A Panel of Inbred Mouse Strains Suitable for Analysis of Complex Genetic Traits

Total Page:16

File Type:pdf, Size:1020Kb

A Panel of Inbred Mouse Strains Suitable for Analysis of Complex Genetic Traits Mamm Genome DOI 10.1007/s00335-012-9411-5 Hybrid mouse diversity panel: a panel of inbred mouse strains suitable for analysis of complex genetic traits Anatole Ghazalpour • Christoph D. Rau • Charles R. Farber • Brian J. Bennett • Luz D. Orozco • Atila van Nas • Calvin Pan • Hooman Allayee • Simon W. Beaven • Mete Civelek • Richard C. Davis • Thomas A. Drake • Rick A. Friedman • Nick Furlotte • Simon T. Hui • J. David Jentsch • Emrah Kostem • Hyun Min Kang • Eun Yong Kang • Jong Wha Joo • Vyacheslav A. Korshunov • Rick E. Laughlin • Lisa J. Martin • Jeffrey D. Ohmen • Brian W. Parks • Matteo Pellegrini • Karen Reue • Desmond J. Smith • Sotirios Tetradis • Jessica Wang • Yibin Wang • James N. Weiss • Todd Kirchgessner • Peter S. Gargalovic • Eleazar Eskin • Aldons J. Lusis • Rene´e C. LeBoeuf Received: 29 March 2012 / Accepted: 4 July 2012 Ó Springer Science+Business Media, LLC 2012 Abstract We have developed an association-based approach strains for a variety of clinical traits as well as intermediate using classical inbred strains of mice in which we correct for phenotypes and have shown that the HMDP has sufficient population structure, which is very extensive in mice, using an power to map genes for highly complex traits with resolution efficient mixed-model algorithm. Our approach includes inbred that is in most cases less than a megabase. In this essay, we parental strains as well as recombinant inbred strains in order to review our experience with the HMDP, describe various capture loci with effect sizes typical of complex traits in mice ongoing projects, and discuss how the HMDP may fit into the (in the range of 5 % of total trait variance). Over the last few larger picture of common diseases and different approaches. years, we have typed the hybrid mouse diversity panel (HMDP) Anatole Ghazalpour, Christoph D. Rau, Charles R. Farber, Brian J. Bennett, Luz D. Orozco, and Atila van Nas contributed equally to this work. A. Ghazalpour Á M. Civelek Á R. C. Davis Á S. T. Hui Á S. W. Beaven B. W. Parks Á A. J. Lusis Division of Digestive Diseases, Department of Medicine, Department of Medicine, David Geffen School of Medicine, David Geffen School of Medicine, University of California, University of California, Los Angeles, CA, USA Los Angeles, CA, USA C. D. Rau Á J. N. Weiss Á A. J. Lusis T. A. Drake Department of Microbiology, Immunology and Molecular Department of Pathology and Laboratory Medicine, Genetics, University of California, Los Angeles, CA, USA David Geffen School of Medicine, University of California, Los Angeles, CA, USA C. R. Farber Departments of Medicine and Biochemistry and Molecular R. A. Friedman Genetics, and Center for Public Health Genomics, University Department of Otology/Skull Base Surgery, of Virginia, Charlottesville, VA, USA House Research Institute, Los Angeles, CA, USA B. J. Bennett N. Furlotte Á E. Kostem Á E. Y. Kang Á E. Eskin Department of Genetics, and Nutrition Research Institute, Department of Computer Sciences, University of California, University of North Carolina, Chapel Hill, NC, USA Los Angeles, CA, USA L. D. Orozco Á A. van Nas Á C. Pan Á J. W. Joo Á J. D. Jentsch L. J. Martin Á K. Reue Á J. Wang Á E. Eskin Á A. J. Lusis Department of Psychology & Behavioral Neuroscience and Department of Human Genetics, David Geffen School of Psychiatry & Biobehavioral Sciences, David Geffen School Medicine, University of California, Los Angeles, CA, USA of Medicine, University of California, Los Angeles, CA, USA H. Allayee H. M. Kang Department of Preventive Medicine, Keck School of Medicine, Department of Biostatistics, School of Public Health, University of Southern California, Los Angeles, CA, USA University of Michigan, Ann Arbor, MI, USA 123 A. Ghazalpour et al.: Hybrid mouse diversity panel Introduction To address these