1
|
Zhang Z, Zhang P, Zhang F, Zhong J, Sun W, Xiong H. Genetic insights into the risk of frailty on metabolic syndrome and its components: Bidirectional Mendelian randomization study. Nutr Metab Cardiovasc Dis 2025; 35:103898. [PMID: 39993952 DOI: 10.1016/j.numecd.2025.103898] [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: 02/21/2024] [Revised: 01/21/2025] [Accepted: 01/25/2025] [Indexed: 02/26/2025]
Abstract
BACKGROUND AND AIMS Previous studies have shown that frailty and metabolic syndrome (Mets) share common pathophysiological mechanisms. However, whether the observed association reflects causality requires clarification. We performed a bidirectional Mendelian randomization study to investigate the causal relationship between frailty, Mets, and their individual components. METHODS AND RESULTS Summary-level data from GWAS to identify genetic variants associated with frailty, Mets, and its components among individuals of European ancestry. Inverse variance weighting was utilized as the main method. Using bidirectional Mendelian randomization analysis, we found that the risk of frailty was causally associated with an increased risk of MetS (OR: 2.092, 95%CI: 1.564-2.799) and its components, including waist circumference (OR: 1.349, 95 % CI: 1.181-1.541), hypertension (OR: 1.099, 95 % CI: 1.075-1.125), triglycerides (OR: 1.297, 95 % CI: 1.179-1.428). Conversely, the risk of MetS was causally associated with an increased risk of frailty (OR: 1.048; 95 % CI: 1.024-1.073). however, when removing SNPs assocaited with BMI at the loci significance level and performed MVMR, Mets and frailty were not associated. CONCLUSION These findings suggest a bidirectional causal relationship between frailty and MetS, indicating that genetic factors contributing to frailty also increase the risk of MetS and its components, and vice versa. Furthermore, BMI-related SNPs may act as effect modifiers in the association between MetS and frailty. These insights into the shared pathophysiology of frailty and MetS have implications for the prevention and treatment strategies in elderly individuals with MetS.
Collapse
Affiliation(s)
- Zihang Zhang
- Department of Cardiovascular Surgery ICU, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230001, China
| | - Pan Zhang
- Department of Neurology, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Feng Zhang
- Department of Neurology, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Jinghui Zhong
- Department of Neurology, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Wen Sun
- Department of Neurology, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Houren Xiong
- Department of Cardiovascular Surgery ICU, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230001, China.
| |
Collapse
|
2
|
Prete V, Di Pietro P, Abate AC, Venturini E, Iside C, Vecchione C, Carrizzo A. TRIB1: a multifaceted regulator of cardiometabolic health. Am J Physiol Cell Physiol 2025; 328:C1973-C1981. [PMID: 40331689 DOI: 10.1152/ajpcell.00231.2025] [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: 03/13/2025] [Revised: 04/02/2025] [Accepted: 05/01/2025] [Indexed: 05/08/2025]
Abstract
Cardiovascular diseases (CVDs) are the leading cause of morbidity and mortality worldwide. The rising prevalence of CVD is primarily driven by several risk factors, including dyslipidemia, atherosclerosis, diabetes, and obesity. Many current studies are focused on unraveling the underlying pathophysiological mechanisms that govern these risk factors, with the main goal of identifying novel biomarkers and therapeutic targets to prevent the onset of CVD in the population. In recent decades, genome-wide association studies (GWASs) have linked the 8q24 locus containing the TRIB1 (Tribbles homolog 1) gene to various cardiometabolic traits in humans, such as plasma triglycerides, LDL cholesterol, HDL cholesterol, total cholesterol, adiponectin, and glycated hemoglobin levels. Emerging research has investigated the role of Trib1 in regulating plasma lipid levels, inflammation, and insulin signaling, opening new avenues for the potential therapeutic role of Trib1 in CVD risk assessment. Accordingly, this review aims to explore the crucial role of Trib1 as a therapeutic biomarker in CVDs, with a focus on its association with lipid metabolism, atherosclerosis, obesity, and diabetes, analyzing in vitro and in vivo studies and offering insights into its underlying molecular mechanisms.
Collapse
Affiliation(s)
- Valeria Prete
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Baronissi, Italy
- Department of Biology, University of Naples Federico II, Complesso Universitario Monte S. Angelo, Naples, Italy
| | - Paola Di Pietro
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Baronissi, Italy
| | - Angela Carmelita Abate
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Baronissi, Italy
| | | | - Concetta Iside
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Baronissi, Italy
| | - Carmine Vecchione
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Baronissi, Italy
- Vascular Pathophysiology Unit, IRCCS Neuromed, Pozzilli, Italy
| | - Albino Carrizzo
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Baronissi, Italy
- Vascular Pathophysiology Unit, IRCCS Neuromed, Pozzilli, Italy
| |
Collapse
|
3
|
Chebii VJ, Wade AN, Crowther NJ, Nonterah EA, Agongo G, Simayi Z, Boua PR, Kisiangani I, Ramsay M, Choudhury A, Sengupta D. Genome-wide association study identifying novel risk variants associated with glycaemic traits in the continental African AWI-Gen cohort. Diabetologia 2025; 68:1184-1196. [PMID: 40025146 PMCID: PMC12069158 DOI: 10.1007/s00125-025-06395-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 01/24/2025] [Indexed: 03/04/2025]
Abstract
AIMS/HYPOTHESIS Glycaemic traits such as high fasting glucose levels and insulin resistance are positively associated with the risk of type 2 diabetes and other cardiometabolic diseases. Genetic association studies have identified hundreds of associations for each glycaemic trait, yet very few studies have involved continental African populations. We report the results of genome-wide association studies (GWASs) in a pan-African cohort for four glycaemic traits, namely fasting glucose, fasting insulin, insulin resistance (HOMA-IR) and beta cell function (HOMA-B), which are quantitative variables that affect the risk of developing type 2 diabetes. METHODS GWASs for the four traits were conducted in approximately 10,000 individuals from the Africa Wits-INDEPTH Partnership for Genomics Studies (AWI-Gen) cohort, with participants from Burkina Faso, Ghana, Kenya and South Africa. Association testing was performed using linear mixed models implemented in BOLT-LMM, with age, sex, BMI and principal components as covariates. Replication, fine mapping and functional annotation were performed using standard approaches. RESULTS We identified a novel signal (rs574173815) in the intron of the ankyrin repeat domain 33B (ANKRD33B) gene associated with fasting glucose, and a novel signal (rs114029796) in the intronic region of the WD repeat domain 7 (WDR7) gene associated with fasting insulin. SNPs in WDR7 have been shown to be associated with type 2 diabetes. A variant (rs74806991) in the intron of ADAM metallopeptidase with thrombospondin type 1 motif 16 (ADAMTS16) and another variant (rs6506934) in the β-1,4-galactosyltransferase 6 gene (B4GALT6) are associated with HOMA-IR. Both ADAMTS16 and B4GALT6 are implicated in the development of type 2 diabetes. In addition, our study replicated several well-established fasting glucose signals in the GCK-YTK6, SLC2A2 and THORLNC gene regions. CONCLUSIONS/INTERPRETATION Our findings highlight the importance of performing GWASs for glycaemic traits in under-represented populations, especially continental African populations, to discover novel associated variants and broaden our knowledge of the genetic aetiology of glycaemic traits. The limited replication of well-known signals in this study hints at the possibility of a unique genetic architecture of these traits in African populations. DATA AVAILABILITY The dataset used in this study is available in the European Genome-Phenome Archive (EGA) database ( https://ega-archive.org/ ) under study accession code EGAS00001002482. The phenotype dataset accession code is EGAD00001006425 and the genotype dataset accession code is EGAD00010001996. The availability of these datasets is subject to controlled access by the Data and Biospecimen Access Committee of the H3Africa Consortium. GWAS summary statistics are accessible through the NHGRI-EBI GWAS Catalog ( https://www.ebi.ac.uk/gwas/ ).
Collapse
Affiliation(s)
- Vivien J Chebii
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.
| | - Alisha N Wade
- Department of Internal Medicine, School of Clinical Medicine, University of the Witwatersrand, Johannesburg, South Africa
- Research in Metabolism and Endocrinology, Division of Endocrinology, Diabetes and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- MRC/Wits Rural Public Health and Health Transitions Research Unit, School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Nigel J Crowther
- Department of Chemical Pathology, National Health Laboratory Service, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Engelbert A Nonterah
- Navrongo Health Research Centre, Ghana Health Service, Navrongo, Ghana
- Department of Epidemiology, School of Public Health, C.K. Tedam University of Technology and Allied Sciences, Navrongo, Ghana
- Julius Global Health, Julius Centre for Health Sciences and Primary Care, University Medical Centre Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Godfred Agongo
- Department of Biochemistry and Forensic Sciences, School of Chemical and Biochemical Sciences, C.K. Tedam University of Technology and Applied Sciences, Navrongo, Ghana
| | - Z Simayi
- Department of Pathology, Faculty of Health Sciences, University of Limpopo, Polokwane, South Africa
| | - Palwende R Boua
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Clinical Research Unit of Nanoro, Institut de Recherche en Sciences de la Santè, Nanoro, Burkina Faso
- MRC Unit The Gambia at London School of Hygiene and Tropical Medicine, Banjul, The Gambia
| | | | - Michèle Ramsay
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Division of Human Genetics, National Health Laboratory Service and School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Ananyo Choudhury
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.
| | - Dhriti Sengupta
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.
| |
Collapse
|
4
|
Huang X, Zhao JV. Exploring the pathways linking fasting insulin to coronary artery disease: a proteome-wide Mendelian randomization study. BMC Med 2025; 23:321. [PMID: 40442727 PMCID: PMC12124044 DOI: 10.1186/s12916-025-04127-6] [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: 02/21/2025] [Accepted: 05/13/2025] [Indexed: 06/02/2025] Open
Abstract
BACKGROUND Insulin is known to be associated with a higher risk of coronary artery disease (CAD), but molecular mechanisms remain unclear. This study aimed to explore protein-mediated pathways linking fasting insulin to CAD using Mendelian randomization (MR). METHODS This MR study examined the association between fasting insulin and CAD using genome-wide association study (GWAS) data from MAGIC and CARDIoGRAMplusC4D. To investigate underlying mechanisms, a two-step proteome-wide MR analysis was conducted. First, associations of fasting insulin with 2940 circulating proteins were assessed using GWAS of proteomics from UKB-PPP. Proteins affected by insulin were then analyzed for their association with CAD risk. Proteins selected in both steps were considered as potential mediators. Sensitivity analyses to test whether associations are robust to pleiotropy and replication using other GWAS data, including GWAS of proteomics from deCODE and GWAS of CAD from FinnGen Biobank, were performed. RESULTS Genetically predicted insulin was associated with a higher risk of CAD (odds ratio 1.79, 95% confidence interval 1.34 to 2.40). At a false discovery rate of 0.05, insulin affected 355 proteins, ten of which were both increased by insulin and linked to a higher risk of CAD. After sensitivity and replication analyses, PLA2G7, GZMA, LDLR, AGRP, and HHEX were identified as reliable mediators. Mediation analyses using non-pleiotropic instruments showed that PLA2G7, GZMA, LDLR, and AGRP explained 19.50%, 6.91%, 19.31%, and 29.66% of insulin's total effect on CAD, respectively. CONCLUSIONS This study identified five protein mediators linking insulin to CAD. These proteins could be considered as potential targets to mitigate insulin-related cardiovascular risk, providing novel insights for drug repurposing.
Collapse
Affiliation(s)
- Xin Huang
- School of Public Health, Li Ka Shing, Faculty of Medicine, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, China
| | - Jie V Zhao
- School of Public Health, Li Ka Shing, Faculty of Medicine, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, China.
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
5
|
Berthold N, MacDermod CM, Thornton LM, Parker R, Morales SAC, Hog L, Kennedy HL, Guintivano J, Sullivan PF, Crowley JJ, Johnson JS, Birgegård A, Fundín BT, Frans E, Xu J, Ngāti Pūkenga MP, Miller AL, Aguilar MV, Barakat S, Abdulkadir M, White JP, Larsen JT, Trujillo E, Winterman B, Zhang R, Lawson R, Wonderlich S, Wonderlich J, Schaefer LM, Mehler PS, Oakes J, Foster M, Gaudiani J, Vacuán ETC, Compte EJ, Petersen LV, Yilmaz Z, Micali N, Jordan J, Kennedy MA, Maguire S, Huckins LM, Lu Y, Dinkler L, Martin NG, Bulik CM. The Eating Disorders Genetics Initiative 2 (EDGI2): study protocol. BMC Psychiatry 2025; 25:532. [PMID: 40419993 PMCID: PMC12105188 DOI: 10.1186/s12888-025-06777-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Accepted: 03/26/2025] [Indexed: 05/28/2025] Open
Abstract
BACKGROUND The Eating Disorders Genetics Initiative 2 (EDGI2) is designed to explore the role of genes and environment in anorexia nervosa, bulimia nervosa, binge-eating disorder, and avoidant/restrictive food intake disorder (ARFID) with a focus on broad population representation and severe and/or longstanding illness. METHODS A total of 20,000 new participants (18,700 cases and 1,300 controls) will be ascertained from the United States (US), Mexico (MX), Australia (AU), Aotearoa New Zealand (NZ), Sweden (SE), and Denmark (DK). Comprehensive phenotyping and genotyping will be performed for participants in US, MX, AU, NZ, and SE using the EDGI2 questionnaire battery and participant saliva samples. In DK, case identification and genotyping will be through the National Patient Register and bloodspots archived near birth. Case-control and case-case genome-wide association studies will be conducted within EDGI2 and enhanced via meta-analysis with external data from the Eating Disorders Working Group of the Psychiatric Genomics Consortium (PGC-ED). Additional analyses will explore genetic correlations between eating disorders (EDs) and other psychiatric and metabolic traits, calculate polygenic risk scores (PRS), and leverage functional biology to evaluate clinical outcomes. Moreover, analyzing PRS for patient stratification and linking identified risk loci to clinically relevant phenotypes highlight the potential of EDGI2 for clinical translation. DISCUSSION EDGI2 is a global expansion of the EDGI study to increase sample size, increase participant representation across multiple ancestral backgrounds, and to include ARFID. ED genetics research has historically lagged behind other psychiatric disorders, and EDGI2 is designed to rapidly advance the study of the genetics of the major EDs. Exploring EDs at both the diagnostic level and the symptom level will provide an unprecedented look at the genetic architecture underlying EDs. TRIAL REGISTRATION EDGI2 is a registered clinical trial: clinicaltrials.gov NCT06594913. https://clinicaltrials.gov/study/NCT06594913 (posted September 19, 2024).
Collapse
Affiliation(s)
- Natasha Berthold
- Department of Psychiatry, University of North Carolina at Chapel Hill, #7160, 101 Manning Drive, Chapel Hill, CBNC, 27599 - 7160, USA
- School of Human Sciences, University of Western Australia, Crawley, WA, 6009, Australia
- Perron Research Institute, Nedlands, WA, 6009, Australia
| | - Casey M MacDermod
- Department of Psychiatry, University of North Carolina at Chapel Hill, #7160, 101 Manning Drive, Chapel Hill, CBNC, 27599 - 7160, USA
| | - Laura M Thornton
- Department of Psychiatry, University of North Carolina at Chapel Hill, #7160, 101 Manning Drive, Chapel Hill, CBNC, 27599 - 7160, USA
| | - Richard Parker
- QIMR Berghofer Medical Research Institute, Royal Brisbane Hospital, Locked Bag 2000, Brisbane, QLD, 4029, Australia
| | - Shantal Anid Cortés Morales
- Research Department, Comenzar de Nuevo, Monterrey, Mexico
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud - , Monterrey, Mexico
| | - Liv Hog
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Hannah L Kennedy
- Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Jerry Guintivano
- Department of Psychiatry, University of North Carolina at Chapel Hill, #7160, 101 Manning Drive, Chapel Hill, CBNC, 27599 - 7160, USA
| | - Patrick F Sullivan
- Department of Psychiatry, University of North Carolina at Chapel Hill, #7160, 101 Manning Drive, Chapel Hill, CBNC, 27599 - 7160, USA
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - James J Crowley
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 171 77, Stockholm, Sweden
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Jessica S Johnson
- Department of Psychiatry, University of North Carolina at Chapel Hill, #7160, 101 Manning Drive, Chapel Hill, CBNC, 27599 - 7160, USA
| | - Andreas Birgegård
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Bengt T Fundín
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Emma Frans
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Jiayi Xu
- Division of Molecular Psychiatry, Department of Psychiatry, Yale University, New Haven, CT, USA
| | | | - Allison L Miller
- Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Mariana Valdez Aguilar
- Department of Psychiatry, University of North Carolina at Chapel Hill, #7160, 101 Manning Drive, Chapel Hill, CBNC, 27599 - 7160, USA
| | - Sarah Barakat
- Insideout Institute for Eating Disorders, The University of Sydney, Sydney, Australia
| | - Mohamed Abdulkadir
- Department of Public Health, National Centre for Register-Based Research, Aarhus University, Aarhus, Denmark
| | - Jennifer P White
- Department of Psychiatry, University of North Carolina at Chapel Hill, #7160, 101 Manning Drive, Chapel Hill, CBNC, 27599 - 7160, USA
- Department of Psychology, University of Albany, State University of New York, Albany, NY, USA
| | - Janne T Larsen
- Department of Public Health, National Centre for Register-Based Research, Aarhus University, Aarhus, Denmark
| | - Elsie Trujillo
- Research Department, Comenzar de Nuevo, Monterrey, Mexico
| | | | - Ruyue Zhang
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Rachel Lawson
- South Island Eating Disorders Service, Health NZ Te Whatu Ora, Christchurch, New Zealand
| | - Stephen Wonderlich
- Center for Biobehavioral Research, Sanford Health, Fargo, ND, USA
- Department of Psychiatry and Behavioral Science, University of North Dakota School of Medicine and Health Sciences, Fargo, ND, USA
| | | | | | - Philip S Mehler
- ACUTE Center for Eating Disorders and Severe Malnutrition, Denver Health and Hospital Authority, Denver, CO, USA
| | - Judy Oakes
- ACUTE Center for Eating Disorders and Severe Malnutrition, Denver Health and Hospital Authority, Denver, CO, USA
| | - Marina Foster
- ACUTE Center for Eating Disorders and Severe Malnutrition, Denver Health and Hospital Authority, Denver, CO, USA
| | | | - Eva Trujillo Chi Vacuán
- Research Department, Comenzar de Nuevo, Monterrey, Mexico
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud - , Monterrey, Mexico
| | - Emilio J Compte
- Research Department, Comenzar de Nuevo, Monterrey, Mexico
- Eating Behavior Research Center, School of Psychology, Universidad Adolfo Ibáñez, Santiago, Chile
| | - Liselotte V Petersen
- Department of Public Health, National Centre for Register-Based Research, Aarhus University, Aarhus, Denmark
| | - Zeynep Yilmaz
- Department of Psychiatry, University of North Carolina at Chapel Hill, #7160, 101 Manning Drive, Chapel Hill, CBNC, 27599 - 7160, USA
- Department of Public Health, National Centre for Register-Based Research, Aarhus University, Aarhus, Denmark
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Nadia Micali
- Center for Eating and Feeding Disorders Research, Mental Health Services of the Capital Region of Denmark, Psychiatric Centre Ballerup, Copenhagen, Denmark
- Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Jennifer Jordan
- Department of Psychological Medicine, University of Otago, Christchurch, New Zealand
- Health NZ - Te Whatu Ora, Christchurch, New Zealand
| | - Martin A Kennedy
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud - , Monterrey, Mexico
| | - Sarah Maguire
- Insideout Institute for Eating Disorders, The University of Sydney, Sydney, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Laura M Huckins
- Division of Molecular Psychiatry, Department of Psychiatry, Yale University, New Haven, CT, USA
| | - Yi Lu
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Lisa Dinkler
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Nicholas G Martin
- QIMR Berghofer Medical Research Institute, Royal Brisbane Hospital, Locked Bag 2000, Brisbane, QLD, 4029, Australia
| | - Cynthia M Bulik
- Department of Psychiatry, University of North Carolina at Chapel Hill, #7160, 101 Manning Drive, Chapel Hill, CBNC, 27599 - 7160, USA.
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 171 77, Stockholm, Sweden.
