(HDAC1), but Not HDAC2, Controls Embryonic Stem Cell Differentiation

Total Page:16

File Type:pdf, Size:1020Kb

(HDAC1), but Not HDAC2, Controls Embryonic Stem Cell Differentiation Histone deacetylase 1 (HDAC1), but not HDAC2, controls embryonic stem cell differentiation Oliver M. Dovey, Charles T. Foster, and Shaun M. Cowley1 Department of Biochemistry, University of Leicester, Leicester LE1 9HN, United Kingdom Edited* by Robert N. Eisenman, Fred Hutchinson Cancer Research Center, Seattle, WA, and approved March 30, 2010 (received for review January 13, 2010) Histone deacetylases (HDAC) 1 and 2 are highly similar enzymes that complexes, including Sin3A (20), SDS3 (21), MBD3 (22), and help regulate chromatin structure as the core catalytic components LSD1 (23, 24). Germ-line deletion of HDAC1 results in early of corepressor complexes. Although tissue-specific deletion of embryonic lethality around embryonic day (e)10.5, although ab- HDAC1 and HDAC2 has demonstrated functional redundancy, errant development occurs as early as e7.5. In contrast to these germ-line deletion of HDAC1 in the mouse causes early embryonic early embryonic phenotypes, constitutive HDAC2 knockout mice lethality, whereas HDAC2 does not. To address the unique re- survive embryogenesis and either die shortly after birth in one quirement for HDAC1 in early embryogenesis we have generated model (19) or survive to adulthood in others (25–27), albeit at conditional knockout embryonic stem (ES) cells in which HDAC1 or reduced Mendelian frequencies. In a number of cell types, de- HDAC2 genes can be inactivated. Deletion of HDAC1, but not letion of both HDAC1 and HDAC2 is required to generate HDAC2, causes a significant reduction in the HDAC activity of Sin3A, a phenotype (14, 19, 28). This result suggests that the activity of NuRD, and CoREST corepressor complexes. This reduced corepressor HDAC1 and HDAC2 is mostly redundant, with the requirement activity results in a specific 1.6-fold increase in histone H3 K56 for both HDAC1 and HDAC2 occurring only at certain key de- acetylation (H3K56Ac), thus providing genetic evidence that velopmental periods, such as gastrulation. To investigate the es- H3K56Ac is a substrate of HDAC1. In culture, ES cell proliferation sential role of HDAC1 during early embryogenesis we have was unaffected by loss of either HDAC1 or HDAC2. Rather, we find compared the biochemical, proliferative, and differentiation that loss of HDAC1 affects ES cell differentiation. ES cells lacking properties of HDAC1 and HDAC2 conditional knockout embry- either HDAC1 or HDAC2 were capable of forming embryoid bodies onic stem (ES) cells. ES cells are the in vitro counterpart of epi- (EBs), which stimulates differentiation into the three primary germ blast cells of the early postimplantation embryo and their fi fi layers. However, HDAC1-de cient EBs were signi cantly smaller, differentiation mimics many of the changes in gene expression showed spontaneous rhythmic contraction, and increased expres- associated with embryonic development (29). We find that loss of sion of both cardiomyocyte and neuronal markers. In summary, our HDAC1, but not HDAC2, reduces the level of HDAC activity genetic study of HDAC1 and HDAC2 in ES cells, which mimic the associated with HDAC1/2 complexes and leads to the enhanced fi embryonic epiblast, has identi ed a unique requirement for HDAC1 differentiation of embryoid bodies. in the optimal activity of HDAC1/2 corepressor complexes and cell fate determination during differentiation. Results Generation of Conditional Knockout ES Cell Lines for HDAC1 and corepressor | acetylation | deacetylation | chromatin HDAC2. Beginning with an E14 ES cell line expressing an inducible Cre/Estrogen Receptor (CreER) construct from the endogenous lass I histone deacetylases (HDAC1, -2, -3, and -8) are highly ROSA26 locus (30), we used homologous recombination to Cconserved enzymes present in the nucleus of all cells, where