1
|
Gard AJM, Lavallee D. Examining the Relationship Between Urinary Incontinence and Women's Physical Activity Engagement: Barriers and Disclosure Patterns. Healthcare (Basel) 2025; 13:856. [PMID: 40281805 PMCID: PMC12026561 DOI: 10.3390/healthcare13080856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 03/27/2025] [Accepted: 04/04/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND This study investigates the relationship between urinary incontinence (UI) and women's participation in physical activity (PA). Women are less active than men across their lifespan, and while interventions aim to bridge this gap, the unique challenge posed by UI remains underexplored. UI disproportionately affects women and often results in reduced self-confidence and avoidance of PA. METHODS Employing a mixed-methods design, the study utilised an online survey (n = 345) and semi-structured interviews (n = 14) to explore women's experiences of UI during PA and its perceived impact relative to other barriers. RESULTS Findings reveal that UI ranks prominently among barriers to PA, yet disclosure is infrequent without direct prompting. Participants highlighted anxiety, embarrassment, and the inadequacy of PA environments in accommodating UI-related needs as critical deterrents. Interviews further uncovered a lack of practitioner knowledge regarding UI, with many women favouring privacy-centric approaches to address their concerns. CONCLUSIONS The study underscores the necessity for tailored interventions, practitioner education, and inclusive PA environments to enhance participation and mitigate UI's impact. These findings contribute to broader efforts to promote gender equity in PA and improve women's health outcomes.
Collapse
Affiliation(s)
| | - David Lavallee
- Department of Health, Sport and Wellbeing, Abertay University, Dundee DD1 1HG, UK;
| |
Collapse
|
2
|
Boyd A, Evans KM, Turner EL, Flaig R, Oakley J, Campbell KC, Thomas R, McLachlan S, Crane M, Whitehorn R, Calkin R, Hill A, Berman S, Ford D, Tobin M, Porteous D, Gomes DF, Garcia MP, Wong A, Sanchez A, Orton C, Thompson S, Gulliver J, Adams K, Badrick E, Batini C, Benzeval M, Boatman S, Breen G, Bristow S, Britten A, Bryant L, Butterworth A, Campbell A, Chave S, Danesh J, Das-Munshi J, Dennison K, Angelantonio ED, Eley TC, Fisher H, Fitzsimons E, Goodman A, Gregg M, Guyatt AL, Hansell A, Harmston R, Heard A, Henderson M, Hill R, Huang SC, John C, Kee F, Kingston N, Kneeshaw J, Kumar R, Lachance G, Lockhart C, Lockhart-Jones H, Markham S, Mason D, McGuinness B, McKenzie M, McMahon A, Malouf CM, Mumme M, Neville C, Northstone K, Oldfield Z, O’Neill D, Pareek M, Pickavance J, Rahman Y, Reilly H, Scott A, Smith D, Steptoe A, Steves C, Sudlow C, Sze G, Timpson NL, Tungamirai T, Venn L, Walker M, Walker N, Wareham N, Watmuff A, Webb T, Williams K, Wright J, Yarand D, Ploubidis GB, Macleod J, Sterne JAC, Chaturvedi N. UK Longitudinal Linkage Collaboration (UK LLC): The National Trusted Research Environment for Longitudinal Research. Int J Popul Data Sci 2025; 10:2468. [PMID: 40129687 PMCID: PMC11931487 DOI: 10.23889/ijpds.v10i1.2468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2025] Open
Abstract
Introduction The UK Longitudinal Linkage Collaboration (UK LLC) is the national Trusted Research Environment (TRE) for the UK's longitudinal research community, supporting the UK's unparalleled collection of Longitudinal Population Studies (LPS). Initially set up as a COVID-19 research resource, UK LLC is now a generic database for any research for the public good. Objectives UK LLC supports longitudinal research by providing record linkage and TRE services. Methods The UK LLC partnership provides a secure analytics environment, a trusted third-party linkage processor and a comprehensive governance framework to minimise risks to participant confidentiality. UK LLC is ISO 27001 certified and accredited by the UK Statistics Authority as a processor under the Digital Economy Act. The active involvement by members of UK LLC's public involvement programme ensures UK LLC is acceptable to LPS participants and the wider public. All UK LPS are eligible for inclusion. Researchers can apply to access the TRE via an approach that fulfils the needs of the LPS, the linked data owners and includes a review by public contributors. Results Twenty-two LPS have so far joined UK LLC. Where permissions allow, participants are linked to their National Health Service (NHS) England, NHS Wales and place-based records, with work ongoing to link to NHS Scotland and non-health administrative records, including Department for Work and Pensions and His Majesty's (HM) Revenue and Customs. UK LLC Explore allows potential researchers to discover the breadth of data available in the TRE. All applications are listed on UK LLC's publicly accessible Data Access Register. Conclusions UK LLC enables researchers to interrogate pooled LPS participant data that are systematically linked to diverse records. UK LLC remains open to additional LPS joining the partnership and will increase the breadth of data to support the longitudinal research community and attract increasing numbers of researchers across multiple disciplines, government departments and industry.
Collapse
Affiliation(s)
- Andy Boyd
- UK Longitudinal Linkage Collaboration, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS8 2PS, UK
| | - Katharine M. Evans
- UK Longitudinal Linkage Collaboration, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS8 2PS, UK
| | - Emma L. Turner
- UK Longitudinal Linkage Collaboration, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS8 2PS, UK
| | - Robin Flaig
- Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh, EH4 2XU, UK
- UK Longitudinal Linkage Collaboration, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, EH16 4UX, UK
| | - Jacqui Oakley
- UK Longitudinal Linkage Collaboration, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS8 2PS, UK
| | - Kirsteen C. Campbell
- UK Longitudinal Linkage Collaboration, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, EH16 4UX, UK
| | - Richard Thomas
- UK Longitudinal Linkage Collaboration, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS8 2PS, UK
| | - Stela McLachlan
- Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh, EH4 2XU, UK
- UK Longitudinal Linkage Collaboration, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, EH16 4UX, UK
| | - Matthew Crane
- UK Longitudinal Linkage Collaboration, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS8 2PS, UK
| | - Rebecca Whitehorn
- Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh, EH4 2XU, UK
- UK Longitudinal Linkage Collaboration, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, EH16 4UX, UK
| | - Rachel Calkin
- UK Longitudinal Linkage Collaboration, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS8 2PS, UK
| | - Abigail Hill
- UK Longitudinal Linkage Collaboration, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS8 2PS, UK
| | - Samantha Berman
- UK Longitudinal Linkage Collaboration, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS8 2PS, UK
| | - David Ford
- Population Data Science, Swansea University, Swansea, SA2 8PP, UK
| | - Martin Tobin
- Population Health Sciences, University of Leicester, Leicester, LE1 7RH, UK
- University Hospitals of Leicester NHS Trust, Leicester, LE1 5WW, UK
| | - David Porteous
- Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh, EH4 2XU, UK
- UK Longitudinal Linkage Collaboration, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, EH16 4UX, UK
| | - Danielle F. Gomes
- Centre for Longitudinal Studies, University College London, London, WC1H 0AL, UK
| | - Maria-Paz Garcia
- Department of Twin Research & Genetic Epidemiology, King’s College London, London, SE1 7EH, UK
| | - Andrew Wong
- MRC Unit of Lifelong Health & Ageing, University College London, London, WC1E 7HB, UK
| | - Aida Sanchez
- Centre for Longitudinal Studies, University College London, London, WC1H 0AL, UK
| | - Chris Orton
- Population Data Science, Swansea University, Swansea, SA2 8PP, UK
| | - Simon Thompson
- Population Data Science, Swansea University, Swansea, SA2 8PP, UK
| | - John Gulliver
- Population Health Research Institute, City St George’s, University of London, Cranmer Terrace, London, SW17 0RE, UK
| | - Kathryn Adams
- Centre for Environment, Sustainability and Health, University of Leicester, Leicester, LE1 7RH, UK
| | - Ellena Badrick
- Bradford Institute for Health Research, Bradford Teaching Hospitals NHS Foundation Trust, Bradford, BD9 6RJ, UK
| | - Chiara Batini
- Population Health Sciences, University of Leicester, Leicester, LE1 7RH, UK
| | | | - Susie Boatman
- MRC Epidemiology Unit, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Gerome Breen
- Social Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, SE5 8AF, UK
- UK National Institute for Health and Social Care Research (NIHR) Biomedical Research Centre for Mental Health, South London and Maudsley Hospital, London, SE5 8AF, UK
| | - Shannon Bristow
- Social Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, SE5 8AF, UK
- UK National Institute for Health and Social Care Research (NIHR) Biomedical Research Centre for Mental Health, South London and Maudsley Hospital, London, SE5 8AF, UK
| | - Abigail Britten
- MRC Epidemiology Unit, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Luke Bryant
- Population Health Sciences, University of Leicester, Leicester, LE1 7RH, UK
| | - Adam Butterworth
- Division of Cardiovascular Medicine, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Archie Campbell
- Generation Scotland, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Sarah Chave
- UK LLC Public Involvement Programme, UK Longitudinal Linkage Collaboration, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, EH16 4UX, UK
| | - John Danesh
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB2 0BB, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, CB2 0BB, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, CB2 0BB, UK
- National Institute for Health and Care Research Blood and Transplant Research Unit in Donor Health and Behaviour, University of Cambridge, Cambridge, CB2 0QQ, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, CB10 1SA, UK
- Department of Human Genetics, Wellcome Sanger Institute, Hinxton, CB10 1SA, UK
| | - Jayati Das-Munshi
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, SE5 8AF, UK
| | - Karen Dennison
- Centre for Longitudinal Studies, University College London, London, WC1H 0AL, UK
| | - Emanuele Di Angelantonio
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB2 0BB, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, CB2 0BB, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, CB2 0BB, UK
- National Institute for Health and Care Research Blood and Transplant Research Unit in Donor Health and Behaviour, University of Cambridge, Cambridge, CB2 0QQ, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, CB10 1SA, UK
- Health Data Science Research Centre, Human Technopole, Milan, 20157, Italy
| | - Thalia C. Eley
- Social Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, SE5 8AF, UK
- UK National Institute for Health and Social Care Research (NIHR) Biomedical Research Centre for Mental Health, South London and Maudsley Hospital, London, SE5 8AF, UK
| | - Helen Fisher
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, SE5 8AF, UK
| | - Emla Fitzsimons
- Millennium Cohort Study, University College London, London, WC1H 0AL, UK
| | - Alissa Goodman
- Centre for Longitudinal Studies, University College London, London, WC1H 0AL, UK
| | - Michael Gregg
- UK LLC Public Involvement Programme, UK Longitudinal Linkage Collaboration, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, EH16 4UX, UK
| | - Anna L Guyatt
- Population Health Sciences, University of Leicester, Leicester, LE1 7RH, UK
| | - Anna Hansell
- Centre for Environment, Sustainability and Health, University of Leicester, Leicester, LE1 7RH, UK
| | - Rebecca Harmston
- UK LLC Public Involvement Programme, UK Longitudinal Linkage Collaboration, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, EH16 4UX, UK
| | - Andy Heard
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, Norfolk Place, London, SW7 2AZ, UK
| | - Morag Henderson
- Centre for Longitudinal Studies, University College London, London, WC1H 0AL, UK
| | - Rosie Hill
- UK LLC Public Involvement Programme, UK Longitudinal Linkage Collaboration, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, EH16 4UX, UK
| | - Szu-Chia Huang
- UK LLC Public Involvement Programme, UK Longitudinal Linkage Collaboration, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, EH16 4UX, UK
| | - Catherine John
- Population Health Sciences, University of Leicester, Leicester, LE1 7RH, UK
- University Hospitals of Leicester NHS Trust, Leicester, LE1 5WW, UK
| | - Frank Kee
- Centre for Public Health, Queens University Belfast, Belfast, BT12 6BA, UK
| | - Nathalie Kingston
- NIHR BioResource, Cambridge University Hospitals NHS Foundation Trust, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK
- Department of Haematology, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK
| | - Jack Kneeshaw
- Understanding Society, University of Essex, Colchester, CO4 3SQ, UK
| | - Rashmi Kumar
- UK LLC Public Involvement Programme, UK Longitudinal Linkage Collaboration, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, EH16 4UX, UK
| | - Genevieve Lachance
- Department of Twin Research & Genetic Epidemiology, King’s College London, London, SE1 7EH, UK
| | - Celestine Lockhart
- Social Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, SE5 8AF, UK
| | | | - Sarah Markham
- UK LLC Public Involvement Programme, UK Longitudinal Linkage Collaboration, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, EH16 4UX, UK
| | - Dan Mason
- Bradford Institute for Health Research, Bradford Teaching Hospitals NHS Foundation Trust, Bradford, BD9 6RJ, UK
| | | | - Maisie McKenzie
- UK LLC Public Involvement Programme, UK Longitudinal Linkage Collaboration, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, EH16 4UX, UK
| | - Amy McMahon
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB2 0BB, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, CB2 0BB, UK
- National Institute for Health and Care Research Blood and Transplant Research Unit in Donor Health and Behaviour, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Chelsea Mika Malouf
- Social Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, SE5 8AF, UK
- UK National Institute for Health and Social Care Research (NIHR) Biomedical Research Centre for Mental Health, South London and Maudsley Hospital, London, SE5 8AF, UK
| | - Mark Mumme
- Avon Longitudinal Study of Parents and Children, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS8 2BN, UK
| | - Charlotte Neville
- Centre for Public Health, Queens University Belfast, Belfast, BT12 6BA, UK
| | - Kate Northstone
- Avon Longitudinal Study of Parents and Children, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS8 2BN, UK
| | - Zoe Oldfield
- Institute for Fiscal Studies, University College London, London, WC1E 7AE, UK
| | - Dara O’Neill
- Social Research Institute, University College London, London, WC1H 0AA, UK
| | - Manish Pareek
- Department of Respiratory Sciences, University of Leicester, Leicester, LE1 9HN, UK
| | - John Pickavance
- Bradford Institute for Health Research, Bradford Teaching Hospitals NHS Foundation Trust, Bradford, BD9 6RJ, UK
| | - Yasmin Rahman
- UK LLC Public Involvement Programme, UK Longitudinal Linkage Collaboration, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, EH16 4UX, UK
| | - Holly Reilly
- Department of Respiratory Sciences, University of Leicester, Leicester, LE1 9HN, UK
| | - Angela Scott
- Centre for Public Health, Queens University Belfast, Belfast, BT12 6BA, UK
| | - Deb Smith
- UK LLC Public Involvement Programme, UK Longitudinal Linkage Collaboration, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, EH16 4UX, UK
| | - Andrew Steptoe
- Department of Behavioural Science and Health, University College London, London, WC1E 6BT, UK
| | - Claire Steves
- Department of Twin Research & Genetic Epidemiology, King’s College London, London, SE1 7EH, UK
| | - Cathie Sudlow
- Generation Scotland, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Gerald Sze
- Population Health Sciences, University of Leicester, Leicester, LE1 7RH, UK
| | - Nicholas L. Timpson
- Avon Longitudinal Study of Parents and Children, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS8 2BN, UK
| | - Tapiwa Tungamirai
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, CB10 1SA, UK
| | - Laura Venn
- Population Health Sciences, University of Leicester, Leicester, LE1 7RH, UK
| | - Matthew Walker
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB2 0BB, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, CB2 0BB, UK
- National Institute for Health and Care Research Blood and Transplant Research Unit in Donor Health and Behaviour, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Neil Walker
- NIHR BioResource, Cambridge University Hospitals NHS Foundation Trust, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK
| | - Nicolas Wareham
- Understanding Society, University of Essex, Colchester, CO4 3SQ, UK
| | - Aidan Watmuff
- Bradford Institute for Health Research, Bradford Teaching Hospitals NHS Foundation Trust, Bradford, BD9 6RJ, UK
| | - Tony Webb
- MRC Epidemiology Unit, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Karen Williams
- UK LLC Public Involvement Programme, UK Longitudinal Linkage Collaboration, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, EH16 4UX, UK
| | - John Wright
- Bradford Institute for Health Research, Bradford Teaching Hospitals NHS Foundation Trust, Bradford, BD9 6RJ, UK
| | - Darioush Yarand
- Department of Twin Research & Genetic Epidemiology, King’s College London, London, SE1 7EH, UK
| | - George B Ploubidis
- Centre for Longitudinal Studies, University College London, London, WC1H 0AL, UK
| | - John Macleod
- UK Longitudinal Linkage Collaboration, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS8 2PS, UK
| | - Jonathan AC. Sterne
- UK Longitudinal Linkage Collaboration, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS8 2PS, UK
| | - Nishi Chaturvedi
- MRC Unit of Lifelong Health & Ageing, University College London, London, WC1E 7HB, UK
| |
Collapse
|
3
|
Karavirta L, Aittokoski T, Pynnönen K, Rantalainen T, Westgate K, Gonzales T, Palmberg L, Neuvonen J, Lipponen JA, Turunen K, Nikander R, Portegijs E, Rantanen T, Brage S. Physical determinants of daily physical activity in older men and women. PLoS One 2025; 20:e0314456. [PMID: 39899499 PMCID: PMC11790164 DOI: 10.1371/journal.pone.0314456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 11/12/2024] [Indexed: 02/05/2025] Open
Abstract
INTRODUCTION The ability to perform bodily movement varies in ageing men and women. We investigated whether physical fitness may explain sex differences in daily physical activity energy expenditure (PAEE) among older people. METHODS In this cross-sectional study, a population-based cohort of 75, 80, and 85-year-old men and women (n = 409, 62% women) underwent laboratory-based assessment of walking speed, maximal knee extension strength, and body fat percentage. Free-living physical activity was assessed as total PAEE, and light (LPA) and moderate-to-vigorous physical activity (MVPA) using individually calibrated combined accelerometry and heart rate sensing. Path modelling was used to examine indirect associations between sex, physical fitness, and physical activity. RESULTS Men had better physical fitness and higher overall PAEE (mean 34.0 (SD 10.8) kJ/kg/day) than women (28.3 (8.4) kJ/kg/day, p<0.001). The path model for PAEE explained 33% of the variance. The direct association between sex and PAEE was non-significant, whereas the association between sex and PAEE mediated by body fat (β = 0.20, p<0.001) and walking speed (β = 0.05, p = 0.001) were statistically significant. Similarly, associations between sex and MVPA mediated by body fat (β = 0.11, p = 0.002) and walking speed (β = 0.05, p = 0.001) were significant, as were the associations between sex and LPA mediated by body fat (β = 0.24, p<0.001) and walking speed (β = 0.03, p = 0.019). CONCLUSION Differences in physical activity between men and women may reflect underlying differences in cardiorespiratory fitness and adiposity. These results highlight the importance of maintaining physical fitness to support active living in older adults.
Collapse
Affiliation(s)
- Laura Karavirta
- Faculty of Sport and Health Sciences and Gerontology Research Center, University of Jyväskylä, Jyväskylä, Finland
| | - Timo Aittokoski
- Faculty of Sport and Health Sciences and Gerontology Research Center, University of Jyväskylä, Jyväskylä, Finland
| | - Katja Pynnönen
- Faculty of Sport and Health Sciences and Gerontology Research Center, University of Jyväskylä, Jyväskylä, Finland
| | - Timo Rantalainen
- Faculty of Sport and Health Sciences and Gerontology Research Center, University of Jyväskylä, Jyväskylä, Finland
| | - Kate Westgate
- MRC Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Tomas Gonzales
- MRC Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Lotta Palmberg
- Faculty of Sport and Health Sciences and Gerontology Research Center, University of Jyväskylä, Jyväskylä, Finland
| | - Joona Neuvonen
- Faculty of Sport and Health Sciences and Gerontology Research Center, University of Jyväskylä, Jyväskylä, Finland
| | - Jukka A. Lipponen
- Department of Technical Physics, University of Eastern Finland, Kuopio, Finland
- Department of Emergency Care, Kuopio University Hospital, Kuopio, Finland
| | - Katri Turunen
- Faculty of Sport and Health Sciences and Gerontology Research Center, University of Jyväskylä, Jyväskylä, Finland
- Seinäjoki University of Applied Sciences, Seinäjoki, Finland
| | - Riku Nikander
- Faculty of Sport and Health Sciences and Gerontology Research Center, University of Jyväskylä, Jyväskylä, Finland
- Wellbeing Region of Central Finland/Wellbeing Services County of Central Finland, Finland
| | - Erja Portegijs
- Faculty of Sport and Health Sciences and Gerontology Research Center, University of Jyväskylä, Jyväskylä, Finland
- University Medical Center Groningen, Center for Human Movement Sciences, University of Groningen, Groningen, the Netherlands
| | - Taina Rantanen
- Faculty of Sport and Health Sciences and Gerontology Research Center, University of Jyväskylä, Jyväskylä, Finland
| | - Søren Brage
- MRC Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
4
|
Dib MJ, Azzo JD, Zhao L, Salman O, Gan S, De Buyzere ML, De Meyer T, Ebert C, Gunawardhana K, Liu L, Gordon D, Seiffert D, Ching-Pin C, Zamani P, Cohen JB, Pourmussa B, Kun S, Gill D, Burgess S, van Empel V, Richards AM, Dennis J, Javaheri A, Mann DL, Cappola TP, Rietzschel E, Chirinos JA. Proteome-Wide Genetic Investigation of Large Artery Stiffness. JACC Basic Transl Sci 2024; 9:1178-1191. [PMID: 39534640 PMCID: PMC11551872 DOI: 10.1016/j.jacbts.2024.05.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 05/28/2024] [Accepted: 05/28/2024] [Indexed: 11/16/2024]
Abstract
The molecular mechanisms contributing to large artery stiffness (LAS) are not fully understood. The aim of this study was to investigate the association between circulating plasma proteins and LAS using complementary proteomic and genomic analyses. A total of 106 proteins associated with carotid-femoral pulse-wave velocity, a noninvasive measure of LAS, were identified in 1,178 individuals from the Asklepios study cohort. Mendelian randomization analyses revealed causal effects of 13 genetically predicted plasma proteins on pulse pressure, including cartilage intermediate layer protein-2, high-temperature requirement A serine peptidase-1, and neuronal growth factor-1. These findings suggest potential novel therapeutic targets to reduce LAS and its related diseases.
