Roles of the Snf2-Like Helicase Hells in Myc-Driven Gene Transcription

Total Page:16

File Type:pdf, Size:1020Kb

Roles of the Snf2-Like Helicase Hells in Myc-Driven Gene Transcription ROLES OF THE SNF2-LIKE HELICASE HELLS IN MYC-DRIVEN GENE TRANSCRIPTION by Jane A. Welch A dissertation submitted to Johns Hopkins University in conformity with the requirements for the degree of Doctor of Philosophy Baltimore, MD October 2017 © Jane A. Welch 2017 All Rights Reserved Abstract Helicase, lymphoid specific (HELLS) is a SNF2-like ATP-dependent chromatin remodeler that participates in transcriptional repression and activation in mammalian cells (von Eyss et al., 2012; Myant and Stancheva, 2008). HELLS has long been described as a proliferation-associated protein (Jarvis et al., 1996; Lee et al., 2000; Raabe et al., 2001), but its role in neoplastic diseases, which are characterized by cellular hyperproliferation, has not been widely investigated. This dissertation presents work that addresses how the expression and molecular function of HELLS may have pathologic relevance to human cancers. Through immunohistochemistry and RNA sequencing (RNA-seq), we found that HELLS is abundantly expressed in proliferative compartments of normal human tissues and broadly overexpressed in a number of cancers. We used chromatin immunoprecipitation followed by sequencing (ChIP-seq) to identify HELLS binding sites in human Burkitt lymphoma, glioblastoma multiforme, and small cell lung carcinoma cell line genomes and found that HELLS predominantly targets the promoters of transcribed genes, leading us to conclude that in human cancers, HELLS function is likely to be associated with transcriptional activation rather than repression. Expressed HELLS-bound genes are significantly enriched for targets of MYC, which has well-described oncogenic effects. Using ChIP- seq and co-immunoprecipitation (co-IP), we found that HELLS and MYC colocalize extensively at transcribed gene promoters and physically interact in human cells, which ii suggests that the proteins may functionally interact as well. We modeled partial loss of HELLS function in human cells through CRISPR/Cas9-mediated engineering of a hypomorphic HELLS allele and found that genes previously identified as HELLS targets exhibited altered expression levels. In human osteosarcoma cells with inducible MYC, we tested the effects of RNA interference (RNAi)-induced HELLS knockdown on MYC- driven transcription. While MYC target gene induction was not wholly impaired, a subset of MYC targets exhibited decreased expression. We conclude that HELLS is a bona fide MYC-interacting protein that may contribute to regulating the expression of some MYC targets, which could have implications for human tumor biology. Thesis advisor: Kathleen H. Burns, M.D., Ph.D. Thesis readers: Kathleen H. Burns, M.D., Ph.D. and Kirby D. Smith, Ph.D. iii Acknowledgements It is often said that it takes a village to raise a child. In my opinion, it also takes a village to raise a scientist at the doctoral level. I helmed my research projects, designed and conducted experiments with my own two hands, prepared my own presentations for meetings and conferences, and wrote this dissertation, but I could not have accomplished any of these things without the support of many others. I will begin by thanking the faculty and staff of the Predoctoral Training Program in Human Genetics housed in the McKusick-Nathans Institute of Genetic Medicine, especially Dr. David Valle and Sandy Muscelli. Earning acceptance into this program became my dream early on in my undergraduate studies. I am so very grateful that I was given the opportunity to study and train in this program, and I will always strive to be a positive reflection of it. I feel that receiving mentorship from my thesis advisor, Dr. Kathleen Burns, has made me one of the luckiest trainees at this institution. Kathy has enthusiastically supported me as I pursued both scientific questions of interest and professional development opportunities. She has always believed in me, even during the times when I found it impossible to believe in myself. She is the most gracious and considerate professional I have ever had the privilege to work with, and I will always look to her as a role model. I will be eternally grateful for the experience of being her mentee. iv