Pan-Cancer Multi-Omics Analysis and Orthogonal Experimental Assessment of Epigenetic Driver Genes

Total Page:16

File Type:pdf, Size:1020Kb

Pan-Cancer Multi-Omics Analysis and Orthogonal Experimental Assessment of Epigenetic Driver Genes Downloaded from genome.cshlp.org on October 9, 2021 - Published by Cold Spring Harbor Laboratory Press Resource Pan-cancer multi-omics analysis and orthogonal experimental assessment of epigenetic driver genes Andrea Halaburkova, Vincent Cahais, Alexei Novoloaca, Mariana Gomes da Silva Araujo, Rita Khoueiry,1 Akram Ghantous,1 and Zdenko Herceg1 Epigenetics Group, International Agency for Research on Cancer (IARC), 69008 Lyon, France The recent identification of recurrently mutated epigenetic regulator genes (ERGs) supports their critical role in tumorigen- esis. We conducted a pan-cancer analysis integrating (epi)genome, transcriptome, and DNA methylome alterations in a cu- rated list of 426 ERGs across 33 cancer types, comprising 10,845 tumor and 730 normal tissues. We found that, in addition to mutations, copy number alterations in ERGs were more frequent than previously anticipated and tightly linked to ex- pression aberrations. Novel bioinformatics approaches, integrating the strengths of various driver prediction and multi- omics algorithms, and an orthogonal in vitro screen (CRISPR-Cas9) targeting all ERGs revealed genes with driver roles with- in and across malignancies and shared driver mechanisms operating across multiple cancer types and hallmarks. This is the largest and most comprehensive analysis thus far; it is also the first experimental effort to specifically identify ERG drivers (epidrivers) and characterize their deregulation and functional impact in oncogenic processes. [Supplemental material is available for this article.] Although it has long been known that human cancers harbor both gression, potentially acting as oncogenes or tumor suppressors genetic and epigenetic changes, with an intricate interplay be- (Plass et al. 2013; Vogelstein et al. 2013). Although several distinct tween the two mechanisms underpinning the hallmarks of cancer definitions of “driver gene” exist in the literature (Sawan et al. (Hanahan and Weinberg 2011), it is only with the fruition of large- 2008; Vogelstein et al. 2013), we define “driver genes” as those scale international sequencing efforts that major enigmas of the genes that, when deregulated (through somatic mutations, copy cancer (epi)genome have started to be solved (Jones et al. 2016; number variations, or aberrant expression), assume primary im- Ng et al. 2018). One of the most remarkable findings of the inter- portance in tumor development such as conferring a selective national high-resolution cancer genome sequencing efforts, spear- growth advantage, immortalization, and invasiveness. This defini- headed by The Cancer Genome Atlas (TCGA), is the high tion relies on inference models for driver prediction and functional frequency of genetic alterations in the genes encoding proteins data (based on the impact of the gene on cellular processes) com- that directly regulate the epigenome (referred to here as epigenetic pared to other methods that are mostly based on statistical models regulator genes [ERGs]) (Gonzalez-Perez et al. 2013; Plass et al. (largely driven by the mutation frequency of a gene) (Parmigiani 2013; Shen and Laird 2013; Timp and Feinberg 2013; Vogelstein et al. 2009; Meyerson et al. 2010; Lahouel et al. 2020). In line et al. 2013; Yang et al. 2015). This high rate of ERG genetic dereg- with this physiological definition, we refer to those ERGs that ulation constitutes a “genetic smoking gun,” indicating that epige- make a net contribution to tumorigenesis as “epigenetic driver netic mechanisms lie at the very heart of cancer biology. These genes” (henceforth called “epidrivers”). Our definition is different discoveries have sparked a debate on the role of ERG deregulation from that used by other investigators (Vogelstein et al. 2013), who (either through mutational or nongenetic events) in ERG expres- define epidrivers as the genes (not necessarily among ERGs) that sion and in the mechanisms underlying tumorigenesis and epige- are aberrantly expressed through changes in DNA methylation nome alterations that are rampant in virtually all human and chromatin modifications and confer a selective growth malignancies (Plass et al. 2013; Timp and Feinberg 2013). We advantage. also still lack a systematic understanding of the functional impor- The products of ERGs are involved in processes such as DNA tance of ERG disruption in tumor development and progression, as methylation, histone modification, chromatin remodeling, and well as its impact on cancer cell phenotype. other chromatin-based modifications, and many ERGs may have ERGs are a group of more than 400 coding genes in the hu- both histone and nonhistone substrates. All of these processes, in man genome, most of which encode enzymes that add (“writers”), turn, are involved in the proper control of not only gene expression modify/revert (“editors”), or recognize (“readers”) epigenetic mod- programs, required for the establishment and maintenance of cell ifications (Plass et al. 2013; Vogelstein et al. 2013) controlling a identity and function, but also DNA repair, recombination, and ge- range of critical cellular processes. Based on the observation that nome integrity (Murr et al. 2006; Bell et al. 2011). Because common many ERGs are frequently disrupted across different malignancies, cancers represent the final outcome of a multistep process, epi- they are candidates to be drivers of cancer development and pro- driver-based disruption of cellular processes may not only assume a primary role at different stages of tumorigenesis but also constitute critical mechanisms underpinning cancer cell plasticity and 1These authors contributed equally to this work. Corresponding author: [email protected] Article published online before print. Article, supplemental material, and publi- © 2020 Halaburkova et al. This article, published in Genome Research, is avail- cation date are at http://www.genome.org/cgi/doi/10.1101/gr.268292.120. able under a Creative Commons License (Attribution 4.0 International), as de- Freely available online through the Genome Research Open Access option. scribed at http://creativecommons.org/licenses/by/4.0/. 30:1–16 Published by Cold Spring Harbor Laboratory Press; ISSN 1088-9051/20; www.genome.org Genome Research 1 www.genome.org Downloaded from genome.cshlp.org on October 9, 2021 - Published by Cold Spring Harbor Laboratory Press Halaburkova et al. emergence of cancer resilience. Here, we conducted a systematic and terations, exceeding 40% of samples in LGG and LUSC, respective- comprehensive pan-cancer investigation of (epi)genetics- and tran- ly. Several ERGs had the highest mutation frequency repeatedly in scriptome-based deregulation of all ERGs using in silico data cura- many cancer types, namely the KMT2C/D family (seven cancers), tion in clinical samples and characterization of the driver ARID1A (five cancers), BAP1 (three cancers), and ATRX (three can- potential by different computational tools. We also developed and cers) (Fig. 2E,G; Supplemental Table S3). A similar observation was tested a conceptual framework for experimental identification and made for deep CNAs in ERGs, namely BOP1 (four cancers), ATAD2 functional characterization of the mechanistically important epi- (four cancers), MECOM (three cancers), and PHF20L1 (three can- drivers that reshape the epigenome and contribute to cancer pheno- cers) (Fig. 2F,H). A larger percentage of ERG alterations was also ob- types. This framework builds on the latest knowledge of the cancer served when both deep and shallow CNAs were included (Fig. 2C, (epi)genome and genomic databases and includes powerful new ex- D; Supplemental Fig. S1C,D). Among the top ERGs altered by deep perimental models including state-of-the-art genome-editing CNAs, the majority showed amplifications, with the exception of screens, phenotyping, and functional genomics. HR, PHF11, and SETB2, which were commonly deleted in many cancer types (Fig. 2H). Frequently amplified ERGs often co-oc- curred in the same tumor sample in many cancer types; in partic- Results ular, the aforementioned pan-cancer recurrent genes BOP1, A four-stage strategy was used to identify and characterize ERGs ATAD2, and PHF20L1 highly co-occurred (Supplemental Fig. with cancer driver potential (Fig. 1A). We assembled a comprehen- S3A). These co-occurrences remained prominent even when the sive compendium of ERG genes by literature mining and manual analysis was focused only on deep amplifications/deletions across curation, resulting in a list of 426 genes coding for histone modifi- tumors (Supplemental Fig. S3B,C). Moreover, the family of TDRs ers, DNA methylation regulators, chromatin remodelers, helicases, (TDRKH, TDRD10, and TDRD5) highly co-occurred together. and other epigenetic entities (Fig. 1B; Supplemental Tables S1, S2). Generally little overlap was observed between the genes with a To identify the candidate epidrivers across different cancer types, high frequency of SNAs and those with a high frequency of – we first used comprehensive in silico data mining of genetic and CNAs (except for a few ERGs) (Fig. 2E H). RNA expression alterations of ERGs using data from TCGA (Fig. When ERGs were stratified by functional groups, similar total 1A). Collectively, the data encompassed 33 different cancer types proportions of genetic alterations were seen among ERG classes from 25 tissue types, with sequencing information from 10,845 tu- (Fig. 2I). DNA methylation writers and editors were characterized mor samples and 730 normal tissues, including a total of by a prominent proportion
Recommended publications
  • A Genetic Screening Identifies a Component of the SWI/SNF Complex, Arid1b As a Senescence Regulator
    A genetic screening identifies a component of the SWI/SNF complex, Arid1b as a senescence regulator Sadaf Khan A thesis submitted to Imperial College London for the degree of Doctor in Philosophy MRC Clinical Sciences Centre Imperial College London, School of Medicine July 2013 Statement of originality All experiments included in this thesis were performed by myself unless otherwise stated. Copyright Declaration The copyright of this thesis rests with the author and is made available under a Creative Commons Attribution Non-Commercial No Derivatives license. Researchers are free to copy, distribute or transmit the thesis on the condition that they attribute it, that they do not use it for commercial purposes and that they do not alter, transform or build upon it. For any reuse or redistribution, researchers must make clear to others the license terms of this work. 2 Abstract Senescence is an important tumour suppressor mechanism, which prevents the proliferation of stressed or damaged cells. The use of RNA interference to identify genes with a role in senescence is an important tool in the discovery of novel cancer genes. In this work, a protocol was established for conducting bypass of senescence screenings, using shRNA libraries together with next-generation sequencing. Using this approach, the SWI/SNF subunit Arid1b was identified as a regulator of cellular lifespan in MEFs. SWI/SNF is a large multi-subunit complex that remodels chromatin. Mutations in SWI/SNF proteins are frequently associated with cancer, suggesting that SWI/SNF components are tumour suppressors. Here the role of ARID1B during senescence was investigated. Depletion of ARID1B extends the proliferative capacity of primary mouse and human fibroblasts.
    [Show full text]
  • A Novel Ve-Zinc Nger-Related-Gene Signature Related to Tumor Immune
    A novel ve-zinc nger-related-gene signature related to tumor immune microenvironment allows for treatment stratication and predicts prognosis in clear cell renal cell carcinoma patients Feilong Zhang Beijing Chaoyang Hospital Jiyue Wu Beijing Chaoyang Hospital Jiandong Zhang Beijing Chaoyang Hospital Peng Cao Beijing Chaoyang Hospital Zejia Sun Beijing Chaoyang Hospital Wei Wang ( [email protected] ) Beijing Chaoyang Hospital https://orcid.org/0000-0003-2642-3338 Research Keywords: Posted Date: February 23rd, 2021 DOI: https://doi.org/10.21203/rs.3.rs-207677/v1 License: This work is licensed under a Creative Commons Attribution 4.0 International License. Read Full License Page 1/25 Abstract Background Clear cell renal cell carcinoma (ccRCC) is one of the most prevalent renal malignant tumors, which survival rate and quality of life of ccRCC patients are not satisfactory. Therefore, identication of prognostic biomarkers of ccRCC patients will contribute to early and accurate clinical intervention and treatment, and then improve their prognosis. Methods We downloaded the original expression data of mRNAs from The Cancer Genome Atlas database and the zinc nger(ZNF)-related genes (ZRGs) from UniProt online database. Differentially expressed ZRGs (DE- ZRGs) was screened from tumor and adjacent nontumor tissues and functional enrichment analysis was conducted out. A ve-ZRG signature were constructed by univariate Cox regression, least absolute shrinkage and selection operator and multivariate Cox regression. Furthermore, we screened out independent prognosis-related factors to build a nomogram by univariate and multivariate Cox regression. Potential biological pathways of ve ZRGs were analyzed by Gene Set Enrichment Analysis (GSEA). Then, we further quantitatively analyze immune inltration and evaluate tumor microenvironment by single sample GSEA.
    [Show full text]
  • Molecular Profile of Tumor-Specific CD8+ T Cell Hypofunction in a Transplantable Murine Cancer Model
    Downloaded from http://www.jimmunol.org/ by guest on September 25, 2021 T + is online at: average * The Journal of Immunology , 34 of which you can access for free at: 2016; 197:1477-1488; Prepublished online 1 July from submission to initial decision 4 weeks from acceptance to publication 2016; doi: 10.4049/jimmunol.1600589 http://www.jimmunol.org/content/197/4/1477 Molecular Profile of Tumor-Specific CD8 Cell Hypofunction in a Transplantable Murine Cancer Model Katherine A. Waugh, Sonia M. Leach, Brandon L. Moore, Tullia C. Bruno, Jonathan D. Buhrman and Jill E. Slansky J Immunol cites 95 articles Submit online. Every submission reviewed by practicing scientists ? is published twice each month by Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts http://jimmunol.org/subscription Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html http://www.jimmunol.org/content/suppl/2016/07/01/jimmunol.160058 9.DCSupplemental This article http://www.jimmunol.org/content/197/4/1477.full#ref-list-1 Information about subscribing to The JI No Triage! Fast Publication! Rapid Reviews! 30 days* Why • • • Material References Permissions Email Alerts Subscription Supplementary The Journal of Immunology The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2016 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. This information is current as of September 25, 2021. The Journal of Immunology Molecular Profile of Tumor-Specific CD8+ T Cell Hypofunction in a Transplantable Murine Cancer Model Katherine A.
    [Show full text]
  • Table S1. 49 Histone Variants Were Identified with High Sequence Coverage Through LC-MS/MS Analysis Electronic Supplementary
    Electronic Supplementary Material (ESI) for Analytical Methods. This journal is © The Royal Society of Chemistry 2020 Table S1. 49 histone variants were identified with high sequence coverage through LC-MS/MS analysis Sequence Uniprot IDs Protein Name Protein Description Coverage Ratio E2+/E2- RSD P07305 H10_HUMAN 67.5% Histone H1.0 OS=Homo sapiens GN=H1F0 PE=1 SV=3 4.85 23.3% Histone H1.1 OS=Homo sapiens GN=HIST1H1A PE=1 Q02539 H11_HUMAN 74.4% SV=3 0.35 92.6% Histone H1.2 OS=Homo sapiens GN=HIST1H1C PE=1 P16403 H12_HUMAN 67.1% SV=2 0.73 80.6% Histone H1.3 OS=Homo sapiens GN=HIST1H1D PE=1 P16402 H13_HUMAN 63.8% SV=2 0.75 77.7% Histone H1.4 OS=Homo sapiens GN=HIST1H1E PE=1 P10412 H14_HUMAN 69.0% SV=2 0.70 80.3% Histone H1.5 OS=Homo sapiens GN=HIST1H1B PE=1 P16401 H15_HUMAN 79.6% SV=3 0.29 98.3% Testis-specific H1 histone OS=Homo sapiens GN=H1FNT Q75WM6 H1FNT_HUMAN 7.8% \ \ PE=2 SV=3 Histone H1oo OS=Homo sapiens GN=H1FOO PE=2 Q8IZA3 H1FOO_HUMAN 5.2% \ \ SV=1 Histone H1t OS=Homo sapiens GN=HIST1H1T PE=2 P22492 H1T_HUMAN 31.4% SV=4 1.42 65.0% Q92522 H1X_HUMAN 82.6% Histone H1x OS=Homo sapiens GN=H1FX PE=1 SV=1 1.15 33.2% Histone H2A type 1 OS=Homo sapiens GN=HIST1H2AG P0C0S8 H2A1_HUMAN 99.2% PE=1 SV=2 0.57 26.8% Q96QV6 H2A1A_HUMAN 58.0% Histone H2A type 1-A OS=Homo sapiens 0.90 11.2% GN=HIST1H2AA PE=1 SV=3 Histone H2A type 1-B/E OS=Homo sapiens P04908 H2A1B_HUMAN 99.2% GN=HIST1H2AB PE=1 SV=2 0.92 30.2% Histone H2A type 1-C OS=Homo sapiens Q93077 H2A1C_HUMAN 100.0% GN=HIST1H2AC PE=1 SV=3 0.76 27.6% Histone H2A type 1-D OS=Homo sapiens P20671
    [Show full text]
  • UNIVERSITY of CALIFORNIA, SAN DIEGO Functional Analysis of Sall4
    UNIVERSITY OF CALIFORNIA, SAN DIEGO Functional analysis of Sall4 in modulating embryonic stem cell fate A dissertation submitted in partial satisfaction of the requirements for the degree Doctor of Philosophy in Molecular Pathology by Pei Jen A. Lee Committee in charge: Professor Steven Briggs, Chair Professor Geoff Rosenfeld, Co-Chair Professor Alexander Hoffmann Professor Randall Johnson Professor Mark Mercola 2009 Copyright Pei Jen A. Lee, 2009 All rights reserved. The dissertation of Pei Jen A. Lee is approved, and it is acceptable in quality and form for publication on microfilm and electronically: ______________________________________________________________ ______________________________________________________________ ______________________________________________________________ ______________________________________________________________ Co-Chair ______________________________________________________________ Chair University of California, San Diego 2009 iii Dedicated to my parents, my brother ,and my husband for their love and support iv Table of Contents Signature Page……………………………………………………………………….…iii Dedication…...…………………………………………………………………………..iv Table of Contents……………………………………………………………………….v List of Figures…………………………………………………………………………...vi List of Tables………………………………………………….………………………...ix Curriculum vitae…………………………………………………………………………x Acknowledgement………………………………………………….……….……..…...xi Abstract………………………………………………………………..…………….....xiii Chapter 1 Introduction ..…………………………………………………………………………….1 Chapter 2 Materials and Methods……………………………………………………………..…12
    [Show full text]
  • Modes of Interaction of KMT2 Histone H3 Lysine 4 Methyltransferase/COMPASS Complexes with Chromatin
    cells Review Modes of Interaction of KMT2 Histone H3 Lysine 4 Methyltransferase/COMPASS Complexes with Chromatin Agnieszka Bochy ´nska,Juliane Lüscher-Firzlaff and Bernhard Lüscher * ID Institute of Biochemistry and Molecular Biology, Medical School, RWTH Aachen University, Pauwelsstrasse 30, 52057 Aachen, Germany; [email protected] (A.B.); jluescher-fi[email protected] (J.L.-F.) * Correspondence: [email protected]; Tel.: +49-241-8088850; Fax: +49-241-8082427 Received: 18 January 2018; Accepted: 27 February 2018; Published: 2 March 2018 Abstract: Regulation of gene expression is achieved by sequence-specific transcriptional regulators, which convey the information that is contained in the sequence of DNA into RNA polymerase activity. This is achieved by the recruitment of transcriptional co-factors. One of the consequences of co-factor recruitment is the control of specific properties of nucleosomes, the basic units of chromatin, and their protein components, the core histones. The main principles are to regulate the position and the characteristics of nucleosomes. The latter includes modulating the composition of core histones and their variants that are integrated into nucleosomes, and the post-translational modification of these histones referred to as histone marks. One of these marks is the methylation of lysine 4 of the core histone H3 (H3K4). While mono-methylation of H3K4 (H3K4me1) is located preferentially at active enhancers, tri-methylation (H3K4me3) is a mark found at open and potentially active promoters. Thus, H3K4 methylation is typically associated with gene transcription. The class 2 lysine methyltransferases (KMTs) are the main enzymes that methylate H3K4. KMT2 enzymes function in complexes that contain a necessary core complex composed of WDR5, RBBP5, ASH2L, and DPY30, the so-called WRAD complex.
    [Show full text]
  • ARID1B Is a Specific Vulnerability in ARID1A-Mutant Cancers The
    ARID1B is a specific vulnerability in ARID1A-mutant cancers The Harvard community has made this article openly available. Please share how this access benefits you. Your story matters. Citation Helming, K. C., X. Wang, B. G. Wilson, F. Vazquez, J. R. Haswell, H. E. Manchester, Y. Kim, et al. 2014. “ARID1B is a specific vulnerability in ARID1A-mutant cancers.” Nature medicine 20 (3): 251-254. doi:10.1038/nm.3480. http://dx.doi.org/10.1038/nm.3480. Published Version doi:10.1038/nm.3480 Accessed February 16, 2015 10:04:32 PM EST Citable Link http://nrs.harvard.edu/urn-3:HUL.InstRepos:12987227 Terms of Use This article was downloaded from Harvard University's DASH repository, and is made available under the terms and conditions applicable to Other Posted Material, as set forth at http://nrs.harvard.edu/urn-3:HUL.InstRepos:dash.current.terms-of- use#LAA (Article begins on next page) NIH Public Access Author Manuscript Nat Med. Author manuscript; available in PMC 2014 September 01. NIH-PA Author ManuscriptPublished NIH-PA Author Manuscript in final edited NIH-PA Author Manuscript form as: Nat Med. 2014 March ; 20(3): 251–254. doi:10.1038/nm.3480. ARID1B is a specific vulnerability in ARID1A-mutant cancers Katherine C. Helming1,2,3,4,*, Xiaofeng Wang1,2,3,*, Boris G. Wilson1,2,3, Francisca Vazquez5, Jeffrey R. Haswell1,2,3, Haley E. Manchester1,2,3, Youngha Kim1,2,3, Gregory V. Kryukov5, Mahmoud Ghandi5, Andrew J. Aguirre5,6,7, Zainab Jagani8, Zhong Wang9, Levi A. Garraway6, William C. Hahn6,7, and Charles W.
    [Show full text]
  • The Landscape of Somatic Mutations in Epigenetic Regulators Across 1,000 Paediatric Cancer Genomes
    ARTICLE Received 24 Sep 2013 | Accepted 12 Mar 2014 | Published 8 Apr 2014 DOI: 10.1038/ncomms4630 The landscape of somatic mutations in epigenetic regulators across 1,000 paediatric cancer genomes Robert Huether1,*, Li Dong2,*, Xiang Chen1, Gang Wu1, Matthew Parker1, Lei Wei1, Jing Ma2, Michael N. Edmonson1, Erin K. Hedlund1, Michael C. Rusch1, Sheila A. Shurtleff2, Heather L. Mulder3, Kristy Boggs3, Bhavin Vadordaria3, Jinjun Cheng2, Donald Yergeau3, Guangchun Song2, Jared Becksfort1, Gordon Lemmon1, Catherine Weber2, Zhongling Cai2, Jinjun Dang2, Michael Walsh4, Amanda L. Gedman2, Zachary Faber2, John Easton3, Tanja Gruber2,4, Richard W. Kriwacki5, Janet F. Partridge6, Li Ding7,8,9, Richard K. Wilson7,8,9, Elaine R. Mardis7,8,9, Charles G. Mullighan2, Richard J. Gilbertson10, Suzanne J. Baker10, Gerard Zambetti6, David W. Ellison2, Jinghui Zhang1 & James R. Downing2 Studies of paediatric cancers have shown a high frequency of mutation across epigenetic regulators. Here we sequence 633 genes, encoding the majority of known epigenetic regulatory proteins, in over 1,000 paediatric tumours to define the landscape of somatic mutations in epigenetic regulators in paediatric cancer. Our results demonstrate a marked variation in the frequency of gene mutations across 21 different paediatric cancer subtypes, with the highest frequency of mutations detected in high-grade gliomas, T-lineage acute lymphoblastic leukaemia and medulloblastoma, and a paucity of mutations in low-grade glioma and retinoblastoma. The most frequently mutated genes are H3F3A, PHF6, ATRX, KDM6A, SMARCA4, ASXL2, CREBBP, EZH2, MLL2, USP7, ASXL1, NSD2, SETD2, SMC1A and ZMYM3. We identify novel loss-of-function mutations in the ubiquitin-specific processing protease 7 (USP7) in paediatric leukaemia, which result in decreased deubiquitination activity.
