The Discovery of SWI/SNF Chromatin Remodeling Activity As a Novel and Targetable Dependency In

Total Page:16

File Type:pdf, Size:1020Kb

The Discovery of SWI/SNF Chromatin Remodeling Activity As a Novel and Targetable Dependency In Author Manuscript Published OnlineFirst on August 3, 2020; DOI: 10.1158/1535-7163.MCT-19-1013 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. 1 Title: The discovery of SWI/SNF chromatin remodeling activity as a novel and targetable dependency in 2 uveal melanoma 3 Authors: Florencia Rago1†, GiNell Elliott1†, Ailing Li1†, Kathleen Sprouffske2†, Grainne Kerr2†, Aurore 4 Desplat2†, Dorothee Abramowski2†, Julie T. Chen1†, Ali Farsidjani1†, Kay X. Xiang1†, Geoffrey Bushold1†, 5 Yun Feng1†, Matthew D. Shirley1†, Anka Bric1†, Anthony Vattay1†, Henrik Mӧbitz2◊, Katsumasa 6 Nakajima1◊, Christopher D. Adair1◊, Simon Mathieu1◊, Rukundo Ntaganda1◊, Troy Smith1◊, Julien P.N. 7 Papillon1◊, Audrey Kauffmann2†, David A. Ruddy1†, Hyo-eun C. Bhang1†, Deborah Castelletti2†, Zainab 8 Jagani1†* 9 † Oncology, ◊ Global Discovery Chemistry 10 1 Novartis Institutes for Biomedical Research, Cambridge, MA, USA 11 2 Novartis Institutes for Biomedical Research, Basel, Switzerland 12 * Corresponding Author: 13 Zainab Jagani 14 Novartis Institutes for Biomedical Research 15 250 Massachusetts Ave 16 600/03/3C-282 17 Cambridge, MA 02139 18 Phone: +16178714276 19 [email protected] 20 21 Running title: SWI/SNF complex is a novel target in uveal melanoma 22 Key words: BRM/SMARCA2, BRG1/SMARCA4, SWI/SNF, Uveal Melanoma, Chromatin Remodeling 23 Conflict of Interest Disclosure Statement: All authors performed the work herein as employees of the 24 Novartis Institutes for BioMedical Research. 25 Figures: 5 main, 6 supplemental 26 Tables: 4 supplemental 27 1 Downloaded from mct.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 3, 2020; DOI: 10.1158/1535-7163.MCT-19-1013 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. 1 Abstract 2 Uveal melanoma is a rare and aggressive cancer that originates in the eye. Currently, there are no 3 approved targeted therapies and very few effective treatments for this cancer. While activating mutations 4 in the G protein alpha subunits, GNAQ and GNA11, are key genetic drivers of the disease, few additional 5 drug targets have been identified. Recently, studies have identified context specific roles for the 6 mammalian SWI/SNF chromatin remodeling complexes (also known as BAF/PBAF) in various cancer 7 lineages. Here we find evidence that the SWI/SNF complex is essential through analysis of functional 8 genomics screens and further validation in a panel of uveal melanoma cell lines using both genetic tools 9 and small molecule inhibitors of SWI/SNF. In addition, we describe a functional relationship between the 10 SWI/SNF complex and the melanocyte lineage specific transcription factor MITF, suggesting that these 11 two factors cooperate to drive a transcriptional program essential for uveal melanoma cell survival. These 12 studies highlight a critical role for SWI/SNF in uveal melanoma, and demonstrate a novel path toward the 13 treatment of this cancer. 14 15 16 17 18 19 20 21 22 23 2 Downloaded from mct.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 3, 2020; DOI: 10.1158/1535-7163.MCT-19-1013 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. 