12 research outputs found

    A SINE-VNTR-Alu in the LRIG2 Promoter Is Associated with Gene Expression at the Locus

    Get PDF
    The hominid SINE-VNTR-Alu (SVA) retrotransposons represent a repertoire of genomic variation which could have significant effects on genome function. A human-specific SVA in the promoter region of the gene leucine-rich repeats and immunoglobulin-like domains 2 (LRIG2), which we termed SVA_LRIG2, is a common retrotransposon insertion polymorphism (RIP), defined as an element which is polymorphic for its presence or absence in the genome. We hypothesised that this RIP might be associated with differential levels of expression of LRIG2. The RIP genotype of SVA_LRIG2 was determined in a subset of frontal cortex DNA samples from the North American Brain Expression Consortium (NABEC) cohort and was imputed for a larger set of that cohort. Utilising available frontal cortex total RNA-seq and CpG methylation data for this cohort, we observed that increased allele dosage of SVA_LRIG2 was non-significantly associated with a decrease in transcription from the region and significantly associated with increased methylation of the CpG probe nearest to SVA_LRIG2, i.e., SVA_LRIG2 is a significant methylation quantitative trait loci (mQTL) at the LRIG2 locus. These data are consistent with SVA_LRIG2 being a transcriptional regulator, which in part may involve epigenetic modulation.</jats:p

    Identification of novel risk loci, causal insights, and heritable risk for Parkinson's disease: a meta-analysis of genome-wide association studies

    Get PDF
    Background Genome-wide association studies (GWAS) in Parkinson's disease have increased the scope of biological knowledge about the disease over the past decade. We aimed to use the largest aggregate of GWAS data to identify novel risk loci and gain further insight into the causes of Parkinson's disease. Methods We did a meta-analysis of 17 datasets from Parkinson's disease GWAS available from European ancestry samples to nominate novel loci for disease risk. These datasets incorporated all available data. We then used these data to estimate heritable risk and develop predictive models of this heritability. We also used large gene expression and methylation resources to examine possible functional consequences as well as tissue, cell type, and biological pathway enrichments for the identified risk factors. Additionally, we examined shared genetic risk between Parkinson's disease and other phenotypes of interest via genetic correlations followed by Mendelian randomisation. Findings Between Oct 1, 2017, and Aug 9, 2018, we analysed 7·8 million single nucleotide polymorphisms in 37 688 cases, 18 618 UK Biobank proxy-cases (ie, individuals who do not have Parkinson's disease but have a first degree relative that does), and 1·4 million controls. We identified 90 independent genome-wide significant risk signals across 78 genomic regions, including 38 novel independent risk signals in 37 loci. These 90 variants explained 16–36% of the heritable risk of Parkinson's disease depending on prevalence. Integrating methylation and expression data within a Mendelian randomisation framework identified putatively associated genes at 70 risk signals underlying GWAS loci for follow-up functional studies. Tissue-specific expression enrichment analyses suggested Parkinson's disease loci were heavily brain-enriched, with specific neuronal cell types being implicated from single cell data. We found significant genetic correlations with brain volumes (false discovery rate-adjusted p=0·0035 for intracranial volume, p=0·024 for putamen volume), smoking status (p=0·024), and educational attainment (p=0·038). Mendelian randomisation between cognitive performance and Parkinson's disease risk showed a robust association (p=8·00 × 10−7). Interpretation These data provide the most comprehensive survey of genetic risk within Parkinson's disease to date, to the best of our knowledge, by revealing many additional Parkinson's disease risk loci, providing a biological context for these risk factors, and showing that a considerable genetic component of this disease remains unidentified. These associations derived from European ancestry datasets will need to be followed-up with more diverse data. Funding The National Institute on Aging at the National Institutes of Health (USA), The Michael J Fox Foundation, and The Parkinson's Foundation (see appendix for full list of funding sources)

    Genome-wide structural variant analysis identifies risk loci for non-Alzheimer’s dementias

