51
|
Loh NY, Vasan SK, Rosoff DB, Roberts E, van Dam AD, Verma M, Phillips D, Wesolowska-Andersen A, Neville MJ, Noordam R, Ray DW, Tobias JH, Gregson CL, Karpe F, Christodoulides C. LRP5 promotes adipose progenitor cell fitness and adipocyte insulin sensitivity. COMMUNICATIONS MEDICINE 2025; 5:51. [PMID: 40000740 PMCID: PMC11862225 DOI: 10.1038/s43856-025-00774-1] [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: 03/04/2024] [Accepted: 02/17/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND WNT signaling plays a key role in postnatal bone formation. Individuals with gain-of-function mutations in the WNT co-receptor LRP5 exhibit increased lower-body fat mass and potentially enhanced glucose metabolism, alongside high bone mass. However, the mechanisms by which LRP5 regulates fat distribution and its effects on systemic metabolism remain unclear. This study aims to explore the role of LRP5 in adipose tissue biology and its impact on metabolism. METHODS Metabolic assessments and imaging were conducted on individuals with gain- and loss-of-function LRP5 mutations, along with age- and BMI-matched controls. Mendelian randomization analyses were used to investigate the relationship between bone, fat distribution, and systemic metabolism. Functional studies and RNA sequencing were performed on abdominal and gluteal adipose cells with LRP5 knockdown. RESULTS Here we show that LRP5 promotes lower-body fat distribution and enhances systemic and adipocyte insulin sensitivity through cell-autonomous mechanisms, independent of its bone-related functions. LRP5 supports adipose progenitor cell function by activating WNT/β-catenin signaling and preserving valosin-containing protein (VCP)-mediated proteostasis. LRP5 expression in adipose progenitors declines with age, but gain-of-function LRP5 variants protect against age-related fat loss in the lower body. CONCLUSIONS Our findings underscore the critical role of LRP5 in regulating lower-body fat distribution and insulin sensitivity, independent of its effects on bone. Pharmacological activation of LRP5 in adipose tissue may offer a promising strategy to prevent age-related fat redistribution and metabolic disorders.
Collapse
Affiliation(s)
- Nellie Y Loh
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Senthil K Vasan
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Daniel B Rosoff
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
- MRC Integrative Epidemiology Unit (IEU), Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Emile Roberts
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Andrea D van Dam
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Manu Verma
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Daniel Phillips
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Agata Wesolowska-Andersen
- Nuffield Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Matt J Neville
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, OUH Foundation Trust, Oxford, UK
| | - Raymond Noordam
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - David W Ray
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, OUH Foundation Trust, Oxford, UK
| | - Jonathan H Tobias
- MRC Integrative Epidemiology Unit (IEU), Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- Musculoskeletal Research Unit, Translational Health Sciences, Bristol Medical School, Southmead Hospital, University of Bristol, Bristol, UK
| | - Celia L Gregson
- MRC Integrative Epidemiology Unit (IEU), Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- Musculoskeletal Research Unit, Translational Health Sciences, Bristol Medical School, Southmead Hospital, University of Bristol, Bristol, UK
| | - Fredrik Karpe
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, OUH Foundation Trust, Oxford, UK
| | - Constantinos Christodoulides
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK.
- NIHR Oxford Biomedical Research Centre, OUH Foundation Trust, Oxford, UK.
| |
Collapse
|
52
|
Moss ST, Minelli C, Leavy OC, Allen RJ, Oliver N, Wain LV, Jenkins G, Stewart I. Assessing causal relationships between diabetes mellitus and idiopathic pulmonary fibrosis: a Mendelian randomisation study. Thorax 2025; 80:133-139. [PMID: 39613458 PMCID: PMC11877114 DOI: 10.1136/thorax-2024-221472] [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: 01/23/2024] [Accepted: 10/30/2024] [Indexed: 12/01/2024]
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is a disease of progressive lung scarring. There is a known association between diabetes mellitus (DM) and IPF, but it is unclear whether a causal relationship exists between these traits. OBJECTIVES The objectives of this study are to examine causal relationships among DM, diabetes-associated traits and IPF using a Mendelian randomisation approach. METHODS Two-sample MR approaches, including bidirectional inverse-variance weighted random effects and routine sensitivity models, used genetic variants identified from genome-wide association studies for type 1 diabetes (T1D), type 2 diabetes (T2D), glycated haemoglobin level (HbA1c), fasting insulin level and body mass index (BMI) to assess for causal effects of these traits on IPF. Further analyses using pleiotropy-robust and multivariable MR (MVMR) methods were additionally performed to account for trait complexity. RESULTS Results did not suggest that either T1D (OR=1.00, 95% CI 0.93 to 1.07, p=0.90) or T2D (1.02, 0.93 to 1.11, p=0.69) are in the causal pathway of IPF. No effects were suggested of HbA1c (1.19, 0.63 to 2.22, p=0.59) or fasting insulin level (0.60, 0.31 to 1.15, p=0.12) on IPF, but potential effects of BMI on IPF were indicated (1.44, 1.12 to 1.85, p=4.00×10-3). Results were consistent in MVMR, although no independent effects of T2D (0.91, 0.68 to 1.21, p=0.51) or BMI (1.01, 0.94 to 1.09, p=0.82) on IPF were observed when modelled together. CONCLUSIONS This study suggests that DM and IPF are unlikely to be causally linked. This comorbid relationship may instead be driven by shared risk factors or treatment effects.
Collapse
Affiliation(s)
- Samuel T Moss
- National Heart and Lung Institute, Imperial College London, London, UK
- Department of Population Health Sciences, University of Leicester, Leicester, UK
| | - Cosetta Minelli
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Olivia C Leavy
- Department of Population Health Sciences, University of Leicester, Leicester, UK
- NIHR Leicester Biomedical Research Centre, Leicester, East Midlands, UK
| | - Richard J Allen
- Department of Population Health Sciences, University of Leicester, Leicester, UK
- NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
| | - Nick Oliver
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Louise V Wain
- Department of Population Health Sciences, University of Leicester, Leicester, UK
- NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
| | - Gisli Jenkins
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Iain Stewart
- National Heart & Lung Institute, Imperial College London, London, UK
| |
Collapse
|
53
|
Jain PR, Ng HK, Tay D, Mina T, Low D, Sadhu N, Kooner IK, Gupta A, Li TF, Bertin N, Chin CWL, Fang CJ, Goh LL, Mok SQ, Peh SQ, Sabanayagam C, Jha V, Kasturiratne A, Katulanda P, Khawaja KI, Lim WK, Leong KP, Cheng CY, Yuan JM, Elliott P, Riboli E, Sing LE, Lee J, Ngeow J, Liu JJ, Best J, Kooner JS, Tai ES, Tan P, van Dam RM, Koh WP, Xueling S, Loh M, Chambers JC. Nuclear regulatory disturbances precede and predict the development of Type-2 diabetes in Asian populations. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.14.25322264. [PMID: 39990582 PMCID: PMC11844604 DOI: 10.1101/2025.02.14.25322264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
To identify biomarkers and pathways to Type-2 diabetes (T2D), a major global disease, we completed array-based epigenome-wide association in whole blood in 5,709 Asian people. We found 323 Sentinel CpGs (from 314 genetic loci) that predict future T2D. The CpGs reveal coherent, nuclear regulatory disturbances in canonical immune activation pathways, as well as metabolic networks involved in insulin signalling, fatty acid metabolism and lipid transport, which are causally linked to development of T2D. The CpGs have potential clinical utility as biomarkers. An array-based composite Methylation Risk Score (MRS) is predictive for future T2D (RR: 5.2 in Q4 vs Q1; P=7×10-25), and is additive to genetic risk. Targeted methylation sequencing revealed multiple additional CpGs predicting T2D, and synthesis of a sequencing-based MRS that is strongly predictive for T2D (RR: 8.3 in Q4 vs Q1; P=1.0×10-11). Importantly, MRS varies between Asian ethnic groups, in a way that explains a large fraction of the difference in T2D risk between populations. We thus provide new insights into the nuclear regulatory disturbances that precede development of T2D, and reveal the potential for sequence-based DNA methylation markers to inform risk stratification in diabetes prevention.
Collapse
Affiliation(s)
- Pritesh R Jain
- Population and Global Health, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Hong Kiat Ng
- Population and Global Health, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Darwin Tay
- Population and Global Health, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Theresia Mina
- Population and Global Health, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Dorrain Low
- Population and Global Health, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Nilanjana Sadhu
- Population and Global Health, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Ishminder K Kooner
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, United Kingdom
| | - Ananya Gupta
- Population and Global Health, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Tai Fei Li
- Population and Global Health, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Nicolas Bertin
- Precision Health Research (PRECISE), Singapore
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore
| | | | - Chai Jin Fang
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
| | - Liuh Ling Goh
- Personalised Medicine Service, Tan Tock Seng Hospital, Singapore
| | - Shi Qi Mok
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore
| | - Su Qin Peh
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore
| | - Charumathi Sabanayagam
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
- Ophthalmology & Visual Sciences Academic Clinical Program, Duke-NUS Medical School, Singapore
| | | | | | | | | | - Weng Khong Lim
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore
- SingHealth Duke-NUS Institute of Precision Medicine, Singapore
- SingHealth Duke-NUS Genomic Medicine Centre, Singapore
- Cancer & Stem Cell Biology Program, Duke-NUS Medical School, Singapore
| | - Khai Pang Leong
- Personalised Medicine Service, Tan Tock Seng Hospital, Singapore
| | - Ching-Yu Cheng
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
- Centre for Innovation and Precision Eye Health, Yong Loo Lin School of Medicine, National University of Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Jian-Min Yuan
- University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Paul Elliott
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, United Kingdom
| | - Elio Riboli
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, United Kingdom
| | - Lee Eng Sing
- Population and Global Health, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Jimmy Lee
- Population and Global Health, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
- Institute of Mental Health, Singapore
| | - Joanne Ngeow
- Population and Global Health, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
- Division of Medical Oncology, National Cancer Centre, Singapore
| | - Jian Jin Liu
- Human Genomics, Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - James Best
- Melbourne Medical School, University of Melbourne, Australia
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Jaspal S Kooner
- Department of Cardiology, Ealing Hospital, London North West Healthcare NHS Trust, Middlesex, United Kingdom
- Imperial College Healthcare NHS Trust, Imperial College London, London, United Kingdom
- Medical Research Council-Public Health England Centre for Environment and Health, Imperial College London, London, United Kingdom
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - E-Shyong Tai
- Precision Health Research (PRECISE), Singapore
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
- Division of Endocrinology, National University Hospital, Singapore
| | - Patrick Tan
- Precision Health Research (PRECISE), Singapore
- Duke-NUS Medical School, Singapore
| | - Rob M van Dam
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
- Department of Exercise and Nutrition Sciences, Milken Institute School of Public Health, The George Washington University, Washington DC, USA
| | - Woon-Puay Koh
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore
| | - Sim Xueling
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
| | - Marie Loh
- Population and Global Health, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, United Kingdom
- National Skin Centre, Research Division, Singapore
| | - John C Chambers
- Population and Global Health, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, United Kingdom
- Precision Health Research (PRECISE), Singapore
| |
Collapse
|
54
|
Liu Z, Chen X, Yuan H, Jin L, Zhang T, Chen X. Dissecting the shared genetic architecture between nonalcoholic fatty liver disease and type 2 diabetes. Hum Mol Genet 2025; 34:338-346. [PMID: 39690818 DOI: 10.1093/hmg/ddae184] [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: 06/11/2024] [Revised: 09/04/2024] [Accepted: 12/05/2024] [Indexed: 12/19/2024] Open
Abstract
Observational studies have reported a bidirectional correlation between nonalcoholic fatty liver disease (NAFLD) and type 2 diabetes (T2D), but the shared genetic basis between the two conditions remains unclear. Using genome-wide association study (GWAS) summary data from European-ancestry populations, we examined the cross-trait genetic correlation and identified genomic overlaps and shared risk loci. We employed a latent causal variable model and Mendelian randomization (MR) analysis to infer causal relationships. Colocalization analysis and conditional/conjunctional false discovery rate (condFDR/conjFDR) were used to identify genomic overlaps and shared risk loci. Two-step MR analysis was utilized to identify potential mediators. We observed a strong positive genomic correlation between NAFLD and T2D (rg = 0.652, P = 5.67 × 10-6) and identified tissue-specific transcriptomic correlations in the pancreas, liver, skeletal muscle, subcutaneous adipose, and blood. Genetic enrichment was observed in NAFLD conditional on associations with T2D and vice versa, indicating significant polygenic overlaps. We found robust evidence for the causal effect of NAFLD on T2D, particularly insulin-related T2D, rather than vice versa. Colocalization analysis identified shared genomic regions between NAFLD and T2D, including GCKR, FTO, MAU2-TM6SF2, and PNPLA3-SAMM50. High-density lipoprotein cholesterol and insulin were partly mediated the association between NAFLD and T2D. These findings unveil a close genetic link between NAFLD and T2D, shedding light on the biological mechanisms connecting NAFLD progression to T2D.
Collapse
Affiliation(s)
- Zhenqiu Liu
- State Key Laboratory of Genetic Engineering and Human Phenome Institute, Fudan University, 825 Zhangheng RD, Pudong New Area, Shanghai 201203, China
- Fudan University Taizhou Institute of Health Sciences, 799 Yaocheng RD, Taizhou 225316, China
| | - Xiaochen Chen
- Clinical Research Institute, Shanghai Jiao Tong University School of Medicine, 227 Chongqing South RD, Shanghai 200025, China
| | - Huangbo Yuan
- State Key Laboratory of Genetic Engineering and Human Phenome Institute, Fudan University, 825 Zhangheng RD, Pudong New Area, Shanghai 201203, China
- Fudan University Taizhou Institute of Health Sciences, 799 Yaocheng RD, Taizhou 225316, China
| | - Li Jin
- State Key Laboratory of Genetic Engineering and Human Phenome Institute, Fudan University, 825 Zhangheng RD, Pudong New Area, Shanghai 201203, China
- Fudan University Taizhou Institute of Health Sciences, 799 Yaocheng RD, Taizhou 225316, China
| | - Tiejun Zhang
- Fudan University Taizhou Institute of Health Sciences, 799 Yaocheng RD, Taizhou 225316, China
- Key Laboratory of Public Health Safety, Fudan University, Ministry of Education, 130 Dong'an RD, Shanghai 200032, China
- Department of Epidemiology, School of Public Health, Fudan University, 130 Dong'an RD, Shanghai 200032, China
- Yiwu Research Institute of Fudan University, 2 Chengbei RD, Yiwu 322000, China
| | - Xingdong Chen
- State Key Laboratory of Genetic Engineering and Human Phenome Institute, Fudan University, 825 Zhangheng RD, Pudong New Area, Shanghai 201203, China
- Fudan University Taizhou Institute of Health Sciences, 799 Yaocheng RD, Taizhou 225316, China
- Yiwu Research Institute of Fudan University, 2 Chengbei RD, Yiwu 322000, China
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, 12 Urumqi RD, Shanghai 200040, China
| |
Collapse
|
55
|
Yuan W, Xu X, Zhang X, Fan W, Zhou W, Zhao F. Exploring the Associations of Obesity and Glycemic Traits with Retinal Vein Occlusion: A Univariate and Multivariable Mendelian Randomization Study. Ophthalmic Epidemiol 2025:1-9. [PMID: 39919303 DOI: 10.1080/09286586.2025.2458245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 11/24/2024] [Accepted: 01/19/2025] [Indexed: 02/09/2025]
Abstract
PURPOSE To explore the genetic links between obesity, glycemic traits and retinal vein occlusion (RVO). METHODS Summary-level statistics for obesity and glycemic traits were extracted from publicly available genome-wide association studies (GWAS) of European participants in the IEU Open GWAS database. Genetic associations with clinically diagnosed RVO were obtained from the FinnGenresearch project (372 cases and 182,573 controls). Two-sample Mendelian randomization (MR) and multivariate MR (MVMR) analysis were performed to determine the total effect and direct effect, respectively. RESULTS After adjustment for the false discovery rate (FDR), the primary inverse-variance-weighted (IVW) methods indicated that the odds ratios of RVO increased with per 1-standard deviation increased in body mass index (BMI) (OR = 1.94, 95% CI: 1.23-3.08,p-FDR = 0.025), waist circumference (OR = 2.4, 95% CI: 1.36-4.24, p-FDR = 0.019), fasting glucose (OR = 5.01, 95% CI: 2-12.55, p-FDR = 0.0067) and two-hour glucose (OR = 3.17, 95% CI: 1.63-6.18,p-FDR = 0.0067). Higher whole-body fat-free mass (OR = 0.45, 95% CI: 0.26-0.8,p-FDR = 0.025) is a potential protective factor for RVO. In addition, the results of MVMR showed that BMI, whole-body fat-free mass, fasting glucose and two-hour glucose were independent factors that had a direct impact on the onset of RVO. CONCLUSIONS Our comprehensive MR analysis suggested significant genetic associations between BMI, whole-body fat-free mass, fasting glucose, two-hour glucose and RVO. This study highlighted the importance of weight, blood glucose management and physical activity for primary prevention and control of RVO.
Collapse
Affiliation(s)
- Weichen Yuan
- Department of Ophthalmology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
- Department of Ophthalmology, Key Lens Research Laboratory of Liaoning Province, Shenyang, China
| | - Xin Xu
- Department of Biochemistry and Molecular Biology, China Medical University, Shenyang, China
| | - Xiran Zhang
- Department of Optometry, China Medical University, Shenyang, China
| | - Wenqi Fan
- Department of Optometry, China Medical University, Shenyang, China
| | - Wenkai Zhou
- Department of Ophthalmology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
- Department of Ophthalmology, Key Lens Research Laboratory of Liaoning Province, Shenyang, China
| | - Fangkun Zhao
- Department of Ophthalmology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
- Department of Ophthalmology, Key Lens Research Laboratory of Liaoning Province, Shenyang, China
| |
Collapse
|
56
|
Liu H, Abedini A, Ha E, Ma Z, Sheng X, Dumoulin B, Qiu C, Aranyi T, Li S, Dittrich N, Chen HC, Tao R, Tarng DC, Hsieh FJ, Chen SA, Yang SF, Lee MY, Kwok PY, Wu JY, Chen CH, Khan A, Limdi NA, Wei WQ, Walunas TL, Karlson EW, Kenny EE, Luo Y, Kottyan L, Connolly JJ, Jarvik GP, Weng C, Shang N, Cole JB, Mercader JM, Mandla R, Majarian TD, Florez JC, Haas ME, Lotta LA, Regeneron Genetics Center, GHS-RGC DiscovEHR Collaboration, Drivas TG, Penn Medicine BioBank, Vy HMT, Nadkarni GN, Wiley LK, Wilson MP, Gignoux CR, Rasheed H, Thomas LF, Åsvold BO, Brumpton BM, Hallan SI, Hveem K, Zheng J, Hellwege JN, Zawistowski M, Zöllner S, Franceschini N, Hu H, Zhou J, Kiryluk K, Ritchie MD, Palmer M, Edwards TL, Voight BF, Hung AM, Susztak K. Kidney multiome-based genetic scorecard reveals convergent coding and regulatory variants. Science 2025; 387:eadp4753. [PMID: 39913582 PMCID: PMC12013656 DOI: 10.1126/science.adp4753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Collaborators] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 11/20/2024] [Indexed: 02/17/2025]
Abstract
Kidney dysfunction is a major cause of mortality, but its genetic architecture remains elusive. In this study, we conducted a multiancestry genome-wide association study in 2.2 million individuals and identified 1026 (97 previously unknown) independent loci. Ancestry-specific analysis indicated an attenuation of newly identified signals on common variants in European ancestry populations and the power of population diversity for further discoveries. We defined genotype effects on allele-specific gene expression and regulatory circuitries in more than 700 human kidneys and 237,000 cells. We found 1363 coding variants disrupting 782 genes, with 601 genes also targeted by regulatory variants and convergence in 161 genes. Integrating 32 types of genetic information, we present the "Kidney Disease Genetic Scorecard" for prioritizing potentially causal genes, cell types, and druggable targets for kidney disease.