limitations, we have developed an association-based approach using classical inbred strains of The human genome-wide association studies (GWAS) of mice (Bennett et al. 2010). We follow previous attempts to the last several years have provided the first unbiased views apply association in mice (Cervino et al. 2007; Grupe et al. of the genetics of common complex diseases, such as 2001; Guo et al. 2007; Liao et al. 2004; Liu et al. 2007; coronary artery disease, diabetes, and cancer. Many of the Pletcher et al. 2004) with two differences. First, we correct loci contain novel genes not previously connected to their for population structure, which is very extensive in mice, respective disease, indicating that there is great potential to using an efficient mixed-model algorithm (EMMA) (Kang discover new pathways and new targets for therapeutic et al. 2008). Second, to capture loci with effect sizes typical intervention. These GWAS do, however, have some of complex traits in mice (in the range of 5 % of total trait important limitations. First, human GWAS are not well variance), we supplemented the population with recombi- powered to study genetic interactions, such as gene-by-gene nant inbred (RI) strains. or gene-by-environment interactions (Zuk et al. 2012). Over the last few years, we have typed the hybrid mouse Second, it will be difficult to move from locus to a disease diversity panel (HMDP) strains for a variety of clinical pathway directly in humans (Altshuler et al. 2008). And traits as well as intermediate phenotypes, and have shown third, for most diseases, GWAS have identified only a small that the HMDP has sufficient power to map genes for fraction of the total genetic contributions and, thus, there is a highly complex traits with resolution that is in most cases great deal more to be discovered (Altshuler et al. 2008; less than a megabase. In this essay, we review our expe- Manolio et al. 2009). rience with the HMDP, describe various ongoing projects, To simplify genetic analysis, natural variations relevant and discuss how the HMDP may fit into the larger picture to disease have been studied in mice and rats (Ahlqvist of common diseases and different approaches. et al. 2011; Flint and Mackay 2009; Keane et al. 2011). This has generally involved traditional linkage mapping methods with crosses between different strains to identify Overview of the HMDP quantitative trait loci (QTLs). An important problem with such analysis has been poor mapping resolution because The hybrid mouse diversity panel (HMDP) consists of a the QTLs generally contain hundreds of genes, making the population of over 100 inbred mouse strains selected for identification of the causal genes difficult. usage in systematic genetic analyses of complex traits J. W. Joo S. Tetradis Á A. J. Lusis Bioinformatics Program, David Geffen School of Medicine, Division of Diagnostic and Surgical Science, School of University of California, Los Angeles, CA, USA Dentistry, University of California, Los Angeles, CA, USA V. A. Korshunov J. Wang Department of Medicine, Aab Cardiovascular Research Institute, Department of Medicine/Cardiology, David Geffen School University of Rochester School of Medicine and Dentistry, of Medicine, University of California, Los Angeles, CA, USA Rochester, NY, USA Y. Wang R. E. Laughlin Division of Molecular Medicine, Department of Anesthesiology, Semel Institute for Neuroscience and Human Behavior, Physiology and Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA University of California, Los Angeles, CA, USA J. D. Ohmen T. Kirchgessner Á P. S. Gargalovic Department of Cell Biology and Genetics, Department of Cardiovascular Drug Discovery, House Research Institute, Los Angeles, CA, USA Bristol-Myers Squibb Co, Pennington, NJ, USA M. Pellegrini R. C. LeBoeuf (&) Department of Molecular, Cell and Developmental Biology, Division of Metabolism, Endocrinology and Nutrition, University of California, Los Angeles, CA, USA Department of Medicine, University of Washington, 850 