Histone H3k27ac Separates Active from Poised Enhancers and Predicts Developmental State

Total Page:16

File Type:pdf, Size:1020Kb

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. regions in ve different cell types. In a candidate approach to Here we interrogate the epigenetic landscape of enhancer elements identify factors that could potentially discriminate between the various possible enhancer states, we establish H3K27ac as an in embryonic stem cells and several adult tissues in the mouse. We important enhancer mark that can distinguish between active and find that histone H3K27ac distinguishes active enhancers from poised enhancer elements. This mark can be deposited by both inactive/poised enhancer elements containing H3K4me1 alone. This p300 and CREB binding protein (CBP) (14) and is associated with indicates that the amount of actively used enhancers is lower than active promoters in mammalian cells (15). Using this modification previously anticipated. Furthermore, poised enhancer networks to distinguish poised from active enhancer networks allows us to provide clues to unrealized developmental programs. Finally, we make predictions on the developmental potency of the cell, as well show that enhancers are reset during nuclear reprogramming. as it current state. Furthermore, it suggests that the active global enhancer network of a cell is smaller than previously anticipated. he genome comprises a large number of noncoding DNA Tregulatory elements, including silencers, insulators, and en- Results hancer regions, that play important regulatory roles in maintain- H3K27ac Distinguishes Active from Inactive Enhancers. We first ing gene expression programs. Enhancers have emerged as key identified cell type-specific enhancers in murine ES cells using cis-regulatory elements that can affect gene transcription in- distal H3K4me1 histone marks identified by ChIP-Seq (Fig. 1A, dependent of their orientation or distance. Global identification Table S1, and SI Materials and Methods) using similar criteria as of these regions as well as their contribution to target gene ex- shown previously (5–9, 16). We identified 25,036 distal enhancer pression has been challenging because enhancers can often reside elements that are roughly equally distributed between introns thousands of base pairs away from their target of regulation (1, 2). and intergenic regions (Fig. S1A). To test the robustness of these Individual enhancers have been identified by detailed studies data we compared our results with a previously published of disease-related elements that reside in noncoding regions H3K4me1 data set in 129/C57BL6 F1 ES cells (17) and found outside of model genes (3), followed on a larger scale by com- that they displayed a remarkably similar distribution pattern parative genomics using ultra-conservation (4). These studies (Pearson correlation, 0.76; Fig. S1 B and C). Second ES cells unequivocally established specific cis-regulatory elements as from an inbred genetic background (C57BL6/N) yielded very important regulators of development and disease. Recently similar results (Pearson correlation, 0.77), indicating that cell efforts to globally identify enhancers have focused on the histone type-specific enhancers pattern the genome with high fidelity mark H3K4me1 to locate many cell type-specific enhancer sites with little effect from genetic background. (5–9). Analysis of human primary and tumor cell lines has shown Proximal gene expression was previously shown to be prefer- that enhancer regions identified by virtue of H3K4me1 enrich- entially affected by enhancer elements (11), and enhancers seem ment, as well as DNaseI hypersensitivity, display a cell type- to cluster near the genes they regulate (5). Similarly, the overall specific distribution across the genome (5, 10). Another study found that the histone acetyltransferase p300 is enriched at enhancers in fetal mouse tissues and found similar tissue-specific Author contributions: M.P.C., A.W.C., and R.J. designed research; M.P.C., A.W.C., G.G.W., marking of these elements. Notably, many of these enhancers B.W.C., E.J.S., J.H., and M.A.L. performed research; G.G.W., T.K., G.M.F., P.A.S., L.A.B., and were shown to contribute to transcriptional activity of proximal R.A.Y. contributed new reagents/analytic tools; M.P.C., A.W.C., T.K., and G.M.F. analyzed genes in a tissue-specific manner (11). data; and M.P.C., A.W.C., L.A.B., R.A.Y., and R.J. wrote the paper. Although most H3K4me1 marked enhancer regions displayed The authors declare no conflict of interest. activity when tested in reporter assays, a significant percentage This article is a PNAS Direct Submission. was not active (5, 11). One hypothesis is that H3K4me1 can mark Data deposition: Raw data are available online at the Gene Expression Omnibus (GEO) active enhancers as well as those