MicroRNA Fingerprints During Human Megakaryocytopoiesis

Described herein are compositions and methods of decreasing expression of MAFB in a subject having a cancer and/or myeloproliferative disorder associated with overexpression of a MAFB gene product where an effective amount of at least one miR-130a gene product or an isolated variant or biologically-active fragment thereof is administered to the subject sufficient to decrease expression of the MAFB gene product in the subject.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
CROSS-REFERENCE TO RELATED APPLICATIONS

This application is a divisional application of U.S. patent application Ser. No. 13/595,200, filed Aug. 27, 2012, now allowed, which is a divisional claiming priority to U.S. patent application Ser. No. 13/169,184, filed Jun. 27, 2011, now U.S. Pat. No. 8,354,224, issued Jan. 15, 2013, which is a divisional claiming priority to U.S. patent application Ser. No. 12/293,471, having a national stage entry date of Oct. 9, 2008, now U.S. Pat. No. 7,985,584, issued Jul. 26, 2011, which is a national stage application filed under 35 USC §371 of international application PCT/US2007/006824 filed Mar. 19, 2007, which claims the benefit of U.S. Provisional Application No. 60/743,585 filed Mar. 20, 2006, the entire disclosures of each of which are expressly incorporated herein by reference for all purposes.

STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH

The invention was supported, in whole or in part, by National Institutes of Health Program Project Grants PO1CA76259, PO1CA16058, PO1CA81534 and P01CA16672. The Government has certain rights in the invention.

STATEMENT REGARDING THE SEQUENCE LISTING

The present application is being filed along with a Sequence Listing in electronic format. The Sequence Listing is provided as a file entitled 60428351_SEQ_ID_LISTING_OSURF-06140.txt, prepared on Oct. 23, 2008, and is 96,224 bytes in size. The information in the electronic format of the Sequence Listing is incorporated herein by reference in its entirety.

BACKGROUND OF THE INVENTION

MicroRNAs (miRNAs) are a small non-coding family of 19-25 nucleotide RNAs that regulate gene expression by targeting messenger RNAs (mRNA) in a sequence specific manner, inducing translational repression or mRNA degradation depending on the degree of complementarity between miRNAs and their targets (Bartel, D. P. (2004) Cell 116, 281-297; Ambros, V. (2004) Nature 431, 350-355). Many miRNAs are conserved in sequence between distantly related organisms, suggesting that these molecules participate in essential processes. Indeed, miRNAs are involved in the regulation of gene expression during development (Xu, P., et al. (2003) Curr. Biol. 13, 790-795), cell proliferation (Xu, P., et al. (2003) Curr. Biol. 13, 790-795), apoptosis (Cheng, A. M., et al. (2005) Nucl. Acids Res. 33, 1290-1297), glucose metabolism (Poy, M. N., et al. (2004) Nature 432, 226-230), stress resistance (Dresios, J., et al. (2005) Proc. Natl. Acad. Sci. USA 102, 1865-1870) and cancer (Calin, G. A, et al. (2002) Proc. Natl. Acad. Sci. USA 99, 1554-15529; Calin, G. A., et al. (2004) Proc. Natl. Acad. Sci. USA 101, 11755-11760; He, L., et al. (2005) Nature 435, 828-833; and Lu, J., et al. (2005) Nature 435:834-838).

There is also strong evidence that miRNAs play a role in mammalian hematopoiesis. In mice, miR-181, miR-223 and miR-142 are differentially expressed in hematopoietic tissues, and their expression is regulated during hematopoiesis and lineage commitment (Chen, C. Z., et al. (2004) Science 303, 83-86). The ectopic expression of miR-181 in murine hematopoietic progenitor cells led to proliferation in the B-cell compartment (Chen, C. Z., et al. (2004) Science 303, 83-86). Systematic miRNA gene profiling in cells of the murine hematopoietic system revealed different miRNA expression patterns in the hematopoietic system compared with neuronal tissues, and identified individual miRNA expression changes that occur during cell differentiation (Monticelli, S., et al. (2005) Genome Biology 6, R71). A recent study has identified down-modulation of miR-221 and miR-222 in human erythropoietic cultures of CD34+ cord blood progenitor cells (Felli, N., et al. (2005) Proc. Natl. Acad. Sci. USA. 102, 18081-18086). These miRNAs were found to target the oncogene c-Kit. Further functional studies indicated that the decline of these two miRNAs in erythropoietic cultures unblocks Kit protein production at the translational level leading to expansion of early erythroid cells (Felli, N., et al. (2005) Proc. Natl. Acad. Sci. USA. 102, 18081-18086). In line with the hypothesis of miRNAs regulating cell differentiation, miR-223 was found to be a key member of a regulatory circuit involving C/EBPa and NFI-A, which controls granulocytic differentiation in all-trans retinoic acid-treated acute promyelocytic leukemic cell lines (Fazi, F., et al. (2005) Cell 123, 819-831).

miRNAs have also been found deregulated in hematopoietic malignancies. Indeed, the first report linking miRNAs and cancer involved the deletion and down regulation of the miR-15a and miR-16-1 cluster, located at chromosome 13q14.3, a commonly-deleted region in chronic lymphocytic leukemia (Cahn, G. A, et al. (2002) Proc. Natl. Acad. Sci. USA 99, 1554-15529). High expression of miR-155 and host gene BIC was also reported in B-cell lymphomas (Metzler M., et al. (2004) Genes Chromosomes and Cancer 39; 167-169). More recently it was shown that the miR-17-92 cluster, which is located in a genomic region of amplification in lymphomas, is overexpressed in human B-cell lymphomas and the enforced expression of this cluster acted in concert with c-MYC expression to accelerate tumor development in a mouse B cell lymphoma model (He, L., et al. (2005) Nature 435, 828-833). These observations indicate that miRNAs are important regulators of hematopoiesis and can be involved in malignant transformation.

Platelets play an essential role in hemostasis and thrombosis. They are produced from in large numbers from their parent cells, bone marrow megakaryocytes, and arise from fragmentation of the cytoplasm. Only recently has the molecular basis of what may turn out to be a large family of related disorders affecting platelet production started to be defined. If the level of circulating platelets drops below a certain number (thrombocytopenia), the patient runs the risk of catastrophic hemorrhage. Patients with cancer who have received chemotherapy or bone marrow transplants usually have thrombocytopenia, and the slow recovery of platelet count in these patients has been a concern. The demand for platelet units for transfusion has been steadily increasing primarily because of the need to maintain a certain platelet level in such patients with cancer or those undergoing major cardiac surgery.

Identification of microRNAs that are differentially-expressed in cancer cells (e.g., leukemia cells) may help pinpoint specific miRNAs that are involved in cancer and other disorders (e.g., platelet disorders). Furthermore, the identification of putative targets of these miRNAs may help to unravel their pathogenic role. In particular, discovering the patterns and sequence of miRNA expression during hematopoietic differentiation may provide insights about the functional roles of these tiny non-coding genes in normal and malignant hematopoiesis.

There is a need for novel methods and compositions for the diagnosis, prognosis and treatment of cancer, myeloproliferative disorders and platelet disorders (e.g., inherited platelet disorders).

SUMMARY OF THE INVENTION

The present invention is based, in part, on the identification of specific miRNAs that are involved in megakaryocytic differentiation and/or have altered expression levels in cancerous cells (e.g., in acute megakaryoblastic leukemia (AMKL cell lines)). In the present study, the miRNA gene expression in human megakaryocyte cultures from bone marrow CD34+ progenitors and acute megakaryoblastic leukemia cell lines was investigated. The results of this analysis indicate that several miRNAs are downregulated during normal megakaryocytic differentiation. The results further demonstrate that these miRNAs target genes involved in megakaryocytopoiesis, while others are over expressed in cancer cells.

Accordingly, the invention encompasses methods of diagnosing or prognosticating cancer and/or a myeloproliferative disorder in a subject (e.g., a human). According to the methods of the invention, the level of at least one miR gene product in a test sample from the subject is compared to the level of a corresponding miR gene product in a control sample. An alteration (e.g., an increase, a decrease) in the level of the miR gene product in the test sample, relative to the level of a corresponding miR gene product in the control sample, is indicative of the subject either having, or being at risk for developing, cancer and/or a myeloproliferative disorder. In one embodiment, the level of the miR gene product in the test sample from the subject is greater than that of the control. In another embodiment, the at least one miR gene product is selected from the group consisting of miR-101, miR-126, miR-99a, miR-99-prec, miR-106, miR-339, miR-99b, miR-149, miR-33, miR-135 and miR-20. In still another embodiment, the at least one miR gene product is selected from the group consisting of miR-101, miR-126, miR-106, miR-20 and miR-135. In yet another embodiment, the at least one miR gene product is selected from the group consisting of miR-106, miR-20 and miR-135. In particular embodiments, the cancer that is diagnosed or prognosticated is a leukemia (e.g., acute myeloid leukemia (e.g., acute megakaryoblastic leukemia)) or multiple myeloma. In other embodiments, the myeloproliferative disorder is selected from the group consisting of essential thrombocytemia (ET), polycythemia vera (PV), myelodisplasia, myelofibrosis (e.g., agnogenic myeloid metaplasia (AMM) (also referred to as idiopathic myelofibrosis)) and chronic myelogenous leukemia (CML).

In another embodiment, the invention is a method of treating a cancer and/or a myeloproliferative disorder in a subject (e.g., a human). In the method, an effective amount of a compound for inhibiting expression of at least one miR gene product selected from the group consisting of miR-101, miR-126, miR-99a, miR-99-prec, miR-106, miR-339, miR-99b, miR-149, miR-33, miR-135 and miR-20 is administered to the subject. In one embodiment, the compound for inhibiting expression of at least one miR gene product inhibits expression of a miR gene product selected from the group consisting of miR-101, miR-126, miR-106, miR-20 and miR-135. In another embodiment, the compound for inhibiting expression of at least one miR gene product inhibits expression of a miR gene product selected from the group consisting of miR-106, miR-20 and miR-135. In particular embodiments, the cancer that is treated is a leukemia (e.g., acute myeloid leukemia (e.g., acute megakaryoblastic leukemia)) or multiple myeloma. In other embodiments, the myeloproliferative disorder is selected from the group consisting of essential thrombocytemia (ET), polycythemia vera (PV), myelodisplasia, myelofibrosis (e.g., agnogenic myeloid metaplasia (AMM)) and chronic myelogenous leukemia (CML).

In another embodiment, the invention is a method of treating a cancer and/or a myeloproliferative disorder associated with overexpression of a MAFB gene product in a subject (e.g., a human). In the method, an effective amount of at least one miR gene product or a variant or biologically-active fragment thereof, which binds to, and decreases expression of, the MAFB gene product, is administered to the subject. In one embodiment, the at least one miR gene product, variant or biologically-active fragment thereof comprises a nucleotide sequence that is complementary to a nucleotide sequence in the MAFB gene product. In another embodiment, the at least one miR gene product is miR-130a or a variant or biologically-active fragment thereof. Cancers and myeloproliferative disorders suitable for treatment using this method include, for example, those described herein.

In another embodiment, the invention is a method of treating a cancer and/or a myeloproliferative disorder associated with overexpression of a HOXA1 gene product in a subject (e.g., a human). In the method, an effective amount of at least one miR gene product or a variant or biologically-active fragment thereof, which binds to, and decreases expression of, the HOXA1 gene product, is administered to the subject. In one embodiment, the at least one miR gene product, variant or biologically-active fragment thereof comprises a nucleotide sequence that is complementary to a nucleotide sequence in the HOXA1 gene product. In another embodiment, the at least one miR gene product is miR-10a or a variant or biologically-active fragment thereof. Cancers and myeloproliferative disorders suitable for treatment using this method include, for example, those described herein.

In one embodiment, the invention is a method of determining and/or predicting megakaryocytic differentiation. In this method, the level of at least one miR gene product in a sample (e.g., a sample from a subject (e.g., a human)) comprising megakaryocyte progeny and/or megakaryocytes is determined. That level is compared to the level of the corresponding miR gene product in a control. An alteration in the level of the at least one miR gene product in the sample, relative to that of the control, is indicative of megakaryocytic differentiation. In one embodiment, the alteration is a decrease in the level of the at least one miR gene product in the sample. In another embodiment, the at least one miR gene product is selected from the group consisting of miR-10a, miR-126, miR-106, miR-010b, miR-130a, miR-130a-prec, miR-124a, miR-032-prec, miR-101, miR-30c, miR-213, miR-132-prec, miR-150, miR-020, miR-339, let-7a, let-7d, miR-181c, miR-181b and miR-017. In still another embodiment, the at least one miR gene product is selected from the group consisting of miR-10a, miR-10b, miR-30c, miR-106, miR-126, miR-130a, miR-132, and miR-143.

The invention further provides pharmaceutical compositions for treating cancer and/or a myeloproliferative disorder. In one embodiment, the pharmaceutical compositions of the invention comprise at least one miR expression-inhibition compound and a pharmaceutically-acceptable carrier. In a particular embodiment, the at least one miR expression-inhibition compound is specific for a miR gene product whose expression is greater in cancer cells (e.g., acute megakaryoblastic leukemia (AMKL) cells) than control cells (i.e., it is upregulated). In one embodiment, the miR expression-inhibition compound is specific for one or more miR gene products selected from the group consisting of miR-101, miR-126, miR-99a, miR-99-prec, miR-106, miR-339, miR-99b, miR-149, miR-33, miR-135 and miR-20. In another embodiment, the miR expression-inhibition compound is specific for one or more miR gene products selected from the group consisting of miR-101, miR-126, miR-106, miR-20, and miR-135. In still another embodiment, the miR expression-inhibition compound is specific for one or more miR gene products selected from the group consisting of miR-106, miR-20 and miR-135. In yet another embodiment, the pharmaceutical composition further comprises at least one anti-cancer agent.

In one embodiment, the invention is a pharmaceutical composition for treating a cancer associated with overexpression of a MAFB gene product and/or a myeloproliferative disorder associated with overexpression of a MAFB gene product. Such pharmaceutical compositions comprise an effective amount of at least one miR gene product and a pharmaceutically-acceptable carrier, wherein the at least one miR gene product binds to, and decreases expression of, the MAFB gene product. In another embodiment, the at least one miR gene product comprises a nucleotide sequence that is complementary to a nucleotide sequence in the MAFB gene product. In still another embodiment, the at least one miR gene product is miR-130a or a variant or biologically-active fragment thereof. In yet another embodiment, the pharmaceutical composition further comprises at least one anti-cancer agent.

In one embodiment, the invention is a pharmaceutical composition for treating a cancer associated with overexpression of a HOXA1 gene product and/or a myeloproliferative disorder associated with overexpression of a HOXA1 gene product. Such pharmaceutical compositions comprise an effective amount of at least one miR gene product and a pharmaceutically-acceptable carrier, wherein the at least one miR gene product binds to, and decreases expression of, the HOXA1 gene product. In another embodiment, the at least one miR gene product comprises a nucleotide sequence that is complementary to a nucleotide sequence in the HOXA1 gene product. In still another embodiment, the at least one miR gene product is miR-10a or a variant or biologically-active fragment thereof. In yet another embodiment, the pharmaceutical composition further comprises at least one anti-cancer agent.

Various objects and advantages of this invention will become apparent to those skilled in the art from the following detailed description of the preferred embodiment, when read in light of the accompanying drawings.

BRIEF DESCRIPTION OF THE DRAWINGS

The patent or application file contains at least one drawing executed in color. Copies of this patent or patent application publication with color drawing(s) will be provided by the Office upon request and payment of the necessary fee.

FIGS. 1A-1D depict Northern Blots and Real Time miRNA-PCR results, which validate microRNA chip data in CD34 progenitor differentiation experiments.

FIG. 1A depicts Northern Blots for miR-130a, miR-10a and miR-223. A loading RNA control was performed with U6.

FIG. 1B is a graph depicting RT-miRNA-PCR for miR-10a, miR-106, miR-126 and miR-130a. miRNA expression is presented as fold difference with respect to CD34+ cells before culture.

FIG. 1C is a graph depicting temporal expression of miR-223 by microarray.

FIG. 1D is a graph depicting temporal expression of miR-15-1 and miR-16-1 by RT-miRNA PCR.

FIGS. 2A-2C demonstrate that MAFB is a target of miR-130a.

FIG. 2A depicts MAFB mRNA and protein expression data in CD34+ progenitors induced to megakaryocytic differentiation. β-Actin was used for RT-PCR and Western blot loading controls.

FIG. 2B is a graph depicting relative repression of luciferase activity in MEG01 cells co-transfected with miR-10a and PGL3 3′UTR MAFB, miR-10a with PGL3 3′UTR, miR-10a seed match mutated and scramble with mutated, and wild type 3′UTR MAFB.

FIG. 2C depicts Western blots of MAFB total protein lysates in K562 cells transfected with miR-130a and scramble.

FIGS. 3A-3G demonstrate that MiR-10a downregulates HOXA1 by mediating RNA cleavage.

FIG. 3A is a graph depicting RT-PCR results for HOXA1 gene expression in differentiated megakaryocytes (Relative amount of transcript with respect to CD34+ progenitors at baseline).

FIG. 3B is a Western blot showing hoxal protein expression in differentiated megakaryocytes.

FIG. 3C is a graph depicting relative repression of luciferase activity of HOXA1 3′ UTR cloned PGL3 reporter plasmid when co-transfected with miR-10a and control scramble.

FIG. 3D is a schematic showing complementarity between miR-10a (SEQ ID NO: 322) and the HOXA1 3′UTR (SEQ ID NO: 507) as predicted by PICTAR.

FIG. 3E depicts RT-PCR results for miR-10a gene expression in scramble and miR-10a precursor transfected K562 cells.

FIG. 3F depicts RT-PCR results for HOXA1 gene expression in scramble and miR-10a precursor transfected K562 cells.

FIG. 3G is a Western blot showing HOXA1 expression in K562 cells transfected with control scramble and precursor miR-10a.

FIGS. 4A and 4B show phenotypic characterization results of in vitro-differentiated CD34+ progenitors.

FIG. 4A depicts May-Giemsa stains that were performed on cytospin preparations from CD34+ progenitors in culture at different days of culture (day 6, day 10, day 12 and day 14). At day 4, most of the cells were immature, as evidenced by the high nucleous:cytoplasmic ratio. Larger and multinuclear cells were observed by day 10. At day 14, predominantly larger, polyploid cells with long cytoplasmic processes and numerous membrane blebs with invaginations and vacuoles (original magnification 400×) were observed.

FIG. 4B depicts FACS analysis of CD34 in vitro-differentiated megakaryocytes. The membrane phenotype of CD34+ progenitor cells that are grown in culture is shown. Cells were harvested at days 10 (D+10), 14 (D+14) and 16 (D+16) and were analyzed by single fluorescent labeling using an anti-CD41 antibody, an anti-CD61a antibody, an anti-CD42a antibody and their respective isotype monoclonal antibodies (D+10 isotype; D+14 isotype; D+16 isotype). Double labeling was performed with anti-CD41a and CD61b monoclonal Abs at day 14 only.

FIG. 5 is a graph depicting RT-PCR expression results for miR-20 and miR-17 in differentiated megakaryocytes. The results are presented as fold difference with respect to CD34+ cells at baseline after normalization with 18S and delta Ct calculations.

FIG. 6A is a graph depicting temporal expression of miR-16-1 during megakaryocytic differentiation. The absolute expression value of miR-16-1 was determined by a per-chip median normalization.

FIG. 6B is a graph depicting temporal expression of miR-142 during megakaryocytic differentiation. The absolute expression value of miR-142 was determined by a per-chip median normalization.

FIG. 6C is a graph depicting temporal expression of miR-181b during megakaryocytic differentiation. The absolute expression value of miR-181b was determined by a per-chip median normalization.

FIG. 7 is a Northern Blot of total RNA obtained from K562 cells transfected with miR-130a precursor and scramble sequences hybridized with the probe for miR-130a. An RNA loading control was performed using U6 hybridization.

FIG. 8 is a schematic depicting microRNAs that are located in the HOXA, HOXB, HOXC and HOXD gene clusters.

FIG. 9A is a graph depicting HOXB4 gene expression in differentiated megakaryocytes. RT-PCR results for HOXB4 are shown as fold difference in the expression level with respect to CD34+ progenitors at baseline (before culture).

FIG. 9B is a graph depicting HOXB5 gene expression in differentiated megakaryocytes. RT-PCR results for HOXB5 are shown as fold difference in the expression levels with respect to CD34+ progenitors at baseline (before culture).

FIG. 10 is a graph depicting microRNA expression in acute megakaryoblastic cell lines by RT-PCR. Results are expressed as fold difference with respect to CD34-differentiated megakaryocytes after normalization with 18S and delta Ct calculations.

DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENT

The present invention is based, in part, on the identification of specific microRNAs (miRNAs) that are involved in megakaryocytic differentiation and/or have altered expression levels in cancerous cells (e.g., in acute megakaryoblastic leukemia (AMKL cell lines)). The invention is further based, in part, on association of these miRNAs with particular diagnostic, prognostic and therapeutic features. As described and exemplified herein:

i) particular miRNA are downregulated during megakaryocytic differentiation;

ii) the transcription factor MAFB is a target for miR-130a;

iii) miR-10a expression parallels that of HOXB gene expression;

iv) miR-10a downregulates HOXA1 expression; and

v) particular miRNA are upregulated in cancerous cells (e.g., acute megakaryoblastic leukemia (AMKL) cells).

As used herein interchangeably, a “miR gene product,” “microRNA,” “miR,” “miR” or “miRNA” refers to the unprocessed or processed RNA transcript from a miR gene. As the miR gene products are not translated into protein, the term “miR gene products” does not include proteins. The unprocessed miR gene transcript is also called a “miR precursor,” and typically comprises an RNA transcript of about 70-100 nucleotides in length. The miR precursor can be processed by digestion with an RNAse (for example, Dicer, Argonaut, RNAse III (e.g., E. coli RNAse III)) into an active 19-25 nucleotide RNA molecule. This active 19-25 nucleotide RNA molecule is also called the “processed” miR gene transcript or “mature” miRNA.

The active 19-25 nucleotide RNA molecule can be obtained from the miR precursor through natural processing routes (e.g., using intact cells or cell lysates) or by synthetic processing routes (e.g., using isolated processing enzymes, such as isolated Dicer, Argonaut, or RNAse III). It is understood that the active 19-25 nucleotide RNA molecule can also be produced directly by biological or chemical synthesis, without having to be processed from the miR precursor. When a microRNA is referred to herein by name, the name corresponds to both the precursor and mature forms, unless otherwise indicated.

Tables 1a and 1b depict the nucleotide sequences of particular precursor and mature human microRNAs.

