Setdb1 Contributes to Repression of Genes Encoding Developmental Regulators and Maintenance of ES Cell State

Total Page:16

File Type:pdf, Size:1020Kb

Setdb1 Contributes to Repression of Genes Encoding Developmental Regulators and Maintenance of ES Cell State Downloaded from genesdev.cshlp.org on September 24, 2021 - Published by Cold Spring Harbor Laboratory Press RESEARCH COMMUNICATION Cole et al. 2008; Dejosez et al. 2008; Jiang et al. 2008; Kim SetDB1 contributes to et al. 2008; Marson et al. 2008; Tam et al. 2008). These repression of genes encoding key transcriptional regulators occupy the promoters of actively transcribed genes encoding transcription factors, developmental regulators and signaling components, and chromatin-modifying en- maintenance of ES cell state zymes that promote ES cell self-renewal. They also oc- cupy genes encoding a large set of developmental regula- Steve Bilodeau,1,3 Michael H. Kagey,1,3 tors that are silent in ES cells, but whose expression is Garrett M. Frampton,1,2 Peter B. Rahl,1 and associated with lineage commitment and cellular dif- 1,2,4 ferentiation. Polycomb-repressive complexes (PRCs) co- Richard A. Young occupy the genes encoding these developmental regula- 1Whitehead Institute for Biomedical Research, Cambridge, tors to help maintain a silent transcriptional state in ES Massachusetts 02142, USA; 2Department of Biology, cells (Azuara et al. 2006; Boyer et al. 2006; Lee et al. 2006; Massachusetts Institute of Technology (MIT), Stock et al. 2007; van der Stoop et al. 2008). Cambridge, Massachusetts 02139, USA The regulation of chromatin structure, which includes nucleosome remodeling and post-translational modifica- Transcription factors that play key roles in regulating tion of histone proteins, is necessary for the establish- embryonic stem (ES) cell state have been identified, but ment and maintenance of heritable gene expression the chromatin regulators that help maintain ES cells are patterns during development (Workman 2006; Dunn and less well understood. A high-throughput shRNA screen Kingston 2007; Kouzarides 2007; Surani et al. 2007). Trithorax group (TrxG) and Polycomb group (PcG) protein was used to identify novel chromatin regulators that complexes are key regulators of chromatin structure that influence ES cell state. Loss of histone H3 Lys 9 (H3K9) are necessary for maintenance of the ES cell gene expres- methyltransferases, particularly SetDB1, had the most sion program and are required for segmental identity in profound effects on ES cells. Chromatin immunoprecip- the developing embryo (for review, see Schuettengruber itation (ChIP) coupled with massively parallel DNA se- et al. 2007). TrxG complexes catalyze histone H3 Lys 4 quencing (ChIP-Seq) and functional analysis revealed trimethylation (H3K4me3) in proximity to promoters of that SetDB1 and histone H3K9-methylated nucleosomes genes that experience transcription initiation by RNA occupy and repress genes encoding developmental regu- polymerase II. Since most genes experience some level lators. These SetDB1-occupied genes are a subset of the of transcription initiation, most promoters are occupied ‘‘bivalent’’ genes, which contain nucleosomes with by nucleosomes with histone H3K4me3 (Guenther H3K4me3 (H3K4 trimethylation) and H3K27me3 modi- et al. 2007). The PcG protein complex PRC2 catalyzes fications catalyzed by Trithorax and Polycomb group H3K27me3, which contributes to repression of genes en- proteins, respectively. These genes are subjected to re- coding key developmental regulators. Disruption of PcG pression by both Polycomb group proteins and SetDB1, function in ES cells leads to derepression of these develop- and loss of either regulator can destabilize ES cell state. mental genes, alteration of the ES cell transcriptional pro- gram, and a loss of the ES cell state (Boyer et al. 2006; Lee Supplemental material is available at