1
|
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
|
2
|
Xu C, Yang ML, Kho PF, Clarke SL, Tcheandjieu C, Peyser PA, Fann CSJ, Chen SP, Saw J, Zhou X, Assimes TL, Ganesh SK. Cross-Ancestry Associations of Spontaneous Coronary Artery Dissection Genetic Risk With Coronary Atherosclerosis and Migraine Headache. J Am Heart Assoc 2025; 14:e036525. [PMID: 40357661 DOI: 10.1161/jaha.124.036525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 03/12/2025] [Indexed: 05/15/2025]
Abstract
BACKGROUND Research studies of spontaneous coronary artery dissection (SCAD) have been primarily focused on European-ancestry individuals, with limited recognition and investigation in non-European-ancestry individuals. While SCAD has not been well ascertained in non-European-ancestry groups, pleiotropic associated traits identified in those of European ancestry have been assessed in individuals of other ancestries. Whether these traits are associated with the complex genetic architecture of SCAD in those of non-European ancestry has not been previously investigated. METHODS We investigated the associations of an established SCAD polygenic score with multiple vascular diseases in ≈900 000 ancestrally diverse participants of large-scale studies. Individual-level data from the UK Biobank and the Million Veteran Program and summary statistics of publicly available databases were analyzed. RESULTS A set of associations between SCAD polygenic score and related vascular diseases were replicated in non-European samples. Notable associations with the SCAD polygenic score included (1) coronary artery disease, myocardial infarction, and migraine headache in a Hispanic group (coronary artery disease: odds ratio [OR], 0.93 [95% CI, 0.90-0.95]; P=2.35×10-7; myocardial infarction: OR, 0.88 [95% CI, 0.80-0.96]; P=5.73×10-3; migraine headache: OR, 1.03 [95% CI, 1.01-1.06]; P=1.86×10-2) of the Million Veteran Program; (2) headache in an African-ancestry group (OR, 1.22 [95% CI, 1.06-1.41]; P=6.94×10-3) and a South Asian-ancestry group (OR, 1.18 [95% CI, 1.02-1.37]; P=2.43×10-2) of the UK Biobank; and (3) coronary artery disease, myocardial infarction, and migraine headache in East Asian-ancestry cohorts (coronary artery disease: OR, 0.95 [95% CI, 0.93-0.98]; P=2.66×10-3; myocardial infarction: OR, 0.86 [95% CI, 0.83-0.89]; P=9.51×10-16; migraine headache: OR, 1.27 [95% CI, 1.10-1.47]; P=1.03×10-3). CONCLUSIONS Pleiotropic associations of SCAD polygenic risk with related vascular diseases previously identified in European-ancestry groups showed notable, largely consistent patterns in non-European-ancestry groups.
Collapse
Affiliation(s)
- Chang Xu
- Department of Biostatistics and Center for Statistical Genetics University of Michigan School of Public Health Ann Arbor MI USA
| | - Min-Lee Yang
- Division of Cardiovascular Medicine, Department of Internal Medicine University of Michigan Medical School Ann Arbor MI USA
- Department of Computational Medicine and Bioinformatics University of Michigan Ann Arbor MI USA
- Department of Human Genetics University of Michigan Medical School Ann Arbor MI USA
| | - Pik Fang Kho
- VA Palo Alto Health Care System Palo Alto CA USA
- Department of Medicine (Division of Cardiovascular Medicine) Stanford University School of Medicine Stanford CA USA
| | - Shoa L Clarke
- VA Palo Alto Health Care System Palo Alto CA USA
- Department of Medicine (Division of Cardiovascular Medicine) Stanford University School of Medicine Stanford CA USA
| | - Catherine Tcheandjieu
- VA Palo Alto Health Care System Palo Alto CA USA
- Gladstone Institute of Data Science and Biotechnology Gladstone Institutes San Francisco CA USA
- Department of Epidemiology and Biostatistics University of California San Francisco San Francisco CA USA
| | - Patricia A Peyser
- Department of Epidemiology University of Michigan School of Public Health Ann Arbor MI USA
| | | | - Shih-Pin Chen
- Department of Medical Research Taipei Veterans General Hospital Taipei Taiwan
- Institute of Clinical Medicine National Yang Ming Chiao Tung University Taipei Taiwan
| | - Jacqueline Saw
- Vancouver General Hospital, Division of Cardiology University of British Columbia Vancouver Canada
| | - Xiang Zhou
- Department of Biostatistics and Center for Statistical Genetics University of Michigan School of Public Health Ann Arbor MI USA
| | - Themistocles L Assimes
- VA Palo Alto Health Care System Palo Alto CA USA
- Department of Medicine (Division of Cardiovascular Medicine) Stanford University School of Medicine Stanford CA USA
| | - Santhi K Ganesh
- Division of Cardiovascular Medicine, Department of Internal Medicine University of Michigan Medical School Ann Arbor MI USA
- Department of Human Genetics University of Michigan Medical School Ann Arbor MI USA
| |
Collapse
|
3
|
Chang X, Tao L, Tian L, Zhao Y, Niku W, Zheng W, Liu P, Wang Y. Identification of hub biomarkers in coronary artery disease patients using machine learning and bioinformatic analyses. Sci Rep 2025; 15:17244. [PMID: 40383719 PMCID: PMC12086195 DOI: 10.1038/s41598-025-02123-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 05/12/2025] [Indexed: 05/20/2025] Open
Abstract
Understanding the molecular underpinnings of CAD is essential for developing effective therapeutic strategies. This study aims to identify and analyze differentially expressed hub biomarkers in the peripheral blood of CAD patients. Based on RNA-seq datasets from the Gene Expression Omnibus database, machine learning algorithms including LASSO, RF, and SVM-RFE were applied. Furthermore, the hub biomarkers were enriched to ascertain their roles in immune cell expression and signaling pathways through GO, KEGG, GSVE, and GSVA. An in vivo experiment was conducted to verify the hub biomarkers. Eleven hub biomarkers (ITM2B, GNA15, PLAU, GNG11, HIST1H2BH, SLC11A1, RPS7, DDIT4, CD83, GNLY, and S100A12) were identified and associated with CD8 + T cells and NK cells. They were mainly involved in immune responses, cardiac muscle contraction, oxidative phosphorylation, and apoptotic signaling pathways. Moreover, ITM2B had the most importance and significance to be the biomarker of CAD patients. In conclusion, these findings point to the possibility of ITM2B as a biomarker on the inflammatory pathogenesis of CAD and suggest new options for therapeutic intervention.
Collapse
Affiliation(s)
- Xindi Chang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Liyu Tao
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | | | - Yingli Zhao
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wangkang Niku
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wang Zheng
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ping Liu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Yiru Wang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
4
|
Yang Q, Wang X, Han M, Sheng H, Sun Y, Su L, Lu W, Li M, Wang S, Chen J, Cui S, Yang BW. Bacterial genome-wide association studies: exploring the genetic variation underlying bacterial phenotypes. Appl Environ Microbiol 2025:e0251224. [PMID: 40377303 DOI: 10.1128/aem.02512-24] [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: 05/18/2025] Open
Abstract
With the continuous advancements in high-throughput genome sequencing technologies and the development of innovative bioinformatics tools, bacterial genome-wide association studies (BGWAS) have emerged as a transformative approach for investigating the genetic variations underlying diverse bacterial phenotypes at the population genome level. This review provides a comprehensive overview of the application of BGWAS in elucidating genetic determinants of bacterial drug resistance, pathogenicity, host specificity, biofilm formation, and probiotic fermentation characteristics. We systematically summarize the BGWAS workflow, including study design, data analysis pipelines, and the bioinformatics software employed at various stages. Furthermore, we highlight specialized tools tailored for BGWAS and discuss their unique features and applications. We also discuss confounding factors that can influence the accuracy and reliability of BGWAS results, including population structure, linkage disequilibrium, and multiple testing. By incorporating recent advancements, this review serves as a comprehensive reference for researchers utilizing BGWAS to uncover the genetic basis of bacterial phenotypes.
Collapse
Affiliation(s)
- Qiuping Yang
- College of Food Science and Engineering, Northwest A&F University, Shaanxi, China
| | - Xiaoqi Wang
- College of Food Science and Engineering, Northwest A&F University, Shaanxi, China
| | - Mengting Han
- College of Food Science and Engineering, Northwest A&F University, Shaanxi, China
| | - Huanjing Sheng
- College of Food Science and Engineering, Northwest A&F University, Shaanxi, China
| | - Yulu Sun
- College of Food Science and Engineering, Northwest A&F University, Shaanxi, China
| | - Li Su
- College of Food Science and Engineering, Northwest A&F University, Shaanxi, China
| | - Wenjing Lu
- College of Food Science and Engineering, Northwest A&F University, Shaanxi, China
| | - Mei Li
- College of Food Science and Engineering, Northwest A&F University, Shaanxi, China
| | - Siyue Wang
- College of Food Science and Engineering, Northwest A&F University, Shaanxi, China
| | - Jia Chen
- College of Chemical Technology, Shijiazhuang University, Shijiazhuang, China
| | - Shenghui Cui
- National Institutes for Food and Drug Control, Beijing, China
| | - Bao-Wei Yang
- College of Food Science and Engineering, Northwest A&F University, Shaanxi, China
| |
Collapse
|
5
|
Damiani I, Solberg EH, Iyer M, Cheng P, Weldy CS, Kim JB. Environmental pollutants and atherosclerosis: Epigenetic mechanisms linking genetic risk and disease. Atherosclerosis 2025; 404:119131. [PMID: 39986958 PMCID: PMC12034486 DOI: 10.1016/j.atherosclerosis.2025.119131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 01/14/2025] [Accepted: 02/11/2025] [Indexed: 02/24/2025]
Abstract
Over the past half-century, significant strides have been made to identify key risk factors, genetic mechanisms, and treatments for atherosclerosis. Yet, coronary artery disease (CAD) remains a leading global public health challenge. While the heritability of CAD is well-documented, there is increasing focus on the role of environmental exposures, such as smoking, air pollution, and heavy metals, on global CAD risk. Recent research has shed light on the interplay between genetic variation and environmental factors, offering insights into gene-environment (GxE) interactions. Moreover, emerging evidence suggests that environmental toxicants can profoundly impact the epigenome, altering gene regulation beyond the genetic sequence itself, revealing novel mechanisms underlying disease. Epigenetic changes - such as modifications in DNA methylation, chromatin structure, and non-coding RNA function - are now recognized as key molecular determinants of atherosclerosis. These observations have created a foundational paradigm that environment, genetics, and epigenetic mechanisms influence risk through a highly complex interaction regulating cellular phenotype, pathology, and disease progression. In this review, we explore the mechanisms by which environmental exposures influence the epigenome and contribute to the regulation of atherosclerotic disease. Additionally, we examine the transgenerational epigenetic effects of these exposures on disease risk. Advancing our understanding of these mechanisms is essential for informing public health strategies aimed at mitigating harmful environmental exposures and reducing the global burden of cardiovascular disease.
Collapse
Affiliation(s)
- Isabella Damiani
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Elena Hurtado Solberg
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA; Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Meghana Iyer
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Paul Cheng
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA; Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Chad S Weldy
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA; Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, 94305, USA; Stanford Center for Inherited Cardiovascular Disease, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| | - Juyong Brian Kim
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA; Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| |
Collapse
|
6
|
Suglia SF, Hidalgo B, Baccarelli AA, Cardenas A, Damrauer S, Johnson A, Key K, Liang M, Magnani JW, Pate B, Sims M, Tajeu GS. Improving Cardiovascular Health Through the Consideration of Social Factors in Genetics and Genomics Research: A Scientific Statement From the American Heart Association. Circ Cardiovasc Qual Outcomes 2025; 18:e000138. [PMID: 40123498 DOI: 10.1161/hcq.0000000000000138] [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: 03/25/2025]
Abstract
Cardiovascular health (CVH) is affected by genetic, social, and genomic factors across the life course, yet little research has focused on the interrelationships among them. An extensive body of work has documented the impact of social determinants of health at both the structural and individual levels on CVH, highlighting pathways in which racism, housing, violence, and neighborhood environments adversely affect CVH and contribute to disparities in cardiovascular disease. Genetic factors have also been identified as contributors to risk for cardiovascular disease. Emerging evidence suggests that social factors can interact with genetic susceptibility to affect disease risk. Increasingly, social factors have been shown to affect epigenetic markers such as DNA methylation, which can regulate gene and protein expression. This is a potential biological mechanism through which exposure to poor social determinants of health becomes physically embodied at the molecular level, potentially contributing to the development of suboptimal CVH and chronic disease, thus reinforcing and propagating health disparities. The objective of this statement is to highlight and summarize key literature that has examined the joint associations between social, genetic, and genomic factors and CVH and cardiovascular disease.
Collapse
|
7
|
Wayne N, Guerraty MA. Functional genomics identifies a protective role for FDX1 in atherosclerosis. NATURE CARDIOVASCULAR RESEARCH 2025; 4:503-504. [PMID: 40360794 DOI: 10.1038/s44161-025-00636-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Affiliation(s)
- Nicole Wayne
- Department of Medicine, Division of Cardiovascular Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Marie A Guerraty
- Department of Medicine, Division of Cardiovascular Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
8
|
Khodursky S, Mimouni N, Levin MG. Recent developments in population biobanks and the genetic architecture of complex disease. Hum Mol Genet 2025:ddaf036. [PMID: 40292753 DOI: 10.1093/hmg/ddaf036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 02/05/2025] [Accepted: 03/09/2025] [Indexed: 04/30/2025] Open
Abstract
Population biobanks have radically transformed our understanding of complex disease genetics. Recent technological advances and the inclusion of diverse populations have accelerated the discovery and interpretation of variant associations. For instance, population-scale whole-genome sequencing now allows deep exploration of rare and structural variant associations, while multi-omics approaches integrating genome-wide association studies with proteomics, metabolomics, and advanced statistical methods like Mendelian randomization provide nuanced insights into genetic disease mechanisms. Additionally, cross-biobank collaborations and meta-analyses have been particularly impactful, dramatically increasing the statistical power for discovery. These efforts have identified novel genetic associations across numerous complex diseases, with significant contributions from non-European populations. However, data integration complexities, privacy concerns, and methodological limitations continue to constrain research. Here we review how recent advances have contributed to genetic discovery.
