US20050100913A1 - RNA interference pathway genes as tools for targeted genetic interference - Google Patents

RNA interference pathway genes as tools for targeted genetic interference Download PDF

Info

Publication number
US20050100913A1
US20050100913A1 US10/645,735 US64573503A US2005100913A1 US 20050100913 A1 US20050100913 A1 US 20050100913A1 US 64573503 A US64573503 A US 64573503A US 2005100913 A1 US2005100913 A1 US 2005100913A1
Authority
US
United States
Prior art keywords
rde
polypeptide
rnai
gene
protein
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
US10/645,735
Inventor
Craig Mello
Andrew Fire
Hiroaki Tabara
Alla Grishok
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.)
University of Massachusetts Medical School UMMS
Original Assignee
University of Massachusetts Medical School UMMS
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 University of Massachusetts Medical School UMMS filed Critical University of Massachusetts Medical School UMMS
Priority to US10/645,735 priority Critical patent/US20050100913A1/en
Publication of US20050100913A1 publication Critical patent/US20050100913A1/en
Priority to US11/710,152 priority patent/US7759463B2/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
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/67General methods for enhancing the expression
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/43504Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from invertebrates
    • C07K14/43536Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from invertebrates from worms
    • C07K14/4354Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from invertebrates from worms from nematodes
    • C07K14/43545Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from invertebrates from worms from nematodes from Caenorhabditis