    Get PDF
    We characterized the role of structural variants, a largely unexplored type of genetic variation, in two non-Alzheimer’s dementias, namely Lewy body dementia (LBD) and frontotemporal dementia (FTD)/amyotrophic lateral sclerosis (ALS). To do this, we applied an advanced structural variant calling pipeline (GATK-SV) to short-read whole-genome sequence data from 5,213 European-ancestry cases and 4,132 controls. We discovered, replicated, and validated a deletion in TPCN1 as a novel risk locus for LBD and detected the known structural variants at the C9orf72 and MAPT loci as associated with FTD/ALS. We also identified rare pathogenic structural variants in both LBD and FTD/ALS. Finally, we assembled a catalog of structural variants that can be mined for new insights into the pathogenesis of these understudied forms of dementia

    Mitochondria function associated genes contribute to Parkinson’s Disease risk and later age at onset

    No full text
    Abstract Mitochondrial dysfunction has been implicated in the etiology of monogenic Parkinson’s disease (PD). Yet the role that mitochondrial processes play in the most common form of the disease; sporadic PD, is yet to be fully established. Here, we comprehensively assessed the role of mitochondrial function-associated genes in sporadic PD by leveraging improvements in the scale and analysis of PD GWAS data with recent advances in our understanding of the genetics of mitochondrial disease. We calculated a mitochondrial-specific polygenic risk score (PRS) and showed that cumulative small effect variants within both our primary and secondary gene lists are significantly associated with increased PD risk. We further reported that the PRS of the secondary mitochondrial gene list was significantly associated with later age at onset. Finally, to identify possible functional genomic associations we implemented Mendelian randomization, which showed that 14 of these mitochondrial function-associated genes showed functional consequence associated with PD risk. Further analysis suggested that the 14 identified genes are not only involved in mitophagy, but implicate new mitochondrial processes. Our data suggests that therapeutics targeting mitochondrial bioenergetics and proteostasis pathways distinct from mitophagy could be beneficial to treating the early stage of PD.Additional information International Parkinson’s Disease Genomics Consortium (IPDGC) Members A. Noyce13, A. Tucci14, B. Middlehurst1, D. Kia15, M. Tan16, H. Houlden14, H. R. Morris16, H. Plun-Favreau14, P. Holmans17, J. Hardy14, D. Trabzuni14,18, J. Bras19, K. Mok14, K. Kinghorn20, N. Wood15, P. Lewis21, R. Guerreiro14,19, R. Lovering22, L. R’Bibo14, M. Rizig14, V. Escott-Price22,23, V. Chelban14, T. Foltynie6, N. Williams24, A. Brice25, F. Danjou25, S. Lesage25, M. Martinez26, A. Giri27,28, C. Schulte27,28, K. Brockmann27,28, J. Simón-Sánchez27,28, P. Heutink27,28, P. Rizzu28, M. Sharma29, T. Gasser27,28, A. Nicolas2, M. Cookson2, F. Faghri2,30, D. Hernandez2, J. Shulman31,32, L. Robak33, S. Lubbe34, S. Finkbeiner35,36,37, N. Mencacci38, C. Lungu39, S. Scholz40, X. Reed2, H. Leonard2, G. Rouleau7, L. Krohan41, J. van Hilten42, J. Marinus42, A. Adarmes-Gómez43, M. Aguilar44, I. Alvarez44, V. Alvarez45, F. Javier Barrero46, J. Bergareche Yarza47, I. Bernal-Bernal43, M. Blazquez45, M. Bonilla-Toribio Bernal43, M. Boungiorno44, Dolores Buiza-Rueda43, A. Cámara48, M. Carcel44, F. Carrillo43, M. Carrión-Claro43, D. Cerdan49, J. Clarimón50,51, Y. Compta48, M. Diez-Fairen44, O. Dols-Icardo50,51, J. Duarte49, R. l. Duran52, F. Escamilla-Sevilla53, M. Ezquerra48, M. Fernández48, R. Fernández-Santiago48, C. Garcia45, P. García-Ruiz54, P. Gómez-Garre43, M. Gomez Heredia55, I. Gonzalez-Aramburu56, A. Gorostidi Pagola57, J. Hoenicka58, J. Infante51,56, S. Jesús43, A. Jimenez-Escrig59, J. Kulisevsky51,60, M. Labrador-Espinosa43, J. Lopez-Sendon59, A. López de Munain Arregui59, D. Macias43, I. Martínez Torres61, J. Marín51,60, M. Jose Marti48, J. Martínez-Castrillo59, C. Méndez-del-Barrio43, M. Menéndez González43, A. Mínguez53, P. Mir43, E. Mondragon Rezola57, E. Muñoz48, J. Pagonabarraga51,60, P. Pastor44, F. Perez Errazquin55, T. Periñán-Tocino43, J. Ruiz-Martínez57, C. Ruz52, A. Sanchez Rodriguez56, M. Sierra56, E. Suarez-Sanmartin4, C. Tabernero59, J. Pablo Tartari44, C. Tejera-Parrado43, E. Tolosa48, F. Valldeoriola48, L. Vargas-González43, L. Vela62, F. Vives52, A. Zimprich63, L. Pihlstrom64, P. Taba65, K. Majamaa66,67, A. Siitonen66, N. Okubadejo68, O. Ojo68 1 Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK 2Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA 3Department of Medical and Molecular Genetics, King’s College London School of Basic and Medical Biosciences, London, SE1 9RT, UK 4Clinical Genetics Unit, Guys and St. Thomas’ NHS Foundation Trust, London, SE1 9RT, UK 5Departamento de Ingeniería de la Información y las Comunicaciones, Universidad de Murcia, 30100, Murcia, Spain 6Department of Neurodegenerative Disease, UCL Institute of Neurology, 10-12 Russell Square House, London, UK 