Collapse
Affiliation(s)
- Hongbo Liu
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Diabetes Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
- Penn-CHOP Kidney Innovation Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Amin Abedini
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Diabetes Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Eunji Ha
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Diabetes Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Ziyuan Ma
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Diabetes Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Xin Sheng
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Diabetes Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
- Liangzhu Laboratory, Zhejiang University, 1369 West Wenyi Road, Hangzhou, Zhejiang, China
- Department of Nephrology, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| | - Bernhard Dumoulin
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Diabetes Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Chengxiang Qiu
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Diabetes Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Tamas Aranyi
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Diabetes Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Molecular Life Sciences, HUN-REN Research Center for Natural Sciences, Budapest, Hungary
- Department of Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Shen Li
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Diabetes Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Nicole Dittrich
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Diabetes Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
- Department of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Hua-Chang Chen
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ran Tao
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Der-Cherng Tarng
- Institute of Clinical Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- Division of Nephrology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Feng-Jen Hsieh
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan, ROC
| | - Shih-Ann Chen
- Cardiovascular Center, Taichung Veterans General Hospital, Taichung, Taiwan, ROC
- National Chung Hsing University, Taichung, Taiwan, ROC
- Heart Rhythm Center, Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Department of Internal Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan, ROC
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan, ROC
| | - Mei-Yueh Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan, ROC
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, ROC
- Department of Internal Medicine, Kaohsiung Medical University Gangshan Hospital, Kaohsiung, Taiwan, ROC
| | - Pui-Yan Kwok
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan, ROC
- Institute for Human Genetics, University of California, San Francisco, CA, USA
| | - Jer-Yuarn Wu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan, ROC
| | - Chien-Hsiun Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan, ROC
| | - Atlas Khan
- Division of Nephrology, Department of Medicine, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Nita A. Limdi
- Department of Neurology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Wei-Qi Wei
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Theresa L. Walunas
- Department of Medicine, Division of General Internal Medicine and Center for Health Information Partnerships, Institute for Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | | - Eimear E. Kenny
- Institute for Genomic Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Genomic Medicine, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of General Internal Medicine, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yuan Luo
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Leah Kottyan
- The Center for Autoimmune Genomics and Etiology, Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - John J. Connolly
- Center for Applied Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Gail P. Jarvik
- Departments of Medicine (Medical Genetics) and Genome Sciences, University of Washington, Seattle, WA, USA
| | - Chunhua Weng
- Department of Biomedical Informatics, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Ning Shang
- Division of Nephrology, Department of Medicine, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Joanne B. Cole
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Division of Endocrinology, Boston Children’s Hospital, Boston, MA, USA
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Josep M. Mercader
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Diabetes Unit, Endocrine Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Ravi Mandla
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Diabetes Unit, Endocrine Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine and Cardiovascular Research Institute, Cardiology Division, University of California, San Francisco, CA, USA
- Graduate Program in Genomics and Computational Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Timothy D. Majarian
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Vertex Pharmaceuticals, Boston, MA, USA
| | - Jose C. Florez
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Diabetes Unit, Endocrine Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Mary E. Haas
- Regeneron Genetics Center, Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | - Luca A. Lotta
- Regeneron Genetics Center, Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | | | | | - Theodore G. Drivas
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | | | - Ha My T. Vy
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Charles Bronfman Institute of Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Girish N. Nadkarni
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Charles Bronfman Institute of Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Hasso Plattner Institute of Digital Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Mount Sinai Clinical Intelligence Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Laura K. Wiley
- Colorado Center for Personalized Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Biomedical Informatics, University of Colorado School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Melissa P. Wilson
- Department of Biomedical Informatics, University of Colorado School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Christopher R. Gignoux
- Colorado Center for Personalized Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Biomedical Informatics, University of Colorado School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Humaira Rasheed
- KGJebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Laurent F. Thomas
- KGJebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Clinical and Molecular Medicine, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- BioCore - Bioinformatics Core Facility, Norwegian University of Science and Technology, Trondheim, Norway
| | - Bjørn Olav Åsvold
- KGJebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Endocrinology, Clinic of Medicine, St. Olav’s Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Ben M. Brumpton
- KGJebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- Clinic of Thoracic and Occupational Medicine, St. Olav’s Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Stein I. Hallan
- Department of Clinical and Molecular Medicine, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Nephrology, St. Olav’s Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Kristian Hveem
- KGJebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
| | - Jie Zheng
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jacklyn N. Hellwege
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Matthew Zawistowski
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Sebastian Zöllner
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
| | - Nora Franceschini
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Hailong Hu
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Diabetes Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Jianfu Zhou
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Diabetes Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Krzysztof Kiryluk
- Division of Nephrology, Department of Medicine, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Marylyn D. Ritchie
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Matthew Palmer
- Pathology and Laboratory Medicine at the Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Todd L. Edwards
- Division of Epidemiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Benjamin F. Voight
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA, USA
| | - Adriana M. Hung
- Division of Nephrology and Hypertension, Vanderbilt Center for Kidney Disease, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- VA Tennessee Valley Healthcare System, Clinical Sciences Research and Development, Nashville, TN, USA
| | - Katalin Susztak
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Diabetes Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
- Penn-CHOP Kidney Innovation Center, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
Collaborators
Aris Baras, Gonçalo Abecasis, Adolfo Ferrando, Giovanni Coppola, Andrew Deubler, Aris Economides, Luca A Lotta, John D Overton, Jeffrey G Reid, Alan Shuldiner, Katherine Siminovitch, Jason Portnoy, Marcus B Jones, Lyndon Mitnaul, Alison Fenney, Jonathan Marchini, Manuel Allen Revez Ferreira, Maya Ghoussaini, Mona Nafde, William Salerno, John D Overton, Christina Beechert, Erin Fuller, Laura M Cremona, Eugene Kalyuskin, Hang Du, Caitlin Forsythe, Zhenhua Gu, Kristy Guevara, Michael Lattari, Alexander Lopez, Kia Manoochehri, Prathyusha Challa, Manasi Pradhan, Raymond Reynoso, Ricardo Schiavo, Maria Sotiropoulos Padilla, Chenggu Wang, Sarah E Wolf, Hang Du, Kristy Guevara, Amelia Averitt, Nilanjana Banerjee, Dadong Li, Sameer Malhotra, Justin Mower, Mudasar Sarwar, Deepika Sharma, Sean Yu, Aaron Zhang, Muhammad Aqeel, Jeffrey G Reid, Mona Nafde, Manan Goyal, George Mitra, Sanjay Sreeram, Rouel Lanche, Vrushali Mahajan, Sai Lakshmi Vasireddy, Gisu Eom, Krishna Pawan Punuru, Sujit Gokhale, Benjamin Sultan, Pooja Mule, Eliot Austin, Xiaodong Bai, Lance Zhang, Sean O'Keeffe, Razvan Panea, Evan Edelstein, Ayesha Rasool, William Salerno, Evan K Maxwell, Boris Boutkov, Alexander Gorovits, Ju Guan, Lukas Habegger, Alicia Hawes, Olga Krasheninina, Samantha Zarate, Adam J Mansfield, Lukas Habegger, Gonçalo Abecasis, Joshua Backman, Kathy Burch, Adrian Campos, Liron Ganel, Sheila Gaynor, Benjamin Geraghty, Arkopravo Ghosh, Salvador Romero Martinez, Christopher Gillies, Lauren Gurski, Joseph Herman, Eric Jorgenson, Tyler Joseph, Michael Kessler, Jack Kosmicki, Adam Locke, Priyanka Nakka, Jonathan Marchini, Karl Landheer, Olivier Delaneau, Maya Ghoussaini, Anthony Marcketta, Joelle Mbatchou, Arden Moscati, Aditeya Pandey, Anita Pandit, Jonathan Ross, Carlo Sidore, Eli Stahl, Timothy Thornton, Sailaja Vedantam, Rujin Wang, Kuan-Han Wu, Bin Ye, Blair Zhang, Andrey Ziyatdinov, Yuxin Zou, Jingning Zhang, Kyoko Watanabe, Mira Tang, Frank Wendt, Suganthi Balasubramanian, Suying Bao, Kathie Sun, Chuanyi Zhang, Adolfo Ferrando, Giovanni Coppola, Luca A Lotta, Alan Shuldiner, Katherine Siminovitch, Brian Hobbs, Jon Silver, William Palmer, Rita Guerreiro, Amit Joshi, Antoine Baldassari, Cristen Willer, Sarah Graham, Ernst Mayerhofer, Erola Pairo Castineira, Mary Haas, Niek Verweij, George Hindy, Jonas Bovijn, Tanima De, Parsa Akbari, Luanluan Sun, Olukayode Sosina, Arthur Gilly, Peter Dornbos, Juan Rodriguez-Flores, Moeen Riaz, Manav Kapoor, Gannie Tzoneva, Momodou W Jallow, Anna Alkelai, Ariane Ayer, Veera Rajagopal, Sahar Gelfman, Vijay Kumar, Jacqueline Otto, Neelroop Parikshak, Aysegul Guvenek, Jose Bras, Silvia Alvarez, Jessie Brown, Jing He, Hossein Khiabanian, Joana Revez, Kimberly Skead, Valentina Zavala, Jae Soon Sul, Lei Chen, Sam Choi, Amy Damask, Nan Lin, Charles Paulding, Marcus B Jones, Esteban Chen, Michelle G LeBlanc, Jason Mighty, Jennifer Rico-Varela, Nirupama Nishtala, Nadia Rana, Jaimee Hernandez, Alison Fenney, Randi Schwartz, Jody Hankins, Anna Han, Samuel Hart, Ann Perez-Beals, Gina Solari, Johannie Rivera-Picart, Michelle Pagan, Sunilbe Siceron, Adam Buchanan, David J Carey, Christa L Martin, Michelle Meyer, Kyle Retterer, David Rolston, Daniel J Rader, Marylyn D Ritchie, JoEllen Weaver, Nawar Naseer, Giorgio Sirugo, Afiya Poindexter, Yi-An Ko, Kyle P Nerz, Meghan Livingstone, Fred Vadivieso, Stephanie DerOhannessian, Teo Tran, Julia Stephanowski, Salma Santos, Ned Haubein, Joseph Dunn, Anurag Verma, Colleen Morse Kripke, Marjorie Risman, Renae Judy, Colin Wollack, Shefali S Verma, Scott M Damrauer, Yuki Bradford, Scott M Dudek, Theodore G Drivas,
Collapse
|
57
|
Fu L, Liu Q, Cheng H, Zhao X, Xiong J, Mi J. Insights Into Causal Effects of Genetically Proxied Lipids and Lipid-Modifying Drug Targets on Cardiometabolic Diseases. J Am Heart Assoc 2025; 14:e038857. [PMID: 39868518 PMCID: PMC12074789 DOI: 10.1161/jaha.124.038857] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 12/13/2024] [Indexed: 01/28/2025]
Abstract
BACKGROUND The differential impact of serum lipids and their targets for lipid modification on cardiometabolic disease risk is debated. This study used Mendelian randomization to investigate the causal relationships and underlying mechanisms. METHODS Genetic variants related to lipid profiles and targets for lipid modification were sourced from the Global Lipids Genetics Consortium. Summary data for 10 cardiometabolic diseases were compiled from both discovery and replication data sets. Expression quantitative trait loci data from relevant tissues were employed to evaluate significant lipid-modifying drug targets. Comprehensive analyses including colocalization, mediation, and bioinformatics were conducted to validate the results and investigate potential mediators and mechanisms. RESULTS Significant causal associations were identified between lipids, lipid-modifying drug targets, and various cardiometabolic diseases. Notably, genetic enhancement of LPL (lipoprotein lipase) was linked to reduced risks of myocardial infarction (odds ratio [OR]1, 0.65 [95% CI, 0.57-0.75], P1=2.60×10-9; OR2, 0.59 [95% CI, 0.49-0.72], P2=1.52×10-7), ischemic heart disease (OR1, 0.968 [95% CI, 0.962-0.975], P1=5.50×10-23; OR2, 0.64 [95% CI, 0.55-0.73], P2=1.72×10-10), and coronary heart disease (OR1, 0.980 [95% CI, 0.975-0.985], P1=3.63×10-14; OR2, 0.64 [95% CI, 0.54-0.75], P2=6.62×10-8) across 2 data sets. Moreover, significant Mendelian randomization and strong colocalization associations for the expression of LPL in blood and subcutaneous adipose tissue were linked with myocardial infarction (OR, 0.918 [95% CI, 0.872-0.967], P=1.24×10-3; PP.H4, 0.99) and coronary heart disease (OR, 0.991 [95% CI, 0.983-0.999], P=0.041; PP.H4=0.92). Glucose levels and blood pressure were identified as mediators in the total effect of LPL on cardiometabolic outcomes. CONCLUSIONS The study substantiates the causal role of lipids in specific cardiometabolic diseases, highlighting LPL as a potent drug target. The effects of LPL are suggested to be influenced by changes in glucose and blood pressure, providing insights into its mechanism of action.
Collapse
Affiliation(s)
- Liwan Fu
- Center for Non‐Communicable Disease ManagementBeijing Children’s Hospital, Capital Medical University, National Center for Children’s HealthBeijingChina
| | - Qin Liu
- Department of UltrasoundChildren’s Hospital of the Capital Institute of PediatricsBeijingChina
| | - Hong Cheng
- Department of EpidemiologyCapital Institute of PediatricsBeijingChina
| | - Xiaoyuan Zhao
- Department of EpidemiologyCapital Institute of PediatricsBeijingChina
| | - Jingfan Xiong
- Child and Adolescent Chronic Disease Prevention and Control DepartmentShenzhen Center for Chronic Disease ControlShenzhenChina
| | - Jie Mi
- Center for Non‐Communicable Disease ManagementBeijing Children’s Hospital, Capital Medical University, National Center for Children’s HealthBeijingChina
- Key Laboratory of Major Diseases in Children, Ministry of EducationChina
| |
Collapse
|
58
|
Rajamoorthi A, Zheng H, Skowronski AA, Zork N, Reddy UM, Tung PW, Kupsco A, Gallagher D, Salem RM, Leibel RL, LeDuc CA, Thaker VV. Association of gestational and childhood circulating C-peptide concentrations in the hyperglycemia and adverse pregnancy outcomes follow-up study. Diabetes Res Clin Pract 2025; 220:111967. [PMID: 39716665 PMCID: PMC11840794 DOI: 10.1016/j.diabres.2024.111967] [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: 09/10/2024] [Revised: 12/02/2024] [Accepted: 12/16/2024] [Indexed: 12/25/2024]
Abstract
AIMS This study examined the association of gravida C-peptide with progeny islet function and insulin sensitivity in the Hyperglycemia and Adverse Pregnancy Outcome Follow-up Study (HAPO FUS). METHODS Pregnancy 3rd trimester oral glucose tolerance test (OGTT), cord blood, and offspring OGTT glucose, C-peptide and insulin at age 10-14 years were analyzed for 4,121 mother-child dyads. Gravida fasting and 1-hour C-peptide concentration correlations with cord blood and childhood C-peptide, insulin, insulinogenic index and insulin sensitivity, and insulin resistance [HOMA-IR]), were assessed by multiple linear regression. Maternal covariates included age, gestational age, BMI and glucose at OGTT; child covariates included age, sex, pubertal stage, BMI z score and glucose. RESULTS Gravida fasting and 1-hour OGTT C-peptide was positively correlated with cord blood C-peptide, offspring OGTT C-peptide and insulin concentrations at fasting, 30 min, 1-hour and 2-hour at 10-14 years of age. Maternal fasting and 1-hour C-peptide concentrations were positively correlated with the insulinogenic index and HOMA-IR but inversely correlated with insulin sensitivity. Maternal C-peptide explained more variance than maternal glucose concentrations (3.0-17.9 % vs 0.2-3.5 %). CONCLUSIONS/INTERPRETATION The correlation between gravida and offspring C-peptide suggests that without crossing the placenta, insulin may influence the offspring pancreatic beta-cell development and insulin sensitivity.
Collapse
Affiliation(s)
- Ananthi Rajamoorthi
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Hao Zheng
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, NY, United States
| | - Alicja A Skowronski
- Department of Pediatrics, Division of Molecular Genetics, Columbia University Irving Medical Center, New York, NY, United States
| | - Noelia Zork
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY, United States
| | - Uma M Reddy
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY, United States
| | - Pei Wen Tung
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, United States
| | - Allison Kupsco
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, United States
| | - Dympna Gallagher
- Department of Medicine, Columbia University, Irving Medical Center, New York, NY, United States
| | - Rany M Salem
- Department of Family Medicine and Public Health, Division of Epidemiology, Herbert Wertheim School of Public Health and Human Longevity Science, University of California at San Diego, San Diego, CA, United States
| | - Rudolph L Leibel
- Department of Pediatrics, Division of Molecular Genetics, Columbia University Irving Medical Center, New York, NY, United States
| | - Charles A LeDuc
- Department of Pediatrics, Division of Molecular Genetics, Columbia University Irving Medical Center, New York, NY, United States
| | - Vidhu V Thaker
- Department of Pediatrics, Division of Molecular Genetics, Columbia University Irving Medical Center, New York, NY, United States; Department of Pediatrics, Division of Pediatric Endocrinology, Columbia University Irving Medical Center, New York, NY, United States.
| |
Collapse
|
59
|
Song ZQ, Chen YQ, Xuan CH, Ni TT, Xu YP, Lu XY, Chen FR, Chen YH. Effect of smoking behaviour and related blood DNA methylation on visceral adipose tissues. Diabetes Obes Metab 2025; 27:619-628. [PMID: 39511847 DOI: 10.1111/dom.16054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 10/22/2024] [Accepted: 10/22/2024] [Indexed: 11/15/2024]
Abstract
BACKGROUND Recent studies have found that tobacco smoking is associated with fat distribution, yet limited research has focused on its relationship with visceral adipose tissues (VATs). Furthermore, the cellular and molecular mechanisms underlying the interactions among smoking, epigenetic modifications, and VATs remain unknown. METHOD We performed univariable Mendelian randomization (MR) analysis to elucidate the causal relationship between smoking behaviours and VATs, including epicardial and pericardial adipose tissue (EPAT), liver fat (LF), and pancreas fat (PF). This approach could minimize the impact of confounders and reverse causality through utilizing genetic variants to proxy the smoking behaviours. Mediation MR analysis were conducted to detect potential mediators. Additionally, summary-data-based MR (SMR) and colocalization analysis were performed to explore the association between smoking-related DNA methylation and VATs. RESULTS We identified a convincing association between smoking initiation and increased EPAT (beta: 0.15, 95% CI: 0.06, 0.23, p = 7.01 × 10-4) and LF area (beta: 0.15, 95% CI = 0.05, 0.24, p = 2.85 × 10-3), respectively. Further mediation analysis suggested type 2 diabetes mellitus (T2DM) as a potential mediator within these co-relationships. When further exploring the associations between the smoking related DNA methylation and VATs, we identified that WT1 methylation at cg05222924 was significantly linked to a lower EPAT area (beta: -0.12, 95% CI: -0.16, -0.06, PFDR = 2.24 × 10-3), while GPX1 methylation at cg18642234 facilitated the deposition of EPAT (beta: 0.15, 95% CI: 0.10, 0.20, PFDR = 1.66 × 10-4). CONCLUSION Our study uncovered a significant causal effect between smoking and VATs, with T2DM identified as a potential mediator. Further investigation into DNA methylation yielded novel insights into the pathogenic role of smoking on EPAT.
Collapse
Affiliation(s)
- Zheng-Qi Song
- Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, China
| | - Yi-Qi Chen
- The Second Clinical Medical College, Wenzhou Medical University, Wenzhou, China
| | - Chen-Hao Xuan
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, China
| | - Tong-Tong Ni
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, China
| | - Yu-Peng Xu
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, China
| | - Xin-Yu Lu
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, China
| | - Fang-Ran Chen
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, China
| | - Yi-He Chen
- Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
60
|
Blanken CPS, Bayer S, Buchner Carro S, Hauner H, Holzapfel C. Associations Between TCF7L2, PPARγ, and KCNJ11 Genotypes and Insulin Response to an Oral Glucose Tolerance Test: A Systematic Review. Mol Nutr Food Res 2025; 69:e202400561. [PMID: 39828593 PMCID: PMC11791742 DOI: 10.1002/mnfr.202400561] [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: 07/19/2024] [Revised: 10/31/2024] [Accepted: 12/06/2024] [Indexed: 01/22/2025]
Abstract
SCOPE Insulin responses to standardized meals differ between individuals. This variability may in part be explained by genotype. This systematic review evaluates associations between genotype and insulin response to an oral glucose tolerance test (OGTT) in terms of insulin area under the curve (AUC). METHODS AND RESULTS Three electronic databases (Web of Science, Embase, PubMed) were searched for studies investigating associations between insulin AUC after an OGTT and single nucleotide polymorphisms (SNPs) belonging to the transcription factor 7 like 2 (TCF7L2) gene, the peroxisome proliferator-activated receptor gamma (PPARγ) gene, or the potassium inwardly rectifying channel subfamily J member 11 (KCNJ11) gene in persons without diabetes. A total of 5199 articles were identified, of which 38 were included. Among them were family-based studies (9), twin studies (2), and studies with unrelated participants (27). Seventeen articles investigated TCF7L2 (7 SNPs), 14 investigated PPARγ (1 SNP), and 8 investigated KCNJ11 (5 SNPs). For all investigated SNPs, at least half of the reports indicated no statistically significant association with postprandial insulin AUC. CONCLUSION No evidence was found for associations between TCF7L2, PPARγ, and KCNJ11 genotypes and insulin AUC after an OGTT. Future studies should investigate the effect of genetic risk scores on postprandial insulin.