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
6
|
Yan W, Kong L, He T, Guo G, Zhu Q, Xi X, Fang M. Association between metabolic syndrome and low back pain: a two-sample Mendelian randomization study. Sci Rep 2025; 15:17686. [PMID: 40399540 PMCID: PMC12095482 DOI: 10.1038/s41598-025-02630-7] [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: 11/05/2024] [Accepted: 05/14/2025] [Indexed: 05/23/2025] Open
Abstract
This study uses two-sample MR analysis with GWAS summary statistics to evaluate the causal relationship between metabolic syndrome and low back pain. A two-sample Mendelian randomization analysis used GWAS summary statistics for low back pain from the FinnGen database and metabolic syndrome data, including waist circumference, hypertension, fasting blood glucose, HDL cholesterol, and triglyceride levels. Various methods like inverse variance weighted, MR-Egger, weighted median, and mode assessed the causal relationship, with sensitivity analyses addressing heterogeneity and pleiotropy. Our analysis found a statistically significant causal association between essential hypertension (OR 2.38, 95% CI 1.42-3.96; Padj = 0.002), metabolic syndrome (OR 1.05, 95% CI 1.01-1.10; Padj = 0.023) and waist circumference (OR 49, 95% CI 1.32-1.68; Padj < 0.001) and low back pain (OR 1.41, 95% CI 1.30-1.53, Padj < 0.001). In contrast, fasting blood glucose (FBG), HDL cholesterol, and triglycerides showed no significant associations with low back pain across all MR methods. The results of sensitivity analyses indicated that the heterogeneity and pleiotropy were unlikely to disturb the causal estimate. Our study indicates that increased essential hypertension, metabolic syndrome and waist circumference is causally associated with a higher risk of low back pain. Interventions targeting metabolic syndrome components, particularly blood pressure control and weight management, could help reduce the risk of low back pain. Further research is needed to explore the underlying biological pathways linking these metabolic factors to low back pain.
Collapse
Affiliation(s)
- Wei Yan
- Department of Clinical Medicine School, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Orthopaedics, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lingjun Kong
- Department of Tuina, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tianxiang He
- Department of Tuina, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guangxin Guo
- Department of Tuina, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- School of Acupuncture-Moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Qingguang Zhu
- Yue Yang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
- Institute of Tuina, Shanghai Institute of Traditional Chinese Medicine, Shanghai, China.
| | - Xiaobing Xi
- Department of Orthopaedics, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Min Fang
- Department of Tuina, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
- Institute of Tuina, Shanghai Institute of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
7
|
Lin MR, Tsai CL, Liao CS, Wei CY, Chou WH, Hsiao TH, Chang WC. Exploring the genomic and transcriptomic profiles of glycemic traits and drug repurposing. J Biomed Sci 2025; 32:50. [PMID: 40399988 PMCID: PMC12096723 DOI: 10.1186/s12929-025-01137-7] [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: 08/29/2024] [Accepted: 03/25/2025] [Indexed: 05/23/2025] Open
Abstract
BACKGROUND Type 2 diabetes is an increasingly prevalent metabolic disorder with moderate to high heritability. Glycemic indices are crucial for diagnosing and monitoring the disease. Previous genome-wide association study (GWAS) have identified several risk loci associated with type 2 diabetes, but data from the Taiwanese population remain relatively sparse and primarily focus on type 2 diabetes status rather than glycemic trait levels. METHODS We conducted a comprehensive genome-wide meta-analysis to explore the genetics of glycemic traits. The study incorporated a community-based cohort of 145,468 individuals and a hospital-based cohort of 35,395 individuals. The study integrated genetics, transcriptomics, biological pathway analyses, polygenic risk score calculation, and drug repurposing for type 2 diabetes. RESULTS This study assessed hemoglobin A1c and fasting glucose levels, validating known loci (FN3K, SPC25, MTNR1B, and FOXA2) and discovering new genes, including MAEA and PRC1. Additionally, we found that diabetes, blood lipids, and liver- and kidney-related traits share genetic foundations with glycemic traits. A higher PRS was associated with an increased risk of type 2 diabetes. Finally, eight repurposed drugs were identified with evidence to regulate blood glucose levels, offering new avenues for the management and treatment of type 2 diabetes. CONCLUSIONS This research illuminates the unique genetic landscape of glucose regulation in Taiwanese Han population, providing valuable insights to guide future treatment strategies for type 2 diabetes.
Collapse
Affiliation(s)
- Min-Rou Lin
- Department of Clinical Pharmacy, School of Pharmacy, Taipei Medical University, Taipei, 110, Taiwan
| | - Cheng-Lin Tsai
- Department of Clinical Pharmacy, School of Pharmacy, Taipei Medical University, Taipei, 110, Taiwan
| | - Cai-Sian Liao
- Bioinformatics Program, Institute of Statistical Science, Taiwan International Graduate Program, Academia Sinica, Taipei, 110, Taiwan
- Bioinformatics Program, Taiwan International Graduate Program, National Taiwan University, Taipei, 110, Taiwan
| | - Chun-Yu Wei
- Department of Clinical Pharmacy, School of Pharmacy, Taipei Medical University, Taipei, 110, Taiwan
- Core Laboratory of Neoantigen Analysis for Personalized Cancer Vaccine, Office of R&D, Taipei Medical University, Taipei, 110, Taiwan
| | - Wan-Hsuan Chou
- Department of Clinical Pharmacy, School of Pharmacy, Taipei Medical University, Taipei, 110, Taiwan
| | - Tzu-Hung Hsiao
- Department of Medical Research, Taichung Veterans General Hospital, 1650 Taiwan Boulevard Sect. 4, Taichung, 407219, Taiwan.
- Department of Public Health, Fu Jen Catholic University, New Taipei City, 242, Taiwan.
- Institute of Genomics and Bioinformatics, National Chung Hsing University, Taichung, 402, Taiwan.
| | - Wei-Chiao Chang
- Department of Clinical Pharmacy, School of Pharmacy, Taipei Medical University, Taipei, 110, Taiwan.
- Master Program in Clinical Genomics and Proteomics, School of Pharmacy, Taipei Medical University, Taipei, 110, Taiwan.
- Core Laboratory of Neoantigen Analysis for Personalized Cancer Vaccine, Office of R&D, Taipei Medical University, Taipei, 110, Taiwan.
- Integrative Research Center in Critical Care, Wan Fang Hospital, Taipei Medical University, Taipei, 116, Taiwan.
- Department of Pharmacy, Wan Fang Hospital, Taipei Medical University, Taipei, 116, Taiwan.
- Department of Pharmacology, National Defense Medical Center, Taipei, 114, Taiwan.
| |
Collapse
|
8
|
Metz S, Belanich JR, Claussnitzer M, Kilpeläinen TO. Variant-to-function approaches for adipose tissue: Insights into cardiometabolic disorders. CELL GENOMICS 2025; 5:100844. [PMID: 40185091 DOI: 10.1016/j.xgen.2025.100844] [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: 10/31/2024] [Revised: 02/14/2025] [Accepted: 03/12/2025] [Indexed: 04/07/2025]
Abstract
Genome-wide association studies (GWASs) have identified thousands of genetic loci associated with cardiometabolic disorders. However, the functional interpretation of these loci remains a daunting challenge. This is particularly true for adipose tissue, a critical organ in systemic metabolism and the pathogenesis of various cardiometabolic diseases. We discuss how variant-to-function (V2F) approaches are used to elucidate the mechanisms by which GWAS loci increase the risk of cardiometabolic disorders by directly influencing adipose tissue. We outline GWAS traits most likely to harbor adipose-related variants and summarize tools to pinpoint the putative causal variants, genes, and cell types for the associated loci. We explain how large-scale perturbation experiments, coupled with imaging and multi-omics, can be used to screen variants' effects on cellular phenotypes and how these phenotypes can be tied to physiological mechanisms. Lastly, we discuss the challenges and opportunities that lie ahead for V2F research and propose a roadmap for future studies.
Collapse
Affiliation(s)
- Sophia Metz
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark; The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| | - Jonathan Robert Belanich
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark; The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Melina Claussnitzer
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Center for Genomic Medicine, Endocrine Division, Massachusetts General Hospital, Harvard Medical School, Cambridge, MA 02142, USA
| | - Tuomas Oskari Kilpeläinen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark; The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| |
Collapse
|
9
|
Zhang Z, Xie Y, Bu Z, Xiang Y, Sheng W, Cao Y, Lian L, Zhang L, Qian W, Ji G. Genetically proxied glucokinase activation and risk of diabetic complications: Insights from phenome-wide and multi-omics mendelian randomization. Diabetes Res Clin Pract 2025; 225:112246. [PMID: 40374125 DOI: 10.1016/j.diabres.2025.112246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 03/31/2025] [Accepted: 05/12/2025] [Indexed: 05/17/2025]
Abstract
AIMS This study aims to assess the benefits and adverse effects of long-term glucokinase (GK) activation from a genetic perspective. METHODS We identified genetic variants in the GCK gene associated with glycated hemoglobin (HbA1c) levels from a genome-wide association study (GWAS) involving 146,806 individuals, which served as proxies for glucokinase activation. To assess the effects and potential pathways of GK activation on a range of diabetic complications and safety outcomes, we integrated drug-target Mendelian randomization (MR), lipidome-wide and proteome-wide MR, phenome-wide MR, and colocalization analyses. RESULTS Genetically proxied GK activation was associated with reduced risks of several predefined diabetic complications, including cardiovascular diseases, stroke and diabetic retinopathy. No kidney-related benefits were observed. Safety analysis revealed a relationship between GK activation and elevated AST levels, while impaired interaction between GK and glucokinase regulatory protein (GKRP) was associated with dyslipidemia, increased liver fat content, AST, systolic blood pressure, and uric acid. Phenome-wide MR suggested that GK activation may have potential benefits for lung function and fluid intelligence score. CONCLUSIONS Our genetic evidence supports GK as a promising target for reducing the risk of specific diabetic complications. These findings require further validation through cohort studies and randomized controlled trials in patients with diabetes.
Collapse
Affiliation(s)
- Ziqi Zhang
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yanxiao Xie
- Department of Respiratory Medicine, Dongguan Hospital of Traditional Chinese Medicine, Dongguan, Guangdong, China; The Ninth Clinical Medical College, Guangzhou University of Chinese Medicine, Dongguan, Guangdong, China
| | - Zhenlin Bu
- Foshan Hospital of Traditional Chinese Medicine, Foshan, Guangdong, China; The Eighth Clinical Medical College, Guangzhou University of Chinese Medicine, Foshan, Guangdong, China
| | - Yingying Xiang
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wei Sheng
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ying Cao
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - LeShen Lian
- Department of Respiratory Medicine, Dongguan Hospital of Traditional Chinese Medicine, Dongguan, Guangdong, China; The Ninth Clinical Medical College, Guangzhou University of Chinese Medicine, Dongguan, Guangdong, China
| | - Li Zhang
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China; State Key Laboratory of Integration and Innovation of Classical Formula and Modern Chinese Medicine, Shanghai, China
| | - Wei Qian
- School of Public Health, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Guang Ji
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China; State Key Laboratory of Integration and Innovation of Classical Formula and Modern Chinese Medicine, Shanghai, China.
| |
Collapse
|
10
|
Myserlis EP, Georgakis MK, Parodi L, Mayerhofer E, Omarov M, Rosand J, Banerjee C, Anderson CD. A Beneficial Role for Gluteofemoral Adipose Tissue in Cerebrovascular Disease: Causal Pathway and Mediation Analysis. Neurology 2025; 104:e213573. [PMID: 40228186 DOI: 10.1212/wnl.0000000000213573] [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: 09/05/2024] [Accepted: 02/18/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND AND OBJECTIVES Previous studies have shown that increased body fat is associated with stroke risk, with evidence suggesting that body fat distribution, rather than total body fat, exerts a more prominent role in cerebrovascular risk prediction. In this study, we explore causal associations between body mass index (BMI)-independent adipose tissue distribution profiles and cerebrovascular disease (CVD) risk, aiming to refine the association between body fat distribution and stroke. METHODS We selected variants associated with BMI-independent visceral adipose tissue (VAT), abdominal subcutaneous adipose tissue (ASAT), and gluteofemoral adipose tissue (GFAT) volumes in UK Biobank, and performed univariable and multivariable Mendelian randomization (MR) analyses with ischemic stroke (IS) and subtypes (large artery stroke [LAS], cardioembolic stroke [CES], and small vessel stroke [SVS]). We used coronary artery disease (CAD), carotid intima media thickness (cIMT), and MRI-confirmed lacunar stroke as positive controls. We explored the mediatory role of common cardiovascular (systolic blood pressure, diabetes, and low-density lipoprotein), insulin resistance, inflammatory (C-reactive protein), and adipose tissue-specific (adiponectin, leptin) factors by performing 2-step mediation MR analyses. Estimates were expressed per standard deviation increase in adjusted adipose tissue volume. RESULTS Genetic predisposition to higher GFAT volume was associated with lower risk of IS (odds ratio [OR] 0.92, 95% CI 0.86-0.98), LAS (OR 0.80, 95% CI 0.66-0.96), and SVS (OR 0.77, 95% CI 0.67-0.88), but not CES, consistent in multivariable analyses. Genetic predisposition to higher GFAT volume was also associated with lower risk of CAD (OR 0.82, 95% CI 0.76-0.88), lacunar stroke (OR 0.78, 95% CI 0.67-0.92), and mean cIMT (β = -0.073, 95% CI -0.114 to -0.031). Associations were largely consistent in sensitivity analyses. No association was observed between genetic predisposition to ASAT or VAT and IS risk. Although common vascular risk factors were the predominant mediators in the GFAT-CVD axis, adiponectin and leptin mediated a proportion of IS and CAD risk (∼15% (1.8%-57%) and ∼4.6% (0.8%-13.5%) mediated by adiponectin, respectively). DISCUSSION This study supports a protective role of gluteofemorally distributed fat volume in CVD risk. Although this role is predominantly mediated by common vascular risk factor modification, adipose tissue-specific factors may exert a mediatory effect, suggesting a possible novel target for attenuating adiposity-related CVD risk.
Collapse
Affiliation(s)
| | - Marios K Georgakis
- Institute for Stroke and Dementia Research (ISD), Ludwig-Maximilians-University (LMU) Hospital, Munich, Germany
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Boston, MA
| | - Livia Parodi
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Boston, MA
- Department of Neurology, Brigham and Women's Hospital, Boston, MA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston
| | - Ernst Mayerhofer
- Center for Genomic Medicine, Massachusetts General Hospital, Boston
| | - Murad Omarov
- Institute for Stroke and Dementia Research, University Hospital of LMU Munich, Germany
| | - Jonathan Rosand
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Boston, MA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston
- Department of Neurology, Massachusetts General Hospital, Boston; and
- McCance Center for Brain Health, Massachusetts General Hospital, Boston
| | - Chirantan Banerjee
- Department of Neurology, Medical University of South Carolina, Charleston
| | - Christopher D Anderson
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Boston, MA
- Department of Neurology, Brigham and Women's Hospital, Boston, MA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston
- McCance Center for Brain Health, Massachusetts General Hospital, Boston
| |
Collapse
|
11
|
Islam MS. Ryanodine Receptors in Islet Cell Function: Calcium Signaling, Hormone Secretion, and Diabetes. Cells 2025; 14:690. [PMID: 40422193 DOI: 10.3390/cells14100690] [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: 03/28/2025] [Revised: 04/29/2025] [Accepted: 05/09/2025] [Indexed: 05/28/2025] Open
Abstract
Ryanodine receptors (RyRs) are large intracellular Ca2+ release channels primarily found in muscle and nerve cells and also present at low levels in pancreatic islet endocrine cells. This review examines the role of RyRs in islet cell function, focusing on calcium signaling and hormone secretion, while addressing the ongoing debate regarding their significance due to their limited expression. We explore conflicting experimental results and their potential causes, synthesizing current knowledge on RyR isoforms in islet cells, particularly in beta and delta cells. The review discusses how RyR-mediated calcium-induced calcium release enhances, rather than drives, glucose-stimulated insulin secretion. We examine the phosphorylation-dependent regulation of beta-cell RyRs, the concept of "leaky ryanodine receptors", and the roles of RyRs in endoplasmic reticulum stress, apoptosis, store-operated calcium entry, and beta-cell electrical activity. The relationship between RyR dysfunction and the development of impaired insulin secretion in diabetes is assessed, noting their limited role in human diabetes pathogenesis given the disease's polygenic nature. We highlight the established role of RyR-mediated CICR in the mechanism of action of common type 2 diabetes treatments, such as glucagon-like peptide-1, which enhances insulin secretion. By integrating findings from electrophysiological, molecular, and clinical studies, this review provides a balanced perspective on RyRs in islet cell physiology and pathology, emphasizing their significance in both normal insulin secretion and current diabetes therapies.
Collapse
Affiliation(s)
- Md Shahidul Islam
- Karolinska Institutet, Department of Clinical Sciences and Education, Södersjukhuset, Research Center, 5th Floor, SE-118 83 Stockholm, Sweden
- Department of Internal Medicine, Uppsala University Hospital, SE-751 85 Uppsala, Sweden
| |
Collapse
|
12
|
Ying X, Wu Q, Li X, Bi Y, Gao L, Yu S, Xu X, Li X, Wang Y, Hua R. Causal Associations Between Pre-Pregnancy Diabetes Mellitus and Pre-Eclampsia Risk: Insights from a Mendelian Randomization Study. Healthcare (Basel) 2025; 13:1085. [PMID: 40361863 PMCID: PMC12072006 DOI: 10.3390/healthcare13091085] [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] [Subscribe] [Scholar Register] [Received: 03/16/2025] [Revised: 04/12/2025] [Accepted: 04/30/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND AND OBJECTIVES Pre-eclampsia (PE) is a serious pregnancy complication defined by the onset of hypertension and multi-organ dysfunction occurring after 20 weeks of gestation. Studies have indicated the correlation between diabetes mellitus (DM) and PE, but the causal relationship remains unclear. MATERIALS AND METHODS The two-sample Mendelian randomization (MR) approach, including the inverse variance weighted random effects (IVW-RE) model and the traditional sensitivity model, was employed to assess the causal effects of pre-pregnancy type 1 diabetes (T1D) and type 2 diabetes (T2D) on PE using summary-level data obtained from genome-wide association studies. Additionally, diabetes-related factors, such as glycated hemoglobin (HbA1c) levels, fasting insulin levels, and body mass index (BMI), were evaluated for their potential causal effects on the risk of PE. Pleiotropy-robust and multivariable Mendelian randomization (MVMR) methods were further used because of the intricate associations among the traits. Insulin and metformin use was also assessed for their causal role in PE risk. RESULTS Our findings show that genetically predicted T1D (OR = 1.06, 95% CI: 1.03-1.09, p < 0.001), T2D (OR = 1.09, 95% CI: 1.04-1.14, p < 0.001), and BMI (OR = 1.64, 95% CI 1.49 to 1.80, p < 0.001) had causal effects on the incidence of PE, while the effects of HbA1c (OR = 0.77, 95% CI 0.59 to 1.02, p = 0.064) and fasting insulin levels (OR = 1.35, 95% CI 0.89 to 2.05, p = 0.153) on the occurrence of PE were not significant. The results were verified by MVMR analysis. Additionally, insulin use increased the risk of pre-eclampsia (OR = 1.11, 95% CI 1.05-1.17, p < 0.001). CONCLUSIONS Our findings demonstrate a causal relationship between pre-pregnancy diabetes (DM) and obesity and the risk of PE from a genetic epidemiological perspective. Adverse maternal factors, including DM and obesity prior to pregnancy, should be considered in mechanistic studies of PE. In addition, comprehensive interventions for risk factors such as pre-pregnancy DM and obesity should be emphasized in clinical practice.
Collapse
Affiliation(s)
- Xiang Ying
- Division of Fetal Medicine, Prenatal Diagnosis Department, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; (X.Y.); (X.L.); (L.G.); (S.Y.); (X.X.)
- Shanghai Key Laboratory of Embryo Original Disease, Shanghai 200030, China;
| | - Quanfeng Wu
- Department of Obstetrics, Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen 518028, China; (Q.W.); (X.L.)
- Department of Obstetrics, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen 361003, China
| | - Xiaohan Li
- Division of Fetal Medicine, Prenatal Diagnosis Department, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; (X.Y.); (X.L.); (L.G.); (S.Y.); (X.X.)
- Shanghai Key Laboratory of Embryo Original Disease, Shanghai 200030, China;
| | - Yan Bi
- Shanghai Key Laboratory of Embryo Original Disease, Shanghai 200030, China;
| | - Li Gao
- Division of Fetal Medicine, Prenatal Diagnosis Department, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; (X.Y.); (X.L.); (L.G.); (S.Y.); (X.X.)
| | - Shushu Yu
- Division of Fetal Medicine, Prenatal Diagnosis Department, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; (X.Y.); (X.L.); (L.G.); (S.Y.); (X.X.)
| | - Xiaona Xu
- Division of Fetal Medicine, Prenatal Diagnosis Department, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; (X.Y.); (X.L.); (L.G.); (S.Y.); (X.X.)
| | - Xiaotian Li
- Department of Obstetrics, Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen 518028, China; (Q.W.); (X.L.)
- Department of Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China
| | - Yanlin Wang
- Division of Fetal Medicine, Prenatal Diagnosis Department, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; (X.Y.); (X.L.); (L.G.); (S.Y.); (X.X.)
