AU2001249622A1 - RNA sequence-specific mediators of RNA interference - Google Patents

RNA sequence-specific mediators of RNA interference

Info

Publication number
AU2001249622A1
AU2001249622A1 AU2001249622A AU2001249622A AU2001249622A1 AU 2001249622 A1 AU2001249622 A1 AU 2001249622A1 AU 2001249622 A AU2001249622 A AU 2001249622A AU 2001249622 A AU2001249622 A AU 2001249622A AU 2001249622 A1 AU2001249622 A1 AU 2001249622A1
Authority
AU
Australia
Prior art keywords
rna
mrna
gene
cell
organism
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.)
Granted
Application number
AU2001249622A
Other versions
AU2001249622B2 (en
Inventor
David P. Bartel
Phillip A. Sharp
Thomas Tuschl
Phillip D. Zamore
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.)
Max Planck Gesellschaft zur Foerderung der Wissenschaften eV
Whitehead Institute for Biomedical Research
Massachusetts Institute of Technology
University of Massachusetts Medical Center
Original Assignee
Max Planck Gesellschaft zur Foerderung der Wissenschaften eV
Whitehead Institute for Biomedical Research
Massachusetts Institute of Technology
University of Massachusetts Medical Center
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 Max Planck Gesellschaft zur Foerderung der Wissenschaften eV, Whitehead Institute for Biomedical Research, Massachusetts Institute of Technology, University of Massachusetts Medical Center filed Critical Max Planck Gesellschaft zur Foerderung der Wissenschaften eV
Priority claimed from PCT/US2001/010188 external-priority patent/WO2001075164A2/en
Publication of AU2001249622A1 publication Critical patent/AU2001249622A1/en
Assigned to WHITEHEAD INSTITUTE FOR BIOMEDICAL RESEARCH, UNIVERSITY OF MASSACHUSETTS MEDICAL CENTER, MAX-PLANCK-GESELLSCHAFT ZUR FORDERUNG DER WISSENSCHAFTEN E.V., MASSACHUSETTS INSTITUTE OF TECHNOLOGY reassignment WHITEHEAD INSTITUTE FOR BIOMEDICAL RESEARCH Amend patent request/document other than specification (104) Assignors: MASSACHUSETTS INSTITUTE OF TECHNOLOGY, MAX-PLANCK-GESELLSCHAFT ZUR FORDERUNG DER WISSENSCHAFTEN E.V., UNIVERSITY OF MASSACHUSSETS MEDICAL CENTER, WHITEHEAD INSTITUTE FOR BIOMEDICAL RESEARCH
Application granted granted Critical
Publication of AU2001249622B2 publication Critical patent/AU2001249622B2/en
Priority to AU2007214287A priority Critical patent/AU2007214287B2/en
Priority to AU2010241526A priority patent/AU2010241526B2/en
Priority to AU2013204199A priority patent/AU2013204199C1/en
Anticipated expiration legal-status Critical
Expired legal-status Critical Current

