151
|
Dixon-Suen SC, Lewis SJ, Martin RM, English DR, Boyle T, Giles GG, Michailidou K, Bolla MK, Wang Q, Dennis J, Lush M, Investigators A, Ahearn TU, Ambrosone CB, Andrulis IL, Anton-Culver H, Arndt V, Aronson KJ, Augustinsson A, Auvinen P, Beane Freeman LE, Becher H, Beckmann MW, Behrens S, Bermisheva M, Blomqvist C, Bogdanova NV, Bojesen SE, Bonanni B, Brenner H, Brüning T, Buys SS, Camp NJ, Campa D, Canzian F, Castelao JE, Cessna MH, Chang-Claude J, Chanock SJ, Clarke CL, Conroy DM, Couch FJ, Cox A, Cross SS, Czene K, Daly MB, Devilee P, Dörk T, Dwek M, Eccles DM, Eliassen AH, Engel C, Eriksson M, Evans DG, Fasching PA, Fletcher O, Flyger H, Fritschi L, Gabrielson M, Gago-Dominguez M, García-Closas M, García-Sáenz JA, Goldberg MS, Guénel P, Gündert M, Hahnen E, Haiman CA, Häberle L, Håkansson N, Hall P, Hamann U, Hart SN, Harvie M, Hillemanns P, Hollestelle A, Hooning MJ, Hoppe R, Hopper J, Howell A, Hunter DJ, Jakubowska A, Janni W, John EM, Jung A, Kaaks R, Keeman R, Kitahara CM, Koutros S, Kraft P, Kristensen VN, Kubelka-Sabit K, Kurian AW, Lacey JV, Lambrechts D, Le Marchand L, Lindblom A, Loibl S, Lubiński J, Mannermaa A, Manoochehri M, et alDixon-Suen SC, Lewis SJ, Martin RM, English DR, Boyle T, Giles GG, Michailidou K, Bolla MK, Wang Q, Dennis J, Lush M, Investigators A, Ahearn TU, Ambrosone CB, Andrulis IL, Anton-Culver H, Arndt V, Aronson KJ, Augustinsson A, Auvinen P, Beane Freeman LE, Becher H, Beckmann MW, Behrens S, Bermisheva M, Blomqvist C, Bogdanova NV, Bojesen SE, Bonanni B, Brenner H, Brüning T, Buys SS, Camp NJ, Campa D, Canzian F, Castelao JE, Cessna MH, Chang-Claude J, Chanock SJ, Clarke CL, Conroy DM, Couch FJ, Cox A, Cross SS, Czene K, Daly MB, Devilee P, Dörk T, Dwek M, Eccles DM, Eliassen AH, Engel C, Eriksson M, Evans DG, Fasching PA, Fletcher O, Flyger H, Fritschi L, Gabrielson M, Gago-Dominguez M, García-Closas M, García-Sáenz JA, Goldberg MS, Guénel P, Gündert M, Hahnen E, Haiman CA, Häberle L, Håkansson N, Hall P, Hamann U, Hart SN, Harvie M, Hillemanns P, Hollestelle A, Hooning MJ, Hoppe R, Hopper J, Howell A, Hunter DJ, Jakubowska A, Janni W, John EM, Jung A, Kaaks R, Keeman R, Kitahara CM, Koutros S, Kraft P, Kristensen VN, Kubelka-Sabit K, Kurian AW, Lacey JV, Lambrechts D, Le Marchand L, Lindblom A, Loibl S, Lubiński J, Mannermaa A, Manoochehri M, Margolin S, Martinez ME, Mavroudis D, Menon U, Mulligan AM, Murphy RA, Collaborators N, Nevanlinna H, Nevelsteen I, Newman WG, Offit K, Olshan AF, Olsson H, Orr N, Patel A, Peto J, Plaseska-Karanfilska D, Presneau N, Rack B, Radice P, Rees-Punia E, Rennert G, Rennert HS, Romero A, Saloustros E, Sandler DP, Schmidt MK, Schmutzler RK, Schwentner L, Scott C, Shah M, Shu XO, Simard J, Southey MC, Stone J, Surowy H, Swerdlow AJ, Tamimi RM, Tapper WJ, Taylor JA, Terry MB, Tollenaar RAEM, Troester MA, Truong T, Untch M, Vachon CM, Joseph V, Wappenschmidt B, Weinberg CR, Wolk A, Yannoukakos D, Zheng W, Ziogas A, Dunning AM, Pharoah PDP, Easton DF, Milne RL, Lynch BM. Physical activity, sedentary time and breast cancer risk: a Mendelian randomisation study. Br J Sports Med 2022; 56:1157-1170. [PMID: 36328784 PMCID: PMC9876601 DOI: 10.1136/bjsports-2021-105132] [Show More Authors] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/29/2022] [Indexed: 02/02/2023]
Abstract
OBJECTIVES Physical inactivity and sedentary behaviour are associated with higher breast cancer risk in observational studies, but ascribing causality is difficult. Mendelian randomisation (MR) assesses causality by simulating randomised trial groups using genotype. We assessed whether lifelong physical activity or sedentary time, assessed using genotype, may be causally associated with breast cancer risk overall, pre/post-menopause, and by case-groups defined by tumour characteristics. METHODS We performed two-sample inverse-variance-weighted MR using individual-level Breast Cancer Association Consortium case-control data from 130 957 European-ancestry women (69 838 invasive cases), and published UK Biobank data (n=91 105-377 234). Genetic instruments were single nucleotide polymorphisms (SNPs) associated in UK Biobank with wrist-worn accelerometer-measured overall physical activity (nsnps=5) or sedentary time (nsnps=6), or accelerometer-measured (nsnps=1) or self-reported (nsnps=5) vigorous physical activity. RESULTS Greater genetically-predicted overall activity was associated with lower breast cancer overall risk (OR=0.59; 95% confidence interval (CI) 0.42 to 0.83 per-standard deviation (SD;~8 milligravities acceleration)) and for most case-groups. Genetically-predicted vigorous activity was associated with lower risk of pre/perimenopausal breast cancer (OR=0.62; 95% CI 0.45 to 0.87,≥3 vs. 0 self-reported days/week), with consistent estimates for most case-groups. Greater genetically-predicted sedentary time was associated with higher hormone-receptor-negative tumour risk (OR=1.77; 95% CI 1.07 to 2.92 per-SD (~7% time spent sedentary)), with elevated estimates for most case-groups. Results were robust to sensitivity analyses examining pleiotropy (including weighted-median-MR, MR-Egger). CONCLUSION Our study provides strong evidence that greater overall physical activity, greater vigorous activity, and lower sedentary time are likely to reduce breast cancer risk. More widespread adoption of active lifestyles may reduce the burden from the most common cancer in women.
Collapse
Affiliation(s)
- Suzanne C Dixon-Suen
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Victoria, Australia
| | - Sarah J Lewis
- Bristol Medical School, Department of Population Health Sciences, University of Bristol, Bristol, UK
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Richard M Martin
- Bristol Medical School, Department of Population Health Sciences, University of Bristol, Bristol, UK
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- NIHR Biomedical Research Centre, University Hospitals Bristol and Weston NHS Foundation Trust and the University of Bristol, Bristol, UK
| | - Dallas R English
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Victoria, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Terry Boyle
- Allied Health and Human Performance, University of South Australia, Adelaide, South Australia, Australia
- Australian Centre for Precision Health, University of South Australia Cancer Research Institute, Adelaide, South Australia, Australia
| | - Graham G Giles
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Victoria, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, Victoria, Australia
| | - Kyriaki Michailidou
- Biostatistics Unit, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
- Cyprus School of Molecular Medicine, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Manjeet K Bolla
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Qin Wang
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Joe Dennis
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Michael Lush
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Abctb Investigators
- Australian Breast Cancer Tissue Bank, Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia
| | - Thomas U Ahearn
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | | | - Irene L Andrulis
- Fred A. Litwin Center for Cancer Genetics, Lunenfeld-Tanenbaum Research Institute of Mount Sinai Hospital, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Hoda Anton-Culver
- Department of Medicine, Genetic Epidemiology Research Institute, University of California Irvine, Irvine, California, USA
| | - Volker Arndt
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Kristan J Aronson
- Department of Public Health Sciences, and Cancer Research Institute, Queen's University, Kingston, Ontario, Canada
| | - Annelie Augustinsson
- Department of Cancer Epidemiology, Clinical Sciences, Lund University, Lund, Sweden
| | - Päivi Auvinen
- Department of Oncology, Cancer Center, Kuopio University Hospital, Kuopio, Finland
- Institute of Clinical Medicine, Oncology, University of Eastern Finland, Kuopio, Finland
| | - Laura E Beane Freeman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Heiko Becher
- Institute of Medical Biometry and Epidemiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Matthias W Beckmann
- Department of Gynecology and Obstetrics, Comprehensive Cancer Center ER-EMN, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Sabine Behrens
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Marina Bermisheva
- Institute of Biochemistry and Genetics, FSBSI Ufa Federal Research Centre of the Russian Academy of Sciences, Ufa, Russian Federation
| | - Carl Blomqvist
- Department of Oncology, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
- Department of Oncology, Örebro University Hospital, Örebro, Sweden
| | - Natalia V Bogdanova
- Department of Radiation Oncology, Hannover Medical School, Hannover, Germany
- Gynaecology Research Unit, Hannover Medical School, Hannover, Germany
| | - Stig E Bojesen
- Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
| | - Bernardo Bonanni
- Division of Cancer Prevention and Genetics, European Institute of Oncology IRCCS, Milan, Italy
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Thomas Brüning
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance (IPA), Institute of the Ruhr University Bochum, Bochum, Germany
| | - Saundra S Buys
- Department of Internal Medicine and Huntsman Cancer Institute, The University of Utah, Salt Lake City, Utah, USA
| | - Nicola J Camp
- Department of Internal Medicine and Huntsman Cancer Institute, The University of Utah, Salt Lake City, Utah, USA
| | - Daniele Campa
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Biology, University of Pisa, Pisa, Italy
| | - Federico Canzian
- Genomic Epidemiology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jose E Castelao
- Oncology and Genetics Unit, Instituto de Investigación Sanitaria Galicia Sur (IISGS), Xerencia de Xestion Integrada de Vigo-SERGAS, Vigo, Spain
| | - Melissa H Cessna
- Department of Pathology, Intermountain Medical Center, Intermountain Healthcare, Salt Lake City, Utah, USA
- Intermountain Biorepository, Intermountain Healthcare, Salt Lake City, Utah, USA
| | - Jenny Chang-Claude
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Cancer Epidemiology Group, University Cancer Center Hamburg (UCCH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stephen J Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Christine L Clarke
- Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia
| | - Don M Conroy
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Fergus J Couch
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Angela Cox
- Sheffield Institute for Nucleic Acids (SInFoNiA), Department of Oncology and Metabolism, The University of Sheffield, Sheffield, UK
| | - Simon S Cross
- Academic Unit of Pathology, Department of Neuroscience, The University of Sheffield, Sheffield, UK
| | - Kamila Czene
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Mary B Daly
- Department of Clinical Genetics, Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA
| | - Peter Devilee
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Thilo Dörk
- Gynaecology Research Unit, Hannover Medical School, Hannover, Germany
| | - Miriam Dwek
- School of Life Sciences, University of Westminster, London, UK
| | - Diana M Eccles
- Faculty of Medicine, University of Southampton, Southampton, UK
| | - A Heather Eliassen
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Christoph Engel
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
- LIFE - Leipzig Research Centre for Civilization Diseases, University of Leipzig, Leipzig, Germany
| | - Mikael Eriksson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - D Gareth Evans
- Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
- North West Genomics Laboratory Hub, Manchester Centre for Genomic Medicine, St Mary's Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Peter A Fasching
- Department of Gynecology and Obstetrics, Comprehensive Cancer Center ER-EMN, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
- David Geffen School of Medicine, Department of Medicine Division of Hematology and Oncology, University of California Los Angeles, Los Angeles, California, USA
| | - Olivia Fletcher
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | - Henrik Flyger
- Department of Breast Surgery, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
| | - Lin Fritschi
- School of Population Health, Curtin University, Perth, Western Australia, Australia
| | - Marike Gabrielson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Manuela Gago-Dominguez
- Genomic Medicine Group, International Cancer Genetics and Epidemiology Group, Fundación Pública Galega de Medicina Xenómica, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complejo Hospitalario Universitario de Santiago, SERGAS, Santiago de Compostela, Spain
- Moores Cancer Center, University of California San Diego, La Jolla, California, USA
| | - Montserrat García-Closas
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - José A García-Sáenz
- Medical Oncology Department, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain
| | - Mark S Goldberg
- Department of Medicine, McGill University, Montréal, Québec, Canada
- Division of Clinical Epidemiology, Royal Victoria Hospital, McGill University, Montréal, Québec, Canada
| | - Pascal Guénel
- Team 'Exposome and Heredity', Center for Research in Epidemiology and Population Health (CESP), Gustave Roussy, INSERM, University Paris-Saclay, Villejuif, France
| | - Melanie Gündert
- Molecular Epidemiology Group, C080, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Molecular Biology of Breast Cancer, University Womens Clinic Heidelberg, University of Heidelberg, Heidelberg, Germany
| | - Eric Hahnen
- Center for Familial Breast and Ovarian Cancer, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Integrated Oncology (CIO), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Christopher A Haiman
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Lothar Häberle
- Department of Gynecology and Obstetrics, Comprehensive Cancer Center ER-EMN, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Niclas Håkansson
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Per Hall
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Oncology, Södersjukhuset, Stockholm, Sweden
| | - Ute Hamann
- Molecular Genetics of Breast Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Steven N Hart
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, USA
| | - Michelle Harvie
- Prevent Breast Cancer Research Unit, Manchester University Hospital Foundation NHS Trust, Manchester, UK
| | - Peter Hillemanns
- Gynaecology Research Unit, Hannover Medical School, Hannover, Germany
| | | | - Maartje J Hooning
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Reiner Hoppe
- Dr Margarete Fischer Bosch Institute of Clinical Pharmacology, Stuttgart, Germany
- University of Tübingen, Tübingen, Germany
| | - John Hopper
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Anthony Howell
- Division of Cancer Sciences, The University of Manchester, Manchester, UK
| | - David J Hunter
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Anna Jakubowska
- Department of Genetics and Pathology, Pomeranian Medical University in Szczecin, Szczecin, Poland
- Independent Laboratory of Molecular Biology and Genetic Diagnostics, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Wolfgang Janni
- Department of Gynaecology and Obstetrics, University Hospital Ulm, Ulm, Germany
| | - Esther M John
- Department of Epidemiology and Population Health, Stanford University School of Medicine, Stanford, California, USA
- Department of Medicine, Division of Oncology, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California, USA
| | - Audrey Jung
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Rudolf Kaaks
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Renske Keeman
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Cari M Kitahara
- Radiation Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| | - Stella Koutros
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Peter Kraft
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Program in Genetic Epidemiology and Statistical Genetics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Vessela N Kristensen
- Department of Medical Genetics, Oslo University Hospital and University of Oslo, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Katerina Kubelka-Sabit
- Department of Histopathology and Cytology, Clinical Hospital Acibadem Sistina, Skopje, Macedonia (the former Yugoslav Republic of)
| | - Allison W Kurian
- Department of Epidemiology and Population Health, Stanford University School of Medicine, Stanford, California, USA
- Department of Medicine, Division of Oncology, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California, USA
| | - James V Lacey
- Department of Computational and Quantitative Medicine, City of Hope, Duarte, California, USA
- City of Hope Comprehensive Cancer Center, City of Hope, Duarte, California, USA
| | - Diether Lambrechts
- VIB Center for Cancer Biology, VIB, Gent, Oost-Vlaanderen, Belgium
- Laboratory for Translational Genetics, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Loic Le Marchand
- Epidemiology Program, University of Hawai'i Cancer Center, Honolulu, Hawaii, USA
| | - Annika Lindblom
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden
| | | | - Jan Lubiński
- Department of Genetics and Pathology, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Arto Mannermaa
- Translational Cancer Research Area, University of Eastern Finland, Kuopio, Finland
- Institute of Clinical Medicine, Pathology and Forensic Medicine, University of Eastern Finland, Kuopio, Finland
| | - Mehdi Manoochehri
- Molecular Genetics of Breast Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sara Margolin
- Department of Oncology, Södersjukhuset, Stockholm, Sweden
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Maria Elena Martinez
- Moores Cancer Center, University of California San Diego, La Jolla, California, USA
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California San Diego, La Jolla, California, USA
| | - Dimitrios Mavroudis
- Department of Medical Oncology, University Hospital of Heraklion, Heraklion, Greece
| | - Usha Menon
- MRC Clinical Trials Unit, Institute of Clinical Trials & Methodology, University College London, London, UK
| | - Anna Marie Mulligan
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Laboratory Medicine Program, University Health Network, Toronto, Ontario, Canada
| | - Rachel A Murphy
- Cancer Control Research, BC Cancer, Vancouver, British Columbia, Canada
- School of Population and Public Health, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Nbcs Collaborators
- Department of Medical Genetics, Oslo University Hospital and University of Oslo, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Heli Nevanlinna
- Department of Obstetrics and Gynecology, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Ines Nevelsteen
- Leuven Multidisciplinary Breast Center, Department of Oncology, Leuven Cancer Institute, University Hospitals Leuven, Leuven, Belgium
| | - William G Newman
- Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
- North West Genomics Laboratory Hub, Manchester Centre for Genomic Medicine, St Mary's Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Kenneth Offit
- Clinical Genetics Research Lab, Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York City, New York, USA
- Clinical Genetics Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York City, New York, USA
| | - Andrew F Olshan
- Department of Epidemiology, Gillings School of Global Public Health and UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Håkan Olsson
- Department of Cancer Epidemiology, Clinical Sciences, Lund University, Lund, Sweden
| | - Nick Orr
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK
| | - Alpa Patel
- Department of Population Science, American Cancer Society, Atlanta, Georgia, USA
| | - Julian Peto
- Department of Non-Communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK
| | - Dijana Plaseska-Karanfilska
- Research Centre for Genetic Engineering and Biotechnology 'Georgi D. Efremov', MASA, Skopje, Macedonia (the former Yugoslav Republic of)
| | - Nadege Presneau
- School of Life Sciences, University of Westminster, London, UK
| | - Brigitte Rack
- Department of Gynaecology and Obstetrics, University Hospital Ulm, Ulm, Germany
| | - Paolo Radice
- Unit of Molecular Bases of Genetic Risk and Genetic Testing, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Erika Rees-Punia
- Department of Population Science, American Cancer Society, Atlanta, Georgia, USA
| | - Gad Rennert
- Clalit National Cancer Control Center, Carmel Medical Center and Technion Faculty of Medicine, Haifa, Israel
| | - Hedy S Rennert
- Clalit National Cancer Control Center, Carmel Medical Center and Technion Faculty of Medicine, Haifa, Israel
| | - Atocha Romero
- Medical Oncology Department, Hospital Universitario Puerta de Hierro, Madrid, Spain
| | | | - Dale P Sandler
- Epidemiology Branch, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, North Carolina, USA
| | - Marjanka K Schmidt
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Division of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Rita K Schmutzler
- Center for Familial Breast and Ovarian Cancer, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Integrated Oncology (CIO), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Lukas Schwentner
- Department of Gynaecology and Obstetrics, University Hospital Ulm, Ulm, Germany
| | - Christopher Scott
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, USA
| | - Mitul Shah
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Xiao-Ou Shu
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Jacques Simard
- Genomics Center, Centre Hospitalier Universitaire de Québec - Université Laval Research Center, Québec City, Québec, Canada
| | - Melissa C Southey
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Victoria, Australia
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, Victoria, Australia
| | - Jennifer Stone
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
- Genetic Epidemiology Group, School of Population and Global Health, The University of Western Australia, Perth, Western Australia, Australia
| | - Harald Surowy
- Molecular Epidemiology Group, C080, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Molecular Biology of Breast Cancer, University Womens Clinic Heidelberg, University of Heidelberg, Heidelberg, Germany
| | - Anthony J Swerdlow
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
- Division of Breast Cancer Research, The Institute of Cancer Research, London, UK
| | - Rulla M Tamimi
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Department of Population Health Sciences, Weill Cornell Medicine, New York City, New York, USA
| | | | - Jack A Taylor
- Epidemiology Branch, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, North Carolina, USA
- Epigenetic and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, North Carolina, USA
| | - Mary Beth Terry
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York City, New York, USA
| | - Rob A E M Tollenaar
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Melissa A Troester
- Department of Epidemiology, Gillings School of Global Public Health and UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Thérèse Truong
- Team 'Exposome and Heredity', Center for Research in Epidemiology and Population Health (CESP), Gustave Roussy, INSERM, University Paris-Saclay, Villejuif, France
| | - Michael Untch
- Department of Gynecology and Obstetrics, Helios Clinics Berlin-Buch, Berlin, Germany
| | - Celine M Vachon
- Department of Quantitative Health Sciences, Division of Epidemiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Vijai Joseph
- Clinical Genetics Research Lab, Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York City, New York, USA
| | - Barbara Wappenschmidt
- Center for Familial Breast and Ovarian Cancer, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Integrated Oncology (CIO), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Clarice R Weinberg
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, North Carolina, USA
| | - Alicja Wolk
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Drakoulis Yannoukakos
- Molecular Diagnostics Laboratory, INRASTES, National Centre for Scientific Research-Demokritos, Athens, Greece
| | - Wei Zheng
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Argyrios Ziogas
- Department of Medicine, Genetic Epidemiology Research Institute, University of California Irvine, Irvine, California, USA
| | - Alison M Dunning
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Paul D P Pharoah
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Douglas F Easton
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Roger L Milne
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Victoria, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, Victoria, Australia
| | - Brigid M Lynch
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Victoria, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
- Physical Activity Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| |
Collapse
|
152
|
Zu T, Lian H, Green B, Yu Y. Ultra-high Dimensional Quantile Regression for Longitudinal Data: an Application to Blood Pressure Analysis. J Am Stat Assoc 2022. [DOI: 10.1080/01621459.2022.2128806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Affiliation(s)
- Tianhai Zu
- Department of Operations, Business Analytics, & Information Systems, University of Cincinnati, Cincinnati, Ohio, USA
| | - Heng Lian
- Department of Mathematics, City University of Hong Kong, Tat Chee Avenue, Kowloon Tong Hong Kong, China
| | - Brittany Green
- Department of Information Systems, Analytics, and Operations, University of Louisville, Louisville, Kentucky, USA
| | - Yan Yu
- Department of Operations, Business Analytics, & Information Systems, University of Cincinnati, Cincinnati, Ohio, USA
| |
Collapse
|
153
|
Endothelial and Vascular Smooth Muscle Dysfunction in Hypertension. Biochem Pharmacol 2022; 205:115263. [PMID: 36174768 DOI: 10.1016/j.bcp.2022.115263] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/19/2022] [Accepted: 09/20/2022] [Indexed: 12/11/2022]
Abstract
The development of essential hypertension involves several factors. Vascular dysfunction, characterized by endothelial dysfunction, low-grade inflammation and structural remodeling, plays an important role in the initiation and maintenance of essential hypertension. Although the mechanistic pathways by which essential hypertension develops are poorly understood, several pharmacological classes available on the clinical settings improve blood pressure by interfering in the cardiac output and/or vascular function. This review is divided in two major sections. The first section depicts the major molecular pathways as renin angiotensin aldosterone system (RAAS), endothelin, nitric oxide signalling pathway and oxidative stress in the development of vascular dysfunction. The second section describes the role of some pharmacological classes such as i) RAAS inhibitors, ii) dual angiotensin receptor-neprilysin inhibitors, iii) endothelin-1 receptor antagonists, iv) soluble guanylate cyclase modulators, v) phosphodiesterase type 5 inhibitors and vi) sodium-glucose cotransporter 2 inhibitors in the context of hypertension. Some classes are already approved in the treatment of hypertension, but others are not yet approved. However, due to their potential benefits these classes were included.
Collapse
|
154
|
Tomaszewski M, Morris AP, Howson JMM, Franceschini N, Eales JM, Xu X, Dikalov S, Guzik TJ, Humphreys BD, Harrap S, Charchar FJ. Kidney omics in hypertension: from statistical associations to biological mechanisms and clinical applications. Kidney Int 2022; 102:492-505. [PMID: 35690124 PMCID: PMC9886011 DOI: 10.1016/j.kint.2022.04.045] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 03/10/2022] [Accepted: 04/22/2022] [Indexed: 02/06/2023]
Abstract
Hypertension is a major cardiovascular disease risk factor and contributor to premature death globally. Family-based investigations confirmed a significant heritable component of blood pressure (BP), whereas genome-wide association studies revealed >1000 common and rare genetic variants associated with BP and/or hypertension. The kidney is not only an organ of key relevance to BP regulation and the development of hypertension, but it also acts as the tissue mediator of genetic predisposition to hypertension. The identity of kidney genes, pathways, and related mechanisms underlying the genetic associations with BP has started to emerge through integration of genomics with kidney transcriptomics, epigenomics, and other omics as well as through applications of causal inference, such as Mendelian randomization. Single-cell methods further enabled mapping of BP-associated kidney genes to cell types, and in conjunction with other omics, started to illuminate the biological mechanisms underpinning associations of BP-associated genetic variants and kidney genes. Polygenic risk scores derived from genome-wide association studies and refined on kidney omics hold the promise of enhanced diagnostic prediction, whereas kidney omics-informed drug discovery is likely to contribute new therapeutic opportunities for hypertension and hypertension-mediated kidney damage.