produce HDAC1Lox/Lox; CreER and HDAC2Lox/Lox; CreER cell they help modulate levels of gene expression (1). The best lines in which exon 2 of each gene is flanked by LoxP sites (see characterized substrates of HDACs are the N-terminal tails of Fig. 1A and Figs. S1 and S2 for detailed methods). Induction of the core histones H2A, H2B, H3 and H4. Deacetylation of his- Cre activity by addition of 4-hydroxy tamoxifen (OHT) to the tone tails results in a “tightening” of chromatin, due to the growth media resulted in complete recombination of each allele Δ Δ Δ Δ electrostatic potential of unacetylated lysine residues to promote and deletion of exon 2 (HDAC1 2/ 2 or HDAC2 2/ 2) within 24 h internucleosomal interactions (2, 3) and the loss of a binding site (Fig.1B). Loss of exon 2 disrupts the ORF of both HDAC1 and for components of the transcriptional machinery containing a HDAC2 such that a premature stop codon is introduced in bromodomain, e.g., TAFII1 (4). exon 3. Following deletion of exon 2 a further 4–5 days of culture Among the class I HDACs, HDAC1 and HDAC2 are the most are required before HDAC1 and HDAC2 protein levels are re- similar (83% amino acid identity), sharing an almost identical duced below 10% of those of control cells (Fig. 1C), suggesting catalytic core domain and a conserved C-terminal tail (5). In that a combination of a long-lived mRNA and recruitment into mammalian cells HDAC1 and HDAC2 interact together (6) to a stable protein complex results in relatively slow protein turn- form the catalytic core of a number of higher-order complexes over. In the subsequent experiments, HDAC1Lox/Lox; CreER and including Sin3A, NuRD, CoREST, and NODE (reviewed in ref. HDAC2Lox/Lox; CreER cells 10–24 days postrecombination (hence Δ Δ Δ Δ 1). These complexes are then targeted to chromatin by sequence- referred to as HDAC1 2/ 2 and HDAC2 2/ 2) were used to assess specific (often cell-specific) transcription factors to repress their biochemical and growth properties. transcription in cooperation with other chromatin modifiers, such as the lysine-specific demethylase, LSD1, in the CoREST – complex (7 9). As part of these multiprotein complexes, the Author contributions: O.M.D. and S.M.C. designed research; O.M.D., C.T.F., and S.M.C. activity of HDAC1 and HDAC2 has been implicated in the performed research; O.M.D. contributed new reagents/analytic tools; O.M.D., C.T.F., regulation of cell cycle progression by tumor suppressors (10– and S.M.C. analyzed data; and O.M.D. and S.M.C. wrote the paper. 12), differentiation (13, 14), cellular aging (15), and cancer (16). The authors declare no conflict of interest. Indeed, a variety of HDAC inhibitors that target both HDAC1 *This Direct Submission article had a prearranged editor. and -2 are currently being tested in the clinic as potential anti- Freely available online through the PNAS open access option. cancer therapeutics (17). 1To whom correspondence should be addressed. E-mail: [email protected]. Mouse genetics has demonstrated an essential role in embryo- This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. genesis for HDAC1 (18, 19) and many components of HDAC1/2 1073/pnas.1000478107/-/DCSupplemental. 8242–8247 | PNAS | May 4, 2010 | vol. 107 | no. 18 www.pnas.org/cgi/doi/10.1073/pnas.1000478107 Downloaded by guest on September 30, 2021 Fig. 1. Generation of HDAC1 and HDAC2 conditional knockout ES cell lines. (A) An E14 ES cell line constitutively expressing a Cre/Estrogen Receptor (CreER) fusion from the ROSA26 locus was used to generate homozygous conditional knockout alleles for HDAC1 and HDAC2. Both copies of exon 2 were flanked by LoxP sites, using consecutive rounds of gene targeting. (B) Southern blots showing wild-type and exon 2 targeted HDAC1Lox/Lox and HDAC2Lox/Lox loci. Addition of 4-hydroxy tamoxifen (OHT) activates the Δ Δ Δ Δ CreER fusion and induces deletion of exon2 (HDAC1 2/ 2, HDAC2 2/ 2)inES cells; >95% recombination is observed after 