO: 227 1894bp CG59253-01, DNA Sequence |ORF Start: ATG at 46 lORF Stop: TAG at 1474
SNP Pos: 1838 SNP Change: T to C
TGGCATTTCTGAGCAGGGGCCACCCTGACTTCACCTTGGCCCACCATGAGGGTCTTCCTGCTTTGTGCCTACA
TACTGCTGCTGATGGTTTCCCAGTTGAGGGCAGTCAGCTTTCCTGAAGATGATGAACCCCTTAATACTGTCGA CTATCACTATTCAAGGCAATATCCGGTTTTTAGAGGACGCCCTTCAGGCAATGAATCGCAGCACAGGCTGGAC TTTCAGCTGATGTTGAAAATTCGAGACACACTTTATATTGCTGGCAGGGATCAAGTTTATACAGTAAACTTAA ATGAAATGCCCAAAACAGAAGTAATACCCAACAAGAAACTGACATGGCGATCAAGACAACAGGATCGAGAAAA CTGTGCTATGAAAGGCAAGCATAAAGATGAATGCCACAACTTTATCAAAGTATTTGTTCCAAGAAACGATGAG ATGGTTTTTGTTTGTGGTACCAATGCATTCAATCCCATGTGTAGATACTACAGGTTGAGTACCTTAGAATATG ATGGGGAAGAAATTAGTGGCCTGGCAAGATGCCCATTTGATGCCAGACAAACCAATGTTGCCCTCTTTGCTGA TGGGAAGCTGTATTCTGCCACAGTGGCTGACTTCTTGGCCAGCGATGCCGTTATTTATCGAAGCATGGGTGAT GGATCTGCCCTTCGCACAATAAAATATGATTCCAAATGGATAAAAGAGCCACACTTTCTTCATGCCATAGAAT ATGGAAACTATGTCTATTTCTTCTTTCGAGAAATCGCTGTCGAACATAATAATTTAGGCAAGGCTGTGTATTC CCGCGTGGCCCGCATATGTAAAAACGACATGGGTGGTTCCCAGCGGGTCCTGGAGAAACACTGGACTTCATTT CTAAAGGCTCGGCTGAACTGTTCTGTCCCTGGAGATTCGTTTTTCTACTTTGATGTTCTGCAGTCTATTACAG ACATAATACAAATCAATGGCATCCCCACTGTGGTCGGGGTGTTTACCACGCAGCTCAATAGCATCCCTGGTTC TGCTGTCTGTGCATTTAGCATGGATGACATTGAAAAAGTATTCAAAGGACGGTTTAAGGAACAGAAAACTCCA GATTCTGTTTGGACAGCAGTTCCCGAAGACAAAGTGCCAAAGCCAAGGCCTGGCTGTTGTGCAAAACACGGCC TTGCCGAAGCTTATAAAACCTCCATCGATTTCCCGGATGAAACTCTGTCATTCATCAAATCTCATCCCCTGAT GGACTCTGCCGTTCCACCCATTGCCGATGAGCCCTGGTTCACAAAGACTCGGGTCAGGTACAGACTGACGGCC ATCTCAGTGGACCATTCAGCCGGACCCTACCAGAACTACACAGTCATCTTTGTTGGCTCTGAAGCTGGCATGG TACTTAAAGTTCTGGCAAAGACCAGTCCTTTCTCTTTGAACGACAGCGTATTACTGGAAGAGATTGAAGCCTA CAACCATGCAAAGTAGGTATATGTTACGAGAACGCCCTTCAGCACTGCTCAAAAATTTTCGGCATGTATTTCA TCTAGTCATGTCCTTTTGGTCCTCTAAATTAGCAGTGGTTTGGCATAATAGTGTTTTGTGTTTTTTTTCTCAT
TGAAATAAATCTTGGGTTTGTTTTTTTCCCGAGCCTGCTAGGGCGAGGGGGGTGAATGGTTGATGAGTTTAAA
AATAATGCAGCCCTTGTTTTTCACCTGTAGAATATGAGAACATTTTAACAGCACCTCTCTTATCTTGCAGATA
TATTCCAAGATGCTACATGCAGCAGACAGCTGTGAGCTTGCATACACACACACACAAATATACATGCACATAC
ATACACAGAATGCAGTACTAGTTAAGTATTTCCTTCCTATCTTTAATAAGTAAGAGAATATTTAGACCA
Figure imgf000261_0001
A ClustalW comparison of the above protem sequences yields the following sequence alignment shown in Table 16B. Table 16B. Comparison of the NOV16 protein sequences.
NOV16a MRVFLLCAYILLLMVSQLRAVSFPEDDEPLNTVDYHY- -SRQYPVFRGRPSGNESQHRLD
NOV16b GSVSFPEDDEPLNTVDYHY--SRQYPVFRGRPSGNESQHRLD
NOV16C MRVFLLCAYILLLMVSQLRAVSFPEDDEPLNTVDYHCKSSRQYPVFRGRPSGNESQHRLD
NOVlSd GSVSFPEDDEPLNTVDYHY--SRQYPVFRGRPSGNESQHRLD
NOV16e VSFPEDDEPLNTVDYHY--SRQYPVFRGRPSGNESQHRLD
NOV16f VSFPEDDEPLNTVDYHY--SRQYPVFRGRPSGNESQHRLD
NOV16g MRVFLLCAYILLLMVSQLRAVSFPEDDEPLNTVDYHCKSSRQYPVFRGRPSGNESQHRLD
NOVl6h MRVFLLCAYILLLMVSQLRAVSFPEDDEPLNTVDYHCKSSRQYPVFRGRPSGNESQHRLD
NOV16i RVFLLCAYILLLMVSQLRAVSFPEDDEPLNTVDYHCKSSRQYPVFRGRPSGNESQHRLD
NOVlδj MRVFLLCAYILLLMVSQLRAVSFPEDDEPLNTVDYHCKSSRQYPVFRGRPSGNESQHRLD
NOV16 MRVFLLCAYILLLMVSQLRAVSFPEDDEPLNTVDYHY- -SRQYPVFRGRPSGNESQHRLD
NOV161 VSFPEDDEPLNTVDYHY--SRQYPVFRGRPSGNESQHRLD
NOV16a FQLMLKIRDTLYIAGRDQVYTVNLNEMPKTEVIPNKKLTWRSRQQDRENCAMKGKHKDEC
NOVl6b FQLMLKIRDTLYIAGRDQVYTVNLNEMPKTEVIPNKKLT RSRQQDRENCAMKGKH DEC
NOV16C FQL LKIRDTLYIAGRDQVYTVNLNEMPKTEVIWQQKLTWRSRQQDRENCAMKGKHKDEC
NOVl6d FQLMLKIRDTLYIAGGDQVYTVNLNEMPKTEVIPNKKLT RSRQQDRENCAMKGKHKDEC
NOV16e FQLMLKIRDTLYIAGRDQVYTVNLNEMPKTEVIPNKKLT RSRQQDRENCAMKGKHKDEC
NOV16f FQLMLKIRDTLYIAGGDQVYTVNLNEMPKTEVIPNKKLT RSRQQDRENCAMKGKHKDEC
NOV16g FQLMLKIRDTLYIAGRDQVYTVNLNEMPKTEVI QQKLTWRSRQQDRENCAMKGKHKDEC
NOV16h FQLMLKIRDTLYIAGRDQVYTVNLNEMPKTEVI QQKLT RSRQQDRENCAMKGKHKDEC
NOV16i FQLMLKIRDTLYIAGRDQVYTVNLNEMPKTEVI QQKLTWRSRQQDRENCAMKGKHKDEC
NOV16J FQLMLKIRDTLYIAGRDQVYTVNLNEMPKTEVIWQQKLT RSRQQDRENCAMKGKHKDEC
NOV16k FQLMLKIRDTLYIAGRDQVYTVNLNEMPKTEVIPNKKLTWRSRQQDRENCAMKGKHKDEC
NOV161 FQLMLKIRDTLYIAGRDQVYTVNLNEMPKTEVIPNKKLTWRSRQQDRENCAMKGKHKDEC
NOVlβa HNFIKVFVPRNDEMVFVCGTNAFNPMCRYYRLSTLEYDGEEISGLARCPFDARQTNVALF
NOVl6b HNFIKVFVPRNDEMVFVCGTNAFNPMCRYYRLSTLEYDGEEISGLARCPFDARQTNVALF
NOVl6c HNFIKVFVPRNDEMVFVCGTNAFNPMCRYYRVSTLEYDGEEISGLARCPFDARQTNVALF
NOVl6d HNFIKVFVPRNDEMVFVCGTNAFNPMCRYYRLSTLEYDGEEISGLARCPFDARQTNVALF
NOV16e HNFIKVFVPRNDEMVFVCGTNAFNPMCRYYRLSTLEYDGEEISGLARCPFDARQTNVALF
NOV16f HNFIKVFVPRNDEMVFVCGTNAFNPMCRYYRLSTLEYDGEEISGLARCPFDARQTNVALF
NOV16g HNFIKVFVPRNDEMVFVCGTNAFNPMCRYYRVSTLEYDGEEISGLARCPFDARQTNVALF
NOV16h HNFIKVFVPRNDEMVFVCGTNAFNPMCRYYRVSTLEYDGEEISGLARCPFDARQTNVALF
NOVl6i HNFIKVFVPRNDEMVFVCGTNAFNPMCRYYRVSTLEYDGEEISGLARCPFDARQTNVALF
NOVl6j HNFIKVFVPRNDEMVFVCGTNAFNPMCRYYRVSTLEYDGEEISGLARCPFDARQTNVALF
NOV16k HNFIKVFVPRNDEMVFVCGTNAFNPMCRYYRLSTLEYDGEEISGLARCPFDARQTNVALF
NOVl61 HNFIKVFVPRNDEMVFVCGTNAFNPMCRYYRLSTLEYDGEEISGLARCPFDARQTNVALF
NOV16a ADGKLYSATVADFLASDAVIYRSMGDGSALRTIKYDSK IKEPHFLHAIEYGNYVYFFFR
NOV16b ADGKLYSATVADFLASDAVIYRSMGDGSALRTIKYDSK IKEPHFLHAIEYGNYVYFFFR
NOV16c ADGKLYSATVADFLASDAVIYRSMGDGSALRTIKYDSK IKEPHFLHAIEYGNYVYFFFR
NOV16d ADGKLYSATVADFLASDAVIYRSMGDGSALRTIKYDSKWIKEPHFLHAIEYGNYVYFFFR
NOV16e ADGKLYSATVADFLASDAVIYRSMGDGSALRTIKYDSK IKEPHFLHAIEYGNYVYFFFR
NOV16f ADGKLYSATVADFLASDAVIYRSMGDGSALRTIKYDSKWIKEPHFLHAIEYGNYVYFFFR
NOV16g ADGKLYSATVADFLASDAVIYRSMGDGSALRTIKYDSK IKEPHFLHAIEYGNYVYFFFR
NOV16h ADGKLYSATVADFLASDAVIYRSMGDGSALRTIKYDSK IKEPHFLHAIEYGNYVYFFFR
NOV16i ADGKLYSATVADFLASDAVIYRSMGDGSALRTIKYDSK IKEPHFLHAIEYGNYVYFFFR
NOVl6j ADGKLYSATVADFLASDAVIYRSMGDGSALRTIKYDSK IKEPHFLHAIEYGNYVYFFFR
NOV16k ADGKLYSATVADFLASDAVIYRSMGDGSALRTIKYDSK IKEPHFLHAIEYGNYVYFFFR
NOV161 ADGKLYSATVADFLASDAVIYRSMGDGSALRTIKYDSK IKEPHFLHAIEYGNYVYFFFR
NOV16a EIAVEHNNLGKAVYSRVARICKNDMGGSQRVLEKH TSFLKARLNCSVPGDSFFYFDVLQ
NOV16b EIAVEHNNLGKAVYSRVARICKNDMGGSQRVLEKHWTSFLKARLNCSVPGDSFFYFDVLQ
NOV16c EIAVEHNNLGKAVYSRVARICKNDMGGSQRVLEKHWTSFLKARLNCSVPGDSFFYFDVLQ
NOVlδd EIAVEHNNLGKAVYSRVARICKNDMGGSQRVLEKH TSFLKARLNCSVPGDSFFYFDVLQ NOV16e EIAVEHNNLGKAVYSRVARICKNDMGGSQRVLEKH TSFLKARLNCSVPGDSFFYFDVLQ
NOV16f EIAVEHNNLGKAVYSRVARICKNDMGGSQRVLEKH TSFLKARLNCSVPGDSFFYFDVLQ
NOV16g EIAVEHNNLGKAVYSRVARICKNDMGGSQRVLEKHWTSFLKARLNCSVPGDSFFYFDVLQ
NOVl6 EIAVEHNNLGKAVYSRVARICKNDMGGSQRVLEKH TSFLKARLNCSVPGDSFFYFDVLQ
NOV16i EIAVEHNNLGKAVYSRVARICKNDMGGSQRVLEKH TSFLKARLNCSVPGDSFFYFDVLQ
NOV16J EIAVEHNNLGKAVYSRVARICKNDMGGSQRVLEKHWTSFLKARLNCSVPGDSFFYFDVLQ
NOVl6k EIAVEHNNLGKAVYSRVARICKNDMGGSQRVLEKH TSFLKARLNCSVPGDSFFYFDVLQ
NOV161 EIAVEHNNLGKAVYSRVARICKNDMGGSQRVLEKHWTSFLKARLNCSVPGDSFFYFDVLQ
NOV16a SITDIIQINGIPTWGVFTTQLNSIPGSAVCAFSMDDIEKVFKGRFKEQKTPDSV TAVP
NOV16b SITDIIQINGIPTWGVFTTQLNSIPGSAVCAFSMDDIEKVFKGRFKEQKTPDSV TAVP
NOVl 6 c SI DIIQINGIPTWGVFTTQLNSIPGSAVCAFSMDDIEKVFKGRFKEQKTPDSVWTAVP
NOV16d SITDIIQINGIPTWGVFTTQLNSIPGSAVCAFSMDDIEKVFKGRFKEQKTPDSV TAVP
NOV16e SITDIIQINGIPTWGVFTTQLNSIPGSAVCAFSMDDIEKVFKGRFKEQKTPDSVWTAVP
NOV16f SITDIIQINGIPTWGVFTTQLNSIPGSAVCAFSMDDIEKVFKGRFKEQKTPDSVWTAVP
NOV16g SITDIIQINGIPTWGVFTTQLNSIPGSAVCAFSMDDIEKVFKGRFKEQKTPDSV TAVP
NOV16h SITDIIQINGIPTWGVFTTQLNSIPGSAVCAFSMDDIEKVFKGRFKEQKTPDSVWTAVP
NOV16 SITDIIQINGIPTWGVFTTQLNSIPGSAVCAFSMDDIEKVFKGRFKEQKTPDSV TAVP
NOV16J SITDIIQINGIPTWGVFTTQLNSIPGSAVCAFSMDDIEKVFKGRFKEQKTPDSVHTAVP
NOVl6k SITDIIQINGIPTWGVFTTQLNSIPGSAVCAFSMDDIEKVFKGRFKEQKTPDSV TAVP
NOV161 SITDIIQINGIPTWGVFTTQLNSIPGSAVCAFSMDDIEKVFKGRFKEQKTPDSV TAVP
NOV16a EDKVPKPRPGCCAKHGLAEAYKTSIDFPDETLSFIKSHPLMDSAVPPIADEPWFTKTRVR
NOV16b EDKVPKPRPGCCAKHGLAEAYKTSIDFPDETLSFIKSHPLMDSAVPPIADEP FTKTRVR
NOV16C EDKVPKPRPGCCAKHGLAEAYKTSIDFPDETLSFIKSHPLMDSAVPPIADEPWFTKTRVR
NOV16d ΞDKVPKPRPGCCAKHGLAEAYKTSIDFPDETLSFIKSHPLMDSAVPPIADEP FTKTRVR
NOV16e EDKVPKPRPGCCAKHGLAEAYKTSIDFPDETLSFIKSHPLMDSAVPPIADEP FTKTRVR
NOV16f EDKVPKPRPGCCAKHGLAEAYKTSIDFPDETLSFIKSHPLMDSAVPPIADEPWFTKTRVR
NOVl6g EDKVPKPRPGCCAKHGLAEAYKTSIDFPDETLSFIKSHPLMDSAVPPIADEP FTKTRVR
NOVl6h EDKVPKPRPGCCAKHGLAEAYKTSIDFPDETLSFIKSHPLMDSAVPPI DEPWFTKTRVR
NOV16i EDKVPKPRPGCCAKHGLAEAYKTSIDFPDETLSFIKSHPLMDSAVPPIADEP FTKTRVR
NOVl6 EDKVPKPRPGCCAKHGLAEAYKTSIDFPDETLSFIKSHPLMDSAVPPIADEP FTKTRVR
NOV16k EDKVPKPRPGCCAKHGLAEAYKTSIDFPDETLSFIKSHPLMDSAVPPIADEP FTKTRVR
NOV161 EDKVPKPRPGCCAKHGLAEAYKTSIDFPDETLSFIKSHPLMDSAVPPIADEP FTKTRVR
NOV16a YRLTAISVDHSAGPYQNYTVIFVGSEAGMVLKVLAKTSPFSLNDSVLLEEIEAYNHAK--
NOV16b YRLTAISVDHSAGPYQNYTVIFVGSEAGMVLKVLAKTSPFSLNDSVLLEEIEAYNHAKCL
NOVl 6c YRLTAISVDHSAGPYQNYTVIFVGSEAGMVLKVLAKTSPFSLNDSVLLEEIEAYNHAKCS
NOVl6d YRLTAISVDHSAGPYQNYTVIFVGSEAGMVLKVLAKTSPFSLNDSVLLEEIEAYNHAKCN
NOV16e YRLTAISVDHSAGPYQNYTVIFVGSEAGMVLKVLAKTSPFSLNDSVLLEEIEAYNHAK- -
NOVlδf YRLTAISVDHSAGPYQNYTVIFVGSEAGMVLKVLAKTSPFSLNDSVLLEEIEAYNHAKCN
NOV16g YRLTAISVDHSAGPYQNYTVIFVGSEAGMVLKVLAKTSPFSLNDSVLLEEIEAYNHAKCS
NOV16 YRLTAISVDHSAGPYQNYTVIFVGSEAGMVLKVLAKTSPFSLNDSVLLEEIEAYNHAKCS
NOV16i YRLTAISVDHSAGPYQNYTVIFVGSEAGMVLKVLAKTSPFSLNDSVLLEEIEAYNHAKCS
NOVl6j YRLTAISVDHSAGPYQNYTVIFVGSEAGMVLKVLAKTSPFSLNDSVLLEEIEAYNHAKCS
NOVl6k YRLTAISVDHSAGPYQNYTVIFVGSEAGMVLKVLAKTSPFSLNDSVLLEEIEAYNHAKCS
NOVl61 YRLTAISVDHSAGPYQNYTVIFVGSEAGMVLKVLAKTSPFSLNDSVLLEEIEAYNHAKCS
NOV16a
NOV16b E
NOVl6c AENEEDKKVISLQLDKDHHALYVAFSSCIIRIPLSRCERYGSCKKSCI SRDPYCGWLSQ
NOVlSd AENEEDKKVISLQLDKDHHALYVAFSSCIIRIPLSRCERYGSCKKSCIASRDPYCGWLSQ
NOV16e
NOV16f AENEEDKKVISLQLDKDHHALYVAFSSCIIRIPLSRCERYGSCKKSCIASRDPYCGWLSQ
NOV16g AENEEDKKVISLQLDKDHHALYVAFSSCIIRIPLSRCERYGSCKKSCIASRDPYCGWLSQ
NOV16h AENEEDKKVISLQLDKDHHALYVAFSSCIIRIPLSRCERYGSCKKSCIASRDPYCGWLSQ
NOV16i AENΞEDKKVISLQLDKDHHALYVAFSSCIIRIPLSRCERYGSCKKSCIASRDPYCGWLSQ
NOV16J AENEEDKKVISLQLDKDHHALYVAFSSCIIRIPLSRCERYGSCKKSCIASRDPYCGWLSQ
NOVl6k AENEEDKKVISLQLDKDHHALYVAFSSCIIRIPLSRCERYGSCKKSCIASRDPYCGWLSQ NOV161 AENEEDKKVISLQLDKDHHALYVAFSSCIIRIPLSRCERYGSCKKSCIASRDPYCGWLSQ
NOV16a
NOV16b
NOVl6c GSCGRVTP- -NHSAEGYEQDTEFGNTAHLGDCHAYEPYEGR- NOV16d GSCGRV P- -GMLAEGYEQDTEFGNTAHLGD NOVl6e NOVl6f GSCGRVTP GMLAEGYEQDTEFGNTAHLGD NOV16g GSCGRVTPGMLLLTEDFFAFHNHSAEGYEQDTEFGNTAHLGDCHEILPTS NOV16 GSCGRVTPGMLLLTEDFFAFHNHSAEGYEQDTEFGNTAHLGDCHEILPTS NOV16i GSCGRVTPGMLLLTEDFFAFHNHSAEGYEQDTEFGNTAHLGDCHEILPTS NOVl6j GSCGRVTPGMLLLTEDFFAFHNHSAEGYEQDTEFGNTAHLGDCHEILPTS NOVl6k GSCGRVTP GMLAEGYEQDTEFGNTAHLGDCHEILPTSTTPDYKIFGG NOVl61 GSCGRVTPGMLLLTEDFFAFHNHSAEGYEQDTEFGNTAHLGDCHEILPTS
NOVl6a NOVl6b NOVl6c -VGSLKAICYLLLFLKSTLFTLSHVSISG NOV16d CHG NOVl6e NOVl6f CHG NOV16g -TTPDYKIFGGPTSG NOV16h -TTPDYKIFGGPTSG NOVl6i --TTP NOVl6j --TTP NOV16k PTSDMEVSSSSVTTMASIPEITPKVIDT RPKLTSSRKFWQDDPNTSDFTDPLSGIPKG
NOVl61 -TTP-
NOVl6a NOVl6b NOVl6c VRWEVQSGESNQMVHMNVLITCVFAAFVLGAFIAGVAVYCYRDMFVRKNRKIHKDAESAQ NOVl6d VRWEVQSGESNQMVHMNLE NOVl6e NOVl6f VRWEVQSGESNQMVHMN NOV16g VRWEVQSGESNQMVHMNVLITCVFAAFVLGAFIAGVAVYCYRDMFVRKNRKIHKDAESAQ NOV16 VRWEVQSGESNQMVHMNVLITCVFAAFVLGAFIAGVAVYCYRDMFVRKNRKIHKDAESAQ NOVl6i -DYKIFGGPTSDMEVSSSSVTTMAS IPEITPKVIDT RPKLTSSRKFWQDDPNTS NOV16J -DYKIFGGPTSDMEVSSSSVTTMAS 1PEITPKVIDT RPKLTSSRKFWQDDPNTS NOVl6k VR EVQSGESNQMVHMNVLITCVFAAFVLGAFIAGVAVYCYRDMFVRKNRKIHKDAESAQ NOVl61 -DYKIFGGPTSDMEVSSSSVTTMAS IPEITPKVIDTWRPKLTSSRKFWQDDPNTS
NOVl6a NOVl6b NOVl6c SCTDSSGSFAKLNGLFDSPVKEYQQNIDSPKLYSNLLTSRKELPPNGDTKSMVMDHRGQP NOV16d NOVl6e NOVl6f NOV16g SCTDSSGSFAKLNGLFDSPVKEYQQNIDSPKLYSNLLTSRKEHEFSGR NOV16h SCTDSSGSFAKLNGLFDSPVKEYQQNIDSPKLYSNLLTSRKELPPNGDSKSMVMDHRGQP NOVl6i DFTDP LSGIPKGVRWEVQSGESNQMVHMNVLITCVFAAFVLGAFIAGVAVYCYR NOVl6j DFTDP LSGIPKGVRWEVQSGESNQMVHMNVLITCVFAAFVLGAFIAGVAVYCYR NOVl6k SCTDSSGSFAKLNGLFDSPVKEYQQNIDSPKLYSNLLTSRKELPPNGDTKSMVMDHRGQP NOVl61 DFTDP LSGIPKGVRWEVQ
NOV16a
NOV16b
NOV16c PELAALPTPESTPVLHQKTLQAMKSHSEKAHGHGASRKETPQFFPSSPPPHSPLSHGHIP
NOV16d
NOV16e NOV16f
NOV16g
NOV16h PELAALPTPESTPVLHQKTLQAMKSHSEKAHGHGASRKETPQFFPSSPPPHSPLSHGHIP
NOV16i DMFVRKNRKIHKDAESAQSCTDSSGSFAKLNGLFDSPVKEYQQNIDSPKLYSNLLTSRKE
NOVl6j DMFVRKNRKIHKDAESAQSCTDSSGSFAKLNGLFDSPVKEYQQNIDSPKLYSNLLTSRKE
NOV16k PELAALPTPESTPVLHQKTLQAMKSHSEKAHGHGASRKETPQFFPSSPPPHSPLSHGHIP
NOV161
NOV16a
NOVlδb
NOV16C SAIVLPNATHDYNTSFSNSNAHKAEKKLQNIDHPLTKSSSKRDHRRSVDSRNTLNDLLKH
NOV16d
NOVlδe
NOV16f
NOV16g
NOVl6h SAIVLPNATHDYNTSFSNSNAHKAEKKLQNIDHPLTKSSSKRDHRRSVDSRNTLNDLLKH
NOV16i HEFSGR
NOV16J LPPNGDTKSMVMDHRGQPPELAALPTPESTPVLHQKTLQAMKSHSEKAHGHGASRKETPQ
NOVl6k SAIVLPNATHDYNTSFSNSNAHKAEKKLQNIDHPLTKSSSKRDHRRSVDSRNTLNDLLKH
NOV161
NOV16a
NOV16b
NOVl6c LNDPNSNPKAIMGDIQMAHQNLMLDPMGSMSEVPPKVPNREASLYSPPSTLPRNSPTKRV
NOV16d
NOV16e
NOV16f
NOV16g
NOV16h LNDPNSNPKAIMGDIQMAHQNLMLDPMGSMSEVPPKVPNREASLYSPPSTLPRNSPTKRV
NOVlδi
NOVl6j FFPSSPPPHSPLSHGHIPSAIVLPNATHDYNTSFSNSNAHKAEKKLQNIDHPLTKSSSKR
NOV16k LNDPNSNPKAIMGDIQMAHQNLMLDPMGSMSEVPPKVPNREASLYSPPSTLPRNSPTKRV
NOV161
NOV16a
NOV16b
NOVl6c DVPTTPGVPMTSLERQRGYHKNSSQRHSISAMPKNLNSPNGVLLSRQPSMNRGGYMPTPT
NOV16d
NOV16e
NOV16f
NOV16g
NOV16h DVPTTPGVPMTSLERQRGYHKNSSQRHSISAMPKNLNSPNGVLLSRQPSMNRGGYMPTPT
NOV16i
NOV16 DHRRSVDSRNTLNDLLKHLNDPNSNPKAIMGDIQMAHQNLMLDPMGSMSEVPPKVPNREA
NOVlδk DVPTTPGVPMTSLERQRGYHKNSSQRHSISAMPKNLNSPNGVLLSRQPSMNRGGYMPTPT
NOV161
NOV16a
NOV16b
NOV16C GAKVDYIQGTPVSVHLQPSLSRQSSYTSNGTLPRTGLKRTPSLKPDVPPKPSFVPQTPSV
NOV16d
NOV16e
NOV16f
NOV16g
NOV16h GAKVDYIQGTPVSVHLQPSLSRQSSYTSNGTLPRTGLKRTPSLKPDVPPKPSFVPQTPSV
NOV16i
NOV16J SLYSPPSTLPRNSPTKRVDVPTTPGVPMTSLERQRGYHKNSSQRHSISAMPKNLNSPNGV
NOV16k GAKVDYIQGTPVSVHLQPSLSRQSSYTSNGTLPRTGLKRTPSLKPDVPPKPSFVPQTPSV
NOV161 NOV16a
NOV16b
NOV16C RPLNKYTY
NOV16d
NOV16e
NOV16f
NOV16g
NOVlδh RPLNKYTY
NOV16i
NOV16J LLSRQPSMNRGGYMPTPTGAKVDYIQGTPVSVHLQPSLSRQSSYTSNGTLPRTGLKRTPS
NOVlδk RPLNKYTY
NOV161
NOV16a
NOVlδb
NOV16C
NOV16d
NOV16e
NOV16f
NOVlδg
NOVlδh
NOVlδi
NOV16J LKPDVPPKPSFVPQTPSVRPLNKYTY
NOV16k ,
NOV161
NOVl6a (SEQ ID NO 196)
NOVl6b (SEQ ID NO 198)
NOVl6c (SEQ ID NO 200)
NOVl6d (SEQ ID NO 202)
NOVl6e (SEQ ID NO 204)
NOVl6f (SEQ ID NO 206)
NOVl6g (SEQ ID NO 208)
NOV16h (SEQ ID NO 210)
NOVl6i (SEQ ID NO 212)
NOVl6j (SEQ ID NO 214)
NOVlδk (SEQ ID NO 216)
NOVl61 (SEQ ID NO 218)
Further analysis of the NOVl 6a protein yielded the following properties shown in Table 16C.
Table 16C. Protein Sequence Properties NOVl 6a
SignalP analysis: Cleavage site between residues 21 and 22
PSORT II analysis:
PSG: a new signal peptide prediction method
N-region: length 2; pos.chg 1; neg.chg 0 H-region: length 16; peak value 9.62 PSG score: 5.22
GvH: von Heijne's method for signal seq. recognition GvH score (threshold: -2.1): -0.82 possible cleavage site: between 20 and 21
>>> Seems to have a cleavable signal peptide (1 to 20)
ALOM: Klein et al's method for TM region allocation Init position for calculation: 21
Tentative number of TMS(s) for the threshold 0.5: 1 Number of TMS(s) for threshold 0.5: 0 PERIPHERAL Likelihood = 1.75 (at 300) ALOM score: -0.32 (number of TMSs: 0)
MTOP: Prediction of membrane topology (Hartmann et al.) Center position for calculation: 10 Charge difference: -5.0 C(-3.0) - N( 2.0) N >= C: N-terminal side will be inside
MITDISC: discrimination of mitochondrial targeting seq R content: 2 Hyd Moment (75): 6.62 Hyd Momen (95) : 8.11 G content: 0 D/E content: 1 S/T content: 2 Score: -2.26
Gavel : prediction of cleavage sites for mitochondrial preseq R-2 motif at 29 LRA|VS
NUCDISC: discrimination of nuclear localization signals pat4 : none pat7 : none bipartite: none content of basic residues: 12.0% NLS Score: -0.47
KDEL: ER retention motif in the C-terminus : none
ER Membrane Retention Signals:
XXRR-like motif in the N-terminus : RVFL none
SKL: peroxisomal targeting signal in the C-terminus : none
PTS2 : 2nd peroxisomal targeting signal : none
VAC: possible vacuolar targeting motif: none
RNA-binding motif: none
Actinin-type actin-binding motif: type 1 : none type : none
NMYR: N-myristoylation pattern : none
Prenylation motif : none memYQRL: transport motif from cell surface to Golgi: none
Tyrosines in the tail : none
Dileucine motif in the tail: none checking 63 PROSITE DNA binding motifs: none checking 71 PROSITE ribosomal protein motifs: none checking 33 PROSITE prokaryotic DNA binding motifs: none NNCN: Reinhardt's method for Cytoplasmic/Nuclear discrimination
Prediction: cytoplasmic
Reliability: 89
COIL: Lupas ' s algorithm to detect coiled-coil regions total : 0 residues
Final Results (k = 9/23) :
21.7 %: mitochondrial
21.7 %: endoplasmic reticulum
17.4 %: extracellular, including cell wall
13.0 %: vacuolar
8.7 %.- cytoplasmic
8.7 %: Golgi
8.7 % : nuclear
» prediction for CG59253-01 is mit (k=23)
A search of the NOVl 6a protein against the Geneseq database, a proprietary database that contains sequences published in patents and patent publication, yielded several homologous proteins shown in Table 16D.
Figure imgf000268_0001
In a BLAST search of public sequence databases, the NOV16a protein was found to have homology to the proteins shown in the BLASTP data in Table 16E.
Figure imgf000269_0001
PFam analysis predicts that the NOVl 6a protein contains the domains shown in the Table 16F.
Figure imgf000269_0002
Example 17. The NOVl 7 clone was analyzed, and the nucleotide and encoded polypeptide sequences are shown in Table 17A. Table 17A. NOV17 Sequence Analysis
NOV17a, CG95430-02 SEQ ID NO: 229 954 bp DNA Sequence ORF Start: at 7 lORF Stop: at 949
GGATCCCAGGACACCTGCAGGCAAGGGCACCCTGGGATCCCTGGGAACCCCGGTCACAATGGTCTGCCTGGAA
GAGATGGACGAGACGGAGCGAAGGGTGACAAAGGCGATGCAGGAGAACCAGGACGTCCTGGCAGCCCGGGGAA GGATGGGACGAGTGGAGAGAAGGGAGAACGAGGAGCAGATGGAAAAGTTGAAGCAAAAGGCATCAAAGGTGAT CAAGGCTCAAGAGGATCCCCAGGAAAACATGGCCCCAAGGGGCTTGCAGGGCCCATGGGAGAGAAAGGCCTCC GAGGAGAGACTGGGCCTCAGGGGCAGAAGGGGAATAAGGGTGACGTGGGTCCCACTGGTCCTGAGGGGCCAAG GGGCAACATTGGGCCTTTGGGCCCAACTGGTTTACCGGGCCCCATGGGCCCTATTGGAAAGCCTGGTCCCAAG GGAGAAGCTGGACCCACGGGGCCCCAGGGTGAGCCAGGAGTCCGGGGAATAAGAGGCTGGAAAGGAGATCGAG GAGAGAAAGGGAAAATCGGTGAGACTCTAGTCTTGCCAAAAAGTGCTTTCACTGTGGGGCTCACGGTGCTGAG CAAGTTTCCTTCTTCAGATGTGCCCATTAAATTTGATAAGATCCTGTATAACGAATTCAACCATTATGATACA GCAGCGGGGAAATTCACGTGCCACATTGCTGGGGTCTATTACTTCACCTACCACATCACTGTTTTCTCCAGGA ATGTTCAGGTGTCTTTGGTCAAAAATGGAGTAAAAATACTGCACACCAAAGATGCTTACATGAGCTCTGAGGA CCAGGCCTCTGGCGGCATTGTCCTGCAGCTGAAGCTCGGGGATGAGGTGTGGCTGCAGGTGACAGGAGGAGAG AGGTTCAATGGCTTGTTTGCTGATGAGGACGATGACACAACTTTCACAGGGTTCCTTCTGTTCAGCAGCCCGC TCGAG
NOVl 7a, CG95430-02 SEQ ID NO: 230 1314 aa MW at 32420.0kD Protein Sequence
QDTCRQGHPGIPGNPGHNGLPGRDGRDGAKGDKGDAGEPGRPGSPGKDGTSGEKGERGADGKVEAKGIKGDQG SRGSPGKHGPKGLAGPMGEKGLRGETGPQGQKGNKGDVGPTGPEGPRGNIGPLGPTGLPGPMGPIGKPGPKGE AGPTGPQGEPGVRGIRGWKGDRGEKGKIGETLVLPKSAFTVGLTVLSKFPSSDVPIKFDKILYNEFNHYDTAA GKFTCHIAGVYYFTYHITVFSRNVQVSLVKNGVKILHTKDAYMSSEDQASGGIVLQLKLGDEV LQVTGGERF NGLFADEDDDTTFTGFLLFSSP
Figure imgf000270_0001
Figure imgf000271_0001
NOV17c, CG95430-01 }SEQ IDNO: 233 818 bp
DNA Sequence ORF Start: ATG at 35 Iθ l stop7τG at 728
TCCCTCTTTCAGTTCAGAGTCTGTCATCTGAACCATGAGGATCTGGTGGCTTCTGCTTGCCATTGAAATCTGC
ACAGGGAACATAAACTCACAGGACACCTGCAGGCAAGGGCACCCTGGAATCCCTGGGAACCCCGGTCACAATG GTCTGCCTGGAAGAGATGGACGAGACGGAGCGAAGGGTGACAAAGGCGATGCAGGTAAGCCTGGTCCCAAAGG AGAAGCTGGACCCACGGGGCCCCAGGGTGAGCCAGGAGTCCGGGGAATAAGAGGCTGGAAAGGAGATCGAGGA GAGAAAGGGAAAATCGGTGAGACTCTAGTCTTGCCAAAAAGTGCTTTCACTGTGGGGCTCACGGTGCTGAGCA AGTTTCCTTCTTCAGATATGCCCATTAAATTTGATAAGATCCTGTATAACGAATTCAACCATTATGATACAGC AGCGGGGAAATTCACGTGCCACATTGCTGGGGTCTATTACTTCACCTACCACATCACTGTTTTCTCCAGAAAT GTTCAGGTGTCTTTGGTCAAAAATGGAGTAAAAATACTGCACACCAAAGATGCTTACATGAGCTCTGAGGACC AGGCCTCTGGCGGCATTGTCCTGCAGCTGAAGCTCGGGGATGAGGTGTGGCTGCAGGTGACAGGAGGAGAGAG GTTCAATGGCTTGTTTGCTGATGAGGACGATGACACAACTTTCACAGGGTTCCTTCTGTTCAGCAGCCCGTGA CAGAGGAGAGTTTAAAAATCCGCCACACCATCCATCAGAATCAGCTTGGGATGAACTTATTCAGATGGTTTTA
CTTTATTAATTCCTC
NOV17c, CG95430-01 SEQ ID NO: 234 231 aa MW at 24946.0kD Protein Sequence
MRIWWLLLAIEICTGNINSQDTCRQGHPGIPGNPGHNGLPGRDGRDGAKGDKGDAGKPGPKGEAGPTGPQGEP GVRGIRG KGDRGEKGKIGETLVLPKSAFTVGLTVLSKFPSSDMPIKFDKILYNEFNHYDTAAGKFTCHIAGV YYFTYHITVFSRNVQVSLVKNGVKILHTKDAYMSSEDQASGGIVLQLKLGDEVLQVTGGERFNGLFADEDDD TTFTGFLLFSβP
NOV17d, 319194717 )SEQ IDNO: 235 jl024bp
DNA Sequence ORF Start: at 2 fORF Stop: TGA at 1013
CACCGGATCCACCATGAGGATCTGGTGGTTTCTGCTTGCCATTGAAATCTGCACAGGGAACATAAACTCTCAG GACACCTGCAGGCAAGGGCACCCTGGAATCCCTGGGAACCCCGGTCACAATGGTCTGCCTGGAAGAGATGGAC GAGACGGAGCGAAGGGTGACAAAGGCGATGCAGGAGAACCAGGACGTCCTGGCAGCCCGGGGAAGGATGGGAC GAGTGGAGAGAAGGGAGAACGAGGAGCAGATGGAAAAGTTGAAGCAAAAGGCATCAAAGGTGATCAAGGCTCA AGAGGATCCCCAGGAAAACATGGCCCCAAGGGGCTTGCAGGGCCCATGGGAGAGAAAGGCCTCCGAGGAGAGA CTGGGCCTCAGGGGCAGAAGGGGAATAAGGGTGACGTGGGTCCCACTGGTCCTGAGGGGCCAAGGGGCAACAT TGGGCCTTTGGGCCCAACTGGTTTACCGGGCCCCATGGGCCCTATTGGAAAGCCTGGTCCCAAGGGAGAAGCT GGACCCACGGGGCCCCAGGGTGAGCCAGGAGTCCGGGGAATAAGAGGCTGGAAAGGAGATCGAGGAGAGAAAG GGAAAATCGGTGAGACTCTAGTCTTGCCAAAAAGTGCTTTCACTGTGGGGCTCACGGTGCTGAGCAAGTTTCC TTCTTCAGATGTGCCCATTAAATTTGATAAGATCCTGTATAACGAATTCAACCATTATGATACAGCAGCGGGG AAATTCACGTGCCACATTGCTGGGGTCTATTACTTCACCTACCACATCACTGTTTTCTCCAGGAATGTTCAGG TGTCTTTGGTCAAAAATGGAGTAAAAATACTGCACACCAAAGATGCTTACATGAGCTCTGAGGACCAGGCCTC TGGCGGCATTGTCCTGCAGCTGAAGCTCGGGGATGAGGTGTGGCTGCAGGTGACAGGAGGAGAGAAGTTCAAT GGCTTGTTTGCTGATGAGGACGATGACACAACTTTCACAGGGTTCCTTCTGTTCAGCAGCCCGTGAGTCGACG GC NOV17d, 319194717 SEQ ID NO: 236 337 aa MW at 35001.0kD Protein Sequence
TGSTMRIW FLLAIEICTGNINSQDTCRQGHPGIPGNPGHNGLPGRDGRDGAKGDKGDAGEPGRPGSPGKDGT SGEKGERGADGKVEAKGIKGDQGSRGSPGKHGPKGLAGPMGEKGLRGETGPQGQKGNKGDVGPTGPEGPRGNI GPLGPTGLPGPMGPIGKPGPKGEAGPTGPQGEPGVRGIRG KGDRGEKGKIGETLVLPKSAFTVGLTVLSKFP SSDVPIKFDKILYNEFNHYDTAAGKFTCHIAGVYYFTYHITVFSRNVQVSLVKNGVKILHTKDAYMSSEDQAS GGIVLQLKLGDEVLQVTGGEKFNGLFADEDDDTTFTGFLLFSSP
Figure imgf000272_0002
OV17e, CG95430-03 SEQ ID NO: 238 131 aa MW at 14607.4kD Protein Sequence
AFTVGLTVLSKFPSSDMPIKFDKILYNEFNHYDTAAGKFTCHIAGVYYFTYHITVFSRNVQVSLVKNGVKILH TKDAYMSSEDQASGGIVLQLKLGDEVWLQVTGGERFNGLFADEDDDTTFTGFLLFSSP
Figure imgf000272_0001
NOV17f, CG95430-05 SEQ ID NO: 240 306 aa MW at 31546.2kD Protein Sequence
MRIW LLLAIEICTGNINSQDTCRQGHPGIPGNPGHNGLPGRDGRDGAKGDKGDAGEPGRPGSPGKDGTSGEK GERGADGKVEAKGIKGDQGSRGSPGKHGPKGLAGPMGEKGLRGETGPQGQKGNKGDVGPTGPEGPRGNIGPLG PTGLPGPMGPIGKPGPKGEAGPTGPQGEPGVRGIRGWKGDRGEKGKIGETLVLPKSAFTVGLTVLSKFPSSDV PIKFDKIHITVFSRNVQVSLVKNGVKILHTRDAYVSSEDQASGSIVLQLKLGDEMWLQVTGGERFNGLFADED DDTTFTGFLLFSSQ
NOV17g, CG95430-06 SEQ ID NO: 241 889 bp DNA Sequence ORF Start: ATG at 16 ORF Stop: TGA at 880
ITCTGTCATCTGAACCATGAGGATCTGGTGGTTTCTGCTTGCCATTGAAATCTGCACAGGGAACATAAACTCAC
AGGACACCTGCAGGCAAGGGCACCCTGGCATCCCTGGGAACCCCGGTCACAATGGTCTGTCTGGAAGAGATGG ACGAGACGGAGCGAAGGGTGACAAAGGCGATGCAGGAGAACCAGGACGTCCTGGCAGCCCGGGGAAGGATGGG ACGAGTGGAGAGAAGGGAGAACGAGGAGCAGATGGAAAAGTTGAAGCAAAAGGCATCAAAGGTGATCAAGGCT CAAGAGGATCCCCAGGAAAACATGGCCCCAAGGGGCTTGCAGGGCCCATGGGAGAGAAAGGCCTCCGAGGAGA GACTGGGCCTCAGGGGCAGAAGGGGAATAAGGGTGAGCCAGGAGTCCGGGGAATAAGAGGCTGGAAAGGAGAT CGAGGAGAGAAAGGGAAAATCGGTGAGACTCTAGTCTTGCCAAAAAGTGCTTTCACTGTGGGGCTCACGGTGC TGAGCAAGTTTCCTTCTTCAGATGTGCCCATTAAATTTGATAAGATCCTGTATAACGAATTCAACCATTATGA TACAGCAGCGGGGAAATTCACGTGCCACATTGCTGGGGTCTATTACTTCACCTACCACATCGCTGTTTTCTCC AGCAATGTTCAGGTGTCTTTGGTCAAAAATGGAGTAAAAATACTGCACACCAAAGATGCTTACATGAGCTCTG AGGACCAGGCCTCTGGCGGCATTGTCCTGCAGCTGAAGCTCGGGGATGAGGTGTGGCTGCAGGTGACAGGAGG AGAGAGGTTCAATGGCTTGTTTGCTGATGAGGACGATGACACAACTTTCACAGGGTTCCTTCTGTTCAGCAGC CAGTGACAGAGGA
NOV17g, CG95430-06 SEQ ID NO: 242 288 aa MW at 30497.9kD Protein Sequence RIW FLLAIEICTGNINSQDTCRQGHPGIPGNPGHNGLSGRDGRDGAKGDKGDAGEPGRPGSPGKDGTSGEK GERGADGKVEAKGIKGDQGSRGSPGKHGPKGLAGPMGEKGLRGETGPQGQKGNKGEPGVRGIRG KGDRGEKG KIGETLVLPKSAFTVGLTVLSKFPSSDVPIKFDKILYNEFNHYDTAAGKFTCHIAGVYYFTYHIAVFSSNVQV SLVKNGVKILHTKDAYMSSEDQASGGIVLQLKLGDEV LQVTGGERFNGLFADEDDDTTFTGFLLFSSQ
Figure imgf000273_0001
Figure imgf000274_0001
NOV17i, CG95430-08 SEQ ID NO: 246 333 aa MW at 34654.7kD
Protein Sequence I
MRIW FLLAIEICTGNINSQDTCRQGHPGIPGNPGHNGLPGRDGRDGAKGDKGDAGEPGRPGSPGKDGTSGEK GERGADGKVEAKGIKGDQGSRGSPGKHGPKGLAGPMGEKGLRGETGPQGQKGNKGDVGPTGPEGPRGNIGPLG PTGLPGPMGPIGKPGPKGEAGPTGPQGEPGVRGIRGWKGDRGEKGKIGETLVLPKSAFTVGLTVLSKFPSSDV PIKFDKILYNEFNHYDTAAGKFTCHIAGVYYFTYHITVFSRNVQVSLVKNGVKILHTKDAYMSSEDQASGGIV ILQLKLGDEVWLQVTGGEKFNGLFADEDDDTTFTGFLLFSSP
NOV17J, CG95430-09 SEQ ID NO: 247 |964bp
DNA Sequence ORF Start^UΪ^
CACCAGATCTCAGGACACCTGCAGGCAAGGGCACCCTGGGATCCCTGGGAACCCCGGTCACAATGGTCTGCCT
GGAAGAGATGGACGAGACGGAGCGAAGGGTGACAAAGGCGATGCAGGAGAACCAGGACGTCCTGGCAGCCCGG GGAAGGATGGGACGAGTGGAGAGAAGGGAGAACGAGGAGCAGATGGAAAAGTTGAAGCAAAAGGCATCAAAGG TGATCAAGGCTCAAGAGGATCCCCAGGAAAACATGGCCCCAAGGGGCTTGCAGGGCCCATGGGAGAGAAAGGC CTCCGAGGAGAGACTGGGCCTCAGGGGCAGAAGGGGAATAAGGGTGACGTGGGTCCCACTGGTCCTGAGGGGC CAAGGGGCAACATTGGGCCTTTGGGCCCAACTGGTTTACCGGGCCCCATGGGCCCTATTGGAAAGCCTGGTCC CAAGGGAGAAGCTGGACCCACGGGGCCCCAGGGTGAGCCAGGAGTCCGGGGAATAAGAGGCTGGAAAGGAGAT CGAGGAGAGAAAGGGAAAATCGGTGAGACTCTAGTCTTGCCAAAAAGTGCTTTCACTGTGGGGCTCACGGTGC TGAGCAAGTTTCCTTCTTCAGATGTGCCCATTAAATTTGATAAGATCCTGTATAACGAATTCAACCATTATGA TACAGCAGCGGGGAAATTCACGTGCCACATTGCTGGGGTCTATTACTTCACCTACCACATCACTGTTTTCTCC AGGAATGTTCAGGTGTCTTTGGTCAAAAATGGAGTAAAAATACTGCACACCAAAGATGCTTACATGAGCTCTG AGGACCAGGCCTCTGGCGGCATTGTCCTGCAGCTGAAGCTCGGGGATGAGGTGTGGCTGCAGGTGACAGGAGG AGAGAGGTTCAATGGCTTGTTTGCTGATGAGGACGATGACACAACTTTCACAGGGTTCCTTCTGTTCAGCAGC CCGTAGGTCGACGGC
NOV17J, CG95430-09 SEQ ID NO: 248 314 aa MW at 32420.0kD Protein Sequence
QDTCRQGHPGIPGNPGHNGLPGRDGRDGAKGDKGDAGEPGRPGSPGKDGTSGEKGERGADGKVEAKGIKGDQG SRGSPGKHGPKGLAGPMGEKGLRGETGPQGQKGNKGDVGPTGPEGPRGNIGPLGPTGLPGPMGPIGKPGPKGE AGPTGPQGEPGVRGIRG KGDRGEKGKIGETLVLPKSAFTVGLTVLSKFPSSDVPIKFDKILYNEFNHYDTAA GKFTCHIAGVYYFTYHITVFSRNVQVSLVKNGVKILHTKDAYMSSEDQASGGIVLQLKLGDEV LQVTGGERF NGLFADEDDDTTFTGFLLFSSP
^V17k, CG95430 0 JSEQ ID NO: 249 " IT"^*11*2^"""" ~
[DNA Sequence fORF Start: ATG at 17 ORF Stop: at 1016
CACCAGATCTCCCACCATGAGGATCTGGTGGTTTCTGCTTGCCATTGAAATCTGCACAGGGAACATAAACTCT
CAGGACACCTGCAGGCAAGGGCACCCTGGAATCCCTGGGAACCCCGGTCACAATGGTCTGCCTGGAAGAGATG GACGAGACGGAGCGAAGGGTGACAAAGGCGATGCAGGAGAACCAGGACGTCCTGGCAGCCCGGGGAAGGATGG GACGAGTGGAGAGAAGGGAGAACGAGGAGCAGATGGAAAAGTTGAAGCAAAAGGCATCAAAGGTGATCAAGGC TCAAGAGGATCCCCAGGAAAACATGGCCCCAAGGGGCTTGCAGGGCCCATGGGAGAGAAAGGCCTCCGAGGAG AGACTGGGCCTCAGGGGCAGAAGGGGAATAAGGGTGACGTGGGTCCCACTGGTCCTGAGGGGCCAAGGGGCAA CATTGGGCCTTTGGGCCCAACTGGTTTACCGGGCCCCATGGGCCCTATTGGAAAGCCTGGTCCCAAGGGAGAA GCTGGACCCACGGGGCCCCAGGGTGAGCCAGGAGTCCGGGGAATAAGAGGGTGGAAAGGAGATCGAGGAGAGA AAGGGAAAATCGGTGAGACTCTAGTCTTGCCAAAAAGTGCTTTCACTGTGGGGCTCACGGTGCTGAGCAAGTT TCCTTCTTCAGATGTGCCCATTAAATTTGATAAGATCCTGTATAACGAATTCAACCATTATGATACAGCAGCG GGGAAATTCACGTGCCACATTGCTGGGGTCTATTACTTCACCTACCACATCACTGTTTTCTCCAGGAATGTTC AGGTGTCTTTGGTCAAAAATGGAGTAAAAATACTGCACACCAAAGATGCTTACATGAGCTCTGAGGACCAGGC CTCTGGCGGCATTGTCCTGCAGCTGAAGCTCGGGGATGAGGTGTGGCTGCAGGTGACAGGAGGAGAGAAGTTC AATGGCTTGTTTGCTGATGAGGACGATGACACAACTTTCACAGGGTTCCTTCTGTTCAGCAGCCCGCTCGAGG GC
Figure imgf000276_0001