7Montreal Neurological Institute, McGill University, Montréal, QC, Canada 8Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada 9Department of Human Genetics, McGill University, Montréal, QC, Canada 10Data Tecnica International, Glen Echo, MD, 20812, USA 11The Perron Institute for Neurological and Translational Science, 8 Verdun Street, Nedlands, WA, 6009, Australia 12Centre for Comparative Genomics, Murdoch University, Murdoch, 6150, Australia 13Preventive Neurology Unit, Wolfson Institute of Preventive Medicine, QMUL, London, UK 14Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK 15UCL Genetics Institute; and Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK 16Department of Clinical Neuroscience, University College London, London, UK 17Biostatistics &amp; Bioinformatics Unit, Institute of Psychological Medicine and Clinical Neuroscience, MRC Centre for Neuropsychiatric Genetics &amp; Genomics, Cardiff, UK 18Department of Genetics, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia 19UK Dementia Research Institute at UCL and Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK 20Institute of Healthy Ageing, University College London, London, UK 21University of Reading, Reading, UK 22University College London, London, UK 23MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff, UK 24Cardiff University School of Medicine, Cardiff, UK 25Institut du Cerveau et de la Moelle épinière, ICM, Inserm U 1127, CNRS, UMR 7225, Sorbonne Universités, UPMC University Paris 06, UMR S 1127, AP-HP, Pitié-Salpêtrière Hospital, Paris, France 26INSERM UMR 1220; and Paul Sabatier University, Toulouse, France 27Department for Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany 28DZNE, German Center for Neurodegenerative Diseases, Tübingen, Germany 29Centre for Genetic Epidemiology, Institute for Clinical Epidemiology and Applied Biometry, University of Tubingen, Tubingen, Germany 30Department of Computer Science, University of Illinois at Urbana-Champaign, Urbana, IL, USA 31Departments of Neurology, Neuroscience, and Molecular &amp; Human Genetics, Baylor College of Medicine, Houston, TX, USA 32Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, USA 33Baylor College of Medicine, Houston, TX, USA 34Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA 35Departments of Neurology and Physiology, University of California, San Francisco, CA, USA 36Gladstone Institute of Neurological Disease, San Francisco, CA, USA 37Taube/Koret Center for Neurodegenerative Disease Research, San Francisco, CA, USA) 38 (Northwestern University Feinberg School of Medicine, Chicago, IL, USA) 39 (National Institutes of Health Division of Clinical Research, NINDS, National Institutes of Health, Bethesda, MD, USA) 40Neurodegenerative Diseases Research Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA 41Department of Human Genetics, McGill University, Montréal, QC H3A 0G4, Canada 42Department of Neurology, Leiden University Medical Center, Leiden, Netherlands 43Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/ Universidad de Sevilla, Seville, Spain 44Fundació Docència i Recerca Mútua de Terrassa and Movement Disorders Unit, Department of Neurology, University Hospital Mutua de Terrassa, Terrassa, Barcelona, Spain 45Hospital Universitario Central de Asturias, Oviedo, Spain 46Hospital Universitario Parque Tecnologico de la Salud, Granada, Spain 47Instituto de Investigación Sanitaria Biodonostia, San Sebastián, Spain 48Hospital Clinic de Barcelona, Barcelona, Spain 49Hospital General de Segovia, Segovia, Spain 50Memory Unit, Department of Neurology, IIB Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain 51Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain 52Centro de Investigacion Biomedica, Universidad de Granada, Granada, Spain 53Hospital Universitario Virgen de las Nieves, Instituto de Investigación Biosanitaria de Granada, Granada, Spain 54Instituto de Investigación Sanitaria Fundación Jiménez Díaz, Madrid, Spain 55Hospital Universitario Virgen de la Victoria, Malaga, Spain 56Hospital Universitario Marqués de Valdecilla-IDIVAL, Santander, Spain 57Instituto de Investigación Sanitaria Biodonostia, San Sebastián, Spain 58Institut de Recerca Sant Joan de Déu, Barcelona, Spain 59Hospital Universitario Ramón y Cajal Madrid, Madrid, Spain 60Movement Disorders Unit, Department of Neurology, IIB Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain 61Department of Neurology, Instituto de Investigación Sanitaria La Fe, Hospital Universitario y Politécnico La Fe, Valencia, Spain 62Department of Neurology, Hospital Universitario Fundación Alcorcón, Madrid, Spain 63Department of Neurology, Medical University of Vienna, Vienna, Austria 64Department of Neurology, Oslo University Hospital, Oslo, Norway 65Department of Neurology and Neurosurgery, University of Tartu, Tartu, Estonia 66Institute of Clinical Medicine, Department of Neurology, University of Oulu, Oulu, Finland 67Department of Neurology and Medical Research Center, Oulu University Hospital, Oulu, Finland 68University of Lagos, Yaba, Lagos State, Nigeri

