Menin-mediated regulation of mirna biogenesis uncovers the IRS2 pathway as a target for regulating pancreatic beta cells

Similar documents
Lack of mutations of exon 2 of the MEN1 gene in endocrine and nonendocrine sporadic tumors

mirna Dr. S Hosseini-Asl

MicroRNA expression profiling and functional analysis in prostate cancer. Marco Folini s.c. Ricerca Traslazionale DOSL

MicroRNA and Male Infertility: A Potential for Diagnosis

MicroRNA dysregulation in cancer. Systems Plant Microbiology Hyun-Hee Lee

micrornas (mirna) and Biomarkers

(,, ) microrna(mirna) 19~25 nt RNA, RNA mirna mirna,, ;, mirna mirna. : microrna (mirna); ; ; ; : R321.1 : A : X(2015)

High AU content: a signature of upregulated mirna in cardiac diseases

MicroRNAs: Regulatory Function and Potential for Gene Therapy

10/31/2017. micrornas and cancer. From the one gene-one enzyme hypothesis to. microrna DNA RNA. Transcription factors.

MicroRNAs, RNA Modifications, RNA Editing. Bora E. Baysal MD, PhD Oncology for Scientists Lecture Tue, Oct 17, 2017, 3:30 PM - 5:00 PM

he micrornas of Caenorhabditis elegans (Lim et al. Genes & Development 2003)

MicroRNA in Cancer Karen Dybkær 2013

Endocrine Surgery. Characteristics of the Germline MEN1 Mutations in Korea: A Literature Review ORIGINAL ARTICLE. The Korean Journal of INTRODUCTION

Post-transcriptional regulation of an intronic microrna

MicroRNAs: novel regulators in skin research

MicroRNA-mediated incoherent feedforward motifs are robust

Detection of let-7a MicroRNA by Real-time PCR in Colorectal Cancer: a Single-centre Experience from China

Bi 8 Lecture 17. interference. Ellen Rothenberg 1 March 2016

Diagnosis and Management of Multiple Endocrine Neoplasia Type 1 (MEN1)

Emerging Role of MicroRNA in Pancreatic Cancer

Crosstalk between Adiponectin and IGF-IR in breast cancer. Prof. Young Jin Suh Department of Surgery The Catholic University of Korea

MicroRNAs: the primary cause or a determinant of progression in leukemia?

NIH Public Access Author Manuscript Diabetologia. Author manuscript; available in PMC 2014 February 01.

FOR REVIEW. BMB Reports - Manuscript Submission. Manuscript Draft. Manuscript Number: BMB

Neoplasia 18 lecture 6. Dr Heyam Awad MD, FRCPath

Association of mir-21 with esophageal cancer prognosis: a meta-analysis

RUNX1 and FPD/AML Translational Research. The Leukemia and Lymphoma Society / Babich Family Foundation Partnership. September 2016

Downregulation of serum mir-17 and mir-106b levels in gastric cancer and benign gastric diseases

Cancer Problems in Indonesia

Genetics and Genomics in Medicine Chapter 6 Questions

3 The abbreviations used are: EGFR, epidermal growth factor receptor; GAP,

Marta Puerto Plasencia. microrna sponges

Multistep nature of cancer development. Cancer genes

BIO360 Fall 2013 Quiz 1

Epigenetic Principles and Mechanisms Underlying Nervous System Function in Health and Disease Mark F. Mehler MD, FAAN

Early Embryonic Development

V16: involvement of micrornas in GRNs

Deciphering the Role of micrornas in BRD4-NUT Fusion Gene Induced NUT Midline Carcinoma

Ch. 18 Regulation of Gene Expression

RNA interference (RNAi)

The Biology and Genetics of Cells and Organisms The Biology of Cancer

of TERT, MLL4, CCNE1, SENP5, and ROCK1 on tumor development were discussed.