NOV171, CG95430-11 SEQ ID NO: 251 1045 bp DNA Sequence ORF Start: ATG at 14 ORF Stop: TAA at 1034
CACCAGATCTACCATGGGCCACCATCACCACCATCACAGGATCTGGTGGTTTCTGCTTGCCATTGAAATCTGC
ACAGGGAACATAAACTCTCAGGACACCTGCAGGCAAGGGCACCCTGGAATCCCTGGGAACCCCGGTCACAATG GTCTGCCTGGAAGAGATGGACGAGACGGAGCGAAGGGTGACAAAGGCGATGCAGGAGAACCAGGACGTCCTGG CAGCCCGGGGAAGGATGGGACGAGTGGAGAGAAGGGAGAACGAGGAGCAGATGGAAAAGTTGAAGCAAAAGGC ATCAAAGGTGATCAAGGCTCAAGAGGATCCCCAGGAAAACATGGCCCCAAGGGGCTTGCAGGGCCCATGGGAG AGAAAGGCCTCCGAGGAGAGACTGGGCCTCAGGGGCAGAAGGGGAATAAGGGTGACGTGGGTCCCACTGGTCC TGAGGGGCCAAGGGGCAACATTGGGCCTTTGGGCCCAACTGGTTTACCGGGCCCCATGGGCCCTATTGGAAAG CCTGGTCCCAAGGGAGAAGCTGGACCCACGGGGCCCCAGGGTGAGCCAGGAGTCCGGGGAATAAGAGGCTGGA AAGGAGATCGAGGAGAGAAAGGGAAAATCGGTGAGACTCTAGTCTTGCCAAAAAGTGCTTTCACTGTGGGGCT CACGGTGCTGAGCAAGTTTCCTTCTTCAGATGTGCCCATTAAATTTGATAAGATCCTGTATAACGAATTCAAC CATTATGATACAGCAGCGGGGAAATTCACGTGCCACATTGCTGGGGTCTATTACTTCACCTACCACATCACTG TTTTCTCCAGGAATGTTCAGGTGTCTTTGGTCAAAAATGGAGTAAAAATACTGCACACCAAAGATGCTTACAT GAGCTCTGAGGACCAGGCCTCTGGCGGCATTGTCCTGCAGCTGAAGCTCGGGGATGAGGTGTGGCTGCAGGTG ACAGGAGGAGAGAAGTTCAATGGCTTGTTTGCTGATGAGGACGATGACACAACTTTCACAGGGTTCCTTCTGT TCAGCAGCCCGTAACTCGAGGGC
NOV17L CG95430-11 SEQ ID NO: 252 340 aa MW at 35534.6kD Protein Sequence
MGHHHHHHRI FLLAIEICTGNINSQDTCRQGHPGIPGNPGHNGLPGRDGRDGAKGDKGDAGEPGRPGSPGK IDGTSGEKGERGADGKVEAKGIKGDQGSRGSPGKHGPKGLAGPMGEKGLRGETGPQGQKGNKGDVGPTGPEGPR GNIGPLGPTGLPGP GPIGKPGPKGEAGPTGPQGEPGVRGIRG KGDRGEKGKIGETLVLPKSAFTVGLTVLS KFPSSDVPIKFDKILYNEFNHYDTAAGKFTCHIAGVYYFTYHITVFSRNVQVSLVKNGVKILHTKDAYMSSED QASGGIVLQLKLGDEV LQVTGGEKFNGLFADEDDDTTFTGFLLFSSP
Figure imgf000277_0001
NOVl 7m, CG95430-12 jSEQ ID NO: 254 320 aa !MW at 33214.9kD
Protein Sequence
HHHHHHQDTCRQGHPGIPGNPGHNGLPGRDGRDGAKGDKGDAGEPGRPGSPGKDGTSGEKGERGADGKVEAKG IKGDQGSRGSPGKHGPKGLAGPMGEKGLRGETGPQGQKGNKGDVGPTGPEGPRGNIGPLGPTGLPGPMGPIGK PGPKGEAGPTGPQGEPG GIRGWKGDRGEKGKIGETLVLPKSAFTVGLTVLSKFPSSDVPIKFDKILYNEFN HYDTAAGKFTCHIAGVYYFTYHITVFSRNVQVSLVKNGVKILHTKDAYMSSEDQASGGIVLQLKLGDEV LQV TGGEKFNGLFADEDDDTTFTGFLLFSSP
NOV17n, CG95430-13 SEQ ID NO: 255 982 bp
DNA Sequence F)RF Start: at 11 ORF Stop: TAA at 971
CACCAGATCTCACCATCACCACCATCACCAGGACACCTGCAGGCAAGGGCACCCTGGAATCCCTGGGAACCCC
GGTCACAATGGTCTGCCTGGAAGAGATGGACGAGACGGAGCGAAGGGTGACAAAGGCGATGCAGGAGAACCAG GACGTCCTGGCAGCCCGGGGAAGGATGGGACGAGTGGAGAGAAGGGAGAACGAGGAGCAGATGGAAAAGTTGA AGCAAAAGGCATCAAAGGTGATCAAGGCTCAAGAGGATCCCCAGGAAAACATGGCCCCAAGGGGCTTGCAGGG CCCATGGGAGAGAAAGGCCTCCGAGGAGAGACTGGGCCTCAGGGGCAGAAGGGGAATAAGGGTGACGTGGGTC CCACTGGTCCTGAGGGGCCAAGGGGCAACATTGGGCCTTTGGGCCCAACTGGTTTACCGGGCCCCATGGGCCC TATTGGAAAGCCTGGTCCCAAGGGAGAAGCTGGACCCACGGGGCCCCAGGGTGAGCCAGGAGTCCGGGGAATA AGAGGCTGGAAAGGAGATCGAGGAGAGAAAGGGAAAATCGGTGAGACTCTAGTCTTGCCAAAAAGTGCTTTCA CTGTGGGGCTCACGGTGCTGAGCAAGTTTCCTTCTTCAGATGTGCCCATTAAATTTGATAAGATCCTGTATAA CGAATTCAACCATTATGATACAGCAGCGGGGAAATTCACGTGCCACATTGCTGGGGTCTATTACTTCACCTAC CACATCACTGTTTTCTCCAGGAATGTTCAGGTGTCTTTGGTCAAAAATGGAGTAAAAATACTGCACACCAAAG ATGCTTACATGAGCTCTGAGGACCAGGCCTCTGGCGGCATTGTCCTGCAGCTGAAGCTCGGGGATGAGGTGTG GCTGCAGGTGACAGGAGGAGAGAGGTTCAATGGCTTGTTTGCTGATGAGGACGATGACACAACTTTCACAGGG TTCCTTCTGTTCAGCAGCCCGTAACTCGAGGGC NOV17n, CG95430-13 SEQ ID NO: 256 320 aa MW at 33242.9kD
Protein Sequence | \ j
HHHHHHQDTCRQGHPGIPGNPGHNGLPGRDGRDGAKGDKGDAGEPGRPGSPGKDGTSGEKGERGADGKVEAKG IKGDQGSRGSPGKHGPKGLAGPMGEKGLRGETGPQGQKGNKGDVGPTGPEGPRGNIGPLGPTGLPGP GPIGK PGPKGEAGPTGPQGEPGVRGIRGWKGDRGEKGKIGETLVLPKSAFTVGLTVLSKFPSSDVPIKFDKILYNEFN HYDTAAGKFTCHIAGVYYFTYHITVFSRNVQVSLVKNGVKILHTKDAYMSSEDQASGGIVLQLKLGDEVWLQV TGGERFNGLFADEDDDTTFTGFLLFSSP
NOV17o, SNP13379412 of SEQ ID NO: 257 818 bp CG95430-01, DNA Sequence ORF Start: ATG at 35 |ORF Stop: TGA at 728
TCCCTCTTTCAGTTCAGAGTCTGTCATCTGAACCATGAGGATCTGGTGGTTTCTGCTTGCCATTGAAATCTGC
ACAGGGAACATAAACTCACAGGACACCTGCAGGCAAGGGCACCCTGGAATCCCTGGGAACCCCGGTCACAATG GTCTGCCTGGAAGAGATGGACGAGACGGAGCGAAGGGTGACAAAGGCGATGCAGGTAAGCCTGGTCCCAAAGG AGAAGCTGGACCCACGGGGCCCCAGGGTGAGCCAGGAGTCCGGGGAATAAGAGGCTGGAAAGGAGATCGAGGA GAGAAAGGGAAAATCGGTGAGACTCTAGTCTTGCCAAAAAGTGCTTTCACTGTGGGGCTCACGGTGCTGAGCA AGTTTCCTTCTTCAGATATGCCCATTAAATTTGATAAGATCCTGTATAACGAATTCAACCATTATGATACAGC AGCGGGGAAATTCACGTGCCACATTGCTGGGGTCTATTACTTCACCTACCACATCACTGTTTTCTCCAGAAAT GTTCAGGTGTCTTTGGTCAAAAATGGAGTAAAAATACTGCACACCAAAGATGCTTACATGAGCTCTGAGGACC AGGCCTCTGGCGGCATTGTCCTGCAGCTGAAGCTCGGGGATGAGGTGTGGCTGCAGGTGACAGGAGGAGAGAG GTTCAATGGCTTGTTTGCTGATGAGGACGATGACACAACTTTCACAGGGTTCCTTCTGTTCAGCAGCCCGTGA CAGAGGAGAGTTTAAAAATCCGCCACACCATCCATCAGAATCAGCTTGGGATGAACTTATTCAGATGGTTTTA
CTTTATTAATTCCTC
Figure imgf000278_0001
NOV17p, SNP13381828 of SEQ ID NO: 259 818 bp CG95430-01, DNA Sequence ORF Start: ATG at 35 jORF Stop: TGA at 728 SNP Pos: 235 JSNP Change: GIO A"
TCCCTCTTTCAGTTCAGAGTCTGTCATCTGAACCATGAGGATCTGGTGGCTTCTGCTTGCCATTGAAATCTGC
ACAGGGAACATAAACTCACAGGACACCTGCAGGCAAGGGCACCCTGGAATCCCTGGGAACCCCGGTCACAATG GTCTGCCTGGAAGAGATGGACGAGACGGAGCGAAGGGTGACAAAGGCGATGCAGGTAAGCCTGGTCCCAAAGG AGAAGCTGGACCCACAGGGCCCCAGGGTGAGCCAGGAGTCCGGGGAATAAGAGGCTGGAAAGGAGATCGAGGA GAGAAAGGGAAAATCGGTGAGACTCTAGTCTTGCCAAAAAGTGCTTTCACTGTGGGGCTCACGGTGCTGAGCA AGTTTCCTTCTTCAGATATGCCCATTAAATTTGATAAGATCCTGTATAACGAATTCAACCATTATGATACAGC AGCGGGGAAATTCACGTGCCACATTGCTGGGGTCTATTACTTCACCTACCACATCACTGTTTTCTCCAGAAAT GTTCAGGTGTCTTTGGTCAAAAATGGAGTAAAAATACTGCACACCAAAGATGCTTACATGAGCTCTGAGGACC AGGCCTCTGGCGGCATTGTCCTGCAGCTGAAGCTCGGGGATGAGGTGTGGCTGCAGGTGACAGGAGGAGAGAG GTTCAATGGCTTGTTTGCTGATGAGGACGATGACACAACTTTCACAGGGTTCCTTCTGTTCAGCAGCCCGTGA CAGAGGAGAGTTTAAAAATCCGCCACACCATCCATCAGAATCAGCTTGGGATGAACTTATTCAGATGGTTTTA CTTTATTAATTCCTC NOV17p, SNP13381828 of SEQ ΪD N07260 J231 aa )MW at 2494 .0M) CG95430-01, Protein Sequence SNP Pos: 67 T SNP Change: Thr to Thr RIW LLLAIEICTGNINSQDTCRQGHPGIPGNPGHNGLPGRDGRDGAKGDKGDAGKPGPKGEAGPTGPQGEP GVRGIRG KGDRGEKGKIGETLVLPKSAFTVGLTVLSKFPSSDMPIKFDKILYNEFNHYDTAAGKFTCHIAGV YYFTYHITVFSRNVQVSLVKNGVKILHTKDAYMSSEDQASGGIVLQLKLGDEVWLQVTGGERFNGLFADEDDD TTFTGFLLFSSP
NOV17q, SNP13379125 of SEQ ID NO: 261 818 bp CG95430-01, DNA Sequence ORF Start: ATG at 35 ORF Stop: TGA at 728
SNP Pos: 383 SNP Change: A to G
TCCCTCTTTCAGTTCAGAGTCTGTCATCTGAACCATGAGGATCTGGTGGCTTCTGCTTGCCATTGAAATCTGC
ACAGGGAACATAAACTCACAGGACACCTGCAGGCAAGGGCACCCTGGAATCCCTGGGAACCCCGGTCACAATG GTCTGCCTGGAAGAGATGGACGAGACGGAGCGAAGGGTGACAAAGGCGATGCAGGTAAGCCTGGTCCCAAAGG AGAAGCTGGACCCACGGGGCCCCAGGGTGAGCCAGGAGTCCGGGGAATAAGAGGCTGGAAAGGAGATCGAGGA GAGAAAGGGAAAATCGGTGAGACTCTAGTCTTGCCAAAAAGTGCTTTCACTGTGGGGCTCACGGTGCTGAGCA AGTTTCCTTCTTCAGATGTGCCCATTAAATTTGATAAGATCCTGTATAACGAATTCAACCATTATGATACAGC AGCGGGGAAATTCACGTGCCACATTGCTGGGGTCTATTACTTCACCTACCACATCACTGTTTTCTCCAGAAAT GTTCAGGTGTCTTTGGTCAAAAATGGAGTAAAAATACTGCACACCAAAGATGCTTACATGAGCTCTGAGGACC AGGCCTCTGGCGGCATTGTCCTGCAGCTGAAGCTCGGGGATGAGGTGTGGCTGCAGGTGACAGGAGGAGAGAG GTTCAATGGCTTGTTTGCTGATGAGGACGATGACACAACTTTCACAGGGTTCCTTCTGTTCAGCAGCCCGTGA CAGAGGAGAGTTTAAAAATCCGCCACACCATCCATCAGAATCAGCTTGGGATGAACTTATTCAGATGGTTTTA
CTTTATTAATTCCTC
NOV17q, SNP13379125 of SEQ ID NO: 262 231 aa MW at 24913.9kD CG95430-01, Protein Sequence SNP Pos: 117 I SNP Change: Met to Val
MRI WLLLAIEICTGNINSQDTCRQGHPGIPGNPGHNGLPGRDGRDGAKGDKGDAGKPGPKGEAGPTGPQGEP GVRGIRG KGDRGEKGKIGETLVLPKSAFTVGLTVLSKFPSSDVPIKFDKILYNEFNHYDTAAGKFTCHIAGV YYFTYHITVFSRNVQVSLVKNGVKILHTKDAYMSSEDQASGGIVLQLKLGDEVLQVTGGERFNGLFADEDDD TTFTGFLLFSSP
NOV17r, SNP13381827 of SEQ ID NO: 263 818 bp CG95430-01, DNA Sequence ORF Start: ATG at 35 |ORF Stop: TGA at 728 SNP Pos: 650 |SNP Change: G to A
TCCCTCTTTCAGTTCAGAGTCTGTCATCTGAACCATGAGGATCTGGTGGCTTCTGCTTGCCATTGAAATCTGC
ACAGGGAACATAAACTCACAGGACACCTGCAGGCAAGGGCACCCTGGAATCCCTGGGAACCCCGGTCACAATG GTCTGCCTGGAAGAGATGGACGAGACGGAGCGAAGGGTGACAAAGGCGATGCAGGTAAGCCTGGTCCCAAAGG AGAAGCTGGACCCACGGGGCCCCAGGGTGAGCCAGGAGTCCGGGGAATAAGAGGCTGGAAAGGAGATCGAGGA GAGAAAGGGAAAATCGGTGAGACTCTAGTCTTGCCAAAAAGTGCTTTCACTGTGGGGCTCACGGTGCTGAGCA AGTTTCCTTCTTCAGATATGCCCATTAAATTTGATAAGATCCTGTATAACGAATTCAACCATTATGATACAGC AGCGGGGAAATTCACGTGCCACATTGCTGGGGTCTATTACTTCACCTACCACATCACTGTTTTCTCCAGAAAT GTTCAGGTGTCTTTGGTCAAAAATGGAGTAAAAATACTGCACACCAAAGATGCTTACATGAGCTCTGAGGACC AGGCCTCTGGCGGCATTGTCCTGCAGCTGAAGCTCGGGGATGAGGTGTGGCTGCAGGTGACAGGAAGAGAGAG GTTCAATGGCTTGTTTGCTGATGAGGACGATGACACAACTTTCACAGGGTTCCTTCTGTTCAGCAGCCCGTGA CAGAGGAGAGTTTAAAAATCCGCCACACCATCCATCAGAATCAGCTTGGGATGAACTTATTCAGATGGTTTTA CTTTATTAATTCCTC NOV17r, SNP13381827 of SEQ ID NO: 264 231 aa MW t 25045.1kD CG95430-01, Protein Sequence SNP Pos: 206 SNP Change: Gly to Arg
MRIWWLLLAIEICTGNINSQDTCRQGHPGIPGNPGHNGLPGRDGRDGAKGDKGDAGKPGPKGEAGPTGPQGEP GVRGIRG KGDRGEKGKIGETLVLPKSAFTVGLTVLSKFPSSD PIKFDKILYNEFNHYDTAAGKFTCHIAGV YYFTYHITVFSRNVQVSLVKNGVKILHTKDAYMSSEDQASGGIVLQLKLGDEV LQVTGRERFNGLFADEDDD TTFTGFLLFSSP
NOV17s, SNP13381822 of SEQ ID NO: 265 818 bp CG95430-01, DNA Sequence ORF Start: ATG at 35 ORF Stop: TGA at 728
SNP Pos: 687 j SNP Change: A to G
TCCCTCTTTCAGTTCAGAGTCTGTCATCTGAACCATGAGGATCTGGTGGCTTCTGCTTGCCATTGAAATCTGC
ACAGGGAACATAAACTCACAGGACACCTGCAGGCAAGGGCACCCTGGAATCCCTGGGAACCCCGGTCACAATG GTCTGCCTGGAAGAGATGGACGAGACGGAGCGAAGGGTGACAAAGGCGATGCAGGTAAGCCTGGTCCCAAAGG AGAAGCTGGACCCACGGGGCCCCAGGGTGAGCCAGGAGTCCGGGGAATAAGAGGCTGGAAAGGAGATCGAGGA GAGAAAGGGAAAATCGGTGAGACTCTAGTCTTGCCAAAAAGTGCTTTCACTGTGGGGCTCACGGTGCTGAGCA AGTTTCCTTCTTCAGATATGCCCATTAAATTTGATAAGATCCTGTATAACGAATTCAACCATTATGATACAGC AGCGGGGAAATTCACGTGCCACATTGCTGGGGTCTATTACTTCACCTACCACATCACTGTTTTCTCCAGAAAT GTTCAGGTGTCTTTGGTCAAAAATGGAGTAAAAATACTGCACACCAAAGATGCTTACATGAGCTCTGAGGACC AGGCCTCTGGCGGCATTGTCCTGCAGCTGAAGCTCGGGGATGAGGTGTGGCTGCAGGTGACAGGAGGAGAGAG GTTCAATGGCTTGTTTGCTGATGAGGACGGTGACACAACTTTCACAGGGTTCCTTCTGTTCAGCAGCCCGTGA CAGAGGAGAGTTTAAAAATCCGCCACACCATCCATCAGAATCAGCTTGGGATGAACTTATTCAGATGGTTTTA
CTTTATTAATTCCTC
NOVl 7s, SNP13381822 of SEQ ID NO: 266 (231 aa (MW at 248873kD CG95430-01, Protein Sequence SNP Pos: 218 ; SNP Change: Asp to Gly RI LLLAIEICTGNINSQDTCRQGHPGIPGNPGHNGLPGRDGRDGAKGDKGDAGKPGPKGEAGPTGPQGEP GVRGIRGWKGDRGEKGKIGETLVLPKSAFTVGLTVLSKFPSSDMPIKFDKILYNEFNHYDTAAGKFTCHIAGV YYFTYHITVFSRNVQVSLVKNGVKILHTKDAYMSSEDQASGGIVLQLKLGDEVWLQVTGGERFNGLFADEDGD TTFTGFLLFSSP
!NOV17t, SNP13381826 of SEQ ID NO: 267 818 bp CG95430-01, DNA Sequence ORF Start: ATG at 35 ORF Stop: TGA at 728
TCCCTCTTTCAGTTCAGAGTCTGTCATCTGAACCATGAGGATCTGGTGGCTTCTGCTTGCCATTGAAATCTGC
ACAGGGAACATAAACTCACAGGACACCTGCAGGCAAGGGCACCCTGGAATCCCTGGGAACCCCGGTCACAATG GTCTGCCTGGAAGAGATGGACGAGACGGAGCGAAGGGTGACAAAGGCGATGCAGGTAAGCCTGGTCCCAAAGG AGAAGCTGGACCCACGGGGCCCCAGGGTGAGCCAGGAGTCCGGGGAATAAGAGGCTGGAAAGGAGATCGAGGA GAGAAAGGGAAAATCGGTGAGACTCTAGTCTTGCCAAAAAGTGCTTTCACTGTGGGGCTCACGGTGCTGAGCA AGTTTCCTTCTTCAGATATGCCCATTAAATTTGATAAGATCCTGTATAACGAATTCAACCATTATGATACAGC AGCGGGGAAATTCACGTGCCACATTGCTGGGGTCTATTACTTCACCTACCACATCACTGTTTTCTCCAGAAAT GTTCAGGTGTCTTTGGTCAAAAATGGAGTAAAAATACTGCACACCAAAGATGCTTACATGAGCTCTGAGGACC AGGCCTCTGGCGGCATTGTCCTGCAGCTGAAGCTCGGGGATGAGGTGTGGCTGCAGGTGACAGGAGGAGAGAG GTTCAATGGCTTGTTTGCTGATGAGGACGATGACACAACTTTCACAGGGTTCCTTCTGTCCAGCAGCCCGTGA CAGAGGAGAGTTTAAAAATCCGCCACACCATCCATCAGAATCAGCTTGGGATGAACTTATTCAGATGGTTTTA CTTTATTAATTCCTC NOV17t, SNP13381826 of SEQ ID, NO: 268 231 aa |MW at 24885.9kD CG95430-01, Protein Sequence SNP Pos: 228 SNP Change: Phe to Ser
MRI LLLAIEICTGNINSQDTCRQGHPGIPGNPGHNGLPGRDGRDGAKGDKGDAGKPGPKGEAGPTGPQGEP GWGIRGWKGDRGEKGKIGETLVLPKSAFTVGLTVLSKFPSSDMPIKFDKILYNEFNHYDTAAGKFTCHIAGV YYFTYHITVFSRNVQVSLVKNGVKILHTKDAY SSEDQASGGIVLQLKLGDEVWLQVTGGERFNGLFADEDDD TTFTGFLLSSSP
A ClustalW comparison of the above protein sequences yields the following sequence alignment shown in Table 17B.
Table 17B. Comparison of the NOV17 protein sequences.
NOVl7a QDTCRQGHPGI GNPGHNGLPGRDGRDGAKGDKG
NOV17b MRIWWFLLAIEICTGNINSQDTCRQGHPGIPGNPGHNGLSGRDGRDGAKGDKG
NOV17c MRI LLLAIEICTGNINSQDTCRQGHPGIPGNPGHNGLPGRDGRDGAKGDKG
NOV17d TGSTMRI FLLAIEICTGNINSQDTCRQGHPGIPGNPGHNGLPGRDGRDGAKGDKG
NOV17e
NOVl7f MRIW LLLAIEICTGNINSQDTCRQGHPGIPGNPGHNGLPGRDGRDGAKGDKG
NOV17g MRI FLLAIEICTGNINSQDTCRQGHPGIPGNPGHNGLSGRDGRDGAKGDKG
NOV17h QDTCRQGHPGIPGNPGHNGLPGRDGRDGAKGDKG
NOV17i MRI FLLAIEICTGNINSQDTCRQGHPGIPGNPGHNGLPGRDGRDGAKGDKG
NOV17j QDTCRQGHPGIPGNPGHNGLPGRDGRDGAKGDKG
NOV17 MRI WFLLAIEICTGNINSQDTCRQGHPGIPGNPGHNGLPGRDGRDGAKGDKG
NOVl71 MGHHHHHHRIWWFLLAIEICTGNINSQDTCRQGHPGIPGNPGHNGLPGRDGRDGAKGDKG
NOVl7m HHHHHHQDTCRQGHPGIPGNPGHNGLPGRDGRDGAKGDKG
NOV17n HHHHHHQDTCRQGHPGIPGNPGHNGLPGRDGRDGAKGDKG
NOV17a DAGEPGRPGSPGKDGTSGEKGERGADGKVEAKGIKGDQGSRGSPGKHGPKGLAGPMGEKG
NOVl7b DAGEPGRPGSPGKDGTSGEKGERGADGKVEAKGIKGDQGSRGSPGKHGPKGLAGPMGEKG
NOV17c DAGKPG PKGEAGPTG
NOVl7d DAGEPGRPGSPGKDGTSGEKGERGADGKVEAKGIKGDQGSRGSPGKHGPKGLAGP GEKG
NOV17e
NOVl7f DAGEPGRPGSPGKDGTSGEKGERGADGKVEAKGIKGDQGSRGSPGKHGPKGLAGPMGEKG
NOV17g DAGEPGRPGSPGKDGTSGEKGERGADGKVEAKGIKGDQGSRGSPGKHGPKGLAGPMGEKG
NOV17h DAGEPGRPGSPGKDGTSGEKGERGADGKVEAKGIKGDQGSRGSPGKHGPKGLAGPMGEKG
NOVl7i DAGEPGRPGSPGKDGTSGEKGΞRGADGKVEAKGIKGDQGSRGSPGKHGPKGLAGPMGEKG
NOV17J DAGEPGRPGSPGKDGTSGEKGERGADGKVEAKGIKGDQGSRGSPGKHGPKGLAGPMGEKG
NOV17k DAGEPGRPGSPGKDGTSGEKGERGADGKVEAKGIKGDQGSRGSPGKHGPKGLAGPMGEKG
NOV171 DAGEPGRPGSPGKDGTSGEKGERGADGKVEAKGIKGDQGSRGSPGKHGPKGLAGPMGEKG
NOVl7m DAGEPGRPGSPGKDGTSGEKGΞRGADGKVEAKGIKGDQGSRGSPGKHGPKGLAGP GEKG
NOVl7n DAGEPGRPGSPGKDGTSGEKGERGADGKVEAKGIKGDQGSRGSPGKHGPKGLAGP GEKG
NOV17a LRGETGPQGQKGNKGDVGPTGPEGPRGNIGPLGPTGLPGP GPIGKPGPKGEAGPTGPQG
NOV17b LRGETGPQGQKGNKGDVGPTGPEGPRGNIGPLGPTGLPGP GPIGKPGPKGEAGPTGPQG
NOV17C PQG
NOV17d LRGETGPQGQKGNKGDVGPTGPEGPRGNIGPLGPTGLPGPMGPIGKPGPKGEAGPTGPQG
NOV17e
NOV17f LRGETGPQGQKGNKGDVGPTGPEGPRGNIGPLGPTGLPGP GPIGKPGPKGEAGPTGPQG
NOV17g LRGETGPQGQKGNKG
NOV17h LRGETGPQGQKGNKGDVGPTGPEGPRGNIGPLGPTGLPGPMGPIGKPGPKGEAGPTGPQG
NOV17i LRGETGPQGQKGNKGDVGPTGPEGPRGNIGPLGPTGLPGPMGPIGKPGPKGEAGPTGPQG
NOV17J LRGETGPQGQKGNKGDVGPTGPEGPRGNIGPLGPTGLPGPMGPIGKPGPKGEAGPTGPQG
NOV17k LRGETGPQGQKGNKGDVGPTGPEGPRGNIGPLGPTGLPGPMGPIGKPGPKGEAGPTGPQG
NOV171 LRGETGPQGQKGNKGDVGPTGPEGPRGNIGPLGPTGLPGPMGPIGKPGPKGEAGPTGPQG ~ " " " 279 ' NOVl7m LRGETGPQGQKGNKGDVGPTGPEGPRGNIGPLGPTGLPGPMGPIGKPGPKGEAGPTGPQG NOV17n LRGETGPQGQKGNKGDVGPTGPEGPRGNIGPLGPTGLPGPMGPIGKPGPKGEAGPTGPQG
NOVl7 EPGVRGIRG KGDRGEKGKIGETLVLPKSAFTVGLTVLSKFPSSDVPIKFDKILYNEFNH NOVl7b EPGVRGIRG KGDRGEKGKIGETLVLPKSAFTVGLTVLSKFPSSDMPIKFDKILYNEFNH NOVl7c EPGVRGIRG KGDRGEKGKIGETLVLPKSAFTVGLTVLSKFPSSDMPIKFDKILYNEFNH NOV17d EPGVRGIRG KGDRGEKGKIGETLVLPKSAFTVGLTVLSKFPSSDVPIKFDKILYNEFNH NOVl7e AFTVGLTVLSKFPSSDMPIKFDKILYNEFNH NOVl7f EPGVRGIRG KGDRGEKGKIGETLVLPKSAFTVGLTVLSKFPSSDVPIKFDK NOV17g EPGVRGIRG KGDRGEKGKIGETLVLPKSAFTVGLTVLSKFPSSDVPIKFDKILYNEFNH NOV17 EPGVRGIRG KGDRGEKGKIGETLVLPKSAFTVGLTVLSKFPSSDVPIKFDKILYNEFNH NOVl7i EPGVRGIRGWKGDRGEKGKIGETLVLPKSAFTVGLTVLSKFPSSDVPIKFDKILYNEFNH NOVl7j EPGVRGIRG KGDRGΞKGKIGETLVLPKSAFTVGLTVLSKFPSSDVPIKFDKILYNEFNH NOVl k EPGVRGIRGWKGDRGEKGKIGETLVLPKSAFTVGLTVLSKFPSSDVPIKFDKILYNEFNH NOVl71 EPGVRGIRG KGDRGEKGKIGETLVLPKSAFTVGLTVLSKFPSSDVPIKFDKILYNEFNH NOV17m EPGVRGIRG KGDRGEKGKIGETLVLPKSAFTVGLTVLSKFPSSDVPIKFDKILYNEFNH NOV17n EPGVRGIRGWKGDRGEKGKIGETLVLPKSAFTVGLTVLSKFPSSDVPIKFDKILYNEFNH
NOVl7a YDTAAGKFTCHIAGVYYFTYHITVFSRNVQVSLVKNGVKILHTKDAYMSSEDQASGGIVL NOVl7b YDTAAGKFTCHIAGVYYFTYHITVFSRNVQVSLVKNGVKILHTKDAYMSSEDQASGGIVL NOVl7c YDTAAGKFTCHIAGVYYFTYHITVFSRNVQVSLVKNGVKILHTKDAYMSSΞDQASGGIVL NOVl7d YDTAAGKFTCHIAGVYYFTYHITVFSRNVQVSLVKNGVKILHTKDAYMSSEDQASGGIVL NOV17e YDTAAGKFTCHIAGVYYFTYHITVFSRNVQVSLVKNGVKILHTKDAYMSSEDQASGGIVL NOVl7f 1 HITVFSRNVQVSLVKNGVKILHTRDAYVSSEDQASGSIVL NOVl7g YDTAAGKFTCHIAGVYYFTYHIAVFSSNVQVSLVKNGVKILHTKDAYMSSEDQASGGIVL N0V17h YDTAAGKFTCHIAGVYYFTYHITVFSRNVQVSLVKNGVKILHTKDAYMSSEDQASGGIVL NOVl7i YDTAAGKFTCHIAGVYYFTYHITVFSRNVQVSLVKNGVKILHTKDAYMSSEDQASGGIVL NOVl7j YDTAAGKFTCHIAGVYYFTYHITVFSRNVQVSLVKNGVKILHTKDAYMSSEDQASGGIVL NOVl7k YDTAAGKFTCHIAGVYYFTYHITVFSRNVQVSLVKNGVKILHTKDAYMSΞEDQASGGIVL NOVl71 YDTAAGKFTCHIAGVYYFTYHITVFSRNVQVSLVKNGVKILHTKDAYMSSEDQASGGIVL NOVl7m YDTAAGKFTCHIAGVYYFTYHITVFSRNVQVSLVKNGVKILHTKDAYMSSΞDQASGGIVL NOV17n YDTAAGKFTCHIAGVYYFTYHITVFSRNVQVSLVKNGVKILHTKDAYMSSEDQASGGIVL
NOVl7a QLKLGDEV LQVTGGERFNGLFADEDDDTTFTGFLLFSSP NOVl7b QLKLGDEVWLQVTGGERFNGLFADEDDDTTFTGFLLFSSQ NOVl7c QLKLGDEV LQVTGGERFNGLFADEDDDTTFTGFLLFSSP NOVl7d QLKLGDEV LQVTGGEKFNGLFADEDDDTTFTGFLLFSSP NOV17e QLKLGDEV LQVTGGERFNGLFADEDDDTTFTGFLLFSSP NOVl7f QLKLGDEMWLQVTGGERFNGLFADEDDDTTFTGFLLFSSQ NOV17g QLKLGDEV LQVTGGERFNGLFADEDDDTTFTGFLLFSSQ N0V17h QLKLGDEV LQVTGGERFNGLFADEDDDTTFTGFLLFSSP NOVl7i QLKLGDEVWLQVTGGEKFNGLFADEDDDTTFTGFLLFSSP NOVl7j QLKLGDEVWLQVTGGERFNGLFADEDDDTTFTGFLLFSSP NOVl7k QLKLGDEVWLQVTGGE FNGLFADEDDDTTFTGFLLFSSP NOVl71 QLKLGDEVWLQVTGGEKFNGLFADEDDDTTFTGFLLFSSP NOVl7m QLKLGDEVWLQVTGGEKFNGLFADEDDDTTFTGFLLFSSP NOV17n QLKLGDEV LQVTGGERFNGLFADEDDDTTFTGFLLFSSP
NOVl7a (SEQ ID NO 230) NOVl7b (SEQ ID NO 232) NOVl7c (SEQ ID NO 234) NOVl7d (SEQ ID NO 236) NOVl7e (SEQ ID NO 238) NOVl7f (SEQ ID NO 240) NOV17g (SEQ ID NO 242) NOV17h (SEQ ID NO 244) NOVl7i (SEQ ID NO 24S) NOVl7j (SEQ ID NO 248) NOV17k (SEQ ID NO 250) NOV171 ( SEQ ID NO NOVl 7m (SEQ ID NO NOV17n ( SEQ ID NO
Further analysis ofthe NOVl7aproteinyielded the followingproperties shown in Table 17C.
Table 17C. Protein Sequence Properties NOV17a
SignalP analysis: No Known Signal Sequence Predicted
PSORT II analysis:
PSG: a new signal peptide prediction method
N-region: length 5; pos.chg 1; neg.chg 1 H-region: length 17; peak value 0.39 PSG score: -4.01
GvH: von Heijne's method for signal seg. recognition GvH score (threshold: -2.1): -14.34 possible cleavage site : between 52 and 53
>>> Seems to have no N-terminal signal peptide
ALOM: Klein et al's method for TM region allocation Init position for calculation: 1
Tentative number of TMS(s) for the threshold 0.5: 0 number of TMS(s) .. fixed PERIPHERAL Likelihood = 2.17 (at 177) ALOM score: 2.17 (number of TMSs: 0)
MITDISC: discrimination of mitochondrial targeting seg R content: 2 Hyd Moment (75): 8.45 Hyd Momen (95) : 12.46 G content: 6 D/E content: 2 S/T content: 1 Score: -6.71
Gavel: prediction of cleavage sites for mitochondrial preseq R-2 motif at 15 CRQ|GH
NUCDISC: discrimination of nuclear localization signals pat4 : none pat7 : none bipartite: none content of basic residues: 12.4% NLS Score: -0.47
KDEL: ER retention motif in the C-terminus : none
ER Membrane Retention Signals :
XXRR-like motif in the N-terminus : DTCR none
SKL: peroxisomal targeting signal in the C-terminus: none
PTS2 : 2nd peroxisomal targeting signal : none
VAC: possible vacuolar targeting motif: none RNA-binding motif: none
Actinin-type actin-binding motif : type 1: none type 2 : none
NMYR: N-myristoylation pattern : none
Prenylation motif: none memYQRL: transport motif from cell surface to Golgi: none
Tyrosines in the tail : none
Dileucine motif in the tail : none checking 63 PROSITE DNA binding motifs: none checking 71 PROSITE ribosomal protein motifs: none checking 33 PROSITE prokaryotic DNA binding motifs: none
NNCN: Reinhardt's method for Cytoplasmic/Nuclear discrimination Prediction: cytoplasmic Reliability: 76.7
COIL: Lupas's algorithm to detect coiled-coil regions total : 0 residues
Final Results (k = 9/23):
56.5 %: cytoplasmic
21.7 %: nuclear
8.7 %: mitochondrial
4.3 %: Golgi
4.3 %: vacuolar
4.3 %: plasma membrane
» prediction for CG95430-02 is cyt (k=23)
A search of the NOVl 7a protein against the Geneseq database, a proprietary database that contains sequences published in patents and patent publication, yielded several homologous proteins shown in Table 17D.
Figure imgf000285_0001
In a BLAST search of public sequence databases, the NOV17a protein was found to have homology to the proteins shown in the BLASTP data in Table 17E.
Figure imgf000286_0001
PFam analysis predicts that the NOVl 7a protein contains the domains shown in the Table 17F.
Figure imgf000286_0002
Example 18.
The NOVl 8 clone was analyzed, and the nucleotide and encoded polypeptide sequences are shown in Table 18A.
Figure imgf000287_0001
NOV18a, CG97111-01 SEQ ID NO: 270 159 aa MW at 17706.9kD Protein Sequence CSLPMARYYRI YADQKALYTRDGQLLVGDPVADNCCAEKICILPNRGLARTKVPIFLGIQGGSRCLACVET EEGPSLQLEPSTLPPQD IEELYKGGEEATRFTFFQSSSGSAFRLEAAAWPGWFLCGPAEPQQPVQLTKESE PSARTKFYFEQSW
NOV18b, CG97111-02 SEQ ID NO: 271 |499 bp_ DNA Sequence ORF Start: ATG at 16 loRF Stop: TAG at 472
CCACTGATTGCAGGAATGTGTTCCCTCCCCATGGCAAGATACTACATAATTAAATATGCAGACCAGAAGGCTC
TATACACAAGAGATGGCCAGCTGCTGGTGGGAGATCCTGTTGCAGACAACTGCTGTGCAGAGAAGATCTGCAT ACTTCCTAACAGAGGCTTGGCCCGCACCAAGGTCCCCATTTTCCTGGGGATCCAGGGAGGGAGCCGCTGCCTG GCATGTGTGGAGACAGAAGAGGGGCCTTCCCTACAGCTGGAGGATGTGAACATTGAGGAACTGTACAAAGGTG GTGAAGAGGCCACACGCTTCACCTTCTTCCAGAGCAGCTCAGGCTCCGCCTTCAGGCTTGAGGCTGCTGCCTG GCCTGGCTGGTTCCTGTGTGGCCCGGCAGAGCCCCAGCAGCCAGTACAGCTCACCAAGGAGAGTGAGCCCTCA GCCCGTACCAAGTTTTACTTTGAACAGAGCTGGTAGGGAGACAGGAAACTGCGTTTTAGCC
NOV18b, CG97111-02 SEQ ID NO: 272 152 aa MW t l6943.1 D Protein Sequence
MCSLPMARYYIIKYADQKALYTRDGQLLVGDPVADNCCAEKICILPNRGLARTKVPIFLGIQGGSRCLACVET EEGPSLQLEDVNIEELYKGGEEATRFTFFQSSSGSAFRLEAAAWPG FLCGPAEPQQPVQLTKESEPSARTKF YFEQS |NOV18c, CG97111-03 |SEQ IDNO: 273 |483 bp
DNA Sequence |0RF Start: at 3__ __]θRF Stop: TAG at 465_
CACTGTCATACTGTTTCAGAATTAAATATGCAGACCAGAAGGCTCTATACACAAGAGATGGCCAGCTGCTGGT GGGAGATCCTGTTGCAGACAACTGCTGTGCAGAGAAGATCTGCATACTTCCTAACAGAGGCTTGGCCCGCACC AAGGTCCCCATTTTCCTGGGGATCCAGGGAGGGAGCCGCTGCCTGGCATGTGTGGAGACAGAAGAGGGGCCTT CCCTACAGCTGGAGC'CATCCACCTTGCCCCCACAGGATGTGAACATTGAGGAACTGTACAAAGGTGGTGAAGA GGCCACACGCTTCACCTTCTTCCAGAGCAGCTCAGGCTCCGCCTTCAGGCTTGAGGCTGCTGCCTGGCCTGGC TGGTTCCTGTGTGGCCCGGCAGAGCCCCAGCAGCCAGTACAGCTCACCAAGGAGAGTGAGCCCTCAGCCCGTA CCAAGTTTTACTTTGAACAGAGCTGGTAGGGAGACAGGAAACTGC
NOVlδc, CG97111-03 SEQ ID NO: 274 154 aa MW at 17104.2kD
Protein Sequence j
LSYCFRIKYADQKALYTRDGQLLVGDPVADNCCAEKICILPNRGLARTKVPIFLGIQGGSRCLACVETEEGPS LQLEPSTLPPQDVNIEELYKGGEEATRFTFFQSSSGSAFRLEAAAWPGWFLCGPAEPQQPVQLTKESEPSART KFYFEQSW
Figure imgf000288_0001
NOV18d, SNP13382516 of j SEQ ID NO: 276 159 aa M at 17706.9kD
CG97111-01, Protein Sequence jSNP Pos:,24 | JSNP Change: Asp to Asp
MCSLPMARYYRIKYADQKALYTRDGQLLVGDPVADNCCAEKICILPNRGLARTKVPIFLGIQGGSRCLACVET SEGPSLQLEPSTLPPQDVNIEΞLYKGGEEATRFTFFQSSSGSAFRLEAAAWPGWFLCGPAEPQQPVQLTKESE PSARTKFYFEQSW NOV18e, SNP13382517 of SEQ ID NO: 277 1019 bp CG97111-01, DNA Sequence ORF Start: ATG at 54 fORF Stop: TAG at 531 SNP Pos: 184 ~ " Jaj chmgeiTto C
GGTTCCAGGAACTCAGGATCTGCAGTGAGGACCAGACACCACTGATTGCAGGAATGTGTTCCCTCCCCATGGC
AAGATACTACAGAATTAAATATGCAGACCAGAAGGCTCTATACACAAGAGATGGCCAGCTGCTGGTGGGAGAT CCTGTTGCAGACAACTGCTGTGCAGAGAAGATCTGCACACTTCCTAACAGAGGCTTGGCCCGCACCAAGGTCC CCATTTTCCTGGGGATCCAGGGAGGGAGCCGCTGCCTGGCATGTGTGGAGACAGAAGAGGGGCCTTCCCTACA GCTGGAGCCATCCACCTTGCCCCCACAGGATGTGAACATTGAGGAACTGTACAAAGGTGGTGAAGAGGCCACA CGCTTCACCTTCTTCCAGAGCAGCTCAGGCTCCGCCTTCAGGCTTGAGGCTGCTGCCTGGCCTGGCTGGTTCC TGTGTGGCCCGGCAGAGCCCCAGCAGCCAGTACAGCTCACCAAGGAGAGTGAGCCCTCAGCCCGTACCAAGTT TTACTTTGAACAGAGCTGGTAGGGAGACAGGAAACTGCGTTTTAGCCTTGTGCCCCCAAACCAAGCTCATCCT GCTCAGGGTCTATGGTAGGCAGAATAATGTCCCCCGAAATATGTCCACATCCTAATCCCAAGATCTGTGCATA
TGTTACCATACATGTCCAAAGAGGTTTTGCAAATGTGATTATGTTAAGGATCTTGAAATGAGGAGACAATCCT iGGGTTATCCTTGTGGGCTCAGTTTAATCACAAGAAGGAGGCAGGAAGGGAGAGTCAGAGAGAGAATGGAAGAT iACCATGCTTCTAATTTTGAAGATGGAGTGAGGGGCCTTGAGCCAACATATGCAGGTGTTTTTAGAAGGAGGAA lAAGCCAAGGGAACGGATTCTCCTCTATAGTCTCCGGAAGGAACACAGCTCTTGACACATGGATTTCAGCTCAG
TGACACCCATTTCAGACTTCTGACCTCCACAACTATAAAATAATAAACTTGTGTTATTGTAAACCTCTGG
Figure imgf000289_0001
NOV18f, SNP13382518 of SEQ ID NO: 279 1019 bp CG97111-01, DNA Sequence ORF Sto ATG at 54 |θRF Stop: TAG at 531
SNP Pos: 205 SNP Change: C to A
GGTTCCAGGAACTCAGGATCTGCAGTGAGGACCAGACACCACTGATTGCAGGAATGTGTTCCCTCCCCATGGC
AAGATACTACAGAATTAAATATGCAGACCAGAAGGCTCTATACACAAGAGATGGCCAGCTGCTGGTGGGAGAT CCTGTTGCAGACAACTGCTGTGCAGAGAAGATCTGCATACTTCCTAACAGAGGCTTGGACCGCACCAAGGTCC CCATTTTCCTGGGGATCCAGGGAGGGAGCCGCTGCCTGGCATGTGTGGAGACAGAAGAGGGGCCTTCCCTACA GCTGGAGCCATCCACCTTGCCCCCACAGGATGTGAACATTGAGGAACTGTACAAAGGTGGTGAAGAGGCCACA CGCTTCACCTTCTTCCAGAGCAGCTCAGGCTCCGCCTTCAGGCTTGAGGCTGCTGCCTGGCCTGGCTGGTTCC TGTGTGGCCCGGCAGAGCCCCAGCAGCCAGTACAGCTCACCAAGGAGAGTGAGCCCTCAGCCCGTACCAAGTT TTACTTTGAACAGAGCTGGTAGGGAGACAGGAAACTGCGTTTTAGCCTTGTGCCCCCAAACCAAGCTCATCCT GCTCAGGGTCTATGGTAGGCAGAATAATGTCCCCCGAAATATGTCCACATCCTAATCCCAAGATCTGTGCATA
TGTTACCATACATGTCCAAAGAGGTTTTGCAAATGTGATTATGTTAAGGATCTTGAAATGAGGAGACAATCCT
GGGTTATCCTTGTGGGCTCAGTTTAATCACAAGAAGGAGGCAGGAAGGGAGAGTCAGAGAGAGAATGGAAGAT lACCATGCTTCTAATTTTGAAGATGGAGTGAGGGGCCTTGAGCCAACATATGCAGGTGTTTTTAGAAGGAGGAA lAAGCCAAGGGAACGGATTCTCCTCTATAGTCTCCGGAAGGAACACAGCTCTTGACACATGGATTTCAGCTCAG
TGACACCCATTTCAGACTTCTGACCTCCACAACTATAAAATAATAAACTTGTGTTATTGTAAACCTCTGG
Figure imgf000289_0002
A ClustalW comparison ofthe above protein sequences yields the following sequence alignment shown in Table 18B.
Table 18B. Comparison ofthe NOV18 protein sequences.
NOVl8a MCSLPMARYYRIKYADQKALYTRDGQLLVGDPVADNCCAEKICILPNRGLARTKVPIFLG
NOVl8b MCSLPMARYYIIKYADQKALYTRDGQLLVGDPVADNCCAEKICILPNRGLARTKVPIFLG OVl8c LSYCFRIKYADQKALYTRDGQLLVGDPVADNCCAEKICILPNRGLARTKVPIFLG
Novisa IQGGSRCLACVETEEGPSLQLEPSTLPPQDVNIEELYKGGEEATRFTFFQSSSGSAFRLE OVl8b IQGGSRCLACVETEEGPSLQLΞ DVNIEELYKGGEEATRFTFFQSSSGSAFRLE
NOV18c IQGGSRCLACVETEEGPSLQLEPSTLPPQDVNIEELYKGGEEATRFTFFQSSSGSAFRLE
NOVI8a AAAWPGWFLCGPAEPQQPVQLTKESEPSARTKFYFEQSW
NOVI8b AAAWPGWFLCGPAEPQQPVQLTKESEPSARTKFYFEQSW
NOVlδc AAAWPGWFLCGPAEPQQPVQLTKESEPSARTKFYFEQSW
NOVl8a (SEQ ID NO 270) NOVl8b (SEQ ID NO 272) NOVl8c (SEQ ID NO 274)
Further analysis of the NOVl 8a protein yielded the following properties shown in Table 18C.
Table 18C. Protein Sequence Properties NOV18a
SignalP analysis: I No Known Signal Sequence Predicted
PSORT II analysis:
PSG: a new signal peptide prediction method
N-region: length 11; pos.chg 2; neg.chg 0 H-region: length 1; peak value -13.22 PSG score: -17.62
GvH: von Heijne's method for signal seq. recognition GvH score (threshold: -2.1): -5.73 possible cleavage site: between 39 and 40
>>> Seems to have no N-terminal signal peptide
ALOM: Klein et al's method for TM region allocation Init position for calculation: 1
Tentative number of TMS(s) for the threshold 0.5: 0 number of TMS(s) .. fixed PERIPHERAL Likelihood = 2.07 (at 55) ALOM score: 2.07 (number of TMSs: 0)
MITDISC: discrimination of mitochondrial targeting seg R content : 2 Hyd Momen (75) : 8.63 Hyd Moment (95): 6.96 . G content: 0 D/E content: 1 S/T content: 1 Score: -2.42
Gavel: prediction of cleavage sites for mitochondrial preseq R-2 motif at 21 YRl|KY
NUCDISC: discrimination of nuclear localization signals pat : none pat7 : none bipartite: none content of basic residues: 10.1%