Collapse
Affiliation(s)
- Carmen P. S. Blanken
- Institute for Nutritional Medicine, School of Medicine and Health, Technical University of MunichMunichGermany
| | - Sandra Bayer
- Institute for Nutritional Medicine, School of Medicine and Health, Technical University of MunichMunichGermany
| | - Sophie Buchner Carro
- Institute for Nutritional Medicine, School of Medicine and Health, Technical University of MunichMunichGermany
| | - Hans Hauner
- Institute for Nutritional Medicine, School of Medicine and Health, Technical University of MunichMunichGermany
| | - Christina Holzapfel
- Institute for Nutritional Medicine, School of Medicine and Health, Technical University of MunichMunichGermany
- Department of Nutritional, Food and Consumer SciencesFulda University of Applied SciencesFuldaGermany
| |
Collapse
|
61
|
Xu F, Wu S, Gao S, Li X, Huang C, Chen Y, Zhu P, Liu G. Causal association between insulin sensitivity index and Alzheimer's disease. J Neurochem 2025; 169:e16254. [PMID: 39479764 DOI: 10.1111/jnc.16254] [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: 04/13/2024] [Revised: 10/06/2024] [Accepted: 10/07/2024] [Indexed: 02/11/2025]
Abstract
Evidence from observational and Mendelian randomization (MR) studies suggested that insulin resistance (IR) was associated with Alzheimer's disease (AD). However, the causal effects of different indicators of IR on AD remain inconsistent. Here, we aim to assess the causal association between the insulin sensitivity index (ISI), a measure of post-prandial IR, and the risk of AD. We first conducted primary and secondary univariable MR analyses. We selected 8 independent genome-wide significant (p < 5E-08, primary analyses) and 61 suggestive (p < 1E-05, secondary analyses) ISI genetic variants from large-scale genome-wide association studies (GWAS; N = 53 657), respectively, and extracted their corresponding GWAS summary statistics from AD GWAS, including IGAP2019 (N = 63 926) and FinnGen_G6_AD_WIDE (N = 412 181). We selected five univariable MR methods and used heterogeneity, horizontal pleiotropy test, and leave-one-out sensitivity analysis to confirm the stability of MR estimates. Finally, we conducted a meta-analysis to combine MR estimates from two non-overlapping AD GWAS datasets. We further performed multivariable MR (MVMR) to assess the potential mediating role of type 2 diabetes (T2D) on the association between ISI and AD using two MVMR methods. In univariable MR, utilizing 8 genetic variants in primary analyses, we found a significant causal association of genetically increased ISI with decreased risk of AD (OR = 0.79, 95% CI: 0.68-0.92, p = 0.003). Utilizing 61 genetic variants in secondary analyses, we found consistent findings of a causal effect of genetically increased ISI on the decreased risk of AD (OR = 0.89, 95% CI: 0.82-0.96, p = 0.003). Heterogeneity, horizontal pleiotropy test, and leave-one-out sensitivity analysis ensured the reliability of the MR estimates. In MVMR, we found no causal relationship between ISI and AD after adjusting for T2D (p > 0.05). We provide genetic evidence that increased ISI is significantly and causally associated with reduced risk of AD, which is mediated by T2D. These findings may inform prevention strategies directed toward IR-associated T2D and AD.
Collapse
Affiliation(s)
- Fang Xu
- Department of Neurology, Xuanwu Hospital, National Center for Neurological Disorders, Capital Medical University, Beijing, China
| | - Shiyang Wu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| | - Shan Gao
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| | - Xuan Li
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| | - Chen Huang
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao SAR, China
| | - Yan Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Wannan Medical College, Wuhu, Anhui, China
| | - Ping Zhu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| | - Guiyou Liu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
- Department of Epidemiology and Biostatistics, School of Public Health, Wannan Medical College, Wuhu, Anhui, China
- Brain Hospital, Shengli Oilfield Central Hospital, Dongying, China
- Beijing Key Laboratory of Hypoxia Translational Medicine, National Engineering Laboratory of Internet Medical Diagnosis and Treatment Technology, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
62
|
Vasudevan A, Venkatesan P. Association of organ iron levels with type 2 diabetes mellitus and glycemic traits: A bidirectional two-sample Mendelian randomization study. J Trace Elem Med Biol 2025; 87:127586. [PMID: 39754912 DOI: 10.1016/j.jtemb.2024.127586] [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: 06/05/2024] [Revised: 12/25/2024] [Accepted: 12/26/2024] [Indexed: 01/06/2025]
Abstract
INTRODUCTION Observational studies have found that higher iron levels are associated with an increased risk of diabetes mellitus. Given the limitations of causal inferences from observational studies and the expensive and time-consuming nature of randomized controlled trials, Mendelian randomization analysis presents a reasonable alternative to study causal relationships. Previous MR analyses studying iron levels and diabetes have used indirect markers of iron levels, such as serum ferritin, and found conflicting results. In this study, we performed bidirectional Mendelian Randomization analyses using organ iron (liver, spleen, and pancreas) levels, which are more direct markers of iron status, to study the causal association of iron levels with type 2 diabetes mellitus and glycaemic traits. METHODS Two sample MR analyses were employed bi-directionally to study the causal effect of liver, spleen, and pancreas iron levels on type 2 diabetes and glycaemic traits and the causal effect of type 2 diabetes on organ iron levels, using summary data from genome-wide association studies (UK-Biobank, DIAGRAM, and MAGIC consortia). SNPs associated with organ iron levels with a cut-off of P < 5 × 10-7 were used as instrumental variables for the MR analyses of the effect of organ iron levels on type 2 diabetes/glycaemic traits, and SNPs associated with diabetes mellitus with a cut-off of P < 5 × 10-8 were used as instrumental variables for the MR analyses of the causal effect of type 2 diabetes on organ iron levels. Serum ferritin (GWAS meta-analysis of deCODE, UK INTERVAL, and Denmark studies) and haemoglobin (Blood Cell consortium) were used as positive controls for the MR analysis with liver iron as the exposure. Primary analyses used the inverse variance weighted means of Wald's ratio. Sensitivity analyses included inverse variance weighted median, weighted mode, and MR-Egger methods. RESULTS Our findings reveal no causal association between liver and pancreas iron levels with type 2 diabetes (Liver iron: OR = 1.02, P = 0.1, Pancreas iron: OR = 1.11, P = 0.5). This also holds for glycaemic traits, except for the negative causal effect of liver iron levels on HbA1c (OR = 0.93, P = 0.001). Spleen iron levels had a negative causal effect on type 2 diabetes (OR = 0.94, P = 0.049). However, these exceptions are likely due to possible pleiotropy, as these associations can be explained by the effect of the genetic variants on factors that falsely decrease HbA1c levels. No causal association was found for the effect of type 2 diabetes on organ iron levels. CONCLUSION Organ iron levels, which are relatively more direct indicators of iron status, showed no causal association with type 2 diabetes in the European population.
Collapse
Affiliation(s)
- Akshaya Vasudevan
- Department of Community Medicine, Christian Medical College, Vellore, Tamil Nadu, India; Affiliated to The Tamil Nadu Dr. MGR Medical University, Chennai, India.
| | - Padmanaban Venkatesan
- Department of Biochemistry, Christian Medical College, Vellore, Tamil Nadu, India; Affiliated to The Tamil Nadu Dr. MGR Medical University, Chennai, India.
| |
Collapse
|
63
|
Mao W, Wang G, Wang X, Shen Y, Yuan S, Wang L, Yang H, Li Y, Chen K, Liu J, Dong X, Zhao Y, Mu L. Glucokinase Regulatory Protein as a Putative Target for Gestational Diabetes Mellitus and Related Complications: Evidence From the Mendelian Randomization Study. J Diabetes 2025; 17:e70056. [PMID: 39921581 PMCID: PMC11806411 DOI: 10.1111/1753-0407.70056] [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: 10/24/2024] [Revised: 01/04/2025] [Accepted: 01/25/2025] [Indexed: 02/10/2025] Open
Abstract
BACKGROUND Gestational diabetes mellitus (GDM) is one of the most common complications of pregnancy and is highly associated with adverse perinatal outcomes and long-term health problems for the mother and offspring. However, there are respective limitations in the pharmacological strategies for the current treatment of GDM. Glucokinase regulatory protein (GCKR) has been associated with GDM in observational studies and animal experiments and thus represents a potential drug target of interest for investigation. METHODS We applied two-sample Mendelian randomization (MR) and colocalization analysis using summary-level data from genome-wide association studies of GCKR and GDM. Two-step MR was used to explore the mediating effects of several metabolic factors on the association. We also applied MR to explore the associations of GCKR levels with GDM-related outcomes. Finally, we performed a phenome-wide association study (PheWAS) to query the potential effects of altered GCKR levels across multiple health categories. RESULTS We found a significant association between elevated GCKR levels and GDM (OR = 3.466, 95% CI = 2.401-5.002, p = 3.16 × 10-11), also supported by the colocalization analysis ([Pcoloc] = 0.997). The estimates were replicated in an independent study (OR = 2.640, 95% CI = 1.983-3.513, p = 2.84 × 10-11, Pcoloc = 0.983). Elevated GCKR levels were also associated with higher risk of type 2 diabetes (OR = 2.183, 95% CI = 1.846-2.581, p = 6.53 × 10-20). Two-step MR suggested that fasting glucose, fasting insulin, and triglycerides partly mediated the causal relationship. PheWAS found that targeting GCKR may improve renal function and glucose homeostasis but cause dyslipidemia and uric acid abnormalities. CONCLUSIONS This study provided novel evidence that circulating GCKR levels are causally implicated in GDM and related complications, suggesting that it may be a promising target for treatment.
Collapse
Affiliation(s)
- Weian Mao
- Reproductive Medicine Center, Zhongshan HospitalFudan UniversityShanghaiChina
- Department of Obstetrics and GynecologyThe Second Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Guiquan Wang
- Department of Reproductive Medicine, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China; Xiamen Key Laboratory of Reproduction and GeneticsXiamenChina
| | - Xiao Wang
- Department of ObstetricsThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Yan Shen
- The First School of MedicineWenzhou Medical UniversityWenzhouChina
| | - Shuai Yuan
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental MedicineKarolinska InstitutetStockholmSweden
| | - Lin Wang
- Reproductive Medicine Center, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Haiyan Yang
- Reproductive Medicine CenterThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Yan Li
- Reproductive Medicine CenterThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Kai Chen
- Center for Reproductive Medicine, Department of Obstetrics and GynecologyPeking University Third HospitalBeijingChina
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third HospitalBeijingChina
- Key Laboratory of Assisted Reproduction, Ministry of EducationBeijingChina
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive TechnologyBeijingChina
| | - Jun Liu
- Nuffield Department of Population HealthUniversity of OxfordOxfordUK
| | - Xi Dong
- Reproductive Medicine Center, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Yue Zhao
- Center for Reproductive Medicine, Department of Obstetrics and GynecologyPeking University Third HospitalBeijingChina
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third HospitalBeijingChina
- Key Laboratory of Assisted Reproduction, Ministry of EducationBeijingChina
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive TechnologyBeijingChina
| | - Liangshan Mu
- Reproductive Medicine Center, Zhongshan HospitalFudan UniversityShanghaiChina
| |
Collapse
|
64
|
Yang LZ, Yang Y, Hong C, Wu QZ, Shi XJ, Liu YL, Chen GZ. Systematic Mendelian Randomization Exploring Druggable Genes for Hemorrhagic Strokes. Mol Neurobiol 2025; 62:1359-1372. [PMID: 38977622 PMCID: PMC11772512 DOI: 10.1007/s12035-024-04336-9] [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: 12/30/2023] [Accepted: 06/26/2024] [Indexed: 07/10/2024]
Abstract
Patients with hemorrhagic stroke have high rates of morbidity and mortality, and drugs for prevention are very limited. Mendelian randomization (MR) analysis can increase the success rate of drug development by providing genetic evidence. Previous MR analyses only analyzed the role of individual drug target genes in hemorrhagic stroke; therefore, we used MR analysis to systematically explore the druggable genes for hemorrhagic stroke. We sequentially performed summary-data-based MR analysis and two-sample MR analysis to assess the associations of all genes within the database with intracranial aneurysm, intracerebral hemorrhage, and their subtypes. Validated genes were further analyzed by colocalization. Only genes that were positive in all three analyses and were druggable were considered desirable genes. We also explored the mediators of genes affecting hemorrhagic stroke incidence. Finally, the associations of druggable genes with other cardiovascular diseases were analyzed to assess potential side effects. We identified 56 genes that significantly affected hemorrhagic stroke incidence. Moreover, TNFSF12, SLC22A4, SPARC, KL, RELT, and ADORA3 were found to be druggable. The inhibition of TNFSF12, SLC22A4, and SPARC can reduce the risk of intracranial aneurysm, subarachnoid hemorrhage, and intracerebral hemorrhage. Gene-induced hypertension may be a potential mechanism by which these genes cause hemorrhagic stroke. We also found that blocking these genes may cause side effects, such as ischemic stroke and its subtypes. Our study revealed that six druggable genes were associated with hemorrhagic stroke, and the inhibition of TNFSF12, SLC22A4, and SPARC had preventive effects against hemorrhagic strokes.
Collapse
Affiliation(s)
- Lun-Zhe Yang
- Department of Neurosurgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yong Yang
- Department of Neurosurgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Chuan Hong
- Department of Neurosurgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Qi-Zhe Wu
- Department of Neurosurgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Xiong-Jie Shi
- Department of Neurosurgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yi-Lin Liu
- Department of Neurosurgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Guang-Zhong Chen
- Department of Neurosurgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
| |
Collapse
|
65
|
Tan LS, Lau HH, Abdelalim EM, Khoo CM, O'Brien RM, Tai ES, Teo AKK. The role of glucose-6-phosphatase activity in glucose homeostasis and its potential for diabetes therapy. Trends Mol Med 2025; 31:152-164. [PMID: 39426930 DOI: 10.1016/j.molmed.2024.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 09/13/2024] [Accepted: 09/20/2024] [Indexed: 10/21/2024]
Abstract
Glucose-6-phosphatase catalytic subunit (G6PC)1 and G6PC2 are crucial for glucose metabolism, regulating processes like glycolysis, gluconeogenesis, and glycogenolysis. Despite their structural and functional similarities, G6PC1 and G6PC2 exhibit distinct tissue-specific expression patterns, G6P hydrolysis kinetics, and physiological functions. This review provides a comprehensive overview of their enzymology and distinct roles in glucose homeostasis. We examine how inactivating mutations in G6PC1 lead to glycogen storage disease, and how elevated G6PC1 and G6PC2 expression can affect the incidence of diabetic complications, risk for type 2 diabetes mellitus (T2DM) and various cancers. We also discuss the potential of inhibiting G6PC1 and G6PC2 to protect against complications from elevated blood glucose levels, and highlight drug development efforts targeting G6PC1 and G6PC2, and the therapeutic potential of inhibitors for disease prevention.
Collapse
Affiliation(s)
- Lay Shuen Tan
- Stem Cells and Diabetes Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore; Dean's Office, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Hwee Hui Lau
- Stem Cells and Diabetes Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Essam M Abdelalim
- Laboratory of Pluripotent Stem Cell Disease Modeling, Translational Medicine Department, Research Branch, Sidra Medicine, P.O. Box 26999, Doha, Qatar; College of Health and Life Sciences, Hamad Bin Khalifa University (HBKU), Qatar Foundation, Education City, Doha, Qatar
| | - Chin Meng Khoo
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Precision Medicine Translational Research Program (TRP), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Richard M O'Brien
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - E Shyong Tai
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Precision Medicine Translational Research Program (TRP), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Adrian Kee Keong Teo
- Stem Cells and Diabetes Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore; Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Precision Medicine Translational Research Program (TRP), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
66
|
Fragoso-Bargas N, Mcbride NS, Lee-Ødegård S, Lawlor DA, Yousefi PD, Moen GH, Opsahl JO, Jenum AK, Franks PW, Prasad RB, Qvigstad E, Birkeland KI, Richardsen KR, Sommer C. Epigenome-wide association study of objectively measured physical activity in peripheral blood leukocytes. BMC Genomics 2025; 26:62. [PMID: 39844050 PMCID: PMC11755845 DOI: 10.1186/s12864-025-11262-0] [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: 07/16/2024] [Accepted: 01/17/2025] [Indexed: 01/24/2025] Open
Abstract
BACKGROUND Few studies have explored the association between DNA methylation and physical activity. The aim of this study was to evaluate the association of objectively measured hours of sedentary behavior (SB) and moderate physical activity (MPA) with DNA methylation. We further aimed to explore the association between SB or MPA related CpG sites and cardiometabolic traits, gene expression, and genetic variation. RESULTS For discovery, we performed cross sectional analyses in pregnant women from the Epigenetics in pregnancy (EPIPREG) sample with both DNA methylation (Illumina MethylationEPIC BeadChip) and objectively measured physical activity data (SenseWear™ Pro 3 armband) (European = 244, South Asian = 109). For EWAS of SB and MPA, two main models were designed: model (1) a linear mixed model adjusted for age, smoking, blood cell composition, including ancestry as random intercept, and model (2) which was additionally adjusted for the total number of steps per day. In model 1, we did not identify any CpG sites associated with neither SB nor MPA. In model 2, SB was positively associated (false discovery rate, FDR < 0.05) with two CpG sites within the VSX1 gene. Both CpG sites were positively associated with BMI and were associated with several genetic variants in cis. MPA was associated with 122 significant CpG sites at FDR < 0.05 (model 2). We further analyzed the ten most statistically significant MPA related CpG sites and found that they presented opposite associations with sedentary behavior and BMI. We were not able to replicate the SB and MPA-related CpG sites in the Avon Longitudinal Study of Parents and Children (ALSPAC). ALSPAC had available objectively measured physical activity data from Actigraph (without steps/day available) and leucocyte DNA methylation data collected during adolescence (n = 408, European). CONCLUSION This study suggests associations of objectively measured SB and MPA with maternal DNA methylation in peripheral blood leukocytes, that needs to be confirmed in larger samples of similar study design.
Collapse
Affiliation(s)
- Nicolas Fragoso-Bargas
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway.
- Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital, Oslo, Norway.
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.
| | - Nancy S Mcbride
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, United Kingdom
- Population Health Science, Bristol Medical School, University of Bristol, Bristol, UK
| | - Sindre Lee-Ødegård
- Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Deborah A Lawlor
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, United Kingdom
- Population Health Science, Bristol Medical School, University of Bristol, Bristol, UK
| | - Paul D Yousefi
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, United Kingdom
- Population Health Science, Bristol Medical School, University of Bristol, Bristol, UK
- NIHR Bristol Biomedical Research Centre, University Hospitals Bristol and Weston NHS Foundation Trust and University of Bristol, Bristol, UK
| | - Gunn-Helen Moen
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Institute of Molecular Bioscience, The University of Queensland, Brisbane, Australia
- Department of Public Health and Nursing, K.G. Jebsen Center for Genetic Epidemiology, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- The Frazer Institute, The University of Queensland, Woolloongabba, Australia
| | - Julia O Opsahl
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Anne Karen Jenum
- General Practice Research Unit (AFE), Department of General Practice, Institute of Health and Society, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Paul W Franks
- Lund University Diabetes Centre, Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Rashmi B Prasad
- Lund University Diabetes Centre, Department of Clinical Sciences, Lund University, Malmö, Sweden
- Institute for Molecular Medicine Finland FIMM, Helsinki University, Helsinki, Finland
| | - Elisabeth Qvigstad
- Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Kåre I Birkeland
- Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Kåre R Richardsen
- Faculty of Health Sciences, Department of Rehabilitation Science and Health Technology, Oslo Metropolitan University, Oslo, Norway
| | - Christine Sommer
- Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
67
|
Huang JX, Xu SZ, Tian T, Wang J, Jiang LQ, He T, Meng SY, Ni J, Pan HF. Genetic Links Between Metabolic Syndrome and Osteoarthritis: Insights From Cross-Trait Analysis. J Clin Endocrinol Metab 2025; 110:e461-e469. [PMID: 38482593 DOI: 10.1210/clinem/dgae169] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Indexed: 01/22/2025]
Abstract
CONTEXT Previous observational studies have indicated a bidirectional association between metabolic syndrome (MetS) and osteoarthritis (OA). However, it remains unclear whether these bidirectional associations reflect causal relationships or shared genetic factors, and the underlying biological mechanisms of this association are not fully understood. OBJECTIVE We aimed to explore the genetic connection between MetS and OA using genome-wide association study (GWAS) summary data. METHODS Leveraging summary statistics from GWAS conducted by the UK Biobank and the Glucose and Insulin-related Traits Consortium (MAGIC), we performed global genetic correlation analyses, genome-wide cross-trait meta-analyses, and a bidirectional two-sample Mendelian randomization analyses using summary statistics from GWAS to comprehensively assess the relationship of MetS and OA. RESULTS We first detected an extensive genetic correlation between MetS and OA (rg = 0.393, P = 1.52 × 10-18), which was consistent in 4 MetS components, including waist circumference, triglycerides, hypertension, and high-density lipoprotein cholesterol and OA with rg ranging from -0.229 to 0.490. We then discovered 32 variants jointly associated with MetS and OA through Multi-Trait Analysis of GWAS (MTAG). Co-localization analysis found 12 genes shared between MetS and OA, with functional implications in several biological pathways. Finally, Mendelian randomization analysis suggested genetic liability to MetS significantly increased the risk of OA, but no reverse causality was found. CONCLUSION Our results illustrate a common genetic architecture, pleiotropic loci, as well as causality between MetS and OA, potentially enhancing our knowledge of high comorbidity and genetic processes that overlap between the 2 disorders.