1 Introduction 2 Uveal melanoma is a rare cancer that arises from the melanocytes in of the uvea. Although it originates in 3 melanocytes, the underlying mutations and unique immune environment of the eye distinguish this cancer 4 from the more common cutaneous melanoma. Uveal melanoma is mainly characterized by driver 5 mutations in GNAQ or GNA11 which lead to hyperactivation of the G proteins, resulting in downstream 6 mitogen-activated protein kinase (MAPK) pathway activation (1,2). In addition, chromosome 8q 7 amplification and BAP1 loss of heterozygosity (chromosome 3) or silencing are also commonly observed 8 and correlate with increased aggressiveness and poor prognosis (3). To date, targeted agents, primarily 9 against the MAPK pathway and its effectors, such as PKC, and even immunotherapy, have resulted in 10 limited clinical responses, and surgery and radiation remain the most common treatments, with overall 11 poor prognosis upon detection of metastatic disease (4). Due to the paucity of available treatments, 12 further studies to understand the biology of the disease, as well as elucidate novel therapeutic targets, 13 remain critical. 14 We set out to uncover novel dependencies in uveal melanoma through analysis of previously 15 performed unbiased pooled short hairpin RNA (shRNA) screens, and discovered an unanticipated role of 16 the SWI/SNF chromatin-remodeling complex in survival of uveal melanomas. The SWI/SNF complex 17 represents an important tumor suppressor in cancer, with approximately 20% of tumors harboring 18 mutations in one or more of its subunits (5,6). More recently however, BRG1/SMARCA4, the catalytic 19 subunit of the SWI/SNF complex, has been shown to be essential for cancer survival, such as in AML 20 where it works in concert with leukemic transcription factors to facilitate MYC expression (7,8). 21 Additionally, it has been demonstrated that SWI/SNF drives active enhancer state maintenance in a 22 lineage specific manner, further suggesting its context specific role in tumor maintenance (9-11). By this 23 logic, the SWI/SNF complex represents a targetable node in the complex lineage-specific transcriptional 24 machinery that may drive certain cancers. 25 In this study, we show that uveal melanoma models are dependent on the SWI/SNF catalytic 26 subunits BRG1 and BRM (also known as SMARCA4 and SMARCA2, respectively) by genetic 27 knockdown. We also demonstrate broad activity of small molecule inhibitors of BRM/BRG1 ATPase 3 Downloaded from mct.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 3, 2020; DOI: 10.1158/1535-7163.MCT-19-1013 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. 1 activity (12,13) across a panel of uveal melanoma cell lines. In an effort to dissect the mechanism of 2 SWI/SNF dependence, our work reveals a functional link between SWI/SNF and the transcription factor 3 Microphthalmia associated Transcription Factor (MITF). Together, our data identify SWI/SNF as a novel 4 target in uveal melanoma and reveal the therapeutic potential of applying small molecule inhibition of 5 SWI/SNF for the treatment of uveal melanoma. 6 7 Materials and Methods: 8 Cell lines and reagents 9 BRM011, BRM014 and BRM017 (synthesis described in (12,13)) stocks were dissolved at 10 mM in 10 DMSO. Doxycycline stock solution was made at 100 µg/ml in water. Shield 1 (Clontech) was dissolved at 11 0.5 mM in ethanol. 12 Cell lines were obtained from ATCC (MP41, MM28, MP46, MP65, MP38, and SW13), Sigma (Mel202), 13 Leiden University medical center (92.1 and OMM1), and Lonza (human epidermal melanocytes), and 14 cultured in manufacturer recommended media (92.1 and OMM1 were cultured in RPMI1640 + 10% FBS). 15 All parental lines were SNP profiled using the Fluidigm assay decribed previously (14) and tested 16 negative for Mycoplasma infection by qPCR (tests performed by Idexx Biosciences between March 2016 17 – February 2019). Cell lines used for no more than 1-3 months after thawing depending on doubling time. 18 Cell line engineering 19 shRNA cloning into pLKO based inducible vectors and cell line generation were described previously 20 (15). Cell line details annotated in Supplementary Table S3. 