Inactivation of the PBRM1 Tumor Suppressor Gene Amplifies

Total Page:16

File Type:pdf, Size:1020Kb

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. Common examples Results and Discussion of the former are loss-of-function mutations of the PBRM1 and BAP1 We examined the protein levels of BAF180, BAP1, SETD2, and tumor suppressor genes, which occur in a mutually exclusive man- various SWI/SNF components in 16 ccRCC cell lines, together with PBRM1 ner in ccRCC and define biologically distinct subsets of ccRCC. HK-2 immortalized renal epithelial cells, using immunoblot assays. encodes the Polybromo- and BRG1-associated factors-containing All the 16 ccRCCs are pVHL-defective except for SLR20 and SLR21 complex (PBAF) chromatin remodeling complex component BRG1- (21).NPMwasusedasaloadingcontrol. BAF180 was undetectable associated factor 180 (BAF180). Here we identified ccRCC lines in five cell lines (A704, RCC4, SKRC20, SLR24, and SLR25) and whose ability to proliferate in vitro and in vivo is sensitive to barely detectable in another (Caki-2; Fig. 1A and SI Appendix,Fig. wild-type BAF180, but not a tumor-associated BAF180 mutant. Bio- PBRM1 chemical and functional studies linked growth suppression by S1). We also sequenced the cDNAs from these 16 ccRCC BAF180 to its ability to form a canonical PBAF complex containing lines and identified frame-shift, presumably loss-of-function, muta- B BRG1 that dampens the HIF transcriptional signature. tions in the six cell lines with low or undetectable BAF180 (Fig. 1 and C). In addition, we identified a small, in-frame, deletion in the PBRM1 B C kidney cancer | chromatin | PBAF | BAF180 | hypoxia cDNA in SLR26 cells (Fig. 1 and ). The A704 and Caki-2 PBRM1 mutations have been reported previously (10, 22). iallelic inactivation of the VHL tumor suppressor gene is the Four lines had diminished or undetectable levels of the ccRCC Busual initiating or truncal event in clear cell renal cell carci- suppressor protein BAP1 (SLR23, SLR26, UMRC2, and UMRC6), noma (ccRCC), which is the most common form of kidney cancer three had diminished or undetectable levels of the ccRCC (1–6). VHL loss, however, is not sufficient to cause ccRCC (4, 7–9). Other cooperating genetic events in ccRCC include copy Significance number gain of chromosome 5q, copy number loss of chromosome 14q, and intragenic mutations affecting chromatin regulatory genes Mutational inactivation of the VHL tumor suppressor gene is such as PBRM1, BAP1, ARID1A, SETD2, KDM5C,andKDM6A; the signature lesion in the most common form of kidney cancer PI3K pathway genes such as PTEN, PIK3CA, TSC1,andTORC1; and causes inappropriate accumulation of the HIF transcription and redox stress genes such as KEAP1 and NFE2L2 (1, 10–14). factor, which activates genes that normally facilitate adapta- PBRM1 is the gene that, after VHL, is most frequently mu- tion to hypoxia but, in the context of kidney cancer, also pro- tated in ccRCC. Interestingly, PBRM1 and VHL reside at chro- mote tumorigenesis. Additional mutational events are needed, mosomes 3p21 and 3p25, respectively. Accordingly, three genetic in conjunction with VHL loss, to cause kidney cancer. The most hits (intragenic cis mutations affecting PBRM1 and VHL, fol- common of these are inactivating mutations of the PBRM1 tumor lowed by loss of chromosome 3p) can cause biallelic loss of both suppressor gene, which encodes a component [BRG1-associated CELL BIOLOGY PBRM1 and VHL. A similar situation exists for BAP1 and factor 180 (BAF180)] of a multiprotein complex [Polybromo- and SETD2, which are also located on chromosome 3p21. PBRM1 BRG1-associated factors-containing complex (PBAF)] that regu- and BAP1 mutations are largely mutually exclusive in ccRCC and lates the positions of nucleosomes throughout the genome. We define biologically distinct ccRCC subtypes (12). describe here kidney cancer cell-based models for monitoring The PBRM1 gene product, BRG1-associated factor 180 (BAF180), BAF180 function and show that loss of BAF180 accentuates the is part of the multisubunit Polybromo- and BRG1-associated factors- transcriptional response to HIF. containing complex (PBAF) switch/sucrose nonfermentable (SWI/ SNF) chromatin remodeling complex (15–18). Mutations affecting Author