24 h. (C) Quantitative Western blot shows ligand-inducible deletion of HDAC1 and -2 protein in whole-cell extracts from HDAC1Lox/Lox and HDAC2Lox/Lox ES cells, respectively. Cells were cultured for up to 10 days (0–3 days in the presence of OHT). α-Tubulin was used to normalize protein loading. Blots were visualized and quantitated using a LiCOR scanner. Data are representative of three independently tested clones for each genotype. CELL BIOLOGY Reduced HDAC Activity Associated with HDAC1/2 Complexes in the Absence of HDAC1. Consistent with previous reports (18, 31), deletion of HDAC1 results in an increased level of HDAC2 protein (Fig. 2A). However, no increase in HDAC2 mRNA is detected, suggesting that increased protein levels may occur from changes in HDAC2 translation and/or degradation in the ab- sence of HDAC1. Interestingly, there is no change in HDAC1 protein levels in the absence of HDAC2 (Fig. 2A). Most, if not all cellular HDAC1 and -2 is associated with higher-order pro- tein complexes in the nucleus (1). We used coimmunoprecipi- tation of individual components of the Sin3A, NuRD, and Fig. 2. Loss of HDAC1 results in decreased deacetylase activity associated CoREST complexes to assess the associated HDAC activity and with HDAC1/2 complexes and an increase H3K56Ac. (A) Quantitative West- their integrity in cells lacking either HDAC1 or HDAC2. In the ern blot of HDAC1, -2, and -3 obtained from nuclear extracts of the indicated cell types. Protein levels were quantitated using a LiCOR scanner and nor- absence of HDAC1 we observe a decrease in the deacetylase malized to the level of α-tubulin. Data represent independent experiments, activity associated with each of the Sin3A, NuRD, and CoREST using three independent clones. (*, P < 0.05; Student’s t test). Mean values complexes, with the largest reduction in the CoREST complex (n =3)± SEM are plotted. (B) Specific antisera to the indicated proteins were (Fig. 2B). This decrease in activity occurs despite the fact that we used to coimmunoprecipitate Sin3A, NuRD (αMTA-2), and CoREST (α-LSD1) detect increased amounts of HDAC2 in each of these same complexes from HDAC1Lox/Lox, HDAC1Δ2/Δ2, HDAC2Lox/Lox, and HDAC2Δ2/Δ2 ES complexes (Fig.
Recommended publications
  • Screening and Identification of Key Biomarkers in Clear Cell Renal Cell Carcinoma Based on Bioinformatics Analysis
    bioRxiv preprint doi: https://doi.org/10.1101/2020.12.21.423889; this version posted December 23, 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. Screening and identification of key biomarkers in clear cell renal cell carcinoma based on bioinformatics analysis Basavaraj Vastrad1, Chanabasayya Vastrad*2 , Iranna Kotturshetti 1. Department of Biochemistry, Basaveshwar College of Pharmacy, Gadag, Karnataka 582103, India. 2. Biostatistics and Bioinformatics, Chanabasava Nilaya, Bharthinagar, Dharwad 580001, Karanataka, India. 3. Department of Ayurveda, Rajiv Gandhi Education Society`s Ayurvedic Medical College, Ron, Karnataka 562209, India. * Chanabasayya Vastrad [email protected] Ph: +919480073398 Chanabasava Nilaya, Bharthinagar, Dharwad 580001 , Karanataka, India bioRxiv preprint doi: https://doi.org/10.1101/2020.12.21.423889; this version posted December 23, 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 Clear cell renal cell carcinoma (ccRCC) is one of the most common types of malignancy of the urinary system. The pathogenesis and effective diagnosis of ccRCC have become popular topics for research in the previous decade. In the current study, an integrated bioinformatics analysis was performed to identify core genes associated in ccRCC. An expression dataset (GSE105261) was downloaded from the Gene Expression Omnibus database, and included 26 ccRCC and 9 normal kideny samples. Assessment of the microarray dataset led to the recognition of differentially expressed genes (DEGs), which was subsequently used for pathway and gene ontology (GO) enrichment analysis.