http://www.genesdev.org. et al. 2006; Pasini et al. 2007; Stock et al. 2007; Jaenisch and Young 2008; van der Stoop et al. 2008). In ES cells, Received June 26, 2009; revised version accepted September genes encoding lineage-specific developmental regula- 10, 2009. tors contain nucleosomes with both H3K4me3 and H3K27me3 modifications, and have thus been called ‘‘bivalent’’ (Bernstein et al. 2006). These genes apparently Embryonic stem (ES) cells provide an important model experience some level of transcription initiation, and system to study the control of early development and thus TrxG-mediated histone H3K4 methylation, but hold significant potential for clinical therapies because are silenced by PcG complexes, which may occur through of their distinctive capacity to both self-renew and dif- repression of transcription elongation (Stock et al. 2007). ferentiate into multiple lineages (Thomson et al. 1998; Although many chromatin remodeling and modifying Reubinoff et al. 2000; Pera and Trounson 2004; Keller activities have been described, only a few are known 2005; Hochedlinger and Jaenisch 2006; Yang and Smith to contribute to the control of ES cell state. We used 2007). Studies of key ES cell transcription factors— a screening approach with a shRNA library to screen including Oct4, Sox2, Nanog, Ronin, Tcf3, and others— for additional chromatin regulators of ES cell state. have led to models for ES cell transcriptional regula- Loss of histone H3K9 methyltransferases as well as tory circuitry that can account for several important PcG components was found to cause loss of ES cell features of the gene expression program of these unique state in this screen. Further study of the H3K9 methyl- cells (Boyer et al. 2005; Loh et al. 2006; Chen et al. 2008; transferase SetDB1 revealed that it contributes to re- pression of a subset of genes encoding developmental [Keywords: SetDB1; ES cell; H3K9 methylation; polycomb; repression; regulators in ES cells. These results indicate that there is pluripotency] an additional layer of chromatin-mediated repression at 3 These authors contributed equally to this work. these developmental genes, and that H3K9me3-modified 4Corresponding author. E-MAIL [email protected]; FAX (617) 258-9872. nucleosomes are not exclusive to constitutive hetero- Article is online at http://www.genesdev.org/cgi/doi/10.1101/gad.1837309. chromatin. 2484 GENES & DEVELOPMENT 23:2484–2489 Ó 2009 by Cold Spring Harbor Laboratory Press ISSN 0890-9369/09; www.genesdev.org Downloaded from genesdev.cshlp.org on September 24, 2021 - Published by Cold Spring Harbor Laboratory Press SetDB1 represses developmental regulators Results and Discussion cells in each well was measured. Control shRNAs target- ing Oct4 and Stat3, positive regulators of pluripotency in In order to identify chromatin regulators that may be mES cells (Nichols et al. 1998; Niwa et al. 1998; Hay et al. involved in the establishment or maintenance of cell 2004), produced cells that lost Oct4 signal, as expected state, we screened murine ES (mES) cells with a shRNA (Fig. 1B). In contrast, a control shRNA designed against library containing four to six hairpins directed against Tcf3, a negative regulator of Oct4 and Nanog (Pereira each of 197 chromatin regulators (Fig. 1A). ES cells were et al. 2006; Cole et al. 2008; Tam et al. 2008), caused an seeded into 384-well plates, and each well was infected increase in Oct4 staining. To normalize for plate variabil- with an individual lentiviral construct delivering ity, the average Oct4 staining intensity for the cells was a shRNA targeting a chromatin regulator. After selecting used to calculate a Z-score for each well, taking into for stable integration of the shRNA construct, the cells account values for negative control infections on each were fixed, and the average Oct4 staining intensity of the plate (see the Supplemental Material). The majority of the shRNAs