21 DD-BRG1 was assembled by adding Shield destabilization domain (DD) sequence (16) to the N-terminus 22 of BRG1 open reading frame (ORF). ACTL6A cDNA (Invitrogen) and DD-BRG1 were cloned into an in 23 house constitutive expression vector (lentiviral with EF1alpha promoter driving ORF expression). Flag- 24 HA-streptavidin (FHS) tagged MITF-M ORF was cloned into pLNCX-2 (Clontech). 4 Downloaded from mct.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 3, 2020; DOI: 10.1158/1535-7163.MCT-19-1013 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. 1 Growth, viability and apoptosis assays 2 To measure cell growth, cell lines were plated in 96 well plates (92.1 5000 cells, OMM1 2500, MP41 2500), 3 then imaged on an IncuCyte (4x objective) and analyzed using Incucyte Zoom 2016B software. 4 For viability assays, cells were plated in 384 well plates (Corning 3765) (92.1 500 cells, OMM1 500, MP41 5 500, Mel202 1500, MP46 1000, MM28 4000, MP38 3000, MP65 1500, SW13 1000, melanocytes 3000). 6 Plates were dosed with an 11 point, 3-fold serial dilution using the Echo550 (Labcyte). After 5 days, Cell 7 Titer Glo (Promega) was added and luminescence measured (PHERAstar, BMG Labtech). Growth 8 inhibition values were calculated as described previously (17). Normalized data were fit using the three 9 parameters nonlinear regression function in GraphPad Prism 7. Absolute AC50s (AAC50) reported as 10 concentrations of compound where curve fit crosses 0.5. 11 For caspase activity, cells were plated and treated as above (92.1 1500 cells, OMM1 1500, MP41 1500, 12 Mel202 3000, MP46 3000, MM28 5000, MP38 5000, MP65 3500, SW13 3000). After 48 h, Caspase 3/7 13 Glo (Promega) was added and luminescence measured. Normalization was performed relative to 14 untreated wells and plotted as fold activity. 15 Single point viability and caspase activity assays were performed in indicated 92.1 shRNA lines treated 16 with 250 nM BRM011, BRM014, BRM017 or 100 ng/ml doxycycline for 3 days and then assayed as 17 above and analyzed by normalizing relative to untreated wells.
Recommended publications
  • Germline Mutations Affecting Gα11 in Hypoparathyroidism
    T h e new england journal o f medicine of a meta-analysis of studies of the effect of re- terone levels were increased. Although long- duced dietary salt on the incidence of cardiovas- term, modest reductions in salt intake result in cular events and death.1 The authors of the Co- small, physiologic increases in plasma renin ac- chrane report wrote that there was “no strong tivity,3 the preponderance of evidence suggests evidence of benefit.” In a summary statement, we that a reduced salt intake is associated with a wrote that this particular Cochrane analysis con- decreased risk of cardiovascular events and cluded that reducing dietary salt intake did not death. Furthermore, it is worth remembering decrease the risk of death or cardiovascular dis- that diuretics remain one of the most effective ease. Stigler et al. suggest that “indeterminate antihypertensive therapies, and their beneficial results,” rather than no significant effect, would effect on cardiovascular disease is well docu- be a more appropriate interpretation of the analy- mented.4 Nevertheless, as we suggested, in sis. Both interpretations may be correct. Although terms of safety, the lower limit of salt consump- it may not be possible to reject the null hypothe- tion has not been clearly defined. sis with certainty (i.e., no effect of reduced salt), Theodore A. Kotchen, M.D. the analysis should have been powered to detect Allen W. Cowley, Jr., Ph.D. a clinically meaningful difference, and the con- Medical College of Wisconsin clusion of no effect provides more guidance than Milwaukee, WI “indeterminate results” to clinical decision mak- [email protected] ers.