- Shanghai Key Laboratory of Embryo Original Disease, Shanghai 200030, China;
| | - Renyi Hua
- Division of Fetal Medicine, Prenatal Diagnosis Department, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; (X.Y.); (X.L.); (L.G.); (S.Y.); (X.X.)
| |
Collapse
|
13
|
Bao Y, Chen J, Han X, He Y, Yang T, Shi X, Chen J, Gu L, Wang S, Xie L, Wang H, Wang L. Calbindin 2 as a Novel Biomarker and Therapeutic Target for Abdominal Aortic Aneurysm: Integrative Analysis of Human Proteomes and Genetics. J Am Heart Assoc 2025; 14:e039195. [PMID: 40314374 DOI: 10.1161/jaha.124.039195] [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: 10/01/2024] [Accepted: 04/08/2025] [Indexed: 05/03/2025]
Abstract
BACKGROUND Abdominal aortic aneurysm (AAA) is a clinical life-threatening issue. No pharmacological treatments are currently approved for the prevention and treatment of AAA. Therefore, identifying novel biomarkers and therapeutic targets is crucial for improving AAA management and outcomes. METHODS To identify plasma proteins with potential causal effects on AAA, we integrated genetic evidence from proteome-wide Mendelian randomization, genetic correlation, and colocalization analysis. The role of identified proteins in AAA was further explored through the phenome-wide association study and mediation analysis. Multiomics data analysis, including bulk RNA sequencing, single-cell/single-nucleus RNA sequencing, and spatial transcriptomics, was employed to characterize the expression patterns of these proteins. Experimental validation was performed using an AAA model in apolipoprotein E-deficient mice infused with angiotensin II. Druggability analysis was conducted to identify drug candidates, which were tested in preclinical mouse models. RESULTS CALB2 (calbindin 2) was identified as having a causal effect on AAA and may influence the progression of AAA through the regulation of lipid metabolism. Multiomics analysis revealed that CALB2 is predominantly expressed in the mesothelial cells of adipose tissues. Inhibition of CALB2 in an AAA mouse model alleviated AAA progression. Druggability analysis identified lenalidomide and genistein as potential therapeutic candidates, and experiments confirmed their efficacy in preventing AAA development. CONCLUSIONS This study identifies CALB2 as being associated with an increased risk of AAA and suggests that i might be a novel biomarker and therapeutic molecule for AAA management. Lenalidomide and genistein hold promising potential as treatments for patients with AAA.
Collapse
Affiliation(s)
- Yulin Bao
- Department of Cardiology The First Affiliated Hospital with Nanjing Medical University Nanjing Jiangsu China
| | - Jiayi Chen
- Department of Cardiology The First Affiliated Hospital with Nanjing Medical University Nanjing Jiangsu China
| | - Xudong Han
- Department of Cardiology The First Affiliated Hospital with Nanjing Medical University Nanjing Jiangsu China
| | - Ye He
- Department of Cardiology The First Affiliated Hospital with Nanjing Medical University Nanjing Jiangsu China
| | - Tongtong Yang
- Department of Cardiology The First Affiliated Hospital with Nanjing Medical University Nanjing Jiangsu China
| | - Xinying Shi
- Department of Cardiology The First Affiliated Hospital with Nanjing Medical University Nanjing Jiangsu China
| | - Jiawen Chen
- Department of Cardiology The First Affiliated Hospital with Nanjing Medical University Nanjing Jiangsu China
| | - Lingfeng Gu
- Department of Cardiology The First Affiliated Hospital with Nanjing Medical University Nanjing Jiangsu China
| | - Sibo Wang
- Department of Cardiology The First Affiliated Hospital with Nanjing Medical University Nanjing Jiangsu China
| | - Liping Xie
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine Nanjing Medical University Nanjing Jiangsu China
| | - Hao Wang
- Department of Cardiology The First Affiliated Hospital with Nanjing Medical University Nanjing Jiangsu China
| | - Liansheng Wang
- Department of Cardiology The First Affiliated Hospital with Nanjing Medical University Nanjing Jiangsu China
| |
Collapse
|
14
|
Suleman S, Ängquist L, Linneberg A, Hansen T, Grarup N. Exploring the genetic intersection between obesity-associated genetic variants and insulin sensitivity indices. Sci Rep 2025; 15:15761. [PMID: 40328835 PMCID: PMC12056085 DOI: 10.1038/s41598-025-98507-w] [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: 10/20/2024] [Accepted: 04/11/2025] [Indexed: 05/08/2025] Open
Abstract
Insulin sensitivity (IS) is a key determinant of metabolic health and may share genetic factors with obesity-related traits. Previous large-scale genetic studies have identified variants associated with IS as well as obesity related traits like body mass index (BMI) and waist-to-hip ratio (WHR). Notably, many of these associations are shared across traits, indicating a potential genetic overlap. However, the genetic intersection between IS and obesity-related traits remains underexplored. To explore this gap, we investigated associations between six IS indices, including fasting and post-glucose load measures, and genetic variants linked to BMI and WHR to determine their influence on IS and related cardiometabolic traits. To achieve this, we calculated six IS indices using fasting and oral glucose tolerance test (OGTT) data from 5,007 non-diabetic individuals, grouping them into fasting, OGTT0,120, and OGTT0,30,120 categories. A total of 678 BMI-associated and 265 WHR-associated genetic variants were analysed using linear regression, adjusting for age and sex, with sex-specific analyses for WHR. Analyses were conducted with and without BMI adjustments and corrected for multiple testing (padj). Additionally, we explored the relationship between IS-linked variants and their associations with type 2 diabetes (T2D), coronary artery disease (CAD) and stroke. Among the 678 BMI-associated variants, 100 showed nominal associations (p < 0.05) with at least one IS index; and 20 remained significant after multiple testing correction (padj < 0.05) when not adjusting for BMI. After adjusting for BMI, 70 variants retained nominal associations, and six remained significant (padj < 0.05). In sex-specific analyses of the 265 WHR-associated variants, 12 variants were associated in females when adjusted for BMI, whereas no significant associations were observed in males. Furthermore, BMI- and WHR-associated variants linked to decreased IS, such as those in FTO and VPS13C loci, were also associated with increased T2D and stroke risk, whereas IS-increasing variants, including those in VPS13C and PPARG, were linked to lower T2D and stroke risk, with some, like THADA, showing opposing effects on CAD. This study offers insights into genetic variants that influence both IS and obesity-related traits, revealing BMI- and WHR-associated variants with both positive and negative effects on IS and their potential impact on cardiometabolic health.
Collapse
Affiliation(s)
- Sufyan Suleman
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Biomedicine, Human Genetics, Aarhus University, Aarhus, 8000, Denmark
| | - Lars Ängquist
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Allan Linneberg
- Center for Clinical Research and Prevention, Copenhagen University Hospital-Bispebjerg and Frederiksberg, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Niels Grarup
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
15
|
Gu Y, Zheng H, Wang P, Liu Y, Guo X, Wei Y, Yang Z, Cheng S, Chen Y, Hu L, Chen X, Zhang Q, Chen G, Wei F, Zhen J, Liu S. Genetic architecture and risk prediction of gestational diabetes mellitus in Chinese pregnancies. Nat Commun 2025; 16:4178. [PMID: 40325049 PMCID: PMC12053562 DOI: 10.1038/s41467-025-59442-6] [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: 09/11/2024] [Accepted: 04/24/2025] [Indexed: 05/07/2025] Open
Abstract
Gestational diabetes mellitus, a heritable metabolic disorder and the most common pregnancy-related condition, remains understudied regarding its genetic architecture and its potential for early prediction using genetic data. Here we conducted genome-wide association studies on 116,144 Chinese pregnancies, leveraging their non-invasive prenatal test sequencing data and detailed prenatal records. We identified 13 novel loci for gestational diabetes mellitus and 111 for five glycemic traits, with minor allele frequencies of 0.01-0.5 and absolute effect sizes of 0.03-0.62. Approximately 50% of these loci were specific to gestational diabetes mellitus and gestational glycemic levels, distinct from type 2 diabetes and general glycemic levels in East Asians. A machine learning model integrating polygenic risk scores and prenatal records predicted gestational diabetes mellitus before 20 weeks of gestation, achieving an area under the receiver operating characteristic curve of 0.729 and an accuracy of 0.835. Shapley values highlighted polygenic risk scores as key contributors. This model offers a cost-effective strategy for early gestational diabetes mellitus prediction using clinical non-invasive prenatal test.
Collapse
Affiliation(s)
- Yuqin Gu
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Hao Zheng
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
- Central Laboratory, Shenzhen Baoan Women's and Children's Hospital, Shenzhen, Guangdong, 518102, China
| | - Piao Wang
- The Genetics Laboratory, Longgang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College), Shenzhen, Guangdong, 518172, China
| | - Yanhong Liu
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Xinxin Guo
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Yuandan Wei
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Zijing Yang
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Shiyao Cheng
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Yanchao Chen
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Liang Hu
- The Genetics Laboratory, Longgang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College), Shenzhen, Guangdong, 518172, China
| | - Xiaohang Chen
- The Genetics Laboratory, Longgang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College), Shenzhen, Guangdong, 518172, China
| | - Quanfu Zhang
- Central Laboratory, Shenzhen Baoan Women's and Children's Hospital, Shenzhen, Guangdong, 518102, China
| | - Guobo Chen
- Department of Genetic and Genomic Medicine, Center for Productive Medicine, Clinical Research Institute, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Fengxiang Wei
- The Genetics Laboratory, Longgang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College), Shenzhen, Guangdong, 518172, China.
| | - Jianxin Zhen
- Central Laboratory, Shenzhen Baoan Women's and Children's Hospital, Shenzhen, Guangdong, 518102, China.
| | - Siyang Liu
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China.
- Shenzhen Key Laboratory of Pathogenic Microbes and Biosafety, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China.
- GuangDong Engineering Technology Research Center of Nutrition Transformation, Sun Yat-sen University, Shenzhen, 518107, Guangdong Province, China.
| |
Collapse
|
16
|
Jiang B, Wei X, Cao X, Zheng C. Insights into modifiable risk factors of retinal vascular occlusion: A Mendelian randomization study. Medicine (Baltimore) 2025; 104:e41752. [PMID: 40324241 PMCID: PMC12055163 DOI: 10.1097/md.0000000000041752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 01/11/2025] [Accepted: 02/14/2025] [Indexed: 05/07/2025] Open
Abstract
Understanding the etiological risk factors for retinal vascular occlusion (RVO) is critical for prevention and treatment. While the effects of cardiovascular events, hypertension, glaucoma, obesity and glycemic risk factors on RVO are still controversial. This study employed two-sample Mendelian randomization (MR) analysis to investigate these causal risk factors. Single-nucleotide polymorphisms (SNPs) were used as instrumental variables (IVs). Genetic instruments for hypertension, glaucoma, obesity, cardiovascular events and glycemic risk factors were obtained from published genome-wide association studies (GWASs). Summary-level data for RVO and hypertension were obtained from the FinnGen consortium. MR analysis primarily utilized the inverse variance weighted (IVW) method, with MR-Egger and weighted median as supplementary approaches. Multivariable MR (MVMR) adjusting for hypertension or glaucoma of RVO were conducted. Heterogeneity was assessed using Cochrane's Q test and I2, while MR-Egger intercept and MR-PRESSO tested horizontal pleiotropy. All MR analyses were performed within R software (4.1.3) using the R packages "TwoSampleMR" and "MR-PRESSO." Genetic instruments for hypertension and glaucoma were significantly associated with RVO risk. A one-standard deviation (SD) increase in hypertension was associated with a higher risk of RVO [OR = 1.577, 95% CI = (1.342, 1.854), P < .001], while a one-SD increase in the log odds of genetically predicted glaucoma was associated with a higher risk of RVO [OR = 1.24, 95% CI = (1.115, 1.379), P < .001]. Meanwhile, hypertension and glaucoma were still significant in multivariable MR. There was not sufficient evidence to suggest cardiovascular events and obesity were associated with RVO risk. This MR study provided genetic evidence supporting that hypertension and glaucoma were causally associated with the risk of RVO. It may help guide clinical decisions in the management of RVO patients with hypertension and glaucoma.
Collapse
Affiliation(s)
- Bingcai Jiang
- Department of Ophthalmology, Guizhou Provincial People’s Hospital, Guizhou, China
| | - Xin Wei
- Department of Ophthalmology, The People’s Hospital of Tongliang District, Chongqing, China
| | - Xiaochuan Cao
- Department of Ophthalmology, The People’s Hospital of Tongliang District, Chongqing, China
| | - Changwei Zheng
- Department of Ophthalmology, The People’s Hospital of Tongliang District, Chongqing, China
| |
Collapse
|
17
|
Fan Z, Su H, Qiao T, Shi S, Shi P, Zhang A. TEX10: A Novel Drug Target and Potential Therapeutic Direction for Sleep Apnea Syndrome. Nat Sci Sleep 2025; 17:731-746. [PMID: 40330585 PMCID: PMC12053781 DOI: 10.2147/nss.s499895] [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: 10/15/2024] [Accepted: 04/07/2025] [Indexed: 05/08/2025] Open
Abstract
Background Sleep apnea syndrome (SAS) is a prevalent sleep disorder strongly associated with obesity, metabolic dysregulation, and cardiovascular diseases. While its underlying pathophysiological mechanisms remain incompletely understood, genetic factors likely play a pivotal role in SAS pathogenesis. This study investigates the causal relationships between potential drug target genes and SAS using multiple statistical approaches, aiming to provide novel insights for targeted therapeutic development. Methods We conducted a comprehensive genetic analysis integrating multiple methodologies to investigate gene-SAS relationships. Using publicly available GWAS and eQTL databases, we performed Mendelian Randomization (MR) analysis with the inverse variance weighted (IVW) method, validated by weighted median and MR-Egger approaches. Summary-data-based MR (SMR) analysis, coupled with HEIDI testing, assessed direct gene expression-SAS associations while controlling for linkage disequilibrium (LD). Colocalization analysis evaluated the probability of shared causal variants between SNPs, gene expression, and SAS. Statistical significance was determined using Benjamini-Hochberg multiple testing correction (FDR < 0.05). Additionally, mediation analysis explored TEX10's influence on SAS through metabolic intermediates including BMI, waist circumference, and HDL cholesterol. Results We identified 18 candidate drug target genes significantly associated with SAS, with MAPKAPK3, TNXB, MPHOSPH8, and TEX10 showing consistent associations across multiple analyses. TEX10, in particular, exhibited significant associations with SAS risk in blood, cerebral cortex, hippocampus, and basal ganglia (PP.H4 > 0.9). Mediation analysis suggested that TEX10 might influence SAS risk indirectly through BMI, waist circumference, and HDL cholesterol levels. Conclusion Our study identified multiple potential therapeutic targets causally linked to SAS, with TEX10 emerging as a key candidate gene. These findings advance our understanding of SAS pathogenesis and offer promising directions for personalized diagnostics and targeted therapies.
Collapse
Affiliation(s)
- Zhitao Fan
- Department of Otorhinolaryngology, Hebei Eye Hospital, Xingtai, Hebei Province, People’s Republic of China
| | - Hui Su
- Department of Neurosurgery, Xingtai People’s Hospital, Xingtai, Hebei Province, People’s Republic of China
| | - Tong Qiao
- Department of Otorhinolaryngology, Hebei Eye Hospital, Xingtai, Hebei Province, People’s Republic of China
| | - Sunan Shi
- Department of Otorhinolaryngology, Hebei Eye Hospital, Xingtai, Hebei Province, People’s Republic of China
| | - Pengfei Shi
- Department of Ophthalmology, Hebei Eye Hospital, Xingtai, Hebei Province, People’s Republic of China
| | - Anqi Zhang
- Department of Otorhinolaryngology, Hebei Eye Hospital, Xingtai, Hebei Province, People’s Republic of China
| |
Collapse
|
18
|
Baltramonaityte V, Karhunen V, Felix JF, Penninx BWJH, Cecil CAM, Fairchild G, Milaneschi Y, Walton E. Biological pathways underlying the relationship between childhood maltreatment and Multimorbidity: A two-step, multivariable Mendelian randomisation study. Brain Behav Immun 2025; 126:59-69. [PMID: 39900145 DOI: 10.1016/j.bbi.2025.01.024] [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: 12/03/2024] [Revised: 01/29/2025] [Accepted: 01/31/2025] [Indexed: 02/05/2025] Open
Abstract
Childhood maltreatment has been associated with multimorbidity of depression, coronary artery disease and type 2 diabetes. However, the biological mechanisms underlying this association remain unclear. We employed two-step and multivariable Mendelian randomisation (MR) to understand the role of three potential biological mediating mechanisms - inflammation (92 proteins), metabolic processes (54 markers), and cortisol - in the link between childhood maltreatment liability and multimorbidity. Using summary statistics from large-scale genome-wide association studies of European ancestry for childhood maltreatment (N = 185,414) and multimorbidity (Neffective = 156,717), we tested for the presence of an indirect effect via each mediator individually. We found a potential role of metabolic pathways. Up to 11% of the effect of childhood maltreatment on multimorbidity was mediated by triglycerides (indirect effect [95% CI]: 0.018 [0.009-0.027]), 8% by glycated haemoglobin (indirect effect: 0.013 [0.003-0.023]), and up to 7% by high-density lipoprotein cholesterol (indirect effect: 0.011 [0.005-0.017]). We did not find evidence for mediation via any inflammatory protein or cortisol. Our findings shed light on the biological mechanisms linking childhood maltreatment liability to multimorbidity, highlighting the role of metabolic pathways. Future studies may explore underlying pathways via non-biological mediators (e.g., lifestyle factors) or via multiple mediators simultaneously.
Collapse
Affiliation(s)
| | - Ville Karhunen
- MRC Biostatistics Unit, University of Cambridge, United Kingdom; Research Unit of Population Health, University of Oulu, Oulu, Finland; Research Unit of Mathematical Sciences, University of Oulu, Oulu, Finland
| | - Janine F Felix
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Brenda W J H Penninx
- Department of Psychiatry, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, the Netherlands; Amsterdam Public Health, Mental Health Program, Amsterdam, the Netherlands; Amsterdam Neuroscience, Complex Trait Genetics, Amsterdam, the Netherlands
| | - Charlotte A M Cecil
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands
| | - Graeme Fairchild
- Department of Psychology, University of Bath, Bath, United Kingdom
| | - Yuri Milaneschi
- Department of Psychiatry, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, the Netherlands; Amsterdam Public Health, Mental Health Program, Amsterdam, the Netherlands; Amsterdam Neuroscience, Complex Trait Genetics, Amsterdam, the Netherlands
| | - Esther Walton
- Department of Psychology, University of Bath, Bath, United Kingdom.
| |
Collapse
|
19
|
Stener-Victorin E, Deng Q. Epigenetic inheritance of PCOS by developmental programming and germline transmission. Trends Endocrinol Metab 2025; 36:472-481. [PMID: 39732517 DOI: 10.1016/j.tem.2024.12.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 12/02/2024] [Accepted: 12/03/2024] [Indexed: 12/30/2024]
Abstract
Polycystic ovary syndrome (PCOS) is a prevalent endocrine and metabolic disorder, affecting approximately 11-13% of women of reproductive age. Women with PCOS experience a higher prevalence of infertility, pregnancy complications, and cardiometabolic disorders such as obesity, insulin resistance, and type 2 diabetes mellitus. Furthermore, psychiatric comorbidities, including depression and anxiety, significantly impact the quality of life in this population. Although obesity exacerbates these health risks, the exact etiology and pathophysiology of PCOS remain complex and only partially understood. Emerging research suggests potential transgenerational inheritance through genetic and epigenetic mechanisms, highlighting the possibility of PCOS-related risks affecting subsequent generations, including sons. This review synthesizes recent findings on PCOS inheritance patterns and underscores areas for future clinical and research exploration.