Collapse
Affiliation(s)
- Samuel Khodursky
- University of Pennsylvania Perelman School of Medicine, 3400 Civic Center Blvd., Philadelphia, PA 19104, United States
| | - Nour Mimouni
- University of Pennsylvania Perelman School of Medicine, 3400 Civic Center Blvd., Philadelphia, PA 19104, United States
| | - Michael G Levin
- University of Pennsylvania Perelman School of Medicine, 3400 Civic Center Blvd., Philadelphia, PA 19104, United States
- Division of Cardiovascular Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, 3400 Civic Center Blvd., Philadelphia, PA 19104, United States
- Department of Medicine, Corporal Michael J. Crescenz VA Medical Center, 3900 Woodland Ave., Philadelphia, PA 19104, United States
| |
Collapse
|
9
|
Liu D, Yang C, Guo T, Guo Y, Xiong J, Chen R, Deng S. Associations Between Physical Capability Markers and Risk of Coronary Artery Disease: A Prospective Study of 439,295 UK Biobank Participants. Healthcare (Basel) 2025; 13:1018. [PMID: 40361796 PMCID: PMC12071247 DOI: 10.3390/healthcare13091018] [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: 02/13/2025] [Revised: 04/21/2025] [Accepted: 04/22/2025] [Indexed: 05/15/2025] Open
Abstract
Background: The relationship between sarcopenia and the incidence of coronary artery disease (CAD) is not well understood. This study aimed to investigate this relationship and the modifying effect of potential risk factors. Methods: We conducted a prospective study including 439,295 individuals from the UK Biobank. The primary outcome was the incidence of CAD. The main physical capability markers for sarcopenia, grip strength and muscle mass, were investigated as risk factors of interest. Grip strength was measured using a Jamar J00105 (Lafayette, IN, USA) hydraulic hand dynamometer, while muscle mass was estimated through bioelectrical impedance. Cox proportional hazard models were employed to analyze the associations between the exposures and the risk of CAD. Results: A total of 41,564 incident cases of CAD were identified after a median follow-up of 13.15 years (IQR 12.29-13.88 years). Compared with the lowest quintile of grip strength, the adjusted HRs for incidences of CAD from the second to the fifth quintile were 0.81 (95% CI: 0.79-0.83), 0.71 (95% CI: 0.69-0.73), 0.61 (95% CI: 0.60-0.63), and 0.49 (95% CI: 0.48-0.51). The association remained significant in subgroup analysis and interactions were observed between the two exposures and sex, age, smoking status, inflammatory diseases, metabolic syndrome, and genetic predisposition (all p for interactions < 0.05). Conclusions: Physical capability markers of sarcopenia, grip strength and muscle mass, were independently associated with a dose-response decreased risk for CAD incidence, regardless of genetic predisposition and potential modifying risk factors.
Collapse
Affiliation(s)
- Duqiu Liu
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (D.L.); (Y.G.)
- Liyuan Cardiovascular Center, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430077, China
| | - Chenxing Yang
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China;
| | - Tianyu Guo
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Ministry of Education Key Laboratory of Environment and Health, and State Key Laboratory of Environment Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China;
| | - Yi Guo
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (D.L.); (Y.G.)
- Liyuan Cardiovascular Center, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430077, China
| | - Jinjie Xiong
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China;
| | - Ru Chen
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (D.L.); (Y.G.)
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China;
| | - Shan Deng
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (D.L.); (Y.G.)
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China;
| |
Collapse
|
10
|
de Vries P, Hasbani N, Heath A, Hodonsky C, Hahn J, Meena D, Lu H, Dehghan AA, Kavousi M, Voight B, Peyser P, Morrison A, Assimes T, Damrauer S, Miller C. A multi-trait genome-wide association study of coronary artery disease and subclinical atherosclerosis traits. RESEARCH SQUARE 2025:rs.3.rs-6456056. [PMID: 40313769 PMCID: PMC12045367 DOI: 10.21203/rs.3.rs-6456056/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
Measures of subclinical atherosclerosis, such as coronary artery calcification (CAC) and carotid intima-media thickness (CIMT), reflect the underlying pathophysiology of coronary artery disease (CAD) and are genetically correlated with CAD and related risk factors. Leveraging summary statistics from genome-wide association studies of CAD, CIMT, CAC, type 2 diabetes, low-density lipoprotein cholesterol, and systolic blood pressure, we performed 15 separate multi-trait GWAS to identify shared susceptibility loci and elucidate the pleiotropic architecture underlying atherosclerosis. We identified 442 shared risk loci across all analyses that met an experiment-wide Bonferroni threshold of 3.3 × 10-9, uncovering 195 novel atherosclerosis loci. Multi-trait colocalization confirmed a shared causal signal in 25 shared novel loci for atherosclerosis. Trait-eQTL colocalization identified evidence of a shared causal signal in arterial, subcutaneous adipose, and cardiac tissues, implicating genes such as PRRX2, BNC2, CLIC4, SCAI, and PPP6C, and pathways related to vascular remodeling, inflammation, and metabolic regulation.
Collapse
Affiliation(s)
| | | | - Adam Heath
- The University of Texas Health Science Center at Houston
| | | | - Julie Hahn
- The University of Texas Health Science Center at Houston
| | | | | | | | | | | | - Patricia Peyser
- Department of Epidemiology, School of Public Health, University of Michigan
| | | | | | | | | |
Collapse
|
11
|
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
|
12
|
Chen SF, Lee SE, Sadaei HJ, Park JB, Khattab A, Chen JF, Henegar C, Wineinger NE, Muse ED, Torkamani A. Meta-prediction of coronary artery disease risk. Nat Med 2025:10.1038/s41591-025-03648-0. [PMID: 40240837 DOI: 10.1038/s41591-025-03648-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 03/07/2025] [Indexed: 04/18/2025]
Abstract
Coronary artery disease (CAD) is a leading cause of morbidity and mortality worldwide, and accurately predicting individual risk is critical for prevention. Here we aimed to integrate unmodifiable risk factors, such as age and genetics, with modifiable risk factors, such as clinical and biometric measurements, into a meta-prediction framework that produces actionable and personalized risk estimates. In the initial development of the model, ~2,000 predictive features were considered, including demographic data, lifestyle factors, physical measurements, laboratory tests, medication usage, diagnoses and genetics. To power our meta-prediction approach, we stratified the UK Biobank into two primary cohorts: first, a prevalent CAD cohort used to train predictive models for cross-sectional prediction at baseline and prospective estimation of contributing risk factor levels and diagnoses (baseline models) and, second, an incident CAD cohort using, in part, these baseline models as meta-features to train a final CAD incident risk prediction model. The resultant 10-year incident CAD risk model, composed of 15 derived meta-features with multiple embedded polygenic risk scores, achieves an area under the curve of 0.84. In an independent test cohort from the All of Us research program, this model achieved an area under the curve of 0.81 for predicting 10-year incident CAD risk, outperforming standard clinical scores and previously developed integrative models. Moreover, this framework enables the generation of individualized risk reduction profiles by quantifying the potential impact of standard clinical interventions. Notably, genetic risk influences the extent to which these interventions reduce overall CAD risk, allowing for tailored prevention strategies.
Collapse
Affiliation(s)
- Shang-Fu Chen
- Scripps Research Translational Institute, La Jolla, CA, USA
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, CA, USA
| | - Sang Eun Lee
- Department of Cardiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Hossein Javedani Sadaei
- Scripps Research Translational Institute, La Jolla, CA, USA
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, CA, USA
| | - Jun-Bean Park
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Cardiovascular Center, Seoul National University Hospital, Seoul, Republic of Korea
| | - Ahmed Khattab
- Scripps Research Translational Institute, La Jolla, CA, USA
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, CA, USA
| | - Jei-Fu Chen
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Corneliu Henegar
- Scripps Research Translational Institute, La Jolla, CA, USA
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, CA, USA
| | - Nathan E Wineinger
- Scripps Research Translational Institute, La Jolla, CA, USA
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, CA, USA
| | - Evan D Muse
- Scripps Research Translational Institute, La Jolla, CA, USA
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, CA, USA
- Scripps Clinic, La Jolla, CA, USA
| | - Ali Torkamani
- Scripps Research Translational Institute, La Jolla, CA, USA.
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, CA, USA.
| |
Collapse
|
13
|
Omran TZ, Jasmi FSOA, Obaid KM, Ghareeb AKR, Alsailawi HA, Mudhafar M. The interleukin gene landscape: understanding its influence on inflammatory mechanisms in apical periodontitis. Mol Biol Rep 2025; 52:365. [PMID: 40192910 DOI: 10.1007/s11033-025-10477-4] [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: 01/30/2025] [Accepted: 03/27/2025] [Indexed: 04/23/2025]
Abstract
Apical periodontitis is a common inflammatory illness caused by microbial infections in the root canal system, which destroys the periapical tissue. This disease's course and severity are highly regulated by a complex interaction of host immunological responses and genetic variables, particularly interleukin (IL) gene polymorphisms. These genetic variants influence cytokine production, the inflammatory cascade, and the ability to resolve infections. Polymorphisms in important cytokines (e.g., IL-1β, IL-6, IL-10, TNF-α, and IL-17) have been linked to worsening or reducing inflammation, affecting the clinical presentation and chronicity of apical periodontitis. A thorough examination of the molecular and clinical consequences of interleukin polymorphisms in apical periodontitis is given in this article. It emphasizes their function in regulating bone resorption, tissue degradation, and immune cell signaling. Their value in enhancing diagnostic precision, forecasting disease susceptibility, and directing treatment approaches is demonstrated by the incorporation of genetic insights into clinical practice. Targeted therapies, like immunomodulatory drugs and cytokine inhibitors, have great potential to reduce inflammation and encourage periapical healing. Future studies should focus on population-based research to examine genetic variability across ethnic groups, functional investigations to clarify the mechanisms behind polymorphism-driven cytokine regulation, and longitudinal studies to evaluate illness trajectories. Furthermore, developments in precision medicine and bioinformatics could completely transform patient-specific strategies by providing customized treatments and diagnostics. This review highlights the necessity of a multidisciplinary strategy that integrates immunology, genetics, and clinical practice to maximize apical periodontitis therapy and enhance dental health outcomes worldwide.
Collapse
Affiliation(s)
- Tuqa Z Omran
- Department of Basic Sciences, College of Dentistry, University of Kerbala, Karbala, 56001, Iraq
| | | | - Kawthar Mahdi Obaid
- College of Dentistry, Al-Ameed University, Najaf Highway Front of Pole (1238), Karbala, Iraq
| | - Ammr Kareem Rashid Ghareeb
- Department of Medical Physics, Faculty of Medical Applied Sciences, University of Kerbala, Karbala, Karbala, 56001, Iraq
| | - Hasan Ali Alsailawi
- Department of Basic Sciences, College of Dentistry, University of Kerbala, Karbala, 56001, Iraq
- Department of Anesthesia Techniques, AlSafwa University College, Karbala, Iraq
| | - Mustafa Mudhafar
- Department of Medical Physics, Faculty of Medical Applied Sciences, University of Kerbala, Karbala, Karbala, 56001, Iraq.
- Department of Anesthesia Techniques and Intensive Care, Al-Taff University College, Kerbala, 56001, Iraq.
| |
Collapse
|
14
|
Rios Coronado PE, Zhou J, Fan X, Zanetti D, Naftaly JA, Prabala P, Martínez Jaimes AM, Farah EN, Kundu S, Deshpande SS, Evergreen I, Kho PF, Ma Q, Hilliard AT, Abramowitz S, Pyarajan S, Dochtermann D, Damrauer SM, Chang KM, Levin MG, Winn VD, Paşca AM, Plomondon ME, Waldo SW, Tsao PS, Kundaje A, Chi NC, Clarke SL, Red-Horse K, Assimes TL. CXCL12 drives natural variation in coronary artery anatomy across diverse populations. Cell 2025; 188:1784-1806.e22. [PMID: 40049164 PMCID: PMC12029448 DOI: 10.1016/j.cell.2025.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 10/22/2024] [Accepted: 02/06/2025] [Indexed: 03/12/2025]
Abstract
Coronary arteries have a specific branching pattern crucial for oxygenating heart muscle. Among humans, there is natural variation in coronary anatomy with respect to perfusion of the inferior/posterior left heart, which can branch from either the right arterial tree, the left, or both-a phenotype known as coronary dominance. Using angiographic data for >60,000 US veterans of diverse ancestry, we conducted a genome-wide association study of coronary dominance, revealing moderate heritability and identifying ten significant loci. The strongest association occurred near CXCL12 in both European- and African-ancestry cohorts, with downstream analyses implicating effects on CXCL12 expression. We show that CXCL12 is expressed in human fetal hearts at the time dominance is established. Reducing Cxcl12 in mice altered coronary dominance and caused septal arteries to develop away from Cxcl12 expression domains. These findings indicate that CXCL12 patterns human coronary arteries, paving the way for "medical revascularization" through targeting developmental pathways.