in a poised or predetermined ascension viewer series, www.ncbi.nlm.nih.gov/geo (accession nos. GSE24164, GSE24165, state. Indeed, several lines of evidence support the existence of and GSE23907). poised enhancers. Lineage-specific enhancers that become acti- See Commentary on page 21240. vated upon differentiation are protected from DNA methylation 1M.P.C. and A.W.C. contributed equally to this work in the parental ES cells, where the enhancers are not yet active 2To whom correspondence should be addressed. E-mail: [email protected]. (12). Furthermore, large sets of inactive developmental genes in This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. SYSTEMS BIOLOGY hematopoietic stem cells are associated with H3K4me1-enriched 1073/pnas.1016071107/-/DCSupplemental. www.pnas.org/cgi/doi/10.1073/pnas.1016071107 PNAS | December 14, 2010 | vol. 107 | no. 50 | 21931–21936 A H3K4me1 H3K4me3 B C Fig. 1. H3K27ac is a good candidate to 100 100 distinguish active from inactive enhancer elements. (A) Heatmap of 25,036 putative le i le t i enhancers based on H3K4me1 enrichment t and H3K4me3 depletion in ES cells. Eight- percen percen kilobase pairs around the center of the en- hancer region are displayed. (B)Geneex- 0 0 pression of microarray data generated in duplicates in ES cells. Box-plots show all genes (All) and genes found associated with 25036 putave enhancers 25036 putave enhancers (H3K4me1+) either enriched (+) − Short RNA H3K27ac H3K4me1 or unenriched ( ) for Oct4 or (C) H3K27ac. D 40.00% E Solid bars of boxes display the 25–75% of ranked genes with the mean indicated as an 30.00% intersection. (D) Graph showing the per- centage of enhancers identified in A that − 20.00% are called bound at P =10 8 by the indicated ES enhancers ES factors or chromatin marks. (E)Heatmap 21166 TSSs 10.00% showing a composite of distal H3K27ac- and Percentage enrichment 0.00% H3K4me1-enriched regions ordered by pres- ence and absence of these two histone marks. Left: Occurrence of short RNA reads based on this distribution within a −8kbto +8 kb window (SI Materials and Methods). presence of the enhancers identified in this study correlated well 4.06E-7). These data indicate that H3K27ac is a good candidate with increased gene activity of proximal genes when we com- to discriminate between active and poised enhancer states. In pared these ChIP data with microarray expression data (Fig. 1 B support of this, we found that Oct4 (P = 1.8E-8) and Sox2 (P = and C; P = 4.4E-16). Consistent with its role as a master regu- 1.7E-8) are selectively enriched at H3K27ac-negative enhancers. lator of ES cell state, motif search analysis identified the Oct4 Because genes can be associated with multiple enhancers that (POU) binding domain at ES cell enhancers (P = 1.0E-250), are either H3K4me1+, H3K27ac+, or a combination of both, we suggesting a role for this transcription factor in enhancer
Recommended publications
  • Dedifferentiation by Adenovirus E1A Due to Inactivation of Hippo Pathway Effectors YAP and TAZ
    Downloaded from genesdev.cshlp.org on October 3, 2021 - Published by Cold Spring Harbor Laboratory Press Dedifferentiation by adenovirus E1A due to inactivation of Hippo pathway effectors YAP and TAZ Nathan R. Zemke, Dawei Gou, and Arnold J. Berk Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California 90095, USA Adenovirus transformed cells have a dedifferentiated phenotype. Eliminating E1A in transformed human embryonic kidney cells derepressed ∼2600 genes, generating a gene expression profile closely resembling mesenchymal stem cells (MSCs). This was associated with a dramatic change in cell morphology from one with scant cytoplasm and a globular nucleus to one with increased cytoplasm, extensive actin stress fibers, and actomyosin-dependent flat- tening against the substratum. E1A-induced hypoacetylation at histone H3 Lys27 and Lys18 (H3K27/18) was reversed. Most of the increase in H3K27/18ac was in enhancers near TEAD transcription factors bound by Hippo signaling-regulated coactivators YAP and TAZ. E1A causes YAP/TAZ cytoplasmic sequestration. After eliminating E1A, YAP/TAZ were transported into nuclei, where they associated with poised enhancers with DNA-bound TEAD4 and H3K4me1. This activation of YAP/TAZ required RHO family GTPase signaling and caused histone acetylation by p300/CBP, chromatin remodeling, and cohesin loading to establish MSC-associated enhancers and then superenhancers. Consistent results were also observed in primary rat embryo kidney cells, human fibroblasts, and human respiratory tract epithelial cells. These results together with earlier studies suggest that YAP/TAZ function in a developmental checkpoint controlled by signaling from the actin cytoskeleton that prevents differ- entiation of a progenitor cell until it is in the correct cellular and tissue environment.