TABLE 1a Human microRNA Precursor Sequences. Precursor Name Sequence (5′ To 3′)* SEQ ID NO. let-7a-1 CACUGUGGGAUGAGGUAGUAGGUUGUAUAGUUUUAGGG 1 UCACACCCACCACUGGGAGAUAACUAUACAAUCUACUGU CUUUCCUAACGUG let-7a-2 AGGUUGAGGUAGUAGGUUGUAUAGUUUAGAAUUACAUC 2 AAGGGAGAUAACUGUACAGCCUCCUAGCUUUCCU let-7a-3 GGGUGAGGUAGUAGGUUGUAUAGUUUGGGGCUCUGCCC 3 UGCUAUGGGAUAACUAUACAAUCUACUGUCUUUCCU let-7a-4 GUGACUGCAUGCUCCCAGGUUGAGGUAGUAGGUUGUAU 4 AGUUUAGAAUUACACAAGGGAGAUAACUGUACAGCCUC CUAGCUUUCCUUGGGUCUUGCACUAAACAAC let-7b GGCGGGGUGAGGUAGUAGGUUGUGUGGUUUCAGGGCAG 5 UGAUGUUGCCCCUCGGAAGAUAACUAUACAACCUACUGC CUUCCCUG let-7c GCAUCCGGGUUGAGGUAGUAGGUUGUAUGGUUUAGAGU 6 UACACCCUGGGAGUUAACUGUACAACCUUCUAGCUUUCC UUGGAGC let-7d CCUAGGAAGAGGUAGUAGGUUGCAUAGUUUUAGGGCAG 7 GGAUUUUGCCCACAAGGAGGUAACUAUACGACCUGCUGC CUUUCUUAGG let-7d-v1 CUAGGAAGAGGUAGUAGUUUGCAUAGUUUUAGGGCAAA 8 GAUUUUGCCCACAAGUAGUUAGCUAUACGACCUGCAGCC UUUUGUAG let-7d-v2 CUGGCUGAGGUAGUAGUUUGUGCUGUUGGUCGGGUUGU 9 GACAUUGCCCGCUGUGGAGAUAACUGCGCAAGCUACUGC CUUGCUAG let-7e CCCGGGCUGAGGUAGGAGGUUGUAUAGUUGAGGAGGAC 10 ACCCAAGGAGAUCACUAUACGGCCUCCUAGCUUUCCCCA GG let-7f-1 UCAGAGUGAGGUAGUAGAUUGUAUAGUUGUGGGGUAGU 11 GAUUUUACCCUGUUCAGGAGAUAACUAUACAAUCUAUU GCCUUCCCUGA let-7f-2-1 CUGUGGGAUGAGGUAGUAGAUUGUAUAGUUGUGGGGUA 12 GUGAUUUUACCCUGUUCAGGAGAUAACUAUACAAUCUA UUGCCUUCCCUGA let-7f-2-2 CUGUGGGAUGAGGUAGUAGAUUGUAUAGUUUUAGGGUC 13 AUACCCCAUCUUGGAGAUAACUAUACAGUCUACUGUCUU UCCCACGG let-7g UUGCCUGAUUCCAGGCUGAGGUAGUAGUUUGUACAGUU 14 UGAGGGUCUAUGAUACCACCCGGUACAGGAGAUAACUG UACAGGCCACUGCCUUGCCAGGAACAGCGCGC let-7i CUGGCUGAGGUAGUAGUUUGUGCUGUUGGUCGGGUUGU 15 GACAUUGCCCGCUGUGGAGAUAACUGCGCAAGCUACUGC CUUGCUAG miR-1b-1-1 ACCUACUCAGAGUACAUACUUCUUUAUGUACCCAUAUGA 16 ACAUACAAUGCUAUGGAAUGUAAAGAAGUAUGUAUUUU UGGUAGGC miR-1b-1-2 CAGCUAACAACUUAGUAAUACCUACUCAGAGUACAUACU 17 UCUUUAUGUACCCAUAUGAACAUACAAUGCUAUGGAAU GUAAAGAAGUAUGUAUUUUUGGUAGGCAAUA miR-1b-2 GCCUGCUUGGGAAACAUACUUCUUUAUAUGCCCAUAUG 18 GACCUGCUAAGCUAUGGAAUGUAAAGAAGUAUGUAUCU CAGGCCGGG miR-1b UGGGAAACAUACUUCUUUAUAUGCCCAUAUGGACCUGC 19 UAAGCUAUGGAAUGUAAAGAAGUAUGUAUCUCA miR-1d ACCUACUCAGAGUACAUACUUCUUUAUGUACCCAUAUGA 20 ACAUACAAUGCUAUGGAAUGUAAAGAAGUAUGUAUUUU UGGUAGGC miR-7-1a UGGAUGUUGGCCUAGUUCUGUGUGGAAGACUAGUGAUU 21 UUGUUGUUUUUAGAUAACUAAAUCGACAACAAAUCACA GUCUGCCAUAUGGCACAGGCCAUGCCUCUACA miR-7-1b UUGGAUGUUGGCCUAGUUCUGUGUGGAAGACUAGUGAU 22 UUUGUUGUUUUUAGAUAACUAAAUCGACAACAAAUCAC AGUCUGCCAUAUGGCACAGGCCAUGCCUCUACAG miR-7-2 CUGGAUACAGAGUGGACCGGCUGGCCCCAUCUGGAAGAC 23 UAGUGAUUUUGUUGUUGUCUUACUGCGCUCAACAACAA AUCCCAGUCUACCUAAUGGUGCCAGCCAUCGCA miR-7-3 AGAUUAGAGUGGCUGUGGUCUAGUGCUGUGUGGAAGAC 24 UAGUGAUUUUGUUGUUCUGAUGUACUACGACAACAAGU CACAGCCGGCCUCAUAGCGCAGACUCCCUUCGAC miR-9-1 CGGGGUUGGUUGUUAUCUUUGGUUAUCUAGCUGUAUGA 25 GUGGUGUGGAGUCUUCAUAAAGCUAGAUAACCGAAAGU AAAAAUAACCCCA miR-9-2 GGAAGCGAGUUGUUAUCUUUGGUUAUCUAGCUGUAUGA 26 GUGUAUUGGUCUUCAUAAAGCUAGAUAACCGAAAGUAA AAACUCCUUCA miR-9-3 GGAGGCCCGUUUCUCUCUUUGGUUAUCUAGCUGUAUGA 27 GUGCCACAGAGCCGUCAUAAAGCUAGAUAACCGAAAGU AGAAAUGAUUCUCA miR-10a GAUCUGUCUGUCUUCUGUAUAUACCCUGUAGAUCCGAA 28 UUUGUGUAAGGAAUUUUGUGGUCACAAAUUCGUAUCUA GGGGAAUAUGUAGUUGACAUAAACACUCCGCUCU miR-10b CCAGAGGUUGUAACGUUGUCUAUAUAUACCCUGUAGAA 29 CCGAAUUUGUGUGGUAUCCGUAUAGUCACAGAUUCGAU UCUAGGGGAAUAUAUGGUCGAUGCAAAAACUUCA miR-15a-2 GCGCGAAUGUGUGUUUAAAAAAAAUAAAACCUUGGAGU 30 AAAGUAGCAGCACAUAAUGGUUUGUGGAUUUUGAAAAG GUGCAGGCCAUAUUGUGCUGCCUCAAAAAUAC miR-15a CCUUGGAGUAAAGUAGCAGCACAUAAUGGUUUGUGGAU 31 UUUGAAAAGGUGCAGGCCAUAUUGUGCUGCCUCAAAAA UACAAGG miR-15b-1 CUGUAGCAGCACAUCAUGGUUUACAUGCUACAGUCAAG 32 AUGCGAAUCAUUAUUUGCUGCUCUAG miR-15b-2 UUGAGGCCUUAAAGUACUGUAGCAGCACAUCAUGGUUU 33 ACAUGCUACAGUCAAGAUGCGAAUCAUUAUUUGCUGCU CUAGAAAUUUAAGGAAAUUCAU miR-16-1 GUCAGCAGUGCCUUAGCAGCACGUAAAUAUUGGCGUUA 34 AGAUUCUAAAAUUAUCUCCAGUAUUAACUGUGCUGCUG AAGUAAGGUUGAC miR-16-2 GUUCCACUCUAGCAGCACGUAAAUAUUGGCGUAGUGAA 35 AUAUAUAUUAAACACCAAUAUUACUGUGCUGCUUUAGU GUGAC miR-16-13 GCAGUGCCUUAGCAGCACGUAAAUAUUGGCGUUAAGAU 36 UCUAAAAUUAUCUCCAGUAUUAACUGUGCUGCUGAAGU AAGGU miR-17 GUCAGAAUAAUGUCAAAGUGCUUACAGUGCAGGUAGUG 37 AUAUGUGCAUCUACUGCAGUGAAGGCACUUGUAGCAUU AUGGUGAC miR-18 UGUUCUAAGGUGCAUCUAGUGCAGAUAGUGAAGUAGAU 38 UAGCAUCUACUGCCCUAAGUGCUCCUUCUGGCA miR-18-13 UUUUUGUUCUAAGGUGCAUCUAGUGCAGAUAGUGAAGU 39 AGAUUAGCAUCUACUGCCCUAAGUGCUCCUUCUGGCAUA AGAA miR-19a GCAGUCCUCUGUUAGUUUUGCAUAGUUGCACUACAAGA 40 AGAAUGUAGUUGUGCAAAUCUAUGCAAAACUGAUGGUG GCCUGC miR-19a-13 CAGUCCUCUGUUAGUUUUGCAUAGUUGCACUACAAGAA 41 GAAUGUAGUUGUGCAAAUCUAUGCAAAACUGAUGGUGG CCUG miR-19b-1 CACUGUUCUAUGGUUAGUUUUGCAGGUUUGCAUCCAGC 42 UGUGUGAUAUUCUGCUGUGCAAAUCCAUGCAAAACUGA CUGUGGUAGUG miR-19b-2 ACAUUGCUACUUACAAUUAGUUUUGCAGGUUUGCAUUU 43 CAGCGUAUAUAUGUAUAUGUGGCUGUGCAAAUCCAUGC AAAACUGAUUGUGAUAAUGU miR-19b-13 UUCUAUGGUUAGUUUUGCAGGUUUGCAUCCAGCUGUGU 44 GAUAUUCUGCUGUGCAAAUCCAUGCAAAACUGACUGUG GUAG miR-19b-X UUACAAUUAGUUUUGCAGGUUUGCAUUUCAGCGUAUAU 45 AUGUAUAUGUGGCUGUGCAAAUCCAUGCAAAACUGAUU GUGAU miR-20 GUAGCACUAAAGUGCUUAUAGUGCAGGUAGUGUUUAGU 46 (miR-20a) UAUCUACUGCAUUAUGAGCACUUAAAGUACUGC miR-21 UGUCGGGUAGCUUAUCAGACUGAUGUUGACUGUUGAAU 47 CUCAUGGCAACACCAGUCGAUGGGCUGUCUGACA miR-21-17 ACCUUGUCGGGUAGCUUAUCAGACUGAUGUUGACUGUU 48 GAAUCUCAUGGCAACACCAGUCGAUGGGCUGUCUGACAU UUUG miR-22 GGCUGAGCCGCAGUAGUUCUUCAGUGGCAAGCUUUAUG 49 UCCUGACCCAGCUAAAGCUGCCAGUUGAAGAACUGUUGC CCUCUGCC miR-23a GGCCGGCUGGGGUUCCUGGGGAUGGGAUUUGCUUCCUG 50 UCACAAAUCACAUUGCCAGGGAUUUCCAACCGACC miR-23b CUCAGGUGCUCUGGCUGCUUGGGUUCCUGGCAUGCUGAU 51 UUGUGACUUAAGAUUAAAAUCACAUUGCCAGGGAUUAC CACGCAACCACGACCUUGGC miR-23-19 CCACGGCCGGCUGGGGUUCCUGGGGAUGGGAUUUGCUUC 52 CUGUCACAAAUCACAUUGCCAGGGAUUUCCAACCGACCC UGA miR-24-1 CUCCGGUGCCUACUGAGCUGAUAUCAGUUCUCAUUUUAC 53 ACACUGGCUCAGUUCAGCAGGAACAGGAG miR-24-2 CUCUGCCUCCCGUGCCUACUGAGCUGAAACACAGUUGGU 54 UUGUGUACACUGGCUCAGUUCAGCAGGAACAGGG miR-24-19 CCCUGGGCUCUGCCUCCCGUGCCUACUGAGCUGAAACAC 55 AGUUGGUUUGUGUACACUGGCUCAGUUCAGCAGGAACA GGGG miR-24-9 CCCUCCGGUGCCUACUGAGCUGAUAUCAGUUCUCAUUUU 56 ACACACUGGCUCAGUUCAGCAGGAACAGCAUC miR-25 GGCCAGUGUUGAGAGGCGGAGACUUGGGCAAUUGCUGG 57 ACGCUGCCCUGGGCAUUGCACUUGUCUCGGUCUGACAGU GCCGGCC miR-26a AGGCCGUGGCCUCGUUCAAGUAAUCCAGGAUAGGCUGU 58 GCAGGUCCCAAUGGCCUAUCUUGGUUACUUGCACGGGGA CGCGGGCCU miR-26a-1 GUGGCCUCGUUCAAGUAAUCCAGGAUAGGCUGUGCAGG 59 UCCCAAUGGGCCUAUUCUUGGUUACUUGCACGGGGACGC miR-26a-2 GGCUGUGGCUGGAUUCAAGUAAUCCAGGAUAGGCUGUU 60 UCCAUCUGUGAGGCCUAUUCUUGAUUACUUGUUUCUGG AGGCAGCU miR-26b CCGGGACCCAGUUCAAGUAAUUCAGGAUAGGUUGUGUG 61 CUGUCCAGCCUGUUCUCCAUUACUUGGCUCGGGGACCGG miR-27a CUGAGGAGCAGGGCUUAGCUGCUUGUGAGCAGGGUCCA 62 CACCAAGUCGUGUUCACAGUGGCUAAGUUCCGCCCCCCAG miR-27b-1 AGGUGCAGAGCUUAGCUGAUUGGUGAACAGUGAUUGGU 63 UUCCGCUUUGUUCACAGUGGCUAAGUUCUGCACCU miR-27b-2 ACCUCUCUAACAAGGUGCAGAGCUUAGCUGAUUGGUGA 64 ACAGUGAUUGGUUUCCGCUUUGUUCACAGUGGCUAAGU UCUGCACCUGAAGAGAAGGUG miR-27-19 CCUGAGGAGCAGGGCUUAGCUGCUUGUGAGCAGGGUCC 65 ACACCAAGUCGUGUUCACAGUGGCUAAGUUCCGCCCCCC AGG miR-28 GGUCCUUGCCCUCAAGGAGCUCACAGUCUAUUGAGUUAC 66 CUUUCUGACUUUCCCACUAGAUUGUGAGCUCCUGGAGGG CAGGCACU miR-29a-2 CCUUCUGUGACCCCUUAGAGGAUGACUGAUUUCUUUUG 67 GUGUUCAGAGUCAAUAUAAUUUUCUAGCACCAUCUGAA AUCGGUUAUAAUGAUUGGGGAAGAGCACCAUG miR-29a AUGACUGAUUUCUUUUGGUGUUCAGAGUCAAUAUAAUU 68 UUCUAGCACCAUCUGAAAUCGGUUAU miR-29b-1 CUUCAGGAAGCUGGUUUCAUAUGGUGGUUUAGAUUUAA 69 AUAGUGAUUGUCUAGCACCAUUUGAAAUCAGUGUUCUU GGGGG miR-29b-2 CUUCUGGAAGCUGGUUUCACAUGGUGGCUUAGAUUUUU 70 CCAUCUUUGUAUCUAGCACCAUUUGAAAUCAGUGUUUU AGGAG miR-29c ACCACUGGCCCAUCUCUUACACAGGCUGACCGAUUUCUC 71 CUGGUGUUCAGAGUCUGUUUUUGUCUAGCACCAUUUGA AAUCGGUUAUGAUGUAGGGGGAAAAGCAGCAGC miR-30a GCGACUGUAAACAUCCUCGACUGGAAGCUGUGAAGCCAC 72 AGAUGGGCUUUCAGUCGGAUGUUUGCAGCUGC miR-30b-1 AUGUAAACAUCCUACACUCAGCUGUAAUACAUGGAUUG 73 GCUGGGAGGUGGAUGUUUACGU miR-30b-2 ACCAAGUUUCAGUUCAUGUAAACAUCCUACACUCAGCUG 74 UAAUACAUGGAUUGGCUGGGAGGUGGAUGUUUACUUCA GCUGACUUGGA miR-30c AGAUACUGUAAACAUCCUACACUCUCAGCUGUGGAAAG 75 UAAGAAAGCUGGGAGAAGGCUGUUUACUCUUUCU miR-30d GUUGUUGUAAACAUCCCCGACUGGAAGCUGUAAGACAC 76 AGCUAAGCUUUCAGUCAGAUGUUUGCUGCUAC miR-30e CUGUAAACAUCCUUGACUGGAAGCUGUAAGGUGUUCAG 77 AGGAGCUUUCAGUCGGAUGUUUACAG miR-31 GGAGAGGAGGCAAGAUGCUGGCAUAGCUGUUGAACUGG 78 GAACCUGCUAUGCCAACAUAUUGCCAUCUUUCC miR-32 GGAGAUAUUGCACAUUACUAAGUUGCAUGUUGUCACGG 79 CCUCAAUGCAAUUUAGUGUGUGUGAUAUUUUC miR-33b GGGGGCCGAGAGAGGCGGGCGGCCCCGCGGUGCAUUGCU 80 GUUGCAUUGCACGUGUGUGAGGCGGGUGCAGUGCCUCG GCAGUGCAGCCCGGAGCCGGCCCCUGGCACCAC miR-33b-2 ACCAAGUUUCAGUUCAUGUAAACAUCCUACACUCAGCUG 81 UAAUACAUGGAUUGGCUGGGAGGUGGAUGUUUACUUC A GCUGACUUGGA miR-33 CUGUGGUGCAUUGUAGUUGCAUUGCAUGUUCUGGUGGU 82 ACCCAUGCAAUGUUUCCACAGUGCAUCACAG miR-34-a GGCCAGCUGUGAGUGUUUCUUUGGCAGUGUCUUAGCUG 83 GUUGUUGUGAGCAAUAGUAAGGAAGCAAUCAGCAAGUA UACUGCCCUAGAAGUGCUGCACGUUGUGGGGCCC miR-34-b GUGCUCGGUUUGUAGGCAGUGUCAUUAGCUGAUUGUAC 84 UGUGGUGGUUACAAUCACUAACUCCACUGCCAUCAAAAC AAGGCAC miR-34-c AGUCUAGUUACUAGGCAGUGUAGUUAGCUGAUUGCUAA 85 UAGUACCAAUCACUAACCACACGGCCAGGUAAAAAGAUU miR-91-13 UCAGAAUAAUGUCAAAGUGCUUACAGUGCAGGUAGUGA 86 UAUGUGCAUCUACUGCAGUGAAGGCACUUGUAGCAUUA UGGUGA miR-92-1 CUUUCUACACAGGUUGGGAUCGGUUGCAAUGCUGUGUU 87 UCUGUAUGGUAUUGCACUUGUCCCGGCCUGUUGAGUUU GG miR-92-2 UCAUCCCUGGGUGGGGAUUUGUUGCAUUACUUGUGUUC 88 UAUAUAAAGUAUUGCACUUGUCCCGGCCUGUGGAAGA miR-93-1 CUGGGGGCUCCAAAGUGCUGUUCGUGCAGGUAGUGUGA 89 (miR-93-2) UUACCCAACCUACUGCUGAGCUAGCACUUCCCGAGCCCC CGG miR-95-4 AACACAGUGGGCACUCAAUAAAUGUCUGUUGAAUUGAA 90 AUGCGUUACAUUCAACGGGUAUUUAUUGAGCACCCACUC UGUG miR-96-7 UGGCCGAUUUUGGCACUAGCACAUUUUUGCUUGUGUCU 91 CUCCGCUCUGAGCAAUCAUGUGCAGUGCCAAUAUGGGAAA miR-97-6 GUGAGCGACUGUAAACAUCCUCGACUGGAAGCUGUGAA 92 (miR-30*) GCCACAGAUGGGCUUUCAGUCGGAUGUUUGCAGCUGCCU ACU miR-98 GUGAGGUAGUAAGUUGUAUUGUUGUGGGGUAGGGAUAU 93 UAGGCCCCAAUUAGAAGAUAACUAUACAACUUACUACU UUCC miR-99b GGCACCCACCCGUAGAACCGACCUUGCGGGGCCUUCGCC 94 GCACACAAGCUCGUGUCUGUGGGUCCGUGUC miR-99a CCCAUUGGCAUAAACCCGUAGAUCCGAUCUUGUGGUGAA 95 GUGGACCGCACAAGCUCGCUUCUAUGGGUCUGUGUCAGU GUG miR-100-1/2 AAGAGAGAAGAUAUUGAGGCCUGUUGCCACAAACCCGU 96 AGAUCCGAACUUGUGGUAUUAGUCCGCACAAGCUUGUA UCUAUAGGUAUGUGUCUGUUAGGCAAUCUCAC miR-100-11 CCUGUUGCCACAAACCCGUAGAUCCGAACUUGUGGUAUU 97 AGUCCGCACAAGCUUGUAUCUAUAGGUAUGUGUCUGUU AGG miR-101-1/2 AGGCUGCCCUGGCUCAGUUAUCACAGUGCUGAUGCUGUC 98 UAUUCUAAAGGUACAGUACUGUGAUAACUGAAGGAUGG CAGCCAUCUUACCUUCCAUCAGAGGAGCCUCAC miR-101 UCAGUUAUCACAGUGCUGAUGCUGUCCAUUCUAAAGGU 99 ACAGUACUGUGAUAACUGA miR-101-1 UGCCCUGGCUCAGUUAUCACAGUGCUGAUGCUGUCUAUU 100 CUAAAGGUACAGUACUGUGAUAACUGAAGGAUGGCA miR-101-2 ACUGUCCUUUUUCGGUUAUCAUGGUACCGAUGCUGUAU 101 AUCUGAAAGGUACAGUACUGUGAUAACUGAAGAAUGGU GGU miR-101-9 UGUCCUUUUUCGGUUAUCAUGGUACCGAUGCUGUAUAU 102 CUGAAAGGUACAGUACUGUGAUAACUGAAGAAUGGUG miR-102-1 CUUCUGGAAGCUGGUUUCACAUGGUGGCUUAGAUUUUU 103 CCAUCUUUGUAUCUAGCACCAUUUGAAAUCAGUGUUUU AGGAG miR-102-7.1 CUUCAGGAAGCUGGUUUCAUAUGGUGGUUUAGAUUUAA 104 (miR-102-7.2) AUAGUGAUUGUCUAGCACCAUUUGAAAUCAGUGUUCUU GGGGG miR-103-2 UUGUGCUUUCAGCUUCUUUACAGUGCUGCCUUGUAGCA 105 UUCAGGUCAAGCAACAUUGUACAGGGCUAUGAAAGAAC CA miR-103-1 UACUGCCCUCGGCUUCUUUACAGUGCUGCCUUGUUGCAU 106 AUGGAUCAAGCAGCAUUGUACAGGGCUAUGAAGGCAUUG miR-104-17 AAAUGUCAGACAGCCCAUCGACUGGUGUUGCCAUGAGA 107 UUCAACAGUCAACAUCAGUCUGAUAAGCUACCCGACAAGG miR-105-1 UGUGCAUCGUGGUCAAAUGCUCAGACUCCUGUGGUGGC 108 UGCUCAUGCACCACGGAUGUUUGAGCAUGUGCUACGGU GUCUA miR-105-2 UGUGCAUCGUGGUCAAAUGCUCAGACUCCUGUGGUGGC 109 UGCUUAUGCACCACGGAUGUUUGAGCAUGUGCUAUGGU GUCUA miR-106-a CCUUGGCCAUGUAAAAGUGCUUACAGUGCAGGUAGCUU 110 UUUGAGAUCUACUGCAAUGUAAGCACUUCUUACAUUAC CAUGG miR-106-b CCUGCCGGGGCUAAAGUGCUGACAGUGCAGAUAGUGGU 111 CCUCUCCGUGCUACCGCACUGUGGGUACUUGCUGCUCCA GCAGG miR-107 CUCUCUGCUUUCAGCUUCUUUACAGUGUUGCCUUGUGGC 112 AUGGAGUUCAAGCAGCAUUGUACAGGGCUAUCAAAGCA CAGA MIR-108-1- ACACUGCAAGAACAAUAAGGAUUUUUAGGGGCAUUAUG 113 SMALL ACUGAGUCAGAAAACACAGCUGCCCCUGAAAGUCCCUCA UUUUUCUUGCUGU MIR-108-2- ACUGCAAGAGCAAUAAGGAUUUUUAGGGGCAUUAUGAU 114 SMALL AGUGGAAUGGAAACACAUCUGCCCCCAAAAGUCCCUCAU UUU miR-122a-1 CCUUAGCAGAGCUGUGGAGUGUGACAAUGGUGUUUGUG 115 UCUAAACUAUCAAACGCCAUUAUCACACUAAAUAGCUAC UGCUAGGC miR-122a-2 AGCUGUGGAGUGUGACAAUGGUGUUUGUGUCCAAACUA 116 UCAAACGCCAUUAUCACACUAAAUAGCU miR-123 ACAUUAUUACUUUUGGUACGCGCUGUGACACUUCAAAC 117 UCGUACCGUGAGUAAUAAUGCGC miR-124a-1 AGGCCUCUCUCUCCGUGUUCACAGCGGACCUUGAUUUAA 118 AUGUCCAUACAAUUAAGGCACGCGGUGAAUGCCAAGAA UGGGGCUG miR-124a-2 AUCAAGAUUAGAGGCUCUGCUCUCCGUGUUCACAGCGGA 119 CCUUGAUUUAAUGUCAUACAAUUAAGGCACGCGGUGAA UGCCAAGAGCGGAGCCUACGGCUGCACUUGAAG miR-124a-3 UGAGGGCCCCUCUGCGUGUUCACAGCGGACCUUGAUUUA 120 AUGUCUAUACAAUUAAGGCACGCGGUGAAUGCCAAGAG AGGCGCCUCC miR-124a CUCUGCGUGUUCACAGCGGACCUUGAUUUAAUGUCUAU 121 ACAAUUAAGGCACGCGGUGAAUGCCAAGAG miR-124b CUCUCCGUGUUCACAGCGGACCUUGAUUUAAUGUCAUAC 122 AAUUAAGGCACGCGGUGAAUGCCAAGAG miR-125a-1 UGCCAGUCUCUAGGUCCCUGAGACCCUUUAACCUGUGAG 123 GACAUCCAGGGUCACAGGUGAGGUUCUUGGGAGCCUGG CGUCUGGCC miR-125a-2 GGUCCCUGAGACCCUUUAACCUGUGAGGACAUCCAGGGU 124 CACAGGUGAGGUUCUUGGGAGCCUGG miR-125b-1 UGCGCUCCUCUCAGUCCCUGAGACCCUAACUUGUGAUGU 125 UUACCGUUUAAAUCCACGGGUUAGGCUCUUGGGAGCUG CGAGUCGUGCU miR-125b-2 ACCAGACUUUUCCUAGUCCCUGAGACCCUAACUUGUGAG 126 GUAUUUUAGUAACAUCACAAGUCAGGCUCUUGGGACCU AGGCGGAGGGGA miR-126-1 CGCUGGCGACGGGACAUUAUUACUUUUGGUACGCGCUG 127 UGACACUUCAAACUCGUACCGUGAGUAAUAAUGCGCCGU CCACGGCA miR-126-2 ACAUUAUUACUUUUGGUACGCGCUGUGACACUUCAAAC 128 UCGUACCGUGAGUAAUAAUGCGC miR-127-1 UGUGAUCACUGUCUCCAGCCUGCUGAAGCUCAGAGGGCU 129 CUGAUUCAGAAAGAUCAUCGGAUCCGUCUGAGCUUGGC UGGUCGGAAGUCUCAUCAUC miR-127-2 CCAGCCUGCUGAAGCUCAGAGGGCUCUGAUUCAGAAAGA 130 UCAUCGGAUCCGUCUGAGCUUGGCUGGUCGG miR-128a UGAGCUGUUGGAUUCGGGGCCGUAGCACUGUCUGAGAG 131 GUUUACAUUUCUCACAGUGAACCGGUCUCUUUUUCAGCU GCUUC miR-128b GCCCGGCAGCCACUGUGCAGUGGGAAGGGGGGCCGAUAC 132 ACUGUACGAGAGUGAGUAGCAGGUCUCACAGUGAACCG GUCUCUUUCCCUACUGUGUCACACUCCUAAUGG miR-128 GUUGGAUUCGGGGCCGUAGCACUGUCUGAGAGGUUUAC 133 AUUUCUCACAGUGAACCGGUCUCUUUUUCAGC miR-129-1 UGGAUCUUUUUGCGGUCUGGGCUUGCUGUUCCUCUCAAC 134 AGUAGUCAGGAAGCCCUUACCCCAAAAAGUAUCUA MIR-129-2 UGCCCUUCGCGAAUCUUUUUGCGGUCUGGGCUUGCUGUA 135 CAUAACUCAAUAGCCGGAAGCCCUUACCCCAAAAAGCAU UUGCGGAGGGCG miR-130a UGCUGCUGGCCAGAGCUCUUUUCACAUUGUGCUACUGUC 136 UGCACCUGUCACUAGCAGUGCAAUGUUAAAAGGGCAUU GGCCGUGUAGUG miR-131-1 GCCAGGAGGCGGGGUUGGUUGUUAUCUUUGGUUAUCUA 137 GCUGUAUGAGUGGUGUGGAGUCUUCAUAAAGCUAGAUA ACCGAAAGUAAAAAUAACCCCAUACACUGCGCAG miR-131-3 CACGGCGCGGCAGCGGCACUGGCUAAGGGAGGCCCGUUU 138 CUCUCUUUGGUUAUCUAGCUGUAUGAGUGCCACAGAGCC GUCAUAAAGCUAGAUAACCGAAAGUAGAAAUG miR-131 GUUGUUAUCUUUGGUUAUCUAGCUGUAUGAGUGUAUUG 139 GUCUUCAUAAAGCUAGAUAACCGAAAGUAAAAAC miR-132-1 CCGCCCCCGCGUCUCCAGGGCAACCGUGGCUUUCGAUUG 140 UUACUGUGGGAACUGGAGGUAACAGUCUACAGCCAUGG UCGCCCCGCAGCACGCCCACGCGC miR-132-2 GGGCAACCGUGGCUUUCGAUUGUUACUGUGGGAACUGG 141 AGGUAACAGUCUACAGCCAUGGUCGCCC miR-133a-1 ACAAUGCUUUGCUAGAGCUGGUAAAAUGGAACCAAAUC 142 GCCUCUUCAAUGGAUUUGGUCCCCUUCAACCAGCUGUAG CUAUGCAUUGA miR-133a-2 GGGAGCCAAAUGCUUUGCUAGAGCUGGUAAAAUGGAAC 143 CAAAUCGACUGUCCAAUGGAUUUGGUCCCCUUCAACCAG CUGUAGCUGUGCAUUGAUGGCGCCG miR-133 GCUAGAGCUGGUAAAAUGGAACCAAAUCGCCUCUUCAA 144 UGGAUUUGGUCCCCUUCAACCAGCUGUAGC miR-133b CCUCAGAAGAAAGAUGCCCCCUGCUCUGGCUGGUCAAAC 145 GGAACCAAGUCCGUCUUCCUGAGAGGUUUGGUCCCCUUC AACCAGCUACAGCAGGGCUGGCAAUGCCCAGUCCUUGGA GA MIR-133B- GCCCCCUGCUCUGGCUGGUCAAACGGAACCAAGUCCGUC 146 SMALL UUCCUGAGAGGUUUGGUCCCCUUCAACCAGCUACAGCAG GG miR-134-1 CAGGGUGUGUGACUGGUUGACCAGAGGGGCAUGCACUG 147 UGUUCACCCUGUGGGCCACCUAGUCACCAACCCUC miR-134-2 AGGGUGUGUGACUGGUUGACCAGAGGGGCAUGCACUGU 148 GUUCACCCUGUGGGCCACCUAGUCACCAACCCU miR-135a-1 AGGCCUCGCUGUUCUCUAUGGCUUUUUAUUCCUAUGUG 149 AUUCUACUGCUCACUCAUAUAGGGAUUGGAGCCGUGGC GCACGGCGGGGACA miR-135a-2 AGAUAAAUUCACUCUAGUGCUUUAUGGCUUUUUAUUCC 150 (miR-135-2) UAUGUGAUAGUAAUAAAGUCUCAUGUAGGGAUGGAAGC CAUGAAAUACAUUGUGAAAAAUCA miR-135 CUAUGGCUUUUUAUUCCUAUGUGAUUCUACUGCUCACUC 151 AUAUAGGGAUUGGAGCCGUGG miR-135b CACUCUGCUGUGGCCUAUGGCUUUUCAUUCCUAUGUGAU 152 UGCUGUCCCAAACUCAUGUAGGGCUAAAAGCCAUGGGCU ACAGUGAGGGGCGAGCUCC miR-136-1 UGAGCCCUCGGAGGACUCCAUUUGUUUUGAUGAUGGAU 153 UCUUAUGCUCCAUCAUCGUCUCAAAUGAGUCUUCAGAGG GUUCU miR-136-2 GAGGACUCCAUUUGUUUUGAUGAUGGAUUCUUAUGCUC 154 CAUCAUCGUCUCAAAUGAGUCUUC miR-137 CUUCGGUGACGGGUAUUCUUGGGUGGAUAAUACGGAUU 155 ACGUUGUUAUUGCUUAAGAAUACGCGUAGUCGAGG miR-138-1 CCCUGGCAUGGUGUGGUGGGGCAGCUGGUGUUGUGAAU 156 CAGGCCGUUGCCAAUCAGAGAACGGCUACUUCACAACAC CAGGGCCACACCACACUACAGG miR-138-2 CGUUGCUGCAGCUGGUGUUGUGAAUCAGGCCGACGAGC 157 AGCGCAUCCUCUUACCCGGCUAUUUCACGACACCAGGGU