Collapse
Affiliation(s)
| | - Jiayan Zhou
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA; VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Xiaochen Fan
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Daniela Zanetti
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA; VA Palo Alto Health Care System, Palo Alto, CA, USA; Institute of Genetic and Biomedical Research, National Research Council, Cagliari, Sardinia, Italy
| | | | - Pratima Prabala
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Azalia M Martínez Jaimes
- Department of Biology, Stanford University, Stanford, CA, USA; Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Elie N Farah
- Department of Medicine, Division of Cardiology, University of California, San Diego, La Jolla, CA, USA
| | - Soumya Kundu
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA; Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Salil S Deshpande
- Institute for Computational and Mathematical Engineering, Stanford University School of Medicine, Stanford, CA, USA
| | - Ivy Evergreen
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Pik Fang Kho
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Qixuan Ma
- Department of Medicine, Division of Cardiology, University of California, San Diego, La Jolla, CA, USA
| | | | - Sarah Abramowitz
- Department of Medicine, Division of Cardiovascular Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Sarnoff Cardiovascular Research Foundation, McLean, VA, USA; Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Saiju Pyarajan
- Center for Data and Computational Sciences, VA Boston Healthcare System, Boston, MA, USA
| | - Daniel Dochtermann
- Center for Data and Computational Sciences, VA Boston Healthcare System, Boston, MA, USA
| | - Scott M Damrauer
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA; Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Kyong-Mi Chang
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA; Department of Medicine, Division of Gastroenterology and Hepatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Michael G Levin
- Department of Medicine, Division of Cardiovascular Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
| | - Virginia D Winn
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| | - Anca M Paşca
- Department of Pediatrics, Neonatology, Stanford University School of Medicine, Stanford, CA, USA
| | - Mary E Plomondon
- Department of Medicine, Rocky Mountain Regional VA Medical Center, Aurora, CO, USA; CART Program, VHA Office of Quality and Patient Safety, Washington, DC, USA
| | - Stephen W Waldo
- Department of Medicine, Rocky Mountain Regional VA Medical Center, Aurora, CO, USA; CART Program, VHA Office of Quality and Patient Safety, Washington, DC, USA; Division of Cardiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Philip S Tsao
- VA Palo Alto Health Care System, Palo Alto, CA, USA; Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA; Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Anshul Kundaje
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA; Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Neil C Chi
- Department of Medicine, Division of Cardiology, University of California, San Diego, La Jolla, CA, USA
| | - Shoa L Clarke
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA; VA Palo Alto Health Care System, Palo Alto, CA, USA; Department of Medicine, Stanford Prevention Research Center, Stanford University School of Medicine, Stanford, CA, USA
| | - Kristy Red-Horse
- Department of Biology, Stanford University, Stanford, CA, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA.
| | - Themistocles L Assimes
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA; VA Palo Alto Health Care System, Palo Alto, CA, USA; Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA; Department of Epidemiology and Population Health, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
15
|
Sun Q, Horimoto ARVR, Chen B, Ockerman F, Mohlke KL, Blue E, Raffield LM, Li Y. Opportunities and challenges of local ancestry in genetic association analyses. Am J Hum Genet 2025; 112:727-740. [PMID: 40185073 DOI: 10.1016/j.ajhg.2025.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 03/05/2025] [Accepted: 03/05/2025] [Indexed: 04/07/2025] Open
Abstract
Recently, admixed populations make up an increasing percentage of the US and global populations, and the admixture is not uniform over space or time or across genomes. Therefore, it becomes indispensable to evaluate local ancestry in addition to global ancestry to improve genetic epidemiological studies. Recent advances in representing human genome diversity, coupled with large-scale whole-genome sequencing initiatives and improved tools for local ancestry inference, have enabled studies to demonstrate that incorporating local ancestry information enhances both genetic association analyses and polygenic risk predictions. Along with the opportunities that local ancestry provides, there exist challenges preventing its full usage in genetic analyses. In this review, we first summarize methods for local ancestry inference and illustrate how local ancestry can be utilized in various analyses, including admixture mapping, association testing, and polygenic risk score construction. In addition, we discuss current challenges in research involving local ancestry, both in terms of the inference itself and its role in genetic association studies. We further pinpoint some future study directions and methodology development opportunities to help more effectively incorporate local ancestry in genetic analyses. It is worth the effort to pursue those future directions and address these analytical challenges because the appropriate use of local ancestry estimates could help mitigate inequality in genomic medicine and improve our understanding of health and disease outcomes.
Collapse
Affiliation(s)
- Quan Sun
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Center for Computational and Genomic Medicine, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA.
| | - Andrea R V R Horimoto
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Brian Chen
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Frank Ockerman
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Elizabeth Blue
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA 98195, USA; Brotman Baty Institute, Seattle, WA 98195, USA
| | - Laura M Raffield
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yun Li
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
16
|
Perry RN, Lenert G, Benavente ED, Ma A, Barbera N, Mokry M, de Kleijn DPV, de Winther MPJ, Mayr M, Björkegren JLM, den Ruijter HM, Civelek M. Female-biased vascular smooth muscle cell gene regulatory networks predict MYH9 as a key regulator of fibrous plaque phenotype. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.28.645955. [PMID: 40236025 PMCID: PMC11996327 DOI: 10.1101/2025.03.28.645955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Atherosclerosis, a chronic inflammatory condition driving coronary artery disease (CAD), manifests in two primary plaque types: unstable atheromatous plaques and stable fibrous plaques. While significant research has focused on atheromatous plaques, recent studies emphasize the growing importance of fibrous plaques, particularly in females under 50 years of age, where erosion on fibrous plaques significantly contributes to coronary thrombosis. The molecular mechanisms underlying sex differences in atherosclerotic plaque characteristics, including vascular smooth muscle cell (VSMC) contributions, remain understudied. Therefore, we utilized sex-specific gene regulatory networks (GRNs) derived from VSMC gene expression data from 119 male and 32 female heart transplant donors to identify molecular drivers of fibrous plaques. GRN analysis revealed two female-biased networks in VSMC, GRN floralwhite and GRN yellowgreen , enriched for inflammatory signaling and actin remodeling pathways, respectively. Single-cell RNA sequencing of carotid plaques from female and male patients confirmed the sex specificity of these networks in VSMCs. Further sub cellular phenotyping of the single-cell RNA sequencing revealed a sex-specific gene expression signature within GRN yellowgreen for VSMCs enriched for contractile and vasculature development pathways. Bayesian network modeling of the GRN yellowgreen identified MYH9 as a key driver gene. Indeed, elevated MYH9 protein expression in atherosclerotic plaques was associated with higher smooth muscle cell content and lower lipid content in female plaques, suggesting its involvement in fibrous plaque formation. Further proteomic analysis confirmed MYH9's upregulation in female fibrous plaques only and its correlation with stable plaque features. These findings provide novel insights into sex-specific molecular mechanisms regulating fibrous plaque formation.
Collapse
|
17
|
Naderian M, Hamed ME, Vaseem AA, Norland K, Dikilitas O, Teymourzadeh A, Bailey KR, Kullo IJ. Effect of Disclosing a Polygenic Risk Score for Coronary Heart Disease on Adverse Cardiovascular Events. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2025; 18:e004968. [PMID: 40151934 DOI: 10.1161/circgen.124.004968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 02/13/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND In the Myocardial Infarction Genes clinical trial (URL: https://www.clinicaltrials.gov; Unique identifier: NCT01936675), participants at intermediate risk of coronary heart disease (CHD) were randomized to receive a Framingham risk score (Framingham risk score group, n=103) or an integrated risk score (integrated risk score group [IRSg], n=104) that additionally included a polygenic risk score. After 6 months, IRSg participants had higher statin initiation and lower low-density lipoprotein cholesterol. We conducted a post hoc 10-year follow-up analysis to investigate whether disclosure of a polygenic risk score for CHD was associated with a reduction in major adverse cardiovascular events (MACE). METHODS Participants were followed from randomization in October 2013 to September 2023 to ascertain MACE, testing for CHD, and changes in risk factors. The primary outcome was time to first MACE, defined as cardiovascular death, nonfatal myocardial infarction, coronary revascularization, and nonfatal stroke. Statistical analyses included Cox proportional hazards regression and linear mixed-effects models. RESULTS We followed all participants who completed the trial, 100 in Framingham risk score group and 103 in IRSg (mean age at the end of follow-up, 68.2±5.2; 48% male). During a median follow-up of 9.5 years, 9 MACEs occurred in Framingham risk score group and 2 in IRSg (hazard ratio, 0.20 [95% CI, 0.04-0.94]; P=0.042). In Framingham risk score group, 47 (47%) underwent at least 1 diagnostic test for CHD, compared with 30 (29%) in IRSg (hazard ratio, 0.51 [95% CI, 0.32-0.81]; P=0.004). A higher proportion of IRSg participants were on statin therapy during the first 4 years postrandomization and had a greater reduction in low-density lipoprotein cholesterol for up to 3 years postrandomization. No significant differences were observed between 2 groups in other traditional cardiovascular risk factors during follow-up. CONCLUSIONS Disclosure of an integrated risk score that included a polygenic risk score to individuals at intermediate risk for CHD was associated with lower MACE incidence after 10 years, likely due to higher statin initiation, leading to lower low-density lipoprotein cholesterol levels.
Collapse
Affiliation(s)
- Mohammadreza Naderian
- Department of Cardiovascular Medicine (M.N., M.E.H., A.A.V., K.N., A.T., I.J.K.), Mayo Clinic, Rochester, MN
| | - Marwan E Hamed
- Department of Cardiovascular Medicine (M.N., M.E.H., A.A.V., K.N., A.T., I.J.K.), Mayo Clinic, Rochester, MN
| | - Ali A Vaseem
- Department of Cardiovascular Medicine (M.N., M.E.H., A.A.V., K.N., A.T., I.J.K.), Mayo Clinic, Rochester, MN
| | - Kristjan Norland
- Department of Cardiovascular Medicine (M.N., M.E.H., A.A.V., K.N., A.T., I.J.K.), Mayo Clinic, Rochester, MN
| | - Ozan Dikilitas
- Department of Internal Medicine (O.D.), Mayo Clinic, Rochester, MN
| | - Azin Teymourzadeh
- Department of Cardiovascular Medicine (M.N., M.E.H., A.A.V., K.N., A.T., I.J.K.), Mayo Clinic, Rochester, MN
| | - Kent R Bailey
- Department of Quantitative Health Sciences (K.R.B.), Mayo Clinic, Rochester, MN
| | - Iftikhar J Kullo
- Department of Cardiovascular Medicine (M.N., M.E.H., A.A.V., K.N., A.T., I.J.K.), Mayo Clinic, Rochester, MN
- Gonda Vascular Center (I.J.K.), Mayo Clinic, Rochester, MN
| |
Collapse
|
18
|
Wadström BN, Borges MC, Wulff AB, Smith GD, Sanderson E, Nordestgaard BG. Elevated Remnant and LDL Cholesterol and the Risk of Peripheral Artery Disease: A Mendelian Randomization Study. J Am Coll Cardiol 2025; 85:1353-1368. [PMID: 40139892 DOI: 10.1016/j.jacc.2024.12.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 12/16/2024] [Accepted: 12/21/2024] [Indexed: 03/29/2025]
Abstract
BACKGROUND Elevated remnant cholesterol and low-density lipoprotein (LDL) cholesterol both increase the risk of coronary artery disease (CAD), but it is not known if the same is true for peripheral artery disease (PAD). OBJECTIVES This study tested the hypothesis that elevated remnant cholesterol and LDL cholesterol, each independent of the other, have causal effects on risk of PAD. METHODS The authors constructed genetic scores from variants near genes known to directly affect levels of remnant cholesterol and LDL cholesterol, identified through a genome-wide association study of individuals in the UK Biobank. Univariable (remnant cholesterol and LDL cholesterol genetic scores separately) and multivariable (remnant cholesterol and LDL cholesterol genetic scores combined) Mendelian randomization were used to estimate the causal effects of higher remnant cholesterol and LDL cholesterol levels on ORs for PAD (n = 38,414 cases and 758,308 controls) and CAD (n = 221,445 cases and 770,615 controls). RESULTS Increments in remnant and LDL genetic scores corresponding to 1 mmol/L (39 mg/dL) higher remnant and LDL cholesterol, respectively, were associated with univariable ORs for PAD of 2.72 (95% CI: 2.10-3.52) and 1.37 (95% CI: 1.25-1.51); corresponding multivariable ORs were 2.16 (95% CI: 1.49-3.12) and 1.14 (95% CI: 1.00-1.30). For CAD, corresponding univariable ORs were 2.92 (95% CI: 2.34-3.64) and 1.67 (95% CI: 1.56-1.79), whereas multivariable ORs were 1.86 (95% CI: 1.39-2.47) and 1.44 (95% CI: 1.29-1.60). Scaled to 1 SD increments in remnant cholesterol and LDL cholesterol, corresponding univariable ORs were 1.37 (95% CI: 1.27-1.49) and 1.29 (95% CI: 1.20-1.39) for PAD, and 1.40 (95% CI: 1.31-1.51) and 1.51 (95% CI: 1.43-1.59) for CAD; corresponding multivariable ORs were 1.28 (95% CI: 1.14-1.43) and 1.11 (95% CI: 1.00-1.23) for PAD, and 1.22 (95% CI: 1.11-1.33) and 1.34 (95% CI: 1.23-1.46) for CAD. CONCLUSIONS Elevated remnant cholesterol had a causal effect on risk of PAD even after accounting for elevated LDL cholesterol, whereas most of the causal effect of elevated LDL cholesterol on risk of PAD was dependent on simultaneously elevated remnant cholesterol. These results indicate that remnant cholesterol may be the major cholesterol fraction responsible for increased risk of PAD. Future studies should investigate the biological mechanisms behind these findings to find improved therapies for prevention and treatment of PAD.
Collapse
Affiliation(s)
- Benjamin Nilsson Wadström
- Medical Research Council (MRC) Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom; Department of Clinical Biochemistry, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark; The Copenhagen General Population Study, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Maria Carolina Borges
- Medical Research Council (MRC) Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom; Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Anders Berg Wulff
- Department of Clinical Biochemistry, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark; The Copenhagen General Population Study, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - George Davey Smith
- Medical Research Council (MRC) Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom; Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom; National Institute for Health Research (NIHR), Biomedical Research Centre, University of Bristol, Bristol, United Kingdom
| | - Eleanor Sanderson
- Medical Research Council (MRC) Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom; Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Børge Grønne Nordestgaard
- Department of Clinical Biochemistry, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark; The Copenhagen General Population Study, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
19
|
Pepin ME, Schwartzman WE, Fang S, Vellarikkal SK, Atri DS, Reddy A, Xu Q, Hamel AR, Billaud M, Segrè AV, Gupta RM. Integrative analysis of single-cell transcriptomics and genetic associations identify cell states associated with vascular disease. Atherosclerosis 2025; 403:119108. [PMID: 40120433 DOI: 10.1016/j.atherosclerosis.2025.119108] [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: 09/04/2024] [Revised: 11/06/2024] [Accepted: 01/08/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Vascular diseases are accompanied by alterations in cellular phenotypes which underlie disease pathogenesis, with single-cell technologies aiding in the discovery of cellular heterogeneity among endothelial cell (EC) and vascular smooth muscle cell (VSMC) populations. In atherosclerotic disease, VSMCs are hypothesized to transition between contractile and synthetic states; however, the specific vascular subpopulations and intermediate cell states responsible for early vascular dysfunction remain unclear. METHODS We integrated newly generated and published single-nuclear RNA-sequencing (snRNA-seq) datasets to analyze normal (n = 7), aneurysmal (n = 9), and atherosclerotic (n = 2) flash-frozen human ascending thoracic aortas. Cell types and subtypes were defined using both top marker genes and canonical gene markers. Disease enrichment and relevant cell types were identified using newly developed computational tools to integrate GWAS data from multiple vascular disease-relevant studies with the single nuclei aortic expression profiles. RESULTS Nuclear dissociation and snRNA-seq identified ten distinct transcriptomic clusters from the integrated analysis representing all major vascular cell populations. Three distinct VSMC populations emerged that exhibited differential expression of extracellular matrix, contractile and pro-proliferative genes. Aneurysmal specimens were enriched for one fibroblast and one VSMC subpopulation compared to healthy tissue. RNA-trajectory analysis inferred a phenotypic continuum of gene expression between VSMC A and VSMC B or C and between two of the identified fibroblast types. VSMCs and Fibroblast C exhibited the greatest cell type-specific enrichment of genes mapped to GWAS loci for coronary artery disease (CAD), blood pressure, and migraine. Cell type-specific enrichment scores were more robust among the transcriptional profiles from non-diseased vascular tissue. CONCLUSIONS Our use of single-cell isolation and new computational methods prioritizes the cell types that most contribute to vascular disease pathogenesis. Specifically, tissue dissociation and single-nuclear transcriptomics better represent all vascular cell types, from which we demonstrate enrichment of pro-proliferative VSMCs in TAA and further implicate phenotypic switching as a likely pathologic mechanism. Integrated analysis of cell-specific gene expression and vascular disease GWAS data implicate genes and pathways associated with fibroblast and VSMC cell-state transitions.
