Pioneering Function of Isl1 in Epigenetic Regulation During Second Heart Field Development

Total Page:16

File Type:pdf, Size:1020Kb

Pioneering Function of Isl1 in Epigenetic Regulation During Second Heart Field Development Pioneering function of Isl1 in epigenetic regulation during second heart field development Dissertation zur Erlangung des Doktorgrades der Naturwissenschaften - Doctor rerum naturalium - (Dr. rer. nat.) eingereicht am Fachbereich Biologie und Chemie der Justus-Liebig-Universität Gießen von Rui Gao Aus Shandong, China 2019 angefertigt am Max-Planck-Institut für Herz- und Lungenforschung, Bad Nauheim und Medizinische Fakultät Mannheim - Universität Heidelberg, Mannheim 1. Gutachter: Prof. Dr. Katja Sträßer Institut für Biochemie, Justus-Liebig-Universität Gießen 35392 Gießen/ Germany 2. Gutachter: Prof. Dr. Gergana Dobreva Medizinische Fakultät Mannheim, Universität Heidelberg 68167 Mannheim/ Germany Disputation am 31.10.2019 EIDESSTATTLICHE ERKLÄRUNG „Ich erkläre: Ich habe die vorgelegte Dissertation selbständig und ohne unerlaubte fremde Hilfe und nur mit den Hilfen angefertigt, die ich in der Dissertation angegeben habe. Alle Textstellen, die wörtlich oder sinngemäß aus veröffentlichten Schriften entnommen sind, und alle Angaben, die auf mündlichen Auskünften beruhen, sind als solche kenntlich gemacht. Bei den von mir durchgeführten und in der Dissertation erwähnten Untersuchungen habe ich die Grundsätze guter wissenschaftlicher Praxis, wie sie in der „Satzung der Justus-Liebig-Universität Gießen zur Sicherung guter wissenschaftlicher Praxis“ niedergelegt sind, eingehalten.“ Mannheim, den Rui Gao Table of Contents List of Figures ................................................................................................. 5 List of Tables .................................................................................................. 7 Zusammenfassung ......................................................................................... 8 Abstract ......................................................................................................... 9 1. Introduction .............................................................................................. 10 1.1 Heart development ................................................................................................ 10 1.1.1 Mouse heart development ...................................................................................... 10 1.1.2 Congenital heart disease .......................................................................................... 14 1.1.3 Cell types and cardiac lineages in mammalian heart .............................................. 17 1.1.4 Signaling pathways in cardiac development ............................................................ 19 1.1.5 Transcriptional factors involved in heart development .......................................... 25 1.2 Isl1 ........................................................................................................................ 31 1.2.1 Protein structure of ISL1 .......................................................................................... 31 1.2.2 Expression and function of Isl1 during development .............................................. 32 1.2.3 Expression and function of Isl1 during heart development .................................... 33 1.2.4 ISL1 and CHD ............................................................................................................ 36 1.3 Pioneer transcription factors .................................................................................. 36 1.4 Epigenetic regulation ............................................................................................. 40 1.4.1 Chromatin structure ................................................................................................. 40 1.4.2 Epigenetic control mechanisms ............................................................................... 41 2. Objectives ................................................................................................. 51 3. Results ...................................................................................................... 52 3.1 Isl1 regulates cardiogenesis in CPCs by directly targeting a large group of downstream targets .................................................................................................... 52 3.1.1 Generation of Isl1 KO ESC line ................................................................................. 52 3.1.2 Establishing in vitro mESC-based cardiac differentiation protocol ......................... 53 3.1.3 Isl1 KO ESC lines can differentiate into CPCs ........................................................... 54 3.1.4 Isl1 loss-of-function results in downregulation of a large number of genes critical for cardiogenesis in CPCs .................................................................................................. 55 3.1.5 Isl1 directly regulates a large amount of genes in CPCs .......................................... 57 1 3.2 Isl1 is essential in regulating second heart field development in mouse embryos .... 58 3.2.1 Isl1 knockout leads to significant gene expression changes at early stage of mouse embryonic development ................................................................................................... 58 3.2.2 Reduced level of Isl1 results in cardiac defects and deregulation of target genes in E10.5 Isl1 hypomorphic embryos ...................................................................................... 60 3.2.3 ChIP-Seq analysis identified Isl1 primary downstream targets in vivo .................... 62 3.3 Isl1 orchestrates a regulatory network driving cardiogenesis .................................. 