targeting chromatin regulators produced Z-scores that were distributed between those observed for shRNAs targeting Oct4 and Tcf3, as expected (Fig. 1C; Supplemental Tables S1, S2). The chromatin regulators whose knockdowns reduced Oct4 levels nearly as profoundly as knockdown of Oct4 itself were of particular interest because many have been implicated in gene silencing, and many of these are associated with histone H3K9 and H3K27 methylation (Fig. 1C; Supplemental Table S1). The chromatin regula- tors associated with H3K9 methylation include SetDB1/ ESET, Ube2i/Ubc9, Ehmt1, and Suv39h2. Those associ- ated with H3K27 methylation include the Polycomb components Cbx7, Cbx8/Pc3, and Ezh2, and their iden- tification in this screen is consistent with previous evidence implicating Polycomb in control of ES cell state (Schuettengruber et al. 2007). The loss of the histone deacetylase Hdac3 and the Sin3/HDAC complex compo- nents Sin3a and Sap18 had similar effects. The loss of certain chromatin regulators led to increased Oct4 levels, and these included three members of the SWI/SNF family (Smarcd1, Arid1a, and Smarcb1) (Fig. 1C). Thus, the re- sults of this screen implicate multiple chromatin regula- tors in control of ES cell state. One of the largest classes of chromatin regulators identified in the screen was associated with histone H3K9 methylation. From this class we selected the SetDB1 H3K9 methyltransferase (Schultz et al. 2002; Ayyanathan et al. 2003; Wang et al. 2003) for further detailed study because its loss produced the most pro- found effects on ES cell state (Fig. 1C; Supplemental Table S1; Supplemental Fig. S1) and SetDB1-null blastocysts have deficiencies in the inner cell mass (Dodge et al. Figure 1. Identification of chromatin regulators of ES cell state. (A) 2004). Independent experiments demonstrated that mul- Outline of the screening protocol. mES cells were seeded without a mouse embryonic fibroblast (MEF) feeder layer into 384-well tiple shRNAs
Recommended publications
  • Structure of the Tripartite Motif of KAP1/TRIM28 Identifies Molecular Interfaces Required for Transcriptional Silencing of Retrotransposons
    bioRxiv preprint doi: https://doi.org/10.1101/505677; this version posted December 25, 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. Structure of the tripartite motif of KAP1/TRIM28 identifies molecular interfaces required for transcriptional silencing of retrotransposons Guido A. Stoll1, Shun-ichiro Oda1, Zheng-Shan Chong1, Minmin Yu2, Stephen H. McLaughlin2 and Yorgo Modis1,* 1 Molecular Immunity Unit, Department of Medicine, University of Cambridge, MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, CB2 0QH, UK 2 MRC Laboratory oF Molecular Biology, Cambridge Biomedical Campus, Cambridge, CB2 0QH, UK * Correspondence: [email protected] (Y.M.) Abstract Transcription oF transposable elements is tightly regulated to prevent damage to the genome. The family of KRAB domain-containing zinc Finger proteins (KRAB-ZFPs) and KRAB-associated protein 1 (KAP1/TRIM28) play a key role in regulating retrotransposons. KRAB-ZFPs recognize speciFic retrotransposon sequences and recruit KAP1, which controls the assembly of an epigenetic silencing complex including histone H3K9 methyltransferase SETDB1. The chromatin remodeling activities of this complex repress transcription of the targeted transposable element and any adjacent genes. Here, we use biophysical and structural approaches to show that the tripartite motif (TRIM) of KAP1 Forms antiparallel dimers, which Further assemble into tetramers and higher-order oligomers in a concentration-dependent manner. Structure-based mutations in the B-box 1 domain prevented higher-order oligomerization without a signiFicant loss oF retrotransposon silencing activity in a cell-based assay, indicating that, in contrast to other TRIM family members, selF-assembly is not essential For the function of KAP1.