Collapse
Affiliation(s)
- Maciej Tomaszewski
- Division of Cardiovascular Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK; Manchester Heart Centre and Manchester Academic Health Science Centre, Manchester University NHS Foundation Trust, Manchester, UK.
| | - Andrew P Morris
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, Division of Musculoskeletal and Dermatological Sciences, University of Manchester, Manchester, UK
| | - Joanna M M Howson
- Department of Genetics, Novo Nordisk Research Centre Oxford, Novo Nordisk Ltd, Oxford, UK
| | - Nora Franceschini
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, North Carolina, USA
| | - James M Eales
- Division of Cardiovascular Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | - Xiaoguang Xu
- Division of Cardiovascular Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | - Sergey Dikalov
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Tomasz J Guzik
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK; Department of Internal and Agricultural Medicine, Jagiellonian University College of Medicine, Kraków, Poland
| | - Benjamin D Humphreys
- Division of Nephrology, Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Stephen Harrap
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia
| | - Fadi J Charchar
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia; Health Innovation and Transformation Centre, School of Science, Psychology and Sport, Federation University Australia, Ballarat, Victoria, Australia; Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| |
Collapse
|
155
|
Legaki E, Tsaklakidou D, Hatzimanolis A, Segredou E, Petalotis M, Moularogiorgou G, Mouchtouri V, Lykouras L, Stefanis NC, Gazouli M. Association of Alcohol Use Disorder Risk With ADH1B, DRD2, FAAH, SLC39A8, GCKR, and PDYN Genetic Polymorphisms. In Vivo 2022; 36:2092-2104. [PMID: 36099111 PMCID: PMC9463892 DOI: 10.21873/invivo.12935] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/01/2022] [Accepted: 07/04/2022] [Indexed: 11/10/2022]
Abstract
BACKGROUND/AIM Alcohol use disorder (AUD) is a chronic, multifactorial psychiatric condition with an enormous impact on public health and social cost. Genetic studies suggest a heritability, and genome-wide association studies (GWAS) have revealed genetic polymorphisms influencing AUD development. Our study aimed to investigate known variants located in ADH1B, DRD2, FAAH, SLC39A8, GCKR, and PDYN genes (rs1229984, rs7121986, rs324420, rs13107325, rs1260326, rs2281285 respectively) in an AUD Greek cohort in order to shed more light on the genetic predisposition to AUD. MATERIALS AND METHODS Alcohol-dependent individuals (n=251) meeting both the Diagnostic and Statistical Manual of Mental Disorders (DSM-IV) and the ICD-10 guidelines for alcohol abuse and dependence, and control individuals (n=280) were recruited. DNA was extracted from whole blood and PCR-restriction fragment length polymorphism (RFLP-PCR) or allele-specific PCR method was used for genotyping. RESULTS Individuals carrying the FAAH rs324420 A allele were significantly associated with increased risk of AUD (p<0.0001). SLC39A8 rs13107325 T allele and ADH1B rs1229984 T allele are overrepresented in control subjects (p<0.0001 and p<0.0001, respectively). The associations are maintained following an adjustment for age and sex and Bonferroni correction. GCKR rs13107325, DRD2 rs7121986, and PDYN rs2281285 polymorphisms did not show a significant association with AUD in the studied population after Bonferroni correction. CONCLUSION Susceptibility to AUD is related to variations in FAAH, ADH1B, and SLC39A8 genes. These polymorphisms could serve as potential biomarkers for AUD risk.
Collapse
Affiliation(s)
- Evangelia Legaki
- Department of Basic Medical Sciences, Laboratory of Biology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Domna Tsaklakidou
- Addiction Unit, 2nd University Department of Psychiatry, "Attikon" University General Hospital, Athens, Greece
| | - Alex Hatzimanolis
- First Department of Psychiatry, National and Kapodistrian University of Athens Medical School, Eginition Hospital, Athens, Greece
| | - Eirini Segredou
- Department of Alcohol Dependence, Psychiatric Hospital of Attica - Dafni, Haidari, Greece
| | - Markos Petalotis
- Department of Alcohol Dependence, Psychiatric Hospital of Attica - Dafni, Haidari, Greece
| | | | - Varvara Mouchtouri
- Department of Alcohol Dependence, Psychiatric Hospital of Attica - Dafni, Haidari, Greece
| | - Lefteris Lykouras
- Addiction Unit, 2nd University Department of Psychiatry, "Attikon" University General Hospital, Athens, Greece
| | - Nikos C Stefanis
- First Department of Psychiatry, National and Kapodistrian University of Athens Medical School, Eginition Hospital, Athens, Greece;
| | - Maria Gazouli
- Department of Basic Medical Sciences, Laboratory of Biology, Medical School, National and Kapodistrian University of Athens, Athens, Greece;
- 2 Department of Radiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- Department of Sciences, Hellenic Open University, Patras, Greece
| |
Collapse
|
156
|
Xiu X, Zhang H, Xue A, Cooper DN, Yan L, Yang Y, Yang Y, Zhao H. Genetic evidence for a causal relationship between type 2 diabetes and peripheral artery disease in both Europeans and East Asians. BMC Med 2022; 20:300. [PMID: 36042491 PMCID: PMC9429730 DOI: 10.1186/s12916-022-02476-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 07/12/2022] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Observational studies have revealed that type 2 diabetes (T2D) is associated with an increased risk of peripheral artery disease (PAD). However, whether the two diseases share a genetic basis and whether the relationship is causal remain unclear. It is also unclear as to whether these relationships differ between ethnic groups. METHODS By leveraging large-scale genome-wide association study (GWAS) summary statistics of T2D (European-based: Ncase = 21,926, Ncontrol = 342,747; East Asian-based: Ncase = 36,614, Ncontrol = 155,150) and PAD (European-based: Ncase = 5673, Ncontrol = 359,551; East Asian-based: Ncase = 3593, Ncontrol = 208,860), we explored the genetic correlation and putative causal relationship between T2D and PAD in both Europeans and East Asians using linkage disequilibrium score regression and seven Mendelian randomization (MR) models. We also performed multi-trait analysis of GWAS and two gene-based analyses to reveal candidate variants and risk genes involved in the shared genetic basis between T2D and PAD. RESULTS We observed a strong genetic correlation (rg) between T2D and PAD in both Europeans (rg = 0.51; p-value = 9.34 × 10-15) and East Asians (rg = 0.46; p-value = 1.67 × 10-12). The MR analyses provided consistent evidence for a causal effect of T2D on PAD in both ethnicities (odds ratio [OR] = 1.05 to 1.28 for Europeans and 1.15 to 1.27 for East Asians) but not PAD on T2D. This putative causal effect was not influenced by total cholesterol, body mass index, systolic blood pressure, or smoking initiation according to multivariable MR analysis, and the genetic overlap between T2D and PAD was further explored employing an independent European sample through polygenic risk score regression. Multi-trait analysis of GWAS revealed two novel European-specific single nucleotide polymorphisms (rs927742 and rs1734409) associated with the shared genetic basis of T2D and PAD. Gene-based analyses consistently identified one gene ANKFY1 and gene-gene interactions (e.g., STARD10 [European-specific] to AP3S2 [East Asian-specific]; KCNJ11 [European-specific] to KCNQ1 [East Asian-specific]) associated with the trans-ethnic genetic overlap between T2D and PAD, reflecting a common genetic basis for the co-occurrence of T2D and PAD in both Europeans and East Asians. CONCLUSIONS Our study provides the first evidence for a genetically causal effect of T2D on PAD in both Europeans and East Asians. Several candidate variants and risk genes were identified as being associated with this genetic overlap. Our findings emphasize the importance of monitoring PAD status in T2D patients and suggest new genetic biomarkers for screening PAD risk among patients with T2D.
Collapse
Affiliation(s)
- Xuehao Xiu
- Department of Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangzhou, China
| | - Haoyang Zhang
- Department of Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangzhou, China.,School of Data and Computer Science, Sun Yat-sen University, Guangzhou, 510000, China
| | - Angli Xue
- Garvan-Weizmann Centre for Cellular Genomics, Garvan Institute of Medical Research, Sydney, NSW, Australia.,Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - David N Cooper
- Institute of Medical Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff, CF14 4XN, UK
| | - Li Yan
- Department of Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangzhou, China
| | - Yuedong Yang
- School of Data and Computer Science, Sun Yat-sen University, Guangzhou, 510000, China.
| | - Yuanhao Yang
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia. .,Mater Research Institute, Translational Research Institute, Brisbane, QLD, Australia.
| | - Huiying Zhao
- Department of Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangzhou, China.
| |
Collapse
|
157
|
Life-Course Associations between Blood Pressure-Related Polygenic Risk Scores and Hypertension in the Bogalusa Heart Study. Genes (Basel) 2022; 13:genes13081473. [PMID: 36011384 PMCID: PMC9408577 DOI: 10.3390/genes13081473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 07/30/2022] [Accepted: 08/15/2022] [Indexed: 11/16/2022] Open
Abstract
Genetic information may help to identify individuals at increased risk for hypertension in early life, prior to the manifestation of elevated blood pressure (BP) values. We examined 369 Black and 832 White Bogalusa Heart Study (BHS) participants recruited in childhood and followed for approximately 37 years. The multi-ancestry genome-wide polygenic risk scores (PRSs) for systolic BP (SBP), diastolic BP (DBP), and hypertension were tested for an association with incident hypertension and stage 2 hypertension using Cox proportional hazards models. Race-stratified analyses were adjusted for baseline age, age2, sex, body mass index, genetic principal components, and BP. In Black participants, each standard deviation increase in SBP and DBP PRS conferred a 38% (p = 0.009) and 22% (p = 0.02) increased risk of hypertension and a 74% (p < 0.001) and 50% (p < 0.001) increased risk of stage 2 hypertension, respectively, while no association was observed with the hypertension PRSs. In Whites, each standard deviation increase in SBP, DBP, and hypertension PRS conferred a 24% (p < 0.05), 29% (p = 0.01), and 25% (p < 0.001) increased risk of hypertension, and a 27% (p = 0.08), 29% (0.01), and 42% (p < 0.001) increased risk of stage 2 hypertension, respectively. The addition of BP PRSs to the covariable-only models generally improved the C-statistics (p < 0.05). Multi-ancestry BP PRSs demonstrate the utility of genomic information in the early life prediction of hypertension.
Collapse
|
158
|
Nordengen K, Pålhaugen L, Bettella F, Bahrami S, Selnes P, Jarholm J, Athanasiu L, Shadrin A, Andreassen OA, Fladby T. Phenotype-informed polygenic risk scores are associated with worse outcome in individuals at risk of Alzheimer's disease. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2022; 14:e12350. [PMID: 35991219 PMCID: PMC9376972 DOI: 10.1002/dad2.12350] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 06/30/2022] [Accepted: 07/07/2022] [Indexed: 11/11/2022]
Abstract
Introduction Patients with predementia Alzheimer's disease (AD) and at-risk subjects are targets for promising disease-modifying treatments, and improved polygenic risk scores (PRSs) could improve early-stage case selection. Methods Phenotype-informed PRSs were developed by selecting AD-associated variants conditional on relevant inflammatory or cardiovascular traits. The primary outcome was longitudinal changes in measures of AD pathology, namely development of pathological amyloid deposition, medial temporal lobe atrophy, and cognitive decline in a prospective cohort study including 394 adults without AD dementia. Results High-risk groups defined by phenotype-informed AD PRSs had significantly steeper volume decline in medial temporal cortices, and the high-risk group defined by the cardiovascular-informed AD PRS had significantly increased hazard ratio of pathological amyloid deposition, compared to low-risk groups. Discussion AD PRSs informed by inflammatory disorders or cardiovascular risk factors and diseases are associated with development of AD pathology markers and may improve identification of subjects at risk for progression of AD.
Collapse
Affiliation(s)
- Kaja Nordengen
- Department of NeurologyAkershus University HospitalLørenskogNorway
- Institute of Clinical MedicineUniversity of OsloOsloNorway
| | - Lene Pålhaugen
- Department of NeurologyAkershus University HospitalLørenskogNorway
- Institute of Clinical MedicineUniversity of OsloOsloNorway
| | - Francesco Bettella
- Norwegian Centre for Mental Disorders Research (NORMENT)Division of Mental Health and AddictionOslo University HospitalOsloNorway
| | - Shahram Bahrami
- Norwegian Centre for Mental Disorders Research (NORMENT)Division of Mental Health and AddictionOslo University HospitalOsloNorway
| | - Per Selnes
- Department of NeurologyAkershus University HospitalLørenskogNorway
- Institute of Clinical MedicineUniversity of OsloOsloNorway
| | - Jonas Jarholm
- Department of NeurologyAkershus University HospitalLørenskogNorway
- Institute of Clinical MedicineUniversity of OsloOsloNorway
| | - Lavinia Athanasiu
- Norwegian Centre for Mental Disorders Research (NORMENT)Division of Mental Health and AddictionOslo University HospitalOsloNorway
| | - Alexey Shadrin
- Norwegian Centre for Mental Disorders Research (NORMENT)Division of Mental Health and AddictionOslo University HospitalOsloNorway
| | - Ole A. Andreassen
- Institute of Clinical MedicineUniversity of OsloOsloNorway
- Norwegian Centre for Mental Disorders Research (NORMENT)Division of Mental Health and AddictionOslo University HospitalOsloNorway
| | - Tormod Fladby
- Department of NeurologyAkershus University HospitalLørenskogNorway
- Institute of Clinical MedicineUniversity of OsloOsloNorway
| |
Collapse
|
159
|
Wang H, Luo Q, Ding X, Chen L, Zhang Z. Trimethylamine N-oxide and its precursors in relation to blood pressure: A mendelian randomization study. Front Cardiovasc Med 2022; 9:922441. [PMID: 35935641 PMCID: PMC9354484 DOI: 10.3389/fcvm.2022.922441] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 07/01/2022] [Indexed: 11/13/2022] Open
Abstract
Objective Previous studies have demonstrated that trimethylamine N-oxide (TMAO) and its precursors, including choline, betaine, and carnitine, are closely associated with blood pressure (BP) changes. Nevertheless, with the limitation of reverse causality and confounder in observational studies, such a relationship remains unclear. We aimed to assess the causal relationship of TMAO and its precursors with BP by the Mendelian Randomization (MR) approach. Method In this study, two-sample MR was used to reveal the causal effect of TMAO and its precursors on BP. Pooled data of TMAO and its precursors was from genome-wide association studies (GWAS) which includes summary data of human metabolome in 2,076 European participants from Framingham Heart Study. Summary-level data for BP was extracted from the International Consortium of Blood Pressure-Genome Wide Association Studies. Inverse variance weighted (IVW), MR Egger regression, Maximum likelihood, Weighted median, and MR pleiotropy residual sum and outlier test (MR-PRESSO) were used in this MR analysis. Results A total of 160 independent SNP loci were associated with TMAO and three precursors, including 58 associated with TMAO, 29 associated with choline, 44 associated with betaine, and 29 associated with carnitine, were selected. MR results suggested that a 1 unit increase in TMAO should be associated with a 1SD increase in systolic BP mmHg (beta: 0.039, SE, 0.072, p = 0.020). Additionally, our findings also indicated that a 1 unit increase in carnitine should be associated with a 1SD increase in systolic BP mmHg (beta: 0.055, SE: 0.075, p = 0.039). This result was also confirmed by sensitivity analysis methods such as Maximum likelihood, MR-PRESSO, and Weighted median. No effects of betaine or choline on systolic or diastolic BP were observed in the present study. Conclusion Our study provides evidence of a causal relationship of TMAO and its precursors with BP, suggesting that mediating the generation of TMAO would be beneficial for lowering BP.
Collapse
Affiliation(s)
- Han Wang
- Department of Cardiology, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
| | - Qiang Luo
- Department of Cardiology, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
| | - Xunshi Ding
- Department of Cardiology, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
| | - Lifang Chen
- Department of Cardiology, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
| | - Zheng Zhang
- Department of Cardiology, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
| |
Collapse
|
160
|
Francis CM, Futschik ME, Huang J, Bai W, Sargurupremraj M, Teumer A, Breteler MMB, Petretto E, Ho ASR, Amouyel P, Engelter ST, Bülow R, Völker U, Völzke H, Dörr M, Imtiaz MA, Aziz NA, Lohner V, Ware JS, Debette S, Elliott P, Dehghan A, Matthews PM. Genome-wide associations of aortic distensibility suggest causality for aortic aneurysms and brain white matter hyperintensities. Nat Commun 2022; 13:4505. [PMID: 35922433 PMCID: PMC9349177 DOI: 10.1038/s41467-022-32219-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 07/20/2022] [Indexed: 12/13/2022] Open
Abstract
Aortic dimensions and distensibility are key risk factors for aortic aneurysms and dissections, as well as for other cardiovascular and cerebrovascular diseases. We present genome-wide associations of ascending and descending aortic distensibility and area derived from cardiac magnetic resonance imaging (MRI) data of up to 32,590 Caucasian individuals in UK Biobank. We identify 102 loci (including 27 novel associations) tagging genes related to cardiovascular development, extracellular matrix production, smooth muscle cell contraction and heritable aortic diseases. Functional analyses highlight four signalling pathways associated with aortic distensibility (TGF-β, IGF, VEGF and PDGF). We identify distinct sex-specific associations with aortic traits. We develop co-expression networks associated with aortic traits and apply phenome-wide Mendelian randomization (MR-PheWAS), generating evidence for a causal role for aortic distensibility in development of aortic aneurysms. Multivariable MR suggests a causal relationship between aortic distensibility and cerebral white matter hyperintensities, mechanistically linking aortic traits and brain small vessel disease.
Collapse
Affiliation(s)
- Catherine M Francis
- National Heart and Lung Institute, Imperial College London, Programme in Cardiovascular Genetics and Genomics, London, UK
- Royal Brompton & Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, SW3 6NP, UK
| | - Matthias E Futschik
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- MRC London Institute of Medical Sciences (LMS), Imperial College London, London, W12 0NN, UK
| | - Jian Huang
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Wenjia Bai
- Department of Brain Sciences, Imperial College London, London, UK
- Department of Computing, Imperial College London, London, UK
| | - Muralidharan Sargurupremraj
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, TX, 78229, USA
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, team VINTAGE, UMR 1219, 33000, Bordeaux, France
| | - Alexander Teumer
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
- Department of Population Medicine and Lifestyle Diseases Prevention, Medical University of Bialystok, Bialystok, Poland
| | - Monique M B Breteler
- Population Health Sciences, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Institute for Medical Biometry, Informatics and Epidemiology (IMBIE), Faculty of Medicine, University of Bonn, Bonn, Germany
| | - Enrico Petretto
- Programme in Cardiovascular & Metabolic Disorders and Centre for Computational Biology, Duke-NUS Medical School, Singapore, 169857, Republic of Singapore
- Institute of Big Data and Artificial Intelligence, China Pharmaceutical University (CPU), 211198, Nanjing, China
- Computational Biology Programme, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Amanda S R Ho
- Computational Biology Programme, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Philippe Amouyel
- LabEx DISTALZ-U1167, RID-AGE-Risk Factors and Molecular Determinants of Aging-Related Diseases, University of Lille, Lille, France
- Inserm, U1167, Lille, France
- Centre Hospitalier Universitaire Lille, Lille, France
- Institut Pasteur de Lille, Lille, France
| | - Stefan T Engelter
- Department of Neurology and Stroke Center, University Hospital and University of Basel, Petersgraben 4, CH - 4031, Basel, Switzerland
- Department of Clinical Neurology and Neurorehabilitation, University Department of Geriatric Medicine FELIX PLATTER, University of Basel, Basel, Switzerland
| | - Robin Bülow
- Department of Radiology and Neuroradiology, University Medicine Greifswald, Greifswald, Germany
| | - Uwe Völker
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Henry Völzke
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Marcus Dörr
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany
| | - Mohammed-Aslam Imtiaz
- Population Health Sciences, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - N Ahmad Aziz
- Population Health Sciences, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Neurology, Faculty of Medicine, University of Bonn, Bonn, Germany
| | - Valerie Lohner
- Population Health Sciences, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - James S Ware
- National Heart and Lung Institute, Imperial College London, Programme in Cardiovascular Genetics and Genomics, London, UK
- Royal Brompton & Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, SW3 6NP, UK
- MRC London Institute of Medical Sciences (LMS), Imperial College London, London, W12 0NN, UK
| | - Stephanie Debette
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, team VINTAGE, UMR 1219, 33000, Bordeaux, France
- Department of Neurology, Institute for Neurodegenerative Diseases, Bordeaux University Hospital - CHU Bordeaux, 33000, Bordeaux, France
| | - Paul Elliott
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- UK Dementia Research Institute at Imperial College London, London, UK
- Health Data Research (HDR) UK London at Imperial College London, London, UK
- Britsh Heart Foundation Centre of Research Excellence at Imperial College London, London, UK
- National Institute for Health Research Imperial Biomedical Research Centre, Imperial College London, London, UK
- MRC Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
| | - Abbas Dehghan
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK.
- UK Dementia Research Institute at Imperial College London, London, UK.
| | - Paul M Matthews
- Department of Brain Sciences, Imperial College London, London, UK.
- UK Dementia Research Institute at Imperial College London, London, UK.
- National Institute for Health Research Imperial Biomedical Research Centre, Imperial College London, London, UK.
| |
Collapse
|
161
|
Kelly TN, Sun X, He KY, Brown MR, Taliun SAG, Hellwege JN, Irvin MR, Mi X, Brody JA, Franceschini N, Guo X, Hwang SJ, de Vries PS, Gao Y, Moscati A, Nadkarni GN, Yanek LR, Elfassy T, Smith JA, Chung RH, Beitelshees AL, Patki A, Aslibekyan S, Blobner BM, Peralta JM, Assimes TL, Palmas WR, Liu C, Bress AP, Huang Z, Becker LC, Hwa CM, O'Connell JR, Carlson JC, Warren HR, Das S, Giri A, Martin LW, Craig Johnson W, Fox ER, Bottinger EP, Razavi AC, Vaidya D, Chuang LM, Chang YPC, Naseri T, Jain D, Kang HM, Hung AM, Srinivasasainagendra V, Snively BM, Gu D, Montasser ME, Reupena MS, Heavner BD, LeFaive J, Hixson JE, Rice KM, Wang FF, Nielsen JB, Huang J, Khan AT, Zhou W, Nierenberg JL, Laurie CC, Armstrong ND, Shi M, Pan Y, Stilp AM, Emery L, Wong Q, Hawley NL, Minster RL, Curran JE, Munroe PB, Weeks DE, North KE, Tracy RP, Kenny EE, Shimbo D, Chakravarti A, Rich SS, Reiner AP, Blangero J, Redline S, Mitchell BD, Rao DC, Ida Chen YD, Kardia SLR, Kaplan RC, Mathias RA, He J, Psaty BM, Fornage M, Loos RJF, Correa A, Boerwinkle E, Rotter JI, Kooperberg C, Edwards TL, et alKelly TN, Sun X, He KY, Brown MR, Taliun SAG, Hellwege JN, Irvin MR, Mi X, Brody JA, Franceschini N, Guo X, Hwang SJ, de Vries PS, Gao Y, Moscati A, Nadkarni GN, Yanek LR, Elfassy T, Smith JA, Chung RH, Beitelshees AL, Patki A, Aslibekyan S, Blobner BM, Peralta JM, Assimes TL, Palmas WR, Liu C, Bress AP, Huang Z, Becker LC, Hwa CM, O'Connell JR, Carlson JC, Warren HR, Das S, Giri A, Martin LW, Craig Johnson W, Fox ER, Bottinger EP, Razavi AC, Vaidya D, Chuang LM, Chang YPC, Naseri T, Jain D, Kang HM, Hung AM, Srinivasasainagendra V, Snively BM, Gu D, Montasser ME, Reupena MS, Heavner BD, LeFaive J, Hixson JE, Rice KM, Wang FF, Nielsen JB, Huang J, Khan AT, Zhou W, Nierenberg JL, Laurie CC, Armstrong ND, Shi M, Pan Y, Stilp AM, Emery L, Wong Q, Hawley NL, Minster RL, Curran JE, Munroe PB, Weeks DE, North KE, Tracy RP, Kenny EE, Shimbo D, Chakravarti A, Rich SS, Reiner AP, Blangero J, Redline S, Mitchell BD, Rao DC, Ida Chen YD, Kardia SLR, Kaplan RC, Mathias RA, He J, Psaty BM, Fornage M, Loos RJF, Correa A, Boerwinkle E, Rotter JI, Kooperberg C, Edwards TL, Abecasis GR, Zhu X, Levy D, Arnett DK, Morrison AC. Insights From a Large-Scale Whole-Genome Sequencing Study of Systolic Blood Pressure, Diastolic Blood Pressure, and Hypertension. Hypertension 2022; 79:1656-1667. [PMID: 35652341 PMCID: PMC9593435 DOI: 10.1161/hypertensionaha.122.19324] [Show More Authors] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 05/12/2022] [Indexed: 12/30/2022]
Abstract
BACKGROUND The availability of whole-genome sequencing data in large studies has enabled the assessment of coding and noncoding variants across the allele frequency spectrum for their associations with blood pressure. METHODS We conducted a multiancestry whole-genome sequencing analysis of blood pressure among 51 456 Trans-Omics for Precision Medicine and Centers for Common Disease Genomics program participants (stage-1). Stage-2 analyses leveraged array data from UK Biobank (N=383 145), Million Veteran Program (N=318 891), and Reasons for Geographic and Racial Differences in Stroke (N=10 643) participants, along with whole-exome sequencing data from UK Biobank (N=199 631) participants. RESULTS Two blood pressure signals achieved genome-wide significance in meta-analyses of stage-1 and stage-2 single variant findings (P<5×10-8). Among them, a rare intergenic variant at novel locus, LOC100506274, was associated with lower systolic blood pressure in stage-1 (beta [SE]=-32.6 [6.0]; P=4.99×10-8) but not stage-2 analysis (P=0.11). Furthermore, a novel common variant at the known INSR locus was suggestively associated with diastolic blood pressure in stage-1 (beta [SE]=-0.36 [0.07]; P=4.18×10-7) and attained genome-wide significance in stage-2 (beta [SE]=-0.29 [0.03]; P=7.28×10-23). Nineteen additional signals suggestively associated with blood pressure in meta-analysis of single and aggregate rare variant findings (P<1×10-6 and P<1×10-4, respectively). DISCUSSION We report one promising but unconfirmed rare variant for blood pressure and, more importantly, contribute insights for future blood pressure sequencing studies. Our findings suggest promise of aggregate analyses to complement single variant analysis strategies and the need for larger, diverse samples, and family studies to enable robust rare variant identification.
