SNF5/INI1 Deficiency Redefines Chromatin Remodeling Complex Composition During Tumor Development

Total Page:16

File Type:pdf, Size:1020Kb

SNF5/INI1 Deficiency Redefines Chromatin Remodeling Complex Composition During Tumor Development Published OnlineFirst July 9, 2014; DOI: 10.1158/1541-7786.MCR-14-0005 Molecular Cancer Chromatin, Gene, and RNA Regulation Research SNF5/INI1 Deficiency Redefines Chromatin Remodeling Complex Composition during Tumor Development Darmood Wei1, Dennis Goldfarb2, Shujie Song3,4, Courtney Cannon4,5, Feng Yan4, Donastas Sakellariou- Thompson4, Michael Emanuele4,5, Michael B. Major4,6, Bernard E. Weissman4,7, and Yasumichi Kuwahara4,8 Abstract Malignant rhabdoid tumors (MRT), a pediatric cancer that most frequently appears in the kidney and brain, generally lack SNF5 (SMARCB1/INI1), a subunit of the SWI/SNF chromatin-remodeling complex. Recent studies have established that multiple SWI/SNF complexes exist due to the presence or absence of different complex members. Therefore, the effect of SNF5 loss upon SWI/SNF complex formation was investigated in human MRT cells. MRT cells and primary human tumors exhibited reduced levels of many complex proteins. Furthermore, reexpression of SNF5 increased SWI/SNF complex protein levels without concomitant increases in mRNA. Proteomic analysis, using mass spectrometry, of MRT cells before and after SNF5 reexpression indicated the recruitment of different components into the complex along with the expulsion of others. IP–Western blotting confirmed these results and demonstrated similar changes in other MRT cell lines. Finally, reduced expression of SNF5 in normal human fibroblasts led to altered levels of these same complex members. These data establish that SNF5 loss during MRT development alters the repertoire of available SWI/SNF complexes, generally disrupting those associated with cellular differentiation. These findings support a model where SNF5 inactivation blocks the conversion of growth-promoting SWI/SNF complexes to differentiation-inducing ones. Therefore, restoration of these complexes in tumors cells provides an attractive approach for the treatment of MRTs. Implications: SNF5 loss dramatically alters SWI/SNF complex composition and prevents formation of complexes required for cellular differentiation. Mol Cancer Res; 12(11); 1574–85. Ó2014 AACR. Introduction within a year of diagnosis (1). MRTs are characterized by the Malignant rhabdoid tumors (MRT) are a highly aggressive loss of SNF5/BAF47/INI1/SMARCB1, a core member of pediatric cancer commonly found in the kidneys (RTK) and the SWI/SNF complex, hereafter referred to as SNF5 (2). brain (AT/RT). These tumors have a median onset of 11 Interestingly, SNF5 loss does not affect genetic stability in months and 80% to 90% of children die from the disease MRT, but causes epigenetic instability (3). Thus, this con- sistent and specific molecular change, SNF5 inactivation, provides a unique model for understanding the role of 1Curriculum in Toxicology, University of North Carolina, Chapel Hill, North epigenetics in human tumor development. Numerous stud- Carolina. 2Department of Computer Science, University of North Carolina ies have shown that SNF5 loss affects multiple signaling at Chapel Hill, North Carolina. 3Oncology Center, ZhuJiang Hospital, Southern Medical University, Guangzhou, Guangdong, China. 4Lineberger pathways (4). However, little else is known about the etiology Comprehensive Cancer Center, University of North Carolina, Chapel Hill, of this cancer. North Carolina. 5Department of Pharmacology, University of North Car- The SWI/SNF complex has multiple variants and is olina, Chapel Hill, North Carolina. 6Department of Cell Biology and Phys- fi iology, University of North Carolina, Chapel Hill, North Carolina. 7Depart- classi ed by their mutually exclusive ATPase subunits, BRM ment of Pathology and Laboratory Medicine, University of North Carolina, and BRG1. BRG1-containing SWI/SNF complexes can be Chapel Hill, North Carolina. 