Download The

Total Page:16

File Type:pdf, Size:1020Kb

Download The BIOCHEMICAL CHARACTERIZATION AND REGULATION OF TRANSCRIPTION OF POLYCOMB GROUP RING FINGER 5 by Christopher Larson Cochrane B.Sc., The University of British Columbia, 2004 A THESIS SUBMITTED IN PARTIAL FULFILLMENT OF THE REQUIREMENTS FOR THE DEGREE OF DOCTOR OF PHILOSOPHY in THE FACULTY OF GRADUATE AND POSTDOCTORAL STUDIES (Experimental Medicine) THE UNIVERSITY OF BRITISH COLUMBIA (Vancouver) August 2013 © Christopher Larson Cochrane, 2013 Abstract The Polycomb Group (PcG) is a highly conserved group of genes which serve to repress transcription via specific modifications of histones in chromatin. The PcG has well-established roles in development and is involved, by mutation or dysregulation, in many human diseases including cancer. This study identifies the gene PCGF5, which is a paralogue of the oncogene Bmi1, as a transcriptional target of Notch signalling in T cell acute lymphoblastic leukemia (T-ALL). Evidence suggests that this regulation is direct and that the Notch transactivation complex binds DNA at several regions near the PCGF5 gene. PCGF5 is found to be expressed at a higher level in T-ALL than other hematopoietic malignancies. PCGF5 is found to associate with the PcG proteins RING1A and RING1B and its overexpression results in increased ubiquitylation of histone H2A, suggesting it shares functional similarity to Bmi1. Despite their similarities, Bmi1 and PCGF5 have a different spectrum of binding partners and are targeted to different locations in the genome. Overexpression of PCGF5 does not significantly alter hematopoietic development in vivo; however, enforced expression of PCGF5 in bone marrow progenitors results in the generation of fewer colonies in a myeloid colony forming assay. This study suggests that PCGF5 may have as yet unappreciated roles in PcG biology and merits further study into its effects on development and hematopoietic neoplasia. ii Table of Contents Abstract ......................................................................................................................................................... ii List of Tables .............................................................................................................................................. viii List of Figures ............................................................................................................................................... ix Abbreviations ..............................................................................................................................................xiii Acknowledgments ........................................................................................................................................ xx Dedication ................................................................................................................................................... xxi Chapter 1 Introduction ................................................................................................................................. 1 1.1 Rationale ............................................................................................................................................. 1 1.2 The Polycomb Group of genes ............................................................................................................ 2 1.2.1 Genetics ....................................................................................................................................... 2 1.2.2 Polycomb genes in mammals....................................................................................................... 3 1.2.3 Defining the PcG complex ............................................................................................................ 5 1.2.4 Histone modification .................................................................................................................... 9 1.2.5 Targeting and recruitment ......................................................................................................... 11 1.3 Epigenetics and the histone code ..................................................................................................... 12 1.3.1 The epigenetic code ................................................................................................................... 12 1.3.2 Histone modifications ................................................................................................................ 13 1.4 The Notch signalling pathway ........................................................................................................... 17 1.4.1 Notch genetics ........................................................................................................................... 17 iii 1.4.2 The Notch transactivation complex ........................................................................................... 