Collapse
Affiliation(s)
- Tanika N Kelly
- Department of Epidemiology (T.N.K., X.S., X.M., Z.H., A.C.R., J.L.N., M.S., Y.P., J.H.), Tulane University, New Orleans, LA
- Translational Sciences Institute (T.N.K., J.H.), Tulane University, New Orleans, LA
| | - Xiao Sun
- Department of Epidemiology (T.N.K., X.S., X.M., Z.H., A.C.R., J.L.N., M.S., Y.P., J.H.), Tulane University, New Orleans, LA
| | - Karen Y He
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH (K.Y.H., X.Z.)
| | - Michael R Brown
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health (M.R.B., P.D.d.V., J.E.H., E.B., A.C.M.), The University of Texas Health Science Center at Houston' Houston' TX
| | - Sarah A Gagliano Taliun
- Department of Biostatistics (S.A.G.T., S.D., H.M.K., J.L., G.R.A.), University of Michigan, Ann Arbor' MI
| | - Jacklyn N Hellwege
- Division of Genetic Medicine, Department of Medicine (J.N.H.), Vanderbilt University Medical Center, Nashville, TN
- Biomedical Laboratory Research and Development, Tennessee Valley Healthcare System (626)/Vanderbilt University, Nashville' TN (J.N.H., A.G., A.M.H., T.L.E.)
| | - Marguerite R Irvin
- Department of Epidemiology (M.R.I., S.A., N.D.A.), University of Alabama at Birmingham' AL
| | - Xuenan Mi
- Department of Epidemiology (T.N.K., X.S., X.M., Z.H., A.C.R., J.L.N., M.S., Y.P., J.H.), Tulane University, New Orleans, LA
| | - Jennifer A Brody
- Cardiovascular Health Research Unit, Department of Medicine (J.A.B., K.E.N.), University of Washington, Seattle' WA
| | - Nora Franceschini
- Department of Epidemiology, University of North Carolina, Chapel Hill (N.F.)
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance' CA (X.G., Y.-D.I.C., J.I.R., D.L.)
| | - Shih-Jen Hwang
- National Heart, Lung and Blood Institute, Population Sciences Branch, National Institutes of Health, Framingham, MA (S.-J.H.)
| | - Paul S de Vries
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health (M.R.B., P.D.d.V., J.E.H., E.B., A.C.M.), The University of Texas Health Science Center at Houston' Houston' TX
| | - Yan Gao
- Department of Physiology and Biophysics (Y.G., E.E.K., R.J.F.L.), University of Mississippi Medical Center, Jackson' MS
| | - Arden Moscati
- The Charles Bronfman Institute for Personalized Medicine (A.M., G.N.N.), The Icahn School of Medicine at Mount Sinai, New York, NY
| | - Girish N Nadkarni
- The Charles Bronfman Institute for Personalized Medicine (A.M., G.N.N.), The Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Medicine (G.N.N.), The Icahn School of Medicine at Mount Sinai, New York, NY
| | - Lisa R Yanek
- Division of General Internal Medicine, Department of Medicine (L.R.Y., D.V.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Tali Elfassy
- Division of Epidemiology, Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami' FL (T.E.)
| | - Jennifer A Smith
- Department of Epidemiology (J.A.S., S.L.R.K.), University of Michigan, Ann Arbor' MI
| | - Ren-Hua Chung
- Institute of Population Sciences, National Health Research Institutes, Taiwan (R.-H.C.)
| | - Amber L Beitelshees
- Division of Endocrinology, Diabetes, and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore' MD (A.L.B., J.R.O., Y.-P.C.C., M.E.M., B.D.M.)
| | - Amit Patki
- Department of Biostatistics (A.P., V.S.), University of Alabama at Birmingham' AL
| | - Stella Aslibekyan
- Department of Epidemiology (M.R.I., S.A., N.D.A.), University of Alabama at Birmingham' AL
| | - Brandon M Blobner
- Cardiovascular Health Research Unit, Departments of Medicine, Epidemiology, and Health Services (B.M.P.), University of Washington, Seattle' WA
- Department of Human Genetics (B.M.B., R.L.M., D.E.W.), University of Pittsburgh, PA
| | - Juan M Peralta
- Department of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley, Brownsville' TX (J.M.P., J.E.C., J.B.)
| | - Themistocles L Assimes
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University, Stanford' CA (T.L.A.)
- Division of Cardiology Medicine, Palo Alto VA HealthCare System, Palo Alto' CA (T.L.A.)
| | - Walter R Palmas
- Division of General Medicine, Department of Medicine, Columbia University, New York, NY (W.R.P.)
| | - Chunyu Liu
- Department of Biostatistics, Boston University, Boston' MA (C.L.)
| | - Adam P Bress
- Division of Health System Innovation and Research, Department of Population Health Sciences, University of Utah School of Medicine, Salt Lake City' UT (A.P.B.)
| | - Zhijie Huang
- Department of Epidemiology (T.N.K., X.S., X.M., Z.H., A.C.R., J.L.N., M.S., Y.P., J.H.), Tulane University, New Orleans, LA
| | - Lewis C Becker
- Division of Cardiology, Department of Medicine (L.C.B.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Chii-Min Hwa
- Taichung Veterans General Hospital, Taichung, Taiwan (C.-M.H.)
| | - Jeffrey R O'Connell
- Division of Endocrinology, Diabetes, and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore' MD (A.L.B., J.R.O., Y.-P.C.C., M.E.M., B.D.M.)
| | - Jenna C Carlson
- Department of Biostatistics, Graduate School of Public Health (J.C.C.), University of Pittsburgh, PA
| | - Helen R Warren
- Department of Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry (H.R.W., P.B.M.), Queen Mary University of London, United Kingdom
- National Institute for Health Research Barts Cardiovascular Biomedical Research Centre (H.R.W., P.B.M.), Queen Mary University of London, United Kingdom
| | - Sayantan Das
- Department of Biostatistics (S.A.G.T., S.D., H.M.K., J.L., G.R.A.), University of Michigan, Ann Arbor' MI
| | - Ayush Giri
- Biomedical Laboratory Research and Development, Tennessee Valley Healthcare System (626)/Vanderbilt University, Nashville' TN (J.N.H., A.G., A.M.H., T.L.E.)
- Division of Quantitative Sciences, Department of Obstetrics and Gynecology, Vanderbilt University, Nashville, TN (A.G.)
| | - Lisa W Martin
- Division of Cardiology, Department of Medicine, George Washington University, Washington, DC (L.W.M.)
| | - W Craig Johnson
- Department of Biostatistics, School of Public Health (W.C.J., D.J., B.D.H., K.M.R., F.F.E., A.T.K., C.C.L., A.M.S., L.E., Q.W.), University of Washington, Seattle' WA
| | - Ervin R Fox
- Division of Cardiology, Department of Medicine (E.R.F.), University of Mississippi Medical Center, Jackson' MS
| | - Erwin P Bottinger
- Hasso Plattner Institute for Digital Health at Mount Sinai (E.P.B.), The Icahn School of Medicine at Mount Sinai, New York, NY
| | - Alexander C Razavi
- Department of Epidemiology (T.N.K., X.S., X.M., Z.H., A.C.R., J.L.N., M.S., Y.P., J.H.), Tulane University, New Orleans, LA
| | - Dhananjay Vaidya
- Division of General Internal Medicine, Department of Medicine (L.R.Y., D.V.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Lee-Ming Chuang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei' Taiwan (L.-M.C.)
| | - Yen-Pei C Chang
- Division of Endocrinology, Diabetes, and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore' MD (A.L.B., J.R.O., Y.-P.C.C., M.E.M., B.D.M.)
| | - Take Naseri
- Ministry of Health, Government of Samoa, Apia' Samoa (T.N.)
| | - Deepti Jain
- Department of Biostatistics, School of Public Health (W.C.J., D.J., B.D.H., K.M.R., F.F.E., A.T.K., C.C.L., A.M.S., L.E., Q.W.), University of Washington, Seattle' WA
| | - Hyun Min Kang
- Department of Biostatistics (S.A.G.T., S.D., H.M.K., J.L., G.R.A.), University of Michigan, Ann Arbor' MI
| | - Adriana M Hung
- Division of Nephrology and Hypertension, Department of Medicine (A.M.H.), Vanderbilt University Medical Center, Nashville, TN
- Biomedical Laboratory Research and Development, Tennessee Valley Healthcare System (626)/Vanderbilt University, Nashville' TN (J.N.H., A.G., A.M.H., T.L.E.)
| | | | - Beverly M Snively
- Division of Public Health Sciences, Department of Biostatistics and Data Science, Wake Forest School of Medicine, Winston-Salem, NC (B.M.S.)
| | - Dongfeng Gu
- Department of Epidemiology and Key Laboratory of Cardiovascular Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (D.G., J.H.)
| | - May E Montasser
- Division of Endocrinology, Diabetes, and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore' MD (A.L.B., J.R.O., Y.-P.C.C., M.E.M., B.D.M.)
| | | | - Benjamin D Heavner
- Department of Biostatistics, School of Public Health (W.C.J., D.J., B.D.H., K.M.R., F.F.E., A.T.K., C.C.L., A.M.S., L.E., Q.W.), University of Washington, Seattle' WA
| | - Jonathon LeFaive
- Department of Biostatistics (S.A.G.T., S.D., H.M.K., J.L., G.R.A.), University of Michigan, Ann Arbor' MI
| | - James E Hixson
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health (M.R.B., P.D.d.V., J.E.H., E.B., A.C.M.), The University of Texas Health Science Center at Houston' Houston' TX
| | - Kenneth M Rice
- Department of Biostatistics, School of Public Health (W.C.J., D.J., B.D.H., K.M.R., F.F.E., A.T.K., C.C.L., A.M.S., L.E., Q.W.), University of Washington, Seattle' WA
| | - Fei Fei Wang
- Department of Biostatistics, School of Public Health (W.C.J., D.J., B.D.H., K.M.R., F.F.E., A.T.K., C.C.L., A.M.S., L.E., Q.W.), University of Washington, Seattle' WA
| | - Jonas B Nielsen
- Department of Internal Medicine: Cardiology (J.B.N.), University of Michigan, Ann Arbor' MI
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark (J.B.N.)
| | - Jianfeng Huang
- Translational Sciences Institute (T.N.K., J.H.), Tulane University, New Orleans, LA
- Department of Epidemiology and Key Laboratory of Cardiovascular Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (D.G., J.H.)
| | - Alyna T Khan
- Department of Biostatistics, School of Public Health (W.C.J., D.J., B.D.H., K.M.R., F.F.E., A.T.K., C.C.L., A.M.S., L.E., Q.W.), University of Washington, Seattle' WA
| | - Wei Zhou
- Department of Computational Medicine and Bioinformatics (W.Z.), University of Michigan, Ann Arbor' MI
| | - Jovia L Nierenberg
- Department of Epidemiology (T.N.K., X.S., X.M., Z.H., A.C.R., J.L.N., M.S., Y.P., J.H.), Tulane University, New Orleans, LA
| | - Cathy C Laurie
- Department of Biostatistics, School of Public Health (W.C.J., D.J., B.D.H., K.M.R., F.F.E., A.T.K., C.C.L., A.M.S., L.E., Q.W.), University of Washington, Seattle' WA
| | - Nicole D Armstrong
- Department of Epidemiology (M.R.I., S.A., N.D.A.), University of Alabama at Birmingham' AL
| | - Mengyao Shi
- Department of Epidemiology (T.N.K., X.S., X.M., Z.H., A.C.R., J.L.N., M.S., Y.P., J.H.), Tulane University, New Orleans, LA
| | - Yang Pan
- Department of Epidemiology (T.N.K., X.S., X.M., Z.H., A.C.R., J.L.N., M.S., Y.P., J.H.), Tulane University, New Orleans, LA
| | - Adrienne M Stilp
- Department of Biostatistics, School of Public Health (W.C.J., D.J., B.D.H., K.M.R., F.F.E., A.T.K., C.C.L., A.M.S., L.E., Q.W.), University of Washington, Seattle' WA
| | - Leslie Emery
- Department of Biostatistics, School of Public Health (W.C.J., D.J., B.D.H., K.M.R., F.F.E., A.T.K., C.C.L., A.M.S., L.E., Q.W.), University of Washington, Seattle' WA
| | - Quenna Wong
- Department of Biostatistics, School of Public Health (W.C.J., D.J., B.D.H., K.M.R., F.F.E., A.T.K., C.C.L., A.M.S., L.E., Q.W.), University of Washington, Seattle' WA
| | - Nicola L Hawley
- Department of Chronic Disease Epidemiology, Yale University, New Haven, CT (N.L.H.)
| | - Ryan L Minster
- Department of Human Genetics (B.M.B., R.L.M., D.E.W.), University of Pittsburgh, PA
| | - Joanne E Curran
- Department of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley, Brownsville' TX (J.M.P., J.E.C., J.B.)
| | - Patricia B Munroe
- Department of Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry (H.R.W., P.B.M.), Queen Mary University of London, United Kingdom
- National Institute for Health Research Barts Cardiovascular Biomedical Research Centre (H.R.W., P.B.M.), Queen Mary University of London, United Kingdom
| | - Daniel E Weeks
- Department of Human Genetics (B.M.B., R.L.M., D.E.W.), University of Pittsburgh, PA
- Department of Biostatistics (D.E.W.), University of Pittsburgh, PA
| | - Kari E North
- Cardiovascular Health Research Unit, Department of Medicine (J.A.B., K.E.N.), University of Washington, Seattle' WA
| | - Russell P Tracy
- Department of Pathology & Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington' VT (R.P.T.)
| | - Eimear E Kenny
- Department of Physiology and Biophysics (Y.G., E.E.K., R.J.F.L.), University of Mississippi Medical Center, Jackson' MS
- Department of Genetics and Genomics (E.E.K.), The Icahn School of Medicine at Mount Sinai, New York, NY
| | - Daichi Shimbo
- Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY (D.S.)
| | - Aravinda Chakravarti
- Department of Medicine (A.C.), University of Mississippi Medical Center, Jackson' MS
| | - Stephen S Rich
- Center for Public Health, University of Virginia, Charlottesville' VA (S.S.R.)
| | - Alex P Reiner
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA (A.P.R., C.K.)
| | - John Blangero
- Department of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley, Brownsville' TX (J.M.P., J.E.C., J.B.)
| | - Susan Redline
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital, Boston, MA (S.R.)
| | - Braxton D Mitchell
- Division of Endocrinology, Diabetes, and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore' MD (A.L.B., J.R.O., Y.-P.C.C., M.E.M., B.D.M.)
- Geriatrics Research and Education Clinical Center, Baltimore Veterans Administration Medical Center, Baltimore' MD (B.D.M.)
| | - Dabeeru C Rao
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO (D.C.R.)
| | - Yii-Der Ida Chen
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance' CA (X.G., Y.-D.I.C., J.I.R., D.L.)
| | - Sharon L R Kardia
- Department of Epidemiology (J.A.S., S.L.R.K.), University of Michigan, Ann Arbor' MI
| | - Robert C Kaplan
- Division of Social Medicine, Albert Einstein College of Medicine, Bronx, NY (R.C.K.)
| | - Rasika A Mathias
- Division of Allergy & Clinical Immunology, Department of Medicine (R.A.M.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jiang He
- Department of Epidemiology (T.N.K., X.S., X.M., Z.H., A.C.R., J.L.N., M.S., Y.P., J.H.), Tulane University, New Orleans, LA
| | - Bruce M Psaty
- Department of Epidemiology (T.N.K., X.S., X.M., Z.H., A.C.R., J.L.N., M.S., Y.P., J.H.), Tulane University, New Orleans, LA
- Kaiser Permanente Washington Health Research Institute, Seattle' WA (B.M.P.)
| | - Myriam Fornage
- Brown Foundation Institute of Molecular Medicine (M.F.), The University of Texas Health Science Center at Houston' Houston' TX
- Human Genetics Center (M.F.), The University of Texas Health Science Center at Houston' Houston' TX
| | - Ruth J F Loos
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance' CA (X.G., Y.-D.I.C., J.I.R., D.L.)
- The Mindich Child Health and Development Institute (R.J.F.L.), The Icahn School of Medicine at Mount Sinai, New York, NY
| | - Adolfo Correa
- Center for Human Genetics and Genomics, New York University Grossman School of Medicine, New York, NY (A.C.)
| | - Eric Boerwinkle
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health (M.R.B., P.D.d.V., J.E.H., E.B., A.C.M.), The University of Texas Health Science Center at Houston' Houston' TX
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX (E.B.)
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance' CA (X.G., Y.-D.I.C., J.I.R., D.L.)
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA (A.P.R., C.K.)
| | - Todd L Edwards
- Division of Epidemiology, Department of Medicine (T.L.E.), Vanderbilt University Medical Center, Nashville, TN
- Biomedical Laboratory Research and Development, Tennessee Valley Healthcare System (626)/Vanderbilt University, Nashville' TN (J.N.H., A.G., A.M.H., T.L.E.)
| | - Gonçalo R Abecasis
- Department of Biostatistics (S.A.G.T., S.D., H.M.K., J.L., G.R.A.), University of Michigan, Ann Arbor' MI
| | - Xiaofeng Zhu
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH (K.Y.H., X.Z.)
| | - Daniel Levy
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance' CA (X.G., Y.-D.I.C., J.I.R., D.L.)
| | - Donna K Arnett
- College of Public Health, University of Kentucky, Lexington, KY (D.K.A.)
| | - Alanna C Morrison
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health (M.R.B., P.D.d.V., J.E.H., E.B., A.C.M.), The University of Texas Health Science Center at Houston' Houston' TX
| |
Collapse
|
162
|
Sudano I. Hypertension and Hypercholesterolemia: Let's Make Therapy Easy and Efficient. PRAXIS 2022; 111:558-563. [PMID: 35920009 DOI: 10.1024/1661-8157/a003894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Arterial hypertension and hypercholesterolemia increase the risk of cardiovascular morbidity and mortality. Effectively reducing both blood pressure and circulating low-density lipoprotein cholesterol may considerably reduce cardiovascular risk. As this is particularly true if the intervention starts early, it is very important to identify and treat hypertension and hypercholesterolemia as early as possible. By improving adherence, a single-pill formulation that offers a combination of different drugs could be an effective way to manage patients with multiple cardiovascular risks.
Collapse
Affiliation(s)
- Isabella Sudano
- University Heart Centre, Clinic for Cardiology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
163
|
Sapena V, Iavarone M, Boix L, Facchetti F, Guarino M, Sanduzzi Zamparelli M, Granito A, Samper E, Scartozzi M, Corominas J, Marisi G, Díaz A, Casadei-Gardini A, Gramantieri L, Lampertico P, Morisco F, Torres F, Bruix J, Reig M. Polymorphism AGT2 (rs4762) is involved in the development of dermatologic events: Proof-of-concept in hepatocellular carcinoma patients treated with sorafenib. World J Hepatol 2022; 14:1438-1458. [PMID: 36158918 PMCID: PMC9376774 DOI: 10.4254/wjh.v14.i7.1438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 01/24/2022] [Accepted: 07/06/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Dermatologic adverse events (DAEs) are associated with a better outcome in patients with hepatocellular carcinoma (HCC) irrespective of the therapeutic agent received. The exact mechanisms associated with the development of DAEs are unknown although several studies point to direct toxicity of tyrosine kinase inhibitors (TKIs) to the skin or an immune-mediated reaction triggered by the oncologic treatment. As is the case in other conditions, individual genetic variants may partially explain a higher risk of DAEs. AIM To evaluate the contribution of several gene variants to the risk of developing DAEs in HCC patients treated with TKIs. METHODS We first analyzed 27 single-nucleotide polymorphisms (SNPs) from 12 genes selected as potential predictors of adverse event (AE) development in HCC patients treated with sorafenib [Barcelona Clinic Liver Cancer 1 (BCLC1) cohort]. Three additional cohorts were analyzed for AGT1 (rs699) and AGT2 (rs4762) polymorphisms-initially identified as predictors of DAEs: BCLC2 (n = 79), Northern Italy (n = 221) and Naples (n = 69) cohorts, respectively. The relation between SNPs and DAEs and death were assessed by univariate and multivariate Cox regression models, and presented with hazard ratios and their 95% confidence intervals (95%CI). RESULTS The BCLC1 cohort showed that patients with arterial hypertension (AHT) (HR = 1.61; P value = 0.007) and/or AGT SNPs had an increased risk of DAEs. Thereafter, AGT2 (rs4762) AA genotype was found to be linked to a statistically significant increased probability of DAEs (HR = 5.97; P value = 0.0201, AA vs GG) in the Northern Italy cohort by multivariate analysis adjusted for BCLC stage, ECOG-PS, diabetes and AHT. The value of this genetic marker was externally validated in the cohort combining the BCLC1, BCLC2 and Naples cohorts [HR = 3.12 (95%CI: 1.2-8.14), P value = 0.0199, AGT2 (rs4762) AA vs AG genotype and HR = 2.73 (95%CI: 1.18-6.32) P value = 0.0188, AGT2 (rs4762) AA vs GG genotype]. None of the other gene variants tested were found to be associated with the risk of DAE development. CONCLUSION DAE development in HCC patients receiving TKIs could be explained by the AGT2 (rs4762) gene variant. If validated in other anti-oncogenic treatments, it might be considered a good prognosis marker.