Collapse
MESH Headings
- Humans
- Single-Cell Analysis
- Transcriptome
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Endothelial Cells/metabolism
- Endothelial Cells/pathology
- Gene Expression Profiling/methods
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Phenotype
- Atherosclerosis/genetics
- Atherosclerosis/pathology
- Atherosclerosis/metabolism
- Male
- Aorta, Thoracic/pathology
- Aorta, Thoracic/metabolism
- Female
- Aortic Aneurysm, Thoracic/genetics
- Aortic Aneurysm, Thoracic/pathology
- Aortic Aneurysm, Thoracic/metabolism
- Genome-Wide Association Study
- RNA-Seq
Collapse
Affiliation(s)
- Mark E Pepin
- The Broad Institute of Harvard and MIT, Cambridge, MA, USA; Divisions of Genetics and Cardiovascular Medicine, Brigham & Women's Hospital, Boston, MA, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - William E Schwartzman
- The Broad Institute of Harvard and MIT, Cambridge, MA, USA; Divisions of Genetics and Cardiovascular Medicine, Brigham & Women's Hospital, Boston, MA, USA
| | - Shi Fang
- The Broad Institute of Harvard and MIT, Cambridge, MA, USA; Divisions of Genetics and Cardiovascular Medicine, Brigham & Women's Hospital, Boston, MA, USA
| | - Shamsudheen K Vellarikkal
- The Broad Institute of Harvard and MIT, Cambridge, MA, USA; Divisions of Genetics and Cardiovascular Medicine, Brigham & Women's Hospital, Boston, MA, USA
| | - Deepak S Atri
- The Broad Institute of Harvard and MIT, Cambridge, MA, USA; Divisions of Genetics and Cardiovascular Medicine, Brigham & Women's Hospital, Boston, MA, USA
| | - Ankith Reddy
- Divisions of Genetics and Cardiovascular Medicine, Brigham & Women's Hospital, Boston, MA, USA
| | - Qiaohan Xu
- Ocular Genomics Institute, Department of Ophthalmology, Massachusetts Eye and Ear, Boston, MA, USA
| | - Andrew R Hamel
- The Broad Institute of Harvard and MIT, Cambridge, MA, USA; Ocular Genomics Institute, Department of Ophthalmology, Massachusetts Eye and Ear, Boston, MA, USA; Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Marie Billaud
- Division of Cardiothoracic Surgery, Brigham & Women's Hospital, Boston, MA, USA
| | - Ayellet V Segrè
- The Broad Institute of Harvard and MIT, Cambridge, MA, USA; Ocular Genomics Institute, Department of Ophthalmology, Massachusetts Eye and Ear, Boston, MA, USA; Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Rajat M Gupta
- The Broad Institute of Harvard and MIT, Cambridge, MA, USA; Divisions of Genetics and Cardiovascular Medicine, Brigham & Women's Hospital, Boston, MA, USA.
| |
Collapse
|
20
|
Patel S, Bull L, Salimi K, Shui AM, Siao K, Yang B, Maher JJ, Khalili M. Exploring the impact of graded alcohol use on atherogenic lipid profiles among Latinos with underlying chronic liver disease. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2025; 49:792-803. [PMID: 40022301 DOI: 10.1111/acer.70010] [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: 11/10/2024] [Accepted: 01/31/2025] [Indexed: 03/03/2025]
Abstract
BACKGROUND Alcohol use and hepatitis C virus (HCV) often coexist and are associated with cardiovascular disease. One of the underlying drivers is dyslipidemia. We assessed lipid and lipoprotein levels and the relationship between alcohol use and atherogenic lipid profiles, specifically small dense low-density lipoprotein cholesterol (sdLDL-C), in Latinos with and without HCV. METHODS From June 1, 2002, to January 1, 2016, 150 Latino adults underwent demographic, clinical, metabolic, lipid/lipoprotein, and genetic evaluations. Linear regression (adjusted for age, sex, and recent alcohol use) assessed factors associated with sdLDL-C. RESULTS Participant characteristics were as follows: median age 44 years, 64% male, 39% HCV+, and alcohol use in the last 12 months was 19% heavy and 47% moderate. Ancestries were as follows: 52% European, 40% Native American (NA), and 4.3% African. 29% had non-CC PNPLA3, 89% non-CC TM6SF2, and 73% non-CC IL-28b genotypes. High-density lipoprotein (HDL) cholesterol, HDL-3, apolipoprotein A-1, and lipoprotein-associated phospholipase A2 levels differed by alcohol use groups (p < 0.05). On multivariable analysis, female sex (est. -6.08, p < 0.001), HCV+ status (est. -8.49, p < 0.001), and heavy alcohol use (vs. none) (est. -4.32, p = 0.03) were associated with lower, while NA ancestry (est. 0.92; p = 0.01) and adipose tissue insulin resistance (est. 3.30, p < 0.001) were associated with higher sdLDL-C levels. The positive association between NA ancestry and sdLDL-C was dampened by the presence of a non-CC IL28b genotype (interaction est. -1.95, p = 0.01). CONCLUSIONS In this Latino cohort, ancestry and metabolic dysfunction, independent of alcohol use and HCV, were associated with atherogenic risk. In addition to HCV treatment in this population, cardiometabolic health should be optimized.
Collapse
Affiliation(s)
- Shyam Patel
- Department of Medicine, California Pacific Medical Center, San Francisco, California, USA
| | - Laura Bull
- Institute for Human Genetics, University of California, San Francisco, San Francisco, California, USA
- Division of Gastroenterology and Hepatology, Department of Medicine, University of California, San Francisco, San Francisco, California, USA
- UCSF Liver Center, University of California, San Francisco, San Francisco, California, USA
| | - Kian Salimi
- Division of Gastroenterology and Hepatology, Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Amy M Shui
- UCSF Liver Center, University of California, San Francisco, San Francisco, California, USA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California, USA
| | - Kevin Siao
- Division of Gastroenterology and Hepatology, Department of Medicine, University of California, San Francisco, San Francisco, California, USA
- UCSF Liver Center, University of California, San Francisco, San Francisco, California, USA
| | - Bokun Yang
- Division of Gastroenterology and Hepatology, Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Jacquelyn J Maher
- Division of Gastroenterology and Hepatology, Department of Medicine, University of California, San Francisco, San Francisco, California, USA
- UCSF Liver Center, University of California, San Francisco, San Francisco, California, USA
| | - Mandana Khalili
- Division of Gastroenterology and Hepatology, Department of Medicine, University of California, San Francisco, San Francisco, California, USA
- UCSF Liver Center, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
21
|
Sanin V, Schmieder RS, Koenig W, Li L, Schunkert H, Chen Z. [Role of genetics in precision medicine of coronary artery disease]. Herz 2025; 50:79-87. [PMID: 40019575 DOI: 10.1007/s00059-025-05297-y] [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] [Accepted: 02/03/2025] [Indexed: 03/01/2025]
Abstract
Coronary artery disease (CAD) develops multifactorially through an interplay of lifestyle, environmental and genetic factors. Smoking, hypertension, hyperlipidemia, obesity and diabetes mellitus are modifiable risk factors for CAD. In addition, both rare mutations and multiple frequently occurring genetic variants can cause CAD, whereby the heritability of CAD is ca. 50%. Genetic diagnostics enable the early identification of affected children and adults and, based on a greatly increased cardiovascular risk, initiation of preventive treatment. In recent years, genome-wide association studies have identified hundreds of significant variants that together greatly increase the risk of CAD. In the general population the many frequently occurring risk alleles in combination with modifiable risk factors result in a widespread genetic predisposition to CAD. Their relevance arises in the context of an integrative risk assessment, whereby the additional genetic risk can be calculated by polygenic risk scores (PRS), which provide a hazard ratio that can be multiplied with the clinically determined risk. This overview article discusses the diagnostic principles of rare and frequent genetic causes of CAD as well as their implications in the precision treatment of the disease.
Collapse
Affiliation(s)
- V Sanin
- Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Universitätsklinikum der Technischen Universität München, Lazarettstr. 36, 80636, München, Deutschland
- Deutsches Zentrum für Herz- und Kreislauf-Forschung (DZHK) e. V. (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, München, Deutschland
| | - R S Schmieder
- Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Universitätsklinikum der Technischen Universität München, Lazarettstr. 36, 80636, München, Deutschland
| | - W Koenig
- Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Universitätsklinikum der Technischen Universität München, Lazarettstr. 36, 80636, München, Deutschland
- Deutsches Zentrum für Herz- und Kreislauf-Forschung (DZHK) e. V. (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, München, Deutschland
| | - L Li
- Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Universitätsklinikum der Technischen Universität München, Lazarettstr. 36, 80636, München, Deutschland
| | - H Schunkert
- Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Universitätsklinikum der Technischen Universität München, Lazarettstr. 36, 80636, München, Deutschland.
- Deutsches Zentrum für Herz- und Kreislauf-Forschung (DZHK) e. V. (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, München, Deutschland.
| | - Z Chen
- Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Universitätsklinikum der Technischen Universität München, Lazarettstr. 36, 80636, München, Deutschland
- Deutsches Zentrum für Herz- und Kreislauf-Forschung (DZHK) e. V. (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, München, Deutschland
| |
Collapse
|
22
|
Lee DSM, Cardone KM, Zhang DY, Tsao NL, Abramowitz S, Sharma P, DePaolo JS, Conery M, Aragam KG, Biddinger K, Dilitikas O, Hoffman-Andrews L, Judy RL, Khan A, Kullo IJ, Puckelwartz MJ, Reza N, Satterfield BA, Singhal P, Arany Z, Cappola TP, Carruth ED, Day SM, Do R, Haggerty CM, Joseph J, McNally EM, Nadkarni G, Owens AT, Rader DJ, Ritchie MD, Sun YV, Voight BF, Levin MG, Damrauer SM. Common-variant and rare-variant genetic architecture of heart failure across the allele-frequency spectrum. Nat Genet 2025; 57:829-838. [PMID: 40195560 PMCID: PMC12049093 DOI: 10.1038/s41588-025-02140-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 02/21/2025] [Indexed: 04/09/2025]
Abstract
Heart failure is a complex trait, influenced by environmental and genetic factors, affecting over 30 million individuals worldwide. Here we report common-variant and rare-variant association studies of all-cause heart failure and examine how different classes of genetic variation impact its heritability. We identify 176 common-variant risk loci at genome-wide significance in 2,358,556 individuals and cluster these signals into five broad modules based on pleiotropic associations with anthropomorphic traits/obesity, blood pressure/renal function, atherosclerosis/lipids, immune activity and arrhythmias. In parallel, we uncover exome-wide significant associations for heart failure and rare predicted loss-of-function variants in TTN, MYBPC3, FLNC and BAG3 using exome sequencing of 376,334 individuals. We find that total burden heritability of rare coding variants is highly concentrated in a small set of Mendelian cardiomyopathy genes, while common-variant heritability is diffusely spread throughout the genome. Finally, we show that common-variant background modifies heart failure risk among carriers of rare pathogenic truncating variants in TTN. Together, these findings discern genetic links between dysregulated metabolism and heart failure and highlight a polygenic component to heart failure not captured by current clinical genetic testing.
Collapse
Affiliation(s)
- David S M Lee
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Kathleen M Cardone
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - David Y Zhang
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Noah L Tsao
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Sarah Abramowitz
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Pranav Sharma
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - John S DePaolo
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Mitchell Conery
- Genomics and Computational Biology Graduate Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Krishna G Aragam
- Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kiran Biddinger
- Program in Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ozan Dilitikas
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Lily Hoffman-Andrews
- Division of Cardiovascular Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Renae L Judy
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Atlas Khan
- Division of Nephrology, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York City, NY, USA
| | - Iftikhar J Kullo
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Megan J Puckelwartz
- Department of Pharmacology, Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Nosheen Reza
- Division of Cardiovascular Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | | | - Pankhuri Singhal
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Zoltan Arany
- Division of Cardiovascular Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Cardiovascular Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Thomas P Cappola
- Division of Cardiovascular Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Eric D Carruth
- Department of Genomic Health, Geisinger, Danville, PA, USA
| | - Sharlene M Day
- Division of Cardiovascular Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Cardiovascular Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Ron Do
- The Charles Bronfman Institute for Personalized Medicine, Mount Sinai Icahn School of Medicine, New York City, NY, USA
- BioMe Phenomics Center, Mount Sinai Icahn School of Medicine, New York City, NY, USA
- Department of Genetics and Genomic Sciences, Mount Sinai Icahn School of Medicine, New York City, NY, USA
| | | | - Jacob Joseph
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Massachusetts Veterans Epidemiology Research and Information Center, VA Boston Healthcare System, Boston, MA, USA
| | - Elizabeth M McNally
- Center for Genetic Medicine, Bluhm Cardiovascular Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Girish Nadkarni
- Division of Nephrology, Department of Medicine, Mount Sinai Icahn School of Medicine, New York City, NY, USA
| | - Anjali T Owens
- Cardiovascular Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Daniel J Rader
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Division of Translational Medicine and Human Genetics, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Marylyn D Ritchie
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Institute for Biomedical Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Yan V Sun
- Atlanta VA Health Care System, Decatur, GA, USA
- Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA, USA
| | - Benjamin F Voight
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Michael G Levin
- Division of Cardiovascular Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
- Cardiovascular Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA.
| | - Scott M Damrauer
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Cardiovascular Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
| |
Collapse
|
23
|
Gummesson A, Lundmark P, Chen QS, Björnson E, Dekkers KF, Hammar U, Adiels M, Wang Y, Andersson T, Bergström G, Carlhäll CJ, Erlinge D, Jernberg T, Landfors F, Lind L, Mannila M, Melander O, Pirazzi C, Sundström J, Östgren CJ, Gunnarsson C, Orho-Melander M, Söderberg S, Fall T, Gigante B. A genome-wide association study of imaging-defined atherosclerosis. Nat Commun 2025; 16:2266. [PMID: 40164586 PMCID: PMC11958696 DOI: 10.1038/s41467-025-57457-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 02/22/2025] [Indexed: 04/02/2025] Open
Abstract
Imaging-defined atherosclerosis represents an intermediate phenotype of atherosclerotic cardiovascular disease (ASCVD). Genome-wide association studies (GWAS) on directly measured coronary plaques using coronary computed tomography angiography (CCTA) are scarce. In the so far largest population-based cohort with CCTA data, we performed a GWAS on coronary plaque burden as determined by the segment involvement score (SIS) in 24,811 European individuals. We identified 20 significant independent genetic markers for SIS, three of which were found in loci not implicated in ASCVD before. Further GWAS on coronary artery calcification showed similar results to that of SIS, whereas a GWAS on ultrasound-assessed carotid plaques identified both shared and non-shared loci with SIS. In two-sample Mendelian randomization studies using SIS-associated markers in UK Biobank and CARDIoGRAMplusC4D, one extra coronary segment with atherosclerosis corresponded to 1.8-fold increased odds of myocardial infarction. This GWAS data can aid future studies of causal pathways in ASCVD.