    The Parkinson's Disease Genome‐Wide Association Study Locus Browser

    No full text
    International Parkinson's Disease Genomics Consortium (IPDGC).[Background] Parkinson's disease (PD) is a neurodegenerative disease with an often complex component identifiable by genome‐wide association studies. The most recent large‐scale PD genome‐wide association studies have identified more than 90 independent risk variants for PD risk and progression across more than 80 genomic regions. One major challenge in current genomics is the identification of the causal gene(s) and variant(s) at each genome‐wide association study locus. The objective of the current study was to create a tool that would display data for relevant PD risk loci and provide guidance with the prioritization of causal genes and potential mechanisms at each locus.[Methods] We included all significant genome‐wide signals from multiple recent PD genome‐wide association studies including themost recent PD risk genome‐wide association study, age‐at‐onset genome‐wide association study, progression genome‐wide association study, and Asian population PD risk genome‐wide association study. We gathered data for all genes 1 Mb up and downstream of each variant to allow users to assess which gene(s) are most associated with the variant of interest based on a set of self‐ranked criteria. Multiple databases were queried for each gene to collect additional causal data.[Results] We created a PD genome‐wide association study browser tool (https://pdgenetics.shinyapps.io/GWASBrowser/) to assist the PD research community with the prioritization of genes for follow‐up functional studies to identify potential therapeutic targets.[Conclusions] Our PD genome‐wide association study browser tool provides users with a useful method of identifying potential causal genes at all known PD risk loci from large‐scale PD genome‐wide association studies. We plan to update this tool with new relevant data as sample sizes increase and new PD risk loci are discovered. © 2020 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society. This article has been contributed to by US Government employees and their work is in the public domain in the USA.This work was supported in part by the Intramural Research Programs of the National Institute of Neurological Disorders and Stroke (NINDS), the National Institute on Aging (NIA), and the National Institute of Environmental Health Sciences, both part of the National Institutes of Health, Department of Health and Human Services (project numbers 1ZIA‐NS003154, Z01‐AG000949‐02, and Z01‐ES101986). We thank the research participants and employees of 23andMe for making this work possible. C.W. is supported by the UK Dementia Research Institute funded by the Medical Research Council (MRC), Alzheimer's Society and Alzheimer's Research UK. C.S. is supported by the Ser Cymru II program, which is partly funded by Cardiff University and the European Regional Development Fund through the Welsh Government. Data were generated as part of the PsychENCODE Consortium supported by: U01MH103339, U01MH103365, U01MH103392, U01MH103340, U01MH103346, R01MH105472, R01MH094714, R01MH105898, R21MH102791, R21MH105881, R21MH103877, and P50MH106934 awarded to Schahram Akbarian (Icahn School of Medicine at Mount Sinai), Gregory Crawford (Duke), Stella Dracheva (Icahn School of Medicine at Mount Sinai), Peggy Farnham (USC), Mark Gerstein (Yale), Daniel Geschwind (UCLA), Thomas M. Hyde (LIBD), Andrew Jaffe (LIBD), James A. Knowles (USC), Chunyu Liu (UIC), Dalila Pinto (Icahn School of Medicine at Mount Sinai), Nenad Sestan (Yale), Pamela Sklar (Icahn School of Medicine at Mount Sinai), Matthew State (UCSF), Patrick Sullivan (UNC), Flora Vaccarino (Yale), Sherman Weissman (Yale), Kevin White (UChicago), and Peter