US20080153147A1 - Compositions, organisms and methodologies employing a novel human kinase - Google Patents

Compositions, organisms and methodologies employing a novel human kinase Download PDF

Info

Publication number
US20080153147A1
US20080153147A1 US11/982,836 US98283607A US2008153147A1 US 20080153147 A1 US20080153147 A1 US 20080153147A1 US 98283607 A US98283607 A US 98283607A US 2008153147 A1 US2008153147 A1 US 2008153147A1
Authority
US
United States
Prior art keywords
mrck1
seq
sequence
gene
kinase
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
US11/982,836
Inventor
Wei Liu
Leeying Wu
Huimin Chen
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Wyeth LLC
Original Assignee
Wyeth LLC
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
Application filed by Wyeth LLC filed Critical Wyeth LLC
Priority to US11/982,836 priority Critical patent/US20080153147A1/en
Publication of US20080153147A1 publication Critical patent/US20080153147A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/10Transferases (2.)
    • C12N9/12Transferases (2.) transferring phosphorus containing groups, e.g. kinases (2.7)
    • C12N9/1205Phosphotransferases with an alcohol group as acceptor (2.7.1), e.g. protein kinases

Definitions

  • the present invention relates to compositions, organisms and methodologies employing a novel human protein kinase, MRCK1, which has 65% sequence homology to rat myotonic dystrophy kinase-related Cdc42 binding kinase (MRCK).
  • This invention can be used for diagnosing, prognosing and treating kinase-related diseases and, in particular, diseases associated with aberrant expression of MRCK1.
  • ATP adenosine triphosphate molecules
  • Phosphorylation occurs in response to extracellular signals (hormones, neurotransmitters, growth and differentiation factors, etc.), cell cycle checkpoints, and environmental or nutritional stresses.
  • the phosphorylation process is roughly analogous to turning on a molecular switch. When the switch goes on, the appropriate protein kinase activates a metabolic enzyme, regulatory protein, receptor, cytoskeletal protein, ion channel or pump, or transcription factor.
  • the kinases comprise the largest known protein group, a superfamily of enzymes with widely varied functions and specificities. They are usually named after their substrate, their regulatory molecules, or some aspect of a mutant phenotype. With regard to substrates, the protein kinases may be roughly divided into two groups: those that phosphorylate tyrosine residues (protein tyrosine kinases, PTK) and those that phosphorylate serine or threonine residues (serine/threonine kinases, STK). A few protein kinases have dual specificity and phosphorylate threonine and tyrosine residues. Almost all kinases contain a similar 250-300 amino acid catalytic domain.
  • the primary structure of the kinase domains is conserved and can be further subdivided into 11 subdomains.
  • the N-terminal of the kinase domain which contains subdomains I-IV, generally folds into a lobe-like structure that binds and orients the ATP (or GTP) donor molecule.
  • the C terminal of the kinase domain forms a larger lobe, which contains subdomains VI-XI, binds the protein substrate and carries out the transfer of the gamma phosphate from ATP to the hydroxyl group of a serine, threonine, or tyrosine residue.
  • Subdomain V spans the two lobes.
  • Each of the 11 subdomains contains specific residues and motifs or patterns of amino acids that are characteristic of that subdomain and are highly conserved.
  • the kinases may be categorized into families by the different amino acid sequences (generally between 5 and 100 residues) located on either side of, or inserted into loops of, the kinase domain. These added amino acid sequences allow the regulation of each kinase as it recognizes and interacts with its target protein.
  • phosphate moiety modulates protein function in multiple ways.
  • a common mechanism involves changes in the catalytic properties (Vmax and Km) of an enzyme, leading to its activation or inactivation.
  • a second widely recognized mechanism involves promoting protein-protein interactions.
  • An example of this is the tyrosine autophosphorylation of the ligand-activated EGF receptor tyrosine kinase.
  • This event triggers the high-affinity binding to the phosphotyrosine residue on the receptor's C-terminal intracellular domain to the SH2 motif of an adaptor molecule Grb2.
  • Grb2 in turn, binds through its SH3 motif to a second adaptor molecule, such as SHC.
  • SHC second adaptor molecule
  • Serine and threonine phosphorylation events also have been recently recognized to exert their biological function through protein-protein interaction events that are mediated by the high-affinity binding of phosphoserine and phosphothreonine to the WW motifs present in a large variety of proteins.
  • a third important outcome of protein phosphorylation is changes in the subcellular localization of the substrate.
  • nuclear import and export events in a large diversity of proteins are regulated by protein phosphorylation.
  • kinases are involved in regulatory cascades wherein their substrates may include other kinases whose activities are regulated by their phosphorylation state. Ultimately the activities of some downstream effectors are modulated by phosphorylation resulting from activation of such a pathway.
  • MRCKs Myotonic dystrophy kinase-related Cdc42 binding kinases
  • MRCKs are serine/threonine kinases.
  • MRCKs have been implicated in the morphological activities of Cdc42 in non-neural cells and are suggest to be downstream effectors of Cdc42 in cytoskeletal reorganization.
  • MRCKs interact with the GTP-bound form of Cdc42 and, to a lesser extent, the GTP-bound form of Rac.
  • the catalytic domain of MRCKs phosphorylates non-muscle myosin light chain 2 at serine 19. The phosphorylation is believed to be involved in myosin contractile activity and associated changes in the organization of actin microfilaments in intact cells.
  • MRCK alpha and Rho-binding kinase (ROK) alpha are believed to have contrasting roles in regulating neurite morphology.
  • ROK alpha acts downstream of RhoA in inducing neurite retraction
  • MRCK alpha acts downstream of Cdc42/Rac1 in promoting neurite outgrowth.
  • the neurite outgrowth induced by either kinase-dead ROK alpha or nerve growth factor can be effectively blocked by a kinase-dead and p21-binding deficient MRCK alpha mutant.
  • expression of kinase-dead MRCK alpha blocks Cdc42 V12 -dependent formation of focal complexes and peripheral microspikes. Microinjection of a plasmid encoding MRCK alpha results in actin and myosin reorganization.
  • MRCKs have multiple functional domains. These domains include three coiled-coil alpha-helix domains, a cysteine-rich motif resembling those of protein kinase C and n-chimaerin, and a Pleckstrin homology domain. Native MRCK kinases tend to form high-molecular-weight multimers. The intermolecular interactions among the three coiled-coil domains and the N-terminal region preceding the kinase domain in MRCK alpha are believed to be responsible for the multimerization.
  • MRCK alpha can be activated by the N-terminus-mediated dimerization.
  • the dimerization leads to trans-autophosphorylation of MRCK kinases.
  • MRCK alpha kinases can be negatively regulated through intramolecular interactions between the two distal coiled-coil domains. Deletion of these coiled-coil domains leads to a more active kinase, showing the negative autoregulatory role of these domains.
  • the N-terminus-mediated dimerization and the intramolecular interaction between the distal coiled-coil domains are considered to be two mutually exclusive events, which regulate the catalytic state of MRCK kinases.
  • the present invention discloses compositions, organisms and methodologies employing a novel human protein kinase.
  • the new human protein kinase shares sequence homology with rat MRCK alpha.
  • the gene encoding the new protein is localized at 11q13.1 in human chromosome 11. This new gene is hereinafter referred to as the MRCK1 gene, and its encoded protein(s) is referred to as MRCK1 or MRCK1 kinase.
  • MRCK1 has multiple domains including at least a kinase domain, a protein kinase C terminal domain, a myosin tail domain, a phorbol esters/diacylglycerol binding domain (DAG_PE binding domain), a Pleckstrin homology domain, a CNH domain, and a P21-Rho-binding domain.
  • DAG_PE binding domain phorbol esters/diacylglycerol binding domain
  • Pleckstrin homology domain a CNH domain
  • P21-Rho-binding domain The sequence and structure similarity between MRCK1 and rat MRCK alpha indicates that MRCK1 may be involved in Cdc42-mediated cytoskeleton reorganization in human cells.
  • the kinase domain in MRCK1 shows 100% sequence alignment with the consensus sequences of the catalytic domains of at least two subfamilies of protein kinases.
  • the utility of various kinase domains is known in the art.
  • the utilities of other domains, such as protein kinase C terminal domains, Phorbol esters/diacylglycerol binding domains, Pleckstrin homology domains, CNH domains, and P21-Rho-binding domains are also known in the art.
  • the unique peptide sequences, and nucleic acid sequences that encode the peptides can be used as models for the development of human therapeutic targets, aid in the identification of therapeutic proteins, and serve as targets for the development of human therapeutic agents that modulate kinase activity in cells and tissues that express the kinase.
  • the invention provides isolated polynucleotides comprising a nucleotide sequence encoding MRCK1 or a variant of MRCK1.
  • the invention provides isolated polypeptides comprising the amino acid sequence of MRCK1 or a variant of MRCK1.
  • the invention provides agents that modulate expression levels of the MRCK1 gene or an activity of MRCK1.
  • the invention also provides methods for (a) detecting polynucleotides comprising a nucleotide sequence encoding MRCK1 or a variant of MRCK1 and (b) detecting polypeptides comprising an amino acid sequence of MRCK1 or a variant of MRCK1 in a biological sample.
  • the invention further provides methods for screening agents that modulate expression level of the MRCK1 gene or an activity of MRCK1.
  • the invention further provides cell lines harboring the MRCK1 gene, animals transgenic for the MRCK1 gene, and animals with interrupted MRCK1 gene (MRCK1 knockout animals). These cell lines and animals can be used to study the functions of MRCK1.
  • the invention provides polynucleotides capable of inhibiting MRCK1 gene expression by RNA interference.
  • the invention further provides methods of inhibiting MRCK1 gene expression by introducing siRNAs or other RNAi sequences into target cells.
  • FIG. 1 is the sequence alignment between a fragment of MRCK1 and its corresponding sequence in rat MRCK alpha.
  • FIG. 2 depicts the sequence alignment between another fragment of MRCK1 and its corresponding sequence in rat MRCK alpha.
  • FIG. 3 compares MRCK1's kinase domain to the catalytic domain of a family of Ser/Thr protein kinases.
  • FIG. 4 compares MRCK1 's kinase domain to the kinase domain of another family of protein kinases.
  • FIG. 5 shows the sequence alignment between the kinase domain of MRCK1 and the catalytic domain of a family of tyrosine kinases.
  • FIG. 6 illustrates the sequence alignment between amino acid residues 339 to 398 of MRCK1 and the extension to Ser/Thr-type protein kinases
  • FIG. 7 compares amino acid residues 339 to 398 of MRCK1 to a protein kinase C terminal domain.
  • FIG. 8 shows the sequence alignment between the amino acid residues 882-920 of MRCK1 and a consensus sequence of the DAG_PE bind domains of other proteins.
  • FIG. 9 depicts the sequence alignment between the amino acid residues 953-1060 of MRCK1 and a consensus sequence of the PH bind domains.
  • FIG. 10 illustrates the sequence alignment between the amino acid residues 954-1060 of MRCK1 and another consensus sequence of the PH bind domain.
  • FIG. 11 compares the amino acid residues 1102-1345 of MRCK1 to a consensus sequence of the CNH domains.
  • FIG. 12 shows the sequence alignment between the amino acid residues 1440-1471 of MRCK1 and a consensus sequence of the P21-Rho-binding domains.
  • FIG. 13 compares the amino acid residues 648-786 of MRCK1 to a consensus sequence of the myosin tail domains.
  • FIG. 14 shows the hydrophobicity profile of MRCK1.
  • FIG. 15 shows the position and nucleotide sequence of human MRCK1 probe and PCR primers used for the amplification of the probe sequences.
  • the present invention is based on the sequence information obtained from a newly-developed genomic prediction pipeline. Briefly, the X-ray crystal structures of the catalytic domains of protein kinases were collected and aligned together according to their structural identity/similarities. The alignment was converted into a “scoring matrix” which carried the structural profile of the kinase catalytic domains. This scoring matrix was then used to search the Celera Human Genome database and pull out sequences that have kinase catalytic domains.
  • the present invention provides the amino acid sequence of a human kinase peptide that is highly homologous to rat myotonic dystrophy kinase-related Cdc42 binding kinase (MRCK), cDNA sequences and genomic sequences that encode the kinase peptide, and information about the closest art known protein/peptide/domain that has structural or sequence homology to the kinase of the present invention.
  • MRCK rat myotonic dystrophy kinase-related Cdc42 binding kinase
  • the peptide of the present invention is selected based on its ability to be used for the development of commercially important products and services. Specifically, the present peptides are selected based on homology to a known kinase protein of rat MRCK alpha.
  • a polynucleotide or a polypeptide is “isolated” if it is removed from its native environment. For instance, a polynucleotide or a polypeptide is isolated through a purification process such that the polynucleotide or polypeptide is substantially free of cellular material or free of chemical precursors.
  • the polynucleotide/polypeptide of the present invention can be purified to homogeneity or other degrees of purity. The level of purification will be based on the intended use. As appreciated by one of ordinary skill in the art, a polynucleotide/polypeptide can perform its desired function(s) even in the presence of considerable amounts of other components or molecules.
  • a polynucleotide/polypeptide that is “substantially free of cellular material” includes preparations which have less than about 30% (by weight) other polynucleotides/polypeptides including contaminating polynucleotides/polypeptides. For instance, the preparations can have less than about 20%, less than about 10%, or less than about 5% other polynucleotides/polypeptides. If a polynucleotide/polypeptide preparation is recombinantly produced, it can be substantially free of culture medium, i.e., culture medium components representing less than about 20% by weight of the polynucleotide/polypeptide preparation.
  • the language “substantially free of chemical precursors” includes preparations in which the polynucleotide/polypeptide is separated from chemical precursors or other chemicals that are involved in the synthesis of the polynucleotide/polypeptide.
  • the language “substantially free of chemical precursors” includes kinase preparations having less than about 30% (by weight), less than about 20% (by weight), less than about 10% (by weight), or less than about 5% (by weight) of chemical precursors or other chemicals used in the synthesis.
  • a “polynucleotide” can include any number of nucleotides. For instance, a polynucleotide can have at least 10, 20, 25, 30, 40, 50, 100 or more nucleotides.
  • a polynucleotide can be DNA or RNA, double-stranded or single-stranded.
  • a polynucleotide encodes a polypeptide if the polypeptide is capable of being transcribed and/or translated from the polynucleotide. Transcriptional and/or translational regulatory sequences, such as promoter and/or enhancer(s), can be added to the polynucleotide before said transcription and/or translation occurs.
  • the polynucleotide is singled-stranded, the corresponding double-stranded DNA containing the original polynucleotide and its complementary sequence can be prepared before said transcription and/or translation.
  • a variant of a polynucleotide refers to a polynucleotide that differs from the original polynucleotide by one or more substitutions, additions, and/or deletions.
  • a variant of a polynucleotide can have 1, 2, 5, 10, 15, 20, 25 or more nucleotide substitutions, additions or deletions.
  • the modification(s) is in-frame, i.e., the modified polynucleotide can be transcribed and translated to the original or intended stop codon.
  • the polypeptide encoded by a variant of the original polynucleotide substantially retains such activity.
  • the biological activity is reduced/enhanced by less than 50%, or more preferably, less than 20%, relative to the original activity.
  • a variant of a polynucleotide can be a polynucleotide that is capable of hybridizing to the original polynucleotide, or the complementary sequence thereof, under reduced stringent conditions, preferably stringent conditions, or more preferably, highly stringent conditions.
  • reduced stringent conditions are those that are at least as stringent as conditions A-F; stringent conditions are at least as stringent as conditions G-L; and reduced stringency conditions are at least as stringent as conditions M-R.
  • hybridization is carried out under a given hybridization condition for about 2 hours, followed by two 15-minute washes under the corresponding washing condition(s).
  • the hybrid length is assumed to be that of the hybridizing polynucleotide.
  • the hybrid length can be determined by aligning the sequences of the polynucleotides and identifying the region or regions of optimal sequence complementarity.
  • H SSPE (1xSSPE is 0.15M NaCl, 10 mM NaH 2 PO 4 , and 1.25 mM EDTA, pH 7.4) can be substituted for SSC (1xSSC is 0.15M NaCl and 15 mM sodium citrate) in the hybridization and wash buffers.
  • T B * ⁇ T R *The hybridization temperature for hybrids anticipated to be less than 50 base pairs in length should be 5-10° C. less than the melting temperature (T m ) of the hybrid, where T m is determined according to the following equations.
  • T m melting temperature
  • T m (° C.) 2(# of A + T bases) + 4(# of G + C bases).
  • polypeptide can include any number of amino acid residues.
  • a polypeptide can have at least 5, 10, 15, 20, 30, 40, 50 or more amino acid residues.
  • a “variant of a polypeptide” is a polypeptide that differs from the original polypeptide by one or more substitutions, deletions, and/or insertions. Preferably, these modifications do not substantially change (e.g. reduce or enhance) the original biological function of the polypeptide.
  • a variant can reduce or enhance or maintain the biological activities of the original polypeptide.
  • the biological activities of the variant are reduced or enhanced by less than 50%, or more preferably, less than 20%, relative to the original polypeptide.
  • the ability of a variant to react with antigen-specific antisera can be enhanced or reduced by less than 50%, preferably less than 20%, relative to the original polypeptide.
  • These variants can be prepared and evaluated by modifying the original polypeptide sequence and then determining the reactivity of the modified polypeptide with the antigen-specific antibodies or antisera.
  • a variant polypeptide contains one or more conservative substitutions.
  • a “conservative substitution” is one in which an amino acid is substituted for another amino acid which has similar properties, such that one skilled in the art would expect that the secondary structure and hydropathic nature of the substituted polypeptide will not be substantially changed.
  • Conservative amino acid substitutions can be made on the basis of similarity in polarity, charge, solubility, hydrophobicity, hydrophilicity and/or the amphipathic nature of the residues.
  • Negatively charged amino acids include aspartic acid and glutamic acid, and positively charged amino acids include lysine and arginine.
  • Amino acids having uncharged polar head groups and similar hydrophilicity values include leucine, isoleucine and valine, or glycine and alanine, or asparagine and glutamine, or serine, threonine, phenylalanine and tyrosine.
  • Other groups of amino acids that can produce conservative changes include: (1) ala, pro, gly, glu, asp, gln, asn, ser, thr; (2) cys, ser, tyr, thr; (3) val, ile, leu, met, ala, phe; (4) lys, arg, his; and (5) phe, tyr, trp, his.
  • a polypeptide variant can also contain nonconservative changes.
  • Polypeptide variants can be prepared by the deletion and/or addition of amino acids that have minimal influence on the biological activity, immunogenicity, secondary structure and/or hydropathic nature of the polypeptide. Variants can be for instance by substituting, modifying, deleting or adding one or more amino acids residues in the original sequence. Polypeptide variants preferably exhibit at least 96%, more preferably at least 97%, and most preferably at least 98% sequence homology to the original polypeptide.
  • Polypeptide variants include polypeptides that are modified from the original polypeptides either by a natural process, such as a post-translational modification, or by a chemical modification. These modifications are well-known in the art. Modifications can occur anywhere in the polypeptide, including the backbone, the amino acid side-chains and the amino or carboxyl termini. It will be appreciated that the same type of modification can be present in the same or varying degrees at several sites in a given polypeptide. Also, a given polypeptide can contain many types of modifications. Polypeptides may be branched, for example, as a result of ubiquitination, and they may be cyclic, with or without branching.
  • Cyclic, branched, and branched cyclic polypeptides can result from natural post-translational processes or be made through synthetic methods. Suitable modifications for this invention include acetylation, acylation, ADP-ribosylation, amidation, covalent attachment of flavin, covalent attachment of a heme moiety, covalent attachment of a nucleotide or nucleotide derivative, covalent attachment of a lipid or lipid derivative, covalent attachment of phosphatidylinositol, cross-linking, cyclization, disulfide bond formation, demethylation, formation of covalent cross-links, formation of cysteine, formation of pyroglutamate, formylation, gamma-carboxylation, glycosylation, GPI anchor formation, hydroxylation, iodination, methylation, myristoylation, oxidation, pegylation, proteolytic processing, phosphorylation, prenylation, racemization, selenoylation,
  • modulation includes up-regulation, induction, stimulation, potentiation, inhibition, down-regulation or suppression, or relief of inhibition.
  • a nucleotide sequence is “operably linked” to another nucleotide sequence if the two sequences are placed into a functional relationship.
  • a coding sequence is operably linked to a 5′ regulatory sequence if the 5′ regulatory sequence can initiate transcription of the coding sequence in an in vitro transcription/translation system or in a host cell.
  • “Operably linked” does not require that the DNA sequences being linked are contiguous to each other. Intervening sequences may exist between two operably linked sequences.
  • a “disease-free” human refers to a human who does not have MRCK1-related diseases.
  • Disease-free, tissues or samples refer to cells, tissues or samples obtained from disease-free human(s).
  • a polynucleotide is “capable of hybridizing” to a gene if the polynucleotide can hybridize to at least one of the following sequences: (1) the sequence of an RNA transcript of the gene, (2) the complementary sequence of an RNA transcript of the gene, (3) the cDNA sequence of an RNA transcript of the gene, (4) the complementary sequence of the cDNA sequence of an RNA transcript of the gene, (5) a genomic sequence of the gene, and (6) the complementary sequence of a genomic sequence of the gene.
  • sequence “identity” or “percentage alignment” in an alignment can be determined by the standard protein-protein or nucleotide-nucleotide BLAST programs (i.e., blastp or blastn, respectively). Sequence identity can also be determined by the BLAST2 program. Suitable BLAST program can be found at the BLAST web site maintained by the National Center of Biotechnology Information (NCBI) (National Library of Medicine)
  • the present invention identifies a new human gene (MRCK1 gene) that encodes a protein that is highly homologous to rat MRCK alpha.
  • the nucleotide and amino acid sequences of the protein encoded by the MRCK1 gene are depicted in SEQ ID NOS:1 and 2, respectively.
  • FIGS. 1 and 2 show the sequence alignment between SEQ ID NO:2 (denoted as “Query”) and the amino acid sequence of rat MRCK alpha (denoted as “Sbjct”).
  • the alignment is a result of blast search of the “all non-redundant GenBank CDS database” in Entrez.
  • the blast search uses BLASTP 2.2.3 algorithm [Apr. 24, 2002], which is described in Altschul et al., Nucleic Acids Res., 25: 3389-3402 (1997).
  • FIG. 1 shows that the sequence consisting of amino acid residues 40 to 434 of MRCK1 (Query) and the sequence consisting of amino acid residues 46 to 463 of rat MRCK alpha (Sbjct) have 65% sequence identity with 78% positives, a score of 588 bits (1516) and an E value of 1 ⁇ 10 ⁇ 172 .
  • FIG. 2 shows that the sequence consisting of amino acid residues 551 to 1457 of MRCK1 (Query) and the sequence consisting of amino acid residues 684 to 1588 of rat MRCK alpha (Sbjct) have 38% sequence identity with 54% positives, a score of 615 bits (1586) and an E value of 1 ⁇ 10 ⁇ 166 .
  • the rat MRCK alpha sequence used in the alignment has Entrez Database accession number AAC02941.1 or NP — 446109.1.
  • the same approach is used to compare MRCK1 to a predicted mouse protein, which is similar to Ser/Thr protein kinase related to the myotonic dystrophy protein kinase.
  • the sequence of the mouse protein has Entrez Database accession number XP — 140553.1. Alignment shows that amino acid residues 40 to 1574 of the mouse sequence (Sbjct) and the sequence consisting of amino acid residues 40 to 1548 of MRCK1 (Query) have 70% sequence identity with 73% positives, a score of 1981 bits (5133) and an E value of 0.0.
  • the same blast search also identifies sequence similarity between MRCK1 and other proteins.
  • proteins include, but are not limited to, human Cdc42-binding protein kinase beta (Entrez accession number: NM — 006035) and C. elegans serine/threonine-protein kinase (Entrez accession number: NM — 072198).
  • kinase domain of MRCK1 has sequence homology to the catalytic domains of various protein kinases.
  • protein kinases include, but are not limited to, human myotonic dystrophy kinase (Entrez accession number: AAC14450.1 or L08835, 62% sequence identity), human dystrophia myotonica-protein (Entrez accession number: NM — 004409, 61% sequence identity), human myotonin-protein kinase, Form VI (Entrez accession number: AAA75239.1 or L00727, 62% sequence identity), and human myotonin-protein kinase, Form VIII (Entrez accession number: AAA75237.1 or L00727, 62% sequence identity).
  • sequence consisting of amino acid residues 113 to 399 of MRCK1 also aligns to the amino acid sequence having Entrez Protein Database accession number CAA73006.1 (Sbjct). These two sequences are 100% identical to each other. Sequence CAA73006.01 was disclosed in Kedra et al., Hum. Genet. 100: 611-619 (1997), and is localized to 11q13 in human chromosome 11.
  • MRCK1 gene is also localized near or at 11q13 in human chromosome 11. Specifically, the MRCK1 gene is located between genes LOC256612 and EHD1, and overlaps with gene LOC196205.
  • the MRCK1 gene encompasses nucleotides 979139 to 999235 in human chromosome 11.
  • the nucleotide numbering in human chromosome 11 is based on Entrez Human Genome Sequence Database maintained by NCBI.
  • the minus-strand sequence of human chromosome 11 that consists of nucleotides 979139 to 999235 is shown in SEQ ID NO:3.
  • the genomic sequence in SEQ ID NO:3 is listed from 5′ to 3′, i.e. from nucleotide 999235 to nucleotide 979139 in the minus strand of human chromosome 11.
  • Human MRCK1 gene has at least 35 exons. Table 2 lists the location of each of these 35 exons in the genomic sequence SEQ ID NO:3. SEQ ID NO:1 shows an MRCK1-coding sequence produced by fusing the 35 exons in consecutive order. Translation of SEQ ID NO:1 produces the amino acid sequence SEQ ID NO:2. Table 2 also illustrates the corresponding location of each exon in the MRCK1-coding sequence SEQ ID NO:1.
  • MRCK1 kinase depicted by SEQ ID NO:2 comprises multiple structural/functional domains. These structural/functional domains include at least a kinase domain (comprising amino acid residues 71 to 337), a protein kinase C terminal domain (comprising amino acid residues 339 to 398), a myosin tail domain (comprising amino acid residues 648 to 786), a DAG_PE binding domain (comprising amino acid residues 882 to 920), a Pleckstrin homology domain (comprising amino acid residues 953 to 1060), a CNH domain (comprising amino acid residues 1102 to 1345), and a P21-Rho-binding domain (comprising amino acid residues 1440 to 1471).
  • a kinase domain comprising amino acid residues 71 to 337)
  • a protein kinase C terminal domain comprising amino acid residues 339 to 398
  • FIGS. 3 , 4 , and 5 illustrate the sequence alignments between MRCK1's kinase domain and various protein kinase domains.
  • Query denotes to the sequence of MRCK1
  • Sbjct refers to the sequence being compared to the MRCK1 sequence.
  • FIG. 3 is the alignment between MRCK1's kinase domain and the consensus sequence of the catalytic domains of a subfamily of Serine/Threonine protein kinases.
  • This subfamily includes C-Jun N-terminal kinase (JNK3), abelson tyrosine kinase, a calmodulin-binding, vesicle-associated, protein kinase-like protein (1G5), serine/threonine-protein kinase prp4, Cdc2/Cdc28 subfamily of Ser/Thr protein kinases in Caenorhabditis elegans , and ribosomal S6 kinase of C elegans .
  • the consensus sequence has CD NO: smart00220.4, S_TKc, and can be retrieved from the conserveed Domain Database maintained by NCBI.
  • the alignment was performed using standard protein-protein BLAST (blastp) algorithm provided by NCBI.
  • MRCK1's kinase domain has 100% sequence identities to the consensus sequence smart00220.4, with a score of 263 bits and an E value of 3 ⁇ 10 ⁇ 71 .
  • FIG. 4 shows the alignment between MRCK1 's kinase domain and the consensus sequence of the catalytic domains of another subfamily of protein kinases.
  • This subfamily includes protein kinase Ck2, wee1-like protein kinase (WEE1hu), and tyrosine-protein kinase RYK.
  • the consensus sequence has CD NO: pfam00069.4, pkinase.
  • MRCK1's kinase domain has 100% sequence identities to pfam00069.4, with a score of 213 bits and an E value of 2 ⁇ 10 ⁇ 56 .
  • FIG. 5 shows the alignment between MRCK1's kinase domain and the catalytic domain (CD NO: smart00219.4, TyrKc) of a subfamily of tyrosine kinases.
  • This subfamily includes the tyrosine kinase domain of fibroblast growth factor receptor 1, tyrosine-protein kinase (KIN15/KIN16 subfamily), and a Drosophila receptor protein-tyrosine kinase family member (drl-P1).
  • the amino acid residues 72-303 in MRCK1's kinase domain has 89.1% sequence identities to smart00219.4, with a score of 112 bits and an E value of 9 ⁇ 10 ⁇ 26 .
  • FIG. 6 illustrates the sequence alignment between the amino acid residues 339-398 of MRCK1 and the consensus sequence for the extension to a family of Ser/Thr-type protein kinases.
  • the consensus sequence has CD NO: smart00133.4, S_TK_X.
  • This family of protein kinases includes cAMP-Dependent protein kinase, protein kinase cek1, and cell cycle protein kinase DBF2.
  • the two sequences share 95.2% sequence identities with a score of 57.7 bits and an E value of 3 ⁇ 10 ⁇ 9 .
  • the amino acid residues 339-398 of MRCK1 further aligned to a consensus sequence (CD NO: pfam00433.4, pkinase_C) of the protein kinase C terminal domain.
  • FIG. 7 shows 91% sequence identities between these two sequences. The alignment has a score of 40.6 bits and an E value of 4 ⁇ 10 ⁇ 04 .
  • Ribosomal protein S6 kinase (S6K), serine/threonine-protein kinase YPK1, and protein kinase C, zeta type (NPKC-ZETA) share the consensus sequence pfam00433.4, pkinase_C.
  • FIG. 8 demonstrates the sequence alignment between the amino acid residues 882-920 and a consensus sequence of the DAG_PE binding domains.
  • the two sequences show 78% sequence identities with a score of 51.9 bits and an E value of 2 ⁇ 10 ⁇ 07 .
  • the CD number for the consensus sequence is pfam00130.4.
  • the DAG_PE binding domain is also known as the protein kinase C conserved region 1 (C1 or cysteine-rich) domain.
  • MRCK1 shows sequence homology to the Pleckstrin homology domain (PH domain).
  • PH domains are commonly found in eukaryotic signaling proteins. The domain family possesses multiple functions including the abilities to bind inositol phosphates and various other proteins. PH domains have been found to possess inserted domains (such as in PLC gamma, syntrophins) and/or to be inserted within other domains. Mutations in Brutons tyrosine kinase (Btk) within its PH domain cause X-linked agammaglobulinaemia (XLA) in patients.
  • Btk Brutons tyrosine kinase
  • FIG. 9 shows the comparison of the amino acid residues 953-1060 of MRCK to the consensus sequence of the PH domains of a family of proteins which include Rac1 and GTPase activating protein BEM2/IPL2.
  • the consensus sequence has CD NO: smart00233.4, PH.
  • the comparison indicates 87.5% sequence identities with a score of 47.8 bits and an E value of 2 ⁇ 10 ⁇ 6 .
  • FIG. 10 depicts the comparison between the amino acid residues 954-1060 of MRCK1 and another consensus sequence of the PH domains.
  • the consensus sequence has CD NO: pfam00169.4, PH, which is shared by proteins including Still life protein type 1 (SIF type 1) and C. elegans LET-502 protein.
  • the comparison shows 88% sequence identities with a score of 42.6 bits and an E value of 9 ⁇ 10 ⁇ 5 .
  • FIG. 11 shows the comparison between the amino acid residues 1102-1345 of MRCK1 and a consensus CNH domain sequence (CD NO: pfam00780.4, CNH).
  • the alignment shows 85.7% sequence identities with a score of 60.0 bits and an E value of 6 ⁇ 10 ⁇ 10 .
  • the consensus CNH domain sequence is found in NIK1-like kinase, mouse citron (Rho-interacting, serine/threonine kinase 21), and yeast ROM1 and ROM2.
  • FIG. 12 shows the sequence alignment between the amino acid residues 1440-1475 of MRCK1 and the consensus sequence of other P21-Rho-binding domain (CD NO: smart00285, PBD).
  • the two sequences have 86.1% sequence identities with a score of 38.3 bits and an E value of 0.002.
  • P21-Rho-binding domain is a domain that binds Cdc42p- and/or Rho-like small GTPases.
  • the domain also knows as the Cdc42/Rac interactive binding domain (CRIB domain).
  • the amino acid residues 648 to 786 in MRCK1 also weakly resemble a myosin tail domain (CD NO: pfam01576).
  • the myosin molecule is a multi-subunit complex made up of two heavy chains and four light chains. It is a fundamental contractile protein found in all eukaryotic cell types.
  • the myosin tail domain consists of the coiled-coil myosin heavy chain tail region.
  • the coiled-coil is composed of the tail from two molecules of myosin. These can then assemble into the macromolecular thick filament.
  • the coiled-coil region provides the structural backbone the thick filament.
  • the alignment shows in FIG. 13 .
  • the two sequences being compared have 17.8% sequence identities with a score of 39.9 bits and an E value of 7 ⁇ 10 ⁇ 4 .
  • the MRCK1 sequence also shows high homology to PKIN20, a human kinase disclosed in PCT patent application No. WO 02/08399.
  • the two proteins share 90% sequence identities in amino acids and 95% identities at the cDNA level.
  • Hydrophobicity analysis indicates that MRCK1 kinase is not likely a membrane or transmembrane protein.
  • the hydrophobicity profile of MRCK1 is illustrated in FIG. 14 .
  • MRCK1 Two transcripts of MRCK1, a 4 kb and a 6 kb transcript, were detected in human brain, heart, skeletal muscle, colon, thymus, spleen, kidney, liver, small intestine, placenta, lung, and peripheral blood leukocyte by Multiple Tissue Northern analysis (MTN). The highest expression was in placenta while the lowest expression was in small intestine.
  • MTN Multiple Tissue Northern analysis
  • the MRCK1 expression was confirmed by an multiple tissue expression array (MTA), in which MRCK1 expression was found in all 76 tissues contained in the array.
  • Protein kinases are involved in the regulation of many critical biological processes such as signal transduction pathways. Malfunctions of cellular signaling have been associated with many diseases. Regulation of signal transduction by cytokines and association of signal molecules with protooncogenes and tumor suppressor genes have been the subjects of intense research. Many therapeutic strategies can now be developed through the synthesis of compounds which activate or inactivate protein kinases.
  • kinases are a major target for drug action and development.
  • Trials are ongoing in a wide variety of therapeutic indications including asthma, Parkinson's, inflammation, psoriasis, rheumatoid arthritis, spinal cord injuries, muscle conditions, osteoporosis, graft versus host disease, cardiovascular disorders, autoimmune disorders, retinal detachment, stroke, epilepsy, ischemia/reperfusion, breast cancer, ovarian cancer, glioblastoma, non-Hodgkin's lymphoma, colorectal cancer, non-small cell lung cancer, brain cancer, Kaposi's sarcoma, pancreatic cancer, liver cancer, and other tumors.
  • Numerous kinds of modulators of kinase activity are currently in clinical trials including antisense molecules, antibodies, small molecules, and even gene therapy. Accordingly, it is valuable to the field of pharmaceutical development to identify and characterize previously unknown members of the kinase family proteins.
  • the present invention advances the state of the art by providing novel human kinase proteins which are structurally related to MRCKs
  • kinase inhibitors are aimed at critical components in signal transduction pathways.
  • Approaches for regulating kinase gene expression include specific antisense oligonucleotides for inhibiting post-transcriptional processing of the messenger RNA, naturally, occurring products and their chemical derivatives to inhibit kinase activity and monoclonal antibodies to inhibit receptor linked kinases.
  • kinase inhibitors also allow other therapeutic agents additional time to become effective and act synergistically with current treatments.
  • the second messenger dependent protein kinases primarily mediate the effects of second messengers such as cyclic AMP (cAMP), cyclic GMP, inositol triphosphate, phosphatidylinositol, 3,4,5-triphosphate, cyclic-ADPribose, arachidonic acid, diacylglycerol and calcium-calmodulin.
  • cyclic AMP cyclic AMP
  • GMP cyclic GMP
  • inositol triphosphate phosphatidylinositol
  • cyclic-ADPribose cyclic-ADPribose
  • arachidonic acid diacylglycerol
  • calcium-calmodulin calcium-calmodulin.
  • the cyclic-AMP dependent protein kinases (PKA) are important members of the STK family. Cyclic-AMP is an intra-cellular mediator of hormone action in all prokaryotic and animal cells that have been studied.
  • Such hormone-induced cellular responses include thyroid hormone secretion, cortisol secretion, progesterone secretion, glycogen breakdown, bone resorption, and regulation of heart rate and force of heart muscle contraction.
  • PKA is found in all animal cells and is thought to account for the effects of cyclic-AMP in most of these cells. Altered PKA expression is implicated in a variety of disorders and diseases including cancer, thyroid disorders, diabetes, atherosclerosis, and cardiovascular disease.
  • CaM dependent protein kinases are also members of STK family. Calmodulin is a calcium receptor that mediates many calcium regulated processes by binding to target proteins in response to the binding of calcium. The principle target protein in these processes is CaM dependent protein kinases. CaM-kinases are involved in regulation of smooth muscle contraction (MLC kinase), glycogen breakdown (phosphorylase kinase), and neurotransmission (CaM kinase I and CaM kinase II). CaM kinase I phosphorylates a variety of substrates including the neurotransmitter related proteins synapsin I and II, the gene transcription regulator, CREB, and the cystic fibrosis conductance regulator protein, CFTR.
  • MLC kinase smooth muscle contraction
  • phosphorylase kinase glycogen breakdown
  • CaM kinase I and CaM kinase II neurotransmission
  • CaM kinase I phosphorylates a variety of substrates including the neuro
  • CaM II kinase also phosphorylates synapsin at different sites, and controls the synthesis of catecholamines in the brain through phosphorylation and activation of tyrosine hydroxylase. Many of the CaM kinases are activated by phosphorylation in addition to binding to CaM. The kinase may autophosphorylate itself, or be phosphorylated by another kinase as part of a “kinase cascade”.
  • AMPK 5′-AMP-activated protein kinase
  • Mammalian AMPK is a regulator of fatty acid and sterol synthesis through phosphorylation of the enzymes acetyl-CoA carboxylase and hydroxymethylglutaryl-CoA reductase and mediates responses of these pathways to cellular stresses such as heat shock and depletion of glucose and ATP.
  • AMPK is a heterotrimeric complex comprised of a catalytic alpha subunit and two non-catalytic beta and gamma subunits that are believed to regulate the activity of the alpha subunit.
  • Subunits of AMPK have a much wider distribution in non-lipogenic tissues, such as brain, heart, spleen, and lung, than expected. This distribution suggests that AMPK's functions may extend beyond regulation of lipid metabolism alone.
  • MAP kinases are also members of the STK family. MAP kinases also regulate intra-cellular signaling pathways. They mediate signal transduction from the cell surface to the nucleus via phosphorylation cascades. Several subgroups have been identified, and each manifests different substrate specificities and responds to distinct extracellular stimuli. MAP kinase signaling pathways are present in mammalian cells as well as in yeast.
  • the extracellular stimuli that activate mammalian pathways include epidermal growth factor (EGF), ultraviolet light, hyperosmolar medium, heat shock, endotoxic lipopolysaccharide (LPS), and pro-inflammatory cytokines, such as tumor necrosis factor (TNF) and interleukin-1 (IL-1).
  • EGF epidermal growth factor
  • UV light ultraviolet light
  • hyperosmolar medium hyperosmolar medium
  • heat shock endotoxic lipopolysaccharide
  • pro-inflammatory cytokines such as tumor necrosis factor (TNF) and interleukin-1 (IL-1).
  • TNF tumor necrosis factor
  • IL-1 interleukin-1
  • EGF receptor is found in over half of breast tumors unresponsive to hormone. EGF is found in many tumors, and EGF may be required for tumor cell growth. Antibody to EGF blocked the growth of tumor xenografts in mice. An antisense oligonucleotide for amphiregulin inhibited growth of a pancreatic cancer cell line.
  • Tamoxifen a protein kinase C inhibitor with anti-estrogen activity
  • Tamoxifen a protein kinase C inhibitor with anti-estrogen activity
  • the use of this compound may increase the risk of developing cancer in other tissues such as the endometrium.
  • Raloxifene a related compound, has been shown to protect against osteoporosis. The tissue specificity of inhibitors must be considered when identifying therapeutic targets.
  • MAP kinases are a family of protein serine/threonine kinases which mediate signal transduction from extracellular receptors or heat shock, or UV radiation.
  • Cell proliferation and differentiation in normal cells are under the regulation and control of multiple MAP kinase cascades. Aberrant and deregulated functioning of MAP kinases can initiate and support carcinogenesis.
  • Insulin and IGF-1 also activate a mitogenic MAP kinase pathway that may be important in acquired insulin resistance occurring in type 2 diabetes.
  • PI-3 kinase/Akt signaling is equally important in diabetes.
  • the pathway activated by RTKs subsequently regulates glycogen synthase 3 (GSK3) and glucose uptake. Since AKT has decreased activity in type 2 diabetes, it provides a therapeutic target.
  • Protein kinase inhibitors provide much of our knowledge about in vivo regulation and coordination of physiological functions of endogenous peptide inhibitors.
  • a pseudosubstrate sequence within PKC acts to inhibit the kinase in the absence of its lipid activator.
  • a PKC inhibitor such as chelerythrine, acts on the catalytic domain to block substrate interaction, while calphostin acts on the regulatory domain to mimic the pseudosubstrate sequence and block ATPase activity, or to inhibit cofactor binding.
  • protein kinases Although some protein kinases have, to date, no known system of physiological regulation, many are activated or inactivated by autophosphorylation or phosphorylation by upstream protein kinases.
  • the regulation of protein kinases also occurs during the transcription, post-transcription, and post-translation processes.
  • the mechanism of post-transcriptional regulation is alternative splicing of precursor mRNA.
  • protein kinase C ⁇ I and ⁇ II are two isoforms of a single PKC ⁇ gene derived from differences in the splicing of the exon encoding the C-terminal 50-52 amino acids. Splicing can be regulated by a kinase cascade in response to peptide hormones, such as insulin and IGF-1.
  • PKC ⁇ I and ⁇ II have different specificities for phosphorylating members of the mitogen activated protein (MAP) kinase family, for glycogen synthase 3 ⁇ , for nuclear transcription factors, such as TLS/Fus, and for other nuclear kinases.
  • MAP mitogen activated protein
  • Antisense oligonucleotides are short lengths of synthetically manufactured, chemically modified DNA or RNA designed to specifically interact with mRNA transcripts encoding target proteins.
  • the interaction of the antisense moiety with mRNA inhibits protein translation and, in some cases, post-transcriptional processing (e.g., alternative splicing and stability) of mRNA.
  • Antisense oligonucleotides have been developed to alter alternative splicing of mRNA forms for inhibiting the translation of PKC ⁇ .
  • Protein kinase C isoforms have been implicated in cellular changes observed in the vascular complications of diabetes. Hyperglycemia is associated with increased levels of PKC ⁇ and ⁇ isoforms in renal glomeruli of diabetic rats. Oral administration of a PKC ⁇ inhibitor prevented the increased mRNA expression of TGF- ⁇ 1 and extracellular matrix component genes. Administration of the specific PKC ⁇ inhibitor (LY333531) also normalized levels of cytokines, caldesmon, and hemodynamics of retinal and renal blood flow. Overexpression of the PKC ⁇ isoform in the myocardium resulted in cardiac hypertrophy and failure. The use of LY333531 to prevent adverse effects of cardiac PKC ⁇ overexpression in diabetic subjects is under investigation. The compound is also in Phase I/II clinical trials for diabetic retinopathy and diabetic macular edema indicating that it may be pharmacodynamically active.
  • PRK proliferation-related kinase
  • PRK is a serum/cytokine inducible STK that is involved in regulation of the cell cycle and cell proliferation in human megakaroytic cells.
  • PRK is related to the polo (derived from humans polo gene) family of STKs implicated in cell division.
  • PRK is down-regulated in lung tumor tissue and may be a proto-oncogene whose deregulated expression in normal tissue leads to oncogenic transformation.
  • Altered MAP kinase expression is implicated in a variety of disease conditions including cancer, inflammation, immune disorders, and disorders affecting growth and development.
  • DNA-dependent protein kinase is involved in the repair of double-strand breaks in mammalian cells.
  • This enzyme requires ends of double-stranded DNA or transitions from single-stranded to double-stranded DNA in order to act as a serine/threonine kinase.
  • Cells with defective or deficient DNA-PK activity are unable to repair radiation induced DNA double-strand breaks and are consequently very sensitive to the lethal effects of ionizing radiation.
  • Inhibition of DNA-PK has the potential to increase the efficacy of anti-tumor treatment with radiation or chemotherapeutic agents.
  • CDKs The cyclin-dependent protein kinases
  • Cyclins are small regulatory proteins that act by binding to and activating CDKs that then trigger various phases of the cell cycle by phosphorylating and activating selected proteins involved in the mitotic process.
  • CDKs are unique in that they require multiple inputs to become activated. In addition to the binding of cyclin, CDK activation requires the phosphorylation of a specific threonine residue and the dephosphorylation of a specific tyrosine residue.
  • CDKs Cellular inhibitors of CDKs also play a major role in cell cycle progression. Alterations in the expression, function, and structure of cyclin and CDK are encountered in the cancer phenotype. Therefore CDKs may be important targets for new cancer therapeutic agents.
  • Chemotherapy resistant cells tend to escape apoptosis. Under certain circumstances, inappropriate CDK activation may even promote apoptosis by encouraging the progression of the cell cycle under unfavorable conditions, i.e., attempting mitosis while DNA damage is largely unrepaired.
  • Flavopiridol is a flavonoid that causes 50% growth inhibition of tumor cells at 60 nM. It also inhibits EGFR and protein kinase A. Flavopiridel induces apoptosis and inhibits lymphoid, myeloid, colon, and prostate cancer cells grown in vivo as tumor xenografts in nude mice.
  • Staurosporine is toxic, but its derivative 7-hydroxystaurosporine (UCN-01) has anti-tumor properties and is in clinical trials.
  • UCN-01 affects the phosphorylation of CDKs and alters the cell cycle checkpoint functioning.
  • Protein tyrosine kinases specifically phosphorylate tyrosine residues on their target proteins and may be divided into transmembrane, receptor PTKs and non-transmembrane, non-receptor PTKs.
  • Transmembrane protein tyrosine kinases are receptors for most growth factors. Binding of a growth factor to the receptor activates the transfer of a phosphate group from ATP to selected tyrosine side chains of the receptor and other specific proteins.
  • Growth factors (GF) associated with receptor PTKs include; epidermal GF, platelet-derived GF, fibroblast GF, hepatocyte GF, insulin and insulin-like GFs, nerve GF, vascular endothelial GF, and macrophage colony stimulating factor.
  • RTKs stimulate tumor cell proliferation
  • Inhibitors of RTKs are also useful in preventing tumor angiogenesis and can eliminate support from the host tissue by targeting RTKs located on vascular cells, such as blood vessel endothelial cells and stromal fibroblasts.
  • vascular cells such as blood vessel endothelial cells and stromal fibroblasts.
  • VEGF stimulates endothelial cell growth during angiogenesis, and increases the permeability of tumor vasculature so that proteins and other growth factors become accessible to the tumor.
  • Broad-spectrum antitumor efficacy of an oral dosage form of an inhibitor of VEGF signaling has been reported.
  • inhibition of VEGF receptor signaling presents an important therapeutic target.
  • An extracellular receptor can also be a target for inhibition.
  • the EGF receptor family and its ligands are overexpressed and exist as an autocrine loop in many tumor types.
  • RTKs Increasing knowledge of the structure and activation mechanism of RTKs and the signaling pathways controlled by tyrosine kinases provided the possibility for the development of target specific drugs and new anti-cancer therapies.