Collapse
Affiliation(s)
- Marie-Joe Dib
- Division of Cardiovascular Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Joe David Azzo
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Lei Zhao
- Bristol Myers Squibb, Lawrenceville, New Jersey, USA
| | - Oday Salman
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Sushrima Gan
- Division of Cardiovascular Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Marc L. De Buyzere
- Department of Cardiovascular Diseases, Ghent University Hospital, Ghent, Belgium
| | - Tim De Meyer
- Department of Mathematical Modelling, Statistics and Bioinformatics, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | | | | | - Laura Liu
- Bristol Myers Squibb, Lawrenceville, New Jersey, USA
| | - David Gordon
- Bristol Myers Squibb, Lawrenceville, New Jersey, USA
| | | | | | - Payman Zamani
- Division of Cardiovascular Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jordana B. Cohen
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Renal-Electrolyte and Hypertension Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Bianca Pourmussa
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Seavmeiyin Kun
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Dipender Gill
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, United Kingdom
| | - Stephen Burgess
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Vanessa van Empel
- Department of Cardiology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - A. Mark Richards
- Cardiovascular Research Institute, National University of Singapore, Singapore, Singapore
- Christchurch Heart Institute, University of Otago, Christchurch, New Zealand
| | | | - Ali Javaheri
- Washington University School of Medicine, St. Louis, Missouri, USA
- John J. Cochran Veterans Hospital, St. Louis, Missouri, USA
| | - Douglas L. Mann
- Washington University School of Medicine, St. Louis, Missouri, USA
| | - Thomas P. Cappola
- Division of Cardiovascular Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ernst Rietzschel
- Department of Cardiovascular Diseases, Ghent University Hospital, Ghent, Belgium
| | - Julio A. Chirinos
- Division of Cardiovascular Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
5
|
Carrasco-Zanini J, Wheeler E, Uluvar B, Kerrison N, Koprulu M, Wareham NJ, Pietzner M, Langenberg C. Mapping biological influences on the human plasma proteome beyond the genome. Nat Metab 2024; 6:2010-2023. [PMID: 39327534 PMCID: PMC11496106 DOI: 10.1038/s42255-024-01133-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 08/23/2024] [Indexed: 09/28/2024]
Abstract
Broad-capture proteomic platforms now enable simultaneous assessment of thousands of plasma proteins, but most of these are not actively secreted and their origins are largely unknown. Here we integrate genomic with deep phenomic information to identify modifiable and non-modifiable factors associated with 4,775 plasma proteins in ~8,000 mostly healthy individuals. We create a data-driven map of biological influences on the human plasma proteome and demonstrate segregation of proteins into clusters based on major explanatory factors. For over a third (N = 1,575) of protein targets, joint genetic and non-genetic factors explain 10-77% of the variation in plasma (median 19.88%, interquartile range 14.01-31.09%), independent of technical factors (median 2.48%, interquartile range 0.78-6.41%). Together with genetically anchored causal inference methods, our map highlights potential causal associations between modifiable risk factors and plasma proteins for hundreds of protein-disease associations, for example, COL6A3, which possibly mediates the association between reduced kidney function and cardiovascular disease. We provide a map of biological and technical influences on the human plasma proteome to help contextualize findings from proteomic studies.
Collapse
Affiliation(s)
- Julia Carrasco-Zanini
- Precision Healthcare University Research Institute, Queen Mary University of London, London, UK
- Computational Medicine, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge, UK
| | - Eleanor Wheeler
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge, UK
| | - Burulça Uluvar
- Computational Medicine, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Nicola Kerrison
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge, UK
| | - Mine Koprulu
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge, UK
| | - Nicholas J Wareham
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge, UK
| | - Maik Pietzner
- Precision Healthcare University Research Institute, Queen Mary University of London, London, UK
- Computational Medicine, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge, UK
| | - Claudia Langenberg
- Precision Healthcare University Research Institute, Queen Mary University of London, London, UK.
- Computational Medicine, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany.
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge, UK.
| |
Collapse
|
6
|
Lockhart SM, Muso M, Zvetkova I, Lam BYH, Ferrari A, Schoenmakers E, Duckett K, Leslie J, Collins A, Romartínez-Alonso B, Tadross JA, Jia R, Gardner EJ, Kentistou K, Zhao Y, Day F, Mörseburg A, Rainbow K, Rimmington D, Mastantuoni M, Harrison J, Nus M, Guma'a K, Sherratt-Mayhew S, Jiang X, Smith KR, Paul DS, Jenkins B, Koulman A, Pietzner M, Langenberg C, Wareham N, Yeo GS, Chatterjee K, Schwabe J, Oakley F, Mann DA, Tontonoz P, Coll AP, Ong K, Perry JRB, O'Rahilly S. Damaging mutations in liver X receptor-α are hepatotoxic and implicate cholesterol sensing in liver health. Nat Metab 2024; 6:1922-1938. [PMID: 39322746 PMCID: PMC11496107 DOI: 10.1038/s42255-024-01126-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 08/05/2024] [Indexed: 09/27/2024]
Abstract
Liver X receptor-α (LXRα) regulates cellular cholesterol abundance and potently activates hepatic lipogenesis. Here we show that at least 1 in 450 people in the UK Biobank carry functionally impaired mutations in LXRα, which is associated with biochemical evidence of hepatic dysfunction. On a western diet, male and female mice homozygous for a dominant negative mutation in LXRα have elevated liver cholesterol, diffuse cholesterol crystal accumulation and develop severe hepatitis and fibrosis, despite reduced liver triglyceride and no steatosis. This phenotype does not occur on low-cholesterol diets and can be prevented by hepatocyte-specific overexpression of LXRα. LXRα knockout mice exhibit a milder phenotype with regional variation in cholesterol crystal deposition and inflammation inversely correlating with steatosis. In summary, LXRα is necessary for the maintenance of hepatocyte health, likely due to regulation of cellular cholesterol content. The inverse association between steatosis and both inflammation and cholesterol crystallization may represent a protective action of hepatic lipogenesis in the context of excess hepatic cholesterol.
Collapse
Affiliation(s)
- Sam M Lockhart
- Medical Research Council (MRC) Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
| | - Milan Muso
- Medical Research Council (MRC) Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
| | - Ilona Zvetkova
- Medical Research Council (MRC) Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Brian Y H Lam
- Medical Research Council (MRC) Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Alessandra Ferrari
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA, USA
| | - Erik Schoenmakers
- Medical Research Council (MRC) Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Katie Duckett
- Medical Research Council (MRC) Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Jack Leslie
- Newcastle Fibrosis Research Group, Bioscience Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Amy Collins
- Newcastle Fibrosis Research Group, Bioscience Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Beatriz Romartínez-Alonso
- Institute of Structural and Chemical Biology, Department of Molecular and Cell Biology, University of Leicester, Leicester, UK
| | - John A Tadross
- Medical Research Council (MRC) Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Department of Histopathology and Cambridge Genomics Laboratory, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Raina Jia
- Medical Research Council (MRC) Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Eugene J Gardner
- Medical Research Council (MRC) Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Katherine Kentistou
- Medical Research Council (MRC) Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Yajie Zhao
- Medical Research Council (MRC) Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Felix Day
- Medical Research Council (MRC) Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Alexander Mörseburg
- Medical Research Council (MRC) Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Medical Research Council (MRC) Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Kara Rainbow
- Medical Research Council (MRC) Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Debra Rimmington
- Medical Research Council (MRC) Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Matteo Mastantuoni
- Medical Research Council (MRC) Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - James Harrison
- VPD Heart and Lung Research Institute, Dept. Medicine, University of Cambridge, Cambridge, UK
| | - Meritxell Nus
- VPD Heart and Lung Research Institute, Dept. Medicine, University of Cambridge, Cambridge, UK
| | - Khalid Guma'a
- Medical Research Council (MRC) Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Sam Sherratt-Mayhew
- Medical Research Council (MRC) Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Xiao Jiang
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Katherine R Smith
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Dirk S Paul
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Benjamin Jenkins
- Medical Research Council (MRC) Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- NIHR BRC Core Metabolomics and Lipidomics Laboratory, Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Albert Koulman
- Medical Research Council (MRC) Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- NIHR BRC Core Metabolomics and Lipidomics Laboratory, Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Maik Pietzner
- Medical Research Council (MRC) Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Computational Medicine, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- Precision Healthcare University Research Institute, Queen Mary University of London, London, UK
| | - Claudia Langenberg
- Medical Research Council (MRC) Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Computational Medicine, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- Precision Healthcare University Research Institute, Queen Mary University of London, London, UK
| | - Nicholas Wareham
- Medical Research Council (MRC) Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Giles S Yeo
- Medical Research Council (MRC) Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Krishna Chatterjee
- Medical Research Council (MRC) Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - John Schwabe
- Department of Histopathology and Cambridge Genomics Laboratory, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Fiona Oakley
- Newcastle Fibrosis Research Group, Bioscience Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Derek A Mann
- Newcastle Fibrosis Research Group, Bioscience Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Peter Tontonoz
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA, USA
| | - Anthony P Coll
- Medical Research Council (MRC) Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Ken Ong
- Medical Research Council (MRC) Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - John R B Perry
- Medical Research Council (MRC) Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Stephen O'Rahilly
- Medical Research Council (MRC) Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
- NIHR Cambridge Biomedical Research Centre, Cambridge, UK.
| |
Collapse
|
7
|
Perry AS, Farber-Eger E, Gonzales T, Tanaka T, Robbins JM, Murthy VL, Stolze LK, Zhao S, Huang S, Colangelo LA, Deng S, Hou L, Lloyd-Jones DM, Walker KA, Ferrucci L, Watts EL, Barber JL, Rao P, Mi MY, Gabriel KP, Hornikel B, Sidney S, Houstis N, Lewis GD, Liu GY, Thyagarajan B, Khan SS, Choi B, Washko G, Kalhan R, Wareham N, Bouchard C, Sarzynski MA, Gerszten RE, Brage S, Wells QS, Nayor M, Shah RV. Proteomic analysis of cardiorespiratory fitness for prediction of mortality and multisystem disease risks. Nat Med 2024; 30:1711-1721. [PMID: 38834850 PMCID: PMC11186767 DOI: 10.1038/s41591-024-03039-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 04/30/2024] [Indexed: 06/06/2024]
Abstract
Despite the wide effects of cardiorespiratory fitness (CRF) on metabolic, cardiovascular, pulmonary and neurological health, challenges in the feasibility and reproducibility of CRF measurements have impeded its use for clinical decision-making. Here we link proteomic profiles to CRF in 14,145 individuals across four international cohorts with diverse CRF ascertainment methods to establish, validate and characterize a proteomic CRF score. In a cohort of around 22,000 individuals in the UK Biobank, a proteomic CRF score was associated with a reduced risk of all-cause mortality (unadjusted hazard ratio 0.50 (95% confidence interval 0.48-0.52) per 1 s.d. increase). The proteomic CRF score was also associated with multisystem disease risk and provided risk reclassification and discrimination beyond clinical risk factors, as well as modulating high polygenic risk of certain diseases. Finally, we observed dynamicity of the proteomic CRF score in individuals who undertook a 20-week exercise training program and an association of the score with the degree of the effect of training on CRF, suggesting potential use of the score for personalization of exercise recommendations. These results indicate that population-based proteomics provides biologically relevant molecular readouts of CRF that are additive to genetic risk, potentially modifiable and clinically translatable.
Collapse
Affiliation(s)
- Andrew S Perry
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Eric Farber-Eger
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Tomas Gonzales
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Toshiko Tanaka
- Longtidudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Jeremy M Robbins
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | | | - Lindsey K Stolze
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Shilin Zhao
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Shi Huang
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Laura A Colangelo
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Shuliang Deng
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Lifang Hou
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Donald M Lloyd-Jones
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Keenan A Walker
- Multimodal Imaging of Neurodegenerative Disease (MIND) Unit, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Luigi Ferrucci
- Longtidudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Eleanor L Watts
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Jacob L Barber
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Prashant Rao
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Michael Y Mi
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Kelley Pettee Gabriel
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Bjoern Hornikel
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - Nicholas Houstis
- Cardiology Division, Massachusetts General Hospital, Boston, MA, USA
| | - Gregory D Lewis
- Cardiology Division, Massachusetts General Hospital, Boston, MA, USA
| | - Gabrielle Y Liu
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, University of California Davis, Sacramento, CA, USA
| | - Bharat Thyagarajan
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minnesota, MN, USA
| | - Sadiya S Khan
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Bina Choi
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - George Washko
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Ravi Kalhan
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Nick Wareham
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Claude Bouchard
- Human Genomic Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Mark A Sarzynski
- Department of Exercise Science, University of South Carolina Columbia, Columbia, SC, USA
| | - Robert E Gerszten
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Soren Brage
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Quinn S Wells
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Matthew Nayor
- Sections of Cardiovascular Medicine and Preventive Medicine and Epidemiology, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Ravi V Shah
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University School of Medicine, Nashville, TN, USA.
| |
Collapse
|
8
|
Yuan H, Chan S, Creagh AP, Tong C, Acquah A, Clifton DA, Doherty A. Self-supervised learning for human activity recognition using 700,000 person-days of wearable data. NPJ Digit Med 2024; 7:91. [PMID: 38609437 PMCID: PMC11015005 DOI: 10.1038/s41746-024-01062-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 02/22/2024] [Indexed: 04/14/2024] Open
Abstract
Accurate physical activity monitoring is essential to understand the impact of physical activity on one's physical health and overall well-being. However, advances in human activity recognition algorithms have been constrained by the limited availability of large labelled datasets. This study aims to leverage recent advances in self-supervised learning to exploit the large-scale UK Biobank accelerometer dataset-a 700,000 person-days unlabelled dataset-in order to build models with vastly improved generalisability and accuracy. Our resulting models consistently outperform strong baselines across eight benchmark datasets, with an F1 relative improvement of 2.5-130.9% (median 24.4%). More importantly, in contrast to previous reports, our results generalise across external datasets, cohorts, living environments, and sensor devices. Our open-sourced pre-trained models will be valuable in domains with limited labelled data or where good sampling coverage (across devices, populations, and activities) is hard to achieve.
Collapse
Affiliation(s)
- Hang Yuan
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
- Department of Computer Science, University of Oxford, Oxford, UK
| | - Shing Chan
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
| | - Andrew P Creagh
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
- Department of Engineering Science, University of Oxford, Oxford, UK
| | - Catherine Tong
- Department of Computer Science, University of Oxford, Oxford, UK
| | - Aidan Acquah
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
- Department of Engineering Science, University of Oxford, Oxford, UK
| | - David A Clifton
- Department of Engineering Science, University of Oxford, Oxford, UK
| | - Aiden Doherty
- Nuffield Department of Population Health, University of Oxford, Oxford, UK.
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK.
| |
Collapse
|
9
|
Vicente-Gabriel S, Lugones-Sánchez C, Tamayo-Morales O, Vicente Prieto A, González-Sánchez S, Conde Martín S, Gómez-Sánchez M, Rodríguez-Sánchez E, García-Ortiz L, Gómez-Sánchez L, Gómez-Marcos MA, EVA-Adic Investigators Group. Relationship between addictions and obesity, physical activity and vascular aging in young adults (EVA-Adic study): a research protocol of a cross-sectional study. Front Public Health 2024; 12:1322437. [PMID: 38344236 PMCID: PMC10853417 DOI: 10.3389/fpubh.2024.1322437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 01/08/2024] [Indexed: 02/15/2024] Open
Abstract
Background Behavioral and substance addictions are prevalent health problems that, alongside obesity, are linked to reduced physical activity and increased sedentary time. Similarly, arterial stiffness and vascular aging are processes that begin gradually at an early age and are closely associated with morbidity and mortality from cardiovascular diseases. The main objective of this study is to analyze how addictions are related to obesity and body fat distribution, physical activity, sedentary time, arterial stiffness and vascular aging, as well as sleep quality, cognitive function and gender differences in young adults aged between 18 and 34 years. Methods This cross-sectional descriptive observational study will analyze data from 500 subjects (250 men and 250 women) aged 18-34 without cardiovascular disease, selected by simple random sampling with replacement from the urban population of the city center of Salamanca (34,044 people aged 18-34, with 18,450 women and 15,594 men). Behavioral and substance addictions, as well as sleep quality and cognitive impairment will be assessed using questionnaires. The Pittisburg Sleep Quality Index (PSQI) will be used to measure sleep quality and the Ford questionnaire will be used to measure insomnia in response to stress. For obesity, weight, height, waist and hip circumference, body composition will be measured with the Inbody 230® impedance meter. For physical activity and sedentary time, we will use the Actigraph® accelerometer alongside the international physical activity questionnaire (IPAQ) and the Marshall questionnaire. The Sphygmocor System® will be used for pulse wave analysis and carotid-femoral pulse wave velocity (cfPWV), while the Vasera VS-2000® will measure cardio ankle vascular index (CAVI) and brachial-ankle pulse wave velocity (baPWV). Vascular aging will be calculated with the 10th and 90th percentiles of cfPWV or baPWV. Demographic, analytical variables will be collected, as will data to assess vascular, cardiac, renal, and brain injury. Discussion Addictions are on the rise in today's society, affecting the mental health and well-being of those who suffer from them, generating important social problems such as job loss, family dysfunction, debt and social isolation. Together with obesity, they are prevalent health problems in young adults and are associated with lower physical activity and higher sedentary time. Meanwhile, arterial stiffness and vascular aging are processes that begin gradually at an early age and determine morbidity and mortality caused by cardiovascular diseases. The results of this project will allow us to understand the situation regarding behavioral and substance addictions in young adults. Better understanding of these addictions will in turn facilitate the development of more effective prevention strategies and intervention programs, which can then reduce the negative impact at both the individual and societal levels. Clinical trial registration [ClinicalTrials.gov], identifier [NCT05819840].
Collapse
Affiliation(s)
- Sara Vicente-Gabriel
- Primary Care Research Unit of Salamanca (APISAL), Salamanca Primary Care Management, Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Intensive Care Unit, Salamanca University Hospital, Salamanca, Spain
| | - Cristina Lugones-Sánchez
- Primary Care Research Unit of Salamanca (APISAL), Salamanca Primary Care Management, Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Research Network on Chronicity, Primary Care and Health Promotion (RICAPPS), Barcelona, Spain
- Salamanca Primary Care Management, Castilla and León Health Service–SACYL, Salamanca, Spain
| | - Olaya Tamayo-Morales
- Primary Care Research Unit of Salamanca (APISAL), Salamanca Primary Care Management, Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Research Network on Chronicity, Primary Care and Health Promotion (RICAPPS), Barcelona, Spain
| | - Alberto Vicente Prieto
- Primary Care Research Unit of Salamanca (APISAL), Salamanca Primary Care Management, Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Salamanca Primary Care Management, Castilla and León Health Service–SACYL, Salamanca, Spain
| | - Susana González-Sánchez
- Primary Care Research Unit of Salamanca (APISAL), Salamanca Primary Care Management, Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Research Network on Chronicity, Primary Care and Health Promotion (RICAPPS), Barcelona, Spain
| | - Sandra Conde Martín
- Primary Care Research Unit of Salamanca (APISAL), Salamanca Primary Care Management, Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Salamanca Primary Care Management, Castilla and León Health Service–SACYL, Salamanca, Spain
| | - Marta Gómez-Sánchez
- Primary Care Research Unit of Salamanca (APISAL), Salamanca Primary Care Management, Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Home Hospitalization Unit, Marqués de Valdecilla University Hospital, Santander, Spain
| | - Emiliano Rodríguez-Sánchez
- Primary Care Research Unit of Salamanca (APISAL), Salamanca Primary Care Management, Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Research Network on Chronicity, Primary Care and Health Promotion (RICAPPS), Barcelona, Spain
- Salamanca Primary Care Management, Castilla and León Health Service–SACYL, Salamanca, Spain
- Department of Medicine, University of Salamanca, Salamanca, Spain
| | - Luis García-Ortiz
- Primary Care Research Unit of Salamanca (APISAL), Salamanca Primary Care Management, Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Research Network on Chronicity, Primary Care and Health Promotion (RICAPPS), Barcelona, Spain
- Salamanca Primary Care Management, Castilla and León Health Service–SACYL, Salamanca, Spain
- Department of Biomedical and Diagnostic Sciences, University of Salamanca, Salamanca, Spain
| | - Leticia Gómez-Sánchez
- Primary Care Research Unit of Salamanca (APISAL), Salamanca Primary Care Management, Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Emergency Department, La Paz University Hospital, Madrid, Spain
| | - Manuel A. Gómez-Marcos
- Primary Care Research Unit of Salamanca (APISAL), Salamanca Primary Care Management, Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Research Network on Chronicity, Primary Care and Health Promotion (RICAPPS), Barcelona, Spain
- Salamanca Primary Care Management, Castilla and León Health Service–SACYL, Salamanca, Spain
- Department of Medicine, University of Salamanca, Salamanca, Spain
| | | |
Collapse
|
10
|
Mueller J, Ahern AL, Jones RA, Sharp SJ, Davies A, Zuckerman A, Perry BI, Khandaker GM, Rolfe EDL, Wareham NJ, Rennie KL. The relationship of within-individual and between-individual variation in mental health with bodyweight: An exploratory longitudinal study. PLoS One 2024; 19:e0295117. [PMID: 38198439 PMCID: PMC10781195 DOI: 10.1371/journal.pone.0295117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 11/15/2023] [Indexed: 01/12/2024] Open
Abstract
BACKGROUND Poor mental health is associated with obesity, but existing studies are either cross-sectional or have long time periods between measurements of mental health and weight. It is, therefore, unclear how small fluctuations in mental wellbeing within individuals predict bodyweight over short time periods, e.g. within the next month. Studying this could identify modifiable determinants of weight changes and highlight opportunities for early intervention. METHODS 2,133 UK adults from a population-based cohort completed monthly mental health and weight measurements using a mobile app over a period of 6-9 months. We used random intercept regression models to examine longitudinal associations of depressive symptoms, anxiety symptoms and stress with subsequent weight. In sub-group analyses, we included interaction terms of mental health variables with baseline characteristics. Mental health variables were split into "between-individual" measurements (= the participant's median score across all timepoints) and "within-individual" measurements (at each timepoint, the difference between the participant's current score and their median). RESULTS Within-individual variation in depressive symptoms predicted subsequent weight (0.045kg per unit of depressive symptom severity, 95% CI 0.021-0.069). We found evidence of a moderation effect of baseline BMI on the association between within-individual fluctuation in depressive symptoms and subsequent weight: The association was only apparent in those with overweight/obesity, and it was stronger in those with obesity than those with overweight (BMI<25kg/m2: 0.011kg per unit of depressive symptom severity [95% CI -0.017 to 0.039]; BMI 25-29.9kg/m2: 0.052kg per unit of depressive symptom severity [95%CI 0.010-0.094kg]; BMI≥30kg/m2: 0.071kg per unit of depressive symptom severity [95%CI 0.013-0.129kg]). We found no evidence for other interactions, associations of stress and anxiety with weight, or for a reverse direction of association. CONCLUSION In this exploratory study, individuals with overweight or obesity were more vulnerable to weight gain following higher-than-usual (for that individual) depressive symptoms than individuals with a BMI<25kg/m2.
