Epigenetic Aging of Classical Monocytes from Healthy Individuals

Total Page:16

File Type:pdf, Size:1020Kb

Epigenetic Aging of Classical Monocytes from Healthy Individuals bioRxiv preprint doi: https://doi.org/10.1101/2020.05.10.087023; this version posted May 11, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder.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. Epigenetic aging of classical monocytes from healthy individuals Irina Shchukina1,*, Juhi Bagaitkar2,*, Oleg Shpynov1,3,*, Ekaterina Loginicheva1, Sofia Porter1, Denis A. Mogilenko1, Erica Wolin4, Patrick Collins1, German Demidov1,6,7, Mykyta Artomov8,9, Konstantin Zaitsev1, Sviatoslav Sidorov5, Christina Camell5, Monika Bambouskova1, Laura Arthur1, Amanda Swain1, Alexandra Panteleeva1, Aleksei Dievskii3, Evgeny Kurbatsky3, Petr Tsurinov3, Roman Chernyatchik3, Vishwa Deep Dixit5, Marko Jovanovic4, Sheila A. Stewart10, Mark J. Daly8,9,11, Sergey Dmitriev3, Eugene M. Oltz1 and Maxim N. Artyomov1,# AFFILIATIONS 1Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA 2Department of Oral Immunology and Infectious Diseases, University of Louisville, Louisville, KY, USA 3JetBrains Research, St. Petersburg, Russia 4Department of Biological Sciences, Columbia University, New York, NY, USA 5Yale Center for Research on Aging, Yale School of Medicine, New Haven, CT, USA 6Bioinformatics and Genomics Program, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, 08003, Barcelona, Spain. 7Universitat Pompeu Fabra (UPF), 08003, Barcelona, Spain. 8Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA 9Broad Institute, Cambridge, MA, USA 10Department of Cell Biology and Physiology, Washington University School of Medicine, Saint Louis, MO, USA 11Institute for Molecular Medicine, Helsinki, Finland *These authors contributed equally #Correspondence to [email protected] RUNNING TITLE Epigenetic changes in aging human monocytes KEYWORDS Aging, human monocytes, DNA methylation, transcriptomic, epigenetic, metabolomics, proteomic, plasma 1 bioRxiv preprint doi: https://doi.org/10.1101/2020.05.10.087023; this version posted May 11, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder.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. ABSTRACT The impact of healthy aging on molecular programming of immune cells is poorly understood. Here, we report comprehensive characterization of healthy aging in human classical monocytes, with a focus on epigenomic, transcriptomic, and proteomic alterations, as well as the corresponding proteomic and metabolomic data for plasma, using healthy cohorts of 20 young and 20 older individuals (~27 and ~64 years old on average). For each individual, we performed eRRBS-based DNA methylation profiling, which allowed us to identify a set of age-associated differentially methylated regions (DMRs) – a novel, cell-type specific signature of aging in DNA methylome. Optimized ultra-low-input ChIP-seq (ULI-ChIP-seq) data acquisition and analysis pipelines applied to 5 chromatin marks for each individual revealed lack of large- scale age-associated changes in chromatin modifications and allowed us to link hypo- and hypermethylated DMRs to distinct chromatin modification patterns. Specifically, hypermethylation events were associated with H3K27me3 in the CpG islands near promoters of lowly-expressed genes, while hypomethylated DMRs were enriched in H3K4me1 marked regions and associated with normal pattern of expression. Furthermore, hypo- and hypermethylated DMRs followed distinct functional and genetic association patterns. Hypomethylation events were associated with age-related increase of expression of the corresponding genes, providing a link between DNA methylation and age-associated transcriptional changes in primary human cells. Furthermore, these locations were also enriched in genetic regions associated by GWAS with asthma, total blood protein, hemoglobin levels and MS. On the other side, acceleration of epigenetic age in HIV and asthma stems only from changes in hypermethylated DMRs but not from hypomethylated loci. 2 bioRxiv preprint doi: https://doi.org/10.1101/2020.05.10.087023; this version posted May 11, 2020. 