  • Approaches towards the prevention or interception of deregulated RTK signaling include the development of selective components that target either the extracellular ligand-binding domain or the intra-cellular substrate binding region.
  • mAbs monoclonal antibodies
  • RTKs reactive hypothalamic hormones
  • recombinant antibody technology has made an enormous progress in the design, selection and production of newly engineered antibodies. It is also possible to generate humanized antibodies, human-mouse chimeric or bispecific antibodies for targeted cancer therapy.
  • anti-RTK mAbs might work by blocking the ligand-receptor interaction and therefore inhibiting ligand-induced RTK signaling and increasing RTK down-regulation and internalization.
  • binding of mAbs to certain epitopes on the cancer cells may induce immune-mediated responses, such as opsonization and complement-mediated lysis, and trigger antibody-dependent cellular cytotoxicity by macrophages or natural killer cells.
  • immune-mediated responses such as opsonization and complement-mediated lysis
  • mAbs control tumor growth by altering the intra-cellular signaling pattern inside the targeted tumor cell, leading to growth inhibition and/or apoptosis.
  • bispecific antibodies can bridge selected surface molecules on a target cell with receptors on an effector cell, thus triggering cytotoxic responses against the target cell.
  • advances in antibody engineering, characterization of tumor antigens and immunology might help to produce rationally designed bispecific antibodies for anti-cancer therapy.
  • Another promising approach to inhibiting aberrant RTK signaling is to develop small molecule drugs that selectively interfere with the intrinsic tyrosine kinase activity and thereby block receptor autophosphorylation and activation of downstream signal transducers.
  • the tyrphostins which belong to the quinazolines, are one important group of such inhibitors that compete with ATP for the ATP binding site at the receptor's tyrosine kinase domain and some members of the group have been shown to specifically inhibit the EGFR.
  • Potent and selective inhibitors of receptors involved in neovascularization have been developed and are now undergoing clinical evaluation.
  • TKIs tyrosine kinase inhibitors
  • Recombinant immunotoxins provide another possibility of target-selective drug design.
  • Recombinant immunotoxins are composed of a bacterial or plant toxin either fused or chemically conjugated to a specific ligand, such as the variable domains of the heavy and light chains of mAbs or to a growth factor.
  • Immunotoxins may contain bacterial toxins, such as Pseudomouas exotoxin A or diphtheria toxin, or plant toxins, such as ricin A or clavin. These recombinant molecules can selectively kill their target cells when internalized after binding to cell surface receptors of the target cells.
  • antisense oligonucleotides represent another strategy to inhibit the activation of RTKs.
  • Antisense oligonucleotides are short pieces of synthetic DNA or RNA that are designed to interact with the mRNA to block the transcription and thus the expression of the target proteins.
  • Antisense oligonucleotides interact with the mRNA by Watson-Crick base-pairing and are therefore highly specific to the target protein.
  • Several preclinical and clinical studies suggest that antisense therapy might be therapeutically useful for the treatment of solid tumors.
  • RTKs and their relevant signaling as selective anti-cancer targets for therapeutic intervention has been recognized.
  • mAbs and RTK inhibitors have been developed and are currently being evaluated in clinical trials.
  • Table 3 summarizes the most successful drugs against receptor tyrosine kinase signaling which are currently evaluated in clinical phases or have already been approved by the FDA.
  • Non-receptor PTKs lack transmembrane regions and, instead, form complexes with the intra-cellular regions of cell surface receptors.
  • Receptors that function through non-receptor PTKs include those for cytokines, hormones (growth hormone and prolactin) and antigen-specific receptors on T and B lymphocytes.
  • PTKs were first identified as the products of mutant oncogenes in cancer cells where their activation was no longer subject to normal cellular controls. In fact, about one third of the known oncogenes encode PTKs, and it is well-known that cellular transformation (oncogenesis) is often accompanied by increased tyrosine phosphorylation activity.
  • tyrosine kinase inhibitors such as flavopiridol, geissem, erbstatin, lavendustin A, staurosporine, and UCN-01, are derived from natural products. Inhibitors directed to the ATP binding site are also available. Signals from RTKs can also be inhibited at other target sites such as nuclear tyrosine kinases, membrane anchors (inhibition of farnesylation) and transcription factors.
  • tyrosine kinases such as EGFR, HER2, PDGFR, src, and abl
  • IGF-1 and TRK will interfere with tumor cell survival. Inhibition of these kinases will lead to tumor shrinkage and apoptosis.
  • FklI/KDR and src are kinases necessary for neovascularization (angiogenesis) of tumors. Inhibition of these kinases will slow tumor growth and decrease metastases.
  • Inhibitors of RTKs suppress tumor development by preventing cell migration, invasion and metastases. These drugs are likely to increase the time required for tumor progression, and may inhibit or attenuate the aggressiveness of the disease but may not initially result in measurable tumor regression.
  • ALK positive lymphomas a class of ALK positive lymphomas referred to as “AL Komas” which display inappropriate expression of a neural-specific tyrosine kinase, anaplastic lymphoma kinase (ALK).
  • Iressa ZD1839 is an orally active selective EGF-R inhibitor. This compound disrupts signaling involved in cancer cell proliferation. The clinical efficacy of this agent shows that it is well tolerated by patients undergoing Phase I/II clinical trials. The compound has shown promising cytotoxicity towards several cancer cell lines.
  • neuron-specific kinases are well established as targets for the development of pharmacologically active modulators.
  • kinase proteins are a major target for drug action and development. Accordingly, it is valuable to the field of pharmaceutical development to identify and characterize previously unknown members of kinase proteins.
  • the present invention advances the state of the art by providing a previously unidentified human kinase protein that has homology to rat MRCK.
  • MRCKs are serine/threonine kinases with multiple functional domains. MRCKs are thought to act as a downstream effector of Cdc42 in cytoskeletal reorganization.
  • Cdc42 is an essential GTPase that belongs to the Rho/Rac subfamily of Ras-like GTPases. These proteins act as molecular switches by responding to exogenous and/or endogenous signals and relaying those signals to activate downstream components of a biological pathway.
  • the 11 current members of the Cdc42 family display between 75 and 100% amino acid identity and are functional, as well as structural, homologs.
  • Cdc42 transduces signals to the actin cytoskeleton to initiate and maintain polarized growth and to mitogen-activated protein morphogenesis.
  • Cdc 42 plays an important role in multiple actin-dependent morphogenetic events such as bud emergence, mating-projection formation, and pseudohyphal growth.
  • Cdc42 regulates a variety of actin-dependent events and induces the JNK/SAPK protein kinase cascade, which leads to the activation of transcription factors within the nucleus.
  • Cdc42 mediates these processes through interactions with a myriad of downstream effectors.
  • Cdc42 has been implicated in a number of human diseases through interactions with its regulators and downstream effectors.
  • the MRCK family includes at least two related protein kinases: MRCK alpha and MRCK beta, which were isolated from a human brain cDNA library using a monoclonal antibody directed against myotonic dystrophy protein kinase (DMPK).
  • DMPK myotonic dystrophy protein kinase
  • the epitope shared by DMPK and MRCKs was located at the catalytic site of DMPK using a phage-displayed random peptide library (Lam L T, Hum Mol Genet. 9:2167-2173, 2000).
  • MRCKs are involved in Cdc42-mediated myosin light chain phosphorylation (Dong et al., Eur J Cell Biol 81:231-42, 2002). Specifically, MRCK alpha was implicated in Cdc42-mediated peripheral actin formation and neurite outgrowth in HeLa and PC12 cells, respectively. It was suggest that MRCK alpha may act as a downstream effector of Cdc42 in cytoskeletal reorganization (Leung et al., Mol Cell Biol 18:130-40, 1998).
  • the native MRCK exists in high-molecular-weight complexes.
  • the three independent coiled-coil (CC) domains and the N-terminal region preceding the kinase domain are responsible for intermolecular interactions leading to MRCK alpha multimerization.
  • N-terminus-mediated dimerization and consequent trans-autophosphorylation are critical processes regulating MRCK alpha catalytic activities.
  • a region containing the two distal CC domains (CC2 and CC3; residues 658 to 930) was found to interact intramolecularly with the kinase domain and negatively regulates its activity. Its deletion also resulted in an active kinase, confirming a negative auto-regulatory role.
  • MRCK and the negative kinase-distal CC domain interaction are two mutually exclusive events that tightly regulate the catalytic state of the kinase. Disruption of this interaction by a mutant kinase domain resulted in increased kinase activity.
  • MRCK kinase activity was also elevated when cells were treated with phorbol ester, which can interact directly with a cysteine-rich domain next to the distal CC domain. It therefore appears that binding of phorbol ester to MRCK releases its auto-inhibition, allowing N-terminal dimerization and subsequent kinase activation. (Tan et al., Mol. Cell. Biol., 21:2767-78, 2001)
  • the approximately 190-kD MRCK kinases preferentially phosphorylate non-muscle myosin light chain at serine 19, which is known to be crucial for activating actin-myosin contractility.
  • the p21-binding domain binds GTP-Cdc42 but not GDP-Cdc42.
  • the multidomain structure includes a cysteine-rich motif resembling those of protein kinase C and n-chimaerin and a putative Pleckstrin homology domain.
  • MRCK alpha and Cdc42 V12 co-localize, particularly at the cell periphery in transfected HeLa cells.
  • P130 and p85 Two major substrates, p130 and p85, for MRCK alpha-kinase have been identified.
  • P130 is identified as the myosin binding subunit p130, whereas p85 is a novel related protein.
  • P85 contains N-terminal ankyrin repeats, an alpha-helical C terminus with leucine repeats, and a centrally located conserved motif with the MRCK alpha kinase phosphorylation site.
  • p85 is specifically associated with protein phosphatase 1delta (PP1delta), and this requires the N-terminus, including the ankyrin repeats. This association is required for the regulation of both the catalytic activities and the assembly of actin cytoskeleton.
  • the N-terminus, in association with PP1delta, is essential for actin depolymerization, whereas the C-terminus antagonizes this action.
  • the C-terminal effects consist of two independent events that involved both a conserved phosphorylation inhibitory motif and an alpha-helical leucine repeats. The former was able to interact with PP1delta only in the phosphorylated state and result in inactivation of PP1delta activity. This provides further evidence that phosphorylation of a myosin binding subunit protein by specific kinases confers conformational changes in a highly conserved region that plays an essential role in the regulation of its catalytic subunit activities (Tan et al., J. Biol. Chem., 276:21209-16, 2001).
  • MRCKs appear to be an important player in cytoskeletal reorganization, neuronal differentiation, and myotonic dystrophy. MRCKs may serve as a major target for drug action and development.
  • the present invention provides a new human kinase (MRCK1) which has sequence and structure similarities to rat MRCK alpha and other kinases.
  • the multiple domains in MRCK1 share high sequence identities with the corresponding domains in other kinases. Each of these domains, either in its native form or in a mutant form, can be used to affect the function of the corresponding domain in other kinases.
  • the kinase domain in MRCK1 can be used to phosphorylate suitable substrates, including p130 and p85 or substrate peptides containing MRCK alpha phosphorylation sites.
  • the substrate peptides can be conjugated to antibodies, and the phosphate groups added to the substrate peptides can be radioactively or fluorescently labeled. Antibodies thus labeled can be used in various detection assays, as appreciated by one of skilled in the art.
  • the MRCK1 gene and gene products can be used as molecular markers for diagnosing, prognosing, and monitoring the treatment of disorders related to the aberrant expression of MRCK1.
  • the MRCK1 gene can be used to screen for potential agents or drugs capable of enhancing or inhibiting the MRCK1 gene expression in human cells.
  • the MRCK1 gene products (polynucleotide and polypeptide) can be used to screen for potential agents or drugs capable of enhancing or inhibiting MRCK1 activity.
  • various therapeutic methods for treating disorders related to the aberrant expression of MRCK1 can be designed based on the MRCK1 gene, its variants, or the agents/drugs that affect the expression of the MRCK1 gene or the activity of the MRCK1 gene products.
  • One aspect of the invention pertains to isolated polynucleotide probes capable of hybridizing to the MRCK1 gene or its transcripts, such as MRCK1 mRNAs. These probes can be used to detect the expression level of the MRCK1 gene in human tissue or cells.
  • the present invention also contemplates polynucleotide fragments for use as PCR primers for the amplification or mutation of the MRCK1 gene or the MRCK1 kinase-coding sequences.
  • Another aspect of the invention pertains to isolated polynucleotides that encode MRCK1, or a fragment or mutant thereof. These polynucleotides can be used for expressing MRCK1, or a fragment or mutant thereof.
  • the protein products thus expressed can be used to screen for agents/drugs that modulate an activity of MRCK1.
  • these polynucleotides can be used to designing gene therapy vectors which target the expression of the MRCK1 gene or an activity of MRCK1 in humans.
  • a polynucleotide comprising SEQ ID NO:1 or SEQ ID NO:3 can be prepared using standard molecular biology techniques as appreciated by one of ordinary skill in the art. For instance, primers derived from the 5′ and 3′ ends of SEQ ID NO:1 can be used to amplify mRNAs isolated from human tissues. The cDNA thus produced contains SEQ ID NO:1. Likewise, primers for amplifying the human genomic sequence containing SEQ ID NO:3 can be designed and used to prepare the genomic sequence of the MRCK1 gene. A variant (such as a homolog) or a fragment of SEQ ID NO:1 or SEQ ID NO:3 can be similarly prepared.
  • probes can be designed to screen for cDNA or genomic sequence libraries in order to identify polynucleotide molecules comprising the full-length or fragments of SEQ ID NO:1 or SEQ ID NO:3.
  • the molecules thus identified can be used to create suitable vectors comprising the full-length SEQ ID NO:1 or SEQ ID NO:3.
  • Polynucleotides capable of hybridizing to the MRCK1 gene can be prepared by standard synthetic techniques, e.g., using an automated DNA synthesizer.
  • the polynucleotide probes can hybridize to the MRCK1 gene under reduced stringent conditions, stringent conditions, or highly stringent conditions.
  • the polynucleotides comprise at least 15, 20, 25, 30, 50, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, 900, 950, 1000 or more consecutive nucleotides of SEQ ID NO:1. Any fragments of SEQ ID NO:1 and SEQ ID NO:3 may be used as hybridization probes or PCR primers for the MRCK1 gene or its transcripts.
  • the probes/primers can be substantially purified.
  • the hybridization probes for the MRCK1 gene comprise a label group.
  • the label group can be a radioisotope, a fluorescent compound, an enzyme, or an enzyme co-factor. Probes thus labeled can be used as part of a diagnostic kit for determining the expression level of the MRCK1 gene in human tissues.
  • This invention encompasses human MRCK1 gene homologs in other species. These homologs can be determined by search different sequence databases, such as the Entrez/GenBank sequence databases maintained by the NCBI.
  • the invention also encompasses polynucleotide molecules which are structurally different from the molecules described above, but have the substantially same properties as the molecules described above. Such molecules include allelic variants, which will be described below in greater detail.
  • DNA sequence polymorphism in human MRCK1 gene exists among different individuals due to natural allelic variations.
  • An allele is one of a group of genes which occur alternatively at a given genetic locus.
  • DNA polymorphisms that affect the RNA expression level of the MRCK1 gene can also exist, e.g. through affecting the regulation or degradation of expression of the gene.
  • the present invention contemplates all allelic variants of human MRCK1 gene. Allelic variants and other homologs of the MRCK1 gene can be isolated using probes/primers derived from SEQ ID NO:1 or SEQ ID NO:3.
  • SEQ ID NO:1 and SEQ ID NO:3 can be modified.
  • the modified polynucleotides can comprise one or more mutations. These mutations can be substitutions, additions or deletions of 1, 2, 3, 5, 10, 15, 20 or more nucleotide residues in SEQ ID NO:1 or SEQ ID NO:3. Standard techniques can be used, such as site-directed mutagenesis or PCR-mediated mutagenesis. Preferably, these mutations create conservative amino acid substitutions. Alternatively, mutations can be introduced randomly along all or part of the MRCK1 gene or its cDNA, such as by saturation mutagenesis. Following mutagenesis, the encoded proteins can be expressed recombinantly and their activities can be determined.
  • nucleotide substitutions leading to amino acid substitutions at “non-essential” amino acid residues can be introduced.
  • a “non-essential” amino acid residue is a residue that can be altered without changing the biological activity of the protein.
  • an “essential” amino acid residue is required for the biological activity of the protein. Amino acid residues that are conserved among allelic variants or homologs of the MRCK1 gene from different species preferably are not changed in the present invention.
  • MRCK1 proteins that contain changes in amino acid residues that are not essential for the biological activity of MRCK1. These proteins differ in amino acid sequence from the original human MRCK1 kinase, but retain its biological activity.
  • the modified protein comprises an amino acid sequence at least about 91%, 95%, 98%, 99% or more homologous to SEQ ID NO:2.
  • MRCK1 proteins contain mutations in amino acid residues which result in inhibition of MRCK1 activity. These mutated MRCK1 proteins can be used to inhibit MRCK1 activity in patients with disorders related to the aberrant expression of MRCK1.
  • a polynucleotide of this invention can be further modified to increase its stability in vivo. Possible modifications include, but are not limited to, the addition of flanking sequences at the 5′ and/or 3′ ends; the use of phosphorothioate or 2-o-methyl rather than phosphodiester linkages in the backbone; and/or the inclusion of nontraditional bases such as inosine, queosine and wybutosine, as well as acetyl- methyl-, thio- and other modified forms of adenine, cytidine, guanine, thymine and uridine.
  • Polynucleotide molecules which are antisense to the MRCK1 gene can be prepared.
  • An “antisense” polynucleotide comprises a nucleotide sequence which is complementary to a “sense” polynucleotide which encodes a protein.
  • An antisense polynucleotide can bind via hydrogen bonds to the sense polynucleotide.
  • Antisense polynucleotides of the invention can be designed according to the rules of Watson and Crick base pairing.
  • the antisense polynucleotide molecule can be complementary to the entire coding region or part of the coding region of the MRCK1 gene.
  • the antisense polynucleotide molecule can also be complementary to a “noncoding region” in the coding strand of the MRCK1 gene.
  • the antisense polynucleotide is an oligonucleotide which is antisense to only a portion of the MRCK1 gene.
  • An antisense polynucleotide can be, for example, about 5, 10, 15, 20, 25, 30, 35, 40, 45 or 50 nucleotides in length.
  • an antisense polynucleotide of the invention can be constructed using chemical synthesis and enzymatic ligation reactions as appreciated by one of ordinary skill in the art.
  • an antisense polynucleotide 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 polynucleotides.
  • modified nucleotides which can be used to generate the antisense polynucleotide include 5-fluorouracil, 5-bromouracil, 5-chlorouracil, 5-iodouracil, hypoxanthine, xanthine, 4-acetylcytosine, 5-(carboxyhydroxymethyl)uracil, 5-carboxymethylaminomethyl-2-thiouridine, 5-carboxymethylaminomethyluracil, dihydrouracil, beta-D-galactosylqueosine, inosine, N6-isopentenyladenine, 1-methylguanine, 1-methylinosine, 2,2-dimethylguanine, 2-methyladenine, 2-methylguanine, 3-methylcytosine, 5-methylcytosine, N6-adenine, 7-methylguanine, 5-methylaminomethyluracil, 5-methoxyaminomethyl-2-thiouracil, beta-D-mannosylqueosine, 5′-methoxy
  • the antisense polynucleotide can be produced biologically using an expression vector into which a polynucleotide has been subcloned in an antisense orientation (i.e., RNA transcribed from the inserted polynucleotide will be of an antisense orientation to the target polynucleotide of interest).
  • the antisense polynucleotides of the invention can be administered to a subject or applied in situ such that they hybridize or bind to cellular mRNAs and/or genomic DNA's that encode MRCK1 kinase, thereby inhibiting the expression of MRCK1 kinase.
  • the hybridization can result in a stable duplex via conventional nucleotide complementarity.
  • An example route for administering antisense polynucleotides includes direct injection at a tissue site.
  • Antisense polynucleotides can also be modified first, 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.
  • Suitable modifications include linking the antisense polynucleotides to peptides or antibodies which bind to the cell surface receptors or antigens.
  • the antisense polynucleotides can be delivered to cells using vectors.
  • strong pol II or pol III promoters may be used in the vectors.
  • the antisense polynucleotides are ⁇ -anomeric polynucleotides.
  • An ⁇ -anomeric polynucleotide molecule forms specific double-stranded hybrid with a complementary RNA in which, contrary to the usual ⁇ -units, the strands run parallel to each other.
  • the antisense polynucleotide molecule can also comprise a 2-o-methylribonucleotide or a chimeric RNA-DNA analog.
  • the antisense polynucleotide is a ribozyme.
  • Ribozymes are catalytic RNA molecules with ribonuclease activity which are capable of cleaving a single-stranded polynucleotide, such as an mRNA, to which they have a complementary region.
  • ribozymes e.g., hammerhead ribozymes described in Haselhoif and Gerlach Nature 334:585-591, 1988
  • a ribozyme having specificity for the MRCK1 gene or its transcripts can be designed based upon SEQ ID NO:1 or 3. mRNAs transcribed from the MRCK1 gene can be used to select from a pool of RNA molecules a catalytic RNA having a specific ribonuclease activity.
  • the expression of the MRCK1 gene can be inhibited by using nucleotide sequences complementary to the regulatory region (e.g., the promoter and/or enhancers). These nucleotide sequences can form triple helical structures that prevent transcription of the gene in the target cells.
  • RNAi is a phenomenon in which the introduction of double-stranded RNA (dsRNA) into certain organisms or cell types causes degradation of the homologous mRNA.
  • dsRNA double-stranded RNA
  • RNAi has since been found to operate in a wide range of organisms. For example, in mammalian cells, introduction of long dsRNA (>30 nucleotides) can initiate a potent antiviral response, exemplified by nonspecific inhibition of protein synthesis and RNA degradation.
  • RNA interference provides a mechanism of gene silencing at the mRNA level.
  • RNAi has become an endogenous and potent gene-specific silencing technique that uses double-stranded RNAs (dsRNA) to mark a particular transcript for degradation in vivo. It also offers an efficient and broadly applicable approach for gene knock-out.
  • RNAi technology can be used for therapeutic purposes. For example, RNAi targeting Fas-mediated apoptosis has been shown to protect mice from fulminant hepatitis.
  • RNAi technology has been disclosed in numerous publications, such as U.S. Pat. Nos. 5,919,619, 6,506,559 and PCT Publication Nos. WO99/14346, WO01/70949, WO01/36646, WO00/63364, WO00/44895, WO01/75164, WO01/92513, WO01/68836 and WO01/29058.
  • a sequence capable of inhibiting gene expression by RNA interference can have any length.
  • the sequence can have at least 10, 15, 20, 25, 30, 35, 40, 45, 50, 100 or more consecutive nucleotides.
  • the sequence can be dsRNA or any other type of polynucleotide, provided that the sequence can form a functional silencing complex to degrade the target mRNA transcript.
  • the sequence comprises or consists of a short interfering RNAs (siRNA).
  • siRNA can be dsRNA having 19-25 nucleotides.
  • siRNAs can be produced endogenously by degradation of longer dsRNA molecules by an RNase III-related nuclease called Dicer.
  • siRNAs can also be introduced into a cell exogenously, or by transcription of an expression construct. Once formed, the siRNAs assemble with protein components into endoribonuclease-containing complexes known as RNA-induced silencing complexes (RISCs).
  • RISCs RNA-induced silencing complexes
  • siRNAs can be synthesized in vitro and introduced into cells to transiently suppress gene expression.
  • Synthetic siRNA provides an easy and efficient way to achieve RNAi.
  • siRNA are duplexes of short mixed oligonucleotides which can include, for example, 19 RNAs nucleotides with symmetric dinucleotide 3′ overhangs.
  • synthetic 21 bp siRNA duplexes e.g., 19 RNA bases followed by a UU or dTdT 3′ overhang
  • sequence specific gene silencing can be achieved in mammalian cells.
  • These siRNAs can specifically suppress targeted gene translation in mammalian cells without activation of DNA-dependent protein kinase (PKR) by longer dsRNA, which may result in non-specific repression of translation of many proteins.
  • PLR DNA-dependent protein kinase
  • siRNAs can be expressed in vivo from vectors. This approach can be used to stably express siRNAs in cells or transgenic animals.
  • siRNA expression vectors are engineered to drive siRNA transcription from polymerase III (pol III) transcription units.
  • Pol III transcription units are suitable for hairpin siRNA expression, since they deploy a short AT rich transcription termination site that leads to the addition of 2 bp overhangs (e.g., UU) to hairpin siRNAs—a feature that is helpful for siRNA function.
  • the Pol III expression vectors can also be used to create transgenic mice that express siRNA.
  • siRNAs can be expressed in a tissue-specific manner.
  • long double-stranded RNAs dsRNAs
  • a promoter such as CMV (pol II)
  • the long dsRNAs are processed into siRNAs in the nuclei (e.g., by Dicer).
  • the siRNAs exit from the nuclei and mediate gene-specific silencing.
  • tissue-specific (pol II) promoters can be used in conjunction with tissue-specific (pol II) promoters to create tissue-specific knockdown mice.
  • Any 3′ dinucleotide overhang such as UU, can be used for siRNA design.
  • G residues in the overhang are avoided because of the potential for the siRNA to be cleaved by RNase at single-stranded G residues.
  • siRNAs with 30-50% GC content can be more active than those with a higher G/C content in certain cases.
  • a 4-6 nucleotide poly(T) tract may act as a termination signal for RNA pol III, stretches of ⁇ 4 Ts or As in the target sequence may be avoided in certain cases when designing sequences to be expressed from an RNA pol III promoter.
  • some regions of mRNA may be either highly structured or bound by regulatory proteins.
  • the potential target sites can be compared to the appropriate genome database (human, mouse, rat, etc.). Any target sequences with more than 16-17 contiguous base pairs of homology to other coding sequences may be eliminated from consideration in certain cases.
  • siRNA can be designed to have two inverted repeats separated by a short spacer sequence and end with a string of Ts that serve as a transcription termination site. This design produces an RNA transcript that is predicted to fold into a short hairpin siRNA.
  • the selection of siRNA target sequence, the length of the inverted repeats that encode the stem of a putative hairpin, the order of the inverted repeats, the length and composition of the spacer sequence that encodes the loop of the hairpin, and the presence or absence of 5′-overhangs, can vary to achieve desirable results.
  • the siRNA targets can be selected by scanning an mRNA sequence for AA dinucleotides and recording the 19 nucleotides immediately downstream of the AA. Other methods can also been used to select the siRNA targets. In one example, the selection of the siRNA target sequence is purely empirically determined (see e.g., Sui et al., Proc. Natl. Acad. Sci. USA 99: 5515-5520, 2002), as long as the target sequence starts with GG and does not share significant sequence homology with other genes as analyzed by BLAST search. In another example, a more elaborate method is employed to select the siRNA target sequences. This procedure exploits an observation that any accessible site in endogenous mRNA can be targeted for degradation by synthetic oligodeoxyribonucleotide/RNase H method (Lee et al., Nature Biotechnology 20:500-505, 2002).
  • the hairpin siRNA expression cassette is constructed to contain the sense strand of the target, followed by a short spacer, the antisense strand of the target, and 5-6 Ts as transcription terminator.
  • the order of the sense and antisense strands within the siRNA expression constructs can be altered without affecting the gene silencing activities of the hairpin siRNA. In certain instances, the reversal of the order may cause partial reduction in gene silencing activities.
  • the length of nucleotide sequence being used as the stem of siRNA expression cassette can range, for instance, from 19 to 29.
  • the loop size can range from 3 to 23 nucleotides. Other lengths and/or loop sizes can also be used.
  • a 5′ overhang in the hairpin siRNA construct can be used, provided that the hairpin siRNA is functional in gene silencing.
  • the 5′ overhang includes about 6 nucleotide residues.
  • the target sequence for RNAi is a 21-mer sequence fragment selected from SEQ ID NO:1.
  • the 5′ end of the target sequence has dinucleotide “NA,” where “N” can be any base and “A” represents adenine.
  • the remaining 19-mer sequence has a GC content of between 35% and 55%.
  • the remaining 19-mer sequence does not include any four consecutive A or T (i.e., AAAA or TTTT), three consecutive G or C (i.e., GGG or CCC), or seven “GC” in a role.
  • Exemplary RNAi target sequences identified according to the above-described criteria (“relaxed” criteria) are illustrated in Table 4.
  • the siRNA sequences for each target sequence (listed in the same row as the target sequence and including the sense strand and the antisense strand) are also indicated in Table 4.
  • RNAi target sequences can be selected from additional criteria. For instance, the GC content of the remaining 19-mer sequence can be limited to between 45% and 55%. Moreover, any 19-mer sequence having three consecutive identical bases (i.e., GGG, CCC, TTT, or AAA) or a palindrome sequence with 5 or more bases is excluded. Furthermore, the remaining 19-mer sequence can be selected to have low sequence homology to other human genes.
  • potential target sequences are searched by BLASTN against NCBI's human UniGene cluster sequence database.
  • the human UniGene database contains non-redundant sets of gene-oriented clusters. Each UniGene cluster includes sequences that represent a unique gene.
  • 19-mer sequences producing no hit to other human genes under the BLASTN search can be selected.
  • the e-value may be set at a stringent value (such as “1”).
  • Exemplary target sequences derived using these additional conditions (“stringent” criteria) are shown in Table 5.
  • the siRNA sequences for each target sequence (listed in the same row as the target sequence and including the sense strand and the antisense strand) are also indicated in Table 5.
  • an siRNA sequence of the present invention can be introduced into a cell that expresses the MRCK1 gene.
  • the polypeptide or mRNA level of the MRCK1 gene in the cell can be detected.
  • a substantial change in the expression level of the MRCK1 gene before and after the introduction of the siRNA sequence is indicative of the effectiveness of the siRNA sequence in suppressing the expression of the MRCK1 gene.
  • the expression levels of other genes are also monitored before and after the introduction of the siRNA sequence.
  • siRNA sequence which has inhibitory effect on MRCK1 gene expression but does not significantly affect the expression of other genes can be selected.
  • multiple siRNA or other RNAi sequences can be introduced into the same target cell. These siRNA or RNAi sequences specifically inhibit MRCK1 gene expression but not the expression of other genes.
  • siRNA or other RNAi sequences that inhibit the expression of both the MRCK1 gene and other gene or genes can be used.
  • the polynucleotides of the present invention can be modified at the base moiety, sugar moiety or phosphate backbone to improve the stability, hybridization, or solubility of the molecules.
  • the deoxyribose phosphate backbone of the polynucleotide molecules can be modified to generate peptide polynucleotides (see, Hyrup et al, Bioorganic & Medicinal Chemistry, 4:523, 1996).
  • peptide polynucleotides or “PNAs” refer to polynucleotide mimics, e.g., DNA mimics, in which the deoxyribose phosphate backbone is replaced by a pseudopeptide backbone and only the four natural nucleobases 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.
  • PNA oligomers can be synthesized using standard solid phase peptide synthesis protocols.
  • PNAs can be used in therapeutic and diagnostic applications.
  • PNAs can be used as antisense agents for sequence-specific modulation of the MRCK1 gene expression.
  • PNAs can also be used in the analysis of single base pair mutations in a gene, (e.g., by PNA-directed PCR clamping); as artificial restriction enzymes when used in combination with other enzymes, (e.g., S1 nucleases); or as probes or primers for DNA sequencing or hybridization.
  • PNAs can be modified 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 drug delivery techniques known in the art.
  • PNA-DNA chimeras of the polynucleotides of the invention can be generated. These chimeras allow DNA recognition enzymes, such as RNase H and DNA polymerases, to interact with the DNA portion while the PNA portion provides high binding affinity and specificity.
  • PNA-DNA chimeras can be linked using linkers of appropriate lengths which are selected based on base stacking, number of bonds between the nucleobases, and orientations.
  • the PNA-DNA chimeras can be synthesized as follows. A DNA chain is synthesized on a solid support using standard phosphoramidite coupling chemistry and modified nucleoside analogs. PNA monomers are then coupled in a stepwise manner to produce a chimeric molecule with a 5′ PNA segment and a 3′ DNA segment. Alternatively, chimeric molecules can be synthesized with a 5′ DNA segment and a 3′ PNA segment.
  • the polynucleotides of this invention may include other appended groups such as peptides (e.g., for targeting host cell receptors in vivo), or agents facilitating transportation across the cell membrane or the blood-kidney barrier (see, e.g., PCT Publication No. WO89/10134).
  • polynucleotides can be modified using hybridization-triggered cleavage agents or intercalating agents.
  • the polynucleotides can be conjugated to another molecule (e.g., a peptide, hybridization triggered cross-linking agent, transport agent, or hybridization-triggered cleavage agent).
  • the polynucleotide can be detectably labeled.
  • MRCK1 polypeptides and mutated MRCK1 polypeptides capable of inhibiting normal MRCK1 activity.
  • the present invention also contemplates immunogenic polypeptide fragments suitable for raising anti-MRCK1 antibodies.
  • native MRCK1 polypeptides can be isolated from cells or tissue sources by using standard protein purification techniques.
  • Standard purification methods include electrophoresis, molecular, immunological and chromatographic techniques. Specific examples include ion exchange, hydrophobic, affinity or reverse-phase HPLC chromatography, and chromatofocusing.
  • MRCK1 polypeptides are purified using a standard affinity column coupled with anti-MRCK1 antibodies. Ultrafiltration and diafiltration techniques can also be used. The degree of purification depends on the purpose of the use of the MRCK1 polypeptides. In some instances, purification is not necessary.
  • MRCK1 polypeptides or mutated MRCK1 polypeptides capable of inhibiting normal MRCK1 activity are produced by recombinant DNA techniques.
  • MRCK1 polypeptides or mutated MRCK1 polypeptides can be synthesized chemically using standard peptide synthesis techniques.
  • the invention provides MRCK1 polypeptides encoded by the human MRCK1 gene, or homologs thereof.
  • the polypeptides of this invention can be substantially homologous to human MRCK1 kinase (SEQ ID NO:2).
  • these polypeptides retain the biological activity of the native MRCK1 kinase.
  • the polypeptides comprise an amino acid sequence which is at least about 91%, 95%, 98%, 99% or more homologous to SEQ ID NO:2.
  • Comparison of sequences and determination of percent identity between two sequences can be accomplished using a mathematical algorithm.
  • the percent identity between two amino acid sequences can be determined using the Needleman and Wunsch (J. Mol. Biol. 48:444-453, 1970) algorithm, or the GAP program in the GCG software package which uses either a Blossom 62 matrix or a PAM250 matrix, and a gap weight of 16, 14, 12, 10, 8, 6, or 4 and a length weight of 1, 2, 3, 4, 5, or 6.
  • the percent identity between two nucleotide sequences can be determined using the GAP program in the GCG software package, which uses a NWSgapdna.CMP matrix and a gap weight of 40, 50, 60, 70, or 80 and a length weight of 1, 2, 3, 4, 5, or 6.
  • the percent identity between two amino acid or nucleotide sequences can also be determined using the algorithm of E. Meyers and W. Miller (CABIOS, 4:11-17, 1989) which has been incorporated into the ALIGN program (version 2.0), or the pairwise BLAST program available at NCBI's BLAST web site.
  • polypeptide and polynucleotide sequences of the present invention can be used as query sequences for searching public databases in order to identify similar sequences.
  • the search can be conducted using BLAST programs, such as the protein BLAST, nucleotide BLAST, pairwise BLAST, and genomic BLAST, that are available at the BLAST web site maintained by the NCBI.
  • BLAST programs such as the protein BLAST, nucleotide BLAST, pairwise BLAST, and genomic BLAST, that are available at the BLAST web site maintained by the NCBI.
  • the default parameters of the respective programs can also be used.
  • the invention further provides chimeric or fusion MRCK1 polypeptides.
  • a fusion MRCK1 polypeptide contains an MRCK1-related polypeptide and a non-MRCK1 polypeptide.
  • the MRCK1-related polypeptides include all or a portion of SEQ ID NO:2 or its variant.
  • a peptide linker sequence can be employed to separate the MRCK1-related polypeptide from the non-MRCK1 polypeptide components by a distance sufficient to ensure that each polypeptide folds into its native secondary and tertiary structures. Such a peptide linker sequence is incorporated into the fusion protein using standard techniques well-known in the art.
  • Suitable peptide linker sequences can be chosen based on the following factors: (1) their ability to adopt a flexible extended conformation; (2) their inability to adopt a secondary structure that could interact with functional epitopes on the MRCK1-related polypeptide and non-MRCK1 polypeptide; and (3) the lack of hydrophobic or charged residues that might react with the polypeptide functional epitopes.
  • Preferred peptide linker sequences contain Gly, Asn and Ser residues. Other near neutral amino acids, such as Thr and Ala can also be used in the linker sequence.
  • Amino acid sequences suitable as linkers include those disclosed in Maratea et al., Gene, 40:39-46, 1985; Murphy et al., Proc. Natl. Acad. Sci.
  • the linker sequences may be from 1 to about 50 amino acids in length. Linker sequences are not required when the MRCK1-related polypeptide or the non-MRCK1 polypeptide has non-essential N-terminal amino acid regions that can be used to separate the respective functional domains and thereby prevent steric interference.
  • the fusion protein is a GST-MRCK1 fusion protein in which an MRCK1-related sequence, such as SEQ ID NO:2, is fused to the C-terminus of the GST sequence.
  • This fusion protein can facilitate the purification of the recombinant MRCK1.
  • the MRCK1-fusion proteins of the invention can be incorporated into pharmaceutical compositions and administered to a subject.
  • the MRCK1-fusion proteins can be used to affect the bioavailability of an MRCK1 substrate.
  • the MRCK1-fusion proteins can also be used for the treatment or prevention of damages caused by (i) aberrant modification or mutation of MRCK1, or (ii) aberrant post-translational modification of MRCK1. It is also conceivable that a fusion protein containing a normal or mutated MRCK1 polypeptide, or a fragment thereof, can be used to inhibit MRCK1 activity in a human subject.
  • the MRCK1-fusion proteins can be used as immunogens to produce anti-MRCK1 antibodies. They can also be used to purify MRCK1 ligands and to screen for molecules capable of inhibiting the interaction between MRCK1 and its substrates.
  • the MRCK1-chimeric or fusion proteins of the invention are produced using standard recombinant DNA techniques.
  • Commercially available expression vectors which encode a fusion moiety e.g., a GST polypeptide can be used.
  • a signal sequence can be used to facilitate secretion and isolation of the secreted protein or other proteins of interest.
  • Signal sequences are typically characterized by a core of hydrophobic amino acids which are generally cleaved from the mature protein.
  • Such signal peptides contain processing sites that allow cleavage of the signal sequence from the mature proteins as they pass through the secretory pathway.
  • the present invention encompasses MRCK1 polypeptides having a signal sequence, or the polynucleotide sequences encoding the same.
  • the present invention also pertains to MRCK1 mutants which function as antagonists to MRCK1.
  • antagonists of MRCK1 are used as therapeutic agents.
  • a mutant of MRCK1 that forms a non-functional dimer with a wide-type MRCK1 can decrease the activity of MRCK1 and may ameliorate diseases in a subject wherein MRCK1 are abnormally increased in level or activity.
  • Dominant negative MRCK1 mutants can be generated by mutagenesis, as appreciated by one skilled in the art.
  • MRCK1 mutants which function as either MRCK1 agonists or antagonists can be identified by screening combinatorial libraries of mutants.
  • a variegated library of MRCK1 mutants can be produced by, for example, enzymatically ligating a mixture of synthetic oligonucleotides into gene sequences such that a degenerate set of potential MRCK1 sequences is expressible as individual polypeptides, or alternatively, as a set of larger fusion proteins containing the set of MRCK1 sequences therein.
  • a degenerate gene sequence can be chemically synthesized using an automatic DNA synthesizer. The synthetic gene can then be ligated into an appropriate expression vector.
  • a library of coding sequences can be generated using nucleases. For instance, double-stranded PCR fragments of the MRCK1 coding sequence can be treated by a nuclease which produces about one nick per molecule. The double-stranded DNAs then are subject to a cycle of denaturing and re-naturing. The newly reformed DNAs, which may include sense/antisense pairs from different nicked products, are treated with S1 nuclease to remove single-stranded portions. Using this method, an expression library which encodes N-terminal, C-terminal or internal fragments of MRCK1 can be derived.
  • REM recursive ensemble mutagenesis
  • MRCK1 fragments can also be generated using synthetic means, such as solid-phase synthesis methods.
  • the synthesized fragment has less than about 100 amino acids, or preferably, less than about 50 amino acids.
  • antibodies specific to MRCK1 or its variants are prepared.
  • An antibody is considered to bind “specifically” to an antigen if the binding affinity between the antibody and the antigen is equal to, or greater than 10 5 M ⁇ 1 .
  • the antibodies can be monoclonal or polyclonal.
  • the antibodies are monoclonal. More preferably, the antibodies are humanized antibodies.
  • Polyclonal anti-MRCK1 antibodies can be prepared by immunizing a suitable subject with MRCK1 or fragments thereof.
  • the anti-MRCK1 antibody titer in the immunized subject can be monitored over the time using standard techniques, such as ELISA.
  • the anti-MRCK1 antibody can be isolated from the immunized subject using well-known techniques.
  • hybridomas capable of producing anti-MRCK1 antibodies are prepared.
  • Purified MRCK1 or its variants, or fragments thereof, are used to immunize a vertebrate, such as a mammal. Suitable mammals include mice, rabbits and sheep.
  • the fragment used for immunization comprises at least 8 amino acid residues, more preferably at least 12 amino acid residues, highly preferably at least 16 amino acid residues, and most preferably at least 20 amino acid residues.
  • Immunogenic fragments (epitopes) of MRCK1 can be identified using well-known techniques.
  • any fragment of SEQ ID NO:2 can be used to raise antibodies specific to MRCK1.
  • Preferred epitopes are regions that are located on the surface of MRCK1. These regions are usually hydrophilic.
  • Splenocytes are isolated from the immunized vertebrate and fused with an immortalized cell line (such as a myeloma) to form hybridomas.
  • an immortalized cell line such as a myeloma
  • the immortal cell line is derived from the same mammalian species as the lymphocytes.
  • murine hybridomas can be made by fusing an immortalized mouse cell line with lymphocytes isolated from a mouse that is immunized with an immunogenic preparation of the present invention.
  • Preferred immortalized cell lines include mouse myeloma cell lines that are sensitive to culture medium containing hypoxanthine, aminopterin and thymidine (“HAT medium”).
  • Suitable myeloma cell lines include, but are not limited to, the P3-NS1/1-Ag4-1, P3-x63-Ag8.653 or Sp210-Ag14 myeloma lines, all of which are available from ATCC.
  • HAT-sensitive mouse myeloma cells are fused to mouse splenocytes using polyethylene glycol (“PEG”).
  • PEG polyethylene glycol
  • Hybridoma cells thus produced are selected against HAT medium, which kills unfused or unproductively fused myeloma cells.
  • Hybridoma cells which produce monoclonal anti-MRCK1 antibodies are then detected by screening the hybridoma culture supernatants.
  • a monoclonal anti-MRCK1 antibody can also be prepared by screening a recombinant combinatorial immunoglobulin library (e.g., an antibody phase display library).
  • Kits for generating and screening phage display libraries are commercially available (e.g., the Pharmacia Recombinant Phage Antibody System, Catalog No. 27-9400-01; and the Stratagene SurfZAPTM Phage Display Kit, Catalog No. 240612).
  • the anti-MRCK1 antibodies of the present invention also include “single-chain Fv” or “scFv.”
  • the scFv fragments comprise the V H and V L domains of an antibody.
  • the scFv polypeptide further comprises a polypeptide linker between the V H and V L domains.
  • the polypeptide linker enables the scFv to form the desired structure for antigen binding.
  • recombinant anti-MRCK1 antibodies such as chimeric and humanized monoclonal antibodies, can be prepared, as appreciated by one of ordinary skill in the art.
  • Humanized antibodies are particularly desirable for therapeutic treatment of human subjects.
  • Humanized forms of non-human (e.g., murine) antibodies are chimeric immunoglobulins, immunoglobulin chains, or fragments thereof (such as Fv, Fab, Fab′, F(ab) 2 or other antigen-binding subsequences of antibodies) which contain minimal sequence derived from non-human immunoglobulin.
  • Humanized antibodies are derived from human immunoglobulins in which the residues forming the complementary determining regions (CDRs) are replaced by the residues from CDRs of a non-human antibody, such as a mouse, rat or rabbit antibody having the desired specificity, affinity and capacity.
  • CDRs complementary determining regions
  • Fv framework residues of the human immunoglobulin are replaced by corresponding non-human residues.
  • Humanized antibodies may also comprise residues which are found neither in the recipient antibody nor in the imported CDR or framework sequences.
  • the humanized antibody can comprise at least one or 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 constant regions are those of a human immunoglobulin consensus sequence.
  • the humanized antibody preferably comprises at least a portion of an immunoglobulin constant region (Fc) of a human immunoglobulin.
  • Humanized antibodies can be produced using transgenic mice which are incapable of expressing endogenous immunoglobulin heavy and light chains but can express human heavy and light chains.
  • the transgenic mice are immunized in the normal fashion with a selected antigen.
  • Monoclonal antibodies directed against the antigen can be obtained using conventional hybridoma technology.
  • the human immunoglobulin transgenes harbored in the transgenic mice rearrange during B cell differentiation, and subsequently undergo class switching and somatic mutation. Using this technique, therapeutically useful IgG, IgA and IgE antibodies can be prepared.
  • humanized antibodies which recognize a selected epitope can be generated using a technique referred to as “guided selection.”
  • a selected non-human monoclonal antibody e.g., a murine antibody, is used to guide the selection of a humanized antibody recognizing the same epitope.
  • the antibodies to MRCK1 are capable of reducing or eliminating the biological function of MRCK1.
  • the antibodies reduce at least 25% of MCRK1 activity. More preferably, the antibodies reduce at least about 50% of the activity. Highly preferably, the antibodies reduce about 95-100% of MRCK1 activity.
  • Anti-MRCK1 antibodies can be used to isolate MRCK1. Suitable methods include affinity chromatography and immunoprecipitation. Moreover, anti-MRCK1 antibodies can be used to evaluate the expression level of MRCK1. Anti-MRCK1 antibodies can also be used to monitor MRCK1 level as part of a clinical testing procedure, or to determine the efficacy of a given treatment regimen. Detection can be facilitated by coupling the antibody to a detectable substance. Examples of 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 streptavidin/biotin and avidin/biotin;
  • suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin;
  • an example of a luminescent material includes luminol;
  • examples of bioluminescent materials include luciferase, luciferin, and aequorin; and