Collapse
Affiliation(s)
- Julia Mueller
- MRC Epidemiology Unit, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Amy L. Ahern
- MRC Epidemiology Unit, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Rebecca A. Jones
- MRC Epidemiology Unit, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Stephen J. Sharp
- MRC Epidemiology Unit, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Alan Davies
- Division of Informatics, Imaging & Data Sciences, School of Health Sciences, University of Manchester, Manchester, United Kingdom
| | - Arabella Zuckerman
- School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Benjamin I. Perry
- Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
- Cambridgeshire and Peterborough NHS Foundation Trust, Cambridge, United Kingdom
| | - Golam M. Khandaker
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- NIHR Bristol Biomedical Research Centre, Bristol, United Kingdom
| | - Emanuella De Lucia Rolfe
- MRC Epidemiology Unit, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Nick J. Wareham
- MRC Epidemiology Unit, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Kirsten L. Rennie
- MRC Epidemiology Unit, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
11
|
Trichia E, Koulman A, Stewart ID, Brage S, Griffin SJ, Griffin JL, Khaw K, Langenberg C, Wareham NJ, Imamura F, Forouhi NG. Plasma Metabolites Related to the Consumption of Different Types of Dairy Products and Their Association with New-Onset Type 2 Diabetes: Analyses in the Fenland and EPIC-Norfolk Studies, United Kingdom. Mol Nutr Food Res 2024; 68:e2300154. [PMID: 38054622 PMCID: PMC10909549 DOI: 10.1002/mnfr.202300154] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 07/07/2023] [Indexed: 12/07/2023]
Abstract
SCOPE To identify metabolites associated with habitual dairy consumption and investigate their associations with type 2 diabetes (T2D) risk. METHODS AND RESULTS Metabolomics assays were conducted in the Fenland (n = 10,281) and EPIC-Norfolk (n = 1,440) studies. Using 82 metabolites assessed in both studies, we developed metabolite scores to classify self-reported consumption of milk, yogurt, cheese, butter, and total dairy (Fenland Study-discovery set; n = 6035). Internal and external validity of the scores was evaluated (Fenland-validation set, n = 4246; EPIC-Norfolk, n = 1440). The study assessed associations between each metabolite score and T2D incidence in EPIC-Norfolk (n = 641 cases; 16,350 person-years). The scores classified low and high consumers for all dairy types with internal validity, and milk, butter, and total dairy with external validity. The scores were further associated with lower incident T2D: hazard ratios (95% confidence interval) per standard deviation: milk 0.71 (0.65, 0.77); butter 0.62 (0.57, 0.68); total dairy 0.66 (0.60, 0.72). These associations persisted after adjustment for known dairy-fat biomarkers. CONCLUSION Metabolite scores identified habitual consumers of milk, butter, and total dairy products, and were associated with lower T2D risk. These findings hold promise for identifying objective indicators of the physiological response to dairy consumption.
Collapse
Affiliation(s)
- Eirini Trichia
- MRC Epidemiology UnitInstitute of Metabolic ScienceUniversity of Cambridge School of Clinical MedicineCambridgeCB2 0SLUK
| | - Albert Koulman
- MRC Epidemiology UnitInstitute of Metabolic ScienceUniversity of Cambridge School of Clinical MedicineCambridgeCB2 0SLUK
| | - Isobel D. Stewart
- MRC Epidemiology UnitInstitute of Metabolic ScienceUniversity of Cambridge School of Clinical MedicineCambridgeCB2 0SLUK
| | - Soren Brage
- MRC Epidemiology UnitInstitute of Metabolic ScienceUniversity of Cambridge School of Clinical MedicineCambridgeCB2 0SLUK
| | - Simon J. Griffin
- MRC Epidemiology UnitInstitute of Metabolic ScienceUniversity of Cambridge School of Clinical MedicineCambridgeCB2 0SLUK
| | | | - Kay‐Tee Khaw
- MRC Epidemiology UnitInstitute of Metabolic ScienceUniversity of Cambridge School of Clinical MedicineCambridgeCB2 0SLUK
| | - Claudia Langenberg
- MRC Epidemiology UnitInstitute of Metabolic ScienceUniversity of Cambridge School of Clinical MedicineCambridgeCB2 0SLUK
| | - Nicholas J. Wareham
- MRC Epidemiology UnitInstitute of Metabolic ScienceUniversity of Cambridge School of Clinical MedicineCambridgeCB2 0SLUK
| | - Fumiaki Imamura
- MRC Epidemiology UnitInstitute of Metabolic ScienceUniversity of Cambridge School of Clinical MedicineCambridgeCB2 0SLUK
| | - Nita G. Forouhi
- MRC Epidemiology UnitInstitute of Metabolic ScienceUniversity of Cambridge School of Clinical MedicineCambridgeCB2 0SLUK
| |
Collapse
|
12
|
Zhao J, Zeng J, Zhu C, Li X, Liu D, Zhang J, Li F, Targher G, Fan JG. Genetically predicted plasma levels of amino acids and metabolic dysfunction-associated fatty liver disease risk: a Mendelian randomization study. BMC Med 2023; 21:469. [PMID: 38017422 PMCID: PMC10685523 DOI: 10.1186/s12916-023-03185-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 11/21/2023] [Indexed: 11/30/2023] Open
Abstract
BACKGROUND Emerging metabolomics-based studies suggested links between amino acid metabolism and metabolic dysfunction-associated fatty liver disease (MAFLD) risk; however, whether there exists an aetiological role of amino acid metabolism in MAFLD development remains unknown. The aim of the present study was to assess the causal relationship between circulating levels of amino acids and MAFLD risk. METHODS We conducted a two-sample Mendelian randomization (MR) analysis using summary-level data from genome-wide association studies (GWAS) to evaluate the causal relationship between genetically predicted circulating levels of amino acids and the risk of MAFLD. In the discovery MR analysis, we used data from the largest MAFLD GWAS (8434 cases and 770,180 controls), while in the replication MR analysis, we used data from a GWAS on MAFLD (1483 cases and 17,781 controls) where MAFLD cases were diagnosed using liver biopsy. We used Wald ratios or inverse variance-weighted (IVW) methods in the MR main analysis and weighted median and MR-Egger regression analyses in sensitivity analyses. Furthermore, we performed a conservative MR analysis by restricting genetic instruments to those directly involved in amino acid metabolism pathways. RESULTS We found that genetically predicted higher alanine (OR = 1.43, 95% CI 1.13-1.81) and lower glutamine (OR = 0.83, 95% CI 0.73-0.96) levels were associated with a higher risk of developing MAFLD based on the results from the MR main and conservative analysis. The results from MR sensitivity analyses and complementary analysis using liver proton density fat fraction as a continuous outcome proxying for MAFLD supported the main findings. CONCLUSIONS Novel causal metabolites related to MAFLD development were uncovered through MR analysis, suggesting future potential for evaluating these metabolites as targets for MAFLD prevention or treatment.
Collapse
Affiliation(s)
- Jian Zhao
- The Ministry of Education and Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No. 1665, Kongjiang Road, Yangpu District, Shanghai, 200092, China.
- Department of Maternal and Child Health, School of Public Health, Shanghai Jiao Tong University, Shanghai, China.
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK.
| | - Jing Zeng
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No. 1665, Kongjiang Road, Yangpu District, Shanghai, 200092, China
| | - Cairong Zhu
- Department of Epidemiology and Biostatistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Xuechao Li
- Department of Epidemiology and Biostatistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Dong Liu
- The Ministry of Education and Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No. 1665, Kongjiang Road, Yangpu District, Shanghai, 200092, China
| | - Jun Zhang
- The Ministry of Education and Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No. 1665, Kongjiang Road, Yangpu District, Shanghai, 200092, China
- Department of Maternal and Child Health, School of Public Health, Shanghai Jiao Tong University, Shanghai, China
| | - Fei Li
- The Ministry of Education and Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No. 1665, Kongjiang Road, Yangpu District, Shanghai, 200092, China
- Department of Maternal and Child Health, School of Public Health, Shanghai Jiao Tong University, Shanghai, China
- Department of Developmental and Behavioral Pediatric & Child Primary Care, Brain and Behavioral Research Unit of Shanghai Institute for Pediatric Research, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Giovanni Targher
- Department of Medicine, University of Verona, Verona, Italy
- Metabolic Diseases Research Unit, IRCCS Ospedale Sacro Cuore - Don Calabria, Negrar di Valpolicella, Italy
| | - Jian-Gao Fan
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No. 1665, Kongjiang Road, Yangpu District, Shanghai, 200092, China.
- Shanghai Key Lab of Pediatric Gastroenterology and Nutrition, Shanghai, China.
| |
Collapse
|
13
|
Cheng TS, Brage S, van Sluijs EMF, Ong KK. Pre-pubertal accelerometer-assessed physical activity and timing of puberty in British boys and girls: the Millennium Cohort Study. Int J Epidemiol 2023; 52:1316-1327. [PMID: 37208864 PMCID: PMC10555885 DOI: 10.1093/ije/dyad063] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/02/2023] [Indexed: 05/21/2023] Open
Abstract
BACKGROUND Early puberty timing is associated with adverse health outcomes. We aimed to examine prospective associations between objectively measured physical activity and puberty timing in boys and girls. METHODS In the UK Millennium Cohort Study, physical activity volume and intensities at 7 years were measured using accelerometers. Status of several pubertal traits and age at menarche were reported at 11, 14 and 17 years. Age at menarche in girls was categorized into tertiles. Other puberty traits were categorized into earlier or later than the median ages calculated from probit models, separately in boys and girls. Multivariable regression models, with adjustment for maternal and child characteristics including body mass index (BMI) at age 7 years as potential confounders, were performed to test the associations of total daily activity counts and fractions of activity counts across intensities (in compositional models) with puberty timing, separately in boys (n = 2531) and girls (n = 3079). RESULTS Higher total daily activity counts were associated with lower risks for earlier (vs later) growth spurt, body hair growth, skin changes and menarche in girls, and more weakly with lower risks for earlier skin changes and voice breaking in boys (odds ratios = 0.80-0.87 per 100 000 counts/day). These associations persisted on additional adjustment for BMI at 11 years as a potential mediator. No association with puberty timing was seen for any physical activity intensity (light, moderate or vigorous). CONCLUSIONS More physical activity regardless of intensity may contribute to the avoidance of earlier puberty timing, independently of BMI, particularly in girls.
Collapse
Affiliation(s)
- Tuck Seng Cheng
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Soren Brage
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Esther M F van Sluijs
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Ken K Ong
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| |
Collapse
|
14
|
Collings PJ, Backes A, Malisoux L. Arterial stiffness and the reallocation of time between device-measured 24-hour movement behaviours: A compositional data analysis. Atherosclerosis 2023; 379:117185. [PMID: 37531669 DOI: 10.1016/j.atherosclerosis.2023.117185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 07/03/2023] [Accepted: 07/04/2023] [Indexed: 08/04/2023]
Abstract
BACKGROUND AND AIMS Arterial stiffness predicts cardiovascular morbidity and mortality. We aimed to quantify the differences in arterial stiffness associated with reallocating time between 24-h movement behaviours. METHODS This observational cross-sectional study included Luxembourg residents aged 25-79y who each provided ≥4 valid days of triaxial accelerometry (n = 1001). Covariable adjusted compositional isotemporal substitution models were used to examine if theoretical reallocations of time between device-measured sedentariness, the sleep period, light physical activity (PA), and moderate-to-vigorous PA (MVPA) were associated with the percentage difference in carotid-femoral pulse wave velocity (cfPWV). We further investigated if replacing sedentary time accumulated in prolonged (≥30 min) with non-prolonged (<30 min) bouts was associated with arterial stiffness. The results are presented as 30 min time exchanges (β (95% confidence interval)). RESULTS Beneficial associations with lower cfPWV were observed when reallocating time to MVPA from the sleep period (-1.38 (-2.63 to -0.12) %), sedentary time (-1.70 (-2.76 to -0.62) %), and light PA (-2.51 (-4.55 to -0.43) %), respectively. Larger associations in the opposite direction were observed when reallocating MVPA to the same behaviours (for example, replacing MVPA with sedentary time: 2.50 (0.85-4.18) %). Replacing prolonged with non-prolonged sedentary time was not associated with cfPWV (-0.27 (-0.86 to 0.32) %). In short sleepers, reallocating sedentary time to the sleep period was favourable (-1.96 (-3.74 to -0.15) %). CONCLUSIONS Increasing or at least maintaining MVPA appears to be important for arterial health in adults. Extending sleep in habitually short sleepers, specifically by redistributing sedentary time, may also be important.
Collapse
Affiliation(s)
- Paul J Collings
- Physical Activity, Sport and Health Research Group, Department of Precision Health, Luxembourg Institute of Health, Luxembourg
| | - Anne Backes
- Physical Activity, Sport and Health Research Group, Department of Precision Health, Luxembourg Institute of Health, Luxembourg
| | - Laurent Malisoux
- Physical Activity, Sport and Health Research Group, Department of Precision Health, Luxembourg Institute of Health, Luxembourg.
| |
Collapse
|
15
|
Paudel S, Ahmadi M, Phongsavan P, Hamer M, Stamatakis E. Do associations of physical activity and sedentary behaviour with cardiovascular disease and mortality differ across socioeconomic groups? A prospective analysis of device-measured and self-reported UK Biobank data. Br J Sports Med 2023; 57:921-929. [PMID: 36754587 PMCID: PMC10359566 DOI: 10.1136/bjsports-2022-105435] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/24/2023] [Indexed: 02/10/2023]
Abstract
OBJECTIVE To examine if individual-level and area-level socioeconomic status (SES) modifies the association of moderate-to-vigorous physical activity (MVPA), domain-specific physical activity and sedentary behaviour with all-cause mortality (ACM) and incident cardiovascular disease (CVD). METHODS We used self-reported (International Physical Activity Questionnaire short form) and accelerometer-measured physical activity and sedentary behaviour data from the UK Biobank. We created an individual-level composite SES index using latent class analysis of household income, education and employment status. The Townsend Index was the measure of area-level SES. Cox proportional hazards regression models stratified across SES were used. RESULTS In 328 228 participants (mean age 55.9 (SD 8.1) years, 45% men) with an average follow-up of 12.1 (1.4) years, 18 033 deaths and 98 922 incident CVD events occurred. We found an increased ACM risk of low physical activity and high sedentary behaviour and an increased incident CVD risk of low accelerometer-measured moderate-to-vigorous physical activity (ACCEL_MVPA) and high sitting time. We observed statistically significant interactions for all exposures in ACM analyses by individual-level SES (p<0.05) but only for screen time in area-level SES-ACM analysis (p<0.001). Compared with high self-reported moderate-to-vigorous physical activity (IPAQ_MVPA), adjusted ACM HRs for low IPAQ_MVPA were 1.14 (95% CI 1.05 to .25), 1.15 (95% CI 1.06 to 1.24) and 1.22 (95% CI 1.13 to 1.31) in high, medium and low individual-level SES, respectively. There were higher detrimental associations of low ACCEL_MVPA with decreasing area-level SES for both outcomes and of high screen time with ACM in low area-level SES. CONCLUSION We found modest evidence suggesting that the detrimental associations of low MVPA and high screen time with ACM and incident CVD are accentuated in low SES groups.
Collapse
Affiliation(s)
- Susan Paudel
- Institute for Physical Activity and Nutrition (IPAN), Deakin University, Burwood, Victoria, Australia
- School of Health Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Matthew Ahmadi
- School of Health Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Philayrath Phongsavan
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- School of Public Health, Prevention Research Collaboration, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Mark Hamer
- Institute of Sport Exercise and Health, Division of Surgery and Interventional Science, University College London, London, UK
| | - Emmanuel Stamatakis
- School of Health Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
16
|
Dolezalova N, Gkrania-Klotsas E, Morelli D, Moore A, Cunningham AC, Booth A, Plans D, Reed AB, Aral M, Rennie KL, Wareham NJ. Feasibility of using intermittent active monitoring of vital signs by smartphone users to predict SARS-CoV-2 PCR positivity. Sci Rep 2023; 13:10581. [PMID: 37386099 PMCID: PMC10310739 DOI: 10.1038/s41598-023-37301-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 06/20/2023] [Indexed: 07/01/2023] Open
Abstract
Early detection of highly infectious respiratory diseases, such as COVID-19, can help curb their transmission. Consequently, there is demand for easy-to-use population-based screening tools, such as mobile health applications. Here, we describe a proof-of-concept development of a machine learning classifier for the prediction of a symptomatic respiratory disease, such as COVID-19, using smartphone-collected vital sign measurements. The Fenland App study followed 2199 UK participants that provided measurements of blood oxygen saturation, body temperature, and resting heart rate. Total of 77 positive and 6339 negative SARS-CoV-2 PCR tests were recorded. An optimal classifier to identify these positive cases was selected using an automated hyperparameter optimisation. The optimised model achieved an ROC AUC of 0.695 ± 0.045. The data collection window for determining each participant's vital sign baseline was increased from 4 to 8 or 12 weeks with no significant difference in model performance (F(2) = 0.80, p = 0.472). We demonstrate that 4 weeks of intermittently collected vital sign measurements could be used to predict SARS-CoV-2 PCR positivity, with applicability to other diseases causing similar vital sign changes. This is the first example of an accessible, smartphone-based remote monitoring tool deployable in a public health setting to screen for potential infections.
Collapse
Affiliation(s)
| | - Effrossyni Gkrania-Klotsas
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Department of Infectious Diseases, Addenbrooke's Hospital, Box 25, Cambridge, UK
| | - Davide Morelli
- Huma Therapeutics Ltd., Millbank Tower, 21-24 Millbank, London, UK
- Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Oxford, UK
| | - Alex Moore
- Huma Therapeutics Ltd., Millbank Tower, 21-24 Millbank, London, UK.
| | | | - Adam Booth
- Huma Therapeutics Ltd., Millbank Tower, 21-24 Millbank, London, UK
| | - David Plans
- Huma Therapeutics Ltd., Millbank Tower, 21-24 Millbank, London, UK
- Department of Experimental Psychology, University of Oxford, Oxford, UK
- INDEX Group, Department of Science, Innovation, Technology, and Entrepreneurship, University of Exeter, Exeter, UK
| | - Angus B Reed
- Huma Therapeutics Ltd., Millbank Tower, 21-24 Millbank, London, UK
| | - Mert Aral
- Huma Therapeutics Ltd., Millbank Tower, 21-24 Millbank, London, UK
| | - Kirsten L Rennie
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Nicholas J Wareham
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| |
Collapse
|
17
|
Williamson A, Norris DM, Yin X, Broadaway KA, Moxley AH, Vadlamudi S, Wilson EP, Jackson AU, Ahuja V, Andersen MK, Arzumanyan Z, Bonnycastle LL, Bornstein SR, Bretschneider MP, Buchanan TA, Chang YC, Chuang LM, Chung RH, Clausen TD, Damm P, Delgado GE, de Mello VD, Dupuis J, Dwivedi OP, Erdos MR, Fernandes Silva L, Frayling TM, Gieger C, Goodarzi MO, Guo X, Gustafsson S, Hakaste L, Hammar U, Hatem G, Herrmann S, Højlund K, Horn K, Hsueh WA, Hung YJ, Hwu CM, Jonsson A, Kårhus LL, Kleber ME, Kovacs P, Lakka TA, Lauzon M, Lee IT, Lindgren CM, Lindström J, Linneberg A, Liu CT, Luan J, Aly DM, Mathiesen E, Moissl AP, Morris AP, Narisu N, Perakakis N, Peters A, Prasad RB, Rodionov RN, Roll K, Rundsten CF, Sarnowski C, Savonen K, Scholz M, Sharma S, Stinson SE, Suleman S, Tan J, Taylor KD, Uusitupa M, Vistisen D, Witte DR, Walther R, Wu P, Xiang AH, Zethelius B, Ahlqvist E, Bergman RN, Chen YDI, Collins FS, Fall T, Florez JC, Fritsche A, Grallert H, Groop L, Hansen T, Koistinen HA, Komulainen P, Laakso M, Lind L, Loeffler M, März W, Meigs JB, Raffel LJ, Rauramaa R, Rotter JI, Schwarz PEH, Stumvoll M, et alWilliamson A, Norris DM, Yin X, Broadaway KA, Moxley AH, Vadlamudi S, Wilson EP, Jackson AU, Ahuja V, Andersen MK, Arzumanyan Z, Bonnycastle LL, Bornstein SR, Bretschneider MP, Buchanan TA, Chang YC, Chuang LM, Chung RH, Clausen TD, Damm P, Delgado GE, de Mello VD, Dupuis J, Dwivedi OP, Erdos MR, Fernandes Silva L, Frayling TM, Gieger C, Goodarzi MO, Guo X, Gustafsson S, Hakaste L, Hammar U, Hatem G, Herrmann S, Højlund K, Horn K, Hsueh WA, Hung YJ, Hwu CM, Jonsson A, Kårhus LL, Kleber ME, Kovacs P, Lakka TA, Lauzon M, Lee IT, Lindgren CM, Lindström J, Linneberg A, Liu CT, Luan J, Aly DM, Mathiesen E, Moissl AP, Morris AP, Narisu N, Perakakis N, Peters A, Prasad RB, Rodionov RN, Roll K, Rundsten CF, Sarnowski C, Savonen K, Scholz M, Sharma S, Stinson SE, Suleman S, Tan J, Taylor KD, Uusitupa M, Vistisen D, Witte DR, Walther R, Wu P, Xiang AH, Zethelius B, Ahlqvist E, Bergman RN, Chen YDI, Collins FS, Fall T, Florez JC, Fritsche A, Grallert H, Groop L, Hansen T, Koistinen HA, Komulainen P, Laakso M, Lind L, Loeffler M, März W, Meigs JB, Raffel LJ, Rauramaa R, Rotter JI, Schwarz PEH, Stumvoll M, Sundström J, Tönjes A, Tuomi T, Tuomilehto J, Wagner R, Barroso I, Walker M, Grarup N, Boehnke M, Wareham NJ, Mohlke KL, Wheeler E, O'Rahilly S, Fazakerley DJ, Langenberg C. Genome-wide association study and functional characterization identifies candidate genes for insulin-stimulated glucose uptake. Nat Genet 2023; 55:973-983. [PMID: 37291194 PMCID: PMC7614755 DOI: 10.1038/s41588-023-01408-9] [Show More Authors] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 04/26/2023] [Indexed: 06/10/2023]
Abstract
Distinct tissue-specific mechanisms mediate insulin action in fasting and postprandial states. Previous genetic studies have largely focused on insulin resistance in the fasting state, where hepatic insulin action dominates. Here we studied genetic variants influencing insulin levels measured 2 h after a glucose challenge in >55,000 participants from three ancestry groups. We identified ten new loci (P < 5 × 10-8) not previously associated with postchallenge insulin resistance, eight of which were shown to share their genetic architecture with type 2 diabetes in colocalization analyses. We investigated candidate genes at a subset of associated loci in cultured cells and identified nine candidate genes newly implicated in the expression or trafficking of GLUT4, the key glucose transporter in postprandial glucose uptake in muscle and fat. By focusing on postprandial insulin resistance, we highlighted the mechanisms of action at type 2 diabetes loci that are not adequately captured by studies of fasting glycemic traits.