    [Show full text]
  • Mapping Influenza-Induced Posttranslational Modifications On
    viruses Article Mapping Influenza-Induced Posttranslational Modifications on Histones from CD8+ T Cells Svetlana Rezinciuc 1, Zhixin Tian 2, Si Wu 2, Shawna Hengel 2, Ljiljana Pasa-Tolic 2 and Heather S. Smallwood 1,3,* 1 Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN 38163, USA; [email protected] 2 Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, WA 99354, USA; [email protected] (Z.T.); [email protected] (S.W.); [email protected] (S.H.); [email protected] (L.P.-T.) 3 Children’s Foundation Research Institute, Memphis, TN 38105, USA * Correspondence: [email protected]; Tel.: +1-(901)-448–3068 Academic Editor: Italo Tempera Received: 10 October 2020; Accepted: 2 December 2020; Published: 8 December 2020 Abstract: T cell function is determined by transcriptional networks that are regulated by epigenetic programming via posttranslational modifications (PTMs) to histone proteins and DNA. Bottom-up mass spectrometry (MS) can identify histone PTMs, whereas intact protein analysis by MS can detect species missed by bottom-up approaches. We used a novel approach of online two-dimensional liquid chromatography-tandem MS with high-resolution reversed-phase liquid chromatography (RPLC), alternating electron transfer dissociation (ETD) and collision-induced dissociation (CID) on precursor ions to maximize fragmentation of uniquely modified species. The first online RPLC separation sorted histone families, then RPLC or weak cation exchange hydrophilic interaction liquid chromatography (WCX-HILIC) separated species heavily clad in PTMs. Tentative identifications were assigned by matching proteoform masses to predicted theoretical masses that were verified with tandem MS. We used this innovative approach for histone-intact protein PTM mapping (HiPTMap) to identify and quantify proteoforms purified from CD8 T cells after in vivo influenza infection.
    [Show full text]
  • Exogenous Hydrogen Sulfide Plays an Important Role by Regulating
    International Journal of Molecular Sciences Review Exogenous Hydrogen Sulfide Plays an Important Role by Regulating Autophagy in Diabetic-Related Diseases Shuangyu Lv , Huiyang Liu and Honggang Wang * Henan International Joint Laboratory of Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475000, China; [email protected] (S.L.); [email protected] (H.L.) * Correspondence: [email protected] Abstract: Autophagy is a vital cell mechanism which plays an important role in many physiological processes including clearing long-lived, accumulated and misfolded proteins, removing damaged organelles and regulating growth and aging. Autophagy also participates in a variety of biological functions, such as development, cell differentiation, resistance to pathogens and nutritional hunger. Recently, autophagy has been reported to be involved in diabetes, but the mechanism is not fully understood. Hydrogen sulfide (H2S) is a colorless, water-soluble, flammable gas with the typical odor of rotten eggs, which has been known as a highly toxic gas for many years. However, it has been reported recently that H2S, together with nitric oxide and carbon monoxide, is an important gas signal transduction molecule. H2S has been reported to play a protective role in many diabetes- related diseases, but the mechanism is not fully clear. Recent studies indicate that H2S plays an important role by regulating autophagy in many diseases including cancer, tissue fibrosis diseases and glycometabolic diseases; however, the related mechanism has not been fully studied. In this review, we summarize recent research on the role of H2S in regulating autophagy in diabetic-related diseases to provide references for future related research.
