1
|
Kojima N, Koido M, He Y, Shimmori Y, Hachiya T, BioBank Japan, Debette S, Kamatani Y. Recurrent Stroke Prediction by Applying a Stroke Polygenic Risk Score in the Japanese Population. Stroke 2025; 56:1483-1491. [PMID: 40135360 DOI: 10.1161/strokeaha.124.047786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 01/19/2025] [Accepted: 02/12/2025] [Indexed: 03/27/2025]
Abstract
BACKGROUND Recently, various polygenic risk score (PRS)-based methods were developed to improve stroke prediction. However, current PRSs (including cross-ancestry PRS) poorly predict recurrent stroke. Here, we aimed to determine whether the best PRS for Japanese individuals can also predict stroke recurrence in this population by extensively comparing the methods and maximizing the predictive performance for stroke onset. METHODS We used data from the disease-oriented BBJ1 (BioBank Japan first cohort; recruited between 2003 and 2007, n=179 938) to derive and optimize the PRSs using a 10-fold cross-validation. We integrated the optimized PRSs for multiple traits, such as vascular risk factors and stroke subtypes to generate a single PRS using the meta-scoring approach (metaGRS). We used an independent BBJ2 (BBJ second cohort; recruited between 2012 and 2017, n=41 929) as a test sample to evaluate the association of the metaGRS with stroke and recurrent stroke. In addition, we analyzed its association stratified by risk factors. We administered 3 distinct tests to consider the potential index event bias. RESULTS We analyzed recurrent stroke cases (n=174) and nonrecurrent stroke controls (n=1153) among subjects within the BBJ2. After adjusting for known risk factors, metaGRS was associated with stroke recurrence (adjusted odds ratio per SD, 1.18 [95% CI, 1.00-1.39]; P=0.044), although no significant correlation was observed with the published PRSs. The outcomes derived from these examinations did not provide any significant indication of the influence of index event bias. The high metaGRS group without a history of hypertension had a higher risk of stroke recurrence than that of the low metaGRS group (adjusted odds ratio, 2.24 [95% CI, 1.07-4.66]; P=0.032). There was no association at all in the hypertension group (adjusted odds ratio, 1.21 [95% CI, 0.69-2.13]; P=0.50). CONCLUSIONS The metaGRS developed in a Japanese cohort predicted stroke recurrence in an independent cohort of patients. In particular, it predicted an increased risk of recurrence among stroke patients without hypertension. These findings provide clues for additional genetic risk stratification and help in developing personalized strategies for stroke recurrence prevention.
Collapse
Affiliation(s)
- Naoki Kojima
- Laboratory of Complex Trait Genomics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Japan (N.K., M.K., Y.H., Y.S., T.H., Y.K.)
| | - Masaru Koido
- Laboratory of Complex Trait Genomics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Japan (N.K., M.K., Y.H., Y.S., T.H., Y.K.)
| | - Yunye He
- Laboratory of Complex Trait Genomics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Japan (N.K., M.K., Y.H., Y.S., T.H., Y.K.)
| | - Yuka Shimmori
- Laboratory of Complex Trait Genomics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Japan (N.K., M.K., Y.H., Y.S., T.H., Y.K.)
| | - Tsuyoshi Hachiya
- Laboratory of Complex Trait Genomics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Japan (N.K., M.K., Y.H., Y.S., T.H., Y.K.)
| | | | - Stéphanie Debette
- Bordeaux Population Health Research Center, University of Bordeaux, Inserm, France (S.D.)
- Department of Neurology, Institute for Neurodegenerative Diseases, CHU de Bordeaux, France (S.D.)
| | - Yoichiro Kamatani
- Laboratory of Complex Trait Genomics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Japan (N.K., M.K., Y.H., Y.S., T.H., Y.K.)
| |
Collapse
|
2
|
Daghlas I, Karhunen V, Kim AS, Gill D. Application of Human Genetics to Prioritize Coagulation Cascade Protein Targets for Ischemic Stroke Prevention. Stroke 2025; 56:1542-1553. [PMID: 40188416 PMCID: PMC7617607 DOI: 10.1161/strokeaha.124.049808] [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: 10/25/2024] [Revised: 02/04/2025] [Accepted: 03/12/2025] [Indexed: 04/08/2025]
Abstract
BACKGROUND While interindividual variations in concentration and function of coagulation cascade proteins are established risk factors for venous thromboembolism (VTE), their associations with arterial ischemic stroke are less well defined. METHODS We identified and validated genetic proxies for lifelong, randomized perturbations of coagulation cascade proteins in genome-wide association studies of circulating protein levels (deCODE, n=35 559; UK Biobank, n=46 218) and of VTE risk (81 190 cases and 1 419 671 controls). Study participants were all of European ancestry. We performed 2-sample Mendelian randomization and colocalization analyses to test associations of these genetic proxies with risk of ischemic stroke (62 100 cases and 1 234 808 controls from the GIGASTROKE consortium) and ischemic stroke subtypes, and further contextualized associations with VTE and secondary efficacy and safety outcomes. RESULTS We identified genetic proxies for 30 coagulation factors, with cross-trait associations recapitulating canonical coagulation biology. Mendelian randomization and colocalization analyses supported causal associations of genetically proxied levels of 5 proteins with risk of ischemic stroke, with all proteins associating with the cardioembolic stroke subtype: factor XI (odds ratio [OR] of cardioembolic stroke per 1-SD increase, 1.31 [95% CI, 1.19-1.44]; P=3.30×10-8), high-molecular-weight kininogen (OR, 1.19 [95% CI, 1.09-1.30]; P=7.79×10-5), prothrombin (OR, 1.83 [95% CI, 1.31-2.57]; P=4.20×10-4), soluble PROCR (protein C receptor; OR, 0.88 [95% CI, 0.82-0.95]; P=6.19×10-4), and γ' fibrinogen (OR per doubling in VTE risk due to lower γ' fibrinogen levels, 1.44 [95% CI, 1.25-1.66]; P=3.96×10-7). γ' Fibrinogen and prothrombin also associated with large artery atherosclerotic stroke, and no proteins were associated with small vessel stroke risk. By contrast, genetic proxies for several coagulation factors (including proteins C and S and factors V and VII) showed selective associations with VTE. CONCLUSIONS These data highlight specific coagulation cascade components implicated in ischemic stroke pathogenesis, while identifying proteins with distinct roles in VTE. These findings may inform development of novel anticoagulants and optimize their use in targeted populations with stroke.
Collapse
Affiliation(s)
- Iyas Daghlas
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California San Francisco (I.D., A.S.K.)
| | - Ville Karhunen
- MRC Biostatistics Unit, School of Clinical Medicine, University of Cambridge, United Kingdom (V.K.)
| | - Anthony S. Kim
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California San Francisco (I.D., A.S.K.)
| | - Dipender Gill
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, United Kingdom (D.G.)
| |
Collapse
|
3
|
Sanghvi MM, Young WJ, Naderi H, Burns R, Ramírez J, Bell CG, Munroe PB. Using Genomics to Develop Personalized Cardiovascular Treatments. Arterioscler Thromb Vasc Biol 2025; 45:866-881. [PMID: 40244646 DOI: 10.1161/atvbaha.125.319221] [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: 02/04/2025] [Accepted: 04/07/2025] [Indexed: 04/18/2025]
Abstract
Advances in genomic technologies have significantly enhanced our understanding of both monogenic and polygenic etiologies of cardiovascular disease. In this review, we explore how the utilization of genomic information is bringing personalized medicine approaches to the forefront of cardiovascular disease management. We describe how genomic data can resolve diagnostic uncertainty, support cascade screening, and inform treatment strategies. We discuss how genome-wide association studies have identified thousands of genetic variants associated with polygenic cardiovascular diseases, and how integrating these insights into polygenic risk scores can enhance personalized risk prediction beyond traditional clinical algorithms. We detail how pharmacogenomics approaches leverage genotype information to guide drug selection and mitigate adverse events. Finally, we present the paradigm-shifting approach of gene therapy, which holds the promise of being a curative intervention for cardiovascular conditions.
Collapse
Affiliation(s)
- Mihir M Sanghvi
- William Harvey Research Institute (M.M.S., W.J.Y., H.N., R.B., J.R., C.G.B., P.B.M.), Queen Mary University of London, United Kingdom
- NIHR Barts Biomedical Research Centre (M.M.S., W.J.Y., H.N., R.B., C.G.B., P.B.M.), Queen Mary University of London, United Kingdom
- Barts Heart Centre, Barts Health NHS Trust, London, United Kingdom (M.M.S., W.J.Y., H.N.)
| | - William J Young
- William Harvey Research Institute (M.M.S., W.J.Y., H.N., R.B., J.R., C.G.B., P.B.M.), Queen Mary University of London, United Kingdom
- NIHR Barts Biomedical Research Centre (M.M.S., W.J.Y., H.N., R.B., C.G.B., P.B.M.), Queen Mary University of London, United Kingdom
- Barts Heart Centre, Barts Health NHS Trust, London, United Kingdom (M.M.S., W.J.Y., H.N.)
| | - Hafiz Naderi
- William Harvey Research Institute (M.M.S., W.J.Y., H.N., R.B., J.R., C.G.B., P.B.M.), Queen Mary University of London, United Kingdom
- NIHR Barts Biomedical Research Centre (M.M.S., W.J.Y., H.N., R.B., C.G.B., P.B.M.), Queen Mary University of London, United Kingdom
- Barts Heart Centre, Barts Health NHS Trust, London, United Kingdom (M.M.S., W.J.Y., H.N.)
| | - Richard Burns
- William Harvey Research Institute (M.M.S., W.J.Y., H.N., R.B., J.R., C.G.B., P.B.M.), Queen Mary University of London, United Kingdom
- NIHR Barts Biomedical Research Centre (M.M.S., W.J.Y., H.N., R.B., C.G.B., P.B.M.), Queen Mary University of London, United Kingdom
| | - Julia Ramírez
- William Harvey Research Institute (M.M.S., W.J.Y., H.N., R.B., J.R., C.G.B., P.B.M.), Queen Mary University of London, United Kingdom
- Aragon Institute of Engineering Research, University of Zaragoza, Spain (J.R.)
- Centro de Investigación Biomédica en Red, Biomedicina, Bioingeniería y Nanomedicina, Zaragoza, Spain (J.R.)
| | - Christopher G Bell
- William Harvey Research Institute (M.M.S., W.J.Y., H.N., R.B., J.R., C.G.B., P.B.M.), Queen Mary University of London, United Kingdom
- NIHR Barts Biomedical Research Centre (M.M.S., W.J.Y., H.N., R.B., C.G.B., P.B.M.), Queen Mary University of London, United Kingdom
| | - Patricia B Munroe
- William Harvey Research Institute (M.M.S., W.J.Y., H.N., R.B., J.R., C.G.B., P.B.M.), Queen Mary University of London, United Kingdom
- NIHR Barts Biomedical Research Centre (M.M.S., W.J.Y., H.N., R.B., C.G.B., P.B.M.), Queen Mary University of London, United Kingdom
| |
Collapse
|
4
|
Lee H, Fernandes M, Lee J, Merino J, Kwak SH. Exploring the shared genetic landscape of diabetes and cardiovascular disease: findings and future implications. Diabetologia 2025; 68:1087-1100. [PMID: 40088285 PMCID: PMC12069157 DOI: 10.1007/s00125-025-06403-9] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 01/28/2025] [Indexed: 03/17/2025]
Abstract
Diabetes is a rapidly growing global health concern projected to affect one in eight adults by 2045, which translates to roughly 783 million people. The profound metabolic alterations often present in dysglycaemia significantly increase the risk of cardiovascular complications. While genetic susceptibility plays a crucial role in diabetes and its vascular complications, identifying genes and molecular mechanisms that influence both diseases simultaneously has proven challenging. A key reason for this challenge is the pathophysiological heterogeneity underlying these diseases, with multiple processes contributing to different forms of diabetes and specific cardiovascular complications. This molecular heterogeneity has limited the effectiveness of large-scale genome-wide association studies (GWAS) in identifying shared underlying mechanisms. Additionally, our limited knowledge of the causal genes, cell types and disease-relevant states through which GWAS signals operate has hindered the discovery of common molecular pathways. This review highlights recent advances in genetic epidemiology, including studies of causal associations that have uncovered genetic and molecular factors influencing both dysglycaemia and cardiovascular complications. We explore how disease subtyping approaches can be critical in pinpointing the unique molecular signatures underlying both diabetes and cardiovascular complications. Finally, we address critical research gaps and future opportunities to advance our understanding of both diseases and translate these discoveries into tangible benefits for patient care and population health.
Collapse
Affiliation(s)
- Hyunsuk Lee
- Department of Internal Medicine, Seoul National University College of Medicine and Seoul National University Hospital, Seoul, Korea
- Department of Translational Medicine, Seoul National University College of Medicine, Seoul, Korea
- Genomic Medicine Institute, Medical Research Center, Seoul National University College of Medicine, Seoul, Korea
| | - Maria Fernandes
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Jeongeun Lee
- Department of Internal Medicine, Seoul National University College of Medicine and Seoul National University Hospital, Seoul, Korea
| | - Jordi Merino
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark.
- Diabetes Unit, Endocrine Division, Massachusetts General Hospital, Boston, MA, USA.
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA.
| | - Soo Heon Kwak
- Department of Internal Medicine, Seoul National University College of Medicine and Seoul National University Hospital, Seoul, Korea.
| |
Collapse
|
5
|
Shang J, Wu Y, Zhang L, Jiang X, Zhang R. Joint effect of modifiable risk factors and genetic susceptibility on ischaemic stroke. J Stroke Cerebrovasc Dis 2025; 34:108313. [PMID: 40252871 DOI: 10.1016/j.jstrokecerebrovasdis.2025.108313] [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: 02/05/2025] [Revised: 03/31/2025] [Accepted: 04/11/2025] [Indexed: 04/21/2025] Open
Abstract
PURPOSE To investigate the effects of modifiable risk factors and genetic susceptibility on ischaemic stroke (IS). METHODS A total of 490365 participants from the UK Biobank, with a 17-year follow-up, were included in this study. Data on 115 modifiable exposures were collected from five domains: early life, environment, lifestyle, socioeconomic status, and physical measures. Additionaly, genetic data were collected. An exposure-wide association analysis was conducted to identify potential risk factors. Risk scores for each domain and genes were calculated. The effect of each domain score on IS and the joint effects among the five domains were analyzed using multi-variate Cox models. The population attributable fraction was estimated to quantize the impact of eliminating unfavorable factors. RESULTS Sixty-four of the 115 modifiable exposures were found to be significantly associated with the risk of IS (P < 4.35 × 10-4 for Bonferroni correction). Newly identified factors included maternal smoking and being either overweight or underweight at age 10, which could significantly increase the risk of IS by 4.78 % to 14.74 %, 11.01 % to 23.75 %, and 3.29 % to 12.80 %, respectively. Additionally, exposure to hard water was associated with a decreased risk of IS by 6.96 % to 11.48 % compared to exposure to soft water. The associations varied across domains, with socioeconomic factors accounting for 5.2 % of IS cases, lifestyle accounting for 2.8 %, and physical measures accounting for 2.5 %, representing the top three contributing factors. Overall, it was estimated that 10.6 % to 11.3 % of IS cases could be prevented by eliminating the identified risks. CONCLUSIONS Interactions between risk factors and genetic susceptibility elevated the risk of IS. Risk factors from different domains contributed variably to IS, with socioeconomic factors accounting for the largest proportion.
Collapse
Affiliation(s)
- Jiao Shang
- Department of Rehabilitation, Panjin Liao-he Oil Field Gem Flower Hospital, Panjin, Liao Ning, China.
| | - Yanmei Wu
- Department of Rheumatology and Immunology, Panjin Liao-he Oil Field Gem Flower Hospital, Panjin, Liao Ning, China.
| | - Lixin Zhang
- Department of Rehabilitation, Sheng Jing Hospital of China Medical University, Shenyang, Liao Ning, China.
| | - Xueting Jiang
- Department of Rehabilitation, Panjin Liao-he Oil Field Gem Flower Hospital, Panjin, Liao Ning, China.
| | - Ruiping Zhang
- Department of Rehabilitation, Panjin Liao-he Oil Field Gem Flower Hospital, Panjin, Liao Ning, China.
| |
Collapse
|
6
|
Bragazzi NL, Zhang L, Omarov M, Georgakis MK. Genetic Risk Scores in Stroke Research and Care. Stroke 2025. [PMID: 40396275 DOI: 10.1161/strokeaha.125.050961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2025]
Abstract
Stroke remains a leading cause of death and disability worldwide. While well-established risk factors play a major role, genetic predisposition is a crucial determinant of stroke susceptibility, with heritability estimates up to 39% for ischemic stroke and 29% for intracerebral hemorrhage. Advances in next-generation sequencing and genome-wide association studies have identified numerous genetic loci associated with stroke risk, paving the way for the development of genetic risk scores. These scores aggregate information from multiple genetic variants to estimate an individual's stroke risk, offering a promising tool for personalized risk stratification that complements traditional clinical models. While GRSs have demonstrated strong predictive potential for primary stroke events in population-based settings, their integration into clinical practice remains limited. Emerging evidence suggests that GRSs could add value in clinical decision-making, for instance, for stratifying ischemic stroke risk in patients with atrial fibrillation, assessing intracerebral hemorrhage risk in anticoagulant users, and predicting vascular risk factor control in stroke survivors. The incorporation of GRSs with multiomics data and machine learning may further refine risk assessment, driving personalized prevention strategies for both primary and secondary stroke preventions. A major challenge is the limited applicability of GRS across diverse populations, as most genome-wide association studies have been conducted in individuals of European ancestry. Addressing this limitation is critical for ensuring equitable and effective implementation of GRSs in clinical settings. As methodologies continue to evolve, integrating GRS into stroke research could significantly enhance risk assessment and support precision medicine approaches tailored to individual patients.
Collapse
Affiliation(s)
- Nicola Luigi Bragazzi
- Institute for Stroke and Dementia Research, LMU University Hospital, LMU Munich, Germany (N.L.B., L.Z., M.O., M.K.G.)
| | - Lanyue Zhang
- Institute for Stroke and Dementia Research, LMU University Hospital, LMU Munich, Germany (N.L.B., L.Z., M.O., M.K.G.)
| | - Murad Omarov
- Institute for Stroke and Dementia Research, LMU University Hospital, LMU Munich, Germany (N.L.B., L.Z., M.O., M.K.G.)
| | - Marios K Georgakis
- Institute for Stroke and Dementia Research, LMU University Hospital, LMU Munich, Germany (N.L.B., L.Z., M.O., M.K.G.)
- Program in Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA (M.K.G.)
- Munich Cluster for Systems Neurology, Germany (M.K.G.)
| |
Collapse
|
7
|
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
|
8
|
Liang YY, Li MJ, Ma DR, Guo MN, Hao XY, Li SJ, Zuo CY, Hao CW, Wang ZY, Feng YM, Mao C, Zhang C, Song B, Xu Y, Shi C. Cardiovascular risk factors modulate the effect of brain imaging-derived phenotypes on ischaemic stroke risk. Brain Commun 2025; 7:fcaf183. [PMID: 40395631 PMCID: PMC12089767 DOI: 10.1093/braincomms/fcaf183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 04/13/2025] [Accepted: 05/12/2025] [Indexed: 05/27/2025] Open
Abstract
Studies have shown that cardiovascular risk factors are closely related to the occurrence of stroke, especially ischaemic stroke, as they can lead to changes in brain structure and function. However, the role of cardiovascular risk factors-induced changes in brain structure and function in the development of ischaemic stroke has not been studied. The aim of this study is thus to explore the causal association among cardiovascular risk factors, brain phenotypes and ischaemic stroke by assessing Mendelian randomization. We used univariate Mendelian randomization to sequentially investigate the causal effects of the 12 most common cardiovascular risk factors on brain structure and 3935 brain imaging-derived phenotypes in the development of ischaemic stroke. We also examined the mediating effect of brain structure on blood pressure-induced ischaemic stroke using a multivariable Mendelian randomization test. We tested the reliability of our results using the Steiger test, heterogeneity test, horizontal pleiotropy test and leave-one-out method. We found that 8 of the 12 examined cardiovascular risk factors were associated with 538 brain imaging-derived phenotypes, and 9 of the 12 cardiovascular risk factors were associated with IS. The main cardiovascular risk factors associated with brain imaging-derived phenotypes and ischaemic stroke was blood pressure (systolic and diastolic), which can affect the occurrence of ischaemic stroke through 6 types of brain imaging-derived phenotypes. However, extrapolation of our findings to other ethnic groups is challenging, and the possibility of reverse causality cannot be completely ruled out. This study identifies the role of cardiovascular risk factors, especially blood pressure, in affecting brain structure and ischaemic stroke risk. The findings assist in early risk detection and enhance stroke prevention strategies, also hinting at non-vascular factors' involvement.
Collapse
Affiliation(s)
- Yuan-yuan Liang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Meng-jie Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan 450000, China
- Department of Neurology, Academy of Medical Sciences of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Dong-rui Ma
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Meng-nan Guo
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Xiao-yan Hao
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan 450000, China
- Department of Neurology, Academy of Medical Sciences of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Shuang-jie Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Chun-yan Zuo
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Chen-wei Hao
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Zhi-yun Wang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Yan-mei Feng
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Chenyuan Mao
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Chan Zhang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Bo Song
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan 450000, China
- NHC Key Laboratory of Prevention and treatment of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan 450000, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan 450000, China
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Yuming Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan 450000, China
- NHC Key Laboratory of Prevention and treatment of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan 450000, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan 450000, China
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Changhe Shi
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan 450000, China
- NHC Key Laboratory of Prevention and treatment of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan 450000, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan 450000, China
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, Henan 450000, China
| |
Collapse
|
9
|
Myserlis EP, Georgakis MK, Parodi L, Mayerhofer E, Omarov M, Rosand J, Banerjee C, Anderson CD. A Beneficial Role for Gluteofemoral Adipose Tissue in Cerebrovascular Disease: Causal Pathway and Mediation Analysis. Neurology 2025; 104:e213573. [PMID: 40228186 DOI: 10.1212/wnl.0000000000213573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 02/18/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND AND OBJECTIVES Previous studies have shown that increased body fat is associated with stroke risk, with evidence suggesting that body fat distribution, rather than total body fat, exerts a more prominent role in cerebrovascular risk prediction. In this study, we explore causal associations between body mass index (BMI)-independent adipose tissue distribution profiles and cerebrovascular disease (CVD) risk, aiming to refine the association between body fat distribution and stroke. METHODS We selected variants associated with BMI-independent visceral adipose tissue (VAT), abdominal subcutaneous adipose tissue (ASAT), and gluteofemoral adipose tissue (GFAT) volumes in UK Biobank, and performed univariable and multivariable Mendelian randomization (MR) analyses with ischemic stroke (IS) and subtypes (large artery stroke [LAS], cardioembolic stroke [CES], and small vessel stroke [SVS]). We used coronary artery disease (CAD), carotid intima media thickness (cIMT), and MRI-confirmed lacunar stroke as positive controls. We explored the mediatory role of common cardiovascular (systolic blood pressure, diabetes, and low-density lipoprotein), insulin resistance, inflammatory (C-reactive protein), and adipose tissue-specific (adiponectin, leptin) factors by performing 2-step mediation MR analyses. Estimates were expressed per standard deviation increase in adjusted adipose tissue volume. RESULTS Genetic predisposition to higher GFAT volume was associated with lower risk of IS (odds ratio [OR] 0.92, 95% CI 0.86-0.98), LAS (OR 0.80, 95% CI 0.66-0.96), and SVS (OR 0.77, 95% CI 0.67-0.88), but not CES, consistent in multivariable analyses. Genetic predisposition to higher GFAT volume was also associated with lower risk of CAD (OR 0.82, 95% CI 0.76-0.88), lacunar stroke (OR 0.78, 95% CI 0.67-0.92), and mean cIMT (β = -0.073, 95% CI -0.114 to -0.031). Associations were largely consistent in sensitivity analyses. No association was observed between genetic predisposition to ASAT or VAT and IS risk. Although common vascular risk factors were the predominant mediators in the GFAT-CVD axis, adiponectin and leptin mediated a proportion of IS and CAD risk (∼15% (1.8%-57%) and ∼4.6% (0.8%-13.5%) mediated by adiponectin, respectively). DISCUSSION This study supports a protective role of gluteofemorally distributed fat volume in CVD risk. Although this role is predominantly mediated by common vascular risk factor modification, adipose tissue-specific factors may exert a mediatory effect, suggesting a possible novel target for attenuating adiposity-related CVD risk.