Definitions

  • This invention relates to the discovery of genes whose expression products are involved in mediation of genetic interference.
  • RNA interference represents an efficient mechanism for inactivating this transfer process for a specific targeted gene. Targeting is mediated by the sequence of the RNA molecule introduced to the cell. Double-stranded (ds) RNA can induce sequence-specific inhibition of gene function (genetic interference) in several organisms including the nematode, C. elegans (Fire, et al., 1998 , Nature 391:806-811), plants, trypanosomes, Drosophila , and planaria (Waterhouse et al., 1998 , Proc. Natl. Acad. Sci. USA 94:13959-13964; Ngo et al., 1998 , Proc.
  • dsRNA can induce genetic interference in organisms from several distinct phyla suggests a conserved mechanism and perhaps a conserved physiological role for the interference process. Although several models of RNAi have been proposed (Baulcombe, 1999 , Curr. Biol. 9:R599-R601; Sharp, 1999 , Genes & Dev. 13:139-141) the mechanisms of action of specific components of the pathway are not known.
  • transgene silencing leads to problems with the commercial or therapeutic application of transgenic technology to alter the genetic makeup of a cell, organism, or human patient.
  • RNAi RNA interference pathway genes which are involved in mediating double-stranded RNA-dependent gene silencing (genetic interference).
  • RNAi requires a set of conserved cellular factors to suppress gene expression. These factors are the components of the RNAi pathway.
  • the RNAi pathway mutations and genes described herein e.g., rde-1, rde-2, rde-3, rde-4, rde-5, mut-2, and mut-7
  • RDE-1 and RDE-4 protein products
  • RNAi pathway RNAi pathway mediated by dsRNA
  • sequences and methods described herein are useful for modulating the RNAi pathway and may be used in conjunction with other methods involving the use of genetic inhibition by dsRNA (e.g., see U.S. Ser. No. 09/215,257, filed Dec. 18, 1998, incorporated herein by reference in its entirety).
  • RNAi pathway components provide activities necessary for interference. These activities may be absent or not sufficiently activated in many cell types, including those of organisms such as humans in which genetic interference may have potential therapeutic value. Components of the RNAi pathway in C. elegans may be sufficient when provided through transgenesis or as direct RNA:protein complexes to activate or directly mediate genetic interference in heterologous cells that are deficient in RNAi.
  • RNAi pathway components e.g., RDE-1, RDE-4
  • RDE-1, RDE-4 RNAi pathway components
  • Such sequences can also be used to generate knockout strains of animals such as C. elegans.
  • the nucleic acids of the invention include nucleic acids that hybridize, e.g., under stringent hybridization conditions (as defined herein), to all or a portion of the nucleotide sequence of SEQ ID NO:1 ( FIG. 5A -C) or its complement; SEQ ID NO:2 ( FIG. 6A -D) or its complement, or SEQ ID NO:4 or its complement.
  • the hybridizing portion of the hybridizing nucleic acids are preferably 20, 30, 50, or 70 bases long.
  • the hybridizing portion of the hybridizing nucleic acid is 80%, more preferably 95%, or even 98% or 100% identical to the sequence of a portion or all of a nucleic acid encoding an RDE-1 polypeptide or an RDE-4 polypeptide.
  • Hybridizing nucleic acids of the type described above can be used as a cloning probe, a primer (e.g., a PCR primer), or a diagnostic probe.
  • Preferred hybridizing nucleic acids encode a polypeptide having some or all of the biological activities possessed by a naturally-occurring RDE-1 polypeptide or an RDE-4 polypeptide e.g., as determined in the assays described below.
  • Hybridizing nucleic acids may encode a protein that is shorter or longer than the RDE-1 protein or RDE-4 protein described herein.
  • Hybridizing nucleic acids may also encode proteins that are related to RDE-1 or RDE-4 (e.g., proteins encoded by genes that include a portion having a relatively high degree of identity to the rde-1 gene or rde-4 gene described herein).
  • the invention also features purified or isolated RDE-1 polypeptides and RDE-4 polypeptides.
  • RDE-1 and RDE-4 polypeptides are useful for generating and testing antibodies that specifically bind to an RDE-1 or an RDE-4. Such antibodies can be used, e.g., for studying the RNAi pathway in C. elegans and other organisms.
  • protein and “polypeptide” mean any chain of amino acids, regardless of length or post-translational modification (e.g., glycosylation or phosphorylation).
  • RNAi pathway polypeptide includes a full-length, naturally occurring RNAi pathway polypeptide such as RDE-1 protein or RDE-4 protein, as well as recombinantly or synthetically produced polypeptides that correspond to a full-length, naturally occurring RDE-1 protein, RDE-4 protein, or to particular domains or portions of a naturally occurring RNAi pathway protein.
  • RNAi pathway mutations and strains harboring those mutations are useful for studying the RNAi pathway, including identification of modulators of the RNAi pathway.
  • RNAi pathway components e.g., those associated with mut-7 and rde-2
  • RNAi pathway components can be used to desilence or prevent silencing of transgenes.
  • RNAi pathway components are inhibited using specific inhibitors of an RNAi pathway gene or its product.
  • the invention includes an isolated nucleic acid molecule comprising a nucleotide sequence encoding an RDE-1 polypeptide.
  • the nucleic acid molecule hybridizes under high stringency conditions to the nucleic acid sequence of Genbank Accession No. AF180730 (SEQ ID NO:2) or its complement, or the sequence of SEQ ID NO:1 or its complement.
  • the isolated nucleic acid can complement an rde-1 mutation.
  • the invention also encompasses an isolated nucleic acid whose nucleotide sequence encodes the amino acid sequence of SEQ ID NO:3.
  • the invention also encompasses a substantially pure RDE-1 polypeptide encoded by the isolated nucleic acids described herein.
  • the invention features an antibody that specifically binds to an RDE-1 polypeptide.
  • the invention also includes a method of enhancing the expression of a transgene in a cell, the method comprising decreasing activity of the RNAi pathway.
  • rde-2 expression or activity is decreased.
  • the invention also features an isolated nucleic acid molecule comprising a nucleotide sequence encoding an RDE-4 polypeptide, wherein the nucleic acid molecule hybridizes under high stringency conditions to the nucleic acid sequence of SEQ ID NO:4 or its complement.
  • the invention also encompasses an isolated nucleic acid encoding an RDE-4 polypeptide, wherein the nucleic acid can complement an rde-4 mutation.
  • the invention also encompasses an isolated nucleic acid encoding an RDE-4 polypeptide, in which the nucleotide sequence encodes the amino acid sequence of SEQ ID NO:5.
  • the invention also features a substantially pure RDE-4 polypeptide encoded by the isolated nucleic acids described herein.
  • the invention features an antibody that specifically binds to an RDE-4 polypeptide.
  • the invention also features a method of preparing an RNAi agent, the method includes incubating a dsRNA in the presence of an RDE-1 protein and an RDE-4 protein.
  • the invention also features a method of inhibiting the activity of a gene by introducing an RNAi agent into a cell, such that the dsRNA component of the RNAi agent is targeted to the gene.
  • the cell contains an exogenous RNAi pathway sequence.
  • the exogenous RNAi pathway sequence can be an RDE-1 polypeptide or an RDE-4 polypeptide.
  • a dsRNA is introduced into a cell containing an exogenous RNAi pathway sequence such as nucleic acid sequence expressing an RDE-1 or RDE-4.
  • RNAi pathway component is a protein or nucleic acid that is involved in promoting dsRNA-mediated genetic interference.
  • a nucleic acid component can be an RNA or DNA molecule.
  • a mutation in a gene encoding an RNAi pathway component may decrease or increase RNAi pathway activity.
  • RNAi pathway protein is a protein that is involved in promoting dsRNA mediated genetic interference.
  • a “substantially pure DNA” is a DNA that is not immediately contiguous with (i.e., covalently linked to) both of the coding sequences with which it is immediately contiguous (i.e., one at the 5′ end and one at the 3′ end) in the naturally-occurring genome of the organism from which the DNA of the invention is derived.
  • the term therefore includes, for example, a recombinant DNA which is incorporated into a vector, into an autonomously replicating plasmid or virus, or into the genomic DNA of a prokaryote or eukaryote; or which exists as a separate molecule (e.g., a cDNA or a genomic or cDNA fragment produced by PCR (polymerase chain reaction) or restriction endonuclease digestion) independent of other sequences. It also includes a recombinant DNA which is part of a hybrid gene encoding additional polypeptide sequences.
  • inhibitory activity of a dsRNA decreases inhibitory activity of a dsRNA which results in at least two-fold less inhibition by a dsRNA relative to its ability to cause inhibition in a wild type cell
  • the pathway can be inhibited by inhibiting a component of the pathway (e.g., RDE-1) or mutating the component so that its function is reduced.
  • a “substantially pure polypeptide” is a polypeptide, e.g., an RNAi pathway polypeptide or fragment thereof, that is at least 60%, by weight, free from the proteins and naturally-occurring organic molecules with which it is naturally associated.
  • the preparation is at least 75%, more preferably at least 90%, and most preferably at least 99%, by weight, RNAi pathway polypeptide or fragment.
  • a substantially pure RNAi pathway polypeptide or fragment thereof is obtained, for example, by extraction from a natural source; by expression of a recombinant nucleic acid encoding an RNAi pathway polypeptide or fragment thereof; or by chemically synthesizing the polypeptide or fragment. Purity can be measured by any appropriate method, e.g., column chromatography, polyacrylamide gel electrophoresis, or HPLC analysis.
  • RNAi pathway polypeptide binds to a particular entity, e.g., an RNAi pathway polypeptide, but which does not substantially recognize or bind to other molecules in a sample, e.g., a biological sample, which includes the particular entity, e.g., RDE-1.
  • RNAi agent is a dsRNA molecule that has been treated with those components of the RNAi pathway that are required to confer RNAi activity on the dsRNA.
  • treatment of a dsRNA under conditions that include RDE-1 and RDE-4 results in an RNAi agent.
  • Injection of such an agent into an animal that is mutant for RDE-1 and RDE-4 will result in activation of the RNAi pathway with respect to a targeted gene.
  • the dsRNA used to trigger the formation of the RNAi agent is selected to be an RNA corresponding to all or a portion of the nucleotide sequence of the targeted gene.
  • FIG. 1A illustrates the genetic scheme used to identify rde mutants.
  • FIG. 1B is an illustration summarizing data from the genetic mapping of rde and mut mutations.
  • the vertical bars represent chromosomes; LGI, LGIII, and LGV. Reference genetic markers are indicated at the right of each chromosome and the relative genetic positions of the rde and mut alleles are indicated at the left.
  • FIG. 2A is a graphical representation of experiments investigating the sensitivity of rde and mut strains to RNAi by microinjection.
  • the RNA species indicated above each graph was injected at high concentration (pos-1: 7 mg/ml, par-2: 3 mg/ml, sqt-3: 7 mg/ml).
  • the strains receiving injection are indicated at the left and the horizontal bar graphs reflect the percent of progeny that exhibited genetic interference.
  • the Unc marker mutants used are also indicated.
  • the percent embryonic lethality of F1 progeny is plotted as shaded bars and the fraction of affected progeny is indicated at the right of each graph.
  • FIG. 2B is a graphical representation of experiments demonstrating that animals homozygous for rde and mut alleles are resistant to RNAi targeting maternally expressed genes, pos-1 and par-2.
  • the percent embryonic lethality of F1 progeny is plotted as shaded bars and the fraction of affected progeny is indicated at the right of each graph.
  • FIG. 3 is a schematic representation of homozygous rde-1 (ne219) and rde-4(ne299) mutant mothers receiving injections of dsRNA targeting the body muscle structural gene unc-22.
  • FIG. 4A is a schematic representation of the physical map of the rde-1 region.
  • C. elegans YAC and cosmid DNA clones that were positive for rescue are indicated by an asterisk.
  • a representation of the expanded interval showing a minimal, 25 kb, rescuing interval defined by the overlap between cosmids T10A5 and C 27 H 6 is shown beneath the YAC and cosmid map. Predicted genes within this sequenced interval are illustrated above and below the hatch marked line.
  • a single, rescuing, 4.5 kb PCR fragment containing the KO8H.107 predicted gene is shown enlarged. Exon and intron (box/line) boundaries are shown as well as the positions of rde-1 point mutation in the predicted coding sequences.
  • FIG. 4B is an illustration of the predicted sequence of RDE-1 and its alignment with four related proteins.
  • the sequences are RDE-1 ( C. elegans ; Genbank Accession No. AF180730), F48F7.1 ( C. elegans ; Genbank Accession No. Z69661), eIF2C (rabbit; Genbank Accession No. AF005355), ZWILLE ( Arabidopsis ; Genbank Accession No. AJ223508), and Sting ( Drosophila ; Genbank Accession No. AF145680).
  • Identities with RDE-1 are shaded in black, and identities among the homologs are shaded in gray.
  • FIGS. 5A-5C are an illustration of the genomic sequence from cosmid K08H10 (Genbank accession Z83113.1; SEQ ID NO:1) corresponding to the rde-1 gene from the first nucleotide of 5′ untranslated region to the polyadenylation site.
  • FIGS. 6A-6D are an illustration of the cDNA sequence of rde-1 (SEQ ID NO:2), including the first 20 nucleotides constituting the 5′ untranslated sequence (5′UTR) and the predicted amino acid sequence encoded by rde-1 (RDE-1; SEQ ID NO:3).
  • the nucleotide sequence is numbered starting with the first nucleotide of the translated region.
  • FIG. 7A is an illustration of the protocol for injection of a wild-type hermaphrodite with dsRNA.
  • FIG. 7B is an illustration of a genetic scheme demonstrating extragenic inheritance of RNAi.
  • the fraction shown represents the number of RNAi affected F2 hermaphrodites over the total number of cross progeny scored for each genotype class. Phenotypically uncoordinated (Unc).
  • FIGS. 8A-8B are illustrations of a genetic scheme to determine if the wild-type activities of rde-1, rde-2, rde-4, and mut-7 are sufficient in the injected animal for interference among the F1 self progeny
  • A illustrates crosses of heterozygous hermaphrodites
  • B illustrates crosses using homzygous F1 progeny from heterozygous mothers. The fraction shown represents the number of RNAi affected animals over the total number of cross progeny scored for each genotype class.
  • FIG. 9A depicts experiments of a the genetic scheme to determine if the wild-type activities of rde-1, rde-2, rde-4, and mut-7 are sufficient in the injected animal for interference among the F1 self progeny.
  • the fraction shown represents the number of RNAi affected animals over the total number of cross progeny scored for each genotype class.
  • FIG. 9B depicts experiments designed to determine the requirements for rde-1, rde-2, rde-4, and mut-7 in F2 ( FIG. 10A ) and F1 ( FIG. 10B ) interference.
  • the fraction shown represents the number of RNAi affected animals over the total number of cross progeny scored for each genotype class.
  • FIGS. 10A-10B are a depiction of the cDNA sequence of a wild type rde-4 nucleic acid sequence (SEQ ID NO:4) and the predicted RDE-4 amino acid sequence (SEQ ID NO:5) of C. elegans . “*” indicates ambiguous base assignment.
  • FIG. 11 is a depiction of regions of homology between the predicted RDE-4 amino acid sequence, X1RBPA (SEQ ID NO:6), HsPKR (SEQ ID NO:7), and a consensus sequence (SEQ ID NO:8).
  • a predicted secondary structure for RDE-4 is also shown illustrating predicted regions of ⁇ helix and ⁇ pleated sheet.
  • FIG. 12 illustrates a scheme for rescue of an rde-4.
  • RNAi pathway genes encode products involved in genetic interference and are useful for mediating or enhancing genetic interference. These genes encode mediators of double-stranded RNA-mediated interference. The mediators can be nucleic acid or protein. RNAi pathway genes are also useful for mediating specific processes, e.g., a gene that mediates dsRNA uptake by cells may be useful for transporting other RNAs into cells or for facilitating entry of agents such as drugs into cells.
  • the methods and examples described below illustrate the identification of RNAi pathway components, the uses of RNAi pathway components, mutants, genes and their products.
  • RNAi pathway genes were identified using screens for C. elegans strains mutant for RNAi (Examples 2 and 3). The mutations were further characterized for germline and somatic effects, effects on transposon mobilization, X chromosome loss and transgene silencing, and target tissue activity (Examples 4 and 5).
  • the rde-1 gene was identified using YACs (yeast artificial chromosomes) and cosmids to rescue rde-1 mutants. Based on the identified sequence, a cDNA sequence was identified in a C. elegans cDNA library and the complete cDNA sequence determined (Example 6).
  • RNAi Pathway Genes Homologous to rde-1, rde-2, rde-3, and rde-4
  • RNAi pathway genes from C. elegans (such as those described herein) and from other organisms (e.g. plant, mammalian, especially human) are useful for the elucidation of the biochemical pathways involved in genetic interference and for developing the uses of RNAi pathway genes described herein.
  • RNAi pathway genes including two-hybrid screens, complementation of C. elegans mutants by expression libraries of cloned heterologous (e.g., plant, mammalian, human) cDNAs, polymerase chain reactions (PCR) primed with degenerate oligonucleotides, low stringency hybridization screens of heterologous cDNA or genomic libraries with a C. elegans RNAi pathway gene, and database screens for sequences homologous to an RNAi pathway gene.
  • Hybridization is performed under stringent conditions.
  • a labeled fragment can be used to screen a genomic library derived from the organism of interest, again, using appropriately stringent conditions. Such stringent conditions are well known, and will vary predictably depending on the specific organisms from which the library and the labeled sequences are derived.
  • T m Nucleic acid duplex or hybrid stability is expressed as the melting temperature or T m , which is the temperature at which a probe dissociates from a target DNA. This melting temperature is used to define the required stringency conditions. If sequences are to be identified that are related and substantially identical to the probe, rather than identical, then it is useful to first establish the lowest temperature at which only homologous hybridization occurs with a particular SSC or SSPE concentration. Then assume that 1% mismatching results in 1° C. decrease in the T m and reduce the temperature of the final wash accordingly (for example, if sequences with ⁇ 95% identity with the probe are sought, decrease the final wash temperature by 5° C.). Note that this assumption is very approximate, and the actual change in T m can be between 0.5° and 1.5° C. per 1% mismatch.
  • high stringency conditions include hybridizing at 68° C. in 5 ⁇ SSC/5 ⁇ Denhardt solution/1.0% SDS, or in 0.5 M NaHPO 4 (pH 7.2)/1 mM EDTA/7% SDS, or in 50% formamide/0.25 M NaHPO 4 (pH 7.2)/0.25 M NaCl/1 mM EDTA/7% SDS; and washing in 0.2 ⁇ SSC/0.1% SDS at room temperature or at 42° C., or in 0.1 ⁇ SSC/0.1% SDS at 68° C., or in 40 mM NaHPO 4 (pH 7.2)/1 mM EDTA/5% SDS at 50° C., or in 40 mM NaHPO 4 (pH 7.2) 1 mM EDTA/1% SDS at 50° C.
  • Moderately stringent conditions include washing in 3 ⁇ SSC at 42° C. The parameters of salt concentration and temperature can be varied to achieve the desired level of identity between the probe and the target nucleic acid.
  • RNAi genes e.g., rde-1
  • complementation of mutants, described in the Examples can be performed using nucleic acid sequences from organisms other than C. elegans .
  • Methods of inhibiting expression of a target gene in a cell using dsRNA are known in the art and are exemplified in U.S. Ser. No. 09/215,257, filed Dec. 18, 1998, which is incorporated herein by reference in its entirety.
  • Another method of screening is to use an identified RNAi pathway gene sequence to screen a cDNA or genomic library using low stringency hybridizations. Such methods are known in the art.
  • PCR with degenerate oligonucleotides is another method of identifying homologs of RNAi pathway genes (e.g., human rde-1). Homologs of an RNAi pathway gene identified in other species are compared to identify specific regions with a high degree of homology (as in the sequence comparison shown in FIG. 4 ). These regions of high homology are selected for designing PCR primers that maximize possible base-pairing with heterologous genes. Construction of such primers involves the use of oligonucleotide mixtures that account for degeneracy in the genetic code, i.e., allow for the possible base changes in an RNAi pathway gene that does not affect the amino acid sequence of the RNAi pathway protein.
  • Such primers may be used to amplify and clone possible RNAi pathway gene fragments from DNA isolated from another organism (e.g., mouse or human). The latter are sequenced and those encoding protein fragments with high degrees of homology to fragments of the RNAi pathway protein are used as nucleic acid probes in subsequent screens of genomic DNA and cDNA libraries (e.g., mouse or human). Full-length genes and cDNAs having substantial homology to the previously identified RNAi pathway gene are identified in these screens.
  • RNAi pathway gene product e.g., RDE-1
  • a sequence encoding the gene is placed in an expression vector and the gene expressed in an appropriate cell type.
  • the gene product is isolated from such cell lines using methods known to those in the art, and used in the assays and procedures described herein.
  • the gene product can be a complete RNAi pathway protein (e.g., RDE-1) or a fragment of such a protein.
  • Full-length polypeptides and polypeptides corresponding to one or more domains of a full-length RNAi pathway protein are also within the scope of the invention.
  • fusion proteins in which a portion (e.g., one or more domains) of an RDE-1 or RDE-4) is fused to an unrelated protein or polypeptide (i.e., a fusion partner) to create a fusion protein.
  • the fusion partner can be a moiety selected to facilitate purification, detection, or solubilization, or to provide some other function.
  • Fusion proteins are generally produced by expressing a hybrid gene in which a nucleotide sequence encoding all or a portion of of an RNAi pathway protein is joined in-frame to a nucleotide sequence encoding the fusion partner.
  • Fusion partners include, but are not limited to, the constant region of an immunoglobulin (IgFc).
  • IgFc immunoglobulin
  • a fusion protein in which an RNAi pathway polypeptide is fused to IgFc can be more stable and have a longer half-life in the body than the polypeptide on its own.
  • RNAi pathway proteins (e.g., RDE-1, RDE-4) according to the invention can be produced by transformation (transfection, transduction, or infection) of a host cell with all or part of an RNAi pathway protein-encoding DNA fragment (e.g., one of the cDNAs described herein) in a suitable expression vehicle.
  • suitable expression vehicles include: plasmids, viral particles, and phage.
  • baculovirus expression vectors are suitable.
  • the entire expression vehicle, or a part thereof, can be integrated into the host cell genome.
  • an inducible expression vector e.g., the LACSWITCHTM Inducible Expression System (Stratagene; LaJolla, Calif.).
  • RNAi pathway protein can be produced in a prokaryotic host (e.g., E. coli or B. subtilis ) or in a eukaryotic host (e.g., Saccharomyces or Pichia ; mammalian cells, e.g., COS, NIH 3T3 CHO, BHK, 293, or HeLa cells; or insect cells).
  • a prokaryotic host e.g., E. coli or B. subtilis
  • a eukaryotic host e.g., Saccharomyces or Pichia
  • mammalian cells e.g., COS, NIH 3T3 CHO, BHK, 293, or HeLa cells
  • insect cells e.g., insect cells.
  • Proteins and polypeptides can also be produced in plant cells.
  • viral expression vectors e.g., cauliflower mosaic virus and tobacco mosaic virus
  • plasmid expression vectors e.g., Ti plasmid
  • Such cells are available from a wide range of sources (e.g., the American Type Culture Collection, Rockland, Md.; also, see, e.g., Ausubel et al., Current Protocols in Molecular Biology , John Wiley & Sons, New York, 1994).
  • the methods of transformation or transfection and the choice of expression vehicle will depend on the host system selected.
  • Transformation and transfection methods are described, e.g., in Ausubel et al., supra; expression vehicles may be chosen from those provided, e.g., in Cloning Vectors : A Laboratory Manual (P. H. Pouwels et al., 1985, Supp. 1987).
  • the host cells harboring the expression vehicle can be cultured in conventional nutrient media adapted as need for activation of a chosen gene, repression of a chosen gene, selection of transformants, or amplification of a chosen gene.
  • pMAMneo provides an RSV-LTR enhancer linked to a dexamethasone-inducible MMTV-LTR promotor, an SV40 origin of replication which allows replication in mammalian systems, a selectable neomycin gene, and SV40 splicing and polyadenylation sites.
  • DNA encoding an RNAi pathway protein would be inserted into the pMAMneo vector in an orientation designed to allow expression.
  • the recombinant RNAi pathway protein would be isolated as described herein.
  • Other preferable host cells that can be used in conjunction with the pMAMneo expression vehicle include COS cells and CHO cells (ATCC Accession Nos. CRL 1650 and CCL 61, respectively).
  • RNAi pathway polypeptides can be produced as fusion proteins.
  • the expression vector pUR278 (Ruther et al., EMBO J. 2:1791, 1983), can be used to create lacZ fusion proteins.
  • the pGEX vectors can be used to express foreign polypeptides as fusion proteins with glutathione S-transferase (GST). In general, such fusion proteins are soluble and can be easily purified from lysed cells by adsorption to glutathione-agarose beads followed by elution in the presence of free glutathione.
  • GST glutathione S-transferase
  • the pGEX vectors are designed to include thrombin or factor Xa protease cleavage sites so that the cloned target gene product can be released from the GST moiety.
  • RNAi pathway protein coding sequence can be cloned individually into non-essential regions (for example the polyhedrin gene) of the virus and placed under control of an AcNPV promoter, e.g., the polyhedrin promoter.
  • an AcNPV promoter e.g., the polyhedrin promoter.
  • Successful insertion of a gene encoding an RNAi pathway polypeptide or protein will result in inactivation of the polyhedrin gene and production of non-occluded recombinant virus (i.e., virus lacking the proteinaceous coat encoded by the polyhedrin gene).
  • RNAi pathway protein nucleic acid sequence can be ligated to an adenovirus transcription/translation control complex, e.g., the late promoter and tripartite leader sequence.
  • This chimeric gene can then be inserted into the adenovirus genome by in vitro or in vivo recombination. Insertion into a non-essential region of the viral genome (e.g., region E1 or E3) will result in a recombinant virus that is viable and capable of expressing an RNAi pathway gene product in infected hosts (see, e.g., Logan, Proc. Natl. Acad. Sci. USA 81:3655, 1984).
  • Specific initiation signals may be required for efficient translation of inserted nucleic acid sequences. These signals include the ATG initiation codon and adjacent sequences. In cases where an entire native RNAi pathway protein gene or cDNA, including its own initiation codon and adjacent sequences, is inserted into the appropriate expression vector, no additional translational control signals may be needed. In other cases, exogenous translational control signals, including, perhaps, the ATG initiation codon, must be provided. Furthermore, the initiation codon must be in phase with the reading frame of the desired coding sequence to ensure translation of the entire insert. These exogenous translational control signals and initiation codons can be of a variety of origins, both natural and synthetic. The efficiency of expression may be enhanced by the inclusion of appropriate transcription enhancer elements, transcription terminators (Bittner et al., Methods in Enzymol. 153:516, 1987).
  • RNAi pathway polypeptides can be expressed directly or as a fusion with a heterologous polypeptide, such as a signal sequence or other polypeptide having a specific cleavage site at the N-and/or C-terminus of the mature protein or polypeptide. Included within the scope of this invention are RNAi pathway polypeptides with a heterologous signal sequence.
  • the heterologous signal sequence selected should be one that is recognized and processed, i.e., cleaved by a signal peptidase, by the host cell.
  • a prokaryotic signal sequence is selected, for example, from the group of the alkaline phosphatase, penicillinase, lpp, or heat-stable enterotoxin II leaders.
  • yeast secretion a yeast invertase, alpha factor, or acid phosphatase leaders may be selected.
  • a host cell may be chosen which modulates the expression of the inserted sequences, or modifies and processes the gene product in a specific, desired fashion. Such modifications (e.g., glycosylation) and processing (e.g., cleavage) of protein products may be important for the function of the protein. Different host cells have characteristic and specific mechanisms for the post-translational processing and modification of proteins and gene products. Appropriate cell lines or host systems can be chosen to ensure the correct modification and processing of the foreign protein expressed. To this end, eukaryotic host cells that possess the cellular machinery for proper processing of the primary transcript, glycosylation, and phosphorylation of the gene product can be used. Such mammalian host cells include, but are not limited to, CHO, VERO, BHK, HeLa, COS, MDCK, 293, 3T3, W138, and in particular, choroid plexus cell lines.
  • an RNAi pathway protein can be produced by a stably-transfected mammalian cell line.
  • a number of vectors suitable for stable transfection of mammalian cells are available to the public, see e.g., Pouwels et al. (supra); methods for constructing such cell lines are also publicly available, e.g., in Ausubel et al. (supra).
  • cDNA encoding an RNAi pathway protein e.g., RDE-1 or RDE-4
  • DHFR dihydrofolate reductase
  • RNAi pathway protein-encoding gene into the host cell chromosome is selected for by including 0.01-300 ⁇ M methotrexate in the cell culture medium (as described in Ausubel et al., supra). This dominant selection can be accomplished in most cell types.
  • Recombinant protein expression can be increased by DHFR-mediated amplification of the transfected gene.
  • Methods for selecting cell lines bearing gene amplifications are described in Ausubel et al. (supra); such methods generally involve extended culture in medium containing gradually increasing levels of methotrexate.
  • DHFR-containing expression vectors commonly used for this purpose include pCVSEII-DHFR and pAdD26SV(A) (described in Ausubel et al., supra).
  • Any of the host cells described above or, preferably, a DHFR-deficient CHO cell line e.g., CHO DHFR ⁇ cells, ATCC Accession No. CRL 9096 are among the host cells preferred for DHFR selection of a stably-transfected cell line or DHFR-mediated gene amplification.
  • a number of other selection systems can be used, including but not limited to the herpes simplex virus thymidine kinase, hypoxanthine-guanine phosphoribosyl-transferase, and adenine phosphoribosyltransferase genes can be employed in tk, hgprt, or aprt cells, respectively.
  • gpt which confers resistance to mycophenolic acid (Mulligan et al., Proc. Natl. Acad. Sci. USA, 78:2072, 1981); neo, which confers resistance to the aminoglycoside G-418 (Colberre-Garapin et al., J. Mol. Biol., 150:1, 1981); and hygro, which confers resistance to hygromycin (Santerre et al., Gene, 30:147, 1981), can be used.
  • any fusion protein can be readily purified by utilizing an antibody specific for the fusion protein being expressed.
  • a system described in Janknecht et al., Proc. Natl. Acad. Sci. USA, 88:8972 (1981) allows for the ready purification of non-denatured fusion proteins expressed in human cell lines.
  • the gene of interest is subcloned into a vaccinia recombination plasmid such that the gene's open reading frame is translationally fused to an amino-terminal tag consisting of six histidine residues. Extracts from cells infected with recombinant vaccinia virus are loaded onto Ni 2+ nitriloacetic acid-agarose columns, and histidine-tagged proteins are selectively eluted with imidazole-containing buffers.
  • RNAi pathway protein or a portion thereof can be fused to an immunoglobulin Fc domain.
  • a fusion protein can be readily purified using a protein A column.
  • the invention also includes humanized or chimeric antibodies, single chain antibodies, Fab fragments, F(ab′) 2 fragments, and molecules produced using a Fab expression library.
  • Antibodies can be raised against a short peptide epitope of an RNAi pathway gene (e.g., rde-1), an epitope linked to a known immunogen to enhance immunogenicity, a long fragment of an RNAi pathway gene, or the intact protein.
  • RNAi pathway gene e.g., rde-1
  • Such antibodies are useful for e.g., localizing RNAi pathway polypeptides in tissue sections or fractionated cell preparations, determining whether an RNAi pathway gene is expressed (e.g., after transfection with an RNAi pathway gene), and evaluating the expression of an RNAi pathway gene in disorders (e.g., genetic conditions) where the RNAi pathway may be affected.
  • RNAi pathway protein e.g., RDE-1
  • RDE-1 an isolated RNAi pathway protein
  • the RNAi pathway immunogen can also be a mutant RNAi pathway protein or a fragment of a mutant RNAi pathway protein.
  • a full-length RNAi pathway protein can be used or, alternatively, antigenic peptide fragments of RNAi pathway protein can be used as immunogens.
  • the antigenic peptide of an RNAi pathway protein comprises at least 8 (preferably 10, 15, 20, or 30) amino acid residues.
  • RDE-1 these residues are drawn from the amino acid sequence shown in SEQ ID NO:3 and encompass an epitope such that an antibody raised against the peptide forms a specific immune complex with RDE-1.
  • Preferred epitopes encompassed by the antigenic peptide are regions of the protein that are located on the surface of the protein, e.g., hydrophilic regions.
  • RNAi pathway protein immunogen typically is used to prepare antibodies by immunizing a suitable subject (e.g., rabbit, goat, mouse or other mammal) with the immunogen.
  • An appropriate immunogenic preparation can contain, for example, recombinantly expressed RNAi pathway protein or a chemically synthesized RNAi polypeptide.
  • the preparation can further include an adjuvant, such as Freund's complete or incomplete adjuvant, or similar immunostimulatory agent. Immunization of a suitable subject with an immunogenic RNAi pathway protein preparation induces a polyclonal anti-RNAi pathway protein antibody response.
  • RNAi pathway antibodies Polyclonal antibodies that recognize an RNAi pathway protein
  • RNAi pathway antibodies can be prepared as described above by immunizing a suitable subject with an RNAi pathway protein immunogen.
  • the RNAi pathway antibody titer in the immunized subject can be monitored over time by standard techniques, such as with an enzyme-linked immunosorbent assay (ELISA) using immobilized RNAi pathway protein from which the immunogen was derived.
  • ELISA enzyme-linked immunosorbent assay
  • the antibody molecules directed against the RNAi pathway protein can be isolated from the mammal (e.g., from the blood) and further purified by well-known techniques, such as protein A chromatography to obtain the IgG fraction.
  • antibody-producing cells can be obtained from the subject and used to prepare monoclonal antibodies by standard techniques, such as the hybridoma technique originally described by Kohler and Milstein (1975) Nature 256:495-497, the human B cell hybridoma technique (Kozbor et al. (1983) Immunol. Today 4:72), the EBV-hybridoma technique (Cole et al. (1985), Monoclonal Antibodies and Cancer Therapy , Alan R. Liss, Inc., pp. 77-96) or trioma techniques.
  • standard techniques such as the hybridoma technique originally described by Kohler and Milstein (1975) Nature 256:495-497, the human B cell hybridoma technique (Kozbor et al. (1983) Immunol. Today 4:72), the EBV-hybridoma technique (Cole et al. (1985), Monoclonal Antibodies and Cancer Therapy , Alan R. Liss, Inc., pp. 77-96) or trioma techniques
  • hybridomas The technology for producing hybridomas is well known (see generally Current Protocols in Immunology (1994) Coligan et al. (eds.) John Wiley & Sons, Inc., New York, N.Y.). Briefly, an immortal cell line (typically a myeloma) is fused to lymphocytes (typically splenocytes) from a mammal immunized with an RNAi pathway immunogen as described above, and the culture supernatants of the resulting hybridoma cells are screened to identify a hybridoma producing a monoclonal antibody that binds to the RNAi pathway protein.
  • lymphocytes typically splenocytes
  • RNAi pathway protein Any of the many well known protocols used for fusing lymphocytes and immortalized cell lines can be applied for the purpose of generating a monoclonal antibody against an RNAi pathway protein (see, e.g., Current Protocols in Immunology , supra; Galfre et al., 1977, Nature 266:55052; R. H. Kenneth, in Monoclonal Antibodies: A New Dimension In Biological Analyses , Plenum Publishing Corp., New York, N.Y., 1980; and Lerner, 1981 , Yale J. Biol. Med., 54:387-402. Moreover, one in the art will appreciate that there are many variations of such methods which also would be useful. Hybridoma cells producing a monoclonal antibody of the invention are detected by screening the hybridoma culture supernatants for antibodies that bind to the RNAi pathway protein, e.g., using a standard ELISA assay.
  • RNAi pathway antibody can be identified and isolated by screening a recombinant combinatorial immunoglobulin library (e.g., an antibody phage display library) with an RNAi pathway protein to thereby isolate immunoglobulin library members that bind to the RNAi pathway protein.
  • 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). Additionally, examples of methods and reagents particularly amenable for use in generating and screening antibody display library can be found in, for example, U.S.
  • chimeric antibodies can be used to splice the genes from a mouse antibody molecule of appropriate antigen specificity together with genes from a human antibody molecule of appropriate biological activity.
  • a chimeric antibody is a molecule in which different portions are derived from different animal species, such as those having a variable region derived from a murine mAb and a human immunoglobulin constant region.
  • single chain antibodies can be adapted to produce single chain antibodies against an RNAi pathway protein or polypeptide.
  • Single chain antibodies are formed by linking the heavy and light chain fragments of the Fv region via an amino acid bridge, resulting in a single chain polypeptide.
  • Antibody fragments that recognize and bind to specific epitopes can be generated by known techniques.
  • such fragments can include but are not limited to F(ab′) 2 fragments, which can be produced by pepsin digestion of the antibody molecule, and Fab fragments, which can be generated by reducing the disulfide bridges of F(ab′) 2 fragments.
  • Fab expression libraries can be constructed (Huse et al., Science, 246:1275, 1989) to allow rapid and easy identification of monoclonal Fab fragments with the desired specificity.
  • RNAi pathway components can be identified in C. elegans and other animals (e.g., a mammal) using the methods described in the Examples below. Pathway components can also be identified using methods known in the art and the information provided herein. Such components include those involved in protein:protein and protein:RNA interactions. Specifically, RDE-1 can be used to identify additional proteins and RNA molecules that bind to the RDE-1 protein and so facilitate genetic interference.
  • RNAi pathway mutant strains described herein can be used in genetic screens to identify additional RNAi pathway components.
  • a strain deficient for rde-1 activity can be mutagenized and screened for the recovery of genetic interference. This type of screen can identify allele-specific suppressors in other genes or second site mutations within the rde-1 gene that restore its activity.
  • the resulting strains may define new genes that activate RNAi to overcome or bypass the rde-1 defect.
  • the mutations identified by these methods can be used to identify their corresponding gene sequences.
  • Two-hybrid screens can also be used to identify proteins that bind to RNAi pathway proteins such as RDE-1.
  • Genes encoding proteins that interact with RDE-1 or human homologs of the C. elegans RDE-1 are identified using the two-hybrid method (Fields and Song, 1989 , Nature 340:245-246; Chien et al., 1991 , Proc. Natl. Acad. Sci. USA 88:9578-9582; Fields and Stemglanz, 1994 , Trends Genet. 10:286-292; Bartel and Fields, 1995 , Methods Enzymol. 254:241-263).
  • DNA encoding the RDE-1 protein is cloned and expressed from plasmids harboring GAL4 or lexA DNA-binding domains and co-transformed into cells harboring lacZ and HIS3 reporter constructs along with libraries of cDNAs that have been cloned into plasmids harboring the GAL4 activation domain.
  • Libraries used for such co-transformation include those made from C. elegans or a vertebrate embryonic cell.
  • RNAi pathway Specific cellular functions associated with the RNAi pathway include the specific targeting of a nucleic acid by a dsRNA, uptake of dsRNA, transport of dsRNA, amplification of the dsRNA signal, and genetic interference.
  • the mechanism of interference may involve translation inhibition, or interference with RNA processing.
  • direct effects on the corresponding gene may contribute to interference.
  • the following assays are designed to identify compounds that are effective inhibitors of the RNAi pathway.
  • Such inhibitors may act by, but are not limited to, binding to an RDE-1 polypeptide (e.g., from C. elegans , mouse, or human), binding to intracellular proteins that bind to an RNAi pathway component, compounds that interfere with the interaction between RNAi pathway components including between an RNAI pathway component and a dsRNA, and compounds that modulate the activity or expression of an RNAi pathway gene such as rde-1.
  • An inhibitor of the RNAi pathway can also be used to promote expression of a transgene.
  • RNAi pathway gene regulatory sequences e.g., promoter sequences
  • Assays can also be used to identify molecules that bind to RNAi pathway gene regulatory sequences (e.g., promoter sequences), thus modulating gene expression. See, e.g., Platt, 1994 , J. Biol. Chem. 269:28558-28562, incorporated herein by reference in its entirety.
  • the compounds which may be screened by the methods described herein include, but are not limited to, peptides and other organic compounds (e.g., peptidomimetics) that bind to an RNAi pathway protein (e.g., that bind to an RDE-1), or inhibit its activity in any way.
  • peptides and other organic compounds e.g., peptidomimetics
  • Such compounds may include, but are not limited to, peptides; for example, soluble peptides, including but not limited to members of random peptide libraries; (see, e.g., Lam et al., 1991 , Nature 354:82-94; Houghten et al., 1991 , Nature 354:84-86), and combinatorial chemistry-derived molecular libraries made of D-and/or L-amino acids, phosphopeptides (including, but not limited to, members of random or partially degenerate, directed phosphopeptide libraries; see e.g., Songyang et al., 1993 , Cell 72:767-778), and small organic or inorganic molecules.
  • peptides for example, soluble peptides, including but not limited to members of random peptide libraries; (see, e.g., Lam et al., 1991 , Nature 354:82-94; Houghten et al., 1991 , Nature 354:84-86),
  • Organic molecules are screened to identify candidate molecules that affect expression of an RNAi pathway gene (e.g., rde-1), e.g., by interacting with the regulatory region or transcription factors of a gene.
  • RNAi pathway gene e.g., rde-1
  • Compounds are also screened to identify those that affect the activity of such proteins, (e.g., by inhibiting rde-1 activity) or the activity of a molecule involved in the regulation of, for example, rde-1.
  • RNAi pathway protein Computer modeling or searching technologies are used to identify compounds, or identify modifications of compounds that modulate the expression or activity of an RNAi pathway protein. For example, compounds likely to interact with the active site of a protein (e.g., RDE-1) are identified.
  • the active site of an RNAi pathway protein can be identified using methods known in the art including, for example, analysis of the amino acid sequence of a molecule, from a study of complexes of an RNAi pathway, with its native ligand (e.g., a dsRNA). Chemical or X-ray crystallographic methods can be used to identify the active site of an RNAi pathway protein by the location of a bound ligand such as a dsRNA.
  • the three-dimensional structure of the active site is determined. This can be done using known methods, including X-ray crystallography which may be used to determine a complete molecular structure. Solid or liquid phase NMR can be used to determine certain intra-molecular distances. Other methods of structural analysis can be used to determine partial or complete geometrical structures. Geometric structure can be determined with an RNAi pathway protein bound to a natural or artificial ligand which may provide a more accurate active site structure determination.
  • Computer-based numerical modeling can also be used to predict protein structure (especially of the active site), or be used to complete an incomplete or insufficiently accurate structure.
  • Modeling methods that may be used are, for example, parameterized models specific to particular biopolymers such as proteins or nucleic acids, molecular dynamics models based on computing molecular motions, statistical mechanics models based on thermal ensembles, or combined models.
  • standard molecular force fields representing the forces between constituent atoms and groups are necessary, and can be selected for the model from among the force fields known in physical chemistry.
  • Information on incomplete or less accurate structures determined as above can be incorporated as constraints on the structures computed by these modeling methods.
  • candidate modulating compounds can be identified by searching databases containing compounds along with information on their molecular structure.
  • the compounds identified in such a search are those that have structures that match the active site structure, fit into the active site, or interact with groups defining the active site.
  • the compounds identified by the search are potential RNAi pathway modulating compounds.
  • RNAi pathway protein e.g., an RDE-1
  • Systematic variations in composition can be evaluated to obtain modified modulating compounds or ligands of preferred specificity or activity.
  • Examples of molecular modeling systems are the QUANTA programs, e.g., CHARMm, MCSS/HOOK, and X-LIGAND, (Molecular Simulations, Inc., San Diego, Calif.).
  • QUANTA analyzes the construction, graphic modeling, and analysis of molecular structure.
  • CHARMm analyzes energy minimization and molecular dynamics functions.
  • MCSS/HOOK characterizes the ability of an active site to bind a ligand using energetics calculated via CHARMm.
  • X-LIGAND fits ligand molecules to electron density of protein-ligand complexes. It also allows interactive construction, modification, visualization, and analysis of the behavior of molecules with each other.
  • libraries of known compounds including natural products, synthetic chemicals, and biologically active materials including peptides, can be screened for compounds that are inhibitors or activators of the RNAi pathway components identified herein.
  • RNAi pathway gene products e.g., an RDE-1
  • Assays for testing the effectiveness of compounds such as those described herein are further described below.
  • RNAi pathway proteins e.g., bind to proteins or genes encoding those proteins (e.g., rde-1 and its protein product).
  • Such compounds are useful, for example, for modulating the activity of these entities, elaborating their biochemistry, treating disorders in which a decrease or increase in dsRNA mediated genetic interference is desired.
  • Such compounds may also be useful to treat diseases in animals, especially humans, involving nematodes, e.g., trichinosis, trichuriasis, and toxocariasis.
  • Compounds such as those described herein may also be useful to treat plant diseases caused by nematodes. These compounds can be used in screens for compounds that disrupt normal function, or may themselves disrupt normal function.
  • Assays to identify compounds that bind to RNAi pathway proteins involve preparation of a reaction mixture of the protein and the test compound under conditions sufficient to allow the two components to interact and bind thus forming a complex which can be removed and/or detected.
  • RNAi pathway protein from an organism e.g., RDE-1
  • peptide, or fusion protein can be immobilized onto a solid phase, reacted with the test compound, and complexes detected by direct or indirect labeling of the test compound.
  • the test compound can be immobilized, reacted with the RNAi pathway molecule, and the complexes detected.
  • Microtiter plates may be used as the solid phase and the immobilized component anchored by covalent or noncovalent interactions. Non-covalent attachment may be achieved by coating the solid phase with a solution containing the molecule and drying.
  • an antibody for example, one specific for an RNAi pathway protein such as RDE-1 is used to anchor the molecule to the solid surface. Such surfaces may be prepared in advance of use, and stored.
  • the non-immobilized component is added to the coated surface containing the immobilized component under conditions sufficient to permit interaction between the two components.
  • the unreacted components are then removed (e.g., by washing) under conditions such that any complexes formed will remain immobilized on the solid phase.
  • the detection of the complexes may be accomplished by a number of methods known to those in the art.
  • the nonimmobilized component of the assay may be prelabeled with a radioactive or enzymatic entity and detected using appropriate means. If the non-immobilized entity was not prelabeled, an indirect method is used. For example, if the non-immobilized entity is an RDE-1, an antibody against the RDE-1 is used to detect the bound molecule, and a secondary, labeled antibody used to detect the entire complex.
  • a reaction can be conducted in a liquid phase, the reaction products separated from unreacted components, and complexes detected (e.g., using an immobilized antibody specific for an RNAi pathway protein).
  • Cell-based assays can be used to identify compounds that interact with RNAi pathway proteins.
  • Cell lines that naturally express such proteins or have been genetically engineered to express such proteins e.g., by transfection or transduction of an rde-1 DNA
  • test compounds can be administered to cell cultures and the amount of mRNA derived from an RNAi pathway gene analyzed, e.g., by Northern analysis. An increase in the amount of RNA transcribed from such a gene compared to control cultures that did not contain the test compound indicates that the test compound is an inhibitor of the RNAi pathway.
  • the amount of a polypeptide encoded by an RNAi pathway gene, or the activity of such a polypeptide can be analyzed in the presence and absence of a test compound. An increase in the amount or activity of the polypeptide indicates that the test compound is an inhibitor of the RNAi pathway.
  • Ectopic expression i.e., expression of an RNAi pathway gene in a cell where it is not normally expressed or at a time when it is not normally expressed
  • a mutant RNAi pathway gene i.e., an RNAi pathway gene that suppresses genetic interference
  • Ectopic expression can be used to block or reduce endogenous interference in a host organism. This is useful, e.g., for enhancing transgene expression in those cases where the RNAi pathway is interfering with expression of a transgene.
  • Another method of accomplishing this is to knockout or down regulate an RNAi pathway gene using methods known in the art. These methods are useful in both plants and animals (e.g., in an invertebrate such as a nematode, a mouse, or a human).
  • Ectopic expression of an RNAi pathway gene can also be used to activate the RNAi pathway.
  • targeting can be used to activate the pathway in specific cell types, e.g., tumor cells.
  • a non-viral RNAi pathway gene construct can be targeted in vivo to specific tissues or organs, e.g., the liver or muscle, in patients. Examples of delivery systems for targeting such constructs include receptor mediated endocytosis, liposome encapsulation (described below), or direct insertion of non-viral expression vectors.
  • liposome encapsulation of nucleic acid is liposome encapsulation of nucleic acid.
  • Successful in vivo gene transfer has been achieved with the injection of DNA, e.g., as a linear construct or a circular plasmid, encapsulated in liposomes (Ledley, Human Gene Therapy 6:1129-1144 (1995) and Farhood, et al., Ann. NY Acad. Sci. 716:23-35 (1994)).
  • a number of cationic liposome amphiphiles are being developed (Ledley, Human Gene Therapy 6:1129-1144 (1995); Farhood, et al., Ann. NY Acad. Sci., 716:23-35 (1994) that can be used for this purpose.
  • Targeted gene transfer has been shown to occur using such methods.
  • intratracheal administration of cationic lipid-DNA complexes was shown to effect gene transfer and expression in the epithelial cells lining the bronchus (Brigham, et al., Am. J. Respir. Cell Mol. Biol. 8:209-213 (1993); and Canonico, et al., Am. J. Respir. Cell Mol. Biol. 10:24-29 (1994)).
  • Expression in pulmonary tissues and the endothelium was reported after intravenous injection of the complexes (Brigham, et al., Am. J. Respir. Cell Mol. Biol.
  • RNAi pathway sequence in linear, plasmid or viral DNA forms can be condensed through ionic interactions with the cationic lipid to form a particulate complex for in vivo delivery (Stewart, et al., Human Gene Therapy 3:267-275 (1992)).
  • liposome formulations for example, proteoliposomes which contain viral envelope receptor proteins, i.e., virosomes, have been found to effectively deliver genes into hepatocytes and kidney cells after direct injection (Nicolau, et al., Proc. Natl. Acad. Sci. USA 80:1068-1072 (1993); Kaneda, et al., Science 243:375-378 (1989); Mannino, et al., Biotechniques 6:682 (1988); and Tomita, et al., Biochem. Biophys. Res. Comm. 186:129-134 (1992)).
  • Direct injection can also be used to administer an RNAi pathway nucleic acid sequence in a DNA expression vectors, e.g., into the muscle or liver, either as a solution or as a calcium phosphate precipitate (Wolff, et al., Science 247:1465-1468 (1990); Ascadi, et al., The New Biologist 3:71-81 (1991); and Benvenisty, et al., Proc. Natl. Acad. Sci. USA 83:9551-9555 (1986).
  • a DNA expression vectors e.g., into the muscle or liver, either as a solution or as a calcium phosphate precipitate (Wolff, et al., Science 247:1465-1468 (1990); Ascadi, et al., The New Biologist 3:71-81 (1991); and Benvenisty, et al., Proc. Natl. Acad. Sci. USA 83:9551-9555 (1986).
  • RNAi pathway components can be used to prepare RNAi agents.
  • agents are dsRNAs that have been treated with RNAi pathway components rendering the treated dsRNA capable of activity in the RNAi pathway and can be used as sequence-specific interfering agents useful for targeted genetic interference.
  • treating a dsRNA with an RDE-1 and RDE-4 is useful for making an RNAi agent.
  • An RNAi agent can be produced by preincubating a dsRNA in vitro in the presence of RDE-1 and RDE-4.
  • Another method of preparing an RNAi agent is to activate the RNAi pathway in a target cell (i.e., a cell in which it is desirable to activate the RNAi pathway such as a tumor cell) by transgenesis of an rde-1 coding sequence and an rde-4 coding sequence into the target cell.
  • a target cell i.e., a cell in which it is desirable to activate the RNAi pathway such as a tumor cell
  • RNAi pathway polypeptides can be modified, e.g., to enhance their stability or cellular uptake, by attaching lipophilic or other helper groups to the polypeptide, by the formation of chimeras with proteins or other moieties that are taken up by cells, or by the use of liposomes or other techniques of drug delivery known in the art.
  • RNAi agents appear to spread from cell to cell, thus, active RNAi agents can diffuse or be actively transported from conditioned media or serum directly into target cells.
  • RNAi agents can be injected into an organism or cell. They may also be incorporated into a cell using liposomes or other such methods known in the art.
  • Such methods are useful for stimulating the RNAi pathway in C. elegans cells, and in heterologous cells including plants and vertebrate cells. Such methods are useful in mammalian, e.g., human cells.
  • RNAi pathway components that mediate the transport of dsRNA into cells and tissues can be used to promote the entry of dsRNA into cells and tissues, including dsRNA that is linked to another compound.
  • the method is accomplished by linking dsRNA to a cargo compound (e.g., a drug or DNA molecule), e.g., by a covalent bond.
  • a cargo compound e.g., a drug or DNA molecule
  • the endogenous RNAi pathway gene expressing dsRNA transport function is activated using methods known in the art. Alternatively, other methods can be used such as transfecting the target cell with the gene that affects transport thus permitting the cell or tissue to take up the dsDNA.
  • the invention is further described in the Examples below which describe methods of identifying mutations in the RNAi pathway and methods of identifying genes encoding components of the RNAi pathway.
  • the Bristol strain N2 was used as standard wild-type strain.
  • the marker mutations and deficiencies used are listed by chromosomes as follows: LGI: dpy-14(e188), unc-13(e51); LGIII: dpy-17(e164), unc-32(e189); LGV: dpy-11(e224), unc-42(e270), daf-11(m87), eDf1, mDf3, nDf31, sDf29, sDf35, unc-76(e911).
  • the C. elegans strain DP13 was used to generate hybrids for STS linkage-mapping (Williams et al., 1992 , Genetics 131:609-624).
  • RNAi Sensitivity to RNAi was tested in the following strains. MT3126: mut-2(r459) (obtained from John Collins, Department of Biochemistry & Molecular Biology, University of New Hampshire, Durham, N. H.); dpy-19(n1347), TW410: mut-2([459) sem-4(n1378), NL917: mut-7(pk204), SS552: mes-2(bn76) rol-1(e91)/mnC1 (obtained from S. Strome, Biology Dept., Indiana University), SS449: mes-3(bn88) dpy-5(e61) (from S.
  • mut-6, mes-2, 3, 4, 6 and him-1 showed sensitivity to RNAi by injection of pos-1 dsRNA.
  • the dose of injected RNA was about 0.7 mg/ml. This dose lies within the range where reduced concentration leads to reduced interference effects.
  • the results of the injection of pos-1 dsRNA into these mutants were as follows: mut-6: 422/437, mes-2: 781/787, mes-3: 462/474, mes-4: 810/814, mes-6: 900/1,002, him-1: 241/248, N2 (control): 365/393.
  • TR1175 unc-22(r765::Tc4).
  • Strains TW410 and TR1175 were gifts from Q. Boese and J. Collins (Department of Biochemistry & Molecular Biology, University of New Hampshire, Durham, N.H.).
  • RNAi administered by microinjection was performed as described in Fire et al., 1998, supra and Rocheleau et al., 1997 , Cell 90:707-716.
  • pos-1 cDNA clone yk61h1, par-2 cDNA clone yk96h7, sqt-3 cDNA clone yk75f2 were used to prepare dsRNA in vitro.
  • These cDNA clones were obtained from the C. elegans cDNA project (Y. Kohara, Gene Network Lab, National Institute of Genetics, Mishima 411, Japan).
  • RNAi administered by feeding was performed as described in Timmons and Fire, 1998 , Nature 395:854.
  • pos-1 cDNA was cloned into a plasmid that contains two T7 promoter sequences arranged in head-to-head configuration.
  • the plasmid was transformed into an E. coli strain, BL21 (DE3), and the transformed bacteria were seeded on NGM (nematode growth medium) plates containing 60 ⁇ g/ml ampicillin and 80 ⁇ g/ml IPTG. The bacteria were grown overnight at room temperature to induce pos-1 dsRNA. Seeded plates (BL21(DE3)[dsRNA] plates) stored at 4° C. remained effective for inducing interference for up to two weeks. To test RNAi sensitivity, C. elegans larvae were transferred onto BL21 (DE3)[dsRNA] plates and embryonic lethality was assayed in the next generation.
  • Transgenic lines expressing interfering RNA for unc-22 were engineered using a mixture of three plasmids: pPD[L4218] (unc-22 antisense segment, driven by myo-3 promoter); pPD[L4218] (corresponding unc-22 sense segment, driven by myo-3 promoter); pRF4 (semidominant transformation marker). DNA concentrations in the injected mixture were 100 ⁇ g/ml each. Injections were as described (Mello et al., 1991, EMBO J. 10:3959; Mello and Fire, 1995 , Methods in Cell Biol. 48:451-482).
  • RNAi pathway A method of screening for mutants defective in the RNAi pathway was devised that would permit the large-scale application of dsRNA to mutagenized populations.
  • the feeding method was optimized to deliver interfering RNA for an essential gene, pos-1.
  • C. elegans hermaphrodites that ingest bacteria expressing dsRNA corresponding to a segment of pos-1 are themselves unaffected but produce dead embryos with the distinctive pos-1 embryonic lethal phenotype.
  • RNAi pathway mutants The genetic screen used to isolate RNAi pathway mutants was similar to one designed by James R. Preiss for the identification of maternal effect mutants (Kemphues et al., 1988 , Cell 52:311-320).
  • An Eg 1 strain, lin-2(e]309) was mutagenized with EMS and the F2 generation was cultured on a bacterial lawn expressing pos-1 dsRNA. Mutagenized populations were then screened for rare individuals that were able to produce complete broods of viable progeny forming a distinctive “bag of worms” phenotype. To make sure that the animals were truly resistant to RNAi, candidate strains were next assayed for resistance to RNAi by injection. Independent EMS induced alleles of rde-1 were found in two separate pools of mutagenized animals at a frequency of approximately one allele in 2,000 to 4,000 haploid genomes.
  • Seven mutant strains were selected for genetic mapping. These seven mutants defined four complementation groups; rde-1, with three alleles, rde-4, with two alleles, and rde-2, and rde-3, with one allele each ( FIG. 1B ).
  • RNAi resistant phenotype was assayed either by feeding bacteria expressing pos-1 dsRNA or by injection of a dsRNA mixture of pos-1 and unc-22. The same assays were used for complementation tests. In vivo expression of unc-22 dsRNA was also used for mapping of rde-1. Mapping with visible marker mutations was performed as described in Brenner (1974, Genetics, 77:71-94) and mapping with STS marker was performed as described in Williams et al. (1992, supra).
  • ne219, ne297 and ne300 failed to complement each other, defining the rde-1 locus.
  • rde-1 mutations mapped near unc-42 V.
  • Three factor mapping was used to locate rde-1 (ne300) one eighth of the distance from unc-42 in the unc-42/daf-11 interval (3/24 Unc-non-Daf recombinants analyzed).
  • the rde-1 (ne300) allele complemented the chromosomal deficiency sDf29 and failed to complement eDf1, mDJ3, nDf31 and sDf35.
  • rde-2(ne221) and rde-3(ne298) mapped near unc-13 I.
  • rde-4(ne299) and (ne301) mapped near unc-69 III and failed to complement each other.
  • ne299 complemented mut-7(pk
  • the rde-1 (+) activity is sufficient maternally or zygotically.
  • animals heterozygous for rde-1 (ne219) were injected with dsRNA targeting the zygotic gene, sqt-3, and self progeny were assayed for the Sqt phenotype. 100% of the self progeny including rde-1 homozygous progeny were found to exhibit the Sqt phenotype.
  • rde-1(+) activity is sufficient to mediate interference with a zygotic target gene.
  • Zygotic sufficiency was assayed by injecting homozygous rde-1 mothers with dsRNA targeting the zygotic unc-22 gene ( FIG. 3 ).
  • Injected animals were allowed to produce self-progeny or instead were mated after 12 hours to wild-type males, to produce heterozygous rde/+cross-progeny. Each class of progeny was scored for the unc-22 twitching phenotype as indicated by the fraction shown if FIG. 3 (Unc progeny/total progeny). The injected animals were then mated with wild-type males. Self progeny from homozygous injected mothers were unaffected, however, 68% of the cross progeny were Unc. This result indicates that zygotically provided rde-1(+) activity is also sufficient.
  • rde-1(+) and rde-4(+) activities are not needed for dsRNA uptake, transport or stability.
  • RNAi sensitivity of several existing C. elegans mutants was also examined. Most of these mutant strains were fully sensitive to RNAi. However, RNAi resistance was identified in two strains that had previously been shown to exhibit elevated levels of transposon mobilization (mutator strains): mut-2 (described in Collins et al., 1987, Nature 328:726-728) and mut-7 (described in Ketting et al., Cell, in press for release on Oct. 15, 1999). Another mutator strain, mut-6(st702), was fully sensitive to RNAi. Since mutator strains continually accumulate mutations, the resistance of mut-2 and mut-7 may have been due to the presence of secondary mutations.
  • This method can be used to identify additional mutations in RNAi pathway genes.
  • Microinjection was used to assay the sensitivity of each rde strain to several distinct dsRNA species.
  • the pos-1 and par-2 genes are expressed in the maternal germline and are required for proper embryonic development (Tabara et al., 1999 , Development 126: 1-11; Boyd et al., 1996 , Development 122:3075-3084). All rde-strains tested (as well as mut-2 and mut-7) showed significant resistance to dsRNA targeting of these germline-specific genes ( FIG. 2B ), as well as to several other germline specific genes tested.
  • the rde-3 data (asterisk in FIG. 2B ) includes a 10% non-specific embryonic lethality present in the rde-3 strain.
  • rde-1, rde-3, rde-4 and mut-2 strains showed strong resistance to both sqt-3 and unc-22 dsRNA, while rde-2 and mut-7 strains showed partial resistance.
  • rde-2 and mut-7 appeared to be partially tissue- or gene-specific in that they were required for effective RNAi against germline but not somatically expressed genes.
  • the rde-1, rde-3, rde-4, and mut-2 (+) activities appeared to be required for interference for all genes analyzed.
  • the rde and mut strains differ from one another in sensitivity to sqt-2 dsRNA.
  • Mutator strains (including mut-2, mut-7) rde-2 and rde-3) exhibit a second phenotype: a high incidence of males reflecting an increased frequency of X-chromosome loss during meiosis (Collins et al., 1987, supra; Ketting et al., supra). This phenotype was observed in rde-2 and rde-3 strains, but not observed in the rde-1 and rde-4 strains which showed a wild-type incidence of males (Table 1).
  • homozygous mut-7 lines carrying various GFP reporters transgenes were generated as follows: N2 (Bristol strain) males were mated to mut-7 (pk204) unc-32 (e]89) hermaphrodites; cross progeny males were then mated to strains carrying the GFP transgenes. mut-7 unc-32/++ cross progeny from these matings were cloned, and mut-7 unc-32 homozygous animals carrying the transgenes were isolated from their self-progeny.
  • GFP reporter transgenes After the GFP reporter transgenes were introduced into different genetic backgrounds, activation of GFP transgene expression in germ cells was assayed at 25 ⁇ C by fluorescence microscopy. The tested GFP reporter transgenes were each active in some or all somatic tissues, but had become silenced in the germline.
  • the plasmids used and transgene designations are as follows: 1) pBK48 which contains an in-frame insertion of GFP into a ubiquitously expressed gene, let-858 (Kelly, et al., 1997 , Genetics 146:227-238).
  • ccExPD7271 contains more than 100 copies of pBK48 in a high copy repetitive array that is carried extrachromosomally.
  • pJH3.92 is an in-frame fusion of GFP with the maternal pie-1 gene (M. Dunn and G. Seydoux, Johns Hopkins University, Baltimore, Md.).
  • jhEx1070 carries pJH3.92 in a low copy “complex” extrachromosomal array generated by the procedure of Kelly et al. (1997, supra)
  • pJKL380.4 is a fusion of GFP with the C. elegans nuclear laminin gene, lam-1, which is expressed in all tissues (J. Liu and A. Fire).
  • ccIn4810 carries pJKL380.4 in a complex array that has been integrated into the X chromosome by gamma irradiation using standard techniques.
  • mut-7 and rde-2 which differ from rde-1 in having transposon mobilization and a high incidence of X-chromosome loss also differ from rde-1 in their ability to partially reactivate silent germline transgenes.
  • the rde-1 and rde-4 mutants differ from other RNAi deficient strains identified herein in that they do not cause transposon mobilization nor do they cause chromosome loss.
  • the role of these genes in upstream events such as dsRNA uptake, transport or stability was examined. Such events could be required for interference induced by exogenous trigger RNAs but might be dispensable for natural functions of RNAi.
  • rde-1 and rde-4 homozygotes were exposed to dsRNA. The next generation was scored for interference.
  • dsRNA targeting the unc-22 gene was injected into the intestinal cells of homozygous rde-1 and rde-4 hermaphrodites and the injected animals were then mated to wild-type males ( FIG. 3 ).
  • the self-progeny for both strains exhibited no interference with the targeted gene.
  • rde-1 and rde-4 mutants have intact mechanisms for transporting the interference effect from the site of injection (the intestine) into the embryos of the injected animal and then into the tissues of the resulting progeny.
  • the stability of the resulting interference also appeared to be normal in rde-1 and rde-4 as the homozygous injected mothers continued to produce affected cross progeny for several days after the time of injection.
  • the muscle-specific promoter from the myo-3 gene was used to drive the expression of both strands of the muscle structural gene unc-22 in the body wall muscles (Moerman et al., 1986, supra; Fire et al., 1991 , Development 113:503-514).
  • the rde-1 gene was cloned using standard genetic mapping to define a physical genetic interval likely to contain the gene using YACs and cosmids that rescue rde-1 mutants. These were used to identify a cloned rde-1 cDNA sequence and a cloned rde-4 sequence. These methods can also be used to identify the genes for rde-2, rde-3, and rde-5 using the mutant strains provided herein.
  • yeast artificial chromosome clones containing C. elegans DNA from this interval were used to rescue the rde-1 mutant phenotype.
  • candidate rescuing YACs were co-injected with plasmids designed to express unc-22(RNAi).
  • YAC and cosmid clones that mapped near the rde-1 locus were obtained from A. Coulson. rde-1 (ne219) was rescued by YAC clones: Y97C12 and Y50B5.
  • the two overlapping YAC clones provided rde-1 rescuing activity as indicated by unc-22 genetic interference with characteristic body paralysis and twitching in the F 1 and F2 transgenic animals.
  • a non-overlapping YAC clone failed to rescue resulting in 100% non-twitching transgenic strains ( FIG. 4A ).
  • the rescuing activity was further localized to two overlapping cosmid clones, cosmid C27H6 and T10A5, and finally to a single 4.5 kb genomic PCR fragment predicted to contain a single gene, designated K08H10.7 (SEQ ID NO:1; FIGS. 5A-5C )
  • K08H10.7 SEQ ID NO:1; FIGS. 5A-5C
  • the K08H10.7 PCR product gave strong rescue when amplified from wild-type genomic DNA. This rescue was greatly diminished using a PCR fragment amplified from any of the three rde-1 alleles and was abolished by a 4 bp insertion at a unique NheI site in the rde-1 coding region.
  • a wild-type PCR product from an adjacent gene C 27 H 6.4 also failed to rescue.
  • the K08H10.7 gene from each of the rde-1 mutant strains was sequenced, and distinct point mutations were identified that are predicted to alter coding sequences in K08H10.7 ( FIG. 4A ). Based on these findings rde-1 can be identified as the K08H10.7 gene.
  • a full-length cDNA sequence was determined for rde-1 using the cDNA clones, yk296b10 and yk595h5.
  • cDNA clones for rde-1 were obtained from Y. Kohara (Gene Network Lab, National Institute of Genetics, Mishima 411, Japan).
  • the cDNA sequence of coding region and 3'UTR was determined on yk296b10 except that the sequence of 5′UTR was determined on yk595h5.
  • the GenBank accession number for rde-1 cDNA is AF180730 (SEQ ID NO:2).
  • the rde-1 cDNA sequence was used to generate a predicted translation product (SEQ ID NO:3), referred to as RDE-1, consisting of 1020 amino acids.
  • the RDE-1 sequence was used to query Genbank and identify numerous related genes in C. elegans as well as other animals and plants.
  • This gene family includes at least 23 predicted C. elegans genes, several of which appear to be members of conserved subfamilies. Within subfamilies, conservation extends throughout the protein and all family members have a carboxy-terminal region that is highly conserved ( FIG. 4B ).
  • other related genes include ARGONAUTE 1 ( Arabidopsis ), SPCC736.11 ( S. pombe ), and Piwi ( Drosophila ). A portion of the N terminal region of RDE-1 showed no significant similarity to any of the identified related genes.
  • RDE-1 may provide sequence-specific inhibition of translation initiation in response to dsRNA.
  • the rde-1 mutations appear likely to reduce or eliminate rde-1(+) activity.
  • Two rde-1 alleles ne219 and ne297 are predicted to cause amino acid substitutions within the RDE-1 protein and were identified at a frequency similar to that expected for simple loss-of-function mutations.
  • the rde-1(ne219) lesion alters a conserved glutamate to a lysine ( FIG. 4B ).
  • the rde-1(ne297) lesion changes a non-conserved glycine, located four residues from the end of the protein, to a glutamate ( FIG. 4B ).
  • ne300 contains the strongest molecular lesion and is predicted to cause a premature stop codon prior to the most highly conserved region within the protein (Q>Ochre in FIG. 4B ). Consistent with the idea that rde-1 (ne300) is a strong loss of function mutation, we found that when placed in trans to a chromosomal deficiency the resulting deficiency trans-heterozyotes were RNAi deficient but showed no additional phenotypes. These observations suggest that rde-1 alleles are simple loss-of-function mutations affecting a gene required for RNAi but that is otherwise non-essential.
  • the RDE-1 protein and fragments thereof can be used to prepare dsRNA that is useful as an RNAi agent.
  • RNAi RNAi effect
  • linkage to the target gene e.g., was involved in a reversible alteration of the gene or associated chromatin
  • a strain was constructed such that the F1 males that carry the RNAi effect also bear a chromosomal deletion that removes the target gene ( FIG. 7B ).
  • the RNAi effect would be transmitted as a dominant factor.
  • dsRNA pos-1 dsRNA, mom-2 dsRNA, or sgg-1 dsRNA
  • the dsRNA was delivered by injection through a needle inserted into the intestine.
  • dsRNA was synthesized in vitro using T3 and T7 polymerases. Template DNA was removed from the RNA samples by DNase treatment (30 minutes at 37° C.). Equal amounts of sense and antisense RNAs were then mixed and annealed to obtain dsRNA.
  • dsRNA at a concentration of 1-5 mg/ml was injected into the intestine of animals.
  • FIG. 7A illustrates this experiment.
  • the gonad of the parent (P0) hermaphrodite has symmetrical anterior and posterior U-shaped arms as shown in FIG. 7A .
  • Several fertilized eggs are shown in FIG. 7A , centrally located in the uterus.
  • the rectangular mature oocytes are cued up in the gonad arms most proximal to the uterus.
  • the embryos present in P0 at the time of injection gave rise to unaffected F 1 progeny.
  • Oocytes in the proximal arms of the injected P0 gonad inherit the RNAi effect but also carry a functional maternal mRNA (F 1 carriers of RNAi).
  • the injected P0 After a clearance period during which carrier and unaffected F1 progeny are produced, the injected P0 begins to exclusively produce dead F1 embryos with the phenotype corresponding to the inactivation of the gene targeted by the injected RNA (Tabara et al. 1999, Development 126:1; C. Rocheleau, 1997, Cell 90:707). Potential F1 and F2 carriers of the interference effect were identified within the brood of the injected animal. In the case of hermaphrodites, carriers were defined as “affected” if the animals produced at least 20% dead embryos with phenotypes corresponding to maternal loss of function for the targeted locus.
  • carriers were defined as animals whose cross progeny included at least one affected F2 hermaphrodite.
  • the total number of carriers identified in each generation for each of three dsRNAs injected is shown in FIG. 7A as a fraction of the total number of animals assayed.
  • RNAi pos-1
  • chromosome carrying the deficiency for pos-1 also carried a deficiency for phenotypically uncoordinated (unc).
  • F2 progeny of the carrier male includes two genotypes: phenotypically wild-type animals that inherit the (+) chromosome, and phenotypically uncoordinated (Unc) progeny that inherit the mDf3 chromosome.
  • the deficiency bearing sperm were just as capable as wild-type sperm of transferring interference to the F2 hermaphrodite progeny ( FIG. 7B ).
  • the target locus was not needed for inheritance of the interference effect.
  • RNAi body muscle structural gene unc-22
  • cuticle collagen gene sqt-3 maternal genes pos-1 and sgg-1.
  • Males of the pes-10::gfp strain (Seydoux, G. and Dunn, Mass., 1997, Development 124:2191-2201 were injected with gfp dsRNA. Injected males were affected by unc-22 and gfp dsRNA to the same extent as injected hermaphrodites.
  • RNAi interference was detected in F1 progeny or injected males (40 to 200 F1 animals scored for each RNA tested. Therefore, the initial transmission of RNAi to F1 progeny may involve a mechanism active only in hermaphrodites while subsequent transmission to the F2 progeny appears to involve a distinct mechanism, active in both hermaphrodites and males.
  • the hermaphrodite-specific step may indicate the existence of a maternal germline process that amplifies the RNAi agent.
  • the germline factors that amplify the RNAi agents can be identified by mutations that result in an RNAi deficient mutant phenotype. Such factors can be used as additional components of an in vitro system for the efficient amplification of RNAi agents.
  • the heterozygous hermaphrodites from each genotype class rde-1, unc-42/+; rde-2, unc-13/+; mut-7, dpy-71/+; and rde-4, unc-69/+ (the following alleles were used in this study: rde-1(ne300) unc-42, rde-1(ne219), rde-2(ne221), rde-4(ne299), and mut-7(pk2040) were injected with pos-1 dsRNA. In each case, two types of F1 self progeny, distinguished by the presence of the linked marker mutations, were scored for interference ( FIG. 8A ).
  • F 1 male progeny were generated that carry the interference effect as well as one mutant copy of each respective locus; rde-1, rde-2, and mut-7 ( FIG. 9A ).
  • Each of these males was then backcrossed with uninjected hermaphrodites homozygous for each corresponding mutant ( FIG. 9A ).
  • the resulting cross progeny (F1) included 50% heterozygotes and 50% homozygotes that were distinguished by the presence of the linked marker mutations.
  • the heterozygous siblings served as controls and in each case exhibited interference at a frequency similar to that seen in wild-type animals ( FIG. 9A ).
  • rde-2 and mut-7 homozygous F2 progeny failed to exhibit interference, indicating that the activities of these two genes are required for interference in the F2 generation.
  • homozygous rde-1 F2 animals exhibited wild-type levels of F2 interference ( FIG. 9A ).
  • Control rde-1 homozygotes generated through identical crosses were completely resistant to pos-1::RNAi when challenged de novo with dsRNA in the F2 generation.
  • 35 rde-1 homozygous animals generated through crosses shown in FIG. 9A were tested by feeding bacteria expressing pos-1 dsRNA, and 21 similar animals were tested by direct injections of pos-1 dsRNA. All animals tested were resistant to pos-1 (RNAi).
  • RNAi pos-1 activity in the preceding generations was sufficient to allow interference to occur in rde-1 mutant F2 animals while the wild-type activities of rde-2 and mut-7 were required directly in the F2 animals for interference.
  • rde-1(+);rde-2( ⁇ ) animals were injected with pos-1 dsRNA and then crossed to generate F1 hermaphrodites homozygous for rde-1( ⁇ ); rde-2(+).
  • rde-1(+) activity in the injected animals was sufficient for F1 interference even when the injected animals were homozygous for rde-2 or mut-7 mutations ( FIG. 10B ).
  • rde-1(+) activity in the injected animals was not sufficient when the injected animals were homozygous for rde-4 mutant ( FIG. 10B ).
  • rde-1 can act independently of rde-2 and mut-7 in the injected animal, but rde-1 and rde-4 must function together.
  • the above Examples provide genetic evidence for the formation and transmission of extragenic interfering agents in the C. elegans germline.
  • Two C elegans genes, rde-1, and rde-4 appear to be necessary for the formation of these extragenic agents but not for interference mediated by them.
  • the activities of two other genes, rde-2 and mut-7 are required only downstream for interference.
  • rde-1 and rde-4 gene products or their homologs can be used to prepare agents effective in activating the RNAi pathway.
  • An rde-4 gene was cloned using methods similar to those described in Example 6.
  • the nucleic acid sequence (SEQ ID NO:4) and predicted amino acid sequence (SEQ ID NO:5) are illustrated in FIG. 10 .
  • rde-4 nucleic acid sequence shows that it encodes a protein (RDE-4) with similarities to dsRNA binding proteins.
  • RDE-4 protein
  • RDE-4 Three regions have been identified within the predicted RDE-4 protein corresponding to conserved regions found in all members of this dsRNA binding domain family. These regions appear to be important for proper folding of the dsRNA binding domain. conserveed amino acid residues, important for interactions with the backbone of the dsRNA helix, are found in all members of the protein family including RDE-4 (see consensus residues in FIG. 11 ). This motif is thought to provide for general non-sequence-specific interactions with dsRNA. The RDE-4 protein contains conserved protein folds that are thought to be important for the assembly of the dsRNA binding domain in this family of proteins. conserveed amino acid residues in RDE-4 are identical to those that form contacts with the dsRNA in the crystal structure of the X1 RBP dsRNA complex. These findings strongly suggest that RDE-4 is likely to have dsRNA binding activity.
  • RDE-4 contains a motif that is likely to bind in a general fashion to any dsRNA and because RDE-4 appears to function upstream in the generation of RNAi agents
  • the RDE-4 protein or fragments thereof can be used to convert any dsRNA into an RNAi agent.
  • RDE-4 contains other functional domains that may mediate the formation of RNAi agents. These domains may provide for interaction between RDE-4 and RDE-1 or for binding to enzymes such as nucleases that convert the dsRNA into the RNAi agent. Because of its RNA binding function in RNA interference, the RDE-4 protein and fragments thereof can be used to prepare dsRNA that is useful in preparing an RNAi agent.
  • In vivo and in vitro assays are used to identify regions in RDE-1 and RDE-4 that are important for the generation of RNAi agents.
  • rde-1 and rde-4 are introduced into the corresponding C. elegans mutant strains via transgenes (Tabara et al., Cell 99:123 (1999); and Example 13).
  • Important functional domains in RDE-1 and RDE-4 are defined by systematically altering the proteins followed by reintroduction into mutant animals to test for rescue of the RNAi deficient phenotype. A series of nested deletions are analyzed for rescue activity for both rde-1 and rde-4. Specific point mutations are used to analyze the importance of specific amino acids.
  • Chimera's are produced between RDE proteins and related proteins and genes. For example, coding sequences from RDE-1 homologs from the worm or from human are tested for their ability to rescue rde-1 mutants. Replacing the RDE-4 dsRNA binding motif with a distinct RNA binding motif, e.g., one that recognizes a specific viral dsRNA sequence or a ssRNA sequence will alter the specificity of the RNAi response perhaps causing sequence-specific or ssRNA-induced gene targeting. In one form of the in vitro assay, whole protein extracts from rde-1 or rde-4 deficient worm strains are used.
  • Recombinant RDE-1 or RDE-4 is then added back to reconstitute the extract.
  • Altered RDE-1 and RDE-4 proteins (described above, including deletions, point mutants and chimeras) are made in vitro and then tested for their ability to function when added back to these extracts.
  • RNAi activity is analyzed by injecting the reconstituted extracts directly into animals or by assaying for the destruction of an added in vitro synthesized target mRNA.
  • RNAi pathway rescue of animals e.g., C. elegans
  • RNAi pathway genes that encode functional polypeptides, e.g., polypeptides that can eliminate the mutant phenotype.
  • PCR using primers located 1 kb upstream and 500 nucleotides downstream of the open reading frame (T20G5.11; illustrated in FIG. 12 ) are used to amplify the rde-4 gene from C. elegans genomic DNA.
  • the resulting PCR product is then injected along with reporter constructs described in Tabara et al. (Cell 99:123 (1999); incorporated herein in its entirety by reference), and the progeny of the injected animal are assayed for rescue of the RNAi deficient phenotype.
  • the PCR product can also be cloned into a plasmid vector for site directed mutational analysis of RDE-4 (see Example 12). Co-injection of such a wild type RDE-4 plasmid and altered derivatives can be used to identify functional domains of rde-4. Similar methods can be used to identify functional domains of rde-1 and other RNAi pathway components.