8Department of Pediatrics, Kyoto Prefectural — University of Medicine, Kyoto, Japan. subdivided into two mutually exclusive groups BAF180/ PBRM1- and BAF250A/ARID1A-containing complexes. Note: Supplementary data for this article are available at Molecular Cancer Research Online (http://mcr.aacrjournals.org/). Although SNF5 appears in all the SWI/SNF complexes, little is known about the effects of SNF5 loss upon SWI/SNF Corresponding Authors: Bernard E. Weissman, Lineberger Comprehen- sive Cancer Center, Room 32-048, University of North Carolina, 450 West activity. One previous study by Doan and colleagues (5) Drive, Chapel Hill, NC 27599-7295. Phone: 919-966-7533; Fax: 919-966- showed that the SWI/SNF complex appeared intact in MRT 3015; E-mail: [email protected]; and Yasumichi Kuwahara, Depart- cell lines despite the loss of SNF5. They also demonstrated ment of Pediatrics, Kyoto Prefectural University of Medicine, 465 Kajii-cyo, Kawaramachi-douri Hirokoji-agaru, Kamigyo-ku, Kyoto-city, Kyoto 602- that BRG1-dependent genes did not show altered expression 8856, Japan. Phone: 81-75-251-5571; Fax: 81-75-252-1399; E-mail: in MRT cell lines (5). However, they did not address the [email protected] stoichiometry of the complex components in MRT cell lines doi: 10.1158/1541-7786.MCR-14-0005 or the interaction of the complex with chromatin. Other Ó2014 American Association for Cancer Research. studies have indicated that loss of a SWI/SNF complex 1574 Mol Cancer Res; 12(11) November 2014 Downloaded from mcr.aacrjournals.org on October 1, 2021. © 2014 American Association for Cancer Research. Published OnlineFirst July 9, 2014; DOI: 10.1158/1541-7786.MCR-14-0005 SNF5/INI1 Regulates SWI/SNF Complex Composition in MRTs member could affect the stability of other complex members Protein extracts and Western blotting in mammalian cells and in flies (6–12). Furthermore, a Isolation of nuclear protein and Western blotting were growing number of studies have identified multiple SWI/ carried out as described previously (16, 23). Western blot SNF complexes defined by the presence or absence of analyses for protein expression were carried out using the different components (4, 13–15). Importantly, these differ- antibodies listed in Supplementary Table S1. Densitometry ent complexes can promote either growth or differentiation was carried out using the Bio-Rad Image Lab 4.1 software. depending upon their composition (4, 13–15). To address whether SNF5 loss affects SWI/SNF complex RNA extraction and quantitative real-time reverse composition during MRT development, we examined the transcription PCR analysis effects of SNF5 expression in MRT cell lines. Our studies RNA was extracted using the RNeasy Mini Kit (Qiagen), demonstrate that reexpression of SNF5 in MRT cell lines and 1 mg was used for cDNA synthesis primed with random altered total protein levels of many components, whereas primers (Invitrogen). cDNA was analyzed using TaqMan knockdown of SNF5 in normal human fibroblasts showed (Applied Biosystems) quantitative real-time reverse tran- the opposite effect without altering mRNA expression. After scription PCR (qPCR) analysis, with b-actin as the reference SNF5 reexpression, many components were recruited into gene in each reaction. Reactions were performed on an ABI SWI/SNF complexes that are generally associated with 7900 HT sequence detection system (Applied Biosystems), ÀDDCt induction of differentiation. Therefore, SNF5 loss may and relative quantification was determined using the 2 promote MRT development by preventing a switch from method (Chai and colleagues, ref. 16). The TaqMan gene SWI/SNF complexes that promote proliferation to those expression assay primer/probe sets used in these studies are associated with differentiation. These studies emphasize the listed in Supplementary Table S2. need to resolve the scope and composition of SWI/SNF complexes among different tissues and may account for the Immunoprecipitation low incidence of these deadly childhood cancers. Cells were infected with adenovirus expressing either HA- tagged SNF5 (Ad-SNF5-HA) or empty vector (Ad-CMV). Materials and Methods Proteins were isolated from the infected cells using IP buffer Cell lines [50 mmol/L Tris, 400 mmol/L NaCl, 2 mmol/L EDTA, Our laboratory previously described the A204.1, D98OR, 10% Glycerol, 1% NP-40, 0.5% sodium deoxycholate, 0.1 and G401.6 cell lines (16–18). The MCF-7, A673, Jurkat, mmol/L PMSF, 4 mmol/L sodium fluoride, 40 nmol/L DAOY, and RD cell lines were obtained from the American sodium orthovanadate, and 1Â complete Mini protease Type Tissue Collection (ATCC). The TTC642 and inhibitor cocktail (Roche Diagnostics)]. The isolated protein TTC549 cell lines were kindly provided from Dr. Timothy was quantified and adjusted to 1 mg/mL. One milligram of Triche, Children's Hospital of Los Angeles. Dr. Peter protein from each sample was incubated with BRG-1 (A303- Houghton, Nationwide Childrens Hospital, kindly provid- 877A; Bethyl), or normal rabbit IgG (sc-2027; Santa Cruz ed the BT-12 AT/RT cell line. 293FT cells were kindly Biotechnology) rotating overnight at 4 C with 30 mLofa provided by Dr. Inder Verma, Salk Institute. All cell lines 50% slurry of protein A/G Sepharose beads. The beads were were cultured in RPMI-1640 plus 10% fetal bovine serum then washed three times with IP wash buffer (1Â PBS, 10% (Gibco; or Benchmark FBS, Gemini Biosciences) and were glycerol, and 1% Triton) and then suspended in 1Â used within 30 passages of their initial arrival to minimize the NuPAGE LDS Loading Buffer supplemented with 125 chances of cross-contamination. mmol/L DTT and boiled for 5 minutes. The supernatants were then run on 4% to 12% Bis-Tris polyacrylamide gel, Adenovirus infection transferred to polyvinylidene