  • suitable radioactive materials include 125 I, 131 I, 35 S or 3 H.
  • Anti-MRCK1 antibodies are also useful for targeting a therapeutic agent/drug to a particular cell or tissue.
  • the therapeutic agent/drug may be coupled to an antibody, either covalently or non-covalently.
  • a therapeutic agent can be coupled to an antibody via a linker group.
  • a linker group can function as a spacer to separate the antibody from the agent so as to avoid interference with antibody's binding capabilities.
  • the linker group can also serve to increase the chemical reactivity of a substituent on the agent or the antibody, and thus increase the coupling efficiency.
  • a variety of bifunctional or polyfunctional reagents either homo- or hetero-functional (such as those described in the catalog of the Pierce Chemical Co., Rockford, Ill.), can be employed as the linker group.
  • Coupling may be effected, for example, through amino groups, carboxyl groups, sulfhydryl groups or oxidized carbohydrate residues.
  • This methodology See e.g., U.S. Pat. No. 4,671,958.
  • linker group which is cleavable during or upon internalization into the target cell.
  • a number of different cleavable linker groups have been described.
  • the mechanisms for the intra-cellular release of an agent from these linker groups include cleavage by reduction of a disulfide bond (e.g., U.S. Pat. No. 4,489,710), by irradiation of a photolabile bond (e.g., U.S. Pat. No. 4,625,014), by hydrolysis of derivatized amino acid side chains (e.g., U.S. Pat. No.
  • immunoconjugates coupled with more than one agent can be prepared in a variety of ways, as appreciated by one of ordinary skill in the art.
  • Another aspect of the invention pertains to vectors containing a polynucleotide encoding MRCK1 or a portion thereof.
  • One type of vector is a “plasmid,” which includes a circular double-stranded DNA into which additional DNA segments can be introduced.
  • Vectors also include expression vectors and gene delivery vectors.
  • the expression vectors of the present invention comprise a polynucleotide encoding MRCK1 or a portion thereof.
  • the expression vectors also include one or more regulatory sequences operably linked to the polynucleotide being expressed. These regulatory sequences are selected based on the type of host cells. It will be appreciated by those skilled in the art that the design of the expression vector depends on such factors as the choice of the host cells and the desired expression levels.
  • MRCK1 can be expressed in bacterial cells such as E. coli , insect cells (using baculovirus expression vectors), yeast cells or mammalian cells.
  • the expression vector can also be transcribed and translated in vitro, for example, by 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 purposes: 1) to increase expression of the recombinant protein; 2) to increase the solubility of the recombinant protein; and 3) 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.
  • Suitable cleavage enzymes include Factor Xa, thrombin and enterokinase.
  • Examples of fusion expression vectors include pGEX (Pharmacia Piscataway, N.J.), pMAL (New England Biolabs, Beverly, Mass.) and pRITS (Pharmacia, Piscataway, N.J.). Purified fusion proteins can be utilized in MRCK1 activity assays, or to generate antibodies specific for MRCK1.
  • suitable inducible non-fusion E. coli expression vectors include pTrc and pET 11d.
  • Target gene expression from the pTrc vector relies on host RNA polymerase transcription from a hybrid trp-lac fusion promoter.
  • Target gene expression from the pET 11d vector relies on transcription from a T7 gn10-lac fusion promoter mediated by a co-expressed viral RNA polymerase (T7 gn1). This viral polymerase is supplied by host strains BL21(DE3) or HSLE174(DE3) from a resident prophage harboring a T7 gn1 gene under the transcriptional control of the lacUV 5 promoter.
  • One strategy to maximize recombinant protein expression in E. coli is to express the protein in host bacteria that have an impaired capacity to proteolytically cleave the recombinant protein. Another strategy is to alter the polynucleotide sequence encoding the protein so that the individual codons for each amino acid are those preferentially utilized in E. coli.
  • the MRCK1 expression vector is a yeast expression vector.
  • yeast expression vectors include pYepSec1, pMFa, pJRY88, pYES2 (Invitrogen Corporation, San Diego, Calif.), and picZ (Invitrogen Corp, San Diego, Calif.).
  • MRCK1 or its variant can be expressed in insect cells using baculovirus expression vectors.
  • Suitable baculovirus vectors include the pAc series and the pVL series.
  • MRCK1 or its variant is expressed in mammalian cells using a mammalian expression vector.
  • mammalian expression vectors include pCDM8 and pMT2PC.
  • the expression vector's control functions are often provided by viral regulatory elements.
  • commonly used promoters are derived from polyoma, adenovirus 2, cytomegalovirus and Simian Virus 40.
  • the mammalian expression vector contains tissue-specific regulatory elements.
  • tissue-specific promoters include the liver-specific albumin promoter, lymphoid-specific promoters, promoters of T cell receptors and immunoglobulins, neuron-specific promoters (e.g., the neurofilament promoter), pancreas-specific promoters, and mammary gland-specific promoters (e.g., milk whey promoter).
  • Developmentally-regulated promoters are also contemplated, which include, for example, the ⁇ -fetoprotein promoter.
  • the present invention also provides a recombinant expression vector comprising a polynucleotide which encodes MRCK1 but is cloned into the expression vector in an antisense orientation.
  • Regulatory sequences that are operatively linked to the antisense-oriented polynucleotide can be chosen to direct continuous expression of the antisense RNA molecule in a variety of cell types. Suitable regulatory sequences include viral promoters and/or enhancers. Regulatory sequences can also be chosen to direct constitutive, tissue specific or cell type specific expression of the antisense RNA.
  • the antisense expression vector can be in the form of a recombinant plasmid, phagemid, or attenuated virus in which antisense polynucleotides are produced under the control of a highly efficient regulatory region.
  • the present invention further provides gene delivery vehicles for delivering polynucleotides to mammals.
  • a polynucleotide sequence of the invention can be administered either locally or systemically via a gene delivery vehicle. Expression of the polynucleotide can be induced using endogenous mammalian or heterologous promoters. Expression of the polynucleotide in vivo can be either constituted or regulated.
  • the gene delivery vehicles preferably are viral vectors, including retroviral, lentiviral, adenoviral, adeno-associated viral (AAV), herpes viral, or alphavirus vectors.
  • the viral vectors can also be astrovirus, coronavirus, orthomyxovirus, papovavirus, paramyxovirus, parvovirus, picornavirus, poxvirus, or togavirus vectors.
  • Delivery of gene therapy constructs is not limited to the above mentioned viral vectors.
  • Other delivery methods can also be employed. These methods include nucleic acid expression vectors, polycationic condensed DNA linked or unlinked to killed adenovirus, ligand linked DNA, liposome-DNA conjugates, gene guns, ionizing radiation, nucleic charge neutralization, or fusion with cell membranes.
  • Naked DNA can also be employed. Uptake efficiency of the naked DNA may be improved using biodegradable latex beads. This method can be further improved by treating the beads to increase their hydrophobicity.
  • Another aspect of the present invention pertains to the use of regulatable expression systems to express desirable polynucleotides or polypeptides in cells.
  • Systems suitable for this invention are briefly described below:
  • Tet-on/off system The Tet-system is based on two regulatory elements derived from the tetracycline-resistance operon of the E. coli Tn10 transposon: the tet repressor protein (TetR) and the Tet operator DNA sequence (tetO) to which TetR binds (Gossen et al., Science, 268: 1766-1769, 1995).
  • the system consists of two components, a “regulator” and a “reporter” plasmid.
  • the “regulator” plasmid encodes a hybrid protein containing a mutated Tet repressor (rtetR) fused to the VP16 activation domain of herpes simplex virus.
  • the “reporter” plasmid contains a tet-responsive element (TRE), which controls the “reporter” gene of choice.
  • TRE tet-responsive element
  • the rtetR-VP16 fusion protein can only bind to the TRE, therefore activating the transcription of the “reporter” gene in the presence of tetracycline.
  • the system has been incorporated into a number of viral vectors including retrovirus, adenovirus and AAV.
  • Ecdysone system The ecdysone system is based on the molting induction system found in Drosophila , but modified for inducible expression in mammalian cells.
  • the system uses an analog of the Drosophila steroid hormone ecdysone, muristerone A, to activate expression of the gene of interest via a heterodimeric nuclear receptor. Expression levels have been reported to exceed 200-fold over basal levels with no effect on mammalian cell physiology (No et al., Proc. Natl. Acad. Sci. USA, 93: 3346-3351, 1996).
  • the progesterone receptor is normally stimulated to bind to a specific DNA sequence and to activate transcription through an interaction with its hormone ligand.
  • the progesterone antagonist mifepristone (RU486) is able to block hormone-induced nuclear transport and subsequent DNA binding.
  • a mutant form of the progesterone receptor that can be stimulated to bind through an interaction with RU486 has been generated.
  • the RU486-binding domain of the progesterone receptor has been fused to the DNA-binding domain of the yeast transcription factor GAL4 and the transactivation domain of the HSV protein VP16.
  • the chimeric factor is inactive in the absence of RU486.
  • Rapamycin-system Immunosuppressive agents, such as FK506 and rapamycin, act by binding to specific cellular proteins and facilitating their dimerization. For example, the binding of rapamycin to FK506-binding protein (FKBP) results in its heterodimerization with another rapamycin binding protein FRAP, which can be reversed by removal of the drug. The ability to bring two proteins together by addition of a drug potentiates the regulation of a number of biological processes, including transcription.
  • a chimeric DNA-binding domain has been fused to the FKBP, which enables binding of the fusion protein to a specific DNA-binding sequence.
  • a transcriptional activation domain also has been fused to FRAP.
  • a fully functional transcription factor can be formed by heterodimerization mediated by addition of rapamycin.
  • the dimerized chimeric transcription factor can then bind to a synthetic promoter sequence containing copies of the synthetic DNA-binding sequence.
  • This system has been successfully integrated into adenoviral and AAV vectors. Long term regulatable gene expression has been achieved in both mice and baboons (Ye et al., Science, 283: 88-91, 1999).
  • MRCK1 In patients with disorders related to the aberrant expression of MRCK1.
  • the expression level of MRCK1 can be used as an indicator for detecting the presence of MRCK1-related diseases in humans. Detection and measurement of the relative amount of the MRCK1 gene product can be carried out using various methods known in the art.
  • Typical methodologies for detecting the transcription level of a gene include extracting RNA from a cell or tissue sample, hybridizing a labeled probe to the extracted RNA or derivative thereof (such as cDNA or cRNA), and detecting the probe. Suitable methods include Northern Blot and quantitative PCR or RT-PCR. In situ hybridization can also be used to detect the transcription level of the MRCK1 gene in human tissues.
  • Typical methodologies for detecting a polypeptide include extracting proteins from a cell or tissue sample, binding an antibody to the target polypeptide and detecting the antibody. Suitable methods include enzyme linked immunosorbent assays (ELISAs), Western blots, immunoprecipitations, and immunofluorescence.
  • the antibody can be polyclonal, or preferably, monoclonal.
  • the antibody can be an intact antibody, or a fragment thereof (e.g. Fab or F(ab′) 2 ).
  • the antibody can be labeled with a radioisotope, a fluorescent compound, an enzyme, an enzyme co-factor, or a detectable ligand.
  • labeling with regard to a probe or antibody, is intended to encompass direct labeling such as through covalent coupling, as well as indirect labeling such as being mediated by another reagent which is directly labeled.
  • indirect labeling include labeling a primary antibody using a fluorescently labeled secondary antibody, or attaching a DNA probe with a biotin which can be detected, for example, by a fluorescence-labeled streptavidin.
  • the binding affinity of the antibody to MRCK1 is at least 10 5 M ⁇ 1 . More preferably, the binding affinity is at least 10 6 M ⁇ 1 .
  • Other methods such as electrophoresis, chromatography or direct sequencing can also be used to detect the amount of a polypeptide in a biological sample.
  • Anti-MRCK1 antibodies can also be directly introduced into a subject. The antibody can be labeled with a radioactive marker whose presence and location in the subject can be detected using standard imaging techniques.
  • the genomic copies of the MRCK1 gene in the genome of a human subject may indicate the presence or predisposition of a disease. Detection of the presence or number of copies of the MRCK1 gene in the genome can be performed using methods known in the art. For instance, it can be assessed using Southern Blot.
  • the probes for Southern Blot can be labeled with a radioisotope, a fluorescent compound, an enzyme, or an enzyme co-factor.
  • the above-described detection methods can be used to determine the severity of MRCK1-related diseases.
  • a biological sample is isolated from a test subject, and the presence, quantity and/or activity of MRCK1 in the sample relative to a normal or control sample is evaluated.
  • the expression level of MRCK1 in the biological sample can indicate the presence or severity of MRCK1-related diseases in the test subject.
  • the term “biological sample” is intended to include tissues, cells or biological fluids isolated from the subject.
  • a preferred biological sample is a serum sample isolated from the subject using conventional means.
  • the present invention also provides methods for identifying MRCK1 modulators.
  • the activity of MRCK1 can be evaluated using various methods, such as those disclosed in Leung et al., Mol. Cell. Biol., 18:130-140, 1998, and Chen et al., J. Biol. Chem., 274: 19901-19905, 1999, both of which are incorporated herein by reference.
  • Suitable modulators include compounds or agents comprising therapeutic moieties, such as peptides, peptidomimetics, peptoids, polynucleotides, small molecules or other drugs. These moieties can either bind to MRCK1, or have a modulatory (e.g., stimulatory or inhibitory) effect on the activity of MRCK1. In one embodiment, the moieties have a modulatory effect on the interactions of MRCK1 with one or more of its natural substrates. These moieties can also exert a modulatory effect on the expression of MRCK1.
  • the screen assays of the present invention comprise detecting the interactions between MRCK1 and test components.
  • test compounds of the present invention can be either small molecules or bioactive agents.
  • the test compound is a small organic or inorganic molecule.
  • the test compound is a polypeptides, oligopeptides, polysaccharides, nucleotides or polynucleotides.
  • kits for screening for compounds that inhibit the biological activities of MRCK1 are provided.
  • Pharmaceutical compositions comprising these compounds can subsequently be prepared.
  • the screening method comprises (1) contacting a sample with a compound, and (2) comparing expression profile or biological activity of MRCK1 in the sample to determine whether the compound substantially decreases the expression level or activities of MRCK1.
  • the screening method can be carried out either in vivo or in vitro.
  • the present invention further includes a method for screening for compounds capable of modulating the binding between MRCK1 and a binding partner.
  • binding partner refers to a bioactive agent which serves as either a substrate for MRCK1, or a ligand having a binding affinity to MRCK1.
  • the bioactive agent may be selected from a variety of naturally-occurring or synthetic compounds, proteins, peptides, polysaccharides, nucleotides or polynucleotides.
  • Inhibitors of the expression, activity or binding ability of MRCK1 may be used as therapeutic compositions. These inhibitors can be formulated in suitable pharmaceutical compositions, as described herein below.
  • the present invention also provides methods for conducting high-throughput screening for compounds capable of inhibiting activity or expression of MRCK1.
  • the high-throughput screening method involves contacting test compounds with MRCK1, and then detecting the effect of the test compounds on MRCK1.
  • Functional assays such as cytosensor microphysiometer-based assays, calcium flux assays (e.g. FLIPR®, Molecular Devices Corp, Sunnyvale, Calif.), or the TUNEL assay, can be employed to measure MRCK1 cellular activity.
  • Fluorescence-based techniques can be used for high-throughput and ultra high-throughput screening. They include, but are not limited to, BRET® and FRET® (both by Packard Instrument Co., Meriden, Conn.).
  • the high-throughput screening assay uses label-free plasmon resonance technology as provided by BIACORE® systems (Biacore International AB, Uppsala, Sweden). Plasmon free resonance occurs when surface plasmon waves are excited at a metal/liquid interface. By reflecting directed light from the surface as a result of contact with a sample, the surface plasmon resonance causes a change in the refractive index at the surface layer.
  • the refractive index change for a given change of mass concentration at the surface layer is similar for many bioactive agents (including proteins, peptides, lipids and polynucleotides), and since the BIACORE® sensor surface can be functionalized to bind a variety of these bioactive agents, detection of a wide selection of test compounds can thus be accomplished.
  • the efficacy of a therapeutic agent for MRCK1-related diseases can be monitored during clinical trials.
  • the therapeutic agent may be a drug, small molecule, agonist, antagonist, peptidomimetic, protein, peptide, or polynucleotide.
  • the changes in the expression or activity of the MRCK1 gene in response to the treatment of the agent can be used to evaluate the therapeutic effect of the agent on patients with MRCK1-related diseases.
  • the expression or activity of MRCK1 in response to the agent can be measured at various points during the clinical trial.
  • the method for monitoring the effectiveness of the therapeutic agent includes the steps of (i) obtaining a pre-administration sample from a subject; (ii) detecting the level of expression or activity of MRCK1 in the pre-administration sample; (iii) obtaining one or more post-administration samples from the subject; (iv) detecting the level of expression or activity of MRCK1 in the post-administration samples; (v) comparing the level of expression or activity of MRCK1 in the pre-administration sample to the level of expression or activity of MRCK1 in the post administration samples.
  • the dose or frequency of the administration of the agent may be adjusted based on the effectiveness of the agent in a particular patient. Therefore, MRCK1 expression or activity can be used as an indicator of the effectiveness of a therapeutic agent for MRCK1-related diseases, even if the agent does not produce an observable phenotypic response.
  • the detection methods described herein can be used to identify subjects having or at risk of developing MRCK1-related diseases.
  • the detection methods can be used to determine whether an agent (e.g., an agonist, antagonist, peptidomimetic, protein, peptide, polynucleotide, small molecule, or other drug candidate) can be administered to a subject for effectively treating or preventing MRCK1-related diseases.
  • an agent e.g., an agonist, antagonist, peptidomimetic, protein, peptide, polynucleotide, small molecule, or other drug candidate
  • MRCK1 expression profiles at different progression stages of MRCK1-related diseases can be established.
  • MRCK1 expression profiles in different patients who have different responses to a drug treatment are determined.
  • a pattern may emerge such that a particular expression profile may be correlated to an increased likelihood of a poor prognosis. Therefore, the prognostic assay of the present invention may be used to determine whether a subject undergoing a treatment for an MRCK1-related disease has a poor outlook for long term survival or disease progression.
  • prognosis is performed shortly after diagnosis, such as within a few days after diagnosis. The result of prognosis can then be used to devise individualized treatment program, thereby enhancing the effectiveness of the treatment as well as the likelihood of long-term survival and well being.
  • the method of the invention can also be used to detect genetic alterations in the MRCK1 gene, thereby determining if a subject with the altered gene is at risk for damages characterized by aberrant regulation in MRCK1 activity or expression.
  • the method includes detecting the presence or absence of a genetic alteration that affects the integrity of the MRCK1 gene, or detecting the aberrant expression of the MRCK1 gene.
  • the genetic alteration can be detected by ascertaining the existence of at least one of the following: 1) deletion of one or more nucleotides from the MRCK1 gene; 2) addition of one or more nucleotides to the MRCK1 gene; 3) substitution of one or more nucleotides of the MRCK1 gene, 4) a chromosomal rearrangement in the MRCK1 gene; 5) alteration in the level of a messenger RNA transcript of the MRCK1 gene, 6) aberrant modification of the MRCK1 gene, 7) the presence of a non-wild-type splicing pattern of a messenger RNA transcript of the MRCK1 gene, 8) non-wild-type level MRCK1, 9) allelic loss of an MRCK1 gene, and 10) inappropriate post-translational modification of MRCK1.
  • detection of the alteration involves the use of a probe/primer in a polymerase chain reaction (such as anchor PCR or RACE PCR) or alternatively, in a ligation chain reaction (LCR).
  • LCR can be particularly useful for detecting point mutations in the MRCK1 gene.
  • This method includes the steps of collecting a sample from a subject, isolating polynucleotides (e.g., genomic DNA, mRNA, or both) from the sample, contacting the polynucleotide with one or more primers which specifically hybridize to the MRCK1 gene or gene product, and detecting the presence or absence of an amplification product, or detecting the size of the amplification product and comparing its length to a control. It is understood that PCR and/or LCR can be used as a preliminary amplification step in conjunction with any other techniques described herein.
  • Alternative amplification methods include: self sustained sequence replication (Guatelli et al., Proc. Natl. Acad. Sci. USA, 87:1874-1878, 1990), transcriptional amplification system (Kwoh et al., Proc. Natl. Acad. Sci. USA, 86:1173-1177, 1989), and Q-Beta Replicase (Lizardi et al., Bio-Technology, 6:1197, 1988).
  • mutations in the MRCK1 gene can be identified using restriction enzymes. Differences in restriction enzyme digestion patterns indicates mutation(s) in the MRCK1 gene or its transcripts. Moreover, sequence specific ribozymes can be used to detect the presence of specific mutations. See e.g., U.S. Pat. No. 5,498,531.
  • genetic mutations in the MRCK1 gene can be identified using high density arrays which contain a large number of oligonucleotides probes.
  • genetic mutations in the MRCK1 gene can be identified in two dimensional arrays.
  • a first hybridization array of probes is used to scan through long stretches of DNA in a sample and a control in order to identify base changes between the two sequences. This step allows the identification of point mutations.
  • This step is followed by a second hybridization array that allows the characterization of specific mutations by using smaller and specialized probe arrays which are complementary to all variants or mutations detected.
  • Each mutation array is composed of parallel probe sets, one complementary to the wild-type gene and the other complementary to the mutant gene.
  • any sequencing reactions known in the art can be used to directly sequence the MRCK1 gene in order to detect mutations. It is contemplated that any automated sequencing procedures can be utilized, including sequencing by mass spectrometry.
  • protection from cleavage agents is used to detect mismatched bases in RNA/RNA or RNA/DNA heteroduplexes.
  • the “mismatch cleavage” technique involves forming heteroduplexes by hybridizing an RNA or DNA (labeled) containing the wild-type MRCK1 gene sequence to a potentially mutant RNA or DNA obtained from a tissue sample.
  • the double-stranded duplexes are treated with an agent which cleaves single-stranded regions of the duplex.
  • the agent may be RNase (for RNA/DNA duplexes), or S1 nuclease (for DNA/DNA hybrids).
  • DNA/DNA or RNA/DNA duplexes are treated with piperidine and hydroxylamine, or piperidine and osmium tetroxide, in order to digest mismatched regions. After the digestion, the resulting material is separated by size on a denaturing polyacrylamide gel from which the site(s) of mutation may be determined.
  • the mismatch cleavage reaction employs one or more proteins that recognize mismatched base pairs in double-stranded DNA.
  • these proteins include “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.
  • cDNAs are prepared from mRNAs isolated from test cells. The cDNAs are then hybridized to a probe derived from the MRCK1 gene. The duplex thus formed 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. Pat. No. 5,459,039.
  • alterations in electrophoretic mobility are used to identify mutations in the MRCK1 gene. Differences in electrophoretic mobility between mutant and wild-type polynucleotides can be detected using single strand conformation polymorphism (SSCP). The resulting alteration in electrophoretic mobility enables the detection of a single base change.
  • the DNA fragments can be labeled or detected with probes.
  • the sensitivity of the assay is enhanced by using RNA, in which the secondary structure is more sensitive to a change in sequence.
  • the assay utilizes heteroduplex analysis to separate double-stranded heteroduplex molecules on the basis of changes in electrophoretic mobility (Keen et al., Trends Genet., 7:5, 1991).
  • the movement of mutant or wild-type fragments is evaluated using denaturing gradient gel electrophoresis (DGGE).
  • DGGE denaturing gradient gel electrophoresis
  • DNA fragments can be modified to insure that they do not completely denature.
  • a GC clamp of approximately 40 GC-rich base pairs can be added to the DNA fragment using PCR.
  • a temperature gradient is used in place of a denaturing gradient (Rosenbaum and Reissner Biophys Chem, 265:12753, 1987).
  • oligonucleotide primers for specific amplification carry the mutation of interest in the center of the molecule (so that amplification depends on differential hybridization) or at the extreme 3′ end of one primer where, under appropriate conditions, mismatch can prevent or reduce polymerase extension. See e.g., Saiki et al., Proc. Natl. Acad. Sci. USA, 86:6230, 1989.
  • kits which comprise at least one polynucleotide probe or one antibody of the present invention. These kits can be used in clinical settings to diagnose subjects exhibiting symptoms or family history of an MRCK1-related disease. Any cell type or tissue in which MRCK1 is expressed can be used for prognostic or diagnostic purposes.
  • This invention also provides methods for preventing diseases associated with aberrant MRCK1 expression or activity.
  • the methods comprise administering to a target subject an agent which modulates MRCK1 expression or activity.
  • a prophylactic agent can be administered prior to the manifestation of MRCK1-related disease symptoms in order to prevent or delay MRCK1-related diseases.
  • Suitable prophylactic agents include mutant MRCK1 proteins, MRCK1 antagonist agents, or MRCK1 antisense polynucleotides.
  • the prophylactic methods of this invention can be specifically tailored or modified, based on knowledge obtained from the study of pharmacogenomics.
  • Pharmacogenomics includes the application of genomics technologies, such as gene sequencing, statistical genetics, and gene expression analysis, to drugs which are either in clinical development or on the market.
  • Pharmacogenomics can be used to determine a subject's response to a drug (e.g., a subject's “drug response phenotype” or “drug response genotype”).
  • another aspect of this invention is to provide methods for tailoring an individual's prophylactic or therapeutic treatment using MRCK1 modulators according to the individual's drug response genotype.
  • Pharmacogenomics allows a clinician or physician to target prophylactic or therapeutic treatments to subjects who will most benefit from the treatment and to avoid treatment of subjects who will experience toxic drug-related side effects.
  • a genome-wide association relies primarily on a high-resolution map of the human genome consisting of already known gene-related sites (e.g., a “bi-allelic” gene marker map which consists of 60,000-100,000 polymorphic or variable sites on the human genome, each of which has two variants).
  • a high-resolution genetic map can be compared to a map of the genome of each of a statistically substantial number of subjects taking part in a Phase II/III drug trial in order to identify genes associated with a particular observed drug response or side effect.
  • such a high resolution map can be generated from a combination of some ten-million known single nucleotide polymorphisms (SNPs) in the human genome.
  • SNP single nucleotide polymorphisms
  • a “SNP” is a common alteration that occurs in a single nucleotide base in a stretch of DNA. For example, a SNP may occur once per every 1000 bases of DNA.
  • a SNP may be involved in a disease process. However, the vast majority of SNPs may be not related to diseases. Given a genetic map based on the occurrence of SNPs, individuals can be grouped into genetic categories depending on a particular pattern of SNPs in their individual genome.
  • treatment regimens can be tailored to groups of genetically similar individuals, taking into account traits that may be common among such genetically similar individuals.
  • mapping of the MRCK1 gene to SNP maps of patients with MRCK1-related diseases may facilitate the identification of drug-response-prediction genes.
  • the “candidate gene approach” can be utilized to identify genes that predict drug response. According to this method, if a gene that encodes a drug target is known, all common variants of that gene can be easily identified in the population. It then can be determined if a particular drug response is associated with one version of the gene versus another.
  • the activity of drug metabolizing enzymes is a major determinant of both the intensity and duration of drug action.
  • drug metabolizing enzymes e.g., N-acetyltransferase 2 (NAT 2) and cytochrome P450 enzymes CYP2D6 and CYPZC19
  • NAT 2 N-acetyltransferase 2
  • CYP2D6 and CYPZC19 cytochrome P450 enzymes
  • the gene coding for CYP2D6 is highly polymorphic and several mutations have been identified in poor metabolizers, 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, poor metabolizers show no therapeutic response. 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 “gene expression profiling” method can be utilized to identify genes that predict drug response.
  • the gene expression profile of an animal dosed with a drug can give an indication of whether the gene pathways related to toxicity have been turned on.
  • Information generated from the above pharmacogenomics approaches can be used to determine the appropriate dosage or treatment regimen suitable for a particular individual. This knowledge can avoid adverse reactions or therapeutic failure, and therefore enhance therapeutic or prophylactic efficiency when treating a subject with an MRCK1 modulator.
  • the present invention includes therapeutic methods for treating a subject at risk for, susceptible to, or diagnosed with MRCK1-related diseases.
  • the therapeutic methods can be individually tailored based on the subject's drug response genotype.
  • the therapeutic methods comprise modulating the expression or activity of MRCK1 in the subject.
  • the method comprises contacting a plurality of cells in the subject with an agent that inhibits the expression or activity of MRCK1.
  • Suitable agents include polynucleotides (e.g., an antisense oligonucleotides of MRCK1), polypeptides (e.g., a dominant negative mutant of MRCK1), or polysaccharides, naturally-occurring target molecules of MRCK1 protein (e.g., an MRCK1 protein substrate or receptor), anti-MRCK1 antibodies, MRCK1 antagonists, or other small organic and inorganic molecule. They may also include vectors comprising polynucleotides encoding MRCK1 inhibitors or antisense sequences. Moreover, the agents can be anti-MRCK1 antibodies conjugated with therapeutic moieties. Suitable agents can be identified using the screening assays of the present invention.
  • compositions comprising an MRCK1 modulator and a pharmaceutically acceptable carrier.
  • a “pharmaceutically acceptable carrier” is intended to include any and all solvents, solubilizers, fillers, stabilizers, binders, absorbents, bases, buffering agents, lubricants, controlled release vehicles, diluents, emulsifying agents, humectants, lubricants, dispersion media, coatings, antibacterial or antifungal agents, isotonic and absorption delaying agents, and the like, compatible with pharmaceutical administration.
  • 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 agents can also be incorporated 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 (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 bisulfate; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose. 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 or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion.
  • suitable carriers include physiological saline, bacteriostatic water, Cremophor ELTM (BASF, Parsippany, N.J.) or phosphate buffered saline (PBS).
  • the injectable composition should be sterile and should be fluid to the extent that easy syringability 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 absorption of the injectable compositions can be brought about by including in the composition an agent which delays absorption, for example, aluminum monostearate and gelatin.
  • Sterile injectable solutions can be prepared by incorporating the active modulator (e.g., an anti-MRCK1 antibody, an MRCK1 activity inhibitor, or a gene therapy vector expressing antisense nucleotide to MRCK1) in the required amount in an appropriate solvent, followed by filtered sterilization.
  • the active modulator e.g., an anti-MRCK1 antibody, an MRCK1 activity inhibitor, or a gene therapy vector expressing antisense nucleotide to MRCK1
  • dispersions are prepared by incorporating the active compound into a sterile vehicle which contains a basic dispersion medium and the required other ingredients from those enumerated above.
  • the preferred methods of preparation are vacuum drying and freeze-drying which 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 purpose of oral therapeutic administration, the active compound can be incorporated 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 tragacanth 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 Stertes; 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 tragacanth 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 Stertes
  • 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 transdermal 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 bioactive 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 therapeutic moieties which may contain a bioactive compound, 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 e.g. Alza Corporation 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. Pat. No. 4,522,811.
  • Dosage unit form as used herein includes 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.
  • Toxicity and therapeutic efficacy of such compounds can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population).
  • the dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD50/ED50.
  • Compounds which exhibit large therapeutic indices are preferred. While compounds that exhibit toxic side effects may be used, care should be taken to design a delivery system that targets such compounds to the site of affected tissue in order to minimize potential damage to uninfected cells and, thereby, reduce side effects.
  • the data obtained from the cell culture assays and animal studies can be used in formulating a range of dosage for use in humans.
  • the dosage of such compounds lies preferably within a range of circulating concentrations that include the ED50 with little or no toxicity.
  • the dosage may vary within this range depending upon the dosage form employed and the route of administration utilized.
  • the therapeutically effective dose can be estimated initially from cell culture assays.
  • a dose may be formulated in animal models to achieve a circulating plasma concentration range that includes the IC50 (i.e., the concentration of the test compound which achieves a half-maximal inhibition of symptoms) as determined in cell culture.
  • IC50 i.e., the concentration of the test compound which achieves a half-maximal inhibition of symptoms
  • levels in plasma may be measured, for example, by high performance liquid chromatography.
  • compositions can be included in a container, pack, or dispenser together with instructions for administration.
  • kits for detecting the presence of an MRCK1 gene product in a biological sample comprises reagents for assessing expression of MRCK1 at mRNA or protein level.
  • the reagents include an antibody or fragment thereof, wherein the antibody or fragment specifically binds to MRCK1.
  • the kits may comprise a polynucleotide probe capable of specifically binding to a transcript of the MRCK1 gene.
  • the kit may also contain means for determining the amount of MRCK1 protein or mRNA in the test sample, and/or means for comparing the amount of MRCK1 protein or mRNA in the test sample to a control or standard.
  • the compound or agent can be packaged in a suitable container.
  • kits for assessing the suitability of each of a plurality of compounds for inhibiting MRCK1-related diseases in cells or human subjects include a plurality of compounds to be tested, and a reagent (such as an antibody specific to MRCK1 proteins, or a polynucleotide probe or primer capable of hybridizing to the MRCK1 gene) for assessing expression of MRCK1.
  • a reagent such as an antibody specific to MRCK1 proteins, or a polynucleotide probe or primer capable of hybridizing to the MRCK1 gene
  • Another aspect of the invention pertains to host cells into which a polynucleotide molecule of the invention is introduced, e.g., an MRCK1 gene or homolog thereof, within an expression vector, a gene delivery vector, or a polynucleotide molecule of the invention containing sequences which allow it to homologously recombine into a specific site of the host cell's genome.
  • a polynucleotide molecule of the invention e.g., an MRCK1 gene or homolog thereof, within an expression vector, a gene delivery vector, or a polynucleotide molecule of the invention containing sequences which allow it to homologously recombine into a specific site of the host cell's genome.
  • 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 to the progeny or potential progeny of such a cell. Because certain modifications may occur in succeeding generations due to either
  • a host cell can be any prokaryotic or eukaryotic cell.
  • an MRCK1 gene can be expressed in bacterial cells such as E. coli , insect cells, yeast or mammalian cells (e.g., Chinese hamster ovary cells (CHO), COS cells, Fischer 344 rat cells, HLA-B27 rat cells, HeLa cells, A549 cells, or 293 cells).
  • bacterial cells such as E. coli , insect cells, yeast or mammalian cells (e.g., Chinese hamster ovary cells (CHO), COS cells, Fischer 344 rat cells, HLA-B27 rat cells, HeLa cells, A549 cells, or 293 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 polynucleotide (e.g., DNA) into a host cell, including calcium phosphate or calcium chloride co-precipitation, DEAE-dextran-mediated transfection, lipofection, or electoporation.
  • a gene that encodes a selectable flag (e.g., resistance to antibiotics) is generally introduced into the host cells along with the gene of interest.
  • selectable flags include those which confer resistance to drugs, such as G418, hygromycin and methotrexate.
  • Polynucleotides encoding a selectable flag can be introduced into a host cell by the same vector as that encoding MRCK1 or can be introduced by a separate vector. Cells stably transfected with the introduced polynucleotide can be identified by drug selection (e.g., cells that have incorporated the selectable flag 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) MRCK1.
  • the invention further provides methods for producing MRCK1 using the host cells of the invention.
  • the method comprises culturing the host cell of invention (into which a recombinant expression vector containing an MRCK1 gene has been introduced) in a suitable medium such that MRCK1 is produced.
  • the method further comprises isolating MRCK1 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 MRCK1-coding sequences have been introduced.
  • Such host cells can then be used to create non-human transgenic animals in which exogenous sequences encoding MRCK1 have been introduced into their genome or homologous recombinant animals in which endogenous sequences encoding MRCK1 have been altered.
  • Such animals are useful for studying the function and/or activity of MRCK1 and for identifying and/or evaluating modulators of MRCK1 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, and the like.
  • a transgene is exogenous DNA which is integrated into the genome of a cell from which a transgenic animal develops and which 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” or “knockout animal” is a non-human animal, preferably a mammal, more preferably a mouse, in which an endogenous MRCK1 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 an MRCK1-encoding polynucleotide into the mate pronuclei of a fertilized oocyte, e.g., by microinjection or retroviral infection, and allowing the oocyte to develop in a pseudopregnant female foster animal.
  • Intronic sequences and polyadenylation signals can also be included in the transgene to increase the efficiency of expression of the transgene.
  • a tissue-specific regulatory sequence(s) can be operably linked to a transgene to direct expression of MRCK1 to particular cells.
  • a transgenic founder animal can be identified based upon the presence of a transgene of the invention in its genome and/or expression of mRNA corresponding to a gene of the invention 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 MRCK1 can further be bred to other transgenic animals carrying other transgenes.
  • a vector is prepared which contains at least a portion of a gene of the invention into which a deletion, addition or substitution has been introduced to thereby alter, e.g., functionally disrupt, the gene.
  • the gene can be a human gene, but more preferably, is a non-human homolog of a human gene of the invention (e.g., a homolog of the MRCK1 gene).
  • a mouse gene can be used to construct a homologous recombination polynucleotide molecule, e.g., a vector, suitable for altering an endogenous gene of the invention in the mouse genome.
  • the homologous recombination polynucleotide molecule is designed such that, upon homologous recombination, the endogenous gene of the invention is functionally disrupted (i.e., no longer encodes a functional protein; also referred to as a “knockout” vector).
  • the homologous recombination polynucleotide molecule can be designed such that, upon homologous recombination, the endogenous 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 MRCK1 gene).
  • the altered portion of the gene of the invention is flanked at its 5′ and 3′ ends by additional polynucleotide sequence of the gene of the invention to allow for homologous recombination to occur between the exogenous gene carried by the homologous recombination polynucleotide molecule and an endogenous gene in a cell, e.g., an embryonic stem cell.
  • the additional flanking polynucleotide sequence is of sufficient length for successful homologous recombination with the endogenous gene.
  • flanking DNA both at the 5′ and 3′ ends
  • the homologous recombination polynucleotide molecule is introduced into embryonic stem cells by electroporation.
  • the cells in which the introduced gene has homologously recombined with the endogenous gene are selected.
  • the selected cells can then be injected into a blastocyst of an animal (e.g., a mouse) to form aggregation chimeras.
  • 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 homologously recombined DNA.
  • Methods for constructing homologous recombination polynucleotide molecules, e.g., vectors, or homologous recombinant animals are well-known in the art.
  • transgenic non-human animals can be produced which contain selected systems which allow for regulated expression of the transgene.
  • a system is the cre/loxP recombinase system of bacteriophage P1.
  • Another example of a recombinase system is the FLP recombinase system of Saccharomyces cerevisiae (See e.g., O'Gorman et al., Science, 251:1351-1355, 1991). 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 encoding 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., Nature, 385:810-813, 1997, and PCT International Publication Nos. WO97/07668 and WO97/07669.
  • 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.
  • the nucleic acid sequence of MRCK1 is obtained from a newly developed genomic prediction pipeline. Briefly, the X-ray crystal structures of the catalytic domains of protein kinases were collected and aligned together according to their structural identity/similarities. The alignment was converted into a “scoring matrix” which carried the structural profile of the kinase catalytic domains. This scoring matrix was then used to search the Celera Human Genome database for sequences that have kinase catalytic domains.
  • MRCK alpha XP — 140553.1, 70% alignment to amino acid residues 40-1548 of MRCK1
  • two putative human MRCK-related proteins Entrez accession number Y12337, 100% alignment to amino acid residues 113-399 of MRCK1 and Entrez accession number U59305, 65% alignment to amino acid residues 40-434 of MRCK1.
  • the amino acid residues 339-398 of MRCK1 are highly homologous to the consensus sequence of an extension to ser/thr type protein kinase (accession number smart00133, 95.2% alignment, FIG. 6 ).
  • the amino acid residues 339-398 of MRCK1 also aligns to the consensus sequence of the protein kinase C terminal domain (accession number pfam00433, 91.0% alignment, FIG. 7 ).
  • the amino acid residues 882-920 of MRCK1 aligns to the consensus sequence of DAG_PE-binding domain (accession number pfam00130, 78.0% alignment, FIG. 8 ).
  • the amino acid residues 953-1060 of MRCK1 aligns to the consensus sequence of the Pleckstrin homology (PH) domain (accession number smart00233, 87.5% alignment, FIG. 9 ).
  • the amino acid residues 953-1060 of MRCK1 also aligns to another consensus sequence of the PH domain (accession number pfam00169, 88.0% alignment, FIG. 10 ).
  • the amino acid residues 1102-1345 of MRCK1 aligns to the consensus sequence of the CNH domain (accession number pfam00780, 85.7% alignment, FIG. 11 ).
  • the amino acid residues 1440-1471 of MRCK1 aligns to the consensus sequence of the P21-Rho-binding domain (accession number smart00285, 86.1% alignment, FIG. 12 ).
  • the amino acid residues 648-786 of MRCK1 also weakly aligns to the consensus sequence of the myosin tail (accession number pfam01576, 17.8% alignment, FIG. 13 ).
  • Pairwise BLAST program (BLASTN setting: Match: 1, Mismatch: ⁇ 2, gap open: 5, gap extension: 2, x_dropoff: 500, expect: 10.0, wordsize: 11, filter: unchecked; BALSTP setting: Matrix: BLOSUM62, gap open: 11, gap extension: 1, x_dropoff: 50, expect: 10.0, wordsize: 3, filter: unchecked) showed sequence homologies of 95% at nucleotide level and 90% at amino acid level between MRCK1 and PKIN-20.
  • the amino acid and nucleotide sequences of PKIN-20 are recited in SEQ ID NOS:4 and 5.
  • a human genome search was carried out using blastn program with Expect setting at 0.01, Filter setting at default, Descriptions setting at 100, and Alignment settings at 100.
  • the MRCK1 gene was mapped to or near loci 11p13 of human chromosome 11. Specifically, MRCK1 gene is located between genes LOC196204 and LOC143732, and overlaps with gene LOC196205.
  • the exons/introns in the MRCK1 gene were determined using the program “sim4” described by Florea et al., in “A computer program for aligning a cDNA sequence with a genomic DNA sequence” Genome Res. 8:967-974, 1998.
  • the hydrophobicity profile of MRCK1 sequence was generated using the GES (Goldman, Engelman and Steitz) hydrophobicity scale (Engelman et al., Ann. Rev. Biophys. Biophys. Chem. 15:321-353, 1986). Briefly, the GES scale is used to identify nonpolar transbilayer helices. The curve is the average of a residue-specific hydrophobicity scale over a window of 20 residues. When the line is in the upper half of the frame (positive), it indicates a hydrophobic region and when it is in the lower half (negative), a hydrophilic region.
  • the X-axis represents the length of the protein in amino acids (aa), while the Y-axis represents the GES score.
  • the curve line shows the GES pattern of the entire protein, while the straight line represents certain cutoff for potential membrane spanning domains.
  • the hydrophobicity profile indicates that MRCK1 is probably not a membrane protein.
  • the DNA probe for the MTN and MTA analysis is a PCR amplified 447 nucleotide fragment (SEQ ID NO:9) from human cDNA library.
  • the probe located at exon 29-33, pos#3898-4344 of the MRCK1 cDNA.
  • the PCR primers were designed based on the cDNA prediction.
  • the Forward primer sequence is: 5′ GCTGGCATCTACGTGGATG 3′ (SEQ ID NO:7).
  • the reverse primer sequence is: 5′ GTGGTTGAAGTTGGTAGGCG 3′ (SEQ ID NO:8).
  • the PCR amplified probe was sequence verified. The positions of the probe and the primers relative to the MRCK1 gene are shown in FIG. 15 .
  • the MTN analysis was performed using the Multiple Tissue Northern (MTN®) Blot Kit (Clontech Laboratories, Inc., Palo Alto, Calif.) under conditions specified in the User Manual. Briefly, the human MRCK1 probe was labeled with 32 P. The MTN Blot was prehybridized in ExpressHyb solution at 68° C. for 30 min, and then hybridized with the labeled probe at 68° C. for 1 hr. The blot was washed two times with Wash Solution 1 for 30 min at room temperature and two times with Wash Solution 2 for 30 min at 50° C., and was then exposed to X-ray film.
  • MTN® Multiple Tissue Northern
  • MTA analysis was performed using the Multiple Tissue Expression (MTE®) Array Kit (Clontech Laboratories, Inc., Palo Alto, Calif.) under conditions specified in the User Manual. Briefly, the MTE array was hybridized to 32 P-labeled human MRCK1 probe in ExpressHyb solution at 65° C. overnight with continuous agitation.
  • MTE® Multiple Tissue Expression
  • the array was washed four times with Wash Solution 1 for 20 min at 65° C. and two times with Wash Solution 2 for 20 min at 55° C., and was then exposed to X-ray film.
  • MRCK1 expression was found in all 76 tissues contained in the array.