Links

Description

. RNA Sequence-Specific Mediators of RNA Interference
RELATED APPLICATIONS
This application claims the benefit of U.S. Provisional Application No. 60/265,232, filed January 31, 2001 and U.S. Provisional Application No. 60/193,594, filed March 30, 2000, and claims priority under 35 U.S.C. §119 to European Application No. 00 126 325.0 filed December 1, 2000. The entire teachings of the above applications are incorporated herein by reference.
GOVERNMENT SUPPORT
Work described herein was funded in part by grants from the National Institutes of Health through a United States Public Health Service MERIT award (Grant No. RO1-GM34277) from the National Institutes of Health. The United States government has certain rights in the invention.
BACKGROUND OF THE INVENTION
RNA interference or "RNAi" is a term initially coined by Fire and co-workers to describe the observation that double-stranded RNA (dsRNA) can block gene expression when it is introduced into worms (Fire et al. (1998) Nature 391, 806-811). dsRNA directs gene-specific, post-transcriptional silencing in many organisms, including vertebrates, and has provided a new tool for studying gene function. RNAi involves mRNA degradation, but many of the biochemical mechanisms underlying this interference are unknown. The recapitulation of the essential features of RNAi in vitro is needed for a biochemical analysis of the phenomenon.
SUMMARY OF THE INVENTION
Described herein is gene-specific, dsRNA-mediated interference in a cell-free system derived from syncytial blastoderm Drosophila embryos. The in vitro system complements genetic approaches to dissecting the molecular basis of RNAi. As described herein, the molecular mechanisms underlying RNAi were examined using the Drosophila in vitro system. Results showed that RNAi is ATP-dependent yet uncoupled from mRNA translation. That is, protein synthesis is not required for RNAi in vitro, hi the RNAi reaction, both strands (sense and antisense) of the dsRNA are processed to small RNA fragments or segments of from about 21 to about 23 nucleotides (nt) in length (RNAs with mobility in sequencing gels that correspond to markers that are 21-23 nt in length, optionally referred to as 21-23 nt RNA). Processing of the dsRNA to the small RNA fragments does not require the targeted mRNA, which demonstrates that the small RNA species is generated by processing of the dsRNA and not as a product of dsRNA-targeted mRNA degradation. The mRNA is cleaved only within the region of identity with the dsRNA. Cleavage occurs at sites 21-23 nucleotides apart, the same interval observed for the dsRNA itself, suggesting that the 21-23 nucleotide fragments from the dsRNA are guiding mRNA cleavage. That purified 21-23 nt RNAs mediate RNAi confirms that these fragments are guiding mRNA cleavage.
Accordingly, the present invention relates to isolated RNA molecules (double- stranded; single-stranded) of from about 21 to about 23 nucleotides which mediate RNAi. That is, the isolated RNAs of the present invention mediate degradation of mRNA of a gene to which the mRNA corresponds (mediate degradation of mRNA that is the transcriptional product of the gene, which is also referred to as a target gene). For convenience, such mRNA is also referred to herein as mRNA to be degraded. As used herein, the terms RNA, RNA molecule(s), RNA segment(s) and RNA fragment(s) are used interchangeably to refer to RNA that mediates RNA interference. These terms include double-stranded RNA, single-stranded RNA, isolated RNA (partially purified RNA, essentially pure RNA, synthetic RNA, recombinantly produced RNA), as well as altered RNA that differs from naturally occurring RNA by the addition, deletion, substitution and/or alteration of one or more nucleotides. Such alterations can include addition of non-nucleotide material, such as to the end(s) of the 21-23 nt RNA or internally (at one or more nucleotides of the RNA). Nucleotides in the RNA molecules of the present invention can also comprise non-standard nucleotides, including non-naturally occurring nucleotides or deoxyribonucleo tides. Collectively, all such altered RNAs are referred to as analogs or analogs of naturally-occurring RNA. RNA of 21-23 nucleotides of the present invention need only be sufficiently similar to natural RNA that it has the ability to mediate (mediates) RNAi. As used herein the phrase "mediates RNAi" refers to (indicates) the ability to distinguish which RNAs are to be degraded by the RNAi machinery or process. RNA that mediates RNAi interacts with the RNAi machinery such that it directs the machinery to degrade particular mRNAs. In one embodiment, the present invention relates to RNA molecules of about 21 to about 23 nucleotides that direct cleavage of specific mRNA to which their sequence corresponds. It is not necessary that there be perfect correspondence of the sequences, but the correspondence must be sufficient to enable the RNA to direct RNAi cleavage of the target mRNA. In a particular embodiment, the 21-23 nt RNA molecules of the present invention comprise a 3' hydroxyl group. The present invention also relates to methods of producing RNA molecules of about 21 to about 23 nucleotides with the ability to mediate RNAi cleavage. In one embodiment, the Drosophila in vitro system is used. In this embodiment, dsRNA is combined with a soluble extract derived. from Drosophila embryo, thereby producing a combination. The combination is maintained under conditions in which the dsRNA is processed to RNA molecules of about 21 to about 23 nucleotides. hi another embodiment, the Drosophila in vitro system is used to obtain RNA sequences of about 21 to about 23 nucleotides which mediate RNA interference of the mRNA of a particular gene (e.g., oncogene, viral gene), hi this embodiment, double-stranded RNA that corresponds to a sequence of the gene to be targeted is combined with a soluble extract derived from Drosophila embryo, thereby producing a combination. The combination is maintained under conditions in which the double-stranded RNA is processed to RNA of about 21 to about 23 nucleotides in length. As shown herein, 21- 23 nt RNA mediates RNAi of the mRNA of the targeted gene (the gene whose mRNA is to be degraded). The method of obtaining 21-23 nt RNAs using the Drosophila in vitro system can further comprise isolating the RNA sequence from the combination. The present invention also relates to 21-23 nt RNA produced by the methods of the present invention, as well as to 21-23 nt RNAs, produced by other methods, such as chemical synthesis or recombinant DNA techniques, that have the same or substantially the same sequences as naturally-occurring RNAs that mediate RNAi, such as those produced by the methods of the present invention. All of these are referred to as 21-23 nt RNAs that mediate RNA interference. As used herein, the term isolated RNA includes RNA obtained by any means, including processing or cleavage of dsRNA as described herein; production by chemical synthetic methods; and production by recombinant DNA techniques. The invention further relates to uses of the 21-23 nt RNAs, such as for therapeutic or prophylactic treatment and compositions comprising 21-23 nt RNAs that mediate RNAi, such as pharmaceutical compositions comprising 21-23 nt RNAs and an appropriate carrier (e.g., a buffer or water).
The present invention also relates to a method of mediating RNA interference of mRNA of a gene in a cell or organism (e.g., mammal such as a mouse or a human). In one embodiment, RNA of about 21 to about 23 nt which targets the mRNA to be degraded is introduced into the cell or organism. The cell or organism is maintained under conditions under which degradation of the mRNA occurs, thereby mediating RNA interference of the mRNA of the gene in the cell or organism. The cell or organism can be one in which RNAi occurs as the cell or organism is obtained or a cell or organism can be one that has been modified so that RNAi occurs (e.g., by addition of components obtained from a cell or cell extract that mediate RNAi or activation of endogenous components). As used herein, the tenn "cell or organism in which RNAi occurs" includes both a cell or organism in which RNAi occurs as the cell or organism is obtained, or a cell or organism that has been modified so that RNAi occurs. In another embodiment, the method of mediating RNA interference of a gene in a cell comprises combining double-stranded RNA that corresponds to a sequence of the gene with a soluble extract derived from Drosophila embryo, thereby producing a combination. The combination is maintained under conditions in which the double-stranded RNA is processed to RNAs of about 21 to about 23 nucleotides. 21 to 23 nt RNA is then isolated and introduced into the cell or organism. The cell or organism is maintained under conditions in which degradation of mRNA of the gene occurs, thereby mediating RNA interference of the gene in the cell or organism. As described for the previous embodiment, the cell or organism is one in which RNAi occurs naturally (in the cell or organism as obtained) or has been modified in such a manner that RNAi occurs. 21 to 23 nt RNAs can also be produced by other methods, such as chemical synthetic methods or recombinant DNA techniques.
The present invention also relates to biochemical components of a cell, such as a Drosophila cell, that process dsRNA to RNA of about 21 to about 23 nucleotides. In addition, biochemical components of a cell that are involved in targeting of mRNA by RNA of about 21 to about 23 nucleotides are the subject of the present invention, hi both embodiments, the biochemical components can be obtained from a cell in which they occur or can be produced by other methods, such as chemical synthesis or recombinant DNA methods. As used herein, the term "isolated" includes materials (e.g., biochemical components, RNA) obtained from a source in which they occur and materials produced by methods such as chemical synthesis or recombinant nucleic acid (DNA, RNA) methods.
The present invention also relates to a method for knocking down, (partially or completely) the targeted gene, thus providing an alternative to presently available methods of knocking down (or out) a gene or genes. This method of knocking down gene expression can be used therapeutically or for research (e.g., to generate models of disease states, to examine the function of a gene, to assess whether an agent acts on a gene, to validate targets for drug discovery), fh those instances in which gene function is eliminated, the resulting cell or organism can also be referred to as a knockout. One embodiment of the method of producing knockdown cells and organisms comprises introducing into a cell or organism in which a gene (referred to as a targeted gene) is to be knocked down, RNA of about 21 to about 23 nt that targets the gene and maintaining the resulting cell or organism under conditions under which RNAi occurs, resulting in degradation of the mRNA of the targeted gene, thereby producing knockdown cells or organisms. Knockdown cells and organisms produced by the present method are also the subject of this invention. The present invention also relates to a method of examining or assessing the function of a gene in a cell or organism. In one embodiment, RNA of about 21 to about 23 nt which targets mRNA of the gene for degradation is introduced into a cell or organism in which RNAi occurs. The cell or organism is referred to as a test cell or organism. The test cell or organism is maintained under conditions under which degradation of mRNA of the gene occurs. The phenotype of the test cell or organism is then observed and compared to that of an appropriate control cell or organism, such as a corresponding cell or organism that is treated in the same manner except that the targeted (specific) gene is not targeted. A 21 to 23 nt RNA that does not target the mRNA for degradation can be introduced into the control cell or organism in place of the RNA introduced into the test cell or organism, although it is not necessary to do so. A difference between the phenotypes of the test and control cells or organisms provides information about the function of the degraded mRNA. In another embodiment, double-stranded RNA that corresponds to a sequence of the gene is combined with a soluble extract that mediates RNAi, such as the soluble extract derived from Drosophila embryo described herein, under conditions in which the double-stranded RNA is processed to generate RNA of about 21 to about 23 nucleotides. The RNA of about 21 to about 23 nucleotides is isolated and then introduced into a cell or organism in which RNAi occurs (test cell or test organism). The test cell or test organism is maintained under conditions under which degradation of the mRNA occurs. The phenotype of the test cell or organism is then observed and compared to that of an appropriate control, such as a corresponding cell or organism that is treated in the same manner as the test cell or organism except that the targeted gene is not targeted. A difference between the phenotypes of the test and control cells or organisms provides information about the function of the targeted gene. The information provided may be sufficient to identify (define) the function of the gene or may be used in conjunction with information obtained from other assays or analyses to do so.
Also the subject of the present invention is a method of validating whether an agent acts on a gene, hi this method, RNA of from about 21 to about 23 nucleotides that targets the mRNA to be degraded is introduced into a cell or organism in which RNAi occurs. The cell or organism (which contains the introduced RNA) is maintained under conditions under which degradation of mRNA occurs, and the agent is introduced into the cell or organism. Whether the agent has an effect on the cell or organism is determined; if the agent has no effect on the cell or organism, then the agent acts on the gene.
The present invention also relates to a method of validating whether a gene product is a target for drug discovery or development. RNA of from about 21 to about 23 nucleotides that targets the mRNA that corresponds to the gene for degradation is introduced into a cell or orgamsm. The cell or organism is maintained under conditions in which degradation of the mRNA occurs, resulting in decreased expression of the gene. Whether decreased expression of the gene has an effect on the cell or organism is determined, wherein if decreased expression of the gene has an effect, then the gene product is a target for drug discovery or development. The present invention also encompasses a method of treating a disease or condition associated with the presence of a protein in an individual comprising administering to the individual RNA of from about 21 to about 23 nucleotides which targets the mRNA of the protein (the mRNA that encodes the protein) for degradation. As a result, the protein is not produced or is not produced to the extent it would be in the absence of the treatment.
Also encompassed by the present invention is a gene identified by the sequencing of endogenous 21 to 23 nucleotide RNA molecules that mediate RNA interference.
Also encompassed by the present invention is a method of identifying target sites within an mRNA that are particularly suitable for RNAi as well as a method of assessing the ability of 21-23 nt RNAs to mediate RNAi.
BRIEF DESCRIPTION OF THE DRAWINGS
The file of this patent contains at least one drawing,executed in color. Copies of this patent with color drawing(s) will be provided by the Patent and Trademark Office upon request and payment of the necessary fee. Figure 1 is a schematic representation of reporter mRNAs and dsRNAs Rr-Luc and Pp-Luc. Lengths and positions of the ssRNA, asRNA, and dsRNAs are shown as black bars relative to the Rr-Luc and Pp-Luc reporter mRNA sequences. Black rectangles indicate the two unrelated luciferase coding sequences, lines correspond to the 5' and 3' untranslated regions of the mRNAs.
Figure 2 A is a graph of the ratio of luciferase activities after targeting 50 pM Pp- Luc mRNA with 10 nM ssRNA, asRNA, or dsRNA from the 505 bp segment of the Pp- Luc gene showing gene-specific interference by dsRNA in vitro. The data are the average values of seven trials ± standard deviation. Four independently prepared lysates were used. Luciferase activity was normalized to the buffer control; a ratio equal to one indicates no gene-specific interference.
Figure 2B is a graph of the ratio of luciferase activities after targeting 50 pM Rr- Luc mRNA with 10 nM ssRNA, asRNA, or dsRNA from the 501 bp segment of the Rr- Luc gene showing gene-specific interference by dsRNA in vitro. The data are the average values of six trials ± standard deviation. A Rr-Luc/Pp-Luc ratio equal to one indicates no gene-specific interference.
Figure 3 A is a schematic representation of the experimental strategy used to show that incubation in the Drosophila embryo lysate potentiates dsRNA for gene- specific interference. The same dsRNAs used in Figure 2 (or buffer) was serially preincubated using two-fold dilutions in six successive reactions with Drosophila embryo lysate, then tested for its capacity to block mRNA expression. As a control, the same amount of dsRNA (10 nM) or buffer was diluted directly in buffer and incubated with Pp-Luc and Rr-Luc mRNAs and lysate.
Figure 3B is a graph of potentiation when targeting Pp-Luc mRNA. Black columns indicate the dsRNA or the buffer was serially preincubated; white columns correspond to a direct 32-fold dilution of the dsRNA. Values were normalized to those of the buffer controls.
Figure 3C is a graph of potentiation when targeting Rr-Luc mRNA. The corresponding buffer control is shown in Figure 3B. Figure 4 is a graph showing effect of competitor dsRNA on gene- specific interference. Increasing concentrations of nanos dsRNA ( 508 bp) were added to reactions containing 5 nM dsRNA (the same dsRNAs used in Figures 2A and 2B) targeting Pp-Luc mRNA (black columns, left axis) or Rr-Luc mRNA (white columns, right axis). Each reaction contained both a target mRNA (Pp-Luc for the black columns, Rr-Luc for the white) and an unrelated control mRNA (Rr-Luc for the black columns, Pp-Luc for the white). Values were normalized to the buffer control (not shown). The reactions were incubated under standard conditions (see Methods).
Figure 5 A is a graph showing the effect of dsRNA on mRNA stability. Circles, Pp-Luc mRNA; squares, Rr-Luc mRNA; filled symbols, buffer incubation; open symbols, incubation with Pp-dsRNA.
Figure 5B is a graph showing the stability of Rr-Luc mRNA incubated with Rr- dsRNA or Pp-dsRNA. Filled squares, buffer; open squares, Pp-dsRNA (10 nM); open circles, Rr-dsRNA (10 nM).
Figure 5C is a graph showing the dependence on dsRNA length. The stability of the Pp-Luc mRNA was assessed after incubation in lysate in the presence of buffer or dsRNAs of different lengths. Filled squares, buffer; open circles, 49 bp dsRNA (10 nM); open inverted triangles, 149 bp dsRNA (10 nM); open triangles, 505 bp dsRNA (10 nM); open diamonds, 997 bp dsRNA (10 nM). Reactions were incubated under standard conditions (see Methods). Figure 6 is a graph showing that RNAi Requires ATP. Creatine kinase (CK) uses creatine phosphate (CP) to regenerate ATP. Circles, +ATP, +CP, +CK; squares, -ATP, +CP, +CK; triangles, -ATP, -CP, +CK; inverted angles, -ATP, +CP, -CK.
Figure 7 A is a graph of protein synthesis, as reflected by luciferase activity produced after incubation of Rr-luc mRNA in the in vitro RNAi reaction for 1 hour, in the presence of the protein synthesis inhibitors anisomycin, cycloheximide, or chloramphenicol, relative to a reaction without any inhibitor showing that RNAi does not require mRNA translation.
Figure 7B is a graph showing translation of 7-methyl-guanosine- and adenosine- capped Pp-luc mRNAs (circles and squares, respectively) in the RNAi reaction in the absence of dsRNA, as measured by luciferase activity produced in a one-hour incubation.
Figure 7C is a graph showing incubation in an RNAi reaction of uniformly 32P- radiolabeled 7-methyl-guanosine-capped Pp-luc mRNA (circles) and adenosine-capped Pp-luc mRNA (squares), in the presence (open symbols) and absence (filled symbols) of 505 bp Pp-luc dsRNA.
Figure 8 A is a graph of the of the denaturing agarose-gel analysis of Pp-luc mRNA incubated in a standard RNAi reaction with buffer, 505 nt Pp-asRNA, or 505 bp Pp-dsRNA for the times indicated showing that asRNA causes a small amount of RNAi in vitro.
Figure 8B is a graph of the of the denaturing agarose-gel analysis of Rr-luc mRNA incubated in a standard RNAi reaction with buffer, 505 nt Pp-asRNA, or 505 bp Pp-dsRNA for the times indicated showing that asRNA causes a small amount of RNAi in vitro. Figure 9 is a schematic of the positions of the three dsRNAs, 'A,' 'B,' and 'C,' relative to the Rr-luc mRNA.
FigurelO indicates the cleavage sites mapped onto the first 267 nt of the Rr-luc mRNA (SEQ ID NO: 1). The blue bar below the sequence indicates the position of dsRNA 'C,' and blue circles indicate the position of cleavage sites caused by this dsRNA. The green bar denotes the position of dsRNA 'B,' and green circles, the cleavage sites. The magenta bar indicates the position of dsRNA 'A,' and magenta circles, the cleavages. An exceptional cleavage within a run of 7 uracils is marked with a red arrowhead.
Figure 11 is a proposed model for RNAi. RNAi is envisioned to begin with cleavage of the dsRNA to 21-23 nt products by a dsRNA-specific nuclease, perhaps in a multiprotein complex. These short dsRNAs might then be dissociated by an ATP- dependent helicase, possibly a component of the initial complex, to 21-23 nt asRNAs that could then target the mRNA for cleavage. The short asRNAs are imagined to remain associated with the RNAi-specific proteins (circles) that were originally bound by the full-length dsRNA, thus explaining the inefficiency of asRNA to trigger RNAi in vivo and in vitro. Finally, a nuclease (triangles) would cleave the mRNA.
Figure 12 is a bar graph showing sequence-specific gene silencing by 21-23 nt fragments. Ratio of luciferase activity after targeting of Pp-Luc and Rr-Luc mRNA by 5 nM Pp-Luc or Rr-Luc dsRNA (500 bp) or 21-23 nt fragments isolated from a previous incubation of the respective dsRNA in Drosophila lysate. The amount of isolated 21-23 mers present in the incubation reaction correspond to approximately the same amount of 21-23 mers generated during an incubation reaction with 5 nM 500 bp dsRNA. The data are average values of 3 trials and the standard deviation is given by error bars. Luciferase activity was normalized to the buffer control.
Figure 13A illustrates the purification of RNA fragments on a Superdex HR 200 10/30 gel filtration column (Pharmacia) using the method described in Example 4. dsRNA was 32P-labeled, and the radioactivity recovered in each column fraction is graphed. The fractions were also analyzed by denaturing gel electrophoresis (inset).
Figure 13B demonstrates the ability of the Rr-luciferase RNA, after incubation in the Drosophila lysate and fractionation as in Fig. 13 A, to mediate sequence-specific interference with the expression of a Rr-luciferase target mRNA. One microliter of each resuspended fraction was tested in a 10 microliter in vitro RNAi reaction (see Example 1). This procedure yields a concentration of RNA in the standard in vitro RNAi reaction that is approximately equal to the concentration of that RNA species in the original reaction prior to loading on the column. Relative luminescence per second has been normalized to the average value of the two buffer controls.
Figure 13C is the specificity control for Fig 13B. It demonstrates that the fractionated RNA of Fig 13B does not efficiently mediate sequence-specific interference with the expression of a Pp-luciferase mRNA. Assays are as in Fig 13B. Figures 14A and 14B are schematic representations of reporter constructs and siRNA duplexes. Figure 14A illustrates the firefly (Pp-luc) and sea pansy (Rr- luc) luciferase reporter gene regions from plasmids pGL2-Control, pGL3-Control, and pRL-TK (Promega). SV40 regulatory elements, the HSV thymidine. kinase promoter, and two introns (lines) are indicated. The sequence of GL3 luciferase is 95% identical to GL2, but RL is completely unrelated to both. Luciferase expression from pGL2 is approximately 10-fold lower than from pGL3 in transfected mammalian cells. The region targeted by the siRNA duplexes is indicated as black bar below the coding region of the "luciferase genes. Figure 14B shows the sense (top) and antisense (bottom) sequences of the siRNA duplexes targeting GL2 (SEQ ID Nos: 10 and 11), GL3 (SEQ ID Nos: 12 and 13), and RL (SEQ ID Nos: 14 and 15) luciferase are shown. The GL2 and GL3 siRNA duplexes differ by only 3 single nucleotide substitutions (boxed in gray). As unspecific control, a duplex with the inverted GL2 sequence, invGL2 (SEQ ID Nos: 16 and 17), was synthesized. The 2 nt 3' overhang of 2'-deoxythymidine is indicated as TT; uGL2 (SEQ ID Nos: 18 and 19) is similar to GL2 siRNA but contains ribo-uridine 3' overhangs. Figures 15A-15J are graphs showing RNA interference by siRNA duplexes.
Ratios of target to control luciferase were normalized to a buffer control (bu, black bars); gray bars indicate ratios of Photinus pyralis (Rp-luc) GL2 or GL3 luciferase to Renilla reniformis (Rr-luc) RL luciferase (left axis), white bars indicate RL to GL2 or GL3 ratios (right axis). Figures 15A, 15C, 15E, 15G, and 151 show results of experiments perfomied with the combination of pGL2-Control and pRL-TK reporter plasmids, Figures 15B, 15D, 15F, 15H, and 15 J with pGL3-Control and pRL-TK reporter plasmids. The cell line used for the interference experiment is indicated at the top of each plot. The ratios of Pp-luc/Rr-luc for the buffer control (bu) varied between 0.5 and 10 for pGL2/pRL, and between 0.03 and 1 for pGL3/pRL, respectively, before nonnalization and between the various cell lines tested. The plotted data were averaged from three independent experiments ± S.D.
Figures 16A-16F are graphs showing the effects of 21 nt siRNAs, 50 bp, and 500 bp dsRNAs on luciferase expression in HeLa cells. The exact length of the long dsRNAs is indicated below the bars. Figures 16A, 16C, and 16E describe experiments performed with pGL2-Control and pRL-TK reporter plasmids, Figures 16B, 16D, and 16F with pGL3-Control and pRL-TK reporter plasmids. The data were averaged from two independent experiments ± S.D. Figures 16A, 16B, Absolute Pp-luc expression, plotted in arbitrary luminescence units. Figure 16C, 16D, Rr-luc expression, plotted in arbitrary luminescence units. Figures 16E, 16F, Ratios of normalized target to control luciferase. The ratios of luciferase activity for siRNA duplexes were normalized to a buffer control (bu, black bars); the luminescence ratios for 50 or 500 bp dsRNAs were normalized to the respective ratios observed for 50 and 500 bp dsRNA from humanized GFP (hG, black bars). It should be noted, that the overall differences in sequence between the 49 and 484 bp dsRNAs targeting GL2 and GL3 are not sufficient to confer specificity between GL2 and GL3 targets (43 nt uninterrupted identity in 49 bp segment, 239 nt longest uninterrupted identity in 484 bp segment) (Parrish, S., et al, Mol. Cell, 6Λ 077-1087 (2000)).
DETAILED DESCRIPTION OF THE INVENTION
Double-stranded (dsRNA) directs the sequence-specific degradation of mRNA through a process known as RNA interference (RNAi). The process is known to occur in a wide variety of organisms, including embryos of mammals and other vertebrates. Using the Drosophila in vitro system described herein, it has been demonstrated that dsRNA is processed to RNA segments 21-23 nucleotides (nt) in length, and furthermore, that when these 21-23 nt fragments are purified and added back to Drosophila extracts, they mediate RNA interference in the absence of longer dsRNA. Thus, these 21-23 nt fragments are sequence-specific mediators of RNA degradation. A molecular signal, which may be the specific length of the fragments, must be present in these 21-23 nt fragments to recruit cellular factors involved in RNAi. This present invention encompasses these 21-23 nt fragments and their use for specifically inactivating gene function. The use of these fragments (or recombinantly produced or chemically synthesized oligonucleotides of the same or similar nature) enables the targeting of specific mRNAs for degradation in mammalian cells. Use of long dsRNAs in mammalian cells to elicit RNAi is usually not practical, presumably because of the deleterious effects of the interferon response. Specific targeting of a particular gene function, which is possible with 21-23 nt fragments of the present invention, is useful in functional genomic and therapeutic applications.
In particular, the present invention relates to RNA molecules of about 21 to about 23 nucleotides that mediate RNAi. In one embodiment, the present invention relates to RNA molecules of about 21 to about 23 nucleotides that direct cleavage of specific mRNA to which they correspond. The 21-2*3 nt RNA molecules of the present invention can also comprise a 3' hydroxyl group. The 21-23 nt RNA molecules can be single-stranded or double stranded (as two 21-23 nt RNAs); such molecules can be blunt ended or comprise overhanging ends (e.g., 5 3'). In specific embodiments, the RNA molecule is double stranded and either blunt ended or comprises overhanging ends (as two 21-23 nt RNAs).
In one embodiment, at least one strand of the RNA molecule has a 3' overhang from about 1 to about 6 nucleotides (e.g., pyrimidine nucleotides, purine nucleotides) in length, h other embodiments, the 3' overhang is from about 1 to about 5 nucleotides, from about 1 to about 3 nucleotides and from about 2 to about 4 nucleotides in length, hi one embodiment the RNA molecule is double stranded, one strand has a 3' overhang and the other strand can be blunt-ended or have an overhang, hi the embodiment in which the RNA molecule is double stranded and both strands comprise an overhang, the length of the overhangs maybe the same or different for each strand, hi a particular embodiment, the RNA of the present invention comprises 21 nucleotide strands which are paired and which have overhangs of from about 1 to about 3, particularly about 2, nucleotides on both 3' ends of the RNA. In order to further enhance the stability of the RNA of the present invention, the 3' overhangs can be stabilized against degradation, hi one embodiment, the RNA is stabilized by including purine nucleotides, such as adenosine or guanosine nucleotides. Alternatively, substitution of pyrimidine nucleotides by modified analogues, e.g., substitution of uridine 2 nucleotide 3' overhangs by 2'-deoxythymidine is tolerated and does not affect the efficiency of RNAi. The absence of a 2' hydroxyl significantly enhances the nuclease resistance of the overhang in tissue culture medium. The 21-23 nt RNA molecules of the present invention can be obtained using a number of techniques known to those of skill in the art. For example, the RNA can be chemically synthesized or recombinantly produced using methods known in the art. The 21-23 nt RNAs can also be obtained using the Drosophila in vitro system described herein. Use of the Drosophila in vitro system entails combining dsRNA with a soluble extract derived from Drosophila embryo, thereby producing a combination. The combination is maintained under conditions in which the dsRNA is processed to RNA of about 21 to about 23 nucleotides. The Drosophila in vitro system can also be used to obtain RNA of about 21 to about 23 nucleotides in length which mediates RNA interference of the mRNA of a particular gene (e.g., oncogene, viral gene), hi this embodiment, double-stranded RNA that corresponds to a sequence of the gene is combined with a soluble extract derived from Drosophila embryo, thereby producing a combination. The combination is maintained under conditions in which the double- stranded RNA is processed to the RNA of about 21 to about 23 nucleotides. As shown herein, 21-23 nt RNA mediates RNAi of the mRNA to be degraded. The present invention also relates to the 21-23 nt RNA molecules produced by the methods described herein. hi one embodiment, the methods described herein are used to identify or obtain 21-23 nt RNA molecules that are useful as sequence-specific mediators of RNA degradation and, thus, for inhibiting mRNAs, such as human mRNAs, that encode products associated with or causative of a disease or an undesirable condition. For example, production of an oncoprotein or viral protein can be inhibited in humans in order to prevent the disease or condition from occurring, limit the extent to which it occurs or reverse it. If the sequence of the gene to be targeted in humans is lαiown, 21-23 nt RNAs can be produced and tested for their ability to mediate RNAi in a cell, such as a human or other primate cell. Those 21-23 nt human RNA molecules shown to mediate RNAi can be tested, if desired, in an appropriate animal model to further assess their in vivo effectiveness. Additional copies of 21-23 nt RNAs shown to mediate RNAi can be produced by the methods described herein. The method of obtaining the 21-23 nt RNA sequence using the Drosophila in vitro system can further comprise isolating the RNA sequence from the combination. The 21-23 nt RNA molecules can be isolated using a number of techniques known to those of skill in the art. For example, gel electrophoresis can be used to separate 21-23 nt RNAs from the combination, gel slices comprising the RNA sequences removed and RNAs eluted from the gel slices. Alternatively, non-denaturing methods, such as non-denaturing column chromatography, can be used to isolate the RNA produced, hi addition, chromatography (e.g., size exclusion chromatography), glycerol gradient centrifugation, affinity purification with antibody can be used to isolate 21 -23 nt RNAs. The RNA-protein complex isolated from the Drosophila in vitro system can also be used directly in the methods described herein (e.g., method of mediating RNAi of mRNA of a gene). Soluble extracts derived from Drosophila embryo that mediate or RNAi are encompassed by the invention. The soluble Drosophila extract can be obtained in a variety of ways. For example, the soluble extract canhe obtained from syncytial blastoderm Drosopliila embryos as described in Examples 1, 2, and 3. Soluble extracts can be derived from other cells in which RNAi occurs. Alternatively, soluble extracts can be obtained from a cell that does not carry out RNAi. In this instance, the factors needed to mediate RNAi can be introduced into such a cell and the soluble extract is then obtained. The components of the extract can also be chemically synthesized and/or combined using methods known in the art.
Any dsRNA can be used in the methods of the present invention, provided that it has sufficient homology to the targeted gene to mediate RNAi. The sequence of the dsRNA for use in the methods of the present invention need not be known. Alternatively, the dsRNA for use in the present invention can correspond to a known sequence, such as that of an entire gene (one or more) or portion thereof. There is no upper limit on the length of the dsRNA that can be used. For example, the dsRNA can range from about 21 base pairs (bp) of the gene to the full length of the gene or more. In one embodiment, the dsRNA used in the methods of the present invention is about 1000 bp in length. In another embodiment, the dsRNA is about 500 bp in length. In yet another embodiment, the dsRNA is about 22 bp in length. The 21 to 23 nt RNAs described herein can be used in a variety of ways. For example, the 21 to 23 nt RNA molecules can be used to mediate RNA interference of mRNA of a gene in a cell or organism, h a specific embodiment, the 21 to 23 nt RNA is introduced into human cells or a human in order to mediate RNA interference in the cells or in cells in the individual, such as to prevent or treat a disease or undesirable condition, hi this method, a gene (or genes) that cause or contribute to the disease or undesirable condition is targeted and the corresponding mRNA (the transcriptional product of the targeted gene) is degraded by RNAi. In this embodiment, an RNA of about 21 to about 23 nucleotides that targets the corresponding mRNA (the mRNA of the targeted gene) for degradation is introduced into the cell or organism. The cell or organism is maintained under conditions under which degradation of the corresponding mRNA occurs, thereby mediating RNA interference of the mRNA of the gene in the cell or organism. In a particular embodiment, the method of mediating RNA interference of a gene in a cell comprises combining double-stranded RNA that corresponds to a sequence of the gene with a soluble extract derived from Drosophila embryo, thereby producing a combination. The combination is maintained under conditions in which the double-stranded RNA is processed to RNA of about 21 to about 23 nucleotides. The 21 to 23 nt RNA is then isolated and introduced into the cell or organism. The cell or organism is maintained under conditions in which degradation of mRNA of the gene occurs, thereby mediating RNA interference of the gene in the cell or organism, hi the event that the 21-23nt RNA is introduced into a cell in which RNAi, does not nonnally occur, the factors needed to mediate RNAi are introduced into such a cell or the expression of the needed factors is induced in such a cell. Alternatively, 21 to 23 nt RNA produced by other methods (e.g., chemical synthesis, recombinant DNA production) to have a composition the same as or sufficiently similar to a 21 to 23 nt RNA known to mediate RNAi can be similarly used to mediate RNAi. Such 21 to 23 nt RNAs can be altered by addition, deletion, substitution or modification of one or more nucleotides and/or can comprise non-nucleotide materials. A further embodiment of this invention is an ex vivo method of treating cells from an individual to degrade a gene(s) that causes or is associated with a disease or undesirable condition, such as leukemia or AIDS, hi this embodiment, cells to be treated are obtained from the individual using known methods (e.g., phlebotomy or collection of bone marrow) and 21-23 nt RNAs that mediate degradation ofthe corresponding nιRNA(s) are introduced into the cells, which are then re-introduced into the individual. If necessary, biochemical components needed for RNAi to occur can also be introduced into the cells.
The mRNA of any gene can be targeted for degradation using the methods of mediating interference of mRNA described herein. For example, any cellular or viral mRNA, can be targeted, and, as a result, the encoded protein (e.g., an oncoprotein, a viral protein), expression will be diminished, h addition, the mRNA of any protein associated with/causative of a disease or undesirable condition can be targeted for degradation using the methods described herein.
The present invention also relates to a method of examining the function of a gene in a cell or organism. In one embodiment, an RNA sequence of about 21 to about 23 nucleotides that targets mRNA ofthe gene for degradation is introduced into the cell or organism. The cell or organism is maintained under conditions under which degradation of mRNA ofthe gene occurs. The phenotype ofthe cell or organism is then observed and compared to an appropriate control, thereby providing information about the function ofthe gene, hi another embodiment, double-stranded RNA that corresponds to a sequence ofthe gene is combined with a soluble extract derived from Drosophila embryo under conditions in which the double-stranded RNA is processed to generate RNA of about 21 to about 23 nucleotides. The RNA of about 21 to about 23 nucleotides is isolated and then introduced into the cell or organism. The cell or organism is maintained under conditions in which degradation ofthe mRNA ofthe gene occurs. The phenotype of the cell or organism is then observed and compared to an appropriate control, thereby identifying the function ofthe gene.
A further aspect of this invention is a method of assessing the ability of 21-23 nt RNAs to mediate RNAi and, particularly, determining which 21-23 nt RNA(s) most efficiently mediate RNAi. In one embodiment ofthe method, dsRNA corresponding to a sequence of an mRNA to be degraded is combined with detectably labeled (e.g., end-labeled, such as radiolabeled) mRNA and the soluble extract of this invention, thereby producing a combination. The combination is maintained under conditions under which the double-stranded RNA is processed and the mRNA is degraded. The sites ofthe most effective cleavage are mapped by comparing the migration ofthe labeled mRNA cleavage products to markers of known length. 21 mers spanning these sites are then designed and tested for their efficiency in mediating RNAi.
Alternatively, the extract ofthe present invention can be used to determine whether there is a particular segment or particular segments ofthe mRNA corresponding to a gene which are more efficiently targeted by RNAi than other regions and, thus, can be especially useful target sites. In one embodiment, dsRNA corresponding to a sequence of a gene to be degraded, labeled mRNA ofthe gene is combined with a soluble extract that mediates RNAi, thereby producing a combination. The resulting combination is maintained under conditions under which the dsRNA is degraded and the sites on the mRNA that are most efficiently cleaved are identified, using known methods, such as comparison to known size standards on a sequencing gel.
OVERVIEW OF EXAMPLES
Biochemical analysis of RNAi has become possible with the development of the in vitro Drosophila embryo lysate that recapitulates dsRNA-dependent silencing of gene expression described in Example 1 (Tuschl et al., Genes Dev., 13:3191-7 (1999)). In the in vitro system, dsRNA, but not sense or asRNA, targets a corresponding mRNA for degradation, yet does not affect the stability of an unrelated control mRNA. Furthermore, pre-incubation ofthe dsRNA in the lysate potentiates its activity for target mRNA degradation, suggesting that the dsRNA must be converted to an active form by binding proteins in the extract or by covalent modification (Tuschl et al, Genes Dev., 13:3191-7 (1999)).
The. development of a cell-free system from syncytial blastoderm Drosophila embryos that recapitulates many ofthe features of RNAi is described herein. The interference observed in this reaction is sequence-specific, is promoted by dsRNA, but not by single-stranded RNA, functions by specific mRNA degradation, requires a minimum length of dsRNA and is most efficient with long dsRNA. Furthermore, preincubation of dsRNA potentiates its activity. These results demonstrate that RNAi is mediated by sequence specific processes in soluble reactions. As described in Example 2, the in vitro system was used to analyze the requirements of RNAi and to determine the fate ofthe dsRNA and the mRNA. RNAi in vitro requires ATP, but does not require either mRNA translation or recognition ofthe 7-methyl-guanosine cap ofthe targeted mRNA. The dsRNA, but not single-stranded RNA, is processed in vitro to a population of 21-23 nt species. Deamination of adenosines within the dsRNA does not appear to be required for formation ofthe 21-23 nt RNAs. As described herein, the mRNA is cleaved only in the region corresponding to the sequence ofthe dsRNA and that the mRNA is cleaved at 21-23 nt intervals, strongly indicating that the 21-23 nt fragments from the dsRNA are targeting the cleavage ofthe mRNA. Furthermore, as described in Examples 3 and 4, when the 21-23 nt fragments are purified and added back to the soluble extract, they mediate RNA.
The present invention is illustrated by the following examples, which are not intended to be limiting in any way.
Example 1 Targeted mRNA degradation by double-stranded RNA in vitro Materials and Methods RNAs
Rr-Luc mRNA consisted ofthe 926 nt Rr luciferase coding sequence flanked by 25 nt of 5' untranslated sequence from the ρSP64 plasmid polylinker and 25 nt of 3' untranslated sequence consisting of 19 nt of pSP64 plasmid polylinker sequence followed by a 6 nt Sac I site. Pp-Luc mRNA contained the 1653 nt Pp luciferase coding sequence with a Kpn I site introduced immediately before the Pp luciferase stop codon. The Pp coding sequence was flanked by 5' untranslated sequences consisting of 21 nt of pSP64 plasmid polylinker followed by the 512 nt ofthe 5' untranslated region (UTR) from the Drosophila hunchback mRNA and 3' untranslated sequences consisting ofthe 562 nt hunchback 3' UTR followed by a 6 nt Sac I site. The hunchback 3' UTR sequences used contained six G-to-U mutations that disrupt function ofthe Nanos Response Elements in vivo and in vitro. Both reporter mRNAs terminated in a 25 nt poly(A) tail encoded in the transcribed plasmid. For both Rx-Luc and Pp -Luc mRNAs, the transcripts were generated by run-off transcription from plasmid templates cleaved at an Nsi I site that immediately followed the 25 nt encoded poly(A) tail. To ensure that the transcripts ended with a poly(A) tail, the Nsi I-cleaved transcription templates were resected with T4 DNA Polymerase in the presence of dNTPs. The SP6 mMessage mMachine kit (A bion) was used for in vitro transcription. Using this kit, about 80% ofthe resulting transcripts are 7-methyl guanosine capped. 32P-radiolabeling was accomplished by including α-32P-UTP in the transcription reaction.
For Pp -Luc, ss, as, and dsRNA corresponded to positions 93 to 597 relative to the start of translation, yielding a 505 bp dsRNA. For Rr -Luc, ss, as, and dsRNA corresponded to positions 118 to 618 relative to the start of translation, yielding a 501 bp dsRNA. The Drosophila nanos competitor dsRNA corresponded to positions 122 to 629 relative to the start of translation, yielding a 508 bp dsRNA. ssRNA, asRNA, and dsRNA (diagrammed in Figure 1) were transcribed in vitro with T7 RNA polymerase from templates generated by the polymerase chain reaction. After gel purification ofthe T7 RNA transcripts, residual DNA template was removed by treatment with RQl DNase (Promega). The RNA was then extracted with phenol and chloroform, and then precipitated and dissolved in water. RNA annealing and native gel electrophoresis. ssRNA and asRNA (0.5 μM) in 10 mM Tris-HCl (pH 7.5) with 20 mM NaCl were heated to 95 ° C for 1 min then cooled and annealed at room temperature for 12 to lό h. The RNAs were precipitated and resuspended in lysis buffer (below). To monitor annealing, RNAs were electrophoresed in a 2% agarose gel in TBE buffer and stained with ethidium bromide (Sambrook et al, Molecular Cloning. Cold Spring Harbor Laboratory Press, Plainview, NY. (1989)). Lysate preparation
Zero- to two-hour old embryos from Oregon R flies were collected on yeasted molasses agar at 25°C. Embryos were dechorionated for 4 to 5 min in 50% (v/v) bleach, washed with water, blotted dry, and transferred to a chilled Potter-Elvehjem tissue grinder (Kontes). Embryos were lysed at 4°C in one ml of lysis buffer (100 mM potassium acetate, 30 mM HEPES-KOH, pH 7.4, 2 mM magnesium acetate) containing 5 mM dithiothreitol (DTT) and 1 mg/ml Pefabloc SC (Boehringer-Mannheim) per gram of damp embryos. The lysate was centrifuged for 25 min at 14,500 x g at 4° C, and the supernatant flash frozen in aliquots in liquid nitrogen and stored at -80°C.
Reaction conditions
Lysate preparation and reaction conditions were derived from those described by Hussain and Leibowitz (Hussain and Leibowitz, Gene 46: 13-23 (1986)). Reactions contained 50% (v/v) lysate, mRNAs (10 to 50 pM final concentration), and 10% (v/v) lysis buffer containing the ssRNA, asRNA, or dsRNA (10 nM final concentration). Each reaction also contained 10 mM creatine phosphate, 10 μg/ml creatine phosphokinase, 100 μM GTP, 100 μM UTP, 100 μM CTP, 500 μM ATP, 5 μM DTT, 0.1 U/mL RNasin (Promega), and 100 μM of each amino acid. The final concentration of potassium acetate was adjusted to 100 mM. For standard conditions, the reactions were assembled on ice and then pre-incubated at 25° C for 10 min before adding mRNA. After adding mRNAs, the incubation was continued for an additional 60 min. The 10 min preincubation step was omitted for the experiments in Figures 3A-3C and 5A-5C. Reactions were quenched with four volumes of 1.25x Passive Lysis Buffer (Promega). Pp and Rr luciferase activity was detected in a Monolight 2010 Luminometer (Analytical Luminescence Laboratory) using the Dual-Luciferase Reporter Assay System (Promega).
RNA stability
Reactions with 32P-radiolabeled mRNA were quenched by the addition of 40 volumes of 2x PK buffer (200 mM Tris-HCl, pH 7.5, 25 mM EDTA, 300 mM NaCl, 2% w/v sodium dodecyl sulfate). Proteinase K (E.M. Merck; dissolved in water) was added to a final concentration of 465 μg/ml. The reactions were then incubated for 15 min at 65° C, extracted with phenol/chloroform/isoamyl alcohol (25:24:1), and precipitated with an equal volume of isopropanol. Reactions were analyzed by electrophoresis in a formaldehyde/agarose (0.8% w/v) gel (Sambrook et al.,
Molecular Cloning. Cold Spring Harbor Laboratory Press, Plainview, NY. (1989)). Radioactivity was detected by exposing the agarose gel [dried under vacuum onto Nytran Plus membrane (Amersham)] to an image plate (Fujix) and quantified using a Fujix Bas 2000 and Image Gauge 3.0 (Fujix) software.
Commercial lysates
Untreated rabbit reticulocyte lysate (Ambion) and wheat germ extract (A bion) reactions were assembled according to the manufacturer's directions. dsRNA was incubated in the lysate at 27°C (wheat germ) or 30°C (reticulocyte lysate) for 10 min prior to the addition of mRNAs.
Results and Discussion
To evaluate if dsRNA could specifically block gene expression in vitro, reporter mRNAs derived from two different luciferase genes that are unrelated both in sequence and in luciferin substrate specificity were used: Renilla reniformis (sea pansy) luciferase (Rr-Luc) and Photuris pennsylvanica (firefly) luciferase (Pp-Luc). dsRNA generated from one gene was used to target that luciferase mRNA whereas the other luciferase mRNA was an internal control co-translated in the same reaction. dsRNAs of approximately 500 bp were prepared by transcription of polymerase-chain reaction products from the Rr-Luc and Pp-Luc genes. Each dsRNA began -100 bp downstream ofthe start of translation (Figure 1). Sense (ss) and anti-sense (as) RNA were transcribed in vitro and annealed to each other to produce the dsRNA. Native gel electrophoresis ofthe individual Rr 501 and Pp 505 nt as RNA and ssRNA used to form the Rr and Pp dsRNAs was preformed. The ssRNA, asRNA, and dsRNAs were each tested for their ability to block specifically expression of their cognate mRNA but not the expression ofthe unrelated internal control mRNA.
The ssRNA, asRNA, or dsRNA was incubated for 10 min in a reaction containing Drosophila embryo lysate, then both Pp-Luc and Rr-Luc mRNAs were added and the incubation continued for an additional 60 min. The Drosophila embryo lysate efficiently translates exogenously transcribed mRNA under the conditions used. The amounts of Pp-Luc and Rr-Luc enzyme activities were measured and were used to calculate ratios of either Pp-Luc/Rr-Luc (Figure 2A) or Rr-Luc/Pp-Luc (Figure 2B). To facilitate comparison of different experiments, the ratios from each experiment were normalized to the ratio observed for a control in which buffer was added to the reaction in place of ssRNA, asRNA, or dsRNA.
Figure 2A shows that a 10 nM concentration ofthe 505 bp dsRNA identical to a portion ofthe sequence ofthe Pp-Luc gene specifically inhibited expression of the Pp- Luc mRNA but did not affect expression ofthe Rr-Luc internal control. Neither ssRNA nor asRNA affected expression of Pp-Luc or the Rr-Luc internal control. Thus, Pp-Luc expression was specifically inhibited by its cognate dsRNA. Conversely, a 10 nM concentration ofthe 501 bp dsRNA directed against the Rr-Luc mRNA specifically inhibited Rr-Luc expression hut not that ofthe Pp-Luc internal control (Figure 2B). Again, comparable levels of ssRNA or asRNA had little or no effect on expression of either reporter mRNA. On average, dsRNA reduced specific luciferase expression by 70% in these experiments, in which luciferase activity was measured after 1 h incubation. In other experiments in which the translational capacity ofthe reaction was replenished by the addition of fresh lysate and reaction components, a further reduction in targeted luciferase activity relative to the internal control was observed.
The ability of dsRNA but not asRNA to inhibit gene expression in these lysates is not merely a consequence ofthe greater stability ofthe dsRNA (half-life about 2 h) relative to the single-stranded RNAs (half-life - 10 min). ssRNA and asRNA transcribed with a 7-methyl guanosine cap were as stable in the lysate as uncapped dsRNA, but do not inhibit gene expression, hi contrast, dsRNA formed from the capped ssRNA and asRNA specifically blocks expression ofthe targeted mRNA.
