PRC2-Mediated Repression of SMARCA2 Predicts EZH2 Inhibitor Activity in SWI/SNF Mutant Tumors

Total Page:16

File Type:pdf, Size:1020Kb

PRC2-Mediated Repression of SMARCA2 Predicts EZH2 Inhibitor Activity in SWI/SNF Mutant Tumors PRC2-mediated repression of SMARCA2 predicts EZH2 inhibitor activity in SWI/SNF mutant tumors Thomas Januarioa,1, Xiaofen Yea,1, Russell Bainerb, Bruno Alickec, Tunde Smitha, Benjamin Haleyd, Zora Modrusand, Stephen Gouldc, and Robert L. Yaucha,2 aDepartment of Discovery Oncology, Genentech, Inc., South San Francisco, CA 94080; bDepartment of Bioinformatics, Genentech, Inc., South San Francisco, CA 94080; cDepartment of Translational Oncology, Genentech, Inc., South San Francisco, CA 94080; and dDepartment of Molecular Biology, Genentech, Inc., South San Francisco, CA 94080 Edited by Joan S. Brugge, Harvard Medical School, Boston, MA, and approved October 3, 2017 (received for review March 8, 2017) Subunits of the SWI/SNF chromatin remodeling complex are fre- of the ovary, hypercalcemic-type (SCCOHT) (3, 7–9). Although the quently mutated in human cancers leading to epigenetic dependen- mechanisms underlying tumorigenesis in these specific contexts cies that are therapeutically targetable. The dependency on the have yet to be fully elucidated, data are further supportive of a polycomb repressive complex (PRC2) and EZH2 represents one such tumor-suppressive function (10, 11). vulnerability in tumors with mutations in the SWI/SNF complex Efforts to therapeutically target SWI/SNF-defective cancers subunit, SNF5; however, whether this vulnerability extends to other have focused on identifying novel vulnerabilities that may be a SWI/SNF subunit mutations is not well understood. Here we show consequence of the altered chromatin state caused by muta- that a subset of cancers harboring mutations in the SWI/SNF ATPase, tions in BAF complex subunits. One such described vulnera- bility was based on the initial discovery in Drosophila of an SMARCA4, is sensitive to EZH2 inhibition. EZH2 inhibition results in a opposing and antagonistic role of BAF and polycomb com- heterogenous phenotypic response characterized by senescence plexes in regulating gene expression (12). Subsequent studies and/or apoptosis in different models, and also leads to tumor revealed that this antagonistic relationship may result in human growth inhibition in vivo. Lower expression of the SMARCA2 cancers with specific defects in BAF subunits to become de- paralog was associated with cellular sensitivity to EZH2 inhibition pendent on the activity of the polycomb repressive group 2 in SMARCA4 mutant cancer models, independent of tissue deriva- (PRC2) complex. This is best exemplified in MRTs, as loss of tion. SMARCA2 is suppressed by PRC2 in sensitive models, and SNF5 results in the altered genomic occupancy of the repressive MEDICAL SCIENCES induced SMARCA2 expression can compensate for SMARCA4 and chromatin mark deposited by PRC2 at histone H3 lysine 27 resi- antagonize PRC2 targets. The induction of SMARCA2 in response to dues (H3K27me3), leading to the repression of lineage-specific EZH2 inhibition is required for apoptosis, but not for growth arrest, targets (13). In these models, disruption of the histone methyl- through a mechanism involving the derepression of the lysomal transferase activity of EZH2, the catalytic subunit of PRC2, im- protease cathepsin B. Expression of SMARCA2 also delineates EZH2 paired tumor growth, thereby demonstrating that SNF5 mutant inhibitor sensitivity for other SWI/SNF complex subunit mutant tumors depend on EZH2 activity (13, 14). More recently, a similar tumors, including SNF5 and ARID1A mutant cancers. Our data support dependency on EZH2 was described in the context of ARID1A monitoring SMARCA2 expression as a predictive biomarker for EZH2- mutant cancers, suggesting that PRC2 activity may be a common targeted therapies in the context of SWI/SNF mutant cancers. vulnerability in SWI/SNF-defective lesions (15). However, whether targeting EZH2 will be effective in all cancers harboring these EZH2 | SMARCA4 | SMARCA2 | PRC2 | ARID1A specific mutations or in other SWI/SNF subunit mutant contexts remains an open question. WI/SNF, or BAF (Brg/Brahma-associated factors), com- Significance Splexes compose a family of ATP-dependent chromatin remodeling complexes that play critical roles in controlling gene transcription and DNA repair through their ability to regulate Targeting epigenetic dependencies caused by mutations in the accessibility of DNA in the nucleus (1, 2). In mammals, these chromatin-modifying enzymes represents a novel therapeutic complexes are composed of approximately 15 subunits, including approach in cancer. Notably, cancers harboring mutations in the either the SMARCA4 or