MicroRNAs Modulate Hematopoietic Lineage Differentiation

Low levels of serum mir-99a is a predictor of poor prognosis in breast cancer

Overview: Conducting the Genetic Orchestra Prokaryotes and eukaryotes alter gene expression in response to their changing environment

MicroRNA Cancer qpcr Array

Original Article Expression and importance of serum MicroRNAs in prostate cancer

CRS4 Seminar series. Inferring the functional role of micrornas from gene expression data CRS4. Biomedicine. Bioinformatics. Paolo Uva July 11, 2012

PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland

Profiles of gene expression & diagnosis/prognosis of cancer. MCs in Advanced Genetics Ainoa Planas Riverola

Menin and p53 have non-synergistic effects on tumorigenesis in mice

reported to underlie diverse aspects of biology, including developmental timing, differentiation, proliferation, cell death, and metabolism.

Transcriptional control in Eukaryotes: (chapter 13 pp276) Chromatin structure affects gene expression. Chromatin Array of nuc

Myelodysplastic Syndromes: Hematopathology. Analysis of SHIP1 as a potential biomarker of Disease Progression

Repressive Transcription

Concurrent Type 2 Diabetes and Multiple Endocrine Neoplasia Type 1 With a Novel MEN1 Gene Mutation

Regulation of Gene Expression in Eukaryotes

SUPPLEMENTARY INFORMATION

Are you the way you are because of the

Phenomena first observed in petunia

SUPPLEMENTARY FIGURE LEGENDS

MicroRNA-29a Reveals Oncogenic Role on Myeloid Malignancies by Regulating DNMT3A

Evidence for dietary regulation of microrna expression in cancer cells

PRC2 crystal clear. Matthieu Schapira

Suresh Alahari Editor. MicroRNA in Cancer

microrna Presented for: Presented by: Date:

Epigenetics: Basic Principals and role in health and disease

Decreased expression of mir-490-3p in osteosarcoma and its clinical significance

RNA interference induced hepatotoxicity results from loss of the first synthesized isoform of microrna-122 in mice

Quantification of early stage lesions for loss of p53 should be shown in the main figures.

Refining Prognosis of Early Stage Lung Cancer by Molecular Features (Part 2): Early Steps in Molecularly Defined Prognosis

Problem Set 5 KEY

Hepatitis B virus X gene in the development of hepatocellular carcinoma. Citation Hong Kong Medical Journal, 2011, v. 17 n. 6, suppl. 6, p.

Molecular Mechanism of MicroRNA-203 Mediated Epidermal Stem Cell Differentiation

Morphogens: What are they and why should we care?

Structure and Function of Fusion Gene Products in. Childhood Acute Leukemia

Gene therapy: State of the art. Ramon Gomis MD, PhD, MAE. Hospital Clínic. IDIBAPS. University of Barcelona.

Chapter 11 How Genes Are Controlled

DOES THE BRCAX GENE EXIST? FUTURE OUTLOOK

micrornas as tumor inhibitors, oncogenes, biomarkers for drug efficacy and outcome predictors in lung cancer (Review)

Introduction to Cancer Biology

MicroRNAs and Cancer

The Central Role of Menin and Wild-Type MLL in MLL-AF9-Induced Leukemia

micrornas: the future for cancer therapy?

Association between downexpression of mir-1301 and poor prognosis in patients with glioma

THYROID TUMOR DIAGNOSIS: MARKER OF THE MONTH CLUB

Identification of mirnas in Eucalyptus globulus Plant by Computational Methods

STAT1 regulates microrna transcription in interferon γ stimulated HeLa cells

Introduction. Cancer Biology. Tumor-suppressor genes. Proto-oncogenes. DNA stability genes. Mechanisms of carcinogenesis.