Abstract

Genes involved in double-stranded RNA interference (RNAi pathway genes) are identified and used to investigate the RNAi pathway. The genes and their products are also useful for modulating RNAi pathway activity.

Description

    RELATED APPLICATION INFORMATION
  • This application claims priority from provisional application Ser. No. 60/159,776, filed Oct. 15, 1999, and 60/193,218, filed Mar. 30, 2000.
  • STATEMENT AS TO FEDERALLY SPONSORED RESEARCH
  • Funding for the work described herein was provided by the federal government (GM58800 and GM37706), which has certain rights in the invention.
  • FIELD OF THE INVENTION
  • This invention relates to the discovery of genes whose expression products are involved in mediation of genetic interference.
  • BACKGROUND OF THE INVENTION
  • All eukaryotic organisms share similar mechanisms for information transfer from DNA to RNA to protein. RNA interference represents an efficient mechanism for inactivating this transfer process for a specific targeted gene. Targeting is mediated by the sequence of the RNA molecule introduced to the cell. Double-stranded (ds) RNA can induce sequence-specific inhibition of gene function (genetic interference) in several organisms including the nematode, C. elegans (Fire, et al., 1998, Nature 391:806-811), plants, trypanosomes, Drosophila, and planaria (Waterhouse et al., 1998, Proc. Natl. Acad. Sci. USA 94:13959-13964; Ngo et al., 1998, Proc. Natl. Acad. Sci. USA 95:14687-14692; Kennerdell and Carthew, 1998, Cell 95:1017-1026; Misquitta and Patterson, 1999, Proc. Natl. Acad. Sci. USA 96:1451-1456; Sanchez-Alvorado and Newmark, 1999, Proc. Natl. Acad. Sci. USA 96:5049-5054). The discovery that dsRNA can induce genetic interference in organisms from several distinct phyla suggests a conserved mechanism and perhaps a conserved physiological role for the interference process. Although several models of RNAi have been proposed (Baulcombe, 1999, Curr. Biol. 9:R599-R601; Sharp, 1999, Genes & Dev. 13:139-141) the mechanisms of action of specific components of the pathway are not known.
  • Attempts to overexpress a gene (e.g., a transgene) often lead only to transient expression of the gene. Furthermore, the even more undesirable effect of “cosuppression” can occur in which a corresponding endogenous copy of the transgene becomes inactivated. In some cases, transgene silencing leads to problems with the commercial or therapeutic application of transgenic technology to alter the genetic makeup of a cell, organism, or human patient.
  • SUMMARY OF THE INVENTION
  • The present invention relates to the discovery of RNA interference (RNAi) pathway genes which are involved in mediating double-stranded RNA-dependent gene silencing (genetic interference). RNAi requires a set of conserved cellular factors to suppress gene expression. These factors are the components of the RNAi pathway. The RNAi pathway mutations and genes described herein (e.g., rde-1, rde-2, rde-3, rde-4, rde-5, mut-2, and mut-7), and their protein products (e.g., RDE-1 and RDE-4) are useful tools for investigating the mechanisms involved in RNAi and developing methods of modulating the RNAi pathway. The sequences and methods described herein are useful for modulating the RNAi pathway and may be used in conjunction with other methods involving the use of genetic inhibition by dsRNA (e.g., see U.S. Ser. No. 09/215,257, filed Dec. 18, 1998, incorporated herein by reference in its entirety).
  • RNAi pathway components (e.g., RDE-1, RDE-4) provide activities necessary for interference. These activities may be absent or not sufficiently activated in many cell types, including those of organisms such as humans in which genetic interference may have potential therapeutic value. Components of the RNAi pathway in C. elegans may be sufficient when provided through transgenesis or as direct RNA:protein complexes to activate or directly mediate genetic interference in heterologous cells that are deficient in RNAi.
  • Nucleic acid sequences encoding RNAi pathway components (e.g., RDE-1, RDE-4) are useful, e.g., for studying the regulation of the RNAi pathway. Such sequences can also be used to generate knockout strains of animals such as C. elegans.
  • The nucleic acids of the invention include nucleic acids that hybridize, e.g., under stringent hybridization conditions (as defined herein), to all or a portion of the nucleotide sequence of SEQ ID NO:1 (FIG. 5A-C) or its complement; SEQ ID NO:2 (FIG. 6A-D) or its complement, or SEQ ID NO:4 or its complement. The hybridizing portion of the hybridizing nucleic acids are preferably 20, 30, 50, or 70 bases long. Preferably, the hybridizing portion of the hybridizing nucleic acid is 80%, more preferably 95%, or even 98% or 100% identical to the sequence of a portion or all of a nucleic acid encoding an RDE-1 polypeptide or an RDE-4 polypeptide. Hybridizing nucleic acids of the type described above can be used as a cloning probe, a primer (e.g., a PCR primer), or a diagnostic probe. Preferred hybridizing nucleic acids encode a polypeptide having some or all of the biological activities possessed by a naturally-occurring RDE-1 polypeptide or an RDE-4 polypeptide e.g., as determined in the assays described below.
  • Hybridizing nucleic acids may encode a protein that is shorter or longer than the RDE-1 protein or RDE-4 protein described herein. Hybridizing nucleic acids may also encode proteins that are related to RDE-1 or RDE-4 (e.g., proteins encoded by genes that include a portion having a relatively high degree of identity to the rde-1 gene or rde-4 gene described herein).
  • The invention also features purified or isolated RDE-1 polypeptides and RDE-4 polypeptides. RDE-1 and RDE-4 polypeptides are useful for generating and testing antibodies that specifically bind to an RDE-1 or an RDE-4. Such antibodies can be used, e.g., for studying the RNAi pathway in C. elegans and other organisms. As used herein, both “protein” and “polypeptide” mean any chain of amino acids, regardless of length or post-translational modification (e.g., glycosylation or phosphorylation). Thus, the term “RNAi pathway polypeptide” includes a full-length, naturally occurring RNAi pathway polypeptide such as RDE-1 protein or RDE-4 protein, as well as recombinantly or synthetically produced polypeptides that correspond to a full-length, naturally occurring RDE-1 protein, RDE-4 protein, or to particular domains or portions of a naturally occurring RNAi pathway protein.
  • RNAi pathway mutations and strains harboring those mutations (e.g., rde-1, rde-2, rde-3, rde-4, rde-5) are useful for studying the RNAi pathway, including identification of modulators of the RNAi pathway.
  • RNAi pathway components (e.g., those associated with mut-7 and rde-2) can be used to desilence or prevent silencing of transgenes. To facilitate this function, such RNAi pathway components are inhibited using specific inhibitors of an RNAi pathway gene or its product.
  • In one embodiment, the invention includes an isolated nucleic acid molecule comprising a nucleotide sequence encoding an RDE-1 polypeptide. The nucleic acid molecule hybridizes under high stringency conditions to the nucleic acid sequence of Genbank Accession No. AF180730 (SEQ ID NO:2) or its complement, or the sequence of SEQ ID NO:1 or its complement. In one embodiment, the isolated nucleic acid can complement an rde-1 mutation. The invention also encompasses an isolated nucleic acid whose nucleotide sequence encodes the amino acid sequence of SEQ ID NO:3.
  • The invention also encompasses a substantially pure RDE-1 polypeptide encoded by the isolated nucleic acids described herein.
  • The invention features an antibody that specifically binds to an RDE-1 polypeptide.
  • The invention also includes a method of enhancing the expression of a transgene in a cell, the method comprising decreasing activity of the RNAi pathway. In one embodiment of this invention, rde-2 expression or activity is decreased.
  • The invention also features an isolated nucleic acid molecule comprising a nucleotide sequence encoding an RDE-4 polypeptide, wherein the nucleic acid molecule hybridizes under high stringency conditions to the nucleic acid sequence of SEQ ID NO:4 or its complement. The invention also encompasses an isolated nucleic acid encoding an RDE-4 polypeptide, wherein the nucleic acid can complement an rde-4 mutation. The invention also encompasses an isolated nucleic acid encoding an RDE-4 polypeptide, in which the nucleotide sequence encodes the amino acid sequence of SEQ ID NO:5.
  • The invention also features a substantially pure RDE-4 polypeptide encoded by the isolated nucleic acids described herein.
  • In another embodiment the invention features an antibody that specifically binds to an RDE-4 polypeptide.
  • The invention also features a method of preparing an RNAi agent, the method includes incubating a dsRNA in the presence of an RDE-1 protein and an RDE-4 protein.
  • The invention also features a method of inhibiting the activity of a gene by introducing an RNAi agent into a cell, such that the dsRNA component of the RNAi agent is targeted to the gene. In another embodiment of the invention, the cell contains an exogenous RNAi pathway sequence. The exogenous RNAi pathway sequence can be an RDE-1 polypeptide or an RDE-4 polypeptide. In still another embodiment, a dsRNA is introduced into a cell containing an exogenous RNAi pathway sequence such as nucleic acid sequence expressing an RDE-1 or RDE-4.
  • An RNAi pathway component is a protein or nucleic acid that is involved in promoting dsRNA-mediated genetic interference. A nucleic acid component can be an RNA or DNA molecule. A mutation in a gene encoding an RNAi pathway component may decrease or increase RNAi pathway activity.
  • An RNAi pathway protein is a protein that is involved in promoting dsRNA mediated genetic interference.
  • A “substantially pure DNA” is a DNA that is not immediately contiguous with (i.e., covalently linked to) both of the coding sequences with which it is immediately contiguous (i.e., one at the 5′ end and one at the 3′ end) in the naturally-occurring genome of the organism from which the DNA of the invention is derived. The term therefore includes, for example, a recombinant DNA which is incorporated into a vector, into an autonomously replicating plasmid or virus, or into the genomic DNA of a prokaryote or eukaryote; or which exists as a separate molecule (e.g., a cDNA or a genomic or cDNA fragment produced by PCR (polymerase chain reaction) or restriction endonuclease digestion) independent of other sequences. It also includes a recombinant DNA which is part of a hybrid gene encoding additional polypeptide sequences.
  • By “inhibited RNAi pathway” is meant decreased inhibitory activity of a dsRNA which results in at least two-fold less inhibition by a dsRNA relative to its ability to cause inhibition in a wild type cell Techniques for measuring RNAi pathway activity are described herein. The pathway can be inhibited by inhibiting a component of the pathway (e.g., RDE-1) or mutating the component so that its function is reduced.
  • A “substantially pure polypeptide” is a polypeptide, e.g., an RNAi pathway polypeptide or fragment thereof, that is at least 60%, by weight, free from the proteins and naturally-occurring organic molecules with which it is naturally associated. Preferably, the preparation is at least 75%, more preferably at least 90%, and most preferably at least 99%, by weight, RNAi pathway polypeptide or fragment. A substantially pure RNAi pathway polypeptide or fragment thereof is obtained, for example, by extraction from a natural source; by expression of a recombinant nucleic acid encoding an RNAi pathway polypeptide or fragment thereof; or by chemically synthesizing the polypeptide or fragment. Purity can be measured by any appropriate method, e.g., column chromatography, polyacrylamide gel electrophoresis, or HPLC analysis.
  • By “specifically binds” is meant a molecule that binds to a particular entity, e.g., an RNAi pathway polypeptide, but which does not substantially recognize or bind to other molecules in a sample, e.g., a biological sample, which includes the particular entity, e.g., RDE-1.
  • An RNAi agent is a dsRNA molecule that has been treated with those components of the RNAi pathway that are required to confer RNAi activity on the dsRNA. For example, treatment of a dsRNA under conditions that include RDE-1 and RDE-4 results in an RNAi agent. Injection of such an agent into an animal that is mutant for RDE-1 and RDE-4 will result in activation of the RNAi pathway with respect to a targeted gene. Typically, the dsRNA used to trigger the formation of the RNAi agent is selected to be an RNA corresponding to all or a portion of the nucleotide sequence of the targeted gene.
  • Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, suitable methods and materials are described below. All publications, patent applications, patents, and other references mentioned herein are incorporated by reference. In addition, the materials, methods, and examples are illustrative only and not intended to be limiting.
  • Other features and advantages of the invention will be apparent from the detailed description, and from the claims.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1A illustrates the genetic scheme used to identify rde mutants.
  • FIG. 1B is an illustration summarizing data from the genetic mapping of rde and mut mutations. The vertical bars represent chromosomes; LGI, LGIII, and LGV. Reference genetic markers are indicated at the right of each chromosome and the relative genetic positions of the rde and mut alleles are indicated at the left.
  • FIG. 2A is a graphical representation of experiments investigating the sensitivity of rde and mut strains to RNAi by microinjection. The RNA species indicated above each graph was injected at high concentration (pos-1: 7 mg/ml, par-2: 3 mg/ml, sqt-3: 7 mg/ml). The strains receiving injection are indicated at the left and the horizontal bar graphs reflect the percent of progeny that exhibited genetic interference. The Unc marker mutants used are also indicated. The percent embryonic lethality of F1 progeny is plotted as shaded bars and the fraction of affected progeny is indicated at the right of each graph.
  • FIG. 2B is a graphical representation of experiments demonstrating that animals homozygous for rde and mut alleles are resistant to RNAi targeting maternally expressed genes, pos-1 and par-2. The percent embryonic lethality of F1 progeny is plotted as shaded bars and the fraction of affected progeny is indicated at the right of each graph.
  • FIG. 3 is a schematic representation of homozygous rde-1 (ne219) and rde-4(ne299) mutant mothers receiving injections of dsRNA targeting the body muscle structural gene unc-22.
  • FIG. 4A is a schematic representation of the physical map of the rde-1 region. C. elegans YAC and cosmid DNA clones that were positive for rescue are indicated by an asterisk. A representation of the expanded interval showing a minimal, 25 kb, rescuing interval defined by the overlap between cosmids T10A5 and C27H6 is shown beneath the YAC and cosmid map. Predicted genes within this sequenced interval are illustrated above and below the hatch marked line. A single, rescuing, 4.5 kb PCR fragment containing the KO8H.107 predicted gene is shown enlarged. Exon and intron (box/line) boundaries are shown as well as the positions of rde-1 point mutation in the predicted coding sequences.
  • FIG. 4B is an illustration of the predicted sequence of RDE-1 and its alignment with four related proteins. The sequences are RDE-1 (C. elegans; Genbank Accession No. AF180730), F48F7.1 (C. elegans; Genbank Accession No. Z69661), eIF2C (rabbit; Genbank Accession No. AF005355), ZWILLE (Arabidopsis; Genbank Accession No. AJ223508), and Sting (Drosophila; Genbank Accession No. AF145680). Identities with RDE-1 are shaded in black, and identities among the homologs are shaded in gray.
  • FIGS. 5A-5C are an illustration of the genomic sequence from cosmid K08H10 (Genbank accession Z83113.1; SEQ ID NO:1) corresponding to the rde-1 gene from the first nucleotide of 5′ untranslated region to the polyadenylation site.
  • FIGS. 6A-6D are an illustration of the cDNA sequence of rde-1 (SEQ ID NO:2), including the first 20 nucleotides constituting the 5′ untranslated sequence (5′UTR) and the predicted amino acid sequence encoded by rde-1 (RDE-1; SEQ ID NO:3). The nucleotide sequence is numbered starting with the first nucleotide of the translated region.
  • FIG. 7A is an illustration of the protocol for injection of a wild-type hermaphrodite with dsRNA.
  • FIG. 7B is an illustration of a genetic scheme demonstrating extragenic inheritance of RNAi. The fraction shown represents the number of RNAi affected F2 hermaphrodites over the total number of cross progeny scored for each genotype class. Phenotypically uncoordinated (Unc).
  • FIGS. 8A-8B are illustrations of a genetic scheme to determine if the wild-type activities of rde-1, rde-2, rde-4, and mut-7 are sufficient in the injected animal for interference among the F1 self progeny (A) illustrates crosses of heterozygous hermaphrodites; (B) illustrates crosses using homzygous F1 progeny from heterozygous mothers. The fraction shown represents the number of RNAi affected animals over the total number of cross progeny scored for each genotype class.
  • FIG. 9A depicts experiments of a the genetic scheme to determine if the wild-type activities of rde-1, rde-2, rde-4, and mut-7 are sufficient in the injected animal for interference among the F1 self progeny. The fraction shown represents the number of RNAi affected animals over the total number of cross progeny scored for each genotype class.
  • FIG. 9B depicts experiments designed to determine the requirements for rde-1, rde-2, rde-4, and mut-7 in F2 (FIG. 10A) and F1 (FIG. 10B) interference. The fraction shown represents the number of RNAi affected animals over the total number of cross progeny scored for each genotype class.
  • FIGS. 10A-10B are a depiction of the cDNA sequence of a wild type rde-4 nucleic acid sequence (SEQ ID NO:4) and the predicted RDE-4 amino acid sequence (SEQ ID NO:5) of C. elegans. “*” indicates ambiguous base assignment.
  • FIG. 11 is a depiction of regions of homology between the predicted RDE-4 amino acid sequence, X1RBPA (SEQ ID NO:6), HsPKR (SEQ ID NO:7), and a consensus sequence (SEQ ID NO:8). A predicted secondary structure for RDE-4 is also shown illustrating predicted regions of α helix and β pleated sheet.
  • FIG. 12 illustrates a scheme for rescue of an rde-4.
  • DETAILED DESCRIPTION
  • Mutations have been discovered that identify genes involved in dsRNA-mediated genetic interference (RNAi). RNAi pathway genes encode products involved in genetic interference and are useful for mediating or enhancing genetic interference. These genes encode mediators of double-stranded RNA-mediated interference. The mediators can be nucleic acid or protein. RNAi pathway genes are also useful for mediating specific processes, e.g., a gene that mediates dsRNA uptake by cells may be useful for transporting other RNAs into cells or for facilitating entry of agents such as drugs into cells. The methods and examples described below illustrate the identification of RNAi pathway components, the uses of RNAi pathway components, mutants, genes and their products.
  • Identification of an RNAi-Deficient Mutants and an RNAi Pathway Gene rde-1
  • RNAi pathway genes were identified using screens for C. elegans strains mutant for RNAi (Examples 2 and 3). The mutations were further characterized for germline and somatic effects, effects on transposon mobilization, X chromosome loss and transgene silencing, and target tissue activity (Examples 4 and 5).
  • The rde-1 gene was identified using YACs (yeast artificial chromosomes) and cosmids to rescue rde-1 mutants. Based on the identified sequence, a cDNA sequence was identified in a C. elegans cDNA library and the complete cDNA sequence determined (Example 6).
  • Identification of RNAi Pathway Genes Homologous to rde-1, rde-2, rde-3, and rde-4
  • RNAi pathway genes from C. elegans (such as those described herein) and from other organisms (e.g. plant, mammalian, especially human) are useful for the elucidation of the biochemical pathways involved in genetic interference and for developing the uses of RNAi pathway genes described herein.
  • Several approaches can be used to isolate RNAi pathway genes including two-hybrid screens, complementation of C. elegans mutants by expression libraries of cloned heterologous (e.g., plant, mammalian, human) cDNAs, polymerase chain reactions (PCR) primed with degenerate oligonucleotides, low stringency hybridization screens of heterologous cDNA or genomic libraries with a C. elegans RNAi pathway gene, and database screens for sequences homologous to an RNAi pathway gene. Hybridization is performed under stringent conditions. Alternatively, a labeled fragment can be used to screen a genomic library derived from the organism of interest, again, using appropriately stringent conditions. Such stringent conditions are well known, and will vary predictably depending on the specific organisms from which the library and the labeled sequences are derived.
  • Nucleic acid duplex or hybrid stability is expressed as the melting temperature or Tm, which is the temperature at which a probe dissociates from a target DNA. This melting temperature is used to define the required stringency conditions. If sequences are to be identified that are related and substantially identical to the probe, rather than identical, then it is useful to first establish the lowest temperature at which only homologous hybridization occurs with a particular SSC or SSPE concentration. Then assume that 1% mismatching results in 1° C. decrease in the Tm and reduce the temperature of the final wash accordingly (for example, if sequences with ≧95% identity with the probe are sought, decrease the final wash temperature by 5° C.). Note that this assumption is very approximate, and the actual change in Tm can be between 0.5° and 1.5° C. per 1% mismatch.
  • As used herein, high stringency conditions include hybridizing at 68° C. in 5×SSC/5× Denhardt solution/1.0% SDS, or in 0.5 M NaHPO4 (pH 7.2)/1 mM EDTA/7% SDS, or in 50% formamide/0.25 M NaHPO4 (pH 7.2)/0.25 M NaCl/1 mM EDTA/7% SDS; and washing in 0.2×SSC/0.1% SDS at room temperature or at 42° C., or in 0.1×SSC/0.1% SDS at 68° C., or in 40 mM NaHPO4 (pH 7.2)/1 mM EDTA/5% SDS at 50° C., or in 40 mM NaHPO4 (pH 7.2) 1 mM EDTA/1% SDS at 50° C. Moderately stringent conditions include washing in 3×SSC at 42° C. The parameters of salt concentration and temperature can be varied to achieve the desired level of identity between the probe and the target nucleic acid.
  • For guidance regarding such conditions see, for example, Sambrook et al., 1989, Molecular Cloning, A Laboratory Manual, Cold Springs Harbor Press, N.Y.; and Ausubel et al. (eds.), 1995, Current Protocols in Molecular Biology, (John Wiley & Sons, N.Y.) at Unit 2.10.
  • Methods of screening for and identifying homologs of C. elegans RNAi genes (e.g., rde-1) are known in the art. For example, complementation of mutants, described in the Examples can be performed using nucleic acid sequences from organisms other than C. elegans. Methods of inhibiting expression of a target gene in a cell using dsRNA are known in the art and are exemplified in U.S. Ser. No. 09/215,257, filed Dec. 18, 1998, which is incorporated herein by reference in its entirety.
  • Another method of screening is to use an identified RNAi pathway gene sequence to screen a cDNA or genomic library using low stringency hybridizations. Such methods are known in the art.
  • PCR with degenerate oligonucleotides is another method of identifying homologs of RNAi pathway genes (e.g., human rde-1). Homologs of an RNAi pathway gene identified in other species are compared to identify specific regions with a high degree of homology (as in the sequence comparison shown in FIG. 4). These regions of high homology are selected for designing PCR primers that maximize possible base-pairing with heterologous genes. Construction of such primers involves the use of oligonucleotide mixtures that account for degeneracy in the genetic code, i.e., allow for the possible base changes in an RNAi pathway gene that does not affect the amino acid sequence of the RNAi pathway protein. Such primers may be used to amplify and clone possible RNAi pathway gene fragments from DNA isolated from another organism (e.g., mouse or human). The latter are sequenced and those encoding protein fragments with high degrees of homology to fragments of the RNAi pathway protein are used as nucleic acid probes in subsequent screens of genomic DNA and cDNA libraries (e.g., mouse or human). Full-length genes and cDNAs having substantial homology to the previously identified RNAi pathway gene are identified in these screens.
  • To produce an RNAi pathway gene product (e.g., RDE-1) a sequence encoding the gene is placed in an expression vector and the gene expressed in an appropriate cell type. The gene product is isolated from such cell lines using methods known to those in the art, and used in the assays and procedures described herein. The gene product can be a complete RNAi pathway protein (e.g., RDE-1) or a fragment of such a protein.
  • Methods of Expressing RNAi Pathway Proteins
  • Full-length polypeptides and polypeptides corresponding to one or more domains of a full-length RNAi pathway protein, e.g., the RNA-binding domain of RDE-4, are also within the scope of the invention. Also within the invention are fusion proteins in which a portion (e.g., one or more domains) of an RDE-1 or RDE-4) is fused to an unrelated protein or polypeptide (i.e., a fusion partner) to create a fusion protein. The fusion partner can be a moiety selected to facilitate purification, detection, or solubilization, or to provide some other function. Fusion proteins are generally produced by expressing a hybrid gene in which a nucleotide sequence encoding all or a portion of of an RNAi pathway protein is joined in-frame to a nucleotide sequence encoding the fusion partner. Fusion partners include, but are not limited to, the constant region of an immunoglobulin (IgFc). A fusion protein in which an RNAi pathway polypeptide is fused to IgFc can be more stable and have a longer half-life in the body than the polypeptide on its own.
  • In general, RNAi pathway proteins (e.g., RDE-1, RDE-4) according to the invention can be produced by transformation (transfection, transduction, or infection) of a host cell with all or part of an RNAi pathway protein-encoding DNA fragment (e.g., one of the cDNAs described herein) in a suitable expression vehicle. Suitable expression vehicles include: plasmids, viral particles, and phage. For insect cells, baculovirus expression vectors are suitable. The entire expression vehicle, or a part thereof, can be integrated into the host cell genome. In some circumstances, it is desirable to employ an inducible expression vector, e.g., the LACSWITCH™ Inducible Expression System (Stratagene; LaJolla, Calif.).
  • Those skilled in the field of molecular biology will understand that any of a wide variety of expression systems can be used to provide the recombinant protein. The precise host cell used is not critical to the invention. The RNAi pathway protein can be produced in a prokaryotic host (e.g., E. coli or B. subtilis) or in a eukaryotic host (e.g., Saccharomyces or Pichia; mammalian cells, e.g., COS, NIH 3T3 CHO, BHK, 293, or HeLa cells; or insect cells).
  • Proteins and polypeptides can also be produced in plant cells. For plant cells viral expression vectors (e.g., cauliflower mosaic virus and tobacco mosaic virus) and plasmid expression vectors (e.g., Ti plasmid) are suitable. Such cells are available from a wide range of sources (e.g., the American Type Culture Collection, Rockland, Md.; also, see, e.g., Ausubel et al., Current Protocols in Molecular Biology, John Wiley & Sons, New York, 1994). The methods of transformation or transfection and the choice of expression vehicle will depend on the host system selected. Transformation and transfection methods are described, e.g., in Ausubel et al., supra; expression vehicles may be chosen from those provided, e.g., in Cloning Vectors: A Laboratory Manual (P. H. Pouwels et al., 1985, Supp. 1987).
  • The host cells harboring the expression vehicle can be cultured in conventional nutrient media adapted as need for activation of a chosen gene, repression of a chosen gene, selection of transformants, or amplification of a chosen gene.
  • One preferred expression system is the mouse 3T3 fibroblast host cell transfected with a pMAMneo expression vector (Clontech, Palo Alto, Calif.). pMAMneo provides an RSV-LTR enhancer linked to a dexamethasone-inducible MMTV-LTR promotor, an SV40 origin of replication which allows replication in mammalian systems, a selectable neomycin gene, and SV40 splicing and polyadenylation sites. DNA encoding an RNAi pathway protein would be inserted into the pMAMneo vector in an orientation designed to allow expression. The recombinant RNAi pathway protein would be isolated as described herein. Other preferable host cells that can be used in conjunction with the pMAMneo expression vehicle include COS cells and CHO cells (ATCC Accession Nos. CRL 1650 and CCL 61, respectively).
  • RNAi pathway polypeptides can be produced as fusion proteins. For example, the expression vector pUR278 (Ruther et al., EMBO J. 2:1791, 1983), can be used to create lacZ fusion proteins. The pGEX vectors can be used to express foreign polypeptides as fusion proteins with glutathione S-transferase (GST). In general, such fusion proteins are soluble and can be easily purified from lysed cells by adsorption to glutathione-agarose beads followed by elution in the presence of free glutathione. The pGEX vectors are designed to include thrombin or factor Xa protease cleavage sites so that the cloned target gene product can be released from the GST moiety.
  • In an insect cell expression system, Autographa californica nuclear polyhidrosis virus (AcNPV), which grows in Spodoptera frugiperda cells, is used as a vector to express foreign genes. An RNAi pathway protein coding sequence can be cloned individually into non-essential regions (for example the polyhedrin gene) of the virus and placed under control of an AcNPV promoter, e.g., the polyhedrin promoter. Successful insertion of a gene encoding an RNAi pathway polypeptide or protein will result in inactivation of the polyhedrin gene and production of non-occluded recombinant virus (i.e., virus lacking the proteinaceous coat encoded by the polyhedrin gene). These recombinant viruses are then used to infect spodoptera frugiperda cells in which the inserted gene is expressed (see, e.g., Smith et al., J. Virol. 46:584, 1983; Smith, U.S. Pat. No. 4,215,051).
  • In mammalian host cells, a number of viral-based expression systems can be utilized. When an adenovirus is used as an expression vector, the RNAi pathway protein nucleic acid sequence can be ligated to an adenovirus transcription/translation control complex, e.g., the late promoter and tripartite leader sequence. This chimeric gene can then be inserted into the adenovirus genome by in vitro or in vivo recombination. Insertion into a non-essential region of the viral genome (e.g., region E1 or E3) will result in a recombinant virus that is viable and capable of expressing an RNAi pathway gene product in infected hosts (see, e.g., Logan, Proc. Natl. Acad. Sci. USA 81:3655, 1984).
  • Specific initiation signals may be required for efficient translation of inserted nucleic acid sequences. These signals include the ATG initiation codon and adjacent sequences. In cases where an entire native RNAi pathway protein gene or cDNA, including its own initiation codon and adjacent sequences, is inserted into the appropriate expression vector, no additional translational control signals may be needed. In other cases, exogenous translational control signals, including, perhaps, the ATG initiation codon, must be provided. Furthermore, the initiation codon must be in phase with the reading frame of the desired coding sequence to ensure translation of the entire insert. These exogenous translational control signals and initiation codons can be of a variety of origins, both natural and synthetic. The efficiency of expression may be enhanced by the inclusion of appropriate transcription enhancer elements, transcription terminators (Bittner et al., Methods in Enzymol. 153:516, 1987).
  • RNAi pathway polypeptides can be expressed directly or as a fusion with a heterologous polypeptide, such as a signal sequence or other polypeptide having a specific cleavage site at the N-and/or C-terminus of the mature protein or polypeptide. Included within the scope of this invention are RNAi pathway polypeptides with a heterologous signal sequence. The heterologous signal sequence selected should be one that is recognized and processed, i.e., cleaved by a signal peptidase, by the host cell. For prokaryotic host cells a prokaryotic signal sequence is selected, for example, from the group of the alkaline phosphatase, penicillinase, lpp, or heat-stable enterotoxin II leaders. For yeast secretion a yeast invertase, alpha factor, or acid phosphatase leaders may be selected. In mammalian cells, it is generally desirable to select a mammalian signal sequences.
  • A host cell may be chosen which modulates the expression of the inserted sequences, or modifies and processes the gene product in a specific, desired fashion. Such modifications (e.g., glycosylation) and processing (e.g., cleavage) of protein products may be important for the function of the protein. Different host cells have characteristic and specific mechanisms for the post-translational processing and modification of proteins and gene products. Appropriate cell lines or host systems can be chosen to ensure the correct modification and processing of the foreign protein expressed. To this end, eukaryotic host cells that possess the cellular machinery for proper processing of the primary transcript, glycosylation, and phosphorylation of the gene product can be used. Such mammalian host cells include, but are not limited to, CHO, VERO, BHK, HeLa, COS, MDCK, 293, 3T3, W138, and in particular, choroid plexus cell lines.
  • Alternatively, an RNAi pathway protein can be produced by a stably-transfected mammalian cell line. A number of vectors suitable for stable transfection of mammalian cells are available to the public, see e.g., Pouwels et al. (supra); methods for constructing such cell lines are also publicly available, e.g., in Ausubel et al. (supra). In one example, cDNA encoding an RNAi pathway protein (e.g., RDE-1 or RDE-4) is cloned into an expression vector that includes the dihydrofolate reductase (DHFR) gene. Integration of the plasmid and, therefore, the RNAi pathway protein-encoding gene into the host cell chromosome is selected for by including 0.01-300 μM methotrexate in the cell culture medium (as described in Ausubel et al., supra). This dominant selection can be accomplished in most cell types.
  • Recombinant protein expression can be increased by DHFR-mediated amplification of the transfected gene. Methods for selecting cell lines bearing gene amplifications are described in Ausubel et al. (supra); such methods generally involve extended culture in medium containing gradually increasing levels of methotrexate. DHFR-containing expression vectors commonly used for this purpose include pCVSEII-DHFR and pAdD26SV(A) (described in Ausubel et al., supra). Any of the host cells described above or, preferably, a DHFR-deficient CHO cell line (e.g., CHO DHFR cells, ATCC Accession No. CRL 9096) are among the host cells preferred for DHFR selection of a stably-transfected cell line or DHFR-mediated gene amplification.
  • A number of other selection systems can be used, including but not limited to the herpes simplex virus thymidine kinase, hypoxanthine-guanine phosphoribosyl-transferase, and adenine phosphoribosyltransferase genes can be employed in tk, hgprt, or aprt cells, respectively. In addition, gpt, which confers resistance to mycophenolic acid (Mulligan et al., Proc. Natl. Acad. Sci. USA, 78:2072, 1981); neo, which confers resistance to the aminoglycoside G-418 (Colberre-Garapin et al., J. Mol. Biol., 150:1, 1981); and hygro, which confers resistance to hygromycin (Santerre et al., Gene, 30:147, 1981), can be used.
  • Alternatively, any fusion protein can be readily purified by utilizing an antibody specific for the fusion protein being expressed. For example, a system described in Janknecht et al., Proc. Natl. Acad. Sci. USA, 88:8972 (1981), allows for the ready purification of non-denatured fusion proteins expressed in human cell lines. In this system, the gene of interest is subcloned into a vaccinia recombination plasmid such that the gene's open reading frame is translationally fused to an amino-terminal tag consisting of six histidine residues. Extracts from cells infected with recombinant vaccinia virus are loaded onto Ni2+ nitriloacetic acid-agarose columns, and histidine-tagged proteins are selectively eluted with imidazole-containing buffers.
  • Alternatively, an RNAi pathway protein or a portion thereof, can be fused to an immunoglobulin Fc domain. Such a fusion protein can be readily purified using a protein A column.
  • Antibodies that Recognize RNAi Pathway Proteins
  • Techniques for generating both monoclonal and polyclonal antibodies specific for a particular protein are well known. The invention also includes humanized or chimeric antibodies, single chain antibodies, Fab fragments, F(ab′)2 fragments, and molecules produced using a Fab expression library.
  • Antibodies can be raised against a short peptide epitope of an RNAi pathway gene (e.g., rde-1), an epitope linked to a known immunogen to enhance immunogenicity, a long fragment of an RNAi pathway gene, or the intact protein. Such antibodies are useful for e.g., localizing RNAi pathway polypeptides in tissue sections or fractionated cell preparations, determining whether an RNAi pathway gene is expressed (e.g., after transfection with an RNAi pathway gene), and evaluating the expression of an RNAi pathway gene in disorders (e.g., genetic conditions) where the RNAi pathway may be affected.
  • An isolated RNAi pathway protein (e.g., RDE-1), or a portion or fragment thereof, can be used as an immunogen to generate antibodies that bind to an RNAi pathway protein using standard techniques for polyclonal and monoclonal antibody preparation. The RNAi pathway immunogen can also be a mutant RNAi pathway protein or a fragment of a mutant RNAi pathway protein. A full-length RNAi pathway protein can be used or, alternatively, antigenic peptide fragments of RNAi pathway protein can be used as immunogens. The antigenic peptide of an RNAi pathway protein comprises at least 8 (preferably 10, 15, 20, or 30) amino acid residues. In the case of RDE-1, these residues are drawn from the amino acid sequence shown in SEQ ID NO:3 and encompass an epitope such that an antibody raised against the peptide forms a specific immune complex with RDE-1. Preferred epitopes encompassed by the antigenic peptide are regions of the protein that are located on the surface of the protein, e.g., hydrophilic regions.
  • An RNAi pathway protein immunogen typically is used to prepare antibodies by immunizing a suitable subject (e.g., rabbit, goat, mouse or other mammal) with the immunogen. An appropriate immunogenic preparation can contain, for example, recombinantly expressed RNAi pathway protein or a chemically synthesized RNAi polypeptide. The preparation can further include an adjuvant, such as Freund's complete or incomplete adjuvant, or similar immunostimulatory agent. Immunization of a suitable subject with an immunogenic RNAi pathway protein preparation induces a polyclonal anti-RNAi pathway protein antibody response.
  • Polyclonal antibodies that recognize an RNAi pathway protein (“RNAi pathway antibodies”) can be prepared as described above by immunizing a suitable subject with an RNAi pathway protein immunogen. The RNAi pathway antibody titer in the immunized subject can be monitored over time by standard techniques, such as with an enzyme-linked immunosorbent assay (ELISA) using immobilized RNAi pathway protein from which the immunogen was derived. If desired, the antibody molecules directed against the RNAi pathway protein can be isolated from the mammal (e.g., from the blood) and further purified by well-known techniques, such as protein A chromatography to obtain the IgG fraction. At an appropriate time after immunization, e.g., when the RNAi pathway antibody titers are highest, antibody-producing cells can be obtained from the subject and used to prepare monoclonal antibodies by standard techniques, such as the hybridoma technique originally described by Kohler and Milstein (1975) Nature 256:495-497, the human B cell hybridoma technique (Kozbor et al. (1983) Immunol. Today 4:72), the EBV-hybridoma technique (Cole et al. (1985), Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, Inc., pp. 77-96) or trioma techniques. The technology for producing hybridomas is well known (see generally Current Protocols in Immunology (1994) Coligan et al. (eds.) John Wiley & Sons, Inc., New York, N.Y.). Briefly, an immortal cell line (typically a myeloma) is fused to lymphocytes (typically splenocytes) from a mammal immunized with an RNAi pathway immunogen as described above, and the culture supernatants of the resulting hybridoma cells are screened to identify a hybridoma producing a monoclonal antibody that binds to the RNAi pathway protein.
  • Any of the many well known protocols used for fusing lymphocytes and immortalized cell lines can be applied for the purpose of generating a monoclonal antibody against an RNAi pathway protein (see, e.g., Current Protocols in Immunology, supra; Galfre et al., 1977, Nature 266:55052; R. H. Kenneth, in Monoclonal Antibodies: A New Dimension In Biological Analyses, Plenum Publishing Corp., New York, N.Y., 1980; and Lerner, 1981, Yale J. Biol. Med., 54:387-402. Moreover, one in the art will appreciate that there are many variations of such methods which also would be useful. Hybridoma cells producing a monoclonal antibody of the invention are detected by screening the hybridoma culture supernatants for antibodies that bind to the RNAi pathway protein, e.g., using a standard ELISA assay.
  • Alternative to preparing monoclonal antibody-secreting hybridomas, a monoclonal RNAi pathway antibody can be identified and isolated by screening a recombinant combinatorial immunoglobulin library (e.g., an antibody phage display library) with an RNAi pathway protein to thereby isolate immunoglobulin library members that bind to the RNAi pathway protein. 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). Additionally, examples of methods and reagents particularly amenable for use in generating and screening antibody display library can be found in, for example, U.S. Pat. No. 5,223,409; PCT Publication No. WO 92/18619; PCT Publication No. WO 91/17271; PCT Publication No. WO 92/20791; PCT Publication No. WO 92/15679; PCT Publication No. WO 93/01288; PCT Publication No. WO 92/01047; PCT Publication No. WO 92/09690; PCT Publication No. WO 90/02809; Fuchs et al., 1991, Bio/Technology 9:1370-1372; Hay et al., 1992, Hum. Antibod. Hybridomas 3:81-85; Huse et al., 1989, Science 246:1275-1281; Griffiths et al., 1993, EMBO J. 12:725-734.
  • Techniques developed for the production of “chimeric antibodies” (Morrison et al., Proc. Natl. Acad. Sci. USA, 81:6851, 1984; Neuberger et al., Nature, 312:604, 1984; Takeda et al., Nature, 314:452, 1984) can be used to splice the genes from a mouse antibody molecule of appropriate antigen specificity together with genes from a human antibody molecule of appropriate biological activity. A chimeric antibody is a molecule in which different portions are derived from different animal species, such as those having a variable region derived from a murine mAb and a human immunoglobulin constant region.
  • Alternatively, techniques described for the production of single chain antibodies (U.S. Pat. No. 4,946,778; and U.S. Pat. Nos. 4,946,778 and 4,704,692) can be adapted to produce single chain antibodies against an RNAi pathway protein or polypeptide. Single chain antibodies are formed by linking the heavy and light chain fragments of the Fv region via an amino acid bridge, resulting in a single chain polypeptide.
  • Antibody fragments that recognize and bind to specific epitopes can be generated by known techniques. For example, such fragments can include but are not limited to F(ab′)2 fragments, which can be produced by pepsin digestion of the antibody molecule, and Fab fragments, which can be generated by reducing the disulfide bridges of F(ab′)2 fragments. Alternatively, Fab expression libraries can be constructed (Huse et al., Science, 246:1275, 1989) to allow rapid and easy identification of monoclonal Fab fragments with the desired specificity.
  • Identification of RNAi Pathway Components
  • RNAi pathway components can be identified in C. elegans and other animals (e.g., a mammal) using the methods described in the Examples below. Pathway components can also be identified using methods known in the art and the information provided herein. Such components include those involved in protein:protein and protein:RNA interactions. Specifically, RDE-1 can be used to identify additional proteins and RNA molecules that bind to the RDE-1 protein and so facilitate genetic interference.
  • The RNAi pathway mutant strains described herein (e.g., rde-1, rde-2, rde-3, rde-4, and rde-5; also mut-2 and mut-7) can be used in genetic screens to identify additional RNAi pathway components. For example, a strain deficient for rde-1 activity can be mutagenized and screened for the recovery of genetic interference. This type of screen can identify allele-specific suppressors in other genes or second site mutations within the rde-1 gene that restore its activity. The resulting strains may define new genes that activate RNAi to overcome or bypass the rde-1 defect. The mutations identified by these methods can be used to identify their corresponding gene sequences.
  • Two-hybrid screens can also be used to identify proteins that bind to RNAi pathway proteins such as RDE-1. Genes encoding proteins that interact with RDE-1 or human homologs of the C. elegans RDE-1, are identified using the two-hybrid method (Fields and Song, 1989, Nature 340:245-246; Chien et al., 1991, Proc. Natl. Acad. Sci. USA 88:9578-9582; Fields and Stemglanz, 1994, Trends Genet. 10:286-292; Bartel and Fields, 1995, Methods Enzymol. 254:241-263). DNA encoding the RDE-1 protein is cloned and expressed from plasmids harboring GAL4 or lexA DNA-binding domains and co-transformed into cells harboring lacZ and HIS3 reporter constructs along with libraries of cDNAs that have been cloned into plasmids harboring the GAL4 activation domain. Libraries used for such co-transformation include those made from C. elegans or a vertebrate embryonic cell.
  • Mechanisms of Action of RNAi Pathway Components
  • Specific cellular functions associated with the RNAi pathway include the specific targeting of a nucleic acid by a dsRNA, uptake of dsRNA, transport of dsRNA, amplification of the dsRNA signal, and genetic interference. The mechanism of interference may involve translation inhibition, or interference with RNA processing. In addition, direct effects on the corresponding gene may contribute to interference. These mechanisms can be identified investigated using the methods described herein and methods known in the art.
  • Methods of Screening for Molecules that Inhibit the RNAi Pathway
  • The following assays are designed to identify compounds that are effective inhibitors of the RNAi pathway. Such inhibitors may act by, but are not limited to, binding to an RDE-1 polypeptide (e.g., from C. elegans, mouse, or human), binding to intracellular proteins that bind to an RNAi pathway component, compounds that interfere with the interaction between RNAi pathway components including between an RNAI pathway component and a dsRNA, and compounds that modulate the activity or expression of an RNAi pathway gene such as rde-1. An inhibitor of the RNAi pathway can also be used to promote expression of a transgene.
  • Assays can also be used to identify molecules that bind to RNAi pathway gene regulatory sequences (e.g., promoter sequences), thus modulating gene expression. See, e.g., Platt, 1994, J. Biol. Chem. 269:28558-28562, incorporated herein by reference in its entirety.
  • The compounds which may be screened by the methods described herein include, but are not limited to, peptides and other organic compounds (e.g., peptidomimetics) that bind to an RNAi pathway protein (e.g., that bind to an RDE-1), or inhibit its activity in any way.
  • Such compounds may include, but are not limited to, peptides; for example, soluble peptides, including but not limited to members of random peptide libraries; (see, e.g., Lam et al., 1991, Nature 354:82-94; Houghten et al., 1991, Nature 354:84-86), and combinatorial chemistry-derived molecular libraries made of D-and/or L-amino acids, phosphopeptides (including, but not limited to, members of random or partially degenerate, directed phosphopeptide libraries; see e.g., Songyang et al., 1993, Cell 72:767-778), and small organic or inorganic molecules.
  • Organic molecules are screened to identify candidate molecules that affect expression of an RNAi pathway gene (e.g., rde-1), e.g., by interacting with the regulatory region or transcription factors of a gene. Compounds are also screened to identify those that affect the activity of such proteins, (e.g., by inhibiting rde-1 activity) or the activity of a molecule involved in the regulation of, for example, rde-1.
  • Computer modeling or searching technologies are used to identify compounds, or identify modifications of compounds that modulate the expression or activity of an RNAi pathway protein. For example, compounds likely to interact with the active site of a protein (e.g., RDE-1) are identified. The active site of an RNAi pathway protein can be identified using methods known in the art including, for example, analysis of the amino acid sequence of a molecule, from a study of complexes of an RNAi pathway, with its native ligand (e.g., a dsRNA). Chemical or X-ray crystallographic methods can be used to identify the active site of an RNAi pathway protein by the location of a bound ligand such as a dsRNA.
  • The three-dimensional structure of the active site is determined. This can be done using known methods, including X-ray crystallography which may be used to determine a complete molecular structure. Solid or liquid phase NMR can be used to determine certain intra-molecular distances. Other methods of structural analysis can be used to determine partial or complete geometrical structures. Geometric structure can be determined with an RNAi pathway protein bound to a natural or artificial ligand which may provide a more accurate active site structure determination.
  • Computer-based numerical modeling can also be used to predict protein structure (especially of the active site), or be used to complete an incomplete or insufficiently accurate structure. Modeling methods that may be used are, for example, parameterized models specific to particular biopolymers such as proteins or nucleic acids, molecular dynamics models based on computing molecular motions, statistical mechanics models based on thermal ensembles, or combined models. For most types of models, standard molecular force fields, representing the forces between constituent atoms and groups are necessary, and can be selected for the model from among the force fields known in physical chemistry. Information on incomplete or less accurate structures determined as above can be incorporated as constraints on the structures computed by these modeling methods.
  • Having determined the structure of the active site of an RNAi pathway protein (e.g., RDE-1), either experimentally, by modeling, or by a combination of methods, candidate modulating compounds can be identified by searching databases containing compounds along with information on their molecular structure. The compounds identified in such a search are those that have structures that match the active site structure, fit into the active site, or interact with groups defining the active site. The compounds identified by the search are potential RNAi pathway modulating compounds.
  • These methods may also be used to identify improved modulating compounds from an already known modulating compound or ligand. The structure of the known compound is modified and effects are determined using experimental and computer modeling methods as described above. The altered structure may be compared to the active site structure of an RNAi pathway protein (e.g., an RDE-1) to determine or predict how a particular modification to the ligand or modulating compound will affect its interaction with that protein. Systematic variations in composition, such as by varying side groups, can be evaluated to obtain modified modulating compounds or ligands of preferred specificity or activity.
  • Other experimental and computer modeling methods useful to identify modulating compounds based on identification of the active sites of an RNAi pathway protein and related transduction and transcription factors will be apparent to those of skill in the art.
  • Examples of molecular modeling systems are the QUANTA programs, e.g., CHARMm, MCSS/HOOK, and X-LIGAND, (Molecular Simulations, Inc., San Diego, Calif.). QUANTA analyzes the construction, graphic modeling, and analysis of molecular structure. CHARMm analyzes energy minimization and molecular dynamics functions. MCSS/HOOK characterizes the ability of an active site to bind a ligand using energetics calculated via CHARMm. X-LIGAND fits ligand molecules to electron density of protein-ligand complexes. It also allows interactive construction, modification, visualization, and analysis of the behavior of molecules with each other.