SMARCA2 ATPase and additional SNF5 subunit of the SWI/SNF chromatin remodeling complex core and accessory subunits whose specialized protein domains have been shown to be susceptible to small-molecule inhibitors enable interactions with chromatin and/or DNA substrates. SWI/ of the EZH2 histone methyltransferase that are currently in clin- SNF complexes notably control lineage specification and dif- ical development. We demonstrate that EZH2 inhibition can be ferentiation programs in tissues, and these highly specific func- effective in SMARCA4 mutant cancers that concurrently tran- tions can be achieved through combinatorial subunit assembly, as scriptionally silence the paralog helicase SMARCA2. SMARCA2 is well as tissue/lineage-specific expression of some subunits. directly suppressed by EZH2, and SMARCA2 expression levels Large-scale cancer genome sequencing efforts have revealed that predict EZH2 inhibitor activity in other SWI/SNF mutant contexts, multiple SWI/SNF complex proteins are recurrently mutated in including ARID1A mutant tumors. These data provide insight into ∼20% of human cancers (3). Mutations often render the encoded the utility of EZH2 inhibitors in SWI/SNF mutant tumors and have protein nonfunctional and associate with unique tumor spectra, important implications regarding predictive diagnostics. suggesting distinct tumor-suppressor functions across these cancers. For example, the SWI/SNF complex subunit SNF5 is subject to Author contributions: T.J., X.Y., R.B., S.G., and R.L.Y. designed research; T.J., X.Y., B.A., T.S., B.H., Z.M., S.G., and R.L.Y. performed research; T.J., X.Y., R.B., B.A., T.S., and R.L.Y. biallelic mutational inactivation in nearly all malignant rhabdoid analyzed data; and R.L.Y. wrote the paper. tumors (MRTs), and studies in genetically engineered mouse models of SNF5 inactivation have supported its role as a bona fide The authors declare no conflict of interest. tumor suppressor (3–6). Similarly, recurrent inactivating mutations This article is a PNAS Direct Submission. in additional BAF subunits have been identified in other indica- Published under the PNAS license. tions, including ARID1A mutations in ovarian clear-cell and 1T.J and X.Y. contributed equally to this work. endometroid cancers, as well as hepatocellular and gastric cancers. 2To whom correspondence should be addressed. Email: [email protected]. SMARCA4 mutations likewise are observed across a spectrum of This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. cancers of the lung and bladder, and nearly all small cell carcinomas 1073/pnas.1703966114/-/DCSupplemental. www.pnas.org/cgi/doi/10.1073/pnas.1703966114 PNAS Early Edition | 1of6 Downloaded by guest on September 26, 2021 Multiple inhibitors targeting the enzymatic activity of EZH2 are AB currently in clinical development, with EPZ-6438 (tazemontstat) 20 20 representing the most clinically advanced molecule. Early clinical 15 7 days 15 13 days 10 10 data presented with EPZ-6438 has shown promise, as objective 5 5 4 4 responses have been observed in a subset of SNF5 mutant and H1299 TOV-112D TOV-112D 3 3 SMARCA4 mutant patients treated with EPZ-6438 as a single 2 2 1 1 Caspase 3/7 activity (fc) agent. These data not only begin to provide early clinical proof of 0 0 NA EPZ-6438: concept, but indicate that EZH2 inhibition may be effective in the A549 SNU-484 A549 A549 HCC15 context of SMARCA4 mutant cancers, a preclinical finding that has HCC15 COV434 COV434 SNU-484 SNU-484 UM-UC-3 UM-UC-3 NCI-H522 NCI-H661 NCI-H522 NCI-H661 TOV-112D TOV-112D NCI-H1703 NCI-H1299 NCI-H1568 yet to be published. Notably, not all patients with tumors harboring NCI-H1703 NCI-H1299 NCI-H1568 EPZ-6438 EPZ-6438 EPZ-6438 EPZ-6438 SNF5 or SMARCA4 defects responded to therapy, suggesting that sensitive resistant sensitive resistant identifying a biomarker predictive of response to EZH2 inhibition NCI-H1568 could provide significant benefit. In the present study, we demon- NCI-H1703 C control EPZ-6438 strate that EZH2 inhibition is effective in a subset of SMARCA4 mutant cancer models, and that the PRC2-mediated transcriptional UM-UC-3 NCI-H522 suppression of the paralog ATPase, SMARCA2, can predict the Cov-434 preclinical activity of EZH2 inhibitors. Importantly, we show that the level of SMARCA2 expression may be a global predictive HCC-15 COV434 biomarker of EZH2 activity in other BAF mutant cancers. NCI-H522 Results A Subset of SMARCA4 Mutant Cancers Is Responsive to EZH2 Inhibition. NCI-H661 We evaluated the effect of EZH2 inhibition using the EZH2- NCI-H1568 targeting histone methyltransferase inhibitor, EPZ-6438, on clo- SNF5mut control EPZ-6438 ( M): D vehicle nogenic growth across a panel of 11 SMARCA4 mutated cancer EPZ-6438 (30mg/kg) cell lines derived from different