Republican Street, Seattle, WA 98109-4725, USA D. J. Smith e-mail: [email protected] Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA S. Tetradis Molecular Biology Institute, David Geffen School of Medicine, University of California, Los Angeles, CA, USA 123 A. Ghazalpour et al.: Hybrid mouse diversity panel (Table 1). Our goals in selecting the strains were to (1) greater than 5 % minor allele frequency (mean and median increase resolution of genetic mapping, (2) have a renew- minor allele frequency of 28 and 31 %) and are polymor- able resource that is available to all investigators world- phic between the strains. Using 0.8 as the r2 cutoff to define wide, and (3) provide a shared data repository that would LD, there are a total of 13,706 LD blocks with the median allow the integration of data across multiple scales, including size of 42.8 kb per block (mean of 143.3 kb per block) genomic, transcriptomic, metabolomic, proteomic, and scattered throughout the genome (Fig. 1). Since LD blocks clinical phenotypes. The core of our panel for association are more likely to define the window in which a candidate mapping (Bennett et al. 2010; Cervino et al. 2007; Grupe gene for a locus resides, the presence of small LD in the et al. 2001) consists of 29 classic parental inbred strains HMDP suggests that the number of causal candidate genes which are a subset of a group of mice commonly called the will be on average fewer than five genes per locus. This is a mouse diversity panel. We settled on our strains by elimi- considerable improvement over mapping resolution in nating closely related strains and removing wild-derived traditional linkage studies and/or in outbred stock mice strains. The decision to remove wild-derived stains is based where, on average, the number of candidate genes for each on the tradeoff between statistical power and genetic diver- locus ranges from 10 to 50. Large blocks (defined as LD sity. While we were sacrificing the genetic diversity by blocks[1 Mb) are also present in the HMDP panel but not leaving out wild-derived strains, our panel increased the frequently.
Recommended publications
  • Disruption of Adipose Rab10-Dependent Insulin
    Page 1 of 35 Diabetes Vazirani et al. Disruption of Adipose Rab10-Dependent Insulin Signaling Causes Hepatic Insulin Resistance Reema P. Vazirani1, Akanksha Verma2, L. Amanda Sadacca1, Melanie S. Buckman3, Belen Picatoste1, Muheeb Beg1, Christopher Torsitano1, Joanne H. Bruno1, Rajesh T. Patel1, Kotryna Simonyte4, Joao P. Camporez5, Gabriela Moreira5, Domenick J. Falcone6, Domenico Accili7, Olivier Elemento2, Gerald I. Shulman6,8, Barbara B. Kahn4 and Timothy E. McGraw1,3* 1Department of Biochemistry, Weill Cornell Medical College 2Department of Physiology and Biophysics, Weill Cornell Medical College 3Department of Cardiothoracic Surgery, Weill Cornell Medical College 4Beth Israel Deaconess Medical Center, Harvard Medical School 5Departments of Internal Medicine and Cellular & Molecular Physiology, Yale University 6Department of Pathology, Weill Cornell Medical College 7Department of Medicine and Berrie Diabetes Center, Columbia University 8Howard Hughes Medical Institute, Yale University *Contact: Timothy E. McGraw, PhD 1300 York Ave New York, NY 10065 212-746-4982 [email protected] The authors have declared that no conflict of interest exists. 1 Diabetes Publish Ahead of Print, published online March 25, 2016 Diabetes Page 2 of 35 Vazirani et al. Abstract Insulin controls glucose uptake into adipose and muscle cells by regulating the amount of the Glut4 glucose transporter in the plasma membrane. The effect of insulin is to promote translocation of intracellular Glut4 to the plasma membrane. The small Rab GTPase Rab10 is required for insulin-stimulated Glut4 translocation in cultured 3T3-L1 adipocytes. Here we demonstrate that both insulin-stimulated glucose uptake and Glut4 translocation to the plasma membrane are reduced by about half in adipocytes from adipose-specific Rab10 knockout mice.