NLS Score: -0.47
KDEL: ER retention motif in the C-terminus: none
ER Membrane Retention Signals: none
SKL: peroxisomal targeting signal in the C-terminus : none
PTS2 : 2nd peroxisomal targeting signal: none
VAC: possible vacuolar targeting motif: found ILPN at 44
RNA-binding motif: none
Actinin-type actin-binding motif : type 1 : none type 2 : none
NMYR: N-myristoylation pattern .- none
Prenylation motif: none memYQRL: transport motif from cell surface to Golgi: none
Tyrosines in the tail : none
Dileucine motif in the tail : none checking 63 PROSITE DNA binding motifs: none checking 71 PROSITE ribosomal protein motifs: none checking 33 PROSITE prokaryotic DNA binding motifs: none
NNCN: Reinhardt's method for Cytoplasmic/Nuclear discrimination Prediction: cytoplasmic Reliability: 55.5
COIL: Lupas's algorithm to detect coiled-coil regions total: 0 residues
Final Results (k = 9/23) :
47.8 %: nuclear
39.1 %: mitochondrial
4.3 %: vacuolar
4.3 %: vesicles of secretory system
4.3 %: cytoplasmic
» prediction for CG97111-01 is nuc (k=23)
A search of the NOVl 8a protein against the Geneseq database, a proprietary database that contains sequences published in patents and patent publication, yielded several homologous proteins shown in Table 18D.
Figure imgf000292_0001
In a BLAST search of public sequence databases, the NOVl 8a protein was found to have homology to the proteins shown in the BLASTP data in Table 18E.
Figure imgf000293_0001
PFam analysis predicts that the NOVl 8a protein contains the domains shown in the Table 18F.
Figure imgf000293_0002
Example 19: The NOVl 9 clone was analyzed, and the nucleotide and encoded polypeptide sequences are shown in Table 19A. Table 19A:
NOV19a, pCR2.1 - CG10132038.0.67- S540u2, a domain SEQ ID NO: 281 1377 bp of CG50513-05
TGGGAACATAATCCTTGGACTGCATGTTCCGTGTCCTGTGGAGGAGGGATTCAGAGACGGAGCTTTGTGTGTG TAGAGGAATCCATGCATGGAGAGATATTGCAGGTGGAAGAATGGAAGTGCATGTACGCACCCAAACCCAAGGT TATGCAAACTTGTAATCTGTTTGATTGCCCCAAGTGGATTGCCATGGAGTGGTCTCAGTGCACAGTGACTTGT GGCCGAGGGTTACGGTACCGGGTTGTTCTGTGTATTAACCACCGCGGAGAGCATGTTGGGGGCTGCAATCCAC AACTGAAGTTACACATCAAAGAAGAATGTGTCATTCCCATCCCGTGTTATAAACCAAAAGAAAAAAGTCCAGT GGAAGCAAAATTGCCTTGGCTGAAACAAGCACAAGAACTAGAAGAGACCAGAATAGCAACAGAAGAACCAACG TTCATTCCAGAACCCTGGTCAGCCTGCAGTACCACGTGTGGGCCGGGTGTGCAGGTCCGTGAGGTGAAGTGCC GTGTGCTCCTCACATTCACGCAGACTGAGACTGAGCTGCCCGAGGAAGAGTGTGAAGGCCCCAAGCTGCCCAC CGAACGGCCCTGCCTCCTGGAAGCATGTGATGAGAGCCCGGCCTCCCGAGAGCTAGACATCCCTCTCCCTGAG GACAGTGAGACGACTTACGACTGGGAGTACGCTGGGTTCACCCCTTGCACAGCAACATGCGTGGGAGGCCATC AAGAAGCCATAGCAGTGTGCTTACATATCCAGACCCAGCAGACAGTCAATGACAGCTTGTGTGATATGGTCCA CCGTCCTCCAGCCATGAGCCAGGCCTGTAACACAGAGCCCTGTCCCCCCAGGTGGCATGTGGGCTCTTGGGGG CCCTGCTCAGCTACCTGTGGAGTTGGAATTCAGACCCGAGATGTGTACTGCCTGCACCCAGGGGAGACCCCTG CCCCTCCTGAGGAGTGCCGAGATGAAAAGCCCCATGCTTTACAAGCATGCAATCAGTTTGACTGCCCTCCTGG CTGGCACATTGAAGAATGGCAGCAGTGTTCCAGGACTTGTGGCGGGGGAACTCAGAACAGAAGAGTCACCTGT CGGCAGCTGCTAACGGATGGCAGCTTTTTGAATCTCTCAGATGAATTGTGCCAAGGACCCAAGGCATCGTCTC ACAAGTCCTGTGCCAGGACAGACTGTCCTCCACATTTAGCTGTGGGAGACTGGTCGAAGTGTTCTGTCAGTTG TGGTGTTGGAATCCAGAGAAGAAAGCAGGTGTGTCAAAGGCTGGCAGCCAAAGGTCGGCGCATCCCCCTCAGT GAGATGATGTGCAGGGATCTACCAGGGCTCCCTCTTGTAAGATCTTGCCAGATGCCTGAGTGC
Figure imgf000294_0001
Example B: Sequencing Methodology and Identification of NOVX Clones
1. GeneCalling™ Technology: This is a proprietary method of performing differential gene expression profiling between two or more samples developed at CuraGen and described by Shimkets, et al., "Gene expression analysis by transcript profiling coupled to a gene database query" Nature Biotechnology 17:198-803 (1999). cDNA was derived from various human samples representing multiple tissue types, normal and diseased states, physiological states, and developmental states from different donors. Samples were obtained as whole tissue, primary cells or tissue cultured primary cells or cell lines. Cells and cell lines may have been treated with biological or chemical agents that regulate gene expression, for example, growth factors, chemokines or steroids. The cDNA thus derived was then digested with up to as many as 120 pairs of restriction enzymes and pairs of linker-adaptors specific for each pair of restriction enzymes were ligated to the appropriate end. The restriction digestion generates a mixture of unique cDNA gene fragments. Limited PCR amplification is performed with primers homologous to the linker adapter sequence where one primer is biotinylated and the other is fluorescently labeled. The doubly labeled material is isolated and the fluorescently labeled single strand is resolved by capillary gel electrophoresis. A computer algorithm compares the electropherograms from an experimental and control group for each of the restriction digestions. This and additional sequence-derived information is used to predict the identity of each differentially expressed gene fragment using a variety of genetic databases. The identity of the gene fragment is confirmed by additional, gene-specific competitive PCR or by isolation and sequencing of the gene fragment.
2. SeqCalling™ Technology: cDNA was derived from various human samples representing multiple tissue types, normal and diseased states, physiological states, and developmental states from different donors. Samples were obtained as whole tissue, primary cells or tissue cultured primary cells or cell lines. Cells and cell lines may have been treated with biological or chemical agents that regulate gene expression, for example, growth factors, chemokines or steroids. The cDNA thus derived was then sequenced using CuraGen's proprietary SeqCalling technology. Sequence traces were evaluated manually and edited for corrections if appropriate. cDNA sequences from all samples were assembled together, sometimes including public human sequences, using bioinformatic programs to produce a consensus sequence for each assembly. Each assembly is included in CuraGen Corporation's database. Sequences were included as components for assembly when the extent of identity with another component was at least 95% over 50 bp. Each assembly represents a gene or portion thereof and includes information on variants, such as splice forms single nucleotide polymorphisms (SNPs), insertions, deletions and other sequence variations.
3. PathCalling™ Technology: The NOVX nucleic acid sequences are derived by laboratory screening of cDNA library by the two-hybrid approach. cDNA fragments covering either the full length of the DNA sequence, or part of the sequence, or both, are sequenced. In silico prediction was based on sequences available in CuraGen Corporation's proprietary sequence databases or in the public human sequence databases, and provided either the full length DNA sequence, or some portion thereof. The laboratory screening was performed using the methods summarized below: cDNA libraries were derived from various human samples representing multiple tissue types, normal and diseased states, physiological states, and developmental states from different donors. Samples were obtained as whole tissue, primary cells or tissue cultured primary cells or cell lines. Cells and cell lines may have been treated with biological or chemical agents that regulate gene expression, for example, growth factors, chemokines or steroids. The cDNA thus derived was then directionally cloned into the appropriate two-hybrid vector (Gal4-activation domain (Gal4-AD) fusion). Such cDNA libraries as well as commercially available cDNA libraries from Clontech (Palo Alto, CA) were then transferred from E.coli into a CuraGen Coφoration proprietary yeast strain (disclosed in U. S. Patents 6,057,101 and 6,083,693, incoφorated herein by reference in their entireties). Gal4-binding domain (Gal4-BD) fusions of a CuraGen Coφoration proprietary library of human sequences was used to screen multiple Gal4-AD fusion cDNA libraries resulting in the selection of yeast hybrid diploids in each of which the Gal4-AD fusion contains an individual cDNA. Each sample was amplified using the polymerase chain reaction (PCR) using non-specific primers at the cDNA insert boundaries. Such PCR product was sequenced; sequence traces were evaluated manually and edited for corrections if appropriate. cDNA sequences from all samples were assembled together, sometimes including public human sequences, using bioinformatic programs to produce a consensus sequence for each assembly. Each assembly is included in CuraGen Coφoration's database. Sequences were included as components for assembly when the extent of identity with another component was at least 95% over 50 bp. Each assembly represents a gene or portion thereof and includes information on variants, such as splice forms single nucleotide polymoφhisms (SNPs), insertions, deletions and other sequence variations.
Physical clone: the cDNA fragment derived by the screening procedure, covering the entire open reading frame is, as a recombinant DNA, cloned into pACT2 plasmid (Clontech) used to make the cDNA library. The recombinant plasmid is inserted into the host and selected by the yeast hybrid diploid generated during the screening procedure by the mating of both CuraGen Coφoration proprietary yeast strains N106' and YULH (U. S. Patents 6,057,101 and 6,083,693).
4. RACE: Techniques based on the polymerase chain reaction such as rapid amplification of cDNA ends (RACE), were used to isolate or complete the predicted sequence of the cDNA of the invention. Usually multiple clones were sequenced from one or more human samples to derive the sequences for fragments. Various human tissue samples from different donors were used for the RACE reaction. The sequences derived from these procedures were included in the SeqCalling Assembly process described in preceding paragraphs. 5. Exon Linking: The NOVX target sequences identified in the present invention were subjected to the exon linking process to confirm the sequence. PCR primers were designed by starting at the most upstream sequence available, for the forward primer, and at the most downstream sequence available for the reverse primer. In each case, the sequence was examined, walking inward from the respective termini toward the coding sequence, until a suitable sequence that is either unique or highly selective was encountered, or, in the case of the reverse primer, until the stop codon was reached. Such primers were designed based on in silico predictions for the full length cDNA, part (one or more exons) of the DNA or protein sequence of the target sequence, or by translated homology of the predicted exons to closely related human sequences from other species. These primers were then employed in PCR amplification based on the following pool of human cDNAs: adrenal gland, bone marrow, brain - amygdala, brain - cerebellum, brain - hippocampus, brain - substantia nigra, brain - thalamus, brain -whole, fetal brain, fetal kidney, fetal liver, fetal lung, heart, kidney, lymphoma - Raji, mammary gland, pancreas, pituitary gland, placenta, prostate, salivary gland, skeletal muscle, small intestine, spinal cord, spleen, stomach, testis, thyroid, trachea, uterus. Usually the resulting amplicons were gel purified, cloned and sequenced to high redundancy. The PCR product derived from exon linking was cloned into the pCR2.1 vector from Invitrogen. The resulting bacterial clone has an insert covering the entire open reading frame cloned into the pCR2.1 vector. The resulting sequences from all clones were assembled with themselves, with other fragments in CuraGen Coφoration's database and with public ESTs. Fragments and ESTs were included as components for an assembly when the extent of their identity with another component of the assembly was at least 95% over 50 bp. In addition, sequence traces were evaluated manually and edited for corrections if appropriate. These procedures provide the sequence reported herein.
6. Physical Clone: Exons were predicted by homology and the intron/exon boundaries were determined using standard genetic rules. Exons were further selected and refined by means of similarity determination using multiple BLAST (for example, tBlastN, BlastX, and BlastN) searches, and, in some instances, GeneScan and Grail. Expressed sequences from both public and proprietary databases were also added when available to further define and complete the gene sequence. The DNA sequence was then manually corrected for apparent inconsistencies thereby obtaining the sequences encoding the full-length protein.
The PCR product derived by exon linking, covering the entire open reading frame, was cloned into the pCR2.1 vector from Invitrogen to provide clones used for expression and screening puφoses.
Example C: Quantitative expression analysis of clones in various cells and tissues
The quantitative expression of various NOV genes was assessed using microtiter plates containing RNA samples from a variety of normal and pathology-derived cells, cell lines and tissues using real time quantitative PCR (RTQ-PCR) performed on an Applied Biosystems (Foster City, CA) ABI PRISM® 7700 or an ABI PRISM® 7900 HT Sequence Detection System.
RNA integrity of all samples was determined by visual assessment of agarose gel electropherograms using 28S and 18S ribosomal RNA staining intensity ratio as a guide (2:1 to 2.5:1 28s:18s) and the absence of low molecular weight RNAs (degradation products). Control samples to detect genomic DNA contamination included RTQ-PCR reactions run in the absence of reverse transcriptase using probe and primer sets designed to amplify across the span of a single exon.
RNA samples were normalized in reference to nucleic acids encoding constitutively expressed genes (i.e., β-actin and GAPDH). Alternatively, non-normalized RNA samples were converted to single strand cDNA (sscDNA) using Superscript II (Invitrogen Coφoration, Carlsbad, CA, Catalog No. 18064-147) and random hexamers according to the manufacturer's instructions. Reactions containing up to 10 μg of total RNA in a volume of 20 μl or were scaled up to contain 50 μg of total RNA in a volume of 100 μl and were incubated for 60 minutes at 42°C. sscDNA samples were then normalized in reference to nucleic acids as described above.
Probes and primers were designed according to Applied Biosystems Primer
Express Software package (version I for Apple Computer's Macintosh Power PC) or a similar algorithm using the target sequence as input. Default reaction condition settings and the following parameters were set before selecting primers: 250 nM primer concentration; 58°-60° C primer melting temperature (Tm) range; 59° C primer optimal Tm; 2° C maximum primer difference (if probe does not have 5' G, probe Tm must be 10° C greater than primer Tm; and 75 bp to 100 bp amplicon size. The selected probes and primers were synthesized by Synthegen (Houston, TX). Probes were double purified by HPLC to remove uncoupled dye and evaluated by mass spectroscopy to verify coupling of reporter and quencher dyes to the 5' and 3' ends of the probe, respectively. Their final concentrations were: 900 nM forward and reverse primers, and 200nM probe.
Normalized RNA was spotted in individual wells of a 96 or 384-well PCR plate (Applied Biosystems, Foster City, CA). PCR cocktails included a single gene-specific probe and primers set or two multiplexed probe and primers sets. PCR reactions were done using TaqMan® One-Step RT-PCR Master Mix (Applied Biosystems, Catalog No. 4313803) following manufacturer's instructions. Reverse transcription was performed at 48° C for 30 minutes followed by amplification/PCR cycles: 95° C IO min, then 40 cycles at 95° C for 15 seconds, followed by 60° C for 1 minute. Results were recorded as CT values (cycle at which a given sample crosses a threshold level of fluorescence) and plotted using a log scale, with the difference in RNA concentration between a given sample and the sample with the lowest CT value being represented as 2 to the power of delta CT. The percent relative expression was the reciprocal of the RNA difference multiplied by 100. CT values below 28 indicate high expression, between 28 and 32 indicate moderate expression, between 32 and 35 indicate low expression and above 35 reflect levels of expression that were too low to be measured reliably. Normalized sscDNA was analyzed by RTQ-PCR using IX TaqMan® Universal Master mix (Applied Biosystems; catalog No. 4324020), following the manufacturer's instructions. PCR amplification and analysis were done as described above.
Panels 1, 1.1, 1.2, and 1.3D
Panels 1, 1.1, 1.2 and 1.3D included 2 control wells (genomic DNA control and chemistry control) and 94 wells of cDNA samples from cultured cell lines and primary normal tissues. Cell lines were derived from carcinomas (ca) including: lung, small cell (s cell var), non small cell (non-s or non-sm); breast; melanoma; colon; prostate; glioma (glio), astrocytoma (astro) and neuroblastoma (neuro); squamous cell (squam); ovarian; liver; renal; gastric and pancreatic from the American Type Culture Collection (ATCC, Bethesda, MD). Normal tissues were obtained from individual adults or fetuses and included: adult and fetal skeletal muscle, adult and fetal heart, adult and fetal kidney, adult and fetal liver, adult and fetal lung, brain, spleen, bone marrow, lymph node, pancreas, salivary gland, pituitary gland, adrenal gland, spinal cord, thymus, stomach, small intestine, colon, bladder, trachea, breast, ovary, uterus, placenta, prostate, testis and adipose. The following abbreviations are used in reporting the results: metastasis (met); pleural effusion (pi. eff or pi effusion) and * indicates established from metastasis.
General_Screening_Panel_vl.4, vl.5, vl.6 and vl.7
Panels 1.4, 1.5, 1.6 and 1.7 were as described for Panels 1, 1.1, 1.2 and 1.3D, above except that normal tissue samples were pooled from 2 to 5 different adults or fetuses.
Panels 2D, 2.2, 2.3, and 2.4
Panels 2D, 2.2, 2.3 and 2.4 included 2 control wells and 94 wells containing RNA or cDNA from human surgical specimens procured through the National Cancer Institute's Cooperative Human Tissue Network (CHTN) or the National Disease Research Initiative (NDRI), Ardais (Lexington, MA) or Clinomics BioSciences (Frederick, MD). Tissues included human malignancies and in some cases matched adjacent normal tissue (NAT). Information regarding histopathological assessment of tumor differentiation grade as well as the clinical stage of the patient from which samples were obtained was generally available. Normal tissue RNA and cDNA samples were purchased from various commercial sources such as Clontech (Palo Alto, CA), Research Genetics and Invitrogen (Carlsbad, CA).
HASS Panel vl.O
The HASS Panel vl .0 included 93 cDNA samples and two controls including: 81 samples of cultured human cancer cell lines subjected to serum starvation, acidosis and anoxia according to established procedures for various lengths of time; 3 human primary cells; 9 malignant brain cancers (4 medulloblastomas and 5 glioblastomas); and 2 controls. Cancer cell lines (ATCC) were cultured using recommended conditions and included: breast, prostate, bladder, pancreatic and CNS. Primary human cells were obtained from Clonetics (WalkersviUe, MD). Malignant brain samples were gifts from the Henry Ford Cancer Center.
ARDAIS Panel vl.O and vl.l
The ARDAIS Panel vl .0 and vl.l included 2 controls and 22 test samples including: human lung adenocarcinomas, lung squamous cell carcinomas, and in some cases matched adjacent normal tissues (NAT) obtained from Ardais (Lexington, MA). Unmatched malignant and non-malignant RNA samples from lungs with gross histopathological assessment of tumor differentiation grade and stage and clinical state of the patient were obtained from Ardais.
ARDAIS Prostate vl.O
ARDAIS Prostate vl .0 panel included 2 controls and 68 test samples of human prostate malignancies and in some cases matched adjacent normal tissues (NAT) obtained from Ardais (Lexington, MA). RNA from unmatched malignant and non-malignant prostate samples with gross histopathological assessment of tumor differentiation grade and stage and clinical state of the patient were also obtained from Ardais. ARDAIS Kidney vl.O
ARDAIS Kidney vl.O panel included 2 control wells and 44 test samples of human renal cell carcinoma and in some cases matched adjacent normal tissue (NAT) obtained from Ardais (Lexington, MA). RNA from unmatched renal cell carcinoma and normal tissue with gross histopathological assessment of tumor differentiation grade and stage and clinical state of the patient were also obtained from Ardais.
ARDAIS Breast vl.O ARDAIS Breast vl .0 panel included 2 control wells and 71 test samples of human breast malignancies and in some cases matched adjacent normal tissue (NAT) obtained from Ardais (Lexington, MA). RNA from unmatched malignant and non-malignant breast samples with gross histopathological assessment of tumor differentiation grade and stage and clinical state of the patient were also obtained from Ardais.
Panels 3D, 3.1 and 3.2
Panels 3D, 3.1, and 3.2 included two controls, 92 cDNA samples of cultured human cancer cell lines and 2 samples of human primary cerebellum. Cell lines (ATCC, National Cancer Institute (NCI), German tumor cell bank) were cultured as recommended and were derived from: squamous cell carcinoma of the tongue, melanoma, sarcoma, leukemia, lymphoma, and epidermoid, bladder, pancreas, kidney, breast, prostate, ovary, uterus, cervix, stomach, colon, lung and CNS carcinomas.
Panels 4D, 4R, and 4.1D
Panels 4D, 4R, and 4.1D included 2 control wells and 94 test samples of RNA (Panel 4R) or cDNA (Panels 4D and 4. ID) from human cell lines or tissues related to inflammatory conditions. Controls included total RNA from normal tissues such as colon, lung (Stratagene, La Jolla, CA), thymus and kidney (Clontech, Palo Alto, CA). Total RNA from cirrhotic and lupus kidney was obtained from BioChain Institute, Inc.,
(Hayward, CA). Crohn's intestinal and ulcerative colitis samples were obtained from the National Disease Research Interchange (NDRI, Philadelphia, PA). Cells purchased from Clonetics (WalkersviUe, MD) included: astrocytes, lung fibroblasts, dermal fibroblasts, coronary artery smooth muscle cells, small airway epithelium, bronchial epithelium, microvascular dermal endothelial cells, microvascular lung endothelial cells, human pulmonary aortic endothelial cells, and human umbilical vein endothelial. These primary cell types were activated by incubating with various cytokines (IL-1 beta -1-5 ngml, TNF alpha -5-10 ng/ml, IFN gamma -20-50 ng/ml, IL-4 -5-10 ng/ml, IL-9 -5-10 ng/ml, IL-13 5-10 ng/ml) or combinations of cytokines as indicated. Starved endothelial cells were cultured in the basal media (Clonetics, WalkersviUe, MD) with 0.1% serum.
Mononuclear cells were prepared from blood donations using Ficoll. LAK cells were cultured in culture media [DMEM, 5% FCS (Hyclone, Logan, UT), 100 μM non essential amino acids (Gibco/Life Technologies, Rockville, MD), 1 mM sodium pyruvate (Gibco), mercaptoethanol 5.5 x 10"5 M (Gibco), and 10 mM Hepes (Gibco)] and interleukin 2 for 4-6 days. Cells were activated with 10-20 ng/ml PMA and 1-2 μg/ml ionomycin, 5-10 ng/ml IL-12, 20-50 ng/ml IFN gamma or 5-10 ng ml IL-18 for 6 hours. In some cases, mononuclear cells were cultured for 4-5 days in culture media with -5 μg/ml PHA (phytohemagglutinin) or PWM (pokeweed mitogen; Sigma-Aldrich Coφ., St. Louis, MO). Samples were taken at 24, 48 and 72 hours for RNA preparation. MLR (mixed lymphocyte reaction) samples were obtained by taking blood from two donors, isolating the mononuclear cells using Ficoll and mixing them 1:1 at a final concentration of -2x106 cells/ml in culture media. The MLR samples were taken at various time points from 1- 7 days for RNA preparation.
Monocytes were isolated from mononuclear cells using CD 14 Miltenyi Beads, +ve VS selection columns and a Vario Magnet (Miltenyi Biotec, Auburn, CA) according to the manufacturer's instructions. Monocytes were differentiated into dendritic cells by culturing in culture media with 50 ng/ml GMCSF and 5 ng/ml IL-4 for 5-7 days. Macrophages were prepared by culturing monocytes for 5-7 days in culture media with -50 ng/ml 10% type AB Human Serum (Life technologies, Rockville, MD) or MCSF (Macrophage colony stimulating factor; R&D, Minneapolis, MN). Monocytes, macrophages and dendritic cells were stimulated for 6 or 12-14 hours with 100 ng/ml lipopolysaccharide (LPS). Dendritic cells were also stimulated with 10 μg/ml anti-CD40 monoclonal antibody (Pharmingen, San Diego, CA) for 6 or 12-14 hours. CD4+ lymphocytes, CD8+ lymphocytes and NK cells were also isolated from mononuclear cells using CD4, CD8 and CD56 Miltenyi beads, positive VS selection columns and a Vario Magnet (Miltenyi Biotec, Auburn, CA) according to the manufacturer's instructions. CD45+RA and CD45+RO CD4+ lymphocytes were isolated by depleting mononuclear cells of CD8+, CD56+, CD14+ and CDl 9+ cells using CD8, CD56, CD 14 and CD 19 Miltenyi beads and positive selection. CD45RO Miltenyi beads were then used to separate the CD45+RO CD4+ lymphocytes from CD45+RA CD4+ lymphocytes. CD45+RA CD4+, CD45+RO CD4 +and CD8+ lymphocytes were cultured in culture media at 106 cells/ml in culture plates precoated overnight with 0.5 μg/ml anti-CD28 (Pharmingen, San Diego, C A) and 3 μg/ml anti-CD3 (OKT3, ATCC) in PBS. After 6 and 24 hours, the cells were harvested for RNA preparation. To prepare chronically activated CD8+ lymphocytes, isolated CD8+ lymphocytes were activated for 4 days on anti-CD28, anti-CD3 coated plates and then harvested and expanded in culture media with IL-2 (1 ng/ml). These CD8+ cells were activated again with plate bound anti-CD3 and anti-CD28 for 4 days and expanded as described above. RNA was isolated 6 and 24 hours after the second activation and after 4 days of the second expansion culture. Isolated NK cells were cultured in culture media with 1 ng/ml IL-2 for 4-6 days before RNA was prepared.