Collapse
Affiliation(s)
- Ji-Xiang Huang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China
- Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, Anhui 230032, China
| | - Shu-Zhen Xu
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China
- Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, Anhui 230032, China
| | - Tian Tian
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China
- Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, Anhui 230032, China
| | - Jing Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China
- Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, Anhui 230032, China
| | - Ling-Qiong Jiang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China
- Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, Anhui 230032, China
| | - Tian He
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China
- Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, Anhui 230032, China
| | - Shi-Yin Meng
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China
- Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, Anhui 230032, China
| | - Jing Ni
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China
- Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, Anhui 230032, China
| | - Hai-Feng Pan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China
- Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, Anhui 230032, China
| |
Collapse
|
68
|
Zhang S, Bian W, Wang Y, Shen M, Qian Y, Dai H, Zheng S, Fu Q, Xu K, Yang T, Jiang H. The MTNR1B Rs724030 variant is associated with islet function and women waist-to-hip ratio in healthy subjects. Front Endocrinol (Lausanne) 2025; 15:1398687. [PMID: 39886031 PMCID: PMC11779613 DOI: 10.3389/fendo.2024.1398687] [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: 03/10/2024] [Accepted: 12/26/2024] [Indexed: 02/01/2025] Open
Abstract
Objective This study aims to investigate the associations between MTNR1B rs724030 A>G variant and prediabetes risk, along with their correlations with clinical features, including plasma glucose and serum insulin levels during oral glucose tolerance test (OGTT), islet function, insulin resistance, and plasma lipid levels. In particular, we investigated whether there are sex dimorphisms in the impact of this variant on islet function/insulin resistance. Methods We included 3415 glucose-tolerant healthy and 1744 prediabetes individuals based on OGTT. Binary logistic regression was performed to evaluate the relationships between rs724030 in MTNR1B and prediabetes under the additive model. Additionally, multiple linear regression was utilized to investigate the associations between this variant and glycemic-related quantitative traits and lipid levels. Results While no association was observed between the rs724030 variant in MTNR1B and prediabetes risk in the overall cohort (P > 0.05), we found the G allele of this variant was associated with higher fasting and 30-minute plasma glucose levels, decreased Insulinogenic Index (IGI), and oral disposition index (DIo) (P = 0.009, 0.001, 0.001, and 0.007, respectively) in the normal glucose tolerance (NGT) individuals with normal BMI levels. Furthermore, we also found significant associations between this variant and IGI, corrected insulin response (CIR), and DIo (All P < 0.001) in female individuals whose waist-to-hip ratio (WHR) is greater than 0.85, with considerable heterogeneity (Phet = 0.009, 0.030, and 0.049, respectively) to male participants in the NGT individuals, but not in the impaired fasting glucose (IFG)/impaired glucose tolerance (IGT) individuals. Additionally, no association was observed between this variant and insulin clearance (All P > 0.05). Conclusions The MTNR1B rs724030 variant contributes to glycemic traits and islet function, and its effects have sex dimorphisms in the NGT individuals after stratifying by WHR. All these findings provide a basis for accurately assessing islet function in healthy populations and offer a new perspective on precision prevention.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Tao Yang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hemin Jiang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
69
|
Yuan C, Shu X, Wang X, Chen W, Li X, Pei W, Su X, Hu Z, Jie Z. The impact of metabolic syndrome on hepatocellular carcinoma: a mendelian randomization study. Sci Rep 2025; 15:1941. [PMID: 39809981 PMCID: PMC11733230 DOI: 10.1038/s41598-025-86317-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 01/09/2025] [Indexed: 01/16/2025] Open
Abstract
Traditional epidemiological studies are susceptible to confounding factors. To clarify the impact of metabolic syndrome and its diagnostic components on hepatocellular carcinoma, we conducted a preliminary mendelian randomization analysis with metabolic syndrome and its diagnostic components as exposures and hepatocellular carcinoma as the outcome. Another set of genetic data related to hepatocellular carcinoma was used as a validation cohort, repeating the mendelian randomization analysis and combining the two groups for a meta-analysis. Preliminary analysis showed that metabolic syndrome (P-value = 0.002) and waist circumference (P-value = 0.026) are significantly positively correlated with an increased risk of hepatocellular carcinoma. After multiple testing corrections, metabolic syndrome (PFDR-value = 0.013) remained significant, although the association between waist circumference (PFDR-value = 0.079) and hepatocellular carcinoma was considered suggestive, the meta-analysis further confirmed the impact of metabolic syndrome (P-value = 0.0002) and waist circumference (P-value = 0.0038) in increasing the risk of hepatocellular carcinoma. After adjusting for the genetic predictive effects of all exposures, waist circumference was found to be a key factor significantly influencing the relationship between metabolic syndrome and hepatocellular carcinoma. In summary, our study indicates that metabolic syndrome increases the risk of hepatocellular carcinoma, particularly among individuals with a larger waist circumference.
Collapse
Affiliation(s)
- Chendong Yuan
- Medical Innovation Center, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwai Main Street, Nanchang, 330006, Jiangxi, China
- Department of General Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Xufeng Shu
- Medical Innovation Center, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwai Main Street, Nanchang, 330006, Jiangxi, China
- Department of General Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Xiaoqiang Wang
- Medical Innovation Center, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwai Main Street, Nanchang, 330006, Jiangxi, China
- Department of General Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Wenzheng Chen
- Medical Innovation Center, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwai Main Street, Nanchang, 330006, Jiangxi, China
- Department of General Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Xin Li
- Department of Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Wenguang Pei
- Medical Innovation Center, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwai Main Street, Nanchang, 330006, Jiangxi, China
- Department of General Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Xujie Su
- Medical Innovation Center, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwai Main Street, Nanchang, 330006, Jiangxi, China
- Department of General Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Zhenzhen Hu
- Department of Anesthesiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwai Main Street, Nanchang, 330006, Jiangxi, China.
| | - Zhigang Jie
- Medical Innovation Center, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwai Main Street, Nanchang, 330006, Jiangxi, China.
- Department of General Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
70
|
Khan SR, Ye WW, Van JAD, Singh I, Rabiee Y, Rodricks KL, Zhang X, Nicholson RJ, Razani B, Summers SA, Futerman AH, Gunderson EP, Wheeler MB. Reduced circulating sphingolipids and CERS2 activity are linked to T2D risk and impaired insulin secretion. SCIENCE ADVANCES 2025; 11:eadr1725. [PMID: 39792658 PMCID: PMC11790001 DOI: 10.1126/sciadv.adr1725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 12/09/2024] [Indexed: 01/12/2025]
Abstract
Gestational diabetes mellitus (GDM), a transient form of diabetes that resolves postpartum, is a major risk factor for type 2 diabetes (T2D) in women. While the progression from GDM to T2D is not fully understood, it involves both genetic and environmental components. By integrating clinical, metabolomic, and genome-wide association study (GWAS) data, we identified associations between decreased sphingolipid biosynthesis and future T2D, in part through the rs267738 allele of the CERS2 gene in Hispanic women shortly after a GDM pregnancy. To understand the impact of the CERS2 gene and risk allele on glucose regulation, we examined whole-body Cers2 knockout and rs267738 knock-in mice. Both models exhibited glucose intolerance and impaired insulin secretion in vivo. Islets isolated from these models also demonstrated reduced β cell function, as shown by decreased insulin secretion ex vivo. Overall, reduced circulating sphingolipids may indicate a high risk of GDM-to-T2D progression and reflect deficits in CERS2 activity that negatively affect glucose homeostasis and β cell function.
Collapse
Affiliation(s)
- Saifur R. Khan
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- VA Medical Center, Pittsburgh, PA, USA
- Center for Immunometabolism, University of Pittsburgh, Pittsburgh, PA, USA
| | - Wenyue W. Ye
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Julie A. D. Van
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Ishnoor Singh
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Yasmin Rabiee
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | | | - Xiangyu Zhang
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- VA Medical Center, Pittsburgh, PA, USA
- Center for Immunometabolism, University of Pittsburgh, Pittsburgh, PA, USA
| | - Rebekah J. Nicholson
- Departments of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
| | - Babak Razani
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- VA Medical Center, Pittsburgh, PA, USA
- Center for Immunometabolism, University of Pittsburgh, Pittsburgh, PA, USA
| | - Scott A. Summers
- Departments of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
| | - Anthony H. Futerman
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Erica P. Gunderson
- Division of Research, Kaiser Permanente Northern California, Pleasanton, CA, USA
- Department of Health Systems Science, Kaiser Permanente Bernard J. Tyson School of Medicine, Pasadena, CA, USA
| | - Michael B. Wheeler
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
71
|
Biswas S, Hilser JR, Woodward NC, Wang Z, Gukasyan J, Nemet I, Schwartzman WS, Huang P, Han Y, Fouladian Z, Charugundla S, Spencer NJ, Pan C, Tang WHW, Lusis AJ, Hazen SL, Hartiala JA, Allayee H. Exploring the Role of Glycine Metabolism in Coronary Artery Disease: Insights from Human Genetics and Mouse Models. Nutrients 2025; 17:198. [PMID: 39796632 PMCID: PMC11723402 DOI: 10.3390/nu17010198] [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: 11/21/2024] [Revised: 12/19/2024] [Accepted: 12/31/2024] [Indexed: 01/13/2025] Open
Abstract
Background: Circulating glycine levels have been associated with reduced risk of coronary artery disease (CAD) in humans but these associations have not been observed in all studies. We evaluated whether the relationship between glycine levels and atherosclerosis was causal using genetic analyses in humans and feeding studies in mice. Methods: Serum glycine levels were evaluated for association with risk of CAD in the UK Biobank. Genetic determinants of glycine levels were identified through a genome-wide association study (GWAS) and used to evaluate the causal relationship between glycine and risk of CAD by Mendelian randomization (MR). A dietary supplementation study was carried out with atherosclerosis-prone apolipoprotein E deficient (ApoE-/-) mice to determine the effects of increased circulating glycine levels on cardiometabolic traits and aortic lesion formation. Results: Among 105,718 UK Biobank subjects, elevated serum glycine levels were associated with significantly reduced risk of prevalent CAD (Quintile 5 vs. Quintile 1 OR = 0.76, 95% CI 0.67-0.87; p < 0.0001) and incident CAD (Quintile 5 vs. Quintile 1 HR = 0.70, 95% CI 0.65-0.77; p < 0.0001) after adjustment for age, sex, ethnicity, anti-hypertensive and lipid-lowering medications, blood pressure, kidney function, and diabetes. A GWAS meta-analysis with 230,947 subjects identified 61 loci for glycine levels, of which 26 were novel. MR analyses provided modest evidence that genetically elevated glycine levels were causally associated with reduced systolic blood pressure and risk of type 2 diabetes, but did not provide significant evidence for an association with decreased risk of CAD. Glycine supplementation in mice had no effects on cardiometabolic traits or atherosclerotic lesion development. Conclusions: While expanding the genetic architecture of glycine metabolism, MR analyses and in vivo feeding studies did not provide evidence that the clinical association of this amino acid with atherosclerosis represents a causal relationship.
Collapse
Affiliation(s)
- Subarna Biswas
- Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - James R. Hilser
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Nicholas C. Woodward
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Zeneng Wang
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Microbiome and Human Health, Cleveland Clinic, Cleveland, OH 44195, USA
- Department of Cardiovascular Medicine, Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Janet Gukasyan
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Ina Nemet
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Microbiome and Human Health, Cleveland Clinic, Cleveland, OH 44195, USA
| | - William S. Schwartzman
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Pin Huang
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Yi Han
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Zachary Fouladian
- Department of Medicine, David Geffen School of Medicine of UCLA, Los Angeles, CA 90095, USA
| | - Sarada Charugundla
- Department of Medicine, David Geffen School of Medicine of UCLA, Los Angeles, CA 90095, USA
| | - Neal J. Spencer
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Calvin Pan
- Department of Human Genetics, David Geffen School of Medicine of UCLA, Los Angeles, CA 90095, USA
| | - W. H. Wilson Tang
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Microbiome and Human Health, Cleveland Clinic, Cleveland, OH 44195, USA
- Department of Cardiovascular Medicine, Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Aldons J. Lusis
- Department of Medicine, David Geffen School of Medicine of UCLA, Los Angeles, CA 90095, USA
- Department of Human Genetics, David Geffen School of Medicine of UCLA, Los Angeles, CA 90095, USA
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine of UCLA, Los Angeles, CA 90095, USA
| | - Stanley L. Hazen
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Microbiome and Human Health, Cleveland Clinic, Cleveland, OH 44195, USA
- Department of Cardiovascular Medicine, Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Jaana A. Hartiala
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Hooman Allayee
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
72
|
Wang R, Sun J, Yu X. Mediators of the association between nut consumption and cardiovascular diseases: a two-step mendelian randomization study. Sci Rep 2025; 15:829. [PMID: 39755742 PMCID: PMC11700201 DOI: 10.1038/s41598-024-85070-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 12/31/2024] [Indexed: 01/06/2025] Open
Abstract
Previous observational studies have reported inconsistent associations between nut consumption and cardiovascular diseases (CVD). This study aims to identify the causal relationship between different types of nuts consumption and CVD, and to quantify the potential mediating effects of cardiometabolic factors. We utilized Genome-Wide Association Study (GWAS) data to assess the causal effects of nut consumption on CVD using two-sample Mendelian randomization (MR) and a two-step MR analysis. The inverse variance weighted (IVW) method indicated that processed (salted or roasted) peanuts were potentially and positively associated with ischaemic heart disease (IHD) (OR 1.4866; 95%CI 1.0491-2.1065). No causal relationships were found between nuts consumption and other CVD outcomes, including atrial fibrillation, angina, coronary atherosclerosis, coronary heart disease, IHD, myocardial infarction, subarachnoid hemorrhage, intracerebral haemorrhage and stroke. Both MR-Egger and median-based methods yielded similar results to IVW. Furthermore, in the two-step MR analysis, fasting insulin, low-density lipoprotein cholesterol and fasting blood glucose were identified as mediators in the potential causal relationship between processed peanuts and IHD, explaining 16.98%, 6.38% and 4.91% of the mediation, respectively. In total, these mediators accounted for 28.27% of the association between salted or roasted peanuts and IHD.
Collapse
Affiliation(s)
- Ruizhe Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Southeast University, Nanjing, People's Republic of China
| | - Jinfang Sun
- Department of Epidemiology and Biostatistics, School of Public Health, Southeast University, Nanjing, People's Republic of China
| | - Xiaojin Yu
- Department of Epidemiology and Biostatistics, School of Public Health, Southeast University, Nanjing, People's Republic of China.
| |
Collapse
|
73
|
Xing X, Xu S, Wang Y, Shen Z, Wen S, Zhang Y, Ruan G, Cai G. Evaluating the Causal Effect of Circulating Proteome on Glycemic Traits: Evidence From Mendelian Randomization. Diabetes 2025; 74:108-119. [PMID: 39418314 DOI: 10.2337/db24-0262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 09/30/2024] [Indexed: 10/19/2024]
Abstract
Exploring the mechanisms underlying abnormal glycemic traits is important for deciphering type 2 diabetes and characterizing novel drug targets. This study aimed to decipher the causal associations of circulating proteins with fasting glucose (FG), 2-h glucose after an oral glucose challenge (2hGlu), fasting insulin (FI), and glycated hemoglobin (HbA1c) using large-scale proteome-wide Mendelian randomization (MR) analyses. Genetic data on plasma proteomes were obtained from 10 proteomic genome-wide association studies. Both cis-protein quantitative trait loci (pQTLs) and cis + trans-pQTLs MR analyses were conducted. Bayesian colocalization, Steiger filtering analysis, assessment of protein-altering variants, and mapping expression QTLs to pQTLs were performed to investigate the reliability of the MR findings. Protein-protein interaction, pathway enrichment analysis, and evaluation of drug targets were performed. Thirty-three proteins were identified with causal effects on FG, FI, or HbA1c but not 2hGlu in the cis-pQTL analysis, and 93 proteins had causal effects on glycemic traits in the cis + trans-pQTLs analysis. Most proteins were either considered druggable or drug targets. In conclusion, many novel circulating protein biomarkers were identified to be causally associated with glycemic traits. These biomarkers enhance the understanding of molecular etiology and provide insights into the screening, monitoring, and treatment of diabetes. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Xing Xing
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Siqi Xu
- Clinical Research Centre, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Yining Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Ziyuan Shen
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Simin Wen
- Clinical Research Centre, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Yan Zhang
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Guangfeng Ruan
- Clinical Research Centre, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Guoqi Cai
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| |
Collapse
|
74
|
Niu Z, Cao L, Guo W, Zhang H. Associations between Type 2 Diabetes Mellitus, Metabolic Traits, and Abdominal Aortic Aneurysm: A Cross-Ethnic Mendelian Randomization Analysis. Ann Vasc Surg 2025; 110:405-413. [PMID: 39103013 DOI: 10.1016/j.avsg.2024.07.105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/23/2024] [Accepted: 07/12/2024] [Indexed: 08/07/2024]
Abstract
BACKGROUND Evidence suggests that type 2 diabetes mellitus (T2DM) may protect from abdominal aortic aneurysm (AAA). However, it is unclear whether a causal relationship exists between these 2 conditions and, if so, whether it remains consistent among racial groups. METHODS Cross-ethnic Mendelian randomization (MR) was used to examine the causal relationships between T2DM, metabolic traits, and AAA. Inverse variance weighted (IVW) was the primary analysis tool, supplemented by MR-Egger, weighted median, and MR Pleiotropy RESidual Sum and Outlier. Heterogeneity and horizontal pleiotropy were assessed using the Cochran's Q test and MR-Egger intercept, respectively. RESULTS According to IVW, an inverse correlation between T2DM and AAA was detected in Europeans (odds ratio [OR] 0.91, 95% confidence interval [CI] 0.84-0.99; P = 0.034) and East Asians (OR 0.87, 95% CI 0.77-0.99; P = 0.038). Fasting glucose was inversely associated with AAA in Europeans (OR 0.56, 95% CI 0.33-0.96; P = 0.034) but not in East Asians. In Europeans, fasting insulin was a risk factor for AAA (OR 3.03, 95% CI 1.53-6.01; P = 0.001), while 2-hour glucose was protective (OR 0.67, 95% CI 0.49-0.91; P = 0.011). Glycated hemoglobin (HbA1c) had no effect. Insufficient instrumental variables prevented the evaluation of the relationships of fasting insulin, HbA1c, and 2-hour glucose with AAA in East Asians. CONCLUSIONS T2DM protects against AAA in Europeans and East Asians. The effects of different glucose metabolism characteristics on AAA may inform AAA treatment.