Recommended publications
  • 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]
  • 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]
  • 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]
  • 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]
  • 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]
  • Downloading and Running the Code
    bioRxiv preprint doi: https://doi.org/10.1101/2020.04.30.069690; this version posted December 10, 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-ND 4.0 International license. Identification of disease treatment mechanisms through the multiscale interactome Camilo Ruiz,1;2 Marinka Zitnik,3 Jure Leskovec1;4;˚ 1 Computer Science Department, Stanford University, Stanford, CA 94305, USA 2 Bioengineering Department, Stanford University, Stanford, CA 94305, USA 3 Biomedical Informatics Department, Harvard University, Boston, MA 02115, USA 4 Chan Zuckerberg Biohub, San Francisco, CA 94158, USA ˚Corresponding author. Email: [email protected] 1 Most diseases disrupt multiple proteins, and drugs treat such diseases by restoring the func- 2 tions of the disrupted proteins. How drugs restore these functions, however, is often unknown 3 as a drug’s therapeutic effects are not limited only to the proteins that the drug directly tar- 4 gets. Here, we develop the multiscale interactome, a powerful approach to explain disease 5 treatment. We integrate disease-perturbed proteins, drug targets, and biological functions 6 into a multiscale interactome network, which contains 478,728 interactions between 1,661 7 drugs, 840 diseases, 17,660 human proteins, and 9,798 biological functions. We find that 8 a drug’s effectiveness can often be attributed to targeting proteins that are distinct from 9 disease-associated proteins but that affect the same biological functions. We develop a ran- 10 dom walk-based method that captures how drug effects propagate through a hierarchy of 11 biological functions and are coordinated by the protein-protein interaction network in which 12 drugs act.
    [Show full text]
  • Identification and Characterization of Novel Potentially Oncogenic Mutations in the Human Gene in a Breast Cancer Patient M
    Identification and characterization of novel potentially oncogenic mutations in the human gene in a breast cancer patient M. Ángeles Villaronga, Irene López-Mateo, Linn Markert, Enrique Espinosa, Juan Ángel Fresno Vara, Borja Belandia To cite this version: M. Ángeles Villaronga, Irene López-Mateo, Linn Markert, Enrique Espinosa, Juan Ángel Fresno Vara, et al.. Identification and characterization of novel potentially oncogenic mutations in the human gene in a breast cancer patient. Breast Cancer Research and Treatment, Springer Verlag, 2011, 128 (3), pp.891-898. 10.1007/s10549-011-1492-4. hal-00629080 HAL Id: hal-00629080 https://hal.archives-ouvertes.fr/hal-00629080 Submitted on 5 Oct 2011 HAL is a multi-disciplinary open access L’archive ouverte pluridisciplinaire HAL, est archive for the deposit and dissemination of sci- destinée au dépôt et à la diffusion de documents entific research documents, whether they are pub- scientifiques de niveau recherche, publiés ou non, lished or not. The documents may come from émanant des établissements d’enseignement et de teaching and research institutions in France or recherche français ou étrangers, des laboratoires abroad, or from public or private research centers. publics ou privés. Identification and characterization of novel potentially oncogenic mutations in the human BAF57 gene in a breast cancer patient 1* 1* 1 2 Mª Ángeles Villaronga , Irene López-Mateo , Linn Markert , Enrique Espinosa , Juan Ángel Fresno-Vara3 and Borja Belandia1 1Department of Cancer Biology, Instituto de Investigaciones Biomédicas Alberto Sols, CSIC-UAM, Arturo Duperier 4, 28029 Madrid, Spain 2Service of Oncology, IdiPAZ, Hospital Universitario La Paz, Paseo de la Castellana 261, 28046 Madrid, Spain 3Laboratory of Molecular Pathology & Oncology, IdiPAZ, Hospital Universitario La Paz, Paseo de la Castellana 261, 28046 Madrid, Spain *Mª Ángeles Villaronga and Irene López-Mateo contributed equally to this study and should be considered as co-first authors.