Collapse
Affiliation(s)
- Alice Williamson
- MRC Epidemiology Unit Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
- Metabolic Research Laboratories Wellcome Trust-MRC Institute of Metabolic Science, Department of Clinical Biochemistry, University of Cambridge, Cambridge, UK
| | - Dougall M Norris
- Metabolic Research Laboratories Wellcome Trust-MRC Institute of Metabolic Science, Department of Clinical Biochemistry, University of Cambridge, Cambridge, UK
| | - Xianyong Yin
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
- Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - K Alaine Broadaway
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Anne H Moxley
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | | | - Emma P Wilson
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Anne U Jackson
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
- Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Vasudha Ahuja
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Mette K Andersen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Zorayr Arzumanyan
- Department of Pediatrics, Genomic Outcomes, The Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Lori L Bonnycastle
- Center for Precision Health Research National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Stefan R Bornstein
- Department of Internal Medicine III, Metabolic and Vascular Medicine, Medical Faculty Carl Gustav Carus, Dresden, Germany
- Helmholtz Zentrum München Paul Langerhans Institute Dresden (PLID), University Hospital and Faculty of Medicine TU Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Maxi P Bretschneider
- Department of Internal Medicine III, Metabolic and Vascular Medicine, Medical Faculty Carl Gustav Carus, Dresden, Germany
- Helmholtz Zentrum München Paul Langerhans Institute Dresden (PLID), University Hospital and Faculty of Medicine TU Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Thomas A Buchanan
- Department of Medicine, Division of Endocrinology and Diabetes, Keck School of Medicine USC, Los Angeles, CA, USA
| | - Yi-Cheng Chang
- Graduate Institute of Medical Genomics and Proteomics, National Taiwan University, Taipei City, Taiwan
- Internal Medicine, National Taiwan University Hospital, Taipei City, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Taipei City, Taiwan
| | - Lee-Ming Chuang
- Department of Internal Medicine, Division of Endocrinology and Metabolism, National Taiwan University Hospital, Taipei City, Taiwan
| | - Ren-Hua Chung
- Institute of Population Health Sciences, National Health Research Institutes, Toufen, Taiwan
| | - Tine D Clausen
- Department of Gynecology and Obstetrics, Nordsjaellands Hospital, Hillerød, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Peter Damm
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Center for Pregnant Women with Diabetes, Rigshospitalet, Copenhagen, Denmark
- Department of Obstetrics, Rigshospitalet, Copenhagen, Denmark
| | - Graciela E Delgado
- Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Vanessa D de Mello
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Josée Dupuis
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montréal, Quebec, Canada
| | - Om P Dwivedi
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Michael R Erdos
- Center for Precision Health Research National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | | | | | - Christian Gieger
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Mark O Goodarzi
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Xiuqing Guo
- Department of Pediatrics, Genomic Outcomes, The Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Stefan Gustafsson
- Department of Medical Sciences, Clinical Epidemiology, Uppsala University, Uppsala, Sweden
| | - Liisa Hakaste
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Ulf Hammar
- Department of Medical Sciences, Molecular Epidemiology, Uppsala University, Uppsala, Sweden
| | - Gad Hatem
- Clinical Sciences Malmö, Genomics, Diabetes and Endocrinology, Lund University, Malmö, Sweden
| | - Sandra Herrmann
- Helmholtz Zentrum München Paul Langerhans Institute Dresden (PLID), University Hospital and Faculty of Medicine TU Dresden, Dresden, Germany
- Department of Internal Medicine III, Prevention and Care of Diabetes, Medical Faculty Carl Gustav Carus, Dresden, Germany
| | - Kurt Højlund
- Steno Diabetes Center Odense, Odense University Hospital, Odense, Denmark
| | - Katrin Horn
- Medical Faculty Institute for Medical Informatics, Statistics and Epidemiology, Leipzig, Germany
- LIFE Leipzig Research Center for Civilization Diseases, Medical Faculty, Leipzig, Germany
| | - Willa A Hsueh
- Internal Medicine, Endocrinology, Diabetes and Metabolism, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Yi-Jen Hung
- Institute of Preventive Medicine, National Defense Medical Center, New Taipei City, Taiwan
| | - Chii-Min Hwu
- Department of Medicine Section of Endocrinology and Metabolism, Taipei Veterans General Hospital, Taipei City, Taiwan
| | - Anna Jonsson
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Line L Kårhus
- Center for Clinical Research and Prevention, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Marcus E Kleber
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
- SYNLAB MVZ Humangenetik Mannheim, Mannheim, Germany
| | - Peter Kovacs
- Medical Department III-Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany
| | - Timo A Lakka
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
- Department of Clinical Physiology and Nuclear Medicine, Kuopio University Hospital, Kuopio, Finland
- Foundation for Research in Health Exercise and Nutrition, Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
| | - Marie Lauzon
- Department of Pediatrics, Genomic Outcomes, The Institute for Translational Genomics and Population Sciences, The Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - I-Te Lee
- Department of Internal Medicine Division of Endocrinology and Metabolism, Taichung Veterans General Hospital, Taichung City, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei City, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung City, Taiwan
| | - Cecilia M Lindgren
- Big Data Institute Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Wellcome Trust Centre Human Genetics, University of Oxford, Oxford, UK
- Broad Institute, Cambridge, MA, USA
| | | | - Allan Linneberg
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Center for Clinical Research and Prevention, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Ching-Ti Liu
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Jian'an Luan
- MRC Epidemiology Unit Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Dina Mansour Aly
- Clinical Sciences Malmö, Genomics, Diabetes and Endocrinology, Lund University, Malmö, Sweden
| | - Elisabeth Mathiesen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Center for Pregnant Women with Diabetes, Rigshospitalet, Copenhagen, Denmark
- Department of Endocrinology Rigshospitalet, Copenhagen, Denmark
| | - Angela P Moissl
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
- Institute of Nutritional Sciences, Friedrich-Schiller-University, Jena, Germany
- Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD) Halle-Jena, Jena, Germany
| | - Andrew P Morris
- Centre for Genetics and Genomics Versus Arthritis Centre for Musculoskeletal Research, The University of Manchester, Manchester, UK
| | - Narisu Narisu
- Center for Precision Health Research National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Nikolaos Perakakis
- Department of Internal Medicine III, Metabolic and Vascular Medicine, Medical Faculty Carl Gustav Carus, Dresden, Germany
- Helmholtz Zentrum München Paul Langerhans Institute Dresden (PLID), University Hospital and Faculty of Medicine TU Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Annette Peters
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Rashmi B Prasad
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- Clinical Sciences Malmö, Genomics, Diabetes and Endocrinology, Lund University, Malmö, Sweden
| | - Roman N Rodionov
- Department of Internal Medicine III, University Center for Vascular Medicine, Medical Faculty Carl Gustav Carus, Dresden, Germany
- College of Medicine and Public Health, Flinders University and Flinders Medical Centre, Adelaide, Australia
| | - Kathryn Roll
- Pediatrics, Genomic Outcomes, The Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Carsten F Rundsten
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Chloé Sarnowski
- Department of Epidemiology, Human Genetics and Environmental Sciences, The University of Texas Health Science Center, Houston, TX, USA
| | - Kai Savonen
- Foundation for Research in Health Exercise and Nutrition, Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
| | - Markus Scholz
- Medical Faculty Institute for Medical Informatics, Statistics and Epidemiology, Leipzig, Germany
- LIFE Leipzig Research Center for Civilization Diseases, Medical Faculty, Leipzig, Germany
| | - Sapna Sharma
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
- Food Chemistry and Molecular and Sensory Science, Technical University of Munich, Freising-Weihenstephan, München, Germany
| | - Sara E Stinson
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sufyan Suleman
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jingyi Tan
- Department of Pediatrics, Genomic Outcomes, The Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Kent D Taylor
- Department of Pediatrics, Genomic Outcomes, The Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Matti Uusitupa
- Department of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Dorte Vistisen
- Clinical Research, Steno Diabetes Center Copenhagen, Herlev, Denmark
- Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Daniel R Witte
- Steno Diabetes Center Aarhus, Aarhus, Denmark
- Department of Public Health, Aarhus University, Aarhus, Denmark
| | - Romy Walther
- Helmholtz Zentrum München Paul Langerhans Institute Dresden (PLID), University Hospital and Faculty of Medicine TU Dresden, Dresden, Germany
- Department of Internal Medicine III, Pathobiochemistry, Medical Faculty Carl Gustav Carus, Dresden, Germany
| | - Peitao Wu
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Anny H Xiang
- Research and Evaluation, Division of Biostatistics, Kaiser Permanente Southern California, Pasadena, CA, USA
| | - Björn Zethelius
- Department of Public Health and Caring Sciences, Geriatrics, Uppsala University, Uppsala, Sweden
| | - Emma Ahlqvist
- Clinical Sciences Malmö, Genomics, Diabetes and Endocrinology, Lund University, Malmö, Sweden
| | - Richard N Bergman
- Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Yii-Der Ida Chen
- Department of Pediatrics, Genomic Outcomes, The Institute for Translational Genomics and Population Sciences, The Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Francis S Collins
- Center for Precision Health Research National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Tove Fall
- Department of Medical Sciences, Molecular Epidemiology, Uppsala University, Uppsala, Sweden
| | - Jose C Florez
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Programs in Metabolism and Medical and Population Genetics, The Broad Institute, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Andreas Fritsche
- Department of Internal Medicine IV, University Hospital Tübingen, Tübingen, Germany
| | - Harald Grallert
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Leif Groop
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- Clinical Sciences Malmö, Genomics, Diabetes and Endocrinology, Lund University, Lund, Sweden
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Heikki A Koistinen
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
- Department of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Pirjo Komulainen
- Foundation for Research in Health Exercise and Nutrition, Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
| | - Markku Laakso
- Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Lars Lind
- Department of Medical Sciences, Clinical Epidemiology, Uppsala University, Uppsala, Sweden
| | - Markus Loeffler
- Medical Faculty Institute for Medical Informatics, Statistics and Epidemiology, Leipzig, Germany
- LIFE Leipzig Research Center for Civilization Diseases, Medical Faculty, Leipzig, Germany
| | - Winfried März
- Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Synlab Academy, SYNLAB Holding Deutschland GmbH, Mannheim, Germany
| | - James B Meigs
- Department of Internal Medicine IV, University Hospital Tübingen, Tübingen, Germany
- Clinical Sciences Malmö, Genomics, Diabetes and Endocrinology, Lund University, Lund, Sweden
- Department of Medicine Division of General Internal Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Leslie J Raffel
- Department of Pediatrics, Genetic and Genomic Medicine, University of California, Irvine, CA, USA
| | - Rainer Rauramaa
- Foundation for Research in Health Exercise and Nutrition, Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
| | - 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
| | - Peter E H Schwarz
- Helmholtz Zentrum München Paul Langerhans Institute Dresden (PLID), University Hospital and Faculty of Medicine TU Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
- Department of Internal Medicine III, Prevention and Care of Diabetes, Medical Faculty Carl Gustav Carus, Dresden, Germany
| | - Michael Stumvoll
- Medical Department III-Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany
| | - Johan Sundström
- Department of Medical Sciences, Clinical Epidemiology, Uppsala University, Uppsala, Sweden
| | - Anke Tönjes
- Medical Department III-Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany
| | - Tiinamaija Tuomi
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland
| | - Jaakko Tuomilehto
- Department of Public Health, University of Helsinki, Helsinki, Finland
- Population Health Unit, Finnish Institute for Health and Welfare, Helsinki, Finland
- Diabetes Research Group, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Robert Wagner
- Department of Internal Medicine IV, University Hospital Tübingen, Tübingen, Germany
| | - Inês Barroso
- Exeter Centre of Excellence for Diabetes Research (EXCEED), Genetics of Complex Traits, University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Mark Walker
- Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Niels Grarup
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Michael Boehnke
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
- Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Nicholas J Wareham
- MRC Epidemiology Unit Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA.
| | - Eleanor Wheeler
- MRC Epidemiology Unit Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK.
| | - Stephen O'Rahilly
- Metabolic Research Laboratories Wellcome Trust-MRC Institute of Metabolic Science, Department of Clinical Biochemistry, University of Cambridge, Cambridge, UK.
| | - Daniel J Fazakerley
- Metabolic Research Laboratories Wellcome Trust-MRC Institute of Metabolic Science, Department of Clinical Biochemistry, University of Cambridge, Cambridge, UK.
| | - Claudia Langenberg
- MRC Epidemiology Unit Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK.
- Computational Medicine, Berlin Institute of Health at Charité-Universitätsmedizin, Berlin, Germany.
- Precision Healthcare University Research Institute, Queen Mary University of London, London, UK.
| |
Collapse
|
18
|
Gonzales TI, Jeon JY, Lindsay T, Westgate K, Perez-Pozuelo I, Hollidge S, Wijndaele K, Rennie K, Forouhi N, Griffin S, Wareham N, Brage S. Resting heart rate is a population-level biomarker of cardiorespiratory fitness: The Fenland Study. PLoS One 2023; 18:e0285272. [PMID: 37167327 PMCID: PMC10174582 DOI: 10.1371/journal.pone.0285272] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 04/19/2023] [Indexed: 05/13/2023] Open
Abstract
INTRODUCTION Few large studies have evaluated the relationship between resting heart rate (RHR) and cardiorespiratory fitness. Here we examine cross-sectional and longitudinal relationships between RHR and fitness, explore factors that influence these relationships, and demonstrate the utility of RHR for remote population monitoring. METHODS In cross-sectional analyses (The UK Fenland Study: 5,722 women, 5,143 men, aged 29-65y), we measured RHR (beats per min, bpm) while seated, supine, and during sleep. Fitness was estimated as maximal oxygen consumption (ml⋅min-1⋅kg-1) from an exercise test. Associations between RHR and fitness were evaluated while adjusting for age, sex, adiposity, and physical activity. In longitudinal analyses (6,589 participant subsample), we re-assessed RHR and fitness after a median of 6 years and evaluated the association between within-person change in RHR and fitness. During the coronavirus disease-2019 pandemic, we used a smartphone application to remotely and serially measure RHR (1,914 participant subsample, August 2020 to April 2021) and examined differences in RHR dynamics by pre-pandemic fitness level. RESULTS Mean RHR while seated, supine, and during sleep was 67, 64, and 57 bpm. Age-adjusted associations (beta coefficients) between RHR and fitness were -0.26, -0.29, and -0.21 ml⋅kg-1⋅beat-1 in women and -0.27, -0.31, and -0.19 ml⋅kg-1⋅beat-1 in men. Adjustment for adiposity and physical activity attenuated the RHR-to-fitness relationship by 10% and 50%, respectively. Longitudinally, a 1-bpm increase in supine RHR was associated with a 0.23 ml⋅min-1⋅kg-1 decrease in fitness. During the pandemic, RHR increased in those with low pre-pandemic fitness but was stable in others. CONCLUSIONS RHR is a valid population-level biomarker of cardiorespiratory fitness. Physical activity and adiposity attenuate the relationship between RHR and fitness.
Collapse
Affiliation(s)
- Tomas I. Gonzales
- MRC Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Justin Y. Jeon
- MRC Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
- Department of Sport Industry Studies, Exercise Medicine Center for Diabetes and Cancer Patients (ICONS), Yonsei University, Seoul, Korea
| | - Timothy Lindsay
- MRC Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Kate Westgate
- MRC Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
| | | | - Stefanie Hollidge
- MRC Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Katrien Wijndaele
- MRC Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Kirsten Rennie
- MRC Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Nita Forouhi
- MRC Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Simon Griffin
- MRC Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Nick Wareham
- MRC Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Soren Brage
- MRC Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
19
|
Montenegro Mendoza R, Roa R, Fontes F, Moreno Velásquez I, Quintana H. Physical Inactivity and Sedentary Behaviour among Panamanian Adults: Results from the National Health Survey of Panama (ENSPA) 2019. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:ijerph20085554. [PMID: 37107836 PMCID: PMC10138807 DOI: 10.3390/ijerph20085554] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/06/2023] [Accepted: 03/10/2023] [Indexed: 05/07/2023]
Abstract
Physical inactivity (PI) has been described as an independent risk factor for a large number of major non-communicable diseases and is associated with an increased risk of premature death. Additionally, sedentary behaviour has been associated with increased overall mortality. We estimated the national prevalence of PI and sedentary behaviour using the Global Physical Activity Questionnaire version 2. Using unconditional logistic regressions, the possible risk factors for PI were assessed. Over half of the people included in this study (54.9%; 95% CI: 54.1-57.3%) were physically inactive, with the median time spent engaged in sedentary behaviour being 120 min per day. Statistically significant associations with PI were observed with regard to sex, living area, and alcohol consumption. PI prevalence in Panama was elevated and showed a sex difference (women: 64.7%, 95% CI: 63.7-66.7%; men: 43.4%, 95% CI: 41.5-47.5%). According to our analysis of three-domain-related physical activities, the main contribution to the total estimated energy expenditure of physical activity/week came from the transport domain, followed by the work/household domain, and the least significant contributor was consistently the domain of exercise- and sports-related physical activities.
Collapse
Affiliation(s)
- Roger Montenegro Mendoza
- Department of Research and Health Technology Assessment, Gorgas Memorial Institute for Health Studies, Panama City 0816-02593, Panama
- Correspondence: ; Tel.: +507-527-4961
| | - Reina Roa
- Planning Directorate, Ministry of Health, Panama City 4444, Panama
| | - Flavia Fontes
- Dietetic and Nutrition Department, Faculty of Medicine, University of Panama, Panama City 3366, Panama
| | - Ilais Moreno Velásquez
- Department of Research and Health Technology Assessment, Gorgas Memorial Institute for Health Studies, Panama City 0816-02593, Panama
| | - Hedley Quintana
- Department of Research and Health Technology Assessment, Gorgas Memorial Institute for Health Studies, Panama City 0816-02593, Panama
| |
Collapse
|
20
|
The moderating role of eating behaviour traits in the association between exposure to hot food takeaway outlets and body fatness. Int J Obes (Lond) 2023; 47:496-504. [PMID: 36918687 DOI: 10.1038/s41366-023-01290-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 02/21/2023] [Accepted: 02/28/2023] [Indexed: 03/16/2023]
Abstract
BACKGROUND Previous studies demonstrated a relation between takeaway outlet exposure and health outcomes. Individual characteristics, such as eating behaviour traits, could make some people more susceptible to the influence of the food environment. Few studies have investigated this topic. We aimed to investigate the moderating role of eating behaviour traits (cognitive restraint, uncontrolled eating and emotional eating) in the association between neighbourhood exposure to hot food takeaway outlets (hereafter referred to as takeaway outlets), and takeaway food consumption and adiposity. METHODS We used cross-sectional data from a cohort in Cambridgeshire, UK (The Fenland study). Takeaway outlet exposure was derived using participants' residential address and data from local authorities and divided into quarters. The Three Factor Eating questionnaire (TFEQ-R18) was used to measure eating behaviour traits. Primary outcomes were consumption of takeaway-like foods (derived from food frequency questionnaire), and body fat percentage (measured using dual-energy X-ray absorptiometry). RESULTS Mean age of participants (n = 4791) was 51.0 (SD = 7.2) and 53.9% were female. Higher exposure to takeaway outlets in the neighbourhood and higher eating behaviour trait scores were independently associated with greater takeaway consumption and body fat percentage. Uncontrolled eating did not moderate the associations between takeaway outlet exposure and takeaway consumption or body fat percentage. The association between takeaway outlet exposure and takeaway consumption was slightly stronger in those with higher cognitive restraint scores, and the association between takeaway outlet exposure and body fat percentage was slightly stronger in those with lower emotional eating scores. CONCLUSION Eating behaviour traits and exposure to takeaway outlets were associated with greater takeaway consumption and body fat, but evidence that individuals with certain traits are more susceptible to takeaway outlets was weak. The findings indicate that interventions at both the individual and environmental levels are needed to comprehensively address unhealthy diets. TRIAL REGISTRY ISRCTN72077169.
Collapse
|
21
|
GONZALES TOMASI, WESTGATE KATE, HOLLIDGE STEFANIE, LINDSAY TIM, WIJNDAELE KATRIEN, FOROUHI NITAG, GRIFFIN SIMON, WAREHAM NICK, BRAGE SOREN. Descriptive Epidemiology of Cardiorespiratory Fitness in UK Adults: The Fenland Study. Med Sci Sports Exerc 2023; 55:507-516. [PMID: 36730941 PMCID: PMC9924962 DOI: 10.1249/mss.0000000000003068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
INTRODUCTION Cardiorespiratory fitness (CRF) is rarely measured in population studies. Most studies of CRF do not examine differences by population subgroups or seasonal trends. We examined how estimated CRF levels vary by anthropometric, sociodemographic, and behavioral characteristics in a population-based cohort of UK adults (the Fenland Study). METHODS We used a validated submaximal exercise test to obtain CRF estimates (CRF estimated ) in 5976 women and 5316 men, residing in the East of England. CRF estimated was defined as estimated maximal oxygen consumption per kilogram total body mass (V̇O 2 max tbm ) and fat-free mass (V̇O 2 max ffm ). Descriptive statistics were computed across anthropometric and sociodemographic characteristics, and across the year. Progressive multivariable analyses were performed to examine associations with physical activity energy expenditure (PAEE) and body mass index (BMI). RESULTS Mean ± SD V̇O 2 max tbm was lower in women (35.2 ± 7.5 mL·min -1 ·kg -1 ) than men (41.7 ± 7.3 mL·min -1 ·kg -1 ) but V̇O 2 max ffm was similar (women: 59.2 ± 11.6 mL·min -1 ·kg -1 ; men: 62.0 ± 10.3 mL·min -1 ·kg -1 ). CRF estimated was inversely associated with age but not after adjustment for PAEE. People in more physically demanding jobs were fitter compared with those in sedentary jobs, but this association was attenuated in women and reversed in men after adjustment for total PAEE. Physical activity energy expenditure and BMI were positively associated with CRF estimated at all levels of adjustment when expressed relative to fat-free mass. CRF estimated was 4% higher in summer than in winter among women, but did not differ by season among men. CONCLUSIONS CRF estimated was inversely associated with age but less steeply than anticipated, suggesting older generations are comparatively fitter than younger generations. Physical activity energy expenditure and BMI were stronger determinants of the variance in CRF estimated than other characteristic including age. This emphasizes the importance of modifiable physical activity behaviors in public health interventions.