UGCAUCA miR-138 CAGCUGGUGUUGUGAAUCAGGCCGACGAGCAGCGCAUCC 158 UCUUACCCGGCUAUUUCACGACACCAGGGUUG miR-139 GUGUAUUCUACAGUGCACGUGUCUCCAGUGUGGCUCGG 159 AGGCUGGAGACGCGGCCCUGUUGGAGUAAC miR-140 UGUGUCUCUCUCUGUGUCCUGCCAGUGGUUUUACCCUAU 160 GGUAGGUUACGUCAUGCUGUUCUACCACAGGGUAGAAC CACGGACAGGAUACCGGGGCACC miR-140as UCCUGCCAGUGGUUUUACCCUAUGGUAGGUUACGUCAU 161 GCUGUUCUACCACAGGGUAGAACCACGGACAGGA miR-140s CCUGCCAGUGGUUUUACCCUAUGGUAGGUUACGUCAUGC 162 UGUUCUACCACAGGGUAGAACCACGGACAGG miR-141-1 CGGCCGGCCCUGGGUCCAUCUUCCAGUACAGUGUUGGAU 163 GGUCUAAUUGUGAAGCUCCUAACACUGUCUGGUAAAGA UGGCUCCCGGGUGGGUUC miR-141-2 GGGUCCAUCUUCCAGUACAGUGUUGGAUGGUCUAAUUG 164 UGAAGCUCCUAACACUGUCUGGUAAAGAUGGCCC miR-142 ACCCAUAAAGUAGAAAGCACUACUAACAGCACUGGAGG 165 GUGUAGUGUUUCCUACUUUAUGGAUG miR-143-1 GCGCAGCGCCCUGUCUCCCAGCCUGAGGUGCAGUGCUGC 166 AUCUCUGGUCAGUUGGGAGUCUGAGAUGAAGCACUGUA GCUCAGGAAGAGAGAAGUUGUUCUGCAGC miR-143-2 CCUGAGGUGCAGUGCUGCAUCUCUGGUCAGUUGGGAGU 167 CUGAGAUGAAGCACUGUAGCUCAGG miR-144-1 UGGGGCCCUGGCUGGGAUAUCAUCAUAUACUGUAAGUU 168 UGCGAUGAGACACUACAGUAUAGAUGAUGUACUAGUCC GGGCACCCCC miR-144-2 GGCUGGGAUAUCAUCAUAUACUGUAAGUUUGCGAUGAG 169 ACACUACAGUAUAGAUGAUGUACUAGUC miR-145-1 CACCUUGUCCUCACGGUCCAGUUUUCCCAGGAAUCCCUU 170 AGAUGCUAAGAUGGGGAUUCCUGGAAAUACUGUUCUUG AGGUCAUGGUU miR-145-2 CUCACGGUCCAGUUUUCCCAGGAAUCCCUUAGAUGCUAA 171 GAUGGGGAUUCCUGGAAAUACUGUUCUUGAG miR-146-1 CCGAUGUGUAUCCUCAGCUUUGAGAACUGAAUUCCAUG 172 GGUUGUGUCAGUGUCAGACCUCUGAAAUUCAGUUCUUC AGCUGGGAUAUCUCUGUCAUCGU miR-146-2 AGCUUUGAGAACUGAAUUCCAUGGGUUGUGUCAGUGUC 173 AGACCUGUGAAAUUCAGUUCUUCAGCU miR-147 AAUCUAAAGACAACAUUUCUGCACACACACCAGACUAUG 174 GAAGCCAGUGUGUGGAAAUGCUUCUGCUAGAUU miR-148a GAGGCAAAGUUCUGAGACACUCCGACUCUGAGUAUGAU 175 (miR-148) AGAAGUCAGUGCACUACAGAACUUUGUCUC miR-148b CAAGCACGAUUAGCAUUUGAGGUGAAGUUCUGUUAUAC 176 ACUCAGGCUGUGGCUCUCUGAAAGUCAGUGCAUCACAGA ACUUUGUCUCGAAAGCUUUCUA MIR-148B- AAGCACGAUUAGCAUUUGAGGUGAAGUUCUGUUAUACA 177 SMALL CUCAGGCUGUGGCUCUCUGAAAGUCAGUGCAU miR-149-1 GCCGGCGCCCGAGCUCUGGCUCCGUGUCUUCACUCCCGU 178 GCUUGUCCGAGGAGGGAGGGAGGGACGGGGGCUGUGCU GGGGCAGCUGGA miR-149-2 GCUCUGGCUCCGUGUCUUCACUCCCGUGCUUGUCCGAGG 179 AGGGAGGGAGGGAC miR-150-1 CUCCCCAUGGCCCUGUCUCCCAACCCUUGUACCAGUGCU 180 GGGCUCAGACCCUGGUACAGGCCUGGGGGACAGGGACCU GGGGAC miR-150-2 CCCUGUCUCCCAACCCUUGUACCAGUGCUGGGCUCAGAC 181 CCUGGUACAGGCCUGGGGGACAGGG miR-151 UUUCCUGCCCUCGAGGAGCUCACAGUCUAGUAUGUCUCA 182 UCCCCUACUAGACUGAAGCUCCUUGAGGACAGG MIR-151-2 CCUGUCCUCAAGGAGCUUCAGUCUAGUAGGGGAUGAGA 183 CAUACUAGACUGUGAGCUCCUCGAGGGCAGG miR-152-1 UGUCCCCCCCGGCCCAGGUUCUGUGAUACACUCCGACUC 184 GGGCUCUGGAGCAGUCAGUGCAUGACAGAACUUGGGCCC GGAAGGACC miR-152-2 GGCCCAGGUUCUGUGAUACACUCCGACUCGGGCUCUGGA 185 GCAGUCAGUGCAUGACAGAACUUGGGCCCCGG miR-153-1-1 CUCACAGCUGCCAGUGUCAUUUUUGUGAUCUGCAGCUAG 186 UAUUCUCACUCCAGUUGCAUAGUCACAAAAGUGAUCAU UGGCAGGUGUGGC miR-153-1-2 UCUCUCUCUCCCUCACAGCUGCCAGUGUCAUUGUCACAA 187 AAGUGAUCAUUGGCAGGUGUGGCUGCUGCAUG miR-153-2-1 AGCGGUGGCCAGUGUCAUUUUUGUGAUGUUGCAGCUAG 188 UAAUAUGAGCCCAGUUGCAUAGUCACAAAAGUGAUCAU UGGAAACUGUG miR-153-2-2 CAGUGUCAUUUUUGUGAUGUUGCAGCUAGUAAUAUGAG 189 CCCAGUUGCAUAGUCACAAAAGUGAUCAUUG miR-154-1 GUGGUACUUGAAGAUAGGUUAUCCGUGUUGCCUUCGCU 190 UUAUUUGUGACGAAUCAUACACGGUUGACCUAUUUUUC AGUACCAA miR-154-2 GAAGAUAGGUUAUCCGUGUUGCCUUCGCUUUAUUUGUG 191 ACGAAUCAUACACGGUUGACCUAUUUUU miR-155 CUGUUAAUGCUAAUCGUGAUAGGGGUUUUUGCCUCCAA 192 CUGACUCCUACAUAUUAGCAUUAACAG MIR-156 = CCUAACACUGUCUGGUAAAGAUGGCUCCCGGGUGGGUUC 193 MIR- UCUCGGCAGUAACCUUCAGGGAGCCCUGAAGACCAUGGA 157 = OVERLA GGAC P MIR-141 MIR-158- GCCGAGACCGAGUGCACAGGGCUCUGACCUAUGAAUUGA 194 SMALL = CAGCCAGUGCUCUCGUCUCCCCUCUGGCUGCCAAUUCCA MIR-192 UAGGUCACAGGUAUGUUCGCCUCAAUGCCAGC MIR-159-1- UCCCGCCCCCUGUAACAGCAACUCCAUGUGGAAGUGCCC 195 SMALL ACUGGUUCCAGUGGGGCUGCUGUUAUCUGGGGCGAGGG CCA MIR-161- AAAGCUGGGUUGAGAGGGCGAAAAAGGAUGAGGUGACU 196 SMALL GGUCUGGGCUACGCUAUGCUGCGGCGCUCGGG MIR-163-1B- CAUUGGCCUCCUAAGCCAGGGAUUGUGGGUUCGAGUCCC 197 SMALL ACCCGGGGUAAAGAAAGGCCGAAUU MIR-163-3- CCUAAGCCAGGGAUUGUGGGUUCGAGUCCCACCUGGGGU 198 SMALL AGAGGUGAAAGUUCCUUUUACGGAAUUUUUU miR-162 CAAUGUCAGCAGUGCCUUAGCAGCACGUAAAUAUUGGC 199 GUUAAGAUUCUAAAAUUAUCUCCAGUAUUAACUGUGCU GCUGAAGUAAGGUUGACCAUACUCUACAGUUG MIR-175- GGGCUUUCAAGUCACUAGUGGUUCCGUUUAGUAGAUGA 200 SMALL = MIR- UUGUGCAUUGUUUCAAAAUGGUGCCCUAGUGACUACAA 224 AGCCC MIR-177- ACGCAAGUGUCCUAAGGUGAGCUCAGGGAGCACAGAAA 201 SMALL CCUCCAGUGGAACAGAAGGGCAAAAGCUCAUU MIR-180- CAUGUGUCACUUUCAGGUGGAGUUUCAAGAGUCCCUUCC 202 SMALL UGGUUCACCGUCUCCUUUGCUCUUCCACAAC miR-181a AGAAGGGCUAUCAGGCCAGCCUUCAGAGGACUCCAAGGA 203 ACAUUCAACGCUGUCGGUGAGUUUGGGAUUUGAAAAAA CCACUGACCGUUGACUGUACCUUGGGGUCCUUA miR-181b-1 CCUGUGCAGAGAUUAUUUUUUAAAAGGUCACAAUCAAC 204 AUUCAUUGCUGUCGGUGGGUUGAACUGUGUGGACAAGC UCACUGAACAAUGAAUGCAACUGUGGCCCCGCUU miR-181b-2 CUGAUGGCUGCACUCAACAUUCAUUGCUGUCGGUGGGU 205 UUGAGUCUGAAUCAACUCACUGAUCAAUGAAUGCAAAC UGCGGACCAAACA miR-181c CGGAAAAUUUGCCAAGGGUUUGGGGGAACAUUCAACCU 206 GUCGGUGAGUUUGGGCAGCUCAGGCAAACCAUCGACCGU UGAGUGGACCCUGAGGCCUGGAAUUGCCAUCCU miR-182-as GAGCUGCUUGCCUCCCCCCGUUUUUGGCAAUGGUAGAAC 207 UCACACUGGUGAGGUAACAGGAUCCGGUGGUUCUAGAC UUGCCAACUAUGGGGCGAGGACUCAGCCGGCAC miR-182 UUUUUGGCAAUGGUAGAACUCACACUGGUGAGGUAACA 208 GGAUCCGGUGGUUCUAGACUUGCCAACUAUGG miR-183 CCGCAGAGUGUGACUCCUGUUCUGUGUAUGGCACUGGU 209 AGAAUUCACUGUGAACAGUCUCAGUCAGUGAAUUACCG AAGGGCCAUAAACAGAGCAGAGACAGAUCCACGA miR-184-1 CCAGUCACGUCCCCUUAUCACUUUUCCAGCCCAGCUUUG 210 UGACUGUAAGUGUUGGACGGAGAACUGAUAAGGGUAGG UGAUUGA miR-184-2 CCUUAUCACUUUUCCAGCCCAGCUUUGUGACUGUAAGUG 211 UUGGACGGAGAACUGAUAAGGGUAGG miR-185-1 AGGGGGCGAGGGAUUGGAGAGAAAGGCAGUUCCUGAUG 212 GUCCCCUCCCCAGGGGCUGGCUUUCCUCUGGUCCUUCCC UCCCA miR-185-2 AGGGAUUGGAGAGAAAGGCAGUUCCUGAUGGUCCCCUC 213 CCCAGGGGCUGGCUUUCCUCUGGUCCUU miR-186-1 UGCUUGUAACUUUCCAAAGAAUUCUCCUUUUGGGCUUU 214 CUGGUUUUAUUUUAAGCCCAAAGGUGAAUUUUUUGGGA AGUUUGAGCU miR-186-2 ACUUUCCAAAGAAUUCUCCUUUUGGGCUUUCUGGUUUU 215 AUUUUAAGCCCAAAGGUGAAUUUUUUGGGAAGU miR-187 GGUCGGGCUCACCAUGACACAGUGUGAGACUCGGGCUAC 216 AACACAGGACCCGGGGCGCUGCUCUGACCCCUCGUGUCU UGUGUUGCAGCCGGAGGGACGCAGGUCCGCA miR-188-1 UGCUCCCUCUCUCACAUCCCUUGCAUGGUGGAGGGUGAG 217 CUUUCUGAAAACCCCUCCCACAUGCAGGGUUUGCAGGAU GGCGAGCC miR-188-2 UCUCACAUCCCUUGCAUGGUGGAGGGUGAGCUUUCUGA 218 AAACCCCUCCCACAUGCAGGGUUUGCAGGA miR-189-1 CUGUCGAUUGGACCCGCCCUCCGGUGCCUACUGAGCUGA 219 UAUCAGUUCUCAUUUUACACACUGGCUCAGUUCAGCAGG AACAGGAGUCGAGCCCUUGAGCAA miR-189-2 CUCCGGUGCCUACUGAGCUGAUAUCAGUUCUCAUUUUAC 220 ACACUGGCUCAGUUCAGCAGGAACAGGAG miR-190-1 UGCAGGCCUCUGUGUGAUAUGUUUGAUAUAUUAGGUUG 221 UUAUUUAAUCCAACUAUAUAUCAAACAUAUUCCUACAG UGUCUUGCC miR-190-2 CUGUGUGAUAUGUUUGAUAUAUUAGGUUGUUAUUUAAU 222 CCAACUAUAUAUCAAACAUAUUCCUACAG miR-191-1 CGGCUGGACAGCGGGCAACGGAAUCCCAAAAGCAGCUGU 223 UGUCUCCAGAGCAUUCCAGCUGCGCUUGGAUUUCGUCCC CUGCUCUCCUGCCU miR-191-2 AGCGGGCAACGGAAUCCCAAAAGCAGCUGUUGUCUCCAG 224 AGCAUUCCAGCUGCGCUUGGAUUUCGUCCCCUGCU miR-192-2/3 CCGAGACCGAGUGCACAGGGCUCUGACCUAUGAAUUGAC 225 AGCCAGUGCUCUCGUCUCCCCUCUGGCUGCCAAUUCCAU AGGUCACAGGUAUGUUCGCCUCAAUGCCAG miR-192 GCCGAGACCGAGUGCACAGGGCUCUGACCUAUGAAUUGA 226 CAGCCAGUGCUCUCGUCUCCCCUCUGGCUGCCAAUUCCA UAGGUCACAGGUAUGUUCGCCUCAAUGCCAGC miR-193-1 CGAGGAUGGGAGCUGAGGGCUGGGUCUUUGCGGGCGAG 227 AUGAGGGUGUCGGAUCAACUGGCCUACAAAGUCCCAGU UCUCGGCCCCCG miR-193-2 GCUGGGUCUUUGCGGGCGAGAUGAGGGUGUCGGAUCAA 228 CUGGCCUACAAAGUCCCAGU miR-194-1 AUGGUGUUAUCAAGUGUAACAGCAACUCCAUGUGGACU 229 GUGUACCAAUUUCCAGUGGAGAUGCUGUUACUUUUGAU GGUUACCAA miR-194-2 GUGUAACAGCAACUCCAUGUGGACUGUGUACCAAUUUCC 230 AGUGGAGAUGCUGUUACUUUUGAU miR-195-1 AGCUUCCCUGGCUCUAGCAGCACAGAAAUAUUGGCACAG 231 GGAAGCGAGUCUGCCAAUAUUGGCUGUGCUGCUCCAGGC AGGGUGGUG miR-195-2 UAGCAGCACAGAAAUAUUGGCACAGGGAAGCGAGUCUG 232 CCAAUAUUGGCUGUGCUGCU miR-196-1 CUAGAGCUUGAAUUGGAACUGCUGAGUGAAUUAGGUAG 233 UUUCAUGUUGUUGGGCCUGGGUUUCUGAACACAACAAC AUUAAACCACCCGAUUCACGGCAGUUACUGCUCC miR-196a-1 GUGAAUUAGGUAGUUUCAUGUUGUUGGGCCUGGGUUUC 234 UGAACACAACAACAUUAAACCACCCGAUUCAC miR-196a-2 UGCUCGCUCAGCUGAUCUGUGGCUUAGGUAGUUUCAUG 235 (miR-196-2) UUGUUGGGAUUGAGUUUUGAACUCGGCAACAAGAAACU GCCUGAGUUACAUCAGUCGGUUUUCGUCGAGGGC miR-196 GUGAAUUAGGUAGUUUCAUGUUGUUGGGCCUGGGUUUC 236 UGAACACAACAACAUUAAACCACCCGAUUCAC miR-196b ACUGGUCGGUGAUUUAGGUAGUUUCCUGUUGUUGGGAU 237 CCACCUUUCUCUCGACAGCACGACACUGCCUUCAUUACU UCAGUUG miR-197 GGCUGUGCCGGGUAGAGAGGGCAGUGGGAGGUAAGAGC 238 UCUUCACCUUCACCACCUUCUCCACCCAGCAUGGCC MIR-197-2 GUGCAUGUGUAUGUAUGUGUGCAUGUGCAUGUGUAUGU 239 GUAUGAGUGCAUGCGUGUGUGC miR-198 UCAUUGGUCCAGAGGGGAGAUAGGUUCCUGUGAUUUUU 240 CCUUCUUCUCUAUAGAAUAAAUGA miR-199a-1 GCCAACCCAGUGUUCAGACUACCUGUUCAGGAGGCUCUC 241 AAUGUGUACAGUAGUCUGCACAUUGGUUAGGC miR-199a-2 AGGAAGCUUCUGGAGAUCCUGCUCCGUCGCCCCAGUGUU 242 CAGACUACCUGUUCAGGACAAUGCCGUUGUACAGUAGUC UGCACAUUGGUUAGACUGGGCAAGGGAGAGCA miR-199b CCAGAGGACACCUCCACUCCGUCUACCCAGUGUUUAGAC 243 UAUCUGUUCAGGACUCCCAAAUUGUACAGUAGUCUGCAC AUUGGUUAGGCUGGGCUGGGUUAGACCCUCGG miR-199s GCCAACCCAGUGUUCAGACUACCUGUUCAGGAGGCUCUC 244 AAUGUGUACAGUAGUCUGCACAUUGGUUAGGC miR-200a GCCGUGGCCAUCUUACUGGGCAGCAUUGGAUGGAGUCA 245 GGUCUCUAAUACUGCCUGGUAAUGAUGACGGC miR-200b CCAGCUCGGGCAGCCGUGGCCAUCUUACUGGGCAGCAUU 246 GGAUGGAGUCAGGUCUCUAAUACUGCCUGGUAAUGAUG ACGGCGGAGCCCUGCACG miR-200c CCCUCGUCUUACCCAGCAGUGUUUGGGUGCGGUUGGGAG 247 UCUCUAAUACUGCCGGGUAAUGAUGGAGG miR-202 GUUCCUUUUUCCUAUGCAUAUACUUCUUUGAGGAUCUG 248 GCCUAAAGAGGUAUAGGGCAUGGGAAGAUGGAGC miR-203 GUGUUGGGGACUCGCGCGCUGGGUCCAGUGGUUCUUAA 249 CAGUUCAACAGUUCUGUAGCGCAAUUGUGAAAUGUUUA GGACCACUAGACCCGGCGGGCGCGGCGACAGCGA miR-204 GGCUACAGUCUUUCUUCAUGUGACUCGUGGACUUCCCUU 250 UGUCAUCCUAUGCCUGAGAAUAUAUGAAGGAGGCUGGG AAGGCAAAGGGACGUUCAAUUGUCAUCACUGGC miR-205 AAAGAUCCUCAGACAAUCCAUGUGCUUCUCUUGUCCUUC 251 AUUCCACCGGAGUCUGUCUCAUACCCAACCAGAUUUCAG UGGAGUGAAGUUCAGGAGGCAUGGAGCUGACA miR-206-1 UGCUUCCCGAGGCCACAUGCUUCUUUAUAUCCCCAUAUG 252 GAUUACUUUGCUAUGGAAUGUAAGGAAGUGUGUGGUUU CGGCAAGUG miR-206-2 AGGCCACAUGCUUCUUUAUAUCCCCAUAUGGAUUACUUU 253 GCUAUGGAAUGUAAGGAAGUGUGUGGUUUU miR-208 UGACGGGCGAGCUUUUGGCCCGGGUUAUACCUGAUGCUC 254 ACGUAUAAGACGAGCAAAAAGCUUGUUGGUCA miR-210 ACCCGGCAGUGCCUCCAGGCGCAGGGCAGCCCCUGCCCA 255 CCGCACACUGCGCUGCCCCAGACCCACUGUGCGUGUGAC AGCGGCUGAUCUGUGCCUGGGCAGCGCGACCC miR-211 UCACCUGGCCAUGUGACUUGUGGGCUUCCCUUUGUCAUC 256 CUUCGCCUAGGGCUCUGAGCAGGGCAGGGACAGCAAAGG GGUGCUCAGUUGUCACUUCCCACAGCACGGAG miR-212 CGGGGCACCCCGCCCGGACAGCGCGCCGGCACCUUGGCU 257 CUAGACUGCUUACUGCCCGGGCCGCCCUCAGUAACAGUC UCCAGUCACGGCCACCGACGCCUGGCCCCGCC miR-213-2 CCUGUGCAGAGAUUAUUUUUUAAAAGGUCACAAUCAAC 258 AUUCAUUGCUGUCGGUGGGUUGAACUGUGUGGACAAGC UCACUGAACAAUGAAUGCAACUGUGGCCCCGCUU miR-213 GAGUUUUGAGGUUGCUUCAGUGAACAUUCAACGCUGUC 259 GGUGAGUUUGGAAUUAAAAUCAAAACCAUCGACCGUUG AUUGUACCCUAUGGCUAACCAUCAUCUACUCC miR-214 GGCCUGGCUGGACAGAGUUGUCAUGUGUCUGCCUGUCU 260 ACACUUGCUGUGCAGAACAUCCGCUCACCUGUACAGCAG GCACAGACAGGCAGUCACAUGACAACCCAGCCU miR-215 AUCAUUCAGAAAUGGUAUACAGGAAAAUGACCUAUGAA 261 UUGACAGACAAUAUAGCUGAGUUUGUCUGUCAUUUCUU UAGGCCAAUAUUCUGUAUGACUGUGCUACUUCAA miR-216 GAUGGCUGUGAGUUGGCUUAAUCUCAGCUGGCAACUGU 262 GAGAUGUUCAUACAAUCCCUCACAGUGGUCUCUGGGAU UAUGCUAAACAGAGCAAUUUCCUAGCCCUCACGA miR-217 AGUAUAAUUAUUACAUAGUUUUUGAUGUCGCAGAUACU 263 GCAUCAGGAACUGAUUGGAUAAGAAUCAGUCACCAUCA GUUCCUAAUGCAUUGCCUUCAGCAUCUAAACAAG miR-218-1 GUGAUAAUGUAGCGAGAUUUUCUGUUGUGCUUGAUCUA 264 ACCAUGUGGUUGCGAGGUAUGAGUAAAACAUGGUUCCG UCAAGCACCAUGGAACGUCACGCAGCUUUCUACA miR-218-2 GACCAGUCGCUGCGGGGCUUUCCUUUGUGCUUGAUCUAA 265 CCAUGUGGUGGAACGAUGGAAACGGAACAUGGUUCUGU CAAGCACCGCGGAAAGCACCGUGCUCUCCUGCA miR-219 CCGCCCCGGGCCGCGGCUCCUGAUUGUCCAAACGCAAUU 266 CUCGAGUCUAUGGCUCCGGCCGAGAGUUGAGUCUGGACG UCCCGAGCCGCCGCCCCCAAACCUCGAGCGGG miR-219-1 CCGCCCCGGGCCGCGGCUCCUGAUUGUCCAAACGCAAUU 267 CUCGAGUCUAUGGCUCCGGCCGAGAGUUGAGUCUGGACG UCCCGAGCCGCCGCCCCCAAACCUCGAGCGGG miR-219-2 ACUCAGGGGCUUCGCCACUGAUUGUCCAAACGCAAUUCU 268 UGUACGAGUCUGCGGCCAACCGAGAAUUGUGGCUGGAC AUCUGUGGCUGAGCUCCGGG miR-220 GACAGUGUGGCAUUGUAGGGCUCCACACCGUAUCUGACA 269 CUUUGGGCGAGGGCACCAUGCUGAAGGUGUUCAUGAUG CGGUCUGGGAACUCCUCACGGAUCUUACUGAUG miR-221 UGAACAUCCAGGUCUGGGGCAUGAACCUGGCAUACAAU 270 GUAGAUUUCUGUGUUCGUUAGGCAACAGCUACAUUGUC UGCUGGGUUUCAGGCUACCUGGAAACAUGUUCUC miR-222 GCUGCUGGAAGGUGUAGGUACCCUCAAUGGCUCAGUAG 271 CCAGUGUAGAUCCUGUCUUUCGUAAUCAGCAGCUACAUC UGGCUACUGGGUCUCUGAUGGCAUCUUCUAGCU miR-223 CCUGGCCUCCUGCAGUGCCACGCUCCGUGUAUUUGACAA 272 GCUGAGUUGGACACUCCAUGUGGUAGAGUGUCAGUUUG UCAAAUACCCCAAGUGCGGCACAUGCUUACCAG miR-224 GGGCUUUCAAGUCACUAGUGGUUCCGUUUAGUAGAUGA 273 UUGUGCAUUGUUUCAAAAUGGUGCCCUAGUGACUACAA AGCCC MIR-294-1 CAAUCUUCCUUUAUCAUGGUAUUGAUUUUUCAGUGCUU 274 (CHR16) CCCUUUUGUGUGAGAGAAGAUA miR-296 AGGACCCUUCCAGAGGGCCCCCCCUCAAUCCUGUUGUGC 275 CUAAUUCAGAGGGUUGGGUGGAGGCUCUCCUGAAGGGC UCU miR-299 AAGAAAUGGUUUACCGUCCCACAUACAUUUUGAAUAUG 276 UAUGUGGGAUGGUAAACCGCUUCUU miR-301 ACUGCUAACGAAUGCUCUGACUUUAUUGCACUACUGUAC 277 UUUACAGCUAGCAGUGCAAUAGUAUUGUCAAAGCAUCU GAAAGCAGG miR-302a CCACCACUUAAACGUGGAUGUACUUGCUUUGAAACUAA 278 AGAAGUAAGUGCUUCCAUGUUUUGGUGAUGG miR-302b GCUCCCUUCAACUUUAACAUGGAAGUGCUUUCUGUGACU 279 UUAAAAGUAAGUGCUUCCAUGUUUUAGUAGGAGU miR-302c CCUUUGCUUUAACAUGGGGGUACCUGCUGUGUGAAACA 280 AAAGUAAGUGCUUCCAUGUUUCAGUGGAGG miR-302d CCUCUACUUUAACAUGGAGGCACUUGCUGUGACAUGACA 281 AAAAUAAGUGCUUCCAUGUUUGAGUGUGG miR-320 GCUUCGCUCCCCUCCGCCUUCUCUUCCCGGUUCUUCCCG 282 GAGUCGGGAAAAGCUGGGUUGAGAGGGCGAAAAAGGAU GAGGU miR-321 UUGGCCUCCUAAGCCAGGGAUUGUGGGUUCGAGUCCCAC 283 CCGGGGUAAAGAAAGGCCGA miR-323 UUGGUACUUGGAGAGAGGUGGUCCGUGGCGCGUUCGCU 284 UUAUUUAUGGCGCACAUUACACGGUCGACCUCUUUGCAG UAUCUAAUC miR-324 CUGACUAUGCCUCCCCGCAUCCCCUAGGGCAUUGGUGUA 285 AAGCUGGAGACCCACUGCCCCAGGUGCUGCUGGGGGUUG UAGUC miR-325 AUACAGUGCUUGGUUCCUAGUAGGUGUCCAGUAAGUGU 286 UUGUGACAUAAUUUGUUUAUUGAGGACCUCCUAUCAAU CAAGCACUGUGCUAGGCUCUGG miR-326 CUCAUCUGUCUGUUGGGCUGGAGGCAGGGCCUUUGUGA 287 AGGCGGGUGGUGCUCAGAUCGCCUCUGGGCCCUUCCUCC AGCCCCGAGGCGGAUUCA miR-328 UGGAGUGGGGGGGCAGGAGGGGCUCAGGGAGAAAGUGC 288 AUACAGCCCCUGGCCCUCUCUGCCCUUCCGUCCCCUG miR-330 CUUUGGCGAUCACUGCCUCUCUGGGCCUGUGUCUUAGGC 289 UCUGCAAGAUCAACCGAGCAAAGCACACGGCCUGCAGAG AGGCAGCGCUCUGCCC miR-331 GAGUUUGGUUUUGUUUGGGUUUGUUCUAGGUAUGGUCC 290 CAGGGAUCCCAGAUCAAACCAGGCCCCUGGGCCUAUCCU AGAACCAACCUAAGCUC miR-335 UGUUUUGAGCGGGGGUCAAGAGCAAUAACGAAAAAUGU 291 UUGUCAUAAACCGUUUUUCAUUAUUGCUCCUGACCUCCU CUCAUUUGCUAUAUUCA miR-337 GUAGUCAGUAGUUGGGGGGUGGGAACGGCUUCAUACAG 292 GAGUUGAUGCACAGUUAUCCAGCUCCUAUAUGAUGCCU UUCUUCAUCCCCUUCAA miR-338 UCUCCAACAAUAUCCUGGUGCUGAGUGAUGACUCAGGCG 293 ACUCCAGCAUCAGUGAUUUUGUUGAAGA miR-339 CGGGGCGGCCGCUCUCCCUGUCCUCCAGGAGCUCACGUG 294 UGCCUGCCUGUGAGCGCCUCGACGACAGAGCCGGCGCCU GCCCCAGUGUCUGCGC miR-340 UUGUACCUGGUGUGAUUAUAAAGCAAUGAGACUGAUUG 295 UCAUAUGUCGUUUGUGGGAUCCGUCUCAGUUACUUUAU AGCCAUACCUGGUAUCUUA miR-342 GAAACUGGGCUCAAGGUGAGGGGUGCUAUCUGUGAUUG 296 AGGGACAUGGUUAAUGGAAUUGUCUCACACAGAAAUCG CACCCGUCACCUUGGCCUACUUA miR-345 ACCCAAACCCUAGGUCUGCUGACUCCUAGUCCAGGGCUC 297 GUGAUGGCUGGUGGGCCCUGAACGAGGGGUCUGGAGGC CUGGGUUUGAAUAUCGACAGC miR-346 GUCUGUCUGCCCGCAUGCCUGCCUCUCUGUUGCUCUGAA 298 GGAGGCAGGGGCUGGGCCUGCAGCUGCCUGGGCAGAGCG GCUCCUGC miR-367 CCAUUACUGUUGCUAAUAUGCAACUCUGUUGAAUAUAA 299 AUUGGAAUUGCACUUUAGCAAUGGUGAUGG miR-368 AAAAGGUGGAUAUUCCUUCUAUGUUUAUGUUAUUUAUG 300 GUUAAACAUAGAGGAAAUUCCACGUUUU miR-369 UUGAAGGGAGAUCGACCGUGUUAUAUUCGCUUUAUUGA 301 CUUCGAAUAAUACAUGGUUGAUCUUUUCUCAG miR-370 AGACAGAGAAGCCAGGUCACGUCUCUGCAGUUACACAGC 302 UCACGAGUGCCUGCUGGGGUGGAACCUGGUCUGUCU miR-371 GUGGCACUCAAACUGUGGGGGCACUUUCUGCUCUCUGGU 303 GAAAGUGCCGCCAUCUUUUGAGUGUUAC miR-372 GUGGGCCUCAAAUGUGGAGCACUAUUCUGAUGUCCAAG 304 UGGAAAGUGCUGCGACAUUUGAGCGUCAC miR-373 GGGAUACUCAAAAUGGGGGCGCUUUCCUUUUUGUCUGU 305 ACUGGGAAGUGCUUCGAUUUUGGGGUGUCCC miR-374 UACAUCGGCCAUUAUAAUACAACCUGAUAAGUGUUAUA 306 GCACUUAUCAGAUUGUAUUGUAAUUGUCUGUGUA mir-hes1 AUGGAGCUGCUCACCCUGUGGGCCUCAAAUGUGGAGGA 307 ACUAUUCUGAUGUCCAAGUGGAAAGUGCUGCGACAUUU GAGCGUCACCGGUGACGCCCAUAUCA mir-hes2 GCAUCCCCUCAGCCUGUGGCACUCAAACUGUGGGGGCAC 308 UUUCUGCUCUCUGGUGAAAGUGCCGCCAUCUUUUGAGU GUUACCGCUUGAGAAGACUCAACC mir-hes3 CGAGGAGCUCAUACUGGGAUACUCAAAAUGGGGGCGCU 309 UUCCUUUUUGUCUGUUACUGGGAAGUGCUUCGAUUUUG GGGUGUCCCUGUUUGAGUAGGGCAUC *An underlined sequence within a precursor sequence corresponds to a mature processed miR transcript (see Table 1b). Some precursor sequences have two underlined sequences denoting two different mature miRs that are derived from the same precursor. All sequences are human.