    [Show full text]
  • Investigation Into the Effect of LRRK2-Rab10 Protein Interactions on the Proboscis Extension Response of the Fruit Fly Drosophila Melanogaster
    Investigation into the effect of LRRK2-Rab10 protein interactions on the Proboscis Extension Response of the fruit fly Drosophila melanogaster Laura Covill Masters by Research University of York Biology December 2018 Abstract Parkinson’s Disease (PD) is a debilitating disease which affects 1% of the population worldwide and is characterised by stiffness, tremor and bradykinesia. PD is a complex disease with many suspected genetic and environmental causes, and it is critical to understand all the pathways involved in disease progression to develop effective therapies for PD, which currently has no cure. A kinase- coding gene, LRRK2 has emerged as a focal point for much PD research, particularly PD-associated SNP LRRK2-G2019S, which leads to LRRK2 overactivity. Rab proteins, a series of small GTPases, have been identified among the proteins phosphorylated by LRRK2. These interactions may be modelled in the fruit fly Drosophila melanogaster. Using optogenetics in the fly, this project investigates the relationship between the LRRK2-G2019S and Rab10 interaction, and the speed and degree of tremor of Proboscis Extension Response (PER) by triggering a PER in fly lines of different genotypes. Significant bradykinesia in Rab10 null flies which was not recreated in flies with dopaminergic neuron Rab10RNAi suggests that the bradykinesia PER phenotype is caused by off-target effect of Rab10-KO in another tissue of the fly than the dopaminergic neurons. Over-expression of Rab10 in dopaminergic neurons of flies also expressing LRRK2-G2019S produced
    [Show full text]
  • Open Data for Differential Network Analysis in Glioma
    International Journal of Molecular Sciences Article Open Data for Differential Network Analysis in Glioma , Claire Jean-Quartier * y , Fleur Jeanquartier y and Andreas Holzinger Holzinger Group HCI-KDD, Institute for Medical Informatics, Statistics and Documentation, Medical University Graz, Auenbruggerplatz 2/V, 8036 Graz, Austria; [email protected] (F.J.); [email protected] (A.H.) * Correspondence: [email protected] These authors contributed equally to this work. y Received: 27 October 2019; Accepted: 3 January 2020; Published: 15 January 2020 Abstract: The complexity of cancer diseases demands bioinformatic techniques and translational research based on big data and personalized medicine. Open data enables researchers to accelerate cancer studies, save resources and foster collaboration. Several tools and programming approaches are available for analyzing data, including annotation, clustering, comparison and extrapolation, merging, enrichment, functional association and statistics. We exploit openly available data via cancer gene expression analysis, we apply refinement as well as enrichment analysis via gene ontology and conclude with graph-based visualization of involved protein interaction networks as a basis for signaling. The different databases allowed for the construction of huge networks or specified ones consisting of high-confidence interactions only. Several genes associated to glioma were isolated via a network analysis from top hub nodes as well as from an outlier analysis. The latter approach highlights a mitogen-activated protein kinase next to a member of histondeacetylases and a protein phosphatase as genes uncommonly associated with glioma. Cluster analysis from top hub nodes lists several identified glioma-associated gene products to function within protein complexes, including epidermal growth factors as well as cell cycle proteins or RAS proto-oncogenes.
    [Show full text]
  • Genome-Wide Association and Gene Enrichment Analyses of Meat Sensory Traits in a Crossbred Brahman-Angus
    Proceedings of the World Congress on Genetics Applied to Livestock Production, 11. 124 Genome-wide association and gene enrichment analyses of meat tenderness in an Angus-Brahman cattle population J.D. Leal-Gutíerrez1, M.A. Elzo1, D. Johnson1 & R.G. Mateescu1 1 University of Florida, Department of Animal Sciences, 2250 Shealy Dr, 32608 Gainesville, Florida, United States. [email protected] Summary The objective of this study was to identify genomic regions associated with meat tenderness related traits using a whole-genome scan approach followed by a gene enrichment analysis. Warner-Bratzler shear force (WBSF) was measured on 673 steaks, and tenderness and connective tissue were assessed by a sensory panel on 496 steaks. Animals belong to the multibreed Angus-Brahman herd from University of Florida and range from 100% Angus to 100% Brahman. All animals were genotyped with the Bovine GGP F250 array. Gene enrichment was identified in two pathways; the first pathway is involved in negative regulation of transcription from RNA polymerase II, and the second pathway groups several cellular component of the endoplasmic reticulum membrane. Keywords: tenderness, gene enrichment, regulation of transcription, cell growth, cell proliferation Introduction Identification of quantitative trait loci (QTL) for any complex trait, including meat tenderness, is the first most important step in the process of understanding the genetic architecture underlying the phenotype. Given a large enough population and a dense coverage of the genome, a genome-wide association study (GWAS) is usually successful in uncovering major genes and QTLs with large and medium effect on these type of traits. Several GWA studies on Bos indicus (Magalhães et al., 2016; Tizioto et al., 2013) or crossbred beef cattle breeds (Bolormaa et al., 2011b; Hulsman Hanna et al., 2014; Lu et al., 2013) were successful at identifying QTL for meat tenderness; and most of them include the traditional candidate genes µ-calpain and calpastatin.
