Thromb Haemost 2019; 119(05): 716-725
DOI: 10.1055/s-0039-1678694
Cellular Haemostasis and Platelets
Georg Thieme Verlag KG Stuttgart · New York

Identification of the Regulatory Elements and Target Genes of Megakaryopoietic Transcription Factor MEF2C

Xianguo Kong
1   Department of Medicine, Cardeza Foundation for Hematologic Research, Sidney Kimmel Medical School at Thomas Jefferson University, Philadelphia, Pennsylvania, United States
,
Lin Ma
1   Department of Medicine, Cardeza Foundation for Hematologic Research, Sidney Kimmel Medical School at Thomas Jefferson University, Philadelphia, Pennsylvania, United States
,
Edward Chen
2   Department of Human & Molecular Genetics, Baylor College of Medicine, Houston, Texas, United States
,
Chad A. Shaw
2   Department of Human & Molecular Genetics, Baylor College of Medicine, Houston, Texas, United States
3   Department of Statistics, Rice University, Houston, Texas, United States
,
Leonard C. Edelstein
1   Department of Medicine, Cardeza Foundation for Hematologic Research, Sidney Kimmel Medical School at Thomas Jefferson University, Philadelphia, Pennsylvania, United States
› Author Affiliations
Funding This work was funded by a grant from the National Institute of Health (HL128234).
Further Information

Publication History

26 July 2018

06 January 2019

Publication Date:
07 February 2019 (online)

Abstract

Megakaryopoiesis produces specialized haematopoietic stem cells in the bone marrow that give rise to megakaryocytes which ultimately produce platelets. Defects in megakaryopoiesis can result in altered platelet counts and physiology, leading to dysfunctional haemostasis and thrombosis. Additionally, dysregulated megakaryopoiesis is also associated with myeloid pathologies. Transcription factors play critical roles in cell differentiation by regulating the temporal and spatial patterns of gene expression which ultimately decide cell fate. Several transcription factors have been described as regulating megakaryopoiesis including myocyte enhancer factor 2C (MEF2C); however, the genes regulated by MEF2C that influence megakaryopoiesis have not been reported. Using chromatin immunoprecipitation-sequencing and Gene Ontology data we identified five candidate genes that are bound by MEF2C and regulate megakaryopoiesis: MOV10, AGO3, HDAC1, RBBP5 and WASF2. To study expression of these genes, we silenced MEF2C gene expression in the Meg01 megakaryocytic cell line and in induced pluripotent stem cells by CRISPR/Cas9 editing. We also knocked down MEF2C expression in cord blood-derived haematopoietic stem cells by siRNA. We found that absent or reduced MEF2C expression resulted in defects in megakaryocytic differentiation and reduced levels of the candidate target genes. Luciferase assays confirmed that genomic sequences within the target genes are regulated by MEF2C levels. Finally, we demonstrate that small deletions linked to a platelet count-associated single nucleotide polymorphism alter transcriptional activity, suggesting a mechanism by which genetic variation in MEF2C alters platelet production. These data help elucidate the mechanism behind MEF2C regulation of megakaryopoiesis and genetic variation driving platelet production.

