1
|
Naeini MK, Cecelja M, Freidin MB, Smith IG, Hysi P, Nielsen CS, Williams FMK. Chronic widespread musculoskeletal pain shares a highly heritable latent pathway with atherosclerosis and arterial stiffness. Pain 2025; 166:1425-1435. [PMID: 39620366 PMCID: PMC12067610 DOI: 10.1097/j.pain.0000000000003486] [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: 06/18/2024] [Revised: 09/27/2024] [Accepted: 10/10/2024] [Indexed: 05/14/2025]
Abstract
ABSTRACT Chronic widespread pain (CWP) is prevalent and associated with reduced life expectancy. Cardiovascular disease is one possible mechanism for this. The purpose of this study was to examine the association of CWP with arterial stiffness and carotid plaque measured using ultrasound to determine if shared environmental or genetic factors might account for any observed association. Around 3000 participants from the TwinsUK with CWP information and measures of carotid-femoral pulse wave velocity (cfPWV), carotid intima-media thickness (cIMT), and plaque were considered. The relationship between CWP and cfPWV, cIMT, and plaque was determined. UK Biobank data were used to replicate the association. Cholesky decomposition and multivariate pathway twin models were examined. Using a 2-sample Mendelian randomisation approach, the causal association between CWP and coronary artery disease was assessed. TwinsUK participants demonstrated a significant association between CWP and increased cfPWV consistent with arterial stiffening (OR = 1.35, P -value = 0.012), as well as the presence of carotid plaque (OR = 1.45, P -value = 0.8e-5). The twin modelling showed a common latent component and pathway underlying CWP, cfPWV, and carotid plaque, with genetic factors accounting for 68% and 90% of the latent factor variation, respectively. The 2-sample MR revealed a potential causal association between CWP and coronary artery disease. This study found that those with CWP have increased the risk of arterial stiffness and atherosclerosis and suggests that CWP leads to an increased risk of cardiovascular disease through genetic factors.
Collapse
Affiliation(s)
- Maryam Kazemi Naeini
- Department of Twin Research and Genetic Epidemiology, School of Life Course and Population Sciences, King's College London, London, United Kingdom
| | - Marina Cecelja
- Department of Twin Research and Genetic Epidemiology, School of Life Course and Population Sciences, King's College London, London, United Kingdom
| | - Maxim B. Freidin
- Department of Twin Research and Genetic Epidemiology, School of Life Course and Population Sciences, King's College London, London, United Kingdom
- Department of Biology, School of Biological and Behavioural Sciences, Queen Mary University of London, London, United Kingdom
| | - Isabelle Granville Smith
- Department of Twin Research and Genetic Epidemiology, School of Life Course and Population Sciences, King's College London, London, United Kingdom
| | - Pirro Hysi
- Department of Twin Research and Genetic Epidemiology, School of Life Course and Population Sciences, King's College London, London, United Kingdom
| | - Christopher Sivert Nielsen
- Division of Mental and Physical Health, Norwegian Institute of Public Health, Oslo, Norway
- Division of Emergencies and Critical Care, Department of Pain Management and Research, Oslo University Hospital, Oslo, Norway
| | - Frances M. K. Williams
- Department of Twin Research and Genetic Epidemiology, School of Life Course and Population Sciences, King's College London, London, United Kingdom
| |
Collapse
|
2
|
Lerga-Jaso J, Terpolovsky A, Novković B, Osama A, Manson C, Bohn S, De Marino A, Kunitomi M, Yazdi PG. Optimization of multi-ancestry polygenic risk score disease prediction models. Sci Rep 2025; 15:17495. [PMID: 40394127 PMCID: PMC12092622 DOI: 10.1038/s41598-025-02903-1] [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: 05/16/2025] [Indexed: 05/22/2025] Open
Abstract
Polygenic risk scores (PRS) have ushered in a new era in genetic epidemiology, offering insights into individual predispositions to a wide range of diseases. However, despite recent marked enhancements in predictive power, PRS-based models still need to overcome several hurdles before they can be broadly applied in the clinic. Chiefly, they need to achieve sufficient accuracy, easy interpretability and portability across diverse populations. Leveraging trans-ancestry genome-wide association study (GWAS) meta-analysis, we generated novel, diverse summary statistics for 30 medically-related traits and benchmarked the performance of six existing PRS algorithms using UK Biobank. We built an ensemble model using logistic regression to combine outputs of top-performing algorithms and validated it on the diverse eMERGE and PAGE MEC cohorts. It surpassed current state-of-the-art PRS models, with minimal performance drops in external cohorts, indicating good calibration. To enhance predictive accuracy for clinical application, we incorporated easily-accessible clinical characteristics such as age, gender, ancestry and risk factors, creating disease prediction models intended as prospective diagnostic tests, with easily interpretable positive or negative outcomes. After adding clinical characteristics, 12 out of 30 models surpassed 80% AUC. Further, 25 traits exceeded the diagnostic odds ratio (DOR) of five, and 19 traits exceeded DOR of 10 for all ancestry groups, indicating high predictive value. Our PRS model for coronary artery disease identified 55-80 times more true coronary events than rare pathogenic variant models, reinforcing its clinical potential. The polygenic component modulated the effect of high-risk rare variants, stressing the need to consider all genetic components in clinical settings. These findings show that newly developed PRS-based disease prediction models have sufficient accuracy and portability to warrant consideration of being used in the clinic.
Collapse
Affiliation(s)
| | | | | | - Alex Osama
- Research & Development, Omics Edge, Miami, FL, USA
| | | | - Sandra Bohn
- Research & Development, Omics Edge, Miami, FL, USA
| | | | | | - Puya G Yazdi
- Research & Development, Omics Edge, Miami, FL, USA.
| |
Collapse
|
3
|
Xu C, Yang ML, Kho PF, Clarke SL, Tcheandjieu C, Peyser PA, Fann CSJ, Chen SP, VA Million Veteran Program, Saw J, Zhou X, Assimes TL, Ganesh SK. Cross-Ancestry Associations of Spontaneous Coronary Artery Dissection Genetic Risk With Coronary Atherosclerosis and Migraine Headache. J Am Heart Assoc 2025; 14:e036525. [PMID: 40357661 DOI: 10.1161/jaha.124.036525] [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: 05/10/2024] [Accepted: 03/12/2025] [Indexed: 05/15/2025]
Abstract
BACKGROUND Research studies of spontaneous coronary artery dissection (SCAD) have been primarily focused on European-ancestry individuals, with limited recognition and investigation in non-European-ancestry individuals. While SCAD has not been well ascertained in non-European-ancestry groups, pleiotropic associated traits identified in those of European ancestry have been assessed in individuals of other ancestries. Whether these traits are associated with the complex genetic architecture of SCAD in those of non-European ancestry has not been previously investigated. METHODS We investigated the associations of an established SCAD polygenic score with multiple vascular diseases in ≈900 000 ancestrally diverse participants of large-scale studies. Individual-level data from the UK Biobank and the Million Veteran Program and summary statistics of publicly available databases were analyzed. RESULTS A set of associations between SCAD polygenic score and related vascular diseases were replicated in non-European samples. Notable associations with the SCAD polygenic score included (1) coronary artery disease, myocardial infarction, and migraine headache in a Hispanic group (coronary artery disease: odds ratio [OR], 0.93 [95% CI, 0.90-0.95]; P=2.35×10-7; myocardial infarction: OR, 0.88 [95% CI, 0.80-0.96]; P=5.73×10-3; migraine headache: OR, 1.03 [95% CI, 1.01-1.06]; P=1.86×10-2) of the Million Veteran Program; (2) headache in an African-ancestry group (OR, 1.22 [95% CI, 1.06-1.41]; P=6.94×10-3) and a South Asian-ancestry group (OR, 1.18 [95% CI, 1.02-1.37]; P=2.43×10-2) of the UK Biobank; and (3) coronary artery disease, myocardial infarction, and migraine headache in East Asian-ancestry cohorts (coronary artery disease: OR, 0.95 [95% CI, 0.93-0.98]; P=2.66×10-3; myocardial infarction: OR, 0.86 [95% CI, 0.83-0.89]; P=9.51×10-16; migraine headache: OR, 1.27 [95% CI, 1.10-1.47]; P=1.03×10-3). CONCLUSIONS Pleiotropic associations of SCAD polygenic risk with related vascular diseases previously identified in European-ancestry groups showed notable, largely consistent patterns in non-European-ancestry groups.
Collapse
Affiliation(s)
- Chang Xu
- Department of Biostatistics and Center for Statistical Genetics University of Michigan School of Public Health Ann Arbor MI USA
| | - Min-Lee Yang
- Division of Cardiovascular Medicine, Department of Internal Medicine University of Michigan Medical School Ann Arbor MI USA
- Department of Computational Medicine and Bioinformatics University of Michigan Ann Arbor MI USA
- Department of Human Genetics University of Michigan Medical School Ann Arbor MI USA
| | - Pik Fang Kho
- VA Palo Alto Health Care System Palo Alto CA USA
- Department of Medicine (Division of Cardiovascular Medicine) Stanford University School of Medicine Stanford CA USA
| | - Shoa L Clarke
- VA Palo Alto Health Care System Palo Alto CA USA
- Department of Medicine (Division of Cardiovascular Medicine) Stanford University School of Medicine Stanford CA USA
| | - Catherine Tcheandjieu
- VA Palo Alto Health Care System Palo Alto CA USA
- Gladstone Institute of Data Science and Biotechnology Gladstone Institutes San Francisco CA USA
- Department of Epidemiology and Biostatistics University of California San Francisco San Francisco CA USA
| | - Patricia A Peyser
- Department of Epidemiology University of Michigan School of Public Health Ann Arbor MI USA
| | | | - Shih-Pin Chen
- Department of Medical Research Taipei Veterans General Hospital Taipei Taiwan
- Institute of Clinical Medicine National Yang Ming Chiao Tung University Taipei Taiwan
| | | | - Jacqueline Saw
- Vancouver General Hospital, Division of Cardiology University of British Columbia Vancouver Canada
| | - Xiang Zhou
- Department of Biostatistics and Center for Statistical Genetics University of Michigan School of Public Health Ann Arbor MI USA
| | - Themistocles L Assimes
- VA Palo Alto Health Care System Palo Alto CA USA
- Department of Medicine (Division of Cardiovascular Medicine) Stanford University School of Medicine Stanford CA USA
| | - Santhi K Ganesh
- Division of Cardiovascular Medicine, Department of Internal Medicine University of Michigan Medical School Ann Arbor MI USA
- Department of Human Genetics University of Michigan Medical School Ann Arbor MI USA
| |
Collapse
|
4
|
Li XC, Wang B, Tang Y. Causal relationships between four types of lipids and breast cancer risk with potential mediators: evidence from Mendelian randomization study and bioinformatics analysis. Discov Oncol 2025; 16:791. [PMID: 40379931 PMCID: PMC12084463 DOI: 10.1007/s12672-025-02597-x] [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: 02/04/2025] [Accepted: 05/06/2025] [Indexed: 05/19/2025] Open
Abstract
BACKGROUND AND PURPOSE Breast cancer (BC) is the primary cause of cancer-related deaths among women worldwide, with increasing evidence pointing to the effect of metabolic factors, particularly lipid levels, in its pathogenesis. In this research, Mendelian randomization (MR) was employed to explore the causality between four plasma lipid traits-total cholesterol (TC), triglycerides (TG), high-density lipoprotein cholesterol (HDL-C) and low-density lipoprotein cholesterol (LDL-C)-and the risk of BC. Additionally, we explored the potential mediating effects of coronary artery disease (CAD), total testosterone (TT) on these associations and possible mechanisms through bioinformatics analyses. METHODS Data of genome-wide association study (GWAS) on lipids, CAD, TT and BC were obtained from public sources and websites as part of a genome-wide association research. The inference of causality was primarily assessed through the inverse variance weighting (IVW) approach, with supplementary tests for horizontal pleiotropy and heterogeneity. To verify the directionality of causal relationships, the MR Steiger test was applied. Additionally, reverse causality was evaluated by regarding BC as the exposure. To adjust for confounders, multivariate MR (MVMR) was performed, followed by a two-step mediation analysis to investigate the mediating roles of CAD in the lipid-BC association, and of TT in the CAD-BC relationship. The intersecting SNP (rs11556924) between causal pathways was established through a Venn diagram and its associated gene (Zinc Finger C3HC-Type Containing 1, ZC3HC1) was identified through the g:Profiler database. The expression of ZC3HC1 was further explored using the TIMER, GEPIA2 and HPA database. Finally, enrichment analyses of Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) and protein-protein interactions (PPI) network analysis were conducted on ZC3HC1 and its related genes. RESULTS The random-effects IVW analysis yielded the following results: HDL-C on CAD (OR = 0.843, 95% CI 0.771-0.921, P < 0.001), CAD on BC (OR = 0.935, 95% CI 0.892-0.980, P = 0.005), HDL-C on BC (OR = 1.127, 95% CI 1.059-1.199, P < 0.001), CAD on TT (OR = 0.987, 95% CI 0.975-0.998, P = 0.020) and TT on BC (OR = 1.354, 95% CI 1.148-1.598, P < 0.001). The MR Steiger test results support the validity of the inferred causal direction (P < 0.001). There were no discernible causal relationships between BC and HDL-C/CAD according to reverse MR analysis (P > 0.05). Following MVMR adjustment, the causal effects of HDL-C, CAD, and TT on BC were still statistically significant (P < 0.05). Besides, the two-step mediation analysis indicated that CAD mediated 7.8% of the causal effect of HDL-C on BC, whereas TT mediated 6.1% of the causal effect between CAD and BC. The expression of ZC3HC1 showed no significant expression difference between normal and BC tissues (P > 0.05), which might indicate a carcinogenic effect independent of expression levels but driven by functional alterations induced by variants (C > T). Functional network analysis suggested that ZC3HC1 was associated with multiple signal pathways in cancers, such as PI3K-Akt and MAPK signal pathways. CONCLUSIONS From a genetic perspective, our study reveals that there is causality between HDL-C levels and BC risk, with CAD and TT acting as partial mediators in this relationship. Moreover, our study firstly establishes a potential link between CAD-associated SNP (rs11556924), the corresponding gene (ZC3HC1) functional dysregulation, and the initiation of BC. These findings shed light on the biological links between lipids and BC, potentially contributing to future prevention and treatment strategies.
Collapse
Affiliation(s)
- Xu-Chu Li
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 South Panjiayuan Lane, Chaoyang District, Beijing, 100021, China
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Bangqi Wang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yu Tang
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 South Panjiayuan Lane, Chaoyang District, Beijing, 100021, China.
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
5
|
Leyden GM, Sobczyk MK, Richardson TG, Gaunt TR. Distinct pathway-based effects of blood pressure and body mass index on cardiovascular traits: comparison of novel Mendelian randomization approaches. Genome Med 2025; 17:54. [PMID: 40375348 DOI: 10.1186/s13073-025-01472-2] [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: 12/21/2023] [Accepted: 04/11/2025] [Indexed: 05/18/2025] Open
Abstract
BACKGROUND Mendelian randomization (MR) leverages trait associated genetic variants as instrumental variables (IVs) to determine causal relationships in epidemiology. However, genetic IVs for complex traits are typically highly heterogeneous and, at a molecular level, exert effects on different biological processes. Exploration of the biological underpinnings of such heterogeneity can enhance our understanding of disease mechanisms and inform therapeutic strategies. Here, we introduce a new approach to instrument partitioning based on enrichment of Mendelian disease categories (pathway-partitioned) and compare it to an existing method based on genetic colocalization in contrasting tissues (tissue-partitioned). METHODS We employed individual- and summary-level MR methodologies using SNPs grouped by pathway informed by proximity to Mendelian disease genes affecting the renal system or vasculature (for blood pressure (BP)), or mental health and metabolic disorders (for body mass index (BMI)). We compared the causal effects of pathway-partitioned SNPs on cardiometabolic outcomes with those derived using tissue-partitioned SNPs informed by colocalization with gene expression in kidney, artery (BP), or adipose and brain tissues (BMI). Additionally, we assessed the likelihood that estimates observed for partitioned exposures could emerge by chance using random SNP sampling. RESULTS Our pathway-partitioned findings suggest the causal relationship between systolic BP and heart disease is predominantly driven by vessel over renal pathways. The stronger effect attributed to kidney over artery tissue in our tissue-partitioned MR hints at a multifaceted interplay between pathways in the disease aetiology. We consistently identified a dominant role for vessel (pathway) and artery (tissue) driving the negative directional effect of diastolic BP on left ventricular stroke volume and positive directional effect of systolic BP on type 2 diabetes. We also found when dissecting the BMI pathway contribution to atrial fibrillation that metabolic-pathway and brain-tissue IVs predominantly drove the causal effects relative to mental health and adipose in pathway- and tissue-partitioned MR analyses, respectively. CONCLUSIONS This study presents a novel approach to dissecting heterogeneity in MR by integrating clinical phenotypes associated with Mendelian disease. Our findings emphasize the importance of understanding pathway-/tissue-specific contributions to complex exposures when interpreting causal relationships in MR. Importantly, we advocate caution and robust validation when interpreting pathway-partitioned effect size differences.
