Genome-Wide CRISPR Screening Identifies BRD9 As a Druggable Component Of

Total Page:16

File Type:pdf, Size:1020Kb

Genome-Wide CRISPR Screening Identifies BRD9 As a Druggable Component Of bioRxiv preprint doi: https://doi.org/10.1101/2021.02.04.429732; this version posted February 4, 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 The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity.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 4.0 International license. 1 Genome-Wide CRISPR Screening Identifies BRD9 as a Druggable Component of 2 Interferon-Stimulated Gene Expression and Antiviral Activity 3 Jacob Börolda,b,†, Davide Elettoa,†,#, Idoia Busnadiegoa, Nina K. Maira,b, Eva Moritza, 4 Samira Schiefera,b, Nora Schmidta,$, Philipp P. Petricc,d, W. Wei-Lynn Wonge, Martin 5 Schwemmlec & Benjamin G. Halea,* 6 7 aInstitute of Medical Virology, University of Zurich, Zurich, Switzerland. 8 bLife Science Zurich Graduate School, ETH and University of Zurich, Zurich, 9 Switzerland. 10 cInstitute of Virology, Freiburg University Medical Center, Faculty of Medicine, University 11 of Freiburg, Freiburg, Germany. 12 dSpemann Graduate School of Biology and Medicine, University of Freiburg, Freiburg, 13 Germany. 14 eInstitute of Experimental Immunology, University of Zurich, Zurich, Switzerland. 15 *Correspondence: Benjamin G. Hale, [email protected] 16 †Equal first authors listed in alphabetical order. 17 #Present address: Department of Biosystems Science and Engineering, ETH Zurich, 18 Basel, Switzerland. 19 $Present address: Helmholtz Institute for RNA-based Infection Research, Helmholtz- 20 Center for Infection Research, Wurzburg, Germany. 1 bioRxiv preprint doi: https://doi.org/10.1101/2021.02.04.429732; this version posted February 4, 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 The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity.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 4.0 International license. 21 ABSTRACT 22 Transient interferon (IFN) induction of IFN-stimulated genes (ISGs) creates a formidable 23 protective antiviral state. However, loss of appropriate control mechanisms can result in 24 constitutive pathogenic ISG upregulation. Here, we used genome-wide loss-of-function 25 screening to establish genes critical for IFN signaling, identifying all expected members 26 of the JAK-STAT pathway and the previously unappreciated bromodomain-containing 27 protein 9 (BRD9), a defining subunit of non-canonical BAF (ncBAF) chromatin 28 remodeling complexes. Genetic knock-out or small-molecule mediated degradation of 29 BRD9 limited IFN-induced expression of a subset of ISGs in multiple cell-types, and 30 prevented IFN from exerting full antiviral activity against several RNA and DNA viruses. 31 Mechanistically, BRD9 acts at the level of ISG transcription, exhibits a proximal 32 association with STAT2 following IFN stimulation, and relies on its intact acetyl-binding 33 bromodomain and unique ncBAF scaffolding function for activity. Given its druggability, 34 BRD9 may be an attractive target for dampening constitutive ISG expression under 35 certain pathogenic autoinflammatory conditions. 36 37 38 39 40 2 bioRxiv preprint doi: https://doi.org/10.1101/2021.02.04.429732; this version posted February 4, 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 4.0 International license. 