Effective RNAi in Drosophila requires the injection of about 0.2 fmol of dsRNA into a syncytial blastoderm embryo (Kennerdell and Carthew, Cell 95:1017-1026 (1998); Carthew, wwwl.pitt.edu/~carthew/manual/RNAi_Protocol.html (1999)). Since the average volume of a Drosophila embryo is approximately 7.3 nl, this corresponds to an intracellular concentration of about 25 nM (Mazur et al., Cryobiology 25:543-544 (1988)). Gene expression in the Drosophila lysate was inhibited by a comparable concentration of dsRNA (10 nM), but lowering the dsRNA concentration ten-fold decreased the amount of specific interference. Ten nanomolar dsRNA corresponds to a 200-fold excess of dsRNA over target mRNA added to the lysate. To test if this excess of dsRNA might reflect a time- and/or concentration-dependent step in which the input dsRNA was converted to a fonn active for gene-specific interference, the effect of preincubation ofthe dsRNA on its ability to inhibit expression of its cognate mRNA was examined. Because the translational capacity ofthe lysates is significantly reduced after 30 min of incubation at 25 °C (unpublished observations), it was desired to ensure that all factors necessary for RNAi remained active throughout the pre-incubation period. Therefore, every 30 min, a reaction containing dsRNA and lysate was mixed with a fresh reaction containing unincubated lysate (Figure 3A). After six successive serial transfers spanning 3 hours of preincubation, the dsRNA, now diluted 64-fold relative to its original concentration, was incubated with lysate and 50 pM of target mRNA for 60 min. Finally, the Pp-Luc and Rr-Luc enzyme levels were measured. For comparison, the input amount of dsRNA (10 nM) was diluted 32-fold in buffer, and its capacity to generate gene-specific dsRNA interference in the absence of any preincubation step was assessed.
The preincubation ofthe dsRNA in lysate significantly potentiated its capacity to inhibit specific gene expression. Whereas the dsRNA diluted 32-fold showed no effect, the preincubated dsRNA was, within experimental error, as potent as undiluted dsRNA, despite having undergone a 64-fold dilution. Potentiation of the dsRNA by preincubation was observed for dsRNAs targeting both the Pp-Luc mRNA (Figure 3B) and the Rr-Luc mRNA (Figure 3C). Taking into account the 64-fold dilution, the activation conferred by preincubation allowed a 156 pM concentration of dsRNA to inhibit 50 pM target mRNA. Further, dilution ofthe "activated" dsRNA may be effective but has not been tested. We note that although both dsRNAs tested were activated by the preincubation procedure, each fully retained its specificity to interfere with expression only ofthe mRNA to which it is homologous. Further study ofthe reactions may provide a route to identifying the mechanism of dsRNA potentiation. One possible explanation for the observation that preincubation of the dsRNA enhances its capacity to inhibit gene expression in these lysates is that ' specific factors either modify and/or associate with the dsRNA. Accordingly, the addition of increasing amounts of dsRNA to the reaction might titrate such factors and decrease the amount of gene-specific interference caused by a second dsRNA of unrelated sequence. For both Pp-Luc mRNA and Rr-Luc mRNA, addition of increasing concentrations ofthe unrelated Drosophila nanos dsRNA to the reaction decreased the amount of gene- specific interference caused by dsRNA targeting the reporter mRNA (Figure 4). None ofthe tested concentrations of nanos dsRNA affected the levels of translation ofthe untargeted mRNA, demonstrating that the nanos dsRNA specifically titrated factors involved in gene-specific interference and not components ofthe translational machinery. The limiting factor(s) was titrated by addition of approximately 1000 nM dsRNA, a 200-fold excess over the 5 nM of dsRNA used to produce specific interference.
Interference in vitro might reflect either a specific inhibition of mRNA translation or the targeted destruction ofthe specific mRNA. To distinguish these two possibilities, the fates ofthe Pp-Luc and Rr-Luc mRNAs were examined directly using 32P-radiolabeled substrates. Stability of 10 nM Pp-Luc mRNA or Rr-Luc mRNA incubated in lysate with either buffer or 505 bp Pp-dsRNA (10 nM). Samples were deproteinized after the indicated times and the 32P-radiolabeled mRNAs were then resolved by denaturing gel electrophoresis. In the absence of dsRNA, both the Pp-Luc and Rr-Luc mRNAs were stable in the lysates, with - 75% of the input mRNA remaining after 3 h of incubation. (About 25% ofthe input mRNA is rapidly degraded in the reaction and likely represents uncapped mRNA generated by the in vitro transcription process.) In the presence of dsRNA (10 nM, 505 bp) targeting the Pp-Luc mRNA, less than 15% ofthe Pp-Luc mRNA remained after 3 h (Figure 5A). As expected, the Rr-Luc mRNA remained stable in the presence ofthe dsRNA targeting Pp-Luc mRNA. Conversely, dsRNA (10 nM, 501 bp) targeting the Rr-Luc mRNA caused the destruction ofthe Rr-Luc mRNA but had no effect on the stability of Pp-Luc mRNA (Figure 5B). Thus, the dsRNA specifically caused accelerated decay ofthe mRNA to which it is homologous with no effect on the stability ofthe unrelated control mRNA. This finding indicates that in vivo, at least in Drosophila, the effect of dsRNA is to directly destabilize the target mRNA, not to change the subcellular localization ofthe mRNA, for example, by causing it to be specifically retained in the nucleus, resulting in non-specific degradation. These results are consistent with the observation that RNAi leads to reduced cytoplasmic mRNA levels in vivo, as measured by in situ hybridization (Montgomery et al, Proc. Natl. Acad. Sci. USA 95:15502-15507 (1998)) and Northern blotting (Ngo et al, Proc. Natl. Acad. Sci. USA 95:14687-14692 (1998)). Northern blot analyses in trypanosomes and hydra suggest that dsRNA typically decreases mRNA levels by less than 90% (Ngo et al., Proc. Natl. Acad. Sci. USA 95:14687-14692 (1998); Lohmann et al., Dev. Biol. 214:211-214 (1999)). The data presented here show that in vitro mRNA levels are reduced 65 to 85% after three hours incubation, an effect comparable with observations in vivo. They also agree with the finding that RNAi in C. elegans is post- transcriptional (Montgomery et al, Proc. Natl. Acad. Sci. USA 95:15502-15507 (1998)). The simplest explanation for the specific effects on protein synthesis is that it reflects the accelerated rate of RNA decay. However, the results do not exclude independent but specific effects on translation as well as stability.
In vivo, RNAi appears to require a minimum length of dsRNA (Ngo et al., Proc. Natl. Acad. Sci., USA, 95: 14687-14692 (1998)). The ability of RNA duplexes of lengths 49 bp, 149 bp, 505 bp, and 997 bp (diagrammed in Figure 1) to target the degradation ofthe Pp-Luc mRNA in vitro was assessed, hi good agreement with in vivo observations, the 49 bp dsRNA was ineffective in vitro, while the 149 bp dsRNA enhanced mRNA decay only slightly, and both the 505 and 997 bp dsRNAs caused robust mRNA degradation (Figure 5C). 50bp dsRNA targeting other portions ofthe mRNA cause detectable mRNA degradation, though not as robust as that seen for 500bp dsRNA. Thus, although some short dsRNA do not mediate RNAi, others of approximately the same length, but different composition, will be able to do so.
Whether the gene-specific interference observed i Drosophila lysates was a general property of cell-free translation systems was examined. The effects of dsRNAs on expression of Pp-Luc and Rr-Luc mRNA were examined in commercially available wheat germ extracts and rabbit reticulocyte lysates. There was no effect of addition of 10 nM of either ssRNA, asRNA, or dsRNA on the expression of either mRNA reporter in wheat germ extracts. In contrast, the addition of 10 nM of dsRNA to the rabbit reticulocyte lysate caused a profound and rapid, non-specific decrease in mRNA stability. For example, addition of Rr-Luc dsRNA caused degradation of both Rr-Luc and Pp-Luc mRNAs within 15 min. The same non-specific effect was observed upon addition of Pp-Luc dsRNA. The non-specific destruction of mRNA induced by the addition of dsRNA to the rabbit reticulocyte lysate presumably reflects the previously observed activation of RNase L by dsRNA (Clemens and Williams, Cell 13:565-572 (1978); Williams et al, Nucleic Acids Res. 6:1335-1350 (1979); Zhou et al., Cell 72:753-765 (1993); Matthews, Interactions between Viruses and the Cellular Machinery for Protein Synthesis. In Translational Control (eds. J. Hershey, M. Mathews andN. Sonenberg), pp. 505-548. Cold Spring Harbor Laboratory Press, Plainview, NY. (1996)). Mouse cell lines lacking dsRNA-induced anti-viral pathways have recently been described (Zhou et al., Virology 258:435-440 (1999)) and maybe useful in the search for mammalian RNAi. Although RNAi is known to exist in some mammalian cells (Wianny and Zernicka-Goetz Nat. Cell Biol. 2: 70-75 (2000)), in many mammalian cell types its presence is likely obscured by the rapid induction by dsRNA of non-specific anti-viral responses. dsRNA-targeted destruction of specific mRNA is characteristic of RNAi, which has been observed in vivo in many organisms, including Drosophila. The system described above recapitulates in a reaction in vitro many aspects of RNAi. The targeted mRNA is specifically degraded whereas unrelated control mRNAs present in the same solution are not affected. The process is most efficient with dsRNAs greater than 150 bp in length. The dsRNA-specific degradation reaction in vitro is probably general to many, if not all, mRNAs since it was observed using two unrelated genes.
The magnitude ofthe effects on mRNA stability in vitro described herein are comparable with those reported in vivo (Ngo et al., Proc. Natl. Acad. Sci., USA,
95:14687-14692 (1998); Lohmann et al., Dev. Biol, 214:211-214 (1999). However, the reaction in vitro requires an excess of dsRNA relative to mRNA. contrast, a few molecules of dsRNA per cell can inhibit gene expression in vivo (Fire et al., Nature, 391: 806-811 (1998); Kennerdell and Carthew, Cell, 95:1017-1026 (1998)). The difference between the stoichiometry of dsRNA to target mRNA in vivo and in vitro should not be surprising in that most in vitro reactions are less efficient than their corresponding in vivo processes, hiterestringly, incubation ofthe dsRNA in the lysate greatly potentiated its activity for RNAi, indicating that it is either modified or becomes associated with other factors or both. Perhaps a small number of molecules is effective in inhibiting the targeted mRNA in vivo because the injected dsRNA has been activated by a process similar to that reported here for RNAi in Drosophila lysates.
Example 2 Double-Stranded RNA directs the ATP-dependent cleavage of mRNA at 21 to 23 nucleotide intervals Methods and Material hi vitro RNAi
In vitro RNAi reactions and lysate preparation were as described in Example 1 (Tuschl et al, Genes Dev., 13:3191-7 (1999)) except that the reaction contained 0.03 g/ml creatine kinase, 25 μM creatine phosphate (Fluka), and 1 mM ATP. Creatine phosphate was freshly dissolved at 500 mM in water for each experiment. GTP was omitted from the reactions, except in Figures 2 and 3.
RNA Synthesis.
Pp-luc and Rr-luc mRNAs and Pp- and Rr-dsRNAs (including dsRNA 'B' in Figure 6) were synthesized by in vitro transcription as described previously (Tuschl et al., Genes Dev., 13:3191-7 (1999)). To generate transcription templates for dsRNA 'C,' the 5' sense RNA primer was gcgtaatacgactcactataGAACAAAGGAAACGGATGAT (SEQ ID NO: 2) and the 3' sense RNA primer was GAAGAAGTTATTCTCCAAAA (SEQ ID NO: 3); the 5' asRNA primer was gcgtaatacgactcactataGAAGAAGTTATTCTCCAAAA (SEQ ID
NO: 4)and the 3' asRNA primer was GAACAAAGGAAACGGATGAT (SEQ ID
NO: 5). For dsRNA 'A' the 5' sense RNA primer was gcgtaatacgactcactataGTAGCGCGGTGTATTATACC (SEQ ID NO: 6)and the 3' sense RNA primer was GTACAACGTCAGGTTTACCA (SEQ ID NO: 7); the 5' asRNA primer was gcgtaatacgactcactataGTACAACGTCAGGTTTACCA (SEQ ID
NO: 8)and the 3' asRNA primer was GTAGCGCGGTGTATTATACC (SEQ ID
NO: 9) (lowercase, T7 promoter sequence). mRNAs were 5 '-end-labeled using guanylyl transferase (Gibco/BRL), S- adenosyl methionine (Sigma), and α-32P-GTP (3000 Ci/mmol; New England Nuclear) according to the manufacturer's directions. Radiolabeled RNAs were purified by poly(A) selection using the Poly(A) Tract HI kit (Promega).
Nonradio active 7-methyl- guanosine- and adenosine-capped RNAs were synthesized in in vitro transcription reactions with a 5-fold excess of 7-methyl-G(5')ρpp(5')G or
A(5')ppp(5')G relative to GTP. Cap analogs were purchased from New England Biolabs.
ATP depletion and Protein Synthesis Inhibition
ATP was depleted by incubating the lysate for 10 minutes at 25 °C with 2 mM glucose and 0.1 U/ml hexokinase (Sigma). Protein synthesis inhibitors were purchased from Sigma and dissolved in absolute ethanol as 250-fold concentrated stocks. The final concentrations of inhibitors in the reaction were: anisomycin, 53 mg/ml; cycloheximide, 100 mg/ml; chloramphenicol, 100 mg/ml. Relative protein synthesis was determined by measuring the activity of Rr luciferase protein produced by translation ofthe Rr-luc mRNA in the RNAi reaction after 1 hour as- described previously (Tuschl et al., Genes Dev., 13:3191-7 (1999)).
Analysis of dsRNA Processing
Internally α-32P-ATP-labeled dsRNAs (505 bp Pp-luc or 501 Rr-luc) or 7-methyl- guanosine-capped Rr-luc antisense RNA (501 nt) were incubated at 5 nM final concentration in the presence or absence of unlabeled mRNAs in Drosophila lysate for 2 hours in standard conditions. Reactions were stopped by the addition of 2x proteinase K buffer and deproteinized as described previously (Tuschl et al., Genes Dev., 13:3191- 3197 (1999)). Products were analyzed by electrophoresis in 15% or 18% polyacrylamide sequencing gels. Length standards were generated by complete RNase Tl digestion of α-32P-ATP-labeled 501 nt Rr-luc sense RNA and asRNA. For analysis of mRNA cleavage, 5'-32P-radiolabeled mRNA (described above) was incubated with dsRNA as described previously (Tuschl et al., Genes Dev., 13:3191- 3197 (1999)) and analyzed by electrophoresis in 5% (Figure 5B) and 6% (Figure 6C) polyacrylamide sequencing gels. Length standards included commercially available RNA size standards (FMC Bioproducts) radiolabeled with guanylyl transferase as described above and partial base hydrolysis and RNase Tl ladders generated from the 5 '-radiolabeled mRNA.
Deamination Assay
Internally α-32P -ATP -labeled dsRNAs (5 nM) were incubated in Drosophila lysate for 2 hours at standard conditions. After deproteinization, samples were run on 12% sequencing gels to separate full-length dsRNAs from the 21-23 nt products. RNAs were eluted from the gel slices in 0.3 M NaCl overnight, ethanol-precipitated, collected by centrifugation, and redissolved in 20 μl water. The RNA was hydrolyzed into nucleoside 5 -phosphates with nuclease PI (10 μl reaction containing 8 μl RNA in water, 30 mM KOAc pH 5.3, 10 mM ZnSO4, 10 μg or 3 units nuclease PI, 3 hours, 50° C). Samples (1 ml) were co-spotted with non-radioactive 5 -mononucleotides [0.05 O.D. units (A260) of pA, pC, pG, pi, and pU] on cellulose HPTLC plates (EM Merck) and separated in the first dimension in isobutyric acid/25% ammonia/water (66/1/33, v/v/v) and in the second dimension in 0.1M sodium phosphate, pH 6.8/ammonium sulfate/1-propanol (100/60/2, v/w/v; Silberklang et al., 1979). Migration ofthe non- radioactive internal standards was determined by UV-shadowing.
Results and Discussion RNAi Requires ATP As described in Example 1, Drosophila embryo lysates faithfully recapitulate
RNAi (Tuschl et al., Genes Dev., 13:3191-7 (1999)). Previously, dsRNA-mediated gene silencing was monitored by measuring the synthesis of luciferase protein from the targeted mRNA. Thus, these RNAi reactions contained an ATP-regenerating system, needed for the efficient translation ofthe mRNA. To test if ATP was, in fact, required for RNAi, the lysates were depleted for ATP by treatment with hexokinase and glucose, which converts ATP to ADP, and RNAi was monitored directly by following the fate of 32P -radiolabeled Renilla reniformis luciferase (Rr-luc) mRNA (Figure 6). Treatment with hexokinase and glucose reduced the endogenous ATP level in the lysate from 250 μM to below 10 μM. ATP regeneration required both exogenous creatine phosphate and creatine kinase, which acts to transfer a high-energy phosphate from creatine phosphate to ADP. When ATP-depleted extracts were supplemented with either creatine phosphate or creatine kinase separately, no RNAi was observed. Therefore, RNAi requires ATP in vitro. When ATP, creatine phosphate, and creatine kinase were all added together to reactions containing the ATP-depleted lysate, dsRNA-dependent degradation ofthe Rr-luc mRNA was restored (Figure 6). The addition of exogenous ATP was not required for efficient RNAi in the depleted lysate, provided that both creatine phosphate and creatine kinase were present, demonstrating that the endogenous concentration (250 mM) of adenosine nucleotide is sufficient to support RNAi. RNAi with a Photinus pyralis luciferase (Pp-luc) mRNA was also ATP-dependent. The stability ofthe Rr-luc mRNA in the absence of Rr-dsRNA was reduced in ATP-depleted lysates relative to that observed when the energy regenerating system was included, but decay ofthe mRNA under these conditions did not display the rapid decay kinetics characteristic of RNAi in vitro, nor did it generate the stable mRNA cleavage products characteristic of dsRNA-directed RNAi. These experiments do not establish if the ATP requirement for RNAi is direct, implicating ATP in one or more steps in the RNAi mechanism, or indirect, reflecting a role for ATP in maintaining high concentrations of another nucleoside triphosphate in the lysate.
Translation Is Not Required for RNAi In Vitro
The requirement for ATP suggested that RNAi might be coupled to mRNA translation, a highly energy-dependent process. To test this possibility, various inhibitors of protein synthesis were added to the reaction by preparing a denaturing agarose-gel analysis of 5' -32P-radio labeled Pp-luc mRNA after incubation for indicated times in a standard RNAi reaction with and without protein synthesis inhibitors. The eukaryotic translation inhibitors anisomycin, an inhibitor of initial peptide bond formation, cycloheximide, an inhibitor of peptide chain elongation, and puromycin, a tRNA mimic which causes premature termination of translation (Cundliffe, Antibiotic Inhibitors of Ribosome Function, hi The Molecular Basis of Antibiotic Action, E. Gale, E. Cundliffe, P. Reynolds, M. Richmond and M.
Warning, eds. (New York: Wiley), pp. 402-547. (1981)) were tested. Each of these inhibitors reduced protein synthesis in the Drosophila lysate by more than 1 ,900-fold (Figure 7A). In contrast, chloramphenicol, an inhibitor of Drosophila mitochondrial protein synthesis (Page and Orr-Weaver, Dev. Biol, 183:195-207 (1997)), had no effect on translation in the lysates (Figure 7A). Despite the presence of anisomycin, cycloheximide, or chloramphenicol, RNAi proceeded at normal efficiency. Puromycin also did not perturb efficient RNAi. Thus, protein synthesis is not required for RNAi in vitro.
Translational initiation is an ATP-dependent process that involves recognition ofthe 7-methyl guanosine cap ofthe mRNA (Kozak, Gene, 234:187-208 (1999); Merrick and Hershey, The Pathway and Mechanism of Eukaryotic Protein Synthesis. In Translational Control, J. Hershey, M. Mathews and N. Soneriberg, eds. (Cold Spring Harbor, NY: Cold Spring Harbor Laboratory Press), pp. 31-69 (1996)). The Drosophila lysate used to support RNAi in vitro also recapitulates the cap-dependence of translation; Pp-luc mRNA with a 7-methyl-guanosine cap was translated greater than ten-fold more efficiently than was the same mRNA with an A(5')ppp(5')G cap (Figure 7B). Both RNAs were equally stable in the Drosophila lysate, showing that this difference in efficiency cannot be merely explained by more rapid decay ofthe mRNA with an adenosine cap (see also Gebauer et al., EMBO J., 18:6146-54 (1999)). Although the translational machinery can discriminate between Pp-luc mRNAs with 7- methyl-guanosine and adenosine caps, the two mRNAs were equally susceptible to RNAi in the presence of Pp-dsRNA (Figure 7C). These results suggest that steps in cap recognition are not involved in RNAi.
dsRNA Is Processed to 21-23 nt Species RNAs 25 nt in length are generated from both the sense and anti-sense strands of genes undergoing post-transcriptional gene silencing in plants (Hamilton and Baulcombe, Science, 286:950-2 (1999)). Denaturing acrylamide-gel analysis of the products formed in a two-hour incubation of unifonnly 3 P-radiolabeled dsRNAs and capped asRNA in lysate under standard RNAi conditions, in the presence or absence of target mRNAs. It was found that dsRNA is also processed to small RNA fragments. When incubated in lysate, approximately 15% ofthe input radioactivity of both the 501 bp Rr-dsRNA and the 505 bp Pp-dsRNA appeared in 21 to 23. nt RNA fragments. Because the dsRNAs are more than 500 bp in length, the 15% yield of fragments implies that multiple.21-23 nt RNAs are produced from each full-length dsRNA molecule. No other stable products were detected. The small RNA species were produced from dsRNAs in which both strands were uniformly 32P-radiolabeled. Formation ofthe 21-23 nt RNAs from the dsRNA did not require the presence ofthe corresponding mRNA, demonstrating that the small RNA species is generated by processing ofthe dsRNA, rather than as a product of dsRNA-targeted mRNA degradation. It was noted that 22 nucleotides corresponds to two turns of an A-form RNA-RNA helix.
When dsRNAs radiolabeled within either the sense or the anti-sense strand were incubated with lysate in a standard RNAi reaction, 21-23 nt RNAs were generated with comparable efficiency. These data support the idea that the 21-23 nt RNAs are generated by symmetric processing ofthe dsRNA. A variety of data support the idea that the 21-23 nt RNA is efficiently generated only from dsRNA and is not the consequence of an interaction between single-stranded RNA and the dsRNA. First, a 32P-radiolabeled 505 nt Pp-luc sense RNA or asRNA was not efficiently converted to the 21-23 nt product when it was incubated with 5 nM nonradioactive 505 bp Pp- dsRNA. Second, in the absence of mRNA, a 501 nt 7-methyl-guanosine-caρped Rr- asRNA produced only a barely detectable amount of 21-23 nt RNA (capped single- stranded RNAs are as stable in the lysate as dsRNA, Tuschl et al., Genes Dev., 13:3191- 7 (1999)), probably due to a small amount of dsRNA contaminating the anti-sense preparation. However, when Rr-luc niRNA was included in the reaction with the 32P- radiolabeled, capped Rr-asRNA, a small amount of 21-23 nt product was generated, corresponding to 4% ofthe amount of 21-23 nt RNA produced from an equimolar amount of Rr-dsRNA. This result is unlikely to reflect the presence of contaminating dsRNA in the Rr-asRNA preparation, since significantly more product was generated from the asRNA in the presence ofthe Rr-luc mRNA than in the absence. Instead, the data suggest that asRNA can interact with the complementary mRNA sequences to form dsRNA in the reaction and that the resulting dsRNA is subsequently processed to the small RNA speeies. Rr-asRNA can support a low level of bona fide RNAi in vitro (see below), consistent with this explanation.
It was next asked if production ofthe 21-23 nt RNAs from dsRNA required ATP. When the 505 bp Pp-dsRNA was incubated in a lysate depleted for ATP by treatment with hexokinase and glucose, 21-23 nt RNA was produced, albeit 6 times slower than when ATP was regenerated in the depleted lysate by the inclusion of creatine kinase and creatine phosphate. Therefore, ATP may not be required for production ofthe 21-23 nt RNA species, but may instead simply enhance its formation. Alternatively, ATP may be required for processing ofthe dsRNA, but at a concentration less than that remaining after hexokinase treatment. The molecular basis for the slower mobility ofthe small RNA fragments generated in the ATP-depleted lysate is not understood. Wagner and Sun (Wagner and Sun, Nature, 391 :744-745 (1998)) and Sharp
(Sharp, Genes Dev., 13:139-41 (1999)) have speculated that the requirement for dsRNA in gene silencing by RNAi reflects the involvement of a dsRNA-specific adenosine deaminase in the process. dsRNA adenosine deaminases unwind dsRNA by converting adenosine to inosine, which does not base-pair with uracil. dsRNA adenosine deaminases function in the post-xranscriptional editing of mRNA (for review see Bass, Trends Biochem. Sci., 22:157-62 (1997)). To test for the involvement of dsRNA adenosine deaminase in RNAi, the degree of conversion of adenosine to inosine in the 501 bp Rr-luc and 505 bp Pp-luc dsRNAs after incubation with Drosophila embryo lysate in a standard in vitro RNAi reaction was examined. Adenosine deamination in full-length dsRNA and the 21-23 nt RNA species was assessed by two-dimensional thin-layer chromatography. Inorganic phosphate (Pj,) was produced by the degradation of monόnucleotides by phosphatases that contaminate commercially available nuclease PI (Auxilien et al., J. Mol. Biol, 262:437-458 (1996)). The degree of adenosine deamination in the 21-23 nt species was also determined. The full-length dsRNA radiolabeled with [32P]-adenosine was incubated in the lysate, and both the full-length dsRNA and the 21-23 nt RNA products were purified from a denaturing acrylamide gel, cleaved to mononucleotides with nuclease PI, and analyzed by two-dimensional thin- layer chromatography. A significant fraction ofthe adenosines in the full-length dsRNA were converted to inosine after 2 hours (3.1% and 5.6% conversion for Pp-luc and Rr-luc dsRNAs, respectively). In contrast, only 0.4% (Pp-dsRNA) or 0.7% (Rr-dsRNA) of the adenosines in the 21-23 nt species were deaminated. These data imply that fewer than 1 in 27 molecules ofthe 21-23 nt RNA species contain an inosine. Therefore, it is unlikely that dsRNA-dependent adenosine deamination within the 21-23 nt species is required for its production. asRNA Generates a Small Amount of RNAi in vitro
When mRNA was 32P-radiolabeled within the 5'-7-methyl-guanosine cap, stable 5' decay products accumulated during the RNAi reaction. Such stable 5 'decay products were observed for both the Pp-luc and Rr-luc mRNAs when they were incubated with their cognate dsRNAs. Previously, it was reported that efficient RNAi does not occur when asRNA is used in place of dsRNA (Tuschl et al., Genes Dev., 13:3191-7 (1999)). Nevertheless, mRNA was measurably less stable when incubated with asRNA than with buffer (Figures 8A and 8B). This was particularly evident for the Rr-luc mRNA: approximately 90% ofthe RNA remained intact after a 3-hour incubation in lysate, but only 50% when asRNA was added. Less than 5% remained when dsRNA was added. Interestingly, the decrease in mRNA stability caused by asRNA was accompanied by the formation of a small amount ofthe stable 5 '-decay products characteristic ofthe RNAi reaction with dsRNA. This finding parallels the observation that a small amount of 21- 23 nt product formed from the asRNA when it was incubated with the mRNA (see above) and lends strength to the idea that asRNA can enter the RNAi pathway, albeit inefficiently.
mRNA Cleavage Sites Are Determined by the Sequence of the dsRNA
The sites of mRNA cleavage were examined using three different dsRNAs, 'A,' 'B,' and 'C,' displaced along the Rr-luc sequence by approximately 100 nts. Denaturing acrylamide-gel analysis ofthe stable, 5'-cleavage products produced after incubation ofthe Rr-luc mRNA for the indicated times with each ofthe three dsRNAs, 'A,' 'B,' and 'C,' or with buffer (0) was performed. The positions of these relative to the Rr-luc mRNA sequence are shown in Figure 9. Each ofthe three dsRNAs was incubated in a standard RNAi reaction with Rr-luc mRNA 32P -radiolabeled within the 5'-cap. In the absence of dsRNA, no stable 5'-cleavage products were detected for the mRNA, even after 3 hours of incubation in lysate. In contrast, after a 20-minute incubation, each ofthe three dsRNAs produced a ladder of bands corresponding to a set of mRNA cleavage products characteristic for that particular dsRNA. For each dsRNA, the stable, 5' mRNA cleavage products were restricted to the region ofthe Rr-luc mRNA that corresponded to the dsRNA (Figures 9 and 10). For dsRNA 'A,' the lengths ofthe 5'- cleavage products ranged from 236 to just under -750 nt; dsRNA 'A spans nucleotides 233 to 729 ofthe Rr-luc mRNA. Incubation ofthe mRNA with dsRNA 'B' produced mRNA 5 '-cleavage products ranging in length from 150 to -600 nt; dsRNA 'B' spans nucleotides 143 to 644 ofthe mRNA. Finally, dsRNA 'C produced mRNA cleavage products from 66 to -500 nt in length. This dsRNA spans nucleotides 50 to 569 of the Rr-luc mRNA. Therefore, the dsRNA not only provides specificity for the RNAi reaction, selecting which mRNA from the total cellular mRNA pool will be degraded, but also determines the precise positions of cleavage along the mRNA sequence.
The mRNA Is Cleaved at 21-23 Nucleotide Intervals
To gain further insight into the mechanism of RNAi, the positions of several mRNA cleavage sites for each ofthe three dsRNAs were mapped (Figure 10). High resolution denaturing acrylamide-gel analysis of a subset ofthe 5 '-cleavage products described above was performed. Remarkably, most ofthe cleavages occurred at 21-23 nt intervals (Figure 10). This spacing is especially striking in light of our observation that the dsRNA is processed to a 21-23 nt RNA species and the finding of Hamilton and Baulcombe that a 25 nt RNA correlates with post-transcriptional gene silencing in plants (Hamilton and Baulcombe, Science, 286:950-2 (1999)). Of the 16 cleavage sites we mapped (2 for dsRNA 'A,' 5 for dsRNA 'B,' and 9 for dsRNA 'C'), all but two reflect the 21-23 nt interval. One ofthe two exceptional cleavages was a weak cleavage site produced by dsRNA 'C (indicated by an open blue circle in Figure 10). This cleavage occuned 32 nt 5' to the next cleavage site. The other exception is particularly intriguing. After four cleavages spaced 21-23 nt apart, dsRNA 'C caused cleavage ofthe mRNA just nine nt 3' to the previous cleavage site (red arrowhead in Figure 10). This cleavage occurred in a run of seven uracil residues and appears to "reset" the ruler for cleavage; the next cleavage site was 21-23 nt 3' to the exceptional site. The three subsequent cleavage sites that we mapped were also spaced 21-23 nt apart. Curiously, ofthe sixteen cleavage sites caused by the three different dsRNAs, fourteen occur at uracil residues. The significance of this finding is not understood, but it suggests that mRNA cleavage is determined by a process which measures 21-23 nt intervals and which has a sequence preference for cleavage at uracil. Results show that the 21-23 nt RNA species produced by incubation of -500 bp dsRNA in the lysate caused sequence- specific interference in vitro when isolated from an acrylamide gel and added to a new RNAi reaction in place ofthe full-length dsRNA.
A Model for dsRNA-directed mRNA Cleavage
Without wishing to be bound by theory, the biochemical data described herein, together with recent genetic experiments in C. elegans and Neurospora (Cogoni and Macino, Nature, 399: 166-9 (1999); Grishok et al., Science, 287: 2494-7 (2000); Ketting et al., Cell, 99:133-41 (1999); Tabara et al, Cell, 99:123-32 (1999)), suggest a model for how dsRNA targets mRNA for destruction (Figure 11). In this model, the dsRNA is first cleaved to 21-23 nt long fragments in a process likely to involve genes such as the C. elegans loci rde-1 and rde-4. The resulting fragments, probably as short asRNAs bound by RNAi-specific proteins, would then pair with the mRNA and recruit a nuclease that cleaves the mRNA. Alternatively, strand exchange could occur in a protein-RNA complex that transiently holds a 21-23 nt dsRNA fragment close to the mRNA. Separation ofthe two strands ofthe dsRNA following fragmentation might be assisted by an ATP-dependent RNA helicase, explaining the observed ATP enhancement of 21-23 nt RNA production.
It is likely that each small RNA fragment produces one, or at most two, cleavages in the mRNA, perhaps at the 5' or 3' ends ofthe 21-23 nt fragment. The small RNAs may be amplified by an RNA-directed RNA polymerase such as that encoded by the ego-1 gene in C. elegans (Smardon et al, Current Biology, 10:169- 178 (2000)) or the qde- 1 gene in Neurospora (Cogoni and Macino, Nature, 399:166-9 (1999)), producing long-lasting post-transcriptional gene silencing in the absence ofthe dsRNA that initiated the RNAi effect. Heritable RNAi in C. elegans requires the rde-1 and rde-4 genes to initiate, but not to persist in subsequent generations. The rde-2, rde- 3, and mut-7 genes in C. elegans are required in the tissue where RNAi occurs, but are not required for initiation of heritable RNAi (Grishok et al., Science, in press 2000). These 'effector' genes (Grishok et al., Science, in press 2000) are likely to encode proteins functioning in the actual selection of mRNA targets and in their subsequent cleavage. ATP may be required at any of a number of steps during RNAi, including complex formation on the dsRNA, strand dissociation during or after dsRNA cleavage, pairing ofthe 21-23 nt RNAs with the target mRNA, mRNA cleavage, and recycling ofthe targeting complex. Testing these ideas with the in vitro RNAi system will be an important challenge for the future. Some genes involved in RNAi are also important for transposon silencing and co-suppresion. Co-suppression is a broad biological phenomenon spanning plants, insects and perhaps humans. The most likely mechanism in Drosophila melanogaster is transcriptional silencing (Pal-Bhanra et al, Cell 99: 35-36. Thus, 21-23 nt fragments are likely to be involved in transcriptional control, as well as in post-transcriptional cotrol.
Example 3 Isolated 21-23 mers caused sequence-specific interference when added to a new RNAi reaction
Isolation of 21-23 nt fragments from incubation reaction of 500 bp dsRNA in lysate. Double-stranded RNA (500 bp from) was incubated at 10 nM concentration in Drosophila embryo lysate for 3 h at 25° C under standard conditions as described herein. After deproteinization ofthe sample, the 21-23 nt reaction products were separated from unprocessed dsRNA by denaturing polyacrylamide (15%) gel electrophoresis. For detection ofthe non-radiolabeled 21-23 nt fragments, an incubation reaction with radiolabeled dsRNA was loaded in a separate lane ofthe same gel. Gel slices containing the non-radioactive 21-23 nt fragments were cut out and the 21-23 nt fragments were eluted from the gel slices at 4° C overnight in 0.4 ml 0.3 M NaCl. The RNA was recovered from the supernatant by ethanol precipitation and centrifugation. The RNA pellet was dissolved in 10 μl of lysis buffer. As control, gel slices slightly above and below the 21-23 nt band were also cut out and subjected to the same elution and precipitation procedures. Also, a non-incubated dsRNA loaded on the 15% gel and a gel slice corresponding to 21-23 nt fragments was cut out and eluted. All pellets from the control experiments were dissolved in 10 μl lysis buffer. The losses of RNA during recovery from gel slices by elution are approx. 50%.
Incubation of purified 21-23 nt fragments in a translation-based RNAi assay
1 μl ofthe eluted 21-23 mer or control RNA solution was used for a standard 10 μl RNAi incubation reaction (see above). The 21-23 mers were preincubated in the lysate containing reaction mixture for 10 or 30 min before the addition ofthe target and control mRNA. During pre-incubation, proteins involved in RNA interference may re- associate with the 21-23 mers due to a specific signal present on these RNAs. The incubation was continued for another hour to allow translation of the target and control mRNAs. The reaction was quenched by the addition of passive lysis buffer (Promega), and luciferase activity was measured. The RNA interference is the expressed as the ratio of target to control luciferase activity normalized by an RNA-free buffer control. Specific suppression ofthe target gene was observed with either 10 or 30 minutes pre- incubation. The suppression was reproducible and reduced the relative ratio of target to control by 2-3 fold. None of the RNA fragments isolated as controls showed specific interference. For comparison, incubation of 5 nM 500 bp dsRNA (10 min pre- incubation) affects the relative ratio of control to target gene approx. 30-fold.
Stability of isolated 21-23 nt fragments in a new lysate incubation reaction. Consistent with the observation of RNAi mediated by purified 21 -23 nt RNA fragment, it was found that 35% ofthe input 21-23 nt RNA persists for more than 3 h in such an incubation reaction. This suggests that cellular factors associate with the deproteinized 21-23 nt fragments and reconstitute a functional mRNA-degrading particle. Signals connected with these 21-23 nt fragments, or their possible double stranded nature or specific lengths are likely responsible for this observation. The 21-23 nt fragments have a terminal 3' hydroxyl group, as evidenced by altered mobility on a sequencing gel following periodate treatment and beta-elimination. Example 4 21 -23 -mers purified by non-denaturing methods caused sequence-specific interference when added to a new RNAi reaction.
Fifty nanomolar double-stranded RNA (501 bp Rr-luc dsRNA, as described in example 1) was incubated in a 1 ml in vitro reaction with lysate at 25 °C (see example 1). The reaction was then stopped by the addition of an equal volume of 2x PK buffer (see example 1) and proteinase K was added to a final concentration of 1.8 μg/μl. The reaction was incubated for an additional 1 h at 25°C, phenol extracted, and then the RNAs were precipitated with 3 volumes of ethanol. The ethanol precipitate was collected by centrifugation, and the pellet was resuspended in 100 μl of lysis buffer and applied to a Superdex HR 200 10/30 gel filtration column (Pharmacia) run in lysis buffer at 0.75 ml/min. 200 μl fractions were collected from the column. Twenty μl of 3 M sodium acetate and 20 μg glycogen was added to each fraction, and the RNA was recovered by precipitation with 3 volumes of ethanol. The precipitates were resuspended in 30 μl of lysis buffer. Column profiles following the fractionation of 32P -labeled input RNA are shown in Figure 13 A.
One microliter of each resuspended fraction was tested in a 10 μl standard in vitro RNAi reaction (see example 1). This procedure yields a concentration of RNA in the in vitro RNAi reaction that is approximately equal to the concentration of that RNA species in the original reaction prior to loading on the column. The fractions were preincubated in the lysate containing reaction mixture for 30 min before the addition of 10 nM Rr-luc mRNA target and 10 nM Pp-luc control mRNA. During pre-incubation, proteins involved in RNA interference may re-associate with the 21-23-mers due to a specific signal present on these RNAs. The incubation was continued for another three hours to allow translation ofthe target and control mRNAs. The reaction was quenched by the addition of passive lysis buffer (Promega), and luciferase activity was measured. The suppression of Rr-luc mRNA target expression by the purified 21-23 nt fragments was reproducible and reduced the relative ratio of target to control by >30-fold, an amount comparable to a 50 nM 500 bp dsRNA control. Suppression of target mRNA expression was specific: little or no effect on the expression ofthe Pp-luc mRNA control was observed. The data show that the both the fractions containing uncleaved dsRNA (fractions 3 - 5) or long, partially cleaved dsRNA (fractions 7 - 13) and the fractions containing the fully processed 21-23 nt siRNAs (fractions 41 - 50) mediate effective RNA interference in vitro (Figure 13B). Suppression of target mRNA expression was specific: little or no effect on the expression ofthe Pp-luc mRNA control was observed (Figure 13C). These data, together with those in the earlier examples, demonstrate that the 21-23 nt siRNAs are (1) true intermediates in the RNAi pathway and (2) effective mediators of RNA interference in vitro.
Example 5 21 -nucleotide siRNA duplexes mediate RNA interference in human tissue cultures
Methods RNA preparation
21 nt RNAs were chemically synthesized using Expedite RNA phosphoramidites and thymidine phosphoramidite (Proligo, Germany). Synthetic oligonucleotides were deprotected and gel-purified (Elbashir, S.M., Lendeckel, W. & Tuschl, T., Genes & Dev. 15, 188-200 (2001)), followed by Sep-Pak C18 cartridge (Waters, Milford, MA, USA) purification (Tuschl, t, et al, Biochemistry, 32:11658- 11668 (1993)). The siRNA sequences targeting GL2 (Ace. X65324) and GL3 luciferase (Ace. U47296) corresponded to the coding regions 153-173 relative to the first nucleotide ofthe start codon, siRNAs targeting RL (Ace. AF025846) corresponded to region 119-129 after the start codon. Longer RNAs were transcribed with T7 RNA polymerase from PCR products, followed by gel and Sep-Pak purification. The 49 and 484 bp GL2 or GL3 dsRNAs corresponded to position 113- 161 and 113-596, respectively, relative to the start of translation; the 50 and 501 bp RL dsRNAs corresponded to position 118-167 and 118-618, respectively. PCR templates for dsRNA synthesis targeting humanized GFP (hG) were amplified from pAD3 (Kehlenbach, R.H., et al, J. Cell Biol, 747.863-874 (1998)), whereby 50 and 501 bp hG dsRNA corresponded to position 118-167 and 118-618, respectively, to the start codon. For annealing of siRNAs, 20 μM single strands were incubated in annealing buffer (100 mM potassium acetate, 30 mM HEPES-KOH at pH 7.4, 2 mM magnesium acetate) for 1 min at 90°C followed by 1 h at 37 °C. The 37 °C incubation step was extended overnight for the 50 and 500 bp dsRNAs, and these annealing reactions were performed at 8.4 μM and 0.84 μM strand concentrations, respectively.
Cell culture
S2 cells were propagated in Schneider's Drosophila medium (Life Technologies) supplemented with 10% FBS, 100 units/ml penicillin, and 100 μg/ml streptomycin at 25 °C. 293, NIH/3T3, HeLa S3, COS-7 cells were grown at 37 °C in Dulbecco's modified Eagle's medium supplemented with 10% FBS, 100 units/ml penicillin, and 100 μg/ml streptomycin. Cells were regularly passaged to maintain exponential growth. 24 h before transfection at approx. 80% confluency, mammalian cells were trypsinized and diluted 1:5 with fresh medium without antibiotics (1-3 x 105 cells/ml) and transferred to 24-well plates (500 μl/well). S2 cells were not trypsinized before splitting. Transfection was carried out with Lipofectamine 2000 reagent (Life Technologies) as described by the manufacturer for adherent cell lines. Per well, 1.0 μg pGL2-Control (Promega) or pGL3 -Control (Promega), 0.1 μgpRL- TK (Promega), and 0.28 μg siRNA duplex or dsRNA, formulated into liposomes, were apphed; the final volume was 600 μl per well. Cells were incubated 20 h after transfection and appeared healthy thereafter. Luciferase expression was subsequently monitored with the Dual luciferase assay (Promega). Transfection efficiencies were determined by fluorescence microscopy for mammalian cell lines after co- transfection of 1.1 μg liGFP-encoding pAD322 and 0.28 μg invGL2 siRNA, and were 70-90%. Reporter plasmids were amplified in XL-1 Blue (Strategene) and purified using the Qiagen EndoFree Maxi Plasmid Kit.
Results
RNA interference (RNAi) is the process of sequence-specific, post- transcriptional gene silencing in animals and plants, initiated by double-stranded RNA (dsRNA) homologous in sequence to the silenced gene (Fire, A., Trends Genet, 75:358-363 (1999); Sharp, P.A. & Zamore, P.D., Science, 287:2431-2433 (2000); Sijen, T. & Kooter, J.M., Bioessays, 22:520-531 (2000); Bass, B.L., Cell, 707:235-238 (2000); Hammond, S.M., et al, Nat. Rev. Genet, 2:110-119 (2001)). The mediators of sequence-specific mRNA degradation are 21 and 22 nt small interfering RNAs (siRNAs) generated by RNase HI cleavage from longer dsRNAs6"10 (Hamilton, AJ. &Baulcombe, D.C., Science, 286:950-952 (1999); Hammond, S.M., et al, Nature, 404:293-296 (2000); Zamore, P.D., et al, Cell, 101:25-33 (2000); Bernstein, E., et al, Naature, 409:363-366 (2001); Elbashir, S.M., et al, Genes & Dev., 75:188-200 (2001)). As shown herein, 21 nt siRNA duplexes are able to specifically suppress reporter gene expression in multiple mammalian tissue cultures, including human embryonic kidney (293) and HeLa cells. In contrast to 50 or 500 bp dsRNAs, siRNAs do not activate the interferon response. These results indicate that siRNA duplexes are a general tool for sequence-specific inactivation of gene function in mammalian cells.
Base-paired 21 and 22 nt siRNAs with overhanging 3' ends mediate efficient sequence-specific mRNA degradation in lysates prepared from D. melanogaster embryos (Elbashir, S.M., et al, Genes & Dev., 75:188-200 (2001)). To test whether siRNAs are also capable of mediating RNAi in tissue culture, 21 nt siRNA duplexes with symmetric 2 nt 3' overhangs directed against reporter genes coding for sea pansy (Renilla reniformis) and two sequence variants of firefly (Photinus pyralis, GL2 and GL3) luciferases (Figures 14A, 14B) were constructed. The siRNA duplexes were co-transfected with the reporter plasmid combhiations pGL2/pRL or pGL3/pRL, into D. melanogaster Schneider S2 cells or mammalian cells using cationic liposomes. Luciferase activities were determined 20 h after transfection. hi all cell lines tested, specific reduction ofthe expression ofthe reporter genes in the presence of cognate siRNA duplexes was observed (Figures 15A-15J). Remarkably, the absolute luciferase expression levels were unaffected by non-cognate siRNAs, indicating the absence of harmful side effects by 21 nt RNA duplexes (e.g. Figures 16A-16D, for HeLa cells). In D. melanogaster S2 cells (Figures 15A, 15B), the specific inhibition of luciferases was complete, and similar to results previously obtained for longer dsRNAs (Hammond, S.M., et al, Nature, 404:293-296 (2000); Caplen, N.J., et al, Gene, 252:95-105 (2000); Clemens, M & Williams, B., Cell, i3:565-572 (1978); Ui-Tei, K., et al, FEBS Letters, 479:79-82 (2000)). In mammalian cells, where the reporter genes were 50- to 100-fold stronger expressed, the specific suppression was less complete (Figures 15C-15J). GL2 expression was reduced 3- to 12-fold, GL3 expression 9- to 25-fold, and RL expression 1- to 3-fold, in response to the cognate siRNAs. For 293 cells, targeting of RL luciferase by RL siRNAs was ineffective, although GL2 and GL3 targets responded specifically (Figures 151, 15J). It is likely that the lack of reduction of RL expression in 293 cells is due to its 5- to 20-fold higher expression compared to any other mammalian cell line tested and/or to limited accessibility ofthe target sequence due to RNA secondary structure or associated proteins. Nevertheless, specific targeting of GL2 and GL3 luciferase by the cognate, siRNA duplexes indicated that RNAi is also functioning in 293 cells. The 2 nt 3' overhang in all siRNA duplexes, except for uGL2, was composed of (2'-deoxy) thymidine. Substitution of uridine by thymidine in the 3' overhang was well tolerated in the D. melanogaster in vitro system, and the sequence ofthe overhang was uncritical for target recognition (Elbashir, S.M., et al, Genes & Dev., 75:188-200 (2001)). The thymidine overhang was chosen, because it is supposed to enhance nuclease resistance of siRNAs in the tissue culture medium and within transfected cells. Indeed, the thymidine-modified GL2 siRNA was slightly more potent than the unmodified uGL2 siRNA in all cell lines tested (Figures 15 A, 15C, 15E, 15G, 151). It is conceivable that further modifications ofthe 3' overhanging nucleotides will provide additional benefits to the delivery and stability of siRNA duplexes.
In co-transfection experiments, 25 nM siRNA duplexes with respect to the final volume of tissue culture medium were used (Figures 15A-15J, 16A-16F). Increasing the siRNA concentration to 100 nM did not enhance the specific silencing effects, but started to affect transfection efficiencies due to competition for liposome encapsulation between plasmid DNA and siRNA. Decreasing the siRNA concentration to 1.5 nM did not reduce the specific silencing effect, even though the siRNAs were now only 2- to 20-fold more concentrated than the DNA plasmids. This indicates that siRNAs are extraordinarily powerful reagents for mediating gene silencing, and that siRNAs are effective at concentrations that are several orders of magnitude below the concentrations applied in conventional antisense or ribozyme gene targeting experiments. order to monitor the effect of longer dsRNAs on mammalian cells, 50 and 500 bp dsRNAs cognate to the reporter genes were prepared. As non-specific control, dsRNAs from humanized GFP (hG) (Kehlenbach, R.H., et al, J. Cell Biol, 141:863-874 (1998)) was used. When dsRNAs were co-transfected, in identical amounts (not concentrations) to the siRNA duplexes, the reporter gene expression was strongly and unspecifically reduced. This effect is illustrated for HeLa cells as a representative example (Figures 16A-16D). The absolute luciferase activities were decreased unspecifically 10- to 20-fold by 50 bp dsRNA, and 20- to 200-fold by 500 bp dsRNA co-transfection, respectively. Similar unspecific effects were observed for COS-7 and NIH/3T3 cells. For 293 cells, a 10- to 20-fold unspecific reduction was observed only for 500 bp dsRNAs. Unspecific reduction in reporter gene expression by dsRNA > 30 bp was expected as part ofthe interferon response (Matthews, M., Interactions between viruses and the cellular machinery for protein synthesis in Translational Control (eds., Hershey, J., Matthews,M. & Sonenberg, N.) 505-548 (Cold Spring Harbor Laboratory Press, Plainview, NY; 1996); Kumar, M. &
Carmichael, G.G., Microbiol. Mol. Biol. Rev., 52:1415-1434 (1998); Stark, G.R., et al, Annu. Rev. Biochem., 67:227-264 (1998)). Surprisingly, despite the strong unspecific decrease in reporter gene expression, additional sequence-specific, dsRNA-mediated silencing were reproducibly detected. The specific silencing effects, however, were only apparent when the relative reporter gene activities were nonnalized to the hG dsRNA controls (Figures 16E, 16F). A 2- to 10-fold specific reduction in response to cognate dsRNA was observed, also in the other three mammalian cell lines tested. Specific silencing effects with dsRNAs (356-1662 bp) were previously reported in CHO-K1 cells, but the amounts of dsRNA required to detect a 2- to 4-fold specific reduction were about 20-fold higher than in our experiments (Ui-Tei, K., et al, FEBS Letters, 479:79-82 (2000)). Also, CHO-K1 cells appear to be deficient in the interferon response, hi another report, 293, NIH/3T3, and BHK-21 cells were tested for RNAi using luciferase/lacZ reporter combinations and 829 bp specific lacZ or 717 bp unspecific GFP dsRNA(Caplen, N.J., et al, Gene, 252:95-105 (2000)). The failure of detecting RNAi in this case is likely due to the less sensitive luciferase/lacZ reporter assay and the length differences of target and control dsRNA. Taken together, the results described herein indicate that RNAi is active in mammalian cells, but that the silencing effect is difficult to detect if the interferon system is activated by dsRNA >30 bp.
The mechanism ofthe 21 nt siRNA-mediated interference process in mammalian cells remains to be uncovered, and silencing may occur post- transcriptional and/or transcriptional. hi D. melanogaster lysate, siRNA duplexes mediate post-transcriptional gene silencing by reconstitution of a siRNA-protein complexes (siRNPs), which are guiding mRNA recognition and targeted cleavage (Hammond, S.M., et al, Nature, 404:293-296 (2000); Zamore, P.D., et al, Cell, 101:25-33 (2000); Elbashir, S.M., et al, Genes & Dev., 75:188-200 (2001)). In plants, dsRNA-mediated post-transcriptional silencing has also been linked to RNA- directed DNA methylation, which may also be directed by 21 nt siRNAs (Wassenegger, M., Plant Mol. Biol, 43:203-220 (2000); Finnegan, E.J., et al, Curr. Biol, 77.R99-R102 (2000)). Methylation of promoter regions can lead to transcriptional silencing (Metter, M.F., et al, EMBO J., 19:5194-5201 (2000)), but methylation in coding sequences must not (Wang, M.-B., RNA, 7.T6-28 (2001)). DNA methylation and transcriptional silencing in mammals are well-documented processes (Kass, S.U., et al, Trends Genet, 73:444-449 (1997); Razin, A., EMBO J, 77:4905-4908 (1998)), yet they have not been linked to post-transcriptional silencing. Methylation in mammals is predominantly directed towards CpG residues. Because there is no CpG in the RL siRNA, but RL siRNA mediates specific silencing in mammalian tissue culture, it is unlikely that DNA methylation is critical for our observed silencing process. In summary, described herein, is siRNA- mediated gene silencing in mammalian cells. The use of 21 nt siRNAs holds great promise for inactivation of gene function in human tissue culture and the development of gene-specific therapeutics. While this invention has been particularly shown and described with reference to preferred embodiments thereof, it will be understood by those skilled in the art that various changes in form and details may be made therein without departing from the scope ofthe invention encompassed by the appended claims