63 3.4 Baf60c is a key Isl1 downstream target ................................................................... 66 3.4.1 Baf60c is directly regulated by Isl1 .......................................................................... 66 3.4.2 RNA-Seq analysis identified Baf60c regulated genes in CPCs .................................. 67 3.4.3 Isl1 and Baf60c share common targets .................................................................... 69 3.5 Isl1 works in concert with the Brg1-based SWI/SNF complex to regulate its target genes .......................................................................................................................... 70 3.5.1 Isl1 interacts with Brg1 and Baf60c ......................................................................... 70 3.5.2 The interaction of Isl1 with Brg1 does not depend on Baf60c ................................ 71 3.5.3 Co-occupancy of Isl1 and Brg1 in CPCs .................................................................... 72 3.5.4 Isl1 works cooperatively with Brg1 to regulate its target gene expression ............ 74 3.5.5 Isl1 recruits the Brg1 complex to its target sequences ............................................ 75 3.6 Isl1 acts as a pioneer factor in cardiogenesis .......................................................... 76 3.6.1 Isl1 binding correlates with sites of open chromatin .............................................. 76 3.6.2 Isl1 and Brg1/Baf60c complex work synergistically to open the chromatin during SHF development .............................................................................................................. 77 3.6.3 Isl1 plays a pioneering function in all three cardiovascular lineages ...................... 80 3.6.4 Isl1 acts as a pioneer factor independently of Gata4 .............................................. 81 3.7 Working model ...................................................................................................... 83 4. Discussion ................................................................................................. 84 4.1 Isl1 is critical for SHF development and depletion of Isl1 leads to cardiac defects as seen in human CHD patients ........................................................................................ 85 4.2 Isl1 organizes a regulatory gene network driving second heart field development .. 87 4.3 Baf60c is a key downstream target of Isl1 ............................................................... 89 4.4 Isl1 works together with Brg1-Baf60c-based SWI/SNF complex to regulate gene expression program during cardiogenesis .................................................................... 90 4.5 Isl1 functions as a pioneer factor during cardiogenesis ........................................... 91 2 4.6 Isl1 plays a pioneering function independently of Gata4 ......................................... 93 4.7 Conclusions and perspectives ................................................................................. 95 5. Materials and Methods ............................................................................. 97 5.1 Chemicals used in this study .................................................................................. 97 5.2 Kits used in this study ............................................................................................ 98 5.3 Cell culture medium and supplements ................................................................... 99 5.4 Further materials .................................................................................................. 100 5.5 Equipment used in this study ................................................................................ 101 5.6 RNA extraction, cDNA synthesis and qPCR ............................................................ 102 5.6.1 RNA extraction ....................................................................................................... 102 5.6.2 cDNA synthesis
Recommended publications
  • A Genetic Screening Identifies a Component of the SWI/SNF Complex, Arid1b As a Senescence Regulator
    A genetic screening identifies a component of the SWI/SNF complex, Arid1b as a senescence regulator Sadaf Khan A thesis submitted to Imperial College London for the degree of Doctor in Philosophy MRC Clinical Sciences Centre Imperial College London, School of Medicine July 2013 Statement of originality All experiments included in this thesis were performed by myself unless otherwise stated. Copyright Declaration The copyright of this thesis rests with the author and is made available under a Creative Commons Attribution Non-Commercial No Derivatives license. Researchers are free to copy, distribute or transmit the thesis on the condition that they attribute it, that they do not use it for commercial purposes and that they do not alter, transform or build upon it. For any reuse or redistribution, researchers must make clear to others the license terms of this work. 2 Abstract Senescence is an important tumour suppressor mechanism, which prevents the proliferation of stressed or damaged cells. The use of RNA interference to identify genes with a role in senescence is an important tool in the discovery of novel cancer genes. In this work, a protocol was established for conducting bypass of senescence screenings, using shRNA libraries together with next-generation sequencing. Using this approach, the SWI/SNF subunit Arid1b was identified as a regulator of cellular lifespan in MEFs. SWI/SNF is a large multi-subunit complex that remodels chromatin. Mutations in SWI/SNF proteins are frequently associated with cancer, suggesting that SWI/SNF components are tumour suppressors. Here the role of ARID1B during senescence was investigated. Depletion of ARID1B extends the proliferative capacity of primary mouse and human fibroblasts.
