Analysis of Normal Human Mammary Epigenomes Reveals Cell-Specific

Total Page:16

File Type:pdf, Size:1020Kb

Analysis of Normal Human Mammary Epigenomes Reveals Cell-Specific Resource Analysis of Normal Human Mammary Epigenomes Reveals Cell-Specific Active Enhancer States and Associated Transcription Factor Networks Graphical Abstract Authors Davide Pellacani, Misha Bilenky, Nagarajan Kannan, ..., Samuel Aparicio, Martin Hirst, Connie J. Eaves Correspondence [email protected] (M.H.), [email protected] (C.J.E.) In Brief Pellacani et al. present comprehensive histone and DNA modification profiles for four cell types in normal human breast tissue and three immortalized human mammary epithelial cell lines. Analysis of activated enhancers place luminal progenitors in between bipotent progenitor-containing basal cells and nonproliferative luminal cells. Explore consortium data at the Cell Press IHEC webportal at http://www.cell.com/ consortium/IHEC. Highlights d Epigenomes of four normal human breast cell fractions and three cell lines are reported d Luminal progenitor and mature luminal cell epigenomes differ greatly d Enhancers define luminal progenitors as intermediate between basal and luminal cells d Transcription factor binding sites in active enhancers point to distinct regulators Pellacani et al., 2016, Cell Reports 17, 2060–2074 November 15, 2016 ª 2016 The Author(s). http://dx.doi.org/10.1016/j.celrep.2016.10.058 Cell Reports Resource Analysis of Normal Human Mammary Epigenomes Reveals Cell-Specific Active Enhancer States and Associated Transcription Factor Networks Davide Pellacani,1 Misha Bilenky,2 Nagarajan Kannan,1 Alireza Heravi-Moussavi,2 David J.H.F. Knapp,1 Sitanshu Gakkhar,2 Michelle Moksa,3 Annaick Carles,3 Richard Moore,2 Andrew J. Mungall,2 Marco A. Marra,2 Steven J.M. Jones,2 Samuel Aparicio,4,5 Martin Hirst,2,3,* and Connie J. Eaves1,6,* 1Terry Fox Laboratory, BC Cancer Agency, Vancouver, BC V5Z 1L3, Canada 2Canada’s Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, BC V5Z 1L3, Canada 3Michael Smith Laboratories, Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada 4Department of Molecular Oncology, BC Cancer Agency, Vancouver, BC V5Z 1L3, Canada 5Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 2B5, Canada 6Lead Contact *Correspondence: [email protected] (M.H.), [email protected] (C.J.E.) http://dx.doi.org/10.1016/j.celrep.2016.10.058 SUMMARY 3D cultures (Eirew et al., 2008; Kannan et al., 2013; Raouf et al., 2008). Most colonies derived from the BCs contain a The normal adult human mammary gland is a contin- mixture of cells with either basal or luminal features, although uous bilayered epithelial system. Bipotent and sometimes only basal progeny are produced. Human LPs myoepithelial progenitors are prominent and unique contain all of the in vitro progenitor activity of the luminal components of the outer (basal) layer. The inner compartment and produce colonies of cells expressing luminal (luminal) layer includes both luminal-restricted pro- features exclusively. Because LCs lack substantial proliferative genitors and a phenotypically separable fraction potential in vitro, they are assumed to represent a mature subset of luminal cells (Fridriksdottir et al., 2015; Kannan et al., 2013). that lacks progenitor activity. We now report an epi- When transplanted into immunodeficient mice, a small propor- genomic comparison of these three subsets with one tion (<1%) of human BCs, but not LPs or LCs, can also individu- another, with their associated stromal cells, and with ally regenerate normal-appearing bilayered mammary structures three immortalized, non-tumorigenic human mam- that contain the same spectrum of cell types and progenitors mary cell lines. Each genome-wide analysis contains found in the normal human gland. This property suggests the profiles for six histone marks, methylated DNA, and cells of origin of these structures represent a human mammary RNA transcripts. Analysis of these datasets shows stem cell population (Eirew et al., 2008; Kuperwasser et al., that each cell type has unique features, primarily 2004; Lim et al., 2009; Nguyen et al., 2014). within genomic regulatory regions, and that the cell Despite these advances, the molecular regulators that specify lines group together. Analyses of the promoter and the unique features of each of these normal human mammary enhancer profiles place the luminal progenitors in subsets have only recently become amenable to interrogation. Transcriptome profiling (Choudhury et al., 2013; Gascard et al., between the basal cells and the non-progenitor 2015; Huh et al., 2015; Kannan et al., 2013; Lim et al., 2009, luminal subset. Integrative analysis reveals networks 2010; Makarem et al., 2013; Maruyama et al., 2011; Raouf of subset-specific transcription factors. et al., 2008; Dos Santos et al., 2015), together with functional studies, has enabled several key transcription factors (TFs) to INTRODUCTION be identified in the