    [Show full text]
  • ACTL6A Promotes the Proliferation of Esophageal Squamous Cell Carcinoma Cells and Correlates with Poor Clinical Outcomes
    OncoTargets and Therapy Dovepress open access to scientific and medical research Open Access Full Text Article ORIGINAL RESEARCH ACTL6A Promotes the Proliferation of Esophageal Squamous Cell Carcinoma Cells and Correlates with Poor Clinical Outcomes This article was published in the following Dove Press journal: OncoTargets and Therapy Rui-zhe Li1 Background: ACTL6A, a regulatory subunit of ATP-dependent chromatin-remodeling Yun-yun Li1,2 complexes SWI/SNF, has been identified as a central oncogenic driver in many tumor types. Hui Qin1 Materials and Methods: We used immunohistochemistry (IHC) to detect ACTL6A Shan-shan Li1 expression in esophageal squamous cell carcinoma (ESCC) tissues. Then, the effect of ACTL6A on proliferation and DNA synthesis was explored by using cell counting kit 8 1 Department of Pathology, School of (CCK8) and EdU retention assays. The potential oncogenic mechanism of ACTL6A in Basic Medical Sciences, Zhengzhou University and First Affiliated Hospital of ESCC cells was also analyzed by flow cytometry and Western blotting. We further estab­ Zhengzhou University, Zhengzhou, lished an ESCC xenograft mouse model to validate the in vitro results. Henan 450000, People’s Republic of China; 2Department of Stomatology, First Results: ACTL6A expression, localized in cancer cell nuclei, was markedly higher in ESCC Affiliated Hospital of Zhengzhou tissues than in the corresponding noncancerous tissues (P<0.001) and was positively asso­ University, Zhengzhou, Henan 450000, ciated with tumor size, histological differentiation, T stage and tumor-node-metastasis People’s Republic of China (TNM) stage. Kaplan–Meier analysis revealed that high ACTL6A expression was signifi­ cantly associated with poor overall survival (OS) (P = 0.008, HR= 2.562, 95% CI: 1.241–­ 5.289), and decision curve analysis (DCA) demonstrated that ACTL6A could increase the clinical prognostic efficiency of the original clinical prediction model.
    [Show full text]
  • C/EBP Creates Elite Cells for Ipsc Reprogramming by Upregulating
    ARTICLES C/EBPα creates elite cells for iPSC reprogramming by upregulating Klf4 and increasing the levels of Lsd1 and Brd4 Bruno Di Stefano1,2,8,9,10, Samuel Collombet3,8, Janus Schou Jakobsen4,5,6,8, Michael Wierer7, Jose Luis Sardina1,2, Andreas Lackner1,2,9, Ralph Stadhouders1,2, Carolina Segura-Morales1,2, Mirko Francesconi1,2, Francesco Limone1,2, Matthias Mann7, Bo Porse4,5,6, Denis Thieffry3 and Thomas Graf1,2,10 Reprogramming somatic cells into induced pluripotent stem cells (iPSCs) is typically inefficient and has been explained by elite-cell and stochastic models. We recently reported that B cells exposed to a pulse of C/EBPα (Bα0 cells) behave as elite cells, in that they can be rapidly and efficiently reprogrammed into iPSCs by the Yamanaka factors OSKM. Here we show that C/EBPα post-transcriptionally increases the abundance of several hundred proteins, including Lsd1, Hdac1, Brd4, Med1 and Cdk9, components of chromatin-modifying complexes present at super-enhancers. Lsd1 was found to be required for B cell gene silencing and Brd4 for the activation of the pluripotency program. C/EBPα also promotes chromatin accessibility in pluripotent cells and upregulates Klf4 by binding to two haematopoietic enhancers. Bα0 cells share many properties with granulocyte/macrophage progenitors, naturally occurring elite cells that are obligate targets for leukaemic transformation, whose formation strictly requires C/EBPα. The ability to reprogram somatic into pluripotent cells has revolu- complex process, where multiple players synergistically establish new tionized stem cell research with major implications for almost all transcriptional networks and remove epigenetic barriers14. Among the fields of modern biology.