Collapse
|
22
|
Lin X, Yang Y, Fuh-Ngwa V, Yin X, Simpson-Yap S, van der Mei I, Broadley SA, Ponsonby AL, Burdon KP, Taylor BV, Zhou Y. Genetically determined serum serine level has a novel causal effect on multiple sclerosis risk and predicts disability progression. J Neurol Neurosurg Psychiatry 2023:jnnp-2022-330259. [PMID: 36732044 DOI: 10.1136/jnnp-2022-330259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 01/19/2023] [Indexed: 02/04/2023]
Abstract
BACKGROUND There are currently no specific biomarkers for multiple sclerosis (MS). Identifying robust biomarkers for MS is crucial to improve disease diagnosis and management. METHODS This study first used six Mendelian randomisation methods to assess causal relationship of 174 metabolites with MS, incorporating data from European-ancestry metabolomics (n=8569-86 507) and MS (n=14 802 MS cases, 26 703 controls) genomewide association studies. Genetic scores for identified causal metabolite(s) were then computed to predict MS disability progression in an independent longitudinal cohort (AusLong study) of 203 MS cases with up to 15-year follow-up. RESULTS We found a novel genetic causal effect of serine on MS onset (OR=1.67, 95% CI 1.51 to 1.84, p=1.73×10-20), such that individuals whose serine level is 1 SD above the population mean will have 1.67 times the risk of developing MS. This is robust across all sensitivity methods (OR ranges from 1.49 to 1.67). In an independent longitudinal MS cohort, we then constructed time-dynamic and time-fixed genetic scores based on serine genetic instrument single-nucleotide polymorphisms, where higher scores for raised serum serine level were associated with increased risk of disability worsening, especially in the time-dynamic model (RR=1.25, 95% CI 1.10 to 1.42, p=7.52×10-4). CONCLUSIONS These findings support investigating serine as an important candidate biomarker for MS onset and disability progression.
Collapse
Affiliation(s)
- Xin Lin
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Yuanhao Yang
- Mater Research Institute, Translational Research Institute, Woolloongabba, Queensland, Australia.,Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland, Australia
| | - Valery Fuh-Ngwa
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Xianyong Yin
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, USA
| | - Steve Simpson-Yap
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia.,Neuroepidemiology Unit, The University of Melbourne School of Population and Global Health, Melbourne, Victoria, Australia
| | - Ingrid van der Mei
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Simon A Broadley
- School of Medicine, Griffith University, Gold Coast, Queensland, Australia
| | - Anne-Louise Ponsonby
- Murdoch Children's Research Institute, Royal Children's Hospital, University of Melbourne, Parkville, Victoria, Australia.,Neuroepidemiology Group, The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
| | | | - Kathryn P Burdon
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Bruce V Taylor
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Yuan Zhou
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| |
Collapse
|
23
|
Watts EL, Saint-Maurice PF, Doherty A, Fensom GK, Freeman JR, Gorzelitz JS, Jin D, McClain KM, Papier K, Patel S, Shiroma EJ, Moore SC, Matthews CE. Association of Accelerometer-Measured Physical Activity Level With Risks of Hospitalization for 25 Common Health Conditions in UK Adults. JAMA Netw Open 2023; 6:e2256186. [PMID: 36795414 PMCID: PMC9936337 DOI: 10.1001/jamanetworkopen.2022.56186] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 12/29/2022] [Indexed: 02/17/2023] Open
Abstract
Importance Higher physical activity levels are associated with lower risks of cancer, cardiovascular disease, and diabetes, but associations with many common and less severe health conditions are not known. These conditions impose large health care burdens and reduce quality of life. Objectives To investigate the association between accelerometer-measured physical activity and the subsequent risk of hospitalization for 25 common reasons for hospitalization and to estimate the proportion of these hospitalizations that might have been prevented if participants had higher levels of physical activity. Design, Setting, and Participants This prospective cohort study used data from a subset of 81 717 UK Biobank participants aged 42 to 78 years. Participants wore an accelerometer for 1 week (between June 1, 2013, and December 23, 2015) and were followed up over a median (IQR) of 6.8 (6.2-7.3) years; follow-up for the current study ended in 2021 (exact date varied by location). Exposures Mean total and intensity-specific accelerometer-measured physical activity. Main Outcomes and Measures Hospitalization for the most common health conditions. Cox proportional hazards regression analysis was used to estimate hazard ratios (HRs) and 95% CIs for mean accelerometer-measured physical activity (per 1-SD increment) and risks of hospitalization for 25 conditions. Population-attributable risks were used to estimate the proportion of hospitalizations for each condition that might be prevented if participants increased their moderate to vigorous physical activity (MVPA) by 20 minutes per day. Results Among 81 717 participants, the mean (SD) age at accelerometer assessment was 61.5 (7.9) years; 56.4% were female, and 97.0% self-identified as White. Higher levels of accelerometer-measured physical activity were associated with lower risks of hospitalization for 9 conditions: gallbladder disease (HR per 1 SD, 0.74; 95% CI, 0.69-0.79), urinary tract infections (HR per 1 SD, 0.76; 95% CI, 0.69-0.84), diabetes (HR per 1 SD, 0.79; 95% CI, 0.74-0.84), venous thromboembolism (HR per 1 SD, 0.82; 95% CI, 0.75-0.90), pneumonia (HR per 1 SD, 0.83; 95% CI, 0.77-0.89), ischemic stroke (HR per 1 SD, 0.85; 95% CI, 0.76-0.95), iron deficiency anemia (HR per 1 SD, 0.91; 95% CI, 0.84-0.98), diverticular disease (HR per 1 SD, 0.94; 95% CI, 0.90-0.99), and colon polyps (HR per 1 SD, 0.96; 95% CI, 0.94-0.99). Positive associations were observed between overall physical activity and carpal tunnel syndrome (HR per 1 SD, 1.28; 95% CI, 1.18-1.40), osteoarthritis (HR per 1 SD, 1.15; 95% CI, 1.10-1.19), and inguinal hernia (HR per 1 SD, 1.13; 95% CI, 1.07-1.19), which were primarily induced by light physical activity. Increasing MVPA by 20 minutes per day was associated with reductions in hospitalization ranging from 3.8% (95% CI, 1.8%-5.7%) for colon polyps to 23.0% (95% CI, 17.1%-28.9%) for diabetes. Conclusions and Relevance In this cohort study of UK Biobank participants, those with higher physical activity levels had lower risks of hospitalization across a broad range of health conditions. These findings suggest that aiming to increase MVPA by 20 minutes per day may be a useful nonpharmaceutical intervention to reduce health care burdens and improve quality of life.
Collapse
Affiliation(s)
- Eleanor L. Watts
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| | - Pedro F. Saint-Maurice
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| | - Aiden Doherty
- Big Data Institute, Nuffield Department of Population Health, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Georgina K. Fensom
- Cancer Epidemiology Unit, Nuffield Department of Population Health, Oxford, United Kingdom
| | - Joshua R. Freeman
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| | | | - David Jin
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| | - Kathleen M. McClain
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| | - Keren Papier
- Cancer Epidemiology Unit, Nuffield Department of Population Health, Oxford, United Kingdom
| | - Shreya Patel
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| | - Eric J. Shiroma
- Laboratory of Epidemiology and Population Science, National Institute on Aging, Bethesda, Maryland
| | - Steven C. Moore
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| | - Charles E. Matthews
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| |
Collapse
|
24
|
Collings PJ, Backes A, Aguayo GA, Malisoux L, on behalf of the ORISCAV-LUX study group AlkerwiAla’aNoppeStephanieDelagardelleCharlesBeisselJeanChiotiAnnaStrangesSaverioSchmitJean-ClaudeLairMarie-LiseD’IncauMarylènePastoreJessicaLe CorollerGwenaëlleAppenzellerBriceCouffignalSophieGantenbeinManonDevauxYvanVaillantMichelHuiartLaetitiaBejkoDritanBohnTorstenSamoudaHanenFagherazziGuyPerquinMagaliRuizMariaErnensIsabelle. Device-measured physical activity and sedentary time in a national sample of Luxembourg residents: the ORISCAV-LUX 2 study. Int J Behav Nutr Phys Act 2022; 19:161. [PMID: 36581944 PMCID: PMC9798598 DOI: 10.1186/s12966-022-01380-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 11/05/2022] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Existing information about population physical activity (PA) levels and sedentary time in Luxembourg are based on self-reported data. METHODS This observational study included Luxembourg residents aged 18-79y who each provided ≥4 valid days of triaxial accelerometry in 2016-18 (n=1122). Compliance with the current international PA guideline (≥150 min moderate-to-vigorous PA (MVPA) per week, irrespective of bout length) was quantified and variability in average 24h acceleration (indicative of PA volume), awake-time PA levels, sedentary time and accumulation pattern were analysed by linear regression. Data were weighted to be nationally representative. RESULTS Participants spent 51% of daily time sedentary (mean (95% confidence interval (CI)): 12.1 (12.0 to 12.2) h/day), 11% in light PA (2.7 (2.6 to 2.8) h/day), 6% in MVPA (1.5 (1.4 to 1.5) h/day), and remaining time asleep (7.7 (7.6 to 7.7) h/day). Adherence to the PA guideline was high (98.1%). Average 24h acceleration and light PA were higher in women than men, but men achieved higher average accelerations across the most active periods of the day. Women performed less sedentary time and shorter sedentary bouts. Older participants (aged ≥55y) registered a lower average 24h acceleration and engaged in less MVPA, more sedentary time and longer sedentary bouts. Average 24h acceleration was higher in participants of lower educational attainment, who also performed less sedentary time, shorter bouts, and fewer bouts of prolonged sedentariness. Average 24h acceleration and levels of PA were higher in participants with standing and manual occupations than a sedentary work type, but manual workers registered lower average accelerations across the most active periods of the day. Standing and manual workers accumulated less sedentary time and fewer bouts of prolonged sedentariness than sedentary workers. Active commuting to work was associated with higher average 24h acceleration and MVPA, both of which were lower in participants of poorer self-rated health and higher weight status. Obesity was associated with less light PA, more sedentary time and longer sedentary bouts. CONCLUSIONS Adherence to recommended PA is high in Luxembourg, but half of daily time is spent sedentary. Specific population subgroups will benefit from targeted efforts to replace sedentary time with PA.
Collapse
Affiliation(s)
- Paul J. Collings
- grid.451012.30000 0004 0621 531XPhysical Activity, Sport and Health Research Group, Department of Precision Health, Luxembourg Institute of Health, 1A-B, rue Thomas Edison, Strassen, L-1445 Luxembourg
| | - Anne Backes
- grid.451012.30000 0004 0621 531XPhysical Activity, Sport and Health Research Group, Department of Precision Health, Luxembourg Institute of Health, 1A-B, rue Thomas Edison, Strassen, L-1445 Luxembourg
| | - Gloria A. Aguayo
- grid.451012.30000 0004 0621 531XDeep Digital Phenotyping Research Unit, Department of Precision Health, Luxembourg Institute of Health, 1A-B, rue Thomas Edison, Strassen, L-1445 Luxembourg
| | - Laurent Malisoux
- grid.451012.30000 0004 0621 531XPhysical Activity, Sport and Health Research Group, Department of Precision Health, Luxembourg Institute of Health, 1A-B, rue Thomas Edison, Strassen, L-1445 Luxembourg
| | | |
Collapse
|
25
|
Spathis D, Perez-Pozuelo I, Gonzales TI, Wu Y, Brage S, Wareham N, Mascolo C. Longitudinal cardio-respiratory fitness prediction through wearables in free-living environments. NPJ Digit Med 2022; 5:176. [PMID: 36460766 PMCID: PMC9718831 DOI: 10.1038/s41746-022-00719-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 10/31/2022] [Indexed: 12/04/2022] Open
Abstract
Cardiorespiratory fitness is an established predictor of metabolic disease and mortality. Fitness is directly measured as maximal oxygen consumption (VO2max), or indirectly assessed using heart rate responses to standard exercise tests. However, such testing is costly and burdensome because it requires specialized equipment such as treadmills and oxygen masks, limiting its utility. Modern wearables capture dynamic real-world data which could improve fitness prediction. In this work, we design algorithms and models that convert raw wearable sensor data into cardiorespiratory fitness estimates. We validate these estimates' ability to capture fitness profiles in free-living conditions using the Fenland Study (N=11,059), along with its longitudinal cohort (N = 2675), and a third external cohort using the UK Biobank Validation Study (N = 181) who underwent maximal VO2max testing, the gold standard measurement of fitness. Our results show that the combination of wearables and other biomarkers as inputs to neural networks yields a strong correlation to ground truth in a holdout sample (r = 0.82, 95CI 0.80-0.83), outperforming other approaches and models and detects fitness change over time (e.g., after 7 years). We also show how the model's latent space can be used for fitness-aware patient subtyping paving the way to scalable interventions and personalized trial recruitment. These results demonstrate the value of wearables for fitness estimation that today can be measured only with laboratory tests.
Collapse
Affiliation(s)
- Dimitris Spathis
- Department of Computer Science and Technology, University of Cambridge, Cambridge, UK.
| | - Ignacio Perez-Pozuelo
- MRC Epidemiology Unit, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Tomas I Gonzales
- MRC Epidemiology Unit, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Yu Wu
- Department of Computer Science and Technology, University of Cambridge, Cambridge, UK
| | - Soren Brage
- MRC Epidemiology Unit, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Nicholas Wareham
- MRC Epidemiology Unit, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Cecilia Mascolo
- Department of Computer Science and Technology, University of Cambridge, Cambridge, UK
| |
Collapse
|
26
|
Carrasco-Zanini J, Pietzner M, Lindbohm JV, Wheeler E, Oerton E, Kerrison N, Simpson M, Westacott M, Drolet D, Kivimaki M, Ostroff R, Williams SA, Wareham NJ, Langenberg C. Proteomic signatures for identification of impaired glucose tolerance. Nat Med 2022; 28:2293-2300. [PMID: 36357677 PMCID: PMC7614638 DOI: 10.1038/s41591-022-02055-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 09/27/2022] [Indexed: 11/12/2022]
Abstract
The implementation of recommendations for type 2 diabetes (T2D) screening and diagnosis focuses on the measurement of glycated hemoglobin (HbA1c) and fasting glucose. This approach leaves a large number of individuals with isolated impaired glucose tolerance (iIGT), who are only detectable through oral glucose tolerance tests (OGTTs), at risk of diabetes and its severe complications. We applied machine learning to the proteomic profiles of a single fasted sample from 11,546 participants of the Fenland study to test discrimination of iIGT defined using the gold-standard OGTTs. We observed significantly improved discriminative performance by adding only three proteins (RTN4R, CBPM and GHR) to the best clinical model (AUROC = 0.80 (95% confidence interval: 0.79-0.86), P = 0.004), which we validated in an external cohort. Increased plasma levels of these candidate proteins were associated with an increased risk for future T2D in an independent cohort and were also increased in individuals genetically susceptible to impaired glucose homeostasis and T2D. Assessment of a limited number of proteins can identify individuals likely to be missed by current diagnostic strategies and at high risk of T2D and its complications.
Collapse
Affiliation(s)
- Julia Carrasco-Zanini
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Maik Pietzner
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
- Computational Medicine, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Joni V Lindbohm
- Clinicum, Department of Public Health, University of Helsinki, Helsinki, Finland
- Department of Epidemiology and Public Health, University College London, London, UK
- The Klarman Cell Observatory, Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - Eleanor Wheeler
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Erin Oerton
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Nicola Kerrison
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | | | | | | | - Mika Kivimaki
- Clinicum, Department of Public Health, University of Helsinki, Helsinki, Finland
- Department of Epidemiology and Public Health, University College London, London, UK
| | | | | | - Nicholas J Wareham
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Claudia Langenberg
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK.
- Computational Medicine, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany.
- Precision Healthcare University Research Institute, Queen Mary University of London, London, UK.
| |
Collapse
|
27
|
Li R, Deng M, Lin Y, Gao W, Liu B, Xia H. Genetically predicted circulating levels of glycine, glutamate, and serotonin in relation to the risks of three major neurodegenerative diseases: A Mendelian randomization analysis. Front Aging Neurosci 2022; 14:938408. [PMID: 36158554 PMCID: PMC9490425 DOI: 10.3389/fnagi.2022.938408] [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: 05/07/2022] [Accepted: 08/22/2022] [Indexed: 11/13/2022] Open
Abstract
It has been previously postulated that blood neurotransmitters might affect risks of neurodegenerative diseases. Here, a Mendelian Randomization (MR) study was conducted to explore whether genetically predicted concentrations of glycine, glutamate and serotonin were associated with risks of Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS). From three genome-wide association studies of European ancestry, single nucleotide polymorphisms strongly associated with glycine, glutamate and serotonin were selected as genetic instrumental variables. Corresponding summary statistics were also obtained from the latest genome-wide association meta-analyses of AD, PD and ALS. The inverse-variance weighted MR and multiple sensitivity analyses were performed to evaluate causal effects of genetically predicted levels of neurotransmitters on risks of neurodegenerative diseases. The statistical significance threshold was set at P < 0.0056 using the Bonferroni-correction, while 0.0056 < P < 0.05 was considered suggestive evidence for a causal association. There was a causal association of elevated blood glutamate levels with higher AD risks. The odds ratio (OR) of AD was 1.311 [95% confidence interval (CI), 1.087-1.580; P = 0.004] per one standard deviation increase in genetically predicted glutamate concentrations. There was suggestive evidence in support of a protective effect of blood serotonin on AD (OR = 0.607; 95% CI, 0.396-0.932; P = 0.022). Genetically predicted glycine levels were not associated with the risk of AD (OR = 1.145; 95% CI, 0.939-1.396; P = 0.180). Besides, MR analyses indicated no causal roles of three blood neurotransmitters in PD or ALS. In conclusion, the MR study provided evidence supporting the association of elevated blood glutamate levels with higher AD risks and the association of increased blood serotonin levels with lower AD risks. Triangulating evidence across further study designs is still warranted to elucidate the role of blood neurotransmitters in risks of neurodegenerative diseases.
Collapse
Affiliation(s)
- Ruizhuo Li
- School of Medicine, South China University of Technology, Guangzhou, China.,Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Provincial Key Laboratory of Research in Structure Birth Defect Disease, Guangzhou, China
| | - Mengjuan Deng
- Department of Anesthesiology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Yuhong Lin
- Zhongshan School of Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wenjing Gao
- Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Provincial Key Laboratory of Research in Structure Birth Defect Disease, Guangzhou, China
| | - Bohao Liu
- Xiangya School of Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Huimin Xia
- School of Medicine, South China University of Technology, Guangzhou, China.,Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Provincial Key Laboratory of Research in Structure Birth Defect Disease, Guangzhou, China
| |
Collapse
|
28
|
Li Y, Cheng Y, Consolato F, Schiano G, Chong MR, Pietzner M, Nguyen NQH, Scherer N, Biggs ML, Kleber ME, Haug S, Göçmen B, Pigeyre M, Sekula P, Steinbrenner I, Schlosser P, Joseph CB, Brody JA, Grams ME, Hayward C, Schultheiss UT, Krämer BK, Kronenberg F, Peters A, Seissler J, Steubl D, Then C, Wuttke M, März W, Eckardt KU, Gieger C, Boerwinkle E, Psaty BM, Coresh J, Oefner PJ, Pare G, Langenberg C, Scherberich JE, Yu B, Akilesh S, Devuyst O, Rampoldi L, Köttgen A. Genome-wide studies reveal factors associated with circulating uromodulin and its relationships to complex diseases. JCI Insight 2022; 7:e157035. [PMID: 35446786 PMCID: PMC9220927 DOI: 10.1172/jci.insight.157035] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 04/07/2022] [Indexed: 11/28/2022] Open
Abstract
Uromodulin (UMOD) is a major risk gene for monogenic and complex forms of kidney disease. The encoded kidney-specific protein uromodulin is highly abundant in urine and related to chronic kidney disease, hypertension, and pathogen defense. To gain insights into potential systemic roles, we performed genome-wide screens of circulating uromodulin using complementary antibody-based and aptamer-based assays. We detected 3 and 10 distinct significant loci, respectively. Integration of antibody-based results at the UMOD locus with functional genomics data (RNA-Seq, ATAC-Seq, Hi-C) of primary human kidney tissue highlighted an upstream variant with differential accessibility and transcription in uromodulin-synthesizing kidney cells as underlying the observed cis effect. Shared association patterns with complex traits, including chronic kidney disease and blood pressure, placed the PRKAG2 locus in the same pathway as UMOD. Experimental validation of the third antibody-based locus, B4GALNT2, showed that the p.Cys466Arg variant of the encoded N-acetylgalactosaminyltransferase had a loss-of-function effect leading to higher serum uromodulin levels. Aptamer-based results pointed to enzymes writing glycan marks present on uromodulin and to their receptors in the circulation, suggesting that this assay permits investigating uromodulin's complex glycosylation rather than its quantitative levels. Overall, our study provides insights into circulating uromodulin and its emerging functions.