tumor types (Dataset S1). A dose- 500 EPZ-6438 (100mg/kg) 0 .06 .18 ) 3 EPZ-6438 (450mg/kg) dependent inhibition of clonogenic growth independent of tissue 400 derivation was observed in a subset of these SMARCA4 mutant cell .56 1.67 5 lines (Fig. 1A). The degree of growth inhibition on EPZ-6438 300 treatment was similar to that previously described in models 200 100 characterized by mutations in SNF5 (G401). No activity was ob- (mm volume tumor = 0 served in a panel of SWI/SNF wild-type models (n 8). The dif- 0 102030 ferential sensitivity to EPZ-6438 was not due to differences in Day target engagement, as a similar dose-dependent inhibition of H3K27 methylation was observed in sensitive and resistant models Fig. 1. EZH2 inhibition suppresses growth in a subset of SMARCA4 mutant (SI Appendix,Fig.S1).
Recommended publications
  • Chromatin Remodeling in Mice
    University of Tennessee, Knoxville TRACE: Tennessee Research and Creative Exchange Supervised Undergraduate Student Research Chancellor’s Honors Program Projects and Creative Work Spring 5-2005 Chromatin Remodeling in Mice Stephen James Dolgner University of Tennessee - Knoxville Follow this and additional works at: https://trace.tennessee.edu/utk_chanhonoproj Recommended Citation Dolgner, Stephen James, "Chromatin Remodeling in Mice" (2005). Chancellor’s Honors Program Projects. https://trace.tennessee.edu/utk_chanhonoproj/843 This is brought to you for free and open access by the Supervised Undergraduate Student Research and Creative Work at TRACE: Tennessee Research and Creative Exchange. It has been accepted for inclusion in Chancellor’s Honors Program Projects by an authorized administrator of TRACE: Tennessee Research and Creative Exchange. For more information, please contact [email protected]. Chromatin Remodeling in Mice Stephen Dolgner Advisor: Dr. Sundaresan Venkatachalam May 2005 ABSTRACT Controlling gene regulation is an important aspect in the life of cells that provides them the ability to carry out their functional roles within an organism. Unregulated or misregulated gene expression can lead to cell immortalization or death. Chromatin remodeling functions as a regulator for many important DNA functions including transcription, the first step of gene expression in cells. The Chromodomain-Helicase­ DNA binding domain gene family (CHD) is evolutionarily conserved and has distinct structural motifs that indicate a role in chromatin remodeling and DNA repair. The CHD proteins have both helicase activity, allowing the winding and unwinding of DNA, and an effect on histone acetylation through their role of the Nucleosome Remodeling and Histone Deacetylation (NuRD) complex. The NuRD complex participates in the deacetylation of chromatin histones, in addition to orchestrating ATP-dependent remodeling of the the chromatin structure.
    [Show full text]
  • 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.
    [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]
  • The Gene Repressor Complex Nurd Interacts with the Histone Variant H3.3
    Downloaded from genome.cshlp.org on October 3, 2021 - Published by Cold Spring Harbor Laboratory Press The gene repressor complex NuRD interacts with the histone variant H3.3 at promoters of active genes Daniel C. Kraushaar1,3,4, Zuozhou Chen2,4, Qingsong Tang1, Kairong Cui1, Junfang Zhang2,*, Keji Zhao1,* 1 Systems Biology Center, National Heart, Lung, and Blood Institute, NIH, MD 2 Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Shanghai Ocean University), Ministry of Education; International Research Center for Marine Biosciences at Shanghai Ocean University, Ministry of Science and Technology; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai 201306, China. 3 Current address: Genomic and RNA Profiling Core, Baylor College of Medicine, TX 4These authors contributed equally to this work *Correspondence: Junfang Zhang ([email protected]); Keji Zhao ([email protected]) Key words: histone variant H3.3; chromatin modification; epigenetics; ChIP-seq; protein interaction; NuRD; histone modification 1 Downloaded from genome.cshlp.org on October 3, 2021 - Published by Cold Spring Harbor Laboratory Press Abstract The histone variant H3.3 is deposited across active genes, regulatory regions and telomeres. It remains unclear how H3.3 interacts with chromatin modifying enzymes and thereby modulates gene activity. In this study, we performed a co- immunoprecipitation-mass spectrometry analysis of proteins associated with H3.3-containing nucleosomes and identified the Nucleosome Remodeling and Deacetylase complex (NuRD) as a major H3.3-interactor. We show that the H3.3- NuRD interaction is dependent on the H3.3 lysine 4 residue and that NuRD binding occurs when lysine 4 is in its unmodified state.