Collapse
Affiliation(s)
- Víctor Sapena
- Barcelona Clinic Liver Cancer Group, Liver Unit, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi Sunyer, Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas, Barcelona 08036, Spain
- Universidad de Barcelona, Barcelona 08036, Spain
| | - Massimo Iavarone
- Division of Gastroenterology and Hepatology, Foundation Istituto di Ricovero e Cura a Carattere Scientifico di Natura Pubblica Ca' Granda Ospedale Maggiore Policlinico, Milano 20122, Italy
| | - Loreto Boix
- Barcelona Clinic Liver Cancer Group, Liver Unit, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi Sunyer, Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas, Barcelona 08036, Spain
| | - Floriana Facchetti
- Gastroenterology and Hepatology Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico di Natura Pubblica Cà Granda Ospedale Maggiore Policlinico, University of Milan, Milan 20100, Italy
| | - Maria Guarino
- Department of Clinical Medicine and Surgery, Gastroenterology Unit, University of Naples "Federico II", Napoli 80100, Italy
| | - Marco Sanduzzi Zamparelli
- Barcelona Clinic Liver Cancer Group, Liver Unit, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi Sunyer, Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas, Barcelona 08036, Spain
- Universidad de Barcelona, Barcelona 08036, Spain
- Department of Clinical Medicine and Surgery, Gastroenterology and Hepatology, Federico II University of Naples, Naples 80131, Italy
| | - Alessandro Granito
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, Istituto di Ricovero e Cura a Carattere Scientifico di Natura Pubblica Azienda Ospedaliero-Universitaria di Bologna, Bologna 40139, Italy
- Department of Medical and Surgical Sciences, University of Bologna, Bologna 40139, Italy
| | - Esther Samper
- Barcelona Clinic Liver Cancer Group, Liver Unit, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi Sunyer, Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas, Barcelona 08036, Spain
| | - Mario Scartozzi
- Department of Medical Oncology, University of Cagliari, Cagliari 45698, Italy
| | - Josep Corominas
- Barcelona Clinic Liver Cancer Group, Liver Unit, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi Sunyer, Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas, Barcelona 08036, Spain
| | - Giorgia Marisi
- Biosciences Laboratory, Istituto di Ricovero e Cura a Carattere Scientifico di Natura Pubblica, Istituto Romagnolo per lo Studio dei Tumori "Dino Amadori", Meldola 47014, Italy
| | - Alba Díaz
- Barcelona Clinic Liver Cancer Group, Liver Unit, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi Sunyer, Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas, Barcelona 08036, Spain
- Universidad de Barcelona, Barcelona 08036, Spain
- Department of Pathology, Hospital Clínic de Barcelona, Universitat de Barcelona, Barcelona 08036, Spain
| | - Andrea Casadei-Gardini
- School of Medicine, Vita-Salute San Raffaele University, Milan 20132, Italy
- Unit of Oncology, Università Vita-Salute, Istituto di Ricovero e Cura a Carattere Scientifico di Natura Pubblica-San Raffaele Scientific Institute, Milan 20132, Italy
| | - Laura Gramantieri
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, Istituto di Ricovero e Cura a Carattere Scientifico di Natura Pubblica Azienda Ospedaliero, Bologna 40138, Italy
| | - Pietro Lampertico
- Division of Gastroenterology and Hepatology, Foundation Istituto di Ricovero e Cura a Carattere Scientifico di Natura Pubblica Ca' Granda Ospedale Maggiore Policlinico, Milano 20122, Italy
- Department of Pathophysiology and Transplantation, Colorectal Cancer "A. M. and A. Migliavacca" Center for Liver Disease, University of Milan, Milano 20122, Italy
| | - Filomena Morisco
- Department of Clinical Medicine and Surgery, Gastroenterology Unit, University of Naples Federico II, Naples 80131, Italy
| | - Ferran Torres
- Medical Statistics Core Facility, Institut d'Investigacions Biomèdiques August Pi Sunyer (IDIBAPS), Hospital Clinic Barcelona, Barcelona 08036, Spain
- Biostatistics Unit, Faculty of Medicine, Universitat Autònoma de Barcelona, Cerdanyola 08193, Spain
| | - Jordi Bruix
- Barcelona Clinic Liver Cancer Group, Liver Unit, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi Sunyer, Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas, Barcelona 08036, Spain
- Universidad de Barcelona, Barcelona 08036, Spain
| | - María Reig
- Barcelona Clinic Liver Cancer Group, Liver Unit, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi Sunyer, Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas, Barcelona 08036, Spain
- Universidad de Barcelona, Barcelona 08036, Spain
| |
Collapse
|
164
|
Petraina A, Nogales C, Krahn T, Mucke H, Lüscher TF, Fischmeister R, Kass DA, Burnett JC, Hobbs AJ, Schmidt HHHW. Cyclic GMP modulating drugs in cardiovascular diseases: mechanism-based network pharmacology. Cardiovasc Res 2022; 118:2085-2102. [PMID: 34270705 PMCID: PMC9302891 DOI: 10.1093/cvr/cvab240] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 07/14/2021] [Indexed: 12/13/2022] Open
Abstract
Mechanism-based therapy centred on the molecular understanding of disease-causing pathways in a given patient is still the exception rather than the rule in medicine, even in cardiology. However, recent successful drug developments centred around the second messenger cyclic guanosine-3'-5'-monophosphate (cGMP), which is regulating a number of cardiovascular disease modulating pathways, are about to provide novel targets for such a personalized cardiovascular therapy. Whether cGMP breakdown is inhibited or cGMP synthesis is stimulated via guanylyl cyclases or their upstream regulators in different cardiovascular disease phenotypes, the outcomes seem to be so far uniformly protective. Thus, a network of cGMP-modulating drugs has evolved that act in a mechanism-based, possibly causal manner in a number of cardiac conditions. What remains a challenge is the detection of cGMPopathy endotypes amongst cardiovascular disease phenotypes. Here, we review the growing clinical relevance of cGMP and provide a glimpse into the future on how drugs interfering with this pathway may change how we treat and diagnose cardiovascular diseases altogether.
Collapse
Affiliation(s)
- Alexandra Petraina
- Department of Pharmacology and Personalised Medicine, School for Mental Health and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Cristian Nogales
- Department of Pharmacology and Personalised Medicine, School for Mental Health and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Thomas Krahn
- Department of Pharmacology and Personalised Medicine, School for Mental Health and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Hermann Mucke
- H.M. Pharma Consultancy, Enenkelstrasse 28/32, A-1160, Vienna, Austria
| | - Thomas F Lüscher
- Royal Brompton & Harefield Hospitals, Heart Division and National Heart and Lung Institute, Guy Scadding Building, Imperial College, Dovehouse Street London SW3 6LY, United Kingdom
- Center for Molecular Cardiology, Schlieren Campus, University of Zurich, Wagistreet 12, CH-8952 Schlieren, Switzerland
| | - Rodolphe Fischmeister
- INSERM UMR-S 1180, Faculty of Pharmacy, Université Paris-Saclay, F-92296 Châtenay-Malabry, France
| | - David A Kass
- Division of Cardiology, Department of Medicine, Ross Research Building, Rm 858, Johns Hopkins Medical Institutions, 720 Rutland Avenue, Baltimore, MD 21205, USA
| | - John C Burnett
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First St. SW, Rochester, MN 55905, USA
| | - Adrian J Hobbs
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, EC1M 6BQ, London, UK
| | - Harald H H W Schmidt
- Department of Pharmacology and Personalised Medicine, School for Mental Health and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| |
Collapse
|
165
|
Berillo O, Huo KG, Richer C, Fraulob-Aquino JC, Briet M, Lipman ML, Sinnett D, Paradis P, Schiffrin EL. Distinct transcriptomic profile of small arteries of hypertensive patients with chronic kidney disease identified miR-338-3p targeting GPX3 and PTPRS. J Hypertens 2022; 40:1394-1405. [PMID: 35703228 DOI: 10.1097/hjh.0000000000003160] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
OBJECTIVE Hypertension is associated with vascular injury, which contributes to end-organ damage. MicroRNAs regulating mRNAs have been shown to play a role in vascular injury in hypertensive mice. We aimed to identify differentially expressed microRNAs and their mRNA targets in small arteries of hypertensive patients with/without chronic kidney disease (CKD) to shed light on the pathophysiological molecular mechanisms of vascular remodeling. METHODS AND RESULTS Normotensive individuals and hypertensive patients with/without CKD were recruited ( n = 15-16 per group). Differentially expressed microRNAs and mRNAs were identified uniquely associated with hypertension (microRNAs: 10, mRNAs: 68) or CKD (microRNAs: 68, mRNAs: 395), and in both groups (microRNAs: 2, mRNAs: 32) with a P less than 0.05 and a fold change less than or greater than 1.3 in subcutaneous small arteries ( n = 14-15). One of the top three differentially expressed microRNAs, miR-338-3p that was down-regulated in CKD, presented the best correlation between RNA sequencing and reverse transcription-quantitative PCR (RT-qPCR, R2 = 0.328, P < 0.001). Profiling of human aortic vascular cells showed that miR-338-3p was mostly expressed in endothelial cells. Two of the selected top nine up-regulated miR-338-3p predicted targets, glutathione peroxidase 3 ( GPX3 ) and protein tyrosine phosphatase receptor type S ( PTPRS ), were validated with mimics by RT-qPCR in human aortic endothelial cells ( P < 0.05) and by a luciferase assay in HEK293T cells ( P < 0.05). CONCLUSION A distinct transcriptomic profile was observed in gluteal subcutaneous small arteries of hypertensive patients with CKD. Down-regulated miR-338-3p could contribute to GPX3 and PTPRS up-regulation via the canonical microRNA targeting machinery in hypertensive patients with CKD. http://links.lww.com/HJH/C27.
Collapse
Affiliation(s)
- Olga Berillo
- Hypertension and Vascular Research Unit, Lady Davis Institute for Medical Research
| | - Ku-Geng Huo
- Hypertension and Vascular Research Unit, Lady Davis Institute for Medical Research
| | - Chantal Richer
- Division of Hematology-Oncology, Research Center, CHU Ste-Justine
| | | | - Marie Briet
- Hypertension and Vascular Research Unit, Lady Davis Institute for Medical Research
- INSERM U1083, CNRS UMR 6214, Centre Hospitalo-Universitaire d'Angers, Université d'Angers, Angers, France
| | - Mark L Lipman
- Hypertension and Vascular Research Unit, Lady Davis Institute for Medical Research
- Department of Medicine, Sir Mortimer B. Davis-Jewish General Hospital, McGill University
| | - Daniel Sinnett
- Division of Hematology-Oncology, Research Center, CHU Ste-Justine
- Department of Pediatrics, Faculty of Medicine, Université de Montréal, Montréal, Canada
| | - Pierre Paradis
- Hypertension and Vascular Research Unit, Lady Davis Institute for Medical Research
| | - Ernesto L Schiffrin
- Hypertension and Vascular Research Unit, Lady Davis Institute for Medical Research
- Department of Medicine, Sir Mortimer B. Davis-Jewish General Hospital, McGill University
| |
Collapse
|
166
|
Samuelson DR, Haq S, Knoell DL. Divalent Metal Uptake and the Role of ZIP8 in Host Defense Against Pathogens. Front Cell Dev Biol 2022; 10:924820. [PMID: 35832795 PMCID: PMC9273032 DOI: 10.3389/fcell.2022.924820] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 05/26/2022] [Indexed: 01/13/2023] Open
Abstract
Manganese (Mn) and Zinc (Zn) are essential micronutrients whose concentration and location within cells are tightly regulated at the onset of infection. Two families of Zn transporters (ZIPs and ZnTs) are largely responsible for regulation of cytosolic Zn levels and to a certain extent, Mn levels, although much less is known regarding Mn. The capacity of pathogens to persevere also depends on access to micronutrients, yet a fundamental gap in knowledge remains regarding the importance of metal exchange at the host interface, often referred to as nutritional immunity. ZIP8, one of 14 ZIPs, is a pivotal importer of both Zn and Mn, yet much remains to be known. Dietary Zn deficiency is common and commonly occurring polymorphic variants of ZIP8 that decrease cellular metal uptake (Zn and Mn), are associated with increased susceptibility to infection. Strikingly, ZIP8 is the only Zn transporter that is highly induced following bacterial exposure in key immune cells involved with host defense against leading pathogens. We postulate that mobilization of Zn and Mn into key cells orchestrates the innate immune response through regulation of fundamental defense mechanisms that include phagocytosis, signal transduction, and production of soluble host defense factors including cytokines and chemokines. New evidence also suggests that host metal uptake may have long-term consequences by influencing the adaptive immune response. Given that activation of ZIP8 expression by pathogens has been shown to influence parenchymal, myeloid, and lymphoid cells, the impact applies to all mucosal surfaces and tissue compartments that are vulnerable to infection. We also predict that perturbations in metal homeostasis, either genetic- or dietary-induced, has the potential to impact bacterial communities in the host thereby adversely impacting microbiome composition. This review will focus on Zn and Mn transport via ZIP8, and how this vital metal transporter serves as a "go to" conductor of metal uptake that bolsters host defense against pathogens. We will also leverage past studies to underscore areas for future research to better understand the Zn-, Mn- and ZIP8-dependent host response to infection to foster new micronutrient-based intervention strategies to improve our ability to prevent or treat commonly occurring infectious disease.
Collapse
Affiliation(s)
- Derrick R. Samuelson
- Division of Pulmonary, Critical Care, and Sleep, Department of Internal Medicine, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Sabah Haq
- Department of Pharmacy Practice and Science, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, United States
| | - Daren L. Knoell
- Department of Pharmacy Practice and Science, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, United States,*Correspondence: Daren L. Knoell,
| |
Collapse
|
167
|
Parcha V, Irvin MR, Lange LA, Armstrong ND, Pampana A, Meyer M, Judd SE, Arora G, Arora P. Corin Missense Variants, Blood Pressure, and Hypertension in 11 322 Black Individuals: Insights From REGARDS and the Jackson Heart Study. J Am Heart Assoc 2022; 11:e025582. [PMID: 35699180 PMCID: PMC9238660 DOI: 10.1161/jaha.121.025582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 04/27/2022] [Indexed: 11/18/2022]
Abstract
Background Corin enzyme contributes to the processing of inactive natriuretic peptides to bioactive hormones. In Black individuals, Corin gene variants (rs111253292 [Q568P] and rs75770792 [T555I]) have been previously reported to have a modest association with blood pressure (BP) and hypertension. Methods and Results We evaluated the association of Corin genotype with BP traits, prevalent hypertension, and incident hypertension among self-identified 11 322 Black Americans in the REGARDS (Reasons for Geographic and Racial Differences in Stroke) study and the JHS (Jackson Heart Study) using multivariable-adjusted regression modeling. Multivariable-adjusted genotype-stratified differences in NT-proBNP (N-terminal pro-B-type natriuretic peptide) and BNP (B-type natriuretic peptide) levels were assessed. Genotype-stratified NPPA and NPPB expression differences in healthy organ donor left atrial and left ventricular heart tissue (N=15) were also examined. The rs111253292 genotype was not associated with systolic BP (β±SE, 0.42±0.58; -1.24±0.82), diastolic BP (0.51±0.33; -0.41±0.46), mean arterial pressure (0.48±0.38; -0.68±0.51), and prevalent hypertension (odds ratio [OR], 0.93 [95% CI, 0.80-1.09]; OR, 0.79 [95% CI, 0.61-1.01]) in both REGARDS and JHS, respectively. The rs75770792 genotype was not associated with systolic BP (0.48±0.58; -1.26±0.81), diastolic BP (0.52±0.33; -0.33±0.45), mean arterial pressure (0.50±0.38; -0.63±0.50), and prevalent hypertension (OR, 1.02 [95% CI, 0.84-1.23]; OR, 0.87 [95% CI, 0.67-1.13]) in both cohorts, respectively. The Corin genotype was also not associated with incident hypertension (OR, 1.35 [95% CI, 0.94-1.93]; OR, 0.95 [95% CI, 0.64-1.39]) in the study cohorts. The NT-proBNP levels in REGARDS and BNP levels in JHS were similar between the Corin genotype groups. In heart tissue, the NPPA and NPPB expression was similar between the genotype groups. Conclusions Corin gene variants observed more commonly in Black individuals are not associated with differences in NP expression, circulating NP levels, and BP or hypertension as previously reported in candidate gene studies. Understanding the genetic determinants of complex cardiovascular traits in underrepresented populations requires further evaluation.
Collapse
Affiliation(s)
- Vibhu Parcha
- Division of Cardiovascular DiseaseUniversity of Alabama at BirminghamBirminghamAL
| | | | - Leslie A. Lange
- Division of Biomedical Informatics and Personalized MedicineDepartment of MedicineUniversity of Colorado School of MedicineAuroraCO
- Department of EpidemiologyUniversity of Colorado School of Public HealthAuroraCO
| | | | - Akhil Pampana
- Division of Cardiovascular DiseaseUniversity of Alabama at BirminghamBirminghamAL
| | - Mariah Meyer
- Division of Biomedical Informatics and Personalized MedicineDepartment of MedicineUniversity of Colorado School of MedicineAuroraCO
| | - Suzanne E. Judd
- Department of BiostatisticsUniversity of Alabama at BirminghamBirminghamAL
| | - Garima Arora
- Division of Cardiovascular DiseaseUniversity of Alabama at BirminghamBirminghamAL
| | - Pankaj Arora
- Division of Cardiovascular DiseaseUniversity of Alabama at BirminghamBirminghamAL
- Section of CardiologyBirmingham Veterans Affairs Medical CenterBirminghamAL
| |
Collapse
|
168
|
Kurniansyah N, Goodman MO, Kelly TN, Elfassy T, Wiggins KL, Bis JC, Guo X, Palmas W, Taylor KD, Lin HJ, Haessler J, Gao Y, Shimbo D, Smith JA, Yu B, Feofanova EV, Smit RAJ, Wang Z, Hwang SJ, Liu S, Wassertheil-Smoller S, Manson JE, Lloyd-Jones DM, Rich SS, Loos RJF, Redline S, Correa A, Kooperberg C, Fornage M, Kaplan RC, Psaty BM, Rotter JI, Arnett DK, Morrison AC, Franceschini N, Levy D, Sofer T. A multi-ethnic polygenic risk score is associated with hypertension prevalence and progression throughout adulthood. Nat Commun 2022; 13:3549. [PMID: 35729114 PMCID: PMC9213527 DOI: 10.1038/s41467-022-31080-2] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 05/31/2022] [Indexed: 12/12/2022] Open
Abstract
In a multi-stage analysis of 52,436 individuals aged 17-90 across diverse cohorts and biobanks, we train, test, and evaluate a polygenic risk score (PRS) for hypertension risk and progression. The PRS is trained using genome-wide association studies (GWAS) for systolic, diastolic blood pressure, and hypertension, respectively. For each trait, PRS is selected by optimizing the coefficient of variation (CV) across estimated effect sizes from multiple potential PRS using the same GWAS, after which the 3 trait-specific PRSs are combined via an unweighted sum called "PRSsum", forming the HTN-PRS. The HTN-PRS is associated with both prevalent and incident hypertension at 4-6 years of follow up. This association is further confirmed in age-stratified analysis. In an independent biobank of 40,201 individuals, the HTN-PRS is confirmed to be predictive of increased risk for coronary artery disease, ischemic stroke, type 2 diabetes, and chronic kidney disease.
Collapse
Affiliation(s)
- Nuzulul Kurniansyah
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, USA
| | - Matthew O Goodman
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Tanika N Kelly
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
| | - Tali Elfassy
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Kerri L Wiggins
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Joshua C Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Walter Palmas
- Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Kent D Taylor
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Henry J Lin
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Jeffrey Haessler
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Yan Gao
- The Jackson Heart Study, University of Mississippi Medical Center, Jackson, MS, USA
| | - Daichi Shimbo
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Jennifer A Smith
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Bing Yu
- Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Elena V Feofanova
- Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Roelof A J Smit
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Zhe Wang
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Shih-Jen Hwang
- Department of Biostatistics, Boston University, Boston, MA, USA
| | - Simin Liu
- Center for Global Cardiometabolic Health and Departments of Epidemiology, Medicine, and Surgery, Brown University, Providence, RI, USA
| | - Sylvia Wassertheil-Smoller
- Department of Epidemiology & Population Health, Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - JoAnn E Manson
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | | | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Ruth J F Loos
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Susan Redline
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Adolfo Correa
- Departments of Medicine and Pediatrics, University of Mississippi Medical Center, Jackson, MS, USA
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Myriam Fornage
- Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, USA
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Robert C Kaplan
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Departments of Medicine, Epidemiology, and Health Systems and Population Health, University of Washington, Seattle, WA, USA
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Donna K Arnett
- College of Public Health, University of Kentucky, Lexington, KY, USA
| | - Alanna C Morrison
- Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Nora Franceschini
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
| | - Daniel Levy
- The Population Sciences Branch of the National Heart, Lung and Blood Institute, Bethesda, MD, USA
- The Framingham Heart Study, Framingham, MA, USA
| | - Tamar Sofer
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, USA.
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
169
|
Khor BH, Komnenov D, Rossi NF. Impact of Dietary Fructose and High Salt Diet: Are Preclinical Studies Relevant to Asian Societies? Nutrients 2022; 14:2515. [PMID: 35745245 PMCID: PMC9227020 DOI: 10.3390/nu14122515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/08/2022] [Accepted: 06/11/2022] [Indexed: 02/01/2023] Open
Abstract
Fructose consumption, especially in food additives and sugar-sweetened beverages, has gained increasing attention due to its potential association with obesity and metabolic syndrome. The relationship between fructose and a high-salt diet, leading to hypertension and other deleterious cardiovascular parameters, has also become more evident, especially in preclinical studies. However, these studies have been modeled primarily on Western diets. The purpose of this review is to evaluate the dietary habits of individuals from China, Japan, and Korea, in light of the existing preclinical studies, to assess the potential relevance of existing data to East Asian societies. This review is not intended to be exhaustive, but rather to highlight the similarities and differences that should be considered in future preclinical, clinical, and epidemiologic studies regarding the impact of dietary fructose and salt on blood pressure and cardiovascular health worldwide.
Collapse
Affiliation(s)
- Ban Hock Khor
- Faculty of Food Science and Nutrition, Universiti Malaysia Sabah, Kota Kinabalu 88400, Malaysia;
| | - Dragana Komnenov
- Department of Internal Medicine, Wayne State University, Detroit, MI 48201, USA;
| | - Noreen F. Rossi
- Department of Internal Medicine, Wayne State University, Detroit, MI 48201, USA;
- Department of Physiology, Wayne State University, Detroit, MI 48201, USA
- Division of Research, John D. Dingell VA Medical Center, Detroit, MI 38201, USA
| |
Collapse
|
170
|
Dam V, Onland-Moret NC, Burgess S, Chirlaque MD, Peters SAE, Schuit E, Tikk K, Weiderpass E, Oliver-Williams C, Wood AM, Tjønneland A, Dahm CC, Overvad K, Boutron-Ruault MC, Schulze MB, Trichopoulou A, Ferrari P, Masala G, Krogh V, Tumino R, Matullo G, Panico S, Boer JMA, Verschuren WMM, Waaseth M, Pérez MJS, Amiano P, Imaz L, Moreno-Iribas C, Melander O, Harlid S, Nordendahl M, Wennberg P, Key TJ, Riboli E, Santiuste C, Kaaks R, Katzke V, Langenberg C, Wareham NJ, Schunkert H, Erdmann J, Willenborg C, Hengstenberg C, Kleber ME, Delgado G, März W, Kanoni S, Dedoussis G, Deloukas P, Nikpay M, McPherson R, Scholz M, Teren A, Butterworth AS, van der Schouw YT. Genetically Determined Reproductive Aging and Coronary Heart Disease: A Bidirectional 2-sample Mendelian Randomization. J Clin Endocrinol Metab 2022; 107:e2952-e2961. [PMID: 35306566 PMCID: PMC9202700 DOI: 10.1210/clinem/dgac171] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Indexed: 12/20/2022]
Abstract
BACKGROUND Accelerated reproductive aging, in women indicated by early natural menopause, is associated with increased coronary heart disease (CHD) risk in observational studies. Conversely, an adverse CHD risk profile has been suggested to accelerate menopause. OBJECTIVES To study the direction and evidence for causality of the relationship between reproductive aging and (non-)fatal CHD and CHD risk factors in a bidirectional Mendelian randomization (MR) approach, using age at natural menopause (ANM) genetic variants as a measure for genetically determined reproductive aging in women. We also studied the association of these variants with CHD risk (factors) in men. DESIGN Two-sample MR, using both cohort data as well as summary statistics, with 4 methods: simple and weighted median-based, standard inverse-variance weighted (IVW) regression, and MR-Egger regression. PARTICIPANTS Data from EPIC-CVD and summary statistics from UK Biobank and publicly available genome-wide association studies were pooled for the different analyses. MAIN OUTCOME MEASURES CHD, CHD risk factors, and ANM. RESULTS Across different methods of MR, no association was found between genetically determined reproductive aging and CHD risk in women (relative risk estimateIVW = 0.99; 95% confidence interval (CI), 0.97-1.01), or any of the CHD risk factors. Similarly, no associations were found in men. Neither did the reversed analyses show evidence for an association between CHD (risk factors) and reproductive aging. CONCLUSION Genetically determined reproductive aging is not causally associated with CHD risk (factors) in women, nor were the genetic variants associated in men. We found no evidence for a reverse association in a combined sample of women and men.
