AU2007299873A1 - miR-143 regulated genes and pathways as targets for therapeutic intervention - Google Patents

miR-143 regulated genes and pathways as targets for therapeutic intervention Download PDF

Info

Publication number
AU2007299873A1
AU2007299873A1 AU2007299873A AU2007299873A AU2007299873A1 AU 2007299873 A1 AU2007299873 A1 AU 2007299873A1 AU 2007299873 A AU2007299873 A AU 2007299873A AU 2007299873 A AU2007299873 A AU 2007299873A AU 2007299873 A1 AU2007299873 A1 AU 2007299873A1
Authority
AU
Australia
Prior art keywords
protein
isoform
carcinoma
cell
hypothetical protein
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
AU2007299873A
Inventor
Andreas G. Bader
David Brown
Mike Byrom
Charles D. Johnson
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.)
Asuragen Inc
Original Assignee
Asuragen Inc
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 Asuragen Inc filed Critical Asuragen Inc
Publication of AU2007299873A1 publication Critical patent/AU2007299873A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/18Drugs for disorders of the alimentary tract or the digestive system for pancreatic disorders, e.g. pancreatic enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/08Drugs for disorders of the urinary system of the prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/10Drugs for disorders of the urinary system of the bladder
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/10Antimycotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/111General methods applicable to biologically active non-coding nucleic acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2320/00Applications; Uses
    • C12N2320/10Applications; Uses in screening processes
    • C12N2320/11Applications; Uses in screening processes for the determination of target sites, i.e. of active nucleic acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2330/00Production
    • C12N2330/10Production naturally occurring

