The Mef2b Regulatory Network

Total Page:16

File Type:pdf, Size:1020Kb

The Mef2b Regulatory Network THE MEF2B REGULATORY NETWORK by Julia Pon B.Sc., The University of Alberta, 2010 A THESIS SUBMITTED IN PARTIAL FULFILLMENT OF THE REQUIREMENTS FOR THE DEGREE OF DOCTOR OF PHILOSOPHY in The Faculty of Graduate and Postdoctoral Studies (Genome Science and Technology) THE UNIVERSITY OF BRITISH COLUMBIA (Vancouver) June 2015 © Julia Pon, 2015 Abstract Myocyte enhancer factor 2B (MEF2B) is a transcription factor with somatic mutation hotspots at K4, Y69 and D83 in diffuse large B-cell lymphoma (DLBCL) and follicular lymphoma. The recurrence of these mutations indicates that they may drive lymphoma development. However, inferring the mechanisms by which they may drive lymphoma development was complicated by our limited understanding of MEF2B’s normal functions. To expand our understanding of the cellular activities of wildtype and mutant MEF2B, I developed and addressed two hypotheses: (1) identifying genes regulated by wildtype MEF2B will allow identification of cellular phenotypes affected by MEF2B activity and (2) contrasting the DNA binding sites, effects on gene expression and effects on cellular phenotypes of mutant and wildtype MEF2B will indicate mechanisms through which MEF2B mutations may contribute to lymphoma development. To address these hypotheses, I first identified genome-wide MEF2B binding sites and transcriptome-wide gene expression changes mediated by MEF2B. Using these data I identified and validated novel MEF2B target genes. I found that target genes of MEF2B included the cancer genes MYC, TGFB1, CARD11, NDRG1, RHOB, BCL2 and JUN. The identification of target genes led to findings that MEF2B promotes expression of mesenchymal markers, promotes HEK293A cell migration, and inhibits DLBCL cell chemotaxis. I then investigated how K4E, Y69H and D83V mutations change MEF2B’s activity. I found that K4E, Y69H and D83V mutations decreased MEF2B’s capacity to promote gene expression in both HEK293A and DLBCL cells. These mutations also reduced MEF2B’s capacity to alter HEK293A and DLBCL cell movement. Overall, these data support the concept that MEF2B mutations may promote lymphoma development by reducing expression of MEF2B target genes that would otherwise function to help confine germinal centre B-cells to germinal centres. My research demonstrates how observations from genome-scale data can aid in the functional characterization of candidate driver mutations. Moreover, my work provides a unique resource for exploring the role of MEF2B in cell biology. I map for the first time the MEF2B regulome, demonstrating connections between a relatively understudied transcription factor and genes significant to oncogenesis. ii Preface Portions of Chapter 1 have been published: J. Pon, M.A. Marra. Driver and Passenger Mutations in Cancer. Annu. Rev. Pathology: Mechanisms of Disease. 10:25-50, 2014. © by Annual Reviews, http://www.annualreviews.org. I wrote most of the text for this review with guidance from M.A. Marra. Figure 1.5 was produced by Jianghong An. Portions of Chapters 2-5 have been accepted for publication pending formatting: J.R. Pon, J. Wong, S. Saberi, O. Alder, M. Moksa, S.W.G. Cheng, G.B. Morin, P.A. Hoodless, M. Hirst, M.A. Marra. MEF2B Mutations in Non-Hodgkin Lymphoma Dysregulate Cell Migration by Decreasing Transcriptional Activation of MEF2B Target Genes. Nature Communications. I designed and performed the research, analyzed and interpreted data, and drafted the manuscript. J.W. produced data in Figures 5.1b and 5.2b and optimized protein detection. S.S. ran MACS2 peak calling and IDR analysis for ChIP-seq data shown in Figure 3.9b. O.A. optimized conditions for gel shift assays. M.M. performed ChIP library construction. RNA-seq library construction, sequencing, alignment of sequence data, and the production of quality control metrics was performed by staff of the British Columbia Genome Sciences Centre. S.W.G.C. and G.B.M. provided advice on gel shift assays and purification of MEF2B-V5 expressed in E. coli. P.A.H. provided advice on gel shift assays and contributed to research design. M.H. advised and co-ordinated ChIP-seq data collection. M.A.M. conceived of the research questions and participated in research design and data interpretation. Dr. Suganthi Chittaranjan also provided guidance on experimental design and data presentation. Dr. Ryan Morin identified the MEF2C mutations reported in Table 1.2. Data in Figure 4.2 and the empty vector cell line was produced by Katie O’Brien, and data in Figure 5.1d was produced by Annie Moradian, Jessica Tamura- Wells and Marlo Firme. iii Table of Contents Abstract .......................................................................................................................................... ii Preface ........................................................................................................................................... iii Table of Contents ......................................................................................................................... iv List of Tables ................................................................................................................................ ix List of Figures ................................................................................................................................ x List of Abbreviations ................................................................................................................. xiii Acknowledgements ..................................................................................................................... xv Dedication .................................................................................................................................. xvii Chapter 1: Transcriptional Dysregulation in Cancer and the Potential Role of MEF2B Mutations as Drivers of Non-Hodgkin Lymphoma ................................................................... 1 1.1 Introduction ........................................................................................................................... 1 1.2 Cancer biology ...................................................................................................................... 1 1.3 Regulation of transcription by chromatin structure…………………………………………3 1.4 Non-Hodgkin lymphomas ..................................................................................................... 5 1.4.1 Clinical characteristics of DLBCL, FL and MCL .......................................................... 6 1.4.2 The cellular origin of DLBCL, FL and MCL................................................................. 7 1.4.3 Driver genes of FL, DLBCL and MCL .......................................................................... 9 1.5 Technologies and model systems for characterizing transcription factors ......................... 10 1.5.1 Technologies for identifying transcription factor target genes .................................... 10 1.5.2 Distinguishing direct and indirect target genes ............................................................ 12 1.5.3 Target gene identification as a means of characterizing how transcription factor filller th mutations may contribute to lymphoma development .......................................................... 13 1.5.4 Model systems for examining cancer gene function .................................................... 14 1.6 MEF2 family proteins ......................................................................................................... 16 1.6.1 Roles of MEF2 proteins in vertebrate organisms ......................................................... 16 iv 1.6.2 Expression patterns of MEF2B .................................................................................... 17 1.6.3 Target genes of MEF2 family proteins ........................................................................ 18 1.6.4 Functions of the MADS and MEF2 domains in MEF2 proteins ................................. 19 1.6.5 Functions of the transactivation domains of MEF2 proteins ....................................... 22 1.7 Roles of MEF2 family proteins in human disease .............................................................. 23 1.7.1 MEF2B may act as an oncogene in some types of carcinoma ..................................... 24 1.7.2 MEF2B may act as a tumor suppressor in some types of non-Hodgkin lymphoma .... 24 1.8 Thesis roadmap and chapter summaries.............................................................................. 28 Chapter 2: Materials and Methods ........................................................................................... 40 2.1 Production of stably transfected HEK293A cell lines ........................................................ 40 2.2 Production of stably transduced DoHH2 cells .................................................................... 40 2.3 Cell culture and treatments .................................................................................................. 41 2.4 MEF2-dependent luciferase reporter assay ......................................................................... 41 2.5 Expression microarrays and qRT-PCR validation .............................................................. 42 2.6 RNA-seq library construction ............................................................................................. 43 2.7 Differential expression analysis using HEK293A RNA-seq data ....................................... 44 2.8 Chromatin immunoprecipitation (ChIP) for sequencing
Recommended publications
  • REPORT Germline Mutation of INI1/SMARCB1 in Familial Schwannomatosis