contributions: W.G. and W.G.K. designed research; W.G. and T.X. performed re- search; T.X. performed MNase-Seq; W.G., W.L., X.S.L., and W.G.K. analyzed data; and W.G. SWI/SNF components have been linked to multiple forms of cancers and W.G.K. wrote the paper. – (15 18). For reasons that are not clear, however, there is a strong bias Reviewers: C.W.M.R., St. Jude Children’s Research Hospital; and A.S., Stowers Institute for to mutate specific SWI/SNF components in specific types of cancer. Medical Research. PBRM1 In this regard, inactivating mutations are most common in Conflict of interest statement: W.G.K. receives consulting income and equity from Peloton ccRCC, followed by cholangiocarcinoma (19, 20), but are otherwise Therapeutics, which is developing HIF2 inhibitors for treatment of kidney cancer. relatively uncommon in cancer. 1To whom correspondence should be addressed. Email: [email protected]. PBRM1 siRNA-mediated knockdown of wild-type was repor- This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. ted to increase the proliferation of multiple ccRCC cell lines in 1073/pnas.1619726114/-/DCSupplemental. www.pnas.org/cgi/doi/10.1073/pnas.1619726114 PNAS | January 31, 2017 | vol. 114 | no. 5 | 1027–1032 Downloaded by guest on September 29, 2021 could be immunoprecipitated with either an anti-FLAG antibody A or an anti-HA antibody from these cells (786-O-BKI), but not A498 HK-2 A704 Caki-2 RCC4 SKRC20SLR24SLR25769-P786-O SLR20SLR21SLR23SLR26UMRC2UMRC6UOK101 from parental 786-O cells, although recovery of the tagged BAF180 BAF180 was reproducibly higher with the anti-FLAG antibody than with the anti-HA antibody (Fig. 2B). In parallel, we infected BAP1 A704 cells, which lack detectable endogenous BAF180 (Fig. 1A), SETD2 with a lentivirus encoding FLAG-HA-tagged wild-type BAF180, a BAF250a tumor-associated BAF180 mutant with a frame-shift mutation (Q1298*) (10), or the empty vector (EV) (Fig. 2C). BAF200 We next performed preparative anti-FLAG immunoprecipi- BAF155 tations with these cell lines under stringent wash conditions. BAF47 The bound proteins were then eluted with a FLAG-peptide, BRG1 immunoprecipitated with an anti-HA antibody, eluted with an HA peptide, and either resolved by SDS/PAGE and detected BRM by silver staining (Fig. 2D)oridentifiedbymassspectrometry NPM (Fig. 2F and SI Appendix,TableS1). As expected, we observed silver-stained protein bands with predicted molecular weights B consistent with epitope-tagged BAF180 in immunoprecipitates Cell Protein Annotation cDNA Annotation prepared from the 786-O-BKI and A704-BAF180 cells and a A704 p.E572fs*16 c.1713-1714insTT slightly faster migrating band in the immunoprecipitates from Caki-2 p.E860fs*4 c. 2582-2585delTATA A704-BAF180 (Q1298*) cells (Fig. 2D). No such bands were RCC4 p.H897fs*3 c.2685-2686insA observed with the control samples prepared from parental 786-O SKRC20 p.F1012fs*1 c.3034delT cells and A704-EV cells. SLR24 p.H897fs*3 c.2685-2686insA Mass spectrometry analysis confirmed the recovery of BAF180 SLR25 p.Q477fs*17 c.1431_1443delGCAAGTTATGCAG itself, as well as other members of the canonical PBAF complex, SLR26 p.M1209_E1214delIMFYKKE c.3625_3642delATGTTCTACAAAAAAGAA including BAF170, BAF155, BAF200, BAF57, BAF60a, BAF45a, BAF60b, BAF53a, and BAF47 and the SWI/SNF-associated C proteins BCL7A and BCL7C. Surprisingly, we also recovered multiple peptides that are unique to the BRM DNA-dependent ATPase or unique to the BRG1 DNA-dependent ATPase, even A704 SLR25 SKRC20 RCC4/SLR24 SLR26 Caki-2 though only BRG1 is believed to participate in PBAF com- 1 1634 plexes (15–18). BAF180WT We next confirmed that exogenous wild-type BAF180, but not BAF180Δ2BD mutant (Q1298*) BAF180, coimmunoprecipitated with PBAF E BAF180Δ6BD components in A704 cells (Fig. 2 ). Consistent with this finding, the exogenous wild-type BAF180 in A704 cells, similar to the BAF180Q1298* endogenous wild-type BAF180 in 786-O cells and UMRC2 cells, was detected in a high-molecular-weight complex containing Bromodomain (BD): binding to acetylated histone other PBAF components after glycerol gradient centrifugation. In contrast, BAF180 (Q1298*) was not detected in this higher- Bromo-Adjacent Homology (BAH): putative protein-protein interaction order complex (Fig. 2 G–J).