    [Show full text]
  • ADHD) Gene Networks in Children of Both African American and European American Ancestry
    G C A T T A C G G C A T genes Article Rare Recurrent Variants in Noncoding Regions Impact Attention-Deficit Hyperactivity Disorder (ADHD) Gene Networks in Children of both African American and European American Ancestry Yichuan Liu 1 , Xiao Chang 1, Hui-Qi Qu 1 , Lifeng Tian 1 , Joseph Glessner 1, Jingchun Qu 1, Dong Li 1, Haijun Qiu 1, Patrick Sleiman 1,2 and Hakon Hakonarson 1,2,3,* 1 Center for Applied Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA; [email protected] (Y.L.); [email protected] (X.C.); [email protected] (H.-Q.Q.); [email protected] (L.T.); [email protected] (J.G.); [email protected] (J.Q.); [email protected] (D.L.); [email protected] (H.Q.); [email protected] (P.S.) 2 Division of Human Genetics, Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA 3 Department of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA * Correspondence: [email protected]; Tel.: +1-267-426-0088 Abstract: Attention-deficit hyperactivity disorder (ADHD) is a neurodevelopmental disorder with poorly understood molecular mechanisms that results in significant impairment in children. In this study, we sought to assess the role of rare recurrent variants in non-European populations and outside of coding regions. We generated whole genome sequence (WGS) data on 875 individuals, Citation: Liu, Y.; Chang, X.; Qu, including 205 ADHD cases and 670 non-ADHD controls. The cases included 116 African Americans H.-Q.; Tian, L.; Glessner, J.; Qu, J.; Li, (AA) and 89 European Americans (EA), and the controls included 408 AA and 262 EA.
    [Show full text]
  • Linkage, Whole Genome Sequence, and Biological Data Implicate Variants in RAB10 in Alzheimer’S Disease Resilience Perry G
    Ridge et al. Genome Medicine (2017) 9:100 DOI 10.1186/s13073-017-0486-1 RESEARCH Open Access Linkage, whole genome sequence, and biological data implicate variants in RAB10 in Alzheimer’s disease resilience Perry G. Ridge1†, Celeste M. Karch2†, Simon Hsu2, Ivan Arano1, Craig C. Teerlink3, Mark T. W. Ebbert1,4, Josue D. Gonzalez Murcia1, James M. Farnham3, Anna R. Damato2, Mariet Allen5, Xue Wang6, Oscar Harari2, Victoria M. Fernandez2, Rita Guerreiro7, Jose Bras7, John Hardy7, Ronald Munger8, Maria Norton9, Celeste Sassi7,10, Andrew Singleton10, Steven G. Younkin5, Dennis W. Dickson5, Todd E. Golde11, Nathan D. Price12, Nilüfer Ertekin-Taner13, Carlos Cruchaga2, Alison M. Goate14, Christopher Corcoran15, JoAnn Tschanz16, Lisa A. Cannon-Albright3,17, John S. K. Kauwe18* and for the Alzheimer’s Disease Neuroimaging Initative Abstract Background: While age and the APOE ε4 allele are major risk factors for Alzheimer’s disease (AD), a small percentage of individuals with these risk factors exhibit AD resilience by living well beyond 75 years of age without any clinical symptoms of cognitive decline. Methods: We used over 200 “AD resilient” individuals and an innovative, pedigree-based approach to identify genetic variants that segregate with AD resilience. First, we performed linkage analyses in pedigrees with resilient individuals and a statistical excess of AD deaths. Second, we used whole genome sequences to identify candidate SNPs in significant linkage regions. Third, we replicated SNPs from the linkage peaks that reduced risk for AD in an independent dataset and in a gene-based test. Finally, we experimentally characterized replicated SNPs. Results: Rs142787485 in RAB10 confers significant protection against AD (p value = 0.0184, odds ratio = 0.5853).
