Molecular Function and Regulation of the Histone Methyltransferase, Ezh2

Total Page:16

File Type:pdf, Size:1020Kb

Molecular Function and Regulation of the Histone Methyltransferase, Ezh2 Molecular function and regulation of the histone methyltransferase, Ezh2 By Susan C. Wu A dissertation submitted to the faculty of the University of North Carolina at Chapel Hill in partial fulfillment of the requirements for the degree of Doctor of Philosophy in the Department of Biochemistry and Biophysics Chapel Hill 2011 Approved by: Dr. Yi Zhang Dr. Yue Xiong Dr. William Marzluff Dr. Adrienne Cox Dr. Young Whang ©2011 Susan C. Wu ALL RIGHTS RESERVED ii ABSTRACT Susan C. Wu Molecular function and regulation of the histone methyltransferase, Ezh2 (Under the direction of Dr. Yi Zhang) EZH2 (Enhancer of zeste 2) is an important developmental regulator that plays an important role in numerous biological processes including multi-cellular development, stem cell biology, and cancer development. As an H3K27 histone methyltransferase, EZH2 is an epigenetic regulator that functions to repress transcription. It is often upregulated in human cancers and its oncogenic potential is thought to stem from its role in cell proliferation and repression of tumor suppressor genes. Hence, regulation of EZH2 function is important for normal cellular growth. In this dissertation, the molecular function and regulation of EZH2 are investigated. I demonstrate that genomic targets of EZH2 not only include protein- coding genes, but also encompasses lncRNAs (long non-coding RNAs), a class of RNAs that account for the vast majority of the mammalian transcriptome. Using a custom-designed microarray, I show that lncRNAs exhibit cell type specific expression patterns and that their expression levels correlate with the epigenetic marks, H3K4me3 and H3K27me3. Consistent with this finding, knockdown of EZH2 results in the derepression of many lncRNAs. With regard to EZH2 regulation, I provide evidence that EZH2 contains numerous phosphorylation sites although their functions are currently unknown. Characterization of two phosphorylation sites, threonines 345 and 487, has revealed important aspects of EZH2 iii regulation. Through in vitro and in vivo experiments, I show that phosphorylation of these two residues can be mediated by the cyclin dependent kinase, CDK1. Consistent with the cell cycle phase that exhibits peak CDK1 activity, mitotic cell extracts are enriched for phospho-EZH2. Importantly, when EZH2 is phosphorylated at T345 and T487, the stability of the protein is compromised as EZH2 is targeted for ubiquitination and degradation by the proteasome. Collectively, this work reveals a novel class of EZH2 targets and provides a mechanism by which EZH2 protein levels are regulated in the cell. iv To my father, Y. John Wu, who persistently instilled my interest in science at an early age. Without his support and the countless lectures encompassing broad scientific topics, I would not be where I am today. v ACKNOWLEDGEMENTS I must take a moment to acknowledge the extraordinary role that the following people had in shaping this work. First and foremost, I would like to thank Dr. Yi Zhang for his guidance during my tenure as a graduate student. I am indebted to him for his critical comments of my work as well as the opportunities that he has provided with me, both of which have pushed me to become a better scientist. I would also like to acknowledge my dissertation committee members: Drs. Yue Xiong, William Marzluff, Adrienne Cox, and Young Whang. Their continued support and scientific guidance were undoubtedly instrumental to the completion of this work. In addition, I would also like to thank Dr. Jean Cook who generously invested her time in providing me with the scientific expertise that I needed. I was also fortunate to work with an outstanding group of scientists on a day to day basis. Thank you to past and present members of the Zhang lab who provided me with both the encouragement and technical expertise that I needed over the years. In particular, I would like to thank Dr. Eric Kallin who I not only had the privilege of collaborating with, but who also contributed to my personal well-being. Drs. Nara Lee and Rob Klose were instrumental in my scientific development and I would be remiss to not thank them. I also have to thank vi Jake Dmochowski, an outstanding undergraduate student, who not only worked with me, but also kept me on my toes with his inquisitive questions. In retrospect, I have learned that graduate school is not for the faint of heart and that the challenges brought forth in front of me undoubtedly required the loving support of the wonderful people in my life. First and foremost, I have to thank my parents, Y. John and Grace Wu who have pushed me to realize my potential and unconditionally supported my decision to attend graduate school. I also have to thank my brothers Jeff and Greg as well as my sister-in-law, Helen, who have provided me with so much laughter over the years. I am extremely grateful to be a part of such an incredible family. In addition, I want to thank all of the friends that I have made during my time in Chapel Hill. In particular, I have to acknowledge Elizabeth Dorn who has been a fabulous co-pilot to have in this journey. Finally, none of this would be possible without the constant support and encouragement from my husband, Chris Winkler. I would like to thank him for his extraordinary patience and eternal positivity over the years. His companionship is truly special and I am sincerely grateful for it. vii TABLE OF CONTENTS Page LIST OF TABLES....................................................................................................................xi LIST OF FIGURES.................................................................................................................xii