Description

WO 2008/036718 PCT/US2007/078859 DESCRIPTION MIR-143 REGULATED GENES AND PATHWAYS AS TARGETS FOR THERAPEUTIC INTERVENTION BACKGROUND OF THE INVENTION 5 This application claims the benefit of priority to U.S. Provisional Patent Application Serial No. 60/939,573 filed May 22, 2007 and U.S. Provisional Patent Application Serial No. 60/826,173 filed September 19, 2006, which are hereby incorporated by reference in their entirety. I. FIELD OF THE INVENTION 10 The present invention relates to the fields of molecular biology and medicine. More specifically, the invention relates to methods and compositions for the treatment of diseases or conditions that are affected by miR-143 microRNAs, microRNA expression, and genes and cellular pathways directly and indirectly modulated by such. 15 II. BACKGROUND In 2001, several groups used a cloning method to isolate and identify a large group of "microRNAs" (miRNAs) from C. elegant, Drosophila, and humans (Lagos Quintana et al., 2001; Lau et al., 2001; Lee and Ambros, 2001). Several hundred miRNAs have been identified in plants and animals-including humans-that do not 20 appear to have endogenous siRNAs. Thus, while similar to siRNAs, miRNAs are distinct. miRNAs thus far observed have been approximately 21-22 nucleotides in length, and they arise from longer precursors transcribed from non-protein-encoding genes. See review of Carrington et al. (2003). The precursors form structures that 25 fold back on themselves in self-complementary regions; they are then processed by the nuclease Dicer (in animals) or DCL1 (in plants) to generate the short double stranded miRNA. One of the miRNA strands is incorporated into a complex of proteins and miRNA called the RNA-induced silencing complex (RISC). The miRNA guides the RISC complex to a target mRNA, which is then cleaved or 30 translationally silenced, depending on the degree of sequence complementarity of the miRNA to its target mRNA. Currently, it is believed that perfect or nearly perfect - 1- WO 2008/036718 PCT/US2007/078859 complementarity leads to mRNA degradation, as is most commonly observed in plants. In contrast, imperfect base pairing, as is primarily found in animals, leads to translational silencing. However, recent data suggest additional complexity (Bagga et al., 2005; Lim et al., 2005), and mechanisms of gene silencing by miRNAs remain 5 under intense study. Recent studies have shown that expression levels of numerous miRNAs are associated with various cancers (reviewed in Esquela-Kerscher and Slack, 2006; Calin and Croce, 2006). miRNAs have also been implicated in regulating cell growth and cell and tissue differentiation - cellular processes that are associated with the 10 development of cancer. The inventors previously demonstrated that hsa-miR-143 is involved with the regulation of numerous cell activities that represent intervention points for cancer therapy and for therapy of other diseases and disorders (U.S. Patent Applications serial number 11/141,707 filed May 31, 2005 and serial number 11/273,640 filed 15 November 14, 2005). Upon evaluation of 24 different human tissues, hsa-miR-143 was found to be preferentially expressed in human prostate and colon tissue samples. The inventors observed that hsa-miR-143 expression is lower in many human cancer tumor samples including lung, colon, breast, bladder, and thyroid tumors, than in normal cells from the same patients. Overexpression of hsa-miR-143 in human 20 leukemia cells (Jurkat) increased proliferation of those cells. The inventors also found hsa-miR-143 to be up-regulated in brain tissues of Alzheimer's patients. Other investigators have also observed that miR- 143 is down-regulated in colorectal tumors when compared with matched normal samples (Michael et al., 2003; Akao et al., 2006) and that miR-143 may be involved in the differentiation of human adipocytes 25 (fat storage cells) (Esau et al., 2004). Bioinformatics analyses suggest that any given miRNA may bind to and alter the expression of up to several hundred different genes. In addition, a single gene may be regulated by several miRNAs. Thus, each miRNA may regulate a complex interaction among genes, gene pathways, and gene networks. Mis-regulation or 30 alteration of these regulatory pathways and networks, involving miRNAs, are likely to contribute to the development of disorders and diseases such as cancer. Although bioinformatics tools are helpful in predicting miRNA binding targets, all have -2- WO 2008/036718 PCT/US2007/078859 limitations. Because of the imperfect complementarity with their target binding sites, it is difficult to accurately predict the mRNA targets of miRNAs with bioinformatics tools alone. Furthermore, the complicated interactive regulatory networks among miRNAs and target genes make it difficult to accurately predict which genes will 5 actually be mis-regulated in response to a given miRNA. Correcting gene expression errors by manipulating miRNA expression or by repairing miRNA mis-regulation represent promising methods to repair genetic disorders and cure diseases like cancer. A current, disabling limitation of this approach is that, as mentioned above, the details of the regulatory pathways and 10 networks that are affected by any given miRNA, including miR-143, remain largely unknown. This represents a significant limitation for treatment of cancers in which miR-143 may play a role. A need exists to identify the genes, genetic pathways, and genetic networks that are regulated by or that may regulate hsa-miR-143 expression. SUMMARY OF THE INVENTION 15 The present invention provides additional compositions and methods by identifying genes that are direct targets for miR-143 regulation or that are indirect or downstream targets of regulation following the miR-143-mediated modification of another gene(s) expression. Furthermore, the invention describes gene, disease, and/or physiologic pathways and networks that are influenced by miR-143 and its 20 family members. In certain aspects, compositions of the invention are administered to a subject having, suspected of having, or at risk of developing a metabolic, an immunologic, an infectious, a cardiovascular, a digestive, an endocrine, an ocular, a genitourinary, a blood, a musculoskeletal, a nervous system, a congenital, a respiratory, a skin, or a cancerous disease or condition. 25 In particular aspects, a subject or patient may be selected for treatment based on expression and/or aberrant expression of one or more miRNA or mRNA. In a further aspect, a subject or patient may be selected for treatment based on aberrations in one or more biologic or physiologic pathway(s), including aberrant expression of one or more gene associated with a pathway, or the aberrant expression of one or 30 more protein encoded by one or more gene associated with a pathway. In still a further aspect, a subject or patient may be selected based on aberrations in miRNA expression, or biologic and/or physiologic pathway(s). A subject may be assessed for -3 - WO 2008/036718 PCT/US2007/078859 sensitivity, resistance, and/or efficacy of a therapy or treatment regime based on the evaluation and/or analysis of miRNA or mRNA expression or lack thereof. A subject may be evaluated for amenability to certain therapy prior to, during, or after administration of one or therapy to a subject or patient. Typically, evaluation or 5 assessment may be done by analysis of miRNA and/or mRNA, as well as combination of other assessment methods that include but are not limited to histology, immunohistochemistry, blood work, etc. In some embodiments, an infectious disease or condition includes a bacterial, viral, parasite, or fungal infection. Many of these genes and pathways are associated 10 with various cancers and other diseases. Cancerous conditions include, but are not limited to astrocytoma, acute myelogenous leukemia, breast carcinoma, bladder carcinoma, cervical carcinoma, colorectal carcinoma, endometrial carcinoma, esophageal squamous cell carcinoma, glioma, glioblastoma, gastric carcinoma, hepatocellular carcinoma, Hodgkin lymphoma, leukemia, lipoma, melanoma, mantle 15 cell lymphoma, myxofibrosarcoma, multiple myeloma, neuroblastoma, non-Hodgkin lymphoma, lung carcinoma, non-small cell lung carcinoma, ovarian carcinoma, esophageal carcinoma, osteosarcoma, pancreatic carcinoma, prostate carcinoma, squamous cell carcinoma of the head and neck, thyroid carcinoma, urothelial carcinoma wherein the modulation of one or more gene is sufficient for a therapeutic 20 response. Typically a cancerous condition is an aberrant hyperproliferative condition associated with the uncontrolled growth or inability to undergo cell death, including apoptosis. The present invention provides methods and compositions for identifying genes that are direct targets for miR-143 regulation or that are downstream targets of 25 regulation following the miR-143-mediated modification of upstream gene expression. Furthermore, the invention describes gene pathways and networks that are influenced by miR-143 expression in biological samples. Many of these genes and pathways are associated with various cancers and other diseases. The altered expression or function of miR-143 in cells would lead to changes in the expression of 30 these key genes and contribute to the development of disease. Introducing miR-143 (for diseases where the miRNA is down-regulated) or a miR-143 inhibitor (for diseases where the miRNA is up-regulated) into disease cells or tissues would result -4- WO 2008/036718 PCT/US2007/078859 in a therapeutic response. The identities of key genes that are regulated directly or indirectly by miR-143 and the disease with which they are associated are provided herein. In certain aspects a cell may be an epithelial, stromal, or mucosal cell. The cell can be, but is not limited to brain, a neuronal, a blood, an esophageal, a lung, a 5 cardiovascular, a liver, a breast, a bone, a thyroid, a glandular, an adrenal, a pancreatic, a stomach, a intestinal, a kidney, a bladder, a prostate, a uterus, an ovarian, a testicular, a splenic, a skin, a smooth muscle, a cardiac muscle, or a striated muscle cell. In certain aspects, the cell, tissue, or target may not be defective in miRNA expression yet may still respond therapeutically to expression or over expression of a 10 miRNA. miR-143 could be used as a therapeutic target for any of these diseases. In certain embodiments miR-143 can be used to modulate the activity of miR-143 in a subject, organ, tissue, or cell. A cell, tissue, or subject may be a cancer cell, a cancerous tissue, harbor cancerous tissue, or be a subject or patient diagnosed or at risk of developing a disease 15 or condition. In certain aspects a cancer cell is a neuronal, glial, lung, liver, brain, breast, bladder, blood, leukemic, colon, endometrial, stomach, skin, ovarian, fat, bone, cervical, esophageal, pancreatic, prostate, kidney, or thyroid cell. In still a further aspect cancer includes, but is not limited to astrocytoma, acute myelogenous leukemia, breast carcinoma, bladder carcinoma, cervical carcinoma, colorectal 20 carcinoma, endometrial carcinoma, esophageal squamous cell carcinoma, glioma, glioblastoma, gastric carcinoma, hepatocellular carcinoma, Hodgkin lymphoma, leukemia, lipoma, melanoma, mantle cell lymphoma, myxofibrosarcoma, multiple myeloma, neuroblastoma, non-Hodgkin lymphoma, lung carcinoma, non-small cell lung carcinoma, ovarian carcinoma, esophageal carcinoma, osteosarcoma, pancreatic 25 carcinoma, prostate carcinoma, squamous cell carcinoma of the head and neck, thyroid carcinoma, urothelial carcinoma. Embodiments of the invention include methods of modulating gene expression, or biologic or physiologic pathways in a cell, a tissue, or a subject comprising administering to the cell, tissue, or subject an amount of an isolated 30 nucleic acid or mimetic thereof comprising a miR-143 nucleic acid, mimetic, or inhibitor in an amount sufficient to modulate the expression of a gene positively or negatively modulated by a miR-143 miRNA. A "miR-143 nucleic acid sequence" or -5- WO 2008/036718 PCT/US2007/078859 "miR-143 inhibitor" includes the full length precursor of miR-143, or complement thereof, as well as 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29 or more nucleotides of a precursor miRNA or its processed sequence, or complement thereof, including all ranges and integers there between. In 5 certain embodiments, the miR-143 nucleic acid sequence or miR-143 inhibitor contains the full-length processed miRNA sequence or complement thereof and is referred to as the "miR- 143 full-length processed nucleic acid sequence" or "miR- 143 full-length processed inhibitor sequence." In still further aspects, the miR-143 nucleic acid comprises at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 10 232, 24, 25, 50 nucleotide (including all ranges and integers there between) segment or complementary segment of miR-143 that is at least 75, 80, 85, 90, 95, 98, 99 or 100% identical to SEQ ID NO:l to SEQ ID NO:13. The general term miR-143 includes all members of the miR-143 family that share at least part of a mature miR 143 sequence (UGAGAUGAAGCACUGUAGCUCA (SEQ ID NO:1)) or a 15 complement thereof. A "miR-143 nucleic acid sequence" includes the full length precursor of miR 143 and other family members that include lla-mir-143 (MI0002552) GCGCAGCGCCCUGUC UCCCAGCCUGAGGUGCAGUGCUGCAUCUCUGGUCAGUUGGGAGUCUGA 20 GAUGAAGCACUGUAGCUCAGGAAGAGAGAAGUUGUUCUGCAGC (SEQ ID NO:2); xtr-mir-143 (MI0004937) UGUCUCCCAGCCCAAGGUGCAGUGCUGCAUCUCUGGUCAGUUGUGA GUCUGAGAUGAAGCACUGUAGCUCGGGAAGGGGGAAU (SEQ ID NO:3); dre-mir-143-2 (MI0002008) 25 GAUCUACAGUCGUCUGGCCCGCGGUGCAGUGCUGCAUCUCUG GUCAACUGGGAGUCUGAGAUGAAGCACUGUAGCUCGGGAGGACAACAC UGUCAGCUC (SEQ ID NO:4); rno-mir-143 (MI0000916) GCGGAGCGCCUG UCUCCCAGCCUGAGGUGCAGUGCUGCAUCUCUGGUCAGUUGGGAGUCU GAGAUGAAGCACUGUAGCUCAGGAAGGGAGAAGAUGUUCUGCAGC (SEQ 30 ID NO:5); ptr-mir-143 (MI0002549) GCGCAGCGCCCUGUCUCCCAGCCUGAGGUGCAGUGCUG CAUCUCUGGUCAGUUGGGAGUCUGAGAUGAAGCACUGUAGCUCAGGAA GAGAGAAGUUUUUCUGCAGC (SEQ ID NO:6); ppy-mir-143 (MI0002551) -6- WO 2008/036718 PCT/US2007/078859 GCGCAGC GCCCUGUCUCCCAGCCUGAGGUGCAGUGCUGCAUCUCUGGUCAGUUGGG AGUCUGAGAUGAAGCACUGUAGCUCAGGAAGAGAGAAGUUGUUCUGCA GC (SEQ ID NO:7); ggo-mir-143 (MI0002550) 5 GCGCAGCGCCCUGUCUCCCAGCCUGAGGUGCAGUGCU GCAUCUCUGGUCAGUUGGGAGUCUGAGAUGAAGCACUGUAGCUCAGGA AGAGAGAAGUUGUUCUGCAGC (SEQ ID NO:8); dre-mir-143-1 (MI0002007) GAUCUACAGUCGUCUGGCCCGCGGUGCAGUGCUGCAUCUCUGGUCAACU GGGAGUCUGAGAUGAAGCACUGUAGCUCGGGAGGACAACACUGUCAGC 10 UC (SEQ ID NO:9); hsa-mir-143 (MI0000459) GCGCAGCGCCCUGUCUCCCAGCCUGAGGUGCAGUG CUGCAUCUCUGGUCAGUUGGGAGUCUGAGAUGAAGCACUGUAGCUCAG GAAGAGAGAAGUUGUUCUGCAGC (SEQ ID NO:10); ppa-mir-143 (MI0002553) 15 GCGCAGCGCCCUGUCUCCCAGCCUGAGGUGCAGUGCUGCAUCUCUGGUC AGUUGGGAGUCUGAGAUGAAGCACUGUAGCUCAGGAAGAGAGAAGUUU UUCUGCAGC (SEQ ID NO:11); mdo-mir-143 (MI0005302) CCCGAGGUGCAGUGCUGCAUCUCUGGUC AGUUGUGAGUCUGAGAUGAAGCACUGUAGCUCGGG (SEQ ID NO:12); 20 mmu-mir-143 (MI0000257) CCUGAGGUGCAGUGCUGCAUCUCUGGUCAGUUGGGAGUCUGA GAUGAAGCACUGUAGCUCAGG (SEQ ID NO:13). In certain aspects, a nucleic acid or mimetic of the present invention will comprise 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29 or more nucleotides of the 25 precursor miRNA or its processed sequence, including all ranges and integers there between. In certain embodiments, the miR-143 nucleic acid sequence contains the full-length processed miRNA sequence and is referred to as the "miR-143 full-length processed nucleic acid sequence." In still further aspects, a miR-143 comprises at least one 5,6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 50 30 nucleotide (including all ranges and integers there between) segment of miR-143 that is at least 75, 80, 85, 90, 95, 98, 99 or 100% identical to SEQ ID NOs provided herein. -7- WO 2008/036718 PCT/US2007/078859 In specific embodiments, a miR-143 or miR-143 inhibitor containing nucleic acid is hsa-miR-143 or hsa-miR-143 inhibitor, or a variation thereof. In a further aspect, a miR-143 nucleic acid or miR-143 inhibitor can be administered with 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more miRNAs or miRNA inhibitors. miRNA or its complement 5 can be administer concurrently, in sequence or in an ordered progression. In certain aspects, a miR-143 or miR-143 inhibitor can be administered in combination with one or more of let-7, miR-15a, miR-16, miR-20, miR-21, miR-26a, miR-31, miR-34a, miR-126, miR-145, miR-147, miR-188, miR-200b, miR-200c, miR-215, miR-216, miR-292-3p, and/or miR-331. All or combinations of miRNAs or inhibotrs thereof 10 may be administered in a single formulation. Administration may be before, during or after a second therapy. miR-143 nucleic acids or complement thereof may also include various heterologous nucleic acid sequence, i.e., those sequences not typically found operatively coupled with miR-143 in nature, such as promoters, enhancers, and the 15 like. The miR-143 nucleic acid is a recombinant nucleic acid, and can be a ribonucleic acid or a deoxyribonucleic acid. The recombinant nucleic acid may comprise a miR-143 or miR-143 inhibitor expression cassette, i.e., a nucleic acid segment that expresses a nucleic acid when introduce into an environment containing components for nucleic acid synthesis. In a further aspect, the expression cassette is 20 comprised in a viral, or plasmid DNA vector or other therapeutic nucleic acid vector or delivery vehicle, including liposomes and the like. In certain aspects, viral vectors can be administered at lx102, x103, 1x104 1xl1, 1x1, lx107, x10 8, 1x10 9 , 1x10 1 m, 1x10 1 , 1xlO1 2 , 1xl1', 1xO1 4 pfu or viral particle (vp). In a particular aspect, the miR-143 nucleic acid or miR-143 inhibitor is a 25 synthetic nucleic acid. Moreover, nucleic acids of the invention may be fully or partially synthetic. In still further aspects, a nucleic acid of the invention or a DNA encoding such can be administered at 0.001, 0.01, 0.1, 1, 10, 20, 30, 40, 50, 100, 200, 400, 600, 800, 1000, 2000, to 4000 pg or mg, including all values and ranges there between. In yet a further aspect, nucleic acids of the invention, including synthetic 30 nucleic acid, can be administered at 0.001, 0.01, 0.1, 1, 10, 20, 30, 40, 50, 100, to 200 Vg or mg per kilogram (kg) of body weight. Each of the amounts described herein may be administered over a period of time, including 0.5, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, -8- WO 2008/036718 PCT/US2007/078859 minutes, hours, days, weeks, months or years, including all values and ranges there between. In certain embodiments, administration of the composition(s) can be enteral or parenteral. In certain aspects, enteral administration is oral. In further aspects, 5 parenteral administration is intralesional, intravascular, intracranial, intrapleural, intratumoral, intraperitoneal, intramuscular, intralymphatic, intraglandular, subcutaneous, topical, intrabronchial, intratracheal, intranasal, inhaled, or instilled. Compositions of the invention may be administered regionally or locally and not necessarily directly into a lesion. 10 In certain aspects, the gene or genes modulated comprises 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, 30, 35, 40, 45, 50, 100, 150, 200 or more genes or combinations of genes identified in Tables 1, 3, 4, and/or 5. In still further aspects, the gene or genes modulated may exclude 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, 30, 35, 40, 45, 50, 100, 150, 175 or more genes or combinations of genes 15 identified in Tables 1, 3, 4, and/or 5. Modulation includes modulating transcription, mRNA levels, mRNA translation, and/or protein levels in a cell, tissue, or organ. In certain aspects the expression of a gene or level of a gene product, such as mRNA or encoded protein, is down-regulated or up-regulated. In a particular aspect the gene modulated comprises or is selected from (and may even exclude) 1, 2, 3, 4, 5, 6, 7, 8, 20 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26. 27, 28, or all of the genes identified in Tables 1, 3, 4, and/or 5, or any combinations thereof In certain embodiments a gene modulated or selected to be modulated is from Table 1. In further embodiments a gene modulated or selected to be modulated is from Table 3. In still further embodiments a gene modulated or selected to be modulated is from 25 Table 4. In yet further embodiments a gene modulated or selected to be modulated is from Table 5. Embodiments of the invention may also include obtaining or assessing a gene expression profile or miRNA profile of a target cell prior to selecting the mode of treatment, e.g., administration of a miR-143 nucleic acid, inhibitor of miR-143, or mimetics thereof. The database content related to nucleic acids and genes designated 30 by an accession number or a database submission are incorporated herein by reference as of the filing date of this application. In certain aspects of the invention one or more miRNA or miRNA inhibitor may modulate a single gene. In a further aspect, one or -9- WO 2008/036718 PCT/US2007/078859 more genes in one or more genetic, cellular, or physiologic pathways can be modulated by one or more miRNAs or complements thereof, including miR-143 nucleic acids and miR-143 inhibitors in combination with other miRNAs. miR-143 nucleic acids may also include various heterologous nucleic acid 5 sequence, i.e., those sequences not typically found operatively coupled with miR-143 in nature, such as promoters, enhancers, and the like. The miR-143 nucleic acid is a recombinant nucleic acid, and can be a ribonucleic acid or a deoxyribonucleic acid. The recombinant nucleic acid may comprise a miR-143 expression cassette. In a further aspect, the expression cassette is comprised in a viral, or plasmid DNA vector 10 or other therapeutic nucleic acid vector or delivery vehicle, including liposomes and the like. In a particular aspect, the miR-143 nucleic acid is a synthetic nucleic acid. Moreover, nucleic acids of the invention may be fully or partially synthetic. 15 Table 1. Genes with increased (positive values) or decreased (negative values) expression following transfection of human cancer cells with pre-miR hsa-miR-143. Gene RefSeq Transcript ID A log2 Symbol AKAP12 NM 005100 /// NM 144497 0.725245496 ANKRD46 NM 198401 0.791492237 ANXA6 NM 001155 ///NM 004033 0.727214714 ARL2BP NM 012106 0.800772424 ASNA1 NM 004317 -1.07942093 ATP6V1A NM 001690 -1.126127932 ATXN1 NM 000332 0.850968582 AXL NM 001699///NM 021913 1.156039698 BCL2L1 NM 001191 ///NM 138578 -0.821265359 CCND1 NM 053056 -0.938024465 CCNG1 NM 004060 //NM 199246 0.862627632 CLIC4 NM 013943 0.825614765 CXCL1 NM 001511 0.938115811 CXCL2 NM 002089 0.706326327 DAZAP2 NM 014764 -0.916764957 DCP2 NM 152624 0.797770229 DDAH1 NM 012137 0.765730627 DDX3Y NM 004660 0.848651105 DICERI NM 030621 //NM 177438 0.929848609 DSC2 NM 004949 /// NM 024422 0.902830281 FLJ13910 NM 022780 0.866839654 GALC NM 000153 -1.161432175 GATM NM 001482 -1.970548228 -10- WO 2008/036718 PCT/US2007/078859 GOLPH2 NM 016548//NM 177937 -1.126884613 GREBI NM 014668 /// NM 033090 /// NM_148903 0.755673527 GREMI NM 013372 1.051739161 HIPK2 NM 022740 -0.904313564 HIPK3 NM 005734 0.826433357 IFIHI NM 022168 0.706653845 IGFBP3 NM 000598 ///NM 001013398 -0.809607512 IL32 NM 001012631 ///NM 001012632/// 0.757126883 NM 001012633 /// NM001012634 // NM 001012635 IL6ST NM 002184///NM 175767 0.751854493 IL8 NM 000584 1.104016175 INSIGI NM 005542///NM 198336///NM 198337 0.875027481 LEPR NM 001003679 /// NM 001003680 /// 0.797930372 NM 002303 LMO4 NM 006769 -1.012706499 LOC137886 XM 059929 -0.752855433 MCLI NM 021960 ///NM 182763 0.761759353 MGC5618 --- 0.797855581 MTUS1 NM 001001924 //NM 001001925/// 0.70655 NM 001001927 ///NM001001931 /I NM 020749 NIDI NM 002508 1.090976167 NT5E NM 002526 0.878049429 PDCD2 NM 002598 /// NM 144781 -0.723484401 PDCD4 NM 014456 ///NM 145341 0.728228239 PDK4 NM 002612 0.961974975 PELI1 NM 020651 0.768582445 PMCH NM 002674 0.790936704 PROSC NM 007198 -1.645677869 PTPN12 NM 002835 0.769808986 RABI1FIP1 NM 001002233 /// NM001002814 /// -0.83733308 NM 025151 RAB2 NM 002865 0.827382805 RBL1 NM 002895 /// NM 183404 -1.302328709 RDX NM 002906 0.760806942 RECK NM 021111 1.103484746 RHEB NM 005614 0.825468322 RHOB NM 004040 0.921813933 RHOBTB1 NM 001032380///NM 014836//NM_198225 0.744478582 RP2 NM 006915 0.822851399 SERPINEl NM 000602 -0.856846452 SLC11A2 NM 000617 0.716682705 SLC30A1 NM 021194 -0.841163945 SLC35BI NM 005827 -1.07644709 TAF1O NM 006284 -1.695883532 TBC1D2 NM 018421 -0.746279363 TGFBR2 NM 001024847///NM 003242 0.854509353 -11- WO 2008/036718 PCT/US2007/078859 TMEM45A NM 018004 -0.748492283 TMF1 NM 007114 -0.939693594 TNC NM 002160 0.86901183 TNRC9 XM 049037 0.740367787 TRAI NM 003299 0.875188144 TTMP NM 024616 0.844059608 TXN NM 003329 0.92541735 UGT1A8 /// NM 019076 //NM 021027 -0.961897449 UGT1A9 WASPIP NM 003387 1.04160055 WDR50 NM 016001 -1.049152791 WEE1 NM 003390 0.722369746 A further embodiment of the invention is directed to methods of modulating a cellular pathway comprising administering to the cell an amount of an isolated nucleic acid comprising a miR-143 nucleic acid sequence or a miR-143 inhibitor. A cell, 5 tissue, or subject may be a cancer cell, a cancerous tissue or harbor cancerous tissue, or a cancer patient. The database content related to all nucleic acids and genes designated by an accession number or a database submission are incorporated herein by reference as of the filing date of this application. A further embodiment of the invention is directed to methods of modulating a 10 cellular pathway comprising administering to the cell an amount of an isolated nucleic acid comprising a miR-143 nucleic acid sequence in an amount sufficient to modulate the expression, function, status, or state of a cellular pathway, in particular those pathways described in Table 2 or the pathways known to include one or more genes from Table 1, 3, 4, and/or 5. Modulation of a cellular pathway includes, but is not 15 limited to modulating the expression of one or more gene(s). Modulation of a gene can include inhibiting the function of an endogenous miRNA or providing a functional miRNA to a cell, tissue, or subject. Modulation refers to the expression levels or activities of a gene or its related gene product (e.g., mRNA) or protein, e.g., the mRNA levels may be modulated or the translation of an mRNA may be 20 modulated. Modulation may increase or up regulate a gene or gene product or it may decrease or down regulate a gene or gene product (e.g., protein levels or activity). Still a further embodiment includes methods of administering an miRNA or mimic thereof, and/or treating a subject or patient having, suspected of having, or at risk of developing a pathological condition comprising one or more of step (a) - 12- WO 2008/036718 PCT/US2007/078859 administering to a patient or subject an amount of an isolated nucleic acid comprising a miR-143 nucleic acid sequence or a miR-143 inhibitor in an amount sufficient to modulate expression of a cellular pathway; and (b) administering a second therapy, wherein the modulation of the cellular pathway sensitizes the patient or subject, or 5 increases the efficacy of a second therapy. An increase in efficacy can include a reduction in toxicity, a reduced dosage or duration of the second therapy, or an additive or synergistic effect. A cellular pathway may include, but is not limited to one or more pathway described in Table 2 below or a pathway that is know to include one or more genes of Tables 1, 3, 4, and/or 5. The second therapy may be 10 administered before, during, and/or after the isolated nucleic acid or miRNA or inhibitor is administered A second therapy can include administration of a second miRNA or therapeutic nucleic acid such as a siRNA or antisense oligonucleotide, or may include various standard therapies, such as pharmaceuticals, chemotherapy, radiation therapy, 15 drug therapy, immunotherapy, and the like. Embodiments of the invention may also include the determination or assessment of gene expression or gene expression profile for the selection of an appropriate therapy. In a particular aspect, a second therapy is a chemotherapy. A chemotherapy can include, but is not limited to paclitaxel, cisplatin, carboplatin, doxorubicin, oxaliplatin, larotaxel, taxol, lapatinib, docetaxel, 20 methotrexate, capecitabine, vinorelbine, cyclophosphamide, gemcitabine, amrubicin, cytarabine, etoposide, camptothecin, dexamethasone, dasatinib, tipifarnib, bevacizumab, sirolimus, temsirolimus, everolimus, lonafarnib, cetuximab, erlotinib, gefitinib, imatinib mesylate, rituximab, trastuzumab, nocodazole, sorafenib, sunitinib, bortezomib, alemtuzumab, gemtuzumab, tositumomab or ibritumomab. 25 Embodiments of the invention include methods of treating a subject with a disease or condition comprising one or more of the steps of (a) determining an expression profile of one or more genes selected from Table 1, 3, 4, and/or 5; (b) assessing the sensitivity of the subject to therapy based on the expression profile; (c) selecting a therapy based on the assessed sensitivity; and (d) treating the subject using 30 a selected therapy. Typically, the disease or condition will have as a component, indicator, or resulting mis-regulation of one or more gene of Table 1, 3, 4, and/or 5. - 13 - WO 2008/036718 PCT/US2007/078859 In certain aspects, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more miRNA may be used in sequence or in combination. For instance, any combination of miR-143 or a miR-143 inhibitor with another miRNA Further embodiments include the identification and assessment of an expression profile indicative of miR-143 status in a cell or tissue 5 comprising expression assessment of one or more gene from Table 1, 3, 4, and/or 5, or any combination thereof. The term "miRNA" is used according to its ordinary and plain meaning and refers to a microRNA molecule found in eukaryotes that is involved in RNA-based gene regulation. See, e.g., Carrington et al., 2003, which is hereby incorporated by 10 reference. The term can be used to refer to the single-stranded RNA molecule processed from a precursor or in certain instances the precursor itself. In some embodiments, it may be useful to know whether a cell expresses a particular miRNA endogenously or whether such expression is affected under particular conditions or when it is in a particular disease state. Thus, in some 15 embodiments of the invention, methods include assaying a cell or a sample containing a cell for the presence of one or more marker gene or mRNA or other analyte indicative of the expression level of a gene of interest. Consequently, in some embodiments, methods include a step of generating an RNA profile for a sample. The term "RNA profile" or "gene expression profile" refers to a set of data regarding the 20 expression pattern for one or more gene or genetic marker in the sample (e.g., a plurality of nucleic acid probes that identify one or more markers from Tables 1, 3, 4, and/or 5); it is contemplated that the nucleic acid profile can be obtained using a set of RNAs, using for example nucleic acid amplification or hybridization techniques well know to one of ordinary skill in the art. The difference in the expression profile in the 25 sample from the patient and a reference expression profile, such as an expression profile from a normal or non-pathologic sample, is indicative of a pathologic, disease, or cancerous condition. A nucleic acid or probe set comprising or inhibitor can be selected based on observing two given miRNAs share a set of target genes or pathways listed in Tables 1, 2, 4 and/or 5 that are altered in a particular disease or 30 condition. These two miRNAs may result in an improved therapy (e.g., reduced toxicity, greater efficacy, prolong remission, or other improvements in a subjects condition), result in an increased efficacy, an additive efficacy, or a synergistic - 14- WO 2008/036718 PCT/US2007/078859 efficacy providing an additional or an improved therapeutic response. Without being bound by any particular theorty, synergy of two miRNA can be a consequence of regulating the same genes or related genes (related by a common pathway or biologic end result) more effectively (e.g., due to distinct binding sites on the same target or 5 related target(s)) and/or a consequence of regulating different genes, but all of which have been implicated in a disease or condition. In certain aspects, miR-143 or a miR-143 inhibitor and let-7 can be administered to patients with acute myeloid leukemia, breast carcinoma, bladder carcinoma, cervical carcinoma, colorectal carcinoma, endometrial carcinoma, glioma, 10 glioblastoma, gastric carcinoma, hepatocellular carcinoma, Hodgkin lymphoma, leukemia, melanoma, myxofibrosarcoma, multiple myeloma, neuroblastoma, non Hodgkin lymphoma, non-small cell lung carcinoma, ovarian carcinoma, esophageal carcinoma, pancreatic carcinoma, prostate carcinoma, squamous cell carcinoma of the head and neck, thyroid carcinoma, or urothelial carcinoma. 15 Further aspects include administering miR-143 or a miR-143 inhibitor and miR-15 to patients with astrocytoma, acute myeloid leukemia, breast carcinoma, bladder carcinoma, cervical carcinoma, colorectal carcinoma, endometrial carcinoma, glioma, glioblastoma, gastric carcinoma, hepatocellular carcinoma, Hodgkin lymphoma, melanoma, mantle cell lymphoma, myxofibrosarcoma, multiple myeloma, 20 neuroblastoma, non-Hodgkin lymphoma, non-small cell lung carcinoma, ovarian carcinoma, esophageal carcinoma, osteosarcoma, pancreatic carcinoma, prostate carcinoma, squamous cell carcinoma of the head and neck, or thyroid carcinoma. In still further aspects, miR-143 or a miR-143 inhibitor and miR-16 are administered to patients with astrocytoma, breast carcinoma, bladder carcinoma, 25 colorectal carcinoma, endometrial carcinoma, glioblastoma, gastric carcinoma, hepatocellular carcinoma, Hodgkin lymphoma, melanoma, mantle cell lymphoma, myxofibrosarcoma, multiple myeloma, non-small cell lung carcinoma, ovarian carcinoma, esophageal carcinoma, pancreatic carcinoma, prostate carcinoma, squamous cell carcinoma of the head and neck, or thyroid carcinoma. 30 Aspects of the invention include methods where miR-143 or a miR-143 inhibitor and miR-20 are administered to patients with astrocytoma, acute myeloid - 15 - WO 2008/036718 PCT/US2007/078859 leukemia, breast carcinoma, bladder carcinoma, colorectal carcinoma, endometrial carcinoma, glioma, glioblastoma, gastric carcinoma, hepatocellular carcinoma, melanoma, mantle cell lymphoma, neuroblastoma, non-small cell lung carcinoma, ovarian carcinoma, esophageal carcinoma, pancreatic carcinoma, prostate carcinoma, 5 or squamous cell carcinoma of the head and neck. In a further aspect, miR-143 or a miR-143 inhibitor and miR-21 are administered to patients with astrocytoma, acute mycloid leukemia, breast carcinoma, bladder carcinoma, cervical carcinoma, colorectal carcinoma, endometrial carcinoma, glioma, glioblastoma, gastric carcinoma, hepatocellular carcinoma, Hodgkin 10 lymphoma, leukemia, melanoma, mantle cell lymphoma, multiple myeloma, non Hodgkin lymphoma, non-small cell lung carcinoma, ovarian carcinoma, esophageal carcinoma, osteosarcoma, pancreatic carcinoma, prostate carcinoma, squamous cell carcinoma of the head and neck, or thyroid carcinoma. In still further aspects, miR-143 or a miR-143 inhibitor and miR-26a are 15 administered to patients with acute myeloid leukemia, breast carcinoma, bladder carcinoma, cervical carcinoma, colorectal carcinoma, glioma, glioblastoma, gastric carcinoma, hepatocellular carcinoma, leukemia, melanoma, multiple myeloma, neuroblastoma, non-Hodgkin lymphoma, non-small cell lung carcinoma, ovarian carcinoma, esophageal carcinoma, osteosarcoma, pancreatic carcinoma, or prostate 20 carcinoma. In yet further aspects, miR-143 or a miR-143 inhibitor and miR-34a are administered to patients with astrocytoma, acute myeloid leukemia, breast carcinoma, bladder carcinoma, cervical carcinoma, colorectal carcinoma, endometrial carcinoma, glioma, glioblastoma, gastric carcinoma, hepatocellular carcinoma, Hodgkin 25 lymphoma, leukemia, melanoma, mantle cell lymphoma, multiple myeloma, non Hodgkin lymphoma, non-small cell lung carcinoma, ovarian carcinoma, esophageal carcinoma, osteosarcoma, pancreatic carcinoma, prostate carcinoma, squamous cell carcinoma of the head and neck, thyroid carcinoma, or urothelial carcinoma. In certain aspects, miR-143 or a miR-143 inhibitor and miR-126 are 30 administered to patients with astrocytoma, acute mycloid leukemia, breast carcinoma, bladder carcinoma, cervical carcinoma, colorectal carcinoma, endometrial carcinoma, - 16- WO 2008/036718 PCT/US2007/078859 glioma, glioblastoma, gastric carcinoma, hepatocellular carcinoma, Hodgkin lymphoma, leukemia, melanoma, mantle cell lymphoma, non-Hodgkin lymphoma, non-small cell lung carcinoma, ovarian carcinoma, esophageal carcinoma, osteosarcoma, pancreatic carcinoma, prostate carcinoma, squamous cell carcinoma of 5 the head and neck, or thyroid carcinoma. In still a further aspect, miR-143 or a miR-143 inhibitor and miR-147 are administered to patients with astrocytoma, breast carcinoma, bladder carcinoma, cervical carcinoma, colorectal carcinoma, endometrial carcinoma, esophageal squamous cell carcinoma, glioma, glioblastoma, gastric carcinoma, hepatocellular 10 carcinoma, Hodgkin lymphoma, leukemia, lipoma, melanoma, mantle cell lymphoma, myxofibrosarcoma, multiple myeloma, non-Hodgkin lymphoma, non-small cell lung carcinoma, ovarian carcinoma, esophageal carcinoma, osteosarcoma, pancreatic carcinoma, prostate carcinoma, squamous cell carcinoma of the head and neck, or thyroid carcinoma. 15 In yet another aspect, miR-143 or a miR-143 inhibitor and miR-188 are administered to patients with astrocytoma, acute myeloid leukemia, breast carcinoma, bladder carcinoma, cervical carcinoma, colorectal carcinoma, endometrial carcinoma, esophageal squamous cell carcinoma, glioma, glioblastoma, gastric carcinoma, hepatocellular carcinoma, leukemia, melanoma, multiple myeloma, non-Hodgkin 20 lymphoma, non-small cell lung carcinoma, ovarian carcinoma, esophageal carcinoma, pancreatic carcinoma, prostate carcinoma, squamous cell carcinoma of the head and neck, or thyroid carcinoma. In other aspects, miR-143 or a miR-143 inhibitor and miR-215 are administered to patients with astrocytoma, acute myeloid leukemia, breast carcinoma, 25 bladder carcinoma, cervical carcinoma, colorectal carcinoma, endometrial carcinoma, esophageal squamous cell carcinoma, glioma, glioblastoma, gastric carcinoma, hepatocellular carcinoma, Hodgkin lymphoma, leukemia, lipoma, melanoma, mantle cell lymphoma, myxofibrosarcoma, multiple myeloma, neuroblastoma, non-Hodgkin lymphoma, non-small cell lung carcinoma, ovarian carcinoma, esophageal carcinoma, 30 osteosarcoma, pancreatic carcinoma, prostate carcinoma, squamous cell carcinoma of the head and neck, thyroid carcinoma, or urothelial carcinoma. - 17- WO 2008/036718 PCT/US2007/078859 In certain aspects, miR-143 or a miR-143 inhibitor and miR-216 are administered to patients with astrocytoma, breast carcinoma, cervical carcinoma, colorectal carcinoma, endometrial carcinoma, glioma, glioblastoma, gastric carcinoma, hepatocellular carcinoma, Hodgkin lymphoma, leukemia, non-Hodgkin 5 lymphoma, non-small cell lung carcinoma, ovarian carcinoma, esophageal carcinoma, osteosarcoma, prostate carcinoma, or squamous cell carcinoma of the head and neck. In a further aspect, miR-143 or a miR-143 inhibitor and miR-292-3p are administered to patients with astrocytoma, acute myeloid leukemia, breast carcinoma, bladder carcinoma, cervical carcinoma, colorectal carcinoma, endometrial carcinoma, 10 glioma, glioblastoma, gastric carcinoma, hepatocellular carcinoma, leukemia, lipoma, melanoma, myxofibrosarcoma, multiple myeloma, neuroblastoma, non-Hodgkin lymphoma, non-small cell lung carcinoma, ovarian carcinoma, esophageal carcinoma, osteosarcoma, pancreatic carcinoma, prostate carcinoma, squamous cell carcinoma of the head and neck, thyroid carcinoma, or urothelial carcinoma. 15 In still a further aspect, miR-143 or a miR-143 inhibitor and miR-331 are administered to patients with astrocytoma, acute myeloid leukemia, breast carcinoma, bladder carcinoma, cervical carcinoma, colorectal carcinoma, endometrial carcinoma, glioma, glioblastoma, gastric carcinoma, hepatocellular carcinoma, leukemia, melanoma, myxofibrosarcoma, multiple myeloma, neuroblastoma, non-Hodgkin 20 lymphoma, ovarian carcinoma, esophageal carcinoma, osteosarcoma, pancreatic carcinoma, prostate carcinoma, squamous cell carcinoma of the head and neck, or thyroid carcinoma. In yet a further aspect, miR-143 or a miR-143 inhibitor and miR-200b/c are administered to patients with breast carcinoma, cervical carcinoma, colorectal 25 carcinoma, glioma, glioblastoma, gastric carcinoma, hepatocellular carcinoma, leukemia, lipoma, multiple myeloma, non-small cell lung carcinoma, ovarian carcinoma, esophageal carcinoma, osteosarcoma, pancreatic carcinoma, prostate carcinoma, squamous cell carcinoma of the head and neck, or thyroid carcinoma. It is contemplated that when miR-143 or a miR-143 inhibitor is given in 30 combination with one or more other miRNA molecules, the two different miRNAs or inhibitors may be given at the same time or sequentially. In some embodiments, -18- WO 2008/036718 PCT/US2007/078859 therapy proceeds with one miRNA or inhibitor and that therapy is followed up with therapy with the other miRNA or inhibitor 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55 minutes, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24 hours, 1, 2, 3, 4, 5, 6, 7 days, 1, 2, 3, 4, 5 weeks, or 1, 2, 3, 4, 5, 5 6, 7, 8, 9, 10, 11, or 12 months or any such combination later. Further embodiments include the identification and assessment of an expression profile indicative of miR-143 status in a cell or tissue comprising expression assessment of one or more gene from Table 1, 3, 4, and/or 5, or any combination thereof. 10 The term "miRNA" is used according to its ordinary and plain meaning and refers to a microRNA molecule found in eukaryotes that is involved in RNA-based gene regulation. See, e.g., Carrington et al., 2003, which is hereby incorporated by reference. The term can be used to refer to the single-stranded RNA molecule processed from a precursor or in certain instances the precursor itself or a mimetic 15 thereof. In some embodiments, it may be useful to know whether a cell expresses a particular miRNA endogenously or whether such expression is affected under particular conditions or when it is in a particular disease state. Thus, in some embodiments of the invention, methods include assaying a cell or a sample containing 20 a cell for the presence of one or more miRNA marker gene or mRNA or other analyte indicative of the expression level of a gene of interest. Consequently, in some embodiments, methods include a step of generating an RNA profile for a sample. The term "RNA profile" or "gene expression profile" refers to a set of data regarding the expression pattern for one or more gene or genetic marker in the sample (e.g., a 25 plurality of nucleic acid probes that identify one or more markers or genes from Tables 1, 3, 4, and/or 5); it is contemplated that the nucleic acid profile can be obtained using a set of RNAs, using for example nucleic acid amplification or hybridization techniques well know to one of ordinary skill in the art. The difference in the expression profile in the sample from a patient and a reference expression 30 profile, such as an expression profile from a normal or non-pathologic sample, or a digitized reference, is indicative of a pathologic, disease, or cancerous condition. In certain aspects the expression profile is an indicator of a propensity to or probability - 19- WO 2008/036718 PCT/US2007/078859 of (i.e., risk factor for a disease or condition) developing such a condition(s). Such a risk or propensity may indicate a treatment, increased monitoring, prophylactic measures, and the like. A nucleic acid or probe set may comprise or identify a segment of a corresponding mRNA and may include all or part of 1, 2, 3, 4, 5, 6, 7, 8, 5 9, 10, 11, 12 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 100, 200, 500, or more segments, including any integer or range derivable there between, of a gene or genetic marker, or a nucleic acid, mRNA or a probe representative thereof that is listed in Tables 1, 3, 4, and/or 5 or 10 identified by the methods described herein. Certain embodiments of the invention are directed to compositions and methods for assessing, prognosing, or treating a pathological condition in a patient comprising measuring or determining an expression profile of one or more miRNA or marker(s) in a sample from the patient, wherein a difference in the expression profile 15 in the sample from the patient and an expression profile of a normal sample or reference expression profile is indicative of pathological condition and particularly cancer (e.g., In certain aspects of the invention, the miRNAs, cellular pathway, gene, or genetic marker is or is representative of one or more pathway or marker described in Table 1, 2, 3, 4, and/or 5, including any combination thereof 20 Aspects of the invention include diagnosing, assessing, or treating a pathologic condition or preventing a pathologic condition from manifesting. For example, the methods can be used to screen for a pathological condition; assess prognosis of a pathological condition; stage a pathological condition; assess response of a pathological condition to therapy; or to modulate the expression of a gene, genes, or 25 related pathway as a first therapy or to render a subject sensitive or more responsive to a second therapy. In particular aspects, assessing the pathological condition of the patient can be assessing prognosis of the patient. Prognosis may include, but is not limited to an estimation of the time or expected time of survival, assessment of response to a therapy, and the like. In certain aspects, the altered expression of one or 30 more gene or marker is prognostic for a patient having a pathologic condition, wherein the marker is one or more of Table 1, 3, 4, and/or 5, including any combination thereof. - 20 - WO 2008/036718 PCT/US2007/078859 Table 2. Significantly affected functional cellular pathways following hsa-miR-143 over-expression in human cancer cells. Number of Genes Pathway Functions Cellular Movement, Hematological System Development and 9 Function, Immune Response Gene Expression, Cellular Growth and Proliferation, Developmental 2 Disorder Table 3. Predicted target genes of hsa-miR-143 for Ref Seq ID reference - Pruitt et 5 al, 2005. Gene RefSeq Symbol Transcript ID Description 76P NM 014444 gamma tubulin ring complex protein (76p gene) AACS NM 023928 acetoacetyl-CoA sthetase AADACL1 NM_020792 arylacetamide deacetylase-like 1 AARSL NM_020745 alanyl-tRNA synthetase like ABAT NM_000663 4-aminobutyate aminotransferase precursor ABCA1 NM 005502 ATP-binding cassette, sub-family A member 1 ABCB 11 NM_003742 ATP-binding cassette, sub-family 3 (MDR/TAP), ABCB9 NM_203445 ATP-binding cassette, sub-family B (MDRITAP), ABCC1 NM _004996 ATP-binding cassette, sub-family C, member 1 ABCC13 NM _172024 ATP-binding cassette protein C13 isoforn b ABCC3 NM 020038 ATP-binding cassette, sub-family C, member 3 ABCC4 NM 005845 ATP-binding cassette, sub-family C, member 4 ABCG4 NM_022169 ATP-hinding cassette, subfamily G, member 4 ABCG5 NM_022436 sterolin 1 ABHD14A NM_015407 abhydrolase domain containing 14A ABHD14B NM 032750 abhydrolase domain containing 14B ABHD8 NM 024527 mtalase domain 10 ABLIMI NM_001003407 A d Met oei I isoform 1 ABR NM 001092 aAM eapoptidse doma so ABTB32 NM 145804 akrnrpa n T PZ oancnann ACACB NM001093 aey-onyeAcroyaebt ACADSBi NM 001609 agl o-m dyr oa.,sotbace ACCN1 NM 001094 metoeptis ih th roa ACE NM 152831 ACE2 NM 021804 nitniIcovrigezm2peusr ACIN1 NM 014977 aottccrmtncnesto nue ACOXL NM 018308 ay-onyeAoiaelk ACP1 NM_004300 ai hshts sfr AC-SL6 NM 001009185 ay-o yteaeln-hi aiymme ACTL8 NM 030812 atnlk rti ACTN2 NM 001103 atnn lh ACTR8 NM 022899 ci-eadprtn8 ACVR1BN 004302 ciiAtye13rcpo sfrapeusr AC-YL2 NM 001010853 hpteia rti O159 ADAM10 NM 001110 DMmtloetds oan1 ADAM12 NM 003474ADMmtloetdsdoan1 ofr1 ADAM9 |NM_001005845 AA ealppiaedmi sfr ADAMTS1 NM 006988 AyA etad ectylase-lie 1hobsodntp aa-tRAsytets-lk WO 2008/036718 PCT/US2007/078859 ADAMTS3 NM 014243 ADAM metallopeptidase with thrombosponidin type 1 ADAMTS4 _NM 005099 ADAM metallopeptidase with thrombospondin type 1 ADAMTSL1 NM 052866 ADAMTS-like 1 isoform 2 ADAR NM 001025107 adenosine deaminase, RNA-specific isoform d ADARB1 NM 001033049 RNA-specific adenosine deaminase B1I isoform 4 ADAT1 NM 012091 adenosine deaminase, tRNA-specific I ADCY1 |NM_ 021116 brain adenylate cyclase 1 ADCY2 NM 020546 adenylate cyclase 2 ADCY6 NM 015270 adenylate cyclase 6 isoform a ADCY9 NM-00 1116 adenylate cyclase 9 ADD2 NM 001617 adducin 2 isoform a ADD3 NM 001121 adducin 3 (gamma) isoform b AD 1l NM 018269 membrane-type I matrix metalloproteinase AIlPOQ NM 004797 adiponectin precursor ADORA3 NM 000677 adenosine A3 receptor isoform 2 ADRA2B3 NM 000682 alpha-2B3-adrenergic receptor ADSSL1 NM 152328 adenylosuccinate synthase-like 1 isoformn 2 AFAP |NM_021638 actin filament associated protein AdF 1 'NM 005935 myeloid/lymphoid or mixed-lineage leukemia AFF2 NM 002025 fragile X mental retardation 2 AFG3 L2 NM 006796 AFG WATPase family gene 3-like 2 AGBL4 NM032785 -hypothetical protein LOC84871 AGMAT NM 024758 agmatine ureohydrolase (agmatinase) AGPAT1 NM 006411 1 -acylglycerol-3 -phosphate O-acyltransferase 1 AGPAT3 |NM_020132 1 -acylglycerol-3 -phosphate O-acyltransferase 3 AGPAT4 ENM 001012733 1 -acylglycerol-3-phosphate O-acyltransferase 4 AGR2 NM 006408 anterior gradient 2 homolog AGRN NM_198576 agrm AHC-TF1 NM 015446 transcription factor ELYS AHCYL1 NM 006621 S-adenosylhomocysteine hydrolase-like 1 AIC-DA NM 020661 activation-induced cytidine deaminase AIF1 NM 004847 allograft inflammatory factor 1 isoform 2 AIG1 1NM 016108 androgen-induced 1 AIP-L NM 001033054 -aryl -hydrocarbon receptor interacting AIRE NM 000383 autoimmune regulator AIRE isoformn I A 1 NM 000476 _adenylate kinase I AK3 NM 016282 adenylate kinase 3 AKAP11I NM 144490 A-kinase anchor protein 11I isoform 2 AKJAP 13 NM 006738 A-linase anchor protein 13 isoformn 1 AK AP 6 NM 004274 A-kinase anchor protein 6 AKT1 NM 001014431 v-akt murine thymoma viral oncogene homolog 1 ALB NM 000477 albumin precursor ALDH3A2 NM 000382 aldehyde dehydrogenase 3A2 isoform 2 ALDH5A1 NM 001080 aldehyde dehydrogenase 5A1 precursor, isoform 2 A KBH4 NM 017621 -hypothetical protein LOC54784 AKLP-L NM 000478 -tissue non-specific alkaline phosphatase A LS2 NM 020919 -alsin ALX3 NM 006492 aristaless-like homeobox 3 AMDHD1 NM 152435 hypothetical protein LOC 144193 AM, FR NM 001144 autocrine motility factor receptor AMICAl NM 153206 adhesion molecule, interacts with CXADR antigen ASMECR1 NM 001025580 AMM -ECR1 protein isoform 2 AMOTL1 NM 130847 angiomotin like 1 AMPD2 NM 004037 adenosine monophosphate deaminase 2 (isoform L) - 22 - WO 2008/036718 PCT/US2007/078859 AMT NM_000481 aminomethyltransferase (glycine cleavage system AMZ1 NM 133463 archaemetzincin-I ANGEL1 NM 015305 angel homolog I ANGPTL1 NM 004673 angiopoietin-like 1 precursor ANGPTL2 NM 012098 angiopoictin-like 2 precursor ANGPTL7 NM 021146 angiopoietin-like 7 ANKH NM 054027 ankylosis, progressive homolog ANKRD12 NM 015208 ankyrinrepeat domain 12 ANKRD13 NM 033121 ankyrinrepeat domain 13 ANKRD20A3 NM 001012419 hypothetical protein L0C441425 ANKRD25 NM 015493 ankyrin repeat domain 25 ANKRD28 NM 015199 ankyrin repeat domain 28 ANKRD29 NM 173505 ankyrin repeat domain 29 ANKRD41 NM_152363 ankyrinrepeatdomain41 ANKRD50 NM 020337 ankyrin repeat domain 50 ANKS6 NM 173551 sterile alpha motif domain containing 6 ANXA3 NM 005139 annexin A3 ANXA9 NM 003568 amiexin A9 AOC2 NM 001158 amine oxidase, copper containing 2 isoform a AP2B1 NM_001030006 adaptor-related protein complex 2, beta 1 AP3D1 NM 003938 adaptor-related protein complex 3, delta 1 AP3M1 NM_012095 adaptor-related protein complex 3, mu 1 subunit APAFI NM 001160 apoptotic protease activating factor isoform b APOAlBP NM 144772 apolipoprotein A-I binding protein precursor APOA5 NM 052968 apolipoprotein AV APOBEC3A NM 145699 plorbolin 1 APOBEC3F NM_145298 apolipoprotein B nRNA editing enzyme, catalytic APOBEC4 NM 203454 apolipoprotein B mRNA editing enzyme, catalytic APOL1 NM 003661 apolipoprotein LI isoform a precursor APOL6 NM 030641 apolipoprotein L6 APOLD1 NM 030817 apolipoprotein L domain containing 1 APPL NM 012096 adaptor protein containing pH domain, PTB domain APTX NM_175069 aprataxin isoform b AQP1O NM_080429 aquaporin10 AQP2 NM_000486 aquaporin 2 AQP3 NM_004925 aquaporin3 ARCN1 NM 001655 archain ARFGAP3 NM 014570 ADP-ribosylation factor GTPase activating AR IP2 NM_012402 ADP-ribosylation fsetor interacting protein 2 ARHGAP18 NM 033515 Rho GTPase activating protein 18 ARHGAP20 NM 020809 R GTPase activating protein 20 ARHGAP25 NM 001007231 Rho GTPaae activating protein 25 isoform a ARHGAP26 NM 015071 GTPase regulator associated with the focal ARHGAP28 NM 001010000 Rho GiPase activating protein 28 isoform a ARHGAP9 NM_032496 Rho GTPaae activating protein 9 ARHGDIB NM 001175 Rho GDP dissociation inhibitor (GDI) beta ARHGEF1 NM 004706 Rho guanine nucleotide exchange factor 1 isoform ARHGEF7 NM 003899 Rho guanine nucleotide exchange factor 7 isoform ARID3B NM 006465 AT rich interactive domain 3B (BRIGHT- like) ARID5B NM 032199 AT rich interactive domain SB (MRF1-like) ARL15 NM 019087 ADP-rihosylation factor related protein 2 ARL3 NM 004311 ADP-riboaylation factor-like 3 ARL6 NM 032146 ADP-rihosylation factor-like 6 ARL6IP2 NM 022374 -ADP-riboylation factor-like 6 interacting -a23 - WO 2008/036718 PCT/US2007/078859 ARMC5 NM_024742 armadillo repeat containing 5 ARMC8 NM_014154 armadillo repeat containing 8 isoform 1 ARNT NM_001668 aryl hydrocarbon receptor nuclear translocator ARRDC4 NM 183376 arrestin domain containing 4 ARSD NM 001669 arylsulfatase D isoform a precursor ARTS-i NM_016442 type 1 tumor necrosis factor receptor shedding ASAM NM 024769 adipocyte-specific adhesion molecule ASB4 NM 145872 ankyrin repeat and SOCS box-containing protein 4 ASB6 NM_017873 ankyrin repeat and SOCS box-containing 6 isoform ASCC3 NM_006828 activating signal cointegrator 1 complex subunit ASL NM_000048 argininosuccinate lyase isoform 1 ASPH NM_004318 aspartate beta-hydroxylase isoform a ASTN . NM 004319 astrotactin isoform 1 ASXL1 NM 015338 additional sex combs like 1 ASXL2 NM 018263 additional sex combs like 2 ATCAY NM_033064 caytaxin ATF3 NM_001030287 activating transcription factor 3 isoform 1 ATG1O NM_031482 APG10 autophagy 10-like ATG12 NM_004707 APG12 autophagy 12-like ATG9A NM 024085 APG9 autophagy 9-like 1 ATG9B NM_173681 nitric oxide synthase 3 antisense ATHL1 NM 025092 hypothetical protein LOC80162 ATM NM 000051 ataxia telangiectasia mutated protein isoform 1 ATOH8 NM 032827 atonal homolog 8 ATP10A NM_024490 ATPase, Class V, type 1OA ATP11B NM _014616 ATPase, Class VI, type 11 B ATP11C NM_001010986 ATPase, Class VI, type 1IC isoform b ATP1A2 NM_000702 Na+/K+ -ATPase alpha 2 subunit proprotein ATP1A3 NM_152296 Na+/K+ -ATPase alpha 3 subunit ATP2B2 NM_001001331 plasma membrane calcium ATPase 2 isoform a ATP6AP1 NM_001183 ATPase, H+ transporting, lysosomal accessory ATP6VOE NM_003945 ATPase, H+ transporting, lysosomal, VO subunit ATP6V1A NM_001690 ATPase, H+ transporting, lysosomal 70kD, VI ATP6V1C2 NM_144583 vacuolar H+ ATPase C2 isoform b ATP6V1F NM_004231 ATPase, H+ transporting, lysosomal 14kD, Vt ATP8A1 NM 006095 ATPase, aminophospholipid transporter (APLT), ATPBD4 NM 080650 ATP binding domain 4 ATPIF1 NM_178191 ATPase inhibitory factor 1 isoform 3 precursor ATXN1 NM 000332 ataxin 1 AVPR1B NM_000707 arginine vasopressin receptor lB AZGPI NM_001185 alpha-2-glycoprotein 1, zinc B3GNT6 NM 138706 UDP-GlcNAc:betaGal B4GALT1 NM_001497 UDP-Gal:betaGlcNAe beta 1,4 B4GALT5 NM_004776 UDP-Gal:betaGlcNAc beta 1,4 BAAT NM_001701 bile acid Coenzyme A: amino acid BACE1 NM 012104 beta-site APP-cleaving enzyme 1 isoform A BACHI NM_001011545 BTB and CNC homology 1 isoform b BACH2 NM 021813 BTB and CNC homology 1, basic leucine zipper BAG1 NM_004323 BCL2-associated athanogene isoform IL BAG3 NM 004281 BCL2-associated athanogene 3 BAG5 NM 001015048 BCL2-associated athanogene 5 isoform b BAGE4 NM_181704 B melanoma antigen family, member 4 BAR.HL2 NM 020063 BarH-like 2 BAT2D1 NM 015172 HBxAg transactivated protein 2 - 24 - WO 2008/036718 PCT/US2007/078859 BATF2 NM_138456 basic leucine zipper transcription factor, BAZ2A NM_013449 bromodomain adjacent to zinc finger domain, 2A BBC3 NM_014417 BCL2 binding component 3 BBS1 NM_024649 Bardet-Biedl syndrome 1 BBS5 NM_152384 Bardet-Biedl syndrome 5 BCAN NM_198427 brevican isoform 2 BCAP29 NM 001008406 B-cell receptor-associated protein BAP29 isoform BCAP3 1 NM_005745 B-cell receptor-associated protein 31 BCL2 NM_000633 B-cell lymphoma protein 2 alpha isoform BCL3 NM_005178 B-cell CLL/lymphoma 3 BCORL1 NM 021946 BCL6 co-repressor-like 1 BCR NM_004327 breakpoint cluster region isoform 1 BDH2 NM 020139 3-hydroxybutyrate dehydrogenase, type 2 BET1L NM_016526 blocked early in transport I homolog (S. BFAR NM 016561 apoptosis regulator BGN NM_001711 biglycan preproprotein BHLHB9 NM_030639 basic helix-loop-helix domain containing, class BHMT2 NM 017614 betaine-homocysteine methyltransferase 2 BICD1 NM 001003398 bicaudal D homolog 1 isoform 2 BIRCI NM_004536 baculoviral IAP repeat-containing 1 BIRC2 NM 001166 baculoviral IAP repeat-containing protein 2 BIRC4 NM 001167 baculoviral IAP repeat-containing protein 4 BIRC4BP NM 017523 XIAP associated factor-I isoform 1 BIRC5 NM_001012270 baculoviral IAP repeat-containing protein 5 BLMH NM 000386 bleomycin hydrolase BLOC1S2 NM_001001342 biogenesis of lysosome-related organelles BLR1 NM 001716 Burkitt lymphoma receptor 1 isoform 1 BLZF1 NM_003666 basic leucine zipper nuclear factor 1 BMPRlA NM 004329 bone morphogenetic protein receptor, type IA BMPR2 NM_001204 bone morphogenetic protein receptor type It BOK NM 032515 BCL2-related ovarian killer BOLA2 NM 001031833 BolA-like protein 2 isoform b BOLL NM 033030 boule isoform 2 BPNT1 NM_006085 3'(2'), 5-bisphosphate nucleotidase 1 BRCA1 NM_007306 breast cancer 1, early onset isoform BRD2 NM 005104 bromodomain containing protein 2 BRD4 NM_014299 bromodomain-containing protein 4 isoform short BSN NM_003458 bassoon protein BTBD14B NM 052876 transcriptional repressor NACl BTBD15 NM_014155 BTB (POZ) domain containing 15 BTBD4 NM_025224 BTB (POZ) domain containing 4 BTBD6 NM_033271 BTB domain protein BDPL BTF3L4 NM_152265 transcription factor BTF3-like BTG2 NM 006763 B-cell translocation gene 2 BTN1A1 NM 001732 butyrophilin, subfamily 1, member Al BTN2A1 NM_007049 butyrophilin, subfamily 2, member Al isoform 1 BTN2A2 NM 006995 butyrophilin, subfamily 2, member A2 isoform a BTN3A2 NM_007047 butyrophilin, subfamily 3, member A2 precursor BTNL8 NM_024850 butyrophilin-like 8 short form BTRC NM 003939 beta-transducin repeat containing protein BVES NM 007073 blood vessel epicardial substance C1Oorfl0 NM_007021 fasting induced gene ClOorflO4 NM 173473 hypothetical protein LOC1 19504 ClOorfl 11 NM_153244 hypothetical protein LOC221060 -25- WO 2008/036718 PCT/US2007/078859 CIOorfl 14 NM 001010911 hypothetical protein L0C399726 ClOorfl2 NM 015652 hypothetical protein L0C26148 C10orfl29 NM 207321 hypotheticalproteinL0C142827 C1Oorf38 NM 001010924 hypotheticallproteinl0C221061 C1Oorf39 NM 194303 hypotheticallproteinL0C282973 C10orf42 NM 138357 hypothetical protein L0C90550 C10orf46 NM 153810 hypothetica protein L0C143384 C1Oorf53 NM 182554 hypothetical protein L0C282966 C1Oorf54 NM 022153 hypothetical protein L0C64115 ClOorf56 NM_153367 hypothetical protein L0C219654 COorf65 NM_138413 hypothetical protein LOC 12817 C1Oorf83 NM_178832 hypothetical protein LOCI 11812 ClOorf99 NM_207373 hypothetical protein L0C387695 Cl lorfl NM 022761 hypothetical protein L0C64776 Clorfl7 NM_182901 chromosome 11 open reading frame 17 C1 1orf45 NM 145013 hypothetical protein L0C219833 Cl lorf46 NM 152316 hypothetical protein L0C120534 C1 1orf49 NM 001003676 hypothetical protein L0C79096 isoform 1 C1 1orf54 NM 014039 hypothetical protein LOC28970 Cl lorf55 NM 207428 hypothetical protein L0C399879 C1 lorf69 NM 152314 hypothetical protein L0C120196 Cl2orf22 NM_030809 TGF-beta induced apoptosia protein 12 Cl2orf29 NM_001009894 hypothetical protein L0C91298 Cl2orf3l NM_032338 hypothetical protein L0C84298 Cl2orf4l NM 017822 hypothetical protein L0C54934 C12orf5 NM 020375 chromosome 12 open reading frame 5 Cl2orf59 NM 153022 hypothetical protein L0C120939 Cl3orf3 NM_145061 hypothetical protein L0C221150 C14orflO3 NM 018036 hypothetical protein L0C55102 C14orfl1 NM 018453 hypotheticalproteinLOC55837 C14orfl 15 NM 018228 hypothetical protein L0C55237 C14orfI43 NM 145231 hypothetical protein L0C90141 Cl4orfl50 NM_001008726 hypothetical protein LOCI 12840 C14orfl62 NM 020181 chromosome 14 open reading frame 162 Cl4orf43 NM 194278 hypothetical protein LOC91748 Cl4orf58 NM 017791 hypothetical protein L0C55640 C14orf8 NM 173846 chromosome 14 open reading frame 8 C15orfI5 NM 016304 ribosomal protein L24-like C15orf2O NM 025049 DNA helicase homolog PIFI C15orf27 NM 152335 hypothetical protein LOCI23591 C15orf38 NM 182616 hypothetical protein L0C348 10 Cl5orf39 NM 015492 hypothetical protein L0C56905 Cl5orf42 NM 152259 leucine-rich repeat kinase 1 C16orf53 NM_024516 hypothetical protein L0C79447 C16orf54 NM 175900 hypothetical protein L0C283897 C16orf58 NM 022744 hypothetical protein L0C64755 Cl7orf28 NM 030630 hypothetical protein L0C283987 C17orf42 NM 024683 hypothetical protein L0C79736 C17orf45 NM 152350 hypothetical protein LOCI25144 Cl7orf53 NM 024032 hypothetical protein L0C78995 C17orf56 NM 144679 hypothetical protein L0C146705 C17orf59 NM 017622 hypothetical protein LOC54785 C17orf69 NM_152466 _hypothetical protein LOC147081 Cl -8orfl NM_001003674 -hypothetical protein LOC753 isoform gaa I - 26 - WO 2008/036718 PCT/US2007/078859 C18orf24 NM_145060 hypothetical protein LOC220134 Cl8orf25 NM 001008239 chromosome 18 open reading frame 25 isoform b C18orf45 NM 032933 hypothetical protein LOC85019 Cl9orfl0 NM 019107 chromosome 19 open reading frame 10 C19orf23 NM 152480 hypothetical protein LOC148046 C19orf35 NM 198532 hypothetical protein LOC374872 C19orf39 NM 175871 hypothetical protein LOCl26074 C19orf4 NM 012109 brain-specific membrane-anchored protein Clorfl06 NM 018265 hypothetical protein LOC55765 Clorfl07 NM 014388 hypothetical protein LOC27042 ClorflO8 NM 024595 hypothetical protein LOC79647 Clorfl09 NM 017850 hypothetical protein LOC54955 Cl orfl 15 NM_024709 hypothetical protein LOC79762 Clorfl 16 NM 023938 specifically androgen-regulated protein Cl orfl17 NM 182623 hypothetical protein LOC348487 Cl orfl19 NM 020141 hypothetical protein LOC56900 CIorfl3O NM 001010980 hypothetical protein LOC400746 Clorfl35 NM_024037 hypothetical protein LOC79000 Clorfl40 NM 001010913 hypothetical protein LOC400804 Clorfl44 NM 015609 putative MAPK activating protein PM20,PM21 Clorfl45 NM_001025495 hypothetical protein LOC574407 Clorfl49 NM 022756 hypothetical protein LOC64769 Clorfl51 NM 001032363 chromosome 1 open reading frame 151 protein Clorf157 NM_182579 hypothetical protein LOC284573 CIorfl62 NM_174896 hypothetical protein LOC128346 Cl orfl66 NM_024544 hypothetical protein LOC79594 Clorfl72 NM_152365 hypothetical protein LOC126695 Clorfl73 NM_001002912 hypothetical protein LOC127254 Clorfl83 NM 019099 hypothetical protein LOC55924 isoform 1 Clorfl87 NM 198545 chromosome 1 open reading frame 187 Clorf2l NM 030806 chromosome 1 open reading frame 21 Clorf36 NM 183059 chromosome I open reading frame 36 Clorf38 NM_004848 basement membrane-induced gene isoform 1 Clorf45 NM 001025231 hypothetical protein LOC448834 Cl orf49 NM_032126 hypothetical protein LOC84066 Clorf52 NM_198077 hypothetical protein LOC148423 Clorf53 NM_001024594 hypothetical protein LOC388722 Clorf56 NM 017860 hypothetical protein LOC54964 Clorf61 NM 006365 transcriptional activator of the c-fos promoter Clorf66 NM 015997 hypothetical protein LOC51093 Clorf69 NM_001010867 hypothetical protein LOC200205 Clorf74 NM 152485 hypothetical protein LOC148304 Clorf76 NM 173509 hypothetical proteinMGC16664 Clorf80 NM_022831 hypothetical protein LOC64853 Clorf83 NM_153035 hypothetical protein LOC127428 Cl orf95 NM 001003665 hypothetical protein LOC375057 Clorf96 NM_145257 hypothetical protein LOC126731 C1QTNF1 NM 030968 CIq and tumor necrosis factor related protein 1 ClRL NM 016546 complement component 1, r subcomponent-like C20orfl8 NM 080821 hypothetical protein LOC116151 C20orfl 1 NM 017896 chromosome 20 open reading frame It C20orfl11 |NM 016470 oxidative stress responsive 1 C20orfl2 NM 018152 hypothetical protein LOC55184 C200rf28 NM 015417 hypothetical protein LOC25876 - 27 - WO 2008/036718 PCT/US2007/078859 C20orf29 NM_018347 hypothetical protein LOC55317 C20orf4 NM_015511 hypothetical protein LOC25980 C20orf42 NM 017671 chromosome 20 open reading frame 42 C20orf43 NM_016407 hypothetical protein LOC51507 C20orf44 NM_018244 basic FGF-repressed Zic binding protein isoform C20orf98 NM_024958 hypothetical protein LOC80023 C21orfl 14 NM 001012707 hypothetical protein LOC378826 C21orf24 NM 001001789 hypothetical protein LOC400866 C21orf29 NM 144991 chromosome 21 open reading frame 29 C21orf62 NM_019596 hypothetical protein LOC56245 C21orf69 NM_058189 chromosome 21 open reading frame 69 C21orf93 NM_145179 hypothetical protein LOC246704 C22orfl3 NM_031444 chromosome 22 open reading frame 13 C22orfl8 NM_001002876 proliferation associated nuclear element 1 C22orf25 NM 152906 hypothetical protein LOC128989 C22orf9 NM 001009880 hypothetical protein LOC233 13 isoform b C2orfl 1 NM_144629 hypothetical protein LOC130132 C2orfl5 NM_144706 hypothetical protein LOC150590 C2orfl7 NM_024293 hypothetical protein LOC79137 C2orfl8 NM_017877 hypothetical protein LOC54978 C2orf27 NM_013310 hypothetical protein LOC29798 C2orf37 NM_025000 hypothetical protein LOC80067 C3orfl7 NM_001025072 hypothetical protein LOC25871 isoform b C3orf21 NM 152531 hypothetical protein LOC152002 C3orf23 NM 001029839 hypothetical protein LOC285343 isoform 2 C3orf34 NM 032898 hypothetical protein LOC84984 C4orfl3 NM_001030316 hypothetical protein LOC84068 isoform a C5orf2l NM 032042 hypothetical protein LOC83989 C5orf24 NM 152409 hypothetical protein LOC134553 C5orf4 NM_016348 hypothetical protein LOC10826 isoform 1 C6orfl30 NM 145063 hypothetical protein LOC221443 C6orfl49 NM 020408 hypothetical protein LOC57128 C6orfl5 NM 014070 STG protein C6orfl55 NM_024882 hypothetical protein LOC79940 C6orfl57 NM_198920 hypothetical protein LOC90025 C6orfl65 NM_178823 hypothetical protein LOC154313 isoform 2 C6orf2Ol NM_206834 hypothetical protein LOC404220 C6orf205 NM_001010909 hypothetical protein LOC394263 C6orf69 NM 173562 hypothetical protein LOC222658 C6orf96 NM 017909 hypothetical protein LOC55005 C6orf97 NM 025059 hypothetical protein LOC80129 C7 NM_000587 complement component 7 precursor C7orf34 NM_178829 hypothetical protein LOC135927 C7orf38 NM_145111 hypothetical protein LOC221786 C8orfl NM_004337 hypothetical protein LOC734 C8orfl7 NM_020237 MOST-1 protein C8orf44 NM_019607 hypothetical protein LOC56260 C8orf5l NM_024035 hypothetical protein LOC78998 C9orfl06 NM_001012715 hypothetical protein LOC414318 C9orfl28 NM_001012446 hypothetical protein LOC392307 C9orfl4O NM_178448 hypothetical protein LOC89958 C9orfl52 NM 001012993 hypothetical protein LOC401546 C9orfl63 NM 152571 hypothetical protein LOC158055 C9orf25 NM 147202 hypothetical protein LOC203259 -28- WO 2008/036718 PCT/US2007/078859 C9orf27 NM 021208 chromosome 9 open reading frame 27 C9orf42 NM_138333 hypothetical protein LOCI 16224 C9orf5 NM_032012 hypothetical protein LOC23731 C9orf50 NM 199350 hypothetical protein LOC375759 C9orf58 NM 001002260 chromosome 9 open reading frame 58 isoform 2 C9orf65 NM 138818 hypothetical protein LOC158471 C9orf89 NM 032310 chromosome 9 open reading frame 89 C9orf91 NM 153045 hypothetical protein LOC203197 CA12 NM 001218 carbonic anhydrase XII isoform 1 precursor CA2 NM_000067 carbonic anhydrase II CABLES2 NM_031215 Cdk5 and Abl enzyme substrate 2 CACHD1 NM_020925 cache domain containing 1 CACNAlE NM 000721 calcium channel, voltage-dependent, alpha 1E CACNA2D2 NM 001005505 calcium channel, voltage-dependent, alpha CACNA2D3 NM 018398 calcium channel, voltage-dependent, alpha CACNG4 NM_014405 voltage-dependent calcium channel gamma-4 CALCB NM 000728 calcitonin-related polypeptide, beta CALDI NM 004342 caldesmon 1 isoform 2 CALM3 NM 005184 calmodulin 3 CALML4 NM 033429 calmodulin-like 4 isoform 2 CALN1 NM 001017440 calneuron 1 CALR NM 004343 calreticulin precursor CAMK2A NM 015981 calcium/calmodulin-dependent protein kinase IIA CAMK2D NM 172127 calcium/calmodulin-dependent protein kinase It CAMK2G NM_001222 calcium/calmodulin-dependent protein kinase II CAMKK1 |NM_032294 calcium/calmodulin-dependent protein kinase 1 CAMKK2 NM 006549 calcium/calmodulin-dependent protein kinase CAMLG NM _001745 calcium modulating ligand CAMSAP1 NM 015447 calmodulin regulated spectrin-associated protein CANDI NM 018448 TIP120 protein CAPNI1 NM_007058 calpain 11 CAPN3 NM 212464 calpain 3 isoform g CAPZB INM_004930 F-actin capping protein beta subunit CARKL NM_013276 carbohydrate kinase-like CASC2 NM 178816 cancer susceptibility candidate 2 isoform 1 CASC3 NM_007359 cancer susceptibility candidate 3 CASKIN2 NM_020753 cask-interacting protein 2 CASP2 NM 032982 caspase 2 isoform 1 preproprotein CASP8 NM_001228 caspase 8 isoform A CASQ2 NM 001232 cardiac calsequestrin 2 CAST1 NM 015576 cytomatrix protein p110 CBFA2T2 NM 001032999 core-binding factor, runt domain, alpha subunit CBFB NM 001755 core-binding factor, beta subunit isoform 2 CBL NM_005188 Cas-Br-M (murine) ecotropic retroviral CBLL1 NM 024814 Cas-Br-M (murine) ecotropic retroviral CBX7 NM_175709 chromobox homolog 7 CC2D1B NM_032449 coiled-coil and C2 domain containing lB CCBL1 NM_004059 cytoplasmic cysteine conjugate-beta lyase OCBP2 NM_001296 chemokine binding protein 2 CCDC1O2B NM 024781 hypothetical protein LOC79839 CCDC14 NM 022757 coiled-coil domain containing 14 CCDC21 NM 022778 coiled-coil domain containing 21 CCDC25 NM 001031708 coiled-coil domain containing 25 isoform 1 CCDC33 NM 182791 hypothetical protein LOC80125 - 29 - WO 2008/036718 PCT/US2007/078859 CCDC49 NM 017748 hypothetical protein LOC54883 CCDC58 NM 001017928 hypothetical protein LOC131076 CCDC68 NM 025214 CTCL tumor antigen se57-1 CCDC72 NM_015933 hypothetical protein LOC51372 CCDC93 NM_019044 hypothetical protein LOC54520 CCDC94 NM_018074 hypothetical protein LOC55702 CCDC97 NM_052848 hypothetical protein LOC90324 CCDC98 NM_139076 coiled-coil domain containing 98 CCKAR NM_000730 cholecystokinin A receptor CCL18 NM 002988 small inducible cytokine A18 precursor CCL22 NM_002990 small inducible cytokine A22 precursor CCL4Ll NM_001001435 chemokine (C-C motif) ligand 4-like 1 precursor CCL4L2 NM_207007 chemokine (C-C motif) ligand 4-like 2 precursor CCL7 NM 006273 chemokine (C-C motif) ligand 7 precursor CCND1 NM_053056 cyclinD1 CCND2 NM 001759 cyclin D2 CCNT2 NM_001241 cyclin T2 isoform a CCPG1 NM 004748 cell cycle progression 1 isoform 1 CCR1 NM_001295 chemokine (C-C motif) receptor 1 CCR2 NM_000647 chemokine (C-C motif) receptor 2 isoform A CCR6 NM 004367 chemokine (C-C motif) receptor 6 CCT5 NM 012073 chaperonin containing TCP1, subunit 5 (epsilon) CD109 NM 133493 CD109 CD164L2 NM_207397 CD164 sialomucin-like 2 CD22 NM_001771 CD22 antigen CD244 NM_016382 