    REPORT Germline Mutation of INI1/SMARCB1 in Familial Schwannomatosis Theo J. M. Hulsebos, Astrid S. Plomp, Ruud A. Wolterman, Els C. Robanus-Maandag, Frank Baas, and Pieter Wesseling Patients with schwannomatosis develop multiple schwannomas but no vestibular schwannomas diagnostic of neurofi- bromatosis type 2. We report an inactivating germline mutation in exon 1 of the tumor-suppressor gene INI1 in a father and daughter who both had schwannomatosis. Inactivation of the wild-type INI1 allele, by a second mutation in exon 5 or by clear loss, was found in two of four investigated schwannomas from these patients. All four schwannomas displayed complete loss of nuclear INI1 protein expression in part of the cells. Although the exact oncogenetic mechanism in these schwannomas remains to be elucidated, our findings suggest that INI1 is the predisposing gene in familial schwannomatosis. Schwannomatosis (MIM 162091) is characterized by the 10 Only two families with an INI1 germline mutation, an development of multiple spinal, peripheral, and cranial- exon 4 frameshift mutation,11 and an exon 7 donor splice nerve schwannomas in the absence of vestibular schwan- site mutation12 have been described in which multiple nomas.1 The presence of vestibular schwannomas is di- generations were affected by malignant (rhabdoid) tumors agnostic of neurofibromatosis type 2 (NF2 [MIM 101000]). in infancy. In both of these families, clear cases of no- Molecular analyses identified somatically acquired mu- nexpressing obligate carriers of the INI1 mutation were
    [Show full text]
  • A Computational Approach for Defining a Signature of Β-Cell Golgi Stress in Diabetes Mellitus
    Page 1 of 781 Diabetes A Computational Approach for Defining a Signature of β-Cell Golgi Stress in Diabetes Mellitus Robert N. Bone1,6,7, Olufunmilola Oyebamiji2, Sayali Talware2, Sharmila Selvaraj2, Preethi Krishnan3,6, Farooq Syed1,6,7, Huanmei Wu2, Carmella Evans-Molina 1,3,4,5,6,7,8* Departments of 1Pediatrics, 3Medicine, 4Anatomy, Cell Biology & Physiology, 5Biochemistry & Molecular Biology, the 6Center for Diabetes & Metabolic Diseases, and the 7Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202; 2Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202; 8Roudebush VA Medical Center, Indianapolis, IN 46202. *Corresponding Author(s): Carmella Evans-Molina, MD, PhD ([email protected]) Indiana University School of Medicine, 635 Barnhill Drive, MS 2031A, Indianapolis, IN 46202, Telephone: (317) 274-4145, Fax (317) 274-4107 Running Title: Golgi Stress Response in Diabetes Word Count: 4358 Number of Figures: 6 Keywords: Golgi apparatus stress, Islets, β cell, Type 1 diabetes, Type 2 diabetes 1 Diabetes Publish Ahead of Print, published online August 20, 2020 Diabetes Page 2 of 781 ABSTRACT The Golgi apparatus (GA) is an important site of insulin processing and granule maturation, but whether GA organelle dysfunction and GA stress are present in the diabetic β-cell has not been tested. We utilized an informatics-based approach to develop a transcriptional signature of β-cell GA stress using existing RNA sequencing and microarray datasets generated using human islets from donors with diabetes and islets where type 1(T1D) and type 2 diabetes (T2D) had been modeled ex vivo. To narrow our results to GA-specific genes, we applied a filter set of 1,030 genes accepted as GA associated.
    [Show full text]
  • The H3.3 Chaperone Hira Complex Orchestrates Oocyte Developmental Competence
    bioRxiv preprint doi: https://doi.org/10.1101/2020.05.25.114124; this version posted May 26, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. The H3.3 chaperone Hira complex orchestrates oocyte developmental competence Rowena Smith1, *, Zongliang Jiang2, *, Andrej Susor3, Hao Ming2, Janet Tait1, Marco Conti4, and Chih-Jen Lin1,5 1MRC Centre For Reproductive Health, University oF Edinburgh Queen’s Medical Research Institute 47 Little France Crescent, Edinburgh, United Kingdom, EH16 4TJ 2 School oF Animal Sciences, AgCenter, Louisiana State University, Baton Rouge, LA, 70803, USA 3 Laboratory oF Biochemistry and Molecular Biology oF Germ Cells, Institute oF Animal Physiology and Genetics, CAS, Rumburska 89, 277 21 Libechov, Czech Republic 4Center For Reproductive Sciences, University oF CaliFornia, San Francisco, CA 94143, USA 5To whom correspondence should be addressed. Tel: +44 131 242 6237; Email: [email protected] *equal contribution bioRxiv preprint doi: https://doi.org/10.1101/2020.05.25.114124; this version posted May 26, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. Abstract Reproductive success relies on a healthy oocyte competent For Fertilisation and capable of sustaining early embryo development. By the end oF oogenesis, the oocyte is characterised by a transcriptionally silenced state, but the signiFicance oF this state and how it is achieved remains poorly understood.