LIST OF ABBREVIATIONS.................................................................................................xiv CHAPTER I. INTRODUCTION..................................................................................................1 i. Epigenetic regulation of gene expression.................................................3 ii. Histone methylation.................................................................................5 iii. Polycomb group of proteins.....................................................................8 iv. Polycomb repressive complexes............................................................10 v. Functions of H3K27 methylation...........................................................15 vi. Recruitment of PRC2 to genomic targets...............................................18 vii. ES cell pluripotency, self-renewal, and differentiation..........................22 viii. EZH2 and human cancers......................................................................23 ix. Research summary.................................................................................25 x. References..............................................................................................35 II. ROLE OF H3K27 METHYLATION IN THE REGULATION OF LONG NON-CODING RNA EXPRESSION..................................................................50 viii i. Abstract..................................................................................................51 ii. Introduction............................................................................................52 iii. Results....................................................................................................54 iv. Discussion..............................................................................................60 v. Experimental procedures........................................................................63 vi. References..............................................................................................73 II. EZH2 IS A THREONINE PHOSPHORYLATED..............................................77 i. Abstract..................................................................................................78 ii. Introduction............................................................................................79 iii. Results....................................................................................................83 iv. Discussion..............................................................................................89 v. Experimental procedures........................................................................92 vi. References............................................................................................103 III. CDK1-MEDIATED PHOSPHORYLATION OF EZH2 LEADS TO DEGRADATION...............................................................................................108 i. Abstract................................................................................................109 ii. Introduction..........................................................................................110 iii. Results..................................................................................................113 iv. Discussion............................................................................................123 v. Experimental procedures......................................................................128 vi. References............................................................................................148 IV. SUMMARY AND CONCLUDING REMARKS..............................................152 i. Epigenetic regulation of long non-coding RNAs..................................153 ii. Long non-coding RNAs and ES cell biology.......................................156 ix iii. Investigating
Recommended publications
  • O-Glcnacylation Regulates the Stability and Enzymatic Activity of the Histone Methyltransferase EZH2
    O-GlcNAcylation regulates the stability and enzymatic activity of the histone methyltransferase EZH2 Pei-Wen Loa, Jiun-Jie Shieb, Chein-Hung Chena, Chung-Yi Wua, Tsui-Ling Hsua, and Chi-Huey Wonga,1 aGenomics Research Center, Academia Sinica, Taipei 115, Taiwan; and bInstitute of Chemistry, Academia Sinica, Taipei 115, Taiwan Contributed by Chi-Huey Wong, May 16, 2018 (sent for review February 1, 2018; reviewed by Michael D. Burkart, Benjamin G. Davis, and Gerald W. Hart) Protein O-glycosylation by attachment of β-N-acetylglucosamine maintenance and differentiation in embryonic stem cells (14, 15). (GlcNAc) to the Ser or Thr residue is a major posttranslational It was suggested that O-GlcNAcylation might play an important glycosylation event and is often associated with protein folding, role in the regulation of PRC1-mediated gene expression, and stability, and activity. The methylation of histone H3 at Lys-27 along this line the O-GlcNAcylation of EZH2 at S76 in the PRC2 catalyzed by the methyltransferase EZH2 was known to suppress complex was reported to stablize EZH2 in our previous study (16). gene expression and cancer development, and we previously The PRC2 complex is composed of Enhancer of zeste 2 (EZH2), reported that the O-GlcNAcylation of EZH2 at S76 stabilized Suppressor of Zeste 12 (Suz12), Extraembryonic endoderm (EED), EZH2 and facilitated the formation of H3K27me3 to inhibit tumor AE binding protein 2 (AEBP2), and retinoblastoma binding protein suppression. In this study, we employed a fluorescence-based method 4/7 (RBBP4/7) (17, 18). Within the PRC2 complex, EZH2 catalyzes the di- and trimethylation of histone H3 at lysine 27 (K27) to form of sugar labeling combined with mass spectrometry to investigate H3K27me2/3 to regulate embryonic and cancer development EZH2 glycosylation and identified five O-GlcNAcylation sites.