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. INTRODUCTION Advanced age, even in healthy individuals, is accompanied by progressive decline of cognitive, metabolic and physiological abilities, and can enhance susceptibility to neurodegenerative, cardiovascular, and chronic inflammatory diseases1,2. Operationally, it is often difficult to determine whether age-associated signatures reflect changes of individual cells or changes in cell type abundances, especially when performing whole tissue transcriptional or epigenetic characterization. For example, T cell numbers are reduced in human blood with age3, complicating comparative analyses of whole blood transcriptional or DNA methylation profiles. And even despite a large amount of clinical and epidemiological data4-8, we understand very little about the nature of age-associated changes in specific primary cell populations of healthy individuals, particularly with respect to age-associated alterations of the epigenetic landscape. To address this question directly, we focused on classical CD14+CD16- monocytes, as they are homogeneous, easily accessible, and relatively abundant in blood, which permits multi-omics profiling of these cells obtained from a single blood draw. Also, monocytes have a short differentiation trajectory from bone marrow progenitors, rendering them informative proxies to understand epigenetic changes that occur with age in human hematopoietic stem cells. Epigenetic aging can manifest in two key aspects: via age-associated changes in chromatin modifications and in DNA methylation. Robustness of the connection between aging and DNA methylation has been well acknowledged9-13, yet in spite the large number of studies, cell specific regions of age- associated DNA methylation/demethylation have not been reported to date. Previous studies have been predominantly using DNA methylation arrays which detect changes of a predefined set of distant solitary cytosines across the genome5. This design, however, inherently prevents identification of differentially methylated regions (DMRs), which are expected to be more biologically relevant compared to changes in single isolated CpG sites. Furthermore, while total level of various chromatin modifications has been characterized across age groups and cell populations using CyTOF14, no genomically resolved profiling of major chromatin modifications (e.g. via ChIP-seq) has been reported for primary human cell types and extent of chromatin changes in primary human cells during aging remains unclear. In this study, we used parallel multi-omics approaches to characterize intracellular states and extracellular environments of monocytes along healthy aging. We profiled DNA methylation, five major chromatin modifications, as well as transcriptional and proteomic states of purified CD14+CD16- monocytes from 20 young (24-30 y. o.) and 20 old (57-70 y. o.) donors. To allow identification of continuous DNA regions that change methylation level with age and comprehensive characterization chromatin state, we utilized enhanced Reduced Representation Bisulfite Sequencing (eRRBS) coupled with the Ultra-Low-Input ChIP-Sequencing (ULI-ChIP-seq)15 approach to profile chromatin modifications from limited input material. Our approach led to identification of more than a thousand DMRs, which could not be achieved via 3 bioRxiv preprint doi: https://doi.org/10.1101/2020.05.10.087023; this version posted May 11, 2020. 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. methylation arrays technology (only ~3% of the DMRs are sufficiently covered in the most commonly used methylation array design). We found no evidence of large-scale remodeling of chromatin modification landscape along the healthy aging yet revealed distinct chromatin features that were characteristic of age- associated DNA hyper- and hypomethylated regions. Integration of the obtained DMR signatures with transcriptional data highlighted connection between age-associated transcriptional alterations and hypomethylated DMRs, while hypermethylated DMRs were not readily associated with transcriptional changes. Together with parallel profiling of plasma proteins and unbiased metabolic profiling from the same individuals, the compendium of data collected in this work comprises a comprehensive aging data resource obtained under stringent inclusion criteria. Easily accessible visualization and exploration of all data from this study, and detailed descriptions of protocols and computational pipelines are available online at http://artyomovlab.wustl.edu/aging/. RESULTS Study design, cohort characteristics, and systemic age-associated changes It has been described that genetic factors, race, sex, body-mass and lifestyles can significantly affect the outcomes of human studies focused on various aspects of aging7,8,10,16,17. Here, we have attempted to assemble a cohort as thoroughly controlled as possible to limit non-age-related contributions in the downstream characterization of monocytes. We used stringent inclusion criteria to eliminate the effect of confounding variables such as inherited genetic traits,