Collapse
Affiliation(s)
| | - Qiaolin Deng
- Department of Physiology and Pharmacology, Karolinska Institutet, 171 77 Stockholm, Sweden.
| |
Collapse
|
20
|
Wang S, Lenzini P, Thyagarajan B, Lee JH, Vardarajan BN, Yashin A, Miljkovic I, Daw EW, Lin SJ, Patti GJ, Brent MR, Zmuda JM, Perls TT, Christensen K, Province MA, An P. Evidence of a novel gene locus ARHGAP44 for longitudinal change in hemoglobin A1c levels among subjects without diabetes from the Long Life Family Study. Physiol Genomics 2025; 57:293-298. [PMID: 40019798 DOI: 10.1152/physiolgenomics.00137.2024] [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: 08/30/2024] [Revised: 10/24/2024] [Accepted: 02/24/2025] [Indexed: 04/01/2025] Open
Abstract
Glycated hemoglobin A1c (HbA1c) indicates average glucose levels over 3 mo and is associated with insulin resistance and type 2 diabetes (T2D). Longitudinal change in circulating HbA1c (ΔHbA1c) is also associated with aging processes, cognitive performance, and mortality. We analyzed ΔHbA1c in 1,886 nondiabetic Europeans from the Long Life Family Study (LLFS) to uncover gene loci influencing ΔHbA1c. Using growth curve modeling adjusted for multiple covariates, we derived ΔHbA1c and conducted linkage-guided sequence analysis. Our genome-wide linkage scan identified a significant locus on 17p12. In-depth analysis revealed a gene locus ARHGAP44 (rs56340929, explaining 27% of the linkage peak) that was significantly associated with ΔHbA1c. Interestingly, RNA transcription of ARHGAP44 was also significantly associated with ΔHbA1c in the LLFS, and this discovery was replicable on the gene locus level in the Framingham Offspring Study (FOS). Taking together, we successfully identified a novel gene locus ARHGAP44 for ΔHbA1c in family members without T2D. Further follow-up studies using longitudinal omics data in large independent cohorts are warranted.NEW & NOTEWORTHY HbA1c is clinically used in T2D diagnosis and monitoring. Its longitudinal change (ΔHbA1c) is associated with T2D-related aging processes and mortality. Targeted association tests under significant linkage peaks in extended families permit identification of unique gene loci. We uncovered a novel gene locus ARHGAP44 for ΔHbA1c with gene-level validations from the FOS and RNAseq data in the LLFS. The finding provides genetically informed biological insight into mechanistic inference of glycemia/HbA1c homeostasis and potential T2D pathophysiology.
Collapse
Affiliation(s)
- Siyu Wang
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Petra Lenzini
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Bharat Thyagarajan
- Department of Laboratory Medicine and Pathology, School of Medicine, University of Minnesota, Minneapolis, Minnesota, United States
| | - Joseph H Lee
- Gertrude H. Sergievsky Center and the Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Departments of Neurology and Epidemiology, Columbia University Medical Center, New York City, New York, United States
| | - Badri N Vardarajan
- Gertrude H. Sergievsky Center and the Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Departments of Neurology and Epidemiology, Columbia University Medical Center, New York City, New York, United States
| | - Anatoli Yashin
- Social Science Research Institute, Duke University, Durham, North Carolina, United States
| | - Iva Miljkovic
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - E Warwick Daw
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Shiow J Lin
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Gary J Patti
- Department of Chemistry, Washington University School of Arts & Sciences, St. Louis, Missouri, United States
| | - Michael R Brent
- Department of Computer Science & Engineering, McKelvey School of Engineering, Washington University, St. Louis, Missouri, United States
| | - Joseph M Zmuda
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Thomas T Perls
- Department of Medicine, Section of Geriatrics, Boston Medical Center, Boston University School of Medicine, Boston, Massachusetts, United States
| | - Kaare Christensen
- Unit of Epidemiology, Biostatistics, and Biodemography, Department of Public Health, Southern Denmark University, Odense, Denmark
| | - Michael A Province
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Ping An
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, United States
| |
Collapse
|
21
|
Siddiqui N, Lee B, Yi V, Farek J, Khan Z, Kalla SE, Wang Q, Walker K, Meldrim J, Kachulis C, Gatzen M, Lennon NJ, Mehtalia S, Catreux S, Mehio R, Gibbs RA, Venner E. Celeste: A cloud-based genomics infrastructure with variant-calling pipeline suited for population-scale sequencing projects. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.04.29.25326690. [PMID: 40343041 PMCID: PMC12060955 DOI: 10.1101/2025.04.29.25326690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 05/11/2025]
Abstract
Background The All of Us Research Program (All of Us) is one of the world's largest sequencing efforts that will generate genetic data for over one million individuals from diverse backgrounds. This historic megaproject will create novel research platforms that integrate an unprecedented amount of genetic data with longitudinal health information. Here, we describe the design of Celeste, a resilient, open-source cloud architecture for implementing genomics workflows that has successfully analyzed petabytes of participant genomic information for All of Us - thereby enabling other large-scale sequencing efforts with a comprehensive set of tools to power analysis. The Celeste infrastructure is tremendously scalable and has routinely processed fluctuating workloads of up to 9,000 whole-genome sequencing (WGS) samples for All of Us, monthly. It also lends itself to multiple projects. Serverless technology and container orchestration form the basis of Celeste's system for managing this volume of data. Results In 12 months of production (within a single Amazon Web Services (AWS) Region), around 200 million serverless functions and over 20 million messages coordinated the analysis of 1.8 million bioinformatics, quality control, and clinical reporting jobs. Adapting WGS analysis to clinical projects requires adaptation of variant-calling methods to enrich the reliable detection of variants with known clinical importance. Thus, we also share the process by which we tuned the variant-calling pipeline in use by the multiple genome centers supporting All of Us to maximize precision and accuracy for low fraction variant calls with clinical significance. Conclusions When combined with hardware-accelerated implementations for genomic analysis, Celeste had far-reaching, positive implications for turn-around time, dynamic scalability, security, and storage of analysis for one hundred-thousand whole-genome samples and counting. Other groups may align their sequencing workflows to this harmonized pipeline standard, included within the Celeste framework, to meet clinical requisites for population-scale sequencing efforts. Celeste is available as an Amazon Web Services (AWS) deployment in GitHub, and includes command-line parameters and software containers.
Collapse
Affiliation(s)
- Noora Siddiqui
- Prostate Cancer Clinical Trials Consortium, New York, NY. USA
| | - Breanna Lee
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX. USA
| | - Victoria Yi
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX. USA
| | - Jesse Farek
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX. USA
| | - Ziad Khan
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX. USA
| | - Sara E Kalla
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX. USA
| | - Qiaoyan Wang
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX. USA
| | - Kimberly Walker
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX. USA
| | - James Meldrim
- Broad Institute of MIT and Harvard, Cambridge, MA. USA
| | | | | | | | | | | | | | - Richard A Gibbs
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX. USA
| | - Eric Venner
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX. USA
| |
Collapse
|
22
|
Lima ACDS, Cruvinel NT, da Silva NR, Mendes MM, Duarte ACS, Coelho ASG, Vimaleswaran KS, Horst MA. Interaction Between Dietary Fiber Intake and MTNR1B rs10830963 Polymorphism on Glycemic Profiles in Young Brazilian Adults. Genes (Basel) 2025; 16:497. [PMID: 40428319 PMCID: PMC12110926 DOI: 10.3390/genes16050497] [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: 03/29/2025] [Revised: 04/25/2025] [Accepted: 04/25/2025] [Indexed: 05/29/2025] Open
Abstract
BACKGROUND/OBJECTIVE The single-nucleotide polymorphism (SNP) rs10830963 in the melatonin receptor 1B (MTNR1B) gene influences insulin secretion and glucose metabolism and has been associated with an increased risk of type-2 diabetes. This study aimed to explore the interaction between dietary intake and the MTNR1B rs10830963 polymorphism on glycemic profiles in young Brazilian adults. METHODS This cross-sectional study assessed 200 healthy young adults (19-24 years), evaluating the MTNR1B rs10830963 genotype, anthropometric parameters, glycemic markers (fasting insulin, glucose, HOMA-IR, and HOMA-β), and dietary intake via three 24 h dietary recalls. Genotype-diet interactions were tested using multivariate linear regression models adjusted for confounders. RESULTS The carriers of the G allele exhibited a positive association with fasting insulin levels (p = 0.003), insulin/glucose ratio (p = 0.004), HOMA-IR (p = 0.003), and HOMA-β (p = 0.018). Energy-adjusted fiber intake showed a significant genotype-specific interaction only in carriers of the G allele, where higher dietary fiber intake was significantly associated with lower fasting insulin (pinteraction = 0.034) and HOMA-IR (pinteraction = 0.028). CONCLUSION Our findings indicate that the MTNR1B rs10830963 polymorphism is associated with glycemic markers, and dietary fiber intake may attenuate the adverse effects of the MTNR1B rs10830963 G allele on glycemic profiles in young Brazilian adults. This highlights the potential role of fiber in improving health outcomes for individuals carrying this risk allele. To validate these results and assess the broader implications for the Brazilian population, further intervention studies and larger-scale research are essential.
Collapse
Affiliation(s)
- Ana Carolina da Silva Lima
- Nutritional Genomics Research Group, Faculty of Nutrition, Federal University of Goiás, Goiania 74605-080, GO, Brazil; (A.C.d.S.L.); (N.T.C.); (N.R.d.S.); (M.M.M.); (A.C.S.D.)
| | - Nathália Teixeira Cruvinel
- Nutritional Genomics Research Group, Faculty of Nutrition, Federal University of Goiás, Goiania 74605-080, GO, Brazil; (A.C.d.S.L.); (N.T.C.); (N.R.d.S.); (M.M.M.); (A.C.S.D.)
| | - Nara Rubia da Silva
- Nutritional Genomics Research Group, Faculty of Nutrition, Federal University of Goiás, Goiania 74605-080, GO, Brazil; (A.C.d.S.L.); (N.T.C.); (N.R.d.S.); (M.M.M.); (A.C.S.D.)
| | - Marcela Moraes Mendes
- Nutritional Genomics Research Group, Faculty of Nutrition, Federal University of Goiás, Goiania 74605-080, GO, Brazil; (A.C.d.S.L.); (N.T.C.); (N.R.d.S.); (M.M.M.); (A.C.S.D.)
| | - Amélia Cristina Stival Duarte
- Nutritional Genomics Research Group, Faculty of Nutrition, Federal University of Goiás, Goiania 74605-080, GO, Brazil; (A.C.d.S.L.); (N.T.C.); (N.R.d.S.); (M.M.M.); (A.C.S.D.)
- Health Research Coordination, Organization: State Department of Health from Goiás (SES-GO), Goiânia 74853-070, GO, Brazil
| | | | - Karani S. Vimaleswaran
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences, University of Reading, Reading RG6 6DZ, UK
- The Institute for Food, Nutrition, and Health, University of Reading, Reading RG6 6AH, UK
| | - Maria Aderuza Horst
- Nutritional Genomics Research Group, Faculty of Nutrition, Federal University of Goiás, Goiania 74605-080, GO, Brazil; (A.C.d.S.L.); (N.T.C.); (N.R.d.S.); (M.M.M.); (A.C.S.D.)
| |
Collapse
|
23
|
Kjaergaard AD, Ellervik C, Jessen N, Lessard SJ. Cardiorespiratory Fitness, Body Composition, Diabetes, and Longevity: A 2-Sample Mendelian Randomization Study. J Clin Endocrinol Metab 2025; 110:1451-1459. [PMID: 38864459 PMCID: PMC12012764 DOI: 10.1210/clinem/dgae393] [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: 05/20/2024] [Accepted: 06/07/2024] [Indexed: 06/13/2024]
Abstract
CONTEXT Cardiorespiratory fitness, commonly assessed as maximal volume of oxygen consumption (VO2max), has emerged as an important predictor of morbidity and mortality. OBJECTIVE We investigated the causality and directionality of the associations of VO2max with body composition, physical activity, diabetes, performance enhancers, and longevity. METHODS Using publicly available summary statistics from the largest genome-wide association studies publicly available, we conducted a bidirectional 2-sample Mendelian randomization (MR) study. Bidirectional MR tested directionality, and estimated the total causal effects, whereas multivariable MR (MVMR) estimated independent causal effects. Cardiorespiratory fitness (VO2max) was estimated from a submaximal cycle ramp test (N ≈ 70 000) and scaled to total body weight, and in additional analyses to fat-free mass (mL/min/kg). RESULTS Genetically predicted higher (per 1 SD increase) body fat percentage was associated with lower VO2max (β = -0.36; 95% CI: -0.40, -0.32, P = 6 × 10-77). Meanwhile, genetically predicted higher appendicular lean mass (β = 0.10; 95% CI: 0.08 to 0.13), physical activity (β = 0.29; 95% CI: 0.07 to 0.52), and performance enhancers (fasting insulin, hematocrit, and free testosterone in men) were all positively associated with VO2max (all P < .01). Genetic predisposition to diabetes had no effect on VO2max. MVMR showed independent causal effects of body fat percentage, appendicular lean mass, physical activity, and hematocrit on VO2max, as well as of body fat percentage and type 2 diabetes (T2D) on longevity. Genetically predicted VO2max showed no associations. CONCLUSION Cardiorespiratory fitness can be improved by favorable body composition, physical activity, and performance enhancers. Despite being a strong predictor of mortality, VO2max is not causally associated with T2D or longevity.
Collapse
Affiliation(s)
- Alisa D Kjaergaard
- Steno Diabetes Center Aarhus, Aarhus University Hospital, 8200 Aarhus, Denmark
- Joslin Diabetes Center, Boston, MA 02115, USA
| | - Christina Ellervik
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Department of Clinical Biochemistry, Zealand University Hospital, 4600 Køge, Denmark
- Department of Laboratory Medicine, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pathology, Harvard Medical School, Boston, MA 02115, USA
| | - Niels Jessen
- Steno Diabetes Center Aarhus, Aarhus University Hospital, 8200 Aarhus, Denmark
- Department of Biomedicine, Faculty of Health, Aarhus University, 8000 Aarhus, Denmark
| | - Sarah J Lessard
- Joslin Diabetes Center, Boston, MA 02115, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
24
|
Wang W, Xiao W, Song Z, Zhuang Z, Huang N, Zhao Y, Huang T. Fetal/Maternal-Determined Birth Weight and Adulthood Type 2 Diabetes and Its Subtypes: A Mendelian Randomization Study. J Clin Endocrinol Metab 2025; 110:1287-1294. [PMID: 38961757 DOI: 10.1210/clinem/dgae455] [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: 03/31/2024] [Revised: 06/12/2024] [Accepted: 07/02/2024] [Indexed: 07/05/2024]
Abstract
BACKGROUND Lower birth weight (BW) might increase the risk of adulthood type 2 diabetes, but its associations with the highly heterogeneous type 2 diabetes subtypes remain to be studied. In addition, whether the associations between lower BW and adulthood type 2 diabetes risks depend on fetal or maternal effect is largely unknown. METHODS In this study, we performed a two-sample Mendelian randomization analysis to study the associations between overall, fetal-determined, and maternal-determined BW and the risks of type 2 diabetes and its subtypes, namely mild age-related diabetes (MARD), mild obesity-related diabetes (MOD), severe insulin-deficient diabetes (SIDD), and severe insulin-resistant diabetes (SIRD). RESULTS Lower BW was genetically associated with increased risks of type 2 diabetes (odds ratio [OR]: 1.86; 95% CI: 1.53, 2.26), MARD (OR: 2.15; 95% CI: 1.43, 3.23), MOD (OR: 1.75; 95% CI: 1.10, 2.77), SIDD (OR: 1.86; 95% CI: 1.11, 3.10), and SIRD (OR: 1.66; 95% CI: 1.06, 2.60). When examining the fetal-determined genetic effects independently, lower BW remained associated with type 2 diabetes and its subtypes, except for MOD. Using maternal-determined BW-lowering genotypes to proxy for an adverse intrauterine environment provided no evidence that it raised offspring risks of type 2 diabetes. CONCLUSION Fetal-determined but not maternal-determined lower BW were associated with increased risks of adulthood type 2 diabetes and its subtypes. Our results underscored the importance of early targeted management among people with a low BW in the prevention of type 2 diabetes.
Collapse
Affiliation(s)
- Wenxiu Wang
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing 100191, China
| | - Wendi Xiao
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing 100191, China
| | - Zimin Song
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing 100191, China
| | - Zhenhuang Zhuang
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing 100191, China
| | - Ninghao Huang
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing 100191, China
| | - Yimin Zhao
- Department of Sports Medicine, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Tao Huang
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing 100191, China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing 100191, China
- State Key Laboratory of General Artificial Intelligence, Peking University, Beijing 100191, China
| |
Collapse
|
25
|
Gozdecka M, Dudek M, Wen S, Gu M, Stopforth RJ, Rak J, Damaskou A, Grice GL, McLoughlin MA, Bond L, Wilson R, Giotopoulos G, Shanmugiah VM, Bakar RB, Yankova E, Cooper JL, Narayan N, Horton SJ, Asby R, Pask DC, Mupo A, Duddy G, Marando L, Georgomanolis T, Carter P, Ramesh AP, Dunn WG, Barcena C, Gallipoli P, Yusa K, Petrovski S, Wright P, Quiros PM, Frezza C, Nathan JA, Kaser A, Kar S, Tzelepis K, Mitchell J, Fabre MA, Huntly BJP, Vassiliou GS. Mitochondrial metabolism sustains DNMT3A-R882-mutant clonal haematopoiesis. Nature 2025:10.1038/s41586-025-08980-6. [PMID: 40239706 DOI: 10.1038/s41586-025-08980-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 04/04/2025] [Indexed: 04/18/2025]
Abstract
Somatic DNMT3A-R882 codon mutations drive the most common form of clonal haematopoiesis (CH) and are associated with increased acute myeloid leukaemia (AML) risk1,2. Preventing expansion of DNMT3A-R882-mutant haematopoietic stem/progenitor cells (HSPCs) may therefore avert progression to AML. To identify DNMT3A-R882-mutant-specific vulnerabilities, we conducted a genome-wide CRISPR screen on primary mouse Dnmt3aR882H/+ HSPCs. Among the 640 vulnerability genes identified, many were involved in mitochondrial metabolism, and metabolic flux analysis confirmed enhanced oxidative phosphorylation use in Dnmt3aR882H/+ versus Dnmt3a+/+ (WT) HSPCs. We selected citrate/malate transporter Slc25a1 and complex I component Ndufb11, for which pharmacological inhibitors are available, for downstream studies. In vivo administration of SLC25A1 inhibitor CTPI2 and complex I inhibitors IACS-010759 and metformin suppressed post-transplantation clonal expansion of Dnmt3aR882H/+, but not WT, long-term haematopoietic stem cells. The effect of metformin was recapitulated using a primary human DNMT3A-R882 CH sample. Notably, analysis of 412,234 UK Biobank participants showed that individuals taking metformin had a markedly lower prevalence of DNMT3A-R882-mutant CH, after controlling for potential confounders including glycated haemoglobin, diabetes and body mass index. Collectively, our data propose modulation of mitochondrial metabolism as a therapeutic strategy for prevention of DNMT3A-R882-mutant AML.
Collapse
Affiliation(s)
- Malgorzata Gozdecka
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK.
- Department of Haematology, University of Cambridge, Cambridge, UK.
| | - Monika Dudek
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Sean Wen
- Department of Haematology, University of Cambridge, Cambridge, UK
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals Research and Development, AstraZeneca, Cambridge, UK
| | - Muxin Gu
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Richard J Stopforth
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
| | - Justyna Rak
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Aristi Damaskou
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Guinevere L Grice
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
| | - Matthew A McLoughlin
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Laura Bond
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Rachael Wilson
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - George Giotopoulos
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Vijaya Mahalingam Shanmugiah
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Rula Bany Bakar
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
| | - Eliza Yankova
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
- Milner Therapeutics Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
| | - Jonathan L Cooper
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Nisha Narayan
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Sarah J Horton
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Ryan Asby
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Dean C Pask
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | | | | | - Ludovica Marando
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Theodoros Georgomanolis
- Cluster of Excellence Cellular Stress Responses in Aging-associated Diseases (CECAD), University Hospital Cologne, Cologne, Germany
| | - Paul Carter
- Section of Cardiovascular Medicine, The Victor Phillip Dahdalleh Heart and Lung Research Institute, The University of Cambridge, Papworth Road, Cambridge Biomedical Campus, Cambridge, UK
| | - Amirtha Priya Ramesh
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - William G Dunn
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Clea Barcena
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo, Oviedo, Spain
| | - Paolo Gallipoli
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Kosuke Yusa
- Stem Cell Genetics, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Slavé Petrovski
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals Research and Development, AstraZeneca, Cambridge, UK
| | - Penny Wright
- Department of Anatomic Pathology, Canterbury Health Laboratories, Christchurch, New Zealand
| | - Pedro M Quiros
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo, Oviedo, Spain
| | - Christian Frezza
- Cluster of Excellence Cellular Stress Responses in Aging-associated Diseases (CECAD), University Hospital Cologne, Cologne, Germany
- Institute of Genetics, Faculty of Mathematics and Natural Sciences, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - James A Nathan
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
| | - Arthur Kaser
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Siddhartha Kar
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Konstantinos Tzelepis
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
- Milner Therapeutics Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
| | - Jonathan Mitchell
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals Research and Development, AstraZeneca, Cambridge, UK
| | - Margarete A Fabre
- Department of Haematology, University of Cambridge, Cambridge, UK
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals Research and Development, AstraZeneca, Cambridge, UK
- Department of Haematology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Brian J P Huntly
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK.
- Department of Haematology, University of Cambridge, Cambridge, UK.