    [Show full text]
  • Methylation of the Homeobox Gene, HOPX, Is Frequently Detected in Poorly Differentiated Colorectal Cancer
    ANTICANCER RESEARCH 31: 2889-2892 (2011) Methylation of the Homeobox Gene, HOPX, Is Frequently Detected in Poorly Differentiated Colorectal Cancer YOSHIKUNI HARADA, KAZUHIRO KIJIMA, KAZUKI SHINMURA, MAKIKO SAKATA, KAZUMA SAKURABA, KAZUAKI YOKOMIZO, YOUHEI KITAMURA, ATSUSHI SHIRAHATA, TETSUHIRO GOTO, HIROKI MIZUKAMI, MITSUO SAITO, GAKU KIGAWA, HIROSHI NEMOTO and KENJI HIBI Gastroenterological Surgery, Showa University Fujigaoka Hospital, 1-30 Fujigaoka, Aoba-ku, Yokohama 227-8501, Japan Abstract. Background: Homeodomein only protein x pathogenesis of colorectal cancer (4-8). It is also known that (HOPX) gene methylation has frequently been detected in gene promoter hypermethylation is involved in the cancer tissues. The methylation status of the HOPX gene in development and progression of cancer (9). An investigation colorectal cancer was examined and compared to the of genetic changes is important in order to clarify the clinocopathological findings. Materials and Methods: tumorigenic pathway of colorectal cancer (10). Eighty-nine tumor samples and corresponding normal tissues The homeodomain only protein x (HOPX) gene, also were obtained from colorectal cancer patients who known as NECC1 (not expressed in choriocarcinoma clone underwent surgery at our hospital. The methylation status of 1), LAGY (lung cancer-associated gene Y), and OB1 (odd the HOPX gene in these samples was examined by homeobox 1 protein), was initially identified as an essential quantitative methylation-specific PCR (qMSP). Subsequently, gene for the modulation of cardiac growth and development the clinicopathological findings were correlated with the (11). Three spliced transcript variants, HOPX-α, HOPX-β methylation status of the HOPX gene. Results: HOPX gene and HOPX-γ, encode the same protein, which contains a methylation was found in 46 (52%) out of the 89 colorectal putative homeodomain motif that acts as an adapter protein carcinomas, suggesting that it was frequently observed in to mediate transcriptional repression (12).