Abstract

This invention provides compositions, organisms and methodologies employing a novel human protein kinase, MCRK1. The novel human kinase has sequence homology to rat myotonic dystrophy kinase-related Cdc42 binding kinase (MRCK) alpha. The gene encoding the novel kinase is localized in locus 11q13 of human chromosome 11. The novel protein kinase comprises multiple functional/structural domains that include a kinase domain, a pkinase_C domain, a DAG-PE binding domain, and a CNH domain. The sequence and structure similarity between the novel human protein and rat MRCK alpha indicates that the novel human protein may function as a downstream effector of Cdc42 in cytoskeleton reorganization.

Description

  • The present invention incorporates by reference U.S. Provisional Application Ser. No. 60/429,381, filed Nov. 27, 2002 and entitled “Compositions, Organisms and Methodologies Employing A Novel Human Kinase.”
  • FIELD OF THE INVENTION
  • The present invention relates to compositions, organisms and methodologies employing a novel human protein kinase, MRCK1, which has 65% sequence homology to rat myotonic dystrophy kinase-related Cdc42 binding kinase (MRCK). This invention can be used for diagnosing, prognosing and treating kinase-related diseases and, in particular, diseases associated with aberrant expression of MRCK1.
  • BACKGROUND OF THE INVENTION
  • Protein kinases regulate many different cell proliferation, differentiation, and signaling processes by adding phosphate groups to proteins. Uncontrolled signaling has been implicated in a variety of disease conditions including inflammation, cancer, arteriosclerosis, and psoriasis. Reversible protein phosphorylation is the main strategy for controlling activities of eukaryotic cells. It is estimated that more than 1,000 of the 10,000 proteins active in a typical mammalian cell are phosphorylated. As is well known in the art, the high energy phosphate, which drives activation, is generally transferred from adenosine triphosphate molecules (ATP) to a particular protein by protein kinases and removed from that protein by protein phosphatases. Phosphorylation occurs in response to extracellular signals (hormones, neurotransmitters, growth and differentiation factors, etc.), cell cycle checkpoints, and environmental or nutritional stresses. The phosphorylation process is roughly analogous to turning on a molecular switch. When the switch goes on, the appropriate protein kinase activates a metabolic enzyme, regulatory protein, receptor, cytoskeletal protein, ion channel or pump, or transcription factor.
  • The kinases comprise the largest known protein group, a superfamily of enzymes with widely varied functions and specificities. They are usually named after their substrate, their regulatory molecules, or some aspect of a mutant phenotype. With regard to substrates, the protein kinases may be roughly divided into two groups: those that phosphorylate tyrosine residues (protein tyrosine kinases, PTK) and those that phosphorylate serine or threonine residues (serine/threonine kinases, STK). A few protein kinases have dual specificity and phosphorylate threonine and tyrosine residues. Almost all kinases contain a similar 250-300 amino acid catalytic domain. The primary structure of the kinase domains is conserved and can be further subdivided into 11 subdomains. The N-terminal of the kinase domain, which contains subdomains I-IV, generally folds into a lobe-like structure that binds and orients the ATP (or GTP) donor molecule. The C terminal of the kinase domain forms a larger lobe, which contains subdomains VI-XI, binds the protein substrate and carries out the transfer of the gamma phosphate from ATP to the hydroxyl group of a serine, threonine, or tyrosine residue. Subdomain V spans the two lobes. Each of the 11 subdomains contains specific residues and motifs or patterns of amino acids that are characteristic of that subdomain and are highly conserved.
  • The kinases may be categorized into families by the different amino acid sequences (generally between 5 and 100 residues) located on either side of, or inserted into loops of, the kinase domain. These added amino acid sequences allow the regulation of each kinase as it recognizes and interacts with its target protein.
  • The presence or absence of a phosphate moiety modulates protein function in multiple ways. A common mechanism involves changes in the catalytic properties (Vmax and Km) of an enzyme, leading to its activation or inactivation.
  • A second widely recognized mechanism involves promoting protein-protein interactions. An example of this is the tyrosine autophosphorylation of the ligand-activated EGF receptor tyrosine kinase. This event triggers the high-affinity binding to the phosphotyrosine residue on the receptor's C-terminal intracellular domain to the SH2 motif of an adaptor molecule Grb2. Grb2, in turn, binds through its SH3 motif to a second adaptor molecule, such as SHC. The formation of this ternary complex activates the signaling events that are responsible for the biological effects of EGF. Serine and threonine phosphorylation events also have been recently recognized to exert their biological function through protein-protein interaction events that are mediated by the high-affinity binding of phosphoserine and phosphothreonine to the WW motifs present in a large variety of proteins.
  • A third important outcome of protein phosphorylation is changes in the subcellular localization of the substrate. As an example, nuclear import and export events in a large diversity of proteins are regulated by protein phosphorylation.
  • Many kinases are involved in regulatory cascades wherein their substrates may include other kinases whose activities are regulated by their phosphorylation state. Ultimately the activities of some downstream effectors are modulated by phosphorylation resulting from activation of such a pathway.
  • Myotonic dystrophy kinase-related Cdc42 binding kinases (MRCKs) are serine/threonine kinases. MRCKs have been implicated in the morphological activities of Cdc42 in non-neural cells and are suggest to be downstream effectors of Cdc42 in cytoskeletal reorganization. At least two types of MRCKs, MRCK alpha and MRCK beta, have been identified. MRCKs interact with the GTP-bound form of Cdc42 and, to a lesser extent, the GTP-bound form of Rac. The catalytic domain of MRCKs phosphorylates non-muscle myosin light chain 2 at serine 19. The phosphorylation is believed to be involved in myosin contractile activity and associated changes in the organization of actin microfilaments in intact cells.
  • MRCK alpha and Rho-binding kinase (ROK) alpha are believed to have contrasting roles in regulating neurite morphology. ROK alpha acts downstream of RhoA in inducing neurite retraction, while MRCK alpha acts downstream of Cdc42/Rac1 in promoting neurite outgrowth. The neurite outgrowth induced by either kinase-dead ROK alpha or nerve growth factor can be effectively blocked by a kinase-dead and p21-binding deficient MRCK alpha mutant. In addition, expression of kinase-dead MRCK alpha blocks Cdc42V12-dependent formation of focal complexes and peripheral microspikes. Microinjection of a plasmid encoding MRCK alpha results in actin and myosin reorganization.
  • MRCKs have multiple functional domains. These domains include three coiled-coil alpha-helix domains, a cysteine-rich motif resembling those of protein kinase C and n-chimaerin, and a Pleckstrin homology domain. Native MRCK kinases tend to form high-molecular-weight multimers. The intermolecular interactions among the three coiled-coil domains and the N-terminal region preceding the kinase domain in MRCK alpha are believed to be responsible for the multimerization.
  • MRCK alpha can be activated by the N-terminus-mediated dimerization. The dimerization leads to trans-autophosphorylation of MRCK kinases. In addition, MRCK alpha kinases can be negatively regulated through intramolecular interactions between the two distal coiled-coil domains. Deletion of these coiled-coil domains leads to a more active kinase, showing the negative autoregulatory role of these domains. The N-terminus-mediated dimerization and the intramolecular interaction between the distal coiled-coil domains are considered to be two mutually exclusive events, which regulate the catalytic state of MRCK kinases.
  • SUMMARY OF THE INVENTION
  • The present invention discloses compositions, organisms and methodologies employing a novel human protein kinase. The new human protein kinase shares sequence homology with rat MRCK alpha. The gene encoding the new protein is localized at 11q13.1 in human chromosome 11. This new gene is hereinafter referred to as the MRCK1 gene, and its encoded protein(s) is referred to as MRCK1 or MRCK1 kinase. MRCK1 has multiple domains including at least a kinase domain, a protein kinase C terminal domain, a myosin tail domain, a phorbol esters/diacylglycerol binding domain (DAG_PE binding domain), a Pleckstrin homology domain, a CNH domain, and a P21-Rho-binding domain. The sequence and structure similarity between MRCK1 and rat MRCK alpha indicates that MRCK1 may be involved in Cdc42-mediated cytoskeleton reorganization in human cells.
  • The kinase domain in MRCK1 shows 100% sequence alignment with the consensus sequences of the catalytic domains of at least two subfamilies of protein kinases. The utility of various kinase domains is known in the art. The utilities of other domains, such as protein kinase C terminal domains, Phorbol esters/diacylglycerol binding domains, Pleckstrin homology domains, CNH domains, and P21-Rho-binding domains are also known in the art. The unique peptide sequences, and nucleic acid sequences that encode the peptides, can be used as models for the development of human therapeutic targets, aid in the identification of therapeutic proteins, and serve as targets for the development of human therapeutic agents that modulate kinase activity in cells and tissues that express the kinase.
  • In one aspect, the invention provides isolated polynucleotides comprising a nucleotide sequence encoding MRCK1 or a variant of MRCK1.
  • In another aspect, the invention provides isolated polypeptides comprising the amino acid sequence of MRCK1 or a variant of MRCK1.
  • In yet another aspect, the invention provides agents that modulate expression levels of the MRCK1 gene or an activity of MRCK1.
  • The invention also provides methods for (a) detecting polynucleotides comprising a nucleotide sequence encoding MRCK1 or a variant of MRCK1 and (b) detecting polypeptides comprising an amino acid sequence of MRCK1 or a variant of MRCK1 in a biological sample.
  • The invention further provides methods for screening agents that modulate expression level of the MRCK1 gene or an activity of MRCK1.
  • The invention further provides cell lines harboring the MRCK1 gene, animals transgenic for the MRCK1 gene, and animals with interrupted MRCK1 gene (MRCK1 knockout animals). These cell lines and animals can be used to study the functions of MRCK1.
  • In still another aspect, the invention provides polynucleotides capable of inhibiting MRCK1 gene expression by RNA interference.
  • The invention further provides methods of inhibiting MRCK1 gene expression by introducing siRNAs or other RNAi sequences into target cells.
  • The preferred embodiments of the invention are described below in the Detailed Description of the Invention. Unless specifically noted, it is intended that the words and phrases in the specification and claims be given the ordinary and accustomed meaning to those of ordinary skill in the applicable art or arts. If any other meaning is intended, the specification will specifically state that a special meaning is being applied to a word or phrase.
  • It is further intended that the invention not be limited only to the specific structure, material or methods that are described in the preferred embodiments, but include any and all structures, materials or methods that perform the claimed function, along with any and all known or later-developed equivalent structures, materials or methods for performing the claimed function.
  • Further examples exist throughout the disclosure, and it is not applicant's intention to exclude from the scope of the invention the use of structures, materials, or methods that are not expressly identified in the specification, but nonetheless are capable of performing the claimed function.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • The inventions of this application are better understood in conjunction with the following drawings, in which:
  • FIG. 1 is the sequence alignment between a fragment of MRCK1 and its corresponding sequence in rat MRCK alpha.
  • FIG. 2 depicts the sequence alignment between another fragment of MRCK1 and its corresponding sequence in rat MRCK alpha.
  • FIG. 3 compares MRCK1's kinase domain to the catalytic domain of a family of Ser/Thr protein kinases.
  • FIG. 4 compares MRCK1 's kinase domain to the kinase domain of another family of protein kinases.
  • FIG. 5 shows the sequence alignment between the kinase domain of MRCK1 and the catalytic domain of a family of tyrosine kinases.
  • FIG. 6 illustrates the sequence alignment between amino acid residues 339 to 398 of MRCK1 and the extension to Ser/Thr-type protein kinases
  • FIG. 7 compares amino acid residues 339 to 398 of MRCK1 to a protein kinase C terminal domain.
  • FIG. 8 shows the sequence alignment between the amino acid residues 882-920 of MRCK1 and a consensus sequence of the DAG_PE bind domains of other proteins.
  • FIG. 9 depicts the sequence alignment between the amino acid residues 953-1060 of MRCK1 and a consensus sequence of the PH bind domains.
  • FIG. 10 illustrates the sequence alignment between the amino acid residues 954-1060 of MRCK1 and another consensus sequence of the PH bind domain.
  • FIG. 11 compares the amino acid residues 1102-1345 of MRCK1 to a consensus sequence of the CNH domains.
  • FIG. 12 shows the sequence alignment between the amino acid residues 1440-1471 of MRCK1 and a consensus sequence of the P21-Rho-binding domains.
  • FIG. 13 compares the amino acid residues 648-786 of MRCK1 to a consensus sequence of the myosin tail domains.
  • FIG. 14 shows the hydrophobicity profile of MRCK1.
  • FIG. 15 shows the position and nucleotide sequence of human MRCK1 probe and PCR primers used for the amplification of the probe sequences.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The following detailed description is presented to enable any person skilled in the art to make and use the invention. For purposes of explanation, specific nomenclature is set forth to provide a thorough understanding of the present invention. However, it will be apparent to one skilled in the art that these specific details are not required to practice the invention. Descriptions of specific applications are provided only as representative examples. Various modifications to the preferred embodiments will be readily apparent to one skilled in the art, and the general principles defined herein may be applied to other embodiments and applications without departing from the scope of the invention. The present invention is not intended to be limited to the embodiments shown, but is to be accorded the widest possible scope consistent with the principles and features disclosed herein.
  • The present invention is based on the sequence information obtained from a newly-developed genomic prediction pipeline. Briefly, the X-ray crystal structures of the catalytic domains of protein kinases were collected and aligned together according to their structural identity/similarities. The alignment was converted into a “scoring matrix” which carried the structural profile of the kinase catalytic domains. This scoring matrix was then used to search the Celera Human Genome database and pull out sequences that have kinase catalytic domains.
  • Based on this analysis, the present invention provides the amino acid sequence of a human kinase peptide that is highly homologous to rat myotonic dystrophy kinase-related Cdc42 binding kinase (MRCK), cDNA sequences and genomic sequences that encode the kinase peptide, and information about the closest art known protein/peptide/domain that has structural or sequence homology to the kinase of the present invention.
  • In addition to being previously unknown, the peptide of the present invention is selected based on its ability to be used for the development of commercially important products and services. Specifically, the present peptides are selected based on homology to a known kinase protein of rat MRCK alpha.
  • Various aspects of the invention are described in detail in the following subsections. It should of course be understood that the use of subsections is not meant to limit the invention. Rather, each subsection applies to any aspect of the invention, as is appropriate.
  • DEFINITIONS AND TERMS
  • To facilitate the understanding of the present invention, a number of terms and phrases are defined below:
  • As used herein, a polynucleotide or a polypeptide is “isolated” if it is removed from its native environment. For instance, a polynucleotide or a polypeptide is isolated through a purification process such that the polynucleotide or polypeptide is substantially free of cellular material or free of chemical precursors. The polynucleotide/polypeptide of the present invention can be purified to homogeneity or other degrees of purity. The level of purification will be based on the intended use. As appreciated by one of ordinary skill in the art, a polynucleotide/polypeptide can perform its desired function(s) even in the presence of considerable amounts of other components or molecules.
  • In some uses, a polynucleotide/polypeptide that is “substantially free of cellular material” includes preparations which have less than about 30% (by weight) other polynucleotides/polypeptides including contaminating polynucleotides/polypeptides. For instance, the preparations can have less than about 20%, less than about 10%, or less than about 5% other polynucleotides/polypeptides. If a polynucleotide/polypeptide preparation is recombinantly produced, it can be substantially free of culture medium, i.e., culture medium components representing less than about 20% by weight of the polynucleotide/polypeptide preparation.
  • The language “substantially free of chemical precursors” includes preparations in which the polynucleotide/polypeptide is separated from chemical precursors or other chemicals that are involved in the synthesis of the polynucleotide/polypeptide. In one embodiment, the language “substantially free of chemical precursors” includes kinase preparations having less than about 30% (by weight), less than about 20% (by weight), less than about 10% (by weight), or less than about 5% (by weight) of chemical precursors or other chemicals used in the synthesis.
  • A “polynucleotide” can include any number of nucleotides. For instance, a polynucleotide can have at least 10, 20, 25, 30, 40, 50, 100 or more nucleotides. A polynucleotide can be DNA or RNA, double-stranded or single-stranded. A polynucleotide encodes a polypeptide if the polypeptide is capable of being transcribed and/or translated from the polynucleotide. Transcriptional and/or translational regulatory sequences, such as promoter and/or enhancer(s), can be added to the polynucleotide before said transcription and/or translation occurs. Moreover, if the polynucleotide is singled-stranded, the corresponding double-stranded DNA containing the original polynucleotide and its complementary sequence can be prepared before said transcription and/or translation.
  • As used herein, “a variant of a polynucleotide” refers to a polynucleotide that differs from the original polynucleotide by one or more substitutions, additions, and/or deletions. For instance, a variant of a polynucleotide can have 1, 2, 5, 10, 15, 20, 25 or more nucleotide substitutions, additions or deletions. Preferably, the modification(s) is in-frame, i.e., the modified polynucleotide can be transcribed and translated to the original or intended stop codon. If the original polynucleotide encodes a polypeptide with a biological activity, the polypeptide encoded by a variant of the original polynucleotide substantially retains such activity. Preferably, the biological activity is reduced/enhanced by less than 50%, or more preferably, less than 20%, relative to the original activity.
  • A variant of a polynucleotide can be a polynucleotide that is capable of hybridizing to the original polynucleotide, or the complementary sequence thereof, under reduced stringent conditions, preferably stringent conditions, or more preferably, highly stringent conditions. Examples of conditions of different stringency are listed in Table 1. Highly stringent conditions are those that are at least as stringent as conditions A-F; stringent conditions are at least as stringent as conditions G-L; and reduced stringency conditions are at least as stringent as conditions M-R. As used in Table 1, hybridization is carried out under a given hybridization condition for about 2 hours, followed by two 15-minute washes under the corresponding washing condition(s).
  • TABLE 1
    Stringency Conditions
    Stringency Polynucleotide Hybrid Hybridization Wash Temp.
    Condition Hybrid Length (bp)1 Temperature and BufferH and BufferH
    A DNA:DNA >50 65° C.; 1xSSC -or- 65° C.; 0.3xSSC
    42° C.; 1xSSC, 50% formamide
    B DNA:DNA <50 TB*; 1xSSC TB*; 1xSSC
    C DNA:RNA >50 67° C.; 1xSSC -or- 67° C.; 0.3xSSC
    45° C.; 1xSSC, 50% formamide
    D DNA:RNA <50 TD*; 1xSSC TD*; 1xSSC
    E RNA:RNA >50 70° C.; 1xSSC -or- 70° C.; 0.3xSSC
    50° C.; 1xSSC, 50% formamide
    F RNA:RNA <50 TF*; 1xSSC TF*; 1xSSC
    G DNA:DNA >50 65° C.; 4xSSC -or- 65° C.; 1xSSC
    42° C.; 4xSSC, 50% formamide
    H DNA:DNA <50 TH*; 4xSSC TH*; 4xSSC
    I DNA:RNA >50 67° C.; 4xSSC -or- 67° C.; 1xSSC
    45° C.; 4xSSC, 50% formamide
    J DNA:RNA <50 TJ*; 4xSSC TJ*; 4xSSC
    K RNA:RNA >50 70° C.; 4xSSC -or- 67° C.; 1xSSC
    50° C.; 4xSSC, 50% formamide
    L RNA:RNA <50 TL*; 2xSSC TL*; 2xSSC
    M DNA:DNA >50 50° C.; 4xSSC -or- 50° C.; 2xSSC
    40° C.; 6xSSC, 50% formamide
    N DNA:DNA <50 TN*; 6xSSC TN*; 6xSSC
    O DNA:RNA >50 55° C.; 4xSSC -or- 55° C.; 2xSSC
    42° C.; 6xSSC, 50% formamide
    P DNA:RNA <50 TP*; 6xSSC TP*; 6xSSC
    Q RNA:RNA >50 60° C.; 4xSSC -or- 60° C.; 2xSSC
    45° C.; 6xSSC, 50% formamide
    R RNA:RNA <50 TR*; 4xSSC TR*; 4xSSC
    1The hybrid length is that anticipated for the hybridized region(s) of the hybridizing polynucleotides. When hybridizing a polynucleotide to a target polynucleotide of unknown sequence, the hybrid length is assumed to be that of the hybridizing polynucleotide. When polynucleotides of known sequence are hybridized, the hybrid length can be determined by aligning the sequences of the polynucleotides and identifying the region or regions of optimal sequence complementarity.
    HSSPE (1xSSPE is 0.15M NaCl, 10 mM NaH2PO4, and 1.25 mM EDTA, pH 7.4) can be substituted for SSC (1xSSC is 0.15M NaCl and 15 mM sodium citrate) in the hybridization and wash buffers.
    TB* − TR*The hybridization temperature for hybrids anticipated to be less than 50 base pairs in length should be 5-10° C. less than the melting temperature (Tm) of the hybrid, where Tm is determined according to the following equations. For hybrids less than 18 base pairs in length, Tm(° C.) = 2(# of A + T bases) + 4(# of G + C bases). For hybrids between 18 and 49 base pairs in length, Tm(° C.) = 81.5 + 16.6(log10Na+) + 0.41(% G + C) − (600/N), where N is the number of bases in the hybrid, and Na+ is the concentration of sodium ions in the hybridization buffer (Na+ for 1xSSC = 0.165M).
  • It will be appreciated by those of ordinary skill in the art that, as a result of the degeneracy of the genetic code, there are many polynucleotide variants that encode the same polypeptide. Some of these polynucleotide variants bear minimal sequence homology to the original polynucleotide. Nonetheless, polynucleotides that vary due to differences in codon usage are specifically contemplated by the present invention.
  • As used herein, a “polypeptide” can include any number of amino acid residues. For instance, a polypeptide can have at least 5, 10, 15, 20, 30, 40, 50 or more amino acid residues.
  • As used herein, a “variant of a polypeptide” is a polypeptide that differs from the original polypeptide by one or more substitutions, deletions, and/or insertions. Preferably, these modifications do not substantially change (e.g. reduce or enhance) the original biological function of the polypeptide. For instance, a variant can reduce or enhance or maintain the biological activities of the original polypeptide. Preferably, the biological activities of the variant are reduced or enhanced by less than 50%, or more preferably, less than 20%, relative to the original polypeptide.
  • Similarly, the ability of a variant to react with antigen-specific antisera can be enhanced or reduced by less than 50%, preferably less than 20%, relative to the original polypeptide. These variants can be prepared and evaluated by modifying the original polypeptide sequence and then determining the reactivity of the modified polypeptide with the antigen-specific antibodies or antisera.
  • Preferably, a variant polypeptide contains one or more conservative substitutions. A “conservative substitution” is one in which an amino acid is substituted for another amino acid which has similar properties, such that one skilled in the art would expect that the secondary structure and hydropathic nature of the substituted polypeptide will not be substantially changed. Conservative amino acid substitutions can be made on the basis of similarity in polarity, charge, solubility, hydrophobicity, hydrophilicity and/or the amphipathic nature of the residues. Negatively charged amino acids include aspartic acid and glutamic acid, and positively charged amino acids include lysine and arginine. Amino acids having uncharged polar head groups and similar hydrophilicity values include leucine, isoleucine and valine, or glycine and alanine, or asparagine and glutamine, or serine, threonine, phenylalanine and tyrosine. Other groups of amino acids that can produce conservative changes include: (1) ala, pro, gly, glu, asp, gln, asn, ser, thr; (2) cys, ser, tyr, thr; (3) val, ile, leu, met, ala, phe; (4) lys, arg, his; and (5) phe, tyr, trp, his. A polypeptide variant can also contain nonconservative changes.
  • Polypeptide variants can be prepared by the deletion and/or addition of amino acids that have minimal influence on the biological activity, immunogenicity, secondary structure and/or hydropathic nature of the polypeptide. Variants can be for instance by substituting, modifying, deleting or adding one or more amino acids residues in the original sequence. Polypeptide variants preferably exhibit at least 96%, more preferably at least 97%, and most preferably at least 98% sequence homology to the original polypeptide.
  • Polypeptide variants include polypeptides that are modified from the original polypeptides either by a natural process, such as a post-translational modification, or by a chemical modification. These modifications are well-known in the art. Modifications can occur anywhere in the polypeptide, including the backbone, the amino acid side-chains and the amino or carboxyl termini. It will be appreciated that the same type of modification can be present in the same or varying degrees at several sites in a given polypeptide. Also, a given polypeptide can contain many types of modifications. Polypeptides may be branched, for example, as a result of ubiquitination, and they may be cyclic, with or without branching. Cyclic, branched, and branched cyclic polypeptides can result from natural post-translational processes or be made through synthetic methods. Suitable modifications for this invention include acetylation, acylation, ADP-ribosylation, amidation, covalent attachment of flavin, covalent attachment of a heme moiety, covalent attachment of a nucleotide or nucleotide derivative, covalent attachment of a lipid or lipid derivative, covalent attachment of phosphatidylinositol, cross-linking, cyclization, disulfide bond formation, demethylation, formation of covalent cross-links, formation of cysteine, formation of pyroglutamate, formylation, gamma-carboxylation, glycosylation, GPI anchor formation, hydroxylation, iodination, methylation, myristoylation, oxidation, pegylation, proteolytic processing, phosphorylation, prenylation, racemization, selenoylation, sulfation, transfer-RNA mediated addition of amino acids to proteins such as arginylation, and ubiquitination.
  • As used herein, the term “modulation” includes up-regulation, induction, stimulation, potentiation, inhibition, down-regulation or suppression, or relief of inhibition.
  • A nucleotide sequence is “operably linked” to another nucleotide sequence if the two sequences are placed into a functional relationship. For example, a coding sequence is operably linked to a 5′ regulatory sequence if the 5′ regulatory sequence can initiate transcription of the coding sequence in an in vitro transcription/translation system or in a host cell. “Operably linked” does not require that the DNA sequences being linked are contiguous to each other. Intervening sequences may exist between two operably linked sequences.
  • As used herein, a “disease-free” human refers to a human who does not have MRCK1-related diseases. Disease-free, tissues or samples refer to cells, tissues or samples obtained from disease-free human(s).
  • A polynucleotide is “capable of hybridizing” to a gene if the polynucleotide can hybridize to at least one of the following sequences: (1) the sequence of an RNA transcript of the gene, (2) the complementary sequence of an RNA transcript of the gene, (3) the cDNA sequence of an RNA transcript of the gene, (4) the complementary sequence of the cDNA sequence of an RNA transcript of the gene, (5) a genomic sequence of the gene, and (6) the complementary sequence of a genomic sequence of the gene.
  • As used herein, sequence “identity” or “percentage alignment” in an alignment can be determined by the standard protein-protein or nucleotide-nucleotide BLAST programs (i.e., blastp or blastn, respectively). Sequence identity can also be determined by the BLAST2 program. Suitable BLAST program can be found at the BLAST web site maintained by the National Center of Biotechnology Information (NCBI) (National Library of Medicine)
  • Human MRCK1 Gene and MRCK1 Kinase
  • The present invention identifies a new human gene (MRCK1 gene) that encodes a protein that is highly homologous to rat MRCK alpha. The nucleotide and amino acid sequences of the protein encoded by the MRCK1 gene are depicted in SEQ ID NOS:1 and 2, respectively. FIGS. 1 and 2 show the sequence alignment between SEQ ID NO:2 (denoted as “Query”) and the amino acid sequence of rat MRCK alpha (denoted as “Sbjct”). The alignment is a result of blast search of the “all non-redundant GenBank CDS database” in Entrez. The blast search uses BLASTP 2.2.3 algorithm [Apr. 24, 2002], which is described in Altschul et al., Nucleic Acids Res., 25: 3389-3402 (1997).
  • FIG. 1 shows that the sequence consisting of amino acid residues 40 to 434 of MRCK1 (Query) and the sequence consisting of amino acid residues 46 to 463 of rat MRCK alpha (Sbjct) have 65% sequence identity with 78% positives, a score of 588 bits (1516) and an E value of 1×10−172. FIG. 2 shows that the sequence consisting of amino acid residues 551 to 1457 of MRCK1 (Query) and the sequence consisting of amino acid residues 684 to 1588 of rat MRCK alpha (Sbjct) have 38% sequence identity with 54% positives, a score of 615 bits (1586) and an E value of 1×10−166. The rat MRCK alpha sequence used in the alignment has Entrez Database accession number AAC02941.1 or NP446109.1.
  • The same approach is used to compare MRCK1 to a predicted mouse protein, which is similar to Ser/Thr protein kinase related to the myotonic dystrophy protein kinase. The sequence of the mouse protein has Entrez Database accession number XP140553.1. Alignment shows that amino acid residues 40 to 1574 of the mouse sequence (Sbjct) and the sequence consisting of amino acid residues 40 to 1548 of MRCK1 (Query) have 70% sequence identity with 73% positives, a score of 1981 bits (5133) and an E value of 0.0.
  • The same blast search also identifies sequence similarity between MRCK1 and other proteins. These proteins include, but are not limited to, human Cdc42-binding protein kinase beta (Entrez accession number: NM006035) and C. elegans serine/threonine-protein kinase (Entrez accession number: NM072198).
  • In addition, the kinase domain of MRCK1 (including amino acid residues 77 to 337, see below) has sequence homology to the catalytic domains of various protein kinases. These protein kinases include, but are not limited to, human myotonic dystrophy kinase (Entrez accession number: AAC14450.1 or L08835, 62% sequence identity), human dystrophia myotonica-protein (Entrez accession number: NM 004409, 61% sequence identity), human myotonin-protein kinase, Form VI (Entrez accession number: AAA75239.1 or L00727, 62% sequence identity), and human myotonin-protein kinase, Form VIII (Entrez accession number: AAA75237.1 or L00727, 62% sequence identity).
  • The sequence consisting of amino acid residues 113 to 399 of MRCK1 (Query) also aligns to the amino acid sequence having Entrez Protein Database accession number CAA73006.1 (Sbjct). These two sequences are 100% identical to each other. Sequence CAA73006.01 was disclosed in Kedra et al., Hum. Genet. 100: 611-619 (1997), and is localized to 11q13 in human chromosome 11.
  • MRCK1 gene is also localized near or at 11q13 in human chromosome 11. Specifically, the MRCK1 gene is located between genes LOC256612 and EHD1, and overlaps with gene LOC196205. The MRCK1 gene encompasses nucleotides 979139 to 999235 in human chromosome 11. The nucleotide numbering in human chromosome 11 is based on Entrez Human Genome Sequence Database maintained by NCBI. The minus-strand sequence of human chromosome 11 that consists of nucleotides 979139 to 999235 is shown in SEQ ID NO:3. The genomic sequence in SEQ ID NO:3 is listed from 5′ to 3′, i.e. from nucleotide 999235 to nucleotide 979139 in the minus strand of human chromosome 11.
  • Human chromosome loci near or at 11q13 harbor multiple disease-related genes. These diseases include insulin-dependent diabetes mellitus, familial paraganglioma type 2, spinocerebellar ataxia type 5, Bardet-Biedl syndrome, and multiple endocrine neoplasia type 1. In addition, there is a report of a translocation, t(11;17), at this loci in B-cell non-Hodgkin's lymphoma.
  • Human MRCK1 gene has at least 35 exons. Table 2 lists the location of each of these 35 exons in the genomic sequence SEQ ID NO:3. SEQ ID NO:1 shows an MRCK1-coding sequence produced by fusing the 35 exons in consecutive order. Translation of SEQ ID NO:1 produces the amino acid sequence SEQ ID NO:2. Table 2 also illustrates the corresponding location of each exon in the MRCK1-coding sequence SEQ ID NO:1.
  • TABLE 2
    Exons in Human MRCK1 Gene
    Corresponding Sequence in SEQ
    Exon ID NO: 3 Comprised in Human Corresponding Sequence in
    Numbers Chromosome 11 SEQ ID NO: 1
    1  1-160  1-160
    2 2,666-2,757 161-252
    3 2,837-2,920 253-336
    4 3,922-4,017 337-432
    5 4,302-4,450 433-581
    6 4,999-5,092 582-675
    7 5,337-5,537 676-876
    8 5,668-5,916   877-1,125
    9 6,408-6,487 1,126-1,205
    10 7,551-7,648 1,206-1,303
    11 7,692-7,831 1,304-1,443
    12 8,276-8,455 1,444-1,623
    13 8,721-8,810 1,624-1,713
    14 8,951-9,072 1,714-1,835
    15 9,153-9,225 1,836-1,908
    16 9,419-9,525 1,909-2,015
    17 9,609-9,723 2,016-2,130
    18  9,995-10,143 2,131-2,279
    19 10,218-10,306 2,280-2,368
    20 10,595-10,647 2,369-2,421
    21 10,736-10,834 2,422-2,520
    22 10,913-10,972 2,521-2,580
    23 11,182-11,341 2,581-2,740
    24 11,613-11,746 2,741-2,874
    25 11,829-11,968 2,875-3,014
    26 12,063-12,144 3,015-3,096
    27 12,851-13,067 3,097-3,313
    28 14,317-14,379 3,314-3,376
    29 14,503-15,102 3,377-3,976
    30 16,786-16,883 3,977-4,074
    31 16,958-17,042 4,075-4,159
    32 17,171-17,291 4,160-4,280
    33 17,402-17,519 4,281-4,398
    34 17,857-18,099 4,399-4,641
    35 20,041-20,097 4,642-4,698
  • MRCK1 kinase depicted by SEQ ID NO:2 comprises multiple structural/functional domains. These structural/functional domains include at least a kinase domain (comprising amino acid residues 71 to 337), a protein kinase C terminal domain (comprising amino acid residues 339 to 398), a myosin tail domain (comprising amino acid residues 648 to 786), a DAG_PE binding domain (comprising amino acid residues 882 to 920), a Pleckstrin homology domain (comprising amino acid residues 953 to 1060), a CNH domain (comprising amino acid residues 1102 to 1345), and a P21-Rho-binding domain (comprising amino acid residues 1440 to 1471).
  • FIGS. 3, 4, and 5 illustrate the sequence alignments between MRCK1's kinase domain and various protein kinase domains. As used in other figures of this invention, “Query” denotes to the sequence of MRCK1, and “Sbjct” refers to the sequence being compared to the MRCK1 sequence.
  • FIG. 3 is the alignment between MRCK1's kinase domain and the consensus sequence of the catalytic domains of a subfamily of Serine/Threonine protein kinases. This subfamily includes C-Jun N-terminal kinase (JNK3), abelson tyrosine kinase, a calmodulin-binding, vesicle-associated, protein kinase-like protein (1G5), serine/threonine-protein kinase prp4, Cdc2/Cdc28 subfamily of Ser/Thr protein kinases in Caenorhabditis elegans, and ribosomal S6 kinase of C elegans. The consensus sequence has CD NO: smart00220.4, S_TKc, and can be retrieved from the Conserved Domain Database maintained by NCBI. The alignment was performed using standard protein-protein BLAST (blastp) algorithm provided by NCBI. MRCK1's kinase domain has 100% sequence identities to the consensus sequence smart00220.4, with a score of 263 bits and an E value of 3×10−71.
  • FIG. 4 shows the alignment between MRCK1 's kinase domain and the consensus sequence of the catalytic domains of another subfamily of protein kinases. This subfamily includes protein kinase Ck2, wee1-like protein kinase (WEE1hu), and tyrosine-protein kinase RYK. The consensus sequence has CD NO: pfam00069.4, pkinase. MRCK1's kinase domain has 100% sequence identities to pfam00069.4, with a score of 213 bits and an E value of 2×10−56.
  • FIG. 5 shows the alignment between MRCK1's kinase domain and the catalytic domain (CD NO: smart00219.4, TyrKc) of a subfamily of tyrosine kinases. This subfamily includes the tyrosine kinase domain of fibroblast growth factor receptor 1, tyrosine-protein kinase (KIN15/KIN16 subfamily), and a Drosophila receptor protein-tyrosine kinase family member (drl-P1). The amino acid residues 72-303 in MRCK1's kinase domain has 89.1% sequence identities to smart00219.4, with a score of 112 bits and an E value of 9×10−26.
  • FIG. 6 illustrates the sequence alignment between the amino acid residues 339-398 of MRCK1 and the consensus sequence for the extension to a family of Ser/Thr-type protein kinases. The consensus sequence has CD NO: smart00133.4, S_TK_X. This family of protein kinases includes cAMP-Dependent protein kinase, protein kinase cek1, and cell cycle protein kinase DBF2. The two sequences share 95.2% sequence identities with a score of 57.7 bits and an E value of 3×10−9.
  • The amino acid residues 339-398 of MRCK1 further aligned to a consensus sequence (CD NO: pfam00433.4, pkinase_C) of the protein kinase C terminal domain. FIG. 7 shows 91% sequence identities between these two sequences. The alignment has a score of 40.6 bits and an E value of 4×10−04. Ribosomal protein S6 kinase (S6K), serine/threonine-protein kinase YPK1, and protein kinase C, zeta type (NPKC-ZETA) share the consensus sequence pfam00433.4, pkinase_C.
  • FIG. 8 demonstrates the sequence alignment between the amino acid residues 882-920 and a consensus sequence of the DAG_PE binding domains. The two sequences show 78% sequence identities with a score of 51.9 bits and an E value of 2×10−07. The CD number for the consensus sequence is pfam00130.4. The DAG_PE binding domain is also known as the protein kinase C conserved region 1 (C1 or cysteine-rich) domain.
  • In addition, MRCK1 shows sequence homology to the Pleckstrin homology domain (PH domain). PH domains are commonly found in eukaryotic signaling proteins. The domain family possesses multiple functions including the abilities to bind inositol phosphates and various other proteins. PH domains have been found to possess inserted domains (such as in PLC gamma, syntrophins) and/or to be inserted within other domains. Mutations in Brutons tyrosine kinase (Btk) within its PH domain cause X-linked agammaglobulinaemia (XLA) in patients.
  • FIG. 9 shows the comparison of the amino acid residues 953-1060 of MRCK to the consensus sequence of the PH domains of a family of proteins which include Rac1 and GTPase activating protein BEM2/IPL2. The consensus sequence has CD NO: smart00233.4, PH. The comparison indicates 87.5% sequence identities with a score of 47.8 bits and an E value of 2×10−6.
  • FIG. 10 depicts the comparison between the amino acid residues 954-1060 of MRCK1 and another consensus sequence of the PH domains. The consensus sequence has CD NO: pfam00169.4, PH, which is shared by proteins including Still life protein type 1 (SIF type 1) and C. elegans LET-502 protein. The comparison shows 88% sequence identities with a score of 42.6 bits and an E value of 9×10−5.
  • FIG. 11 shows the comparison between the amino acid residues 1102-1345 of MRCK1 and a consensus CNH domain sequence (CD NO: pfam00780.4, CNH). The alignment shows 85.7% sequence identities with a score of 60.0 bits and an E value of 6×10−10. The consensus CNH domain sequence is found in NIK1-like kinase, mouse citron (Rho-interacting, serine/threonine kinase 21), and yeast ROM1 and ROM2.
  • FIG. 12 shows the sequence alignment between the amino acid residues 1440-1475 of MRCK1 and the consensus sequence of other P21-Rho-binding domain (CD NO: smart00285, PBD). The two sequences have 86.1% sequence identities with a score of 38.3 bits and an E value of 0.002. P21-Rho-binding domain is a domain that binds Cdc42p- and/or Rho-like small GTPases. The domain also knows as the Cdc42/Rac interactive binding domain (CRIB domain).
  • The amino acid residues 648 to 786 in MRCK1 also weakly resemble a myosin tail domain (CD NO: pfam01576). The myosin molecule is a multi-subunit complex made up of two heavy chains and four light chains. It is a fundamental contractile protein found in all eukaryotic cell types. The myosin tail domain consists of the coiled-coil myosin heavy chain tail region. The coiled-coil is composed of the tail from two molecules of myosin. These can then assemble into the macromolecular thick filament. The coiled-coil region provides the structural backbone the thick filament. The alignment shows in FIG. 13. The two sequences being compared have 17.8% sequence identities with a score of 39.9 bits and an E value of 7×10−4.
  • The MRCK1 sequence also shows high homology to PKIN20, a human kinase disclosed in PCT patent application No. WO 02/08399. The two proteins share 90% sequence identities in amino acids and 95% identities at the cDNA level.
  • Hydrophobicity analysis indicates that MRCK1 kinase is not likely a membrane or transmembrane protein. The hydrophobicity profile of MRCK1 is illustrated in FIG. 14.
  • The existence and expression of the MRCK1 gene in humans are supported by various EST sequences. For instance, nucleotides 289-1205 of SEQ ID NO:1 are supported by the EST sequence disclosed under GenBank accession number BF994269; nucleotides 2449-2798 of SEQ ID NO:1 are supported by the EST sequences disclosed under GenBank accession numbers BF869661, BF357216, BF357213, BG952299, and BG014499; nucleotides 3042-3313 of SEQ ID NO:1 are supported by the EST sequence disclosed under GenBank accession number BF991223; nucleotides 4043-4403 of SEQ ID NO:1 are supported by the EST sequences disclosed under GenBank accession numbers BE793390, BG752641, and AW814108; nucleotides 4398-440 of SEQ ID NO:1 are supported by the EST sequences disclosed under GenBank accession numbers BG752641, AW516225, BE793390, BI792977, and BI793270; nucleotides 4556-4698 of SEQ ID NO:1 are supported by the EST sequences disclosed under GenBank accession numbers BG752641, BI793270, BE793390, AW516225, BI792977, and AA809737.
  • Two transcripts of MRCK1, a 4 kb and a 6 kb transcript, were detected in human brain, heart, skeletal muscle, colon, thymus, spleen, kidney, liver, small intestine, placenta, lung, and peripheral blood leukocyte by Multiple Tissue Northern analysis (MTN). The highest expression was in placenta while the lowest expression was in small intestine. The MRCK1 expression was confirmed by an multiple tissue expression array (MTA), in which MRCK1 expression was found in all 76 tissues contained in the array.
  • Utility of Protein Kinases
  • Protein kinases are involved in the regulation of many critical biological processes such as signal transduction pathways. Malfunctions of cellular signaling have been associated with many diseases. Regulation of signal transduction by cytokines and association of signal molecules with protooncogenes and tumor suppressor genes have been the subjects of intense research. Many therapeutic strategies can now be developed through the synthesis of compounds which activate or inactivate protein kinases.