  • Articles reviewing computer modeling of compounds interacting with specific protein can provide additional guidance. For example, see Rotivinen et al., 1988, Acta Pharmaceutical Fennica 97:159-166; Ripka, New Scientist June 16, 1988 pp. 54-57; McKinaly and Rossmann, 1989, Ann. Rev. Pharmacol. Toxicol. 29:111-122; Perry and Davies. OSAR Quantitative Structure-Activity Relationships in Drug Design pp. 189-193 (Alan R. Liss, Inc., 1989); Lewis and Dean, 1989, Proc. R. Soc. Lond. 236:125-140, 141-152; and, regarding a model receptor for nucleic acid components, Askew et al., Am. J. Chem. Soc. 111: 1082-1090. Computer programs designed to screen and depict chemicals are available from companies such as MSI (supra), Allelix, Inc. (Mississauga, Ontario, Canada), and Hypercube, Inc. (Gainesville, Fla.).
  • These applications are largely designed for drugs specific to particular proteins; however, they can be adapted to the design of drugs specific to identified regions of DNA or RNA. Chemical libraries that can be used in the protocols described herein include those available, e.g., from ArQule, Inc. (Medford, Mass.) and Oncogene Science, Inc. (Uniondale, N.Y.).
  • In addition to designing and generating compounds that alter binding, as described above, libraries of known compounds, including natural products, synthetic chemicals, and biologically active materials including peptides, can be screened for compounds that are inhibitors or activators of the RNAi pathway components identified herein.
  • Compounds identified by methods described above can be used, for example, for elaborating the biological function of RNAi pathway gene products (e.g., an RDE-1), and to treat genetic disorders involving an RNAi pathway protein. Assays for testing the effectiveness of compounds such as those described herein are further described below.
  • In Vitro Screening Assays for Compounds that Bind to RNAi Pathway Proteins and Genes
  • In vitro systems can be used to identify compounds that interact with (e.g., bind to) RNAi pathway proteins or genes encoding those proteins (e.g., rde-1 and its protein product). Such compounds are useful, for example, for modulating the activity of these entities, elaborating their biochemistry, treating disorders in which a decrease or increase in dsRNA mediated genetic interference is desired. Such compounds may also be useful to treat diseases in animals, especially humans, involving nematodes, e.g., trichinosis, trichuriasis, and toxocariasis. Compounds such as those described herein may also be useful to treat plant diseases caused by nematodes. These compounds can be used in screens for compounds that disrupt normal function, or may themselves disrupt normal function.
  • Assays to identify compounds that bind to RNAi pathway proteins involve preparation of a reaction mixture of the protein and the test compound under conditions sufficient to allow the two components to interact and bind thus forming a complex which can be removed and/or detected.
  • Screening assays can be performed using a number of methods. For example, an RNAi pathway protein from an organism (e.g., RDE-1), peptide, or fusion protein can be immobilized onto a solid phase, reacted with the test compound, and complexes detected by direct or indirect labeling of the test compound. Alternatively, the test compound can be immobilized, reacted with the RNAi pathway molecule, and the complexes detected. Microtiter plates may be used as the solid phase and the immobilized component anchored by covalent or noncovalent interactions. Non-covalent attachment may be achieved by coating the solid phase with a solution containing the molecule and drying. Alternatively, an antibody, for example, one specific for an RNAi pathway protein such as RDE-1 is used to anchor the molecule to the solid surface. Such surfaces may be prepared in advance of use, and stored.
  • In these screening assays, the non-immobilized component is added to the coated surface containing the immobilized component under conditions sufficient to permit interaction between the two components. The unreacted components are then removed (e.g., by washing) under conditions such that any complexes formed will remain immobilized on the solid phase. The detection of the complexes may be accomplished by a number of methods known to those in the art. For example, the nonimmobilized component of the assay may be prelabeled with a radioactive or enzymatic entity and detected using appropriate means. If the non-immobilized entity was not prelabeled, an indirect method is used. For example, if the non-immobilized entity is an RDE-1, an antibody against the RDE-1 is used to detect the bound molecule, and a secondary, labeled antibody used to detect the entire complex.
  • Alternatively, a reaction can be conducted in a liquid phase, the reaction products separated from unreacted components, and complexes detected (e.g., using an immobilized antibody specific for an RNAi pathway protein).
  • Cell-based assays can be used to identify compounds that interact with RNAi pathway proteins. Cell lines that naturally express such proteins or have been genetically engineered to express such proteins (e.g., by transfection or transduction of an rde-1 DNA) can be used. For example, test compounds can be administered to cell cultures and the amount of mRNA derived from an RNAi pathway gene analyzed, e.g., by Northern analysis. An increase in the amount of RNA transcribed from such a gene compared to control cultures that did not contain the test compound indicates that the test compound is an inhibitor of the RNAi pathway. Similarly, the amount of a polypeptide encoded by an RNAi pathway gene, or the activity of such a polypeptide, can be analyzed in the presence and absence of a test compound. An increase in the amount or activity of the polypeptide indicates that the test compound is an inhibitor of the RNAi pathway.
  • Ectopic Expression of an RNAi Pathway Gene
  • Ectopic expression (i.e., expression of an RNAi pathway gene in a cell where it is not normally expressed or at a time when it is not normally expressed) of a mutant RNAi pathway gene (i.e., an RNAi pathway gene that suppresses genetic interference) can be used to block or reduce endogenous interference in a host organism. This is useful, e.g., for enhancing transgene expression in those cases where the RNAi pathway is interfering with expression of a transgene. Another method of accomplishing this is to knockout or down regulate an RNAi pathway gene using methods known in the art. These methods are useful in both plants and animals (e.g., in an invertebrate such as a nematode, a mouse, or a human).
  • Ectopic expression of an RNAi pathway gene, e.g., rde-1 or rde-4 can also be used to activate the RNAi pathway. In some cases, targeting can be used to activate the pathway in specific cell types, e.g., tumor cells. For example, a non-viral RNAi pathway gene construct can be targeted in vivo to specific tissues or organs, e.g., the liver or muscle, in patients. Examples of delivery systems for targeting such constructs include receptor mediated endocytosis, liposome encapsulation (described below), or direct insertion of non-viral expression vectors.
  • An example of one such method is liposome encapsulation of nucleic acid. Successful in vivo gene transfer has been achieved with the injection of DNA, e.g., as a linear construct or a circular plasmid, encapsulated in liposomes (Ledley, Human Gene Therapy 6:1129-1144 (1995) and Farhood, et al., Ann. NY Acad. Sci. 716:23-35 (1994)). A number of cationic liposome amphiphiles are being developed (Ledley, Human Gene Therapy 6:1129-1144 (1995); Farhood, et al., Ann. NY Acad. Sci., 716:23-35 (1994) that can be used for this purpose.
  • Targeted gene transfer has been shown to occur using such methods. For example, intratracheal administration of cationic lipid-DNA complexes was shown to effect gene transfer and expression in the epithelial cells lining the bronchus (Brigham, et al., Am. J. Respir. Cell Mol. Biol. 8:209-213 (1993); and Canonico, et al., Am. J. Respir. Cell Mol. Biol. 10:24-29 (1994)). Expression in pulmonary tissues and the endothelium was reported after intravenous injection of the complexes (Brigham, et al., Am. J. Respir. Cell Mol. Biol. 8:209-213 (1993); Zhu, et al., Science, 261:209-211 (1993); Stewart, et al., Human Gene Therapy 3:267-275 (1992); Nabel, et al., Human Gene Therapy 3:649-656 (1992); and Canonico, et al., J. Appl. Physiol. 77:415-419 (1994)). An expression cassette for an RNAi pathway sequence in linear, plasmid or viral DNA forms can be condensed through ionic interactions with the cationic lipid to form a particulate complex for in vivo delivery (Stewart, et al., Human Gene Therapy 3:267-275 (1992)).
  • Other liposome formulations, for example, proteoliposomes which contain viral envelope receptor proteins, i.e., virosomes, have been found to effectively deliver genes into hepatocytes and kidney cells after direct injection (Nicolau, et al., Proc. Natl. Acad. Sci. USA 80:1068-1072 (1993); Kaneda, et al., Science 243:375-378 (1989); Mannino, et al., Biotechniques 6:682 (1988); and Tomita, et al., Biochem. Biophys. Res. Comm. 186:129-134 (1992)).
  • Direct injection can also be used to administer an RNAi pathway nucleic acid sequence in a DNA expression vectors, e.g., into the muscle or liver, either as a solution or as a calcium phosphate precipitate (Wolff, et al., Science 247:1465-1468 (1990); Ascadi, et al., The New Biologist 3:71-81 (1991); and Benvenisty, et al., Proc. Natl. Acad. Sci. USA 83:9551-9555 (1986).
  • Preparation of RNAi Agents
  • RNAi pathway components can be used to prepare RNAi agents. Such agents are dsRNAs that have been treated with RNAi pathway components rendering the treated dsRNA capable of activity in the RNAi pathway and can be used as sequence-specific interfering agents useful for targeted genetic interference. Specifically, treating a dsRNA with an RDE-1 and RDE-4 is useful for making an RNAi agent. An RNAi agent can be produced by preincubating a dsRNA in vitro in the presence of RDE-1 and RDE-4.
  • Another method of preparing an RNAi agent is to activate the RNAi pathway in a target cell (i.e., a cell in which it is desirable to activate the RNAi pathway such as a tumor cell) by transgenesis of an rde-1 coding sequence and an rde-4 coding sequence into the target cell.
  • RNAi pathway polypeptides can be modified, e.g., to enhance their stability or cellular uptake, by attaching lipophilic or other helper groups to the polypeptide, by the formation of chimeras with proteins or other moieties that are taken up by cells, or by the use of liposomes or other techniques of drug delivery known in the art.
  • In C. elegans, RNAi agents appear to spread from cell to cell, thus, active RNAi agents can diffuse or be actively transported from conditioned media or serum directly into target cells. Alternatively, RNAi agents can be injected into an organism or cell. They may also be incorporated into a cell using liposomes or other such methods known in the art.
  • Such methods are useful for stimulating the RNAi pathway in C. elegans cells, and in heterologous cells including plants and vertebrate cells. Such methods are useful in mammalian, e.g., human cells.
  • Enhanced Delivery of a Cargo Compound
  • RNAi pathway components that mediate the transport of dsRNA into cells and tissues can be used to promote the entry of dsRNA into cells and tissues, including dsRNA that is linked to another compound. The method is accomplished by linking dsRNA to a cargo compound (e.g., a drug or DNA molecule), e.g., by a covalent bond. The endogenous RNAi pathway gene expressing dsRNA transport function is activated using methods known in the art. Alternatively, other methods can be used such as transfecting the target cell with the gene that affects transport thus permitting the cell or tissue to take up the dsDNA.
  • EXAMPLES
  • The invention is further described in the Examples below which describe methods of identifying mutations in the RNAi pathway and methods of identifying genes encoding components of the RNAi pathway.
  • EXAMPLE 1 Strains and Alleles
  • The Bristol strain N2 was used as standard wild-type strain. The marker mutations and deficiencies used are listed by chromosomes as follows: LGI: dpy-14(e188), unc-13(e51); LGIII: dpy-17(e164), unc-32(e189); LGV: dpy-11(e224), unc-42(e270), daf-11(m87), eDf1, mDf3, nDf31, sDf29, sDf35, unc-76(e911). The C. elegans strain DP13 was used to generate hybrids for STS linkage-mapping (Williams et al., 1992, Genetics 131:609-624).
  • Sensitivity to RNAi was tested in the following strains. MT3126: mut-2(r459) (obtained from John Collins, Department of Biochemistry & Molecular Biology, University of New Hampshire, Durham, N. H.); dpy-19(n1347), TW410: mut-2([459) sem-4(n1378), NL917: mut-7(pk204), SS552: mes-2(bn76) rol-1(e91)/mnC1 (obtained from S. Strome, Biology Dept., Indiana University), SS449: mes-3(bn88) dpy-5(e61) (from S. Strome, supra); hDp20, SS268: dpy-11(e224) mes-4(bn23) unc-76(e91)/nT1, SS360: mes-6(bn66) dpy-20(e1282)/nT1, CB879: him-1 (e879). A non-Unc mut-6 strain used was derived from RW7096: mut-6(st702) unc-22(st192::Tc1), due to the loss of Tc1 insertion in unc-22.
  • Homozygous mutants of mut-6, mes-2, 3, 4, 6 and him-1 showed sensitivity to RNAi by injection of pos-1 dsRNA. The dose of injected RNA was about 0.7 mg/ml. This dose lies within the range where reduced concentration leads to reduced interference effects. The results of the injection of pos-1 dsRNA into these mutants (dead embryos/F1 progeny) were as follows: mut-6: 422/437, mes-2: 781/787, mes-3: 462/474, mes-4: 810/814, mes-6: 900/1,002, him-1: 241/248, N2 (control): 365/393.
  • To test mutator activity, a mutant that was caused by Tc4 transposon insertion was used; TR1175: unc-22(r765::Tc4). Strains TW410 and TR1175 were gifts from Q. Boese and J. Collins (Department of Biochemistry & Molecular Biology, University of New Hampshire, Durham, N.H.).
  • EXAMPLE 2 RNA Interference Assay
  • Genetic interference using RNAi administered by microinjection was performed as described in Fire et al., 1998, supra and Rocheleau et al., 1997, Cell 90:707-716. pos-1 cDNA clone yk61h1, par-2 cDNA clone yk96h7, sqt-3 cDNA clone yk75f2 were used to prepare dsRNA in vitro. These cDNA clones were obtained from the C. elegans cDNA project (Y. Kohara, Gene Network Lab, National Institute of Genetics, Mishima 411, Japan).
  • Genetic interference using RNAi administered by feeding was performed as described in Timmons and Fire, 1998, Nature 395:854. pos-1 cDNA was cloned into a plasmid that contains two T7 promoter sequences arranged in head-to-head configuration. The plasmid was transformed into an E. coli strain, BL21 (DE3), and the transformed bacteria were seeded on NGM (nematode growth medium) plates containing 60 μg/ml ampicillin and 80 μg/ml IPTG. The bacteria were grown overnight at room temperature to induce pos-1 dsRNA. Seeded plates (BL21(DE3)[dsRNA] plates) stored at 4° C. remained effective for inducing interference for up to two weeks. To test RNAi sensitivity, C. elegans larvae were transferred onto BL21 (DE3)[dsRNA] plates and embryonic lethality was assayed in the next generation.
  • Transgenic lines expressing interfering RNA for unc-22 were engineered using a mixture of three plasmids: pPD[L4218] (unc-22 antisense segment, driven by myo-3 promoter); pPD[L4218] (corresponding unc-22 sense segment, driven by myo-3 promoter); pRF4 (semidominant transformation marker). DNA concentrations in the injected mixture were 100 μg/ml each. Injections were as described (Mello et al., 1991, EMBO J. 10:3959; Mello and Fire, 1995, Methods in Cell Biol. 48:451-482).
  • EXAMPLE 3 Identification of RNAi-Deficient Mutants
  • A method of screening for mutants defective in the RNAi pathway was devised that would permit the large-scale application of dsRNA to mutagenized populations. Feeding worms E. coli which express a dsRNA, or simply soaking worms in dsRNA solution, are both sufficient to induce interference in C. elegans (Timmons and Fire, 1998, supra; Tabara et al., 1998, Science 282:430-431). To carry out a selection, the feeding method was optimized to deliver interfering RNA for an essential gene, pos-1. C. elegans hermaphrodites that ingest bacteria expressing dsRNA corresponding to a segment of pos-1 are themselves unaffected but produce dead embryos with the distinctive pos-1 embryonic lethal phenotype.
  • To identify strains defective in the RNAi pathway, wild-type animals were mutagenized, backcrossed, and the F2 generation examined for rare individuals that were able to produce complete broods of viable progeny. Chemical mutagenesis was used to generate the mutations as well as spontaneous mutations arising in the mut-6 strain in which Tc1 transposons are activated (Mori et al., 1988, Genetics 120:397-407). To facilitate screens for mutations, an egg laying starting strain was used. In the absence of egg laying, the F3 progeny remained trapped within the mother's cuticle. Candidate mutants had internally hatched broods of viable embryos and were thus easily distinguished from the background population of individuals filled primarily with dead embryos (FIG. 1A). Candidates were then re-tested for resistance to injected dsRNA.
  • The genetic screen used to isolate RNAi pathway mutants was similar to one designed by James R. Preiss for the identification of maternal effect mutants (Kemphues et al., 1988, Cell 52:311-320). An Eg1 strain, lin-2(e]309) was mutagenized with EMS and the F2 generation was cultured on a bacterial lawn expressing pos-1 dsRNA. Mutagenized populations were then screened for rare individuals that were able to produce complete broods of viable progeny forming a distinctive “bag of worms” phenotype. To make sure that the animals were truly resistant to RNAi, candidate strains were next assayed for resistance to RNAi by injection. Independent EMS induced alleles of rde-1 were found in two separate pools of mutagenized animals at a frequency of approximately one allele in 2,000 to 4,000 haploid genomes.
  • In addition, a search was made for spontaneous mutants using a mut-6 strain in which Tc1 transposons are activated (Mori et al., 1988). 100,000 mut-6; lin-2 animals (Mello et al., 1994) were cultured on bacteria expressing pos-1 dsRNA. After one generation of growth, surviving animals were transferred again to plates with bacteria expressing the dsRNA and screened for resistant mutants. Three resulting strains were genetically mapped. One of these strains (ne300) mapped to LGV and failed to complement rde-1(ne219). Two strains ne299 and ne301 mapped to LGIII and define the rde-4 complementation group. Because the screen was clonal in nature and involved rounds of enrichment it is possible that both rde-4 strains are related.
  • Seven mutant strains were selected for genetic mapping. These seven mutants defined four complementation groups; rde-1, with three alleles, rde-4, with two alleles, and rde-2, and rde-3, with one allele each (FIG. 1B).
  • To map the RNAi defective mutations, the RNAi resistant phenotype was assayed either by feeding bacteria expressing pos-1 dsRNA or by injection of a dsRNA mixture of pos-1 and unc-22. The same assays were used for complementation tests. In vivo expression of unc-22 dsRNA was also used for mapping of rde-1. Mapping with visible marker mutations was performed as described in Brenner (1974, Genetics, 77:71-94) and mapping with STS marker was performed as described in Williams et al. (1992, supra).
  • ne219, ne297 and ne300 failed to complement each other, defining the rde-1 locus. rde-1 mutations mapped near unc-42 V. Three factor mapping was used to locate rde-1 (ne300) one eighth of the distance from unc-42 in the unc-42/daf-11 interval (3/24 Unc-non-Daf recombinants analyzed). The rde-1 (ne300) allele complemented the chromosomal deficiency sDf29 and failed to complement eDf1, mDJ3, nDf31 and sDf35. rde-2(ne221) and rde-3(ne298) mapped near unc-13 I. rde-2 complemented rde-3. rde-4(ne299) and (ne301) mapped near unc-69 III and failed to complement each other. ne299 complemented mut-7(pk204).
  • The rde-1 (+) activity is sufficient maternally or zygotically. To test the maternal sufficiency, animals heterozygous for rde-1 (ne219) were injected with dsRNA targeting the zygotic gene, sqt-3, and self progeny were assayed for the Sqt phenotype. 100% of the self progeny including rde-1 homozygous progeny were found to exhibit the Sqt phenotype. Thus, maternally provided rde-1(+) activity is sufficient to mediate interference with a zygotic target gene. Zygotic sufficiency was assayed by injecting homozygous rde-1 mothers with dsRNA targeting the zygotic unc-22 gene (FIG. 3). Injected animals were allowed to produce self-progeny or instead were mated after 12 hours to wild-type males, to produce heterozygous rde/+cross-progeny. Each class of progeny was scored for the unc-22 twitching phenotype as indicated by the fraction shown if FIG. 3 (Unc progeny/total progeny). The injected animals were then mated with wild-type males. Self progeny from homozygous injected mothers were unaffected, however, 68% of the cross progeny were Unc. This result indicates that zygotically provided rde-1(+) activity is also sufficient. However both maternal and zygotic rde-1 (+) activity contribute to zygotic interference as 100% of progeny from wild-type injected mothers exhibit unc-22 interference (606/606). Thus, rde-1(+) and rde-4(+) activities are not needed for dsRNA uptake, transport or stability.
  • RNAi sensitivity of several existing C. elegans mutants was also examined. Most of these mutant strains were fully sensitive to RNAi. However, RNAi resistance was identified in two strains that had previously been shown to exhibit elevated levels of transposon mobilization (mutator strains): mut-2 (described in Collins et al., 1987, Nature 328:726-728) and mut-7 (described in Ketting et al., Cell, in press for release on Oct. 15, 1999). Another mutator strain, mut-6(st702), was fully sensitive to RNAi. Since mutator strains continually accumulate mutations, the resistance of mut-2 and mut-7 may have been due to the presence of secondary mutations. To test this possibility we examined the genetic linkage between the mutator and RNAi resistance phenotypes of mut-2 and mut-7. We found that independently outcrossed mut-2(r459) mutator strains TW410 and MT3126 both showed resistance to RNAi. We mapped the RNAi resistance phenotype of mut-7(pk204) to the center of linkage group III (FIG. 1B), the position that had been defined for the mutator activity of mut-7(pk204) by Ketting et al. (supra). Together, these observations indicate that the RNAi resistance phenotypes of the mut-2 and mut-7 strains are genetically linked to their mutator activities. Animals heterozygous for the rde and mut alleles were generated by crossing wild-type males with Unc-Rde or Unc-Mut hermaphrodites. The rde and mut mutations appeared to be simple recessive mutations with the exception of mut-2([459), which appeared to be weakly dominant (FIG. 2A).
  • These data demonstrate that some genes are non-essential (e.g., rde-1 and rde-4).
  • This method can be used to identify additional mutations in RNAi pathway genes.
  • EXAMPLE 4 Identification of Properties of RNAi-Deficient Mutants
  • Effects of rde Mutations in Germline and Somatic Tissues
  • Microinjection was used to assay the sensitivity of each rde strain to several distinct dsRNA species. The pos-1 and par-2 genes are expressed in the maternal germline and are required for proper embryonic development (Tabara et al., 1999, Development 126: 1-11; Boyd et al., 1996, Development 122:3075-3084). All rde-strains tested (as well as mut-2 and mut-7) showed significant resistance to dsRNA targeting of these germline-specific genes (FIG. 2B), as well as to several other germline specific genes tested. The rde-3 data (asterisk in FIG. 2B) includes a 10% non-specific embryonic lethality present in the rde-3 strain.
  • To examine the effect of these mutations on genetic interference of somatically expressed genes, cells were injected with dsRNA targeting the cuticle collagen gene sqt-3 and the body muscle structural gene unc-22. sqt-3 hypomorphic mutants exhibit a short, dumpy body shape (dpy; van der Keyl et al., 1994, Dev. Dyn. 201:86-94). unc-22 mutations exhibit severe paralysis with a distinctive body twitching phenotype (Moerman et al., 1986, Proc. Natl. Acad. Sci. USA 83:2579-2583). rde-1, rde-3, rde-4 and mut-2 strains showed strong resistance to both sqt-3 and unc-22 dsRNA, while rde-2 and mut-7 strains showed partial resistance. Thus rde-2 and mut-7 appeared to be partially tissue- or gene-specific in that they were required for effective RNAi against germline but not somatically expressed genes. The rde-1, rde-3, rde-4, and mut-2 (+) activities appeared to be required for interference for all genes analyzed. The rde and mut strains differ from one another in sensitivity to sqt-2 dsRNA.
  • Effect of rde on Transposon Mobilization
  • The effect of rde mutations on transposon mobilization was examined. Two of the newly identified mutants, rde-2 and rde-3 exhibited a level of transposon activation similar to that of mut-7 (Table 1). In contrast, transposon mobilization was not observed in the presence of rde-1 or rde-4 (Table 1).
    TABLE 1
    TRANSPOSON MOBILIZATION AND MALE
    INCIDENCE IN rde AND mut STRAINS
    Percentage of Non-Unc Revertants
    unc-22 (r765::Tc4) 0 (0/2000)
    rde-1 (ne219); 0 (0/4000)
    unc-22 (r765::Tc4)
    rde-2 (ne221; 0.96 (8/830)
    unc-22 (r765::Tc4)
    rde-3 (ne298); 1.6 (35/2141)
    unc-22 (r765::Tc4)
    rde-4 (ne299); 0 (0/2885)
    unc-22 (r765::Tc4)
    mut-7 (pk204); 1.0 (40/3895)
    unc-22 (r765::Tc4)
    Percentage of Male Animals
    Wild type (n2) 0.21 (2/934)
    rde-1 (ne219) 0.07 (1/1530)
    rde-2 (ne221) 3.2 (25/788)
    rde-3 (ne298) 7.8 (71/912)
    rde-4 (ne299) 0.24 (5/2055)
  • X-Chromosome Loss
  • Mutator strains (including mut-2, mut-7) rde-2 and rde-3) exhibit a second phenotype: a high incidence of males reflecting an increased frequency of X-chromosome loss during meiosis (Collins et al., 1987, supra; Ketting et al., supra). This phenotype was observed in rde-2 and rde-3 strains, but not observed in the rde-1 and rde-4 strains which showed a wild-type incidence of males (Table 1).
  • A previously described gene-silencing process appears to act on transgenes in the germline of C. elegans. Although the silencing mechanisms are not well understood, they are known to depend on the products of the genes mes-2, 3, 4 and 6 (Kelly and Fire, 1998, Development 125:2451-2456). To examine the possibility that the RNAi and germline transgene-silencing might share common mechanistic features, we first asked if the mes mutants were resistant to RNAi. We found normal levels of RNA interference in each of these strains. We next asked if RNAi deficient strains were defective in transgene-silencing. Three strains were analyzed: mut-7(pk204), rde-1(ne219) and rde-2(ne221).
  • To analyze transgene silencing in mut-7 worms, homozygous mut-7 lines carrying various GFP reporters transgenes were generated as follows: N2 (Bristol strain) males were mated to mut-7 (pk204) unc-32 (e]89) hermaphrodites; cross progeny males were then mated to strains carrying the GFP transgenes. mut-7 unc-32/++ cross progeny from these matings were cloned, and mut-7 unc-32 homozygous animals carrying the transgenes were isolated from their self-progeny. After the GFP reporter transgenes were introduced into different genetic backgrounds, activation of GFP transgene expression in germ cells was assayed at 25□C by fluorescence microscopy. The tested GFP reporter transgenes were each active in some or all somatic tissues, but had become silenced in the germline. The plasmids used and transgene designations are as follows: 1) pBK48 which contains an in-frame insertion of GFP into a ubiquitously expressed gene, let-858 (Kelly, et al., 1997, Genetics 146:227-238). ccExPD7271 contains more than 100 copies of pBK48 in a high copy repetitive array that is carried extrachromosomally. 2) pJH3.92 is an in-frame fusion of GFP with the maternal pie-1 gene (M. Dunn and G. Seydoux, Johns Hopkins University, Baltimore, Md.). jhEx1070 carries pJH3.92 in a low copy “complex” extrachromosomal array generated by the procedure of Kelly et al. (1997, supra) pJKL380.4 is a fusion of GFP with the C. elegans nuclear laminin gene, lam-1, which is expressed in all tissues (J. Liu and A. Fire). ccIn4810 carries pJKL380.4 in a complex array that has been integrated into the X chromosome by gamma irradiation using standard techniques.
  • The mut-7 strain was analyzed most extensively and was found to exhibit desilencing of three different germline transgenes tested (Table 2). The rde-2 strain exhibited a similar level of desilencing for a single transgene. In contrast, no transgene desilencing was observed in rde-1 mutants (Table 2). Thus, mut-7 and rde-2 which differ from rde-1 in having transposon mobilization and a high incidence of X-chromosome loss also differ from rde-1 in their ability to partially reactivate silent germline transgenes.
    TABLE 2
    REACTIVATION OF SILENCED TRANSGENES
    IN THE GERMLINE OF mut-7(pk204)
    Percentage of
    Germline
    Genotype Transgene Array Desilencing
    +/+ ccEx7271 8.3 (4/48)
    mut-71+ ccEx7271 14.5 (7/48)
    mut-7/mut-7 ccEx7271 91.0 (71/78)
    +/+ jhEx1070 3.9 (2/51)
    mut-7/mut-7 jhEx1070 86.5 (32/37)
    +/+ ccin4810 4.3 (2/46)
    mut-7/mut-7 ccin4810 73.3 (33/45)
    rde-1/rde-1 ccEx7271 0 (0/34)
  • EXAMPLE 5 Requirement for rde-1(+) and rde-4(+) Activities in Target Tissue
  • The rde-1 and rde-4 mutants differ from other RNAi deficient strains identified herein in that they do not cause transposon mobilization nor do they cause chromosome loss. The role of these genes in upstream events such as dsRNA uptake, transport or stability was examined. Such events could be required for interference induced by exogenous trigger RNAs but might be dispensable for natural functions of RNAi. To evaluate these upstream events, rde-1 and rde-4 homozygotes were exposed to dsRNA. The next generation was scored for interference. dsRNA targeting the unc-22 gene was injected into the intestinal cells of homozygous rde-1 and rde-4 hermaphrodites and the injected animals were then mated to wild-type males (FIG. 3). The self-progeny for both strains exhibited no interference with the targeted gene. However, there was potent interference in the rde-1/+ and rde-4/+ cross progeny (FIG. 3). These observations indicated that rde-1 and rde-4 mutants have intact mechanisms for transporting the interference effect from the site of injection (the intestine) into the embryos of the injected animal and then into the tissues of the resulting progeny. The stability of the resulting interference also appeared to be normal in rde-1 and rde-4 as the homozygous injected mothers continued to produce affected cross progeny for several days after the time of injection.
  • To examine whether rde-1 and rde-4 mutants could block interference caused by dsRNA expressed directly in the target tissue, the muscle-specific promoter from the myo-3 gene (Dibb et al., 1989, J. Mol. Biol. 205:605-613) was used to drive the expression of both strands of the muscle structural gene unc-22 in the body wall muscles (Moerman et al., 1986, supra; Fire et al., 1991, Development 113:503-514). A mixture of three plasmids was injected: [myo-3 promoter::unc-22 antisense], [myo-3::unc-22 sense], and a marker plasmid (pRF4[rol-6(sul 006gf)] [Mello et al., 1991]). Frequencies of Unc transgenic animals were followed in F1 and F2 generations. The aUnc phenotype was weak. Wild-type animals bearing this transgene exhibit a strong twitching phenotype consistent with unc-22 interference. The twitching phenotype was strongly suppressed by both rde-1 and rde-4 mutants (Table 3). The mut-7 and rde-2 mutants which are both sensitive to unc-22(RNAi) by microinjection were also sensitive to promoter driven unc-22 interference in the muscle (Table 3). Taken together these findings suggest that rde-1(+) and rde-4(+) activities are not necessary for uptake or stability of the interfering RNA and may function directly in the target tissue.
    TABLE 3
    SENSITIVITY OF rde AND mut STRAINS TO
    TRANSGENE-DRIVEN INTERFERING RNA
    Unc Animals in Unc F2 Lines in
    Transgenic F1 Inherited Lines
    Wild type (N2) 26/59 10/11
    rde-1 (ne219)  0/25 0/3
    rde-2 (ne221) 35/72 14/14
    rde-3 (ne298) 1a/38 1a/9  
    rde-4 (ne299)  0/51 0/4
    mut-7 (pk204)  9/13 3/3
  • EXAMPLE 6 Molecular Identification of the rde-1 Gene
  • The rde-1 gene was cloned using standard genetic mapping to define a physical genetic interval likely to contain the gene using YACs and cosmids that rescue rde-1 mutants. These were used to identify a cloned rde-1 cDNA sequence and a cloned rde-4 sequence. These methods can also be used to identify the genes for rde-2, rde-3, and rde-5 using the mutant strains provided herein.
  • To clone an rde-1 gene, yeast artificial chromosome clones (YACs) containing C. elegans DNA from this interval were used to rescue the rde-1 mutant phenotype. To facilitate this analysis candidate rescuing YACs were co-injected with plasmids designed to express unc-22(RNAi). YAC and cosmid clones that mapped near the rde-1 locus were obtained from A. Coulson. rde-1 (ne219) was rescued by YAC clones: Y97C12 and Y50B5. The two overlapping YAC clones provided rde-1 rescuing activity as indicated by unc-22 genetic interference with characteristic body paralysis and twitching in the F 1 and F2 transgenic animals. In contrast a non-overlapping YAC clone failed to rescue resulting in 100% non-twitching transgenic strains (FIG. 4A).
  • The rescuing activity was further localized to two overlapping cosmid clones, cosmid C27H6 and T10A5, and finally to a single 4.5 kb genomic PCR fragment predicted to contain a single gene, designated K08H10.7 (SEQ ID NO:1; FIGS. 5A-5C) The K08H10.7 PCR product gave strong rescue when amplified from wild-type genomic DNA. This rescue was greatly diminished using a PCR fragment amplified from any of the three rde-1 alleles and was abolished by a 4 bp insertion at a unique NheI site in the rde-1 coding region. A wild-type PCR product from an adjacent gene C27H6.4, also failed to rescue.
  • The K08H10.7 gene from each of the rde-1 mutant strains was sequenced, and distinct point mutations were identified that are predicted to alter coding sequences in K08H10.7 (FIG. 4A). Based on these findings rde-1 can be identified as the K08H10.7 gene.
  • A full-length cDNA sequence was determined for rde-1 using the cDNA clones, yk296b10 and yk595h5. cDNA clones for rde-1 were obtained from Y. Kohara (Gene Network Lab, National Institute of Genetics, Mishima 411, Japan). The cDNA sequence of coding region and 3'UTR was determined on yk296b10 except that the sequence of 5′UTR was determined on yk595h5. The GenBank accession number for rde-1 cDNA is AF180730 (SEQ ID NO:2). The rde-1 cDNA sequence was used to generate a predicted translation product (SEQ ID NO:3), referred to as RDE-1, consisting of 1020 amino acids. The RDE-1 sequence was used to query Genbank and identify numerous related genes in C. elegans as well as other animals and plants. This gene family includes at least 23 predicted C. elegans genes, several of which appear to be members of conserved subfamilies. Within subfamilies, conservation extends throughout the protein and all family members have a carboxy-terminal region that is highly conserved (FIG. 4B). Besides the genes shown in FIG. 4B, other related genes include ARGONAUTE 1 (Arabidopsis), SPCC736.11 (S. pombe), and Piwi (Drosophila). A portion of the N terminal region of RDE-1 showed no significant similarity to any of the identified related genes. There are no defined functional motifs within this gene family, but members including RDE-1 are predicted to be cytoplasmic or nuclear by PSORT analysis (Nakai and Horton, 1999, Trends Biochem. Sci. 24:34-36). Furthermore, one family member named eIF2C has been identified as a component of a cytoplasmic protein fraction isolated from rabbit reticulocyte lysates. The RDE-1 protein is most similar to the rabbit eIF2C. However, two other C. elegans family members are far more similar to eIF2C than is RDE-1 (FIG. 4B). RDE-1 may provide sequence-specific inhibition of translation initiation in response to dsRNA.
  • The rde-1 mutations appear likely to reduce or eliminate rde-1(+) activity. Two rde-1 alleles ne219 and ne297 are predicted to cause amino acid substitutions within the RDE-1 protein and were identified at a frequency similar to that expected for simple loss-of-function mutations. The rde-1(ne219) lesion alters a conserved glutamate to a lysine (FIG. 4B). The rde-1(ne297) lesion changes a non-conserved glycine, located four residues from the end of the protein, to a glutamate (FIG. 4B). The third allele, ne300, contains the strongest molecular lesion and is predicted to cause a premature stop codon prior to the most highly conserved region within the protein (Q>Ochre in FIG. 4B). Consistent with the idea that rde-1 (ne300) is a strong loss of function mutation, we found that when placed in trans to a chromosomal deficiency the resulting deficiency trans-heterozyotes were RNAi deficient but showed no additional phenotypes. These observations suggest that rde-1 alleles are simple loss-of-function mutations affecting a gene required for RNAi but that is otherwise non-essential.
  • Because of its upstream role RNA interference (see Examples 8-10 below), the RDE-1 protein and fragments thereof can be used to prepare dsRNA that is useful as an RNAi agent.
  • EXAMPLE 7 Maternal Establishment and Paternal Transmission of RNAi
  • To examine whether the interference effect induced by RNAi exhibited linkage to the target gene (e.g., was involved in a reversible alteration of the gene or associated chromatin), a strain was constructed such that the F1 males that carry the RNAi effect also bear a chromosomal deletion that removes the target gene (FIG. 7B). In the case of linkage to the target gene, the RNAi effect would be transmitted as a dominant factor.
  • In experiments testing the linkage of the interference effect to the target gene, three different species of dsRNA (pos-1 dsRNA, mom-2 dsRNA, or sgg-1 dsRNA) were delivered into C. elegans in independent experiments. The dsRNA was delivered by injection through a needle inserted into the intestine. In general, dsRNA was synthesized in vitro using T3 and T7 polymerases. Template DNA was removed from the RNA samples by DNase treatment (30 minutes at 37° C.). Equal amounts of sense and antisense RNAs were then mixed and annealed to obtain dsRNA. dsRNA at a concentration of 1-5 mg/ml was injected into the intestine of animals. In control experiments, mixtures of linearized template DNA plasmids used for synthesizing RNA failed to induce interference in P0, F1, or F2 animals when injected into the intestine of hermaphrodites at a concentration of 0.2 mg/ml. FIG. 7A illustrates this experiment. The gonad of the parent (P0) hermaphrodite has symmetrical anterior and posterior U-shaped arms as shown in FIG. 7A. Several fertilized eggs are shown in FIG. 7A, centrally located in the uterus. The rectangular mature oocytes are cued up in the gonad arms most proximal to the uterus. The embryos present in P0 at the time of injection gave rise to unaffected F 1 progeny. Oocytes in the proximal arms of the injected P0 gonad inherit the RNAi effect but also carry a functional maternal mRNA (F 1 carriers of RNAi).
  • After a clearance period during which carrier and unaffected F1 progeny are produced, the injected P0 begins to exclusively produce dead F1 embryos with the phenotype corresponding to the inactivation of the gene targeted by the injected RNA (Tabara et al. 1999, Development 126:1; C. Rocheleau, 1997, Cell 90:707). Potential F1 and F2 carriers of the interference effect were identified within the brood of the injected animal. In the case of hermaphrodites, carriers were defined as “affected” if the animals produced at least 20% dead embryos with phenotypes corresponding to maternal loss of function for the targeted locus. In the case of males, carriers were defined as animals whose cross progeny included at least one affected F2 hermaphrodite. The total number of carriers identified in each generation for each of three dsRNAs injected is shown in FIG. 7A as a fraction of the total number of animals assayed.
  • To examine the extragenic inheritance of RNAi, experiments were carried out investigating whether sperm that inherit the deletion and therefore have no copies of the target locus could carry the interference effect into the F2 generation. F 1 males that carried both pos-1 (RNAi) and a chromosomal deficiency for the pos-1 locus were generated. The chromosome carrying the deficiency for pos-1 also carried a deficiency for phenotypically uncoordinated (unc). F2 progeny of the carrier male includes two genotypes: phenotypically wild-type animals that inherit the (+) chromosome, and phenotypically uncoordinated (Unc) progeny that inherit the mDf3 chromosome. In these experiments, the deficiency bearing sperm were just as capable as wild-type sperm of transferring interference to the F2 hermaphrodite progeny (FIG. 7B). Thus, the target locus was not needed for inheritance of the interference effect.
  • Surprisingly, although males were sensitive to RNAi and could inherit and transmit RNAi acquired from their mothers, direct injections into males failed to cause transmission of RNAi to the F1 for several genes tested. In an example of this type of protocol, wild type males were injected with targeting dsRNA: body muscle structural gene unc-22, cuticle collagen gene sqt-3, maternal genes pos-1 and sgg-1. Males of the pes-10::gfp strain (Seydoux, G. and Dunn, Mass., 1997, Development 124:2191-2201 were injected with gfp dsRNA. Injected males were affected by unc-22 and gfp dsRNA to the same extent as injected hermaphrodites. No RNAi interference was detected in F1 progeny or injected males (40 to 200 F1 animals scored for each RNA tested. Therefore, the initial transmission of RNAi to F1 progeny may involve a mechanism active only in hermaphrodites while subsequent transmission to the F2 progeny appears to involve a distinct mechanism, active in both hermaphrodites and males. The hermaphrodite-specific step may indicate the existence of a maternal germline process that amplifies the RNAi agent. These data show that extracts from the maternal germline tissues of C. elegans may be used in conjunction with RDE-1 and RDE-4 activity to create and to then amplify RNAi agents.
  • In addition, the germline factors that amplify the RNAi agents can be identified by mutations that result in an RNAi deficient mutant phenotype. Such factors can be used as additional components of an in vitro system for the efficient amplification of RNAi agents.
  • EXAMPLE 8 Sufficiency of Wild-Type Activities of rde-1, rde-2, mut-7, and rde-4 in Injected Animals for Interference Among F1 Self Progeny
  • To investigate whether the activities of rde-1, rde-2, rde-4, and mut-7, respectively, are sufficient in injected hermaphrodites for interference in the F1 and F2 generations, crosses were designed such that wild-type activities of these genes would be present in the injected animal but absent in the F1 or F2 generations. To examine inheritance in the F1 generation, (hermaphrodite) mothers heterozygous for each mutant (P0) were injected, allowed to produce self-progeny (F1) and the homozygous mutant progeny in the F1 generation were examined for genetic interference (FIG. 8A). To do this, the heterozygous hermaphrodites from each genotype class, rde-1, unc-42/+; rde-2, unc-13/+; mut-7, dpy-71/+; and rde-4, unc-69/+ (the following alleles were used in this study: rde-1(ne300) unc-42, rde-1(ne219), rde-2(ne221), rde-4(ne299), and mut-7(pk2040) were injected with pos-1 dsRNA. In each case, two types of F1 self progeny, distinguished by the presence of the linked marker mutations, were scored for interference (FIG. 8A). In these experiments the rde-1 and rde-4 mutant F1 progeny exhibited robust interference, comparable to wild-type, while the rde-2 and mut-7 F1 progeny failed to do so. In control experiments, homozygous F1 progeny from heterozygous (uninjected) mothers were directly injected with pos-1 dsRNA (FIG. 8B). Injection of dsRNA directly into the rde-1 and rde-4 mutant progeny of uninjected heterozygous mothers failed to result in interference. Thus, injection of dsRNA into heterozygous hermaphrodites resulted in an inherited interference effect that triggered gene silencing in otherwise RNAi resistant rde-1 and rde-4 mutant F1 progeny while rde-2 and mut-7 mutant F1 progeny remained resistant.
  • In this experiment, the expression of rde-1(+) and rde-4(+) in the injected animal was sufficient for interference in later generations.
  • These data suggest that treatment of a dsRNA with functional rde-1 and rde-4 gene products can produce an agent that activates the remainder of the RNAi pathway.
  • EXAMPLE 9 Requirements for rde-1, rde-2, rde-4, and mut-7 in F1 and F2 interference
  • To examine the genetic requirements for RNAi genes in the F2 generation, F 1 male progeny were generated that carry the interference effect as well as one mutant copy of each respective locus; rde-1, rde-2, and mut-7 (FIG. 9A). Each of these males was then backcrossed with uninjected hermaphrodites homozygous for each corresponding mutant (FIG. 9A). The resulting cross progeny (F1) included 50% heterozygotes and 50% homozygotes that were distinguished by the presence of the linked marker mutations. The heterozygous siblings served as controls and in each case exhibited interference at a frequency similar to that seen in wild-type animals (FIG. 9A). In these crosses, rde-2 and mut-7 homozygous F2 progeny failed to exhibit interference, indicating that the activities of these two genes are required for interference in the F2 generation. In contrast, we found that homozygous rde-1 F2 animals exhibited wild-type levels of F2 interference (FIG. 9A). Control rde-1 homozygotes generated through identical crosses were completely resistant to pos-1::RNAi when challenged de novo with dsRNA in the F2 generation. In these experiments, 35 rde-1 homozygous animals generated through crosses shown in FIG. 9A were tested by feeding bacteria expressing pos-1 dsRNA, and 21 similar animals were tested by direct injections of pos-1 dsRNA. All animals tested were resistant to pos-1 (RNAi). Thus, rde-1 activity in the preceding generations was sufficient to allow interference to occur in rde-1 mutant F2 animals while the wild-type activities of rde-2 and mut-7 were required directly in the F2 animals for interference.
  • In this experiment, the expression of rde-1(+) and rde-4(+) in the injected animal was sufficient for interference in later generations. The wild-type activities of the rde-2 and mut-7 genes were required for interference in all generations asayed. Thus, rde-2 and mut-7 might be required only downstream or might also function along with rde-1 and rde-4.
  • These data lend additional support to the concept that an appropriately treated dsRNA could be used as an RNAi agent.
  • EXAMPLE 10 Sufficiency of rde-1 Activity to Initiate RNA Interference in Injected Animals That Lack the Wild-Type Activities of rde-2, mut-7, or rde-4
  • To ask if rde-2 and mut-7 activities function along with or downstream of rde-1, genetic cross experiments were designed in which the activities of these genes were present sequentially (FIG. 9B). For example, rde-1(+);rde-2(−) animals were injected with pos-1 dsRNA and then crossed to generate F1 hermaphrodites homozygous for rde-1(−); rde-2(+). In these experiments rde-1(+) activity in the injected animals was sufficient for F1 interference even when the injected animals were homozygous for rde-2 or mut-7 mutations (FIG. 10B). In contrast, rde-1(+) activity in the injected animals was not sufficient when the injected animals were homozygous for rde-4 mutant (FIG. 10B). Thus, rde-1 can act independently of rde-2 and mut-7 in the injected animal, but rde-1 and rde-4 must function together. These findings are consistent with the model that rde-1 and rde-4 function in the formation of the inherited interfering agent (i.e., an RNAi agent) while rde-2 and mut-7 function at a later step necessary for interference.
  • In summary, the above Examples provide genetic evidence for the formation and transmission of extragenic interfering agents in the C. elegans germline. Two C elegans genes, rde-1, and rde-4, appear to be necessary for the formation of these extragenic agents but not for interference mediated by them. In contrast, the activities of two other genes, rde-2 and mut-7, are required only downstream for interference.
  • These examples provide evidence that the rde-1 and rde-4 gene products or their homologs (e.g., from a mammal) can be used to prepare agents effective in activating the RNAi pathway.
  • EXAMPLE 11 rde-4 Sequences
  • An rde-4 gene was cloned using methods similar to those described in Example 6. The nucleic acid sequence (SEQ ID NO:4) and predicted amino acid sequence (SEQ ID NO:5) are illustrated in FIG. 10.
  • Analysis of the rde-4 nucleic acid sequence shows that it encodes a protein (RDE-4) with similarities to dsRNA binding proteins. Examples of the homology to X1 RBPA (SEQ ID NO:6; Swissprot: locus TRBP_XENLA, accession Q91836; Eckmann and Jantsch, 1997, J. Cell Biol. 138:239-253) and HSPKR (SEQ ID NO:7; AAF13156.1; Xu and Williams, 1998, J. Interferon Cytokine Res. 18:609-616), and a consensus sequence (SEQ ID NO:8) are shown in FIG. 11. Three regions have been identified within the predicted RDE-4 protein corresponding to conserved regions found in all members of this dsRNA binding domain family. These regions appear to be important for proper folding of the dsRNA binding domain. Conserved amino acid residues, important for interactions with the backbone of the dsRNA helix, are found in all members of the protein family including RDE-4 (see consensus residues in FIG. 11). This motif is thought to provide for general non-sequence-specific interactions with dsRNA. The RDE-4 protein contains conserved protein folds that are thought to be important for the assembly of the dsRNA binding domain in this family of proteins. Conserved amino acid residues in RDE-4 are identical to those that form contacts with the dsRNA in the crystal structure of the X1 RBP dsRNA complex. These findings strongly suggest that RDE-4 is likely to have dsRNA binding activity.
  • Because RDE-4 contains a motif that is likely to bind in a general fashion to any dsRNA and because RDE-4 appears to function upstream in the generation of RNAi agents, the RDE-4 protein or fragments thereof can be used to convert any dsRNA into an RNAi agent. In addition to the dsRNA binding domain, RDE-4 contains other functional domains that may mediate the formation of RNAi agents. These domains may provide for interaction between RDE-4 and RDE-1 or for binding to enzymes such as nucleases that convert the dsRNA into the RNAi agent. Because of its RNA binding function in RNA interference, the RDE-4 protein and fragments thereof can be used to prepare dsRNA that is useful in preparing an RNAi agent.
  • EXAMPLE 12 Identification of Regions of RDE-1 and RDE-4 that are Required for Creating an RNAi Agent
  • In vivo and in vitro assays are used to identify regions in RDE-1 and RDE-4 that are important for the generation of RNAi agents. In the in vivo assay, rde-1 and rde-4 are introduced into the corresponding C. elegans mutant strains via transgenes (Tabara et al., Cell 99:123 (1999); and Example 13). Important functional domains in RDE-1 and RDE-4 are defined by systematically altering the proteins followed by reintroduction into mutant animals to test for rescue of the RNAi deficient phenotype. A series of nested deletions are analyzed for rescue activity for both rde-1 and rde-4. Specific point mutations are used to analyze the importance of specific amino acids. Chimera's are produced between RDE proteins and related proteins and genes. For example, coding sequences from RDE-1 homologs from the worm or from human are tested for their ability to rescue rde-1 mutants. Replacing the RDE-4 dsRNA binding motif with a distinct RNA binding motif, e.g., one that recognizes a specific viral dsRNA sequence or a ssRNA sequence will alter the specificity of the RNAi response perhaps causing sequence-specific or ssRNA-induced gene targeting. In one form of the in vitro assay, whole protein extracts from rde-1 or rde-4 deficient worm strains are used.
  • Recombinant RDE-1 or RDE-4 is then added back to reconstitute the extract. Altered RDE-1 and RDE-4 proteins (described above, including deletions, point mutants and chimeras) are made in vitro and then tested for their ability to function when added back to these extracts. RNAi activity is analyzed by injecting the reconstituted extracts directly into animals or by assaying for the destruction of an added in vitro synthesized target mRNA.
  • EXAMPLE 13 Rescue of rde-4 Animals
  • Rescue of animals (e.g., C. elegans) that are mutant for an RNAi pathway is a useful method for identifying sequences from RNAi pathway genes that encode functional polypeptides, e.g., polypeptides that can eliminate the mutant phenotype.
  • An example of such a method for identifying rde-4 mutant animals is as follows. PCR using primers located 1 kb upstream and 500 nucleotides downstream of the open reading frame (T20G5.11; illustrated in FIG. 12) are used to amplify the rde-4 gene from C. elegans genomic DNA. The resulting PCR product is then injected along with reporter constructs described in Tabara et al. (Cell 99:123 (1999); incorporated herein in its entirety by reference), and the progeny of the injected animal are assayed for rescue of the RNAi deficient phenotype. The PCR product can also be cloned into a plasmid vector for site directed mutational analysis of RDE-4 (see Example 12). Co-injection of such a wild type RDE-4 plasmid and altered derivatives can be used to identify functional domains of rde-4. Similar methods can be used to identify functional domains of rde-1 and other RNAi pathway components.
  • Other Embodiments
  • It is to be understood that while the invention has been described in conjunction with the detailed description thereof, the foregoing description is intended to illustrate and not limit the scope of the invention, which is defined by the scope of the appended claims. Other aspects, advantages, and modifications are within the scope of the following claims.