    [Show full text]
  • Table S1. List of Proteins in the BAHD1 Interactome
    Table S1. List of proteins in the BAHD1 interactome BAHD1 nuclear partners found in this work yeast two-hybrid screen Name Description Function Reference (a) Chromatin adapters HP1α (CBX5) chromobox homolog 5 (HP1 alpha) Binds histone H3 methylated on lysine 9 and chromatin-associated proteins (20-23) HP1β (CBX1) chromobox homolog 1 (HP1 beta) Binds histone H3 methylated on lysine 9 and chromatin-associated proteins HP1γ (CBX3) chromobox homolog 3 (HP1 gamma) Binds histone H3 methylated on lysine 9 and chromatin-associated proteins MBD1 methyl-CpG binding domain protein 1 Binds methylated CpG dinucleotide and chromatin-associated proteins (22, 24-26) Chromatin modification enzymes CHD1 chromodomain helicase DNA binding protein 1 ATP-dependent chromatin remodeling activity (27-28) HDAC5 histone deacetylase 5 Histone deacetylase activity (23,29,30) SETDB1 (ESET;KMT1E) SET domain, bifurcated 1 Histone-lysine N-methyltransferase activity (31-34) Transcription factors GTF3C2 general transcription factor IIIC, polypeptide 2, beta 110kDa Required for RNA polymerase III-mediated transcription HEYL (Hey3) hairy/enhancer-of-split related with YRPW motif-like DNA-binding transcription factor with basic helix-loop-helix domain (35) KLF10 (TIEG1) Kruppel-like factor 10 DNA-binding transcription factor with C2H2 zinc finger domain (36) NR2F1 (COUP-TFI) nuclear receptor subfamily 2, group F, member 1 DNA-binding transcription factor with C4 type zinc finger domain (ligand-regulated) (36) PEG3 paternally expressed 3 DNA-binding transcription factor with
    [Show full text]
  • Mutant P53 Protects Cells from 12-O-Tetradecanoylphorbol-13- Acetate–Induced Death by Attenuatingactivating Transcription Factor 3 Induction
    Research Article Mutant p53 Protects Cells from 12-O-Tetradecanoylphorbol-13- Acetate–Induced Death by AttenuatingActivating Transcription Factor 3 Induction Yosef Buganim,1 Eyal Kalo,1 Ran Brosh,1 Hila Besserglick,1 Ido Nachmany,3 Yoach Rais,2 Perry Stambolsky,1 Xiaohu Tang,1 Michael Milyavsky,1 Igor Shats,1 Marina Kalis,1 Naomi Goldfinger,1 and Varda Rotter1 1Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel; 2Department of Life Science, Bar-Ilan University, Ramat Gan, Israel; and 3Department of General Surgery B, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel Abstract mutated. Notably, the predominant mode of p53 inactivation is by Mutations in p53 are ubiquitous in human tumors. Some p53 point mutation rather than by deletion or truncation. These data mutations not only result in loss of wild-type (WT) activity but coupled with the observation that mutant p53 is generally highly also grant additional functions, termed ‘‘gain of function.’’ overexpressed in tumors have led to the hypothesis that mutant In this study, we explore how the status of p53 affects the p53 possesses gain-of-function activities. This hypothesis is immediate response gene activating transcription factor 3 supported by the results of in vivo and in vitro studies. For (ATF3) in the 12-O-tetradecanoylphorbol-13-acetate (TPA)- example, mice harboring mutant p53 display allele-specific tumor protein kinase C (PKC) pathway. We show that high doses of spectra, higher metastatic frequency, enhanced cell proliferation, TPA induce ATF3 in a WT p53-independent manner correlat- and higher transformation potential compared with their p53-null ingwith PKCs depletion and cell death.
    [Show full text]
  • Molecular Profile of Tumor-Specific CD8+ T Cell Hypofunction in a Transplantable Murine Cancer Model
    Downloaded from http://www.jimmunol.org/ by guest on September 25, 2021 T + is online at: average * The Journal of Immunology , 34 of which you can access for free at: 2016; 197:1477-1488; Prepublished online 1 July from submission to initial decision 4 weeks from acceptance to publication 2016; doi: 10.4049/jimmunol.1600589 http://www.jimmunol.org/content/197/4/1477 Molecular Profile of Tumor-Specific CD8 Cell Hypofunction in a Transplantable Murine Cancer Model Katherine A. Waugh, Sonia M. Leach, Brandon L. Moore, Tullia C. Bruno, Jonathan D. Buhrman and Jill E. Slansky J Immunol cites 95 articles Submit online. Every submission reviewed by practicing scientists ? is published twice each month by Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts http://jimmunol.org/subscription Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html http://www.jimmunol.org/content/suppl/2016/07/01/jimmunol.160058 9.DCSupplemental This article http://www.jimmunol.org/content/197/4/1477.full#ref-list-1 Information about subscribing to The JI No Triage! Fast Publication! Rapid Reviews! 30 days* Why • • • Material References Permissions Email Alerts Subscription Supplementary The Journal of Immunology The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2016 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. This information is current as of September 25, 2021. The Journal of Immunology Molecular Profile of Tumor-Specific CD8+ T Cell Hypofunction in a Transplantable Murine Cancer Model Katherine A.