Collapse
Affiliation(s)
- Veerle Dam
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, GA 3508 Utrecht, the Netherlands
- Netherlands Heart Institute, DG 3501 Utrecht, the Netherlands
| | - N Charlotte Onland-Moret
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, GA 3508 Utrecht, the Netherlands
| | - Stephen Burgess
- MRC Biostatistics Unit, University of Cambridge, Cambridge CB2 0SR, UK
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Homerton College, Cambridge, UK
| | - Maria-Dolores Chirlaque
- Department of Epidemiology, Regional Health Authority, IMIB-Arrixaca, Murcia University, 30001 Murcia, Spain
- Department of Public Health and Clinical Medicine, Umea University, 901 87 Umea, Sweden
| | - Sanne A E Peters
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, GA 3508 Utrecht, the Netherlands
- The George Institute for Global Health, Imperial College London, London W12 0BZ, UK
| | - Ewoud Schuit
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, GA 3508 Utrecht, the Netherlands
| | - Kaja Tikk
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Centre (DKFZ), 69120 Heidelberg, Germany
- German Cancer Consortium, DKFZ, 69120 Heidelberg, Germany
| | - Elisabete Weiderpass
- International Agency for Research on Cancer, World Health Organization, 69372 Lyon, France
| | - Clare Oliver-Williams
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Homerton College, Cambridge, UK
| | - Angela M Wood
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Anne Tjønneland
- Danish Cancer Society Research Center, DK-2100 Copenhagen, Denmark
- Department of Public Health, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Christina C Dahm
- Department of Public Health, Aarhus University, 8000 Aarhus, Denmark
| | - Kim Overvad
- Department of Public Health, Aarhus University, 8000 Aarhus, Denmark
- Department of Cardiology, Aalborg University Hospital, 9000 Aalborg, Denmark
| | - Marie-Christine Boutron-Ruault
- INSERM, Centre for Research in Epidemiology and Population Health, U1018, Nutrition, Hormones, and Women’s Health Team, Institut Gustave Roussy, 94 805 Villejuif, France
| | - Matthias B Schulze
- Department of Epidemiology, German Institute of Human Nutrition, Potsdam-Rehbruecke, Nuthetal, Germany; Institute of Nutritional Science, University of Potsdam, 14558 Nuthetal, Germany
| | - Antonia Trichopoulou
- Hellenic Health Foundation, 115 27 Athens, Greece
- WHO Collaborating Center for Nutrition and Health, Unit of Nutritional Epidemiology and Nutrition in Public Health, Department of Hygiene, Epidemiology and Medical Statistics, School of Medicine, National and Kapodistrian University of Athens, Athens 115 27, Greece
| | - Pietro Ferrari
- International Agency for Research on Cancer, World Health Organization, 69372 Lyon, France
| | - Giovanna Masala
- Cancer Risk Factors and Life-Style Epidemiology Unit, Institute for Cancer Research, Prevention and Clinical Network - ISPRO, 50139 Florence, Italy
| | - Vittorio Krogh
- Epidemiology and Prevention Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Rosario Tumino
- Cancer Registry and Histopathology Department, “Civic - M.P. Arezzo” hospital, ASPRagusa, 97100 Ragusa, Italy
| | - Giuseppe Matullo
- Department of Medical Sciences, University of Torino, 10124 Torino, Italy
- Italian Institute for Genomic Medicine–IIGM/HuGeF, 10126 Torino, Italy
| | - Salvatore Panico
- Dipartimento di medicina clinica e chirurgia, Federico II University, 80126 Naples, Italy
| | - Jolanda M A Boer
- National Institute for Public Health and the Environment, 3720 BA Bilthoven, the Netherlands
| | - W M Monique Verschuren
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, GA 3508 Utrecht, the Netherlands
- National Institute for Public Health and the Environment, 3720 BA Bilthoven, the Netherlands
| | - Marit Waaseth
- Department of Pharmacy, Faculty of Health Sciences, UiT the Arctic University of Norway, N-9037 Tromsø, Norway
| | - Maria José Sánchez Pérez
- Escuela Andaluza de Salud Pública. Instituto de Investigación Biosanitaria ibs.GRANADA, Universidad de Granada, 18011 Granada, Spain
- CIBER de Epidemiología y Salud Pública (CIBERESP), 28029 Madrid, Spain
| | - Pilar Amiano
- CIBER de Epidemiología y Salud Pública (CIBERESP), 28029 Madrid, Spain
- Public Health Division of Gipuzkoa, Biodonostia Research Institute, 20014 San Sebastian, Spain
| | - Liher Imaz
- CIBER de Epidemiología y Salud Pública (CIBERESP), 28029 Madrid, Spain
- Public Health Division of Gipuzkoa, Biodonostia Research Institute, 20014 San Sebastian, Spain
| | - Conchi Moreno-Iribas
- Instituto de Salud Pública de Navarra, IdiSNA, Navarre Institute for Health Research, REDISSEC, 31008, Pamplona, Spain
| | - Olle Melander
- Department of Clinical Sciences, Lund University, SE-221 00 Malmö, Sweden
| | - Sophia Harlid
- Department of Radiation Sciences, Oncology, Umea University, 901 87 Umea, Sweden
| | - Maria Nordendahl
- Department of Public Health and Clinical Medicine, Umea University, 901 87 Umea, Sweden
| | - Patrik Wennberg
- Department of Public Health and Clinical Medicine, Umea University, 901 87 Umea, Sweden
| | - Timothy J Key
- Nuffield Department of Population Health, University of Oxford, OX3 7LF Oxford, England
| | - Elio Riboli
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, SW7 2AZ London, UK
| | - Carmen Santiuste
- CIBER de Epidemiología y Salud Pública (CIBERESP), 28029 Madrid, Spain
- Department of Epidemiology, Murcia Regional Health Authority, IMIB-Arrixaca, 30001 Murcia, Spain
| | - Rudolf Kaaks
- Division of Cancer Epidemiology, DKFZ, Foundation under Public Law, D-69120 Heidelberg, Germany
| | - Verena Katzke
- Division of Cancer Epidemiology, DKFZ, Foundation under Public Law, D-69120 Heidelberg, Germany
| | - Claudia Langenberg
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, CB2 0SL Cambridge, UK
| | - Nicholas J Wareham
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, CB2 0SL Cambridge, UK
| | - Heribert Schunkert
- Deutsches Herzzentrum München, Technische Universität München, 80636 Munich, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, 80636 Munich, Germany
| | - Jeanette Erdmann
- Institute for Cardiogenetics, University of Lübeck, 23562 Lübeck, Germany
| | | | - Christian Hengstenberg
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, 1090 Vienna, Austria
| | - Marcus E Kleber
- Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Graciela Delgado
- Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Winfried März
- Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Synlab Academy, Synlab Holding Deutschland GmbH, 68167 Mannheim, Germany
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, 8036 Graz, Austria
| | - Stavroula Kanoni
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 4NS, UK
| | - George Dedoussis
- Department of Nutrition-Dietetics/Harokopio University, 17671 Athens, Greece
| | - Panos Deloukas
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 4NS, UK
- Centre for Genomic Health, Queen Mary University of London, London E1 4NS, UK
- Princess Al-Jawhara Al-Brahim Centre of Excellence in Research of Hereditary Disorders, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Majid Nikpay
- Ruddy Canadian Cardiovascular Genetics Centre, University of Ottawa Heart Institute, Ottawa, Ontario K1Y 4W7, Canada
| | - Ruth McPherson
- Ruddy Canadian Cardiovascular Genetics Centre, University of Ottawa Heart Institute, Ottawa, Ontario K1Y 4W7, Canada
| | - Markus Scholz
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, 04107 Leipzig, Germany
- LIFE Research Center for Civilization Diseases, University of Leipzig, 04103 Leipzig, Germany
| | - Andrej Teren
- LIFE Research Center for Civilization Diseases, University of Leipzig, 04103 Leipzig, Germany
- Heart Center Leipzig, 04289 Leipzig, Germany
| | - Adam S Butterworth
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Yvonne T van der Schouw
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, GA 3508 Utrecht, the Netherlands
| |
Collapse
|
171
|
A Review of Vascular Traits and Assessment Techniques, and Their Heritability. Artery Res 2022. [DOI: 10.1007/s44200-022-00016-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
AbstractVarious tools are available to assess atherosclerosis, arterial stiffening, and endothelial function. They offer utility in the assessment of hypertensive phenotypes, in cardiovascular risk prediction, and as surrogate endpoints in clinical trials. We explore the relative influence of participant genetics, with reference to large-scale genomic studies, population-based cohorts, and candidate gene studies. We find heritability estimates highest for carotid intima-media thickness (CIMT 35–65%), followed by pulse wave velocity as a measure of arterial stiffness (26–43%), and flow mediated dilatation as a surrogate for endothelial function (14–39%); data were lacking for peripheral artery tonometry. We furthermore examine genes and polymorphisms relevant to each technique. We conclude that CIMT and pulse wave velocity dominate the existing evidence base, with fewer published genomic linkages for measures of endothelial function. We finally make recommendations regarding planning and reporting of data relating to vascular assessment techniques, particularly when genomic data are also available, to facilitate integration of these tools into cardiovascular disease research.
Collapse
|
172
|
Galley JC, Hahn SA, Miller MP, Durgin BG, Jackson EK, Stocker SD, Straub AC. Angiotensin II augments renal vascular smooth muscle soluble GC expression via an AT 1 receptor-forkhead box subclass O transcription factor signalling axis. Br J Pharmacol 2022; 179:2490-2504. [PMID: 33963547 PMCID: PMC8883839 DOI: 10.1111/bph.15522] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/18/2021] [Accepted: 04/23/2021] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND AND PURPOSE Reduced renal blood flow triggers activation of the renin-angiotensin-aldosterone system (RAAS) leading to renovascular hypertension. Renal vascular smooth muscle expression of the NO receptor, soluble GC (sGC), modulates the vasodilator response needed to control renal vascular tone and blood flow. Here, we tested if angiotensin II (Ang II) affects sGC expression via an AT1 receptor-forkhead box subclass O (FoxO) transcription factor dependent mechanism. EXPERIMENTAL APPROACH Using a murine two-kidney-one-clip (2K1C) renovascular hypertension model, we measured renal artery vasodilatory function and sGC expression. Additionally, we conducted cell culture studies using rat renal pre-glomerular smooth muscle cells (RPGSMCs) to test the in vitro mechanistic effects of Ang II treatment on sGC expression and downstream function. KEY RESULTS Contralateral, unclipped renal arteries in 2K1C mice showed increased NO-dependent vasorelaxation compared to sham control mice. Immunofluorescence studies revealed increased sGC protein expression in 2K1C contralateral renal arteries over sham controls. RPGSMCs treated with Ang II caused a significant up-regulation of sGC mRNA and protein expression as well as downstream sGC-dependent signalling. Ang II signalling effects on sGC expression occurred through an AT1 receptor and FoxO transcription factor-dependent mechanism at both the mRNA and protein expression levels. CONCLUSION AND IMPLICATIONS Renal artery smooth muscle, in vivo and in vitro, up-regulates expression of sGC following RAAS activity. In both cases, up-regulation of sGC leads to increased downstream cGMP signalling, suggesting a previously unrecognized protective mechanism to improve renal blood flow in the uninjured contralateral renal artery. LINKED ARTICLES This article is part of a themed issue on cGMP Signalling in Cell Growth and Survival. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v179.11/issuetoc.
Collapse
Affiliation(s)
- Joseph C. Galley
- Heart, Lung, Blood and Vascular Medicine Institute,
University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Pharmacology and Chemical Biology, University
of Pittsburgh, Pittsburgh, Pennsylvania
| | - Scott A. Hahn
- Heart, Lung, Blood and Vascular Medicine Institute,
University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Megan P. Miller
- Heart, Lung, Blood and Vascular Medicine Institute,
University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Brittany G. Durgin
- Heart, Lung, Blood and Vascular Medicine Institute,
University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Edwin K. Jackson
- Department of Pharmacology and Chemical Biology, University
of Pittsburgh, Pittsburgh, Pennsylvania
| | - Sean D. Stocker
- Department of Medicine, Renal-Electrolyte Division,
University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Adam C. Straub
- Heart, Lung, Blood and Vascular Medicine Institute,
University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Pharmacology and Chemical Biology, University
of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
173
|
Tapela NM, Collister J, Liu X, Clifton L, Stiby A, Murgia F, Hopewell JC, Hunter DJ. Are polygenic risk scores for systolic blood pressure and LDL-cholesterol associated with treatment effectiveness, and clinical outcomes among those on treatment? Eur J Prev Cardiol 2022; 29:925-937. [PMID: 34864974 DOI: 10.1093/eurjpc/zwab192] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Accepted: 10/25/2021] [Indexed: 12/27/2022]
Abstract
AIMS Many studies have investigated associations between polygenic risk scores (PRS) and the incidence of cardiovascular disease (CVD); few have examined whether risk factor-related PRS predict CVD outcomes among adults treated with risk-modifying therapies. We assessed whether PRS for systolic blood pressure (PRSSBP) and for low-density lipoprotein cholesterol (PRSLDL-C) were associated with achieving SBP and LDL-C-related targets, and with major adverse cardiovascular events (MACE: non-fatal stroke or myocardial infarction, CVD death, and revascularization procedures). METHODS AND RESULTS Using observational data from the UK Biobank (UKB), we calculated PRSSBP and PRSLDL-C and constructed two sub-cohorts of unrelated adults of White British ancestry aged 40-69 years and with no history of CVD, who reported taking medications used in the treatment of hypertension or hypercholesterolaemia. Treatment effectiveness in achieving adequate risk factor control was ascertained using on-treatment blood pressure (BP) or LDL-C levels measured at enrolment (uncontrolled hypertension: BP ≥ 140/90 mmHg; uncontrolled hypercholesterolaemia: LDL-C ≥ 3 mmol/L). We conducted multivariable logistic and Cox regression modelling for incident events, adjusting for socioeconomic characteristics, and CVD risk factors. There were 55 439 participants using BP lowering therapies (51.0% male, mean age 61.0 years, median follow-up 11.5 years) and 33 787 using LDL-C lowering therapies (58.5% male, mean age 61.7 years, median follow-up 11.4 years). PRSSBP was associated with uncontrolled hypertension (odds ratio 1.70; 95% confidence interval: 1.60-1.80) top vs. bottom quintile, equivalent to a 5.4 mmHg difference in SBP, and with MACE [hazard ratio (HR) 1.13; 1.04-1.23]. PRSLDL-C was associated with uncontrolled hypercholesterolaemia (HR 2.78; 2.58-3.00) but was not associated with subsequent MACE. CONCLUSION We extend previous findings in the UKB cohort to examine PRSSBP and PRSLDL-C with treatment effectiveness. Our results indicate that both PRSSBP and PRSLDL-C can help identify individuals who, despite being on treatment, have inadequately controlled SBP and LDL-C, and for SBP are at higher risk for CVD events. This extends the potential role of PRS in clinical practice from identifying patients who may need these interventions to identifying patients who may need more intensive intervention.
Collapse
Affiliation(s)
- Neo M Tapela
- Nuffield Department of Population Health, University of Oxford Big Data Institute, Old Road Campus, Headington, Oxford, OX3 7LF, UK
- Botswana Harvard AIDS Institute Partnership, Princess Marina Hospital, Plot No. 1836, Northring Road, Gaborone, Botswana
- Department of Medicine, Division of Global Health Equity, Brigham and Women's Hospital, 75 Francis Street, Boston, MA 02115, USA
| | - Jennifer Collister
- Nuffield Department of Population Health, University of Oxford Big Data Institute, Old Road Campus, Headington, Oxford, OX3 7LF, UK
| | - Xiaonan Liu
- Nuffield Department of Population Health, University of Oxford Big Data Institute, Old Road Campus, Headington, Oxford, OX3 7LF, UK
| | - Lei Clifton
- Nuffield Department of Population Health, University of Oxford Big Data Institute, Old Road Campus, Headington, Oxford, OX3 7LF, UK
| | - Alexander Stiby
- Nuffield Department of Population Health, University of Oxford Big Data Institute, Old Road Campus, Headington, Oxford, OX3 7LF, UK
| | - Federico Murgia
- Nuffield Department of Population Health, University of Oxford Big Data Institute, Old Road Campus, Headington, Oxford, OX3 7LF, UK
| | - Jemma C Hopewell
- Nuffield Department of Population Health, University of Oxford Big Data Institute, Old Road Campus, Headington, Oxford, OX3 7LF, UK
| | - David J Hunter
- Nuffield Department of Population Health, University of Oxford Big Data Institute, Old Road Campus, Headington, Oxford, OX3 7LF, UK
- Department of Epidemiology, Harvard TH Chan School of Public Health, 677 Huntington Avenue, Boston, MA 02115, USA
| |
Collapse
|
174
|
Georgakis MK, Parodi L, Frerich S, Mayerhofer E, Tsivgoulis G, Pirruccello JP, Slowik A, Rundek T, Malik R, Dichgans M, Rosand J, Anderson CD. Genetic Architecture of Stroke of Undetermined Source: Overlap with Known Stroke Etiologies and Associations with Modifiable Risk Factors. Ann Neurol 2022; 91:640-651. [PMID: 35178771 DOI: 10.1002/ana.26332] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 02/16/2022] [Accepted: 02/16/2022] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Ischemic stroke etiology remains undetermined in 30% of cases. We explored the genetic architecture of stroke classified as undetermined to test if mechanisms and risk factors underlying large-artery atherosclerotic (LAAS), cardioembolic (CES), and small-vessel stroke (SVS) contribute to its pathogenesis. METHODS We analyzed genome-wide data from 16,851 ischemic stroke cases and 32,473 controls. Using polygenic risk scores for LAAS, CES, and SVS, we assessed the genetic overlap with stroke of undetermined source and used pairwise genomewide association study (GWAS-PW) to search for shared loci. We then applied Mendelian randomization (MR) to identify potentially causal risk factors of stroke of undetermined source. RESULTS Genetic risk for LAS, CES, and SVS was associated with stroke of undetermined source pointing to overlap in their genetic architecture. Pairwise analyses revealed 19 shared loci with LAAS, 2 with CES, and 5 with SVS that have been implicated in atherosclerosis-related phenotypes. Genetic liability to both carotid atherosclerosis and atrial fibrillation was associated with stroke of undetermined source, but the association with atrial fibrillation was attenuated after excluding cases with incomplete diagnostic workup. MR analyses showed effects of genetically determinants of blood pressure, diabetes, waist-to-hip ratio, inflammatory pathways (IL-6 signaling, MCP-1/CCL2 levels), and factor XI levels on stroke of undetermined source. INTERPRETATION Stroke of undetermined source shares genetic and vascular risk factors with other stroke subtypes, especially LAAS, thus highlighting the diagnostic limitations of current subtyping approaches. The potentially causal associations with carotid atherosclerosis and atherosclerotic risk factors might have implications for prevention strategies targeting these mechanisms. ANN NEUROL 2022;91:640-651.
Collapse
Affiliation(s)
- Marios K Georgakis
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA
- Program in Medical and Population Genetics, Broad Institute of Harvard and the Massachusetts Institute of Technology, Boston, MA
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
| | - Livia Parodi
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA
- Program in Medical and Population Genetics, Broad Institute of Harvard and the Massachusetts Institute of Technology, Boston, MA
| | - Simon Frerich
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
| | - Ernst Mayerhofer
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA
- Program in Medical and Population Genetics, Broad Institute of Harvard and the Massachusetts Institute of Technology, Boston, MA
| | - Georgios Tsivgoulis
- Second Department of Neurology, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
- Department of Neurology, University of Tennessee Health Sciences Center, Memphis, TN, USA
| | - James P Pirruccello
- Cardiology Division, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad institute of Harvard and the Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Agnieszka Slowik
- Department of Neurology, Jagiellonian University, Krakow, Poland
| | - Tatjana Rundek
- Department of Neurology, Evelyn F. McKnight Brain Institute, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Rainer Malik
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
| | - Martin Dichgans
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- German Centre for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Jonathan Rosand
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA
- Program in Medical and Population Genetics, Broad Institute of Harvard and the Massachusetts Institute of Technology, Boston, MA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA, USA
| | - Christopher D Anderson
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA
- Program in Medical and Population Genetics, Broad Institute of Harvard and the Massachusetts Institute of Technology, Boston, MA
- McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
| |
Collapse
|
175
|
Hypertension: The most common chronic health problem in Spain. A call to action. HIPERTENSION Y RIESGO VASCULAR 2022; 39:121-127. [DOI: 10.1016/j.hipert.2022.03.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 03/22/2022] [Indexed: 11/23/2022]
|
176
|
Wang A, Liu W, Liu Z. A two-sample robust Bayesian Mendelian Randomization method accounting for linkage disequilibrium and idiosyncratic pleiotropy with applications to the COVID-19 outcomes. Genet Epidemiol 2022; 46:159-169. [PMID: 35192729 PMCID: PMC9648496 DOI: 10.1002/gepi.22445] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 11/03/2021] [Accepted: 01/20/2022] [Indexed: 01/02/2023]
Abstract
Mendelian randomization (MR) is a statistical method exploiting genetic variants as instrumental variables to estimate the causal effect of modifiable risk factors on an outcome of interest. Despite wide uses of various popular two-sample MR methods based on genome-wide association study summary level data, however, those methods could suffer from potential power loss or/and biased inference when the chosen genetic variants are in linkage disequilibrium (LD), and also have relatively large direct effects on the outcome whose distribution might be heavy-tailed which is commonly referred to as the idiosyncratic pleiotropy phenomenon. To resolve those two issues, we propose a novel Robust Bayesian Mendelian Randomization (RBMR) model that uses the more robust multivariate generalized t $t$ -distribution to model such direct effects in a probabilistic model framework which can also incorporate the LD structure explicitly. The generalized t $t$ -distribution can be represented as a Gaussian scaled mixture so that our model parameters can be estimated by the expectation maximization (EM)-type algorithms. We compute the standard errors by calibrating the evidence lower bound using the likelihood ratio test. Through extensive simulation studies, we show that our RBMR has robust performance compared with other competing methods. We further apply our RBMR method to two benchmark data sets and find that RBMR has smaller bias and standard errors. Using our proposed RBMR method, we find that coronary artery disease is associated with increased risk of critically ill coronavirus disease 2019. We also develop a user-friendly R package RBMR (https://github.com/AnqiWang2021/RBMR) for public use.
Collapse
Affiliation(s)
- Anqi Wang
- Department of Statistics and Actuarial ScienceUniversity of Hong KongHong KongSARChina
| | - Wei Liu
- Department of Statistics and Actuarial ScienceUniversity of Hong KongHong KongSARChina
| | - Zhonghua Liu
- Department of Statistics and Actuarial ScienceUniversity of Hong KongHong KongSARChina
| |
Collapse
|
177
|
Coto E, Albaiceta GM, Amado-Rodríguez L, García-Clemente M, Cuesta-Llavona E, Vázquez-Coto D, Alonso B, Iglesias S, Melón S, Alvarez-Argüelles ME, Boga JA, Rojo-Alba S, Pérez-Oliveira S, Alvarez V, Gómez J. FURIN gene variants (rs6224/rs4702) as potential markers of deat hand cardiovascular traits in severe COVID-19. J Med Virol 2022; 94:3589-3595. [PMID: 35355278 PMCID: PMC9088626 DOI: 10.1002/jmv.27748] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/28/2022] [Accepted: 03/28/2022] [Indexed: 11/08/2022]
Abstract
Furin is a protease that plays a key role in the infection cycle of SARS‐CoV‐2 by cleaving the viral proteins during the virus particle assembly. In addition, Furin regulates several physiological processes related to cardio‐metabolic traits. DNA variants in the FURIN gene are candidates to regulate the risk of developing these traits as well as the susceptibility to severe COVID‐19. We genotyped two functional FURIN variants (rs6224/rs4702) in 428 COVID‐19 patients in the intensive care unit. The association with death (N = 106) and hypertension, diabetes, and hyperlipidaemia was statistically evaluated. The risk of death was associated with age, hypertension, and hypercholesterolemia. The two FURIN alleles linked to higher expression (rs6224 T and rs4702 A) were significantly increased in the death cases (odds ratio= 1.40 and 1.43). Homozygosis for the two high expression genotypes (rs6224 TT and rs4702 AA) and for the T‐A haplotype was associated with an increased risk of hypercholesterolemia. In the multiple logistic regression both, hypercholesterolemia and the TT + AA genotype were significantly associated with death. In conclusion, besides its association with hypercholesterolemia, FURIN variants might be independent risk factors for the risk of death among COVID‐19 patients. Furin plays an important role in the life‐cycle of SARS‐CoV‐2 Furin activity might regulate the risk for cardiovascular traits, such as hypertension and hypercholesterolemia. Two functional FURIN variants were associated with the risk of death among critical COVID‐19 patients. The observed association with mortality was independent of higher cholesterol levels. FURIN gene variants might be predictors of COVID‐19 severity and mortaility.