Collapse
Affiliation(s)
- Zelin Niu
- Department of Vascular and Endovascular Surgery, Chinese PLA General Hospital, Beijing, China; Medical School of Chinese PLA, Beijing, China
| | - Long Cao
- Department of Vascular and Endovascular Surgery, Chinese PLA General Hospital, Beijing, China; Department of General Surgery, Chinese PLA No. 983 Hospital, Tianjin, China
| | - Wei Guo
- Department of Vascular and Endovascular Surgery, Chinese PLA General Hospital, Beijing, China
| | - Hongpeng Zhang
- Department of Vascular and Endovascular Surgery, Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
75
|
Brotman SM, El-Sayed Moustafa JS, Guan L, Broadaway KA, Wang D, Jackson AU, Welch R, Currin KW, Tomlinson M, Vadlamudi S, Stringham HM, Roberts AL, Lakka TA, Oravilahti A, Fernandes Silva L, Narisu N, Erdos MR, Yan T, Bonnycastle LL, Raulerson CK, Raza Y, Yan X, Parker SCJ, Kuusisto J, Pajukanta P, Tuomilehto J, Collins FS, Boehnke M, Love MI, Koistinen HA, Laakso M, Mohlke KL, Small KS, Scott LJ. Adipose tissue eQTL meta-analysis highlights the contribution of allelic heterogeneity to gene expression regulation and cardiometabolic traits. Nat Genet 2025; 57:180-192. [PMID: 39747594 DOI: 10.1038/s41588-024-01982-6] [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: 09/21/2023] [Accepted: 10/11/2024] [Indexed: 01/04/2025]
Abstract
Complete characterization of the genetic effects on gene expression is needed to elucidate tissue biology and the etiology of complex traits. In the present study, we analyzed 2,344 subcutaneous adipose tissue samples and identified 34,774 conditionally distinct expression quantitative trait locus (eQTL) signals at 18,476 genes. Over half of eQTL genes exhibited at least two eQTL signals. Compared with primary eQTL signals, nonprimary eQTL signals had lower effect sizes, lower minor allele frequencies and less promoter enrichment; they corresponded to genes with higher heritability and higher tolerance for loss of function. Colocalization of eQTLs with genome-wide association study (GWAS) signals for 28 cardiometabolic traits identified 1,835 genes. Inclusion of nonprimary eQTL signals increased discovery of colocalized GWAS-eQTL signals by 46%. Furthermore, 21 genes with ≥2 colocalized GWAS-eQTL signals showed a mediating gene dosage effect on the GWAS trait. Thus, expanded eQTL identification reveals more mechanisms underlying complex traits and improves understanding of the complexity of gene expression regulation.
Collapse
Affiliation(s)
- Sarah M Brotman
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | | | - Li Guan
- Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - K Alaine Broadaway
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Dongmeng Wang
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Anne U Jackson
- Department of Biostatistics and Center for Statistical Genetics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Ryan Welch
- Department of Biostatistics and Center for Statistical Genetics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Kevin W Currin
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Max Tomlinson
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
- Department of Medical and Molecular Genetics, King's College London, London, UK
| | | | - Heather M Stringham
- Department of Biostatistics and Center for Statistical Genetics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Amy L Roberts
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - 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
| | - Anniina Oravilahti
- Institute of Clinical Medicine, Kuopio University Hospital, University of Eastern Finland, Kuopio, Finland
| | - Lilian Fernandes Silva
- Institute of Clinical Medicine, Kuopio University Hospital, University of Eastern Finland, Kuopio, Finland
| | - Narisu Narisu
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Michael R Erdos
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Tingfen Yan
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Lori L Bonnycastle
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | | | - Yasrab Raza
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Xinyu Yan
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Stephen C J Parker
- Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, MI, USA
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Johanna Kuusisto
- Department of Medicine and Clinical Research, Kuopio University Hospital, Kuopio, Finland
| | - Päivi Pajukanta
- Department of Human Genetics and Institute for Precision Health, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Jaakko Tuomilehto
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
- Department of Public Health, University of Helsinki, Helsinki, Finland
- Diabetes Research Group, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Francis S Collins
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Michael I Love
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
- Department of Biostatistics, University of North Carolina, Chapel Hill, NC, USA
| | - Heikki A Koistinen
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
- University of Helsinki and Department of Medicine, Helsinki University Hospital, Helsinki, Finland
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Markku Laakso
- Institute of Clinical Medicine, Kuopio University Hospital, University of Eastern Finland, Kuopio, Finland
- Department of Medicine and Clinical Research, Kuopio University Hospital, Kuopio, Finland
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA.
| | - Kerrin S Small
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK.
| | - Laura J Scott
- Department of Biostatistics and Center for Statistical Genetics, School of Public Health, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
76
|
Ru X, Huang L, Su Z, Ye C, Guo Y. Exploring the causal relationship between asthma in the metabolic syndrome: a Mendelian randomization study. J Asthma 2025; 62:167-177. [PMID: 39163002 DOI: 10.1080/02770903.2024.2394143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 08/14/2024] [Indexed: 08/21/2024]
Abstract
BACKGROUND Previous observational studies have indicated a potential association between metabolic syndrome (MetS) and asthma, though the causal nature of this connection is still uncertain. Our study used Mendelian randomization (MR) to examine the causal relationship between metabolic syndrome (MetS) and its components with asthma. METHODS This study utilized single nucleotide polymorphisms (SNPs) related to MetS and its components, sourced from publicly available genome-wide association studies (GWAS) data, in combination with asthma data from the FinnGen database. Statistical analyses were conducted using the inverse variance weighted method (IVW), MR-Egger, and weighted median method. The robustness of the findings was confirmed through various sensitivity analyses. RESULTS The IVW analysis indicated that MetS was associated with an increased risk of asthma (OR = 1.0781, 95% CI = 1.0255-1.1333, p = 0.0032). Among the components of MetS, waist circumference (WC) showed a strong association with asthma (OR = 1.4777, 95% CI = 1.3412-1.6281, p = 2.8707 × 10-15). Conversely, high-density lipoprotein cholesterol (HDL-C) was found to be inversely related to the risk of asthma (OR = 0.9186, 95% CI = 0.8669-0.9734, p = 0.0041). CONCLUSION The findings of this study support that MetS and its specific components, particularly abdominal obesity, are linked to a higher risk of asthma, while HDL-C might offer protective effects against asthma. These findings provide a foundation both for further research and possible therapeutic interventions.
Collapse
Affiliation(s)
- Xiaosong Ru
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Luyi Huang
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ziying Su
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chenxiao Ye
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yong Guo
- Department of Medical Oncology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
77
|
Guo X, Feng Y, Ji X, Jia N, Maimaiti A, Lai J, Wang Z, Yang S, Hu S. Shared genetic architecture and bidirectional clinical risks within the psycho-metabolic nexus. EBioMedicine 2025; 111:105530. [PMID: 39731856 PMCID: PMC11743124 DOI: 10.1016/j.ebiom.2024.105530] [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] [Received: 09/05/2024] [Revised: 12/12/2024] [Accepted: 12/12/2024] [Indexed: 12/30/2024] Open
Abstract
BACKGROUND Increasing evidence suggests a complex interplay between psychiatric disorders and metabolic dysregulations. However, most research has been limited to specific disorder pairs, leaving a significant gap in our understanding of the broader psycho-metabolic nexus. METHODS This study leveraged large-scale cohort data and genome-wide association study (GWAS) summary statistics, covering 8 common psychiatric disorders and 43 metabolic traits. We introduced a comprehensive analytical strategy to identify shared genetic bases sequentially, from key genetic correlation regions to local pleiotropy and pleiotropic genes. Finally, we developed polygenic risk score (PRS) models to translate these findings into clinical applications. FINDINGS We identified significant bidirectional clinical risks between psychiatric disorders and metabolic dysregulations among 310,848 participants from the UK Biobank. Genetic correlation analysis confirmed 104 robust trait pairs, revealing 1088 key genomic regions, including critical hotspots such as chr3: 47588462-50387742. Cross-trait meta-analysis uncovered 388 pleiotropic single nucleotide variants (SNVs) and 126 shared causal variants. Among variants, 45 novel SNVs were associated with psychiatric disorders and 75 novel SNVs were associated with metabolic traits, shedding light on new targets to unravel the mechanism of comorbidity. Notably, RBM6, a gene involved in alternative splicing and cellular stress response regulation, emerged as a key pleiotropic gene. When psychiatric and metabolic genetic information were integrated, PRS models demonstrated enhanced predictive power. INTERPRETATION The study highlights the intertwined genetic and clinical relationships between psychiatric disorders and metabolic dysregulations, emphasising the need for integrated approaches in diagnosis and treatment. FUNDING The National Key Research and Development Program of China (2023YFC2506200, SHH). The National Natural Science Foundation of China (82273741, SY).
Collapse
Affiliation(s)
- Xiaonan Guo
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yu Feng
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Department of Psychiatry, Melbourne Neuropsychiatry Centre, The University of Melbourne, Carlton South, VIC, Australia
| | - Xiaolong Ji
- Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ningning Jia
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin, China
| | - Aierpati Maimaiti
- Department of Neurosurgery, Xinjiang Medical University Affiliated First Hospital, Urumqi, Xinjiang, China
| | - Jianbo Lai
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zheng Wang
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| | - Sheng Yang
- Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Shaohua Hu
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Nanhu Brain-Computer Interface Institute, Hangzhou, Zhejiang, China; Zhejiang Key Laboratory of Precision Psychiatry, Hangzhou, 310003, China; Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 311121, China; Brain Research Institute of Zhejiang University, Hangzhou, 310058, China; MOE Frontier Science Center for Brain Science and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou, 310058, China; Department of Psychology and Behavioral Sciences, Graduate School, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
78
|
Lankinen MA, Nuotio P, Kauppinen S, Koivu N, Tolonen U, Malkki-Keinänen K, Oravilahti A, Kuulasmaa T, Uusitupa M, Schwab U, Laakso M. Effects of Genetic Risk on Incident Type 2 Diabetes and Glycemia: The T2D-GENE Lifestyle Intervention Trial. J Clin Endocrinol Metab 2024; 110:130-138. [PMID: 38888187 PMCID: PMC11651687 DOI: 10.1210/clinem/dgae422] [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: 04/03/2024] [Revised: 06/11/2024] [Accepted: 06/14/2024] [Indexed: 06/20/2024]
Abstract
CONTEXT Lifestyle intervention prevents or delays type 2 diabetes (T2D) in subjects at a high risk of T2D. However, it is not known whether genetic variants modify the effect on incident T2D during lifestyle intervention. OBJECTIVE To investigate whether a low or high genetic risk has effects on incident T2D in a group-based lifestyle intervention study. METHODS The T2D-GENE trial involved 973 men from the Metabolic Syndrome in Men (METSIM) cohort, aged 50-75 years, body mass index ≥25 kg/m2, fasting plasma glucose 5.6-6.9 mmol/L, hemoglobin A1c < 48 mmol/mol, and either a low or high genetic risk score for T2D. There were 2 intervention groups, a low (n = 315) and high genetic risk for T2D (n = 313). They were provided with a 3-year group-based intervention with access to a web portal focused on healthy diet and physical activity. There were also corresponding population-based control groups at low (n = 196) and high (n = 149) genetic risk for T2D who had two laboratory visits (0 and 3 years) and general health advice as a part of their METSIM cohort protocol. The primary outcome was incident T2D, and a secondary outcome was glycemia. RESULTS The intervention significantly lowered the risk of T2D among the participants with a high genetic risk for T2D [hazards ratio (HR) 0.30, 95% confidence interval (CI) 0.16-0.56, P < .001) whereas in the low genetic risk group the effect was not significant (HR 0.69, 95% CI 0.36-1.32, P = .262). The intervention effect was not significantly different between the high and low genetic risk groups (P = .135). The intervention significantly ameliorated the worsening of glycemia and decreased weight both in the low and high genetic risk groups. CONCLUSION Our results showed that individuals with a high genetic risk for T2D benefitted from a low-cost group-based intervention focusing on healthy diet and physical activity. Therefore, all individuals at risk of T2D should be encouraged to make lifestyle changes regardless of genetic risk.
Collapse
Affiliation(s)
- Maria Anneli Lankinen
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70211 Kuopio, Finland
| | - Petrus Nuotio
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70211 Kuopio, Finland
| | - Susanna Kauppinen
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70211 Kuopio, Finland
| | - Noora Koivu
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70211 Kuopio, Finland
| | - Ulla Tolonen
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70211 Kuopio, Finland
| | - Katriina Malkki-Keinänen
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70211 Kuopio, Finland
| | - Anniina Oravilahti
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, 70211 Kuopio, Finland
| | - Teemu Kuulasmaa
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, 70211 Kuopio, Finland
| | - Matti Uusitupa
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70211 Kuopio, Finland
| | - Ursula Schwab
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70211 Kuopio, Finland
- Department of Medicine, Endocrinology and Clinical Nutrition, Kuopio University Hospital, Wellbeing Services County of North Savo, 70210 Kuopio, Finland
| | - Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland and Kuopio University Hospital, 70211 Kuopio, Finland
| |
Collapse
|
79
|
Varshney A, Manickam N, Orchard P, Tovar A, Ventresca C, Zhang Z, Feng F, Mears J, Erdos MR, Narisu N, Nishino K, Rai V, Stringham HM, Jackson AU, Tamsen T, Gao C, Yang M, Koues OI, Welch JD, Burant CF, Williams LK, Jenkinson C, DeFronzo RA, Norton L, Saramies J, Lakka TA, Laakso M, Tuomilehto J, Mohlke KL, Kitzman JO, Koistinen HA, Liu J, Boehnke M, Collins FS, Scott LJ, Parker SCJ. Population-scale skeletal muscle single-nucleus multi-omic profiling reveals extensive context specific genetic regulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.15.571696. [PMID: 38168419 PMCID: PMC10760134 DOI: 10.1101/2023.12.15.571696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Skeletal muscle, the largest human organ by weight, is relevant in several polygenic metabolic traits and diseases including type 2 diabetes (T2D). Identifying genetic mechanisms underlying these traits requires pinpointing cell types, regulatory elements, target genes, and causal variants. Here, we use genetic multiplexing to generate population-scale single nucleus (sn) chromatin accessibility (snATAC-seq) and transcriptome (snRNA-seq) maps across 287 frozen human skeletal muscle biopsies representing nearly half a million nuclei. We identify 13 cell types and integrate genetic variation to discover >7,000 expression quantitative trait loci (eQTL) and >100,000 chromatin accessibility QTLs (caQTL) across cell types. Learning patterns of e/caQTL sharing across cell types increased precision of effect estimates. We identify high-resolution cell-states and context-specific e/caQTL with significant genotype by context interaction. We identify nearly 2,000 eGenes colocalized with caQTL and construct causal directional maps for chromatin accessibility and gene expression. Almost 3,500 genome-wide association study (GWAS) signals across 38 relevant traits colocalize with sn-e/caQTL, most in a cell-specific manner. These signals typically colocalize with caQTL and not eQTL, highlighting the importance of population-scale chromatin profiling for GWAS functional studies. Finally, our GWAS-caQTL colocalization data reveal distinct cell-specific regulatory paradigms. Our results illuminate the genetic regulatory architecture of human skeletal muscle at high resolution epigenomic, transcriptomic, and cell-state scales and serve as a template for population-scale multi-omic mapping in complex tissues and traits.
Collapse
Affiliation(s)
- Arushi Varshney
- Dept. of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Nandini Manickam
- Dept. of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Peter Orchard
- Dept. of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Adelaide Tovar
- Dept. of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Christa Ventresca
- Dept. of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
- Dept. of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Zhenhao Zhang
- Dept. of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Fan Feng
- Dept. of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Joseph Mears
- Dept. of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Michael R Erdos
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Narisu Narisu
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Kirsten Nishino
- Dept. of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Vivek Rai
- Dept. of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Heather M Stringham
- Department of Biostatistics, Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Anne U Jackson
- Department of Biostatistics, Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Tricia Tamsen
- Biomedical Research Core Facilities Advanced Genomics Core, University of Michigan, Ann Arbor, MI, USA
| | - Chao Gao
- Dept. of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Mao Yang
- Department of Internal Medicine, Center for Individualized and Genomic Medicine Research, Henry Ford Hospital, Detroit, MI, USA
| | - Olivia I Koues
- Biomedical Research Core Facilities Advanced Genomics Core, University of Michigan, Ann Arbor, MI, USA
| | - Joshua D Welch
- Dept. of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Charles F Burant
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - L Keoki Williams
- Department of Internal Medicine, Center for Individualized and Genomic Medicine Research, Henry Ford Hospital, Detroit, MI, USA
| | - Chris Jenkinson
- South Texas Diabetes and Obesity Research Institute, School of Medicine, University of Texas, Rio Grande Valley, TX, USA
| | - Ralph A DeFronzo
- Department of Medicine/Diabetes Division, University of Texas Health, San Antonio, TX, USA
| | - Luke Norton
- Department of Medicine/Diabetes Division, University of Texas Health, San Antonio, TX, USA
| | - Jouko Saramies
- Savitaipale Health Center, South Karelia Central Hospital, Lappeenranta, Finland
| | - Timo A Lakka
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Markku Laakso
- Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Jaakko Tuomilehto
- Dept. of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
- Dept. of Public Health, University of Helsinki, Helsinki, Finland
- Diabetes Research Group, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Karen L Mohlke
- Dept. of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Jacob O Kitzman
- Dept. of Human Genetics, University of Michigan, Ann Arbor, MI, USA
- Dept. of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Heikki A Koistinen
- Dept. 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
| | - Jie Liu
- Dept. of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Michael Boehnke
- Department of Biostatistics, Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Francis S Collins
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Laura J Scott
- Department of Biostatistics, Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Stephen C J Parker
- Dept. of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
- Dept. of Human Genetics, University of Michigan, Ann Arbor, MI, USA
- Department of Biostatistics, Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
80
|
Zhao C, Hatzikotoulas K, Balasubramanian R, Bertone-Johnson E, Cai N, Huang L, Huerta-Chagoya A, Janiczek M, Ma C, Mandla R, Paluch A, Rayner NW, Southam L, Sturgeon SR, Suzuki K, Taylor HJ, VanKim N, Yin X, Lee CH, Collins F, Spracklen CN. Associations of Combined Genetic and Lifestyle Risks with Incident Type 2 Diabetes in the UK Biobank. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.12.16.24319115. [PMID: 39763538 PMCID: PMC11702748 DOI: 10.1101/2024.12.16.24319115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
Background Type 2 diabetes (T2D) results from a complex interplay between genetic predisposition and lifestyle factors. Both genetic susceptibility and unhealthy lifestyle are known to be associated with elevated T2D risk. However, their combined effects on T2D risk are not well studied. We aimed to determine whether unhealthy modifiable health behaviors were associated with similar increases in the risk of incident T2D among individuals with different levels of genetic risk. Methods We performed a genetic risk score (GRS) by lifestyle interaction analysis within 332,251 non-diabetic individuals at baseline from the UK Biobank. Multi-ancestry GRS were calculated by summing the effects of 783 T2D-associated variants and ranked into tertiles. We used baseline self-reported data on smoking, BMI, physical activity level, and diet quality to categorize participants as having a healthy, intermediate, or unhealthy lifestyle. Cox proportional hazards regression models were used to generate adjusted hazards ratios (HR) of T2D risk and associated 95% confidence intervals (CI). Results During follow-up (median 13.6 years), 13,128 (4.0%) participants developed T2D. GRS (P < 0.001) and lifestyle classification (P < 0.001) were independently associated with increased risk for T2D. Compared with healthy lifestyle, unhealthy lifestyle was associated with increased T2D risk in all genetic risk strata, with adjusted HR ranging from 7.11 (low genetic risk) to 16.33 (high genetic risk). Conclusions High genetic risk and unhealthy lifestyle were the most significant contributors to the development of T2D. Individuals at all levels of genetic risk can greatly mitigate their risk for T2D through lifestyle modifications.