CD244 natural killer cell receptor 2B4 CD276 NM 001024736 CD276 antigen isoform a CD28 NM 006139 CD28 antigen CD300C NM_006678 CD300C antigen CD300LG NM_145273 triggering receptor expressed on myeloid cells CD34 NM_001025109 CD34 antigen isoform a CD3D NM 000732 CD3D antigen, delta polypeptide (TiT3 complex) CD4 NM 000616 CD4 antigen precursor CD40 NM_152854 CD40 antigen isoform 2 precursor CD44 NM 000610 CD44 antigen isoform 1 precursor CD47 NM 001025079 CD47 molecule isoform 3 precursor CD53 NM 000560 CD53 antigen CD80 NM_005191 CD80 antigen (CD28 antigen ligand 1, B7-1 CD82 NM 001024844 CD82 antigen isoform 2 CD84 NM 003874 CD84 antigen (leukocyte antigen) CD8A NM 001768 CD8 antigen alpha polypeptide isoform 1 CD93 NM 012072 CD93 antigen precursor CDAN1 NM 138477 codanin 1 CDC25A NM 001789 cell division cycle 25A isoform a CDC25B NM_004358 cell division cycle 25B isoform 2 CDC42BPA NM 003607 CDC42-binding protein kinase alpha isoform B CDC42SEI NM 020239 CDC42 small effector 1 CDCA5 NM 080668 cell division cycle associated 5 CDGAP NM_020754 Cdc42 GTPase-activating protein CDH1 NM 004360 cadherin 1, type 1 preproprotein CDH17 NM_004063 cadherin 17 precursor CDH3 NM_001793 cadherin 3, type 1 preproprotein CDH5 NM 001795 cadherin 5, type 2 preproprotein CDK2AP1 NM_004642 CDK2-associated protein 1 -30- WO 2008/036718 PCT/US2007/078859 CDK5R2 NM 003936 cyclin-dependent kinase 5, regulatory subunit 2 CDK5RAP3 NM 025197 CDK5 regulatory subunit associated protein 3 CDK6 NM 001259 cyclin-dependent kinase 6 CDKAL1 NM 017774 CDK5 regulatory subunit associated protein CDON NM 016952 surface glycoprotein, Ig superfamily member CDR2L NM 014603 paraneoplastic antigen CDRTl NM 006382 CMT1A duplicated region transcript 1 CDRT4 NM 173622 hypothetical protein LOC284040 CDX1NM 001804 caudal type home box transcription factor CEACAMS NM 004363 carcinoembryonic antigen-related cell adhesion CELSR1 NM 014246 cadherin EGF LAG seven-pass G-type rceptor 1 CELSR2 NM 001408 cadherin EGF LAG seven-pass G-type receptor 2 CELSR3 NM 001407 cadherin EGF LAG seven-pass G-type receptor 3 CENTA2 NM 018404 centaurin-alpha 2 protein CENTD1 NM 015230 centaurin delta 1 isoform a CENTG1 NM 014770 centaurin, gamma I CEP135 NM 025009 centrosome protein 4 CEP192 NM 018069 hypothetical protein LOC55125 isoform 2 CEP350 NM 014810 centrosome-associated protein 350 CFD NM 001928 complement factor D preproprotein CG018 NM_052818 hypothetical protein LOC90634 CGN NM 020770 cingulin CGNL1 NM032866 cingulin-like 1 CHD5 NM_015557 chromodomain helicase DNA binding protein 5 CHD6 NM032221 chromodomain helicase DNA binding protein 6 CHKA NM_001277 choline kinase alpha isoform a CHKB NM 152253 choline/ethanolamin se efor isoform b CHML NM 001821 choroideremia-like Rab escort protein 2 CHPF NM 024536 chondroitin polymerizing factor CHRNB1 NM 000747 nicotinic acetylcholine receptor beta subunit CHRNB2 NM 000748 cholinergic receptor, nicotinic, beta CH1RNG NM 005199 cholinergic receptor, nicotinic, gamma CHST10 NM 004854 HNK-1 sulfotransferase CHST13 NM_152889 carbohydrate (chondroitin 4) sulfotransferase CHST3 NM004273 carbohydrate (chondroitin 6) sulfotransferase 3 CHST4 NM 005769 carbohydrate (N-acetylglucosamine 6-O) C-HURCl- NM 145165 churchill domain containing 1 CIAPIN1 NM_020313 cytokine induced apoptosis inhibitor 1 CIAS1 NM 004895 cryopyrin isoform a CIDEA ,NM_001279 cell death-inducing DFFA-like effector a isoform CIR NM 004882 CBF interacting corepressor CIT NM 007174 citron CITED4 NM_133467 Cbp/p300-interacting transactivator, with CLASP1 NM 015282 CLIP-associating protein 1 CLCN6 NM 001286 chloride channel 6 isoform CC-6a CLE C12A NM_138337 myeloid inhibitory C-type lectin-like receptor CL-EC12B NM 205852 macrophage antigen ht C _LEC4E NM_014358 C-6-type lectin domain family 4, member E CLEC4F NM_173535 CG-type lectin, superfamily member 13 CL-EC5A NM 013252 C-type lectin, superfamily member 5 CL-IC4 NM 013943 chloride intracellular channel 4 C _LN5 NM 006493 croid-lipofuscinosis, neuronal 5 CL-N6 NM 017882 CLN6 protein CLN8 NM 018941 CLN8 protein - 31 - WO 2008/036718 PCT/US2007/078859 CLPS NM_001832 colipase preproprotein CLYBL NM 138280 citrate lyase beta like CMYA5 NM 153610 cardiomyopathy associated 5 CNDP2 NM 018235 CNDP dipeptidase 2 (metallopeptidase M20 CNGA2 NM 005140 cyclic nucleotide gated channel alpha 2 CNGA3 NM 001298 cyclic nucleotide gated channel alpha 3 CNGB1 NM_001297 cyclic nucleotide gated channel beta 1 CNNM1 NM_020348 cyclinMI CNNM3 NM 017623 cyclin M3 isoform 1 CNOT4 NM 013316 CCR4-NOT transcription complex, subunit 4 CNP NM 033133 2',3'-cyclic nucleotide 3' phosphodiesterase CNTD1 NM 173478 hypothetical protein LOC124817 CNTD2 NM_024877 hypothetical protein LOC79935 CNTNAP2 NM_014141 cell recognition molecule Caspr2 precursor COG4 NM 015386 component of oligomeric golgi complex 4 COG5 NM 006348 component of oligomeric golgi complex 5 isoform COL12A1 NM 004370 collagen, type XII, alpha 1 long isoform COL18A1 NM 030582 alpha 1 type XVIII collagen isoform 1 precursor COL1Al NM_000088 alpha 1 type I collagen preproprotein COL21A1 NM_030820 collagen, type XXI, alpha 1 precursor COL24A1 NM 152890 collagen, type XXIV, alpha 1 COL4A4 NM_000092 alpha 4 type IV collagen precursor COL4A5 NM_000495 alpha 5 type IV collagen isoforn 1, precursor COL5A2 NM 000393 alpha 2 type V collagen preproprotein COL5A3 NM 015719 collagen, type V, alpha 3 preproprotein COL9A1 NM_001851 alpha 1 type IX collagen isoform 1 precursor COL9A2 NM 001852 alpha 2 type IX collagen COMMD2 NM 016094 COMM domain containing 2 COMMD5 NM 014066 hypertension-related calcium-regulated gene COMMD7 NM053041 COMM domain containing 7 COPA NM 004371 coatomer protein complex, subunit alpha COPZ1 NM 016057 coatomer protein complex, subunit zeta 1 COQ5 NM 032314 hypothetical protein LOC84274 COQ9 NM 020312 hypothetical protein LOC57017 CORIN NM 006587 corm COROiB NM 001018070 coronin, actin binding protein, B CORO2B NM 006091 coronin, actin binding protein, 2B COTL1 NM 021149 coactosin-like 1 COVA1 NM_006375 cytosolic ovarian carcinoma antigen I isoform a COX4NB NM 006067 neighbor of COX4 COX7A2L NM 004718 cytochrome c oxidase subunit VIla polypeptide 2 CPl10 NM 014711 CP110 protein CPAMD8 NM 015692 C3 and PZP-like, alpha-2-macroglobulin domain CPB2 NM 001872 plasma carboxypeptidase B2 isoform a CPD NM001304 carboxypeptidase D precursor CPLX2 NM 001008220 complexin 2 CPM NM 001005502 carboxypeptidase M precursor CPNE3 NM 003909 copine III CPOX NM 000097 coproporphyrinogen oxidase CPSF2 NM 017437 cleavage and polyadenylation specific factor 2 CPSF3L NM 032179 related to CPSF subunits 68 kDa isoform 2 CRAMP1L NM 020825 Crm, cramped-like CREB 1 NM 004379 cAMP responsive element binding protein 1 CREB3L2 NM 194071 cAMP responsive element binding protein 3-like - 32 - WO 2008/036718 PCT/US2007/078859 CREB5 NM 001011666 cAMP responsive element binding protein 5 CREBL2 NM_001310 cAMP responsive element binding protein-like 2 CREG2 NM 153836 cellular repressor of ElA-stimulated genes 2 CRELD1 NM_001031717 cysteine-rich with EGF-like domains 1 isoform 1 CRISPLD2 NM 031476 cysteine-rich secretory protein LCCL domain CRK NM_005206 v-crk sarcoma virus CT1O oncogene homolog CRLF3 NM 015986 cytokine receptor-like factor 3 CRNKL1 NM 016652 crooked neck-like 1 protein CRSP2 NM_004229 cofactor required for Sp1 transcriptional CRSP7 NM_004831 cofactor required for Sp 1 transcriptional CRTC3 NM_022769 transducer of regulated CREB protein 3 CRX NM_000554 cone-rod homeobox protein CSDC2 NM 014460 RNA-binding protein pippin CSF1 NM 172212 colony stimulating factor I isoform a precursor CSF2RA NM_006140 colony stimulating factor 2 receptor alpha chain CSMDI NM_033225 CUB and Sushi multiple domains 1 CSNKIG1 NM_001011664 casein kinase 1, gamma 1 isoform L CSNK1G3 NM_001031812 casein kinase 1, gamma 3 isoform 2 CSNK2A1 NM_001895 casein kinase II alpha 1 subunit isoform a CSPG3 NM 004386 chondroitin sulfate proteoglycan 3 (neurocan) CSRP3 NM 003476 cysteine and glycine-rich protein 3 CSTB NM_000100 cystatin B CTAGE1 NM_172241 cutaneous T-cell lymphoma-associated antigen 1 CTDSP2 NM_005730 nuclear LIM interactor-interacting factor 2 CTF1 NM_001330 cardiotrophin 1 CTGF NM_001901 connective tissue growth factor CTH NM_001902 cystathionase isoform 1 CTLA4 NM_005214 cytotoxic T-lymphocyte-associated protein 4 CTNNBIPI NM 001012329 catenin, beta interacting protein 1 CTNNDI NM_001331 catenin (cadherin-associated protein), delta 1 CTSB NM_001908 cathepsin B preproprotein CTSC NM_148170 cathepsin C isoform b precursor CTSD NM_001909 cathepsin D preproprotein CTSS NM_004079 cathepsin S preproprotein CTTN NM 005231 cortactin isoform a CTXN1 NM 206833 cortexin 1 CUBN NM 001081 cubilin CUGBP2 NM 001025076 CUG triplet repeat, RNA binding protein 2 CUL3 NM 003590 cullin 3 CUL5 NM_003478 Vasopressin-activated calcium-mobilizing CWF19L1 NM_018294 CWF19-like 1, cell cycle control CX3CL1 NM 002996 chemokine (C-X3-C motif) ligand 1 CXCL12 NM_000609 chemokine (C-X-C motif) ligand 12 (stromal CXCL14 NM 004887 small inducible cytokine B14 precursor CXCL9 NM_002416 small inducible cytokine B9 precursor CXorf2l NM 025159 hypothetical protein LOC80231 CXorf23 NM 198279 hypothetical protein LOC256643 CXorf34 NM 024917 hypothetical protein LOC79979 CXorf38 NM 144970 hypothetical protein LOC159013 CXorf53 NM_001018055 BRCAl/BRCA2-containing complex subunit 36 CXXC5 NM_016463 CXXC finger 5 CXXC6 NM 030625 CXXC finger 6 CYB561D1 NM 182580 cytochrome b-561 domain containing 1 CYB5B NM 030579 cytoebrome b5 outer mitochondrial membrane - 33 - WO 2008/036718 PCT/US2007/078859 CYB5D1 NM 144607 hypothetical protein LOCI24637 CYBRD1 NM_024843 cytoebrome b reductase 1 CYCS NM_018947 cytochrome c CYFIP2 NM 014376 cytoplasmic FMRI interacting protein 2 CYLC2 NM 001340 cylicin 2 CYLD NM 015247 ubiquitin carboxyl-terminal hydrolase CYLD CYLN2 NM 003388 cytoplasmic linker 2 isoform 1 CYP11 B1 NM 000497 cytoebrome P450, family 11, subfamily B, CYP1i1B2 NM 000498 cytoebrome P450, subfamily XIB polypeptide 2 CYP1A2 NM 000761 cytoebrome P450, family 1, subfamily A, CYP26B1 NM 019885 cytoebrome P450, family 26, subfamily b, CYP2B6 NM 000767 cytoebrome P450, family 2, subfamily B, CYP2C9 NM_000771 cytoebrome P450, family 2, subfamily C, CYP8B1 NM 004391 cytochrome P450, family 8, subfamily B, D2HGDH NM_152783 D-2-hydroxyglutarate dehydrogenase DAB2 NM 001343 disabled homolog 2 DAPK1 NM 004938 death-associated protein kinase 1 DAPK2 NM 014326 death-associated protein kinase 2 DBF4 NM_006716 activator of S phase kinase DBT NM 001918 dihydrolipoamide branched chain trausacylase DCAKD NM 024819 dephospho-CoA kinase domain containing DCAMKL1 NM 004734 doublecortin and CaM kinase-like 1 DCLRE1C NM 001033855 anemia protein isoform a DCST2 NM 144622 hypothetical protein LOC127579 DCTD NM 001012732 dCMP deaminase isoform a DCTN4 NM 016221 dynactin 4(p 6 2 ) DCTN5 NM 032486 dyn 4 DCX NM 000555 doublecortin isofonn a DDAH1L NM 012137 dimethylarginine dimethylamiohydrolase I DDEFL1 NM 017707 deeomnandifrnitnehncg DDI NM 0010711 hypothetical protein LC41430 DDI2 NM 032341 NAD-dend einl yrogena s DDIT4L NM 145244 D -DR1 NM 001954 dicimdmi eetrfmlmme DDX11 N 004399 DA/ ApGuAaApHs o oyetd DDX17 NM 006386 DEDbxplppie1 sfrp8 DDX23 NM 004818 DED(sGlAlAs)bxpyeti 2 DEDD2 NM 133328 -dahefcodoancnannDNbiig DEFA3 NM 005217 deesnalh3prrooti DEFA6 NM 001926 dfniapa6perpoe DEGS1 NM 003676 dgnrtv praoyehmlg1 ii DENNDlC NM 024898 hyohtclptenLC95 DENND2C NM 198459 DN/ADdmi otiig2 DERA NM 015954 -exrbs--hsht doa orl9 DERL3 NM4001002862 dri- rti sfr DFFA NM 213566 DAfamnainfco,4ka lh DFFB NM 001004285 DNfrgettofao,40k ba D5GKB NM 004080 iyllerlknsbtiofmI DGKQ NM_001347 daygyeo iae ht DHCR24 . NM 014762 4dhdohlseo euteprcsr DHDDS -NM 024887 dhdooihldpopaesnhs sfr DHFR NM 000791 iyrfltreuas DHRSB N 01510 hypothetical protein LOC246379 DHRS9 NM005771 NAD-cytchroend retnldhroea/eductase cyocroe- WO 2008/036718 PCT/US2007/078859 DHTKD1 NM_018706 dehydrogenase El and transketolase domain DHX30 NM 138614 DEAH (Asp-Glu-Ala-His) box polypeptide 30 DHX37 NM_032656 DEAH (Asp-Glu-Ala-His) box polypeptide 37 DIAPHIl NM 005219 diaphanous 1 DIDO1 NM 033081 death inducer-obliterator 1 isoform c DIP13B NM 018171 DIP13 beta DIP2B NM_173602 hypothetical protein LOC57609 DIP2C NM 014974 hypothetical protein LOC22982 DIRASI NM 145173 small GTP-binding tumor suppressor 1 DIRAS2 NM 017594 Di-Ras2 DIRCI NM_052952 hypothetical protein LOCI 16093 DISCI NM_ 001012957 disrupted in schizophrenia 1 isoform Lv DIXDCl NM_033425 DIX domain containing 1 isoform b DJ12208.2 NM 020466 hypothetical protein LOC57226 DKFZP434B0335 NM 015395 hypothetical protein LOC25851 DKFZp434I1020 NM_194295 hypothetical protein LOC196968 DKFZp547H025 NM_020161 hypothetical protein LOC56918 DKFZp564K142 NM 032121 implantation-associated protein DKFZp686K16132 NM_001012987 hypothetical protein LOC388957 DKFZp686024166 NM_001009913 hypothetical protein LOC374383 DKFZp761B107 NM 173463 hypothetical protein LOC91050 DKFZp761E198 NM_138368 hypothetical protein LOC91056 DKFZp779B1540 NM 001010903 hypothetical protein LOC389384 DLC1 NM 006094 deleted in liver cancer 1 isoform 2 DLEC1 NM 007335 deleted in lung and esophageal cancer 1 isoform DLG3 NM 021120 synapse-associated protein 102 DLGAP2 NM 004745 discs large-associated protein 2 DLX1 NM 178120 distal-less homeobox 1 isoform 1 DMBXl NM 147192 diencephalon/mesencephalon homeobox I isoform b DMTF1 NM 021145 cyclin D binding myb-like transcription factor DNAH1 1 NM 003777 dynein, axonemal, heavy polypeptide 11 DNAJA4 NM_018602 DnaJ (Hsp40) homolog, subfamily A, member 4 DNAJC 11 NM 018198 DnaJ (Hsp40) homolog, subfamily C, member 11 DNAJC14 NM_032364 dopamine receptor interacting protein DNAJC18 NM_152686 DnaJ (Hsp40) homolog, subfamily C, member 18 DNAL4 NM 005740 dynein light chain 4, axonemal DNASE1L1 NM 001009932 deoxyribonuclease I-like 1 precursor DNASE2 NM 001375 deoxyribonuclease II, lysosomal precursor DNMT3A INM_022552 DNA cytosine methyltransferase 3 alpha isoform DOC2B NM 003585 double C2-like domains, beta DOCKI NM 001380 dedicator of cytokinesis 1 DOCK2 NM 004946 dedicator of cytokinesis 2 DOCK3 NM 004947 dedicator of cytokinesis 3 DOCK9 NM_015296 dedicator of cytokinesis 9 DPCR1 NM_080870 diffuse panbronchiolitis critical region 1 DPF3 NM_012074 D4, zinc and double PHD fingers, family 3 DPY19L2 NM 173812 hypothetical protein LOC283417 DPYSL3 NM_001387 dihydropyrimidinase-like 3 DQX1 NM_133637 DEAQ box polypeptide 1 (RNA-dependent ATPase) DSCAM NM 206887 Down syndrome cell adhesion molecule isoform DTNA NM 001390 dystrobrevin alpha isoform 1 DTNB NM 021907 dystrobrevin, beta isoform 1 DTWD2 NM 173666 DTW domain containing 2 DTX1 NM 004416 deltex homolog 1 -35- WO 2008/036718 PCT/US2007/078859 DTX3L NM 138287 deltex 3-like DUSP13 NM_001007271 muscle-restricted dual specificity phosphatase DUSP4 NM 001394 dual specificity phosphatase 4 isoform 1 DUT NM 001025248 dUTP pyrophosphatase isoform 1 precursor DUXI NM_012146 double homeobox, 1 DUXA NM 001012729 hypothetical protein LOC503835 DVL3 NM 004423 dishevelled 3 DYNC2LIl NM 016008 dynein 2 light intermediate chain isoform 1 DYRK1B NM 004714 dual-specificity tyrosine-(Y)-phosphorylation DZIP1 NM_014934 DAZ interacting protein 1 isoform 1 E2F2 NM_004091 E2F transcription factor 2 E2F3 NM_001949 E2F transcription factor 3 EAFl NM_033083 ELL associated factor 1 EARS2 NM_133451 hypothetical protein LOC124454 EBI3 NM_005755 Epstein-Barr virus induced gene 3 precursor ECM2 NM 001393 extracellular matrix protein 2 precursor ECOP NM_030796 EGFR-coamplified and overexpressed protein EDA2R NM_021783 X-linked ectodysplasin receptor EDARADD NM 080738 EDAR-associated death domain isoform B EDEMI NM_014674 ER degradation enhancer, mannosidase alpha-like EDG4 NM_004720 endothelial differentiation, lysophosphatidic EDN3 NM_000114 endothelin 3 isoform 1 preproprotein EEF2K NM_013302 elongation factor-2 kinase EFCAB5 NM_001033562 EF-hand calcium binding domain 5 isoform 2 EFEMPI NM_004105 EGF-containing fibulin-like extracellular matrix EFNA1 NM_004428 ephrin Al isoform a precursor EFNA3 NM_004952 ephrin A3 EFNB3 NM_001406 ephrin-B3 precursor EFS NM_005864 embryonal Fyn-associated substrate isoform 1 EGFR NM_201284 epidermal growth factor receptor isoform d EGLN1 NM_022051 egl nine homolog 1 EGR1 NM_001964 early growth response 1 EHD2 NM_014601 EH-domain containing 2 EIF2AK2 NM_002759 eukaryotic translation initiation factor 2-alpha EIF2AK3 NM_004836 eukaryotic translation initiation factor 2-alpha EIF2AK4 NM_001013703 eukaryotic translation initiation factor 2 alpha EIF2C1 NM_012199 eukaryotic translation initiation factor 2C, 1 EIF4EBP2 NM_004096 eukaryotic translation initiation factor 4E EIF4ENIF1 NM_019843 eukaryotic translation initiation factor 4E EIF5 NM_001969 eukaryotic translation initiation factor 5 ELACl NM 018696 elaC homolog 1 ELF4 NM_001421 E74-like factor 4 (ets domain transcription ELF5 NM 001422 E74-like factor 5 ESE-2b ELKI NM 005229 ELKI protein ELK4 NM_021795 ELK4 protein isoform b ELMOL NM_014800 engulfment and cell motility 1 isoform 1 ELMO2 NM_133171 engulfment and cell motility 2 ELMODI NM_018712 ELMO domain containing 1 ELOF1 NM 032377 elongation factor 1 homolog (ELF1, S. ELOVL5 NM_021814 homolog of yeast long chain polyunsaturated ELOVL6 NM_024090 ELOVL family member 6, elongation of long chain EME1 NM_152463 essential meiotic endonuclease 1 homolog I EMIDI NM 133455 EMI domain containing 1 EMPI NM_001423 epithelial membrane protein 1 -36 - WO 2008/036718 PCT/US2007/078859 EMR2 NM 013447 egf-like module containing, mucin-like, hormone ENAH NM 001008493 enabled homolog isoform a ENAM NM 031889 enamelin ENO1 NM 001428 enolase 1 ENPP1 NM 006208 ectonucleotide pyrophosphatase/phosphodiesterase ENPP5 NM 021572 ectonucleotide pyrophosphatase/phosphodiesterase ENPP6 NM 153343 ectonucleotide pyrophosphatase/phosphodiesterase ENSA 'NM_207043 endosulfine alpha isoform 2 ENTPD3 NM 001248 ectonucleoside triphosphate diphosphohydrolase EP400 NM 015409 E1A binding protein p400 EPB41 NM 004437 erythrocyte membrane protein band 4.1 EPB4lLS NM 020909 erythrocyte membrane protein band 4.1 like 5 EPHA2 NM 004431 ephrin receptor EphA2 EPHA3 NM_005233 ephrin receptor EphA3 isoform a precursor EPHB4 NM 004444 ephrin receptor EphB4 precursor EPM2AIP1 NM 014805 EPM2A interacting protein 1 EPO NM 000799 erythropoietin precursor ERBB3 NM 001982 erbB-3 isoform 1 precursor ERGIC1 NM 020462 endoplasmic reticulum-golgi intermediate ESAM NM 138961 endothelial cell adhesion molecule ESRRG NM 001438 estrogen-related receptor gamma isoform 1 ET NM 024311 hypothetical protein LOC79157 ETV1 NM 004956 ets variant gene 1 ETV3 NM 005240 ets variant gene 3 ETV6 NM 001987 ets variant gene 6 EVC NM 153717 Ellis van Creveld syndrome protein EXOC2 NM 018303 Sec5 protein EXOC4 NM 021807 SEC8 protein isoform a EXTL3 NM_001440 Reg receptor EYA2 NM 005244 eyes absent 2 isoform a EZH1 NM 001991 enhancer of zeste homolog 1 FUR NM 016946 F1I receptor isoform a precursor F13AI NM 000129 coagulation factor XIII Al subunit precursor F2R NM 001992 coagulation factor II receptor precursor F2RLl NM 005242 coagulation factor II (thrombin) receptor-like 1 F2RL3 NM 003950 coagulation factor II (thrombin) receptor-like 3 FADS2 NM 004265 fatty acid desaturase 2 FADS6 NM 178128 fatty acid desaturase domain family, member 6 FAIM2 NM 012306 Fas apoptotic inhibitory molecule 2 FAMIOOB NM 182565 hypothetical protein LOC283991 FAM102A NM_203305 early estrogen-induced gene 1 protein isoform b FAM102B NM 001010883 hypothetical protein LOC28461-1 FAM104A NM 032837 hypothetical protein LOC84923 FAM106A NM 024974 hypothetical protein LOC80039 FAM107A NM 007177 downregulated in renal cell carcinoma FAM107B NM 031453 hypothetical protein LOC83641 FAMI I 11 A NM 022074 hypothetical protein LOC63901 FAM117A NM 030802 CiEBP-induced protein FAM11A NM 032508 family with sequence similarity 11, member A FAM19Al NM 213609 family with sequence similarity 19 (chemokine FAM20B NM 014864 family with sequence similarity 20, member B FAM36A NM 198076 family with sequence similarity 36, member A FAM3B NM 058186 family with sequence similarity 3, member B FAM40A NM 033088 hypothetical protein LOC85369 - 37 - WO 2008/036718 PCT/US2007/078859 FAM43A NM 153690 hypothetical protein LOC 131583 FAM53B NM 014661 hypothetical protein LOC9679 FAM55C NM 145037 hypothetical protein LOC91775 FAM5B NM 021165 BMP/retinoic acid-inducible neural-specific FAM60A NM 021238 family with sequence similarity 60, member A FAM62C NM 031913 family with sequence similarity 62 (C2 domain FAM71C NM 153364 hypothetical protein LOC196472 FAM81A NM 152450 hypothetical protein LOC145773 FAM83E NM_017708 hypothetical protein LOC54854 FAM83F NM_138435 hypothetical protein LOCI 13828 FAM83H NM_198488 hypothetical protein LOC286077 FAM89B NM_152832 Mouse Mammary Turmor Virus Receptor homolog 1 FAM98B NM 173611 hypothetical protein LOC283742 FANCC NM_000136 Fanconi anemia, complementation group C FANCD2 NM_033084 Fanconi anemia complementation group D2 isoform FATE1 NM 033085 fetal and adult testis expressed transcript FBS 1 NM 022452 fibrosin I FBXL11 NM 012308 F-box and leucine-rich repeat protein 11 FBX016 NM_172366 F-box only protein 16 FBXO21 NM 015002 F-box only protein 21 isoform 2 FBXO27 NM 178820 F-box protein 27 FBXO31 NM_024735 F-box protein 31 FBXO34 NM_017943 F-box only protein 34 FBXO44 NM 001014765 F-box protein 44 isoform 1 FBXO9 NM_012347 F-box only protein 9 isoform 1 FBXWI1 NM 012300 F-box and WD-40 domain protein lB isoform C FBXW8 NM 012174 F-box and WD-40 domain protein 8 isoform 2 FCER2 NM_002002 Fc fragment of IgE, low affinity II, receptor FCGR3A NM _000569 Fc fragment of IgG, low affinity IIla, receptor FCGR3B NM 000570 low affinity immunoglobulin gamma Fe region FCHSDl NM 033449 FCH and double SH3 domains 1 FCMD NM 006731 fukutin FEMIC NM 020177 feminization 1 homolog a FGA NM 021871 fibrinogen, alpha polypeptide isoform alpha FGD6 NM _018351 FYVE, RhoGEF and PH domain containing 6 FGFI NM 000800 fibroblast growth factor 1 (acidic) isoform 1 FGF19 NM 005117 fibroblast growth factor 19 precursor FGFR1 NM_023107 fibroblast growth factor receptor I isoform 5 FHIT NM 002012 fragile histidine triad gene FIS NM 175616 hypothetical protein LOC202299 FKBP1O NM_021939 FK506 binding protein 10, 65 kDa FKBP1A NM_000801 FK506-binding protein 1A FKBP5 NM 004117 FK506 binding protein 5 FKBP9 NM _007270 FK506 binding protein 9 FKBP9L NM 182827 FK506 binding protein 9-like FKRP NM_024301 fukutin-related protein FKSG44 NM _031904 FKSG44 protein FLJ10159 NM 018013 hypothetical protein LOC55084 FLJ10324 NM_018059 hypothetical protein LOC55698 FLJ10357 NM 018071 hypothetical protein LOC55701 FLJ10490 NM 018111 hypothetical protein LOC55150 FLJ10803 NM 018224 hypothetical protein LOC55744 FLJ10815 NM 018231 amino acid transporter FLJ11021 NM 023012 hypothetical protein LOC65117 isoform a -38 - WO 2008/036718 PCT/US2007/078859 FLJ 11151 NM 018340 hypothetical protein L0C55313 FLJ 11171 NM 018348 hypothetical protein L0C55783 FLJ1 1259 NM 018370 hypotheticalproteinL0C55332 FLJ 11292 NM 018382 hypothetical protein L0C55338 FLJ1 1806 NM 024824 nuclear protein Up68 isoform 1 FLJ12505 NM 024749 hypothetical protein L0C79805 FLJ12681 NM 022773 hypothetical protein L0C64788 FLJ12700 NM_024910 hypothetical protein L0C79970 FLJ12949 NM_023008 hypothetical protein L0C65095 isoform 1 FLJ13197 NM 024614 hypothetical protein L0C79667 FLJ14001 NM_024677 hypothetical protein L0C79730 FLJ14213 NM 024841 hypothetical protein L0C79899 FLJ14397 NM 032779 hypothetical protein L0C84865 FLJ14816 NM 032845 hypothetical protein L0C84931 FLJ14834 NM 032849 hypothetical protein L0C84935 FLJ20032 NM 017628 hypothetical protein L0C54790 FLJ20035 NM 017631 hypothetical protein L0C55601 FLJ20160 NM 017694 hypothetical protein L0C54842 FLJ20186 NM 207514 differentially expressed in FDCP 8 isoform 1 FLJ20297 NM 017751 hypothetical protein L0C55627 isoform I FLJ20581 NM 017888 hypothetical protein L0C54988 FLJ20582 NM 014106 hypothetical protein L0C54989 FLJ20628 NM_017910 hypothetical protein L0C55006 FLJ20701 NM_017933 hypothetical protein L0C55022 FLJ20758 NM 017952 hypothetical protein L0C55037 FLJ20972 NM 025030 hypothetical protein L0C80098 FLJ21865 NM 022759 endo-beta-N-acetylglucosaminidase FLJ21963 NM 024560 hypothetical protein L0C7961 1 FLJ22795 NM 025084 hypotheticalprotein LOC80154 FLJ23322 NM 024955 hypothetical protein L0C80020 FLJ23834 NM 152750 hypothetical protein L0C222256 FLJ25996 NM 001001699 hypothetical protein LOC401109 FLJ26175 NM 001001668 hypothetia protein LOC3 88566 FLJ27365 NM 207477 hypothetical protein L0C400931 FLJ31222 NM 207388 hypothetical protein L0C388387 FLJ31568 NM 152509 hypothetical protein LOCI50244 FLJ31875 NM 182531 hypothetical protein LOCI97320 FLJ32011 NM 182516 hypothetical protein LOC 148930 FLJ32130 NM 152458 hypothetical protein L0C16540 FLJ32312 NM 144709 ypteiaprenLO106 -FL-J32447 NM_153038prtiLO127 FLJ32569 NM 152491 hypothetical protein LC48811 FLJ32894 NM14466 7 hpteia rti 146 FLJ32926 NM 207 hypothetical protein LOC144577 FLJ32955 NM 153041 FLJ33387 NM 182526hyohtclptenLC614 FL-J33860 NM_173644 hpteia rti O245 FLJ34931 NM_001029883 hpteia rti O383 FLJ35409 NM_001001688 hpteia rti O406 FLJ35429 NM_001003807 hpteia rti O253 FILJ35740 NM_147195 FJ54 rti FLJ3773 M 15599 hypothetical protein LOC53138 FLJ3580 NM 53264 hypothetical protein LOC57 FLJ3268 M 20511 hypothetical protein LOC53 hyoheialpoei9OC53 WO 2008/036718 PCT/US2007/078859 FLJ36492 NM 182568 hypothetical protein LOC284047 FLJ36874 NM_152716 hypothetical protein LOC219988 FLJ37927 NM_152623 hypothetical protein LOC166979 FLJ38288 NM_173632 hypothetical protein LOC284309 FLJ38663 NM_152269 hypothetical protein LOC91574 FLJ38973 NM 153689 hypothetical protein LOC205327 FLJ38991 NM 001033760 mitochondrial COX18 isoform 5 FLJ39370 NM 152400 hypothetical protein LOC132720 FLJ39531 NM 207445 hypothetical protein LOC400360 FLJ39743 NM 182562 hypothetical protein LOC283777 FLJ40142 NM 207435 hypothetical protein LOC400073 FLJ40852 NM 173677 hypothetical protein LOC285962 FLJ41423 NM_001001679 hypothetical protein LOC399886 FLJ41733 NM 207473 hypothetical protein LOC400870 FLJ41841 NM_207499 hypothetical protein LOC401263 FLJ41993 NM_001001694 hypothetical protein LOC400935 FLJ42102 NM_001001680 hypothetical protein LOC399923 FLJ42418 NM_001001695 hypothetical protein LOC400941 FLJ42953 NM 207474 hypothetical protein LOC400892 FLJ43339 NM_207380 hypothetical protein LOC388115 FLJ43505 NM 207468 hypothetical protein LOC400823 FLJ43582 NM 207412 hypothetical protein LOC389649 FLJ43879 NM 001001698 hypothetical protein LOC401039 FLJ43980 NM_001004299 hypothetical protein LOC1 24149 FLJ44691 NM 198506 hypothetical protein LOC345193 FLJ45079 NM 001001685 hypothetical protein LOC400624 FLJ45121 NM_207451 hypothetical protein LOC400556 FLJ45139 NM_001001692 hypothetical protein LOC400867 FLJ45202 NM 207507 hypothetical protein LOC401508 FLJ45422 NM 001004349 hypothetical protein LOC441140 FLJ45645 NM 198557 hypothetical protein LOC375287 FLJ45684 NM 207462 hypothetical protein LOC400666 FLJ45831 NM_001001684 hypothetical protein LOC400576 FLJ45850 NM_207395 hypothetical protein LOC388569 FLJ45909 NM_198445 hypothetical protein LOC126432 FLJ45910 NM_207390 hypothetical protein LOC388512 FLJ45964 NM 207483 hypothetical protein LOC401040 FLJ46010 NM_001001703 hypothetical protein LOC401191 FIJ46026 NM_207458 hypothetical protein LOC400627 FLJ46154 NM_198462 FLJ46154 protein FLJ46230 NM_207463 hypothetical protein LOC400679 FLJ46257 NM_001001693 hypothetical protein LOC400932 FLJ46266 NM_207430 hypothetical protein LOC399949 FLJ46347 NM_001005303 hypothetical protein LOC389064 FLJ46363 NM_207434 hypothetical protein LOC400002 FLJ46365 NM_207504 hypothetical protein LOC401459 FLJ46481 NM 207405 hypothetical protein LOC389197 FLJ46688 NM_001004330 hypothetical protein LOC440107 FLJ46831 NM 207426 forkhead box 12 FLJ46838 NM_001007546 hypothetical protein LOC440865 FLJ90757 |NM_001004336 hypothetical protein LOC440465 FLOT1 NM 005803 flotillin 1 FLOT2 NM 004475 flotillin 2 FLTl NM 002019 fmis-related tyrosine kinase 1 (vascular - 40 - WO 2008/036718 PCT/US2007/078859 FLT4 NM 182925 fms-related tyrosine kinase 4 isoform 1 FLYWCHI NM 032296 FLYWCH-type zinc finger 1 isoform a FMNL3 NM 175736 formin-like 3 isoform 1 FMO4 NM_002022 flavin containing monooxygenase 4 FMOD NM 002023 fibromodulin precursor FNI NM_002026 fibronectin 1 isoform 3 preproprotein FNDC1 NM_032532 fibronectin type III domain containing 1 FNDC5 NM_153756 fibronectin type III domain containing 5 FNDC8 NM_017559 hypothetical protein LOC54752 FNTB NM_002028 famesyltransferase, CAAX box, beta FOSB NM_006732 FBJ murine osteosarcoma viral oncogene homolog FOSL2 NM_005253 FOS-like antigen 2 FOXJ2 NM 018416 forkhead box J2 FOXJ3 NM 014947 forkhead box J3 FOXK2 NM 181430 forkhead box K2 isoform 2 FOXO1A NM 002015 forkhead box OlA FOXP1 NM 032682 forkhead box P1 isoform 1 FRMD1 NM 024919 FERM domain containing 1 FRMPD2 NM 001017929 FERM and PDZ domain containing 2 isoform 2 FSCN1 NM 003088 fascin 1 FSD1L NM 207647 fibronectin type III and SPRY domain containing FST NM_006350 follistatin isoform FST317 precursor FSTL4 NM 015082 follistatin-like 4 FTSJI NM_012280 FtsJ homolog 1 isoform a FUNDC2 NM_023934 FUN 14 domain containing 2 FUSIP 1 NM_006625 FUS interacting protein (serine-arginine rich) 1 FUT2 NM 000511 fucosyltransferase 2 (secretor status included) FUT4 NM_002033 fucosyltransferase 4 FUT6 NM 000150 fucosyltransferase 6 (alpha (1,3) FXYD3 NM 005971 FXYD domain containing ion transport regulator 3 FYCO1 NM 024513 FYVE and coiled-coil domain containing 1 FZD1 NM 003505 frizzled 1 GAB2 NM_012296 GRB2-associated binding protein 2 isoform b GABARAPL1 NM 031412 GABA(A) receptor-associated protein like 1 GABBR1 NM 001470 gamma-aminobutyric acid (GABA) B receptor 1 GABRA4 NM 000809 gamma-aminobutyric acid A receptor, alpha 4 GABRB3 NM_000814 gamma-aminobutyric acid (GABA) A receptor, beta GABRE NM_004961 gamma-aminobutyric acid (GABA) A receptor, GABRG1 NM_173536 gamma-aminobutyric acid A receptor, gamma I GABRG2 NM_000816 gamma-aminobutyric acid A receptor, gamma 2 GABRR2 NM_002043 gamma-aminobutyric acid (GABA) receptor, rho 2 GALC NM_000153 galactosylceramidase isoform a precursor GALM NM_138801 galactose mutarotase (aldose 1-epimerase) GALNT13 NM 052917 UDP-N-acetyl-alpha-D-galactosamine:poypeptide GALNT3 NM 004482 polypeptide N-acetylgalactosaminyltransferase 3 GALNT6 NM 007210 polypeptide N-acetylgalactosaminyltransferase 6 GALNTL2 NM_054110 UDP-N-acetyl-alpha-D-galactosamine:polypeptide GALT NM_000155 galactose-1-phosphate uridylyltransferase GAPVD1 NM_015635 GTPase activating protein and VPS9 domains 1 GARNL1 NM_014990 GTPase activating Rap/RanGAP domain-like 1 GARNL4 NM_015085 GTPase activating Rap/RanGAP domain-like 4 GAS2L1 NM 152237 growth arrest-specific 2 like 1 isoform b GAS7 NM 003644 growth arrest-specific 7 isoform a GATA4 NM 002052 GATA binding protein 4 -41- WO 2008/036718 PCT/US2007/078859 GATAD1 NM 021167 GATA zinc finger domain containing 1 GATM NM 001482 glycine amidinotransferase (L-arginine:glycine GATS NM_178831 opposite strand transcription unit to STAG3 GCLM NM 002061 glutamate-cysteine ligase regulatory protein GCM - NM 003643 glial cells missing homolog a GCNT1 NM 001490 beta-1,3-galactosyl-O-glycosyl-glycoprotein GCNT2 NM 001491 glucosaminyl (N-acetyl) transferase 2, Gcoml NM 001018097 GRINLIA combined protein isoform 8 GDAP2 NM 017686 ganglioside induced differentiation associated GDF1O NM_004962 growth differentiation factor 10 precursor GDF6 NM_001001557 growth differentiation factor 6 GDPD4 NM 182833 glycerophosphodiester phosphodiesterase domain Gene symbol hsa-miR-143 targets Genename GFODI NM 018988 glucose-fructose oxidoreductase domain GFOD2 NM 030819 hypothetical protein LOC81577 GFPT1 NM 002056 glucosamine-fructose-6-phosphate GFPT2 NM 005110 glutamine-fructose-6-phosphate transaminase 2 GGA2 NM 015044 ADP-ribosylation factor binding protein 2 GGT6 NM_153338 gamma-glutamyltransferase 6 homolog GGTL3 NM_178025 gamma-glutamyltransferase-like 3 isoform b GHR NM_000163 growth hormone receptor precursor GIF NM 005142 gastric intrinsic factor (vitamin B synthesis) GIMAP6 NM 001007224 GTPase, IMAP family member 6 isoform 3 GIT2 NM 014776 G protein-coupled receptor kinase-interactor 2 GJCl NM 152219 gap junction protein, chi 1, 31.9kDa (connexin GLB1L NM_024506 galactosidase, beta 1-like GLDC NM 000170 glycine dehydrogenase (decarboxylating; glycine GLI3 NM_000168 GLI-Kruppel family member GLI3 GLP1R NM 002062 glucagon-like peptide 1 receptor GLT25D2 NM_015101 glycosyltransferase 25 domain containing 2 GLYATL2 NM_145016 hypothetical protein LOC219970 GMEB2 NM 012384 glucocorticoid modulatory element binding GMFB NM_004124 glia maturation factor, beta GNA15 NM 002068 guanine nucleotide binding protein (G protein), GNAIl NM 002069 guanine nucleotide binding protein (G protein), GNAL NM 002071 guanine nucleotide binding protein (G protein), GNAS NM 016592 guanine nucleotide binding protein, alpha GNB3 NM 002075 guanine nucleotide-binding protein, beta-3 GNB4 NM 021629 guanine nucleotide-binding protein, beta-4 GNB5 NM 006578 guanine nucleotide-binding protein, beta-5 GNG12 NM 018841 G-protein gamma-12 subunit GNG4 NM 004485 guanine nucleotide binding protein (G protein), GNG7 NM 052847 guanine nucleotide binding protein (G protein), GNL3 NM_014366 guanine nucleotide binding protein-like 3 GNPNATl NM 198066 glucosamine-phosphate N-acetyltransferase 1 GNS NM 002076 glucosamine (N-acetyl)-6-sulfatase precursor GOLGA NM 018652 golgin-like protein GOLGA1 NM 002077 golgin 97 GOLGA4 NM 002078 golgi autoantigen, golgin subfamily a, 4 GOLPH2 NM 016548 golgi phosphoprotein 2 GORASP1 NM 031899 Golgi reassembly stacking protein 1 GOSR1 NM 001007024 golgi SNAP receptor complex member 1 isoform 3 GOT1 NM 002079 aspartate aminotransferase 1 LGOT2 NM 002080 aspartate aminotransferase 2 precursor -42 - WO 2008/036718 PCT/US2007/078859 GP5 NM_004488 glycoprotein V (platelet) GP6 NM_016363 glycoprotein VI (platelet) GPA33 NM 005814 transmembrane glycoprotein A33 precursor GPC1 NM 002081 glypican 1 precursor GPC2 NM 152742 glypican 2 GPIAPI NM_005898 membrane component chromosome 11 surface marker GPR109A NM_177551 G protein-coupled receptor 109A GPR109B NM_006018 G protein-coupled receptor 109B GPR135 NM_022571 G protein-coupled receptor 135 GPR176 NM 007223 putative G protein coupled receptor GPR180 NM 180989 G protein-coupled receptor 180 precursor GPR26 NM 153442 G protein-coupled receptor 26 GPR62 NM 080865 G protein-coupled receptor 62 GPR83 NM_016540 G protein-coupled receptor 83 GPRC5A NM_003979 G protein-coupled receptor, family C, group 5, GPRC5B NM_016235 G protein-coupled receptor, family C, group 5, GPSM3 NM 022107 G-protein signalling modulator 3 (AGS3-like, C. GPX3 NM_002084 plasma glutathione peroxidase 3 precursor GRAMD1A NM_020895 hypothetical protein LOC57655 GRAMD2 NM_001012642 hypothetical protein LOC196996 GRHL2 NM 024915 transcription factor CP2-like 3 GRIA2 NM 000826 glutamate receptor, ionotropic, AMPA 2 GRIN2B NM_000834 N-methyl-D-aspartate receptor subunit 2B GRINLIA NM_001018103 glutamate receptor, ionotropic, N-methyl GRIPAP1 NM 020137 GRIP1 associated protein I isoform I GRK1 NM 002929 rhodopsin kinase GSDMDCl NM_024736 gasdermin domain containing 1 GSTA4 NM_001512 glutathione S-transferase A4 GSTM4 NM_147149 glutathione S-transferase M4 isoform 3 GTF2I NM_001518 general transcription factor II, i isoform 4 GTPBP1 NM 004286 GTP binding protein 1 GTPBP3 NM_032620 GTP binding protein 3 (mitochondrial) isoform V GUSBL2 NM 206910 hypothetical protein LOC375513 isoform 2 H2AFY2 NM_018649 core histone macroH2A2.2 H2BFWT NM_001002916 H2B histone family, member W, testis-specific H6PD NM 004285 hexose-6-phosphate dehydrogenase precursor HABP2 NM_004132 hyaluronan binding protein 2 HAGHL NM_207112 hydroxyacylglutathione hydrolase-like isoform 1 HAPLN4 NM 023002 brain link protein 2 HAS3 NM_005329 hyaluronan synthase 3 isoform a HBS1L NM 006620 HBS1-like hCAP-H2 NM 152299 kleisin beta isoform 2 HCCS NM_005333 holocytochrome c synthase (cytochrome c HCG9 NM 005844 hypothetical protein LOC10255 HCP 1 NM_080669 heme carrier protein 1 HDAC4 NM_006037 histone deacetylase 4 HDAC7A NM_015401 histone deacetylase 7A isoform a HECA NM 016217 headcase HECTD1 NM 015382 HECT domain containing 1 HECW2 NM_020760 HECT, C2 and WW domain containing E3 ubiquitin HEMK1 NM 016173 HemK methyltransferase family member 1 HES2 NM 019089 hairy and enhancer of split homolog 2 HFE NM_000410 hemochromatosis protein isoform I precursor HGF NM_001010934 hepatocyte growth factor isoform 5 precursor -43 - WO 2008/036718 PCT/US2007/078859 HGS NM 004712 eaoyegotfatrrgledyosn HHAT NM 018194 hearan sul transferase HHLA2 NM 007023 hERa s coaminyl HIATL1 NM 01558 h etscac protein 1 HIG2 NM 013332 hyoxytid (bete dg 1 HIGD2A NM 1424 hea soc ain family, member 7 HIP-P1 NM012267 hsp-interacting protein HIPK1 NM_1157 hoetial protein 18135870 HISTlH4E NM 003545 hydoxyramine (met r HK2 NM 000189 hyoxtpamn ( HKDC NM_025130 HKR2 NM 181846 GIKuplfml ebrHR HLA-A NM 002116 mjrhsooptblt opecas1 HTLA-B NM_005514 mjrhsooptblt opecas1 HLA-C NM_002117 mjrhsooptblt opecas1 HfLA-DOA NM 002119 mao hsooptbltco lelss1,D HLA-DPA1 NM 033554 -r jrit~optblt opecasID HLA-DPB1 NM 002121 mjrhsooptblt opecas1,D HLA-DQA2 NM 020056 mjrhsooptblt opecas1,D HLA-DQB1 NM 002123 mjrhsooptblt opecas1,D HLA-E NM 005516 mjrhsooptblt opecas1 HL-F NM 002126heailekmafco HM _BS NM 000190 ydoyehliaesnaeiofr1 _HMG2L1 NM 001003681 hihmblt -goppoen2ikI sfrb HMGA1 NM 002131 hg oiiygopA-okIio~m _ HMGA2 NM 001015886 hihmbltgruATok2 sfmC HMGB1 NM 002128 hihmbltgruboI HMGCS2 NM 005518 3hdoy3mtyguay-onyeAsnhs HTMMR NM 012484 yauoa-eitdmoiiyrcprisfma HN -1 NM 001002033 hraooia adnuooia xrse HNF4_A NM 000457 Wepaoyencerfco4alhisorb HNMT NM 001024074 hitmn -ehlrnfasioom2 HENRPA0 NM 006805 heeoeeunula inceprtn HfOXA5 NM 019102 hoebxA HfOXB13 NM 006361 -hmoo B1 HOXB9 NM 024017 -h-cbxB HOXC5 NM 018953 oobxC HPCAL4 NM 016257 hpoacnlk rti HPS5 NM 007216 ernsk-ulksnrm5iofmb HPSE NM 006665 hprns HR NM 005144 hils rti sfr HRB _NM 004504 HV e idn rti HRH4 NM 021624 hitmnH4rcpo H _S2ST1 _ NM 012262 hprnslae20slornfrs HS3ST2 NM 006043 hprnslaeDguoaiy HSBP1 NM 001537 ha hc atrbnigpoe HS-D17B1 NM 000413 Iydxseod( -ea eyrgrae H _SDL2 NM 032303 hdoytri dhdoeaelk HSH2D NM 032855 b -aooei H oancnann HfSPB7 NM 014424 _ -a sok2kapoenfmlmme HSPBP1 NM 012267hs7ineatgpren _HSPC065 NM 014157hyohtclptenLC97 H _TR2C NM 000868 _ _ _yrxtytmn srtnn eetr2 HTR3A NM 000869 -_hypox-ndcilptein 2srtn eetr3 HI1doan4 aiymmbr2 WO 2008/036718 PCT/US2007/078859 HTR3B NM 006028 5-hydroxytryptamine (serotonin) receptor 3B HTR4 NM 000870 serotonin 5-HT4 receptor isoform b HTR6 NM 000871 5-hydroxytryptamine (serotonin) receptor 6 HTR7 NM 000872 5-hydroxytryptamine receptor 7 isoform a HUNK NM 014586 hormonally upregulated Neu-associated kinase HYOUI NM 006389 oxygen regulated protein precursor HYPK NM_016400 Huntingtin interacting protein K IAPP NM 000415 islet amyloid polypeptide precursor IBRDC1 NM 152553 IBR domain containing 1 ICAl NM 022308 islet cell autoantigen 1 isoform 3 ID4 NM 001546 inhibitor of DNA binding 4, dominant negative IER3 NM_003897 immediate early response 3 isoform short IFIT3 NM_001031683 interferon-induced protein with IFIT5 NM_012420 interferon-induced protein with IFNA14 NM 002172 interferon, alpha 14 IFNA16 NM_002173 interferon, alpha 16 IFNA7 NM 021057 interferon, alpha 7 IGF1 NM_000618 insulin-like growth factor 1 (somatomedin C) IGF2BP1 NM_006546 insulin-like growth factor 2 mRNA binding IGF2R NM_000876 insulin-like growth factor 2 receptor IGFBP3 NM_000598 insulin-like growth factor binding protein 3 IGFBP5 NM_000599 insulin-like growth factor binding protein 5 IGFL1 NM_198541 insulin growth factor-like family member 1 IGSF4D NM_153184 immunoglobulin superfamily, member 4D IHPKl NM_001006115 inositol hexaphosphate kinase 1 isoform 2 IHPK2 NM_001005910 inositol hexaphosphate kinase 2 isoform b IHPK3 NM_054111 inositol hexaphosphate kinase 3 ILIORA NM 001558 interleukin 10 receptor, alpha precursor IL1ORB NM_000628 interleukin 10 receptor, beta precursor ILl lRA NM_147162 interleukin 11 receptor, alpha isoform 2 IL12RBl NM_153701 interleukin 12 receptor, beta 1 isoform 2 IL12RB2 NM 001559 interleukin 12 receptor, beta 2 precursor IL13RAI NM_001560 interleukin 13 receptor, alpha I precursor IL16 NM 004513 interleukin 16 isoform 1 precursor IL17C NM 013278 interleukin 17C IL17RD NM_017563 interleukin 17 receptor D IL18 NM 001562 interleukin 18 proprotein ILIF5 NM_012275 interleukin 1 family, member 5 ILIF9 NM 019618 interleukin 1 family, member 9 ILIRAP NM 002182 interleukin 1 receptor accessory protein isoform ILIRLI NM_003856 interleukin 1 receptor-like 1 isoform 2 ILlRN NM_000577 interleukin 1 receptor antagonist isoform 3 IL22RA2 NM_052962 interleukin 22-binding protein isoform 1 IL27RA NM_004843 class I cytokine receptor IL28RA NM_170743 interleukin 28 receptor, alpha isoform 1 IL2RA NM_000417 interleukin 2 receptor, alpha chain precursor IL3 NM 000588 interleukin 3 precursor IL6R NM 181359 interleukin 6 receptor isoform 2 precursor IL8RA NM 000634 interleukin 8 receptor alpha INCAl NM_213726 inhibitor of CDK interacting with cyclin Al ING5 NM_032329 inhibitor of growth family, member 5 INOCl NM 017553 INO80 complex homolog 1 INPP5E NM_019892 inositol polyphosphate-5-phosphatase E INSL4 NM 002195 insulin-like 4 precursor - 45 - WO 2008/036718 PCT/US2007/078859 INTS2 NM_020748 integrator complex subunit 2 IQCC NM 018134 IQ motif containing C IQCE NM_152558 IQ motif containing E IRAK1 NM 001025242 interleukin-1 receptor-associated kinase 1 IRF5 NM 002200 interferon regulatory factor 5 isoform a IRF8 NM 002163 interferon regulatory factor 8 IRX6 NM 024335 iroquois homeobox protein 6 ITGA1 1 NM 001004439 integrin, alpha 11 precursor ITGA3 NM 002204 integrin alpha 3 isoform a precursor ITGA5 NM 002205 integrin alpha 5 precursor ITGA6 NM 000210 integrin alpha chain, alpha 6 ITGAM NM_000632 integrin alpha M precursor ITGAV NM 002210 integrin alpha-V precursor ITM2B NM_021999 integral membrane protein 2B ITPR1 NM 002222 inositol 1,4,5-triphosphate receptor, type 1 JAGI NM_000214 jagged 1 precursor JAGNI NM 032492 jagunal homolog 1 JMl 1 NM 033626 hypothetical protein LOC90060 JMJD2B NM 015015 jumonji domain containing 2B JMJD2C NM 015061 jumonji domain containing 2C JOSD1 NM 014876 Josephin domain containing 1 JOSD3 NM_024116 Josephin domain containing 3 JPH1 NM 020647 junctophilin 1 JPH3 NM 020655 junctophilin 3 JRK NM 003724 jerky homolog K6IRS4 NM 175053 keratin 6 irs4 KA36 NM _182497 type I hair keratin KA36 KAL1 NM 000216 Kallmann syndrome 1 protein KATNALl NM 001014380 katanin p60 subunit A-like 1 KBTBD3 NM 152433 BTB and kelch domain containing 3 KBTBD6 NM_152903 kelch repeat and BTB (POZ) domain-containing 6 KBTBD8 NM 032505 T-cell activation kelch repeat protein KCNA7 NM 031886 potassium voltage-gated channel, shaker-related KCNBI NM_004975 potassium voltage-gated channel, Shab-related KCNDI NM_004979 potassium voltage-gated channel, Shal-related KCND2 NM 012281 potassium voltage-gated channel, Shal-related KCND3 NM 004980 potassium voltage-gated channel, Shal-related KCNElL NM 012282 potassium voltage-gated channel, Isk-related KCNE3 NM_005472 potassium voltage-gated channel, Isk-related KCNH5 NM_172375 potassium voltage-gated channel, subfamily H, KCNH6 NM 173092 potassium voltage-gated channel, subfamily H, KCNH7 NM_033272 potassium voltage-gated channel, subfamily H, KCNH8 NM _144633 potassium voltage-gated channel, subfamily H, KCNIP1 :NM 014592 Kv channel interacting protein 1 isoform 2 KCNIP2 NM 014591 Kv channel interacting protein 2 isoform 1 KCNJ10 NM_002241 potassium inwardly-rectifying channel, subfamily KCNJ13 NM_002242 potassium inwardly-rectifying channel J13 KCNJ4 NM 004981 potassium inwardly-rectifying channel J4 KCNJ5 NM 000890 potassium inwardly-rectifying channel J5 KCNJ8 NM_004982 potassium inwardly-rectifying channel J8 KCNK2 NM 001017424 potassium channel, subfamily K, member 2 isoform KCNK3 NM 002246 potassium channel, subfamily K, member 3 KCNMA1 NM 001014797 large conductance calcium-activated potassium KCNS2 NM 020697 potassium voltage-gated channel, - 46 - WO 2008/036718 PCT/US2007/078859 KCTDIO NM 031954 potassium channel tetramerisation domain KIDELC2 NM 153705 KDEL (Lys-Asp-Glu-Lea) containing 2 KEAP1 NM 012289 kelch-likc ECH-associated protein 1 KENAE NM 176816 hypothetical protein LOC202243 KIAA0125 NM 014792 hypothetical protein LOC9834 KIAA0232 NM 014743 hypothetical protein LOC9778 KIAA0256 NM 014701 hypothetical protein LOC9728 KIAA0265 NM 014997 hypothetical protein LOC23008 KIAA0286 NM 015257 hypothetical protein L0C336 KIAA0319 NM 014809 h IAlon9 KIAA0319L NM 024874 oyytckdediesI-leiofra KIAA0329 NM 014844 hypothetical protein L0C9895 KIAA0350 NM 015226 hypothetical protein L0C23274 KIAA0355 NM 014686 hypothetical protein L0C9761 KIAA0427 NM 014772 hypothetical protein L0C9811 KIAA0446 NM 014655 hypothetical protein L0C93 KIAA0467 NM_015284 h iA l protein KIAA0494 NM 014774 hypothetical protein L0C9813 KIAA0495 NM 207306 h IAlon5 KIAA0513 NM 014732 hypothetical protein L0C964 KIAA0514 NM 014696 hypothetical protein L0C2 i KIAA0523 NM 015253 hypothetical protein L0C23302 KIAA0553 NM0127 hypotheticalproteinLOC2 KIAA0644 NM 014817 hypothetical protein L0C98 KIAA0652 NM 014741 h icalo t KIAA0676 NM 015043 hypothetical protein L0C23178 KIAA0701 NM 001006947 hypothetical protein LC230 KIAA0703 NM 014861 galcelltantier pse 14 KIAA0738 NM 014719 hypothetical protein L0C9747 KIAA0773 NM 001031690 hepatoeucarin anign12 |KIAA0789 NM 01 465 3 hpteia rti 97 KIAA0804 NM 001009921 hypothetical protein L0C23355 isoform a KIAA0831 NM 014924 hypothetical protein L0C2863 KIAA0889 NM 027 hypothetical protein LC55722 KIAA0892 NM 015329 hypotheticalproteinLOC23 KIAA1008 NM 014953 lIk h f r e u KIAA1012 NM_0244 hypothetical protein L0C14973 KIAA1024 NM 015206 hypothetical protein L0C2321 KIAA1128 NM 018999 gauecl nieu oiie1 KIAA1161 NM 020702 hypothetical protein L0C5762 KIAA1166 NM 018684 eaoellrcrioaasctdangn12 KIAA1189 NM 001009959 hypothetical protein L0C5747 KIAA1267 NM 015443 hypothetical protein L0C28405 KIAA1274 NM_014431 KA17 KIA-A1328 NM_020776hyohtclptenLC73 KIAA1333 NM 017769hyohtclptenLC53 KIAA1446 NM 020836lieyotooofrtbaneicd uyae KIAA1456 NM 020844 hetik a t protein isof7rm4 KIAA1467 NM 020853hyohtclptenLC71 KIAA1522 NM 020888hyohtclptenLC74 KIAA1576 NM 020927hyohtclptenLC78 KIAA104 00943 hypothetical protein LOC23306 KIAA1622 NM 020958 HpoTleetia -oann protein LOC9895 KIAA1641 NM 020970 hypothetical protein LOC57730 -K47 - WO 2008/036718 PCT/US2007/078859 KIAA1706 NM 030636 hypothetical protein LOC80820 KIAA1715 NM 030650 Lunapark KIAA1727 NM_033393 hypothetical protein LOC85462 KIAA1729 NM_053042 hypothetical protein LOC85460 KIAA1737 NM_033426 KIAA1737 protein KIAA1853 NM_194286 KIAA1853 protein KIAA1875 NM 032529 KIAA1875 protein KIAA1909 NM_052909 hypothetical protein LOC153478 KIAA1914 NM_001001936 KIAA1914 protein isoform I KIAA1920 NM_052919 hypothetical protein LOCI 14817 KIAA2022 NM_001008537 hypothetical protein LOC340533 KIFIB NM_015074 kinesin family member lB isoform b KIF3B NM_004798 kinesin family member 3B KIF3C NM_002254 kinesin family member 3C KIF4A NM_012310 kinesin family member 4 KIF9 NM 022342 kinesin family member 9 isoform 1 KIRREL NM 018240 kin ofIRRE like KLC2 NM 022822 likely ortholog of kinesin light chain 2 KLC3 NM_177417 kinesin light chain 3 KLF12 NM_007249 Kruppel-like factor 12 isoform a KLF13 NM_015995 Kruppel-like factor 13 KLF17 NM_173484 zinc finger protein 393 KLF5 NM_001730 Kruppel-like factor 5 KLHDC6 NM 207335 hypothetical protein LOC166348 KLHL20 NM 014458 kelch-like 20 KLHL21 NM 014851 kelch-like 21 KLHL22 NM 032775 kelch-like KLHL24 NM 017644 DREl protein KLHL25 NM_022480 BTB/POZ KELCH domain protein KLHL26 NM_018316 hypothetical protein LOC55295 KLHL6 NM_130446 kelch-like 6 KLHL7 NM_001031710 SBBI26 protein isoform 1 KLK13 NM_015596 kallikrein 13 precursor KLK5 NM 012427 kallikrein 5 preproprotein KLRG1 NM 005810 killer cell lectin-like receptor subfamily G, KM-HN-1 NM_152775 KM-HN-1 protein KNDC1 NM_152643 kinase non-catalytic C-lobe domain (KIND) KPNA1 NM_002264 karyopherin alpha 1 KPNA6 NM_012316 karyopherin alpha 6 KRAS NM_004985 c-K-ras2 protein isoform b KREMEN2 NM_024507 kringle-containing transmembrane protein 2 KRIT1 NM_001013406 krev interaction trapped 1 isoform 2 KRT25A NM 181534 keratin25A KRT2A NM 000423 keratin 2a KRT2B NM 015848 cytokeratin 2 KRT4 NM 002272 keratin 4 KRTAP1-1 NM_030967 keratin associated protein 1-1 KRTAP4-14 NM_033059 keratin associated protein 4-14 KRTAP4-4 NM 032524 keratin associated protein 4.4 KRTAP9-2 NM 031961 keratin associated protein 9.2 KRTAP9-3 NM_031962 keratin associated protein 9.3 L3MBTL4 NM 173464 hypothetical protein LOC91133 LACEl NM 145315 lactation elevated 1 LAMB3 NM 000228 laminin subunit beta 3 precursor - 48 - WO 2008/036718 PCT/US2007/078859 LAMCl NM 002293 laminin, gamma 1 precursor LANCL2 NM 018697 LanC lantibiotic synthetase component C-like 2 LARP1 NM 015315 la related protein isoform 1 LARP4 NM 052879 c-Mpl binding protein isoform a LARPS NM 015155 La ribonucleoprotein domain family, member 5 LASP1 NM 006148 LIM and SH3 protein 1 LASS3 NM_178842 hypothetical protein LOC204219 LBH NM 030915 hypothetical protein DKFZp566J091 LCT NM 002299 lactase-phlorizin hydrolase preproprotein LDB3 NM 007078 LIM domain binding 3 LDLR NM 000527 low density lipoprotein receptor precursor LDLRAP1 NM 015627 low density lipoprotein receptor adaptor protein LDOC1L NM 032287 hypothetical protein LOC84247 LECT2 NM 002302 leukocyte cell-derived chemotaxin 2 precursor LENEP NM 018655 lens epithelial protein LEREPO4 NM 018471 crythropoietin 4 immediate early response LETMI NM 012318 leucine zipper-EF-hand containing transmembrane LGALS8 NM 006499 galectin 8 isoform a LHFPL2 NM 005779 lipoma HMGIC fusion partner-like 2 LHFPL3 NM 199000 lipoma HMGIC fusion partner-like 3 LHFPL5 NM 182548 lipoma HMGIC fusion partner-like 5 LHX3 NM 014564 LIM homeobox protein 3 isoform b LHX4 NM 033343 LIM homeobox protein 4 LIAS NM 006859 lipoic acid synthetase isoform 1 precursor LIF NM 002309 leukemia inhibitory factor (cholinergic LIFR NM 002310 leukemia inhibitory factor receptor precursor LILRB 1 NM 006669 leukocyte immunoglobulin-like receptor, LILRB4 NM 006847 leukocyte immunoglobulin-like receptor, LIMDl NM 014240 LIM domains containing 1 LIMD2 NM 030576 LIM domain containing 2 LIMK1 NM 002314 LIM domain kinase 1 LIMK2 NM 005569 LIM domain kinase 2 isoform 2a LIMS2 NM 017980 LIM and senescent cell antigen-like domains 2 LIMS3 NM 033514 LIM and senescent cell antigen-like domains 3 LIN28 NM 024674 lin-28 homolog LIN9 NM_173083 lin-9 homolog LIXI NM 153234 limb expression I LLGL1 NM 004140 lethal giant larvae homolog 1 LMNB2 NM_032737 lamin B2 LMO4 NM 006769 LIM domain only 4 LMO7 NM 005358 LIM domain only 7 LMOD3 NM 198271 leiomodin 3 (fetal) LOC116236 NM_198147 hypothetical protein LOC1 16236 LOC124491 NM 145254 hypothetical protein LOC124491 LOC129138 NM 138797 hypothetical protein LOC129138 LOC129607 NM 207315 thymidylate kinase family LPS-inducible member LOC130576 NM 177964 hypothetical protein LOC 30576 LOC133619 NM 130809 hypothetical protein LOC133619 LOC144501 N M 182507 hypothetical protein LOC144501 LOCI 51194 NM 145280 hypothetical protein LOC 151194 LOC152485 NM 178835 hypothetical protein LOC152485 LOC153561 NM 207331 hypothetical protein LOC153561 LOC158318 NM 001024608 hypothetical protein LOC158318 LOC162427 NM_178126 hypothetical protein LOC162427 - 49 - WO 2008/036718 PCT/US2007/078859 LOC196463 NM 173542 hypothetical protein L0C196463 LOC196752 NM 001010864 pothetical protein LOG 196752 LOC197322 NM_174917 hypothetical protein LOC197322 LOC201164 NM_178836 hypothetical protein LOC201164 LOC203427 NM 145305 mitochondrial solute carrier protein LOC221091 NM 203422 hypothetical protein T0C221091 LOC222967 NM 173565 hypothetical protein L0C222967 LOC283219 NM 001029859 hypothetical protein L0C283219 LOC283537 NM_181785 hypotheticalproteilOC283537 LOC283551 NM 001012706 hypothetical protein L0C283551 LOC284296 NM 175908 hypothetical protein L0C284296 LOC284434 NM 001007525 hypothetical protein L0C284434 LOC284757 NM 001004305 hypothetical protein L0C284757 LOC286076 NM 001024610 hypothetical protein L0C286076 LOC339524 NM 207357 hypothetical protein L0C339524 LOC340156 NM 001012418 hypothetical protein L0C340156 LOC342897 NM 001001414 similar to F-box only protein 2 LOC345222 NM_001012982 hypothetical protein L0C345222 LOC348262 NM 207368 hypothetical protein L0C348262 LOC387856 NM 001013635 hypothetical protein L0C387856 LOC388503 NM_001013640 hypothetical protein L0C388503 LOC389118 NM 001007540 hypothetical protein L0C389118 LOC389199 NM 203423 hypothetical protein L0C389199 LOC389791 NM 001013652 hypothetical protein L0C389791 LOC389834 NM 001013655 pothetical protein L0C389834 LOC392395 NM 001013664 hypothetical proteinL0G392395 LOC399706 NM 001010910 pothetical protein L0C399706 LOC399898 NM 001013666 hypothetical protein L0C399898 LOC400145 NM 001013669 hypothetical protein L0C400145 LOC400499 NM 001013671 hypothetical protein L0C400499 LOC400657 NM 001008234 pothetical protein L0C400657 LOC400891 NM 001013675 hypothetical protein L0C400891 LOC400924 NM 001013676 hypothetical protein L0C400924 LOC400965 NM_001013677 hypothetical protein L0C400965 LOC401137 NM 214711 hypothetical protein LOC401137 LOC401398 NM 001023566 hypothetical protein L0C401398 LC4013- NM 001008 745 hypothetical protein LOC401431 L0C40150 NM_001012278 hypothetical protein LOC401507 L0C401589 NM_001013687 hypothetical protein LOC401589 L0C401520 NM 001013688 hypothetical protein LOC401620 OC40172-0 NM_001013690 hypothetical protein L pC401720 L0C440313 NM 001013704 hypothetical protein LOC440313 L0C440337 NM 001013705 hypothetical protein LOC440337 LC440570 NM_00 10 13708 hypothetical protein LOC440570 L0C440742 NM 001013710 hypothetical protein LOC440742 LO-C440925 NM 001013712 hypothetical protein LOC440925 L0C440944 NM 001013713 hypothetical protein LOC440944 L0C441070 NM 001013715 hypothetical protein LOC441070 LO-C441136 NM_001013719 hypothetical protein LOC441136 L0C441268 NM 001013725 hypothetical protein LOC441268 L0C441459 NM_001013728 hypothetical protein LOC441459 L0C442247 NM 001013734 hypothetical protein LOC442247 L0C504189 NM 001013404 hypothetical protein LOC504188 L -C54103 NM 017439 hypothetical protein LOC54103 -50- WO 2008/036718 PCT/US2007/078859 LOC541473 NM 001013748 FKBP6-like LOC554251 NM 001024680 hypothetical protein LOC554251 LOC55908 NM_018687 hepatocellular carcinoma-associated gene TD26 LOC613206 NM 001033016 myeloproliferative disease associated tumor LOC613266 NM 001033516 hypothetical protein LOC613266 LOC63928 NM 022097 hepatocellular carcinoma antigen gene 520 LOC90167 NM_194277 hypothetical protein LOC90167 LOC90639 NM 001031617 hypothetical protein LOC90639 LOH12CR1 NM 058169 LOH1CR12 LOXL4 NM 032211 lysyl oxidase-like 4 precursor LPIN3 NM 022896 lipin 3 LPP NM 005578 LIM domain containing preferred translocation LRAT NM_004744 lecithin retinol acyltransferase LRBA NM_006726 LPS-responsive vesicle trafficking, beach and LRCH4 NM_002319 leucine-rich repeats and calponin homology (CH) LRPI11 NM_032832 low density lipoprotein receptor-related protein LRP12 NM 013437 suppression of tumorigenicity LRP2BP NM 018409 LRP2 binding protein LRRC14 NM_014665 leucine rich repeat containing 14 LRRC2 NM 024512 leucine rich repeat containing 2 LRRC20 NM 018205 leucine rich repeat containing 20 isoform 3 LRRC27 NM_030626 leucine rich repeat containing 27 LRRC3B NM_052953 leucine rich repeat containing 3B LRRC48 NM_031294 leucine rich repeat containing 48 LRRC54 NM 015516 tsukushi LRRIQ2 NM_024548 leucine-rich repeats and IQ motif containing 2 LRRN5 NM_006338 leucine rich repeat neuronal 5 precursor LRRTM3 NM_178011 leucine rich repeat transmembrane neuronal 3 LSM12 NM_152344 hypothetical protein LOC124801 LSM16 NM_025083 LSM16 homolog (EDC3, S. cerevisiae) LTBP2 NM 000428 latent transforming growth factor beta binding LUZP1 NM 033631 leucine zipper protein 1 LY6H NM 002347 lymphocyte antigen 6 complex, locus H LY86 NM 004271 MD-1, RP105-associated LYCAT NM_001002257 lysocardiolipin acyltransferase isoform 2 LYPLA3 NM 012320 lysophospholipase 3 (lysosomal phospholipase LYSMD1 NM_212551 LysM, putative peptidoglycan-binding, domain LYSMD4 NM_152449 hypothetical protein LOC145748 LYZ NM 000239 lysozyme precursor LZTR2 NM 033127 regucalcin gene promotor region related protein LZTS1 NM 021020 leucine zipper, putative tumor suppressor 1 M6PR NM_002355 cation-dependent mannose-6-phosphate receptor M6PRBP1 NM_005817 mannose 6 phosphate receptor binding protein 1 MAB2lL1 NM_005584 mab-21-like protein 1 MAF NM_001031804 v-mafmusculoaponeurotic fibrosarcoma oncogene MAGEA8 NM_005364 melanoma antigen family A, 8 MAGEA9 NM_005365 melanoma antigen family A, 9 MAGEL2 NM_019066 MAGE-like protein 2 MAGI2 NM_012301 membrane associated guanylate kinase, WW and PDZ MALL NM_005434 mal, T-cell differentiation protein-like MAN1C1 NM 020379 mannosidase, alpha, class 1C, member 1 MANEA NM 024641 mannosidase, endo-alpha MAPIB NM 005909 microtubule-associated protein 1B isoform I MAP3K3 NM 002401 mitogen-activated protein kinase kinase kinase 3 -51 - WO 2008/036718 PCT/US2007/078859 MAP3K7 NM_003188 mitogen-activated protein kinase kinase kinase 7 MAP4K1 NM_007181 mitogen-activated protein kinase kinase kinase MAPK1 NM_002745 mitogen-activated protein kinase 1 MAPK14 NM_001315 mitogen-activated protein kinase 14 isoform I MAPK3 NM 002746 mitogen-activated protein kinase 3 isoform 1 MAPK7 NM_002749 mitogen-activated protein kinase 7 isoform 1 MAPKAPK2 NM 004759 mitogen-activated protein kinase-activated MAPKBP1 NM 014994 mitogen-activated protein kinase binding protein MAPT NM_005910 microtubule-associated protein tau isoform 2 MARCH3 NM 178450 membrane-associated ring finger (C3HC4) 3 MARCH5 NM_017824 ring finger protein 153 MARCKS NM_002356 myristoylated alanine-rich protein kinase C MARK3 NM_002376 MAP/microtubule affinity-regulating kinase 3 MARVELDI NM_031484 MARVEL domain containing 1 MARVELD3 NM_052858 MARVEL domain containing 3 isoform 2 MASI NM_002377 MASI oncogene MASPI NM_001879 mannan-binding lectin serme protease 1 isoform MATlA NM_000429 methionine adenosyltransferase I, alpha MATN2 NM_002380 matrilin 2 isoform a precursor MBD3 NM_003926 methyl-CpG binding domain protein 3 MBNL3 NM 018388 muscleblind-like 3 isoform G MCART6 NM 001012755 hypothetical protein LOC401612 MCCC2 NM 022132 methylcrotonoyl-Coenzyme A carboxylase 2 (beta) MCF2 NM 005369 MCF.2 cell line derived transforming sequence MCFD2 NM_139279 multiple coagulation factor deficiency 2 MCL1 NM 021960 myeloid cell leukemia sequence 1 isoform 1 MCM4 NM 005914 minichromosome maintenance protein 4 MCM8 NM_032485 minichromosome maintenance protein 8 isoform 1 MDFIC NM_199072 MyoD family inhibitor domain containing isoform MDGAl NM 153487 MAM domain containing MECP2 NM 004992 methyl CpG binding protein 2 MED12L NM 053002 mediator of RNA polymerase II transcription, MEF2C NM 002397 MADS box transcription enhancer factor 2, MEF2D NM 005920 MADS box transcription enhancer factor 2, MEGF1O NM 032446 MEGF10 protein MEP1A NM 005588 meprin A, alpha (PABA peptide hydrolase) METT5Dl NM 152636 methyltransferase 5 domain containing 1 METTL5 NM_014168 methyltransferase like 5 MFAP3 NM_005927 microfibrillar-associated protein 3 MFI2 NM 033316 melanoma-associated antigen p97 isoform 2, MFN2 NM 014874 mitofusin 2 MFSD4 NM 181644 hypothetical protein DKFZp76 lN1 114 MGAM NM 004668 maltase-glucoamylase MGC10334 NM 001029885 hypothetical protein LOC80772 MGC 11102 NM 032325 hypothetical protein LOC84285 MGC13379 NM 016499 hypothetical protein LOC51259 MGC15875 NM 032921 hypothetical protein LOC85007 isoform 1 MGC16028 NM 052873 hypothetical protein LOC1 12752 MGC16703 NM 145042 hypothetical protein LOC1 13691 MGC20470 NM 145053 hypothetical protein LOC143630 MGC23280 NM 144683 hypothetical protein LOC147015 MGC24039 NM 144973 hypothetical protein LOC160518 MGC26694 NM_178526 hypothetical protein LOC284439 MGC26718 NM_001029999 hypothetical protein LOC440482 - 52 - WO 2008/036718 PCT/US2007/078859 MGC26733 NM_144992 hypothetical protein LOC200403 MGC27121 NM 001001343 hypothetical protein LOC408263 MGC2752 NM_023939 hypothetical protein LOC65996 MGC29891 NM_144618 GA repeat binding protein, beta 2 MGC29898 NM_145048 hypothetical protein LOC133015 MGC3207 NM_001031727 hypothetical protein LOC84245 isoform 1 MGC33214 NM_153354 hypothetical protein LOC153396 MGC33530 NM_182546 hypothetical protein LOC222008 MGC34646 NM_173519 hypothetical protein LOC157807 MGC35295 NM_152717 hypothetical protein LOC219995 MGC39900 NM_194324 hypothetical protein LOC286527 MGC4562 NM 133375 hypothetical protein LOCI 15752 MGC4655 NM_033309 hypothetical protein LOC84752 MGC50273 NM 214461 hypothetical protein LOC408029 MGC9712 NM_152689 hypothetical protein LOC202915 MGLL NM_001003794 monoglyceride lipase isoform 2 MIBl NM_020774 mindbomb homolog 1 MICAL2 NM_014632 microtubule associated monoxygenase, calponin MICAL-Ll NM_033386 molecule interacting with Rabl3 MID1IP1 NM_021242 MIDI interacting G12-like protein MIER3 NM_152622 hypothetical protein LOC166968 MIPOL1 NM 138731 mirror-image polydactyly I MKLl NM_020831 megakaryoblastic leukemia 1 protein MKL2 NM_014048 megakaryoblastic leukemia 2 protein MKLN1 NM_013255 muskelin 1, intracellular mediator containing MKRN3 NM_005664 makorin, ring finger protein, 3 MLC1 NM 015166 megalencephalic leukoencephalopathy with MLL4 NM 014727 myeloid/lymphoid or mixed-lineage leukemia 4 MLLT3 NM 004529 myeloid/lymphoid or mixed-lineage leukemia MLSTD2 NM_032228 male sterility domain containing 2 MLX NM_170607 transcription factor-like protein 4 isoform MLXIPL NM 032951 Williams Beuren syndrome chromosome region 14 MMD2 NM_198403 monocyte-to-macrophage differentiation factor 2 MMP14 NM 004995 matrix metalloproteinase 14 preproprotein MMP17 NM_016155 matrix metalloproteinase 17 preproprotein MMP19 NM_001032360 matrix metalloproteinase 19 isoform 2 precursor MMP2 NM_004530 matrix metalloproteinase 2 preproprotein MMP8 NM_002424 matrix metalloproteinase 8 preproprotein MN1 NM 002430 meningioma 1 MOBKL2A NM 130807 MOB-LAK MOBKL2B NM 024761 MOB 1, Mps One Binder kinase activator-like 2B MOCS1 NM 005942 molybdenum cofactor synthesis-step 1 protein MOCS2 NM 176806 molybdopterin synthase small subunit MOCS2A MOG NM 001008228 myelin oligodendrocyte glycoprotein isoform MON1B NM_014940 MONI homolog B MOSPD1 NM_019556 motile sperm domain containing 1 MPP2 NM_005374 palmitoylated membrane protein 2 MPPED1 NM_001585 hypothetical protein LOC758 MPST NM_001013436 3-mercaptopyruvate sulfurtransferase MRAS NM 012219 muscle RAS oncogene homolog MRO NM 031939 maestro MRP63 NM 024026 mitochondrial ribosomal protein 63 MRPL30 NM 145212 mitochondrial ribosomal protein L30 MRPL41 NM_032477 mitochondrial ribosomal protein L41 - 53 - WO 2008/036718 PCT/US2007/078859 MRPL52 NM_178336 mitochondrial ribosomal protein L52 isoform a MRPS 11 NM 022839 mitochondrial ribosomal protein S 11 isoform a MRPS26 NM 030811 mitochondrial ribosomal protein S26 MRPS33 NM 016071 mitochondrial ribosomal protein S33 MS4A10 NM 206893 membrane-spanning 4-domains, subfamily A, member MS4A2 NM 000139 membrane-spanning 4-domains, subfamily A, member MS4A4A NM 024021 membrane-spanning 4-domains, subfamily A, member MS4A7 NM 021201 membrane-spanning 4-domains, subfamily A, member MSH3 NM 002439 mutS homolog 3 MSI2 NM_138962 musashi 2 isoform a MSL3L1 NM 078628 male-specific lethal 3-like 1 isoform d MSR1 NM 002445 macrophage scavenger receptor 1 isoform type 2 MSRB3 NM 001031679 methionine sulfoxide reductase B3 isoform 2 MTAC2DL NM 152332 membrane targeting (tandem) C2 domain containing MTHFR NM 005957 5,1 0-methylenetetrahydrofolate reductase MTHFSD NM 022764 hypothetical protein LOC64779 MTMI NM_000252 myotubularin MTMR12 NM 019061 myotubularin related protein 12 MTMR2 NM 016156 myotubularin-related protein 2 isoform 1 MTMR3 NM 021090 myotubularin-related protein 3 isoform c MTMR9 NM 015458 myotubularin-related protein 9 MTPN NM 145808 myotrophin MTRR NM 002454 methionine synthase reductase isoform 1 MUCDHL NM 031265 mu-protocadherin isoform 4 MUMILl NM 152423 melanoma associated antigen (mutated) 1-like 1 MUTED NM 201280 muted MX2 NM_002463 myxovirus resistance protein 2 MXDI NM 002357 MAX dimerization protein 1 MXD4 NM_006454 MAD4 MYADM NM 001020818 myeloid-associated differentiation marker MYBBP1A NM 014520 MYB binding protein Is MYBL2 NM 002466 MYB-related protein B MYCLI NM 001033081 -myc-I proto-oncogeus isoform 1 MYD88 NM 002468 mycloid differentiation primary response gene MYL2 NM 000432 myosin light chain 2 MYL3 NM 000258 myosin light chain 3 MYO18A NM 078471 myosin 18A isoform a MYO1B NM 012223 myosin IB MYO1E NM 004998 myosin IE MYO3A NM_017433 myosin IIIA MYO5C NM 018728 myosinVC MYO6 NM 004999 myosin VI MYO7A NM 000260 myosin VIIA MYOM2 NM 003970 myomesin 2 MYST2 NM 007067 MYST histone acetyltransferase 2 MYST3 NM 006766 MYST histone acetyltransferase (monocytic MYT1L NM 015025 myelin transcription factor 1-like N4BP1 NM_153029 Nedd4 binding protein 1 NAALADL2 NM_207015 N-acetylated alpha-linked acidic dipeptidase 2 NAG6 NM 022742 hypothetical protein DKFZp434G156 NAG8 NM_014411 nasopharyngeal carcinoma associated gene NALPI NM 014922 death effector filament-forming Ced-4-like NALP12 NM 144687 PYRIN-containing APAFI-like protein 7 isoform 2 NANOSI NM 199461 nanos homolog I isoform 1 - 54 - WO 2008/036718 PCT/US2007/078859 NANP NM 152667 haloacid dehalogenase-like hydrolase domain NAP1L4 NM 005969 nucleosome assembly protein 1-like 4 NAPE-PLD NM 198990 N-aeyl-phosphatidylethanolamine-hydrolyzing NARGI NM 057175 NMDA receptor regulated 1 NARGIL NM 024561 NMDA receptor regulated 1-like protein iaoform NARG2 NM_001018089 NMA receptor regulated 2 iaoform b NAT10 NM 024662 N-acetyltransfersse-like protein NAT12 NM_001011713 hypothetical protein L0C122830 NAV3 NM 014903 neuron navigator 3 NCAM1 NM_181351 neural cell adhesion molecule I isoform 2 NCOAl NM 003743 nuclear receptor coactivator 1 isoform 1 NCOA6IP NM_024831 PRIP-interacting protein PIPMT NCOA7 NM 181782 nuclear receptor coactivator 7 NCR1 NM 004829 natural cytotoxicity triggering receptor 1 NCSTN NM 015331 nicaatrin precursor NDE1 NM_017668 nuclear distribution gene E homolog 1 NDEL1 NM 001025579 nudE nuclear distribution gene E homolog like 1 NDFIP1 NM_030571 Nedd4 family interacting protein 1 NDRG4 NM 020465 NDRG family member 4 NDSTl NM_001543 N-deacetylase/N-aulfotransferase (heparan NEBL NM 006393 nebulette aarcomeric isoform NECAPI NM 015509 adaptin-ear-binding coat-associated protein 1 NECAP2 NM 018090 adaptin-ear-binding coat-associated protein 2 NEDD4 NM 006154 neural precursor cell expressed, developmentally NEDD9 NM 182966 neual precursor cell expressed, developmentally NEIL2 NM 145043 nes-like 2 NEK8 NM 178170 NIMA-related kinase 8 NES NM_006617 nesting NETOl NM_138999 neuropilin- and tolloid-like protein I isoform 1 NETO2 NM_018092 neuropilin- and tolloid-like protein 2 NEURL NM 004210 neuralized-like NEUROG2 NM 024019 neurogenin 2 NF2 NM 000268 neurofibromin 2 isoform 1 NFAM1 NM 145912 NFAT activation molecule 1 precursor NFASC NM 015090 neurofascin precursor NFAT5 NM 006599 nuclear factor of activated T-cells 5 isoform c NFATC1 NM_006162 nuclear factor of activated T-cells, cytosolic NFIC NM 005597 nuclear factor I/C isoform 1I NFKBIL1 NM 005007 nuclear factor of kappa light polypeptide gene N XL1 NM 152995 nuclear transcription factor, X-box binding-like NFYA NM 002505 nuclear transcription factor Y, alpha isoform I N YB NM 006166 nuclear transcription factorY, beta NGFR NM_002507 nerve growth factor receptor precursor N HLH1 NM_005598 nescient helix loop helix 1 NIPAl NMP144599 non-imprinted in Prader-Willi/Angeman syndrome NIPSNAP1 NM_003634 nipanap homolog 1 NXIRAS2 NM 001001349 NFKB inhibitor interacting Ras-like 2 N TR NM 001012651 natural killer-tumor recognition sequence NLGN2 NM 020795 neuroligin 2 NMNATI NM 022787 nicotinamide nucleotide adenylyltrangferase 1 NMTl NM 021079 N-myristoyltransferase I NMT2 N N004808 glycylpeptide N-tetradecanoyltransferase 2 NNAT NM 005386 neuronatin isoform alpha NOBI 014062 nin one binding protein - 55 - WO 2008/036718 PCT/US2007/078859 NOLl 1 NM 015462 nucleolar protein 11 NOL6 NM 022917 nucleolar RNA-associated protein alpha isoform NOMI NM_138400 nucleolar protein with MIF4G domain 1 NOVAl NM 002515 neuro-oncological ventral antigen 1 isoform 1 NOXI NM 007052 NADPH oxidase 1 isoform long NPAL3 NM_020448 NIPA-like domain containing 3 NPAS2 NM 002518 neuronal PAS domain protein 2 NPCl NM 000271 Niemann-Pick disease, type C1 NPHPl NM 000272 nephrocystin isoform 1 NPLOC4 NM_017921 nuclear protein localization 4 NPR3 NM 000908 natriuretic peptide receptor C/guanylate cyclase NPTX1 NM 002522 neuronal pentraxin I precursor NPTXR NM_014293 neuronal pentraxin receptor isoform 1 NQOI NM_000903 NAD(P)H menadione oxidoreductase 1, NR3C1 NM 000176 nuclear receptor subfamily 3, group C, member 1 NRG1 NM_013958 neuregulin 1 isoform HRG-beta3 NRIPI NM_003489 receptor interacting protein 140 NRIP2 NM_031474 nuclear receptor interacting protein 2 NRP2 NM_003872 neuropilin 2 isoform 2 precursor NSF NM_006178 N-ethylmaleimide-sensitive factor NT5C2 NM 012229 5'-nucleotidase, cytosolic II NTRK2 NM_001007097 neurotrophic tyrosine kinase, receptor, type 2 NUAK2 NM_030952 NUAK family, SNF1-like kinase, 2 NUCB1 NM 006184 nucleobindin 1 NUDTIO NM_153183 nudix-type motif 10 NUDT12 NM_031438 nudix -type motif 12 NUDT15 NM_018283 nudix-type motif 15 NUTDT16 NM 152395 nudix-type motif 16 NUDT16L1 NM 032349 syndesmos NUDT18 NM 024815 nudix (nucleoside diphosphate linked moiety NUDT4 NM 019094 nudix-type motif 4 isoform alpha NUMB NM_001005743 numb homolog isoform 1 NUMBL NM_004756 numb homolog (Drosophila)-like NUP35 NM_001008544 nucleoporin 35kDa isoform b NUP43 NM_198887 nucleoporin 43kDa NXF1 NM_006362 nuclear RNA export factor 1 NYD-SP18 NM 032599 testes development-related NYD-SP18 NY-REN-7 NM 173663 hypothetical protein LOC285596 OACT2 NM 138799 O-acyltransferase (membrane bound) domain OACT5 NM_005768 gene rich cluster, C3f gene OAF NM 178507 hypothetical protein LOC220323 OAS3 NM 006187 2'-5'oligoadenylate synthetase 3 OAZI NM_004152 omithine decarboxylase antizyme 1 OBFC2B NM_024068 hypothetical protein LOC79035 OCRL NM_000276 phosphatidylinositol polyphosphate 5-phosphatase OLIGI NM_138983 oligodendrocyte transcription factor 1 OPCML NM_001012393 opioid binding protein/cell adhesion OPRD1 NM_000911 opioid receptor, delta I OPTC NM 014359 opticin precursor OR2Hl NM 030883 olfactory receptor, family 2, subfamily H, OR51E2 NM 030774 olfactory receptor, family 51, subfamily E, OR7D2 NM 175883 hypothetical protein LOC162998 ORAOV1 NM 153451 oral cancer overexpressed I ORC2L NM_006190 origin recognition complex, subunit 2 -56- WO 2008/036718 PCT/US2007/078859 OSBP2 NM_ 030758 oxysterol binding protein 2 isoform a OSBPL2 NM 014835 oxysterol-binding protein-like protein 2 isoform OSBPL3 NM 015550 oxysterol-binding protein-like protein 3 isoform OSBPL7 NM 145798 oxysterol-binding protein-like protein 7 OSCAR NM 206817 osteoclast-associated receptor isoform 2 OTUD4 NM 199324 OTU domain containing 4 protein isoform 1 OTUD6B NM_016023 OTU domain containing 6B OXGR1 NM 080818 oxoglutarate (alpha-ketoglutarate) receptor 1 P2RX2 NM_012226 purinergic receptor P2X2 isoform I P2RX7 NM_002562 purinergic receptor P2X7 P2RY13 NM 023914 purinergic receptor P2Y, G-protein coupled, 13 P2RYl4 NM 014879 purinergic