18 1.4.3 Mechanism of signal transduction ............................................................................................. 18 1.4.4 Notch and T cell Acute Lymphoblastic Leukemia ...................................................................... 19 1.4.5 Hypotheses................................................................................................................................. 20 Chapter 2 Materials and methods ............................................................................................................. 23 Chapter 3 Notch signalling regulates expression of PCGF5 in T cell Acute Lymphoblastic Leukemia cells33 3.1 Introduction ...................................................................................................................................... 33 3.2 Results ............................................................................................................................................... 35 3.2.1 Identification of PCGF5 as a target of Notch ............................................................................. 35 3.2.2 Notch activation of PCGF5 expression does not require do novo protein translation .............. 42 3.2.3 Notch binds upstream of the PCGF5 locus ................................................................................ 42 3.2.4 Notch1 and CSL binding of PCGF5 locus is significantly higher than expected by chance ........ 50 3.2.5 PCGF5 expression in T-ALL cases ............................................................................................... 52 3.2.6 Tissue distribution ...................................................................................................................... 55 3.2.7 Splice variants and genomic organization ................................................................................. 58 3.3 Discussion .......................................................................................................................................... 59 3.3.1 Notch transcriptional regulation of PCGF5 ................................................................................ 59 3.3.2 Notch and CSL binding of the PCGF5 locus ................................................................................ 67 3.3.3 Notch expression in cancer ........................................................................................................ 69 3.3.4 PCGF5 and Notch linked in normal development ...................................................................... 70 iv Chapter 4 PCGF5 overexpression does not alter steady state hematopoiesis but results in reduced myeloid colony output by bone marrow progenitors ................................................................................ 72 4.1 Introduction ...................................................................................................................................... 72 4.1.1 The role of Notch in lymphopoiesis ........................................................................................... 72 4.1.2 Notch and T-cell Acute Lymphoblastic Leukemia ...................................................................... 73 4.1.3 The role of Bmi1 and the Polycomb Group in lymphopoiesis ................................................... 74 4.1.4 Experimental approaches .......................................................................................................... 76 4.1.5 Hypotheses................................................................................................................................. 77 4.2 Results ............................................................................................................................................... 77 4.2.1 Overexpression of neither PCGF5 nor Bmi1 causes significant alterations to hematopoiesis in bone marrow transplant recipients .................................................................................................... 77 4.2.2 PCGF5 does not alter lymphoid progenitor performance in OP9 coculture assay .................... 89 4.2.3 PCGF5 causes reduced myeloid CFC output in bone marrow progenitors ................................ 97 4.3 Discussion ........................................................................................................................................ 101 4.3.1 Comment on Experimental Design .......................................................................................... 101 4.3.2 PCGF5 overexpression in bone marrow does not alter engraftment or multilineage reconstitution in transplant recipients ............................................................................................
Recommended publications
  • Specification in Embryonic Stem Cells Through Activation and Repression