During my graduate studies, I have also had the privilege to work with a number of talented and generous collaborators on my projects: Dr. Amy Duffield at the Johns Hopkins Hospital’s clinical IHC laboratory; Daniel Ardeljan, Dr. Kathy Burns, David Esopi, Dr. Michael Haffner, Dr. Chunhong Liu, Dr. Paul Schaughency, Alyza Skaist, Reema Sharma, Peilin Shen, Dr. Sarah Wheelan, William Wu, and Dr. Srinivasan Yegnasubramanian here at the Johns Hopkins University School of Medicine; Dr. Brian Altman, Dr. Annie Hsieh, and Dr. Chi Dang, who worked together at the University of Pennsylvania during our time of collaboration; and Dr. David Fenyö, Zuojian Tang, and Xuya Wang at the New York University School of Medicine. The work presented in this dissertation would not have been possible without their collaboration, and it is for that reason that I use “we” in presenting the rationales for experiments, hypotheses, findings, and conclusions throughout this tome. I would like to thank the members of my thesis committee– Dr. Kathy Burns, Dr. Elana Fertig, Dr. Haig Kazazian (my committee chairman), Dr. Kirby Smith, Dr. Sarah Wheelan, and Dr. Srinivasan Yegnasubramanian– for the thoughtful discussions, constructive criticisms, and steadfast encouragement they have provided over the past several years. I am especially grateful that Kathy and Kirby were willing to read this lengthy thesis and offer suggestions for improvement. I am very grateful to have worked alongside my colleagues, both former and current, in the Burns lab for all these years. They have always been generous with offering feedback on my experiments and analyses, and I have learned a great deal by working with them. In particular, I would like to thank Dan Ardeljan, Dr. Chunhong Liu, v Reema Sharma, Peilin Shen, and William Wu for collaborating with me on several of my projects. I am also incredibly grateful to Dr. Sarah Wheelan and the members of her lab– particularly Dr. Bracha Avigdor, Lauren Ciotti, Dr. Paul Schaughency, Alyza Skaist, and Heather Wick– for working closely with me during much of my time here at the School of Medicine. Sarah co-mentored me for many years, and it is entirely thanks to her and her lab members that I learned enough programming to complete the bioinformatics components of my thesis work. I would like to thank everyone at OMIM®, especially Joanna Amberger, Dr. Nara Sobreira, Matt Gross, and Carol Bocchini, for giving me the opportunity to write for their organization. I wholeheartedly believe that reviewing journal articles and drafting gene entries for OMIM® have made an important contribution to my education as a scientist. I feel that I would be remiss if I did not acknowledge Dr. Kerry Smith and Dr. Cheryl Ingram-Smith, who welcomed me into their laboratory as a completely inexperienced rising freshman at Clemson University. My dearest ambition was– and still is today– to find a way to help others through my work. Kerry and Cheryl taught me that research could provide a way to achieve that ambition, as our efforts to understand the bases of human diseases can ultimately lead us to ways to help people who are suffering. Without their guidance and support, I am not sure that I would ever have taken the path to becoming a biomedical scientist. As such, I will always be grateful to them both. Last, but unequivocally not least, I am incredibly grateful to my family and friends for their unwavering support throughout my lifetime and, in particular, during the vi long, challenging process of earning my Ph.D. My parents, Rob and Anne Welch, and their personal experiences with genetic disorders were really what sparked my interest in human genetics at a pretty early age. They have done everything they possibly could have to foster this interest of mine and to help me achieve my dream of becoming a geneticist, and I will always, always be thankful to them. My brother, David, has also been a substantial source of encouragement, along with my wonderful and loveable extended family: Jim and Pam Welch; Rosemary Nellist; Terry and Beth Dismukes; Matt, Sonia, Cannon, Chandler, and Saylor Dismukes; Jim, Ginnie, and Michelle Dismukes; Lauren and Jimmy Marshall; Bobby Dismukes and Jim White; Glenn, Adam, Jessica, and Dr. Joey Dismukes; and Brian and Will Dismukes. I have no doubt that my late grandparents, James and Laureign Welch and John and Doris Dismukes, would have been incredibly supportive and very proud of me for taking on the challenge of earning a Ph.D.