receptor P2Y, G-protein coupled, 14 P2RY4 NM 002565 pyrimidinergic receptor P2Y4 P2RY8 NM 178129 G-protein coupled purinergic receptor P2Y8 P4HAl NM 000917 prolyl 4-hydroxylase, alpha I subunit isoform 1 P4HA3 NM 182904 prolyl 4-hydroxylase, alpha III subunit P53AIP1 NM_022112 p53-regulated apoptosis-inducing protein 1 PACRG NM 152410 PARK2 co-regulated PACSI NM_018026 phosphofurin acidic cluster sorting protein I PAFAH1B2 NM 002572 platelet-activating factor acetylhydrolase, PAG1 NM 018440 phosphoprotein associated with glycosphingolipid PAICS NM_006452 phosphoribosylaminoimidazole carboxylase PALMD NM 017734 palmdelphin PAN3 NM 175854 PABPl-dependent poly A-specific ribonuclease PAP2D NM_001010861 phosphatidic acid phosphatase type 2d isoform 2 PAPLN NM 173462 papilin PAPOLB NM 020144 poly(A) polymerase beta (testis specific) PAPPA NM _002581 pregnancy-associated plasma protein A PAQR5 NM_017705 membrane progestin receptor gamma PAQR6 NM_198406 progestin and adipoQ receptor family member VI PARD6G NM 032510 PAR-6 gamma protein PARP6 NM_020213 poly (ADP-ribose) polymerase family, member 6 PARVA NM 018222 parvin, alpha PATE NM_138294 expressed in prostate and testis PAX5 NM 016734 paired box 5 PBK NM 018492 T-LAK cell-originated protein kinase PC NM 000920 pyruvate carboxylase precursor PCDH 1lX NM 032967 protocadherin 11 X-linked isoform b precursor PCDH1 lY NM 032971 protocadherin 11 Y-linked isoform a PCDH21 NM_033100 protocadherin 21 precursor PCDHA9 NM 014005 protocadherin alpha 9 isoform 2 precursor PCDHB1O NM 018930 protocadherin beta 10 precursor PCGF3 NM_006315 ring finger protein 3 PCGF6 NM_001011663 polycomb group ring finger 6 isoform a PCMT1 NM_005389 protein-L-isoaspartate (D-aspartate) PCNXL2 NM_014801 pecanex-like 2 PCQAP NM_001003891 positive cofactor 2, glutamine/Q-rich-associated PCSK2 NM_002594 proprotein convertase subtilisin/kexin type 2 PCSK6 NM_138323 paired basic amino acid cleaving system 4 PCSK7 NM_004716 proprotein convertase subtilisin/kexin type 7 PCSK9 NM 174936 proprotein convertase subtilisin/kexin type 9 PCYOX1 NM_016297 prenylcysteine oxidase 1 PDAP1 NM 014891 PDGFA associated protein 1 PDCD6IP NM_013374 programmed cell death 6 interacting protein - 57 - WO 2008/036718 PCT/US2007/078859 PDCL NM_005388 phosducin-like PDDCl NM_182612 hypothetical protein LOC347862 PDEllA NM_016953 phosphodiesterase 11A PDElB NM_000924 phosphodiesterase 1B, calmodulin-dependent PDE4DIP NM_001002811 phosphodiesterase 4D interacting protein isoform PDE5A NM_001083 phosphodiesterase 5A isoform 1 PDE7A NM_002604 phosphodiesterase 7A isoform b PDE8B NM_001029851 phosphodiesterase 8B isoform 3 PDGFB NM 002608 platelet-derived growth factor beta isoform 1, PDGFRA NM 006206 platelet-derived growth factor receptor alpha PDGFRB NM_002609 platelet-derived growth factor receptor beta PDIA6 NM_005742 protein disulfide isomerase-associated 6 PDK1 NM_002610 pyruvate dehydrogenase kinase, isozyme 1 PDLIM2 NM_176871 PDZ and LIM domain 2 isoform 1 PDLIM5 NM_001011513 PDZ and LIM domain 5 isoform b PDP2 NM_020786 pyruvate dehydrogenase phosphatase isoenzyme 2 PDPK1 NM 002613 3-phosphoinositide dependent protein kinase-1 PDPR NM_017990 pyruvate dehydrogenase phosphatase regulatory PDXK NM_003681 pyridoxal kinase PDYN NM_024411 beta-neoendorphin-dynorphin preproprotein PDZD2 NM 178140 PDZ domain containing 2 PDZD4 NM_032512 PDZ domain containing 4 PEBP1 NM 002567 prostatic binding protein PECR NM_018441 peroxisomal trans-2-enoyl-CoA reductase PEG3 NM 006210 paternally expressed 3 PER2 NM 022817 period 2 isoform 1 PEX1O NM_002617 peroxisome biogenesis factor 10 isoform 2 PEX5 NM_000319 peroxisomal biogenesis factor 5 PFKFB2 NM_001018053 6-phosphofructo-2-kinase/fructose-2, PGAP1 NM_024989 GPI deacylase PGBD4 NM_152595 piggyBac transposable element derived 4 PGD NM_002631 phosphogluconate dehydrogenase PGK1 NM_000291 phosphoglycerate kinase 1 PGK2 NM 138733 phosphoglycerate kinase 2 PGLYRP2 NM 052890 peptidoglycan recognition protein L precursor PGLYRP4 NM_020393 peptidoglycan recognition protein-I-beta PGM2L1 NM_173582 phosphoglucomutase 2-like 1 PGRMC2 NM_006320 progesterone membrane binding protein PHC2 NM 004427 polyhomeotic 2-like isoform b PHIIll NM_016119 PHD finger protein 11 PHF13 NM_153812 PHD finger protein 13 PHF20 NM_016436 PHD finger protein 20 PHF20L1 NM 016018 PHD finger protein 20-like 1 isoform 1 PHF6 NM_001015877 PHD finger protein 6 isoform 1 PHF8 NM 015107 PHD finger protein 8 PHGDHL1 NM_177967 hypothetical protein LOC337867 PHLDB1 NM_015157 pleckstrin homology-like domain, family B, PHTF2 NM_020432 putative homeodomain transcription factor 2 PI4KII NM_018425 phosphatidylinositol 4-kinase type II PIAS3 NM_006099 protein inhibitor of activated STAT, 3 PIGQ NM_004204 phosphatidylinositol glycan, class Q isoform 2 PIGW NM 178517 phosphatidylinositol glycan, class W PIK3CG NM 002649 phosphoinositide-3-kinase, catalytic, gamma PIK3R1 NM_181504 phosphoinositide-3-kinase, regulatory subunit, - 58 - WO 2008/036718 PCT/US2007/078859 PIK3R3 NM _003629 phosphoinositide-3-kinase, regulatory subunit 3 PILRA NM_013439 paired immunoglobulin-like type 2 receptor alpha PIP3-E NM_015553 phosphoinositide-binding protein PIP3-E PIP5K1C NM_012398 phosphatidylinositol-4-phosphate 5-kinase, type PIP5K2B NM 003559 phosphatidylinositol-4-phosphate 5-kinase type PIP5KLl NM_173492 phosphatidylinositol-4-phosphate 5-kinase-like PITPNA NM 006224 phosphatidylinositol transfer protein, alpha PITX1 NM_002653 paired-like homeodomain transcription factor 1 PKD2 NM 000297 polycystin 2 PKNOX1 NM 004571 PBX/knotted I homeobox 1 isoform 1 PKP1 NM 000299 plakophilin 1 isoform lb PLA2G1B NM_000928 phospholipase A2, group IB PLA2G2D NM 012400 phospholipase A2, group IID PLA2G4D NM_178034 phospholipase A2, group IVD PLAGL2 NM_002657 pleiomorphic adenoma gene-like 2 PLAU NM_002658 urokinase plasminogen activator preproprotein PLAUR NM_001005376 plasminogen activator, urokinase receptor PLCB4 NM_000933 phospholipase C beta 4 isoform a PLCD3 NM_133373 phospholipase C delta 3 PLCXD3 NM 001005473 phosphatidylinositol-specific phospholipase C, X PLDS NM 152666 phospholipase D family, member 5 PLEKHA1 NM_001001974 pleckstrin homology domain containing, family A PLEKHA6 NM_014935 phosphoinositol 3-phosphate-binding protein-3 PLEKHB2 NM_ 017958 pleckstrin homology domain containing, family B PLEKHQ1 NM 025201 PH domain-containing protein PL1N NM 002666 perilipin PLXNAl NM 032242 plexin Al PML NM 033238 promyclocytic leukemia protein isoform 1 PNKfD NM 015488 myofibrillogenesis regulator 1 isoform 1 PNMA2 NM_007257 paraneoplastic antigen MA2 PNPLAl NM 173676 patatin-like phospholipase domain containing 1 PNPO NM 018129 pyridoxine 5'-phosphate oxidase PNRC 1 NM 006813 proline-rich nuclear receptor coactivator 1 PODXL NM 001018111 podocalyxin-like precursor isoform 1 POFUTI NM_015352 protein 0-fucosyltransferase 1 isoform 1 POFUT2 NM_015227 protein 0-fucosyltransferase 2 isoform A POGK NM 017542 pogo transposable element with KRAB domain POGZ NM 145796 pogo transposable element with ZNF domain POLDIP2 NM 015584 DNA polymerase delta interacting protein 2 POLDIP3 NM 032311 DNA polymerase delta interacting protein 3 POLH NM 006502 polymerase (DNA directed), eta POLRiB NM 019014 RNA polymerase I polypeptide B POLRlE NM 022490 RNA polymerase I associated factor 53 POLR2L NM 021128 DNA directed RNA polymerase It polypeptide L POLR3E NM 018119 polymerase (RNA) III (DNA directed) polypeptide POLR3GL NM 032305 polymerase (RNA) III (DNA directed) polypeptide POM121 NM 172020 nuclear pore membrane protein 121 POU2F2 NM 002698 POU domain, class 2, transcription factor 2 POU2F3 NM 014352 POU transcription factor PPAPDC2 NM 203453 phosphatidic acid phosphatase type 2 domain PPARA NM 001001928 peroxisome proliferative activated receptor, PPCDC NM 021823 phosphopantothenoylcysteine decarboxylase PPEF2 NM_152933 serine/threonine protein phosphatase with PPFIBP2 NM 003621 PTPRF interacting protein, binding protein 2 -59- WO 2008/036718 PCT/US2007/078859 PPIL2 NM_014337 peptidylprolyl isomerase-like 2 isoform a PPIL4 NM_139126 peptidylprolyl isomerase-like 4 PPL NM_002705 periplakin PPM1B NM 177968 protein phosphatase lB isoform 2 PPM1E NM_014906 protein phosphatase IE PPM2C NM 018444 pyruvate dehydrogenase phosphatase precursor PPP1R12B NM 002481 protein phosphatase 1, regulatory (inhibitor) PPP1R12C NM 017607 protein phosphatase 1, regulatory subunit 12C PPPIR13L NM 006663 protein phosphatase 1, regulatory (inhibitor) PPPIR15B NM 032833 protein phosphatase 1, regulatory subunit 15B PPPIR16B NM_015568 protein phosphatase 1 regulatory inhibitor PPP1R3A NM_002711 protein phosphatase 1 glycogen-binding PPPlR3B NM_024607 protein phosphatase 1, regulatory (inhibitor) PPP2CB NM_001009552 protein phosphatase 2, catalytic subunit, beta PPP2R1B NM_002716 beta isoform of regulatory subunit A, protein PPP2R2A NM_002717 alpha isoform of regulatory subunit B55, protein PPP2R3A NM_002718 protein phosphatase 2, regulatory subunit B", PPP2R4 NM_021131 protein phosphatase 2A, regulatory subunit B' PPP2R5C NM 002719 gamma isoform of regulatory subunit B56, protein PPP4R1L NM_018498 hypothetical protein LOC55370 PPT2 NM_005155 palmitoyl-protein thioesterase 2 isoform a PRCI NM_003981 protein regulator of cytokinesis 1 isoform 1 PRDM12 NM_021619 PR domain containing 12 PRDM16 NM_022114 PR domain containing 16 isoform 1 PRDM9 NM 020227 PR domain containing 9 PREB NM 013388 prolactin regulatory element binding protein PRELP NM_002725 proline arginine-rich end leucine-rich repeat PREPL NM_006036 prolyl endopeptidase-like PRICKLE2 NM_198859 prickle-like 2 PRKAA2 NM_006252 AMP-activated protein kinase alpha 2 catalytic PRKCA NM 002737 protein kinase C, alpha PRKCE NM_005400 protein kinase C, epsilon PRKD2 NM_016457 protein kinase D2 PRKRIP1 NM 024653 PRKR interacting protein 1 (ILl 1 inducible) PRKRIR NM 004705 protein-kinase, interferon-inducible double PRKX NM_005044 protein kinase, X-linked PRKY NM_002760 protein kinase, Y-linked PRND NM_012409 prion-like protein doppel preproprotein PROSC NM 007198 proline synthetase co-transcribed homolog PRPF19 NM_014502 PRP19/PSO4 pre-mRNA processing factor 19 PRPF4 NM_004697 PRP4 pre-mRNA processing factor 4 homolog PRRG4 NM_024081 proline rich Gla (G-carboxyglutamic acid) 4 PRRT2 NM_145239 hypothetical protein LOC1 12476 PRRXl NM_006902 paired mesoderm homeobox 1 isoform pmx-la PRSS23 NM_007173 protease, serine, 23 precursor PRX NM 020956 periaxin isoform 1 PRY NM_004676 PTPN13-like, Y-linked PRY2 NM_001002758 PTPN13-like, Y-linked 2 PSCD1 NM 004762 pleckstrin homology, Sec7 and coiled/coil PSCD4 NM_013385 pleckstrin homology, Sec7 and coiled/coil PSD3 NM 015310 ADP-ribosylation factor guanine nucleotide PSG4 NM 002780 pregnancy specific beta-1-glycoprotein 4 isoform PSG7 NM 002783 pregnancy specific beta-1-glycoprotein 7 PSMD5 NM_005047 proteasome 26S non-ATPase subunit 5 - 60 - WO 2008/036718 PCT/US2007/078859 PSME4 NM_014614 proteasome (prosome, macropain) activator PTAFR NM_000952 platelet-activating factor receptor PTCH NM 000264 patched PTDO04 NM 001011708 GTP-binding protein PTDO04 isoform 2 PTDSS2 NM 030783 phosphatidylserne synthase 2 PTGDR NM 000953 prostaglandin D2 receptor PTGER3 NM 198718 prostaglandin E receptor 3, subtype EP3 isoform PTGES2 NM_025072 prostaglandin E synthase 2 isoform 1 PTGES3 NM 006601 unactive progesterone receptor, 23 kD PTGIS NM_000961 prostaglandin 12 (prostacyclin) synthase PTHB1 NM_001033604 parathyroid hormone-responsive B1 isoform 3 PTK6 NM 005975 PTK6 protein tyrosine kinase 6 PTK7 NM_002821 PTK7 protein tyrosine kinase 7 isoform a PTK9L NM_007284 twinfilin-like protein PTPDC1 NM_152422 protein tyrosine phosphatase domain containing 1 PTPLB NM_198402 protein tyrosine phosphatase-like (proline PTPN1 1 NM_002834 protein tyrosine phosphatase, non-receptor type PTPN2 NM 002828 protein tyrosine phosphatase, non-receptor type PTPN23 NM 015466 protein tyrosine phosphatase, non-receptor type PTPN4 NM 002830 protein tyrosine phosphatase, non-receptor type PTPN7 NM 002832 protein tyrosine phosphatase, non-receptor type PTPRE NM_006504 protein tyrosine phosphatase, receptor type, E PTPRN NM_002846 protein tyrosine phosphatase, receptor type, N PTPRT NM_007050 protein tyrosine phosphatase, receptor type, T PTRF NM 012232 polymerase I and transcript release factor PTTG IIP NM_004339 pituitary tumor-transforming gene 1 PXMP4 NM 183397 peroxisomal membrane protein 4 isoform b PXT1 NM 152990 peroxisomal, testis specific 1 PYCRL NM 023078 pyrroline-5-carboxylate reductase-like QDPR NM 000320 quinoid dihydropteridine reductase QKI NM 006775 quaking homolog, KH domain RNA binding isoform QPCTL NM 017659 glutaminyl-peptide cyclotransferase-like QPRT NM_014298 quinolinate phosphoribosyltransferase QRSL1 NM_018292 glutaminyl-tRNA synthase QSCN6 NM 002826 quiescin Q6 isoform a QSCN6L1 NM_181701 quiescin Q6-like 1 RABI1A NM 004663 Ras-related protein Rab-1 1A RAB1IFIPi NM_001002814 Rab coupling protein isoform 3 RAB11FIP4 NM_032932 RAB 11 family interacting protein 4 (class II) RAB15 NM_198686 Ras-related protein Rab-15 RAB22A NM 020673 RAS-related protein RAB-22A RAB23 NM_016277 Ras-related protein Rab-23 RAB27A NM_004580 Ras-related protein Rab-27A RAB28 NM 001017979 RAB28, member RAS oncogene family isoform 1 RAB33B NM 031296 RAB33B, member RAS oncogene family RAB37 NM 001006638 RAB37, member RAS oncogene family isoform 2 RAB40B NM_006822 RAB40B, member RAS oncogene family RAB40C NM 021168 RAR (RAS like GTPASE) like RAB41 NM_001032726 RAB41, member RAS homolog family RAB43 NM 198490 RAB43 protein RAB6B NM 016577 RAB6B, member RAS oncogene family RAB6IP2 NM_015064 RAB6-interacting protein 2 isoform alpha RAB7 NM_004637 RAB7, member RAS oncogene family RAB7L1 NM_003929 RAB7, member RAS oncogene family-like 1 -61 - WO 2008/036718 PCT/US2007/078859 RABEP1 NM 004703 rabaptin, RAB GTPase binding effector protein 1 RABIF NM 002871 RAB-interacting factor RABL5 N-M_022777 RAB, member RAS oncogene family-like 5 RAD1 NM 002853 RADI homolog isoform I RAD23B NM_002874 UV excision repair protein RAD23 homolog B RAD51 NM 002875 RAD51 homolog protein isoform 1 RAD51L3 NM 002878 RAD5 1-like 3 isoform 1 RAE1 NM_001015885 RAEI (RNA export 1, S.pombe) homolog RAFI NM 002880 v-raf-1 marine leukemia viral oncogene homolog RAIl7 NM 020338 retinoic acid induced 17 RALBP1 NM 006788 ralA binding protein 1 RALGPS1 NM 014636 Ral GEF with PH domain and SH3 binding motif 1 RANBP1O NM_020850 RAN binding protein 10 RAP2B NM 002886 RAP2B, member of RAS oncogene family RAPGEF1 NM_005312 guanine nucleotide-releasing factor 2 isoform a RAPGEF6 NM_016340 PDZ domain-containing guanine nucleotide RARG NM_000966 retinoic acid receptor, gamma RARRESI NM_206963 retinoic acid receptor responder (tazarotene RASD2 NM 014310 RASD family, member 2 RASGEFIB NM 152545 RasGEF domain family, member 1B RASGRP1 NM_005739 RAS guanyl releasing protein 1 RASGRP4 NM_052949 RAS guanyl releasing protein 4 isoform 3 RASLIOB NM_033315 RAS-like, family 10, member B RASSF2 NM_014737 Ras association domain family 2 RASSF4 NM 032023 Ras association domain family 4 isoform a RASSF5 NM 031437 Ras association (RaIGDS/AF-6) domain family 5 RASSF6 NM 177532 Ras -association (RaIGDS/AF-6) domain family 6 RASSF8 NM 007211 Ras association (RaIGDS/AF-6) domain family 8 RAVER1 NM_133452 RAVER1 RAXLX NM_001008494 hypothetical protein LOC91464 RBI NM_000321 retinoblastoma 1 RBBP9 NM_006606 retinoblastoma binding protein 9 RBL1 NM_002895 retinoblastoma-like protein 1 isoform a RBM14 NM 006328 RNA binding motif protein 14 RBM16 NM 014892 RNA-binding motif protein 16 RBM17 NM_032905 RNA binding motif protein 17 RBM19 NM_016196 RNA binding motif protein 19 RBM24 NM 153020 hypothetical protein LOC221662 RBM3 NM 001017430 RNA binding motif protein 3 isoform b RBM33 NM_001008408 hypothetical protein LOC155435 RBM5 NM 005778 RNA binding motif protein 5 RCC2 NM_018715 RCCl-like RCD-8 NM 014329 autoantigen RCD8 RCHY1 NM_001008925 ring finger and CHY zinc finger domain RDBP NM_002904 RD RNA-binding protein RDH12 NM_152443 retinol dehydrogenase 12 (all-trans and 9-cis) RECQL5 NM_001003715 RecQ protein-like 5 isoform 2 REEPI NM 022912 receptor expression enhancing protein 1 REEP3 NM_001001330 receptor expression enhancing protein 3 REG4 NM 032044 regenerating islet-derived family, member 4 REPSI NM_031922 RALBP1 associated Eps domain containing 1 RER1 NM 007033 RERI retention in endoplasmic reticulum 1 RETNLB NM 032579 colon and small intestine-specific cysteine-rich REXOlL1 NM 172239 exonuclease GOR - 62 - WO 2008/036718 PCT/US2007/078859 REXO2 NM_015523 small fragment nuclease RFC3 NM_181558 replication factor C 3 isoform 2 RFK NM 018339 riboflavin kinase RFNG NM_002917 radical fringe homolog RFWD3 NM_018124 ring finger and WD repeat domain 3 RFX2 NM_000635 regulatory factor X2 isoform a RG9MTD3 NM 144964 RNA (guanine-9-) methyltransferase domain RGAG1 NM_020769 retrotransposon gag domain containing 1 RGL1 NM_015149 ral guanine nucleotide dissociation RGMB NM 001012761 RGM domain family, member B isoform 1 precursor RGS11 NM_003834 regulator of G-protein signalling 11 isoform 2 RGS12 NM 198432 regulator of G-protein signalling 12 isoform 5 RGS18 NM_130782 regulator of G-protein signalling 18 RGS3 NM 017790 regulator of G-protein signalling 3 isoform 3 RGSL1 NM 181572 regulator of G-protein signalling like 1 RHBDDI NM 032276 rhomboid domain containing 1 RHBDL3 NM 138328 rhomboid, veinlet-like 3 RHCG NM_016321 Rhesus blood group, C glycoprotein RHOBTBl NM_001032380 Rho-related BTB domain containing 1 RHOG NM 001665 ras homolog gene family, member G RHOJ NM 020663 TCO-like Rho GTPase RHOU NM 021205 ras homolog gene family, member U RIC8A NM_021932 resistance to inhibitors of cholinesterase 8 RICTOR NM_152756 rapamycin-insensitive companion of mTOR RIMBP2 NM 015347 RIM-binding protein 2 RIMS3 NM_014747 regulating synaptic membrane exocytosis 3 RIN2 NM 018993 RAB5 interacting protein 2 RIN3 NM_024832 Ras and Rab interactor 3 RIPK5 NM_015375 receptor interacting protein kinase 5 isoform 1 RKHD2 NM_016626 ring finger and KH domain containing 2 RLN2 NM 005059 relaxin 2 isoform 2 RMND5A NM 022780 hypothetical protein LOC64795 RNASE7 NM_032572 ribonuclease 7 RND2 NM 005440 Rho family GTPase 2 RNF1O NM_014868 ring finger protein 10 RNFl 1 NM_014372 ring finger protein 11 RNF121 NM 018320 ring finger protein 121 isoform 1 RNF125 NM 017831 ring finger protein 125 RNF135 NM 197939 ring finger protein 135 isoform 2 RNF138 NM 016271 ring finger protein 138 isoform 1 RNF144 NM 014746 ring finger protein 144 RNF165 NM_152470 ring finger protein 165 RNF185 NM_152267 ring finger protein 185 RNF2 NM_007212 ring finger protein 2 RNF24 NM_007219 ring finger protein 24 RNF26 NM_032015 ring finger protein 26 RNF4 NM_002938 ring finger protein 4 RNF40 NM_014771 ring finger protein 40 RNF6 NM_005977 ring finger protein 6 isoform 1 RNF8 NM_003958 ring finger protein 8 isoforn 1 RNGTT NM_003800 RNA guanylyltransferase and 5'-phosphatase RNMT NM_003799 RNA (guanine-7-) methyltransferase RNPC2 NM_004902 RNA-binding region containing protein 2 isoform ROBO4 NM 019055 roundabout homolog 4, magic roundabout - 63 - WO 2008/036718 PCT/US2007/078859 RODI NM 005156 ROD1 regulator of differentiation 1 RORC NM_001001523 RAR-related orphan receptor C isoform b RP1 1-19J3.3 NM_001012267 hypothetical protein LOC401541 RP 11-31 1P8.3 NM_145052 hypothetical protein LOC139596 RP13-15M17.2 NM 001010866 hypothetical protein LOC199953 RPA1 NM_002945 replication protein A1, 70kDa RPL28 NM 000991 ribosomal protein L28 RPL32 NM 000994 ribosomal protein L32 RPL34 NM 000995 ribosomal protein L34 RPL37 NM 000997 ribosomal protein L37 RPL7L1 NM _198486 ribosomal protein L7-like 1 RPLP2 NM_001004 ribosomal protein P2 RPP25 NM_017793 ribonuclease P 25kDa subunit RPS27 NM _001030 ribosomal protein S27 RPS6KA3 NM_004586 ribosomal protein S6 kinase, 90kDa, polypeptide RRAS2 NM_012250 related RAS viral (r-ras) oncogene homolog 2 RRH NM 006583 peropsin RRM2 NM 001034 ribonucleotide reductase M2 polypeptide RRM2B NM 015713 ribonucleotide reductase M2 B (TP53 inducible) RRP22 NM_001007279 RAS-related on chromosome 22 isoform b RS1 NM_000330 X-linked juvenile retinoschisis protein RSAD1 NM_018346 radical S-adenosyl methionine domain containing RTEL1 NM_032957 regulator of telomere elongation helicase 1 RTF1 NM_015138 Pafl/RNA polymerase II complex component RTN2 NM 206902 reticulon 2 isoform D RTN4RL1 NM 178568 reticulon 4 receptor-like 1 RUNDC1 NM_173079 RUN domain containing 1 RUNX3 NM_001031680 runt-related transcription factor 3 isoform 1 RWDD4A NM_152682 RWD domain containing 4A S1OOAl 1 NM_005620 S100 calcium binding protein Al1 (calgizzarin) S1OOA14 NM 020672 S100 calcium binding protein A14 S10OA7L1 NM_176823 S100 calcium binding protein A7-like 1 S1OOPBP NM_022753 S1OOP binding protein Riken isoform a SALL4 NM_020436 sal-like 4 SAMD13 NM_001010971 dnaj-like protein SAP130 NM_024545 mSin3A-associated protein 130 SAP30BP NM_013260 transcriptional regulator protein SARM1 NM_015077 sterile alpha and TIR motif containing 1 SARTl NM 005146 squamous cell carcinoma antigen recognized by T SASH1 NM 015278 SAM and SH3 domain containing 1 SATL1 NM_001012980 spermidine/spermine NI-acetyl transferase-like SAV1 NM_021818 WW45 protein SC65 NM 006455 synaptonemal complex protein SC65 SCAMP1 NM 004866 secretory carrier membrane protein 1 isoform 1 SCAMP4 NM_079834 secretory carrier membrane protein 4 SCAMP5 NM_138967 secretory carrier membrane protein 5 SCAND2 NM 022050 SCAN domain-containing protein 2 isoform 1 SCAP2 NM_003930 src family associated phosphoprotein 2 SCC-1l2 NM 015200 SCC-112 protein SCG3 NM 013243 secretogranin III SCML1 NM_006746 sex comb on midleg-like 1 isoform b SCML4 NM_198081 sex comb on midleg-like 4 SCN11A NM 014139 sodium channel, voltage-gated, type XI, alpha SCN2B NM 004588 sodium channel, voltage-gated, type II, beta - 64- WO 2008/036718 PCT/US2007/078859 SCN4A NM 000334 voltage-gated sodium channel type 4 alpha SCN4B NM_174934 sodium channel, voltage-gated, type IV, beta SCOC NM 032547 short coiled-coil protein SCRT1 NM 031309 scratch SCYLl NM 020680 SCYl-like 1 SDF4 NM 016176 calcium binding protein Cab45 precursor SDS NM_006843 serine dehydratase SEC14L1 NM_003003 SEC 14 (S. cerevisiae)-like I isoform a SEC 14L4 NM 174977 SEC l4p-like protein TAP3 SELIL NM_005065 sel-1 suppressor of lin-12-like SELI NM_033505 selenoprotein I SELL NM 000655 selectin L SELP NM_003005 selectin P precursor SELT NM_016275 selenoprotein T SEMA3E NM_012431 semaphorin 3E SEMA3G NM 020163 semaphorin sem2 SEMA4F NM 004263 semaphorin W SEMA5A NM_003966 semaphorin 5A SEMA7A NM_003612 semaphorin 7A SEPT10 NM_144710 septin 10 isoform 1 SEPT11 NM_018243 septin 1I SEPT3 NM_019106 septin 3 isoform B SEPT4 NM_080417 septin 4 isoform 4 SEPT6 NM_145799 septin 6 isoform A SEPT9 NM_006640 septin 9 SEPX1 NM 016332 selenoprotein X, 1 SERFIA NM_021967 small EDRK-rich factor IA, telomeric SERFIB NM_022978 small EDRK-rich factor IB, centromeric SERPINB13 NM_012397 serine (or cysteine) proteinase inhibitor, clade SERPINB8 NM_002640 serine (or cysteine) proteinase inhibitor, clade SERPINCl NM_000488 serine (or cysteine) proteinase inhibitor, clade SERPINEl NM_000602 plasminogen activator inhibitor-i SETD1A NM_014712 SET domain containing 1A SF1 NM_004630 splicing factor 1 isoform 1 SF3A1 NM_001005409 splicing factor 3a, subunit 1, 120kDa isoform 2 SF3A3 NM_006802 splicing factor 3a, subunit 3 SF4 NM_ 182812 splicing factor 4 isoform c SFMBT1 NM_001005158 Scm-like with four mbt domains 1 SFMBT2 NM 001029880 Scm-like with four mbt domains 2 SFRP4 NM_003014 secreted frizzled-related protein 4 SFRS11 NM_004768 splicing factor p54 SFRS14 NM_001017392 splicing factor, arginine/serine-rich 14 SFTPB NM_198843 surfactant, pulmonary-associated protein B SFXN1 NM 022754 sideroflexin 1 SFXN5 NM 144579 sideroflexin 5 SGCB NM 000232 sarcoglycan, beta (43kDa dystrophin-associated SGEF NM 015595 Src homology 3 domain-containing guanine SGK2 NM 016276 serum/glucocorticoid regulated kinase 2 isoform SGK3 NM_001033578 serum/glucocorticoid regulated kinase 3 isoform SH2BP1 NM_014633 SH2 domain binding protein 1 SH2D3A NM_005490 SH2 domain containing 3A SH2D3C NM_170600 SH2 domain containing 3C isoform 2 SH2D4A NM 022071 SH2 domain containing 4A SH2D4B NM 207372 SH2 domain containing 4B - 65 - WO 2008/036718 PCT/US2007/078859 SH3BGRL2 NM 031469 SH3 domain binding glutamic acid-rich protein SH3BP2 NM_003023 SH3-domain binding protein 2 SH3GL2 NM 003026 SH3-domain GRB2-like 2 SH3PX3 NM 153271 SH3 and PX domain containing 3 SH3PXD2A NM_014631 SH3 multiple domains 1 SH3PXD2B NM 001017995 SH3 and PX domains 2B SHANK2 NM_012309 SH3 and multiple ankyrin repeat domains 2 SHE NM 001010846 Sre homology 2 domain containing E SIDTl NM_017699 SIDI transmembrane family, member 1 SIGLECI 1 NM 052884 sialic acid binding Ig-like lectin 11 SIGLEC6 NM 198846 sialic acid binding Ig-like lectin 6 isoform 3 SIPAIL3 NM 015073 signal-induced proliferation-associated 1 like SIRPA NM_080792 signal-regulatory protein alpha precursor SIRPB1 NM_006065 signal-regulatory protein beta 1 precursor SIRPG NM_018556 signal-regulatory protein gamma isoform 1 SIRT2 NM 012237 sirtuin 2 isoform 1 SIRT5 NM 031244 sirtuin 5 isoform 2 SITI NM_014450 SHP2-interacting transmembrane adaptor protein SITPEC NM 016581 evolutionarily conserved signaling intermediate SIX4 NM 017420 sine oculis homeobox homolog 4 SKIP NM_016532 skeletal muscle and kidney enriched inositol SLAMF7 NM_021181 SLAM family member 7 SLC12A5 NM_020708 solute carrier family 12 member 5 SLC13A5 NM_177550 solute carrier family 13 (sodium-dependent SLC14A2 NM_007163 solute carrier family 14 (urea transporter), SLC15A4 NM 145648 solute carrier family 15, member 4 SLC16A12 NM 213606 solute carrier family 16 (monocarboxylic acid SLCl6Al4 NM 152527 solute carrier family 16 (monocarboxylic acid SLCI6A2 NM 006517 solute carrier family 16, member 2 SLC17A5 NM 012434 solute carrier family 17 (anion/sugar SLC17A6 NM 020346 differentiation-associated Na-dependent SLC17A7 NM_020309 solute carrier family 17, member 7 SLCIAl NM_004170 solute carrier family 1, member 1 SLCIA2 NM_004171 solute carrier family 1, member 2 SLCIA3 NM_004172 solute carrier family 1 (glial high affinity SLC22A15 NM_018420 solute carrier family 22 (organic cation SLC22A16 NM 033125 solute carrier family 22, member 16 SLC22A3 NM 021977 solute carrier family 22 member 3 SLC22A7 NM_006672 solute carrier family 22 member 7 isoform a SLC24A1 NM_004727 solute carrier family 24 SLC24A4 NM 153646 solute carrier family 24 member 4 isoform 1 SLC25A13 NM 014251 solute carrier family 25, member 13 (citrin) SLC25A15 NM 014252 solute carrier family 25 (mitochondrial carrier; SLC25A23 NM_024103 solute carrier family 25 (mitochondrial carrier; SLC25A25 NM_001006641 solute carrier family 25, member 25 isoform b SLC25A3 NM_213612 solute carrier family 25 member 3 isoform e SLC26A2 NM_000112 solute carrier family 26 member 2 SLC26A4 NM_000441 pendrin SLC26A8 NM_052961 solute carrier family 26, member 8 isoform a SLC27A1 NM_198580 solute carrier family 27 (fatty acid SLC27A4 NM_005094 solute carrier family 27 (fatty acid SLC2A3 NM_006931 solute carrier family 2 (facilitated glucose SLC2A5 NM_003039 solute carrier family 2 (facilitated SLC30A3 NM_003459 solute carrier family 30 (zinc transporter), - 66 - WO 2008/036718 PCT/US2007/078859 SLC30A8 NM_ 173851 solute carrier family 30 member 8 SLC30A9 NM 006345 solute carrier family 30 (zinc transporter), SLC31A1 NM 001859 solute carrier family 31 (copper transporters), SLC31A2 NM 001860 solute carrier family 31 (copper transporters), SLC35A4 NM 080670 solute carrier family 35, member A4 SLC35A5 NM 017945 solute carrier family 35, member A5 SLC35Bl NM 005827 solute carrier family 35, member BI SLC35B4 NM 032826 solute carrier family 35, member B4 SLC35D2 NM_007001 solute carrier family 35, member D2 SLC35EI NM 024881 solute carrier family 35, member El SLC35Fl NM_001029858 solute carrier family 35, member F1 SLC35F5 NM_025181 solute carrier family 35, member F5 SLC36Al NM 078483 solute carrier family 36 member I SLC37A2 NM 198277 solute carrier family 37 (glycerol-3-phosphate SLC38A2 NM 018976 solute carrier family 38, member 2 SLC38A3 NM 006841 solute carrier family 38, member 3 SLC39A1O NM 020342 solute carrier family 39 (zinc transporter), SLC39All NM_139177 solute carrier family 39 (metal ion SLC39A3 NM 213568 solute carrier family 39 (zinc transporter), SLC41A1 NM 173854 solute carrier family 41 member 1 SLC45A3 NM_033102 protein SLC5A6 NM 021095 solute carrier family 5 (sodium-dependent SLC5A8 NM 145913 solute carrier family 5 (iodide transporter), SLC6Al NM_003042 solute carrier family 6 (neurotransmitter SLC6A20 NM 020208 solute carrier family 6, member 20 isoform 1 SLC6A6 NM_003043 solute carrier family 6 (neurotransmitter SLC6A7 NM 014228 solute carrier family 6, member 7 SLC7A5 NM_003486 solute carrier family 7 (cationic amino acid SLC7A6 NM 003983 solute carrier family 7 (cationic amino acid SLC8A2 NM 015063 solute carrier family 8 member 2 SLC8A3 NM _033262 solute carrier family 8 member 3 isoform A SLC9Al NM 003047 solute carrier family 9, isoform Al SLC9A5 NM_004594 solute carrier family 9 (sodiuni/hydrogen SLC9A8 NM 015266 Na+/H+ exchanger isoform 8 SLCO2A1 NM 005630 solute carrier organic anion transporter family, SLCO2B1 NM 007256 solute carrier organic anion transporter family, SLFN13 NM 144682 schlafen family member 13 SLFN5 NM_144975 schlafen family member 5 SLFNLI NM_144990 hypothetical protein LOC200172 SLITRK3 NM 014926 slit and trk like 3 protein SMA4 NM 021652 SMA4 SMAD2 NM _001003652 Sma- and Mad-related protein 2 SMAD3 NM_005902 MAD, mothers against decapentaplegic homolog 3 SMARCBI NM_001007468 SWI/SNF related, matrix associated, actin SMARCD2 NM 003077 SWI/SNF-related matrix-associated SMClL1 NM_006306 SMC1 structural maintenance of chromosomes SMC2L1 NM 006444 structural maintenance of chromosomes 2-like 1 SMCR7 NM 139162 Smith-Magenis syndrome chromosome region, SMG7 NM 014837 SMG-7 homolog isoform 3 SMNDCl NM 005871 survival motor neuron domain containing 1 SMO NM 005631 smoothened SMPD3 NM 018667 sphingomyelin phosphodiesterase 3, neutral SMURF1 NM 020429 Smad ubiquitination regulatory factor 1 isoform SMYD4 NM 052928 SET and MYND domain containing 4 - 67 - WO 2008/036718 PCT/US2007/078859 SNF1LK2 NM 015191 SNF1-like kinase 2 SNIP NM 025248 SNAP25-interacting protein SNPH NM 014723 syntaphilin SNRPN NM 003097 small nuclear ribonucleoprotein polypeptide N SNTB2 NM_130845 basic beta 2 syntrophin isoform b SNURF NM_005678 SNRPN upstream reading frame protein SNXl1 NM_013323 sorting nexin I1 SNX13 NM_015132 sorting nexin 13 SNX27 NM_030918 sorting nexin family member 27 SOHLH2 NM_017826 hypothetical protein LOC54937 SON NM_003103 SON DNA-binding protein isoform G SORBS1 NM_015385 sorbin and SH3 domain containing 1 isoform 2 SORCS1 NM_001013031 SORCS receptor 1 isoform b SORCS2 NM 020777 VPS10 domain receptor protein SORCS 2 SOST NM_025237 sclerostin precursor SOXI NM 005986 SRY (sex determining region Y)-box 1 SOX13 NM 005686 SRY-box 13 SOX8 NM_014587 SRY (sex determining region Y)-box 8 SPI NM 138473 Spl transcription factor SP4 NM_003112 Sp4 transcription factor SP6 NM_199262 Sp6 transcription factor SP7 NM 152860 osterix SPACA4 NM 133498 sperm acrosomal membrane protein 14 SPAG16 NM_001025436 sperm associated antigen 16 isoform 2 SPANXAI NM 013453 sperm protein associated with the nucleus, X SPANXA2 NM_145662 sperm protein associated with the nucleus, X SPANXC NM_022661 sperm protein associated with the nucleus, X SPANXD NM 032417 sperm protein associated with the nucleus, X SPANXE NM 145665 sperm protein associated with the nucleus, X SPATA12 NM 181727 spermatogenesis associated 12 SPATA18 NM 145263 spermatogenesis associated 18 homolog SPATA2 NM_006038 spermatogenesis associated 2 SPECCl NM_001033554 spectrin domain with coiled-coils I NSP5a3a SPG21 NM_016630 acid cluster protein 33 SPIB NM 003121 Spi-B transcription factor (Spi-1/PU.1 related) SPINLW1 NM 020398 serine peptidase inhibitor-like, with Kunitz and SPIREl NM 020148 spire homolog I SPN NM_001030288 sialophorin SPOCK1 NM_004598 sparc/osteonectin, cwcv and kazal-like domains SPOCK2 NM_014767 sparc/osteonectin, cwcv and kazal-like domains SPRN NM_001012508 shadow of prion protein SPRY3 NM_005840 sprouty homolog 3 SPRYD3 NM_032840 hypothetical protein LOC84926 SPTB NM_001024858 spectrin beta isoform a SPTBN2 NM 006946 spectrin, beta, non-erythrocytic 2 SPTLC2 NM_004863 serine palmitoyltransferase, long chain base SPTY2D1 NM_194285 hypothetical protein LOC144108 SRD5Al NM 001047 steroid-5-alpha-reductase 1 SRD5A2L2 NM_001010874 steroid 5 alpha-reductase 2-like 2 SRGAP2 NM 015326 SLIT-ROBO Rho GTPase activating protein 2 SRM NM 003132 spermidine synthase SRP72 NM 006947 signal recognition particle 72kDa SS18L1 NM 015558 SS18-like protein 1 SSBP3 NM_001009955 _ single stranded DNA binding protein 3 isoform c - 68 - WO 2008/036718 PCT/US2007/078859 SSH2 NM 033389 slingshot 2 SSR3 NM 007107 signal sequence receptor gamma subunit SSTR1 NM 001049 somatostatin receptor 1 SSX1 NM 005635 synovial sarcoma, X breakpoint 1 SSX8 NM_174961 synovial sarcoma, X breakpoint 8 ST6GAL1 NM 003032 sialyltransferase 1 isoform a ST6GALNAC4 NM_175040 sialyltransferase 7D isoform b ST7L NM 017744 suppression of tumorigenicity 7-like isoform 1 ST8SIA2 NM_006011 ST8 alpha-N-acetyl-neuraminide ST8SIA4 NM_005668 ST8 alpha-N-acetyl-neuraminide STAB2 NM_017564 stabilin 2 precursor STAC NM_003149 SH3 and cysteine rich domain STAR NM 000349 steroidogenic acute regulator isoform 1 STARD13 NM_052851 START domain containing 13 isoform gamma STARD4 NM_139164 START domain containing 4, sterol regulated STARD5 NM_030574 StAR-related lipid transfer protein 5 isoform 2 STAT5A NM_003152 signal transducer and activator of transcription STAU2 NM_014393 staufen homolog 2 STCH NM_006948 stress 70 protein chaperone, STEAP3 NM_001008410 dudulin 2 isoform b STIPI NM 006819 stress-induced-phosphoprotein 1 STK1O NM_005990 serine/threonine kinase 10 STK16 NM 001008910 serine/threonine kinase 16 STK32B NM_018401 serine/threonine kinase 32B STK35 NM_080836 serine/threonine kinase 35 STK4 NM 006282 serine/threonine kinase 4 STMN3 NM_015894 SCGIO-like-protein STONI NM 006873 stonin 1 STOX2 NM 020225 storkhead box 2 STRN NM 003162 striatin, calmodulin binding protein STRN3 NM_014574 nuclear autoantigen STS NM_000351 steryl-sulfatase precursor STX17 NM_017919 syntaxin 17 STXBP I NM 001032221 syntaxin binding protein 1 isoform b STXBP5 NM_139244 tomosyn SUFU NM 016169 suppressor of fused SUHW1 NM 080740 suppressor of hairy wing homolog 1 SULT1A3 NM 001017387 sulfotransferase family, cytosolic, IA, SULT1A4 NM 001017389 sulfotransferase family, cytosolic, 1A, SULTIE1 NM 005420 sulfotransferase, estrogen-preferring SULT2A1 NM_003167 sulfotransferase family, cytosolic, 2A, SUMO3 NM_006936 small ubiquitin-like modifier protein 3 SURB7 NM_004264 SRB7 suppressor of RNA polymerase B homolog SURF4 NM 033161 surfeit 4 SURF5 NM_133640 surfeit 5 isoform b SUSD2 NM 019601 sushi domain containing 2 SUSD4 NM 017982 sushi domain containing 4 isoform a SUV420H1I NM 016028 suppressor of variegation 4-20 homolog 1 isoform SV2A NM 014849 synaptic vesicle glycoprotein 2 SV2B NM 014848 synaptic vesicle protein 2B homolog SVOP NM 018711 SV2 related protein SWAP70 NM_015055 SWAP-70 protein SYBL1 NM_005638 synaptobrevin-like 1 SYN2 NM 003178 synapsin II isoform IIb - 69 - WO 2008/036718 PCT/US2007/078859 SYN3 NM_133632 synapsin III isoform IIIb SYNGRI NM_004711 synaptogyrin 1 isoform l a SYNJ2 NM 003898 synaptojanin 2 SYNJ2BP NM_018373 synaptojanin 2 binding protein SYT10 NM_198992 synaptotagmin 10 SYT11 NM_152280 synaptotagmin 12 SYT3 NM 032298 synaptotagmin 3 SYT6 NM 205848 synaptotagmin VI SYT7 NM_004200 synaptotagmin VII SYT9 NM 175733 synaptotagmin IX TACCI NM 006283 transforming, acidic coiled-coil containing TACSTD2 NM 002353 tumor-associated calcium signal transducer 2 TADA3L NM 133480 transcriptional adaptor 3-like isoform b TAF12 NM 005644 TAF12 RNA polymerase II, TATA box binding TAF1L NM 153809 TBP-associated factor RNA polymerase 1-like TAOK2 NM_004783 TAO kinase 2 isoform 1 TAPBP NM_003190 tapasin isoform 1 precursor TARDBP NM_007375 TAR DNA binding protein TATDN2 NM_014760 TatD DNase domain containing 2 TAZ NM 181314 tafazzin isoform 5 TBC1DI NM 015173 TBC1 (tre-2/USP6, BUB2, cdc16) domain family, TBC1D1OB NM 015527 TBC1 domain family, member 1OB TBC1D14 NM_020773 TBCI domain family, member 14 TBC1D20 NM_144628 TBC1 domain family, member 20 TBC1D22A NM_014346 TBC1 domain family, member 22A TBC1D22B NM_017772 TBC1 domain family, member 22B TBClD2B NM_015079 TBC1 domain family, member 2B TBL1X NM 005647 transducin beta-like IX TBX21 NM 013351 T-box 21 TBX3 NM_005996 T-box 3 protein isoform 1 TCEAL7 NM_152278 hypothetical protein LOC56849 TCF15 NM_004609 basic helix-loop-helix transcription factor 15 TCF20 NM_005650 transcription factor 20 isoform 1 TCF21 NM_198392 transcription factor 21 TCF7 NM_003202 transcription factor 7 (T-cell specific, TCHHL1 NM_001008536 trichohyalin-like 1 TCHP NM_032300 trichoplein TCN2 NM_000355 transcobalamin II precursor TCP1O NM 004610 t-complex 10 TCTA NM_022171 T-cell leukemia translocation altered gene TEADI NM_021961 TEA domain family member 1 TEAD3 NM 003214 TEA domain family member 3 TERT NM 198253 telomerase reverse transcriptase isoform 3 TEX2 NM_018469 testis expressed sequence 2 TEX261 NM_144582 tests expressed sequence 261 TFAP2B NM_003221 transcription factor AP-2 beta (activating TFF3 NM_003226 trefoil factor 3 precursor TGIF2 NM 021809 TGFB-induced factor 2 TGM2 NM_004613 transglutaminase 2 isofonn a THADA NM_198554 thyroid adenoma associated isoform 2 THAP6 NM 144721 THAP domain containing 6 THBSI NM 003246 thrombospondin 1 precursor THEDCl NM_018324 thioesterase domain containing 1 isoform 1 THEM4 NM_053055 thioesterase superfamily member 4 isoform a - 70 - WO 2008/036718 PCT/US2007/078859 THEM5 NM 182578 thioesterase superfamily member 5 THYl NM_006288 Thy-i cell surface antigen TIAl NM 022037 TIAl protein isoform 1 TIGD5 NM_032862 tigger transposable element derived 5 TIMM17A NM 006335 translocase of inner mitochondrial membrane 17 TK2 NM 004614 thymidine kinase 2, mitochondrial TKTL1 NM 012253 transketolase-like 1 TKTL2 NM 032136 transketolase-like 2 TLK2 NM 006852 tousled-like kinase 2 TLN2 NM 015059 talin 2 TLR1O NM 001017388 toll-like receptor 10 precursor TLX2 NM_016170 T-cell leukemia, homeobox 2 TM2D2 NM_001024380 TM2 domain containing 2 isoform b TM4SFl1 NM 015993 plasmolipin TM4SF20 NM_024795 transmembrane 4 L six family member 20 TM7SF4 NM_030788 dendritic cell-specific transmembrane protein TMBIM1 NM_022152 transmembrane BAX inhibitor motif containing 1 TMC5 NM 024780 transmembrane channel-like 5 TMCC3 NM_020698 transmembrane and coiled-coil domains 3 TMED2 NM 006815 coated vesicle membrane protein TMEMI NM_001001723 transmembrane protein 1 isoform b TMEM105 NM_178520 hypothetical protein LOC284186 TMEM106A NM_145041 hypothetical protein LOC113277 TMEM113 NM_025222 hypothetical protein PR02730 TMEM 116 NM_138341 hypothetical protein LOC89894 TMEM119 NM_181724 hypothetical protein LOC338773 TMEM12 NM 152311 transmembrane protein 12 TMEM121 NM_025268 hole protein TMEM127 NM 017849 hypothetical protein LOC55654 TMEM132D NM_133448 hypothetical protein LOC121256 TMEM134 NM_025124 hypothetical protein LOC80194 TMEM140 NM_018295 hypothetical protein LOC55281 TMEM148 NM_153238 hypothetical protein LOC197196 TMEM16B NM_020373 transmembrane protein 16B TMEM16F NM 001025356 transmembrane protein 16F TMEM16G NM_001001891 transmembrane protein 16G isoform NGEP long TMEM19 NM 018279 transmembrane protein 19 TMEM29 NM_014138 hypothetical protein LOC29057 TMEM30B NM_001017970 transmembrane protein 30B TMEM33 NM 018126 transmembrane protein 33 TMEM40 NM 018306 transmembrane protein 40 TMEM41B NM 015012 transmembrane protein 41B TMEM43 NM 024334 transmembrane protein 43 TMEM53 NM_024587 transmembrane protein 53 TMEM56 NM_152487 transmembrane protein 56 TMEM58 NM_198149 transmembrane protein 58 TMEM60 NM_032936 transmembrane protein 60 TMEM63A NM_014698 transmembrane protein 63A TMEM69 NM 016486 transmembrane protein 69 TMEM80 NM 174940 hypothetical protein LOC283232 TMEM97 NM_014573 hypothetical protein MAC30 TMLHE NM 018196 trimethyllysine hydroxylase, epsilon TMOD2 NM 014548 tropomodulin 2 (neuronal) TMPRSS1lB NM 182502 transmembrane protease, shrine 1 B -71 - WO 2008/036718 PCT/US2007/078859 TMPRSS3 NM_024022 transmembrane protease, serine 3 isoform 1 TMPRSS4 NM 019894 transmembrane protease, serine 4 isoform 1 TNFAIP1 NM_021137 tumor necrosis factor, alpha-induced protein 1 TNFAIP8L1 NM 152362 tumor necrosis factor, alpha-induced protein TNFAIP8L3 NM_207381 tumor necrosis factor, alpha-induced protein TNFRSF1OB NM_003842 tumor necrosis factor receptor superfamily, TNFRSF1OC NM 003841 tumor necrosis factor receptor superfamily, TNFRSFIOD NM_003840 tumor necrosis factor receptor superfamily, TNFRSF19 NM 148957 tumor necrosis factor receptor superfamily, TNFRSF8 NM 001243 tumor necrosis factor receptor superfamily, TNFSF1O NM_003810 tumor necrosis factor (ligand) superfamily, TNFSF4 NM 003326 tumor necrosis factor (ligand) superfamily, TNFSF9 NM 003811 tumor necrosis factor (ligand) superfamily, TNIP3 NM 024873 hypothetical protein LOC79931 TNNI1 NM 003281 troponin I, skeletal, slow TNP1 NM_003284 transition protein 1 (during histone to .TNPO2 NM_013433 transportin 2 (importin 3, karyopherin beta 2b) TNRC15 NM_015575 trinucleotide repeat containing 15 TNRC6B NM_001024843 trinucleotide repeat containing 6B isoform 2 TNS3 NM 022748 tensin-like SH2 domain containing I TOB2 NM 016272 transducer of ERBB2, 2 TOLLIP NM_019009 toll interacting protein TOM1L2 NM 001033551 target of mybI-like 2 isoform 1 TOMM40L NM 032174 translocase of outer mitochondrial membrane 40 TOP2A NM_001067 DNA topoisomerase II, alpha isozyme TOR2A NM_130459 torsin family 2, member A TOR3A NM 022371 torsin family 3, member A TP53 NM_000546 tumor protein p53 TP53INP1 NM_033285 tumor protein p53 inducible nuclear protein 1 TP53RK NM_033550 p53-related protein kinase TPD52L3 NM_033516 protein kinase NYD-SP25 isoform 1 TPM3 NM 153649 tropomyosin 3 isoform 2 TPM4 NM_003290 tropomyosin 4 TPP1 NM 000391 tripeptidyl-peptidase I precursor TRAF7 NM_032271 ring finger and WD repeat domain 1 isoform 1 TRAIP NM 005879 TRAF interacting protein TRAM2 NM 012288 translocation-associated membrane protein 2 TRAPPC3 NM_014408 BET3 homolog TRIAD3 NM_207111 TRIAD3 protein isoform a TRIB3 NM 021158 tribbles 3 TRIM1O NM_006778 tripartite motif-containing 10 isoform 1 TRIM14 NM 033220 tripartite motif protein TRIM14 isoform alpha TRIM22 NM 006074 tripartite motif-containing 22 TRIM24 NM_003852 transcriptional intermediary factor 1 alpha TRIM25 NM 005082 tripartite motif-containing 25 TRIM26 NM 003449 tripartite motif-containing 26 TRIM29 NM_012101 tripartite motif protein TRIM29 isoform alpha TRIM35 NM 015066 tripartite motif-containing 35 isoform 1 TRIM37 NM_015294 tripartite motif-containing 37 protein TRIM44 NM_017583 DIPB protein TRIM5 NM_033034 tripartite motif protein TRIM5 isoform alpha TRIM52 NM_032765 hypothetical protein LOC84851 TRIM55 NM_033058 ring finger protein 29 isoform 2 TRIM56 NM 030961 tripartite motif-containing 56 - 72 - WO 2008/036718 PCT/US2007/078859 TRIM58 NM 015431 tripartite motif-containing 58 TRIM62 NM 018207 tripartite motif-containing 62 TRIM65 NM 173547 tripartite motif-containing 65 TRIM67 NM 001004342 hypothetical protein LOC440730 TRIM73 NM_198924 hypothetical protein LOC375593 TRIM74 NM 198853 hypothetical protein LOC378108 TRIM9 NM_052978 tripartite motif protein 9 isoform 2 TRIO NM 007118 triple functional domain (PTPRF interacting) TRITI NM 017646 tRNA isopentenyltransferase 1 TRMT5 NM 020810 tRNA-(N1 G37) methyltransferase TRPC5 NM 012471 transient receptor potential cation channel, TRPM1 NM 002420 transient receptor potential cation channel, TRPM2 NM 001001188 transient receptor potential cation channel, TRPS1 NM 014112 zinc finger transcription factor TRPSI TRPV5 NM_019841 transient receptor potential cation channel, TRPV6 NM_018646 transient receptor potential cation channel, TRUB2 NM_015679 TruB pseudouridine (psi) synthase homolog 2 TSCl NM_000368 tuberous sclerosis 1 protein isoform 1 TSC22D3 NM_001015881 TSC22 domain family, member 3 isoform 3 TSN NM 004622 translin TSNAX NM 005999 translin-associated factor X TSPAN13 NM_014399 tetraspan NET-6 TSPAN15 NM 012339 transmembrane 4 superfamily member 15 TSPAN2 NM_005725 tetraspan 2 TSPAN9 NM_006675 tetraspanin 9 TSPYL5 NM 033512 TSPY-like 5 TTBK1 NM 032538 tau tubulin kinase 1 TTBK2 NM 173500 tau tubulin kinase 2 TTC12 NM_017868 tetratricopeptide repeat domain 12 TTC19 NM_017775 tetratricopeptide repeat domain 19 TTC21B NM_024753 tetratricopeptide repeat domain 21B TTF2 NM_003594 transcription termination factor, RNA polymerase TTL NM 153712 tubulin tyrosine ligase TTLL2 NM_031949 tubulin tyrosine ligase-like family, member 2 TTLL3 NM 001025930 tubulin tyrosine ligase-like family, member 3 TTLL6 NM_173623 hypothetical protein LOC284076 TTLL9 NM_001008409 tubulin tyrosine ligase-like family, member 9 TTYH2 NM 032646 tweety 2 isoform 1 TTYH3 NM 025250 tweety 3 TUB NM_003320 tubby isoform a TUBB NM_178014 tubulin, beta polypeptide TUBB 1 NM 030773 beta tubulin 1, class VI TUBB4 NM 006087 tubulin, beta 4 TUBG1 NM_001070 tubulin, gamma 1 TUBG2 NM 016437 tubulin, gamma 2 TUBGCP6 NM_001008658 tubulin, gamma complex associated protein 6 TUFTI NM 020127 tuftelin 1 TULP3 NM_003324 tubby like protein 3 TUSC5 NM 172367 LOST TXLNA NM_175852 taxilin TXLNB NM_153235 muscle-derived protein 77 TXNDC13 NM_021156 thioredoxin domain containing 13 TXNDC4 NM_015051 thioredoxin domain containing 4 (endoplasmic TXNL4B NM 017853 thioredoxin-like 4B - 73 - WO 2008/036718 PCT/US2007/078859 TXNRD1 NM 003330 thioredoxin reductase 1 TYSND1 NM 173555 trypsin domain containing 1 isoform a UACA NM 001008224 uveal autoantigen with coiled-coil domains and UAP1L1 NM 207309 UDP-N-acteylglucosamine pyrophosphorylase 1-like UBE2E1 NM 003341 ubiquitin-conjugating enzyme E2E 1 isoform 1 UBE2E3 NM 006357 ubiquitin-conjugating enzyme E2E 3 UBE2G1 NM 003342 ubiquitin-conjugating enzyme E2G 1 isoform 1 UBE2I NM 003345 ubiquitin-conjugating enzyme E21 UBE2Jl NM_016021 ubiquitin-conjugating enzyme E2, Jt UBE2Q1 NM 017582 ubiquitin-conjugating enzyme E2Q UBE2R2 NM_017811 ubiquitin-conjugating enzyme UBC3B UBE3B NM 183414 ubiquitin protein ligase E3B isoform b UBE3C NM 014671 ubiquitin protein ligase E3C UBL3 NM 007106 ubiquitin-like 3 UBL7 NM_032907 ubiquitin-like 7 (bone marrow stromal UBNl NM 016936 ubinuclein 1 UBOX5 NM_014948 U-box domain containing 5 isoform a UBXD2 NM_014607 UBX domain containing 2 UBXD8 NM_014613 UBX domain containing 8 UGDH NM 003359 UDP-glucose dehydrogenase UGT1A1 NM 000463 UDP glycosyltransferase 1 family, polypeptide Al UGT1A1O NM 019075 UDP glycosyltransferase 1 family, polypeptide UGT1A3 NM_019093 UDP glycosyltransferase 1 family, polypeptide A3 UGTlA4 NM_007120 UDP glycosyltransferase 1 family, polypeptide A4 UGT1A5 NM_019078 UDP glycosyltransferase 1 family, polypeptide A5 UGTlA6 NM_001072 UDP glycosyltransferase 1 family, polypeptide A6 UGT1A7 NM 019077 UDP glycosyltransferase 1 family, polypeptide A7 UGT1A8 NM_019076 UDP glycosyltransferase 1 family, polypeptide A8 UGT1A9 NM_021027 UDP glycosyltransferase 1 family, polypeptide A9 ULBP1 NM_025218 UL16 binding protein 1 UMOD NM 001008389 uromodulin precursor UNCl3D NM_199242 unc-13 homolog D UNC45B NM 001033576 cardiomyopathy associated 4 isoform 2 UNC5A NM_133369 netrin receptor Unc5hl UNC5D NM_080872 netrin receptor Unc5h4 UNC93A NM_018974 unc-93 homolog A UPF1 NM 002911 regulator of nonsense transcripts I UPF2 NM_015542 UPF2 regulator of nonsense transcripts homolog USFI NM_007122 upstream stimulatory factor 1 isoform 1 USP18 NM_017414 ubiquitin specific protease 18 USP2 NM_004205 ubiquitin specific protease 2 isoform a USP37 NM 020935 ubiquitin specific protease 37 USP46 NM 022832 ubiquitin specific protease 46 USP47 NM 017944 ubiquitin specific protease 47 USP49 NM_018561 ubiquitin specific protease 49 UTP14C NM_021645 UTP14, U3 small nucleolar ribonucleoprotein, UTS2D NM_198152 urotensin 2 domain containing UVRAG NM 003369 UV radiation resistance associated gene VANGL2 NM 020335 vang-like 2 (van gogh, Drosophila) VAPB NM 004738 VAMP-associated protein B/C VASH1 NM 014909 vasohibin 1 VATI NM 006373 vesicle amine transport protein 1 VAXI NM 199131 ventral anterior homeobox 1 VBP1 NM_003372 von Hippel-Lindau binding protein 1 - 74- WO 2008/036718 PCT/US2007/078859 VCPIP1 NM 025054 valosin containing protein (p97)/p47 complex VDACl NM 003374 voltage-dependent anion channel 1 VEGF NM 001025366 vascular endothelial growth factor isoform a VGLL3 NM 016206 colon carcinoma related protein VHL NM_000551 von Hippel-Lindau tumor suppressor isoform 1 VIPRI NM 004624 vasoactive intestinal peptide receptor 1 VISA NM_020746 virus-induced signaling adapter VMD2L3 NM 152439 vitelliform macular dystrophy 2-like 3 VPREB1 NM_007128 immunoglobulin iota chain preproprotein VPS13A NI 001018037 vacuolar protein sorting 13A isoform C VPS13D NM_015378 vacuolar protein sorting 13D isoform I VPS16 NM_022575 vacuolar protein sorting 16 isoform 1 VPS26A NM_004896 vacuolar protein sorting 26 homolog A isoform 1 VPS37A NM_152415 hepatocellular carcinoma related protein 1 VPS45A NM 007259 vacuolar protein sorting 45A VPS4B NM 004869 vacuolar protein sorting factor 4B VPS52 NM_022553 suppressor of actin mutations 2-like VPS72 NM_005997 transcription factor-like 1 VSIG4 NM_ 007268 V-set and immunoglobulin domain containing 4 VSIG9 NM_173799 hypothetical protein LOC201633 VTCN1 NM 024626 V-set domain containing T cell activation WASF3 NM 006646 WAS protein family, member 3 WASPIP NM_003387 WASP-interacting protein WBP2 NM_012478 WW domain binding protein 2 WBP5 NM_001006612 WW domain binding protein 5 WBSCR17 NM_022479 UDP-GaINAc:polypeptide WDFY3 NM_014991 WD repeat and FYVE domain containing 3 isoform WDR17 NM_170710 WD repeat domain 17 isoform 1 WDR22 NM_003861 Breakpoint cluster region protein, uterine WDR23 NM_025230 WD repeat domain 23 isoform 1 WDR33 NM_018383 WD repeat domain 33 isoform 1 WDR36 NM_139281 WD repeat domain 36 WDR42B NM 001017930 WD repeat domain 42B WDR48 NM 020839 WD repeat domain 48 WDR50 NM_016001 WD repeat domain 50 WDR6 NM_018031 WD repeat domain 6 protein WDR64 NM_144625 hypothetical protein LOC128025 WDR68 NM_005828 WD-repeat protein WDR7 NM 015285 rabconnectin-3 beta isoform 1 WDR81 NM_152348 alpha-2-plasmin inhibitor WDTC1 NM 015023 WD and tetratricopeptide repeats 1 WFDC1 NM 021197 WAP four-disulfide core domain 1 precursor WFSI NM 006005 wolframin WHSC1 NM 014919 Wolf-Hirschhorn syndrome candidate 1 protein WIGI NM_022470 p53 target zinc finger protein isoform 1 WIRE NM_133264 WIRE protein WNK4 NM_032387 WNK lysine deficient protein kinase 4 WNT2 NM_003391 wingless-type MMTV integration site family WNT5B NM_030775 wingless-type MMTV integration site family, WSBl NM 015626 WD repeat and SOCS box-containing 1 isoform 1 WWC3 NM_015691 hypothetical protein LOC55841 WWP2 NM 007014 WW domain containing E3 ubiquitin protein ligase XK NM 021083 McLeod syndrome-associated, Kell blood group XKR5 NM 207411 XK-related protein Sa - 75 - WO 2008/036718 PCT/US2007/078859 XLKD1 NM 006691 extracellular link domain containing 1 XPO4 NM 022459 exportin 4 XPO5 NM_020750 exportin 5 XRCC2 NM_005431 X-ray repair cross complementing protein 2 XRN1 NM 019001 5'-3' exoribonuclease 1 XYLB NM 005108 xylulokinase homolog YAF2 NM 001012424 YY1 associated factor 2 isoform b YARS NM 003680 tyrosyl-tRNA synthetase YEATS2 NM_018023 YEATS domain containing 2 YIFlB NM 033557 Yip1 interacting factor homolog B isoform 2 YPEL1 NM_013313 yippee-like 1 YPEL2 NM_001005404 yippee-like 2 YPEL5 NM_016061 yippee-like 5 YTHDC2 NM_022828 YTH domain containing 2 YWHAB NM 003404 tyrosine 3-monooxygenase/tryptophan ZADHI NM_152444 zinc binding alcohol dehydrogenase, domain ZADH2 NM 175907 zinc binding alcohol dehydrogenase, domain ZAK NM 016653 MLK-related kinase isoform 1 ZBTB40 NM_014870 zinc finger and BTB domain containing 40 ZBTB41 NM 194314 zinc finger and BTB domain containing 41 ZBTB5 NM_014872 zinc finger and BTB domain containing 5 ZBTB6 NM 006626 zinc finger protein 482 ZC3H12A NM_025079 zinc finger CCCH-type containing 12A ZCCHC14 NM 015144 zinc finger, CCHC domain containing 14 ZDHHC 11 NM 024786 zinc finger, DHHC domain containing 11 ZDHHC2 NM_016353 rec ZDHHC23 NM_173570 zinc finger, DHHC domain containing 23 ZDHHC4 NM_018106 zinc finger, DHHC domain containing 4 ZDHHC9 NM_001008222 zinc finger, DHHC domain containing 9 ZFAND2B NM_138802 zinc finger, AN1-type domain 2B ZFP30 NM_014898 zinc finger protein 30 homolog ZFP36L1 NM_004926 butyrate response factor 1 ZFP41 NM 173832 zinc finger protein 41 homolog ZFP91 NM_053023 zinc finger protein 91 isoform 1 ZFP95 NM_014569 zinc finger protein 95 homolog ZFYVE16 NM 014733 endosome-associated FYVE-domain protein ZFYVE27 NM_001002261 zinc finger, FYVE domain containing 27 isoform ZFYVE28 NM_020972 zinc finger, FYVE domain containing 28 ZGPAT NM_181484 zinc finger, CCCH-type with G patch domain ZHX3 NM_015035 zinc fingers and homeoboxes 3 ZICi NM_003412 zinc finger protein of the cerebellum 1 ZIC3 NM_003413 zinc finger protein of the cerebellum 3 ZIC4 NM_032153 zinc finger protein of the cerebellum 4 ZIM3 NM_052882 zinc finger, imprinted 3 ZKSCAN1 NM_003439 zinc finger protein 36 ZMYM3 NM 005096 zinc finger protein 261 ZMYM4 NM_005095 zinc finger protein 262 ZMYND11 NM_006624 zinc finger, MYND domain containing 11 isoform ZMYND19 NM_138462 zinc finger, MYND domain containing 19 ZNF132 NM 003433 zinc finger protein 132 (clone pHZ-12) ZNF136 NM 003437 zinc finger protein 136 (clone pHZ-20) ZNF137 NM 003438 zinc finger protein 137 (clone pHZ-30) ZNF157 NM 003446 zinc finger protein 157 ZNF160 NM 033288 zinc finger protein 160 - 76 - WO 2008/036718 PCT/US2007/078859 ZNF 167 NM 018651 zinc finger protein ZFP isoform 1 ZNF17 NM 006959 zinc finger protein 17 ZNF182 NM 001007088 zinc finger protein 21 isoform 2 ZNF187 NM 001023560 zinc finger protein 187 ZNF192 NM 006298 zinc finger protein 192 ZNF200 NM 003454 zinc finger protein 200 isoform 1 ZNF202 NM_003455 zinc finger protein 202 ZNF217 NM_006526 zinc finger protein 217 ZNF226 NM 001032374 zinc finger protein 226 isoform b ZNF236 NM_007345 zinc finger protein 236 ZNF264 NM 003417 zinc finger protein 264 ZNF265 NM_005455 zinc finger protein 265 isoform 2 ZNF272 NM 006635 zinc finger protein 272 ZNF276 NM 152287 zinc finger protein 276 homolog ZNF294 NM 015565 zinc finger protein 294 ZNF300 NM_052860 zinc finger protein 300 ZNF31 NM 145238 zinc finger protein 31 ZNF313 NM_018683 zinc finger protein 313 ZNF317 NM 020933 zinc finger protein 317 ZNF318 NM 014345 zinc finger protein 318 ZNF320 NM_207333 zinc finger protein 320 ZNF322A NM 024639 zinc finger protein 322A ZNF322B NM 199005 zinc finger protein 322B ZNF329 NM_024620 zinc finger protein 329 ZNF333 NM 032433 zinc finger protein 333 ZNF33A NM_006974 zinc finger protein 33A ZNF33B NM_006955 zinc finger protein 33B ZNF346 NM 012279 zinc finger protein 346 ZNF365 NM_199451 zinc finger protein 365 isoform C ZNF37A NM 001007094 zinc finger protein 37a ZNF384 NM_133476 nuclear matrix transcription factor 4 isoform a ZNF385 NM 015481 zinc finger protein 385 ZNF394 NM_032164 zinc finger protein 99 ZNF397 NM 032347 zinc finger protein 397 ZNF41 NM 007130 zinc finger protein 41 ZNF425 NM_001001661 zinc finger protein 425 ZNF426 NM_024106 zinc finger protein 426 ZNF43 NM_003423 zinc finger protein 43 (HTF6) ZNF430 NM_025189 zinc finger protein 430 ZNF445 NM 181489 zinc finger protein 445 ZNF471 NM_020813 zinc finger protein 471 ZNF480 NM_144684 zinc finger protein 480 ZNF483 NM 001007169 zinc finger protein 483 isoform b ZNF485 NM 145312 zinc finger protein 485 ZNF490 NM 020714 zinc finger protein 490 ZNF493 NM 175910 zinc finger protein 493 ZNF497 NM 198458 zinc finger protein 497 ZNF498 NM 145115 zinc finger protein 498 ZNF500 NM 021646 zinc finger protein 500 ZNF514 NM 032788 zinc finger protein 514 ZNF526 NM_133444 zinc finger protein 526 ZNF529 NM_020951 zinc finger protein 529 ZNF543 NM_213598 zinc finger protein 543 ZNF545 NM 133466 zinc finger protein 545 - 77 - WO 2008/036718 PCT/US2007/078859 ZNF547 NM_173631 zinc finger protein 547 ZNF562 NM_017656 zinc finger protein 562 ZNF565 NM_152477 zinc finger protein 565 ZNF570 NM 144694 zinc finger protein 570 ZNF571 NM 016536 zinc finger protein 571 ZNF577 NM_032679 zinc finger protein 577 ZNF581 NM_016535 zinc finger protein 581 ZNF583 NM_152478 zinc finger protein 583 ZNF592 NM 014630 zinc finger protein 592 ZNF599 NM_001007247 zinc finger protein 599 isoform b ZNF600 NM_198457 zinc finger protein 600 ZNF605 NM_183238 zinc finger protein 605 ZNF607 NM_032689 zinc finger protein 607 ZNF621 NM_198484 zinc finger protein 621 ZNF622 NM_033414 zinc finger protein 622 ZNF623 NM 014789 zinc finger protein 623 ZNF650 NM_172070 zinc finger protein 650 ZNF651 NM_145166 zinc finger protein 651 ZNF652 NM_014897 zinc finger protein 652 ZNF660 NM_173658 zinc finger protein 660 ZNF662 NM_207404 zinc finger protein 662 ZNF677 NM_182609 zinc finger protein 677 ZNF694 NM_001012981 zinc finger protein 694 ZNF696 NM_030895 zinc finger protein 696 ZNF702 NM 024924 zinc finger protein 702 ZNF705A NM_001004328 hypothetical protein LOC440077 ZNF708 NM_021269 zinc finger protein 15-like 1 (KOX 8) ZNF81 NM_007137 zinc finger protein 81 (HFZ20) ZNF93 NM_001004126 zinc finger protein 93 isoform b ZNRF2 NM_147128 zinc finger/RING finger 2 ZSCAN2 NM_181877 zinc finger protein 29 isoform 1 ZSWIM4 NM_023072 zinc finger, SWIM domain containing 4 ZWILCH NM 017975 Zwilch ZWINT NM 001005414 ZW10 interactor isoform c ZYG11A NM_001004339 hypothetical protein LOC440590 ZYGI1B NM 024646 hypothetical protein LOC79699 Table 4. hsa-miR-143 targets that exhibited altered mRNA expression levels in human cancer cells after transfection with pre-miR hsa-miR-143. for Ref Seq ID reference - Pruitt et aL, 2005. RefSeq Gene Symbol Transcript ID Description ATP6VlA NM 001690 ATPase, H+ transporting, lysosomal 70kD, VI ATXN1 NM 000332 ataxin 1 CCND1 NM 053056 cyclin DI CLIC4 NM 013943 chloride intracellular channel 4 DDAH1 NM 012137 dimethylarginine dimethylaminohydrolase 1 GALC NM 000153 galactosylceramidase isoform a precursor GATM NM_001482 glycine amidinotransferase (L-arginine:glycine GOLPH2 NM 016548 golgi phosphoprotein 2 IGFBP3 NM 000598 insulin-like growth factor binding protein 3 LMO4 NM 006769 LIM domain only 4 MCL1 NM 021960 myeloid cell leukemia sequence 1 isoform 1 - 78 - WO 2008/036718 PCT/US2007/078859 PROSC NM 007198 proline synthetase co-transcribed homolog RAB 11FIPi NM_001002814 Rab coupling protein isoform 3 RBLl NM_002895 retinoblastoma-like protein 1 isoform a RHOBTB1 NM 001032380 Rho-related BTB domain containing 1 SERPINEl NM 000602 plasminogen activator inhibitor-i SLC35B1 NM 005827 salute carrier family 35, member BI WASPIP NM 003387 WASP-interacting protein WDR50 NM 016001 WD repeat domain 50 The predicted gene targets of hsa-miR-143 whose mRNA expression levels are affected by hsa-miR-143 represent particularly useful candidates for cancer therapy and therapy of other diseases through manipulation of their expression levels. 5 Certain embodiments of the invention include determining expression of one or more marker, gene, or nucleic acid segment representative of one or more genes, by using an amplification assay, a hybridization assay, or protein assay, a variety of which are well known to one of ordinary skill in the art. In certain aspects, an amplification assay can be a quantitative amplification assay, such as quantitative RT 10 PCR or the like. In still further aspects, a hybridization assay can include array hybridization assays or solution hybridization assays. The nucleic acids from a sample may be labeled from the sample and/or hybridizing the labeled nucleic acid to one or more nucleic acid probes. Nucleic acids, mRNA, and/or nucleic acid probes may be coupled to a support. Such supports are well known to those of ordinary skill 15 in the art and include, but are not limited to glass, plastic, metal, or latex. In particular aspects of the invention, the support can be planar or in the form of a bead or other geometric shapes or configurations known in the art. Proteins are typically assayed by immunoblotting, chromatography, or mass spectrometry or other methods known to those of ordinary skill in the art. 20 The present invention also concerns kits containing compositions of the invention or compositions to implement methods of the invention. In some embodiments, kits can be used to evaluate one or more marker molecules, and/or express one or more miRNA or miRNA inhibitor. In certain embodiments, a kit contains, contains at least or contains at most 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 25 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 100, 150, 200 or more probes, recombinant nucleic acid, or synthetic nucleic acid molecules related to the markers to be assessed or an miRNA or miRNA inhibitor to - 79 - WO 2008/036718 PCT/US2007/078859 be expressed or modulated, and may include any range or combination derivable therein. Kits may comprise components, which may be individually packaged or placed in a container, such as a tube, bottle, vial, syringe, or other suitable container means. Individual components may also be provided in a kit in concentrated 5 amounts; in some embodiments, a component is provided individually in the same concentration as it would be in a solution with other components. Concentrations of components may be provided as lx, 2x, 5x, lOx, or 20x or more. Kits for using probes, synthetic nucleic acids, recombinant nucleic acids, or non-synthetic nucleic acids of the invention for therapeutic, prognostic, or diagnostic applications are 10 included as part of the invention. Specifically contemplated are any such molecules corresponding to any miRNA reported to influence biological activity or expression of one or more marker gene or gene pathway described herein. In certain aspects, negative and/or positive controls are included in some kit embodiments. The control molecules can be used to verify transfection efficiency and/or control for transfection 15 induced changes in cells. Certain embodiments are directed to a kit for assessment of a pathological condition or the risk of developing a pathological condition in a patient by nucleic acid profiling of a sample comprising, in suitable container means, two or more nucleic acid hybridization or amplification reagents. The kit can comprise reagents 20 for labeling nucleic acids in a sample and/or nucleic acid hybridization reagents. The hybridization reagents typically comprise hybridization probes. Amplification reagents include, but are not limited to amplification primers, reagents, and enzymes. In some embodiments of the invention, an expression profile is generated by steps that include: (a) labeling nucleic acid in the sample; (b) hybridizing the nucleic 25 acid to a number of probes, or amplifying a number of nucleic acids, and (c) detennining and/or quantitating nucleic acid hybridization to the probes or detecting and quantitating amplification products, wherein an expression profile is generated. See U.S. Provisional Patent Application 60/575,743 and the U.S. Provisional Patent Application 60/649,584, and U.S. Patent Application Serial No. 11/141,707 and U.S. 30 Patent Application Serial No. 11/273,640, all of which are hereby incorporated by reference. - S0 - WO 2008/036718 PCT/US2007/078859 Methods of the invention involve diagnosing and/or assessing the prognosis of a patient based on a miRNA and/or a marker nucleic acid expression profile. In certain embodiments, the elevation or reduction in the level of expression of a particular gene or genetic pathway or set of nucleic acids in a cell is correlated with a 5 disease state or pathological condition compared to the expression level of the same in a normal or non-pathologic cell or tissue sample. This correlation allows for diagnostic and/or prognostic methods to be carried out when the expression level of one or more nucleic acid is measured in a biological sample being assessed and then compared to the expression level of a normal or non-pathologic cell or tissue sample. 10 It is specifically contemplated that expression profiles for patients, particularly those suspected of having or having a propensity for a particular disease or condition such as cancer, can be generated by evaluating any of or sets of the miRNAs and/or nucleic acids discussed in this application. The expression profile that is generated from the patient will be one that provides information regarding the particular disease or 15 condition. In many embodiments, the profile is generated using nucleic acid hybridization or amplification, (e.g., array hybridization or RT-PCR). In certain aspects, an expression profile can be used in conjunction with other diagnostic and/or prognostic tests, such as histology, protein profiles in the serum and/or cytogenetic assessment. -81 - WO 2008/036718 PCT/US2007/078859 11 0 4b4 CSo >~ ow o~~~~~ ~' id~2C oO (N ~ ( 5) Su U Co Z3 0 (. 0. o (N 'Z o Oo m m 51C mo~ w0 0~ ~~~~~ .( C o s~C~ .2 ~~~~ -2s El~. ~>~ 00C 19,--0 u 0 Co~ oU. 0o 0- o WO 2008/036718 PCT/US2007/078859 The methods can further comprise one or more of the steps including: (a) obtaining a sample from the patient, (b) isolating nucleic acids from the sample, (c) labeling the nucleic acids isolated from the sample, and (d) hybridizing the labeled nucleic acids to one or more probes. Nucleic acids of the invention include one or more nucleic acid comprising at least 5 one segment having a sequence or complementary sequence of to a nucleic acid representative of one or more of genes or markers in Table 1, 3, 4, and/or 5. It is contemplated that any method or composition described herein can be implemented with respect to any other method or composition described herein and that different embodiments may be combined. It is specifically contemplated that any methods 10 and compositions discussed herein with respect to miRNA molecules, miRNA, genes, and nucleic acids representative of genes may be implemented with respect to synthetic nucleic acids. In some embodiments the synthetic nucleic acid is exposed to the proper conditions to allow it to become a processed or mature nucleic acid, such as a miRNA under physiological circumstances. The claims originally filed are contemplated to cover claims that are multiply 15 dependent on any filed claim or combination of filed claims. Also, any embodiment of the invention involving specific genes (including representative fragments there of), mRNA, or miRNAs by name is contemplated also to cover embodiments involving miRNAs whose sequences are at least 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99% identical to the mature sequence of the 20 specified miRNA. It will be further understood that shorthand notations are employed such that a generic description of a gene or marker thereof, or of an miRNA refers to any of its gene family members (distinguished by a number) or representative fragments thereof, unless otherwise indicated. It is understood by those of skill in the art that a "gene family" refers to a group of 25 genes having the same coding sequence or miRNA coding sequence. Typically, miRNA members of a gene family are identified by a number following the initial designation. For example, miR-16-1 and miR-16-2 are members of the miR-16 gene family and "mir-7" refers to miR-7-1, miR-7-2 and miR-7-3. Moreover, unless otherwise indicated, a shorthand notation refers to related miRNAs (distinguished by a letter). Exceptions to these shorthand 30 notations will be otherwise identified. - 83 - WO 2008/036718 PCT/US2007/078859 Other embodiments of the invention are discussed throughout this application. Any embodiment discussed with respect to one aspect of the invention applies to other aspects of the invention as well and vice versa. The embodiments in the Example and Detailed Description section are understood to be embodiments of the invention that are applicable to 5 all aspects of the invention. The terms "inhibiting," "reducing," or "prevention," or any variation of these terms, when used in the claims and/or the specification includes any measurable decrease or complete inhibition to achieve a desired result. The use of the word "a" or "an" when used in conjunction with the term "comprising" 10 in the claims and/or the specification may mean "one," but it is also consistent with the meaning of "one or more," "at least one," and "one or more than one." Throughout this application, the term "about" is used to indicate that a value includes the standard deviation of error for the device or method being employed to determine the value. 15 The use of the term "or" in the claims is used to mean "and/or" unless explicitly indicated to refer to alternatives only or the alternatives are mutually exclusive, although the disclosure supports a definition that refers to only alternatives and "and/or." As used in this specification and claim(s), the words "comprising" (and any form of comprising, such as "comprise" and "comprises"), "having" (and any form of having, such as 20 "have" and "has"), "including" (and any form of including, such as "includes" and "include") or "containing" (and any form of containing, such as "contains" and "contain") are inclusive or open-ended and do not exclude additional, unrecited elements or method steps. Other objects, features and advantages of the present invention will become apparent from the following detailed description. It should be understood, however, that the detailed 25 description and the specific examples, while indicating specific embodiments of the invention, are given by way of illustration only, since various changes and modifications within the spirit and scope of the invention will become apparent to those skilled in the art from this detailed description. - 84 - WO 2008/036718 PCT/US2007/078859 DETAILED DESCRIPTION OF THE INVENTION The present invention is directed to compositions and methods relating to the identification and characterization of genes and biological pathways related to these genes as represented by the expression of the identified genes, as well as use of miRNAs related to 5 such, for therapeutic, prognostic, and diagnostic applications, particularly those methods and compositions related to assessing and/or identifying pathological conditions directly or indirectly related to miR-143 expression or the aberrant expression thereof. In certain aspects, the invention is directed to methods for the assessment, analysis, and/or therapy of a cell or subject where certain genes have a reduced or increased expression 10 (relative to normal) as a result of an increased or decreased expression of any one or a combination of miR-143 family members (including, but not limited to lla-mir-143 M10002552; xtr-mir-143 M10004937; dre-mir-143-2 M10002008; rno-mir-143 MI0000916; ptr-mir-143 M10002549; ppy-mir-143 MI0002551; ggo-mir-143 M10002550; dre-mir-143-1 M10002007; hsa-mir-143 M10000459; ppa-mir-143 M10002553; mdo-mir-143 MI0005302; 15 and mmu-mir-143 M10000257) and/or genes with an increased expression (relative to normal) as a result of decreased expression thereof. The expression profile and/or response to miR-143 expression or lack of expression may be indicative of an individual with a pathological condition, e.g., cancer. Prognostic assays featuring any one or combination of the miRNAs listed or the 20 markers listed (including nucleic acids representative thereof) could be used in assessment of a patient to determine what if any treatment regimen is justified. As with the diagnostic assays mentioned above, the absolute values that define low expression will depend on the platform used to measure the miRNA(s). The same methods described for the diagnostic assays could be used for prognostic assays. 