  • The importance of kinases in the etiology of diseases has been well established. Kinase proteins are a major target for drug action and development. A January 2002 survey of ongoing clinical trials in the USA revealed more than 100 clinical trials involving the modulation of kinases. Trials are ongoing in a wide variety of therapeutic indications including asthma, Parkinson's, inflammation, psoriasis, rheumatoid arthritis, spinal cord injuries, muscle conditions, osteoporosis, graft versus host disease, cardiovascular disorders, autoimmune disorders, retinal detachment, stroke, epilepsy, ischemia/reperfusion, breast cancer, ovarian cancer, glioblastoma, non-Hodgkin's lymphoma, colorectal cancer, non-small cell lung cancer, brain cancer, Kaposi's sarcoma, pancreatic cancer, liver cancer, and other tumors. Numerous kinds of modulators of kinase activity are currently in clinical trials including antisense molecules, antibodies, small molecules, and even gene therapy. Accordingly, it is valuable to the field of pharmaceutical development to identify and characterize previously unknown members of the kinase family proteins. The present invention advances the state of the art by providing novel human kinase proteins which are structurally related to MRCKs.
  • Many therapeutic strategies are aimed at critical components in signal transduction pathways. Approaches for regulating kinase gene expression include specific antisense oligonucleotides for inhibiting post-transcriptional processing of the messenger RNA, naturally, occurring products and their chemical derivatives to inhibit kinase activity and monoclonal antibodies to inhibit receptor linked kinases. In some cases, kinase inhibitors also allow other therapeutic agents additional time to become effective and act synergistically with current treatments.
  • Among the areas of pharmaceutical research that are currently receiving a great deal of attention are the role of phosphorylation in transcriptional control, apoptosis, protein degradation, nuclear import and export, cytoskeletal regulation, and checkpoint signaling. The accumulating knowledge about signaling networks and the proteins involved will be put to practical use in the development of potent and specific pharmacological modulators of phosphorylation-dependent signaling. The rational structure-based design and development of highly specific kinase modulators is becoming routine and drugs that intercede in signaling pathways are becoming a major class of drug. The functions of some of the kinases are described below.
  • The second messenger dependent protein kinases primarily mediate the effects of second messengers such as cyclic AMP (cAMP), cyclic GMP, inositol triphosphate, phosphatidylinositol, 3,4,5-triphosphate, cyclic-ADPribose, arachidonic acid, diacylglycerol and calcium-calmodulin. The cyclic-AMP dependent protein kinases (PKA) are important members of the STK family. Cyclic-AMP is an intra-cellular mediator of hormone action in all prokaryotic and animal cells that have been studied. Such hormone-induced cellular responses include thyroid hormone secretion, cortisol secretion, progesterone secretion, glycogen breakdown, bone resorption, and regulation of heart rate and force of heart muscle contraction. PKA is found in all animal cells and is thought to account for the effects of cyclic-AMP in most of these cells. Altered PKA expression is implicated in a variety of disorders and diseases including cancer, thyroid disorders, diabetes, atherosclerosis, and cardiovascular disease.
  • Calcium-calmodulin (CaM) dependent protein kinases are also members of STK family. Calmodulin is a calcium receptor that mediates many calcium regulated processes by binding to target proteins in response to the binding of calcium. The principle target protein in these processes is CaM dependent protein kinases. CaM-kinases are involved in regulation of smooth muscle contraction (MLC kinase), glycogen breakdown (phosphorylase kinase), and neurotransmission (CaM kinase I and CaM kinase II). CaM kinase I phosphorylates a variety of substrates including the neurotransmitter related proteins synapsin I and II, the gene transcription regulator, CREB, and the cystic fibrosis conductance regulator protein, CFTR. CaM II kinase also phosphorylates synapsin at different sites, and controls the synthesis of catecholamines in the brain through phosphorylation and activation of tyrosine hydroxylase. Many of the CaM kinases are activated by phosphorylation in addition to binding to CaM. The kinase may autophosphorylate itself, or be phosphorylated by another kinase as part of a “kinase cascade”.
  • Another ligand-activated protein kinase is 5′-AMP-activated protein kinase (AMPK). Mammalian AMPK is a regulator of fatty acid and sterol synthesis through phosphorylation of the enzymes acetyl-CoA carboxylase and hydroxymethylglutaryl-CoA reductase and mediates responses of these pathways to cellular stresses such as heat shock and depletion of glucose and ATP. AMPK is a heterotrimeric complex comprised of a catalytic alpha subunit and two non-catalytic beta and gamma subunits that are believed to regulate the activity of the alpha subunit. Subunits of AMPK have a much wider distribution in non-lipogenic tissues, such as brain, heart, spleen, and lung, than expected. This distribution suggests that AMPK's functions may extend beyond regulation of lipid metabolism alone.
  • The mitogen-activated protein kinases (MAP) are also members of the STK family. MAP kinases also regulate intra-cellular signaling pathways. They mediate signal transduction from the cell surface to the nucleus via phosphorylation cascades. Several subgroups have been identified, and each manifests different substrate specificities and responds to distinct extracellular stimuli. MAP kinase signaling pathways are present in mammalian cells as well as in yeast. The extracellular stimuli that activate mammalian pathways include epidermal growth factor (EGF), ultraviolet light, hyperosmolar medium, heat shock, endotoxic lipopolysaccharide (LPS), and pro-inflammatory cytokines, such as tumor necrosis factor (TNF) and interleukin-1 (IL-1).
  • EGF receptor is found in over half of breast tumors unresponsive to hormone. EGF is found in many tumors, and EGF may be required for tumor cell growth. Antibody to EGF blocked the growth of tumor xenografts in mice. An antisense oligonucleotide for amphiregulin inhibited growth of a pancreatic cancer cell line.
  • Tamoxifen, a protein kinase C inhibitor with anti-estrogen activity, is currently a standard treatment for hormone-dependent breast cancer. The use of this compound may increase the risk of developing cancer in other tissues such as the endometrium. Raloxifene, a related compound, has been shown to protect against osteoporosis. The tissue specificity of inhibitors must be considered when identifying therapeutic targets.
  • Signal transduction to the nucleus in response to extracellular stimulus by a growth factor involves the mitogen activated protein (MAP) kinases. MAP kinases are a family of protein serine/threonine kinases which mediate signal transduction from extracellular receptors or heat shock, or UV radiation. Cell proliferation and differentiation in normal cells are under the regulation and control of multiple MAP kinase cascades. Aberrant and deregulated functioning of MAP kinases can initiate and support carcinogenesis. Insulin and IGF-1 also activate a mitogenic MAP kinase pathway that may be important in acquired insulin resistance occurring in type 2 diabetes.
  • Many cancers become refractory to chemotherapy by developing a survival strategy involving the constitutive activation of the phosphatidylinositol 3-kinase-protein kinase B/Akt signaling cascade. This survival signaling pathway thus becomes an important target for the development of specific inhibitors that would block its function. PI-3 kinase/Akt signaling is equally important in diabetes. The pathway activated by RTKs subsequently regulates glycogen synthase 3 (GSK3) and glucose uptake. Since AKT has decreased activity in type 2 diabetes, it provides a therapeutic target.
  • Protein kinase inhibitors provide much of our knowledge about in vivo regulation and coordination of physiological functions of endogenous peptide inhibitors. A pseudosubstrate sequence within PKC acts to inhibit the kinase in the absence of its lipid activator. A PKC inhibitor, such as chelerythrine, acts on the catalytic domain to block substrate interaction, while calphostin acts on the regulatory domain to mimic the pseudosubstrate sequence and block ATPase activity, or to inhibit cofactor binding.
  • Although some protein kinases have, to date, no known system of physiological regulation, many are activated or inactivated by autophosphorylation or phosphorylation by upstream protein kinases. The regulation of protein kinases also occurs during the transcription, post-transcription, and post-translation processes. The mechanism of post-transcriptional regulation is alternative splicing of precursor mRNA. For example, protein kinase C βI and βII are two isoforms of a single PKCβ gene derived from differences in the splicing of the exon encoding the C-terminal 50-52 amino acids. Splicing can be regulated by a kinase cascade in response to peptide hormones, such as insulin and IGF-1. PKC βI and βII have different specificities for phosphorylating members of the mitogen activated protein (MAP) kinase family, for glycogen synthase 3β, for nuclear transcription factors, such as TLS/Fus, and for other nuclear kinases. By inhibiting the post-transcriptional alternative splicing of PKC βII mRNA, PKC βII-dependent processes are inhibited.
  • The development of antisense oligonucleotides to inhibit the expression of various protein kinases has been successful. Antisense oligonucleotides are short lengths of synthetically manufactured, chemically modified DNA or RNA designed to specifically interact with mRNA transcripts encoding target proteins. The interaction of the antisense moiety with mRNA inhibits protein translation and, in some cases, post-transcriptional processing (e.g., alternative splicing and stability) of mRNA. Antisense oligonucleotides have been developed to alter alternative splicing of mRNA forms for inhibiting the translation of PKCα.
  • Protein kinase C isoforms have been implicated in cellular changes observed in the vascular complications of diabetes. Hyperglycemia is associated with increased levels of PKCα and β isoforms in renal glomeruli of diabetic rats. Oral administration of a PKCβ inhibitor prevented the increased mRNA expression of TGF-β1 and extracellular matrix component genes. Administration of the specific PKCβ inhibitor (LY333531) also normalized levels of cytokines, caldesmon, and hemodynamics of retinal and renal blood flow. Overexpression of the PKCβ isoform in the myocardium resulted in cardiac hypertrophy and failure. The use of LY333531 to prevent adverse effects of cardiac PKCβ overexpression in diabetic subjects is under investigation. The compound is also in Phase I/II clinical trials for diabetic retinopathy and diabetic macular edema indicating that it may be pharmacodynamically active.
  • PRK (proliferation-related kinase) is a serum/cytokine inducible STK that is involved in regulation of the cell cycle and cell proliferation in human megakaroytic cells. PRK is related to the polo (derived from humans polo gene) family of STKs implicated in cell division. PRK is down-regulated in lung tumor tissue and may be a proto-oncogene whose deregulated expression in normal tissue leads to oncogenic transformation. Altered MAP kinase expression is implicated in a variety of disease conditions including cancer, inflammation, immune disorders, and disorders affecting growth and development.
  • DNA-dependent protein kinase (DNA-PK) is involved in the repair of double-strand breaks in mammalian cells. This enzyme requires ends of double-stranded DNA or transitions from single-stranded to double-stranded DNA in order to act as a serine/threonine kinase. Cells with defective or deficient DNA-PK activity are unable to repair radiation induced DNA double-strand breaks and are consequently very sensitive to the lethal effects of ionizing radiation. Inhibition of DNA-PK has the potential to increase the efficacy of anti-tumor treatment with radiation or chemotherapeutic agents.
  • The cyclin-dependent protein kinases (CDKs) are another group of STKs that control the progression of cells through the cell cycle. Cyclins are small regulatory proteins that act by binding to and activating CDKs that then trigger various phases of the cell cycle by phosphorylating and activating selected proteins involved in the mitotic process. CDKs are unique in that they require multiple inputs to become activated. In addition to the binding of cyclin, CDK activation requires the phosphorylation of a specific threonine residue and the dephosphorylation of a specific tyrosine residue.
  • Cellular inhibitors of CDKs also play a major role in cell cycle progression. Alterations in the expression, function, and structure of cyclin and CDK are encountered in the cancer phenotype. Therefore CDKs may be important targets for new cancer therapeutic agents.
  • Chemotherapy resistant cells tend to escape apoptosis. Under certain circumstances, inappropriate CDK activation may even promote apoptosis by encouraging the progression of the cell cycle under unfavorable conditions, i.e., attempting mitosis while DNA damage is largely unrepaired.
  • Purines and purine analogs act as CDK inhibitors. Flavopiridol is a flavonoid that causes 50% growth inhibition of tumor cells at 60 nM. It also inhibits EGFR and protein kinase A. Flavopiridel induces apoptosis and inhibits lymphoid, myeloid, colon, and prostate cancer cells grown in vivo as tumor xenografts in nude mice.
  • Staurosporine and its derivative, UCN-01, in addition to inhibiting protein kinase C, inhibit cyclin B/CDK (IC50=3 to 6 nM). Staurosporine is toxic, but its derivative 7-hydroxystaurosporine (UCN-01) has anti-tumor properties and is in clinical trials. UCN-01 affects the phosphorylation of CDKs and alters the cell cycle checkpoint functioning. These compounds illustrate that multiple intra-cellular targets may be affected as the concentration of an inhibitor is increased within cells.
  • Protein tyrosine kinases, PTKs, specifically phosphorylate tyrosine residues on their target proteins and may be divided into transmembrane, receptor PTKs and non-transmembrane, non-receptor PTKs. Transmembrane protein tyrosine kinases are receptors for most growth factors. Binding of a growth factor to the receptor activates the transfer of a phosphate group from ATP to selected tyrosine side chains of the receptor and other specific proteins. Growth factors (GF) associated with receptor PTKs include; epidermal GF, platelet-derived GF, fibroblast GF, hepatocyte GF, insulin and insulin-like GFs, nerve GF, vascular endothelial GF, and macrophage colony stimulating factor.
  • Since RTKs stimulate tumor cell proliferation, inhibitors of RTKs may inhibit the growth and proliferation of such cancers. Inhibitors of RTKs are also useful in preventing tumor angiogenesis and can eliminate support from the host tissue by targeting RTKs located on vascular cells, such as blood vessel endothelial cells and stromal fibroblasts. For example, VEGF stimulates endothelial cell growth during angiogenesis, and increases the permeability of tumor vasculature so that proteins and other growth factors become accessible to the tumor. Broad-spectrum antitumor efficacy of an oral dosage form of an inhibitor of VEGF signaling has been reported. Thus, inhibition of VEGF receptor signaling presents an important therapeutic target. An extracellular receptor can also be a target for inhibition. For example, the EGF receptor family and its ligands are overexpressed and exist as an autocrine loop in many tumor types.
  • Increasing knowledge of the structure and activation mechanism of RTKs and the signaling pathways controlled by tyrosine kinases provided the possibility for the development of target specific drugs and new anti-cancer therapies. Approaches towards the prevention or interception of deregulated RTK signaling include the development of selective components that target either the extracellular ligand-binding domain or the intra-cellular substrate binding region.
  • The most successful strategy to selectively kill tumor cells is the use of monoclonal antibodies (mAbs) that are directed against the extracellular domain of RTKs which are critically involved in cancer and are expressed at the surface of tumor cells. In the past years, recombinant antibody technology has made an enormous progress in the design, selection and production of newly engineered antibodies. It is also possible to generate humanized antibodies, human-mouse chimeric or bispecific antibodies for targeted cancer therapy. Mechanistically, anti-RTK mAbs might work by blocking the ligand-receptor interaction and therefore inhibiting ligand-induced RTK signaling and increasing RTK down-regulation and internalization. In addition, binding of mAbs to certain epitopes on the cancer cells may induce immune-mediated responses, such as opsonization and complement-mediated lysis, and trigger antibody-dependent cellular cytotoxicity by macrophages or natural killer cells. In recent years, it became evident that mAbs control tumor growth by altering the intra-cellular signaling pattern inside the targeted tumor cell, leading to growth inhibition and/or apoptosis. In addition, bispecific antibodies can bridge selected surface molecules on a target cell with receptors on an effector cell, thus triggering cytotoxic responses against the target cell. Despite the toxicity that has been seen in clinical trials of bispecific antibodies, advances in antibody engineering, characterization of tumor antigens and immunology might help to produce rationally designed bispecific antibodies for anti-cancer therapy.
  • Another promising approach to inhibiting aberrant RTK signaling is to develop small molecule drugs that selectively interfere with the intrinsic tyrosine kinase activity and thereby block receptor autophosphorylation and activation of downstream signal transducers. The tyrphostins, which belong to the quinazolines, are one important group of such inhibitors that compete with ATP for the ATP binding site at the receptor's tyrosine kinase domain and some members of the group have been shown to specifically inhibit the EGFR. Potent and selective inhibitors of receptors involved in neovascularization have been developed and are now undergoing clinical evaluation. New classes of tyrosine kinase inhibitors (TKIs) with increased potency and selectivity, higher in vitro and in vivo efficacy and decreased toxicity have been developed using the advantages of structure-based drug design, crystallographic structure information, combinatorial chemistry and high-throughput screening.
  • Recombinant immunotoxins provide another possibility of target-selective drug design. Recombinant immunotoxins are composed of a bacterial or plant toxin either fused or chemically conjugated to a specific ligand, such as the variable domains of the heavy and light chains of mAbs or to a growth factor. Immunotoxins may contain bacterial toxins, such as Pseudomouas exotoxin A or diphtheria toxin, or plant toxins, such as ricin A or clavin. These recombinant molecules can selectively kill their target cells when internalized after binding to cell surface receptors of the target cells.
  • The use of antisense oligonucleotides represents another strategy to inhibit the activation of RTKs. Antisense oligonucleotides are short pieces of synthetic DNA or RNA that are designed to interact with the mRNA to block the transcription and thus the expression of the target proteins. Antisense oligonucleotides interact with the mRNA by Watson-Crick base-pairing and are therefore highly specific to the target protein. Several preclinical and clinical studies suggest that antisense therapy might be therapeutically useful for the treatment of solid tumors.
  • The potential of RTKs and their relevant signaling as selective anti-cancer targets for therapeutic intervention has been recognized. As a consequence, a variety of successful target specific drugs such as mAbs and RTK inhibitors have been developed and are currently being evaluated in clinical trials. Table 3 summarizes the most successful drugs against receptor tyrosine kinase signaling which are currently evaluated in clinical phases or have already been approved by the FDA.
  • TABLE 3
    RTK Drugs Currently Under Clinical Evaluation
    RTK Drug Company Description Status
    EGFR ZA18539 Iressa AstraZeneca TKI that inhibits EGFR Phase III
    signaling
    EGFR Cetuximab C225 ImClone Mab directed against EGFR Phase III
    Systems
    EGFR EGF fusion protein Seragen Recombinant diphtheria Phase II
    toxin-hEGF fusion protein
    HER2 Trastuzumab Genetech Mab directed against HER2 Approved
    Herceptin by the FDA
    in 1998
    IGF-IR INX-4437 INEX USA Antisense oligonucleotides Phase I
    targeting IGR-IR
    VEGFR SU5416 SUGEN TKI that inhibits VEGFR2 Phase II
    VEGFR/ SU6668 SUGEN RTK inhibition of Phase I
    FGFR/ VEGFR, FGFR, and PDGFR
    PDGFR
  • Non-receptor PTKs lack transmembrane regions and, instead, form complexes with the intra-cellular regions of cell surface receptors. Receptors that function through non-receptor PTKs include those for cytokines, hormones (growth hormone and prolactin) and antigen-specific receptors on T and B lymphocytes.
  • Many of the PTKs were first identified as the products of mutant oncogenes in cancer cells where their activation was no longer subject to normal cellular controls. In fact, about one third of the known oncogenes encode PTKs, and it is well-known that cellular transformation (oncogenesis) is often accompanied by increased tyrosine phosphorylation activity.
  • Many tyrosine kinase inhibitors, such as flavopiridol, genistem, erbstatin, lavendustin A, staurosporine, and UCN-01, are derived from natural products. Inhibitors directed to the ATP binding site are also available. Signals from RTKs can also be inhibited at other target sites such as nuclear tyrosine kinases, membrane anchors (inhibition of farnesylation) and transcription factors.
  • Targeting the signaling potential of growth promoting tyrosine kinases such as EGFR, HER2, PDGFR, src, and abl, will block tumor growth while blocking IGF-1 and TRK will interfere with tumor cell survival. Inhibition of these kinases will lead to tumor shrinkage and apoptosis. FklI/KDR and src are kinases necessary for neovascularization (angiogenesis) of tumors. Inhibition of these kinases will slow tumor growth and decrease metastases.
  • Inhibitors of RTKs suppress tumor development by preventing cell migration, invasion and metastases. These drugs are likely to increase the time required for tumor progression, and may inhibit or attenuate the aggressiveness of the disease but may not initially result in measurable tumor regression.
  • An example of cancer arising from a defective tyrosine kinase is a class of ALK positive lymphomas referred to as “ALKomas” which display inappropriate expression of a neural-specific tyrosine kinase, anaplastic lymphoma kinase (ALK).
  • Iressa (ZD1839) is an orally active selective EGF-R inhibitor. This compound disrupts signaling involved in cancer cell proliferation. The clinical efficacy of this agent shows that it is well tolerated by patients undergoing Phase I/II clinical trials. The compound has shown promising cytotoxicity towards several cancer cell lines.
  • Since the majority of protein kinases are expressed in the brain, often in a neuron-specific fashion, protein phosphorylation must play a key role in the development and function of the vertebrate central nervous system. Thus neuron-specific kinases are well established as targets for the development of pharmacologically active modulators.
  • In summary, kinase proteins are a major target for drug action and development. Accordingly, it is valuable to the field of pharmaceutical development to identify and characterize previously unknown members of kinase proteins. The present invention advances the state of the art by providing a previously unidentified human kinase protein that has homology to rat MRCK.
  • Utility of the MRCK1 Gene and MRCK1 Kinase
  • MRCKs are serine/threonine kinases with multiple functional domains. MRCKs are thought to act as a downstream effector of Cdc42 in cytoskeletal reorganization. Cdc42 is an essential GTPase that belongs to the Rho/Rac subfamily of Ras-like GTPases. These proteins act as molecular switches by responding to exogenous and/or endogenous signals and relaying those signals to activate downstream components of a biological pathway. The 11 current members of the Cdc42 family display between 75 and 100% amino acid identity and are functional, as well as structural, homologs. Cdc42 transduces signals to the actin cytoskeleton to initiate and maintain polarized growth and to mitogen-activated protein morphogenesis. In the budding yeast Saccharomyces cerevisiae, Cdc42 plays an important role in multiple actin-dependent morphogenetic events such as bud emergence, mating-projection formation, and pseudohyphal growth. In mammalian cells, Cdc42 regulates a variety of actin-dependent events and induces the JNK/SAPK protein kinase cascade, which leads to the activation of transcription factors within the nucleus. Cdc42 mediates these processes through interactions with a myriad of downstream effectors. In addition, Cdc42 has been implicated in a number of human diseases through interactions with its regulators and downstream effectors.
  • The MRCK family includes at least two related protein kinases: MRCK alpha and MRCK beta, which were isolated from a human brain cDNA library using a monoclonal antibody directed against myotonic dystrophy protein kinase (DMPK). The epitope shared by DMPK and MRCKs was located at the catalytic site of DMPK using a phage-displayed random peptide library (Lam L T, Hum Mol Genet. 9:2167-2173, 2000).
  • MRCKs are involved in Cdc42-mediated myosin light chain phosphorylation (Dong et al., Eur J Cell Biol 81:231-42, 2002). Specifically, MRCK alpha was implicated in Cdc42-mediated peripheral actin formation and neurite outgrowth in HeLa and PC12 cells, respectively. It was suggest that MRCK alpha may act as a downstream effector of Cdc42 in cytoskeletal reorganization (Leung et al., Mol Cell Biol 18:130-40, 1998).
  • The native MRCK exists in high-molecular-weight complexes. The three independent coiled-coil (CC) domains and the N-terminal region preceding the kinase domain are responsible for intermolecular interactions leading to MRCK alpha multimerization. N-terminus-mediated dimerization and consequent trans-autophosphorylation are critical processes regulating MRCK alpha catalytic activities. A region containing the two distal CC domains (CC2 and CC3; residues 658 to 930) was found to interact intramolecularly with the kinase domain and negatively regulates its activity. Its deletion also resulted in an active kinase, confirming a negative auto-regulatory role. The N-terminus-mediated dimerization and activation of MRCK and the negative kinase-distal CC domain interaction are two mutually exclusive events that tightly regulate the catalytic state of the kinase. Disruption of this interaction by a mutant kinase domain resulted in increased kinase activity. MRCK kinase activity was also elevated when cells were treated with phorbol ester, which can interact directly with a cysteine-rich domain next to the distal CC domain. It therefore appears that binding of phorbol ester to MRCK releases its auto-inhibition, allowing N-terminal dimerization and subsequent kinase activation. (Tan et al., Mol. Cell. Biol., 21:2767-78, 2001)
  • The approximately 190-kD MRCK kinases preferentially phosphorylate non-muscle myosin light chain at serine 19, which is known to be crucial for activating actin-myosin contractility. The p21-binding domain binds GTP-Cdc42 but not GDP-Cdc42. The multidomain structure includes a cysteine-rich motif resembling those of protein kinase C and n-chimaerin and a putative Pleckstrin homology domain. MRCK alpha and Cdc42V12 co-localize, particularly at the cell periphery in transfected HeLa cells. Microinjection of a plasmid encoding MRCK alpha resulted in actin and myosin reorganization. Expression of kinase-dead MRCK alpha blocked Cdc42V12-dependent formation of focal complexes and peripheral microspikes. This was not due to possible sequestration of the p21, as a kinase-dead MRCK alpha mutant defective in Cdc42 binding was an equally effective blocker. Coinjection of an MRCK alpha plasmid with a Cdc42 plasmid, at concentrations where Cdc42 plasmid by itself elicited no effect, led to the formation of the peripheral structures associated with a Cdc42-induced morphological phenotype. These Cdc42-type effects were not promoted upon coinjection with plasmids of kinase-dead or Cdc42-binding-deficient MRCK alpha mutants. These results suggest that MRCK alpha may act as a downstream effector of Cdc42 in cytoskeletal reorganization (Leung et al., Mol. Cell. Biol., 18:130-40, 1998).
  • Two major substrates, p130 and p85, for MRCK alpha-kinase have been identified. P130 is identified as the myosin binding subunit p130, whereas p85 is a novel related protein. P85 contains N-terminal ankyrin repeats, an alpha-helical C terminus with leucine repeats, and a centrally located conserved motif with the MRCK alpha kinase phosphorylation site. Like MBS130, p85 is specifically associated with protein phosphatase 1delta (PP1delta), and this requires the N-terminus, including the ankyrin repeats. This association is required for the regulation of both the catalytic activities and the assembly of actin cytoskeleton. The N-terminus, in association with PP1delta, is essential for actin depolymerization, whereas the C-terminus antagonizes this action. The C-terminal effects consist of two independent events that involved both a conserved phosphorylation inhibitory motif and an alpha-helical leucine repeats. The former was able to interact with PP1delta only in the phosphorylated state and result in inactivation of PP1delta activity. This provides further evidence that phosphorylation of a myosin binding subunit protein by specific kinases confers conformational changes in a highly conserved region that plays an essential role in the regulation of its catalytic subunit activities (Tan et al., J. Biol. Chem., 276:21209-16, 2001).
  • Taken together, MRCKs appear to be an important player in cytoskeletal reorganization, neuronal differentiation, and myotonic dystrophy. MRCKs may serve as a major target for drug action and development.
  • The present invention provides a new human kinase (MRCK1) which has sequence and structure similarities to rat MRCK alpha and other kinases. The multiple domains in MRCK1 share high sequence identities with the corresponding domains in other kinases. Each of these domains, either in its native form or in a mutant form, can be used to affect the function of the corresponding domain in other kinases. The kinase domain in MRCK1 can be used to phosphorylate suitable substrates, including p130 and p85 or substrate peptides containing MRCK alpha phosphorylation sites. The substrate peptides can be conjugated to antibodies, and the phosphate groups added to the substrate peptides can be radioactively or fluorescently labeled. Antibodies thus labeled can be used in various detection assays, as appreciated by one of skilled in the art.
  • The MRCK1 gene and gene products can be used as molecular markers for diagnosing, prognosing, and monitoring the treatment of disorders related to the aberrant expression of MRCK1. In addition, the MRCK1 gene can be used to screen for potential agents or drugs capable of enhancing or inhibiting the MRCK1 gene expression in human cells. The MRCK1 gene products (polynucleotide and polypeptide) can be used to screen for potential agents or drugs capable of enhancing or inhibiting MRCK1 activity. Furthermore, various therapeutic methods for treating disorders related to the aberrant expression of MRCK1 can be designed based on the MRCK1 gene, its variants, or the agents/drugs that affect the expression of the MRCK1 gene or the activity of the MRCK1 gene products.
  • The following subsections illustrate examples of the utilities of human MRCK1 gene and MRCK1 kinase. Various changes and modifications within the spirit and scope of the present invention will become apparent to those skilled in the art from the present description.
  • Polynucleotides and Variants Thereof
  • One aspect of the invention pertains to isolated polynucleotide probes capable of hybridizing to the MRCK1 gene or its transcripts, such as MRCK1 mRNAs. These probes can be used to detect the expression level of the MRCK1 gene in human tissue or cells. The present invention also contemplates polynucleotide fragments for use as PCR primers for the amplification or mutation of the MRCK1 gene or the MRCK1 kinase-coding sequences. Another aspect of the invention pertains to isolated polynucleotides that encode MRCK1, or a fragment or mutant thereof. These polynucleotides can be used for expressing MRCK1, or a fragment or mutant thereof. The protein products thus expressed can be used to screen for agents/drugs that modulate an activity of MRCK1. In addition, these polynucleotides can be used to designing gene therapy vectors which target the expression of the MRCK1 gene or an activity of MRCK1 in humans.
  • A polynucleotide comprising SEQ ID NO:1 or SEQ ID NO:3 can be prepared using standard molecular biology techniques as appreciated by one of ordinary skill in the art. For instance, primers derived from the 5′ and 3′ ends of SEQ ID NO:1 can be used to amplify mRNAs isolated from human tissues. The cDNA thus produced contains SEQ ID NO:1. Likewise, primers for amplifying the human genomic sequence containing SEQ ID NO:3 can be designed and used to prepare the genomic sequence of the MRCK1 gene. A variant (such as a homolog) or a fragment of SEQ ID NO:1 or SEQ ID NO:3 can be similarly prepared. Alternatively, probes can be designed to screen for cDNA or genomic sequence libraries in order to identify polynucleotide molecules comprising the full-length or fragments of SEQ ID NO:1 or SEQ ID NO:3. The molecules thus identified can be used to create suitable vectors comprising the full-length SEQ ID NO:1 or SEQ ID NO:3.
  • Polynucleotides capable of hybridizing to the MRCK1 gene can be prepared by standard synthetic techniques, e.g., using an automated DNA synthesizer. Preferably, the polynucleotide probes can hybridize to the MRCK1 gene under reduced stringent conditions, stringent conditions, or highly stringent conditions. In one embodiment, the polynucleotides comprise at least 15, 20, 25, 30, 50, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, 900, 950, 1000 or more consecutive nucleotides of SEQ ID NO:1. Any fragments of SEQ ID NO:1 and SEQ ID NO:3 may be used as hybridization probes or PCR primers for the MRCK1 gene or its transcripts. The probes/primers can be substantially purified.
  • In a preferred embodiment, the hybridization probes for the MRCK1 gene comprise a label group. The label group can be a radioisotope, a fluorescent compound, an enzyme, or an enzyme co-factor. Probes thus labeled can be used as part of a diagnostic kit for determining the expression level of the MRCK1 gene in human tissues.
  • This invention encompasses human MRCK1 gene homologs in other species. These homologs can be determined by search different sequence databases, such as the Entrez/GenBank sequence databases maintained by the NCBI. The invention also encompasses polynucleotide molecules which are structurally different from the molecules described above, but have the substantially same properties as the molecules described above. Such molecules include allelic variants, which will be described below in greater detail.
  • DNA sequence polymorphism in human MRCK1 gene exists among different individuals due to natural allelic variations. An allele is one of a group of genes which occur alternatively at a given genetic locus. DNA polymorphisms that affect the RNA expression level of the MRCK1 gene can also exist, e.g. through affecting the regulation or degradation of expression of the gene. The present invention contemplates all allelic variants of human MRCK1 gene. Allelic variants and other homologs of the MRCK1 gene can be isolated using probes/primers derived from SEQ ID NO:1 or SEQ ID NO:3.
  • It should, of course, be understood that SEQ ID NO:1 and SEQ ID NO:3 can be modified. The modified polynucleotides can comprise one or more mutations. These mutations can be substitutions, additions or deletions of 1, 2, 3, 5, 10, 15, 20 or more nucleotide residues in SEQ ID NO:1 or SEQ ID NO:3. Standard techniques can be used, such as site-directed mutagenesis or PCR-mediated mutagenesis. Preferably, these mutations create conservative amino acid substitutions. Alternatively, mutations can be introduced randomly along all or part of the MRCK1 gene or its cDNA, such as by saturation mutagenesis. Following mutagenesis, the encoded proteins can be expressed recombinantly and their activities can be determined.
  • In one embodiment, nucleotide substitutions leading to amino acid substitutions at “non-essential” amino acid residues can be introduced. A “non-essential” amino acid residue is a residue that can be altered without changing the biological activity of the protein. In contrast, an “essential” amino acid residue is required for the biological activity of the protein. Amino acid residues that are conserved among allelic variants or homologs of the MRCK1 gene from different species preferably are not changed in the present invention.
  • Accordingly, another aspect of the invention pertains to MRCK1 proteins that contain changes in amino acid residues that are not essential for the biological activity of MRCK1. These proteins differ in amino acid sequence from the original human MRCK1 kinase, but retain its biological activity. In one embodiment, the modified protein comprises an amino acid sequence at least about 91%, 95%, 98%, 99% or more homologous to SEQ ID NO:2.
  • In another embodiment, MRCK1 proteins contain mutations in amino acid residues which result in inhibition of MRCK1 activity. These mutated MRCK1 proteins can be used to inhibit MRCK1 activity in patients with disorders related to the aberrant expression of MRCK1.
  • A polynucleotide of this invention can be further modified to increase its stability in vivo. Possible modifications include, but are not limited to, the addition of flanking sequences at the 5′ and/or 3′ ends; the use of phosphorothioate or 2-o-methyl rather than phosphodiester linkages in the backbone; and/or the inclusion of nontraditional bases such as inosine, queosine and wybutosine, as well as acetyl- methyl-, thio- and other modified forms of adenine, cytidine, guanine, thymine and uridine.
  • Polynucleotide molecules which are antisense to the MRCK1 gene can be prepared. An “antisense” polynucleotide comprises a nucleotide sequence which is complementary to a “sense” polynucleotide which encodes a protein. An antisense polynucleotide can bind via hydrogen bonds to the sense polynucleotide.
  • Antisense polynucleotides of the invention can be designed according to the rules of Watson and Crick base pairing. The antisense polynucleotide molecule can be complementary to the entire coding region or part of the coding region of the MRCK1 gene. The antisense polynucleotide molecule can also be complementary to a “noncoding region” in the coding strand of the MRCK1 gene. Preferably, the antisense polynucleotide is an oligonucleotide which is antisense to only a portion of the MRCK1 gene. An antisense polynucleotide can be, for example, about 5, 10, 15, 20, 25, 30, 35, 40, 45 or 50 nucleotides in length. An antisense polynucleotide of the invention can be constructed using chemical synthesis and enzymatic ligation reactions as appreciated by one of ordinary skill in the art. For example, an antisense polynucleotide 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 polynucleotides. Examples of modified nucleotides which can be used to generate the antisense polynucleotide include 5-fluorouracil, 5-bromouracil, 5-chlorouracil, 5-iodouracil, hypoxanthine, xanthine, 4-acetylcytosine, 5-(carboxyhydroxymethyl)uracil, 5-carboxymethylaminomethyl-2-thiouridine, 5-carboxymethylaminomethyluracil, dihydrouracil, beta-D-galactosylqueosine, inosine, N6-isopentenyladenine, 1-methylguanine, 1-methylinosine, 2,2-dimethylguanine, 2-methyladenine, 2-methylguanine, 3-methylcytosine, 5-methylcytosine, N6-adenine, 7-methylguanine, 5-methylaminomethyluracil, 5-methoxyaminomethyl-2-thiouracil, beta-D-mannosylqueosine, 5′-methoxycarboxymethyluracil, 5-methoxyuracil, 2-methylthio-N6-isopentenyl adenosine, uracil-5-oxyacetic acid, wybutoxosine, pseudouracil, queosine, 2-thiocytosine, 5-methyl-2-thiouracil, 2-thiouracil, 4-thiouracil, 5-methyluracil, uracil-5-oxyacetic acid methylester, 3-(3-amino-3-N2-carboxypropyl)uracil, (acp3)w, and 2,6-diaminopurine. Phosphorothioate derivatives and acridine substituted nucleotides can also be used. Alternatively, the antisense polynucleotide can be produced biologically using an expression vector into which a polynucleotide has been subcloned in an antisense orientation (i.e., RNA transcribed from the inserted polynucleotide will be of an antisense orientation to the target polynucleotide of interest).
  • The antisense polynucleotides of the invention can be administered to a subject or applied in situ such that they hybridize or bind to cellular mRNAs and/or genomic DNA's that encode MRCK1 kinase, thereby inhibiting the expression of MRCK1 kinase. The hybridization can result in a stable duplex via conventional nucleotide complementarity. An example route for administering antisense polynucleotides includes direct injection at a tissue site. Antisense polynucleotides can also be modified first, 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. Suitable modifications include linking the antisense polynucleotides to peptides or antibodies which bind to the cell surface receptors or antigens. In addition, the antisense polynucleotides can be delivered to cells using vectors. To achieve sufficient intra-cellular concentrations of the antisense molecules, strong pol II or pol III promoters may be used in the vectors.
  • In one embodiment, the antisense polynucleotides are α-anomeric polynucleotides. An α-anomeric polynucleotide molecule forms specific double-stranded hybrid with a complementary RNA in which, contrary to the usual β-units, the strands run parallel to each other. The antisense polynucleotide molecule can also comprise a 2-o-methylribonucleotide or a chimeric RNA-DNA analog.
  • In another embodiment, the antisense polynucleotide is a ribozyme. Ribozymes are catalytic RNA molecules with ribonuclease activity which are capable of cleaving a single-stranded polynucleotide, such as an mRNA, to which they have a complementary region. Thus, ribozymes (e.g., hammerhead ribozymes described in Haselhoif and Gerlach Nature 334:585-591, 1988) can be used to catalytically cleave mRNA transcripts of MRCK1 in order to inhibit its expression. A ribozyme having specificity for the MRCK1 gene or its transcripts can be designed based upon SEQ ID NO:1 or 3. mRNAs transcribed from the MRCK1 gene can be used to select from a pool of RNA molecules a catalytic RNA having a specific ribonuclease activity.
  • Alternatively, the expression of the MRCK1 gene can be inhibited by using nucleotide sequences complementary to the regulatory region (e.g., the promoter and/or enhancers). These nucleotide sequences can form triple helical structures that prevent transcription of the gene in the target cells.
  • Expression of the MRCK1 gene can also be inhibited using RNA interference (“RNAi”). RNAi is a phenomenon in which the introduction of double-stranded RNA (dsRNA) into certain organisms or cell types causes degradation of the homologous mRNA. First discovered in the nematode Caenorhabditis elegans, RNAi has since been found to operate in a wide range of organisms. For example, in mammalian cells, introduction of long dsRNA (>30 nucleotides) can initiate a potent antiviral response, exemplified by nonspecific inhibition of protein synthesis and RNA degradation. RNA interference provides a mechanism of gene silencing at the mRNA level. In recent years, RNAi has become an endogenous and potent gene-specific silencing technique that uses double-stranded RNAs (dsRNA) to mark a particular transcript for degradation in vivo. It also offers an efficient and broadly applicable approach for gene knock-out. In addition, RNAi technology can be used for therapeutic purposes. For example, RNAi targeting Fas-mediated apoptosis has been shown to protect mice from fulminant hepatitis. RNAi technology has been disclosed in numerous publications, such as U.S. Pat. Nos. 5,919,619, 6,506,559 and PCT Publication Nos. WO99/14346, WO01/70949, WO01/36646, WO00/63364, WO00/44895, WO01/75164, WO01/92513, WO01/68836 and WO01/29058.
  • A sequence capable of inhibiting gene expression by RNA interference can have any length. For instance, the sequence can have at least 10, 15, 20, 25, 30, 35, 40, 45, 50, 100 or more consecutive nucleotides. The sequence can be dsRNA or any other type of polynucleotide, provided that the sequence can form a functional silencing complex to degrade the target mRNA transcript.