    [Show full text]
  • Action on Muscle Metabolism and Insulin Sensitivity E Strong Enough for a Man, Made for a Woman
    Review The impact of ERa action on muscle metabolism and insulin sensitivity e Strong enough for a man, made for a woman Andrea L. Hevener*, Zhenqi Zhou, Timothy M. Moore, Brian G. Drew, Vicent Ribas ABSTRACT Background: The incidence of chronic disease is elevated in women after menopause. Natural variation in muscle expression of the estrogen receptor (ER)a is inversely associated with plasma insulin and adiposity. Moreover, reduced muscle ERa expression levels are observed in women and animals presenting clinical features of the metabolic syndrome (MetSyn). Considering that metabolic dysfunction impacts nearly a quarter of the U.S. adult population and elevates chronic disease risk including type 2 diabetes, heart disease, and certain cancers, treatment strategies to combat metabolic dysfunction and associated pathologies are desperately needed. Scope of the review: This review will provide evidence supporting a critical and protective role for skeletal muscle ERa in the regulation of metabolic homeostasis and insulin sensitivity, and propose novel ERa targets involved in the maintenance of metabolic health. Major conclusions: Studies identifying ERa-regulated pathways essential for disease prevention will lay the important foundation for the rational design of novel therapeutics to improve the metabolic health of women while limiting secondary complications that have plagued traditional hormone replacement interventions. Ó 2018 Published by Elsevier GmbH. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/). Keywords Estrogen action; Estrogen receptors; Insulin sensitivity; Metabolic homeostasis 1. INTRODUCTION new-onset T2DM in postmenopausal women following HRT compared with placebo [7]. The mechanism by which HRT reduces T2D incidence For over two decades researchers have shown strong relationships in postmenopausal women is not yet known however molecular between estrogen action and metabolic health in women.
    [Show full text]
  • ARID1B Is a Specific Vulnerability in ARID1A-Mutant Cancers The
    ARID1B is a specific vulnerability in ARID1A-mutant cancers The Harvard community has made this article openly available. Please share how this access benefits you. Your story matters. Citation Helming, K. C., X. Wang, B. G. Wilson, F. Vazquez, J. R. Haswell, H. E. Manchester, Y. Kim, et al. 2014. “ARID1B is a specific vulnerability in ARID1A-mutant cancers.” Nature medicine 20 (3): 251-254. doi:10.1038/nm.3480. http://dx.doi.org/10.1038/nm.3480. Published Version doi:10.1038/nm.3480 Accessed February 16, 2015 10:04:32 PM EST Citable Link http://nrs.harvard.edu/urn-3:HUL.InstRepos:12987227 Terms of Use This article was downloaded from Harvard University's DASH repository, and is made available under the terms and conditions applicable to Other Posted Material, as set forth at http://nrs.harvard.edu/urn-3:HUL.InstRepos:dash.current.terms-of- use#LAA (Article begins on next page) NIH Public Access Author Manuscript Nat Med. Author manuscript; available in PMC 2014 September 01. NIH-PA Author ManuscriptPublished NIH-PA Author Manuscript in final edited NIH-PA Author Manuscript form as: Nat Med. 2014 March ; 20(3): 251–254. doi:10.1038/nm.3480. ARID1B is a specific vulnerability in ARID1A-mutant cancers Katherine C. Helming1,2,3,4,*, Xiaofeng Wang1,2,3,*, Boris G. Wilson1,2,3, Francisca Vazquez5, Jeffrey R. Haswell1,2,3, Haley E. Manchester1,2,3, Youngha Kim1,2,3, Gregory V. Kryukov5, Mahmoud Ghandi5, Andrew J. Aguirre5,6,7, Zainab Jagani8, Zhong Wang9, Levi A. Garraway6, William C. Hahn6,7, and Charles W.