    Article Polycomb Regulates Mesoderm Cell Fate- Specification in Embryonic Stem Cells through Activation and Repression Mechanisms Graphical Abstract Authors Lluis Morey, Alexandra Santanach, Enrique Blanco, ..., Elphe` ge P. Nora, Benoit G. Bruneau, Luciano Di Croce Correspondence [email protected] (L.M.), [email protected] (L.D.C.) In Brief Morey et al. reveal that Mel18, a Polycomb-complex-associated protein, is an essential epigenetic regulator of cardiac differentiation. During directed differentiation of embryonic stem cells, Morey et al. find that Mel18-PRC1 complexes exchange subunits in a stage- specific manner and instruct sequential gene activation and repression programs to specify mesoderm fate, prevent alternate lineage commitment, and promote cardiac differentiation. Highlights Accession Numbers d Mel18 is required for PRC1 stability and maintenance of gene GSE67868 repression in ESCs d Mel18 is essential for ESC differentiation into early cardiac- mesoderm precursors d Different Mel18-PRC1 complexes are assembled during cardiac differentiation d Distinctive PRC1 complexes bind to highly active and repressed genes in MES cells Morey et al., 2015, Cell Stem Cell 17, 300–315 September 3, 2015 ª2015 Elsevier Inc. http://dx.doi.org/10.1016/j.stem.2015.08.009 Cell Stem Cell Article Polycomb Regulates Mesoderm Cell Fate-Specification in Embryonic Stem Cells through Activation and Repression Mechanisms Lluis Morey,1,2,6,* Alexandra Santanach,1,2 Enrique Blanco,1,2 Luigi Aloia,1,2 Elphe` ge P. Nora,3,4 Benoit G. Bruneau,3,4
    [Show full text]
  • Noelia Díaz Blanco
    Effects of environmental factors on the gonadal transcriptome of European sea bass (Dicentrarchus labrax), juvenile growth and sex ratios Noelia Díaz Blanco Ph.D. thesis 2014 Submitted in partial fulfillment of the requirements for the Ph.D. degree from the Universitat Pompeu Fabra (UPF). This work has been carried out at the Group of Biology of Reproduction (GBR), at the Department of Renewable Marine Resources of the Institute of Marine Sciences (ICM-CSIC). Thesis supervisor: Dr. Francesc Piferrer Professor d’Investigació Institut de Ciències del Mar (ICM-CSIC) i ii A mis padres A Xavi iii iv Acknowledgements This thesis has been made possible by the support of many people who in one way or another, many times unknowingly, gave me the strength to overcome this "long and winding road". First of all, I would like to thank my supervisor, Dr. Francesc Piferrer, for his patience, guidance and wise advice throughout all this Ph.D. experience. But above all, for the trust he placed on me almost seven years ago when he offered me the opportunity to be part of his team. Thanks also for teaching me how to question always everything, for sharing with me your enthusiasm for science and for giving me the opportunity of learning from you by participating in many projects, collaborations and scientific meetings. I am also thankful to my colleagues (former and present Group of Biology of Reproduction members) for your support and encouragement throughout this journey. To the “exGBRs”, thanks for helping me with my first steps into this world. Working as an undergrad with you Dr.
    [Show full text]
  • Mai Muudatuntuu Ti on Man Mini
    MAIMUUDATUNTUU US009809854B2 TI ON MAN MINI (12 ) United States Patent ( 10 ) Patent No. : US 9 ,809 ,854 B2 Crow et al. (45 ) Date of Patent : Nov . 7 , 2017 Whitehead et al. (2005 ) Variation in tissue - specific gene expression ( 54 ) BIOMARKERS FOR DISEASE ACTIVITY among natural populations. Genome Biology, 6 :R13 . * AND CLINICAL MANIFESTATIONS Villanueva et al. ( 2011 ) Netting Neutrophils Induce Endothelial SYSTEMIC LUPUS ERYTHEMATOSUS Damage , Infiltrate Tissues, and Expose Immunostimulatory Mol ecules in Systemic Lupus Erythematosus . The Journal of Immunol @(71 ) Applicant: NEW YORK SOCIETY FOR THE ogy , 187 : 538 - 552 . * RUPTURED AND CRIPPLED Bijl et al. (2001 ) Fas expression on peripheral blood lymphocytes in MAINTAINING THE HOSPITAL , systemic lupus erythematosus ( SLE ) : relation to lymphocyte acti vation and disease activity . Lupus, 10 :866 - 872 . * New York , NY (US ) Crow et al . (2003 ) Microarray analysis of gene expression in lupus. Arthritis Research and Therapy , 5 :279 - 287 . * @(72 ) Inventors : Mary K . Crow , New York , NY (US ) ; Baechler et al . ( 2003 ) Interferon - inducible gene expression signa Mikhail Olferiev , Mount Kisco , NY ture in peripheral blood cells of patients with severe lupus . PNAS , (US ) 100 ( 5 ) : 2610 - 2615. * GeneCards database entry for IFIT3 ( obtained from < http : / /www . ( 73 ) Assignee : NEW YORK SOCIETY FOR THE genecards. org /cgi - bin / carddisp .pl ? gene = IFIT3 > on May 26 , 2016 , RUPTURED AND CRIPPLED 15 pages ) . * Navarra et al. (2011 ) Efficacy and safety of belimumab in patients MAINTAINING THE HOSPITAL with active systemic lupus erythematosus : a randomised , placebo FOR SPECIAL SURGERY , New controlled , phase 3 trial . The Lancet , 377 :721 - 731. * York , NY (US ) Abramson et al . ( 1983 ) Arthritis Rheum .