    [Show full text]
  • 4-6 Weeks Old Female C57BL/6 Mice Obtained from Jackson Labs Were Used for Cell Isolation
    Methods Mice: 4-6 weeks old female C57BL/6 mice obtained from Jackson labs were used for cell isolation. Female Foxp3-IRES-GFP reporter mice (1), backcrossed to B6/C57 background for 10 generations, were used for the isolation of naïve CD4 and naïve CD8 cells for the RNAseq experiments. The mice were housed in pathogen-free animal facility in the La Jolla Institute for Allergy and Immunology and were used according to protocols approved by the Institutional Animal Care and use Committee. Preparation of cells: Subsets of thymocytes were isolated by cell sorting as previously described (2), after cell surface staining using CD4 (GK1.5), CD8 (53-6.7), CD3ε (145- 2C11), CD24 (M1/69) (all from Biolegend). DP cells: CD4+CD8 int/hi; CD4 SP cells: CD4CD3 hi, CD24 int/lo; CD8 SP cells: CD8 int/hi CD4 CD3 hi, CD24 int/lo (Fig S2). Peripheral subsets were isolated after pooling spleen and lymph nodes. T cells were enriched by negative isolation using Dynabeads (Dynabeads untouched mouse T cells, 11413D, Invitrogen). After surface staining for CD4 (GK1.5), CD8 (53-6.7), CD62L (MEL-14), CD25 (PC61) and CD44 (IM7), naïve CD4+CD62L hiCD25-CD44lo and naïve CD8+CD62L hiCD25-CD44lo were obtained by sorting (BD FACS Aria). Additionally, for the RNAseq experiments, CD4 and CD8 naïve cells were isolated by sorting T cells from the Foxp3- IRES-GFP mice: CD4+CD62LhiCD25–CD44lo GFP(FOXP3)– and CD8+CD62LhiCD25– CD44lo GFP(FOXP3)– (antibodies were from Biolegend). In some cases, naïve CD4 cells were cultured in vitro under Th1 or Th2 polarizing conditions (3, 4).
    [Show full text]
  • Global Mef2 Target Gene Analysis in Skeletal and Cardiac Muscle
    GLOBAL MEF2 TARGET GENE ANALYSIS IN SKELETAL AND CARDIAC MUSCLE STEPHANIE ELIZABETH WALES A DISSERTATION SUBMITTED TO THE FACULTY OF GRADUATE STUDIES IN PARTIAL FULFILLMENT OF THE REQUIREMENTS FOR THE DEGREE OF DOCTOR OF PHILOSOPHY GRADUATE PROGRAM IN BIOLOGY YORK UNIVERSITY TORONTO, ONTARIO FEBRUARY 2016 © Stephanie Wales 2016 ABSTRACT A loss of muscle mass or function occurs in many genetic and acquired pathologies such as heart disease, sarcopenia and cachexia which are predominantly found among the rapidly increasing elderly population. Developing effective treatments relies on understanding the genetic networks that control these disease pathways. Transcription factors occupy an essential position as regulators of gene expression. Myocyte enhancer factor 2 (MEF2) is an important transcription factor in striated muscle development in the embryo, skeletal muscle maintenance in the adult and cardiomyocyte survival and hypertrophy in the progression to heart failure. We sought to identify common MEF2 target genes in these two types of striated muscles using chromatin immunoprecipitation and next generation sequencing (ChIP-seq) and transcriptome profiling (RNA-seq). Using a cell culture model of skeletal muscle (C2C12) and primary cardiomyocytes we found 294 common MEF2A binding sites within both cell types. Individually MEF2A was recruited to approximately 2700 and 1600 DNA sequences in skeletal and cardiac muscle, respectively. Two genes were chosen for further study: DUSP6 and Hspb7. DUSP6, an ERK1/2 specific phosphatase, was negatively regulated by MEF2 in a p38MAPK dependent manner in striated muscle. Furthermore siRNA mediated gene silencing showed that MEF2D in particular was responsible for repressing DUSP6 during C2C12 myoblast differentiation. Using a p38 pharmacological inhibitor (SB 203580) we observed that MEF2D must be phosphorylated by p38 to repress DUSP6.