    [Show full text]
  • Subcellular Distribution of HDAC1 in Neurotoxic Conditions Is Dependent on Serine Phosphorylation
    The Journal of Neuroscience, August 2, 2017 • 37(31):7547–7559 • 7547 Neurobiology of Disease Subcellular Distribution of HDAC1 in Neurotoxic Conditions Is Dependent on Serine Phosphorylation Yunjiao Zhu,1* Oscar G. Vidaurre,1* XKadidia P. Adula,1 XNebojsa Kezunovic,1 XMaureen Wentling,1 X George W. Huntley,1 and XPatrizia Casaccia1,2 1Department of Neuroscience, Friedman Brain Institute, Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029, and 2Advanced Science Research Center at the Graduate Center of the City University of New York, New York, New York 10031 Calcium-dependent nuclear export of histone deacetylase 1 (HDAC1) was shown previously to precede axonal damage in culture, but the in vivo relevance of these findings and the potential posttranslational modifications of HDAC1 remained elusive. Using acute hippocam- pal slices from mice of either sex with genetic conditional ablation of Hdac1 in CA1 hippocampal neurons (i.e., Camk2a-cre;Hdac1 fl/fl), we show significantly diminished axonal damage in response to neurotoxic stimuli. The protective effect of Hdac1 ablation was detected also in CA3 neurons in Grik4-cre;Hdac1 fl/f mice, which were more resistant to the excitotoxic damage induced by intraventricular injection of kainic acid. The amino acid residues modulating HDAC1 subcellular localization were identified by site-directed mutagenesis, which identified serine residues 421 and 423 as critical for its nuclear localization. The physiological phosphorylation of HDAC1 was decreased by neurotoxic stimuli, which stimulated the phosphatase enzymatic activity of calcineurin. Treatment of neurons with the calcineurin inhibitors FK506 or cyclosporin A resulted in nuclear accumulation of phospho-HDAC1 and was neuroprotective.
    [Show full text]
  • Determining HDAC8 Substrate Specificity by Noah Ariel Wolfson A
    Determining HDAC8 substrate specificity by Noah Ariel Wolfson A dissertation submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy (Biological Chemistry) in the University of Michigan 2014 Doctoral Committee: Professor Carol A. Fierke, Chair Professor Robert S. Fuller Professor Anna K. Mapp Associate Professor Patrick J. O’Brien Associate Professor Raymond C. Trievel Dedication My thesis is dedicated to all my family, mentors, and friends who made getting to this point possible. ii Table of Contents Dedication ....................................................................................................................................... ii List of Figures .............................................................................................................................. viii List of Tables .................................................................................................................................. x List of Appendices ......................................................................................................................... xi Abstract ......................................................................................................................................... xii Chapter 1 HDAC8 substrates: Histones and beyond ...................................................................... 1 Overview ..................................................................................................................................... 1 HDAC introduction
    [Show full text]
  • Up-Regulation of UVRAG by HDAC1 Inhibition Attenuates 5FU-Induced
    ANTICANCER RESEARCH 38 : 271-277 (2018) doi:10.21873/anticanres.12218 Up-regulation of UVRAG by HDAC1 Inhibition Attenuates 5FU-induced Cell Death in HCT116 Colorectal Cancer Cells YOON KYUNG JO 1* , NA YEON PARK 1* , JI HYUN SHIN 1, DOO SIN JO 1, JI-EUN BAE 1, EUN SUN CHOI 1, SUNGHO MAENG 1, HONG BAE JEON 2, SEON AE ROH 3, JONG WOOK CHANG 4, JIN CHEON KIM 3,5 and DONG-HYUNG CHO 1 1Department of Gerontology, Graduate School of East-West Medical Science, Kyung Hee University, Seoul, Republic of Korea; 2Biomedical Research Institute, MEDIPOST Corporation, Seongnam, Republic of Korea; 3Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea; 4Stem Cell & Regenerative Medicine Institute, Samsung Medical Center, Seoul, Republic of Korea; 5Department of Surgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea Abstract. The ultraviolent irradiation resistance-associated Autophagy has important roles in cellular homeostasis gene (UVRAG), a component of the Beclin 1/autophagy- through the degradation of useless or damaged proteins and related 6 complex, regulates the autophagy initiation step organelles via lysosomes (1, 2). The primordial function of and functions in the DNA-damage response. UVRAG is autophagy is a response to stress, such as starvation, frequently mutated in various cancer types, and mutations of oxidative stress, and ion stress (1, 2). Given that multiple UVRAG increase sensitivity to chemotherapy by impairing signaling pathways are involved in the regulation of DNA-damage repair. In this study, we addressed the autophagy progress, various autophagy-related (ATG) epigenetic regulation of UVRAG in colorectal cancer cells.