41 INTRODUCTION 42 The type I interferon (IFN-α/β) system is a key component of the human innate immune 43 response, and acts as a first line of defense against invading pathogens such as 44 viruses. At a general level, following host recognition of infection, secreted type I IFNs 45 relay signals to target cells via their binding to cognate cell surface receptors 46 (IFNAR1/IFNAR2) and the subsequent triggering of a well-defined phosphorylation- 47 activation JAK-STAT signaling cascade involving the Janus kinases (JAK1/TYK2) and 48 Signal Transducer and Activator of Transcription (STAT1/STAT2) proteins. Together 49 with IFN-regulatory factor 9 (IRF9), activated STAT1 and STAT2 form a heterotrimeric 50 transcription factor complex, termed ISGF3, that facilitates the transcription of hundreds 51 of antiviral IFN-stimulated genes (ISGs)1-3. The critical nature of the IFN system in 52 protecting humans against infection is exemplified by findings that some individuals with 53 genetic defects in IFN pathway components can exhibit severe, or even fatal, viral 54 diseases, particularly in the absence of pre-existing humoral immunity, as may be the 55 case in young infants or following infection with an antigenically-novel pandemic virus4,5. 56 It is essential that the IFN system is tightly regulated at the molecular level to prevent 57 exuberant proinflammatory responses following infection6-8. Furthermore, uncontrolled 58 activation of the IFN system caused by loss- or gain- of-function mutations in key 59 regulators of the IFN pathway can be associated with aberrantly high levels of 60 circulating IFNs and/or the constitutive expression of ISGs, leading to a broad range of 61 autoinflammatory disorders known as interferonopathies9-17. Proposed treatments for 62 interferonopathies include JAK inhibitors (such as baricitinib18, ruxolitinib19 or 3 bioRxiv preprint doi: https://doi.org/10.1101/2021.02.04.429732; this version posted February 4, 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 4.0 International license. 63 tofacitinib20) to limit the signaling action of high levels of constitutively circulating IFN or 64 inappropriate JAK-STAT regulation. However, while effective in treating 65 interferonopathies, inhibiting critical components of the IFN signaling pathway can 66 increase patient susceptibility to some viral infections21,22. Therefore a nuanced 67 approach targeting factors involved in transcription of specific ISGs may be more 68 appropriate22. 69 In this study, we sought to leverage CRISPR/Cas9-mediated genome-wide loss-of- 70 function screening to interrogate the IFN signaling cascade and identify human host 71 factors required for ISG expression. Our screening results led us to focus on 72 Bromodomain-containing protein 9 (BRD9), a defining subunit of specific chromatin- 73 remodeling complexes23-25, which has not previously been implicated in IFN responses. 74 Many factors involved in transcriptional regulation via chromatin modification or 75 remodeling play important roles in STAT-mediated ISG expression22,26-39, and some 76 (such as histone deacetylases (HDACs) and bromodomain-containing proteins22,26,35,39) 77 can be targeted pharmacologically. Here, we provide evidence that BRD9 contributes to 78 the expression of a subset of ISGs (and thus the full antiviral action of IFN) via its 79 unique functions in non-canonical BAF (BRG1- or BRM- associated factor; ncBAF) 80 chromatin remodeling complexes. Importantly, this contribution of BRD9 is conserved 81 across numerous human cell types, including primary cells, and against multiple distinct 82 viruses. Moreover, BRD9 can be specifically targeted for degradation by small-molecule 83 compounds without impacting cell viability. Our data add mechanistic insights into 84 human factors required for the full activity of IFN, and suggest a plausible therapeutic 4 bioRxiv preprint doi: https://doi.org/10.1101/2021.02.04.429732; this version posted February 4, 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 4.0 International license. 85 target to rationally temper detrimentally-high ISG levels in some autoinflammatory 86 disorders, including interferonopathies. 