Collapse
Affiliation(s)
- Eliecer Coto
- Genética Molecular, Hospital Universitario Central Asturias, Oviedo, Spain.,Instituto de Investigación Sanitaria del Principado deAsturias, ISPA, Oviedo, Spain.,Universidad de Oviedo, Oviedo, Spain
| | - Guillermo M Albaiceta
- Unidad de Cuidados Intensivos Cardiológicos, Hospital Universitario Central Asturias, Oviedo, Spain.,Instituto de Investigación Sanitaria del Principado deAsturias, ISPA, Oviedo, Spain.,Universidad de Oviedo, Oviedo, Spain.,CIBER-Enfermedades Respiratorias. Instituto de Salud Carlos III, Madrid, Spain.,Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain
| | - Laura Amado-Rodríguez
- Unidad de Cuidados Intensivos Cardiológicos, Hospital Universitario Central Asturias, Oviedo, Spain.,Instituto de Investigación Sanitaria del Principado deAsturias, ISPA, Oviedo, Spain.,Universidad de Oviedo, Oviedo, Spain.,CIBER-Enfermedades Respiratorias. Instituto de Salud Carlos III, Madrid, Spain.,Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain
| | - Marta García-Clemente
- Neumología, Hospital Universitario Central Asturias, Oviedo, Spain.,Instituto de Investigación Sanitaria del Principado deAsturias, ISPA, Oviedo, Spain
| | - Elías Cuesta-Llavona
- Genética Molecular, Hospital Universitario Central Asturias, Oviedo, Spain.,Instituto de Investigación Sanitaria del Principado deAsturias, ISPA, Oviedo, Spain
| | | | - Belén Alonso
- Genética Molecular, Hospital Universitario Central Asturias, Oviedo, Spain
| | - Sara Iglesias
- Genética Molecular, Hospital Universitario Central Asturias, Oviedo, Spain
| | - Santiago Melón
- Microbiologia, Hospital Universitario Central Asturias, Oviedo, Spain.,Instituto de Investigación Sanitaria del Principado deAsturias, ISPA, Oviedo, Spain
| | - Marta E Alvarez-Argüelles
- Microbiologia, Hospital Universitario Central Asturias, Oviedo, Spain.,Instituto de Investigación Sanitaria del Principado deAsturias, ISPA, Oviedo, Spain
| | - José A Boga
- Microbiologia, Hospital Universitario Central Asturias, Oviedo, Spain.,Instituto de Investigación Sanitaria del Principado deAsturias, ISPA, Oviedo, Spain
| | - Susana Rojo-Alba
- Microbiologia, Hospital Universitario Central Asturias, Oviedo, Spain.,Instituto de Investigación Sanitaria del Principado deAsturias, ISPA, Oviedo, Spain
| | - Sergio Pérez-Oliveira
- Genética Molecular, Hospital Universitario Central Asturias, Oviedo, Spain.,Instituto de Investigación Sanitaria del Principado deAsturias, ISPA, Oviedo, Spain
| | - Victoria Alvarez
- Genética Molecular, Hospital Universitario Central Asturias, Oviedo, Spain.,Instituto de Investigación Sanitaria del Principado deAsturias, ISPA, Oviedo, Spain
| | - Juan Gómez
- Genética Molecular, Hospital Universitario Central Asturias, Oviedo, Spain.,Instituto de Investigación Sanitaria del Principado deAsturias, ISPA, Oviedo, Spain
| |
Collapse
|
178
|
He Y, Li Y, Zhang J, Chen L, Li J, Zhang M, Zhang Q, Lu Y, Jiang J, Zhang X, Hu J, Ding Y, Zhang M, Peng H. FURIN Promoter Methylation Predicts the Risk of Incident Diabetes: A Prospective Analysis in the Gusu Cohort. Front Endocrinol (Lausanne) 2022; 13:873012. [PMID: 35399937 PMCID: PMC8990793 DOI: 10.3389/fendo.2022.873012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 02/25/2022] [Indexed: 12/02/2022] Open
Abstract
Background Furin has been associated with diabetes but the underlying mechanisms are unclear. As a mediator linking fixed genome and dynamic environment, DNA methylation of its coding gene FURIN may be involved. Here, we aimed to examine the prospective association between DNA methylation in FURIN promoter and incident diabetes during 4 years of follow-up in Chinese adults. Methods DNA methylation levels in FURIN promoter were quantified by target bisulfite sequencing using peripheral blood from 1836 participants in the Gusu cohort who were free of diabetes at baseline. To examine the association between DNA methylation levels in FURIN promoter and incident diabetes, we constructed a logistic regression model adjusting for the conventional factors. Multiple testing was controlled by adjusting for the total number of CpG sites assayed using the false-discovery rate approach. Results Among the 1836 participants free of diabetes at baseline, 109 (5.94%) participants developed diabetes during the average of 4 years of follow-up. Hypermethylation at two of the eight CpG sites assayed in the FURIN promoter was associated with an increased risk of diabetes, after multivariable adjustment and multiple testing correction. Every 5% increment in methylation levels at CpG1 and CpG2 were associated with a 22% (OR=1.22, 95%CI: 1.05-1.43, P=0.009, q=0.038) and 39% (OR=1.39, 95%CI: 1.08-1.77, P=0.009, q=0.038) higher risk of incident diabetes, respectively. The gene-based association analysis revealed that DNA methylation at multiple CpG loci was jointly associated with incident diabetes (P<0.001). Using the average methylation level of the 8 CpG loci in FURIN promoter revealed a similar association (OR=1.28, 95% CI: 1.02-1.62, P=0.037). Conclusions These results suggested that the hypermethylation levels in FURIN promoter were associated with an increased risk for incident diabetes in Chinese adults.
Collapse
Affiliation(s)
- Yan He
- Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, China
| | - Yinan Li
- Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Jianan Zhang
- Department of Chronic Disease, Taicang Center for Disease Control and Prevention, Suzhou, China
| | - Linan Chen
- Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Jing Li
- Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Min Zhang
- Department of Central Office, Suzhou National New and Hi-Tech Industrial Development Zone Center for Disease Control and Prevention, Suzhou, China
| | - Qiu Zhang
- Department of Chronic Disease, Gusu Center for Disease Control and Prevention, Suzhou, China
| | - Ying Lu
- Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Jun Jiang
- Department of Tuberculosis Control, Suzhou Center for Disease Control and Prevention, Suzhou, China
| | - Xiaolong Zhang
- Department of Tuberculosis Control, Suzhou Center for Disease Control and Prevention, Suzhou, China
| | - Jianwei Hu
- Department of Central Office, Maternal and Child Health Bureau of Kunshan, Suzhou, China
| | - Yi Ding
- Department of Preventive Medicine, College of Clinical Medicine, Suzhou Vocational Health College, Suzhou, China
| | - Mingzhi Zhang
- Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Hao Peng
- Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, China
| |
Collapse
|
179
|
Chillo P, Mashili F, Kwesigabo G, Ruggajo P, Kamuhabwa A. Developing a Sustainable Cardiovascular Disease Research Strategy in Tanzania Through Training: Leveraging From the East African Centre of Excellence in Cardiovascular Sciences Project. Front Cardiovasc Med 2022; 9:849007. [PMID: 35402575 PMCID: PMC8990919 DOI: 10.3389/fcvm.2022.849007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 03/01/2022] [Indexed: 11/17/2022] Open
Abstract
Introduction Cardiovascular disease (CVD) contribute the largest mortality burden globally, with most of the deaths (80% of all deaths) occurring in low and middle-income countries (LMICs), including Tanzania. Despite the increasing burden, to date, CVD research output is still limited in Tanzania, as it is for many sub-Saharan Africa (SSA) countries. This trend hinders the establishment of locally informed CVD management and policy changes. Here, we aim to review the existing gaps while highlighting the available opportunities for a sustainable CVD research strategy in Tanzania. Methods A rapid review of available literature on CVD research in SSA was conducted, with emphasis on the contribution of Tanzania in the world literature of CVD. Through available literature, we identify strategic CVD research priorities in Tanzania and highlight challenges and opportunities for sustainable CVD research output. Findings Shortage of skilled researchers, inadequate research infrastructure, limited funding, and lack of organized research strategies at different levels (regional, country, and institutional) are among the existing key bottlenecks contributing to the low output of CVD research in Tanzania. There is generally strong global, regional and local political will to address the CVD epidemic. The establishment of the East African Centre of Excellence in Cardiovascular Sciences (EACoECVS) offers a unique opportunity for setting strategies and coordinating CVD research and training for Tanzania and the East African region. Conclusion There is a light of hope for long-term sustainable CVD research output from Tanzania, taking advantage of the ongoing activities and plans for the evolving EACoECVS. The Tanzanian experience can be taken as a lesson for other SSA countries.
Collapse
Affiliation(s)
- Pilly Chillo
- Department of Internal Medicine, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
- East African Centre of Excellence in Cardiovascular Sciences, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
- *Correspondence: Pilly Chillo, ;
| | - Fredirick Mashili
- East African Centre of Excellence in Cardiovascular Sciences, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
- Department of Physiology, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Gideon Kwesigabo
- East African Centre of Excellence in Cardiovascular Sciences, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
- Department of Epidemiology and Biostatistics, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Paschal Ruggajo
- Department of Internal Medicine, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Appolinary Kamuhabwa
- East African Centre of Excellence in Cardiovascular Sciences, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
- Department of Clinical Pharmacy and Pharmacology, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| |
Collapse
|
180
|
Liu Y, Wang Y, Qin S, Jin X, Jin L, Gu W, Mu Y. Insights Into Genome-Wide Association Study for Diabetes: A Bibliometric and Visual Analysis From 2001 to 2021. Front Endocrinol (Lausanne) 2022; 13:817620. [PMID: 35360064 PMCID: PMC8963272 DOI: 10.3389/fendo.2022.817620] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 02/07/2022] [Indexed: 12/24/2022] Open
Abstract
Hundreds of research and review articles concerning genome-wide association study (GWAS) in diabetes have been published in the last two decades. We aimed to evaluate the hotspots and future trends in GWAS in diabetes research through bibliometric analysis. Accordingly, 567 research and review articles published between 2001 and 2021 were included. A rising trend was noted in the annual number of publications and citations on GWAS in diabetes during this period. Harvard University and Harvard Medical School have played leading roles in genome research. Hotspot analyses indicated that DNA methylation and genetic variation, especially in type 2 diabetes mellitus, are likely to remain the research hotspots. Moreover, the identification of genetic phenotypes associated with adiposity, metabolic memory, pancreatic islet, and inflammation is the leading trend in this research field. Through this review, we provide predictions on the main research trends in the future so as to shed light on new directions and ideas for further investigations on the genetic etiology of diabetes for its prevention and treatment.
Collapse
Affiliation(s)
- Yang Liu
- Department of Endocrinology, the First Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Department of Endocrinology, the Eighth Medical Center of People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Yun Wang
- Department of Endocrinology, the First Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Shan Qin
- Department of Endocrinology, the First Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Xinye Jin
- Department of Endocrinology, the First Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Department of Nephrology, Hainan Hospital of Chinese People’s Liberation Army (PLA) General Hospital, Academician Chen Xiangmei of Hainan Province Kidney Diseases Research Team Innovation Center, Sanya, China
| | - Lingzi Jin
- Department of International Medical Services, Peking Union Medical College Hospital, Beijing, China
| | - Weijun Gu
- Department of Endocrinology, the First Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Yiming Mu
- Department of Endocrinology, the First Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| |
Collapse
|
181
|
Comprehensive Landscape of Modules-Dysregulated Functions Reveals a Profound Role of ceRNAs in Coronary Heart Disease. JOURNAL OF HEALTHCARE ENGINEERING 2022. [DOI: 10.1155/2022/4547413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Coronary heart disease (CHD) is one of the most common severe cardiovascular diseases. Competitive endogenous RNAs (ceRNA) play critical roles in complex diseases. However, our understanding of the dysregulated functions of ceRNAs in CHD remains limited. Here, we systematically analyzed the alterations of ceRNAs and identified the specific functions based on dysregulated modules from the ceRNA network. A total of 2457 significantly differential expressed genes and 212 differential expressed lncRNAs were identified. We got 76679 regulator relationship between different expression genes and miRNAs and 336 regulator relationship between differential expressed lncRNAs and miRNAs. We constructed the ceRNA network and selected five dysregulated modules. Furthermore, CHD specific functions based on dysregulated modules from the ceRNA network were identified, including histone acetylation, platelet degranulation, cAMP-dependent protein kinase complex, xenobiotic transport and so on. Our results will provide novel insight for a better understanding of the mechanism of ceRNAs and facilitate the identification of novel diagnostic and therapeutic biomarkers in CHD.
Collapse
|
182
|
Cataloging the potential SNPs (single nucleotide polymorphisms) associated with quantitative traits, viz. BMI (body mass index), IQ (intelligence quotient) and BP (blood pressure): an updated review. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2022. [DOI: 10.1186/s43042-022-00266-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Single nucleotide polymorphism (SNP) variants are abundant, persistent and widely distributed across the genome and are frequently linked to the development of genetic diseases. Identifying SNPs that underpin complex diseases can aid scientists in the discovery of disease-related genes by allowing for early detection, effective medication and eventually disease prevention.
Main body
Various SNP or polymorphism-based studies were used to categorize different SNPs potentially related to three quantitative traits: body mass index (BMI), intelligence quotient (IQ) and blood pressure, and then uncovered common SNPs for these three traits. We employed SNPedia, RefSNP Report, GWAS Catalog, Gene Cards (Data Bases), PubMed and Google Scholar search engines to find relevant material on SNPs associated with three quantitative traits. As a result, we detected three common SNPs for all three quantitative traits in global populations: SNP rs6265 of the BDNF gene on chromosome 11p14.1, SNP rs131070325 of the SL39A8 gene on chromosome 4p24 and SNP rs4680 of the COMT gene on chromosome 22q11.21.
Conclusion
In our review, we focused on the prevalent SNPs and gene expression activities that influence these three quantitative traits. These SNPs have been used to detect and map complex, common illnesses in communities for homogeneity testing and pharmacogenetic studies. High blood pressure, diabetes and heart disease, as well as BMI, schizophrenia and IQ, can all be predicted using common SNPs. Finally, the results of our work can be used to find common SNPs and genes that regulate these three quantitative features across the genome.
Collapse
|
183
|
Zhu X, Zhu L, Wang H, Cooper RS, Chakravarti A. Genome-wide pleiotropy analysis identifies novel blood pressure variants and improves its polygenic risk scores. Genet Epidemiol 2022; 46:105-121. [PMID: 34989438 PMCID: PMC8863647 DOI: 10.1002/gepi.22440] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 12/07/2021] [Indexed: 01/21/2023]
Abstract
Systolic and diastolic blood pressure (S/DBP) are highly correlated modifiable risk factors for cardiovascular disease (CVD). We report here a bidirectional Mendelian Randomization (MR) and horizontal pleiotropy analysis of S/DBP summary statistics from the UK Biobank (UKB)-International Consortium for Blood Pressure (ICBP) (UKB-ICBP) BP genome-wide association study and construct a composite genetic risk score (GRS) by including pleiotropic variants. The composite GRS captures greater (1.11-3.26 fold) heritability for BP traits and increases (1.09- and 2.01-fold) Nagelkerke's R2 for hypertension and CVD. We replicated 118 novel BP horizontal pleiotropic variants including 18 novel BP loci using summary statistics from the Million Veteran Program (MVP) study. An additional 219 novel BP signals and 40 novel loci were identified after a meta-analysis of the UKB-ICBP and MVP summary statistics but without further independent replication. Our study provides further insight into BP regulation and provides a novel way to construct a GRS by including pleiotropic variants for other complex diseases.
Collapse
Affiliation(s)
- Xiaofeng Zhu
- Department of Population and Quantitative Health SciencesCase Western Reserve UniversityClevelandOhioUSA
| | - Luke Zhu
- Department of Medicine, Center for Human Genetics & GenomicsNew York University Langone HealthNew YorkNew YorkUSA
| | - Heming Wang
- Division of Sleep and Circadian DisordersBrigham and Women's HospitalBostonMassachusettsUSA
| | - Richard S. Cooper
- Department of Public Health Sciences, Stritch School of MedicineLoyola University ChicagoMaywoodIllinoisUSA
| | - Aravinda Chakravarti
- Department of Medicine, Center for Human Genetics & GenomicsNew York University Langone HealthNew YorkNew YorkUSA
| |
Collapse
|
184
|
Chen K, Zhuang Z, Shao C, Zheng J, Zhou Q, Dong E, Huang T, Tang YD. Roles of Cardiometabolic Factors in Mediating the Causal Effect of Type 2 Diabetes on Cardiovascular Diseases: A Two-Step, Two-Sample Multivariable Mendelian Randomization Study. Front Cardiovasc Med 2022; 9:813208. [PMID: 35282373 PMCID: PMC8909643 DOI: 10.3389/fcvm.2022.813208] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 01/17/2022] [Indexed: 12/17/2022] Open
Abstract
ObjectiveThe objective of this study is to investigate the roles of cardiometabolic factors (including blood pressure, blood lipids, thyroid function, body mass, and insulin sensitivity) in mediating the causal effect of type 2 diabetes (T2DM) on cardiovascular disease (CVD) outcomes.DesignTwo-step, two-sample multivariable Mendelian randomization (MVMR) study.SettingInternational genome-wide association study (GWAS) consortia data.ExposureType 2 diabetes, blood pressure: systolic blood pressure (SBP), diastolic blood pressure (DBP); blood lipids: low-density lipoprotein (LDL), high-density lipoprotein (HDL), total cholesterol (TC), triglycerides (TG); thyroid function: hyperthyroidism, hypothyroidism; body mass index (BMI), waist-hip-ratio (WHR), and insulin sensitivity.Main OutcomesCardiovascular disease includes coronary heart disease (CHD), myocardial infarction (MI), and stroke.MethodsSummary-level data for exposures and main outcomes were extracted from GWAS consortia. We used two-sample MR to illustrate the causal effect of T2DM on CVD subtypes and regression-based MVMR to quantify the possible mediation effects of cardiometabolic factors on CVD.ResultsEach additional unit of log odds of T2DM increased 16% risk of CHD [odds ratio (OR): 1.16, 95% CI: 1.12–1.21], 15% risk of myocardial infarction (MI) (OR: 1.15, 95% CI: 1.10–1.20), and 10% risk of stroke (OR: 1.10, 95% CI: 1.06–1.13). In mediation analysis, SBP, DBP, and TG were found as main mediators, while the mediation effects of other cardiometabolic factors were not significant. The proportion of total effect of T2DM on CHD mediated by SBP, DBP, and TG was 16% (95% CI: 8–24%), 7% (95% CI: 1–13%) and 10% (95% CI: 2–18%), respectively. Mediation effect of SBP and DBP on MI and stroke, TG on MI was also prominent, while mediation effect of TG on stroke was not significant. The combined mediation effect of all the three mediators accounted for 29%, 26%, and 13% of the total effect of T2DM on CHD, MI, and stroke, respectively.ConclusionSystolic blood pressure, DBP, and TG mediate a substantial proportion of the causal effect of T2DM on CVD and thus interventions on these factors might reduce the considerable excess risk of CVD among patients with T2DM.
Collapse
Affiliation(s)
- Ken Chen
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Cardiology, Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhenhuang Zhuang
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Chunli Shao
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Cardiology, Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Jilin Zheng
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Cardiology, Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qing Zhou
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Cardiology, Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Erdan Dong
- Department of Cardiology and Institute of Vascular Medicine, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University Third Hospital, Beijing, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, The Institute of Cardiovascular Sciences, Peking University, Beijing, China
| | - Tao Huang
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing, China
- Department of Global Health, School of Public Health, Peking University, Beijing, China
- Key Laboratory of Molecular Cardiovascular Sciences (Peking University), Ministry of Education, Beijing, China
- Center for Intelligent Public Health, Institute for Artificial Intelligence, Peking University, Beijing, China
| | - Yi-Da Tang
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Cardiology, Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| |
Collapse
|
185
|
Singh V, Akash R, Chaudhary G, Singh R, Choudhury S, Shukla A, Prabhu SN, Gangwar N, Garg SK. Sepsis downregulates aortic Notch signaling to produce vascular hyporeactivity in mice. Sci Rep 2022; 12:2941. [PMID: 35190630 PMCID: PMC8861011 DOI: 10.1038/s41598-022-06949-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 02/02/2022] [Indexed: 11/25/2022] Open
Abstract
Inhibition of Notch signaling in macrophages is known to reduce inflammation, however, its role in regulating vascular hyporeactivity in sepsis is unknown. Thus we aimed to evaluate the effect of sepsis on vascular Notch signaling. Polymicrobial sepsis was induced by caecal ligation and puncture (CLP) in mice. mRNA expressions of Notch receptors (Notch1,3) and ligands (Jag1, Dll4), and downstream effector genes (Hey1, MLCK, MYPT1) were assessed by RT-qPCR. Protein level of activated Notch (NICD) was assessed by Western blot and immuno-histochemistry. Isometric tension in isolated aortic rings was measured by wire myography.CLP down-regulated aortic expression of Notch3, Jag1 and Dll4 as compared to control mice. Additionally, the protein level of NICD was found to be lesser in aortic tissue sections from CLP mice. Expression of Hey1 and MLCK were attenuated whereas MYPT1 expression was increased in septic mouse aorta. DAPT pretreatment did not improve CLP-induced vascular hyporeactivity to NA, CaCl2 and high K+ (80 mM), rather significantly attenuated the aortic response to these vasoconstrictors in control mice. Treatment with 1400 W reversed attenuated Notch3 (but not Jag1 and MLCK) expression in septic mouse aorta. In conclusion, sepsis significantly attenuated the Notch (especially Notch3) signaling in mouse aorta along with reduction in contractile gene expression and vasoconstriction response. Further, iNOS/NO pathway was involved in sepsis-induced down-regulation of Notch3 receptor. Thus systemic inhibition of Notch signaling during sepsis may have serious impact on sepsis-induced vascular hyporeactivity.
Collapse
Affiliation(s)
- Vandana Singh
- Smooth Muscle Pharmacology and Molecular Pharmacology Laboratory, Department of Veterinary Pharmacology and Toxicology, College of Veterinary Science and Animal Husbandry, U.P. Pandit Deen Dayal Upadhyaya Pashu Chikitsa Vigyan Vishwavidyalaya Evam Go-Anusandhan Sansthan (DUVASU), Mathura, 281001, India
| | - Raut Akash
- Smooth Muscle Pharmacology and Molecular Pharmacology Laboratory, Department of Veterinary Pharmacology and Toxicology, College of Veterinary Science and Animal Husbandry, U.P. Pandit Deen Dayal Upadhyaya Pashu Chikitsa Vigyan Vishwavidyalaya Evam Go-Anusandhan Sansthan (DUVASU), Mathura, 281001, India
| | - Gaurav Chaudhary
- Smooth Muscle Pharmacology and Molecular Pharmacology Laboratory, Department of Veterinary Pharmacology and Toxicology, College of Veterinary Science and Animal Husbandry, U.P. Pandit Deen Dayal Upadhyaya Pashu Chikitsa Vigyan Vishwavidyalaya Evam Go-Anusandhan Sansthan (DUVASU), Mathura, 281001, India
| | - Rajneesh Singh
- Smooth Muscle Pharmacology and Molecular Pharmacology Laboratory, Department of Veterinary Pharmacology and Toxicology, College of Veterinary Science and Animal Husbandry, U.P. Pandit Deen Dayal Upadhyaya Pashu Chikitsa Vigyan Vishwavidyalaya Evam Go-Anusandhan Sansthan (DUVASU), Mathura, 281001, India
| | - Soumen Choudhury
- Smooth Muscle Pharmacology and Molecular Pharmacology Laboratory, Department of Veterinary Pharmacology and Toxicology, College of Veterinary Science and Animal Husbandry, U.P. Pandit Deen Dayal Upadhyaya Pashu Chikitsa Vigyan Vishwavidyalaya Evam Go-Anusandhan Sansthan (DUVASU), Mathura, 281001, India.
| | - Amit Shukla
- Smooth Muscle Pharmacology and Molecular Pharmacology Laboratory, Department of Veterinary Pharmacology and Toxicology, College of Veterinary Science and Animal Husbandry, U.P. Pandit Deen Dayal Upadhyaya Pashu Chikitsa Vigyan Vishwavidyalaya Evam Go-Anusandhan Sansthan (DUVASU), Mathura, 281001, India
| | - Shyama N Prabhu
- Department of Veterinary Pathology, College of Veterinary Science and Animal Husbandry, U.P. Pandit Deen Dayal Upadhyaya Pashu Chikitsa Vigyan Vishwavidyalaya Evam Go-Anusandhan Sansthan, Mathura, 281001, India
| | - Neeraj Gangwar
- Department of Veterinary Pathology, College of Veterinary Science and Animal Husbandry, U.P. Pandit Deen Dayal Upadhyaya Pashu Chikitsa Vigyan Vishwavidyalaya Evam Go-Anusandhan Sansthan, Mathura, 281001, India
| | - Satish K Garg
- Smooth Muscle Pharmacology and Molecular Pharmacology Laboratory, Department of Veterinary Pharmacology and Toxicology, College of Veterinary Science and Animal Husbandry, U.P. Pandit Deen Dayal Upadhyaya Pashu Chikitsa Vigyan Vishwavidyalaya Evam Go-Anusandhan Sansthan (DUVASU), Mathura, 281001, India
| |
Collapse
|
186
|
Yu M, Tcheandjieu C, Georges A, Xiao K, Tejeda H, Dina C, Le Tourneau T, Fiterau M, Judy R, Tsao NL, Amgalan D, Munger CJ, Engreitz JM, Damrauer SM, Bouatia-Naji N, Priest JR. Computational estimates of annular diameter reveal genetic determinants of mitral valve function and disease. JCI Insight 2022; 7:146580. [PMID: 35132965 PMCID: PMC8855800 DOI: 10.1172/jci.insight.146580] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 12/21/2021] [Indexed: 11/17/2022] Open
Abstract
The fibrous annulus of the mitral valve plays an important role in valvular function and cardiac physiology, while normal variation in the size of cardiovascular anatomy may share a genetic link with common and rare disease. We derived automated estimates of mitral valve annular diameter in the 4-chamber view from 32,220 MRI images from the UK Biobank at ventricular systole and diastole as the basis for GWAS. Mitral annular dimensions corresponded to previously described anatomical norms, and GWAS inclusive of 4 population strata identified 10 loci, including possibly novel loci (GOSR2, ERBB4, MCTP2, MCPH1) and genes related to cardiac contractility (BAG3, TTN, RBFOX1). ATAC-Seq of primary mitral valve tissue localized multiple variants to regions of open chromatin in biologically relevant cell types and rs17608766 to an algorithmically predicted enhancer element in GOSR2. We observed strong genetic correlation with measures of contractility and mitral valve disease and clinical correlations with heart failure, cerebrovascular disease, and ventricular arrhythmias. Polygenic scoring of mitral valve annular diameter in systole was predictive of risk mitral valve prolapse across 4 cohorts. In summary, genetic and clinical studies of mitral valve annular diameter revealed genetic determinants of mitral valve biology, while highlighting clinical associations. Polygenic determinants of mitral valve annular diameter may represent an independent risk factor for mitral prolapse. Overall, computationally estimated phenotypes derived at scale from medical imaging represent an important substrate for genetic discovery and clinical risk prediction.