Recommended publications
  • 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]
  • Transcriptome Analyses of Rhesus Monkey Pre-Implantation Embryos Reveal A
    Downloaded from genome.cshlp.org on September 23, 2021 - Published by Cold Spring Harbor Laboratory Press Transcriptome analyses of rhesus monkey pre-implantation embryos reveal a reduced capacity for DNA double strand break (DSB) repair in primate oocytes and early embryos Xinyi Wang 1,3,4,5*, Denghui Liu 2,4*, Dajian He 1,3,4,5, Shengbao Suo 2,4, Xian Xia 2,4, Xiechao He1,3,6, Jing-Dong J. Han2#, Ping Zheng1,3,6# Running title: reduced DNA DSB repair in monkey early embryos Affiliations: 1 State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China 2 Key Laboratory of Computational Biology, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center for Genetics and Developmental Biology, Chinese Academy of Sciences-Max Planck Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China 3 Yunnan Key Laboratory of Animal Reproduction, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China 4 University of Chinese Academy of Sciences, Beijing, China 5 Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China 6 Primate Research Center, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China * Xinyi Wang and Denghui Liu contributed equally to this work 1 Downloaded from genome.cshlp.org on September 23, 2021 - Published by Cold Spring Harbor Laboratory Press # Correspondence: Jing-Dong J. Han, Email: [email protected]; Ping Zheng, Email: [email protected] Key words: rhesus monkey, pre-implantation embryo, DNA damage 2 Downloaded from genome.cshlp.org on September 23, 2021 - Published by Cold Spring Harbor Laboratory Press ABSTRACT Pre-implantation embryogenesis encompasses several critical events including genome reprogramming, zygotic genome activation (ZGA) and cell fate commitment.
    [Show full text]
  • Mediator of DNA Damage Checkpoint 1 (MDC1) Is a Novel Estrogen Receptor Co-Regulator in Invasive 6 Lobular Carcinoma of the Breast 7 8 Evelyn K
    bioRxiv preprint doi: https://doi.org/10.1101/2020.12.16.423142; this version posted December 16, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC 4.0 International license. 1 Running Title: MDC1 co-regulates ER in ILC 2 3 Research article 4 5 Mediator of DNA damage checkpoint 1 (MDC1) is a novel estrogen receptor co-regulator in invasive 6 lobular carcinoma of the breast 7 8 Evelyn K. Bordeaux1+, Joseph L. Sottnik1+, Sanjana Mehrotra1, Sarah E. Ferrara2, Andrew E. Goodspeed2,3, James 9 C. Costello2,3, Matthew J. Sikora1 10 11 +EKB and JLS contributed equally to this project. 12 13 Affiliations 14 1Dept. of Pathology, University of Colorado Anschutz Medical Campus 15 2Biostatistics and Bioinformatics Shared Resource, University of Colorado Comprehensive Cancer Center 16 3Dept. of Pharmacology, University of Colorado Anschutz Medical Campus 17 18 Corresponding author 19 Matthew J. Sikora, PhD.; Mail Stop 8104, Research Complex 1 South, Room 5117, 12801 E. 17th Ave.; Aurora, 20 CO 80045. Tel: (303)724-4301; Fax: (303)724-3712; email: [email protected]. Twitter: 21 @mjsikora 22 23 Authors' contributions 24 MJS conceived of the project. MJS, EKB, and JLS designed and performed experiments. JLS developed models 25 for the project. EKB, JLS, SM, and AEG contributed to data analysis and interpretation. SEF, AEG, and JCC 26 developed and performed informatics analyses. MJS wrote the draft manuscript; all authors read and revised the 27 manuscript and have read and approved of this version of the manuscript.