Collapse
Affiliation(s)
| | - Marios K Georgakis
- Institute for Stroke and Dementia Research (ISD), Ludwig-Maximilians-University (LMU) Hospital, Munich, Germany
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Boston, MA
| | - Livia Parodi
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Boston, MA
- Department of Neurology, Brigham and Women's Hospital, Boston, MA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston
| | - Ernst Mayerhofer
- Center for Genomic Medicine, Massachusetts General Hospital, Boston
| | - Murad Omarov
- Institute for Stroke and Dementia Research, University Hospital of LMU Munich, Germany
| | - Jonathan Rosand
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Boston, MA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston
- Department of Neurology, Massachusetts General Hospital, Boston; and
- McCance Center for Brain Health, Massachusetts General Hospital, Boston
| | - Chirantan Banerjee
- Department of Neurology, Medical University of South Carolina, Charleston
| | - Christopher D Anderson
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Boston, MA
- Department of Neurology, Brigham and Women's Hospital, Boston, MA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston
- McCance Center for Brain Health, Massachusetts General Hospital, Boston
| |
Collapse
|
10
|
Suleman S, Ängquist L, Linneberg A, Hansen T, Grarup N. Exploring the genetic intersection between obesity-associated genetic variants and insulin sensitivity indices. Sci Rep 2025; 15:15761. [PMID: 40328835 PMCID: PMC12056085 DOI: 10.1038/s41598-025-98507-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Accepted: 04/11/2025] [Indexed: 05/08/2025] Open
Abstract
Insulin sensitivity (IS) is a key determinant of metabolic health and may share genetic factors with obesity-related traits. Previous large-scale genetic studies have identified variants associated with IS as well as obesity related traits like body mass index (BMI) and waist-to-hip ratio (WHR). Notably, many of these associations are shared across traits, indicating a potential genetic overlap. However, the genetic intersection between IS and obesity-related traits remains underexplored. To explore this gap, we investigated associations between six IS indices, including fasting and post-glucose load measures, and genetic variants linked to BMI and WHR to determine their influence on IS and related cardiometabolic traits. To achieve this, we calculated six IS indices using fasting and oral glucose tolerance test (OGTT) data from 5,007 non-diabetic individuals, grouping them into fasting, OGTT0,120, and OGTT0,30,120 categories. A total of 678 BMI-associated and 265 WHR-associated genetic variants were analysed using linear regression, adjusting for age and sex, with sex-specific analyses for WHR. Analyses were conducted with and without BMI adjustments and corrected for multiple testing (padj). Additionally, we explored the relationship between IS-linked variants and their associations with type 2 diabetes (T2D), coronary artery disease (CAD) and stroke. Among the 678 BMI-associated variants, 100 showed nominal associations (p < 0.05) with at least one IS index; and 20 remained significant after multiple testing correction (padj < 0.05) when not adjusting for BMI. After adjusting for BMI, 70 variants retained nominal associations, and six remained significant (padj < 0.05). In sex-specific analyses of the 265 WHR-associated variants, 12 variants were associated in females when adjusted for BMI, whereas no significant associations were observed in males. Furthermore, BMI- and WHR-associated variants linked to decreased IS, such as those in FTO and VPS13C loci, were also associated with increased T2D and stroke risk, whereas IS-increasing variants, including those in VPS13C and PPARG, were linked to lower T2D and stroke risk, with some, like THADA, showing opposing effects on CAD. This study offers insights into genetic variants that influence both IS and obesity-related traits, revealing BMI- and WHR-associated variants with both positive and negative effects on IS and their potential impact on cardiometabolic health.
Collapse
Affiliation(s)
- Sufyan Suleman
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Biomedicine, Human Genetics, Aarhus University, Aarhus, 8000, Denmark
| | - Lars Ängquist
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Allan Linneberg
- Center for Clinical Research and Prevention, Copenhagen University Hospital-Bispebjerg and Frederiksberg, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Niels Grarup
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
11
|
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
|
12
|
Kaiser R, Gold C, Stark K. Recent Advances in Immunothrombosis and Thromboinflammation. Thromb Haemost 2025. [PMID: 40311639 DOI: 10.1055/a-2523-1821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
Inflammation and thrombosis are traditionally considered two separate entities of acute host responses to barrier breaks. While inciting inflammatory responses is a prerequisite to fighting invading pathogens and subsequent restoration of tissue homeostasis, thrombus formation is a crucial step of the hemostatic response to prevent blood loss following vascular injury. Though originally designed to protect the host, excessive induction of either inflammatory signaling or thrombus formation and their reciprocal activation contribute to a plethora of disorders, including cardiovascular, autoimmune, and malignant diseases. In this state-of-the-art review, we summarize recent insights into the intricate interplay of inflammation and thrombosis. We focus on the protective aspects of immunothrombosis as well as evidence of detrimental sequelae of thromboinflammation, specifically regarding recent studies that elucidate its pathophysiology beyond coronavirus disease 2019 (COVID-19). We introduce recently identified molecular aspects of key cellular players like neutrophils, monocytes, and platelets that contribute to both immunothrombosis and thromboinflammation. Further, we describe the underlying mechanisms of activation involving circulating plasma proteins and immune complexes. We then illustrate how these factors skew the inflammatory state toward detrimental thromboinflammation across cardiovascular as well as septic and autoimmune inflammatory diseases. Finally, we discuss how the advent of new technologies and the integration with clinical data have been used to investigate the mechanisms and signaling cascades underlying immunothrombosis and thromboinflammation. This review highlights open questions that will need to be addressed by the field to translate our mechanistic understanding into clinically meaningful therapeutic targeting.
Collapse
Affiliation(s)
- Rainer Kaiser
- Medizinische Klinik und Poliklinik I, University Hospital Ludwig-Maximilian University, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Christoph Gold
- Medizinische Klinik und Poliklinik I, University Hospital Ludwig-Maximilian University, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Konstantin Stark
- Medizinische Klinik und Poliklinik I, University Hospital Ludwig-Maximilian University, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
13
|
Fan Y, Lu D, Yang C, Song Z, Chen Y, Ma Y, Li J, Zhang H. Multiomic Underpinnings of Drug Targets for Intracranial Aneurysm: Evidence From Diversified Mendelian Randomization. CNS Neurosci Ther 2025; 31:e70430. [PMID: 40346920 PMCID: PMC12064948 DOI: 10.1111/cns.70430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 04/27/2025] [Accepted: 04/28/2025] [Indexed: 05/12/2025] Open
Abstract
AIMS The absence of pharmaceutics poses challenges in preventing intracranial aneurysm (IA) progression and rupture. This research emphasized identifying drug targets for IA through a druggable genome-wide Mendelian randomization (MR) analysis. METHODS A two-sample MR analysis was performed leveraging cis-expression quantitative trait loci in the blood (n = 31,684) and arteries (n = 584) aligned with 5883 druggable genes as exposure and the largest IA summary statistics (n = 7495) as outcome. Bayesian colocalization analysis, plasma cis-protein quantitative trait loci (n = 35,559), and external IA cohorts (FinnGen, n = 2582; Zhou, n = 380) were used for validation. A phenome-wide MR (Phe-MR) incorporating 783 diseases uncovered side effects. Multivariable MR addressed unmeasured pleiotropy. RESULTS Five druggable genes in blood and one in the coronary artery showed significant association with IA risk (p-FDR ≤ 0.05). NT5C2, PRCP, and CRMP1 shared a common variant with IA (PPH4 ≥ 0.8). The external validation cohorts confirmed the effects of NT5C2 on IA (FinnGen cohort, Odds Ratio [OR], 0.81, 95% Confidential Interval [95% CI] 95% CI, 0.707-0.930; p = 0.003; Zhou cohort, OR, 0.68, 95% CI, 0.469-0.983; p = 0.041). The genetically predicted protein level of PRCP validated an inverse association with IA risk (OR, 0.734; 95% CI, 0.561-0.959; p = 0.023). The Phe-MR revealed insignificance for NT5C2 or PRCP. Direct causal effects on IA were 0.60 (95% CI, 0.457-0.797; p = 1.36E-05) for PRCP and 0.67 (95% CI, 0.527-0.860; p = 0.002) for NT5C2 after adjusting for IA modifiable risk factors. CONCLUSIONS NT5C2 and PRCP were identified as potential drug targets for IA, with effects independent of known modifiable risk factors. Targeting NT5C2 and PRCP appeared exclusively effective and safe.
Collapse
Affiliation(s)
- Yu‐Xiang Fan
- Department of Neurosurgery, Xuanwu HospitalCapital Medical UniversityBeijingChina
- China International Neuroscience Institute (China‐INI), Xuanwu HospitalCapital Medical UniversityBeijingChina
| | - Di Lu
- Department of Neurosurgery, Xuanwu HospitalCapital Medical UniversityBeijingChina
- China International Neuroscience Institute (China‐INI), Xuanwu HospitalCapital Medical UniversityBeijingChina
| | - Cheng‐Bin Yang
- Department of Neurosurgery, Xuanwu HospitalCapital Medical UniversityBeijingChina
- China International Neuroscience Institute (China‐INI), Xuanwu HospitalCapital Medical UniversityBeijingChina
| | - Zi‐Hao Song
- Department of Neurosurgery, Xuanwu HospitalCapital Medical UniversityBeijingChina
- China International Neuroscience Institute (China‐INI), Xuanwu HospitalCapital Medical UniversityBeijingChina
| | - Yi‐Guang Chen
- Department of Neurosurgery, Xuanwu HospitalCapital Medical UniversityBeijingChina
| | - Yong‐Jie Ma
- Department of Neurosurgery, Xuanwu HospitalCapital Medical UniversityBeijingChina
- China International Neuroscience Institute (China‐INI), Xuanwu HospitalCapital Medical UniversityBeijingChina
| | - Jing‐Wei Li
- Department of Neurosurgery, Xuanwu HospitalCapital Medical UniversityBeijingChina
- China International Neuroscience Institute (China‐INI), Xuanwu HospitalCapital Medical UniversityBeijingChina
| | - Hong‐Qi Zhang
- Department of Neurosurgery, Xuanwu HospitalCapital Medical UniversityBeijingChina
- China International Neuroscience Institute (China‐INI), Xuanwu HospitalCapital Medical UniversityBeijingChina
| |
Collapse
|
14
|
Koloi A, Rydin A, Milaneschi Y, Lamers F, Bosch JA, Pruin E, van der Laan SW, Mishra PP, Lehtimäki T, Kähönen M, Raitakari OT, Fotiadis DI, Quax R. Morbidity-bridging metabolic pathways: linking early cardiovascular disease risk and depression symptoms using a multi-modal approach. EUROPEAN HEART JOURNAL OPEN 2025; 5:oeaf038. [PMID: 40329991 PMCID: PMC12053008 DOI: 10.1093/ehjopen/oeaf038] [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: 03/26/2025] [Accepted: 03/31/2025] [Indexed: 05/08/2025]
Abstract
Aims Prevalence of cardiovascular diseases (CVDs) and depression is rising globally. Their co-occurrence associates with poorer outcomes, potentially due to shared metabolic pathways. This study aimed to identify metabolic pathways linking depression symptoms and CVD risk factors. Methods and results Data from the Young Finns Study (YFS, n = 1,599, mean age 37 ± 5, 54% female) served as input for a network (mixed graphical models). Confirmatory analysis through covariate-adjusted regression was done with UK Biobank (UKB, n = 69,513, mean age 63 ± 7, 64% female). Mendelian randomization assessed causality using genome-wide association studies data. The study examined 52 plasma metabolites measured by nuclear magnetic resonance spectroscopy. Outcomes included depression symptoms (BDI in YFS, PHQ-9 in UKB) and CVD risk factors [systolic/diastolic blood pressure, carotid intima-media thickness (cIMT)]. Mendelian randomization inferred causal links between metabolites and depression or (intermediate markers of) CVD. Two bridge metabolites were identified: glucose linked to sleep pattern (P = 0.034); omega-3 fatty acids (FAs) linked to appetite change (P < 0.001); and both connected to cIMT (both P = 0.002). Mendelian randomization suggested glucose as causal in coronary artery disease (CAD) risk, while omega-3 FAs showed potential causal links to CAD, coronary artery calcification, and cIMT. Conclusion This study integrated three statistical techniques and identified two metabolic markers (glucose, omega-3 FAs) connecting depression and CVD on a symptom and risk factor level. The associations, established in a relatively young cohort, were replicated in a predominantly middle-aged cohort and emphasize both the generalizability of the findings across different populations and value of symptom-level analysis in depression and CVD comorbidity research.
Collapse
Affiliation(s)
- Angela Koloi
- Unit of Medical Technology and Intelligent Information Systems, Department of Materials Science and Engineering, University of Ioannina, Ioannina, Greece
- Department of Biological Applications and Technology, University of Ioannina, Ioannina, Greece
- Department of Clinical Psychology, University of Amsterdam, Amsterdam, The Netherlands
| | - Arja Rydin
- Department of Psychiatry, Amsterdam UMC Location Vrije Universiteit Amsterdam, Boelelaan, Amsterdam 1117, The Netherlands
- Amsterdam Public Health, Mental Health Program, Amsterdam, The Netherlands
| | - Yuri Milaneschi
- Department of Psychiatry, Amsterdam UMC Location Vrije Universiteit Amsterdam, Boelelaan, Amsterdam 1117, The Netherlands
- Amsterdam Public Health, Mental Health Program, Amsterdam, The Netherlands
- Amsterdam Public Health, Methodology Program, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Mood, Anxiety, Psychosis, Sleep & Stress Program, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Complex Trait Genetics, Amsterdam, The Netherlands
| | - Femke Lamers
- Department of Psychiatry, Amsterdam UMC Location Vrije Universiteit Amsterdam, Boelelaan, Amsterdam 1117, The Netherlands
- Amsterdam Public Health, Mental Health Program, Amsterdam, The Netherlands
| | - Jos A Bosch
- Department of Clinical Psychology, University of Amsterdam, Amsterdam, The Netherlands
- Department of medical Psychology, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Emma Pruin
- Department of Psychiatry, Amsterdam UMC Location Vrije Universiteit Amsterdam, Boelelaan, Amsterdam 1117, The Netherlands
- Amsterdam Public Health, Mental Health Program, Amsterdam, The Netherlands
- Amsterdam Public Health, Methodology Program, Amsterdam, The Netherlands
| | - Sander W van der Laan
- Central Diagnostic Laboratory, Division Laboratories, Pharmacy, and Biomedical Genetics, University Medical Center Utrecht, Utrecht University, The Netherlands
- Department of Genomic Sciences, University of Virginia, Charlottesville, VA, USA
| | - Pashupati P Mishra
- Department of Clinical Chemistry, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Faculty of Medicine and Health Technology, Finnish Cardiovascular Research Center Tampere, Tampere University, Tampere, Finland
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Faculty of Medicine and Health Technology, Finnish Cardiovascular Research Center Tampere, Tampere University, Tampere, Finland
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
| | - Mika Kähönen
- Faculty of Medicine and Health Technology, Finnish Cardiovascular Research Center Tampere, Tampere University, Tampere, Finland
- Department of Clinical Physiology, Tampere University Hospital, Tampere, Finland
| | - Olli T Raitakari
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
| | - Dimitrios I Fotiadis
- Unit of Medical Technology and Intelligent Information Systems, Department of Materials Science and Engineering, University of Ioannina, Ioannina, Greece
- Biomedical Research Institute, Foundation for Research and Technology - Hellas (FORTH), Ioannina, Greece
| | - Rick Quax
- Computational Science Lab, Institute of Informatics, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
15
|
Tang C, Xu M, Ruan R, Pan B, Luo J, Huang J, Cheng J, Li H, Zhang Y, Zhang Z. The relationship between COVID-19 and stroke and its risk factors, a Mendelian randomization analysis. INTERNATIONAL JOURNAL OF ENVIRONMENTAL HEALTH RESEARCH 2025:1-12. [PMID: 40304158 DOI: 10.1080/09603123.2025.2490187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 04/03/2025] [Indexed: 05/02/2025]
Abstract
BACKGROUND It remains unclear about the association between stroke and Coronavirus disease 2019 (COVID-19). Therefore, a Mendelian randomization (MR) analysis was conducted to explore the relationship between them. METHODS In this study, the most recent large-scale genome-wide association studies (GWASs) were selected from the publicly available COVID-19 GWAS meta-analysis (Round 7) as the exposure. Data from a recent original stroke GWAS were used as the outcome for the experimental group, while the stroke GWAS data from the IEU GWAS database were used as the validation group. In addition, an MR analysis was conducted to explore the causal relationships of susceptibility, hospitalization, and severity of COVID-19 with stroke and its subtypes. In addition, the results of the experimental and validation groups were integrated to perform a meta-analysis. RESULTS The MR analysis results corroborated that there was a positively causal relationship between the hospitalization (OR, 1.11; p = 0.015) (ORmeta, 1.11; p < 0.001), and severity (OR, 1.06; p = 0.043) (ORmeta, 1.07; p = 0.002) of COVID-19 and cardioembolic stroke (CES). This result is supported by the validation group and sensitivity analysis. CONCLUSION This study demonstrates for the first time that COVID-19 hospitalization and COVID-19 severity are risk factors for CES.
Collapse
Affiliation(s)
- Chao Tang
- Jinzhou Medical University, Jinzhou, China
| | | | | | - Bingxiao Pan
- The Second Affiliated Hospital of China Medical University, Shenyang, China
| | - Jia Luo
- Shenyang University of technology, Shenyang, China
| | - Jing Huang
- Jinzhou Medical University, Jinzhou, China
| | - Junyao Cheng
- The People's hospital of Yuechi County, Yuechi, China
| | - Hangxu Li
- The Third Clinical Medical College of Jinzhou Medical University, Jinzhou, China
| | - Yinan Zhang
- Department of Neurosurgery, General Hospital of Fushun Mining Bureau of Liaoning Health Industry Group, Fushun, China
| | - Zhenxing Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| |
Collapse
|
16
|
Yao P, Mazidi M, Pozarickij A, Iona A, Wright N, Lin K, Millwood I, Fry H, Kartsonaki C, Chen Y, Yang L, Du H, Avery D, Schmidt D, Sun D, Lv J, Yu C, Hill M, Bennett D, Walters R, Li L, Clarke R, Chen Z. Proteome-Wide Genetic Study in East Asians and Europeans Identified Multiple Therapeutic Targets for Ischemic Stroke. Stroke 2025. [PMID: 40304040 DOI: 10.1161/strokeaha.125.050982] [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: 02/05/2025] [Revised: 03/18/2025] [Accepted: 04/11/2025] [Indexed: 05/02/2025]
Abstract
BACKGROUND Analyses of genomic and proteomics data in prospective biobank studies in diverse populations may discover novel or repurposing drug targets for stroke. METHODS We extracted individual cis-protein quantitative trait locus for 2923 proteins measured using Olink Explore panel from a genome-wide association study in prospective China Kadoorie Biobank and UK Biobank, both established ≈20 years ago. These cis-protein quantitative trait loci were used in ancestry-specific 2-sample Mendelian randomization analyses of ischemic stroke (IS) in East Asians (n=22 664 cases) and Europeans (n=62 100 cases). We further undertook colocalization analyses to examine the shared causal variants of cis-protein quantitative trait locus with stroke, along with various downstream analyses (eg, phenome-wide association study, drug development lookups) to clarify mechanisms of action and druggability. RESULTS In Mendelian randomization analyses, the genetically predicted plasma levels of 10 proteins were significantly associated with IS in East Asians (n=2) and Europeans (n=9), with 6 proteins (FGF5 [fibroblast growth factor 5], TMPRSS5 [transmembrane protease serine 5], FURIN, F11 [coagulation factor XI], ALDH2 [aldehyde dehydrogenase 2], and ABO) showing positive and 4 (GRK5 [G protein-coupled receptor kinase 5], KIAA0319 [dyslexia-associated protein KIAA0319], PROCR [endothelial protein C receptor], and MMP12 [macrophage metalloelastase 12]) showing inverse associations, all directionally consistent between East Asians and Europeans. Colocalization analyses provided strong evidence (posterior probabilities for the H4 hypothesis ≥0.7) of shared genetic variants with IS for 9 out of 10 proteins (except ABO). Moreover, 8 proteins were also causally associated, in the expected directions, with systolic blood pressure (positive/inverse: 4/2), low-density lipoprotein cholesterol (1 positive), body mass index (1 inverse), type 2 diabetes (2/1), or atrial fibrillation (3/1). Phenome-wide association study analyses and lookups in knock-out mouse models confirmed their importance for IS or stroke-related traits (eg, hematologic phenotypes). Of these 10 proteins, 1 was not druggable (ABO), 3 had known primary (F11) or potentially repurposed (ALDH2, MMP12) drug targets for stroke, and 6 (PROCR, GRK5, FGF5, FURIN, KIAA0319, and TMPRSS5) had no evidence of any drug targets. CONCLUSIONS Proteogenomic investigation in diverse ancestry populations identified the causal relevance of 10 proteins for IS, with several being potentially novel or repurposed targets that could be prioritized for further investigation.
Collapse
Affiliation(s)
- Pang Yao
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (P.Y., M.M., A.P., A.I., N.W., K.L., I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, M.H., D.B., R.W., R.C., Z.C.)
| | - Mohsen Mazidi
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (P.Y., M.M., A.P., A.I., N.W., K.L., I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, M.H., D.B., R.W., R.C., Z.C.)
| | - Alfred Pozarickij
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (P.Y., M.M., A.P., A.I., N.W., K.L., I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, M.H., D.B., R.W., R.C., Z.C.)
| | - Andri Iona
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (P.Y., M.M., A.P., A.I., N.W., K.L., I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, M.H., D.B., R.W., R.C., Z.C.)
| | - Neil Wright
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (P.Y., M.M., A.P., A.I., N.W., K.L., I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, M.H., D.B., R.W., R.C., Z.C.)
| | - Kuang Lin
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (P.Y., M.M., A.P., A.I., N.W., K.L., I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, M.H., D.B., R.W., R.C., Z.C.)
| | - Iona Millwood
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (P.Y., M.M., A.P., A.I., N.W., K.L., I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, M.H., D.B., R.W., R.C., Z.C.)
