H3K27 Acetylation and Gene Expression Analysis Reveals Differences in Placental Chromatin Activity in Fetal Growth Restriction N

Total Page:16

File Type:pdf, Size:1020Kb

H3K27 Acetylation and Gene Expression Analysis Reveals Differences in Placental Chromatin Activity in Fetal Growth Restriction N Paauw et al. Clinical Epigenetics (2018) 10:85 https://doi.org/10.1186/s13148-018-0508-x RESEARCH Open Access H3K27 acetylation and gene expression analysis reveals differences in placental chromatin activity in fetal growth restriction N. D. Paauw1,6*, A. T. Lely1, J. A. Joles2, A. Franx1, P. G. Nikkels3, M. Mokry4 and B. B. van Rijn1,5,6* Abstract Background: Posttranslational modification of histone tails such as histone 3 lysine 27 acetylation (H3K27ac) is tightly coupled to epigenetic regulation of gene expression. To explore whether this is involved in placenta pathology, we probed genome-wide H3K27ac occupancy by chromatin immunoprecipitation sequencing (ChIP- seq) in healthy placentas and placentas from pathological pregnancies with fetal growth restriction (FGR). Furthermore, we related specific acetylation profiles of FGR placentas to gene expression changes. Results: Analysis of H3K27ac occupancy in FGR compared to healthy placentas showed 970 differentially acetylated regions distributed throughout the genome. Principal component analysis and hierarchical clustering revealed complete segregation of the FGR and control group. Next, we identified 569 upregulated genes and 521 downregulated genes in FGR placentas by RNA sequencing. Differential gene transcription largely corresponded to expected direction based on H3K27ac status. Pathway analysis on upregulated transcripts originating from hyperacetylated sites revealed genes related to the HIF-1-alpha transcription factor network and several other genes with known involvement in placental pathology (LEP, FLT1, HK2, ENG, FOS). Downregulated transcripts in the vicinity of hypoacetylated sites were related to the immune system and growth hormone receptor signaling. Additionally, we found enrichment of 141 transcription factor binding motifs within differentially acetylated regions. Of the corresponding transcription factors, four were upregulated, SP1, ARNT2, HEY2, and VDR, and two downregulated, FOSL and NR4A1. Conclusion: We demonstrate a key role for genome-wide alterations in H3K27ac in FGR placentas corresponding with changes in transcription profiles of regions relevant to placental function. Future studies on the role of H3K27ac in FGR and placental-fetal development may help to identify novel targets for therapy of this currently incurable disease. Keywords: ChIP-seq, Growth restriction, H3K27ac, Epigenetics, Histone acetylation, Placenta, Placental pathology, RNA-seq Background and distal enhancers, the most important and best under- The dynamics of histone 3 lysine 27 acetylation stood regulatory domains. To study involvement of this (H3K27ac) in DNA regulatory regions is one of the com- regulatory level/layer in placental pathology, we mapped ponents playing a fundamental role in the precise timing H3K27ac occupancy in healthy placentas and placentas and level of gene transcription [1, 2]. Consequently, aber- from pregnancies with fetal growth restriction (FGR). rant H3K27ac has been suggested to be involved in dis- FGR, a condition in which the fetus is unable to ease pathology by eliciting pathological gene expression achieve its full growth potential through inadequate sup- programs [3, 4]. H3K27ac marks both active promoters ply of nutrients and growth factors, occurs in approxi- mately 5% of pregnancies [5]. FGR imposes a major risk of perinatal morbidity and mortality [6] and programs * Correspondence: [email protected]; [email protected] the health of the fetus throughout life, by being associ- 1Department of Obstetrics, Wilhelmina Children’s Hospital Birth Center, University Medical Center Utrecht, Utrecht, the Netherlands ated with a future risk of type 2 diabetes and Full list of author information is available at the end of the article © The Author(s). 