    [Show full text]
  • Neutralizing Gatad2a-Chd4-Mbd3 Axis Within the Nurd Complex
    bioRxiv preprint doi: https://doi.org/10.1101/192781; this version posted February 9, 2018. 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. 1 Neutralizing Gatad2a-Chd4-Mbd3 Axis 2 within the NuRD Complex Facilitates 3 Deterministic Induction of Naïve Pluripotency 4 1* 1* 1 1,2 1 5 Nofar Mor , Yoach Rais , Shani Peles , Daoud Sheban , Alejandro Aguilera-Castrejon , 1 3 1 1 1 6 Asaf Zviran , Dalia Elinger , Sergey Viukov , Shay Geula , Vladislav Krupalnik , Mirie 1 4,5 1 1 1 1 7 Zerbib , Elad Chomsky , Lior Lasman , Tom Shani , Jonathan Bayerl , Ohad Gafni , 6 7,8 9 10 10 8 Suhair Hanna , Jason D. Buenrostro , Tzachi Hagai , Hagit Masika , Yehudit Bergman , 11,12 13 3 1 2 9 William J. Greenleaf , Miguel A. Esteban , Yishai Levin , Rada Massarwa , Yifat Merbl , 1#@ 1#@% 10 Noa Novershtern and Jacob H. Hanna . 1 11 The Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel. 2 12 The Department of Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel. 13 3 The Nancy and Stephen Grand Israel National Center for Personalized Medicine (INCPM), 14 Weizmann Institute of Science, Rehovot 7610001, Israel. 15 4 Department of Biological Regulation, Weizmann Institute of Science, Rehovot 7610001, Israel 16 5 Department of Computer Science, Weizmann Institute of Science, Rehovot 7610001, Israel 17 6 Department of Pediatrics and the Pediatric Immunology Unit, Rambam Health Care Campus & 18 Bruce
    [Show full text]
  • Rabbit Mab A
    Revision 1 C 0 2 - t SIN3A (D1B7) Rabbit mAb a e r o t S Orders: 877-616-CELL (2355) [email protected] Support: 877-678-TECH (8324) 1 9 Web: [email protected] 6 www.cellsignal.com 7 # 3 Trask Lane Danvers Massachusetts 01923 USA For Research Use Only. Not For Use In Diagnostic Procedures. Applications: Reactivity: Sensitivity: MW (kDa): Source/Isotype: UniProt ID: Entrez-Gene Id: WB, ChIP H M R Mk Endogenous 145 Rabbit IgG Q96ST3 25942 Product Usage Information nucleosome binding of the complex. The SIN3 complex functions to repress transcription, in part, by deacetylating histones at target gene promoters (3,4). In addition, recent For optimal ChIP results, use 5 μl of antibody and 10 μg of chromatin (approximately 4 x studies have shown that SIN3 is recruited to the coding regions of repressed and active 106 cells) per IP. This antibody has been validated using SimpleChIP® Enzymatic genes, where it deacetylates histones and suppresses spurious transcription by RNA Chromatin IP Kits. polymerase II (3,5). In addition to histone deacetylase activity, the SIN3 complex associates with histone methyltransferase (ESET), histone demethylase Application Dilution (JARID1A/RBP2), ATP-dependent chromatin remodeling (SWI/SNF), methylcytosine dioxygenase (TET1), and O-GlcNAc transferase (OGT) activities, all of which appear to Western Blotting 1:1000 contribute to the regulation of target genes (5-9). The SIN3 complex is critical for proper Chromatin IP 1:100 regulation of embryonic development, cell growth and proliferation, apoptosis, DNA replication, DNA repair, and DNA methylation (imprinting and X-chromosome inactivation) Storage (3,4).