25 I. THERAPEUTIC METHODS Embodiments of the invention concern nucleic acids that perform the activities of or inhibit endogenous miRNAs when introduced into cells, In certain aspects, nucleic acids are synthetic or non-synthetic miRNA. Sequence-specific miRNA inhibitors can be used to inhibit sequentially or in combination the activities of one or more endogenous miRNAs in 30 cells, as well those genes and associated pathways modulated by the endogenous miRNA. - 85 - WO 2008/036718 PCT/US2007/078859 The present invention concerns, in some embodiments, short nucleic acid molecules that function as miRNAs or as inhibitors of miRNA in a cell. The term "short" refers to a length of a single polynucleotide that is 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 50, 100, or 150 nucleotides or fewer,, including all integers or ranges derivable there between. The 5 nucleic acid molecules are typically synthetic. The term "synthetic" refers to a nucleic acid molecule that is not produced naturally in a cell. In certain aspects the chemical structure deviates from a naturally-occurring nucleic acid molecule, such as an endogenous precursor miRNA or miRNA molecule or complement thereof. While in some embodiments, nucleic acids of the invention do not have an entire sequence that is identical or complementary to a 10 sequence of a naturally-occurring nucleic acid, such molecules may encompass all or part of a naturally-occurring sequence or a complement thereof. It is contemplated, however, that a synthetic nucleic acid administered to a cell may subsequently be modified or altered in the cell such that its structure or sequence is the same as non-synthetic or naturally occurring nucleic acid, such as a mature miRNA sequence. For example, a synthetic nucleic acid may 15 have a sequence that differs from the sequence of a precursor miRNA, but that sequence may be altered once in a cell to be the same as an endogenous, processed miRNA or an inhibitor thereof. The term "isolated" means that the nucleic acid molecules of the invention are initially separated from different (in terms of sequence or structure) and unwanted nucleic acid molecules such that a population of isolated nucleic acids is at least about 90% 20 homogenous, and may be at least about 95, 96, 97, 98, 99, or 100% homogenous with respect to other polynucleotide molecules. In many embodiments of the invention, a nucleic acid is isolated by virtue of it having been synthesized in vitro separate from endogenous nucleic acids in a cell. It will be understood, however, that isolated nucleic acids may be subsequently mixed or pooled together. In certain aspects, synthetic miRNA of the invention 25 are RNA or RNA analogs. miRNA inhibitors may be DNA or RNA, or analogs thereof miRNA and miRNA inhibitors of the invention are collectively referred to as "synthetic nucleic acids." In some embodiments, there is a miRNA or a synthetic miRNA having a length of between 17 and 130 residues. The present invention concerns miRNA or synthetic miRNA 30 molecules that are, are at least, or are at most 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46,47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, - 86 - WO 2008/036718 PCT/US2007/078859 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114, 115, 116, 117, 118, 119, 120, 121,122,123,124,125,126,127,128,129,130,140,145,150,160,170, 180, 190,200 or more residues in length, including any integer or any range there between. In certain embodiments, synthetic miRNA have (a) a "miRNA region" whose 5 sequence or binding region from 5' to 3' is identical or complementary to all or a segment of a mature miRNA sequence, and (b) a "complementary region" whose sequence from 5' to 3' is between 60% and 100% complementary to the miRNA sequence in (a). In certain embodiments, these synthetic miRNA are also isolated, as defined above. The term "miRNA region" refers to a region on the synthetic miRNA that is at least 75, 80, 85, 90, 95, or 100% 10 identical, including all integers there between, to the entire sequence of a mature, naturally occurring miRNA sequence or a complement thereof. In certain embodiments, the miRNA region is or is at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 99.1, 99.2, 99.3, 99.4, 99.5, 99.6, 99.7, 99.8, 99.9 or 100% identical to the sequence of a naturally-occurring miRNA or complement thereof. 15 The term "complementary region" or "complement" refers to a region of a nucleic acid or mimetic that is or is at least 60% complementary to the mature, naturally occurring miRNA sequence. The complementary region is or is at least 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 99.1, 99.2, 99.3, 99.4, 99.5, 99.6, 99.7, 99.8, 99.9 or 100% 20 complementary, or any range derivable therein. With single polynucleotide sequences, there may be a hairpin loop structure as a result of chemical bonding between the miRNA region and the complementary region. In other embodiments, the complementary region is on a different nucleic acid molecule than the miRNA region, in which case the complementary region is on the complementary strand and the miRNA region is on the active strand. 25 In other embodiments of the invention, there are synthetic nucleic acids that are miRNA inhibitors. A miRNA inhibitor is between about 17 to 25 nucleotides in length and comprises a 5' to 3' sequence that is at least 90% complementary to the 5' to 3' sequence of a mature miRNA. In certain embodiments, a miRNA inhibitor molecule is 17, 18, 19, 20, 21, 22, 23, 24, or 25 nucleotides in length, or any range derivable therein. Moreover, an miRNA 30 inhibitor may have a sequence (from 5' to 3') that is or is at least 70, 75, 80, 85, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 99.1, 99.2, 99.3, 99.4, 99.5, 99.6, 99.7, 99.8, 99.9 or 100% complementary, or any range derivable therein, to the 5' to 3' sequence of a mature miRNA, - 87 - WO 2008/036718 PCT/US2007/078859 particularly a mature, naturally occurring miRNA. One of skill in the art could use a portion of the miRNA sequence that is complementary to the sequence of a mature miRNA as the sequence for a miRNA inhibitor. Moreover, that portion of the nucleic acid sequence can be altered so that it is still comprises the appropriate percentage of complementarity to the 5 sequence of a mature miRNA. In some embodiments, of the invention, a synthetic miRNA or inhibitor contains one or more design element(s). These design elements include, but are not limited to: (i) a replacement group for the phosphate or hydroxyl of the nucleotide at the 5' terminus of the complementary region; (ii) one or more sugar modifications in the first or last 1 to 6 residues 10 of the complementary region; or, (iii) noncomplementarity between one or more nucleotides in the last 1 to 5 residues at the 3' end of the complementary region and the corresponding nucleotides of the miRNA region. A variety of design modifications are known in the art, see below. In certain embodiments, a synthetic miRNA has a nucleotide at its 5' end of the 15 complementary region in which the phosphate and/or hydroxyl group has been replaced with another chemical group (referred to as the "replacement design"). In some cases, the phosphate group is replaced, while in others, the hydroxyl group has been replaced. In particular embodiments, the replacement group is biotin, an amine group, a lower alkylamine group, an acetyl group, 2'O-Me (2'oxygen-methyl), DMTO (4,4'-dimethoxytrityl with 20 oxygen), fluoroscein, a thiol, or acridine, though other replacement groups are well known to those of skill in the art and can be used as well. This design element can also be used with a miRNA inhibitor. . Additional embodiments concern a synthetic miRNA having one or more sugar modifications in the first or last 1 to 6 residues of the complementary region (referred to as 25 the "sugar replacement design"). In certain cases, there is one or more sugar modifications in the first 1, 2, 3, 4, 5, 6 or more residues of the complementary region, or any range derivable therein. In additional cases, there is one or more sugar modifications in the last 1, 2, 3, 4, 5, 6 or more residues of the complementary region, or any range derivable therein, have a sugar modification. It will be understood that the terms "first" and "last" are with respect to the 30 order of residues from the 5' end to the 3' end of the region. In particular embodiments, the sugar modification is a 2'O-Me modification. In further embodiments, there is one or more sugar modifications in the first or last 2 to 4 residues of the complementary region or the first - 88 - WO 2008/036718 PCT/US2007/078859 or last 4 to 6 residues of the complementary region. This design element can also be used with an miRNA inhibitor. Thus, an miRNA inhibitor can have this design element and/or a replacement group on the nucleotide at the 5' terminus, as discussed above. In other embodiments of the invention, there is a synthetic miRNA or inhibitor in 5 which one or more nucleotides in the last 1 to 5 residues at the 3' end of the complementary region are not complementary to the corresponding nucleotides of the miRNA region ("noncomplementarity") (referred to as the "noncomplementarity design"). The noncomplementarity may be in the last 1, 2, 3, 4, and/or 5 residues of the complementary miRNA. In certain embodiments, there is noncomplementarity with at least 2 nucleotides in 10 the complementary region. It is contemplated that synthetic miRNA of the invention have one or more of the replacement, sugar modification, or noncomplementarity designs. In certain cases, synthetic RNA molecules have two of them, while in others these molecules have all three designs in place. 15 The miRNA region and the complementary region may be on the same or separate polynucleotides. In cases in which they are contained on or in the same polynucleotide, the miRNA molecule will be considered a single polynucleotide. In embodiments in which the different regions are on separate polynucleotides, the synthetic miRNA will be considered to be comprised of two polynucleotides. 20 When the RNA molecule is a single polynucleotide, there can be a linker region between the miRNA region and the complementary region. In some embodiments, the single polynucleotide is capable of forming a hairpin loop structure as a result of bonding between the miRNA region and the complementary region. The linker constitutes the hairpin loop. It is contemplated that in some embodiments, the linker region is, is at least, or is at most 2, 3, 25 4, 5,6,7, 8,9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, or 40 residues in length, or any range derivable therein. In certain embodiments, the linker is between 3 and 30 residues (inclusive) in length, In addition to having a miRNA or inhibitor region and a complementary region, there may be flanking sequences as well at either the 5' or 3' end of the region. In some 30 embodiments, there is or is at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 nucleotides or more, or any range derivable therein, flanking one or both sides of these regions. - 89 - WO 2008/036718 PCT/US2007/078859 Methods of the invention include reducing or eliminating activity of one or more miRNAs in a cell comprising introducing into a cell a miRNA inhibitor (which may be described generally herein as an miRNA, so that a description of miRNA, where appropriate, also will refer to a miRNA inhibitor); or supplying or enhancing the activity of one or more 5 miRNAs in a cell. The present invention also concerns inducing certain cellular characteristics by providing to a cell a particular nucleic acid, such as a specific synthetic miRNA molecule or a synthetic miRNA inhibitor molecule. However, in methods of the invention, the miRNA molecule or miRNA inhibitor need not be synthetic. They may have a sequence that is identical to a naturally occurring miRNA or they may not have any design 10 modifications. In certain embodiments, the miRNA molecule and/or the miRNA inhibitor are synthetic, as discussed above. The particular nucleic acid molecule provided to the cell is understood to correspond to a particular miRNA in the cell, and thus, the miRNA in the cell is referred to as the "corresponding miRNA." In situations in which a named miRNA molecule is introduced into 15 a cell, the corresponding miRNA will be understood to be the induced or inhibited miRNA or induced or inhibited miRNA function. It is contemplated, however, that the miRNA molecule introduced into a cell is not a mature miRNA but is capable of becoming or functioning as a mature miRNA under the appropriate physiological conditions. In cases in which a particular corresponding miRNA is being inhibited by a miRNA inhibitor, the 20 particular miRNA will be referred to as the "targeted miRNA." It is contemplated that multiple corresponding miRNAs may be involved. In particular embodiments, more than one miRNA molecule is introduced into a cell. Moreover, in other embodiments, more than one miRNA inhibitor is introduced into a cell. Furthermore, a combination of miRNA molecule(s) and miRNA inhibitor(s) may be introduced into a cell. The inventors 25 contemplate that a combination of miRNA may act at one or more points in cellular pathways of cells with aberrant phenotypes and that such combination may have increased efficacy on the target cell while not adversely effecting normal cells. Thus, a combination of miRNA may have a minimal adverse effect on a subject or patient while supplying a sufficient therapeutic effect, such as amelioration of a condition, growth inhibition of a cell, death of a 30 targeted cell, alteration of cell phenotype or physiology, slowing of cellular growth, sensitization to a second therapy, sensitization to a particular therapy, and the like. - 90 - WO 2008/036718 PCT/US2007/078859 Methods include identifying a cell or patient in need of inducing those cellular characteristics. Also, it will be understood that an amount of a synthetic nucleic acid that is provided to a cell or organism is an "effective amount," which refers to an amount needed (or a sufficient amount) to achieve a desired goal, such as inducing a particular cellular 5 characteristic(s). Certain embodiments of the methods include providing or introducing to a cell a nucleic acid molecule corresponding to a mature miRNA in the cell in an amount effective to achieve a desired physiological result. Moreover, methods can involve providing synthetic or nonsynthetic miRNA molecules. It is contemplated that in these embodiments, that the methods may or may not be 10 limited to providing only one or more synthetic miRNA molecules or only one or more nonsynthetic miRNA molecules. Thus, in certain embodiments, methods may involve providing both synthetic and nonsynthetic miRNA molecules. In this situation, a cell or cells are most likely provided a synthetic miRNA molecule corresponding to a particular miRNA and a nonsynthetic miRNA molecule corresponding to a different miRNA. Furthermore, any 15 method articulated using a list of miRNAs using Markush group language may be articulated without the Markush group language and a disjunctive article (i.e., or) instead, and vice versa. Typically, an endogenous gene, miRNA or mRNA is modulated in the cell. In particular embodiments, the nucleic acid sequence comprises at least one segment that is at least 70, 75, 80, 85, 90, 95, or 100% identical in nucleic acid sequence to one or more 20 miRNA or gene sequence. Modulation of the expression or processing of an endogenous gene, miRNA, or mRNA can be through modulation of the processing of a mRNA, such processing including transcription, transportation and/or translation with in a cell. Modulation may also be effected by the inhibition or enhancement of miRNA activity with a cell, tissue, or organ. Such processing may affect the expression of an encoded product or the 25 stability of the mRNA. In still other embodiments, a nucleic acid sequence can comprise a modified nucleic acid sequence. In certain aspects, one or more miRNA sequence may include or comprise a modified nucleobase or nucleic acid sequence. It will be understood in methods of the invention that a cell or other biological matter such as an organism (including patients) can be provided a miRNA or miRNA molecule 30 corresponding to a particular miRNA by administering to the cell or organism a nucleic acid molecule that functions as the corresponding miRNA once inside the cell. The form of the molecule provided to the cell may not be the form that acts a miRNA once inside the cell. -91 - WO 2008/036718 PCT/US2007/078859 Thus, it is contemplated that in some embodiments, a synthetic miRNA or a nonsynthetic miRNA is provided such that it becomes processed into a mature and active miRNA once it has access to the cell's miRNA processing machinery. In certain embodiments, it is specifically contemplated that the miRNA molecule provided is not a mature miRNA 5 molecule but a nucleic acid molecule that can be processed into the mature miRNA once it is accessible to miRNA processing machinery. The term "nonsynthetic" in the context of miRNA means that the miRNA is not "synthetic," as defined herein. Furthermore, it is contemplated that in embodiments of the invention that concern the use of synthetic miRNAs, the use of corresponding nonsynthetic miRNAs is also considered an aspect of the invention, 10 and vice versa. It will be understand that the term "providing" an agent is used to include "administering" the agent to a patient. In certain embodiments, methods also include targeting a miRNA to modulate in a cell or organism. The term "targeting a miRNA to modulate" means a nucleic acid of the invention will be employed so as to modulate the selected miRNA. In some embodiments the 15 modulation is achieved with a synthetic or non-synthetic miRNA that corresponds to the targeted miRNA, which effectively provides the targeted miRNA to the cell or organism (positive modulation). In other embodiments, the modulation is achieved with a miRNA inhibitor, which effectively inhibits the targeted miRNA in the cell or organism (negative modulation). 20 In some embodiments, the miRNA targeted to be modulated is a miRNA that affects a disease, condition, or pathway. In certain embodiments, the miRNA is targeted because a treatment can be provided by negative modulation of the targeted miRNA. In other embodiments, the miRNA is targeted because a treatment can be provided by positive modulation of the targeted miRNA or its targets. 25 In certain methods of the invention, there is a further step of administering the selected miRNA modulator to a cell, tissue, organ, or organism (collectively "biological matter") in need of treatment related to modulation of the targeted miRNA or in need of the physiological or biological results discussed herein (such as with respect to a particular cellular pathway or result like decrease in cell viability). Consequently, in some methods of 30 the invention there is a step of identifying a patient in need of treatment that can be provided by the miRNA modulator(s). It is contemplated that an effective amount of a miRNA modulator can be administered in some embodiments. In particular embodiments, there is a - 92 - WO 2008/036718 PCT/US2007/078859 therapeutic benefit conferred on the biological matter, where a "therapeutic benefit" refers to an improvement in the one or more conditions or symptoms associated with a disease or condition or an improvement in the prognosis, duration, or status with respect to the disease. It is contemplated that a therapeutic benefit includes, but is not limited to, a decrease in pain, 5 a decrease in morbidity, a decrease in a symptom. For example, with respect to cancer, it is contemplated that a therapeutic benefit can be inhibition of tumor growth, prevention of metastasis, reduction in number of metastases, inhibition of cancer cell proliferation, induction of cell death in cancer cells, inhibition of angiogenesis near cancer cells, induction of apoptosis of cancer cells, reduction in pain, reduction in risk of recurrence, induction of 10 chemo- or radiosensitivity in cancer cells, prolongation of life, and/or delay of death directly or indirectly related to cancer. Furthermore, it is contemplated that the miRNA compositions may be provided as part of a therapy to a patient, in conjunction with traditional therapies or preventative agents. Moreover, it is contemplated that any method discussed in the context of therapy may be 15 applied preventatively, particularly in a patient identified to be potentially in need of the therapy or at risk of the condition or disease for which a therapy is needed. In addition, methods of the invention concern employing one or more nucleic acids corresponding to a miRNA and a therapeutic drug. The nucleic acid can enhance the effect or efficacy of the drug, reduce any side effects or toxicity, modify its bioavailability, and/or 20 decrease the dosage or frequency needed. In certain embodiments, the therapeutic drug is a cancer therapeutic. Consequently, in some embodiments, there is a method of treating cancer in a patient comprising administering to the patient the cancer therapeutic and an effective amount of at least one miRNA molecule that improves the efficacy of the cancer therapeutic or protects non-cancer cells. Cancer therapies also include a variety of combination therapies 25 with both chemical and radiation based treatments. Combination chemotherapies include but are not limited to, for example, 5-fluorouracil, alemtuzumab, amrubicin, bevacizumab, bleomycin, bortezomib, busulfan, camptothecin, capecitabine, carboplatin, cetuximab, chlorambucil, cisplatin (CDDP), COX-2 inhibitors (e.g., celecoxib), cyclophosphamide, cytarabine, dactinomycin, dasatinib, daunorubicin, dexamethasone, docetaxel, doxorubicin 30 (adriamycin), EGFR inhibitors (gefitinib and cetuximab), erlotinib, estrogen receptor binding agents, etoposide (VP16), everolimus, farnesyl-protein transferase inhibitors, gefitinib, gemcitabine, gemtuzumab, ibritumomab, ifosfamide, imatinib mesylate, larotaxel, lapatinib, - 93 - WO 2008/036718 PCT/US2007/078859 lonafarnib, mechlorethamine, melphalan, methotrexate, mitomycin, navelbine, nitrosurea, nocodazole, oxaliplatin, paclitaxel, plicomycin, procarbazine, raloxifene, rituximab, sirolimus, sorafenib, sunitinib, tamoxifen, taxol, taxotere, temsirolimus, tipifarib, tositumomab, transplatinum, trastuzumab, vinblastin, vincristin, or vinorelbine or any analog 5 or derivative variant of the foregoing. Generally, inhibitors of miRNAs can be given to decrease the activity of an endogenous miRNA. For example, inhibitors of miRNA molecules that increase cell proliferation can be provided to cells to decrease cell proliferation. The present invention contemplates these embodiments in the context of the different physiological effects observed 10 with the different miRNA molecules and miRNA inhibitors disclosed herein. These include, but are not limited to, the following physiological effects: increase and decreasing cell proliferation, increasing or decreasing apoptosis, increasing transformation, increasing or decreasing cell viability, activating or inhibiting a kinase (e.g., Erk), activating/inducing or inhibiting hTert, inhibit stimulation of growth promoting pathway (e.g., Stat 3 signaling), 15 reduce or increase viable cell number, and increase or decrease number of cells at a particular phase of the cell cycle. Methods of the invention are generally contemplated to include providing or introducing one or more different nucleic acid molecules corresponding to one or more different miRNA molecules. It is contemplated that the following, at least the following, or at most the following number of different nucleic acid or miRNA molecules 20 may be provided or introduced: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20,21,22,23,24,25,26,27,28,29,30, 31,32,33,34,35,36,37,38,39,40,41,42,43,44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, or any range derivable therein. This also applies to the number of 25 different miRNA molecules that can be provided or introduced into a cell. II. PHARMACEUTICAL FORMULATIONS AND DELIVERY Methods of the present invention include the delivery of an effective amount of a miRNA or an expression construct encoding the same. An "effective amount" of the pharmaceutical composition, generally, is defined as that amount sufficient to detectably and 30 repeatedly to achieve the stated desired result, for example, to ameliorate, reduce, minimize or limit the extent of the disease or its symptoms. Other more rigorous definitions may apply, including elimination, eradication or cure of disease. - 94 - WO 2008/036718 PCT/US2007/078859 A. Administration In certain embodiments, it is desired to kill cells, inhibit cell growth, inhibit metastasis, decrease tumor or tissue size, and/or reverse or reduce the malignant or disease phenotype of cells. The routes of administration will vary, naturally, with the location and 5 nature of the lesion or site to be targeted, and include, e.g., intradermal, subcutaneous, regional, parenteral, intravenous, intramuscular, intranasal, systemic, and oral administration and formulation. Direct injection, intratumoral injection, or injection into tumor vasculature is specifically contemplated for discrete, solid, accessible tumors, or other accessible target areas. Local, regional, or systemic administration also may be appropriate. For tumors of >4 10 cm, the volume to be administered will be about 4-10 ml (preferably 10 ml), while for tumors of<4 cm, a volume of about 1-3 ml will be used (preferably 3 ml). Multiple injections delivered as a single dose comprise about 0.1 to about 0.5 ml volumes. Compositions of the invention may be administered in multiple injections to a tumor or a targeted site. In certain aspects, injections may be spaced at approximately 1 cm 15 intervals. In the case of surgical intervention, the present invention may be used preoperatively, to render an inoperable tumor subject to resection. Alternatively, the present invention may be used at the time of surgery, and/or thereafter, to treat residual or metastatic disease. For example, a resected tumor bed may be injected or perfused with a formulation comprising a 20 miRNA or combinations thereof. Administration may be continued post-resection, for example, by leaving a catheter implanted at the site of the surgery. Periodic post-surgical treatment also is envisioned. Continuous perfusion of an expression construct or a viral construct also is contemplated. Continuous administration also maybe applied where appropriate, for example, where 25 a tumor or other undesired affected area is excised and the tumor bed or targeted site is treated to eliminate residual, microscopic disease. Delivery via syringe or catherization is contemplated. Such continuous perfusion may take place for a period from about 1-2 hours, to about 2-6 hours, to about 6-12 hours, to about 12-24 hours, to about 1-2 days, to about 1-2 wk or longer following the initiation of treatment. Generally, the dose of the therapeutic 30 composition via continuous perfusion will be equivalent to that given by a single or multiple injections, adjusted over a period of time during which the perfusion occurs. - 95 - WO 2008/036718 PCT/US2007/078859 Treatment regimens may vary as well and often depend on tumor type, tumor location, immune condition, target site, disease progression, and health and age of the patient. Certain tumor types will require more aggressive treatment. The clinician will be best suited to make such decisions based on the known efficacy and toxicity (if any) of the therapeutic 5 formulations. In certain embodiments, the tumor or affected area being treated may not, at least initially, be resectable. Treatments with compositions of the invention may increase the resectability of the tumor due to shrinkage at the margins or by elimination of certain particularly invasive portions. Following treatments, resection may be possible. Additional 10 treatments subsequent to resection may serve to eliminate microscopic residual disease at the tumor or targeted site. Treatments may include various "unit doses," A unit dose is defined as containing a predetermined quantity of a therapeutic composition(s). The quantity to be administered, and the particular route and formulation, are within the skill of those in the clinical arts. A unit 15 dose need not be administered as a single injection but may comprise continuous infusion over a set period of time. With respect to a viral component of the present invention, a unit dose may conveniently be described in terms of ig or mg of miRNA or miRNA mimetic. Alternatively, the amount specified may be the amount administered as the average daily, average weekly, or average monthly dose. 20 miRNA can be administered to the patient in a dose or doses of about or of at least about 0.5, 1, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300, 310, 320, 330, 340, 350, 360, 370, 380, 390, 400, 410, 420, 430, 440, 450, 460, 470, 480, 490, 500, 510, 520, 530, 540, 550, 560, 570, 580, 590, 600, 610, 620, 630, 640, 650, 660, 670, 680, 690, 25 700, 710, 720, 730, 740, 750, 760, 770, 780, 790, 800, 810, 820, 830, 840, 850, 860, 870, 880, 890, 900, 910, 920, 930, 940, 950, 960, 970, 980, 990, 1000 pLg or mg, or more, or any range derivable therein. Alternatively, the amount specified may be the amount administered as the average daily, average weekly, or average monthly dose, or it may be expressed in terms of mg/kg, where kg refers to the weight of the patient and the mg is specified above. In 30 other embodiments, the amount specified is any number discussed above but expressed as mg/m 2 (with respect to tumor size or patient surface area). -96- WO 2008/036718 PCT/US2007/078859 B. Injectable Compositions and Formulations In some embodiments, the method for the delivery of a miRNA or an expression construct encoding such or combinations thereof is via systemic administration. However, the pharmaceutical compositions disclosed herein may also be administered parenterally, 5 subcutaneously, directly, intratracheally, intravenously, intradermally, intramuscularly, or even intraperitoneally as described in U.S. Patents 5,543,158; 5,641,515 and 5,399,363 (each specifically incorporated herein by reference in its entirety). Injection of nucleic acids may be delivered by syringe or any other method used for injection of a solution, as long as the nucleic acid and any associated components can pass 10 through the particular gauge of needle required for injection. A syringe system has also been described for use in gene therapy that permits multiple injections of predetermined quantities of a solution precisely at any depth (U.S. Patent 5,846,225). Solutions of the active compounds as free base or pharmacologically acceptable salts may be prepared in water suitably mixed with a surfactant, such as hydroxypropyleellulose. 15 Dispersions may also be prepared in glycerol, liquid polyethylene glycols, mixtures thereof, and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms. The pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions (U.S. Patent 20 5,466,468, specifically incorporated herein by reference in its entirety). In all cases the form must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (e.g., glycerol, 25 propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and/or vegetable oils. Proper fluidity may be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. The prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, 30 chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars or sodium chloride. Prolonged - 97 - WO 2008/036718 PCT/US2007/078859 absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin. In certain formulations, a water-based formulation is employed while in others, it may be lipid-based. In particular embodiments of the invention, a composition comprising a 5 tumor suppressor protein or a nucleic acid encoding the same is in a water-based formulation. In other embodiments, the formulation is lipid based. For parenteral administration in an aqueous solution, for example, the solution should be suitably buffered if necessary and the liquid diluent first rendered isotonic with sufficient saline or glucose. These particular aqueous solutions are especially suitable for intravenous, 10 intramuscular, subcutaneous, intratumoral, intralesional, and intraperitoneal administration. In this connection, sterile aqueous media which can be employed will be known to those of skill in the art in light of the present disclosure, For example, one dosage may be dissolved in 1 ml of isotonic NaCl solution and either added to 1000 ml of hypodermoclysis fluid or injected at the proposed site of infusion, (see for example, "Remington's Pharmaceutical 15 Sciences" 15th Edition, pages 1035-1038 and 1570-1580). Some variation in dosage will necessarily occur depending on the condition of the subject being treated. The person responsible for administration will, in any event, determine the appropriate dose for the individual subject. Moreover, for human administration, preparations should meet sterility, pyrogenicity, general safety and purity standards as required by FDA Office of Biologics 20 standards. As used herein, a "carrier" includes any and all solvents, dispersion media, vehicles, coatings, diluents, antibacterial and antifungal agents, isotonic and absorption delaying agents, buffers, carrier solutions, suspensions, colloids, and the like. The use of such media and agents for pharmaceutical active substances is well known in the art. Except insofar as 25 any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions is contemplated. Supplementary active ingredients can also be incorporated into the compositions. The phrase "pharmaceutically acceptable" refers to molecular entities and compositions that do not produce an allergic or similar untoward reaction when administered 30 to a human. -98 - WO 2008/036718 PCT/US2007/078859 The nucleic acid(s) are administered in a manner compatible with the dosage formulation, and in such amount as will be therapeutically effective. The quantity to be administered depends on the subject to be treated, including, e.g., the aggressiveness of the disease or cancer, the size of any tumor(s) or lesions, the previous or other courses of 5 treatment. Precise amounts of active ingredient required to be administered depend on the judgment of the practitioner. Suitable regimes for initial administration and subsequent administration are also variable, but are typified by an initial administration followed by other administrations. Such administration may be systemic, as a single dose, continuous over a period of time spanning 10, 20, 30, 40, 50, 60 minutes, and/or 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 10 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24 or more hours, and/or 1, 2, 3, 4, 5, 6, 7, days or more. Moreover, administration may be through a time release or sustained release mechanism, implemented by formulation and/or mode of administration. C. Combination Treatments In certain embodiments, the compositions and methods of the present invention 15 involve a miRNA, or expression construct encoding such. These miRNA composition can be used in combination with a second therapy to enhance the effect of the miRNA therapy, or increase the therapeutic effect of another therapy being employed. These compositions would be provided in a combined amount effective to achieve the desired effect, such as the killing of a cancer cell and/or the inhibition of cellular hyperproliferation. This process may 20 involve contacting the cells with the miRNA or second therapy at the same or different time. This may be achieved by contacting the cell with one or more compositions or pharmacological formulation that includes or more of the agents, or by contacting the cell with two or more distinct compositions or formulations, wherein one composition provides (1) miRNA; and/or (2) a second therapy. A second composition or method may be 25 administered that includes a chemotherapy, radiotherapy, surgical therapy, immunotherapy or gene therapy. It is contemplated that one may provide a patient with the miRNA therapy and the second therapy within about 12-24 h of each other and, more preferably, within about 6-12 h of each other. In some situations, it may be desirable to extend the time period for treatment 30 significantly, however, where several days (2, 3, 4, 5, 6 or 7) to several weeks (1, 2, 3, 4, 5, 6, 7 or 8) lapse between the respective administrations. - 99 - WO 2008/036718 PCT/US2007/078859 In certain embodiments, a course of treatment will last 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 5 87, 88, 89, 90 days or more. It is contemplated that one agent may be given on day 1, 2, 3, 4, 5,6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, and/or 90, any combination thereof, and another agent is 10 given on day 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, and/or 90, or any combination thereof. Within a single day (24-hour period), the patient may be given one or multiple 15 administrations of the agent(s). Moreover, after a course of treatment, it is contemplated that there is a period of time at which no treatment is administered. This time period may last 1, 2, 3, 4, 5, 6, 7 days, and/or 1, 2, 3, 4, 5 weeks, and/or 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 months or more, depending on the condition of the patient, such as their prognosis, strength, health, etc. 20 Various combinations may be employed, for example miRNA therapy is "A" and a second therapy is "B": A/B/A B/A/B B/B/A A/A/B A/B/B B/A/A A/B/B/B B/A/B/B B/B/B/A B/B/A/B A/A/B/B A/B/A/B A/B/B/A B/B/A/A B/A/B/A B/A/A/B A/A/A/B B/A/A/A A/B/A/A A/A/B/A 25 Administration of any compound or therapy of the present invention to a patient will follow general protocols for the administration of such compounds, taking into account the toxicity, if any, of the vector or any protein or other agent. Therefore, in some embodiments there is a step of monitoring toxicity that is attributable to combination therapy. It is expected that the treatment cycles would be repeated as necessary. It also is contemplated 30 that various standard therapies, as well as surgical intervention, may be applied in combination with the described therapy. - 100- WO 2008/036718 PCT/US2007/078859 In specific aspects, it is contemplated that a second therapy, such as chemotherapy, radiotherapy, immunotherapy, surgical therapy or other gene therapy, is employed in combination with the miRNA therapy, as described herein. 1. Chemotherapy 5 A wide variety of chemotherapeutic agents may be used in accordance with the present invention. The term "chemotherapy" refers to the use of drugs to treat cancer. A "chemotherapeutic agent" is used to connote a compound or composition that is administered in the treatment of cancer. These agents or drugs are categorized by their mode of activity within a cell, for example, whether and at what stage they affect the cell cycle. Alternatively, 10 an agent may be characterized based on its ability to directly cross-link DNA, to intercalate into DNA, or to induce chromosomal and mitotic aberrations by affecting nucleic acid synthesis. Most chemotherapeutic agents fall into the following categories: alkylating agents, antimetabolites, antitumor antibiotics, mitotic inhibitors, and nitrosoureas. a. Alkylating agents 15 Alkylating agents are drugs that directly interact with genomic DNA to prevent the cancer cell from proliferating. This category of chemotherapeutic drugs represents agents that affect all phases of the cell cycle, that is, they are not phase-specific. Alkylating agents can be implemented to treat chronic leukemia, non-Hodgkin's lymphoma, Hodgkin's disease, multiple myeloma, and particular cancers of the breast, lung, and ovary. They include: 20 busulfan, chlorambucil, cisplatin, cyclophosphamide (cytoxan), dacarbazine, ifosfamide, mechlorethamine (mustargen), and melphalan. Troglitazaone can be used to treat cancer in combination with any one or more of these alkylating agents. b. Antimetabolites Antimetabolites disrupt DNA and RNA synthesis. Unlike alkylating agents, they 25 specifically influence the cell cycle during S phase. They have been used to combat chronic leukemias in addition to tumors of breast, ovary and the gastrointestinal tract. Antimetabolites include 5-fluorouracil (5-FU), cytarabine (Ara-C), fludarabine, gemcitabine, and methotrexate. 5-Fluorouracil (5-FU) has the chemical name of 5-fluoro-2,4(lH,3H) 30 pyrimidinedione. Its mechanism of action is thought to be by blocking the methylation - 101 - WO 2008/036718 PCT/US2007/078859 reaction of deoxyuridylic acid to thymidylic acid. Thus, 5-FU interferes with the synthesis of deoxyribonucleic acid (DNA) and to a lesser extent inhibits the formation of ribonucleic acid (RNA). Since DNA and RNA are essential for cell division and proliferation, it is thought that the effect of 5-FU is to create a thymidine deficiency leading to cell death. Thus, the 5 effect of 5-FU is found in cells that rapidly divide, a characteristic of metastatic cancers. c. Antitumor Antibiotics Antitumor antibiotics have both antimicrobial and cytotoxic activity. These drugs also interfere with DNA by chemically inhibiting enzymes and mitosis or altering cellular membranes. These agents are not phase specific so they work in all phases of the cell cycle. 10 Thus, they are widely used for a variety of cancers. Examples of antitumor antibiotics include bleomycin, dactinomycin, daunorubicin, doxorubicin (Adriamycin), and idarubicin, some of which are discussed in more detail below. Widely used in clinical setting for the treatment of neoplasms, these compounds are administered through bolus injections intravenously at doses ranging from 25-75 mg/m 2 at 21 day intervals for adriamycin, to 35 15 100 mg/m 2 for etoposide intravenously or orally. d. Mitotic Inhibitors Mitotic inhibitors include plant alkaloids and other natural agents that can inhibit either protein synthesis required for cell division or mitosis. They operate during a specific phase during the cell cycle. Mitotic inhibitors comprise docetaxel, etoposide (VP16), 20 paclitaxel, taxol, taxotere, vinblastine, vincristine, and vinorelbine. e. Nitrosureas Nitrosureas, like alkylating agents, inhibit DNA repair proteins. They are used to treat non-Hodgkin's lymphomas, multiple myeloma, malignant melanoma, in addition to brain tumors. Examples include carmustine and lomustine. 25 2. Radiotherapy Radiotherapy, also called radiation therapy, is the treatment of cancer and other diseases with ionizing radiation. Ionizing radiation deposits energy that injures or destroys cells in the area being treated by damaging their genetic material, making it impossible for these cells to continue to grow. Although radiation damages both cancer cells and normal 30 cells, the latter are able to repair themselves and function properly. Radiotherapy may be - 102 - WO 2008/036718 PCT/US2007/078859 used to treat localized solid tumors, such as cancers of the skin, tongue, larynx, brain, breast, or cervix. It can also be used to treat leukemia and lymphoma (cancers of the blood-forming cells and lymphatic system, respectively). Radiation therapy used according to the present invention may include, but is not 5 limited to, the use of y-rays, X-rays, and/or the directed delivery of radioisotopes to tumor cells. Other forms of DNA damaging factors are also contemplated such as microwaves, proton beam irradiation (U.S. Patents 5,760,395 and 4,870,287) and UV-irradiation. It is most likely that all of these factors effect a broad range of damage on DNA, on the precursors of DNA, on the replication and repair of DNA, and on the assembly and maintenance of 10 chromosomes. Dosage ranges for X-rays range from daily doses of 50 to 200 roentgens for prolonged periods of time (3 to 4 wk), to single doses of 2000 to 6000 roentgens. Dosage ranges for radioisotopes vary widely, and depend on the half-life of the isotope, the strength and type of radiation emitted, and the uptake by the neoplastic cells. Radiotherapy may comprise the use of radiolabeled antibodies to deliver doses of radiation directly to the cancer 15 site (radioimmunotherapy). Once injected into the body, the antibodies actively seek out the cancer cells, which are destroyed by the cell-killing (cytotoxic) action of the radiation. This approach can minimize the risk of radiation damage to healthy cells. Stereotactic radio-surgery (gamma knife) for brain and other tumors does not use a knife, but very precisely targeted beams of gamma radiotherapy from hundreds of different 20 angles. Only one session of radiotherapy, taking about four to five hours, is needed. For this treatment a specially made metal frame is attached to the head. Then, several scans and x rays are carried out to find the precise area where the treatment is needed. During the radiotherapy for brain tumors, the patient lies with their head in a large helmet, which has hundreds of holes in it to allow the radiotherapy beams through. Related approaches permit 25 positioning for the treatment of tumors in other areas of the body. 3. Immunotherapy In the context of cancer treatment, immunotherapeutics, generally, rely on the use of immune effector cells and molecules to target and destroy cancer cells. Trastuzumab (Herceptin T M ) is such an example. The immune effector may be, for example, an antibody 30 specific for some marker on the surface of a tumor cell. The antibody alone may serve as an effector of therapy or it may recruit other cells to actually affect cell killing. The antibody - 103 - WO 2008/036718 PCT/US2007/078859 also may be conjugated to a drug or toxin (chemotherapeutic, radionuclide, ricin A chain, cholera toxin, pertussis toxin, etc.) and serve merely as a targeting agent. Alternatively, the effector may be a lymphocyte carrying a surface molecule that interacts, either directly or indirectly, with a tumor cell target. Various effector cells include cytotoxic T cells and NK 5 cells. The combination of therapeutic modalities, i.e., direct cytotoxic activity and inhibition or reduction of ErbB2 would provide therapeutic benefit in the treatment of ErbB2 overexpressing cancers. In one aspect of immunotherapy, the tumor or disease cell must bear some marker that is amenable to targeting, i.e., is not present on the majority of other cells. Many tumor 10 markers exist and any of these may be suitable for targeting in the context of the present invention. Common tumor markers include carcinoembryonic antigen, prostate specific antigen, urinary tumor associated antigen, fetal antigen, tyrosinase (p97), gp68, TAG-72, HMFG, Sialyl Lewis Antigen, MucA, MucB, PLAP, estrogen receptor, laminin receptor, erb B and p 1 55. An alternative aspect of immunotherapy is to combine anticancer effects with 15 immune stimulatory effects. Immune stimulating molecules also exist including: cytokines such as IL-2, IL-4, IL-12, GM-CSF, gamma-IFN, chemokines such as MIP-1, MCP-1, IL-8 and growth factors such as FLT3 ligand. Combining immune stimulating molecules, either as proteins or using gene delivery in combination with a tumor suppressor such as MDA-7 has been shown to enhance anti-tumor effects (Ju et al., 2000). Moreover, antibodies against 20 any of these compounds can be used to target the anti-cancer agents discussed herein. Examples of immunotherapies currently under investigation or in use are immune adjuvants e.g., Mycobacterium bovis, Plasmodium falciparum, dinitrochlorobenzene and aromatic compounds (U.S. Patents 5,801,005 and 5,739,169; Hui and Hashimoto, 1998; Christodoulides et al., 1998), cytokine therapy e.g., interferons c, P and y; IL-1, GM-CSF 25 and TNF (Bukowski et al., 1998; Davidson et al., 1998; Hellstrand et al., 1998) gene therapy e.g., TNF, IL-1, IL-2, p 53 (Qin et al., 1998; Austin-Ward and Villaseca, 1998; U.S. Patents 5,830,880 and 5,846,945) and monoclonal antibodies e.g., anti-ganglioside GM2, anti-HER 2, anti-p185; Pietras et al., 1998; Hanibuchi et al., 1998; U.S. Patent 5,824,311). Herceptin (trastuzumab) is a chimeric (mouse-human) monoclonal antibody that blocks the HER2-neu 30 receptor. It possesses anti-tumor activity and has been approved for use in the treatment of malignant tumors (Dillman, 1999). Table 6 is a non-limiting list of several known anti - 104 - WO 2008/036718 PCT/US2007/078859 cancer immunotherapeutic agents and their targets. It is contemplated that one or more of these therapies may be employed with the miRNA therapies described herein. A number of different approaches for passive immunotherapy of cancer exist. They may be broadly categorized into the following: injection of antibodies alone; injection of 5 antibodies coupled to toxins or chemotherapeutic agents; injection of antibodies coupled to radioactive isotopes; injection of anti-idiotype antibodies; and finally, purging of tumor cells in bone marrow. TABLE 6 Generic Name Target Cetuximab EGFR Panitumumab EGFR Trastuzumab erbB2 receptor Bevacizumab VEGF Alemtuzumab CD52 Gemtuzumab ozogamicin CD33 Rituximab CD20 Tositumomab CD20 Matuzumab EGFR Ibritumomab tiuxetan CD20 Tositumomab CD20 HuPAM4 MUC1 MORAb-009 Mesothelin G250 carbonic anhydrase IX mAb 8H9 8H9 antigen M195 CD33 Ipilimumab CTLA4 HuLuc63 CS1 Alemtuzumab CD53 Epratuzumab CD22 BC8 CD45 HuJ591 Prostate specific membrane antigen hA20 CD20 Lexatumumab TRAIL receptor-2 Pertuzumab HER-2 receptor Mik-beta-1 IL-2R RAV12 RAAG12 SGN-30 CD30 AME-133v CD20 HeFi-I CD30 BMS-663513 CD137 Volociximab anti-a5p1 integrin GC1008 TGFp HCD122 CD40 Siplizumab CD2 MORAb-003 Folate receptor alpha CNTO 328 IL-6 MDX-060 CD30 Ofatumumab CD20 SGN-33 CD33 - 105- WO 2008/036718 PCT/US2007/078859 4. Gene Therapy In yet another embodiment, a combination treatment involves gene therapy in which a therapeutic polynucleotide is administered before, after, or at the same time as one or more therapeutic miRNA. Delivery of a therapeutic polypeptide or encoding nucleic acid in 5 conjunction with a miRNA may have a combined therapeutic effect on target tissues, A variety of proteins are encompassed within the invention, some of which are described below. Various genes that may be targeted for gene therapy of some form in combination with the present invention include, but are not limited to inducers of cellular proliferation, inhibitors of cellular proliferation, regulators of programmed cell death, cytokines and other therapeutic 10 nucleic acids or nucleic acid that encode therapeutic proteins. The tumor suppressor oncogenes function to inhibit excessive cellular proliferation. The inactivation of these genes destroys their inhibitory activity, resulting in unregulated proliferation. The tumor suppressors (e.g., therapeutic polypeptides) p53, FHIT, p16 and C CAM can be employed. 15 In addition to p53, another inhibitor of cellular proliferation is p16. The major transitions of the eukaryotic cell cycle are triggered by cyclin-dependent kinases, or CDK's. One CDK, cyclin-dependent kinase 4 (CDK4), regulates progression through the G1. The activity of this enzyme may be to phosphorylate Rb at late G1. The activity of CDK4 is controlled by an activating subunit, D-type cyclin, and by an inhibitory subunit, the p161NK4 20 has been biochemically characterized as a protein that specifically binds to and inhibits CDK4, and thus may regulate Rb phosphorylation (Serrano et al., 1993; Serrano et al., 1995). Since the p16INK4 protein is a CDK4 inhibitor (Serrano, 1993), deletion of this gene may increase the activity of CDK4, resulting in hyperphosphorylation of the Rb protein. p16 also is known to regulate the function of CDK6. 25 p161NK4 belongs to a newly described class of CDK-inhibitory proteins that also includes pl6B, p19, p21WAF1, and p27KIPI. The pl6INK4 gene maps to 9p2t, a chromosome region frequently deleted in many tumor types. Homozygous deletions and mutations of the p16INK4 gene are frequent in human tumor cell lines. This evidence suggests that the p161NK4 gene is a tumor suppressor gene. This interpretation has been 30 challenged, however, by the observation that the frequency of the p16INK4 gene alterations is much lower in primary uncultured tumors than in cultured cell lines (Caldas et al., 1994; Cheng et al., 1994; Hussussian et al., 1994; Kamb et al., 1994; Mori et al., 1994; Okamoto et - 106- WO 2008/036718 PCT/US2007/078859 al., 1994; Nobori et al., 1995; Orlow et al., 1994; Arap et al., 1995). Restoration of wild-type p1 61NK4 function by transfection with a plasmid expression vector reduced colony formation by some human cancer cell lines (Okamoto, 1994; Arap, 1995). Other genes that may be employed according to the present invention include Rb, 5 APC, DCC, NF-1, NF-2, WT-1, MEN-I, MEN-It, zacl, p73, VHL, MMAC1 / PTEN, DBCCR-1, FCC, rsk-3, p27, p27/pl6 fusions, p2l/p27 fusions, anti-thrombotic genes (e.g., COX-1, TFPI), PGS, Dp, E2F, ras, myc, neu, raf, erb, fns, trk, ret, gsp, hst, abl, ElA, p300, genes involved in angiogenesis (e.g., VEGF, FGF, thrombospondin, BAI-1, GDAIF, or their receptors) and MCC. 10 5. Surgery Approximately 60% of persons with cancer will undergo surgery of some type, which includes preventative, diagnostic or staging, curative and palliative surgery. Curative surgery is a cancer treatment that may be used in conjunction with other therapies, such as the treatment of the present invention, chemotherapy, radiotherapy, hormonal therapy, gene 15 therapy, immunotherapy and/or alternative therapies. Curative surgery includes resection in which all or part of cancerous tissue is physically removed, excised, and/or destroyed. Tumor resection refers to physical removal of at least part of a tumor. In addition to tumor resection, treatment by surgery includes laser surgery, cryosurgery, electrosurgery, and microscopically controlled surgery (Mohs' 20 surgery). It is further contemplated that the present invention may be used in conjunction with removal of superficial cancers, precancers, or incidental amounts of normal tissue. Upon excision of part of all of cancerous cells, tissue, or tumor, a cavity may be formed in the body. Treatment may be accomplished by perfusion, direct injection or local application of the area with an additional anti-cancer therapy. Such treatment may be 25 repeated, for example, every 1, 2, 3, 4, 5, 6, or 7 days, or every 1, 2, 3, 4, and 5 weeks or every 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 months. These treatments may be of varying dosages as well. 6. Other Agents It is contemplated that other agents may be used in combination with the present 30 invention to improve the therapeutic efficacy of treatment. These additional agents include -107- WO 2008/036718 PCT/US2007/078859 immunomodulatory agents, agents that affect the upregulation of cell surface receptors and GAP junctions, cytostatic and differentiation agents, inhibitors of cell adhesion, agents that increase the sensitivity of the hyperproliferative cells to apoptotic inducers, or other biological agents. Immunomodulatory agents include tumor necrosis factor; interferon alpha, 5 beta, and gamma; IL-2 and other cytokines; F42K and other cytokine analogs; or MIP-1, MIP-ibeta, MCP-1, RANTES, and other chemokines. It is further contemplated that the upregulation of cell surface receptors or their ligands such as Fas / Fas ligand, DR4 or DR5 / TRAIL (Apo-2 ligand) would potentiate the apoptotic inducing abilities of the present invention by establishment of an autocrine or paracrine effect on hyperproliferative cells. 10 Increases intercellular signaling by elevating the number of GAP junctions would increase the anti-hyperproliferative effects on the neighboring hyperproliferative cell population. In other embodiments, cytostatic or differentiation agents can be used in combination with the present invention to improve the anti-hyperproliferative efficacy of the treatments. Inhibitors of cell adhesion are contemplated to improve the efficacy of the present invention. Examples 15 of cell adhesion inhibitors are focal adhesion kinase (FAKs) inhibitors and Lovastatin. It is further contemplated that other agents that increase the sensitivity of a hyperproliferative cell to apoptosis, such as the antibody c225, could be used in combination with the present invention to improve the treatment efficacy. Apo2 ligand (Apo2L, also called TRAIL) is a member of the tumor necrosis factor 20 (TNF) cytokine family. TRAIL activates rapid apoptosis in many types of cancer cells, yet is not toxic to normal cells. TRAIL mRNA occurs in a wide variety of tissues. Most normal cells appear to be resistant to TRAIL's cytotoxic action, suggesting the existence of mechanisms that can protect against apoptosis induction by TRAIL. The first receptor described for TRAIL, called death receptor 4 (DR4), contains a cytoplasmic "death domain"; 25 DR4 transmits the apoptosis signal carried by TRAIL. Additional receptors have been identified that bind to TRAIL. One receptor, called DR5, contains a cytoplasmic death domain and signals apoptosis much like DR4. The DR4 and DR5 mRNAs are expressed in many normal tissues and tumor cell lines. Recently, decoy receptors such as DcR1 and DcR2 have been identified that prevent TRAIL from inducing apoptosis through DR4 and DR5. 30 These decoy receptors thus represent a novel mechanism for regulating sensitivity to a pro apoptotic cytokine directly at the cell's surface. The preferential expression of these inhibitory receptors in normal tissues suggests that TRAIL may be useful as an anticancer agent that induces apoptosis in cancer cells while sparing normal cells. (Marsters et al., 1999). -108- WO 2008/036718 PCT/US2007/078859 There have been many advances in the therapy of cancer following the introduction of cytotoxic chemotherapeutic drugs. However, one of the consequences of chemotherapy is the development/acquisition of drug-resistant phenotypes and the development of multiple drug resistance. The development of drug resistance remains a major obstacle in the treatment of 5 such tumors and therefore, there is an obvious need for alternative approaches such as gene therapy. Another form of therapy for use in conjunction with chemotherapy, radiation therapy or biological therapy includes hyperthermia, which is a procedure in which a patient's tissue is exposed to high temperatures (up to 106*F). External or internal heating devices may be 10 involved in the application of local, regional, or whole-body hyperthermia. Local hyperthermia involves the application of heat to a small area, such as a tumor. Heat may be generated externally with high-frequency waves targeting a tumor from a device outside the body. Internal heat may involve a sterile probe , including thin, heated wires or hollow tubes filled with warm water, implanted microwave antennae, or radiofrequency electrodes. 15 A patient's organ or a limb is heated for regional therapy, which is accomplished using devices that produce high energy, such as magnets. Alternatively, some of the patient's blood may be removed and heated before being perfused into an area that will be internally heated. Whole-body heating may also be implemented in cases where cancer has spread throughout the body. Warm-water blankets, hot wax, inductive coils, and thermal chambers 20 may be used for this purpose. Hormonal therapy may also be used in conjunction with the present invention or in combination with any other cancer therapy previously described. The use of hormones may be employed in the treatment of certain cancers such as breast, prostate, ovarian, or cervical cancer to lower the level or block the effects of certain hormones such as testosterone or 25 estrogen. This treatment is often used in combination with at least one other cancer therapy as a treatment option or to reduce the risk of metastases. This application incorporates U.S. Application Serial No. 11/349,727 filed on February 8, 2006 claiming priority to U.S. Provisional Application Serial No. 60/650,807 filed February 8, 2005 herein by references in its entirety. -109- WO 2008/036718 PCT/US2007/078859 III. MIRNA MOLECULES MicroRNA molecules ("miRNAs") are generally 21 to 22 nucleotides in length, though lengths of 19 and up to 23 nucleotides have been reported. The miRNAs are each processed from a longer precursor RNA molecule ("precursor miRNA"). Precursor miRNAs 5 are transcribed from non-protein-encoding genes. The precursor miRNAs have two regions of complementarity that enables them to form a stem-loop- or fold-back-like structure, which is cleaved in animals by a ribonuclease III-like nuclease enzyme called Dicer. The processed miRNA is typically a portion of the stem. The processed miRNA (also referred to as "mature miRNA") becomes part of a large 10 complex to down-regulate a particular target gene or its gene product. Examples of animal miRNAs include those that imperfectly basepair with the target, which halts translation (Olsen et al., 1999; Seggerson et al, 2002). siRNA molecules also are processed by Dicer, but from a long, double-stranded RNA molecule. siRNAs are not naturally found in animal cells, but they can direct the sequence-specific cleavage of an mRNA target through a RNA 15 induced silencing complex (RISC) (Denli et al, 2003). A. Array Preparation Certain embodiments of the present invention concerns the preparation and use of mRNA or nucleic acid arrays, miRNA or nucleic acid arrays, and/or miRNA or nucleic acid probe arrays, which are macroarrays or microarrays of nucleic acid molecules (probes) that 20 are fully or nearly complementary (over the length of the prove) or identical (over the length of the prove) to a plurality of nucleic acid, mRNA or miRNA molecules, precursor miRNA molecules, or nucleic acids derived from the various genes and gene pathways modulated by miR-143 miRNAs and that are positioned on a support or support material in a spatially separated organization. Macroarrays are typically sheets of nitrocellulose or nylon upon 25 which probes have been spotted. Microarrays position the nucleic acid probes more densely such that up to 10,000 nucleic acid molecules can be fit into a region typically 1 to 4 square centimeters. Microarrays can be fabricated by spotting nucleic acid molecules, e.g., genes, oligonucleotides, etc., onto substrates or fabricating oligonucleotide sequences in situ on a substrate. Spotted or fabricated nucleic acid molecules can be applied in a high density 30 matrix pattern of up to about 30 non-identical nucleic acid molecules per square centimeter or higher, e.g. up to about 100 or even 1000 per square centimeter. Microarrays typically use coated glass as the solid support, in contrast to the nitrocellulose-based material of filter -110- WO 2008/036718 PCT/US2007/078859 arrays. By having an ordered array of marker RNA and/or miRNA-complementing nucleic acid samples, the position of each sample can be tracked and linked to the original sample. A variety of different array devices in which a plurality of distinct nucleic acid probes are stably associated with the surface of a solid support are known to those of skill in the art. 5 Useful substrates for arrays include nylon, glass, metal, plastic, latex, and silicon. Such arrays may vary in a number of different ways, including average probe length, sequence or types of probes, nature of bond between the probe and the array surface, e.g. covalent or non covalent, and the like. The labeling and screening methods of the present invention and the arrays are not limited in its utility with respect to any parameter except that the probes detect 10 miRNA, or genes or nucleic acid representative of genes; consequently, methods and compositions may be used with a variety of different types of nucleic acid arrays. Representative methods and apparatus for preparing a microarray have been described, for example, in U.S. Patents 5,143,854; 5,202,231; 5,242,974; 5,288,644; 5,324,633; 5,384,261; 5,405,783; 5,412,087; 5,424,186; 5,429,807; 5,432,049; 5,436,327; 15 5,445,934; 5,468,613; 5,470,710; 5,472,672; 5,492,806; 5,525,464; 5,503,980; 5,510,270; 5,525,464; 5,527,681; 5,529,756; 5,532,128; 5,545,531; 5,547,839; 5,554,501; 5,556,752; 5,561,071; 5,571,639; 5,580,726; 5,580,732; 5,593,839; 5,599,695; 5,599,672; 5,610;287; 5,624,711; 5,631,134; 5,639,603; 5,654,413; 5,658,734; 5,661,028; 5,665,547; 5,667,972; 5,695,940; 5,700,637; 5,744,305; 5,800,992; 5,807,522; 5,830,645; 5,837,196; 5,871,928; 20 5,847,219; 5,876,932; 5,919,626; 6,004,755; 6,087,102; 6,368,799; 6,383,749; 6,617,112; 6,638,717; 6,720,138, as well as WO 93/17126; WO 95/11995; WO 95/21265; WO 95/21944; WO 95/35505; WO 96/31622; WO 97/10365; WO 97/27317; WO 99/35505; WO 09923256; WO 09936760; W00138580; WO 0168255; WO 03020898; WO 03040410; WO 03053586; WO 03087297; WO 03091426; W003100012; WO 04020085; WO 04027093; 25 EP 373 203; EP 785 280; EP 799 897 and UK 8 803 000; the disclosures of which are all herein incorporated by reference. It is contemplated that the arrays can be high density arrays, such that they contain 2, 20, 25, 50, 80, 100 or more different probes. It is contemplated that they may contain 1000, 16,000, 65,000, 250,000 or 1,000,000 or more different probes. The probes can be directed 30 to mRNA and/or miRNA targets in one or more different organisms or cell types. The oligonucleotide probes range from 5 to 50, 5 to 45, 10 to 40, 9 to 34, or 15 to 40 nucleotides in length in some embodiments. In certain embodiments, the oligonucleotide probes are 5, -111- WO 2008/036718 PCT/US2007/078859 10, 15, 20 to 20, 25, 30, 35, 40 nucleotides in length including all integers and ranges there between. The location and sequence of each different probe sequence in the array are generally known. Moreover, the large number of different probes can occupy a relatively small area 5 providing a high density array having a probe density of generally greater than about 60, 100, 600, 1000, 5,000, 10,000, 40,000, 100,000, or 400,000 different oligonucleotide probes per cm 2 . The surface area of the array can be about or less than about 1, 1.6, 2, 3, 4, 5, 6, 7, 8, 9, or 10 cm 2 . Moreover, a person of ordinary skill in the art could readily analyze data generated 10 using an array. Such protocols are disclosed above, and include information found in WO 9743450; WO 03023058; WO 03022421; WO 03029485; WO 03067217; WO 03066906; WO 03076928; WO 03093810; WO 03100448A1, all of which are specifically incorporated by reference. B. Sample Preparation 15 It is contemplated that the RNA and/or miRNA of a wide variety of samples can be analyzed using the arrays, index of probes, or array technology of the invention. While endogenous miRNA is contemplated for use with compositions and methods of the invention, recombinant miRNA - including nucleic acids that are complementary or identical to endogenous miRNA or precursor miRNA - can also be handled and analyzed as described 20 herein. Samples may be biological samples, in which case, they can be from biopsy, fine needle aspirates, exfoliates, blood, tissue, organs, semen, saliva, tears, other bodily fluid, hair follicles, skin, or any sample containing or constituting biological cells, particularly cancer or hyperproliferative cells. In certain embodiments, samples may be, but are not limited to, biopsy, or cells purified or enriched to some extent from a biopsy or other bodily fluids or 25 tissues. Alternatively, the sample may not be a biological sample, but be a chemical mixture, such as a cell-free reaction mixture (which may contain one or more biological enzymes). C. Hybridization After an array or a set of probes is prepared and/or the nucleic acid in the sample or probe is labeled, the population of target nucleic acids is contacted with the array or probes 30 under hybridization conditions, where such conditions can be adjusted, as desired, to provide for an optimum level of specificity in view of the particular assay being performed. Suitable -112- WO 2008/036718 PCT/US2007/078859 hybridization conditions are well known to those of skill in the art and reviewed in Sambrook et al. (2001) and WO 95/21944. Of particular interest in many embodiments is the use of stringent conditions during hybridization. Stringent conditions are known to those of skill in the art. 5 It is specifically contemplated that a single array or set of probes may be contacted with multiple samples. The samples may be labeled with different labels to distinguish the samples. For example, a single array can be contacted with a tumor tissue sample labeled with Cy3, and normal tissue sample labeled with Cy5. Differences between the samples for particular miRNAs corresponding to probes on the array can be readily ascertained and 10 quantified. The small surface area of the array permits uniform hybridization conditions, such as temperature regulation and salt content. Moreover, because of the small area occupied by the high density arrays, hybridization may be carried out in extremely small fluid volumes (e.g., about 250 p1 or less, including volumes of about or less than about 5, 10, 25, 50, 60, 70, 80, 15 90, 100 [d, or any range derivable therein). In small volumes, hybridization may proceed very rapidly. D. Differential Expression Analyses Arrays of the invention can be used to detect differences between two samples. Specifically contemplated applications include identifying and/or quantifying differences 20 between miRNA or gene expression from a sample that is normal and from a sample that is not normal, between a disease or condition and a cell not exhibiting such a disease or condition, or between two differently treated samples. Also, miRNA or gene expression may be compared between a sample believed to be susceptible to a particular disease or condition and one believed to be not susceptible or resistant to that disease or condition. A sample that 25 is not normal is one exhibiting phenotypic or genotypic trait(s) of a disease or condition, or one believed to be not normal with respect to that disease or condition. It may be compared to a cell that is normal with respect to that disease or condition. Phenotypic traits include symptoms of, or susceptibility to, a disease or condition of which a component is or may or may not be genetic, or caused by a hyperproliferative or neoplastic cell or cells. 30 An array comprises a solid support with nucleic acid probes attached to the support. Arrays typically comprise a plurality of different nucleic acid probes that are coupled to a - 113 - WO 2008/036718 PCT/US2007/078859 surface of a substrate in different, known locations. These arrays, also described as "microarrays" or colloquially "chips" have been generally described in the art, for example, U.S. Patents 5,143,854, 5,445,934, 5,744,305, 5,677,195, 6,040,193, 5,424,186 and Fodor et al., (1991), each of which is incorporated by reference in its entirety for all purposes. 