Claims (50)

CLAIMSWhat is claimed is:
1. Isolated RNA of from about 21 to about 23 nucleotides that mediates RNA interference of an mRNA to which it corresponds.
2. Isolated RNA of claim 1 that comprises a temiinal 3' hydroxyl group.
3. Isolated RNA of claim 1 which is chemically synthesized RNA or an analog of a naturally occurring RNA.
4. An analog of isolated RNA of claim 1, wherein the analog differs from the RNA of claim 1 by the addition, deletion, substitution or alteration of one or more nucleotides.
5. Isolated RNA of from about 21 to about 23 nucleotides that inactivates a corresponding gene by transcriptional silencing.
6. A soluble extract that mediates RNA interference.
7. The soluble extract of Claim 6, wherein the extract is derived from Drosophila embryos.
8. The soluble extract of Claim 7 wherein the extract is derived from syncytial blastoderm Drosophila embryos.
9. A method of producing RNA of from about 21 to about 23 nucleotides in length comprising: (a) combining double-stranded RNA with a soluble extract that mediates
RNA interference, thereby producing a combination; and (b) mamtaining the combination of a) under conditions in which the double-stranded RNA is processed to RNA of from about 21 to about 23 nucleotides in length.
10. The method of Claim 9, wherein the soluble extract is derived from syncytial blastoderm Drosophila embryos.
11. The method of Claim 9 further comprising isolating the RNA of from about 21 to about 23 nucleotides from the combination.
12. RNA of about 21 to about 23 nucleotides produced by the method of Claim 9.
13. A method of producing RNA of from about 21 to about 23 nucleotides in length that mediates RNA interference of mRNA of a gene to be degraded, comprising:
(a) combining double-stranded RNA that corresponds to a sequence of the gene to be degraded with a soluble extract that mediates RNA interference, thereby producing a combination; and
(b) maintaining the combination of (a) under conditions under which the double-stranded RNA is processed to RNA of from about 21 to about 23 nucleotides that mediates RNA interference ofthe mRNA ofthe gene to be degraded, thereby producing RNA of from about 21 to about 23 nucleotides that mediates RNA interference ofthe mRNA.
14. The method of Claim 13, wherein the soluble extract is derived from syncytial blastoderm Drosophila embryos.
15. The method of Claim 13 further comprising isolating RNA of from about 21 to about 23 nucleotides from the combination.
16. Isolated RNA of from about 21 to about 23 nucleotides produced by the method of Claim 15.
17. A method of mediating RNA interference of mRNA of a gene in a cell or organism comprising: (a) introducing RNA of from about 21 to about 23 nucleotides which targets the mRNA ofthe gene for degradation into the cell or organism;
(b) maintaining the cell or organism produced in (a) under conditions under winch degradation ofthe mRNA occurs, thereby mediating RNA interference ofthe mRNA ofthe gene in the cell or organism.
18. The method of Claiml 7 wherein the RNA of (a) is a chemically synthesized RNA or an analog of naturally occurring RNA.
19. The method of Claim 17, wherein the gene encodes a cellular mRNA or a viral mRNA.
20. A method of mediating RNA interference of mRNA of a gene in a cell or organism in which RNA interference occurs, comprising: (a) combining double-stranded RNA that corresponds to a sequence of the gene with a soluble extract that mediates RNA interference, thereby producing a combination; (b) maintaining the combination produced in (a) under conditions under which the double- stranded RNA is processed to RNA of from about 21 to about 23 nucleotides, thereby producing RNA of from about 21 to about 23 nucleotides;
(c) isolating RNA of from about 21 to about 23 nucleotides produced in (b);
(d) introducing RNA isolated in ( c) into the cell or organism; and (e) maintaining the cell or organism produced in (d) under conditions under which degradation of mRNA ofthe gene occurs, thereby mediating RNA interference ofthe mRNA ofthe gene in the cell or organism.
21. The method of Claim 20, wherein the soluble extract is derived from syncytial blastoderm Drosophila embryos.
22. The method of Claim 20, wherein the RNA is isolated using gel electrophoresis.
23. A method of mediating RNA interference of mRNA of a gene in a cell or organism in which RNA interference occurs, comprising: (a) introducing into the cell or organism RNA of from about 21 to about 23 nucleotides that mediates RNA interference of mRNA ofthe gene, thereby producing a cell or organism that contains the RNA and (b) maintaining the cell or organism that contains the RNA under conditions under which RNA interference occurs, thereby mediatmg RNA interference of mRNA ofthe gene in the cell or organism.
24. The method of claim 23, wherein the RNA of from about 21 to about 23 nucleotides is chemically synthesized RNA or an analog of RNA that mediates RNA interference.
25. The method of Claim 23, wherein the gene encodes a cellular mRNA or a viral mRNA.
26. A knockdown cell or organism generated by the method of claim 23.
27. The knockdown cell or organism of claim 26, wherein the cell or organism mimics a disease.
28. A method of examining the function of a gene in a cell or organism comprising:
(a) introducing RNA of from about 21 to about 23 nucleotides that targets mRNA ofthe gene for degradation into the cell or organism, thereby producing a test cell or test organism;
(b) maintaining the test cell or test organism under conditions under which degradation of mRNA ofthe gene occurs, thereby producing a test cell or test organism in which mRNA ofthe gene is degraded; and (c) observing the phenotype ofthe test cell or test organism produced in
(b) and, optionally, comparing the phenotype observed to that of an appropriate control cell or control organism, thereby providing information about the function ofthe gene.
29. The method of Claim 28 wherein the RNA introduced in (a) is chemically synthesized or an analog of RNA that mediates RNA interference.
30. A method of examining the function of a gene in a cell or organism comprising
(a) combining double-stranded RNA that corresponds to a sequence of the gene with a soluble extract that mediates RNA interference, thereby producing a combination;
(b) maintaining the combination produced in (a) under conditions under which the double- stranded RNA is processed to RNA of about 21 to about 23 nucleotides, whereby RNA of about 21 to about 23 nucleotides is produced; (c) isolating RNA of about 21 to about 23 nucleotides produced in (b);
(d) introducing the RNA isolated in (c) into the cell or organism, thereby producing a test cell or test organism; (e) maintaining the test cell or test organism under conditions under which degradation of mRNA ofthe gene occurs, thereby producing a test cell or test organism in which mRNA ofthe gene is degraded; and (f) observing the phenotype of the test cell or test organism produced in
(e) and, optionally, comparing the phenotype observed to that of an appropriate control, thereby providing information about the function ofthe gene.
31. The method of claim 30, wherein the RNA comprises a terminal 3' hydroxyl group.
32. The method of claim 30, wherein the soluble extract is derived from syncytial blastoderm Drosophila embryos.
33. The method of claim 30, wherein the RNA is isolated using gel electrophoresis.
34. A composition comprising biochemical components of a Drosophila cell that process dsRNA to RNA of about 21 to about 23 nucleotides and a suitable carrier.
35. A composition comprising biochemical components of a cell that target mRNA of a gene to be degraded by RNA of about 21 to about 23 nucleotides.
36. A method of treating a disease or condition associated with the presence of a protein in an individual comprising administering to the individual RNA of from about 21 to about 23 nucleotides that targets the mRNA of the protein for degradation.
37. The method of claim 36 wherein RNA of from about 21 to about 23 nucleotides is chemically synthesized or an analog of RNA that mediates RNA interference.
38. A method of assessing whether an agent acts on a gene product comprising: (a) introducing RNA of from about 21 to about 23 nucleotides which targets the mRNA ofthe gene for degradation into a cell or organism;
(b) maintaining the cell or organism of (a) under conditions in which degradation ofthe mRNA occurs,
(c) introducing the agent into the cell or organism of (b); and (d) determining whether the agent has an effect on the cell or organism, wherein if the agent has no effect on the cell or organism then the agent acts on the gene product or on a biological pathway that involves the gene product.
39. The method of claim 38, wherein the RNA of from about 21 to about 23 nucleotides is chemically synthesized or an analog of RNA that mediates
RNA interference.
40. A method of assessing whether a gene product is a suitable target for drug discovery comprising:
(a) introducing RNA of from about 21 to about 23 nucleotides which targets the mRNA ofthe gene for degradation into a cell or organism;
(b) maintaining the cell or organism of (a) under conditions in which degradation ofthe mRNA occurs resulting in decreased expression of the gene; and
(c) determining the effect ofthe decreased expression ofthe gene on the cell or organism, wherein if decreased expression has an effect, then the gene product is a target for drug discovery.
41. The method of claim 40, wherein the RNA of from about 21 to about 23 nucleotides is synthetic RNA or an analog of RNA that mediates RNA interference.
42. A gene identified by the sequencing of endogenous 21 to 23 nucleotide RNA molecules that mediate RNA interference.
43. A pharmaceutical composition comprising RNA of from about 21 to about 23 nucleotides that mediates RNA interference and an appropriate carrier.
44. A method of producing knockdown cells, comprising introducing into cells in which a gene is to be knocked down RNA of about 21 to about 23 nt that targets the mRNA corresponding to the gene and maintaining the resulting cells under conditions under which RNAi occurs, resulting in degradation of the mRNA ofthe gene, thereby producing knockdown cells.
45. The method of claim 44, wherein the RNA of about 21 to about 23 nucleotides is synthetic RNA or an analog of RNA that mediates RNA interference.
46. A method of identifying target sites within mRNA that are efficiently cleaved by the RNAi process, comprising combining dsRNA corresponding to a sequence of a gene to be degraded, labeled mRNA corresponding to the gene and a soluble extract that mediates RNA interference, thereby producing a combination; maintaining the combination under conditions under which the dsRNA is degraded and identifying sites in the mRNA that are efficiently cleaved.
47. A method of identifying 21 -23 nt RNAs that efficiently mediate RNAi, wherein said 21-23 nt RNAs span the target sites identified within the mRNA by the method of claim 46.
48. . RNA of claim 16, isolated using gel electrophoresis.
49. RNA of claim 16, isolated using non-denaturing methods.
50. RNA of claim 16, isolated using non-denaturing column chromatography.
AU2001249622A 2000-03-30 2001-03-30 RNA sequence-specific mediators of RNA interference Expired AU2001249622B2 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
AU2007214287A AU2007214287B2 (en) 2000-03-30 2007-08-28 RNA sequence-specific mediators of RNA interference
AU2010241526A AU2010241526B2 (en) 2000-03-30 2010-11-18 RNA sequence-specific mediators of RNA interference
AU2013204199A AU2013204199C1 (en) 2000-03-30 2013-04-12 RNA sequence-specific mediators of RNA interference

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
US19359400P 2000-03-30 2000-03-30
US60/193,594 2000-03-30
EP00126325.0 2000-12-01
EP00126325 2000-12-01
US26523201P 2001-01-31 2001-01-31
US60/265,232 2001-01-31
PCT/US2001/010188 WO2001075164A2 (en) 2000-03-30 2001-03-30 Rna sequence-specific mediators of rna interference

Related Child Applications (1)

Application Number Title Priority Date Filing Date
AU2007214287A Division AU2007214287B2 (en) 2000-03-30 2007-08-28 RNA sequence-specific mediators of RNA interference

Publications (2)

Publication Number Publication Date
AU2001249622A1 true AU2001249622A1 (en) 2002-01-03
AU2001249622B2 AU2001249622B2 (en) 2007-06-07

Family

ID=38068195

Family Applications (4)

Application Number Title Priority Date Filing Date
AU2001249622A Expired AU2001249622B2 (en) 2000-03-30 2001-03-30 RNA sequence-specific mediators of RNA interference
AU4962201A Pending AU4962201A (en) 2000-03-30 2001-03-30 Rna sequence-specific mediators of rna interference
AU2010241526A Expired AU2010241526B2 (en) 2000-03-30 2010-11-18 RNA sequence-specific mediators of RNA interference
AU2013204199A Expired AU2013204199C1 (en) 2000-03-30 2013-04-12 RNA sequence-specific mediators of RNA interference

Family Applications After (3)

Application Number Title Priority Date Filing Date
AU4962201A Pending AU4962201A (en) 2000-03-30 2001-03-30 Rna sequence-specific mediators of rna interference
AU2010241526A Expired AU2010241526B2 (en) 2000-03-30 2010-11-18 RNA sequence-specific mediators of RNA interference
AU2013204199A Expired AU2013204199C1 (en) 2000-03-30 2013-04-12 RNA sequence-specific mediators of RNA interference

Country Status (17)

Country Link
US (19) US20020086356A1 (en)
EP (3) EP2345742B1 (en)
JP (6) JP5500750B2 (en)
KR (3) KR101215789B1 (en)
AT (1) ATE450621T2 (en)
AU (4) AU2001249622B2 (en)
BR (1) BR0107536A (en)
CA (1) CA2404890C (en)
CY (2) CY1109864T1 (en)
DE (1) DE60140676D1 (en)
DK (2) DK1309726T4 (en)
ES (2) ES2745378T3 (en)
HK (5) HK1161288A1 (en)
IL (4) IL151928A0 (en)
NZ (2) NZ553687A (en)
PT (2) PT1309726E (en)
WO (1) WO2001075164A2 (en)