87 RESULTS 88 A Genome-Wide Loss-of-Function Screen Identifies Human Genes Important for 89 Interferon-Stimulated Gene Expression. The type I IFN signaling pathway leading to 90 induction of ISG expression is comprised of several canonical gene components, 91 including receptors (IFNAR1, IFNAR2), kinases (JAK1, TYK2), and transcription factors 92 (ISGF3: STAT1, STAT2, IRF9) (Figure 1A). To identify additional, previously 93 uncharacterized, genes critical for the ability of IFN to signal and induce ISG 94 expression, we performed a genome-wide FACS-based loss-of-function screen in the 95 human lung epithelial cell-line, A549. We used the GeCKOv2 CRISPR-Cas9 library, 96 which contains a total of 123,411 CRISPR single-guide RNAs (sgRNAs) targeting 97 19,050 genes in the human genome with 6 guide RNAs per gene40. First, we generated 98 a sub-clone of an A549-based reporter cell-line that expresses eGFP under control of 99 an IFN-stimulated response element (A549/pr(ISRE).eGFP.A1)41, and confirmed that 100 treatment of this sub-clone with IFN-α2 leads to high and homogenous expression of 101 eGFP, with clear separation of IFN-stimulated and non-stimulated cell populations in 102 flow cytometry analysis (Figure 1B). As a pre-screen validation, we next transduced 103 this reporter cell-line with all-in-one puromycin-resistant lentiviral vectors expressing 104 Cas9 and several control sgRNAs selected from the GeCKOv2 human library. Following 105 puromycin selection for 10 days to ensure protein depletion after CRISPR gene editing, 106 we observed that a significant proportion of reporter cells transduced with sgRNAs 5 bioRxiv preprint doi: https://doi.org/10.1101/2021.02.04.429732; this version posted February 4, 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 4.0 International license. 107 targeting the canonical pathway components, IFNAR1 or STAT1, failed to express 108 eGFP in response to IFN-α2 stimulation (Figure 1C). As expected, reporter cells 109 transduced with an sgRNA targeting a non-pathway component, IFNLR1, responded 110 similarly to IFN-α2 stimulation as the parental cell-line (Figure 1C).
Recommended publications
  • The Role of BRD7 in Embryo Development and Glucose Metabolism
    The role of BRD7 in embryo development and glucose metabolism The Harvard community has made this article openly available. Please share how this access benefits you. Your story matters Citation Kim, Yoo, Mario Andrés Salazar Hernández, Hilde Herrema, Tuncay Delibasi, and Sang Won Park. 2016. “The role of BRD7 in embryo development and glucose metabolism.” Journal of Cellular and Molecular Medicine 20 (8): 1561-1570. doi:10.1111/jcmm.12907. http://dx.doi.org/10.1111/jcmm.12907. Published Version doi:10.1111/jcmm.12907 Citable link http://nrs.harvard.edu/urn-3:HUL.InstRepos:29002534 Terms of Use This article was downloaded from Harvard University’s DASH repository, and is made available under the terms and conditions applicable to Other Posted Material, as set forth at http:// nrs.harvard.edu/urn-3:HUL.InstRepos:dash.current.terms-of- use#LAA J. Cell. Mol. Med. Vol 20, No 8, 2016 pp. 1561-1570 The role of BRD7 in embryo development and glucose metabolism Yoo Kim a, Mario Andres Salazar Hernandez a, Hilde Herrema a, Tuncay Delibasi b, Sang Won Park a, * a Division of Endocrinology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA b Department of Internal Medicine, School of Medicine, Kastamonu, Hacettepe University, Ankara, Turkey Received: May 4, 2016; Accepted: May 17, 2016 Abstract Bromodomain-containing protein 7 (BRD7) is a member of bromodomain-containing protein family and its function has been implicated in sev- eral diseases. We have previously shown that BRD7 plays a role in metabolic processes. However, the effect of BRD7 deficiency in glucose metabolism and its role in in vivo have not been fully revealed.