- Department of Haematology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.
| | - George S Vassiliou
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK.
- Department of Haematology, University of Cambridge, Cambridge, UK.
- Department of Haematology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK.
| |
Collapse
|
26
|
Ma XB, Lv YL, Qian L, Yang JF, Song Q, Liu YM. Evaluating the effects of coffee consumption on the structure and function of the heart from multiple perspectives. Front Cardiovasc Med 2025; 12:1453106. [PMID: 40303614 PMCID: PMC12037506 DOI: 10.3389/fcvm.2025.1453106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 04/01/2025] [Indexed: 05/02/2025] Open
Abstract
Objective To assess the causal relationship between coffee consumption and cardiac structure and function in elderly European populations using multiple genetic methodologies. Methods Leveraging genome-wide association study (GWAS) data from elderly European populations, we conducted linkage disequilibrium score regression (LDSC), two-step Mendelian randomization (MR), and colocalization analyses to investigate genetic associations, causal relationships, and mediating effects among these factors. Robustness of findings was verified through comprehensive sensitivity analyses. Results LDSC regression analysis revealed positive genetic correlations between coffee consumption and cardiac parameters, excluding left ventricular (LV) ejection fraction and right ventricular (RV) ejection fraction. MR results demonstrated favorable associations between increased coffee consumption and cardiac parameters. After applying the Bonferroni adjustment to IVW analysis, as coffee consumption increased by each 1-cup/day, LV end-diastolic volume increased (β = 0.128; 95% CI: 0.043-0.212; P = 0.002), an increase in LV end-systolic volume (β = 0.143; 95% CI: 0.053-0.232; P = 0.001), an increase in RV end-diastolic volume (β = 0.200; 95% CI: 0.095-0.305; P < 0.001), and an increase in RV stroke volume (β = 0.209; 95% CI: 0.104-0.313; P < 0.001). Mediation analyses indicated that each 1-cup/day increase in coffee consumption significantly correlated with reduced diastolic blood pressure (DBP) and elevated body mass index (BMI). Notably, higher DBP exhibited inverse associations with ventricular systolic/diastolic functional parameters, whereas increased BMI demonstrated positive associations with these parameters, collectively mitigating age-related ventricular volume loss. No U-shaped associations were detected in linear MR frameworks. Colocalization analyses confirmed shared causal genetic variants between coffee intake and cardiac remodeling phenotypes. Conclusions Genetically predicted coffee consumption may counteract age-associated ventricular volume loss in elderly Europeans through dual mediation pathways involving DBP reduction and BMI elevation. These structural adaptations suggest potential cardioprotective mechanisms against senile cardiac atrophy. Future studies should prioritize the integration of coffee consumption into cardiovascular risk assessment frameworks and develop personalized recommendations based on individual health profiles.
Collapse
Affiliation(s)
- Xiong-Bin Ma
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
| | - Yan-Lin Lv
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
| | - Lin Qian
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
| | - Jing-Fen Yang
- The First Clinical Medical College of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Qian Song
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
| | - Yong-Ming Liu
- Geriatric Cardiovascular Department and Gansu Clinical Research Center for Geriatric Diseases, First Hospital of Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
27
|
Ji Y, Liu N, Yang Y, Wang M, Cheng J, Zhu W, Qiu S, Geng Z, Cui G, Yu Y, Liao W, Zhang H, Gao B, Xu X, Han T, Yao Z, Zhang Q, Qin W, Liu F, Liang M, Wang S, Xu Q, Xu J, Fu J, Zhang P, Li W, Shi D, Wang C, Lui S, Yan Z, Chen F, Zhang J, Shen W, Miao Y, Wang D, Gao JH, Zhang X, Xu K, Zuo XN, Zhang L, Ye Z, Li MJ, Xian J, Zhang B, Yu C. Cross-ancestry and sex-stratified genome-wide association analyses of amygdala and subnucleus volumes. Nat Genet 2025; 57:839-850. [PMID: 40097784 DOI: 10.1038/s41588-025-02136-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: 11/14/2023] [Accepted: 02/19/2025] [Indexed: 03/19/2025]
Abstract
The amygdala is a small but critical multi-nucleus structure for emotion, cognition and neuropsychiatric disorders. Although genetic associations with amygdala volumetric traits have been investigated in sex-combined European populations, cross-ancestry and sex-stratified analyses are lacking. Here we conducted cross-ancestry and sex-stratified genome-wide association analyses for 21 amygdala volumetric traits in 6,923 Chinese and 48,634 European individuals. We identified 191 variant-trait associations (P < 2.38 × 10-9), including 47 new associations (12 new loci) in sex-combined univariate analyses and seven additional new loci in sex-combined and sex-stratified multivariate analyses. We identified 12 ancestry-specific and two sex-specific associations. The identified genetic variants include 16 fine-mapped causal variants and regulate amygdala and fetal brain gene expression. The variants were enriched for brain development and colocalized with mood, cognition and neuropsychiatric disorders. These results indicate that cross-ancestry and sex-stratified genetic association analyses may provide insight into the genetic architectures of amygdala and subnucleus volumes.
Collapse
Affiliation(s)
- Yuan Ji
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging and State Key Laboratory of Experimental Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Nana Liu
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging and State Key Laboratory of Experimental Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yunjun Yang
- Department of Radiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Meiyun Wang
- Department of Radiology, Henan Provincial People's Hospital & Zhengzhou University People's Hospital, Zhengzhou, China
- Biomedical Institute, Henan Academy of Sciences, Zhengzhou, China
| | - Jingliang Cheng
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wenzhen Zhu
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shijun Qiu
- Department of Medical Imaging, The First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Zuojun Geng
- Department of Medical Imaging, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Guangbin Cui
- Functional and Molecular Imaging Key Lab of Shaanxi Province & Department of Radiology, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Yongqiang Yu
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Weihua Liao
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, China
- Molecular Imaging Research Center of Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Hui Zhang
- Department of Radiology, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Bo Gao
- Department of Radiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
- Department of Radiology, Yantai Yuhuangding Hospital, Yantai, China
| | - Xiaojun Xu
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Tong Han
- Department of Radiology, Tianjin Huanhu Hospital, Tianjin, China
| | - Zhenwei Yao
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Quan Zhang
- Department of Radiology, Characteristic Medical Center of Chinese People's Armed Police Force, Tianjin, China
| | - Wen Qin
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging and State Key Laboratory of Experimental Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Feng Liu
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging and State Key Laboratory of Experimental Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Meng Liang
- School of Medical Imaging and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University, Tianjin, China
| | - Sijia Wang
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging and State Key Laboratory of Experimental Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Qiang Xu
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging and State Key Laboratory of Experimental Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Jiayuan Xu
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging and State Key Laboratory of Experimental Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Jilian Fu
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging and State Key Laboratory of Experimental Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Peng Zhang
- Department of Radiology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Wei Li
- Department of Radiology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Dapeng Shi
- Department of Radiology, Henan Provincial People's Hospital & Zhengzhou University People's Hospital, Zhengzhou, China
| | - Caihong Wang
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Su Lui
- Department of Radiology, Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, Chengdu, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, China
| | - Zhihan Yan
- Department of Radiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Feng Chen
- Department of Radiology, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China
| | - Jing Zhang
- Department of Magnetic Resonance, Lanzhou University Second Hospital, Lanzhou, China
- Gansu Province Clinical Research Center for Functional and Molecular Imaging, Lanzhou, China
| | - Wen Shen
- Department of Radiology, Tianjin First Center Hospital, Tianjin, China
| | - Yanwei Miao
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Dawei Wang
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, China
| | - Jia-Hong Gao
- Center for MRI Research, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Xiaochu Zhang
- Division of Life Science and Medicine, University of Science & Technology of China, Hefei, China
| | - Kai Xu
- Department of Radiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xi-Nian Zuo
- Developmental Population Neuroscience Research Center at IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
- Institute of Psychology, Chinese Academy of Sciences, Beijing, China
| | - Longjiang Zhang
- Department of Radiology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Zhaoxiang Ye
- Department of Radiology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Mulin Jun Li
- Department of Bioinformatics, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Junfang Xian
- Department of Radiology, Beijing Tongren Hospital, Capital Medical University, Beijing, China.
| | - Bing Zhang
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| | - Chunshui Yu
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging and State Key Laboratory of Experimental Hematology, Tianjin Medical University General Hospital, Tianjin, China.
- School of Medical Imaging and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
28
|
Xu H, Li B, Lv P, Chen Y, Lin Y, Zhang A, Zhao J, Zhou G, Wu L. Inhibition of Putative Ibrutinib Targets Promotes Atrial Fibrillation, Conduction Blocks, and Proarrhythmic Electrocardiogram Indices: A Mendelian Randomization Analysis. CANCER INNOVATION 2025; 4:e70004. [PMID: 40078362 PMCID: PMC11897533 DOI: 10.1002/cai2.70004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 07/05/2024] [Accepted: 10/16/2024] [Indexed: 03/14/2025]
Abstract
Background The mechanism by which ibrutinib, a Bruton's tyrosine kinase inhibitor, can elevate the risk of arrhythmias is not fully elucidated. In this study, we explored how inhibition of off-target kinases can contribute to this phenomenon. Methods We performed a Mendelian randomization analysis to examine the causal associations between genetically proxied inhibition of six putative ibrutinib drug targets (ErbB2/HER2, CSK, JAK3, TEC, BLK, and PLCG2) and the atrial fibrillation (AF) risk, proarrhythmic ECG indices, and cardiometabolic traits and diseases. Inverse-variance weighted random-effects models and Wald ratio were used to examine the associations between genetically proxied inhibition of these drug targets and the risk of outcomes. Colocalization analyses were employed to examine the robustness of the causally significant findings. ELISAs were used to measure ErbB2 levels in intracardiac plasma samples. Results Genetically proxied ErbB2 inhibition was associated with an increased AF risk, higher P wave terminal force, and prolonged QTc interval. Patients with AF had significantly higher intracardiac ErbB2 levels compared with patients with paroxysmal supraventricular tachycardia. CSK inhibition prolonged the QRS duration, decreased the QTc interval, and was potentially linked to conduction blocks. PLCG2 inhibition led to decreased P wave terminal force, shorter QTc interval, and increased risk of left bundle branch block. BLK inhibition shortened the QTc interval and was also associated with atrioventricular block. Conclusion The off-target effects and downstream targets of ibrutinib, including CSK, PLCG2, ERBB2, TEC, and BLK, may lead to cardiac electrical homeostasis imbalances and lethal cardiovascular diseases. Using drugs that inhibit these targets should be given extra caution.
Collapse
Affiliation(s)
- Hongxuan Xu
- Department of CardiologyPeking University First HospitalBeijingChina
| | - Bingxun Li
- Department of CardiologyPeking University First HospitalBeijingChina
| | - Pinchao Lv
- Department of CardiologyPeking University First HospitalBeijingChina
| | - Ying Chen
- Department of CardiologyPeking University First HospitalBeijingChina
| | - Yanyun Lin
- Department of CardiologyPeking University First HospitalBeijingChina
| | - An Zhang
- Department of CardiologyPeking University First HospitalBeijingChina
| | - Jing Zhao
- Department of CardiologyPeking University First HospitalBeijingChina
| | - Guoxiong Zhou
- Department of CardiologyPeking University First HospitalBeijingChina
| | - Lin Wu
- Department of CardiologyPeking University First HospitalBeijingChina
- State Key Laboratory of Vascular Homeostasis and RemodelingPeking UniversityBeijingChina
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Institute of Cardiovascular ResearchSouthwest Medical UniversityLuzhouChina
| |
Collapse
|
29
|
Venkataraghavan S, Pankow JS, Boerwinkle E, Fornage M, Selvin E, Ray D. Epigenome-wide association study of incident type 2 diabetes in Black and White participants from the Atherosclerosis Risk in Communities Study. Diabetologia 2025; 68:815-834. [PMID: 39971753 PMCID: PMC12054846 DOI: 10.1007/s00125-024-06352-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 05/29/2024] [Indexed: 02/21/2025]
Abstract
AIMS/HYPOTHESIS DNA methylation studies of incident type 2 diabetes in US populations are limited and to our knowledge none include individuals of African descent. We aimed to fill this gap by identifying methylation sites (CpG sites) and regions likely influencing the development of type 2 diabetes using data from Black and White individuals from the USA. METHODS We prospectively followed 2091 Black and 1029 White individuals without type 2 diabetes from the Atherosclerosis Risk in Communities study over a median follow-up period of 17 years, and performed an epigenome-wide association analysis of blood-based methylation levels with incident type 2 diabetes using Cox regression. We assessed whether significant CpG sites were associated with incident type 2 diabetes independently of BMI or fasting glucose at baseline. We estimated variation in incident type 2 diabetes accounted for by the major non-genetic risk factors and the significant CpG sites. We also examined groups of methylation sites that were differentially methylated. We performed replication of previously discovered CpG sites associated with prevalent and/or incident type 2 diabetes. All analyses were adjusted for batch effects, cell-type proportions and relevant confounders. RESULTS At an epigenome-wide threshold (10-7), we detected seven novel diabetes-associated CpG sites, of which the sites at MICOS10 (cg05380846: HR 0.89, p=8.4 × 10-12), ZNF2 (cg01585592: HR 0.88, p=1.6 × 10-9), JPH3 (cg16696007: HR 0.87, p=7.8 × 10-9) and GPX6 (cg02793507: HR 0.85, p=2.7 × 10-8; cg00647063: HR 1.20, p=2.5 × 10-8) were identified in Black adults; chr17q25 (cg16865890: HR 0.8, p=6.9 × 10-8) in White adults; and chr11p15 (cg13738793: HR 1.11, p=7.7 × 10-8) in the meta-analysed group. The JPH3 and GPX6 sites remained epigenome-wide significant on adjustment for BMI, while only the JPH3 site retained significance after adjusting for fasting glucose. We replicated known type 2 diabetes-associated CpG sites, including cg19693031 at TXNIP, cg00574958 at CPT1A, cg16567056 at PLCB2, cg11024682 at SREBF1, cg08857797 at VPS25 and cg06500161 at ABCG1, three of which were replicated in Black adults at the epigenome-wide threshold and all of which had directionally consistent effects. We observed a modest increase in type 2 diabetes variance explained by the significantly associated CpG sites over and above traditional type 2 diabetes risk factors and fasting glucose (26.2% vs 30.5% in Black adults; 36.9% vs 39.4% in White adults). At the Šidák-corrected significance threshold of 5%, our differentially methylated region (DMR) analyses revealed several clusters of significant CpG sites, including a DMR consisting of a previously discovered CpG site at ADCY7 (pBlack=1.8 × 10-4, pWhite=3.6 × 10-3, pAll=1.6 × 10-9) and a DMR consisting of the promoter region of TP63 (pBlack=7.4 × 10-4, pWhite=3.9 × 10-3, pAll=1.4 × 10-5), which were differentially methylated across all racial and ethnic groups. CONCLUSIONS/INTERPRETATION This study illustrates improved discovery of CpG sites and regions by leveraging both individual CpG site analysis and DMR analyses in an unexplored population. Our findings include genes linked to diabetes in experimental studies (e.g. GPX6, JPH3 and TP63). The JPH3 and GPX6 sites were likely associated with incident type 2 diabetes independently of BMI. All the CpG sites except that at JPH3 were likely consequences of elevated glucose. Replication in African-descent individuals of CpG sites previously discovered mostly in individuals of European descent indicates that some of these methylation-type 2 diabetes associations are robust across racial and ethnic groups. This study is a first step towards understanding the influence of methylation on the incidence of type 2 diabetes and its disparity in two major racial and ethnic groups in the USA. It paves the way for future studies to investigate causal relationships between type 2 diabetes and the CpG sites and potentially elucidate molecular targets for intervention.
Collapse
Affiliation(s)
- Sowmya Venkataraghavan
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - James S Pankow
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Eric Boerwinkle
- The University of Texas Health School of Public Health, Houston, TX, USA
| | - Myriam Fornage
- Brown Foundation Institute for Molecular Medicine, The University of Texas Health Science Center, Houston, TX, USA
| | - Elizabeth Selvin
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
- Welch Center for Prevention, Epidemiology, & Clinical Research, Johns Hopkins University, Baltimore, MD, USA
| | - Debashree Ray
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA.
- Department of Biostatistics, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
30
|
Shan Y, Hu H, Chu Y. Cross-ancestry genome-wide association study identifies new susceptibility genes for preeclampsia. BMC Pregnancy Childbirth 2025; 25:379. [PMID: 40170147 PMCID: PMC11959822 DOI: 10.1186/s12884-025-07534-y] [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: 01/03/2025] [Accepted: 03/26/2025] [Indexed: 04/03/2025] Open
Abstract
BACKGROUND Preeclampsia (PE) is a heterogeneous, multi-organ pregnancy disorder that poses a significant health burden globally, with its pathogenesis remaining unclear. This study aimed to identify novel susceptibility genes for PE through a cross-ancestry genome-wide association study (GWAS). METHODS We performed meta-analysis to summarize the PE GWAS data from the United Kingdom, Finland, and Japan. Subsequently, the multi-ancestry sum of the single-effects model was used to perform cross-ancestry fine-mapping. The functional mapping and annotation (FUMA)-expression quantitative trait loci (eQTL) mapping method, transcriptome-wide association study (TWAS)- functional summary-based imputation (FUSION) method, genome-wide complex trait analysis (GCTA)-multivariate set-based association test (mBAT)-combo method, and polygenic priority score (PoPS) method were employed to screen for candidate genes. We utilized biomarker expression level imputation using summary-level statistics (BLISS), based on summary-level protein quantitative trait loci (pQTL) data, to conduct a multi-ancestry proteome-wide association study (PWAS) analysis, followed by candidate drug prediction. RESULTS Six novel susceptibility genes associated with PE risk were identified: NPPA, SWAP70, NPR3, FGF5, REPIN1, and ACAA1. High expression of the NPPA and SWAP70 and low expression of the remaining genes were associated with a reduced risk of PE. Furthermore, we identified drugs that target NPPA, NPR3, and REPIN1. CONCLUSIONS Our study identified NPPA, SWAP70, NPR3, FGF5, REPIN1, and ACAA1 as novel genes whose predicted expression was linked to the risk of PE, offering new insights into the genetic framework of this condition.
Collapse
Affiliation(s)
- Yuping Shan
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hong Hu
- Clinical Medicine, Nantong University, Nantong, China
| | - Yijing Chu
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
31
|
Zhang J, Jiao F, Wang Z, Zou C, Du X, Ye D, Jiang G. Identification of CD209 as an Intervention Target for Type 2 Diabetes After COVID-19 Infection: Insights From Proteome-Wide Mendelian Randomization. Diabetes 2025; 74:619-629. [PMID: 39874030 DOI: 10.2337/db24-0677] [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: 08/08/2024] [Accepted: 12/27/2024] [Indexed: 01/30/2025]
Abstract
ARTICLE HIGHLIGHTS Increasing evidence links coronavirus disease 2019 (COVID-19) infection with heightened type 2 diabetes (T2D) risk; however, the mechanisms underlying this relationship remain poorly understood. We aimed to identify mediating proteins linking COVID-19 infection with T2D, elucidating how COVID-19 might heighten T2D risk. Protein CD209 and central obesity potentially play a crucial role between COVID-19 susceptibility and T2D. Our results highlight CD209 as a potential intervention target for T2D prevention following COVID-19 infection.
Collapse
Affiliation(s)
- Jiaying Zhang
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, China
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Feng Jiao
- Guangzhou Centre for Applied Mathematics, Guangzhou University, Guangzhou, China
| | - Zhenqian Wang
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, China
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Chenfeng Zou
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, China
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiangjun Du
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, China
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, Guangdong, China
- Shenzhen Key Laboratory of Pathogenic Microbes and Biosafety, Shenzhen, Guangdong, China
| | - Dewei Ye
- Institute of Metabolic Science, Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Guozhi Jiang
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, China
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, Guangdong, China
- Shenzhen Key Laboratory of Pathogenic Microbes and Biosafety, Shenzhen, Guangdong, China
| |
Collapse
|
32
|
Yang X, Xiao R, Liu B, Xie B, Yang Z. The causal relationship of inflammation-related factors with osteoporosis: A Mendelian Randomization Analysis. Exp Gerontol 2025; 202:112715. [PMID: 39983802 DOI: 10.1016/j.exger.2025.112715] [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: 11/19/2024] [Revised: 02/10/2025] [Accepted: 02/15/2025] [Indexed: 02/23/2025]
Abstract
BACKGROUND We used Mendelian randomization (MR) approach to examine whether genetically determined inflammation-related risk factors play a role in the onset of osteoporosis (OP) in the European population. METHODS Genome-wide association studies (GWASs) summary statistics of estimated bone mineral density (eBMD) obtained from the public database GEnetic Factors for OSteoporosis Consortium (GEFOS) including 142,487 European people. For exposures, we utilized GWAS data of 9 risk factors including diseases chronic kidney disease (CKD) (41,395 cases and 439,303 controls), type 2 diabetes (T2D) (88,427 cases and 566,778 controls), Alzheimer's disease (AD) (71,880 cases, 383,378 controls) and major depression disorder (MDD) (9240 cases and 9519 controls) and lifestyle behaviors are from different consortiums. Inverse variance weighted (IVW) analysis was principal method in this study and random effect model was applied; MR-Egger method and weighted median method were also performed for reliable results. Cochran's Q test and MR-Egger regression were used to detect heterogeneity and pleiotropy and leave-one-out analysis was performed to find out whether there are influential SNPs. RESULTS We found that T2D (IVW: β = 0.05, P = 0.0014), FI (IVW: β = -0.22, P < 0.001), CKD (IVW: β = 0.02, P = 0.009), ALZ (IVW: β = 0.06, P = 0.005), Coffee consumption (IVW: β = 0.11, P = 0.003) were causally associated with OP (P<0.006after Bonferroni correction). CONCLUSIONS Our study revealed that T2D, FI, CKD, ALZ and coffee consumption are causally associated with OP. Future interventions targeting factors above could provide new clinical strategies for the personalized prevention and treatment of osteoporosis.