Collapse
Affiliation(s)
- Genevieve M Leyden
- MRC Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, Oakfield House, Bristol, BS8 2BN, UK.
| | - Maria K Sobczyk
- MRC Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, Oakfield House, Bristol, BS8 2BN, UK
| | - Tom G Richardson
- MRC Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, Oakfield House, Bristol, BS8 2BN, UK
| | - Tom R Gaunt
- MRC Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, Oakfield House, Bristol, BS8 2BN, UK.
| |
Collapse
|
6
|
Legault MA, Hartford J, Arsenault BJ, Yang AY, Pineau J. A flexible machine learning Mendelian randomization estimator applied to predict the safety and efficacy of sclerostin inhibition. Am J Hum Genet 2025:S0002-9297(25)00171-5. [PMID: 40378846 DOI: 10.1016/j.ajhg.2025.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 04/22/2025] [Accepted: 04/22/2025] [Indexed: 05/19/2025] Open
Abstract
Mendelian randomization (MR) enables the estimation of causal effects while controlling for unmeasured confounding factors. However, traditional MR's reliance on strong parametric assumptions can introduce bias if these are violated. We describe a machine learning MR estimator named quantile instrumental variable (Quantile IV) that achieves a low estimation error in a wide range of plausible MR scenarios. Quantile IV is distinctive in its ability to estimate nonlinear and heterogeneous causal effects and offers a flexible approach for subgroup analysis. Applying quantile IV, we investigate the impact of circulating sclerostin levels on heel bone mineral density, osteoporosis, and cardiovascular outcomes. Employing various MR estimators and colocalization techniques, our analysis reveals that a genetically predicted reduction in sclerostin levels significantly increases heel bone mineral density and reduces the risk of osteoporosis while showing no discernible effect on ischemic cardiovascular diseases. As a second application, we estimated the effect of increases in low-density lipoprotein cholesterol and waist-to-hip ratio on ischemic cardiovascular diseases using this well-known association as a positive control analysis. Quantile IV contributes to the advancement of MR methodology, and the selected applications demonstrate the applicability of our estimator in various MR contexts.
Collapse
Affiliation(s)
- Marc-André Legault
- Department of Computer Science, McGill University, Montreal, QC, Canada; Mila, Montreal, QC, Canada; Faculté de pharmacie, Université de Montréal, Montreal, QC, Canada; Centre de recherche Azrieli du CHU Sainte-Justine, Montreal, QC, Canada.
| | | | - Benoît J Arsenault
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Québec, QC, Canada; Department of Medicine, Faculty of Medicine, Université Laval, Quebec, QC, Canada
| | - Archer Y Yang
- Mila, Montreal, QC, Canada; Department of Mathematics and Statistics, McGill University, Montreal, QC, Canada
| | - Joelle Pineau
- Department of Computer Science, McGill University, Montreal, QC, Canada; Mila, Montreal, QC, Canada
| |
Collapse
|
7
|
Ahmadi MN, Mundell HD, Sutherland GT, Hamer M, Sillanpää E, Blodgett JM, Cruz BDP, Stamatakis E. Physical activity, genetic predisposition, and incident cardiovascular disease: Prospective analyses of the UK Biobank. JOURNAL OF SPORT AND HEALTH SCIENCE 2025:101055. [PMID: 40355085 DOI: 10.1016/j.jshs.2025.101055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 09/20/2024] [Accepted: 10/28/2024] [Indexed: 05/14/2025]
Abstract
BACKGROUND It is unclear whether physical activity can benefit participants with high genetic predisposition to cardiovascular disease. We examined the joint associations of intensity-specific physical activity and genetic predisposition (based on polygenetic risk score) with incident coronary heart disease (CHD), stroke, and atrial fibrillation (AF). METHODS This prospective cohort study included 303,950 adults (age = 56.4 ± 8.0 years, mean ± SD; 52.5% females) from the UK Biobank with physical activity and disease-related genotypes. Moderate-to-vigorous physical activity (MVPA) and intensity-specific activity was classified according to volume (e.g., MVPA was classified as none, low, medium, and high). Genetic predisposition for CHD, stroke, and AF were classified as low (Quintile 1), intermediate (Quintiles 2-4), and high (Quintile 5). RESULTS During 11.6 ± 2.1 years of follow-up: 19,865 CHD; 7907 stroke; and 16,688 AF events occurred. Compared to the no MVPA and high genetic risk group, we observed lower CHD risk for increasing levels of MVPA over and above genetic risk groupings. These associations were primarily driven by vigorous-intensity activity. For example, in the high genetic risk group, those with low vigorous-intensity activity levels (compared to none) had a hazard ratio (HR) of 0.78 (95% confidence interval (95% CI):0.72-0.86) compared to an HR of 0.90 (95% CI: 0.83-0.98) for low moderate-intensity activity levels. For stroke incidence, we observed a protective association for MVPA across genetic risk groups that was mostly driven by moderate-intensity activity volume. Among the high genetic risk group, low moderate-intensity had an HR of 0.77 (95% CI:0.66-0.90), whereas low vigorous-intensity had no association (HR = 0.95, 95% CI:0.82-1.09). We did not observe a consistent joint association of MVPA and AF genetic predisposition. CONCLUSIONS We observed lower CHD and stroke risk for low to high MVPA among participants with high genetic predisposition. The associations of moderate- and vigorous-intensity activity volume differed considerably across cardiovascular disease sub-types. Overall, our findings suggest vigorous intensity activity may mitigate genetic predisposition for CHD while moderate intensity activity may be associated with similar effects for stroke. Joint associations were less consistent across AF genetic predisposition groups. Our results inform precision medicine approaches and future lifestyle modification interventions by quantifying the potential benefits of physical activity among at-risk individuals.
Collapse
Affiliation(s)
- Matthew N Ahmadi
- Mackenzie Wearables Research Hub, Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, 2006, Australia; Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, 2006, Australia.
| | - Hamish D Mundell
- Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, 2006, Australia
| | - Greg T Sutherland
- Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, 2006, Australia
| | - Mark Hamer
- Institute Sport Exercise Health, Division of Surgery and Interventional Science, University College London, London, WC1E 6BT, United Kingdom
| | - Elina Sillanpää
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, 40014, Finland; The Wellbeing Services County of Central Finland, Jyväskylä, 40014, Finland
| | - Joanna M Blodgett
- Institute Sport Exercise Health, Division of Surgery and Interventional Science, University College London, London, WC1E 6BT, United Kingdom
| | - Borja Del Pozo Cruz
- Faculty of Sports Sciences, Universidad Europea de Madrid, Madrid, 28670, Spain; Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, Madrid, 28670, Spain; Department of Sports Science and Clinical Biomechanics, University of Southern Denmark, Odense, 5230, Denmark
| | - Emmanuel Stamatakis
- Mackenzie Wearables Research Hub, Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, 2006, Australia; Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, 2006, Australia
| |
Collapse
|
8
|
Le NN, Tran TQB, McClure J, Gill D, Padmanabhan S. Emerging antihypertensive therapies and cardiovascular, kidney, and metabolic outcomes: a Mendelian randomization study. EUROPEAN HEART JOURNAL. CARDIOVASCULAR PHARMACOTHERAPY 2025; 11:264-274. [PMID: 39963705 PMCID: PMC12046581 DOI: 10.1093/ehjcvp/pvaf015] [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: 11/30/2024] [Revised: 12/18/2024] [Accepted: 02/14/2025] [Indexed: 05/03/2025]
Abstract
AIMS Emerging antihypertensive drug classes offer new opportunities to manage hypertension; however, their long-term effects on cardiovascular, kidney, and metabolic (CKM) outcomes remain to be elucidated. This study aims to explore the effects of phosphodiesterase type 5 inhibitors (PDE5i), soluble guanylate cyclase stimulators (sGCs), endothelin receptor antagonists (ERAs), and angiotensinogen inhibitors (AGTis) on a range of CKM outcomes. METHODS AND RESULTS Mendelian randomization (MR), summary-based MR (SMR), and colocalization analyses were applied to assess the drug effect on coronary artery disease (CAD), myocardial infarction (MI), ischaemic stroke, atrial fibrillation (AF), heart failure (HF), type 2 diabetes (T2D), and chronic kidney disease (CKD). Genetic association and gene expression summary data were obtained from the largest European-ancestry genome-wide association studies (GWAS) and the genotype-tissue expression version 8 for 29 tissues relevant to the outcomes' pathophysiology.Genetically predicted systolic blood pressure (SBP) reduction was associated with reduced risks of all outcomes. PDE5i was associated with reduced risks of CAD (OR per 10-mmHg decrease in SBP: 0.348[95% confidence interval (CI): 0.199-0.607]) and ischaemic stroke (0.588[0.453-0.763]). sGCs showed protective effects against CAD (0.332[0.236-0.469]), MI (0.238[0.168-0.337]), and CKD (0.55[0.398-0.761]). ERA and AGTi showed protective effects against CAD and ischaemic stroke. SMR and colocalization supported the association of gene expression levels of GUCY1A3 and PDE5A with CAD and MI risk. CONCLUSION Our study highlights the potential of PDE5i, sGCs, ERA, and AGTi in reducing cardiovascular and renal risks. These findings underscore the necessity for targeted clinical trials to validate the efficacy and safety of these therapies.
Collapse
Affiliation(s)
- Nhu Ngoc Le
- BHF Cardiovascular Research Centre, School of Cardiovascular and Metabolic Health, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
| | - Tran Quoc Bao Tran
- BHF Cardiovascular Research Centre, School of Cardiovascular and Metabolic Health, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
| | - John McClure
- BHF Cardiovascular Research Centre, School of Cardiovascular and Metabolic Health, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
| | - Dipender Gill
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London W2 1PG, UK
| | - Sandosh Padmanabhan
- BHF Cardiovascular Research Centre, School of Cardiovascular and Metabolic Health, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
| |
Collapse
|
9
|
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
|
10
|
Sadhu N, Dalan R, Jain PR, Lee CJM, Pakkiri LS, Tay KY, Mina TH, Low D, Min Y, Ackers-Johnson M, Thi TT, Kota VG, Shi Y, Liu Y, Yu H, Lai V, Yang Y, Tay D, Ng HK, Wang X, Wong KE, Lam M, Guan XL, Bertin N, Wong E, Best J, Sarangarajan R, Elliott P, Riboli E, Lee J, Lee ES, Ngeow J, Tan P, Cheung C, Drum CL, Foo RS, Michelotti GA, Yu H, Sheridan PA, Loh M, Chambers JC. Metabolome-wide association identifies ferredoxin-1 (FDX1) as a determinant of cholesterol metabolism and cardiovascular risk in Asian populations. NATURE CARDIOVASCULAR RESEARCH 2025; 4:567-583. [PMID: 40360795 DOI: 10.1038/s44161-025-00638-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 03/19/2025] [Indexed: 05/15/2025]
Abstract
The burden of cardiovascular disease is rising in the Asia-Pacific region, in contrast to falling cardiovascular disease mortality rates in Europe and North America. Here we perform quantification of 883 metabolites by untargeted mass spectroscopy in 8,124 Asian adults and investigate their relationships with carotid intima media thickness, a marker of atherosclerosis. Plasma concentrations of 3beta-hydroxy-5-cholestenoate (3BH5C), a cholesterol metabolite, were inversely associated with carotid intima media thickness, and Mendelian randomization studies supported a causal relationship between 3BH5C and coronary artery disease. The observed effect size was 5- to 6-fold higher in Asians than Europeans. Colocalization analyses indicated the presence of a shared causal variant between 3BH5C plasma levels and messenger RNA and protein expression of ferredoxin-1 (FDX1), a protein that is essential for sterol and bile acid synthesis. We validated FDX1 as a regulator of 3BH5C synthesis in hepatocytes and macrophages and demonstrated its role in cholesterol efflux in macrophages and aortic smooth muscle cells, using knockout and overexpression models.
Collapse
Affiliation(s)
- Nilanjana Sadhu
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.
| | - Rinkoo Dalan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Department of Endocrinology, Tan Tock Seng Hospital, Singapore, Singapore
| | - Pritesh R Jain
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Chang Jie Mick Lee
- Cardiovascular Research Institute, National University Health System, Singapore, Singapore
- Cardiovascular Metabolic Disease Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | | | - Kai Yi Tay
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Theresia H Mina
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Dorrain Low
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Yilin Min
- Precision Medicine Translational Research Programme, and Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Matthew Ackers-Johnson
- Cardiovascular Research Institute, National University Health System, Singapore, Singapore
- Cardiovascular Metabolic Disease Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Thi Tun Thi
- Precision Medicine Translational Research Programme, and Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Vishnu Goutham Kota
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yu Shi
- Precision Medicine Translational Research Programme, and Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yan Liu
- Precision Medicine Translational Research Programme, and Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Hanry Yu
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Vicky Lai
- Cardiovascular Research Institute, National University Health System, Singapore, Singapore
- Cardiovascular Metabolic Disease Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yang Yang
- Precision Medicine Translational Research Programme, and Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Darwin Tay
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Hong Kiat Ng
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Xiaoyan Wang
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | | | - Max Lam
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- North Region, Institute of Mental Health, Singapore, Singapore
| | - Xue Li Guan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Nicolas Bertin
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
| | - Eleanor Wong
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
| | - James Best
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | | | - Paul Elliott
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
| | - Elio Riboli
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
| | - Jimmy Lee
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- North Region, Institute of Mental Health, Singapore, Singapore
| | - Eng Sing Lee
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Clinical Research Unit, National Healthcare Group Polyclinic, Singapore, Singapore
| | - Joanne Ngeow
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Cancer Genetics Service, National Cancer Centre, Singapore, Singapore
| | - Patrick Tan
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
- Precision Health Research, Singapore, Singapore
| | - Christine Cheung
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
| | - Chester Lee Drum
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Roger Sy Foo
- Cardiovascular Research Institute, National University Health System, Singapore, Singapore
- Cardiovascular Metabolic Disease Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | | | - Haojie Yu
- Cardiovascular Metabolic Disease Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Precision Medicine Translational Research Programme, and Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | | | - Marie Loh
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- National Skin Centre, Singapore, Singapore
| | - John C Chambers
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK.
- Precision Health Research, Singapore, Singapore.
| |
Collapse
|
11
|
Amente LD, Mills NT, Le TD, Hyppönen E, Lee SH. A latent outcome variable approach for Mendelian randomization using the stochastic expectation maximization algorithm. Hum Genet 2025; 144:559-574. [PMID: 40214754 PMCID: PMC12033120 DOI: 10.1007/s00439-025-02739-9] [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: 10/28/2024] [Accepted: 03/18/2025] [Indexed: 04/27/2025]
Abstract
Mendelian randomization (MR) is a widely used tool to uncover causal relationships between exposures and outcomes. However, existing MR methods can suffer from inflated type I error rates and biased causal effects in the presence of invalid instruments. Our proposed method enhances MR analysis by augmenting latent phenotypes of the outcome, explicitly disentangling horizontal and vertical pleiotropy effects. This allows for explicit assessment of the exclusion restriction assumption and iteratively refines causal estimates through the expectation-maximization algorithm. This approach offers a unique and potentially more precise framework compared to existing MR methods. We rigorously evaluate our method against established MR approaches across diverse simulation scenarios, including balanced and directional pleiotropy, as well as violations of the Instrument Strength Independent of Direct Effect (InSIDE) assumption. Our findings consistently demonstrate superior performance of our method in terms of controlling type I error rates, bias, and robustness to genetic confounding, regardless of whether individual-level or summary data is used. Additionally, our method facilitates testing for directional horizontal pleiotropy and outperforms MR-Egger in this regard, while also effectively testing for violations of the InSIDE assumption. We apply our method to real data, demonstrating its effectiveness compared to traditional MR methods. This analysis reveals the causal effects of body mass index (BMI) on metabolic syndrome (MetS) and a composite MetS score calculated by the weighted sum of its component factors. While the causal relationship is consistent across most methods, our proposed method shows fewer violations of the exclusion restriction assumption, especially for MetS scores where horizontal pleiotropy persists and other methods suffer from inflation.
Collapse
Affiliation(s)
- Lamessa Dube Amente
- Australian Centre for Precision Health, University of South Australia, Adelaide, SA, 5000, Australia.
- UniSA Allied Health and Human Performance, University of South Australia, Adelaide, SA, 5000, Australia.
- South Australian Health and Medical Research Institute, Adelaide, SA, 5000, Australia.
- Epidemiology Department, Jimma University, 378, Jimma, Ethiopia.
| | - Natalie T Mills
- Discipline of Psychiatry, University of Adelaide, Adelaide, South Australia, 5000, Australia
| | - Thuc Duy Le
- UniSA STEM, University of South Australia, Mawson Lakes, SA, 5095, Australia
| | - Elina Hyppönen
- Australian Centre for Precision Health, University of South Australia, Adelaide, SA, 5000, Australia
- South Australian Health and Medical Research Institute, Adelaide, SA, 5000, Australia
- UniSA Clinical and Health Sciences, University of South Australia, Adelaide, SA, 5000, Australia
| | - S Hong Lee
- Australian Centre for Precision Health, University of South Australia, Adelaide, SA, 5000, Australia.
- UniSA Allied Health and Human Performance, University of South Australia, Adelaide, SA, 5000, Australia.