    [Show full text]
  • Increased ACTL6A Occupancy Within Mswi/SNF Chromatin Remodelers
    bioRxiv preprint doi: https://doi.org/10.1101/2021.03.22.435873; this version posted March 22, 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. Increased ACTL6A Occupancy Within mSWI/SNF Chromatin Remodelers Drives Human Squamous Cell Carcinoma Chiung-Ying Chang1,2,7, Zohar Shipony3,7, Ann Kuo1,2, Kyle M. Loh4,5, William J. Greenleaf3,6, Gerald R. Crabtree1,2,5* 1Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, California 94305, USA. 2Department of Pathology, Stanford University School of Medicine, Stanford, California 94305, USA. 3Department of Genetics, Stanford University School of Medicine, Stanford, California 94305, USA. 4Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California 94305, USA. 5Department of Developmental Biology, Stanford University School of Medicine, Stanford, California 94305, USA. 6Department of Applied Physics, Stanford University, Stanford, California 94305, USA. 7These authors contributed equally. *Corresponding author: Gerald R. Crabtree, [email protected] Summary Mammalian SWI/SNF (BAF) chromatin remodelers play dosage-sensitive roles in many human malignancies and neurologic disorders. The gene encoding the BAF subunit, ACTL6A, is amplified at an early stage in the development of squamous cell carcinomas (SCCs), but its oncogenic role remains unclear. Here we demonstrate that ACTL6A overexpression leads to its stoichiometric assembly into BAF complexes and drives its interaction and engagement with specific regulatory regions in the genome. In normal epithelial cells, ACTL6A was sub-stoichiometric to other BAF subunits. However, increased ACTL6A levels by ectopic expression or in SCC cells led to near-saturation of ACTL6A within BAF complexes.
    [Show full text]
  • Ten Commandments for a Good Scientist
    Unravelling the mechanism of differential biological responses induced by food-borne xeno- and phyto-estrogenic compounds Ana María Sotoca Covaleda Wageningen 2010 Thesis committee Thesis supervisors Prof. dr. ir. Ivonne M.C.M. Rietjens Professor of Toxicology Wageningen University Prof. dr. Albertinka J. Murk Personal chair at the sub-department of Toxicology Wageningen University Thesis co-supervisor Dr. ir. Jacques J.M. Vervoort Associate professor at the Laboratory of Biochemistry Wageningen University Other members Prof. dr. Michael R. Muller, Wageningen University Prof. dr. ir. Huub F.J. Savelkoul, Wageningen University Prof. dr. Everardus J. van Zoelen, Radboud University Nijmegen Dr. ir. Toine F.H. Bovee, RIKILT, Wageningen This research was conducted under the auspices of the Graduate School VLAG Unravelling the mechanism of differential biological responses induced by food-borne xeno- and phyto-estrogenic compounds Ana María Sotoca Covaleda Thesis submitted in fulfillment of the requirements for the degree of doctor at Wageningen University by the authority of the Rector Magnificus Prof. dr. M.J. Kropff, in the presence of the Thesis Committee appointed by the Academic Board to be defended in public on Tuesday 14 September 2010 at 4 p.m. in the Aula Unravelling the mechanism of differential biological responses induced by food-borne xeno- and phyto-estrogenic compounds. Ana María Sotoca Covaleda Thesis Wageningen University, Wageningen, The Netherlands, 2010, With references, and with summary in Dutch. ISBN: 978-90-8585-707-5 “Caminante no hay camino, se hace camino al andar. Al andar se hace camino, y al volver la vista atrás se ve la senda que nunca se ha de volver a pisar” - Antonio Machado – A mi madre.
    [Show full text]
  • TP53 Mutations As a Driver of Metastasis Signaling in Advanced Cancer Patients
    cancers Article TP53 Mutations as a Driver of Metastasis Signaling in Advanced Cancer Patients Ritu Pandey 1,2,*, Nathan Johnson 3, Laurence Cooke 1, Benny Johnson 4, Yuliang Chen 1, Manjari Pandey 5, Jason Chandler 5 and Daruka Mahadevan 6,* 1 Cancer Center, University of Arizona, Tucson, AZ 85724, USA; [email protected] (L.C.); [email protected] (Y.C.) 2 Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85724, USA 3 School of Medicine, Vanderbilt University, Nashville, TN 37325, USA; [email protected] 4 MD Anderson Cancer Center, Houston, TX 77030, USA; [email protected] 5 West Cancer Center, 7945 Wolf River Blvd, Germantown, TN 38138, USA; [email protected] (M.P.); [email protected] (J.C.) 6 Mays Cancer Center, University of Texas Health San Antonio, San Antonio, TX 78229, USA * Correspondence: [email protected] (R.P.); [email protected] (D.M.) Simple Summary: The DNA sequencing of cancer provides information about specific genetic changes that could help with treatment decisions. Tumors from different tissues change over time and acquire new genetic changes particularly with treatment. Our study analyzed the gene changes of 171 advanced cancer patients. We predicted that tumors gain new mutations but that TP53 mutations (guardian of the human genome) are conserved as the tumor progresses from primary to metastatic sites and across tissue types. We analyzed the primary and metastatic site gene changes in 25 tissue types and conducted in-depth analysis of colon and lung cancer sites for substantial changes. TP53 site specific mutations were different across tissue types and suggest different molecular changes.
