Your browser doesn't support javascript.
loading
The effects of pathogenic and likely pathogenic variants for inherited hemostasis disorders in 140 214 UK Biobank participants.
Stefanucci, Luca; Collins, Janine; Sims, Matthew C; Barrio-Hernandez, Inigo; Sun, Luanluan; Burren, Oliver S; Perfetto, Livia; Bender, Isobel; Callahan, Tiffany J; Fleming, Kathryn; Guerrero, Jose A; Hermjakob, Henning; Martin, Maria J; Stephenson, James; Paneerselvam, Kalpana; Petrovski, Slavé; Porras, Pablo; Robinson, Peter N; Wang, Quanli; Watkins, Xavier; Frontini, Mattia; Laskowski, Roman A; Beltrao, Pedro; Di Angelantonio, Emanuele; Gomez, Keith; Laffan, Mike; Ouwehand, Willem H; Mumford, Andrew D; Freson, Kathleen; Carss, Keren; Downes, Kate; Gleadall, Nick; Megy, Karyn; Bruford, Elspeth; Vuckovic, Dragana.
Afiliação
  • Stefanucci L; Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom.
  • Collins J; National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom.
  • Sims MC; British Heart Foundation, BHF Centre of Research Excellence, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom.
  • Barrio-Hernandez I; Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom.
  • Sun L; National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom.
  • Burren OS; Department of Haematology, Barts Health NHS Trust, London, United Kingdom.
  • Perfetto L; Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom.
  • Bender I; National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom.
  • Callahan TJ; Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Royal Hallamshire Hospital, Sheffield, United Kingdom.
  • Fleming K; Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom.
  • Guerrero JA; European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, United Kingdom.
  • Hermjakob H; Department of Public Health and Primary Care, BHF Cardiovascular Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom.
  • Martin MJ; Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom.
  • Stephenson J; European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, United Kingdom.
  • Paneerselvam K; Department of Biology and Biotechnology "C.Darwin," Sapienza University of Rome, Rome, Italy.
  • Petrovski S; Department of Biochemistry, University of Oxford, Oxford, United Kingdom.
  • Porras P; Department of Biomedical Informatics, Columbia University Irving Medical Center, New York, NY.
  • Robinson PN; School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom.
  • Wang Q; National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom.
  • Watkins X; Department of Haematology, Barts Health NHS Trust, London, United Kingdom.
  • Frontini M; European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, United Kingdom.
  • Laskowski RA; European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, United Kingdom.
  • Beltrao P; European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, United Kingdom.
  • Di Angelantonio E; European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, United Kingdom.
  • Gomez K; Centre for Genomics Research, Discovery Sciences, AstraZeneca, Cambridge, United Kingdom.
  • Laffan M; Department of Medicine, Austin Health, The University of Melbourne, Melbourne, Australia.
  • Ouwehand WH; European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, United Kingdom.
  • Mumford AD; Genomic Medicine, The Jackson Laboratory, Farmington, CT.
  • Freson K; Institute for Systems Genomics, University of Connecticut, Farmington, CT.
  • Carss K; Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom.
  • Downes K; European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, United Kingdom.
  • Gleadall N; Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom.
  • Megy K; National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom.
  • Bruford E; British Heart Foundation, BHF Centre of Research Excellence, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom.
  • Vuckovic D; Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences RILD Building, University of Exeter Medical School, Exeter, United Kingdom.
Blood ; 142(24): 2055-2068, 2023 12 14.
Article em En | MEDLINE | ID: mdl-37647632
ABSTRACT
Rare genetic diseases affect millions, and identifying causal DNA variants is essential for patient care. Therefore, it is imperative to estimate the effect of each independent variant and improve their pathogenicity classification. Our study of 140 214 unrelated UK Biobank (UKB) participants found that each of them carries a median of 7 variants previously reported as pathogenic or likely pathogenic. We focused on 967 diagnostic-grade gene (DGG) variants for rare bleeding, thrombotic, and platelet disorders (BTPDs) observed in 12 367 UKB participants. By association analysis, for a subset of these variants, we estimated effect sizes for platelet count and volume, and odds ratios for bleeding and thrombosis. Variants causal of some autosomal recessive platelet disorders revealed phenotypic consequences in carriers. Loss-of-function variants in MPL, which cause chronic amegakaryocytic thrombocytopenia if biallelic, were unexpectedly associated with increased platelet counts in carriers. We also demonstrated that common variants identified by genome-wide association studies (GWAS) for platelet count or thrombosis risk may influence the penetrance of rare variants in BTPD DGGs on their associated hemostasis disorders. Network-propagation analysis applied to an interactome of 18 410 nodes and 571 917 edges showed that GWAS variants with large effect sizes are enriched in DGGs and their first-order interactors. Finally, we illustrate the modifying effect of polygenic scores for platelet count and thrombosis risk on disease severity in participants carrying rare variants in TUBB1 or PROC and PROS1, respectively. Our findings demonstrate the power of association analyses using large population datasets in improving pathogenicity classifications of rare variants.
Assuntos

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Trombose / Estudo de Associação Genômica Ampla Tipo de estudo: Prognostic_studies Limite: Humans Idioma: En Revista: Blood Ano de publicação: 2023 Tipo de documento: Article País de afiliação: Reino Unido

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Trombose / Estudo de Associação Genômica Ampla Tipo de estudo: Prognostic_studies Limite: Humans Idioma: En Revista: Blood Ano de publicação: 2023 Tipo de documento: Article País de afiliação: Reino Unido