Claims (17)

1-10. (canceled)
11. A substantially pure polypeptide encoded by a nucleic acid molecule which hybridizes under high stringency conditions to a complement of the nucleic acid molecule set forth in SEQ ID NO: 4
12-16. (canceled)
17. The polypeptide of claim 11, which hybridizes under conditions of hybridization at 68° C. in 5×SSC/5× Denhardt solution/1.0% SDS, followed by washing in 0.2×SSC/0.1% SDS at room temperature.
18. A substantially pure polypeptide encoded by a nucleic acid molecule having at least 80% sequence identity with the nucleic acid molecule set forth as SEQ ID No:4.
19. The polypeptide of claim 18, which is encoded by a nucleic acid molecule having at least 95% identity with the nucleic acid molecule set forth as SEQ ID NO:4.
20. The polypeptide of claim 18, which is encoded by a nucleic acid molecule having at least 98% identity with the nucleic acid molecule set forth as SEQ ID NO:4.
21. The polypeptide of claim 11 or 18, which polypeptide has a dsRNA binding activity.
22. The polypeptide of claim 11 or 18, which polypeptide has the dsRNA binding motif of SEQ ID NOS:8.
23. A substantially pure polypeptide fragment comprising at least 30 contiguous amino acids of SEQ ID NO:5.
24. A substantially pure protein encoded by the nucleic acid molecule set forth in SEQ ID NO:4.
25. A substantially pure protein comprising the amino acid sequence of SEQ ID NO:5.
26. A fragment of claim 24, wherein said fragment comprises amino acids 150 to 212 of SEQ ID NO:5.
27. A substantially pure RDE-4 polypeptide encoded by a nucleic acid molecule which can complement a RDE-4 mutation.
28. A fusion protein comprising a fragment of the polypeptide of any one of the preceding claims and a heterologous polypeptide.
29. The fusion protein of claim 28, wherein the heterologous polypeptide is selected from the group consisting of an immunoglobulin Fc (IgFc) polypeptide, a lacZ polypeptide, a glutathione S-transferase (GST) polypeptide, a six histidine tag polypeptide and a signal sequence polypeptide.
30. A method of preparing an RNAi agent, the method comprising incubating a dsRNA in the presence of an RDE-4 protein.
US10/645,735 1999-10-15 2003-08-20 RNA interference pathway genes as tools for targeted genetic interference Abandoned US20050100913A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US10/645,735 US20050100913A1 (en) 1999-10-15 2003-08-20 RNA interference pathway genes as tools for targeted genetic interference
US11/710,152 US7759463B2 (en) 1999-10-15 2007-02-22 RNA interference pathway genes as tools for targeted genetic interference

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US15977699P 1999-10-15 1999-10-15
US19321800P 2000-03-30 2000-03-30
US68999200A 2000-10-13 2000-10-13
US10/645,735 US20050100913A1 (en) 1999-10-15 2003-08-20 RNA interference pathway genes as tools for targeted genetic interference

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US68999200A Division 1999-10-15 2000-10-13

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US11/710,152 Continuation US7759463B2 (en) 1999-10-15 2007-02-22 RNA interference pathway genes as tools for targeted genetic interference

Publications (1)

Publication Number Publication Date
US20050100913A1 true US20050100913A1 (en) 2005-05-12

Family

ID=26856276

Family Applications (4)

Application Number Title Priority Date Filing Date
US10/645,746 Expired - Fee Related US7282564B2 (en) 1999-10-15 2003-08-20 RNA interference pathway genes as tools for targeted genetic interference
US10/645,735 Abandoned US20050100913A1 (en) 1999-10-15 2003-08-20 RNA interference pathway genes as tools for targeted genetic interference
US11/144,985 Abandoned US20060024798A1 (en) 1999-10-15 2005-06-03 RNA interference pathway genes as tools for targeted genetic interference
US11/710,152 Expired - Fee Related US7759463B2 (en) 1999-10-15 2007-02-22 RNA interference pathway genes as tools for targeted genetic interference

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US10/645,746 Expired - Fee Related US7282564B2 (en) 1999-10-15 2003-08-20 RNA interference pathway genes as tools for targeted genetic interference