- South Australian Health and Medical Research Institute, Adelaide, SA, 5000, Australia.
| |
Collapse
|
12
|
Wang L, Cui F, Chen X, Zhang Q, Yang J, Li S. Examining the causal relationship between hypertension and benign paroxysmal vertigo: a univariate and multivariate Mendelian randomization study. Acta Otolaryngol 2025; 145:400-408. [PMID: 40122118 DOI: 10.1080/00016489.2025.2479015] [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: 01/15/2025] [Revised: 03/05/2025] [Accepted: 03/07/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Patients with benign paroxysmal positional vertigo (BPPV) may be at increased risk for hypertension. OBJECTIVES To examine the causal relationship between hypertensive disorders and benign paroxysmal vertigo (BPV). MATERIALS AND METHODS A bidirectional two-sample Mendelian randomization (MR) study was conducted to assess the causal relationship between hypertension and BPV. Summary statistics for BPV and hypertension were obtained from the FinnGen biobank to support the primary MR analysis. Five complementary methods - inverse variance weighted, weighted median, MR-Egger, simple mode, and weighted mode - were applied to obtain MR estimates. Furthermore, a multivariable MR (MVMR) analysis was conducted to examine whether hypertension independently influences BPV. RESULTS The MR analysis indicated a significant causal effect of hypertension on BPV, with an odds ratio (OR) of 1.11 (95% CI: 1.02-1.22, p = .019), indicating an increased risk of BPV associated with hypertension. MVMR analysis further indicated that the causal effect of hypertension on BPV remained significant even after adjusting for menopausal disorders and ischemic stroke (OR: 1.108, 95% CI: 1.003-1.225, p = .043). CONCLUSIONS AND SIGNIFICANCE This MR study provided evidence supporting a causal effect of hypertension on BPV.
Collapse
Affiliation(s)
- Luyao Wang
- Department of Otorhinolaryngology-Head & Neck Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Feilun Cui
- Urology Department, Taizhou Second People's Hospital Affiliated to Yangzhou University, Taizhou, China
| | - Xiangping Chen
- Department of Otorhinolaryngology-Head & Neck Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Qing Zhang
- Department of Otorhinolaryngology-Head & Neck Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jun Yang
- Department of Otorhinolaryngology-Head & Neck Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Shuna Li
- Department of Otorhinolaryngology-Head & Neck Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
13
|
Wu GJ, Zhang Y, Liang R, Peng L, Zhang SH. Comparison of Survival Outcomes of Early-Stage Non-Small-Cell Lung Cancer in Elderly Patients (≥ 70 years) Treated With Stereotactic Body Radiotherapy Versus Surgical Resection. World J Surg 2025; 49:1160-1171. [PMID: 40214658 DOI: 10.1002/wjs.12584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/21/2025] [Accepted: 03/22/2025] [Indexed: 05/09/2025]
Abstract
OBJECTIVE To evaluate the survival outcomes of stereotactic body radiotherapy (SBRT) versus surgical resection in elderly patients (≥ 70 years old) with early-stage non-small-cell lung cancer (NSCLC). METHODS A systematic search was conducted across databases such as PubMed, Web of Science, EMBASE, Cochrane Library, CNKI, VIP, and Wanfang to collect comparative studies on the survival outcomes of SBRT versus surgery for early-stage NSCLC in elderly patients, with searches up to December 2023 and statistical analysis performed using Stata software. This meta-analysis included 10 comparative studies, encompassing 9410 elderly patients with early-stage NSCLC. RESULTS The results indicated that the SBRT group had worse outcomes compared to the surgery group in terms of 3-year overall survival (OS) [HR = 1.31, 95% CI (1.02-1.70), p < 0.05], 5-year OS [HR = 1.37, 95% CI (1.06-1.77), p < 0.05], 1-year cancer-specific survival (CSS) [HR = 1.32, 95% CI (1.03-1.68), p < 0.05], 3-year CSS [HR = 1.89, 95% CI (1.70-2.11), p < 0.01], and 5-year CSS [HR = 1.92, 95% CI (1.75-2.10), p < 0.01]. There were no statistically significant differences between the two groups in terms of 1-year OS, 1-year, 3-year, and 5-year progression-free survival (PFS), and locoregional control (LC) rates (p > 0.05). CONCLUSION For elderly early-stage NSCLC patients with acceptable risk, surgery remains the more appropriate treatment method. For patients unable to undergo surgery or who refuse it, SBRT serves as a viable alternative treatment option.
Collapse
Affiliation(s)
- Guo-Jun Wu
- Department of Thoracic Surgery, Beijing Anzhen Nanchong Hospital, Capital Medical University & Nanchong Central Hospital, Nanchong, China
- Motor Robotics Institute (MRI), South China Hospital, Shenzhen University, Shenzhen, China
| | - Yi Zhang
- Motor Robotics Institute (MRI), South China Hospital, Shenzhen University, Shenzhen, China
- Department of Urology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou Medical University, Jinzhou, China
| | - Rui Liang
- Motor Robotics Institute (MRI), South China Hospital, Shenzhen University, Shenzhen, China
| | - Lei Peng
- Motor Robotics Institute (MRI), South China Hospital, Shenzhen University, Shenzhen, China
| | - Shao-Hua Zhang
- Motor Robotics Institute (MRI), South China Hospital, Shenzhen University, Shenzhen, China
| |
Collapse
|
14
|
Salido E, de Medeiros Vieira C, Mosquera JV, Zade R, Parikh P, Suryavanshi S, Miller CL, Lo Sardo V. The 9p21.3 Coronary Artery Disease Risk Locus Drives Vascular Smooth Muscle Cells to an Osteochondrogenic State. Arterioscler Thromb Vasc Biol 2025; 45:702-721. [PMID: 40143812 PMCID: PMC12017600 DOI: 10.1161/atvbaha.124.322045] [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: 10/22/2024] [Accepted: 03/12/2025] [Indexed: 03/28/2025]
Abstract
BACKGROUND Genome-wide association studies have identified common genetic variants at ≈300 human genomic loci linked to coronary artery disease susceptibility. Among these genomic regions, the most impactful is the 9p21.3 coronary artery disease risk locus, which spans a 60-kb gene desert and encompasses ≈80 SNPs (single nucleotide polymorphism) in high linkage disequilibrium. Despite ≈2 decades since its discovery, the role of the 9p21.3 locus in cells of the vasculature remains incompletely resolved. METHODS We differentiated induced pluripotent stem cells (iPSCs) from risk, nonrisk donors at 9p21.3, and isogenic knockouts into vascular smooth muscle cells (VSMCs). We performed single-cell transcriptomic profiling, including coembedding and comparison with publicly available human arterial data sets. We conducted functional characterization using migration and calcification assays and confirmed our findings on iPSC-VSMCs derived from additional donors. Finally, we used overexpression of ANRIL followed by gene expression analysis. RESULTS We demonstrated that iPSC-VSMCs harboring the 9p21.3 risk haplotype preferentially adopt an osteochondrogenic state and show remarkable similarity to fibrochondrocytes from human artery tissue. The transcriptional profile and functional assessment of migration and calcification capacity across iPSC-VSMC lines from multiple donors concordantly resemble an osteochondrogenic state. Importantly, we identified numerous transcription factors driving different VSMC state trajectories. Additionally, we prioritized LIMCH1 and CRABP1 as signature genes critical for defining the risk transcriptional program. Finally, overexpression of a short isoform of ANRIL in 9p21.3 knockout cells was sufficient to induce the osteochondrogenic transcriptional signature. CONCLUSIONS Our study provides new insights into the mechanism of the 9p21.3 risk locus and defines its previously undescribed role in driving a disease-prone transcriptional and functional state in VSMCs concordant with an osteochondrogenic-like state. Our data suggest that the 9p21.3 risk haplotype likely promotes arterial calcification, through altered expression of ANRIL, in a cell type-specific and cell-autonomous manner, providing insight into potential risk assessment and treatment for carriers.
Collapse
MESH Headings
- Humans
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Induced Pluripotent Stem Cells/metabolism
- Induced Pluripotent Stem Cells/pathology
- Chromosomes, Human, Pair 9/genetics
- Genetic Predisposition to Disease
- Coronary Artery Disease/genetics
- Coronary Artery Disease/pathology
- Coronary Artery Disease/metabolism
- Cell Movement
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- Polymorphism, Single Nucleotide
- Cells, Cultured
- Phenotype
- Vascular Calcification/genetics
- Vascular Calcification/pathology
- Vascular Calcification/metabolism
- Risk Factors
- Cell Differentiation
- Haplotypes
- Gene Expression Profiling
- Transcriptome
- Genome-Wide Association Study
Collapse
Affiliation(s)
- Elsa Salido
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison (E.S., C.d.M.V., R.Z., P.P., S.S., V.L.S.)
| | - Carolina de Medeiros Vieira
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison (E.S., C.d.M.V., R.Z., P.P., S.S., V.L.S.)
| | - Jose Verdezoto Mosquera
- Department of Genome Sciences, and Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville (J.V.M., C.L.M.)
| | - Rohan Zade
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison (E.S., C.d.M.V., R.Z., P.P., S.S., V.L.S.)
| | - Parth Parikh
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison (E.S., C.d.M.V., R.Z., P.P., S.S., V.L.S.)
| | - Shraddha Suryavanshi
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison (E.S., C.d.M.V., R.Z., P.P., S.S., V.L.S.)
| | - Clint L. Miller
- Department of Genome Sciences, and Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville (J.V.M., C.L.M.)
| | - Valentina Lo Sardo
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison (E.S., C.d.M.V., R.Z., P.P., S.S., V.L.S.)
| |
Collapse
|
15
|
Liu W, Chen S, Yang C, Lei F, Huang X, Zhang X, Sun T, Lin L, Wang C, Cao Y, She ZG, Xiao X, Li H. Elevated high-density lipoprotein triglycerides increase atherosclerotic risk. J Lipid Res 2025; 66:100791. [PMID: 40164335 PMCID: PMC12088758 DOI: 10.1016/j.jlr.2025.100791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 03/25/2025] [Accepted: 03/27/2025] [Indexed: 04/02/2025] Open
Abstract
The relationship between high-density lipoprotein (HDL) and atherosclerotic risk remains incompletely elucidated, potentially due to the inherent heterogeneity of HDL particles. Hypertriglyceridemia is associated with alterations in HDL composition. This study investigated the impact of elevated triglycerides (TG) on HDL and its association with coronary artery disease (CAD) risk using a large prospective cohort study and Mendelian randomization (MR). We found that elevated TG was associated with reduced HDL particle size, decreased concentrations of HDL components, and increased triglycerides in HDL (HDL-TG) (all P for trend < 0.001). The protective effects of HDL particle concentration and HDL cholesterol on CAD are attenuated with increasing serum TG levels. An independent and positive association between HDL-TG levels and incident CAD events (hazard ratio [HR] per 1 standard deviation increase: 1.066, 95% CI: 1.052-1.080, P < 0.001) was confirmed even after adjustment for established cardiovascular disease risk factors. MR analyses supported a causal role for HDL-TG in CAD development (inverse-variance weighted [IVW] method: odds ratios [ORs] of 1.120 (95% CI: 1.053-1.192, P < 0.001) and 1.141 (95% CI: 1.032-1.263, P = 0.010) for dataset groups 1 and 2, respectively). Drug-target MR analyses suggested a potential association between omega-3 fatty acids (OM3-FA) and lower HDL-TG levels, with LPL and DGAT2 as key pharmacological targets. Our findings suggest that elevated TG contributes to adverse alterations in HDL, elevating CAD risk. HDL-TG is an independent positive risk factor for CAD and a potential causal contributor to CAD development. OM3-FA supplementation may offer a therapeutic strategy for mitigating the CAD risk associated with elevated HDL-TG.
Collapse
Affiliation(s)
- Weifang Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China; State Key Laboratory of New Drug Discovery and Development for Major Diseases, Gannan Medical University, Ganzhou, China; Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China
| | - Shaoze Chen
- Department of Cardiology, Huanggang Central Hospital of Yangtze University, Huanggang, China
| | - Chengzhang Yang
- Department of Cardiology, Huanggang Central Hospital of Yangtze University, Huanggang, China
| | - Fang Lei
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xuewei Huang
- Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Xingyuan Zhang
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Tao Sun
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Lijin Lin
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Chuansen Wang
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yuanyuan Cao
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zhi-Gang She
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China; State Key Laboratory of New Drug Discovery and Development for Major Diseases, Gannan Medical University, Ganzhou, China; Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China.
| | - Xuan Xiao
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.
| | - Hongliang Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China; State Key Laboratory of New Drug Discovery and Development for Major Diseases, Gannan Medical University, Ganzhou, China; Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China.
| |
Collapse
|
16
|
Liu MN, Liu ZH, Leng RX, Strijdom H, Weng JP, Xu SW. Revisiting the role of GDF15 in atherosclerosis in mouse and human. Acta Pharmacol Sin 2025:10.1038/s41401-025-01561-3. [PMID: 40307459 DOI: 10.1038/s41401-025-01561-3] [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: 01/17/2025] [Accepted: 04/07/2025] [Indexed: 05/02/2025]
Abstract
Growth differentiation factor 15 (GDF15) is a key regulator of food intake and energy metabolism. GDF15 mimetic drugs for the treatment of metabolic syndrome and obesity are under clinical development. While GDF15 presents a promising target for weight management, its potential cardiovascular actions remain elusive. In this study we investigated the role of GDF15 in macrophage function and atherosclerosis pathogenesis and whether GDF15 acts both as a biomarker and mediator of atherosclerosis severity. ApoE-/- mice were fed a high-cholesterol diet (HCD, 1.25% cholesterol) for 6, 12 or 18 weeks to establish atherosclerotic models. We showed that serum levels of GDF15 were elevated in ApoE-/- mice with atheroprogression; increased serum levels of GDF15 were also observed in patients with coronary artery disease. Enlightened by this finding, we established atherosclerotic model in Gdf15-/- mice by injecting with AAV8-PCSK9D377Y virus and feeding HCD for 12 or 16 weeks. We showed that global Gdf15 knockout, whether in male or female mice, did not alter plaque size in en face aorta, lesion in aortic sinus, size of necrotic core or plaque composition. In macrophage-derived foam cells isolated from atherosclerotic mice, neither Gdf15 deletion nor the treatment with recombinant GDF15 protein (1, 10, 100 ng/mL) affected lipid deposition or macrophage polarization. To translate this finding into a clinically relevant scenario, we performed Mendelian randomization (MR) analysis, and found no significant causal association between circulating GDF15 levels and the incidence of cardiovascular diseases. Furthermore, MR studies suggest that genetic associations between GDF15 and factors such as BMI, ApoB, LDL and HDL were not significant in plasma data from the UK Biobank and the deCODE cohort. In summary, this study demonstrates that global Gdf15 deficiency does not affect the development of atherosclerosis in male or female mice despite the positive association between circulating GDF15 levels and disease progression in mice and human. Thus, GDF15 in circulation is a potential biomarker, but not a causal mediator, of atherosclerosis. Long-term cardiovascular safety of GDF15-targeted therapies warrants further investigation.
Collapse
Affiliation(s)
- Mo-Nan Liu
- Department of Endocrinology, 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, 230001, China
| | - Zheng-Hong Liu
- Department of Endocrinology, 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, 230001, China
| | - Rui-Xue Leng
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, 230032, China
| | - Hans Strijdom
- Centre for Cardio-metabolic Research in Africa, Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Jian-Ping Weng
- Department of Endocrinology, 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, 230001, China
- Anhui Provincial Key Laboratory of Metabolic Health and Panvascular Diseases, Hefei, 230001, China
- Institute of Endocrine and Metabolic Diseases, University of Science and Technology of China, Hefei, 230001, China
| | - Suo-Wen Xu
- Department of Endocrinology, 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, 230001, China.
- Anhui Provincial Key Laboratory of Metabolic Health and Panvascular Diseases, Hefei, 230001, China.
- Institute of Endocrine and Metabolic Diseases, University of Science and Technology of China, Hefei, 230001, China.
| |
Collapse
|
17
|
Wang J, Liu Z, Hu C, Zhao R, Zhu D, Xie Y, Zhang P, Cui M, Xu K, Zhao G, Jin L, Chen X, Suo C, Jiang Y. Healthy lifestyles are associated with alleviating the single-nucleotide polymorphism-based genetic risks of ischaemic stroke, intracerebral haemorrhage and myocardial infarction. Stroke Vasc Neurol 2025; 10:e003257. [PMID: 38925676 DOI: 10.1136/svn-2024-003257] [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: 03/24/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND Both genetic and lifestyle factors contribute to myocardial infarction (MI) and stroke, including ischaemic stroke (IS) and intracerebral haemorrhage (ICH). We explored how and the extent to which a healthy lifestyle, by considering a comprehensive list, could counteract the genetic risk of those diseases, respectively. METHODS 315 044 participants free of stroke and MI at baseline were identified from the UK Biobank. Genetic risk scores (GRS) for those diseases were constructed separately and categorised as low, intermediate and high by tertile. Lifestyle risk scores (LRS) were constructed separately using smoking, alcohol intake, physical activity, dietary patterns and sleep patterns. Similarly, participants were categorised into low, intermediate and high LRS. The data were analysed using Cox proportional hazard models. RESULTS Over a median follow-up of 12.8 years, 4642, 1046 and 9485 participants developed IS, ICH and MI, respectively. Compared with participants with low levels of GRS and LRS, the HRs of those with high levels of GRS and LRS were 3.45 (95% CI 2.71 to 4.41), 2.32 (95% CI 1.40 to 3.85) and 4.89 (95% CI 4.16 to 5.75) for IS, ICH and MI, respectively. Moreover, among participants with high GRS, the standardised 14-year rates of IS events were 4.40% (95% CI 3.45% to 5.36%) among those with high LRS. In contrast, it is only 1.78% (95% CI 1.63% to 1.94%) among those with low LRS. Similarly for MI, the high LRS group had standardised rates of 8.60% (95% CI 7.38% to 9.81%), compared with 3.34% (95% CI 3.12% to 3.56%) in low LRS. Among the high genetic risk group of ICH, the rate is reduced by about half compared low LRS to high LRS, although the rate was low for both (0.36% (95% CI 0.31% to 0.42%) and 0.71% (95% CI 0.36% to 1.05%), respectively). CONCLUSION Healthy lifestyles were substantially associated with a reduction in the risk of IS, ICH and MI and attenuated the genetic risk of IS, ICH and MI by at least half, respectively.