Supplementary Material

 
  • References

  • 1 Gieger C, Radhakrishnan A, Cvejic A. , et al. New gene functions in megakaryopoiesis and platelet formation. Nature 2011; 480 (7376): 201-208
  • 2 Astle WJ, Elding H, Jiang T. , et al. The allelic landscape of human blood cell trait variation and links to common complex disease. Cell 2016; 167 (05) 1415.e19-1429.e19
  • 3 Eicher JD, Chami N, Kacprowski T. , et al; Global Lipids Genetics Consortium; CARDIoGRAM Exome Consortium; Myocardial Infarction Genetics Consortium. Platelet-related variants identified by exomechip meta-analysis in 157,293 individuals. Am J Hum Genet 2016; 99 (01) 40-55
  • 4 Schick UM, Jain D, Hodonsky CJ. , et al. genome-wide association study of platelet count identifies ancestry-specific loci in Hispanic/Latino Americans. Am J Hum Genet 2016; 98 (02) 229-242
  • 5 Vannucchi AM, Barbui T. Thrombocytosis and thrombosis. Hematology (Am Soc Hematol Educ Program) 2007; 363-370
  • 6 Pujol-Moix N, Vázquez-Santiago M, Morera A. , et al. Genetic determinants of platelet large-cell ratio, immature platelet fraction, and other platelet-related phenotypes. Thromb Res 2015; 136 (02) 361-366
  • 7 Buckley MF, James JW, Brown DE. , et al. A novel approach to the assessment of variations in the human platelet count. Thromb Haemost 2000; 83 (03) 480-484
  • 8 Daly ME. Transcription factor defects causing platelet disorders. Blood Rev 2017; 31 (01) 1-10
  • 9 Tijssen MR, Ghevaert C. Transcription factors in late megakaryopoiesis and related platelet disorders. J Thromb Haemost 2013; 11 (04) 593-604
  • 10 Songdej N, Rao AK. Hematopoietic transcription factor mutations: important players in inherited platelet defects. Blood 2017; 129 (21) 2873-2881
  • 11 Canté-Barrett K, Pieters R, Meijerink JP. Myocyte enhancer factor 2C in hematopoiesis and leukemia. Oncogene 2014; 33 (04) 403-410
  • 12 Fuhrken PG, Chen C, Apostolidis PA, Wang M, Miller WM, Papoutsakis ET. Gene ontology-driven transcriptional analysis of CD34+ cell-initiated megakaryocytic cultures identifies new transcriptional regulators of megakaryopoiesis. Physiol Genomics 2008; 33 (02) 159-169
  • 13 Schüler A, Schwieger M, Engelmann A. , et al. The MADS transcription factor Mef2c is a pivotal modulator of myeloid cell fate. Blood 2008; 111 (09) 4532-4541
  • 14 Gekas C, Rhodes KE, Gereige LM. , et al. Mef2C is a lineage-restricted target of Scl/Tal1 and regulates megakaryopoiesis and B-cell homeostasis. Blood 2009; 113 (15) 3461-3471
  • 15 Stehling-Sun S, Dade J, Nutt SL, DeKoter RP, Camargo FD. Regulation of lymphoid versus myeloid fate ‘choice’ by the transcription factor Mef2c. Nat Immunol 2009; 10 (03) 289-296
  • 16 Laszlo GS, Alonzo TA, Gudgeon CJ. , et al. High expression of myocyte enhancer factor 2C (MEF2C) is associated with adverse-risk features and poor outcome in pediatric acute myeloid leukemia: a report from the Children's Oncology Group. J Hematol Oncol 2015; 8: 115
  • 17 Maguire JA, Gagne AL, Jobaliya CD, Gandre-Babbe S, Gadue P, French DL. Generation of human control iPS cell line CHOPWT10 from healthy adult peripheral blood mononuclear cells. Stem Cell Res (Amst) 2016; 16 (02) 338-341
  • 18 Mills JA, Paluru P, Weiss MJ, Gadue P, French DL. Hematopoietic differentiation of pluripotent stem cells in culture. Methods Mol Biol 2014; 1185: 181-194
  • 19 Edelstein LC, Simon LM, Montoya RT. , et al. Racial differences in human platelet PAR4 reactivity reflect expression of PCTP and miR-376c. Nat Med 2013; 19 (12) 1609-1616
  • 20 Chiang TW, le Sage C, Larrieu D, Demir M, Jackson SP. CRISPR-Cas9(D10A) nickase-based genotypic and phenotypic screening to enhance genome editing. Sci Rep 2016; 6: 24356
  • 21 Ogura M, Morishima Y, Okumura M. , et al. Functional and morphological differentiation induction of a human megakaryoblastic leukemia cell line (MEG-01s) by phorbol diesters. Blood 1988; 72 (01) 49-60
  • 22 Becker PB. ; ENCODE Project Consortium. A user's guide to the encyclopedia of DNA elements (ENCODE). PLoS Biol 2011; 9 (04) e1001046
  • 23 Li Q, Brown JB, Huang H, Bickel PJ. Measuring reproducibility of high-throughput experiments. Ann Appl Stat 2011; 5 (03) 1752-1779
  • 24 Tarumoto Y, Lu B, Somerville TDD. , et al. LKB1, salt-inducible kinases, and MEF2C are linked dependencies in acute myeloid leukemia. Mol Cell 2018; 69 (06) 1017.e6-1027.e6
  • 25 Khan A, Fornes O, Stigliani A. , et al. JASPAR 2018: update of the open-access database of transcription factor binding profiles and its web framework. Nucleic Acids Res 2018; 46 (D1): D260-D266