2018 Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated. Paauw et al. Clinical Epigenetics (2018) 10:85 Page 2 of 11 cardiovascular and renal disease [7, 8]. At the histo- Genes annotated to within 20 kb of differentially pathological level, FGR placenta exhibit signs of dis- acetylated regions rupted placental development characterized by increased To identify the biological relevance of differentially acet- infarction area, increased syncytial knotting, inflamma- ylated peaks, we annotated genes to the hyper- and tion, and impaired trophoblast invasion into the spiral hypoacetylated sites using a window of ± 20 kb from arteries of the uterus due to inappropriate maternal-fetal transcription start site (TSS). We annotated 368 genes immune interaction [9–11]. corresponding to hyperacytelated sites and 313 genes as- Previous studies of placentas of FGR pregnancies have sociated with hypoacetylated sites (Additional file 1: reported differences in gene transcription across many re- Table S2). Several of these annotated genes are known to gions across the genome. Pathways associated with altered be involved in placental development. For instance, we placentalgeneexpressioninFGR include angiogenesis, im- found hyperacetylation of regions near HK2, FLT1, and mune modulation, energy production, and growth signaling LEP, previously reported to be upregulated in other pla- [12–15]. Most adaptive responses of the placenta, e.g., to cental disorders [25, 26], and hypoacetylation of regions support restricted fetal growth, are thought to result from near CH2 and CDLN1, which are involved in growth changes in epigenetic regulation [16–18]. Recently, DNA and endothelial cell-to-cell adhesions. The individual methylation has been mapped in the human placenta [19] peaks of selected regions of interest are presented in and a number of studies have suggested differences in Fig. 2a (near HK2) and Fig. 2b (near Flt-1, LEP, CDLN1, genome-wide methylation profiles in FGR placentas [20– and CH1). These figures show that sites identified to be 22]. We assume that disruptions in other epigenetic sys- differentially acetylated are highly similar across each of tems (e.g., histone modifications and other posttranslational the replicates. chromatin modifications) regulating placenta gene expres- sionarealsolikelytobeinvolved[23, 24]. Pathway analysis of differential acetylated regions in FGR In this study, we mapped differential H3K27ac profiles placentas in DNA regulatory regions in relation to disrupted devel- Next, we aimed to assign biological significance to genes opment of the placenta, by exploring the presence of annotated to differentially acetylated peaks by pathway H3K27ac using chromatin immunoprecipitation sequen- analysis using GREAT software. The software annotated cing (ChIP-seq) in FGR and control placentas. Next, we 515 genes to hyperacetylated regions in FGR. Although performed RNA-seq to examine whether differences in no enrichment for GO biological process or for path- H3K27ac also reflects gene expression levels. With this ways was identified using this approach, the annotated approach, we identified previously unstudied alterations in genes were enriched for genes that are transcriptionally promoter and enhancer activity related to placenta regulated by HIF-1-alfa/hypoxia within the MSigDB per- pathology. turbation ontology (full output supplied in Add- itional file 1: Table S3). Other functional pathways Results related to hyperacetylated regions include pathways in- Detection of regions with differential H3K27ac occupancy volved in cancer and immune response. Nearby the dif- First, we performed genome-wide analysis of H3K27ac by ferential hypoacetylated regions, GREAT identified 868 ChIP-seq on placental tissue from control (n = 4) and FGR genes. GO biological processes and pathways related to pregnancies (n = 5) and identified 30,288 H3K27ac peaks these genes consisted of angiogenesis, response to exter- that were present in at least two independent samples. Of nal signals, and immune activation (Fig. 3a, b, full output these, 970 regions showed differential H3K27ac levels in supplied in Additional file 1: Table S3). Interestingly, FGR compared to controls with 366 being hyperacetylated many of these pathways are known to be disrupted in and 604 regions being hypoacetylated in FGR (Fig. 1a,full FGR, especially angiogenesis, HIF-1-alpha signaling, and list supplied in Additional file 1: Table S1). Based on the the immune