5 Techniques for the synthesis of these arrays using mechanical synthesis methods are described in, e.g., U.S. Patent 5,384,261, incorporated herein by reference in its entirety for all purposes. Although a planar array surface is used in certain aspects, the array may be fabricated on a surface of virtually any shape or even a multiplicity of surfaces. Arrays may be nucleic acids on beads, gels, polymeric surfaces, fibers such as fiber optics, glass or any 10 other appropriate substrate, see U.S. Patents 5,770,358, 5,789,162, 5,708,153, 6,040,193 and 5,800,992, which are hereby incorporated in their entirety for all purposes. Arrays may be packaged in such a manner as to allow for diagnostics or other manipulation of an all inclusive device, see for example, U.S. Patents 5,856,174 and 5,922,591 incorporated in their entirety by reference for all purposes. See also U.S. patent application Ser. No. 09/545,207, 15 filed April 7, 2000 for additional information concerning arrays, their manufacture, and their characteristics, which is incorporated by reference in its entirety for all purposes. Particularly, arrays can be used to evaluate samples with respect to pathological condition such as cancer and related conditions. It is specifically contemplated that the invention can be used to evaluate differences between stages or sub-classifications of disease, 20 such as between benign, cancerous, and metastatic tissues or tumors. Phenotypic traits to be assessed include characteristics such as longevity, morbidity, expected survival, susceptibility or receptivity to particular drugs or therapeutic treatments (drug efficacy), and risk of drug toxicity. Samples that differ in these phenotypic traits may also be evaluated using the compositions and methods described. 25 In certain embodiments, miRNA and/or expression profiles may be generated to evaluate and correlate those profiles with pharmacokinetics or therapies. For example, these profiles may be created and evaluated for patient tumor and blood samples prior to the patient's being treated or during treatment to determine if there are miRNA or genes whose expression correlates with the outcome of the patient's treatment. Identification of 30 differential miRNAs or genes can lead to a diagnostic assay for evaluation of tumor and/or blood samples to determine what drug regimen the patient should be provided. In addition, it can be used to identify or select patients suitable for a particular clinical trial. If an -114- WO 2008/036718 PCT/US2007/078859 expression profile is determined to be correlated with drug efficacy or drug toxicity that profile is relevant to whether that patient is an appropriate patient for receiving a drug, for receiving a combination of drugs, or for a particular dosage of the drug. In addition to the above prognostic assay, samples from patients with a variety of 5 diseases can be evaluated to determine if different diseases can be identified based on miRNA and/or related gene expression levels. A diagnostic assay can be created based on the profiles that doctors can use to identify individuals with a disease or who are at risk to develop a disease. Alternatively, treatments can be designed based on miRNA profiling. Examples of such methods and compositions are described in the U.S. Provisional Patent 10 Application entitled "Methods and Compositions Involving miRNA and miRNA Inhibitor Molecules" filed on May 23, 2005, which is hereby incorporated by reference in its entirety. E. Other Assays In addition to the use of arrays and microarrays, it is contemplated that a number of different assays could be employed to analyze miRNAs or related genes, their activities, and 15 their effects. Such assays include, but are not limited to, nucleic acid amplification, polymerase chain reaction, quantitative PCR, RT-PCR, in situ hybridization, Northern hybridization, hybridization protection assay (HPA)(GenProbe), branched DNA (bDNA) assay (Chiron), rolling circle amplification (RCA), single molecule hybridization detection (US Genomics), Invader assay (ThirdWave Technologies), and/or Bridge Litigation Assay 20 (Genaco). IV. NUCLEIC ACIDS The present invention concerns nucleic acids, modified or mimetic nucleic acids, miRNAs, mRNAs, genes, and representative fragments thereof that can be labeled, used in array analysis, or employed in diagnostic, therapeutic, or prognostic applications, particularly 25 those related to pathological conditions such as cancer. The molecules may have been endogenously produced by a cell, or been synthesized or produced chemically or recombinantly. They may be isolated and/or purified. Each of the miRNAs described herein and include the corresponding SEQ ID NO and accession numbers for these miRNA sequences. The name of a miRNA is often abbreviated and referred to without a "hsa-" 30 prefix and will be understood as such, depending on the context. Unless otherwise indicated, -115- WO 2008/036718 PCT/US2007/078859 miRNAs referred to in the application are human sequences identified as miR-X or let-X, where X is a number and/or letter. In certain aspects, a miRNA probe designated by a suffix "SP" or "3P" can be used. "5P" indicates that the mature miRNA derives from the 5' end of the precursor and a 5 corresponding "3P" indicates that it derives from the 3' end of the precursor, as described on the world wide web at sanger.ac.uk. Moreover, in some embodiments, a miRNA probe is used that does not correspond to a known human miRNA. It is contemplated that these non human miRNA probes may be used in embodiments of the invention or that there may exist a human miRNA that is homologous to the non-human miRNA. In other embodiments, any 10 mammalian cell, biological sample, or preparation thereof may be employed. In some embodiments of the invention, methods and compositions involving miRNA may concern miRNA, markers (mRNAs), and/or other nucleic acids. Nucleic acids may be, be at least, or be at most 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47, 15 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250,-260, 270, 280, 290, 300, 310, 320, 330, 340, 350, 360, 370, 380, 390, 400, 410, 420, 430, 440, 450, 460, 470, 480, 490, 500, 510, 520, 530, 20 540, 550, 560, 570, 580, 590, 600, 610, 620, 630, 640, 650, 660, 670, 680, 690, 700, 710, 720, 730, 740, 750, 760, 770, 780, 790, 800, 810, 820, 830, 840, 850, 860, 870, 880, 890, 900, 910, 920, 930, 940, 950, 960, 970, 980, 990, or 1000 nucleotides, or any range derivable therein, in length. Such lengths cover the lengths of processed miRNA, miRNA probes, precursor miRNA, miRNA containing vectors, mRNA, mRNA probes, control nucleic acids, 25 and other probes and primers. In many embodiments, miRNA are 19-24 nucleotides in length, while miRNA probes are 19-35 nucleotides in length, depending on the length of the processed miRNA and any flanking regions added. miRNA precursors are generally between 62 and 110 nucleotides in humans. 30 Nucleic acids of the invention may have regions of identity or complementarity to another nucleic acid. It is contemplated that the region of complementarity or identity can be -116- WO 2008/036718 PCT/US2007/078859 at least 5 contiguous residues, though it is specifically contemplated that the region is, is at least, or is at most 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 5 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300, 310, 320, 330, 340, 350, 360, 370, 380, 390, 400, 410, 420, 430, 440, 441, 450, 460, 470, 480, 490, 500, 510, 520, 530, 540, 550, 560, 570, 580, 590, 600, 610, 620, 630, 640, 650, 660, 670, 680, 690, 700, 710, 720, 730, 740, 750, 760, 770, 780, 790, 800, 810, 10 820, 830, 840, 850, 860, 870, 880, 890, 900, 910, 920, 930, 940, 950, 960, 970, 980, 990, or 1000 contiguous nucleotides. It is further understood that the length of complementarity within a precursor miRNA or other nucleic acid or between a miRNA probe and a miRNA or a miRNA gene are such lengths. Moreover, the complementarity may be expressed as a percentage, meaning that the complementarity between a probe and its target is 90% or 15 greater over the length of the probe. In some embodiments, complementarity is or is at least 90%, 95% or 100%. In particular, such lengths may be applied to any nucleic acid comprising a nucleic acid sequence identified in any of SEQ ID NO:1-13, accession number, or any other sequence disclosed herein. Typically, the commonly used name of the miRNA is given (with its identifying source in the prefix, for example, "hsa" for human sequences) 20 and the processed miRNA sequence. Unless otherwise indicated, a miRNA without a prefix will be understood to refer to a human miRNA. Moreover, a lowercase letter in a miRNA name may or may not be lowercase; for example, hsa-mir-130b can also be referred to as miR-130B. The term "miRNA probe" refers to a nucleic acid probe that can identify a particular miRNA or structurally related miRNAs. 25 It is understood that some nucleic acids are derived from genomic sequences or a gene. In this respect, the term "gene" is used for simplicity to refer to the genomic sequence encoding the precursor nucleic acid or miRNA for a given miRNA or gene. However, embodiments of the invention may involve genomic sequences of a miRNA that are involved in its expression, such as a promoter or other regulatory sequences. 30 The term "recombinant" may be used and this generally refers to a molecule that has been manipulated in vitro or that is a replicated or expressed product of such a molecule. -117- WO 2008/036718 PCT/US2007/078859 The term "nucleic acid" is well known in the art. A "nucleic acid" as used herein will generally refer to a molecule (one or more strands) of DNA, RNA or a derivative or analog thereof, comprising a nucleobase. A nucleobase includes, for example, a naturally occurring purine or pyrimidine base found in DNA (e.g., an adenine "A," a guanine "G," a thymine "T" 5 or a cytosine "C") or RNA (e.g., an A, a G, an uracil "U" or a C). The term "nucleic acid" encompasses the terms "oligonucleotide" and "polynucleotide," each as a subgenus of the term "nucleic acid." The term "miRNA" generally refers to a single-stranded molecule, but in specific embodiments, molecules implemented in the invention will also encompass a region or an 10 additional strand that is partially (between 10 and 50% complementary across length of strand), substantially (greater than 50% but less than 100% complementary across length of strand) or fully complementary to another region of the same single-stranded molecule or to another nucleic acid. Thus, miRNA may encompass a molecule that comprises one or more complementary or self-complementary strand(s) or "complement(s)" of a particular sequence. 15 For example, precursor miRNA may have a self-complementary region, which is up to 100% complementary, miRNA probes or nucleic acids of the invention can include, can be or can be at least 60, 65, 70, 75, 80, 85, 90, 95, 96, 97, 98, 99 or 100% complementary to their target. It is understood that a "synthetic nucleic acid" of the invention means that the nucleic 20 acid does not have all or part of a chemical structure or sequence of a naturally occurring nucleic acid. Consequently, it will be understood that the term "synthetic miRNA" refers to a "synthetic nucleic acid" that functions in a cell or under physiological conditions as a naturally occurring miRNA. While embodiments of the invention may involve synthetic miRNAs or synthetic 25 nucleic acids, in some embodiments of the invention, the nucleic acid molecule(s) need not be "synthetic." In certain embodiments, a non-synthetic nucleic acid or miRNA employed in methods and compositions of the invention may have the entire sequence and structure of a naturally occurring mRNA or miRNA precursor or the mature mRNA or miRNA. For example, non-synthetic miRNAs used in methods and compositions of the invention may not 30 have one or more modified nucleotides or nucleotide analogs. In these embodiments, the non-synthetic miRNA may or may not be recombinantly produced. In particular embodiments, the nucleic acid in methods and/or compositions of the invention is specifically -118- WO 2008/036718 PCT/US2007/078859 a synthetic miRNA and not a non-synthetic miRNA (that is, not an miRNA that qualifies as "synthetic"); though in other embodiments, the invention specifically involves a non synthetic miRNA and not a synthetic miRNA. Any embodiments discussed with respect to the use of synthetic miRNAs can be applied with respect to non-synthetic miRNAs, and vice 5 versa. It will be understood that the term "naturally occurring" refers to something found in an organism without any intervention by a person; it could refer to a naturally-occurring wildtype or mutant molecule. In some embodiments a synthetic miRNA molecule does not have the sequence of a naturally occurring miRNA molecule. In other embodiments, a 10 synthetic miRNA molecule may have the sequence of a naturally occurring miRNA molecule, but the chemical structure of the molecule, particularly in the part unrelated specifically to the precise sequence (non-sequence chemical structure) differs from chemical structure of the naturally occurring miRNA molecule with that sequence. In some cases, the synthetic miRNA has both a sequence and non-sequence chemical structure that are not 15 found in a naturally-occurring miRNA. Moreover, the sequence of the synthetic molecules will identify which miRNA is effectively being provided or inhibited; the endogenous miRNA will be referred to as the "corresponding miRNA." Corresponding miRNA sequences that can be used in the context of the invention include, but are not limited to, all or a portion of those sequences in the SEQ IDs provided herein, as well as any other miRNA 20 sequence, miRNA precursor sequence, or any sequence complementary thereof In some embodiments, the sequence is or is derived from or contains all or part of a sequence identified herein to target a particular miRNA (or set of miRNAs) that can be used with that sequence. As used herein, "hybridization", "hybridizes" or "capable of hybridizing" is 25 understood to mean the forming of a double or triple stranded molecule or a molecule with partial double or triple stranded nature. The term "anneal" as used herein is synonymous with "hybridize." The term "hybridization", "hybridize(s)" or "capable of hybridizing" encompasses the terms "stringent condition(s)" or "high stringency" and the terms "low stringency" or "low stringency condition(s)." 30 As used herein "stringent condition(s)" or "high stringency" are those conditions that allow hybridization between or within one or more nucleic acid strand(s) containing complementary sequence(s), but preclude hybridization of random sequences. Stringent -119- WO 2008/036718 PCT/US2007/078859 conditions tolerate little, if any, mismatch between a nucleic acid and a target strand. Such conditions are well known to those of ordinary skill in the art, and are preferred for applications requiring high selectivity. Non-limiting applications include isolating a nucleic acid, such as a gene or a nucleic acid segment thereof, or detecting at least one specific 5 mRNA transcript or a nucleic acid segment thereof, and the like. Stringent conditions may comprise low salt and/or high temperature conditions, such as provided by about 0.02 M to about 0.5 M NaCl at temperatures of about 42 0 C to about 70'C. It is understood that the temperature and ionic strength of a desired stringency are determined in part by the length of the particular nucleic acid(s), the length and nucleobase 10 content of the target sequence(s), the charge composition of the nucleic acid(s), and to the presence or concentration of formamide, tetramethylammonium chloride or other solvent(s) in a hybridization mixture. It is also understood that these ranges, compositions and conditions for hybridization are mentioned by way of non-limiting examples only, and that the desired stringency for a 15 particular hybridization reaction is often determined empirically by comparison to one or more positive or negative controls. Depending on the application envisioned it is preferred to employ varying conditions of hybridization to achieve varying degrees of selectivity of a nucleic acid towards a target sequence. In a non-limiting example, identification of a related target nucleic acid that does not hybridize to a nucleic acid under stringent conditions may be 20 achieved by hybridization at low temperature and/or high ionic strength. Such conditions are termed "low stringency" or "low stringency conditions," and non-limiting examples of low stringency include hybridization performed at about 0.15 M to about 0.9 M NaCl at a temperature range of about 20*C to about 50oC. Of course, it is within the skill of one in the art to further modify the low or high stringency conditions to suite a particular application. 25 A. Nucleobase, Nucleoside, Nucleotide, and Modified Nucleotides As used herein a "nucleobase" refers to a heterocyclic base, such as for example a naturally occurring nucleobase (i.e., an A, T, G, C or U) found in at least one naturally occurring nucleic acid (i.e., DNA and RNA), and naturally or non-naturally occurring derivative(s) and analogs of such a nucleobase. A nucleobase generally can form one or 30 more hydrogen bonds ("anneal" or "hybridize") with at least one naturally occurring nucleobase in a manner that may substitute for naturally occurring nucleobase pairing (e.g., the hydrogen bonding between A and T, G and C, and A and U). - 120 - WO 2008/036718 PCT/US2007/078859 "Purine" and/or "pyrimidine" nucleobase(s) encompass naturally occurring purine and/or pyrimidine nucleobases and also derivative(s) and analog(s) thereof, including but not limited to, those a purine or pyrimidine substituted by one or more of an alkyl, caboxyalkyl, amino, hydroxyl, halogen (i.e., fluoro, chloro, bromo, or iodo), thiol or alkylthiol moiety. 5 Preferred alkyl (e.g., alkyl, caboxyalkyl, etc.) moieties comprise of from about 1, about 2, about 3, about 4, about 5, to about 6 carbon atoms. Other non-limiting examples of a purine or pyrimidine include a deazapurine, a 2,6-diaminopurine, a 5-fluorouracil, a xanthine, a hypoxanthine, a 8-bromoguanine, a 8-chloroguanine, a bromothymine, a 8-aminoguanine, a 8-hydroxyguanine, a 8-methylguanine, a 8-thioguanine, an azaguanine, a 2-aminopurine, a 5 10 ethylcytosine, a 5-methylcyosine, a 5-bromouracil, a 5-ethyluracil, a 5-iodouracil, a 5 chlorouracil, a 5-propyluracil, a thiouracil, a 2-methyladenine, a methylthioadenine, a N,N diemethyladenine, an azaadenines, a 8-bromoadenine, a 8-hydroxyadenine, a 6 hydroxyaminopurine, a 6-thiopurine, a 4-(6-aminohexyl/cytosine), and the like. Other examples are well known to those of skill in the art. 15 As used herein, a "nucleoside" refers to an individual chemical unit comprising a nucleobase covalently attached to a nucleobase linker moiety. A non-limiting example of a "nucleobase linker moiety" is a sugar comprising 5-carbon atoms (i.e., a "5-carbon sugar"), including but not limited to a deoxyribose, a ribose, an arabinose, or a derivative or an analog of a 5-carbon sugar. Non-limiting examples of a derivative or an analog of a 5-carbon sugar 20 include a 2'-fluoro-2'-deoxyribose or a carbocyclic sugar where a carbon is substituted for an oxygen atom in the sugar ring. Different types of covalent attachment(s) of a nucleobase to a nucleobase linker moiety are known in the art (Kornberg and Baker, 1992). As used herein, a "nucleotide" refers to a nucleoside further comprising a "backbone moiety". A backbone moiety generally covalently attaches a nucleotide to another molecule 25 comprising a nucleotide, or to another nucleotide to form a nucleic acid. The "backbone moiety" in naturally occurring nucleotides typically comprises a phosphorus moiety, which is covalently attached to a 5-carbon sugar. The attachment of the backbone moiety typically occurs at either the 3'- or 5'-position of the 5-carbon sugar. However, other types of attachments are known in the art, particularly when a nucleotide comprises derivatives or 30 analogs of a naturally occurring 5-carbon sugar or phosphorus moiety. A nucleic acid may comprise, or be composed entirely of, a derivative or analog of a nucleobase, a nucleobase linker moiety and/or backbone moiety that may be present in a - 121 - WO 2008/036718 PCT/US2007/078859 naturally occurring nucleic acid. RNA with nucleic acid analogs may also be labeled according to methods of the invention. As used herein a "derivative" refers to a chemically modified or altered form of a naturally occurring molecule, while the terms "mimic" or "analog" refer to a molecule that may or may not structurally resemble a naturally occurring 5 molecule or moiety, but possesses similar functions. As used herein, a "moiety" generally refers to a smaller chemical or molecular component of a larger chemical or molecular structure. Nucleobase, nucleoside and nucleotide analogs or derivatives are well known in the art, and have been described (see for example, Scheit, 1980, incorporated herein by reference). 10 Additional non-limiting examples of nucleosides, nucleotides or nucleic acids include those in: U.S. Patents 5,681,947, 5,652,099 and 5,763,167, 5,614,617, 5,670,663, 5,872,232, 5,859,221, 5,446,137, 5,886,165, 5,714,606, 5,672,697, 5,466,786, 5,792,847, 5,223,618, 5,470,967, 5,378,825, 5,777,092, 5,623,070, 5,610,289, 5,602,240, 5,858,988, 5,214,136, 5,700,922, 5,708,154, 5,728,525, 5,637,683, 6,251,666, 5,480,980, and 5,728,525, each of 15 which is incorporated herein by reference in its entirety. Labeling methods and kits of the invention specifically contemplate the use of nucleotides that are both modified for attachment of a label and can be incorporated into a miRNA molecule. Such nucleotides include those that can be labeled with a dye, including a fluorescent dye, or with a molecule such as biotin. Labeled nucleotides are readily available; 20 they can be acquired commercially or they can be synthesized by reactions known to those of skill in the art. Modified nucleotides for use in the invention are not naturally occurring nucleotides, but instead, refer to prepared nucleotides that have a reactive moiety on them. Specific reactive functionalities of interest include: amino, sulfhydryl, sulfoxyl, aminosulfhydryl, 25 azido, epoxide, isothiocyanate, isocyanate, anhydride, monochlorotriazine, dichlorotriazine, mono-or dihalogen substituted pyridine, mono- or disubstituted diazine, maleimide, epoxide, aziridine, sulfonyl halide, acid halide, alkyl halide, aryl halide, alkylsulfonate, N hydroxysuccinimide ester, imido ester, hydrazine, azidonitrophenyl, azide, 3-(2-pyridyl dithio)-propionamide, glyoxal, aldehyde, iodoacetyl, cyanomethyl ester, p-nitrophenyl ester, 30 o-nitrophenyl ester, hydroxypyridine ester, carbonyl imidazole, and the other such chemical groups. In some embodiments, the reactive functionality may be bonded directly to a nucleotide, or it may be bonded to the nucleotide through a linking group. The functional -122- WO 2008/036718 PCT/US2007/078859 moiety and any linker cannot substantially impair the ability of the nucleotide to be added to the miRNA or to be labeled. Representative linking groups include carbon containing linking groups, typically ranging from about 2 to 18, usually from about 2 to 8 carbon atoms, where the carbon containing linking groups may or may not include one or more heteroatoms, e.g. 5 S, 0, N etc., and may or may not include one or more sites of unsaturation. Of particular interest in many embodiments are alkyl linking groups, typically lower alkyl linking groups of 1 to 16, usually 1 to 4 carbon atoms, where the linking groups may include one or more sites of unsaturation. The functionalized nucleotides (or primers) used in the above methods of functionalized target generation may be fabricated using known protocols or purchased 10 from commercial vendors, e.g., Sigma, Roche, Ambion, Biosearch Technologies and NEN. Functional groups may be prepared according to ways known to those of skill in the art, including the representative information found in U.S. Patents 4,404,289; 4,405,711; 4,337,063 and 5,268,486, and U.K.. Patent 1,529,202, which are all incorporated by reference. 15 Amine-modified nucleotides are used in several embodiments of the invention. The amine-modified nucleotide is a nucleotide that has a reactive amine group for attachment of the label. It is contemplated that any ribonucleotide (G, A, U, or C) or deoxyribonucleotide (G, A, T, or C) can be modified for labeling. Examples include, but are not limited to, the following modified ribo- and deoxyribo-nucleotides: 5-(3-aminoallyl)-UTP; 8-[(4 20 amino)butyl]-amino-ATP and 8-[(6-amino)butyl]-amino-ATP; N6-(4-amino)butyl-ATP, N6 (6-amino)butyl-ATP, N4-[2,2-oxy-bis-(ethylamine)]-CTP; N6-(6-Amino)hexyl-ATP; 8-[(6 Amino)hexyl]-amino-ATP; 5-propargylamino-CTP, 5-propargylamino-UTP; 5-(3 aminoallyl)-dUTP; 8-[(4-amino)butyl]-amino-dATP and 8-[(6-amino)butyl]-amino-dATP; N6-(4-amino)butyl-dATP, N6-(6-amino)butyl-dATP, N4-[2,2-oxy-bis-(ethylamine)]-dCTP; 25 N6-(6-Amino)hexyl-dATP; 8-[(6-Amino)hexyl]-amino-dATP; 5-propargylamino-dCTP, and 5-propargylamino-dUTP. Such nucleotides can be prepared according to methods known to those of skill in the art. Moreover, a person of ordinary skill in the art could prepare other nucleotide entities with the same amine-modification, such as a 5-(3-aminoallyl)-CTP, GTP, ATP, dCTP, dGTP, dTTP, or dUTP in place of a 5-(3-aminoallyl)-UTP. 30 B. Preparation of Nucleic Acids A nucleic acid may be made by any technique known to one of ordinary skill in the art, such as for example, chemical synthesis, enzymatic production, or biological production. - 123- WO 2008/036718 PCT/US2007/078859 It is specifically contemplated that miRNA probes of the invention are chemically synthesized. In some embodiments of the invention, miRNAs are recovered or isolated from a biological sample. The miRNA may be recombinant or it may be natural or endogenous to 5 the cell (produced from the cell's genome). It is contemplated that a biological sample may be treated in a way so as to enhance the recovery of small RNA molecules such as miRNA. U.S. Patent Application Serial No. 10/667,126 describes such methods and it is specifically incorporated by reference herein. Generally, methods involve lysing cells with a solution having guanidinium and a detergent. 10 Alternatively, nucleic acid synthesis is performed according to standard methods. See, for example, Itakura and Riggs (1980) and U.S. Patents 4,704,362, 5,221,619, and 5,583,013, each of which is incorporated herein by reference. Non-limiting examples of a synthetic nucleic acid (e.g., a synthetic oligonucleotide), include a nucleic acid made by in vitro chemically synthesis using phosphotriester, phosphite, or phosphoramidite chemistry 15 and solid phase techniques such as described in EP 266,032, incorporated herein by reference, or via deoxynucleoside H-phosphonate intermediates as described by Froehler et al, 1986 and U.S. Patent 5,705,629, each incorporated herein by reference. Various different mechanisms of oligonucleotide synthesis have been disclosed in for example, U.S. Patents 4,659,774, 4,816,571, 5,141,813, 5,264,566, 4,959,463, 5,428,148, 5,554,744, 5,574,146, 20 5,602,244, each of which is incorporated herein by reference. A non-limiting example of an enzymatically produced nucleic acid include one produced by enzymes in amplification reactions such as PCRTM (see for example, U.S. Patents 4,683,202 and 4,682,195, each incorporated herein by reference), or the synthesis of an oligonucleotide described in U.S. Patent 5,645,897, incorporated herein by reference. See 25 also Sambrook et al, 2001, incorporated herein by reference). Oligonucleotide synthesis is well known to those of skill in the art. Various different mechanisms of oligonucleotide synthesis have been disclosed in for example, U.S. Patents 4,659,774, 4,816,571, 5,141,813, 5,264,566, 4,959,463, 5,428,148, 5,554,744, 5,574,146, 5,602,244, each of which is incorporated herein by reference. 30 Recombinant methods for producing nucleic acids in a cell are well known to those of skill in the art. These include the use of vectors (viral and non-viral), plasmids, cosmids, and - 124- WO 2008/036718 PCT/US2007/078859 other vehicles for delivering a nucleic acid to a cell, which may be the target cell (e.g., a cancer cell) or simply a host cell (to produce large quantities of the desired RNA molecule). Alternatively, such vehicles can be used in the context of a cell free system so long as the reagents for generating the RNA molecule are present. Such methods include those described 5 in Sambrook, 2003, Sambrook, 2001 and Sambrook, 1989, which are hereby incorporated by reference. C. Isolation of Nucleic Acids Nucleic acids may be isolated using techniques well known to those of skill in the art, though in particular embodiments, methods for isolating small nucleic acid molecules, and/or 10 isolating RNA molecules can be employed. Chromatography is a process often used to separate or isolate nucleic acids from protein or from other nucleic acids. Such methods can involve electrophoresis with a gel matrix, filter columns, alcohol precipitation, and/or other chromatography. If miRNA from cells is to be used or evaluated, methods generally involve lysing the cells with a chaotropic (e.g., guanidinium isothiocyanate) and/or detergent (e.g., N 15 lauroyl sarcosine) prior to implementing processes for isolating particular populations of RNA. In particular methods for separating miRNA from other nucleic acids, a gel matrix is prepared using polyacrylamide, though agarose can also be used. The gels may be graded by concentration or they may be uniform. Plates or tubing can be used to hold the gel matrix for 20 electrophoresis. Usually one-dimensional electrophoresis is employed for the separation of nucleic acids. Plates are used to prepare a slab gel, while the tubing (glass or rubber, typically) can be used to prepare a tube gel. The phrase "tube electrophoresis" refers to the use of a tube or tubing, instead of plates, to form the gel. Materials for implementing tube electrophoresis can be readily prepared by a person of skill in the art or purchased, such as 25 from C.B.S. Scientific Co., Inc. or Scie-Plas. Methods may involve the use of organic solvents and/or alcohol to isolate nucleic acids, particularly miRNA used in methods and compositions of the invention. Some embodiments are described in U.S. Patent Application Serial No. 10/667,126, which is hereby incorporated by reference. Generally, this disclosure provides methods for efficiently 30 isolating small RNA molecules from cells comprising: adding an alcohol solution to a cell lysate and applying the alcohol/lysate mixture to a solid support before eluting the RNA molecules from the solid support. In some embodiments, the amount of alcohol added to a - 125 - WO 2008/036718 PCT/US2007/078859 cell lysate achieves an alcohol concentration of about 55% to 60%. While different alcohols can be employed, ethanol works well. A solid support may be any structure, and it includes beads, filters, and columns, which may include a mineral or polymer support with electronegative groups. A glass fiber filter or column has worked particularly well for such 5 isolation procedures. In specific embodiments, miRNA isolation processes include: a) lysing cells in the sample with a lysing solution comprising guanidinium, wherein a lysate with a concentration of at least about I M guanidinium is produced; b) extracting miRNA molecules from the lysate with an extraction solution comprising phenol; c) adding to the lysate an alcohol 10 solution for forming a lysate/alcohol mixture, wherein the concentration of alcohol in the mixture is between about 35% to about 70%; d) applying the lysate/alcohol mixture to a solid support; e) eluting the miRNA molecules from the solid support with an ionic solution; and, f) capturing the miRNA molecules. Typically the sample is dried and resuspended in a liquid and volume appropriate for subsequent manipulation. 15 V. LABELS AND LABELING TECHNIQUES In some embodiments, the present invention concerns miRNA that are labeled. It is contemplated that miRNA may first be isolated and/or purified prior to labeling. This may achieve a reaction that more efficiently labels the miRNA, as opposed to other RNA in a sample in which the miRNA is not isolated or purified prior to labeling. In many 20 embodiments of the invention, the label is non-radioactive. Generally, nucleic acids may be labeled by adding labeled nucleotides (one-step process) or adding nucleotides and labeling the added nucleotides (two-step process). A. Labeling Techniques In some embodiments, nucleic acids are labeled by catalytically adding to the nucleic 25 acid an already labeled nucleotide or nucleotides. One or more labeled nucleotides can be added to miRNA molecules. See U.S. Patent 6,723,509, which is hereby incorporated by reference. In other embodiments, an unlabeled nucleotide or nucleotides is catalytically added to a miRNA, and the unlabeled nucleotide is modified with a chemical moiety that enables it to 30 be subsequently labeled. In embodiments of the invention, the chemical moiety is a reactive amine such that the nucleotide is an amine-modified nucleotide. Examples of amine -126 - WO 2008/036718 PCT/US2007/078859 modified nucleotides are well known to those of skill in the art, many being commercially available such as from Ambion, Sigma, Jena Bioscience, and TriLink. In contrast to labeling of cDNA during its synthesis, the issue for labeling miRNA is how to label the already existing molecule. The present invention concerns the use of an 5 enzyme capable of using a di- or tri-phosphate ribonucleotide or deoxyribonucleotide as a substrate for its addition to a miRNA. Moreover, in specific embodiments, it involves using a modified di- or tri-phosphate ribonucleotide, which is added to the 3' end of a miRNA. Enzymes capable of adding such nucleotides include, but are not limited to, poly(A) polymerase, terminal transferase, and polynucleotide phosphorylase. In specific 10 embodiments of the invention, a ligase is contemplated as not being the enzyme used to add the label, and instead, a non-ligase enzyme is employed. Terminal transferase catalyzes the addition of nucleotides to the 3' terminus of a nucleic acid. Polynucleotide phosphorylase can polymerize nucleotide diphosphates without the need for a primer. B. Labels 15 Labels on miRNA or miRNA probes may be colorimetric (includes visible and UV spectrum, including fluorescent), luminescent, enzymatic, or positron emitting (including radioactive). The label may be detected directly or indirectly. Radioactive labels include 125, , 3 P, and 3IS. Examples of enzymatic labels include alkaline phosphatase, luciferase, horseradish peroxidase, and p-galactosidase. Labels can also be proteins with luminescent 20 properties, e.g., green fluorescent protein and phicoerythrin. The colorimetric and fluorescent labels contemplated for use as conjugates include, but are not limited to, Alexa Fluor dyes, BODIPY dyes, such as BODIPY FL; Cascade Blue; Cascade Yellow; coumarin and its derivatives, such as 7-amino-4-methylcoumarin, aminocoumarin and hydroxycoumarin; cyanine dyes, such as Cy3 and Cy5; eosins and 25 erythrosins; fluorescein and its derivatives, such as fluorescein isothiocyanate; macrocyclic chelates of lanthanide ions, such as Quantum DyeTM; Marina Blue; Oregon Green; rhodamine dyes, such as rhodamine red, tetramethylrhodamine and rhodamine 6G; Texas Red; , fluorescent energy transfer dyes, such as thiazole orange-ethidium heterodimer; and, TOTAB. 30 Specific examples of dyes include, but are not limited to, those identified above and the following: Alexa Fluor 350, Alexa Fluor 405, Alexa Fluor 430, Alexa Fluor 488, Alexa - 127- WO 2008/036718 PCT/US2007/078859 Fluor 500. Alexa Fluor 514, Alexa Fluor 532, Alexa Fluor 546, Alexa Fluor 555, Alexa Fluor 568, Alexa Fluor 594, Alexa Fluor 610, Alexa Fluor 633, Alexa Fluor 647, Alexa Fluor 660, Alexa Fluor 680, Alexa Fluor 700, and, Alexa Fluor 750; amine-reactive BODIPY dyes, such as BODIPY 493/503, BODIPY 530/550, BODIPY 558/568, BODIPY 564/570, BODIPY 5 576/589, BODIPY 581/591, BODIPY 630/650, BODIPY 650/655, BODIPY FL, BODIPY R6G, BODIPY TMR, and, BODIPY-TR; Cy3, Cy5, 6-FAM, Fluorescein Isothiocyanate, HEX, 6-JOE, Oregon Green 488, Oregon Green 500, Oregon Green 514, Pacific Blue, REG, Rhodamine Green, Rhodamine Red, Renographin, ROX, SYPRO, TAMRA, 2',4',5',7' Tetrabromosulfonefluorescein, and TET. 10 Specific examples of fluorescently labeled ribonucleotides are available from Molecular Probes, and these include, Alexa Fluor 488-5-UTP, Fluorescein-12-UTP, BODIPY FL-14-UTP, BODIPY TMR-14-UTP, Tetramethylrhodamine-6-UTP, Alexa Fluor 546-14 UTP, Texas Red-5-UTP, and BODIPY TR-14-UTP. Other fluorescent ribonucleotides are available from Amersham Biosciences, such as Cy3-UTP and Cy5-UTP. 15 Examples of fluorescently labeled deoxyribonucleotides include Dinitrophenyl (DNP)-ll-dUTP, Cascade Blue-7-dUTP, Alexa Fluor 488-5-dUTP, Fluorescein-12-dUTP, Oregon Green 488-5-dUTP, BODIPY FL-14-dUTP, Rhodamine Green-5-dUTP, Alexa Fluor 532-5-dUTP, BODIPY TMR-14-dUTP, Tetramethylrhodamine-6-dUTP, Alexa Fluor 546 14-dUTP, Alexa Fluor 568-5-dUTP, Texas Red-l 2-dUTP, Texas Red-5-dUTP, BODIPY TR 20 14-dUTP, Alexa Fluor 594-5-dUTP, BODIPY 630/650-14-dUTP, BODIPY 650/665-14 dUTP; Alexa Fluor 488-7-OBEA-dCTP, Alexa Fluor 546-16-OBEA-dCTP, Alexa Fluor 594 7-OBEA-dCTP, Alexa Fluor 647-12-OBEA-dCTP. It is contemplated that nucleic acids may be labeled with two different labels. Furthermore, fluorescence resonance energy transfer (FRET) may be employed in methods of 25 the invention (e.g., Klostermeier et al., 2002; Emptage, 2001; Didenko, 2001, each incorporated by reference). Alternatively, the label may not be detectable per se, but indirectly detectable or allowing for the isolation or separation of the targeted nucleic acid. For example, the label could be biotin, digoxigenin, polyvalent cations, chelator groups and the other ligands, 30 include ligands for an antibody. - 128- WO 2008/036718 PCT/US2007/078859 C. Visualization Techniques A number of techniques for visualizing or detecting labeled nucleic acids are readily available. Such techniques include, microscopy, arrays, Fluorometry, Light cyclers or other real time PCR machines, FACS analysis, scintillation counters, Phosphoimagers, Geiger 5 counters, MRI, CAT, antibody-based detection methods (Westerns, immunofluorescence, immunohistochemistry), histochemical techniques, HPLC (Griffey et al, 1997), spectroscopy, capillary gel electrophoresis (Cummins et al, 1996), spectroscopy; mass spectroscopy; radiological techniques; and mass balance techniques. When two or more differentially colored labels are employed, fluorescent resonance 10 energy transfer (FRET) techniques may be employed to characterize association of one or more nucleic acid. Furthermore, a person of ordinary skill in the art is well aware of ways of visualizing, identifying, and characterizing labeled nucleic acids, and accordingly, such protocols may be used as part of the invention. Examples of tools that may be used also include fluorescent microscopy, a BioAnalyzer, a plate reader, Storm (Molecular Dynamics), 15 Array Scanner, FACS (fluorescent activated cell sorter), or any instrument that has the ability to excite and detect a fluorescent molecule. V. KITS Any of the compositions described herein may be comprised in a kit. In a non limiting example, reagents for isolating miRNA, labeling miRNA, and/or evaluating a 20 miRNA population using an array, nucleic acid amplification, and/or hybridization can be included in a kit, as well reagents for preparation of samples from blood samples. The kit may further include reagents for creating or synthesizing miRNA probes. The kits will thus comprise, in suitable container means, an enzyme for labeling the miRNA by incorporating labeled nucleotide or unlabeled nucleotides that are subsequently labeled. In certain aspects, 25 the kit can include amplification reagents. In other aspects, the kit may include various supports, such as glass, nylon, polymeric beads, and the like, and/or reagents for coupling any probes and/or target nucleic acids. It may also include one or more buffers, such as reaction buffer, labeling buffer, washing buffer, or a hybridization buffer, compounds for preparing the miRNA probes, and components for isolating miRNA. Other kits of the invention may 30 include components for making a nucleic acid array comprising miRNA, and thus, may include, for example, a solid support. -129- WO 2008/036718 PCT/US2007/078859 Kits for implementing methods of the invention described herein are specifically contemplated. In some embodiments, there are kits for preparing miRNA for multi-labeling and kits for preparing miRNA probes and/or miRNA arrays. In these embodiments, kit comprise, in suitable container means, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 or more of the 5 following: (1) poly(A) polymerase; (2) unmodified nucleotides (G, A, T, C, and/or U); (3) a modified nucleotide (labeled or unlabeled); (4) poly(A) polymerase buffer; and, (5) at least one microfilter; (6) label that can be attached to a nucleotide; (7) at least one miRNA probe; (8) reaction buffer; (9) a miRNA array or components for making such an array; (10) acetic acid; (11) alcohol; (12) solutions for preparing, isolating, enriching, and purifying miRNAs 10 or miRNA probes or arrays. Other reagents include those generally used for manipulating RNA, such as formamide, loading dye, ribonuclease inhibitors, and DNase. In specific embodiments, kits of the invention include an array containing miRNA probes, as described in the application. An array may have probes corresponding to all known miRNAs of an organism or a particular tissue or organ in particular conditions, or to a 15 subset of such probes. The subset of probes on arrays of the invention may be or include those identified as relevant to a particular diagnostic, therapeutic, or prognostic application. For example, the array may contain one or more probes that is indicative or suggestive of (1) a disease or condition (acute myeloid leukemia), (2) susceptibility or resistance to a particular drug or treatment; (3) susceptibility to toxicity from a drug or substance; (4) the stage of 20 development or severity of a disease or condition (prognosis); and (5) genetic predisposition to a disease or condition. For any kit embodiment, including an array, there can be nucleic acid molecules that contain or can be used to amplify a sequence that is a variant of, identical to or complementary to all or part of any of SEQ IDs described herein. In certain embodiments, a 25 kit or array of the invention can contain one or more probes for the miRNAs identified by the SEQ IDs described herein. Any nucleic acid discussed above may be implemented as part of a kit. The components of the kits may be packaged either in aqueous media or in lyophilized form. The container means of the kits will generally include at least one vial, test 30 tube, flask, bottle, syringe or other container means, into which a component may be placed, and preferably, suitably aliquoted. Where there is more than one component in the kit (labeling reagent and label may be packaged together), the kit also will generally contain a -130- WO 2008/036718 PCT/US2007/078859 second, third or other additional container into which the additional components may be separately placed. However, various combinations of components may be comprised in a vial. The kits of the present invention also will typically include a means for containing the nucleic acids, and any other reagent containers in close confinement for commercial sale. 5 Such containers may include injection or blow molded plastic containers into which the desired vials are retained. When the components of the kit are provided in one and/or more liquid solutions, the liquid solution is an aqueous solution, with a sterile aqueous solution being particularly preferred. 10 However, the components of the kit may be provided as dried powder(s). When reagents and/or components are provided as a dry powder, the powder can be reconstituted by the addition of a suitable solvent. It is envisioned that the solvent may also be provided in another container means. In some embodiments, labeling dyes are provided as a dried power. It is contemplated that 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 120, 120, 130, 140, 150, 160, 15 170, 180, 190, 200, 300, 400, 500, 600, 700, 800, 900, 1000 pg or at least or at most those amounts of dried dye are provided in kits of the invention. The dye may then be resuspended in any suitable solvent, such as DMSO. Such kits may also include components that facilitate isolation of the labeled miRNA. It may also include components that preserve or maintain the miRNA or that protect against 20 its degradation. Such components may be RNAse-free or protect against RNAses. Such kits generally will comprise, in suitable means, distinct containers for each individual reagent or solution. A kit will also include instructions for employing the kit components as well the use of any other reagent not included in the kit. Instructions may include variations that can be 25 implemented. Kits of the invention may also include one or more of the following: Control RNA; nuclease-free water; RNase-free containers, such as 1.5 ml tubes; RNase-free elution tubes; PEG or dextran; ethanol; acetic acid; sodium acetate; ammonium acetate; guanidinium; detergent; nucleic acid size marker; RNase-free tube tips; and RNase or DNase inhibitors. -131- WO 2008/036718 PCT/US2007/078859 It is contemplated that such reagents are embodiments of kits of the invention. Such kits, however, are not limited to the particular items identified above and may include any reagent used for the manipulation or characterization of miRNA. VII. EXAMPLES 5 The following examples are included to demonstrate preferred embodiments of the invention. It should be appreciated by those of skill in the art that the techniques disclosed in the examples which follow represent techniques discovered by the inventor to function well in the practice of the invention, and thus can be considered to constitute preferred modes for its practice. However, those of skill in the art should, in light of the present disclosure, 10 appreciate that many changes can be made in the specific embodiments which are disclosed and still obtain a like or similar result without departing from the spirit and scope of the invention. EXAMPLE 1: 15 GENE EXPRESSION ANALYSIS FOLLOWING TRANSFECTION WITH HSA-MIR-143 miRNAs are believed to regulate gene expression by binding to target mRNA transcripts and (1) initiating transcript degradation or (2) altering protein translation from the transcript. Translational regulation leading to an up or down change in protein expression 20 may lead to changes in activity and expression of downstream gene products and genes that are in turn regulated by those proteins. These numerous regulatory effects may be revealed as changes in the global mRNA expression profile. Microarray gene expression analyses were performed to identify genes that are mis-regulated by hsa-miR- 143 expression. Synthetic Pre-miR-143 (Ambion) or two negative control miRNAs (pre-miR-NCI, 25 Ambion cat. no. AM17110 and pre-miR-NC2, Ambion, cat. no. AM17111) were reverse transfected into quadruplicate samples of A549 cells for each of three time points. Cells were transfected using siPORT NeoFX (Ambion) according to the manufacturer's recommendations using the following parameters: 200,000 cells per well in a 6 well plate, 5.0 pi of NeoFX, 30 nM final concentration of miRNA in 2.5 ml. Cells were harvested at 4 h, 24 30 h, and 72 h post transfection. Total RNA was extracted using RNAqueous-4PCR (Ambion) according to the manufacturer's recommended protocol. - 132- WO 2008/036718 PCT/US2007/078859 mRNA array analyses were performed by Asuragen Services (Austin, TX), according to the company's standard operating procedures. Using the MessageAmp TM 11-96 aRNA Amplification Kit (Ambion, cat #1819) 2 pg of total RNA were used for target preparation and labeling with biotin. cRNA yields were quantified using an Agilent Bioanalyzer 2100 5 capillary electrophoresis protocol. Labeled target was hybridized to Affymetrix mRNA arrays (Human HG-Ul33A 2.0 arrays) using the manufacturer's recommendations and the following parameters. Hybridizations were carried out at 45"C for 16 hr in an Affymetrix Model 640 hybridization oven. Arrays were washed and stained on an Affymetrix FS450 Fluidics station, running the wash script Midi euk2v3_450. The arrays were scanned on a 10 Affymetrix GeneChip Scanner 3000. Summaries of the image signal data, group mean values, p-values with significance flags, log ratios and gene annotations for every gene on the array were generated using the Affymetrix Statistical Algorithm MAS 5.0 (GCOS vl.3). Data were reported in a file (cabinet) containing the Affymetrix data and result files and in files (.eel) containing the primary image and processed cell intensities of the arrays. Data 15 were normalized for the effect observed by the average of two negative control microRNA sequences and then were averaged together for presentation. A list of genes whose expression levels varied by at least 0.7 log2 from the average negative control was assembled. Results of the microarray gene expression analysis are shown in Table 1 above. Manipulation of the expression levels of the genes listed in Table I represents a 20 potentially useful therapy for cancer and other diseases in which increased or reduced expression of hsa-miR-143 has a role in the disease. EXAMPLE 2: CELLULAR PATHWAYS AFFECTED BY HSA-miR-143 25 The mis-regulation of gene expression by hsa-miR-143 (Table 1) affects many cellular pathways that represent potential therapeutic targets for the control of cancer and other diseases and disorders. The inventors determined the identity and nature of the cellular genetic pathways affected by the regulatory cascade induced by hsa-miR-143 expression. Cellular pathway analyses were performed using Ingenuity Pathways Analysis (Version 4.0, 30 Ingenuity* Systems, Redwood City, CA). Alteration of a given pathway was determined by Fisher's Exact test (Fisher, 1922). The most significantly affected pathways following over expression of hsa-miR-143 in A549 cells are shown in Table 2. -133 - WO 2008/036718 PCT/US2007/078859 These data demonstrate that hsa-miR-143 directly or indirectly affects the expression of several, cellular proliferation-, development-, and cell growth-related genes and thus primarily affects functional pathways related to cellular growth, cellular development, and cell proliferation. Those cellular processes have integral roles in the development and 5 progression of various cancers. Manipulation of the expression levels of genes in the cellular pathways shown in Table 2 represents a potentially useful therapy for cancer and other diseases in which increased or reduced expression of hsa-miR-143 has a role in the disease. EXAMPLE 3: 10 PREDICTED GENE TARGETS OF HSA-MIR-143 Gene targets for binding of and regulation by hsa-miR-143 were predicted using the proprietary algorithm miRNATargetTM (Asuragen), which is an implementation of the method proposed by Krek et al. (2005). Predicted target genes are shown in Table 3. The predicted gene targets that exhibited altered mRNA expression levels in human 15 cancer cells, following transfection with pre-miR hsa-miR-143, are shown in Table 4. The predicted gene targets of hsa-miR-143 whose mRNA expression levels are affected by hsa-miR-143 represent particularly useful candidates for cancer therapy and therapy of other diseases through manipulation of their expression levels. 20 EXAMPLE 4: CANCER RELATED GENE EXPRESSION ALTERED BY HSA-MIR-143 Cell proliferation, survival, and growth pathways are commonly altered in tumors (Hanahan and Weinberg, 2000). The inventors have shown that hsa-miR-143 directly or indirectly regulates the transcripts of proteins that are critical in the regulation of these 25 pathways. Many of these targets have inherent oncogenic or tumor suppressor activity. Hsa miR-143 targets that have prognostic and/or therapeutic value for the treatment of various malignancies are shown in Table 5. Hsa-miR-143 targeted cancer genes are regulators of the cell cycle, transcription, intracellular signaling, apoptosis and the thioredoxin redox pathway. Hsa-miR-143 regulates 30 cell cycle progression by altering the expression of Weel, the retinoblastoma-like 1 protein (RBLl) as well as the cyclins Dl and GI. RBLI, also known as p107, is a member of the - 134- WO 2008/036718 PCT/US2007/078859 retinoblastoma tumor suppressor protein family that includes the pocket proteins p107, p130 and pRb. Similar to the pRb prototype, RBL1 interacts with the E2F family of transcription factors and blocks cell cycle progression and DNA replication (Sherr and McCormick, 2002). A subset of cancers show deregulated expression of RBL1 (Takimoto et al., 1998; Claudio et 5 al., 2002; Wu et al., 2002; Ito et al., 2003). Transient transfection of hsa-miR-143 leads to a decrease in RBL1 mRNA levels which may suggest a proliferative function for hsa-miR-143. In contrast, negative regulation of cyclin D1 and positive regulation of cyclin G1 are indicators of a growth-inhibitory role for hsa-miR-143. Cyclins are co-factors of cyclin dependent kinases (CDKs) and function in the progression of the cell cycle. Cyclin Dl is 10 required for the transition from G1 into S phase and is overexpressed in numerous cancer types (Donnellan and Chetty, 1998).(Donnellan and Chetty, 1998). Hsa-miR-143 negatively regulates cyclin Dl expression and therefore might interfere with abnormal cell growth that depends on high levels of cyclin Dl. In accordance, cyclin G1 has growth inhibitory activity and is upregulated by hsa-miR-143 (Zhao et al., 2003). Weel is a tyrosine kinase that 15 functions as a mitotic inhibitor by phosphorylating the CDKl(cdc2)/cyclinB1 complex (Parker and Piwnica-Worms, 1992; McGowan and Russell, 1993). Lack of Weel expression in lung cancer is correlated with a higher proliferation index, a higher relapse rate and poor prognosis (Yoshida et al., 2004). Another hsa-miR-143 target is LMO-4 (LIM domain only 4), a zinc finger protein regulating transcription. LMO-4 is inherently oncogenic and 20 inactivates the BRCA-1 tumor suppressor protein (breast cancer 1) (Sum et al., 2002; Sum et al., 2005). LMO-4 is frequently overexpressed in multiple cancer types and predicts poor outcome in breast cancer (Visvader et al., 2001; Mizunuma et al., 2003; Sum et al., 2005; Taniwaki et al., 2006). Accordingly, RNAi directed against LMO-4 leads to reduced breast cancer cell growth and migration (Sum et al., 2005). Our data indicate that hsa-miR-143 25 diminishes LMO-4 transcripts and therefore may intercept with the oncogenic properties of LMO-4. Hsa-miR-143 also governs the expression of PDCD4, BCL2L1 and MCL1, all of which are functionally linked to the apoptotic pathway. Pdcd-4 (programmed cell death 4) is a tumor suppressor that is induced in response to apoptosis in normal cells. The growth 30 inhibitory properties of Pdcd-4 are due to Pdcd-4 mediated inhibition of the c-Jun proto oncoprotein, inhibition of cap-dependent mRNA translation and activation of the p2lWafl/Cipl CDK inhibitor (Yang et al., 2003; Bitomsky et al., 2004; Goke et al., 2004). Pdcd-4 frequently shows reduced or lost expression in various human malignancies, such as - 135- WO 2008/036718 PCT/US2007/078859 gliomas, hepatocellular carcinomas, lung and renal cell carcinomas (Jansen et al., 2004; Zhang et al., 2006; Gao et al., 2007). Expression of Pdcd-4 interferes with skin carcinogenesis in a mouse model and suppresses growth of human colon carcinoma cells (Jansen et al., 2005; Yang et al., 2006). Loss of Pdcd-4 also correlates with lung tumor 5 progression (Chen et al., 2003). Since hsa-miR-143 positively regulates Pdcd-4 expression, a hsa-miR-143 based therapy may reconstitute Pdcd-4 function. BCL2L1 and MCL1 are members of the anti-apoptotic BCL-2 (B cell lymphoma 2) gene family that give rise to two alternatively spliced gene products with opposing functions (Boise et al., 1993; Bae et al., 2000). The predominantly expressed protein encoded by BCL2L1 is Bcl-XL which - next to 10 BCL-2 - is a major inhibtor of programmed cell death. Overexpression of Bel-XL is detected in numerous cancer types and correlates with tumor progression as well as poor survival (Manion and Hockenbery, 2003). Increased levels of Bcl-XL are also associated with resistance to chemo- and radiotherapy (Fesik, 2005). Transient transfection of hsa-miR-143 leads to a reduction of Bcl-XL transcripts and therefore might provide a therapeutic benefit to 15 oncogenic cells with increased expression of Bcl-XL. Mcl-1 (myeloid leukemia 1) is overexpressed in hepatocellular carcinoma, prostate cancer, testicular tumor, multiple myeloma and various leukemias [see refs in Table 5]. Similar to Bcl-XL, high levels of Mel 1 is correlated with poor prognosis of patients with ovarian carcinoma and is indicative for leukemic relapse (Kauftnann et al., 1998; Shigemasa et al., 2002). RNA interference against 20 Mcl-1 induces a therapeutic response in gastric and hepatocellular carcinoma cells (Schulze Bergkamen et al., 2006; Zangemeister-Wittke and Huwiler, 2006). Molecules regulated by hsa-miR-143 that function in intracellular signal transduction include the inflammatory interleukin 8 (IL-8), transforming growth factor beta (TGF-D) receptor 2 (TGFBR2) and A-kinase anchor protein 12 (AKAP12). IL-8 is frequently 25 upregulated in various cancers and correlates with tumor vascularization, metastasis and poor prognosis (Rosenkilde and Schwartz, 2004; Sparmann and Bar-Sagi, 2004). TGFBR-2 forms a functional complex with TGFBR-1 and is the primary receptor for TGF-P (Massague et al., 2000). Central role of TGF-p is inhibition of cellular growth of numerous cell types, such as epithelial, endothelial, hematopoietic neural and mesenchymal cells. Many mammary and 30 colorectal carcinomas with microsatellite instability harbor inactivating mutations of TGFBR-2, and therefore escape the growth-inhibitory function of TGF-P (Markowitz et al., 1995; Lucke et al., 2001). AKAP12, also referred to as gravin or SSeCKS (Src suppressed C kinase substrate), functions as a kinase scaffold protein that tethers the enzyme-substrate - 136- WO 2008/036718 PCT/US2007/078859 interaction (Nauert et al., 1997). Expression of AKAP12 interferes with oncogenic cell transformation induced by the Src or Jun oncoproteins in vitro and is lost or reduced in numerous cancers, such as leukemia and carcinomas of the rectum, lung and stomach (Lin and Gelman, 1997; Cohen et al., 2001; Xia et al., 2001; Wikman et al., 2002; Boultwood et 5 al., 2004; Choi et al., 2004; Mori et al., 2006). An apparent anti-oncogenic activity of AKAP12 in prostate and gastric cancers marks this protein as a putative tumor suppressor (Xia et al., 2001; Choi et al., 2004). Based on the functions for most of these targets and how they are regulated by hsa miR-143, hsa-miR-143 appears to have tumor suppressor potential. This view is supported 10 by our observation that most cancers show reduced expression of miR-143. However, hsa miR-143 also regulates gene expression in a manner that suggests a role for hsa-miR-143 in the development or progression of disease. For instance, hsa-miR-143 stimulates the expression of thioredoxin (TXN), a 12-kDa thiol reductase targeting various proteins and multiple pathways. Thioredoxin modulates the activity of transcription factors, induces the 15 expression of angiogenic Hif-la (hypoxia induced factor la) as well as VEGF (vascular endothelial growth factor) and can act as a proliferative and anti-apoptotic agent (Marks, 2006). In accord, carcinomas of the lung, pancreas, cervix, and liver show increased levels of thioredoxin. Thioredoxin expression is also correlated with aggressive tumor growth, poor prognosis, and chemoresistance (Marks, 2006). Therefore, a hsa-miR-143 antagonist may 20 have therapeutic potential in cancers that show altered expression of thioredoxin. In summary and not intending to limit the invention by any particular theory, hsa miR-143 governs the activity of proteins that are critical regulators of cell proliferation and survival. These targets are frequently deregulated in human cancer. Based on this review of the genes and related pathways that are regulated by miR-143, introduction of hsa-miR-143 25 or an anti-hsa-miR-143 into a variety of cancer cell types would likely result in a therapeutic response. - 137- WO 2008/036718 PCT/US2007/078859 REFERENCES The following references, to the extent that they provide exemplary procedural or other details supplementary to those set forth herein, are specifically incorporated.herein by reference. 5 U.S. Patent 4,337,063 U.S. Patent 4,404,289 U.S. Patent 4,405,711 U.S. Patent 4,659,774 10 U.S. Patent 4,682,195 U.S. Patent 4,683,202 U.S. Patent 4,704,362 U.S. Patent 4,816,571 U.S. Patent 4,870,287 15 U.S. Patent 4,959,463 U.S. Patent 5,141,813 U.S. Patent 5,143,854 U.S. Patent 5,202,231 U.S. Patent 5,214,136 20 U.S. Patent 5,221,619 U.S. Patent 5,223,618 U.S. Patent 5,242,974 U.S. Patent 5,264,566 U.S. Patent 5,268,486 25 U.S. Patent 5,288,644 U.S. Patent 5,324,633 U.S. Patent 5,378,825 U.S. Patent 5,384,261 U.S. Patent 5,399,363 30 U.S. Patent 5,405,783 U.S. Patent 5,412,087 U.S. Patent 5,424,186 U.S. Patent 5,428,148 -138- WO 2008/036718 PCT/US2007/078859 U.S. Patent 5,429,807 U.S. Patent 5,432,049 U.S. Patent 5,436,327 U.S. Patent 5,445,934 5 U.S. Patent 5,446,137 U.S. Patent 5,466,468 U.S. Patent 5,466,786 U.S. Patent 5,468,613 U.S. Patent 5,470,710 10 U.S. Patent 5,470,967 U.S. Patent 5,472,672 U.S. Patent 5,480,980 U.S. Patent 5,492,806 U.S. Patent 5,503,980 15 U.S. Patent 5,510,270 U.S. Patent 5,525,464 U.S. Patent 5,527,681 U.S. Patent 5,529,756 U.S. Patent 5,532,128 20 U.S. Patent 5,543,158 U.S. Patent 5,545,531 U.S. Patent 5,574,146 U.S. Patent 5,580,726 U.S. Patent 5,580,732 25 U.S. Patent 5,583,013 U.S. Patent 5,593,839 U.S. Patent 5,599,672 U.S. Patent 5,599,695 U.S. Patent 5,602,240 30 U.S. Patent 5,602,244 U.S. Patent 5,610,287 U.S. Patent 5,610,289 U.S. Patent 5,614,617 U.S. Patent 5,623,070 - 139- WO 2008/036718 PCT/US2007/078859 U.S. Patent 5,624,711 U.S. Patent 5,631,134 U.S. Patent 5,637,683 U.S. Patent 5,639,603 5 U.S. Patent 5,641,515 U.S. Patent 5,645,897 U.S. Patent 5,652,099 U.S. Patent 5,654,413 U.S. Patent 5,658,734 10 U.S. Patent 5,661,028 U.S. Patent 5,665,547 U.S. Patent 5,667,972 U.S. Patent 5,670,663 U.S. Patent 5,672,697 15 U.S. Patent 5,677,195 U.S. Patent 5,681,947 U.S. Patent 5,695,940 U.S. Patent 5,700,637 U.S. Patent 5,700,922 20 U.S. Patent 5,705,629 U.S. Patent 5,708,153 U.S. Patent 5,708,154 U.S. Patent 5,714,606 U.S. Patent 5,728,525 25 U.S. Patent 5,728,525 U.S. Patent 5,739,169 U.S. Patent 5,744,305 U.S. Patent 5,744,305 U.S. Patent 5,760,395 30 U.S. Patent 5,763,167 U.S. Patent 5,770,358 U.S. Patent 5,777,092 U.S. Patent 5,789,162 U.S. Patent 5,792,847 -140- WO 2008/036718 PCT/US2007/078859 U.S. Patent 5,800,992 U.S. Patent 5,801,005 U.S. Patent 5,807,522 U.S. Patent 5,824,311 5 U.S. Patent 5,830,645 U.S. Patent 5,830,880 U.S. Patent 5,837,196 U.S. Patent 5,846,225 U.S. Patent 5,846,945 10 U.S. Patent 5,847,219 U.S. Patent 5,856,174 U.S. Patent 5,858,988 U.S. Patent 5,859,221 U.S. Patent 5,871,928 15 U.S. Patent 5,872,232 U.S. Patent 5,876,932 U.S. Patent 5,886,165 U.S. Patent 5,919,626 U.S. Patent 5,922,591 20 U.S. Patent 6,004,755 U.S. Patent 6,040,193 U.S. Patent 6,040,193 U.S. Patent 6,087,102 U.S. Patent 6,251,666 25 U.S. Patent 6,368,799 U.S. Patent 6,383,749 U.S. Patent 6,617,112 U.S. Patent 6,638,717 U.S. Patent 6,720,138 30 U.S. Patent 6,723,509 U.S. Patent Serial 09/545,207 U.S. Patent Serial 10/667,126 U.S. Patent Serial 11/141,707 U.S. Patent Serial 11/273,640 - 141 - WO 2008/036718 PCT/US2007/078859 U.S. Patent Serial 11/349,727 U.S. Patent Serial 60/575,743 U.S. Patent Serial 60/649,584 U.S. Patent Serial 60/650,807 5 Akao et al., Oncol Reports, 16:845-850, 2006. Akiba etal., IntJOncol, 18(2):257-264, 2001. Arap et al., Cancer Res., 55(6):1351-1354, 1995. Austin-Ward and Villaseca, Revista Medica de Chile, 126(7):838-845, 1998. 10 Bae et al., JBiol Chem, 275(33):25255-25261, 2000. Bagga et al., Cell, 122(4):553-563, 2005. Biswas et al., Cancer Res, 64(14):4687-4692, 2004. Bitomsky et al., Oncogene, 23(45):7484-7493, 2004. Boise et al., Cell, 74(4):597-608, 1993. 15 Boultwood et al., Br JHaematol, 126(4):508-511, 2004. Bukowski et al., Clinical Cancer Res., 4(10):2337-2347, 1998. Caldas et al., Cancer Res., 54:3568-3573, 1994. Calin and Croce, Nat Rev Cancer, 6(11):857-866, 2006. Carrington and Ambros, Science, 301(5631):336-338, 2003. 20 Chen et al., JPathol, 200(5):640-646, 2003. Cheng et al., Cancer Res., 54(21):5547-5551, 1994. Choi et al., Oncogene, 23(42):7095-7103, 2004. Cho-Vega et al., Hum Pathol, 35(9):1095-1100, 2004. Christodoulides et al., Microbiology, 144(Pt 11):3027-3037, 1998. 25 Claudio et al., Clin Cancer Res, 8(6):1808-1815, 2002. Cohen et al., Oncogene, 20(2):141-146, 2001. Cummins et al., In: IRT: Nucleosides and nucleosides, La Jolla CA, 72, 1996. Davidson et al., J. Immunother., 21(5):389-398, 1998. Denli et al., Trends Biochem. Sci., 28:196, 2003. 30 Didenko, Biotechniques, 31(5):1106-1116, 1118, 1120-1121, 2001. Dillman, Cancer Biother. Radiopharm., 14(1):5-10, 1999. Donnellan and Chetty, Mol Pathol, 51(1):1-7, 1998. Emptage et al., Neuron, 29(1):197-208, 2001. EP 266,032 - 142 - WO 2008/036718 PCT/US2007/078859 EP 373 203 EP 785 280 EP 799 897 Esau et al., JBiol Chem, 279(50):52361-52365, 2004. 5 Esquela-Kerscher and Slack, Nat Rev Cancer, 6(4):259-269, 2006. Fesik, Nat Rev Cancer, 5(11):876-885, 2005. Firth and Baxter, Endocr Rev, 23(6):824-854, 2002. Fisher, J Royal Statistical Soc 85(1):87-94, 1922. Fleischer et al., Int J Oncol, 28(1):25-32, 2006. 10 Fodor et al., Biochemistry, 30(33):8102-8108, 1991. Froehler et al., Nucleic Acids Res., 14(13):5399-5407, 1986. Gao et al., Oncol Rep, 17(1):123-128, 2007. Goke et al., Am JPhysiol Cell Physiol, 287(6):C1 541-1546, 2004. Griffey et al., J Mass Spectrom, 32(3):305-13, 1997. 15 Hanahan and Weinberg, Cell, 100(1):57-70, 2000. Hanibuchi et al., Int. J. Cancer, 78(4):480-485, 1998. Hellstrand et al., Acta Oncologica, 37(4):347-353, 1998. Hui and Hashimoto, Infection Immun., 66(11):5329-5336, 1998. Hussussian et al., Nat. Genet., 8(1):15-21, 1994. 20 Itakura and Riggs, Science, 209:1401-1405, 1980. Ito et al., Anticancer Res, 23(5A):3819-3824, 2003. Jansen et al., Cancer Res, 65(14):6034-6041, 2005. Jansen et al., Mol Cancer Ther, 3(2):103-110, 2004. Ju et al., Gene Ther., 7(19):1672-1679, 2000. 25 Kamb et al., Nat. Genet., 8(1):23-26, 1994. Kamb et al., Science, 2674:436-440, 1994. Kaufmann et al., Blood, 91(3):991-1000, 1998. Kitada et al., Blood, 91(9):3379-3389, 1998. Klostermeier and Millar, Biopolymers, 61(3):159-79, 2001-2002. 30 Kornberg and Baker, In: DNA Replication, 2d Ed., Freeman, San Francisco, 1992. Krajewska et al., Am JPathol, 148(5):1567-1576, 1996. Krek et al., Nature Genet 37:495-500, 2005. Lagos-Quintana et al., Science, 294(5543):853-858, 2001. Lau et al., Science, 294(5543):858-862, 2001. - 143 - WO 2008/036718 PCT/US2007/078859 Lee and Ambros, Science, 294(5543):862-864, 2001. Lim et al., Nature, 433(7027):769-773, 2005. Lin and Gelman, Cancer Res, 57(11):2304-2312, 1997. Lucke et al., Cancer Res, 61(2):482-485, 2001. 5 Manion and Hockenbery, Cancer Biol Ther, 2(4 Suppl 1):S105-114, 2003. Markowitz, Biochim Biophys Acta, 1470(1):M13-20, 2000. Marks, Semin Cancer Biol, 16(6):436-443, 2006. Marsters et al., Recent Prog. Horm. Res., 54:225-234, 1999. Massague et al., Cell, 103(2):295-309, 2000. 10 McGowan and Russell, Embo J, 12(1):75-85, 1993. Michael et al., Mol Cancer Res, 1:882-891, 2003. Mizunuma et al., Br J Cancer, 88(10):1543-1548, 2003. Mori et al., Cancer Res., 54(13):3396-3397, 1994. Mori et al., Gastroenterology, 131(3):797-808, 2006. 15 Nauert et al., Curr Biol, 7(1):52-62, 1997. Nobori et al., Nature, 368(6473):753-756, 1994. Okamoto et al., Proc. Nati. Acad. Sci. USA, 91(23):11045-11049, 1994. Olsen et al., Dev. Biol., 216:671, 1999. Orlow et al., Cancer Res, 54(11):2848-2851, 1994. 20 Orlow et al., Int. J. Oncol., 15(1):17-24, 1994. Parker and Piwnica-Worms, Science, 257(5078):1955-1957, 1992. PCT Appln. WO 0138580 PCT Appln. WO 0168255 PCT Appln. WO 03020898 25 PCT Appln. WO 03022421 PCT Appln. WO 03023058 PCT Appln. WO 03029485 PCT Appln. WO 03040410 PCT Appln. WO 03053586 30 PCT Appln. WO 03066906 PCT Appln. WO 03067217 PCT Appln. WO 03076928 PCT Appln. WO 03087297 PCT Appln. WO 03091426 -144- WO 2008/036718 PCT/US2007/078859 PCT AppIn. WO 03093810 PCT AppIn. WO 03100012 PCT AppIn. WO 03100448A1 PCT AppIn. WO 04020085 5 PCT AppIn. WO 04027093 PCT AppIn. WO 09923256 PCT AppIn. WO 09936760 PCT AppIn. WO 93/17126 PCT AppIn. WO 95/11995 10 PCT Appln. WO 95/21265 PCT AppIn. WO 95/21944 PCT Appln. WO 95/21944 PCT AppIn. WO 95/35505 PCT AppIn. WO 96/31622 15 PCT Appln. WO 97/10365 PCT Appln. WO 97/27317 PCT Appln. WO 9743450 PCT Appln. WO 99/35505 Pietras et al., Oncogene, 17(17):2235-2249, 1998. 20 Pruitt et al., Nucleic Acids Res, 33(1):D501-D504, 2005. Qin et al., Proc. Natl. Acad. Sci. USA, 95(24):14411-14416, 1998. Reimer et al., JBiol Chem, 274(16):11022-11029, 1999. Remington's Pharmaceutical Sciences" 15th Edition, pages 1035-1038 and 1570-1580, 1990. Rosenkilde and Schwartz, Apmis, 112(7-8):481-495, 2004. 25 Rust et al., J Clin Pathol, 58(5):520-524, 2005. Sambrook and Russell, Molecular Cloning: A Laboratory Manual 3 rd Ed., Cold Spring Harbor Laboratory Press, 2001. Sambrook et al., In: DNA microaarays: a molecular cloning manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, 2003. 30 Sambrook et al., In: Molecular cloning: a laboratory manual, 2 "nd Ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, 1989. Sano et al., Histopathology, 46(5):532-539, 2005. Scheit, In: Synthesis and Biological Function, Wiley-Interscience, New York, 171-172, 1980. Schulze-Bergkamen et al., BMC Cancer, 6:232, 2006. - 145 - WO 2008/036718 PCT/US2007/078859 Seggerson et al., Dev. Biol., 243:215, 2002. Serrano et al., Nature, 366:704-707, 1993. Serrano et al., Science, 267(5195):249-252, 1995. Sherr and McCormick, Cancer Cell, 2(2):103-112, 2002. 5 Shigemasa et al., Jpn J Cancer Res, 93(5):542-550, 2002. Sieghart et al., JHepatol, 44(1):151-157, 2006. Skotzko et al., Cancer Res, 55(23):5493-5498, 1995. Spannann and Bar-Sagi, Cancer Cell, 6(5):447-458, 2004. Sum et al., JBiol Chem, 277(10):7849-7856, 2002. 10 Sum et al., Proc Natl Acad Sci US A, 102(21):7659-7664, 2005. Takimoto et al., Biochem Biophys Res Commun, 251(1):264-268, 1998. Taniwaki et al., Int J Oncol, 29(3):567-575, 2006. U.K.. Patent 1,529,202 U.K.. Patent 8 803 000 15 Visvader et al., Proc Natl Acad Sci USA, 98(25):14452-14457, 2001. Wikman et al., Oncogene, 21(37):5804-5813, 2002. Wu et al., Eur J Cancer, 38(14):1838-1848, 2002. Wuilleme-Toumi et al., Leukemia, 19(7):1248-1252, 2005. Xia et al., Cancer Res, 61(14):5644-5651, 2001 a. 20 Xia et al., Cancer, 91(8):1429-1436, 2001b. Yang et al., Cancer Cell, 9(6):445-457, 2006a. Yang et al., Mol Cell Biol, 23(1):26-37, 2003. Yang et al., Mol Cell Biol, 26(4):1297-1306, 2006b. Yoshida et al., Ann Oncol, 15(2):252-256, 2004. 25 Zangemeister-Wittke and Huwiler, Cancer Biol Ther, 5(10):1355-1356, 2006. Zhang et al., Oncogene, 25(45):6101-6112, 2006. Zhao et al., Mol Cancer Res, 1(3):195-206, 2003. - 146-