Collapse
Affiliation(s)
| | - Catherine Tcheandjieu
- Department of Pediatrics and.,Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Adrien Georges
- Paris Cardiovascular Research Center, INSERM, University of Paris, Paris, France
| | - Ke Xiao
- College of Information & Computer Sciences at University of Massachusetts Amherst, Amherst, Massachusetts, USA
| | | | - Christian Dina
- University of Nantes, INSERM, CNRS, CHU Nantes, The Thorax Institute, Nantes, France
| | - Thierry Le Tourneau
- University of Nantes, INSERM, CNRS, CHU Nantes, The Thorax Institute, Nantes, France
| | - Madalina Fiterau
- College of Information & Computer Sciences at University of Massachusetts Amherst, Amherst, Massachusetts, USA
| | - Renae Judy
- Department of Surgery, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Noah L Tsao
- Department of Surgery, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Dulguun Amgalan
- Department of Genetics, Stanford University School of Medicine, Stanford, California, USA.,Basic Science & Engineering Initiative & Betty Irene Moore Children's Heart Center, Lucile Packard Children's Hospital, Stanford, California, USA
| | - Chad J Munger
- Department of Genetics, Stanford University School of Medicine, Stanford, California, USA.,Basic Science & Engineering Initiative & Betty Irene Moore Children's Heart Center, Lucile Packard Children's Hospital, Stanford, California, USA
| | - Jesse M Engreitz
- Department of Genetics, Stanford University School of Medicine, Stanford, California, USA.,Basic Science & Engineering Initiative & Betty Irene Moore Children's Heart Center, Lucile Packard Children's Hospital, Stanford, California, USA
| | - Scott M Damrauer
- Department of Surgery, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Nabila Bouatia-Naji
- Paris Cardiovascular Research Center, INSERM, University of Paris, Paris, France
| | - James R Priest
- Department of Pediatrics and.,Chan-Zuckerberg Biohub, San Francisco, California, USA
| |
Collapse
|
187
|
Hettwer MD, Lancaster TM, Raspor E, Hahn PK, Mota NR, Singer W, Reif A, Linden DEJ, Bittner RA. Evidence From Imaging Resilience Genetics for a Protective Mechanism Against Schizophrenia in the Ventral Visual Pathway. Schizophr Bull 2022; 48:551-562. [PMID: 35137221 PMCID: PMC9077432 DOI: 10.1093/schbul/sbab151] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
INTRODUCTION Illuminating neurobiological mechanisms underlying the protective effect of recently discovered common genetic resilience variants for schizophrenia is crucial for more effective prevention efforts. Current models implicate adaptive neuroplastic changes in the visual system and their pro-cognitive effects as a schizophrenia resilience mechanism. We investigated whether common genetic resilience variants might affect brain structure in similar neural circuits. METHOD Using structural magnetic resonance imaging, we measured the impact of an established schizophrenia polygenic resilience score (PRSResilience) on cortical volume, thickness, and surface area in 101 healthy subjects and in a replication sample of 33 224 healthy subjects (UK Biobank). FINDING We observed a significant positive whole-brain correlation between PRSResilience and cortical volume in the right fusiform gyrus (FFG) (r = 0.35; P = .0004). Post-hoc analyses in this cluster revealed an impact of PRSResilience on cortical surface area. The replication sample showed a positive correlation between PRSResilience and global cortical volume and surface area in the left FFG. CONCLUSION Our findings represent the first evidence of a neurobiological correlate of a genetic resilience factor for schizophrenia. They support the view that schizophrenia resilience emerges from strengthening neural circuits in the ventral visual pathway and an increased capacity for the disambiguation of social and nonsocial visual information. This may aid psychosocial functioning, ameliorate the detrimental effects of subtle perceptual and cognitive disturbances in at-risk individuals, and facilitate coping with the cognitive and psychosocial consequences of stressors. Our results thus provide a novel link between visual cognition, the vulnerability-stress concept, and schizophrenia resilience models.
Collapse
Affiliation(s)
- Meike D Hettwer
- Department of Psychiatry, Psychosomatic Medicine, and Psychotherapy, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany,Max Planck School of Cognition, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany,Institute of Neuroscience and Medicine, Brain & Behaviour (INM-7), Research Centre Jülich, Jülich, Germany,Institute of Systems Neuroscience, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Thomas M Lancaster
- School of Psychology, Bath University, Bath, UK,MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neuroscience, School of Medicine, Cardiff University, Cardiff, UK
| | - Eva Raspor
- Department of Psychiatry, Psychosomatic Medicine, and Psychotherapy, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Peter K Hahn
- Department of Psychiatry, Psychosomatic Medicine, and Psychotherapy, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Nina Roth Mota
- Department of Human Genetics, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands,Department of Psychiatry, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - Wolf Singer
- Ernst Strüngmann Institute for Neuroscience (ESI) in Cooperation with Max Planck Society, Frankfurt am Main, Germany,Max Planck Institute for Brain Research (MPI BR), Frankfurt am Main, Germany,Frankfurt Institute for Advanced Studies (FIAS), Frankfurt am Main, Germany
| | - Andreas Reif
- Department of Psychiatry, Psychosomatic Medicine, and Psychotherapy, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - David E J Linden
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neuroscience, School of Medicine, Cardiff University, Cardiff, UK,School for Mental Health and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
| | - Robert A Bittner
- To whom correspondence should be addressed; Heinrich-Hoffmann-Str. 10, D-60528 Frankfurt am Main, Germany; tel: 69-6301-84713, fax: 69-6301-81775, e-mail:
| |
Collapse
|
188
|
Canoy D, Harvey NC, Prieto-Alhambra D, Cooper C, Meyer HE, Åsvold BO, Nazarzadeh M, Rahimi K. Elevated blood pressure, antihypertensive medications and bone health in the population: revisiting old hypotheses and exploring future research directions. Osteoporos Int 2022; 33:315-326. [PMID: 34642814 PMCID: PMC8813726 DOI: 10.1007/s00198-021-06190-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 10/01/2021] [Indexed: 11/17/2022]
Abstract
Blood pressure and bone metabolism appear to share commonalities in their physiologic regulation. Specific antihypertensive drug classes may also influence bone mineral density. However, current evidence from existing observational studies and randomised trials is insufficient to establish causal associations for blood pressure and use of blood pressure-lowering drugs with bone health outcomes, particularly with the risks of osteoporosis and fractures. The availability and access to relevant large-scale biomedical data sources as well as developments in study designs and analytical approaches provide opportunities to examine the nature of the association between blood pressure and bone health more reliably and in greater detail than has ever been possible. It is unlikely that a single source of data or study design can provide a definitive answer. However, with appropriate considerations of the strengths and limitations of the different data sources and analytical techniques, we should be able to advance our understanding of the role of raised blood pressure and its drug treatment on the risks of low bone mineral density and fractures. As elevated blood pressure is highly prevalent and blood pressure-lowering drugs are widely prescribed, even small effects of these exposures on bone health outcomes could be important at a population level.
Collapse
Affiliation(s)
- D Canoy
- Deep Medicine, Nuffield Department of Women's and Reproductive Health, University of Oxford, Hayes House 1F, George St., Oxford, OX1 2BQ, UK.
- NIHR Oxford Biomedical Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK.
| | - N C Harvey
- MRC Life Course Epidemiology Unit, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - D Prieto-Alhambra
- Centre for Statistics in Medicine, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
- Department of Medical Informatics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - C Cooper
- NIHR Oxford Biomedical Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
- MRC Life Course Epidemiology Unit, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - H E Meyer
- Department of Community Medicine and Global Health, Faculty of Medicine, Oslo, Norway
- Norwegian Institute of Public Health, Oslo, Norway
| | - B O Åsvold
- Department of Endocrinology, Clinic of Medicine, St. Olav's Hospital, Trondheim University Hospital, Trondheim, Norway
- Department of Public Health and Nursing, K.G. Jebsen Center for Genetic Epidemiology, Norwegian University of Science and Technology, Trondheim, Norway
| | - M Nazarzadeh
- Deep Medicine, Nuffield Department of Women's and Reproductive Health, University of Oxford, Hayes House 1F, George St., Oxford, OX1 2BQ, UK
| | - K Rahimi
- Deep Medicine, Nuffield Department of Women's and Reproductive Health, University of Oxford, Hayes House 1F, George St., Oxford, OX1 2BQ, UK
- NIHR Oxford Biomedical Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| |
Collapse
|
189
|
Daghals I, Sargurupremraj M, Danning R, Gormley P, Malik R, Amouyel P, Metso T, Pezzini A, Kurth T, Debette S, Chasman D. Migraine, Stroke, and Cervical Arterial Dissection: Shared Genetics for a Triad of Brain Disorders With Vascular Involvement. Neurol Genet 2022; 8:e653. [PMID: 35128049 PMCID: PMC8808356 DOI: 10.1212/nxg.0000000000000653] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 12/07/2021] [Indexed: 12/15/2022]
Abstract
Background and Objectives Migraine, stroke, and cervical artery dissection (CeAD) represent a triad of cerebrovascular disorders with pairwise comorbid relationships and vascular involvement. Larger samples and recent advances in methodology invite systematic exploration of their shared genetics. Methods Genetic analyses leveraged summary statistics from genome-wide association studies of the largest available samples of each disorder, including subtypes of stroke (ischemic stroke, large artery stroke, small vessel stroke, and cardioembolic stroke) and migraine (with aura and without aura). For each pair of disorders, genetic correlation was assessed both on a genome-wide basis and within independent segments across the genome including known specific loci for each disorder. A cross-trait meta-analysis was used to identify novel candidate loci. Finally, potential causality of migraine susceptibility on stroke and CeAD was assessed by Mendelian randomization. Results Among all pairs of disorders, genome-wide genetic correlation was observed only between CeAD and migraine, particularly MO. Local genetic correlations were more extensive between migraine and CeAD than those between migraine and stroke or CeAD and stroke and revealed evidence for novel CeAD associations at rs6693567 (ADAMTSL4/ECM1), rs11187838 (PLCE1), and rs7940646 (MRVI1) while strengthening prior subthreshold evidence at rs9486725 (FHL5) and rs650724 (LRP1). At known migraine loci, novel associations with stroke had concordant risk alleles for small vessel stroke at rs191602009 (CARF) and for cardioembolic stroke at rs55884259 (NKX2-5). Known migraine loci also revealed novel associations but with opposite risk alleles for all stroke, ischemic stroke, and small vessel stroke at rs55928386 (HTRA1), for large artery stroke at rs11172113 (LRP1), and for all stroke and ischemic stroke at rs1535791 and rs4942561 (both LRCH1), respectively. rs182923402 (near PTCH1) was a novel concordant locus for migraine and cardioembolic stroke. Mendelian randomization supported potential causal influences of migraine on CeAD (odds ratio [95% confidence interval] per doubling migraine prevalence = 1.69 [1.24-2.3], p = 0.0009) with concordant risk, but with opposite risk on large artery stroke (0.86 [0.76-0.96], p = 0.0067). Discussion The findings emphasize shared genetic risk between migraine and CeAD while identifying loci with likely vascular function in migraine and shared but opposite genetic risk between migraine and stroke subtypes, and a central role of LRP1 in all 3 cerebrovascular disorders.
Collapse
Affiliation(s)
| | | | - Rebecca Danning
- From the Harvard Medical School (I.D., D.C.), Boston, MA; Division of Preventive Medicine (I.D., R.D., D.C.), Brigham and Women's Hospital, Boston, MA; University of Bordeaux (M.S., S.D.), Inserm, Bordeaux Population Health Research Center, Team VINTAGE, UMR 1219, France; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (M.S.), University of Texas Health, San Antonio; Massachusetts General Hospital (P.G.), Boston; Institute for Stroke and Dementia Research (R.M.), Klinikum der Universität München, Ludwig-Maximilians-University, Germany; LabEx DISTALZ-U1167 (P.A.), RID-AGE-Risk Factors and Molecular Determinants of Aging-Related Diseases, University of Lille; Inserm U1167 (P.A.), Lille; Centre Hospitalier Universitaire Lille (P.A.); Institut Pasteur de Lille (P.A.), France; Department of Neurology (T.M.), Helsinki University Central Hospital, Finland; Department of Clinical and Experimental Sciences (A.P.), Neurology Clinic, Brescia University Hospital, Italy; Institute of Public Health (T.K.), Charité—Universitätsmedizin Berlin, Germany; and Department of Neurology (S.D.), CHU de Bordeaux, France
| | - Padhraig Gormley
- From the Harvard Medical School (I.D., D.C.), Boston, MA; Division of Preventive Medicine (I.D., R.D., D.C.), Brigham and Women's Hospital, Boston, MA; University of Bordeaux (M.S., S.D.), Inserm, Bordeaux Population Health Research Center, Team VINTAGE, UMR 1219, France; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (M.S.), University of Texas Health, San Antonio; Massachusetts General Hospital (P.G.), Boston; Institute for Stroke and Dementia Research (R.M.), Klinikum der Universität München, Ludwig-Maximilians-University, Germany; LabEx DISTALZ-U1167 (P.A.), RID-AGE-Risk Factors and Molecular Determinants of Aging-Related Diseases, University of Lille; Inserm U1167 (P.A.), Lille; Centre Hospitalier Universitaire Lille (P.A.); Institut Pasteur de Lille (P.A.), France; Department of Neurology (T.M.), Helsinki University Central Hospital, Finland; Department of Clinical and Experimental Sciences (A.P.), Neurology Clinic, Brescia University Hospital, Italy; Institute of Public Health (T.K.), Charité—Universitätsmedizin Berlin, Germany; and Department of Neurology (S.D.), CHU de Bordeaux, France
| | - Rainer Malik
- From the Harvard Medical School (I.D., D.C.), Boston, MA; Division of Preventive Medicine (I.D., R.D., D.C.), Brigham and Women's Hospital, Boston, MA; University of Bordeaux (M.S., S.D.), Inserm, Bordeaux Population Health Research Center, Team VINTAGE, UMR 1219, France; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (M.S.), University of Texas Health, San Antonio; Massachusetts General Hospital (P.G.), Boston; Institute for Stroke and Dementia Research (R.M.), Klinikum der Universität München, Ludwig-Maximilians-University, Germany; LabEx DISTALZ-U1167 (P.A.), RID-AGE-Risk Factors and Molecular Determinants of Aging-Related Diseases, University of Lille; Inserm U1167 (P.A.), Lille; Centre Hospitalier Universitaire Lille (P.A.); Institut Pasteur de Lille (P.A.), France; Department of Neurology (T.M.), Helsinki University Central Hospital, Finland; Department of Clinical and Experimental Sciences (A.P.), Neurology Clinic, Brescia University Hospital, Italy; Institute of Public Health (T.K.), Charité—Universitätsmedizin Berlin, Germany; and Department of Neurology (S.D.), CHU de Bordeaux, France
| | - Philippe Amouyel
- From the Harvard Medical School (I.D., D.C.), Boston, MA; Division of Preventive Medicine (I.D., R.D., D.C.), Brigham and Women's Hospital, Boston, MA; University of Bordeaux (M.S., S.D.), Inserm, Bordeaux Population Health Research Center, Team VINTAGE, UMR 1219, France; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (M.S.), University of Texas Health, San Antonio; Massachusetts General Hospital (P.G.), Boston; Institute for Stroke and Dementia Research (R.M.), Klinikum der Universität München, Ludwig-Maximilians-University, Germany; LabEx DISTALZ-U1167 (P.A.), RID-AGE-Risk Factors and Molecular Determinants of Aging-Related Diseases, University of Lille; Inserm U1167 (P.A.), Lille; Centre Hospitalier Universitaire Lille (P.A.); Institut Pasteur de Lille (P.A.), France; Department of Neurology (T.M.), Helsinki University Central Hospital, Finland; Department of Clinical and Experimental Sciences (A.P.), Neurology Clinic, Brescia University Hospital, Italy; Institute of Public Health (T.K.), Charité—Universitätsmedizin Berlin, Germany; and Department of Neurology (S.D.), CHU de Bordeaux, France
| | - Tiina Metso
- From the Harvard Medical School (I.D., D.C.), Boston, MA; Division of Preventive Medicine (I.D., R.D., D.C.), Brigham and Women's Hospital, Boston, MA; University of Bordeaux (M.S., S.D.), Inserm, Bordeaux Population Health Research Center, Team VINTAGE, UMR 1219, France; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (M.S.), University of Texas Health, San Antonio; Massachusetts General Hospital (P.G.), Boston; Institute for Stroke and Dementia Research (R.M.), Klinikum der Universität München, Ludwig-Maximilians-University, Germany; LabEx DISTALZ-U1167 (P.A.), RID-AGE-Risk Factors and Molecular Determinants of Aging-Related Diseases, University of Lille; Inserm U1167 (P.A.), Lille; Centre Hospitalier Universitaire Lille (P.A.); Institut Pasteur de Lille (P.A.), France; Department of Neurology (T.M.), Helsinki University Central Hospital, Finland; Department of Clinical and Experimental Sciences (A.P.), Neurology Clinic, Brescia University Hospital, Italy; Institute of Public Health (T.K.), Charité—Universitätsmedizin Berlin, Germany; and Department of Neurology (S.D.), CHU de Bordeaux, France
| | - Alessandro Pezzini
- From the Harvard Medical School (I.D., D.C.), Boston, MA; Division of Preventive Medicine (I.D., R.D., D.C.), Brigham and Women's Hospital, Boston, MA; University of Bordeaux (M.S., S.D.), Inserm, Bordeaux Population Health Research Center, Team VINTAGE, UMR 1219, France; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (M.S.), University of Texas Health, San Antonio; Massachusetts General Hospital (P.G.), Boston; Institute for Stroke and Dementia Research (R.M.), Klinikum der Universität München, Ludwig-Maximilians-University, Germany; LabEx DISTALZ-U1167 (P.A.), RID-AGE-Risk Factors and Molecular Determinants of Aging-Related Diseases, University of Lille; Inserm U1167 (P.A.), Lille; Centre Hospitalier Universitaire Lille (P.A.); Institut Pasteur de Lille (P.A.), France; Department of Neurology (T.M.), Helsinki University Central Hospital, Finland; Department of Clinical and Experimental Sciences (A.P.), Neurology Clinic, Brescia University Hospital, Italy; Institute of Public Health (T.K.), Charité—Universitätsmedizin Berlin, Germany; and Department of Neurology (S.D.), CHU de Bordeaux, France
| | - Tobias Kurth
- From the Harvard Medical School (I.D., D.C.), Boston, MA; Division of Preventive Medicine (I.D., R.D., D.C.), Brigham and Women's Hospital, Boston, MA; University of Bordeaux (M.S., S.D.), Inserm, Bordeaux Population Health Research Center, Team VINTAGE, UMR 1219, France; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (M.S.), University of Texas Health, San Antonio; Massachusetts General Hospital (P.G.), Boston; Institute for Stroke and Dementia Research (R.M.), Klinikum der Universität München, Ludwig-Maximilians-University, Germany; LabEx DISTALZ-U1167 (P.A.), RID-AGE-Risk Factors and Molecular Determinants of Aging-Related Diseases, University of Lille; Inserm U1167 (P.A.), Lille; Centre Hospitalier Universitaire Lille (P.A.); Institut Pasteur de Lille (P.A.), France; Department of Neurology (T.M.), Helsinki University Central Hospital, Finland; Department of Clinical and Experimental Sciences (A.P.), Neurology Clinic, Brescia University Hospital, Italy; Institute of Public Health (T.K.), Charité—Universitätsmedizin Berlin, Germany; and Department of Neurology (S.D.), CHU de Bordeaux, France
| | | | | |
Collapse
|
190
|
Zhang H, Wang A, Xu T, Mo X, Zhang Y. Promoter DNA Methylation in GWAS-Identified Genes as Potential Functional Elements for Blood Pressure: An Observational and Mendelian Randomization Study. Front Genet 2022; 12:791146. [PMID: 35087571 PMCID: PMC8787193 DOI: 10.3389/fgene.2021.791146] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 12/21/2021] [Indexed: 01/03/2023] Open
Abstract
Genome-wide association studies have identified numerous genetic loci for blood pressure (BP). However, the relationships of functional elements inside these loci with BP are not fully understood. This study represented an effort to determine if promoter DNA methylations inside BP-associated loci were associated with BP.We conducted a cross-sectional study investigating the association between promoter DNA methylations of 10 candidate genes and BP in 1,241 Chinese individuals. Twenty-one genomic fragments in the CpG Islands were sequenced. The associations of methylation levels with BP and hypertension were assessed in regression models. Mendelian randomization (MR) analysis was then applied to find supporting evidence for the identified associations.A total of 413 DNA methylation sites were examined in an observational study. Methylation levels of 24 sites in PRDM6, IGFBP3, SYT7, PDE3A, TBX2 and C17orf82 were significantly associated with BP. Methylation levels of PRDM6 and SYT7 were significantly associated with hypertension. Methylation levels of five sites (including cg06713098) in IGFBP3 were significantly associated with DBP. MR analysis found associations between the methylation levels of six CpG sites (cg06713098, cg14228300, cg23193639, cg21268650, cg10677697 and cg04812164) around the IGFBP3 promoter and DBP. Methylation levels of cg14228300 and cg04812164 were associated with SBP. By further applying several MR methods we showed that the associations may not be due to pleiotropy. Association between IGFBP3 mRNA levels in blood cells and BP was also found in MR analysis. This study identified promoter methylation as potential functional element for BP. The identified methylations may be involved in the regulatory pathway linking genetic variants to BP.
Collapse
Affiliation(s)
- Huan Zhang
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China.,Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Aili Wang
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China.,Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Tan Xu
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China.,Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Xingbo Mo
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China.,Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, China.,Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Yonghong Zhang
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China.,Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, China
| |
Collapse
|
191
|
P.O.F.T. Guimarães J, Sprooten E, Beckmann CF, Franke B, Bralten J. Shared genetic influences on resting‐state functional networks of the brain. Hum Brain Mapp 2022; 43:1787-1803. [PMID: 35076988 PMCID: PMC8933256 DOI: 10.1002/hbm.25712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 10/23/2021] [Accepted: 10/24/2021] [Indexed: 11/17/2022] Open
Abstract
The amplitude of activation in brain resting state networks (RSNs), measured with resting‐state functional magnetic resonance imaging, is heritable and genetically correlated across RSNs, indicating pleiotropy. Recent univariate genome‐wide association studies (GWASs) explored the genetic underpinnings of individual variation in RSN activity. Yet univariate genomic analyses do not describe the pleiotropic nature of RSNs. In this study, we used a novel multivariate method called genomic structural equation modeling to model latent factors that capture the shared genomic influence on RSNs and to identify single nucleotide polymorphisms (SNPs) and genes driving this pleiotropy. Using summary statistics from GWAS of 21 RSNs reported in UK Biobank (N = 31,688), the genomic latent factor analysis was first conducted in a discovery sample (N = 21,081), and then tested in an independent sample from the same cohort (N = 10,607). In the discovery sample, we show that the genetic organization of RSNs can be best explained by two distinct but correlated genetic factors that divide multimodal association networks and sensory networks. Eleven of the 17 factor loadings were replicated in the independent sample. With the multivariate GWAS, we found and replicated nine independent SNPs associated with the joint architecture of RSNs. Further, by combining the discovery and replication samples, we discovered additional SNP and gene associations with the two factors of RSN amplitude. We conclude that modeling the genetic effects on brain function in a multivariate way is a powerful approach to learn more about the biological mechanisms involved in brain function.