TABLE 1b Human Mature microRNA Sequences. Mature miRNA Mature miRNA Sequence SEQ ID Corresponding precursor Name (5′ to 3′) NO. microRNA(s); see Table 1a let-7a UGAGGUAGUAGGUUGUA 310 let-7a-1; let-7a-2; let-7a-3; let-7a-4 UAGUU let-7b UGAGGUAGUAGGUUGUG 311 let-7b UGGUU let-7c UGAGGUAGUAGGUUGUA 312 let-7c UGGUU let-7d AGAGGUAGUAGGUUGCA 313 let-7d; let-7d-v1 UAGU let-7e UGAGGUAGGAGGUUGUA 314 let-7e UAGU let-7f UGAGGUAGUAGAUUGUA 315 let-7f-1; let-7f-2-1; UAGUU let-7f-2-2 let-7g UGAGGUAGUAGUUUGUA 316 let-7g CAGU let-7i UGAGGUAGUAGUUUGUG 317 let-7i CU miR-1 UGGAAUGUAAAGAAGUA 318 miR-1b; miR-1b-1; UGUA miR-1b-2 miR-7 UGGAAGACUAGUGAUUU 319 miR-7-1; miR-7-1a; UGUU miR-7-2; miR-7-3 miR-9 UCUUUGGUUAUCUAGCU 320 miR-9-1; miR-9-2; GUAUGA miR-9-3 miR-9* UAAAGCUAGAUAACCGA 321 miR-9-1; miR-9-2; AAGU miR-9-3 miR-10a UACCCUGUAGAUCCGAA 322 miR-10a UUUGUG miR-10b UACCCUGUAGAACCGAA 323 miR-10b UUUGU miR-15a UAGCAGCACAUAAUGGU 324 miR-15a; miR-15a-2 UUGUG miR-15b UAGCAGCACAUCAUGGU 325 miR-15b UUACA miR-16 UAGCAGCACGUAAAUAU 326 miR-16-1; miR-16-2; UGGCG miR-16-13 miR-17-5p CAAAGUGCUUACAGUGC 327 miR-17 AGGUAGU miR-17-3p ACUGCAGUGAAGGCACU 328 miR-17 UGU miR-18 UAAGGUGCAUCUAGUGC 329 miR-18; miR-18-13 AGAUA miR-19a UGUGCAAAUCUAUGCAA 330 miR-19a; miR-19a-13 AACUGA miR-19b UGUGCAAAUCCAUGCAA 331 miR-19b-1; miR-19b-2 AACUGA miR-20 UAAAGUGCUUAUAGUGC 332 miR-20 (miR-20a) AGGUA miR-21 UAGCUUAUCAGACUGAU 333 miR-21; miR-21-17 GUUGA miR-22 AAGCUGCCAGUUGAAGA 334 miR-22 ACUGU miR-23a AUCACAUUGCCAGGGAU 335 miR-23a UUCC miR-23b AUCACAUUGCCAGGGAU 336 miR-23b UACCAC miR-24 UGGCUCAGUUCAGCAGG 337 miR-24-1; miR-24-2; AACAG miR-24-19; miR-24-9 miR-25 CAUUGCACUUGUCUCGG 338 miR-25 UCUGA miR-26a UUCAAGUAAUCCAGGAU 339 miR-26a; miR-26a-1; AGGCU miR-26a-2 miR-26b UUCAAGUAAUUCAGGAU 340 miR-26b AGGU miR-27a UUCACAGUGGCUAAGUU 341 miR-27a CCGCC miR-27b UUCACAGUGGCUAAGUU 342 miR-27b-1; miR-27b-2 CUG miR-28 AAGGAGCUCACAGUCUA 343 miR-28 UUGAG miR-29a CUAGCACCAUCUGAAAU 344 miR-29a-2; miR-29a CGGUU miR-29b UAGCACCAUUUGAAAUC 345 miR-29b-1; miR-29b-2 AGU miR-29c UAGCACCAUUUGAAAUC 346 miR-29c GGUUA miR-30a-5p UGUAAACAUCCUCGACU 347 miR-30a GGAAGC miR-30a-3p CUUUCAGUCGGAUGUUU 348 miR-30a GCAGC miR-30b UGUAAACAUCCUACACU 349 miR-30b-1; miR-30b-2 CAGC miR-30c UGUAAACAUCCUACACU 350 miR-30c CUCAGC miR-30d UGUAAACAUCCCCGACU 351 miR-30d GGAAG miR-30e UGUAAACAUCCUUGACU 352 miR-30e GGA miR-31 GGCAAGAUGCUGGCAUA 353 miR-31 GCUG miR-32 UAUUGCACAUUACUAAG 354 miR-32 UUGC miR-33 GUGCAUUGUAGUUGCAU 355 miR-33; miR-33b UG miR-34a UGGCAGUGUCUUAGCUG 356 miR-34a GUUGU miR-34b AGGCAGUGUCAUUAGCU 357 miR-34b GAUUG miR-34c AGGCAGUGUAGUUAGCU 358 miR-34c GAUUG miR-92 UAUUGCACUUGUCCCGG 359 miR-92-2; miR-92-1 CCUGU miR-93 AAAGUGCUGUUCGUGCA 360 miR-93-1; miR-93-2 GGUAG miR-95 UUCAACGGGUAUUUAUU 361 miR-95 GAGCA miR-96 UUUGGCACUAGCACAUU 362 miR-96 UUUGC miR-98 UGAGGUAGUAAGUUGUA 363 miR-98 UUGUU miR-99a AACCCGUAGAUCCGAUC 364 miR-99a UUGUG miR-99b CACCCGUAGAACCGACC 365 miR-99b UUGCG miR-100 UACAGUACUGUGAUAAC 366 miR-100 UGAAG miR-101 UACAGUACUGUGAUAAC 367 miR-101-1; miR-101-2 UGAAG miR-103 AGCAGCAUUGUACAGGG 368 miR-103-1 CUAUGA miR-105 UCAAAUGCUCAGACUCC 369 miR-105 UGU miR-106-a AAAAGUGCUUACAGUGC 370 miR-106-a AGGUAGC miR-106-b UAAAGUGCUGACAGUGC 371 miR-106-b AGAU miR-107 AGCAGCAUUGUACAGGG 372 miR-107 CUAUCA miR-122a UGGAGUGUGACAAUGGU 373 miR-122a-1; miR-122a-2 GUUUGU miR-124a UUAAGGCACGCGGUGAA 374 miR-124a-1; miR-124a-2; miR-124a-3 UGCCA miR-125a UCCCUGAGACCCUUUAA 375 miR-125a-1; miR-125a-2 CCUGUG miR-125b UCCCUGAGACCCUAACU 376 miR-125b-1; miR-125b-2 UGUGA miR-126* CAUUAUUACUUUUGGUA 377 miR-126-1; miR-126-2 CGCG miR-126 UCGUACCGUGAGUAAUA 378 miR-126-1; miR-126-2 AUGC miR-127 UCGGAUCCGUCUGAGCU 379 miR-127-1; miR-127-2 UGGCU miR-128a UCACAGUGAACCGGUCU 380 miR-128; miR-128a CUUUU miR-128b UCACAGUGAACCGGUCU 381 miR-128b CUUUC miR-129 CUUUUUGCGGUCUGGGC 382 miR-129-1; miR-129-2 UUGC miR-130a CAGUGCAAUGUUAAAAG 383 miR-130a GGC miR-130b CAGUGCAAUGAUGAAAG 384 miR-130b GGCAU miR-132 UAACAGUCUACAGCCAU 385 miR-132-1 GGUCG miR-133a UUGGUCCCCUUCAACCA 386 miR-133a-1; miR-133a-2 GCUGU miR-133b UUGGUCCCCUUCAACCA 387 miR-133b GCUA miR-134 UGUGACUGGUUGACCAG 388 miR-134-1; miR-134-2 AGGG miR-135a UAUGGCUUUUUAUUCCU 389 miR-135a; miR-135a-2 (miR-135-2) AUGUGA miR-135b UAUGGCUUUUCAUUCCU 390 miR-135b AUGUG miR-136 ACUCCAUUUGUUUUGAU 391 miR-136-1; miR-136-2 GAUGGA miR-137 UAUUGCUUAAGAAUACG 392 miR-137 CGUAG miR-138 AGCUGGUGUUGUGAAUC 393 miR-138-1; miR-138-2 miR-139 UCUACAGUGCACGUGUCU 394 miR-139 miR-140 AGUGGUUUUACCCUAUG 395 miR-140; miR-140as; GUAG miR-140s miR-141 AACACUGUCUGGUAAAG 396 miR-141-1; miR-141-2 AUGG miR-142-3p UGUAGUGUUUCCUACUU 397 miR-142 UAUGGA miR-142-5p CAUAAAGUAGAAAGCAC 398 miR-142 UAC miR-143 UGAGAUGAAGCACUGUA 399 miR-143-1 GCUCA miR-144 UACAGUAUAGAUGAUGU 400 miR-144-1; miR-144-2 ACUAG miR-145 GUCCAGUUUUCCCAGGA 401 miR-145-1; miR-145-2 AUCCCUU miR-146 UGAGAACUGAAUUCCAU 402 miR-146-1; miR-146-2 GGGUU miR-147 GUGUGUGGAAAUGCUUC 403 miR-147 UGC miR-148a UCAGUGCACUACAGAAC 404 miR-148a (miR-148) UUUGU miR-148b UCAGUGCAUCACAGAAC 405 miR-148b UUUGU miR-149 UCUGGCUCCGUGUCUUC 406 miR-149 ACUCC miR-150 UCUCCCAACCCUUGUACC 407 miR-150-1; miR-150-2 AGUG miR-151 ACUAGACUGAAGCUCCU 408 miR-151 UGAGG miR-152 UCAGUGCAUGACAGAAC 409 miR-152-1; miR-152-2 UUGG miR-153 UUGCAUAGUCACAAAAG 410 miR-153-1-1; miR-153-1-2; miR-153- UGA 2-1; miR-153-2-2 miR-154 UAGGUUAUCCGUGUUGC 411 miR-154-1; miR-154-2 CUUCG miR-154 * AAUCAUACACGGUUGAC 412 miR-154-1; miR-154-2 CUAUU miR-155 UUAAUGCUAAUCGUGAU 413 miR-155 AGGGG miR-181a AACAUUCAACGCUGUCG 414 miR-181a GUGAGU miR-181b AACAUUCAUUGCUGUCG 415 miR-181b-1; miR-181b-2 GUGGGUU miR-181c AACAUUCAACCUGUCGG 416 miR-181c UGAGU miR-182 UUUGGCAAUGGUAGAAC 417 miR-182; miR-182as UCACA miR-182* UGGUUCUAGACUUGCCA 418 miR-182; miR-182as ACUA miR-183 UAUGGCACUGGUAGAAU 419 miR-183 UCACUG miR-184 UGGACGGAGAACUGAUA 420 miR-184-1; miR-184-2 AGGGU miR-185 UGGAGAGAAAGGCAGUUC 421 miR-185-1; miR-185-2 miR-186 CAAAGAAUUCUCCUUUU 422 miR-186-1; miR-186-2 GGGCUU miR-187 UCGUGUCUUGUGUUGCA 423 miR-187 GCCG miR-188 CAUCCCUUGCAUGGUGG 424 miR-188 AGGGU miR-189 GUGCCUACUGAGCUGAU 425 miR-189-1; miR-189-2 AUCAGU miR-190 UGAUAUGUUUGAUAUAU 426 miR-190-1; miR-190-2 UAGGU miR-191 CAACGGAAUCCCAAAAG 427 miR-191-1; miR-191-2 CAGCU miR-192 CUGACCUAUGAAUUGAC 428 miR-192 AGCC miR-193 AACUGGCCUACAAAGUC 429 miR-193-1; miR-193-2 CCAG miR-194 UGUAACAGCAACUCCAU 430 miR-194-1; miR-194-2 GUGGA miR-195 UAGCAGCACAGAAAUAU 431 miR-195-1; miR-195-2 UGGC miR-196a UAGGUAGUUUCAUGUUG 432 miR-196a; miR-196a-2 (miR-196-2) UUGG miR-196b UAGGUAGUUUCCUGUUG 433 miR-196b UUGG miR-197 UUCACCACCUUCUCCACC 434 miR-197 CAGC miR-198 GGUCCAGAGGGGAGAUA 435 miR-198 GG miR-199a CCCAGUGUUCAGACUAC 436 miR-199a-1; miR-199a-2 CUGUUC miR-199a* UACAGUAGUCUGCACAU 437 miR-199a-1; miR-199a-2; miR-199s; UGGUU miR-199b miR-199b CCCAGUGUUUAGACUAU 438 miR-199b CUGUUC miR-200a UAACACUGUCUGGUAAC 439 miR-200a GAUGU miR-200b CUCUAAUACUGCCUGGU 440 miR-200b AAUGAUG miR-200c AAUACUGCCGGGUAAUG 441 miR-200c AUGGA miR-202 AGAGGUAUAGGGCAUGG 442 miR-202 GAAGA miR-203 GUGAAAUGUUUAGGACC 443 miR-203 ACUAG miR-204 UUCCCUUUGUCAUCCUA 444 miR-204 UGCCU miR-205 UCCUUCAUUCCACCGGA 445 miR-205 GUCUG miR-206 UGGAAUGUAAGGAAGUG 446 miR-206-1; miR-206-2 UGUGG miR-208 AUAAGACGAGCAAAAAG 447 miR-208 CUUGU miR-210 CUGUGCGUGUGACAGCG 448 miR-210 GCUG miR-211 UUCCCUUUGUCAUCCUU 449 miR-211 CGCCU miR-212 UAACAGUCUCCAGUCAC 450 miR-212 GGCC miR-213 ACCAUCGACCGUUGAUU 451 miR-213 GUACC miR-214 ACAGCAGGCACAGACAG 452 miR-214 GCAG miR-215 AUGACCUAUGAAUUGAC 453 miR-215 AGAC miR-216 UAAUCUCAGCUGGCAAC 454 miR-216 UGUG miR-217 UACUGCAUCAGGAACUG 455 miR-217 AUUGGAU miR-218 UUGUGCUUGAUCUAACC 456 miR-218-1; miR-218-2 AUGU miR-219 UGAUUGUCCAAACGCAA 457 miR-219; miR-219-1; UUCU miR-219-2 miR-220 CCACACCGUAUCUGACA 458 miR-220 CUUU miR-221 AGCUACAUUGUCUGCUG 459 miR-221 GGUUUC miR-222 AGCUACAUCUGGCUACU 460 miR-222 GGGUCUC miR-223 UGUCAGUUUGUCAAAUA 461 miR-223 CCCC miR-224 CAAGUCACUAGUGGUUC 462 miR-224 CGUUUA miR-296 AGGGCCCCCCCUCAAUCC 463 miR-296 UGU miR-299 UGGUUUACCGUCCCACA 464 miR-299 UACAU miR-301 CAGUGCAAUAGUAUUGU 465 miR-301 CAAAGC miR-302a UAAGUGCUUCCAUGUUU 466 miR-302a UGGUGA miR-302b* ACUUUAACAUGGAAGUG 467 miR-302b CUUUCU miR-302b UAAGUGCUUCCAUGUUU 468 miR-302b UAGUAG miR-302c* UUUAACAUGGGGGUACC 469 miR-302c UGCUG miR-302c UAAGUGCUUCCAUGUUU 470 miR-302c CAGUGG miR-302d UAAGUGCUUCCAUGUUU 471 miR-302d GAGUGU miR-320 AAAAGCUGGGUUGAGAG 472 miR-320 GGCGAA miR-321 UAAGCCAGGGAUUGUGG 473 miR-321 GUUC miR-323 GCACAUUACACGGUCGA 474 miR-323 CCUCU miR-324-5p CGCAUCCCCUAGGGCAU 475 miR-324 UGGUGU miR-324-3p CCACUGCCCCAGGUGCU 476 miR-324 GCUGG miR-325 CCUAGUAGGUGUCCAGU 477 miR-325 AAGU miR-326 CCUCUGGGCCCUUCCUCC 478 miR-326 AG miR-328 CUGGCCCUCUCUGCCCUU 479 miR-328 CCGU miR-330 GCAAAGCACACGGCCUG 480 miR-330 CAGAGA miR-331 GCCCCUGGGCCUAUCCU 481 miR-331 AGAA miR-335 UCAAGAGCAAUAACGAA 482 miR-335 AAAUGU miR-337 UCCAGCUCCUAUAUGAU 483 miR-337 GCCUUU miR-338 UCCAGCAUCAGUGAUUU 484 miR-338 UGUUGA miR-339 UCCCUGUCCUCCAGGAG 485 miR-339 CUCA miR-340 UCCGUCUCAGUUACUUU 486 miR-340 AUAGCC miR-342 UCUCACACAGAAAUCGC 487 miR-342 ACCCGUC miR-345 UGCUGACUCCUAGUCCA 488 miR-345 GGGC miR-346 UGUCUGCCCGCAUGCCU 489 miR-346 GCCUCU miR-367 AAUUGCACUUUAGCAAU 490 miR-367 GGUGA miR-368 ACAUAGAGGAAAUUCCA 491 miR-368 CGUUU miR-369 AAUAAUACAUGGUUGAU 492 miR-369 CUUU miR-370 GCCUGCUGGGGUGGAAC 493 miR-370 CUGG miR-371 GUGCCGCCAUCUUUUGA 494 miR-371 GUGU miR-372 AAAGUGCUGCGACAUUU 495 miR-372 GAGCGU miR-373* ACUCAAAAUGGGGGCGC 496 miR-373 UUUCC miR-373 GAAGUGCUUCGAUUUUG 497 miR-373 GGGUGU miR-374 UUAUAAUACAACCUGAU 498 miR-374 AAGUG