Families Citing this family (886)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0642589A4 (en) * 1992-05-11 1997-05-21 Ribozyme Pharm Inc Method and reagent for inhibiting viral replication.
US20030206887A1 (en) * 1992-05-14 2003-11-06 David Morrissey RNA interference mediated inhibition of hepatitis B virus (HBV) using short interfering nucleic acid (siNA)
US5639647A (en) * 1994-03-29 1997-06-17 Ribozyme Pharmaceuticals, Inc. 2'-deoxy-2'alkylnucleotide containing nucleic acid
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
US7994295B2 (en) * 1997-12-22 2011-08-09 The University Of Tennessee Research Corporation Recombinant viruses comprising the membrane-proximal domain of VSV G protein
US6506559B1 (en) 1997-12-23 2003-01-14 Carnegie Institute Of Washington Genetic inhibition by double-stranded RNA
CZ295108B6 (en) 1998-03-20 2005-05-18 Benitec Australia Ltd Synthetic gene comprising dispersed or foreign deoxyribonucleic molecule and a gene construct containing such a synthetic gene
AUPP249298A0 (en) * 1998-03-20 1998-04-23 Ag-Gene Australia Limited Synthetic genes and genetic constructs comprising same I
US20030228597A1 (en) * 1998-04-13 2003-12-11 Cowsert Lex M. Identification of genetic targets for modulation by oligonucleotides and generation of oligonucleotides for gene modulation
CA2326823A1 (en) * 1998-04-20 1999-10-28 Ribozyme Pharmaceuticals, Inc. Nucleic acid molecules with novel chemical compositions capable of modulating gene expression
US20040242521A1 (en) * 1999-10-25 2004-12-02 Board Of Regents, The University Of Texas System Thio-siRNA aptamers
US20060172925A1 (en) * 1998-10-26 2006-08-03 Board Of Regents, The University Of Texas System Thio-siRNA aptamers
EP2314700A1 (en) * 1999-01-28 2011-04-27 Medical College of Georgia Research Institute, Inc Composition and method for in vivo and in vitro attenuation of gene expression using double stranded RNA
DE19956568A1 (en) 1999-01-30 2000-08-17 Roland Kreutzer Method and medicament for inhibiting the expression of a given gene
US6987025B1 (en) 1999-02-11 2006-01-17 The Arizona Board Of Regents On Behalf Of The University Of Arizona Dwf4 polynucleotides, polypeptides and uses thereof
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
IL145778A0 (en) * 1999-04-21 2002-07-25 American Home Prod Methods and compositions for inhibiting the function of polynucleotide sequences
US20040138168A1 (en) * 1999-04-21 2004-07-15 Wyeth Methods and compositions for inhibiting the function of polynucleotide sequences
US20040002153A1 (en) * 1999-07-21 2004-01-01 Monia Brett P. Modulation of PTEN expression via oligomeric compounds
US6423885B1 (en) 1999-08-13 2002-07-23 Commonwealth Scientific And Industrial Research Organization (Csiro) Methods for obtaining modified phenotypes in plant cells
GB9925459D0 (en) 1999-10-27 1999-12-29 Plant Bioscience Ltd Gene silencing
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
US7829693B2 (en) 1999-11-24 2010-11-09 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of a target gene
DE10160151A1 (en) * 2001-01-09 2003-06-26 Ribopharma Ag Inhibiting expression of target gene, useful e.g. for inhibiting oncogenes, by administering double-stranded RNA complementary to the target and having an overhang
US20050032733A1 (en) * 2001-05-18 2005-02-10 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (SiNA)
US20080039414A1 (en) * 2002-02-20 2008-02-14 Sima Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA)
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
US8273866B2 (en) * 2002-02-20 2012-09-25 Merck Sharp & Dohme Corp. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (SINA)
WO2005019453A2 (en) * 2001-05-18 2005-03-03 Sirna Therapeutics, Inc. RNA INTERFERENCE MEDIATED INHIBITION OF GENE EXPRESSION USING CHEMICALLY MODIFIED SHORT INTERFERING NUCLEIC ACID (siNA)
US20070026394A1 (en) * 2000-02-11 2007-02-01 Lawrence Blatt Modulation of gene expression associated with inflammation proliferation and neurite outgrowth using nucleic acid based technologies
US20050020525A1 (en) * 2002-02-20 2005-01-27 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA)
EP1272630A2 (en) 2000-03-16 2003-01-08 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
US20030084471A1 (en) * 2000-03-16 2003-05-01 David Beach Methods and compositions for RNA interference
GB2377221B (en) * 2000-03-17 2004-08-18 Benitec Australia Ltd Genetic silencing
ES2461715T3 (en) * 2000-03-30 2014-05-21 Whitehead Institute For Biomedical Research Specific RNA Interference Sequence RNA Mediators
IL151928A0 (en) 2000-03-30 2003-04-10 Whitehead Biomedical Inst Rna sequence-specific mediators of rna interference
US7691991B2 (en) 2000-04-17 2010-04-06 Ceres, Inc. Sequence-determined DNA fragments encoding cytochrome P450 proteins
CN1311081C (en) * 2000-08-19 2007-04-18 爱克斯澳迪亚有限公司 Stem cell differentiation
US20030190635A1 (en) * 2002-02-20 2003-10-09 Mcswiggen James A. RNA interference mediated treatment of Alzheimer's disease using short interfering RNA
US20020165192A1 (en) 2000-09-19 2002-11-07 Kerr William G. Control of NK cell function and survival by modulation of ship activity
EP1666595A1 (en) 2000-10-26 2006-06-07 Beth Israel Deaconess Medical Center, Inc. GAB2 (P97) gene and methods of use thereof
AU2013201799B2 (en) * 2000-12-01 2014-08-14 Europaisches Laboratorium Fur Molekularbiologie (Embl) Rna interference mediating small rna molecules
ES2215494T5 (en) * 2000-12-01 2017-12-28 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. Small RNA molecules that mediate RNA interference
US7385046B2 (en) 2001-01-03 2008-06-10 Ceres, Inc. Sequence-determined DNA fragments encoding ethylene responsive element binding proteins
US8546143B2 (en) 2001-01-09 2013-10-01 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of a target gene
US7767802B2 (en) 2001-01-09 2010-08-03 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of anti-apoptotic genes
US7423142B2 (en) 2001-01-09 2008-09-09 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of anti-apoptotic genes
US20020132257A1 (en) * 2001-01-31 2002-09-19 Tony Giordano Use of post-transcriptional gene silencing for identifying nucleic acid sequences that modulate the function of a cell
JP3765574B2 (en) * 2001-02-22 2006-04-12 三菱化学株式会社 Recombinant gene containing inverted repeat sequence and use thereof
WO2004111237A1 (en) * 2003-04-16 2004-12-23 Sirna Therapeutics, Inc. RNA INTERFERENCE MEDIATED INHIBITION OF PLATELET-DERIVED ENDOTHELIAL CELL GROWTH FACTOR (ECGF1) GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)
US20030124513A1 (en) * 2001-05-29 2003-07-03 Mcswiggen James Enzymatic nucleic acid treatment of diseases or conditions related to levels of HIV
US20050233344A1 (en) * 2001-05-18 2005-10-20 Sirna Therapeutics, Inc. RNA interference mediated inhibition of platelet derived growth factor (PDGF) and platelet derived growth factor receptor (PDGFR) gene expression using short interfering nucleic acid (siNA)
US20060142225A1 (en) * 2001-05-18 2006-06-29 Sirna Therapeutics, Inc. RNA interference mediated inhibition of cyclin dependent kinase-2 (CDK2) gene expression using short interfering nucleic acid (siNA)
US20050209180A1 (en) * 2001-05-18 2005-09-22 Sirna Therapeutics, Inc. RNA interference mediated inhibition of hepatitis C virus (HCV) expression using short interfering nucleic acid (siNA)
US20040019001A1 (en) * 2002-02-20 2004-01-29 Mcswiggen James A. RNA interference mediated inhibition of protein typrosine phosphatase-1B (PTP-1B) gene expression using short interfering RNA
US7109165B2 (en) * 2001-05-18 2006-09-19 Sirna Therapeutics, Inc. Conjugates and compositions for cellular delivery
US20040198682A1 (en) * 2001-11-30 2004-10-07 Mcswiggen James RNA interference mediated inhibition of placental growth factor gene expression using short interfering nucleic acid (siNA)
US20050203040A1 (en) * 2001-05-18 2005-09-15 Sirna Therapeutics, Inc. RNA interference mediated inhibition of vascular cell adhesion molecule (VCAM) gene expression using short interfering nucleic acid (siNA)
US20050164224A1 (en) * 2001-05-18 2005-07-28 Sirna Therapeutics, Inc. RNA interference mediated inhibition of cyclin D1 gene expression using short interfering nucleic acid (siNA)
US20050124566A1 (en) * 2001-05-18 2005-06-09 Sirna Therapeutics, Inc. RNA interference mediated inhibition of myostatin gene expression using short interfering nucleic acid (siNA)
US20050176666A1 (en) * 2001-05-18 2005-08-11 Sirna Therapeutics, Inc. RNA interference mediated inhibition of GPRA and AAA1 gene expression using short interfering nucleic acid (siNA)
US20050282188A1 (en) * 2001-05-18 2005-12-22 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using short interfering nucleic acid (siNA)
US7517864B2 (en) 2001-05-18 2009-04-14 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)
US20050288242A1 (en) * 2001-05-18 2005-12-29 Sirna Therapeutics, Inc. RNA interference mediated inhibition of RAS gene expression using short interfering nucleic acid (siNA)
US20050222066A1 (en) * 2001-05-18 2005-10-06 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)
US20050159378A1 (en) * 2001-05-18 2005-07-21 Sirna Therapeutics, Inc. RNA interference mediated inhibition of Myc and/or Myb gene expression using short interfering nucleic acid (siNA)
US20080161256A1 (en) * 2001-05-18 2008-07-03 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using short interfering nucleic acid (siNA)
US20050196781A1 (en) * 2001-05-18 2005-09-08 Sirna Therapeutics, Inc. RNA interference mediated inhibition of STAT3 gene expression using short interfering nucleic acid (siNA)
US20050176663A1 (en) * 2001-05-18 2005-08-11 Sima Therapeutics, Inc. RNA interference mediated inhibition of protein tyrosine phosphatase type IVA (PRL3) gene expression using short interfering nucleic acid (siNA)
WO2005078097A2 (en) * 2004-02-10 2005-08-25 Sirna Therapeutics, Inc. RNA INTERFERENCE MEDIATED INHIBITION OF GENE EXPRESSION USING MULTIFUNCTIONAL SHORT INTERFERING NUCLEIC ACID (Multifunctional siNA)
US20050261219A1 (en) * 2001-05-18 2005-11-24 Sirna Therapeutics, Inc. RNA interference mediated inhibition of interleukin and interleukin receptor gene expression using short interfering nucleic acid (siNA)
US20050164967A1 (en) * 2001-05-18 2005-07-28 Sirna Therapeutics, Inc. RNA interference mediated inhibition of platelet-derived endothelial cell growth factor (ECGF1) gene expression using short interfering nucleic acid (siNA)
US20050182007A1 (en) * 2001-05-18 2005-08-18 Sirna Therapeutics, Inc. RNA interference mediated inhibition of interleukin and interleukin receptor gene expression using short interfering nucleic acid (SINA)
US20030175950A1 (en) * 2001-05-29 2003-09-18 Mcswiggen James A. RNA interference mediated inhibition of HIV gene expression using short interfering RNA
US20050191618A1 (en) * 2001-05-18 2005-09-01 Sirna Therapeutics, Inc. RNA interference mediated inhibition of human immunodeficiency virus (HIV) gene expression using short interfering nucleic acid (siNA)
US20050196765A1 (en) * 2001-05-18 2005-09-08 Sirna Therapeutics, Inc. RNA interference mediated inhibition of checkpoint Kinase-1 (CHK-1) gene expression using short interfering nucleic acid (siNA)
US20050159380A1 (en) * 2001-05-18 2005-07-21 Sirna Therapeutics, Inc. RNA interference mediated inhibition of angiopoietin gene expression using short interfering nucleic acid (siNA)
US20050136436A1 (en) * 2001-05-18 2005-06-23 Sirna Therapeutics, Inc. RNA interference mediated inhibition of G72 and D-amino acid oxidase (DAAO) gene expression using short interfering nucleic acid (siNA)
US20050176025A1 (en) * 2001-05-18 2005-08-11 Sirna Therapeutics, Inc. RNA interference mediated inhibition of B-cell CLL/Lymphoma-2 (BCL-2) gene expression using short interfering nucleic acid (siNA)
US20050054596A1 (en) * 2001-11-30 2005-03-10 Mcswiggen James RNA interference mediated inhibition of vascular endothelial growth factor and vascular endothelial growth factor receptor gene expression using short interfering nucleic acid (siNA)
US20080188430A1 (en) * 2001-05-18 2008-08-07 Sirna Therapeutics, Inc. RNA interference mediated inhibition of hypoxia inducible factor 1 (HIF1) gene expression using short interfering nucleic acid (siNA)
US9994853B2 (en) 2001-05-18 2018-06-12 Sirna Therapeutics, Inc. Chemically modified multifunctional short interfering nucleic acid molecules that mediate RNA interference
US20050196767A1 (en) * 2001-05-18 2005-09-08 Sirna Therapeutics, Inc. RNA interference mediated inhibition of GRB2 associated binding protein (GAB2) gene expression using short interfering nucleic acis (siNA)
US20050143333A1 (en) * 2001-05-18 2005-06-30 Sirna Therapeutics, Inc. RNA interference mediated inhibition of interleukin and interleukin receptor gene expression using short interfering nucleic acid (SINA)
US20090299045A1 (en) * 2001-05-18 2009-12-03 Sirna Therapeutics, Inc. RNA Interference Mediated Inhibition Of Interleukin and Interleukin Gene Expression Using Short Interfering Nucleic Acid (siNA)
US20050171040A1 (en) * 2001-05-18 2005-08-04 Sirna Therapeutics, Inc. RNA interference mediated inhibition of cholesteryl ester transfer protein (CEPT) gene expression using short interfering nucleic acid (siNA)
US20050159381A1 (en) * 2001-05-18 2005-07-21 Sirna Therapeutics, Inc. RNA interference mediated inhibition of chromosome translocation gene expression using short interfering nucleic acid (siNA)
US20050187174A1 (en) * 2001-05-18 2005-08-25 Sirna Therapeutics, Inc. RNA interference mediated inhibition of intercellular adhesion molecule (ICAM) gene expression using short interfering nucleic acid (siNA)
US20050287128A1 (en) * 2001-05-18 2005-12-29 Sirna Therapeutics, Inc. RNA interference mediated inhibition of TGF-beta and TGF-beta receptor gene expression using short interfering nucleic acid (siNA)
US20050267058A1 (en) * 2001-05-18 2005-12-01 Sirna Therapeutics, Inc. RNA interference mediated inhibition of placental growth factor gene expression using short interfering nucleic acid (sINA)
US20060211642A1 (en) * 2001-05-18 2006-09-21 Sirna Therapeutics, Inc. RNA inteference mediated inhibition of hepatitis C virus (HVC) gene expression using short interfering nucleic acid (siNA)
US20050079610A1 (en) * 2001-05-18 2005-04-14 Sirna Therapeutics, Inc. RNA interference mediated inhibition of Fos gene expression using short interfering nucleic acid (siNA)
US20060217331A1 (en) * 2001-05-18 2006-09-28 Sirna Therapeutics, Inc. Chemically modified double stranded nucleic acid molecules that mediate RNA interference
US20050137155A1 (en) * 2001-05-18 2005-06-23 Sirna Therapeutics, Inc. RNA interference mediated treatment of Parkinson disease using short interfering nucleic acid (siNA)
US20050277133A1 (en) * 2001-05-18 2005-12-15 Sirna Therapeutics, Inc. RNA interference mediated treatment of polyglutamine (polyQ) repeat expansion diseases using short interfering nucleic acid (siNA)
US20060241075A1 (en) * 2001-05-18 2006-10-26 Sirna Therapeutics, Inc. RNA interference mediated inhibition of desmoglein gene expression using short interfering nucleic acid (siNA)
US20050119212A1 (en) * 2001-05-18 2005-06-02 Sirna Therapeutics, Inc. RNA interference mediated inhibition of FAS and FASL gene expression using short interfering nucleic acid (siNA)
US20070270579A1 (en) * 2001-05-18 2007-11-22 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using short interfering nucleic acid (siNA)
US20050158735A1 (en) * 2001-05-18 2005-07-21 Sirna Therapeutics, Inc. RNA interference mediated inhibition of proliferating cell nuclear antigen (PCNA) gene expression using short interfering nucleic acid (siNA)
US20050124569A1 (en) * 2001-05-18 2005-06-09 Sirna Therapeutics, Inc. RNA interference mediated inhibition of CXCR4 gene expression using short interfering nucleic acid (siNA)
US20050164968A1 (en) * 2001-05-18 2005-07-28 Sirna Therapeutics, Inc. RNA interference mediated inhibition of ADAM33 gene expression using short interfering nucleic acid (siNA)
US20050153914A1 (en) * 2001-05-18 2005-07-14 Sirna Therapeutics, Inc. RNA interference mediated inhibition of MDR P-glycoprotein gene expression using short interfering nucleic acid (siNA)
US20070093437A1 (en) * 2001-05-18 2007-04-26 Sirna Therapeutics, Inc. Rna interference mediated inhibition of xiap gene expression using short interfering nucleic acid (sina)
US20070042983A1 (en) * 2001-05-18 2007-02-22 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using short interfering nucleic acid (siNA)
US20050159382A1 (en) * 2001-05-18 2005-07-21 Sirna Therapeutics, Inc. RNA interference mediated inhibition of polycomb group protein EZH2 gene expression using short interfering nucleic acid (siNA)
US20050159379A1 (en) * 2001-05-18 2005-07-21 Sirna Therapeutics, Inc RNA interference mediated inhibition of gastric inhibitory polypeptide (GIP) and gastric inhibitory polypeptide receptor (GIPR) gene expression using short interfering nucleic acid (siNA)
US20050048529A1 (en) * 2002-02-20 2005-03-03 Sirna Therapeutics, Inc. RNA interference mediated inhibition of intercellular adhesion molecule (ICAM) gene expression using short interfering nucleic acid (siNA)
WO2003001877A2 (en) * 2001-06-26 2003-01-09 Gene Logic, Inc. Methods for the diagnosis and treatment of cardiac tissue rejection
IL159756A0 (en) 2001-07-12 2004-06-20 Univ Massachusetts IN VIVO PRODUCTION OF SMALL INTERFERING RNAs THAT MEDIATE GENE SILENCING
US10590418B2 (en) * 2001-07-23 2020-03-17 The Board Of Trustees Of The Leland Stanford Junior University Methods and compositions for RNAi mediated inhibition of gene expression in mammals
US20030153519A1 (en) 2001-07-23 2003-08-14 Kay Mark A. Methods and compositions for RNAi mediated inhibition of gene expression in mammals
US20030198627A1 (en) * 2001-09-01 2003-10-23 Gert-Jan Arts siRNA knockout assay method and constructs
DE10163098B4 (en) 2001-10-12 2005-06-02 Alnylam Europe Ag Method for inhibiting the replication of viruses
US7745418B2 (en) * 2001-10-12 2010-06-29 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting viral replication
WO2003035870A1 (en) * 2001-10-26 2003-05-01 Ribopharma Ag Drug for treating a carcinoma of the pancreas
US20040121348A1 (en) * 2001-10-26 2004-06-24 Ribopharma Ag Compositions and methods for treating pancreatic cancer
US20050119202A1 (en) * 2001-10-26 2005-06-02 Roland Kreutzer Medicament to treat a fibrotic disease
DE10230997A1 (en) * 2001-10-26 2003-07-17 Ribopharma Ag Drug to increase the effectiveness of a receptor-mediates apoptosis in drug that triggers tumor cells
WO2003035868A1 (en) * 2001-10-26 2003-05-01 Ribopharma Ag Medicament that increases the effectiveness of a drug that induces receptor-mediated apoptosis in tumor cells
DE10230996A1 (en) * 2001-10-26 2003-07-17 Ribopharma Ag Method for inhibiting viral replication, useful particularly for treating hepatitis C infection, by altering the 3'-untranslated region of the virus
US20040063654A1 (en) * 2001-11-02 2004-04-01 Davis Mark E. Methods and compositions for therapeutic use of RNA interference
CN1318586C (en) * 2001-11-05 2007-05-30 詹森药业有限公司 Method for the in vitro synthesis of short double stranded RNAs
FR2832154B1 (en) * 2001-11-09 2007-03-16 Centre Nat Rech Scient OLIGONUCLEOTIDES INHIBITORS AND THEIR USE FOR SPECIFICALLY REPRESSING A GENE
AU2002352797A1 (en) * 2001-11-19 2003-06-10 Proteologics, Inc. Methods for identifying and validating potential drug targets
WO2003044188A1 (en) 2001-11-21 2003-05-30 Mitsubishi Chemical Corporation Method of inhibiting gene expression
US20070203333A1 (en) * 2001-11-30 2007-08-30 Mcswiggen James RNA interference mediated inhibition of vascular endothelial growth factor and vascular endothelial growth factor receptor gene expression using short interfering nucleic acid (siNA)
US20040138163A1 (en) * 2002-05-29 2004-07-15 Mcswiggen James RNA interference mediated inhibition of vascular edothelial growth factor and vascular edothelial growth factor receptor gene expression using short interfering nucleic acid (siNA)
US7871619B2 (en) 2001-11-30 2011-01-18 Chemocentryx, Inc. Compositions and methods for detecting and treating diseases and conditions related to chemokine receptors
US20050075304A1 (en) * 2001-11-30 2005-04-07 Mcswiggen James RNA interference mediated inhibition of vascular endothelial growth factor and vascular endothelial growth factor receptor gene expression using short interfering nucleic acid (siNA)
US7294504B1 (en) 2001-12-27 2007-11-13 Allele Biotechnology & Pharmaceuticals, Inc. Methods and compositions for DNA mediated gene silencing
CA2469685C (en) 2002-01-17 2013-03-12 The University Of British Columbia Bispecific antisense oligonucleotides that inhibit igfbp-2 and igfbp-5 and methods of using same
DE10202419A1 (en) * 2002-01-22 2003-08-07 Ribopharma Ag Method of inhibiting expression of a target gene resulting from chromosome aberration
US20030166282A1 (en) 2002-02-01 2003-09-04 David Brown High potency siRNAS for reducing the expression of target genes
ATE443133T1 (en) 2002-02-01 2009-10-15 Life Technologies Corp OLIGONUCLEOTIDE COMPOSITIONS WITH IMPROVED EFFICIENCY
US20060009409A1 (en) 2002-02-01 2006-01-12 Woolf Tod M Double-stranded oligonucleotides
JP2005527198A (en) 2002-02-14 2005-09-15 シティ・オブ・ホープ Methods for producing interfering RNA molecules in mammalian cells and therapeutic uses of the interfering RNA molecules
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)
ATE519774T1 (en) * 2002-02-20 2011-08-15 Sirna Therapeutics Inc RNA DISRUPTION-MEDIATED INHIBITION OF HEPATITIS C VIRUS (HCV) GENE EXPRESSION WITH SHORT INTERFERING NUCLEIC ACID (SINA)
US8067575B2 (en) * 2002-02-20 2011-11-29 Merck, Sharp & Dohme Corp. RNA interference mediated inhibition of cyclin D1 gene expression using 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)
US20050096284A1 (en) * 2002-02-20 2005-05-05 Sirna Therapeutics, Inc. RNA interference mediated treatment of polyglutamine (polyQ) repeat expansion diseases using short interfering nucleic acid (siNA)
US20090099117A1 (en) * 2002-02-20 2009-04-16 Sirna Therapeutics, Inc. RNA INTERFERENCE MEDIATED INHIBITION OF MYOSTATIN GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)
WO2003106476A1 (en) * 2002-02-20 2003-12-24 Sirna Therapeutics, Inc Nucleic acid mediated inhibition of enterococcus infection and cytolysin toxin activity
US20050004008A1 (en) * 2002-03-01 2005-01-06 Frackelton A. Raymond SHC proteins as therapeutic targets in proliferative diseases
WO2003078959A2 (en) 2002-03-11 2003-09-25 Ortho Mcneil Pharmaceutical, Inc Methods for shp1 mediated neuroprotection
US20030180712A1 (en) 2002-03-20 2003-09-25 Biostratum Ab Inhibition of the beta3 subunit of L-type Ca2+ channels
US20040248296A1 (en) * 2002-03-20 2004-12-09 Beresford Paul J. HIV therapeutic
US7541150B2 (en) 2002-04-08 2009-06-02 University Of Louisville Research Foundation, Inc Method for the diagnosis and prognosis of malignant diseases
US7357928B2 (en) 2002-04-08 2008-04-15 University Of Louisville Research Foundation, Inc. Method for the diagnosis and prognosis of malignant diseases
JP5578388B2 (en) * 2002-04-18 2014-08-27 オプコ ファーマシューティカルズ、エルエルシー Means and methods for specific inhibition of genes in the central nervous system and / or cells and tissues of the eye
US20040180438A1 (en) 2002-04-26 2004-09-16 Pachuk Catherine J. Methods and compositions for silencing genes without inducing toxicity
AU2003234336A1 (en) * 2002-05-03 2003-11-17 The Board Of Trustees Of The Leland Stanford Junior University Methods and compositions for use in preparing sirnas
PT1504126E (en) 2002-05-03 2014-06-02 Univ Duke A method of regulating gene expression
AU2003239897A1 (en) 2002-05-23 2003-12-12 Ceptyr, Inc. Modulation of ptp1b signal transduction by rna interference
WO2003099298A1 (en) * 2002-05-24 2003-12-04 Max-Planck Gesellschaft zur Förderung der Wissenschaften e.V. Rna interference mediating small rna molecules
WO2003106617A2 (en) * 2002-06-12 2003-12-24 Tel Aviv Medical Center Research Development Fund Oligonucleotides antibodies and kits including same for treating prostate cancer and determining predisposition thereto
US20040033602A1 (en) * 2002-06-12 2004-02-19 Ambion, Inc. Methods and compositions relating to polypeptides with RNase III domains that mediate RNA interference
US20100075423A1 (en) * 2002-06-12 2010-03-25 Life Technologies Corporation Methods and compositions relating to polypeptides with rnase iii domains that mediate rna interference
US20040248094A1 (en) * 2002-06-12 2004-12-09 Ford Lance P. Methods and compositions relating to labeled RNA molecules that reduce gene expression
AU2003242688A1 (en) * 2002-06-20 2004-01-06 Dsm Ip Assets B.V. Inhibition of nuclear receptors
US20040086911A1 (en) * 2002-06-24 2004-05-06 Baylor College Of Medicine Inhibition of gene expression in vertebrates using double-stranded RNA (RNAi)
CN1678734B (en) 2002-06-27 2012-12-12 韦尔瓦制药有限公司 Differentiation modulating agents and uses thereof
DE60334618D1 (en) 2002-06-28 2010-12-02 Protiva Biotherapeutics Inc METHOD AND DEVICE FOR PREPARING LIPOSOMES
DE10229872A1 (en) * 2002-07-03 2004-01-29 Curevac Gmbh Immune stimulation through chemically modified RNA
CA2496253C (en) 2002-07-19 2016-05-10 Beth Israel Deaconess Medical Center, Inc. Methods of diagnosing and treating pre-eclampsia or eclampsia
US7435419B2 (en) 2002-07-19 2008-10-14 Beth Israel Deaconess Medical Center Methods of diagnosing and treating pre-eclampsia or eclampsia
US7148342B2 (en) 2002-07-24 2006-12-12 The Trustees Of The University Of Pennyslvania Compositions and methods for sirna inhibition of angiogenesis
US7399851B2 (en) 2002-07-25 2008-07-15 Dana Farber Cancer Institute, Inc. Composition and method for imaging cells
EP1532248B1 (en) 2002-07-26 2009-04-01 Novartis Vaccines and Diagnostics, Inc. Modified small interfering rna molecules and methods of use
US20040241854A1 (en) 2002-08-05 2004-12-02 Davidson Beverly L. siRNA-mediated gene silencing
US20040023390A1 (en) * 2002-08-05 2004-02-05 Davidson Beverly L. SiRNA-mediated gene silencing with viral vectors
HUE032289T2 (en) * 2002-08-05 2017-09-28 Silence Therapeutics Gmbh Futher novel forms of interfering RNA molecules
US20050042646A1 (en) 2002-08-05 2005-02-24 Davidson Beverly L. RNA interference suppresion of neurodegenerative diseases and methods of use thereof
US20080176812A1 (en) * 2002-08-05 2008-07-24 Davidson Beverly L Allele-specific silencing of disease genes
US20050106731A1 (en) * 2002-08-05 2005-05-19 Davidson Beverly L. siRNA-mediated gene silencing with viral vectors
DK1389637T3 (en) * 2002-08-05 2012-09-03 Silence Therapeutics Ag Interfering RNA molecules with blunt ends
US20050255086A1 (en) * 2002-08-05 2005-11-17 Davidson Beverly L Nucleic acid silencing of Huntington's Disease gene
PT1527176E (en) 2002-08-05 2007-04-30 Atugen Ag Further novel forms of interfering rna molecules
US20080274989A1 (en) * 2002-08-05 2008-11-06 University Of Iowa Research Foundation Rna Interference Suppression of Neurodegenerative Diseases and Methods of Use Thereof
CN1720257A (en) * 2002-08-06 2006-01-11 因特拉迪格姆公司 Methods of down regulating target gene expression in vivo by introduction of interfering RNA
AU2003261449A1 (en) 2002-08-07 2004-02-25 Compositions for rna interference and methods of use thereof
US20040029275A1 (en) * 2002-08-10 2004-02-12 David Brown Methods and compositions for reducing target gene expression using cocktails of siRNAs or constructs expressing siRNAs
CN1685063B (en) 2002-08-12 2011-12-21 新英格兰生物实验室公司 Methods and compositions relating to gene silencing
WO2004019973A1 (en) 2002-08-14 2004-03-11 Atugen Ag Use of protein kinase n beta
EP1393742A1 (en) 2002-08-14 2004-03-03 atugen AG Use of protein kinase N beta
JP4717633B2 (en) 2002-08-21 2011-07-06 ザ・ユニバーシティ・オブ・ブリティッシュ・コロンビア RNAi probes targeting cancer-related proteins
US9447434B2 (en) 2002-09-05 2016-09-20 California Institute Of Technology Use of chimeric nucleases to stimulate gene targeting
US7923547B2 (en) 2002-09-05 2011-04-12 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA)
US7655772B2 (en) 2002-09-06 2010-02-02 University Of South Florida Materials and methods for treatment of allergic diseases
US20080260744A1 (en) 2002-09-09 2008-10-23 Omeros Corporation G protein coupled receptors and uses thereof
US20040053289A1 (en) * 2002-09-09 2004-03-18 The Regents Of The University Of California Short interfering nucleic acid hybrids and methods thereof
MXPA05002791A (en) 2002-09-13 2005-12-05 Replicor Inc Non-sequence complementary antiviral oligonucleotides.
US20090217404A1 (en) * 2002-09-27 2009-08-27 Lowe Scott W Cell-based RNA interference and related methods and compositions
US20040242518A1 (en) * 2002-09-28 2004-12-02 Massachusetts Institute Of Technology Influenza therapeutic
AU2003279004B2 (en) * 2002-09-28 2009-10-08 Massachusetts Institute Of Technology Influenza therapeutic
US9453251B2 (en) 2002-10-08 2016-09-27 Pfenex Inc. Expression of mammalian proteins in Pseudomonas fluorescens
CA2501752A1 (en) 2002-10-10 2004-04-22 Wyeth Compositions, organisms and methodologies employing a novel human kinase
WO2004035615A2 (en) 2002-10-18 2004-04-29 Atugen Ag Factor involved in metastasis and uses thereof
US20040077082A1 (en) * 2002-10-18 2004-04-22 Koehn Richard K. RNA-based inhibitory oligonucleotides
CA2503491A1 (en) 2002-10-24 2004-05-06 Wyeth Calcineurin-like human phosphoesterase
EP1581654A4 (en) * 2002-10-28 2007-01-03 Invitrogen Corp Array oligomer synthesis and use
WO2004042024A2 (en) * 2002-11-01 2004-05-21 The Trustees Of The University Of Pennsylvania COMPOSITIONS AND METHODS FOR siRNA INHIBITION OF HIF-1 ALPHA
US7892793B2 (en) * 2002-11-04 2011-02-22 University Of Massachusetts Allele-specific RNA interference
CA2504694C (en) 2002-11-05 2013-10-01 Isis Pharmaceuticals, Inc. Polycyclic sugar surrogate-containing oligomeric compounds and compositions 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
AU2003295387A1 (en) 2002-11-05 2004-06-03 Isis Parmaceuticals, Inc. Modified oligonucleotides for use in rna interference
US9827263B2 (en) 2002-11-05 2017-11-28 Ionis Pharmaceuticals, Inc. 2′-methoxy substituted oligomeric compounds and compositions for use in gene modulations
EP1560839A4 (en) 2002-11-05 2008-04-23 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
CN1498964A (en) * 2002-11-07 2004-05-26 本元正阳基因技术股份有限公司 Serial recombined gland related virus inducible path of RNAi, and utilized in gene therapy
US7951935B2 (en) 2002-11-14 2011-05-31 Dharmacon, Inc. siRNA targeting v-myc myelocytomatosis viral oncogene homolog (MYC)
US20090227780A1 (en) * 2002-11-14 2009-09-10 Dharmacon, Inc. siRNA targeting connexin 43
US7906326B2 (en) 2003-05-07 2011-03-15 Rosetta Genomics Ltd. Bioinformatically detectable group of novel regulatory oligonucleotides associated with alzheimer's disease and uses thereof
US7612196B2 (en) * 2002-11-14 2009-11-03 Dharmacon, Inc. siRNA targeting cyclin-dependent kinase inhibitor 1B (p27, Kip1) (CDKN1B)
US7250496B2 (en) 2002-11-14 2007-07-31 Rosetta Genomics Ltd. Bioinformatically detectable group of novel regulatory genes and uses thereof
US9228186B2 (en) 2002-11-14 2016-01-05 Thermo Fisher Scientific Inc. Methods and compositions for selecting siRNA of improved functionality
US7781575B2 (en) 2002-11-14 2010-08-24 Dharmacon, Inc. siRNA targeting tumor protein 53 (p53)
US7619081B2 (en) * 2002-11-14 2009-11-17 Dharmacon, Inc. siRNA targeting coatomer protein complex, subunit beta 2 (COPB2)
US8198427B1 (en) * 2002-11-14 2012-06-12 Dharmacon, Inc. SiRNA targeting catenin, beta-1 (CTNNB1)
US20080268457A1 (en) * 2002-11-14 2008-10-30 Dharmacon, Inc. siRNA targeting forkhead box P3 (FOXP3)
US7691998B2 (en) * 2002-11-14 2010-04-06 Dharmacon, Inc. siRNA targeting nucleoporin 62kDa (Nup62)
US9719094B2 (en) 2002-11-14 2017-08-01 Thermo Fisher Scientific Inc. RNAi targeting SEC61G
US7635770B2 (en) * 2002-11-14 2009-12-22 Dharmacon, Inc. siRNA targeting protein kinase N-3 (PKN-3)
US7977471B2 (en) * 2002-11-14 2011-07-12 Dharmacon, Inc. siRNA targeting TNFα
US10011836B2 (en) 2002-11-14 2018-07-03 Thermo Fisher Scientific Inc. Methods and compositions for selecting siRNA of improved functionality
US9839649B2 (en) 2002-11-14 2017-12-12 Thermo Fisher Scientific Inc. Methods and compositions for selecting siRNA of improved functionality
US9771586B2 (en) 2002-11-14 2017-09-26 Thermo Fisher Scientific Inc. RNAi targeting ZNF205
US9719092B2 (en) 2002-11-14 2017-08-01 Thermo Fisher Scientific Inc. RNAi targeting CNTD2
US8163896B1 (en) 2002-11-14 2012-04-24 Rosetta Genomics Ltd. Bioinformatically detectable group of novel regulatory genes and uses thereof
US7592442B2 (en) * 2002-11-14 2009-09-22 Dharmacon, Inc. siRNA targeting ribonucleotide reductase M2 polypeptide (RRM2 or RNR-R2)
US9879266B2 (en) 2002-11-14 2018-01-30 Thermo Fisher Scientific Inc. Methods and compositions for selecting siRNA of improved functionality
EP1560931B1 (en) * 2002-11-14 2011-07-27 Dharmacon, Inc. Functional and hyperfunctional sirna
US20100113307A1 (en) * 2002-11-14 2010-05-06 Dharmacon, Inc. siRNA targeting vascular endothelial growth factor (VEGF)
US7655785B1 (en) 2002-11-14 2010-02-02 Rosetta Genomics Ltd. Bioinformatically detectable group of novel regulatory oligonucleotides and uses thereof
WO2006006948A2 (en) * 2002-11-14 2006-01-19 Dharmacon, Inc. METHODS AND COMPOSITIONS FOR SELECTING siRNA OF IMPROVED FUNCTIONALITY
US7064337B2 (en) 2002-11-19 2006-06-20 The Regents Of The University Of California Radiation detection system for portable gamma-ray spectroscopy
EP1588142A4 (en) 2002-11-21 2007-10-31 Wyeth Corp Methods for diagnosing rcc and other solid tumors
AU2003298718A1 (en) * 2002-11-22 2004-06-18 University Of Massachusetts Modulation of hiv replication by rna interference
JP4526228B2 (en) * 2002-11-22 2010-08-18 隆 森田 Novel therapeutic methods and therapeutic agents using RNAi
EP2112229A3 (en) 2002-11-25 2009-12-02 Sequenom, Inc. Methods for identifying risk of breast cancer and treatments thereof
US7217807B2 (en) 2002-11-26 2007-05-15 Rosetta Genomics Ltd Bioinformatically detectable group of novel HIV regulatory genes and uses thereof
US20130130231A1 (en) 2002-11-26 2013-05-23 Isaac Bentwich Bioinformatically detectable group of novel viral regulatory genes and uses thereof
US7696334B1 (en) 2002-12-05 2010-04-13 Rosetta Genomics, Ltd. Bioinformatically detectable human herpesvirus 5 regulatory gene
AU2003290664A1 (en) 2002-11-27 2004-06-23 Wei Liu Compositions, organisms and methodologies employing a novel human kinase
US20040110698A1 (en) * 2002-12-10 2004-06-10 Kimron Veterinary Institute Oligonucleotides and methods using same for treating cox-ll associated diseases
WO2004063342A2 (en) * 2003-01-09 2004-07-29 Invitrogen Corporation Cellular delivery and activation polypeptide-nucleic acid complexes
ATE477337T1 (en) * 2003-01-16 2010-08-15 Univ Pennsylvania COMPOSITIONS AND METHODS FOR SIRNA INHIBITION OF ICAM-1
US20070104688A1 (en) 2003-02-13 2007-05-10 City Of Hope Small interfering RNA mediated transcriptional gene silencing in mammalian cells
US20040171118A1 (en) * 2003-02-13 2004-09-02 City Of Hope Methods for directing DNA methylation in mammalian cells using homologous short double stranded RNAs
US20090186839A1 (en) * 2003-02-17 2009-07-23 Cold Spring Harbor Laboratory Model for studying the role of genes in chemoresistance
WO2004074445A2 (en) * 2003-02-17 2004-09-02 Cold Spring Harbor Laboratory Model for studying the role of genes in tumor resistance to chemotherapy
US7521534B1 (en) 2003-03-03 2009-04-21 The University Board Of Regents Of Texas System IKK gamma gene products and methods for making and using same
NZ542665A (en) * 2003-03-05 2008-05-30 Senesco Technologies Inc Use of antisense oligonucleotides or siRNA to suppress expression of eIF-5A1
EP1605978B1 (en) 2003-03-07 2010-09-01 Alnylam Pharmaceuticals Inc. Therapeutic compositions
GB0306715D0 (en) * 2003-03-24 2003-04-30 Novartis Ag Organic compounds
EP1608755B1 (en) 2003-04-01 2012-10-17 Helmholtz-Zentrum für Infektionsforschung GmbH Tak1-mediated inhibition of osteogenesis
JP4912873B2 (en) * 2003-04-09 2012-04-11 アルナイラム ファーマシューティカルズ, インコーポレイテッド iRNA complex
CA2521464C (en) 2003-04-09 2013-02-05 Alnylam Pharmaceuticals, Inc. Irna conjugates
WO2004092383A2 (en) * 2003-04-15 2004-10-28 Sirna Therapeutics, Inc. RNA INTERFERENCE MEDIATED INHIBITION OF SEVERE ACUTE RESPIRATORY SYNDROME (SARS) VIRUS GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)
EP2664672A1 (en) * 2003-04-17 2013-11-20 Alnylam Pharmaceuticals Inc. Modified iRNA agents
AU2004232964B2 (en) 2003-04-17 2011-09-22 Alnylam Pharmaceuticals, Inc. Protected monomers
US8017762B2 (en) * 2003-04-17 2011-09-13 Alnylam Pharmaceuticals, Inc. Modified iRNA agents
US7723509B2 (en) 2003-04-17 2010-05-25 Alnylam Pharmaceuticals IRNA agents with biocleavable tethers
US7851615B2 (en) * 2003-04-17 2010-12-14 Alnylam Pharmaceuticals, Inc. Lipophilic conjugated iRNA agents
AU2013205517B2 (en) * 2003-04-17 2015-07-16 Alnylam Pharmaceuticals, Inc. Modified irna agents
US8796436B2 (en) 2003-04-17 2014-08-05 Alnylam Pharmaceuticals, Inc. Modified iRNA agents
WO2004094606A2 (en) * 2003-04-18 2004-11-04 The Trustees Of The University Of Pennsylvania COMPOSITIONS AND METHODS FOR siRNA INHIBITION OF ANGIOPOIETIN 1 AND 2 AND THEIR RECEPTOR TIE2
ATE516047T1 (en) 2003-05-09 2011-07-15 Diadexus Inc OVR110 ANTIBODY COMPOSITIONS AND METHODS OF USE
AU2003902253A0 (en) 2003-05-12 2003-05-29 The University Of Queensland Method for increasing product yield
EP1628993A4 (en) * 2003-05-16 2010-04-07 Rosetta Inpharmatics Llc Methods and compositions for rna interference
EP1627916B1 (en) 2003-05-28 2009-11-25 Takeda Pharmaceutical Company Limited Anti-BAMBI antibodies or RNA for diagnosis and therapy of colon or liver cancer
JP4505749B2 (en) 2003-05-30 2010-07-21 日本新薬株式会社 Oligo double-stranded RNA that suppresses expression of Bcl-2 and pharmaceutical composition containing the same
ES2864206T3 (en) * 2003-06-02 2021-10-13 Univ Massachusetts Methods and compositions to improve the efficacy and specificity of RNAi
US7750144B2 (en) * 2003-06-02 2010-07-06 University Of Massachusetts Methods and compositions for enhancing the efficacy and specificity of RNA silencing
ES2712695T3 (en) 2003-06-02 2019-05-14 Univ Massachusetts Methods and compositions to control the efficiency of RNA silencing
US20050059044A1 (en) * 2003-06-03 2005-03-17 Graham Michael Wayne Double-stranded nucleic acid
BRPI0410886A (en) 2003-06-03 2006-07-04 Isis Pharmaceuticals Inc double stranded compound, pharmaceutical composition, pharmaceutically acceptable salt, methods of modifying human survivin-encoding nucleic acid, inhibiting suvivin expression in cells or tissues, and treating a condition associated with suvivin expression or overexpression, and single stranded RNA oligonucleotide
AU2004252481B2 (en) 2003-06-06 2011-01-27 Arborgen Inc. Compositions and methods for regulating polysaccharides of a plant cell
US8575327B2 (en) 2003-06-12 2013-11-05 Alnylam Pharmaceuticals, Inc. Conserved HBV and HCV sequences useful for gene silencing
EP2284268A3 (en) * 2003-06-12 2012-05-30 Alnylam Pharmaceuticals Inc. Conserved HBV and HCV sequences useful for gene silencing