  • In one embodiment, the sequence comprises or consists of a short interfering RNAs (siRNA). The siRNA can be dsRNA having 19-25 nucleotides. siRNAs can be produced endogenously by degradation of longer dsRNA molecules by an RNase III-related nuclease called Dicer. siRNAs can also be introduced into a cell exogenously, or by transcription of an expression construct. Once formed, the siRNAs assemble with protein components into endoribonuclease-containing complexes known as RNA-induced silencing complexes (RISCs). An ATP-generated unwinding of the siRNA activates the RISCs, which in turn target the complementary mRNA transcript by Watson-Crick base-pairing, thereby cleaving and destroying the mRNA. Cleavage of the mRNA takes place near the middle of the region bound by the siRNA strand. This sequence specific mRNA degradation results in gene silencing.
  • At least two ways can be employed to achieve siRNA-mediated gene silencing. First, siRNAs can be synthesized in vitro and introduced into cells to transiently suppress gene expression. Synthetic siRNA provides an easy and efficient way to achieve RNAi. siRNA are duplexes of short mixed oligonucleotides which can include, for example, 19 RNAs nucleotides with symmetric dinucleotide 3′ overhangs. Using synthetic 21 bp siRNA duplexes (e.g., 19 RNA bases followed by a UU or dTdT 3′ overhang), sequence specific gene silencing can be achieved in mammalian cells. These siRNAs can specifically suppress targeted gene translation in mammalian cells without activation of DNA-dependent protein kinase (PKR) by longer dsRNA, which may result in non-specific repression of translation of many proteins.
  • Second, siRNAs can be expressed in vivo from vectors. This approach can be used to stably express siRNAs in cells or transgenic animals. In one embodiment, siRNA expression vectors are engineered to drive siRNA transcription from polymerase III (pol III) transcription units. Pol III transcription units are suitable for hairpin siRNA expression, since they deploy a short AT rich transcription termination site that leads to the addition of 2 bp overhangs (e.g., UU) to hairpin siRNAs—a feature that is helpful for siRNA function. The Pol III expression vectors can also be used to create transgenic mice that express siRNA.
  • In another embodiment, siRNAs can be expressed in a tissue-specific manner. Under this approach, long double-stranded RNAs (dsRNAs) are first expressed from a promoter (such as CMV (pol II)) in the nuclei of selected cell lines or transgenic mice. The long dsRNAs are processed into siRNAs in the nuclei (e.g., by Dicer). The siRNAs exit from the nuclei and mediate gene-specific silencing. A similar approach can be used in conjunction with tissue-specific (pol II) promoters to create tissue-specific knockdown mice.
  • Any 3′ dinucleotide overhang, such as UU, can be used for siRNA design. In some cases, G residues in the overhang are avoided because of the potential for the siRNA to be cleaved by RNase at single-stranded G residues.
  • With regard to the siRNA sequence itself, it has been found that siRNAs with 30-50% GC content can be more active than those with a higher G/C content in certain cases. Moreover, since a 4-6 nucleotide poly(T) tract may act as a termination signal for RNA pol III, stretches of ≧4 Ts or As in the target sequence may be avoided in certain cases when designing sequences to be expressed from an RNA pol III promoter. In addition, some regions of mRNA may be either highly structured or bound by regulatory proteins. Thus, it may be helpful to select siRNA target sites at different positions along the length of the gene sequence. Finally, the potential target sites can be compared to the appropriate genome database (human, mouse, rat, etc.). Any target sequences with more than 16-17 contiguous base pairs of homology to other coding sequences may be eliminated from consideration in certain cases.
  • In one embodiment, siRNA can be designed to have two inverted repeats separated by a short spacer sequence and end with a string of Ts that serve as a transcription termination site. This design produces an RNA transcript that is predicted to fold into a short hairpin siRNA. The selection of siRNA target sequence, the length of the inverted repeats that encode the stem of a putative hairpin, the order of the inverted repeats, the length and composition of the spacer sequence that encodes the loop of the hairpin, and the presence or absence of 5′-overhangs, can vary to achieve desirable results.
  • The siRNA targets can be selected by scanning an mRNA sequence for AA dinucleotides and recording the 19 nucleotides immediately downstream of the AA. Other methods can also been used to select the siRNA targets. In one example, the selection of the siRNA target sequence is purely empirically determined (see e.g., Sui et al., Proc. Natl. Acad. Sci. USA 99: 5515-5520, 2002), as long as the target sequence starts with GG and does not share significant sequence homology with other genes as analyzed by BLAST search. In another example, a more elaborate method is employed to select the siRNA target sequences. This procedure exploits an observation that any accessible site in endogenous mRNA can be targeted for degradation by synthetic oligodeoxyribonucleotide/RNase H method (Lee et al., Nature Biotechnology 20:500-505, 2002).
  • In another embodiment, the hairpin siRNA expression cassette is constructed to contain the sense strand of the target, followed by a short spacer, the antisense strand of the target, and 5-6 Ts as transcription terminator. The order of the sense and antisense strands within the siRNA expression constructs can be altered without affecting the gene silencing activities of the hairpin siRNA. In certain instances, the reversal of the order may cause partial reduction in gene silencing activities.
  • The length of nucleotide sequence being used as the stem of siRNA expression cassette can range, for instance, from 19 to 29. The loop size can range from 3 to 23 nucleotides. Other lengths and/or loop sizes can also be used.
  • In yet another embodiment, a 5′ overhang in the hairpin siRNA construct can be used, provided that the hairpin siRNA is functional in gene silencing. In one specific example, the 5′ overhang includes about 6 nucleotide residues.
  • In still yet another embodiment, the target sequence for RNAi is a 21-mer sequence fragment selected from SEQ ID NO:1. The 5′ end of the target sequence has dinucleotide “NA,” where “N” can be any base and “A” represents adenine. The remaining 19-mer sequence has a GC content of between 35% and 55%. In addition, the remaining 19-mer sequence does not include any four consecutive A or T (i.e., AAAA or TTTT), three consecutive G or C (i.e., GGG or CCC), or seven “GC” in a role. Exemplary RNAi target sequences identified according to the above-described criteria (“relaxed” criteria) are illustrated in Table 4. The siRNA sequences for each target sequence (listed in the same row as the target sequence and including the sense strand and the antisense strand) are also indicated in Table 4.
  • Additional criteria can also be used for selecting RNAi target sequences. For instance, the GC content of the remaining 19-mer sequence can be limited to between 45% and 55%. Moreover, any 19-mer sequence having three consecutive identical bases (i.e., GGG, CCC, TTT, or AAA) or a palindrome sequence with 5 or more bases is excluded. Furthermore, the remaining 19-mer sequence can be selected to have low sequence homology to other human genes. In one specific example, potential target sequences are searched by BLASTN against NCBI's human UniGene cluster sequence database. The human UniGene database contains non-redundant sets of gene-oriented clusters. Each UniGene cluster includes sequences that represent a unique gene. 19-mer sequences producing no hit to other human genes under the BLASTN search can be selected. During the search, the e-value may be set at a stringent value (such as “1”). Exemplary target sequences derived using these additional conditions (“stringent” criteria) are shown in Table 5. The siRNA sequences for each target sequence (listed in the same row as the target sequence and including the sense strand and the antisense strand) are also indicated in Table 5.
  • The effectiveness of the siRNA sequences listed in Tables 4 and 5, as well as any other RNAi sequence derived according to the present invention, can be evaluated using various methods known in the art. For instance, an siRNA sequence of the present invention can be introduced into a cell that expresses the MRCK1 gene. The polypeptide or mRNA level of the MRCK1 gene in the cell can be detected. A substantial change in the expression level of the MRCK1 gene before and after the introduction of the siRNA sequence is indicative of the effectiveness of the siRNA sequence in suppressing the expression of the MRCK1 gene. In one specific example, the expression levels of other genes are also monitored before and after the introduction of the siRNA sequence. An siRNA sequence which has inhibitory effect on MRCK1 gene expression but does not significantly affect the expression of other genes can be selected. In another specific example, multiple siRNA or other RNAi sequences can be introduced into the same target cell. These siRNA or RNAi sequences specifically inhibit MRCK1 gene expression but not the expression of other genes. In yet another specific example, siRNA or other RNAi sequences that inhibit the expression of both the MRCK1 gene and other gene or genes can be used.
  • TABLE 4
    Exemplary RNAi Target Sequences in the MRCK1 Gene and the
    Corresponding siRNAs (Under Relaxed Criteria)
    Target Sequence siRNA Sense Strand siRNA Antisense Strand
    SEQ ID NO: 10 SEQ ID NO: 11 SEQ ID NO: 12
    SEQ ID NO: 13 SEQ ID NO: 14 SEQ ID NO: 15
    SEQ ID NO: 16 SEQ ID NO: 17 SEQ ID NO: 18
    SEQ ID NO: 19 SEQ ID NO: 20 SEQ ID NO: 21
    SEQ ID NO: 22 SEQ ID NO: 23 SEQ ID NO: 24
    SEQ ID NO: 25 SEQ ID NO: 26 SEQ ID NO: 27
    SEQ ID NO: 28 SEQ ID NO: 29 SEQ ID NO: 30
    SEQ ID NO: 31 SEQ ID NO: 32 SEQ ID NO: 33
    SEQ ID NO: 34 SEQ ID NO: 35 SEQ ID NO: 36
    SEQ ID NO: 37 SEQ ID NO: 38 SEQ ID NO: 39
    SEQ ID NO: 40 SEQ ID NO: 41 SEQ ID NO: 42
    SEQ ID NO: 43 SEQ ID NO: 44 SEQ ID NO: 45
    SEQ ID NO: 46 SEQ ID NO: 47 SEQ ID NO: 48
    SEQ ID NO: 49 SEQ ID NO: 50 SEQ ID NO: 51
    SEQ ID NO: 52 SEQ ID NO: 53 SEQ ID NO: 54
    SEQ ID NO: 55 SEQ ID NO: 56 SEQ ID NO: 57
    SEQ ID NO: 58 SEQ ID NO: 59 SEQ ID NO: 60
    SEQ ID NO: 61 SEQ ID NO: 62 SEQ ID NO: 63
    SEQ ID NO: 64 SEQ ID NO: 65 SEQ ID NO: 66
    SEQ ID NO: 67 SEQ ID NO: 68 SEQ ID NO: 69
    SEQ ID NO: 70 SEQ ID NO: 71 SEQ ID NO: 72
    SEQ ID NO: 73 SEQ ID NO: 74 SEQ ID NO: 75
    SEQ ID NO: 76 SEQ ID NO: 77 SEQ ID NO: 78
    SEQ ID NO: 79 SEQ ID NO: 80 SEQ ID NO: 81
    SEQ ID NO: 82 SEQ ID NO: 83 SEQ ID NO: 84
    SEQ ID NO: 85 SEQ ID NO: 86 SEQ ID NO: 87
    SEQ ID NO: 88 SEQ ID NO: 89 SEQ ID NO: 90
    SEQ ID NO: 91 SEQ ID NO: 92 SEQ ID NO: 93
    SEQ ID NO: 94 SEQ ID NO: 95 SEQ ID NO: 96
    SEQ ID NO: 97 SEQ ID NO: 98 SEQ ID NO: 99
    SEQ ID NO: 100 SEQ ID NO: 101 SEQ ID NO: 102
    SEQ ID NO: 103 SEQ ID NO: 104 SEQ ID NO: 105
    SEQ ID NO: 106 SEQ ID NO: 107 SEQ ID NO: 108
    SEQ ID NO: 109 SEQ ID NO: 110 SEQ ID NO: 111
    SEQ ID NO: 112 SEQ ID NO: 113 SEQ ID NO: 114
    SEQ ID NO: 115 SEQ ID NO: 116 SEQ ID NO: 117
    SEQ ID NO: 118 SEQ ID NO: 119 SEQ ID NO: 120
    SEQ ID NO: 121 SEQ ID NO: 122 SEQ ID NO: 123
    SEQ ID NO: 124 SEQ ID NO: 125 SEQ ID NO: 126
    SEQ ID NO: 127 SEQ ID NO: 128 SEQ ID NO: 129
    SEQ ID NO: 130 SEQ ID NO: 131 SEQ ID NO: 132
    SEQ ID NO: 133 SEQ ID NO: 134 SEQ ID NO: 135
    SEQ ID NO: 136 SEQ ID NO: 137 SEQ ID NO: 138
    SEQ ID NO: 139 SEQ ID NO: 140 SEQ ID NO: 141
    SEQ ID NO: 142 SEQ ID NO: 143 SEQ ID NO: 144
    SEQ ID NO: 145 SEQ ID NO: 146 SEQ ID NO: 147
    SEQ ID NO: 148 SEQ ID NO: 149 SEQ ID NO: 150
    SEQ ID NO: 151 SEQ ID NO: 152 SEQ ID NO: 153
    SEQ ID NO: 154 SEQ ID NO: 155 SEQ ID NO: 156
    SEQ ID NO: 157 SEQ ID NO: 158 SEQ ID NO: 159
    SEQ ID NO: 160 SEQ ID NO: 161 SEQ ID NO: 162
    SEQ ID NO: 163 SEQ ID NO: 164 SEQ ID NO: 165
    SEQ ID NO: 166 SEQ ID NO: 167 SEQ ID NO: 168
    SEQ ID NO: 169 SEQ ID NO: 170 SEQ ID NO: 171
    SEQ ID NO: 172 SEQ ID NO: 173 SEQ ID NO: 174
    SEQ ID NO: 175 SEQ ID NO: 176 SEQ ID NO: 177
    SEQ ID NO: 178 SEQ ID NO: 179 SEQ ID NO: 180
    SEQ ID NO: 181 SEQ ID NO: 182 SEQ ID NO: 183
    SEQ ID NO: 184 SEQ ID NO: 185 SEQ ID NO: 186
    SEQ ID NO: 187 SEQ ID NO: 188 SEQ ID NO: 189
    SEQ ID NO: 190 SEQ ID NO: 191 SEQ ID NO: 192
    SEQ ID NO: 193 SEQ ID NO: 194 SEQ ID NO: 195
    SEQ ID NO: 196 SEQ ID NO: 197 SEQ ID NO: 198
    SEQ ID NO: 199 SEQ ID NO: 200 SEQ ID NO: 201
    SEQ ID NO: 202 SEQ ID NO: 203 SEQ ID NO: 204
    SEQ ID NO: 205 SEQ ID NO: 206 SEQ ID NO: 207
    SEQ ID NO: 208 SEQ ID NO: 209 SEQ ID NO: 210
    SEQ ID NO: 211 SEQ ID NO: 212 SEQ ID NO: 213
    SEQ ID NO: 214 SEQ ID NO: 215 SEQ ID NO: 216
    SEQ ID NO: 217 SEQ ID NO: 218 SEQ ID NO: 219
    SEQ ID NO: 220 SEQ ID NO: 221 SEQ ID NO: 222
    SEQ ID NO: 223 SEQ ID NO: 224 SEQ ID NO: 225
    SEQ ID NO: 226 SEQ ID NO: 227 SEQ ID NO: 228
    SEQ ID NO: 229 SEQ ID NO: 230 SEQ ID NO: 231
    SEQ ID NO: 232 SEQ ID NO: 233 SEQ ID NO: 234
    SEQ ID NO: 235 SEQ ID NO: 236 SEQ ID NO: 237
    SEQ ID NO: 238 SEQ ID NO: 239 SEQ ID NO: 240
    SEQ ID NO: 241 SEQ ID NO: 242 SEQ ID NO: 243
    SEQ ID NO: 244 SEQ ID NO: 245 SEQ ID NO: 246
    SEQ ID NO: 247 SEQ ID NO: 248 SEQ ID NO: 249
    SEQ ID NO: 250 SEQ ID NO: 251 SEQ ID NO: 252
    SEQ ID NO: 253 SEQ ID NO: 254 SEQ ID NO: 255
    SEQ ID NO: 256 SEQ ID NO: 257 SEQ ID NO: 258
    SEQ ID NO: 259 SEQ ID NO: 260 SEQ ID NO: 261
    SEQ ID NO: 262 SEQ ID NO: 263 SEQ ID NO: 264
  • TABLE 5
    Exemplary RNAi Target Sequences in the MRCK1
    Gene and the Corresponding siRNAs (Under Stringent Criteria)
    Target Sequence siRNA Sense Strand siRNA Antisense Strand
    SEQ ID NO: 265 SEQ ID NO: 266 SEQ ID NO: 267
    SEQ ID NO: 268 SEQ ID NO: 269 SEQ ID NO: 270
    SEQ ID NO: 271 SEQ ID NO: 272 SEQ ID NO: 273
    SEQ ID NO: 274 SEQ ID NO: 275 SEQ ID NO: 276
    SEQ ID NO: 277 SEQ ID NO: 278 SEQ ID NO: 279
    SEQ ID NO: 280 SEQ ID NO: 281 SEQ ID NO: 282
    SEQ ID NO: 283 SEQ ID NO: 284 SEQ ID NO: 285
    SEQ ID NO: 286 SEQ ID NO: 287 SEQ ID NO: 288
    SEQ ID NO: 289 SEQ ID NO: 290 SEQ ID NO: 291
    SEQ ID NO: 292 SEQ ID NO: 293 SEQ ID NO: 294
    SEQ ID NO: 295 SEQ ID NO: 296 SEQ ID NO: 297
    SEQ ID NO: 298 SEQ ID NO: 299 SEQ ID NO: 300
    SEQ ID NO: 301 SEQ ID NO: 302 SEQ ID NO: 303
    SEQ ID NO: 304 SEQ ID NO: 305 SEQ ID NO: 306
  • In yet another embodiment, the polynucleotides of the present invention can be modified at the base moiety, sugar moiety or phosphate backbone to improve the stability, hybridization, or solubility of the molecules. For instance, the deoxyribose phosphate backbone of the polynucleotide molecules can be modified to generate peptide polynucleotides (see, Hyrup et al, Bioorganic & Medicinal Chemistry, 4:523, 1996). As used herein, the terms “peptide polynucleotides” or “PNAs” refer to polynucleotide mimics, e.g., DNA mimics, in which the deoxyribose phosphate backbone is replaced by a pseudopeptide backbone and only the four natural nucleobases 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. PNA oligomers can be synthesized using standard solid phase peptide synthesis protocols.
  • PNAs can be used in therapeutic and diagnostic applications. For example, PNAs can be used as antisense agents for sequence-specific modulation of the MRCK1 gene expression. PNAs can also be used in the analysis of single base pair mutations in a gene, (e.g., by PNA-directed PCR clamping); as artificial restriction enzymes when used in combination with other enzymes, (e.g., S1 nucleases); or as probes or primers for DNA sequencing or hybridization.
  • In one embodiment, PNAs can be modified 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 drug delivery techniques known in the art. For example, PNA-DNA chimeras of the polynucleotides of the invention can be generated. These chimeras allow DNA recognition enzymes, such as RNase H and DNA polymerases, to interact with the DNA portion while the PNA portion provides high binding affinity and specificity. PNA-DNA chimeras can be linked using linkers of appropriate lengths which are selected based on base stacking, number of bonds between the nucleobases, and orientations. The PNA-DNA chimeras can be synthesized as follows. A DNA chain is synthesized on a solid support using standard phosphoramidite coupling chemistry and modified nucleoside analogs. PNA monomers are then coupled in a stepwise manner to produce a chimeric molecule with a 5′ PNA segment and a 3′ DNA segment. Alternatively, chimeric molecules can be synthesized with a 5′ DNA segment and a 3′ PNA segment.
  • In other embodiments, the polynucleotides of this invention may include other appended groups such as peptides (e.g., for targeting host cell receptors in vivo), or agents facilitating transportation across the cell membrane or the blood-kidney barrier (see, e.g., PCT Publication No. WO89/10134). In addition, polynucleotides can be modified using hybridization-triggered cleavage agents or intercalating agents. To this end, the polynucleotides can be conjugated to another molecule (e.g., a peptide, hybridization triggered cross-linking agent, transport agent, or hybridization-triggered cleavage agent). Furthermore, the polynucleotide can be detectably labeled.
  • Polypeptides and Variants Thereof
  • Several aspects of the invention pertain to isolated MRCK1 polypeptides and mutated MRCK1 polypeptides capable of inhibiting normal MRCK1 activity. The present invention also contemplates immunogenic polypeptide fragments suitable for raising anti-MRCK1 antibodies.
  • In one embodiment, native MRCK1 polypeptides can be isolated from cells or tissue sources by using standard protein purification techniques. Standard purification methods include electrophoresis, molecular, immunological and chromatographic techniques. Specific examples include ion exchange, hydrophobic, affinity or reverse-phase HPLC chromatography, and chromatofocusing. In one embodiment, MRCK1 polypeptides are purified using a standard affinity column coupled with anti-MRCK1 antibodies. Ultrafiltration and diafiltration techniques can also be used. The degree of purification depends on the purpose of the use of the MRCK1 polypeptides. In some instances, purification is not necessary.
  • In another embodiment, MRCK1 polypeptides or mutated MRCK1 polypeptides capable of inhibiting normal MRCK1 activity are produced by recombinant DNA techniques. Alternative to recombinant expression, MRCK1 polypeptides or mutated MRCK1 polypeptides can be synthesized chemically using standard peptide synthesis techniques.
  • The invention provides MRCK1 polypeptides encoded by the human MRCK1 gene, or homologs thereof. The polypeptides of this invention can be substantially homologous to human MRCK1 kinase (SEQ ID NO:2). Preferably, these polypeptides retain the biological activity of the native MRCK1 kinase. In one embodiment, the polypeptides comprise an amino acid sequence which is at least about 91%, 95%, 98%, 99% or more homologous to SEQ ID NO:2.
  • Comparison of sequences and determination of percent identity between two sequences can be accomplished using a mathematical algorithm. The percent identity between two amino acid sequences can be determined using the Needleman and Wunsch (J. Mol. Biol. 48:444-453, 1970) algorithm, or the GAP program in the GCG software package which uses either a Blossom 62 matrix or a PAM250 matrix, and a gap weight of 16, 14, 12, 10, 8, 6, or 4 and a length weight of 1, 2, 3, 4, 5, or 6. The percent identity between two nucleotide sequences can be determined using the GAP program in the GCG software package, which uses a NWSgapdna.CMP matrix and a gap weight of 40, 50, 60, 70, or 80 and a length weight of 1, 2, 3, 4, 5, or 6. The percent identity between two amino acid or nucleotide sequences can also be determined using the algorithm of E. Meyers and W. Miller (CABIOS, 4:11-17, 1989) which has been incorporated into the ALIGN program (version 2.0), or the pairwise BLAST program available at NCBI's BLAST web site.
  • The polypeptide and polynucleotide sequences of the present invention can be used as query sequences for searching public databases in order to identify similar sequences. The search can be conducted using BLAST programs, such as the protein BLAST, nucleotide BLAST, pairwise BLAST, and genomic BLAST, that are available at the BLAST web site maintained by the NCBI. When using BLAST programs, the default parameters of the respective programs can also be used.
  • The invention further provides chimeric or fusion MRCK1 polypeptides. A fusion MRCK1 polypeptide contains an MRCK1-related polypeptide and a non-MRCK1 polypeptide. The MRCK1-related polypeptides include all or a portion of SEQ ID NO:2 or its variant. A peptide linker sequence can be employed to separate the MRCK1-related polypeptide from the non-MRCK1 polypeptide components by a distance sufficient to ensure that each polypeptide folds into its native secondary and tertiary structures. Such a peptide linker sequence is incorporated into the fusion protein using standard techniques well-known in the art. Suitable peptide linker sequences can be chosen based on the following factors: (1) their ability to adopt a flexible extended conformation; (2) their inability to adopt a secondary structure that could interact with functional epitopes on the MRCK1-related polypeptide and non-MRCK1 polypeptide; and (3) the lack of hydrophobic or charged residues that might react with the polypeptide functional epitopes. Preferred peptide linker sequences contain Gly, Asn and Ser residues. Other near neutral amino acids, such as Thr and Ala can also be used in the linker sequence. Amino acid sequences suitable as linkers include those disclosed in Maratea et al., Gene, 40:39-46, 1985; Murphy et al., Proc. Natl. Acad. Sci. USA, 83:8258-8262, 1986; and U.S. Pat. Nos. 4,935,233 and 4,751,180. The linker sequences may be from 1 to about 50 amino acids in length. Linker sequences are not required when the MRCK1-related polypeptide or the non-MRCK1 polypeptide has non-essential N-terminal amino acid regions that can be used to separate the respective functional domains and thereby prevent steric interference.
  • In one embodiment, the fusion protein is a GST-MRCK1 fusion protein in which an MRCK1-related sequence, such as SEQ ID NO:2, is fused to the C-terminus of the GST sequence. This fusion protein can facilitate the purification of the recombinant MRCK1.
  • The MRCK1-fusion proteins of the invention can be incorporated into pharmaceutical compositions and administered to a subject. The MRCK1-fusion proteins can be used to affect the bioavailability of an MRCK1 substrate. The MRCK1-fusion proteins can also be used for the treatment or prevention of damages caused by (i) aberrant modification or mutation of MRCK1, or (ii) aberrant post-translational modification of MRCK1. It is also conceivable that a fusion protein containing a normal or mutated MRCK1 polypeptide, or a fragment thereof, can be used to inhibit MRCK1 activity in a human subject.
  • Moreover, the MRCK1-fusion proteins can be used as immunogens to produce anti-MRCK1 antibodies. They can also be used to purify MRCK1 ligands and to screen for molecules capable of inhibiting the interaction between MRCK1 and its substrates.
  • Preferably, the MRCK1-chimeric or fusion proteins of the invention are produced using standard recombinant DNA techniques. Commercially available expression vectors which encode a fusion moiety (e.g., a GST polypeptide) can be used.
  • A signal sequence can be used to facilitate secretion and isolation of the secreted protein or other proteins of interest. Signal sequences are typically characterized by a core of hydrophobic amino acids which are generally cleaved from the mature protein. Such signal peptides contain processing sites that allow cleavage of the signal sequence from the mature proteins as they pass through the secretory pathway. The present invention encompasses MRCK1 polypeptides having a signal sequence, or the polynucleotide sequences encoding the same.
  • The present invention also pertains to MRCK1 mutants which function as antagonists to MRCK1. In one embodiment, antagonists of MRCK1 are used as therapeutic agents. For example, a mutant of MRCK1 that forms a non-functional dimer with a wide-type MRCK1 (the so-called dominant negative mutant) can decrease the activity of MRCK1 and may ameliorate diseases in a subject wherein MRCK1 are abnormally increased in level or activity. Dominant negative MRCK1 mutants can be generated by mutagenesis, as appreciated by one skilled in the art.
  • MRCK1 mutants which function as either MRCK1 agonists or antagonists can be identified by screening combinatorial libraries of mutants. A variegated library of MRCK1 mutants can be produced by, for example, enzymatically ligating a mixture of synthetic oligonucleotides into gene sequences such that a degenerate set of potential MRCK1 sequences is expressible as individual polypeptides, or alternatively, as a set of larger fusion proteins containing the set of MRCK1 sequences therein. There are a variety of methods which can be used to produce libraries of potential MRCK1 mutants from a degenerate oligonucleotide sequence. A degenerate gene sequence can be chemically synthesized using an automatic DNA synthesizer. The synthetic gene can then be ligated into an appropriate expression vector.
  • In one embodiment, a library of coding sequences can be generated using nucleases. For instance, double-stranded PCR fragments of the MRCK1 coding sequence can be treated by a nuclease which produces about one nick per molecule. The double-stranded DNAs then are subject to a cycle of denaturing and re-naturing. The newly reformed DNAs, which may include sense/antisense pairs from different nicked products, are treated with S1 nuclease to remove single-stranded portions. Using this method, an expression library which encodes N-terminal, C-terminal or internal fragments of MRCK1 can be derived.
  • In addition, recursive ensemble mutagenesis (REM), a technique which enhances the frequency of functional mutants in the libraries, can be used to prepare MRCK1 mutants (Delgrave et al., Protein Engineering, 6:327-331, 1993).
  • MRCK1 fragments, or variants thereof, can also be generated using synthetic means, such as solid-phase synthesis methods. Preferably, the synthesized fragment has less than about 100 amino acids, or preferably, less than about 50 amino acids.
  • Antibodies
  • In accordance with another aspect of the present invention, antibodies specific to MRCK1 or its variants are prepared. An antibody is considered to bind “specifically” to an antigen if the binding affinity between the antibody and the antigen is equal to, or greater than 105 M−1. The antibodies can be monoclonal or polyclonal. Preferably, the antibodies are monoclonal. More preferably, the antibodies are humanized antibodies.
  • Polyclonal anti-MRCK1 antibodies can be prepared by immunizing a suitable subject with MRCK1 or fragments thereof. The anti-MRCK1 antibody titer in the immunized subject can be monitored over the time using standard techniques, such as ELISA. The anti-MRCK1 antibody can be isolated from the immunized subject using well-known techniques.
  • In one embodiment, hybridomas capable of producing anti-MRCK1 antibodies are prepared. Purified MRCK1 or its variants, or fragments thereof, are used to immunize a vertebrate, such as a mammal. Suitable mammals include mice, rabbits and sheep. Preferably, the fragment used for immunization comprises at least 8 amino acid residues, more preferably at least 12 amino acid residues, highly preferably at least 16 amino acid residues, and most preferably at least 20 amino acid residues.
  • Immunogenic fragments (epitopes) of MRCK1 can be identified using well-known techniques. In general, any fragment of SEQ ID NO:2 can be used to raise antibodies specific to MRCK1. Preferred epitopes are regions that are located on the surface of MRCK1. These regions are usually hydrophilic.
  • Splenocytes are isolated from the immunized vertebrate and fused with an immortalized cell line (such as a myeloma) to form hybridomas. Preferably, the immortal cell line is derived from the same mammalian species as the lymphocytes. For example, murine hybridomas can be made by fusing an immortalized mouse cell line with lymphocytes isolated from a mouse that is immunized with an immunogenic preparation of the present invention. Preferred immortalized cell lines include mouse myeloma cell lines that are sensitive to culture medium containing hypoxanthine, aminopterin and thymidine (“HAT medium”). Suitable myeloma cell lines include, but are not limited to, the P3-NS1/1-Ag4-1, P3-x63-Ag8.653 or Sp210-Ag14 myeloma lines, all of which are available from ATCC. In one embodiment, HAT-sensitive mouse myeloma cells are fused to mouse splenocytes using polyethylene glycol (“PEG”). Hybridoma cells thus produced are selected against HAT medium, which kills unfused or unproductively fused myeloma cells. Hybridoma cells which produce monoclonal anti-MRCK1 antibodies are then detected by screening the hybridoma culture supernatants.
  • A monoclonal anti-MRCK1 antibody can also be prepared by screening a recombinant combinatorial immunoglobulin library (e.g., an antibody phase display library). Kits for generating and screening phage display libraries are commercially available (e.g., the Pharmacia Recombinant Phage Antibody System, Catalog No. 27-9400-01; and the Stratagene SurfZAP™ Phage Display Kit, Catalog No. 240612).
  • The anti-MRCK1 antibodies of the present invention also include “single-chain Fv” or “scFv.” The scFv fragments comprise the VH and VL domains of an antibody. Generally, the scFv polypeptide further comprises a polypeptide linker between the VH and VL domains. The polypeptide linker enables the scFv to form the desired structure for antigen binding. Additionally, recombinant anti-MRCK1 antibodies, such as chimeric and humanized monoclonal antibodies, can be prepared, as appreciated by one of ordinary skill in the art.
  • Humanized antibodies are particularly desirable for therapeutic treatment of human subjects. Humanized forms of non-human (e.g., murine) antibodies are chimeric immunoglobulins, immunoglobulin chains, or fragments thereof (such as Fv, Fab, Fab′, F(ab)2 or other antigen-binding subsequences of antibodies) which contain minimal sequence derived from non-human immunoglobulin. Humanized antibodies are derived from human immunoglobulins in which the residues forming the complementary determining regions (CDRs) are replaced by the residues from CDRs of a non-human antibody, such as a mouse, rat or rabbit antibody having the desired specificity, affinity and capacity. In some instances, Fv framework residues of the human immunoglobulin are replaced by corresponding non-human residues. Humanized antibodies may also comprise residues which are found neither in the recipient antibody nor in the imported CDR or framework sequences. The humanized antibody can comprise at least one or 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 constant regions are those of a human immunoglobulin consensus sequence. The humanized antibody preferably comprises at least a portion of an immunoglobulin constant region (Fc) of a human immunoglobulin.
  • Humanized antibodies can be produced using transgenic mice which are incapable of expressing endogenous immunoglobulin heavy and light chains but can express human heavy and light chains. The transgenic mice are immunized in the normal fashion with a selected antigen. Monoclonal antibodies directed against the antigen can be obtained using conventional hybridoma technology. The human immunoglobulin transgenes harbored in the transgenic mice rearrange during B cell differentiation, and subsequently undergo class switching and somatic mutation. Using this technique, therapeutically useful IgG, IgA and IgE antibodies can be prepared.
  • In addition, humanized antibodies which recognize a selected epitope can be generated using a technique referred to as “guided selection.” In this approach a selected non-human monoclonal antibody, e.g., a murine antibody, is used to guide the selection of a humanized antibody recognizing the same epitope.
  • In a preferred embodiment, the antibodies to MRCK1 are capable of reducing or eliminating the biological function of MRCK1. Preferably, the antibodies reduce at least 25% of MCRK1 activity. More preferably, the antibodies reduce at least about 50% of the activity. Highly preferably, the antibodies reduce about 95-100% of MRCK1 activity.
  • Anti-MRCK1 antibodies can be used to isolate MRCK1. Suitable methods include affinity chromatography and immunoprecipitation. Moreover, anti-MRCK1 antibodies can be used to evaluate the expression level of MRCK1. Anti-MRCK1 antibodies can also be used to monitor MRCK1 level as part of a clinical testing procedure, or to determine the efficacy of a given treatment regimen. Detection can be facilitated by coupling 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, galactosidase, or acetylcholinesterase; examples of suitable prosthetic group complexes include streptavidin/biotin and avidin/biotin; examples of suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin; an example of a luminescent material includes luminol; examples of bioluminescent materials include luciferase, luciferin, and aequorin; and examples of suitable radioactive materials include 125I, 131I, 35S or 3H.
  • Anti-MRCK1 antibodies are also useful for targeting a therapeutic agent/drug to a particular cell or tissue. The therapeutic agent/drug may be coupled to an antibody, either covalently or non-covalently. For instance, a therapeutic agent can be coupled to an antibody via a linker group. A linker group can function as a spacer to separate the antibody from the agent so as to avoid interference with antibody's binding capabilities. The linker group can also serve to increase the chemical reactivity of a substituent on the agent or the antibody, and thus increase the coupling efficiency. A variety of bifunctional or polyfunctional reagents, either homo- or hetero-functional (such as those described in the catalog of the Pierce Chemical Co., Rockford, Ill.), can be employed as the linker group. Coupling may be effected, for example, through amino groups, carboxyl groups, sulfhydryl groups or oxidized carbohydrate residues. There are numerous references describing this methodology. See e.g., U.S. Pat. No. 4,671,958.
  • Where a therapeutic agent is more potent when free from the antibody, it may be desirable to use a linker group which is cleavable during or upon internalization into the target cell. A number of different cleavable linker groups have been described. The mechanisms for the intra-cellular release of an agent from these linker groups include cleavage by reduction of a disulfide bond (e.g., U.S. Pat. No. 4,489,710), by irradiation of a photolabile bond (e.g., U.S. Pat. No. 4,625,014), by hydrolysis of derivatized amino acid side chains (e.g., U.S. Pat. No. 4,638,045), by serum complement-mediated hydrolysis (e.g., U.S. Pat. No. 4,671,958), or by acid-catalyzed hydrolysis (e.g., U.S. Pat. No. 4,569,789).
  • It may also be desirable to couple more than one agent to an antibody. In one embodiment, multiple agents are coupled to one antibody molecule. In another embodiment, at least two different types of agents are coupled to one antibody. Regardless of the particular embodiment, immunoconjugates coupled with more than one agent can be prepared in a variety of ways, as appreciated by one of ordinary skill in the art.
  • Vectors, Expression Vectors and Gene Delivery Vectors
  • Another aspect of the invention pertains to vectors containing a polynucleotide encoding MRCK1 or a portion thereof. One type of vector is a “plasmid,” which includes a circular double-stranded DNA into which additional DNA segments can be introduced. Vectors also include expression vectors and gene delivery vectors.
  • The expression vectors of the present invention comprise a polynucleotide encoding MRCK1 or a portion thereof. The expression vectors also include one or more regulatory sequences operably linked to the polynucleotide being expressed. These regulatory sequences are selected based on the type of host cells. It will be appreciated by those skilled in the art that the design of the expression vector depends on such factors as the choice of the host cells and the desired expression levels. MRCK1 can be expressed in bacterial cells such as E. coli, insect cells (using baculovirus expression vectors), yeast cells or mammalian cells. The expression vector can also be transcribed and translated in vitro, for example, by using T7 promoter regulatory sequences and T7 polymerase. Expression of proteins in prokaryotes is most often carried out in E. 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 purposes: 1) to increase expression of the recombinant protein; 2) to increase the solubility of the recombinant protein; and 3) 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. Suitable cleavage enzymes include Factor Xa, thrombin and enterokinase. Examples of fusion expression vectors include pGEX (Pharmacia Piscataway, N.J.), pMAL (New England Biolabs, Beverly, Mass.) and pRITS (Pharmacia, Piscataway, N.J.). Purified fusion proteins can be utilized in MRCK1 activity assays, or to generate antibodies specific for MRCK1.
  • Examples of suitable inducible non-fusion E. coli expression vectors include pTrc and pET 11d. Target gene expression from the pTrc vector relies on host RNA polymerase transcription from a hybrid trp-lac fusion promoter. Target gene expression from the pET 11d vector relies on transcription from a T7 gn10-lac fusion promoter mediated by a co-expressed viral RNA polymerase (T7 gn1). This viral polymerase is supplied by host strains BL21(DE3) or HSLE174(DE3) from a resident prophage harboring a T7 gn1 gene under the transcriptional control of the lacUV 5 promoter.
  • One strategy to maximize recombinant protein expression in E. coli is to express the protein in host bacteria that have an impaired capacity to proteolytically cleave the recombinant protein. Another strategy is to alter the polynucleotide sequence encoding the protein so that the individual codons for each amino acid are those preferentially utilized in E. coli.
  • In another embodiment, the MRCK1 expression vector is a yeast expression vector. Examples of yeast expression vectors include pYepSec1, pMFa, pJRY88, pYES2 (Invitrogen Corporation, San Diego, Calif.), and picZ (Invitrogen Corp, San Diego, Calif.).
  • Alternatively, MRCK1 or its variant can be expressed in insect cells using baculovirus expression vectors. Suitable baculovirus vectors include the pAc series and the pVL series.
  • In yet another embodiment, MRCK1 or its variant is expressed in mammalian cells using a mammalian expression vector. Examples of mammalian expression vectors include pCDM8 and pMT2PC. 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.
  • In another embodiment, the mammalian expression vector contains tissue-specific regulatory elements. Examples of suitable tissue-specific promoters include the liver-specific albumin promoter, lymphoid-specific promoters, promoters of T cell receptors and immunoglobulins, neuron-specific promoters (e.g., the neurofilament promoter), pancreas-specific promoters, and mammary gland-specific promoters (e.g., milk whey promoter). Developmentally-regulated promoters are also contemplated, which include, for example, the α-fetoprotein promoter.
  • The present invention also provides a recombinant expression vector comprising a polynucleotide which encodes MRCK1 but is cloned into the expression vector in an antisense orientation. Regulatory sequences that are operatively linked to the antisense-oriented polynucleotide can be chosen to direct continuous expression of the antisense RNA molecule in a variety of cell types. Suitable regulatory sequences include viral promoters and/or enhancers. Regulatory sequences can also be chosen to direct constitutive, tissue specific or cell type specific expression of the antisense RNA. The antisense expression vector can be in the form of a recombinant plasmid, phagemid, or attenuated virus in which antisense polynucleotides are produced under the control of a highly efficient regulatory region.
  • The present invention further provides gene delivery vehicles for delivering polynucleotides to mammals. A polynucleotide sequence of the invention can be administered either locally or systemically via a gene delivery vehicle. Expression of the polynucleotide can be induced using endogenous mammalian or heterologous promoters. Expression of the polynucleotide in vivo can be either constituted or regulated. The gene delivery vehicles preferably are viral vectors, including retroviral, lentiviral, adenoviral, adeno-associated viral (AAV), herpes viral, or alphavirus vectors. The viral vectors can also be astrovirus, coronavirus, orthomyxovirus, papovavirus, paramyxovirus, parvovirus, picornavirus, poxvirus, or togavirus vectors.
  • Delivery of gene therapy constructs is not limited to the above mentioned viral vectors. Other delivery methods can also be employed. These methods include nucleic acid expression vectors, polycationic condensed DNA linked or unlinked to killed adenovirus, ligand linked DNA, liposome-DNA conjugates, gene guns, ionizing radiation, nucleic charge neutralization, or fusion with cell membranes. Naked DNA can also be employed. Uptake efficiency of the naked DNA may be improved using biodegradable latex beads. This method can be further improved by treating the beads to increase their hydrophobicity.
  • Regulatable Expression Systems
  • Another aspect of the present invention pertains to the use of regulatable expression systems to express desirable polynucleotides or polypeptides in cells. Systems suitable for this invention are briefly described below:
  • Tet-on/off system. The Tet-system is based on two regulatory elements derived from the tetracycline-resistance operon of the E. coli Tn10 transposon: the tet repressor protein (TetR) and the Tet operator DNA sequence (tetO) to which TetR binds (Gossen et al., Science, 268: 1766-1769, 1995). The system consists of two components, a “regulator” and a “reporter” plasmid. The “regulator” plasmid encodes a hybrid protein containing a mutated Tet repressor (rtetR) fused to the VP16 activation domain of herpes simplex virus. The “reporter” plasmid contains a tet-responsive element (TRE), which controls the “reporter” gene of choice. The rtetR-VP16 fusion protein can only bind to the TRE, therefore activating the transcription of the “reporter” gene in the presence of tetracycline. The system has been incorporated into a number of viral vectors including retrovirus, adenovirus and AAV.
  • Ecdysone system. The ecdysone system is based on the molting induction system found in Drosophila, but modified for inducible expression in mammalian cells. The system uses an analog of the Drosophila steroid hormone ecdysone, muristerone A, to activate expression of the gene of interest via a heterodimeric nuclear receptor. Expression levels have been reported to exceed 200-fold over basal levels with no effect on mammalian cell physiology (No et al., Proc. Natl. Acad. Sci. USA, 93: 3346-3351, 1996).
  • Progesterone-system. The progesterone receptor is normally stimulated to bind to a specific DNA sequence and to activate transcription through an interaction with its hormone ligand. Conversely, the progesterone antagonist mifepristone (RU486) is able to block hormone-induced nuclear transport and subsequent DNA binding. A mutant form of the progesterone receptor that can be stimulated to bind through an interaction with RU486 has been generated. To generate a specific, regulatable transcription factor, the RU486-binding domain of the progesterone receptor has been fused to the DNA-binding domain of the yeast transcription factor GAL4 and the transactivation domain of the HSV protein VP16. The chimeric factor is inactive in the absence of RU486. The addition of hormone, however, induces a conformational change in the chimeric protein, and this change allows binding to a GAL4-binding site and the activation of transcription from promoters containing the GAL4-binding site (Wang et al., Nat. Biotech., 15: 239-243, 1997).
  • Rapamycin-system. Immunosuppressive agents, such as FK506 and rapamycin, act by binding to specific cellular proteins and facilitating their dimerization. For example, the binding of rapamycin to FK506-binding protein (FKBP) results in its heterodimerization with another rapamycin binding protein FRAP, which can be reversed by removal of the drug. The ability to bring two proteins together by addition of a drug potentiates the regulation of a number of biological processes, including transcription. A chimeric DNA-binding domain has been fused to the FKBP, which enables binding of the fusion protein to a specific DNA-binding sequence. A transcriptional activation domain also has been fused to FRAP. When these two fusion proteins are co-expressed in the same cell, a fully functional transcription factor can be formed by heterodimerization mediated by addition of rapamycin. The dimerized chimeric transcription factor can then bind to a synthetic promoter sequence containing copies of the synthetic DNA-binding sequence. This system has been successfully integrated into adenoviral and AAV vectors. Long term regulatable gene expression has been achieved in both mice and baboons (Ye et al., Science, 283: 88-91, 1999).