environment/response [27–29]. differentially acetylated regions, the FGR and healthy pla- centas could be clearly segregated using both supervised Cross-validation of functional effects by combining and unsupervised analysis (Fig. 1b–c,Additionalfile2) H3K27ac and mRNA profiles These findings indicate clear distinction between the two To cross-validate functional meaning of the differentially groups and suggest a specific and highly reproducible acetylated peaks and to examine whether hyper- or H3K27ac pattern in FGR placentas. Differentially acety- hypoacetylated state of H3K27 was also accompanied by lated regions were distributed throughout the whole gen- differential gene expression, we performed RNA-seq. We ome as shown by Manhattan
Recommended publications
  • Genome-Wide Profiling of Active Enhancers in Colorectal Cancer
    Genome-wide proling of active enhancers in colorectal cancer Min Wu ( [email protected] ) Wuhan University https://orcid.org/0000-0003-1372-4764 Qinglan Li Wuhan University Xiang Lin Wuhan University Ya-Li Yu Zhongnan Hospital, Wuhan University Lin Chen Wuhan University Qi-Xin Hu Wuhan University Meng Chen Zhongnan Hospital, Wuhan University Nan Cao Zhongnan Hospital, Wuhan University Chen Zhao Wuhan University Chen-Yu Wang Wuhan University Cheng-Wei Huang Wuhan University Lian-Yun Li Wuhan University Mei Ye Zhongnan Hospital, Wuhan University https://orcid.org/0000-0002-9393-3680 Article Keywords: Colorectal cancer, H3K27ac, Epigenetics, Enhancer, Transcription factors Posted Date: December 10th, 2020 DOI: https://doi.org/10.21203/rs.3.rs-119156/v1 License: This work is licensed under a Creative Commons Attribution 4.0 International License. Read Full License Genome-wide profiling of active enhancers in colorectal cancer Qing-Lan Li1, #, Xiang Lin1, #, Ya-Li Yu2, #, Lin Chen1, #, Qi-Xin Hu1, Meng Chen2, Nan Cao2, Chen Zhao1, Chen-Yu Wang1, Cheng-Wei Huang1, Lian-Yun Li1, Mei Ye2,*, Min Wu1,* 1 Frontier Science Center for Immunology and Metabolism, Hubei Key Laboratory of Cell Homeostasis, Hubei Key Laboratory of Developmentally Originated Disease, Hubei Key Laboratory of Intestinal and Colorectal Diseases, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China 2Division of Gastroenterology, Department of Geriatrics, Hubei Clinical Centre & Key Laboratory of Intestinal and Colorectal Diseases, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430072, China #Equal contribution to the study. Contact information *Correspondence should be addressed to Dr. Min Wu, Email: [email protected], Tel: 86-27-68756620, or Dr.
    [Show full text]
  • Recognition of Histone Acetylation by the GAS41 YEATS Domain Promotes H2A.Z Deposition in Non-Small Cell Lung Cancer
    Downloaded from genesdev.cshlp.org on October 5, 2021 - Published by Cold Spring Harbor Laboratory Press Recognition of histone acetylation by the GAS41 YEATS domain promotes H2A.Z deposition in non-small cell lung cancer Chih-Chao Hsu,1,2,8 Jiejun Shi,3,8 Chao Yuan,1,2,7,8 Dan Zhao,4,5,8 Shiming Jiang,1,2 Jie Lyu,3 Xiaolu Wang,1,2 Haitao Li,4,5 Hong Wen,1,2 Wei Li,3 and Xiaobing Shi1,2,6 1Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA; 2Center for Cancer Epigenetics, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA; 3Dan L. Duncan Cancer Center, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, USA; 4MOE Key Laboratory of Protein Sciences, Beijing Advanced Innovation Center for Structural Biology, Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing 100084, China; 5Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing 100084, China; 6Genetics and Epigenetics Graduate Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas 77030, USA Histone acetylation is associated with active transcription in eukaryotic cells. It helps to open up the chromatin by neutralizing the positive charge of histone lysine residues and providing binding platforms for “reader” proteins. The bromodomain (BRD) has long been thought to be the sole protein module that recognizes acetylated histones. Re- cently, we identified the YEATS domain of AF9 (ALL1 fused gene from chromosome 9) as a novel acetyl-lysine- binding module and showed that the ENL (eleven-nineteen leukemia) YEATS domain is an essential acetyl-histone reader in acute myeloid leukemias.