- Medical Research Council Health Research Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, D.B., R.W., Z.C.)
| | - Hannah Fry
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (P.Y., M.M., A.P., A.I., N.W., K.L., I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, M.H., D.B., R.W., R.C., Z.C.)
- Medical Research Council Health Research Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, D.B., R.W., Z.C.)
| | - Christiana Kartsonaki
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (P.Y., M.M., A.P., A.I., N.W., K.L., I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, M.H., D.B., R.W., R.C., Z.C.)
- Medical Research Council Health Research Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, D.B., R.W., Z.C.)
| | - Yiping Chen
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (P.Y., M.M., A.P., A.I., N.W., K.L., I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, M.H., D.B., R.W., R.C., Z.C.)
- Medical Research Council Health Research Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, D.B., R.W., Z.C.)
| | - Ling Yang
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (P.Y., M.M., A.P., A.I., N.W., K.L., I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, M.H., D.B., R.W., R.C., Z.C.)
- Medical Research Council Health Research Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, D.B., R.W., Z.C.)
| | - Huaidong Du
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (P.Y., M.M., A.P., A.I., N.W., K.L., I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, M.H., D.B., R.W., R.C., Z.C.)
- Medical Research Council Health Research Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, D.B., R.W., Z.C.)
| | - Daniel Avery
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (P.Y., M.M., A.P., A.I., N.W., K.L., I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, M.H., D.B., R.W., R.C., Z.C.)
- Medical Research Council Health Research Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, D.B., R.W., Z.C.)
| | - Dan Schmidt
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (P.Y., M.M., A.P., A.I., N.W., K.L., I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, M.H., D.B., R.W., R.C., Z.C.)
- Medical Research Council Health Research Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, D.B., R.W., Z.C.)
| | - Dianjianyi Sun
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China (D. Sun, J.L., C.Y., L.L.)
- Peking University Center for Public Health and Epidemic Preparedness & Response, Beijing, China (D. Sun, P.P., J.L., C.Y., L.L.)
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China (D. Sun, J.L., C.Y., L.L.)
| | - Jun Lv
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China (D. Sun, J.L., C.Y., L.L.)
- Peking University Center for Public Health and Epidemic Preparedness & Response, Beijing, China (D. Sun, P.P., J.L., C.Y., L.L.)
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China (D. Sun, J.L., C.Y., L.L.)
| | - Canqing Yu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China (D. Sun, J.L., C.Y., L.L.)
- Peking University Center for Public Health and Epidemic Preparedness & Response, Beijing, China (D. Sun, P.P., J.L., C.Y., L.L.)
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China (D. Sun, J.L., C.Y., L.L.)
| | - Michael Hill
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (P.Y., M.M., A.P., A.I., N.W., K.L., I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, M.H., D.B., R.W., R.C., Z.C.)
| | - Derrick Bennett
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (P.Y., M.M., A.P., A.I., N.W., K.L., I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, M.H., D.B., R.W., R.C., Z.C.)
- Medical Research Council Health Research Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, D.B., R.W., Z.C.)
| | - Robin Walters
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (P.Y., M.M., A.P., A.I., N.W., K.L., I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, M.H., D.B., R.W., R.C., Z.C.)
- Medical Research Council Health Research Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, D.B., R.W., Z.C.)
| | - Liming Li
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China (D. Sun, J.L., C.Y., L.L.)
- Peking University Center for Public Health and Epidemic Preparedness & Response, Beijing, China (D. Sun, P.P., J.L., C.Y., L.L.)
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China (D. Sun, J.L., C.Y., L.L.)
| | - Robert Clarke
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (P.Y., M.M., A.P., A.I., N.W., K.L., I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, M.H., D.B., R.W., R.C., Z.C.)
| | - Zhengming Chen
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (P.Y., M.M., A.P., A.I., N.W., K.L., I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, M.H., D.B., R.W., R.C., Z.C.)
- Medical Research Council Health Research Unit, Nuffield Department of Population Health, University of Oxford, United Kingdom. (I.M., H.F., C.K., Y.C., L.Y., H.D., D.A., D. Schmidt, D.B., R.W., Z.C.)
| |
Collapse
|
17
|
Clarke R, Wright N, Lin K, Yu C, Walters RG, Lv J, Hill M, Kartsonaki C, Millwood IY, Bennett DA, Avery D, Yang L, Chen Y, Du H, Sherliker P, Yang X, Sun D, Li L, Qu C, Marcovina S, Collins R, Chen Z, Parish S. Causal Relevance of Lp(a) for Coronary Heart Disease and Stroke Types in East Asian and European Ancestry Populations: A Mendelian Randomization Study. Circulation 2025. [PMID: 40297899 DOI: 10.1161/circulationaha.124.072086] [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: 10/11/2024] [Accepted: 03/25/2025] [Indexed: 04/30/2025]
Abstract
BACKGROUND Elevated plasma levels of Lp(a) [lipoprotein(a)] are a causal risk factor for coronary heart disease and stroke in European individuals, but the causal relevance of Lp(a) for different stroke types and in East Asian individuals with different Lp(a) genetic architecture is uncertain. METHODS We measured plasma levels of Lp(a) in a nested case-control study of 18 174 adults (mean [SD] age, 57 [10] years; 49% female) in the China Kadoorie Biobank (CKB) and performed a genome-wide association analysis to identify genetic variants affecting Lp(a) levels, with replication in ancestry-specific subsets in UK Biobank. We further performed 2-sample Mendelian randomization analyses, associating ancestry-specific Lp(a)-associated instrumental variants derived from CKB or from published data in European individuals with risk of myocardial infarction (n=17 091), ischemic stroke (IS [n=29 233]) and its subtypes, or intracerebral hemorrhage (n=5845) in East Asian and European individuals using available data from CKB and genome-wide association analysis consortia. RESULTS In CKB observational analyses, plasma levels of Lp(a) were log-linearly and positively associated with higher risks of myocardial infarction and IS, but not with ICH. In genome-wide association analysis, we identified 29 single nucleotide polymorphisms independently associated with Lp(a) that together explained 33% of variance in Lp(a) in Chinese individuals. In UK Biobank, the lead Chinese variants identified in CKB were replicated in 1260 Chinese individuals, but explained only 10% of variance in Lp(a) in European individuals. In Mendelian randomization analyses, however, there were highly concordant effects of Lp(a) across both ancestries for all cardiovascular disease outcomes examined. In combined analyses of both ancestries, the proportional reductions in risk per 100 nmol/L lower genetically predicted Lp(a) levels for myocardial infarction were 3-fold greater than for total IS (rate ratio, 0.78 [95% CI, 0.76-0.81] versus 0.94 [0.92-0.96]), but were similar to those for large-artery IS (0.80 [0.73-0.87]; n=8134). There were weaker associations with cardioembolic IS (0.92 [95% CI, 0.86-0.98]; n=11 730), and no association with small-vessel IS (0.99 [0.91-1.07]; n=12 343) or with intracerebral hemorrhage (1.08 [0.96-1.21]; n=5845). CONCLUSIONS The effects of Lp(a) on risk of myocardial infarction and large-artery IS were comparable in East Asian and European individuals, suggesting that people with either ancestry could expect comparable proportional benefits for equivalent reductions in Lp(a), but there was little effect on other stroke types.
Collapse
Affiliation(s)
- Robert Clarke
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, UK (R. Clarke, N.W., K.L., R.G.W., M.H., C.K., I.Y.M., D.A.B., D.A., L.Y., Y.C., H.D., P.S., X.Y., R. Collins, Z.C. S.P.)
| | - Neil Wright
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, UK (R. Clarke, N.W., K.L., R.G.W., M.H., C.K., I.Y.M., D.A.B., D.A., L.Y., Y.C., H.D., P.S., X.Y., R. Collins, Z.C. S.P.)
| | - Kuang Lin
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, UK (R. Clarke, N.W., K.L., R.G.W., M.H., C.K., I.Y.M., D.A.B., D.A., L.Y., Y.C., H.D., P.S., X.Y., R. Collins, Z.C. S.P.)
| | - Canqing Yu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China (C.Y., J.L., D.S., L.L.)
- Peking University Center for Public Health and Epidemic Preparedness and Response, Beijing, China (C.Y., J.L., D.S., L.L.)
- Key Laboratory of Major Diseases (Peking University), Ministry of Education, Beijing, China (C.Y., J.L., D.S., L.L.)
| | - Robin G Walters
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, UK (R. Clarke, N.W., K.L., R.G.W., M.H., C.K., I.Y.M., D.A.B., D.A., L.Y., Y.C., H.D., P.S., X.Y., R. Collins, Z.C. S.P.)
| | - Jun Lv
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China (C.Y., J.L., D.S., L.L.)
- Peking University Center for Public Health and Epidemic Preparedness and Response, Beijing, China (C.Y., J.L., D.S., L.L.)
- Key Laboratory of Major Diseases (Peking University), Ministry of Education, Beijing, China (C.Y., J.L., D.S., L.L.)
| | - Michael Hill
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, UK (R. Clarke, N.W., K.L., R.G.W., M.H., C.K., I.Y.M., D.A.B., D.A., L.Y., Y.C., H.D., P.S., X.Y., R. Collins, Z.C. S.P.)
| | - Christiana Kartsonaki
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, UK (R. Clarke, N.W., K.L., R.G.W., M.H., C.K., I.Y.M., D.A.B., D.A., L.Y., Y.C., H.D., P.S., X.Y., R. Collins, Z.C. S.P.)
| | - Iona Y Millwood
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, UK (R. Clarke, N.W., K.L., R.G.W., M.H., C.K., I.Y.M., D.A.B., D.A., L.Y., Y.C., H.D., P.S., X.Y., R. Collins, Z.C. S.P.)
| | - Derrick A Bennett
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, UK (R. Clarke, N.W., K.L., R.G.W., M.H., C.K., I.Y.M., D.A.B., D.A., L.Y., Y.C., H.D., P.S., X.Y., R. Collins, Z.C. S.P.)
| | - Daniel Avery
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, UK (R. Clarke, N.W., K.L., R.G.W., M.H., C.K., I.Y.M., D.A.B., D.A., L.Y., Y.C., H.D., P.S., X.Y., R. Collins, Z.C. S.P.)
| | - Ling Yang
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, UK (R. Clarke, N.W., K.L., R.G.W., M.H., C.K., I.Y.M., D.A.B., D.A., L.Y., Y.C., H.D., P.S., X.Y., R. Collins, Z.C. S.P.)
| | - Yiping Chen
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, UK (R. Clarke, N.W., K.L., R.G.W., M.H., C.K., I.Y.M., D.A.B., D.A., L.Y., Y.C., H.D., P.S., X.Y., R. Collins, Z.C. S.P.)
| | - Huaidong Du
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, UK (R. Clarke, N.W., K.L., R.G.W., M.H., C.K., I.Y.M., D.A.B., D.A., L.Y., Y.C., H.D., P.S., X.Y., R. Collins, Z.C. S.P.)
| | - Paul Sherliker
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, UK (R. Clarke, N.W., K.L., R.G.W., M.H., C.K., I.Y.M., D.A.B., D.A., L.Y., Y.C., H.D., P.S., X.Y., R. Collins, Z.C. S.P.)
| | - Xiaoming Yang
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, UK (R. Clarke, N.W., K.L., R.G.W., M.H., C.K., I.Y.M., D.A.B., D.A., L.Y., Y.C., H.D., P.S., X.Y., R. Collins, Z.C. S.P.)
| | - Dianjianyi Sun
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China (C.Y., J.L., D.S., L.L.)
- Peking University Center for Public Health and Epidemic Preparedness and Response, Beijing, China (C.Y., J.L., D.S., L.L.)
- Key Laboratory of Major Diseases (Peking University), Ministry of Education, Beijing, China (C.Y., J.L., D.S., L.L.)
| | - Liming Li
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China (C.Y., J.L., D.S., L.L.)
- Peking University Center for Public Health and Epidemic Preparedness and Response, Beijing, China (C.Y., J.L., D.S., L.L.)
- Key Laboratory of Major Diseases (Peking University), Ministry of Education, Beijing, China (C.Y., J.L., D.S., L.L.)
| | - Chan Qu
- NCDs Prevention and Control Department, Liuyang CDC, China (C.Q.)
| | | | - Rory Collins
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, UK (R. Clarke, N.W., K.L., R.G.W., M.H., C.K., I.Y.M., D.A.B., D.A., L.Y., Y.C., H.D., P.S., X.Y., R. Collins, Z.C. S.P.)
| | - Zhengming Chen
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, UK (R. Clarke, N.W., K.L., R.G.W., M.H., C.K., I.Y.M., D.A.B., D.A., L.Y., Y.C., H.D., P.S., X.Y., R. Collins, Z.C. S.P.)
| | - Sarah Parish
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, UK (R. Clarke, N.W., K.L., R.G.W., M.H., C.K., I.Y.M., D.A.B., D.A., L.Y., Y.C., H.D., P.S., X.Y., R. Collins, Z.C. S.P.)
| |
Collapse
|
18
|
MacCarthy G, Pazoki R. Evaluation of Machine Learning and Traditional Statistical Models to Assess the Value of Stroke Genetic Liability for Prediction of Risk of Stroke Within the UK Biobank. Healthcare (Basel) 2025; 13:1003. [PMID: 40361781 PMCID: PMC12071721 DOI: 10.3390/healthcare13091003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 04/18/2025] [Accepted: 04/19/2025] [Indexed: 05/15/2025] Open
Abstract
Background and Objective: Stroke is one of the leading causes of mortality and long-term disability in adults over 18 years of age globally, and its increasing incidence has become a global public health concern. Accurate stroke prediction is highly valuable for early intervention and treatment. There is a scarcity of studies evaluating the prediction value of genetic liability in the prediction of the risk of stroke. Materials and Methods: Our study involved 243,339 participants of European ancestry from the UK Biobank. We created stroke genetic liability using data from MEGASTROKE genome-wide association studies (GWASs). In our study, we built four predictive models with and without stroke genetic liability in the training set, namely a Cox proportional hazard (Coxph) model, gradient boosting model (GBM), decision tree (DT), and random forest (RF), to estimate time-to-event risk for stroke. We then assessed their performances in the testing set. Results: Each unit (standard deviation) increase in genetic liability increases the risk of incident stroke by 7% (HR = 1.07, 95% CI = 1.02, 1.12, p-value = 0.0030). The risk of stroke was greater in the higher genetic liability group, demonstrated by a 14% increased risk (HR = 1.14, 95% CI = 1.02, 1.27, p-value = 0.02) compared with the low genetic liability group. The Coxph model including genetic liability was the best-performing model for stroke prediction achieving an AUC of 69.54 (95% CI = 67.40, 71.68), NRI of 0.202 (95% CI = 0.12, 0.28; p-value = 0.000) and IDI of 1.0 × 10-4 (95% CI = 0.000, 3.0 × 10-4; p-value = 0.13) compared with the Cox model without genetic liability. Conclusions: Incorporating genetic liability in prediction models slightly improved prediction models of stroke beyond conventional risk factors.
Collapse
Affiliation(s)
- Gideon MacCarthy
- Cardiovascular and Metabolic Research Group, Department of Biosciences, College of Health, Medicine, and Life Sciences, Brunel University of London, Uxbridge UB8 3PH, UK;
| | - Raha Pazoki
- Cardiovascular and Metabolic Research Group, Department of Biosciences, College of Health, Medicine, and Life Sciences, Brunel University of London, Uxbridge UB8 3PH, UK;
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London W2 1PG, UK
| |
Collapse
|
19
|
He W, Shi J, Qian Y, Fan T, Cai X, Li H, Huang P, Shi Q. Evidence to shared genetic correlation of ischemic stroke and intracerebral hemorrhage and cardiovascular related traits. PLoS One 2025; 20:e0320479. [PMID: 40267100 PMCID: PMC12017486 DOI: 10.1371/journal.pone.0320479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 02/20/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND Previous studies have demonstrated the genetic basis of stroke and also revealed their genetic correlation with some cardiovascular related diseases or traits at the entire genome, which, however, would not give the answer which regions may mainly account for the genetic overlap. This study aims to identify specific genetic loci that contribute to the shared genetic basis between ischemic stroke subtypes and common cardiovascular traits. METHODS We used Local Analysis of [co]Variant Annotation (LAVA), a recent developed local genetic correlation method, to perform a system local genetic correlation analysis on GWAS summary data of two major subtypes of stroke, including any ischemic stroke (AIS) and intracerebral hemorrhage (ICH), and ten common cardiovascular related diseases or traits (CRTs). We further used colocalization analysis to explore potential shared causal genes in loci with significant local genetic correlation. In addition, we also performed Transcriptome-wide association (TWAS) analysis and fine-mapping for each phenotype to functionally annotate significant loci. RESULTS LAVA analysis identified a total of 3 significant local genetic correlations (Bonferroni-adjusted P < 0.05) across 3 chromosomes between AIS and systolic blood pressure (SBP), AIS and hypertension (HT), and ICH and body mass index (BMI), among which locus 7.24 explicated to harbor a shared causal variant for AIS and SBP. TWIST1 in locus 7.24 was defined to be nominally associated with SBP, but not for AIS. Fine-mapping analysis also only identified TWIST1 a credible causal gene for BMI. CONCLUSIONS Our study revealed the local genetic correlations between two stroke subtypes and ten common CRTs. Gene-level analyses indicated that biological explanations underlying these identified local genetic correlations may existed elsewhere beyond a common pattern of genetic-gene expression regulation.
Collapse
Affiliation(s)
- Wei He
- Department of Physical Medicine and Rehabilitation, The Affiliated Jiangyin People’s Hospital of Southeast University Medical College, Wuxi, China
| | - Jiajia Shi
- Department of Physical Medicine and Rehabilitation, Kunshan Rehabilitation Hospital, Suzhou, China
| | - Yiming Qian
- Department of Physical Medicine and Rehabilitation, The Affiliated Jiangyin People’s Hospital of Southeast University Medical College, Wuxi, China
| | - Tao Fan
- Department of Neurology, Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
| | - Xuehong Cai
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Haochang Li
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Peng Huang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Qin Shi
- Department of Neurology, Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
| |
Collapse
|
20
|
Adkar SS, Lynch J, Choi RB, Roychowdhury T, Judy RL, Paruchuri K, Go DC, Bamezai S, Cabot J, Sorondo S, Levin MG, Milewicz DM, Willer CJ, Natarajan P, Pyarajan S, Chang KM, Damrauer S, Tsao P, Skirboll S, Leeper NJ, Klarin D. Dissecting the Genetic Architecture of Intracranial Aneurysms. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2025:e004626. [PMID: 40255156 DOI: 10.1161/circgen.123.004626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 04/03/2025] [Indexed: 04/22/2025]
Abstract
BACKGROUND The genetic risk of intracranial aneurysm (IA) development has been ascribed to the genetic risk of smoking exposure and hypertension. The relationship of IA to other cardiovascular traits and the contribution of IA risk loci to aberrant gene programs within cerebrovascular cell types remains unclear. METHODS We performed a genome-wide association study in the Million Veteran Program and Finnish cohort study testing association of roughly 25 million DNA variants with unruptured IA (4694 cases and 877 091 controls) in individuals of European, African, and Hispanic ancestries. Meta-analysis with publicly available summary statistics generated a final cohort of 15 438 cases and 1 183 973 controls. We constructed a cerebrovascular single-nuclear RNA sequencing data set and integrated IA summary statistics to prioritize candidate causal cell types. We constructed a polygenic risk score to identify patients at risk of developing IA. RESULTS We identified 5 novel associations with IA, increasing the number of known susceptibility loci to 22. At these susceptibility loci, we prioritized 17 candidate causal genes. We found a significant positive genetic correlation of IA with coronary artery disease and abdominal aortic aneurysm. Integration of an IA gene set with cerebrovascular single-nuclear RNA sequencing data revealed a significant association with pericytes and smooth muscle cells. Finally, a polygenic risk score was significantly associated with IA across European (odds ratio, 1.87 [95% CI, 1.61-2.17]; P=8.8×10-17), African (odds ratio, 1.62 [95% CI, 1.19-2.15]; P=1.2×10-3), and Hispanic (odds ratio, 2.28 [95% CI, 1.47-3.38]; P=1.0×10-4) ancestries. CONCLUSIONS Here, we identify 5 novel loci associated with IA. Integration of summary statistics with cerebrovascular single-nuclear RNA sequencing reveals an association of cell types involved in matrix production. We validated a polygenic risk score that predicts IA, controlling for demographic variables including smoking status and blood pressure. Our findings suggest that a deficit in matrix production may drive IA pathogenesis independent of hypertension and smoking.
Collapse
Affiliation(s)
- Shaunak S Adkar
- Division of Vascular Surgery, Department of Surgery, Stanford University School of Medicine, Palo Alto, CA. (S.S.A., J.C., S. Sorondo, N.J.L., D.K.)
- Stanford Cardiovascular Institute, Stanford University, CA (S.S.A., S.B., J.C., S. Sorondo. P.T., N.J.L.)
- Veterans Affairs (VA) Palo Alto Healthcare System, CA (S.S.A., J.C., S. Sorondo, P.T., S. Skirboll, D.K.)
| | - Julie Lynch
- VA Salt Lake City Healthcare System The University of Utah, Salt Lake City (J.L.)
- Epidemiology, School of Medicine, The University of Utah, Salt Lake City (J.L.)
| | - Ryan B Choi
- Stanford University School of Medicine, Palo Alto, CA (R.B.C.)
| | - Tanmoy Roychowdhury
- Department of Biology and Koita Centre for Digital Health, Trivedi School of Biosciences, Ashoka University, Sonepat, India (T.R.)
| | - Renae L Judy
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia. (R.L.J.)
- Research, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA. (R.L.J.)
| | - Kaavya Paruchuri
- Department of Medicine, Massachusetts General Hospital, Boston. (K.P., P.N.)
- Cardiovascular Research Center, Massachusetts General Hospital, Boston. (K.P., P.N.)
- Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA. (K.P., P.N.)
| | - Dong-Chuan Go
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, University of Texas Health Science Center at Houston (D.-C.G., D.M.M.)
| | - Sharika Bamezai
- Stanford Cardiovascular Institute, Stanford University, CA (S.S.A., S.B., J.C., S. Sorondo. P.T., N.J.L.)
- University of Michigan School of Medicine, Ann Arbor (S.B.)
| | - John Cabot
- Division of Vascular Surgery, Department of Surgery, Stanford University School of Medicine, Palo Alto, CA. (S.S.A., J.C., S. Sorondo, N.J.L., D.K.)
- Veterans Affairs (VA) Palo Alto Healthcare System, CA (S.S.A., J.C., S. Sorondo, P.T., S. Skirboll, D.K.)
| | - Sabina Sorondo
- Division of Vascular Surgery, Department of Surgery, Stanford University School of Medicine, Palo Alto, CA. (S.S.A., J.C., S. Sorondo, N.J.L., D.K.)
- Veterans Affairs (VA) Palo Alto Healthcare System, CA (S.S.A., J.C., S. Sorondo, P.T., S. Skirboll, D.K.)
| | - Michael G Levin
- Division of Cardiovascular Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia. (M.G.L.)
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia. (M.G.L.)
| | - Dianna M Milewicz
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, University of Texas Health Science Center at Houston (D.-C.G., D.M.M.)
| | - Cristen J Willer
- Division of Cardiology, Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor (C.J.W.)
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor. (C.J.W.)