– especially my Grandma Welch, who strongly advocated for women’s education. I would also like to offer heartfelt thanks to my dear friends; Randy, Becky, and Zach Hawkins; Rico, Sikithea, Skylar, and Sydney Zackery; and Nichola, Griffin, and Jillie Conze for all their love and support. In particular, Skylar, Sydney, and Nichola have brought me so much joy and encouragement, and they have made my hardest days so much easier to bear. Finally, I want to acknowledge my beloved collie, Toby, for always being able to make me smile– and for always reminding me of what matters most in life. vii Table of Contents Abstract ii Acknowledgements iv Table of Contents viii List of Figures xv List of Tables xx Chapter 1 Introduction 1 1.1 SWI/SNF chromatin remodelers and gene expression 1 1.2 Discovery and early characterization of HELLS 2 1.3 HELLS-associated phenotypes 4 1.4 Transcriptional regulation by
Recommended publications
  • The Title of the Article
    Mechanism-Anchored Profiling Derived from Epigenetic Networks Predicts Outcome in Acute Lymphoblastic Leukemia Xinan Yang, PhD1, Yong Huang, MD1, James L Chen, MD1, Jianming Xie, MSc2, Xiao Sun, PhD2, Yves A Lussier, MD1,3,4§ 1Center for Biomedical Informatics and Section of Genetic Medicine, Department of Medicine, The University of Chicago, Chicago, IL 60637 USA 2State Key Laboratory of Bioelectronics, Southeast University, 210096 Nanjing, P.R.China 3The University of Chicago Cancer Research Center, and The Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL 60637 USA 4The Institute for Genomics and Systems Biology, and the Computational Institute, The University of Chicago, Chicago, IL 60637 USA §Corresponding author Email addresses: XY: [email protected] YH: [email protected] JC: [email protected] JX: [email protected] XS: [email protected] YL: [email protected] - 1 - Abstract Background Current outcome predictors based on “molecular profiling” rely on gene lists selected without consideration for their molecular mechanisms. This study was designed to demonstrate that we could learn about genes related to a specific mechanism and further use this knowledge to predict outcome in patients – a paradigm shift towards accurate “mechanism-anchored profiling”. We propose a novel algorithm, PGnet, which predicts a tripartite mechanism-anchored network associated to epigenetic regulation consisting of phenotypes, genes and mechanisms. Genes termed as GEMs in this network meet all of the following criteria: (i) they are co-expressed with genes known to be involved in the biological mechanism of interest, (ii) they are also differentially expressed between distinct phenotypes relevant to the study, and (iii) as a biomodule, genes correlate with both the mechanism and the phenotype.
    [Show full text]
  • Coupling of Spliceosome Complexity to Intron Diversity
    bioRxiv preprint doi: https://doi.org/10.1101/2021.03.19.436190; this version posted March 20, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. Coupling of spliceosome complexity to intron diversity Jade Sales-Lee1, Daniela S. Perry1, Bradley A. Bowser2, Jolene K. Diedrich3, Beiduo Rao1, Irene Beusch1, John R. Yates III3, Scott W. Roy4,6, and Hiten D. Madhani1,6,7 1Dept. of Biochemistry and Biophysics University of California – San Francisco San Francisco, CA 94158 2Dept. of Molecular and Cellular Biology University of California - Merced Merced, CA 95343 3Department of Molecular Medicine The Scripps Research Institute, La Jolla, CA 92037 4Dept. of Biology San Francisco State University San Francisco, CA 94132 5Chan-Zuckerberg Biohub San Francisco, CA 94158 6Corresponding authors: [email protected], [email protected] 7Lead Contact 1 bioRxiv preprint doi: https://doi.org/10.1101/2021.03.19.436190; this version posted March 20, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. SUMMARY We determined that over 40 spliceosomal proteins are conserved between many fungal species and humans but were lost during the evolution of S. cerevisiae, an intron-poor yeast with unusually rigid splicing signals. We analyzed null mutations in a subset of these factors, most of which had not been investigated previously, in the intron-rich yeast Cryptococcus neoformans.
