Your browser doesn't support javascript.
loading
Understanding the genetic complexity of puberty timing across the allele frequency spectrum.
Kentistou, Katherine A; Kaisinger, Lena R; Stankovic, Stasa; Vaudel, Marc; de Oliveira, Edson M; Messina, Andrea; Walters, Robin G; Liu, Xiaoxi; Busch, Alexander S; Helgason, Hannes; Thompson, Deborah J; Santon, Federico; Petricek, Konstantin M; Zouaghi, Yassine; Huang-Doran, Isabel; Gudbjartsson, Daniel F; Bratland, Eirik; Lin, Kuang; Gardner, Eugene J; Zhao, Yajie; Jia, Raina; Terao, Chikashi; Riggan, Margie; Bolla, Manjeet K; Yazdanpanah, Mojgan; Yazdanpanah, Nahid; Bradfield, Jonath P; Broer, Linda; Campbell, Archie; Chasman, Daniel I; Cousminer, Diana L; Franceschini, Nora; Franke, Lude H; Girotto, Giorgia; He, Chunyan; Järvelin, Marjo-Riitta; Joshi, Peter K; Kamatani, Yoichiro; Karlsson, Robert; Luan, Jian'an; Lunetta, Kathryn L; Mägi, Reedik; Mangino, Massimo; Medland, Sarah E; Meisinger, Christa; Noordam, Raymond; Nutile, Teresa; Concas, Maria Pina; Polasek, Ozren; Porcu, Eleonora.
Afiliación
  • Kentistou KA; MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Box 285 Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK.
  • Kaisinger LR; MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Box 285 Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK.
  • Stankovic S; MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Box 285 Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK.
  • Vaudel M; Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, NO-5020, Bergen, Norway.
  • de Oliveira EM; Department of Genetics and Bioinformatics, Health Data and Digitalization, Norwegian Institute of Public Health, NO-0213, Oslo, Norway.
  • Messina A; University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK.
  • Walters RG; Division of Endocrinology, Diabetology, and Metabolism, Lausanne University Hospital, 1011 Lausanne, Switzerland.
  • Liu X; Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK.
  • Busch AS; MRC Population Health Research Unit, University of Oxford, Oxford OX3 7LF, UK.
  • Helgason H; Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.
  • Thompson DJ; Department of General Pediatrics, University of Münster, Münster, Germany.
  • Santon F; Deptartment of Growth and Reproduction, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark.
  • Petricek KM; deCODE Genetics/Amgen, Inc., Reykjavik, Iceland.
  • Zouaghi Y; School of Engineering and Natural Sciences, University of Iceland, Reykjavik, Iceland.
  • Huang-Doran I; Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge CB1 8RN, UK.
  • Gudbjartsson DF; Division of Endocrinology, Diabetology, and Metabolism, Lausanne University Hospital, 1011 Lausanne, Switzerland.
  • Bratland E; Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pharmacology, Berlin, Germany.
  • Lin K; Division of Endocrinology, Diabetology, and Metabolism, Lausanne University Hospital, 1011 Lausanne, Switzerland.
  • Gardner EJ; University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK.
  • Zhao Y; deCODE Genetics/Amgen, Inc., Reykjavik, Iceland.
  • Jia R; School of Engineering and Natural Sciences, University of Iceland, Reykjavik, Iceland.
  • Terao C; Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, NO-5020, Bergen, Norway.
  • Riggan M; Department of Medical Genetics, Haukeland University Hospital, NO-5021, Bergen, Norway.
  • Bolla MK; Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK.
  • Yazdanpanah M; MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Box 285 Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK.
  • Yazdanpanah N; MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Box 285 Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK.
  • Bradfield JP; MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Box 285 Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK.
  • Broer L; Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.
  • Campbell A; Clinical Research Center, Shizuoka General Hospital, Shizuoka, Japan.
  • Chasman DI; The Department of Applied Genetics, The School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan.
  • Cousminer DL; Department of Gynecology, Duke University Medical Center, Durham, North Carolina, USA.
  • Franceschini N; Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge CB1 8RN, UK.
  • Franke LH; Research Center of the Sainte-Justine University Hospital, University of Montreal, Montreal, Quebec, Canada.
  • Girotto G; Research Center of the Sainte-Justine University Hospital, University of Montreal, Montreal, Quebec, Canada.
  • He C; Quantinuum Research, Wayne, PA, USA.
  • Järvelin MR; Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
  • Joshi PK; Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands.
  • Kamatani Y; Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands.
  • Karlsson R; Centre for Genomic and Experimental Medicine, Institute of Genetics & Cancer, University of Edinburgh, Edinburgh, UK.
  • Luan J; Division of Preventive Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02215, USA.
  • Lunetta KL; Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
  • Mägi R; Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA.
  • Mangino M; Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
  • Medland SE; Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA.
  • Meisinger C; Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
  • Noordam R; Institute for Maternal and Child Health - IRCCS ''Burlo Garofolo", Trieste, Italy.
  • Nutile T; Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, Italy.
  • Concas MP; Department of Epidemiology and Biostatistics, Department of Big Data in Health Science, School of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, China.
  • Polasek O; Departments of Medical Oncology and Hematology, Sir Runrun Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China.
  • Porcu E; Department of Epidemiology and Biostatistics, MRC Health Protection Agency (HPA) Centre for Environment and Health, School of Public Health, Imperial College London, UK.
medRxiv ; 2023 Jun 20.
Article en En | MEDLINE | ID: mdl-37503126
ABSTRACT
Pubertal timing varies considerably and has been associated with a range of health outcomes in later life. To elucidate the underlying biological mechanisms, we performed multi-ancestry genetic analyses in ~800,000 women, identifying 1,080 independent signals associated with age at menarche. Collectively these loci explained 11% of the trait variance in an independent sample, with women at the top and bottom 1% of polygenic risk exhibiting a ~11 and ~14-fold higher risk of delayed and precocious pubertal development, respectively. These common variant analyses were supported by exome sequence analysis of ~220,000 women, identifying several genes, including rare loss of function variants in ZNF483 which abolished the impact of polygenic risk. Next, we implicated 660 genes in pubertal development using a combination of in silico variant-to-gene mapping approaches and integration with dynamic gene expression data from mouse embryonic GnRH neurons. This included an uncharacterized G-protein coupled receptor GPR83, which we demonstrate amplifies signaling of MC3R, a key sensor of nutritional status. Finally, we identified several genes, including ovary-expressed genes involved in DNA damage response that co-localize with signals associated with menopause timing, leading us to hypothesize that the ovarian reserve might signal centrally to trigger puberty. Collectively these findings extend our understanding of the biological complexity of puberty timing and highlight body size dependent and independent mechanisms that potentially link reproductive timing to later life disease.

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Tipo de estudio: Prognostic_studies Idioma: En Revista: MedRxiv Año: 2023 Tipo del documento: Article País de afiliación: Reino Unido

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Tipo de estudio: Prognostic_studies Idioma: En Revista: MedRxiv Año: 2023 Tipo del documento: Article País de afiliación: Reino Unido