Collapse
Affiliation(s)
- Yong Li
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, and
| | - Yurong Cheng
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, and
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Francesco Consolato
- Molecular Genetics of Renal Disorders group, Division of Genetics and Cell Biology, IRCCS Ospedale San Raffaele, Milan, Italy
| | | | - Michael R. Chong
- Population Health Research Institute and Thrombosis and Atherosclerosis Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton Health Sciences, Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences and
- Department of Pathology and Molecular Medicine, Faculty of Health Science, McMaster University, Hamilton, Ontario, Canada
| | - Maik Pietzner
- Medical Research Council (MRC) Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
- Computational Medicine, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Ngoc Quynh H. Nguyen
- Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Nora Scherer
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, and
- Spemann Graduate School of Biology and Medicine, University of Freiburg, Freiburg, Germany
| | - Mary L. Biggs
- Cardiovascular Health Research Unit, Department of Medicine, and
- Department of Biostatistics, University of Washington, Seattle, Washington, USA
| | - Marcus E. Kleber
- SYNLAB MVZ Humangenetik Mannheim GmbH, Mannheim, Germany
- Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Stefan Haug
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, and
| | - Burulça Göçmen
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, and
| | - Marie Pigeyre
- Population Health Research Institute and Thrombosis and Atherosclerosis Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton Health Sciences, Hamilton, Ontario, Canada
- Department of Medicine, Michael G. DeGroote School of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Peggy Sekula
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, and
| | - Inga Steinbrenner
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, and
| | - Pascal Schlosser
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, and
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Christina B. Joseph
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh, United Kingdom
| | | | - Morgan E. Grams
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Division of Nephrology, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Caroline Hayward
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh, United Kingdom
| | - Ulla T. Schultheiss
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, and
- Department of Medicine IV: Nephrology and Primary Care, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Bernhard K. Krämer
- Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Florian Kronenberg
- Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
- Chair of Epidemiology, Institute for Medical Information Processing, Biometry, and Epidemiology, Faculty of Medicine, Ludwig-Maximilians-Universität (LMU), Munich, Germany
| | - Jochen Seissler
- Medical Clinic and Policlinic IV, Hospital of the University of Munich, LMU Munich, Munich, Germany
| | - Dominik Steubl
- Division of Nephrology, Tufts Medical Center, Boston, Massachusetts, USA
- Department of Nephrology, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
- Boehringer Ingelheim International GmbH, Ingelheim, Germany
| | - Cornelia Then
- Medical Clinic and Policlinic IV, Hospital of the University of Munich, LMU Munich, Munich, Germany
| | - Matthias Wuttke
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, and
- Department of Medicine IV: Nephrology and Primary Care, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Winfried März
- Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
- SYNLAB Academy, SYNLAB Holding Deutschland GmbH, Augsburg and Mannheim, Germany
| | - Kai-Uwe Eckardt
- Department of Nephrology and Medical Intensive Care, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Department of Nephrology and Hypertension, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Christian Gieger
- Institute of Epidemiology, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
- Research Unit of Molecular Epidemiology, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Partner Munich, Neuherberg, Germany
| | - Eric Boerwinkle
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, USA
| | - Bruce M. Psaty
- Cardiovascular Health Research Unit, Department of Medicine, and
- Department of Epidemiology and
- Department of Health Systems and Population Health, School of Public Health, University of Washington, Seattle, Washington, USA
| | - Josef Coresh
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Peter J. Oefner
- Institute of Functional Genomics, University of Regensburg, Regensburg, Germany
| | - Guillaume Pare
- Population Health Research Institute and Thrombosis and Atherosclerosis Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton Health Sciences, Hamilton, Ontario, Canada
- Department of Pathology and Molecular Medicine, Faculty of Health Science, McMaster University, Hamilton, Ontario, Canada
| | - Claudia Langenberg
- Medical Research Council (MRC) Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
- Computational Medicine, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
| | | | - Bing Yu
- Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Shreeram Akilesh
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Olivier Devuyst
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Luca Rampoldi
- Molecular Genetics of Renal Disorders group, Division of Genetics and Cell Biology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Anna Köttgen
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, and
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Centre for Integrative Biological Signalling Studies (CIBSS), University of Freiburg, Freiburg, Germany
| |
Collapse
|
29
|
Sandercock GRH, Moran J, Cohen DD. Who is meeting the strengthening physical activity guidelines by definition: A cross-sectional study of 253 423 English adults? PLoS One 2022; 17:e0267277. [PMID: 35507575 PMCID: PMC9067886 DOI: 10.1371/journal.pone.0267277] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 04/05/2022] [Indexed: 11/19/2022] Open
Abstract
The current UK physical activity guidelines recommend that adults aged 19 to 65 years perform activity to strengthen muscle and bone a minimum of twice weekly. The number of adults meeting strengthening activity guidelines is lower than for aerobic activity, but estimates vary between studies partly due to differences in how muscle-strengthening activity is defined. We aimed to provide estimates for strengthening activity prevalence in English adults based on a nationally representative sample of n = 253,423 18-65-year-olds. We attempted to quantify the variation in estimates attributable to differences in the way strengthening activity is defined. Finally, we aim to provide a brief descriptive epidemiology of the factors associated with strengthening activity. Adults met guidelines for aerobic activity if they reported the activity equivalent to >150 min/week moderate-intensity exercise. Respondents met strengthening guidelines if they reported at least two bouts per week of strengthening activity. We defined strengthening activity, first, according to criteria used in the Health Survey for England (HSE). Second, we counted bouts of strengthening activities for which we could find evidence of health-related benefits (Evidence). Third, we included bouts of strengthening activity as defined in current UK physical activity guidelines (Guideline). Two-thirds (67%) of adults met guidelines for aerobic activity (69% of men, 65% of women). Less than one-third (29% of men and 24% of women) met guidelines for the HSE definition of strengthening activity. Under the Evidence definition, 16% of men and 9% of women met strengthening guidelines. Using the most-stringent definition (Guideline) just 7.3% of men and 4.1% of women achieved the recommendations for strengthening activity. We found females and older adults (50–65 years) were less likely to meet guidelines for aerobic, strengthening, and combined aerobic plus strengthening activity. The prevalence of meeting activity guidelines was lower in adults from more deprived areas (compared with the least deprived); Adults with lower academic qualifications (Level 1) were less likely to meet activity guidelines than those educated to Level 4 (Degree Level) or higher. Having a limiting disability was associated with a lower prevalence of meeting activity guidelines. Associations between socio-demographic measures and the prevalence of adults meeting activity guidelines were stronger for strengthening activity than for aerobic 51(or combined aerobic plus strengthening) activity Compared with aerobic activity, fewer adults engage in strengthening activity regardless of how it is defined. The range in estimates for how many adults meet strengthening activity guidelines can be explained by variations in the definition of ‘strengthening’ that are used and the specific sports or activities identified as strengthening exercise. When strengthening activity is included, the proportion of English adults meeting current physical activity guidelines could be as high as 1 in 3 but possibly as low as just 1 in 20. A harmonized definition of strengthening activity, that is aligned with physical activity guidelines, is required to provide realistic and comparable prevalence estimates.
Collapse
Affiliation(s)
- Gavin R. H. Sandercock
- School of Sport Rehabilitation & Exercise Science, University of Essex, Colchester, United Kingdom
- * E-mail:
| | - Jason Moran
- School of Sport Rehabilitation & Exercise Science, University of Essex, Colchester, United Kingdom
| | - Daniel D. Cohen
- Masira Research Institute, Faculty of Health Sciences, Universidad de Santander (UDES), Bucaramanga, Colombia
| |
Collapse
|
30
|
Seyres D, Cabassi A, Lambourne JJ, Burden F, Farrow S, McKinney H, Batista J, Kempster C, Pietzner M, Slingsby O, Cao TH, Quinn PA, Stefanucci L, Sims MC, Rehnstrom K, Adams CL, Frary A, Ergüener B, Kreuzhuber R, Mocciaro G, D’Amore S, Koulman A, Grassi L, Griffin JL, Ng LL, Park A, Savage DB, Langenberg C, Bock C, Downes K, Wareham NJ, Allison M, Vacca M, Kirk PDW, Frontini M. Transcriptional, epigenetic and metabolic signatures in cardiometabolic syndrome defined by extreme phenotypes. Clin Epigenetics 2022; 14:39. [PMID: 35279219 PMCID: PMC8917653 DOI: 10.1186/s13148-022-01257-z] [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] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 02/25/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND This work is aimed at improving the understanding of cardiometabolic syndrome pathophysiology and its relationship with thrombosis by generating a multi-omic disease signature. METHODS/RESULTS We combined classic plasma biochemistry and plasma biomarkers with the transcriptional and epigenetic characterisation of cell types involved in thrombosis, obtained from two extreme phenotype groups (morbidly obese and lipodystrophy) and lean individuals to identify the molecular mechanisms at play, highlighting patterns of abnormal activation in innate immune phagocytic cells. Our analyses showed that extreme phenotype groups could be distinguished from lean individuals, and from each other, across all data layers. The characterisation of the same obese group, 6 months after bariatric surgery, revealed the loss of the abnormal activation of innate immune cells previously observed. However, rather than reverting to the gene expression landscape of lean individuals, this occurred via the establishment of novel gene expression landscapes. NETosis and its control mechanisms emerge amongst the pathways that show an improvement after surgical intervention. CONCLUSIONS We showed that the morbidly obese and lipodystrophy groups, despite some differences, shared a common cardiometabolic syndrome signature. We also showed that this could be used to discriminate, amongst the normal population, those individuals with a higher likelihood of presenting with the disease, even when not displaying the classic features.
Collapse
Affiliation(s)
- Denis Seyres
- National Institute for Health Research BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK. .,Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK. .,NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK.
| | - Alessandra Cabassi
- grid.5335.00000000121885934MRC Biostatistics Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - John J. Lambourne
- grid.24029.3d0000 0004 0383 8386National Institute for Health Research BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK ,grid.5335.00000000121885934Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK ,grid.436365.10000 0000 8685 6563NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK
| | - Frances Burden
- grid.24029.3d0000 0004 0383 8386National Institute for Health Research BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK ,grid.5335.00000000121885934Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK ,grid.436365.10000 0000 8685 6563NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK
| | - Samantha Farrow
- grid.24029.3d0000 0004 0383 8386National Institute for Health Research BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK ,grid.5335.00000000121885934Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK ,grid.436365.10000 0000 8685 6563NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK
| | - Harriet McKinney
- grid.5335.00000000121885934Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Joana Batista
- grid.5335.00000000121885934Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Carly Kempster
- grid.5335.00000000121885934Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Maik Pietzner
- grid.5335.00000000121885934MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Oliver Slingsby
- grid.9918.90000 0004 1936 8411Department of Cardiovascular Sciences, Glenfield Hospital, University of Leicester, Leicester, UK ,grid.412925.90000 0004 0400 6581National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Thong Huy Cao
- grid.9918.90000 0004 1936 8411Department of Cardiovascular Sciences, Glenfield Hospital, University of Leicester, Leicester, UK ,grid.412925.90000 0004 0400 6581National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Paulene A. Quinn
- grid.9918.90000 0004 1936 8411Department of Cardiovascular Sciences, Glenfield Hospital, University of Leicester, Leicester, UK ,grid.412925.90000 0004 0400 6581National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Luca Stefanucci
- grid.5335.00000000121885934Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK ,grid.436365.10000 0000 8685 6563NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK ,British Heart Foundation Centre of Excellence, Cambridge Biomedical Campus, Cambridge, UK
| | - Matthew C. Sims
- grid.5335.00000000121885934Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK ,grid.436365.10000 0000 8685 6563NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK ,grid.454382.c0000 0004 7871 7212Oxford Haemophilia and Thrombosis Centre, Oxford University Hospitals NHS Foundation Trust, NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Karola Rehnstrom
- grid.5335.00000000121885934Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Claire L. Adams
- grid.5335.00000000121885934Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ UK
| | - Amy Frary
- grid.5335.00000000121885934Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Bekir Ergüener
- grid.418729.10000 0004 0392 6802CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Roman Kreuzhuber
- grid.5335.00000000121885934Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK ,grid.225360.00000 0000 9709 7726European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton, UK
| | - Gabriele Mocciaro
- grid.5335.00000000121885934Department of Biochemistry and the Cambridge Systems Biology Centre, University of Cambridge, The Sanger Building, 80 Tennis Court Road, Cambridge, CB2 1GA UK
| | - Simona D’Amore
- grid.24029.3d0000 0004 0383 8386Addenbrooke’s Hospital, NIHR Cambridge Biomedical Research Centre, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK ,grid.7644.10000 0001 0120 3326Department of Medicine, Aldo Moro University of Bari, Piazza Giulio Cesare 11, 70124 Bari, Italy ,National Cancer Research Center, IRCCS Istituto Tumori ‘Giovanni Paolo II’, Viale Orazio Flacco, 65, 70124 Bari, Italy
| | - Albert Koulman
- grid.5335.00000000121885934MRC Epidemiology Unit, University of Cambridge, Cambridge, UK ,grid.415055.00000 0004 0606 2472MRC Elsie Widdowson Laboratory, Cambridge, UK ,grid.5335.00000000121885934National Institute for Health Research Biomedical Research Centres Core Nutritional Biomarker Laboratory, Addenbrooke’s Hospital, University of Cambridge, Cambridge, UK ,grid.5335.00000000121885934National Institute for Health Research Biomedical Research Centres Core Metabolomics and Lipidomics Laboratory, Addenbrooke’s Hospital, University of Cambridge, Cambridge, UK
| | - Luigi Grassi
- grid.24029.3d0000 0004 0383 8386National Institute for Health Research BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK ,grid.5335.00000000121885934Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK ,grid.436365.10000 0000 8685 6563NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK
| | - Julian L. Griffin
- grid.5335.00000000121885934Department of Biochemistry and the Cambridge Systems Biology Centre, University of Cambridge, The Sanger Building, 80 Tennis Court Road, Cambridge, CB2 1GA UK
| | - Leong Loke Ng
- grid.9918.90000 0004 1936 8411Department of Cardiovascular Sciences, Glenfield Hospital, University of Leicester, Leicester, UK ,grid.412925.90000 0004 0400 6581National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Adrian Park
- grid.24029.3d0000 0004 0383 8386Addenbrooke’s Hospital, NIHR Cambridge Biomedical Research Centre, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - David B. Savage
- grid.5335.00000000121885934Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ UK
| | - Claudia Langenberg
- grid.5335.00000000121885934MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Christoph Bock
- grid.418729.10000 0004 0392 6802CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria ,grid.511293.d0000 0004 6104 8403Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria ,grid.22937.3d0000 0000 9259 8492Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Kate Downes
- grid.24029.3d0000 0004 0383 8386National Institute for Health Research BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK ,grid.5335.00000000121885934Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK ,grid.24029.3d0000 0004 0383 8386East Midlands and East of England Genomic Laboratory Hub, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Nicholas J. Wareham
- grid.5335.00000000121885934MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Michael Allison
- grid.24029.3d0000 0004 0383 8386Addenbrooke’s Hospital, NIHR Cambridge Biomedical Research Centre, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Michele Vacca
- grid.5335.00000000121885934Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ UK ,grid.5335.00000000121885934Department of Biochemistry and the Cambridge Systems Biology Centre, University of Cambridge, The Sanger Building, 80 Tennis Court Road, Cambridge, CB2 1GA UK
| | - Paul D. W. Kirk
- grid.5335.00000000121885934MRC Biostatistics Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK ,grid.5335.00000000121885934Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge Biomedical Campus, Puddicombe Way, Cambridge, CB2 0AW UK
| | - Mattia Frontini
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK. .,NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK. .,British Heart Foundation Centre of Excellence, Cambridge Biomedical Campus, Cambridge, UK. .,Institute of Biomedical & Clinical Science, College of Medicine and Health, University of Exeter Medical School, RILD Building, Barrack Road, Exeter, EX2 5DW, UK.
| |
Collapse
|
31
|
Dempsey PC, Aadland E, Strain T, Kvalheim OM, Westgate K, Lindsay T, Khaw KT, Wareham NJ, Brage S, Wijndaele K. Physical activity intensity profiles associated with cardiometabolic risk in middle-aged to older men and women. Prev Med 2022; 156:106977. [PMID: 35131206 PMCID: PMC8907866 DOI: 10.1016/j.ypmed.2022.106977] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 01/20/2022] [Accepted: 01/29/2022] [Indexed: 11/16/2022]
Abstract
Accelerometers provide detailed data about physical activity (PA) across the full intensity spectrum. However, when examining associations with health, results are often aggregated to only a few summary measures [e.g. time spent "sedentary" or "moderate-to-vigorous" intensity PA]. Using multivariate pattern analysis, which can handle collinear exposure variables, we examined associations between the full PA intensity spectrum and cardiometabolic risk (CMR) in a population-based sample of middle-aged to older adults. Participants (n = 3660; mean ± SD age = 69 ± 8y and BMI = 26.7 ± 4.2 kg/m2; 55% female) from the EPIC-Norfolk study (UK) with valid accelerometry (ActiGraph-GT1M) data were included. We used multivariate pattern analysis with partial least squares regression to examine cross-sectional multivariate associations (r) across the full PA intensity spectrum [minutes/day at 0-5000 counts-per-minute (cpm); 5 s epoch] with a continuous CMR score (reflecting waist, blood pressure, lipid, and glucose metabolism). Models were sex-stratified and adjusted for potential confounders. There was a positive (detrimental) association between PA and CMR at 0-12 cpm (maximally-adjusted r = 0.08 (95%CI 0.06-0.10). PA was negatively (favourably) associated with CMR at all intensities above 13 cpm ranging between r = -0.09 (0.07-0.12) at 800-999 cpm and r = -0.14 (0.11-0.16) at 75-99 and 4000-4999 cpm. The strongest favourable associations were from 50 to 800 cpm (r = 0.10-0.12) in men, but from ≥2500 cpm (r = 0.18-0.20) in women; with higher proportions of model explained variance for women (R2 = 7.4% vs. 2.3%). Most of the PA intensity spectrum was beneficially associated with CMR in middle-aged to older adults, even at intensities lower than what has traditionally been considered "sedentary" or "light-intensity" activity. This supports encouragement of PA at almost any intensity in this age-group.
Collapse
Affiliation(s)
- Paddy C Dempsey
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK; Physical Activity & Behavioural Epidemiology Laboratories, Baker Heart & Diabetes Institute, Melbourne, Australia; Diabetes Research Centre, University of Leicester, Leicester General Hospital, Leicester, UK.
| | - Eivind Aadland
- Faculty of Education, Arts and Sports, Department of Sport, Food and Natural Sciences, Campus Sogndal, Western Norway University of Applied Sciences, Sogndal, Norway
| | - Tessa Strain
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Olav M Kvalheim
- Department of Chemistry, University of Bergen, Bergen, Norway
| | - Kate Westgate
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Tim Lindsay
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Kay-Tee Khaw
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Nicholas J Wareham
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Søren Brage
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Katrien Wijndaele
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| |
Collapse
|
32
|
Lindsay T, Wijndaele K, Westgate K, Dempsey P, Strain T, De Lucia Rolfe E, Forouhi NG, Griffin S, Wareham NJ, Brage S. Joint associations between objectively measured physical activity volume and intensity with body fatness: the Fenland study. Int J Obes (Lond) 2022; 46:169-177. [PMID: 34593963 PMCID: PMC8748201 DOI: 10.1038/s41366-021-00970-8] [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] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 08/16/2021] [Accepted: 09/14/2021] [Indexed: 02/02/2023]
Abstract
BACKGROUND/OBJECTIVES Physical activity energy expenditure (PAEE) represents the total volume of all physical activity. This can be accumulated as different underlying intensity profiles. Although volume and intensity have been studied in isolation, less is known about their joint association with health. We examined this association with body fatness in a population-based sample of middle-aged British adults. METHODS In total, 6148 women and 5320 men from the Fenland study with objectively measured physical activity from individually calibrated combined heart rate and movement sensing and DXA-derived body fat percentage (BF%) were included in the analyses. We used linear and compositional isocaloric substitution analysis to examine associations of PAEE and its intensity composition with body fatness. Sex-stratified models were adjusted for socio-economic and dietary covariates. RESULTS PAEE was inversely associated with body fatness in women (beta = -0.16 (95% CI: -0.17; -0.15) BF% per kJ day-1 kg-1) and men (beta = -0.09 (95% CI: -0.10; -0.08) BF% per kJ day-1 kg-1). Intensity composition was significantly associated with body fatness, beyond that of PAEE; the reallocation of energy to vigorous physical activity (>6 METs) from other intensities was associated with less body fatness, whereas light activity (1.5-3 METs) was positively associated. However, light activity was the main driver of overall PAEE volume, and the relative importance of intensity was marginal compared to that of volume; the difference between PAEE in tertile 1 and 2 in women was associated with 3 percentage-point lower BF%. Higher vigorous physical activity in the same group to the maximum observed value was associated with 1 percentage-point lower BF%. CONCLUSIONS In this large, population-based cohort study with objective measures, PAEE was inversely associated with body fatness. Beyond the PAEE association, greater levels of intense activity were also associated with lower body fatness. This contribution was marginal relative to PAEE. These findings support current guidelines for physical activity which emphasise that any movement is beneficial, rather than specific activity intensity or duration targets.
Collapse
Affiliation(s)
- Tim Lindsay
- MRC Epidemiology Unit, University of Cambridge, School of Clinical Medicine, Cambridge, UK
| | - Katrien Wijndaele
- MRC Epidemiology Unit, University of Cambridge, School of Clinical Medicine, Cambridge, UK
| | - Kate Westgate
- MRC Epidemiology Unit, University of Cambridge, School of Clinical Medicine, Cambridge, UK
| | - Paddy Dempsey
- MRC Epidemiology Unit, University of Cambridge, School of Clinical Medicine, Cambridge, UK
- Physical Activity and Behavioural Epidemiology Laboratories, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Diabetes Research Centre, University of Leicester, Leicester General Hospital, Leicester, UK
| | - Tessa Strain
- MRC Epidemiology Unit, University of Cambridge, School of Clinical Medicine, Cambridge, UK
| | | | - Nita G Forouhi
- MRC Epidemiology Unit, University of Cambridge, School of Clinical Medicine, Cambridge, UK
| | - Simon Griffin
- MRC Epidemiology Unit, University of Cambridge, School of Clinical Medicine, Cambridge, UK
| | - Nick J Wareham
- MRC Epidemiology Unit, University of Cambridge, School of Clinical Medicine, Cambridge, UK
| | - Søren Brage
- MRC Epidemiology Unit, University of Cambridge, School of Clinical Medicine, Cambridge, UK.
| |
Collapse
|
33
|
Porta M, Orrù PF, Pau M. Use of wearable sensors to assess patterns of trunk flexion in young and old workers in the Metalworking Industry. ERGONOMICS 2021; 64:1543-1554. [PMID: 34180361 DOI: 10.1080/00140139.2021.1948107] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 06/19/2021] [Indexed: 06/13/2023]
Abstract
Workers exposed to repeated trunk flexions are at risk of onset of low-back disorders and in individuals aged over 50 this issue is exacerbated by the physiologic decline of the musculoskeletal system and longer lifetime occupational exposure. In this study, we investigated the existence of possible age-related differences in patterns of trunk flexion of workers in the metalworking industry. Thirty-three subjects were monitored during an actual shift using a wearable Inertial Measurement Unit (IMU) to assess trunk flexion angles (i.e. between 30° and 60°, 60°-90° and > 90°). Results show that older workers spent less time with their trunk flexed, regardless of the class of flexion considered, with respect to their younger colleagues. Although further studies are necessary to clarify the existence of strategies aimed at optimising trunk movements during ageing, the IMU-based approach appears useful in highlighting potentially harmful conditions, especially in workers with marked signs of decline in their physical capacities. Practitioner summary: Wearable sensors, which are well tolerated and minimally intrusive, represent a valid option to continuously monitor trunk posture in workers employed in metalworking industry. The results of this study show that they provide valuable information about the patterns of flexion of young and old individuals engaged in physically demanding tasks.