Claims (44)

1. A method of modulating gene expression in a cell comprising administering to the cell an amount of an isolated nucleic acid comprising a miR-143 in an amount sufficient to modulate the expression of one or more genes identified in Table 1, 3, 4, or 5. 5
2. The method of claim 1, wherein the cell is in a subject having, suspected of having, or at risk of developing a metabolic, an immunologic, an infectious, a cardiovascular, a digestive, an endocrine, an ocular, a genitourinary, a blood, a musculoskeletal, a nervous system, a congenital, a respiratory, a skin, or a cancerous disease or condition.
3. The method of claim 2, wherein the infectious disease or condition is a parasitic, 10 bacterial, viral, or fungal infection.
4. The method of claim 2, wherein the cancerous condition is astrocytoma, anaplastic large cell lymphoma, acute lymphoblastic leukemia, acute myelogenous leukemia, breast carcinoma, B-cell lymphoma, bladder carcinoma, cervical carcinoma, chronic lymphoblastic leukemia, colorectal carcinoma, endometrial carcinoma, glioma, glioblastoma, gastric 15 carcinoma, hepatocellular carcinoma, Hodgkin lymphoma, leukemia, lung carcinoma, melanoma, medulloblastoma, mantle cell lymphoma, multiple myeloma, myeloma, non Hodgkin lymphoma, non-small cell lung carcinoma, ovarian carcinoma, oligodendroglioma, oesophageal carcinoma, osteosarcoma, pancreatic carcinoma, prostate carcinoma, renal cell carcinoma, squamous cell carcinoma of the head and neck, small cell lung carcinoma, thyroid 20 carcinoma, testicular tumor wherein the modulation of one or more gene is sufficient for a therapeutic response.
5. The method of claim 1, wherein the expression of a gene is up-regulated.
6. The method of claim 1, wherein the cell is an epithelial, a stromal, or a mucosal cell.
7. The method of claim 1, wherein the cell is a brain, a neuronal, a blood, an esophageal, 25 a lung, a cardiovascular, a liver, a breast, a bone, a thyroid, a glandular, an adrenal, a pancreatic, a stomach, an intestinal, a kidney, a bladder, a prostate, a cervical, a uterine, an ovarian, a testicular, a splenic, a skin, a smooth muscle, a cardiac muscle, a striated muscle cell.
8. The method of claim 1, wherein the cell is a cancer cell. -147- WO 2008/036718 PCT/US2007/078859
9. The method of claim 8, wherein the cancer cell is a neuronal, glial, lung, liver, brain, breast, bladder, blood, leukemic, colon, endometrial, stomach, skin, ovarian, fat, bone, cervical, esophageal, pancreatic, prostate, kidney, or thyroid cell.
10. The method of claim 1, wherein the isolated miR- 143 is a recombinant nucleic acid. 5
11. The method of claim 10, wherein the recombinant nucleic acid is RNA.
12. The method of claim 10, wherein the recombinant nucleic acid is DNA.
13. The method of claim 12, wherein the recombinant nucleic acid comprises a miR-143 inhibitor expression cassette.
14. The method of claim 13, wherein the expression cassette is comprised in a viral 10 vector, or plasmid DNA vector.
15. The method of claim 14, wherein the viral vector is administered at a dose of 1x10 5 to 1xIO1 4 viral particles per dose or the plasmid DNA vector is administered at a dose of 100 mg per patient to 4000 mg per patient.
16. The method of claim 1, wherein the miR-143 nucleic acid is a synthetic nucleic acid. 15
17. The method of claim 16, wherein the nucleic acid is administered at a dose of 0.01 mg/kg of body weight to 10 mg/kg of body weight.
18. The method of claim 1, wherein the miR-143 is a hsa-miR-143.
19. The method of claim 1, wherein the nucleic acid is administered enterally or parenterally.
20 20. The method of claim 19, wherein enteral administration is orally.
21. The method of claim 19, wherein parenteral administration is intravascular, intracranial, intrapleural, intratumoral, intraperitoneal, intramuscular, intralymphatic, intraglandular, subcutaneous, topical, intrabronchial, intratracheal, intranasal, inhaled, or instilled. 25
22. The method of claim 1, wherein the nucleic acid is comprised in a phannaceutical formulation. -148 - WO 2008/036718 PCT/US2007/078859
23. The method of claim 22, wherein the pharmaceutical formulation is a lipid composition.
24. A method of modulating a cellular pathway or a physiologic pathway comprising administering to a cell an amount of an isolated nucleic acid comprising a miR-143 inhibitor 5 in an amount sufficient to modulate the cellular pathway or physiologic pathway that includes one or more genes identified or gene products related to one or more genes identified in Table 1, 3, 4, or 5.
25. The method of claim 24, further comprising administering 2, 3, 4, 5, 6, or more miRNAs. 10
26. The method claim 25 wherein the miRNAs are comprised in a single composition.
27. The method of 23, wherein at least two cellular pathways or physiologic pathways are modulated.
28. The method of claim 25, wherein at least one gene is modulated by multiple miRNAs.
29. The method of claim 24, wherein the expression of a gene or a gene product is down 15 regulated.
30. The method of claim 24, wherein the expression of a gene or a gene product is up regulated.
31. The method of claim 24, wherein the cell is a cancer cell.
32. The method of claim 31, wherein viability of the cell is reduced, proliferation of the 20 cell is reduced, metastasis of the cell is reduced, or the cell's sensitivity to therapy is increased.
33. The method of claim 31, wherein the cancer cell is a neuronal, glial, lung, liver, brain, breast, bladder, blood, leukemic, colon, endometrial, stomach, intestinal, skin, ovarian, fat, bone, cervical, esophageal, pancreatic, prostate, kidney, testicular, or thyroid cell. 25
34. The method of claim 24, wherein the miR-143 is a recombinant nucleic acid.
35. The method of claim 34, wherein the recombinant nucleic acid is DNA. -149 - WO 2008/036718 PCT/US2007/078859
36. The method of claim 35, wherein the recombinant nucleic acid is a viral vector or a plasmid DNA vector.
37. A method of treating a patient diagnosed with or suspected of having or suspected of developing a pathological condition or disease related to a gene modulated by a miRNA 5 comprising the steps of: (a) administering to the patient an amount of an isolated nucleic acid comprising a miR-143 in an amount sufficient to modulate a cellular pathway or a physiologic pathway; and (b) administering a second therapy, wherein the modulation of the cellular 10 pathway or physiologic pathway sensitizes the patient to the second therapy.
38. The method of claim 37, wherein one or more cellular pathway or physiologic pathway includes one or more genes identified in Table 1, 3, 4, or 5.
39. A method of selecting a miRNA to be administered to a subject with, suspected of having, or having a propensity for developing a pathological condition or disease comprising: 15 (a) determining an expression profile of one or more genes selected from Table 1, 3, 4, or 5; (b) assessing the sensitivity of the subject to miRNA therapy based on the expression profile; and (c) selecting one or more miRNA based on the assessed sensitivity. 20
40. The method of claim 39 further comprising treating the subject with 1, 2, 4, 5, 6, 7, 8, 9, 10, or more miRNAs.
41. The method of claim 40, wherein each miRNA is administered individually or one or more combinations.
42. The method of claim 41, wherein the miRNAs are in a single composition. 25
43. A method of assessing a cell, tissue, or subject comprising assessing expression of miR- 143 in combination with assessing expression of one or more gene from Table 1, 3, 4, or 5 in at least one sample.
44. A method of assessing miR-143 status in a sample comprising the steps of: -150- WO 2008/036718 PCT/US2007/078859 (a) assessing expression of one or more genes from Table 1, 3, 4, or 5 in a sample; and (b) determining miR-143 status based on level of miR-143 expression in the sample. - 151 -
AU2007299873A 2006-09-19 2007-09-19 miR-143 regulated genes and pathways as targets for therapeutic intervention Abandoned AU2007299873A1 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US82617306P 2006-09-19 2006-09-19
US60/826,173 2006-09-19
US93957307P 2007-05-22 2007-05-22
US60/939,573 2007-05-22
PCT/US2007/078859 WO2008036718A2 (en) 2006-09-19 2007-09-19 Mir-143 regulated genes and pathways as targets for therapeutic intervention