    [Show full text]
  • Ubiquitylome Profiling of Parkin-Null Brain Reveals Dysregulation Of
    Neurobiology of Disease 127 (2019) 114–130 Contents lists available at ScienceDirect Neurobiology of Disease journal homepage: www.elsevier.com/locate/ynbdi Ubiquitylome profiling of Parkin-null brain reveals dysregulation of calcium T homeostasis factors ATP1A2, Hippocalcin and GNA11, reflected by altered firing of noradrenergic neurons Key J.a,1, Mueller A.K.b,1, Gispert S.a, Matschke L.b, Wittig I.c, Corti O.d,e,f,g, Münch C.h, ⁎ ⁎ Decher N.b, , Auburger G.a, a Exp. Neurology, Goethe University Medical School, 60590 Frankfurt am Main, Germany b Institute for Physiology and Pathophysiology, Vegetative Physiology and Marburg Center for Mind, Brain and Behavior - MCMBB; Clinic for Neurology, Philipps-University Marburg, 35037 Marburg, Germany c Functional Proteomics, SFB 815 Core Unit, Goethe University Medical School, 60590 Frankfurt am Main, Germany d Institut du Cerveau et de la Moelle épinière, ICM, Paris, F-75013, France e Inserm, U1127, Paris, F-75013, France f CNRS, UMR 7225, Paris, F-75013, France g Sorbonne Universités, Paris, F-75013, France h Institute of Biochemistry II, Goethe University Medical School, 60590 Frankfurt am Main, Germany ARTICLE INFO ABSTRACT Keywords: Parkinson's disease (PD) is the second most frequent neurodegenerative disorder in the old population. Among Parkinson's disease its monogenic variants, a frequent cause is a mutation in the Parkin gene (Prkn). Deficient function of Parkin Mitochondria triggers ubiquitous mitochondrial dysfunction and inflammation in the brain, but it remains unclear howse- Parkin lective neural circuits become vulnerable and finally undergo atrophy. Ubiquitin We attempted to go beyond previous work, mostly done in peripheral tumor cells, which identified protein Calcium targets of Parkin activity, an ubiquitin E3 ligase.
    [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]
  • Swi/Snf Chromatin Remodeling/Tumor Suppressor Complex Establishes Nucleosome Occupancy at Target Promoters
    Swi/Snf chromatin remodeling/tumor suppressor complex establishes nucleosome occupancy at target promoters Michael Y. Tolstorukova,b,1,2, Courtney G. Sansamc,d,e,1, Ping Luc,d,e,1, Edward C. Koellhofferc,d,e, Katherine C. Helmingc,d,e, Burak H. Alvera, Erik J. Tillmanc,d,e, Julia A. Evansc,d,e, Boris G. Wilsonc,d,e, Peter J. Parka,b,3, and Charles W. M. Robertsc,d,e,3 aCenter for Biomedical Informatics, Harvard Medical School, Boston, MA 02115; bDivision of Genetics, Brigham and Women’s Hospital, Boston, MA 02115; cDepartment of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02115; dDivision of Hematology/Oncology, Boston Children’s Hospital, Boston, MA 02115; and eDepartment of Pediatrics, Harvard Medical School, Boston, MA 02115 Edited by Mark Groudine, Fred Hutchinson Cancer Research Center, Seattle, WA, and approved May 2, 2013 (received for review February 6, 2013) Precise nucleosome-positioning patterns at promoters are thought Brg1 haploinsufficient mice are tumor prone, establishing these to be crucial for faithful transcriptional regulation. However, the subunits of the complex as bona fide tumor suppressors (1, 12–17). mechanisms by which these patterns are established, are dynam- It is noteworthy that recent exome sequencing of 35 human SNF5- ically maintained, and subsequently contribute to transcriptional deficient rhabdoid tumors identified a remarkably low rate of control are poorly understood. The switch/sucrose non-fermentable mutations, with loss of SNF5 being essentially the sole recurrent event (18). Indeed, in two of the cancers, there were no other chromatin remodeling complex, also known as the Brg1 associated fi factors complex, is a master developmental regulator and tumor identi ed mutations.