    [Show full text]
  • Control Association Analysis of CABIN1 with Schizophrenia in a Japanese Population
    Journal of Human Genetics (2010) 55, 179–181 & 2010 The Japan Society of Human Genetics All rights reserved 1434-5161/10 $32.00 www.nature.com/jhg SHORT COMMUNICATION A case–control association analysis of CABIN1 with schizophrenia in a Japanese population Yuichiro Watanabe1,2, Ayako Nunokawa1, Naoshi Kaneko1 and Toshiyuki Someya1 Calcineurin (CN) is a calcium/calmodulin-dependent serine/threonine protein phosphatase and regulates neuronal structure, neurotransmission and activity-dependent gene expression. Several studies have indicated that CN signaling is likely to be involved in the pathogenesis of schizophrenia. The gene encoding CN-binding protein 1 (CABIN1) is located on 22q11.23, one of the common susceptibility loci for schizophrenia. Therefore, CABIN1 is a promising functional and positional candidate gene for schizophrenia. To assess whether CABIN1 is implicated in vulnerability to schizophrenia, we conducted a case–control association study between CABIN1 and schizophrenia. The results showed no evidence of an association between CABIN1 and schizophrenia using 11 tagging single nucleotide polymorphisms in 1193 Japanese subjects. Our results suggest that CABIN1 may not confer increased susceptibility for schizophrenia in the Japanese population. Journal of Human Genetics (2010) 55, 179–181; doi:10.1038/jhg.2009.136; published online 15 January 2010 Keywords: CABIN1; case–control study; schizophrenia; tagging SNP Schizophrenia is a complex genetic disorder that affects approximately one study. Fallin et al.10 examined seven polymorphisms in CABIN1 1% of the global population. The pathogenesis of schizophrenia is with an average density of one marker per 20.9 kb and failed to find currently unclear, but there is cumulative evidence that calcineurin any association with schizophrenia.
    [Show full text]
  • Molecular-Genetic Mechanisms of Memory Formation in Mouse Models of Neurodevelopmental and Neuropsychiatric Disorders
    University of Pennsylvania ScholarlyCommons Publicly Accessible Penn Dissertations 2014 Molecular-Genetic Mechanisms of Memory Formation in Mouse Models of Neurodevelopmental and Neuropsychiatric Disorders Hannah Schoch University of Pennsylvania, [email protected] Follow this and additional works at: https://repository.upenn.edu/edissertations Part of the Neuroscience and Neurobiology Commons Recommended Citation Schoch, Hannah, "Molecular-Genetic Mechanisms of Memory Formation in Mouse Models of Neurodevelopmental and Neuropsychiatric Disorders" (2014). Publicly Accessible Penn Dissertations. 1435. https://repository.upenn.edu/edissertations/1435 This paper is posted at ScholarlyCommons. https://repository.upenn.edu/edissertations/1435 For more information, please contact [email protected]. Molecular-Genetic Mechanisms of Memory Formation in Mouse Models of Neurodevelopmental and Neuropsychiatric Disorders Abstract Neurodevelopmental and neuropsychiatric disorders are a significant and expanding global health crisis. Many individuals affected by these disorders have social and cognitive symptoms represent significant sources of ongoing disability that are refractory to available treatment options. The search for cures and therapies for disorders fundamentally requires an understanding of the core neuropathology and insight into the underlying molecular mechanisms at work. In this dissertation, I describe experiments that we performed to explore molecular and genetic mechanisms underlying memory impairment and enhancement
    [Show full text]
  • Whole-Exome Sequencing of 81 Individuals from 27 Multiply Affected Bipolar Disorder Families Andreas J
    Forstner et al. Translational Psychiatry (2020) 10:57 https://doi.org/10.1038/s41398-020-0732-y Translational Psychiatry ARTICLE Open Access Whole-exome sequencing of 81 individuals from 27 multiply affected bipolar disorder families Andreas J. Forstner1,2,3,4, Sascha B. Fischer 3,5,LorenaM.Schenk2,JanaStrohmaier 6,7, Anna Maaser-Hecker2, Céline S. Reinbold3,5,8, Sugirthan Sivalingam 2, Julian Hecker9,FabianStreit 6, Franziska Degenhardt2, Stephanie H. Witt 6, Johannes Schumacher1,2, Holger Thiele10, Peter Nürnberg10, José Guzman-Parra11, Guillermo Orozco Diaz12, Georg Auburger13,MargotAlbus14, Margitta Borrmann-Hassenbach14,MariaJoséGonzaleź 11, Susana Gil Flores15, Francisco J. Cabaleiro Fabeiro16, Francisco del Río Noriega17, Fermin Perez Perez18, Jesus Haro González19,FabioRivas20, Fermin Mayoral20,MichaelBauer 21, Andrea Pfennig21, Andreas Reif 22, Stefan Herms 2,3,5, Per Hoffmann2,3,5,23, Mehdi Pirooznia24, Fernando S. Goes 24, Marcella Rietschel 6, Markus M. Nöthen2 and Sven Cichon2,3,5,23 Abstract Bipolar disorder (BD) is a highly heritable neuropsychiatric disease characterized by recurrent episodes of depression and mania. Research suggests that the cumulative impact of common alleles explains 25–38% of phenotypic variance, and that rare variants may contribute to BD susceptibility. To identify rare, high-penetrance susceptibility variants for BD, whole-exome sequencing (WES) was performed in three affected individuals from each of 27 multiply affected families from Spain and Germany. WES identified 378 rare, non-synonymous, and potentially functional variants. These spanned 368 genes, and were carried by all three affected members in at least one family. Eight of the 368 genes harbored rare variants that were implicated in at least two independent families.