Collapse
Affiliation(s)
- Anders Gummesson
- Region Västra Götaland, Sahlgrenska University Hospital, Department of Clinical Genetics and Genomics, Gothenburg, Sweden.
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - Per Lundmark
- Department of Medical Sciences, Molecular Epidemiology, Uppsala University, Uppsala, Sweden
| | - Qiao Sen Chen
- Division of Cardiology, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Elias Björnson
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Koen F Dekkers
- Department of Medical Sciences, Molecular Epidemiology, Uppsala University, Uppsala, Sweden
| | - Ulf Hammar
- Department of Medical Sciences, Molecular Epidemiology, Uppsala University, Uppsala, Sweden
| | - Martin Adiels
- School of Public Health and Community Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Yunzhang Wang
- Department of Clinical Sciences, Danderyd University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Therese Andersson
- Department of Public Medicine and Clinical Health, Umeå University, Umeå, Sweden
| | - Göran Bergström
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Region Västra Götaland, Sahlgrenska University Hospital, Department of Clinical Physiology, Gothenburg, Sweden
| | - Carl-Johan Carlhäll
- Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden
- Department of Clinical Physiology in Linköping, Linköping University, Linköping, Sweden
- Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - David Erlinge
- Department of Clinical Sciences Lund, Cardiology, Lund University, Lund, Sweden
| | - Tomas Jernberg
- Department of Clinical Sciences, Danderyd University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Fredrik Landfors
- Department of Public Medicine and Clinical Health, Umeå University, Umeå, Sweden
| | - Lars Lind
- Department of Medical Sciences, Clinical Epidemiology, Uppsala University, Uppsala, Sweden
| | - Maria Mannila
- Department of Cardiology, Karolinska University Hospital, Stockholm, Sweden
| | - Olle Melander
- Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden
- Department of Emergency and Internal Medicine, Skåne University Hospital, Malmö, Sweden
| | - Carlo Pirazzi
- Region Västra Götaland, Sahlgrenska University Hospital, Department of Cardiology, Gothenburg, Sweden
| | - Johan Sundström
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
- The George Institute for Global Health, University of New South Wales, Sydney, Australia
| | - Carl Johan Östgren
- Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden
- Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - Cecilia Gunnarsson
- Department of Biomedical and Clinical Sciences, Division of Clinical Genetics, Linköping University, Linköping, Sweden
| | | | - Stefan Söderberg
- Department of Public Medicine and Clinical Health, Umeå University, Umeå, Sweden
| | - Tove Fall
- Department of Medical Sciences, Molecular Epidemiology, Uppsala University, Uppsala, Sweden
| | - Bruna Gigante
- Division of Cardiology, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Cardiology, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
24
|
Aminbakhsh AP, Théberge ET, Burden E, Adejumo CK, Gravely AK, Lehman A, Sedlak TL. Exploring associations between estrogen and gene candidates identified by coronary artery disease genome-wide association studies. Front Cardiovasc Med 2025; 12:1502985. [PMID: 40182431 PMCID: PMC11965610 DOI: 10.3389/fcvm.2025.1502985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 03/04/2025] [Indexed: 04/05/2025] Open
Abstract
Introduction Coronary artery disease (CAD) is the leading cause of death around the world, with epidemiological sex and gender differences in prevalence, pathophysiology and outcomes. It has been hypothesized that sex steroids, like estrogen, may contribute to these sex differences. There is a relatively large genetic component to developing CAD, with heritability estimates ranging between 40%-60%. In the last two decades, genome-wide association studies (GWAS) have contributed substantially to advancing the understanding of genetic candidates contributing to CAD. The aim of this study was to determine if genes discovered in CAD GWASs are affected by estrogen via direct modulation or indirect down-stream targets. Methods A scoping review was conducted using MEDLINE and EMBASE for studies of atherosclerotic coronary artery disease and a genome-wide association study (GWAS) design. Analysis was limited to candidate genes with corresponding single nucleotide polymorphisms (SNPs) surpassing genome-wide significance and had been mapped to genes by study authors. The number of studies that conducted sex-stratified analyses with significant genes were quantified. A literature search of the final gene lists was done to examine any evidence suggesting estrogen may modulate the genes and/or gene products. Results There were 60 eligible CAD GWASs meeting inclusion criteria for data extraction. Of these 60, only 36 had genome-wide significant SNPs reported, and only 3 of these had significant SNPs from sex-stratified analyses mapped to genes. From these 36 studies, a total of 61 genes were curated, of which 26 genes (43%) were found to have modulation by estrogen. All 26 were discovered in studies that adjusted for sex. 12/26 genes were also discovered in studies that conducted sex-stratified analyses. 12/26 genes were classified as having a role in lipid synthesis, metabolism and/or lipoprotein mechanisms, while 11/26 were classified as having a role in vascular integrity, and 3/26 were classified as having a role in thrombosis. Discussion This study provides further evidence of the relationship between estrogen, genetic risk and the development of CAD. More sex-stratified research will need to be conducted to further characterize estrogen's relation to sex differences in the pathology and progression of CAD.
Collapse
Affiliation(s)
- Ava P. Aminbakhsh
- Department of Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Emilie T. Théberge
- Department of Medical Genetics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Elizabeth Burden
- Division of Internal Medicine, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
- Vancouver Coastal Health, Vancouver, BC, Canada
| | - Cindy Kalenga Adejumo
- Division of Internal Medicine, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
- Vancouver Coastal Health, Vancouver, BC, Canada
| | - Annabel K. Gravely
- Department of Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Anna Lehman
- Department of Medical Genetics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- Vancouver Coastal Health, Vancouver, BC, Canada
| | - Tara L. Sedlak
- Vancouver Coastal Health, Vancouver, BC, Canada
- Division of Cardiology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
25
|
Marston NA, Kamanu FK, Melloni GEM, Schnitzler G, Hakim A, Ma RX, Kang H, Chasman DI, Giugliano RP, Ellinor PT, Ridker PM, Engreitz JM, Sabatine MS, Ruff CT, Gupta RM. Endothelial cell-related genetic variants identify LDL cholesterol-sensitive individuals who derive greater benefit from aggressive lipid lowering. Nat Med 2025; 31:963-969. [PMID: 40011692 DOI: 10.1038/s41591-025-03533-w] [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: 02/22/2024] [Accepted: 01/24/2025] [Indexed: 02/28/2025]
Abstract
The role of endothelial cell (EC) dysfunction in contributing to an individual's susceptibility to coronary atherosclerosis and how low-density lipoprotein cholesterol (LDL-C) concentrations might modify this relationship have not been previously studied. Here, from an examination of genome-wide significant single nucleotide polymorphisms associated with coronary artery disease (CAD), we identified variants with effects on EC function and constructed a 35 single nucleotide polymorphism polygenic risk score comprising these EC-specific variants (EC PRS). The association of the EC PRS with the risk of incident cardiovascular disease was tested in 3 cohorts: a primary prevention population in the UK Biobank (UKBB; n = 348,967); a primary prevention cohort from a trial that tested a statin (JUPITER, n = 8,749); and a secondary prevention cohort that tested a PCSK9 inhibitor (FOURIER, n = 14,298). In the UKBB, the EC PRS was independently associated with the risk of incident CAD (adjusted hazard ratio (aHR) per 1 s.d. of 1.24 (95% CI 1.21-1.26), P < 2 × 10-16). Moreover, LDL-C concentration significantly modified this risk: the aHR per 1 s.d. was 1.26 (1.22-1.30) when LDL-C was 150 mg dl-1 but 1.00 (0.85-1.16) when LDL-C was 50 mg dl-1 (Pinteraction = 0.004). The clinical benefit of LDL-C lowering was significantly greater in individuals with a high EC PRS than in individuals with low or intermediate EC PRS, with relative risk reductions of 68% (HR 0.32 (0.18-0.59)) versus 29% (HR 0.71 (0.52-0.95)) in the primary prevention cohort (Pinteraction = 0.02) and 33% (HR 0.67 (0.53-0.83)) versus 8% (HR 0.92 (0.82-1.03)) in the secondary prevention cohort (Pinteraction = 0.01). We conclude that EC PRS quantifies an independent axis of CAD risk that is not currently captured in medical practice and identifies individuals who are more sensitive to the atherogenic effects of LDL-C and who would potentially derive substantially greater benefit from aggressive LDL-C lowering.
Collapse
Affiliation(s)
- Nicholas A Marston
- Division of Cardiology, TIMI Study Group, Boston, MA, USA.
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Frederick K Kamanu
- Division of Cardiology, TIMI Study Group, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Giorgio E M Melloni
- Division of Cardiology, TIMI Study Group, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Gavin Schnitzler
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Aaron Hakim
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Rosa X Ma
- BASE Initiative, Betty Irene Moore Children's Heart Center, Lucile Packard Children's Hospital, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Helen Kang
- BASE Initiative, Betty Irene Moore Children's Heart Center, Lucile Packard Children's Hospital, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Computational and Systems Biology PhD Program, MIT, Cambridge, MA, USA
| | - Daniel I Chasman
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Robert P Giugliano
- Division of Cardiology, TIMI Study Group, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Patrick T Ellinor
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Paul M Ridker
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jesse M Engreitz
- BASE Initiative, Betty Irene Moore Children's Heart Center, Lucile Packard Children's Hospital, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Marc S Sabatine
- Division of Cardiology, TIMI Study Group, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Christian T Ruff
- Division of Cardiology, TIMI Study Group, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Rajat M Gupta
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
26
|
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
|
27
|
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
|
28
|
Hecker D, Song X, Baumgarten N, Diagel A, Katsaouni N, Li L, Li S, Maji RK, Behjati Ardakani F, Ma L, Tews D, Wabitsch M, Björkegren JL, Schunkert H, Chen Z, Schulz MH. Cell type-specific epigenetic regulatory circuitry of coronary artery disease loci. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.20.639228. [PMID: 40027824 PMCID: PMC11870499 DOI: 10.1101/2025.02.20.639228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Coronary artery disease (CAD) is the leading cause of death worldwide. Recently, hundreds of genomic loci have been shown to increase CAD risk, however, the molecular mechanisms underlying signals from CAD risk loci remain largely unclear. We sought to pinpoint the candidate causal coding and non-coding genes of CAD risk loci in a cell type-specific fashion. We integrated the latest statistics of CAD genetics from over one million individuals with epigenetic data from 45 relevant cell types to identify genes whose regulation is affected by CAD-associated single nucleotide variants (SNVs) via epigenetic mechanisms. Applying two statistical approaches, we identified 1,580 genes likely involved in CAD, about half of which have not been associated with the disease so far. Enrichment analysis and phenome-wide association studies linked the novel candidate genes to disease-specific pathways and CAD risk factors, corroborating their disease relevance. We showed that CAD-SNVs are enriched to regulate gene expression by affecting the binding of transcription factors (TFs) with cellular specificity. Of all the candidate genes, 23.5% represented non-coding RNAs (ncRNA), which likewise showed strong cell type specificity. We conducted a proof-of-concept biological validation for the novel CAD ncRNA gene IQCH-AS1 . CRISPR/Cas9-based gene knockout of IQCH-AS1 , in a human preadipocyte strain, resulted in reduced preadipocyte proliferation, less adipocyte lipid accumulation, and atherogenic cytokine profile. The cellular data are in line with the reduction of IQCH-AS1 in adipose tissues of CAD patients and the negative impact of risk alleles on its expression, suggesting IQCH-AS1 to be protective for CAD. Our study not only pinpoints CAD candidate genes in a cell type-specific fashion but also spotlights the roles of the understudied ncRNA genes in CAD genetics.