Zandi (JHU). The Genotype‐Tissue Expression (GTEx) Project was supported by the Common Fund of the Office of the Director of the National Institutes of Health, and by NCI, NHGRI, NHLBI, NIDA, NIMH, and NINDS. The data used for the analyses described in this article were obtained from the GTEx Portal on February 12, 2020. Molecular data for the Trans‐Omics in Precision Medicine (TOPMed) program was supported by the National Heart, Lung, and Blood Institute (NHLBI). Genome sequencing for “NHLBI TOPMed: Atherosclerosis Risk in Communities (ARIC)” (phs001211.v2.p2) was performed at the Broad Institute of MIT and Harvard (3R01HL092577‐06S1)and at the Baylor Human Genome Sequencing Center (3U54HG003273‐12S2, HHSN268201500015C). Genome sequencing for the “NHLBI TOPMed: Cleveland Clinic Atrial Fibrillation (CCAF) Study” (phs001189.v1.p1) was performed at the Broad Institute of MIT and Harvard (3R01HL092577‐06S1). Genome sequencing for “NHLBI TOPMed: Trans‐Omics for Precision Medicine (TOPMed) Whole Genome Sequencing Project: Cardiovascular Health Study (phs001368.v1.p1) was performed at the Baylor Human Genome Sequencing Center (3U54HG003273‐12S2, HHSN268201500015C). Genome sequencing for “NHLBI TOPMed: Partners HealthCare Biobank” (phs001024.v3.p1) was performed at the Broad Institute of MIT and Harvard (3R01HL092577‐06S1). Genome sequencing for “NHLBI TOPMed: Whole Genome Sequencing of Venous Thromboembolism (WGS of VTE)” (phs001402.v1.p1) was performed at the Baylor Human Genome Sequencing Center (3U54HG003273‐12S2, HHSN268201500015C). Genome sequencing for “NHLBI TOPMed: Novel Risk Factors for the Development of Atrial Fibrillation in Women” (phs001040.v3.p1) was performed at the Broad Institute of MIT and Harvard (3R01HL092577‐06S1). Genome sequencing for “NHLBI TOPMed: The Genetics and Epidemiology of Asthma in Barbados” (phs001143.v2.p1) was performed by Illumina Genomic Services (3R01HL104608‐04S1). Genome sequencing for “NHLBI TOPMed: The Vanderbilt Genetic Basis of Atrial Fibrillation” (phs001032.v4.p2) was performed at the Broad Institute of MIT and Harvard (3R01HL092577‐06S1). Genome sequencing for “NHLBI TOPMed: Heart and Vascular Health Study (HVH)” (phs000993.v3.p2) was performed at the Broad Institute of MIT and Harvard (3R01HL092577‐06S1) and at the Baylor Human Genome Sequencing Center (3U54HG003273‐12S2, HHSN268201500015C). Genome sequencing for “NHLBI TOPMed: Genetic Epidemiology of COPD (COPDGene)” (phs000951.v3.p3) was performed at the University of Washington Northwest Genomics Center (3R01HL089856‐08S1) and at the Broad Institute of MIT and Harvard (HHSN268201500014C). Genome sequencing for “NHLBI TOPMed: The Vanderbilt Atrial Fibrillation Ablation Registry” (phs000997.v3.p2) was performed at the Broad Institute of MIT and Harvard (3U54HG003067‐12S2, 3U54HG003067‐13S1). Genome sequencing for “NHLBI TOPMed: The Jackson Heart Study” (phs000964.v3.p1) was performed at the University of Washington Northwest Genomics Center (HHSN268201100037C). Genome sequencing for “NHLBI TOPMed: Genetics of Cardiometabolic Health in the Amish” (phs000956.v3.p1) was performed at the Broad Institute of MIT and Harvard (3R01HL121007‐01S1). Genome sequencing for “NHLBI TOPMed: Massachusetts General Hospital Atrial Fibrillation (MGH AF) Study” (phs001062.v3.p2) was performed at the Broad Institute of MIT and Harvard (3R01HL092577‐06S1, 3U54HG003067‐12S2, 3U54HG003067‐13S1, 3UM1HG008895‐01S2). Genome sequencing for “NHLBI TOPMed: The Framingham Heart Study” (phs000974.v3.p2) was performed at the Broad Institute of MIT and Harvard (3U54HG003067‐12S2). Core support including centralized genomic read mapping and genotype calling, along with variant quality metrics and filtering, were provided by the TOPMed Informatics Research Center (3R01HL‐117626‐02S1; contract