B cells were prepared from minced and sieved tonsil tissue (NDRI). Tonsil cells were pelleted and resupended at 106 cells/ml in culture media. Cells were activated using 5 μg/ml PWM (Sigma-Aldrich Coφ., St. Louis, MO) or -10 μg/ml anti-CD40 (Pharmingen, San Diego, CA) and 5-10 ng/ml IL-4. Cells were harvested for RNA preparation after 24, 48 and 72 hours.
To prepare primary and secondary Thl/Th2 and Trl cells, umbilical cord blood CD4+ lymphocytes (Poietic Systems, German Town, MD) were cultured at 105106cells/ml in culture media with IL-2 (4 ng/ml) in 6-well Falcon plates (precoated overnight with 10 μg/ml anti-CD28 (Pharmingen) and 2 μg/ml anti-CD3 (OKT3; ATCC) then washed twice with PBS) .
To stimulate Thl phenotype differentiation, IL-12 (5 ng/ml) and anti-IL4 (1 μg/ml) were used; for Th2 phenotype differentiation, IL-4 (5 ng/ml) and anti-IFN gamma (1 μg/ml) were used; and for Trl phenotype differentiation, IL-10 (5 ng/ml) was used. After 4-5 days, the activated Thl, Th2 and Trl lymphocytes were washed once with DMEM and expanded for 4-7 days in culture media with IL-2 (1 ng/ml). Activated Thl, Th2 and Trl lymphocytes were re-stimulated for 5 days with anti-CD28/CD3 and cytokines as described above with the addition of anti-CD95L (1 μg/ml) to prevent apoptosis. After 4-5 days, the Thl, Th2 and Trl lymphocytes were washed and expanded in culture media with IL-2 for 4-7 days. Activated Thl and Th2 lymphocytes were maintained for a maximum of three cycles. RNA was prepared from primary and secondary Thl, Th2 and Trl after 6 and 24 hours following the second and third activations with plate-bound anti-CD3 and anti-CD28 mAbs and 4 days into the second and third expansion cultures.
Leukocyte cells lines Ramos, EOL-1, KU-812 were obtained from the ATCC. EOL-1 cells were further differentiated by culturing in culture media at 5 xlO5 cells/ml with 0.1 mM dbcAMP for 8 days, changing the media every 3 days and adjusting the cell concentration to 5 xlO5 cells/ml. RNA was prepared from resting cells or cells activated with PMA (10 ng/ml) and ionomycin (1 μg ml) for 6 and 14 hours. RNA was prepared from resting CCD 1106 keratinocyte cell line (ATCC) or from cells activated with -5 ng/ml TNF alpha and 1 ng/ml IL-1 beta. RNA was prepared from resting NCI-H292, airway epithelial tumor cell line (ATCC) or from cells activated for 6 and 14 hours in culture media with 5 ng/ml IL-4, 5 ng/ml IL-9, 5 ng/ml IL-13, and 25 ng/ml IFN gamma.
RNA was prepared by lysing approximately 107 cells/ml using Trizol (Gibco BRL) then adding 1/10 volume of bromochloropropane (Molecular Research Coφoration, Cincinnati, OH), vortexing, incubating for 10 minutes at room temperature and then spinning at 14,000 φm in a Sorvall SS34 rotor. The aqueous phase was placed in a 15 ml Falcon Tube and an equal volume of isopropanol was added and left at -20° C overnight. The precipitated RNA was spun down at 9,000 φm for 15 min and washed in 70% ethanol. The pellet was redissolved in 300 μl of RNAse-free water with 35 μl buffer (Promega, Madison, WI) 5 μl DTT, 7 μl RNAsin and 8 μl DNAse and incubated at 37° C for 30 minutes to remove contaminating genomic DNA, extracted once with phenol chloroform and re-precipitated with 1/10 volume of 3 M sodium acetate and 2 volumes of
100% ethanol. The RNA was spun down, placed in RNAse free water and stored at
-80° C. Al comprehensive panel vl.O
Autoimmunity (Al) comprehensive panel vl.O included two controls and 89 cDNA test samples isolated from male (M) and female (F) surgical and postmortem human tissues that were obtained from the Backus Hospital and Clinomics (Frederick, MD). Tissue samples included : normal, adjacent (Adj); matched normal adjacent (match control); joint tissues (synovial (Syn) fluid, synovium, bone and cartilage, osteoarthritis (OA), rheumatoid arthritis (RA)); psoriatic; ulcerative colitis colon; Crohns disease colon; and emphysmatic, asthmatic, allergic and chronic obstructive pulmonary disease (COPD) lung.
Pulmonary and General inflammation (PGI) panel vl.O
Pulmonary and General inflammation (PGI) panel vl .0 included two controls and 39 test samples isolated as surgical or postmortem samples. Tissue samples include: five normal lung samples obtained from Maryland Brain and Tissue Bank, University of Maryland (Baltimore, MD), International Bioresource systems, IBS (Tuscon, AZ), and Asterand (Detroit, MI), five normal adjacent intestine tissues (NAT) from Ardais (Lexington, MA), ulcerative colitis samples (UC) from Ardais (Lexington, MA); Crohns disease colon from NDRI, National Disease Research Interchange (Philadelphia, PA); emphysematous tissue samples from Ardais (Lexington, MA) and Genomic Collaborative Inc. (Cambridge, MA), asthmatic tissue from Maryland Brain and Tissue Bank, University of Maryland (Baltimore, MD) and Genomic Collaborative Inc (Cambridge, MA) and fibrotic tissue from Ardais (Lexinton, MA) and Genomic Collaborative (Cambridge, MA).
AI.05 chondrosarcoma
AI.05 chondrosarcoma plates included SW1353 cells (ATCC) subjected to serum starvation and treated for 6 and 18 h with cytokines that are known to induce MMP (1, 3 and 13) synthesis (e.g. ILlbeta). These treatments included: IL-lbeta (10 ng/ml), IL-lbeta + TNF-alpha (50 ng/ml), IL-lbeta + Oncostatin (50 ng/ml) and PMA (100 ng/ml). Supernatants were collected and analyzed for MMP 1, 3 and 13 production. RNA was prepared from these samples using standard procedures. Panels 5D and 51
Panel 5D and 51 included two controls and cDNAs isolated from human tissues, human pancreatic islets cells, cell lines, metabolic tissues obtained from patients enrolled in the Gestational Diabetes study (described below), and cells from different stages of adipocyte differentiation, including differentiated (AD), midway differentiated (AM), and undifferentiated (U; human mesenchymal stem cells).
Gestational Diabetes study subjects were young (18 - 40 years), otherwise healthy women with and without gestational diabetes undergoing routine (elective) Caesarean section. Uterine wall smooth muscle (UT), visceral (Vis) adipose, skeletal muscle (SK), placenta (PI) greater omentum adipose (GO Adipose) and subcutaneous (SubQ) adipose samples (<1 cc) were collected, rinsed in sterile saline, blotted and flash frozen in liquid nitrogen. Patients included: Patient 2, an overweight diabetic Hispanic not on insulin; Patient 7-9, obese non-diabetic Caucasians with body mass index (BMI) greater than 30; Patient 10, an overweight diabetic Hispanic, on insulin; Patient 11, an overweight nondiabetic African American; and Patient 12, a diabetic Hispanic on insulin.
Differentiated adipocytes were obtained from induced donor progenitor cells (Clonetics, WalkersviUe, MD). Differentiated human mesenchymal stem cells (HuMSCs) were prepared as described in Mark F. Pittenger, et al., Multilineage Potential of Adult Human Mesenchymal Stem Cells Science Apr 2 1999: 143-147. mRNA was isolated and sscDNA was produced from Trizol lysates or frozen pellets. Human cell lines (ATCC, NCI or German tumor cell bank) included: kidney proximal convoluted tubule, uterine smooth muscle cells, small intestine, liver HepG2 cancer cells, heart primary stromal cells and adrenal cortical adenoma cells. Cells were cultured, RNA extracted and sscDNA was produced using standard procedures
Panel 51 also contains pancreatic islets (Diabetes Research Institute at the University of Miami School of Medicine). Human Metabolic RTQ-PCR Panel
Human Metabolic RTQ-PCR Panel included two controls (genomic DNA control and chemistry control) and 211 cDNAs isolated from human tissues and cell lines relevant to metabolic diseases. This panel identifies genes that play a role in the etiology and pathogenesis of obesity and/or diabetes. Metabolic tissues including placenta (PI), uterine wall smooth muscle (Ut), visceral adipose, skeletal muscle (Sk) and subcutaneous (SubQ) adipose were obtained from the Gestational Diabetes study (described above). Included in the panel are: Patients 7 and 8, obese non-diabetic Caucasians; Patient 12 a diabetic Caucasian with unknown BMI, on insulin (treated); Patient 13, an overweight diabetic Caucasian, not on insulin (untreated); Patient 15, an obese, untreated, diabetic Caucasian; Patient 17 and 25, untreated diabetic Caucasians of normal weight; Patient 18, an obese, untreated, diabetic Hispanic; Patient 19, a non-diabetic Caucasian of normal weight; Patient 20, an overweight, treated diabetic Caucasian; Patient 21 and 23, overweight non-diabetic Caucasians; Patient 22, a treated diabetic Caucasian of normal weight;
Patient 23, an overweight non-diabetic Caucasian; and Patients 26 and 27, obese , treated, diabetic Caucasians.
Total RNA was isolated from metabolic tissues including: hypothalamus, liver, pancreas, pancreatic islets, small intestine, psoas muscle, diaphragm muscle, visceral (Vis) adipose, subcutaneous (SubQ) adipose and greater omentum (Go) from 12 Type II diabetic (Diab) patients and 12 non diabetic (Norm) at autopsy. Control diabetic and non-diabetic subjects were matched where possible for: age; sex, male (M); female (F); ethnicity, Caucasian (CC); Hispanic (HI); African American (AA); Asian (AS); and BMI, 20-25 (Low BM), 26-30 (Med BM) or overweight (Overwt), BMI greater than 30 (Hi BMI) (obese).
RNA was extracted and ss cDNA was produced from cell lines (ATCC) by standard methods.
CNS Panels
CNS Panels CNSD.01, CNS Neurodegeneration Vl.O and CNS Neurodegeneration V2.0 included two controls and 46 to 94 test cDNA samples isolated from postmortem human brain tissue obtained from the Harvard Brain Tissue Resource Center (McLean Hospital). Brains were removed from calvaria of donors between 4 and 24 hours after death, and frozen at -80°C in liquid nitrogen vapor.
Panel CNSD.01
Panel CNSD.01 included two specimens each from: Alzheimer's disease, Parkinson's disease, Huntington's disease, Progressive Superauclear Palsy (PSP), Depression, and normal controls. Collected tissues included: cingulate gyrus (Cing Gyr), temporal pole (Temp Pole), globus palladus (Glob palladus), substantia nigra (Sub Nigra), primary motor strip (Brodman Area 4), parietal cortex (Brodman Area 7), prefrontal cortex (Brodman Area 9), and occipital cortex (Brodman area 17). Not all brain regions are represented in all cases.
Panel CNS Neurodegeneration Vl.O The CNS Neurodegeneration V 1.0 panel included: six Alzheimer's disease (AD) brains and eight normals which included no dementia and no Alzheimer's like pathology (control) or no dementia but evidence of severe Alzheimer's like pathology (Control Path), specifically senile plaque load rated as level 3 on a scale of 0-3; 0 no evidence of plaques, 3 severe AD senile plaque load. Tissues collected included: hippocampus, temporal cortex (Brodman Area 21), parietal cortex (Brodman area 7), occipital cortex (Brodman area 17) superior temporal cortex (Sup Temporal Ctx) and inferior temporal cortex (Inf Temproal Ctx).
Gene expression was analyzed after normalization using a scaling factor calculated by subtracting the Well mean (CT average for the specific tissue) from the Grand mean (average CT value for all wells across all runs). The scaled CT value is the result of the raw CT value plus the scaling factor.
Panel CNS Neurodegeneration V2.0 The CNS Neurodegeneration V2.0 panel included sixteen cases of Alzheimer's disease (AD) and twenty-nine normal controls (no evidence of dementia prior to death) including fourteen controls (Control) with no dementia and no Alzheimer's like pathology and fifteen controls with no dementia but evidence of severe Alzheimer's like pathology (AH3), specifically senile plaque load rated as level 3 on a scale of 0-3; 0 no evidence of plaques, 3 severe AD senile plaque load. Tissues from the temporal cortex (Brodman Area 21) included the inferior and superior temporal cortex that was pooled from a given individual (Inf & Sup Temp Ctx Pool).
A. CGI 03945-02: Semaphorin sem2.
Expression of gene CG103945-02 was assessed using the primer-probe set Ag7442, described in Table AA. Results of the RTQ-PCR runs are shown in Tables AB and AC.
Table AA. Probe Name Ag7442
Figure imgf000310_0001
Table AB. General screening panel yl.7
Figure imgf000310_0002
Figure imgf000311_0001
Table AC. Panel 4.1D
Figure imgf000311_0002
Figure imgf000312_0001
General_screening_panel_vl.7 Summary: Ag7442 Highest expression of the CG103945-02 gene was detected in adipose tissue (CT=29.5). In addition, significant expression of this gene was also seen in skeletal muscle and thyroid. Therapeutic modulation of the activity of this gene or its protein product is useful in the treatment of metabolic disorders, including diabetes and obesity. Moderate levels of expression of this gene were also seen in all regions of the central nervous system examined, including amygdala, hippocampus, substantia nigra, thalamus, cerebellum, cerebral cortex, and spinal cord. Therapeutic modulation of the activity of this gene or its protein product is useful in the treatment of central nervous system disorders such as Alzheimer's disease, Parkinson's disease, epilepsy, multiple sclerosis, schizophrenia and depression.
Moderate to low expression of this gene was seen in number of cancer cell lines derived from colon, lung, breast and ovarian cancers. Therapeutic modulation of the activity of this gene or its protein product is useful in the treatment of colon, lung, breast and ovarian cancers.
Panel 4.1D Summary: Ag7442 Highest expression of this gene was seen in lung microvascular endothelium (CT=33.6) and its expression was down-regulated upon activation of these cells. Endothelial cells are known to play important roles in inflammatory responses by altering the expression of surface proteins that are involved in activation and recruitment of effector inflammatory cells. Higher expression of this gene in resting cells suggests a role for this gene in the maintenance of the integrity of the lung microvasculature. Therapeutic modulation of the activity of this gene or its protein product is beneficial for the treatment of diseases associated with damaged microvasculature including heart diseases or inflammatory diseases, such as psoriasis, asthma, and chronic obstructive pulmonary diseases.
B. CG106951-01 and CG106951-04: Semaphorin 5B.
Expression of gene CG106951-01 and CG106951-04 was assessed using the primer-probe sets Agl216, described in Tables BA. Table BA. Probe Name Agl216
Figure imgf000314_0001
Table BB. AI comprehensive panel yl.O
Figure imgf000314_0002
Figure imgf000315_0002
Table BC. General screening panel yl.4
Figure imgf000315_0001
Figure imgf000316_0001
Table BD. Panel 3D
Figure imgf000316_0002
Figure imgf000317_0001
Table BE. Panel 4D
Figure imgf000318_0001
Figure imgf000319_0001
Table BF. Panel 5 Islet
Figure imgf000319_0002
Figure imgf000320_0001
Table BG. general oncology screening panel v 2.4
Figure imgf000320_0002
AI_comprehensive panel_vl.0 Summary: Agl216 Highest expression of the CG106951-01 and CG106951-04 genes was detected in a sample orthoarthritis bone (CT=31.2). Moderate to low levels of expression of these genes were detected in samples derived from normal and orthoarthitis/ rheumatoid arthritis bone, cartilage, synovium and synovial fluid samples, as well as in normal lung, COPD lung, emphysema, atopic asthma, asthma, allergy, Crohn's disease (normal matched control and diseased), ulcerative colitis (normal matched confrol and diseased), and psoriasis (normal matched control and diseased). Therapeutic modulation of the activity of these genes or their protein products will ameliorate symptoms/conditions associated with autoimmune and inflammatory disorders including psoriasis, allergy, asthma, inflammatory bowel disease, rheumatoid arthritis and osteoarthritis.
General_screening_panel_vl.4 Summary: Agl216 Highest expression of these genes was detected in renal cancer cell line 786-0 (CT=26.4). High to moderate expression of these genes was also seen in number of cancer cell lines derived from ovarian, breast, brain and kidney cancers. Therapeutic modulation of the activity of these genes or their protein products is useful in the treatment of these cancers.
Among tissues with metabolic or endocrine function, these genes were expressed at moderate to low levels in pancreas, adipose, adrenal gland, pituitary gland, skeletal muscle, heart, fetal liver and the gastrointestinal tract. Therapeutic modulation of the activity of these genes or their protein products is useful in the treatment of endocrine/metabolically related diseases, such as obesity and diabetes.
In addition, these genes were expressed at moderate levels in all regions of the central nervous system examined, including amygdala, hippocampus, substantia nigra, thalamus, cerebellum, cerebral cortex, and spinal cord. Therapeutic modulation of the activity of these genes or their protein products is useful in the treatment of central nervous system disorders such as Alzheimer's disease, Parkinson's disease, epilepsy, multiple sclerosis, schizophrenia and depression.
The CGI 06951-01 and CGI 06951-04 genes were also expressed at higher levels in fetal (CTs=29-32) liver, lung, heart, kidney and skeletal muscle when compared to adult tissues (CTs=33-37). The relative overexpression of these genes in fetal tissue suggests that the expressed proteins may enhance growth or development of these tissues in the fetus and thus may also act in a regenerative capacity in the adult. Therapeutic modulation of the activity of these genes or their protein products is useful in treatment of liver, lung, kidney, heart and skeletal muscle related diseases. Panel 3D Summary: Agl216 Moderate expression of these genes were detected mainly in a lung cancer DMS-79 cell line (CT=30.4). Therapeutic modulation of the activity of these genes or their protein products is useful in the freatment of lung cancer.
Panel 4D Summary: Agl216 Highest expression of these genes was detected in thymus (CTs=31-32). These genes also show low expression in normal lung as well as in astrocytes and bronchial epithelium treated with TNF- and IL-1 β. Therapeutic modulation of the activity of these genes or their protein products is useful in the freatment of inflammatory diseases including asthma, allergies, inflammatory bowel disease, lupus erythematosus, psoriasis, rheumatoid arthritis, and osteoarthritis.
Panel 5 Islet Summary: Agl216 Highest expression of these genes were detected in placenta (CT=34). Low expression of these genes was also seen in adipose and uterus. Please see panel 1.4 for further discussion of these genes.
General oncolo y screening panel_v_2.4 Summary: Agl216 Highest expression of these genes was detected in a kidney cancer sample (CT=27). Expression of these genes was higher in 4/4 kidney cancer, 3/3 colon cancer, and 3/3 lung cancer samples relative to corresponding normal adjacent tissue. In addition, significant expression of these genes was also seen in metastatic melanoma and prostate cancers. Gene or protein expression levels are useful as a marker to detect the presence of these cancers. Therapeutic modulation of the activity of these genes or their protein products using nucleic acid, protein, antibody or small molecule drugs is useful in the treatment of kidney, lung, colon, metastatic melanoma and prostate cancers.
C. CG124756-01: Complement component 1, q subcomponent, beta polypeptide.
Expression of gene CGI 24756-01 was assessed using the primer-probe set Ag4901, described in Table CA. Results of the RTQ-PCR runs are shown in Tables CB and CC. Table CA. Probe Name Ag4901
Figure imgf000323_0001
Table CB. CNS neurodegeneration yl.O
Figure imgf000323_0002
Table CC. General screening panel yl.6
Figure imgf000324_0001
Figure imgf000325_0001
CNS_neurodegeneration_vl.0 Summary: Ag4901 Expression of the CG124756-01 gene was upregulated in the temporal cortex of Alzheimer's disease patients compared to normal patients. Inhibition of this gene or its protein product is useful in the treatment of Alzheimer's disease and can decrease neuronal death.
General_screenin _panel_vl.6 Summary: Ag4901 The highest expression of this gene was detected in bladder (CT=26). In addition, this gene was expressed at high to moderate levels in pancreas, adipose, adrenal gland, thyroid, pituitary gland, skeletal muscle, heart, liver and the gastrointestinal tract. Therapeutic modulation of the activity of this gene or its protein product is useful in the treatment of endocrine/metabolically related diseases, such as obesity and diabetes.
This gene was also expressed at moderate levels in all regions of the central nervous system examined, including amygdala, hippocampus, substantia nigra, thalamus, cerebellum, cerebral cortex, and spinal cord. Thereapeutic modulation of the activity of this gene or its protein product using nucleic acid, protein, antibody or small molecule drugs is useful in the treatment of central nervous system disorders such as Alzheimer's disease, Parkinson's disease, epilepsy, multiple sclerosis, schizophrenia and depression.
High expression of this gene was also seen in a colon cancer cell line. Therapeutic modulation of the activity of this gene or its protein product is useful in the treatment of colon cancer.
D. CG50353-01: 129293352_EXT, Wnt 7a like protein.
Expression of gene CG50353-01 was assessed using the primer-probe set Ag3093, described in Table DA. Results of the RTQ-PCR runs are shown in Tables DB and DC. Table DA. Probe Name Ag3093
Figure imgf000326_0001
Table DB. Panel 1.3D
Figure imgf000326_0002
Figure imgf000327_0001
Table DC. Panel 4D
Figure imgf000327_0002
Figure imgf000328_0001
Panel 1.3D Summary: Ag3093 Highest expression of the CG50353-01 gene was detected in the SK-OV-3 ovarian cancer cell line derived from ascites fluid (CT=30.28). This gene was also expressed in two additional ovarian cancer cell lines. Gene or protein expression levels are useful as a marker for ovarian cancer or for ascites. Therapeutic modulation of the activity of this gene or its protein product using nucleic acid, protein, antibody or small molecule drugs is useful in the treatment of ovarian cancer.
Panel 4D Summary: This gene was expressed at the highest level in TNF alpha + IL-1 beta-treated small airway epithelial cells (CT=32.6) and bronchial epithelial cells as well as in CCDl 106 keratinocytes, independent of treatment. Expression of this gene in keratinocytes suggests that it is important in skin disorders including psoriasis. Expression of this gene in airway/bronchial cell types suggests that this gene also plays a role in inflammatory lung disorders, including, for example, chronic obstructive pulmonary disease (COPD), asthma, allergy and emphysema. Therapeutic modulation of the activity of this gene or its protein product is useful in the treatment of skin disorders, such as psoriasis, and inflammatory lung disorders, including COPD, asthma, allergy and emphysema.
E. CG50709-03 and CG50709-05: WNT14B
Expression of genes CG50709-03 and CG50709-05 was assessed using the primer-probe sets Ag2262, and Ag2316, described in Tables EA, and EB. Results of the RTQ-PCR runs are shown in Tables EC, ED, EE, and EF.
Table EA. Probe Name Ag2262
Figure imgf000329_0002
Table EB. Probe Name Ag2316
Figure imgf000329_0003
Table EC. Ardais Kidney 1.0
Figure imgf000329_0001
Figure imgf000330_0002
Table ED. Panel 1.3D
Figure imgf000330_0001
Figure imgf000331_0001
Table EE. Panel 2D
Figure imgf000331_0002
Figure imgf000332_0001
Table EF. Panel 4D
Figure imgf000333_0001
Figure imgf000334_0001
Ardais Kidney 1.0 Summary: Ag2262 Highest expression of the CG50709-03 and CG50709-05 genes was detected in a normal kidney sample (CT=27.6). In many cases, expression of these genes was higher in normal adjacent kidney samples relative to the tumors. The results from Panel 1.3D indicate that these genes were also more highly expressed in fetal as compared to adult kidney. This expression profile suggests that the function of these genes is to drive and/or maintain differentiation of kidney epithelium, since loss of differentiation is a hallmark of kidney cancer. Gene or protein expression levels are useful to distinguish normal kidney from kidney cancer. Therapeutic modulation of the activity of these genes or their protein products using nucleic acid, protein, antibody, or small molecule drugs is useful in the treatment of kidney cancer.
Panel 1.3D Summary: Ag2262 Significant expression of these genes was seen mainly in spleen and fetal kidney (CTs=30-32) with upregulated expression in fetal relative to adult kidney. Please see Ardias Kidney vl.O panel for further discussion of these genes.
Panel 2D Summary: Ag2262 Expression of these genes was highest in a sample derived from normal kidney tissue (CT=32.6) and was generally higher in normal kidney tissue relative to adjacent malignant tissue. This expression profile is in agreement with that seen in the Ardias kidney vl .0 panel. Therapeutic modulation of these genes or their protein products using nucleic acid, protein, antibody or small molecule drugs that increase the activity of these genes is useful in the treatment of kidney cancers. Panel 4D Summary: Ag2316 Significant expression of these genes was seen exclusively in thymus (CT=33). These genes encode variants of a Wntl4B-like protein; other members of this protein family are known to regulate cell differentiation. The encoded Wnt 14-like proteins may play an important role in T cell development. Therapeutic modulation of the activity of these genes or their protein products is useful to modulate immune function (T cell development) and for organ transplant, AIDS freatment or post chemotherapy immune reconstitiution.