The present invention encompasses methods of diagnosing or prognosticating whether a subject has, or is at risk for developing, a cancer and/or myeloproliferative disorder. The methods comprise determining the level of at least one miR gene product in a sample from the subject and comparing the level of the miR gene product in the sample to a control. As used herein, a “subject” can be any mammal that has, or is suspected of having, a cancer and/or myeloproliferative disorder. In a preferred embodiment, the subject is a human who has, or is suspected of having, a cancer, myeloproliferative disorder and/or a platelet disorder.

The level of at least one miR gene product can be measured in cells of a biological sample obtained from the subject. For example, a tissue sample can be removed from a subject suspected of having cancer and/or a myeloproliferative disorder by conventional biopsy techniques. In another embodiment, a blood sample can be removed from the subject, and white blood cells can be isolated for DNA extraction by standard techniques. In one embodiment, the blood or tissue sample is obtained from the subject prior to initiation of radiotherapy, chemotherapy or other therapeutic treatment. A corresponding control tissue or blood sample, or a control reference sample (e.g., obtained from a population of control samples), can be obtained from unaffected tissues of the subject, from a normal human individual or population of normal individuals, or from cultured cells corresponding to the majority of cells in the subject's sample. The control tissue or blood sample can then processed along with the sample from the subject, so that the levels of miR gene product produced from a given miR gene in cells from the subject's sample can be compared to the corresponding miR gene product levels from cells of the control sample. Alternatively, a reference sample can be obtained and processed separately (e.g., at a different time) from the test sample and the level of a miR gene product produced from a given miR gene in cells from the test sample can be compared to the corresponding miR gene product level from the reference sample.

In one embodiment, the level of the at least one miR gene product in the test sample is greater than the level of the corresponding miR gene product in the control sample (i.e., expression of the miR gene product is “upregulated”). As used herein, expression of a miR gene product is “upregulated” when the amount of miR gene product in a cell or tissue sample from a subject is greater than the amount of the same gene product in a control (e.g., a reference standard, a control cell sample, a control tissue sample). In another embodiment, the level of the at least one miR gene product in the test sample is less than the level of the corresponding miR gene product in the control sample (i.e., expression of the miR gene product is “downregulated”). As used herein, expression of a miR gene is “downregulated” when the amount of miR gene product produced from that gene in a cell or tissue sample from a subject is less than the amount produced from the same gene in a control cell or tissue sample. The relative miR gene expression in the control and normal samples can be determined with respect to one or more RNA expression standards. The standards can comprise, for example, a zero miR gene expression level, the miR gene expression level in a standard cell line, the miR gene expression level in unaffected tissues of the subject, or the average level of miR gene expression previously obtained for a population of normal human controls (e.g., a control reference standard).

An alteration (i.e., an increase or decrease) in the level of a miR gene product in the sample obtained from the subject, relative to the level of a corresponding miR gene product in a control sample, is indicative of the presence of cancer and/or a myeloproliferative disorder in the subject. In one embodiment, the level of the at least one miR gene product in the test sample is greater than the level of the corresponding miR gene product in the control sample. miR gene products having higher expression levels in cancer cell lines (e.g., AMKL cell lines) than control cells (e.g., in vitro CD34+-differentiated megakaryocytes) are described and exemplified herein (see, e.g., Example 5). In one embodiment, the at least one miR gene product is selected from the group consisting of miR-101, miR-126, miR-99a, miR-99-prec, miR-106, miR-339, miR-99b, miR-149, miR-33, miR-135, miR-20 and combinations thereof. In another embodiment, the at least one miR gene product is selected from the group consisting of miR-101, miR-126, miR-106, miR-20 and miR-135 and combinations thereof. In yet another embodiment, the at least one miR gene product is selected from the group consisting of miR-106, miR-20 and miR-135 and combinations thereof. As described and exemplified herein, the increased expression of such miR gene products discriminates cancerous cells from corresponding non-cancerous cells.

As described herein, the diagnostic and prognostic methods of the invention can be used to diagnose or prognosticate cancers and/or myeloproliferative disorders. In particular embodiments, the diagnostic and prognostic methods are used to diagnose or prognosticate a cancer in a subject, tissue sample, cell sample or fluid sample. The diagnostic and prognostic methods can be used to diagnose or prognosticate any type of cancer. In particular embodiments, the diagnostic and prognostic methods can be used to diagnose or prognosticate a leukemia. In one embodiment, the leukemia that is diagnosed or prognosticated is acute myeloid leukemia (e.g., acute megakaryoblastic leukemia). In other embodiments, the diagnostic and prognostic methods can be used to diagnose or prognosticate multiple myeloma.

The diagnostic and prognostic methods of the invention can also be used to diagnose or prognosticate hematologic malignancies (e.g., myeloproliferative disorders). In one embodiment, the myeloproliferative disorder that is diagnosed or prognosticated is selected from the group consisting of essential thrombocytemia (ET), polycythemia vera (PV), myelodisplasia, myelofibrosis (e.g., agnogenic myeloid metaplasia (AMM) (also referred to as idiopathic myelofibrosis)) and chronic myelogenous leukemia (CML).

In particular embodiments, the diagnostic, prognostic and therapeutic methods of the invention can also be used to diagnose, prognosticate and/or treat platelet disorders (e.g., inherited platelet disorders). For example, the diagnostic, prognostic and therapeutic methods can be used to diagnose, prognosticate and/or treat defects in platelet-vessel wall interactions (i.e., disorders of adhesion). Such adhesion disorders include, e.g., von Willebrand disease (deficiency or defect in plasma vWF) and Bernard-Soulier syndrome (deficiency or defect in GPIb). In other embodiments, the diagnostic, prognostic and therapeutic methods can be used to diagnose, prognosticate and/or treat defects in platelet-platelet interaction (i.e., disorders of aggregation). Such aggregation disorders include, e.g., congenital afibrinogenemia (deficiency of plasma fibrinogen) and glanzmann thrombasthenia (deficiency or defect in GPIIb-IIIa). In other embodiments, the diagnostic, prognostic and therapeutic methods can be used to diagnose, prognosticate and/or treat disorders of platelet secretion and abnormalities of granules. Such disorders of platelet secretion and abnormalities of granules include, e.g., storage pool deficiency and Quebec platelet disorder. In yet other embodiments, the diagnostic, prognostic and therapeutic methods can be used to diagnose, prognosticate and/or treat disorders of platelet secretion and signal transduction (primary secretion defects). Such primary secretion defects include, e.g., defects in platelet-agonist interaction (receptor defects) (e.g., thromboxane A2, collagen, ADP, epinephrine), defects in G-protein activation (e.g., Gαq deficiency, Gas abnormalities, Gαi deficiency), defects in phosphatidylinositol metabolism (e.g., phospholipase C-2 deficiency), defects in calcium mobilization, defects in protein phosphorylation (pleckstrin) PKC-y deficiency, and abnormalities in arachidonic acid pathways and thromboxane synthesis (e.g., cyclooxygenase deficiency, thromboxane synthase deficiency). In other embodiments, the diagnostic, prognostic and therapeutic methods can be used to diagnose, prognosticate and/or treat defects in cytoskeletal regulation (e.g., Wiskott-Aldrich syndrome). In still other embodiments, the diagnostic, prognostic and therapeutic methods can be used to diagnose, prognosticate and/or treat disorders of platelet coagulant-protein interaction (membrane phospholipid defects) (e.g., Scott syndrome). Other platelet disorders (e.g., inherited platelet disorders) can also be diagnosed, prognosticated and/or treated using the methods of the invention.

The invention also provides methods of determining the prognosis of a subject with cancer and/or a myeloproliferative disorder. In this method, the level of at least one miR gene product, which is associated with a particular prognosis in cancer and/or a myeloproliferative disorder (e.g., a good or positive prognosis, a poor or adverse prognosis), is measured in a test sample from the subject. An alteration (e.g., an increase, a decrease) in the level of the miR gene product in the test sample, relative to the level of a corresponding miR gene product in a control sample, is indicative of the subject having a cancer and/or myeloproliferative disorder with a particular prognosis. In one embodiment, the miR gene product is associated with an adverse (i.e., poor) prognosis. Examples of an adverse prognosis include, but are not limited to, low survival rate and rapid disease progression. In one embodiment, the level of the at least one miR gene product in the test sample is greater than the level of the corresponding miR gene product in a control sample (i.e., it is upregulated). In a particular embodiment, the at least one miR gene product that is upregulated is selected from the group consisting of miR-101, miR-126, miR-99a, miR-99-prec, miR-106, miR-339, miR-99b, miR-149, miR-33, miR-135, miR-20 and combinations thereof. In another embodiment, the at least one miR gene product that is upregulated is selected from the group consisting of miR-101, miR-126, miR-106, miR-20 and miR-135 and combinations thereof. In yet another embodiment, the at least one miR gene product that is upregulated is selected from the group consisting of miR-106, miR-20 and miR-135 and combinations thereof. The increased expression of such miR gene products can correlate with an adverse prognosis and the severity of a subject's cancer and/or myeloproliferative disorder.

In certain embodiments of the diagnostic and prognostic methods described herein, the level of the at least one miR gene product is measured by reverse transcribing RNA from a test sample obtained from the subject to provide a set of target oligodeoxynucleotides, hybridizing the target oligodeoxynucleotides to a microarray that comprises miRNA-specific probe oligonucleotides to provide a hybridization profile for the test sample, and comparing the test sample hybridization profile to a hybridization profile generated from a control sample.

Identification of targets of particular miR gene products (e.g., those miR gene products exhibiting upregulated or downregulated expression relative to a control sample) can aid in elucidating mechanisms of action of microRNAs. As described and exemplified herein, particular targets and putative targets of select microRNAs were identified (see, e.g., Tables 2, 3 and 5 and Exemplification). For example, the transcription factor MAFB was identified as a target of mi-130a (Example 2). Similarly, HOXA1 was identified as a target of miR-10a (Example 5). For both miRs, direct interaction of the miR with the 3′ UTR of its respective target was demonstrated (Examples 2 and 5). Moreover, an inverse relation in the expression of the miR and its respective target were demonstrated. Thus, expression of pre-miR-130a resulted in decreased expression of MAFB (see, e.g., FIG. 2C) while expression of pre-miR-10a resulted in decreased expression of HOXA1 (see, e.g., FIGS. 3C, 3F and 3G). Thus, in one embodiment, expression of target genes of particular microRNAs (e.g., those listed in Tables 2, 3 and 5) can be used to diagnose cancer and/or a myeloproliferative disorder. Such target genes display inverse expression to the respective miR that targets it. One of skill in the art can measure the expression levels of any of these target genes using known methods and/or methods described herein for measuring the expression levels of microRNAs (e.g., quantitative or semi-quantitative RT-PCR, Northern blot analysis, solution hybridization detection, microarray analysis), without undue experimentation. In particular embodiments, the target gene that is measured is MAFB or HOXA1.

The level of the at least one miR gene product can be measured using a variety of techniques that are well known to those of skill in the art (e.g., quantitative or semi-quantitative RT-PCR, Northern blot analysis, solution hybridization detection). In a particular embodiment, the level of at least one miR gene product is measured by reverse transcribing RNA from a test sample obtained from the subject to provide a set of target oligodeoxynucleotides, hybridizing the target oligodeoxynucleotides to one or more miRNA-specific probe oligonucleotides (e.g., a microarray that comprises miRNA-specific probe oligonucleotides) to provide a hybridization profile for the test sample, and comparing the test sample hybridization profile to a hybridization profile generated from a control sample. An alteration in the signal of at least one miRNA in the test sample relative to the control sample is indicative of the subject either having, or being at risk for developing cancer and/or a myeloproliferative disorder. In one embodiment, the signal of at least one miRNA is upregulated, relative to the signal generated from the control sample. In another embodiment, the signal of at least one miRNA is downregulated, relative to the signal generated from the control sample. In a particular embodiment, the microarray comprises miRNA-specific probe oligonucleotides for a substantial portion of all known human miRNAs (e.g., the miRNAs listed in Tables 1a and 1b plus other known or discovered miRNAs). In a further embodiment, the microarray comprises miRNA-specific probe oligonucleotides for one or more miRNAs selected from the group consisting of miR-101, miR-126, miR-99a, miR-99-prec, miR-106, miR-339, miR-99b, miR-149, miR-33, miR-135, miR-20 and a combination thereof. In one embodiment, the microarray comprises miRNA-specific probe oligonucleotides for one or more miRNAs selected from the group consisting of miR-101, miR-126, miR-106, miR-20, miR-135 and a combination thereof.

The microarray can be prepared from gene-specific oligonucleotide probes generated from known miRNA sequences. The array may contain two different oligonucleotide probes for each miRNA, one containing the active, mature sequence and the other being specific for the precursor of the miRNA. The array may also contain controls, such as one or more mouse sequences differing from human orthologs by only a few bases, which can serve as controls for hybridization stringency conditions. tRNAs and other RNAs (e.g., rRNAs, mRNAs) from both species may also be printed on the microchip, providing an internal, relatively stable, positive control for specific hybridization. One or more appropriate controls for non-specific hybridization may also be included on the microchip. For this purpose, sequences are selected based upon the absence of any homology with any known miRNAs.

The microarray may be fabricated using techniques known in the art. For example, probe oligonucleotides of an appropriate length, e.g., 40 nucleotides, are 5′-amine modified at position C6 and printed using commercially available microarray systems, e.g., the GeneMachine OmniGrid™ 100 Microarrayer and Amersham CodeLink™ activated slides. Labeled cDNA oligomer corresponding to the target RNAs is prepared by reverse transcribing the target RNA with labeled primer. Following first strand synthesis, the RNA/DNA hybrids are denatured to degrade the RNA templates. The labeled target cDNAs thus prepared are then hybridized to the microarray chip under hybridizing conditions, e.g., 6×SSPE/30% formamide at 25° C. for 18 hours, followed by washing in 0.75×TNT at 37° C. for 40 minutes. At positions on the array where the immobilized probe DNA recognizes a complementary target cDNA in the sample, hybridization occurs. The labeled target cDNA marks the exact position on the array where binding occurs, allowing automatic detection and quantification. The output consists of a list of hybridization events, indicating the relative abundance of specific cDNA sequences, and therefore the relative abundance of the corresponding complementary miRs, in the patient sample. According to one embodiment, the labeled cDNA oligomer is a biotin-labeled cDNA, prepared from a biotin-labeled primer. The microarray is then processed by direct detection of the biotin-containing transcripts using, e.g., Streptavidin-Alexa647 conjugate, and scanned utilizing conventional scanning methods. Image intensities of each spot on the array are proportional to the abundance of the corresponding miR in the patient sample.

The use of the array has several advantages for miRNA expression detection. First, the global expression of several hundred genes can be identified in the same sample at one time point. Second, through careful design of the oligonucleotide probes, expression of both mature and precursor molecules can be identified. Third, in comparison with Northern blot analysis, the chip requires a small amount of RNA, and provides reproducible results using 2.5 μg of total RNA. The relatively limited number of miRNAs (a few hundred per species) allows the construction of a common microarray for several species, with distinct oligonucleotide probes for each. Such a tool would allow for analysis of trans-species expression for each known miR under various conditions.

In addition to use for quantitative expression level assays of specific miRs, a microchip containing miRNA-specific probe oligonucleotides corresponding to a substantial portion of the miRNome, preferably the entire miRNome, may be employed to carry out miR gene expression profiling, for analysis of miR expression patterns. Distinct miR signatures can be associated with established disease markers, or directly with a disease state.

According to the expression profiling methods described herein, total RNA from a sample from a subject suspected of having a cancer and/or a myeloproliferative disorder is quantitatively reverse transcribed to provide a set of labeled target oligodeoxynucleotides complementary to the RNA in the sample. The target oligodeoxynucleotides are then hybridized to a microarray comprising miRNA-specific probe oligonucleotides to provide a hybridization profile for the sample. The result is a hybridization profile for the sample representing the expression pattern of miRNA in the sample. The hybridization profile comprises the signal from the binding of the target oligodeoxynucleotides from the sample to the miRNA-specific probe oligonucleotides in the microarray. The profile may be recorded as the presence or absence of binding (signal vs. zero signal). More preferably, the profile recorded includes the intensity of the signal from each hybridization. The profile is compared to the hybridization profile generated from a normal (e.g., noncancerous, non-myeloproliferative disorder) control sample or reference sample. An alteration in the signal is indicative of the presence of, or propensity to develop, cancer in the subject.

Other techniques for measuring miR gene expression are also within the skill in the art, and include various techniques for measuring rates of RNA transcription and degradation.

The invention also provides methods of diagnosing whether a subject has, or is at risk for developing, a cancer and/or myeloproliferative disorder with an adverse prognosis. In this method, the level of at least one miR gene product, which is associated with an adverse prognosis in a cancer and/or myeloproliferative disorder, is measured by reverse transcribing RNA from a test sample obtained from the subject to provide a set of target oligodeoxynucleotides. The target oligodeoxynucleotides are then hybridized to one or more miRNA-specific probe oligonucleotides (e.g., a microarray that comprises miRNA-specific probe oligonucleotides) to provide a hybridization profile for the test sample, and the test sample hybridization profile is compared to a hybridization profile generated from a control sample. An alteration in the signal of at least one miRNA in the test sample relative to the control sample is indicative of the subject either having, or being at risk for developing, a cancer and/or myeloproliferative disorder with an adverse prognosis. miRs suitable for use in this method include, e.g., those that are upregulated in cancerous cells (e.g., AMKL cells).

In particular embodiments of the diagnostic, prognostic and therapeutic methods of the invention, as well as the pharmaceutical compositions of the invention, the miR gene product is not one or more of let7a-2, let-7c, let-7g, let-7i, miR-7-2, miR-7-3, miR-9, miR-9-1, miR-10a, miR-15a, miR-15b, miR-16-1, miR-16-2, miR-17-5p, miR-20a, miR-21, miR-24-1, miR-24-2, miR-25, miR-29b-2, miR-30, miR-30a-5p, miR-30c, miR-30d, miR-31, miR-32, miR-34, miR-34a, miR-34a prec, miR-34a-1, miR-34a-2, miR-92-2, miR-96, miR-99a, miR-99b prec, miR-100, miR-103, miR-106a, miR-107, miR-123, miR-124a-1, miR-125b-1, miR-125b-2, miR-126*, miR-127, miR-128b, miR-129, miR-129-1/2 prec, miR-132, miR-135-1, miR-136, miR-137, miR-141, miR-142-as, miR-143, miR-146, miR-148, miR-149, miR-153, miR-155, miR 159-1, miR-181, miR-181b-1, miR-182, miR-186, miR-191, miR-192, miR-195, miR-196-1, miR-196-1 prec, miR-196-2, miR-199a-1, miR-199a-2, miR-199b, miR-200b, miR-202, miR-203, miR-204, miR-205, miR-210, miR-211, miR-212, miR-214, miR-215, miR-217, miR-221 and/or miR-223.

As described herein, the level of a miR gene product in a sample can be measured using any technique that is suitable for detecting RNA expression levels in a biological sample. Suitable techniques (e.g., Northern blot analysis, RT-PCR, in situ hybridization) for determining RNA expression levels in a biological sample (e.g., cells, tissues) are well known to those of skill in the art. In a particular embodiment, the level of at least one miR gene product is detected using Northern blot analysis. For example, total cellular RNA can be purified from cells by homogenization in the presence of nucleic acid extraction buffer, followed by centrifugation. Nucleic acids are precipitated, and DNA is removed by treatment with DNase and precipitation. The RNA molecules are then separated by gel electrophoresis on agarose gels according to standard techniques, and transferred to nitrocellulose filters. The RNA is then immobilized on the filters by heating. Detection and quantification of specific RNA is accomplished using appropriately labeled DNA or RNA probes complementary to the RNA in question. See, for example, Molecular Cloning: A Laboratory Manual, J. Sambrook et al., eds., 2nd edition, Cold Spring Harbor Laboratory Press, 1989, Chapter 7, the entire disclosure of which is incorporated by reference.

Suitable probes (e.g., DNA probes, RNA probes) for Northern blot hybridization of a given miR gene product can be produced from the nucleic acid sequences provided in Table 1a and Table 1b and include, but are not limited to, probes having at least about 70%, 75%, 80%, 85%, 90%, 95%, 98% or 99% complementarity to a miR gene product of interest, as well as probes that have complete complementarity to a miR gene product of interest. Methods for preparation of labeled DNA and RNA probes, and the conditions for hybridization thereof to target nucleotide sequences, are described in Molecular Cloning: A Laboratory Manual, J. Sambrook et al., eds., 2nd edition, Cold Spring Harbor Laboratory Press, 1989, Chapters 10 and 11, the disclosures of which are incorporated herein by reference.

For example, the nucleic acid probe can be labeled with, e.g., a radionuclide, such as 3H, 32P, 33P, 14C, or 35S; a heavy metal; a ligand capable of functioning as a specific binding pair member for a labeled ligand (e.g., biotin, avidin or an antibody); a fluorescent molecule; a chemiluminescent molecule; an enzyme or the like.

Probes can be labeled to high specific activity by either the nick translation method of Rigby et al. (1977), J. Mol. Biol. 113:237-251 or by the random priming method of Fienberg et al. (1983), Anal. Biochem. 132:6-13, the entire disclosures of which are incorporated herein by reference. The latter is the method of choice for synthesizing 32P-labeled probes of high specific activity from single-stranded DNA or from RNA templates. For example, by replacing preexisting nucleotides with highly radioactive nucleotides according to the nick translation method, it is possible to prepare 32P-labeled nucleic acid probes with a specific activity well in excess of 108 cpm/microgram. Autoradiographic detection of hybridization can then be performed by exposing hybridized filters to photographic film. Densitometric scanning of the photographic films exposed by the hybridized filters provides an accurate measurement of miR gene transcript levels. Using another approach, miR gene transcript levels can be quantified by computerized imaging systems, such as the Molecular Dynamics 400-B 2D_Phosphorimager available from Amersham Biosciences, Piscataway, N.J.

Where radionuclide labeling of DNA or RNA probes is not practical, the random-primer method can be used to incorporate an analogue, for example, the dTTP analogue 5-(N—(N-biotinyl-epsilon-aminocaproyl)-3-aminoallyfldeoxyuridine triphosphate, into the probe molecule. The biotinylated probe oligonucleotide can be detected by reaction with biotin-binding proteins, such as avidin, streptavidin and antibodies (e.g., anti-biotin antibodies) coupled to fluorescent dyes or enzymes that produce color reactions.

In addition to Northern and other RNA hybridization techniques, determining the levels of RNA transcripts can be accomplished using the technique of in situ hybridization. This technique requires fewer cells than the Northern blotting technique and involves depositing whole cells onto a microscope cover slip and probing the nucleic acid content of the cell with a solution containing radioactive or otherwise labeled nucleic acid (e.g., cDNA or RNA) probes. This technique is particularly well-suited for analyzing tissue biopsy samples from subjects. The practice of the in situ hybridization technique is described in more detail in U.S. Pat. No. 5,427,916, the entire disclosure of which is incorporated herein by reference. Suitable probes for in situ hybridization of a given miR gene product can be produced from the nucleic acid sequences provided in Table 1a and Table 1b, and include, but are not limited to, probes having at least about 70%, 75%, 80%, 85%, 90%, 95%, 98% or 99% complementarity to a miR gene product of interest, as well as probes that have complete complementarity to a miR gene product of interest, as described above.

The relative number of miR gene transcripts in cells can also be determined by reverse transcription of miR gene transcripts, followed by amplification of the reverse-transcribed transcripts by polymerase chain reaction (RT-PCR), for example, as exemplified herein. The levels of miR gene transcripts can be quantified in comparison with an internal standard, for example, the level of mRNA from a “housekeeping” gene present in the same sample. A suitable “housekeeping” gene for use as an internal standard includes, e.g., U6 small nuclear RNA, myosin or glyceraldehyde-3-phosphate dehydrogenase (G3PDH). Methods for performing quantitative and semi-quantitative RT-PCR, and variations thereof, are well known to those of skill in the art.

In some instances, it may be desirable to simultaneously determine the expression level of a plurality of different miR gene products in a sample. In other instances, it may be desirable to determine the expression level of the transcripts of all known miR genes correlated with a cancer and/or myeloproliferative disorder. Assessing cancer-specific expression levels for hundreds of miR genes or gene products is time consuming and requires a large amount of total RNA (e.g., at least 20 μg for each Northern blot) and autoradiographic techniques that require radioactive isotopes.

To overcome these limitations, an oligolibrary, in microchip format (i.e., a microarray), may be constructed containing a set of oligonucleotide (e.g., oligodeoxynucleotide) probes that are specific for a set of miR genes. Using such a microarray, the expression level of multiple microRNAs in a biological sample can be determined by reverse transcribing the RNAs to generate a set of target oligodeoxynucleotides, and hybridizing them to probe the oligonucleotides on the microarray to generate a hybridization, or expression, profile. The hybridization profile of the test sample can then be compared to that of a control sample to determine which microRNAs have an altered expression level in cancer cells and/or cells exhibiting a myeloproliferative disorder. As used herein, “probe oligonucleotide” or “probe oligodeoxynucleotide” refers to an oligonucleotide that is capable of hybridizing to a target oligonucleotide. “Target oligonucleotide” or “target oligodeoxynucleotide” refers to a molecule to be detected (e.g., via hybridization). By “miR-specific probe oligonucleotide” or “probe oligonucleotide specific for a miR” is meant a probe oligonucleotide that has a sequence selected to hybridize to a specific miR gene product, or to a reverse transcript of the specific miR gene product.

An “expression profile” or “hybridization profile” of a particular sample is essentially a fingerprint of the state of the sample; while two states may have any particular gene similarly expressed, the evaluation of a number of genes simultaneously allows the generation of a gene expression profile that is unique to the state of the cell. That is, normal tissue, cell or fluid samples may be distinguished from corresponding cancerous and/or myeloproliferative disorder-exhibiting tissue, cell or fluid samples. Within cancerous and/or myeloproliferative disorder-exhibiting tissue, cell or fluid samples, different prognosis states (for example, good or poor long term survival prospects) may be determined. By comparing expression profiles of cancerous and/or myeloproliferative disorder-exhibiting tissue, cell or fluid samples in different states, information regarding which genes are important (including both upregulation and downregulation of genes) in each of these states is obtained. The identification of sequences that are differentially expressed in cancerous and/or myeloproliferative disorder-exhibiting tissue, cell or fluid samples, as well as differential expression resulting in different prognostic outcomes, allows the use of this information in a number of ways. For example, a particular treatment regime may be evaluated (e.g., to determine whether a chemotherapeutic drug acts to improve the long-term prognosis in a particular subject). Similarly, diagnosis may be done or confirmed by comparing samples from a subject with known expression profiles. Furthermore, these gene expression profiles (or individual genes) allow screening of drug candidates that suppress the cancer and/or myeloproliferative disorder expression profile or convert a poor prognosis profile to a better prognosis profile.