HHSN268201800002I). Core support including phenotype harmonization, data management, sample‐identity QC, and general program coordination were provided by the TOPMed Data Coordinating Center (R01HL‐120393; U01HL‐120393; contract HHSN268201800001I). We gratefully acknowledge the studies and participants who provided biological samples and data for TOPMed. The Atherosclerosis Risk in Communities study has been funded in whole or in part with federal funds from the National Heart, Lung, and Blood Institute, National Institute of Health, Department of Health and Human Services, under contract numbers (HHSN268201700001I, HHSN268201700002I, HHSN268201700003I, HHSN268201700004I, and HHSN268201700005I). The authors thank the staff and participants of the ARIC study for their important contributions. The research reported in this article was supported by grants from the National Institutes of Health (NIH) National Heart, Lung, and Blood Institute grants R01 HL090620 and R01 HL111314, the NIH National Center for Research Resources for Case Western Reserve University and Cleveland Clinic Clinical and Translational Science Award (CTSA) UL1‐RR024989, the Department of Cardiovascular Medicine philanthropic research fund, Heart and Vascular Institute, Cleveland Clinic, the Fondation Leducq grant 07‐CVD 03, and the Atrial Fibrillation Innovation Center, state of Ohio. This research was supported by contracts HHSN268201200036C, HHSN268200800007C, N01‐HC85079, N01‐HC‐85080, N01‐HC‐85081, N01‐HC‐85082, N01‐HC‐85083, N01‐HC‐85084, N01‐HC‐85085, N01‐HC‐85086, N01‐HC‐35129, N01‐HC‐15103, N01‐HC‐55222, N01‐HC‐75150, N01‐HC‐45133, and N01‐ HC‐85239; grant numbers U01 HL080295 and U01 HL130014 from the National Heart, Lung, and Blood Institute, and R01 AG023629 from the National Institute on Aging, with additional contribution from the National Institute of Neurological Disorders and Stroke. A full list of principal CHS investigators and institutions can be found at https://chs-nhlbi.org/pi. This article was not prepared in collaboration with CHS investigators and does not necessarily reflect the opinions or views of CHS or the NHLBI. We thank the Broad Institute for generating high‐quality sequence data supported by NHLBI grant 3R01HL092577‐06S1 to Dr. Patrick Ellinor. Funded in part by grants from the National Institutes of Health, National Heart, Lung, and Blood Institute (HL66216 and HL83141), and the National Human Genome Research Institute (HG04735). The Women's Genome Health Study (WGHS) is supported by HL 043851 and HL099355 from the National Heart, Lung, and Blood Institute and CA 047988 from the National Cancer Institute, the Donald W. Reynolds Foundation with collaborative scientific support and funding for genotyping provided by Amgen. AF end‐point confirmation was supported by HL‐093613 and a grant from the Harris Family Foundation and Watkin's Foundation. The Genetics and Epidemiology of Asthma in Barbados is supported by National Institutes of Health (NIH) National Heart, Lung, and Blood Institute TOPMed (R01 HL104608‐S1), and R01 AI20059, K23 HL076322, and RC2 HL101651. The research reported in this article was supported by grants from the American Heart Association to Dr. Darbar (EIA 0940116N), and grants from the National Institutes of Health (NIH) to Dr. Darbar (HL092217), and Dr. Roden (U19 HL65962, and UL1 RR024975). This project was also supported by a CTSA award (UL1TR000445) from the National Center for Advancing Translational Sciences. Its contents are solely the responsibility of the authors and do not necessarily represent official views of the National Center for Advancing Translational Sciences of the NIH. The research reported in this article was supported by grants HL068986, HL085251, HL095080, and HL073410 from the National Heart, Lung, and