    [Show full text]
  • Mouse Pcgf5 Conditional Knockout Project (CRISPR/Cas9)
    https://www.alphaknockout.com Mouse Pcgf5 Conditional Knockout Project (CRISPR/Cas9) Objective: To create a Pcgf5 conditional knockout Mouse model (C57BL/6J) by CRISPR/Cas-mediated genome engineering. Strategy summary: The Pcgf5 gene (NCBI Reference Sequence: NM_029508 ; Ensembl: ENSMUSG00000024805 ) is located on Mouse chromosome 19. 9 exons are identified, with the ATG start codon in exon 2 and the TAG stop codon in exon 9 (Transcript: ENSMUST00000071267). Exon 3 will be selected as conditional knockout region (cKO region). Deletion of this region should result in the loss of function of the Mouse Pcgf5 gene. To engineer the targeting vector, homologous arms and cKO region will be generated by PCR using BAC clone RP24-148L3 as template. Cas9, gRNA and targeting vector will be co-injected into fertilized eggs for cKO Mouse production. The pups will be genotyped by PCR followed by sequencing analysis. Note: Bone marrow cells from mice homozygous for a conditional allele exhibit normal hematopoietic and progenitor cell function. Exon 3 starts from about 15.96% of the coding region. The knockout of Exon 3 will result in frameshift of the gene. The size of intron 2 for 5'-loxP site insertion: 22333 bp, and the size of intron 3 for 3'-loxP site insertion: 2488 bp. The size of effective cKO region: ~597 bp. The cKO region does not have any other known gene. Page 1 of 8 https://www.alphaknockout.com Overview of the Targeting Strategy Wildtype allele gRNA region 5' gRNA region 3' 1 3 9 Targeting vector Targeted allele Constitutive KO allele (After Cre recombination) Legends Exon of mouse Pcgf5 Homology arm cKO region loxP site Page 2 of 8 https://www.alphaknockout.com Overview of the Dot Plot Window size: 10 bp Forward Reverse Complement Sequence 12 Note: The sequence of homologous arms and cKO region is aligned with itself to determine if there are tandem repeats.
    [Show full text]
  • Mammalian PRC1 Complexes: Compositional Complexity and Diverse Molecular Mechanisms
    International Journal of Molecular Sciences Review Mammalian PRC1 Complexes: Compositional Complexity and Diverse Molecular Mechanisms Zhuangzhuang Geng 1 and Zhonghua Gao 1,2,3,* 1 Departments of Biochemistry and Molecular Biology, Penn State College of Medicine, Hershey, PA 17033, USA; [email protected] 2 Penn State Hershey Cancer Institute, Hershey, PA 17033, USA 3 The Stem Cell and Regenerative Biology Program, Penn State College of Medicine, Hershey, PA 17033, USA * Correspondence: [email protected] Received: 6 October 2020; Accepted: 5 November 2020; Published: 14 November 2020 Abstract: Polycomb group (PcG) proteins function as vital epigenetic regulators in various biological processes, including pluripotency, development, and carcinogenesis. PcG proteins form multicomponent complexes, and two major types of protein complexes have been identified in mammals to date, Polycomb Repressive Complexes 1 and 2 (PRC1 and PRC2). The PRC1 complexes are composed in a hierarchical manner in which the catalytic core, RING1A/B, exclusively interacts with one of six Polycomb group RING finger (PCGF) proteins. This association with specific PCGF proteins allows for PRC1 to be subdivided into six distinct groups, each with their own unique modes of action arising from the distinct set of associated proteins. Historically, PRC1 was considered to be a transcription repressor that deposited monoubiquitylation of histone H2A at lysine 119 (H2AK119ub1) and compacted local chromatin. More recently, there is increasing evidence that demonstrates the transcription activation role of PRC1. Moreover, studies on the higher-order chromatin structure have revealed a new function for PRC1 in mediating long-range interactions. This provides a different perspective regarding both the transcription activation and repression characteristics of PRC1.