CA2530248A1 (en) * 2003-06-25 2005-01-06 Gencia Corporation Modified vectors for organelle transfection
FR2857013B1 (en) * 2003-07-02 2005-09-30 Commissariat Energie Atomique SMALL INTERFERING RNA SPECIFIC OF ALPHA, ALPHA PRIME AND BETA SUBUNITS OF PROTEIN KINASE CK2 AND THEIR APPLICATIONS
AU2004257167B2 (en) 2003-07-03 2012-03-29 The Trustees Of The University Of Pennsylvania Inhibition of Syk kinase expression
ES2559828T3 (en) * 2003-07-16 2016-02-16 Protiva Biotherapeutics Inc. RNA interference encapsulated in lipids
US20050026290A1 (en) * 2003-08-01 2005-02-03 Ciardi Joseph Anthony Inhibiting gene expression with dsRNA
WO2005047505A2 (en) * 2003-08-07 2005-05-26 Whitehead Institute For Biomedical Research Methods and products for expression of micro rnas
US7888497B2 (en) * 2003-08-13 2011-02-15 Rosetta Genomics Ltd. Bioinformatically detectable group of novel regulatory oligonucleotides and uses thereof
US7825235B2 (en) * 2003-08-18 2010-11-02 Isis Pharmaceuticals, Inc. Modulation of diacylglycerol acyltransferase 2 expression
EP1670924A1 (en) * 2003-08-28 2006-06-21 Aveo Pharmaceuticals, Inc. Tumor-specific expression of reporter genes
MXPA06002216A (en) 2003-08-28 2006-04-27 Novartis Ag INTERFERING RNA DUPLEX HAVING BLUNT-ENDS AND 3aCO-MODIFICATIONS.
ES2808561T3 (en) * 2003-09-12 2021-03-01 Univ Massachusetts RNA interference for the treatment of gain-of-function disorders
US8680063B2 (en) 2003-09-12 2014-03-25 University Of Massachusetts RNA interference for the treatment of gain-of-function disorders
CA2539651A1 (en) * 2003-09-22 2005-04-07 Rosetta Inpharmatics Llc Synthetic lethal screen using rna interference
US20050282168A1 (en) * 2003-09-29 2005-12-22 Wyeth Cell surface molecules as markers and therapeutic agents against kidney cancers
AU2004280634A1 (en) * 2003-10-09 2005-04-21 E. I. Du Pont De Nemours And Company Gene silencing by using micro-RNA molecules
US8062891B2 (en) 2003-10-24 2011-11-22 Gencia Corporation Nonviral vectors for delivering polynucleotides to plants
EP2418281B1 (en) 2003-10-24 2016-06-01 Gencia Corporation Methods and compositions for delivering polynucleotides
US8133733B2 (en) 2003-10-24 2012-03-13 Gencia Corporation Nonviral vectors for delivering polynucleotides to target tissues
US20090123468A1 (en) * 2003-10-24 2009-05-14 Gencia Corporation Transducible polypeptides for modifying metabolism
US8507277B2 (en) * 2003-10-24 2013-08-13 Gencia Corporation Nonviral vectors for delivering polynucleotides
CN1926551B (en) * 2003-10-27 2010-06-16 罗斯塔生化科技有限责任公司 Method of designing siRNA for gene silencing
US20070083943A1 (en) * 2003-10-31 2007-04-12 Hannah L C Materials and methods for improved sweet corn
DE10351149A1 (en) * 2003-11-03 2005-06-30 Beiersdorf Ag Oligoribonucleotides for the treatment of unwanted pigmentation of the skin and hair by RNA interference
EP1694842B1 (en) * 2003-11-04 2011-03-23 Geron Corporation Rna amidates and thioamidates for rnai
US8227434B1 (en) 2003-11-04 2012-07-24 H. Lee Moffitt Cancer Center & Research Institute, Inc. Materials and methods for treating oncological disorders
EP2514826A3 (en) * 2003-11-21 2013-04-03 Revivicor Inc. Use of interfering RNA molecules to down regulate the expression of specific genes or family of genes
US20080021205A1 (en) * 2003-12-11 2008-01-24 Helen Blau Methods and Compositions for Use in Preparing Hairpin Rnas
WO2005068630A1 (en) * 2003-12-16 2005-07-28 National Institute Of Advanced Industrial Science And Technology Double-stranded rna for interference
US20060134787A1 (en) 2004-12-22 2006-06-22 University Of Massachusetts Methods and compositions for enhancing the efficacy and specificity of single and double blunt-ended siRNA
WO2005062937A2 (en) * 2003-12-22 2005-07-14 University Of Massachusetts Methods and compositions for enhancing the efficacy and specificity of single and double blunt-ended sirna
AR047574A1 (en) 2003-12-30 2006-01-25 Arborgen Llc 2 Genesis Res 1 CELL CYCLE GENES AND RELATED USE METHODS
AU2005204689A1 (en) * 2004-01-07 2005-07-28 Neopharm, Inc. Lipid compositions and use thereof
EP1706511A4 (en) 2004-01-23 2010-04-07 New England Biolabs Inc Compositions and methods for generating short double-stranded rna using mutated rnase iii
DK1713912T3 (en) * 2004-01-30 2013-12-16 Santaris Pharma As Modified Short Interfering RNA (Modified siRNA)
JP4755113B2 (en) * 2004-01-30 2011-08-24 クアーク・ファーマスーティカルス、インコーポレイテッド Oligoribonucleotides and methods of use thereof for the treatment of fibrotic conditions and other diseases
WO2005076979A2 (en) 2004-02-06 2005-08-25 Wyeth Diagnosis and therapeutics for cancer
EP2295604B1 (en) 2004-02-09 2015-04-08 Thomas Jefferson University Diagnosis and treatment of cancers with microRNA located in or near cancer-associated chromosomal features
WO2005078848A2 (en) 2004-02-11 2005-08-25 University Of Tennessee Research Foundation Inhibition of tumor growth and invasion by anti-matrix metalloproteinase dnazymes
US20080194028A1 (en) * 2004-02-12 2008-08-14 New England Biolabs, Inc. Highly Potent Hsirna Mixtures and Method for Gene Splicing
US20050182005A1 (en) * 2004-02-13 2005-08-18 Tuschl Thomas H. Anti-microRNA oligonucleotide molecules
CA2556435C (en) * 2004-02-13 2014-08-12 The Rockefeller University Anti-microrna oligonucleotide molecules
WO2005079532A2 (en) * 2004-02-17 2005-09-01 University Of Massachusetts Methods and compositions for enhancing risc activity in vitro and in vivo
US20060069050A1 (en) * 2004-02-17 2006-03-30 University Of Massachusetts Methods and compositions for mediating gene silencing
CA2559853A1 (en) 2004-02-17 2005-10-13 University Of South Florida Materials and methods for treatment of inflammatory and cell proliferation disorders
US7622301B2 (en) * 2004-02-24 2009-11-24 Basf Plant Science Gmbh Compositions and methods using RNA interference for control of nematodes
EP1737493B1 (en) 2004-02-25 2011-06-29 Dana-Farber Cancer Institute, Inc. Inhibitors of insulin-like growth factor receptor -1 for inhibiting tumor cell growth
US20070265220A1 (en) 2004-03-15 2007-11-15 City Of Hope Methods and compositions for the specific inhibition of gene expression by double-stranded RNA
EP2514758B2 (en) 2004-03-15 2021-06-23 City of Hope Methods and compositions for the specific inhibition of gene expression by double-stranded RNA
WO2005097207A2 (en) 2004-03-26 2005-10-20 Curis, Inc. Rna interference modulators of hedgehog signaling and uses thereof
US7416842B2 (en) * 2004-04-05 2008-08-26 The Rockefeller University DNA virus microRNA
US8088902B2 (en) 2004-04-05 2012-01-03 The Rockefeller University DNA virus microRNA and methods for inhibiting same
CA2561741C (en) 2004-04-05 2016-09-27 Alnylam Pharmaceuticals, Inc. Processes and reagents for oligonucleotide synthesis and purification
US7365058B2 (en) * 2004-04-13 2008-04-29 The Rockefeller University MicroRNA and methods for inhibiting same
BRPI0510152A (en) 2004-04-23 2007-10-02 Ceres Inc methods for modifying plant characteristics
CA2557532A1 (en) 2004-04-23 2005-11-10 Angela M. Christiano Inhibition of hairless protein mrna
JP4584986B2 (en) * 2004-04-27 2010-11-24 アルニラム ファーマスーティカルズ インコーポレイテッド Single-stranded and double-stranded oligonucleotides containing 2-arylpropyl moieties
AU2005323437B2 (en) * 2004-04-30 2011-10-06 Alnylam Pharmaceuticals, Inc. Oligonucleotides comprising a C5-modified pyrimidine
US7605250B2 (en) * 2004-05-12 2009-10-20 Dharmacon, Inc. siRNA targeting cAMP-specific phosphodiesterase 4D
US7687616B1 (en) 2004-05-14 2010-03-30 Rosetta Genomics Ltd Small molecules modulating activity of micro RNA oligonucleotides and micro RNA targets and uses thereof
JP5697297B2 (en) 2004-05-14 2015-04-08 ロゼッタ ジノミクス リミテッド Micro NAS and its use
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
US7795419B2 (en) 2004-05-26 2010-09-14 Rosetta Genomics Ltd. Viral and viral associated miRNAs and uses thereof
EP2290071B1 (en) * 2004-05-28 2014-12-31 Asuragen, Inc. Methods and compositions involving microRNA
US8394947B2 (en) 2004-06-03 2013-03-12 Isis Pharmaceuticals, Inc. Positionally modified siRNA constructs
US20140371299A1 (en) * 2004-06-07 2014-12-18 Senesco Technologies, Inc. Use of Apoptosis-Specific elF-5A siRNA to Down Regulate Expression of Proinflammatory Cytokines to Treat Sepsis
US7615618B2 (en) * 2004-06-30 2009-11-10 Alnylam Pharmaceuticals, Inc. Oligonucleotides comprising a non-phosphate backbone linkage
US7297786B2 (en) * 2004-07-09 2007-11-20 University Of Iowa Research Foundation RNA interference in respiratory epitheial cells
US8361976B2 (en) 2004-07-09 2013-01-29 University Of Massachusetts Therapeutic alteration of transplantable tissues through in situ or ex vivo exposure to RNA interference molecules
US7968762B2 (en) 2004-07-13 2011-06-28 Van Andel Research Institute Immune-compromised transgenic mice expressing human hepatocyte growth factor (hHGF)
AU2005275062A1 (en) 2004-07-14 2006-02-23 University Of Utah Research Foundation Netrin-related compositions and uses
CA2574572A1 (en) 2004-07-19 2006-10-26 Baylor College Of Medicine Modulation of cytokine signaling regulators and applications for immunotherapy
US7579451B2 (en) 2004-07-21 2009-08-25 Alnylam Pharmaceuticals, Inc. Oligonucleotides comprising a modified or non-natural nucleobase
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
WO2006091233A2 (en) * 2004-07-23 2006-08-31 Boston Medical Center Corporation Cellular delivery of reagents that inhibit gene expression utilizing the anthrax toxin protective antigen (pa)
WO2006014899A2 (en) 2004-07-26 2006-02-09 Dow Global Technologies Inc. Process for improved protein expression by strain engineering
CA2576193A1 (en) 2004-08-03 2006-02-16 Biogen Idec Ma Inc. Taj in neuronal function
AU2005330637B2 (en) 2004-08-04 2012-09-20 Alnylam Pharmaceuticals, Inc. Oligonucleotides comprising a ligand tethered to a modified or non-natural nucleobase
WO2006020768A2 (en) 2004-08-10 2006-02-23 Alnylam Pharmaceuticals, Inc. Chemically modified oligonucleotides
EP1778848A2 (en) * 2004-08-13 2007-05-02 BASF Plant Science GmbH Compositions and methods using rna interference for control of nematodes
EP1789592A4 (en) * 2004-08-13 2009-12-23 Univ Delaware Method for identification and quantification of short or small rna molecules
EA012799B1 (en) 2004-08-16 2009-12-30 Кварк Фармасьютикалс, Инк. Therapeutic uses of inhibitors of rtp801
US7893197B2 (en) 2004-08-25 2011-02-22 Janssen Pharmaceutica N.V. Relaxin-3 chimeric polypeptides and their preparation and use
US7323310B2 (en) * 2004-08-31 2008-01-29 Qiagen North American Holdings, Inc. Methods and compositions for RNA amplification and detection using an RNA-dependent RNA-polymerase
US7884086B2 (en) 2004-09-08 2011-02-08 Isis Pharmaceuticals, Inc. Conjugates for use in hepatocyte free uptake assays
US20060059585A1 (en) 2004-09-14 2006-03-16 Boris Jankowski Modulating plant sugar levels
WO2006031859A2 (en) 2004-09-14 2006-03-23 Ceres Inc. Modulation of amino acid and sugar content in plants
US20060057590A1 (en) * 2004-09-14 2006-03-16 Azeddine Si-Ammour RNA probes
FI20041204A0 (en) 2004-09-16 2004-09-16 Riikka Lund Methods for the utilization of new target genes associated with immune-mediated diseases
US7799906B1 (en) 2004-09-22 2010-09-21 Arborgen, Llc Compositions and methods for modulating lignin of a plant
SG155257A1 (en) 2004-09-24 2009-09-30 Beth Israel Hospital Methods of diagnosing and treating complications of pregnancy
NZ553987A (en) 2004-09-28 2011-01-28 Quark Pharmaceuticals Inc Oligoribonucleotides and methods of use thereof for treatment of alopecia, acute renal failure and other diseases
JP2008522585A (en) * 2004-10-12 2008-07-03 ザ ロックフェラー ユニバーシティー MicroRNA
JP2008517629A (en) * 2004-10-27 2008-05-29 シェーリング コーポレイション Compositions and methods for short interfering nucleic acid inhibition of Nav1.8
US9492400B2 (en) 2004-11-04 2016-11-15 Massachusetts Institute Of Technology Coated controlled release polymer particles as efficient oral delivery vehicles for biopharmaceuticals
EP2281888B1 (en) 2004-11-12 2015-01-07 Asuragen, Inc. Methods and compositions involving miRNA and miRNA inhibitor molecules
WO2006060454A2 (en) * 2004-12-02 2006-06-08 B-Bridge International, Inc. Methods of designing small interfering rnas, antisense polynucleotides, and other hybridizing polynucleotides
US7517870B2 (en) 2004-12-03 2009-04-14 Fondazione Telethon Use of compounds that interfere with the hedgehog signaling pathway for the manufacture of a medicament for preventing, inhibiting, and/or reversing ocular diseases related with ocular neovascularization
WO2006062971A2 (en) 2004-12-08 2006-06-15 Ceres Inc. Modulating plant carbon levels
WO2007089610A1 (en) 2006-01-26 2007-08-09 Ceres, Inc. Modulating plant oil levels
EP1833838A2 (en) * 2004-12-14 2007-09-19 Applera Corporation, Applied Biosystems Group Cationic liposomes comprising a charge neutral compound and a cationic phospholipid
EP1828388A2 (en) 2004-12-16 2007-09-05 Ceres, Inc. Modulating plant nitrogen levels
US7335760B2 (en) 2004-12-22 2008-02-26 Ceres, Inc. Nucleic acid sequences encoding zinc finger proteins
US20060142228A1 (en) * 2004-12-23 2006-06-29 Ambion, Inc. Methods and compositions concerning siRNA's as mediators of RNA interference
TWI401316B (en) * 2004-12-23 2013-07-11 Alcon Inc Rnai inhibition of serum amyloid a for treatment of glaucoma
TWI386225B (en) * 2004-12-23 2013-02-21 Alcon Inc Rnai inhibition of ctgf for treatment of ocular disorders
CA2593032C (en) 2004-12-27 2015-12-22 Silence Therapeutics Ag Coated lipid complexes and their use
US20070014795A1 (en) * 2004-12-30 2007-01-18 Dhodapkar Madhav V Compositions and methods for enhanced dendritic cell maturation and function
CN101124339A (en) * 2004-12-30 2008-02-13 托德·M·豪泽 Compositions and methods for modulating gene expression using self-protected oligonucleotides
US8137907B2 (en) * 2005-01-03 2012-03-20 Cold Spring Harbor Laboratory Orthotopic and genetically tractable non-human animal model for liver cancer and the uses thereof
DK1841793T3 (en) 2005-01-07 2010-07-19 Diadexus Inc Ovr110 antibody compositions and methods for their use
US7718625B2 (en) 2005-01-27 2010-05-18 University Of South Florida Polynucleotides targeted against the extended 5′-UTR region of argininosuccinate synthase and uses thereof
TW200639252A (en) * 2005-02-01 2006-11-16 Alcon Inc RNAi-mediated inhibition of ocular hypertension targets
US7199128B2 (en) * 2005-02-02 2007-04-03 Achillion Pharmaceuticals, Inc. 8-N-substituted-2H-isothiazolo[5,4-b]quinolizine-3,4-diones and related compounds as antiinfective agents
US7745389B2 (en) 2005-02-14 2010-06-29 University Of Iowa Research Foundation Methods for treatment of age-related macular degeneration
CN101175769A (en) 2005-03-10 2008-05-07 健泰科生物技术公司 Methods and compositions for modulating vascular integrity
CA2598234A1 (en) * 2005-03-11 2006-09-21 Alcon, Inc. Rnai-mediated inhibition of frizzled related protein-1 for treatment of glaucoma
DE202005004135U1 (en) * 2005-03-11 2005-05-19 Klocke Verpackungs-Service Gmbh Multi-component packaging with applicator
WO2006130201A1 (en) 2005-03-14 2006-12-07 Board Of Regents, The University Of Texas System Antigene oligomers inhibit transcription
GB0505081D0 (en) * 2005-03-14 2005-04-20 Genomica Sau Downregulation of interleukin-12 expression by means of rnai technology
SI2567976T1 (en) 2005-03-23 2017-11-30 Genmab A/S Antibodies against CD38 for treatment of multiple myeloma
US7985852B2 (en) 2005-04-15 2011-07-26 National University Corporation Tottori University hTERT gene expression regulatory gene
US20090203055A1 (en) * 2005-04-18 2009-08-13 Massachusetts Institute Of Technology Compositions and methods for RNA interference with sialidase expression and uses thereof
JP5173793B2 (en) 2005-04-29 2013-04-03 ザ ロックフェラー ユニバーシティ MicroRNA and method for inhibiting the same
US20090042186A1 (en) * 2005-05-06 2009-02-12 Alexander Mankin Mapping new sites for antibiotic action in the ribosome
KR100694804B1 (en) 2005-05-18 2007-03-14 아주대학교산학협력단 A composition for the prophylactic or treatment of endometrial cancer and a method for preventing or treating endometrial cancer using the composition
EP1896587A2 (en) 2005-05-31 2008-03-12 Cold Spring Harbor Laboratory METHODS FOR PRODUCING MICRORNAs
JP5371424B2 (en) 2005-06-01 2013-12-18 ポリプラス トランスフェクション エスアー Oligonucleotides for RNA interference and their biological applications
WO2006135862A2 (en) 2005-06-10 2006-12-21 Children's Hospital And Research Center At Oakland Immunomodulation by altering sphingosine 1-phosphate lyase (spl) activity
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
WO2006138606A2 (en) 2005-06-17 2006-12-28 Arborgen, Llc Cell signaling genes and related methods
US7868159B2 (en) 2005-06-23 2011-01-11 Baylor College Of Medicine Modulation of negative immune regulators and applications for immunotherapy
EP1915448B1 (en) 2005-07-07 2013-09-04 Yissum Research Development Company, of The Hebrew University of Jerusalem Nucleic acid agents for downregulating h19, and methods of using same
EP1907855A4 (en) * 2005-07-12 2009-11-11 Univ Temple Genetic and epigenetic alterations in the diagnosis and treatment of cancer
US8703769B2 (en) 2005-07-15 2014-04-22 The University Of North Carolina At Chapel Hill Use of EGFR inhibitors to prevent or treat obesity
WO2007014370A2 (en) * 2005-07-28 2007-02-01 University Of Delaware Small regulatory rnas and methods of use
US20090176725A1 (en) * 2005-08-17 2009-07-09 Sirna Therapeutics Inc. Chemically modified short interfering nucleic acid molecules that mediate rna interference
EP1937066A4 (en) * 2005-08-18 2008-12-24 Alnylam Pharmaceuticals Inc Methods and compositions for treating neurological disease
US20100129491A1 (en) 2005-09-01 2010-05-27 Yoshihiro Nakao Tryptophan transporter gene and use thereof
ES2523989T3 (en) 2005-09-12 2014-12-03 The Ohio State University Research Foundation Compositions for the therapy of cancers associated with BCL2
EP1762575A1 (en) 2005-09-12 2007-03-14 Ganymed Pharmaceuticals AG Identification of tumor-associated antigens for diagnosis and therapy
CN101268194A (en) 2005-09-20 2008-09-17 巴斯福植物科学有限公司 Methods for controlling gene expression using ta-siRNA
US8168584B2 (en) 2005-10-08 2012-05-01 Potentia Pharmaceuticals, Inc. Methods of treating age-related macular degeneration by compstatin and analogs thereof
US7723314B1 (en) * 2005-10-28 2010-05-25 Transderm, Inc. Methods and compositions for treating pachyonychia congenita
EP1946117B1 (en) * 2005-11-11 2011-08-24 Roger Williams Hospital P66-shc as predictive marker in cancer treatment
EP2431467A3 (en) 2005-11-17 2012-05-02 Board Of Regents, The University Of Texas Modulation of gene expression by oligomers targeted to chromosomal DNA
EP1960778A2 (en) * 2005-11-25 2008-08-27 Institut National De La Sante Et De La Recherche Medicale (Inserm) Method for demonstrating presence or absence of markers associated with the presence and/or the chemosensitivity of tumors
CN101535500B (en) 2005-11-29 2013-12-11 剑桥企业有限公司 Markers for breast cancer
JP4737531B2 (en) 2005-12-05 2011-08-03 サントリーホールディングス株式会社 Method for producing ceramide using transformed yeast
EP1795596A1 (en) 2005-12-08 2007-06-13 Ganymed Pharmaceuticals AG Composition and methods for therapy and diagnosis of cancer
US20090317802A1 (en) * 2005-12-09 2009-12-24 Bhatia Sangeeta N Compositions and Methods to Monitor RNA Delivery to Cells
US9157066B2 (en) 2005-12-13 2015-10-13 The Trustees Of The University Of Pennsylvania Transcriptome transfer produces cellular phenotype conversion
US10646590B2 (en) 2005-12-13 2020-05-12 The Trustees Of The University Of Pennsylvania Methods for phototransfecting nucleic acids into live cells
US10647960B2 (en) 2005-12-13 2020-05-12 The Trustees Of The University Of Pennsylvania Transcriptome transfer produces cellular phenotype conversion
US9267937B2 (en) 2005-12-15 2016-02-23 Massachusetts Institute Of Technology System for screening particles
EP1966379B1 (en) * 2005-12-22 2010-04-28 OPKO Ophthalmics, LLC Compositions and methods for regulating complement system
DE602006020617D1 (en) 2005-12-30 2011-04-21 Evonik Roehm Gmbh LACTOFERRIN PEPTIDES, SUITABLE AS PEPTIDES INTO THE CELL
ES2461189T3 (en) 2006-01-05 2014-05-19 The Ohio State University Research Foundation MicroRNA-based methods and compositions for the diagnosis and treatment of solid breast or lung cancers
EP1968622B1 (en) 2006-01-05 2014-08-27 The Ohio State University Research Foundation Microrna expression abnormalities in pancreatic endocrine and acinar tumors
CN102943108B (en) 2006-01-05 2014-05-21 俄亥俄州立大学研究基金会 Microrna-based methods and compositions for the diagnosis, prognosis and treatment of lung cancer
EP1970078A4 (en) 2006-01-11 2010-11-17 Kyowa Hakko Kirin Co Ltd Composition inhibiting the expression of target gene in eyeball and remedy for disease in eyeball
US20090060921A1 (en) * 2006-01-17 2009-03-05 Biolex Therapeutics, Inc. Glycan-optimized anti-cd20 antibodies
BRPI0707290A2 (en) * 2006-01-17 2011-08-16 Biolex Therapeutics Inc compositions and methods for humanization and optimization of n-glycans in plants
NL2000439C2 (en) 2006-01-20 2009-03-16 Quark Biotech Therapeutic applications of inhibitors of RTP801.
US20120208824A1 (en) 2006-01-20 2012-08-16 Cell Signaling Technology, Inc. ROS Kinase in Lung Cancer
HUE039085T2 (en) 2006-01-20 2018-12-28 Cell Signaling Technology Inc Translocation and mutant ROS kinase in human non-small cell lung carcinoma
US7825099B2 (en) 2006-01-20 2010-11-02 Quark Pharmaceuticals, Inc. Treatment or prevention of oto-pathologies by inhibition of pro-apoptotic genes
DK1984382T3 (en) * 2006-01-27 2012-09-03 Santaris Pharma As LNA modified phosphorothiolated oligonucleotides
CA2913655A1 (en) 2006-01-27 2007-08-09 Biogen Ma Inc. Nogo receptor antagonists
US7884078B2 (en) 2006-02-10 2011-02-08 Massachusetts Institute Of Technology CPG15 compounds as insulin receptor and insulin-like growth factor receptor agonists
US20090047381A1 (en) 2006-02-24 2009-02-19 Suntory Limited Ammonia transporter gene and use thereof
WO2007097113A1 (en) 2006-02-24 2007-08-30 Suntory Limited Gene encoding protein responsible for flocculation property of yeast and use thereof
KR100929997B1 (en) 2006-02-24 2009-12-07 산토리 홀딩스 가부시키가이샤 Genes encoding proteins responsible for the aggregation properties of yeast and uses thereof
EP2522750A1 (en) 2006-03-02 2012-11-14 The Ohio State University MicroRNA expression profile associated with pancreatic cancer
US7910566B2 (en) 2006-03-09 2011-03-22 Quark Pharmaceuticals Inc. Prevention and treatment of acute renal failure and other kidney diseases by inhibition of p53 by siRNA
FI20060246A0 (en) 2006-03-16 2006-03-16 Jukka Westermarck A new growth stimulating protein and its use
EP2369012A1 (en) 2006-03-20 2011-09-28 The Ohio State University Research Foundation Micro-RNA fingerprints during human megakaryocytopoiesis
JP2009529917A (en) 2006-03-22 2009-08-27 ヴァイラル ロジック システムズ テクノロジー コーポレーション Methods for identifying target polypeptides and their use for the treatment of immunological disorders
CA2644347C (en) * 2006-03-23 2017-05-30 Santaris Pharma A/S Small internally segmented interfering rna
FR2898908A1 (en) 2006-03-24 2007-09-28 Agronomique Inst Nat Rech Process, useful to prepare differentiated avian cells from avian stem cells grown in culture medium, comprises induction of stem cells differentiation by inhibiting expression/activity of gene expressed in the stem cells e.g. Nanog gene
WO2008105773A2 (en) 2006-03-31 2008-09-04 Massachusetts Institute Of Technology System for targeted delivery of therapeutic agents
EP2010226B1 (en) 2006-04-07 2014-01-15 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
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
EP2018873A4 (en) 2006-04-07 2009-12-09 Japan Found Cancer Prophylactic/therapeutic agent for cancer
EP2012815A4 (en) 2006-04-14 2009-12-09 Massachusetts Inst Technology Identifying and modulating molecular pathways that mediate nervous system plasticity
EP2450437B1 (en) 2006-04-14 2017-05-17 Cell Signaling Technology, Inc. Gene defects and mutant ALK kinase in human solid tumors
WO2007127487A2 (en) 2006-04-28 2007-11-08 University Of South Florida Materials and methods for reducing inflammation by inhibition of the atrial natriuretic peptide receptor
GB0608838D0 (en) 2006-05-04 2006-06-14 Novartis Ag Organic compounds
JPWO2007132867A1 (en) 2006-05-15 2009-09-24 杉本 芳一 Preventive and therapeutic agents for cancer
JP5630998B2 (en) 2006-05-15 2014-11-26 マサチューセッツ インスティテュート オブ テクノロジー Polymers for functional particles
AU2007257162A1 (en) 2006-06-05 2007-12-13 Cancer Care Ontario Assessment of risk for colorectal cancer
WO2007141796A2 (en) 2006-06-09 2007-12-13 Quark Pharmaceuticals, Inc. Therapeutic uses of inhibitors of rtp801l
US20080152654A1 (en) * 2006-06-12 2008-06-26 Exegenics, Inc., D/B/A Opko Health, Inc. COMPOSITIONS AND METHODS FOR siRNA INHIBITION OF ANGIOGENESIS
US9381477B2 (en) 2006-06-23 2016-07-05 Massachusetts Institute Of Technology Microfluidic synthesis of organic nanoparticles
ES2425387T3 (en) 2006-07-13 2013-10-15 The Ohio State University Research Foundation Mir-106a to diagnose colon adenocarcinoma of poor survival prognosis
SG173371A1 (en) 2006-07-13 2011-08-29 Univ Iowa Res Found Methods and reagents for treatment and diagnosis of vascular disorders and age-related macular degeneration
CN101490253A (en) 2006-07-21 2009-07-22 赛伦斯治疗公司 Means for inhibiting the expression of protein kinase 3
WO2008014979A2 (en) 2006-07-31 2008-02-07 Curevac Gmbh NUCLEIC ACID OF FORMULA (I): GIXmGn, OR (II): CIXmCn, IN PARTICULAR AS AN IMMUNE-STIMULATING AGENT/ADJUVANT
WO2008019142A2 (en) * 2006-08-04 2008-02-14 Massachusetts Institute Of Technology Oligonucleotide systems for targeted intracellular delivery
US7872118B2 (en) * 2006-09-08 2011-01-18 Opko Ophthalmics, Llc siRNA and methods of manufacture
US20100158894A1 (en) 2006-09-15 2010-06-24 Tokai University Preventive or remedy for er-negative and her2-negative breast cancer and method of screening the same
EP2145001A2 (en) * 2006-09-19 2010-01-20 Asuragen, Inc. Mir-15, mir-26, mir -31,mir -145, mir-147, mir-188, mir-215, mir-216 mir-331, mmu-mir-292-3p regulated genes and pathways as targets for therapeutic intervention
AU2007299804A1 (en) * 2006-09-19 2008-03-27 Asuragen, Inc. MiR-200 regulated genes and pathways as targets for therapeutic intervention
EP1911851A1 (en) 2006-10-12 2008-04-16 Ganymed Pharmaceuticals AG Compositions and methods for therapy and diagnosis of cancer and cancer metastasis
JP2010507387A (en) 2006-10-25 2010-03-11 クアーク・ファーマスーティカルス、インコーポレイテッド Novel siRNA and method of using the same
WO2008052774A2 (en) 2006-10-31 2008-05-08 Noxxon Pharma Ag Methods for detection of a single- or double-stranded nucleic acid molecule
WO2008053487A2 (en) 2006-11-01 2008-05-08 The Medical Research Fund At The Tel-Aviv Sourasky Medical Center Adipocyte-specific constructs and methods for inhibiting platelet-type 12 lipoxygenase expression
US20110294782A1 (en) 2006-11-10 2011-12-01 Massachusetts Institute Of Technology Small molecule pak inhibitors
JP5391073B2 (en) 2006-11-27 2014-01-15 ディアデクサス インコーポレーテッド Ovr110 antibody compositions and methods of use
EP2101813B1 (en) 2006-11-27 2014-04-02 Patrys Limited Novel glycosylated peptide target in neoplastic cells
AU2007333225B2 (en) * 2006-12-08 2014-06-12 Massachusetts Institute Of Technology Delivery of nanoparticles and/or agents to cells
CN101675165A (en) * 2006-12-08 2010-03-17 奥斯瑞根公司 The function of LET-7 Microrna and target
CA2671294A1 (en) * 2006-12-08 2008-06-19 Asuragen, Inc. Mir-21 regulated genes and pathways as targets for therapeutic intervention
US20090175827A1 (en) * 2006-12-29 2009-07-09 Byrom Mike W miR-16 REGULATED GENES AND PATHWAYS AS TARGETS FOR THERAPEUTIC INTERVENTION
US8476243B2 (en) 2006-12-29 2013-07-02 Transderm, Inc. Methods and compositions for treating keratin hyperproliferative disorders
US8975068B2 (en) 2007-01-25 2015-03-10 The General Hospital Corporation Isolated stem cell comprising a Xic flanking region transgene
EP2117305A4 (en) 2007-01-26 2011-03-30 Univ Louisville Res Found Modification of exosomal components for use as a vaccine
US8530436B2 (en) 2007-01-29 2013-09-10 Transderm, Inc. Methods and compositions for transdermal delivery of nucleotides
US9217129B2 (en) 2007-02-09 2015-12-22 Massachusetts Institute Of Technology Oscillating cell culture bioreactor
US7872119B2 (en) 2007-02-26 2011-01-18 Quark Pharmaceuticals, Inc. Inhibitors of RTP801 and their use in disease treatment
US20100292301A1 (en) * 2007-02-28 2010-11-18 Elena Feinstein Novel sirna structures
EP2243834A1 (en) 2007-03-05 2010-10-27 Cancer Care Ontario Assessment of risk for colorectal cancer
CA2680600A1 (en) 2007-03-12 2008-09-18 Antigen Express, Inc. Li-rnai involved li suppression in cancer immunotherapy
WO2008115387A2 (en) * 2007-03-15 2008-09-25 University Hospitals Of Cleveland Screening, diagnosing, treating and prognosis of pathophysiologic states by rna regulation
WO2008115556A2 (en) * 2007-03-19 2008-09-25 Cold Spring Harbor Laboratory Identification of genetic alterations that modulate drug sensitivity in cancer treatments
US7812002B2 (en) 2007-03-21 2010-10-12 Quark Pharmaceuticals, Inc. Oligoribonucleotide inhibitors of NRF2 and methods of use thereof for treatment of cancer
EP2136832B1 (en) 2007-03-26 2015-09-02 General Regeneratives Limited Methods for promoting protection and regeneration of bone marrow using cxcl9 and anti-cxcl9 antibodies
JP5344517B2 (en) 2007-03-30 2013-11-20 サントリーホールディングス株式会社 Method for producing ceramide in transformed cells using sphingolipid Δ4-desaturase with endoplasmic reticulum localization signal
EP2159281B1 (en) 2007-03-30 2012-09-26 National University Corporation Okayama University Novel slc17-type transporter protein in mammal and use thereof
WO2008124634A1 (en) 2007-04-04 2008-10-16 Massachusetts Institute Of Technology Polymer-encapsulated reverse micelles
WO2008124639A2 (en) 2007-04-04 2008-10-16 Massachusetts Institute Of Technology Poly (amino acid) targeting moieties
WO2008124129A2 (en) * 2007-04-09 2008-10-16 University Of Massachusetts Treating hiv with a m-csf effector kinase inhibitor like imatinib
JP5889527B2 (en) 2007-04-12 2016-03-22 ザ ブリガム アンド ウィメンズ ホスピタル インコーポレイテッドThe Brigham and Women’s Hospital, Inc. ABCB5 targeting for cancer therapy
AU2008245696B2 (en) 2007-04-27 2013-11-07 Pelican Technology Holdings, Inc. Method for rapidly screening microbial hosts to identify certain strains with improved yield and/or quality in the expression of heterologous proteins
US9580719B2 (en) 2007-04-27 2017-02-28 Pfenex, Inc. Method for rapidly screening microbial hosts to identify certain strains with improved yield and/or quality in the expression of heterologous proteins
US11078262B2 (en) 2007-04-30 2021-08-03 Allergan, Inc. High viscosity macromolecular compositions for treating ocular conditions
WO2008143774A2 (en) * 2007-05-01 2008-11-27 University Of Massachusetts Methods and compositions for locating snp heterozygosity for allele specific diagnosis and therapy
WO2008143786A1 (en) * 2007-05-14 2008-11-27 The Rockefeller University Production of artificial micrornas using synthetic microrna precursors
JP5726520B2 (en) 2007-05-22 2015-06-03 アークトゥラス・セラピューティクス・インコーポレイテッドArcturus Therapeutics,Inc. UNA oligomers for therapeutic agents
US20090131354A1 (en) * 2007-05-22 2009-05-21 Bader Andreas G miR-126 REGULATED GENES AND PATHWAYS AS TARGETS FOR THERAPEUTIC INTERVENTION
CA2687964A1 (en) 2007-06-01 2009-02-19 The Trustees Of Princeton University Treatment of viral infections by modulation of host cell metabolic pathways
WO2008154470A1 (en) * 2007-06-08 2008-12-18 University Of Connecticut Nhibitor of the receptor activity of the s1p2 receptor for inhibiting pathological angiogenesis in the eye
EP2170403B1 (en) 2007-06-27 2014-04-16 Quark Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of pro-apoptotic genes
JP5298014B2 (en) 2007-07-03 2013-09-25 杏林製薬株式会社 Influenza treatment
NZ582390A (en) 2007-07-10 2012-05-25 Neurim Pharma 1991 Cd44 splice variants in neurodegenerative diseases
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
CN101809169B (en) 2007-07-31 2013-07-17 俄亥俄州立大学研究基金会 Methods for reverting methylation by targeting DNMT3A and DNMT3B
US8012474B2 (en) 2007-08-02 2011-09-06 Nov Immune S.A. Anti-RANTES antibodies
ES2627059T3 (en) 2007-08-03 2017-07-26 The Ohio State University Research Foundation Ultraconserved regions encoding RNAnc
US20100186102A1 (en) * 2007-08-21 2010-07-22 Scott And White Memorial Hospital And Scott, Sherwood, And Brindley Foundation Methods and compositions for post-transcriptional gene silencing
JP5770472B2 (en) 2007-08-22 2015-08-26 ジ・オハイオ・ステイト・ユニバーシティ・リサーチ・ファウンデイションThe Ohio State University Research Foundation Methods and compositions for inducing deregulation of EPHA7 and ERK phosphorylation in human acute leukemia
WO2009030254A1 (en) 2007-09-04 2009-03-12 Curevac Gmbh Complexes of rna and cationic peptides for transfection and for immunostimulation
US8183221B2 (en) 2007-09-05 2012-05-22 Medtronic, Inc. Suppression of SCN9A gene expression and/or function for the treatment of pain
CN101939446B (en) 2007-09-06 2015-02-11 俄亥俄州立大学研究基金会 MicroRNA signatures in human ovarian cancer
WO2009036332A1 (en) 2007-09-14 2009-03-19 Asuragen, Inc. Micrornas differentially expressed in cervical cancer and uses thereof
CN101849008A (en) 2007-09-19 2010-09-29 应用生物系统有限公司 SiRNA sequence-independent modification formats for reducing off-target phenotypic effects in RNAi, and stabilized forms thereof
CA2701845A1 (en) 2007-10-03 2009-04-09 Quark Pharmaceuticals, Inc. Novel sirna structures
AU2008310704B2 (en) 2007-10-11 2014-03-20 The Government Of The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Methods and compositions for the diagnosis and treatment of esphageal adenocarcinomas
MX2010003642A (en) 2007-10-12 2010-08-09 Massachusetts Inst Technology Vaccine nanotechnology.
EP3741851A1 (en) 2007-10-18 2020-11-25 Cell Signaling Technology, Inc. Translocation and mutant ros kinase in human non-small cell lung carcinoma
EP2060583A1 (en) 2007-10-23 2009-05-20 Ganymed Pharmaceuticals AG Identification of tumor-associated markers for diagnosis and therapy
US8097712B2 (en) 2007-11-07 2012-01-17 Beelogics Inc. Compositions for conferring tolerance to viral disease in social insects, and the use thereof
US7828840B2 (en) * 2007-11-15 2010-11-09 Med Institute, Inc. Medical devices and methods for local delivery of angiotensin II type 2 receptor antagonists
CN101932339B (en) 2007-11-30 2014-10-29 贝勒医学院 Dendritic cell vaccine compositions and uses of same
US8071562B2 (en) * 2007-12-01 2011-12-06 Mirna Therapeutics, Inc. MiR-124 regulated genes and pathways as targets for therapeutic intervention
US8507455B2 (en) * 2007-12-04 2013-08-13 Alnylam Pharmaceuticals, Inc. Folate conjugates
EP2231195B1 (en) * 2007-12-04 2017-03-29 Arbutus Biopharma Corporation Targeting lipids
AU2008336249B2 (en) 2007-12-10 2015-01-29 The University Of Queensland Treatment and prophylaxis
WO2009074990A2 (en) * 2007-12-12 2009-06-18 Quark Pharmaceuticals, Inc. Rtp801l sirna compounds and methods of use thereof
US8614311B2 (en) 2007-12-12 2013-12-24 Quark Pharmaceuticals, Inc. RTP801L siRNA compounds and methods of use thereof
WO2009086156A2 (en) * 2007-12-21 2009-07-09 Asuragen, Inc. Mir-10 regulated genes and pathways as targets for therapeutic intervention
EP2242854A4 (en) * 2008-01-15 2012-08-15 Quark Pharmaceuticals Inc Sirna compounds and methods of use thereof
EP3346005A1 (en) 2008-01-31 2018-07-11 CureVac AG Nucleic acids of formula (i) (nuglxmgnnv)a and derivatives thereof as an immunostimulating agent/adjuvant
US20090263803A1 (en) * 2008-02-08 2009-10-22 Sylvie Beaudenon Mirnas differentially expressed in lymph nodes from cancer patients
CN104975020B (en) 2008-02-11 2020-01-17 菲奥医药公司 Modified RNAi polynucleotides and uses thereof
WO2009111643A2 (en) * 2008-03-06 2009-09-11 Asuragen, Inc. Microrna markers for recurrence of colorectal cancer
JP2011517404A (en) * 2008-03-20 2011-06-09 クォーク・ファーマシューティカルズ・インク Novel siRNA compound for inhibiting RTP801
US20090253780A1 (en) * 2008-03-26 2009-10-08 Fumitaka Takeshita COMPOSITIONS AND METHODS RELATED TO miR-16 AND THERAPY OF PROSTATE CANCER
US20090258928A1 (en) * 2008-04-08 2009-10-15 Asuragen, Inc. Methods and compositions for diagnosing and modulating human papillomavirus (hpv)
EP2108701A1 (en) 2008-04-10 2009-10-14 Ganymed Pharmaceuticals AG Methods involving MS4A12 and agents targeting MS4A12 for therapy, diagnosis and testing
EP3604533A1 (en) 2008-04-11 2020-02-05 Arbutus Biopharma Corporation Site-specific delivery of nucleic acids by combining targeting ligands with endosomolytic components
EP2285385A4 (en) * 2008-04-15 2013-01-16 Quark Pharmaceuticals Inc siRNA COMPOUNDS FOR INHIBITING NRF2
WO2009127060A1 (en) 2008-04-15 2009-10-22 Protiva Biotherapeutics, Inc. Novel lipid formulations for nucleic acid delivery
EP2116602A1 (en) 2008-05-07 2009-11-11 Institut Gustave Roussy Combination products for treating cancer
US8258111B2 (en) * 2008-05-08 2012-09-04 The Johns Hopkins University Compositions and methods related to miRNA modulation of neovascularization or angiogenesis
AU2009256243A1 (en) 2008-06-04 2009-12-10 The Board Of Regents Of The University Of Texas System Modulation of gene expression through endogenous small RNA targeting of gene promoters
WO2009147684A2 (en) 2008-06-06 2009-12-10 Quark Pharmaceuticals, Inc. Compositions and methods for treatment of ear disorders
TWI455944B (en) 2008-07-01 2014-10-11 Daiichi Sankyo Co Ltd Double-stranded polynucleotides
US8815818B2 (en) 2008-07-18 2014-08-26 Rxi Pharmaceuticals Corporation Phagocytic cell delivery of RNAI
CN105030809A (en) 2008-08-01 2015-11-11 协和发酵麒麟株式会社 Composition for inhibiting expression of target gene
EP2323695B1 (en) 2008-08-19 2018-12-05 Nektar Therapeutics Complexes of small-interfering nucleic acids
CN103429270B (en) * 2008-08-25 2016-11-23 埃克斯雷德制药有限公司 Stop antisense nucleotide of Connective Tissue Growth Factor and application thereof
WO2011028218A1 (en) 2009-09-02 2011-03-10 Alnylam Pharmaceuticals, Inc. Process for triphosphate oligonucleotide synthesis
US20100068200A1 (en) * 2008-09-12 2010-03-18 The University Of Connecticut Methods and Compositions for Inhibiting Atherosclerosis and Vascular Inflammation
CA2746527A1 (en) 2008-09-22 2010-03-25 Rxi Pharmaceuticals Corporation Rna interference in skin indications
CA2737661C (en) 2008-09-23 2019-08-20 Alnylam Pharmaceuticals, Inc. Chemical modifications of monomers and oligonucleotides with cycloaddition
WO2010037408A1 (en) 2008-09-30 2010-04-08 Curevac Gmbh Composition comprising a complexed (m)rna and a naked mrna for providing or enhancing an immunostimulatory response in a mammal and uses thereof
US8343498B2 (en) 2008-10-12 2013-01-01 Massachusetts Institute Of Technology Adjuvant incorporation in immunonanotherapeutics
US8343497B2 (en) 2008-10-12 2013-01-01 The Brigham And Women's Hospital, Inc. Targeting of antigen presenting cells with immunonanotherapeutics
US8277812B2 (en) 2008-10-12 2012-10-02 Massachusetts Institute Of Technology Immunonanotherapeutics that provide IgG humoral response without T-cell antigen