- Department of Human Genetics, University of Michigan, Ann Arbor. (C.J.W.)
| | - Pradeep Natarajan
- Department of Medicine, Massachusetts General Hospital, Boston. (K.P., P.N.)
- Cardiovascular Research Center, Massachusetts General Hospital, Boston. (K.P., P.N.)
- Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA. (K.P., P.N.)
- Cardiovascular Disease Initiative, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA. (P.N.)
| | - Saiju Pyarajan
- Center for Data and Computational Sciences, VA Boston Health Care System, MA (S.P.)
- Research, Harvard Medical School, Boston, MA (S.P.)
| | - Kyong-Mi Chang
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia. (K.-M.C.)
- Research and Medicine, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA. (K.-M.C.)
| | - Scott Damrauer
- Division of Vascular Surgery and Endovascular Therapy, Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia. (S.D.)
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia. (S.D.)
- Penn Cardiovascular Institute, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA. (S.D.)
- Department of Surgery, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA. (S.D.)
| | - Phil Tsao
- Department of Medicine, Stanford University School of Medicine, Palo Alto, CA. (P.T., N.J.L.)
- Stanford Cardiovascular Institute, Stanford University, CA (S.S.A., S.B., J.C., S. Sorondo. P.T., N.J.L.)
- Veterans Affairs (VA) Palo Alto Healthcare System, CA (S.S.A., J.C., S. Sorondo, P.T., S. Skirboll, D.K.)
| | - Stephen Skirboll
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, CA. (S. Skirboll)
- Veterans Affairs (VA) Palo Alto Healthcare System, CA (S.S.A., J.C., S. Sorondo, P.T., S. Skirboll, D.K.)
| | - Nicholas J Leeper
- Division of Vascular Surgery, Department of Surgery, Stanford University School of Medicine, Palo Alto, CA. (S.S.A., J.C., S. Sorondo, N.J.L., D.K.)
- Department of Medicine, Stanford University School of Medicine, Palo Alto, CA. (P.T., N.J.L.)
- Stanford Cardiovascular Institute, Stanford University, CA (S.S.A., S.B., J.C., S. Sorondo. P.T., N.J.L.)
| | - Derek Klarin
- Division of Vascular Surgery, Department of Surgery, Stanford University School of Medicine, Palo Alto, CA. (S.S.A., J.C., S. Sorondo, N.J.L., D.K.)
- Veterans Affairs (VA) Palo Alto Healthcare System, CA (S.S.A., J.C., S. Sorondo, P.T., S. Skirboll, D.K.)
| |
Collapse
|
21
|
Wang H, Guo J, Zhang Y, Fu Z, Yao Y. Closed-loop rehabilitation of upper-limb dyskinesia after stroke: from natural motion to neuronal microfluidics. J Neuroeng Rehabil 2025; 22:87. [PMID: 40253334 PMCID: PMC12008995 DOI: 10.1186/s12984-025-01617-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 03/27/2025] [Indexed: 04/21/2025] Open
Abstract
This review proposes an innovative closed-loop rehabilitation strategy that integrates multiple subdomains of stroke science to address the global challenge of upper-limb dyskinesia post-stroke. Despite advancements in neural remodeling and rehabilitation research, the compartmentalization of subdomains has limited the effectiveness of current rehabilitation strategies. Our approach unites key areas-including the post-stroke brain, upper-limb rehabilitation robotics, motion sensing, metrics, neural microfluidics, and neuroelectronics-into a cohesive framework designed to enhance upper-limb motion rehabilitation outcomes. By leveraging cutting-edge technologies such as lightweight rehabilitation robotics, advanced motion sensing, and neural microfluidic models, this strategy enables real-time monitoring, adaptive interventions, and personalized rehabilitation plans. Furthermore, we explore the potential of closed-loop systems to drive neural plasticity and functional recovery, offering a transformative perspective on stroke rehabilitation. Finally, we discuss future directions, emphasizing the integration of emerging technologies and interdisciplinary collaboration to advance the field. This review highlights the promise of closed-loop strategies in achieving unprecedented integration of subdomains and improving post-stroke upper-limb rehabilitation outcomes.
Collapse
Affiliation(s)
- Honggang Wang
- State Key Laboratory of Robotics and System, Harbin Institute of Technology, Harbin, 150001, China
| | - Junlong Guo
- State Key Laboratory of Robotics and System, Harbin Institute of Technology, Harbin, 150001, China
| | - Yangqi Zhang
- State Key Laboratory of Robotics and System, Harbin Institute of Technology, Harbin, 150001, China
| | - Ze Fu
- Institute of Biological and Medical Technology, Harbin Institute of Technology (Weihai), Weihai, 264200, China
| | - Yufeng Yao
- State Key Laboratory of Robotics and System, Harbin Institute of Technology, Harbin, 150001, China.
| |
Collapse
|
22
|
Schmidt AF, Finan C, van Setten J, Puyol-Antón E, Ruijsink B, Bourfiss M, Alasiri AI, Velthuis BK, Asselbergs FW, Te Riele ASJM. A Mendelian randomization analysis of cardiac MRI measurements as surrogate outcomes for heart failure and atrial fibrillation. COMMUNICATIONS MEDICINE 2025; 5:130. [PMID: 40253538 PMCID: PMC12009341 DOI: 10.1038/s43856-025-00855-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 04/07/2025] [Indexed: 04/21/2025] Open
Abstract
BACKGROUND Drug development and disease prevention of heart failure (HF) and atrial fibrillation (AF) are impeded by a lack of robust early-stage surrogates. We determined to what extent cardiac magnetic resonance (CMR) measurements act as surrogates for the development of HF or AF. METHODS Genetic data were sourced on the association with 21 atrial and ventricular CMR measurements. Mendelian randomization was used to determine CMR associations with AF, HF, non-ischaemic cardiomyopathy (NICM), and dilated cardiomyopathy (DCM), noting that the definition of NICM includes DCM as a subset. Additionally, for the CMR surrogates of AF and HF, we explored their association with non-cardiac traits potentially influenced by cardiac disease liability. RESULTS In total we find that 7 CMR measures (biventricular ejection fraction (EF) and end-systolic volume (ESV), as well as LV systolic volume (SV), end-diastolic volume (EDV), and mass to volume ratio (MVR)) associate with the development of HF, 5 with the development of NICM (biventricular EDV and ESV, LV-EF), 7 with DCM (biventricular EF, ESV, EDV, and LV end-diastolic mass (EDM), and 3 associate with AF (LV-ESV, RV-EF, RV-ESV). Higher EF of both ventricles associate with lower risk of HF and DCM, with biventricular ESV associating with all four cardiac outcomes. Higher values of biventricular EDV associate with lower risk of HF, and DCM. Exploring the associations of these CMR cardiac disease surrogates with non-cardiac traits confirms a strong link with diastolic blood pressure, as well as more specific associations with lung function (LV-ESV), HbA1c (LV-EDM), and type 2 diabetes (LV-SV). CONCLUSIONS The current paper identifies key CMR measurements that may act as surrogate endpoints for the development of HF (including NICM and DCM) or AF.
Collapse
Affiliation(s)
- A F Schmidt
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London, UK.
- UCL BHF Research Accelerator Centre, London, UK.
- Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.
- Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam, The Netherlands.
| | - C Finan
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London, UK
- UCL BHF Research Accelerator Centre, London, UK
- Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - J van Setten
- Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - E Puyol-Antón
- Department of Biomedical Engineering, School of Biomedical Engineering and Imaging Sciences, King's College London, King's Health Partners, London, UK
| | - B Ruijsink
- School of Imaging Sciences and Biomedical Engineering, King's College London, London, UK
| | - M Bourfiss
- Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - A I Alasiri
- Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Medical Genomics Research Department, King Abdullah International Medical Research Center, King Saud Bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - B K Velthuis
- Department of Radiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - F W Asselbergs
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London, UK
- Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
- Institute of Health Informatics, Faculty of Population Health, University College London, London, UK
| | - A S J M Te Riele
- Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Netherlands Heart Institute, Utrecht, the Netherlands
| |
Collapse
|
23
|
Schunkert H, Di Angelantonio E, Inouye M, Patel RS, Ripatti S, Widen E, Sanderson SC, Kaski JP, McEvoy JW, Vardas P, Wood A, Aboyans V, Vassiliou VS, Visseren FLJ, Lopes LR, Elliott P, Kavousi M. Clinical utility and implementation of polygenic risk scores for predicting cardiovascular disease: A clinical consensus statement of the ESC Council on Cardiovascular Genomics, the ESC Cardiovascular Risk Collaboration, and the European Association of Preventive Cardiology. Eur Heart J 2025; 46:1372-1383. [PMID: 39906985 PMCID: PMC11997548 DOI: 10.1093/eurheartj/ehae649] [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: 02/06/2025] Open
Abstract
Genome-wide association studies have revealed hundreds of genetic variants associated with cardiovascular diseases (CVD). Polygenic risk scores (PRS) can capture this information in a single metric and hold promise for use in CVD risk prediction. Importantly, PRS information can reflect the causally mediated risk to which the individual is exposed throughout life. Although European Society of Cardiology guidelines do not currently advocate their use in routine clinical practice, PRS are commercially available and increasingly sought by clinicians, health systems, and members of the public to inform personalized health care decision-making. This clinical consensus statement provides an overview of the scientific basis of PRS and evidence to date on their role in CVD risk prediction for the purposes of disease prevention. It provides the reader with a summary of the opportunities and challenges for implementation and identifies current gaps in supporting evidence. The document also lays out a potential roadmap by which the scientific and clinical community can navigate any future transition of PRS into routine clinical care. Finally, clinical scenarios are presented where information from PRS may hold most value and discuss organizational frameworks to enable responsible use of PRS testing while more evidence is being generated by clinical studies.
Collapse
Affiliation(s)
- Heribert Schunkert
- Department of Cardiology, Deutsches Herzzentrum München, Universitätsklinikum der Technischen Universität München, 80636 Munich, Lazarettstrasse 36, Germany
- Deutsches Zentrum für Herz- und Kreislauferkrankungen (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Emanuele Di Angelantonio
- BHF 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
- BHF Centre of Research Excellence, School of Clinical Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
- NIHR Blood and Transplant Research Unit in Donor Health and Behaviour, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Hinxton, UK
- Health Data Science Centre, Human Technopole, Milan, Italy
| | - Michael Inouye
- BHF 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
- BHF Centre of Research Excellence, School of Clinical Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Hinxton, 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, Australia
| | - Riyaz S Patel
- Institute of Cardiovascular Sciences, University College London, London, UK
- Barts Heart Centre, St Bartholomew's Hospital, London, UK
- National Institute of Health Research Biomedical Research Centre, University College London Hospitals, London, UK
| | - Samuli Ripatti
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Finland
- Faculty of Medicine, University of Helsinki, Finland
- Massachusetts General Hospital & Broad Institute of MIT and Harvard, MA, USA
| | - Elisabeth Widen
- Institute for Molecular Medicine Finland FIMM, University of Helsinki, Helsinki, Finland
| | - Saskia C Sanderson
- Public Health Genomics (PHG) Foundation, Cambridge, UK
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Department of Behavioural Science and Health, University College London, London, UK
| | - Juan Pablo Kaski
- Centre for Paediatric Inherited and Rare Cardiovascular Disease, UCL Institute of Cardiovascular Science, London, and Centre for Inherited Cardiovascular Diseases, Great Ormond Street Hospital, London, UK
| | - John W McEvoy
- National Institute for Prevention and Cardiovascular Health, University of Galway School of Medicine, Galway, Ireland
| | - Panos Vardas
- University of Crete, Greece
- European Society of Cardiology Health Policy Unit, European Heart Health Institute, Brussels, Belgium
| | - Angela Wood
- BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- BHF Centre of Research Excellence, School of Clinical Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
- NIHR Blood and Transplant Research Unit in Donor Health and Behaviour, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Hinxton, UK
- Cambridge Centre of Artificial Intelligence in Medicine, University of Cambridge, Cambridge, UK
| | - Victor Aboyans
- Inserm U1094, IRD U270, Univ. Limoges, CHU Limoges, EpiMaCT—Epidemiology of Chronic Diseases in Tropical Zone, Institute of Epidemiology and Tropical Neurology, OmegaHealth, Limoges, France
- Department of Cardiology, Dupuytren-2 University Hospital Center, Limoges, France
| | - Vassilios S Vassiliou
- Department of Cardiology, Norwich Medical School, University of East Anglia and Norfolk and Norwich University Hospital, Norwich, UK
| | - Frank L J Visseren
- Department of Vascular Medicine, University Medical Centre Utrecht, The Netherlands
| | - Luis R Lopes
- Institute of Cardiovascular Sciences, University College London, London, UK
- Barts Heart Centre, St Bartholomew's Hospital, London, UK
| | - Perry Elliott
- Institute of Cardiovascular Sciences, University College London, London, UK
| | - Maryam Kavousi
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
24
|
Xie T, Pan Y, Lu K, Wei Y, Chen F, Tian Z, Wu P, Li Q, Wu B, Liu Y, Xue J, Bai J, Dong W, Liu Y, Shao J, Chen Y, Zhou X, Du Y, Liu Z, Gao S, Cheng Y, Huang R, Zhang Z, Yue Y, Zhong S, Deng Z, Zhou K, Jin J, Li C, Xu T, Zhou K. Cohort Profile: Kunshan Aging Research with E-health (KARE). Int J Epidemiol 2025; 54:dyaf041. [PMID: 40286341 DOI: 10.1093/ije/dyaf041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 03/21/2025] [Indexed: 04/29/2025] Open
Affiliation(s)
- Tian Xie
- Guangzhou National Laboratory, Guangzhou, Guangdong Province, China
| | - Ying Pan
- Affiliated Kunshan Hospital of Jiangsu University, Suzhou, Jiangsu Province, China
- Kunshan Biomedical Big Data Innovation Application Laboratory, Jiangsu, China
| | - Ke Lu
- Affiliated Kunshan Hospital of Jiangsu University, Suzhou, Jiangsu Province, China
- Kunshan Biomedical Big Data Innovation Application Laboratory, Jiangsu, China
| | - Yinlin Wei
- Centre for Health Statistics and Information, Health Commission of Kunshan, Suzhou, Jiangsu Province, China
| | - Fei Chen
- Department of Endocrinology, Peking University First Hospital, Beijing, China
| | - Zijian Tian
- National Laboratory of Biomacromolecules, Institute of Biophysics Chinese Academy of Sciences, Beijing, China
| | - Peng Wu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Qian Li
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Benrui Wu
- National Laboratory of Biomacromolecules, Institute of Biophysics Chinese Academy of Sciences, Beijing, China
| | - Yiying Liu
- National Laboratory of Biomacromolecules, Institute of Biophysics Chinese Academy of Sciences, Beijing, China
| | - Jingnan Xue
- Guangzhou National Laboratory, Guangzhou, Guangdong Province, China
| | - Jie Bai
- Guangzhou National Laboratory, Guangzhou, Guangdong Province, China
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Wanqing Dong
- Luoyang Key Laboratory of Clinical Multiomics and Translational Medicine, Henan Key Laboratory of Rare Diseases, Endocrinology and Metabolism Center, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Yang Liu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Jian Shao
- Guangzhou National Laboratory, Guangzhou, Guangdong Province, China
| | - Ying Chen
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Xiaozhou Zhou
- Guangzhou National Laboratory, Guangzhou, Guangdong Province, China
| | - Yuxuan Du
- Guangzhou National Laboratory, Guangzhou, Guangdong Province, China
| | - Ziqing Liu
- Guangzhou National Laboratory, Guangzhou, Guangdong Province, China
| | - Shiteng Gao
- Guangzhou National Laboratory, Guangzhou, Guangdong Province, China
- College of Public Health, Guangzhou Medical University, Guangzhou, China
| | - Yao Cheng
- Guangzhou National Laboratory, Guangzhou, Guangdong Province, China
| | - Rong Huang
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong Province, China
| | - Zhiqin Zhang
- Affiliated Kunshan Hospital of Jiangsu University, Suzhou, Jiangsu Province, China
| | - Yushan Yue
- Affiliated Kunshan Hospital of Jiangsu University, Suzhou, Jiangsu Province, China
- Kunshan Biomedical Big Data Innovation Application Laboratory, Jiangsu, China
| | - Shao Zhong
- Affiliated Kunshan Hospital of Jiangsu University, Suzhou, Jiangsu Province, China
| | - Zhiyong Deng
- Affiliated Kunshan Hospital of Jiangsu University, Suzhou, Jiangsu Province, China
| | - Kaiyun Zhou
- Affiliated Kunshan Hospital of Jiangsu University, Suzhou, Jiangsu Province, China
| | - Jian Jin
- Centre for Health Statistics and Information, Health Commission of Kunshan, Suzhou, Jiangsu Province, China
| | - Chong Li
- Affiliated Kunshan Hospital of Jiangsu University, Suzhou, Jiangsu Province, China
- Kunshan Biomedical Big Data Innovation Application Laboratory, Jiangsu, China
| | - Tao Xu
- Guangzhou National Laboratory, Guangzhou, Guangdong Province, China
- National Laboratory of Biomacromolecules, Institute of Biophysics Chinese Academy of Sciences, Beijing, China
| | - Kaixin Zhou
- Guangzhou National Laboratory, Guangzhou, Guangdong Province, China
- College of Public Health, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
25
|
Milani L, Alver M, Laur S, Reisberg S, Haller T, Aasmets O, Abner E, Alavere H, Allik A, Annilo T, Fischer K, Hofmeister R, Hudjashov G, Jõeloo M, Kals M, Karo-Astover L, Kasela S, Kolde A, Krebs K, Krigul KL, Kronberg J, Kruusmaa K, Kukuškina V, Kõiv K, Lehto K, Leitsalu L, Lind S, Luitva LB, Läll K, Lüll K, Metsalu K, Metspalu M, Mõttus R, Nelis M, Nikopensius T, Nurm M, Nõukas M, Oja M, Org E, Palover M, Palta P, Pankratov V, Pantiukh K, Pervjakova N, Pujol-Gualdo N, Reigo A, Reimann E, Smit S, Rogozina D, Särg D, Taba N, Talvik HA, Teder-Laving M, Tõnisson N, Vaht M, Vainik U, Võsa U, Yelmen B, Esko T, Kolde R, Mägi R, Vilo J, Laisk T, Metspalu A. The Estonian Biobank's journey from biobanking to personalized medicine. Nat Commun 2025; 16:3270. [PMID: 40188112 PMCID: PMC11972354 DOI: 10.1038/s41467-025-58465-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 03/04/2025] [Indexed: 04/07/2025] Open
Abstract
Large biobanks have set a new standard for research and innovation in human genomics and implementation of personalized medicine. The Estonian Biobank was founded a quarter of a century ago, and its biological specimens, clinical, health, omics, and lifestyle data have been included in over 800 publications to date. What makes the biobank unique internationally is its translational focus, with active efforts to conduct clinical studies based on genetic findings, and to explore the effects of return of results on participants. In this review, we provide an overview of the Estonian Biobank, highlight its strengths for studying the effects of genetic variation and quantitative phenotypes on health-related traits, development of methods and frameworks for bringing genomics into the clinic, and its role as a driving force for implementing personalized medicine on a national level and beyond.
Collapse
Affiliation(s)
- Lili Milani
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia.
- Estonian Biobank, Institute of Genomics, University of Tartu, Tartu, Estonia.
| | - Maris Alver
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Sven Laur
- Institute of Computer Science, University of Tartu, Tartu, Estonia
- STACC, Tartu, Estonia
| | - Sulev Reisberg
- Institute of Computer Science, University of Tartu, Tartu, Estonia
- STACC, Tartu, Estonia
| | - Toomas Haller
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Oliver Aasmets
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Erik Abner
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Helene Alavere
- Estonian Biobank, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Annely Allik
- Estonian Biobank, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Tarmo Annilo
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Krista Fischer
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
- Institute of Mathematics and Statistics, University of Tartu, Tartu, Estonia
| | - Robin Hofmeister
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
| | - Georgi Hudjashov
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
- Estonian Biocentre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Maarja Jõeloo
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Mart Kals
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Liis Karo-Astover
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Silva Kasela
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Anastassia Kolde
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
- Institute of Mathematics and Statistics, University of Tartu, Tartu, Estonia
| | - Kristi Krebs
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Kertu Liis Krigul
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Jaanika Kronberg
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Karoliina Kruusmaa
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Viktorija Kukuškina
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Kadri Kõiv
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Kelli Lehto
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Liis Leitsalu
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Sirje Lind
- Estonian Biobank, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Laura Birgit Luitva
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
- Institute of Mathematics and Statistics, University of Tartu, Tartu, Estonia
| | - Kristi Läll
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Kreete Lüll
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Kristjan Metsalu
- Estonian Biobank, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Mait Metspalu
- Estonian Biocentre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - René Mõttus
- Institute of Psychology, University of Tartu, Tartu, Estonia
- Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - Mari Nelis
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Tiit Nikopensius
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Miriam Nurm
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Margit Nõukas
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Marek Oja
- Institute of Computer Science, University of Tartu, Tartu, Estonia
| | - Elin Org
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Marili Palover
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Priit Palta
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Vasili Pankratov
- Centre for Genomics, Evolution and Medicine, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Kateryna Pantiukh
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Natalia Pervjakova
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Natàlia Pujol-Gualdo
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Anu Reigo
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Ene Reimann
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Steven Smit
- Estonian Biobank, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Diana Rogozina
- Estonian Biobank, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Dage Särg
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Nele Taba
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Harry-Anton Talvik
- Institute of Computer Science, University of Tartu, Tartu, Estonia
- STACC, Tartu, Estonia
| | - Maris Teder-Laving
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Neeme Tõnisson
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Mariliis Vaht
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Uku Vainik
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
- Institute of Psychology, University of Tartu, Tartu, Estonia
- Montreal Neurological Institute, McGill University, Montreal, Canada
| | - Urmo Võsa
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Burak Yelmen
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Tõnu Esko
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Raivo Kolde
- Institute of Computer Science, University of Tartu, Tartu, Estonia
| | - Reedik Mägi
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Jaak Vilo
- Institute of Computer Science, University of Tartu, Tartu, Estonia
- STACC, Tartu, Estonia
| | - Triin Laisk
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Andres Metspalu
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia.
| |
Collapse
|
26
|
Wei J, Zhao J, Yang K, Ma S, Zhang M, Sun Y, He C, Pan X, Zhu X. Metabolic Factors Mediate the Causal Effect of Physical Activity and Sedentary Behavior on Stroke and Its Subtypes: Evidence from Mendelian Randomization Study. Mol Neurobiol 2025:10.1007/s12035-025-04881-x. [PMID: 40172820 DOI: 10.1007/s12035-025-04881-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 03/21/2025] [Indexed: 04/04/2025]
Abstract
In observational studies, physical activity and sedentary behavior are associated with the risk of stroke (overall and stroke subtypes). However, whether it is causal relevance remains to be established. Given that exercise habits can be an intervention towards preventing strokes, we assessed the causality of behavioral habits with stroke and its subtypes using Mendelian randomization (MR) approaches. Stroke was analyzed as all-cause ischemic stroke (IS), three IS subtypes including large artery stroke, small vessel stroke and cardioembolic stroke, and intracerebral hemorrhage (ICH). Univariable MR analyses revealed an association between genetically predicted leisure screen time and higher overall IS risk (odds ratio (OR), 1.12; 95% confidence interval (CI), 1.06 to 1.19; P = 1.65 × 10-4). A protective association was also reported between genetically linked moderate-to-vigorous intensity physical activity and the risk of small vessel stroke (OR, 0.52; 95% CI, 0.32 to 0.84; P = 0.008). Further reverse analyses found no causal effect of IS on leisure screen time and small vessel stroke on moderate-to-vigorous intensity physical activity. We also studied the mediating role of metabolic traits including obesity, blood lipids, blood glucose, and blood pressure via two-step MR. It was found in mediation analysis that BMI partly mediated the causal relationship between leisure screen time and all-cause IS; the mediated proportion was 26.1% (95% CI: 18.1 to 35.0%). We found evidence that a sedentary lifestyle is associated with a higher risk of overall IS, and BMI plays a mediating role in the causal pathway. Our findings provide genetic evidence for the point that active lifestyles may be an effective prevention strategy for IS.