    [Show full text]
  • Association of the NPAS3 Gene and Five Other Loci with Response to The
    Molecular Psychiatry (2009) 14, 804–819 & 2009 Nature Publishing Group All rights reserved 1359-4184/09 $32.00 www.nature.com/mp ORIGINAL ARTICLE Association of the NPAS3 gene and five other loci with response to the antipsychotic iloperidone identified in a whole genome association study C Lavedan, L Licamele, S Volpi, J Hamilton, C Heaton, K Mack, R Lannan, A Thompson, CD Wolfgang and MH Polymeropoulos Vanda Pharmaceuticals Inc., Rockville, MD, USA A whole genome association study was performed in a phase 3 clinical trial conducted to evaluate a novel antipsychotic, iloperidone, administered to treat patients with schizophrenia. Genotypes of 407 patients were analyzed for 334 563 single nucleotide polymorphisms (SNPs). SNPs associated with iloperidone efficacy were identified within the neuronal PAS domain protein 3 gene (NPAS3), close to a translocation breakpoint site previously observed in a family with schizophrenia. Five other loci were identified that include the XK, Kell blood group complex subunit-related family, member 4 gene (XKR4), the tenascin-R gene (TNR), the glutamate receptor, inotropic, AMPA 4 gene (GRIA4), the glial cell line-derived neurotrophic factor receptor-alpha2 gene (GFRA2), and the NUDT9P1 pseudogene located in the chromosomal region of the serotonin receptor 7 gene (HTR7). The study of these polymorphisms and genes may lead to a better understanding of the etiology of schizophrenia and of its treatment. These results provide new insight into response to iloperidone, developed with the ultimate goal of directing therapy to patients with the highest benefit-to-risk ratio. Molecular Psychiatry (2009) 14, 804–819; doi:10.1038/mp.2008.56; published online 3 June 2008 Keywords: iloperidone; antipsychotic; NPAS3; pharmacogenomics; schizophrenia Introduction the most optimal drug and dosage with less trial and error.
    [Show full text]
  • Mouse Pcgf5 Knockout Project (CRISPR/Cas9)
    https://www.alphaknockout.com Mouse Pcgf5 Knockout Project (CRISPR/Cas9) Objective: To create a Pcgf5 knockout Mouse model (C57BL/6J) by CRISPR/Cas-mediated genome engineering. Strategy summary: The Pcgf5 gene (NCBI Reference Sequence: NM_029508 ; Ensembl: ENSMUSG00000024805 ) is located on Mouse chromosome 19. 9 exons are identified, with the ATG start codon in exon 2 and the TAG stop codon in exon 9 (Transcript: ENSMUST00000071267). Exon 2 will be selected as target site. Cas9 and gRNA will be co-injected into fertilized eggs for KO Mouse production. The pups will be genotyped by PCR followed by sequencing analysis. Note: Bone marrow cells from mice homozygous for a conditional allele exhibit normal hematopoietic and progenitor cell function. Exon 2 starts from the coding region. Exon 2 covers 15.82% of the coding region. The size of effective KO region: ~281 bp. The KO region does not have any other known gene. Page 1 of 9 https://www.alphaknockout.com Overview of the Targeting Strategy Wildtype allele 5' gRNA region gRNA region 3' 1 2 9 Legends Exon of mouse Pcgf5 Knockout region Page 2 of 9 https://www.alphaknockout.com Overview of the Dot Plot (up) Window size: 15 bp Forward Reverse Complement Sequence 12 Note: The 2000 bp section upstream of Exon 2 is aligned with itself to determine if there are tandem repeats. No significant tandem repeat is found in the dot plot matrix. So this region is suitable for PCR screening or sequencing analysis. Overview of the Dot Plot (down) Window size: 15 bp Forward Reverse Complement Sequence 12 Note: The 2000 bp section downstream of Exon 2 is aligned with itself to determine if there are tandem repeats.