    [Show full text]
  • Gene Silencing Associated with SWI/SNF Complex Loss During NSCLC Development
    Published OnlineFirst January 20, 2014; DOI: 10.1158/1541-7786.MCR-13-0427 Molecular Cancer Genomics Research Gene Silencing Associated with SWI/SNF Complex Loss during NSCLC Development Shujie Song1,2, Vonn Walter2, Mehmet Karaca3, Ying Li2, Christopher S. Bartlett4, Dominic J. Smiraglia7, Daniel Serber5, Christopher D. Sproul3, Christoph Plass8, Jiren Zhang1, D. Neil Hayes2,6, Yanfang Zheng1, and Bernard E. Weissman2,3 Abstract The SWI/SNF chromatin-remodeling complex regulates gene expression and alters chromatin structures in an ATP-dependent manner. Recent sequencing efforts have shown mutations in BRG1 (SMARCA4), one of two mutually exclusive ATPase subunits in the complex, in a significant number of human lung tumor cell lines and primary non–small cell lung carcinoma (NSCLC) clinical specimens. To determine how BRG1 loss fuels tumor progression in NSCLC, molecular profiling was performed after restoration of BRG1 expression or treatment with a histone deacetylase inhibitor or a DNA methyltransferase (DNMT) inhibitor in a BRG1-deficient NSCLC cells. Importantly, validation studies from multiple cell lines revealed that BRG1 reexpression led to substantial changes in the expression of CDH1, CDH3, EHF, and RRAD that commonly undergo silencing by other epigenetic mechanisms during NSCLC development. Furthermore, treatment with DNMT inhibitors did not restore expression of these transcripts, indicating that this common mechanism of gene silencing did not account for their loss of expression. Collectively, BRG1 loss is an important mechanism for the epigenetic silencing of target genes during NSCLC development. Implications: Inactivation of the SWI/SNF complex provides a novel mechanism to induce gene silencing during NSCLC development. Mol Cancer Res; 12(4); 1–11.
    [Show full text]
  • 140503 IPF Signatures Supplement Withfigs Thorax
    Supplementary material for Heterogeneous gene expression signatures correspond to distinct lung pathologies and biomarkers of disease severity in idiopathic pulmonary fibrosis Daryle J. DePianto1*, Sanjay Chandriani1⌘*, Alexander R. Abbas1, Guiquan Jia1, Elsa N. N’Diaye1, Patrick Caplazi1, Steven E. Kauder1, Sabyasachi Biswas1, Satyajit K. Karnik1#, Connie Ha1, Zora Modrusan1, Michael A. Matthay2, Jasleen Kukreja3, Harold R. Collard2, Jackson G. Egen1, Paul J. Wolters2§, and Joseph R. Arron1§ 1Genentech Research and Early Development, South San Francisco, CA 2Department of Medicine, University of California, San Francisco, CA 3Department of Surgery, University of California, San Francisco, CA ⌘Current address: Novartis Institutes for Biomedical Research, Emeryville, CA. #Current address: Gilead Sciences, Foster City, CA. *DJD and SC contributed equally to this manuscript §PJW and JRA co-directed this project Address correspondence to Paul J. Wolters, MD University of California, San Francisco Department of Medicine Box 0111 San Francisco, CA 94143-0111 [email protected] or Joseph R. Arron, MD, PhD Genentech, Inc. MS 231C 1 DNA Way South San Francisco, CA 94080 [email protected] 1 METHODS Human lung tissue samples Tissues were obtained at UCSF from clinical samples from IPF patients at the time of biopsy or lung transplantation. All patients were seen at UCSF and the diagnosis of IPF was established through multidisciplinary review of clinical, radiological, and pathological data according to criteria established by the consensus classification of the American Thoracic Society (ATS) and European Respiratory Society (ERS), Japanese Respiratory Society (JRS), and the Latin American Thoracic Association (ALAT) (ref. 5 in main text). Non-diseased normal lung tissues were procured from lungs not used by the Northern California Transplant Donor Network.