Chapt 15: Molecular Genetics of Cell Cycle and Cancer

Mir-595 is a significant indicator of poor patient prognosis in epithelial ovarian cancer

On the Reproducibility of TCGA Ovarian Cancer MicroRNA Profiles

Insulin Resistance. Biol 405 Molecular Medicine

Menin Epigenetically Represses Hedgehog Signaling in MEN1 Tumor Syndrome

Research Communication

Histones modifications and variants

Yiying Zhang, PhD Research Scientist. Research Summary:

TARGETS OF CYCLIN D1-CDK

HALLA KABAT * Outreach Program, mircore, 2929 Plymouth Rd. Ann Arbor, MI 48105, USA LEO TUNKLE *

Transcription:

2014, Vol.1, No.9 Menin-mediated regulation of mirna biogenesis uncovers the IRS2 pathway as a target for regulating pancreatic beta cells Buddha Gurung 1, Bryson W. Katona 1,2, and Xianxin Hua 1 1 Abramson Family Cancer Research Institute, Department of Cancer Biology, University of Pennsylvania Perelman School of Medicine, 421 Curie Blvd., BRB II/III, Philadelphia, PA-19104, USA. 2 Division of Gastroenterology, University of Pennsylvania Perelman School of Medicine, 421 Curie Blvd., BRB II/III, Philadelphia, PA-19104, USA. Correspondence to: Xianxin Hua, email: huax@mail.med.upenn.edu Keywords: Menin, ARS2, mirna processing, IRS1, Let7a, beta cell Received: September 4, 2014 Accepted: September 8, 2014 Published: September 15, 2014 This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited. ABSTRACT Menin, a protein encoded by the MEN1 gene, is mutated in patients with multiple endocrine neoplasia type 1 (MEN1). Menin acts as a tumor suppressor in endocrine organs while it is also required for transformation of a subgroup of leukemia. The recently solved crystal structure of menin with different binding partners reveals that menin is a key scaffold protein that cross-talks with various partners, including transcription factors, to regulate gene transcription. Our recent findings unravel a previously undiscovered mechanism for menin-mediated control of gene expression via processing of certain microrna s, thus adding to the plethora of ways in which menin regulates gene expression. By interacting with ARS2, an RNA binding protein, menin facilitates the processing of pri-let 7a and pri-mir155 to pre-let 7a and pre-mir155 respectively. Consistently, excision of the Men1 gene results in upregulation of IRS2, a let-7a target. As IRS2 is known to mediate both insulin signaling and insulin-induced cell proliferation, and let-7a targets include oncogenes like RAS and HMGA2, a deeper understanding of the menin-ars2 complex in regulating mirna biogenesis will yield further insights into the pathogenesis of the MEN1 syndrome and other menin-associated malignancies. Multiple endocrine neoplasia type 1 (MEN1) is an inherited syndrome that is characterized by the development of neuroendocrine tumors in the parathyroid glands, pancreatic islet cells, anterior pituitary gland, and other sites such as the duodenum [1]. The gene mutated in this syndrome, MEN1, is located on chromosome 11q13 and encodes the primarily nuclear protein, menin [2]. A germline MEN1 mutation, which is also often detected in sporadic pancreatic neuroendocrine tumors [3], can lead to the development of familial MEN1 in an autosomal dominant fashion. While biochemical tests have suggested a prevalence of the MEN1 syndrome at 0.01 0.175 per thousand [4, 5], the incidence of the syndrome based on genetic testing of the MEN1 gene in a large population is not yet clear. Since the original discovery of the MEN1 gene in 1997 [2], extensive work from multiple groups has been devoted toward understanding the biochemical functions of the MEN1 encoded protein, menin, as well as its underlying mechanisms of action. The recently solved crystal structure of menin demonstrates that it acts as a scaffold protein in regulating gene transcription, cell proliferation, apoptosis and genome stability vis-à-vis its interaction with various protein partners [6-10]. However, the precise mechanism by which menin mediates these functions remains to be further explored. Excellent reviews on the molecular mechanisms whereby menin controls gene transcription, proliferation, and apoptosis have previously been published [11-16]. Defining a novel role for menin as a posttranscriptional regulator of mirna Recently, we identified ARS2 (arsenite-resistance protein-2) as a new menin-binding partner [17]. ARS2 is a component of the nuclear RNA cap-binding complex 562