    [Show full text]
  • DNA Methylation Regulates Discrimination of Enhancers From
    Sharifi-Zarchi et al. BMC Genomics (2017) 18:964 DOI 10.1186/s12864-017-4353-7 RESEARCHARTICLE Open Access DNA methylation regulates discrimination of enhancers from promoters through a H3K4me1-H3K4me3 seesaw mechanism Ali Sharifi-Zarchi1,2,3,4†, Daniela Gerovska5†, Kenjiro Adachi6, Mehdi Totonchi3, Hamid Pezeshk7,8, Ryan J. Taft9, Hans R. Schöler6,10, Hamidreza Chitsaz2, Mehdi Sadeghi8,11, Hossein Baharvand3,12* and Marcos J. Araúzo-Bravo5,13,14* Abstract Background: DNA methylation at promoters is largely correlated with inhibition of gene expression. However, the role of DNA methylation at enhancers is not fully understood, although a crosstalk with chromatin marks is expected. Actually, there exist contradictory reports about positive and negative correlations between DNA methylation and H3K4me1, a chromatin hallmark of enhancers. Results: We investigated the relationship between DNA methylation and active chromatin marks through genome- wide correlations, and found anti-correlation between H3K4me1 and H3K4me3 enrichment at low and intermediate DNA methylation loci. We hypothesized “seesaw” dynamics between H3K4me1 and H3K4me3 in the low and intermediate DNA methylation range, in which DNA methylation discriminates between enhancers and promoters, marked by H3K4me1 and H3K4me3, respectively. Low methylated regions are H3K4me3 enriched, while those with intermediate DNA methylation levels are progressively H3K4me1 enriched. Additionally, the enrichment of H3K27ac, distinguishing active from primed enhancers, follows a plateau in the lower range of the intermediate DNA methylation level, corresponding to active enhancers, and decreases linearly in the higher range of the intermediate DNA methylation. Thus, the decrease of the DNA methylation switches smoothly the state of the enhancers from a primed to an active state.
    [Show full text]
  • Modes of Interaction of KMT2 Histone H3 Lysine 4 Methyltransferase/COMPASS Complexes with Chromatin
    cells Review Modes of Interaction of KMT2 Histone H3 Lysine 4 Methyltransferase/COMPASS Complexes with Chromatin Agnieszka Bochy ´nska,Juliane Lüscher-Firzlaff and Bernhard Lüscher * ID Institute of Biochemistry and Molecular Biology, Medical School, RWTH Aachen University, Pauwelsstrasse 30, 52057 Aachen, Germany; [email protected] (A.B.); jluescher-fi[email protected] (J.L.-F.) * Correspondence: [email protected]; Tel.: +49-241-8088850; Fax: +49-241-8082427 Received: 18 January 2018; Accepted: 27 February 2018; Published: 2 March 2018 Abstract: Regulation of gene expression is achieved by sequence-specific transcriptional regulators, which convey the information that is contained in the sequence of DNA into RNA polymerase activity. This is achieved by the recruitment of transcriptional co-factors. One of the consequences of co-factor recruitment is the control of specific properties of nucleosomes, the basic units of chromatin, and their protein components, the core histones. The main principles are to regulate the position and the characteristics of nucleosomes. The latter includes modulating the composition of core histones and their variants that are integrated into nucleosomes, and the post-translational modification of these histones referred to as histone marks. One of these marks is the methylation of lysine 4 of the core histone H3 (H3K4). While mono-methylation of H3K4 (H3K4me1) is located preferentially at active enhancers, tri-methylation (H3K4me3) is a mark found at open and potentially active promoters. Thus, H3K4 methylation is typically associated with gene transcription. The class 2 lysine methyltransferases (KMTs) are the main enzymes that methylate H3K4. KMT2 enzymes function in complexes that contain a necessary core complex composed of WDR5, RBBP5, ASH2L, and DPY30, the so-called WRAD complex.
    [Show full text]
  • 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]
  • 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.