subpopulations of normal human as well as analogous mouse mammary cell types (Fu et al., 2014; Visvader The normal adult human female mammary gland is a continuous and Stingl, 2014). Thus implicated in BCs are SLUG, TP63 and bilayered, branching epithelial structure containing three pheno- TP53, SOX9, STAT3, MYC, and TAZ. In cells with luminal fea- typically distinct subsets of cells. These can now be isolated and tures, CEBPB and NOTCH3 (Raouf et al., 2008), together with biologically distinguished (Fu et al., 2014). They are referred to as GATA3, ELF5 (Chakrabarti et al., 2012), and FOXA1, which reg- basal cells (BCs), luminal progenitors (LPs), and luminal cells ulates estrogen receptor activity (Theodorou et al., 2013), have (LCs) because their surface marker expression profiles identify been identified as important. their location within the outer (basal) or inner (luminal) layer of Previous human mammary epigenome profiles produced as the gland. A prominent fourth cell population in the breast is part of the Roadmap Epigenomics Consortium (Roadmap Epi- comprised of mesodermally derived stromal cells (SCs). Approx- genomics Consortium et al., 2015) have provided an initial global imately 20%–30% of human mammary BCs and LPs have annotation of active enhancer regions (Gascard et al., 2015). We progenitor (clonogenic) activity demonstrable in both 2D and now report a more extensive set of reference epigenome profiles 2060 Cell Reports 17, 2060–2074, November 15, 2016 ª 2016 The Author(s). This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/). A Normal FACS 6 samples pooled Molecular analyses Integrative analyses breast ( 5x107 cells per fraction) samples RNA Datasets n = 6 RNA-seq CD45- CD31- LC DNA Expression Chromatin states single cells WGBS LC LP Chromatin LP H3K4me1 H3K4me3 Enhancers BC BC H3K9me3 EpCAM-PE Promoters H3K27ac SC SC H3K27me3 Transcription H3K36me3 factor CD49f-APC networks Colony assays B C Type Study LP vs BC LP vs LC Up: 806 BC CEMT Do Do BC Nguyen BC 1 Nguyen 1 BC Nguyen (RPKM) (RPKM) 10 myo REMC 10 myo REMC myo REMC LC log LP CEMT BC log LP Nguyen LP Nguyen LP Nguyen 1 1 lum REMC LP log10(RPKM) LP log10(RPKM) LC CEMT lum REMC BC vs LC DE genes lum REMC Do MCF10A CEMT LP vs BC 184−L9 CEMT 184−L2 CEMT 1 vHMEC REMC (RPKM) vHMEC REMC 10 LP vs LC SC CEMT log fibr REMC LC fibr REMC BC vs LC 0.20 0.15 0.10 0.05 0.00 1 246810 BC log10(RPKM) Dist. = 1−Spearman | Link. = complete absolute log2( D E Type Study myo REMC BC CEMT myo REMC SC CEMT LP CEMT LC CEMT lum REMC lum REMC fibr REMC fibr REMC vHMEC REMC MCF10A CEMT 184−L9 CEMT 184−L2 CEMT 0.4 0.3 0.2 0.1 0.0 Dist. = 1−Spearman | Link. = complete Figure 1. Isolation of Purified Cell Subsets and Epigenomic Profiling (A) Experimental design. (B) Unsupervised clustering of RNA-sequencing experiments comparing the samples analyzed in this study (CEMT) with other studies (Nguyen et al., 2015; Roadmap Epigenomics Consortium et al., 2015; Gascard et al., 2015). (legend continued on next page) Cell Reports 17, 2060–2074, November 15, 2016 2061 that include a separate analysis of human LPs and LCs and pro- These findings are consistent with and extend previously re- files for SCs isolated from the same normal breast tissue, as well ported findings obtained both by RNA-seq (Gascard et al., as profiles generated from three widely used immortalized, but 2015; Nguyen et al., 2015) and by 30 tag-based and microarray non-tumorigenic, human mammary epithelial cell lines: i.e., analysis (Kannan et al., 2013; Lim et al., 2009; Raouf et al., MCF-10A cells (Debnath et al., 2003) and 184-hTERT-L2 (184- 2008), including an identification of many of the same genes as L2) and 184-hTERT-L9 (184-L9) cells (Burleigh et al., 2015). ‘‘BC specific’’ (e.g., TP63 (deltaN), ACTA2, MME, KRT14, The results demonstrate that the cell lines are not representative THY1, and KRT5) or ‘‘LP specific’’ (e.g., MUC1, PROM1, KIT, of any freshly isolated normal human mammary subset. They KRT18, EPCAM, and KRT8)(Figure S2A). Similar clustering have also allowed a hierarchy of enhancer states in normal and differences between the same four cell types were human mammary tissue and their corresponding cell-specific observed when either microRNA (miRNA) or large intergenic TF networks to be identified. noncoding RNA (lincRNA) profiles were analyzed (Figures 1D, S2D, and S2E). RESULTS AND DISCUSSION Notably, the transcriptomes for all three immortalized human mammary cell lines clustered independently from both the pri- Generation and Validation of Comprehensive Reference mary epithelial subsets and the SCs isolated directly from the Epigenomes same freshly dissociated normal human breast tissue (Figure 1B). Reference epigenomic profiles were generated following the rec- Moreover, 66 of the 100 genes whose differential expression ommendations of the International Human Epigenome Con- contributed most to the separate clustering of the mammary sortium (IHEC; http://ihec-epigenomes.org).