    [Show full text]
  • Investigation of KRAS Dependency Bypass and Functional Characterization of All Possible KRAS Missense Variants
    Investigation of KRAS Dependency Bypass and Functional Characterization of All Possible KRAS Missense Variants The Harvard community has made this article openly available. Please share how this access benefits you. Your story matters Citable link http://nrs.harvard.edu/urn-3:HUL.InstRepos:40050098 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 Investigation of KRAS Dependency Bypass and Functional Characterization of All Possible KRAS Missense Variants A dissertation presented by Seav Huong Ly to The Division of Medical Sciences in partial fulfillment of the requirements for the degree of Doctor of Philosophy in the subject of Biological and Biomedical Sciences Harvard University Cambridge, Massachusetts April 2018 © 2018 Seav Huong Ly All rights reserved. Dissertation Advisor: William C. Hahn Seav Huong Ly Investigation of KRAS Dependency Bypass and Functional Characterization of All Possible KRAS Missense Variants Abstract The importance of oncogenic KRAS in human cancers have prompted intense efforts to target KRAS and its effectors. To anticipate the development of resistance to these strategies, we previously performed a genome-scale expression screen to identify genes that bypass KRAS oncogenic dependency. Here we test thirty-seven genes that scored over five standard deviations and find that overexpression of LIM homeobox 9 (LHX9), a transcription factor involved in embryonic development, robustly rescues the suppression of KRAS in vitro and xenograft models. Furthermore, LHX9 substantially decreases cell sensitivity to KRASG12C and MEK1/2 inhibitors in KRAS-dependent cells.
    [Show full text]
  • Dynamics of Transcription-Dependent H3k36me3 Marking by the SETD2:IWS1:SPT6 Ternary Complex
    bioRxiv preprint doi: https://doi.org/10.1101/636084; this version posted May 14, 2019. 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. Dynamics of transcription-dependent H3K36me3 marking by the SETD2:IWS1:SPT6 ternary complex Katerina Cermakova1, Eric A. Smith1, Vaclav Veverka2, H. Courtney Hodges1,3,4,* 1 Department of Molecular & Cellular Biology, Center for Precision Environmental Health, and Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA 2 Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic 3 Center for Cancer Epigenetics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA 4 Department of Bioengineering, Rice University, Houston, TX, 77005, USA * Lead contact; Correspondence to: [email protected] Abstract The genome-wide distribution of H3K36me3 is maintained SETD2 contributes to gene expression by marking gene through various mechanisms. In human cells, H3K36 is bodies with H3K36me3, which is thought to assist in the mono- and di-methylated by eight distinct histone concentration of transcription machinery at the small portion methyltransferases; however, the predominant writer of the of the coding genome. Despite extensive genome-wide data trimethyl mark on H3K36 is SETD21,11,12. Interestingly, revealing the precise localization of H3K36me3 over gene SETD2 is a major tumor suppressor in clear cell renal cell bodies, the physical basis for the accumulation, carcinoma13, breast cancer14, bladder cancer15, and acute maintenance, and sharp borders of H3K36me3 over these lymphoblastic leukemias16–18. In these settings, mutations sites remains rudimentary.