Collapse
Affiliation(s)
- Jin Wei
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Jie Zhao
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Kaiying Yang
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Shiyin Ma
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Meng Zhang
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Yu Sun
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Chang He
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Xudong Pan
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China.
| | - Xiaoyan Zhu
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China.
| |
Collapse
|
27
|
Li Y, Wang S, Liu L, Cai H, Huang Y, Gao M, Zhang X, Wu Q, Qiu G. (Apo)Lipoprotein Profiling with Multi-Omics Analysis Identified Medium-HDL-Targeting PSRC1 with Therapeutic Potential for Coronary Artery Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413491. [PMID: 39985383 PMCID: PMC12005818 DOI: 10.1002/advs.202413491] [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: 10/23/2024] [Revised: 02/05/2025] [Indexed: 02/24/2025]
Abstract
Identification of (apo)lipoprotein subclasses causally underpinning atherosclerosis may lead to identification of novel drug targets for treatment of atherosclerotic cardiovascular disease (ASCVD). In this study, observational and genetic associations between (apo)lipoprotein profile and carotid intima-media thickness-assessed atherosclerosis, and risks of coronary artery disease (CAD) and ischemic stroke (IS) are assessed, using data from the UK Biobank study, with further exploration of potential drug target for these two ASCVD subtypes through multi-omics analysis integrating genetic, transcriptomic, and proteomic data. Cholesteryl ester content in medium high-density lipoprotein causally protective of atherosclerosis is identified, plus a target gene, PSRC1, with therapeutic potential for CAD, but not IS, supported by consistent evidence from multi-omics layers of data, which also reveals that such therapeutic potential may be through downregulation of circulating proteins including TRP1, GRNs, and Pla2g12b, and upregulation of Neo1. The results provide strong evidence as well as mechanistic clues of PSRC1's therapeutic potential for CAD.
Collapse
Affiliation(s)
- Yingmei Li
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating)School of Public HealthTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Sihan Wang
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating)School of Public HealthTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Ling Liu
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating)School of Public HealthTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Hao Cai
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating)School of Public HealthTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Yacan Huang
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating)School of Public HealthTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Mingjing Gao
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating)School of Public HealthTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | | | - Qingqing Wu
- Department of CardiologyZhongnan Hospital of Wuhan UniversityWuhan430062China
- Institute of Myocardial Injury and RepairWuhan UniversityWuhan430062China
| | - Gaokun Qiu
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating)School of Public HealthTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| |
Collapse
|
28
|
Lin Y, Li X, Hu M, Zhao J, Zhu C. Reassessing the Association of Sedentary Behavior and Physical Activity with Ischemic Stroke: A Mendelian Randomization Study. Med Sci Sports Exerc 2025; 57:781-790. [PMID: 39809232 DOI: 10.1249/mss.0000000000003601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
PURPOSE Findings from previous Mendelian randomization (MR) studies disagreed with the current scientific consensus regarding the role of physical activity (PA) and sedentary behavior in ischemic stroke (IS). We reassessed these associations with a focus on etiological subtypes of IS and the potential mediating roles of cardiometabolic traits and brain imaging-derived phenotypes (IDPs). METHODS We performed MR analyses using summary statistics from genome-wide association studies of sedentary behavior and PA ( n = 88,411 ~ 608,595), cardiometabolic traits ( n = 393,193 ~ 694,649), brain IDPs ( n = 33,224), and the latest IS data (62,100 cases and 1,234,808 controls). Inverse-variance weighted regression was used as the primary method, complemented by several sensitivity analyses. A two-step MR approach was employed to assess the mediating effects of cardiometabolic traits and brain IDPs. RESULTS Genetic liability to leisure-time moderate-to-vigorous PA (MVPA) and higher overall PA (OPA) were associated with reduced risks of IS and small vessel stroke (Benjamini-Hochberg adjusted P < 0.05). Suggestive associations were observed between longer leisure-screen time and higher IS risk and between higher OPA and lower cardioembolic stroke risk ( P < 0.05). The isotropic volume fraction in the anterior limb of the left internal capsule, as well as some cardiometabolic metrics, partially mediated these associations. There was no evidence for causal effects of overall MVPA, overall light-intensity PA, or overall sedentary duration on IS. CONCLUSIONS Longer leisure screen time, less OPA, and not engaging in MVPA during leisure time were associated with higher risk of IS. The associations between PA and IS depended on different subtypes and were mediated by changes in anterior limb of the left internal capsule and cardiometabolic biomarkers.
Collapse
Affiliation(s)
- Yidie Lin
- Department of Epidemiology and Health Statistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, CHINA
| | - Xuechao Li
- Department of Epidemiology and Health Statistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, CHINA
| | - Meijing Hu
- Department of Epidemiology and Health Statistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, CHINA
| | | | - Cairong Zhu
- Department of Epidemiology and Health Statistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, CHINA
| |
Collapse
|
29
|
Xiao J, Li J, Wu X, Hao Y, Zhao X, Zhang W, Xu B, Ma T, Zhang L, Xiang R, Cui H, Yang C, Yan P, Tang M, Wang Y, Qu Y, Chen L, Liu Y, Zou Y, Zhang L, Liu Z, Yao Y, Yang C, Zhang B, Jiang X. Adult Height, Cardiovascular Disease, and the Underlying Mechanism: A Comprehensive Epidemiological and Genetic Analysis. Can J Cardiol 2025:S0828-282X(25)00237-5. [PMID: 40174860 DOI: 10.1016/j.cjca.2025.03.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 02/13/2025] [Accepted: 03/06/2025] [Indexed: 04/04/2025] Open
Abstract
BACKGROUND Adult height measures the complete growth of an individual and influences the development of cardiovascular disease (CVD). Despite recent within-sibling studies that have suggested minimal effects from environmental confounders, biological mechanisms underlying the height-CVD relationship remain elusive. METHODS Leveraging the large-scale UK Biobank data set and summary statistics from the latest genome-wide association studies, we reevaluated the effect of height on 8 major CVD subtypes. Phenotypic associations were determined using Cox proportional hazard analysis. Putative causal relationships were assessed using univariable Mendelian randomization. Mediation analysis and 2-step Mendelian randomization were further performed to investigate the mediation effect of 15 common cardiometabolic or pulmonary risk factors. RESULTS Height was consistently associated with a decreased risk of coronary artery disease (CAD), confirmed in epidemiological (hazard ratio, 0.90; 95% confidence interval [CI], 0.88-0.91) and genetic (odds ratio, 0.89, 95% CI, 0.86-0.92) analysis. Forced vital capacity was identified as the most significant mediator for the height-CAD relationship in epidemiological (proportion-mediated, 65.6%; 95% CI, 53.1%-78.0%) and genetic (proportion-mediated, 46.2%; 95% CI, 5.0%-87.5%) analysis. Notably, obesity, and blood pressure, lipid, and C-reactive protein levels also exhibited significant mediatory effects. Despite a consistent risk effect of height on atrial fibrillation and venous thromboembolism, no promising mediator was identified. CONCLUSIONS Our study confirms the health effects of height on CAD, atrial fibrillation, and venous thromboembolism and emphasizes forced vital capacity as the primary pathway that links height to CAD. Importantly, it indicates that the CAD risk associated with nonmodifiable height could be mitigated through enhanced lung function and cardiometabolic conditions.
Collapse
Affiliation(s)
- Jinyu Xiao
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Jiayuan Li
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Xueyao Wu
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Yu Hao
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Xunying Zhao
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Wenqiang Zhang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Bin Xu
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Tianpei Ma
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Li Zhang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Rong Xiang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Huijie Cui
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Chao Yang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Peijing Yan
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Mingshuang Tang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Yutong Wang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Yang Qu
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Lin Chen
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Yunjie Liu
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Yanqiu Zou
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Ling Zhang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China; Department of Iatrical Polymer Material and Artificial Apparatus, School of Polymer Science and Engineering, Sichuan University, Chengdu, China
| | - Zhenmi Liu
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China; Department of Maternal, Child and Adolescent Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Yuqin Yao
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China; Department of Occupational and Environmental Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Chunxia Yang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Ben Zhang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China; Department of Cardiology, Department of Neurology, and Department of Oncology, Hainan General Hospital and Hainan Affiliated Hospital, Hainan Medical University, Haikou, China.
| | - Xia Jiang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China; Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China; Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
30
|
Wang M, Collings PJ, Day FR, Ong KK, Brage S, Sharp SJ, Jang H, Suh S, Luo S, Au Yeung SL, Kim Y. Genetic Susceptibility to Type 2 Diabetes, Television Viewing, and Atherosclerotic Cardiovascular Disease Risk. J Am Heart Assoc 2025; 14:e036811. [PMID: 40071666 DOI: 10.1161/jaha.124.036811] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 01/06/2025] [Indexed: 03/19/2025]
Abstract
BACKGROUND Type 2 diabetes (T2D) is a major risk factor for atherosclerotic cardiovascular disease (ASCVD). This study examined the interplay between watching television and T2D genetic risk for risk of ASCVD. METHODS We included 346 916 White British individuals from UK Biobank. A weighted polygenic risk score for T2D was calculated on the basis of 138 genetic variants associated with T2D. Time spent watching television was self-reported and categorized into 2 groups: ≤1 h/d and ≥2 h/d. Over a median 13.8-year follow-up, 21 265 incident ASCVD events were identified. Models using Cox regression with age as the underlying time scale adjusted for potential confounders (demographic, anthropometric, lifestyle factors, and medication use) were fit. RESULTS Compared with watching television for ≤1 h/d, watching ≥2 h/d was associated with 12% (95% CI, 1.07-1.16) higher hazards of ASCVD, independently of T2D genetic risk. Joint analyses (with low T2D genetic risk and ≤1 h/d of television viewing as reference) indicated that medium and high T2D genetic risk was not associated with higher hazards of ASCVD as long as television viewing was ≤1 h/d. The P values for multiplicative and additive interactions between T2D genetic risk and television viewing were 0.050 and 0.038, respectively. The 10-year absolute risk of ASCVD was lower for high T2D genetic risk combined with ≤1 h/d of television viewing (2.13%) than for low T2D genetic risk combined with ≥2 h/d of television viewing (2.46%). CONCLUSIONS Future clinical trials of lifestyle-modification interventions targeting specific types of screen-based sedentary activities could be implemented to individuals at high genetic risk of T2D for primary prevention of ASCVD.
Collapse
Affiliation(s)
- Mengyao Wang
- School of Public Health The University of Hong Kong Li Ka Shing Faculty of Medicine Hong Kong SAR China
| | - Paul James Collings
- School of Public Health The University of Hong Kong Li Ka Shing Faculty of Medicine Hong Kong SAR China
| | - Felix R Day
- MRC Epidemiology Unit Institute of Metabolic Science, University of Cambridge Cambridge Cambridgeshire United Kingdom
| | - Ken K Ong
- MRC Epidemiology Unit Institute of Metabolic Science, University of Cambridge Cambridge Cambridgeshire United Kingdom
| | - Soren Brage
- MRC Epidemiology Unit Institute of Metabolic Science, University of Cambridge Cambridge Cambridgeshire United Kingdom
| | - Stephen J Sharp
- MRC Epidemiology Unit Institute of Metabolic Science, University of Cambridge Cambridge Cambridgeshire United Kingdom
| | - Haeyoon Jang
- School of Public Health The University of Hong Kong Li Ka Shing Faculty of Medicine Hong Kong SAR China
| | - Siyeon Suh
- School of Public Health The University of Hong Kong Li Ka Shing Faculty of Medicine Hong Kong SAR China
| | - Shan Luo
- School of Public Health The University of Hong Kong Li Ka Shing Faculty of Medicine Hong Kong SAR China
| | - Shiu Lun Au Yeung
- School of Public Health The University of Hong Kong Li Ka Shing Faculty of Medicine Hong Kong SAR China
| | - Youngwon Kim
- School of Public Health The University of Hong Kong Li Ka Shing Faculty of Medicine Hong Kong SAR China
- MRC Epidemiology Unit Institute of Metabolic Science, University of Cambridge Cambridge Cambridgeshire United Kingdom
| |
Collapse
|
31
|
Song Z, Han Y, Li W, Xu Y, He Y, Wang Y. A cross-tissue transcriptome-wide association study identifies new key genes in ischemic stroke. Gene 2025; 941:149207. [PMID: 39755263 DOI: 10.1016/j.gene.2024.149207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 12/20/2024] [Accepted: 12/31/2024] [Indexed: 01/06/2025]
Abstract
BACKGROUND Ischemic stroke (IS) is an important disease causing death and disability worldwide, and further investigation of IS-related genes through genome-wide association study (GWAS) data is valuable. METHODS The study included GWAS data from 62,100 IS patients of European origin and 1,234,808 controls in a cross-tissue transcriptome association study (TWAS). A joint analysis was first performed by the Unified Test for Molecular Markers (UTMOST) and FUSION methods. The results of the joint analysis were also validated by fine-mapping through FOCUS. Mendelian randomisation analysis was performed to determine whether the obtained genes were causally related to IS. Genome Annotated Multiple Marker Analysis (MAGMA) explored which biological functions the genes associated with IS. We used Coloc to co-localise GWAS and eQTL of the genes. We also biologically validated the results by Western blotting and immunofluorescence staining in the middle cerebral artery occlusion/reperfusion (MCAO/R) mouse model. RESULTS Four TWAS methods identified only one new susceptibility gene (USP38) associated with IS risk. Mendelian randomization and colocalization analysis found that USP38 may be protective against IS development. Functional enrichment analysis indicated IS-related genes were mainly associated with the intrinsic fibrinogen activation, acute myocardial infarction, exogenous fibrinogen activation, coagulation cascade response, TNF signalling pathway and GRB2 signalling pathway. Western blotting and immunofluorescence staining demonstrated a reduction in USP38 expression in MCAO/R mice. CONCLUSION Our research indicates that USP38 is an essential gene related to IS, with its expression strongly connected with IS risk, thus providing new perspectives on the genetic framework of IS.
Collapse
Affiliation(s)
- Zhiwei Song
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, China; Department of Neurology, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, China
| | - Yupeng Han
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, China; Department of Anesthesiology, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, China
| | - Wangyu Li
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, China; Department of Painology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Yiya Xu
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, China; Department of Neurology, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, China
| | - Yingchao He
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, China; Department of Neurology, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, China
| | - Yinzhou Wang
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, China; Department of Neurology, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, China; Fujian Key Laboratory of Medical Analysis, Fujian Academy of Medical Sciences, Fuzhou, Fujian, China.
| |
Collapse
|
32
|
Stamova B, Knepp B, Rodriguez F. Molecular heterogeneity in human stroke - What can we learn from the peripheral blood transcriptome? J Cereb Blood Flow Metab 2025:271678X251322598. [PMID: 40079561 PMCID: PMC11907527 DOI: 10.1177/0271678x251322598] [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] [Indexed: 03/15/2025]
Abstract
Stroke is a multifaceted disease with genetic and environmental components like diet and lifestyle. The central nervous and immune systems display complex interactions, with the peripheral immune response participating in brain injury and repair mechanisms following stroke. The bidirectional communication between the injured brain and peripheral blood presents an opportunity to investigate the molecular changes in the latter. There is substantial heterogeneity in stroke pathogenesis, pathophysiology, comorbidities, and response to treatment and outcome. This is captured and underscored by heterogeneity in the peripheral blood transcriptome. The current review highlights the role of the human peripheral blood transcriptome architecture for molecular phenotyping of different stroke etiologies and comorbidities, and for identifying underlying molecular correlates with clinically important variables and outcomes. Specific transcriptome features can potentially provide targets for clinical translation and for prioritizing genes and pathways for evaluation in experimental models. We also propose an approach to study the patient-specific transcriptional architecture and uncover the combinatorial heterogeneity in altered pathways in stroke patients that can also guide the search for treatment and prevention targets. Deciphering the molecular heterogeneity of stroke in a tissue that can be easily accessed and monitored, such as peripheral blood, may improve clinical trial success.
Collapse
Affiliation(s)
- Boryana Stamova
- Department of Neurology, School of Medicine, University of California at Davis, Sacramento, CA, USA
| | - Bodie Knepp
- Department of Neurology, School of Medicine, University of California at Davis, Sacramento, CA, USA
| | - Fernando Rodriguez
- Department of Neurology, School of Medicine, University of California at Davis, Sacramento, CA, USA
| |
Collapse
|
33
|
Asare Y, Yan G, Schlegl C, Prestel M, van der Vorst EPC, Teunissen AJP, Aronova A, Tosato F, Naser N, Caputo J, Prevot G, Azzun A, Wefers B, Wurst W, Schneider M, Forne I, Bidzhekov K, Naumann R, van der Laan SW, Brandhofer M, Cao J, Roth S, Malik R, Tiedt S, Mulder WJM, Imhof A, Liesz A, Weber C, Bernhagen J, Dichgans M. A cis-regulatory element controls expression of histone deacetylase 9 to fine-tune inflammasome-dependent chronic inflammation in atherosclerosis. Immunity 2025; 58:555-567.e9. [PMID: 39879983 DOI: 10.1016/j.immuni.2025.01.003] [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: 01/24/2024] [Revised: 09/03/2024] [Accepted: 01/03/2025] [Indexed: 01/31/2025]
Abstract
Common genetic variants in a conserved cis-regulatory element (CRE) at histone deacetylase (HDAC)9 are a major risk factor for cardiovascular disease, including stroke and coronary artery disease. Given the consistency of this association and its proinflammatory properties, we examined the mechanisms whereby HDAC9 regulates vascular inflammation. HDAC9 bound and mediated deacetylation of NLRP3 in the NACHT and LRR domains leading to inflammasome activation and lytic cell death. Targeted deletion of the critical CRE in mice increased Hdac9 expression in myeloid cells to exacerbate inflammasome-dependent chronic inflammation. In human carotid endarterectomy samples, increased HDAC9 expression was associated with atheroprogression and clinical plaque instability. Incorporation of TMP195, a class IIa HDAC inhibitor, into lipoprotein-based nanoparticles to target HDAC9 at the site of myeloid-driven vascular inflammation stabilized atherosclerotic plaques, implying a lower risk of plaque rupture and cardiovascular events. Our findings link HDAC9 to atherogenic inflammation and provide a paradigm for anti-inflammatory therapeutics for atherosclerosis.
Collapse
Affiliation(s)
- Yaw Asare
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilian-University (LMU), Munich, Germany.
| | - Guangyao Yan
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilian-University (LMU), Munich, Germany
| | - Christina Schlegl
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilian-University (LMU), Munich, Germany
| | - Matthias Prestel
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilian-University (LMU), Munich, Germany
| | - Emiel P C van der Vorst
- Institute for Cardiovascular Prevention (IPEK), LMU, Munich, Germany; Institute for Molecular Cardiovascular Research (IMCAR), Aachen-Maastricht Institute for CardioRenal Disease (AMICARE) & Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, Aachen, Germany
| | - Abraham J P Teunissen
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Arailym Aronova
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilian-University (LMU), Munich, Germany
| | - Federica Tosato
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilian-University (LMU), Munich, Germany
| | - Nawraa Naser
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilian-University (LMU), Munich, Germany
| | - Julio Caputo
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilian-University (LMU), Munich, Germany
| | - Geoffrey Prevot
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anthony Azzun
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Benedikt Wefers
- Deutsches Zentrum für Neurodegenerative Erkrankungen e. V. (DZNE), Munich, Germany
| | - Wolfgang Wurst
- Deutsches Zentrum für Neurodegenerative Erkrankungen e. V. (DZNE), Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Melanie Schneider
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilian-University (LMU), Munich, Germany
| | - Ignasi Forne
- Protein Analysis Unit, Faculty of Medicine, Biomedical Center, LMU, Martinsried, Germany
| | - Kiril Bidzhekov
- Institute for Cardiovascular Prevention (IPEK), LMU, Munich, Germany
| | - Ronald Naumann
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Sander W van der Laan
- Central Diagnostics Laboratory, Division of Laboratory, Pharmacy, and Biomedical Genetics, University Medical Center Utrecht, University of Utrecht, Utrecht, the Netherlands
| | - Markus Brandhofer
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilian-University (LMU), Munich, Germany
| | - Jiayu Cao
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilian-University (LMU), Munich, Germany
| | - Stefan Roth
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilian-University (LMU), Munich, Germany
| | - Rainer Malik
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilian-University (LMU), Munich, Germany
| | - Steffen Tiedt
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilian-University (LMU), Munich, Germany
| | - Willem J M Mulder
- Department of Internal Medicine, Radboud Institute of Molecular Life Sciences (RIMLS) and Radboud Center for Infectious Diseases (RCI), Radboud University Nijmegen Medical Center, Laboratory of Chemical Biology, Nijmegen, the Netherlands
| | - Axel Imhof
- Protein Analysis Unit, Faculty of Medicine, Biomedical Center, LMU, Martinsried, Germany
| | - Arthur Liesz
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilian-University (LMU), Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), LMU, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance (MHA), Munich, Germany; Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Jürgen Bernhagen
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilian-University (LMU), Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance (MHA), Munich, Germany
| | - Martin Dichgans
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilian-University (LMU), Munich, Germany; Deutsches Zentrum für Neurodegenerative Erkrankungen e. V. (DZNE), Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance (MHA), Munich, Germany.
| |
Collapse
|
34
|
Bortnick AE, Austin TR, Hamerton E, Gudmundsdottir V, Emilsson V, Jennings LL, Gudnason V, Owens DS, Massera D, Dufresne L, Yang TY, Engert JC, Thanassoulis G, Tracy RP, Gerszten RE, Psaty BM, Kizer JR. Plasma Proteomic Assessment of Calcific Aortic Valve Disease in Older Adults. J Am Heart Assoc 2025; 14:e036336. [PMID: 40008515 DOI: 10.1161/jaha.124.036336] [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: 09/11/2024] [Accepted: 01/02/2025] [Indexed: 02/27/2025]
Abstract
BACKGROUND Calcific aortic valve disease (CAVD), and ensuing severe aortic stenosis (AS), is the foremost valvular disorder of aging, yet preventive therapies are lacking. A better understanding of the molecular underpinnings of aortic valve calcification (AVC) is necessary to develop pharmacologic interventions. METHODS AND RESULTS We undertook large-scale plasma proteomics in a cohort study of adults ≥65 years old, the CHS (Cardiovascular Health Study), to identify individual proteins associated with echocardiographic AVC and incident moderate/severe AS. Proteomics measurements were performed with the aptamer-based SomaLogic platform of ~5000 proteins. Significant proteins were validated in a second cohort, the AGES-RS (Age, Gene/Environment Susceptibility-Reykjavik Study), which assessed AVC and AS by computed tomography. The potential causal associations of replicated proteins were tested in 2-sample Mendelian randomization using identified cis protein quantitative trait loci in consortia having computed tomography-quantified AVC or AS as outcomes. Six proteins showed Bonferroni-corrected significant relationships with AVC in CHS. Three of these, CXCL-12 (C-X-C chemokine ligand 12), KLKB1 (kallikrein), and leptin, replicated in AGES-RS, of which the former 2 are novel. Only 1 protein, CXCL6, which showed a near-significant association with AS in the replication cohort, was significantly (positively) associated with incident AS. Mendelian randomization analysis was conducted for KLKB1, CXCL12, and CXCL6, which supported a causal relationship for higher KLKB1 with lower AVC (beta=-0.25, P=0.009). CONCLUSIONS This study of older adults newly identified and largely replicated associations of 3 circulating proteins with calcific aortic valve disease, of which the relationship of plasma KLKB1 may have a causal basis. Additional investigation is necessary to determine if KLKB1 could be harnessed for calcific aortic valve disease therapeutics.