Collapse
Affiliation(s)
- Xinyue Yang
- Department of Neurology, Yongchuan Hospital of Chongqing Medical University, Chongqing Medical University, China
| | - Rui Xiao
- Department of General Practice, Yongchuan Hospital of Chongqing Medical University, Chongqing Medical University, China
| | - Beizhong Liu
- Central Laboratory of Yongchuan Hospital, Chongqing Medical University, China
| | - Bo Xie
- Department of General Practice, Yongchuan Hospital of Chongqing Medical University, Chongqing Medical University, China.
| | - Zhao Yang
- Department of Neurology, Yongchuan Hospital of Chongqing Medical University, Chongqing Medical University, China.
| |
Collapse
|
33
|
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, Kullo IJ, Puckelwartz MJ, Reza N, Satterfield BA, Singhal P, Arany Z, Cappola TP, Carruth ED, Day SM, Do R, Haggerty CM, Joseph J, McNally EM, Nadkarni G, Owens AT, Rader DJ, Ritchie MD, Sun YV, Voight BF, Levin MG, Damrauer SM. Common-variant and rare-variant genetic architecture of heart failure across the allele-frequency spectrum. Nat Genet 2025; 57:829-838. [PMID: 40195560 PMCID: PMC12049093 DOI: 10.1038/s41588-025-02140-2] [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: 04/25/2024] [Accepted: 02/21/2025] [Indexed: 04/09/2025]
Abstract
Heart failure is a complex trait, influenced by environmental and genetic factors, affecting over 30 million individuals worldwide. Here we report common-variant and rare-variant association studies of all-cause heart failure and examine how different classes of genetic variation impact its heritability. We identify 176 common-variant risk loci at genome-wide significance in 2,358,556 individuals and cluster these signals into five broad modules based on pleiotropic associations with anthropomorphic traits/obesity, blood pressure/renal function, atherosclerosis/lipids, immune activity and arrhythmias. In parallel, we uncover exome-wide significant associations for heart failure and rare predicted loss-of-function variants in TTN, MYBPC3, FLNC and BAG3 using exome sequencing of 376,334 individuals. We find that total burden heritability of rare coding variants is highly concentrated in a small set of Mendelian cardiomyopathy genes, while common-variant heritability is diffusely spread throughout the genome. Finally, we show that common-variant background modifies heart failure risk among carriers of rare pathogenic truncating variants in TTN. Together, these findings discern genetic links between dysregulated metabolism and heart failure and highlight a polygenic component to heart failure 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, USA
| | - Kathleen M Cardone
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - David Y Zhang
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Noah L Tsao
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Sarah Abramowitz
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Pranav Sharma
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - John S DePaolo
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Mitchell Conery
- Genomics and Computational Biology Graduate Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Krishna G Aragam
- Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kiran Biddinger
- Program in Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ozan Dilitikas
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Lily Hoffman-Andrews
- Division of Cardiovascular Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Renae L Judy
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Atlas Khan
- Division of Nephrology, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York City, NY, USA
| | - Iftikhar J Kullo
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Megan J Puckelwartz
- Department of Pharmacology, Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Nosheen Reza
- Division of Cardiovascular Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | | | - Pankhuri Singhal
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Zoltan Arany
- Division of Cardiovascular Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Cardiovascular Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Thomas P Cappola
- Division of Cardiovascular Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Eric D Carruth
- Department of Genomic Health, Geisinger, Danville, PA, USA
| | - Sharlene M Day
- Division of Cardiovascular Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Cardiovascular Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Ron Do
- The Charles Bronfman Institute for Personalized Medicine, Mount Sinai Icahn School of Medicine, New York City, NY, USA
- BioMe Phenomics Center, Mount Sinai Icahn School of Medicine, New York City, NY, USA
- Department of Genetics and Genomic Sciences, Mount Sinai Icahn School of Medicine, New York City, NY, USA
| | | | - Jacob Joseph
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Massachusetts Veterans Epidemiology Research and Information Center, VA Boston Healthcare System, Boston, MA, USA
| | - Elizabeth M McNally
- Center for Genetic Medicine, Bluhm Cardiovascular Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Girish Nadkarni
- Division of Nephrology, Department of Medicine, Mount Sinai Icahn School of Medicine, New York City, NY, USA
| | - Anjali T Owens
- Cardiovascular Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Daniel J Rader
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Division of Translational Medicine and Human Genetics, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Marylyn D Ritchie
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Institute for Biomedical Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Yan V Sun
- Atlanta VA Health Care System, Decatur, GA, USA
- Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA, USA
| | - Benjamin F Voight
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Michael G Levin
- Division of Cardiovascular Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
- Cardiovascular Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA.
| | - Scott M Damrauer
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Cardiovascular Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
| |
Collapse
|
34
|
Gummesson A, Lundmark P, Chen QS, Björnson E, Dekkers KF, Hammar U, Adiels M, Wang Y, Andersson T, Bergström G, Carlhäll CJ, Erlinge D, Jernberg T, Landfors F, Lind L, Mannila M, Melander O, Pirazzi C, Sundström J, Östgren CJ, Gunnarsson C, Orho-Melander M, Söderberg S, Fall T, Gigante B. A genome-wide association study of imaging-defined atherosclerosis. Nat Commun 2025; 16:2266. [PMID: 40164586 PMCID: PMC11958696 DOI: 10.1038/s41467-025-57457-7] [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: 04/16/2024] [Accepted: 02/22/2025] [Indexed: 04/02/2025] Open
Abstract
Imaging-defined atherosclerosis represents an intermediate phenotype of atherosclerotic cardiovascular disease (ASCVD). Genome-wide association studies (GWAS) on directly measured coronary plaques using coronary computed tomography angiography (CCTA) are scarce. In the so far largest population-based cohort with CCTA data, we performed a GWAS on coronary plaque burden as determined by the segment involvement score (SIS) in 24,811 European individuals. We identified 20 significant independent genetic markers for SIS, three of which were found in loci not implicated in ASCVD before. Further GWAS on coronary artery calcification showed similar results to that of SIS, whereas a GWAS on ultrasound-assessed carotid plaques identified both shared and non-shared loci with SIS. In two-sample Mendelian randomization studies using SIS-associated markers in UK Biobank and CARDIoGRAMplusC4D, one extra coronary segment with atherosclerosis corresponded to 1.8-fold increased odds of myocardial infarction. This GWAS data can aid future studies of causal pathways in ASCVD.
Collapse
Affiliation(s)
- Anders Gummesson
- Region Västra Götaland, Sahlgrenska University Hospital, Department of Clinical Genetics and Genomics, Gothenburg, Sweden.
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - Per Lundmark
- Department of Medical Sciences, Molecular Epidemiology, Uppsala University, Uppsala, Sweden
| | - Qiao Sen Chen
- Division of Cardiology, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Elias Björnson
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Koen F Dekkers
- Department of Medical Sciences, Molecular Epidemiology, Uppsala University, Uppsala, Sweden
| | - Ulf Hammar
- Department of Medical Sciences, Molecular Epidemiology, Uppsala University, Uppsala, Sweden
| | - Martin Adiels
- School of Public Health and Community Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Yunzhang Wang
- Department of Clinical Sciences, Danderyd University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Therese Andersson
- Department of Public Medicine and Clinical Health, Umeå University, Umeå, Sweden
| | - Göran Bergström
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Region Västra Götaland, Sahlgrenska University Hospital, Department of Clinical Physiology, Gothenburg, Sweden
| | - Carl-Johan Carlhäll
- Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden
- Department of Clinical Physiology in Linköping, Linköping University, Linköping, Sweden
- Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - David Erlinge
- Department of Clinical Sciences Lund, Cardiology, Lund University, Lund, Sweden
| | - Tomas Jernberg
- Department of Clinical Sciences, Danderyd University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Fredrik Landfors
- Department of Public Medicine and Clinical Health, Umeå University, Umeå, Sweden
| | - Lars Lind
- Department of Medical Sciences, Clinical Epidemiology, Uppsala University, Uppsala, Sweden
| | - Maria Mannila
- Department of Cardiology, Karolinska University Hospital, Stockholm, Sweden
| | - Olle Melander
- Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden
- Department of Emergency and Internal Medicine, Skåne University Hospital, Malmö, Sweden
| | - Carlo Pirazzi
- Region Västra Götaland, Sahlgrenska University Hospital, Department of Cardiology, Gothenburg, Sweden
| | - Johan Sundström
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
- The George Institute for Global Health, University of New South Wales, Sydney, Australia
| | - Carl Johan Östgren
- Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden
- Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - Cecilia Gunnarsson
- Department of Biomedical and Clinical Sciences, Division of Clinical Genetics, Linköping University, Linköping, Sweden
| | | | - Stefan Söderberg
- Department of Public Medicine and Clinical Health, Umeå University, Umeå, Sweden
| | - Tove Fall
- Department of Medical Sciences, Molecular Epidemiology, Uppsala University, Uppsala, Sweden
| | - Bruna Gigante
- Division of Cardiology, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Cardiology, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
35
|
Xiao J, Li J, Wu X, Hao Y, Zhao X, Zhang W, Xu B, Ma T, Zhang L, Xiang R, Cui H, Yang C, Yan P, Tang M, Wang Y, Qu Y, Chen L, Liu Y, Zou Y, Zhang L, Liu Z, Yao Y, Yang C, Zhang B, Jiang X. Adult Height, Cardiovascular Disease, and the Underlying Mechanism: A Comprehensive Epidemiological and Genetic Analysis. Can J Cardiol 2025:S0828-282X(25)00237-5. [PMID: 40174860 DOI: 10.1016/j.cjca.2025.03.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 02/13/2025] [Accepted: 03/06/2025] [Indexed: 04/04/2025] Open
Abstract
BACKGROUND Adult height measures the complete growth of an individual and influences the development of cardiovascular disease (CVD). Despite recent within-sibling studies that have suggested minimal effects from environmental confounders, biological mechanisms underlying the height-CVD relationship remain elusive. METHODS Leveraging the large-scale UK Biobank data set and summary statistics from the latest genome-wide association studies, we reevaluated the effect of height on 8 major CVD subtypes. Phenotypic associations were determined using Cox proportional hazard analysis. Putative causal relationships were assessed using univariable Mendelian randomization. Mediation analysis and 2-step Mendelian randomization were further performed to investigate the mediation effect of 15 common cardiometabolic or pulmonary risk factors. RESULTS Height was consistently associated with a decreased risk of coronary artery disease (CAD), confirmed in epidemiological (hazard ratio, 0.90; 95% confidence interval [CI], 0.88-0.91) and genetic (odds ratio, 0.89, 95% CI, 0.86-0.92) analysis. Forced vital capacity was identified as the most significant mediator for the height-CAD relationship in epidemiological (proportion-mediated, 65.6%; 95% CI, 53.1%-78.0%) and genetic (proportion-mediated, 46.2%; 95% CI, 5.0%-87.5%) analysis. Notably, obesity, and blood pressure, lipid, and C-reactive protein levels also exhibited significant mediatory effects. Despite a consistent risk effect of height on atrial fibrillation and venous thromboembolism, no promising mediator was identified. CONCLUSIONS Our study confirms the health effects of height on CAD, atrial fibrillation, and venous thromboembolism and emphasizes forced vital capacity as the primary pathway that links height to CAD. Importantly, it indicates that the CAD risk associated with nonmodifiable height could be mitigated through enhanced lung function and cardiometabolic conditions.
Collapse
Affiliation(s)
- Jinyu Xiao
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Jiayuan Li
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Xueyao Wu
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Yu Hao
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Xunying Zhao
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Wenqiang Zhang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Bin Xu
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Tianpei Ma
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Li Zhang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Rong Xiang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Huijie Cui
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Chao Yang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Peijing Yan
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Mingshuang Tang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Yutong Wang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Yang Qu
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Lin Chen
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Yunjie Liu
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Yanqiu Zou
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Ling Zhang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China; Department of Iatrical Polymer Material and Artificial Apparatus, School of Polymer Science and Engineering, Sichuan University, Chengdu, China
| | - Zhenmi Liu
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China; Department of Maternal, Child and Adolescent Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Yuqin Yao
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China; Department of Occupational and Environmental Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Chunxia Yang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Ben Zhang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China; Department of Cardiology, Department of Neurology, and Department of Oncology, Hainan General Hospital and Hainan Affiliated Hospital, Hainan Medical University, Haikou, China.
| | - Xia Jiang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China; Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China; Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
36
|
Ma X, Ding L, Li S, Fan Y, Wang X, Han Y, Yuan H, Sun L, He Q, Liu M. Druggable genome-wide Mendelian randomization identifies therapeutic targets for metabolic dysfunction-associated steatotic liver disease. Lipids Health Dis 2025; 24:113. [PMID: 40140823 PMCID: PMC11938603 DOI: 10.1186/s12944-025-02515-8] [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: 11/06/2024] [Accepted: 03/06/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD) affects > 25% of the global population, potentially leading to severe hepatic and extrahepatic complications, including metabolic dysfunction-associated steatohepatitis. Given that the pathophysiology of MASLD is incompletely understood, identifying therapeutic targets and optimizing treatment strategies are crucial for addressing this severe condition. METHODS Mendelian randomization (MR) analysis was conducted using two genome-wide association study datasets: a European meta-analysis (8,434 cases; 770,180 controls) and an additional study (3,954 cases; 355,942 controls), identifying therapeutic targets for MASLD. Of 4302 drug-target genes, 2,664 genetic instrument variables were derived from cis-expression quantitative trait loci (cis-eQTLs). Colocalization analyses assessed shared causal variants between MASLD-associated single nucleotide polymorphisms and eQTLs. Using the drug target gene cis-eQTL of liver tissue from the genotype-tissue expression project, we performed MR and summary MR to validate the significance of the gene results of the blood eQTL MR. RNA-sequencing data from liver biopsies were validated using immunohistochemistry and quantitative polymerase chain reaction (qPCR) tests to confirm gene expression findings. RESULT MR analysis across both datasets identified significant MR associations between MASLD and two drug targets-milk fat globule-EGF factor 8 (MFGE8) (odds ratio [OR] 0.89, 95% confidence interval [CI] 0.85-0.94; P = 2.15 × 10-6) and cluster of differentiation 33 (CD33) (OR 1.17, 95% CI 1.10-1.25; P = 1.39 × 10-6). Both targets exhibited strong colocalization with MASLD. Genetic manipulation indicating MFGE8 activation and CD33 inhibition did not increase the risk for other metabolic disorders. RNA-sequencing, qPCR, and immunohistochemistry validation demonstrated consistent differential expressions of MFGE8 and CD33 in MASLD. CONCLUSION CD33 inhibition can reduce MASLD risk, while MFGE8 activation may offer therapeutic benefits for MASLD treatment.
Collapse
Affiliation(s)
- Xiaohui Ma
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
- Department of Endocrinology and Metabolism, Baotou Central Hospital, Baotou, China
| | - Li Ding
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Shuo Li
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Yu Fan
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Xin Wang
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Yitong Han
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Hengjie Yuan
- Department of Pharmacy, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.
| | - Longhao Sun
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.
| | - Qing He
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.
| | - Ming Liu
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| |
Collapse
|
37
|
Chen X, Cheng Z, Xu J, Wang Q, Zhao Z, Cheng Q, Jiang Q. The dual role of diabetes on oral potentially malignant disorders. Eur J Med Res 2025; 30:199. [PMID: 40122861 PMCID: PMC11931820 DOI: 10.1186/s40001-025-02462-1] [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: 10/14/2024] [Accepted: 03/13/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND Observational studies suggest a link between diabetes and oral potentially malignant disorders (OPMDs), such as oral lichen planus (OLP) and oral leukoplakia (OLK). The causal relationship, as well as the type of diabetes that promotes OPMDs development, remains unclear. This Mendelian randomization (MR) study estimated the causal effects of diabetes-related traits on OPMDs. METHODS Large-scale genome-wide association study data on type 1 diabetes (T1D), type 2 diabetes (T2D), fasting glucose (FG), fasting insulin (FI), glycated hemoglobin (HbA1c), OLP, OLK, and actinic cheilitis (AC) were used. Causal effects were assessed using inverse-variance weighted (IVW), weighted median, and MR-Egger methods. Multivariable MR analyses evaluated the independent roles of these traits, with extensive sensitivity analyses. RESULTS Genetic susceptibility to T1D (IVW OR = 1.09, 95% CI 1.02-1.17, P = 0.007) and T2D (IVW OR = 0.91, 95% CI 0.86-0.97, P = 0.002) showed protective effects against AC. T1D was associated with an increased risk of OLP (IVW OR = 1.09, 95% CI 1.02-1.17, P = 0.007). The effect of T1D on AC and OLP remained robust after adjusting for FI, FG, and HbA1c, while T2D's effect on AC was not significant when considering these glycemic traits. No potential pleiotropy was detected (P > 0.05). CONCLUSIONS T1D may have a causal role in the development of OLP independent of glycemic traits, emphasizing the need for routine oral examinations in T1D patients. Conversely, genetically predicted T1D and T2D are significantly associated with a reduced risk of AC, challenging previous assumptions and offering new insights into the relationship between diabetes and OPMDs. Further extensive investigations are required to address the limitations of this study and to clarify these associations.
Collapse
Affiliation(s)
- Xin Chen
- Department of Oral and Maxillofacial Surgery, Jiangyin People's Hospital Affiliated to Nantong University, No. 163, Shoushan Road, Jiangyin, 214400, Jiangsu Province, China
| | - Zheng Cheng
- Department of Oral and Maxillofacial Surgery, Jiangyin People's Hospital Affiliated to Nantong University, No. 163, Shoushan Road, Jiangyin, 214400, Jiangsu Province, China
| | - Junyu Xu
- Department of Oral and Maxillofacial Surgery, Jiangyin People's Hospital Affiliated to Nantong University, No. 163, Shoushan Road, Jiangyin, 214400, Jiangsu Province, China
| | - Qianyi Wang
- Department of Cardiology, Jiangyin People's Hospital Affiliated to Nantong University, Jiangyin, China
| | - Zhibai Zhao
- Department of Oral Mucosal Diseases, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
| | - Qing Cheng
- Department of Oral and Maxillofacial Surgery, Jiangyin People's Hospital Affiliated to Nantong University, No. 163, Shoushan Road, Jiangyin, 214400, Jiangsu Province, China.
| | - Qianglin Jiang
- Department of Oral and Maxillofacial Surgery, Jiangyin People's Hospital Affiliated to Nantong University, No. 163, Shoushan Road, Jiangyin, 214400, Jiangsu Province, China.
| |
Collapse
|
38
|
Lin X, Liang B, Lam TH, Cheng KK, Zhang W, Xu L. The mediating roles of anthropo-metabolic biomarkers on the association between beverage consumption and breast cancer risk. Nutr J 2025; 24:46. [PMID: 40121496 PMCID: PMC11929343 DOI: 10.1186/s12937-025-01110-y] [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: 01/21/2025] [Accepted: 03/02/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND Breast cancer (BC) is the most common malignancy in women, yet the role of beverage consumption in BC risk remains unclear. Additionally, the contribution of anthropo-metabolic biomarkers as mediators is unknown, limiting the development of effective prevention strategies. METHODS This study included 13,567 participants from the Guangzhou Biobank Cohort Study (GBCS), where beverage consumption was assessed at baseline using a food frequency questionnaire. BC cases were identified through cancer registry linkage over a mean follow-up of 14.8 years. Mendelian randomization (MR) analyses were performed to evaluate the causal effects of beverage consumption on BC risk, with a two-step MR approach used to estimate mediation effects. RESULTS During follow-up, 243 BC cases were identified. Weekly consumption of ≥ 1 portion of sugar sweetened beverages (SSB), versus < 1 portion, was significantly associated with a higher risk of BC (hazard ratio [HR] 1.58, 95% confidence interval [CI] 1.12-2.23). This association was partly mediated by body mass index (proportion mediated [PM] 4.2%, 95% CI 0.9-17.1%) and uric acid (PM 18.8%, 95% CI 1.5-77.5%). Weekly consumption of > 6 portions of dairy-based milk was associated with a non-significantly higher BC risk (HR 1.41, 95% CI 0.99-2.03), while 3-6 portions of soy milk were associated with a lower BC risk (HR 0.31, 95% CI 0.10-0.98). No significant associations were found for pure fruit juice, coffee, tea, or alcoholic drinks. MR analyses supported the detrimental effect of SSB on BC risk, with high-density lipoprotein cholesterol, polyunsaturated fatty acids to total fatty acids (TFAs) ratio, and omega-6 fatty acids to TFAs ratio mediating 2.44%, 2.73%, and 3.53% of the association, respectively. CONCLUSION This study suggested that SSB consumption was a risk factor for BC and identified key anthropo-metabolic biomarkers mediating this relationship. Reducing SSB consumption and addressing associated metabolic pathways may offer effective strategies for BC prevention.