    [Show full text]
  • SMARCA4 Regulates Spatially Restricted Metabolic Plasticity in 3D Multicellular Tissue
    bioRxiv preprint doi: https://doi.org/10.1101/2021.03.21.436346; this version posted March 22, 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. SMARCA4 regulates spatially restricted metabolic plasticity in 3D multicellular tissue Katerina Cermakova1,2,*, Eric A. Smith1,2,*, Yuen San Chan1,2, Mario Loeza Cabrera1,2, Courtney Chambers1,2, Maria I. Jarvis3, Lukas M. Simon4, Yuan Xu5, Abhinav Jain6, Nagireddy Putluri1, Rui Chen7, R. Taylor Ripley5, Omid Veiseh3, and H. Courtney Hodges1,2,3,8,9,‡ 1. Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA 2. Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, USA 3. Department of Bioengineering, Rice University, Houston, TX, USA 4. Therapeutic Innovation Center, Baylor College of Medicine, Houston, TX, USA 5. Department of Surgery, Division of General Thoracic Surgery, Baylor College of Medicine, Houston, TX, USA 6. Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, USA 7. Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA 8. Center for Cancer Epigenetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA 9. Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA * These authors contributed equally to this work ‡ Corresponding author: H. Courtney Hodges, Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA, [email protected]. Abstract SWI/SNF and related chromatin remodeling complexes act as tissue-specific tumor suppressors and are frequently inactivated in different cancers.
    [Show full text]
  • BRG1 Knockdown Inhibits Proliferation Through Multiple Cellular Pathways in Prostate Cancer Katherine A
    Giles et al. Clin Epigenet (2021) 13:37 https://doi.org/10.1186/s13148-021-01023-7 RESEARCH Open Access BRG1 knockdown inhibits proliferation through multiple cellular pathways in prostate cancer Katherine A. Giles1,2,3, Cathryn M. Gould1, Joanna Achinger‑Kawecka1,4, Scott G. Page2, Georgia R. Kafer2, Samuel Rogers2, Phuc‑Loi Luu1,4, Anthony J. Cesare2, Susan J. Clark1,4† and Phillippa C. Taberlay3*† Abstract Background: BRG1 (encoded by SMARCA4) is a catalytic component of the SWI/SNF chromatin remodelling com‑ plex, with key roles in modulating DNA accessibility. Dysregulation of BRG1 is observed, but functionally uncharacter‑ ised, in a wide range of malignancies. We have probed the functions of BRG1 on a background of prostate cancer to investigate how BRG1 controls gene expression programmes and cancer cell behaviour. Results: Our investigation of SMARCA4 revealed that BRG1 is over‑expressed in the majority of the 486 tumours from The Cancer Genome Atlas prostate cohort, as well as in a complementary panel of 21 prostate cell lines. Next, we utilised a temporal model of BRG1 depletion to investigate the molecular efects on global transcription programmes. Depleting BRG1 had no impact on alternative splicing and conferred only modest efect on global expression. How‑ ever, of the transcriptional changes that occurred, most manifested as down‑regulated expression. Deeper examina‑ tion found the common thread linking down‑regulated genes was involvement in proliferation, including several known to increase prostate cancer proliferation (KLK2, PCAT1 and VAV3). Interestingly, the promoters of genes driving proliferation were bound by BRG1 as well as the transcription factors, AR and FOXA1.