that stabilizes certain primary microrna (pri-mirna) transcripts for processing by the Microprocessor complex consisting of Drosha and DGCR8, thereby playing an important role in mirna-mediated gene silencing [18, 19]. We further demonstrated that the menin-ars2 complex functionally controls the processing of prilet 7a [17]. This is of importance since the let-7 family of micrornas, which was first discovered in Caenorhabditis elegans as a key developmental regulator, has significantly decreased expression in human cancers and cancer stem cells with elevated levels of oncogenes including RAS and HMGA2 [20-23]. Furthermore, the lin28/let 7 pathway acts as a central regulator of mammalian glucose metabolism and insulin signaling therefore implicating this pathway in diabetes mellitus [24, 25]. To specifically examine processing of pri-let 7a, we demonstrated that acute excision of the Men1 gene results in reduced levels of certain mirnas including let- 7a and mir-155, and this reduction was due to impaired processing of pri-let 7a to pre-let-7a [17]. However, levels of the pri-mirna transcripts, including pri-let7a and pri-mir155, are not affected by menin, indicating that menin-mediated regulation does not occur at the level of transcription. Unlike menin s previously described role in regulating gene expression at the level of gene transcription via epigenetic histone modifications or direct transcriptional activation [26], this is the first report demonstrating menin s role in regulating target gene expression at the post-transcriptional level. Through menin s binding to ARS2, this data also highlights menin s role as a scaffold protein and its versatility in regulating gene expression via association with its multiple binding partners. We subsequently explored the mechanism whereby menin post-transcriptionally regulates the levels of these specific mirnas. In assays using 32 P-radiolabeled prilet7a as the substrate, we found that menin is required for processing of pri-let7a to pre-let7a. However, menin does not affect the levels of Drosha, an RNase III enzyme that executes the initial step of the mirna processing in the nucleus [27]. This finding indicates that menin likely enhances the stability of the pri-let7a transcript and subsequent delivery of the pri-let7a transcript to the catalytic Microprocessor complex. Through targeted gene knockdown analysis, the interaction between menin and ARS2 was found essential for processing of pri-let7a, as Ars2 knockdown reduced mature let-7a levels in meninexpressing cells, but not in menin-null cells. It was recently reported that in addition to its involvement in the biogenesis of mirnas, ARS2 also regulates a number of mrnas, in particular histone-encoding mrnas by contributing to histone mrna 3 end formation and facilitating its cleavage [28]. Given the role of menin in epigenetic regulation of target gene expression is well characterized [7, 9, 29], it would be of interest to evaluate whether menin regulates certain genes by facilitating both modification of histone tails as well as histone expression in conjunction with ARS2. Elucidating the role of menin-mediated let-7a processing in regulating beta cell proliferation and insulin signaling Using prediction algorithms, IRS2, a protein involved in the insulin-signaling pathway, was previously reported as a let-7a target based on sequence complementarity analysis [25]. Furthermore, inhibition of let-7a prevents the down-regulation of IRS2 in the liver of mice on a high-fat diet, highlighting Irs2 as a bona fide target of let-7a [25]. Physiologically, IRS2 has been shown to play a central role in peripheral insulin signaling and pancreatic beta cell proliferation, as knockout of Irs2 in mice results in resistance to insulin and the development of type 2 diabetes [30]. We have previously shown that excision of the Men1 gene ameliorated pre-existing hyperglycemia and increased both glucose-stimulated insulin release and circulating insulin levels in mouse diabetes models [31]. As such, we set to explore a possible link between menin s role in let-7a processing and its overall role in controlling pancreatic beta cell proliferation and function. These studies demonstrate that meninexcision led to increased IRS2 expression. Furthermore, ectopic expression of menin in insulin-producing βhc9 cells, which are derived from pancreatic islets with beta cell hyperplasia, and BON cells, a carcinoid cell line, results in decreased levels of IRS2. Our findings thus suggest that the elevated levels of let-7a in meninexpressing cells targets the Irs2 mrna for degradation, and the excision of the Men1 gene subsequently relieves this targeted mrna degradation, resulting in enhanced IRS2 expression. Additionally, we showed that anti-mirmediated knockdown of let-7a results in a significant increase of IRS2 in menin-expressing cells but not in menin-null cells. These findings clearly indicate that the decreased levels of IRS2 in menin-expressing cells, at least partly, result from the elevated levels of let-7a (Figure 1). In line with decreased levels of let-7 in various tumors, and oncogenes (including RAS) amongst let- 7 target genes, it is plausible that menin s role in tumor suppression can be attributed, in part, to enhanced levels of let-7a biogenesis via its interaction with ARS2 [21, 22]. Accordingly, it has been shown that reduced menin expression is associated with enhanced expression of Ras in human lung adenocarcinoma samples [32]. Examining whether increased Ras expression in lung adenocarcinoma harboring reduced menin levels correlates with diminished let-7a biogenesis might yield some new insight into the pathogenesis of the disease. Furthermore, mir-155, whose biogenesis is also regulated by menin [17], was among the top down-regulated mirna s in pancreatic neuroendocrine 563