Collapse
Affiliation(s)
- Xiaoyi Lin
- School of Public Health, Sun Yat-sen University, No. 74 Zhongshan 2nd Road, Guangzhou, Guangdong Province, China
- Greater Bay Area, Greater Bay Area Public Health Research Collaboration, Guangzhou, China
| | - Boheng Liang
- Guangzhou Center for Disease Control and Prevention, Guangzhou, China
| | - Tai Hing Lam
- School of Public Health, the University of Hong Kong, Hong Kong, China
- Guangzhou Twelfth People's Hospital, Guangzhou, China
- Greater Bay Area, Greater Bay Area Public Health Research Collaboration, Guangzhou, China
| | - Kar Keung Cheng
- School of Health Sciences, College of Medicine and Health, University of Birmingham, Birmingham, UK
| | - Weisen Zhang
- Guangzhou Twelfth People's Hospital, Guangzhou, China
- Greater Bay Area, Greater Bay Area Public Health Research Collaboration, Guangzhou, China
| | - Lin Xu
- School of Public Health, Sun Yat-sen University, No. 74 Zhongshan 2nd Road, Guangzhou, Guangdong Province, China.
- School of Public Health, the University of Hong Kong, Hong Kong, China.
- School of Health Sciences, College of Medicine and Health, University of Birmingham, Birmingham, UK.
- Greater Bay Area, Greater Bay Area Public Health Research Collaboration, Guangzhou, China.
| |
Collapse
|
39
|
Sevilla-González M, Smith K, Wang N, Jensen AE, Litkowski EM, Kim H, DiCorpo DA, Hsu S, Cui J, Liu CT, Yu C, McNeil JJ, Lacaze P, Westerman KE, Chang KM, Tsao PS, Phillips LS, Goodarzi MO, Sladek R, Rotter JI, Dupuis J, Florez JC, Merino J, Meigs JB, Zhou JJ, Raghavan S, Udler MS, Manning AK. Heterogeneous effects of genetic variants and traits associated with fasting insulin on cardiometabolic outcomes. Nat Commun 2025; 16:2569. [PMID: 40089507 PMCID: PMC11910595 DOI: 10.1038/s41467-025-57452-y] [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: 04/30/2024] [Accepted: 02/21/2025] [Indexed: 03/17/2025] Open
Abstract
Elevated fasting insulin levels (FI), indicative of altered insulin secretion and sensitivity, may precede type 2 diabetes (T2D) and cardiovascular disease onset. In this study, we group FI-associated genetic variants based on their genetic and phenotypic similarities and identify seven clusters with distinct mechanisms contributing to elevated FI levels. Clusters fall into two types: "non-diabetogenic hyperinsulinemia," where clusters are not associated with increased T2D risk, and "diabetogenic hyperinsulinemia," where T2D associations are driven by body fat distribution, liver function, circulating lipids, or inflammation. In over 1.1 million multi-ancestry individuals, we demonstrated that diabetogenic hyperinsulinemia cluster-specific polygenic scores exhibit varying risks for cardiovascular conditions, including coronary artery disease, myocardial infarction (MI), and stroke. Notably, the visceral adiposity cluster shows sex-specific effects for MI risk in males without T2D. This study underscores processes that decouple elevated FI levels from T2D and cardiovascular risk, offering new avenues for investigating process-specific pathways of disease.
Collapse
Affiliation(s)
- Magdalena Sevilla-González
- Clinical and Translational Epidemiology Unit, Mongan Institute, Massachusetts General Hospital, Boston, MA, 02114, USA
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
- Programs in Metabolism and Medical & Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Kirk Smith
- Programs in Metabolism and Medical & Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Ningyuan Wang
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, 02118, USA
| | - Aubrey E Jensen
- Phoenix Veterans Affairs Medical Center, Phoenix, AZ, 85012, USA
- Department of Biostatistics, UCLA Fielding School of Public Health, Los Angeles, CA, 90095, USA
| | - Elizabeth M Litkowski
- Veterans Affairs Eastern Colorado Health Care System, Aurora, CO, 80045, USA
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Hyunkyung Kim
- Programs in Metabolism and Medical & Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Daniel A DiCorpo
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, 02118, USA
| | - Sarah Hsu
- Programs in Metabolism and Medical & Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Jinrui Cui
- Division of Endocrinology, Diabetes, and Metabolism, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Ching-Ti Liu
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, 02118, USA
| | - Chenglong Yu
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, 3004, Australia
| | - John J McNeil
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, 3004, Australia
| | - Paul Lacaze
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, 3004, Australia
| | - Kenneth E Westerman
- Clinical and Translational Epidemiology Unit, Mongan Institute, Massachusetts General Hospital, Boston, MA, 02114, USA
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
- Programs in Metabolism and Medical & Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Kyong-Mi Chang
- Corporal Michael J Crescenz VA Medical Center, Philadelphia, PA, 19104, USA
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Philip S Tsao
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Mark O Goodarzi
- Division of Endocrinology, Diabetes, and Metabolism, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Rob Sladek
- Department of Human Genetics and Department of Medicine, McGill University, Montréal, QC, Canada
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Josée Dupuis
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, 02118, USA
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montréal, QC, Canada
| | - Jose C Florez
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
- Programs in Metabolism and Medical & Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Jordi Merino
- Programs in Metabolism and Medical & Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - James B Meigs
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
- Programs in Metabolism and Medical & Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Division of General Internal Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Jin J Zhou
- Phoenix Veterans Affairs Medical Center, Phoenix, AZ, 85012, USA
- Department of Biostatistics, UCLA Fielding School of Public Health, Los Angeles, CA, 90095, USA
| | - Sridharan Raghavan
- Veterans Affairs Eastern Colorado Health Care System, Aurora, CO, 80045, USA
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Miriam S Udler
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA.
- Programs in Metabolism and Medical & Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA.
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA.
| | - Alisa K Manning
- Clinical and Translational Epidemiology Unit, Mongan Institute, Massachusetts General Hospital, Boston, MA, 02114, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA.
- Programs in Metabolism and Medical & Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA.
| |
Collapse
|
40
|
Lee Y, Seo JH, Lee J, Kim HS. Causal Effects of 25-Hydroxyvitamin D on Metabolic Syndrome and Metabolic Risk Traits: A Bidirectional Two-Sample Mendelian Randomization Study. Biomedicines 2025; 13:723. [PMID: 40149699 PMCID: PMC11940704 DOI: 10.3390/biomedicines13030723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/10/2025] [Accepted: 03/13/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objectives: Individuals with metabolic syndrome (MetS) present reduced 25(OH)D levels. We performed a two-sample Mendelian randomization (MR) study to investigate whether causal relationships exist between 25(OH)D levels and MetS/MetS risk traits, including waist circumference, body mass index (BMI), hypertension (systolic/diastolic blood pressure), triglyceride, high-density lipoprotein cholesterol, and glucose levels. Methods: We employed genetic variants related to 25(OH)D levels from the SUNLIGHT Consortium and a European genome-wide association study meta-analysis, including UK Biobank (UKB) data, as well as variants for MetS and MetS risk traits from UKB and multiple European consortia. Several MR methods were used, i.e., inverse-variance weighted, weighted median, and MR-Egger regression. Heterogeneity and horizontal pleiotropy analyses were performed to ensure the stability of candidate single-nucleotide polymorphisms (SNPs) as the instrumental variable. We first conducted univariable MR to investigate the relationship between 25(OH)D levels and MetS, including its related risk traits, and subsequently performed multivariable MR to adjust for potential confounders. Results: This study did not provide evidence of a causal relationship between 25(OH)D levels and MetS/MetS risk traits. However, we found that several risk traits of MetS, such as waist circumference, BMI, and TG, had an inverse-causal relationship with 25(OH)D levels, suggesting that 25(OH)D levels could be secondary consequences of metabolic illnesses. Conclusions: We identified no causal relationship between 25(OH)D levels and MetS/MetS risk factors. However, 25(OH)D levels may result from MetS traits.
Collapse
Affiliation(s)
- Young Lee
- Veterans Medical Research Institute, Veterans Health Service Medical Center, Seoul 05368, Republic of Korea; (Y.L.); (J.H.S.)
| | - Je Hyun Seo
- Veterans Medical Research Institute, Veterans Health Service Medical Center, Seoul 05368, Republic of Korea; (Y.L.); (J.H.S.)
| | - Junyong Lee
- Department of Family Medicine, Veterans Health Service Medical Center, Seoul 05368, Republic of Korea;
| | - Hwa Sun Kim
- Department of Family Medicine, Veterans Health Service Medical Center, Seoul 05368, Republic of Korea;
| |
Collapse
|
41
|
Arthur TD, Nguyen JP, Henson BA, D'Antonio-Chronowska A, Jaureguy J, Silva N, Panopoulos AD, Izpisua Belmonte JC, D'Antonio M, McVicker G, Frazer KA. Multiomic QTL mapping reveals phenotypic complexity of GWAS loci and prioritizes putative causal variants. CELL GENOMICS 2025; 5:100775. [PMID: 39986281 PMCID: PMC11960542 DOI: 10.1016/j.xgen.2025.100775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 10/18/2024] [Accepted: 01/24/2025] [Indexed: 02/24/2025]
Abstract
Most GWAS loci are presumed to affect gene regulation; however, only ∼43% colocalize with expression quantitative trait loci (eQTLs). To address this colocalization gap, we map eQTLs, chromatin accessibility QTLs (caQTLs), and histone acetylation QTLs (haQTLs) using molecular samples from three early developmental-like tissues. Through colocalization, we annotate 10.4% (n = 540) of GWAS loci in 15 traits by QTL phenotype, temporal specificity, and complexity. We show that integration of chromatin QTLs results in a 2.3-fold higher annotation rate of GWAS loci because they capture distal GWAS loci missed by eQTLs, and that 5.4% (n = 13) of GWAS colocalizing eQTLs are early developmental specific. Finally, we utilize the iPSCORE multiomic QTLs to prioritize putative causal variants overlapping transcription factor motifs to elucidate the potential genetic underpinnings of 296 GWAS-QTL colocalizations.
Collapse
Affiliation(s)
- Timothy D Arthur
- Biomedical Sciences Program, University of California, San Diego, La Jolla, CA 92093, USA; Department of Biomedical Informatics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jennifer P Nguyen
- Department of Biomedical Informatics, University of California, San Diego, La Jolla, CA 92093, USA; Bioinformatics and Systems Biology Graduate Program, University of California, San Diego, La Jolla, CA 92093, USA
| | - Benjamin A Henson
- Institute of Genomic Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | | | - Jeffrey Jaureguy
- Bioinformatics and Systems Biology Graduate Program, University of California, San Diego, La Jolla, CA 92093, USA; Integrative Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Nayara Silva
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Athanasia D Panopoulos
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | | | - Matteo D'Antonio
- Department of Biomedical Informatics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Graham McVicker
- Integrative Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Kelly A Frazer
- Institute of Genomic Medicine, University of California San Diego, La Jolla, CA 92093, USA; Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
42
|
He Y, Wei Y, Liang H, Wan Y, Zhang Y, Zhang J. Causal association between metabolic syndrome and ovarian dysfunction: a bidirectional two-sample mendelian randomization. J Ovarian Res 2025; 18:50. [PMID: 40069881 PMCID: PMC11895234 DOI: 10.1186/s13048-025-01614-5] [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: 11/05/2024] [Accepted: 01/31/2025] [Indexed: 03/15/2025] Open
Abstract
BACKGROUND The relationship between Metabolic Syndrome (MetS) and ovarian dysfunction has been widely reported in observational studies, yet it remains not fully understood. This study employs genetic prediction methods and utilizes summary data from genome-wide association studies (GWAS) to investigate this causal link. METHODS We employed a bidirectional two-sample Mendelian Randomization (MR) analysis utilizing MetS and ovarian dysfunction summary data from GWAS. Inverse variance weighted (IVW) was employed as the primary MR method, supplemented by Weighted Median, Weighted Mode, and MR-Egger methods. The robustness of the results was further assessed through sensitivity analyses including MR-Egger regression, MR-PRESSO, Cochran's Q, and leave-one-out test. RESULTS Our MR analysis identified a causal relationship between genetically determined insulin resistance (OR = 0.26, 95% CI: 0.08-0.89, P = 0.03), waist circumference (OR = 2.14, 95% CI: 1.45-3.15, P < 0.001), BMI (OR = 2.1, 95% CI: 1.56-2.83, P < 0.001) and ovarian dysfunction. Conversely, reverse MR analysis confirmed causal effects of ovarian dysfunction on metabolic syndrome (OR = 0.98, 95% CI: 0.97-0.99, P < 0.001) and waist circumference (OR = 0.99, 95% CI: 0.98-0.99, P = 0.02). The results of MR-Egger regression test indicated that the whole analysis was not affected by horizontal pleiotropy. Additionally, the MR-PRESSO test identified outliers in SNPs, but after removal of outliers, results remained unchanged. CONCLUSION This study reveals a bidirectional causal connection between metabolic syndrome and ovarian dysfunction via genetic prediction methods. These findings are crucial for advancing our understanding of the interactions between these conditions and developing strategies for prevention and treatment.
Collapse
Affiliation(s)
- Ying He
- Department of Obstetrics and Gynecology, Xijing 986 Hospital Department, Air force Medical University, No. 6 Jianshe West Road, Xi'an, 710054, Shaanxi, China
| | - Yanling Wei
- Department of Obstetrics and Gynecology, Xijing Hospital, Air force Medical University, No. 15 Changle West Road, Xi'an, 710033, Shaanxi, China
| | - Haixia Liang
- Department of Obstetrics and Gynecology, Xijing 986 Hospital Department, Air force Medical University, No. 6 Jianshe West Road, Xi'an, 710054, Shaanxi, China
| | - Yi Wan
- Department of Health Service, Air force Medical University, Xi'an, 710032, Shaanxi, China
| | - Ying Zhang
- Department of Obstetrics and Gynecology, Xijing 986 Hospital Department, Air force Medical University, No. 6 Jianshe West Road, Xi'an, 710054, Shaanxi, China.
| | - Jianfang Zhang
- Department of Obstetrics and Gynecology, Xijing Hospital, Air force Medical University, No. 15 Changle West Road, Xi'an, 710033, Shaanxi, China.
| |
Collapse
|
43
|
Jiang W, Ding K, Yang M, Hu Z, Yue R. Exploring the Potential Effect of GLP1R Agonism on Common Aging-Related Diseases via Glucose Reduction: A Mendelian Randomization Study. J Gerontol A Biol Sci Med Sci 2025; 80:glaf007. [PMID: 39797952 DOI: 10.1093/gerona/glaf007] [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: 09/09/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND Glucagon-like peptide-1 receptor agonists (GLP1RAs) are widely used in managing type 2 diabetes mellitus and weight control. Their potential in treating aging-related diseases has been gaining attention in recent years. However, the long-term effects of GLP1RAs on these diseases have yet to be fully revealed. METHODS Using a genetic variant in the GLP1R gene to model the long-term effects of GLP1RAs, this Mendelian randomization (MR) study systematically explored potential causal associations between GLP1R agonism and 12 aging-related diseases and indicators. Genetic summary data sets used in this study were obtained from previous genome-wide association studies. RESULTS The primary MR analysis results suggested that GLP1R agonism was potentially positively causally associated with appendicular lean mass (Beta = 0.246, 95% confidence interval [CI] = 0.096-0.396), whole-body fat-free mass (Beta = 0.202, 95% CI = 0.048-0.355), and lung function (forced vital capacity [FVC]; Beta = 0.179, 95% CI = 0.152-0.205; p < .05). Additionally, a potential negative causal association was observed with myocardial infarction (odds ratio = 0.430, 95% CI = 0.249-0.745; p < .05). CONCLUSIONS The present MR study provides exploratory evidence suggesting potential causal associations between GLP1R agonism and appendicular lean mass, whole-body fat-free mass, lung function (FVC), and myocardial infarction. Given the exploratory nature of these findings and the limitations of the MR methodology, further research is needed to validate these results and investigate the underlying biological mechanisms.
Collapse
Affiliation(s)
- Wei Jiang
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Clinical Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Kaixi Ding
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Maoyi Yang
- Department of Clinical Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhipeng Hu
- Department of Clinical Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Rensong Yue
- Department of Clinical Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
44
|
Reed ZE, Sallis HM, Richmond RC, Attwood AS, Lawlor DA, Munafò MR. Investigating whether smoking and alcohol behaviours influence risk of type 2 diabetes using a Mendelian randomisation study. Sci Rep 2025; 15:7985. [PMID: 40055374 PMCID: PMC11889105 DOI: 10.1038/s41598-025-90437-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 02/13/2025] [Indexed: 03/15/2025] Open
Abstract
Previous studies suggest that smoking and higher alcohol consumption are associated with greater type 2 diabetes (T2D) risk. However, studies examining whether this reflects causal relationships are limited and often do not consider continuous glycaemic traits. We conducted both two-sample and one-sample Mendelian randomisation (MR), using publicly available GWAS data and UK Biobank data, respectively, to examine the potential causal effects of lifetime smoking index (LSI) and alcoholic drinks per week (DPW) on T2D and continuous traits (fasting glucose, fasting insulin and glycated haemoglobin, HbA1c). Two-sample MR results suggested possible causal effects of higher LSI on T2D risk (OR per 1SD higher LSI: 1.42, 95% CI 1.22 to 1.64); however, sensitivity analyses did not consistently support this finding. There was no robust evidence that higher DPW influenced T2D risk (OR per 1 SD higher log-transformed DPW: 1.04, 95% CI 0.40 to 2.65). There was evidence of a potential causal effect on higher fasting glucose (difference in mean fasting glucose in mmol/l per 1SD higher log-transformed DPW: 0.34, 95% CI 0.09 to 0.59), though, this was attenuated when accounting for body mass index (BMI), suggesting BMI confounding might explain the potential effect. One-sample MR results suggested a possible causal effect of higher DPW on T2D risk (OR per 1 SD higher log-transformed DPW: 1.71, 95% CI 1.24 to 2.36), but lower HbA1c levels (difference in mean SD of log transformed HbA1c (mmol/mol) per 1 SD higher log-transformed DPW: -0.07, 95% CI -0.11 to -0.02). Our results suggest effective public health interventions to prevent and/or reduce smoking and alcohol consumption are unlikely to reduce T2D prevalence.
Collapse
Affiliation(s)
- Zoe E Reed
- School of Psychological Science, University of Bristol, Bristol, UK.