    [Show full text]
  • PWWP2A Binds Distinct Chromatin Moieties and Interacts with an MTA1-Specific Core Nurd Complex
    ARTICLE DOI: 10.1038/s41467-018-06665-5 OPEN PWWP2A binds distinct chromatin moieties and interacts with an MTA1-specific core NuRD complex Stephanie Link1,2, Ramona M.M. Spitzer1,2, Maryam Sana3, Mario Torrado3, Moritz C. Völker-Albert1, Eva C. Keilhauer4,9, Thomas Burgold5,10, Sebastian Pünzeler1,11, Jason K.K. Low3, Ida Lindström 3, Andrea Nist6, Catherine Regnard1, Thorsten Stiewe 6,7, Brian Hendrich5, Axel Imhof 1,8, Matthias Mann 4,8, Joel P. Mackay 3, Marek Bartkuhn2 & Sandra B. Hake 2,8 1234567890():,; Chromatin structure and function is regulated by reader proteins recognizing histone mod- ifications and/or histone variants. We recently identified that PWWP2A tightly binds to H2A. Z-containing nucleosomes and is involved in mitotic progression and cranial–facial devel- opment. Here, using in vitro assays, we show that distinct domains of PWWP2A mediate binding to free linker DNA as well as H3K36me3 nucleosomes. In vivo, PWWP2A strongly recognizes H2A.Z-containing regulatory regions and weakly binds H3K36me3-containing gene bodies. Further, PWWP2A binds to an MTA1-specific subcomplex of the NuRD complex (M1HR), which consists solely of MTA1, HDAC1, and RBBP4/7, and excludes CHD, GATAD2 and MBD proteins. Depletion of PWWP2A leads to an increase of acetylation levels on H3K27 as well as H2A.Z, presumably by impaired chromatin recruitment of M1HR. Thus, this study identifies PWWP2A as a complex chromatin-binding protein that serves to direct the deacetylase complex M1HR to H2A.Z-containing chromatin, thereby promoting changes in histone acetylation levels. 1 Department of Molecular Biology, BioMedical Center (BMC), Ludwig-Maximilians-University Munich, 82152 Planegg-Martinsried, Germany.
    [Show full text]
  • Aberrations of EZH2 in Cancer
    Published OnlineFirst March 2, 2011; DOI: 10.1158/1078-0432.CCR-10-2156 Clinical Cancer Molecular Pathways Research Aberrations of EZH2 in Cancer Andrew Chase and Nicholas C.P. Cross Abstract Control of gene expression is exerted at a number of different levels, one of which is the accessibility of genes and their controlling elements to the transcriptional machinery. Accessibility is dictated broadly by the degree of chromatin compaction, which is influenced in part by polycomb group proteins. EZH2, together with SUZ12 and EED, forms the polycomb repressive complex 2 (PRC2), which catalyzes trimethylation of histone H3 lysine 27 (H3K27me3). PRC2 may recruit other polycomb complexes, DNA methyltransferases, and histone deacetylases, resulting in additional transcriptional repressive marks and chromatin compaction at key developmental loci. Overexpression of EZH2 is a marker of advanced and metastatic disease in many solid tumors, including prostate and breast cancer. Mutation of EZH2 Y641 is described in lymphoma and results in enhanced activity, whereas inactivating mutations are seen in poor prognosis myeloid neoplasms. No histone demethylating agents are currently available for treatment of patients, but 3-deazaneplanocin (DZNep) reduces EZH2 levels and H3K27 trimethylation, resulting in reduced cell proliferation in breast and prostate cancer cells in vitro. Furthermore, synergistic effects are seen for combined treatment with DNA demethylating agents and histone deacetylation inhibitors, opening up the possibility of refined epigenetic treatments in the future. Clin Cancer Res; 17(9); 2613–8. Ó2011 AACR. Background trithorax homolog myeloid-lymphoid leukemia (MLL), is associated with transcriptional activation (Fig. 1A). Impor- DNA is wrapped around histone complexes termed tantly, many genes involved in development, stem cell nucleosomes, and gene accessibility is determined largely maintenance, and differentiation are targets of H3K27 by the local chromatin configuration.