    [Show full text]
  • The Mutational Landscape of Myeloid Leukaemia in Down Syndrome
    cancers Review The Mutational Landscape of Myeloid Leukaemia in Down Syndrome Carini Picardi Morais de Castro 1, Maria Cadefau 1,2 and Sergi Cuartero 1,2,* 1 Josep Carreras Leukaemia Research Institute (IJC), Campus Can Ruti, 08916 Badalona, Spain; [email protected] (C.P.M.d.C); [email protected] (M.C.) 2 Germans Trias i Pujol Research Institute (IGTP), Campus Can Ruti, 08916 Badalona, Spain * Correspondence: [email protected] Simple Summary: Leukaemia occurs when specific mutations promote aberrant transcriptional and proliferation programs, which drive uncontrolled cell division and inhibit the cell’s capacity to differentiate. In this review, we summarize the most frequent genetic lesions found in myeloid leukaemia of Down syndrome, a rare paediatric leukaemia specific to individuals with trisomy 21. The evolution of this disease follows a well-defined sequence of events and represents a unique model to understand how the ordered acquisition of mutations drives malignancy. Abstract: Children with Down syndrome (DS) are particularly prone to haematopoietic disorders. Paediatric myeloid malignancies in DS occur at an unusually high frequency and generally follow a well-defined stepwise clinical evolution. First, the acquisition of mutations in the GATA1 transcription factor gives rise to a transient myeloproliferative disorder (TMD) in DS newborns. While this condition spontaneously resolves in most cases, some clones can acquire additional mutations, which trigger myeloid leukaemia of Down syndrome (ML-DS). These secondary mutations are predominantly found in chromatin and epigenetic regulators—such as cohesin, CTCF or EZH2—and Citation: de Castro, C.P.M.; Cadefau, in signalling mediators of the JAK/STAT and RAS pathways.
    [Show full text]
  • Automethylation of PRC2 Promotes H3K27 Methylation and Is Impaired in H3K27M Pediatric Glioma
    Downloaded from genesdev.cshlp.org on October 5, 2021 - Published by Cold Spring Harbor Laboratory Press Automethylation of PRC2 promotes H3K27 methylation and is impaired in H3K27M pediatric glioma Chul-Hwan Lee,1,2,7 Jia-Ray Yu,1,2,7 Jeffrey Granat,1,2,7 Ricardo Saldaña-Meyer,1,2 Joshua Andrade,3 Gary LeRoy,1,2 Ying Jin,4 Peder Lund,5 James M. Stafford,1,2,6 Benjamin A. Garcia,5 Beatrix Ueberheide,3 and Danny Reinberg1,2 1Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, New York 10016, USA; 2Howard Hughes Medical Institute, Chevy Chase, Maryland 20815, USA; 3Proteomics Laboratory, New York University School of Medicine, New York, New York 10016, USA; 4Shared Bioinformatics Core, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA; 5Department of Biochemistry and Molecular Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA The histone methyltransferase activity of PRC2 is central to the formation of H3K27me3-decorated facultative heterochromatin and gene silencing. In addition, PRC2 has been shown to automethylate its core subunits, EZH1/ EZH2 and SUZ12. Here, we identify the lysine residues at which EZH1/EZH2 are automethylated with EZH2-K510 and EZH2-K514 being the major such sites in vivo. Automethylated EZH2/PRC2 exhibits a higher level of histone methyltransferase activity and is required for attaining proper cellular levels of H3K27me3. While occurring inde- pendently of PRC2 recruitment to chromatin, automethylation promotes PRC2 accessibility to the histone H3 tail. Intriguingly, EZH2 automethylation is significantly reduced in diffuse intrinsic pontine glioma (DIPG) cells that carry a lysine-to-methionine substitution in histone H3 (H3K27M), but not in cells that carry either EZH2 or EED mutants that abrogate PRC2 allosteric activation, indicating that H3K27M impairs the intrinsic activity of PRC2.