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK.
| | - Hannah M Sallis
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- Centre for Academic Mental Health, Bristol Medical School, University of Bristol, Bristol, UK
| | - Rebecca C Richmond
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Angela S Attwood
- School of Psychological Science, University of Bristol, Bristol, UK
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Deborah A Lawlor
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Marcus R Munafò
- School of Psychological Science, University of Bristol, Bristol, UK
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- National Institute for Health Research Bristol Biomedical Research Centre, University Hospitals Bristol NHS Foundation Trust and University of Bristol, Bristol, UK
| |
Collapse
|
45
|
Fu S, Li Q, Cheng L, Wan S, Wang Q, Min Y, Xie Y, Liu H, Hu T, Liu H, Chen W, Zhang Y, Xiong F. Causal Relationship Between Intelligence, Noncognitive Education, Cognition and Urinary Tract or Kidney Infection: A Mendelian Randomization Study. Int J Nephrol Renovasc Dis 2025; 18:71-85. [PMID: 40070673 PMCID: PMC11895678 DOI: 10.2147/ijnrd.s511736] [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] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
Background The occurrence of urinary tract or kidney infection is correlated with intelligence, noncognitive education and cognition, but the causal relationship between them remains uncertain, and which risk factors mediate this causal relationship remains unknown. Methods The intelligence (n=269,867), noncognitive education (n=510,795) and cognition data (n=257,700) were obtained from genome-wide association studies (GWAS) conducted in individuals of European ethnicities. The genetic associations between these factors and urinary tract or kidney infection (UK Biobank, n=397,867) were analyzed using linkage disequilibrium score regression. We employed a two-sample univariate and multivariate Mendelian randomization to evaluate the causal relationship, and utilized a two-step Mendelian randomization to examine the involvement of 28 potential mediators and their respective mediating proportions. Results The genetic correlation coefficients of intelligence, noncognitive education, cognition, and urinary tract or kidney infection were -0.338, -0.218, and -0.330. The Mendelian randomization using the inverse variance weighted method revealed each 1-SD increase in intelligence, the risk of infection decreased by 15.9%, while after adjusting for noncognitive education, the risk decreased by 20%. For each 1-SD increase in noncognitive education, the risk of infection decreased by 8%, which further reduced to 7.1% after adjusting for intelligence and to 6.7% after adjusting for cognition. For each 1-SD increase in cognition, the risk of infection decreased by 10.8%, increasing to 11.9% after adjusting for noncognitive education. The effects of intelligence and cognition are interdependent. 2 out of 28 potential mediating factors exhibited significant mediation effects in the causal relationship between noncognitive education and urinary tract or kidney infection, with body mass index accounting for 12.1% of the mediation effect and smoking initiation accounting for 14.7%. Conclusion Enhancing intelligence, noncognitive education, and cognition can mitigate the susceptibility to urinary tract or kidney infection. Noncognitive education exhibited independent effect, while body mass index and smoking initiation assuming a mediating role.
Collapse
Affiliation(s)
- Shuai Fu
- Department of Nephrology, Wuhan No. 1 Hospital, Wuhan, Hubei Province, People’s Republic of China
| | - Qiang Li
- Department of Nephrology, Wuhan No. 1 Hospital, Wuhan, Hubei Province, People’s Republic of China
| | - Li Cheng
- Department of Nephrology, Wuhan No. 1 Hospital, Wuhan, Hubei Province, People’s Republic of China
| | - Sheng Wan
- Department of Nephrology, Wuhan No. 1 Hospital, Wuhan, Hubei Province, People’s Republic of China
| | - Quan Wang
- Department of Nephrology, Wuhan No. 1 Hospital, Wuhan, Hubei Province, People’s Republic of China
| | - Yonglong Min
- Department of Nephrology, Wuhan No. 1 Hospital, Wuhan, Hubei Province, People’s Republic of China
| | - Yanghao Xie
- Department of Nephrology, Wuhan No. 1 Hospital, Wuhan, Hubei Province, People’s Republic of China
| | - Huizhen Liu
- Department of Nephrology, Wuhan No. 1 Hospital, Wuhan, Hubei Province, People’s Republic of China
| | - Taotao Hu
- Department of Nephrology, Wuhan No. 1 Hospital, Wuhan, Hubei Province, People’s Republic of China
| | - Hong Liu
- Department of Nephrology, Wuhan No. 1 Hospital, Wuhan, Hubei Province, People’s Republic of China
| | - Weidong Chen
- Department of Nephrology, Wuhan No. 1 Hospital, Wuhan, Hubei Province, People’s Republic of China
| | - Yanmin Zhang
- Department of Nephrology, Wuhan No. 1 Hospital, Wuhan, Hubei Province, People’s Republic of China
| | - Fei Xiong
- Department of Nephrology, Wuhan No. 1 Hospital, Wuhan, Hubei Province, People’s Republic of China
| |
Collapse
|
46
|
Dou C, Liu D, Kong L, Chen M, Ye C, Zhu Z, Zheng J, Xu M, Xu Y, Li M, Zhao Z, Lu J, Chen Y, Ning G, Wang W, Bi Y, Wang T. Shared genetic architecture of type 2 diabetes with muscle mass and function and frailty reveals comorbidity etiology and pleiotropic druggable targets. Metabolism 2025; 164:156112. [PMID: 39710002 DOI: 10.1016/j.metabol.2024.156112] [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: 10/08/2024] [Revised: 12/19/2024] [Accepted: 12/19/2024] [Indexed: 12/24/2024]
Abstract
BACKGROUND Delineating the shared genetic architecture of type 2 diabetes with muscle mass and function and frailty is essential for unraveling the common etiology and developing holistic therapeutic strategies for these co-existing conditions. METHODS In this genome-wide pleiotropic association study, we performed multi-level pairwise trait pleiotropic analyses using genome-wide association study summary statistics from up to 461,026 European ancestry individuals to dissect the shared genetic factors and causal relationships of type 2 diabetes and seven glycemic traits with four muscle mass- and function-related phenotypes and the frailty index. RESULTS We first identified 27 pairs with significant genetic correlations through the linkage disequilibrium score regression and high-definition likelihood analysis. Then we determined 79 pleiotropic loci and 109 pleiotropic genes across linkage pairs via the pleiotropic analysis under the composite null hypothesis (PLACO), the colocalization, and the Multi-marker Analysis of GenoMic Annotation (MAGMA) analyses. We subsequently performed transcriptome-wide association study (TWAS) analyses using joint-tissue imputation, refined by gene-based integrative fine-mapping through a conditional TWAS approach, and identified 44 unique causal shared genes across 13 tissues in linkage pairs, including eight druggable genes (ABO, AOC1, FTO, GCKR, MTOR, POLK, PPARG, and APEH), with MTOR and PPARG categorized as clinically actionable. Two-sample Mendelian randomization analysis supported bidirectional causality between diabetes and frailty index and unidirectional causal effects of muscle phenotypes on glycemic profiles. CONCLUSIONS Our findings highlight the common genetic underpinnings between type 2 diabetes and muscle loss and frailty and inform drug targets with pleiotropic effects on both of these aging-related challenges.
Collapse
Affiliation(s)
- Chun Dou
- 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, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dong Liu
- 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, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lijie Kong
- 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, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mingling Chen
- 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, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chaojie Ye
- 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, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zheng Zhu
- 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, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Zheng
- 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, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Xu
- 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, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Xu
- 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, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mian Li
- 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, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhiyun Zhao
- 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, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jieli Lu
- 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, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuhong Chen
- 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, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guang Ning
- 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, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiqing Wang
- 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, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yufang Bi
- 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, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tiange Wang
- 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, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
47
|
Hu Y, Cui X, Lu M, Guan X, Li Y, Zhang L, Lin L, Zhang Z, Zhang M, Hao J, Wang X, Huan J, Li Y, Li C. Body Fat Distribution and Ectopic Fat Accumulation as Mediator of Diabetogenic Action of Lipid-Modifying Drugs: A Mediation Mendelian Randomization Study. Mayo Clin Proc 2025; 100:424-439. [PMID: 39918451 DOI: 10.1016/j.mayocp.2024.10.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 10/06/2024] [Accepted: 10/25/2024] [Indexed: 05/08/2025]
Abstract
OBJECTIVE To investigate the causal relationship between various lipid-modifying drugs and new-onset diabetes, as well as the mediators contributing to this relationship. METHODS Mediation Mendelian randomization was performed to investigate the causal effect of lipid-modifying drug targets on type 2 diabetes (T2D) outcomes and the proportion of this association that is mediated through ectopic fat accumulation traits. Specific sets of variants in or near genes that encode 11 lipid-modifying drug targets (LDLR, HMGCR, NPC1L1, PCSK9, APOB, ABCG5/ABCG8, LPL, PPARA, ANGPTL3, APOC3, and CETP; for expansion of gene symbols, use search tool at www.genenames.org) were extracted. Random effects inverse variance weighted were performed to evaluate the causal effects among outcomes. Mediation analyses were performed to identify the mediators of the association between lipid-modifying drugs and T2D. The study was conducted from November 10, 2023, to April 2, 2024 RESULTS: The genetic mimicry of HMGCR and APOB inhibition was associated with an increased T2D risk, whereas the genetic mimicry of LPL enhancement was linked to a lower T2D risk. Gluteofemoral adipose tissue volume was a mediator for explaining 9.52% (P=.002), 16.90% (P=.03), and 10.50% (P=.003) of the total effect of HMGCR, APOB, and LPL on T2D susceptibility, respectively. Liver fat was a mediator for explaining 21.12% (P=.005), 12.28% (P=.03), and 9.84% (P=.005) of the total effect of HMGCR, APOB, and LPL on T2D susceptibility, respectively. CONCLUSION Our findings support the hypothesis that liver fat and gluteofemoral adipose tissue play a mediating role in the prodiabetic effects of HMGCR and APOB inhibition, as well as in the antidiabetic effects of LPL enhancement.
Collapse
Affiliation(s)
- Yuanlong Hu
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Xinhai Cui
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Mengkai Lu
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Xiuya Guan
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yuan Li
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Lei Zhang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Lin Lin
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Zhiyuan Zhang
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Muxin Zhang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Jiaqi Hao
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Xiaojie Wang
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macau, China
| | - Jiaming Huan
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yunlun Li
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China; Department of Cardiovascular, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China.
| | - Chao Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China.
| |
Collapse
|
48
|
Zheng SL, Jurgens SJ, McGurk KA, Xu X, Grace C, Theotokis PI, Buchan RJ, Francis C, de Marvao A, Curran L, Bai W, Pua CJ, Tang HC, Jorda P, van Slegtenhorst MA, Verhagen JMA, Harper AR, Ormondroyd E, Chin CWL, Pantazis A, Baksi J, Halliday BP, Matthews P, Pinto YM, Walsh R, Amin AS, Wilde AAM, Cook SA, Prasad SK, Barton PJR, O'Regan DP, Lumbers RT, Goel A, Tadros R, Michels M, Watkins H, Bezzina CR, Ware JS. Evaluation of polygenic scores for hypertrophic cardiomyopathy in the general population and across clinical settings. Nat Genet 2025; 57:563-571. [PMID: 39966645 PMCID: PMC11906360 DOI: 10.1038/s41588-025-02094-5] [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: 03/30/2023] [Accepted: 01/21/2025] [Indexed: 02/20/2025]
Abstract
Hypertrophic cardiomyopathy (HCM) is an important cause of morbidity and mortality, with pathogenic variants found in about a third of cases. Large-scale genome-wide association studies (GWAS) demonstrate that common genetic variation contributes to HCM risk. Here we derive polygenic scores (PGS) from HCM GWAS and genetically correlated traits and test their performance in the UK Biobank, 100,000 Genomes Project, and clinical cohorts. We show that higher PGS significantly increases the risk of HCM in the general population, particularly among pathogenic variant carriers, where HCM penetrance differs 10-fold between those in the highest and lowest PGS quintiles. Among relatives of HCM probands, PGS stratifies risks of developing HCM and adverse outcomes. Finally, among HCM cases, PGS strongly predicts the risk of adverse outcomes and death. These findings support the broad utility of PGS across clinical settings, enabling tailored screening and surveillance and stratification of risk of adverse outcomes.
Collapse
Affiliation(s)
- Sean L Zheng
- National Heart Lung Institute, Imperial College London, London, UK
- Medical Research Council Laboratory of Medical Sciences, Imperial College London, London, UK
- Royal Brompton & Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Sean J Jurgens
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kathryn A McGurk
- National Heart Lung Institute, Imperial College London, London, UK
- Medical Research Council Laboratory of Medical Sciences, Imperial College London, London, UK
| | - Xiao Xu
- National Heart Lung Institute, Imperial College London, London, UK
- Medical Research Council Laboratory of Medical Sciences, Imperial College London, London, UK
| | - Chris Grace
- Radcliffe Department of Medicine, University of Oxford, Division of Cardiovascular Medicine, John Radcliffe Hospital, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Pantazis I Theotokis
- National Heart Lung Institute, Imperial College London, London, UK
- Medical Research Council Laboratory of Medical Sciences, Imperial College London, London, UK
- Royal Brompton & Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Rachel J Buchan
- National Heart Lung Institute, Imperial College London, London, UK
- Medical Research Council Laboratory of Medical Sciences, Imperial College London, London, UK
- Royal Brompton & Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Catherine Francis
- National Heart Lung Institute, Imperial College London, London, UK
- Royal Brompton & Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Antonio de Marvao
- National Heart Lung Institute, Imperial College London, London, UK
- Medical Research Council Laboratory of Medical Sciences, Imperial College London, London, UK
- Department of Women and Children's Health, King's College London, London, UK
- School of Cardiovascular and Metabolic Medicine and Sciences, King's College London, London, UK
| | - Lara Curran
- National Heart Lung Institute, Imperial College London, London, UK
- Medical Research Council Laboratory of Medical Sciences, Imperial College London, London, UK
- Royal Brompton & Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Wenjia Bai
- Biomedical Image Analysis Group, Department of Computing, Imperial College London, London, UK
- Department of Brain Sciences, Imperial College London, London, UK
| | - Chee Jian Pua
- National Heart Research Institute Singapore, National Heart Center, Singapore, Singapore
| | - Hak Chiaw Tang
- Department of Cardiology, National Heart Centre, Singapore, Singapore
| | - Paloma Jorda
- Cardiovascular Genetics Centre, Montreal Heart Institute, Montreal, Quebec, Canada
- Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Marjon A van Slegtenhorst
- Department of Clinical Genetics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Judith M A Verhagen
- Department of Clinical Genetics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Andrew R Harper
- Radcliffe Department of Medicine, University of Oxford, Division of Cardiovascular Medicine, John Radcliffe Hospital, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Elizabeth Ormondroyd
- Radcliffe Department of Medicine, University of Oxford, Division of Cardiovascular Medicine, John Radcliffe Hospital, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Calvin W L Chin
- Department of Cardiology, National Heart Centre, Singapore, Singapore
| | - Antonis Pantazis
- Royal Brompton & Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - John Baksi
- National Heart Lung Institute, Imperial College London, London, UK
- Royal Brompton & Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Brian P Halliday
- National Heart Lung Institute, Imperial College London, London, UK
- Royal Brompton & Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Paul Matthews
- Department of Brain Sciences, Imperial College London, London, UK
| | - Yigal M Pinto
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- Department of Clinical Cardiology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart, Paris, France
| | - Roddy Walsh
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Ahmad S Amin
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- Department of Clinical Cardiology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart, Paris, France
| | - Arthur A M Wilde
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- Department of Clinical Cardiology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart, Paris, France
| | - Stuart A Cook
- Medical Research Council Laboratory of Medical Sciences, Imperial College London, London, UK
- Department of Cardiology, National Heart Centre, Singapore, Singapore
- Duke-National University of Singapore Medical School, Singapore, Singapore
| | - Sanjay K Prasad
- National Heart Lung Institute, Imperial College London, London, UK
- Royal Brompton & Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Paul J R Barton
- National Heart Lung Institute, Imperial College London, London, UK
- Medical Research Council Laboratory of Medical Sciences, Imperial College London, London, UK
- Royal Brompton & Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Declan P O'Regan
- Medical Research Council Laboratory of Medical Sciences, Imperial College London, London, UK
| | - R T Lumbers
- Institute of Health Informatics, University College London, London, UK
- Health Data Research UK London, University College London, London, UK
- British Heart Foundation Research Accelerator, University College London, London, UK
| | - Anuj Goel
- Radcliffe Department of Medicine, University of Oxford, Division of Cardiovascular Medicine, John Radcliffe Hospital, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Rafik Tadros
- Cardiovascular Genetics Centre, Montreal Heart Institute, Montreal, Quebec, Canada
- Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Michelle Michels
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart, Paris, France
- Department of Cardiology, Thorax Center, Cardiovascular Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Hugh Watkins
- Radcliffe Department of Medicine, University of Oxford, Division of Cardiovascular Medicine, John Radcliffe Hospital, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Connie R Bezzina
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart, Paris, France
| | - James S Ware
- National Heart Lung Institute, Imperial College London, London, UK.
- Medical Research Council Laboratory of Medical Sciences, Imperial College London, London, UK.
- Royal Brompton & Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK.
- Imperial College Healthcare NHS Trust, London, UK.
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
49
|
Li Y, Zhao K. Two-Sample Bidirectional Mendelian Randomization Study With Causal Association Between Metabolic Syndrome and Cerebral Aneurysm. Brain Behav 2025; 15:e70396. [PMID: 40038846 PMCID: PMC11879889 DOI: 10.1002/brb3.70396] [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: 11/12/2024] [Revised: 01/23/2025] [Accepted: 02/17/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND We used a two-sample mendelian randomization (MR) method to comprehensively investigate the causality of metabolic syndrome (MetS) or its components, including MetS, triglyceride (TG), high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDL-C), fasting blood glucose (FBG), waist circumference (WC), and hypertension (HP), with cerebral aneurysm including nonruptured and ruptured aneurysmal subarachnoid hemorrhage (SAH). METHODS By leveraging large-scale genome-wide association study (GWAS) summary statistics of MetS or its components and cerebral aneurysm (nonruptured and SAH) from European, MR, reverse-direction MR, and sensitivity analysis were utilized to quantify the genetic correlations and causal relationships. In addition, we adjusted for multiple comparisons using the false discovery rate (FDR) correction. RESULTS Two-sample MR analysis showed that HP was a risk factor for cerebral aneurysm (nonruptured and SAH) with odds ratio (OR) of 58.959 (95% confidence interval [95% CI] = 12.073-287.920, p < 0.001, q < 0.001), and 32.290 (95% CI = 5.615-185.671, p < 0.001, q < 0.001), respectively. HDL-C (OR = 0.836, 95% CI = 0.728-0.960, p = 0.011, q = 0.039) and FBG (OR = 0.626, 95% CI = 0.426-0.919, p = 0.017, q = 0.039) were negatively correlated with cerebral aneurysm (nonruptured). The HDL-C result was inconsistent after adjusting for TG and LDL-C by multivariable MR analysis. In reverse MR analysis, we found that there was no statistical causal association between cerebral aneurysm (nonruptured) and MetS or its components. Genetic liability to cerebral aneurysm (SAH) was inversely associated with HDL-C and FBG but was not associated with others, however, sensitivity analysis showed that few instrumental variables made a big difference. CONCLUSIONS Genetically determined elevated FBG level reduces the risk of cerebral aneurysm (nonruptured). However, hypertension increases the risk of cerebral aneurysm (nonruptured and SAH).
Collapse
Affiliation(s)
- Yu Li
- Department of Neurosurgery, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Kai Zhao
- Department of Neurosurgery, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
50
|
Martin SS, Aday AW, Allen NB, Almarzooq ZI, Anderson CAM, Arora P, Avery CL, Baker-Smith CM, Bansal N, Beaton AZ, Commodore-Mensah Y, Currie ME, Elkind MSV, Fan W, Generoso G, Gibbs BB, Heard DG, Hiremath S, Johansen MC, Kazi DS, Ko D, Leppert MH, Magnani JW, Michos ED, Mussolino ME, Parikh NI, Perman SM, Rezk-Hanna M, Roth GA, Shah NS, Springer MV, St-Onge MP, Thacker EL, Urbut SM, Van Spall HGC, Voeks JH, Whelton SP, Wong ND, Wong SS, Yaffe K, Palaniappan LP. 2025 Heart Disease and Stroke Statistics: A Report of US and Global Data From the American Heart Association. Circulation 2025; 151:e41-e660. [PMID: 39866113 DOI: 10.1161/cir.0000000000001303] [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] [Indexed: 01/28/2025]
Abstract
BACKGROUND The American Heart Association (AHA), in conjunction with the National Institutes of Health, annually reports the most up-to-date statistics related to heart disease, stroke, and cardiovascular risk factors, including core health behaviors (smoking, physical activity, nutrition, sleep, and obesity) and health factors (cholesterol, blood pressure, glucose control, and metabolic syndrome) that contribute to cardiovascular health. The AHA Heart Disease and Stroke Statistical Update presents the latest data on a range of major clinical heart and circulatory disease conditions (including stroke, brain health, complications of pregnancy, kidney disease, congenital heart disease, rhythm disorders, sudden cardiac arrest, subclinical atherosclerosis, coronary heart disease, cardiomyopathy, heart failure, valvular disease, venous thromboembolism, and peripheral artery disease) and the associated outcomes (including quality of care, procedures, and economic costs). METHODS The AHA, through its Epidemiology and Prevention Statistics Committee, continuously monitors and evaluates sources of data on heart disease and stroke in the United States and globally to provide the most current information available in the annual Statistical Update with review of published literature through the year before writing. The 2025 AHA Statistical Update is the product of a full year's worth of effort in 2024 by dedicated volunteer clinicians and scientists, committed government professionals, and AHA staff members. This year's edition includes a continued focus on health equity across several key domains and enhanced global data that reflect improved methods and incorporation of ≈3000 new data sources since last year's Statistical Update. RESULTS Each of the chapters in the Statistical Update focuses on a different topic related to heart disease and stroke statistics. CONCLUSIONS The Statistical Update represents a critical resource for the lay public, policymakers, media professionals, clinicians, health care administrators, researchers, health advocates, and others seeking the best available data on these factors and conditions.
Collapse
|