    [Show full text]
  • Supplementary Information Integrative Analyses of Splicing in the Aging Brain: Role in Susceptibility to Alzheimer’S Disease
    Supplementary Information Integrative analyses of splicing in the aging brain: role in susceptibility to Alzheimer’s Disease Contents 1. Supplementary Notes 1.1. Religious Orders Study and Memory and Aging Project 1.2. Mount Sinai Brain Bank Alzheimer’s Disease 1.3. CommonMind Consortium 1.4. Data Availability 2. Supplementary Tables 3. Supplementary Figures Note: Supplementary Tables are provided as separate Excel files. 1. Supplementary Notes 1.1. Religious Orders Study and Memory and Aging Project Gene expression data1. Gene expression data were generated using RNA- sequencing from Dorsolateral Prefrontal Cortex (DLPFC) of 540 individuals, at an average sequence depth of 90M reads. Detailed description of data generation and processing was previously described2 (Mostafavi, Gaiteri et al., under review). Samples were submitted to the Broad Institute’s Genomics Platform for transcriptome analysis following the dUTP protocol with Poly(A) selection developed by Levin and colleagues3. All samples were chosen to pass two initial quality filters: RNA integrity (RIN) score >5 and quantity threshold of 5 ug (and were selected from a larger set of 724 samples). Sequencing was performed on the Illumina HiSeq with 101bp paired-end reads and achieved coverage of 150M reads of the first 12 samples. These 12 samples will serve as a deep coverage reference and included 2 males and 2 females of nonimpaired, mild cognitive impaired, and Alzheimer's cases. The remaining samples were sequenced with target coverage of 50M reads; the mean coverage for the samples passing QC is 95 million reads (median 90 million reads). The libraries were constructed and pooled according to the RIN scores such that similar RIN scores would be pooled together.
    [Show full text]
  • Identification and Characterization of TPRKB Dependency in TP53 Deficient Cancers
    Identification and Characterization of TPRKB Dependency in TP53 Deficient Cancers. by Kelly Kennaley A dissertation submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy (Molecular and Cellular Pathology) in the University of Michigan 2019 Doctoral Committee: Associate Professor Zaneta Nikolovska-Coleska, Co-Chair Adjunct Associate Professor Scott A. Tomlins, Co-Chair Associate Professor Eric R. Fearon Associate Professor Alexey I. Nesvizhskii Kelly R. Kennaley [email protected] ORCID iD: 0000-0003-2439-9020 © Kelly R. Kennaley 2019 Acknowledgements I have immeasurable gratitude for the unwavering support and guidance I received throughout my dissertation. First and foremost, I would like to thank my thesis advisor and mentor Dr. Scott Tomlins for entrusting me with a challenging, interesting, and impactful project. He taught me how to drive a project forward through set-backs, ask the important questions, and always consider the impact of my work. I’m truly appreciative for his commitment to ensuring that I would get the most from my graduate education. I am also grateful to the many members of the Tomlins lab that made it the supportive, collaborative, and educational environment that it was. I would like to give special thanks to those I’ve worked closely with on this project, particularly Dr. Moloy Goswami for his mentorship, Lei Lucy Wang, Dr. Sumin Han, and undergraduate students Bhavneet Singh, Travis Weiss, and Myles Barlow. I am also grateful for the support of my thesis committee, Dr. Eric Fearon, Dr. Alexey Nesvizhskii, and my co-mentor Dr. Zaneta Nikolovska-Coleska, who have offered guidance and critical evaluation since project inception.
    [Show full text]
  • A Computational Approach for Defining a Signature of Β-Cell Golgi Stress in Diabetes Mellitus
    Page 1 of 781 Diabetes A Computational Approach for Defining a Signature of β-Cell Golgi Stress in Diabetes Mellitus Robert N. Bone1,6,7, Olufunmilola Oyebamiji2, Sayali Talware2, Sharmila Selvaraj2, Preethi Krishnan3,6, Farooq Syed1,6,7, Huanmei Wu2, Carmella Evans-Molina 1,3,4,5,6,7,8* Departments of 1Pediatrics, 3Medicine, 4Anatomy, Cell Biology & Physiology, 5Biochemistry & Molecular Biology, the 6Center for Diabetes & Metabolic Diseases, and the 7Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202; 2Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202; 8Roudebush VA Medical Center, Indianapolis, IN 46202. *Corresponding Author(s): Carmella Evans-Molina, MD, PhD ([email protected]) Indiana University School of Medicine, 635 Barnhill Drive, MS 2031A, Indianapolis, IN 46202, Telephone: (317) 274-4145, Fax (317) 274-4107 Running Title: Golgi Stress Response in Diabetes Word Count: 4358 Number of Figures: 6 Keywords: Golgi apparatus stress, Islets, β cell, Type 1 diabetes, Type 2 diabetes 1 Diabetes Publish Ahead of Print, published online August 20, 2020 Diabetes Page 2 of 781 ABSTRACT The Golgi apparatus (GA) is an important site of insulin processing and granule maturation, but whether GA organelle dysfunction and GA stress are present in the diabetic β-cell has not been tested. We utilized an informatics-based approach to develop a transcriptional signature of β-cell GA stress using existing RNA sequencing and microarray datasets generated using human islets from donors with diabetes and islets where type 1(T1D) and type 2 diabetes (T2D) had been modeled ex vivo. To narrow our results to GA-specific genes, we applied a filter set of 1,030 genes accepted as GA associated.