Collapse
Affiliation(s)
- Dennis Hecker
- Department of Medicine, Institute for Computational Genomic Medicine, Goethe University Frankfurt, 60590 Frankfurt, Germany
- Deutsches Zentrum für Herz- und Kreislaufforschung (DZHK), Rhein-Main, Germany
| | - Xiaoning Song
- Department of Cardiology, German Heart Centre Munich, School of Medicine and Health, Technical University of Munich, 80636 Munich, Germany
- DZHK, Partner Site Munich Heart Alliance, Munich, Germany
| | - Nina Baumgarten
- Department of Medicine, Institute for Computational Genomic Medicine, Goethe University Frankfurt, 60590 Frankfurt, Germany
- Deutsches Zentrum für Herz- und Kreislaufforschung (DZHK), Rhein-Main, Germany
| | - Anastasiia Diagel
- Department of Cardiology, German Heart Centre Munich, School of Medicine and Health, Technical University of Munich, 80636 Munich, Germany
- DZHK, Partner Site Munich Heart Alliance, Munich, Germany
| | - Nikoletta Katsaouni
- Department of Medicine, Institute for Computational Genomic Medicine, Goethe University Frankfurt, 60590 Frankfurt, Germany
- Deutsches Zentrum für Herz- und Kreislaufforschung (DZHK), Rhein-Main, Germany
| | - Ling Li
- Department of Cardiology, German Heart Centre Munich, School of Medicine and Health, Technical University of Munich, 80636 Munich, Germany
- DZHK, Partner Site Munich Heart Alliance, Munich, Germany
| | - Shuangyue Li
- Department of Cardiology, German Heart Centre Munich, School of Medicine and Health, Technical University of Munich, 80636 Munich, Germany
- DZHK, Partner Site Munich Heart Alliance, Munich, Germany
| | - Ranjan Kumar Maji
- Department of Medicine, Institute for Computational Genomic Medicine, Goethe University Frankfurt, 60590 Frankfurt, Germany
- Deutsches Zentrum für Herz- und Kreislaufforschung (DZHK), Rhein-Main, Germany
| | - Fatemeh Behjati Ardakani
- Department of Medicine, Institute for Computational Genomic Medicine, Goethe University Frankfurt, 60590 Frankfurt, Germany
- Deutsches Zentrum für Herz- und Kreislaufforschung (DZHK), Rhein-Main, Germany
| | - Lijiang Ma
- Department of Genetics & Genomic Sciences, Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York 10029, USA
| | - Daniel Tews
- German Center for Child and Adolescent Health (DZKJ), Partner Site Ulm
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, 89075 Ulm, Germany
| | - Martin Wabitsch
- German Center for Child and Adolescent Health (DZKJ), Partner Site Ulm
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, 89075 Ulm, Germany
| | - Johan L.M. Björkegren
- Department of Genetics & Genomic Sciences, Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York 10029, USA
- Department of Medicine, Karolinska Institutet, Karolinska Universitetssjukhuset, 14157 Huddinge, Sweden
| | - Heribert Schunkert
- Department of Cardiology, German Heart Centre Munich, School of Medicine and Health, Technical University of Munich, 80636 Munich, Germany
- DZHK, Partner Site Munich Heart Alliance, Munich, Germany
| | - Zhifen Chen
- Department of Cardiology, German Heart Centre Munich, School of Medicine and Health, Technical University of Munich, 80636 Munich, Germany
- DZHK, Partner Site Munich Heart Alliance, Munich, Germany
| | - Marcel H. Schulz
- Department of Medicine, Institute for Computational Genomic Medicine, Goethe University Frankfurt, 60590 Frankfurt, Germany
- Deutsches Zentrum für Herz- und Kreislaufforschung (DZHK), Rhein-Main, Germany
| |
Collapse
|
29
|
Marderstein AR, Kundu S, Padhi EM, Deshpande S, Wang A, Robb E, Sun Y, Yun CM, Pomales-Matos D, Xie Y, Nachun D, Jessa S, Kundaje A, Montgomery SB. Mapping the regulatory effects of common and rare non-coding variants across cellular and developmental contexts in the brain and heart. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.18.638922. [PMID: 40027628 PMCID: PMC11870466 DOI: 10.1101/2025.02.18.638922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Whole genome sequencing has identified over a billion non-coding variants in humans, while GWAS has revealed the non-coding genome as a significant contributor to disease. However, prioritizing causal common and rare non-coding variants in human disease, and understanding how selective pressures have shaped the non-coding genome, remains a significant challenge. Here, we predicted the effects of 15 million variants with deep learning models trained on single-cell ATAC-seq across 132 cellular contexts in adult and fetal brain and heart, producing nearly two billion context-specific predictions. Using these predictions, we distinguish candidate causal variants underlying human traits and diseases and their context-specific effects. While common variant effects are more cell-type-specific, rare variants exert more cell-type-shared regulatory effects, with selective pressures particularly targeting variants affecting fetal brain neurons. To prioritize de novo mutations with extreme regulatory effects, we developed FLARE, a context-specific functional genomic model of constraint. FLARE outperformed other methods in prioritizing case mutations from autism-affected families near syndromic autism-associated genes; for example, identifying mutation outliers near CNTNAP2 that would be missed by alternative approaches. Overall, our findings demonstrate the potential of integrating single-cell maps with population genetics and deep learning-based variant effect prediction to elucidate mechanisms of development and disease-ultimately, supporting the notion that genetic contributions to neurodevelopmental disorders are predominantly rare.
Collapse
Affiliation(s)
- Andrew R. Marderstein
- Department of Pathology, Stanford University, Stanford, CA, USA
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Soumya Kundu
- Department of Computer Science, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Evin M. Padhi
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Salil Deshpande
- Department of Genetics, Stanford University, Stanford, CA, USA
- Institute for Computational and Mathematical Engineering, Stanford University, Stanford, CA, USA
| | - Austin Wang
- Department of Computer Science, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Esther Robb
- Department of Computer Science, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Ying Sun
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Chang M. Yun
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| | | | - Yilin Xie
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Daniel Nachun
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Selin Jessa
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Anshul Kundaje
- Department of Computer Science, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Stephen B. Montgomery
- Department of Pathology, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| |
Collapse
|
30
|
Iyer KR, Clarke SL, Guarischi‐Sousa R, Gjoni K, Heath AS, Young EP, Stitziel NO, Laurie C, Broome JG, Khan AT, Lewis JP, Xu H, Montasser ME, Ashley KE, Hasbani NR, Boerwinkle E, Morrison AC, Chami N, Do R, Rocheleau G, Lloyd‐Jones DM, Lemaitre RN, Bis JC, Floyd JS, Kinney GL, Bowden DW, Palmer ND, Benjamin EJ, Nayor M, Yanek LR, Kral BG, Becker LC, Kardia SLR, Smith JA, Bielak LF, Norwood AF, Min Y, Carson AP, Post WS, Rich SS, Herrington D, Guo X, Taylor KD, Manson JE, Franceschini N, Pollard KS, Mitchell BD, Loos RJF, Fornage M, Hou L, Psaty BM, Young KA, Regan EA, Freedman BI, Vasan RS, Levy D, Mathias RA, Peyser PA, Raffield LM, Kooperberg C, Reiner AP, Rotter JI, Jun G, de Vries PS, Assimes TL. Unveiling the Genetic Landscape of Coronary Artery Disease Through Common and Rare Structural Variants. J Am Heart Assoc 2025; 14:e036499. [PMID: 39950338 PMCID: PMC12074758 DOI: 10.1161/jaha.124.036499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 10/21/2024] [Indexed: 02/17/2025]
Abstract
BACKGROUND Genome-wide association studies have identified several hundred susceptibility single nucleotide variants for coronary artery disease (CAD). Despite single nucleotide variant-based genome-wide association studies improving our understanding of the genetics of CAD, the contribution of structural variants (SVs) to the risk of CAD remains largely unclear. METHOD AND RESULTS We leveraged SVs detected from high-coverage whole genome sequencing data in a diverse group of participants from the National Heart Lung and Blood Institute's Trans-Omics for Precision Medicine program. Single variant tests were performed on 58 706 SVs in a study sample of 11 556 CAD cases and 42 907 controls. Additionally, aggregate tests using sliding windows were performed to examine rare SVs. One genome-wide significant association was identified for a common biallelic intergenic duplication on chromosome 6q21 (P=1.54E-09, odds ratio=1.34). The sliding window-based aggregate tests found 1 region on chromosome 17q25.3, overlapping USP36, to be significantly associated with coronary artery disease (P=1.03E-10). USP36 is highly expressed in arterial and adipose tissues while broadly affecting several cardiometabolic traits. CONCLUSIONS Our results suggest that SVs, both common and rare, may influence the risk of coronary artery disease.
Collapse
Affiliation(s)
- Kruthika R. Iyer
- Data Science and Biotechnology, Gladstone InstitutesSan FranciscoCAUSA
- Department of Medicine, Division of Cardiovascular MedicineStanford University School of MedicineStanfordCAUSA
| | - Shoa L. Clarke
- Department of Medicine, Division of Cardiovascular MedicineStanford University School of MedicineStanfordCAUSA
- Department of Medicine, Stanford Prevention Research CenterStanford University School of MedicineStanfordCAUSA
| | - Rodrigo Guarischi‐Sousa
- Department of Medicine, Division of Cardiovascular MedicineStanford University School of MedicineStanfordCAUSA
| | - Ketrin Gjoni
- Data Science and Biotechnology, Gladstone InstitutesSan FranciscoCAUSA
- Department of Epidemiology and BiostatisticsUniversity of CaliforniaSan FranciscoCAUSA
| | - Adam S. Heath
- Department of Epidemiology, Human Genetics Center, School of Public HealthThe University of Texas Health Science Center at HoustonHoustonTXUSA
| | - Erica P. Young
- Department of Medicine, Division of CardiologyWashington University School of MedicineSaint LouisMOUSA
- McDonnell Genome Institute, Washington University School of MedicineSaint LouisMOUSA
| | - Nathan O. Stitziel
- Department of Medicine, Division of CardiologyWashington University School of MedicineSaint LouisMOUSA
- McDonnell Genome Institute, Washington University School of MedicineSaint LouisMOUSA
- Department of GeneticsWashington University School of MedicineSaint LouisMOUSA
| | - Cecelia Laurie
- Department of BiostatisticsUniversity of WashingtonSeattleWAUSA
| | - Jai G. Broome
- Department of BiostatisticsUniversity of WashingtonSeattleWAUSA
- Department of Medicine, Division of Internal MedicineUniversity of WashingtonSeattleWAUSA
| | - Alyna T. Khan
- Department of BiostatisticsUniversity of WashingtonSeattleWAUSA
| | - Joshua P. Lewis
- Department of MedicineUniversity of Maryland School of MedicineBaltimoreMDUSA
| | - Huichun Xu
- Department of MedicineUniversity of Maryland School of MedicineBaltimoreMDUSA
| | - May E. Montasser
- Department of MedicineUniversity of Maryland School of MedicineBaltimoreMDUSA
| | - Kellan E. Ashley
- Department of MedicineUniversity of Mississippi Medical CenterJacksonMSUSA
| | - Natalie R. Hasbani
- Department of Epidemiology, Human Genetics Center, School of Public HealthThe University of Texas Health Science Center at HoustonHoustonTXUSA
| | - Eric Boerwinkle
- Department of Epidemiology, Human Genetics Center, School of Public HealthThe University of Texas Health Science Center at HoustonHoustonTXUSA
- Human Genome Sequencing CenterBaylor College of MedicineHoustonTXUSA
| | - Alanna C. Morrison
- Department of Epidemiology, Human Genetics Center, School of Public HealthThe University of Texas Health Science Center at HoustonHoustonTXUSA
| | - Nathalie Chami
- The Charles Bronfman Institute for Personalized MedicineIcahn School of Medicine at Mount SinaiNew YorkNYUSA
| | - Ron Do
- The Charles Bronfman Institute for Personalized MedicineIcahn School of Medicine at Mount SinaiNew YorkNYUSA
| | - Ghislain Rocheleau
- The Charles Bronfman Institute for Personalized MedicineIcahn School of Medicine at Mount SinaiNew YorkNYUSA
- Department of Genetics and Genomic SciencesIcahn School of Medicine at Mount SinaiNew YorkNYUSA
| | | | - Rozenn N. Lemaitre
- Department of Medicine, Cardiovascular Health Research UnitUniversity of WashingtonSeattleWAUSA
| | - Joshua C. Bis
- Department of Medicine, Cardiovascular Health Research UnitUniversity of WashingtonSeattleWAUSA
| | - James S. Floyd
- Department of Medicine, Cardiovascular Health Research UnitUniversity of WashingtonSeattleWAUSA
- Department of EpidemiologyUniversity of WashingtonSeattleWAUSA
| | - Gregory L. Kinney
- Department of EpidemiologyColorado School of Public HealthAuroraCOUSA
| | - Donald W. Bowden
- Department of BiochemistryWake Forest University School of MedicineWinston‐SalemNCUSA
| | - Nicholette D. Palmer
- Department of BiochemistryWake Forest University School of MedicineWinston‐SalemNCUSA
| | - Emelia J. Benjamin
- Department of Medicine, Cardiovascular Medicine, Boston Medical CenterBoston University Chobanian & Avedisian School of MedicineBostonMAUSA
- Department of EpidemiologyBoston University School of Public HealthBostonMAUSA
| | - Matthew Nayor
- Department of Medicine, Cardiovascular MedicineBoston University Chobanian & Avedisian School of MedicineBostonMAUSA
- Department of Medicine, Preventive Medicine & EpidemiologyBoston University Chobanian & Avedisian School of MedicineBostonMAUSA
| | - Lisa R. Yanek
- Department of MedicineJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Brian G. Kral
- Department of MedicineJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Lewis C. Becker
- Department of MedicineJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Sharon L. R. Kardia
- Department of EpidemiologyUniversity of Michigan School of Public HealthAnn ArborMIUSA
| | - Jennifer A. Smith
- Department of EpidemiologyUniversity of Michigan School of Public HealthAnn ArborMIUSA
- Institute for Social ResearchSurvey Research Center, University of MichiganAnn ArborMIUSA
| | - Lawrence F. Bielak
- Department of EpidemiologyUniversity of Michigan School of Public HealthAnn ArborMIUSA
| | - Arnita F. Norwood
- Department of MedicineUniversity of Mississippi Medical CenterJacksonMSUSA
| | - Yuan‐I Min
- Department of MedicineUniversity of Mississippi Medical CenterJacksonMSUSA
| | - April P. Carson
- Department of MedicineUniversity of Mississippi Medical CenterJacksonMSUSA
| | - Wendy S. Post
- Department of Medicine, Division of CardiologyJohns Hopkins UniversityBaltimoreMDUSA