Collapse
Affiliation(s)
- Anna E Bortnick
- Department of Medicine, Divisions of Cardiology and Geriatrics Montefiore Medical Center and Albert Einstein College of Medicine Bronx NY
- Department of Obstetrics and Gynecology and Women's Health Montefiore Medical Center and Albert Einstein College of Medicine Bronx NY
| | - Thomas R Austin
- Cardiovascular Health Research Unit, Department of Epidemiology University of Washington Seattle WA
| | - Emily Hamerton
- Department of Medicine University of California San Francisco San Francisco CA
- Cardiology Section San Francisco Veterans Affairs Health Care System San Francisco CA
| | - Valborg Gudmundsdottir
- Faculty of Medicine University of Iceland Reykjavik Iceland
- Icelandic Heart Association Kopavogur Iceland
| | | | | | - Vilmundur Gudnason
- Faculty of Medicine University of Iceland Reykjavik Iceland
- Icelandic Heart Association Kopavogur Iceland
| | - David S Owens
- Division of Cardiology University of Washington Seattle WA
| | - Daniele Massera
- Leon H. Charney Division of Cardiology New York University Langone Health New York NY
| | - Line Dufresne
- Preventive and Genomic Cardiology McGill University Health Centre Research Institute Montreal Quebec Canada
| | - Ta-Yu Yang
- Preventive and Genomic Cardiology McGill University Health Centre Research Institute Montreal Quebec Canada
- Department of Human Genetics McGill University Montreal Quebec Canada
| | - James C Engert
- Preventive and Genomic Cardiology McGill University Health Centre Research Institute Montreal Quebec Canada
- Department of Human Genetics McGill University Montreal Quebec Canada
- Division of Experimental Medicine McGill University Montreal Quebec Canada
| | - George Thanassoulis
- Preventive and Genomic Cardiology McGill University Health Centre Research Institute Montreal Quebec Canada
- Division of Experimental Medicine McGill University Montreal Quebec Canada
| | - Russell P Tracy
- Department of Pathology and Laboratory Medicine Larner College of Medicine, University of Vermont Burlington VT
| | - Robert E Gerszten
- Department of Medicine, Division of Cardiology Beth Israel Deaconess Hospital and Harvard Medical School Boston MA
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Departments of Medicine, Epidemiology, and Health Systems and Population Health University of Washington Seattle WA
| | - Jorge R Kizer
- Department of Medicine University of California San Francisco San Francisco CA
- Cardiology Section San Francisco Veterans Affairs Health Care System San Francisco CA
- Department of Epidemiology and Biostatistics University of California San Francisco San Francisco CA
| |
Collapse
|
35
|
Saks DG, Sachdev PS. Monogenic causes of cerebral small vessel disease- models for vascular cognitive impairment and dementia? Curr Opin Psychiatry 2025; 38:112-118. [PMID: 39840612 PMCID: PMC11789596 DOI: 10.1097/yco.0000000000000978] [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] [Indexed: 01/23/2025]
Abstract
PURPOSE OF REVIEW Recent advancements in molecular biomarkers and therapeutic options for Alzheimer's disease have brought into focus the need for greater progress in the second most common cause of dementia, vascular cognitive impairment and dementia (VCID). We examine how the study of monogenic causes of VCID has contributed to the understanding of its pathophysiology and potential biomarker and treatment research. RECENT FINDINGS It is widely accepted that conditions which disrupt the cerebral small vessels contribute to vascular pathologies including stroke and cerebral microbleeds, ultimately leading to vascular cognitive impairment and dementia. Among these conditions are a range of monogenic small vessel diseases (SVDs) such as CADASIL, CARASIL, Fabry disease and COL4A-related disorders. SUMMARY This review indicates the importance of furthering research into monogenic SVDs in order to gain insight into the pathomechanisms of VCID more broadly. Monogenic conditions are easier to model than sporadic VCID and can serve as a guide for identifying biomarkers for diagnosis, monitoring and intervention outcomes.
Collapse
Affiliation(s)
- Danit G. Saks
- Centre for Healthy Brain Ageing, University of New South Wales
| | - Perminder S. Sachdev
- Centre for Healthy Brain Ageing, University of New South Wales
- Neuropsychiatric Institute, Prince of Wales Hospital, Sydney, Australia
| |
Collapse
|
36
|
Wei X, He Y, Yu Y, Tang S, Liu R, Guo J, Jiang Q, Zhi X, Wang X, Meng D. The Multifaceted Roles of BACH1 in Disease: Implications for Biological Functions and Therapeutic Applications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412850. [PMID: 39887888 PMCID: PMC11905017 DOI: 10.1002/advs.202412850] [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: 10/13/2024] [Revised: 12/22/2024] [Indexed: 02/01/2025]
Abstract
BTB domain and CNC homolog 1 (BACH1) belongs to the family of basic leucine zipper proteins and is expressed in most mammalian tissues. It can regulate its own expression and play a role in transcriptionally activating or inhibiting downstream target genes. It has a crucial role in various biological processes, such as oxidative stress, cell cycle, heme homeostasis, and immune regulation. Recent research highlights BACH1's significant regulatory roles in a series of conditions, including stem cell pluripotency maintenance and differentiation, growth, senescence, and apoptosis. BACH1 is closely associated with cardiovascular diseases and contributes to angiogenesis, atherosclerosis, restenosis, pathological cardiac hypertrophy, myocardial infarction, and ischemia/reperfusion (I/R) injury. BACH1 promotes tumor cell proliferation and metastasis by altering tumor metabolism and the epithelial-mesenchymal transition phenotype. Moreover, BACH1 appears to show an adverse role in diseases such as neurodegenerative diseases, gastrointestinal disorders, leukemia, pulmonary fibrosis, and skin diseases. Inhibiting BACH1 may be beneficial for treating these diseases. This review summarizes the role of BACH1 and its regulatory mechanism in different cell types and diseases, proposing that precise targeted intervention of BACH1 may provide new strategies for human disease prevention and treatment.
Collapse
Affiliation(s)
- Xiangxiang Wei
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesDepartment of RheumatologyZhongshan HospitalZhongshan Hospital Immunotherapy Translational Research CenterFudan UniversityShanghai200032China
| | - Yunquan He
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesDepartment of RheumatologyZhongshan HospitalZhongshan Hospital Immunotherapy Translational Research CenterFudan UniversityShanghai200032China
| | - Yueyang Yu
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesDepartment of RheumatologyZhongshan HospitalZhongshan Hospital Immunotherapy Translational Research CenterFudan UniversityShanghai200032China
| | - Sichong Tang
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesDepartment of RheumatologyZhongshan HospitalZhongshan Hospital Immunotherapy Translational Research CenterFudan UniversityShanghai200032China
| | - Ruiwen Liu
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesDepartment of RheumatologyZhongshan HospitalZhongshan Hospital Immunotherapy Translational Research CenterFudan UniversityShanghai200032China
| | - Jieyu Guo
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesDepartment of RheumatologyZhongshan HospitalZhongshan Hospital Immunotherapy Translational Research CenterFudan UniversityShanghai200032China
| | - Qingjun Jiang
- Department of Vascular & Endovascular SurgeryChangzheng HospitalNaval Medical UniversityShanghai200003China
| | - Xiuling Zhi
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesDepartment of RheumatologyZhongshan HospitalZhongshan Hospital Immunotherapy Translational Research CenterFudan UniversityShanghai200032China
| | - Xinhong Wang
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesDepartment of RheumatologyZhongshan HospitalZhongshan Hospital Immunotherapy Translational Research CenterFudan UniversityShanghai200032China
| | - Dan Meng
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesDepartment of RheumatologyZhongshan HospitalZhongshan Hospital Immunotherapy Translational Research CenterFudan UniversityShanghai200032China
| |
Collapse
|
37
|
Lin S, Li YE, Wang Y. Multi-Cohort Analysis Reveals Genetic Predispositions to Clonal Hematopoiesis as Mutation-Specific Risk Factors for Stroke. ADVANCED GENETICS (HOBOKEN, N.J.) 2025; 6:2400047. [PMID: 40093911 PMCID: PMC11909397 DOI: 10.1002/ggn2.202400047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 01/17/2025] [Indexed: 03/19/2025]
Abstract
Recent observational studies have found an association between Clonal Hematopoesis (CH) and strokes but with incomplete results. This study aims to comprehensively characterize mutation-specific effects of CH on ischemic and hemorrhagic stroke subtypes and 90-day functional outcomes through publicly available genome-wide association study (GWAS) cohorts and Mendelian Randomization. TET2 is associated with an increased risk of overall stroke (OR = 1.06, P = 0.02), ischemic stroke (OR = 1.05, P = 0.03), transient ischemic attack (OR = 1.07, P = 0.01) and small vessel stroke (OR = 1.29, P = 0.01), as well as adverse 90-day modified Rankin scale (mRS ≥ 3) before (OR = 1.34, P = 0.005) and after adjusted for age, sex, and stroke severity (OR = 1.30, P = 0.02). While the presence of any CH mutation is associated with intracerebral hemorrhage (ICH) (OR = 1.21, P = 0.02), specific mutations, SRSF2 and ASXL1 are protective against ICH (OR = 0.9, P = 0.04) and nontraumatic subarachnoid hemorrhage (OR = 0.92, P = 0.03), respectively. In conclusion, the study provided genetic evidence that TET2 is strongly associated with an increased risk of ischemic stroke and poor functional recovery. Future studies clarifying the relationship between CH and hemorrhagic stroke subtypes are needed.
Collapse
Affiliation(s)
- Shuyang Lin
- Department of HematologyWashington University School of Medicine in St LouisSt. LouisMO63110USA
- Department of GeneticsWashington University School of Medicine in St LouisSt. LouisMO63110USA
| | - Yang E. Li
- Department of GeneticsWashington University School of Medicine in St LouisSt. LouisMO63110USA
- Department of NeurosurgeryWashington University School of Medicine in St LouisSt. LouisMO63110USA
| | - Yan Wang
- Department of NeurologyWashington University School of Medicine in St LouisSt. LouisMO63110USA
| |
Collapse
|
38
|
Boudriot E, Stephan M, Rabe F, Smigielski L, Schmitt A, Falkai P, Ziller MJ, Rossner MJ, Homan P, Papiol S, Raabe FJ. Genetic Analysis of Retinal Cell Types in Neuropsychiatric Disorders. JAMA Psychiatry 2025; 82:285-295. [PMID: 39775833 PMCID: PMC11883512 DOI: 10.1001/jamapsychiatry.2024.4230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 10/29/2024] [Indexed: 01/11/2025]
Abstract
Importance As an accessible part of the central nervous system, the retina provides a unique window to study pathophysiological mechanisms of brain disorders in humans. Imaging and electrophysiological studies have revealed retinal alterations across several neuropsychiatric and neurological disorders, but it remains largely unclear which specific cell types and biological mechanisms are involved. Objective To determine whether specific retinal cell types are affected by genomic risk for neuropsychiatric and neurological disorders and to explore the mechanisms through which genomic risk converges in these cell types. Design, Setting, and Participants This genetic association study combined findings from genome-wide association studies in schizophrenia, bipolar disorder, major depressive disorder, multiple sclerosis, Parkinson disease, Alzheimer disease, and stroke with retinal single-cell transcriptomic datasets from humans, macaques, and mice. To identify susceptible cell types, Multi-Marker Analysis of Genomic Annotation (MAGMA) cell-type enrichment analyses were applied and subsequent pathway analyses performed. The cellular top hits were translated to the structural level using retinal optical coherence tomography (acquired between 2009 and 2010) and genotyping data in the large population-based UK Biobank cohort study. Data analysis was conducted between 2022 and 2024. Main Outcomes and Measures Cell type-specific enrichment of genetic risk loading for neuropsychiatric and neurological disorder traits in the gene expression profiles of retinal cells. Results Expression profiles of amacrine cells (interneurons within the retina) were robustly enriched in schizophrenia genetic risk across mammalian species and in different developmental stages. This enrichment was primarily driven by genes involved in synapse biology. Moreover, expression profiles of retinal immune cell populations were enriched in multiple sclerosis genetic risk. No consistent cell-type associations were found for bipolar disorder, major depressive disorder, Parkinson disease, Alzheimer disease, or stroke. On the structural level, higher polygenic risk for schizophrenia was associated with thinning of the ganglion cell inner plexiform layer, which contains dendrites and synaptic connections of amacrine cells (B, -0.09; 95% CI, -0.16 to -0.03; P = .007; n = 36 349; mean [SD] age, 57.50 [8.00] years; 19 859 female [54.63%]). Higher polygenic risk for multiple sclerosis was associated with increased thickness of the retinal nerve fiber layer (B, 0.06; 95% CI, 0.02 to 0.10; P = .007; n = 36 371; mean [SD] age, 57.51 [8.00] years; 19 843 female [54.56%]). Conclusions and Relevance This study provides novel insights into the cellular underpinnings of retinal alterations in neuropsychiatric and neurological disorders and highlights the retina as a potential proxy to study synaptic pathology in schizophrenia.
Collapse
Affiliation(s)
- Emanuel Boudriot
- Max Planck Institute of Psychiatry, Munich, Germany
- Department of Psychiatry and Psychotherapy, Ludwig Maximilian University Hospital, Ludwig Maximilian University Munich, Munich, Germany
| | - Marius Stephan
- Department of Psychiatry and Psychotherapy, Ludwig Maximilian University Hospital, Ludwig Maximilian University Munich, Munich, Germany
- Systasy Bioscience, Munich, Germany
| | - Finn Rabe
- Department of Adult Psychiatry and Psychotherapy, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Lukasz Smigielski
- Department of Child and Adolescent Psychiatry and Psychotherapy, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Andrea Schmitt
- Max Planck Institute of Psychiatry, Munich, Germany
- Department of Psychiatry and Psychotherapy, Ludwig Maximilian University Hospital, Ludwig Maximilian University Munich, Munich, Germany
- German Center for Mental Health, Partner Site Munich-Augsburg, Germany
- Laboratory of Neurosciences, Institute of Psychiatry, University of São Paulo, São Paulo, Brazil
| | - Peter Falkai
- Max Planck Institute of Psychiatry, Munich, Germany
- Department of Psychiatry and Psychotherapy, Ludwig Maximilian University Hospital, Ludwig Maximilian University Munich, Munich, Germany
- German Center for Mental Health, Partner Site Munich-Augsburg, Germany
| | - Michael J. Ziller
- Max Planck Institute of Psychiatry, Munich, Germany
- Department of Psychiatry, University of Münster, Münster, Germany
- Center for Soft Nanoscience, University of Münster, Münster, Germany
| | - Moritz J. Rossner
- Department of Psychiatry and Psychotherapy, Ludwig Maximilian University Hospital, Ludwig Maximilian University Munich, Munich, Germany
- Systasy Bioscience, Munich, Germany
| | - Philipp Homan
- Department of Adult Psychiatry and Psychotherapy, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and Eidgenössische Technische Hochschule (ETH) Zurich, Zurich, Switzerland
| | - Sergi Papiol
- Max Planck Institute of Psychiatry, Munich, Germany
- Institute of Psychiatric Phenomics and Genomics, Ludwig Maximilian University Munich, Munich, Germany
| | - Florian J. Raabe
- Max Planck Institute of Psychiatry, Munich, Germany
- Department of Psychiatry and Psychotherapy, Ludwig Maximilian University Hospital, Ludwig Maximilian University Munich, Munich, Germany
| |
Collapse
|
39
|
Sutoh Y, Hachiya T, Otsuka-Yamasaki Y, Tokutomi T, Yoshida A, Kotozaki Y, Komaki S, Minabe S, Ohmomo H, Tanno K, Fukushima A, Sasaki M, Shimizu A. Reference-Based Standardization Approach Stabilizing Small Batch Risk Prediction via Polygenic Score. Genet Epidemiol 2025; 49:e70002. [PMID: 39888077 DOI: 10.1002/gepi.70002] [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: 08/29/2024] [Revised: 12/19/2024] [Accepted: 12/20/2024] [Indexed: 02/01/2025]
Abstract
The polygenic score (PGS) holds promise for motivating preventive behavioral changes. However, no clinically validated standardization methodology currently exists. Here, we demonstrate the efficacy of a "reference-based" approach for standardization. This method uses the PGS distribution in the general population as a reference for normalization and percentile determination; however, it has not been validated. We investigated three potential influences on PGS computation: (1) the size of the reference population, (2) biases associated with different genotyping platforms, and (3) inclusion of kinship ties within the reference group. Our results indicate that the reference size affects the bootstrap estimate of standard error for PGS percentiles, peaking around the 50th percentile and diminishing at extreme percentiles (1st or 100th). Discrepancies between genotyping platforms, such as different microarrays and whole-genome sequencing, resulted in deviations in PGS (p < 0.05 in Kolmogorov-Smirnov test). However, these deviations were reduced to a nonsignificant level using shared genetic variants in the calculations when the ancestry of the samples and reference were matched. This approach recovered approximately 9.6% of the positive predictive value of PGS by naïve genotype. Our results provide fundamental insights for establishing clinical guidelines for implementing PGS to communicate reliable risks to individuals.
Collapse
Affiliation(s)
- Yoichi Sutoh
- Division of Biomedical Information Analysis, Iwate Tohoku Medical Megabank Organization, Disaster Reconstruction Center, Iwate Medical University, Yahaba, Japan
- Division of Biomedical Information Analysis, Institute for Biomedical Sciences, Iwate Medical University, Yahaba, Japan
| | - Tsuyoshi Hachiya
- Division of Biomedical Information Analysis, Iwate Tohoku Medical Megabank Organization, Disaster Reconstruction Center, Iwate Medical University, Yahaba, Japan
- Division of Biomedical Information Analysis, Institute for Biomedical Sciences, Iwate Medical University, Yahaba, Japan
| | - Yayoi Otsuka-Yamasaki
- Division of Biomedical Information Analysis, Iwate Tohoku Medical Megabank Organization, Disaster Reconstruction Center, Iwate Medical University, Yahaba, Japan
- Division of Biomedical Information Analysis, Institute for Biomedical Sciences, Iwate Medical University, Yahaba, Japan
| | - Tomoharu Tokutomi
- Division of Innovation & Education, Iwate Tohoku Medical Megabank Organization, Disaster Reconstruction Center, Iwate Medical University, Yahaba, Japan
- Department of Clinical Genetics, School of Medicine, Iwate Medical University, Morioka, Japan
- Department of Pediatrics, Kawasaki Medical School, Kurashiki, Japan
| | - Akiko Yoshida
- Division of Innovation & Education, Iwate Tohoku Medical Megabank Organization, Disaster Reconstruction Center, Iwate Medical University, Yahaba, Japan
- Department of Clinical Genetics, School of Medicine, Iwate Medical University, Morioka, Japan
| | - Yuka Kotozaki
- Division of Clinical Research & Epidemiology, Iwate Tohoku Medical Megabank Organization, Disaster Reconstruction Center, Iwate Medical University, Yahaba, Japan
- Department of Hygiene and Preventive Medicine, School of Medicine, Iwate Medical University, Iwate, Japan
| | - Shohei Komaki
- Division of Biomedical Information Analysis, Iwate Tohoku Medical Megabank Organization, Disaster Reconstruction Center, Iwate Medical University, Yahaba, Japan
- Division of Biomedical Information Analysis, Institute for Biomedical Sciences, Iwate Medical University, Yahaba, Japan
| | - Shiori Minabe
- Division of Biomedical Information Analysis, Iwate Tohoku Medical Megabank Organization, Disaster Reconstruction Center, Iwate Medical University, Yahaba, Japan
- Division of Biomedical Information Analysis, Institute for Biomedical Sciences, Iwate Medical University, Yahaba, Japan
| | - Hideki Ohmomo
- Division of Biomedical Information Analysis, Iwate Tohoku Medical Megabank Organization, Disaster Reconstruction Center, Iwate Medical University, Yahaba, Japan
- Division of Biomedical Information Analysis, Institute for Biomedical Sciences, Iwate Medical University, Yahaba, Japan
| | - Kozo Tanno
- Division of Clinical Research & Epidemiology, Iwate Tohoku Medical Megabank Organization, Disaster Reconstruction Center, Iwate Medical University, Yahaba, Japan
- Department of Hygiene and Preventive Medicine, School of Medicine, Iwate Medical University, Iwate, Japan
| | - Akimune Fukushima
- Division of Innovation & Education, Iwate Tohoku Medical Megabank Organization, Disaster Reconstruction Center, Iwate Medical University, Yahaba, Japan
- Department of Clinical Genetics, School of Medicine, Iwate Medical University, Morioka, Japan
- Kitakami Saiseikai Hospital, Kitakami, Japan
| | - Makoto Sasaki
- Iwate Tohoku Medical Megabank Organization, Disaster Reconstruction Center, Iwate Medical University, Yahaba, Japan
- Division of Ultrahigh Field MRI, Institute for Biomedical Sciences, Iwate Medical University, Yahaba, Japan
| | - Atsushi Shimizu
- Division of Biomedical Information Analysis, Iwate Tohoku Medical Megabank Organization, Disaster Reconstruction Center, Iwate Medical University, Yahaba, Japan
- Division of Biomedical Information Analysis, Institute for Biomedical Sciences, Iwate Medical University, Yahaba, Japan
| |
Collapse
|
40
|
D'Aoust T, Clocchiatti‐Tuozzo S, Rivier CA, Mishra A, Hachiya T, Grenier‐Boley B, Soumaré A, Duperron M, Le Grand Q, Bouteloup V, Proust‐Lima C, Samieri C, Neuffer J, Sargurupremraj M, Chêne G, Helmer C, Thibault M, Amouyel P, Lambert J, Kamatani Y, Jacqmin‐Gadda H, Tregouët D, Inouye M, Dufouil C, Falcone GJ, Debette S. Polygenic score integrating neurodegenerative and vascular risk informs dementia risk stratification. Alzheimers Dement 2025; 21:e70014. [PMID: 40042447 PMCID: PMC11881617 DOI: 10.1002/alz.70014] [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: 08/02/2024] [Revised: 01/24/2025] [Accepted: 01/25/2025] [Indexed: 03/09/2025]
Abstract
INTRODUCTION An integrative polygenic risk score (iPRS) capturing the neurodegenerative and vascular contribution to dementia could identify high-risk individuals and improve risk prediction. METHODS We developed an iPRS for dementia (iPRS-DEM) in Europeans (aged 65+), comprising genetic risk for Alzheimer's disease (AD) and 23 vascular or neurodegenerative traits (excluding apolipoprotein E [APOE]). iPRS-DEM was evaluated across cohorts comprising older community-dwelling people (N = 3702), a multi-ancestry biobank (N = 130,797 Europeans; 105,404 non-Europeans), and dementia-free memory clinic participants (N = 2032). RESULTS iPRS-DEM was associated with dementia risk independently of APOE in the elderly (subdistribution hazard ratio [sHR]per1SD = 1.15, 95% confidence interval [CI]: 1.03 to 1.28), which generalized to Europeans (EUR-sHRper1SD = 1.28, 95% CI: 1.09 to 1.51]), East-Asians (EAS-sHRper1SD = 5.29, 95% CI: 1.43 to 34.36), and memory-clinic participants (sHRper1SD = 1.25, 95% CI: 1.11 to 1.42). Prediction was comparable to clinical risk factors in older community-dwelling people, with improved performance among memory-clinic patients. Risk stratification was enhanced by defining four genetic risk groups with iPRS-DEM and APOE ε4, reaching five-fold increased risk in APOE ε4+/iPRS-DEM+ memory-clinic participants. DISCUSSION Alongside APOE ε4, iPRS-DEM may refine risk stratification for the enrichment of dementia clinical trials and prevention programs. HIGHLIGHTS iPRS-DEM reflects neurodegenerative and vascular contribution to dementia. We show iPRS-DEM captures additional dementia genetic risk beyond APOE and AD-PRS. iPRS-DEM, in combination with APOE ε4, shows promise for dementia risk stratification. Our results generalize across both population-based and memory-clinic settings. We show transportability of iPRS-DEM to East Asian ancestry.