    [Show full text]
  • A Computational Approach for Defining a Signature of Β-Cell Golgi Stress in Diabetes Mellitus
    Page 1 of 781 Diabetes A Computational Approach for Defining a Signature of β-Cell Golgi Stress in Diabetes Mellitus Robert N. Bone1,6,7, Olufunmilola Oyebamiji2, Sayali Talware2, Sharmila Selvaraj2, Preethi Krishnan3,6, Farooq Syed1,6,7, Huanmei Wu2, Carmella Evans-Molina 1,3,4,5,6,7,8* Departments of 1Pediatrics, 3Medicine, 4Anatomy, Cell Biology & Physiology, 5Biochemistry & Molecular Biology, the 6Center for Diabetes & Metabolic Diseases, and the 7Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202; 2Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202; 8Roudebush VA Medical Center, Indianapolis, IN 46202. *Corresponding Author(s): Carmella Evans-Molina, MD, PhD ([email protected]) Indiana University School of Medicine, 635 Barnhill Drive, MS 2031A, Indianapolis, IN 46202, Telephone: (317) 274-4145, Fax (317) 274-4107 Running Title: Golgi Stress Response in Diabetes Word Count: 4358 Number of Figures: 6 Keywords: Golgi apparatus stress, Islets, β cell, Type 1 diabetes, Type 2 diabetes 1 Diabetes Publish Ahead of Print, published online August 20, 2020 Diabetes Page 2 of 781 ABSTRACT The Golgi apparatus (GA) is an important site of insulin processing and granule maturation, but whether GA organelle dysfunction and GA stress are present in the diabetic β-cell has not been tested. We utilized an informatics-based approach to develop a transcriptional signature of β-cell GA stress using existing RNA sequencing and microarray datasets generated using human islets from donors with diabetes and islets where type 1(T1D) and type 2 diabetes (T2D) had been modeled ex vivo. To narrow our results to GA-specific genes, we applied a filter set of 1,030 genes accepted as GA associated.
    [Show full text]
  • A KAP1 Phosphorylation Switch Controls Myod Function During Skeletal Muscle Differentiation
    Downloaded from genesdev.cshlp.org on October 2, 2021 - Published by Cold Spring Harbor Laboratory Press A KAP1 phosphorylation switch controls MyoD function during skeletal muscle differentiation Kulwant Singh,1,5 Marco Cassano,2,5 Evarist Planet,2 Soji Sebastian,1 Suk Min Jang,2 Gurjeev Sohi,1 Herve Faralli,1 Jinmi Choi,3 Hong-Duk Youn,3 F. Jeffrey Dilworth,1,4 and Didier Trono2 1Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, Ottawa, Ontario K1H 8L6, Canada; 2School of Life Sciences, Ecole Polytechnique Fed erale de Lausanne (EPFL), 1015 Lausanne, Switzerland; 3Department of Biomedical Sciences and Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul 110-799, Korea; 4Department of Cellular and Molecular Medicine, University of Ottawa, Ontario K1H 8L6, Canada The transcriptional activator MyoD serves as a master controller of myogenesis. Often in partnership with Mef2 (myocyte enhancer factor 2), MyoD binds to the promoters of hundreds of muscle genes in proliferating myoblasts yet activates these targets only upon receiving cues that launch differentiation. What regulates this off/on switch of MyoD function has been incompletely understood, although it is known to reflect the action of chromatin modifiers. Here, we identify KAP1 (KRAB [Kruppel-like€ associated box]-associated protein 1)/TRIM28 (tripartite motif protein 28) as a key regulator of MyoD function. In myoblasts, KAP1 is present with MyoD and Mef2 at many muscle genes, where it acts as a scaffold to recruit not only coactivators such as p300 and LSD1 but also corepressors such as G9a and HDAC1 (histone deacetylase 1), with promoter silencing as the net outcome.