| | - Stephen S. Rich
- Department of Genome SciencesUniversity of Virginia School of MedicineCharlottesvilleVAUSA
| | - David Herrington
- Department of MedicineWake Forest University School of MedicineWinston‐SalemNCUSA
| | - Xiuqing Guo
- Department of Pediatrics, The Institute for Translational Genomics and Population SciencesThe Lundquist Institute for Biomedical Innovation at Harbor‐UCLA Medical CenterTorranceCAUSA
| | - Kent D. Taylor
- Department of Pediatrics, The Institute for Translational Genomics and Population SciencesThe Lundquist Institute for Biomedical Innovation at Harbor‐UCLA Medical CenterTorranceCAUSA
| | - JoAnn E. Manson
- Department of MedicineBrigham and Women’s Hospital, Harvard Medical SchoolBostonMAUSA
| | - Nora Franceschini
- Department of EpidemiologyUniversity of North Carolina at Chapel HillChapel HillNCUSA
| | - Katherine S. Pollard
- Data Science and Biotechnology, Gladstone InstitutesSan FranciscoCAUSA
- Department of Epidemiology and BiostatisticsUniversity of CaliforniaSan FranciscoCAUSA
- Chan Zuckerberg BiohubSan FranciscoCAUSA
| | - Braxton D. Mitchell
- Department of MedicineUniversity of Maryland School of MedicineBaltimoreMDUSA
- Geriatric Research and Education Clinical CenterBaltimore Veterans Administration Medical CenterBaltimoreMDUSA
| | - Ruth J. F. Loos
- The Charles Bronfman Institute for Personalized MedicineIcahn School of Medicine at Mount SinaiNew YorkNYUSA
- Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Basic Metabolic ResearchUniversity of CopenhagenCopenhagenDenmark
| | - Myriam Fornage
- Brown Foundation Institute of Molecular MedicineUniversity of Texas Health Science Center at HoustonHoustonTXUSA
| | - Lifang Hou
- Department of Preventive MedicineNorthwestern UniversityChicagoILUSA
| | - Bruce M. Psaty
- Department of Medicine, Cardiovascular Health Research UnitUniversity of WashingtonSeattleWAUSA
- Department of EpidemiologyUniversity of WashingtonSeattleWAUSA
- Department of Health Systems and Population HealthUniversity of WashingtonSeattleWAUSA
| | - Kendra A. Young
- Department of EpidemiologyColorado School of Public HealthAuroraCOUSA
| | | | - Barry I. Freedman
- Department of Internal Medicine, Section on NephrologyWake Forest University School of MedicineWinston‐SalemNCUSA
| | | | - Daniel Levy
- Division of Intramural Research, Population Sciences BranchNational Heart, Lung, and Blood Institute, National Institutes of HealthBethesdaMDUSA
| | - Rasika A. Mathias
- Department of MedicineJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Patricia A. Peyser
- Department of EpidemiologyUniversity of Michigan School of Public HealthAnn ArborMIUSA
| | - Laura M. Raffield
- Department of GeneticsUniversity of North Carolina at Chapel HillChapel HillNCUSA
| | | | - Alex P. Reiner
- Division of Public HealthFred Hutchinson Cancer CenterSeattleWAUSA
| | - Jerome I. Rotter
- Department of Pediatrics, The Institute for Translational Genomics and Population SciencesThe Lundquist Institute for Biomedical Innovation at Harbor‐UCLA Medical CenterTorranceCAUSA
| | - Goo Jun
- Department of Epidemiology, Human Genetics Center, School of Public HealthThe University of Texas Health Science Center at HoustonHoustonTXUSA
| | - Paul S. de Vries
- Department of Epidemiology, Human Genetics Center, School of Public HealthThe University of Texas Health Science Center at HoustonHoustonTXUSA
| | - Themistocles L. Assimes
- Department of Medicine, Division of Cardiovascular MedicineStanford University School of MedicineStanfordCAUSA
- VA Palo Alto Healthcare SystemPalo AltoCAUSA
| |
Collapse
|
31
|
Kalwick M, Roth M. A Comprehensive Review of the Genetics of Dyslipidemias and Risk of Atherosclerotic Cardiovascular Disease. Nutrients 2025; 17:659. [PMID: 40004987 PMCID: PMC11858766 DOI: 10.3390/nu17040659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/03/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
Dyslipidemias are often diagnosed based on an individual's lipid panel that may or may not include Lp(a) or apoB. But these values alone omit key information that can underestimate risk and misdiagnose disease, which leads to imprecise medical therapies that reduce efficacy with unnecessary adverse events. For example, knowing whether an individual's dyslipidemia is monogenic can granularly inform risk and create opportunities for precision therapeutics. This review explores the canonical and non-canonical causes of dyslipidemias and how they impact atherosclerotic cardiovascular disease (ASCVD) risk. This review emphasizes the multitude of genetic causes that cause primary hypercholesterolemia, hypertriglyceridemia, and low or elevated high-density lipoprotein (HDL)-cholesterol levels. Within each of these sections, this review will explore the evidence linking these genetic conditions with ASCVD risk. Where applicable, this review will summarize approved therapies for a particular genetic condition.
Collapse
Affiliation(s)
| | - Mendel Roth
- GBinsight, GB Healthwatch, San Diego, CA 92122, USA;
| |
Collapse
|
32
|
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
|
33
|
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
|
34
|
Ruotsalainen AK, Kettunen S, Suoranta T, Kaikkonen MU, Ylä-Herttuala S, Aherrahrou R. The mechanisms of Chr.9p21.3 risk locus in coronary artery disease: where are we today? Am J Physiol Heart Circ Physiol 2025; 328:H196-H208. [PMID: 39656484 DOI: 10.1152/ajpheart.00580.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 11/22/2024] [Accepted: 11/22/2024] [Indexed: 01/15/2025]
Abstract
Despite the advancements and release of new therapeutics in the past few years, cardiovascular diseases (CVDs) have remained the number one cause of death worldwide. Genetic variation of a 9p21.3 genomic locus has been identified as the most significant and robust genetic CVD risk marker on the population level, with the strongest association with coronary artery disease (CAD) and other diseases, including diabetes and cancer. Several mechanisms of 9p21.3 in CVDs have been proposed, but their effects on CVDs have remained elusive. Moreover, most of the single nucleotide polymorphisms (SNPs) associated with CAD are located on a sequence of a long noncoding RNA (lncRNA) called ANRIL. ANRIL has several linear and circular splicing isoforms, which seem to have different effects and implications for CVDs. The mechanisms of the 9p21.3 locus and the interplay of its coding and noncoding transcripts in different diseases require further research. Circular RNAs have generally raised interest due to their beneficial features as biomarkers and therapeutic molecules. Here, we review the literature of 9p21.3 from its identification in 2007 and draw the current knowledge on its function, implications in CVDs, and therapeutic potential.
Collapse
Affiliation(s)
- Anna-Kaisa Ruotsalainen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Sanna Kettunen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Tuisku Suoranta
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Minna U Kaikkonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Seppo Ylä-Herttuala
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
- Heart Centre, Gene Therapy Unit, Kuopio University Hospital, Kuopio, Finland
| | - Rédouane Aherrahrou
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
- Institute for Cardiogenetics, Universität zu Lübeck, Lübeck, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, University Heart Centre Lübeck, Lübeck, Germany
| |
Collapse
|
35
|
Chignon A, Lettre G. Using omics data and genome editing methods to decipher GWAS loci associated with coronary artery disease. Atherosclerosis 2025; 401:118621. [PMID: 39909615 DOI: 10.1016/j.atherosclerosis.2024.118621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/18/2024] [Accepted: 10/03/2024] [Indexed: 02/07/2025]
Abstract
Coronary artery disease (CAD) is due to atherosclerosis, a pathophysiological process that involves several cell-types and results in the accumulation of lipid-rich plaque that disrupt the normal blood flow through the coronary arteries to the heart. Genome-wide association studies have identified 1000s of genetic variants robustly associated with CAD or its traditional risk factors (e.g. blood pressure, blood lipids, type 2 diabetes, smoking). However, gaining biological insights from these genetic discoveries remain challenging because of linkage disequilibrium and the difficulty to interpret the functions of non-coding regulatory elements in the human genome. In this review, we present different statistical methods (e.g. Mendelian randomization) and molecular datasets (e.g. expression or protein quantitative trait loci) that have helped connect CAD-associated variants with genes, biological pathways, and cell-types or tissues. We emphasize that these various strategies make predictions, which need to be validated in orthologous systems. We discuss specific examples where the integration of omics data with GWAS results has prioritized causal CAD variants and genes. Finally, we review how targeted and genome-wide genome editing experiments using the CRISPR/Cas9 toolbox have been used to characterize new CAD genes in human cells. Researchers now have the statistical and bioinformatic methods, the molecular datasets, and the experimental tools to dissect comprehensively the loci that contribute to CAD risk in humans.
Collapse
Affiliation(s)
- Arnaud Chignon
- Montreal Heart Institute, Montreal, Quebec, Canada; Faculté de Médecine, Université de Montréal, Montreal, Quebec, Canada
| | - Guillaume Lettre
- Montreal Heart Institute, Montreal, Quebec, Canada; Faculté de Médecine, Université de Montréal, Montreal, Quebec, Canada.
| |
Collapse
|
36
|
Alemu R, Sharew NT, Arsano YY, Ahmed M, Tekola-Ayele F, Mersha TB, Amare AT. Multi-omics approaches for understanding gene-environment interactions in noncommunicable diseases: techniques, translation, and equity issues. Hum Genomics 2025; 19:8. [PMID: 39891174 PMCID: PMC11786457 DOI: 10.1186/s40246-025-00718-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 01/16/2025] [Indexed: 02/03/2025] Open
Abstract
Non-communicable diseases (NCDs) such as cardiovascular diseases, chronic respiratory diseases, cancers, diabetes, and mental health disorders pose a significant global health challenge, accounting for the majority of fatalities and disability-adjusted life years worldwide. These diseases arise from the complex interactions between genetic, behavioral, and environmental factors, necessitating a thorough understanding of these dynamics to identify effective diagnostic strategies and interventions. Although recent advances in multi-omics technologies have greatly enhanced our ability to explore these interactions, several challenges remain. These challenges include the inherent complexity and heterogeneity of multi-omic datasets, limitations in analytical approaches, and severe underrepresentation of non-European genetic ancestries in most omics datasets, which restricts the generalizability of findings and exacerbates health disparities. This scoping review evaluates the global landscape of multi-omics data related to NCDs from 2000 to 2024, focusing on recent advancements in multi-omics data integration, translational applications, and equity considerations. We highlight the need for standardized protocols, harmonized data-sharing policies, and advanced approaches such as artificial intelligence/machine learning to integrate multi-omics data and study gene-environment interactions. We also explore challenges and opportunities in translating insights from gene-environment (GxE) research into precision medicine strategies. We underscore the potential of global multi-omics research in advancing our understanding of NCDs and enhancing patient outcomes across diverse and underserved populations, emphasizing the need for equity and fairness-centered research and strategic investments to build local capacities in underrepresented populations and regions.
Collapse
Affiliation(s)
- Robel Alemu
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Anderson School of Management, University of California Los Angeles, Los Angeles, CA, USA.
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia.
| | - Nigussie T Sharew
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia
| | - Yodit Y Arsano
- Alpert Medical School, Lifespan Health Systems, Brown University, WarrenProvidence, Rhode Island, USA
| | - Muktar Ahmed
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia
| | - Fasil Tekola-Ayele
- Epidemiology Branch, Division of Population Health Research, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Tesfaye B Mersha
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| | - Azmeraw T Amare
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia.
| |
Collapse
|
37
|
Liu B, Li Y, Han M, Yuan C, Liu B, Ren X, Liu T, Huang K, Li J, Liu F, Lu X, Tian W. Polygenic risk score, dietary inflammatory potential, and incident coronary heart disease. Eur J Prev Cardiol 2025:zwaf009. [PMID: 39881513 DOI: 10.1093/eurjpc/zwaf009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 07/18/2024] [Accepted: 01/06/2025] [Indexed: 01/31/2025]
Abstract
AIMS Dietary inflammatory potential and genetic factors are reported as being linked to coronary heart disease (CHD). We aimed to investigate their joint association with CHD incidence. METHODS AND RESULTS We included 51 889 British participants from the UK Biobank who completed the 24-h dietary assessment at baseline. We used reduced rank regression and stepwise linear regression analyses to generate an empirical dietary inflammatory pattern (EDIP) score to assess dietary inflammatory potential. A polygenic risk score (PRS) for CHD was constructed based on 1.7 million genetic variants. During a median follow-up of 11 years, 1346 incident cases of CHD were observed. High EDIP scores significantly increased the risk of CHD with the hazard ratio (HR) [95% confidence interval (CI)] of 1.26 (1.10-1.45) for high EDIP scores (T3) compared with low EDIP scores (T1). Interestingly, we observed a gradient in the risk of CHD across PRS categories, with the HRs of 1.12 (95% CI: 0.73-1.71), 1.20 (95% CI: 1.01-1.43), and 1.42 (95% CI: 1.10-1.83) in low (Q1), intermediate (Q2-4), and high (Q5) PRS categories, respectively. When the joint effect was examined, individuals with high PRS (Q5) and high EDIP scores (T3) would have the highest risk of CHD with a HR of 3.87 (95% CI: 2.74-5.46) compared with individuals with both low PRS (Q1) and low EDIP scores. CONCLUSION High dietary inflammatory potential was associated with a higher CHD risk, especially in those with high PRS, suggesting that a comprehensive assessment of inflammatory diet and genetic factors may be beneficial in the prevention of CHD.