    [Show full text]
  • Expressed Gene Fusions As Frequent Drivers of Poor Outcomes in Hormone Receptor–Positive Breast Cancer
    Published OnlineFirst December 14, 2017; DOI: 10.1158/2159-8290.CD-17-0535 RESEARCH ARTICLE Expressed Gene Fusions as Frequent Drivers of Poor Outcomes in Hormone Receptor–Positive Breast Cancer Karina J. Matissek1,2, Maristela L. Onozato3, Sheng Sun1,2, Zongli Zheng2,3,4, Andrew Schultz1, Jesse Lee3, Kristofer Patel1, Piiha-Lotta Jerevall2,3, Srinivas Vinod Saladi1,2, Allison Macleay3, Mehrad Tavallai1,2, Tanja Badovinac-Crnjevic5, Carlos Barrios6, Nuran Beşe7, Arlene Chan8, Yanin Chavarri-Guerra9, Marcio Debiasi6, Elif Demirdögen10, Ünal Egeli10, Sahsuvar Gökgöz10, Henry Gomez11, Pedro Liedke6, Ismet Tasdelen10, Sahsine Tolunay10, Gustavo Werutsky6, Jessica St. Louis1, Nora Horick12, Dianne M. Finkelstein2,12, Long Phi Le2,3, Aditya Bardia1,2, Paul E. Goss1,2, Dennis C. Sgroi2,3, A. John Iafrate2,3, and Leif W. Ellisen1,2 ABSTRACT We sought to uncover genetic drivers of hormone receptor–positive (HR+) breast cancer, using a targeted next-generation sequencing approach for detecting expressed gene rearrangements without prior knowledge of the fusion partners. We identified inter- genic fusions involving driver genes, including PIK3CA, AKT3, RAF1, and ESR1, in 14% (24/173) of unselected patients with advanced HR+ breast cancer. FISH confirmed the corresponding chromo- somal rearrangements in both primary and metastatic tumors. Expression of novel kinase fusions in nontransformed cells deregulates phosphoprotein signaling, cell proliferation, and survival in three- dimensional culture, whereas expression in HR+ breast cancer models modulates estrogen-dependent growth and confers hormonal therapy resistance in vitro and in vivo. Strikingly, shorter overall survival was observed in patients with rearrangement-positive versus rearrangement-negative tumors. Cor- respondingly, fusions were uncommon (<5%) among 300 patients presenting with primary HR+ breast cancer.
    [Show full text]
  • Inactivation of the PBRM1 Tumor Suppressor Gene Amplifies
    Inactivation of the PBRM1 tumor suppressor gene − − amplifies the HIF-response in VHL / clear cell renal carcinoma Wenhua Gaoa, Wei Lib,c, Tengfei Xiaoa,b,c, Xiaole Shirley Liub,c, and William G. Kaelin Jr.a,d,1 aDepartment of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115; bCenter for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215; cDepartment of Biostatistics and Computational Biology, Dana-Farber Cancer Institute and Harvard T.H. Chan School of Public Health, Boston, MA 02115; and dHoward Hughes Medical Institute, Chevy Chase, MD 20815 Contributed by William G. Kaelin, Jr., December 1, 2016 (sent for review October 31, 2016; reviewed by Charles W. M. Roberts and Ali Shilatifard) Most clear cell renal carcinomas (ccRCCs) are initiated by somatic monolayer culture and in soft agar (10). These effects were not, inactivation of the VHL tumor suppressor gene. The VHL gene prod- however, proven to be on-target, and were not interrogated in uct, pVHL, is the substrate recognition unit of an ubiquitin ligase vivo. As a step toward understanding the role of BAF180 in that targets the HIF transcription factor for proteasomal degrada- ccRCC, we asked whether BAF180 participates in the canonical tion; inappropriate expression of HIF target genes drives renal car- PBAF complex in ccRCC cell lines and whether loss of BAF180 cinogenesis. Loss of pVHL is not sufficient, however, to cause ccRCC. measurably alters ccRCC behavior in cell culture and in mice. Additional cooperating genetic events, including intragenic muta- tions and copy number alterations, are required.