Collapse
Affiliation(s)
- Tim D'Aoust
- Bordeaux Population Health CenterINSERM, UMR U1219University of BordeauxBordeauxFrance
| | - Santiago Clocchiatti‐Tuozzo
- Department of NeurologyYale School of MedicineNew HavenConnecticutUSA
- Center for Brain and Mind HealthYale School of MedicineNew HavenConnecticutUSA
| | - Cyprien A. Rivier
- Department of NeurologyYale School of MedicineNew HavenConnecticutUSA
- Center for Brain and Mind HealthYale School of MedicineNew HavenConnecticutUSA
| | - Aniket Mishra
- Bordeaux Population Health CenterINSERM, UMR U1219University of BordeauxBordeauxFrance
| | - Tsuyoshi Hachiya
- Iwate Tohoku Medical Megabank OrganizationIwate Medical UniversityYahabaIwateJapan
| | - Benjamin Grenier‐Boley
- U1167‐RID‐AGE facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, INSERMInstitut Pasteur de LilleCHU LilleUniversity of LilleLilleFrance
| | - Aïcha Soumaré
- Bordeaux Population Health CenterINSERM, UMR U1219University of BordeauxBordeauxFrance
| | - Marie‐Gabrielle Duperron
- Bordeaux Population Health CenterINSERM, UMR U1219University of BordeauxBordeauxFrance
- Department of NeurologyInstitute for Neurodegenerative DiseasesCHU de BordeauxBordeauxFrance
| | - Quentin Le Grand
- Bordeaux Population Health CenterINSERM, UMR U1219University of BordeauxBordeauxFrance
| | - Vincent Bouteloup
- Bordeaux Population Health CenterINSERM, UMR U1219University of BordeauxBordeauxFrance
- CIC 1401 ECPôle Santé ´ PubliqueINSERMBordeauxFrance
| | - Cécile Proust‐Lima
- Bordeaux Population Health CenterINSERM, UMR U1219University of BordeauxBordeauxFrance
- CIC 1401 ECPôle Santé ´ PubliqueINSERMBordeauxFrance
| | - Cécilia Samieri
- Bordeaux Population Health CenterINSERM, UMR U1219University of BordeauxBordeauxFrance
| | - Jeanne Neuffer
- Bordeaux Population Health CenterINSERM, UMR U1219University of BordeauxBordeauxFrance
| | - Muralidharan Sargurupremraj
- Biggs Institute for Alzheimer's and Neurodegenerative DiseasesUniversity of Texas‐San AntonioSan AntonioTexasUSA
| | - Geneviève Chêne
- Bordeaux Population Health CenterINSERM, UMR U1219University of BordeauxBordeauxFrance
- Department of Public HealthCHU de BordeauxBordeauxFrance
| | - Catherine Helmer
- Bordeaux Population Health CenterINSERM, UMR U1219University of BordeauxBordeauxFrance
| | - Mura Thibault
- Institute for Neurosciences of Montpellier INMINSERM, UMR U1298University of MontpellierMontpellierFrance
| | - Philippe Amouyel
- U1167‐RID‐AGE facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, INSERMInstitut Pasteur de LilleCHU LilleUniversity of LilleLilleFrance
- Department of Epidemiology and Public HealthCHRU de LilleLilleFrance
| | - Jean‐Charles Lambert
- U1167‐RID‐AGE facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, INSERMInstitut Pasteur de LilleCHU LilleUniversity of LilleLilleFrance
| | - Yoichiro Kamatani
- Graduate School of Frontier SciencesThe University of TokyoKashiwaChibaJapan
| | - Hélène Jacqmin‐Gadda
- Bordeaux Population Health CenterINSERM, UMR U1219University of BordeauxBordeauxFrance
| | | | - Michael Inouye
- Department of Public HealthCambridge UniversityCambridgeUK
- Baker Heart and Diabetes InstituteMelbourneVictoriaAustralia
| | - Carole Dufouil
- Bordeaux Population Health CenterINSERM, UMR U1219University of BordeauxBordeauxFrance
- CIC 1401 ECPôle Santé ´ PubliqueINSERMBordeauxFrance
| | - Guido J. Falcone
- Department of NeurologyYale School of MedicineNew HavenConnecticutUSA
- Center for Brain and Mind HealthYale School of MedicineNew HavenConnecticutUSA
| | - Stéphanie Debette
- Bordeaux Population Health CenterINSERM, UMR U1219University of BordeauxBordeauxFrance
- Department of NeurologyInstitute for Neurodegenerative DiseasesCHU de BordeauxBordeauxFrance
- Institut du Cerveau (ICM), Paris Brain Institute, INSERM U1127, UMR CNRS 7225 ParisSorbonne Université, Assistance Publique des Hôpitaux de ParisParisFrance
| |
Collapse
|
41
|
Gallego-Fabrega C, Cullell N, Fernández-Cadenas I. How epigenetics impacts stroke risk and outcomes through DNA methylation: A systematic review. J Cereb Blood Flow Metab 2025:271678X251322032. [PMID: 40012472 DOI: 10.1177/0271678x251322032] [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] [Indexed: 02/28/2025]
Abstract
The impact of DNA methylation (DNAm) on epigenetics has gained prominence in recent years due to its potential influence on ischemic stroke (IS) and treatment outcomes. DNAm is reversible and a better understanding of its role in IS could help identify novel therapeutic targets. The aim of this systematic review was to compile the available data on DNAm in the risk and prognosis of IS and to explore its therapeutic potential. The review process followed the PRISMA criteria. We searched the Pubmed and Cochrane databases to identify studies that used hypothesis free methodological approaches. Of the 459 studies identified, 34 met the inclusion criteria. The studies were categorized as follows: risk of IS; outcomes; and DNAm age. Most studies used genotyping array technology rather than whole-genome sequencing. DNAm testing was mainly based on blood samples. Most studies involved European cohorts. Most of the studies were performed at a single-center with recruitment at the time of stroke. In a few studies, health status was determined longitudinally. This systematic review shows that IS patients are biologically older than expected and present characteristic DNAm patterns related to stroke risk and outcomes. These patterns could be used to develop new treatments with epidrugs.
Collapse
Affiliation(s)
- Cristina Gallego-Fabrega
- Stroke Pharmacogenomics and Genetics Group, Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain
| | - Natalia Cullell
- Stroke Pharmacogenomics and Genetics Group, Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain
- Neurology Unit, Hospital Universitari MútuaTerrassa, Terrassa, Spain
- Fundació per a Docencia i Recerca, Mútua Terrassa, Terrassa (Barcelona), Spain
| | - Israel Fernández-Cadenas
- Stroke Pharmacogenomics and Genetics Group, Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain
| |
Collapse
|
42
|
Zhao Y, Zhang R, Li P, Zhang Z, Yu H, Su Z, Xia Y, Meng A. A new nomogram for predicting 90-day outcomes of intravenous thrombolysis in patients with acute ischaemic stroke. Front Neurol 2025; 16:1512913. [PMID: 40083457 PMCID: PMC11905897 DOI: 10.3389/fneur.2025.1512913] [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: 10/17/2024] [Accepted: 02/07/2025] [Indexed: 03/16/2025] Open
Abstract
Background The aim of this study was to construct and validate a new nomogram to predict the risk of poor outcome in patients with acute ischemic stroke (AIS) after intravenous thrombolytic therapy (IVT). Methods A total of 425 patients who received IVT within 4.5 h of stroke onset were included in a retrospective study. All the patients were divided into training (70%, n = 298) and validation cohorts (30%, n = 127). Poor outcome (defined as a 90-day modified Rankin Scale score 3-5) was the primary outcome. Logistic regression was used for analysis of independent risk factors for poor outcome in patients with AIS. Nomograms of poor outcome in AIS patients were constructed using R software. Discrimination and calibration of the models were assessed using area under the receiver operating characteristic (ROC) curve (AUC) and calibration plots. Results Multifactorial logistic regression analysis showed that SII (OR = 1.001, 95% CI: 1.000-1.002, p = 0.008), SIRI (OR = 1.584, 95% CI: 1.122-2.236, p = 0.009), NIHSS (OR = 1.101, 95% CI: 1.044-1.160, p < 0.001), and history of diabetes mellitus (OR = 2.582, 95% CI: 1.285-5.188, p = 0.008) were the independent risk factors for the occurrence of poor outcome in AIS patients. The poor outcome nomogram for AIS patients was constructed based on the above independent risk factors. The training and validation cohort AUCs of the nomogram were 0.854 (95% CI: 0.807-0.901) and 0.855 (95% CI: 0.783-0.927), respectively. The prediction models were well calibrated in both the training and validation cohorts. The net benefit of the nomograms was better when the threshold probability ranges were 4.28-66.4% and 4.01-67.8% for the training and validation cohorts, respectively. Conclusion New nomogram includes NIHSS, SII, SIRI and diabetes as variables with the potential to predict the risk of 90-day outcomes in patients with AIS following IVT.
Collapse
Affiliation(s)
- Yingjie Zhao
- Department of Clinical Laboratory, North China University of Science and Technology Affiliated Hospital, Tangshan, China
| | - Rui Zhang
- Department of Clinical Laboratory, North China University of Science and Technology Affiliated Hospital, Tangshan, China
| | - Pan Li
- Department of Clinical Laboratory, North China University of Science and Technology Affiliated Hospital, Tangshan, China
| | - Zhen Zhang
- Department of Clinical Laboratory, North China University of Science and Technology Affiliated Hospital, Tangshan, China
| | - Huan Yu
- Department of Clinical Laboratory, North China University of Science and Technology Affiliated Hospital, Tangshan, China
| | - Zhaoya Su
- Department of Clinical Laboratory, North China University of Science and Technology Affiliated Hospital, Tangshan, China
| | - Yandong Xia
- Department of Clinical Laboratory, North China University of Science and Technology Affiliated Hospital, Tangshan, China
| | - Aiguo Meng
- Department of Clinical Laboratory, North China University of Science and Technology Affiliated Hospital, Tangshan, China
| |
Collapse
|
43
|
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
|
44
|
Wen J, Lin BM, Sun Q, Jiang MZ, Linchangco G, Li G, Chen R, Go AS, Miller-Fleming TW, Shuey MM, Cohen DL, Rao PS, Rahman M, Cox NJ, Lash JP, Guan W, Posner DC, Hui Q, Houghton SC, Hung AM, Cho K, Wilson PWF, Zhou H, Sun YV, Li Y, Franceschini N. Genetics of cardiovascular outcomes in individuals with chronic kidney disease: the Chronic Renal Insufficiency Cohort (CRIC) study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.19.25322572. [PMID: 40034774 PMCID: PMC11875325 DOI: 10.1101/2025.02.19.25322572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Genome-wide association studies (GWAS) identified multiple loci for cardiovascular disease, but their relevance to individuals with chronic kidney disease (CKD), who are at higher risk of cardiovascular disease, is unknown. In this study, we performed GWAS analyses of coronary heart disease (CHD) or all-cause stroke in African (AFR) and European (EUR) American participants with CKD of the Chronic Renal Insufficiency Cohort (CRIC). Mixed- effect logistic regression models were race-stratified and adjusted for age, sex, site of recruitment, estimated glomerular filtration rate (eGFR), and principal components, followed by meta-analysis. We attempted replication in participants from two biobanks with biomarker or ICD-10 (International Classification of Diseases, 10th Revision) diagnostic codes for CKD. We assessed the association of single nucleotide variants (SNVs) at known CHD and stroke loci in CRIC and tested the genetic correlation among CRIC, a biobank-based cohort and published GWAS of cardiovascular disease. Among 3,588 CRIC participants, 1,203 had CHD and 535 had all-cause stroke. We identified six SNVs across three loci ( LINC02744 , AZIN1- AS1 , and ATP6V0A4 ) associated with all-cause stroke, and two intronic SNVs at the PPARG locus associated with CHD. However, SNV associations were not significant in replication studies. Published SNVs for CHD or stroke were not associated with cardiovascular outcomes in CRIC. When testing the genetic correlations between published GWAS and CRIC GWAS, they were significant for CHD (genetic correlations (rg) range of 0.39 to 0.51, p-value< 0.007). These findings suggest some differences in the genetic architecture of CHD and stroke among individuals with CKD compared to those from the general population, although large numbers of CKD participants are needed to assess if findings are related to participant selection and CKD severity, or non-traditional risk factors in people with CKD.
Collapse
|
45
|
Wang M, Daghlas I, Zhang Z, Gill D, Liu D. MTHFR Polymorphisms, Homocysteine Elevation, and Ischemic Stroke Susceptibility in East Asian and European Populations. Neurology 2025; 104:e210245. [PMID: 39787475 DOI: 10.1212/wnl.0000000000210245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 10/31/2024] [Indexed: 01/12/2025] Open
Abstract
BACKGROUND AND OBJECTIVES Methylenetetrahydrofolate reductase (MTHFR) is a key enzyme that regulates folate and homocysteine metabolism. Genetic variation in MTHFR has been implicated in cerebrovascular disease risk, although research in diverse populations is lacking. We thus aimed to investigate the effect of genetically predicted MTHFR activity on risk of ischemic stroke (IS) and its main subtypes using a multiancestry Mendelian randomization (MR) approach. METHODS We proxied reduced MTHFR function using the C677T missense variant that impairs MTHFR function and consequently increases levels of total plasma homocysteine (tHcy) in both East Asian and European populations. Summary data for IS and its subtypes (small vessel stroke [SVS], large artery stroke [LAS], and cardioembolic stroke [CES]) were obtained from the largest available genome-wide association studies. MR estimates were calculated using the Wald ratio and random-effects inverse-variance-weighted methods. We performed sensitivity analyses to evaluate for confounding due to linkage disequilibrium. RESULTS Genetically downregulated MTHFR activity, associated with a consequent SD increase in tHcy levels, was associated with an increased risk of SVS in both East Asian (odds ratio [OR] 1.20, 95% CI 1.08-1.34, p = 8.58 × 10-4) and European populations (OR 1.62, 95% CI 1.24-2.12; p = 3.73 × 10-4). There was no evidence that genetically perturbed MTHFR activity influenced risk of CES or LAS. These findings were consistent in sensitivity analyses. DISCUSSION Our findings provide genetic evidence that reduced MTHFR activity was selectively associated with an increased risk of SVS in both East Asian and European populations. These findings warrant further investigation of genotype-guided nutritional supplementation for the prevention of SVS.
Collapse
Affiliation(s)
- Mengmeng Wang
- Department of Neurology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Iyas Daghlas
- Department of Neurology, University of California, San Francisco
| | - Zhizhong Zhang
- Department of Neurology, Jinling Hospital, Medical School of Nanjing University, China
| | - Dipender Gill
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, United Kingdom; and
| | - Dandan Liu
- Department of Integrated Traditional Chinese and Western Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, China
| |
Collapse
|
46
|
Liu H, Abedini A, Ha E, Ma Z, Sheng X, Dumoulin B, Qiu C, Aranyi T, Li S, Dittrich N, Chen HC, Tao R, Tarng DC, Hsieh FJ, Chen SA, Yang SF, Lee MY, Kwok PY, Wu JY, Chen CH, Khan A, Limdi NA, Wei WQ, Walunas TL, Karlson EW, Kenny EE, Luo Y, Kottyan L, Connolly JJ, Jarvik GP, Weng C, Shang N, Cole JB, Mercader JM, Mandla R, Majarian TD, Florez JC, Haas ME, Lotta LA, Regeneron Genetics Center, GHS-RGC DiscovEHR Collaboration, Drivas TG, Penn Medicine BioBank, Vy HMT, Nadkarni GN, Wiley LK, Wilson MP, Gignoux CR, Rasheed H, Thomas LF, Åsvold BO, Brumpton BM, Hallan SI, Hveem K, Zheng J, Hellwege JN, Zawistowski M, Zöllner S, Franceschini N, Hu H, Zhou J, Kiryluk K, Ritchie MD, Palmer M, Edwards TL, Voight BF, Hung AM, Susztak K. Kidney multiome-based genetic scorecard reveals convergent coding and regulatory variants. Science 2025; 387:eadp4753. [PMID: 39913582 PMCID: PMC12013656 DOI: 10.1126/science.adp4753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Collaborators] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 11/20/2024] [Indexed: 02/17/2025]
Abstract
Kidney dysfunction is a major cause of mortality, but its genetic architecture remains elusive. In this study, we conducted a multiancestry genome-wide association study in 2.2 million individuals and identified 1026 (97 previously unknown) independent loci. Ancestry-specific analysis indicated an attenuation of newly identified signals on common variants in European ancestry populations and the power of population diversity for further discoveries. We defined genotype effects on allele-specific gene expression and regulatory circuitries in more than 700 human kidneys and 237,000 cells. We found 1363 coding variants disrupting 782 genes, with 601 genes also targeted by regulatory variants and convergence in 161 genes. Integrating 32 types of genetic information, we present the "Kidney Disease Genetic Scorecard" for prioritizing potentially causal genes, cell types, and druggable targets for kidney disease.