    [Show full text]
  • EZH2 in Normal Hematopoiesis and Hematological Malignancies
    www.impactjournals.com/oncotarget/ Oncotarget, Vol. 7, No. 3 EZH2 in normal hematopoiesis and hematological malignancies Laurie Herviou2, Giacomo Cavalli2, Guillaume Cartron3,4, Bernard Klein1,2,3 and Jérôme Moreaux1,2,3 1 Department of Biological Hematology, CHU Montpellier, Montpellier, France 2 Institute of Human Genetics, CNRS UPR1142, Montpellier, France 3 University of Montpellier 1, UFR de Médecine, Montpellier, France 4 Department of Clinical Hematology, CHU Montpellier, Montpellier, France Correspondence to: Jérôme Moreaux, email: [email protected] Keywords: hematological malignancies, EZH2, Polycomb complex, therapeutic target Received: August 07, 2015 Accepted: October 14, 2015 Published: October 20, 2015 This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited. ABSTRACT Enhancer of zeste homolog 2 (EZH2), the catalytic subunit of the Polycomb repressive complex 2, inhibits gene expression through methylation on lysine 27 of histone H3. EZH2 regulates normal hematopoietic stem cell self-renewal and differentiation. EZH2 also controls normal B cell differentiation. EZH2 deregulation has been described in many cancer types including hematological malignancies. Specific small molecules have been recently developed to exploit the oncogenic addiction of tumor cells to EZH2. Their therapeutic potential is currently under evaluation. This review summarizes the roles of EZH2 in normal and pathologic hematological processes and recent advances in the development of EZH2 inhibitors for the personalized treatment of patients with hematological malignancies. PHYSIOLOGICAL FUNCTIONS OF EZH2 state through tri-methylation of lysine 27 on histone H3 (H3K27me3) [6].
    [Show full text]
  • Oncogenic Deregulation of EZH2 As an Opportunity for Targeted Therapy in Lung Cancer
    Published OnlineFirst June 16, 2016; DOI: 10.1158/2159-8290.CD-16-0164 RESEARCH ARTICLE Oncogenic Deregulation of EZH2 as an Opportunity for Targeted Therapy in Lung Cancer Haikuo Zhang1,2, Jun Qi1,2, Jaime M. Reyes1, Lewyn Li3, Prakash K. Rao3, Fugen Li3, Charles Y. Lin1, Jennifer A. Perry1, Matthew A. Lawlor1, Alexander Federation1, Thomas De Raedt2,4, Yvonne Y. Li1,2, Yan Liu1,2, Melissa A. Duarte3, Yanxi Zhang1,2, Grit S. Herter-Sprie1,2, Eiki Kikuchi1,2, Julian Carretero5, Charles M. Perou6, Jacob B. Reibel1,2, Joshiawa Paulk1, Roderick T. Bronson7, Hideo Watanabe1,2, Christine Fillmore Brainson8,9,10, Carla F. Kim8,9,10, Peter S. Hammerman1,2, Myles Brown2,3, Karen Cichowski2,4, Henry Long3, James E. Bradner1,2, and Kwok-Kin Wong1,2,11 Downloaded from cancerdiscovery.aacrjournals.org on September 29, 2021. © 2016 American Association for Cancer Research. Published OnlineFirst June 16, 2016; DOI: 10.1158/2159-8290.CD-16-0164 ABSTRACT As a master regulator of chromatin function, the lysine methyltransferase EZH2 orchestrates transcriptional silencing of developmental gene networks. Overex- pression of EZH2 is commonly observed in human epithelial cancers, such as non–small cell lung carci- noma (NSCLC), yet definitive demonstration of malignant transformation by deregulatedEZH2 remains elusive. Here, we demonstrate the causal role of EZH2 overexpression in NSCLC with new genetically engineered mouse models of lung adenocarcinoma. Deregulated EZH2 silences normal developmental pathways, leading to epigenetic transformation independent of canonical growth factor pathway acti- vation. As such, tumors feature a transcriptional program distinct from KRAS- and EGFR-mutant mouse lung cancers, but shared with human lung adenocarcinomas exhibiting high EZH2 expression.