  • Detection Methods
  • In patients with disorders related to the aberrant expression of MRCK1. The expression level of MRCK1 can be used as an indicator for detecting the presence of MRCK1-related diseases in humans. Detection and measurement of the relative amount of the MRCK1 gene product can be carried out using various methods known in the art.
  • Typical methodologies for detecting the transcription level of a gene include extracting RNA from a cell or tissue sample, hybridizing a labeled probe to the extracted RNA or derivative thereof (such as cDNA or cRNA), and detecting the probe. Suitable methods include Northern Blot and quantitative PCR or RT-PCR. In situ hybridization can also be used to detect the transcription level of the MRCK1 gene in human tissues.
  • Typical methodologies for detecting a polypeptide include extracting proteins from a cell or tissue sample, binding an antibody to the target polypeptide and detecting the antibody. Suitable methods include enzyme linked immunosorbent assays (ELISAs), Western blots, immunoprecipitations, and immunofluorescence. The antibody can be polyclonal, or preferably, monoclonal. The antibody can be an intact antibody, or a fragment thereof (e.g. Fab or F(ab′)2). The antibody can be labeled with a radioisotope, a fluorescent compound, an enzyme, an enzyme co-factor, or a detectable ligand. The term “labeled,” with regard to a probe or antibody, is intended to encompass direct labeling such as through covalent coupling, as well as indirect labeling such as being mediated by another reagent which is directly labeled. Examples of indirect labeling include labeling a primary antibody using a fluorescently labeled secondary antibody, or attaching a DNA probe with a biotin which can be detected, for example, by a fluorescence-labeled streptavidin.
  • Preferably, the binding affinity of the antibody to MRCK1 is at least 105 M−1. More preferably, the binding affinity is at least 106 M−1. Other methods such as electrophoresis, chromatography or direct sequencing can also be used to detect the amount of a polypeptide in a biological sample. Anti-MRCK1 antibodies can also be directly introduced into a subject. The antibody can be labeled with a radioactive marker whose presence and location in the subject can be detected using standard imaging techniques.
  • In one embodiment, the genomic copies of the MRCK1 gene in the genome of a human subject may indicate the presence or predisposition of a disease. Detection of the presence or number of copies of the MRCK1 gene in the genome can be performed using methods known in the art. For instance, it can be assessed using Southern Blot. The probes for Southern Blot can be labeled with a radioisotope, a fluorescent compound, an enzyme, or an enzyme co-factor.
  • In the field of diagnostic assays, the above-described detection methods can be used to determine the severity of MRCK1-related diseases. A biological sample is isolated from a test subject, and the presence, quantity and/or activity of MRCK1 in the sample relative to a normal or control sample is evaluated. The expression level of MRCK1 in the biological sample can indicate the presence or severity of MRCK1-related diseases in the test subject. The term “biological sample” is intended to include tissues, cells or biological fluids isolated from the subject. A preferred biological sample is a serum sample isolated from the subject using conventional means.
  • Screening Methods
  • The present invention also provides methods for identifying MRCK1 modulators. The activity of MRCK1 can be evaluated using various methods, such as those disclosed in Leung et al., Mol. Cell. Biol., 18:130-140, 1998, and Chen et al., J. Biol. Chem., 274: 19901-19905, 1999, both of which are incorporated herein by reference.
  • Suitable modulators include compounds or agents comprising therapeutic moieties, such as peptides, peptidomimetics, peptoids, polynucleotides, small molecules or other drugs. These moieties can either bind to MRCK1, or have a modulatory (e.g., stimulatory or inhibitory) effect on the activity of MRCK1. In one embodiment, the moieties have a modulatory effect on the interactions of MRCK1 with one or more of its natural substrates. These moieties can also exert a modulatory effect on the expression of MRCK1. The screen assays of the present invention comprise detecting the interactions between MRCK1 and test components.
  • The test compounds of the present invention can be either small molecules or bioactive agents. In a preferred embodiment, the test compound is a small organic or inorganic molecule. In another preferred embodiment, the test compound is a polypeptides, oligopeptides, polysaccharides, nucleotides or polynucleotides.
  • In accordance with one aspect of this invention, methods for screening for compounds that inhibit the biological activities of MRCK1 are provided. Pharmaceutical compositions comprising these compounds can subsequently be prepared. The screening method comprises (1) contacting a sample with a compound, and (2) comparing expression profile or biological activity of MRCK1 in the sample to determine whether the compound substantially decreases the expression level or activities of MRCK1. The screening method can be carried out either in vivo or in vitro.
  • The present invention further includes a method for screening for compounds capable of modulating the binding between MRCK1 and a binding partner. As used herein, the term “binding partner” refers to a bioactive agent which serves as either a substrate for MRCK1, or a ligand having a binding affinity to MRCK1. The bioactive agent may be selected from a variety of naturally-occurring or synthetic compounds, proteins, peptides, polysaccharides, nucleotides or polynucleotides.
  • Inhibitors of the expression, activity or binding ability of MRCK1 may be used as therapeutic compositions. These inhibitors can be formulated in suitable pharmaceutical compositions, as described herein below.
  • The present invention also provides methods for conducting high-throughput screening for compounds capable of inhibiting activity or expression of MRCK1. In one embodiment, the high-throughput screening method involves contacting test compounds with MRCK1, and then detecting the effect of the test compounds on MRCK1. Functional assays, such as cytosensor microphysiometer-based assays, calcium flux assays (e.g. FLIPR®, Molecular Devices Corp, Sunnyvale, Calif.), or the TUNEL assay, can be employed to measure MRCK1 cellular activity. Fluorescence-based techniques can be used for high-throughput and ultra high-throughput screening. They include, but are not limited to, BRET® and FRET® (both by Packard Instrument Co., Meriden, Conn.).
  • In a preferred embodiment, the high-throughput screening assay uses label-free plasmon resonance technology as provided by BIACORE® systems (Biacore International AB, Uppsala, Sweden). Plasmon free resonance occurs when surface plasmon waves are excited at a metal/liquid interface. By reflecting directed light from the surface as a result of contact with a sample, the surface plasmon resonance causes a change in the refractive index at the surface layer. The refractive index change for a given change of mass concentration at the surface layer is similar for many bioactive agents (including proteins, peptides, lipids and polynucleotides), and since the BIACORE® sensor surface can be functionalized to bind a variety of these bioactive agents, detection of a wide selection of test compounds can thus be accomplished.
  • Monitoring Efficacy of a Drug During Clinical Trials
  • Using the MRCK1 detection methods of this invention, the efficacy of a therapeutic agent for MRCK1-related diseases can be monitored during clinical trials. The therapeutic agent may be a drug, small molecule, agonist, antagonist, peptidomimetic, protein, peptide, or polynucleotide. The changes in the expression or activity of the MRCK1 gene in response to the treatment of the agent can be used to evaluate the therapeutic effect of the agent on patients with MRCK1-related diseases. In addition, the expression or activity of MRCK1 in response to the agent can be measured at various points during the clinical trial.
  • In a preferred embodiment, the method for monitoring the effectiveness of the therapeutic agent includes the steps of (i) obtaining a pre-administration sample from a subject; (ii) detecting the level of expression or activity of MRCK1 in the pre-administration sample; (iii) obtaining one or more post-administration samples from the subject; (iv) detecting the level of expression or activity of MRCK1 in the post-administration samples; (v) comparing the level of expression or activity of MRCK1 in the pre-administration sample to the level of expression or activity of MRCK1 in the post administration samples. The dose or frequency of the administration of the agent may be adjusted based on the effectiveness of the agent in a particular patient. Therefore, MRCK1 expression or activity can be used as an indicator of the effectiveness of a therapeutic agent for MRCK1-related diseases, even if the agent does not produce an observable phenotypic response.
  • Prognostic Assays
  • The detection methods described herein can be used to identify subjects having or at risk of developing MRCK1-related diseases. In addition, the detection methods can be used to determine whether an agent (e.g., an agonist, antagonist, peptidomimetic, protein, peptide, polynucleotide, small molecule, or other drug candidate) can be administered to a subject for effectively treating or preventing MRCK1-related diseases.
  • MRCK1 expression profiles at different progression stages of MRCK1-related diseases can be established. In addition, MRCK1 expression profiles in different patients who have different responses to a drug treatment are determined. A pattern may emerge such that a particular expression profile may be correlated to an increased likelihood of a poor prognosis. Therefore, the prognostic assay of the present invention may be used to determine whether a subject undergoing a treatment for an MRCK1-related disease has a poor outlook for long term survival or disease progression. Preferably, prognosis is performed shortly after diagnosis, such as within a few days after diagnosis. The result of prognosis can then be used to devise individualized treatment program, thereby enhancing the effectiveness of the treatment as well as the likelihood of long-term survival and well being.
  • The method of the invention can also be used to detect genetic alterations in the MRCK1 gene, thereby determining if a subject with the altered gene is at risk for damages characterized by aberrant regulation in MRCK1 activity or expression. In a preferred embodiment, the method includes detecting the presence or absence of a genetic alteration that affects the integrity of the MRCK1 gene, or detecting the aberrant expression of the MRCK1 gene. The genetic alteration can be detected by ascertaining the existence of at least one of the following: 1) deletion of one or more nucleotides from the MRCK1 gene; 2) addition of one or more nucleotides to the MRCK1 gene; 3) substitution of one or more nucleotides of the MRCK1 gene, 4) a chromosomal rearrangement in the MRCK1 gene; 5) alteration in the level of a messenger RNA transcript of the MRCK1 gene, 6) aberrant modification of the MRCK1 gene, 7) the presence of a non-wild-type splicing pattern of a messenger RNA transcript of the MRCK1 gene, 8) non-wild-type level MRCK1, 9) allelic loss of an MRCK1 gene, and 10) inappropriate post-translational modification of MRCK1.
  • In one embodiment, detection of the alteration involves the use of a probe/primer in a polymerase chain reaction (such as anchor PCR or RACE PCR) or alternatively, in a ligation chain reaction (LCR). LCR can be particularly useful for detecting point mutations in the MRCK1 gene. This method includes the steps of collecting a sample from a subject, isolating polynucleotides (e.g., genomic DNA, mRNA, or both) from the sample, contacting the polynucleotide with one or more primers which specifically hybridize to the MRCK1 gene or gene product, and detecting the presence or absence of an amplification product, or detecting the size of the amplification product and comparing its length to a control. It is understood that PCR and/or LCR can be used as a preliminary amplification step in conjunction with any other techniques described herein.
  • Alternative amplification methods include: self sustained sequence replication (Guatelli et al., Proc. Natl. Acad. Sci. USA, 87:1874-1878, 1990), transcriptional amplification system (Kwoh et al., Proc. Natl. Acad. Sci. USA, 86:1173-1177, 1989), and Q-Beta Replicase (Lizardi et al., Bio-Technology, 6:1197, 1988).
  • In another embodiment, mutations in the MRCK1 gene can be identified using restriction enzymes. Differences in restriction enzyme digestion patterns indicates mutation(s) in the MRCK1 gene or its transcripts. Moreover, sequence specific ribozymes can be used to detect the presence of specific mutations. See e.g., U.S. Pat. No. 5,498,531.
  • In yet another embodiment, genetic mutations in the MRCK1 gene can be identified using high density arrays which contain a large number of oligonucleotides probes. For example, genetic mutations in the MRCK1 gene can be identified in two dimensional arrays. In this example, a first hybridization array of probes is used to scan through long stretches of DNA in a sample and a control in order to identify base changes between the two sequences. This step allows the identification of point mutations. This step is followed by a second hybridization array that allows the characterization of specific mutations by using smaller and specialized probe arrays which are complementary to all variants or mutations detected. Each mutation array is composed of parallel probe sets, one complementary to the wild-type gene and the other complementary to the mutant gene.
  • In still another embodiment, any sequencing reactions known in the art can be used to directly sequence the MRCK1 gene in order to detect mutations. It is contemplated that any automated sequencing procedures can be utilized, including sequencing by mass spectrometry.
  • In one embodiment, protection from cleavage agents is used to detect mismatched bases in RNA/RNA or RNA/DNA heteroduplexes. In general, the “mismatch cleavage” technique involves forming heteroduplexes by hybridizing an RNA or DNA (labeled) containing the wild-type MRCK1 gene sequence to a potentially mutant RNA or DNA obtained from a tissue sample. The double-stranded duplexes are treated with an agent which cleaves single-stranded regions of the duplex. The agent may be RNase (for RNA/DNA duplexes), or S1 nuclease (for DNA/DNA hybrids). In one case, either DNA/DNA or RNA/DNA duplexes are treated with piperidine and hydroxylamine, or piperidine and osmium tetroxide, in order to digest mismatched regions. After the digestion, the resulting material is separated by size on a denaturing polyacrylamide gel from which the site(s) of mutation may be determined.
  • In a preferred embodiment, the mismatch cleavage reaction employs one or more proteins that recognize mismatched base pairs in double-stranded DNA. Examples of these proteins include “DNA mismatch repair” enzymes. For instance, 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. In one case, cDNAs are prepared from mRNAs isolated from test cells. The cDNAs are then hybridized to a probe derived from the MRCK1 gene. The duplex thus formed 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. Pat. No. 5,459,039.
  • In another embodiment, alterations in electrophoretic mobility are used to identify mutations in the MRCK1 gene. Differences in electrophoretic mobility between mutant and wild-type polynucleotides can be detected using single strand conformation polymorphism (SSCP). The resulting alteration in electrophoretic mobility enables the detection of a single base change. The DNA fragments can be labeled or detected with probes. In one case, the sensitivity of the assay is enhanced by using RNA, in which the secondary structure is more sensitive to a change in sequence. In a preferred embodiment, the assay utilizes heteroduplex analysis to separate double-stranded heteroduplex molecules on the basis of changes in electrophoretic mobility (Keen et al., Trends Genet., 7:5, 1991).
  • In yet another embodiment, the movement of mutant or wild-type fragments is evaluated using denaturing gradient gel electrophoresis (DGGE). For this purpose, DNA fragments can be modified to insure that they do not completely denature. For instance, a GC clamp of approximately 40 GC-rich base pairs can be added to the DNA fragment using PCR. In a further embodiment, a temperature gradient is used in place of a denaturing gradient (Rosenbaum and Reissner Biophys Chem, 265:12753, 1987).
  • Examples of other techniques for detecting point mutations include, but are not limited to, selective oligonucleotide hybridization, selective amplification, or selective primer extension. In one embodiment, oligonucleotide primers for specific amplification carry the mutation of interest in the center of the molecule (so that amplification depends on differential hybridization) or at the extreme 3′ end of one primer where, under appropriate conditions, mismatch can prevent or reduce polymerase extension. See e.g., Saiki et al., Proc. Natl. Acad. Sci. USA, 86:6230, 1989. In addition, it may be desirable to introduce a novel restriction site in the region of the mutation to create cleavage-based detection.
  • The methods described herein can be performed using prepackaged diagnostic kits which comprise at least one polynucleotide probe or one antibody of the present invention. These kits can be used in clinical settings to diagnose subjects exhibiting symptoms or family history of an MRCK1-related disease. Any cell type or tissue in which MRCK1 is expressed can be used for prognostic or diagnostic purposes.
  • Prophylactic Methods
  • This invention also provides methods for preventing diseases associated with aberrant MRCK1 expression or activity. The methods comprise administering to a target subject an agent which modulates MRCK1 expression or activity.
  • Subjects at risk of diseases which are caused by or attributed to aberrant MRCK1 expression or activity can be identified using the diagnostic or prognostic assays described herein. A prophylactic agent can be administered prior to the manifestation of MRCK1-related disease symptoms in order to prevent or delay MRCK1-related diseases. Suitable prophylactic agents include mutant MRCK1 proteins, MRCK1 antagonist agents, or MRCK1 antisense polynucleotides.
  • The prophylactic methods of this invention can be specifically tailored or modified, based on knowledge obtained from the study of pharmacogenomics. Pharmacogenomics includes the application of genomics technologies, such as gene sequencing, statistical genetics, and gene expression analysis, to drugs which are either in clinical development or on the market. Pharmacogenomics can be used to determine a subject's response to a drug (e.g., a subject's “drug response phenotype” or “drug response genotype”). Thus, another aspect of this invention is to provide methods for tailoring an individual's prophylactic or therapeutic treatment using MRCK1 modulators according to the individual's drug response genotype. Pharmacogenomics allows a clinician or physician to target prophylactic or therapeutic treatments to subjects who will most benefit from the treatment and to avoid treatment of subjects who will experience toxic drug-related side effects.
  • One pharmacogenomics approach to identify genes that predict drug response, known as “a genome-wide association,” relies primarily on a high-resolution map of the human genome consisting of already known gene-related sites (e.g., a “bi-allelic” gene marker map which consists of 60,000-100,000 polymorphic or variable sites on the human genome, each of which has two variants). Such a high-resolution genetic map can be compared to a map of the genome of each of a statistically substantial number of subjects taking part in a Phase II/III drug trial in order to identify genes associated with a particular observed drug response or side effect. Alternatively, such a high resolution map can be generated from a combination of some ten-million known single nucleotide polymorphisms (SNPs) in the human genome. A “SNP” is a common alteration that occurs in a single nucleotide base in a stretch of DNA. For example, a SNP may occur once per every 1000 bases of DNA. A SNP may be involved in a disease process. However, the vast majority of SNPs may be not related to diseases. Given a genetic map based on the occurrence of SNPs, individuals can be grouped into genetic categories depending on a particular pattern of SNPs in their individual genome. In such a manner, treatment regimens can be tailored to groups of genetically similar individuals, taking into account traits that may be common among such genetically similar individuals. Thus, mapping of the MRCK1 gene to SNP maps of patients with MRCK1-related diseases may facilitate the identification of drug-response-prediction genes.
  • Alternatively, the “candidate gene approach” can be utilized to identify genes that predict drug response. According to this method, if a gene that encodes a drug target is known, all common variants of that gene can be easily identified in the population. It then can be determined if a particular drug response is associated with one version of the gene versus another.
  • The activity of drug metabolizing enzymes is a major determinant of both the intensity and duration of drug action. The discovery of genetic polymorphisms of drug metabolizing enzymes (e.g., N-acetyltransferase 2 (NAT 2) and cytochrome P450 enzymes CYP2D6 and CYPZC19) has provided an explanation as to why some subjects do not obtain the expected drug effects or show exaggerated drug response and serious toxicity after taking the standard and safe dose of a drug. These polymorphisms are expressed in two phenotypes in the population, extensive metabolizer and poor metabolizer. The prevalence of poor metabolizer phenotypes is different among different populations. For example, the gene coding for CYP2D6 is highly polymorphic and several mutations have been identified in poor metabolizers, 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, poor metabolizers show no therapeutic response. 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.
  • In one embodiment, the “gene expression profiling” method can be utilized to identify genes that predict drug response. In this regard, the gene expression profile of an animal dosed with a drug can give an indication of whether the gene pathways related to toxicity have been turned on.
  • Information generated from the above pharmacogenomics approaches can be used to determine the appropriate dosage or treatment regimen suitable for a particular individual. This knowledge can avoid adverse reactions or therapeutic failure, and therefore enhance therapeutic or prophylactic efficiency when treating a subject with an MRCK1 modulator.
  • Therapeutic Methods
  • As described above, the present invention includes therapeutic methods for treating a subject at risk for, susceptible to, or diagnosed with MRCK1-related diseases. The therapeutic methods can be individually tailored based on the subject's drug response genotype. Typically, the therapeutic methods comprise modulating the expression or activity of MRCK1 in the subject. In one embodiment, the method comprises contacting a plurality of cells in the subject with an agent that inhibits the expression or activity of MRCK1. Suitable agents include polynucleotides (e.g., an antisense oligonucleotides of MRCK1), polypeptides (e.g., a dominant negative mutant of MRCK1), or polysaccharides, naturally-occurring target molecules of MRCK1 protein (e.g., an MRCK1 protein substrate or receptor), anti-MRCK1 antibodies, MRCK1 antagonists, or other small organic and inorganic molecule. They may also include vectors comprising polynucleotides encoding MRCK1 inhibitors or antisense sequences. Moreover, the agents can be anti-MRCK1 antibodies conjugated with therapeutic moieties. Suitable agents can be identified using the screening assays of the present invention.
  • Pharmaceutical Compositions
  • The present invention is further directed to pharmaceutical compositions comprising an MRCK1 modulator and a pharmaceutically acceptable carrier. As used herein, a “pharmaceutically acceptable carrier” is intended to include any and all solvents, solubilizers, fillers, stabilizers, binders, absorbents, bases, buffering agents, lubricants, controlled release vehicles, diluents, emulsifying agents, humectants, lubricants, dispersion media, coatings, antibacterial or antifungal agents, isotonic and absorption delaying agents, and the like, compatible with pharmaceutical administration. 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 agents can also be incorporated 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 (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 bisulfate; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose. 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 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 injectable composition should be sterile and should be fluid to the extent that easy syringability 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 absorption of the injectable compositions can be brought about by including in the composition an agent which delays absorption, for example, aluminum monostearate and gelatin.
  • Sterile injectable solutions can be prepared by incorporating the active modulator (e.g., an anti-MRCK1 antibody, an MRCK1 activity inhibitor, or a gene therapy vector expressing antisense nucleotide to MRCK1) in the required amount in an appropriate solvent, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the active compound into a sterile vehicle which contains a basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum drying and freeze-drying which 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 purpose of oral therapeutic administration, the active compound can be incorporated 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 tragacanth 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 Stertes; 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 transdermal means. For transmucosal or transdermal 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 transdermal administration, the bioactive 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 therapeutic moieties, which may contain a bioactive compound, 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 e.g. Alza Corporation 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. Pat. 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 includes 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.
  • Toxicity and therapeutic efficacy of such compounds can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD50/ED50. Compounds which exhibit large therapeutic indices are preferred. While compounds that exhibit toxic side effects may be used, care should be taken to design a delivery system that targets such compounds to the site of affected tissue in order to minimize potential damage to uninfected cells and, thereby, reduce side effects.
  • The data obtained from the cell culture assays and animal studies can be used in formulating a range of dosage for use in humans. The dosage of such compounds lies preferably within a range of circulating concentrations that include the ED50 with little or no toxicity. The dosage may vary within this range depending upon the dosage form employed and the route of administration utilized. For any compound used in the method of the invention, the therapeutically effective dose can be estimated initially from cell culture assays. A dose may be formulated in animal models to achieve a circulating plasma concentration range that includes the IC50 (i.e., the concentration of the test compound which achieves a half-maximal inhibition of symptoms) as determined in cell culture. Such information can be used to more accurately determine useful doses in humans. Levels in plasma may be measured, for example, by high performance liquid chromatography.
  • The pharmaceutical compositions can be included in a container, pack, or dispenser together with instructions for administration.
  • Kits
  • The invention also encompasses kits for detecting the presence of an MRCK1 gene product in a biological sample. An example kit comprises reagents for assessing expression of MRCK1 at mRNA or protein level. Preferably, the reagents include an antibody or fragment thereof, wherein the antibody or fragment specifically binds to MRCK1. Optionally, the kits may comprise a polynucleotide probe capable of specifically binding to a transcript of the MRCK1 gene. The kit may also contain means for determining the amount of MRCK1 protein or mRNA in the test sample, and/or means for comparing the amount of MRCK1 protein or mRNA in the test sample to a control or standard. The compound or agent can be packaged in a suitable container.
  • The invention further provides kits for assessing the suitability of each of a plurality of compounds for inhibiting MRCK1-related diseases in cells or human subjects. Such kits include a plurality of compounds to be tested, and a reagent (such as an antibody specific to MRCK1 proteins, or a polynucleotide probe or primer capable of hybridizing to the MRCK1 gene) for assessing expression of MRCK1.
  • It should be understood that the above-described embodiments are given by way illustration, not limitation. Various changes and modifications within the spirit and scope of the present invention will become apparent to those skilled in the art from the present description.
  • Host Cells
  • Another aspect of the invention pertains to host cells into which a polynucleotide molecule of the invention is introduced, e.g., an MRCK1 gene or homolog thereof, within an expression vector, a gene delivery vector, or a polynucleotide molecule of the invention containing sequences which allow it to homologously recombine into a specific site of the host cell's genome. 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 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, an MRCK1 gene can be expressed in bacterial cells such as E. coli, insect cells, yeast or mammalian cells (e.g., Chinese hamster ovary cells (CHO), COS cells, Fischer 344 rat cells, HLA-B27 rat cells, HeLa cells, A549 cells, or 293 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 polynucleotide (e.g., DNA) into a host cell, including calcium phosphate or calcium chloride co-precipitation, DEAE-dextran-mediated transfection, lipofection, or electoporation.
  • 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 flag (e.g., resistance to antibiotics) is generally introduced into the host cells along with the gene of interest. Preferred selectable flags include those which confer resistance to drugs, such as G418, hygromycin and methotrexate. Polynucleotides encoding a selectable flag can be introduced into a host cell by the same vector as that encoding MRCK1 or can be introduced by a separate vector. Cells stably transfected with the introduced polynucleotide can be identified by drug selection (e.g., cells that have incorporated the selectable flag 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) MRCK1. Accordingly, the invention further provides methods for producing MRCK1 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 containing an MRCK1 gene has been introduced) in a suitable medium such that MRCK1 is produced. In another embodiment, the method further comprises isolating MRCK1 from the medium or the host cell.
  • Transgenic and Knockout 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 MRCK1-coding sequences have been introduced. Such host cells can then be used to create non-human transgenic animals in which exogenous sequences encoding MRCK1 have been introduced into their genome or homologous recombinant animals in which endogenous sequences encoding MRCK1 have been altered. Such animals are useful for studying the function and/or activity of MRCK1 and for identifying and/or evaluating modulators of MRCK1 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, and the like. A transgene is exogenous DNA which is integrated into the genome of a cell from which a transgenic animal develops and which 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” or “knockout animal” is a non-human animal, preferably a mammal, more preferably a mouse, in which an endogenous MRCK1 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 an MRCK1-encoding polynucleotide into the mate pronuclei of a fertilized oocyte, e.g., by microinjection or retroviral infection, and allowing the oocyte to develop in a pseudopregnant female foster animal. Intronic sequences and polyadenylation signals can also be included in the transgene to increase the efficiency of expression of the transgene. A tissue-specific regulatory sequence(s) can be operably linked to a transgene to direct expression of MRCK1 to particular cells. Methods for generating transgenic animals via embryo manipulation and microinjection, particularly animals such as mice, have become conventional in the art. Similar methods are used for production of other transgenic animals. A transgenic founder animal can be identified based upon the presence of a transgene of the invention in its genome and/or expression of mRNA corresponding to a gene of the invention 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 MRCK1 can further be bred to other transgenic animals carrying other transgenes.
  • To create a homologous recombinant animal (knockout animal), a vector is prepared which contains at least a portion of a gene of the invention into which a deletion, addition or substitution has been introduced to thereby alter, e.g., functionally disrupt, the gene. The gene can be a human gene, but more preferably, is a non-human homolog of a human gene of the invention (e.g., a homolog of the MRCK1 gene). For example, a mouse gene can be used to construct a homologous recombination polynucleotide molecule, e.g., a vector, suitable for altering an endogenous gene of the invention in the mouse genome. In a preferred embodiment, the homologous recombination polynucleotide molecule is designed such that, upon homologous recombination, the endogenous gene of the invention is functionally disrupted (i.e., no longer encodes a functional protein; also referred to as a “knockout” vector). Alternatively, the homologous recombination polynucleotide molecule can be designed such that, upon homologous recombination, the endogenous 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 MRCK1 gene). In the homologous recombination polynucleotide molecule, the altered portion of the gene of the invention is flanked at its 5′ and 3′ ends by additional polynucleotide sequence of the gene of the invention to allow for homologous recombination to occur between the exogenous gene carried by the homologous recombination polynucleotide molecule and an endogenous gene in a cell, e.g., an embryonic stem cell. The additional flanking polynucleotide sequence is of sufficient length for successful homologous recombination with the endogenous gene.
  • Typically, several kilobases of flanking DNA (both at the 5′ and 3′ ends) are included in the homologous recombination polynucleotide molecule. The homologous recombination polynucleotide molecule is introduced into embryonic stem cells by electroporation. The cells in which the introduced gene has homologously recombined with the endogenous gene are selected. The selected cells can then be injected into a blastocyst of an animal (e.g., a mouse) to form aggregation chimeras. 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 homologously recombined DNA. Methods for constructing homologous recombination polynucleotide molecules, e.g., vectors, or homologous recombinant animals are well-known in the art.
  • In another embodiment, transgenic non-human animals can be produced which contain selected systems which allow for regulated expression of the transgene. One example of such a system is the cre/loxP recombinase system of bacteriophage P1. Another example of a recombinase system is the FLP recombinase system of Saccharomyces cerevisiae (See e.g., O'Gorman et al., Science, 251:1351-1355, 1991). 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 encoding 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., Nature, 385:810-813, 1997, and PCT International Publication Nos. WO97/07668 and WO97/07669. 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.
  • EXAMPLES Example 1 Identification of MRCK1 Sequence in Human Genome Database
  • The nucleic acid sequence of MRCK1 is obtained from a newly developed genomic prediction pipeline. Briefly, the X-ray crystal structures of the catalytic domains of protein kinases were collected and aligned together according to their structural identity/similarities. The alignment was converted into a “scoring matrix” which carried the structural profile of the kinase catalytic domains. This scoring matrix was then used to search the Celera Human Genome database for sequences that have kinase catalytic domains.
  • Example 2 BLAST Analysis
  • Sequence alignments between MRCK1 and other sequences in GenBank database were performed using the standard protein-protein BLAST(blastp), standard nucleotide-nucleotide BLAST(blastn), BLAST2 Sequences, and human genome BLAST programs that are available at NCBI's BLAST website.
  • A standard protein-protein BLAST search in the “nr” database (available at NCBI's BLAST website) with “Filter” setting unchecked, “Expect” setting at 10.0, “Word Size” setting at 3, “Matrix” setting at BLOSUM62, “Gap costs” setting at Existence: 11 and Extension: 1, revealed that MRCK1 shares sequence homologies with rat MRCK alpha (Entrez accession number: NP-446109.1, FIGS. 1 and 2), a putative mouse MRCK alpha (XP140553.1, 70% alignment to amino acid residues 40-1548 of MRCK1), and two putative human MRCK-related proteins (Entrez accession number Y12337, 100% alignment to amino acid residues 113-399 of MRCK1 and Entrez accession number U59305, 65% alignment to amino acid residues 40-434 of MRCK1).
  • A conserved domain search was performed within the standard protein-protein BLAST search with the RPS-BLAST 2.2.3 [Apr-24-2002] program. The amino acid residues 71-337 of MRCK1 are highly homologous to the consensus sequence of the catalytic domain of a family of ser/thr protein kinase (accession number smart00220, 100% alignment, FIG. 3), the pkinase domain (accession number pfam00069, 100% alignment, FIG. 4), and the catalytic domain of tyrosine kinase (accession number smart00219, 89.1% alignment, FIG. 5). The amino acid residues 339-398 of MRCK1 are highly homologous to the consensus sequence of an extension to ser/thr type protein kinase (accession number smart00133, 95.2% alignment, FIG. 6). The amino acid residues 339-398 of MRCK1 also aligns to the consensus sequence of the protein kinase C terminal domain (accession number pfam00433, 91.0% alignment, FIG. 7). The amino acid residues 882-920 of MRCK1 aligns to the consensus sequence of DAG_PE-binding domain (accession number pfam00130, 78.0% alignment, FIG. 8). The amino acid residues 953-1060 of MRCK1 aligns to the consensus sequence of the Pleckstrin homology (PH) domain (accession number smart00233, 87.5% alignment, FIG. 9). The amino acid residues 953-1060 of MRCK1 also aligns to another consensus sequence of the PH domain (accession number pfam00169, 88.0% alignment, FIG. 10). The amino acid residues 1102-1345 of MRCK1 aligns to the consensus sequence of the CNH domain (accession number pfam00780, 85.7% alignment, FIG. 11). The amino acid residues 1440-1471 of MRCK1 aligns to the consensus sequence of the P21-Rho-binding domain (accession number smart00285, 86.1% alignment, FIG. 12). The amino acid residues 648-786 of MRCK1 also weakly aligns to the consensus sequence of the myosin tail (accession number pfam01576, 17.8% alignment, FIG. 13).
  • A standard nucleotide-nucleotide BLAST search in the “geneseqn” database (available at NCBI's BLAST website) with “Matrix” setting at blastn matrix: 1-3, “Gap Penalties” setting at Existence: 5, Extension: 2, identified significant alignment of MRCK1 nucleotide sequence to the human kinase polypeptide PKIN-20 from PCT patent application WO02/08399 (Entrez accession number: AAD30567). Further analysis using Pairwise BLAST program (BLASTN setting: Match: 1, Mismatch: −2, gap open: 5, gap extension: 2, x_dropoff: 500, expect: 10.0, wordsize: 11, filter: unchecked; BALSTP setting: Matrix: BLOSUM62, gap open: 11, gap extension: 1, x_dropoff: 50, expect: 10.0, wordsize: 3, filter: unchecked) showed sequence homologies of 95% at nucleotide level and 90% at amino acid level between MRCK1 and PKIN-20. The amino acid and nucleotide sequences of PKIN-20 are recited in SEQ ID NOS:4 and 5. The standard nucleotide-nucleotide BLAST search in the “geneseqn” database also revealed significant sequence homologies between MRCK1 and a human Cdc42-binding kinase homologue-encoding cDNA (Entrez accession number: ABA08323, SEQ ID NO:6, 99% identities to nucleotide residues 2741-4398 of MRCK1).
  • A human genome search was carried out using blastn program with Expect setting at 0.01, Filter setting at default, Descriptions setting at 100, and Alignment settings at 100. The MRCK1 gene was mapped to or near loci 11p13 of human chromosome 11. Specifically, MRCK1 gene is located between genes LOC196204 and LOC143732, and overlaps with gene LOC196205. The exons/introns in the MRCK1 gene were determined using the program “sim4” described by Florea et al., in “A computer program for aligning a cDNA sequence with a genomic DNA sequence” Genome Res. 8:967-974, 1998.
  • Example 3 Hydrophobicity Analysis
  • The hydrophobicity profile of MRCK1 sequence (FIG. 14) was generated using the GES (Goldman, Engelman and Steitz) hydrophobicity scale (Engelman et al., Ann. Rev. Biophys. Biophys. Chem. 15:321-353, 1986). Briefly, the GES scale is used to identify nonpolar transbilayer helices. The curve is the average of a residue-specific hydrophobicity scale over a window of 20 residues. When the line is in the upper half of the frame (positive), it indicates a hydrophobic region and when it is in the lower half (negative), a hydrophilic region.
  • In FIG. 14, the X-axis represents the length of the protein in amino acids (aa), while the Y-axis represents the GES score. The curve line shows the GES pattern of the entire protein, while the straight line represents certain cutoff for potential membrane spanning domains. The hydrophobicity profile indicates that MRCK1 is probably not a membrane protein.
  • Example 4 MTN and MTA Analysis
  • The DNA probe for the MTN and MTA analysis is a PCR amplified 447 nucleotide fragment (SEQ ID NO:9) from human cDNA library. The probe located at exon 29-33, pos#3898-4344 of the MRCK1 cDNA. The PCR primers were designed based on the cDNA prediction. The Forward primer sequence is: 5′ GCTGGCATCTACGTGGATG 3′ (SEQ ID NO:7). The reverse primer sequence is: 5′ GTGGTTGAAGTTGGTAGGCG 3′ (SEQ ID NO:8). The PCR amplified probe was sequence verified. The positions of the probe and the primers relative to the MRCK1 gene are shown in FIG. 15.
  • The MTN analysis was performed using the Multiple Tissue Northern (MTN®) Blot Kit (Clontech Laboratories, Inc., Palo Alto, Calif.) under conditions specified in the User Manual. Briefly, the human MRCK1 probe was labeled with 32P. The MTN Blot was prehybridized in ExpressHyb solution at 68° C. for 30 min, and then hybridized with the labeled probe at 68° C. for 1 hr. The blot was washed two times with Wash Solution 1 for 30 min at room temperature and two times with Wash Solution 2 for 30 min at 50° C., and was then exposed to X-ray film. Two transcripts of MRCK1, a 4 kb and a 6 kb transcript, were detected in human brain, heart, skeletal muscle, colon, thymus, spleen, kidney, liver, small intestine, placenta, lung, and peripheral blood leukocyte. The highest expression was in placenta while the lowest expression was in small intestine. The MTA analysis was performed using the Multiple Tissue Expression (MTE®) Array Kit (Clontech Laboratories, Inc., Palo Alto, Calif.) under conditions specified in the User Manual. Briefly, the MTE array was hybridized to 32P-labeled human MRCK1 probe in ExpressHyb solution at 65° C. overnight with continuous agitation. The array was washed four times with Wash Solution 1 for 20 min at 65° C. and two times with Wash Solution 2 for 20 min at 55° C., and was then exposed to X-ray film. MRCK1 expression was found in all 76 tissues contained in the array.
  • Having described the preferred embodiments of compositions, organisms and methodologies employing a novel human gene MRCK1 (which are intended to be illustrative and not limiting), it is noted that modifications and variations can be made by persons skilled in the art in light of the above teachings. Therefore, it is understood that changes may be made in the particular embodiments disclosed which are within the scope and spirit of what is described as defined by the appended claims.