Blood Institute. This article was not prepared in collaboration with Heart and Vascular Health (HVH) Study investigators and does not necessarily reflect the opinions or views of the HVH Study or the NHLBI. This research used data generated by the COPDGene study, which was supported by NIH grants U01 HL089856 and U01 HL089897. The COPDGene project is also supported by the COPD Foundation through contributions made by an Industry Advisory Board composed of Pfizer, AstraZeneca, Boehringer Ingelheim, Novartis, and Sunovion. Centralized read mapping and genotype calling, along with variant quality metrics and filtering were provided by the TOPMed Informatics Research Center (3R01HL‐117626‐02S1; contract HHSN268201800002I). Phenotype harmonization, data management, sample‐identity QC, and general study coordination were provided by the TOPMed Data Coordinating Center (3R01HL‐120393‐02S1; contract HHSN268201800001I). We gratefully acknowledge the studies and participants who provided biological samples and data for TOPMed. This study is part of the Centers for Common Disease Genomics (CCDG) program, a large‐scale genome sequencing effort to identify rare risk and protective alleles that contribute to a range of common disease phenotypes. The CCDG program is funded by the National Human Genome Research Institute (NHGRI) and the National Heart, Lung, and Blood Institute (NHLBI). Sequencing was completed at the Human Genome Sequencing Center at Baylor College of Medicine under NHGRI grant UM1 HG008898. The research reported in this article was supported by grants from the American Heart Association to Dr. Shoemaker (11CRP742009) and Dr. Darbar (EIA 0940116N), and grants from the National Institutes of Health (NIH) to Dr. Darbar (R01 HL092217) and Dr. Roden (U19 HL65962 and UL1 RR024975). The project was also supported by a CTSA award (UL1 TR00045) from the National Center for Advancing Translational Sciences. Its contents are solely the responsibility of the authors and do not necessarily represent official views of the National Center for Advancing Translational Sciences or the NIH. The Jackson Heart Study (JHS) is supported and conducted in collaboration with Jackson State University (HHSN268201800013I), Tougaloo College (HHSN268201800014I), the Mississippi State Department of Health (HHSN268201800015I/HHSN26800001), and the University of Mississippi Medical Center (HHSN268201800010I, HHSN268201800011I, and HHSN268201800012I) contracts from the National Heart, Lung, and Blood Institute (NHLBI) and the National Institute for Minority Health and Health Disparities (NIMHD). The authors also thank the staffs and participants of the JHS. The Amish studies on which these data are based were supported by NIH grants R01 AG18728, U01 HL072515, R01 HL088119, R01 HL121007, and P30 DK072488. See publication PMID: 18440328. The research reported in this article was supported by NIH grants K23HL071632, K23HL114724, R21DA027021, R01HL092577, R01HL092577S1, R01HL104156, K24HL105780, and U01HL65962. The research has also been supported by an Established Investigator Award from the American Heart Association (13EIA14220013) and by support from the Fondation Leducq (14CVD01). This article was not prepared in collaboration with MGH AF Study investigators and does not necessarily reflect the opinions or views of the MGH AF Study investigators or the NHLBI. The Framingham Heart Study is conducted and supported by the National Heart, Lung, and Blood Institute (NHLBI) in collaboration with Boston University (contract nos. N01‐HC‐25195, HHSN268201500001I, and 75N92019D00031). This article was not prepared in collaboration with investigators of the Framingham Heart Study and does not necessarily reflect the opinions or views of the Framingham Heart Study, Boston University, or NHLBI