Ag 2262 The Wnt 14B variant recognized by this probe-primer set was significantly expressed in colon, lung and thymus (CT=33-34.7). This gene may play an important role in the normal homeostasis of these tissues. Therapeutic modulation of the activity of this gene or its protein product is useful in maintaining or restoring normal function to these organs during inflammation.
F. CG53054-02: WNT-14 PROTEIN PRECURSOR
Expression of gene CG53054-02 was assessed using the primer-probe sets Ag2261 and Ag3035, described in Tables FA and FB. Results of the RTQ-PCR runs are shown in Tables FC, FD, FE, FF, FG, FH, FI and FJ.
Table FA. Probe Name Ag2261
Start SEQ ID
Primers (Sequences Length Position No
[Forward J5 ' -ggatgactcgcctagcttct-3 20 882 301 Probe lτET-5 ' -gccgtaggtgccaccgtgagaag-3 ' -TAMRA 23 935 1302
Reverse J5 ' -agcagatgctctcgcagtt-3 19 958 303
Table FB. Probe Name Ag3035
Figure imgf000335_0001
Table FC.AI comprehensive panel yl.O
Figure imgf000336_0001
Figure imgf000337_0001
Table FD. Oncology cell line screening panel v3.2
Figure imgf000337_0002
Figure imgf000338_0001
Figure imgf000339_0001
Table FE. Panel 1.3D
Figure imgf000339_0002
Figure imgf000340_0001
Table FF. Panel 2D
Figure imgf000340_0002
Figure imgf000341_0001
Table FG. Panel 4.1D
Figure imgf000342_0001
Figure imgf000343_0001
Table FH. Panel 4D
Figure imgf000343_0002
Figure imgf000344_0001
Table FI. Panel 5 Islet
Figure imgf000344_0002
Figure imgf000345_0001
Table FJ. general oncology screening panel v 2.4
Figure imgf000345_0002
Figure imgf000346_0001
AI_comprehensive panel_vl.0 Summary: Ag3035 Highest expression of the CG53054-02 gene was detected in a COPD sample (CT=30). This gene shows widespread expression in this panel. Moderate levels of expression of this gene were detected in samples derived from normal and orthoarthitis/ rheumatoid arthritis bone, cartilage, synovium and synovial fluid samples, as well as from normal lung, COPD lung, emphysema, atopic asthma, asthma, allergy, Crohn's disease (normal matched control and diseased), ulcerative colitis (normal matched control and diseased), and psoriasis (normal matched control and diseased). Therapeutic modulation of the activity of this gene or its protein product will ameliorate symptoms/conditions associated with autoimmune and inflammatory disorders, including psoriasis, allergy, asthma, inflammatory bowel disease, rheumatoid arthritis and osteoarthritis.
Oncology_cell_line_screeningjpanel_v3.2 Summary: Ag3035 Highest expression of this gene was seen in lung cancer cell line DMS-79 (CT=28.6). Moderate to low expression of this gene was also seen in number of cancer cell lines derived from tongue, bone, bladder, pancreatic, cervical, uterine, gastric, colon and lung cancer. Gene or protein expression levels are useful as a marker to detect the presence of these cancers. Therapeutic modulation of the activity of this gene or its protein product is useful in the treatment of these cancers.
Panel 1.3D Summary: Ag2261 This gene was expressed at moderate levels in a number of metabolic tissues, with highest overall expression seen in fetal skeletal muscle (CTs=30.4-31.8). The higher levels of expression in fetal skeletal muscle when compared to adult skeletal muscle suggests that the protein product encoded by this gene may be useful in treating muscular dystrophy, Lesch-Nyhan syndrome, myasthenia gravis and other conditions that result in weak or dystrophic muscle. This gene was also expressed in adipose, thyroid and heart. Since biologic cross-talk between adipose and thyroid is a component of some forms of obesity, therapeutic modulation of the activity of this gene or its protein product is useful for the treatment of metabolic disease, including obesity and Type 2 diabetes.
Ag3035 This probe/primer set recognizes a distinct portion of this gene that shows a distinctive expression pattern when compared to Ag2261. This observation may indicate that the probe/primer sets can distinguish splice variants of this gene. In contrast to the results obtained with Ag2261, expression of this gene was highest in an ovarian cancer cell line (CT = 30.6). As was the case for Ag2261, expression of this gene using Ag3035 was also relatively high in fetal skeletal muscle. However, Ag3035 showed higher levels of gene expression in adult skeletal muscle as well as in adult and fetal heart. Most other expression is similar using both probe/primer sets. Please see Ag2261 for additional information.
Panel 2D Summary: Ag2261 This gene was consistently expressed in samples of breast cancer, uterine cancer and lung cancer relative to their respective normal adjacent tissue controls. Gene or protein expression levels are useful as marker to detect breast, uterine and lung cancers. Therapeutic modulation of the activity of this gene or its protein product using nucleic acid, protein, antibody or small molecule drugs is useful in the treatment of breast, lung or uterine cancers.
Panel 4.1D Summary: Ag3035 This probe/primer set recognizes a distinct portion of this gene and shows a distinctive expression pattern relative to probe/primer set Ag2261 in this panel. This observation may indicate that the probe/primer sets can distinguish splice variants of this gene. In contrast to the results obtained with Ag2261 (see panel 4D summary), expression of this gene was highest in normal kidney (CT = 30.6). The other expression results for this panel were similar using both probe/primer sets. This gene encodes a WNT-14 homolog and was expressed at moderate to low levels in unstimulated or cytokine-activated keratinocytes as well as in lung and dermal fibroblast preparations (CTs=29-34). Therapeutic modulation of the activity of this gene or its protein product will reduce or eliminate the symptoms of chronic obstructive pulmonary disease, asthma, emphysema, or psoriasis. In addition, due to its known effects on development of vertebrate joints, the protein encoded this gene will reduce or eliminate the symptoms of osetoarthritis (Christine Hartmann and Clifford J. Tabin, 2001, Wnt-14 Plays a Pivotal Role in Inducing Synovial Joint Formation in the Developing Appendicular Skeleton Cell, Nol 104, 341-35).
Panel 4D Summary: Ag2261 This gene was expressed at low levels in colon (CT=33.5). Low but significant levels of expression were also seen in normal lung, keratinocytes and dermal fibroblasts. This gene or the Wnt-14 protein encoded by it may play an important role in the normal homeostasis of these tissues. Therapeutic modulation of the activity of this gene or its protein product is useful to maintain or restore normal function to these organs during inflammation.
Panel 5 Islet Summary: Ag3035 Highest expression of this gene was seen in sample of skeletal muscle from a diabetic patient (CT=31.8). Significant expression of this gene was also seen in pancreatic islet cells. Therapeutic modulation of the activity of this gene or its protein product using nucleic acid, protein, antibody or small molecule drugs is useful in the treatment of metabolic related disease such as obesity and diabetes, especially type II diabetes.
General oncology screening panel_v_2.4 Summary: Ag3035 Highest expression of this gene was detected in a metastatic melanoma sample (CT=31.3). Expression of this gene was also upregulated in prostate, lung and kidney cancers when compared to their appropriate normal adjacent tissue. Gene or protein expression levels are useful for the detection of prostate cancer, lung cancer, kidney cancer and metastatic melanoma. Therapeutic modulation of the activity of this gene or its protein product using nucleic acid, protein, antibody or small molecule drugs is useful for the treatment of these cancers.
G. CG53473-02: NEUROMEDIN B-32 PRECURSOR
Expression of gene CG53473-02 was assessed using the primer-probe set Ag235, described in Table GA. Results of the RTQ-PCR runs are shown in Tables GB and GC.
Table GA. Probe Name Ag235
[Primers (Sequences Length Start Position JSEQ π) No
(Forward (5 ' -ttccagcccatccccatt-3 ' |18 225 |307
(Probe (TET-5 ' -ccccacacctccctgagggacc-3 ' -TAMRA 22 253 (308
Reverse J5 ' -cagatcatgactcagctgcagtc-3 ' |23 278 (309 Table GB. Panel 1.2
Figure imgf000349_0001
Figure imgf000350_0002
Table GC. Panel 2D
Figure imgf000350_0001
Figure imgf000351_0001
Panel 1.2 Summary: Ag235 Highest expression of the CG53473-02 gene was seen in adrenal gland (CTs=23-26). Significant expression of this gene was also detected in pancreas, adrenal gland, thyroid, pituitary gland, skeletal muscle, heart, liver and the gastrointestinal tract. Therapeutic modulation of the activity of this gene or its protein product is useful in the treatment of endocrine/metabolically related diseases, such as obesity and diabetes.
In addition, this gene was expressed at high to moderate levels in all regions of the central nervous system examined, including amygdala, hippocampus, substantia nigra, thalamus, cerebellum, cerebral cortex, and spinal cord. Therapeutic modulation of the activity of this gene or its protein product is useful in the treatment of central nervous system disorders such as Alzheimer's disease, Parkinson's disease, epilepsy, multiple sclerosis, schizophrenia and depression.
High expression of this gene was also seen in a number of cancer cell lines derived from pancreatic, gastric, colon, lung, liver, renal, breast, ovarian, prostate, squamous cell carcinoma, melanoma and brain cancers. Gene or protein expression levels are useful as a marker for these cancers. Therapeutic modulation of the activity of this gene or its protein product is useful in the treatment of pancreatic, gastric, colon, lung, liver, renal, breast, ovarian, prostate, melanoma and brain cancers.
Panel 2D Summary: Ag235 Highest expression of this gene was detected in a kidney cancer sample (CT=27.9). This gene was overexpressed in a number of kidney, gastric, ovarian, bladder, breast and lung cancers relative to the appropriate normal tissues. Gene or protein expression levels are useful for the detection of these cancers. Therapeutic modulation of the activity of this gene or its protein product using nucleic acid, protein, antibody or small molecule drugs is useful in the treatment of kidney, gastric, ovarian, bladder, breast and lung cancer.
H. CG55184-03: Cerebellin
Expression of gene CG55184-03 was assessed using the primer-probe set Agl 161, described in Table HA. Results of the RTQ-PCR runs are shown in Tables HB, HC, HD and HE.
Table HA. Probe Name Agllθl
Figure imgf000352_0002
Table HB. CNS neurodegeneration yl.O
Figure imgf000352_0001
Figure imgf000353_0001
Table HC. General screening panel yl.7
Column A - Rel . Exp. (%) Agllόl, Run 317667428
Tissue Name A Tissue Name A
Adipose 0.4 Gastric ca. (liver met.) NCI-N87 0.0
HUVEC 0.0 Stomach 0.0
Melanoma* Hs688(A).T 0.0 Colon ca. SW-948 0.1
Melanoma* Hs688(B).T 0.0 Colon ca. SW480 0.0
Melanoma (met) SK-MEL-5 0.0 Colon ca. (SW480 met) SW620 0.0
Testis 5.6 Colon ca. HT29 0.0
Prostate ca. (bone met) PC-3 0.0 Colon ca. HCT-116 0.0 Prostate ca. DU145 0.0 Colon cancer tissue 0.1
Prostate pool 0.1 Colon ca. SW1116 0.0
Uterus pool 0.1 Colon ca. Colo-205 0.0
Ovarian ca. OVCAR-3 0.0 Colon ca. SW-48 0.0
Ovarian ca. (ascites) SK-OV-3 0.0 Colon 0.0
Ovarian ca. OVCAR-4 0.0 Small Intestine 0.3
Ovarian ca. OVCAR-5 0.0 Fetal Heart 0.0
Ovarian ca. IGROV-1 0.0 Heart 0.2
Ovarian ca. OVCAR-8 0.0 jJLymph Node pool 1 0.8
Ovary 12.6 Lymph Node pool 2 0.1
Breast ca. MCF-7 0.1 Fetal Skeletal Muscle 0.2
Breast ca. MDA-MB-231 0.0 Skeletal Muscle pool 0.6
Breast ca. BT-549 0.0 Skeletal Muscle 1.8
Breast ca. T47D 0.0 Spleen 3.2
Breast pool 0.8 Thymus 1.1
Trachea 0.6 CNS cancer (glio/astro) SF-268 0.0
Figure imgf000354_0001
Table HP. Panel 2.2
Figure imgf000354_0002
Figure imgf000355_0001
Table HE. Panel 4D
Figure imgf000355_0002
Figure imgf000356_0001
CNS_neurodegeneration_vl.0 Summary: Agl 161 Expression of the CG55184-03 gene was down-regulated in the temporal cortex of Alzheimer's disease patients when compared with normal patients. Therefore, up-regulation of this gene or its protein product or treatment with specific agonists for this receptor is useful in reversing the dementia/memory loss and neuronal death associated with this disease.
General_screening_panel_vl.7 Summary: Agl 161 Highest expression of this gene was detected in whole brain (CT=26). This gene showed brain preferential expression with high expression seen in all the regions of the central nervous system examined, including amygdala, hippocampus, substantia nigra, thalamus, cerebellum, cerebral cortex, and spinal cord. Gene or protein expression levels are useful as a marker for brain.
Therapeutic modulation of the activity of this gene or its protein product is useful in the treatment of central nervous system disorders such as Alzheimer's disease, Parkinson's disease, epilepsy, multiple sclerosis, schizophrenia and depression.
Moderate to low expression of this gene was also seen in tissues with metabolic/endocrine function including pancreas, adipose, adrenal gland, thyroid, skeletal muscle, liver and small intestine. Therapeutic modulation of the activity of this gene or its protein product is useful in the treatment of endocrine/metabolically related diseases, such as obesity and diabetes.
Panel 2.2 Summary: Agl 161 Highest expression of this gene was detected in normal ovarian tissue (CT=32). Expression of this gene was upregulated in normal ovarian and lung samples relative to corresponding cancer samples. Therapeutic modulation of the activity of this gene or its protein product using nucleic acid, protein, antibody or small molecule drugs is useful in the treatment of ovarian and lung cancers.
Low expression of this gene was also detected in a thyroid cancer sample. Therapeutic modulation of the activity of this gene or its protein product using nucleic acid, protein, antibody or small molecule drugs is useful in the treatment of thyroid cancer.
Panel 4D Summary: Agl 161 This gene was expressed at low levels in normal lung and colon (CTs=34). Expression of this gene was downregulated in the colon from a Crohn's disease patient was reduced. Therapeutic modulation of the activity of this gene or its protein product is useful in the treatment of inflammatory bowel diseases, such as Crohn's.
Low expression of this gene was also seen in liver cirrhosis sample. Therapeutic modulation of the activity of this gene or its protein product is useful in the treatment of liver cirrhosis.
I. CG55274-05: Diazepam-binding inhibitor
Expression of gene CG55274-05 was assessed using the primer-probe set Ag497, described in Table IA. Results of the RTQ-PCR runs are shown in Tables IB and IC.
Table IA. Probe Name Ag497
Figure imgf000358_0001
Table IB. Panel 1.1
Figure imgf000358_0002
Figure imgf000359_0001
Table IC. Panel 5 Islet
Column A - Rel. Exp.(%) Ag497, Run 323591176
Tissue Name A Tissue Name |A
97457_Patient-02go_adipose 0.0 94709_Donor 2 AM - A_adipose jo.o
97476_Patient-07sk_skeletal muscle 0.0 94710_Donor 2 AM - B_adipose |o.α
97477 Patient-07ut uterus jo.o 9471 l_Donor 2 AM - C_adipose jo.o
97478_Patient-07pl_placenta (0.0 94712_Donor 2 AD - A_adiρose fo.o
99167_Bayer Patient 1 jo.o 94713_Donor 2 AD - B_adipose jo.o
97482 Patient-08ut uterus 0.0 94714_Donor 2 AD - C_adipose jo.o
Figure imgf000360_0001
Panel 1.1 Summary: Ag497 Low expression of the CG55274-05 gene was restricted to the ovarian cancer cell line OVCAR-5 (CT=33.6). Gene or protein expression levels are useful as a marker to detect the presence of ovarian cancer. Therapeutic modulation of the activity of this gene or its protein product is useful in the treatment of ovarian cancer.
Panel 5 Islet Summary: Ag497 Low expression of this gene was mainly detected in a skeletal muscle sample from a diabetic patient on insulin (CT=33.6). The CG55274-05 gene encodes Diazepam-binding inhibitor, a member of the endozepine/acetyl CoA binding protein (ACBP)/diazepam binding inhibitor (DBI) family. ACBP is known to affect intracellular calcium levels via release from the sarcoplasmic reticulum in muscle, via the ryanodine receptor, and possibly the mitochondria (Fulceri R, Knudsen J, Giunti R,
Volpe P, Nori A, Benedetti A. Fatty acyl-CoA-acyl-CoA-binding protein complexes activate the Ca2+ release channel of skeletal muscle sarcoplasmic reticulum. Biochem J
1997 Jul 15;325 ( Pt 2):423-8; Fulceri R, Giunti R, Knudsen J, Leuzzi R, Kardon T, Benedetti A. Rapamycin inhibits activation of ryanodine receptors from skeletal muscle by the fatty acyl CoA-acyl CoA binding protein complex. Biochem Biophys Res Commun 1999 Oct 22;264(2):409-12). Since the activity of many metabolic enzymes is regulated by intracellular calcium, ACBP could play an important role in many aspects of energy metabolism. Furthermore, the peptides produced from ACBP act as hormones or paracrine factors to influence metabolism globally. One ACBP-derived peptide
(octadecaneuropeptide: ODN - ACBP33-50) exerts this action through several mechanisms. One mechanism influences nutrient absoφtion through the stimulation of CCK secretion and the subsequent secretion by the exocrine pancreas (Herzig KH; Schon I; Tatemoto K; Ohe Y; Li Y; Folsch UR; Owyang C. Diazepam binding inhibitor is a potent cholecystokmin-releasing peptide in the intestine. Proc. Nat. Acad. Sci. 1996; 93: 7927-7932). At the same time ODN inhibits glucose-stimulated insulin secretion from the endocrine pancreas [10]. In addition, intracerebroventricular administration of ODN has anorexigenic effects in rats (de Mateos-Verchere JG, Leprince J, Tonon MC, Vaudry H, Costentin J. The octadecaneuropeptide [diazepam-binding inhibitor (33-50)] exerts potent anorexigenic effects in rodents. Eur J Pharmacol 2001 Mar 2;414(2-3):225-31).
Full-length ACBP and peptides derived from the parent polypeptide participate in several different feedback loops influencing metabolism at many levels. Based upon the specific expression of this gene in skeletal muscle from diabetic patient and that ODN has broad-ranging effects on physiologic processes, ODN-related peptides from the CG55274-05 gene, encoding an ACBP-like protein, are potential protein therapeutics for the treatment of metabolic disorders such as obesity and diabetes.
J. CG55379-01 and CG55379-04: hNOPE
Expression of gene CG55379-01 and CG55379-04 was assessed using the primer-probe set Ag902, described in Table JA. Results of the RTQ-PCR runs are shown in Tables IB, JC, JD, JE, IF, JG and JH.
Table JA. Probe Name Ag902
Figure imgf000361_0001
Table JB. CNS neurodegeneration yl.O
Figure imgf000362_0001
Table JC. General screening panel yl.4
Figure imgf000362_0002
Figure imgf000363_0001
Table JD. HASS Panel vl.O
Figure imgf000364_0001
Figure imgf000365_0001
Table JE. Panel 2D
Figure imgf000365_0002
Figure imgf000366_0001
Table JF. Panel 3D
Figure imgf000366_0002
Figure imgf000367_0001
Table JG. Panel 4.1D
Figure imgf000368_0001
Figure imgf000369_0002
Table JH. Panel 5D
Figure imgf000369_0001
CNS_neurodegeneration_vl.0 Summary: Ag902 Expression of the CG55379-01 and CG55379-04 genes was upregulated in the temporal cortex of Alzheimer's disease patients compared to normal patients. Therefore, therapeutic modulation of the activity of these genes or their protein products using nucleic acid, protein, antibody and small molecule drugs is useful decreasing neuronal death that accompanies Alzheimer's disease.
General_screeningjpanel_vl.4 Summary: Ag902 Highest expression of these genes was detected in pancreatic cancer cell line CAPAN2 (CT=27). Moderate levels of expression of these genes were also seen in cluster of cell lines derived from gastric, colon, lung, renal, breast, ovarian, prostate, and brain cancers and melanomas. Gene or protein expression levels are useful as a marker to detect the presence of these cancers. Therapeutic modulation of the activity of these genes using nucleic acid, protein, antibody or small molecule drugs will be effective in the treatment of pancreatic, gastric, colon, lung, renal, breast, ovarian, prostate, melanoma and brain cancers.
Among tissues with metabolic or endocrine function, these genes were expressed at moderate levels in pancreas, adipose, adrenal gland, thyroid, pituitary gland, skeletal muscle, heart, fetal liver and the gastrointestinal tract. Therapeutic modulation of the activity of these genes or their protein products is useful in the treatment of endocrine/metabolically related diseases, such as obesity and diabetes.
In addition, these gene variants were expressed at moderate levels in all regions of the central nervous system examined, including amygdala, hippocampus, substantia nigra, thalamus, cerebellum, cerebral cortex, and spinal cord. The protein encoded by this gene is a homolog of mouse NOPE protein, which functions as a guidance receptor in the developing CNS (Salbaum JM, Kappen C, 2000, Cloning and expression of nope, a new mouse gene of the immunoglobulin superfamily related to guidance receptors. Genomics. 64(l):15-23, PMID: 10708514). Therapeutic modulation of the activity of these genes or their protein products is useful in the treatment of central nervous system disorders such as Alzheimer's disease, Parkinson's disease, epilepsy, multiple sclerosis, schizophrenia and depression. Expression of these genes was higher in fetal tissues relative to adult tissues, especially in fetal liver, lung, and brain. The relative overexpression of these genes in fetal tissue indicates that the encoded proteins may enhance liver, lung and brain growth or development in the fetus and thus may also act in a regenerative capacity in the adult. Therefore, therapeutic modulation of these genes and/or encoded proteins is useful in treatment of liver, lung and brain related diseases.
HASS Panel vl.O Summary: Ag902 Highest expression of the CG55379-01 and CG55379-04 genes was detected in glioma (CT=28). Moderate to low levels of expression of these variants were also seen in pancreatic cancer cell line CAPaN and glioblastoma/ astrocytoma cell lines. The expression of these genes was not altered by oxygen deprivation, acidic conditions or a serum-starved environment. Therapeutic modulation of the activity of these variants are useful in the treatment of pancreatic cancer, medulloblastoma and glioma.
Panel 2D Summary: Ag902 Highest expression of these genes was detected in a kidney cancer sample (CTs=30). The CG55379-01 and CG55379-04 genes were overexpressed in 7/9 kidney cancer and 2/4 colon cancer samples relative to the corresponding normal adjacent tissues. Gene or protein expression levels are useful in the diagnosis of kidney and colon cancer. Therapeutic modulation of the activity of these variants or their protein products using nucleic acid, protein, antibody and small molecule drugs are useful in the treatment of kidney cancer.
Panel 3D Summary: Ag902 Highest expression of the CG55379-01 and CG55379-04 genes was detected in a colon cancer cell line (CT=30). These variants were also expressed in cancer cell lines derived from kidney, lung, brain, pancreas and bone cancers. This observation indicates a possible role for this gene in the pathogenesis of these cancers. Please see panel 2D for further discussion of this gene.
Panel 4.1D Summary: Ag902 Highest expression of these genes was detected in activated dendritic cells (CT=30). The expression of these variants was also induced in LPS-stimulated dendritic cells and in IL-4-stimulated dermal fibroblasts. Low expression of this gene was also seen in astrocytes and normal thymus. The CG55379-01 and CG55379-04 genes encodes variants homologous to the mouse NOPE protein, a guidance receptors (Salbaum JM, Kappen C, 2000, Cloning and expression of nope, a new mouse gene of the immunoglobulin superfamily related to guidance receptors. Genomics. 64(l):15-23, PMID: 10708514). These proteins may act as a receptor for dendritic cells and dermal fibroblasts and may control interactions between these cells and other cell types during antigen presentation or apoptosis similar to netrins (Livesey F.J., 1999, - Netrins and netrin receptors. Cell Mol. Life Sci. 56: 62-68, PMID: 11213262). Therapeutic modulation of the activity of these variants or their protein products are useful in blocking inflammation in diseases such as asthma, arthritis, psoriasis, allergy and other diseases in which dendritic cell or dermal fibroblasts play important roles.
Panel 5D Summary: Ag902 Highest expression of these genes was seen in placenta of a diabetic patient on insulin (CT=32.3). Significant expression of these genes were also seen in placenta from diabetic and non-diabetic patients. Please see panel 1.4 for further discussion of this gene.
K. CG55688-01: Cyr61 Expression of gene CG55688-01 was assessed using the primer-probe set Agl 148, described in Table KA. Results of the RTQ-PCR runs are shown in Tables KB, KC, KD, KE, KF andKG.
Table KA. Probe Name Agl 148
Figure imgf000372_0001
Table KB. HASS Panel vl.O
Figure imgf000372_0002
Figure imgf000373_0001
371 Table KC.PGI1.0
Figure imgf000374_0001
Table KD. Panel 1.3D
Figure imgf000374_0002
Figure imgf000375_0001
Table KE. Panel 2D
Figure imgf000376_0001
Figure imgf000377_0001
Table KF. Panel 3D
Figure imgf000377_0002
Figure imgf000378_0001
Table KG. Panel 4D
Figure imgf000378_0002
Figure imgf000379_0001