US8591905B2 (en) 2008-10-12 2013-11-26 The Brigham And Women's Hospital, Inc. Nicotine immunonanotherapeutics
US8691514B2 (en) 2008-10-31 2014-04-08 Japan Science And Technology Agency Method for selective control of helper T cell function
EP2727996A1 (en) 2008-11-06 2014-05-07 The Johns-Hopkins University Treatment of chronic inflammatory respiratory disorders with NP1 inhibitors
EP3207944B1 (en) 2008-11-10 2020-01-15 Arbutus Biopharma Corporation Novel lipids and compositions for the delivery of therapeutics
US20100179213A1 (en) * 2008-11-11 2010-07-15 Mirna Therapeutics, Inc. Methods and Compositions Involving miRNAs In Cancer Stem Cells
US9074211B2 (en) 2008-11-19 2015-07-07 Rxi Pharmaceuticals Corporation Inhibition of MAP4K4 through RNAI
SG10201500318SA (en) 2008-12-03 2015-03-30 Arcturus Therapeutics Inc UNA Oligomer Structures For Therapeutic Agents
EP2361306A1 (en) * 2008-12-04 2011-08-31 OPKO Ophthalmics, LLC Compositions and methods for selective inhibition of pro-angiogenic vegf isoforms
TWI686405B (en) 2008-12-09 2020-03-01 建南德克公司 Anti-pd-l1 antibodies and their use to enhance t-cell function
US20100233172A1 (en) 2008-12-16 2010-09-16 Bristol-Myers Squibb Company Methods of inhibiting quiescent tumor proliferation
US20110288155A1 (en) 2008-12-18 2011-11-24 Elena Feinstein Sirna compounds and methods of use thereof
JP5592897B2 (en) 2008-12-26 2014-09-17 サムヤン バイオファーマシューティカルズ コーポレイション Anionic drug-containing pharmaceutical composition and method for producing the same
US9493774B2 (en) 2009-01-05 2016-11-15 Rxi Pharmaceuticals Corporation Inhibition of PCSK9 through RNAi
WO2010090762A1 (en) 2009-02-04 2010-08-12 Rxi Pharmaceuticals Corporation Rna duplexes with single stranded phosphorothioate nucleotide regions for additional functionality
EP2396342B1 (en) 2009-02-12 2014-09-17 Cell Signaling Technology, Inc. Mutant ros expression in human liver cancer
US20110293643A1 (en) 2009-02-13 2011-12-01 Wilkes David S Compounds and methods for inhibiting mmp2 and mmp9
EP2221375A1 (en) 2009-02-20 2010-08-25 Ganymed Pharmaceuticals AG Methods and compositions for diagnosis and treatment of cancer
CN107028969A (en) 2009-02-20 2017-08-11 加尼梅德药物公司 For cancer diagnosis and the method and composition for the treatment of
EP2221063A1 (en) 2009-02-20 2010-08-25 Ganymed Pharmaceuticals AG Methods and compositions for diagnosis and treatment of cancer
EP2669290A1 (en) 2009-03-02 2013-12-04 Alnylam Pharmaceuticals Inc. Nucleic Acid Chemical Modifications
GB2468477A (en) 2009-03-02 2010-09-15 Mina Therapeutics Ltd Double stranded RNA molecule comprising siRNA and miRNA precursors
US8444983B2 (en) 2009-03-23 2013-05-21 Quark Pharmaceuticals, Inc. Composition of anti-ENDO180 antibodies and methods of use for the treatment of cancer and fibrotic diseases
EP2421972A2 (en) 2009-04-24 2012-02-29 The Board of Regents of The University of Texas System Modulation of gene expression using oligomers that target gene regions downstream of 3' untranslated regions
EP2249159A1 (en) 2009-04-29 2010-11-10 Ganymed Pharmaceuticals AG Identification of tumor-associated markers for diagnosis and therapy
MX352992B (en) 2009-05-05 2017-12-15 Beeologics Inc Prevention and treatment of nosema disease in bees.
KR20120013371A (en) * 2009-05-15 2012-02-14 베링거 인겔하임 인터내셔날 게엠베하 Improved cell lines having reduced expression of nocr and use thereof
US20120128673A1 (en) 2009-05-20 2012-05-24 Schering Corporation Modulation of pilr receptors to treat microbial infections
CA2764158A1 (en) 2009-06-01 2010-12-09 Halo-Bio Rnai Therapeutics, Inc. Polynucleotides for multivalent rna interference, compositions and methods of use thereof
EP2258858A1 (en) 2009-06-05 2010-12-08 Universitätsklinikum Freiburg Transgenic LSD1 animal model for cancer
AU2010259984B2 (en) 2009-06-10 2017-03-09 Arbutus Biopharma Corporation Improved lipid formulation
US8435961B2 (en) 2009-06-26 2013-05-07 Massachusetts Institute Of Technology Methods and compositions for increasing the activity of inhibitory RNA
US20110097329A1 (en) 2009-06-26 2011-04-28 Massachusetts Institute Of Technology Compositions and methods for treating cancer and modulating stress granule formation
US8268550B2 (en) 2009-06-26 2012-09-18 Massachusetts Institute Of Technology Compositions and methods for identification of PARP function, inhibitors, and activators
IL292615B2 (en) 2009-07-01 2023-11-01 Protiva Biotherapeutics Inc Nucleic acid-lipid particles, compositions comprising the same and uses thereof
AU2009350151B2 (en) 2009-07-20 2015-07-16 Bristol-Myers Squibb Company Combination of anti-CTLA4 antibody with diverse therapeutic regimens for the synergistic treatment of proliferative diseases
US20110053829A1 (en) 2009-09-03 2011-03-03 Curevac Gmbh Disulfide-linked polyethyleneglycol/peptide conjugates for the transfection of nucleic acids
ES2655079T3 (en) 2009-09-10 2018-02-16 Merck Sharp & Dohme Corp. Use of IL-33 antagonists to treat fibrotic diseases
US8916693B2 (en) 2009-09-17 2014-12-23 Nektar Therapeutics Monoconjugated chitosans as delivery agents for small interfering nucleic acids
US20150025122A1 (en) 2009-10-12 2015-01-22 Larry J. Smith Methods and Compositions for Modulating Gene Expression Using Oligonucleotide Based Drugs Administered in vivo or in vitro
US8962584B2 (en) 2009-10-14 2015-02-24 Yissum Research Development Company Of The Hebrew University Of Jerusalem, Ltd. Compositions for controlling Varroa mites in bees
US9799416B2 (en) * 2009-11-06 2017-10-24 Terrapower, Llc Methods and systems for migrating fuel assemblies in a nuclear fission reactor
US8901097B2 (en) 2009-11-08 2014-12-02 Quark Pharmaceuticals, Inc. Methods for delivery of siRNA to the spinal cord and therapies arising therefrom
EP2499161B8 (en) 2009-11-11 2017-10-25 Ganymed Pharmaceuticals GmbH Antibodies specific for claudin 6 (CLDN6)
CN108707584A (en) 2009-11-17 2018-10-26 Musc研究发展基金会 For the human monoclonal antibodies of people's paranuclein
DK2504435T3 (en) 2009-11-26 2019-12-09 Quark Pharmaceuticals Inc SIRNA CONNECTIONS INCLUDING TERMINAL SUBSTITUTIONS
EP2505660B1 (en) 2009-11-27 2014-05-21 Japan Science And Technology Agency Method for screening of therapeutic agent for hyperlipemia
CA2782728A1 (en) * 2009-12-04 2011-06-09 Opko Ophthalmics, Llc Compositions and methods for inhibition of vegf
WO2011071916A2 (en) 2009-12-07 2011-06-16 The Johns Hopkins University Sr-bi as a predictor of human female infertility and responsiveness to treatment
WO2011071860A2 (en) 2009-12-07 2011-06-16 Alnylam Pharmaceuticals, Inc. Compositions for nucleic acid delivery
TWI543763B (en) 2009-12-09 2016-08-01 日東電工股份有限公司 Modulation of hsp47 expression
WO2011072091A1 (en) 2009-12-09 2011-06-16 Quark Pharmaceuticals, Inc. Methods and compositions for treating diseases, disorders or injury of the cns
WO2011084357A1 (en) 2009-12-17 2011-07-14 Schering Corporation Modulation of pilr to treat immune disorders
KR101605932B1 (en) 2009-12-18 2016-03-24 노파르티스 아게 Organic compositions to treat hsf1-related diseases
AU2010330814B2 (en) 2009-12-18 2017-01-12 Acuitas Therapeutics Inc. Methods and compositions for delivery of nucleic acids
US20130023578A1 (en) 2009-12-31 2013-01-24 Samyang Biopharmaceuticals Corporation siRNA for inhibition of c-Met expression and anticancer composition containing the same
WO2011084193A1 (en) 2010-01-07 2011-07-14 Quark Pharmaceuticals, Inc. Oligonucleotide compounds comprising non-nucleotide overhangs
WO2011094580A2 (en) 2010-01-28 2011-08-04 Alnylam Pharmaceuticals, Inc. Chelated copper for use in the preparation of conjugated oligonucleotides
US9198972B2 (en) 2010-01-28 2015-12-01 Alnylam Pharmaceuticals, Inc. Monomers and oligonucleotides comprising cycloaddition adduct(s)
WO2011098449A1 (en) 2010-02-10 2011-08-18 Novartis Ag Methods and compounds for muscle growth
US9080171B2 (en) 2010-03-24 2015-07-14 RXi Parmaceuticals Corporation Reduced size self-delivering RNAi compounds
EP2550001B1 (en) 2010-03-24 2019-05-22 Phio Pharmaceuticals Corp. Rna interference in ocular indications
AU2011232365A1 (en) 2010-03-24 2012-10-25 Rxi Pharmaceuticals Corporation RNA interference in dermal and fibrotic indications
US9102938B2 (en) 2010-04-01 2015-08-11 Alnylam Pharmaceuticals, Inc. 2′ and 5′ modified monomers and oligonucleotides
EP2558115B1 (en) 2010-04-16 2019-07-31 The Salk Institute for Biological Studies Methods for treating metabolic disorders using fgf
US20130101600A1 (en) 2010-04-19 2013-04-25 Gwendal Lazennec Cxcl5 as a marker of hormone escape in prostate cancer
US20130260460A1 (en) 2010-04-22 2013-10-03 Isis Pharmaceuticals Inc Conformationally restricted dinucleotide monomers and oligonucleotides
US9725479B2 (en) 2010-04-22 2017-08-08 Ionis Pharmaceuticals, Inc. 5′-end derivatives
US10913767B2 (en) 2010-04-22 2021-02-09 Alnylam Pharmaceuticals, Inc. Oligonucleotides comprising acyclic and abasic nucleosides and analogs
HUE040278T2 (en) 2010-04-23 2019-02-28 Cold Spring Harbor Laboratory NOVEL STRUCTURALLY DESIGNED shRNAs
MX350595B (en) 2010-04-23 2017-09-11 Novartis Ag * ORGANIC COMPOSITIONS TO TREAT BETA-ENaC-RELATED DISEASES.
WO2011135530A2 (en) 2010-04-28 2011-11-03 Kimberly-Clark Worldwide, Inc. Device for delivery of rheumatoid arthritis medication
WO2011135531A2 (en) 2010-04-28 2011-11-03 Kimberly-Clark Worldwide, Inc. MEDICAL DEVICES FOR DELIVERY OF siRNA
WO2011135533A2 (en) 2010-04-28 2011-11-03 Kimberly-Clark Worldwide, Inc. Nanopatterned medical device with enhanced cellular interaction
EP2563455A4 (en) 2010-04-28 2014-02-19 Kimberly Clark Co Method for increasing permeability of an epithelial barrier
CA2805267C (en) 2010-05-04 2019-07-30 The Brigham And Women's Hospital, Inc. Detection and treatment of fibrosis
EA030620B1 (en) 2010-05-26 2018-09-28 Селекта Байосайенсиз, Инк. Compositions for generating an immune response to sets of surface antigens comprising synthetic nanocarriers and use thereof
WO2011163121A1 (en) 2010-06-21 2011-12-29 Alnylam Pharmaceuticals, Inc. Multifunctional copolymers for nucleic acid delivery
US9006417B2 (en) 2010-06-30 2015-04-14 Protiva Biotherapeutics, Inc. Non-liposomal systems for nucleic acid delivery
EP2404936A1 (en) 2010-07-06 2012-01-11 Ganymed Pharmaceuticals AG Cancer therapy using CLDN6 target-directed antibodies in vivo
US9011863B2 (en) 2010-07-09 2015-04-21 Exelixis, Inc. Combinations of kinase inhibitors for the treatment of cancer
WO2012016188A2 (en) 2010-07-30 2012-02-02 Alnylam Pharmaceuticals, Inc. Methods and compositions for delivery of active agents
AU2011285200B2 (en) 2010-07-30 2014-08-21 CureVac SE Complexation of nucleic acids with disulfide-crosslinked cationic components for transfection and immunostimulation
US20130323269A1 (en) 2010-07-30 2013-12-05 Muthiah Manoharan Methods and compositions for delivery of active agents
WO2012019132A2 (en) 2010-08-06 2012-02-09 Cell Signaling Technology, Inc. Anaplastic lymphoma kinase in kidney cancer
US20120052079A1 (en) * 2010-08-10 2012-03-01 Dana-Farber Cancer Institute, Inc. Compositions, Kits, and Methods for Predicting Anti-Cancer Response to Anthracyclines
JP6106085B2 (en) 2010-08-24 2017-03-29 サーナ・セラピューティクス・インコーポレイテッドSirna Therapeutics,Inc. Single-stranded RNAi agent containing an internal non-nucleic acid spacer
EP2622076A1 (en) 2010-09-30 2013-08-07 University of Zürich Treatment of b-cell lymphoma with microrna
WO2012046065A2 (en) 2010-10-06 2012-04-12 Omnicyte Limited Culture method
US8835400B2 (en) 2010-10-08 2014-09-16 Mina Therapeutics Limited RNA molecules that upregulate insulin production
US20140134231A1 (en) 2010-10-11 2014-05-15 Sanford-Burnham Medical Research Institute Mir-211 expression and related pathways in human melanoma
WO2012051491A1 (en) 2010-10-14 2012-04-19 The United States Of America, As Represented By The Secretary National Institutes Of Health Compositions and methods for controlling neurotropic viral pathogenesis by micro-rna targeting
EP2634253B1 (en) 2010-10-27 2016-05-11 Jichi Medical University Adeno-associated virus virions for transferring genes into neural cells
DK2632472T3 (en) 2010-10-29 2018-03-19 Sirna Therapeutics Inc RNA INTERFERENCE-MEDIATED INHIBITION OF GENE EXPRESSION USING SHORT INTERFERRING NUCLEIC ACIDS (SINA)
US8569220B2 (en) 2010-11-12 2013-10-29 Jelmar, Llc Hard surface cleaning composition
WO2012071436A1 (en) 2010-11-24 2012-05-31 Genentech, Inc. Method of treating autoimmune inflammatory disorders using il-23r loss-of-function mutants
WO2012072096A1 (en) 2010-12-03 2012-06-07 Biontech Ag Method for cellular rna expression
EP2646557B1 (en) 2010-12-03 2017-08-09 BioNTech RNA Pharmaceuticals GmbH Method for cellular rna expression
CN103298939A (en) 2010-12-06 2013-09-11 夸克医药公司 Double stranded oligonucleotide compounds comprising positional modifications
WO2012090150A2 (en) 2010-12-27 2012-07-05 Compugen Ltd New cell-penetrating peptides and uses thereof
DK3202760T3 (en) 2011-01-11 2019-11-25 Alnylam Pharmaceuticals Inc PEGYLED LIPIDS AND THEIR USE FOR PHARMACEUTICAL SUPPLY
EP2666856A4 (en) 2011-01-19 2015-01-14 Kyowa Hakko Kirin Co Ltd Composition for inhibiting target gene expression
CN103459598B (en) 2011-02-03 2016-08-10 米尔纳医疗股份有限公司 The synthesis analogies of MIR-124
WO2012109495A1 (en) 2011-02-09 2012-08-16 Metabolic Solutions Development Company, Llc Cellular targets of thiazolidinediones
US9796979B2 (en) 2011-03-03 2017-10-24 Quark Pharmaceuticals Inc. Oligonucleotide modulators of the toll-like receptor pathway
CA2828544A1 (en) 2011-03-03 2012-09-07 Quark Pharmaceuticals, Inc. Oligonucleotide modulators of the toll-like receptor pathway
JP6000287B2 (en) 2011-03-03 2016-09-28 クォーク ファーマシューティカルズ インコーポレーティッドQuark Pharmaceuticals,Inc. Compositions and methods for treating lung disease and injury
US10184942B2 (en) 2011-03-17 2019-01-22 University Of South Florida Natriuretic peptide receptor as a biomarker for diagnosis and prognosis of cancer
EP2927323A3 (en) 2011-04-11 2015-12-09 Targeted Growth, Inc. Identification and the use of krp mutants in plants
KR20220116069A (en) 2011-05-13 2022-08-19 가니메드 파마슈티칼스 게엠베하 Antibodies for treatment of cancer expressing claudin 6
WO2012164058A1 (en) 2011-06-01 2012-12-06 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for adjusting expression of mitochondrial genome by microrna
ES2880291T3 (en) 2011-06-02 2021-11-24 Univ Louisville Res Found Inc Nanoparticles conjugated to an antinucleolin agent
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
US10196637B2 (en) 2011-06-08 2019-02-05 Nitto Denko Corporation Retinoid-lipid drug carrier
DK2718442T3 (en) 2011-06-10 2017-08-14 Temasek Life Sciences Laboratory Ltd GENETIC MANIPULATION AND EXPRESSION SYSTEMS FOR SUBPHYLA OF PUCCINIOMYCOTINA AND USTILAGINOMYCOTINA
WO2013003697A1 (en) 2011-06-30 2013-01-03 Trustees Of Boston University Method for controlling tumor growth, angiogenesis and metastasis using immunoglobulin containing and proline rich receptor-1 (igpr-1)
US9120858B2 (en) 2011-07-22 2015-09-01 The Research Foundation Of State University Of New York Antibodies to the B12-transcobalamin receptor
ES2714373T3 (en) 2011-08-01 2019-05-28 Tufts Medical Ct Inc Endogline specific antibody for use in a method of treatment of heart failure and related conditions
EA201490553A1 (en) 2011-09-02 2014-08-29 Новартис Аг ORGANIC COMPOSITIONS FOR THE TREATMENT OF HSF1-CONNECTED DISEASES
US9644241B2 (en) 2011-09-13 2017-05-09 Interpace Diagnostics, Llc Methods and compositions involving miR-135B for distinguishing pancreatic cancer from benign pancreatic disease
EP3456317A1 (en) 2011-09-27 2019-03-20 Alnylam Pharmaceuticals, Inc. Di-aliphatic substituted pegylated lipids
JP2014530612A (en) 2011-10-14 2014-11-20 ジ・オハイオ・ステート・ユニバーシティ Methods and materials for ovarian cancer
JP6100271B2 (en) 2011-10-27 2017-03-22 キンバリー クラーク ワールドワイド インコーポレイテッド Transdermal delivery of highly viscous bioactive agents
US20170246439A9 (en) 2011-10-27 2017-08-31 Kimberly-Clark Worldwide, Inc. Increased Bioavailability of Transdermally Delivered Agents
EP3542851B1 (en) 2011-10-27 2021-12-15 Sorrento Therapeutics, Inc. Implantable devices for delivery of bioactive agents
EP2771482A1 (en) 2011-10-27 2014-09-03 Institut National de la Sante et de la Recherche Medicale (INSERM) Methods for the treatment and diagnosis of atherosclerosis
MX356814B (en) 2011-11-03 2018-06-13 Quark Pharmaceuticals Inc Methods and compositions for neuroprotection.
EP2776565A1 (en) 2011-11-08 2014-09-17 Quark Pharmaceuticals, Inc. Methods and compositions for treating diseases, disorders or injury of the nervous system
CN104302768A (en) 2012-01-09 2015-01-21 诺华股份有限公司 Rnai agents to treat beta-catenin related diseases
US20150126438A1 (en) 2012-01-24 2015-05-07 Beth Israel Deaconess Medical Center, Inc. Novel ChREBP Isoforms and Methods Using the Same
WO2013113326A1 (en) 2012-01-31 2013-08-08 Curevac Gmbh Pharmaceutical composition comprising a polymeric carrier cargo complex and at least one protein or peptide antigen
CN110917362A (en) 2012-02-07 2020-03-27 全球生物疗法有限公司 Compartmentalized methods of nucleic acid delivery, compositions and uses thereof
WO2013123305A1 (en) 2012-02-16 2013-08-22 The Penn State Research Foundation Modulators of acyl-coa lysocardiolipin acyltransferase 1 ( alcat1) and uses thereof
JP2015518475A (en) 2012-04-10 2015-07-02 インサーム(インスティテュ ナシオナル ドゥ ラ サンテ エ ドゥ ラルシェルシェ メディカル)Inserm(Institut National Dela Sante Etde La Recherche Medicale) Treatment method for nonalcoholic steatohepatitis
WO2013153139A1 (en) 2012-04-11 2013-10-17 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for the treatment and diagnosis of acute leukemia
SI2838998T1 (en) 2012-04-18 2018-04-30 Cell Signaling Technology, Inc. Egfr and ros1 in cancer
US20140108091A1 (en) * 2012-04-19 2014-04-17 FullCircle CRM Method and System for Attributing Metrics in a CRM System
US10100321B2 (en) 2012-04-19 2018-10-16 Temasek Life Sciences Laboratory Limited Methods for increasing cotton fiber length
EP3358013B1 (en) 2012-05-02 2020-06-24 Sirna Therapeutics, Inc. Short interfering nucleic acid (sina) compositions
EP2849771A1 (en) 2012-05-16 2015-03-25 Silence Therapeutics GmbH Use of vegfr1 as a biomarker for pkn3 inhibitor administration
US9869519B2 (en) * 2012-07-12 2018-01-16 Google Inc. Thermosiphon systems for electronic devices
US9913907B2 (en) 2012-07-16 2018-03-13 Kyowa Hakko Kirin Co., Ltd. RNAi pharmaceutical composition for suppressing expression of KRAS gene
WO2014018375A1 (en) 2012-07-23 2014-01-30 Xenon Pharmaceuticals Inc. Cyp8b1 and uses thereof in therapeutic and diagnostic methods
WO2014071963A1 (en) 2012-11-09 2014-05-15 Biontech Ag Method for cellular rna expression
AU2013343864B2 (en) 2012-11-09 2019-04-04 BioNTech SE Method for cellular RNA expression
DK2920201T3 (en) 2012-11-15 2020-04-14 Apellis Pharmaceuticals Inc Long-acting compstatin analogs and related compositions and methods
WO2014152391A1 (en) 2013-03-15 2014-09-25 Apellis Pharmaceuticals, Inc. Cell-penetrating compstatin analogs and uses thereof
CA2918194C (en) 2013-03-27 2022-12-06 The General Hospital Corporation Methods and agents for treating alzheimer's disease
CN104211814A (en) 2013-05-29 2014-12-17 三星电子株式会社 Composition for target membrane protein depletion
BR112015029559A2 (en) * 2013-06-19 2017-12-12 Apse Llc virus-like particle, nucleic acid, and methods for producing double-stranded rna and sirna, shrna, sshrna, 1shrna, and mirna
CA2916533C (en) 2013-06-25 2022-12-20 University Of Canberra Methods and compositions for modulating cancer stem cells
EP3608412A3 (en) 2013-07-19 2020-04-08 Monsanto Technology LLC Compositions and methods for controlling leptinotarsa
US20160208247A1 (en) 2013-07-31 2016-07-21 Qbi Enterprises Ltd. Methods of use of sphingolipid polyalkylamine oligonucleotide compounds
WO2015015496A1 (en) 2013-07-31 2015-02-05 Qbi Enterprises Ltd. Sphingolipid-polyalkylamine-oligonucleotide compounds
WO2015014376A1 (en) 2013-07-31 2015-02-05 Biontech Ag Diagnosis and therapy of cancer involving cancer stem cells
CA2920261C (en) 2013-08-08 2018-04-03 Global Bio Therapeutics, Inc. Clamp device for minimally invasive procedures and uses thereof
CN105473158B (en) 2013-08-21 2021-04-13 库瑞瓦格股份公司 Respiratory Syncytial Virus (RSV) vaccine
SG11201601408PA (en) 2013-09-18 2016-04-28 Univ Canberra Stem cell modulation ii
TW202003849A (en) * 2013-10-02 2020-01-16 美商艾爾妮蘭製藥公司 Compositions and methods for inhibiting expression of the LECT2 gene
WO2015051304A1 (en) 2013-10-04 2015-04-09 Aptose Biosciences Inc. Compositions, biomarkers and their use in treatment of cancer
WO2015070158A1 (en) 2013-11-11 2015-05-14 Sirna Therapeutics, Inc. Systemic delivery of myostatin short interfering nucleic acids (sina) conjugated to a lipophilic moiety
US10934550B2 (en) 2013-12-02 2021-03-02 Phio Pharmaceuticals Corp. Immunotherapy of cancer
CA2932904A1 (en) 2013-12-06 2015-06-11 Dicerna Pharmaceuticals, Inc. Methods and compositions for the specific inhibition of transthyretin (ttr) by double-stranded rna
WO2015086828A1 (en) 2013-12-12 2015-06-18 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for the prevention and treatment of diabetic cardiomyopathy using mir-424/322
CA2937118A1 (en) 2014-01-17 2015-07-23 Kyowa Hakko Kirin Co., Ltd. Nucleic acid capable of inhibiting expression of .beta.2gpi
PL3116902T3 (en) 2014-03-11 2020-07-27 Cellectis Method for generating t-cells compatible for allogenic transplantation
WO2015143078A1 (en) 2014-03-18 2015-09-24 University Of Massachusetts Raav-based compositions and methods for treating amyotrophic lateral sclerosis
JP6752151B2 (en) 2014-03-25 2020-09-09 アークトゥラス・セラピューティクス・インコーポレイテッドArcturus Therapeutics,Inc. UNA oligomer with reduced OFF-TARGET effect in gene silencing
EP3122365B1 (en) 2014-03-25 2023-05-03 Arcturus Therapeutics, Inc. Transthyretin allele selective una oligomers for gene silencing
US9856475B2 (en) 2014-03-25 2018-01-02 Arcturus Therapeutics, Inc. Formulations for treating amyloidosis
US11091770B2 (en) 2014-04-01 2021-08-17 Monsanto Technology Llc Compositions and methods for controlling insect pests
WO2015149944A2 (en) 2014-04-01 2015-10-08 Curevac Gmbh Polymeric carrier cargo complex for use as an immunostimulating agent or as an adjuvant
CA2947270A1 (en) 2014-04-28 2015-11-05 Rxi Pharmaceuticals Corporation Methods for treating cancer using nucleic acids targeting mdm2 or mycn
AU2015275128C1 (en) 2014-06-09 2020-02-13 Kyowa Kirin Co., Ltd. The effective and efficient control of serum phosphate for optimal bone formation
JP6264329B2 (en) 2014-06-18 2018-01-24 トヨタ自動車株式会社 Vehicle drive control device
RU2754955C2 (en) 2014-07-29 2021-09-08 Монсанто Текнолоджи Ллс Compositions and methods for combating insect pests
JP2017528452A (en) 2014-08-25 2017-09-28 ユニバーシティ・オブ・キャンベラUniversity of Canberra Composition for modulating cancer stem cells and use thereof
WO2016037071A2 (en) 2014-09-05 2016-03-10 Rxi Pharmaceuticals Corporation Methods for treating aging and skin disorders using nucleic acids targeting tyr or mmp1
WO2016038550A1 (en) 2014-09-11 2016-03-17 Novartis Ag Inhibition of prmt5 to treat mtap-deficiency-related diseases
EP3194581A4 (en) 2014-09-15 2018-04-25 Children's Medical Center Corporation Methods and compositions to increase somatic cell nuclear transfer (scnt) efficiency by removing histone h3-lysine trimethylation
RU2017110868A (en) 2014-09-25 2018-10-25 Колд Спринг Харбор Лаборатори TREATMENT OF RETT SYNDROME
EP3204040B1 (en) 2014-10-10 2021-12-08 Idera Pharmaceuticals, Inc. Treatment of cancer using tlr9 agonists and checkpoint inhibitors
US20170304459A1 (en) 2014-10-10 2017-10-26 Alnylam Pharmaceuticals, Inc. Methods and compositions for inhalation delivery of conjugated oligonucleotide
WO2016061144A1 (en) 2014-10-14 2016-04-21 The Regents Of The University Of California Use of cdk9 and brd4 inhibitors to inhibit inflammation
WO2016062323A1 (en) 2014-10-20 2016-04-28 Biontech Ag Methods and compositions for diagnosis and treatment of cancer
US10266837B2 (en) 2014-10-22 2019-04-23 Temasek Life Sciences Laboratory Limited Terpene synthases from ylang ylang (Cananga odorata var. fruticosa)
JOP20200092A1 (en) 2014-11-10 2017-06-16 Alnylam Pharmaceuticals Inc HEPATITIS B VIRUS (HBV) iRNA COMPOSITIONS AND METHODS OF 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
IL292999A (en) 2014-11-14 2022-07-01 Voyager Therapeutics Inc Compositions and methods of treating amyotrophic lateral sclerosis (als)
JP6863891B2 (en) 2014-11-14 2021-04-21 ボイジャー セラピューティクス インコーポレイテッドVoyager Therapeutics,Inc. Regulatory polynucleotide
EP3227446A1 (en) 2014-12-01 2017-10-11 Novartis AG Compositions and methods for diagnosis and treatment of prostate cancer
WO2016089872A1 (en) 2014-12-03 2016-06-09 Glycomimetics, Inc. Heterobifunctional inhibitors of e-selectins and cxcr4 chemokine receptors
MX2017009521A (en) 2015-01-22 2018-11-09 Monsanto Technology Llc Compositions and methods for controlling leptinotarsa.
AU2016226414B2 (en) 2015-03-02 2021-09-09 180 Therapeutics Lp Method of treating a localized fibrotic disorder using an IL-33 antagonist
WO2016145005A1 (en) 2015-03-09 2016-09-15 University Of Kentucky Research Foundation Rna nanoparticles for brain tumor treatment
EP3277289A4 (en) 2015-04-01 2018-12-05 Arcturus Therapeutics, Inc. Therapeutic una oligomers and uses thereof
MX2017012610A (en) 2015-04-08 2018-03-16 Alnylam Pharmaceuticals Inc Compositions and methods for inhibiting expression of the lect2 gene.
WO2016179394A1 (en) 2015-05-05 2016-11-10 Malik Mohammad Tariq Anti-nucleolin agent-conjugated nanoparticles as radio-sensitizers and mri and/or x-ray contrast agents
AU2016269839B2 (en) 2015-06-03 2021-07-08 The University Of Queensland Mobilizing agents and uses therefor
WO2016193945A2 (en) 2015-06-05 2016-12-08 Novartis Ag Methods and compositions for diagnosing, treating, and monitoring treatment of shank3 deficiency associated disorders
US10808247B2 (en) 2015-07-06 2020-10-20 Phio Pharmaceuticals Corp. Methods for treating neurological disorders using a synergistic small molecule and nucleic acids therapeutic approach
US11001845B2 (en) 2015-07-06 2021-05-11 Phio Pharmaceuticals Corp. Nucleic acid molecules targeting superoxide dismutase 1 (SOD1)
WO2017015671A1 (en) 2015-07-23 2017-01-26 Arcturus Therapeutics, Inc. Compositions for treating amyloidosis
BR112018003784A2 (en) 2015-08-24 2018-09-25 Halo-Bio Rnai Therapeutics, Inc. polynucleotide nanoparticles for the modulation of gene expression and its use
EP3357491B1 (en) 2015-09-15 2021-06-02 Samyang Biopharmaceuticals Corporation Pharmaceutical composition for delivering an anionic drug
AU2016332900C1 (en) 2015-09-29 2024-02-01 Amgen Inc. ASGR inhibitors
EP3359555B1 (en) 2015-10-07 2023-12-20 Apellis Pharmaceuticals, Inc. Dosing regimens
CN109563509B (en) 2015-10-19 2022-08-09 菲奥医药公司 Reduced size self-delivering nucleic acid compounds targeting long non-coding RNAs
AU2016372321B2 (en) 2015-12-18 2019-01-24 Samyang Holdings Corporation Method for preparing polymeric micelle containing anionic drug
US11856951B2 (en) 2015-12-22 2024-01-02 Provivi, Inc. Method for managing resistance to insecticidal traits and chemicals using pheromones
WO2017151708A1 (en) 2016-03-02 2017-09-08 Glycomimetics, Inc. Methods for the treatment and/or prevention of cardiovescular disease by inhibition of e-selectin
US11072777B2 (en) 2016-03-04 2021-07-27 University Of Louisville Research Foundation, Inc. Methods and compositions for ex vivo expansion of very small embryonic-like stem cells (VSELs)
UY37146A (en) 2016-03-07 2017-09-29 Arrowhead Pharmaceuticals Inc ADDRESSING LINKS FOR THERAPEUTIC COMPOUNDS
US10947317B2 (en) 2016-03-15 2021-03-16 Mersana Therapeutics, Inc. NaPi2b-targeted antibody-drug conjugates and methods of use thereof
MA45470A (en) 2016-04-01 2019-02-06 Avidity Biosciences Llc KRAS NUCLEIC ACIDS AND THEIR USES
MA45349A (en) 2016-04-01 2019-02-06 Avidity Biosciences Llc EGFR NUCLEIC ACIDS AND THEIR USES
MA45328A (en) 2016-04-01 2019-02-06 Avidity Biosciences Llc NUCLEIC ACID-POLYPEPTIDE COMPOSITIONS AND USES THEREOF
MA45469A (en) 2016-04-01 2019-02-06 Avidity Biosciences Llc BETA-CATENIN NUCLEIC ACIDS AND THEIR USES
US9988641B2 (en) 2016-04-05 2018-06-05 Corn Products Development, Inc. Compositions and methods for producing starch with novel functionality
ES2900272T3 (en) 2016-04-22 2022-03-16 BioNTech SE Methods for providing single-stranded RNA
PT3445773T (en) 2016-05-13 2023-03-13 4D Molecular Therapeutics Inc Adeno-associated virus variant capsids and methods of use thereof
IL302748A (en) 2016-05-18 2023-07-01 Voyager Therapeutics Inc Modulatory polynucleotides
BR112018073472A2 (en) 2016-05-18 2019-08-27 Voyager Therapeutics Inc Huntington's disease treatment compositions and methods
PT109454A (en) 2016-06-14 2017-12-14 Phyzat Biopharmaceuticals Lda NUCLEIC ACIDS OF INTERFERENCE AND COMPOSITIONS THAT UNDERSTAND THEM
AU2017290828A1 (en) 2016-06-30 2019-01-24 Virogin Biotech Canada Ltd Pseudotyped oncolytic viral delivery of therapeutic polypeptides
US20190367930A1 (en) 2016-07-29 2019-12-05 Danmarks Tekniske Universitet Methods for decoupling cell growth from production of biochemicals and recombinant polypeptides
WO2018031445A1 (en) 2016-08-08 2018-02-15 Glycomimetics, Inc. Combination of t-cell checkpoint inhibitors with inhibitors of e-selectin or cxcr4, or with heterobifunctional inhibitors of both e-selectin and cxcr4
KR102557906B1 (en) 2016-09-02 2023-07-20 애로우헤드 파마슈티컬스 인코포레이티드 Targeting ligands
WO2018057575A1 (en) 2016-09-21 2018-03-29 Alnylam Pharmaceuticals, Inc Myostatin irna compositions and methods of use thereof
WO2018068010A1 (en) 2016-10-07 2018-04-12 Glycomimetics, Inc. Highly potent multimeric e-selectin antagonists
WO2018083606A1 (en) 2016-11-01 2018-05-11 Novartis Ag Methods and compositions for enhancing gene editing
US11135307B2 (en) 2016-11-23 2021-10-05 Mersana Therapeutics, Inc. Peptide-containing linkers for antibody-drug conjugates
US10450565B2 (en) 2017-01-10 2019-10-22 Arrowhead Pharmaceuticals, Inc. Alpha-1 antitrypsin (AAT) RNAi agents, compositions including AAT RNAi agents, and methods of use
EP3569708A4 (en) 2017-01-13 2020-09-09 Jichi Medical University Aav vector for disrupting clotting-related factor gene on liver genome
US20180271996A1 (en) 2017-02-28 2018-09-27 Mersana Therapeutics, Inc. Combination therapies of her2-targeted antibody-drug conjugates
TW201837172A (en) 2017-03-09 2018-10-16 日商協和醱酵麒麟有限公司 Nucleic acid capable of inhibiting expression of masp2
EP3596096A1 (en) 2017-03-15 2020-01-22 GlycoMimetics, Inc. Galactopyranosyl-cyclohexyl derivatives as e-selectin antagonists
WO2018186032A1 (en) 2017-04-05 2018-10-11 国立大学法人千葉大学 Function inhibitor of swi/snf complexes
BR112019020955A2 (en) 2017-04-07 2020-05-05 Apellis Pharmaceuticals Inc dosage regimens and related compositions and methods
US11324820B2 (en) 2017-04-18 2022-05-10 Alnylam Pharmaceuticals, Inc. Methods for the treatment of subjects having a hepatitis b virus (HBV) infection
CN106973864A (en) * 2017-04-25 2017-07-25 遵义医学院 A kind of breeding apparatus and its application method suitable for white backed planthopper injection RNA interference experiments
CN111108198A (en) 2017-05-05 2020-05-05 沃雅戈治疗公司 Compositions and methods for treating huntington's disease
AU2018261790A1 (en) 2017-05-05 2019-11-28 Voyager Therapeutics, Inc. Compositions and methods of treating amyotrophic lateral sclerosis (ALS)
WO2018208972A1 (en) 2017-05-09 2018-11-15 University Of Massachusetts Methods of treating amyotrophic lateral sclerosis (als)
AU2018277476A1 (en) 2017-05-31 2020-01-02 Kyowa Kirin Co., Ltd. APCS-expression-suppressing nucleic acids
US11530413B2 (en) 2017-07-21 2022-12-20 Novartis Ag Compositions and methods to treat cancer
WO2019060454A2 (en) 2017-09-20 2019-03-28 4D Molecular Therapeutics Inc. Adeno-associated virus variant capsids and methods of use thereof
JP7397488B2 (en) 2017-09-22 2023-12-13 ユニバーシティ オブ マサチューセッツ SOD1 dual expression vector and its use
US20200237799A1 (en) 2017-10-16 2020-07-30 Voyager Therapeutics, Inc. Treatment of amyotrophic lateral sclerosis (als)
WO2019079240A1 (en) 2017-10-16 2019-04-25 Voyager Therapeutics, Inc. Treatment of amyotrophic lateral sclerosis (als)
MX2020004060A (en) 2017-10-20 2020-07-21 Dicerna Pharmaceuticals Inc Methods for treating hepatitis b infection.
SG11202004545XA (en) 2017-11-27 2020-06-29 4D Molecular Therapeutics Inc Adeno-associated virus variant capsids and use for inhibiting angiogenesis
EP3717021A1 (en) 2017-11-27 2020-10-07 Mersana Therapeutics, Inc. Pyrrolobenzodiazepine antibody conjugates
WO2019108750A1 (en) 2017-11-30 2019-06-06 Glycomimetics, Inc. Methods of mobilizing marrow infiltrating lymphocytes and uses thereof
AU2018378812A1 (en) * 2017-12-06 2020-07-09 Avidity Biosciences, Inc. Compositions and methods of treating muscle atrophy and myotonic dystrophy
JP2021506883A (en) 2017-12-21 2021-02-22 メルサナ セラピューティクス インコーポレイテッド Pyrrolobenzodiazepine antibody conjugate
EP3731850A4 (en) 2017-12-29 2021-12-01 Oncorus, Inc. Oncolytic viral delivery of therapeutic polypeptides
JP7304863B2 (en) 2017-12-29 2023-07-07 グリコミメティクス, インコーポレイテッド Heterobifunctional inhibitors of E-selectin and galectin-3
US11661603B2 (en) 2018-01-16 2023-05-30 Dicerna Pharmaceuticals, Inc. Compositions and methods for inhibiting ALDH2 expression
CA3091454A1 (en) 2018-03-05 2019-09-12 Glycomimetics, Inc. Methods for treating acute myeloid leukemia and related conditions
US20210123016A1 (en) 2018-05-02 2021-04-29 Novartis Ag Regulators of human pluripotent stem cells and uses thereof
WO2020026968A1 (en) 2018-07-30 2020-02-06 株式会社遺伝子治療研究所 Method for enhancing gene expression by aav vector
WO2020036862A1 (en) 2018-08-13 2020-02-20 Alnylam Pharmaceuticals, Inc. HEPATITIS B VIRUS (HBV) dsRNA AGENT COMPOSITIONS AND METHODS OF USE THEREOF
EA202191175A1 (en) 2018-10-29 2021-09-08 Мерсана Терапьютикс, Инк. CYSTEINE-DESIGNED ANTIBODY-DRUG CONJUGATES CONTAINING PEPTIDE-CONTAINING LINKERS
US11845771B2 (en) 2018-12-27 2023-12-19 Glycomimetics, Inc. Heterobifunctional inhibitors of E-selectin and galectin-3
JP2022520653A (en) 2019-02-12 2022-03-31 ディセルナ ファーマシューティカルズ インコーポレイテッド Methods and Compositions for Inhibiting Expression of CYP27A1
SG11202110896WA (en) 2019-04-04 2021-10-28 Dicerna Pharmaceuticals Inc Compositions and methods for inhibiting gene expression in the central nervous system
JP2022529917A (en) 2019-04-18 2022-06-27 アンスティチュ ナショナル ドゥ ラ サンテ エ ドゥ ラ ルシェルシュ メディカル Methods for cancer treatment and prognosis
BR112021026720A2 (en) * 2019-07-02 2022-02-15 Na Vaccine Inst heterostructured RNA, homogeneous dsrna, pharmaceutical composition, homogeneous pharmaceutical agent and therapeutic agent for cancer
WO2021009805A1 (en) 2019-07-12 2021-01-21 株式会社遺伝子治療研究所 Adeno-associated virus virion for gene transfer to human liver
EP4081217A1 (en) 2019-12-24 2022-11-02 F. Hoffmann-La Roche AG Pharmaceutical combination of antiviral agents targeting hbv and/or an immune modulator for treatment of hbv
JP2023509870A (en) 2019-12-24 2023-03-10 エフ. ホフマン-ラ ロシュ アーゲー Pharmaceutical combinations of therapeutic oligonucleotides targeting HBV and TLR7 agonists for the treatment of HBV
CN116096889A (en) 2020-03-18 2023-05-09 迪克纳制药公司 Compositions and methods for inhibiting ANGPTL3 expression
IL296387B1 (en) 2020-03-19 2024-04-01 Avidity Biosciences Inc Compositions and methods of treating facioscapulohumeral muscular dystrophy
CA3176356A1 (en) 2020-04-22 2021-10-28 Anne BROOKS Systems and methods for coordinating manufacturing of cells for patient-specific immunotherapy
US20210332364A1 (en) 2020-04-28 2021-10-28 Phyzat Biopharmaceuticals, Lda siNA MOLECULES, METHODS OF PRODUCTION AND USES THEREOF
WO2021228585A1 (en) 2020-05-09 2021-11-18 Phyzat Biopharmaceuticals, Lda Sina molecules, methods of production and uses thereof
CN115702243A (en) 2020-06-09 2023-02-14 罗氏创新中心哥本哈根有限公司 Guanosine analogs for therapeutic polynucleotides
TW202221120A (en) 2020-08-04 2022-06-01 美商黛瑟納製藥公司 Compositions and methods for the treatment of metabolic syndrome
IL300283A (en) 2020-08-04 2023-04-01 Dicerna Pharmaceuticals Inc Systemic delivery of oligonucleotides
WO2022029209A1 (en) 2020-08-05 2022-02-10 F. Hoffmann-La Roche Ag Oligonucleotide treatment of hepatitis b patients
JP7476422B2 (en) 2020-08-05 2024-04-30 ディセルナ ファーマシューティカルズ インコーポレイテッド Compositions and methods for inhibiting LPA expression
WO2022107106A2 (en) 2020-11-23 2022-05-27 Phyzat Biopharmaceuticals, Lda Sina molecules, methods of production and uses thereof
AU2021385572A1 (en) 2020-11-25 2023-06-22 Akagera Medicines, Inc. Lipid nanoparticles for delivery of nucleic acids, and related methods of use
CN112511569B (en) * 2021-02-07 2021-05-11 杭州筋斗腾云科技有限公司 Method and system for processing network resource access request and computer equipment
CA3215103A1 (en) 2021-04-12 2022-10-20 Steffen Panzner Rna compositions comprising a buffer substance and methods for preparing, storing and using the same
US20220340909A1 (en) 2021-04-12 2022-10-27 Boehringer Ingelheim International Gmbh Compositions and methods for inhibiting ketohexokinase (khk)
TW202300645A (en) 2021-04-14 2023-01-01 美商戴瑟納製藥股份有限公司 Compositions and methods for modulating pnpla3 expression
BR112023021614A2 (en) 2021-04-19 2024-01-16 Dicerna Pharmaceuticals Inc COMPOSITIONS AND METHODS FOR INHIBITING THE EXPRESSION OF NUCLEAR RECEPTORS SUFAMILY 1, GROUP H, MEMBER 3 (NR1H3)
WO2022224372A1 (en) 2021-04-21 2022-10-27 学校法人自治医科大学 Adeno-associated virus virion for treating ornithine transcarbamylase deficiency
BR112023024375A2 (en) 2021-05-28 2024-02-15 Shanghai Regenelead Therapies Co Ltd RECOMBINANT ADENO-ASSOCIATED VIRUS HAVING VARIANT CAPSID AND ITS APPLICATION
JP7463621B2 (en) 2021-05-28 2024-04-08 ノヴォ ノルディスク アー/エス Compositions and methods for inhibiting mitochondrial amidoxime reducing component 1 (MARC1) expression
WO2023027759A1 (en) 2021-08-25 2023-03-02 Dicerna Pharmaceuticals, Inc. Compositions and methods for inhibiting αlpha-1 antitrypsin expression
CA3234478A1 (en) 2021-11-11 2023-05-19 Souphalone LUANGSAY Pharmaceutical combinations for treatment of hbv
AR127843A1 (en) 2021-12-01 2024-03-06 Dicerna Pharmaceuticals Inc COMPOSITIONS AND METHODS TO MODULATE THE EXPRESSION OF APOC3
WO2023118546A2 (en) 2021-12-23 2023-06-29 Boehringer Ingelheim International Gmbh METHODS AND MOLECULES FOR RNA INTERFERENCE (RNAi)
WO2023193892A1 (en) 2022-04-05 2023-10-12 BioNTech SE Nucleic acid compositions comprising an inorganic polyphosphate and methods for preparing, storing and using the same
WO2023201043A1 (en) 2022-04-15 2023-10-19 Dicerna Pharmaceuticals, Inc. Compositions and methods for modulating scap activity
US20230416742A1 (en) 2022-05-12 2023-12-28 Dicerna Phrmaceuticals, Inc. Compositions and methods for inhibiting mapt expression
WO2023220351A1 (en) 2022-05-13 2023-11-16 Dicerna Pharmaceuticals, Inc. Compositions and methods for inhibiting snca expression
TW202400193A (en) 2022-06-24 2024-01-01 丹麥商諾佛 儂迪克股份有限公司 Compositions and methods for inhibiting transmembrane serine protease 6 (tmprss6) expression
WO2024028325A1 (en) 2022-08-01 2024-02-08 BioNTech SE Nucleic acid compositions comprising amphiphilic oligo ethylene glycol (oeg)-conjugated compounds and methods of using such compounds and compositions
WO2024107993A1 (en) 2022-11-16 2024-05-23 Dicerna Pharmaceuticals, Inc. Stat3 targeting oligonucleotides and uses thereof