    [Show full text]
  • Quantitative Proteomic Characterization and Comparison of T Helper 17 and Induced Regulatory T Cells
    METHODS AND RESOURCES Quantitative proteomic characterization and comparison of T helper 17 and induced regulatory T cells Imran Mohammad1,2, Kari Nousiainen3, Santosh D. Bhosale1,2, Inna Starskaia1,2, Robert Moulder1, Anne Rokka1, Fang Cheng4, Ponnuswamy Mohanasundaram4, John E. Eriksson4, David R. Goodlett5, Harri LaÈhdesmaÈki3, Zhi Chen1* 1 Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland, 2 Turku Doctoral Programme of Molecular Medicine, University of Turku, Turku, Finland, 3 Department of Computer a1111111111 Science, Aalto University, Espoo, Finland, 4 Cell Biology, Biosciences, Faculty of Science and Engineering, a1111111111 Åbo Akademi University, Turku, Finland, 5 Department of Pharmaceutical Sciences, University of Maryland a1111111111 School of Pharmacy, Baltimore, Maryland, United States of America a1111111111 a1111111111 * [email protected] Abstract OPEN ACCESS The transcriptional network and protein regulators that govern T helper 17 (Th17) cell differ- Citation: Mohammad I, Nousiainen K, Bhosale SD, entiation have been studied extensively using advanced genomic approaches. For a better Starskaia I, Moulder R, Rokka A, et al. (2018) understanding of these biological processes, we have moved a step forward, from gene- to Quantitative proteomic characterization and protein-level characterization of Th17 cells. Mass spectrometry±based label-free quantita- comparison of T helper 17 and induced regulatory T cells. PLoS Biol 16(5): e2004194. https://doi.org/ tive (LFQ) proteomics analysis were made of in vitro differentiated murine Th17 and induced 10.1371/journal.pbio.2004194 regulatory T (iTreg) cells. More than 4,000 proteins, covering almost all subcellular compart- Academic Editor: Paula Oliver, University of ments, were detected. Quantitative comparison of the protein expression profiles resulted in Pennsylvania Perelman School of Medicine, United the identification of proteins specifically expressed in the Th17 and iTreg cells.
    [Show full text]
  • Aging Human Hematopoietic Stem Cells Manifest Profound Epigenetic Reprogramming of Enhancers That May Predispose to Leukemia
    Published OnlineFirst May 13, 2019; DOI: 10.1158/2159-8290.CD-18-1474 RESEARCH ARTICLE Aging Human Hematopoietic Stem Cells Manifest Profound Epigenetic Reprogramming of Enhancers That May Predispose to Leukemia Emmalee R. Adelman1,2,3, Hsuan-Ting Huang1,2, Alejandro Roisman1,2, André Olsson4, Antonio Colaprico1,2, Tingting Qin5, R. Coleman Lindsley6, Rafael Bejar7, Nathan Salomonis8, H. Leighton Grimes4, and Maria E. Figueroa1,2 Downloaded from cancerdiscovery.aacrjournals.org on October 5, 2021. © 2019 American Association for Cancer Research. Published OnlineFirst May 13, 2019; DOI: 10.1158/2159-8290.CD-18-1474 ABSTRACT Aging is associated with functional decline of hematopoietic stem cells (HSC) as well as an increased risk of myeloid malignancies. We performed an integrative characterization of epigenomic and transcriptomic changes, including single-cell RNA sequencing, dur- ing normal human aging. Lineage−CD34+CD38− cells [HSC-enriched (HSCe)] undergo age-associated epigenetic reprogramming consisting of redistribution of DNA methylation and reductions in H3K27ac, H3K4me1, and H3K4me3. This reprogramming of aged HSCe globally targets developmental and can- cer pathways that are comparably altered in acute myeloid leukemia (AML) of all ages, encompassing loss of 4,646 active enhancers, 3,091 bivalent promoters, and deregulation of several epigenetic modi- fiers and key hematopoietic transcription factors, such as KLF6, BCL6, and RUNX3. Notably,in vitro downregulation of KLF6 results in impaired differentiation, increased colony-forming potential, and changes in expression that recapitulate aging and leukemia signatures. Thus, age-associated epigenetic reprogramming may form a predisposing condition for the development of age-related AML. SIGNIFICANCE: AML, which is more frequent in the elderly, is characterized by epigenetic deregulation.