Family Applications After (2)

Application Number Title Priority Date Filing Date
US11/144,985 Abandoned US20060024798A1 (en) 1999-10-15 2005-06-03 RNA interference pathway genes as tools for targeted genetic interference
US11/710,152 Expired - Fee Related US7759463B2 (en) 1999-10-15 2007-02-22 RNA interference pathway genes as tools for targeted genetic interference

Country Status (7)

Country Link
US (4) US7282564B2 (en)
EP (1) EP1235842A4 (en)
JP (1) JP2003516124A (en)
AU (1) AU1086501A (en)
CA (1) CA2386270A1 (en)
HK (1) HK1047109A1 (en)
WO (1) WO2001029058A1 (en)

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010077894A2 (en) 2008-12-16 2010-07-08 Bristol-Myers Squibb Company Methods of inhibiting quiescent tumor proliferation
EP2769737A1 (en) 2009-07-20 2014-08-27 Bristol-Myers Squibb Company Combination of an anti-CTLA4 antibody with etoposide for the synergistic treatment of proliferative diseases
WO2017160754A1 (en) 2016-03-15 2017-09-21 Mersana Therapeutics,Inc. Napi2b-targeted antibody-drug conjugates and methods of use thereof
WO2018098269A2 (en) 2016-11-23 2018-05-31 Mersana Therapeutics, Inc. Peptide-containing linkers for antibody-drug conjugates
WO2018160538A1 (en) 2017-02-28 2018-09-07 Mersana Therapeutics, Inc. Combination therapies of her2-targeted antibody-drug conjugates
WO2019104289A1 (en) 2017-11-27 2019-05-31 Mersana Therapeutics, Inc. Pyrrolobenzodiazepine antibody conjugates
WO2019126691A1 (en) 2017-12-21 2019-06-27 Mersana Therapeutics, Inc. Pyrrolobenzodiazepine antibody conjugates
US10604574B2 (en) 2016-06-30 2020-03-31 Oncorus, Inc. Oncolytic viral delivery of therapeutic polypeptides
WO2020092385A1 (en) 2018-10-29 2020-05-07 Mersana Therapeutics, Inc. Cysteine engineered antibody-drug conjugates with peptide-containing linkers
WO2020226633A1 (en) 2019-05-07 2020-11-12 Immunicom, Inc. Increasing responses to checkpoint inhibitors by extracorporeal apheresis
WO2021216920A1 (en) 2020-04-22 2021-10-28 Iovance Biotherapeutics, Inc. Systems and methods for coordinating manufacturing of cells for patient-specific immunotherapy
EP4029508A1 (en) 2014-10-10 2022-07-20 Idera Pharmaceuticals, Inc. Treatment of cancer using tlr9 agonists and checkpoint inhibitors
US11865081B2 (en) 2017-12-29 2024-01-09 Virogin Biotech Canada Ltd. Oncolytic viral delivery of therapeutic polypeptides