    [Show full text]
  • Intrinsic Disorder of the BAF Complex: Roles in Chromatin Remodeling and Disease Development
    International Journal of Molecular Sciences Article Intrinsic Disorder of the BAF Complex: Roles in Chromatin Remodeling and Disease Development Nashwa El Hadidy 1 and Vladimir N. Uversky 1,2,* 1 Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, 12901 Bruce B. Downs Blvd. MDC07, Tampa, FL 33612, USA; [email protected] 2 Laboratory of New Methods in Biology, Institute for Biological Instrumentation of the Russian Academy of Sciences, Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences”, Pushchino, 142290 Moscow Region, Russia * Correspondence: [email protected]; Tel.: +1-813-974-5816; Fax: +1-813-974-7357 Received: 20 September 2019; Accepted: 21 October 2019; Published: 23 October 2019 Abstract: The two-meter-long DNA is compressed into chromatin in the nucleus of every cell, which serves as a significant barrier to transcription. Therefore, for processes such as replication and transcription to occur, the highly compacted chromatin must be relaxed, and the processes required for chromatin reorganization for the aim of replication or transcription are controlled by ATP-dependent nucleosome remodelers. One of the most highly studied remodelers of this kind is the BRG1- or BRM-associated factor complex (BAF complex, also known as SWItch/sucrose non-fermentable (SWI/SNF) complex), which is crucial for the regulation of gene expression and differentiation in eukaryotes. Chromatin remodeling complex BAF is characterized by a highly polymorphic structure, containing from four to 17 subunits encoded by 29 genes. The aim of this paper is to provide an overview of the role of BAF complex in chromatin remodeling and also to use literature mining and a set of computational and bioinformatics tools to analyze structural properties, intrinsic disorder predisposition, and functionalities of its subunits, along with the description of the relations of different BAF complex subunits to the pathogenesis of various human diseases.
    [Show full text]
  • SMARCA4 Regulates Gene Expression and Higher-Order Chromatin Structure in Proliferating Mammary Epithelial Cells
    Downloaded from genome.cshlp.org on October 1, 2021 - Published by Cold Spring Harbor Laboratory Press Research SMARCA4 regulates gene expression and higher- order chromatin structure in proliferating mammary epithelial cells A. Rasim Barutcu,1 Bryan R. Lajoie,2 Andrew J. Fritz,3 Rachel P. McCord,4 Jeffrey A. Nickerson,1 Andre J. van Wijnen,5 Jane B. Lian,3 Janet L. Stein,3 Job Dekker,2,6 Gary S. Stein,3 and Anthony N. Imbalzano1 1Department of Cell and Developmental Biology, University of Massachusetts Medical School, Worcester, Massachusetts 01655, USA; 2Program in Systems Biology, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA; 3Department of Biochemistry, University of Vermont College of Medicine, Burlington, Vermont 05405, USA; 4Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, Tennessee 37996, USA; 5Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota 55905, USA; 6Howard Hughes Medical Institute, Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA The packaging of DNA into chromatin plays an important role in transcriptional regulation and nuclear processes. Brahma- related gene-1 SMARCA4 (also known as BRG1), the essential ATPase subunit of the mammalian SWI/SNF chromatin re- modeling complex, uses the energy from ATP hydrolysis to disrupt nucleosomes at target regions. Although the transcrip- tional role of SMARCA4 at gene promoters is well-studied, less is known about its role in higher-order genome organization. SMARCA4 knockdown in human mammary epithelial MCF-10A cells resulted in 176 up-regulated genes, includ- ing many related to lipid and calcium metabolism, and 1292 down-regulated genes, some of which encode extracellular ma- trix (ECM) components that can exert mechanical forces and affect nuclear structure.