Figure 1: A model for menin/ars2-mediated processing of Let-7a in regulating endocrine cell proliferation, insulin signaling, and glucose metabolism. tumors, further implicating the role of mirna s in menindependent repression of cell proliferation [33]. Similarly, the biogenesis of several other micrornas, in addition to let-7a and mir-155, in the presence of menin might collectively contribute to its role in tumor suppression. In this regard, a detailed comparison analysis of differential mirna expression in menin-null cells compared to menin wild-type cells will shed light on the identities of mirnas whose biogenesis is possibly regulated by the menin-ars2 pathway thereby providing deeper insight into menin-mediated repression of cell proliferation. Exploiting the menin-ars2 axis to understand the mechanism of MEN1 tumorigenesis and beta cell regeneration A detailed understanding of the menin-ars2 interaction and its effects on beta cell proliferation via mirna biogenesis might yield valuable insight into the mechanisms by which loss of menin function results in MEN1 tumor syndrome. It has previously been shown that let-7 expression is reduced in a population of human lung cancers, and furthermore its decreased expression is associated with shortened post-operative survival [34]. Similarly, decreased levels of let-7a, in part, could lead to MEN1 tumorigenesis as a result of elevated levels of target oncogenes including RAS. Examining the global levels of let-7a in MEN1 patients would provide important clues as to whether menin-associated loss of let-7a processing plays a role in the pathogenesis of the disease. Importantly, microarray analysis of the expression profile of mirnas in menin-containing and menin-null cells would uncover other mirnas in addition to let-7a and mir-155 whose processing is regulated by the menin-ars2 complex, possibly implicating a larger role for menin-mediated mirna processing in tumorigenesis. It has previously been shown that the global knockdown of let-7 was sufficient to prevent as well as ameliorate impaired glucose tolerance in mice on a high fat diet [25]. As a corollary to menin-ars2 complex repressing Irs2, a key player in regulating beta cell mass and function [35], the discovery of a small molecule that interferes with this binding may hold considerable promise for use in the treatment of type 2 diabetes. Acknowledgements We appreciate support from AACR Care for Carcinoid Foundation (11-60-33XH), the NIH (RO1- CA-113962 and RO1-DK085121 to XH, T32-DK007066 to BK), and the NIH/NIDDK Center for Molecular Studies in Digestive and Liver Diseases (P30DK050306) and its core facilities at the University of Pennsylvania. References 1. Marx SJ. Molecular genetics of multiple endocrine neoplasia types 1 and 2. Nat Rev Cancer. 2005; 5: 367-375. 564