    [Show full text]
  • UC San Diego Electronic Theses and Dissertations
    UC San Diego UC San Diego Electronic Theses and Dissertations Title Cardiac Stretch-Induced Transcriptomic Changes are Axis-Dependent Permalink https://escholarship.org/uc/item/7m04f0b0 Author Buchholz, Kyle Stephen Publication Date 2016 Peer reviewed|Thesis/dissertation eScholarship.org Powered by the California Digital Library University of California UNIVERSITY OF CALIFORNIA, SAN DIEGO Cardiac Stretch-Induced Transcriptomic Changes are Axis-Dependent A dissertation submitted in partial satisfaction of the requirements for the degree Doctor of Philosophy in Bioengineering by Kyle Stephen Buchholz Committee in Charge: Professor Jeffrey Omens, Chair Professor Andrew McCulloch, Co-Chair Professor Ju Chen Professor Karen Christman Professor Robert Ross Professor Alexander Zambon 2016 Copyright Kyle Stephen Buchholz, 2016 All rights reserved Signature Page The Dissertation of Kyle Stephen Buchholz is approved and it is acceptable in quality and form for publication on microfilm and electronically: Co-Chair Chair University of California, San Diego 2016 iii Dedication To my beautiful wife, Rhia. iv Table of Contents Signature Page ................................................................................................................... iii Dedication .......................................................................................................................... iv Table of Contents ................................................................................................................ v List of Figures ...................................................................................................................
    [Show full text]
  • A Central Role for Canonical PRC1 in Shaping the 3D Nuclear Landscape
    Downloaded from genesdev.cshlp.org on October 7, 2021 - Published by Cold Spring Harbor Laboratory Press A central role for canonical PRC1 in shaping the 3D nuclear landscape Shelagh Boyle,2 Ilya M. Flyamer,2 Iain Williamson, Dipta Sengupta, Wendy A. Bickmore, and Robert S. Illingworth1 MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, United Kingdom Polycomb group (PcG) proteins silence gene expression by chemically and physically modifying chromatin. A subset of PcG target loci are compacted and cluster in the nucleus; a conformation that is thought to contribute to gene silencing. However, how these interactions influence gross nuclear organization and their relationship with tran- scription remains poorly understood. Here we examine the role of Polycomb-repressive complex 1 (PRC1) in shaping 3D genome organization in mouse embryonic stem cells (mESCs). Using a combination of imaging and Hi-C anal- yses, we show that PRC1-mediated long-range interactions are independent of CTCF and can bridge sites at a megabase scale. Impairment of PRC1 enzymatic activity does not directly disrupt these interactions. We demon- strate that PcG targets coalesce in vivo, and that developmentally induced expression of one of the target loci dis- rupts this spatial arrangement. Finally, we show that transcriptional activation and the loss of PRC1-mediated interactions are separable events. These findings provide important insights into the function of PRC1, while highlighting the complexity of this regulatory system. [Keywords: polycomb; topologically associating domains (TADs); gene repression; nuclear organization; embryonic stem cells; gene regulation; epigenetics; histone modifications] Supplemental material is available for this article.