    [Show full text]
  • 1 Unique Protein Interaction Networks Define the Chromatin Remodeling
    bioRxiv preprint doi: https://doi.org/10.1101/2021.01.27.428018; this version posted January 28, 2021. 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. Unique Protein Interaction Networks Define The Chromatin Remodeling Module of The NuRD Complex Mehdi Sharifi Tabar1,2, Caroline Giardina1, Yue Julie Feng1, Habib Francis1, Hakimeh Moghaddas Sani3, Jason K. K. Low3, Joel P. Mackay3, Charles G. Bailey1,2,4, John E.J. Rasko1,2,5,6 1Gene and Stem Cell Therapy Program Centenary Institute, The University of Sydney, Camperdown, NSW, 2050, Australia 2Faculty of Medicine & Health, The University of Sydney, Sydney, NSW, 2006, Australia 3School of Life & Environmental Sciences, The University of Sydney, Sydney, NSW, 2006, Australia 4Cancer and Gene Regulation Laboratory Centenary Institute, The University of Sydney, Camperdown, NSW, 2050, Australia 5Cell and Molecular Therapies, Royal Prince Alfred Hospital, Camperdown, NSW, 2050, Australia 6Corresponding author 1 bioRxiv preprint doi: https://doi.org/10.1101/2021.01.27.428018; this version posted January 28, 2021. 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 combination of four proteins and their paralogues including MBD2/3, GATAD2A/B, CDK2AP1, and CHD3/4/5, which we refer to as the MGCC module, form the chromatin remodeling module of the Nucleosome Remodeling and Deacetylase (NuRD) complex, a gene repressor complex. Specific paralogues of the MGCC subunits such as MBD2 and CHD4 are amongst the key repressors of adult-stage fetal globin and provide important targets for molecular therapies in beta (β)-thalassemia.
    [Show full text]
  • DNA Binding Selectivity of Mecp2 Due to a Requirement for A/T Sequences Adjacent to Methyl-Cpg
    Molecular Cell, Vol. 19, 667–678, September 2, 2005, Copyright ©2005 by Elsevier Inc. DOI 10.1016/j.molcel.2005.07.021 DNA Binding Selectivity of MeCP2 Due to a Requirement for A/T Sequences Adjacent to Methyl-CpG Robert J. Klose, Shireen A. Sarraf, 2004). Mammalian MBD3 does not bind specifically to Lars Schmiedeberg, Suzanne M. McDermott, methylated DNA, and MBD4 is a DNA repair protein Irina Stancheva,* and Adrian P. Bird* (Hendrich et al., 1999; Millar et al., 2002), although re- Wellcome Trust Centre for Cell Biology cent evidence suggests that MBD4 may also act as a Michael Swann Building transcriptional repressor (Kondo et al., 2005). University of Edinburgh The biomedical relevance of MBD proteins became Mayfield Road apparent with the discovery that the human neurode- Edinburgh EH9 3JR velopmental disorder Rett syndrome is caused by mu- United Kingdom tations in the MECP2 gene (Amir et al., 1999). Muta- tional profiling of Rett syndrome patients identified a significant fraction of point mutations that inactivated Summary the MBD itself (Kriaucionis and Bird, 2003). The DNA and protein features that determine the specificity of DNA methylation is interpreted by a family of methyl- the MBD for methyl-CpG sites have been examined CpG binding domain (MBD) proteins that repress (Free et al., 2001; Meehan et al., 1992; Nan et al., 1993; transcription through recruitment of corepressors Yusufzai and Wolffe, 2000), and the three-dimensional that modify chromatin. To compare in vivo binding of structure of the domain, both alone (Ohki et al., 1999; MeCP2 and MBD2, we analyzed immunoprecipitated Wakefield et al., 1999) and in complex with methylated chromatin from primary human cells.
    [Show full text]