HASS Panel vl.O Summary: Agl 148 Expression of the CG55688-01 gene was highest in T24 cells (CT=27.9). This gene was also expressed at significant level in CaPaN and U87 cancer cell lines, as well as in primary astrocytes, renal epithelial cells and melanocytes in culture. Gene expression was induced by a combination of low oxygen tension and acidic pH in U8 cell lines, suggesting a regulation in vivo may also occur in regions of low pH and low oxygen. Therapeutic modulation of the activity of this gene or its protein product is useful in the treatment of cancer.
PGI1.0 Summary: Agl 148 Highest expression of this gene was detected in emphysema lung (CT=25.2). High expression of this gene was also detected in lung fibrosis, asthma, emphysema and ulcerative colitis samples. Therapeutic modulation of the activity of this gene or its protein product using nucleic acid, protein, antibody or small molecule drugs is useful in the treatment of lung fibrosis, asthma, emphysema and ulcerative colitis.
Panel 1.3D Summary: Agl 148 The expression of this gene was highest in a sample derived from a melanoma cell line (Hs.688(A).T) (CT=27). In addition, there is significant expression in a related melanoma cell line (Hs.688(B).T) as well as a cluster of brain cancer cell lines and renal cancer cell lines. T Therapeutic modulation of the activity of this gene or its protein product is useful in the treatment of melanoma, renal cancer or brain cancer.
This panel shows significant expression of this gene in metabolic tissues, including adipose, pancreas, adrenal, thyroid, pituitary, skeletal muscle and adult and fetal liver. The CG55688-01 gene encodes CYR61, which belongs to the insulin-like growth factor binding protein family and may play myriad roles in metabolic regulation. Therapeutic modulation of the activity of this gene or its protein product using nucleic acid, protein, antibody or small molecule drugs is useful for the treatment of metabolic and endocrine diseases, including obesity and Types 1 and 2 diabetes.
In addition, this gene was expressed at low levels in several brain regions including hippocampus, cortex, substantia nigra, thalamus, amygdala, and the fetal brain. Cry61 is an immediate early gene that has been implicated in memory formation and synaptic plasticity (Albrecht C, von Der Kammer H, Mayhaus M, Klaudiny J, Schweizer M, Nitsch RM. Muscarinic acetylcholine receptors induce the expression of the immediate early growth regulatory gene CYR61. J Biol Chem 2000 Sep 15;275(37):28929-36). It has also been shown to be upregulated during the development of the hippocampus, which is a critical brain region for the formation of long-term memory (Chung KC, Ahn YS. Expression of immediate early gene cyrόl during the differentiation of immortalized embryonic hippocampal neuronal cells. Neurosci Lett 1998 Oct 23 ;255(3): 155-8).
Therefore, this gene, expressed protein, antibodies or small molecule drug targeting this gene or gene product is useful in the treatment of dementia (Alzheimer's, vascular, etc) or for memory enhancement.
Panel 2D Summary: Agl 148 Highest expression of this gene was found in normal bladder tissue and a kidney cancer sample (CTs=28). In addition, there was significant expression of this gene associated with ovarian and prostate derived tissues and a number of kidney samples. Therapeutic modulation of the activity of this gene or its protein product is useful in the treatment of kidney cancer, ovarian cancer or prostate cancer.
Panel 3D Summary: Agl 148 Highest expression of this gene was detected in lung cancer cell line NCI-H292 (CT=28). Significant expression of this gene was also seen in a number of cancer cell lines derived from brain, renal, pancreatic, bladder cancers and sarcomas. Therapeutic modulation of the activity of this gene or its protein product is useful in the treatment of these cancers.
Panel 4D Summary: Agl 148 This gene, a Cyr61 homolog, was expressed at moderate levels (CTs=28-32) in resting and cytokine-stimulated HUVEC, lung microvascular endothelial cells, coronary artery smooth muscle cells, bronchial epithelial cells, small airway epithelial cells, astrocytes, pulmonary artery endothelial cells, lung fibroblasts, and dermal fibroblasts. Based upon this expression pattern and a role for Cyrόl in vascular biology (Babic AM, Kireeva ML, Kolesnikova TV, Lau LF CYR61, a product of a growth factor-inducible immediate early gene, promotes angiogenesis and tumor growth. Proc Natl Acad Sci U S A 1998 May 26;95(11):6355-60), therapeutic modulation of the acitivity of this gene or its protein product is useful in the treatment of inflammatory or autoimmune diseases, including Crohn's disease, ulcerative colitis, multiple sclerosis, chronic obstructive pulmonary disease, asthma, emphysema, rheumatoid arthritis, or psoriasis. L. CG56768-01: Wnt-5A
Expression of gene CG56768-01 was assessed using the primer-probe set Agl450, described in Table LA. Results of the RTQ-PCR runs are shown in Tables LB, LC, LD and LE.
Table LA. Probe Name Agl450
Figure imgf000382_0002
Table LB. Ardais Panel 1.1
Figure imgf000382_0001
Table LC. Panel 1.2
Figure imgf000382_0003
Figure imgf000383_0001
Table LD. Panel 2D
Figure imgf000384_0001
Figure imgf000385_0001
Table LE. Panel 4.1D
Figure imgf000385_0002
Figure imgf000386_0001
Ardais Panel 1.1 Summary: Agl450 Highest expression of the CG56768-01 gene was seen in a lung cancer sample (CT=21). Expression of this gene was higher in this cancer sample relative to the normal adjacent tissue sample (CT=29). Gene or protein expression levels are useful for the detection of lung cancer. Therapeutic modulation of the activity of this gene or its protein product using nucleic acid, protein, antibody or small molecule drags is useful in the treatment of lung cancer.
Panel 1.2 Summary: Agl450 Highest expression of this gene was detected in glioma cell line SF-295 (CT=22). This gene was overexpressed in cell lines derived from CNS malignancies when compared to the low to moderate expression in the samples derived from normal CNS tissue. In addition, there was consistently high expression of this gene in melanoma cell lines, ovarian cancer cell lines and lung cancer cell lines. The CG56768-01 gene encodes a putative Wnt5a-like protein. The Wnt genes belong to a family of protooncogenes with at least 13 known members that are expressed in species ranging from Drosophila to man. The name Wnt denotes the relationship of this family to the Drosophila segment polarity gene 'wingless' and to its vertebrate ortholog, Intl, a mouse protooncogene (OMIM 164975, 164820). Therapeutic modulation of the activity of this gene or its protein product using nucleic acid, protein, antibody or small molecule drugs is useful for the treatment of brain cancer, melanoma, ovarian cancer and/or lung cancer.
Significant levels of expression of this gene were detected in all the regions of the brain examined including amygdala, hippocampus, substantia nigra, thalamus, cerebellum, cerebral cortex, and spinal cord. Wnt-5A signalling is believed to play a critical role in cadherin-mediated cell organization. Cadherins can act as axon guidance and cell adhesion proteins, specifically during development and in the response to injury. Therapeutic modulation of the activity of this gene or its protein product is useful in inducing a compensatory synaptogenic response to neuronal death in Alzheimer's disease, Parkinson's disease, Huntington's disease, spinocerebellar ataxia, progressive supranuclear palsy, ALS, head trauma, stroke, or any other disease/condition associated with neuronal loss.
Among tissues with metabolic or endocrine function, this gene was expressed at moderate levels in pancreas, adrenal gland, thyroid, pituitary and liver. In addition, this gene was expressed at high levels in skeletal muscle (CT = 27). These observations suggest that the Wnt-5A-like protein encoded by this gene may be secreted from skeletal muscle as a paracrine or endocrine signalling molecule acting on other insulin-responsive tissues (i.e., adipose and pancreatic beta cells). Therapeutic modulation of the activity of this gene or its protein product using nucleic acid, protein, antibody or small molecule drags is useful in the treatment of metabolic diseases involving skeletal muscle, including Type 2 diabetes.
Panel 2D Summary: Agl 450 Highest expression of this gene was detected in a lung cancer sample (CTs=28-29). This gene was overexpressed in number of cancer tissues relative to the adjacent normal colon, lung, kidney, breast, and stomach. These results are consistent with the observation that the Wnt-5A gene appears to be up-regulated in a number of human malignancies (Iozzo RN., Eichstetter I., Danielson K.G., 1995,
Aberrant expression of the growth factor Wnt-5A in human malignancy. Cancer Res. 55: 3495-3499). Therapeutic modulation of the activity of this gene or its protein product is of use in the treatment of colon, lung, kidney, breast or gastric cancers.
Panel 4.1D Summary: Agl450 Highest expression of this gene was detected in IL-1 beta activated dermal fibroblasts (CT=26). This gene was expressed mainly in fibroblasts and in LPS-activated monocytes, macrophages and dendritic cells. WNTs are secreted signalling molecules that regulate cell fate and behavior and are involved in embryonic development and hematopoiesis. During inflammation, the Wnt5a-like protein encoded by this gene could potentiate the inflammatory response by acting as an autocrine factor and stimulating monocyte differentiation into dendritic cells as well as by allowing dendritic cells to mature into potent antigen presenting cells. Alternatively, this gene may influence the differentiation of other cell types in the microenvironment including synovial tissues (Sen M., Lauterbach K., El-Gabalawy H., Firestein G.S., Corr M., Carson D.A., 2000, Expression and function of wingless and frizzled homologs in rheumatoid arthritis. Proc. Natl. Acad. Sci. USA 97: 2791-2796). Therapeutic modulation of the activity of this gene or its protein product using nucleic acid, protein, antibody or small molecule drags is important in reducing or blocking inflammation associated with rheumatoid arthritis, asthma, allergy, psoriasis, IBD and Crohn's disease.
M. CG59253-01 and CG59253-02: Semaphorin precursor
Expression of gene CG59253-01 and CG59253-02 was assessed using the primer-probe sets Agl 492 and Ag2441 , described in Tables MA and MB. Results of the RTQ-PCR runs are shown in Tables MC, MD, ME and MF. CG59253-01 represents a full-length physical clone.
Table MA. Probe Name Agl492
Figure imgf000388_0001
Table MB. Probe Name Ag2441
Figure imgf000389_0001
Table MC. AI comprehensive panel yl.O
Figure imgf000389_0002
Figure imgf000390_0002
Table MD. Panel 1.3D
Figure imgf000390_0001
Figure imgf000391_0001
Table ME. Panel 2D
Figure imgf000391_0002
Figure imgf000392_0001
Table MF. Panel 4D
Figure imgf000393_0001
Figure imgf000394_0001
AI_comprehensive panel vl.O Summary: Agl492 Highest expression of the CG59253-01 and CG59253-02 genes was detected in normal lung (CT=27.6). Significant expression of these genes was detected in samples derived from normal and orthoarthitis/ rheumatoid arthritis bone, cartilage, synovium and synovial fluid samples, normal lung, COPD lung, emphysema, atopic asthma, asthma, allergy, Crohn's disease (normal matched control and diseased), ulcerative colitis (normal matched control and diseased), and psoriasis (normal matched control and diseased). Therapeutic modulation of the activity of these gene variants or their protein products is useful in the treatment of autoimmune and inflammatory disorders including psoriasis, allergy, asthma, inflammatory bowel disease, rheumatoid arthritis and osteoarthritis
Panel 1.3D Summary: Agl492/Ag2441 The CG59253-01 and CG59253-02 genes encode a semaphorin homolog that had brain-preferential expression. Highest expression of these variants was seen in the brain and a brain cancer cell line (CTs=28-29). Semaphorins can act as axon guidance proteins, specifically as chemorepellents that inhibit CNS regenerative capacity. Manipulation of levels of these gene variants or their protein products is useful in inducing a compensatory synaptogenic response to neuronal death in Alzheimer's disease, Parkinson's disease, Huntington's disease, spinocerebellar ataxia, progressive supranuclear palsy, multiple sclerosis, ALS, head trauma, stroke, or any other disease/condition associated with neuronal loss. Therapeutic modulation of the activity of these gene variants or their protein products using nucleic acid, protein, antibody or small molecule drugs is useful in the treatment of brain cancer.
This gene was also moderately expressed in several metabolic tissues, including pancreas, adrenal, thyroid, pituitary, adult and fetal heart, adult and fetal skeletal muscle, adult and fetal liver, and adipose. Gene or protein expression levels are important for the pathogenesis, diagnosis, and/or treatment of metabolic diseases including obesity and Types 1 and 2 diabetes.
Panel 2D Summary: Ag2441 Highest expression of the CG59253-01 and CG59253-02 gene variants was detected in normal kidney (CT=27.6). These variants were more highly expressed in normal kidney samples relative to the matched kidney cancers. Gene or protein levels are useful to distinguish normal kidney from kidney cancer. These genes encode variants of the semaphorin SEMA6D protein. The semaphorin family of proteins is characterized as cell surface receptors for their ligands, the pillins, and is involved largely in cell guidance (Tamagnone L, Comoglio PM. Signaling by semaphorin receptors: cell guidance and beyond. Trends Cell Biol 2000 Sep;10(9):377-83). Semaphorins have been implicated in general invasive growth and potentially even tube formation (Comoglio PM, Tamagnone L, Boccaccio C. Plasminogen-related growth factor and semaphorin receptors: a gene superfamily controlling invasive growth. Exp Cell Res 1999 Nov 25;253(l):88-99). Thus, semaphorins are likely agents to promote the differentiation of cells. Normal kidney cells undergo a great deal of tubular morphogenesis. Therefore, the extracellular domain of these protein products may act to promote growth arrest and differentiation of the cancer cells through interaction with a membrane bound ligand or ligand complexed with plexins. Therapeutic modulation of the activity of these gene variants or their protein products using nucleic acid, protein, antibody or small molecule drugs is of use in the treatment of kidney cancer.
Panel 4D Summary: Agl492/2441 Highest expression of the CG59253-01 and CG59253-02 gene variants was detected in thymus (CTs=27-28). Significant expression of these variants was also seen activated lung fibroblasts cells, HUVEC, HPAEC, activated bronchial epithelium, NCI-H292 cell, dermal fibroblasts, IBD Crohn's, liver cirrhosis and lupus samples, normal tissues colon and thymus. Therapeutic modulation of the activity of these gene variants or their protein products is useful to reduce or eliminate the symptoms of chronic obstructive pulmonary disease, asthma, emphysema, and ulcerative colitis
N. CG95430-01: AdipoQ-like
Expression of gene CG95430-01 was assessed using the primer-probe set Ag4020, described in Table NA. Results of the RTQ-PCR runs are shown in Tables NB, NC, ND, NE andNF.
Table NA. Probe Name Ag4020
Figure imgf000396_0001
Table NB. CNS neurodegeneration yl.O
Figure imgf000396_0002
Figure imgf000397_0001
Table NC. Oncology cell line screening panel v3.1
Figure imgf000397_0002
Figure imgf000398_0001
Table ND. Panel 4.1D
Figure imgf000398_0002
Figure imgf000399_0001
Table NE. Panel 5 Islet
Figure imgf000399_0002
Figure imgf000400_0002
Table NF. general oncology screening panel v 2.4
Figure imgf000400_0001
Figure imgf000401_0001
CNS_neurodegeneration_vl.0 Summary: Ag4020 The CG95430-01 gene was not differentially expressed in the Alzheimer's disease samples represented on this panel. However, this gene was expressed in the brain, with highest expression in the hippocampus of an Alzheimer's patient (CT=31.4). Therapeutic modulation of the activity of this gene or its protein product is useful in the treatment of neurological disorders, such as Alzheimer's disease, Parkinson's disease, schizophrenia, multiple sclerosis, stroke and epilepsy.
Oncology_cell_line_screening_panel_v3.1 Summary: Ag4020 Highest expression of the CG95430-01 gene was detected in an epidermoid carcinoma cell line (CT=32.6). Low expression of this gene was also seen in cell lines derived from fibrosarcoma, pancreatic ductal adenocarcinoma, pancreatic, colon and gastric cancers. Therapeutic modulation of the activity of this gene or its protein product is useful in the treatment of these cancers.
Panel 4.1D Summary: Ag4020 This gene was most highly expressed in a normal kidney sample (CT=32.3). Low but significant levels of expression were also seen in untreated and cytokine activated lung fibroblasts and thymus. These results suggest that this gene is involved in the homeostasis of the lung, thymus, and kidney. Down-regulated expression of this gene in cytokine-activated lung fibroblasts indicates that modulation of this gene and its protein product will help to maintain or restore function to the lung during inflammation.
Panel 5 Islet Summary: Ag4020 The CG95430-01 gene was expressed in adipose and skeletal muscle (CTs=31-34). This gene encodes a putative adiponectin [also known as adipocyte complement-related protein (ACRP-30), AdipoQ, apMl (adipose most abundant transcript 1) or GBP28 (28 kDa gelatin binding protein)], a member of the Clq family. This protein is induced over 100-fold in adipocyte differentiation (Scherer et al., J Biol Chem 1995 Nov 10;270(45):26746-9) and is involved in adipocyte signaling (Hu et al., J Biol Chem 1996 May 3;271(18):10697-703). Like other members of the Clq family, it forms a homotrimer and the crystal structure indicates that it likely arose from tumor necrosis factor (TNF; Shapiro and Scherer,Curr Biol 1998 Mar 12;8(6):335-8). Ionomycin increases expression of adiponectin and dibutyryl cAMP and TNF-alpha reduce expression and secretion in 3T3-L1 adipocytes (Kappes and Loftier, Horm Metab Res 2000 Nov-Dec;32(l l-12):548-54). Levels of adiponectin are decreased in obese humans (Arita et al., Biochem Biophys Res Commun 1999 Apr 2;257(l):79-83) and mice (Hu et al., J Biol Chem 1996 May 3;271(18):10697-703). A proteolytic cleavage product of adiponectin is reported to increase fatty acid oxidation in muscle and causes weight loss in mice. (Fruebis et al., Proc Natl Acad Sci U S A 2001 Feb 13;98(4):2005-10). A missense mutation in the protein was correlated with a markedly low plasma adiponectin level (Takahashi et al., Int J Obes Relat Metab Disord 2000 Jul;24(7):861-8). Recent papers have shown that adiponectin reverses insulin resistance in mouse models of lipoatrophy and obesity (Yamauchi et al., Nature Med 2000; 7(8): 941-6), and that it enhances insulin action on the liver (Berg et al., ibid, 947-53). In addition, circulating levels of adiponectin have been shown to be lower in obese than in lean subjects and lower in diabetic patients than in non-diabetic patients, with particularly low levels in subjects with coronary artery disease. Furthermore, in patients who were subjected to a weight loss program that resulted in a 10% reduction of their body mass index, circulating adiponectin levels increased significantly. (Berg AH. Trends Endocrinol Metab. 2002 Mar; 13(2): 84-9). Based on the homology of CG95430-01 to adiponectin and its expression profile, therapeutic modulation of the activity of this gene or its protein product using nucleic acid, protein, antibody or small molecule drugs is useful for the treatment of obesity, type II diabetes and/or their secondary complications.
Adiponectin also seems to have additional cardiovascular and immune system effects. Levels of this protein are reduced in a cohort of Japanese patients with coronary artery disease (CAD), which correlates with the modulation of endothelial adhesion molecules on treatment of human aortic endothelial cells with adiponectin (Ouchi et al., Circulation 1999 Dec 21-28;100(25):2473-6). This protein is found adhering to vascular walls after injury (Okamoto et al. Horm Metab Res 2000 Feb;32(2):47-50) and presence of adiponectin suppresses the macrophage to foam cell transformation (Ouchi et al., Circulation 2001 Feb 27; 103 (8): 1057-63). In addition, levels of adiponectin are lower in diabetic subjects with CAD relative to non-diabetic subjects or diabetic subjects without CAD (Hotta et al., Arterioscler Thromb Vase Biol 2000 Jun;20(6): 1595-9), indicating that lower levels of adiponectin may be an indicator of macroangiopathy in diabetes. Moreover, this protein negatively regulates the growth of myelomonocytic precursors (in part by inducing apoptosis) and macrophage function (Yokota et al., Blood 2000 Sep 1;96(5): 1723-32), potentially via the complement 1Q receptor ClqRp.
The Clq family of proteins includes the complement subunit Clq, gliacolin, Clq-related protein, cerebellin, CORS26 etc., all of which are secreted proteins. These proteins share a common domain, the Clq domain, at the C terminus and collagen triple helix repeats at the C terminus. The repeats enable the proteins to form homotrimers and possibly oligomers. Members of this family have been implicated in tissue differentiation, immune regulation, energy homeostasis, synaptic function and in diseases such as obesity and neurodegeneration. Therapeutic modulation of the activity of this gene or its protein product using nucleic acid, protein, antibody or small molecule drugs is useful in the prevention and/or treatment of obesity and diabetes. Furthermore, development of human monoclonal antibodies that inhibit this Adipo-Q like protein is useful in the therapeutic treatment of cachexia that occurs in many forms of cancer.
General oncology screening panel_v_2.4 Summary: Ag4020 This gene was most highly expressed in a metastatic melanoma (CT=32.7). Significant levels of expression were also seen in a lung cancer and a kidney cancer when compared to normal adjacent tissue. Gene or protein expression levels are useful as a diagnostic marker to detect the presence of these cancers. Therapeutic modulation of the activity of this gene or its protein product is useful in the treatment of kidney cancer, lung cancer, and melanoma.
O. CG95430-02 and CG95430-04
Expression of genes CG95430-02 and CG95430-04 was assessed using the primer-probe set Ag7140, described in Table OA. Results of the RTQ-PCR runs are shown in Table OB. CG95430-02 and CG95430-04 represent the physical clones for mature and full-length gene respectively. Table OA. Probe Name Ag7140
Figure imgf000404_0001
Table OB. General screening panel yl.7
Figure imgf000404_0002
Figure imgf000405_0001
General_screening_panel_vl.7 Summary: Ag7140 Highest expression of the CG95430-02 and CG95430-04 gene variants was detected in adipose tissue (CT=28.9). Moderate to low expression of these variants was also seen in number of tissues that contribute to metabolism including thyroid, skeletal muscle, heart, small intestine, and colon. Therapeutic modulation of the activity of these gene variants or their protein products using nucleic acid, protein, antibody or small molecule drugs is useful in the treatment of endocrine/metabolically related diseases, such as obesity and diabetes.
P. CG97111-01: Interleukin-1 receptor antagonist protein precursor Expression of gene CG97111-01 was assessed using the primer-probe sets Ag4106 described in Tables PA. Results of the RTQ-PCR runs are shown in Tables PB and PC.
Table PA. Probe Name Ag4106
Figure imgf000405_0002
Table PB. General screening panel yl.4
Figure imgf000406_0001
Figure imgf000407_0001
Table PC. Panel 4.1D
Figure imgf000407_0002
Figure imgf000408_0001
General_screening_panel vl.4 Summary: Ag4106 Significant expression of the CG97111-01 gene was seen mainly in thymus (CT=33.4). This gene may therefore play an important role in T cell development. Gene or protein expression levels are useful for the detection of thymus. Therapeutic modulation of the activity of this gene or its protein product is useful to modulate immune function (T cell development) and be important for organ transplant, AIDS treatment or post chemotherapy immune reconstitiution.
Panel 4.1D Summary: Ag4106 Significant expression of this gene was seen in a normal kidney sample (CT=33.4). Therapeutic modulation of the activity of this gene or its protein product is useful to modulate kidney function and for the treatment of inflammatory or autoimmune diseases that affect the kidney, including lupus and glomerulonephritis.
OTHER EMBODIMENTS Although particular embodiments have been disclosed herein in detail, this has been done by way of example for purposes of illustration only, and is not intended to be limiting with respect to the scope of the appended claims, which follow. In particular, it is contemplated by the inventors that various substitutions, alterations, and modifications may be made to the invention without departing from the spirit and scope of the invention as defined by the claims. The choice of nucleic acid starting material, clone of interest, or library type is believed to be a matter of routine for a person of ordinary skill in the art with knowledge of the embodiments described herein. Other aspects, advantages, and modifications considered to be within the scope of the following claims. The claims presented are representative of the inventions disclosed herein. Other, unclaimed inventions are also contemplated. Applicants reserve the right to pursue such inventions in later claims.