Without wishing to be bound by any one theory, it is believed that alterations in the level of one or more miR gene products in cells can result in the deregulation of one or more intended targets for these miRs, which can lead to aberrant megakaryocytic differentiation and/or the formation of cancer, a myeloproliferative disorder and/or a platelet disorder. Therefore, altering the level of the miR gene product (e.g., by decreasing the level of a miR that is upregulated in cancerous and/or myeloproliferative disorder-exhibiting cells, by increasing the level of a miR that is downregulated in cancerous and/or myeloproliferative disorder-exhibiting cells) may successfully treat the cancer, myeloproliferative disorder and/or platelet disorder.

Accordingly, the present invention encompasses methods of treating a cancer and/or myeloproliferative disorder in a subject, wherein at least one miR gene product is deregulated (e.g., downregulated, upregulated) in the cells (e.g., cancerous cells and/or myeloproliferative disorder-exhibiting cells) of the subject. In one embodiment, the level of at least one miR gene product in a test sample (e.g., a sample comprising cancerous and/or myeloproliferative disorder-exhibiting tissues, cells or fluid) is greater than the level of the corresponding miR gene product in a control or reference sample. In another embodiment, the level of at least one miR gene product in a test sample (e.g., a sample comprising cancerous and/or myeloproliferative disorder-exhibiting tissues, cells or fluid) is less than the level of the corresponding miR gene product in a control sample. When the at least one isolated miR gene product is downregulated in the test sample (e.g., a sample comprising cancerous and/or myeloproliferative disorder-exhibiting tissues, cells or fluid), the method comprises administering an effective amount of the at least one isolated miR gene product, or an isolated variant or biologically-active fragment thereof, such that proliferation of the cancerous and/or myeloproliferative disorder-exhibiting cells in the subject is inhibited. For example, when a miR gene product is downregulated in a cancer cell in a subject, administering an effective amount of an isolated miR gene product to the subject can inhibit proliferation of the cancer cell. The isolated miR gene product that is administered to the subject can be identical to an endogenous wild-type miR gene product (e.g., a miR gene product shown in Table 1a or Table 1b) that is downregulated in the cancer cell or it can be a variant or biologically-active fragment thereof. As defined herein, a “variant” of a miR gene product refers to a miRNA that has less than 100% identity to a corresponding wild-type miR gene product and possesses one or more biological activities of the corresponding wild-type miR gene product. Examples of such biological activities include, but are not limited to, inhibition of expression of a target RNA molecule (e.g., inhibiting translation of a target RNA molecule, modulating the stability of a target RNA molecule, inhibiting processing of a target RNA molecule) and inhibition of a cellular process associated with cancer and/or a myeloproliferative disorder (e.g., cell differentiation, cell growth, cell death). These variants include species variants and variants that are the consequence of one or more mutations (e.g., a substitution, a deletion, an insertion) in a miR gene. In certain embodiments, the variant is at least about 70%, 75%, 80%, 85%, 90%, 95%, 98%, or 99% identical to a corresponding wild-type miR gene product.

As defined herein, a “biologically-active fragment” of a miR gene product refers to an RNA fragment of a miR gene product that possesses one or more biological activities of a corresponding wild-type miR gene product. As described above, examples of such biological activities include, but are not limited to, inhibition of expression of a target RNA molecule and inhibition of a cellular process associated with cancer and/or a myeloproliferative disorder. In certain embodiments, the biologically-active fragment is at least about 5, 7, 10, 12, 15, or 17 nucleotides in length. In a particular embodiment, an isolated miR gene product can be administered to a subject in combination with one or more additional anti-cancer treatments. Suitable anti-cancer treatments include, but are not limited to, chemotherapy, radiation therapy and combinations thereof (e.g., chemoradiation).

When the at least one isolated miR gene product is upregulated in the cancer cells, the method comprises administering to the subject an effective amount of a compound that inhibits expression of the at least one miR gene product, such that proliferation of the cancer and/or myeloproliferative disorder-exhibiting cells is inhibited. Such compounds are referred to herein as miR gene expression-inhibition compounds. Examples of suitable miR gene expression-inhibition compounds include, but are not limited to, those described herein (e.g., double-stranded RNA, antisense nucleic acids and enzymatic RNA molecules). In a particular embodiment, a miR gene expression-inhibiting compound can be administered to a subject in combination with one or more additional anti-cancer treatments. Suitable anti-cancer treatments include, but are not limited to, chemotherapy, radiation therapy and combinations thereof (e.g., chemoradiation).

As described, when the at least one isolated miR gene product is upregulated in cancer cells (e.g., AMKL cells), the method comprises administering to the subject an effective amount of at least one compound for inhibiting expression of the at least one miR gene product, such that proliferation of cancer cells is inhibited. In one embodiment, the compound for inhibiting expression of the at least one miR gene product inhibits a miR gene product selected from the group consisting of miR-101, miR-126, miR-99a, miR-99-prec, miR-106, miR-339, miR-99b, miR-149, miR-33, miR-135, miR-20 and a combination thereof. In another embodiment, the compound for inhibiting expression of the at least one miR gene product inhibits a miR gene product selected from the group consisting of miR-101, miR-126, miR-106, miR-20, miR-135 and a combination thereof. In yet another embodiment, the compound for inhibiting expression of the at least one miR gene product inhibits a miR gene product selected from the group consisting of miR-106, miR-20, miR-135 and a combination thereof.

As described and exemplified herein, the transcription factor MAFB, which is upregulated in megakaryocytic differentiation, is a target of miR-130a. Moreover, an inverse relation in the expression of miR-130a and its respective target were demonstrated. Thus, expression of pre-miR-130a resulted in decreased expression of MAFB (see, e.g., FIG. 2C). MAFB is known to be deregulated in cancer (e.g., multiple myeloma and acute myeloid leukemia). For example, ectopic expression of MAFB has been observed in human myeloma cells carrying (14;20)(q32;q11) chromosomal translocations (Hanamura, I., et al. (2001) Jpn. J. Cancer Res. 92(6):638-644 (2001)). Accordingly, in one embodiment, the invention is a method of treating a cancer and/or myeloproliferative disorder in a subject comprising administering an effective amount of at least one miR gene product or an isolated variant or biologically-active fragment thereof to the subject, wherein:

the cancer and/or myeloproliferative disorder is associated with overexpression of a MAFB gene product; and

the at least one miR gene product binds to, and decreases expression of, the MAFB gene product.

In one embodiment, the at least one miR gene product or isolated variant or biologically-active fragment thereof comprises a nucleotide sequence that is complementary to a nucleotide sequence in the MAFB gene product (e.g., complementary to the 3′ UTR of MAFB). In a particular embodiment, the at least one miR gene product is miR-130a or an isolated variant or biologically-active fragment thereof.

Also as described and exemplified herein, mRNA of HOXA1, one of the members of the HOX family of proteins, is upregulated 7-fold in megakaryocytic differentiation (see, e.g., Example 4). Moreover, HOXA1 is a target of miR-10a and its expression is inversely related to the expression of miR-10a. Thus, expression of pre-miR-10a resulted in decreased expression of HOXA1 (see, e.g., FIGS. 3C, 3F and 3G). HOXA1. Expression of HOXA1 has been demonstrated to be sufficient to result in the oncogenic transformation of immortalized human mammary epithelial cells with aggressive in vivo tumor formation (Zhang, X., et al., (2002) J. Biol. Chem. 278(9):7580-7590). Further, forced expression of HOXA1 in mammary carcinoma cells, in a Bcl-2-dependent manner, resulted in a dramatic enhancement of anchorage-independent proliferation and colony formation in soft agar. Id. Accordingly, in one embodiment, the invention is a method of treating a cancer and/or myeloproliferative disorder in a subject comprising administering an effective amount of at least one miR gene product or an isolated variant or biologically-active fragment thereof to the subject, wherein:

the cancer and/or myeloproliferative disorder is associated with overexpression of a HOXA1 gene product; and

the at least one miR gene product binds to, and decreases expression of, the HOXA1 gene product.

In one embodiment, the at least one miR gene product or isolated variant or biologically-active fragment thereof comprises a nucleotide sequence that is complementary to a nucleotide sequence in the HOXA1 gene product (e.g., complementary to the 3′ UTR of HOXA1). In a particular embodiment, the at least one miR gene product is miR-10a or an isolated variant or biologically-active fragment thereof.

In a related embodiment, the methods of treating cancer and/or a myeloproliferative disorder in a subject additionally comprise the step of first determining the amount of at least one miR gene product in a sample from the subject, and comparing that level of the miR gene product to the level of a corresponding miR gene product in a control. If expression of the miR gene product is deregulated (e.g., downregulated, upregulated) in the sample from the subject, the methods further comprise altering the amount of the at least one miR gene product expressed in the sample from the subject. In one embodiment, the amount of the miR gene product expressed in the sample from the subject is less than the amount of the miR gene product expressed in the control, and an effective amount of the miR gene product, or an isolated variant or biologically-active fragment thereof, is administered to the subject. In another embodiment, the amount of the miR gene product expressed in the sample from the subject is greater than the amount of the miR gene product expressed in the control, and an effective amount of at least one compound for inhibiting expression of the at least one miR gene is administered to the subject. Suitable miRs and compounds that inhibit expression of miR genes include, for example, those described herein.

The terms “treat”, “treating” and “treatment”, as used herein, refer to ameliorating symptoms associated with a disease or condition, for example, cancer and/or a myeloproliferative disorder, including preventing or delaying the onset of the disease symptoms, and/or lessening the severity or frequency of symptoms of the disease or condition. The terms “subject”, “patient” and “individual” are defined herein to include animals, such as mammals, including, but not limited to, primates, cows, sheep, goats, horses, dogs, cats, rabbits, guinea pigs, rats, mice or other bovine, ovine, equine, canine, feline, rodent, or murine species. In a preferred embodiment, the animal is a human.

As used herein, an “effective amount” of an isolated miR gene product is an amount sufficient to inhibit proliferation of cells (e.g., cancerous cells, cells exhibiting a myeloproliferative disorder) in a subject suffering from cancer and/or a myeloproliferative disorder. One skilled in the art can readily determine an effective amount of a miR gene product to be administered to a given subject, by taking into account factors, such as the size and weight of the subject; the extent of disease penetration; the age, health and sex of the subject; the route of administration; and whether the administration is regional or systemic.

For example, an effective amount of an isolated miR gene product can be based on the approximate weight of a tumor mass to be treated. The approximate weight of a tumor mass can be determined by calculating the approximate volume of the mass, wherein one cubic centimeter of volume is roughly equivalent to one gram. An effective amount of the isolated miR gene product based on the weight of a tumor mass can be in the range of about 10-500 micrograms/gram of tumor mass. In certain embodiments, the tumor mass can be at least about 10 micrograms/gram of tumor mass, at least about 60 micrograms/gram of tumor mass or at least about 100 micrograms/gram of tumor mass.

An effective amount of an isolated miR gene product can also be based on the approximate or estimated body weight of a subject to be treated. Preferably, such effective amounts are administered parenterally or enterally, as described herein. For example, an effective amount of the isolated miR gene product that is administered to a subject can range from about 5-3000 micrograms/kg of body weight, from about 700-1000 micrograms/kg of body weight, or greater than about 1000 micrograms/kg of body weight.

One skilled in the art can also readily determine an appropriate dosage regimen for the administration of an isolated miR gene product to a given subject. For example, a miR gene product can be administered to the subject once (e.g., as a single injection or deposition). Alternatively, a miR gene product can be administered once or twice daily to a subject for a period of from about three to about twenty-eight days, more particularly from about seven to about ten days. In a particular dosage regimen, a miR gene product is administered once a day for seven days. Where a dosage regimen comprises multiple administrations, it is understood that the effective amount of the miR gene product administered to the subject can comprise the total amount of gene product administered over the entire dosage regimen.

As used herein, an “isolated” miR gene product is one that is synthesized, or altered or removed from the natural state through human intervention. For example, a synthetic miR gene product, or a miR gene product partially or completely separated from the coexisting materials of its natural state, is considered to be “isolated.” An isolated miR gene product can exist in a substantially-purified form, or can exist in a cell into which the miR gene product has been delivered. Thus, a miR gene product that is deliberately delivered to, or expressed in, a cell is considered an “isolated” miR gene product. A miR gene product produced inside a cell from a miR precursor molecule is also considered to be an “isolated” molecule. According to the invention, the isolated miR gene products described herein can be used for the manufacture of a medicament for treating cancer and/or a myeloproliferative disorder in a subject (e.g., a human).

Isolated miR gene products can be obtained using a number of standard techniques. For example, the miR gene products can be chemically synthesized or recombinantly produced using methods known in the art. In one embodiment, miR gene products are chemically synthesized using appropriately protected ribonucleoside phosphoramidites and a conventional DNA/RNA synthesizer. Commercial suppliers of synthetic RNA molecules or synthesis reagents include, e.g., Proligo (Hamburg, Germany), Dharmacon Research (Lafayette, Colo., U.S.A.), Pierce Chemical (part of Perbio Science, Rockford, Ill., U.S.A.), Glen Research (Sterling, Va., U.S.A.), ChemGenes (Ashland, Mass., U.S.A.) and Cruachem (Glasgow, UK).

Alternatively, the miR gene products can be expressed from recombinant circular or linear DNA plasmids using any suitable promoter. Suitable promoters for expressing RNA from a plasmid include, e.g., the U6 or H1 RNA pol III promoter sequences, or the cytomegalovirus promoters. Selection of other suitable promoters is within the skill in the art. The recombinant plasmids of the invention can also comprise inducible or regulatable promoters for expression of the miR gene products in cells (e.g., cancerous cells, cells exhibiting a myeloproliferative disorder).

The miR gene products that are expressed from recombinant plasmids can be isolated from cultured cell expression systems by standard techniques. The miR gene products that are expressed from recombinant plasmids can also be delivered to, and expressed directly in, cells (e.g., cancerous cells, cells exhibiting a myeloproliferative disorder). The use of recombinant plasmids to deliver the miR gene products to cells (e.g., cancerous cells, cells exhibiting a myeloproliferative disorder) is discussed in more detail below.

The miR gene products can be expressed from a separate recombinant plasmid, or they can be expressed from the same recombinant plasmid. In one embodiment, the miR gene products are expressed as RNA precursor molecules from a single plasmid, and the precursor molecules are processed into the functional miR gene product by a suitable processing system, including, but not limited to, processing systems extant within a cancer cell. Other suitable processing systems include, e.g., the in vitro Drosophila cell lysate system (e.g., as described in U.S. Published Patent Application No. 2002/0086356 to Tuschl et al., the entire disclosure of which is incorporated herein by reference) and the E. coli RNAse III system (e.g., as described in U.S. Published Patent Application No. 2004/0014113 to Yang et al., the entire disclosure of which is incorporated herein by reference).

Selection of plasmids suitable for expressing the miR gene products, methods for inserting nucleic acid sequences into the plasmid to express the gene products, and methods of delivering the recombinant plasmid to the cells of interest are within the skill in the art. See, for example, Zeng et al. (2002), Molecular Cell 9:1327-1333; Tuschl (2002), Nat. Biotechnol, 20:446-448; Brummelkamp et al. (2002), Science 296:550-553; Miyagishi et al. (2002), Nat. Biotechnol. 20:497-500; Paddison et al. (2002), Genes Dev. 16:948-958; Lee et al. (2002), Nat. Biotechnol. 20:500-505; and Paul et al. (2002), Nat. Biotechnol. 20:505-508, the entire disclosures of which are incorporated herein by reference.

In one embodiment, a plasmid expressing the miR gene products comprises a sequence encoding a miR precursor RNA under the control of the CMV intermediate-early promoter. As used herein, “under the control” of a promoter means that the nucleic acid sequences encoding the miR gene product are located 3′ of the promoter, so that the promoter can initiate transcription of the miR gene product coding sequences.

The miR gene products can also be expressed from recombinant viral vectors. It is contemplated that the miR gene products can be expressed from two separate recombinant viral vectors, or from the same viral vector. The RNA expressed from the recombinant viral vectors can either be isolated from cultured cell expression systems by standard techniques, or can be expressed directly in cells (e.g., cancerous cells, cells exhibiting a myeloproliferative disorder). The use of recombinant viral vectors to deliver the miR gene products to cells (e.g., cancerous cells, cells exhibiting a myeloproliferative disorder) is discussed in more detail below.

The recombinant viral vectors of the invention comprise sequences encoding the miR gene products and any suitable promoter for expressing the RNA sequences. Suitable promoters include, but are not limited to, the U6 or H1 RNA pol III promoter sequences, or the cytomegalovirus promoters. Selection of other suitable promoters is within the skill in the art. The recombinant viral vectors of the invention can also comprise inducible or regulatable promoters for expression of the miR gene products in a cancer cell.

Any viral vector capable of accepting the coding sequences for the miR gene products can be used; for example, vectors derived from adenovirus (AV); adeno-associated virus (AAV); retroviruses (e.g., lentiviruses (LV), Rhabdoviruses, murine leukemia virus); herpes virus, and the like. The tropism of the viral vectors can be modified by pseudotyping the vectors with envelope proteins or other surface antigens from other viruses, or by substituting different viral capsid proteins, as appropriate.

For example, lentiviral vectors of the invention can be pseudotyped with surface proteins from vesicular stomatitis virus (VSV), rabies, Ebola, Mokola, and the like. AAV vectors of the invention can be made to target different cells by engineering the vectors to express different capsid protein serotypes. For example, an AAV vector expressing a serotype 2 capsid on a serotype 2 genome is called AAV 2/2. This serotype 2 capsid gene in the AAV 2/2 vector can be replaced by a serotype 5 capsid gene to produce an AAV 2/5 vector. Techniques for constructing AAV vectors that express different capsid protein serotypes are within the skill in the art; see, e.g., Rabinowitz, J. E., et al. (2002), J. Virol. 76:791-801, the entire disclosure of which is incorporated herein by reference.

Selection of recombinant viral vectors suitable for use in the invention, methods for inserting nucleic acid sequences for expressing RNA into the vector, methods of delivering the viral vector to the cells of interest, and recovery of the expressed RNA products are within the skill in the art. See, for example, Dornburg (1995), Gene Therapy 2:301-310; Eglitis (1988), Biotechniques 6:608-614; Miller (1990), Hum. Gene Therapy 1:5-14; and Anderson (1998), Nature 392:25-30, the entire disclosures of which are incorporated herein by reference.

Particularly suitable viral vectors are those derived from AV and AAV. A suitable AV vector for expressing the miR gene products, a method for constructing the recombinant AV vector, and a method for delivering the vector into target cells, are described in Xia et al. (2002), Nat. Biotech. 20:1006-1010, the entire disclosure of which is incorporated herein by reference. Suitable AAV vectors for expressing the miR gene products, methods for constructing the recombinant AAV vector, and methods for delivering the vectors into target cells are described in Samulski et al. (1987), J. Virol. 61:3096-3101; Fisher et al. (1996), J. Virol., 70:520-532; Samulski et al. (1989), J. Virol. 63:3822-3826; U.S. Pat. No. 5,252,479; U.S. Pat. No. 5,139,941; International Patent Application No. WO 94/13788; and International Patent Application No. WO 93/24641, the entire disclosures of which are incorporated herein by reference. In one embodiment, the miR gene products are expressed from a single recombinant AAV vector comprising the CMV intermediate early promoter.

In a certain embodiment, a recombinant AAV viral vector of the invention comprises a nucleic acid sequence encoding a miR precursor RNA in operable connection with a polyT termination sequence under the control of a human U6 RNA promoter. As used herein, “in operable connection with a polyT termination sequence” means that the nucleic acid sequences encoding the sense or antisense strands are immediately adjacent to the polyT termination signal in the 5′ direction. During transcription of the miR sequences from the vector, the polyT termination signals act to terminate transcription.

In other embodiments of the treatment methods of the invention, an effective amount of at least one compound that inhibits miR expression can be administered to the subject. As used herein, “inhibiting miR expression” means that the production of the precursor and/or active, mature form of miR gene product after treatment is less than the amount produced prior to treatment. One skilled in the art can readily determine whether miR expression has been inhibited in cells (e.g., cancerous cells, cells exhibiting a myeloproliferative disorder), using, for example, the techniques for determining miR transcript level discussed herein Inhibition can occur at the level of gene expression (i.e., by inhibiting transcription of a miR gene encoding the miR gene product) or at the level of processing (e.g., by inhibiting processing of a miR precursor into a mature, active miR).

As used herein, an “effective amount” of a compound that inhibits miR expression is an amount sufficient to inhibit proliferation of cells (e.g., cancerous cells, cells exhibiting a myeloproliferative disorder) in a subject suffering from cancer and/or a myeloproliferative disorder. One skilled in the art can readily determine an effective amount of a miR expression-inhibiting compound to be administered to a given subject, by taking into account factors, such as the size and weight of the subject; the extent of disease penetration; the age, health and sex of the subject; the route of administration; and whether the administration is regional or systemic.

For example, an effective amount of the expression-inhibiting compound can be based on the approximate weight of a tumor mass to be treated, as described herein. An effective amount of a compound that inhibits miR expression can also be based on the approximate or estimated body weight of a subject to be treated, as described herein.

One skilled in the art can also readily determine an appropriate dosage regimen for administering a compound that inhibits miR expression to a given subject, as described herein. Suitable compounds for inhibiting miR gene expression include double-stranded RNA (such as short- or small-interfering RNA or “siRNA”), antisense nucleic acids, and enzymatic RNA molecules, such as ribozymes. Each of these compounds can be targeted to a given miR gene product and interfere with the expression (e.g., by inhibiting translation, by inducing cleavage and/or degradation) of the target miR gene product.

For example, expression of a given miR gene can be inhibited by inducing RNA interference of the miR gene with an isolated double-stranded RNA (“dsRNA”) molecule which has at least 90%, for example, at least 95%, at least 98%, at least 99%, or 100%, sequence homology with at least a portion of the miR gene product. In a particular embodiment, the dsRNA molecule is a “short or small interfering RNA” or “siRNA.”

siRNA useful in the present methods comprise short double-stranded RNA from about 17 nucleotides to about 29 nucleotides in length, preferably from about 19 to about 25 nucleotides in length. The siRNA comprise a sense RNA strand and a complementary antisense RNA strand annealed together by standard Watson-Crick base-pairing interactions (hereinafter “base-paired”). The sense strand comprises a nucleic acid sequence that is substantially identical to a nucleic acid sequence contained within the target miR gene product.

As used herein, a nucleic acid sequence in an siRNA that is “substantially identical” to a target sequence contained within the target mRNA is a nucleic acid sequence that is identical to the target sequence, or that differs from the target sequence by one or two nucleotides. The sense and antisense strands of the siRNA can comprise two complementary, single-stranded RNA molecules, or can comprise a single molecule in which two complementary portions are base-paired and are covalently linked by a single-stranded “hairpin” area.

The siRNA can also be altered RNA that differs from naturally-occurring RNA by the addition, deletion, substitution and/or alteration of one or more nucleotides. Such alterations can include addition of non-nucleotide material, such as to the end(s) of the siRNA or to one or more internal nucleotides of the siRNA, or modifications that make the siRNA resistant to nuclease digestion, or the substitution of one or more nucleotides in the siRNA with deoxyribonucleotides.

One or both strands of the siRNA can also comprise a 3′ overhang. As used herein, a “3′ overhang” refers to at least one unpaired nucleotide extending from the 3′-end of a duplexed RNA strand. Thus, in certain embodiments, the siRNA comprises at least one 3′ overhang of from 1 to about 6 nucleotides (which includes ribonucleotides or deoxyribonucleotides) in length, from 1 to about 5 nucleotides in length, from 1 to about 4 nucleotides in length, or from about 2 to about 4 nucleotides in length. In a particular embodiment, the 3′ overhang is present on both strands of the siRNA, and is 2 nucleotides in length. For example, each strand of the siRNA can comprise 3′ overhangs of dithymidylic acid (“TT”) or diuridylic acid (“uu”).

The siRNA can be produced chemically or biologically, or can be expressed from a recombinant plasmid or viral vector, as described above for the isolated miR gene products. Exemplary methods for producing and testing dsRNA or siRNA molecules are described in U.S. Published Patent Application No. 2002/0173478 to Gewirtz and in U.S. Published Patent Application No. 2004/0018176 to Reich et al., the entire disclosures of both of which are incorporated herein by reference.

Expression of a given miR gene can also be inhibited by an antisense nucleic acid. As used herein, an “antisense nucleic acid” refers to a nucleic acid molecule that binds to target RNA by means of RNA-RNA, RNA-DNA or RNA-peptide nucleic acid interactions, which alters the activity of the target RNA. Antisense nucleic acids suitable for use in the present methods are single-stranded nucleic acids (e.g., RNA, DNA, RNA-DNA chimeras, peptide nucleic acids (PNA)) that generally comprise a nucleic acid sequence complementary to a contiguous nucleic acid sequence in a miR gene product. The antisense nucleic acid can comprise a nucleic acid sequence that is 50-100% complementary, 75-100% complementary, or 95-100% complementary to a contiguous nucleic acid sequence in a miR gene product. Nucleic acid sequences of particular human miR gene products are provided in Table 1a and Table 1b. Without wishing to be bound by any theory, it is believed that the antisense nucleic acids activate RNase H or another cellular nuclease that digests the miR gene product/antisense nucleic acid duplex.

Antisense nucleic acids can also contain modifications to the nucleic acid backbone or to the sugar and base moieties (or their equivalent) to enhance target specificity, nuclease resistance, delivery or other properties related to efficacy of the molecule. Such modifications include cholesterol moieties, duplex intercalators, such as acridine, or one or more nuclease-resistant groups.

Antisense nucleic acids can be produced chemically or biologically, or can be expressed from a recombinant plasmid or viral vector, as described above for the isolated miR gene products. Exemplary methods for producing and testing are within the skill in the art; see, e.g., Stein and Cheng (1993), Science 261:1004 and U.S. Pat. No. 5,849,902 to Woolf et al., the entire disclosures of which are incorporated herein by reference.

Expression of a given miR gene can also be inhibited by an enzymatic nucleic acid. As used herein, an “enzymatic nucleic acid” refers to a nucleic acid comprising a substrate binding region that has complementarity to a contiguous nucleic acid sequence of a miR gene product, and which is able to specifically cleave the miR gene product. The enzymatic nucleic acid substrate binding region can be, for example, 50-100% complementary, 75-100% complementary, or 95-100% complementary to a contiguous nucleic acid sequence in a miR gene product. The enzymatic nucleic acids can also comprise modifications at the base, sugar, and/or phosphate groups. An exemplary enzymatic nucleic acid for use in the present methods is a ribozyme.

The enzymatic nucleic acids can be produced chemically or biologically, or can be expressed from a recombinant plasmid or viral vector, as described above for the isolated miR gene products. Exemplary methods for producing and testing dsRNA or siRNA molecules are described in Werner and Uhlenbeck (1995), Nucleic Acids Res. 23:2092-96; Hammann et al. (1999), Antisense and Nucleic Acid Drug Dev. 9:25-31; and U.S. Pat. No. 4,987,071 to Cech et al, the entire disclosures of which are incorporated herein by reference.