    [Show full text]
  • Acetyltransferase 10 Protein in DNA Methylation and Genomic Imprinting
    Article The Role of N-a-acetyltransferase 10 Protein in DNA Methylation and Genomic Imprinting Graphical Abstract Authors Chen-Cheng Lee, Shih-Huan Peng, Genomic imprinting Li Shen, ..., Yu-Ting Yan, Yi Zhang, Paternally imprinted gene Maternally imprinted gene Li-Jung Juan ICR ICR Correspondence ICR ICR [email protected] (Y.Z.), Naa10-KO or WT Naa10p clinical mutation: [email protected] (L.-J.J.) S37P, V107F or R116W Dnmt1 Naa10p Naa10p Dnmt1 X In Brief X Mut X X Lee et al. reveal an unexpected function G H19 G H19 for Naa10p, which is primarily known to ICR of imprinted allele (ex: H19) ICR of imprinted allele (ex: H19) acetylate nascent peptides from ribosomes, in maintaining global DNA Methylated CpG H3K9me3/H4K20me3 methylation and marking the imprinted Unmethylated CpG H3K9Ac allele for genomic imprinting Imprinting maintenance Imprinting dysregulation maintenance. Their results suggest that Successful development Developmental defect Embryonic lethality defects in DNA methylation and genomic Growth retardation imprinting may contribute to Naa10p- Brain disorder associated Ogden syndrome. Maternal-effect lethality Highlights d Naa10p KO causes defects in genomic imprinting and embryonic development d Naa10p maintains Dnmt1 activity by facilitating Dnmt1 binding to DNA substrate d Naa10p selectively binds to ICRs of the imprinted allele via non-methylated GCXGXG d Ogden syndrome-causing Naa10p mutation disrupts ICR binding of Naa10p and Dnmt1 Lee et al., 2017, Molecular Cell 68, 1–15 October 5, 2017 ª 2017 Elsevier Inc. http://dx.doi.org/10.1016/j.molcel.2017.08.025 Please cite this article in press as: Lee et al., The Role of N-a-acetyltransferase 10 Protein in DNA Methylation and Genomic Imprinting, Molecular Cell (2017), http://dx.doi.org/10.1016/j.molcel.2017.08.025 Molecular Cell Article The Role of N-a-acetyltransferase 10 Protein in DNA Methylation and Genomic Imprinting Chen-Cheng Lee,1,9 Shih-Huan Peng,1,2,9 Li Shen,3,8,9 Chung-Fan Lee,1 Ting-Huei Du,1 Ming-Lun Kang,1 Guo-Liang Xu,4 Anup K.
    [Show full text]
  • Polycomb Repressor Complex 2 Function in Breast Cancer (Review)
    INTERNATIONAL JOURNAL OF ONCOLOGY 57: 1085-1094, 2020 Polycomb repressor complex 2 function in breast cancer (Review) COURTNEY J. MARTIN and ROGER A. MOOREHEAD Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON N1G2W1, Canada Received July 10, 2020; Accepted September 7, 2020 DOI: 10.3892/ijo.2020.5122 Abstract. Epigenetic modifications are important contributors 1. Introduction to the regulation of genes within the chromatin. The poly- comb repressive complex 2 (PRC2) is a multi‑subunit protein Epigenetic modifications, including DNA methylation complex that is involved in silencing gene expression through and histone modifications, play an important role in gene the trimethylation of lysine 27 at histone 3 (H3K27me3). The regulation. The dysregulation of these modifications can dysregulation of this modification has been associated with result in pathogenicity, including tumorigenicity. Research tumorigenicity through the increased repression of tumour has indicated an important influence of the trimethylation suppressor genes via condensing DNA to reduce access to the modification at lysine 27 on histone H3 (H3K27me3) within transcription start site (TSS) within tumor suppressor gene chromatin. This methylation is involved in the repression promoters. In the present review, the core proteins of PRC2, as of multiple genes within the genome by condensing DNA well as key accessory proteins, will be described. In addition, to reduce access to the transcription start site (TSS) within mechanisms controlling the recruitment of the PRC2 complex gene promoter sequences (1). The recruitment of H1.2, an H1 to H3K27 will be outlined. Finally, literature identifying the histone subtype, by the H3K27me3 modification has been a role of PRC2 in breast cancer proliferation, apoptosis and suggested as a mechanism for mediating this compaction (1).