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]
  • The International Human Epigenome Consortium (IHEC): a Blueprint for Scientific Collaboration and Discovery
    The International Human Epigenome Consortium (IHEC): A Blueprint for Scientific Collaboration and Discovery Hendrik G. Stunnenberg1#, Martin Hirst2,3,# 1Department of Molecular Biology, Faculties of Science and Medicine, Radboud University, Nijmegen, The Netherlands 2Department of Microbiology and Immunology, Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada V6T 1Z4. 3Canada’s Michael Smith Genome Science Center, BC Cancer Agency, Vancouver, BC, Canada V5Z 4S6 #Corresponding authors [email protected] [email protected] Abstract The International Human Epigenome Consortium (IHEC) coordinates the generation of a catalogue of high-resolution reference epigenomes of major primary human cell types. The studies now presented (cell.com/XXXXXXX) highlight the coordinated achievements of IHEC teams to gather and interpret comprehensive epigenomic data sets to gain insights in the epigenetic control of cell states relevant for human health and disease. One of the great mysteries in developmental biology is how the same genome can be read by cellular machinery to generate the plethora of different cell types required for eukaryotic life. As appreciation grew for the central roles of transcriptional and epigenetic mechanisms in specification of cellular fates and functions, researchers around the world encouraged scientific funding agencies to develop an organized and standardized effort to exploit epigenomic assays to shed additional light on this process (Beck, Olek et al. 1999, Jones and Martienssen 2005, American Association for Cancer Research Human Epigenome Task and European Union 2008). In March 2009, leading scientists and international health research funding agency representatives were invited to a meeting in Bethesda (MD, USA) to gauge the level of interest in an international epigenomics project and to identify potential areas of focus.
    [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]
  • A Machine Learning Framework for Precise 3D Domain Boundary Prediction at Base-Level Resolution
    bioRxiv preprint doi: https://doi.org/10.1101/2020.09.03.282186; this version posted September 29, 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 4.0 International license. preciseTAD: A machine learning framework for precise 3D domain boundary prediction at base-level resolution Spiro C. Stilianoudakis1 ([email protected]), Mikhail G. Dozmorov1* (mikhail.dozmorov@ vcuhealth.org) 1 Dept. of Biostatistics, Virginia Commonwealth University, Richmond, VA, 23298, USA * To whom correspondence should be addressed Abstract The low resolution of high-throughput chromatin conformation capture data limits the precise mapping of boundaries of topologically associating domains and chromatin loops. We developed preciseTAD, an optimized random forest model trained on high-resolution genome annotation data (e.g., CTCF ChIP- seq) to predict the location of domain boundaries at base-level resolution. Distance between boundaries and annotations, random under-sampling, and transcription factor binding sites resulted in best model performance. preciseTAD boundaries were more enriched for CTCF, RAD21, SMC3, and ZNF143, and conserved across cell lines. Using genome annotations, pre-trained models can detect boundaries in cells without Hi-C data. preciseTAD is available at https://bioconductor.org/packages/preciseTAD Keywords Hi-C, TAD, machine learning, random forest, DBSCAN Background The advent of chromosome conformation capture (3C) sequencing technologies, and its successor Hi-C, have revealed a hierarchy of the 3-dimensional (3D) structure of the human genome such as chromatin loops [1], Topologically Associating Domains (TADs) [2,3], and A/B compartments [4].