Collapse
Affiliation(s)
- Chi Zhao
- Department of Biostatistics and Epidemiology, University of Massachusetts Amherst, Amherst, MA, USA
| | - Konstantinos Hatzikotoulas
- Institute of Translational Genomics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Raji Balasubramanian
- Department of Biostatistics and Epidemiology, University of Massachusetts Amherst, Amherst, MA, USA
| | | | - Na Cai
- Helmholtz Pioneer Campus, Helmholtz Munich, Neuherberg, Germany
- Computational Health Centre, Helmholtz Munich, Neuherberg, Germany
- School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Lianyun Huang
- Helmholtz Pioneer Campus, Helmholtz Munich, Neuherberg, Germany
- Computational Health Centre, Helmholtz Munich, Neuherberg, Germany
- School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Alicia Huerta-Chagoya
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Margaret Janiczek
- Department of Biostatistics and Epidemiology, University of Massachusetts Amherst, Amherst, MA, USA
| | - Chaoran Ma
- Department of Nutrition, University of Massachusetts Amherst, Amherst, MA, USA
| | - Ravi Mandla
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Graduate Program in Genomics and Computational Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Amanda Paluch
- Department of Kinesiology, Institute for Applied Life Sciences, University of Massachusetts Amherst, Amherst, MA, USA
| | - Nigel W Rayner
- Institute of Translational Genomics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Lorraine Southam
- Institute of Translational Genomics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Susan R. Sturgeon
- Department of Biostatistics and Epidemiology, University of Massachusetts Amherst, Amherst, MA, USA
| | - Ken Suzuki
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
| | - Henry J Taylor
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
| | - Nicole VanKim
- Department of Biostatistics and Epidemiology, University of Massachusetts Amherst, Amherst, MA, USA
| | - Xianyong Yin
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, China
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Chi Hyun Lee
- Department of Applied Statistics, Yonsei University, Seoul, South Korea
| | - Francis Collins
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Cassandra N. Spracklen
- Department of Biostatistics and Epidemiology, University of Massachusetts Amherst, Amherst, MA, USA
| |
Collapse
|
81
|
Kushnir MM, Salihovic S, Bergquist J, Lind PM, Lind L. Environmental contaminants, sex hormones and SHBG in an elderly population. ENVIRONMENTAL RESEARCH 2024; 263:120054. [PMID: 39341538 DOI: 10.1016/j.envres.2024.120054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 09/12/2024] [Accepted: 09/22/2024] [Indexed: 10/01/2024]
Abstract
INTRODUCTION Effects of environmental contaminants (ECs) on endocrine systems have been reported, but few studies assessed associations between ECs and sex hormones (SH) in elderly. Aim of this study was to investigate whether blood concentrations of four classes of ECs were associated with SH concentrations in elderly. METHODS Samples from participants of the cross-sectional population-based Prospective Investigation of the Vasculature in Uppsala Seniors study (PIVUS, 70-year-old men and women, n = 1016) were analyzed using validated mass spectrometry-based methods for SH (testosterone (T), dihydrotestosterone (DHT), estrone and estradiol (E2)); 23 persistent organic pollutants (POPs); 8 perfluoroalkyl substances (PFAS); 4 phthalates and 11 metals. SH binding globulin (SHBG) was analyzed using immunoassay. The measured concentrations were normalized, and the values converted to a z-scale. Linear regression analyses were conducted to assess association between concentration of the SH, SHBG and E2/T (aromatase enzyme index, AEI) with the ECs. Multiple linear regression analyses were performed to model the relationships. RESULTS The strongest associations were observed with the polychlorinated biphenyls (PCBs). In men, the strongest associations with concentrations of SH and SHBG were seen for PCBs containing >5 chlorine, monoethyl phthalate (MEP), Ni and Cd; and in women, with PCBs, MEP, several of the PFAS, Cd, Co, and Ni. Difference in the effect of ECs on AEI between men and women were observed. Area under the ROC curve for the models predicting abnormal values of SH and SHBG >0.75 due to the effects of ECs was observed for T, DHT, and E2 in men, and for E2 and SHBG in women. CONCLUSIONS Results of this study suggest that in elderly subjects, concentrations of many ECs associated with concentrations of SH and SHBG, and AEI. Further studies are needed to confirm the findings and to assess effect of the pollutants on endocrine system function in elderly.
Collapse
Affiliation(s)
- Mark M Kushnir
- ARUP Institute for Clinical and Experimental Pathology, Salt Lake City, UT, USA; Department of Pathology, University of Utah, Salt Lake City, UT, USA.
| | - Samira Salihovic
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Sweden
| | - Jonas Bergquist
- Department of Pathology, University of Utah, Salt Lake City, UT, USA; Department of Chemistry, Analytical Chemistry, Uppsala University, Uppsala, Sweden
| | - P Monica Lind
- Department of Medical Sciences, Occupational and Environmental Medicine, Uppsala University, Uppsala, Sweden
| | - Lars Lind
- Department of Medical Sciences, Cardiovascular Epidemiology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
82
|
Needham EJ, Hingst JR, Onslev JD, Diaz-Vegas A, Leandersson MR, Huckstep H, Kristensen JM, Kido K, Richter EA, Højlund K, Parker BL, Cooke K, Yang G, Pehmøller C, Humphrey SJ, James DE, Wojtaszewski JFP. Personalized phosphoproteomics of skeletal muscle insulin resistance and exercise links MINDY1 to insulin action. Cell Metab 2024; 36:2542-2559.e6. [PMID: 39577414 DOI: 10.1016/j.cmet.2024.10.020] [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: 03/12/2024] [Revised: 07/05/2024] [Accepted: 10/22/2024] [Indexed: 11/24/2024]
Abstract
Type 2 diabetes is preceded by a defective insulin response, yet our knowledge of the precise mechanisms is incomplete. Here, we investigate how insulin resistance alters skeletal muscle signaling and how exercise partially counteracts this effect. We measured parallel phenotypes and phosphoproteomes of insulin-resistant (IR) and insulin-sensitive (IS) men as they responded to exercise and insulin (n = 19, 114 biopsies), quantifying over 12,000 phosphopeptides in each biopsy. Insulin resistance involves selective and time-dependent alterations to signaling, including reduced insulin-stimulated mTORC1 and non-canonical signaling responses. Prior exercise promotes insulin sensitivity even in IR individuals by "priming" a portion of insulin signaling prior to insulin infusion. This includes MINDY1 S441, which we show is an AKT substrate. We found that MINDY1 knockdown enhances insulin-stimulated glucose uptake in rat myotubes. This work delineates the signaling alterations in IR skeletal muscle and identifies MINDY1 as a regulator of insulin action.
Collapse
Affiliation(s)
- Elise J Needham
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Sydney, NSW, Australia; British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK; Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
| | - Janne R Hingst
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Johan D Onslev
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Alexis Diaz-Vegas
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Sydney, NSW, Australia
| | - Magnus R Leandersson
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Hannah Huckstep
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC 3052, Australia
| | - Jonas M Kristensen
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Kohei Kido
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark; Health and Medical Research Institute, Department of Life Science and Biotechnology, National Institute of Advanced Industrial Science and Technology (AIST), Takamatsu, Kagawa, Japan
| | - Erik A Richter
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Kurt Højlund
- Steno Diabetes Center Odense, Odense University Hospital, Odense, Denmark; Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Benjamin L Parker
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Sydney, NSW, Australia; Department of Anatomy and Physiology, School of Biomedical Sciences, Faculty of Medicine Dentistry and Health Sciences, The University of Melbourne, Melbourne, VIC, Australia
| | - Kristen Cooke
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Sydney, NSW, Australia
| | - Guang Yang
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Sydney, NSW, Australia; School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Christian Pehmøller
- Internal Medicine Research Unit, Pfizer Global Research and Development, Cambridge, MA, USA
| | - Sean J Humphrey
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Sydney, NSW, Australia; Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC 3052, Australia.
| | - David E James
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Sydney, NSW, Australia; Sydney Medical School, University of Sydney, Sydney, NSW, Australia.
| | - Jørgen F P Wojtaszewski
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
83
|
Chen G, Wang Y, Wang X. Insulin-related traits and prostate cancer: A Mendelian randomization study. Comput Struct Biotechnol J 2024; 23:2337-2344. [PMID: 38867724 PMCID: PMC11167198 DOI: 10.1016/j.csbj.2024.05.034] [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: 11/21/2023] [Revised: 05/17/2024] [Accepted: 05/17/2024] [Indexed: 06/14/2024] Open
Abstract
Investigating the causal relationship between insulin secretion and prostate cancer (PCa) development is challenging due to the multifactorial nature of PCa, which complicates the isolation of the specific impact of insulin-related factors. We conducted a Mendelian randomization (MR) study to investigate the associations between insulin secretion-related traits and PCa. We used 36, 60, 56, 23, 48, and 49 single nucleotide polymorphisms (SNPs) as instrumental variables for fasting insulin, insulin sensitivity, proinsulin, and proinsulin in nondiabetic individuals, individuals with diabetes, and individuals receiving exogenous insulin, respectively. These SNPs were selected from various genome-wide association studies. To clarify the causal relationship between insulin-related traits and PCa, we utilized a multivariable MR analysis to adjust for obesity and body fat percentage. Additionally, two-step Mendelian randomization was conducted to assess the role of insulin-like growth factor 1 (IGF-1) in the relationship between proinsulin and PCa. Two-sample MR analysis revealed strong associations between genetically predicted fasting insulin, insulin sensitivity, proinsulin, and proinsulin in nondiabetic individuals and the development of PCa. After adjustment for obesity and body fat percentage using multivariable MR analysis, proinsulin remained significantly associated with PCa, whereas other factors were not. Furthermore, two-step MR analysis demonstrated that proinsulin acts as a negative factor in prostate carcinogenesis, largely independent of IGF-1. This study provides evidence suggesting that proinsulin may act as a negative factor contributing to the development of PCa. Novel therapies targeting proinsulin may have potential benefits for PCa patients, potentially reducing the need for unnecessary surgical treatments.
Collapse
Affiliation(s)
- Guihua Chen
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Yi Wang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
- Department of Urology, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China
| | - Xiang Wang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| |
Collapse
|
84
|
Zheng H, Wang W, Chen C, Feng Y. Association between walking pace and heart failure: A Mendelian randomization analysis. Nutr Metab Cardiovasc Dis 2024; 34:2713-2719. [PMID: 39174430 DOI: 10.1016/j.numecd.2024.07.012] [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: 01/24/2024] [Revised: 06/10/2024] [Accepted: 07/15/2024] [Indexed: 08/24/2024]
Abstract
BACKGROUND AND AIM The relationship between walking pace and heart failure (HF) has been recognized, yet the directionality and underlying mediating risk factors remain unclear. METHODS AND RESULTS This study utilized bidirectional two-sample Mendelian randomization (MR) with genome-wide association studies (GWAS) summary statistics to assess the causal relationships between walking pace and HF. Additionally, we employed a two-step Multivariable Mendelian Randomization (MVMR) to explore potential mediating factors. We further validated our findings by conducting two-sample MR with another available GWAS summary data on heart failure. Results indicated that genetically predicted increases in walking pace were associated with a reduced risk of HF (odds ratio (OR), 0.589, 95% confidence interval (CI): 0.417-0.832). Among the considered mediators, the waist-to-hip ratio (WHR) accounts for the largest proportion of the effect (45.7%, 95% CI: 13.2%, 78.2%). This is followed by type 2 diabetes at 24.4% (95% CI: 6.7%, 42.0%) and triglycerides at 18.6% (95% CI: 4.5%, 32.7%). Furthermore, our findings reveal that genetically predicted HF risk (OR, 0.975, 95% CI: 0.960-0.991) is associated with a slower walking pace. Validated findings were consistent with the main results. CONCLUSIONS In conclusion, MR analysis demonstrates that a slow walking pace is a reliable indicator of an elevated risk of HF, and the causal relationship is bidirectional. Interventions focusing on waist-to-hip ratio, type 2 diabetes, and triglycerides may provide valuable strategies for HF prevention in individuals with a slow walking pace.
Collapse
Affiliation(s)
- He Zheng
- School of Medicine, South China University of Technology, Guangzhou, China; Department of Cardiology, Hypertension Research Laboratory, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Wenbin Wang
- Department of Cardiology, Hypertension Research Laboratory, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Chaolei Chen
- Department of Cardiology, Hypertension Research Laboratory, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Yingqing Feng
- School of Medicine, South China University of Technology, Guangzhou, China; Department of Cardiology, Hypertension Research Laboratory, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
| |
Collapse
|
85
|
Lowe WL, Kuang A, Hayes MG, Hivert MF, Scholtens DM. Genetics of glucose homeostasis in pregnancy and postpartum. Diabetologia 2024; 67:2726-2739. [PMID: 39180581 DOI: 10.1007/s00125-024-06256-8] [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: 03/07/2024] [Accepted: 07/02/2024] [Indexed: 08/26/2024]
Abstract
AIMS/HYPOTHESIS Pregnancy is accompanied by maternal metabolic adaptations to ensure fetal growth and development, including insulin resistance, which occurs primarily during the second and third trimesters of pregnancy, and a decrease in fasting blood sugar levels over the course of pregnancy. Glucose-related traits are regulated by genetic and environmental factors and modulated by physiological variations throughout the life course. We addressed the hypothesis that there are both overlaps and differences between genetic variants associated with glycaemia-related traits during and outside of pregnancy. METHODS Genome-wide SNP data were used to identify genetic variations associated with glycaemia-related traits measured during an OGTT performed at ~28 weeks' gestation in 8067 participants in the Hyperglycaemia and Adverse Pregnancy Outcome (HAPO) Study. Associations outside of pregnancy were determined in 3977 individuals who also participated in the HAPO Follow-Up Study at 11-14 years postpartum. A Bayesian classification algorithm was used to determine whether SNPs associated with fasting and 2 h glucose and fasting C-peptide during pregnancy had a pregnancy-predominant effect vs a similar effect during pregnancy and postpartum. RESULTS SNPs in six loci (GCKR, G6PC2, GCK, PPP1R3B, PCSK1 and MTNR1B) were significantly associated with fasting glucose during pregnancy, while SNPs in CDKAL1 and MTNR1B were associated with 1 h glucose and SNPs in MTNR1B and HKDC1 were associated with 2 h glucose. Variants in CDKAL1 and MTNR1B were associated with insulin secretion during pregnancy. Variants in multiple loci were associated with fasting C-peptide during pregnancy, including GCKR, IQSEC1, PPP1R3B, IGF1 and BACE2. GCKR and BACE2 were associated with 1 h C-peptide and GCKR, IQSEC1 and BACE2 with insulin sensitivity during pregnancy. The associations of MTNR1B with 2 h glucose, BACE2 with fasting and 1 h C-peptide and insulin sensitivity, and IQSEC1 with fasting C-peptide and insulin sensitivity that we identified during pregnancy have not been previously reported in non-pregnancy cohorts. The Bayesian classification algorithm demonstrated that the magnitude of effect of the lead SNP was greater during pregnancy compared with 11-14 years postpartum in PCSK1 and PPP1R3B with fasting glucose, in three loci, including MTNR1B, with 2 h glucose, and in six loci, including IGF1, with fasting C-peptide. CONCLUSIONS/INTERPRETATION Our findings support the hypothesis that there are both overlaps and differences between the genetic architecture of glycaemia-related traits during and outside of pregnancy. Genetic variants at several loci, including PCSK1, PPP1R3B, MTNR1B and IGF1, appear to influence glycaemic regulation in a unique fashion during pregnancy. Future studies in larger cohorts will be needed to replicate the present findings, fully characterise the genetics of maternal glycaemia during pregnancy and determine similarities to and differences from the non-gravid state.
Collapse
Affiliation(s)
- William L Lowe
- Department of Medicine, Northwestern University Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| | - Alan Kuang
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - M Geoffrey Hayes
- Department of Medicine, Northwestern University Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Marie-France Hivert
- Diabetes Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine, Harvard Pilgrim Health Care Institute, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada
| | - Denise M Scholtens
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
86
|
Yu L, Liu W, Zhang Y, Tan Q, Song J, Fan L, You X, Zhou M, Wang B, Chen W. Styrene and ethylbenzene exposure and type 2 diabetes mellitus: A longitudinal gene-environment interaction study. ECO-ENVIRONMENT & HEALTH 2024; 3:452-457. [PMID: 39559189 PMCID: PMC11570399 DOI: 10.1016/j.eehl.2024.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 06/12/2024] [Accepted: 07/21/2024] [Indexed: 11/20/2024]
Abstract
Styrene and ethylbenzene (S/EB) are identified as hazardous air contaminants that raise significant concerns. The association between S/EB exposure and the incidence of type 2 diabetes mellitus (T2DM), and the interaction between genes and environment, remains poorly understood. Our study consisted of 2219 Chinese adults who were part of the Wuhan-Zhuhai cohort. A follow-up assessment was conducted after six years. Exposure to S/EB was quantified by determining the concentrations of urinary biomarkers of exposure to S/EB (UBE-S/EB; urinary phenylglyoxylic acid level plus urinary mandelic acid level). Logistic regression models were constructed to investigate the relations of UBE-S/EB and genetic risk score (GRS) with T2DM prevalence and incidence. The interaction effects of UBE-S/EB and GRS on T2DM were investigated on multiplicative and additive scales. UBE-S/EB was dose-dependently and positively related to T2DM prevalence and incidence. Participants with high levels of UBE-S/EB [relative risk (RR) = 1.930, 95% confidence interval (CI): 1.157-3.309] or GRS (1.943, 1.110-3.462) demonstrated the highest risk of incident T2DM, in comparison to those with low levels of UBE-S/EB or GRS. Significant additive interaction between UBE-S/EB and GRS on T2DM incidence was discovered with relative excess risk due to interaction (95% CI) of 0.178 (0.065-0.292). The RR (95% CI) of T2DM incidence was 2.602 (1.238-6.140) for individuals with high UBE-S/EB and high GRS, compared to those with low UBE-S/EB and low GRS. This study presented the initial evidence that S/EB exposure was significantly related to increased risk of T2DM incidence, and the relationship was interactively aggravated by genetic predisposition.
Collapse
Affiliation(s)
- Linling Yu
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Department of Public Health, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Wei Liu
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yongfang Zhang
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qiyou Tan
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jiahao Song
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Lieyang Fan
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiaojie You
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Min Zhou
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Bin Wang
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Weihong Chen
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
87
|
Cao Z, Li Q, Wu J, Li Y. Genetic proxies for therapy of insulin drug targets and risk of osteoarthritis: a drug-target Mendelian randomization analysis. Inflammopharmacology 2024; 32:3717-3728. [PMID: 39127978 PMCID: PMC11550247 DOI: 10.1007/s10787-024-01542-8] [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: 05/12/2024] [Accepted: 07/24/2024] [Indexed: 08/12/2024]
Abstract
BACKGROUND The potential effects of insulin therapy on osteoarthritis (OA) risk are poorly understood. This study aimed to explore the causal relationship between insulin therapy and OA. METHODS Mendelian randomization (MR) analysis was performed to examine the association between genetically proxied inhibition of insulin targets and the risk of overall, hip (HOA) and knee OA (KOA). We then performed univariable MR using summary statistics regarding insulin target genes derived from the DrugBank database. Data related to blood glucose reduction levels were used as a proxy for insulin levels. Two phenotypes, type 2 diabetes, and glycosylated hemoglobin levels, were selected as positive controls to confirm the direction and validity of the proxies. The OA datasets were derived from the UK Biobank cohort. Multivariable MR was adjusted for body mass index, sedentary behavior, cigarette smoking, frequency of alcohol intake, age, and genetic sex. RESULTS Genetically proxied insulin therapy was associated with an increased risk of overall OA [odds ratio (OR):1.2595; 95% confidence interval (CI):1.0810-1.4675] and HOA (OR:1.4218; 95%CI:1.1240-1.7985), which remained consistent across multiple MR methods. After adjusting for confounders, we found evidence supporting a significant causal link with a higher risk of overall OA and HOA. A further two-step MR analysis revealed no significant mediation effects from the six mediators in the associations. CONCLUSION There was a causal association between genetically proxied insulin therapy and a higher risk of OA, especially HOA.
Collapse
Affiliation(s)
- Ziqin Cao
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha, China
| | - Qiangxiang Li
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha, China
| | - Jianhuang Wu
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha, China.
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
| | - Yajia Li
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha, China.
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
| |
Collapse
|
88
|
Wei Z, Hu Y, Zuo F, Wen X, Wu D, Sun X, Liu C. The association between metabolic syndrome and lung cancer risk: a Mendelian randomization study. Sci Rep 2024; 14:28494. [PMID: 39558018 PMCID: PMC11574301 DOI: 10.1038/s41598-024-79260-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: 04/12/2024] [Accepted: 11/07/2024] [Indexed: 11/20/2024] Open
Abstract
Metabolic syndrome (MetS) is closely linked to cancer development, with emerging evidence suggesting its association with pulmonary carcinoma. However, causal relationships remain unclear due to observational study limitations. Employing Mendelian randomization, we investigated the causal link between MetS and lung cancer (LC) susceptibility. The data utilized in this study were obtained from the publicly available genetic variation summary database. The causal relationship was assessed using the inverse variance weighting method (IVW), weighted median method, and MR-Egger regression. A sensitivity analysis was carried out to confirm the robustness of the findings. Furthermore, risk factor analyses were conducted to explore potential mediators. Utilizing various analyses, MetS demonstrated a significant positive association with LC (OR, 1.22; 95% CI, 1.09-1.37, p = 7.57 × 10- 4), lung squamous cell carcinoma (LUSC) (OR, 1.47; 95%, 1.23-1.75, p = 2.22 × 10- 5), and small cell lung cancer (SCLC) (OR, 1.76; 95% CI, 1.37-2.26, p = 8.20 × 10- 6) but not lung adenocarcinoma (LUAD) (OR, 1.08; 95% CI, 0.94-1.24, p = 0.28). Risk factor analyses indicated that smoking, alcohol, body mass index, education, and type 2 diabetes might mediate the association. This study genetically validates and reinforces the evidence of MetS increasing the incidence of LC, including both LUSC) and SCLC, especially among individuals with abdominal obesity. It provides valuable insights for the development of lung cancer prevention strategies and directions for future research.