    [Show full text]
  • Histone H3k27ac Separates Active from Poised Enhancers and Predicts Developmental State
    Histone H3K27ac separates active from poised SEE COMMENTARY enhancers and predicts developmental state Menno P. Creyghtona,1, Albert W. Chenga,b,1, G. Grant Welsteada, Tristan Kooistrac,d, Bryce W. Careya,e, Eveline J. Steinea,e, Jacob Hannaa, Michael A. Lodatoa,e, Garrett M. Framptona,e, Phillip A. Sharpd,e, Laurie A. Boyere, Richard A. Younga,e, and Rudolf Jaenischa,e,2 aWhitehead Institute for Biomedical Research, Cambridge, MA 02142; bComputational and Systems Biology Program, Massachusetts Institute of Technology, Cambridge, MA 02142; cDivision of Health Sciences and Technology, Harvard–Massachusetts Institute of Technology, Boston, MA 02115; dKoch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142; and eDepartment of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139 Contributed by Rudolf Jaenisch, October 26, 2010 (sent for review September 29, 2010) Developmental programs are controlled by transcription factors and distal enhancers (13). Finally distal H3K4me1-enriched Stat1 chromatin regulators, which maintain specific gene expression binding sites become selectively activated upon INF-γ treatment programs through epigenetic modification of the genome. These of HeLa cells, suggesting that these regions maintain the po- regulatory events at enhancers contribute to the specificgene tential to become active (5). This raises the intriguing possibility expression programs that determine cell state and the potential that enhancers contain information about the current and future for differentiation into new cell types. Although enhancer elements developmental potential of a cell, as well as its ability to respond are known to be associated with certain histone modifications and to external cues. fi Here, we identify close to 135,000 candidate distal enhancer transcription factors, the relationship of these modi cations to gene fi expression and developmental state has not been clearly defined.
    [Show full text]
  • Epigenomic Analysis Reveals DNA Motifs Regulating Histone Modifications in Human and Mouse
    Epigenomic analysis reveals DNA motifs regulating histone modifications in human and mouse Vu Ngoa,1, Zhao Chenb,1, Kai Zhanga, John W. Whitakerb, Mengchi Wanga, and Wei Wanga,b,c,2 aGraduate Program of Bioinformatics and Systems Biology, University of California, San Diego, La Jolla, CA 92093-0359; bDepartment of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA 92093-0359; and cDepartment of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093-0359 Edited by Steven Henikoff, Fred Hutchinson Cancer Research Center, Seattle, WA, and approved January 3, 2019 (received for review August 6, 2018) Histones are modified by enzymes that act in a locus, cell-type, and An analogy is that a transcription factor (TF) recognizes the same developmental stage-specific manner. The recruitment of enzymes DNA motif but its binding sites are cell-type–dependent. However, if to chromatin is regulated at multiple levels, including interaction we identify all motifs enriched in the TF binding sites across a large with sequence-specific DNA-binding factors. However, the DNA- and diverse set of cell types, the most common motif is likely the one binding specificity of the regulatory factors that orchestrate spe- recognized by the TF. Histone modifications are more complicated cific histone modifications has not been broadly mapped. We have than a single TF binding and one histone mark can be regulated by analyzed 6 histone marks (H3K4me1, H3K4me3, H3K27ac, H3K27me3, K3H9me3, H3K36me3) across 121 human cell types and tissues from multiple factors recognizing different motifs. Therefore, a compar- the NIH Roadmap Epigenomics Project as well as 8 histone marks ative analysis across diverse cell types/tissues is critical.