  • 26 Fernández JM, de la Torre V, Richardson D. , et al; BLUEPRINT Consortium. The BLUEPRINT data analysis portal. Cell Syst 2016; 3 (05) 491.e5-495.e5
  • 27 Westra HJ, Peters MJ, Esko T. , et al. Systematic identification of trans eQTLs as putative drivers of known disease associations. Nat Genet 2013; 45 (10) 1238-1243
  • 28 Romanoski CE, Glass CK, Stunnenberg HG, Wilson L, Almouzni G. Epigenomics: Roadmap for regulation. Nature 2015; 518 (7539): 314-316
  • 29 Schwieger M, Schüler A, Forster M. , et al. Homing and invasiveness of MLL/ENL leukemic cells is regulated by MEF2C. Blood 2009; 114 (12) 2476-2488
  • 30 Black BL, Olson EN. Transcriptional control of muscle development by myocyte enhancer factor-2 (MEF2) proteins. Annu Rev Cell Dev Biol 1998; 14: 167-196
  • 31 Krivtsov AV, Twomey D, Feng Z. , et al. Transformation from committed progenitor to leukaemia stem cell initiated by MLL-AF9. Nature 2006; 442 (7104): 818-822
  • 32 Norfo R, Zini R, Pennucci V. , et al; Associazione Italiana per la Ricerca sul Cancro Gruppo Italiano Malattie Mieloproliferative Investigators. miRNA-mRNA integrative analysis in primary myelofibrosis CD34+ cells: role of miR-155/JARID2 axis in abnormal megakaryopoiesis. Blood 2014; 124 (13) e21-e32
  • 33 Meister G, Landthaler M, Peters L. , et al. Identification of novel argonaute-associated proteins. Curr Biol 2005; 15 (23) 2149-2155
  • 34 Fabian MR, Sonenberg N, Filipowicz W. Regulation of mRNA translation and stability by microRNAs. Annu Rev Biochem 2010; 79: 351-379
  • 35 Edelstein LC, McKenzie SE, Shaw C, Holinstat MA, Kunapuli SP, Bray PF. MicroRNAs in platelet production and activation. J Thromb Haemost 2013; 11 (Suppl. 01) 340-350
  • 36 Liu J, Carmell MA, Rivas FV. , et al. Argonaute2 is the catalytic engine of mammalian RNAi. Science 2004; 305 (5689): 1437-1441
  • 37 Wu L, Fan J, Belasco JG. Importance of translation and nonnucleolytic ago proteins for on-target RNA interference. Curr Biol 2008; 18 (17) 1327-1332
  • 38 Wang F, Yu J, Yang GH, Wang XS, Zhang JW. Regulation of erythroid differentiation by miR-376a and its targets. Cell Res 2011; 21 (08) 1196-1209
  • 39 Ben-Ami O, Pencovich N, Lotem J, Levanon D, Groner Y. A regulatory interplay between miR-27a and Runx1 during megakaryopoiesis. Proc Natl Acad Sci U S A 2009; 106 (01) 238-243
  • 40 Meier K, Brehm A. Chromatin regulation: how complex does it get?. Epigenetics 2014; 9 (11) 1485-1495
  • 41 Heideman MR, Lancini C, Proost N, Yanover E, Jacobs H, Dannenberg JH. Sin3a-associated Hdac1 and Hdac2 are essential for hematopoietic stem cell homeostasis and contribute differentially to hematopoiesis. Haematologica 2014; 99 (08) 1292-1303
  • 42 Wada T, Kikuchi J, Nishimura N, Shimizu R, Kitamura T, Furukawa Y. Expression levels of histone deacetylases determine the cell fate of hematopoietic progenitors. J Biol Chem 2009; 284 (44) 30673-30683
  • 43 Hu X, Li X, Valverde K. , et al. LSD1-mediated epigenetic modification is required for TAL1 function and hematopoiesis. Proc Natl Acad Sci U S A 2009; 106 (25) 10141-10146
  • 44 Guo H, Friedman AD. Phosphorylation of RUNX1 by cyclin-dependent kinase reduces direct interaction with HDAC1 and HDAC3. J Biol Chem 2011; 286 (01) 208-215
  • 45 Shinsky SA, Hu M, Vought VE. , et al. A non-active-site SET domain surface crucial for the interaction of MLL1 and the RbBP5/Ash2L heterodimer within MLL family core complexes. J Mol Biol 2014; 426 (12) 2283-2299
  • 46 Huang G, Zhao X, Wang L. , et al. The ability of MLL to bind RUNX1 and methylate H3K4 at PU.1 regulatory regions is impaired by MDS/AML-associated RUNX1/AML1 mutations. Blood 2011; 118 (25) 6544-6552
  • 47 Eto K, Nishikii H, Ogaeri T. , et al. The WAVE2/Abi1 complex differentially regulates megakaryocyte development and spreading: implications for platelet biogenesis and spreading machinery. Blood 2007; 110 (10) 3637-3647
  • 48 Ogaeri T, Eto K, Otsu M, Ema H, Nakauchi H. The actin polymerization regulator WAVE2 is required for early bone marrow repopulation by hematopoietic stem cells. Stem Cells 2009; 27 (05) 1120-1129
  • 49 Lambert L, Bienvenu T, Allou L. , et al. MEF2C mutations are a rare cause of Rett or severe Rett-like encephalopathies. Clin Genet 2012; 82 (05) 499-501
  • 50 Orozco G, Barrett JC, Zeggini E. Synthetic associations in the context of genome-wide association scan signals. Hum Mol Genet 2010; 19 (R2): R137-R144