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]
  • 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]
  • 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]
  • Enhancer Priming by H3K4 Methyltransferase MLL4 Controls Cell Fate Transition
    Enhancer priming by H3K4 methyltransferase MLL4 controls cell fate transition Chaochen Wanga, Ji-Eun Leea, Binbin Laia, Todd S. Macfarlanb, Shiliyang Xua, Lenan Zhuanga, Chengyu Liuc, Weiqun Pengd,e, and Kai Gea,1 aLaboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892; bDivision of Developmental Biology, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892; cTransgenic Core, Center for Molecular Medicine, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892; dDepartment of Physics, The George Washington University, Washington, DC 20052; and eDepartment of Anatomy and Regenerative Biology, The George Washington University, Washington, DC 20052 Edited by Eric N. Olson, University of Texas Southwestern Medical Center, Dallas, TX, and approved August 26, 2016 (received for review April 29, 2016) Transcriptional enhancers control cell-type–specific gene expres- excellent models for studying cell fate transition and the underlying sion. Primed enhancers are marked by histone H3 lysine 4 (H3K4) molecular mechanism. mono/di-methylation (H3K4me1/2). Active enhancers are further MLL4 is a major enhancer H3K4 mono- and di-methyltransfer- marked by H3K27 acetylation (H3K27ac). Mixed-lineage leukemia ase with partial functional redundancy with MLL3 in mammalian 4 (MLL4/KMT2D) is a major enhancer H3K4me1/2 methyltransferase cells (7, 8). MLL4 colocalizes with lineage-determining TFs on AEs with functional redundancy with MLL3 (KMT2C). However, its role in during adipogenesis and myogenesis. Furthermore, MLL4 is re- cell fate maintenance and transition is poorly understood. Here, we quired for enhancer activation, cell-type–specific gene expression, show in mouse embryonic stem cells (ESCs) that MLL4 associates and cell differentiation during adipogenesis and myogenesis (8).
    [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]
  • Supplementary Information Hierarchical Chromatin Regulation
    Supplementary information Hierarchical chromatin regulation during blood formation uncovered by single- cell sortChIC Peter Zeller*, Jake Yeung*, Buys Anton de Barbanson, Helena Viñas Gaza, Maria Florescu, and Alexander van Oudenaarden Oncode Institute, Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences) and University Medical Center Utrecht, 3584 CT, Utrecht, The Netherlands *These authors contributed equally Correspondence: [email protected] (A.v.O.) This PDF file includes: Figs. S1 to S6 Tables S2 Other Supplementary Materials for this manuscript include the following: Tables S1 and S3 Figure S1 A FSC SSC counts FSC trigger pulse Hoechst B H3K4me1 H3K4me3 H3K9me3 H3K27me3 1.00 selected excluded 0.50 Fraction of cuts Fraction starting with TA 0.00 10 103 105 10 103 105 10 103 105 10 103 105 Number of Cuts per Cell C 15 H3K4me1 H3K4me3 H3K9me3 H3K27me3 selected excluded 10 density 5 0 0.00 0.25 0.50 0.75 1 0.00 0.25 0.50 0.75 1 0.00 0.25 0.50 0.75 1 0.00 0.25 0.50 0.75 1 Fraction of Cuts in Peaks D H3K4me1 H3K4me3 H3K9me3 H3K27me3 (cuts) 2 scChIC log ChIP ChIP ChIP ChIP log2(counts) log2(counts) log2(counts over input) log2(counts) E H3K9me3 H3K4me1 H3K4me3 H3K27me3 ChIP scChIC ChIP scChIC ChIP scChIC ChIP scChIC 1 0.83 −0.56 −0.69 −0.47 −0.48 −0.58 −0.58 ChIP 0.83 1 −0.58 −0.52 −0.5 −0.39 −0.53 −0.41 H3K9me3 −0.56 −0.58 1 0.82 0.81 0.82 0.5 0.23 ChIP scChIC −0.69 −0.52 0.82 1 0.66 0.88 0.51 0.45 H3K4me1 scChIC −0.47 −0.5 0.81 0.66 1 0.8 0.38 0.15 ChIP −0.48 −0.39 0.82 0.88 0.8 1 0.27 0.14 H3K4me3 scChIC −0.58 −0.53 0.5 0.51 0.38 0.27 1 0.88 ChIP −0.58 −0.41 0.23 0.45 0.15 0.14 0.88 1 H3K27me3 scChIC -1 Pearson Correlation 1 F H3K9me3 Input ChIP H3K9me3/Input (log2) H3K9me3 scChIC 120 mb 140 mb 160 mb 180 mb Chr3 Figure S1.
    [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]