    [Show full text]
  • BRD7 Expression and C-Myc Activation
    Liu et al. Journal of Experimental & Clinical Cancer Research (2018) 37:64 https://doi.org/10.1186/s13046-018-0734-2 RESEARCH Open Access BRD7 expression and c-Myc activation forms a double-negative feedback loop that controls the cell proliferation and tumor growth of nasopharyngeal carcinoma by targeting oncogenic miR-141 Yukun Liu1,2,3, Ran Zhao2,3, Yanmei Wei2,3, Mengna Li2,3, Heran Wang1,3, Weihong Niu2,3, Yao Zhou2,3, Yuanzheng Qiu4, Songqing Fan5, Yihao Zhan6, Wei Xiong1,2,3, Yanhong Zhou1,2,3, Xiaoling Li1,2,3, Zheng Li1,2,3, Guiyuan Li1,2,3 and Ming Zhou1,2,3* Abstract Background: miR-141 is up-regulated and plays crucial roles in nasopharyngeal carcinoma (NPC). However, the molecular mechanism underlying the dysregulation of miR-141 is still obscure. Methods: Thus, the ChIP-PCR was performed to identify the c-Myc-binding sites in miR-141 and BRD7. qRT-PCR, western blot and immunohistochemistry assays were used to detect the expression of miR-141 and its up/down stream molecules. The rescue experiments on the c-Myc/miR-141 axis were performed in vitro and in vivo. Results: Our results showed that the levels of mature miR-141, pre-miR-141 and pri-miR-141 were downregulated in c-Myc knockdown NPC cells. Meanwhile, c-Myc transactivates the expression of miR-141 by binding its promoter region. Moreover, BRD7 was identified as a co-factor of c-Myc to negatively regulate the activation of c-Myc/miR-141 axis, as well as a direct target of c-Myc.
    [Show full text]
  • Emerging Roles of BRD7 in Pathophysiology
    International Journal of Molecular Sciences Review Emerging Roles of BRD7 in Pathophysiology Sang Won Park 1,2,* and Junsik M. Lee 1 1 Division of Endocrinology, Boston Children’s Hospital, Boston, MA 02115, USA; [email protected] 2 Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA * Correspondence: [email protected] Received: 7 September 2020; Accepted: 23 September 2020; Published: 27 September 2020 Abstract: Bromodomain is a conserved structural module found in many chromatin-associated proteins. Bromodomain-containing protein 7 (BRD7) is a member of the bromodomain-containing protein family, and was discovered two decades ago as a protein that is downregulated in nasopharyngeal carcinoma. Since then, BRD7 has been implicated in a variety of cellular processes, including chromatin remodeling, transcriptional regulation, and cell cycle progression. Decreased BRD7 activity underlies the pathophysiological properties of various diseases in different organs. BRD7 plays an important role in the pathogenesis of many cancers and, more recently, its roles in the regulation of metabolism and obesity have also been highlighted. Here, we review the involvement of BRD7 in a variety of pathophysiological conditions, with a focus on glucose homeostasis, obesity, and cancer. Keywords: BRD7; glucose metabolism; type 2 diabetes; XBP1s; PI3K; p85; cancer 1. Introduction Bromodomain is an evolutionarily conserved module, which consists of 110 amino acids [1,2]. It recognizes acetylated lysine residues through its hydrophobic cavity formed by four α-helices and two loops, and modulates the activity of other proteins [1,3–5]. The first protein reported to contain bromodomain was the Brahma protein, a regulator of Drosophila homeotic genes [6].