2. Chandrasekharappa SC, Guru SC, Manickam P, Olufemi SE, Collins FS, Emmert-Buck MR, Debelenko LV, Zhuang Z, Lubensky IA, Liotta LA, Crabtree JS, Wang Y, Roe BA, et al. Positional cloning of the gene for multiple endocrine neoplasia-type 1. Science. 1997; 276: 404-407. 3. Lemos MC, Thakker RV. Multiple endocrine neoplasia type 1 (MEN1): analysis of 1336 mutations reported in the first decade following identification of the gene. Hum Mutat. 2008; 29: 22-32. 4. Oberg K, Skogseid B, Eriksson B. Multiple endocrine neoplasia type 1 (MEN-1). Clinical, biochemical and genetical investigations. Acta Oncol. 1989; 28: 383-387. 5. Betts JB, O Malley BP, Rosenthal FD. Hyperparathyroidism: a prerequisite for Zollinger-Ellison syndrome in multiple endocrine adenomatosis Type 1 report of a further family and a review of th literature. Q J Med. 1980; 49: 69-76. 6. Agarwal SK, Guru SC, Heppner C, Erdos MR, Collins RM, Park SY, Saggar S, Chandrasekharappa SC, Collins FS, Spiegel AM, Marx SJ, Burns AL. Menin interacts with the AP1 transcription factor JunD and represses JunD-activated transcription. Cell. 1999; 96: 143-152. 7. Hughes CM, Rozenblatt-Rosen O, Milne TA, Copeland TD, Levine SS, Lee JC, Hayes DN, Shanmugam KS, Bhattacharjee A, Biondi CA, Kay GF, Hayward NK, Hess JL, et al. Menin associates with a trithorax family histone methyltransferase complex and with the hoxc8 locus. Mol Cell. 2004; 13: 587-597. 8. Yang Y, Hua X. In search of tumor suppressing functions of menin. Mol Cell Endocrinol. 2007; 265-266: 34-41. 9. Gurung B, Feng Z, Iwamoto DV, Thiel A, Jin G, Fan CM, Ng JM, Curran T, Hua X. Menin epigenetically represses Hedgehog signaling in MEN1 tumor syndrome. Cancer Res. 2013; 73: 2650-2658. 10. Huang J, Gurung B, Wan B, Matkar S, Veniaminova NA, Wan K, Merchant JL, Hua X, Lei M. The same pocket in menin binds both MLL and JUND but has opposite effects on transcription. Nature. 2012; 482: 542-546. 11. Thiel AT, Huang J, Lei M, Hua X. Menin as a hub controlling mixed lineage leukemia. Bioessays. 2012; 34: 771-780. 12. Thakker RV. Multiple endocrine neoplasia type 1 (MEN1). Best Pract Res Clin Endocrinol Metab. 24: 355-370. 13. Agarwal SK, Kennedy PA, Scacheri PC, Novotny EA, Hickman AB, Cerrato A, Rice TS, Moore JB, Rao S, Ji Y, Mateo C, Libutti SK, Oliver B, et al. Menin molecular interactions: insights into normal functions and tumorigenesis. Horm Metab Res. 2005; 37: 369-374. 14. Matkar S, Thiel A, Hua X. Menin: a scaffold protein that controls gene expression and cell signaling. Trends Biochem Sci. 38: 394-402. 15. Wu X, Hua X. Menin, histone h3 methyltransferases, and regulation of cell proliferation: current knowledge and perspective. Curr Mol Med. 2008; 8: 805-815. 16. Marx SJ, Stratakis CA. Multiple endocrine neoplasia introduction. J Intern Med. 2005; 257: 2-5. 17. Gurung B, Muhammad AB, Hua X. Menin Is Required for Optimal Processing of the MicroRNA let-7a. J Biol Chem. 2014; 289: 9902-9908. 18. Gruber JJ, Zatechka DS, Sabin LR, Yong J, Lum JJ, Kong M, Zong WX, Zhang Z, Lau CK, Rawlings J, Cherry S, Ihle JN, Dreyfuss G, et al. Ars2 links the nuclear cap-binding complex to RNA interference and cell proliferation. Cell. 2009; 138: 328-339. 19. Sabin LR, Zhou R, Gruber JJ, Lukinova N, Bambina S, Berman A, Lau CK, Thompson CB, Cherry S. Ars2 regulates both mirna and sirna dependent silencing and suppresses RNA virus infection in Drosophila. Cell. 2009; 138: 340-351. 20. Reinhart BJ, Slack FJ, Basson M, Pasquinelli AE, Bettinger JC, Rougvie AE, Horvitz HR, Ruvkun G. The 21-nucleotide let-7 RNA regulates developmental timing in Caenorhabditis elegans. Nature. 2000; 403: 901-906. 21. Esquela-Kerscher A, Slack FJ. Oncomirs micrornas with a role in cancer. Nat Rev Cancer. 2006; 6: 259-269. 22. Johnson SM, Grosshans H, Shingara J, Byrom M, Jarvis R, Cheng A, Labourier E, Reinert KL, Brown D, Slack FJ. RAS is regulated by the let-7 microrna family. Cell. 2005; 120: 635-647. 23. Mayr C, Hemann MT, Bartel DP. Disrupting the pairing between let-7 and Hmga2 enhances oncogenic transformation. Science. 2007; 315: 1576-1579. 24. Zhu H, Shyh-Chang N, Segre AV, Shinoda G, Shah SP, Einhorn WS, Takeuchi A, Engreitz JM, Hagan JP, Kharas MG, Urbach A, Thornton JE, Triboulet R, et al. The Lin28/ let-7 axis regulates glucose metabolism. Cell. 147: 81-94. 25. Frost RJ, Olson EN. Control of glucose homeostasis and insulin sensitivity by the Let-7 family of micrornas. Proc Natl Acad Sci U S A. 2011; 108: 21075-21080. 26. Luzi E, Marini F, Tognarini I, Galli G, Falchetti A, Brandi ML. The regulatory network menin-microrna 26a as a possible target for RNA-based therapy of bone diseases. Nucleic Acid Ther. 22: 103-108. 27. Lee Y, Ahn C, Han J, Choi H, Kim J, Yim J, Lee J, Provost P, Radmark O, Kim S, Kim VN. The nuclear RNase III Drosha initiates microrna processing. Nature. 2003; 425: 415-419. 28. Gruber JJ, Olejniczak SH, Yong J, La Rocca G, Dreyfuss G, Thompson CB. Ars2 promotes proper replication-dependent histone mrna 3 end formation. Mol Cell. 45: 87-98. 29. Thiel AT, Blessington P, Zou T, Feather D, Wu X, Yan J, Zhang H, Liu Z, Ernst P, Koretzky GA, Hua X. MLL-AF9- induced leukemogenesis requires coexpression of the wildtype Mll allele. Cancer Cell. 2010; 17: 148-159. 30. Withers DJ, Gutierrez JS, Towery H, Burks DJ, Ren JM, Previs S, Zhang Y, Bernal D, Pons S, Shulman GI, Bonner- Weir S, White MF. Disruption of IRS-2 causes type 2 diabetes in mice. Nature. 1998; 391: 900-904. 565