    [Show full text]
  • The P53mh Algorithm and Its Application in Detecting P53-Responsive Genes
    The p53MH algorithm and its application in detecting p53-responsive genes J. Hoh*†‡, S. Jin§†, T. Parrado*, J. Edington*, A. J. Levine§, and J. Ott* *Laboratories of Statistical Genetics and §Cancer Biology of The Rockefeller University, 1230 York Avenue, New York, NY 10021 Contributed by A. J. Levine, May 6, 2002 A computer algorithm, p53MH, was developed, which identifies overall binding likelihood is needed for a given gene of arbitrary putative p53 transcription factor DNA-binding sites on a genome- size. Three features have been implemented in an algorithm to wide scale with high power and versatility. With the sequences meet the above requirements: binding propensity plots, weighted from the human and mouse genomes, putative p53 DNA-binding scores, and statistical significance for most likely binding sites. elements were identified in a scan of 2,583 human genes and 1,713 This computer algorithm has been used to identify putative mouse orthologs based on the experimental data of el-Deiry et al. binding elements on a genomewide scale. The algorithm, [el-Deiry, W. S., Kern, S. E., Pietenpol, J. A., Kinzler, K. W. & p53MH, uses an optimal scoring system as an indication of the Vogelstein, B. (1992) Nat. Genet. 1, 45–49] and Funk et al. [Funk, percent similarity to the consensus. It can simultaneously screen W. D., Pak, D. T., Karas, R. H., Wright, W. E. & Shay, J. W. (1992) Mol. thousands of genes for degenerate consensus sequences in the Cell. Biol. 12, 2866–2871] (http:͞͞linkage.rockefeller.edu͞p53). The course of only a few minutes. With this, based on the available p53 DNA-binding motif consists of a 10-bp palindrome and most annotated human sequence databases, a White Page-like direc- commonly a second related palindrome linked by a spacer region.
    [Show full text]
  • RABIF/MSS4 Is a Rab-Stabilizing Holdase Chaperone Required for GLUT4 Exocytosis
    RABIF/MSS4 is a Rab-stabilizing holdase chaperone required for GLUT4 exocytosis Daniel R. Gulbransona, Eric M. Davisa, Brittany A. Demmitta,b, Yan Ouyanga, Yihong Yec, Haijia Yua,d,1, and Jingshi Shena,1 aDepartment of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO 80309; bInstitute for Behavioral Genetics, University of Colorado, Boulder, CO 80309; cLaboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892; and dJiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China Edited by Jennifer Lippincott-Schwartz, Howard Hughes Medical Institute, Ashburn, VA, and approved August 21, 2017 (received for review July 7, 2017) Rab GTPases are switched from their GDP-bound inactive confor- anabolic hormone insulin facilitates glucose uptake by acutely mation to a GTP-bound active state by guanine nucleotide exchange relocating GLUT4 from intracellular compartments to the cell factors (GEFs). The first putative GEFs isolated for Rabs are RABIF surface (6, 20, 21, 23). Upon the termination of insulin signaling, (Rab-interacting factor)/MSS4 (mammalian suppressor of Sec4) and GLUT4 is retrieved from the plasma membrane through endocy- its yeast homolog DSS4 (dominant suppressor of Sec4). However, tosis and returns to intracellular storage vesicles (6). Importantly, the biological function and molecular mechanism of these molecules the components of GLUT4 exocytosis are also involved in the remained unclear. In a genome-wide CRISPR genetic screen, we regulation of other exocytic pathways such as insulin secretion and isolated RABIF as a positive regulator of exocytosis.