    [Show full text]
  • Hippocampal Dysfunction in the Euchromatin Histone Methyltransferase 1 Heterozygous Knockout Mouse Model for Kleefstra Syndrome
    Human Molecular Genetics, 2013, Vol. 22, No. 5 852–866 doi:10.1093/hmg/dds490 Advance Access published on November 21, 2012 Hippocampal dysfunction in the Euchromatin histone methyltransferase 1 heterozygous knockout mouse model for Kleefstra syndrome Monique C.M. Balemans1,2, Nael Nadif Kasri3, Maksym V. Kopanitsa5, Nurudeen O. Afinowi5, Ger Ramakers6, Theo A. Peters4, Andy J. Beynon4, Sanne M. Janssen1, Rik C.J. van Summeren1,2, Jorine M. Eeftens1, Nathalie Eikelenboom1, Marco Benevento3, Makoto Tachibana7, Yoichi Shinkai7, Tjitske Kleefstra1, Hans van Bokhoven1,3,∗,{ and Catharina E.E.M. Van der Zee2,∗,{ 1Department of Human Genetics, 2Department of Cell Biology, Nijmegen Centre for Molecular Life Sciences, 3Department of Cognitive Neurosciences, and 4Department of Otorhinolaryngology, Donders Institute for Brain, Downloaded from Cognition and Behavior, Radboud University, Nijmegen Medical Centre, P.O. Box 9101, 6500 HB Nijmegen, the Netherlands 5Synome Ltd, Moneta Building, Babraham Research Campus, Cambridge CB22 3AT, UK, 6Department of Developmental Psychology, University of Amsterdam, Roetersstraat 15, 1018 WB Amsterdam, the Netherlands and 7Experimental Research Center for Infectious Diseases, Institute for Virus Research, Kyoto, University, 53 Shogoin, http://hmg.oxfordjournals.org/ Kawara-cho, Sakyo-ku, Kyoto, Japan Received October 21, 2012; Revised October 21, 2012; Accepted November 15, 2012 Euchromatin histone methyltransferase 1 (EHMT1) is a highly conserved protein that catalyzes mono- and dimethylation of histone H3 lysine 9, thereby epigenetically regulating transcription. Kleefstra syndrome (KS), is caused by haploinsufficiency of the EHMT1 gene, and is an example of an emerging group of intel- lectual disability (ID) disorders caused by genes encoding epigenetic regulators of neuronal gene activity. by guest on March 24, 2016 Little is known about the mechanisms underlying this disorder, prompting us to study the Euchromatin his- tone methyltransferase 1 heterozygous knockout (Ehmt11/2) mice as a model for KS.
    [Show full text]
  • The Histone Methyltransferase SETDB1 Represses Endogenous and Exogenous Retroviruses in B Lymphocytes
    The histone methyltransferase SETDB1 represses endogenous and exogenous retroviruses in B lymphocytes Patrick L. Collinsa, Katherine E. Kylea, Takeshi Egawaa, Yoichi Shinkaib, and Eugene M. Oltza,1 aDepartment of Pathology, Washington University School of Medicine, St. Louis, MO 63110; and bCellular Memory Laboratory, RIKEN, Saitama 351-0198, Japan Edited by John V. Moran, University of Michigan, Ann Arbor, MI, and accepted by the Editorial Board May 21, 2015 (received for review November 19, 2014) Genome stability relies on epigenetic mechanisms that enforce tumor viruses (MMTVs). Reintegration of these expressed viruses repression of endogenous retroviruses (ERVs). Current evidence can generate mutations with potential oncogenic consequences suggests that distinct chromatin-based mechanisms repress ERVs (7). Thus, suppression of ERVs via epigenetic mechanisms is es- in cells of embryonic origin (histone methylation dominant) vs. pecially important in adult tissues that harbor cells with a high more differentiated cells (DNA methylation dominant). However, proliferative capacity. the latter aspect of this model has not been tested. Remarkably, Recent studies suggest that the mechanisms of ERV re- and in contrast to the prevailing model, we find that repressive pression in differentiated adult tissues are distinct from those histone methylation catalyzed by the enzyme SETDB1 is critical for in embryonic stem cells (ESCs) or early lineage progenitors (3). In fully differentiated cells, such as fibroblasts, DNA methylation suppression of specific ERV families and exogenous retroviruses in appears to be particularly important for ERV suppression, committed B-lineage cells from adult mice. The profile of ERV whereas HMTs responsible for H3K9me3 are largely dispens- activation in SETDB1-deficient B cells is distinct from that observed able (3, 4).