    [Show full text]
  • Engaging Chromatin: PRC2 Structure Meets Function
    www.nature.com/bjc REVIEW ARTICLE Engaging chromatin: PRC2 structure meets function Paul Chammas1, Ivano Mocavini1 and Luciano Di Croce1,2,3 Polycomb repressive complex 2 (PRC2) is a key epigenetic multiprotein complex involved in the regulation of gene expression in metazoans. PRC2 is formed by a tetrameric core that endows the complex with histone methyltransferase activity, allowing it to mono-, di- and tri-methylate histone H3 on lysine 27 (H3K27me1/2/3); H3K27me3 is a hallmark of facultative heterochromatin. The core complex of PRC2 is bound by several associated factors that are responsible for modulating its targeting specificity and enzymatic activity. Depletion and/or mutation of the subunits of this complex can result in severe developmental defects, or even lethality. Furthermore, mutations of these proteins in somatic cells can be drivers of tumorigenesis, by altering the transcriptional regulation of key tumour suppressors or oncogenes. In this review, we present the latest results from structural studies that have characterised PRC2 composition and function. We compare this information with data and literature for both gain-of function and loss-of-function missense mutations in cancers to provide an overview of the impact of these mutations on PRC2 activity. British Journal of Cancer (2020) 122:315–328; https://doi.org/10.1038/s41416-019-0615-2 BACKGROUND and embryonic ectoderm development (EED) (Table 1). These Transcriptional diversity is one of the hallmarks of cellular three proteins form the minimal core that confers histone identity. It is largely regulated at the level of chromatin, where methyltransferase (HMT) activity. A fourth factor, retinoblastoma- different protein complexes act as initiators, enhancers and/or binding protein (RBBP)4/7 (also known as RBAP48/46), has a repressors of transcription.
    [Show full text]
  • Study Identifies a Protein Complex Possibly Crucial for Triggering Embryo Development 6 January 2010
    Study identifies a protein complex possibly crucial for triggering embryo development 6 January 2010 modifications that often prevent them from curbing the proliferation of cancer cells. The discovery may also have implications for stem cell research by providing a tool to quickly reprogram adult cells to possess the same attributes as embryonic stem cells, but without the ethical or safety issues of cells currently used for such studies. The results of the study appear on- line in the Jan. 6, 2010 issue of the journal Nature. "The implications of such research have always been clear, and that is why for years researchers have tried to identify a factor responsible for erasing these epigenetic markers," said senior This is a fertilized human egg, prior to division. Two nuclei (one from the egg and one from the sperm) can author Yi Zhang, Ph.D., Howard Hughes Medical be seen in the center. Credit: Photo courtesy of Stan Institute Investigator and Kenan Distinguished Beyler, Ph.D. UNC A.R.T. laboratory. Professor of biochemistry and biophysics at UNC. He is also a member of the UNC Lineberger Comprehensive Cancer Center. The DNA contained within each of our cells is Epigenetic markers are essentially chemical tags exactly the same, yet different types of cells - skin attached to the genomes of each cell, determining cells, heart cells, brain cells - perform very different which genes will be turned on or off and, ultimately, functions. The ultimate fate of these cells is what role that cell type will have in the body. One encoded not just in the DNA, but in a specific way this comes about is through DNA methylation, pattern of chemical modifications that overlay the a process by which methyl groups are stamped DNA structure.