    [Show full text]
  • Robles JTO Supplemental Digital Content 1
    Supplementary Materials An Integrated Prognostic Classifier for Stage I Lung Adenocarcinoma based on mRNA, microRNA and DNA Methylation Biomarkers Ana I. Robles1, Eri Arai2, Ewy A. Mathé1, Hirokazu Okayama1, Aaron Schetter1, Derek Brown1, David Petersen3, Elise D. Bowman1, Rintaro Noro1, Judith A. Welsh1, Daniel C. Edelman3, Holly S. Stevenson3, Yonghong Wang3, Naoto Tsuchiya4, Takashi Kohno4, Vidar Skaug5, Steen Mollerup5, Aage Haugen5, Paul S. Meltzer3, Jun Yokota6, Yae Kanai2 and Curtis C. Harris1 Affiliations: 1Laboratory of Human Carcinogenesis, NCI-CCR, National Institutes of Health, Bethesda, MD 20892, USA. 2Division of Molecular Pathology, National Cancer Center Research Institute, Tokyo 104-0045, Japan. 3Genetics Branch, NCI-CCR, National Institutes of Health, Bethesda, MD 20892, USA. 4Division of Genome Biology, National Cancer Center Research Institute, Tokyo 104-0045, Japan. 5Department of Chemical and Biological Working Environment, National Institute of Occupational Health, NO-0033 Oslo, Norway. 6Genomics and Epigenomics of Cancer Prediction Program, Institute of Predictive and Personalized Medicine of Cancer (IMPPC), 08916 Badalona (Barcelona), Spain. List of Supplementary Materials Supplementary Materials and Methods Fig. S1. Hierarchical clustering of based on CpG sites differentially-methylated in Stage I ADC compared to non-tumor adjacent tissues. Fig. S2. Confirmatory pyrosequencing analysis of DNA methylation at the HOXA9 locus in Stage I ADC from a subset of the NCI microarray cohort. 1 Fig. S3. Methylation Beta-values for HOXA9 probe cg26521404 in Stage I ADC samples from Japan. Fig. S4. Kaplan-Meier analysis of HOXA9 promoter methylation in a published cohort of Stage I lung ADC (J Clin Oncol 2013;31(32):4140-7). Fig. S5. Kaplan-Meier analysis of a combined prognostic biomarker in Stage I lung ADC.
    [Show full text]
  • SEC16A Is a RAB10 Effector Required for Insulin- Stimulated GLUT4 Trafficking in Adipocytes
    JCB: Article SEC16A is a RAB10 effector required for insulin- stimulated GLUT4 trafficking in adipocytes Joanne Bruno,1,3* Alexandria Brumfield,1* Natasha Chaudhary,1 David Iaea,1 and Timothy E. McGraw1,2 1Department of Biochemistry and 2Department of Cardiothoracic Surgery, Weill Cornell Medical College, New York, NY 10065 3Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY 10065 RAB10 is a regulator of insulin-stimulated translocation of the GLUT4 glucose transporter to the plasma membrane (PM) of adipocytes, which is essential for whole-body glucose homeostasis. We establish SEC16A as a novel RAB10 effector in this process. Colocalization of SEC16A with RAB10 is augmented by insulin stimulation, and SEC16A knockdown attenuates insulin-induced GLUT4 translocation, phenocopying RAB10 knockdown. We show that SEC16A and RAB10 promote insulin-stimulated mobilization of GLUT4 from a perinuclear recycling endosome/TGN compartment. We pro- pose RAB10–SEC16A functions to accelerate formation of the vesicles that ferry GLUT4 to the PM during insulin stimu- lation. Because GLUT4 continually cycles between the PM and intracellular compartments, the maintenance of elevated cell-surface GLUT4 in the presence of insulin requires accelerated biogenesis of the specialized GLUT4 transport vesicles. The function of SEC16A in GLUT4 trafficking is independent of its previously characterized activity in ER exit site forma- tion and therefore independent of canonical COPII-coated​ vesicle function. However, our data support a role for SEC23A, but not the other COP​II components SEC13, SEC23B, and SEC31, in the insulin stimulation of GLUT4 trafficking, sug- gesting that vesicles derived from subcomplexes of COPII​ coat proteins have a role in the specialized trafficking of GLUT4.