    [Show full text]
  • EZH2 Reduction Is an Essential Mechanoresponse for The
    Li et al. Cell Death and Disease (2020) 11:757 https://doi.org/10.1038/s41419-020-02963-3 Cell Death & Disease ARTICLE Open Access EZH2 reduction is an essential mechanoresponse for the maintenance of super-enhancer polarization against compressive stress in human periodontal ligament stem cells Qian Li1,XiwenSun2,YunyiTang2, Yanan Qu2, Yanheng Zhou1 and Yu Zhang2 Abstract Despite the ubiquitous mechanical cues at both spatial and temporal dimensions, cell identities and functions are largely immune to the everchanging mechanical stimuli. To understand the molecular basis of this epigenetic stability, we interrogated compressive force-elicited transcriptomic changes in mesenchymal stem cells purified from human periodontal ligament (PDLSCs), and identified H3K27me3 and E2F signatures populated within upregulated and weakly downregulated genes, respectively. Consistently, expressions of several E2F family transcription factors and EZH2, as core methyltransferase for H3K27me3, decreased in response to mechanical stress, which were attributed to force-induced redistribution of RB from nucleoplasm to lamina. Importantly, although epigenomic analysis on H3K27me3 landscape only demonstrated correlating changes at one group of mechanoresponsive genes, we observed a genome-wide destabilization of super-enhancers along with aberrant EZH2 retention. These super- enhancers were tightly bounded by H3K27me3 domain on one side and exhibited attenuating H3K27ac deposition fl 1234567890():,; 1234567890():,; 1234567890():,; 1234567890():,; and attening H3K27ac peaks along with compensated EZH2 expression after force exposure, analogous to increased H3K27ac entropy or decreased H3K27ac polarization. Interference of force-induced EZH2 reduction could drive actin filaments dependent spatial overlap between EZH2 and super-enhancers and functionally compromise the multipotency of PDLSC following mechanical stress. These findings together unveil a specific contribution of EZH2 reduction for the maintenance of super-enhancer stability and cell identity in mechanoresponse.
    [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]
  • Rapa, a Bacterial Homolog of SWI2/SNF2, Stimulates RNA Polymerase Recycling in Transcription
    Downloaded from genesdev.cshlp.org on October 4, 2021 - Published by Cold Spring Harbor Laboratory Press RapA, a bacterial homolog of SWI2/SNF2, stimulates RNA polymerase recycling in transcription Maxim V. Sukhodolets, Julio E. Cabrera, Huijun Zhi, and Ding Jun Jin1 Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA We report that RapA, an Escherichia coli RNA polymerase (RNAP)-associated homolog of SWI2/SNF2, is capable of dramatic activation of RNA synthesis. The RapA-mediated transcriptional activation in vitro depends on supercoiled DNA and high salt concentrations, a condition that is likely to render the DNA superhelix tightly compacted. Moreover, RapA activates transcription by stimulating RNAP recycling. Mutational analyses indicate that the ATPase activity of RapA is essential for its function as a transcriptional activator, and a rapA null mutant exhibits a growth defect on nutrient plates containing high salt concentrations in vivo. Thus, RapA acts as a general transcription factor and an integral component of the transcription machinery. The mode of action of RapA in remodeling posttranscription or posttermination complexes is discussed. [Key Words: RapA; SWI2/SNF2 homolog; transcriptional activation; RNA polymerase recycling; remodeling posttranscription complexes] Received August 10, 2001; revised version accepted October 17, 2001. In Escherichia coli, core RNA polymerase (RNAP), Muchardt and Yaniv 1999). These proteins are capable of ␣ ␤␤Ј which consists of subunits 2 , is capable of transcrip- altering the configuration of naked DNA, an activity tion elongation and termination at simple terminators. that may be responsible for their chromatin/nucleosome On a sigma factor binding to core RNAP, the resulting remodeling function (Havas et al.
    [Show full text]