    [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]
  • 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]
  • The Capacity of Long-Term in Vitro Proliferation of Acute Myeloid
    The Capacity of Long-Term in Vitro Proliferation of Acute Myeloid Leukemia Cells Supported Only by Exogenous Cytokines Is Associated with a Patient Subset with Adverse Outcome Annette K. Brenner, Elise Aasebø, Maria Hernandez-Valladares, Frode Selheim, Frode Berven, Ida-Sofie Grønningsæter, Sushma Bartaula-Brevik and Øystein Bruserud Supplementary Material S2 of S31 Table S1. Detailed information about the 68 AML patients included in the study. # of blasts Viability Proliferation Cytokine Viable cells Change in ID Gender Age Etiology FAB Cytogenetics Mutations CD34 Colonies (109/L) (%) 48 h (cpm) secretion (106) 5 weeks phenotype 1 M 42 de novo 241 M2 normal Flt3 pos 31.0 3848 low 0.24 7 yes 2 M 82 MF 12.4 M2 t(9;22) wt pos 81.6 74,686 low 1.43 969 yes 3 F 49 CML/relapse 149 M2 complex n.d. pos 26.2 3472 low 0.08 n.d. no 4 M 33 de novo 62.0 M2 normal wt pos 67.5 6206 low 0.08 6.5 no 5 M 71 relapse 91.0 M4 normal NPM1 pos 63.5 21,331 low 0.17 n.d. yes 6 M 83 de novo 109 M1 n.d. wt pos 19.1 8764 low 1.65 693 no 7 F 77 MDS 26.4 M1 normal wt pos 89.4 53,799 high 3.43 2746 no 8 M 46 de novo 26.9 M1 normal NPM1 n.d. n.d. 3472 low 1.56 n.d. no 9 M 68 MF 50.8 M4 normal D835 pos 69.4 1640 low 0.08 n.d.
    [Show full text]
  • Plenary and Platform Abstracts
    American Society of Human Genetics 68th Annual Meeting PLENARY AND PLATFORM ABSTRACTS Abstract #'s Tuesday, October 16, 5:30-6:50 pm: 4. Featured Plenary Abstract Session I Hall C #1-#4 Wednesday, October 17, 9:00-10:00 am, Concurrent Platform Session A: 6. Variant Insights from Large Population Datasets Ballroom 20A #5-#8 7. GWAS in Combined Cancer Phenotypes Ballroom 20BC #9-#12 8. Genome-wide Epigenomics and Non-coding Variants Ballroom 20D #13-#16 9. Clonal Mosaicism in Cancer, Alzheimer's Disease, and Healthy Room 6A #17-#20 Tissue 10. Genetics of Behavioral Traits and Diseases Room 6B #21-#24 11. New Frontiers in Computational Genomics Room 6C #25-#28 12. Bone and Muscle: Identifying Causal Genes Room 6D #29-#32 13. Precision Medicine Initiatives: Outcomes and Lessons Learned Room 6E #33-#36 14. Environmental Exposures in Human Traits Room 6F #37-#40 Wednesday, October 17, 4:15-5:45 pm, Concurrent Platform Session B: 24. Variant Interpretation Practices and Resources Ballroom 20A #41-#46 25. Integrated Variant Analysis in Cancer Genomics Ballroom 20BC #47-#52 26. Gene Discovery and Functional Models of Neurological Disorders Ballroom 20D #53-#58 27. Whole Exome and Whole Genome Associations Room 6A #59-#64 28. Sequencing-based Diagnostics for Newborns and Infants Room 6B #65-#70 29. Omics Studies in Alzheimer's Disease Room 6C #71-#76 30. Cardiac, Valvular, and Vascular Disorders Room 6D #77-#82 31. Natural Selection and Human Phenotypes Room 6E #83-#88 32. Genetics of Cardiometabolic Traits Room 6F #89-#94 Wednesday, October 17, 6:00-7:00 pm, Concurrent Platform Session C: 33.