    [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]
  • 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]
  • 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]
  • Generative Modeling of Multi-Mapping Reads with Mhi-C Advances Analysis of Hi-C Studies Ye Zheng1, Ferhat Ay2,3, Sunduz Keles1,4*
    TOOLS AND RESOURCES Generative modeling of multi-mapping reads with mHi-C advances analysis of Hi-C studies Ye Zheng1, Ferhat Ay2,3, Sunduz Keles1,4* 1Department of Statistics, University of Wisconsin-Madison, Madison, United States; 2La Jolla Institute for Allergy and Immunology, La Jolla, United States; 3School of Medicine, University of California, San Diego, La Jolla, United States; 4Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, United States Abstract Current Hi-C analysis approaches are unable to account for reads that align to multiple locations, and hence underestimate biological signal from repetitive regions of genomes. We developed and validated mHi-C, a multi-read mapping strategy to probabilistically allocate Hi-C multi-reads. mHi-C exhibited superior performance over utilizing only uni-reads and heuristic approaches aimed at rescuing multi-reads on benchmarks. Specifically, mHi-C increased the sequencing depth by an average of 20% resulting in higher reproducibility of contact matrices and detected interactions across biological replicates. The impact of the multi-reads on the detection of significant interactions is influenced marginally by the relative contribution of multi-reads to the sequencing depth compared to uni-reads, cis-to-trans ratio of contacts, and the broad data quality as reflected by the proportion of mappable reads of datasets. Computational experiments highlighted that in Hi-C studies with short read lengths, mHi-C rescued multi-reads can emulate the effect of longer reads. mHi-C also revealed biologically supported bona fide promoter-enhancer interactions and topologically associating domains involving repetitive genomic regions, thereby *For correspondence: unlocking a previously masked portion of the genome for conformation capture studies.
    [Show full text]
  • Next Generation Sequencing: Advances in Characterizing the Methylome
    Genes 2010, 1, 143-165; doi:10.3390/genes1020143 OPEN ACCESS genes ISSN 2073-4425 www.mdpi.com/journal/genes Review Next Generation Sequencing: Advances in Characterizing the Methylome Kristen H. Taylor 1, Huidong Shi 2 and Charles W. Caldwell 1,* 1 University of Missouri-Columbia School of Medicine, Ellis Fischel Cancer Center, Columbia, MO 65212, USA; E-Mail: [email protected] 2 Medical College of Georgia, Augusta, GA 30912, USA; E-Mail: [email protected] * Author to whom correspondence should be addressed; E-Mail: [email protected]; Tel.: +1-573-882-1283; Fax: +1-573-884-4612. Received: 3 May 2010; in revised form: 22 June 2010 / Accepted: 28 June 2010 / Published: 1 July 2010 Abstract: Epigenetic modifications play an important role in lymphoid malignancies. This has been evidenced by the large body of work published using microarray technologies to generate methylation profiles for numerous types and subtypes of lymphoma and leukemia. These studies have shown the importance of defining the epigenome so that we can better understand the biology of lymphoma. Recent advances in DNA sequencing technology have transformed the landscape of epigenomic analysis as we now have the ability to characterize the genome-wide distribution of chromatin modifications and DNA methylation using next-generation sequencing. To take full advantage of the throughput of next-generation sequencing, there are many methodologies that have been developed and many more that are currently being developed. Choosing the appropriate methodology is fundamental to the outcome of next-generation sequencing studies. In this review, published technologies and methodologies applicable to studying the methylome are presented.
    [Show full text]
  • Association of Cnvs with Methylation Variation
    www.nature.com/npjgenmed ARTICLE OPEN Association of CNVs with methylation variation Xinghua Shi1,8, Saranya Radhakrishnan2, Jia Wen1, Jin Yun Chen2, Junjie Chen1,8, Brianna Ashlyn Lam1, Ryan E. Mills 3, ✉ ✉ Barbara E. Stranger4, Charles Lee5,6,7 and Sunita R. Setlur 2 Germline copy number variants (CNVs) and single-nucleotide polymorphisms (SNPs) form the basis of inter-individual genetic variation. Although the phenotypic effects of SNPs have been extensively investigated, the effects of CNVs is relatively less understood. To better characterize mechanisms by which CNVs affect cellular phenotype, we tested their association with variable CpG methylation in a genome-wide manner. Using paired CNV and methylation data from the 1000 genomes and HapMap projects, we identified genome-wide associations by methylation quantitative trait locus (mQTL) analysis. We found individual CNVs being associated with methylation of multiple CpGs and vice versa. CNV-associated methylation changes were correlated with gene expression. CNV-mQTLs were enriched for regulatory regions, transcription factor-binding sites (TFBSs), and were involved in long- range physical interactions with associated CpGs. Some CNV-mQTLs were associated with methylation of imprinted genes. Several CNV-mQTLs and/or associated genes were among those previously reported by genome-wide association studies (GWASs). We demonstrate that germline CNVs in the genome are associated with CpG methylation. Our findings suggest that structural variation together with methylation may affect cellular phenotype. npj Genomic Medicine (2020) 5:41 ; https://doi.org/10.1038/s41525-020-00145-w 1234567890():,; INTRODUCTION influence transcript regulation is DNA methylation, which involves The extent of genetic variation that exists in the human addition of a methyl group to cytosine residues within a CpG population is continually being characterized in efforts to identify dinucleotide.
    [Show full text]