Claims (10)

1. An isolated polynucleotide comprising a nucleic acid sequence which encodes the amino acid sequence depicted in SEQ ID NO:2.
2. The polynucleotide according to claim 1, wherein the nucleic acid sequence is selected from the group consisting of
(a) the nucleic acid sequence as shown in SEQ ID NO:1;
(b) the complement of (a); and
(c) a nucleic acid sequence that differs from (a) or (b) due to the degeneracy of genetic code.
3. The polynucleotide according to claim 1, wherein the nucleic acid sequence is selected from the group consisting of:
(a) the nucleic acid sequence as shown in SEQ ID NO:3;
(b) the complement of (a); and
(c) a nucleic acid sequence that differs from (a) or (b) due to the degeneracy of genetic code.
4. (canceled)
5. An isolated polynucleotide comprising a nucleic acid sequence which has at least 96% sequence identity with the sequence depicted in SEQ ID NO:1, or the complement thereof.
6. The polynucleotide according to claim 5, wherein said nucleic acid sequence encodes a protein having MRCK activity.
7. (canceled)
8. An isolated polypeptide comprising a fragment of SEQ ID NO:2, wherein said fragment comprises at least 260 consecutive amino acid residues.
9. The isolated polypeptide according to claim 8, wherein said fragment comprises amino acid residues 70-337 of SEQ ID NO:2.
10-23. (canceled)
US11/982,836 2002-11-27 2007-11-05 Compositions, organisms and methodologies employing a novel human kinase Abandoned US20080153147A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/982,836 US20080153147A1 (en) 2002-11-27 2007-11-05 Compositions, organisms and methodologies employing a novel human kinase

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US42938102P 2002-11-27 2002-11-27
US10/702,496 US7297525B2 (en) 2002-11-27 2003-11-07 Composition employing a novel human kinase
US11/982,836 US20080153147A1 (en) 2002-11-27 2007-11-05 Compositions, organisms and methodologies employing a novel human kinase

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US10/702,496 Continuation US7297525B2 (en) 2002-11-27 2003-11-07 Composition employing a novel human kinase

Publications (1)

Publication Number Publication Date
US20080153147A1 true US20080153147A1 (en) 2008-06-26

Family

ID=32469312

Family Applications (2)

Application Number Title Priority Date Filing Date
US10/702,496 Expired - Fee Related US7297525B2 (en) 2002-11-27 2003-11-07 Composition employing a novel human kinase
US11/982,836 Abandoned US20080153147A1 (en) 2002-11-27 2007-11-05 Compositions, organisms and methodologies employing a novel human kinase

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US10/702,496 Expired - Fee Related US7297525B2 (en) 2002-11-27 2003-11-07 Composition employing a novel human kinase

Country Status (3)

Country Link
US (2) US7297525B2 (en)
AU (1) AU2003290664A1 (en)
WO (1) WO2004050831A2 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009051825A1 (en) * 2007-10-17 2009-04-23 University Of Miami Use of rac1 inhibitors to treat kaposi's sarcoma

Citations (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4489710A (en) * 1981-06-23 1984-12-25 Xoma Corporation Composition and method for transplantation therapy
US4522811A (en) * 1982-07-08 1985-06-11 Syntex (U.S.A.) Inc. Serial injection of muramyldipeptides and liposomes enhances the anti-infective activity of muramyldipeptides
US4569789A (en) * 1984-08-29 1986-02-11 Dana-Farber Cancer Institute, Inc. Acid-cleavable compound, use in protein conjugates and drug delivery systems
US4625014A (en) * 1984-07-10 1986-11-25 Dana-Farber Cancer Institute, Inc. Cell-delivery agent
US4638045A (en) * 1985-02-19 1987-01-20 Massachusetts Institute Of Technology Non-peptide polyamino acid bioerodible polymers
US4671958A (en) * 1982-03-09 1987-06-09 Cytogen Corporation Antibody conjugates for the delivery of compounds to target sites
US4751180A (en) * 1985-03-28 1988-06-14 Chiron Corporation Expression using fused genes providing for protein product
US4935233A (en) * 1985-12-02 1990-06-19 G. D. Searle And Company Covalently linked polypeptide cell modulators
US5459039A (en) * 1989-05-12 1995-10-17 Duke University Methods for mapping genetic mutations
US5498531A (en) * 1993-09-10 1996-03-12 President And Fellows Of Harvard College Intron-mediated recombinant techniques and reagents
US5919619A (en) * 1981-10-23 1999-07-06 Molecular Biosystems, Inc. Oligonucleotide therapeutic agent and methods of making same
US6506559B1 (en) * 1997-12-23 2003-01-14 Carnegie Institute Of Washington Genetic inhibition by double-stranded RNA

Family Cites Families (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4902505A (en) 1986-07-30 1990-02-20 Alkermes Chimeric peptides for neuropeptide delivery through the blood-brain barrier
GB9517780D0 (en) 1995-08-31 1995-11-01 Roslin Inst Edinburgh Biological manipulation
GB9517779D0 (en) 1995-08-31 1995-11-01 Roslin Inst Edinburgh Biological manipulation
JP2002508299A (en) 1997-09-19 2002-03-19 セクイター, インク. Sense mRNA therapy
FR2771422B1 (en) 1997-11-21 2001-07-27 Centre Nat Rech Scient REAGENTS AND METHODS FOR THE DETECTION OF GENES RELATED TO THE MAJOR COMPLEX OF HISTOCOMPATIBILITY OF BREEDING BIRDS, SUCH AS CHICKEN
DE19956568A1 (en) 1999-01-30 2000-08-17 Roland Kreutzer Method and medicament for inhibiting the expression of a given gene
JP2002542263A (en) 1999-04-21 2002-12-10 ワイス Methods and compositions for inhibiting the function of a polynucleotide sequence
JP2003501038A (en) 1999-05-28 2003-01-14 スージェン・インコーポレーテッド Protein kinase
JP2003516124A (en) 1999-10-15 2003-05-13 ユニバーシティー オブ マサチューセッツ RNA interference pathway genes as a means of targeted genetic interference
GB9927444D0 (en) 1999-11-19 2000-01-19 Cancer Res Campaign Tech Inhibiting gene expression
AU2001241961A1 (en) 2000-03-06 2001-09-17 Sugen, Inc. Novel human protein kinases and protein kinase-like enzymes
WO2001068836A2 (en) 2000-03-16 2001-09-20 Genetica, Inc. Methods and compositions for rna interference
MXPA02009069A (en) 2000-03-17 2004-04-05 Benitec Australia Ltd Genetic silencing.
DE60140676D1 (en) 2000-03-30 2010-01-14 Massachusetts Inst Technology RNA INTERFERENCE MEDIATORS WHICH ARE RNA SEQUENCE SPECIFIC
US6436703B1 (en) 2000-03-31 2002-08-20 Hyseq, Inc. Nucleic acids and polypeptides
WO2001092513A1 (en) 2000-05-30 2001-12-06 Johnson & Johnson Research Pty Limited METHODS FOR MEDIATING GENE SUPPRESION BY USING FACTORS THAT ENHANCE RNAi
CA2416414A1 (en) * 2000-07-21 2002-01-31 Henry Yue Human kinases
JP2004520813A (en) 2000-08-31 2004-07-15 インサイト・ゲノミックス・インコーポレイテッド Human kinase
CA2420862A1 (en) 2000-09-21 2002-03-28 Incyte Genomics, Inc. Protein phosphatases
EP1379628A2 (en) 2000-12-06 2004-01-14 Incyte Genomics, Inc. Kinases and phosphatases sequences, and use thereof
WO2002081517A2 (en) * 2001-01-19 2002-10-17 Curagen Corporation Novel polypeptides and nucleic acids encoded thereby
US6743619B1 (en) 2001-01-30 2004-06-01 Nuvelo Nucleic acids and polypeptides
US20040038881A1 (en) 2001-08-31 2004-02-26 Olga Bandman Human kinases
EP1485474A2 (en) 2001-12-07 2004-12-15 Incyte Genomics, Inc. Kinases and phosphatases

Patent Citations (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4489710A (en) * 1981-06-23 1984-12-25 Xoma Corporation Composition and method for transplantation therapy
US5919619A (en) * 1981-10-23 1999-07-06 Molecular Biosystems, Inc. Oligonucleotide therapeutic agent and methods of making same
US4671958A (en) * 1982-03-09 1987-06-09 Cytogen Corporation Antibody conjugates for the delivery of compounds to target sites
US4522811A (en) * 1982-07-08 1985-06-11 Syntex (U.S.A.) Inc. Serial injection of muramyldipeptides and liposomes enhances the anti-infective activity of muramyldipeptides
US4625014A (en) * 1984-07-10 1986-11-25 Dana-Farber Cancer Institute, Inc. Cell-delivery agent
US4569789A (en) * 1984-08-29 1986-02-11 Dana-Farber Cancer Institute, Inc. Acid-cleavable compound, use in protein conjugates and drug delivery systems
US4638045A (en) * 1985-02-19 1987-01-20 Massachusetts Institute Of Technology Non-peptide polyamino acid bioerodible polymers
US4751180A (en) * 1985-03-28 1988-06-14 Chiron Corporation Expression using fused genes providing for protein product
US4935233A (en) * 1985-12-02 1990-06-19 G. D. Searle And Company Covalently linked polypeptide cell modulators
US5459039A (en) * 1989-05-12 1995-10-17 Duke University Methods for mapping genetic mutations
US5498531A (en) * 1993-09-10 1996-03-12 President And Fellows Of Harvard College Intron-mediated recombinant techniques and reagents
US6506559B1 (en) * 1997-12-23 2003-01-14 Carnegie Institute Of Washington Genetic inhibition by double-stranded RNA

Also Published As

Publication number Publication date
AU2003290664A1 (en) 2004-06-23
WO2004050831A2 (en) 2004-06-17
AU2003290664A8 (en) 2004-06-23
US20040121383A1 (en) 2004-06-24
WO2004050831A3 (en) 2006-09-14
US7297525B2 (en) 2007-11-20

Similar Documents

Publication Publication Date Title
US7407792B2 (en) Compositions, organisms and methodologies employing a novel human kinase
Müller et al. Identification of B-KSR1, a novel brain-specific isoform of KSR1 that functions in neuronal signaling
US20050266409A1 (en) Compositions and methods for diagnosing, preventing, and treating cancers
US20070087988A1 (en) Hematopoietic progenitor kinase 1 for modulation of an immune response
CA2394803A1 (en) Novel human protein kinases and protein kinase-like enzymes
US20080178307A1 (en) Compositions, organisms and methodologies employing a novel human protein phosphatase
US6383791B1 (en) Molecules of the HKID-1-related protein family and uses thereof
US7297525B2 (en) Composition employing a novel human kinase
US20050059025A1 (en) Compositions, organisms and methodologies employing a novel human kinase
US20020115120A1 (en) Novel molecules of the HKID-1-related protein family and uses thereof
EP1294899B1 (en) 16836, a human phospholipase c family member and uses thereof
US20070178515A1 (en) 3714, 16742, 23546, and 13887 novel protein kinase molecules and uses therefor
US7211392B2 (en) 2150, human protein kinase family member and uses therefor
US6730491B2 (en) 2504, 15977, and 14760, novel protein kinase family members and uses therefor
AU2003287050A1 (en) Compositions, organisms and methodologies employing a novel human kinase
US7241585B2 (en) 14171 protein kinase, a novel human protein kinase and uses thereof
US7157240B2 (en) MID 4460, a human tyrosine phosphatase family member and uses therefor
US20050186667A1 (en) Pregnancy up-regulated, nonubiquitous CaM kinase
US7198929B2 (en) 14790, a novel protein kinase molecule and uses therefor
JP2003144168A (en) Cell cycle-relating protein having nuclear export function or nucleus-cytoplasm transport function

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION