NRF1) Coordinates Changes in the Transcriptional and Chromatin Landscape Affecting Development and Progression of Invasive Breast Cancer

Total Page:16

File Type:pdf, Size:1020Kb

NRF1) Coordinates Changes in the Transcriptional and Chromatin Landscape Affecting Development and Progression of Invasive Breast Cancer Florida International University FIU Digital Commons FIU Electronic Theses and Dissertations University Graduate School 11-7-2018 Decipher Mechanisms by which Nuclear Respiratory Factor One (NRF1) Coordinates Changes in the Transcriptional and Chromatin Landscape Affecting Development and Progression of Invasive Breast Cancer Jairo Ramos [email protected] Follow this and additional works at: https://digitalcommons.fiu.edu/etd Part of the Clinical Epidemiology Commons Recommended Citation Ramos, Jairo, "Decipher Mechanisms by which Nuclear Respiratory Factor One (NRF1) Coordinates Changes in the Transcriptional and Chromatin Landscape Affecting Development and Progression of Invasive Breast Cancer" (2018). FIU Electronic Theses and Dissertations. 3872. https://digitalcommons.fiu.edu/etd/3872 This work is brought to you for free and open access by the University Graduate School at FIU Digital Commons. It has been accepted for inclusion in FIU Electronic Theses and Dissertations by an authorized administrator of FIU Digital Commons. For more information, please contact [email protected]. FLORIDA INTERNATIONAL UNIVERSITY Miami, Florida DECIPHER MECHANISMS BY WHICH NUCLEAR RESPIRATORY FACTOR ONE (NRF1) COORDINATES CHANGES IN THE TRANSCRIPTIONAL AND CHROMATIN LANDSCAPE AFFECTING DEVELOPMENT AND PROGRESSION OF INVASIVE BREAST CANCER A dissertation submitted in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY in PUBLIC HEALTH by Jairo Ramos 2018 To: Dean Tomás R. Guilarte Robert Stempel College of Public Health and Social Work This dissertation, Written by Jairo Ramos, and entitled Decipher Mechanisms by Which Nuclear Respiratory Factor One (NRF1) Coordinates Changes in the Transcriptional and Chromatin Landscape Affecting Development and Progression of Invasive Breast Cancer, having been approved in respect to style and intellectual content, is referred to you for judgment. We have read the dissertation and recommend that it be approved. ___________________________________________ Alok Deoraj ___________________________________________ Juan Luizzi ___________________________________________ Quentin Felty __________________________________________ Deodutta Roy, Co-Major Professor __________________________________________ ChangWon Yoo, Co-Major Professor Date of Defense: November 7, 2018 The dissertation of Jairo Ramos is approved. ___________________________________________ Dean Tomás R. Guilarte R. Stempel College of Public Health and Social Work ___________________________________________ Andres G. Gil Vice President for Research and Economic Development and Dean of the University Graduate School Florida International University, 2018 ii © Copyright 2018 by Jairo Ramos All rights reserved. iii DEDICATION This dissertation is dedicated to the memory of my parents, Raul Ramos and Elinor Vega de Ramos, Who alWays encouraged me to pursue my dreams through education; to the memory of my sister, Meibis Ramos, who was alWays there for me through the good and bad times; and to my Wife, Mile, and my two sons, Jairo and Andres, Who Were my inspiration to pursue my doctoral degree. iv ACKNOWLEDGMENTS I Would like to thank first my major professor, Dr. Deodutta Roy, for his continuous support and patience and for sharing With me all his knoWledge in the fields of genetics, epigenetics, and carcinogenesis. I have learned a great deal from him. The research and Writing of this dissertation Would have not been possible Without his help and guidance. I Wish to thank my co-major professor, Dr. ChangWon Yoo, for his assistance and support in the field of biostatistics and for facilitating the tools (hardWare and softWare) I needed for the statistical analysis section of my research. I also Want to express my gratitude to the other members of my dissertation committee: Dr. Quentin Felty, Dr. Alok Deoraj, and Dr. Juan Liuzzi, for their encouragement, comments, and suggestions that helped me to complete successfully this dissertation. I thank too my felloW students from the Environmental Health Science Department and from the SMLG group for the stimulating discussions and camaraderie and for sharing With me their knoWledge in different fields of health sciences and statistics. My thanks also to Marie Alvarez, our former departmental secretary, who helped me greatly With all the administrative matters and the use of the university’s resources. My sincere gratitude goes to the Starr Foundation for its generosity and to the committee members at Stempel College Who selected me as one of the recipients of the STARR scholarship in 2016 and 2017. v Last but not least, I Would like to thank my brother, Dr. Ariel Ramos, and my nepheW, Larry Ramos, for their continuous support and encouragement while I pursued my dream of becoming a scientist. vi ABSTRACT OF THE DISSERTATION DECIPHER MECHANISMS BY WHICH NUCLEAR RESPIRATORY FACTOR ONE (NRF1) COORDINATES CHANGES IN THE TRANSCRIPTIONAL AND CHROMATIN LANDSCAPE AFFECTING DEVELOPMENT AND PROGRESSION OF INVASIVE BREAST CANCER by Jairo Ramos Florida International University, 2018 Miami, Florida Professor Deodutta Roy, Co-Major Professor Professor ChangWon Yoo, Co-Major Professor Despite tremendous progress in the understanding of breast cancer (BC), gaps remain in our knoWledge of the molecular basis underlying the aggressiveness of BC and BC disparities. Nuclear respiratory factor 1 (NRF1) is a transcription factor (TF) knoWn to control breast cancer cell cycle progression. DNA response elements bound by NRF1 positively correlate With the progression of malignant breast cancer. Mechanistic aspects by Which NRF1 contributes to susceptibility to different breast tumor subtypes are still not fully understood. Therefore, the primary objective of this dissertation was to decipher mechanisms by Which NRF1 coordinates changes in the transcriptional and chromatin landscape affecting development and progression of invasive breast cancer. Our hypothesis was that NRF1 reprogramming the transcription of tumor initiating gene(s) and tumor suppressor gene(s) contribute in the development and progression of vii invasive breast cancer. To test this hypothesis, We proposed three specific aims: (a) Decipher regulatory landscape of NRF1 netWorks in breast cancer. (b) Determine the role of NRF1 gene netWorks in different subtypes of breast cancer. (c) Determine differential NRF1 gene netWork sensitivity contributing to breast cancer disparities. Our approach to test these aims consisted of a systematic integration of ChIP DNA-seq, RNA-Seq, NRF1 protein-DNA motif binding, signal pathWay analysis, and Bayesian machine learning. We uncovered a novel oncogenic role for NRF1. This discovery strongly supported the supposition that NRF1 overexpression is sufficient to derive breast tumorigenesis. We also observed neW roles for NRF1 in the acquisition of breast tumor initiating cells, regulation of epithelial to mesenchymal transition (EMT), and invasiveness of BC stem cells. Furthermore, through the use of Bayesian netWork structure learning we found that the NRF1 motif Was enriched in 14 associated With HER2 amplified breast cancer. Three genes—GSK3B, E2F3, and PIK3CA—Were able to predict HER2 breast tumor status With 96% to100% confidence. The findings of this study also shoWed the roles of NRF1 sensitivity to development of lobular A, Her2+, and TNBC in different racial/ethnic groups of breast cancer patients. In summary, our study revealed for the first time the role of NRF1 in the pathogenesis of invasive BC and BC disparities. viii TABLE OF CONTENTS CHAPTER PAGE I. INTRODUCTION ............................................................................................. 1 Overall Goal ................................................................................................ 5 Hypothesis and Specific Aims .................................................................... 5 REFERENCES ........................................................................................... 6 II. CURRENT KNOWLEDGE OF NRF1 INVOLVEMENT IN THE PATHOGENESIS OF BREAST CANCER, INCLUDING GENE ONTOLOGY AND PATHWAY ANALYSIS OF NRF1 REGULATED NETWORKS .................................................................................................... 9 Abstract....................................................................................................... 9 Introduction ............................................................................................... 10 Results and Discussion ............................................................................ 12 Conclusion ................................................................................................ 78 Methods .................................................................................................... 78 REFERENCES ......................................................................................... 79 III. INTEGRATED CHIP-SEQ AND RNA-SEQDATA ANALYSIS TO INVESTIGATE REGULATORY MECHANISMS OF NRF1 TRANSCRIPTION FACTOR ON TARGET GENES IN HER2+ BREAST CANCER CELLS ........................................................................................... 86 Abstract..................................................................................................... 86 Introduction ............................................................................................... 87 Methods .................................................................................................... 88 Results and Discussion .........................................................................
Recommended publications
  • Comparative Transcriptome Analyses Indicate Molecular Homology of Zebrafish Swimbladder and Mammalian Lung
    Comparative Transcriptome Analyses Indicate Molecular Homology of Zebrafish Swimbladder and Mammalian Lung Weiling Zheng1, Zhengyuan Wang1, John E. Collins2, Robert M. Andrews2, Derek Stemple2, Zhiyuan Gong1* 1 Department of Biological Sciences, National University of Singapore, Singapore, Singapore, 2 Vertebrate Development and Genetics, Wellcome Trust Genome Campus, Wellcome Trust Sanger Institute, Hinxton, Cambridge, United Kingdom Abstract The fish swimbladder is a unique organ in vertebrate evolution and it functions for regulating buoyancy in most teleost species. It has long been postulated as a homolog of the tetrapod lung, but the molecular evidence is scarce. In order to understand the molecular function of swimbladder as well as its relationship with lungs in tetrapods, transcriptomic analyses of zebrafish swimbladder were carried out by RNA-seq. Gene ontology classification showed that genes in cytoskeleton and endoplasmic reticulum were enriched in the swimbladder. Further analyses depicted gene sets and pathways closely related to cytoskeleton constitution and regulation, cell adhesion, and extracellular matrix. Several prominent transcription factor genes in the swimbladder including hoxc4a, hoxc6a, hoxc8a and foxf1 were identified and their expressions in developing swimbladder during embryogenesis were confirmed. By comparison of enriched transcripts in the swimbladder with those in human and mouse lungs, we established the resemblance of transcriptome of the zebrafish swimbladder and mammalian lungs. Based on the transcriptomic data of zebrafish swimbladder, the predominant functions of swimbladder are in its epithelial and muscular tissues. Our comparative analyses also provide molecular evidence of the relatedness of the fish swimbladder and mammalian lung. Citation: Zheng W, Wang Z, Collins JE, Andrews RM, Stemple D, et al.
    [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]
  • The Utility of Genetic Risk Scores in Predicting the Onset of Stroke March 2021 6
    DOT/FAA/AM-21/24 Office of Aerospace Medicine Washington, DC 20591 The Utility of Genetic Risk Scores in Predicting the Onset of Stroke Diana Judith Monroy Rios, M.D1 and Scott J. Nicholson, Ph.D.2 1. KR 30 # 45-03 University Campus, Building 471, 5th Floor, Office 510 Bogotá D.C. Colombia 2. FAA Civil Aerospace Medical Institute, 6500 S. MacArthur Blvd Rm. 354, Oklahoma City, OK 73125 March 2021 NOTICE This document is disseminated under the sponsorship of the U.S. Department of Transportation in the interest of information exchange. The United States Government assumes no liability for the contents thereof. _________________ This publication and all Office of Aerospace Medicine technical reports are available in full-text from the Civil Aerospace Medical Institute’s publications Web site: (www.faa.gov/go/oamtechreports) Technical Report Documentation Page 1. Report No. 2. Government Accession No. 3. Recipient's Catalog No. DOT/FAA/AM-21/24 4. Title and Subtitle 5. Report Date March 2021 The Utility of Genetic Risk Scores in Predicting the Onset of Stroke 6. Performing Organization Code 7. Author(s) 8. Performing Organization Report No. Diana Judith Monroy Rios M.D1, and Scott J. Nicholson, Ph.D.2 9. Performing Organization Name and Address 10. Work Unit No. (TRAIS) 1 KR 30 # 45-03 University Campus, Building 471, 5th Floor, Office 510, Bogotá D.C. Colombia 11. Contract or Grant No. 2 FAA Civil Aerospace Medical Institute, 6500 S. MacArthur Blvd Rm. 354, Oklahoma City, OK 73125 12. Sponsoring Agency name and Address 13. Type of Report and Period Covered Office of Aerospace Medicine Federal Aviation Administration 800 Independence Ave., S.W.
    [Show full text]
  • Seq2pathway Vignette
    seq2pathway Vignette Bin Wang, Xinan Holly Yang, Arjun Kinstlick May 19, 2021 Contents 1 Abstract 1 2 Package Installation 2 3 runseq2pathway 2 4 Two main functions 3 4.1 seq2gene . .3 4.1.1 seq2gene flowchart . .3 4.1.2 runseq2gene inputs/parameters . .5 4.1.3 runseq2gene outputs . .8 4.2 gene2pathway . 10 4.2.1 gene2pathway flowchart . 11 4.2.2 gene2pathway test inputs/parameters . 11 4.2.3 gene2pathway test outputs . 12 5 Examples 13 5.1 ChIP-seq data analysis . 13 5.1.1 Map ChIP-seq enriched peaks to genes using runseq2gene .................... 13 5.1.2 Discover enriched GO terms using gene2pathway_test with gene scores . 15 5.1.3 Discover enriched GO terms using Fisher's Exact test without gene scores . 17 5.1.4 Add description for genes . 20 5.2 RNA-seq data analysis . 20 6 R environment session 23 1 Abstract Seq2pathway is a novel computational tool to analyze functional gene-sets (including signaling pathways) using variable next-generation sequencing data[1]. Integral to this tool are the \seq2gene" and \gene2pathway" components in series that infer a quantitative pathway-level profile for each sample. The seq2gene function assigns phenotype-associated significance of genomic regions to gene-level scores, where the significance could be p-values of SNPs or point mutations, protein-binding affinity, or transcriptional expression level. The seq2gene function has the feasibility to assign non-exon regions to a range of neighboring genes besides the nearest one, thus facilitating the study of functional non-coding elements[2]. Then the gene2pathway summarizes gene-level measurements to pathway-level scores, comparing the quantity of significance for gene members within a pathway with those outside a pathway.
    [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]
  • New PDF Document
    888.267.4436 [email protected] www.origene.com Name:CLDN6 mouse monoclonal antibody, clone OTI3E3 (formerly 3E3) Catalog: TA507011 Product Data Sheet - TRUEMAB Components: • CLDN6 mouse monoclonal antibody, clone OTI3E3 (formerly 3E3) (TA507011) • WB positive control also available: 20ug myc-DDK tagged CLDN6 HEK293T over-expression lysate lyophilized in RIPA buffer (LC412034). (Reconstitute into 20ul of 1x SDS sample buffer before loading; load 5ul per lane as WB control or as desired) Amount: 100ul Immunogen: Full length human recombinant protein of human CLDN6(NP_067018) produced in HEK293T cell. Host: Mouse Isotype: IgG1 Species Reactivity: Human Guaranteed WB, IF Applications: Suggested WB 1:1000, IF 1:100, Dilutions: Concentration: 1 mg/ml Buffer: PBS (PH 7.3) containing 1% BSA, 50% glycerol and 0.02% sodium azide. Purification: Purified from mouse ascites fluids by affinity chromatography Storage Condition: Shipped at -20C or with ice packs. Upon delivery store at -20C. Dilute in PBS (pH7.3) if necessary. Stable for 12 months from date of receipt. Avoid repeated freeze-thaws. Target Target Name: Homo sapiens claudin 6 (CLDN6) Alternative Name: OTTHUMP00000159248; claudin 6 Database Link: NP_067018 Entrez Gene 9074 Human Function: Tight junctions represent one mode of cell-to-cell adhesion in epithelial or endothelial cell sheets, forming continuous seals around cells and serving as a physical barrier to prevent solutes and water from passing freely through the paracellular space. These junctions are comprised of sets of continuous networking strands in the outwardly facing cytoplasmic leaflet, with complementary grooves in the inwardly facing extracytoplasmic leaflet. This gene encodes a component of tight junction strands, which This product is to be used for laboratory only.
    [Show full text]
  • Polyclonal Antibody to ARFGAP3 (C-Term) - Aff - Purified
    OriGene Technologies, Inc. OriGene Technologies GmbH 9620 Medical Center Drive, Ste 200 Schillerstr. 5 Rockville, MD 20850 32052 Herford UNITED STATES GERMANY Phone: +1-888-267-4436 Phone: +49-5221-34606-0 Fax: +1-301-340-8606 Fax: +49-5221-34606-11 [email protected] [email protected] AP15932PU-N Polyclonal Antibody to ARFGAP3 (C-term) - Aff - Purified Alternate names: ADP-ribosylation factor GTPase-activating protein 3, ARF GAP 3, ARFGAP1 Quantity: 0.1 mg Concentration: 0.5 mg/ml Background: The protein encoded by this gene is a GTPase-activating protein (GAP) which associates with the Golgi apparatus and which is thought to interact with ADP- ribosylation factor 1 (ARF1). The encoded protein likely promotes hydrolysis of ARF1-bound GTP, which is required for the dissociation of coat proteins from Golgi- derived membranes and vesicles. Dissociation of the coat proteins is a prerequisite for the fusion of these vesicles with target compartments. The activity of this protein is sensitive to phospholipids. This gene was originally known as ARFGAP1, but that is now the name of a related but different gene. Uniprot ID: Q9NP61 NCBI: NP_055385.3 GeneID: 26286 Host: Goat Immunogen: Peptide from C Terminus of the protein sequence according to NP_055385.3; NP_001135765.1 Genename: ARFGAP3 AA Sequence: C-NGVVTSIQDRYGS Format: State: Liquid Ig fraction Purification: Ammonium sulphate precipitation followed by antigen affinity chromatography using the immunizing peptide Buffer System: Tris saline, 0.02% sodium azide, pH7.3 with 0.5% bovine serum albumin Applications: Peptide ELISA: Limit dilution 1:8000. Western blot: 1-3 µg/ml.
    [Show full text]
  • Exome-Wide Meta-Analysis Identifies Rare 3'-UTR Variant In
    View metadata, citation and similar papers at core.ac.uk brought to you by CORE provided by Erasmus University Digital Repository ORIGINAL RESEARCH published: 18 October 2017 doi: 10.3389/fgene.2017.00151 Exome-Wide Meta-Analysis Identifies Rare 3′-UTR Variant in ERCC1/CD3EAP Associated with Symptoms of Sleep Apnea Ashley van der Spek 1, Annemarie I. Luik 2, Desana Kocevska 3, Chunyu Liu 4, 5, 6, Rutger W. W. Brouwer 7, Jeroen G. J. van Rooij 8, 9, 10, Mirjam C. G. N. van den Hout 7, Robert Kraaij 1, 8, 9, Albert Hofman 1, 11, André G. Uitterlinden 1, 8, 9, Wilfred F. J. van IJcken 7, Daniel J. Gottlieb 12, 13, 14, Henning Tiemeier 1, 15, Cornelia M. van Duijn 1 and Najaf Amin 1* 1 Department of Epidemiology, Erasmus Medical Center, Rotterdam, Netherlands, 2 Sleep and Circadian Neuroscience Institute, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom, 3 Department of Child and Adolescent Psychiatry, Erasmus Medical Center, Rotterdam, Netherlands, 4 Framingham Heart Study, National Heart, Lung, and Blood Institute, Framingham, MA, United States, 5 Population Sciences Branch, National Heart, Lung, and Blood Institute, Bethesda, MD, United States, 6 Department of Biostatistics, School of Public Health, Boston University, Boston, MA, United States, 7 Center for Biomics, Erasmus Medical Center, Rotterdam, Netherlands, 8 Department of Internal Medicine, Erasmus Medical Center, Rotterdam, Netherlands, 9 Netherlands Consortium for Healthy Ageing, Rotterdam, Netherlands, 10 Department of Neurology, Erasmus
    [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]
  • Supplementary Table 1: Adhesion Genes Data Set
    Supplementary Table 1: Adhesion genes data set PROBE Entrez Gene ID Celera Gene ID Gene_Symbol Gene_Name 160832 1 hCG201364.3 A1BG alpha-1-B glycoprotein 223658 1 hCG201364.3 A1BG alpha-1-B glycoprotein 212988 102 hCG40040.3 ADAM10 ADAM metallopeptidase domain 10 133411 4185 hCG28232.2 ADAM11 ADAM metallopeptidase domain 11 110695 8038 hCG40937.4 ADAM12 ADAM metallopeptidase domain 12 (meltrin alpha) 195222 8038 hCG40937.4 ADAM12 ADAM metallopeptidase domain 12 (meltrin alpha) 165344 8751 hCG20021.3 ADAM15 ADAM metallopeptidase domain 15 (metargidin) 189065 6868 null ADAM17 ADAM metallopeptidase domain 17 (tumor necrosis factor, alpha, converting enzyme) 108119 8728 hCG15398.4 ADAM19 ADAM metallopeptidase domain 19 (meltrin beta) 117763 8748 hCG20675.3 ADAM20 ADAM metallopeptidase domain 20 126448 8747 hCG1785634.2 ADAM21 ADAM metallopeptidase domain 21 208981 8747 hCG1785634.2|hCG2042897 ADAM21 ADAM metallopeptidase domain 21 180903 53616 hCG17212.4 ADAM22 ADAM metallopeptidase domain 22 177272 8745 hCG1811623.1 ADAM23 ADAM metallopeptidase domain 23 102384 10863 hCG1818505.1 ADAM28 ADAM metallopeptidase domain 28 119968 11086 hCG1786734.2 ADAM29 ADAM metallopeptidase domain 29 205542 11085 hCG1997196.1 ADAM30 ADAM metallopeptidase domain 30 148417 80332 hCG39255.4 ADAM33 ADAM metallopeptidase domain 33 140492 8756 hCG1789002.2 ADAM7 ADAM metallopeptidase domain 7 122603 101 hCG1816947.1 ADAM8 ADAM metallopeptidase domain 8 183965 8754 hCG1996391 ADAM9 ADAM metallopeptidase domain 9 (meltrin gamma) 129974 27299 hCG15447.3 ADAMDEC1 ADAM-like,
    [Show full text]
  • Intrinsic Disorder in PTEN and Its Interactome Confers Structural Plasticity and 63
    OPEN Intrinsic Disorder in PTEN and its SUBJECT AREAS: Interactome Confers Structural Plasticity CELLULAR SIGNALLING NETWORKS and Functional Versatility CANCER GENOMICS Prerna Malaney1*, Ravi Ramesh Pathak1*, Bin Xue3, Vladimir N. Uversky3,4,5 & Vrushank Dave´1,2 SYSTEMS ANALYSIS GENE REGULATORY NETWORKS 1Morsani College of Medicine, Department of Pathology and Cell Biology, 2Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, 3Department of Molecular Medicine, 4USF Health Byrd Alzheimer’s Research Institute, Received University of South Florida, Tampa, FL, 33612, USA, 5Institute for Biological Instrumentation, Russian Academy of Sciences, 142290 11 March 2013 Pushchino, Moscow Region, Russia. Accepted 3 June 2013 IDPs, while structurally poor, are functionally rich by virtue of their flexibility and modularity. However, how mutations in IDPs elicit diseases, remain elusive. Herein, we have identified tumor suppressor PTEN as Published an intrinsically disordered protein (IDP) and elucidated the molecular principles by which its intrinsically 20 June 2013 disordered region (IDR) at the carboxyl-terminus (C-tail) executes its functions. Post-translational modifications, conserved eukaryotic linear motifs and molecular recognition features present in the C-tail IDR enhance PTEN’s protein-protein interactions that are required for its myriad cellular functions. PTEN primary and secondary interactomes are also enriched in IDPs, most being cancer related, revealing that Correspondence and PTEN functions emanate from and are nucleated by the C-tail IDR, which form pliable network-hubs. requests for materials Together, PTEN higher order functional networks operate via multiple IDP-IDP interactions facilitated by should be addressed to its C-tail IDR. Targeting PTEN IDR and its interaction hubs emerges as a new paradigm for treatment of V.D.
    [Show full text]
  • BMC Cell Biology Biomed Central
    BMC Cell Biology BioMed Central Research article Open Access Mutational analysis of βCOP (Sec26p) identifies an appendage domain critical for function Carol J DeRegis1, Peter B Rahl2, Gregory R Hoffman3, Richard A Cerione4,5 and Ruth N Collins*4,6 Address: 1Graduate Program in Comparative Biomedical Sciences, Cornell University, Ithaca NY 14853, USA, 2Graduate Program in Pharmacology, Cornell University, Ithaca, NY 14853, USA, 3Graduate Program in Biophysics, Cornell University, Ithaca, NY 14853, USA, 4Department of Molecular Medicine, Cornell University, Ithaca, NY 14853, USA, 5Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853, USA and 6Department of Molecular Medicine, Cornell University, C4-109 Veterinary Medical Center, Ithaca, NY 14853, USA Email: Carol J DeRegis - [email protected]; Peter B Rahl - [email protected]; Gregory R Hoffman - [email protected]; Richard A Cerione - [email protected]; Ruth N Collins* - [email protected] * Corresponding author Published: 22 January 2008 Received: 2 June 2007 Accepted: 22 January 2008 BMC Cell Biology 2008, 9:3 doi:10.1186/1471-2121-9-3 This article is available from: http://www.biomedcentral.com/1471-2121/9/3 © 2008 De Regis et al; licensee BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Abstract Background: The appendage domain of the γCOP subunit of the COPI vesicle coat bears a striking structural resemblance to adaptin-family appendages despite limited primary sequence homology.
    [Show full text]