    [Show full text]
  • Mosaic Activating Mutations in GNA11 and GNAQ Are Associated with Phakomatosis Pigmentovascularis and Extensive Dermal Melanocytosis Anna C
    ORIGINAL ARTICLE Mosaic Activating Mutations in GNA11 and GNAQ Are Associated with Phakomatosis Pigmentovascularis and Extensive Dermal Melanocytosis Anna C. Thomas1,18, Zhiqiang Zeng2,18, Jean-Baptiste Rivie`re3,18, Ryan O’Shaughnessy4, Lara Al-Olabi1, Judith St.-Onge3, David J. Atherton5,He´le`ne Aubert6, Lorea Bagazgoitia7, Se´bastien Barbarot6, Emmanuelle Bourrat8,9, Christine Chiaverini10, W. Kling Chong11, Yannis Duffourd3, Mary Glover5, Leopold Groesser12, Smail Hadj-Rabia13, Henning Hamm14, Rudolf Happle15, Imran Mushtaq16, Jean-Philippe Lacour10, Regula Waelchli5, Marion Wobser14, Pierre Vabres3,17,19, E. Elizabeth Patton2,19 and Veronica A. Kinsler1,5,19 Common birthmarks can be an indicator of underlying genetic disease but are often overlooked. Mongolian blue spots (dermal melanocytosis) are usually localized and transient, but they can be extensive, permanent, and associated with extracutaneous abnormalities. Co-occurrence with vascular birthmarks defines a subtype of phakomatosis pigmentovascularis, a group of syndromes associated with neurovascular, ophthalmological, overgrowth, and malignant complications. Here, we discover that extensive dermal melanocytosis and pha- komatosis pigmentovascularis are associated with activating mutations in GNA11 and GNAQ, genes that encode Ga subunits of heterotrimeric G proteins. The mutations were detected at very low levels in affected tissues but were undetectable in the blood, indicating that these conditions are postzygotic mosaic disorders. R183C Q209L In vitro expression of mutant GNA11 and GNA11 in human cell lines demonstrated activation of the downstream p38 MAPK signaling pathway and the p38, JNK, and ERK pathways, respectively. Transgenic R183C mosaic zebrafish models expressing mutant GNA11 under promoter mitfa developed extensive dermal melanocytosis recapitulating the human phenotype. Phakomatosis pigmentovascularis and extensive dermal melanocytosis are therefore diagnoses in the group of mosaic heterotrimeric G-protein disorders, joining McCune-Albright and Sturge-Weber syndromes.
    [Show full text]
  • Rats and Axolotls Share a Common Molecular Signature After Spinal Cord Injury Enriched in Collagen-1
    Rats and axolotls share a common molecular signature after spinal cord injury enriched in collagen-1 Athanasios Didangelos1, Katalin Bartus1, Jure Tica1, Bernd Roschitzki2, Elizabeth J. Bradbury1 1Wolfson CARD King’s College London, United Kingdom. 2Centre for functional Genomics, ETH Zurich, Switzerland. Running title: spinal cord injury in rats and axolotls Correspondence: A Didangelos: [email protected] SUPPLEMENTAL FIGURES AND LEGENDS Supplemental Fig. 1: Rat 7 days microarray differentially regulated transcripts. A-B: Protein-protein interaction networks of upregulated (A) and downregulated (B) transcripts identified by microarray gene expression profiling of rat SCI (4 sham versus 4 injured spinal cord samples) 7 days post-injury. Microarray expression data and experimental information is publicly available online (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE45006) and is also summarised in Supplemental Table 1. Protein-protein interaction networks were performed in StringDB using the full range of protein interaction scores (0.15 – 0.99) to capture maximum evidence of proteins’ interactions. Networks were then further analysed for betweeness centrality and gene ontology (GO) annotations (BinGO) in Cytoscape. Node colour indicates betweeness centrality while edge colour and thickness indicate interaction score based on predicted functional links between nodes (green: low values; red: high values). C-D: The top 10 upregulated (C) or downregulated (D) transcripts sorted by betweeness centrality score in protein-protein interaction networks shown in A & B. E-F: Biological process GO analysis was performed on networks of upregulated and downregulated genes using BinGO in Cytoscape. Graphs indicate the 20 most significant GO categories and the number of genes in each category.
    [Show full text]