    [Show full text]
  • Functional Dependency Analysis Identifies Potential Druggable
    cancers Article Functional Dependency Analysis Identifies Potential Druggable Targets in Acute Myeloid Leukemia 1, 1, 2 3 Yujia Zhou y , Gregory P. Takacs y , Jatinder K. Lamba , Christopher Vulpe and Christopher R. Cogle 1,* 1 Division of Hematology and Oncology, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL 32610-0278, USA; yzhou1996@ufl.edu (Y.Z.); gtakacs@ufl.edu (G.P.T.) 2 Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL 32610-0278, USA; [email protected]fl.edu 3 Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL 32610-0278, USA; cvulpe@ufl.edu * Correspondence: [email protected]fl.edu; Tel.: +1-(352)-273-7493; Fax: +1-(352)-273-5006 Authors contributed equally. y Received: 3 November 2020; Accepted: 7 December 2020; Published: 10 December 2020 Simple Summary: New drugs are needed for treating acute myeloid leukemia (AML). We analyzed data from genome-edited leukemia cells to identify druggable targets. These targets were necessary for AML cell survival and had favorable binding sites for drug development. Two lists of genes are provided for target validation, drug discovery, and drug development. The deKO list contains gene-targets with existing compounds in development. The disKO list contains gene-targets without existing compounds yet and represent novel targets for drug discovery. Abstract: Refractory disease is a major challenge in treating patients with acute myeloid leukemia (AML). Whereas the armamentarium has expanded in the past few years for treating AML, long-term survival outcomes have yet to be proven. To further expand the arsenal for treating AML, we searched for druggable gene targets in AML by analyzing screening data from a lentiviral-based genome-wide pooled CRISPR-Cas9 library and gene knockout (KO) dependency scores in 15 AML cell lines (HEL, MV411, OCIAML2, THP1, NOMO1, EOL1, KASUMI1, NB4, OCIAML3, MOLM13, TF1, U937, F36P, AML193, P31FUJ).
    [Show full text]
  • Network Inference Analysis Identifies SETDB1 As a Key Regulator for Reverting Colorectal Cancer Cells Into Differentiated Normal-Like Cells
    MOLECULAR CANCER RESEARCH | NEW HORIZONS IN CANCER BIOLOGY Network Inference Analysis Identifies SETDB1 as a Key Regulator for Reverting Colorectal Cancer Cells into Differentiated Normal-Like Cells Soobeom Lee1, Chansu Lee2, Chae Young Hwang1, Dongsan Kim1, Younghyun Han1, Sung Noh Hong2, Seok-Hyung Kim3, and Kwang-Hyun Cho1 ABSTRACT ◥ Cancer cells exhibit properties of cells in a less differentiated state methyltransferase, hinders the function of the identified TFs. than the adjacent normal cells in the tissue. We explored whether SETDB1 depletion effectively converts stem-like colorectal cancer cancer cells can be converted to a differentiated normal-like state by cells into postmitotic cells and restores normal morphology in restoring the gene regulatory network (GRN) of normal cells. Here, patient-derived colorectal cancer organoids. RNA-sequencing anal- we report that colorectal cancer cells exhibit a range of develop- yses revealed that SETDB1 depletion recapitulates global gene mental states from embryonic and intestinal stem-like cells to expression profiles of normal differentiated cells by restoring the differentiated normal-like cells. To identify the transcription factors transcriptional activity of core TFs on their target genes. (TF) that commit stem-like colorectal cancer cells into a differen- tiated normal-like state, we reconstructed GRNs of normal colon Implications: Our study provides insights into the molecular mucosa and identified core TFs (CDX2, ELF3, HNF4G, PPARG, regulatory mechanism underlying the developmental hierarchy of and VDR) that govern the cellular state. We further found that SET colorectal cancer and suggests that induction of a postmitotic state Domain Bifurcated 1 (SETDB1), a histone H3 lysine 9–specific may be a therapeutic alternative to destruction of cancer cells.