    [Show full text]
  • The Complex SNP and CNV Genetic Architecture of the Increased Risk of Congenital Heart Defects in Down Syndrome
    Downloaded from genome.cshlp.org on September 24, 2021 - Published by Cold Spring Harbor Laboratory Press Research The complex SNP and CNV genetic architecture of the increased risk of congenital heart defects in Down syndrome M. Reza Sailani,1,2 Periklis Makrythanasis,1 Armand Valsesia,3,4,5 Federico A. Santoni,1 Samuel Deutsch,1 Konstantin Popadin,1 Christelle Borel,1 Eugenia Migliavacca,1 Andrew J. Sharp,1,20 Genevieve Duriaux Sail,1 Emilie Falconnet,1 Kelly Rabionet,6,7,8 Clara Serra-Juhe´,7,9 Stefano Vicari,10 Daniela Laux,11 Yann Grattau,12 Guy Dembour,13 Andre Megarbane,12,14 Renaud Touraine,15 Samantha Stora,12 Sofia Kitsiou,16 Helena Fryssira,16 Chariklia Chatzisevastou-Loukidou,16 Emmanouel Kanavakis,16 Giuseppe Merla,17 Damien Bonnet,11 Luis A. Pe´rez-Jurado,7,9 Xavier Estivill,6,7,8 Jean M. Delabar,18 and Stylianos E. Antonarakis1,2,19,21 1–19[Author affiliations appear at the end of the paper.] Congenital heart defect (CHD) occurs in 40% of Down syndrome (DS) cases. While carrying three copies of chromosome 21 increases the risk for CHD, trisomy 21 itself is not sufficient to cause CHD. Thus, additional genetic variation and/or environmental factors could contribute to the CHD risk. Here we report genomic variations that in concert with trisomy 21, determine the risk for CHD in DS. This case-control GWAS includes 187 DS with CHD (AVSD = 69, ASD = 53, VSD = 65) as cases, and 151 DS without CHD as controls. Chromosome 21–specific association studies revealed rs2832616 and rs1943950 as CHD risk alleles (adjusted genotypic P-values <0.05).
    [Show full text]
  • Metastatic Adrenocortical Carcinoma Displays Higher Mutation Rate and Tumor Heterogeneity Than Primary Tumors
    ARTICLE DOI: 10.1038/s41467-018-06366-z OPEN Metastatic adrenocortical carcinoma displays higher mutation rate and tumor heterogeneity than primary tumors Sudheer Kumar Gara1, Justin Lack2, Lisa Zhang1, Emerson Harris1, Margaret Cam2 & Electron Kebebew1,3 Adrenocortical cancer (ACC) is a rare cancer with poor prognosis and high mortality due to metastatic disease. All reported genetic alterations have been in primary ACC, and it is 1234567890():,; unknown if there is molecular heterogeneity in ACC. Here, we report the genetic changes associated with metastatic ACC compared to primary ACCs and tumor heterogeneity. We performed whole-exome sequencing of 33 metastatic tumors. The overall mutation rate (per megabase) in metastatic tumors was 2.8-fold higher than primary ACC tumor samples. We found tumor heterogeneity among different metastatic sites in ACC and discovered recurrent mutations in several novel genes. We observed 37–57% overlap in genes that are mutated among different metastatic sites within the same patient. We also identified new therapeutic targets in recurrent and metastatic ACC not previously described in primary ACCs. 1 Endocrine Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA. 2 Center for Cancer Research, Collaborative Bioinformatics Resource, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA. 3 Department of Surgery and Stanford Cancer Institute, Stanford University, Stanford, CA 94305, USA. Correspondence and requests for materials should be addressed to E.K. (email: [email protected]) NATURE COMMUNICATIONS | (2018) 9:4172 | DOI: 10.1038/s41467-018-06366-z | www.nature.com/naturecommunications 1 ARTICLE NATURE COMMUNICATIONS | DOI: 10.1038/s41467-018-06366-z drenocortical carcinoma (ACC) is a rare malignancy with types including primary ACC from the TCGA to understand our A0.7–2 cases per million per year1,2.
    [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]