    [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]
  • Supplemental Materials ZNF281 Enhances Cardiac Reprogramming
    Supplemental Materials ZNF281 enhances cardiac reprogramming by modulating cardiac and inflammatory gene expression Huanyu Zhou, Maria Gabriela Morales, Hisayuki Hashimoto, Matthew E. Dickson, Kunhua Song, Wenduo Ye, Min S. Kim, Hanspeter Niederstrasser, Zhaoning Wang, Beibei Chen, Bruce A. Posner, Rhonda Bassel-Duby and Eric N. Olson Supplemental Table 1; related to Figure 1. Supplemental Table 2; related to Figure 1. Supplemental Table 3; related to the “quantitative mRNA measurement” in Materials and Methods section. Supplemental Table 4; related to the “ChIP-seq, gene ontology and pathway analysis” and “RNA-seq” and gene ontology analysis” in Materials and Methods section. Supplemental Figure S1; related to Figure 1. Supplemental Figure S2; related to Figure 2. Supplemental Figure S3; related to Figure 3. Supplemental Figure S4; related to Figure 4. Supplemental Figure S5; related to Figure 6. Supplemental Table S1. Genes included in human retroviral ORF cDNA library. Gene Gene Gene Gene Gene Gene Gene Gene Symbol Symbol Symbol Symbol Symbol Symbol Symbol Symbol AATF BMP8A CEBPE CTNNB1 ESR2 GDF3 HOXA5 IL17D ADIPOQ BRPF1 CEBPG CUX1 ESRRA GDF6 HOXA6 IL17F ADNP BRPF3 CERS1 CX3CL1 ETS1 GIN1 HOXA7 IL18 AEBP1 BUD31 CERS2 CXCL10 ETS2 GLIS3 HOXB1 IL19 AFF4 C17ORF77 CERS4 CXCL11 ETV3 GMEB1 HOXB13 IL1A AHR C1QTNF4 CFL2 CXCL12 ETV7 GPBP1 HOXB5 IL1B AIMP1 C21ORF66 CHIA CXCL13 FAM3B GPER HOXB6 IL1F3 ALS2CR8 CBFA2T2 CIR1 CXCL14 FAM3D GPI HOXB7 IL1F5 ALX1 CBFA2T3 CITED1 CXCL16 FASLG GREM1 HOXB9 IL1F6 ARGFX CBFB CITED2 CXCL3 FBLN1 GREM2 HOXC4 IL1F7
    [Show full text]
  • UNIVERSITY of CALIFORNIA, IRVINE Combinatorial Regulation By
    UNIVERSITY OF CALIFORNIA, IRVINE Combinatorial regulation by maternal transcription factors during activation of the endoderm gene regulatory network DISSERTATION submitted in partial satisfaction of the requirements for the degree of DOCTOR OF PHILOSOPHY in Biological Sciences by Kitt D. Paraiso Dissertation Committee: Professor Ken W.Y. Cho, Chair Associate Professor Olivier Cinquin Professor Thomas Schilling 2018 Chapter 4 © 2017 Elsevier Ltd. © 2018 Kitt D. Paraiso DEDICATION To the incredibly intelligent and talented people, who in one way or another, helped complete this thesis. ii TABLE OF CONTENTS Page LIST OF FIGURES vii LIST OF TABLES ix LIST OF ABBREVIATIONS X ACKNOWLEDGEMENTS xi CURRICULUM VITAE xii ABSTRACT OF THE DISSERTATION xiv CHAPTER 1: Maternal transcription factors during early endoderm formation in 1 Xenopus Transcription factors co-regulate in a cell type-specific manner 2 Otx1 is expressed in a variety of cell lineages 4 Maternal otx1 in the endodermal conteXt 5 Establishment of enhancers by maternal transcription factors 9 Uncovering the endodermal gene regulatory network 12 Zygotic genome activation and temporal control of gene eXpression 14 The role of maternal transcription factors in early development 18 References 19 CHAPTER 2: Assembly of maternal transcription factors initiates the emergence 26 of tissue-specific zygotic cis-regulatory regions Introduction 28 Identification of maternal vegetally-localized transcription factors 31 Vegt and OtX1 combinatorially regulate the endodermal 33 transcriptome iii
    [Show full text]
  • The GATA2 Transcription Factor Negatively Regulates the Proliferation of Neuronal Progenitors