31. Yang Y, Gurung B, Wu T, Wang H, Stoffers DA, Hua X. Reversal of preexisting hyperglycemia in diabetic mice by acute deletion of the Men1 gene. Proc Natl Acad Sci U S A. 2010; 107: 20358-20363. 32. Wu Y, Feng ZJ, Gao SB, Matkar S, Xu B, Duan HB, Lin X, Li SH, Hua X, Jin GH. Interplay between menin and K-Ras in regulating lung adenocarcinoma. J Biol Chem. 287: 40003-40011. 33. Roldo C, Missiaglia E, Hagan JP, Falconi M, Capelli P, Bersani S, Calin GA, Volinia S, Liu CG, Scarpa A, Croce CM. MicroRNA expression abnormalities in pancreatic endocrine and acinar tumors are associated with distinctive pathologic features and clinical behavior. J Clin Oncol. 2006; 24: 4677-4684. 34. Takamizawa J, Konishi H, Yanagisawa K, Tomida S, Osada H, Endoh H, Harano T, Yatabe Y, Nagino M, Nimura Y, Mitsudomi T, Takahashi T. Reduced expression of the let- 7 micrornas in human lung cancers in association with shortened postoperative survival. Cancer Res. 2004; 64: 3753-3756. 35. Kubota N, Terauchi Y, Tobe K, Yano W, Suzuki R, Ueki K, Takamoto I, Satoh H, Maki T, Kubota T, Moroi M, Okada- Iwabu M, Ezaki O, et al. Insulin receptor substrate 2 plays a crucial role in beta cells and the hypothalamus. J Clin Invest. 2004; 114: 917-927. 566