Collapse
Affiliation(s)
- Hongbo Liu
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Diabetes Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
- Penn-CHOP Kidney Innovation Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Amin Abedini
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Diabetes Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Eunji Ha
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Diabetes Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Ziyuan Ma
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Diabetes Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Xin Sheng
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Diabetes Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
- Liangzhu Laboratory, Zhejiang University, 1369 West Wenyi Road, Hangzhou, Zhejiang, China
- Department of Nephrology, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| | - Bernhard Dumoulin
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Diabetes Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Chengxiang Qiu
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Diabetes Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Tamas Aranyi
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Diabetes Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Molecular Life Sciences, HUN-REN Research Center for Natural Sciences, Budapest, Hungary
- Department of Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Shen Li
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Diabetes Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Nicole Dittrich
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Diabetes Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
- Department of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Hua-Chang Chen
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ran Tao
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Der-Cherng Tarng
- Institute of Clinical Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- Division of Nephrology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Feng-Jen Hsieh
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan, ROC
| | - Shih-Ann Chen
- Cardiovascular Center, Taichung Veterans General Hospital, Taichung, Taiwan, ROC
- National Chung Hsing University, Taichung, Taiwan, ROC
- Heart Rhythm Center, Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Department of Internal Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan, ROC
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan, ROC
| | - Mei-Yueh Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan, ROC
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, ROC
- Department of Internal Medicine, Kaohsiung Medical University Gangshan Hospital, Kaohsiung, Taiwan, ROC
| | - Pui-Yan Kwok
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan, ROC
- Institute for Human Genetics, University of California, San Francisco, CA, USA
| | - Jer-Yuarn Wu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan, ROC
| | - Chien-Hsiun Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan, ROC
| | - Atlas Khan
- Division of Nephrology, Department of Medicine, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Nita A. Limdi
- Department of Neurology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Wei-Qi Wei
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Theresa L. Walunas
- Department of Medicine, Division of General Internal Medicine and Center for Health Information Partnerships, Institute for Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | | - Eimear E. Kenny
- Institute for Genomic Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Genomic Medicine, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of General Internal Medicine, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yuan Luo
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Leah Kottyan
- The Center for Autoimmune Genomics and Etiology, Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - John J. Connolly
- Center for Applied Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Gail P. Jarvik
- Departments of Medicine (Medical Genetics) and Genome Sciences, University of Washington, Seattle, WA, USA
| | - Chunhua Weng
- Department of Biomedical Informatics, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Ning Shang
- Division of Nephrology, Department of Medicine, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Joanne B. Cole
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Division of Endocrinology, Boston Children’s Hospital, Boston, MA, USA
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Josep M. Mercader
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Diabetes Unit, Endocrine Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Ravi Mandla
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Diabetes Unit, Endocrine Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine and Cardiovascular Research Institute, Cardiology Division, University of California, San Francisco, CA, USA
- Graduate Program in Genomics and Computational Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Timothy D. Majarian
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Vertex Pharmaceuticals, Boston, MA, USA
| | - Jose C. Florez
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Diabetes Unit, Endocrine Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Mary E. Haas
- Regeneron Genetics Center, Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | - Luca A. Lotta
- Regeneron Genetics Center, Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | | | | | - Theodore G. Drivas
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | | | - Ha My T. Vy
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Charles Bronfman Institute of Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Girish N. Nadkarni
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Charles Bronfman Institute of Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Hasso Plattner Institute of Digital Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Mount Sinai Clinical Intelligence Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Laura K. Wiley
- Colorado Center for Personalized Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Biomedical Informatics, University of Colorado School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Melissa P. Wilson
- Department of Biomedical Informatics, University of Colorado School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Christopher R. Gignoux
- Colorado Center for Personalized Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Biomedical Informatics, University of Colorado School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Humaira Rasheed
- KGJebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Laurent F. Thomas
- KGJebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Clinical and Molecular Medicine, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- BioCore - Bioinformatics Core Facility, Norwegian University of Science and Technology, Trondheim, Norway
| | - Bjørn Olav Åsvold
- KGJebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Endocrinology, Clinic of Medicine, St. Olav’s Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Ben M. Brumpton
- KGJebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- Clinic of Thoracic and Occupational Medicine, St. Olav’s Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Stein I. Hallan
- Department of Clinical and Molecular Medicine, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Nephrology, St. Olav’s Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Kristian Hveem
- KGJebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
| | - Jie Zheng
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jacklyn N. Hellwege
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Matthew Zawistowski
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Sebastian Zöllner
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
| | - Nora Franceschini
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Hailong Hu
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Diabetes Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Jianfu Zhou
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Diabetes Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Krzysztof Kiryluk
- Division of Nephrology, Department of Medicine, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Marylyn D. Ritchie
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Matthew Palmer
- Pathology and Laboratory Medicine at the Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Todd L. Edwards
- Division of Epidemiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Benjamin F. Voight
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA, USA
| | - Adriana M. Hung
- Division of Nephrology and Hypertension, Vanderbilt Center for Kidney Disease, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- VA Tennessee Valley Healthcare System, Clinical Sciences Research and Development, Nashville, TN, USA
| | - Katalin Susztak
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Diabetes Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
- Penn-CHOP Kidney Innovation Center, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
Collaborators
Aris Baras, Gonçalo Abecasis, Adolfo Ferrando, Giovanni Coppola, Andrew Deubler, Aris Economides, Luca A Lotta, John D Overton, Jeffrey G Reid, Alan Shuldiner, Katherine Siminovitch, Jason Portnoy, Marcus B Jones, Lyndon Mitnaul, Alison Fenney, Jonathan Marchini, Manuel Allen Revez Ferreira, Maya Ghoussaini, Mona Nafde, William Salerno, John D Overton, Christina Beechert, Erin Fuller, Laura M Cremona, Eugene Kalyuskin, Hang Du, Caitlin Forsythe, Zhenhua Gu, Kristy Guevara, Michael Lattari, Alexander Lopez, Kia Manoochehri, Prathyusha Challa, Manasi Pradhan, Raymond Reynoso, Ricardo Schiavo, Maria Sotiropoulos Padilla, Chenggu Wang, Sarah E Wolf, Hang Du, Kristy Guevara, Amelia Averitt, Nilanjana Banerjee, Dadong Li, Sameer Malhotra, Justin Mower, Mudasar Sarwar, Deepika Sharma, Sean Yu, Aaron Zhang, Muhammad Aqeel, Jeffrey G Reid, Mona Nafde, Manan Goyal, George Mitra, Sanjay Sreeram, Rouel Lanche, Vrushali Mahajan, Sai Lakshmi Vasireddy, Gisu Eom, Krishna Pawan Punuru, Sujit Gokhale, Benjamin Sultan, Pooja Mule, Eliot Austin, Xiaodong Bai, Lance Zhang, Sean O'Keeffe, Razvan Panea, Evan Edelstein, Ayesha Rasool, William Salerno, Evan K Maxwell, Boris Boutkov, Alexander Gorovits, Ju Guan, Lukas Habegger, Alicia Hawes, Olga Krasheninina, Samantha Zarate, Adam J Mansfield, Lukas Habegger, Gonçalo Abecasis, Joshua Backman, Kathy Burch, Adrian Campos, Liron Ganel, Sheila Gaynor, Benjamin Geraghty, Arkopravo Ghosh, Salvador Romero Martinez, Christopher Gillies, Lauren Gurski, Joseph Herman, Eric Jorgenson, Tyler Joseph, Michael Kessler, Jack Kosmicki, Adam Locke, Priyanka Nakka, Jonathan Marchini, Karl Landheer, Olivier Delaneau, Maya Ghoussaini, Anthony Marcketta, Joelle Mbatchou, Arden Moscati, Aditeya Pandey, Anita Pandit, Jonathan Ross, Carlo Sidore, Eli Stahl, Timothy Thornton, Sailaja Vedantam, Rujin Wang, Kuan-Han Wu, Bin Ye, Blair Zhang, Andrey Ziyatdinov, Yuxin Zou, Jingning Zhang, Kyoko Watanabe, Mira Tang, Frank Wendt, Suganthi Balasubramanian, Suying Bao, Kathie Sun, Chuanyi Zhang, Adolfo Ferrando, Giovanni Coppola, Luca A Lotta, Alan Shuldiner, Katherine Siminovitch, Brian Hobbs, Jon Silver, William Palmer, Rita Guerreiro, Amit Joshi, Antoine Baldassari, Cristen Willer, Sarah Graham, Ernst Mayerhofer, Erola Pairo Castineira, Mary Haas, Niek Verweij, George Hindy, Jonas Bovijn, Tanima De, Parsa Akbari, Luanluan Sun, Olukayode Sosina, Arthur Gilly, Peter Dornbos, Juan Rodriguez-Flores, Moeen Riaz, Manav Kapoor, Gannie Tzoneva, Momodou W Jallow, Anna Alkelai, Ariane Ayer, Veera Rajagopal, Sahar Gelfman, Vijay Kumar, Jacqueline Otto, Neelroop Parikshak, Aysegul Guvenek, Jose Bras, Silvia Alvarez, Jessie Brown, Jing He, Hossein Khiabanian, Joana Revez, Kimberly Skead, Valentina Zavala, Jae Soon Sul, Lei Chen, Sam Choi, Amy Damask, Nan Lin, Charles Paulding, Marcus B Jones, Esteban Chen, Michelle G LeBlanc, Jason Mighty, Jennifer Rico-Varela, Nirupama Nishtala, Nadia Rana, Jaimee Hernandez, Alison Fenney, Randi Schwartz, Jody Hankins, Anna Han, Samuel Hart, Ann Perez-Beals, Gina Solari, Johannie Rivera-Picart, Michelle Pagan, Sunilbe Siceron, Adam Buchanan, David J Carey, Christa L Martin, Michelle Meyer, Kyle Retterer, David Rolston, Daniel J Rader, Marylyn D Ritchie, JoEllen Weaver, Nawar Naseer, Giorgio Sirugo, Afiya Poindexter, Yi-An Ko, Kyle P Nerz, Meghan Livingstone, Fred Vadivieso, Stephanie DerOhannessian, Teo Tran, Julia Stephanowski, Salma Santos, Ned Haubein, Joseph Dunn, Anurag Verma, Colleen Morse Kripke, Marjorie Risman, Renae Judy, Colin Wollack, Shefali S Verma, Scott M Damrauer, Yuki Bradford, Scott M Dudek, Theodore G Drivas,
Collapse
|
47
|
Yoshiji S, Lu T, Butler-Laporte G, Carrasco-Zanini-Sanchez J, Su CY, Chen Y, Liang K, Willett JDS, Wang S, Adra D, Ilboudo Y, Sasako T, Koyama S, Nakao T, Forgetta V, Farjoun Y, Zeberg H, Zhou S, Marks-Hultström M, Machiela MJ, Kaalia R, Dashti H, Claussnitzer M, Flannick J, Wareham NJ, Mooser V, Timpson NJ, Langenberg C, Richards JB. Integrative proteogenomic analysis identifies COL6A3-derived endotrophin as a mediator of the effect of obesity on coronary artery disease. Nat Genet 2025; 57:345-357. [PMID: 39856218 PMCID: PMC11821532 DOI: 10.1038/s41588-024-02052-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 12/04/2024] [Indexed: 01/27/2025]
Abstract
Obesity strongly increases the risk of cardiometabolic diseases, yet the underlying mediators of this relationship are not fully understood. Given that obesity strongly influences circulating protein levels, we investigated proteins mediating the effects of obesity on coronary artery disease, stroke and type 2 diabetes. By integrating two-step proteome-wide Mendelian randomization, colocalization, epigenomics and single-cell RNA sequencing, we identified five mediators and prioritized collagen type VI α3 (COL6A3). COL6A3 levels were strongly increased by body mass index and increased coronary artery disease risk. Notably, the carboxyl terminus product of COL6A3, endotrophin, drove this effect. COL6A3 was highly expressed in disease-relevant cell types and tissues. Finally, we found that body fat reduction could reduce plasma levels of COL6A3-derived endotrophin, indicating a tractable way to modify endotrophin levels. In summary, we provide actionable insights into how circulating proteins mediate the effects of obesity on cardiometabolic diseases and prioritize endotrophin as a potential therapeutic target.
Collapse
Grants
- 169303 Gouvernement du Canada | Instituts de Recherche en Santé du Canada | CIHR Skin Research Training Centre (Skin Research Training Centre)
- 365825 Gouvernement du Canada | Instituts de Recherche en Santé du Canada | CIHR Skin Research Training Centre (Skin Research Training Centre)
- K99 HL169733 NHLBI NIH HHS
- 100558 Gouvernement du Canada | Instituts de Recherche en Santé du Canada | CIHR Skin Research Training Centre (Skin Research Training Centre)
- 409511 Gouvernement du Canada | Instituts de Recherche en Santé du Canada | CIHR Skin Research Training Centre (Skin Research Training Centre)
- 202460267 MEXT | Japan Society for the Promotion of Science (JSPS)
- Wellcome Trust
- The Richards research group is supported by the Canadian Institutes of Health Research (CIHR: 365825, 409511, 100558, 169303), the McGill Interdisciplinary Initiative in Infection and Immunity (MI4), the Lady Davis Institute of the Jewish General Hospital, the Jewish General Hospital Foundation, the Canadian Foundation for Innovation, the NIH Foundation, Cancer Research UK, Genome Québec, the Public Health Agency of Canada, McGill University, Cancer Research UK [grant number C18281/A29019] and the Fonds de Recherche Québec Santé (FRQS). J.B.R. is supported by an FRQS Mérite Clinical Research Scholarship. Support from Calcul Québec and Compute Canada is acknowledged. TwinsUK is funded by the Welcome Trust, Medical Research Council, European Union, the National Institute for Health Research (NIHR)-funded BioResource, Clinical Research Facility and Biomedical Research Centre based at Guy’s and St Thomas’ NHS Foundation Trust in partnership with King’s College London. NJT is a Wellcome Trust Investigator (202802/Z/16/Z), is the PI of the Avon Longitudinal Study of Parents and Children (MRC & WT 217065/Z/19/Z), is supported by the University of Bristol NIHR Biomedical Research Centre (BRC-1215-2001), the MRC Integrative Epidemiology Unit (MC_UU_00011/1) and works within the CRUK Integrative Cancer Epidemiology Programme (C18281/A29019).
- T.L. is supported by a Schmidt AI in Science Postdoctoral Fellowship, a Vanier Canada Graduate Scholarship, an FRQS doctoral training fellowship, and a McGill University Faculty of Medicine Studentship.
- G.B.L. is supported by scholarships from the FRQS, the CIHR, and Québec’s ministry of health and social services.
- Y.C. is supported by an FRQS doctoral training fellowship and the Lady Davis Institute/TD Bank Studentship Award.
- C-Y.S. is supported by a CIHR Canada Graduate Scholarship Doctoral Award, an FRQS doctoral training fellowship, and a Lady Davis Institute/ TD Bank Studentship Award.
Collapse
Affiliation(s)
- Satoshi Yoshiji
- Department of Human Genetics, McGill University, Montréal, Québec, Canada.
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada.
- Canada Excellence Research Chair in Genomic Medicine, Victor Phillip Dahdaleh Institute of Genomic Medicine, McGill University, Montréal, Québec, Canada.
- Kyoto-McGill International Collaborative Program in Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
- Programs in Metabolism and Medical & Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Tianyuan Lu
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
- Department of Statistical Sciences, University of Toronto, Toronto, ON, Canada
- Department of Population Health Sciences, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Department of Biostatistics and Medical Informatics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Guillaume Butler-Laporte
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
- Division of Infectious Diseases, McGill University Health Centre, Montréal, Québec, Canada
- Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Julia Carrasco-Zanini-Sanchez
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Precision Healthcare University Research Institute, Queen Mary University of London, London, UK
| | - Chen-Yang Su
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
- Canada Excellence Research Chair in Genomic Medicine, Victor Phillip Dahdaleh Institute of Genomic Medicine, McGill University, Montréal, Québec, Canada
- Quantitative Life Sciences Program, McGill University, Montréal, Québec, Canada
| | - Yiheng Chen
- Department of Human Genetics, McGill University, Montréal, Québec, Canada
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
- 5 Prime Sciences, Montréal, Québec, Canada
| | - Kevin Liang
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
- Quantitative Life Sciences Program, McGill University, Montréal, Québec, Canada
| | - Julian Daniel Sunday Willett
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
- Quantitative Life Sciences Program, McGill University, Montréal, Québec, Canada
- Department of Anatomic Pathology and Laboratory Medicine, New York Presbyterian - Weill Cornell Medical Center, New York, NY, USA
| | | | - Darin Adra
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
| | - Yann Ilboudo
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
| | - Takayoshi Sasako
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
| | - Satoshi Koyama
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Tetsushi Nakao
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | | | - Yossi Farjoun
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
- Fulcrum Genomics, Somerville, MA, USA
| | - Hugo Zeberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Sirui Zhou
- Department of Human Genetics, McGill University, Montréal, Québec, Canada
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
- Canada Excellence Research Chair in Genomic Medicine, Victor Phillip Dahdaleh Institute of Genomic Medicine, McGill University, Montréal, Québec, Canada
| | - Michael Marks-Hultström
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
- Anaesthesiology and Intensive Care Medicine, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
- Integrative Physiology, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Mitchell J Machiela
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Rama Kaalia
- Type 2 Diabetes Systems Genomics Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Hesam Dashti
- Type 2 Diabetes Systems Genomics Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Genomic Medicine and Endocrine Division, Massachusetts General Hospital, Boston, MA, USA
| | - Melina Claussnitzer
- Type 2 Diabetes Systems Genomics Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Genomic Medicine and Endocrine Division, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Jason Flannick
- Programs in Metabolism and Medical & Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Nicholas J Wareham
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Vincent Mooser
- Department of Human Genetics, McGill University, Montréal, Québec, Canada
- Canada Excellence Research Chair in Genomic Medicine, Victor Phillip Dahdaleh Institute of Genomic Medicine, McGill University, Montréal, Québec, Canada
| | - Nicholas J Timpson
- Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Claudia Langenberg
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Precision Healthcare University Research Institute, Queen Mary University of London, London, UK
- Computational Medicine, Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - J Brent Richards
- Department of Human Genetics, McGill University, Montréal, Québec, Canada.
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada.
- Quantitative Life Sciences Program, McGill University, Montréal, Québec, Canada.
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montréal, Québec, Canada.
- Department of Twin Research, King's College London, London, UK.
| |
Collapse
|
48
|
Wang S, Mu J, Wu Q, Chen L, Yin X. Circulating plasma protein identified as a therapeutic target for intracranial aneurysm through Mendelian Randomization analysis. J Clin Neurosci 2025; 132:110998. [PMID: 39721116 DOI: 10.1016/j.jocn.2024.110998] [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: 08/04/2024] [Revised: 11/21/2024] [Accepted: 12/18/2024] [Indexed: 12/28/2024]
Abstract
BACKGROUND Intracranial aneurysms are the main cause of subarachnoid hemorrhage (SAH), a severe stroke with devastating effects. However, there are no existing medications for intracranial aneurysms (IAs) and novel therapeutic targets are required. METHODS We performed a summary data-based Mendelian Randomization (MR) analysis to explore the causal association between circulating plasma proteins and the risk of IAs and SAH. Colocalization analysis was conducted to identify shared causal variants between circulating plasma proteins and IAs, as well as SAH. Finally, mediation MR analyses were conducted to clarify the role of potential plasma proteins in aneurysm formation. RESULTS Proteome-wide MR analysis showed that FGF5 (fibroblast growth factor 5) had a causal effect on IA and SAH risk (Pfdr < 0.05). Moreover, genetic variants affecting FGF5 expression levels showed strong evidence of colocalization with IA risk (PPH4 = 0.993) and SAH risk (PPH = 0.988), suggesting that this protein represents a potential direct target for IA intervention. Mediation analysis using two-step MR showed that systolic blood pressure and diastolic blood pressure mediate the effects of FGF5 on IA and SAH. CONCLUSION Our investigation identified a causal connection between FGF5 and IAs.
Collapse
Affiliation(s)
- Songquan Wang
- Department of Neurosurgery, Second Hospital of Shanxi Medical University, Taiyuan, PR China
| | - Jiali Mu
- Department of Cardiology, Shanxi Cardiovascular Hospital, Taiyuan, PR China
| | - Quansheng Wu
- Department of Neurosurgery, Second Hospital of Shanxi Medical University, Taiyuan, PR China
| | - Laizhao Chen
- Department of Neurosurgery, Second Hospital of Shanxi Medical University, Taiyuan, PR China
| | - Xiaofeng Yin
- Department of Neurosurgery, Second Hospital of Shanxi Medical University, Taiyuan, PR China.
| |
Collapse
|
49
|
Moura S, Nasciben LB, Ramirez AM, Coombs L, Rivero J, Van Booven DJ, DeRosa BA, Hamilton‐Nelson KL, Whitehead PL, Adams LD, Starks TD, Mena PR, Illanes‐Manrique M, Tejada S, Byrd GS, Cornejo‐Olivas MR, Feliciano‐Astacio BE, Nuytemans K, Wang L, Pericak‐Vance MA, Dykxhoorn DM, Rajabli F, Griswold AJ, Young JI, Vance JM. Comparing Alzheimer's genes in African, European, and Amerindian induced pluripotent stem cell-derived microglia. Alzheimers Dement 2025; 21:e70031. [PMID: 40008916 PMCID: PMC11863361 DOI: 10.1002/alz.70031] [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: 09/26/2024] [Revised: 01/14/2025] [Accepted: 01/29/2025] [Indexed: 02/27/2025]
Abstract
INTRODUCTION Genome-wide association studies (GWAS) studies in Alzheimer's disease (AD) demonstrate ancestry-specific loci. Previous studies in the regulatory architecture have only been conducted in Europeans (EUs), thus studies in additional ancestries are needed. Given the prevalence of AD genes expressed in microglia, we initiated our studies in induced pluripotent stem cell (iPSC) -derived microglia. METHODS We created iPSC-derived microglia from 13 individuals of either high Amerindian (AI), African (AF), or EU global ancestry, including both AD and controls. RNA-seq, ATAC-seq, and pathway analyses were compared between ancestries in both AD and non-AD genes. RESULTS Twelve AD genes were differentially expressed genes (DEGs) and/or accessible between ancestries, including ABI3, CTSB, and MS4A6A. A total of 5% of all genes had differential ancestral expression, but differences in accessibility were less than 1%. The DEGs were enriched in known AD pathways. DISCUSSION This resource will be valuable in evaluating AD in admixed populations and other neurological disorders and understanding the AD risk differences between populations. HIGHLIGHTS First comparison of the genomics of AI, AF, and EU microglia. Report differences in expression and accessibility of AD genes between ancestries. Ancestral expression differences are greater than differences in accessibility. Good transcriptome correlation was seen between brain and iPSC-derived microglia. Differentially expressed AD genes were in known AD pathways.
Collapse
Affiliation(s)
- Sofia Moura
- John P. Hussman Institute for Human GenomicsUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Luciana Bertholim Nasciben
- John P. Hussman Institute for Human GenomicsUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Aura M. Ramirez
- John P. Hussman Institute for Human GenomicsUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Lauren Coombs
- John P. Hussman Institute for Human GenomicsUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Joe Rivero
- John P. Hussman Institute for Human GenomicsUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Derek J. Van Booven
- John P. Hussman Institute for Human GenomicsUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Brooke A. DeRosa
- John P. Hussman Institute for Human GenomicsUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Kara L. Hamilton‐Nelson
- John P. Hussman Institute for Human GenomicsUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Patrice L. Whitehead
- John P. Hussman Institute for Human GenomicsUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Larry D. Adams
- John P. Hussman Institute for Human GenomicsUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Takiyah D. Starks
- Maya Angelou Center for Health EquityWake Forest UniversityWinston‐SalemNorth CarolinaUSA
| | - Pedro R. Mena
- John P. Hussman Institute for Human GenomicsUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Maryenela Illanes‐Manrique
- Neurogenetics Working GroupUniversidad Científica del SurVilla EL SalvadorPeru
- Neurogenetics Research CenterInstituto Nacional de Ciencias NeurológicasLimaPeru
| | - Sergio Tejada
- John P. Hussman Institute for Human GenomicsUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Goldie S. Byrd
- Maya Angelou Center for Health EquityWake Forest UniversityWinston‐SalemNorth CarolinaUSA
| | - Mario R. Cornejo‐Olivas
- Neurogenetics Working GroupUniversidad Científica del SurVilla EL SalvadorPeru
- Neurogenetics Research CenterInstituto Nacional de Ciencias NeurológicasLimaPeru
| | | | - Karen Nuytemans
- John P. Hussman Institute for Human GenomicsUniversity of Miami Miller School of MedicineMiamiFloridaUSA
- Dr. John T. Macdonald Foundation Department of Human GeneticsUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Liyong Wang
- John P. Hussman Institute for Human GenomicsUniversity of Miami Miller School of MedicineMiamiFloridaUSA
- Dr. John T. Macdonald Foundation Department of Human GeneticsUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Margaret A. Pericak‐Vance
- John P. Hussman Institute for Human GenomicsUniversity of Miami Miller School of MedicineMiamiFloridaUSA
- Dr. John T. Macdonald Foundation Department of Human GeneticsUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Derek M. Dykxhoorn
- John P. Hussman Institute for Human GenomicsUniversity of Miami Miller School of MedicineMiamiFloridaUSA
- Dr. John T. Macdonald Foundation Department of Human GeneticsUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Farid Rajabli
- John P. Hussman Institute for Human GenomicsUniversity of Miami Miller School of MedicineMiamiFloridaUSA
- Dr. John T. Macdonald Foundation Department of Human GeneticsUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Anthony J. Griswold
- John P. Hussman Institute for Human GenomicsUniversity of Miami Miller School of MedicineMiamiFloridaUSA
- Dr. John T. Macdonald Foundation Department of Human GeneticsUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Juan I. Young
- John P. Hussman Institute for Human GenomicsUniversity of Miami Miller School of MedicineMiamiFloridaUSA
- Dr. John T. Macdonald Foundation Department of Human GeneticsUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Jeffery M. Vance
- John P. Hussman Institute for Human GenomicsUniversity of Miami Miller School of MedicineMiamiFloridaUSA
- Dr. John T. Macdonald Foundation Department of Human GeneticsUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| |
Collapse
|
50
|
Ran L, Fang Y, Cheng C, He Y, Shao Z, Kong Y, Huang H, Xu S, Luo X, Wang W, Hao X, Wang M. Genome-wide and phenome-wide studies provided insights into brain glymphatic system function and its clinical associations. SCIENCE ADVANCES 2025; 11:eadr4606. [PMID: 39823331 PMCID: PMC11740961 DOI: 10.1126/sciadv.adr4606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 12/16/2024] [Indexed: 01/19/2025]
Abstract
We applied an MRI technique diffusion tensor imaging along the perivascular space (DTI-ALPS) for assessing glymphatic system (GS) in a genome-wide association study (GWAS) and phenome-wide association study (PheWAS) of 40,486 European individuals. Exploratory analysis revealed 17 genetic loci significantly associating with the regional DTI-ALPS index. We found 58 genes, including SPPL2C and EFCAB5, which prioritized in the DTI-ALPS index subtypes and associated with neurodegenerative diseases. PheWAS of 241 traits suggested that body mass index and blood pressure phenotypes closely related to GS function. Moreover, we detected disrupted GS function in 44 of 625 predefined disease conditions. Notably, Mendelian randomization and mediation analysis indicated that lower DTI-ALPS index was a risk factor for ischemic stroke (odds ratio = 1.56, P = 0.028) by partly mediating the risk factor of obesity. Results provide insights into the genetic architecture and mechanism for the DIT-ALPS index and highlight its great clinical value, especially in cerebral stroke.
Collapse
Affiliation(s)
- Lusen Ran
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuanyuan Fang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chang Cheng
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuqin He
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhonghe Shao
- Department of Epidemiology and Biostatistics, Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yifan Kong
- Department of Epidemiology and Biostatistics, Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hao Huang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shabei Xu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiang Luo
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xingjie Hao
- Department of Epidemiology and Biostatistics, Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Minghuan Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|