    [Show full text]
  • SETDB1 Regulates Microtubule Dynamics
    bioRxiv preprint doi: https://doi.org/10.1101/2021.05.15.444210; this version posted May 16, 2021. 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. SETDB1 regulates microtubule dynamics Rosari Hernandez-Vicens1, Nomi Pernicone1,2, Tamar Listovsky1,2 and Gabi Gerlitz1,* 1 Department of Molecular Biology, Faculty of Life Sciences and Ariel Center for Applied Cancer Research, Ariel University, Ariel 40700, Israel. 2 Adelson School of Medicine, Ariel University, Ariel 40700, Israel *Correspondence to: Department of Molecular Biology, Faculty of Natural Sciences, Ariel University, Kiryat Hamada, Ariel 40700, Israel. E-mail addresses: [email protected] bioRxiv preprint doi: https://doi.org/10.1101/2021.05.15.444210; this version posted May 16, 2021. 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. Summary SETDB1 is a methyltransferase responsible for the methylation of histone H3-lysine-9, which is mainly related to heterochromatin formation. SETDB1 is overexpressed in various cancer types and is associated with an aggressive phenotype. In agreement with its activity, it mainly exhibits a nuclear localization; however, in several cell types a cytoplasmic localization was reported. Here we show that a substantial cytoplasmic pool of SETDB1 is colocalized with microtubules.
    [Show full text]
  • Fetal Alcohol Spectrum Disorder (FASD) Associated Neural Defects: Complex Mechanisms and Potential Therapeutic Targets
    Brain Sci. 2013, 3, 964-991; doi:10.3390/brainsci3020964 OPEN ACCESS brain sciences ISSN 2076-3425 www.mdpi.com/journal/brainsci/ Review Fetal Alcohol Spectrum Disorder (FASD) Associated Neural Defects: Complex Mechanisms and Potential Therapeutic Targets Pooja Muralidharan 1,†, Swapnalee Sarmah 1,†, Feng C. Zhou 2 and James A. Marrs 1,* 1 Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA; E-Mails: [email protected] (P.M.); [email protected] (S.S.) 2 Department of Anatomy and Cell Biology, Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA; E-Mail: [email protected] † These authors contributed equally to this work. * Author to whom correspondence should be addressed; E-Mail: [email protected]; Tel.: +1-317-278-0031; Fax: +1-317-274-2846. Received: 1 April 2013; in revised form: 3 June 2013 / Accepted: 4 June 2013 / Published: 19 June 2013 Abstract: Fetal alcohol spectrum disorder (FASD), caused by prenatal alcohol exposure, can result in craniofacial dysmorphism, cognitive impairment, sensory and motor disabilities among other defects. FASD incidences are as high as 2% to 5 % children born in the US, and prevalence is higher in low socioeconomic populations. Despite various mechanisms being proposed to explain the etiology of FASD, the molecular targets of ethanol toxicity during development are unknown. Proposed mechanisms include cell death, cell signaling defects and gene expression changes. More recently, the involvement of several other molecular pathways was explored, including non-coding RNA, epigenetic changes and specific vitamin deficiencies. These various pathways may interact, producing a wide spectrum of consequences.
    [Show full text]
  • Discovery of Potential Epigenetic Inhibitors Against Histone Methyltransferases Through Molecular Docking and Molecular Dynamics Simulations
    bioRxiv preprint doi: https://doi.org/10.1101/2020.04.13.040030; this version posted April 14, 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. Discovery of potential epigenetic inhibitors against histone methyltransferases through molecular docking and molecular dynamics simulations Tirumalasetty Muni Chandra Babua,#, Zaiping Zhangb,#, Danian Qina,*, Chengyang Huanga,b,c,* a Department of Physiology, Shantou University Medical College, Shantou 515041, China b Center for Neurobiology, Shantou University Medical College, Shantou 515041, China c Department of Biological Chemistry, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles California (UCLA) 90095, USA. # Equal contribution, T.M.C.B. and Z.P.Z. contributed equally in this work. *Corresponding author: Danian Qin ([email protected]) and Chengyang Huang ([email protected]) bioRxiv preprint doi: https://doi.org/10.1101/2020.04.13.040030; this version posted April 14, 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 Histone methyltransferases (HMTases) catalyze histone methylations that are the important epigenetic marks regulating gene expression, cell fate, and disease progression. In this study, we investigated potential epigenetic inhibitors against HMTases through in silico approaches, including ensembled molecular docking and molecular dynamics simulations (MDS).We identified three candidate compounds, including echinomycin, emetine, and streptonigrin, which showed interactions with HMTases. Echinomycin showed similar binding affinity with H3K4-HMTase NSD3 and H3K9-HMTase SETDB1 but streptonigrin and emetine had preferential binding affinity with NSD3 and SETDB1, respectively.