Collapse
Affiliation(s)
- Jingru Wang
- Department of Epidemiology and Ministry of Education Key Laboratory of Public Health Safety, School of Public Health, Fudan University, Shanghai, Shanghai, China
| | - Zhenqiu Liu
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, Shanghai, China
- Fudan University Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China
| | - Chengxin Hu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Renjia Zhao
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, Shanghai, China
| | - Dongliang Zhu
- Department of Epidemiology and Ministry of Education Key Laboratory of Public Health Safety, School of Public Health, Fudan University, Shanghai, Shanghai, China
| | - Yijing Xie
- Department of Epidemiology and Ministry of Education Key Laboratory of Public Health Safety, School of Public Health, Fudan University, Shanghai, Shanghai, China
| | - Pengyan Zhang
- Department of Epidemiology and Ministry of Education Key Laboratory of Public Health Safety, School of Public Health, Fudan University, Shanghai, Shanghai, China
| | - Mei Cui
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, Shanghai, China
| | - Kelin Xu
- Department of Biostatistics, School of Public Health, Fudan University, Shanghai, Shanghai, China
| | - Genming Zhao
- Department of Epidemiology and Ministry of Education Key Laboratory of Public Health Safety, School of Public Health, Fudan University, Shanghai, Shanghai, China
| | - Li Jin
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, Shanghai, China
- Fudan University Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China
| | - Xingdong Chen
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, Shanghai, China
- Fudan University Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China
- Yiwu Research Institute of Fudan University, Yiwu, Zhejiang, China
| | - Chen Suo
- Department of Epidemiology and Ministry of Education Key Laboratory of Public Health Safety, School of Public Health, Fudan University, Shanghai, Shanghai, China
- Fudan University Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China
- Shanghai Institute of Infectious Disease and Biosecurity, Shanghai, Shanghai, China
| | - Yanfeng Jiang
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, Shanghai, China
- Fudan University Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China
| |
Collapse
|
18
|
Tao Y, Liu Q, Wang Y, Gong Y, Xu M, Jiang R, Zhang K. Multiomics insight into the role of glucagon-like peptide-1 receptor agonists in heart failure. ESC Heart Fail 2025. [PMID: 40290076 DOI: 10.1002/ehf2.15310] [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: 11/04/2024] [Revised: 02/22/2025] [Accepted: 04/08/2025] [Indexed: 04/30/2025] Open
Abstract
AIMS Cardiovascular diseases, such as atrial fibrillation, coronary artery disease, heart failure (HF) and ischaemic stroke, are leading causes of death globally and exert major global health burden. Recent studies suggest that glucagon-like peptide-1 receptor agonists (GLP1Ra), a novel class of antidiabetic drugs, may not only help manage blood glucose but also reduce the risks of cardiovascular diseases. However, the mechanisms through which GLP1Ra protects against cardiovascular diseases, remain incompletely understood. METHODS AND RESULTS Genome-wide association study dates were used to investigate the impact of GLP1Ra on cardiovascular diseases, including atrial fibrillation (1,202,168 European and 28,612 East Asian), coronary artery disease (501,756 European and 183,134 East Asian), heart failure (HF) (1,350,497 European and 203,040 East Asian) and ischaemic stroke (689,168 European and 192,383 East Asian). Genetic instruments were selected from the GLP1R gene region, and two-sample Mendelian randomization (MR) analyses were conducted to assess the causal effects of GLP1Ra on cardiovascular diseases in European and East Asian populations. Summary-data-based MR analyses were performed for further validation using expression quantitative trait loci data. Mediation analysis evaluates the role of circulating inflammatory proteins and metabolites in mediating the effects of GLP1Ra. Meantime, we performed a series of sensitivity analyses to confirm the robustness of the results. The result demonstrated significant causal association between GLP1Ra and HF in European populations (odds ratio: 0.60, 95% CI 0.51-0.72, P = 2.76 × 10-8), and this result was confirmed by SMR analyses. No significant associations were found for atrial fibrillation, coronary artery disease, ischaemic stroke in European populations or all cardiovascular diseases in East Asian populations. GLP1Ra was found to influence 27 circulating inflammatory proteins and 146 circulating metabolites. Among them, 4 inflammatory proteins and 17 metabolites are associated with HF. Mediation analysis indicated that the protective effect of GLP1Ra on HF was mediated by circulating fibroblast growth factor 5 (FGF5) and N-acetylglycine (NAC), with a mediated proportion of 4.37% and 8% of the total effect, respectively. CONCLUSIONS This study provides multiomics insight into the role of GLP1Ra in cardiovascular diseases, especially in HF and the underlying pathway. The results suggest that GLP1Ra may exert anti-HF effects by reducing the concentration of circulating FGF5 and increasing the levels of circulating NAC, and enriches the potential mechanisms through which GLP1Ra alleviates HF.
Collapse
Affiliation(s)
- Yuhang Tao
- Department of Cardiology, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Zhejiang, Hangzhou, China
| | - Qi Liu
- Department of Cardiology, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Zhejiang, Hangzhou, China
| | - Yuxing Wang
- Department of Emergency Medicine, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yingchao Gong
- Department of Cardiology, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Zhejiang, Hangzhou, China
| | - Mingying Xu
- Department of General Practice, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, Zhejiang, China
| | - Ruhong Jiang
- Department of Cardiology, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Zhejiang, Hangzhou, China
| | - Kai Zhang
- Department of Cardiology, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Zhejiang, Hangzhou, China
- Department of General Practice, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
19
|
Titova OE, Yuan S, Larsson SC. Applying Mendelian randomization to study the relationship between circulating proteins and myocardial infarction. Eur Heart J 2025:ehaf187. [PMID: 40249358 DOI: 10.1093/eurheartj/ehaf187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/19/2025] Open
Affiliation(s)
- Olga E Titova
- Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Dag Hammarskjölds väg 14 B, 75185 Uppsala, Sweden
| | - Shuai Yuan
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Susanna C Larsson
- Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Dag Hammarskjölds väg 14 B, 75185 Uppsala, Sweden
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
20
|
Lu C, Wang X, Chen X, Qin T, Ye P, Liu J, Wang S, Luo W. Causal Analysis Between Gut Microbes, Aging Indicator, and Age-Related Disease, Involving the Discovery and Validation of Biomarkers. Aging Cell 2025:e70057. [PMID: 40202110 DOI: 10.1111/acel.70057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 03/13/2025] [Accepted: 03/18/2025] [Indexed: 04/10/2025] Open
Abstract
The influence of gut microbes on aging has been reported in several studies, but the mediating pathways of gut microbiota, whether there is a causal relationship between the two, and biomarker screening and validation have not been fully discussed. In this study, Mendelian Randomization (MR) and Linkage Disequilibrium Score Regression (LDSC) are used to systematically investigate the associations between gut microbiota, three aging indicators, and 14 age-related diseases. Additionally, this study integrates machine learning algorithms to explore the potential of MR and LDSC methods for biomarker screening. Gut microbiota is found to be a potential risk factor for 14 age-related diseases. The causal effects of gut microbiota on chronic kidney disease, cirrhosis, and heart failure are partially mediated by aging indicators. Additionally, gut microbiota identified through MR and LDSC methods exhibit biomarker properties for disease prediction (average AUC = 0.731). These methods can serve as auxiliary tools for conventional biomarker screening, effectively enhancing the performance of disease models (average AUC increased from 0.808 to 0.832). This study provides evidence that supports the association between the gut microbiota and aging and highlights the potential of genetic correlation and causal relationship analysis in biomarker discovery. These findings may help to develop new approaches for healthy aging detection and intervention.
Collapse
Affiliation(s)
- Chunrong Lu
- AIage Life Science Corporation Ltd., Guangxi Free Trade Zone, Aisheng Biotechnology Corporation Ltd., Nanning, Guangxi, China
| | - Xiaojun Wang
- AIage Life Science Corporation Ltd., Guangxi Free Trade Zone, Aisheng Biotechnology Corporation Ltd., Nanning, Guangxi, China
- Guangxi Key Laboratory of Longevity Science and Technology, Nanning, Guangxi, P.R. China
| | - Xiaochun Chen
- AIage Life Science Corporation Ltd., Guangxi Free Trade Zone, Aisheng Biotechnology Corporation Ltd., Nanning, Guangxi, China
- Guangxi Key Laboratory of Longevity Science and Technology, Nanning, Guangxi, P.R. China
| | - Tao Qin
- AIage Life Science Corporation Ltd., Guangxi Free Trade Zone, Aisheng Biotechnology Corporation Ltd., Nanning, Guangxi, China
| | - Pengpeng Ye
- AIage Life Science Corporation Ltd., Guangxi Free Trade Zone, Aisheng Biotechnology Corporation Ltd., Nanning, Guangxi, China
- Guangxi Key Laboratory of Longevity Science and Technology, Nanning, Guangxi, P.R. China
| | - Jianqun Liu
- AIage Life Science Corporation Ltd., Guangxi Free Trade Zone, Aisheng Biotechnology Corporation Ltd., Nanning, Guangxi, China
| | - Shuai Wang
- AIage Life Science Corporation Ltd., Guangxi Free Trade Zone, Aisheng Biotechnology Corporation Ltd., Nanning, Guangxi, China
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Weifei Luo
- AIage Life Science Corporation Ltd., Guangxi Free Trade Zone, Aisheng Biotechnology Corporation Ltd., Nanning, Guangxi, China
- Guangxi Key Laboratory of Longevity Science and Technology, Nanning, Guangxi, P.R. China
| |
Collapse
|
21
|
He X, Li L, Zhou D, Yan Z, Liu M, Yun L. Risk Factors and Genetic Insights into Coronary Artery Disease-Related Sudden Cardiac Death: A Molecular Analysis of Forensic Investigation. Int J Mol Sci 2025; 26:3470. [PMID: 40331954 PMCID: PMC12027380 DOI: 10.3390/ijms26083470] [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: 02/21/2025] [Revised: 04/02/2025] [Accepted: 04/03/2025] [Indexed: 05/08/2025] Open
Abstract
Sudden cardiac death (SCD) is a major cause of mortality among patients with coronary artery disease (CAD). This study aimed to identify risk factors for CAD-related SCD (SCDCAD) through autopsy data and genetic screening with a particular emphasis on rare variants (minor allele frequency < 0.01). We included 241 SCDCAD cases (mean age 54.6 ± 12.8 years, 74.7% male) verified by medico-legal examination and 241 silent CAD controls (mean age 53.6 ± 15.2 years, 25.3% female) who died from severe craniocerebral trauma. Information about death characteristics was obtained from questionnaires, police reports and autopsy data. Whole-exome sequencing was performed on myocardial tissue samples. Polygenic risk score (PRS) from a previously validated model was applied and rare variant pathogenicity was predicted using in silico tools. SCDCAD victims predominantly died at night and showed higher mortality rates during summer and winter months, with more complex coronary disease. Nocturnal time (adjusted odds ratio [AOR] = 3.53, 95% CI: 2.37-5.25, p < 0.001), winter (AOR = 2.06, 95% CI: 1.33-3.20, p = 0.001), multiple vessel occlusion (AOR = 1.79, 95% CI: 1.16-2.77, p = 0.009), right coronary artery stenosis (AOR = 2.38, 95% CI: 1.54-3.68, p < 0.001) and unstable plaque (AOR = 2.17, 95% CI: 1.46-3.23, p < 0.001) were identified as risk factors of SCDCAD. The PRS score was associated with a 60% increased risk of SCDCAD (OR = 1.632 per SD, 95%CI: 1.631-1.633, p < 0.001). Genetic analysis identified MUC19 and CGN as being associated with SCDCAD. We identified both hereditary and acquired risk factors that may contribute to cardiac dysfunction and precipitate SCD in CAD patients, thereby facilitating the prevention and early recognition of high-risk individuals.
Collapse
Affiliation(s)
| | | | | | | | | | - Libing Yun
- Department of Forensic Medicine, West China School of Basic Medical Sciences & Forensic Science, Sichuan University, Chengdu 610041, China
| |
Collapse
|
22
|
Zhou S, Zhou P, Yang T, Si J, An W, Jiang Y. Glucosamine supplementation contributes to reducing the risk of type 2 diabetes: Evidence from Mendelian randomization combined with a meta-analysis. J Int Med Res 2025; 53:3000605251334460. [PMID: 40300556 PMCID: PMC12041707 DOI: 10.1177/03000605251334460] [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: 03/21/2025] [Indexed: 05/01/2025] Open
Abstract
ObjectiveObservational studies on glucosamine supplementation and type 2 diabetes risk have shown inconsistent results, necessitating the use of Mendelian randomization to clarify the true causal relationship.MethodsThe glucosamine supplementation-related genome-wide association study dataset was obtained from the MRC Integrative Epidemiology Unit consortium, whereas type 2 diabetes-related genome-wide association study datasets were obtained from the FinnGen consortium (discovery) and Xue et al.'s meta-analysis (validation). Two-sample Mendelian randomization analyses were performed separately in the discovery and validation datasets, followed by meta-analysis and multivariable Mendelian randomization analyses to verify the robustness of the results of two-sample Mendelian randomization. The estimation of the causal relationship was conducted through the inverse variance weighted method.ResultsGlucosamine supplementation exhibited a significant protective effect against type 2 diabetes, as identified by two-sample Mendelian randomization analysis in the FinnGen consortium (odds ratio: 0.13, 95% confidence interval: 0.02-0.89) and validated in Xue et al.'s meta-analysis (odds ratio: 0.06, 95%; confidence interval: 0.01-0.29). A combined meta-analysis (odds ratio: 0.08, 95%; confidence interval: 0.02-0.27) of the results of two-sample Mendelian randomization confirmed the robustness of these findings. Additionally, multivariable Mendelian randomization analysis (odds ratio: 0.12, 95%; confidence interval: 0.02-0.94), after adjusting for confounding factors, supported the results of two-sample Mendelian randomization. No evidence of heterogeneity or pleiotropy was observed.ConclusionOverall, our results revealed that genetically predicted glucosamine supplementation was inversely associated with the risk of type 2 diabetes, highlighting the potential importance of glucosamine supplementation in preventing type 2 diabetes.
Collapse
Affiliation(s)
- Shuai Zhou
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, Genetic Diagnosis Center, The First Hospital of Jilin University, China
| | - Peiwen Zhou
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, Genetic Diagnosis Center, The First Hospital of Jilin University, China
| | - Tianshi Yang
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, Genetic Diagnosis Center, The First Hospital of Jilin University, China
| | - Junzhuo Si
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, Genetic Diagnosis Center, The First Hospital of Jilin University, China
| | - Wenyan An
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, Genetic Diagnosis Center, The First Hospital of Jilin University, China
| | - Yanfang Jiang
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, Genetic Diagnosis Center, The First Hospital of Jilin University, China
| |
Collapse
|
23
|
Zhang P, Wang W, Xu Q, Cui J, Zhu M, Li Y, Liu Y, Liu Y. Genetic association of circulating lipids and lipid-lowering drug targets with vascular calcification. Atherosclerosis 2025; 403:119136. [PMID: 39985880 DOI: 10.1016/j.atherosclerosis.2025.119136] [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: 08/16/2024] [Revised: 01/27/2025] [Accepted: 02/16/2025] [Indexed: 02/24/2025]
Abstract
BACKGROUND AND AIMS Vascular calcification (VC) significantly increases the incidence and mortality of many diseases. The causal relationships of dyslipidaemia and lipid-lowering drug use with VC severity remain unclear. This study explores the genetic causal associations of different circulating lipids and lipid-lowering drug targets with coronary artery calcification (CAC) and abdominal aortic artery calcification (AAC). METHODS We obtained single-nucleotide polymorphisms (SNPs) and expression quantitative trait loci (eQTLs) associated with seven circulating lipids and 13 lipid-lowering drug targets from publicly available genome-wide association studies and eQTL databases. Causal associations were investigated by univariable, multivariable, drug-target, and summary data-based Mendelian randomization (MR) analyses. Potential mediation effects of metabolic risk factors were evaluated. RESULTS MR analysis revealed that genetic proxies for low-density lipoprotein cholesterol (LDL-C), triglycerides (TC) and Lipoprotein (a) (Lp(a)) were causally associated with CAC severity, and apolipoprotein B (apoB) level was causally associated with AAC severity. A significant association was detected between hepatic Lipoprotein(A) (LPA) gene expression and CAC severity. Colocalisation analysis supported the hypothesis that the association between LPA expression and CAC quantity is driven by different causal variant sites within the ±1 Mb flanking region of LPA. Serum calcium and phosphorus had causal associations with CAC severity. CONCLUSIONS Inhibitors targeting LPA might represent CAC drug candidates. Moreover, T2DM, hypercalcemia, and hyperphosphatemia are positively causally associated with CAC severity, while chronic kidney disease and estimated glomerular filtration rate are not.