    RESEARCH ARTICLE 2155 Development 133, 2155-2165 (2006) doi:10.1242/dev.02377 The GATA2 transcription factor negatively regulates the proliferation of neuronal progenitors Abeer El Wakil*, Cédric Francius*,†, Annie Wolff, Jocelyne Pleau-Varet† and Jeannette Nardelli†,§ Postmitotic neurons are produced from a pool of cycling progenitors in an orderly fashion that requires proper spatial and temporal coordination of proliferation, fate determination, differentiation and morphogenesis. This probably relies on complex interplay between mechanisms that control cell cycle, specification and differentiation. In this respect, we have studied the possible implication of GATA2, a transcription factor that is involved in several neuronal specification pathways, in the control of the proliferation of neural progenitors in the embryonic spinal cord. Using gain- and loss-of-function manipulations, we have shown that Gata2 can drive neural progenitors out of the cycle and, to some extent, into differentiation. This correlates with the control of cyclin D1 transcription and of the expression of the p27/Kip1 protein. Interestingly, this functional aspect is not only associated with silencing of the Notch pathway but also appears to be independent of proneural function. Consistently, GATA2 also controls the proliferation capacity of mouse embryonic neuroepithelial cells in culture. Indeed, Gata2 inactivation enhances the proliferation rate in these cells. By contrast, GATA2 overexpression is sufficient to force such cells and neuroblastoma cells to stop dividing but not to drive either type of cell into differentiation. Furthermore, a non-cell autonomous effect of Gata2 expression was observed in vivo as well as in vitro. Hence, our data have provided evidence for the ability of Gata2 to inhibit the proliferation of neural progenitors, and they further suggest that, in this regard, Gata2 can operate independently of neuronal differentiation.
    [Show full text]
  • Genome-Wide DNA Methylation Analysis of KRAS Mutant Cell Lines Ben Yi Tew1,5, Joel K
    www.nature.com/scientificreports OPEN Genome-wide DNA methylation analysis of KRAS mutant cell lines Ben Yi Tew1,5, Joel K. Durand2,5, Kirsten L. Bryant2, Tikvah K. Hayes2, Sen Peng3, Nhan L. Tran4, Gerald C. Gooden1, David N. Buckley1, Channing J. Der2, Albert S. Baldwin2 ✉ & Bodour Salhia1 ✉ Oncogenic RAS mutations are associated with DNA methylation changes that alter gene expression to drive cancer. Recent studies suggest that DNA methylation changes may be stochastic in nature, while other groups propose distinct signaling pathways responsible for aberrant methylation. Better understanding of DNA methylation events associated with oncogenic KRAS expression could enhance therapeutic approaches. Here we analyzed the basal CpG methylation of 11 KRAS-mutant and dependent pancreatic cancer cell lines and observed strikingly similar methylation patterns. KRAS knockdown resulted in unique methylation changes with limited overlap between each cell line. In KRAS-mutant Pa16C pancreatic cancer cells, while KRAS knockdown resulted in over 8,000 diferentially methylated (DM) CpGs, treatment with the ERK1/2-selective inhibitor SCH772984 showed less than 40 DM CpGs, suggesting that ERK is not a broadly active driver of KRAS-associated DNA methylation. KRAS G12V overexpression in an isogenic lung model reveals >50,600 DM CpGs compared to non-transformed controls. In lung and pancreatic cells, gene ontology analyses of DM promoters show an enrichment for genes involved in diferentiation and development. Taken all together, KRAS-mediated DNA methylation are stochastic and independent of canonical downstream efector signaling. These epigenetically altered genes associated with KRAS expression could represent potential therapeutic targets in KRAS-driven cancer. Activating KRAS mutations can be found in nearly 25 percent of all cancers1.
    [Show full text]