Families Citing this family (617)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7812149B2 (en) 1996-06-06 2010-10-12 Isis Pharmaceuticals, Inc. 2′-Fluoro substituted oligomeric compounds and compositions for use in gene modulations
US9096636B2 (en) 1996-06-06 2015-08-04 Isis Pharmaceuticals, Inc. Chimeric oligomeric compounds and their use in gene modulation
US5898031A (en) 1996-06-06 1999-04-27 Isis Pharmaceuticals, Inc. Oligoribonucleotides for cleaving RNA
US20050059016A1 (en) * 2002-11-05 2005-03-17 Ecker David J. Structural motifs and oligomeric compounds and their use in gene modulation
AUPP249298A0 (en) 1998-03-20 1998-04-23 Ag-Gene Australia Limited Synthetic genes and genetic constructs comprising same I
ATE526406T1 (en) 1998-03-20 2011-10-15 Commw Scient Ind Res Org CONTROL OF GENE EXPRESSION
DE19956568A1 (en) 1999-01-30 2000-08-17 Roland Kreutzer Method and medicament for inhibiting the expression of a given gene
US7601494B2 (en) 1999-03-17 2009-10-13 The University Of North Carolina At Chapel Hill Method of screening candidate compounds for susceptibility to biliary excretion
US6423885B1 (en) 1999-08-13 2002-07-23 Commonwealth Scientific And Industrial Research Organization (Csiro) Methods for obtaining modified phenotypes in plant cells
US7829693B2 (en) 1999-11-24 2010-11-09 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of a target gene
DE10100586C1 (en) 2001-01-09 2002-04-11 Ribopharma Ag Inhibiting gene expression in cells, useful for e.g. treating tumors, by introducing double-stranded complementary oligoRNA having unpaired terminal bases
US7491805B2 (en) 2001-05-18 2009-02-17 Sirna Therapeutics, Inc. Conjugates and compositions for cellular delivery
US7833992B2 (en) 2001-05-18 2010-11-16 Merck Sharpe & Dohme Conjugates and compositions for cellular delivery
US8202979B2 (en) 2002-02-20 2012-06-19 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid
CA2403397A1 (en) 2000-03-16 2001-09-20 Genetica, Inc. Methods and compositions for rna interference
US8202846B2 (en) 2000-03-16 2012-06-19 Cold Spring Harbor Laboratory Methods and compositions for RNA interference
US20030190635A1 (en) * 2002-02-20 2003-10-09 Mcswiggen James A. RNA interference mediated treatment of Alzheimer's disease using short interfering RNA
US8546143B2 (en) 2001-01-09 2013-10-01 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of a target gene
US7423142B2 (en) 2001-01-09 2008-09-09 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of anti-apoptotic genes
US7767802B2 (en) 2001-01-09 2010-08-03 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of anti-apoptotic genes
EP1860199B1 (en) 2001-01-12 2014-03-05 Exelixis, Inc. Modulating insulin receptor signaling
EP1229134A3 (en) 2001-01-31 2004-01-28 Nucleonics, Inc Use of post-transcriptional gene silencing for identifying nucleic acid sequences that modulate the function of a cell
US9994853B2 (en) 2001-05-18 2018-06-12 Sirna Therapeutics, Inc. Chemically modified multifunctional short interfering nucleic acid molecules that mediate RNA interference
US20050148530A1 (en) 2002-02-20 2005-07-07 Sirna Therapeutics, Inc. RNA interference mediated inhibition of vascular endothelial growth factor and vascular endothelial growth factor receptor gene expression using short interfering nucleic acid (siNA)
US7109165B2 (en) 2001-05-18 2006-09-19 Sirna Therapeutics, Inc. Conjugates and compositions for cellular delivery
AU2002316135B9 (en) 2001-05-18 2009-05-28 Sirna Therapeutics, Inc. Conjugates and compositions for cellular delivery
AU2002320264B2 (en) 2001-06-05 2008-05-01 Exelixis, Inc. GFATs as modifiers of the p53 pathway and methods of use
JP2004534955A (en) 2001-07-12 2004-11-18 エクセリクシス・インコーポレイテッド HADHs as Modifiers of the p21 Pathway and Methods of Use
US7745418B2 (en) 2001-10-12 2010-06-29 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting viral replication
DE10163098B4 (en) 2001-10-12 2005-06-02 Alnylam Europe Ag Method for inhibiting the replication of viruses
KR20050041999A (en) * 2001-11-05 2005-05-04 얀센 파마슈티카 엔.브이. Method for the in vitro synthesis of short double stranded rnas
US7294504B1 (en) 2001-12-27 2007-11-13 Allele Biotechnology & Pharmaceuticals, Inc. Methods and compositions for DNA mediated gene silencing
DE10202419A1 (en) 2002-01-22 2003-08-07 Ribopharma Ag Method of inhibiting expression of a target gene resulting from chromosome aberration
EP1432724A4 (en) 2002-02-20 2006-02-01 Sirna Therapeutics Inc Rna interference mediated inhibition of map kinase genes
US9181551B2 (en) 2002-02-20 2015-11-10 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA)
US9657294B2 (en) 2002-02-20 2017-05-23 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA)
WO2003076592A2 (en) * 2002-03-06 2003-09-18 Rigel Pharmaceuticals, Inc. Novel method for delivery and intracellular synthesis of sirna molecules
US20040180438A1 (en) 2002-04-26 2004-09-16 Pachuk Catherine J. Methods and compositions for silencing genes without inducing toxicity
EP1504126B1 (en) 2002-05-03 2014-02-26 Duke University A method of regulating gene expression
WO2004016735A2 (en) 2002-05-23 2004-02-26 Ceptyr, Inc. Modulation of biological signal transduction by rna interference
US7655790B2 (en) 2002-07-12 2010-02-02 Sirna Therapeutics, Inc. Deprotection and purification of oligonucleotides and their derivatives
US7399851B2 (en) 2002-07-25 2008-07-15 Dana Farber Cancer Institute, Inc. Composition and method for imaging cells
US20080274989A1 (en) 2002-08-05 2008-11-06 University Of Iowa Research Foundation Rna Interference Suppression of Neurodegenerative Diseases and Methods of Use Thereof
US20040241854A1 (en) 2002-08-05 2004-12-02 Davidson Beverly L. siRNA-mediated gene silencing
US20050042646A1 (en) 2002-08-05 2005-02-24 Davidson Beverly L. RNA interference suppresion of neurodegenerative diseases and methods of use thereof
ATE435303T1 (en) 2002-08-12 2009-07-15 New England Biolabs Inc METHODS AND COMPOSITIONS RELATED TO GENE SILENCING
CA2497892A1 (en) * 2002-09-04 2004-03-18 Provost, Fellows And Scholars Of The College Of The Holy And Undivided T Rinity Of Queen Elizabeth Near Dublin Compositions and methods for tissue specific or inducible inhibition of gene expression
US20080260744A1 (en) 2002-09-09 2008-10-23 Omeros Corporation G protein coupled receptors and uses thereof
WO2004032877A2 (en) 2002-10-10 2004-04-22 Wyeth Compositions, organisms and methodologies employing a novel human kinase
US7208306B2 (en) 2002-10-24 2007-04-24 Wyeth Compositions employing a novel human protein phosphatase
AU2003287505A1 (en) 2002-11-05 2004-06-03 Isis Pharmaceuticals, Inc. Chimeric oligomeric compounds and their use in gene modulation
US9150605B2 (en) 2002-11-05 2015-10-06 Isis Pharmaceuticals, Inc. Compositions comprising alternating 2′-modified nucleosides for use in gene modulation
US9150606B2 (en) 2002-11-05 2015-10-06 Isis Pharmaceuticals, Inc. Compositions comprising alternating 2'-modified nucleosides for use in gene modulation
WO2004044139A2 (en) 2002-11-05 2004-05-27 Isis Parmaceuticals, Inc. Modified oligonucleotides for use in rna interference
AU2003291753B2 (en) 2002-11-05 2010-07-08 Isis Pharmaceuticals, Inc. Polycyclic sugar surrogate-containing oligomeric compounds and compositions for use in gene modulation
CA2505416A1 (en) 2002-11-21 2004-06-10 Wyeth Methods for diagnosing rcc and other solid tumors
US7297525B2 (en) 2002-11-27 2007-11-20 Wyeth Composition employing a novel human kinase
US7803365B2 (en) 2002-12-02 2010-09-28 Biovec, Llc Ex vivo and in vivo expression of the thrombomodulin gene for the treatment of cardiovascular and peripheral vascular diseases
US9388427B2 (en) 2002-12-02 2016-07-12 Biovec, Llc In vivo and ex vivo gene transfer into renal tissue using gutless adenovirus vectors
WO2004058147A2 (en) 2002-12-16 2004-07-15 Halozyme, Inc. Human chondroitinase glycoprotein (chasegp), process for preparing the same, and pharmaceutical compositions comprising thereof
KR101250818B1 (en) 2002-12-24 2013-04-15 리나트 뉴로사이언스 코프. Anti-ngf antibodies and methods using same
WO2004058948A2 (en) * 2002-12-24 2004-07-15 Genpath Pharmaceuticals, Incorporated In vivo methods for validating the role of a tumor maintenance gene
US9498530B2 (en) 2002-12-24 2016-11-22 Rinat Neuroscience Corp. Methods for treating osteoarthritis pain by administering a nerve growth factor antagonist and compositions containing the same
AU2004213452A1 (en) 2003-02-14 2004-09-02 Sagres Discovery, Inc. Therapeutic GPCR targets in cancer
JP2006525960A (en) 2003-02-19 2006-11-16 ライナット ニューロサイエンス コーポレイション Nerve growth factor agonists and NSAIDs and methods for treating pain by administration of compositions comprising them
CN102943067B (en) 2003-03-05 2016-06-22 海洋酶公司 Soluble hyaluronidase glycoprotein (sHASEGP), prepare their method, their purposes and the pharmaceutical composition comprising them
US20060104968A1 (en) 2003-03-05 2006-05-18 Halozyme, Inc. Soluble glycosaminoglycanases and methods of preparing and using soluble glycosaminogly ycanases
US7871607B2 (en) 2003-03-05 2011-01-18 Halozyme, Inc. Soluble glycosaminoglycanases and methods of preparing and using soluble glycosaminoglycanases
US8158420B2 (en) 2003-04-04 2012-04-17 The Trustees Of Columbia University In The City Of New York Methods for inhibiting the differentation of proliferative telencephalic cells in vitro by addition of ATF5
EP3222294A1 (en) 2003-04-30 2017-09-27 Sirna Therapeutics, Inc. Conjugates and compositions for cellular delivery
IL155783A (en) 2003-05-05 2010-11-30 Technion Res & Dev Foundation Multicellular systems of pluripotent human embryonic stem cells and cancer cells and uses thereof
WO2004101756A2 (en) 2003-05-09 2004-11-25 Diadexus, Inc. Ovr110 antibody compositions and methods of use
EP1631306A4 (en) 2003-05-19 2009-08-12 Univ Columbia Compositions and methods for treating and preventing heart tissue degeneration, and uses thereof
EP1631659A4 (en) * 2003-05-22 2008-02-13 Isis Pharmaceuticals Inc Modulation of the rna interference pathway
EP1633307A4 (en) 2003-06-03 2009-06-24 Isis Pharmaceuticals Inc Modulation of survivin expression
JP5183064B2 (en) * 2003-07-02 2013-04-17 エムユーエスシー ファウンデイション フォー リサーチ デべロップメント Specific and non-specific immunity induced by dsRNA in crustaceans and other invertebrates, and biological delivery vehicles used therein
US20080050313A1 (en) 2003-08-14 2008-02-28 Exelixis, Inc. Sulfs as Modifiers of the Beta-Catenin Pathway and Methods of Use
US8071652B2 (en) 2003-08-21 2011-12-06 The Board Of Regents Of The University Of Texas System Method of treating irritable bowel syndrome
WO2005035769A2 (en) 2003-10-09 2005-04-21 E. I. Du Pont De Nemours And Company Gene silencing by using micro-rna molecules
EP2395112B1 (en) 2003-12-31 2017-02-15 The Penn State Research Foundation Methods for predicting and overcoming resistance to chemotherapy in ovarian cancer and for predicting colon cancer occurrence
US7695964B2 (en) 2004-01-23 2010-04-13 New England Biolabs, Inc. Compositions and methods for generating short double-stranded RNA using mutated RNAse III
JP2007523649A (en) 2004-02-10 2007-08-23 サーナ・セラピューティクス・インコーポレイテッド Inhibition of gene expression via RNA interference using multifunctional short interfering nucleic acids (multifunctional siNA)
WO2005078848A2 (en) 2004-02-11 2005-08-25 University Of Tennessee Research Foundation Inhibition of tumor growth and invasion by anti-matrix metalloproteinase dnazymes
US7781393B2 (en) 2004-02-25 2010-08-24 Dana-Farber Cancer Institute, Inc. Methods for inhibiting tumor cell growth
WO2006074418A2 (en) 2005-01-07 2006-07-13 Diadexus, Inc. Ovr110 antibody compositions and methods of use
WO2005094391A2 (en) 2004-04-02 2005-10-13 The Regents Of The University Of California METHODS AND COMPOSITIONS FOR TREATING AND PREVENTING DISEASE ASSOCIATED WITH αVβ5 INTEGRIN
US20070031417A2 (en) * 2004-04-14 2007-02-08 University Of Massachusetts Dicer interacting proteins and uses therefor
EP1747022A4 (en) 2004-04-23 2010-03-31 Univ Columbia INHIBITION OF HAIRLESS PROTEIN mRNA
DE102004025881A1 (en) 2004-05-19 2006-01-05 Beiersdorf Ag Oligoribonucleotides for influencing hair growth
US10508277B2 (en) 2004-05-24 2019-12-17 Sirna Therapeutics, Inc. Chemically modified multifunctional short interfering nucleic acid molecules that mediate RNA interference
EP2471923B1 (en) 2004-05-28 2014-08-20 Asuragen, Inc. Methods and compositions involving microRNA
US8394947B2 (en) 2004-06-03 2013-03-12 Isis Pharmaceuticals, Inc. Positionally modified siRNA constructs
EP2441847B1 (en) 2004-06-21 2015-09-23 Proteome Sciences Plc Screening methods using syk in combination with tau protein
WO2006009575A1 (en) 2004-06-22 2006-01-26 The Board Of Trustees Of The University Of Illinois METHODS OF INHIBITING TUMOR CELL PROLIFERATION WITH FOXM1 siRNA
US7968762B2 (en) 2004-07-13 2011-06-28 Van Andel Research Institute Immune-compromised transgenic mice expressing human hepatocyte growth factor (hHGF)
AU2005330703A1 (en) 2004-07-19 2006-10-26 Baylor College Of Medicine Modulation of cytokine signaling regulators and applications for immunotherapy
US20060024677A1 (en) 2004-07-20 2006-02-02 Morris David W Novel therapeutic targets in cancer
EP2484780A1 (en) 2004-07-23 2012-08-08 The University of North Carolina At Chapel Hill Methods and materials for determining pain sensibility and predicting and treating related disorders
US7846438B2 (en) 2004-08-03 2010-12-07 Biogen Idec Ma Inc. Methods of promoting neurite outgrowth with soluble TAJ polypeptides
WO2006017932A1 (en) 2004-08-18 2006-02-23 Genesense Technologies Inc. Small interfering rna molecules against ribonucleotide reductase and uses thereof
US7884086B2 (en) 2004-09-08 2011-02-08 Isis Pharmaceuticals, Inc. Conjugates for use in hepatocyte free uptake assays
US8975471B2 (en) * 2004-10-12 2015-03-10 The Rockefeller University MicroRNAs
AU2005295796B2 (en) 2004-10-13 2010-07-22 University Of Georgia Research Foundation, Inc. Nematode resistant transgenic plants
EP2287303B1 (en) * 2004-11-12 2014-07-02 Asuragen, Inc. Methods and compositions involving miRNA and miRNA inhibitor molecules
US9944713B2 (en) 2004-11-24 2018-04-17 Medicinal Bioconvergence Research Center Antibody specific to the AIMP2-DX2
US7939490B2 (en) 2004-12-13 2011-05-10 University Of Maryland, Baltimore TWEAK as a therapeutic target for treating central nervous system diseases associated with cerebral edema and cell death
AU2006236453B2 (en) * 2005-01-25 2012-02-23 Board Of Regents, The University Of Texas System Delivery of siRNA by neutral lipid compositions
ES2381201T3 (en) 2005-03-31 2012-05-24 Calando Pharmaceuticals, Inc. Inhibitors of the subunit 2 of the ribonucleotide reductase and uses thereof
US20090214536A1 (en) 2005-04-07 2009-08-27 Guoying Yu CACNA1E in Cancer Diagnosis, Detection and Treatment
US20090220495A1 (en) 2005-04-07 2009-09-03 Abdallah Fanidi Cancer Related Genes (PRLR)
EP1888052A2 (en) 2005-05-12 2008-02-20 Wisconsin Alumni Research Foundation Blockade of pin1 prevents cytokine production by activated immune cells
FR2885808B1 (en) 2005-05-19 2007-07-06 Oreal VECTORIZATION OF DSRNA BY CATIONIC PARTICLES AND TOPICAL USE.
WO2006131925A2 (en) * 2005-06-10 2006-12-14 Quark Pharmaceuticals, Inc. Oligoribonucleotides and methods of use thereof for treatment of fibrotic conditions and other diseases
US7868159B2 (en) 2005-06-23 2011-01-11 Baylor College Of Medicine Modulation of negative immune regulators and applications for immunotherapy
WO2007011702A2 (en) 2005-07-15 2007-01-25 The University Of North Carolina At Chapel Hill Use of egfr inhibitors to prevent or treat obesity
EP1947942B1 (en) 2005-08-18 2015-07-29 Accelalox, Inc. Methods for bone treatment by modulating an arachidonic acid metabolic or signaling pathway
EP2980220A1 (en) 2005-09-20 2016-02-03 BASF Plant Science GmbH Improved methods controlling gene expression
CA2660661A1 (en) 2005-09-26 2007-04-05 The Trustees Of Columbia University In The City Of New York Side population cells in cardiac repair
EP2522722A3 (en) 2005-12-09 2012-12-12 BASF Plant Science GmbH Nucleic acid molecules encoding polypeptides involved in regulation of sugar and lipid metabolism and methods of use VIII
US20090012029A1 (en) 2006-01-06 2009-01-08 Hussey Richard S Cyst Nematode Resistant Transgenic Plants
US8669345B2 (en) 2006-01-27 2014-03-11 Biogen Idec Ma Inc. Nogo receptor antagonists
WO2007123777A2 (en) 2006-03-30 2007-11-01 Duke University Inhibition of hif-1 activation for anti-tumor and anti-inflammatory responses
US9044461B2 (en) 2006-04-07 2015-06-02 The Research Foundation Of State University Of New York Transcobalamin receptor polypeptides, nucleic acids, and modulators thereof, and related methods of use in modulating cell growth and treating cancer and cobalamin deficiency
US8524454B2 (en) 2006-04-07 2013-09-03 The Research Foundation Of State University Of New York Transcobalamin receptor polypeptides, nucleic acids, and modulators thereof, and related methods of use in modulating cell growth and treating cancer and cobalamin deficiency
WO2007120787A2 (en) 2006-04-13 2007-10-25 Novartis Vaccines & Diagnostics, Inc. Methods of treating, diagnosing or detecting cancers associated with liv-1 overexpression
US7572618B2 (en) 2006-06-30 2009-08-11 Bristol-Myers Squibb Company Polynucleotides encoding novel PCSK9 variants
CA2657319C (en) 2006-07-11 2016-06-21 University Of Medicine And Dentistry Of New Jersey Mg53 compositions and methods of use
EP2412383A1 (en) 2006-07-28 2012-02-01 Children's Memorial Hospital Methods of inhibiting tumor cell aggressiveness using the microenvironment of human embryonic stem cells
US9580515B2 (en) 2006-08-21 2017-02-28 Zensun (Shanghai) Science & Technology, Co., Ltd. Neukinase, a downstream protein of neuregulin
EP2182056B1 (en) 2006-10-06 2015-12-23 BASF Plant Science GmbH Method for producing polyunsaturated fatty acids in transgenic non-human organisms
EP2101813B1 (en) 2006-11-27 2014-04-02 Patrys Limited Novel glycosylated peptide target in neoplastic cells
EP2962697A1 (en) 2006-11-27 2016-01-06 diaDexus, Inc. Ovr110 antibody compositions and methods of use
EP2913341A1 (en) 2006-12-22 2015-09-02 University of Utah Research Foundation Method of detecting ocular diseases and pathologic conditions and treatment of same
EP2117305A4 (en) 2007-01-26 2011-03-30 Univ Louisville Res Found Modification of exosomal components for use as a vaccine
US20100292301A1 (en) * 2007-02-28 2010-11-18 Elena Feinstein Novel sirna structures
US8067390B2 (en) * 2007-03-02 2011-11-29 The Board Of Regents Of The University Of Texas System Therapeutic targeting of interleukins using siRNA in neutral liposomes
JP2010521180A (en) 2007-03-14 2010-06-24 ノバルティス アーゲー APCDD1 inhibitor for treating, diagnosing or detecting cancer
US8796442B2 (en) 2007-03-21 2014-08-05 Brookhaven Science Associates, Llc. Combined hairpin-antisense compositions and methods for modulating expression
WO2008118324A2 (en) 2007-03-26 2008-10-02 Macrogenics, Inc. Composition and method of treating cancer with an anti-uroplakin ib antibody
US20100322949A1 (en) 2007-04-26 2010-12-23 Ludwig Institute For Cancer Research Ltd. Methods for diagnosing and treating astrocytomas
WO2008137115A1 (en) 2007-05-03 2008-11-13 The Brigham And Women's Hospital, Inc. Multipotent stem cells and uses thereof
EP2492346A3 (en) 2007-05-04 2013-03-27 BASF Plant Science GmbH Seed enhancement by combinations of pyruvate kinases
WO2008143786A1 (en) * 2007-05-14 2008-11-27 The Rockefeller University Production of artificial micrornas using synthetic microrna precursors
US8097422B2 (en) 2007-06-20 2012-01-17 Salk Institute For Biological Studies Kir channel modulators
US9689031B2 (en) 2007-07-14 2017-06-27 Ionian Technologies, Inc. Nicking and extension amplification reaction for the exponential amplification of nucleic acids
WO2009012263A2 (en) 2007-07-18 2009-01-22 The Trustees Of Columbia University In The City Of New York Tissue-specific micrornas and compositions and uses thereof
CN101802211B (en) 2007-08-02 2014-10-01 诺维莫尼公司 Anti-RANTES antibodies and methods of use thereof
EP2192916A4 (en) 2007-08-23 2012-04-04 Univ Leland Stanford Junior Modulation of synaptogenesis
US8361714B2 (en) 2007-09-14 2013-01-29 Asuragen, Inc. Micrornas differentially expressed in cervical cancer and uses thereof
CN101815521B (en) * 2007-10-03 2014-12-10 夸克制药公司 Novel siRNA structures
EP2065037A1 (en) 2007-11-27 2009-06-03 DKFZ Deutsches Krebsforschungszentrum Means and methods for treating or preventing brain tumors based on the nuclear receptor tailless (Tlx)
CN101932339B (en) 2007-11-30 2014-10-29 贝勒医学院 Dendritic cell vaccine compositions and uses of same
WO2009070805A2 (en) * 2007-12-01 2009-06-04 Asuragen, Inc. Mir-124 regulated genes and pathways as targets for therapeutic intervention
US8614311B2 (en) 2007-12-12 2013-12-24 Quark Pharmaceuticals, Inc. RTP801L siRNA compounds and methods of use thereof
US20110105584A1 (en) * 2007-12-12 2011-05-05 Elena Feinstein Rtp80il sirna compounds and methods of use thereof
CA2708093A1 (en) 2007-12-17 2009-06-25 Basf Plant Science Gmbh Lipid metabolism protein and uses thereof i (bzip transcription factor)
EP2072528A1 (en) 2007-12-19 2009-06-24 Labeit, Mr. Siegfried A host cell deficient for MuRF1 and MuRF2
WO2009090639A2 (en) * 2008-01-15 2009-07-23 Quark Pharmaceuticals, Inc. Sirna compounds and methods of use thereof
US20110028531A1 (en) * 2008-03-20 2011-02-03 Elena Feinstein Novel sirna compounds for inhibiting rtp801
EP2283133A2 (en) 2008-04-04 2011-02-16 Calando Pharmaceuticals, Inc. Compositions and use of epas1 inhibitors
EP2285385A4 (en) * 2008-04-15 2013-01-16 Quark Pharmaceuticals Inc siRNA COMPOUNDS FOR INHIBITING NRF2
GB0807018D0 (en) 2008-04-17 2008-05-21 Fusion Antibodies Ltd Antibodies and treatment
WO2009137807A2 (en) 2008-05-08 2009-11-12 Asuragen, Inc. Compositions and methods related to mirna modulation of neovascularization or angiogenesis
US20090291073A1 (en) * 2008-05-20 2009-11-26 Ward Keith W Compositions Comprising PKC-theta and Methods for Treating or Controlling Ophthalmic Disorders Using Same
EP2293800B1 (en) 2008-06-06 2016-10-05 Quark Pharmaceuticals, Inc. Compositions and methods for treatment of ear disorders
CN102209522B (en) 2008-09-10 2016-04-06 布拉德福德大学 The Pigmented composition of regulation of skin and method
EP2727996A1 (en) 2008-11-06 2014-05-07 The Johns-Hopkins University Treatment of chronic inflammatory respiratory disorders with NP1 inhibitors
CA2743057C (en) 2008-11-07 2019-11-26 Research Development Foundation Compositions and methods for the inhibition of cripto/grp78 complex formation and signaling
WO2010054379A2 (en) 2008-11-10 2010-05-14 The United States Of America, As Represensted By The Secretary, Department Of Health And Human Services Gene signature for predicting prognosis of patients with solid tumors
CA2745111A1 (en) 2008-12-03 2010-06-10 Research Development Foundation Modulation of olfml-3 mediated angiogenesis
AU2009326129B2 (en) 2008-12-12 2016-05-12 Basf Plant Science Gmbh Desaturases and process for the production of polyunsaturated fatty acids in transgenic organisms
WO2010080452A2 (en) 2008-12-18 2010-07-15 Quark Pharmaceuticals, Inc. siRNA COMPOUNDS AND METHODS OF USE THEREOF
WO2010074783A1 (en) 2008-12-23 2010-07-01 The Trustees Of Columbia University In The City Of New York Phosphodiesterase inhibitors and uses thereof
JP2012513464A (en) 2008-12-23 2012-06-14 ザ トラスティーズ オブ コロンビア ユニヴァーシティ イン ザ シティ オブ ニューヨーク Phosphodiesterase inhibitors and uses thereof
AU2010226604A1 (en) 2009-03-19 2011-10-13 Merck Sharp & Dohme Corp. RNA interference mediated inhibition of BTB and CNC homology 1, basic leucine zipper transcription factor 1 (Bach 1) gene expression using short interfering nucleic acid (siNA) sequence listing
US20120035247A1 (en) 2009-03-19 2012-02-09 Merck Sharp & Dohme Corp. RNA Interference Mediated Inhibition of Signal Transducer and Activator of Transcription 6 (STAT6) Gene Expression Using Short Interfering Nucleic Acid (siNA)
US20120016011A1 (en) 2009-03-19 2012-01-19 Merck Sharp & Dohme Corp. RNA Interference Mediated Inhibition of Connective Tissue Growth Factor (CTGF) Gene Expression Using Short Interfering Nucleic Acid (siNA)
US20120029054A1 (en) 2009-03-19 2012-02-02 Merck Sharp & Dohme Corp. RNA Interference Mediated Inhibition of GATA Binding Protein 3 (GATA3) Gene Expression Using Short Intefering Nucleic Acid (siNA)
SG174367A1 (en) 2009-03-23 2011-10-28 Quark Pharmaceuticals Inc Compounds compositions and methods of treating cancer and fibrotic diseases
US20110045080A1 (en) * 2009-03-24 2011-02-24 William Marsh Rice University Single-Walled Carbon Nanotube/Bioactive Substance Complexes and Methods Related Thereto
US20120010272A1 (en) 2009-03-27 2012-01-12 Merck Sharp & Dohme Corp. RNA Interference Mediated Inhibition of Apoptosis Signal-Regulating Kinase 1 (ASK1) Gene Expression Using Short Interfering Nucleic Acid (siNA)
WO2010111490A2 (en) 2009-03-27 2010-09-30 Merck Sharp & Dohme Corp. RNA INTERFERENCE MEDIATED INHIBITION OF THE THYMIC STROMAL LYMPHOPOIETIN (TSLP) GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)
WO2010111468A2 (en) 2009-03-27 2010-09-30 Merck Sharp & Dohme Corp. RNA INTERFERENCE MEDIATED INHIBITION OF THE NERVE GROWTH FACTOR BETA CHAIN (NGFß) GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (SINA)
EP2411019A2 (en) 2009-03-27 2012-02-01 Merck Sharp&Dohme Corp. RNA INTERFERENCE MEDIATED INHIBITION OF SIGNAL TRANSDUCER AND ACTIVATOR OF TRANSCRIPTION 1 (STAT1) GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)
MX2011010072A (en) 2009-03-27 2011-10-06 Merck Sharp & Dohme RNA INTERFERENCE MEDIATED INHIBITION OF THE INTERCELLULAR ADHESION MOLECULE 1 (ICAM-1)GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA).
WO2010118243A2 (en) 2009-04-08 2010-10-14 Genentech, Inc. Use of il-27 antagonists to treat lupus
US8283332B2 (en) 2009-04-17 2012-10-09 University Of Louisville Research Foundation, Inc. PFKFB4 inhibitors and methods of using the same
EP2243828A1 (en) 2009-04-24 2010-10-27 DKFZ Deutsches Krebsforschungszentrum Use of mixed lineage like kinase polypeptides (MLKL polypeptides) in cancer therapy
EP2258858A1 (en) 2009-06-05 2010-12-08 Universitätsklinikum Freiburg Transgenic LSD1 animal model for cancer
CN102458099B (en) 2009-06-05 2015-08-19 佛罗里达大学研究基金公司 The separation of sugarcane Lignin biosynthesis gene and targeted inhibition
SI2453923T1 (en) 2009-07-14 2016-04-29 Mayo Foundation For Medical Education And Research Peptide-mediated non-covalent delivery of active agents across the blood brain barrier
KR101752515B1 (en) 2009-07-24 2017-06-29 더 리젠츠 오브 더 유니버시티 오브 캘리포니아 Methods and compositions for treating and preventing disease associated with avb5 integrin
US20120309073A1 (en) * 2009-09-10 2012-12-06 Whitehead Institute For Biomedical Research Rnai in budding yeast
WO2011038002A1 (en) 2009-09-22 2011-03-31 Yale University Immunogenic epitopes as targets for universal cancer vaccines
WO2011057171A1 (en) 2009-11-08 2011-05-12 Quark Pharmaceuticals, Inc. METHODS FOR DELIVERY OF siRNA TO THE SPINAL CORD AND THERAPIES ARISING THEREFROM
EP2501414B1 (en) 2009-11-17 2018-01-10 The Trustees Of The University Of Pennsylvania Smndelta7 degron: novel compositions and methods of use
JP5889198B2 (en) 2009-11-23 2016-03-22 アクアバウンティ テクノロジーズ インコーポレイテッド Maternally induced sterility in animals
CA2776568A1 (en) 2009-11-26 2011-06-03 Quark Pharmaceuticals, Inc. Sirna compounds comprising terminal substitutions
CA2785996C (en) 2009-12-07 2021-04-13 The Johns Hopkins University Sr-bi as a predictor of human female infertility and responsiveness to treatment
WO2011072091A1 (en) 2009-12-09 2011-06-16 Quark Pharmaceuticals, Inc. Methods and compositions for treating diseases, disorders or injury of the cns
PT2509991E (en) 2009-12-09 2016-01-12 Nitto Denko Corp Modulation of hsp47 expression
ES2764999T3 (en) 2009-12-10 2020-06-05 Univ Columbia Histone acetyltransferase activators and uses thereof
US10640457B2 (en) 2009-12-10 2020-05-05 The Trustees Of Columbia University In The City Of New York Histone acetyltransferase activators and uses thereof
EP2513309A2 (en) 2009-12-18 2012-10-24 Novartis AG Organic compositions to treat hsf1-related diseases
CA2785492C (en) 2009-12-23 2018-07-24 Novartis Ag Lipids, lipid compositions, and methods of using them
WO2011084193A1 (en) 2010-01-07 2011-07-14 Quark Pharmaceuticals, Inc. Oligonucleotide compounds comprising non-nucleotide overhangs
EP2529033B1 (en) 2010-01-26 2017-05-24 National Jewish Health Methods for risk prediction, diagnosis, prognosis of pulmonary disorders
KR20120125357A (en) 2010-02-10 2012-11-14 노파르티스 아게 Methods and compounds for muscle growth
WO2011133931A1 (en) 2010-04-22 2011-10-27 Genentech, Inc. Use of il-27 antagonists for treating inflammatory bowel disease
KR101223660B1 (en) 2010-05-20 2013-01-17 광주과학기술원 Pharmaceutical Compositions for Preventing or Treating Arthritis Comprising HIF-2α Inhibitor as an Active Ingredient
CA2800065A1 (en) 2010-05-21 2011-11-24 Peptimed, Inc. Reagents and methods for treating cancer
EP2575818A4 (en) 2010-06-03 2013-11-06 Pharmacyclics Inc The use of inhibitors of bruton's tyrosine kinase (btk)
WO2011163466A1 (en) 2010-06-23 2011-12-29 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Regulation of skin pigmentation by neuregulin-1 (nrg-1)
WO2012018754A2 (en) 2010-08-02 2012-02-09 Merck Sharp & Dohme Corp. RNA INTERFERENCE MEDIATED INHIBITION OF CATENIN (CADHERIN-ASSOCIATED PROTEIN), BETA 1 (CTNNB1) GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)
RU2624045C2 (en) 2010-08-17 2017-06-30 Сирна Терапьютикс,Инк RNA INTERFERENCE MEDIATED INHIBITION OF HEPATITIS B VIRUS (HBV) GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)
EP4050109A1 (en) 2010-08-18 2022-08-31 Fred Hutchinson Cancer Center Agents for use in treating facioscapulohumeral dystrophy (fshd)
US9243246B2 (en) 2010-08-24 2016-01-26 Sirna Therapeutics, Inc. Single-stranded RNAi agents containing an internal, non-nucleic acid spacer
EP2609106A4 (en) 2010-08-26 2014-03-19 Merck Sharp & Dohme RNA INTERFERENCE MEDIATED INHIBITION OF PROLYL HYDROXYLASE DOMAIN 2 (PHD2) GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)
US20140134231A1 (en) 2010-10-11 2014-05-15 Sanford-Burnham Medical Research Institute Mir-211 expression and related pathways in human melanoma
BR112013009196A2 (en) 2010-10-15 2020-08-25 The Trustees Of Columbia University In The City Of New York uses of polypeptide to reduce fatty acid acquisition and food intake, as well as promoting satiety related to obesity
WO2012058210A1 (en) 2010-10-29 2012-05-03 Merck Sharp & Dohme Corp. RNA INTERFERENCE MEDIATED INHIBITION OF GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACIDS (siNA)
SG190088A1 (en) 2010-11-01 2013-06-28 Peptimed Inc Compositions of a peptide targeting system for treating cancer
SI2635299T1 (en) 2010-11-02 2020-02-28 The Trustees Of Columbia University In The City Of New York Methods for treating hair loss disorders
US9198911B2 (en) 2010-11-02 2015-12-01 The Trustees Of Columbia University In The City Of New York Methods for treating hair loss disorders
CN103298939A (en) 2010-12-06 2013-09-11 夸克医药公司 Double stranded oligonucleotide compounds comprising positional modifications
US9150926B2 (en) 2010-12-06 2015-10-06 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Diagnosis and treatment of adrenocortical tumors using human microRNA-483
SI2649086T1 (en) 2010-12-09 2018-01-31 The Trustees Of The University Of Pennsylvania Center For Technology Transfer Use of chimeric antigen receptor-modified t cells to treat cancer
US9969677B2 (en) 2010-12-22 2018-05-15 The Trustees Of Columbia University In The City Of New York Histone acetyltransferase modulators and uses thereof
MX2013008376A (en) 2011-01-18 2013-08-12 Univ Pennsylvania Compositions and methods for treating cancer.
US9222085B2 (en) 2011-02-03 2015-12-29 Mirna Therapeutics, Inc. Synthetic mimics of MIR-124
EP2487242A1 (en) 2011-02-09 2012-08-15 Ruprecht-Karls-Universität Heidelberg B-type plexin antagonists and uses thereof
US9796979B2 (en) 2011-03-03 2017-10-24 Quark Pharmaceuticals Inc. Oligonucleotide modulators of the toll-like receptor pathway
CA2828002A1 (en) 2011-03-03 2012-09-07 Quark Pharmaceuticals, Inc. Compositions and methods for treating lung disease and injury
SG193280A1 (en) 2011-03-03 2013-10-30 Quark Pharmaceuticals Inc Oligonucleotide modulators of the toll-like receptor pathway
KR101291668B1 (en) 2011-04-21 2013-08-01 서울대학교산학협력단 Shuttle Vectors for Mycobacteria-Escherichia coli and Uses Thereof
US10196637B2 (en) 2011-06-08 2019-02-05 Nitto Denko Corporation Retinoid-lipid drug carrier
TWI658830B (en) 2011-06-08 2019-05-11 日東電工股份有限公司 Retinoid-liposomes for enhancing modulation of hsp47 expression
GB201109966D0 (en) 2011-06-10 2011-07-27 Cancer Res Inst Royal Materials and methods for treating pten mutated or deficient cancer
GB201109965D0 (en) 2011-06-10 2011-07-27 Cancer Res Inst Royal Materials and methods for treating estrogen receptor alpher(ER) positive cancer
WO2013055420A2 (en) 2011-07-12 2013-04-18 Philadelphia Health & Education Corporation Novel clostridium difficile dna vaccine
US9120858B2 (en) 2011-07-22 2015-09-01 The Research Foundation Of State University Of New York Antibodies to the B12-transcobalamin receptor
EA201490364A1 (en) 2011-07-29 2014-08-29 Дзе Трастиз Оф Дзе Юниверсити Оф Пенсильвания COSTIMULATING SWITCHING RECEPTORS
BR112014005104A2 (en) 2011-09-02 2017-07-04 Salk Inst For Biological Studi camkii, ip3r, calcineurin, p38 and mk2 / 3 inhibitors to treat bind metabolic disorders of obesity
RU2640913C2 (en) 2011-09-07 2018-01-12 Дойчес Кребсфоршунгсцентрум Means and methods for cancer treatment and/or prevention depending on natural ahr ligand
WO2013040251A2 (en) 2011-09-13 2013-03-21 Asurgen, Inc. Methods and compositions involving mir-135b for distinguishing pancreatic cancer from benign pancreatic disease
US9708384B2 (en) 2011-09-22 2017-07-18 The Trustees Of The University Of Pennsylvania Universal immune receptor expressed by T cells for the targeting of diverse and multiple antigens
US9352312B2 (en) 2011-09-23 2016-05-31 Alere Switzerland Gmbh System and apparatus for reactions
SG11201401477XA (en) 2011-10-14 2014-07-30 Genentech Inc ANTI-HtrA1 ANTIBODIES AND METHODS OF USE
AU2012325997C1 (en) 2011-10-18 2018-07-05 Dicerna Pharmaceuticals, Inc. Amine cationic lipids and uses thereof
WO2013063419A2 (en) 2011-10-28 2013-05-02 The Trustees Of The University Of Pennsylvania A fully human, anti-mesothelin specific chimeric immune receptor for redirected mesothelin-expressing cell targeting
ES2861435T3 (en) 2011-11-03 2021-10-06 Univ Pennsylvania Specific compositions of isolated B7-H4 and methods of using them
BR112014010769B1 (en) 2011-11-03 2021-08-03 Quark Pharmaceuticals, Inc USE OF A DOUBLE TAPE RNA COMPOUND TO PREPARE A DRUG TO TREAT MEN'S DISEASE
US20140322216A1 (en) 2011-11-08 2014-10-30 The Trustees Of The University Of Pennsylvania Glypican-3-specific antibody and uses thereof
WO2013070821A1 (en) 2011-11-08 2013-05-16 Quark Pharmaceuticals, Inc. Methods and compositions for treating diseases, disorders or injury of the nervous system
WO2013078230A1 (en) 2011-11-23 2013-05-30 The Trustees Of The University Of Pennsylvania Use of pdl1 expressing cells to convert t cells into regulatory t cells
AU2013216920B2 (en) 2012-02-07 2016-09-22 Global Bio Therapeutics, Inc. Compartmentalized method of nucleic acid delivery and compositions and uses thereof
KR20140127829A (en) 2012-02-22 2014-11-04 더 트러스티스 오브 더 유니버시티 오브 펜실바니아 Use of the CD2 Signaling Domain in Second-Generation Chimeric Antigen Receptors
KR20140135715A (en) 2012-02-22 2014-11-26 더 트러스티스 오브 더 유니버시티 오브 펜실바니아 Use of icos-based cars to enhance antitumor activity and car persistence
CA2863799C (en) 2012-02-22 2020-09-01 Matthew J. FRIGAULT Compositions and methods for generating a persisting population of t cells useful for the treatment of cancer
PT2830662T (en) 2012-03-29 2018-11-29 Univ Columbia Methods for treating hair loss disorders
EP2650013A1 (en) 2012-04-10 2013-10-16 Deutsches Krebsforschungszentrum Inhibitors of Receptor for Advanced Glycation-End products (RAGE) for use in treating and/or preventing inflammation- and/or damage-induced cancer
EP3453762B1 (en) 2012-05-02 2021-04-21 Sirna Therapeutics, Inc. Short interfering nucleic acid (sina) compositions
WO2013187556A1 (en) 2012-06-14 2013-12-19 Scripps Korea Antibody Institute Novel antibody specific for clec14a and uses thereof
US20150204851A1 (en) 2012-06-27 2015-07-23 Metanomics Health Gmbh Methods for identifying diabetes drugs
EP3699284A1 (en) 2012-07-05 2020-08-26 The Trustees of the University of Pennsylvania U1 snrnp regulates gene expression and modulates oncogenicity
EA201590208A1 (en) 2012-07-13 2015-11-30 Дзе Трастиз Оф Дзе Юниверсити Оф Пенсильвания CAR ACTIVITY STRENGTHENING BY CAR BY JOINT INTRODUCTION OF BISPECIFIC ANTIBODIES
MX370495B (en) 2012-07-13 2019-12-13 Univ Pennsylvania Methods of assessing the suitability of transduced t cells for administration.
EP2877494B1 (en) 2012-07-23 2020-07-15 La Jolla Institute for Allergy and Immunology Ptprs and proteoglycans in autoimmune disease
EP2877598A1 (en) 2012-07-24 2015-06-03 Pharmacyclics, Inc. Mutations associated with resistance to inhibitors of bruton's tyrosine kinase (btk)
US9937205B2 (en) 2012-09-04 2018-04-10 The Trustees Of The University Of Pennsylvania Inhibition of diacylglycerol kinase to augment adoptive T cell transfer
WO2014045126A2 (en) 2012-09-18 2014-03-27 Uti Limited Partnership Treatment of pain by inhibition of usp5 de-ubiquitinase
TW201414837A (en) 2012-10-01 2014-04-16 Univ Pennsylvania Compositions and methods for targeting stromal cells for the treatment of cancer
US9598489B2 (en) 2012-10-05 2017-03-21 The Trustees Of The Univeristy Of Pennsylvania Human alpha-folate receptor chimeric antigen receptor
EP3677310A1 (en) 2012-10-08 2020-07-08 St. Jude Children's Research Hospital Therapies based on control of regulatory t cell stability and function via a neuropilin-1:semaphorin axis
WO2015116178A1 (en) 2014-01-31 2015-08-06 Thomas Jefferson University Fusion proteins for modulating regulatory and effector t cells
EP2951199A4 (en) 2013-01-31 2016-07-20 Univ Jefferson Fusion proteins for modulating regulatory and effector t cells
EP2958943B1 (en) 2013-02-20 2019-09-11 The Trustees Of The University Of Pennsylvania Treatment of cancer using humanized anti-egfrviii chimeric antigen receptor
ES2814962T3 (en) 2013-02-20 2021-03-29 Novartis Ag Efficient targeting of primary human leukemia using anti-CD123 chimeric antigen receptor modified T cells
EP2961853B1 (en) 2013-02-28 2018-09-19 The Board of Regents of The University of Texas System Methods for classifying a cancer as susceptible to tmepai-directed therapies and treating such cancers
US8957044B2 (en) 2013-03-01 2015-02-17 Wake Forest University Health Sciences Systemic gene replacement therapy for treatment of X-linked myotubular myopathy (XLMTM)
EP3608308B1 (en) 2013-03-08 2021-07-21 Novartis AG Lipids and lipid compositions for the delivery of active agents
ES2769574T3 (en) 2013-03-15 2020-06-26 Michael C Milone Recognition of cytotoxic cells with chimeric receptors for adoptive immunotherapy
AU2014233198B2 (en) 2013-03-15 2019-06-27 Sutter West Bay Hospitals Falz for use as a target for therapies to treat cancer
US9446105B2 (en) 2013-03-15 2016-09-20 The Trustees Of The University Of Pennsylvania Chimeric antigen receptor specific for folate receptor β
CA2907152A1 (en) 2013-03-15 2014-09-25 The Trustees Of Columbia University In The City Of New York Fusion proteins and methods thereof
UY35468A (en) 2013-03-16 2014-10-31 Novartis Ag CANCER TREATMENT USING AN ANTI-CD19 CHEMERIC ANTIGEN RECEIVER
WO2015002969A1 (en) 2013-07-03 2015-01-08 City Of Hope Anticancer combinations
CN105452465B (en) 2013-07-31 2019-06-21 奇比艾企业有限公司 Poly- alkylamine-the oligonucleotide compound of sphingolipid-
WO2015015498A1 (en) 2013-07-31 2015-02-05 Qbi Enterprises Ltd. Methods of use of sphingolipid polyalkylamine oligonucleotide compounds
PT3030661T (en) * 2013-08-08 2019-06-06 Danisco Us Inc Increasing the expression of a transgene in eukaryotic cells by reducing rna interference
CA2920261C (en) 2013-08-08 2018-04-03 Global Bio Therapeutics, Inc. Clamp device for minimally invasive procedures and uses thereof
JP6694811B2 (en) 2013-10-04 2020-05-20 ノバルティス アーゲー 3'end cap for RNAi agents for use in RNA interference
WO2015051044A2 (en) 2013-10-04 2015-04-09 Novartis Ag Novel formats for organic compounds for use in rna interference
US10519446B2 (en) 2013-10-04 2019-12-31 Novartis Ag Organic compounds to treat hepatitis B virus
US9988627B2 (en) 2013-10-04 2018-06-05 Novartis Ag Formats for organic compounds for use in RNA interference
EP3055426B1 (en) 2013-10-09 2019-06-19 The United States of America as represented by The Secretary Department of Health and Human Services Detection of hepatitis delta virus (hdv) for the diagnosis and treatment of sjögren's syndrome and lymphoma
RU2016117978A (en) 2013-10-11 2017-11-17 Дженентек, Инк. NSP4 INHIBITORS AND WAYS OF THEIR APPLICATION
EP3068407A1 (en) 2013-11-11 2016-09-21 Sirna Therapeutics, Inc. Systemic delivery of myostatin short interfering nucleic acids (sina) conjugated to a lipophilic moiety
US10919929B2 (en) 2013-12-11 2021-02-16 Sloan-Kettering Institute For Cancer Research Glucocorticoid inhibitors for treatment of prostate cancer
US10426737B2 (en) 2013-12-19 2019-10-01 Novartis Ag Lipids and lipid compositions for the delivery of active agents
CN116478927A (en) 2013-12-19 2023-07-25 诺华股份有限公司 Human mesothelin chimeric antigen receptor and application thereof
EP3872066A1 (en) 2013-12-19 2021-09-01 Novartis AG Lipids and lipid compositions for the delivery of active agents
US9682123B2 (en) 2013-12-20 2017-06-20 The Trustees Of Columbia University In The City Of New York Methods of treating metabolic disease
US10287354B2 (en) 2013-12-20 2019-05-14 Novartis Ag Regulatable chimeric antigen receptor
US9274117B2 (en) 2013-12-21 2016-03-01 Catholic University Industry Academic Use of SIRT7 as novel cancer therapy target and method for treating cancer using the same
EP3088001B1 (en) 2013-12-27 2020-02-12 National University Corporation Kochi University Anti-lipolysis stimulated liporotein receptor antibodies for treatment of ovarian cancer
US11028143B2 (en) 2014-01-21 2021-06-08 Novartis Ag Enhanced antigen presenting ability of RNA CAR T cells by co-introduction of costimulatory molecules
JP2017513818A (en) 2014-03-15 2017-06-01 ノバルティス アーゲー Treatment of cancer using chimeric antigen receptors
ES2857226T3 (en) 2014-03-15 2021-09-28 Novartis Ag Regulable chimeric antigen receptor
JP2017509336A (en) 2014-03-20 2017-04-06 ファーマサイクリックス エルエルシー Mutations associated with phospholipase C gamma 2 and resistance
PL3129470T3 (en) 2014-04-07 2021-11-29 Novartis Ag Treatment of cancer using anti-cd19 chimeric antigen receptor
EP4008725A1 (en) 2014-05-02 2022-06-08 The Trustees of the University of Pennsylvania Compositions and methods of chimeric autoantibody receptor t cells
US10258639B2 (en) 2014-05-06 2019-04-16 Research Development Foundation Methods for treating insulin resistance and for sensitizing patients to GLP1 agonist therapy
KR101633876B1 (en) 2014-05-08 2016-06-28 고려대학교 산학협력단 REV-ERB Use of REV-ERB for treating affective and addictive disorders
KR101633881B1 (en) 2014-05-08 2016-06-28 고려대학교 산학협력단 REV-ERB Use of REV-ERB for treating dopamine-dependent disorders
US10342761B2 (en) 2014-07-16 2019-07-09 Novartis Ag Method of encapsulating a nucleic acid in a lipid nanoparticle host
WO2016011432A2 (en) 2014-07-17 2016-01-21 Czerniecki Brian J Identification of immunogenic mhc class ii peptides for immune-based therapy
US9777061B2 (en) 2014-07-21 2017-10-03 Novartis Ag Treatment of cancer using a CD33 chimeric antigen receptor
BR112017001183A2 (en) 2014-07-21 2017-11-28 Novartis Ag cancer treatment using humanized anti-bcma chimeric antigen receptor
WO2016014553A1 (en) 2014-07-21 2016-01-28 Novartis Ag Sortase synthesized chimeric antigen receptors
WO2016014530A1 (en) 2014-07-21 2016-01-28 Novartis Ag Combinations of low, immune enhancing. doses of mtor inhibitors and cars
EP3660042B1 (en) 2014-07-31 2023-01-11 Novartis AG Subset-optimized chimeric antigen receptor-containing t-cells
AU2015301460B2 (en) 2014-08-14 2021-04-08 Novartis Ag Treatment of cancer using GFR alpha-4 chimeric antigen receptor
CN112410363A (en) 2014-08-19 2021-02-26 诺华股份有限公司 anti-CD 123 Chimeric Antigen Receptor (CAR) for cancer therapy
CN107427552B (en) 2014-08-22 2022-12-20 广州英恩迈生物医药科技有限公司 Methods and compositions for the prevention and treatment of diseases or conditions associated with abnormal levels and/or activity of the IFP35 protein family
WO2016037053A1 (en) 2014-09-05 2016-03-10 Novartis Ag Lipids and lipid compositions for the delivery of active agents
DK3194443T3 (en) 2014-09-17 2021-09-27 Novartis Ag TARGETING OF CYTOTOXIC CELLS WITH CHIMARY RECEPTORS IN CONNECTION WITH ADOPTIVE IMMUNTERAPHY
RU2743657C2 (en) 2014-10-08 2021-02-20 Новартис Аг Biomarkers predicting a therapeutic response to therapy with a chimeric antigen receptor, and use thereof
CA2974716A1 (en) 2014-11-12 2016-05-19 Nmc, Inc. Transgenic plants with engineered redox sensitive modulation of photosynthetic antenna complex pigments and methods for making the same
US20180334490A1 (en) 2014-12-03 2018-11-22 Qilong H. Wu Methods for b cell preconditioning in car therapy
WO2016094342A1 (en) 2014-12-08 2016-06-16 The Board Of Regents Of The University Of Texas System Lipocationic polymers and uses thereof
US20190054117A1 (en) 2014-12-19 2019-02-21 Novartis Ag Dimerization switches and uses thereof
WO2016105517A1 (en) 2014-12-23 2016-06-30 The Trustees Of Columbia University In The City Of New York Fusion proteins and methods thereof
US10264976B2 (en) 2014-12-26 2019-04-23 The University Of Akron Biocompatible flavonoid compounds for organelle and cell imaging
WO2016109410A2 (en) 2014-12-29 2016-07-07 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
GB201501004D0 (en) 2015-01-21 2015-03-04 Cancer Rec Tech Ltd Inhibitors
WO2016126608A1 (en) 2015-02-02 2016-08-11 Novartis Ag Car-expressing cells against multiple tumor antigens and uses thereof
DK3253865T3 (en) 2015-02-06 2022-08-15 Nat Univ Singapore METHODS OF IMPROVING THE EFFECT OF THERAPEUTIC IMMUNE CELLS
KR20240018680A (en) 2015-02-27 2024-02-13 아이셀 진 테라퓨틱스 엘엘씨 Chimeric antigen receptors (cars) targeting hematologic malignancies, compositions and methods of use thereof
MA41629A (en) 2015-03-04 2018-01-09 Center For Human Reproduction COMPOSITIONS AND METHODS OF USE OF ANTI-MÜLLERIAN HORMONE FOR THE TREATMENT OF INFERTILITY
US11085044B2 (en) 2015-03-09 2021-08-10 University Of Kentucky Research Foundation miRNA for treatment of breast cancer
CN111961103B (en) 2015-03-09 2023-06-16 肯塔基大学研究基金会 RNA nanoparticles for brain tumor treatment
WO2016145003A1 (en) 2015-03-09 2016-09-15 University Of Kentucky Research Foundation Rna nanoparticle for treatment of gastric cancer
WO2016145578A1 (en) 2015-03-13 2016-09-22 Syz Cell Therapy Co. Methods of cancer treatment using activated t cells
KR101797569B1 (en) 2015-03-18 2017-11-22 한국교통대학교산학협력단 Liver Targeting Metal Nano-particle Based Nucleic Acid Delivery System And Manufacturing Method Thereof
US11279768B1 (en) 2015-04-03 2022-03-22 Precision Biologics, Inc. Anti-cancer antibodies, combination therapies, and uses thereof
JP6903587B2 (en) 2015-04-03 2021-07-14 ユーリカ セラピューティックス, インコーポレイテッド Constructs targeting AFP peptide / MHC complexes and their use
EP3280432B8 (en) 2015-04-06 2021-04-14 Subdomain, LLC De novo binding domain containing polypeptides and uses thereof
US20180140602A1 (en) 2015-04-07 2018-05-24 Novartis Ag Combination of chimeric antigen receptor therapy and amino pyrimidine derivatives
RU2021121771A (en) 2015-04-08 2022-01-12 Новартис Аг CD20 THERAPY, CD22 THERAPY AND COMBINATION THERAPY WITH CELLS EXPRESSING CHIMERIC CD19 ANTIGEN RECEPTOR (CAR)
AU2016249005B2 (en) 2015-04-17 2022-06-16 Novartis Ag Methods for improving the efficacy and expansion of chimeric antigen receptor-expressing cells
US10738290B2 (en) 2015-04-21 2020-08-11 Novartis Ag RNA-guided gene editing system and uses thereof
EP3286211A1 (en) 2015-04-23 2018-02-28 Novartis AG Treatment of cancer using chimeric antigen receptor and protein kinase a blocker
US20180156807A1 (en) 2015-04-29 2018-06-07 New York University Method for treating high-grade gliomas
WO2016174652A1 (en) 2015-04-30 2016-11-03 Technion Research & Development Foundation Limited Chimeric antigen receptors and methods of their use
RU2754661C2 (en) 2015-05-01 2021-09-06 Дзе Реджентс Оф Дзе Юниверсити Оф Калифорния Glycan-dependent immunotherapy molecules
PT3298033T (en) 2015-05-18 2020-09-22 Tcr2 Therapeutics Inc Compositions and methods for tcr reprogramming using fusion proteins
JP2018518491A (en) 2015-06-12 2018-07-12 アレクトル エルエルシー Anti-CD33 antibody and method of use thereof
CA2988982A1 (en) 2015-06-12 2016-12-15 Alector Llc Anti-cd33 antibodies and methods of use thereof
EP3313420B1 (en) 2015-06-25 2024-03-13 The Children's Medical Center Corporation Methods and compositions relating to hematopoietic stem cell expansion, enrichment, and maintenance
CN107849112B (en) 2015-06-25 2022-04-01 美商生物细胞基因治疗有限公司 Chimeric Antigen Receptors (CAR), compositions and methods of use thereof
WO2017009853A1 (en) 2015-07-16 2017-01-19 Yeda Research And Development Co. Ltd. Genetically modified anti-third party central memory t cells and use of same in immunotherapy
AU2016297014B2 (en) 2015-07-21 2021-06-17 Novartis Ag Methods for improving the efficacy and expansion of immune cells
US10072065B2 (en) 2015-08-24 2018-09-11 Mayo Foundation For Medical Education And Research Peptide-mediated delivery of immunoglobulins across the blood-brain barrier
AU2016316768A1 (en) 2015-08-28 2018-03-29 Alector Llc Anti-Siglec-7 antibodies and methods of use thereof
US11649435B2 (en) 2015-08-28 2023-05-16 The Trustees Of The University Of Pennsylvania Methods and compositions for cells expressing a chimeric intracellular signaling molecule
EP3344996A2 (en) 2015-09-03 2018-07-11 The Trustees Of The University Of Pennsylvania Biomarkers predictive of cytokine release syndrome
AU2016321298A1 (en) 2015-09-09 2018-03-29 The Trustees Of Columbia University In The City Of New York Reduction of ER-MAM localized APP-C99 and methods of treating Alzheimer's Disease
US11247968B2 (en) 2015-09-14 2022-02-15 The Board Of Regents Of The University Of Texas System Lipocationic dendrimers and uses thereof
CA2999496A1 (en) 2015-09-22 2017-03-30 The Trustees Of The University Of Pennsylvania Method of redirecting t cells to treat hiv infection
US11285142B2 (en) 2015-09-29 2022-03-29 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Compositions and methods for identifying and treating dystonia disorders
CN116059316A (en) 2015-10-23 2023-05-05 优瑞科生物技术公司 antibody/T cell receptor chimeric constructs and uses thereof
CA3003458A1 (en) 2015-10-29 2017-05-04 Alector Llc Anti-siglec-9 antibodies and methods of use thereof
WO2017075421A1 (en) 2015-10-29 2017-05-04 Temple University-Of The Commonwealth System Of Higher Education Modification of 3' terminal ends of nucleic acids by dna polymerase theta
MA43135A (en) 2015-10-30 2018-09-05 Aleta Biotherapeutics Inc COMPOSITIONS AND METHODS FOR THE TREATMENT OF CANCER
NZ741780A (en) 2015-10-30 2019-11-29 Genentech Inc Anti-htra1 antibodies and methods of use thereof
CA3003144A1 (en) 2015-10-30 2017-05-04 Aleta Biotherapeutics Inc. Compositions and methods for tumor transduction
CN109152824B (en) 2015-11-27 2022-12-06 卡瑟里克斯私人有限公司 Genetically modified cells and uses thereof
JP6983779B2 (en) 2015-11-30 2021-12-17 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニアThe Regents Of The University Of California Tumor-specific payload delivery and immune activation using human antibodies targeting highly specific tumor cell surface antigens
ES2865030T3 (en) 2015-12-13 2021-10-14 Nitto Denko Corp SiRNA structures for high activity and low nonspecificity
GB201522186D0 (en) 2015-12-16 2016-01-27 Singapore Health Services Pte Ltd And Nat University Of Singapore The Treatment of fibrosis
US11413340B2 (en) 2015-12-22 2022-08-16 Novartis Ag Mesothelin chimeric antigen receptor (CAR) and antibody against PD-L1 inhibitor for combined use in anticancer therapy
CN117025539A (en) 2015-12-28 2023-11-10 诺华股份有限公司 Method for preparing chimeric antigen receptor expressing cells
IL260369B2 (en) 2016-01-06 2024-03-01 Lonza Ag Inhibition of protein degradation for improved production
US11441146B2 (en) 2016-01-11 2022-09-13 Christiana Care Health Services, Inc. Compositions and methods for improving homogeneity of DNA generated using a CRISPR/Cas9 cleavage system
EP3423568A4 (en) 2016-03-04 2019-11-13 University Of Louisville Research Foundation, Inc. Methods and compositions for ex vivo expansion of very small embryonic-like stem cells (vsels)
MX2018010733A (en) 2016-03-04 2019-07-04 Novartis Ag Cells expressing multiple chimeric antigen receptor (car) molecules and uses therefore.
US10689873B2 (en) 2016-03-10 2020-06-23 Lonza Ltd Customizable facility
US11236520B2 (en) 2016-03-10 2022-02-01 Lonza Ltd Customizable facility
US20190119642A1 (en) 2016-03-15 2019-04-25 Children's Medical Center Corporation Methods and compositions relating to hematopoietic stem cell expansion
EP3436070A4 (en) 2016-03-29 2019-11-27 University of Southern California Chimeric antigen receptors targeting cancer
US10883108B2 (en) 2016-03-31 2021-01-05 The Schepens Eye Research Institute, Inc. Endomucin inhibitor as an anti-angiogenic agent
KR102408260B1 (en) 2016-04-14 2022-06-10 론자 리미티드 Compositions and Methods for Detection of Host Cell Proteins
KR20180134385A (en) 2016-04-15 2018-12-18 노파르티스 아게 Compositions and methods for selective protein expression
CN109328230A (en) 2016-04-15 2019-02-12 宾夕法尼亚大学董事会 The composition and method of chimeric alloantigen recipient T cells
US20190119364A1 (en) 2016-04-29 2019-04-25 The Regents Of The University Of Colorado, A Body Corporate Compounds and compositions useful for treating metabolic syndrome, and methods using same
IL262646B (en) 2016-05-03 2022-08-01 Lonza Ag Modulation of lipid metabolism for protein production
MA44740A (en) 2016-05-13 2019-02-27 4D Molecular Therapeutics Inc ADENO-ASSOCIATED VIRUS CAPSIDE VARIANTS AND THEIR METHODS OF USE
CN114656378A (en) 2016-05-16 2022-06-24 得克萨斯州大学系统董事会 Cationic sulfonamide amino lipids and amphiphilic zwitterionic amino lipids
WO2017210617A2 (en) 2016-06-02 2017-12-07 Porter, David, L. Therapeutic regimens for chimeric antigen receptor (car)- expressing cells
JP2019517267A (en) 2016-06-08 2019-06-24 イントレキソン コーポレーション CD33-specific chimeric antigen receptor
JP7219376B2 (en) 2016-07-15 2023-02-08 ノバルティス アーゲー Treatment and prevention of cytokine release syndrome using chimeric antigen receptors in combination with kinase inhibitors
JP7178568B2 (en) 2016-07-18 2022-11-28 ナショナル リサーチ カウンシル オブ カナダ CAR immune cells targeting carcinoembryonic antigen-related cell adhesion molecule 6 for treating cancer
KR20190034588A (en) 2016-07-28 2019-04-02 노파르티스 아게 Combination therapy of chimeric antigen receptor and PD-1 inhibitor
SG11201900885VA (en) 2016-08-01 2019-02-27 Novartis Ag Treatment of cancer using a chimeric antigen receptor in combination with an inhibitor of a pro-m2 macrophage molecule
JP2019525765A (en) 2016-08-02 2019-09-12 ロンザ リミテッドLonza Limited Customizable facilities
AU2017306432A1 (en) 2016-08-02 2019-03-21 TCR2 Therapeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
WO2018031858A1 (en) 2016-08-12 2018-02-15 Lonza Ltd Proteomic analysis of host cell proteins
EP3516068A4 (en) 2016-09-19 2020-10-14 University of Southern California Non-radioactive cytotoxicity assays
EP3516062A1 (en) 2016-09-21 2019-07-31 Alnylam Pharmaceuticals, Inc. Myostatin irna compositions and methods of use thereof
EP3757120B8 (en) 2016-10-04 2022-06-15 Precision Biosciences, Inc. Co-stimulatory domains for use in genetically-modified cells
EP3445787B1 (en) 2016-10-07 2020-12-02 TCR2 Therapeutics Inc. Compositions and methods for t-cell receptors reprogramming using fusion proteins
AU2017341047A1 (en) 2016-10-07 2019-05-02 Novartis Ag Chimeric antigen receptors for the treatment of cancer
EP3535396A1 (en) 2016-11-01 2019-09-11 Novartis AG Methods and compositions for enhancing gene editing
US10550183B2 (en) 2016-11-22 2020-02-04 National University Of Singapore Blockade of CD7 expression and chimeric antigen receptors for immunotherapy of T-cell malignancies
AU2017363311A1 (en) 2016-11-22 2019-06-13 TCR2 Therapeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
US11147249B2 (en) 2016-12-08 2021-10-19 Alector Llc Siglec transgenic mice and methods of use thereof
WO2018119298A1 (en) 2016-12-21 2018-06-28 TCR2 Therapeutics Inc. Engineered t cells for the treatment of cancer
WO2018127585A1 (en) 2017-01-06 2018-07-12 Txcell Monospecific regulatory t cell population with cytotoxicity for b cells
EP3346001A1 (en) 2017-01-06 2018-07-11 TXCell Monospecific regulatory t cell population with cytotoxicity for b cells
KR20190102259A (en) 2017-01-10 2019-09-03 인트렉손 코포레이션 Regulation of Polypeptide Expression Through a Novel Gene Switch Expression System
US20190359973A1 (en) 2017-01-10 2019-11-28 Christiana Care Health Services, Inc. Methods for in vitro site-directed mutagenesis using gene editing technologies
US11274157B2 (en) 2017-01-12 2022-03-15 Eureka Therapeutics, Inc. Constructs targeting histone H3 peptide/MHC complexes and uses thereof
EP3571295A1 (en) 2017-01-18 2019-11-27 Yeda Research and Development Co. Ltd Genetically modified veto cells and use of same in immunotherapy
WO2018140725A1 (en) 2017-01-26 2018-08-02 Novartis Ag Cd28 compositions and methods for chimeric antigen receptor therapy
CN108395482B (en) 2017-02-08 2021-02-05 西比曼生物科技(香港)有限公司 Construction of targeting CD20 antigen chimeric antigen receptor and activity identification of engineered T cell thereof
US11597911B2 (en) 2017-02-27 2023-03-07 Life Technologies Corporation Expansion of populations of T cells by the use of modified serum free media
WO2018191490A1 (en) 2017-04-13 2018-10-18 The Trustees Of The University Of Pennsylvania Use of gene editing to generate universal tcr re-directed t cells for adoptive immunotherapy
WO2018200586A1 (en) 2017-04-26 2018-11-01 Eureka Therapeutics, Inc. Constructs specifically recognizing glypican 3 and uses thereof
EP3615046A4 (en) 2017-04-26 2021-03-03 Eureka Therapeutics, Inc. Cells expressing chimeric activating receptors and chimeric stimulating receptors and uses thereof
EP3615055A1 (en) 2017-04-28 2020-03-04 Novartis AG Cells expressing a bcma-targeting chimeric antigen receptor, and combination therapy with a gamma secretase inhibitor
EP3634493A1 (en) 2017-05-08 2020-04-15 Precision BioSciences, Inc. Nucleic acid molecules encoding an engineered antigen receptor and an inhibitory nucleic acid molecule and methods of use thereof
WO2018213316A1 (en) 2017-05-16 2018-11-22 Alector Llc Anti-siglec-5 antibodies and methods of use thereof
US20210079057A1 (en) 2017-06-13 2021-03-18 TCR2 Therapeutics Inc. Compositions and methods for tcr reprogramming using fusion proteins
EP3622072A1 (en) 2017-06-16 2020-03-18 Lonza Ltd Universal self-regulating mammalian cell line platform for the production of biologics
WO2019007991A1 (en) 2017-07-03 2019-01-10 Universite De Strasbourg Mtmr2-s polypeptide for use in the treatment of myopathies
CA3057447A1 (en) 2017-08-03 2019-02-07 Alector Llc Anti-cd33 antibodies and methods of use thereof
EP3444347A1 (en) 2017-08-18 2019-02-20 Universität Heidelberg Use of anti-crispr polypeptides for specific activation of cas nucleases
CN109456943A (en) 2017-09-06 2019-03-12 亘喜生物科技(上海)有限公司 Universal Chimeric antigen receptor T cell technology of preparing
KR20200044973A (en) 2017-09-15 2020-04-29 라이프 테크놀로지스 코포레이션 Compositions and methods for culturing and expanding cells
WO2019056106A1 (en) 2017-09-20 2019-03-28 The University Of British Columbia Novel anti-hla-a2 antibodies and uses thereof
CN111836649A (en) 2017-09-20 2020-10-27 4D分子治疗有限公司 Adeno-associated virus variant capsids and methods of use thereof
CN109554348A (en) 2017-09-27 2019-04-02 亘喜生物科技(上海)有限公司 It can induce the engineering immunocyte of secretion anti-cd 47 antibody
EP4269560A3 (en) 2017-10-03 2024-01-17 Precision Biosciences, Inc. Modified epidermal growth factor receptor peptides for use in genetically-modified cells
CA3078460A1 (en) 2017-10-04 2019-04-11 Opko Pharmaceuticals, Llc Articles and methods directed to personalized therapy of cancer
EP3692058A1 (en) 2017-10-06 2020-08-12 Oslo Universitetssykehus HF Chimeric antigen receptors
KR20200069358A (en) 2017-10-25 2020-06-16 노파르티스 아게 Method for producing chimeric antigen receptor expressing cells
WO2019089798A1 (en) 2017-10-31 2019-05-09 Novartis Ag Anti-car compositions and methods
US11666628B2 (en) 2017-11-11 2023-06-06 Universite De Strasbourg Compositions and method for the treatment of x-linked centronuclear myopathy
WO2019099433A2 (en) 2017-11-14 2019-05-23 Arcellx, Inc. D-domain containing polypeptides and uses thereof
CN111511383A (en) 2017-11-14 2020-08-07 阿奇利克斯股份有限公司 Multifunctional immune cell therapy
SG11202004512XA (en) 2017-11-15 2020-06-29 Novartis Ag Bcma-targeting chimeric antigen receptor, cd19-targeting chimeric antigen receptor, and combination therapies
CA3083472A1 (en) 2017-11-27 2019-05-31 4D Molecular Therapeutics Inc. Adeno-associated virus variant capsids and use for inhibiting angiogenesis
AU2018375738A1 (en) 2017-11-30 2020-06-11 Novartis Ag BCMA-targeting chimeric antigen receptor, and uses thereof
CA3083236A1 (en) 2017-12-05 2019-06-13 Lonza Ltd Methods of assaying tropolone
US11470827B2 (en) 2017-12-12 2022-10-18 Alector Llc Transgenic mice expressing human TREM proteins and methods of use thereof
CA3086267A1 (en) 2017-12-29 2019-07-04 Cellectis Method for improving production of car t cells
EP3737408A1 (en) 2018-01-08 2020-11-18 Novartis AG Immune-enhancing rnas for combination with chimeric antigen receptor therapy
GB201801067D0 (en) 2018-01-23 2018-03-07 Price Nicola Kaye Biomarkers predictive of tumour infiltrating lymphocyte therapy and uses thereof
CA3090249A1 (en) 2018-01-31 2019-08-08 Novartis Ag Combination therapy using a chimeric antigen receptor
GB2577236A (en) 2018-02-02 2020-03-18 Lonza Ag Methods of cell selection and modifying cell metabolism
EP3752532A1 (en) 2018-02-12 2020-12-23 Diabetes-Free, Inc. Improved antagonistic anti-human cd40 monoclonal antibodies
WO2019160956A1 (en) 2018-02-13 2019-08-22 Novartis Ag Chimeric antigen receptor therapy in combination with il-15r and il15
MX2020009514A (en) 2018-03-14 2020-12-07 Beijing Xuanyi Pharmasciences Co Ltd Anti-claudin 18.2 antibodies.
SG11202009697RA (en) 2018-03-30 2020-10-29 Univ Geneve Micro rna expression constructs and uses thereof
WO2019183924A1 (en) 2018-03-30 2019-10-03 Syz Cell Therapy Co. Improved multiple antigen specific cell therapy methods
US20210162007A1 (en) 2018-04-09 2021-06-03 President And Fellows Of Harvard College Modulating nuclear receptors and methods of using same
CN116836297A (en) 2018-04-12 2023-10-03 上海赛比曼生物科技有限公司 Chimeric antigen receptor targeting BCMA and preparation method and application thereof
WO2019196088A1 (en) 2018-04-13 2019-10-17 Syz Cell Therapy Co. Methods of obtaining tumor-specific t cell receptors
WO2019196087A1 (en) 2018-04-13 2019-10-17 Syz Cell Therapy Co. Methods of cancer treatment using tumor antigen-specific t cells
AU2019250692A1 (en) 2018-04-13 2020-11-05 Sangamo Therapeutics France Chimeric antigen receptor specific for Interleukin-23 receptor
EP3784351A1 (en) 2018-04-27 2021-03-03 Novartis AG Car t cell therapies with enhanced efficacy
WO2019213180A1 (en) 2018-04-30 2019-11-07 Amicus Therapeutics, Inc. Gene therapy constructs and methods of use
EP3788369A1 (en) 2018-05-01 2021-03-10 Novartis Ag Biomarkers for evaluating car-t cells to predict clinical outcome
CN112424342A (en) 2018-05-08 2021-02-26 生命科技公司 Compositions and methods for culturing and expanding cells
WO2019227003A1 (en) 2018-05-25 2019-11-28 Novartis Ag Combination therapy with chimeric antigen receptor (car) therapies
GB201808927D0 (en) 2018-05-31 2018-07-18 Institute Of Cancer Res Royal Cancer Hospital Materials and methods for monitoring the development of resistance of cancers to treatment
CN112203725A (en) 2018-06-13 2021-01-08 诺华股份有限公司 BCMA chimeric antigen receptors and uses thereof
WO2019241549A1 (en) 2018-06-15 2019-12-19 A2 Biotherapeutics, Inc. Foxp3-expressing car-t regulatory cells
GB201810181D0 (en) 2018-06-21 2018-08-08 Immetacyte Ltd Cells expressing chimeric recominant growth factor receptors
EA202190183A1 (en) 2018-07-27 2021-05-18 Алектор Ллс ANTIBODIES TO SIGLEC-5 AND METHODS OF THEIR APPLICATION
MX2021001592A (en) 2018-08-10 2021-09-21 Sangamo Therapeutics France New car constructs comprising tnfr2 domains.
CA3110565A1 (en) 2018-08-30 2020-03-05 TCR2 Therapeutics Inc. Compositions and methods for tcr reprogramming using fusion proteins
KR20210112295A (en) 2018-08-31 2021-09-14 잉벡띠스 에스아 Chimeric antigen receptors for multiple HLA-G isoforms
CN112639083A (en) 2018-08-31 2021-04-09 诺华股份有限公司 Method for producing cells expressing chimeric antigen receptor
WO2020047449A2 (en) 2018-08-31 2020-03-05 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
WO2020061381A1 (en) 2018-09-19 2020-03-26 La Jolla Institute For Immunology Ptprs and proteoglycans in rheumatoid arthritis
CN110950953B (en) 2018-09-26 2022-05-13 福州拓新天成生物科技有限公司 Monoclonal antibody against B7-H3 and application thereof in cell therapy
US20220047633A1 (en) 2018-09-28 2022-02-17 Novartis Ag Cd22 chimeric antigen receptor (car) therapies
US20210347851A1 (en) 2018-09-28 2021-11-11 Novartis Ag Cd19 chimeric antigen receptor (car) and cd22 car combination therapies
EP3863663A2 (en) 2018-10-10 2021-08-18 Amicus Therapeutics, Inc. Disulfide bond stabilized polypeptide compositions and methods of use
US20210387020A1 (en) 2018-10-19 2021-12-16 University Of Rochester Immune modulators in combination with radiation treatment for advanced pancreatic cancer
JP2022513376A (en) 2018-10-22 2022-02-07 ユニバーシティ オブ ロチェスター Genome editing by directional non-homologous DNA insertion using retrovirus integrase-Cas9 fusion protein
US20220170097A1 (en) 2018-10-29 2022-06-02 The Broad Institute, Inc. Car t cell transcriptional atlas
WO2020102454A1 (en) 2018-11-13 2020-05-22 Regents Of The University Of Minnesota Cd40 targeted peptides and uses thereof
WO2020102555A1 (en) 2018-11-16 2020-05-22 Memorial Sloan Kettering Cancer Center Antibodies to mucin-16 and methods of use thereof
TW202031271A (en) 2018-11-16 2020-09-01 日商日東電工股份有限公司 Rna interference delivery formulation and methods for malignant tumors
CA3119338A1 (en) 2018-11-16 2020-05-22 Celgene Corporation Improved t cell manufacturing process
WO2020140007A1 (en) 2018-12-28 2020-07-02 University Of Rochester Gene therapy for best1 dominant mutations
US11883401B2 (en) 2019-01-08 2024-01-30 Duke University Compositions for the treatment of pathogenic- and/or chemical-induced lung injury and for the treatment of cancer and methods of using same
MX2021008487A (en) 2019-01-14 2021-11-12 Univ Rochester Targeted nuclear rna cleavage and polyadenylation with crispr-cas.
KR20210138579A (en) 2019-01-21 2021-11-19 싱가포르 헬스 서비시즈 피티이 엘티디 Treatment of hepatotoxicity
GB201900858D0 (en) 2019-01-22 2019-03-13 Price Nicola Kaye Receptors providing targeted costimulation for adoptive cell therapy
US20220184119A1 (en) 2019-02-08 2022-06-16 Biontech Cell & Gene Therapies Gmbh Chimeric antigen receptor-modified cells for the treatment of cldn6-expressing cancers
US20220136011A1 (en) 2019-02-08 2022-05-05 Board Of Regents, The University Of Texas System Telomerase-containing exosomes for treatment of diseases associated with aging and age-related organ dysfunction
US20220088075A1 (en) 2019-02-22 2022-03-24 The Trustees Of The University Of Pennsylvania Combination therapies of egfrviii chimeric antigen receptors and pd-1 inhibitors
GB201902419D0 (en) 2019-02-22 2019-04-10 Singapore Health Serv Pte Ltd Treatment of kidney injury
RU2742000C2 (en) 2019-03-13 2021-02-01 Общество С Ограниченной Ответственностью "Анабион" Isolated alternative intracellular signalling domain of chimeric antigen receptor and chimeric antigen receptor including it
JP7379654B2 (en) 2019-03-15 2023-11-14 カーティザン セラピューティクス,インコーポレーテッド Anti-BCMA chimeric antigen receptor
US20220184176A1 (en) 2019-03-20 2022-06-16 Centre National De La Recherche Scientifique Amphiphysin / bin1 for the treatment of autosomal dominant centronuclear myopathy
JP2022527297A (en) 2019-03-27 2022-06-01 ザ トラスティーズ オブ ザ ユニバーシティ オブ ペンシルバニア Tn-MUC1 Chimeric Antigen Receptor (CAR) T Cell Therapy
CN111793627A (en) 2019-04-08 2020-10-20 中国科学院上海生命科学研究院 RNA fixed-point editing by utilizing artificially constructed RNA editing enzyme and related application
US20220168389A1 (en) 2019-04-12 2022-06-02 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
US20220251152A1 (en) 2019-04-24 2022-08-11 Novartis Ag Compositions and methods for selective protein degradation
JP2022531591A (en) 2019-05-03 2022-07-07 シンガポール・ヘルス・サービシーズ・ピーティーイー・リミテッド Treatment and prevention of metabolic disorders
GB201906291D0 (en) 2019-05-03 2019-06-19 Singapore Health Serv Pte Ltd Treatment and prevention of metabolic diseases
US20220202864A1 (en) 2019-05-07 2022-06-30 Gracell Biotechnologies (Shanghai) Co., Ltd. Bcma-targeting engineered immune cell and use thereof
EP3967329A4 (en) 2019-05-07 2023-06-14 Gracell Biotechnologies (Shanghai) Co., Ltd. Engineered immune cell targeting bcma and use thereof
CA3145662A1 (en) 2019-07-02 2021-01-07 M6P Therapeutics Vector compositions and methods of using same for treatment of lysosomal storage disorders
JP2022544015A (en) 2019-07-23 2022-10-17 ユニバーシティ オブ ロチェスター Targeted RNA Cleavage with CRISPR-Cas
US20220273715A1 (en) 2019-07-25 2022-09-01 Precision Biosciences, Inc. Compositions and methods for sequential stacking of nucleic acid sequences into a genomic locus
CN112300997A (en) 2019-08-01 2021-02-02 上海赛比曼生物科技有限公司 Universal CAR-T cell and preparation and application thereof
WO2021030182A1 (en) 2019-08-09 2021-02-18 A2 Biotherapeutics, Inc. Bifunctional single variable domain t cell receptors and uses thereof
MX2022001781A (en) 2019-08-19 2022-03-11 Univ Basel Cell therapy methods.
CA3148179A1 (en) 2019-08-20 2021-02-25 Bruce J. Mccreedy Jr. Lymphodepletion dosing regimens for cellular immunotherapies
WO2021035170A1 (en) 2019-08-21 2021-02-25 Precision Biosciences, Inc. Compositions and methods for tcr reprogramming using fusion proteins
GB201913030D0 (en) 2019-09-10 2019-10-23 Francis Crick Institute Ltd Treatment of hr deficient cancer
KR102100163B1 (en) 2019-09-24 2020-04-13 테고사이언스 (주) Compositions of Prevention or Treatment of Keloid or Hypertrophic scar
EP3808766A1 (en) 2019-10-15 2021-04-21 Sangamo Therapeutics France Chimeric antigen receptor specific for interleukin-23 receptor
US20220389075A1 (en) 2019-11-12 2022-12-08 A2 Biotherapeutics, Inc. Engineered t cell receptors and uses thereof
IL292890A (en) 2019-11-14 2022-07-01 Lonza Ag Methods of cell selection
CN110760007B (en) 2019-11-21 2022-08-26 博生吉医药科技(苏州)有限公司 CD7-CAR-T cell and preparation and application thereof
TW202134285A (en) 2019-11-26 2021-09-16 瑞士商諾華公司 Cd19 and cd22 chimeric antigen receptors and uses thereof
TW202134264A (en) 2019-11-26 2021-09-16 瑞士商諾華公司 Chimeric antigen receptors and uses thereof
EP4073103A1 (en) 2019-12-11 2022-10-19 A2 Biotherapeutics, Inc. Lilrb1-based chimeric antigen receptor
KR20220114065A (en) 2019-12-18 2022-08-17 노파르티스 아게 3-(5-Methoxy-1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
JP2023506842A (en) 2019-12-18 2023-02-20 ノバルティス アーゲー Compositions and methods for treating hemoglobinopathies
EP4087652A1 (en) 2020-01-08 2022-11-16 Regeneron Pharmaceuticals, Inc. Treatment of fibrodysplasia ossificans progressiva
WO2021150300A1 (en) 2020-01-22 2021-07-29 Massachusetts Institute Of Technology Inducible tissue constructs and uses thereof
EP4100533A1 (en) 2020-02-07 2022-12-14 University of Rochester Ribozyme-mediated rna assembly and expression
JP2023513211A (en) 2020-02-07 2023-03-30 ユニバーシティ オブ ロチェスター Target RNA translation by CRISPR-Cas13 to enhance protein synthesis
WO2021163618A1 (en) 2020-02-14 2021-08-19 Novartis Ag Method of predicting response to chimeric antigen receptor therapy
WO2021173674A1 (en) 2020-02-26 2021-09-02 A2 Biotherapeutics, Inc. Polypeptides targeting mage-a3 peptide-mhc complexes and methods of use thereof
JP2023515211A (en) 2020-02-27 2023-04-12 ノバルティス アーゲー Method for producing chimeric antigen receptor-expressing cells
KR20220146530A (en) 2020-02-27 2022-11-01 노파르티스 아게 Methods of Making Chimeric Antigen Receptor-Expressing Cells
KR20220145859A (en) 2020-02-28 2022-10-31 상하이 헨리우스 바이오테크, 인크. Anti-CD137 constructs, multispecific antibodies and uses thereof
US11642407B2 (en) 2020-02-28 2023-05-09 Massachusetts Institute Of Technology Identification of variable influenza residues and uses thereof
KR20220148209A (en) 2020-02-28 2022-11-04 상하이 헨리우스 바이오테크, 인크. Anti-CD137 constructs and uses thereof
AU2021237818A1 (en) 2020-03-19 2022-09-29 Trizell Ltd. Temperature-responsive virus storage system
MX2022011683A (en) 2020-03-20 2022-12-08 Inst Nat Sante Rech Med Chimeric antigen receptor specific for human cd45rc and uses thereof.
US20230103771A1 (en) 2020-03-27 2023-04-06 University Of Rochester CRISPR-Cas13 crRNA Arrays
WO2021195519A1 (en) 2020-03-27 2021-09-30 University Of Rochester Targeted destruction of viral rna by crispr-cas13
CN113527507A (en) 2020-04-16 2021-10-22 上海赛比曼生物科技有限公司 Chimeric antigen receptor targeting CD22 and preparation method and application thereof
US20220023204A1 (en) 2020-04-20 2022-01-27 Board Of Regents, The University Of Texas System Biologically active dry powder compositions and method of their manufacture and use
WO2021222576A1 (en) 2020-05-01 2021-11-04 A2 Biotherapeutics, Inc. Pag1 fusion proteins and methods of making and using same
IL298558A (en) 2020-05-27 2023-01-01 Antion Biosciences Sa Adapter molecules to re-direct car t cells to an antigen of interest
US20230220057A1 (en) 2020-05-27 2023-07-13 Staidson (Beijing) Biopharmaceuticals Co., Ltd. Antibodies specifically recognizing nerve growth factor and uses thereof
EP4165169A1 (en) 2020-06-11 2023-04-19 Novartis AG Zbtb32 inhibitors and uses thereof
WO2021252635A1 (en) 2020-06-11 2021-12-16 A2 Biotherapeutics, Inc. Compositions and methods for treating cancers
GB202009292D0 (en) 2020-06-18 2020-08-05 Singapore Health Serv Pte Ltd Treatment and prevention of disease caused by type IV collagen dysfunction
WO2022006286A1 (en) 2020-06-30 2022-01-06 Lunglife Ai Methods for detecting lung cancer
CA3185589A1 (en) 2020-07-15 2022-01-20 Douglas Matthew ANDERSON Targeted rna cleavage with dcasl3-rnase fusion proteins
JP2023534055A (en) 2020-07-16 2023-08-07 上海交通大学 Immunotherapy targeting chemokine and cytokine delivery by mesenchymal stem cells
KR20230088333A (en) 2020-07-17 2023-06-19 인스틸 바이오 유케이 리미티드 Receptors that provide targeted costimulation for adoptive cell therapy
US20230277670A1 (en) 2020-07-17 2023-09-07 John Bridgeman Chimeric molecules providing targeted costimulation for adoptive cell therapy
WO2022036068A1 (en) 2020-08-13 2022-02-17 A2 Biotherapeutics, Inc. Gene fusions for control of genetically modified cells
KR20230052289A (en) 2020-08-20 2023-04-19 에이투 바이오쎄라퓨틱스, 인크. Compositions and methods for treating EGFR positive cancer
CA3188862A1 (en) 2020-08-20 2022-02-24 Carl Alexander Kamb Compositions and methods for treating mesothelin positive cancers
AU2021329375A1 (en) 2020-08-20 2023-04-20 A2 Biotherapeutics, Inc. Compositions and methods for treating ceacam positive cancers
WO2022079308A1 (en) 2020-10-16 2022-04-21 Institut Pasteur Chimeric constructs useful in vaccination and cancer therapy
GB202017244D0 (en) 2020-10-30 2020-12-16 Nat Univ Singapore Methods to extend health-span and treat age-related diseases
KR20230107617A (en) 2020-11-13 2023-07-17 노파르티스 아게 Combination therapy using chimeric antigen receptor (CAR)-expressing cells
CN114525306A (en) 2020-11-23 2022-05-24 博生吉医药科技(苏州)有限公司 Preparation method and application of CD7-CAR-T cell
TW202237639A (en) 2020-12-09 2022-10-01 日商武田藥品工業股份有限公司 Compositions of guanylyl cyclase c (gcc) antigen binding agents and methods of use thereof
TW202237638A (en) 2020-12-09 2022-10-01 日商武田藥品工業股份有限公司 Compositions of guanylyl cyclase c (gcc) antigen binding agents and methods of use thereof
WO2022147481A1 (en) 2020-12-30 2022-07-07 Ansun Biopharma Inc. Combination therapy of an oncolytic virus delivering a foreign antigen and an engineered immune cell expressing a chimeric receptor targeting the foreign antigen
EP4271482A2 (en) 2020-12-31 2023-11-08 Alamar Biosciences, Inc. Binder molecules with high affinity and/ or specificity and methods of making and use thereof
CN114763381A (en) 2021-01-13 2022-07-19 博生吉医药科技(苏州)有限公司 B7-H3 chimeric antigen receptor modified T cell and application thereof
CA3204730A1 (en) 2021-01-15 2022-07-21 Gowrishankar MUTHUKRISHNAN Staphylococcus aureus antigen-based nucleic acid vaccines
WO2022165111A1 (en) 2021-01-28 2022-08-04 Precision Biosciences, Inc. Modulation of tgf beta signaling in genetically-modified eukaryotic cells
GB2603166A (en) 2021-01-29 2022-08-03 Thelper As Therapeutic and Diagnostic Agents and Uses Thereof
CN114907485A (en) 2021-02-08 2022-08-16 浙江大学 Chimeric antigen receptor with endogenous protein molecule replacing single domain antibody
IL304857A (en) 2021-02-16 2023-10-01 A2 Biotherapeutics Inc Compositions and methods for treating her2 positive cancers
CA3216146A1 (en) 2021-04-23 2022-10-27 Douglas Anderson Genome editing by directed non-homologous dna insertion using a retroviral integrase-cas fusion protein and methods of treatment
TW202313096A (en) 2021-05-28 2023-04-01 大陸商江蘇恆瑞醫藥股份有限公司 Recombinant adeno-associated virus with capsid variant and its application
WO2022254337A1 (en) 2021-06-01 2022-12-08 Novartis Ag Cd19 and cd22 chimeric antigen receptors and uses thereof
CN115477705B (en) 2021-06-16 2024-02-23 四川大学华西医院 Preparation and application of chimeric antigen receptor immune cells constructed based on granzyme B
CN115477704B (en) 2021-06-16 2024-02-23 四川大学华西医院 Preparation and application of chimeric antigen receptor immune cells constructed based on LOX1
WO2022266660A1 (en) 2021-06-17 2022-12-22 Amberstone Biosciences, Inc. Anti-cd3 constructs and uses thereof
WO2022271955A1 (en) 2021-06-23 2022-12-29 Musc Foundation For Research Development Novel targeted shrna nanoparticles for cancer therapy
WO2022269518A2 (en) 2021-06-23 2022-12-29 Novartis Ag Compositions and methods for the treatment of hemoglobinopathies
WO2023278641A1 (en) 2021-06-29 2023-01-05 Flagship Pioneering Innovations V, Inc. Immune cells engineered to promote thanotransmission and uses thereof
WO2023283611A1 (en) 2021-07-08 2023-01-12 Staidson Biopharma Inc. Antibodies specifically recognizing tnfr2 and uses thereof
WO2023284714A1 (en) 2021-07-14 2023-01-19 舒泰神(北京)生物制药股份有限公司 Antibody that specifically recognizes cd40 and application thereof
WO2023288278A1 (en) 2021-07-16 2023-01-19 Instil Bio (Uk) Limited Chimeric molecules providing targeted costimulation for adoptive cell therapy
WO2023006765A1 (en) 2021-07-26 2023-02-02 Boehringer Ingelheim International Gmbh Treatment and prevention of alcoholic liver disease
WO2023012165A1 (en) 2021-08-02 2023-02-09 Universite De Montpellier Compositions and methods for treating cmt1a or cmt1e diseases with rnai molecules targeting pmp22
WO2023056329A1 (en) 2021-09-30 2023-04-06 Akouos, Inc. Compositions and methods for treating kcnq4-associated hearing loss
US20230241118A1 (en) 2021-10-20 2023-08-03 University Of Rochester Rejuvenation treatment of age-related white matter loss
WO2023081633A1 (en) 2021-11-02 2023-05-11 University Of Rochester Tcf7l2 mediated remyelination in the brain
WO2023086939A1 (en) 2021-11-12 2023-05-19 Amicus Therapeutics, Inc. Compositions and methods for treating mucopolysaccharidosis iiia
WO2023089556A1 (en) 2021-11-22 2023-05-25 Pfizer Inc. Reducing risk of antigen mimicry in immunogenic medicaments
WO2023105005A1 (en) 2021-12-09 2023-06-15 BioNTech SE Chimeric antigen receptor-modified cells for the treatment of cldn6 expressing cancer
WO2024046572A1 (en) 2022-09-01 2024-03-07 BioNTech SE Chimeric antigen receptor-modified cells for the treatment of cldn6 expressing cancer
WO2023111196A1 (en) 2021-12-16 2023-06-22 Singapore Health Services Pte. Ltd. Treatment and prevention of glomerular disease
WO2023144779A1 (en) 2022-01-28 2023-08-03 Pfizer Inc. Coronavirus antigen variants
GB202201137D0 (en) 2022-01-28 2022-03-16 Thelper As Therapeutic and diagnostic agents and uses thereof
WO2023177954A1 (en) 2022-03-18 2023-09-21 University Of Rochester Combination therapy for treatment of cancer, methods and systems of delivery thereof
GB202206507D0 (en) 2022-05-04 2022-06-15 Antion Biosciences Sa Expression construct
WO2023215860A1 (en) 2022-05-05 2023-11-09 University Of Rochester Modified bacteria and methods of use for bioglass microlenses
WO2023230531A1 (en) 2022-05-24 2023-11-30 Lunglife Ai, Inc. Methods for detecting circulating genetically abnormal cells
WO2024020407A1 (en) 2022-07-19 2024-01-25 Staidson Biopharma Inc. Antibodies specifically recognizing b- and t-lymphocyte attenuator (btla) and uses thereof
WO2024028794A1 (en) 2022-08-02 2024-02-08 Temple Therapeutics BV Methods for treating endometrial and ovarian hyperproliferative disorders
GB202212077D0 (en) 2022-08-18 2022-10-05 Tribune Therapeutics Ab Agents that inhibit ccn ligand-induced signalling for treating disease
WO2024059618A2 (en) 2022-09-13 2024-03-21 Arsenal Biosciences, Inc. Immune cells having co-expressed tgfbr shrnas
WO2024056809A1 (en) 2022-09-15 2024-03-21 Novartis Ag Treatment of autoimmune disorders using chimeric antigen receptor therapy
WO2024059824A2 (en) 2022-09-16 2024-03-21 Arsenal Biosciences, Inc. Immune cells with combination gene perturbations