Collapse
Affiliation(s)
- Pengfei Zhang
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Wenting Wang
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Qian Xu
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Jing Cui
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Mengmeng Zhu
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Yiwen Li
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Yanfei Liu
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, 100091, China; The Second Department of Gerontology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Yue Liu
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, 100091, China.
| |
Collapse
|
24
|
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
|
25
|
Fang Q, Fan H, Li Q, Zhang M, Zhou Z, Du J, Huang J. Multi-Omic Insight Into the Molecular Networks in the Pathogenesis of Coronary Artery Disease. J Am Heart Assoc 2025; 14:e037203. [PMID: 40135555 DOI: 10.1161/jaha.124.037203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 01/17/2025] [Indexed: 03/27/2025]
Abstract
BACKGROUND Genome-wide association studies have revealed numerous loci associated with coronary artery disease (CAD). However, some potential causal/risk genes remain unidentified, and causal therapies are lacking. METHODS AND RESULTS We integrated multi-omics data from gene methylation, expression, and protein levels using summary data-based Mendelian randomization and colocalization analysis. Candidate genes were prioritized based on protein-level associations, colocalization probability, and links to methylation and expression. Single-cell RNA sequencing data were used to assess differential expression in the coronary arteries of patients with CAD. TAGLN2 (Transgelin 2), APOB (Apolipoprotein B), and GIP (Glucose-dependent insulinotropic polypeptide) were identified as the genes most strongly associated with CAD, with TAGLN2 exhibiting the most significant association. Higher methylation levels of TAGLN2 at specific Cytosine-phosphate-Guanine sites were negatively correlated with its gene expression and associated with a lower risk of CAD, whereas higher circulating TAGLN2 protein levels were positively associated with CAD risk (odds ratio,1.66 [95% CI, 1.32-2.08). These results suggest distinct regulatory mechanisms for TAGLN2. In contrast, APOB and GIP showed positive associations with CAD risk, whereas DHX58 (DExH-box helicase 58) and SWAP70 (Switch-associated protein 70) were associated with decreased risk. CONCLUSIONS Our findings provide multi-omics evidence suggesting that TAGLN2, APOB, GIP, DHX58, and SWAP70 genes are associated with CAD risk. This work provides novel insights into the molecular mechanisms of CAD and highlights the potential of integrating multi-omics data to uncover potential causal relationships that cannot be fully captured by traditional genome-wide association studies.
Collapse
Affiliation(s)
- Qinghua Fang
- Department of Cardiology The Second Affiliated Hospital of Chongqing Medical University Chongqing China
| | - Hongdan Fan
- Department of Hepatobiliary Surgery The Second Affiliated Hospital of Chongqing Medical University Chongqing China
| | - Qiaoqiao Li
- Department of Cardiology The Second Affiliated Hospital of Chongqing Medical University Chongqing China
| | - Muzi Zhang
- Department of Cardiology The Second Affiliated Hospital of Chongqing Medical University Chongqing China
| | - Zhengzhong Zhou
- Department of Cardiology The Second Affiliated Hospital of Chongqing Medical University Chongqing China
| | - Jianlin Du
- Department of Cardiology The Second Affiliated Hospital of Chongqing Medical University Chongqing China
| | - Jing Huang
- Department of Cardiology The Second Affiliated Hospital of Chongqing Medical University Chongqing China
| |
Collapse
|
26
|
Choi J, Wen W, Jia G, Tao R, Long J, Shu XO, Zheng W. Associations of Blood Lipid-Related Polygenic Scores, Lifestyle Factors and Their Combined Effects with Risk of Coronary Artery Disease in the UK Biobank Cohort. J Cardiovasc Transl Res 2025; 18:331-340. [PMID: 39680354 DOI: 10.1007/s12265-024-10578-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 11/29/2024] [Indexed: 12/17/2024]
Abstract
Circulating lipids play a crucial role in the development of coronary artery disease (CAD). However, it is unclear whether the genetic susceptibility to hyperlipidemia may interact with lifestyle factors in CAD risk. Using UK Biobank data from 328,606 participants, we evaluated combined effects of genetic susceptibility to hyperlipidemia and lifestyle factors with risk of CAD. We found that both blood lipid-related polygenic score (PGS) and healthy lifestyle score (HLS) are independently associated with CAD risk, and individuals with the highest-risk lipid-related PGS and the least healthy HLS had the highest CAD risk. This association was stronger in younger (< 60 years, hazard ratio: 4.46, 95% confidence interval: 3.44-5.78) than older adults (2.54, 2.13-3.03). Our study suggests that individuals, particularly younger adults, with higher-risk PGSs of blood lipid traits would benefit more substantially by adherence to a healthy lifestyle than those with lower PGSs.
Collapse
Affiliation(s)
- Jungyoon Choi
- Division of Oncology, Department of Internal Medicine, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-Do, Korea
| | - Wanqing Wen
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Guochong Jia
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ran Tao
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jirong Long
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Xiao-Ou Shu
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Wei Zheng
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
27
|
Khan TG, Bragazzi Cunha J, Raut C, Burroughs M, Vyas HS, Leix K, Goonewardena SN, Smrcka AV, Speliotes EK, Emmer BT. Functional interrogation of cellular Lp(a) uptake by genome-scale CRISPR screening. Atherosclerosis 2025; 403:119174. [PMID: 40174266 PMCID: PMC12011201 DOI: 10.1016/j.atherosclerosis.2025.119174] [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: 07/18/2024] [Revised: 02/04/2025] [Accepted: 03/07/2025] [Indexed: 04/04/2025]
Abstract
BACKGROUND AND AIMS An elevated level of lipoprotein(a), or Lp(a), in the bloodstream has been causally linked to the development of atherosclerotic cardiovascular disease and calcific aortic valve stenosis. Steady state levels of circulating lipoproteins are modulated by their rate of clearance, but the identity of the Lp(a) uptake receptor(s) has been controversial. METHODS We performed a genome-scale CRISPR screen to functionally interrogate all potential Lp(a) uptake regulators in HuH7 cells. Screen validation was performed by single gene disruption and overexpression. Direct binding between purified lipoproteins and recombinant protein was tested using biolayer interferometry. An association between human genetic variants and circulating Lp(a) levels was analyzed in the UK Biobank cohort. RESULTS The top positive and negative regulators of Lp(a) uptake in our screen were LDLR and MYLIP, encoding the LDL receptor and its ubiquitin ligase IDOL, respectively. We also found a significant correlation for other genes with established roles in LDLR regulation. No other gene products, including those previously proposed as Lp(a) receptors, exhibited a significant effect on Lp(a) uptake in our screen. We validated the functional influence of LDLR expression on HuH7 Lp(a) uptake, confirmed in vitro binding between the LDLR extracellular domain and purified Lp(a), and detected an association between loss-of-function LDLR variants and increased circulating Lp(a) levels in the UK Biobank cohort. CONCLUSIONS Our findings support a central role for the LDL receptor in mediating Lp(a) uptake by hepatocytes.
Collapse
Affiliation(s)
- Taslima G Khan
- Program in Chemical Biology, University of Michigan, Ann Arbor, MI, USA
| | - Juliana Bragazzi Cunha
- Division of Hospital Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Chinmay Raut
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA; Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Michael Burroughs
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, USA
| | - Hitarthi S Vyas
- Division of Hospital Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Kyle Leix
- Division of Hospital Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Sascha N Goonewardena
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA; Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI, USA
| | - Alan V Smrcka
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, USA
| | - Elizabeth K Speliotes
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA; Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Brian T Emmer
- Division of Hospital Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA; Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
28
|
Ao L, van Heemst D, Jukema JW, Rensen PCN, Willems van Dijk K, Noordam R. Potential causal evidence for an ApoB-independent and HDL-related risk profile associated with coronary artery disease. J Lipid Res 2025; 66:100778. [PMID: 40089107 PMCID: PMC12000745 DOI: 10.1016/j.jlr.2025.100778] [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: 05/14/2024] [Revised: 02/09/2025] [Accepted: 03/12/2025] [Indexed: 03/17/2025] Open
Abstract
Plasma 1H-NMR metabolomic measures have yielded significant insight into the pathophysiology of cardiometabolic disease, but their inter-related nature complicates causal inference and clinical interpretation. This study aimed to investigate the associations of unrelated 1H-NMR metabolomic profiles with coronary artery disease (CAD) and ischemic stroke (IS). Principal component (PC) analysis was performed on 168 1H-NMR metabolomic measures in 56,712 unrelated European participants from UK Biobank to retrieve uncorrelated PCs, which were used in Cox-proportional hazard models. For each outcome, two-sample Mendelian randomization analyses were then conducted based on three nonoverlapping databases, followed by a meta-analysis. The first six PCs collectively explaining 88% of the total variance were identified. For CAD, results from Cox and Mendelian randomization analyses were generally directionally consistent. The pooled odds ratios (95% CI) for CAD per one-SD increase in genetically influenced PC1 and PC3 (both characterized by distinct apolipoprotein B [ApoB]-associated lipoprotein profiles) were 1.04 (1.03, 1.05) and 0.94 (0.93, 0.96), respectively. Besides, the pooled odds ratio (95% CI) for CAD per one-SD increase in genetically influenced PC4, characterized by simultaneously decreased small HDL and increased large HDL, and independent of ApoB, was 1.05 (1.03, 1.07). For IS, increases of PC3 and PC5 (characterized by increased amino acids) were associated with a lower risk and a higher risk, respectively. This study confirms associations of ApoB-associated lipoprotein profiles with CAD and IS, and highlights the possible existence of an ApoB-independent lipoprotein profile, characterized by a distinctive HDL subparticle distribution, driving CAD.
Collapse
Affiliation(s)
- Linjun Ao
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands.
| | - Diana van Heemst
- Section of Gerontology and Geriatrics, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, the Netherlands; Netherlands Heart Institute, Utrecht, the Netherlands
| | - Patrick C N Rensen
- Division of Endocrinology, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Ko Willems van Dijk
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands; Division of Endocrinology, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Raymond Noordam
- Section of Gerontology and Geriatrics, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
29
|
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
|
30
|
Wei L, Ding E, Lu D, Rui Z, Shen J, Fan G. Assessing the effect of modifiable risk factors on hepatocellular carcinoma: evidence from a bidirectional Mendelian randomization analysis. Discov Oncol 2025; 16:437. [PMID: 40164825 PMCID: PMC11958933 DOI: 10.1007/s12672-025-02177-z] [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: 12/12/2024] [Accepted: 03/18/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND The pathogenesis of hepatocellular carcinoma (HCC) involves a variety of environmental risk factors, some of which have yet to be fully clarified. Using the Mendelian randomization (MR) approach, this study comprehensively investigates the causal effect of genetically predicted modifiable risk factors on HCC. METHODS Genetic variants related to the 50 risk factors that had been identified in previous research were derived from genome-wide association studies. Summary statistics for the discovery cohort and validation cohort of HCC were sourced from the FinnGen consortium and the UK Biobank, respectively. Bidirectional MR analysis and sensitivity analysis were performed to establish causative risk factors for HCC. RESULTS Through the inverse variance weighted method, the results of the discovery cohort indicated that waist circumference, nonalcoholic fatty liver disease (NAFLD), alanine aminotransferase (ALT) levels, and aspartate aminotransferase (AST) levels were significantly linked to HCC occurrence risk. Furthermore, body fat percentage, glycated hemoglobin (HbA1c), obesity class 1-3, waist-to-hip ratio, iron, ferritin, transferrin saturation, and urate had suggestive associations with HCC. The validation cohort further confirmed that NAFLD and ALT levels were strongly related to HCC. Reverse MR indicated that genetic susceptibility to HCC was connected to NAFLD and transferrin saturation. Sensitivity analyses showed that most of the findings were robust. CONCLUSION This MR study delivers evidence of the complex causal relationship between modifiable risk factors and HCC. These findings offer new insights into potential prevention and treatment strategies for HCC.
Collapse
Affiliation(s)
- Lijuan Wei
- Department of Nuclear Medicine, Tianjin First Central Hospital, Tianjin, China
| | - Enci Ding
- Department of Nuclear Medicine, Tianjin First Central Hospital, Tianjin, China
| | - Dongyan Lu
- Department of Nuclear Medicine, Tianjin First Central Hospital, Tianjin, China
| | - Zhongying Rui
- Department of Nuclear Medicine, Tianjin First Central Hospital, Tianjin, China
| | - Jie Shen
- Department of Nuclear Medicine, Tianjin First Central Hospital, Tianjin, China
| | - Guoju Fan
- Department of Vascular Surgery, The Second Hospital of Tianjin Medical University, No. 23, Pingjiang Road, Hexi District, Tianjin, 300211, China.
| |
Collapse
|
31
|
Uffelmann E, Price AL, Posthuma D, Peyrot WJ. Estimating Disorder Probability Based on Polygenic Prediction Using the BPC Approach. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2024.01.12.24301157. [PMID: 38260678 PMCID: PMC10802765 DOI: 10.1101/2024.01.12.24301157] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Polygenic Scores (PGSs) summarize an individual's genetic propensity for a given trait in a single value based on SNP effect sizes derived from Genome-Wide Association Study (GWAS) results. Methods have been developed that apply Bayesian approaches to improve the prediction accuracy of PGSs through optimization of estimated effect sizes. While these methods are generally well-calibrated for continuous traits (implying the predicted values are, on average, equal to the true trait values), they are not well-calibrated for binary disorder traits in ascertained samles. This is a problem because well-calibrated PGSs are needed to reliably compute the absolute disorder probability for an individual to facilitate future clinical implementation. Here, we introduce the Bayesian polygenic score Probability Conversion (BPC) approach, which computes an individual's predicted disorder probability using GWAS summary statistics, an existing Bayesian PGS method (e.g., PRScs, SBayesR), the individual's genotype data, and a prior disorder probability (which can be specified flexibly, based on e.g., literature, small reference samples, or prior elicitation). The BPC approach transforms the PGS to its underlying liability scale, computes the variances of the PGS in cases and controls, and applies Bayes' Theorem to compute the absolute disorder probability; it is practical in its application as it does not require a tuning sample with both genotype and phenotype data. We applied the BPC approach to extensive simulated data and empirical data of nine disorders. The BPC approach yielded well-calibrated results that were consistently better than the results of another recently published approach.
Collapse
|
32
|
Aminbakhsh AP, Théberge ET, Burden E, Adejumo CK, Gravely AK, Lehman A, Sedlak TL. Exploring associations between estrogen and gene candidates identified by coronary artery disease genome-wide association studies. Front Cardiovasc Med 2025; 12:1502985. [PMID: 40182431 PMCID: PMC11965610 DOI: 10.3389/fcvm.2025.1502985] [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: 09/27/2024] [Accepted: 03/04/2025] [Indexed: 04/05/2025] Open
Abstract
Introduction Coronary artery disease (CAD) is the leading cause of death around the world, with epidemiological sex and gender differences in prevalence, pathophysiology and outcomes. It has been hypothesized that sex steroids, like estrogen, may contribute to these sex differences. There is a relatively large genetic component to developing CAD, with heritability estimates ranging between 40%-60%. In the last two decades, genome-wide association studies (GWAS) have contributed substantially to advancing the understanding of genetic candidates contributing to CAD. The aim of this study was to determine if genes discovered in CAD GWASs are affected by estrogen via direct modulation or indirect down-stream targets. Methods A scoping review was conducted using MEDLINE and EMBASE for studies of atherosclerotic coronary artery disease and a genome-wide association study (GWAS) design. Analysis was limited to candidate genes with corresponding single nucleotide polymorphisms (SNPs) surpassing genome-wide significance and had been mapped to genes by study authors. The number of studies that conducted sex-stratified analyses with significant genes were quantified. A literature search of the final gene lists was done to examine any evidence suggesting estrogen may modulate the genes and/or gene products. Results There were 60 eligible CAD GWASs meeting inclusion criteria for data extraction. Of these 60, only 36 had genome-wide significant SNPs reported, and only 3 of these had significant SNPs from sex-stratified analyses mapped to genes. From these 36 studies, a total of 61 genes were curated, of which 26 genes (43%) were found to have modulation by estrogen. All 26 were discovered in studies that adjusted for sex. 12/26 genes were also discovered in studies that conducted sex-stratified analyses. 12/26 genes were classified as having a role in lipid synthesis, metabolism and/or lipoprotein mechanisms, while 11/26 were classified as having a role in vascular integrity, and 3/26 were classified as having a role in thrombosis. Discussion This study provides further evidence of the relationship between estrogen, genetic risk and the development of CAD. More sex-stratified research will need to be conducted to further characterize estrogen's relation to sex differences in the pathology and progression of CAD.