Collapse
Affiliation(s)
- Micaela Porta
- Department of Mechanical, Chemical and Materials Engineering University of Cagliari, Cagliari, Italy
| | - Pier Francesco Orrù
- Department of Mechanical, Chemical and Materials Engineering University of Cagliari, Cagliari, Italy
| | - Massimiliano Pau
- Department of Mechanical, Chemical and Materials Engineering University of Cagliari, Cagliari, Italy
| |
Collapse
|
34
|
Pietzner M, Wheeler E, Carrasco-Zanini J, Kerrison ND, Oerton E, Koprulu M, Luan J, Hingorani AD, Williams SA, Wareham NJ, Langenberg C. Synergistic insights into human health from aptamer- and antibody-based proteomic profiling. Nat Commun 2021; 12:6822. [PMID: 34819519 PMCID: PMC8613205 DOI: 10.1038/s41467-021-27164-0] [Citation(s) in RCA: 113] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 11/03/2021] [Indexed: 01/09/2023] Open
Abstract
Affinity-based proteomics has enabled scalable quantification of thousands of protein targets in blood enhancing biomarker discovery, understanding of disease mechanisms, and genetic evaluation of drug targets in humans through protein quantitative trait loci (pQTLs). Here, we integrate two partly complementary techniques-the aptamer-based SomaScan® v4 assay and the antibody-based Olink assays-to systematically assess phenotypic consequences of hundreds of pQTLs discovered for 871 protein targets across both platforms. We create a genetically anchored cross-platform proteome-phenome network comprising 547 protein-phenotype connections, 36.3% of which were only seen with one of the two platforms suggesting that both techniques capture distinct aspects of protein biology. We further highlight discordance of genetically predicted effect directions between assays, such as for PILRA and Alzheimer's disease. Our results showcase the synergistic nature of these technologies to better understand and identify disease mechanisms and provide a benchmark for future cross-platform discoveries.
Collapse
Affiliation(s)
- Maik Pietzner
- grid.5335.00000000121885934MRC Epidemiology Unit, University of Cambridge, Cambridge, UK ,grid.6363.00000 0001 2218 4662Computational Medicine, Berlin Institute of Health (BIH) at Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Eleanor Wheeler
- grid.5335.00000000121885934MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Julia Carrasco-Zanini
- grid.5335.00000000121885934MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Nicola D. Kerrison
- grid.5335.00000000121885934MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Erin Oerton
- grid.5335.00000000121885934MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Mine Koprulu
- grid.5335.00000000121885934MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Jian’an Luan
- grid.5335.00000000121885934MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Aroon D. Hingorani
- grid.83440.3b0000000121901201Institute of Cardiovascular Science, Faculty of Population Health, University College London, London, WC1E 6BT UK ,grid.83440.3b0000000121901201UCL BHF Research Accelerator Centre, London, UK ,grid.507332.0Health Data Research UK, London, UK
| | | | - Nicholas J. Wareham
- grid.5335.00000000121885934MRC Epidemiology Unit, University of Cambridge, Cambridge, UK ,grid.507332.0Health Data Research UK, London, UK
| | - Claudia Langenberg
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK. .,Computational Medicine, Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, Berlin, Germany. .,Health Data Research UK, London, UK.
| |
Collapse
|
35
|
Pietzner M, Wheeler E, Carrasco-Zanini J, Cortes A, Koprulu M, Wörheide MA, Oerton E, Cook J, Stewart ID, Kerrison ND, Luan J, Raffler J, Arnold M, Arlt W, O’Rahilly S, Kastenmüller G, Gamazon ER, Hingorani AD, Scott RA, Wareham NJ, Langenberg C. Mapping the proteo-genomic convergence of human diseases. Science 2021; 374:eabj1541. [PMID: 34648354 PMCID: PMC9904207 DOI: 10.1126/science.abj1541] [Citation(s) in RCA: 269] [Impact Index Per Article: 67.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Characterization of the genetic regulation of proteins is essential for understanding disease etiology and developing therapies. We identified 10,674 genetic associations for 3892 plasma proteins to create a cis-anchored gene-protein-disease map of 1859 connections that highlights strong cross-disease biological convergence. This proteo-genomic map provides a framework to connect etiologically related diseases, to provide biological context for new or emerging disorders, and to integrate different biological domains to establish mechanisms for known gene-disease links. Our results identify proteo-genomic connections within and between diseases and establish the value of cis-protein variants for annotation of likely causal disease genes at loci identified in genome-wide association studies, thereby addressing a major barrier to experimental validation and clinical translation of genetic discoveries.
Collapse
Affiliation(s)
- Maik Pietzner
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK,Computational Medicine, Berlin Institute of Health (BIH) at Charité – Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Eleanor Wheeler
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | - Julia Carrasco-Zanini
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | | | - Mine Koprulu
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | - Maria A. Wörheide
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Erin Oerton
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | - James Cook
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | - Isobel D. Stewart
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | - Nicola D. Kerrison
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | - Jian’an Luan
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | - Johannes Raffler
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany,Institut für Digitale Medizin, Universitätsklinikum Augsburg, 86156 Augsburg, Germany
| | - Matthias Arnold
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany,Department of Psychiatry and Behavioural Sciences, Duke University, Durham, NC 27710, USA
| | - Wiebke Arlt
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Stephen O’Rahilly
- MRC Metabolic Diseases Unit, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Gabi Kastenmüller
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany,German Centre for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Eric R. Gamazon
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN 37203, USA,Clare Hall, University of Cambridge, Cambridge CB3 9AL, United Kingdom
| | - Aroon D. Hingorani
- UCL British Heart Foundation Research Accelerator, Institute of Cardiovascular Science, University College London, WC1E 6BT, UK.,Health Data Research UK, Gibbs Building, 215 Euston Road, London NW1 2BE, UK,Institute of Health Informatics, University College London, 222 Euston Road, London NW1 2DA, UK
| | | | - Nicholas J. Wareham
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK,Health Data Research UK, Gibbs Building, 215 Euston Road, London NW1 2BE, UK
| | - Claudia Langenberg
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK,Computational Medicine, Berlin Institute of Health (BIH) at Charité – Universitätsmedizin Berlin, 10117 Berlin, Germany,Health Data Research UK, Gibbs Building, 215 Euston Road, London NW1 2BE, UK,Correspondence to Dr. Claudia Langenberg ()
| |
Collapse
|
36
|
Lam BYH, Williamson A, Finer S, Day FR, Tadross JA, Gonçalves Soares A, Wade K, Sweeney P, Bedenbaugh MN, Porter DT, Melvin A, Ellacott KLJ, Lippert RN, Buller S, Rosmaninho-Salgado J, Dowsett GKC, Ridley KE, Xu Z, Cimino I, Rimmington D, Rainbow K, Duckett K, Holmqvist S, Khan A, Dai X, Bochukova EG, Trembath RC, Martin HC, Coll AP, Rowitch DH, Wareham NJ, van Heel DA, Timpson N, Simerly RB, Ong KK, Cone RD, Langenberg C, Perry JRB, Yeo GS, O'Rahilly S. MC3R links nutritional state to childhood growth and the timing of puberty. Nature 2021; 599:436-441. [PMID: 34732894 PMCID: PMC8819628 DOI: 10.1038/s41586-021-04088-9] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 10/01/2021] [Indexed: 02/02/2023]
Abstract
The state of somatic energy stores in metazoans is communicated to the brain, which regulates key aspects of behaviour, growth, nutrient partitioning and development1. The central melanocortin system acts through melanocortin 4 receptor (MC4R) to control appetite, food intake and energy expenditure2. Here we present evidence that MC3R regulates the timing of sexual maturation, the rate of linear growth and the accrual of lean mass, which are all energy-sensitive processes. We found that humans who carry loss-of-function mutations in MC3R, including a rare homozygote individual, have a later onset of puberty. Consistent with previous findings in mice, they also had reduced linear growth, lean mass and circulating levels of IGF1. Mice lacking Mc3r had delayed sexual maturation and an insensitivity of reproductive cycle length to nutritional perturbation. The expression of Mc3r is enriched in hypothalamic neurons that control reproduction and growth, and expression increases during postnatal development in a manner that is consistent with a role in the regulation of sexual maturation. These findings suggest a bifurcating model of nutrient sensing by the central melanocortin pathway with signalling through MC4R controlling the acquisition and retention of calories, whereas signalling through MC3R primarily regulates the disposition of calories into growth, lean mass and the timing of sexual maturation.
Collapse
Affiliation(s)
- B Y H Lam
- MRC Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - A Williamson
- MRC Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- NIHR Cambridge Biomedical Research Centre, Cambridge, UK
- MRC Epidemiology Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - S Finer
- Wolfson Institute of Population Health, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - F R Day
- MRC Epidemiology Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - J A Tadross
- MRC Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- NIHR Cambridge Biomedical Research Centre, Cambridge, UK
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - A Gonçalves Soares
- MRC Integrative Epidemiology Unit and Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - K Wade
- MRC Integrative Epidemiology Unit and Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - P Sweeney
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | - M N Bedenbaugh
- Department of Molecular Physiology and Biophysics, School of Medicine, Vanderbilt University, Nashville, TN, USA
| | - D T Porter
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | - A Melvin
- MRC Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - K L J Ellacott
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Exeter, UK
| | - R N Lippert
- Department of Neurocircuit Development and Function, German Institute of Human Nutrition, Potsdam, Germany
| | - S Buller
- MRC Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - J Rosmaninho-Salgado
- Medical Genetics Unit, Hospital Pediátrico, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - G K C Dowsett
- MRC Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - K E Ridley
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - Z Xu
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - I Cimino
- MRC Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - D Rimmington
- MRC Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - K Rainbow
- MRC Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - K Duckett
- MRC Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - S Holmqvist
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - A Khan
- Wolfson Institute of Population Health, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - X Dai
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, London, UK
| | - E G Bochukova
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, London, UK
| | - R C Trembath
- School of Basic and Medical Biosciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - H C Martin
- Wellcome Sanger Institute, Hinxton, Cambridge, UK
| | - A P Coll
- MRC Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - D H Rowitch
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - N J Wareham
- MRC Epidemiology Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - D A van Heel
- Wolfson Institute of Population Health, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, London, UK
| | - N Timpson
- MRC Integrative Epidemiology Unit and Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - R B Simerly
- Department of Molecular Physiology and Biophysics, School of Medicine, Vanderbilt University, Nashville, TN, USA
| | - K K Ong
- MRC Epidemiology Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - R D Cone
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Molecular and Integrative Physiology, School of Medicine, University of Michigan, Ann Arbor, MI, USA
| | - C Langenberg
- MRC Epidemiology Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Computational Medicine, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - J R B Perry
- MRC Epidemiology Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - G S Yeo
- MRC Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - S O'Rahilly
- MRC Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
- NIHR Cambridge Biomedical Research Centre, Cambridge, UK.
| |
Collapse
|
37
|
Kyriakidou Y, Cooper I, Kraev I, Lange S, Elliott BT. Preliminary Investigations Into the Effect of Exercise-Induced Muscle Damage on Systemic Extracellular Vesicle Release in Trained Younger and Older Men. Front Physiol 2021; 12:723931. [PMID: 34650440 PMCID: PMC8507150 DOI: 10.3389/fphys.2021.723931] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 07/31/2021] [Indexed: 01/11/2023] Open
Abstract
Background: Exercise-induced muscle damage (EIMD) results in transient muscle inflammation, strength loss, and muscle soreness and may cause subsequent exercise avoidance. Research has recently proven that skeletal muscle can also release extracellular vesicles (EVs) into the circulation following a bout of exercise. However, EV’s potential role, including as a biomarker, in the response to eccentric resistance exercise stimulus remains unclear. Methods: Twelve (younger, n=7, 27.0±1.5years and older, n=5, 63.0±1.0years) healthy, physically active males, undertaking moderate, regular physical activity (3–5 times per week) performed a unilateral high intensity eccentric exercise protocol. Venous plasma was collected for assessment of EVs and creatine kinase (CK) prior to EIMD, immediately after EIMD, and 1–72h post-EIMD, and maximal voluntary isometric contraction (MVIC) and delayed onset muscle soreness (DOMS) were assessed at all time points, except 1 and 2h post-EIMD. Results: A significant effect of both time (p=0.005) and group (p<0.001) was noted for MVIC, with younger participants’ MVIC being higher throughout. Whilst a significant increase was observed in DOMS in the younger group (p=0.014) and in the older group (p=0.034) following EIMD, no significant differences were observed between groups. CK was not different between age groups but was altered following the EIMD (main effect of time p=0.026), with increased CK seen immediately post-, at 1 and 2h post-EIMD. EV count tended to be lower in older participants at rest, relative to younger participants (p=0.056), whilst EV modal size did not differ between younger and older participants pre-EIMD. EIMD did not substantially alter EV modal size or EV count in younger or older participants; however, the alteration in EV concentration (ΔCount) and EV modal size (ΔMode) between post-EIMD and pre-EIMD negatively associated with CK activity. No significant associations were noted between MVIC or DOMS and either ΔCount or ΔMode of EVs at any time point. Conclusion: These findings suggest that profile of EV release, immediately following exercise, may predict later CK release and play a role in the EIMD response. Exercise-induced EV release profiles may therefore serve as an indicator for subsequent muscle damage.
Collapse
Affiliation(s)
- Yvoni Kyriakidou
- Translational Physiology Research Group, School of Life Sciences, University of Westminster, London, United Kingdom
| | - Isabella Cooper
- Translational Physiology Research Group, School of Life Sciences, University of Westminster, London, United Kingdom
| | - Igor Kraev
- Electron Microscopy Suite, Faculty of Science, Technology, Engineering and Mathematics, Open University, Milton Keynes, United Kingdom
| | - Sigrun Lange
- Tissue Architecture and Regeneration Research Group, School of Life Sciences, University of Westminster, London, United Kingdom
| | - Bradley T Elliott
- Translational Physiology Research Group, School of Life Sciences, University of Westminster, London, United Kingdom
| |
Collapse
|
38
|
Gonzales TI, Westgate K, Strain T, Hollidge S, Jeon J, Christensen DL, Jensen J, Wareham NJ, Brage S. Cardiorespiratory fitness assessment using risk-stratified exercise testing and dose-response relationships with disease outcomes. Sci Rep 2021; 11:15315. [PMID: 34321526 PMCID: PMC8319417 DOI: 10.1038/s41598-021-94768-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 07/06/2021] [Indexed: 02/07/2023] Open
Abstract
Cardiorespiratory fitness (CRF) is associated with mortality and cardiovascular disease, but assessing CRF in the population is challenging. Here we develop and validate a novel framework to estimate CRF (as maximal oxygen consumption, VO2max) from heart rate response to low-risk personalised exercise tests. We apply the method to examine associations between CRF and health outcomes in the UK Biobank study, one of the world's largest and most inclusive studies of CRF, showing that risk of all-cause mortality is 8% lower (95%CI 5-11%, 2670 deaths among 79,981 participants) and cardiovascular mortality is 9% lower (95%CI 4-14%, 854 deaths) per 1-metabolic equivalent difference in CRF. Associations obtained with the novel validated CRF estimation method are stronger than those obtained using previous methodology, suggesting previous methods may have underestimated the importance of fitness for human health.
Collapse
Affiliation(s)
- Tomas I Gonzales
- MRC Epidemiology Unit, School of Clinical Medicine, Institute of Metabolic Science, University of Cambridge, Cambridge Biomedical Campus, Box 285, Cambridge, CB2 0QQ, UK
| | - Kate Westgate
- MRC Epidemiology Unit, School of Clinical Medicine, Institute of Metabolic Science, University of Cambridge, Cambridge Biomedical Campus, Box 285, Cambridge, CB2 0QQ, UK
| | - Tessa Strain
- MRC Epidemiology Unit, School of Clinical Medicine, Institute of Metabolic Science, University of Cambridge, Cambridge Biomedical Campus, Box 285, Cambridge, CB2 0QQ, UK
| | - Stefanie Hollidge
- MRC Epidemiology Unit, School of Clinical Medicine, Institute of Metabolic Science, University of Cambridge, Cambridge Biomedical Campus, Box 285, Cambridge, CB2 0QQ, UK
| | - Justin Jeon
- MRC Epidemiology Unit, School of Clinical Medicine, Institute of Metabolic Science, University of Cambridge, Cambridge Biomedical Campus, Box 285, Cambridge, CB2 0QQ, UK
- Yonsei University, Seoul, Republic of Korea
| | - Dirk L Christensen
- MRC Epidemiology Unit, School of Clinical Medicine, Institute of Metabolic Science, University of Cambridge, Cambridge Biomedical Campus, Box 285, Cambridge, CB2 0QQ, UK
- University of Copenhagen, Copenhagen, Denmark
| | - Jorgen Jensen
- MRC Epidemiology Unit, School of Clinical Medicine, Institute of Metabolic Science, University of Cambridge, Cambridge Biomedical Campus, Box 285, Cambridge, CB2 0QQ, UK
- Department Physical Performance, Norwegian School of Sport Sciences, Oslo, Norway
| | - Nicholas J Wareham
- MRC Epidemiology Unit, School of Clinical Medicine, Institute of Metabolic Science, University of Cambridge, Cambridge Biomedical Campus, Box 285, Cambridge, CB2 0QQ, UK
| | - Søren Brage
- MRC Epidemiology Unit, School of Clinical Medicine, Institute of Metabolic Science, University of Cambridge, Cambridge Biomedical Campus, Box 285, Cambridge, CB2 0QQ, UK.
| |
Collapse
|
39
|
Freyberg J, Brage S, Kessing LV, Faurholt-Jepsen M. The association between self-reported physical activity and objective measures of physical activity in participants with newly diagnosed bipolar disorder, unaffected relatives, and healthy individuals. Nord J Psychiatry 2021; 75:186-193. [PMID: 33779478 PMCID: PMC7610645 DOI: 10.1080/08039488.2020.1831063] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
BACKGROUND The association between the International Physical Activity Questionnaire Short Form (IPAQ-SF) and objective measures of physical activity has never been evaluated in participants with newly diagnosed bipolar disorder (BD). Our aim was to compare IPAQ-SF to objective measures in participants with newly diagnosed BD, their unaffected first-degree relatives (UR), and healthy control individuals (HC) in groups combined and stratified by group. MATERIALS AND METHODS Physical activity measurements were collected on 20 participants with newly diagnosed BD, 20 of their UR, and 20 HC using individually calibrated combined acceleration and heart rate sensing (Actiheart) for seven days. IPAQ-SF was self-completed at baseline. Correlation between measurements from the two methods was examined with Spearman rank correlation coefficient and agreement levels examined with modified Bland-Altman plots. RESULTS Physical activity energy expenditure (PAEE) from IPAQ-SF was weakly but significantly positively correlated with physical activity estimates measured using acceleration and heart rate in groups combined (Actiheart PAEE) (ρ= 0.301, p = 0.02). Correlations for each group were positive, but only in UR were it statistically significant (BD: p = 0.18, UR: p = 0.007, HC: p = 0.84). Self-reported PAEE and moderate-intensity were markedly underestimated [PAEE in all participants combined: 62.7 (Actiheart) vs. 24.3 kJ/day/kg (IPAQ-SF), p < 0.001], while vigorous-intensity was overestimated. Bland-Altman plots indicated proportional bias. CONCLUSION These results suggest that the use of the IPAQ-SF to monitor levels of physical activity in participants with newly diagnosed BD, in a psychiatric clinical setting, should be used with caution and consideration.
Collapse
Affiliation(s)
- Josefine Freyberg
- The Copenhagen Affective Disorder research Center (CADIC), Psychiatric Center Copenhagen, Rigshospitalet, Copenhagen, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Søren Brage
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Lars Vedel Kessing
- The Copenhagen Affective Disorder research Center (CADIC), Psychiatric Center Copenhagen, Rigshospitalet, Copenhagen, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Maria Faurholt-Jepsen
- The Copenhagen Affective Disorder research Center (CADIC), Psychiatric Center Copenhagen, Rigshospitalet, Copenhagen, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
40
|
Lotta LA, Pietzner M, Stewart ID, Wittemans LBL, Li C, Bonelli R, Raffler J, Biggs EK, Oliver-Williams C, Auyeung VPW, Luan J, Wheeler E, Paige E, Surendran P, Michelotti GA, Scott RA, Burgess S, Zuber V, Sanderson E, Koulman A, Imamura F, Forouhi NG, Khaw KT, Griffin JL, Wood AM, Kastenmüller G, Danesh J, Butterworth AS, Gribble FM, Reimann F, Bahlo M, Fauman E, Wareham NJ, Langenberg C. A cross-platform approach identifies genetic regulators of human metabolism and health. Nat Genet 2021; 53:54-64. [PMID: 33414548 PMCID: PMC7612925 DOI: 10.1038/s41588-020-00751-5] [Citation(s) in RCA: 120] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 11/20/2020] [Indexed: 02/02/2023]
Abstract
In cross-platform analyses of 174 metabolites, we identify 499 associations (P < 4.9 × 10-10) characterized by pleiotropy, allelic heterogeneity, large and nonlinear effects and enrichment for nonsynonymous variation. We identify a signal at GLP2R (p.Asp470Asn) shared among higher citrulline levels, body mass index, fasting glucose-dependent insulinotropic peptide and type 2 diabetes, with β-arrestin signaling as the underlying mechanism. Genetically higher serine levels are shown to reduce the likelihood (by 95%) and predict development of macular telangiectasia type 2, a rare degenerative retinal disease. Integration of genomic and small molecule data across platforms enables the discovery of regulators of human metabolism and translation into clinical insights.