Publications (1)

Publication Number Publication Date
AU2007299873A1 true AU2007299873A1 (en) 2008-03-27

Family

ID=38866587

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2007299873A Abandoned AU2007299873A1 (en) 2006-09-19 2007-09-19 miR-143 regulated genes and pathways as targets for therapeutic intervention

Country Status (6)

Country Link
EP (1) EP2094848A2 (en)
JP (1) JP2010504349A (en)
AU (1) AU2007299873A1 (en)
CA (1) CA2663812A1 (en)
IL (1) IL197691A0 (en)
WO (1) WO2008036718A2 (en)

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10378060B2 (en) 2011-10-14 2019-08-13 Dana-Farber Cancer Institute, Inc. ZNF365/ZFP365 biomarker predictive of anti-cancer response
US11236337B2 (en) 2016-11-01 2022-02-01 The Research Foundation For The State University Of New York 5-halouracil-modified microRNAs and their use in the treatment of cancer
CN110290794A (en) 2016-11-01 2019-09-27 纽约州州立大学研究基金会 The microRNA and its purposes in cancer treatment of 5- halo uracil modification
CN110638828B (en) * 2019-10-16 2023-05-05 中国人民解放军军事科学院军事医学研究院 Use of substances inhibiting CWF19L1 protein activity and/or expression level in inhibiting RSV replication

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7683036B2 (en) * 2003-07-31 2010-03-23 Regulus Therapeutics Inc. Oligomeric compounds and compositions for use in modulation of small non-coding RNAs
AU2006254732A1 (en) * 2005-06-03 2006-12-07 Southern Adelaide Health Service-Flinders Medical Centre Targeting cells with altered microrna expression

Also Published As

Publication number Publication date
WO2008036718A2 (en) 2008-03-27
EP2094848A2 (en) 2009-09-02
IL197691A0 (en) 2011-08-01
JP2010504349A (en) 2010-02-12
CA2663812A1 (en) 2008-03-27
WO2008036718A3 (en) 2008-05-22

Similar Documents

Publication Publication Date Title
US20090232893A1 (en) miR-143 REGULATED GENES AND PATHWAYS AS TARGETS FOR THERAPEUTIC INTERVENTION
US8071562B2 (en) MiR-124 regulated genes and pathways as targets for therapeutic intervention
US20090227533A1 (en) miR-34 Regulated Genes and Pathways as Targets for Therapeutic Intervention
US20090163434A1 (en) miR-20 Regulated Genes and Pathways as Targets for Therapeutic Intervention
US20090192102A1 (en) miR-21 REGULATED GENES AND PATHWAYS AS TARGETS FOR THERAPEUTIC INTERVENTION
EP2104737B1 (en) Functions and targets of let-7 micro rnas
US20090163435A1 (en) miR-200 REGULATED GENES AND PATHWAYS AS TARGETS FOR THERAPEUTIC INTERVENTION
US20090175827A1 (en) miR-16 REGULATED GENES AND PATHWAYS AS TARGETS FOR THERAPEUTIC INTERVENTION
US20090131356A1 (en) 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
US20090131354A1 (en) miR-126 REGULATED GENES AND PATHWAYS AS TARGETS FOR THERAPEUTIC INTERVENTION
CN101627121A (en) As the miRNA regulatory gene and the path for the treatment of the target of intervening
US20090192114A1 (en) miR-10 Regulated Genes and Pathways as Targets for Therapeutic Intervention
AU2007342068A1 (en) miR-16 regulated genes and pathways as targets for therapeutic intervention
US20100305188A1 (en) Nucleic acid capable of regulating the proliferation of cell
EP2104736B1 (en) Mir-126 regulated genes and pathways as targets for therapeutic intervention
WO2012006181A2 (en) Compositions and methods for inhibiting oncogenic micrornas and treatment of cancer
AU2007299873A1 (en) miR-143 regulated genes and pathways as targets for therapeutic intervention

Legal Events

Date Code Title Description
MK1 Application lapsed section 142(2)(a) - no request for examination in relevant period