    [Show full text]
  • Genetic and Genomic Analysis of Hyperlipidemia, Obesity and Diabetes Using (C57BL/6J × TALLYHO/Jngj) F2 Mice
    University of Tennessee, Knoxville TRACE: Tennessee Research and Creative Exchange Nutrition Publications and Other Works Nutrition 12-19-2010 Genetic and genomic analysis of hyperlipidemia, obesity and diabetes using (C57BL/6J × TALLYHO/JngJ) F2 mice Taryn P. Stewart Marshall University Hyoung Y. Kim University of Tennessee - Knoxville, [email protected] Arnold M. Saxton University of Tennessee - Knoxville, [email protected] Jung H. Kim Marshall University Follow this and additional works at: https://trace.tennessee.edu/utk_nutrpubs Part of the Animal Sciences Commons, and the Nutrition Commons Recommended Citation BMC Genomics 2010, 11:713 doi:10.1186/1471-2164-11-713 This Article is brought to you for free and open access by the Nutrition at TRACE: Tennessee Research and Creative Exchange. It has been accepted for inclusion in Nutrition Publications and Other Works by an authorized administrator of TRACE: Tennessee Research and Creative Exchange. For more information, please contact [email protected]. Stewart et al. BMC Genomics 2010, 11:713 http://www.biomedcentral.com/1471-2164/11/713 RESEARCH ARTICLE Open Access Genetic and genomic analysis of hyperlipidemia, obesity and diabetes using (C57BL/6J × TALLYHO/JngJ) F2 mice Taryn P Stewart1, Hyoung Yon Kim2, Arnold M Saxton3, Jung Han Kim1* Abstract Background: Type 2 diabetes (T2D) is the most common form of diabetes in humans and is closely associated with dyslipidemia and obesity that magnifies the mortality and morbidity related to T2D. The genetic contribution to human T2D and related metabolic disorders is evident, and mostly follows polygenic inheritance. The TALLYHO/ JngJ (TH) mice are a polygenic model for T2D characterized by obesity, hyperinsulinemia, impaired glucose uptake and tolerance, hyperlipidemia, and hyperglycemia.
    [Show full text]
  • Loss of Fam60a, a Sin3a Subunit, Results in Embryonic Lethality and Is Associated with Aberrant Methylation at a Subset of Gene
    RESEARCH ARTICLE Loss of Fam60a, a Sin3a subunit, results in embryonic lethality and is associated with aberrant methylation at a subset of gene promoters Ryo Nabeshima1,2, Osamu Nishimura3,4, Takako Maeda1, Natsumi Shimizu2, Takahiro Ide2, Kenta Yashiro1†, Yasuo Sakai1, Chikara Meno1, Mitsutaka Kadota3,4, Hidetaka Shiratori1†, Shigehiro Kuraku3,4*, Hiroshi Hamada1,2* 1Developmental Genetics Group, Graduate School of Frontier Biosciences, Osaka University, Suita, Japan; 2Laboratory for Organismal Patterning, RIKEN Center for Developmental Biology, Kobe, Japan; 3Phyloinformatics Unit, RIKEN Center for Life Science Technologies, Kobe, Japan; 4Laboratory for Phyloinformatics, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan Abstract We have examined the role of Fam60a, a gene highly expressed in embryonic stem cells, in mouse development. Fam60a interacts with components of the Sin3a-Hdac transcriptional corepressor complex, and most Fam60a–/– embryos manifest hypoplasia of visceral organs and die in utero. Fam60a is recruited to the promoter regions of a subset of genes, with the expression of these genes being either up- or down-regulated in Fam60a–/– embryos. The DNA methylation level of the Fam60a target gene Adhfe1 is maintained at embryonic day (E) 7.5 but markedly reduced at –/– *For correspondence: E9.5 in Fam60a embryos, suggesting that DNA demethylation is enhanced in the mutant. [email protected] (SK); Examination of genome-wide DNA methylation identified several differentially methylated regions, [email protected] (HH) which were preferentially hypomethylated, in Fam60a–/– embryos. Our data suggest that Fam60a is †These authors contributed required for proper embryogenesis, at least in part as a result of its regulation of DNA methylation equally to this work at specific gene promoters.
    [Show full text]
  • Inherited Variation in Mir-290 Expression Suppresses Breast Cancer Progression by Targeting the Metastasis Susceptibility Gene Arid4b
    Published OnlineFirst February 27, 2013; DOI: 10.1158/0008-5472.CAN-12-3513 Cancer Tumor and Stem Cell Biology Research Inherited Variation in miR-290 Expression Suppresses Breast Cancer Progression by Targeting the Metastasis Susceptibility Gene Arid4b Natalie Goldberger1, Renard C. Walker1, Chang Hee Kim2, Scott Winter1, and Kent W. Hunter1 Abstract The metastatic cascade is a complex and extremely inefficient process with many potential barriers. Understanding this process is of critical importance because the majority of cancer mortality is associated with metastatic disease. Recently, it has become increasingly clear that microRNAs (miRNA) play important roles in tumorigenesis and metastasis, yet few studies have examined how germline variations may dysregulate miRNAs, in turn affecting metastatic potential. To explore this possibility, the highly metastatic MMTV-PyMT mice were crossed with 25 AKXD (AKR/J Â DBA/2J) recombinant inbred strains to produce F1 progeny with varying metastatic indices. When mammary tumors from the F1 progeny were analyzed by miRNA microarray, miR-290 (containing miR-290-3p and miR-290-5p) was identified as a top candidate progression-associated miRNA. The microarray results were validated in vivo when miR-290 upregulation in two independent breast cancer cell lines suppressed both primary tumor and metastatic growth. Computational analysis identified breast cancer progression gene Arid4b as a top target of miR-290-3p, which was confirmed by luciferase reporter assay. Surprisingly, pathway analysis identified estrogen receptor (ER) signaling as the top canonical pathway affected by miR-290 upregulation. Further analysis showed that ER levels were elevated in miR-290–expressing tumors and positively correlated with apoptosis.
    [Show full text]
  • Advances in Understanding Cancer Genomes Through Second-Generation Sequencing
    Advances in understanding cancer genomes through second-generation sequencing Matthew Meyerson, Stacey Gabriel and Gad Getz Second-generation A major near-term medical impact of the genome comprehensive genome-based diagnosis of cancer is sequencing technology revolution will be the elucidation of mecha- becoming increasingly crucial for therapeutic decisions. Used in this Review to refer to nisms of cancer pathogenesis, leading to improvements During the past decades, there have been major sequencing methods that have in the diagnosis of cancer and the selection of cancer advances in experimental and informatic methods emerged since 2005 that second-generation sequencing parallelize the sequencing treatment. Thanks to for genome characterization based on DNA and RNA 1–5 process and produce millions technologies , recently it has become feasible to microarrays and on capillary-based DNA sequenc- of typically short sequence sequence the expressed genes (‘transcriptomes’)6,7, ing (‘first-generation sequencing’, also known as Sanger reads (50–400 bases) from known exons (‘exomes’)8,9, and complete genomes10–15 sequencing). These technologies provided the ability amplified DNA clones. of cancer samples. to analyse exonic mutations and copy number altera- It is also often known as next-generation sequencing. These technological advances are important for tions and have led to the discovery of many important advancing our understanding of malignant neoplasms alterations in the cancer genome20. because cancer is fundamentally a disease of the genome. However, there are particular challenges for the A wide range of genomic alterations — including point detection and diagnosis of cancer genome alterations. mutations, copy number changes and rearrangements For example, some genomic alterations in cancer are — can lead to the development of cancer.
    [Show full text]