Collapse
Affiliation(s)
- João P.O.F.T. Guimarães
- Department of Cognitive Neuroscience Radboud University Medical Center Nijmegen The Netherlands
- Donders Institute for Brain, Cognition and Behaviour Radboud University Nijmegen The Netherlands
- Department of Human Genetics Radboud University Medical Center Nijmegen The Netherlands
| | - E. Sprooten
- Department of Cognitive Neuroscience Radboud University Medical Center Nijmegen The Netherlands
- Donders Institute for Brain, Cognition and Behaviour Radboud University Nijmegen The Netherlands
- Department of Human Genetics Radboud University Medical Center Nijmegen The Netherlands
| | - C. F. Beckmann
- Department of Cognitive Neuroscience Radboud University Medical Center Nijmegen The Netherlands
- Donders Institute for Brain, Cognition and Behaviour Radboud University Nijmegen The Netherlands
- Centre for Functional MRI of the Brain (FMRIB) University of Oxford Oxford UK
| | - B. Franke
- Donders Institute for Brain, Cognition and Behaviour Radboud University Nijmegen The Netherlands
- Department of Human Genetics Radboud University Medical Center Nijmegen The Netherlands
- Department of Psychiatry Radboud University Medical Center Nijmegen The Netherlands
| | - J. Bralten
- Donders Institute for Brain, Cognition and Behaviour Radboud University Nijmegen The Netherlands
- Department of Human Genetics Radboud University Medical Center Nijmegen The Netherlands
| |
Collapse
|
192
|
Vieira-Rocha MS, Rodriguez-Rodriguez P, Ferreira-Duarte M, Faria M, Sousa JB, Morato M, Arribas SM, Diniz C. Fetal Undernutrition Modifies Vascular RAS Balance Enhancing Oxidative Damage and Contributing to Remodeling. Int J Mol Sci 2022; 23:1233. [PMID: 35163158 PMCID: PMC8835999 DOI: 10.3390/ijms23031233] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/18/2022] [Accepted: 01/21/2022] [Indexed: 12/21/2022] Open
Abstract
Fetal stress is known to increase susceptibility to cardiometabolic diseases and hypertension in adult age in a process known as fetal programming. This study investigated the relationship between vascular RAS, oxidative damage and remodeling in fetal programming. Six-month old Sprague-Dawley offspring from mothers that were fed ad libitum (CONTROL) or with 50% intake during the second half of gestation (maternal undernutrition, MUN) were used. qPCR or immunohistochemistry were used to obtain the expression of receptors and enzymes. Plasma levels of carbonyls were measured by spectrophotometry. In mesenteric arteries from MUN rats we detected an upregulation of ACE, ACE2, AT1 receptors and NADPH oxidase, and lower expression of AT2, Mas and MrgD receptors compared to CONTROL. Systolic and diastolic blood pressure and plasma levels of carbonyls were higher in MUN than in CONTROL. Vascular morphology evidenced an increased media/lumen ratio and adventitia/lumen ratio, and more connective tissue in MUN compared to CONTROL. In conclusion, fetal undernutrition indices RAS alterations and oxidative damage which may contribute to the remodeling of mesenteric arteries, and increase the risk of adverse cardiovascular events and hypertension.
Collapse
Affiliation(s)
- Maria Sofia Vieira-Rocha
- Laboratory of Pharmacology, Department of Drug Science, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (M.F.-D.); (J.B.S.); (M.M.)
- LAQV/REQUIMTE, Faculty of Pharmacy, University of Porto, 4099-002 Porto, Portugal;
| | - Pilar Rodriguez-Rodriguez
- Department of Physiology, Faculty of Medicine, Universidad Autonoma de Madrid, 28049 Madrid, Spain; (P.R.-R.); (S.M.A.)
| | - Mariana Ferreira-Duarte
- Laboratory of Pharmacology, Department of Drug Science, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (M.F.-D.); (J.B.S.); (M.M.)
- LAQV/REQUIMTE, Faculty of Pharmacy, University of Porto, 4099-002 Porto, Portugal;
| | - Miguel Faria
- LAQV/REQUIMTE, Faculty of Pharmacy, University of Porto, 4099-002 Porto, Portugal;
- Laboratory of Bromatology and Hydrology, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4099-002 Porto, Portugal
| | - Joana Beatriz Sousa
- Laboratory of Pharmacology, Department of Drug Science, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (M.F.-D.); (J.B.S.); (M.M.)
- LAQV/REQUIMTE, Faculty of Pharmacy, University of Porto, 4099-002 Porto, Portugal;
| | - Manuela Morato
- Laboratory of Pharmacology, Department of Drug Science, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (M.F.-D.); (J.B.S.); (M.M.)
- LAQV/REQUIMTE, Faculty of Pharmacy, University of Porto, 4099-002 Porto, Portugal;
| | - Silvia Magdalena Arribas
- Department of Physiology, Faculty of Medicine, Universidad Autonoma de Madrid, 28049 Madrid, Spain; (P.R.-R.); (S.M.A.)
| | - Carmen Diniz
- Laboratory of Pharmacology, Department of Drug Science, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (M.F.-D.); (J.B.S.); (M.M.)
- LAQV/REQUIMTE, Faculty of Pharmacy, University of Porto, 4099-002 Porto, Portugal;
| |
Collapse
|
193
|
Chaudhary M. Novel methylation mark and essential hypertension. JOURNAL OF GENETIC ENGINEERING AND BIOTECHNOLOGY 2022; 20:11. [PMID: 35061109 PMCID: PMC8777530 DOI: 10.1186/s43141-022-00301-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 01/14/2022] [Indexed: 12/11/2022]
Abstract
Background Essential hypertension (EH) is an important risk factor for various cardiovascular, cerebral and renal disorders. It is a multi-factorial trait which occurs through complex interplay between genetic, epigenetic, and environmental factors. Even after advancement of technology and deciphering the involvement of multiple signalling pathways in blood pressure regulation, it still remains as a huge global concern. Main body of the abstract Genome-wide association studies (GWAS) have revealed EH-associated genetic variants but these solely cannot explain the variability in blood pressure indicating the involvement of additional factors. The etiopathogenesis of hypertension has now advanced to the level of epigenomics where aberrant DNA methylation is the most defined epigenetic mechanism to be involved in gene regulation. Though role of DNA methylation in cancer and other mechanisms is deeply studied but this mechanism is in infancy in relation to hypertension. Generally, 5-methylcytosine (5mC) levels are being targeted at both individual gene and global level to find association with the disease. But recently, with advanced sequencing techniques another methylation mark, N6-methyladenine (6mA) was found and studied in humans which was earlier considered to be absent in case of eukaryotes. Relation of aberrant 6mA levels with cancer and stem cell fate has drawn attention to target 6mA levels with hypertension too. Conclusion Recent studies targeting hypertension has suggested 6mA levels as novel marker and its demethylase, ALKBH1 as probable therapeutic target to prevent hypertension through epigenetic programming. This review compiles different methylation studies and suggests targeting of both 5mC and 6mA levels to cover role of methylation in hypertension in broader scenario.
Collapse
|
194
|
Oral postmenopausal hormone therapy and genetic risk on venous thromboembolism: gene-hormone interaction results from a large prospective cohort study. Menopause 2022; 29:293-303. [PMID: 35013060 PMCID: PMC8881382 DOI: 10.1097/gme.0000000000001924] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
OBJECTIVE Oral postmenopausal hormone therapy (HT) has been shown to be associated with venous thromboembolism (VTE), but whether this association is modified by VTE-associated genetic susceptibility is unknown. We examined interactions between oral HT use and a genetic risk score (GRS) of VTE. METHOD Eligible women were postmenopausal women who had data on oral HT use, VTE incidence between 1990 and 2012, and genetic data in the Nurses' Health Study. We built a GRS aggregating 16 VTE-related genetic variants. We used Cox regression to estimate associations of HT use with incident VTE and assessed interactions between HT use and VTE GRS. We also estimated incidence of VTE between age 50 and 79 years for groups of women defined by HT use and VTE GRS. RESULTS We identified 432 incident VTE cases. Current HT users were at higher risk of VTE than never users (HR: 1.9, 95% CI: 1.5-2.6), with slightly higher risk for estrogen plus progestin HT than estrogen only (HR: 2.4 vs 1.9). The GRS was associated with VTE risk (HR comparing 4th quartile to 1st: 2.0, 95% CI: 1.2-3.4). We did not observe significant multiplicative interactions between HT use and GRS. The estimated VTE risk difference (per 10,000 person-years) comparing 50-year-old current HT users to never users was 22.5 for women in the highest GRS quartile and 9.8 for women in the lowest GRS quartile. CONCLUSION The VTE GRS might inform clinical guidance regarding the balance of risks and benefits of HT use, especially among younger women.
Collapse
|
195
|
Takei Y. Evolution of the membrane/particulate guanylyl cyclase: From physicochemical sensors to hormone receptors. Gen Comp Endocrinol 2022; 315:113797. [PMID: 33957096 DOI: 10.1016/j.ygcen.2021.113797] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 04/19/2021] [Accepted: 04/28/2021] [Indexed: 12/26/2022]
Abstract
Guanylyl cyclase (GC) is an enzyme that produces 3',5'-cyclic guanosine monophosphate (cGMP), one of the two canonical cyclic nucleotides used as a second messenger for intracellular signal transduction. The GCs are classified into two groups, particulate/membrane GCs (pGC) and soluble/cytosolic GCs (sGC). In relation to the endocrine system, pGCs include hormone receptors for natriuretic peptides (GC-A and GC-B) and guanylin peptides (GC-C), while sGC is a receptor for nitric oxide and carbon monoxide. Comparing the functions of pGCs in eukaryotes, it is apparent that pGCs perceive various environmental factors such as light, temperature, and various external chemical signals in addition to endocrine hormones, and transmit the information into the cell using the intracellular signaling cascade initiated by cGMP, e.g., cGMP-dependent protein kinases, cGMP-sensitive cyclic nucleotide-gated ion channels and cGMP-regulated phosphodiesterases. Among vertebrate pGCs, GC-E and GC-F are localized on retinal epithelia and are involved in modifying signal transduction from the photoreceptor, rhodopsin. GC-D and GC-G are localized in olfactory epithelia and serve as sensors at the extracellular domain for external chemical signals such as odorants and pheromones. GC-G also responds to guanylin peptides in the urine, which alters sensitivity to other chemicals. In addition, guanylin peptides that are secreted into the intestinal lumen, a pseudo-external environment, act on the GC-C on the apical membrane for regulation of epithelial transport. In this context, GC-C and GC-G appear to be in transition from exocrine pheromone receptor to endocrine hormone receptor. The pGCs also exist in various deuterostome and protostome invertebrates, and act as receptors for environmental, exocrine and endocrine factors including hormones. Tracing the evolutionary history of pGCs, it appears that pGCs first appeared as a sensor for physicochemical signals in the environment, and then evolved to function as hormone receptors. In this review, the author proposes an evolutionary history of pGCs that highlights the emerging role of the GC/cGMP system for signal transduction in hormone action.
Collapse
Affiliation(s)
- Yoshio Takei
- Laboratory of Physiology, Department of Marine Bioscience, Atmosphere and Ocean Research Institute, The University of Tokyo, Kashiwa, Chiba 277-8564, Japan.
| |
Collapse
|
196
|
Svoboda LK, Ishikawa T, Dolinoy DC. Developmental toxicant exposures and sex-specific effects on epigenetic programming and cardiovascular health across generations. ENVIRONMENTAL EPIGENETICS 2022; 8:dvac017. [PMID: 36325489 PMCID: PMC9600458 DOI: 10.1093/eep/dvac017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 09/12/2022] [Accepted: 10/01/2022] [Indexed: 05/15/2023]
Abstract
Despite substantial strides in diagnosis and treatment, cardiovascular diseases (CVDs) continue to represent the leading cause of death in the USA and around the world, resulting in significant morbidity and loss of productive years of life. It is increasingly evident that environmental exposures during early development can influence CVD risk across the life course. CVDs exhibit marked sexual dimorphism, but how sex interacts with environmental exposures to affect cardiovascular health is a critical and understudied area of environmental health. Emerging evidence suggests that developmental exposures may have multi- and transgenerational effects on cardiovascular health, with potential sex differences; however, further research in this important area is urgently needed. Lead (Pb), phthalate plasticizers, and perfluoroalkyl substances (PFAS) are ubiquitous environmental contaminants with numerous adverse human health effects. Notably, recent evidence suggests that developmental exposure to each of these toxicants has sex-specific effects on cardiovascular outcomes, but the underlying mechanisms, and their effects on future generations, require further investigation. This review article will highlight the role for the developmental environment in influencing cardiovascular health across generations, with a particular emphasis on sex differences and epigenetic mechanisms. In particular, we will focus on the current evidence for adverse multi and transgenerational effects of developmental exposures to Pb, phthalates, and PFAS and highlight areas where further research is needed.
Collapse
Affiliation(s)
- Laurie K Svoboda
- *Correspondence address. Environmental Health Sciences, University of Michigan, School of Public Health, 1415 Washington Heights, Ann Arbor, MI 48109, USA. Tel: +734-764-2032; E-mail:
| | - Tomoko Ishikawa
- Environmental Health Sciences, University of Michigan, School of Public Health, 1415 Washington Heights, Ann Arbor, MI 48109, USA
| | - Dana C Dolinoy
- Environmental Health Sciences, University of Michigan, School of Public Health, 1415 Washington Heights, Ann Arbor, MI 48109, USA
- Nutritional Sciences, University of Michigan, School of Public Health, 1415 Washington Heights, Ann Arbor, MI 48109, USA
| |
Collapse
|
197
|
Wang G, Bhatta L, Moen GH, Hwang LD, Kemp JP, Bond TA, Åsvold BO, Brumpton B, Evans DM, Warrington NM. Investigating a Potential Causal Relationship Between Maternal Blood Pressure During Pregnancy and Future Offspring Cardiometabolic Health. Hypertension 2022; 79:170-177. [PMID: 34784738 PMCID: PMC8654122 DOI: 10.1161/hypertensionaha.121.17701] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 10/20/2021] [Indexed: 12/20/2022]
Abstract
Observational epidemiological studies have reported that higher maternal blood pressure (BP) during pregnancy is associated with increased future risk of offspring cardiometabolic disease. However, it is unclear whether this association represents a causal relationship through intrauterine mechanisms. We used a Mendelian randomization (MR) framework to examine the relationship between unweighted maternal genetic scores for systolic BP and diastolic BP and a range of cardiometabolic risk factors in the offspring of up to 29 708 genotyped mother-offspring pairs from the UKB study (UK Biobank) and the HUNT study (Trøndelag Health). We conducted similar analyses in up to 21 423 father-offspring pairs from the same cohorts. We confirmed that the BP-associated genetic variants from the general population sample also had similar effects on maternal BP during pregnancy in independent cohorts. We did not detect any association between maternal (or paternal) unweighted genetic scores and cardiometabolic offspring outcomes in the meta-analysis of UKB and HUNT after adjusting for offspring genotypes at the same loci. We find little evidence to support the notion that maternal BP is a major causal risk factor for adverse offspring cardiometabolic outcomes in later life.
Collapse
Affiliation(s)
- Geng Wang
- The University of Queensland Diamantina Institute (G.W., G.-H.M., L.-D.H., J.P.K., T.A.B., D.M.E., N.M.W.), The University of Queensland, Brisbane, Australia
| | - Laxmi Bhatta
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Trondheim, Norway (L.B., G.-H.M., B.O.A., B.B., N.M.W.)
| | - Gunn-Helen Moen
- The University of Queensland Diamantina Institute (G.W., G.-H.M., L.-D.H., J.P.K., T.A.B., D.M.E., N.M.W.), The University of Queensland, Brisbane, Australia
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Trondheim, Norway (L.B., G.-H.M., B.O.A., B.B., N.M.W.)
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Norway (G.-H.M.)
- Population Health Sciences, Bristol Medical School (G.-H.M., T.A.B.), University of Bristol, United Kingdom
| | - Liang-Dar Hwang
- The University of Queensland Diamantina Institute (G.W., G.-H.M., L.-D.H., J.P.K., T.A.B., D.M.E., N.M.W.), The University of Queensland, Brisbane, Australia
- Institute of Molecular Bioscience (L.-D.H., J.P.K., D.M.E., N.M.W.), The University of Queensland, Brisbane, Australia
| | - John P. Kemp
- The University of Queensland Diamantina Institute (G.W., G.-H.M., L.-D.H., J.P.K., T.A.B., D.M.E., N.M.W.), The University of Queensland, Brisbane, Australia
- Institute of Molecular Bioscience (L.-D.H., J.P.K., D.M.E., N.M.W.), The University of Queensland, Brisbane, Australia
- Medical Research Council Integrative Epidemiology Unit (J.P.K., T.A.B., D.M.E., N.M.W.), University of Bristol, United Kingdom
| | - Tom A. Bond
- The University of Queensland Diamantina Institute (G.W., G.-H.M., L.-D.H., J.P.K., T.A.B., D.M.E., N.M.W.), The University of Queensland, Brisbane, Australia
- Population Health Sciences, Bristol Medical School (G.-H.M., T.A.B.), University of Bristol, United Kingdom
- Medical Research Council Integrative Epidemiology Unit (J.P.K., T.A.B., D.M.E., N.M.W.), University of Bristol, United Kingdom
| | - Bjørn Olav Åsvold
- Department of Endocrinology, Clinic of Medicine (B.O.A.), St Olavs Hospital, Trondheim University Hospital, Norway
- HUNT Research Center, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Levanger, Norway (B.O.A., B.B.)
| | - Ben Brumpton
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Trondheim, Norway (L.B., G.-H.M., B.O.A., B.B., N.M.W.)
- Clinic of Medicine (B.B.), St Olavs Hospital, Trondheim University Hospital, Norway
- HUNT Research Center, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Levanger, Norway (B.O.A., B.B.)
| | - David M. Evans
- The University of Queensland Diamantina Institute (G.W., G.-H.M., L.-D.H., J.P.K., T.A.B., D.M.E., N.M.W.), The University of Queensland, Brisbane, Australia
- Institute of Molecular Bioscience (L.-D.H., J.P.K., D.M.E., N.M.W.), The University of Queensland, Brisbane, Australia
- Medical Research Council Integrative Epidemiology Unit (J.P.K., T.A.B., D.M.E., N.M.W.), University of Bristol, United Kingdom
| | - Nicole M. Warrington
- The University of Queensland Diamantina Institute (G.W., G.-H.M., L.-D.H., J.P.K., T.A.B., D.M.E., N.M.W.), The University of Queensland, Brisbane, Australia
- Institute of Molecular Bioscience (L.-D.H., J.P.K., D.M.E., N.M.W.), The University of Queensland, Brisbane, Australia
- Medical Research Council Integrative Epidemiology Unit (J.P.K., T.A.B., D.M.E., N.M.W.), University of Bristol, United Kingdom
| |
Collapse
|
198
|
Fujishiro H, Kambe T. Manganese transport in mammals by zinc transporter family proteins, ZNT and ZIP. J Pharmacol Sci 2021; 148:125-133. [PMID: 34924116 DOI: 10.1016/j.jphs.2021.10.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 10/07/2021] [Accepted: 10/07/2021] [Indexed: 12/14/2022] Open
Abstract
Manganese (Mn) is an essential trace element required for various biological processes. However, excess Mn causes serious side effects in humans, including parkinsonism. Thus, elucidation of Mn homeostasis at the systemic, cellular, and molecular levels is important. Many metal transporters and channels can be involved in the transport and homeostasis of Mn, and an increasing body of evidence shows that several zinc (Zn) transporters belonging to the ZIP and ZNT families, specifically, ZNT10, ZIP8, and ZIP14, play pivotal roles in Mn metabolism. Mutations in the genes encoding these transporter proteins are associated with congenital disorders related to dysregulated Mn homeostasis in humans. Moreover, single nucleotide polymorphisms of ZIP8 are associated with multiple clinical phenotypes. In this review, we discuss the recent literature on the structural and biochemical features of ZNT10, ZIP8, and ZIP14, including transport mechanisms, regulation of expression, and pathophysiological functions. Because a disturbance in Mn homeostasis is closely associated with a variety of phenotypes and risk of human diseases, these transporters constitute a significant target for drug development. An understanding of the roles of these key transporters in Mn metabolism should provide new insights into pharmacological applications of their inhibitors and enhancers in human diseases.
Collapse
Affiliation(s)
- Hitomi Fujishiro
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima, 770-8514, Japan.
| | - Taiho Kambe
- Division of Integrated Life Science, Graduate School of Biostudies, Kyoto University, Kyoto, 606-8502, Japan.
| |
Collapse
|
199
|
Jansen SA, Huiskens B, Trompet S, Jukema JW, Mooijaart SP, Willems van Dijk K, van Heemst D, Noordam R. Classical risk factors for primary coronary artery disease from an aging perspective through Mendelian Randomization. GeroScience 2021; 44:1703-1713. [PMID: 34932184 PMCID: PMC9213623 DOI: 10.1007/s11357-021-00498-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 12/11/2021] [Indexed: 11/25/2022] Open
Abstract
The significance of classical risk factors in coronary artery disease (CAD) remains unclear in older age due to possible changes in underlying disease pathologies. Therefore, we conducted Mendelian Randomization approaches to investigate the causal relationship between classical risk factors and primary CAD in different age groups. A Mendelian Randomization study was conducted in European-ethnicity individuals from the UK Biobank population. Analyses were performed using data of 22,313 CAD cases (71.6% men) and 407,920 controls (44.5% men). Using logistic regression analyses, we investigated the associations between standardized genetic risk score and primary CAD stratified by age of diagnosis. In addition, feature importance and model accuracy were assessed in different age groups to evaluate predictive power of the genetic risk scores with increasing age. We found age-dependent associations for all classical CAD risk factors. Notably, body mass index (OR 1.22 diagnosis < 50 years; OR 1.02 diagnosis > 70 years), blood pressure (OR 1.12 < 50 years; OR 1.04 > 70 years), LDL cholesterol (OR 1.16 < 50 years; OR 1.02 > 70 years), and triglyceride levels (OR 1.11 < 50 years; 1.04 > 70 years). In line with the Mendelian Randomization analyses, model accuracy and feature importance of the classical risk factors decreased with increasing age of diagnosis. Causal determinants for primary CAD are age dependent with classical CAD risk factors attenuating in relation with primary CAD with increasing age. These results question the need for (some) currently applied cardiovascular disease risk reducing interventions at older age.
Collapse
Affiliation(s)
- Swetta A Jansen
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, PO Box 9600, 2300RC, Leiden, The Netherlands
- Data Science Lab, Amsterdam, the Netherlands
| | | | - Stella Trompet
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, PO Box 9600, 2300RC, Leiden, The Netherlands
| | - JWouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, the Netherlands
- Netherlands Heart Institute, Utrecht, the Netherlands
| | - Simon P Mooijaart
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, PO Box 9600, 2300RC, Leiden, The Netherlands
| | - Ko Willems van Dijk
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
- Department of Internal Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, the Netherlands
- Leiden Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Diana van Heemst
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, PO Box 9600, 2300RC, Leiden, The Netherlands
| | - Raymond Noordam
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, PO Box 9600, 2300RC, Leiden, The Netherlands.
| |
Collapse
|
200
|
Geng YJ, Madonna R, Hermida RC, Smolensky MH. Pharmacogenomics and circadian rhythms as mediators of cardiovascular drug-drug interactions. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2021; 2:100025. [PMID: 34909660 PMCID: PMC8663962 DOI: 10.1016/j.crphar.2021.100025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 03/28/2021] [Accepted: 04/07/2021] [Indexed: 11/01/2022] Open
Abstract
This article summarizes the current literature and documents new evidence concerning drug-drug interactions (DDI) stemming from pharmacogenomic and circadian rhythm determinants of therapies used to treat common cardiovascular diseases (CVD), such as atherosclerosis and hypertension. Patients with CVD often have more than one pathophysiologic condition, namely metabolic syndromes, hypertension, hyperlipidemia, and hyperglycemia, among others, which necessitate polytherapeutic or polypharmaceutic management. Interactions between drugs, drugs and food/food supplements, or drugs and genetic/epigenetic factors may have adverse impacts on the cardiovascular and other systems of the body. The mechanisms underlying cardiovascular DDI may involve the formation of a complex pharmacointeractome, including the absorption, distribution, metabolism, and elimination of drugs, which affect their respective bioavailability, efficacy, and/or harmful metabolites. The pharmacointeractome of cardiovascular drugs is likely operated with endogenous rhythms controlled by circadian clock genes. Basic and clinical investigations have improved the knowledge and understanding of cardiovascular pharmacogenomics and pharmacointeractomes, and additionally they have presented new evidence that the staging of deterministic circadian rhythms, according to the dosing time of drugs, e.g., upon awakening vs. at bedtime, cannot only differentially impact their pharmacokinetics and pharmacodynamics but also mediate agonistic/synergetic or antagonistic DDI. To properly manage CVD patients and avoid DDI, it is important that clinicians have sufficient knowledge of their multiple risk factors, i.e., age, gender, and life style elements (like diet, smoking, psychological stress, and alcohol consumption), and comorbidities, such as diabetes, hypertension, dyslipidemia, and depression, and the potential interactions between genetic or epigenetic background of their prescribed therapeutics.
Collapse
Affiliation(s)
- Yong-Jian Geng
- Department of Internal Medicine, McGovern School of Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Rosalinda Madonna
- Department of Internal Medicine, McGovern School of Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA.,Chair of Cardiology, Department of Surgical, Medical and Molecular Pathology, University of Pisa, Pisa, Italy
| | - Ramon C Hermida
- Bioengineering & Chronobiology Laboratories, Atlantic Research Center for Information and Communication Technologies (atlanTTic), Universidade de Vigo, Vigo, Spain.,Department of Biomedical Engineering, Cockrell School of Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Michael H Smolensky
- Department of Internal Medicine, McGovern School of Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA.,Department of Biomedical Engineering, Cockrell School of Engineering, The University of Texas at Austin, Austin, TX, USA
| |
Collapse
|