    [Show full text]
  • Ubinuclein-1 Confers Histone H3.3-Specific-Binding by the HIRA
    ARTICLE Received 22 Apr 2015 | Accepted 1 Jun 2015 | Published 10 Jul 2015 DOI: 10.1038/ncomms8711 OPEN Ubinuclein-1 confers histone H3.3-specific-binding by the HIRA histone chaperone complex M. Daniel Ricketts1,2, Brian Frederick3, Henry Hoff3, Yong Tang3, David C. Schultz3, Taranjit Singh Rai4,5, Maria Grazia Vizioli4, Peter D. Adams4 & Ronen Marmorstein1,2,6 Histone chaperones bind specific histones to mediate their storage, eviction or deposition from/or into chromatin. The HIRA histone chaperone complex, composed of HIRA, ubinuclein-1 (UBN1) and CABIN1, cooperates with the histone chaperone ASF1a to mediate H3.3-specific binding and chromatin deposition. Here we demonstrate that the conserved UBN1 Hpc2-related domain (HRD) is a novel H3.3-specific-binding domain. Biochemical and biophysical studies show the UBN1-HRD preferentially binds H3.3/H4 over H3.1/H4. X-ray crystallographic and mutational studies reveal that conserved residues within the UBN1-HRD and H3.3 G90 as key determinants of UBN1–H3.3-binding specificity. Comparison of the structure with the unrelated H3.3-specific chaperone DAXX reveals nearly identical points of contact between the chaperone and histone in the proximity of H3.3 G90, although the mechanism for H3.3 G90 recognition appears to be distinct. This study points to UBN1 as the determinant of H3.3-specific binding and deposition by the HIRA complex. 1 Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA. 2 Graduate Group in Biochemistry and Molecular Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.
    [Show full text]
  • Discovery and Systematic Characterization of Risk Variants and Genes For
    medRxiv preprint doi: https://doi.org/10.1101/2021.05.24.21257377; this version posted June 2, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. It is made available under a CC-BY 4.0 International license . 1 Discovery and systematic characterization of risk variants and genes for 2 coronary artery disease in over a million participants 3 4 Krishna G Aragam1,2,3,4*, Tao Jiang5*, Anuj Goel6,7*, Stavroula Kanoni8*, Brooke N Wolford9*, 5 Elle M Weeks4, Minxian Wang3,4, George Hindy10, Wei Zhou4,11,12,9, Christopher Grace6,7, 6 Carolina Roselli3, Nicholas A Marston13, Frederick K Kamanu13, Ida Surakka14, Loreto Muñoz 7 Venegas15,16, Paul Sherliker17, Satoshi Koyama18, Kazuyoshi Ishigaki19, Bjørn O Åsvold20,21,22, 8 Michael R Brown23, Ben Brumpton20,21, Paul S de Vries23, Olga Giannakopoulou8, Panagiota 9 Giardoglou24, Daniel F Gudbjartsson25,26, Ulrich Güldener27, Syed M. Ijlal Haider15, Anna 10 Helgadottir25, Maysson Ibrahim28, Adnan Kastrati27,29, Thorsten Kessler27,29, Ling Li27, Lijiang 11 Ma30,31, Thomas Meitinger32,33,29, Sören Mucha15, Matthias Munz15, Federico Murgia28, Jonas B 12 Nielsen34,20, Markus M Nöthen35, Shichao Pang27, Tobias Reinberger15, Gudmar Thorleifsson25, 13 Moritz von Scheidt27,29, Jacob K Ulirsch4,11,36, EPIC-CVD Consortium, Biobank Japan, David O 14 Arnar25,37,38, Deepak S Atri39,3, Noël P Burtt4, Maria C Costanzo4, Jason Flannick40, Rajat M 15 Gupta39,3,4, Kaoru Ito18, Dong-Keun Jang4,
    [Show full text]
  • A Mef2 Gene That Generates a Muscle-Specific Isoform Via Alternative Mrna Splicing JAMES F