Collapse
Affiliation(s)
- Ava P. Aminbakhsh
- Department of Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Emilie T. Théberge
- Department of Medical Genetics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Elizabeth Burden
- Division of Internal Medicine, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
- Vancouver Coastal Health, Vancouver, BC, Canada
| | - Cindy Kalenga Adejumo
- Division of Internal Medicine, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
- Vancouver Coastal Health, Vancouver, BC, Canada
| | - Annabel K. Gravely
- Department of Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Anna Lehman
- Department of Medical Genetics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- Vancouver Coastal Health, Vancouver, BC, Canada
| | - Tara L. Sedlak
- Vancouver Coastal Health, Vancouver, BC, Canada
- Division of Cardiology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
33
|
Wang T, Yao Y, Gao X, Luan H, Wang X, Liu L, Sun C. Genetic association of lipids and lipid-lowering drug target genes with breast cancer. Discov Oncol 2025; 16:331. [PMID: 40095250 PMCID: PMC11914663 DOI: 10.1007/s12672-025-02041-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 03/03/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND Although several preclinical and epidemiological studies have shown that blood lipids and lipid-lowering drugs can reduce the risk of breast cancer, this finding remains controversial. This study aimed to explore the causal relationship between dyslipidemia,lipid-lowering drugs, and breast cancer. We also aimed to evaluate the potential impact of lipid-lowering drug targets on breast cancer. METHOD Data of 431 lipid- and lipid-related phenotypes were obtained from genome-wide association study (GWAS), and mendelian randomization (MR) analyses were performed using two independent breast cancer datasets as endpoints. Genetic variants associated with genes encoding lipid-lowering drug targets were extracted from the Global Lipid Genetics Consortium. Expression quantitative trait loci data in relevant tissues were used to further validate lipid-lowering drug targets that reached significance and combined with bioinformatics approaches for molecular expression and prognostic exploration. Further mediation analyses were performed to explore potential mediators. RESULT In two independent datasets, phosphatidylcholine (18:1_0:0 levels) was associated with breast cancer risk (discovery: odds ratio (OR) = 1.255 [95% confidence interval (CI) 1.120-1.406]; p = 8.936 × 10-5, replication: OR = 1.016 [95% CI, 1.003-1.030]; p = 0.017), HMG- CoA reductase (HMGCR) inhibition was genetically modeled and associated with a reduced risk of breast cancer (discovery: OR = 0.833 [95% CI 0.752-0.923], p = 5.12 × 10-4; replication: OR = 0.975 [95% CI 0.960-0.990], p = 1.65 × 10-3). There was a significant MR correlation between HMGCR expression in whole blood and breast cancer (OR = 1.11 [95% 1.01-1.22] p = 0.04). Bioinformatics analysis revealed that HMGCR expression higher in breast cancer tissues than in normal tissues, along with poor overall survival and relapse-free survival, and was associated with multiple immune cell infiltration. Finally, the mediation analysis showed that HMGCR inhibitors affected breast cancer through different immune cell phenotypes and C-reactive protein levels. CONCLUSION In this study, we found for the first time that phosphatidylcholine (18:1_0:0) levels are associated with breast cancer risk. We found that HMGCR inhibitors are associated with a reduced risk of breast cancer, and part of their action may be through pathways other than lipid-lowering, including modulation of immune function and reduction of inflammation represented by C-reactive protein levels.
Collapse
Affiliation(s)
- Tianhua Wang
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yan Yao
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, China
| | - Xinhai Gao
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Hao Luan
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xue Wang
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang, China
| | - Lijuan Liu
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China.
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, China.
| | - Changgang Sun
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, China.
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang, China.
| |
Collapse
|
34
|
Shan H, Fan S, Li Q, Liang R, Chen Z, Wang S, Wang X, Li Y, Chen S, Yu K, Fei T. Systematic interrogation of functional genes underlying cholesterol and lipid homeostasis. Genome Biol 2025; 26:59. [PMID: 40098013 PMCID: PMC11912599 DOI: 10.1186/s13059-025-03531-8] [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: 08/07/2024] [Accepted: 03/06/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND Dyslipidemia or hypercholesterolemia are among the main risk factors for cardiovascular diseases. Unraveling the molecular basis of lipid or cholesterol homeostasis would help to identify novel drug targets and develop effective therapeutics. RESULTS Here, we adopt a systematic approach to catalog the genes underlying lipid and cholesterol homeostasis by combinatorial use of high-throughput CRISPR screening, RNA sequencing, human genetic variant association analysis, and proteomic and metabolomic profiling. Such integrative multi-omics efforts identify gamma-glutamyltransferase GGT7 as an intriguing potential cholesterol and lipid regulator. As a SREBP2-dependent target, GGT7 positively regulates cellular cholesterol levels and affects the expression of several cholesterol metabolism genes. Furthermore, GGT7 interacts with actin-dependent motor protein MYH10 to control low-density lipoprotein cholesterol (LDL-C) uptake into the cells. Genetic ablation of Ggt7 in mice leads to reduced serum cholesterol levels, supporting an in vivo role of Ggt7 during cholesterol homeostasis. CONCLUSIONS Our study not only provides a repertoire of lipid or cholesterol regulatory genes from multiple angles but also reveals a causal link between a gamma-glutamyltransferase and cholesterol metabolism.
Collapse
Affiliation(s)
- Haihuan Shan
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, China
- National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Northeastern University, Shenyang, 110819, China
- Key Laboratory of Data Analytics and Optimization for Smart Industry (Northeastern University), Ministry of Education, Shenyang, 110819, China
| | - Shuangshuang Fan
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, China
- National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Northeastern University, Shenyang, 110819, China
- Key Laboratory of Data Analytics and Optimization for Smart Industry (Northeastern University), Ministry of Education, Shenyang, 110819, China
| | - Quanrun Li
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, China
- National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Northeastern University, Shenyang, 110819, China
- Key Laboratory of Data Analytics and Optimization for Smart Industry (Northeastern University), Ministry of Education, Shenyang, 110819, China
| | - Ruipu Liang
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, China
- National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Northeastern University, Shenyang, 110819, China
- Key Laboratory of Data Analytics and Optimization for Smart Industry (Northeastern University), Ministry of Education, Shenyang, 110819, China
| | - Zhisong Chen
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, China
- National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Northeastern University, Shenyang, 110819, China
- Key Laboratory of Data Analytics and Optimization for Smart Industry (Northeastern University), Ministry of Education, Shenyang, 110819, China
| | - Shengnan Wang
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, China
- National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Northeastern University, Shenyang, 110819, China
- Key Laboratory of Data Analytics and Optimization for Smart Industry (Northeastern University), Ministry of Education, Shenyang, 110819, China
| | - Xiaofeng Wang
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, China
- National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Northeastern University, Shenyang, 110819, China
- Key Laboratory of Data Analytics and Optimization for Smart Industry (Northeastern University), Ministry of Education, Shenyang, 110819, China
| | - Yurong Li
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, China
- National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Northeastern University, Shenyang, 110819, China
- Key Laboratory of Data Analytics and Optimization for Smart Industry (Northeastern University), Ministry of Education, Shenyang, 110819, China
| | - Shuai Chen
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Shenyang, 110819, China
| | - Kun Yu
- College of Medicine and Bioinformation Engineering, Northeastern University, Shenyang, 110819, China
| | - Teng Fei
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, China.
- National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Northeastern University, Shenyang, 110819, China.
- Key Laboratory of Data Analytics and Optimization for Smart Industry (Northeastern University), Ministry of Education, Shenyang, 110819, China.
| |
Collapse
|
35
|
Zhao H, Wang X, Guo L, Li X, Teopiz KM, McIntyre RS, Wang W, Lu C. Investigating the impact of multidimensional sleep traits on cardiovascular diseases and the mediating role of depression. Open Heart 2025; 12:e002866. [PMID: 40086821 PMCID: PMC11907084 DOI: 10.1136/openhrt-2024-002866] [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: 08/01/2024] [Accepted: 01/31/2025] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND Observational studies have reported that sleep is associated with the risk of major depressive disorder (MDD) and cardiovascular diseases (CVDs). However, the causal relationships among various sleep traits remain contentious, and whether MDD mediates the impact of specific sleep traits on CVDs is unclear. METHODS We performed two-sample Mendelian randomisation analyses to explore whether insomnia, sleep time, daytime napping, daytime sleepiness, chronotype, snoring or obstructive sleep apnoea were causally associated with the risk of five CVDs, including coronary artery disease (CAD), myocardial infarction (MI), heart failure (HF), atrial fibrillation and stroke. Mediation analyses were performed to assess the proportion mediated by MDD. RESULTS Genetically predicted insomnia, short sleep, daytime napping and daytime sleepiness increased the risk of CVDs, with the OR ranging from 1.24 (95% CI 1.06 to 1.45) for insomnia on stroke to 1.55 (95% CI 1.28 to 1.89) for insomnia on MI. In contrast to short sleep, genetically predicted sleep duration decreased the risk of CAD (OR 0.88 (95% CI 0.80 to 0.97)), MI (OR 0.89 (95% CI 0.80 to 0.99)) and HF (OR 0.90 (95% CI 0.83 to 0.98)). However, we found no significant associations of long sleep, chronotype, snoring and obstructive sleep apnoea with increased risk for any CVD subtype. Additionally, the effect of insomnia was partially mediated by MDD for the risk of CAD (proportion mediated: 8.81% (95% CI 1.20% to 16.43%)), MI (9.17% (95% CI 1.71% to 16.63%)) and HF (14.46% (95% CI 3.48% to 25.45%)). Similarly, the effect of short sleep was partially mediated by MDD for the risk of CAD (8.92% (95% CI 0.87% to 16.97%)), MI (11.43% (95% CI 0.28% to 22.57%)) and HF (12.65% (95% CI 1.35% to 23.96%)). MDD also partially mediated the causal effects of insomnia on stroke, sleep duration on CAD, MI and HF, daytime napping on HF and daytime sleepiness on CAD. CONCLUSIONS Our study provides evidence that genetically predicted insomnia, short sleep, frequent daytime napping and sleepiness are associated with a higher risk of certain CVD subtypes, partly mediated by MDD.
Collapse
Affiliation(s)
- Hao Zhao
- Department of Medical Statistics and Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-Sen University, Guangzhou, China
| | - Xiaojie Wang
- Department of Neurology, Shenzhen Shekou People's Hospital, Shenzhen, China
| | - Lan Guo
- Department of Medical Statistics and Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Xiuwen Li
- Department of Medical Statistics and Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Kayla M Teopiz
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada
| | - Roger S McIntyre
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Wanxin Wang
- Department of Medical Statistics and Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Ciyong Lu
- Department of Medical Statistics and Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
36
|
Wang J, Wang P, Lv J, Chen R, Yan W, He D. Exploring the silent connection: unveiling the intricate relationship between gastroesophageal reflux disease and sleep apnea syndrome. Hum Genomics 2025; 19:23. [PMID: 40045407 PMCID: PMC11883946 DOI: 10.1186/s40246-025-00728-7] [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: 10/06/2023] [Accepted: 02/12/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND Gastroesophageal reflux disease (GERD) and Sleep Apnea Syndrome (SAS) are two prevalent medical conditions that significantly affect health and quality of life. GERD involves stomach content reflux into the esophagus, while SAS causes recurrent upper airway obstruction during sleep. Despite recent studies hinting at a link, the precise relationship and causality between GERD and SAS remain unclear. Our research uses bidirectional Mendelian randomization to explore this intricate relationship. Additionally, given SAS's high prevalence in cardiovascular patients (40-80%, as highlighted by the American Heart Association), we also investigated its potential association with various cardiovascular diseases to gain new insights into prevention and treatment. METHODS This study employed genetic data from large-scale genome-wide association studies (GWAS) on GERD (129,080 cases, 473,524 controls) and SAS (25,008 cases, 391,473 controls) for two-sample Mendelian randomization (MR) analysis to estimate the causal effects of GERD on the risk of SAS. All SNPs were selected using a strict clump window (r2 = 0.001 and kb = 10,000). We initially applied the inverse variance weighted (IVW) method and measured horizontal pleiotropy using MR-Egger, weighted median, and weighted mode methods. I2 index and Cochran Q statistics were used for sensitivity analysis. Funnel plot symmetry of IVW MR estimates versus 1/standard error (1/SEIV) was examined to exclude SNPs potentially causing heterogeneity. Additionally, to exclude reverse causality, bidirectional MR was employed to investigate whether genetic susceptibility to SAS causally influenced the risk of GERD. RESULTS GERD was associated with an elevated risk of SAS, demonstrating an odds ratio (OR) of 1.750 (95% CI 1.590-1.930; P < 0.001). Conversely, there was no compelling evidence to indicate a causal link between SAS and the risk of developing GERD, with an OR of 1.000 (95% CI 0.989-1.011; P = 0.964). In addition to the primary findings, our study also revealed significant risks associated with SAS for several cardiovascular conditions, including coronary heart disease, atrial fibrillation, coronary artery disease, heart failure, intracerebral hemorrhage, and ischemic stroke. CONCLUSION We discovered compelling evidence indicating an elevated risk of SAS in individuals with GERD, but no significant evidence supporting an increased risk of GERD in those with SAS. Future investigations into SAS risk should take into account the potential therapeutic targeting of GERD. PPI and histamine antagonists can effectively reduce reflux and airway secretions, preventing airway damage and collapse. Furthermore, it is necessary to investigate the underlying mechanisms by which GERD affects SAS. For example, the inflammatory stimulation caused by gastric acid and pepsin in refluxed fluid, as well as the increased tension of bronchial smooth muscle caused by vagus nerve reflex. Thus, early preventive measures can be implemented for potential complications related to SAS.
Collapse
Affiliation(s)
- Junming Wang
- Center of Emergency and Critical Care Medicine, Jinshan Hospital, Fudan University, Shanghai, 201508, People's Republic of China
- Research Center for Chemical Injury, Emergency and Critical Medicine of Fudan University, Shanghai, 201508, People's Republic of China
- Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health Commission, Shanghai, 201508, China
| | - Pengfei Wang
- Center of Emergency and Critical Care Medicine, Jinshan Hospital, Fudan University, Shanghai, 201508, People's Republic of China
- Research Center for Chemical Injury, Emergency and Critical Medicine of Fudan University, Shanghai, 201508, People's Republic of China
- Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health Commission, Shanghai, 201508, China
| | - Jiang Lv
- Department of General Practice, Jinshan Hospital, Fudan University, No. 1508 Longhang Road, Jinshan District, Shanghai, 201508, People's Republic of China
| | - Ran Chen
- Department of General Practice, Jinshan Hospital, Fudan University, No. 1508 Longhang Road, Jinshan District, Shanghai, 201508, People's Republic of China
| | - Wei Yan
- Department of General Practice, Jinshan Hospital, Fudan University, No. 1508 Longhang Road, Jinshan District, Shanghai, 201508, People's Republic of China
| | - Daikun He
- Department of General Practice, Jinshan Hospital, Fudan University, No. 1508 Longhang Road, Jinshan District, Shanghai, 201508, People's Republic of China.
- Department of General Practice, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China.
- Center of Emergency and Critical Care Medicine, Jinshan Hospital, Fudan University, Shanghai, 201508, People's Republic of China.
- Research Center for Chemical Injury, Emergency and Critical Medicine of Fudan University, Shanghai, 201508, People's Republic of China.
- Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health Commission, Shanghai, 201508, China.
| |
Collapse
|
37
|
Jung HU, Jung H, Baek EJ, Kang JO, Kwon SY, You J, Lim JE, Oh B. Assessment of polygenic risk score performance in East Asian populations for ten common diseases. Commun Biol 2025; 8:374. [PMID: 40045046 PMCID: PMC11882803 DOI: 10.1038/s42003-025-07767-9] [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: 07/22/2024] [Accepted: 02/18/2025] [Indexed: 03/09/2025] Open
Abstract
Polygenic risk score (PRS) uses genetic variants to assess disease susceptibility. While PRS performance is well-studied in Europeans, its accuracy in East Asians is less explored. This study evaluated PRSs for ten diseases in the Health Examinees (HEXA) cohort (n = 55,870) in Korea. Single-population PRSs were constructed using PRS-CS, LDpred2, and Lassosum based on East Asian GWAS summary statistics (sample sizes: 51,442-341,204), while cross-population PRSs were developed using PRS-CSx and CT-SLEB by integrating European and East Asian GWAS data. PRS-CS consistently outperformed other single-population methods across key metrics, including the likelihood ratio test (LRT), odds ratio per standard deviation (perSD OR), net reclassification improvement (NRI), and area under the curve (AUC). Cross-population PRSs further improved predictive performance, with average increases of 1.08-fold (LRT), 1.07-fold (perSD OR), and 1.15-fold (NRI) across seven diseases with statistical significance, and a 1.01-fold improvement in AUC. Differences in R² between single- and cross-population PRSs were statistically significant for five diseases, showing an average increase of 1.13%. Cross-population PRSs achieved 87.8% of the predictive performance observed in European PRSs. These findings highlight the benefits of integrating European GWAS data while underscoring the need for larger East Asian datasets to improve prediction accuracy.
Collapse
Affiliation(s)
- Hae-Un Jung
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Hyein Jung
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | | | - Ji-One Kang
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Shin Young Kwon
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | | | - Ji Eun Lim
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul, Republic of Korea.
| | - Bermseok Oh
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Republic of Korea.
- Mendel Inc, Seoul, Republic of Korea.