Claims

CLAIMSWhat is claimed is:
1. An isolated polypeptide comprising the mature form of an amino acid sequenced selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141.
2. An isolated polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141.
3. An isolated polypeptide comprising an amino acid sequence which is at least 95% identical to an amino acid sequence selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141.
4. An isolated polypeptide, wherein the polypeptide comprises an amino acid sequence comprising one or more conservative substitutions in the amino acid sequence selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141.
5. The polypeptide of claim 1 wherein said polypeptide is naturally occurring.
6. A composition comprising the polypeptide of claim 1 and a carrier.
7. A kit comprising, in one or more containers, the composition of claim 6.
8. The use of a therapeutic in the manufacture of a medicament for treating a syndrome associated with a human disease, the disease selected from a pathology associated with the polypeptide of claim 1, wherein the therapeutic comprises the polypeptide of claim 1.
9. A method for determining the presence or amount of the polypeptide of claim 1 in a sample, the method comprising:
(a) providing said sample;
(b) introducing said sample to an antibody that binds immunospecifically to the polypeptide; and
(c) determining the presence or amount of antibody bound to said polypeptide, thereby determining the presence or amount of polypeptide in said sample.
10. A method for determining the presence of or predisposition to a disease associated with altered levels of expression of the polypeptide of claim 1 in a first mammalian subject, the method comprising: a) measuring the level of expression of the polypeptide in a sample from the first mammalian subject; and b) comparing the expression of said polypeptide in the sample of step (a) to the expression of the polypeptide present in a control sample from a second mammalian subject known not to have, or not to be predisposed to, said disease, wherein an alteration in the level of expression of the polypeptide in the first subject as compared to the control sample indicates the presence of or predisposition to said disease.
11. A method of identifying an agent that binds to the polypeptide of claim 1 , the method comprising:
(a) introducing said polypeptide to said agent; and
(b) determining whether said agent binds to said polypeptide.
12. The method of claim 11 wherein the agent is a cellular receptor or a downstream effector.
13. A method for identifying a potential therapeutic agent for use in treatment of a pathology, wherein the pathology is related to aberrant expression or aberrant physiological interactions of the polypeptide of claim 1, the method comprising:
(a) providing a cell expressing the polypeptide of claim 1 and having a property or function ascribable to the polypeptide;
(b) contacting the cell with a composition comprising a candidate substance; and
(c) determining whether the substance alters the property or function ascribable to the polypeptide; whereby, if an alteration observed in the presence of the substance is not observed when the cell is contacted with a composition in the absence of the substance, the substance is identified as a potential therapeutic agent.
14. A method for screening for a modulator of activity of or of latency or predisposition to a pathology associated with the polypeptide of claim 1, said method comprising:
(a) administering a test compound to a test animal at increased risk for a pathology associated with the polypeptide of claim 1, wherein said test animal recombinantly expresses the polypeptide of claim 1;
(b) measuring the activity of said polypeptide in said test animal after administering the compound of step (a); and
(c) comparing the activity of said polypeptide in said test animal with the activity of said polypeptide in a control animal not administered said polypeptide, wherein a change in the activity of said polypeptide in said test animal relative to said control animal indicates the test compound is a modulator activity of or latency or predisposition to, a pathology associated with the polypeptide of claim 1.
15. The method of claim 14, wherein said test animal is a recombinant test animal that expresses a test protein transgene or expresses said transgene under the control of a promoter at an increased level relative to a wild-type test animal, and wherein said promoter is not the native gene promoter of said transgene.
16. A method for modulating the activity of the polypeptide of claim 1 , the method comprising contacting a cell sample expressing the polypeptide of claim 1 with a compound that binds to said polypeptide in an amount sufficient to modulate the activity of the polypeptide.
17. A method of treating or preventing a pathology associated with the polypeptide of claim 1, the method comprising administering the polypeptide of claim 1 to a subject in which such treatment or prevention is desired in an amount sufficient to treat or prevent the pathology in the subject.
18. The method of claim 17, wherein the subject is a human.
19. A method of treating a pathological state in a mammal, the method comprising administering to the mammal a polypeptide in an amount that is sufficient to alleviate the pathological state, wherein the polypeptide is a polypeptide having an amino acid sequence at least 95% identical to a polypeptide comprising the amino acid sequence selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141, or a biologically active fragment thereof.
20. An isolated nucleic acid molecule comprising a nucleic acid sequence selected from the group consisting of SEQ ID NO:2n-l, wherein n is an integer between 1 and 141.
21. The nucleic acid molecule of claim 20, wherein the nucleic acid molecule is naturally occurring.
22. A nucleic acid molecule, wherein the nucleic acid molecule differs by a single nucleotide from a nucleic acid sequence selected from the group consisting of SEQ ID NO: 2n-l, wherein n is an integer between 1 and 141.
23. An isolated nucleic acid molecule encoding the mature form of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 141.
24. A composition comprising an isolated nucleic acid molecule, said molecule comprising a nucleic acid sequence selected from the group consisting of SEQ ID NO: 2n-l , wherein n is an integer between 1 and 141 , and a carrier.
25. The nucleic acid molecule of claim 20, wherein said nucleic acid molecule hybridizes under stringent conditions to the nucleotide sequence selected from the group consisting of SEQ BO NO: 2n-l, wherein n is an integer between 1 and 141, or a complement of said nucleotide sequence.
26. A vector comprising the nucleic acid molecule of claim 20.
27. The vector of claim 26, further comprising a promoter operably linked to said nucleic acid molecule.
28. A cell comprising the vector of claim 26.
29. An antibody that immunospecifically binds to the polypeptide of claim 1.
30. The antibody of claim 29, wherein the antibody is a monoclonal antibody.
31. The antibody of claim 29, wherein the antibody is a humanized antibody.
32. A method for determining the presence or amount of the nucleic acid molecule of claim 20 in a sample, the method comprising:
(a) providing said sample;
(b) introducing said sample to a probe that binds to said nucleic acid molecule; and
(c) determining the presence or amount of said probe bound to said nucleic acid molecule, thereby determining the presence or amount of the nucleic acid molecule in said sample.
33. The method of claim 32 wherein presence or amount of the nucleic acid molecule is used as a marker for cell or tissue type.
34. The method of claim 33 wherein the cell or tissue type is cancerous.
35. A method for determining the presence of or predisposition to a disease associated with altered levels of expression of the nucleic acid molecule of claim 20 in a first mammalian subject, the method comprising: a) measuring the level of expression of the nucleic acid in a sample from the first mammalian subject; and b) comparing the level of expression of said nucleic acid in the sample of step (a) to the level of expression of the nucleic acid present in a control sample from a second mammalian subject known not to have or not be predisposed to, the disease; wherein an alteration in the level of expression of the nucleic acid in the first subject as compared to the control sample indicates the presence of or predisposition to the disease.
36. A method of producing the polypeptide of claim 1 , the method comprising culturing a cell under conditions that lead to expression of the polypeptide, wherein said cell comprises a vector comprising an isolated nucleic acid molecule comprising a nucleic acid sequence selected from the group consisting of SEQ ID NO:2n-l, wherein n is an integer between 1 and 141.
37. The method of claim 36 wherein the cell is a bacterial cell.
38. The method of claim 36 wherein the cell is an insect cell.
39. The method of claim 36 wherein the cell is a yeast cell.
40. The method of claim 36 wherein the cell is a mammalian cell.
41. A method of producing the polypeptide of claim 2, the method comprising culturing a cell under conditions that lead to expression of the polypeptide, wherein said cell comprises a vector comprising an isolated nucleic acid molecule comprising a nucleic acid sequence selected from the group consisting of SEQ ID NO:2n-l , wherein n is an integer between 1 and 141.
42. The method of claim 41 wherein the cell is a bacterial cell.
43. The method of claim 41 wherein the cell is an insect cell.
44. The method of claim 41 wherein the cell is a yeast cell.
45. The method of claim 41 wherein the cell is a mammalian cell.
46. An isolated polypeptide comprising an amino acid sequence at least 95% similar to SEQ ID NO: 2, wherein said amino acid sequence comprises at least one amino acid substitution, wherein said substitution is at amino acid position 43 when numbered in accordance with SEQ ID NO: 2.
47. An isolated nucleic acid molecule comprising an nucleic acid sequence at least 95% similar to SEQ ID NO: 1, wherein said nucleic acid sequence comprises at least one nucleic acid substitution, wherein said substitution is at nucleic acid position 135 when numbered in accordance with SEQ ID NO: 1.
48. An isolated polypeptide comprising an amino acid sequence at least 95% similar to SEQ ID NO: 14, wherein said amino acid sequence comprises at least one amino acid substitution, wherein said substitution is at the amino acid position selected from the group consisting of 8, 54, 56, 92, 207, 240, 706, 891 and 923 when numbered in accordance with SEQ ID NO: 14.
49. An isolated nucleic acid molecule comprising an nucleic acid sequence at least 95% similar to SEQ ID NO: 13, wherein said nucleic acid sequence comprises at least one nucleic acid substitution, wherein said substitution is at the nucleic acid position selected from the group consisting of 272, 410, 416, 523, 869, 967, 2366, 2921 and 3018 when numbered in accordance with SEQ ID NO: 13.
50. An isolated polypeptide comprising an amino acid sequence at least 95% similar to SEQ JD NO: 58, wherein said amino acid sequence comprises at least one amino acid substitution, wherein said substitution is at the amino acid position selected from the group consisting of 23, 56, 105, 125, 160, 183 and 215 when numbered in accordance with SEQ ID NO: 58.
51. An isolated nucleic acid molecule comprising an nucleic acid sequence at least 95% similar to SEQ ID NO: 57, wherein said nucleic acid sequence comprises at least one nucleic acid substitution, wherein said substitution is at the nucleic acid position selected from the group consisting of 181, 278, 426, 485, 591, 661 and 756 when numbered in accordance with SEQ ID NO: 57.
52. An isolated polypeptide comprising an amino acid sequence at least 95% similar to SEQ ID NO: 80, wherein said amino acid sequence comprises at least one amino acid substitution, wherein said substitution is at amino acid position 219 when numbered in accordance with SEQ ID NO: 80.
53. An isolated nucleic acid molecule comprising an nucleic acid sequence at least 95% similar to SEQ ID NO: 79, wherein said nucleic acid sequence comprises at least one nucleic acid substitution, wherein said substitution is at nucleic acid position 685 when numbered in accordance with SEQ ID NO: 79.
54. An isolated polypeptide comprising an amino acid sequence at least 95% similar to SEQ ID NO: 92, wherein said amino acid sequence comprises at least one amino acid substitution, wherein said substitution is at amino acid position 470 when numbered in accordance with SEQ ID NO: 92.
55. An isolated nucleic acid molecule comprising an nucleic acid sequence at least 95% similar to SEQ ID NO: 91, wherein said nucleic acid sequence comprises at least one nucleic acid substitution, wherein said substitution is at nucleic acid position 1874 when numbered in accordance with SEQ ID NO: 91.
56. An isolated polypeptide comprising an amino acid sequence at least 95% similar to SEQ ID NO: 100, wherein said amino acid sequence comprises at least one amino acid substitution, wherein said substitution is at the amino acid position selected from the group consisting of 11, 112 and 145 when numbered in accordance with SEQ ID NO: 100.
57. An isolated nucleic acid molecule comprising an nucleic acid sequence at least 95% similar to SEQ ID NO: 99, wherein said nucleic acid sequence comprises at least one nucleic acid substitution, wherein said substitution is at the nucleic acid position selected from the group consisting of 80, 383 and 482 when numbered in accordance with SEQ ID NO: 99.
58. An isolated polypeptide comprising an amino acid sequence at least 95% similar to SEQ ID NO: 122, wherein said amino acid sequence comprises at least one amino acid substitution, wherein said substitution is at the amino acid position selected from the group consisting of 12, 38, 54, 65, 66, 69, 80, 90, 91, 96, 100, 101, 102, 114, 122, 125, 126, 134, 135, 144, 148, 154, 155 and 156 when numbered in accordance with SEQ ID NO: 122.
59. An isolated nucleic acid molecule comprising an nucleic acid sequence at least 95% similar to SEQ ID NO: 121, wherein said nucleic acid sequence comprises at least one nucleic acid substitution, wherein said substitution is at the nucleic acid position selected from the group consisting of 35, 112, 160, 194, 197, 206, 240, 269, 273, 287, 298, 301, 305, 340, 365, 374, 376, 400, 404, 431, 442, 461, 463 and 468 when numbered in accordance with SEQ ID NO: 121.
PCT/US2003/017512 2002-03-19 2003-06-04 Therapeutic polypeptides, nucleic acids encoding same, and methods of use WO2004000997A2 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
AU2003272200A AU2003272200A1 (en) 2002-06-04 2003-06-04 Therapeutic polypeptides, nucleic acids encoding same, and methods of use
EP03754372A EP1572948A2 (en) 2002-06-04 2003-06-04 Therapeutic polypeptides, nucleic acids encoding same, and methods of use
CA002486490A CA2486490A1 (en) 2002-03-19 2003-06-04 Therapeutic polypeptides, nucleic acids encoding same, and methods of use