Administration of at least one miR gene product, or at least one compound for inhibiting miR expression, will inhibit the proliferation of cells (e.g., cancerous cells, cells exhibiting a myeloproliferative disorder) in a subject who has a cancer and/or a myeloproliferative disorder. As used herein, to “inhibit the proliferation of cancerous cells or cells exhibiting a myeloproliferative disorder” means to kill the cells, or permanently or temporarily arrest or slow the growth of the cells. Inhibition of cell proliferation can be inferred if the number of such cells in the subject remains constant or decreases after administration of the miR gene products or miR gene expression-inhibiting compounds. An inhibition of proliferation of cancerous cells or cells exhibiting a myeloproliferative disorder can also be inferred if the absolute number of such cells increases, but the rate of tumor growth decreases.

The number of cancer cells in the body of a subject can be determined by direct measurement, or by estimation from the size of primary or metastatic tumor masses. For example, the number of cancer cells in a subject can be measured by immunohistological methods, flow cytometry, or other techniques designed to detect characteristic surface markers of cancer cells.

The size of a tumor mass can be ascertained by direct visual observation, or by diagnostic imaging methods, such as X-ray, magnetic resonance imaging, ultrasound, and scintigraphy. Diagnostic imaging methods used to ascertain size of the tumor mass can be employed with or without contrast agents, as is known in the art. The size of a tumor mass can also be ascertained by physical means, such as palpation of the tissue mass or measurement of the tissue mass with a measuring instrument, such as a caliper.

The miR gene products or miR gene expression-inhibiting compounds can be administered to a subject by any means suitable for delivering these compounds to cells (e.g., cancer cells, cells exhibiting a myeloproliferative disorder) of the subject. For example, the miR gene products or miR expression-inhibiting compounds can be administered by methods suitable to transfect cells of the subject with these compounds, or with nucleic acids comprising sequences encoding these compounds. In one embodiment, the cells are transfected with a plasmid or viral vector comprising sequences encoding at least one miR gene product or miR gene expression-inhibiting compound.

Transfection methods for eukaryotic cells are well known in the art, and include, e.g., direct injection of the nucleic acid into the nucleus or pronucleus of a cell; electroporation; liposome transfer or transfer mediated by lipophilic materials; receptor-mediated nucleic acid delivery, bioballistic or particle acceleration; calcium phosphate precipitation, and transfection mediated by viral vectors.

For example, cells can be transfected with a liposomal transfer compound, e.g., DOTAP (N-[1-(2,3-dioleoyloxy)propyl]-N,N,N-trimethyl-ammonium methylsulfate, Boehringer-Mannheim) or an equivalent, such as LIPOFECTIN. The amount of nucleic acid used is not critical to the practice of the invention; acceptable results may be achieved with 0.1-100 micrograms of nucleic acid/105 cells. For example, a ratio of about 0.5 micrograms of plasmid vector in 3 micrograms of DOTAP per 105 cells can be used.

A miR gene product or miR gene expression-inhibiting compound can also be administered to a subject by any suitable enteral or parenteral administration route. Suitable enteral administration routes for the present methods include, e.g., oral, rectal, or intranasal delivery. Suitable parenteral administration routes include, e.g., intravascular administration (e.g., intravenous bolus injection, intravenous infusion, intra-arterial bolus injection, intra-arterial infusion and catheter instillation into the vasculature); peri- and intra-tissue injection (e.g., peri-tumoral and intra-tumoral injection, intra-retinal injection, or subretinal injection); subcutaneous injection or deposition, including subcutaneous infusion (such as by osmotic pumps); direct application to the tissue of interest, for example by a catheter or other placement device (e.g., a retinal pellet or a suppository or an implant comprising a porous, non-porous, or gelatinous material); and inhalation. Particularly suitable administration routes are injection, infusion and direct injection into the tumor.

In the present methods, a miR gene product or miR gene product expression-inhibiting compound can be administered to the subject either as naked RNA, in combination with a delivery reagent, or as a nucleic acid (e.g., a recombinant plasmid or viral vector) comprising sequences that express the miR gene product or miR gene expression-inhibiting compound. Suitable delivery reagents include, e.g., the Mirus Transit TKO lipophilic reagent; LIPOFECTIN; lipofectamine; cellfectin; polycations (e.g., polylysine) and liposomes.

Recombinant plasmids and viral vectors comprising sequences that express the miR gene products or miR gene expression-inhibiting compounds, and techniques for delivering such plasmids and vectors to cancer cells, are discussed herein and/or are well known in the art.

In a particular embodiment, liposomes are used to deliver a miR gene product or miR gene expression-inhibiting compound (or nucleic acids comprising sequences encoding them) to a subject. Liposomes can also increase the blood half-life of the gene products or nucleic acids. Suitable liposomes for use in the invention can be formed from standard vesicle-forming lipids, which generally include neutral or negatively charged phospholipids and a sterol, such as cholesterol. The selection of lipids is generally guided by consideration of factors, such as the desired liposome size and half-life of the liposomes in the blood stream. A variety of methods are known for preparing liposomes, for example, as described in Szoka et al. (1980), Ann. Rev. Biophys. Bioeng. 9:467; and U.S. Pat. Nos. 4,235,871, 4,501,728, 4,837,028, and 5,019,369, the entire disclosures of which are incorporated herein by reference.

The liposomes for use in the present methods can comprise a ligand molecule that targets the liposome to cancer cells. Ligands that bind to receptors prevalent in cancer cells, such as monoclonal antibodies that bind to tumor cell antigens, are preferred.

The liposomes for use in the present methods can also be modified so as to avoid clearance by the mononuclear macrophage system (“MMS”) and reticuloendothelial system (“RES”). Such modified liposomes have opsonization-inhibition moieties on the surface or incorporated into the liposome structure. In a particularly preferred embodiment, a liposome of the invention can comprise both an opsonization-inhibition moiety and a ligand.

Opsonization-inhibiting moieties for use in preparing the liposomes of the invention are typically large hydrophilic polymers that are bound to the liposome membrane. As used herein, an opsonization-inhibiting moiety is “bound” to a liposome membrane when it is chemically or physically attached to the membrane, e.g., by the intercalation of a lipid-soluble anchor into the membrane itself, or by binding directly to active groups of membrane lipids. These opsonization-inhibiting hydrophilic polymers form a protective surface layer that significantly decreases the uptake of the liposomes by the MMS and RES; e.g., as described in U.S. Pat. No. 4,920,016, the entire disclosure of which is incorporated herein by reference.

Opsonization-inhibiting moieties suitable for modifying liposomes are preferably water-soluble polymers with a number-average molecular weight from about 500 to about 40,000 daltons, and more preferably from about 2,000 to about 20,000 daltons. Such polymers include polyethylene glycol (PEG) or polypropylene glycol (PPG) or derivatives thereof; e.g., methoxy PEG or PPG, and PEG or PPG stearate; synthetic polymers, such as polyacrylamide or poly N-vinyl pyrrolidone; linear, branched, or dendrimeric polyamidoamines; polyacrylic acids; polyalcohols, e.g., polyvinylalcohol and polyxylitol to which carboxylic or amino groups are chemically linked, as well as gangliosides, such as ganglioside GM1. Copolymers of PEG, methoxy PEG, or methoxy PPG, or derivatives thereof, are also suitable. In addition, the opsonization-inhibiting polymer can be a block copolymer of PEG and either a polyamino acid, polysaccharide, polyamidoamine, polyethyleneamine, or polynucleotide. The opsonization-inhibiting polymers can also be natural polysaccharides containing amino acids or carboxylic acids, e.g., galacturonic acid, glucuronic acid, mannuronic acid, hyaluronic acid, pectic acid, neuraminic acid, alginic acid, carrageenan; aminated polysaccharides or oligosaccharides (linear or branched); or carboxylated polysaccharides or oligosaccharides, e.g., reacted with derivatives of carbonic acids with resultant linking of carboxylic groups. Preferably, the opsonization-inhibiting moiety is a PEG, PPG, or a derivative thereof. Liposomes modified with PEG or PEG-derivatives are sometimes called “PEGylated liposomes.”

The opsonization-inhibiting moiety can be bound to the liposome membrane by any one of numerous well-known techniques. For example, an N-hydroxysuccinimide ester of PEG can be bound to a phosphatidyl-ethanolamine lipid-soluble anchor, and then bound to a membrane. Similarly, a dextran polymer can be derivatized with a stearylamine lipid-soluble anchor via reductive amination using Na(CN)BH3 and a solvent mixture, such as tetrahydrofuran and water in a 30:12 ratio at 60° C.

Liposomes modified with opsonization-inhibition moieties remain in the circulation much longer than unmodified liposomes. For this reason, such liposomes are sometimes called “stealth” liposomes. Stealth liposomes are known to accumulate in tissues fed by porous or “leaky” microvasculature. Thus, tissue characterized by such microvasculature defects, for example, solid tumors, will efficiently accumulate these liposomes; see Gabizon, et al. (1988), Proc. Natl. Acad. Sci., U.S.A., 18:6949-53. In addition, the reduced uptake by the RES lowers the toxicity of stealth liposomes by preventing significant accumulation of the liposomes in the liver and spleen. Thus, liposomes that are modified with opsonization-inhibition moieties are particularly suited to deliver the miR gene products or miR gene expression-inhibition compounds (or nucleic acids comprising sequences encoding them) to tumor cells.

The miR gene products or miR gene expression-inhibition compounds can be formulated as pharmaceutical compositions, sometimes called “medicaments,” prior to administering them to a subject, according to techniques known in the art. Accordingly, the invention encompasses pharmaceutical compositions for treating cancer and/or a myeloproliferative disorder.

In one embodiment, the pharmaceutical composition of the invention comprises at least one miR expression-inhibition compound and a pharmaceutically-acceptable carrier. In a particular embodiment, the at least one miR expression-inhibition compound is specific for a miR gene product whose expression is greater in cancer cells than control cells (i.e., it is upregulated). In another embodiment, the miR expression-inhibition compound is specific for one or more miR gene products selected from the group consisting of miR-101, miR-126, miR-99a, miR-99-prec, miR-106, miR-339, miR-99b, miR-149, miR-33, miR-135 and miR-20. In another embodiment, the miR expression-inhibition compound is specific for one or more miR gene products selected from the group consisting of miR-101, miR-126, miR-106, miR-20, and miR-135. In yet another embodiment, the miR expression-inhibition compound is specific for one or more miR gene products selected from the group consisting of miR-106, miR-20 and miR-135.

In other embodiments, the pharmaceutical compositions comprise an effective amount of at least one miR gene product, or an isolated variant or biologically-active fragment thereof, and a pharmaceutically-acceptable carrier. In one embodiment, the invention is a pharmaceutical composition for treating a cancer and/or a myeloproliferative disorder, wherein the cancer and/or myeloproliferative disorder is associated with overexpression of a MAFB gene product. In this embodiment, the pharmaceutical composition comprises at least one miR gene product that binds to, and decreases expression of, the MAFB gene product. In a particular embodiment, the at least one miR gene product comprises a nucleotide sequence that is complementary to a nucleotide sequence in the MAFB gene product. In another embodiment, the at least one miR gene product is miR-130a or an isolated variant or biologically-active fragment thereof.

In one embodiment, the invention is a pharmaceutical composition for treating a cancer and/or a myeloproliferative disorder, wherein the cancer and/or myeloproliferative disorder is associated with overexpression of a HOXA1 gene product. In this embodiment, the pharmaceutical composition comprises at least one miR gene product that binds to, and decreases expression of, the HOXA1 gene product. In a particular embodiment, the at least one miR gene product comprises a nucleotide sequence that is complementary to a nucleotide sequence in the HOXA1 gene product. In another embodiment, the at least one miR gene product is miR-10a or an isolated variant or biologically-active fragment thereof.

Pharmaceutical compositions of the present invention are characterized as being at least sterile and pyrogen-free. As used herein, “pharmaceutical compositions” include formulations for human and veterinary use. Methods for preparing pharmaceutical compositions of the invention are within the skill in the art, for example, as described in Remington's Pharmaceutical Science, 17th ed., Mack Publishing Company, Easton, Pa. (1985), the entire disclosure of which is incorporated herein by reference.

The present pharmaceutical compositions comprise at least one miR gene product or miR gene expression-inhibition compound (or at least one nucleic acid comprising a sequence encoding the miR gene product or miR gene expression-inhibition compound) (e.g., 0.1 to 90% by weight), or a physiologically-acceptable salt thereof, mixed with a pharmaceutically-acceptable carrier. In certain embodiments, the pharmaceutical composition of the invention additionally comprises one or more anti-cancer agents (e.g., chemotherapeutic agents). The pharmaceutical formulations of the invention can also comprise at least one miR gene product or miR gene expression-inhibition compound (or at least one nucleic acid comprising a sequence encoding the miR gene product or miR gene expression-inhibition compound), which are encapsulated by liposomes and a pharmaceutically-acceptable carrier. In one embodiment, the pharmaceutical composition comprises a miR gene or gene product that is not miR-15, miR-16, miR-143 and/or miR-145.

Especially suitable pharmaceutically-acceptable carriers are water, buffered water, normal saline, 0.4% saline, 0.3% glycine, hyaluronic acid and the like.

In a particular embodiment, the pharmaceutical compositions of the invention comprise at least one miR gene product or miR gene expression-inhibition compound (or at least one nucleic acid comprising a sequence encoding the miR gene product or miR gene expression-inhibition compound) that is resistant to degradation by nucleases. One skilled in the art can readily synthesize nucleic acids that are nuclease resistant, for example by incorporating one or more ribonucleotides that is modified at the 2′-position into the miR gene product. Suitable 2′-modified ribonucleotides include those modified at the 2′-position with fluoro, amino, alkyl, alkoxy and O-allyl.

Pharmaceutical compositions of the invention can also comprise conventional pharmaceutical excipients and/or additives. Suitable pharmaceutical excipients include stabilizers, antioxidants, osmolality adjusting agents, buffers, and pH adjusting agents. Suitable additives include, e.g., physiologically biocompatible buffers (e.g., tromethamine hydrochloride), additions of chelants (such as, for example, DTPA or DTPA-bisamide) or calcium chelate complexes (such as, for example, calcium DTPA, CaNaDTPA-bisamide), or, optionally, additions of calcium or sodium salts (for example, calcium chloride, calcium ascorbate, calcium gluconate or calcium lactate). Pharmaceutical compositions of the invention can be packaged for use in liquid form, or can be lyophilized

For solid pharmaceutical compositions of the invention, conventional nontoxic solid pharmaceutically-acceptable carriers can be used; for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharin, talcum, cellulose, glucose, sucrose, magnesium carbonate, and the like.

For example, a solid pharmaceutical composition for oral administration can comprise any of the carriers and excipients listed above and 10-95%, preferably 25%-75%, of the at least one miR gene product or miR gene expression-inhibition compound (or at least one nucleic acid comprising sequences encoding them). A pharmaceutical composition for aerosol (inhalational) administration can comprise 0.01-20% by weight, preferably 1%-10% by weight, of the at least one miR gene product or miR gene expression-inhibition compound (or at least one nucleic acid comprising a sequence encoding the miR gene product or miR gene expression-inhibition compound) encapsulated in a liposome as described above, and a propellant. A carrier can also be included as desired; e.g., lecithin for intranasal delivery.

The pharmaceutical compositions of the invention can further comprise one or more anti-cancer agents. In a particular embodiment, the compositions comprise at least one miR gene product or miR gene expression-inhibition compound (or at least one nucleic acid comprising a sequence encoding the miR gene product or miR gene expression-inhibition compound) and at least one chemotherapeutic agent. Chemotherapeutic agents that are suitable for the methods of the invention include, but are not limited to, DNA-alkylating agents, anti-tumor antibiotic agents, anti-metabolic agents, tubulin stabilizing agents, tubulin destabilizing agents, hormone antagonist agents, topoisomerase inhibitors, protein kinase inhibitors, HMG-CoA inhibitors, CDK inhibitors, cyclin inhibitors, caspase inhibitors, metalloproteinase inhibitors, antisense nucleic acids, triple-helix DNAs, nucleic acids aptamers, and molecularly-modified viral, bacterial and exotoxic agents. Examples of suitable agents for the compositions of the present invention include, but are not limited to, cytidine arabinoside, methotrexate, vincristine, etoposide (VP-16), doxorubicin (adriamycin), cisplatin (CDDP), dexamethasone, arglabin, cyclophosphamide, sarcolysin, methylnitrosourea, fluorouracil, 5-fluorouracil (5FU), vinblastine, camptothecin, actinomycin-D, mitomycin C, hydrogen peroxide, oxaliplatin, irinotecan, topotecan, leucovorin, carmustine, streptozocin, CPT-11, taxol, tamoxifen, dacarbazine, rituximab, daunorubicin, 1-β-D-arabinofuranosylcytosine, imatinib, fludarabine, docetaxel and FOLFOX4.

The invention also encompasses methods of identifying an anti-cancer agent, comprising providing a test agent to a cell and measuring the level of at least one miR gene product in the cell. In one embodiment, the method comprises providing a test agent to a cell and measuring the level of at least one miR gene product associated with increased expression levels in cancer cells (e.g., in AMKL cells). A decrease in the level of the miR gene product that is associated with increased expression levels in cancer, relative to a suitable control (e.g., the level of the miR gene product in control cells), is indicative of the test agent being an anti-cancer agent. In a particular embodiment, the at least one miR gene product associated with increased expression levels in cancer cells is selected from the group consisting of miR-101, miR-126, miR-99a, miR-99-prec, miR-106, miR-339, miR-99b, miR-149, miR-33, miR-135 and miR-20. In another embodiment, the at least one miR gene product associated with increased expression levels in cancer cells is selected from the group consisting of miR-101, miR-126, miR-106, miR-20 and miR-135. In yet another embodiment, the at least one miR gene product associated with increased expression levels in cancer cells is selected from the group consisting of miR-106, miR-20 and miR-135. In one embodiment, the miR gene product is not one or more of let7a-2, let-7c, let-7g, let-7i, miR-7-2, miR-7-3, miR-9, miR-9-1, miR-10a, miR-15a, miR-15b, miR-16-1, miR-16-2, miR-17-5p, miR-20a, miR-21, miR-24-1, miR-24-2, miR-25, miR-29b-2, miR-30, miR-30a-5p, miR-30c, miR-30d, miR-31, miR-32, miR-34, miR-34a, miR-34a prec, miR-34a-1, miR-34a-2, miR-92-2, miR-96, miR-99a, miR-99b prec, miR-100, miR-103, miR-106a, miR-107, miR-123, miR-124a-1, miR-125b-1, miR-125b-2, miR-126*, miR-127, miR-128b, miR-129, miR-129-1/2 prec, miR-132, miR-135-1, miR-136, miR-137, miR-141, miR-142-as, miR-143, miR-146, miR-148, miR-149, miR-153, miR-155, miR 159-1, miR-181, miR-181b-1, miR-182, miR-186, miR-191, miR-192, miR-195, miR-196-1, miR-196-1 prec, miR-196-2, miR-199a-1, miR-199a-2, miR-199b, miR-200b, miR-202, miR-203, miR-204, miR-205, miR-210, miR-211, miR-212, miR-214, miR-215, miR-217, miR-221 and/or miR-223.

In one embodiment, the method comprises providing a test agent to a cell and measuring the level of at least one miR gene product associated with decreased expression levels in cancerous cells. An increase in the level of the miR gene product in the cell, relative to a suitable control (e.g., the level of the miR gene product in a control cell), is indicative of the test agent being an anti-cancer agent.

Suitable agents include, but are not limited to drugs (e.g., small molecules, peptides), and biological macromolecules (e.g., proteins, nucleic acids). The agent can be produced recombinantly, synthetically, or it may be isolated (i.e., purified) from a natural source. Various methods for providing such agents to a cell (e.g., transfection) are well known in the art, and several of such methods are described hereinabove. Methods for detecting the expression of at least one miR gene product (e.g., Northern blotting, in situ hybridization, RT-PCR, expression profiling) are also well known in the art. Several of these methods are also described herein.

The invention will now be illustrated by the following non-limiting examples.

EXEMPLIFICATION

Unless otherwise noted, the following materials and methods were used in the Examples.

Material and Methods

Cell Lines and Human CD34+ Cells.

The human chronic myeloid leukemia (CML) blast crisis cell lines K-562 and MEG-01 were obtained from American Type Tissue Culture (ATCC, Manassas, Va.) and maintained in RPMI 1640 (GIBCO, Carlsbad, Calif.) containing 10% FBS with penicillin-gentamycin at 37° C. with 5% CO2. The human megakaryoblastic leukemia cells UT-7, and CMK, and the chronic myeloid leukemia (CML) in blast crisis LAMA were obtained from DSMZ (Braunsweig, Germany). All cells were maintained in RPMI medium 1640 with 20% FBS and antibiotics, except UT-7 which is factor-dependent and was cultured in MEM-with 20% FBS and 5 ng/ml GM-CSF. Fresh and frozen human bone marrow CD34+ cells were obtained from Stemcell Technologies (Vancouver, B.C., Canada). FACS analysis for CD34 antigen revealed a purity >98%.

Human Progenitor CD34+ Cell Cultures.

Human bone marrow CD34+ cells were grown in STEM-media (Stemcell Technologies), which includes Isocove modified Dulbecco's medium supplemented with human transferrin, insulin, bovine serine albumin, human low density lipoprotein and glutamine, in the presence of 100 ng/ml human recombinant thrombopoietin (TPO) for the first 4 days, followed by a combination of 100 ng/ml TPO, IL3, and SCF (cytokine mixture CC-200, Stemcell Technologies). The initial cell density was 100,000 cells/ml; three times a week, the cell density was adjusted to 100,000 to 200,000 cells/ml. To increase the purity of the cells for microarray analysis, cell sorting was performed at day 10 of culture. Cells were incubated on ice for 45 minutes with anti-human CD34+, anti-human CD41+, anti-human CD61+, and their respective isotypes. After washing twice with PBS 3% FBS, cells were sorted using a FACS Aria sorting machine in bulk in two separate populations; CD34CD61+ and CD34+ CD61+ cells for culture and RNA extraction. The purity of the sorted populations was greater than 95%.

Megakaryocytes Characterization.

Cyto spin preparations of CD34+ progenitors in culture were performed and stained with May-Grunwald Giemsa at different time points during the megakaryocytic differentiation induction. For FACS analysis, the primary antibodies that were used were as follows: CD41A, CD61A, CD42B, and CD34 with their respective isotypes (BD Pharmingen, San Diego, Calif.). Cytometric studies were performed as previously described (Tajima, S., et al. (1996) J. Exp. Med. 184, 1357-1364) using a FACScalibur (BD Biosciences) and the CELLQUEST software (BD Biosciences).

RNA Extraction, Northern Blotting and miRNA Microarray Experiments.

Procedures were performed as described in detail elsewhere (Liu, C. G., et al. (2002) Proc. Natl. Acad. Sci. USA 101, 9740-9744). Raw data were normalized and analyzed in GENESPRING 7.2 software (zcomSilicon Genetics, Redwood City, Calif.). Expression data were median-centered by using both GENESPRING normalization option and global median normalization of the BIOCONDUCTOR package with similar results. Statistical comparisons were done by using the GENESPRING ANOVA tool, predictive analysis of microarray (PAM) and the significance analysis of microarray (SAM) software.

Reverse Transcriptase PCR (RT-PCR) and Real Time PCR.

Total RNA isolated with Trizol reagent (Invitrogen, Carlsbad, Calif.) was processed after DNAase treatment (Ambion, Austin, Tex.) directly to cDNA by reverse transcription using Superscript II (Invitrogen). Comparative real-time PCR was performed in triplicate. Primers and probes were obtained from Applied Biosystems (Foster City, Calif.) for the following genes: HOXA1, HOXA3, HOXB4, HOXB5, and HOXD10. Gene expression levels were quantified by using the ABI Prism 7900 Sequence detection system (Applied Biosystems). Normalization was performed by using the 18S RNA primer kit. Relative expression was calculated by using the computed tomography (CT) method. RT-PCR also was performed by using the following oligonucleotide primers:

MAFB FW; (SEQ ID NO: 499) 5′-AACTTTGTCTTGGGGGACAC-3′; MAFB RW; (SEQ ID NO: 500) 5′-GAGGGGAGGATCTGTTTTCC-3′; HOXA1 FW; (SEQ ID NO: 501) 5′-CCAGGAGCTCAGGAAGAAGA GAT-3′; and HOXA1 RW; (SEQ ID NO: 502) 5′-CCCTCTGAGGCATCTGATTGGGTTT-3′.

Real-Time Quantification of miRNAs by Stem-Loop RT-PCR.

Real time-PCR for pri-miRNAs 10a, miR15a, miR16-1, miR-130a, miR-20, miR-106, miR-17-5, miR-181b, miR-99a, and miR-126 were performed as described (Chen, C., et al. (2005) Nucl. Acids Res. 33, e179. 18S was used for normalization. All reagents and primers were obtained from Applied Biosystems.

Bioinformatics.

miRNA target prediction of the differentially expressed miRNAs was performed by using TARGETSCAN, MIRANDA, and PICTAR software.

Cell Transfection with miRNA Precursors.

miRNA precursors miR-10a and miR-130a were purchased from Ambion. Five million K562 cells were nucleoporated by using Amaxa (Gaithesburg, Md.) with 5 μg of precursor oligonucleotides in a total volume of 10 ml. The expression of the oligonucleotides was assessed by Northern blots and RT-PCR as described.

Luciferase Reporter Experiments.

The 3′ UTR segments containing the target sites for miR-10a and miR-130a from HOXA1 and MAFB genes, respectively, were amplified by PCR from genomic DNA and inserted into the pGL3 control vector (Promega, Madison, Wis.), by using the XbaI site immediately downstream from the stop codon of luciferase. The following oligonucleotide primer sets were used to generate specific fragments:

MAFB FW (SEQ ID NO: 503) 5′-GCATCTAGAGCACCCCAGAGGAGTGT-3′; MAFB RW (SEQ ID NO: 504) 5′-GCATCTAGACAAGCACCATGCGGTTC-3′; HOXA1 FW (SEQ ID NO: 505) 5′-TACTCTAGACCAGGAGCTCAGGAAGA-3′; and HOXA1 RW (SEQ ID NO: 506) 5′-MCATTCTAGATGAGGCATCTGATTGGG-3′.

We also generated two inserts with deletions of 5 by and 9 bp, respectively, from the site of perfect complementarity by using the QuikChange XL-site directed Mutagenesis Kit (Stratagene, La Jolla, Calif.). Wild type (WT) and mutant insert were confirmed by sequencing.

Human chronic myeloid leukemia (CML) in megakaryoblastic crisis cell line (MEG-01) was cotransfected in six-well plates by using Lipofectamine 2000 (Invitrogen) according to the manufacturer's protocol with 0.4 μg of the firefly luciferase report vector and 0.08 μg of the control vector containing Renilla luciferase, pRL-TK (Promega). For each well, 10 nM of the premiR-130a and premiR-10a precursors (Ambion) were used. Firefly and Renilla luciferase activities were measured consecutively by using the dual luciferase assays (Promega) 24 hours after transfection.

Western Blots.