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul, Republic of Korea.
| |
Collapse
|
38
|
Yang Q, Li M, Chen P, Dou N, Liu M, Lu P, Yu C. Systematic Evaluation of the Impact of a Wide Range of Dietary Habits on Myocardial Infarction: A Two-Sample Mendelian Randomization Analysis. J Am Heart Assoc 2025; 14:e035936. [PMID: 40008582 DOI: 10.1161/jaha.124.035936] [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: 07/01/2024] [Accepted: 11/19/2024] [Indexed: 02/27/2025]
Abstract
BACKGROUND Myocardial infarction is a cardiovascular disease that significantly contributes to global morbidity and disability. Given the significant role of diet in the pathogenesis and prevention of cardiovascular diseases, this study rigorously investigates the causal relationship between dietary habits and myocardial infarction. METHODS AND RESULTS This study used large-scale genome-wide association studies with pooled UK Biobank data to explore associations between 9 dietary categories (83 types) and myocardial infarction. A 2-sample Mendelian randomization approach was applied to assess these associations, while multivariate Mendelian randomization and mediation analyses investigated the role of lipids in mediating the effects of diet on myocardial infarction. Univariate Mendelian analyses revealed genetic associations among 9 categories of dietary habits (83 types) and myocardial infarction. Notably, robust evidence indicates the "tablespoons of cooked vegetables per day" as the most significant risk factor for myocardial infarction development. "Coffee consumption(cups per day)" and "frequency of adding salt to food" were also identified as supplementary risk factors. In contrast, "overall alcohol intake" showed a protective effect, potentially by increasing high-density lipoprotein cholesterol (4.48% mediation) and reducing triglycerides (6.24% mediation). Cereal category, particularly "cereal consumption (bowls per week)" was associated with reduced myocardial infarction risk, contributing by raising high-density lipoprotein cholesterol (3.69% mediation) and lowering total cholesterol (8.33% mediation). Additionally, "overall cheese consumption" was also protective against myocardial infarction. CONCLUSIONS Our findings elucidate the influence of dietary habits on myocardial infarction, showing underlying genetic mechanisms and emphasizing the regulatory role of lipids as an intermediate.
Collapse
Affiliation(s)
- Qian Yang
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China. Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan Shandong China
- Shandong Institute of Endocrine and Metabolic Diseases Jinan Shandong China
- Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases Jinan Shandong China
| | - Man Li
- Department of Geratology Qilu Hospital of Shandong University Jinan Shandong China
| | - Pengcheng Chen
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China. Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan Shandong China
- Shandong Institute of Endocrine and Metabolic Diseases Jinan Shandong China
- Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases Jinan Shandong China
| | - Naixin Dou
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China. Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan Shandong China
- Shandong Institute of Endocrine and Metabolic Diseases Jinan Shandong China
- Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases Jinan Shandong China
| | - Mei Liu
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China. Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan Shandong China
- Shandong Institute of Endocrine and Metabolic Diseases Jinan Shandong China
- Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases Jinan Shandong China
| | - Peng Lu
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China. Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan Shandong China
- Shandong Institute of Endocrine and Metabolic Diseases Jinan Shandong China
- Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases Jinan Shandong China
| | - Chunxiao Yu
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China. Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan Shandong China
- Shandong Institute of Endocrine and Metabolic Diseases Jinan Shandong China
- Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases Jinan Shandong China
| |
Collapse
|
39
|
Loesch DP, Garg M, Matelska D, Vitsios D, Jiang X, Ritchie SC, Sun BB, Runz H, Whelan CD, Holman RR, Mentz RJ, Moura FA, Wiviott SD, Sabatine MS, Udler MS, Gause-Nilsson IA, Petrovski S, Oscarsson J, Nag A, Paul DS, Inouye M. Identification of plasma proteomic markers underlying polygenic risk of type 2 diabetes and related comorbidities. Nat Commun 2025; 16:2124. [PMID: 40032831 PMCID: PMC11876343 DOI: 10.1038/s41467-025-56695-z] [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: 04/04/2024] [Accepted: 01/22/2025] [Indexed: 03/05/2025] Open
Abstract
Genomics can provide insight into the etiology of type 2 diabetes and its comorbidities, but assigning functionality to non-coding variants remains challenging. Polygenic scores, which aggregate variant effects, can uncover mechanisms when paired with molecular data. Here, we test polygenic scores for type 2 diabetes and cardiometabolic comorbidities for associations with 2,922 circulating proteins in the UK Biobank. The genome-wide type 2 diabetes polygenic score associates with 617 proteins, of which 75% also associate with another cardiometabolic score. Partitioned type 2 diabetes scores, which capture distinct disease biology, associate with 342 proteins (20% unique). In this work, we identify key pathways (e.g., complement cascade), potential therapeutic targets (e.g., FAM3D in type 2 diabetes), and biomarkers of diabetic comorbidities (e.g., EFEMP1 and IGFBP2) through causal inference, pathway enrichment, and Cox regression of clinical trial outcomes. Our results are available via an interactive portal ( https://public.cgr.astrazeneca.com/t2d-pgs/v1/ ).
Collapse
Affiliation(s)
- Douglas P Loesch
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK.
| | - Manik Garg
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Dorota Matelska
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Dimitrios Vitsios
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Xiao Jiang
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Scott C Ritchie
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | | | - Heiko Runz
- Translational Sciences, Biogen Inc., Cambridge, MA, USA
| | | | - Rury R Holman
- Diabetes Trials Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Robert J Mentz
- Division of Cardiology, Duke University School of Medicine, Durham, NC, USA
| | - Filipe A Moura
- Thrombolysis in Myocardial Infarction (TIMI) Study Group, Division of Cardiovascular Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- VA Connecticut Healthcare System, West Haven, CT, USA
| | - Stephen D Wiviott
- Thrombolysis in Myocardial Infarction (TIMI) Study Group, Division of Cardiovascular Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Marc S Sabatine
- Thrombolysis in Myocardial Infarction (TIMI) Study Group, Division of Cardiovascular Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Miriam S Udler
- Diabetes Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Ingrid A Gause-Nilsson
- Late-Stage Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Slavé Petrovski
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Jan Oscarsson
- Late-Stage Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Abhishek Nag
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Dirk S Paul
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK.
- Precision Medicine and Biosamples, Oncology R&D, AstraZeneca, Cambridge, UK.
| | - Michael Inouye
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| |
Collapse
|
40
|
Tian D, Li J, Lai X, Yang Q, Zhang Z, Deng F. Single nucleotide polymorphisms: Implications in the early diagnosis and targeted intervention of coronary microvascular dysfunction. Genes Dis 2025; 12:101249. [PMID: 39759113 PMCID: PMC11696767 DOI: 10.1016/j.gendis.2024.101249] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 01/12/2024] [Accepted: 01/21/2024] [Indexed: 01/07/2025] Open
Abstract
Coronary microvascular dysfunction (CMD) is a clinical syndrome of myocardial ischemia caused by structural and/or functional abnormalities of pre-coronary arterioles and arterioles. While genetics and other factors play a role in CMD etiology, the key pathogenic mechanism remains unclear. Currently, the diagnostic procedure for CMD is still cumbersome, and there is a lack of effective targeted interventions. Single nucleotide polymorphisms (SNPs) offer promise in addressing these issues. SNPs, reflecting common genetic variations, have garnered extensive investigation across multiple diseases. Several SNPs associated with CMD have been discovered, and some have the potential to be therapeutic targets. Nevertheless, studies on CMD-related SNPs are relatively nascent and limited in number. In this review, we summarize the previously reported CMD-associated SNPs, delineate their pathophysiological mechanisms, and predict potentially important CMD sites by analyzing the SNPs linked to diseases sharing similar pathogenetic mechanisms and risk factors, such as coronary artery disease. We aim to explore reliable genetic markers implicated in CMD risk and prognosis, thereby providing a novel approach for early diagnosis and gene-targeted interventions of CMD in subsequent studies.
Collapse
Affiliation(s)
- Dingyuan Tian
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing 400038, China
- Department of Cardiovascular Medicine, Southwest Hospital, Army Medical University, Chongqing 400038, China
| | - Jie Li
- Department of Cardiovascular Medicine, Southwest Hospital, Army Medical University, Chongqing 400038, China
| | - Xiaoyue Lai
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Qingyuan Yang
- Department of Cardiovascular Medicine, Southwest Hospital, Army Medical University, Chongqing 400038, China
| | - Zhihui Zhang
- Department of Cardiovascular Medicine, Southwest Hospital, Army Medical University, Chongqing 400038, China
- Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing 400038, China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Chongqing 400038, China
| | - Fang Deng
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing 400038, China
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing 400038, China
- Key Laboratory of High Altitude Medicine, PLA, Chongqing 400038, China
| |
Collapse
|
41
|
Qiu Y, Song B, Yin Z, Wang M, Tao Y, Xie M, Duan A, Chen Z, Si K, Wang Z. Novel insights into causal effects of serum lipids, lipid metabolites, and lipid-modifying targets on the risk of intracerebral aneurysm. Eur Stroke J 2025; 10:236-247. [PMID: 39081035 PMCID: PMC11569451 DOI: 10.1177/23969873241265019] [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: 03/31/2024] [Accepted: 06/05/2024] [Indexed: 11/19/2024] Open
Abstract
INTRODUCTION Different serum lipid and lipid-lowering agents are reported to be related to the occurrence of intracerebral aneurysm (IA). However, the causal relationship between them requires further investigation. PATIENTS AND METHODS Mendelian randomization (MR) analysis was performed on IA and its subtypes by using instrumental variants associated with six serum lipids, 249 lipid metabolic traits, and 10 lipid-lowering agents that were extracted from the largest genome-wide association study. Phenome-wide MR analyses were conducted to identify potential phenotypes associated with significant lipid-lowering agents. RESULTS After multiple comparison adjustments (p < 0.0083), genetically proxied triglyceride (TG) (odds ratio [OR] 1.25, 95% confidence interval [CI] 1.07-1.47, p = 0.005) and high-density lipoprotein cholesterol (HDL-C) levels (OR 0.93, 95% CI 0.89-0.98, p = 0.008) showed causal relationships with the risk of IA. Four lipid metabolic traits showed a causal relationship with the risk of IA (p < 0.0002). As confirmed by drug target MR, the causal relationship between the HMGCR target and IA, HMGCR target and subarachnoid hemorrhage (SAH), ANGPTL3 target and SAH, CETP target, and SAH remained statistically significant after multiple adjustments (p < 0.005). Additionally, phenome-wide MR did not identify other diseases linked to the significant lipid-lowering agent (p < 6.39 × 10-5). DISCUSSION AND CONCLUSION This study not only supports that serum lipids (TG and HDL-C) are associated with IA but also confirms the positive effect and absence of safety concerns of intervening HMGCR, ANGPTL3, and CETP targets in IA and its subtypes, opening new avenues for IA treatment.
Collapse
Affiliation(s)
- Youjia Qiu
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Bingyi Song
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Ziqian Yin
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Menghan Wang
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Yuchen Tao
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Minjia Xie
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Aojie Duan
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Zhouqing Chen
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Ke Si
- Department of Cardiac Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Zhong Wang
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
42
|
Phair IR, Sovakova M, Alqurashi N, Nisr RB, McNeilly AD, Lamont D, Rena G. In-depth proteomic profiling identifies potentiation of the LPS response by 7-ketocholesterol. JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY PLUS 2025; 11:100285. [PMID: 39991505 PMCID: PMC11847031 DOI: 10.1016/j.jmccpl.2025.100285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/23/2025] [Accepted: 01/28/2025] [Indexed: 02/25/2025]
Abstract
In patients with stable coronary artery disease, plasma levels of 7-ketocholesterol (7-KC), found at high levels in atherosclerotic lesions, predict risk of incident heart failure dose dependently, potentially contributing to disease aetiology. Previous studies demonstrated that 7-KC can elicit effects on macrophage function; however, effects of 7-KC on the macrophage proteome have not been studied systematically. Here we used quantitative mass spectrometry to establish the effect of 7-KC on the mouse macrophage proteome. 7-KC independently mediated dynamic changes, including on atherogenic/M1 markers, cholesterol metabolism, biosynthesis and transport, as well as nutrient transport more broadly. These changes were however insufficient alone to drive changes in cytokine and chemokine secretion. Rather, they prime the macrophage, potentiating LPS-stimulated TNF alpha secretion and key pro-inflammatory enzymes. Our results indicate that 7-KC has independent metabolic effects on the macrophage; however, effects on the immune system are primarily due to the changes in metabolism priming the response to an inflammatory stimulus. Earlier findings from CANTOS and the recent FDA approval of colchicine highlight that inflammation is a viable target for cardiovascular disease; however, it is currrently unclear which will be the best anti-inflammatory targets to pursue in the future. In this context, our findings suggest that drugs targeting atherogenic markers induced by 7-KC might be well tolerated, as they will not necessarily be expected to be immunosuppressive.
Collapse
Affiliation(s)
- Iain R. Phair
- Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee DD1 9SY, Scotland, United Kingdom
| | - Magdalena Sovakova
- Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee DD1 9SY, Scotland, United Kingdom
| | - Noor Alqurashi
- Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee DD1 9SY, Scotland, United Kingdom
| | - Raid B. Nisr
- Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee DD1 9SY, Scotland, United Kingdom
| | - Alison D. McNeilly
- Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee DD1 9SY, Scotland, United Kingdom
| | - Douglas Lamont
- Centre for Advanced Scientific Technologies, School of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland, United Kingdom
| | - Graham Rena
- Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee DD1 9SY, Scotland, United Kingdom
| |
Collapse
|
43
|
Shi Y, Xiang Y, Ye Y, He T, Sham PC, So HC. A framework for detecting causal effects of risk factors at an individual level based on principles of Mendelian randomisation: applications to modelling individualised effects of lipids on coronary artery disease. EBioMedicine 2025; 113:105616. [PMID: 40020258 PMCID: PMC11919333 DOI: 10.1016/j.ebiom.2025.105616] [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: 05/11/2024] [Revised: 01/30/2025] [Accepted: 02/10/2025] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND Mendelian Randomisation (MR) has been widely used to study the causal effects of risk factors. However, almost all MR studies concentrate on the population's average causal effects. With the advent of precision medicine, the individualised treatment effect (ITE) is often of greater interest. For instance, certain risk factors may pose a higher risk to some individuals than others, and the benefits of treatments may vary across individuals. This study proposes a framework for estimating individualised causal effects in large-scale observational studies where unobserved confounding factors may be present. METHODS We propose a framework (MR-ITE) that expands the scope of MR from estimating average causal effects to individualised causal effects. We present several approaches for estimating ITEs within this MR framework, primarily grounded on the principles of the "R-learner". To evaluate the presence of causal effect heterogeneity, we also proposed two permutation testing methods. We employed polygenic risk score (PRS) as instruments and proposed methods to improve the accuracy of ITE estimates by removal of potentially pleiotropic single nucleotide polymorphisms (SNPs). The validity of our approach was substantiated through comprehensive simulations. The proposed framework also allows the identification of important effect modifiers contributing to individualised differences in treatment effects. We applied our framework to study the individualised causal effects of various lipid traits, including low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C), triglycerides (TG), and total cholesterol (TC), on the risk of coronary artery disease (CAD) based on the UK-Biobank (UKBB). We also studied the ITE of C-reactive protein (CRP) and insulin-like growth factor 1 (IGF-1) on CAD as secondary analyses. FINDINGS Simulation studies demonstrated that MR-ITE outperformed traditional causal forest approaches in identifying ITEs when unobserved confounders were present. The integration of the contamination mixture (ConMix) approach to remove invalid pleiotropic SNPs further enhanced MR-ITE's performance. In real-world applications, we identified positive causal associations between CAD and several factors (LDL-C, Total Cholesterol, and IGF-1 levels). Our permutation tests revealed significant heterogeneity in these causal associations across individuals. Using Shapley value analysis, we identified the top effect modifiers contributing to this heterogeneity. INTERPRETATION We introduced a new framework, MR-ITE, capable of inferring individualised causal effects in observational studies based on the MR approach, utilizing PRS as instruments. MR-ITE extends the application of MR from estimating the average treatment effect to individualised treatment effects. Our real-world application of MR-ITE underscores the importance of identifying ITEs in the context of precision medicine. FUNDING This work was supported partially by a National Natural Science Foundation of China grant (NSFC; grant number 81971706), the KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research of Common Diseases, Kunming Institute of Zoology and The Chinese University of Hong Kong, China, and the Lo Kwee Seong Biomedical Research Fund from The Chinese University of Hong Kong.