    MOLECULAR AND CELLULAR BIOLOGY, Mar. 1994, p. 1647-1656 Vol. 14, No. 3 0270-7306/94/$04.00 + 0 Copyright C) 1994, American Society for Microbiology A Mef2 Gene That Generates a Muscle-Specific Isoform via Alternative mRNA Splicing JAMES F. MARTIN,' JOSEPH M. MIANO,' CAROLYN M. HUSTAD,2 NEAL G. COPELAND,2 NANCY A. JENKINS,2 AND ERIC N. OLSON'* Department of Biochemistry and Molecular Biology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030,1 and Mammalian Genetics Laboratory, ABL-Basic Research Program, NCI-Frederick Cancer Research and Development Center, Frederick, Maryland 217022 Received 21 July 1993/Returned for modification 30 September 1993/Accepted 2 December 1993 Members of the myocyte-specific enhancer-binding factor 2 (MEF2) family of transcription factors bind a conserved A/T-rich sequence in the control regions of numerous muscle-specific genes. Mammalian MEF2 proteins have been shown previously to be encoded by three genes, MeQt, xMeJ2, and Me.2c, each of which gives rise to multiple alternatively spliced transcripts. We describe the cloning of a new member of the MEF2 family from mice, termed MEF2D, which shares extensive homology with other MEF2 proteins but is the product of a separate gene. MEF2D binds to and activates transcription through the MEF2 site and forms heterodimers with other members of the MEF2 family. Deletion mutations show that the carboxyl terminus of MEF2D is required for efficient transactivation. MEF2D transcripts are widely expressed, but alternative splicing of MEF2D transcripts gives rise to a muscle-specific isoform which is induced during myoblast differentiation.
    [Show full text]
  • Morphology Regulation in Vascular Endothelial Cells Kiyomi Tsuji-Tamura1,2* and Minetaro Ogawa1
    Tsuji-Tamura and Ogawa Inflammation and Regeneration (2018) 38:25 Inflammation and Regeneration https://doi.org/10.1186/s41232-018-0083-8 REVIEW Open Access Morphology regulation in vascular endothelial cells Kiyomi Tsuji-Tamura1,2* and Minetaro Ogawa1 Abstract Morphological change in endothelial cells is an initial and crucial step in the process of establishing a functional vascular network. Following or associated with differentiation and proliferation, endothelial cells elongate and assemble into linear cord-like vessels, subsequently forming a perfusable vascular tube. In vivo and in vitro studies have begun to outline the underlying genetic and signaling mechanisms behind endothelial cell morphology regulation. This review focuses on the transcription factors and signaling pathways regulating endothelial cell behavior, involved in morphology, during vascular development. Keywords: Vasculature, Endothelial cells, Angiogenesis, Morphology, Elongation Background known as the hemogenic endothelium reportedly gener- Vascular system development ates hematopoietic stem cells directly [3, 7–10]. During the earliest stages of embryonic development, Specification of angioblasts to either arterial or venous vascular formation occurs in connection with blood cell endothelial cells is established prior to forming blood formation (hematopoiesis) [1, 2]. There are various the- vessel structures [11–13]. The receptor tyrosine kinase ories about the origin of endothelial cells, but the meso- EphB4 and its transmembrane ligand ephrinB2 are derm has been reported to generate an endothelial cell demonstrated to be significant factors for arteriovenous progenitor (angioblast) and a common progenitor of definition [14]. The binding of vascular endothelial hematopoietic cells and endothelial cells (hemangioblast) growth factor (VEGF) to its receptor VEGFR2, also [3] (Fig. 1).
    [Show full text]