    [Show full text]
  • Enhancer Priming by H3K4 Methylation Safeguards Germline Competence
    bioRxiv preprint doi: https://doi.org/10.1101/2020.07.07.192427; this version posted July 7, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. Title: Enhancer priming by H3K4 methylation safeguards germline competence 1* 1 1,2 Authors: Tore Bleckwehl ​ ,​ Kaitlin Schaaf ,​ Giuliano Crispatzu ,​ Patricia ​ ​ ​ ​ 1,3 1,4 5 5 Respuela ,​ Michaela Bartusel ,​ Laura Benson ,​ Stephen J. Clark ,​ Kristel M. ​ ​ ​ ​ 6 7 1,8 7,9 Dorighi ,​ Antonio Barral ,​ Magdalena Laugsch ,​ Miguel Manzanares ,​ Joanna ​ ​ ​ ​ 6,10,11 5,12,13 1,3,14* Wysocka ,​ Wolf Reik ,​ Álvaro Rada-Iglesias ​ ​ ​ ​ ​ ​ Affiliations: 1 Center for Molecular Medicine Cologne (CMMC), University of Cologne, Germany. 2 Department of Internal Medicine 2, University Hospital Cologne. 3 Institute of Biomedicine and Biotechnology of Cantabria (IBBTEC), CSIC/University of Cantabria, Spain. 4 Department of Biology, Massachusetts Institute of Technology, USA. 5 Epigenetics Programme, Babraham Institute, Cambridge CB22 3AT, UK. 6 Department of Chemical and Systems Biology, Stanford University School of Medicine, USA. 7 Centro Nacional de Investigaciones Cardiovasculares (CNIC), Spain. 8 Institute of Human Genetics, Heidelberg University Hospital, Germany. 9 Centro de Biología Molecular Severo Ochoa (CBMSO), CSIC-UAM, Spain. 10 Department of Developmental Biology, Stanford University School of Medicine, USA. 11 Howard Hughes Medical Institute, Stanford University School of Medicine, USA. 12 Centre for Trophoblast Research, University of Cambridge, CB2 3EG, UK 13 Wellcome Trust Sanger Institute, Cambridge CB10 1SA, UK.
    [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]
  • Deciphering the Histone Code to Build the Genome Structure
    bioRxiv preprint doi: https://doi.org/10.1101/217190; this version posted November 20, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC 4.0 International license. Deciphering the histone code to build the genome structure Kirti Prakasha,b,c,* and David Fournierd,* aPhysico-Chimie Curie, Institut Curie, CNRS UMR 168, 75005 Paris, France; bOxford Nanoimaging Ltd, OX1 1JD, Oxford, UK; cMicron Advanced Bioimaging Unit, Department of Biochemistry, University of Oxford, Oxford, UK; dFaculty of Biology and Center for Computational Sciences, Johannes Gutenberg University Mainz, 55128 Mainz, Germany; *Correspondence: [email protected], [email protected] Histones are punctuated with small chemical modifications that alter their interaction with DNA. One attractive hypothesis stipulates that certain combinations of these histone modifications may function, alone or together, as a part of a predictive histone code to provide ground rules for chromatin folding. We consider four features that relate histone modifications to chromatin folding: charge neutrali- sation, molecular specificity, robustness and evolvability. Next, we present evidence for the association among different histone modi- fications at various levels of chromatin organisation and show how these relationships relate to function such as transcription, replica- tion and cell division. Finally, we propose a model where the histone code can set critical checkpoints for chromatin to fold reversibly be- tween different orders of the organisation in response to a biological stimulus. DNA | nucleosomes | histone modifications | chromatin domains | chro- mosomes | histone code | chromatin folding | genome structure Introduction The genetic information within chromosomes of eukaryotes is packaged into chromatin, a long and folded polymer of double-stranded DNA, histones and other structural and non- structural proteins.
    [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]
  • Chromatin Dynamics During DNA Replication Raz Bar-Ziv*, Yoav Voichek* and Naama Barkai†
    Downloaded from genome.cshlp.org on October 5, 2021 - Published by Cold Spring Harbor Laboratory Press Chromatin Dynamics during DNA Replication Raz Bar-Ziv*, Yoav Voichek* and Naama Barkai† Affiliations: Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel. *these authors contributed equally to the manuscript †Correspondence to: [email protected] Keywords: Chromatin; Replication; Summary: Chromatin is composed of DNA and histones, which provide a unified platform for regulating DNA-related processes, mostly through their post translational modification. During DNA replication, histone arrangement is perturbed, to first allow progression of DNA polymerase and then during repackaging of the replicated DNA. To study how DNA replication influences the pattern of histone modification, we followed the cell-cycle dynamics of ten histone marks in budding yeast. We find that histones deposited on newly replicated DNA are modified at different rates; while some marks appear immediately upon replication (e.g. H4K16ac, H3K4me1), others increase with transcription-dependent delays (e.g. H3K4me3, H3K36me3). Notably, H3K9ac was deposited as a wave preceding the replication fork by ~5-6 kb. This replication-guided H3K9ac was fully dependent on the acetyltransferase Rtt109, while expression-guided H3K9ac was deposited by Gcn5. Further, topoisomerase depletion intensified H3K9ac in front of the replication fork, and in sites where RNA polymerase II was trapped, suggesting supercoiling stresses trigger H3K9 acetylation. Our results assign complementary roles for DNA replication and gene expression in defining the pattern of histone modification. Introduction: In eukaryotic cells, DNA is wrapped around histone octamers to form nucleosomes, the basic building blocks of the chromatin structure.
    [Show full text]