    [Show full text]
  • Phase Separation Drives X-Chromosome Inactivation
    comment Phase separation drives X-chromosome inactivation: a hypothesis The long non-coding RNA Xist induces heterochromatinization of the X chromosome by recruiting repressive protein complexes to chromatin. Here we gather evidence, from the literature and from computational analyses, showing that Xist assemblies are similar in size, shape and composition to phase-separated condensates, such as paraspeckles and stress granules. Given the progressive sequestration of Xist’s binding partners during X-chromosome inactivation, we formulate the hypothesis that Xist uses phase separation to perform its function. Andrea Cerase, Alexandros Armaos, Christoph Neumayer, Philip Avner, Mitchell Guttman and Gian Gaetano Tartaglia he ability of protein and RNA to with their environment. They form when isolated hydrophobic regions that, exposed interact affects the formation of locally saturated molecules separate into two to the solvent, promote aggregation9. In membrane-less organelles, such as phases, a process known as ‘liquid–liquid RNAs, the presence of nucleotide repetitions T 7 paraspeckles and stress granules, which are phase separation’ (LLPS) . can induce the formation of structures that involved in various biological functions1, LLPS is promoted by proteins carrying attract proteins2,4. For example, the long including RNA processing and responses intrinsically disordered regions (IDRs) non-coding RNA Neat1 uses repeats to to environmental changes2–4. As studied by that lack a specific three-dimensional (3D) sequester specific proteins and
    [Show full text]
  • Exploring the Role of Polycomb Recruitment in Xist-Mediated Silencing of the X Chromosome in ES Cells
    bioRxiv preprint doi: https://doi.org/10.1101/495739; this version posted December 13, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. Exploring the role of Polycomb recruitment in Xist-mediated silencing of the X chromosome in ES cells Aurélie Bousard1*, Ana Cláudia Raposo2*, Jan Jakub Żylicz1,3*, Christel Picard1, Vanessa Borges Pires2,4, Yanyan Qi5, Laurène Syx1, Howard Y. Chang5,6, Edith Heard1†, Simão Teixeira da Rocha1,2† 1 Mammalian Developmental Epigenetics Group, Institut Curie, PSL University, CNRS UMR3215, INSERM U934, 26, rue d'Ulm, 75005 Paris, France. 2 Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649- 028 Lisboa, Portugal. 3 University of Cambridge, Department of Physiology, Development and Neuroscience, Downing Street, Cambridge CB2 3EG, UK. 4 Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal. 5 Center for Dynamic Personal Regulomes, Stanford University, Stanford, CA 94305, USA. 6 Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA. * these authors contributed equally † co-corresponding authors Correspondence: Edith Heard: [email protected] Simão Teixeira da Rocha: [email protected] bioRxiv preprint doi: https://doi.org/10.1101/495739; this version posted December 13, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. Bousard, Raposo, Zylicz et al., Abstract Xist RNA has been established as the master regulator of X-chromosome inactivation (XCI) in female eutherian mammals but its mechanism of action remains unclear.
    [Show full text]
  • Epigenome Alterations in Aortic Valve Stenosis and Its Related Left
    Gošev et al. Clinical Epigenetics (2017) 9:106 DOI 10.1186/s13148-017-0406-7 REVIEW Open Access Epigenome alterations in aortic valve stenosis and its related left ventricular hypertrophy Igor Gošev1, Martina Zeljko2, Željko Đurić3, Ivana Nikolić4, Milorad Gošev5, Sanja Ivčević6, Dino Bešić7, Zoran Legčević7 and Frane Paić7* Abstract Aortic valve stenosis is the most common cardiac valve disease, and with current trends in the population demographics, its prevalence is likely to rise, thus posing a major health and economic burden facing the worldwide societies. Over the past decade, it has become more than clear that our traditional genetic views do not sufficiently explain the well-known link between AS, proatherogenic risk factors, flow-induced mechanical forces, and disease-prone environmental influences. Recent breakthroughs in the field of epigenetics offer us a new perspective on gene regulation, which has broadened our perspective on etiology of aortic stenosis and other aortic valve diseases. Since all known epigenetic marks are potentially reversible this perspective is especially exciting given the potential for development of successful and non-invasive therapeutic intervention and reprogramming of cells at the epigenetic level even in the early stages of disease progression. This review will examine the known relationships between four major epigenetic mechanisms: DNA methylation, posttranslational histone modification, ATP-dependent chromatin remodeling, and non-coding regulatory RNAs, and initiation and progression of AS. Numerous profiling and functional studies indicate that they could contribute to endothelial dysfunctions, disease-prone activation of monocyte-macrophage and circulatory osteoprogenitor cells and activation and osteogenic transdifferentiation of aortic valve interstitial cells, thus leading to valvular inflammation, fibrosis, and calcification, and to pressure overload-induced maladaptive myocardial remodeling and left ventricular hypertrophy.
    [Show full text]