    [Show full text]
  • STAT3 Targets Suggest Mechanisms of Aggressive Tumorigenesis in Diffuse Large B Cell Lymphoma
    STAT3 Targets Suggest Mechanisms of Aggressive Tumorigenesis in Diffuse Large B Cell Lymphoma Jennifer Hardee*,§, Zhengqing Ouyang*,1,2,3, Yuping Zhang*,4 , Anshul Kundaje*,†, Philippe Lacroute*, Michael Snyder*,5 *Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305; §Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT 06520; and †Department of Computer Science, Stanford University School of Engineering, Stanford, CA 94305 1The Jackson Laboratory for Genomic Medicine, Farmington, CT 06030 2Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269 3Department of Genetics and Developmental Biology, University of Connecticut Health Center, Farmington, CT 06030 4Department of Biostatistics, Yale School of Public Health, Yale University, New Haven, CT 06520 5Corresponding author: Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305. Email: [email protected] DOI: 10.1534/g3.113.007674 Figure S1 STAT3 immunoblotting and immunoprecipitation with sc-482. Western blot and IPs show a band consistent with expected size (88 kDa) of STAT3. (A) Western blot using antibody sc-482 versus nuclear lysates. Lanes contain (from left to right) lysate from K562 cells, GM12878 cells, HeLa S3 cells, and HepG2 cells. (B) IP of STAT3 using sc-482 in HeLa S3 cells. Lane 1: input nuclear lysate; lane 2: unbound material from IP with sc-482; lane 3: material IP’d with sc-482; lane 4: material IP’d using control rabbit IgG. Arrow indicates the band of interest. (C) IP of STAT3 using sc-482 in K562 cells. Lane 1: input nuclear lysate; lane 2: material IP’d using control rabbit IgG; lane 3: material IP’d with sc-482.
    [Show full text]
  • Histone Methyltransferase SETDB1 Promotes Cells Proliferation And
    Zhang et al. BMC Cancer (2018) 18:539 https://doi.org/10.1186/s12885-018-4464-9 RESEARCH ARTICLE Open Access Histone methyltransferase SETDB1 promotes cells proliferation and migration by interacting withTiam1 in hepatocellular carcinoma Yuqin Zhang†, Jing Huang†, Qisheng Li, Keli Chen, Yonghao Liang, Zetao Zhan, Feng Ye, Wen Ni, Longhua Chen* and Yi Ding* Abstract Background: SETDB1 is a histone H3K9 methyltransferase, which plays a significant role in the occurrence and progression of tumors. Previous studies have confirmed that T-lymphom invasion and metastasis gene (Tiam1) is a protein associated with the metastasis of hepatocellular carcinoma (HCC); however, we have not yet been successful in elucidating the specific mechanism of HCC. Methods: Yeast two-hybrid test was conducted to screen proteins that interacted with Tiam1 gene. Glutathione-S-transferase (GST) pull-down and crosslinking-immunoprecipitation (CLIP) assays were performed to determine whether SETDB1 can interact with Tiam1 gene. A series of related experiments were performed to explore role of SETDB1 on cell proliferation, migration, and invasion in HCC. Recovery experiment was performedtoinvestigatetheeffectofTiam1knockdownoncellproliferationandmigration,whichwas caused by SETDB1 overexpression in HCC cells. The expression of SETDB1 was frequently upregulated in HCC tissues and positively correlated with Tiam1. Results: GST pull-down and CLIP assays were performed to elucidate the interaction between SETDB1 and Tiam1. Cell proliferation, migration, and epithelial mesenchymal transformation (EMT) in HCC cells was promoted with the overexpression of SETDB1. Following the knockdown of Tiam1 gene, the effect of SETDB1 on cell proliferation and migration was reversed in HCC cells. The expression of SETDB1 was frequently up-regulated in HCC tissues, and it was positively correlated with Tiam1 gene.
    [Show full text]