    The Parkinson's DiseaseGenome-WideAssociation Study Locus Browser

    No full text
    Background: Parkinson's disease (PD) is a neurodegenerative disease with an often complex component identifiable by genome-wide association studies. The most recent large-scale PD genome-wide association studies have identified more than 90 independent risk variants for PD risk and progression across more than 80 genomic regions. One major challenge in current genomics is the identification of the causal gene(s) and variant(s) at each genome-wide association study locus. The objective of the current study was to create a tool that would display data for relevant PD risk loci and provide guidance with the prioritization of causal genes and potential mechanisms at each locus. Methods: We included all significant genome-wide signals from multiple recent PD genome-wide association studies including themost recent PD risk genome-wide association study, age-at-onset genome-wide association study, progression genome-wide association study, and Asian population PD risk genome-wide association study. We gathered data for all genes 1 Mb up and downstream of each variant to allow users to assess which gene(s) are most associated with the variant of interest based on a set of self-ranked criteria. Multiple databases were queried for each gene to collect additional causal data. Results: We created a PD genome-wide association study browser tool (https://pdgenetics.shinyapps.io/GWASBrowser/) to assist the PD research community with the prioritization of genes for follow-up functional studies to identify potential therapeutic targets. Conclusions: Our PD genome-wide association study browser tool provides users with a useful method of identifying potential causal genes at all known PD risk loci from large-scale PD genome-wide association studies. We plan to update this tool with new relevant data as sample sizes increase and new PD risk loci are discovered

    Moving beyond neurons:the role of cell type-specific gene regulation in Parkinson’s disease heritability

    No full text
    Abstract Parkinson’s disease (PD), with its characteristic loss of nigrostriatal dopaminergic neurons and deposition of α-synuclein in neurons, is often considered a neuronal disorder. However, in recent years substantial evidence has emerged to implicate glial cell types, such as astrocytes and microglia. In this study, we used stratified LD score regression and expression-weighted cell-type enrichment together with several brain-related and cell-type-specific genomic annotations to connect human genomic PD findings to specific brain cell types. We found that PD heritability attributable to common variation does not enrich in global and regional brain annotations or brain-related cell-type-specific annotations. Likewise, we found no enrichment of PD susceptibility genes in brain-related cell types. In contrast, we demonstrated a significant enrichment of PD heritability in a curated lysosomal gene set highly expressed in astrocytic, microglial, and oligodendrocyte subtypes, and in LoF-intolerant genes, which were found highly expressed in almost all tested cellular subtypes. Our results suggest that PD risk loci do not lie in specific cell types or individual brain regions, but rather in global cellular processes detectable across several cell types

    An international virtual hackathon to build tools for the analysis of structural variants within species ranging from coronaviruses to vertebrates

    No full text
    In October 2020, 62 scientists from nine nations worked together remotely in the Second Baylor College of Medicine &amp; DNAnexus hackathon, focusing on different related topics on Structural Variation, Pan-genomes, and SARS-CoV-2 related research. The overarching focus was to assess the current status of the field and identify the remaining challenges. Furthermore, how to combine the strengths of the different interests to drive research and method development forward. Over the four days, eight groups each designed and developed new open-source methods to improve the identification and analysis of variations among species, including humans and SARS-CoV-2. These included improvements in SV calling, genotyping, annotations and filtering. Together with advancements in benchmarking existing methods. Furthermore, groups focused on the diversity of SARS-CoV-2. Daily discussion summary and methods are available publicly at https://github.com/collaborativebioinformatics provides valuable insights for both participants and the research community.</p

    An international virtual hackathon to build tools for the analysis of structural variants within species ranging from coronaviruses to vertebrates

    No full text
    In October 2020, 62 scientists from nine nations worked together remotely in the Second Baylor College of Medicine & DNAnexus hackathon, focusing on different related topics on Structural Variation, Pan-genomes, and SARS-CoV-2 related research.   The overarching focus was to assess the current status of the field and identify the remaining challenges. Furthermore, how to combine the strengths of the different interests to drive research and method development forward. Over the four days, eight groups each designed and developed new open-source methods to improve the identification and analysis of variations among species, including humans and SARS-CoV-2. These included improvements in SV calling, genotyping, annotations and filtering. Together with advancements in benchmarking existing methods. Furthermore, groups focused on the diversity of SARS-CoV-2. Daily discussion summary and methods are available publicly at https://github.com/collaborativebioinformatics provides valuable insights for both participants and the research community
    corecore