    [Show full text]
  • Molecular Network-Based Identification of Competing Endogenous Rnas and Mrna Signatures That Predict Survival in Prostate Cancer
    Xu et al. J Transl Med (2018) 16:274 https://doi.org/10.1186/s12967-018-1637-x Journal of Translational Medicine RESEARCH Open Access Molecular network‑based identifcation of competing endogenous RNAs and mRNA signatures that predict survival in prostate cancer Ning Xu1,2, Yu‑Peng Wu2, Hu‑Bin Yin1, Xue‑Yi Xue2 and Xin Gou1* Abstract Background: The aim of the study is described the regulatory mechanisms and prognostic values of diferentially expressed RNAs in prostate cancer and construct an mRNA signature that predicts survival. Methods: The RNA profles of 499 prostate cancer tissues and 52 non-prostate cancer tissues from TCGA were ana‑ lyzed. The diferential expression of RNAs was examined using the edgeR package. Survival was analyzed by Kaplan– Meier method. microRNA (miRNA), messenger RNA (mRNA), and long non-coding RNA (lncRNA) networks from the miRcode database were constructed, based on the diferentially expressed RNAs between non-prostate and prostate cancer tissues. Results: A total of 773 lncRNAs, 1417 mRNAs, and 58 miRNAs were diferentially expressed between non-prostate and prostate cancer samples. The newly constructed ceRNA network comprised 63 prostate cancer-specifc lncRNAs, 13 miRNAs, and 18 mRNAs. Three of 63 diferentially expressed lncRNAs and 1 of 18 diferentially expressed mRNAs were signifcantly associated with overall survival in prostate cancer (P value < 0.05). After the univariate and multivari‑ ate Cox regression analyses, 4 mRNAs (HOXB5, GPC2, PGA5, and AMBN) were screened and used to establish a predic‑ tive model for the overall survival of patients. Our ROC curve analysis revealed that the 4-mRNA signature performed well.
    [Show full text]
  • Genomic Variants Within Chromosome 14Q32.32 Regulate Bone Mass Through MARK3 Signaling in Osteoblasts
    Genomic variants within chromosome 14q32.32 regulate bone mass through MARK3 signaling in osteoblasts Qian Zhang, … , Charles R. Farber, Thomas L. Clemens J Clin Invest. 2021;131(7):e142580. https://doi.org/10.1172/JCI142580. Research Article Bone Biology Bone mineral density (BMD) is a highly heritable predictor of osteoporotic fracture. GWAS have identified hundreds of loci influencing BMD, but few have been functionally analyzed. In this study, we show that SNPs within a BMD locus on chromosome 14q32.32 alter splicing and expression of PAR-1a/microtubule affinity regulating kinase 3 (MARK3), a conserved serine/threonine kinase known to regulate bioenergetics, cell division, and polarity. Mice lacking Mark3 either globally or selectively in osteoblasts have increased bone mass at maturity. RNA profiling from Mark3-deficient osteoblasts suggested changes in the expression of components of the Notch signaling pathway. Mark3-deficient osteoblasts exhibited greater matrix mineralization compared with controls that was accompanied by reduced Jag1/Hes1 expression and diminished downstream JNK signaling. Overexpression of Jag1 in Mark3-deficient osteoblasts both in vitro and in vivo normalized mineralization capacity and bone mass, respectively. Together, these findings reveal a mechanism whereby genetically regulated alterations in Mark3 expression perturb cell signaling in osteoblasts to influence bone mass. Find the latest version: https://jci.me/142580/pdf The Journal of Clinical Investigation RESEARCH ARTICLE Genomic variants within chromosome 14q32.32 regulate bone mass through MARK3 signaling in osteoblasts Qian Zhang,1,2 Larry D. Mesner,3 Gina M. Calabrese,3 Naomi Dirckx,1 Zhu Li,1,2 Angela Verardo,1 Qian Yang,1 Robert J.
    [Show full text]