    [Show full text]
  • Modeling Renal Cell Carcinoma in Mice: Bap1 and Pbrm1 Inactivation Drive Tumor Grade
    Published OnlineFirst May 4, 2017; DOI: 10.1158/2159-8290.CD-17-0292 RESEARCH ARTICLE Modeling Renal Cell Carcinoma in Mice: Bap1 and Pbrm1 Inactivation Drive Tumor Grade Yi-Feng Gu1,2, Shannon Cohn1,2, Alana Christie2, Tiffani McKenzie2,3, Nicholas Wolff1,2, Quyen N. Do4, Ananth J. Madhuranthakam4, Ivan Pedrosa2,4, Tao Wang2,5, Anwesha Dey6, Meinrad Busslinger7, Xian-Jin Xie2,8, Robert E. Hammer9, Renée M. McKay1,2, Payal Kapur2,3, and James Brugarolas1,2 Downloaded from cancerdiscovery.aacrjournals.org on September 26, 2021. © 2017 American Association for Cancer Research. 17-CD-17-0292_p900-917.indd 900 7/20/17 10:05 AM Published OnlineFirst May 4, 2017; DOI: 10.1158/2159-8290.CD-17-0292 ABSTRACT Clear cell renal cell carcinoma (ccRCC) is characterized by BAP1 and PBRM1 muta- tions, which are associated with tumors of different grade and prognosis. However, whether BAP1 and PBRM1 loss causes ccRCC and determines tumor grade is unclear. We conditionally targeted Bap1 and Pbrm1 (with Vhl ) in the mouse using several Cre drivers. Sglt2 and Villin proximal convoluted tubule drivers failed to cause tumorigenesis, challenging the conventional notion of ccRCC origins. In contrast, targeting with PAX8, a transcription factor frequently overexpressed in ccRCC, led to ccRCC of different grades. Bap1 -defi cient tumors were of high grade and showed greater mTORC1 activation than Pbrm1 -defi cient tumors, which exhibited longer latency. Disrupting one allele of the mTORC1 negative regulator, Tsc1 , in Pbrm1 -defi cient kidneys triggered higher grade ccRCC. This study establishes Bap1 and Pbrm1 as lineage-specifi c drivers of ccRCC and histologic grade, implicates mTORC1 as a tumor grade rheostat, and suggests that ccRCCs arise from Bowman capsule cells.
    [Show full text]
  • Genomic and Expression Profiling of Chromosome 17 in Breast Cancer Reveals Complex Patterns of Alterations and Novel Candidate Genes
    [CANCER RESEARCH 64, 6453–6460, September 15, 2004] Genomic and Expression Profiling of Chromosome 17 in Breast Cancer Reveals Complex Patterns of Alterations and Novel Candidate Genes Be´atrice Orsetti,1 Me´lanie Nugoli,1 Nathalie Cervera,1 Laurence Lasorsa,1 Paul Chuchana,1 Lisa Ursule,1 Catherine Nguyen,2 Richard Redon,3 Stanislas du Manoir,3 Carmen Rodriguez,1 and Charles Theillet1 1Ge´notypes et Phe´notypes Tumoraux, EMI229 INSERM/Universite´ Montpellier I, Montpellier, France; 2ERM 206 INSERM/Universite´ Aix-Marseille 2, Parc Scientifique de Luminy, Marseille cedex, France; and 3IGBMC, U596 INSERM/Universite´Louis Pasteur, Parc d’Innovation, Illkirch cedex, France ABSTRACT 17q12-q21 corresponding to the amplification of ERBB2 and collinear genes, and a large region at 17q23 (5, 6). A number of new candidate Chromosome 17 is severely rearranged in breast cancer. Whereas the oncogenes have been identified, among which GRB7 and TOP2A at short arm undergoes frequent losses, the long arm harbors complex 17q21 or RP6SKB1, TBX2, PPM1D, and MUL at 17q23 have drawn combinations of gains and losses. In this work we present a comprehensive study of quantitative anomalies at chromosome 17 by genomic array- most attention (6–10). Furthermore, DNA microarray studies have comparative genomic hybridization and of associated RNA expression revealed additional candidates, with some located outside current changes by cDNA arrays. We built a genomic array covering the entire regions of gains, thus suggesting the existence of additional amplicons chromosome at an average density of 1 clone per 0.5 Mb, and patterns of on 17q (8, 9). gains and losses were characterized in 30 breast cancer cell lines and 22 Our previous loss of heterozygosity mapping data pointed to the primary tumors.