    [Show full text]
  • Epidyne®-FRET for Nucleosome Remodeling Assays
    Nucleosome Remodeling Assay by EpiDyne®-FRET EpiDyne®-FRET allows unprecedented access to disease-relevant ATP-dependent chromatin remodeling complexes FIGURE 3 SMARCA 2 SMARCA 4 EpiDyne®-FRET nucleosomes (20 nM) were incubated with Figure 3A Figure 3B chromatin remodeling enzyme (Panel 3A, SMARCA2; panel 3B, SMARCA4) at the indicated concentration in 4.0 3.5 the presence of ATP (2 mM). 3.5 Upon ATP addition, reactions 3.0 were immediately read in 3.0 an Envision Multi-label plate 2.5 reader. Data are presented as 2.5 the mean of the Cy3-Cy5 ratio 2.0 (N=2). 2.0 1.5 Cy3/Cy5 Ratio Cy3/Cy5 1.5 Ratio Cy3/Cy5 1.0 1.0 0 1 2 3 4 5 6 7 8 9 10 0 10 20 30 40 Time, Min Time, Min nM Enzyme nM Enzyme 28 14 7 3.5 0.0 12.50 6.25 3.13 1.56 0 ORDERING INFO Chromatin Remodeling Substrate, Fluorescent Readout Enzymes EpiDyne®-FRET Nucleosome Remodeling Assay Substrate SMARCA2 Chromatin Remodeling Enzyme Catalog No. 16-4201 (Human BRM) Pack Size: 50 μg Catalog No. 15-1015 Pack Size: 100 remodeling rxns Chromatin Remodeling Substrates, Non-Fluorescent Readout SMARCA4 Chromatin Remodeling Enzyme (Human BRG1) ST601-GATC1 ST601-GATC1, 50-N-66, Biotinylated Catalog No. 15-1014 Cat. No. 16-4101 Cat. No.: 16-4114 Pack Size: 100 remodeling rxns Pack Size: 50 μg Pack Size: 50 μg ACF Chromatin Remodeling Enzyme Complex ST601-GATC1, Biotinylated ST601-GATC1,2, 50-N-66, Biotinylated Catalog No. 15-1013 Cat.
    [Show full text]
  • Multiple Epigenetic Modifiers Induce Aggressive Viral Extinction
    View metadata, citation and similar papers at core.ac.uk brought to you by CORE provided by Elsevier - Publisher Connector Cell Stem Cell Article Multiple Epigenetic Modifiers Induce Aggressive Viral Extinction in Extraembryonic Endoderm Stem Cells Michael C. Golding,1,2,3,4 Liyue Zhang,3,5 and Mellissa R.W. Mann1,2,3,5,* 1Department of Obstetrics & Gynecology 2Department of Biochemistry University of Western Ontario, Schulich School of Medicine and Dentistry, London, Ontario N6A 4V2, Canada 3Children’s Health Research Institute, London, Ontario N6C 2V5, Canada 4Department of Veterinary Physiology, College of Veterinary Medicine, Texas A&M University, College Station, TX 77843, USA 5Lawson Health Research Institute, London, Ontario N6C 2V5, Canada *Correspondence: [email protected] DOI 10.1016/j.stem.2010.03.014 SUMMARY 5a (TRIM5a), zinc finger antiviral protein (ZAP), TRIM19/PML (promyelocytic leukemia), and the TRIM28-ZFP809 (zinc finger To prevent insertional mutagenesis arising from antiviral protein 809) complex all restrict retroviral tropism via retroviral reactivation, cells of embryonic origin direct biochemical interactions (Best et al., 1996; Kaiser et al., possess a unique capacity to silence retroviruses. 2007; Gao et al., 2002; Turelli et al., 2001; Wolf and Goff, 2009; Given the distinct modes of X chromosome inactiva- Rowe et al., 2010). tion between embryonic and extraembryonic line- Less well understood is the process of retroviral extinction, ages, we investigated paradigms of viral extinction. which is progressive silencing of proviral transcription that occurs over long-term cellular growth or differentiation (Cherry We show that trophectoderm stem cells do not et al., 2000; Laker et al., 1998).
    [Show full text]