Total and nuclear protein extracts from K562 cells transfected with miR-10a and miR-130a, as well as CD34+ cells at different stages of megakaryocytic differentiation were extracted by using RIPA buffer or Nuclear extraction Kit (Pierce, Rockford, Ill.). Protein expression was analyzed by Western blotting with the following primary antibodies: MAFB (Santa Cruz Biotechnology, Santa Cruz, Calif.), HOXA1 (R&D Systems, Minneapolis, Minn.), β-Actin and Nucleolin (Santa Cruz Biotechnology). Appropriate secondary antibodies were used (Santa Cruz Biotechnology).

Example 1 miRNA Expression During In Vitro Megakaryocytic Differentiation of CD34+ Progenitors

Using a combination of a specific megakaryocytic growth factor (thrombopoietin) and nonspecific cytokines (SCF and IL-3), we were able to generate in vitro pure, abundant megakaryocyte progeny from CD34+ bone marrow progenitors suitable for microarray studies (FIG. 4). Total RNA was obtained for miRNA chip analysis from three different CD34 progenitors at baseline and at days 10, 12, 14 and 16 of culture with cytokines. We initially compared the expression of miRNA between the CD34+ progenitors and the pooled CD34+ differentiated megakaryocytes at all points during the differentiation process. 17 miRNAs (Table 1) that are sharply down regulated during megakaryocytic differentiation were identified. There were no statistically significant miRNAs upregulated during megakaryocytic differentiation. Using predictive analysis of microarray (PAM), we identified 8 microRNAs that predicted megakaryocytic differentiation with no misclassification error: miR-10a, miR-10b, miR-30c, miR-106, miR-126, miR-130a, miR-132, and miR-143. All of these miRNAs, except miR-143, are included in the 17 miRNAs identified by significance analysis of microarray (SAM). Northern blots and real-time PCR for several miRNAs confirmed the results obtained by miRNA chip analysis (FIG. 1).

Because we found mainly downregulation of miRNAs during megakaryocytopoiesis, we

TABLE 2 Chromosomal Fold miRNA Location T-test (†) Change Putative targets hsa-mir-010a* 17 q21 −9.10 50.00 HOXA1, HOXA3, HOXD10, CRK, FLT1 hsa-mir-126* 9q34 −2.73 8.33 CRK, EVI2, HOXA9, MAFB, CMAF hsa-mir-106* xq26.2 −2.63 2.86 TAL1, FLT1, SKI, RUNX1, FOG2, FLI, PDGFRA, CRK hsa-mir-010b* 2q31 −2.17 11.11 HOXA1, HOXA3, HOXD10, ETS- 1, CRK, FLT1 hsa-mir-130a* 11q12 −2.08 4.76 MAFB, MYB, FOG2, CBFB, PDGFRA, SDFR1, CXCL12 hsa-mir-130a- 11q12 −2.07 7.69 NA± prec* hsa-mir-124a 8q23 −1.81 2.78 TAL1, SKI, FLT1, FOG2, ETS-1, CBFB, RAF1, MYB hsa-mir-032- 9q31 −1.76 3.57 NA± prec hsa-mir-101 1p31.3 −1.75 3.33 TAL1, CXCL12, MEIS1, MEIS2, ETS-1 RUNX1, MYB hsa-mir-30c 6q13 −1.71 2.56 CBFB, MAFG, HOXA1, SBF1, NCOR2, ERG hsa-mir-213* 1q31.3 −1.69 2.38 MAX-SATB2 hsa-mir-132- 17p13 −1.67 4.17 NA± prec hsa-mir-150* 19q13.3 −1.63 5.26 MYB, SDFR1 hsa-mir-020 13q31 −1.62 2.17 TAL1, SKI, RUNX-1, FLT1, CRK, FOG2, RARB hsa-mir-339 7p22 −1.60 3.03 SKI, ETV6, GATA2, FLT1, RAP1B, JUNB, MEIS2 hsa-let-7a 9q22 −1.58 2.94 HOXA1, HOXA9, MEIS2, ITGB3, PLDN hsa-let-7d 9q22 −1.56 2.17 HOXA1, HOXD1, ITGB3, RUNX1, PDGFRA hsa-mir-181c 19p13 −1.55 2.50 RUNX-1, KIT, HOXA1, MEIS2, ETS-1 ETV6, PDGFRA hsa-mir-181b 1q31.3 −1.53 2.13 RUNX-1, KIT, ITGA3, HOXA1, MEIS2, ETS-1, SDFR1, hsa-mir-017 13q31 −1.38 1.82 TAL1, SKI, FLT1, RUNX1, CRK, FOG1, ETS-1, MEIS1 (†) t test p < 0.05. *These miRNAs were identified by PAM as predictors of a megakaryocytic class with the lowest misclassification error. All, except miR-143 are downregulated during megakaryocytic differentiation. NA±: miRNA precursor sequence that does not contain the mature miRNA, therefore no putative target is shown.

hypothesized that these miRNAs may unblock target genes involved in differentiation. In line with this hypothesis, miRNAs that are sharply downregulated in our system are predicted to target genes with important roles in megakaryocytic differentiation. Among the transcription factors with well-known function in megakaryocytopoiesis, RUNX-1 (Elagib, K. E., et al. (2003) Blood, 101:4333-4341), Fli-1 (Athanasoiu, M., et al. (1996) Cell Growth Differ. 7, 1525-1534), FLT1 (Casella, I., et al. (2003) Blood 101, 1316-1323), ETV6 (Hock, H., et al. (2004) Genes Dev. 18:2336-2341), TAL1 (Begley, C. G., and Green, A. R. (1999) Blood, 93:2760-2770), ETS1 (Jackers, P., et al. (2004) J. Biol. Chem. 279:52183-52190) and CRK (Lannutti, B. J., et al. (2003) Exp. Hematol. 12:1268-1274) are putative targets for several miRNAs downregulated in differentiated megakaryocytes. Moreover, each of these transcription factors has more than one miRNA predicted to be its regulator. For example, RUNX1 (AML1) is predicted to be the target of miR-106, miR-181b, miR-101, let7d and the miR-17-92 cluster. The multiplicity of miRNAs predicted to target AML1 suggests a combinatorial model of regulation.

We then looked at the temporal expression of miRNAs during the megakaryocytic differentiation process from CD34+ progenitors. We focused on miRNAs that have been described in hematopoietic tissues, such as miR-223, miR-181, miR-155, miR-142, miR-15a, miR-16, miR-106 and the cluster of miR-17-92 (FIG. 5). We found sequential changes in the expression of miR-223. Initially, miR-223 is downregulated during megakaryocytic differentiation, but after 14 days in culture, its expression returns to levels comparable with that of CD34 progenitors (FIG. 1C). The miR-15a and miR-16-1 cluster also follows the same pattern of expression as miR-223 (FIG. 1D), whereas miR-181b, miR-155, miR-106a, miR-17, and miR-20 were downregulated during differentiation (FIG. 6). The temporal variation of the expression of miR-223 and miR-15a/mir-16-1 suggests a stage-specific function.

Table 2. miRNAs downregulated during in vitro CD34+ megakaryocytic differentiation. All differentially expressed miRNAs have q value <0.01 (false-positive rate).

Example 2 MAFB Transcription Factor is a Target of miR-130a

By using three target prediction algorithms (TARGETSCAN, MIRANDA, and PICTAR), we identified that miR-130a is predicted to target MAFB, a transcription factor that is upregulated during megakaryocytic differentiation and induces the GPIIb gene, in synergy with GATA1, SP1 and ETS-1 (Sevinsky, J. R., et al. (2004) Mol. Cell. Biol. 24, 4534-4545). To investigate this putative interaction, first, we examined MAFB protein and mRNA levels in CD34+ progenitors at baseline and after cytokine stimulation (FIG. 2A). We found that the MAFB protein is upregulated during in vitro megakaryocytic differentiation. Although the mRNA levels for MAFB by PCR increase with differentiation, this increase does not correlate well with the intensity of its protein expression. The inverse pattern of expression of MAFB and miR-130a suggested in vivo interaction that was further investigated.

To demonstrate a direct interaction between the 3′ UTRs of MAFB with miR-130a, we inserted the 3′ UTR regions predicted to interact with this miRNA into a luciferase vector. This experiment revealed a repression of about ˜60% of luciferase activity compared with control vector (FIG. 2B). As an additional control experiment, we used a mutated target mRNA sequence for MAFB lacking five of the complementary bases. As expected, the mutations completely abolished the interaction between miR-130a and its target 3′UTRs (FIG. 2B).

We also determined the in vivo consequences of overexpressing miR-130a on MAFB expression. The pre-miR-130a and a negative control were transfected by electroporation into K562 cells, which naturally express MAFB and lack miR-130a. Transfection of the pre-miR-130a, but not the control, resulted in a decrease in the protein levels at 48 hours (FIG. 2C). Northern blotting confirmed successful ectopic expression of miR-130a in K562 cells (FIG. 7).

Example 3 MiR-10a Correlates with HOXB Gene Expression

It has been reported that in mouse embryos, miR-10a, miR-10b, and miR-196 are expressed in HOX-like patterns (Mansfield, J. H., et al. (2004) Nature 36, 1079-1083) and closely follow their “host” HOX cluster during evolution (Tanzer, A., et al. (2005) J. Exp. Zool. B Mol. Dev. Evol. 304B, 75-85). These data suggest common regulatory elements across paralog clusters. MiR-10a is located at chromosome 17q21 within the cluster of the HOXB genes (FIG. 8) and miR-10b is located at chromosome 2q31 within the HOXD gene cluster. To determine whether the miR-10a expression pattern correlates with the expression of HOXB genes, we performed RT-PCR for HOXB4 and HOXB5, which are the genes located 5′ and 3′, respectively, to miR-10a in the HOXB cluster. As shown in FIG. 8, HOXB4 and HOXB5 expression paralleled that of miR-10a, suggesting a common regulatory mechanism.

Example 4 MiR-10a Downregulates HOXA1

We determined by miRNA array and Northern blot that miR-10a is sharply down-regulated during megakaryocytic differentiation. Interestingly, we found several HOX genes as putative targets for miR-10a (Table 2). We thus investigated whether miR-10a could target a HOX gene. We performed real-time PCR for the predicted HOX targets of miR-10: HOXA1, HOXA3, and HOXD10. After normalization with 18S RNA, we found that HOXA1 mRNA is upregulated 7-fold during megakaryocytic differentiation compared with CD34 progenitors (FIG. 3A; see also FIG. 9). HOXA1 protein levels were also upregulated during megakaryocytic differentiation (FIG. 3B). These results are in sharp contrast with the downregulation of miR-10a in megakaryocytic differentiation, suggesting that miR-10a could be an inhibitor of HOXA1 expression. To demonstrate a direct interaction of miR-10a and the 3′ UTR sequences of the HOXA1 gene, we carried out a luciferase reporter assay as described in Material and Methods. When the miRNA precursor miR-10a was introduced in the MEG01 cells along with the reporter plasmid containing the 3′ UTR sequence of HOXA1, a 50% reduction in luciferase activity was observed (FIG. 3C). The degree of complementarity between miR-10a and the HOXA1 3′ UTR is shown in FIG. 3D, as predicted by PICTAR.

To confirm in vivo these findings, we transfected K562 cells with the pre-miR-10a precursor using nucleoporation and measured HOXA1 mRNA expression by RT-PCR and HOXA1 protein levels by Western blotting. Successful ectopic expression of miR-10a was documented by Northern Blot (FIG. 3E). A significant reduction at the mRNA and protein levels for HOXA1 was found for K562 cells transfected with the miR-10a precursor but not with the negative control (FIGS. 3F and 3G). These data indicate that miR-10a targets HOXA1 in vitro and in vivo.

It has been reported that miR-196 induces cleavage of HOXB8 mRNA, pointing to a posttranscriptional restriction mechanism of HOX gene expression (Yekta, S., et al. (2004) Science, 304:594-596). Contrary to the miR-196-HOXB8 interaction, where an almost perfect complementarity exists, the degree of pairing between miR-10a and the human HOXA1 3′ UTR is suboptimal (FIG. 4). Although our results indicated target mRNA degradation, further studies are needed to determine whether cleavage or translational repression is the primary mechanism of downregulation of the HOXA1 gene in this system. A previous study using microarray analysis showed that a large number of target mRNA genes are downregulated by miRNA at the level of transcription (Lim, L. P., et al. (2005) Nature: 433, 769-771). These data raise the question whether target degradation is a consequence of translational repression and subsequent relocalization of the miR-target complexes to cytoplasmic processing bodies or is a primary event (Pillai, R. (2005) RNA 11, 1753-1761).

Example 5 miRNA Profiling in Acute Megakaryoblastic Leukemia (AMKL) Cell Lines

After the identification of the microRNA expression profile of CD34+ cells during megakaryocytic differentiation, we then investigated miRNA expression in AMKL cell lines with the goal to identify differentially expressed miRNAs that could have a pathogenic role in megakaryoblastic leukemia. We initially compared miRNA expression in four AMKL cell lines with that of in vitro CD34+-differentiated megakaryocytes. Using significance analysis of microarray (SAM), we identified 10 miRNAs upregulated in AMKL cell lines compared with that of CD34 in vitro-differentiated megakaryocytes (Table 3; see also Table 4). These miRNAs are as follows (in order of the fold increase with respect to differentiated megakaryocytes): miR-101, miR-126, miR-99a, miR-99-prec, miR-106, miR-339, miR-99b, miR-149, miR-33 and miR-135. Results were validated by RT-PCR as shown in FIG. 10. Using PAM, we compared miRNA expression in CD34+ cells with in vitro-differentiated megakaryocytes and AMKL cell lines (FIG. 10). Interestingly, we found five miRNAs involved in the megakaryocytic differentiation signature (miR-101, miR-126, miR-106, miR-20, and miR-135) that were upregulated in the leukemic cell lines (Tables 3, 5 and 6). Whether this profile represents merely a differentiation state of the cells or has a truly pathogenic role remains to be elucidated. Supporting the second hypothesis, miR-106, miR-135, and miR-20 are predicted to target RUNX1, which is one of the genes most commonly associated with leukemia (Nakao, M., et al. (2004) Oncogene 125, 709-719). Moreover, mutations of RUNX1 have been described in familial thrombocytopenias with a propensity to develop acute myeloid leukemia (Song, W. J., et al. (1999) Nat. Genet. 23, 166-175).

Table 3. microRNAs upregulated in acute megakaryoblastic cell lines compared with in vitro-differentiated megakaryocytes.

TABLE 3 Chromosomal t test Fold microRNA Location Score Change Putative Targets hsa-mir-101 1p31.3 6.14 11.85 MEIS2, RUNX1, ETS-1, C-MYB, FOS, BARB, NFE2L2 hsa-mir-126 9q34 4.91 11.97 V-CRK hsa-mir-099a 21q21 3.30 6.83 HOXA1, EIF2C, FOXA1 hsa-mir- 21q21 2.85 7.59 NA 099b-prec hsa-mir-106 xq26.2 2.79 3.33 FLT1, SKI, E2F1, NCOA3, PDGFRA, CRK hsa-mir-339 7p22 2.58 3.36 HOXA1, FLT1, PTP4A1, RAP1B hsa-mir-099b 19q13 2.46 4.19 HOXA1, MYCBP2 hsa-mir-149 2q37 2.29 3.53 RAP1A, MAFF, PDGFRA, SP1, NFIB hsa-mir-033 2q13 2.27 3.23 PDGFRA, HIF1A, MEIS2 hsa-mir-135 3p21 2.12 3.97 SP1, HIF1A, SP3, HNRPA1, HOXA10, RUNX1

All the miRNAs have a q value <0.01 (false discovery rate).

The same miRNAs, except miR-339 and miR-149, were found by using PAM to predict a megakaryoblastic leukemia class with no misclassification error.

The results described herein demonstrate that there is a downregulation of miRNAs during megakaryocytopoiesis. Hypothetically, the downregulation of miRNAs unblocks target genes involved in differentiation. In line with this hypothesis, miRNAs that are sharply downregulated in our system are predicted to target genes with important roles in megakaryocytic differentiation. Thus, we have shown that miR-130a targets MAFB and miR-10a modulates HOXA1. The fact that we found several differentially expressed miRNAs during differentiation and leukemia that are predicted to target HOXA1 suggests a function for HOXA1 in megakaryocytopoiesis. Loss and gain studies will ultimately be needed to define the role of HOXA1 in this differentiation process. Our findings delineate the expression of miRNAs in megakaryocytic differentiation and suggest a role for miRNA modulation of this lineage by targeting megakaryocytic transcription factors. Furthermore, in megakaryoblastic leukemia cell lines, we have found inverse expression of miRNAs involved in normal megakaryocytic differentiation. These data provide a starting point for future studies of miRNAs in megakaryocytopoiesis and leukemia.

Table 4. Signature of megakaryocytic differentiation.

TABLE 4 CD34 Megakaryocytic microRNA Expression Expression hsa-mir-010a up Down hsa-mir-126 up Down hsa-mir-130a-prec up Down hsa-mir-010b up Down hsa-mir-106 up Down hsa-mir-130a up Down hsa-mir-132 up Down hsa-mir-30c up Down hsa-mir-143-prec Down up

PAM selected microRNAs with a very low misclassification error.

Table 5 Signature of megakaryoblastic leukemia cell lines

TABLE 5 Level of t test Fold Expression MicroRNA Score Change in AML M7 Putative Targets hsa-mir-101- 6.14 11.85 up MEIS2, RUNX1, C-MYB, FOS, RARb, NFE2L2 hsa-mir-126 4.91 11.97 up V-CRK hsa-mir-099a 3.30 6.83 up HOXA1, EIF2C, FOXA1 hsa-mir-095 up SHOX2 hsa-mir-033 2.27 3.23 up PDGFRA, HIF1A, MEIS2 hsa-mir-135 2.12 3.97 up SP1, HIF1A, SP3, HNRPA1, HOXA10, RUNX1 hsa-mir-099b 2.85 7.59 up HOXA1, MYCBP2 hsa-mir-339 2.58 3.36 up HOXA1, FLT1, PTP4A1, RAP1B hsa-mir-106 2.79 3.33 up HOXA1, EIF2C, FOXA1 hsa-mir-124a 2.07 2.78 up SDFR1, RXRa hsa-mir-155 down ETS-1 hsa-mir-020 2.00 3.09 up TAL1, SKI, RUNX-1, FLT1, CRK, FOG2, RARB hsa-mir-025 1.98 4.24 up GATA2, hsa-mir-140 down GATA1

PAM selected microRNAs. The fold change of miRNA expression is shown alongside t test score (SAM) and putative targets.

Table 6 Three class analysis showing the different regulated microRNAs among the three cell types: CD34+ progenitors, acute megakaryoblastic leukemia cell lines (AMKL) and in vitro-differentiated megakaryocytes.

TABLE 6 AML In Vitro- M7 cell differentiated Chromosomal CD34+ lines Megakaryocytes microRNA Location Score score Score hsa-mir-010a 17q21 1.0198 0 −0.3562 hsa-mir-101 1p31.3 0 0.814 −0.432 hsa-mir-126 9q34 0.0621 0.4882 −0.4514 hsa-mir-099a 21q21 0 0.4685 −0.2875 hsa-mir-033 22q13 0 0.4258 −0.2294 hsa-mir-095 4p16 0 0.4142 −0.3567 hsa-mir-010b 2q31 0.3308 0 0 hsa-mir-155 21q21 0 −0.3217 0 hsa-mir-130a 11q12 0.2755 0 0 hsa-let-7d 9q22 0.263 −0.274 0 hsa-mir-099b- 21q21 0 0.266 −0.1078 prec hsa-mir-135-2- 12q23 0 0.2279 −0.2566 prec hsa-mir-339 7p22 0 0.2456 −0.1176 hsa-mir-099b 19q13 0 0.2275 −0.1025 hsa-mir-106 xq26 0 0.0575 −0.1891 hsa-let-7c 21q21 0.0289 −0.1753 0 hsa-mir-148 7p15 0 −0.1748 0 hsa-mir-132-prec 17p13 0.1721 0 0 hsa-mir-020 13q31 0 0.0374 −0.1509

There are three patterns of miRNA expression among the three different cell types. The first pattern is defined by miRNA highly expressed in CD34+ cells and downregulated in AMKL and differentiated megakaryocytes. miR-10a and miR-130a follow this pattern of expression; however, miR-10a is upregulated in AMKL relative to differentiated megakaryocytes. The second pattern is miRNA that is upregulated in AMKL, downregulated in CD34+ cells and differentiated megakaryocytes and includes the following miRNAs: miR-126, miR-99, miR-101, let 7A, and miR-100. The last two miRNAs are equally expressed in CD34+ and differentiated megakaryocytes, rather than showing a gradual decline in expression, as evidenced by miR-126, miR-99 and miR-101. The last pattern includes miRNA-106 and miRNA-135-2, which are upregulated in CD34+ cells and AMKL, but low in differentiated megakaryocytes.

MicroRNAs are a highly conserved class of non-coding RNAs with important regulatory functions in proliferation, apoptosis, development and differentiation. As described herein, to discover novel regulatory pathways during megakaryocytic differentiation, we performed microRNA expression profiling of in vitro-differentiated megakaryocytes derived from CD34+ hematopoietic progenitors. One major finding was downregulation of miR-10a, miR-126, miR-106, miR-10b, miR-17 and miR-20. Without wishing to be bound to any theory, it is believed that the downregulation of microRNAs unblocks target genes involved in differentiation. It was confirmed in vitro and in vivo that miR-130a targets the transcription factor MAFB, which is involved in the activation of the GPIIB promoter, a key protein for platelet physiology. In addition, it was shown that miR-10a expression in differentiated megakaryocytes is inverse to that of HOXA1, and HOXA1 is a direct target of miR-10a. Finally, the microRNA expression of megakaryoblastic leukemic cell lines was compared to that of in vitro-differentiated megakaryocytes and CD34+ progenitors. This analysis revealed upregulation of miR-101, miR-126, miR-99a, miR-135, and miR-20 in the cancerous cell line. The data and results described herein delineate the expression of microRNAs during megakaryocytopoiesis and demonstrate a regulatory role of microRNAs in this process by targeting megakaryocytic transcription factors.

The relevant teachings of all publications cited herein that have not explicitly been incorporated by reference, are incorporated herein by reference in their entirety. While this invention has been particularly shown and described with references to preferred embodiments thereof, it will be understood by those skilled in the art that various changes in form and details may be made therein without departing from the scope of the invention encompassed by the appended claims.

While the invention has been described with reference to various and preferred embodiments, it should be understood by those skilled in the art that various changes may be made and equivalents may be substituted for elements thereof without departing from the essential scope of the invention. In addition, many modifications may be made to adapt a particular situation or material to the teachings of the invention without departing from the essential scope thereof. Therefore, it is intended that the invention not be limited to the particular embodiment disclosed herein contemplated for carrying out this invention, but that the invention will include all embodiments falling within the scope of the claims.

Claims

1. A pharmaceutical composition for treating a subject having a cancer and/or a myeloproliferative disorder associated with overexpression of a transcription factor gene product from the Maf family, also known as protooncogene c-Maf or V-maf musculoaponeurotic fibrosarcoma oncogene homolog, (MAFB) gene product,

wherein the cancer and/or myeloproliferative disorder is selected from the group consisting of: acute myeloid leukemia (AML), acute megakaryoblastic leukemia (AMKL), multiple myeloma (MM), and chronic myelogenous leukemia (CML),
the composition comprising an effective amount of at least one miR-130a gene product and a pharmaceutically-acceptable carrier, wherein the at least one miR-130a gene product binds to, and decreases expression of, the MAFB gene product in the subject.

2. The pharmaceutical composition of claim 1, wherein the at least one miR-130a gene product comprises a nucleotide sequence that is complementary to a nucleotide sequence in the MAFB gene product.

3. The pharmaceutical composition of claim 1, wherein the at least one miR-130a gene product comprises a nucleotide sequence that is complementary to at least five nucleotides of the 3′ untranslated region (UTR) of MAFB.

4. The pharmaceutical composition of claim 1, wherein the pharmaceutical composition further comprises at least one anti-cancer agent.

5. The pharmaceutical composition of claim 4, wherein the at least one anti-cancer agent is selected from the group consisting of: cytidine arabinoside, methotrexate, vincristine, etoposide (VP-16), doxorubicin (adriamycin), cisplatin (CDDP), dexamethasone, arglabin, cyclophosphamide, sarcolysin, methylnitrosourea, fluorouracil, 5-fluorouracil (5FU), vinblastine, camptothecin, actinomycin-D, mitomycin C, hydrogen peroxide, oxaliplatin, irinotecan, topotecan, leucovorin, carmustine, streptozocin, CPT-11, taxol, tamoxifen, dacarbazine, rituximab, daunorubicin, 1-β-D-arabinofuranosylcytosine, imatinib, fludarabine, docetaxel, and FOLFOX4.

6. The pharmaceutical composition of claim 1, wherein the subject has been diagnosed with acute myeloid leukemia (AML).

7. The pharmaceutical composition of claim 1, wherein the at least one miR-130a gene product consists of a sequence having at least 90% identity to SEQ ID:136 and/or SEQ ID:383.

8. The pharmaceutical composition of claim 1, wherein the at least one miR-130a gene product comprises a sequence having at least 95% identity to SEQ ID:136 and/or SEQ ID:383.

9. The pharmaceutical composition of claim 1, wherein the wherein the at least one miR-130a gene product comprises one or more chemical modifications selected from the group consisting of: sugar modifications, backbone modifications, and combinations thereof.

10. The pharmaceutical composition of claim 1, wherein the wherein the at least one miR-130a gene product is produced recombinantly.

11. An artificial agonist of miR-130a, wherein the agonist is a polynucleotide comprising a pri-miRNA, pre-miRNA, or mature sequence of miR-130a, and wherein administration of the agonist decreases expression of a MAFB gene product in at least one cell following administration of the agonist.

12. A pharmaceutical composition for treating a subject diagnosed with AML comprising the agonist of claim 11 in a pharmaceutically-acceptable carrier.

13. The pharmaceutical composition of claim 12, wherein the polynucleotide is encapsulated by liposomes.

14. The agonist of claim 11, wherein the polynucleotide is about 18 to about 25 nucleotides in length.

15. The agonist of claim 11, wherein the polynucleotide is double-stranded.

16. The agonist of claim 11, wherein the polynucleotide comprises a sequence of SEQ ID NO: 136 or SEQ ID NO: 383.

17. The agonist of claim 11, wherein the polynucleotide is encoded by an expression vector.

18. The agonist of claim 11, wherein the polynucleotide is nuclease resistant.

Patent History
Publication number: 20140161869
Type: Application
Filed: Jun 25, 2013
Publication Date: Jun 12, 2014
Inventors: Carlo M. Croce (Columbus, OH), Ramiro Garzon (Columbus, OH), George A. Calin (Pearland, TX)
Application Number: 13/925,947
Classifications
Current U.S. Class: Liposomes (424/450); 514/44.00A; Dna Or Rna Fragments Or Modified Forms Thereof (e.g., Genes, Etc.) (536/23.1)
International Classification: C12N 15/113 (20060101);