Collapse
Affiliation(s)
- Yujia Shi
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Yong Xiang
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Yuxin Ye
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Tingwei He
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Pak-Chung Sham
- Department of Psychiatry, University of Hong Kong, Hong Kong SAR, China
| | - Hon-Cheong So
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China; KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research of Common Diseases, Kunming Institute of Zoology and the Chinese University of Hong Kong, China; Department of Psychiatry, The Chinese University of Hong Kong, Hong Kong, China; CUHK Shenzhen Research Institute, Shenzhen, China; Margaret K.L. Cheung Research Centre for Management of Parkinsonism, The Chinese University of Hong Kong, Shatin, Hong Kong, China; Brain and Mind Institute, The Chinese University of Hong Kong, Hong Kong SAR, China; Hong Kong Branch of the Chinese Academy of Sciences Center for Excellence in Animal Evolution and Genetics, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
44
|
Lin S, Lin S, Ge T, Chen C, Lin Y. Causal Mediation Analysis: A Summary-Data Mendelian Randomization Approach. Stat Med 2025; 44:e10317. [PMID: 39910926 PMCID: PMC11799828 DOI: 10.1002/sim.10317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 10/30/2024] [Accepted: 12/13/2024] [Indexed: 02/07/2025]
Abstract
Summary-data Mendelian randomization (MR), a widely used approach in causal inference, has recently attracted attention for improving causal mediation analysis. Two existing methods corresponding to the difference method and product method of linear mediation analysis have been developed to perform MR-based mediation analysis using the inverse-variance weighted method (MR-IVW). Despite these developments, there is still a need for more rigorous, efficient, and precise MR-based mediation methodologies. In this study, we develop summary-data MR-based frameworks for causal mediation analysis. We improve the accuracy, statistical efficiency and robustness of the existing MR-based mediation analysis by implementing novel variance estimators for the mediation effects, deriving rigorous procedures for statistical inference, and accounting for widespread pleiotropic effects. Specifically, we propose Diff-IVW and Prod-IVW to improve upon the existing methods and provide the pleiotropy-robust methods (Diff-Egger, Diff-Median, Prod-Egger, and Prod-Median), adapted from MR-Egger and MR-Median, to enhance the robustness of the MR-based mediation analysis. We conduct comprehensive simulation studies to compare the existing and proposed methods. The results show that the proposed methods, Diff-IVW and Prod-IVW, improve statistical efficiency and type I error control over the existing approaches. Although all IVW-based methods suffer from directional pleiotropy biases, the median-based methods (Diff-Median and Prod-Median) can mitigate such biases. The differences among the methods can lead to discrepant statistical conclusions as demonstrated in real data applications. Based on our simulation results, we recommend the three proposed methods in practice: Diff-IVW, Prod-IVW, and Prod-Median, which are complementary under various scenarios.
Collapse
Affiliation(s)
- Shu‐Chin Lin
- Center for Neuropsychiatric ResearchNational Health Research InstitutesMiaoliTaiwan
- Institute of Statistics and Data ScienceNational Taiwan UniversityTaipeiTaiwan
| | - Sheng‐Hsuan Lin
- Institute of StatisticsNational Yang Ming Chiao Tung UniversityHsinchuTaiwan
| | - Tian Ge
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic MedicineMassachusetts General HospitalBostonMassachusettsUSA
- Stanley Center for Psychiatric ResearchBroad Institute of MIT and HarvardCambridgeMassachusettsUSA
- Department of PsychiatryMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Chia‐Yen Chen
- Translational MedicineBiogenCambridgeMassachusettsUSA
| | - Yen‐Feng Lin
- Center for Neuropsychiatric ResearchNational Health Research InstitutesMiaoliTaiwan
- Department of Public Health & Medical Humanities, School of MedicineNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
- Institute of Behavioral Medicine, College of MedicineNational Cheng Kung UniversityTainanTaiwan
| |
Collapse
|
45
|
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
|
46
|
Bian K, Zhang P, Xu G, Sun W. The association between fatigue and cardiometabolic diseases: Insights from the UK biobank study. J Affect Disord 2025; 371:261-267. [PMID: 39577501 DOI: 10.1016/j.jad.2024.11.040] [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: 08/28/2024] [Revised: 10/09/2024] [Accepted: 11/11/2024] [Indexed: 11/24/2024]
Abstract
BACKGROUND Cardiometabolic diseases (CMD) are major global health concerns with significant morbidity and mortality. Fatigue, a common but often overlooked symptom, has been postulated as both a potential risk factor for and a consequence of these conditions. However, the relationships between fatigue and CMD remain unclear. This study aimed to investigate the relationship between fatigue and CMD using observational and genetic approaches. METHOD Observational study was conducted in the UK biobank. Genetic method was employed a bidirectional MR approach to examine the causal relationship between fatigue and CMD. Genetic variants associated with fatigue were identified through a GWAS, and summary statistics from the largest available GWAS were used to obtain variants associated with stroke, CAD, T2D, and HF. Inverse variance weighting (IVW) was conducted, with weighted median, MR-Egger, and MR-PRESSO as sensitivity analyses. Multivariable MR and mediation analysis were also employed. RESULTS Observational analyses indicated that individuals with fatigue had a significantly increased risk of developing stroke (HR 1.44, 95 % CI 1.27-1.63), T2D (HR 1.46, 95 % CI 1.41-1.51), CAD (HR 1.45, 95 % CI 1.4-1.5), and HF (HR 1.60, 95 % CI 1.52-1.68). Mendelian randomization analyses further supported a causal relationship. Additionally, observational and genetic analyses showed T2D was found to be associated with increased levels of fatigue. Mediation analysis identified lipid metabolites as mediators in the relationship between fatigue and CMD. CONCLUSION This study highlights a bidirectional relationship between fatigue and CMD, underscoring the importance of considering fatigue in the context of cardiometabolic health. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Keyu Bian
- Department of Neurology, Wujin TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Changzhou, Jiangsu, China; Department of Neurology, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China; Department of Neurology, Shenzhen Second People's Hospital, Shenzhen, Guangdong, 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, China
| | - Gelin Xu
- Department of Neurology, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China; Department of Neurology, Shenzhen Second People's Hospital, Shenzhen, Guangdong, China; Department of Neurology, First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, 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, China.
| |
Collapse
|
47
|
Gong Z, Wu D, Ku Y, Zou C, Qiu L, Hao X, Liu L. Lipid-lowering drug targets associated with risk of respiratory disease: a Mendelian randomization study. BMC Pulm Med 2025; 25:71. [PMID: 39934773 PMCID: PMC11817876 DOI: 10.1186/s12890-025-03527-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 01/28/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND Observational studies have identified a possible connection between lipid-lowering medications and respiratory illnesses. However, it remains unclear whether lipid-lowering drugs is causative for respiratory diseases, and we aimed to answer this question. METHODS We performed Mendelian randomization (MR) analyses by integrating data from genome-wide association studies (GWAS). Three statistical approaches were employed for MR analysis: inverse variance weighting (IVW), MR-Egger, and weighted median. The purpose was to evaluate the causal relationships between 10 drug targets that lower lipid levels and the likelihood of developing 7 respiratory diseases. Additional sensitivity analyses were conducted to ensure the robustness and validity of the results. RESULTS After adjusting for multiple testing, our MR analysis identified APOB (odd ratios [OR]: 0.86; 95% confidence interval [CI]: 0.77 to 0.97; PIVW = 0.01) and PCSK9 (OR: 0.84; 95% CI: 0.72 to 0.97; PIVW = 0.02) as significant risk targets for asthma. Additionally, LDLR was found to be a significant risk target for chronic obstructive pulmonary disease (OR: 0.81; 95% CI: 0.67 to 0.98; PIVW = 0.03). The sensitivity analysis validated no proof of heterogeneity or pleiotropy amongst the mentioned results. CONCLUSIONS Our findings suggest a likely causal relationship between respiratory diseases and lipid-lowering drug targets. Further mechanistic and clinical research is needed to confirm and validate these findings.
Collapse
Affiliation(s)
- Zhipeng Gong
- Department of Thoracic Surgery and Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Dongsheng Wu
- Department of Thoracic Surgery and Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Yin Ku
- Department of Thoracic Surgery and Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Congyao Zou
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Lin Qiu
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Xiaohu Hao
- Department of Thoracic Surgery and Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Lunxu Liu
- Department of Thoracic Surgery and Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
48
|
Salido E, de Medeiros Vieira C, Mosquera JV, Zade R, Parikh P, Suryavanshi S, Miller CL, Lo Sardo V. The 9p21.3 coronary artery disease risk locus drives vascular smooth muscle cells to an osteochondrogenic state. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.05.25.595888. [PMID: 38853913 PMCID: PMC11160673 DOI: 10.1101/2024.05.25.595888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Background Genome-wide association studies have identified common genetic variants at ~300 human genomic loci linked to coronary artery disease (CAD) susceptibility. Among these genomic regions, the most impactful is the 9p21.3 CAD risk locus, which spans a 60 kb gene desert and encompasses ~80 SNPs in high linkage disequilibrium. Despite nearly two decades since its discovery, the role of the 9p21.3 locus in cells of the vasculature remains incompletely resolved. Methods We differentiated induced pluripotent stem cells (iPSCs) from risk and non-risk donors at 9p21.3 into vascular smooth muscle cells. We performed single-cell transcriptomic profiling, including co-embedding and comparison with publicly available human arterial datasets. We conducted functional characterization using migration and calcification assays and confirmed our findings on iPSC-VSMCs derived from additional donors. Finally, we used overexpression of ANRIL followed by gene expression analysis. Results We demonstrated that iPSC-VSMCs harboring the 9p21.3 risk haplotype preferentially adopt an osteochondrogenic state and show remarkable similarity to fibrochondrocytes from human artery tissue. The transcriptional profile and functional assessment of migration and calcification capacity across iPSC-VSMCs lines from multiple donors concordantly resemble an osteochondrogenic state. Importantly, we identified numerous transcription factors driving different VSMC state trajectories. Additionally, we prioritized LIMCH1 and CRABP1 as signature genes critical for defining the risk transcriptional program. Finally, overexpression of a short isoform of ANRIL in non-risk cells was sufficient to induce the osteochondrogenic transcriptional signature. Conclusions Our study provides new insights into the mechanism of the 9p21.3 risk locus and defines its previously undescribed role in driving a disease-prone transcriptional and functional state in VSMCs concordant with an osteochondrogenic-like state. Our data suggest that the 9p21.3 risk haplotype likely promotes arterial calcification, through altered expression of ANRIL, in a cell-type specific and cell-autonomous manner, providing insight into potential risk assessment and treatment for carriers.
Collapse
Affiliation(s)
- Elsa Salido
- Department of Cell and Regenerative Biology; University of Wisconsin-Madison; Madison, WI 53705 USA
| | | | - José Verdezoto Mosquera
- Department of Genome Sciences; Department of Biochemistry and Molecular Genetics; University of Virginia; Charlottesville, VA 22908 USA
| | - Rohan Zade
- Department of Cell and Regenerative Biology; University of Wisconsin-Madison; Madison, WI 53705 USA
| | - Parth Parikh
- Department of Cell and Regenerative Biology; University of Wisconsin-Madison; Madison, WI 53705 USA
| | - Shraddha Suryavanshi
- Department of Cell and Regenerative Biology; University of Wisconsin-Madison; Madison, WI 53705 USA
| | - Clint L. Miller
- Department of Genome Sciences; Department of Biochemistry and Molecular Genetics; University of Virginia; Charlottesville, VA 22908 USA
| | - Valentina Lo Sardo
- Department of Cell and Regenerative Biology; University of Wisconsin-Madison; Madison, WI 53705 USA
| |
Collapse
|
49
|
Xiao J, Zhang N, Gao Z, Wei Y, Wei H, Qiu Z, Sundquist K, Sundquist J, Ji J, Huang W. Phosphodiesterase 5 and its inhibitors with ischaemic heart disease: a Mendelian randomization analysis and a real-world study. EUROPEAN HEART JOURNAL. CARDIOVASCULAR PHARMACOTHERAPY 2025; 11:75-83. [PMID: 39424597 PMCID: PMC11805687 DOI: 10.1093/ehjcvp/pvae081] [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: 08/14/2024] [Revised: 09/23/2024] [Accepted: 10/17/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND Accumulating studies reported that several phosphodiesterases (PDEs) inhibitors might have cardiovascular benefits. OBJECTIVES This study aimed to explore the relationship between genetically-predicted PDEs and ischaemia heart disease via drug target Mendelian randomization (MR) approach, and then examine the effect of inhibitors of identified target on the outcomes by using real-world data. METHODS AND RESULTS In the two-sample MR study, the expression of genes encoding PDEs was used to proxy the level of PDEs and available expression quantitative trait loci (eQTLs) for each target gene were identified as the genetic instruments. The outcomes included coronary heart disease (CHD) and myocardial infarction (MI). In the real-world study, a retrospective cohort was conducted to compare the incidence of outcomes between PDE5 inhibitors and alprostadil use by linking Swedish nationwide registers. MR analyses identified two types of PDEs, PDE5, and PDE8, genetically-predicted expression in blood of the encoded genes was significantly associated with the risk of CHD [odds ratio (OR)PDE5A = 1.22,95% confidence interval (CI) = 1.06-1.40; ORPDE8A = 1.26,95% CI = 1.07-1.49] and MI (ORPDE5A = 1.27,95% CI = 1.09-1.48; ORPDE8A = 1.24,95% CI = 1.04-1.48). Notably, the highest expression of PDE5A was observed in artery aorta, which was also positively related to CHD (OR = 1.17,95% CI = 1.05-1.32) and MI (OR = 1.15,95% CI = 1.02-1.30). Real-world study provided supportive evidence that as compared to alprostadil use, PDE5 inhibitors use significantly reduced the incidence of CHD (adjusted HR = 0.70,95% CI = 0.66-0.73) and MI (adjusted HR = 0.79,95% CI = 0.73-0.84). CONCLUSION This study provided observational and genetic evidence about the protective role of PDE5 inhibition against ischaemic heart disease, indicating the potential of these drugs to be repurposed for ischemia heart disease prevention and treatment.
Collapse
Affiliation(s)
- Jun Xiao
- Department of Cardiovascular Surgery, Fujian Medical University, Union Hospital, Fuzhou, Fujian, China
- Heart Center of Fujian Medical University, Fuzhou, Fujian, China
| | - Naiqi Zhang
- Center for Primary Health Care Research, Department of Clinical Sciences Malmö, Lund University, Sweden
- Nutritional Epidemiology, Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Ziting Gao
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Yajing Wei
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Hongye Wei
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Ziyi Qiu
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Kristina Sundquist
- Center for Primary Health Care Research, Department of Clinical Sciences Malmö, Lund University, Sweden
| | - Jan Sundquist
- Center for Primary Health Care Research, Department of Clinical Sciences Malmö, Lund University, Sweden
| | - Jianguang Ji
- Center for Primary Health Care Research, Department of Clinical Sciences Malmö, Lund University, Sweden
- Department of Public Health and Medicinal Administration, Faculty of Health Sciences, University of Macau, Macao, Macao SAR, China
| | - Wuqing Huang
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| |
Collapse
|
50
|
Nakada S, Ward J, Strawbridge RJ, Welsh P, Celis-Morales C, Ho FK, Pell JP. Anxiety disorder, depression and coronary artery disease: associations and modification by genetic susceptibility. BMC Med 2025; 23:73. [PMID: 39915848 PMCID: PMC11804096 DOI: 10.1186/s12916-025-03915-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 01/27/2025] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND Associations of anxiety disorder and depression with coronary artery disease (CAD) are heterogeneous between populations. This study investigated how genetic susceptibility to CAD alters these associations with incident CAD, comparing and combining anxiety disorder and depression. METHODS This is a prospective cohort study using UK Biobank. Diagnoses of anxiety disorder and depression were ascertained through linked hospital admission data. Incident CAD was ascertained through hospital admission and death certificate data after baseline. CAD polygenic risk score (PRSCAD) was obtained from CARDIoGRAMplus4 and categorised into low, intermediate, and high. Cox proportional hazard models were used to examine associations between anxiety disorder and depression and CAD. RESULTS Both anxiety disorder (HR 2.31, 95% CI 1.92-2.78) and depression (HR 2.15, 95% CI 1.90-2.24) were associated with CAD after adjusting for sociodemographic confounders. There was an addictive interaction between depression and PRSCAD (RERI 0.97, 95% CI 0.12-1.81) such that the association between depression and CAD was strongest among those with a high PRSCAD whilst there was no such evidence for anxiety disorder. Anxiety disorder only (HR 1.68, 95% 1.16-2.44), depression only (HR 2.13, 95% CI 1.72-2.64), and concomitant anxiety disorder and depression (HR 3.85, 95% CI 2.48-5.98) were associated with CAD even among people with a low PRSCAD. Adjusting for potential mediators attenuated all these associations across PRS categories. CONCLUSIONS CAD genetic susceptibility might partly contribute to the clustering of depression and CAD but does not provide a full explanation, nor does it explain the association between anxiety disorder and CAD. Therefore, other mechanisms should be explored.
Collapse
Affiliation(s)
- Shinya Nakada
- School of Health and Wellbeing, University of Glasgow, 90 Byres Road, Glasgow, G12 8TB, UK
| | - Joey Ward
- School of Health and Wellbeing, University of Glasgow, 90 Byres Road, Glasgow, G12 8TB, UK
| | - Rona J Strawbridge
- School of Health and Wellbeing, University of Glasgow, 90 Byres Road, Glasgow, G12 8TB, UK
- Division of Cardiovascular Medicine, Department of Medicine Solna, Karolinska Institute, Solna, Sweden
- HDR-UK, London, UK
| | - Paul Welsh
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, G12 8TA, UK
| | - Carlos Celis-Morales
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, G12 8TA, UK
- Human Performance Laboratory, Physical Activity and Health Research Unit, Universidad Católica del Maule, EducationTalca, Chile
- Centro de Investigación en Medicina de Altura (CEIMA), Universidad Arturo Prat, Iquique, Chile
| | - Frederick K Ho
- School of Health and Wellbeing, University of Glasgow, 90 Byres Road, Glasgow, G12 8TB, UK
| | - Jill P Pell
- School of Health and Wellbeing, University of Glasgow, 90 Byres Road, Glasgow, G12 8TB, UK.
| |
Collapse
|