Collapse
Affiliation(s)
- Luca A Lotta
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Maik Pietzner
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | | | - Laura B L Wittemans
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
- The Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
| | - Chen Li
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Roberto Bonelli
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Johannes Raffler
- Institute of Computational Biology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Emma K Biggs
- Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Clare Oliver-Williams
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Homerton College, University of Cambridge, Cambridge, UK
| | | | - Jian'an Luan
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Eleanor Wheeler
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Ellie Paige
- National Centre for Epidemiology and Population Health, The Australian National University, Canberra, Australia
| | - Praveen Surendran
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
- Rutherford Fund Fellow, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | | | - Robert A Scott
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Stephen Burgess
- MRC Biostatistics Unit, University of Cambridge, Cambridge, UK
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Verena Zuber
- MRC Biostatistics Unit, University of Cambridge, Cambridge, UK
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
| | - Eleanor Sanderson
- MRC Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, Bristol, UK
| | - Albert Koulman
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
- Institute of Computational Biology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
- NIHR BRC Nutritional Biomarker Laboratory, University of Cambridge, Cambridge, UK
| | - Fumiaki Imamura
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Nita G Forouhi
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Kay-Tee Khaw
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Julian L Griffin
- Biomolecular Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Angela M Wood
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
- National Institute for Health Research Blood and Transplant Research Unit in Donor Health and Genomics, University of Cambridge, Cambridge, UK
- The Alan Turing Institute, London, UK
| | - Gabi Kastenmüller
- Institute of Computational Biology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - John Danesh
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
- National Institute for Health Research Blood and Transplant Research Unit in Donor Health and Genomics, University of Cambridge, Cambridge, UK
- Cambridge Biomedical Research Centre, National Institute for Health Research, University of Cambridge and Cambridge University Hospitals, Cambridge, UK
- Department of Human Genetics, Wellcome Sanger Institute, Hinxton, UK
| | - Adam S Butterworth
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
- National Institute for Health Research Blood and Transplant Research Unit in Donor Health and Genomics, University of Cambridge, Cambridge, UK
- Cambridge Biomedical Research Centre, National Institute for Health Research, University of Cambridge and Cambridge University Hospitals, Cambridge, UK
- Department of Human Genetics, Wellcome Sanger Institute, Hinxton, UK
| | - Fiona M Gribble
- Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Frank Reimann
- Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Melanie Bahlo
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Eric Fauman
- Internal Medicine Research Unit, Pfizer Worldwide Research, Cambridge, MA, USA
| | | | - Claudia Langenberg
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK.
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK.
- Computational Medicine, Berlin Institute of Health (BIH), Charité University Medicine, Berlin, Germany.
| |
Collapse
|
41
|
Strain T, Wijndaele K, Garcia L, Cowan M, Guthold R, Brage S, Bull FC. Levels of domain-specific physical activity at work, in the household, for travel and for leisure among 327 789 adults from 104 countries. Br J Sports Med 2020; 54:1488-1497. [PMID: 33239355 PMCID: PMC7719912 DOI: 10.1136/bjsports-2020-102601] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/12/2020] [Indexed: 02/07/2023]
Abstract
OBJECTIVE To compare the country-level absolute and relative contributions of physical activity at work and in the household, for travel, and during leisure-time to total moderate-to-vigorous physical activity (MVPA). METHODS We used data collected between 2002 and 2019 from 327 789 participants across 104 countries and territories (n=24 low, n=34 lower-middle, n=30 upper-middle, n=16 high-income) from all six World Health Organization (WHO) regions. We calculated mean min/week of work/household, travel and leisure MVPA and compared their relative contributions to total MVPA using Global Physical Activity Questionnaire data. We compared patterns by country, sex and age group (25-44 and 45-64 years). RESULTS Mean MVPA in work/household, travel and leisure domains across the 104 countries was 950 (IQR 618-1198), 327 (190-405) and 104 (51-131) min/week, respectively. Corresponding relative contributions to total MVPA were 52% (IQR 44%-63%), 36% (25%-45%) and 12% (4%-15%), respectively. Work/household was the highest contributor in 80 countries; travel in 23; leisure in just one. In both absolute and relative terms, low-income countries tended to show higher work/household (1233 min/week, 57%) and lower leisure MVPA levels (72 min/week, 4%). Travel MVPA duration was higher in low-income countries but there was no obvious pattern in the relative contributions. Women tended to have relatively less work/household and more travel MVPA; age groups were generally similar. CONCLUSION In the largest domain-specific physical activity study to date, we found considerable country-level variation in how MVPA is accumulated. Such information is essential to inform national and global policy and future investments to provide opportunities to be active, accounting for country context.
Collapse
Affiliation(s)
- Tessa Strain
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
- Physical Activity for Health Research Centre, University of Edinburgh, Edinburgh, UK
| | | | - Leandro Garcia
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
- Centre for Public Health, Queen's University Belfast, Belfast, UK
| | - Melanie Cowan
- Department of Noncommunicable Diseases, World Health Organization, Geneva, Switzerland
| | - Regina Guthold
- Maternal, Newborn, Child, and Adolescent Health and Ageing Department, World Health Organization, Geneva, Switzerland
| | - Soren Brage
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Fiona C Bull
- Department of Health Promotion, Division of Universal Health Coverage and Healthier Populations, World Health Organization, Geneva, Switzerland
- Physical Activity Unit, The University of Western Australia, Perth, Western Australia, Australia
| |
Collapse
|
42
|
Brage S, Assah F, Msyamboza KP. Quantifying population levels of physical activity in Africa using wearable sensors: implications for global physical activity surveillance. BMJ Open Sport Exerc Med 2020; 6:e000941. [PMID: 33868705 PMCID: PMC8008991 DOI: 10.1136/bmjsem-2020-000941] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 09/16/2020] [Accepted: 09/16/2020] [Indexed: 02/02/2023] Open
Affiliation(s)
- Soren Brage
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Felix Assah
- Department of Public Health, Faculty of Medicine and Biomedical Science, University of Yaounde I, Yaounde, Cameroon
| | | |
Collapse
|
43
|
Strain T, Wijndaele K, Dempsey PC, Sharp SJ, Pearce M, Jeon J, Lindsay T, Wareham N, Brage S. Wearable-device-measured physical activity and future health risk. Nat Med 2020; 26:1385-1391. [PMID: 32807930 PMCID: PMC7116559 DOI: 10.1038/s41591-020-1012-3] [Citation(s) in RCA: 183] [Impact Index Per Article: 36.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 07/07/2020] [Indexed: 02/02/2023]
Abstract
Use of wearable devices that monitor physical activity is projected to increase more than fivefold per half-decade1. We investigated how device-based physical activity energy expenditure (PAEE) and different intensity profiles were associated with all-cause mortality. We used a network harmonization approach to map dominant-wrist acceleration to PAEE in 96,476 UK Biobank participants (mean age 62 years, 56% female). We also calculated the fraction of PAEE accumulated from moderate-to-vigorous-intensity physical activity (MVPA). Over the median 3.1-year follow-up period (302,526 person-years), 732 deaths were recorded. Higher PAEE was associated with a lower hazard of all-cause mortality for a constant fraction of MVPA (for example, 21% (95% confidence interval 4-35%) lower hazard for 20 versus 15 kJ kg-1 d-1 PAEE with 10% from MVPA). Similarly, a higher MVPA fraction was associated with a lower hazard when PAEE remained constant (for example, 30% (8-47%) lower hazard when 20% versus 10% of a fixed 15 kJ kg-1 d-1 PAEE volume was from MVPA). Our results show that higher volumes of PAEE are associated with reduced mortality rates, and achieving the same volume through higher-intensity activity is associated with greater reductions than through lower-intensity activity. The linkage of device-measured activity to energy expenditure creates a framework for using wearables for personalized prevention.
Collapse
Affiliation(s)
- Tessa Strain
- MRC Epidemiology Unit, University of Cambridge, University of
Cambridge, Institute of Metabolic Science, Cambridge
| | - Katrien Wijndaele
- MRC Epidemiology Unit, University of Cambridge, University of
Cambridge, Institute of Metabolic Science, Cambridge
| | - Paddy C. Dempsey
- MRC Epidemiology Unit, University of Cambridge, University of
Cambridge, Institute of Metabolic Science, Cambridge
- Physical Activity & Behavioural Epidemiology Laboratories,
Baker Heart & Diabetes Institute, Melbourne, Australia
| | - Stephen J. Sharp
- MRC Epidemiology Unit, University of Cambridge, University of
Cambridge, Institute of Metabolic Science, Cambridge
| | - Matthew Pearce
- MRC Epidemiology Unit, University of Cambridge, University of
Cambridge, Institute of Metabolic Science, Cambridge
| | - Justin Jeon
- MRC Epidemiology Unit, University of Cambridge, University of
Cambridge, Institute of Metabolic Science, Cambridge
- Department of Sport Industry Studies, Exercise Medicine Center for
Diabetes and Cancer Patients (ICONS), Yonsei University South Korea
| | - Tim Lindsay
- MRC Epidemiology Unit, University of Cambridge, University of
Cambridge, Institute of Metabolic Science, Cambridge
| | - Nick Wareham
- MRC Epidemiology Unit, University of Cambridge, University of
Cambridge, Institute of Metabolic Science, Cambridge
| | - Søren Brage
- MRC Epidemiology Unit, University of Cambridge, University of
Cambridge, Institute of Metabolic Science, Cambridge
| |
Collapse
|
44
|
Brage S, Lindsay T, Venables M, Wijndaele K, Westgate K, Collins D, Roberts C, Bluck L, Wareham N, Page P. Descriptive epidemiology of energy expenditure in the UK: findings from the National Diet and Nutrition Survey 2008-15. Int J Epidemiol 2020; 49:1007-1021. [PMID: 32191299 PMCID: PMC7394951 DOI: 10.1093/ije/dyaa005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/13/2020] [Indexed: 12/27/2022] Open
Abstract
Background Little is known about population levels of energy expenditure, as national surveillance systems typically employ only crude measures. The National Diet and Nutrition Survey (NDNS) in the UK measured energy expenditure in a 10% subsample by gold-standard doubly labelled water (DLW). Methods DLW-subsample participants from the NDNS (383 males, 387 females) aged 4–91 years were recruited between 2008 and 2015 (rolling programme). Height and weight were measured and body-fat percentage estimated by deuterium dilution. Results Absolute total energy expenditure (TEE) increased steadily throughout childhood, ranging from 6.2 and 7.2 MJ/day in 4- to 7-year-olds to 9.7 and 11.7 MJ/day for 14- to 16-year-old girls and boys, respectively. TEE peaked in 17- to 27-year-old women (10.7 MJ/day) and 28- to 43-year-old men (14.4 MJ/day), before decreasing gradually in old age. Physical-activity energy expenditure (PAEE) declined steadily with age from childhood (87 kJ/day/kg in 4- to 7-year-olds) through to old age (38 kJ/day/kg in 71- to 91-year-olds). No differences were observed by time, region and macronutrient composition. Body-fat percentage was strongly inversely associated with PAEE throughout life, irrespective of expressing PAEE relative to body mass or fat-free mass. Compared with females with <30% body fat, females with >40% recorded 29 kJ/day/kg body mass and 18 kJ/day/kg fat-free mass less PAEE in analyses adjusted for age, geographical region and time of assessment. Similarly, compared with males with <25% body fat, males with >35% recorded 26 kJ/day/kg body mass and 10 kJ/day/kg fat-free mass less PAEE. Conclusions This first nationally representative study reports levels of human-energy expenditure as measured by gold-standard methodology; values may serve as a reference for other population studies. Age, sex and body composition are the main determinants of energy expenditure. Key Messages This is the first nationally representative study of human energy expenditure, covering the UK in the period 2008-2015. Total energy expenditure (MJ/day) increases steadily with age throughout childhood and adolescence, peaks in the 3rd decade of life in women and 4th decade of life in men, before decreasing gradually in old age. Physical activity energy expenditure (kJ/day/kg or kJ/day/kg fat-free mass) declines steadily with age from childhood to old age, more steeply so in males. Body-fat percentage is strongly inversely associated with physical activity energy expenditure. We found little evidence that energy expenditure varied by geographical region, over time, or by dietary macronutrient composition.
Collapse
Affiliation(s)
- Soren Brage
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Tim Lindsay
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | | | | | - Kate Westgate
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - David Collins
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Caireen Roberts
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Les Bluck
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Nick Wareham
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Polly Page
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| |
Collapse
|
45
|
Grimaldi-Puyana M, Fernández-Batanero JM, Fennell C, Sañudo B. Associations of Objectively-Assessed Smartphone Use with Physical Activity, Sedentary Behavior, Mood, and Sleep Quality in Young Adults: A Cross-Sectional Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:E3499. [PMID: 32429550 PMCID: PMC7277080 DOI: 10.3390/ijerph17103499] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 05/09/2020] [Accepted: 05/14/2020] [Indexed: 12/11/2022]
Abstract
This study assesses the associations of objectively-measured smartphone time with physical activity, sedentary behavior, mood, and sleep patterns among young adults by collecting real-time data of the smartphone screen-state. The sample consisted of 306 college-aged students (mean age ± SD: 20.7 ± 1.4 years; 60% males). Over seven days of time, the following variables were measured in the participants: objectively-measured smartphone use (Your Hour and Screen Time applications), objective and subjective physical activity (GoogleFit and Apple Health applications, and the International Physical Activity Questionnaire (IPAQ), respectively), the number of hours sitting (IPAQ), mood (The Profile of Mood State (POMS)), and sleep (The Pittsburgh Sleep Quality Index (PSQI)). Multiple regressions analyses showed that the number of hours sitting per day, physical activity, and the POMS Global Score significantly predicted smartphone use (adj.R2 = 0.15). Further, participants with low levels of physical activity were more likely to increase the use of smartphones (OR = 2.981). Moreover, mood state (β = 0.185; 95% CI = 0.05, 0.32) and sleep quality (β = 0.076; 95% CI = -0.06, 0.21) predicted smartphone use, with those reporting poor quality of sleep (PSQI index >5) being more likely to use the smartphone (OR = 2.679). In conclusion, there is an association between objectively-measured smartphone use and physical activity, sedentary behavior, mood, and sleep patterns. Those participants with low levels of physical activity, high levels of sedentary behavior, poor mood state, and poor sleep quality were more likely to spend more time using their smartphones.
Collapse
Affiliation(s)
- Moisés Grimaldi-Puyana
- Department of Physical Education and Sports, Faculty of Educational Sciences, Universidad de Sevilla, 41013 Seville, Spain;
| | | | - Curtis Fennell
- Department of Exercise and Nutrition Science, University of Montevallo, Montevallo, AL 35115, USA;
| | - Borja Sañudo
- Department of Physical Education and Sports, Faculty of Educational Sciences, Universidad de Sevilla, 41013 Seville, Spain;
| |
Collapse
|
46
|
Hamer M, Stamatakis E. The descriptive epidemiology of standing activity during free-living in 5412 middle-aged adults: the 1970 British Cohort Study. J Epidemiol Community Health 2020; 74:757-760. [PMID: 32350124 DOI: 10.1136/jech-2020-213783] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 03/28/2020] [Accepted: 04/14/2020] [Indexed: 12/31/2022]
Abstract
BACKGROUND Standing is often classified as light-intensity physical activity, with potential health benefits compared with sitting. Standing is, however, rarely captured as an independent activity. To better understand free-living standing behaviour at a population level, we incorporated a gold standard postural allocation technique into a national cohort study. METHODS Participants (n=5412, aged 46.8±0.7 years) from the 1970 British Cohort Study were fitted with a water-proofed thigh-mounted accelerometer device (activPAL3 micro) worn 24 hours continuously over 7 days (90.7% provided at least 3 full days). We examined the correlates of free-living standing during waking hours. RESULTS Total daily standing time averaged 4.6±1.5 h/d, accounting for 29% of waking hours, which was largely (98.7%) accumulated in bouts lasting less than 30 min. In mutually adjusted models, male sex, obesity, diabetes, professional occupation, poor self-rated health and disability were associated with lower device-measured standing times. CONCLUSION Middle-aged people in Britain spent a surprisingly large proportion of the day in activities involving standing. Standing merits attention as a health-related posture and may represent a potential target for public health intervention.
Collapse
Affiliation(s)
- Mark Hamer
- Institute Sport Exercise & Health, Division Surgery Interventional Science, University College London, London, UK
| | - Emmanuel Stamatakis
- Faculty of Medicine and Health, School of Health Sciences, The University of Sydney, Sydney, Australia
| |
Collapse
|
47
|
Freyberg J, Brage S, Kessing LV, Faurholt-Jepsen M. Differences in psychomotor activity and heart rate variability in patients with newly diagnosed bipolar disorder, unaffected relatives, and healthy individuals. J Affect Disord 2020; 266:30-36. [PMID: 32056891 PMCID: PMC7116568 DOI: 10.1016/j.jad.2020.01.110] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 11/22/2019] [Accepted: 01/20/2020] [Indexed: 11/24/2022]
Abstract
BACKGROUND Heart rate variability (HRV) and psychomotor activity have been found reduced in bipolar disorder (BD) but has never been investigated in newly diagnosed BD and unaffected relatives. The present study aimed to compare HRV and psychomotor activity between newly diagnosed patients with BD, their unaffected first-degree relatives (UR), and healthy control individuals (HC). METHODS 20 newly diagnosed patients with BD, 20 of their UR, and 20 age- and sex-matched HC were included. Measurements of HRV for five minutes and heart rate and acceleration for seven days were conducted. Activity energy expenditure (AEE) was derived from the latter. Linear mixed effect regression models were conducted to compare the three groups. RESULTS HRV did not differ in any measure between the three groups of participants. Similarly, AEE (kJ/day/kg) did not differ between the three groups in neither daily means (BD: 63.6, UR: 64.1, HC: 62.1) nor when divided into quarter-daily intervals. LIMITATIONS The relatively small size of the study may affect the validity of the results. CONCLUSION Patients with newly diagnosed BD and UR do not present with decreased HRV or AEE. These results contrast prior findings from BD patients with more advanced stages of the disorder, suggesting that these outcomes progress with illness duration.
Collapse
Affiliation(s)
- Josefine Freyberg
- The Copenhagen Affective Disorder research Center (CADIC), Psychiatric Center Copenhagen, Rigshospitalet, and Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Søren Brage
- MRC Epidemiology Unit, Cambridge, United Kingdom
| | - Lars Vedel Kessing
- The Copenhagen Affective Disorder research Center (CADIC), Psychiatric Center Copenhagen, Rigshospitalet, and Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Maria Faurholt-Jepsen
- The Copenhagen Affective Disorder research Center (CADIC), Psychiatric Center Copenhagen, Rigshospitalet, and Faculty of Health and Medical Sciences, University of Copenhagen, Denmark.
| |
Collapse
|
48
|
Pearce M, Strain T, Kim Y, Sharp SJ, Westgate K, Wijndaele K, Gonzales T, Wareham NJ, Brage S. Estimating physical activity from self-reported behaviours in large-scale population studies using network harmonisation: findings from UK Biobank and associations with disease outcomes. Int J Behav Nutr Phys Act 2020; 17:40. [PMID: 32178703 PMCID: PMC7074990 DOI: 10.1186/s12966-020-00937-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 02/17/2020] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND UK Biobank is a large prospective cohort study containing accelerometer-based physical activity data with strong validity collected from 100,000 participants approximately 5 years after baseline. In contrast, the main cohort has multiple self-reported physical behaviours from > 500,000 participants with longer follow-up time, offering several epidemiological advantages. However, questionnaire methods typically suffer from greater measurement error, and at present there is no tested method for combining these diverse self-reported data to more comprehensively assess the overall dose of physical activity. This study aimed to use the accelerometry sub-cohort to calibrate the self-reported behavioural variables to produce a harmonised estimate of physical activity energy expenditure, and subsequently examine its reliability, validity, and associations with disease outcomes. METHODS We calibrated 14 self-reported behavioural variables from the UK Biobank main cohort using the wrist accelerometry sub-cohort (n = 93,425), and used published equations to estimate physical activity energy expenditure (PAEESR). For comparison, we estimated physical activity based on the scoring criteria of the International Physical Activity Questionnaire, and by summing variables for occupational and leisure-time physical activity with no calibration. Test-retest reliability was assessed using data from the UK Biobank repeat assessment (n = 18,905) collected a mean of 4.3 years after baseline. Validity was assessed in an independent validation study (n = 98) with estimates based on doubly labelled water (PAEEDLW). In the main UK Biobank cohort (n = 374,352), Cox regression was used to estimate associations between PAEESR and fatal and non-fatal outcomes including all-cause, cardiovascular diseases, respiratory diseases, and cancers. RESULTS PAEESR explained 27% variance in gold-standard PAEEDLW estimates, with no mean bias. However, error was strongly correlated with PAEEDLW (r = -.98; p < 0.001), and PAEESR had narrower range than the criterion. Test-retest reliability (Λ = .67) and relative validity (Spearman = .52) of PAEESR outperformed two common approaches for processing self-report data with no calibration. Predictive validity was demonstrated by associations with morbidity and mortality, e.g. 14% (95%CI: 11-17%) lower mortality for individuals meeting lower physical activity guidelines. CONCLUSIONS The PAEESR variable has good reliability and validity for ranking individuals, with no mean bias but correlated error at individual-level. PAEESR outperformed uncalibrated estimates and showed stronger inverse associations with disease outcomes.
Collapse
Affiliation(s)
- Matthew Pearce
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge Biomedical Campus, Box 285, Cambridge, CB2 0QQ, UK
| | - Tessa Strain
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge Biomedical Campus, Box 285, Cambridge, CB2 0QQ, UK
| | - Youngwon Kim
- School of Public Health, The University of Hong Kong Li Ka Shing Faculty of Medicine, Room 301D 3/F, Jockey Club Building for Interdisciplinary Research, 5 Sassoon Road, Pokfulam, Hong Kong
| | - Stephen J Sharp
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge Biomedical Campus, Box 285, Cambridge, CB2 0QQ, UK
| | - Kate Westgate
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge Biomedical Campus, Box 285, Cambridge, CB2 0QQ, UK
| | - Katrien Wijndaele
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge Biomedical Campus, Box 285, Cambridge, CB2 0QQ, UK
| | - Tomas Gonzales
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge Biomedical Campus, Box 285, Cambridge, CB2 0QQ, UK
| | - Nicholas J Wareham
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge Biomedical Campus, Box 285, Cambridge, CB2 0QQ, UK
| | - Søren Brage
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge Biomedical Campus, Box 285, Cambridge, CB2 0QQ, UK.
| |
Collapse
|