    [Show full text]
  • Targeting the MTF2-MDM2 Axis Sensitizes Refractory Acute Myeloid Leukemia to Chemotherapy
    Author Manuscript Published OnlineFirst on August 16, 2018; DOI: 10.1158/2159-8290.CD-17-0841 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Targeting the MTF2-MDM2 Axis Sensitizes Refractory Acute Myeloid Leukemia to Chemotherapy Harinad B. Maganti1,2,3†, Hani Jrade1,2,4†, Christopher Cafariello1,2,4, Janet L. Manias Rothberg1,2,4, Christopher J. Porter5, Julien Yockell-Lelièvre1,2, Hannah L. Battaion1,2,4, Safwat T. Khan1, Joel P. Howard1, Yuefeng Li1,2,4, Adrian T. Grzybowski6, Elham Sabri9, Alexander J. Ruthenburg6, F. Jeffrey Dilworth1,2,4, Theodore J. Perkins1,3,5, Mitchell Sabloff7,8, Caryn Y. Ito1,4* & William L. Stanford1,2,3,4* 1The Sprott Center for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada K1H 8L6; 2Ottawa Institute of Systems Biology, Ottawa, Ontario, Canada; 3Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada; 4Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada; 5Ottawa Bioinformatics Core Facility, The Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, Ottawa, ON, Canada K1H 8L6 6Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, Illinois, USA 60637 7Division of Hematology, Department of Medicine, University of Ottawa, Ottawa, Canada 8Ottawa Hospital Research Institute, Ottawa, ON, Canada Canada K1H 8L6 9Clinical Epidemiology Methods Centre, Ottawa Hospital Research Institute, Ottawa, ON, Canada K1H 8L6 †These authors contributed equally to this work. *Correspondence to: Caryn Ito, [email protected]; William L. Stanford, [email protected] Ottawa Hospital, 501 Smyth Rd, Box 511 Ottawa, ON K1H 8L6 CANADA 613-737-8899 ext.
    [Show full text]
  • The Genomic and Epigenomic Landscapes of AML
    The Genomic and Epigenomic Landscapes of AML Luca Mazzarella,a Laura Riva,b Lucilla Luzi,a,c Chiara Ronchini,b and Pier Giuseppe Peliccia,d A progressively better understanding of the genetic and epigenetic abnormalities underlying acute myeloid leukemia has changed clinical practice and affected the outcome of thousands of patients. Over the past decades, approaches focused on cloning, sequencing, and functional characterization of one or a few genes were the preferred (and the only possible) modality of investigation. The advent of disruptive new sequencing technologies brought about an unprecedented acceleration in our learning curve. Our view of the abnormalities required to generate and sustain leukemia is evolving from a piecemeal account based on individual lines of research into a comprehensive view of how all the important components (eg, transcriptional program, chromatin modifications, DNA sequence, alterations in noncoding genome) interact, in each patient and each leukemic cell. In this article, we provide an overall look at this complicated landscape and highlight outstanding issues for future research. Semin Hematol 51:259–272. C 2014 Elsevier Inc. All rights reserved. he idea that underlying genetic abnormalities A complex picture has emerged, with some AML- might dictate clinical decisions, now a common distinctive features. concept in oncology, was pioneered for acute The average number of mutations in coding sequences T 4 1–3 myeloid leukemia (AML) 25 years ago. Treatment of is very low in AML, compared with the majority of solid AML was indeed revolutionized by the recognition of tumors (13 mutations per patient in AML vs 52 in microscopically visible chromosomal abnormalities, as breast cancers, 290 in bladder cancers, and 500 in exemplified by the case of acute promyelocytic leukemia smoking-associated lung adenocarcinomas).
    [Show full text]
  • Jimmunol.1302943.Full.Pdf
    Published April 23, 2014, doi:10.4049/jimmunol.1302943 The Journal of Immunology Ezh2 Regulates Transcriptional and Posttranslational Expression of T-bet and Promotes Th1 Cell Responses Mediating Aplastic Anemia in Mice Qing Tong,*,† Shan He,†,‡ Fang Xie,† Kazuhiro Mochizuki,† Yongnian Liu,†,‡ Izumi Mochizuki,† Lijun Meng,‡,x Hongxing Sun,x Yanyun Zhang,x Yajun Guo,* Elizabeth Hexner,{ and Yi Zhang†,‡ Acquired aplastic anemia (AA) is a potentially fatal bone marrow (BM) failure syndrome. IFN-g–producing Th1 CD4+ T cells mediate the immune destruction of hematopoietic cells, and they are central to the pathogenesis. However, the molecular events that control the development of BM-destructive Th1 cells remain largely unknown. Ezh2 is a chromatin-modifying enzyme that regulates multiple cellular processes primarily by silencing gene expression. We recently reported that Ezh2 is crucial for inflammatory T cell responses after allogeneic BM transplantation. To elucidate whether Ezh2 mediates pathogenic Th1 responses in AA and the mechanism of Ezh2 action in regulating Th1 cells, we studied the effects of Ezh2 inhibition in CD4+ T cells using a mouse model of human AA. Conditionally deleting Ezh2 in mature T cells dramatically reduced the production of BM- destructive Th1 cells in vivo, decreased BM-infiltrating Th1 cells, and rescued mice from BM failure. Ezh2 inhibition resulted in significant decrease in the expression of Tbx21 and Stat4, which encode transcription factors T-bet and STAT4, respectively. Introduction of T-bet but not STAT4 into Ezh2-deficient T cells fully rescued their differentiation into Th1 cells mediating AA. Ezh2 bound to the Tbx21 promoter in Th1 cells and directly activated Tbx21 transcription.