Applications Claiming Priority (42)

Application Number Priority Date Filing Date Title
US36549102P 2002-03-19 2002-03-19
US60/365,491 2002-03-19
US38550402P 2002-06-04 2002-06-04
US60/385,504 2002-06-04
US38604102P 2002-06-05 2002-06-05
US60/386,041 2002-06-05
US38645302P 2002-06-06 2002-06-06
US38697402P 2002-06-06 2002-06-06
US60/386,974 2002-06-06
US60/386,453 2002-06-06
US38700202P 2002-06-07 2002-06-07
US38681602P 2002-06-07 2002-06-07
US60/387,002 2002-06-07
US60/386,816 2002-06-07
US38754002P 2002-06-10 2002-06-10
US60/387,540 2002-06-10
US38765902P 2002-06-11 2002-06-11
US60/387,659 2002-06-11
US38793402P 2002-06-12 2002-06-12
US60/387,934 2002-06-12
US38912302P 2002-06-13 2002-06-13
US60/389,123 2002-06-13
US38974202P 2002-06-17 2002-06-17
US38972902P 2002-06-17 2002-06-17
US60/389,742 2002-06-17
US60/389,729 2002-06-17
US39000602P 2002-06-19 2002-06-19
US60/390,006 2002-06-19
US39670602P 2002-07-17 2002-07-17
US60/396,706 2002-07-17
US40283202P 2002-08-12 2002-08-12
US60/402,832 2002-08-12
US40348602P 2002-08-13 2002-08-13
US60/403,486 2002-08-13
US40352202P 2002-08-14 2002-08-14
US60/403,522 2002-08-14
US40374802P 2002-08-15 2002-08-15
US60/403,748 2002-08-15
US38703702P 2002-11-06 2002-11-06
US60/387,037 2002-11-06
US10/454,246 2003-06-03
US10/454,246 US20050053930A1 (en) 2000-07-18 2003-06-03 Therapeutic polypeptides, nucleic acids encoding same, and methods of use

Publications (2)

Publication Number Publication Date
WO2004000997A2 true WO2004000997A2 (en) 2003-12-31
WO2004000997A3 WO2004000997A3 (en) 2007-08-16

Family

ID=34140035

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2003/017512 WO2004000997A2 (en) 2002-03-19 2003-06-04 Therapeutic polypeptides, nucleic acids encoding same, and methods of use

Country Status (2)

Country Link
CA (1) CA2486490A1 (en)
WO (1) WO2004000997A2 (en)

Cited By (73)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007013575A3 (en) * 2005-07-28 2007-10-25 Oncotherapy Science Inc Method for diagnosing and treating renal cell carcinoma
EP2260858A2 (en) 2003-11-06 2010-12-15 Seattle Genetics, Inc. Monomethylvaline compounds capable of conjugation to ligands
WO2011000054A1 (en) 2009-07-03 2011-01-06 Avipep Pty Ltd Immuno-conjugates and methods for producing them
EP2286844A2 (en) 2004-06-01 2011-02-23 Genentech, Inc. Antibody-drug conjugates and methods
WO2011031870A1 (en) 2009-09-09 2011-03-17 Centrose, Llc Extracellular targeted drug conjugates
WO2011056983A1 (en) 2009-11-05 2011-05-12 Genentech, Inc. Zirconium-radiolabeled, cysteine engineered antibody conjugates
WO2011075786A1 (en) 2009-12-23 2011-06-30 Avipep Pty Ltd Immuno-conjugates and methods for producing them 2
WO2011130598A1 (en) 2010-04-15 2011-10-20 Spirogen Limited Pyrrolobenzodiazepines and conjugates thereof
WO2011156328A1 (en) 2010-06-08 2011-12-15 Genentech, Inc. Cysteine engineered antibodies and conjugates
WO2012074757A1 (en) 2010-11-17 2012-06-07 Genentech, Inc. Alaninyl maytansinol antibody conjugates
WO2012142662A1 (en) 2011-04-21 2012-10-26 Garvan Institute Of Medical Research Modified variable domain molecules and methods for producing and using them b
WO2012155019A1 (en) 2011-05-12 2012-11-15 Genentech, Inc. Multiple reaction monitoring lc-ms/ms method to detect therapeutic antibodies in animal samples using framework signature pepides
WO2013130093A1 (en) 2012-03-02 2013-09-06 Genentech, Inc. Biomarkers for treatment with anti-tubulin chemotherapeutic compounds
WO2014057074A1 (en) 2012-10-12 2014-04-17 Spirogen Sàrl Pyrrolobenzodiazepines and conjugates thereof
WO2014140174A1 (en) 2013-03-13 2014-09-18 Spirogen Sàrl Pyrrolobenzodiazepines and conjugates thereof
WO2014140862A2 (en) 2013-03-13 2014-09-18 Spirogen Sarl Pyrrolobenzodiazepines and conjugates thereof
WO2014159981A2 (en) 2013-03-13 2014-10-02 Spirogen Sarl Pyrrolobenzodiazepines and conjugates thereof
WO2015023355A1 (en) 2013-08-12 2015-02-19 Genentech, Inc. 1-(chloromethyl)-2,3-dihydro-1h-benzo[e]indole dimer antibody-drug conjugate compounds, and methods of use and treatment
WO2015095223A2 (en) 2013-12-16 2015-06-25 Genentech, Inc. Peptidomimetic compounds and antibody-drug conjugates thereof
WO2015095212A1 (en) 2013-12-16 2015-06-25 Genentech, Inc. 1-(chloromethyl)-2,3-dihydro-1h-benzo[e]indole dimer antibody-drug conjugate compounds, and methods of use and treatment
WO2015095227A2 (en) 2013-12-16 2015-06-25 Genentech, Inc. Peptidomimetic compounds and antibody-drug conjugates thereof
US9068991B2 (en) 2009-06-08 2015-06-30 Singulex, Inc. Highly sensitive biomarker panels
WO2016040856A2 (en) 2014-09-12 2016-03-17 Genentech, Inc. Cysteine engineered antibodies and conjugates
WO2016037644A1 (en) 2014-09-10 2016-03-17 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
WO2016040825A1 (en) 2014-09-12 2016-03-17 Genentech, Inc. Anthracycline disulfide intermediates, antibody-drug conjugates and methods
WO2016090050A1 (en) 2014-12-03 2016-06-09 Genentech, Inc. Quaternary amine compounds and antibody-drug conjugates thereof
EP3088004A1 (en) 2004-09-23 2016-11-02 Genentech, Inc. Cysteine engineered antibodies and conjugates
WO2017059289A1 (en) 2015-10-02 2017-04-06 Genentech, Inc. Pyrrolobenzodiazepine antibody drug conjugates and methods of use
WO2017064675A1 (en) 2015-10-16 2017-04-20 Genentech, Inc. Hindered disulfide drug conjugates
WO2017068511A1 (en) 2015-10-20 2017-04-27 Genentech, Inc. Calicheamicin-antibody-drug conjugates and methods of use
US9719999B2 (en) 2006-04-04 2017-08-01 Singulex, Inc. Highly sensitive system and method for analysis of troponin
WO2017165734A1 (en) 2016-03-25 2017-09-28 Genentech, Inc. Multiplexed total antibody and antibody-conjugated drug quantification assay
EP3235820A1 (en) 2014-09-17 2017-10-25 Genentech, Inc. Pyrrolobenzodiazepines and antibody disulfide conjugates thereof
WO2017201449A1 (en) 2016-05-20 2017-11-23 Genentech, Inc. Protac antibody conjugates and methods of use
WO2017205741A1 (en) 2016-05-27 2017-11-30 Genentech, Inc. Bioanalytical method for the characterization of site-specific antibody-drug conjugates
WO2017214024A1 (en) 2016-06-06 2017-12-14 Genentech, Inc. Silvestrol antibody-drug conjugates and methods of use
WO2018031662A1 (en) 2016-08-11 2018-02-15 Genentech, Inc. Pyrrolobenzodiazepine prodrugs and antibody conjugates thereof
US9919056B2 (en) 2012-10-12 2018-03-20 Adc Therapeutics S.A. Pyrrolobenzodiazepine-anti-CD22 antibody conjugates
US9931414B2 (en) 2012-10-12 2018-04-03 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
US9931415B2 (en) 2012-10-12 2018-04-03 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
WO2018065501A1 (en) 2016-10-05 2018-04-12 F. Hoffmann-La Roche Ag Methods for preparing antibody drug conjugates
US9950078B2 (en) 2013-10-11 2018-04-24 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
US9956299B2 (en) 2013-10-11 2018-05-01 Medimmune Limited Pyrrolobenzodiazepine—antibody conjugates
US9977031B2 (en) 2006-04-04 2018-05-22 Singulex, Inc. Highly sensitive system and method for analysis of troponin
US10010624B2 (en) 2013-10-11 2018-07-03 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
US10029018B2 (en) 2013-10-11 2018-07-24 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
WO2019060398A1 (en) 2017-09-20 2019-03-28 Ph Pharma Co., Ltd. Thailanstatin analogs
US10392393B2 (en) 2016-01-26 2019-08-27 Medimmune Limited Pyrrolobenzodiazepines
US10420777B2 (en) 2014-09-12 2019-09-24 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
US10544223B2 (en) 2017-04-20 2020-01-28 Adc Therapeutics Sa Combination therapy with an anti-axl antibody-drug conjugate
US10543279B2 (en) 2016-04-29 2020-01-28 Medimmune Limited Pyrrolobenzodiazepine conjugates and their use for the treatment of cancer
WO2020049286A1 (en) 2018-09-03 2020-03-12 Femtogenix Limited Polycyclic amides as cytotoxic agents
WO2020086858A1 (en) 2018-10-24 2020-04-30 Genentech, Inc. Conjugated chemical inducers of degradation and methods of use
WO2020123275A1 (en) 2018-12-10 2020-06-18 Genentech, Inc. Photocrosslinking peptides for site specific conjugation to fc-containing proteins
US10695439B2 (en) 2016-02-10 2020-06-30 Medimmune Limited Pyrrolobenzodiazepine conjugates
US10695433B2 (en) 2012-10-12 2020-06-30 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
WO2020157491A1 (en) 2019-01-29 2020-08-06 Femtogenix Limited G-a crosslinking cytotoxic agents
US10736903B2 (en) 2012-10-12 2020-08-11 Medimmune Limited Pyrrolobenzodiazepine-anti-PSMA antibody conjugates
US10751346B2 (en) 2012-10-12 2020-08-25 Medimmune Limited Pyrrolobenzodiazepine—anti-PSMA antibody conjugates
US10780096B2 (en) 2014-11-25 2020-09-22 Adc Therapeutics Sa Pyrrolobenzodiazepine-antibody conjugates
US10799595B2 (en) 2016-10-14 2020-10-13 Medimmune Limited Pyrrolobenzodiazepine conjugates
US11059893B2 (en) 2015-04-15 2021-07-13 Bergenbio Asa Humanized anti-AXL antibodies
US11135303B2 (en) 2011-10-14 2021-10-05 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
US11160872B2 (en) 2017-02-08 2021-11-02 Adc Therapeutics Sa Pyrrolobenzodiazepine-antibody conjugates
WO2022023735A1 (en) 2020-07-28 2022-02-03 Femtogenix Limited Cytotoxic agents
US11318211B2 (en) 2017-06-14 2022-05-03 Adc Therapeutics Sa Dosage regimes for the administration of an anti-CD19 ADC
US11352324B2 (en) 2018-03-01 2022-06-07 Medimmune Limited Methods
US11370801B2 (en) 2017-04-18 2022-06-28 Medimmune Limited Pyrrolobenzodiazepine conjugates
US11517626B2 (en) 2016-02-10 2022-12-06 Medimmune Limited Pyrrolobenzodiazepine antibody conjugates
US11524969B2 (en) 2018-04-12 2022-12-13 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof as antitumour agents
US11612665B2 (en) 2017-02-08 2023-03-28 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
US11649250B2 (en) 2017-08-18 2023-05-16 Medimmune Limited Pyrrolobenzodiazepine conjugates
US11702473B2 (en) 2015-04-15 2023-07-18 Medimmune Limited Site-specific antibody-drug conjugates

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2894961C (en) 2012-12-21 2020-09-15 Spirogen Sarl Pyrrolobenzodiazepines and conjugates thereof
AU2013366490B9 (en) 2012-12-21 2018-02-01 Medimmune Limited Unsymmetrical pyrrolobenzodiazepines-dimers for use in the treatment of proliferative and autoimmune diseases

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7041284B2 (en) * 1993-10-25 2006-05-09 Canji, Inc. Recombinant adenoviral vector and method of use

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7041284B2 (en) * 1993-10-25 2006-05-09 Canji, Inc. Recombinant adenoviral vector and method of use

Cited By (95)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2478912A1 (en) 2003-11-06 2012-07-25 Seattle Genetics, Inc. Auristatin conjugates with anti-HER2 or anti-CD22 antibodies and their use in therapy
EP2260858A2 (en) 2003-11-06 2010-12-15 Seattle Genetics, Inc. Monomethylvaline compounds capable of conjugation to ligands
EP3858387A1 (en) 2003-11-06 2021-08-04 Seagen Inc. Monomethylvaline compounds capable of conjugation to ligands
EP3434275A1 (en) 2003-11-06 2019-01-30 Seattle Genetics, Inc. Assay for cancer cells based on the use of auristatin conjugates with antibodies
EP2489364A1 (en) 2003-11-06 2012-08-22 Seattle Genetics, Inc. Monomethylvaline compounds onjugated to antibodies
EP2486933A1 (en) 2003-11-06 2012-08-15 Seattle Genetics, Inc. Monomethylvaline compounds conjugated with antibodies
EP2286844A2 (en) 2004-06-01 2011-02-23 Genentech, Inc. Antibody-drug conjugates and methods
EP3088004A1 (en) 2004-09-23 2016-11-02 Genentech, Inc. Cysteine engineered antibodies and conjugates
WO2007013575A3 (en) * 2005-07-28 2007-10-25 Oncotherapy Science Inc Method for diagnosing and treating renal cell carcinoma
US9719999B2 (en) 2006-04-04 2017-08-01 Singulex, Inc. Highly sensitive system and method for analysis of troponin
US9977031B2 (en) 2006-04-04 2018-05-22 Singulex, Inc. Highly sensitive system and method for analysis of troponin
US9068991B2 (en) 2009-06-08 2015-06-30 Singulex, Inc. Highly sensitive biomarker panels
WO2011000054A1 (en) 2009-07-03 2011-01-06 Avipep Pty Ltd Immuno-conjugates and methods for producing them
WO2011031870A1 (en) 2009-09-09 2011-03-17 Centrose, Llc Extracellular targeted drug conjugates
WO2011056983A1 (en) 2009-11-05 2011-05-12 Genentech, Inc. Zirconium-radiolabeled, cysteine engineered antibody conjugates
WO2011075786A1 (en) 2009-12-23 2011-06-30 Avipep Pty Ltd Immuno-conjugates and methods for producing them 2
WO2011130598A1 (en) 2010-04-15 2011-10-20 Spirogen Limited Pyrrolobenzodiazepines and conjugates thereof
WO2011156328A1 (en) 2010-06-08 2011-12-15 Genentech, Inc. Cysteine engineered antibodies and conjugates
WO2012074757A1 (en) 2010-11-17 2012-06-07 Genentech, Inc. Alaninyl maytansinol antibody conjugates
WO2012142662A1 (en) 2011-04-21 2012-10-26 Garvan Institute Of Medical Research Modified variable domain molecules and methods for producing and using them b
EP3103810A2 (en) 2011-04-21 2016-12-14 Garvan Institute of Medical Research Modified variable domain molecules and methods for producing and using them
WO2012155019A1 (en) 2011-05-12 2012-11-15 Genentech, Inc. Multiple reaction monitoring lc-ms/ms method to detect therapeutic antibodies in animal samples using framework signature pepides
US11135303B2 (en) 2011-10-14 2021-10-05 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
WO2013130093A1 (en) 2012-03-02 2013-09-06 Genentech, Inc. Biomarkers for treatment with anti-tubulin chemotherapeutic compounds
US11779650B2 (en) 2012-10-12 2023-10-10 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
US10780181B2 (en) 2012-10-12 2020-09-22 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
US11690918B2 (en) 2012-10-12 2023-07-04 Medimmune Limited Pyrrolobenzodiazepine-anti-CD22 antibody conjugates
US11771775B2 (en) 2012-10-12 2023-10-03 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
EP2839860A1 (en) 2012-10-12 2015-02-25 Spirogen Sàrl Pyrrolobenzodiazepines and conjugates thereof
US10994023B2 (en) 2012-10-12 2021-05-04 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
WO2014057074A1 (en) 2012-10-12 2014-04-17 Spirogen Sàrl Pyrrolobenzodiazepines and conjugates thereof
US9931415B2 (en) 2012-10-12 2018-04-03 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
US10799596B2 (en) 2012-10-12 2020-10-13 Adc Therapeutics S.A. Pyrrolobenzodiazepine-antibody conjugates
US9919056B2 (en) 2012-10-12 2018-03-20 Adc Therapeutics S.A. Pyrrolobenzodiazepine-anti-CD22 antibody conjugates
US10751346B2 (en) 2012-10-12 2020-08-25 Medimmune Limited Pyrrolobenzodiazepine—anti-PSMA antibody conjugates
US11701430B2 (en) 2012-10-12 2023-07-18 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
US10736903B2 (en) 2012-10-12 2020-08-11 Medimmune Limited Pyrrolobenzodiazepine-anti-PSMA antibody conjugates
US10722594B2 (en) 2012-10-12 2020-07-28 Adc Therapeutics S.A. Pyrrolobenzodiazepine-anti-CD22 antibody conjugates
US10695433B2 (en) 2012-10-12 2020-06-30 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
US10646584B2 (en) 2012-10-12 2020-05-12 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
US10335497B2 (en) 2012-10-12 2019-07-02 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
US9889207B2 (en) 2012-10-12 2018-02-13 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
US9931414B2 (en) 2012-10-12 2018-04-03 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
WO2014140862A2 (en) 2013-03-13 2014-09-18 Spirogen Sarl Pyrrolobenzodiazepines and conjugates thereof
WO2014140174A1 (en) 2013-03-13 2014-09-18 Spirogen Sàrl Pyrrolobenzodiazepines and conjugates thereof
WO2014159981A2 (en) 2013-03-13 2014-10-02 Spirogen Sarl Pyrrolobenzodiazepines and conjugates thereof
WO2015023355A1 (en) 2013-08-12 2015-02-19 Genentech, Inc. 1-(chloromethyl)-2,3-dihydro-1h-benzo[e]indole dimer antibody-drug conjugate compounds, and methods of use and treatment
US9950078B2 (en) 2013-10-11 2018-04-24 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
US10010624B2 (en) 2013-10-11 2018-07-03 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
US10029018B2 (en) 2013-10-11 2018-07-24 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
US9956299B2 (en) 2013-10-11 2018-05-01 Medimmune Limited Pyrrolobenzodiazepine—antibody conjugates
WO2015095227A2 (en) 2013-12-16 2015-06-25 Genentech, Inc. Peptidomimetic compounds and antibody-drug conjugates thereof
WO2015095223A2 (en) 2013-12-16 2015-06-25 Genentech, Inc. Peptidomimetic compounds and antibody-drug conjugates thereof
WO2015095212A1 (en) 2013-12-16 2015-06-25 Genentech, Inc. 1-(chloromethyl)-2,3-dihydro-1h-benzo[e]indole dimer antibody-drug conjugate compounds, and methods of use and treatment
WO2016037644A1 (en) 2014-09-10 2016-03-17 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
US10188746B2 (en) 2014-09-10 2019-01-29 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
WO2016040856A2 (en) 2014-09-12 2016-03-17 Genentech, Inc. Cysteine engineered antibodies and conjugates
WO2016040825A1 (en) 2014-09-12 2016-03-17 Genentech, Inc. Anthracycline disulfide intermediates, antibody-drug conjugates and methods
US10420777B2 (en) 2014-09-12 2019-09-24 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
EP3235820A1 (en) 2014-09-17 2017-10-25 Genentech, Inc. Pyrrolobenzodiazepines and antibody disulfide conjugates thereof
US10780096B2 (en) 2014-11-25 2020-09-22 Adc Therapeutics Sa Pyrrolobenzodiazepine-antibody conjugates
WO2016090050A1 (en) 2014-12-03 2016-06-09 Genentech, Inc. Quaternary amine compounds and antibody-drug conjugates thereof
US11702473B2 (en) 2015-04-15 2023-07-18 Medimmune Limited Site-specific antibody-drug conjugates
US11059893B2 (en) 2015-04-15 2021-07-13 Bergenbio Asa Humanized anti-AXL antibodies
WO2017059289A1 (en) 2015-10-02 2017-04-06 Genentech, Inc. Pyrrolobenzodiazepine antibody drug conjugates and methods of use
WO2017064675A1 (en) 2015-10-16 2017-04-20 Genentech, Inc. Hindered disulfide drug conjugates
WO2017068511A1 (en) 2015-10-20 2017-04-27 Genentech, Inc. Calicheamicin-antibody-drug conjugates and methods of use
US10392393B2 (en) 2016-01-26 2019-08-27 Medimmune Limited Pyrrolobenzodiazepines
US11517626B2 (en) 2016-02-10 2022-12-06 Medimmune Limited Pyrrolobenzodiazepine antibody conjugates
US10695439B2 (en) 2016-02-10 2020-06-30 Medimmune Limited Pyrrolobenzodiazepine conjugates
EP4273551A2 (en) 2016-03-25 2023-11-08 F. Hoffmann-La Roche AG Multiplexed total antibody and antibody-conjugated drug quantification assay
WO2017165734A1 (en) 2016-03-25 2017-09-28 Genentech, Inc. Multiplexed total antibody and antibody-conjugated drug quantification assay
US10543279B2 (en) 2016-04-29 2020-01-28 Medimmune Limited Pyrrolobenzodiazepine conjugates and their use for the treatment of cancer
WO2017201449A1 (en) 2016-05-20 2017-11-23 Genentech, Inc. Protac antibody conjugates and methods of use
WO2017205741A1 (en) 2016-05-27 2017-11-30 Genentech, Inc. Bioanalytical method for the characterization of site-specific antibody-drug conjugates
WO2017214024A1 (en) 2016-06-06 2017-12-14 Genentech, Inc. Silvestrol antibody-drug conjugates and methods of use
WO2018031662A1 (en) 2016-08-11 2018-02-15 Genentech, Inc. Pyrrolobenzodiazepine prodrugs and antibody conjugates thereof
WO2018065501A1 (en) 2016-10-05 2018-04-12 F. Hoffmann-La Roche Ag Methods for preparing antibody drug conjugates
US10799595B2 (en) 2016-10-14 2020-10-13 Medimmune Limited Pyrrolobenzodiazepine conjugates
US11612665B2 (en) 2017-02-08 2023-03-28 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
US11813335B2 (en) 2017-02-08 2023-11-14 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
US11160872B2 (en) 2017-02-08 2021-11-02 Adc Therapeutics Sa Pyrrolobenzodiazepine-antibody conjugates
US11370801B2 (en) 2017-04-18 2022-06-28 Medimmune Limited Pyrrolobenzodiazepine conjugates
US10544223B2 (en) 2017-04-20 2020-01-28 Adc Therapeutics Sa Combination therapy with an anti-axl antibody-drug conjugate
US11318211B2 (en) 2017-06-14 2022-05-03 Adc Therapeutics Sa Dosage regimes for the administration of an anti-CD19 ADC
US11938192B2 (en) 2017-06-14 2024-03-26 Medimmune Limited Dosage regimes for the administration of an anti-CD19 ADC
US11649250B2 (en) 2017-08-18 2023-05-16 Medimmune Limited Pyrrolobenzodiazepine conjugates
WO2019060398A1 (en) 2017-09-20 2019-03-28 Ph Pharma Co., Ltd. Thailanstatin analogs
US11352324B2 (en) 2018-03-01 2022-06-07 Medimmune Limited Methods
US11524969B2 (en) 2018-04-12 2022-12-13 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof as antitumour agents
WO2020049286A1 (en) 2018-09-03 2020-03-12 Femtogenix Limited Polycyclic amides as cytotoxic agents
WO2020086858A1 (en) 2018-10-24 2020-04-30 Genentech, Inc. Conjugated chemical inducers of degradation and methods of use
WO2020123275A1 (en) 2018-12-10 2020-06-18 Genentech, Inc. Photocrosslinking peptides for site specific conjugation to fc-containing proteins
WO2020157491A1 (en) 2019-01-29 2020-08-06 Femtogenix Limited G-a crosslinking cytotoxic agents
WO2022023735A1 (en) 2020-07-28 2022-02-03 Femtogenix Limited Cytotoxic agents

Also Published As

Publication number Publication date
CA2486490A1 (en) 2003-12-31
WO2004000997A3 (en) 2007-08-16

Similar Documents

Publication Publication Date Title
WO2004000997A2 (en) Therapeutic polypeptides, nucleic acids encoding same, and methods of use
WO2003083046A2 (en) Novel proteins and nucleic acids encoding same
US20040048256A1 (en) Novel proteins and nucleic acids encoding same
WO2003022998A2 (en) Therapeutic polypeptides, nucleic acids encoding same, and methods of use
WO2003078572A2 (en) Therapeutic polypeptides, nucleic acids encoding same and methods of use
WO2003031571A2 (en) Novel human proteins, polynucleotides encoding them and methods of using the same
EP1572922A2 (en) Therapeutic polypeptides, nucleic acids encoding same, and methods of use
EP1532238A2 (en) Therapeutic polypeptides, nucleic acids encoding same, and methods of use
WO2003085124A2 (en) Therapeutic polypeptides, nucleic acids encoding same, and methods of use
US20050053930A1 (en) Therapeutic polypeptides, nucleic acids encoding same, and methods of use
US20060234255A1 (en) Novel proteins and nucleic acids encoding same
US20040067505A1 (en) Therapeutic polypeptides, nucleic acids encoding same, and methods of use
US20030229016A1 (en) Novel human proteins, polynucleotides encoding them and methods of using the same
WO2004089282A2 (en) Therapeutic polypeptides, nucleic acids encoding same, and methods of use
US20040229779A1 (en) Therapeutic polypeptides, nucleic acids encoding same, and methods of use
CA2471480A1 (en) Therapeutic polypeptides, nucleic acids encoding same, and methods of use
CA2481039A1 (en) Therapeutic polypeptides, nucleic acids encoding same, and methods of use
EP1581618A2 (en) Therapeutic polypeptides, nucleic acids encoding same, and methods of use
US20040002453A1 (en) Therapeutic polypeptides, nucleic acids encoding same, and methods of use
EP1504026A2 (en) Therapeutic polypeptides, nucleic acids encodings same, and methods of use
EP1572948A2 (en) Therapeutic polypeptides, nucleic acids encoding same, and methods of use
EP1513864A2 (en) Therapeutic polypeptides, nucleic acids encoding same, and methods of use
US20060234257A1 (en) Therapeutic polypeptides, nucleic acids encoding same, and methods of use
AU2003210852A1 (en) Therapeutic polypeptides, nucleic acids encoding same, and methods of use
EP1565199A2 (en) Therapeutic polypeptides,nucleic acids encoding same, and methods of use

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NI NO NZ OM PH PL PT RO RU SC SD SE SG SK SL TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2003272200

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2486490

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2003754372

Country of ref document: EP

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWP Wipo information: published in national office

Ref document number: 2003754372

Country of ref document: EP

NENP Non-entry into the national phase in:

Ref country code: JP

WWW Wipo information: withdrawn in national office

Country of ref document: JP

WWW Wipo information: withdrawn in national office

Ref document number: 2003754372

Country of ref document: EP