    [Show full text]
  • EZH2 Inhibition Results in Genome-Wide PRC2 Redistribution
    bioRxiv preprint doi: https://doi.org/10.1101/713842; this version posted July 31, 2019. 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 4.0 International license. Title EZH2 inhibition results in genome-wide PRC2 redistribution Authors/Affiliations 1* 2* 2,3,† Chih-Chi Yuan ,​ Ah Jung Jeon ,​ Greg Tucker-Kellogg ,​ Barbara ​ ​ ​ 1,†,‡ 1 Bryant ,​ and Patrick Trojer ​ ​ ​ 1 ​ Constellation Pharmaceuticals, Cambridge, Massachusetts, 02142, USA 2 ​ Department of Biological Sciences, National University of Singapore, Singapore 117558, Singapore 3 ​ Computational Biology Programme, Faculty of Science, National University of Singapore, Singapore 117558, Singapore * Equal​ contributor † ​ Corresponding author ‡ ​ Lead Contact Correspondence Barbara Bryant [email protected] ​ or Greg Tucker-Kellogg [email protected] (Twitter: @gtuckerkellogg). ​ ​ Requests for materials should be directed to [email protected]. ​ 1 bioRxiv preprint doi: https://doi.org/10.1101/713842; this version posted July 31, 2019. 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 4.0 International license. Summary Histone methyltransferase polycomb repressive complex 2 (PRC2) plays a critical role in cell fate determination, and its catalytic subunit EZH2 is a key oncogenic driver in GCB-DLBCL. EZH2 inhibition in some GCB-DLBCL cell models leads to a global loss of H3K27me3, the derepression of a subset of silenced PRC2 target genes, and ultimately cell death.
    [Show full text]
  • Histone H3K27 Methyltransferase Ezh2 Represses Wnt Genes to Facilitate Adipogenesis
    Histone H3K27 methyltransferase Ezh2 represses Wnt genes to facilitate adipogenesis Lifeng Wanga, Qihuang Jina, Ji-Eun Leea, I-hsin Sub,1, and Kai Gea,2 aNuclear Receptor Biology Section, Clinical Endocrinology Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892; and bLaboratory of Lymphocyte Signaling, The Rockefeller University, New York, NY 10065 Edited by Mark T. Groudine, Fred Hutchinson Cancer Research Center, Seattle, WA, and approved March 12, 2010 (received for review January 4, 2010) Wnt/β-catenin signaling inhibits adipogenesis. Genome-wide histone methyltransferase subunit Ezh2, PRC2 methylates his- profiling studies have revealed the enrichment of histone H3K27 tone H3 on lysine 27 (H3K27). The resulting H3K27 trimethy- methyltransferase Ezh2 on Wnt genes. However, the functional lation is specifically recognized and bound by the PRC1 complex significance of such a direct link between the two types of devel- to facilitate transcriptional repression (6). PRC2 and PRC1 are opmental regulators in mammalian cells, and the role of Ezh2 in localized on a large number of developmental genes in embry- adipogenesis, remain unclear. Here we show Ezh2 and its H3K27 onic stem (ES) cells. Disruption of PRC2 by deletion of Ezh2, methyltransferase activity are required for adipogenesis. Ezh2 Suz12, or EED in ES cells markedly decreases the global levels directly represses Wnt1,-6,-10a, and -10b genes in preadipocytes of H3K27 di- and trimethylation (H3K27me2 and H3K27me3) and during adipogenesis. Deletion of Ezh2 eliminates H3K27me3 and derepresses many polycomb target genes (7–12). on Wnt promoters and derepresses Wnt expression, which leads Genomewide profiling studies have revealed the enrichment β to activation of Wnt/ -catenin signaling and inhibition of adipo- of H3K27 methyltransferase Ezh2 and associated H3K27me3 on genesis.
    [Show full text]