    [Show full text]
  • A Role for SMARCB1 in Synovial Sarcomagenesis Reveals That SS18-SSX Induces Canonical BAF Destruction
    Author Manuscript Published OnlineFirst on June 2, 2021; DOI: 10.1158/2159-8290.CD-20-1219 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. SMARCB1 in Synovial Sarcomagenesis 1 A role for SMARCB1 in synovial sarcomagenesis reveals that SS18-SSX induces canonical BAF destruction Jinxiu Li*1,2,3, Timothy S. Mulvihill*2,3, Li Li1,2,3, Jared J. Barrott1,2,3, Mary L. Nelson1,2,3, Lena Wagner6, Ian C. Lock1,2,3, Amir Pozner1,2,3, Sydney Lynn Lambert1,2,3, Benjamin B. Ozenberger1,2,3, Michael B. Ward3,4, Allie H. Grossmann3,4, Ting Liu3,4, Ana Banito6, Bradley R. Cairns2,3,5† and Kevin B. Jones1,2,3† 1Department of Orthopaedics, 2Department of Oncological Sciences, 3Huntsman Cancer Institute, 4Department of Pathology, 5Howard Hughes Medical Institute, University of Utah, Salt Lake City, Utah. 6Hopp Children’s Cancer Center (KiTZ), German Cancer Research Center (DFKZ), Heidelberg, Germany. *These authors contributed equally to this work. †These authors are co-corresponding authors. Please Address Correspondence to: Kevin B. Jones and Bradley R. Cairns Address: 2000 Circle of Hope Drive, Salt Lake City, UT 84112 Phone: 801-585-0300 Fax: 801-585-7084 Email: [email protected], [email protected] Running Title: SMARCB1 in Synovial Sarcomagenesis Key Words: SWI/SNF; Chromatin Remodeling; Mouse Genetic Model; Epigenetics; Biochemistry Financial Support: This work was supported by R01CA201396 (Jones and Cairns), U54CA231652 (Jones, Cairns, and Banito), and 2P30CA042014-31, from the National Cancer Institute (NCI/NIH), as well as the Paul Nabil Bustany Fund for Synovial Sarcoma Research (Jones), and the Sarcoma Foundation of America (Barrott).
    [Show full text]
  • Residual Complexes Containing SMARCA2 (BRM) Underlie the Oncogenic Drive of SMARCA4 (BRG1) Mutation
    Residual Complexes Containing SMARCA2 (BRM) Underlie the Oncogenic Drive of SMARCA4 (BRG1) Mutation The Harvard community has made this article openly available. Please share how this access benefits you. Your story matters Citation Wilson, B. G., K. C. Helming, X. Wang, Y. Kim, F. Vazquez, Z. Jagani, W. C. Hahn, and C. W. M. Roberts. 2014. “Residual Complexes Containing SMARCA2 (BRM) Underlie the Oncogenic Drive of SMARCA4 (BRG1) Mutation.” Molecular and Cellular Biology 34 (6): 1136–44. doi:10.1128/MCB.01372-13. Citable link http://nrs.harvard.edu/urn-3:HUL.InstRepos:41542741 Terms of Use This article was downloaded from Harvard University’s DASH repository, and is made available under the terms and conditions applicable to Other Posted Material, as set forth at http:// nrs.harvard.edu/urn-3:HUL.InstRepos:dash.current.terms-of- use#LAA Residual Complexes Containing SMARCA2 (BRM) Underlie the Oncogenic Drive of SMARCA4 (BRG1) Mutation Boris G. Wilson,a Katherine C. Helming,a Xiaofeng Wang,a Youngha Kim,a Francisca Vazquez,c,d Zainab Jagani,f William C. Hahn,c,d,e Charles W. M. Robertsa,b,d,e Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USAa; Division of Hematology/Oncology, Boston Children’s Hospital, Boston, Massachusetts, USAb; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USAc; Cancer Program, Broad Institute of Harvard and Massachusetts Institute of Technology, Boston, Massachusetts, USAd; Center for Cancer Genome Discovery, Dana-Farber Cancer Institute, Boston, Massachusetts, USAe; Novartis Institutes for BioMedical Research, Cambridge, Massachusetts, USAf Collectively, genes encoding subunits of the SWI/SNF (BAF) chromatin remodeling complex are mutated in 20% of all human Downloaded from cancers, with the SMARCA4 (BRG1) subunit being one of the most frequently mutated.
    [Show full text]