Family Cites Families (138)

* 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
JPS59198885A (en) 1983-04-25 1984-11-10 Nec Corp Piezoelectric actuator exciting system
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
GB8704365D0 (en) 1987-02-25 1987-04-01 Exxon Chemical Patents Inc Zeolite l preparation
IE66830B1 (en) 1987-08-12 1996-02-07 Hem Res Inc Topically active compositions of double-stranded RNAs
US5712257A (en) 1987-08-12 1998-01-27 Hem Research, Inc. Topically active compositions of mismatched dsRNAs
US5703055A (en) * 1989-03-21 1997-12-30 Wisconsin Alumni Research Foundation Generation of antibodies through lipid mediated DNA delivery
EP0942000B1 (en) * 1989-10-24 2004-06-23 Isis Pharmaceuticals, Inc. 2'-Modified oligonucleotides
US5457189A (en) * 1989-12-04 1995-10-10 Isis Pharmaceuticals Antisense oligonucleotide inhibition of papillomavirus
US5670633A (en) * 1990-01-11 1997-09-23 Isis Pharmaceuticals, Inc. Sugar modified oligonucleotides that detect and modulate gene expression
BR9105935A (en) 1990-01-11 1992-11-17 Isis Pharmaceuticals Inc COMPOSITION, OLIGONUCLEOTIDEO ANALOG, MIXED SEQUENCED OLIGONUCLEOTIDEO, PROCESS FOR THE PREPARATION OF 2'-SUBSTITUTED NUCLEOSASES PREPARATION, COMPOSITION TO MODULATE THE RNA ACTIVITY, PROCESSES TO MODULAR THE PRODUCTION OF A PROTEIN FOR AN ORGANIC PROTECTION OR ABSENCE OF RNA, OLIGONUCLEOTIDEO OR NUCLEASE RESISTANT OLIGONUCLEOTIDE ANALOG ANALOG AND PROCESS TO TREAT AN ORGANISM HAVING A DISEASE
US5514577A (en) * 1990-02-26 1996-05-07 Isis Pharmaceuticals, Inc. Oligonucleotide therapies for modulating the effects of herpes viruses
CA2093664C (en) * 1990-10-12 2003-07-29 Fritz Eckstein Modified ribozymes
FR2675803B1 (en) 1991-04-25 1996-09-06 Genset Sa CLOSED, ANTISENSE AND SENSE OLIGONUCLEOTIDES AND THEIR APPLICATIONS.
WO1994008003A1 (en) * 1991-06-14 1994-04-14 Isis Pharmaceuticals, Inc. ANTISENSE OLIGONUCLEOTIDE INHIBITION OF THE ras GENE
FR2685346B1 (en) * 1991-12-18 1994-02-11 Cis Bio International PROCESS FOR THE PREPARATION OF DOUBLE-STRANDED RNA, AND ITS APPLICATIONS.
IL104965A (en) 1992-03-05 1999-11-30 Isis Pharmaceuticals Inc Covalently cross-linked oligonucleotides
US5792751A (en) * 1992-04-13 1998-08-11 Baylor College Of Medicine Tranformation of cells associated with fluid spaces
US20040054156A1 (en) * 1992-05-14 2004-03-18 Kenneth Draper Method and reagent for inhibiting hepatitis B viral replication
WO2002081494A1 (en) * 2001-03-26 2002-10-17 Sirna Therapeutics, Inc. Oligonucleotide mediated inhibition of hepatitis b virus and hepatitis c virus replication
US20030068301A1 (en) * 1992-05-14 2003-04-10 Kenneth Draper Method and reagent for inhibiting hepatitis B virus replication
US20030206887A1 (en) * 1992-05-14 2003-11-06 David Morrissey RNA interference mediated inhibition of hepatitis B virus (HBV) using short interfering nucleic acid (siNA)
US20030171311A1 (en) * 1998-04-27 2003-09-11 Lawrence Blatt Enzymatic nucleic acid treatment of diseases or conditions related to hepatitis C virus infection
US5693535A (en) * 1992-05-14 1997-12-02 Ribozyme Pharmaceuticals, Inc. HIV targeted ribozymes
JPH08501928A (en) 1992-07-02 1996-03-05 ハイブライドン インコーポレイテッド Self-stabilizing oligonucleotides as therapeutic agents
US5652355A (en) 1992-07-23 1997-07-29 Worcester Foundation For Experimental Biology Hybrid oligonucleotide phosphorothioates
WO1994015645A1 (en) 1992-12-31 1994-07-21 Texas Biotechnology Corporation Antisense molecules directed against genes of the raf oncogene family
US6056704A (en) 1993-03-03 2000-05-02 Ide; Masatake Foot-pressure massage stand
EP0616026A1 (en) 1993-03-19 1994-09-21 The Procter & Gamble Company Concentrated cleaning compositions
WO1995000638A2 (en) * 1993-06-23 1995-01-05 Genesys Pharma Inc. Antisense oligonucleotides and therapeutic use thereof in human immunodeficiency virus infection
FR2710074B1 (en) 1993-09-15 1995-12-08 Rhone Poulenc Rorer Sa GRB3-3 gene, its variants and their uses.
US5624803A (en) * 1993-10-14 1997-04-29 The Regents Of The University Of California In vivo oligonucleotide generator, and methods of testing the binding affinity of triplex forming oligonucleotides derived therefrom
US5801154A (en) * 1993-10-18 1998-09-01 Isis Pharmaceuticals, Inc. Antisense oligonucleotide modulation of multidrug resistance-associated protein
IL111660A (en) 1993-11-16 2005-05-17 Genta Inc Oligonucleoside compounds for effecting rnaseh-mediated cleavage of a target ribonucleic acid sequence and a pharmaceutical composition containing them
US5578716A (en) * 1993-12-01 1996-11-26 Mcgill University DNA methyltransferase antisense oligonucleotides
US5908779A (en) * 1993-12-01 1999-06-01 University Of Connecticut Targeted RNA degradation using nuclear antisense RNA
JPH10503364A (en) * 1994-05-10 1998-03-31 ザ ジェネラル ホスピタル コーポレーション Antisense inhibition of hepatitis C virus
US6057153A (en) * 1995-01-13 2000-05-02 Yale University Stabilized external guide sequences
US5674683A (en) 1995-03-21 1997-10-07 Research Corporation Technologies, Inc. Stem-loop and circular oligonucleotides and method of using
US5624808A (en) * 1995-03-28 1997-04-29 Becton Dickinson And Company Method for identifying cells committed to apoptosis by determining cellular phosphotyrosine content
ATE285477T1 (en) 1995-06-07 2005-01-15 Inex Pharmaceutical Corp PRODUCTION OF LIPID-NUCLIC ACID PARTICLES USING A HYDROPHOBIC LIPID-NUCLIC ACID COMPLEX INTERMEDIATE PRODUCT AND FOR USE IN GENE TRANSFER
WO1997011170A1 (en) 1995-09-20 1997-03-27 Worcester Foundation For Biomedical Research Antisense oligonucleotide chemotherapy for benign hyperplasia or cancer of the prostate
US5998203A (en) * 1996-04-16 1999-12-07 Ribozyme Pharmaceuticals, Inc. Enzymatic nucleic acids containing 5'-and/or 3'-cap structures
EP0882061B1 (en) 1996-02-14 2004-05-19 Isis Pharmaceuticals, Inc. Sugar-modified gapped oligonucleotides
BR9708701A (en) 1996-04-17 2000-01-04 Hoechst Marion Roussel De Gmbh Anti-sense inhibitors of vascular endothelial growth factor expression.
DE19618797C2 (en) 1996-05-10 2000-03-23 Bertling Wolf Vehicle for the transport of molecular substances
US5898031A (en) 1996-06-06 1999-04-27 Isis Pharmaceuticals, Inc. Oligoribonucleotides for cleaving RNA
US20040266706A1 (en) 2002-11-05 2004-12-30 Muthiah Manoharan Cross-linked oligomeric compounds and their use in gene modulation
DE19631919C2 (en) 1996-08-07 1998-07-16 Deutsches Krebsforsch Anti-sense RNA with secondary structure
US6225290B1 (en) * 1996-09-19 2001-05-01 The Regents Of The University Of California Systemic gene therapy by intestinal cell transformation
EP0972015B1 (en) 1996-10-04 2006-06-07 Derek Nigel John Hart Enzyme having s-adenosyl-l-homocysteine hydrolase (ahcy) type activity
US5814500A (en) * 1996-10-31 1998-09-29 The Johns Hopkins University School Of Medicine Delivery construct for antisense nucleic acids and methods of use
JP4427639B2 (en) 1996-12-12 2010-03-10 イスム リサーチ ディベロップメント カンパニー オブ ザ ヘブライ ユニバーシティ オブ エルサレム Synthetic antisense oligodeoxynucleotides and pharmaceutical compositions containing them
US20030064945A1 (en) * 1997-01-31 2003-04-03 Saghir Akhtar Enzymatic nucleic acid treatment of diseases or conditions related to levels of epidermal growth factor receptors
GB9703146D0 (en) * 1997-02-14 1997-04-02 Innes John Centre Innov Ltd Methods and means for gene silencing in transgenic plants
US6218142B1 (en) * 1997-03-05 2001-04-17 Michael Wassenegger Nucleic acid molecules encoding polypeptides having the enzymatic activity of an RNA-directed RNA polymerase (RDRP)
GB9710475D0 (en) 1997-05-21 1997-07-16 Zeneca Ltd Gene silencing
DE04020014T1 (en) 1997-09-12 2006-01-26 Exiqon A/S Bi-cyclic - nucleoside, nucleotide and oligonucleotide analogs
EP2292771A3 (en) 1997-09-19 2011-07-27 Life Technologies Corporation Sense mRNA therapy
GB9720148D0 (en) * 1997-09-22 1997-11-26 Innes John Centre Innov Ltd Gene silencing materials and methods
US6506559B1 (en) * 1997-12-23 2003-01-14 Carnegie Institute Of Washington Genetic inhibition by double-stranded RNA
US6475726B1 (en) * 1998-01-09 2002-11-05 Cubist Pharmaceuticals, Inc. Method for identifying validated target and assay combinations for drug development
CZ295108B6 (en) 1998-03-20 2005-05-18 Benitec Australia Ltd Synthetic gene comprising dispersed or foreign deoxyribonucleic molecule and a gene construct containing such a synthetic gene
AUPP249298A0 (en) * 1998-03-20 1998-04-23 Ag-Gene Australia Limited Synthetic genes and genetic constructs comprising same I
ES2624549T3 (en) 1998-04-08 2017-07-14 Commonwealth Scientific And Industrial Research Organisati Methods and means to obtain modified phenotypes
US20040214330A1 (en) * 1999-04-07 2004-10-28 Waterhouse Peter Michael Methods and means for obtaining modified phenotypes
CA2326823A1 (en) 1998-04-20 1999-10-28 Ribozyme Pharmaceuticals, Inc. Nucleic acid molecules with novel chemical compositions capable of modulating gene expression
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
EP1024502A4 (en) * 1998-08-05 2001-10-24 Sony Corp Composition for electrolyte, electrolyte and process for producing the same, and cell containing the same
EP1050583A4 (en) 1998-11-24 2005-02-02 Hisamitsu Pharmaceutical Co Hiv infection inhibitors
WO2000032619A1 (en) 1998-11-30 2000-06-08 Ribogene, Inc. Methods and compositions for identification of inhibitors of ribosome assembly
US6939712B1 (en) * 1998-12-29 2005-09-06 Impedagen, Llc Muting gene activity using a transgenic nucleic acid
EP2314700A1 (en) 1999-01-28 2011-04-27 Medical College of Georgia Research Institute, Inc Composition and method for in vivo and in vitro attenuation of gene expression using double stranded RNA
DE19956568A1 (en) * 1999-01-30 2000-08-17 Roland Kreutzer Method and medicament for inhibiting the expression of a given gene
IL145778A0 (en) 1999-04-21 2002-07-25 American Home Prod Methods and compositions for inhibiting the function of polynucleotide sequences
US20040002153A1 (en) * 1999-07-21 2004-01-01 Monia Brett P. Modulation of PTEN expression via oligomeric compounds
US6367949B1 (en) * 1999-08-04 2002-04-09 911 Emergency Products, Inc. Par 36 LED utility lamp
GB9925459D0 (en) * 1999-10-27 1999-12-29 Plant Bioscience Ltd Gene silencing
GB9927444D0 (en) 1999-11-19 2000-01-19 Cancer Res Campaign Tech Inhibiting gene expression
US7829693B2 (en) * 1999-11-24 2010-11-09 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of a target gene
DE10160151A1 (en) 2001-01-09 2003-06-26 Ribopharma Ag Inhibiting expression of target gene, useful e.g. for inhibiting oncogenes, by administering double-stranded RNA complementary to the target and having an overhang
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
RU2164944C1 (en) * 1999-12-09 2001-04-10 Институт молекулярной биологии им. В.А. Энгельгардта РАН Method of alternation of organism genetic features
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
AU2001260114A1 (en) 2000-03-14 2001-09-24 Syngenta Participations Ag Protoporphyrinogen oxidase ("protox") genes
EP1272630A2 (en) * 2000-03-16 2003-01-08 Genetica, Inc. Methods and compositions for rna interference
US20030084471A1 (en) * 2000-03-16 2003-05-01 David Beach Methods and compositions for RNA interference
ES2461715T3 (en) 2000-03-30 2014-05-21 Whitehead Institute For Biomedical Research Specific RNA Interference Sequence RNA Mediators
IL151928A0 (en) 2000-03-30 2003-04-10 Whitehead Biomedical Inst Rna sequence-specific mediators of rna interference
EP1290161B1 (en) 2000-05-30 2011-06-22 Johnson & Johnson Research Pty Limited METHODS FOR MEDIATING GENE SUPPRESION BY USING FACTORS THAT ENHANCE RNAi
ES2215494T5 (en) 2000-12-01 2017-12-28 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. Small RNA molecules that mediate RNA interference
NZ526194A (en) 2000-12-08 2004-10-29 Invitrogen Corp Methods of covalently linking, in one or both strands, two or more double stranded nucleotide sequences using one or more topoisomerases
CA2433680A1 (en) 2000-12-28 2002-08-01 Gregory M Arndt Double-stranded rna-mediated gene suppression
US7423142B2 (en) * 2001-01-09 2008-09-09 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of anti-apoptotic genes
WO2003035869A1 (en) 2001-10-26 2003-05-01 Ribopharma Ag Use of a double-stranded ribonucleic acid for specifically inhibiting the expression of a given target gene
US20020132257A1 (en) * 2001-01-31 2002-09-19 Tony Giordano Use of post-transcriptional gene silencing for identifying nucleic acid sequences that modulate the function of a cell
US20040019001A1 (en) * 2002-02-20 2004-01-29 Mcswiggen James A. RNA interference mediated inhibition of protein typrosine phosphatase-1B (PTP-1B) gene expression using short interfering RNA
US20030124513A1 (en) * 2001-05-29 2003-07-03 Mcswiggen James Enzymatic nucleic acid treatment of diseases or conditions related to levels of HIV
US20040006035A1 (en) * 2001-05-29 2004-01-08 Dennis Macejak Nucleic acid mediated disruption of HIV fusogenic peptide interactions
EP1263250B1 (en) 2001-06-01 2004-03-24 Mobilkom Austria Aktiengesellschaft & Co KG Method to determine the location of a mobile station in a mobile radio system
US20030140362A1 (en) * 2001-06-08 2003-07-24 Dennis Macejak In vivo models for screening inhibitors of hepatitis B virus
US6586684B2 (en) * 2001-06-29 2003-07-01 Intel Corporation Circuit housing clamp and method of manufacture therefor
US6900289B2 (en) * 2001-08-22 2005-05-31 The University Of Hawaii Physalia fluorescent proteins
CA2462144C (en) 2001-09-28 2016-09-20 Max-Planck-Gesellschaft Zur Forderung Der Wissenschaften E.V. Micro-rna molecules
DE10163098B4 (en) 2001-10-12 2005-06-02 Alnylam Europe Ag Method for inhibiting the replication of viruses
US20050119202A1 (en) * 2001-10-26 2005-06-02 Roland Kreutzer Medicament to treat a fibrotic disease
US20040121348A1 (en) * 2001-10-26 2004-06-24 Ribopharma Ag Compositions and methods for treating pancreatic cancer
DE10230997A1 (en) * 2001-10-26 2003-07-17 Ribopharma Ag Drug to increase the effectiveness of a receptor-mediates apoptosis in drug that triggers tumor cells
DE10202419A1 (en) * 2002-01-22 2003-08-07 Ribopharma Ag Method of inhibiting expression of a target gene resulting from chromosome aberration
US20030166282A1 (en) 2002-02-01 2003-09-04 David Brown High potency siRNAS for reducing the expression of target genes
JP2005527198A (en) * 2002-02-14 2005-09-15 シティ・オブ・ホープ Methods for producing interfering RNA molecules in mammalian cells and therapeutic uses of the interfering RNA molecules
AU2003228301A1 (en) * 2002-03-06 2003-09-22 Rigel Pharmaceuticals, Inc. Novel method for delivery and intracellular synthesis of sirna molecules
US20040248296A1 (en) * 2002-03-20 2004-12-09 Beresford Paul J. HIV therapeutic
US20030180756A1 (en) * 2002-03-21 2003-09-25 Yang Shi Compositions and methods for suppressing eukaryotic gene expression
US20040053876A1 (en) * 2002-03-26 2004-03-18 The Regents Of The University Of Michigan siRNAs and uses therof
WO2003099298A1 (en) 2002-05-24 2003-12-04 Max-Planck Gesellschaft zur Förderung der Wissenschaften e.V. Rna interference mediating small rna molecules
EP1539998A4 (en) 2002-06-05 2007-10-31 Invitrogen Corp Methods and compositions for synthesis of nucleic acid molecules using multiple recognition sites
US20040033602A1 (en) 2002-06-12 2004-02-19 Ambion, Inc. Methods and compositions relating to polypeptides with RNase III domains that mediate RNA interference
ES2550609T3 (en) 2002-07-10 2015-11-11 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. RNA interference by single stranded RNA molecules
US20040241854A1 (en) * 2002-08-05 2004-12-02 Davidson Beverly L. siRNA-mediated gene silencing
PT1527176E (en) 2002-08-05 2007-04-30 Atugen Ag Further novel forms of interfering rna molecules
AU2003261449A1 (en) 2002-08-07 2004-02-25 Compositions for rna interference and methods of use thereof
AU2003273336A1 (en) * 2002-09-18 2004-04-08 Isis Pharmaceuticals, Inc. Efficient reduction of target rna's by single- and double-stranded oligomeric compounds
WO2004029212A2 (en) 2002-09-25 2004-04-08 University Of Massachusetts In vivo gene silencing by chemically modified and stable sirna
AU2003295387A1 (en) 2002-11-05 2004-06-03 Isis Parmaceuticals, Inc. Modified oligonucleotides for use in rna interference
JP4262471B2 (en) * 2002-11-12 2009-05-13 富士通株式会社 Biometric feature data acquisition device
EP1560931B1 (en) 2002-11-14 2011-07-27 Dharmacon, Inc. Functional and hyperfunctional sirna
AU2003295539A1 (en) 2002-11-15 2004-06-15 University Of Massachusetts Allele-targeted rna interference
AU2003298718A1 (en) * 2002-11-22 2004-06-18 University Of Massachusetts Modulation of hiv replication by rna interference
US20040224328A1 (en) * 2003-01-15 2004-11-11 Hans Prydz siRNA screening method
WO2004063375A1 (en) 2003-01-15 2004-07-29 Hans Prydz OPTIMIZING siRNA BY RNAi ANTISENSE
DE602004030315D1 (en) 2003-01-17 2011-01-13 Max Planck Gesellschaft INDUCIBLE SIRNA EXPRESSION CONSTRUCTS FOR TARGET GENERAL SWITCHING
WO2004065600A2 (en) 2003-01-17 2004-08-05 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. Rna interference by palindromic or modified rna molecules
KR20050115231A (en) 2003-02-10 2005-12-07 내셔날 인스티튜트 오브 어드밴스드 인더스트리얼 사이언스 앤드 테크놀로지 Regulation of mammalian cells
CN1750752A (en) * 2003-02-19 2006-03-22 联邦科学和工业研究组织 Efficient gene silencing in plants using short dsRNA sequences
ES2712695T3 (en) 2003-06-02 2019-05-14 Univ Massachusetts Methods and compositions to control the efficiency of RNA silencing
EA201201356A1 (en) 2005-01-07 2013-07-30 Элнилэм Фармасьютикалз, Инк. COMPOSITION FOR THE TREATMENT OF RESPIRATORY SYNCTIAL VIRUSES

Similar Documents

Publication Publication Date Title
US20200270602A1 (en) Rna sequence-specific mediators of rna interference
AU2001249622A1 (en) RNA sequence-specific mediators of RNA interference
EP2360253B1 (en) Methods of producing knockdown cells or organisms by means of RNA sequence-specific mediators of RNA interference and uses thereof.
AU2007214287B2 (en) RNA sequence-specific mediators of RNA interference