Citations (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4469863A (en) * 1980-11-12 1984-09-04 Ts O Paul O P Nonionic nucleic acid alkyl and aryl phosphonates and processes for manufacture and use thereof
US4511713A (en) * 1980-11-12 1985-04-16 The Johns Hopkins University Process for selectively controlling unwanted expression or function of foreign nucleic acids in animal or mammalian cells
US5034323A (en) * 1989-03-30 1991-07-23 Dna Plant Technology Corporation Genetic engineering of novel plant phenotypes
US5107065A (en) * 1986-03-28 1992-04-21 Calgene, Inc. Anti-sense regulation of gene expression in plant cells
US5190931A (en) * 1983-10-20 1993-03-02 The Research Foundation Of State University Of New York Regulation of gene expression by employing translational inhibition of MRNA utilizing interfering complementary MRNA
US5208149A (en) * 1983-10-20 1993-05-04 The Research Foundation Of State University Of New York Nucleic acid constructs containing stable stem and loop structures
US5258369A (en) * 1988-08-29 1993-11-02 Hem Pharmaceuticals Corporation Treatment of chronic cerebral dysfunction by dsRNA methodology
US5272065A (en) * 1983-10-20 1993-12-21 Research Foundation Of State University Of New York Regulation of gene expression by employing translational inhibition of MRNA utilizing interfering complementary MRNA
US5365015A (en) * 1989-07-14 1994-11-15 Imperial Chemical Industries Plc Antisense constructs derived from pTOM13 plants and plant cells with reduced ethylene evolution
US5453566A (en) * 1986-03-28 1995-09-26 Calgene, Inc. Antisense regulation of gene expression in plant/cells
US5738985A (en) * 1993-04-02 1998-04-14 Ribogene, Inc. Method for selective inactivation of viral replication
US5795715A (en) * 1991-12-18 1998-08-18 Cis Bio International Process for preparing double-stranded RNA, and its applications
US5874555A (en) * 1987-10-30 1999-02-23 California Institute Of Technology Triple helices and processes for making same
US5976567A (en) * 1995-06-07 1999-11-02 Inex Pharmaceuticals Corp. Lipid-nucleic acid particles prepared via a hydrophobic lipid-nucleic acid complex intermediate and use for gene transfer
US6010908A (en) * 1992-08-21 2000-01-04 The Regents Of The University Of California Gene therapy by small fragment homologous replacement
US6136601A (en) * 1991-08-21 2000-10-24 Epoch Pharmaceuticals, Inc. Targeted mutagenesis in living cells using modified oligonucleotides

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5265015A (en) * 1991-06-27 1993-11-23 Schlumberger Technology Corporation Determining horizontal and/or vertical permeability of an earth formation
WO1998004717A2 (en) 1996-07-30 1998-02-05 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Double-stranded rna dependent protein kinase derived peptides to promote proliferation of cells and tissues in a controlled manner
US6124091A (en) 1997-05-30 2000-09-26 Research Corporation Technologies, Inc. Cell growth-controlling oligonucleotides
US6506559B1 (en) 1997-12-23 2003-01-14 Carnegie Institute Of Washington Genetic inhibition by double-stranded RNA
DE69943389D1 (en) 1998-04-08 2011-06-09 Commw Scient Ind Res Org METHOD AND MEANS FOR OBTAINING MODIFIED PHENOTYPES
AR020078A1 (en) 1998-05-26 2002-04-10 Syngenta Participations Ag METHOD FOR CHANGING THE EXPRESSION OF AN OBJECTIVE GENE IN A PLANT CELL
GB9827152D0 (en) 1998-07-03 1999-02-03 Devgen Nv Characterisation of gene function using double stranded rna inhibition
BR0009884A (en) 1999-04-21 2002-01-08 American Home Prod Processes and compositions for inhibiting the function of polynucleotide sequences
US7144714B2 (en) * 2003-09-25 2006-12-05 Board Of Regents, The University Of Texas System R2D2: an enzyme of RNA silencing

Patent Citations (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4511713A (en) * 1980-11-12 1985-04-16 The Johns Hopkins University Process for selectively controlling unwanted expression or function of foreign nucleic acids in animal or mammalian cells
US4469863A (en) * 1980-11-12 1984-09-04 Ts O Paul O P Nonionic nucleic acid alkyl and aryl phosphonates and processes for manufacture and use thereof
US5272065A (en) * 1983-10-20 1993-12-21 Research Foundation Of State University Of New York Regulation of gene expression by employing translational inhibition of MRNA utilizing interfering complementary MRNA
US5190931A (en) * 1983-10-20 1993-03-02 The Research Foundation Of State University Of New York Regulation of gene expression by employing translational inhibition of MRNA utilizing interfering complementary MRNA
US5208149A (en) * 1983-10-20 1993-05-04 The Research Foundation Of State University Of New York Nucleic acid constructs containing stable stem and loop structures
US5107065A (en) * 1986-03-28 1992-04-21 Calgene, Inc. Anti-sense regulation of gene expression in plant cells
US5453566A (en) * 1986-03-28 1995-09-26 Calgene, Inc. Antisense regulation of gene expression in plant/cells
US5874555A (en) * 1987-10-30 1999-02-23 California Institute Of Technology Triple helices and processes for making same
US5258369A (en) * 1988-08-29 1993-11-02 Hem Pharmaceuticals Corporation Treatment of chronic cerebral dysfunction by dsRNA methodology
US5034323A (en) * 1989-03-30 1991-07-23 Dna Plant Technology Corporation Genetic engineering of novel plant phenotypes
US5365015A (en) * 1989-07-14 1994-11-15 Imperial Chemical Industries Plc Antisense constructs derived from pTOM13 plants and plant cells with reduced ethylene evolution
US6136601A (en) * 1991-08-21 2000-10-24 Epoch Pharmaceuticals, Inc. Targeted mutagenesis in living cells using modified oligonucleotides
US5795715A (en) * 1991-12-18 1998-08-18 Cis Bio International Process for preparing double-stranded RNA, and its applications
US6010908A (en) * 1992-08-21 2000-01-04 The Regents Of The University Of California Gene therapy by small fragment homologous replacement
US5738985A (en) * 1993-04-02 1998-04-14 Ribogene, Inc. Method for selective inactivation of viral replication
US5976567A (en) * 1995-06-07 1999-11-02 Inex Pharmaceuticals Corp. Lipid-nucleic acid particles prepared via a hydrophobic lipid-nucleic acid complex intermediate and use for gene transfer

Cited By (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010077894A2 (en) 2008-12-16 2010-07-08 Bristol-Myers Squibb Company Methods of inhibiting quiescent tumor proliferation
US20100233172A1 (en) * 2008-12-16 2010-09-16 Bristol-Myers Squibb Company Methods of inhibiting quiescent tumor proliferation
EP2769737A1 (en) 2009-07-20 2014-08-27 Bristol-Myers Squibb Company Combination of an anti-CTLA4 antibody with etoposide for the synergistic treatment of proliferative diseases
EP2947098A1 (en) 2009-07-20 2015-11-25 Bristol-Myers Squibb Company Combination of anti-ctla4 antibody with gemcitabine for the synergistic treatment of proliferative diseases
EP3659596A1 (en) 2009-07-20 2020-06-03 Bristol-Myers Squibb Company Combination of anti-ctla4 antibody with paclitaxel for the synergistic treatment of proliferative diseases
EP4029508A1 (en) 2014-10-10 2022-07-20 Idera Pharmaceuticals, Inc. Treatment of cancer using tlr9 agonists and checkpoint inhibitors
WO2017160754A1 (en) 2016-03-15 2017-09-21 Mersana Therapeutics,Inc. Napi2b-targeted antibody-drug conjugates and methods of use thereof
EP4302782A2 (en) 2016-03-15 2024-01-10 Mersana Therapeutics, Inc. Napi2b-targeted antibody-drug conjugates and methods of use thereof
US10604574B2 (en) 2016-06-30 2020-03-31 Oncorus, Inc. Oncolytic viral delivery of therapeutic polypeptides
US11078280B2 (en) 2016-06-30 2021-08-03 Oncorus, Inc. Oncolytic viral delivery of therapeutic polypeptides
WO2018098269A2 (en) 2016-11-23 2018-05-31 Mersana Therapeutics, Inc. Peptide-containing linkers for antibody-drug conjugates
WO2018160538A1 (en) 2017-02-28 2018-09-07 Mersana Therapeutics, Inc. Combination therapies of her2-targeted antibody-drug conjugates
WO2019104289A1 (en) 2017-11-27 2019-05-31 Mersana Therapeutics, Inc. Pyrrolobenzodiazepine antibody conjugates
US11638760B2 (en) 2017-11-27 2023-05-02 Mersana Therapeutics, Inc. Pyrrolobenzodiazepine antibody conjugates
WO2019126691A1 (en) 2017-12-21 2019-06-27 Mersana Therapeutics, Inc. Pyrrolobenzodiazepine antibody conjugates
US11865081B2 (en) 2017-12-29 2024-01-09 Virogin Biotech Canada Ltd. Oncolytic viral delivery of therapeutic polypeptides
WO2020092385A1 (en) 2018-10-29 2020-05-07 Mersana Therapeutics, Inc. Cysteine engineered antibody-drug conjugates with peptide-containing linkers
WO2020226633A1 (en) 2019-05-07 2020-11-12 Immunicom, Inc. Increasing responses to checkpoint inhibitors by extracorporeal apheresis
WO2021216920A1 (en) 2020-04-22 2021-10-28 Iovance Biotherapeutics, Inc. Systems and methods for coordinating manufacturing of cells for patient-specific immunotherapy

Also Published As

Publication number Publication date
JP2003516124A (en) 2003-05-13
US7282564B2 (en) 2007-10-16
EP1235842A4 (en) 2003-04-23
EP1235842A1 (en) 2002-09-04
US20080305543A1 (en) 2008-12-11
HK1047109A1 (en) 2003-02-07
US20040265839A1 (en) 2004-12-30
AU1086501A (en) 2001-04-30
US20060024798A1 (en) 2006-02-02
WO2001029058A1 (en) 2001-04-26
CA2386270A1 (en) 2001-04-26
US7759463B2 (en) 2010-07-20

Similar Documents

Publication Publication Date Title
US7282564B2 (en) RNA interference pathway genes as tools for targeted genetic interference
Lundström et al. Vilse, a conserved Rac/Cdc42 GAP mediating Robo repulsion in tracheal cells and axons
Lu et al. lin-35 and lin-53, two genes that antagonize a C. elegans Ras pathway, encode proteins similar to Rb and its binding protein RbAp48
US20090077679A1 (en) Gene silencing by systemic rna interference
CA2373628A1 (en) Animal models and methods for analysis of lipid metabolism and screening of pharmaceutical and pesticidal agents that modulate lipid metabolism
AU2006201716B2 (en) RNA interference pathway genes as tools for targeted genetic interference
WO2001018549A1 (en) Method for identifying modulators of daf-16
WO2003044168A9 (en) Facilitation of rna interference
WO1999054436A2 (en) Nucleic acids and proteins of c. elegans insulin-like genes and uses thereof
US10273487B2 (en) Transfection vector for pathogenic amoebae and uses thereof
US7125976B2 (en) Method of screening for agents inhibiting chloride intracellular channels
US6489459B1 (en) Identification oh HOP-1 and uses thereof
US20070191295A1 (en) Gene silencing by systemic rna interference
WO2002038600A9 (en) C. elegans genes involved in viability and/or reproduction and uses thereof
US6781028B1 (en) Animal models and methods for analysis of lipid metabolism and screening of pharmaceutical and pesticidal agents that modulate lipid metabolism
US7193126B2 (en) Method for generating overexpression of alleles in genes of unknown function
US6596495B1 (en) EGL-1, a new protein required for programmed cell death in C. elegans that interacts with the BCL-2-like protein CED-9
Tharin Molecular and genetic analyses of the axon guidance gene unc-69
Chen Extragenic suppressors of heat shock activated GOalpha: Topic I. Cyclin in heat shock response. Topic II. Signaling by Go and Gq in Caenorhabditis elegans
JP4877835B2 (en) RNA interference induction element and use thereof
Del Campo Functional analysis of EFF-1: A novel protein necessary and sufficient for somatic cell fusion in the nematode Caenorhabditis elegans
Zheng Identification and study of gamma tubulins in Drosophila melanogaster and Homo sapiens
Tong Cloning and characterization of exc-9, a Caenorhabditis elegans CRIP homologue that regulates tubular structure
Zhen Cloning, characterization and functional analysis of UBC-2, a gene encoding a ubiguitin-conjugating enzyme in the nematode Caenorhabditis elegans
Schinkel et al. Structure and function of P-glycoproteins

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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