    [Show full text]
  • Team Publications Maintenance of Transcriptional Repression by Polycomb Proteins
    Team Publications Maintenance of Transcriptional Repression by Polycomb Proteins Year of publication 2019 Michel Wassef, Eric Pasmant, Raphaël Margueron (2019 Oct 3) “MPNST Epigenetics”-Letter. Molecular cancer research : MCR : 2139 : DOI : 10.1158/1541-7786.MCR-19-0680 Summary Roberta Ragazzini, Raquel Pérez-Palacios, Irem H Baymaz, Seynabou Diop, Katia Ancelin, Dina Zielinski, Audrey Michaud, Maëlle Givelet, Mate Borsos, Setareh Aflaki, Patricia Legoix, Pascal W T C Jansen, Nicolas Servant, Maria-Elena Torres-Padilla, Deborah Bourc'his, Pierre Fouchet, Michiel Vermeulen, Raphaël Margueron (2019 Aug 26) EZHIP constrains Polycomb Repressive Complex 2 activity in germ cells. Nature communications : 10 : 1-18 : DOI : 10.1038/s41467-019-11800-x Summary The Polycomb group of proteins is required for the proper orchestration of gene expression due to its role in maintaining transcriptional silencing. It is composed of several chromatin modifying complexes, including Polycomb Repressive Complex 2 (PRC2), which deposits H3K27me2/3. Here, we report the identification of a cofactor of PRC2, EZHIP (EZH1/2 Inhibitory Protein), expressed predominantly in the gonads. EZHIP limits the enzymatic activity of PRC2 and lessens the interaction between the core complex and its accessory subunits, but does not interfere with PRC2 recruitment to chromatin. Deletion of Ezhip in mice leads to a global increase in H3K27me2/3 deposition both during spermatogenesis and at late stages of oocyte maturation. This does not affect the initial number of follicles but is associated with a reduction of follicles in aging. Our results suggest that mature oocytes Ezhip-/- might not be fully functional and indicate that fertility is strongly impaired in Ezhip-/- females.
    [Show full text]
  • Mammalian PRC1 Complexes: Compositional Complexity and Diverse Molecular Mechanisms
    International Journal of Molecular Sciences Review Mammalian PRC1 Complexes: Compositional Complexity and Diverse Molecular Mechanisms Zhuangzhuang Geng 1 and Zhonghua Gao 1,2,3,* 1 Departments of Biochemistry and Molecular Biology, Penn State College of Medicine, Hershey, PA 17033, USA; [email protected] 2 Penn State Hershey Cancer Institute, Hershey, PA 17033, USA 3 The Stem Cell and Regenerative Biology Program, Penn State College of Medicine, Hershey, PA 17033, USA * Correspondence: [email protected] Received: 6 October 2020; Accepted: 5 November 2020; Published: 14 November 2020 Abstract: Polycomb group (PcG) proteins function as vital epigenetic regulators in various biological processes, including pluripotency, development, and carcinogenesis. PcG proteins form multicomponent complexes, and two major types of protein complexes have been identified in mammals to date, Polycomb Repressive Complexes 1 and 2 (PRC1 and PRC2). The PRC1 complexes are composed in a hierarchical manner in which the catalytic core, RING1A/B, exclusively interacts with one of six Polycomb group RING finger (PCGF) proteins. This association with specific PCGF proteins allows for PRC1 to be subdivided into six distinct groups, each with their own unique modes of action arising from the distinct set of associated proteins. Historically, PRC1 was considered to be a transcription repressor that deposited monoubiquitylation of histone H2A at lysine 119 (H2AK119ub1) and compacted local chromatin. More recently, there is increasing evidence that demonstrates the transcription activation role of PRC1. Moreover, studies on the higher-order chromatin structure have revealed a new function for PRC1 in mediating long-range interactions. This provides a different perspective regarding both the transcription activation and repression characteristics of PRC1.
    [Show full text]