    [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]
  • 1 Canonical BAF Complex in Regulatory T Cells 2 3 Chin
    bioRxiv preprint doi: https://doi.org/10.1101/2020.02.26.964981; this version posted February 27, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. 1 A genome-wide CRISPR screen reveals a role for the BRD9-containing non- 2 canonical BAF complex in regulatory T cells 3 4 Chin-San Loo1,3,#, Jovylyn Gatchalian2,#, Yuqiong Liang1, Mathias Leblanc1, Mingjun 5 Xie1, Josephine Ho2, Bhargav Venkatraghavan1, Diana C. Hargreaves2*, and Ye 6 Zheng1* 7 8 1. NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for 9 Biological Studies 10 2. Molecular and Cellular Biology Laboratory, Salk Institute for Biological Studies 11 3. Division of Biological Sciences, University of California, San Diego 12 # Co-first authors 13 * Co-corresponding authors 14 1 bioRxiv preprint doi: https://doi.org/10.1101/2020.02.26.964981; this version posted February 27, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. 15 Summary 16 Regulatory T cells (Tregs) play a pivotal role in suppressing auto-reactive T cells 17 and maintaining immune homeostasis. Treg development and function are 18 dependent on the transcription factor Foxp3. Here we performed a genome-wide 19 CRISPR/Cas9 knockout screen to identify the regulators of Foxp3 in mouse 20 primary Tregs. The results showed that Foxp3 regulators are highly enriched in 21 genes encoding SWI/SNF and SAGA complex subunits. Among the three 22 SWI/SNF-related complexes, the non-canonical or ncBAF (also called GBAF or 23 BRD9-containing BAF) complex promoted the expression of Foxp3, whereas the 24 PBAF complex repressed its expression.
    [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]
  • 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]
  • SMARCE1 Mutations in Pediatric Clear Cell Meningioma: Case Report
    PEDIATRICS CASE REPORT J Neurosurg Pediatr 16:296–300, 2015 SMARCE1 mutations in pediatric clear cell meningioma: case report Linton T. Evans, MD,1 Jack Van Hoff, MD,2,4,5 William F. Hickey, MD,3,4 Miriam J. Smith, PhD,6 D. Gareth Evans, MD,6 William G. Newman, MD, PhD,6 and David F. Bauer, MD1,2,4,5 1Section of Neurosurgery, 2Department of Pediatrics, and 3Department of Pathology, Dartmouth-Hitchcock Medical Center; 4Norris Cotton Cancer Center; 5Children’s Hospital at Dartmouth-Hitchcock, Lebanon, New Hampshire; and 6Manchester Centre for Genomic Medicine, Manchester Academic Health Sciences Centre (MAHSC), St. Mary’s Hospital, University of Manchester, United Kingdom Clear cell meningioma (CCM) is an uncommon variant of meningioma. The authors describe a case of a pediatric CCM localized to the lumbar spine. After resection, sequencing revealed an inactivating mutation in the SWI/SNF chromatin remodeling complex subunit SMARCE1, with loss of the second allele in the tumor. The authors present a literature review of this mutation that is associated with CCM and a family history of spine tumors. http://thejns.org/doi/abs/10.3171/2015.3.PEDS14417 KEY WORDS clear cell meningioma; SMARCE1; SWI/SNF; oncology LEAR cell meningioma (CCM) is a rare histopatho- of a SMARCE1 mutation and loss of heterozygosity in logical variant originally described in 1990. This a 3-year-old boy with an intraspinal CCM and a relative subtype is estimated to account for less than 1% of with a known spinal tumor. meningiomasC but is frequently difficult to treat. Despite a benign histological appearance, these tumors exhibit Case Report aggressive behavior with up to 60% recurring following resection.
    [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]
  • Insights Into Functional Connectivity in Mammalian Signal Transduction Pathways by Pairwise
    bioRxiv preprint doi: https://doi.org/10.1101/2019.12.30.891200; this version posted December 30, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. Insights into Functional Connectivity in Mammalian Signal Transduction Pathways by Pairwise Comparison of Protein Interaction Partners of Critical Signaling Hubs Chilakamarti V. Ramana * Department of Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH 03766, USA *Correspondence should be addressed: Chilakamarti V .Ramana, Telephone. (603)-738-2507, E-mail: [email protected] ORCID ID: /0000-0002-5153-8252 1 bioRxiv preprint doi: https://doi.org/10.1101/2019.12.30.891200; this version posted December 30, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. Abstract Growth factors and cytokines activate signal transduction pathways and regulate gene expression in eukaryotes. Intracellular domains of activated receptors recruit several protein kinases as well as transcription factors that serve as platforms or hubs for the assembly of multi-protein complexes. The signaling hubs involved in a related biologic function often share common interaction proteins and target genes. This functional connectivity suggests that a pairwise comparison of protein interaction partners of signaling hubs and network analysis of common partners and their expression analysis might lead to the identification of critical nodes in cellular signaling.
    [Show full text]