Collapse
Affiliation(s)
- Zhicheng Wei
- Department of Pharmacy, Dazhou Central Hospital, Dazhou, 635000, People's Republic of China
| | - Yunyun Hu
- Department of Pharmacy, Dazhou Central Hospital, Dazhou, 635000, People's Republic of China
| | - Fang Zuo
- Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, People's Republic of China
| | - Xiushu Wen
- Department of Pharmacy, Dazhou Central Hospital, Dazhou, 635000, People's Republic of China
| | - Desheng Wu
- Department of Pharmacy, Dazhou Central Hospital, Dazhou, 635000, People's Republic of China
| | - Xiaodong Sun
- Department of Pharmacy, Dazhou Central Hospital, Dazhou, 635000, People's Republic of China
| | - Conghai Liu
- Department of Pharmacy, Dazhou Central Hospital, Dazhou, 635000, People's Republic of China.
| |
Collapse
|
89
|
Wang W, Zhang J, Zhang M, Zhang C, Liu H, Li W, Fan Y. Impact of diabetes mellitus on the risk of Alzheimer's disease: a mendelian randomization study. BMC Neurol 2024; 24:448. [PMID: 39558225 PMCID: PMC11571773 DOI: 10.1186/s12883-024-03955-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] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 11/07/2024] [Indexed: 11/20/2024] Open
Abstract
BACKGROUND The impact of diabetes on the risk of Alzheimer's disease remains uncertain. This study aimed to explore this issue from multiple perspectives by using the Mendelian randomization (MR) approach. METHODS Instrumental variables for predicting six diabetic traits (including insulin and blood glucose), eight metabolic risk factors for diabetes (including total cholesterol and blood pressure), and seven diabetic genes were extracted from their summary data. These data were derived from multiple European cohorts and included 31,684 to 810,865 subjects respectively. The two-sample MR, multivariate MR, and summary-data-based Mendelian randomization (SMR) methods were employed to determine the associations of these traits or genes with the risk of Alzheimer's disease. RESULTS The two-sample MR showed that elevated fasting insulin and total cholesterol levels were associated with an increased risk of dementia in Alzheimer's disease (P = 0.022, P = 0.041). Elevated systolic and diastolic blood pressure levels were associated with a decreased risk of dementia in Alzheimer's disease (P = 0.036, P = 0.025). The multivariate MR reported that adjusting for telomere length (a well-established biomarker of aging) did not change these findings (P < 0.05). Additionally, the two-sample MR showed that type 1 and type 2 diabetes did not affect the risk of Alzheimer's disease. The SMR also indicated that the diabetic genes did not affect the risk of this disease. CONCLUSION Multiple MR approaches concluded that fasting insulin, total cholesterol, and blood pressure, rather than diabetes, were potential metabolic variables that had an impact on the risk of Alzheimer's disease. However, aging might not be involved in these correlations.
Collapse
Affiliation(s)
- Weichao Wang
- Department of Endocrinology, Shijiazhuang People's Hospital, 365, Jianhua South Street, Yuhua District, Shijiazhuang, Hebei Province, 050011, China.
| | - Jie Zhang
- Department of Ophthalmology, Shijiazhuang Third Hospital, Shijiazhuang, Hebei Province, 050011, China
| | - Man Zhang
- Graduate School, Hebei Medical University, Shijiazhuang, Hebei Province, 050011, China
| | - Chengyuan Zhang
- Examination Division, Medical Skills Examination and Appraisal Center, Shijiazhuang, Hebei Province, 050011, China
| | - Huanli Liu
- Teaching Division, Shijiazhuang People's Hospital, Shijiazhuang, Hebei Province, 050011, China
| | - Wanlin Li
- Teaching Division, Shijiazhuang People's Hospital, Shijiazhuang, Hebei Province, 050011, China
| | - Yimeng Fan
- Teaching Division, Shijiazhuang People's Hospital, Shijiazhuang, Hebei Province, 050011, China
| |
Collapse
|
90
|
Wang J, Peng L, Yang M, Wang J, Feng R, Xu K, Xu P. Is there a genetic relationship between blood glucose and osteoarthritis? A mendelian randomization study. Diabetol Metab Syndr 2024; 16:274. [PMID: 39543708 PMCID: PMC11562302 DOI: 10.1186/s13098-024-01517-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 11/07/2024] [Indexed: 11/17/2024] Open
Abstract
OBJECTIVE The relationship between blood glucose levels and osteoarthritis (OA) is unclear. This study aimed to investigate the genetic causal relationship between blood glucose-related traits and OA. METHODS We first performed univariate Mendelian randomization (UVMR) analyses using published genome-wide association study (GWAS) datasets with fasting glucose (FG), 2 h-glucose post-challenge glucose (2hGlu), and glycosylated hemoglobin (HbA1c) as exposures, and hip osteoarthritis (HOA) and knee osteoarthritis (KOA) as outcomes; then, we performed inverse analyses of them. We used Inverse-variance weighted (IVW) analysis as the primary analysis, and sensitivity analyses were performed. Moreover, we performed multivariate Mendelian randomization (MVMR) to estimate the independent effect of exposure on outcome after adjusting for body mass index (BMI). Summarized data for blood glucose-related traits were obtained from the MAGIC Consortium study of the glucose trait genome and for OA from the UK Biobank and arcOGEN. Summarized data for BMI were obtained from the GIANT Consortium meta-analysis of individuals of European ancestry. A two-sided p value < 0.05 in UVMR was considered suggestive of significance when p < 0.0167 (Bonferroni correction p = 0.05/3 exposures) was considered statistically significant. RESULTS We found significant negative genetic causality of FG for HOA and KOA, and these associations remained significant after we adjusted for the effect of BMI [odds ratios (ORs) of 0.829 (0.687-0.999, p = 0.049) and 0.741 (0.570-0.964, p = 0.025)]. HbA1c also had an independent negative genetic causal effect on HOA after adjustment for BMI [0.665 (0.463-0.954, p = 0.027)]. At the same time, there was no evidence of reverse genetic causality of OA on blood glucose-related traits. CONCLUSION We further elucidated the relationship between blood glucose-related traits and OA by adjusting for the effect of BMI from a genetic causal perspective. This study provides new insights to further clarify the relationship between blood glucose levels and OA, as well as the pathogenesis, etiology and genetics of OA.
Collapse
Affiliation(s)
- Junxiang Wang
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710054, China
- Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Leixuan Peng
- Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Mingyi Yang
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710054, China.
| | - Jiachen Wang
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710054, China
| | - Ruoyang Feng
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710054, China
| | - Ke Xu
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710054, China
| | - Peng Xu
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710054, China.
| |
Collapse
|
91
|
Alcalde-Herraiz M, Xie J, Newby D, Prats C, Gill D, Gordillo-Marañón M, Prieto-Alhambra D, Català M, Prats-Uribe A. Effect of genetically predicted sclerostin on cardiovascular biomarkers, risk factors, and disease outcomes. Nat Commun 2024; 15:9832. [PMID: 39537602 PMCID: PMC11561231 DOI: 10.1038/s41467-024-53623-5] [Citation(s) in RCA: 3] [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: 07/31/2023] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
Sclerostin inhibitors protect against osteoporotic fractures, but their cardiovascular safety remains unclear. We conducted a cis-Mendelian randomisation analysis to estimate the causal effect of sclerostin levels on cardiovascular risk factors. We meta-analysed three GWAS of sclerostin levels including 49,568 Europeans and selected 2 SNPs to be used as instruments. We included heel bone mineral density and hip fracture risk as positive control outcomes. Public GWAS and UK Biobank patient-level data were used for the study outcomes, which include cardiovascular events, risk factors, and biomarkers. Lower sclerostin levels were associated with higher bone mineral density and 85% reduction in hip fracture risk. However, genetically predicted lower sclerostin levels led to 25-85% excess coronary artery disease risk, 40% to 60% increased risk of type 2 diabetes, and worse cardiovascular biomarkers values, including higher triglycerides, and decreased HDL cholesterol levels. Results also suggest a potential (but borderline) association with increased risk of myocardial infarction. Our study provides genetic evidence of a causal relationship between reduced levels of sclerostin and improved bone health and fracture protection, but increased risk of cardiovascular events and risk factors.
Collapse
Affiliation(s)
- Marta Alcalde-Herraiz
- Centre for Statistics in Medicine and NIHR Biomedical Research Centre Oxford, NDORMS, University of Oxford, Oxford, UK
- Computational Biology and Complex Systems (BIOCOM-SC), Department of Physics, Universitat Politècnica de Catalunya, Castelldefels, Spain
| | - JunQing Xie
- Centre for Statistics in Medicine and NIHR Biomedical Research Centre Oxford, NDORMS, University of Oxford, Oxford, UK
| | - Danielle Newby
- Centre for Statistics in Medicine and NIHR Biomedical Research Centre Oxford, NDORMS, University of Oxford, Oxford, UK
| | - Clara Prats
- Computational Biology and Complex Systems (BIOCOM-SC), Department of Physics, Universitat Politècnica de Catalunya, Castelldefels, Spain
| | - Dipender Gill
- Department of Epidemiology and Biostatistics, School of Public Health, St Mary's Hospital, Imperial College London, London, UK
| | - María Gordillo-Marañón
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London, UK
- Data Analytics and Methods Task Force, European Medicines Agency, Amsterdam, Netherlands
| | - Daniel Prieto-Alhambra
- Centre for Statistics in Medicine and NIHR Biomedical Research Centre Oxford, NDORMS, University of Oxford, Oxford, UK.
- Department of Medical Informatics, Erasmus University Medical Centre, Rotterdam, the Netherlands.
| | - Martí Català
- Centre for Statistics in Medicine and NIHR Biomedical Research Centre Oxford, NDORMS, University of Oxford, Oxford, UK
| | - Albert Prats-Uribe
- Centre for Statistics in Medicine and NIHR Biomedical Research Centre Oxford, NDORMS, University of Oxford, Oxford, UK
| |
Collapse
|
92
|
Sokolowski EK, Kursawe R, Selvam V, Bhuiyan RM, Thibodeau A, Zhao C, Spracklen CN, Ucar D, Stitzel ML. Multi-omic human pancreatic islet endoplasmic reticulum and cytokine stress response mapping provides type 2 diabetes genetic insights. Cell Metab 2024; 36:2468-2488.e7. [PMID: 39383866 PMCID: PMC11798411 DOI: 10.1016/j.cmet.2024.09.006] [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: 12/22/2023] [Revised: 06/14/2024] [Accepted: 09/10/2024] [Indexed: 10/11/2024]
Abstract
Endoplasmic reticulum (ER) and inflammatory stress responses contribute to islet dysfunction in type 2 diabetes (T2D). Comprehensive genomic understanding of these human islet stress responses and whether T2D-associated genetic variants modulate them is lacking. Here, comparative transcriptome and epigenome analyses of human islets exposed ex vivo to these stressors revealed 30% of expressed genes and 14% of islet cis-regulatory elements (CREs) as stress responsive, modulated largely in an ER- or cytokine-specific fashion. T2D variants overlapped 86 stress-responsive CREs, including 21 induced by ER stress. We linked the rs6917676-T T2D risk allele to increased islet ER-stress-responsive CRE accessibility and allele-specific β cell nuclear factor binding. MAP3K5, the ER-stress-responsive putative rs6917676 T2D effector gene, promoted stress-induced β cell apoptosis. Supporting its pro-diabetogenic role, MAP3K5 expression correlated inversely with human islet β cell abundance and was elevated in T2D β cells. This study provides genome-wide insights into human islet stress responses and context-specific T2D variant effects.
Collapse
Affiliation(s)
- Eishani K Sokolowski
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA; Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT 06032, USA
| | - Romy Kursawe
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Vijay Selvam
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Redwan M Bhuiyan
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA; Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT 06032, USA
| | - Asa Thibodeau
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Chi Zhao
- Department of Biostatistics and Epidemiology, University of Massachusetts, Amherst, Amherst, MA 01003, USA
| | - Cassandra N Spracklen
- Department of Biostatistics and Epidemiology, University of Massachusetts, Amherst, Amherst, MA 01003, USA
| | - Duygu Ucar
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA; Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT 06032, USA; Institute of Systems Genomics, University of Connecticut, Farmington, CT 06032, USA.
| | - Michael L Stitzel
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA; Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT 06032, USA; Institute of Systems Genomics, University of Connecticut, Farmington, CT 06032, USA.
| |
Collapse
|
93
|
Ghatan S, Koromani F, Trajanoska K, van Velsen EFS, Kavousi M, Zillikens MC, Medina-Gomez C, Oei L, Rivadeneira F. Evaluating the relationship between glycemic control and bone fragility within the UK Biobank: observational and one-sample Mendelian randomization analyses. JBMR Plus 2024; 8:ziae126. [PMID: 39469527 PMCID: PMC11515132 DOI: 10.1093/jbmrpl/ziae126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 09/02/2024] [Indexed: 10/30/2024] Open
Abstract
We aimed to: (1) examine the relationship between glycemic control, BMD estimated from heel ultrasound (eBMD) and fracture risk in individuals with type 1 (T1D) and type 2 diabetes (T2D) and (2) perform a one-sample Mendelian randomization (MR) study to explore potential causal associations between glycemic control, eBMD, and fractures. This study comprised 452 131 individuals from the UK Biobank with glycated hemoglobin A1C (HbA1c) and eBMD levels. At baseline, 4078 participants were diagnosed with T1D and 23 682 with T2D. HbA1c was used to classify patients into "adequately-" (ACD; n = 17 078; HbA1c < 7.0%/53 mmol/mol) and "inadequately-" (ICD; n = 10 682; HbA1c ≥ 7.0%/53 mmol/mol) controlled diabetes. In individuals with T1D, a 1% unit (11 mmol/mol) increase in HbA1c levels was associated with a 12% increase in fracture risk (HR: 1.12, 95% CI [1.05-1.19]). Fracture risk was highest in individuals with T1D and ICD (HR 2.84, 95%CI [2.53, 3.19]), followed by those with ACD (HR 2.26, 95%CI [1.91, 2.69]), as compared to subjects without diabetes. Evidence for a non-linear association between HbA1c and fracture risk was observed (F-test ANOVA p-value = 0.002) in individuals with T2D, with risk being increased at both low and high levels of HbA1c. Fracture risk between the T2D ACD and ICD groups was not significantly different (HR: 0.97, 95%CI [0.91-1.16]), despite increased BMD. In MR analyses genetically predicted higher HbA1c levels were not significantly associated with fracture risk (causal risk ratio: 1.04, 95%CI [0.95-1.14]). We did observe evidence of a non-linear causal association with eBMD (quadratic test p-value = 0.0002), indicating U-shaped relationship between HbA1c and eBMD. We obtained evidence that lower HbA1c levels will reduce fracture risk in patients with T1D. In individuals with T2D, lowering HbA1c levels can mitigate the risk of fractures up to a threshold, beyond which the risk may begin to rise again.
Collapse
Affiliation(s)
- Samuel Ghatan
- Department of Internal Medicine, Erasmus Medical Center, 3015 GD, Rotterdam, The Netherlands
| | - Fjorda Koromani
- Department of Internal Medicine, Erasmus Medical Center, 3015 GD, Rotterdam, The Netherlands
| | - Katerina Trajanoska
- Canada Excellence Research Chair in Genomic Medicine, Victor Philip Dahdaleh Institute of Genomic Medicine, Department of Human Genetics, Faculty of Medicine and Health Sciences, McGill University, QC H3G 2M1, Montreal, QC, Canada
| | - Evert F S van Velsen
- Department of Internal Medicine, Erasmus Medical Center, 3015 GD, Rotterdam, The Netherlands
| | - Maryam Kavousi
- Department of Epidemiology, Erasmus Medical Center, 3015 GD, Rotterdam, The Netherlands
| | - M Carola Zillikens
- Department of Internal Medicine, Erasmus Medical Center, 3015 GD, Rotterdam, The Netherlands
| | - Carolina Medina-Gomez
- Department of Internal Medicine, Erasmus Medical Center, 3015 GD, Rotterdam, The Netherlands
| | - Ling Oei
- Department of Internal Medicine, Erasmus Medical Center, 3015 GD, Rotterdam, The Netherlands
| | - Fernando Rivadeneira
- Department of Internal Medicine, Erasmus Medical Center, 3015 GD, Rotterdam, The Netherlands
| |
Collapse
|
94
|
Miao J, Wu Y, Sun Z, Miao X, Lu T, Zhao J, Lu Q. Valid inference for machine learning-assisted genome-wide association studies. Nat Genet 2024; 56:2361-2369. [PMID: 39349818 PMCID: PMC11972620 DOI: 10.1038/s41588-024-01934-0] [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: 02/13/2024] [Accepted: 08/29/2024] [Indexed: 11/10/2024]
Abstract
Machine learning (ML) has become increasingly popular in almost all scientific disciplines, including human genetics. Owing to challenges related to sample collection and precise phenotyping, ML-assisted genome-wide association study (GWAS), which uses sophisticated ML techniques to impute phenotypes and then performs GWAS on the imputed outcomes, have become increasingly common in complex trait genetics research. However, the validity of ML-assisted GWAS associations has not been carefully evaluated. Here, we report pervasive risks for false-positive associations in ML-assisted GWAS and introduce Post-Prediction GWAS (POP-GWAS), a statistical framework that redesigns GWAS on ML-imputed outcomes. POP-GWAS ensures valid and powerful statistical inference irrespective of imputation quality and choice of algorithm, requiring only GWAS summary statistics as input. We employed POP-GWAS to perform a GWAS of bone mineral density derived from dual-energy X-ray absorptiometry imaging at 14 skeletal sites, identifying 89 new loci and revealing skeletal site-specific genetic architecture. Our framework offers a robust analytic solution for future ML-assisted GWAS.
Collapse
Affiliation(s)
- Jiacheng Miao
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, USA
| | - Yixuan Wu
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, USA
| | - Zhongxuan Sun
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, USA
| | - Xinran Miao
- Department of Statistics, University of Wisconsin-Madison, Madison, WI, USA
| | - Tianyuan Lu
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
- Department of Statistical Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Jiwei Zhao
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, USA
- Department of Statistics, University of Wisconsin-Madison, Madison, WI, USA
| | - Qiongshi Lu
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, USA.
- Department of Statistics, University of Wisconsin-Madison, Madison, WI, USA.
- Center for Demography of Health and Aging, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
95
|
Yang T, Yuan X, Gao W, Hu MJ, Lu MJ, Sun HS. Mendelian randomization did not support the causal effect of diabetes on aortic diseases. Intern Emerg Med 2024; 19:2185-2192. [PMID: 39210233 DOI: 10.1007/s11739-024-03727-z] [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: 04/14/2023] [Accepted: 12/11/2023] [Indexed: 09/04/2024]
Abstract
Observational studies revealed paradoxically inverse associations between diabetes and aortic diseases (aortic aneurysm or aortic dissection), yet the causality remains to be determined. To investigate the causal associations between diabetes and aortic diseases using Mendelian randomization (MR) analyses. Summary-level data for exposures (type 1 diabetes, type 2 diabetes, fasting glucose, fasting insulin, glycated hemoglobin) and outcomes (aortic dissection and aortic aneurysm) were obtained from public genome-wide association study data. The principal analysis was the inverse-variance weighted (IVW) method. Sensitivity analyses were also carried out, including weighted median, MR-Egger, and multivariable MR methods. According to IVW results, type 1 diabetes (odds ratio [OR]: 0.99; 95% confidence interval [CI] 0.93-1.07; P = 0.87), type 2 diabetes (OR: 0.97; 95% CI 0.77-1.20; P = 0.75), fasting glucose (OR: 1.16; 95% CI 0.48-2.84; P = 0.74), fasting insulin (OR: 2.75; 95% CI 0.53-14.26; P = 0.23), or glycated hemoglobin (OR: 0.33; 95% CI 0.09-1.17; P = 0.09) had no causal effect on aortic dissection. Similarly, type 1 diabetes, type 2 diabetes, fasting glucose, fasting insulin, or glycated hemoglobin had no causal effect on aortic aneurysm. Sensitivity analyses revealed consistent results. MR-Egger method and funnel plot yielded no indication of directional pleiotropy. Diabetes had no causal associations with aortic dissection or aortic aneurysm. The observed inverse associations in previous cohort studies may be explained by confounding factors or reverse causation.
Collapse
Affiliation(s)
- Tao Yang
- Department of Cardiovascular Surgery, Cardiovascular Institute and Fu Wai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, No. 167 North Lishi Road, Xicheng District, Beijing, 100037, China
| | - Xin Yuan
- Department of Cardiovascular Surgery, Cardiovascular Institute and Fu Wai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, No. 167 North Lishi Road, Xicheng District, Beijing, 100037, China
| | - Wei Gao
- Department of Cardiovascular Surgery, Cardiovascular Institute and Fu Wai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, No. 167 North Lishi Road, Xicheng District, Beijing, 100037, China
| | - Meng-Jin Hu
- Department of Cardiovascular Surgery, Cardiovascular Institute and Fu Wai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, No. 167 North Lishi Road, Xicheng District, Beijing, 100037, China
| | - Min-Jie Lu
- Department of Magnetic Resonance Imaging, Cardiovascular Institute and Fu Wai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, No. 167 North Lishi Road, Xicheng District, Beijing, 100037, China.
| | - Han-Song Sun
- Department of Cardiovascular Surgery, Cardiovascular Institute and Fu Wai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, No. 167 North Lishi Road, Xicheng District, Beijing, 100037, China.
| |
Collapse
|
96
|
Olwi DI, Kaisinger LR, Kentistou KA, Vaudel M, Stankovic S, Njølstad PR, Johansson S, Perry JRB, Day FR, Ong KK. Likely causal effects of insulin resistance and IGF-1 bioaction on childhood and adult adiposity: a Mendelian randomization study. Int J Obes (Lond) 2024; 48:1650-1655. [PMID: 39174749 PMCID: PMC11502485 DOI: 10.1038/s41366-024-01605-4] [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: 09/08/2023] [Revised: 07/23/2024] [Accepted: 08/06/2024] [Indexed: 08/24/2024]
Abstract
BACKGROUND Circulating insulin and insulin-like growth factor-1 (IGF-1) concentrations are positively correlated with adiposity. However, the causal effects of insulin and IGF-1 on adiposity are unclear. METHODS We performed two-sample Mendelian randomization analyses to estimate the likely causal effects of fasting insulin and IGF-1 on relative childhood adiposity and adult body mass index (BMI). To improve accuracy and biological interpretation, we applied Steiger filtering (to avoid reverse causality) and 'biological effect' filtering of fasting insulin and IGF-1 associated variants. RESULTS Fasting insulin-increasing alleles (35 variants also associated with higher fasting glucose, indicative of insulin resistance) were associated with lower relative childhood adiposity (P = 3.8 × 10-3) and lower adult BMI (P = 1.4 × 10-5). IGF-1-increasing alleles also associated with taller childhood height (351 variants indicative of greater IGF-1 bioaction) showed no association with relative childhood adiposity (P = 0.077) or adult BMI (P = 0.562). Conversely, IGF-1-increasing alleles also associated with shorter childhood height (306 variants indicative of IGF-1 resistance) were associated with lower relative childhood adiposity (P = 6.7 × 10-3), but effects on adult BMI were inconclusive. CONCLUSIONS Genetic causal modelling indicates negative effects of insulin resistance on childhood and adult adiposity, and negative effects of IGF-1 resistance on childhood adiposity. Our findings demonstrate the need to distinguish between bioaction and resistance when modelling variants associated with biomarker concentrations.
Collapse
Affiliation(s)
- Duaa I Olwi
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK
- King Abdullah International Medical Research Center, Jeddah, Saudi Arabia
- King Saud bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia
| | - Lena R Kaisinger
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Katherine A Kentistou
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Marc Vaudel
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, NO-5020, Bergen, Norway
- Department of Genetics and Bioinformatics, Health Data and Digitalization, Norwegian Institute of Public Health, NO-0213, Oslo, Norway
| | - Stasa Stankovic
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Pål R Njølstad
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, NO-5020, Bergen, Norway
- Department of Pediatrics and Adolescents, Haukeland University Hospital, NO-5021, Bergen, Norway
| | - Stefan Johansson
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, NO-5020, Bergen, Norway
- Department of Medical Genetics, Haukeland University Hospital, NO-5021, Bergen, Norway
| | - John R B Perry
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK
- Metabolic Research Laboratory, Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Felix R Day
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Ken K Ong
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK.
- Department of Paediatrics, University of Cambridge, Cambridge, UK.
| |
Collapse
|
97
|
Sass F, Ma T, Ekberg JH, Kirigiti M, Ureña MG, Dollet L, Brown JM, Basse AL, Yacawych WT, Burm HB, Andersen MK, Nielsen TS, Tomlinson AJ, Dmytiyeva O, Christensen DP, Bader L, Vo CT, Wang Y, Rausch DM, Kristensen CK, Gestal-Mato M, In Het Panhuis W, Sjøberg KA, Kernodle S, Petersen JE, Pavlovskyi A, Sandhu M, Moltke I, Jørgensen ME, Albrechtsen A, Grarup N, Babu MM, Rensen PCN, Kooijman S, Seeley RJ, Worthmann A, Heeren J, Pers TH, Hansen T, Gustafsson MBF, Tang-Christensen M, Kilpeläinen TO, Myers MG, Kievit P, Schwartz TW, Hansen JB, Gerhart-Hines Z. NK2R control of energy expenditure and feeding to treat metabolic diseases. Nature 2024; 635:987-1000. [PMID: 39537932 PMCID: PMC11602716 DOI: 10.1038/s41586-024-08207-0] [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] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 10/11/2024] [Indexed: 11/16/2024]
Abstract
The combination of decreasing food intake and increasing energy expenditure represents a powerful strategy for counteracting cardiometabolic diseases such as obesity and type 2 diabetes1. Yet current pharmacological approaches require conjugation of multiple receptor agonists to achieve both effects2-4, and so far, no safe energy-expending option has reached the clinic. Here we show that activation of neurokinin 2 receptor (NK2R) is sufficient to suppress appetite centrally and increase energy expenditure peripherally. We focused on NK2R after revealing its genetic links to obesity and glucose control. However, therapeutically exploiting NK2R signalling has previously been unattainable because its endogenous ligand, neurokinin A, is short-lived and lacks receptor specificity5,6. Therefore, we developed selective, long-acting NK2R agonists with potential for once-weekly administration in humans. In mice, these agonists elicit weight loss by inducing energy expenditure and non-aversive appetite suppression that circumvents canonical leptin signalling. Additionally, a hyperinsulinaemic-euglycaemic clamp reveals that NK2R agonism acutely enhances insulin sensitization. In diabetic, obese macaques, NK2R activation significantly decreases body weight, blood glucose, triglycerides and cholesterol, and ameliorates insulin resistance. These findings identify a single receptor target that leverages both energy-expending and appetite-suppressing programmes to improve energy homeostasis and reverse cardiometabolic dysfunction across species.
Collapse
Affiliation(s)
- Frederike Sass
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
- Center for Adipocyte Signaling (ADIPOSIGN), University of Southern Denmark, Odense, Denmark
| | - Tao Ma
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Jeppe H Ekberg
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
- Embark Laboratories, Copenhagen, Denmark
| | - Melissa Kirigiti
- Division of Metabolic Health and Disease, Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Mario G Ureña
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Lucile Dollet
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Jenny M Brown
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Astrid L Basse
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Warren T Yacawych
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Hayley B Burm
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Mette K Andersen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Thomas S Nielsen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | | | - Oksana Dmytiyeva
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Dan P Christensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
- Embark Laboratories, Copenhagen, Denmark
| | - Lindsay Bader
- Division of Metabolic Health and Disease, Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Camilla T Vo
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
- Neuroscience Academy Denmark, Copenhagen, Denmark
| | - Yaxu Wang
- Center for Adipocyte Signaling (ADIPOSIGN), University of Southern Denmark, Odense, Denmark
- Center of Excellence for Data Driven Discovery, Department of Structural Biology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Dylan M Rausch
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Cecilie K Kristensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - María Gestal-Mato
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Wietse In Het Panhuis
- Department of Medicine, Division of Endocrinology and Einthoven Laboratory of Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Kim A Sjøberg
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Stace Kernodle
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Jacob E Petersen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Artem Pavlovskyi
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Manbir Sandhu
- Center for Adipocyte Signaling (ADIPOSIGN), University of Southern Denmark, Odense, Denmark
- Center of Excellence for Data Driven Discovery, Department of Structural Biology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Ida Moltke
- Section for Computational and RNA Biology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Marit E Jørgensen
- Clinical Research, Copenhagen University Hospital - Steno Diabetes Center Copenhagen, Herlev, Denmark
- Centre for Public Health in Greenland, National Institute of Public Health, University of Southern Denmark, Copenhagen, Denmark
- Steno Diabetes Center Greenland, Nuuk, Greenland
| | - Anders Albrechtsen
- Section for Computational and RNA Biology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Niels Grarup
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - M Madan Babu
- Center for Adipocyte Signaling (ADIPOSIGN), University of Southern Denmark, Odense, Denmark
- Center of Excellence for Data Driven Discovery, Department of Structural Biology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Patrick C N Rensen
- Department of Medicine, Division of Endocrinology and Einthoven Laboratory of Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Sander Kooijman
- Department of Medicine, Division of Endocrinology and Einthoven Laboratory of Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Randy J Seeley
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Anna Worthmann
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Joerg Heeren
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tune H Pers
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Magnus B F Gustafsson
- Embark Laboratories, Copenhagen, Denmark
- Chemical Process Research and Development, Chemical Process Research & DevelopmentLEO Pharma, Ballerup, Denmark
| | - Mads Tang-Christensen
- Embark Laboratories, Copenhagen, Denmark
- School of Biomedical Sciences Faculty of Medicine, Nursing and Health Sciences Monash University, Melbourne, Victoria, Australia
| | - Tuomas O Kilpeläinen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Martin G Myers
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Paul Kievit
- Division of Metabolic Health and Disease, Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Thue W Schwartz
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
- Embark Laboratories, Copenhagen, Denmark
| | - Jakob B Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark.
- Embark Laboratories, Copenhagen, Denmark.
| | - Zachary Gerhart-Hines
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark.
- Center for Adipocyte Signaling (ADIPOSIGN), University of Southern Denmark, Odense, Denmark.
- Embark Laboratories, Copenhagen, Denmark.
| |
Collapse
|
98
|
Zhao JV, Zhang J. Using Genetics to Assess the Role of Acetate in Ischemic Heart Disease, Diabetes, and Sex-Hormone-Related Cancers: A Mendelian Randomization Study. Nutrients 2024; 16:3674. [PMID: 39519507 PMCID: PMC11547320 DOI: 10.3390/nu16213674] [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: 09/09/2024] [Revised: 10/16/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Acetate, a short-chain fatty acid, has gained attention for its contrasting roles, with evidence suggesting it may offer cardiovascular protection but also promote cancer, particularly those involving sex hormones. However, these influences have been scarcely assessed in epidemiological research. OBJECTIVE To investigate the relationship between acetate and ischemic heart disease (IHD), diabetes, and cancers related to sex hormones. METHODS Mendelian randomization (MR) was used to assess potential causal effects, selecting genetic variants without linkage disequilibrium (r2 < 0.001) and with genome-wide significance for acetate (p < 5 × 10-8). These variants were applied to large genome-wide association studies (GWAS) for ischemic heart disease (IHD; up to 154,373 cases), diabetes (109,731 cases), and five sex-hormone-related cancers (breast, colorectal, prostate, ovarian, and endometrial cancers, ranging from 8679 to 122,977 cases). We employed various methods for analysis, including penalized inverse variance weighting (pIVW), inverse variance weighting, weighted mode, and weighted median. RESULTS This study indicates that acetate may be associated with a lower risk of ischemic heart disease (IHD), with an odds ratio (OR) of 0.62 per standard deviation (SD) increase in acetate and a 95% confidence interval (CI) of 0.39 to 0.98. Additionally, acetate was linked to a higher breast cancer risk, with an OR of 1.26 and a 95% CI ranging from 1.08 to 1.46. This association remained robust across multiple sensitivity analyses. CONCLUSIONS Acetate, along with factors that influence its activity, may serve as possible targets for breast cancer treatment and possibly IHD, offering opportunities for new drug development.
Collapse
Affiliation(s)
- Jie V. Zhao
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China;
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China
| | - Junmeng Zhang
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China;
| |
Collapse
|
99
|
Lee DSM, Cardone KM, Zhang DY, Tsao NL, Abramowitz S, Sharma P, DePaolo JS, Conery M, Aragam KG, Biddinger K, Dilitikas O, Hoffman-Andrews L, Judy RL, Khan A, Kulo I, Puckelwartz MJ, Reza N, Satterfield BA, Singhal P, Arany ZP, Cappola TP, Carruth E, Day SM, Do R, Haggarty CM, Joseph J, McNally EM, Nadkarni G, Owens AT, Rader DJ, Ritchie MD, Sun YV, Voight BF, Levin MG, Damrauer SM. Common- and rare-variant genetic architecture of heart failure across the allele frequency spectrum. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2023.07.16.23292724. [PMID: 37503172 PMCID: PMC10371173 DOI: 10.1101/2023.07.16.23292724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Heart failure (HF) is a complex trait, influenced by environmental and genetic factors, which affects over 30 million individuals worldwide. Historically, the genetics of HF have been studied in Mendelian forms of disease, where rare genetic variants have been linked to familial cardiomyopathies. More recently, genome-wide association studies (GWAS) have successfully identified common genetic variants associated with risk of HF. However, the relative importance of genetic variants across the allele-frequency spectrum remains incompletely characterized. Here, we report the results of common- and rare-variant association studies of all-cause heart failure, applying recently developed methods to quantify the heritability of HF attributable to different classes of genetic variation. We combine GWAS data across multiple populations including 207,346 individuals with HF and 2,151,210 without, identifying 176 risk loci at genome-wide significance (P-value < 5×10-8). Signals at newly identified common-variant loci include coding variants in Mendelian cardiomyopathy genes (MYBPC3, BAG3) and in regulators of lipoprotein (LPL) and glucose metabolism (GIPR, GLP1R). These signals are enriched in myocyte and adipocyte cell types and can be clustered into 5 broad modules based on pleiotropic associations with anthropomorphic traits/obesity, blood pressure/renal function, atherosclerosis/lipids, immune activity, and arrhythmias. Gene burden studies across three biobanks (PMBB, UKB, AOU), including 27,208 individuals with HF and 349,126 without, uncover exome-wide significant (P-value < 1.57×10-6) associations for HF and rare predicted loss-of-function (pLoF) variants in TTN, MYBPC3, FLNC, and BAG3. Total burden heritability of rare coding variants (2.2%, 95% CI 0.99-3.5%) is highly concentrated in a small set of Mendelian cardiomyopathy genes, while common variant heritability (4.3%, 95% CI 3.9-4.7%) is more diffusely spread throughout the genome. Finally, we show that common-variant background, in the form of a polygenic risk score (PRS), significantly modifies the risk of HF among carriers of pathogenic truncating variants in the Mendelian cardiomyopathy gene TTN. Together, these findings provide a genetic link between dysregulated metabolism and HF, and suggest a significant polygenic component to HF exists that is not captured by current clinical genetic testing.
Collapse
Affiliation(s)
- David S M Lee
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Kathleen M Cardone
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - David Y Zhang
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Noah L Tsao
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Sarah Abramowitz
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Pranav Sharma
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - John S DePaolo
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Mitchell Conery
- Genomics and Computational Biology Graduate Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Krishna G Aragam
- Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Program in Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA
| | - Kiran Biddinger
- Program in Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA
| | - Ozan Dilitikas
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | - Lily Hoffman-Andrews
- Division of Cardiovascular Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Renae L Judy
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Atlas Khan
- Division of Nephrology, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY
| | - Iftikhar Kulo
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | - Megan J Puckelwartz
- Department of Pharmacology, Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Nosheen Reza
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | | | - Pankhuri Singhal
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Zoltan P Arany
- Division of Cardiovascular Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
- Cardiovascular Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Thomas P Cappola
- Division of Cardiovascular Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Eric Carruth
- Department of Translational Data Science and Informatics, Geisinger, Danville, PA
| | - Sharlene M Day
- Division of Cardiovascular Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
- Cardiovascular Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Ron Do
- The Charles Bronfman Institute for Personalized Medicine, Mount Sinai Icahn School of Medicine, New York, NY
- Biome Phenomics Center, Mount Sinai Icahn School of Medicine, New York, NY
- Department of Genetics and Genomic Sciences, Mount Sinai Icahn School of Medicine, New York, NY
| | | | - Jacob Joseph
- Massachusetts Veterans Epidemiology Research and Information Center, VA Boston Healthcare System, Boston, MA
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Elizabeth M McNally
- Center for Genetic Medicine, Bluhm Cardiovascular Institute, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Girish Nadkarni
- Division of Nephrology, Department of Medicine, Mount Sinai Icahn School of Medicine, New York, NY
| | - Anjali T Owens
- Cardiovascular Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Daniel J Rader
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
- Division of Translational Medicine and Human Genetics, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Marylyn D Ritchie
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
- Institute for Biomedical Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Yan V Sun
- Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA
- Atlanta VA Health Care System, Decatur, GA
| | - Benjamin F Voight
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA
| | - Michael G Levin
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA
| | - Scott M Damrauer
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
- Cardiovascular Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA
| |
Collapse
|
100
|
Jacobs BM, Stow D, Hodgson S, Zöllner J, Samuel M, Kanoni S, Bidi S, Walter K, Langenberg C, Dobson R, Finer S, Morton C, Siddiqui MK, Martin HC, Pietzner M, Mathur R, van Heel DA. Genetic architecture of routinely acquired blood tests in a British South Asian cohort. Nat Commun 2024; 15:8929. [PMID: 39414775 PMCID: PMC11484750 DOI: 10.1038/s41467-024-53091-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: 09/30/2024] [Indexed: 10/18/2024] Open
Abstract
Understanding the genetic basis of routinely-acquired blood tests can provide insights into several aspects of human physiology. We report a genome-wide association study of 42 quantitative blood test traits defined using Electronic Healthcare Records (EHRs) of ~50,000 British Bangladeshi and British Pakistani adults. We demonstrate a causal variant within the PIEZO1 locus which was associated with alterations in red cell traits and glycated haemoglobin. Conditional analysis and within-ancestry fine mapping confirmed that this signal is driven by a missense variant - chr16-88716656-G-TT - which is common in South Asian ancestries (MAF 3.9%) but ultra-rare in other ancestries. Carriers of the T allele had lower mean HbA1c values, lower HbA1c values for a given level of random or fasting glucose, and delayed diagnosis of Type 2 Diabetes Mellitus. Our results shed light on the genetic basis of clinically-relevant traits in an under-represented population, and emphasise the importance of ancestral diversity in genetic studies.
Collapse
Grants
- 210561/Z/18/Z Wellcome Trust
- WT102627 Wellcome Trust (Wellcome)
- MR/V028766/1 RCUK | Medical Research Council (MRC)
- Wellcome Trust
- M009017 RCUK | Medical Research Council (MRC)
- Genes & Health is/has recently been core-funded by Wellcome (WT102627, WT210561), the Medical Research Council (UK) (M009017, MR/X009777/1, MR/X009920/1), Higher Education Funding Council for England Catalyst, Barts Charity (845/1796), Health Data Research UK (for London substantive site), and research delivery support from the NHS National Institute for Health Research Clinical Research Network (North Thames). Genes & Health is/has recently been funded by Alnylam Pharmaceuticals, Genomics PLC; and a Life Sciences Industry Consortium of Astra Zeneca PLC, Bristol-Myers Squibb Company, GlaxoSmithKline Research and Development Limited, Maze Therapeutics Inc, Merck Sharp & Dohme LLC, Novo Nordisk A/S, Pfizer Inc, Takeda Development Centre Americas Inc.
Collapse
Affiliation(s)
- Benjamin M Jacobs
- Wolfson Institute of Population Health, Queen Mary University of London, London, UK
- Department of Neurology, Royal London Hospital, Barts Health NHS Trust, London, UK
| | - Daniel Stow
- Wolfson Institute of Population Health, Queen Mary University of London, London, UK
| | - Sam Hodgson
- Wolfson Institute of Population Health, Queen Mary University of London, London, UK
| | - Julia Zöllner
- Wolfson Institute of Population Health, Queen Mary University of London, London, UK
- University College London, London, UK
| | - Miriam Samuel
- Wolfson Institute of Population Health, Queen Mary University of London, London, UK
| | - Stavroula Kanoni
- William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Saeed Bidi
- Wolfson Institute of Population Health, Queen Mary University of London, London, UK
| | - Klaudia Walter
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, 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
| | - Ruth Dobson
- Wolfson Institute of Population Health, Queen Mary University of London, London, UK
- Department of Neurology, Royal London Hospital, Barts Health NHS Trust, London, UK
| | - Sarah Finer
- Wolfson Institute of Population Health, Queen Mary University of London, London, UK
- Blizard Institute, Queen Mary University of London, London, UK
| | - Caroline Morton
- Wolfson Institute of Population Health, Queen Mary University of London, London, UK
| | - Moneeza K Siddiqui
- Wolfson Institute of Population Health, Queen Mary University of London, London, UK
| | - Hilary C Martin
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, 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
| | - Rohini Mathur
- Wolfson Institute of Population Health, Queen Mary University of London, London, UK
| | - David A van Heel
- Wolfson Institute of Population Health, Queen Mary University of London, London, UK.
- Blizard Institute, Queen Mary University of London, London, UK.
| |
Collapse
|