Collapse
Affiliation(s)
- Bangquan Liu
- Department of Epidemiology, School of Public Health, Harbin Medical University, 157 Baojian Road, Harbin 150081, China
- Key Laboratory of Cardiovascular Epidemiology and Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Xicheng District, Beijing 100037, China
| | - Yun Li
- School of Public Health, North China University of Science and Technology, Tangshan 063210, China
| | - Minghui Han
- Department of Epidemiology, School of Public Health, Center for Global Health, Nanjing Medical University, Nanjing 211166, China
| | - Chenxi Yuan
- Department of Epidemiology, School of Public Health, Center for Global Health, Nanjing Medical University, Nanjing 211166, China
| | - Bisen Liu
- Key Laboratory of Cardiovascular Epidemiology and Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Xicheng District, Beijing 100037, China
| | - Xiyun Ren
- Department of Epidemiology, School of Public Health, Harbin Medical University, 157 Baojian Road, Harbin 150081, China
| | - Tianyu Liu
- Department of Epidemiology, School of Public Health, Harbin Medical University, 157 Baojian Road, Harbin 150081, China
| | - Keyong Huang
- Key Laboratory of Cardiovascular Epidemiology and Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Xicheng District, Beijing 100037, China
| | - Jianxin Li
- Key Laboratory of Cardiovascular Epidemiology and Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Xicheng District, Beijing 100037, China
| | - Fangchao Liu
- Key Laboratory of Cardiovascular Epidemiology and Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Xicheng District, Beijing 100037, China
| | - Xiangfeng Lu
- Key Laboratory of Cardiovascular Epidemiology and Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Xicheng District, Beijing 100037, China
| | - Wenjing Tian
- Department of Epidemiology, School of Public Health, Harbin Medical University, 157 Baojian Road, Harbin 150081, China
| |
Collapse
|
38
|
Clarke SL, Huang RDL, Hilliard AT, Levin MG, Sharma D, Thomson B, Lynch J, Tsao PS, Gaziano JM, Assimes TL. Genetically predicted lipoprotein(a) associates with coronary artery plaque severity independent of low-density lipoprotein cholesterol. Eur J Prev Cardiol 2025; 32:116-127. [PMID: 39158116 DOI: 10.1093/eurjpc/zwae271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 06/11/2024] [Accepted: 08/15/2024] [Indexed: 08/20/2024]
Abstract
AIMS Elevated lipoprotein(a) [Lp(a)] is a causal risk factor for atherosclerotic cardiovascular disease, but the mechanisms of risk are debated. Studies have found inconsistent associations between Lp(a) and measurements of atherosclerosis. We aimed to assess the relationship between Lp(a), low-density lipoprotein cholesterol (LDL-C), and coronary artery plaque severity. METHODS AND RESULTS The study population consisted of participants of the Million Veteran Program who have undergone an invasive angiogram. The primary exposure was genetically predicted Lp(a) estimated by a polygenic score. Genetically predicted LDL-C was also assessed for comparison. The primary outcome was coronary artery plaque severity categorized as normal, non-obstructive disease, one-vessel disease, two-vessel disease, and three-vessel or left main disease. Among 18 927 adults of genetically inferred European ancestry and 4039 adults of genetically inferred African ancestry, we observed consistent associations between genetically predicted Lp(a) and obstructive coronary plaque, with effect sizes trending upward for increasingly severe categories of disease. Associations were independent of risk factors, clinically measured LDL-C and genetically predicted LDL-C. However, we did not find strong or consistent evidence for an association between genetically predicted Lp(a) and risk for non-obstructive plaque. CONCLUSION Genetically predicted Lp(a) is positively associated with coronary plaque severity independent of LDL-C, consistent with Lp(a) promoting atherogenesis. However, the effects of Lp(a) may be greater for progression of plaque to obstructive disease than for the initial development of non-obstructive plaque. A limitation of this study is that Lp(a) was estimated using genetic markers and could not be directly assayed nor could apo(a) isoform size.
Collapse
Affiliation(s)
- Shoa L Clarke
- VA Palo Alto Healthcare System, 3801 Miranda Avenue, Palo Alto, CA, 94304, USA
- Department of Medicine, Stanford Prevention Research Center, Stanford University School of Medicine, 300 Pasteur Drive, Palo Alto, CA, 94304, USA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Rose D L Huang
- VA Palo Alto Healthcare System, 3801 Miranda Avenue, Palo Alto, CA, 94304, USA
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA, USA
| | - Austin T Hilliard
- VA Palo Alto Healthcare System, 3801 Miranda Avenue, Palo Alto, CA, 94304, USA
| | - Michael G Levin
- Corporal Michael J. Crescenz VA Medical Center, 3900 Woodland Avenue, Philadelphia, PA, 19104, USA
- Department of Medicine, Division of Cardiovascular Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Disha Sharma
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Blake Thomson
- Stanford University School of Medicine, Stanford, CA, USA
| | - Julie Lynch
- VA Informatics and Computing Infrastructure, VA Salt Lake City Health Care System, Salt Lake City, UT, USA
| | - Philip S Tsao
- VA Palo Alto Healthcare System, 3801 Miranda Avenue, Palo Alto, CA, 94304, USA
| | - J Michael Gaziano
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA, USA
| | - Themistocles L Assimes
- VA Palo Alto Healthcare System, 3801 Miranda Avenue, Palo Alto, CA, 94304, USA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
39
|
Yang C, Ji L, Han X. Low C-Reactive Protein Alleles in Hepatocyte Nuclear Factor 1A Are Associated With an Increased Risk of Cardiovascular Disease. J Clin Endocrinol Metab 2025; 110:592-600. [PMID: 39210612 DOI: 10.1210/clinem/dgae602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 07/10/2024] [Accepted: 08/29/2024] [Indexed: 09/04/2024]
Abstract
CONTEXT Rare variants in HNF1A cause both maturity onset diabetes of the young 3 (HNF1A-MODY) and reduced serum C-reactive protein (CRP) levels. Common variants of HNF1A are associated with serum CRP and type 2 diabetes mellitus (T2DM), but inconsistently with cardiovascular disease (CVD). OBJECTIVE Our study aimed to investigate the association of low CRP alleles in HNF1A with CVD and indirectly evaluate the CVD risk of HNF1A-MODY patients because of unavailability of enough cases to study their clinical outcomes. METHODS A literature search was performed using PubMed, Embase, and Cochrane Library databases from inception to December 2023. All relevant studies concerning the association of HNF1A with CRP, CVD, lipids, and T2DM were included. Odds ratios (ORs), 95% CIs, and study characteristics were extracted. RESULTS Three common coding variants of HNF1A (rs1169288, rs2464196, and rs1169289) were examined. The minor alleles of these variants correlated with low CRP levels (OR 0.89; 95% CI, 0.86-0.91; OR 0.89; 95% CI, 0.88-0.91; OR 0.89; 95% CI, 0.88-0.91, respectively). Their low CRP alleles were associated with increased risk of CVD (OR 1.03; 95% CI, 1.03-1.04), higher low-density lipoprotein cholesterol levels (OR 1.07; 95% CI, 1.04-1.10), and elevated risk of T2DM (OR 1.04; 95%, CI 1.01-1.08). CONCLUSION Our study revealed an association between low CRP alleles in HNF1A and a high CVD risk, which indicated that antidiabetic drugs with CV benefits such as glucagon-like peptide-1 receptor agonists should be recommended as a first-line choice for HNF1A-MODY.
Collapse
Affiliation(s)
- Chaochao Yang
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center, Beijing 100044, China
| | - Linong Ji
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center, Beijing 100044, China
| | - Xueyao Han
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center, Beijing 100044, China
| |
Collapse
|
40
|
Liu H, Zhang Y, Zhao Y, Li Y, Zhang X, Bao L, Yan R, Yang Y, Zhou H, Zhang J, Song S. Research Progress and Clinical Translation Potential of Coronary Atherosclerosis Diagnostic Markers from a Genomic Perspective. Genes (Basel) 2025; 16:98. [PMID: 39858645 PMCID: PMC11764800 DOI: 10.3390/genes16010098] [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: 12/05/2024] [Revised: 12/31/2024] [Accepted: 01/15/2025] [Indexed: 01/27/2025] Open
Abstract
Objective: Coronary atherosclerosis (CAD) is characterized by arterial intima lipid deposition, chronic inflammation, and fibrous tissue proliferation, leading to arterial wall thickening and lumen narrowing. As the primary cause of coronary heart disease and acute coronary syndrome, CAD significantly impacts global health. Recent genetic studies have demonstrated CAD's polygenic and multifactorial nature, providing molecular insights for early diagnosis and risk assessment. This review analyzes recent advances in CAD-related genetic markers and evaluates their diagnostic potential, focusing on their applications in diagnosis and risk stratification within precision medicine. Methods: We conducted a systematic review of CAD genomic studies from PubMed and Web of Science databases, analyzing findings from genome-wide association studies (GWASs), gene sequencing, transcriptomics, and epigenomics research. Results: GWASs and sequencing studies have identified key genetic variations associated with CAD, including JCAD/KIAA1462, GUCY1A3, PCSK9, and SORT1, which regulate inflammation, lipid metabolism, and vascular function. Transcriptomic and epigenomic analyses have revealed disease-specific gene expression patterns, DNA methylation signatures, and regulatory non-coding RNAs (miRNAs and lncRNAs), providing new approaches for early detection. Conclusions: While genetic marker research in CAD has advanced significantly, clinical implementation faces challenges including marker dynamics, a lack of standardization, and integration with conventional diagnostics. Future research should prioritize developing standardized guidelines, conducting large-scale prospective studies, and enhancing multi-omics data integration to advance genomic diagnostics in CAD, ultimately improving patient outcomes through precision medicine.
Collapse
Affiliation(s)
- Hanxiang Liu
- School of Medical Imaging, Xuzhou Medical University, No. 209 Tongshan Road, Xuzhou 221004, China
| | - Yuchen Zhang
- School of Medical Imaging, Xuzhou Medical University, No. 209 Tongshan Road, Xuzhou 221004, China
| | - Yueyan Zhao
- Medical and Information College, Xuzhou Medical University, No. 209 Tongshan Road, Xuzhou 221004, China
| | - Yuzhen Li
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Xiaofeng Zhang
- Greenwich Hospital, Yale New Haven Health, Greenwich, CT 06519, USA
| | - Lingyu Bao
- Department of Internal Medicine, Montefiore Medical Center Wakefield Campus, 600 East 233rd Street, Bronx, NY 10466, USA (H.Z.)
| | - Rongkai Yan
- Department of Radiology, Ohio State University, Columbus, OH 43210, USA
| | - Yixin Yang
- Department of Clinical Medicine, The First Clinical Medical College, Norman Bethune University of Medical Sciences, Jilin 130021, China
| | - Huixian Zhou
- Department of Internal Medicine, Montefiore Medical Center Wakefield Campus, 600 East 233rd Street, Bronx, NY 10466, USA (H.Z.)
| | - Jinming Zhang
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77030, USA
| | - Siyuan Song
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
41
|
Gunn S, Wang X, Posner DC, Cho K, Huffman JE, Gaziano M, Wilson PW, Sun YV, Peloso G, Lunetta KL. Comparison of methods for building polygenic scores for diverse populations. HGG ADVANCES 2025; 6:100355. [PMID: 39323095 PMCID: PMC11532986 DOI: 10.1016/j.xhgg.2024.100355] [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: 02/13/2024] [Revised: 09/22/2024] [Accepted: 09/22/2024] [Indexed: 09/27/2024] Open
Abstract
Polygenic scores (PGSs) are a promising tool for estimating individual-level genetic risk of disease based on the results of genome-wide association studies (GWASs). However, their promise has yet to be fully realized because most currently available PGSs were built with genetic data from predominantly European-ancestry populations, and PGS performance declines when scores are applied to target populations different from the populations from which they were derived. Thus, there is a great need to improve PGS performance in currently under-studied populations. In this work we leverage data from two large and diverse cohorts the Million Veterans Program (MVP) and All of Us (AoU), providing us the unique opportunity to compare methods for building PGSs for multi-ancestry populations across multiple traits. We build PGSs for five continuous traits and five binary traits using both multi-ancestry and single-ancestry approaches with popular Bayesian PGS methods and both MVP META GWAS results and population-specific GWAS results from the respective African, European, and Hispanic MVP populations. We evaluate these scores in three AoU populations genetically similar to the respective African, Admixed American, and European 1000 Genomes Project superpopulations. Using correlation-based tests, we make formal comparisons of the PGS performance across the multiple AoU populations. We conclude that approaches that combine GWAS data from multiple populations produce PGSs that perform better than approaches that utilize smaller single-population GWAS results matched to the target population, and specifically that multi-ancestry scores built with PRS-CSx outperform the other approaches in the three AoU populations.
Collapse
Affiliation(s)
- Sophia Gunn
- Biostatistics, Boston University School of Public Health, Boston, MA, USA; VA Boston Healthcare System, Boston, MA, USA.
| | - Xin Wang
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA; Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Daniel C Posner
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC) , Boston, MA, USA
| | - Kelly Cho
- Department of Medicine, Harvard Medical School, Boston, MA, USA; MVP Boston Coordinating Center, VA Boston Healthcare System, Boston, MA, USA; Department of Medicine, Division of Aging, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Jennifer E Huffman
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC) , Boston, MA, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA; Palo Alto Veterans Institute for Research (PAVIR), Palo Alto Health Care System, Palo Alto, CA, USA
| | - Michael Gaziano
- Department of Medicine, Harvard Medical School, Boston, MA, USA; MVP Boston Coordinating Center, VA Boston Healthcare System, Boston, MA, USA; Department of Medicine, Division of Aging, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Peter W Wilson
- VA Atlanta Healthcare System, Decatur, GA, USA; Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA; Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Yan V Sun
- VA Atlanta Healthcare System, Decatur, GA, USA; Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Gina Peloso
- Biostatistics, Boston University School of Public Health, Boston, MA, USA; VA Boston Healthcare System, Boston, MA, USA
| | - Kathryn L Lunetta
- Biostatistics, Boston University School of Public Health, Boston, MA, USA
| |
Collapse
|
42
|
Abramowitz SA, Boulier K, Keat K, Cardone KM, Shivakumar M, DePaolo J, Judy R, Bermudez F, Mimouni N, Neylan C, Kim D, Rader DJ, Ritchie MD, Voight BF, Pasaniuc B, Levin MG, Damrauer SM. Evaluating Performance and Agreement of Coronary Heart Disease Polygenic Risk Scores. JAMA 2025; 333:60-70. [PMID: 39549270 PMCID: PMC11569413 DOI: 10.1001/jama.2024.23784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 10/23/2024] [Indexed: 11/18/2024]
Abstract
Importance Polygenic risk scores (PRSs) for coronary heart disease (CHD) are a growing clinical and commercial reality. Whether existing scores provide similar individual-level assessments of disease susceptibility remains incompletely characterized. Objective To characterize the individual-level agreement of CHD PRSs that perform similarly at the population level. Design, Setting, and Participants Cross-sectional study of participants from diverse backgrounds enrolled in the All of Us Research Program (AOU), Penn Medicine BioBank (PMBB), and University of California, Los Angeles (UCLA) ATLAS Precision Health Biobank with electronic health record and genotyping data. Exposures Polygenic risk for CHD from published PRSs and new PRSs developed separately from testing samples. Main Outcomes and Measures PRSs that performed population-level prediction similarly were identified by comparing calibration and discrimination of models of prevalent CHD. Individual-level agreement was tested with intraclass correlation coefficient (ICC) and Light κ. Results A total of 48 PRSs were calculated for 171 095 AOU participants. The mean (SD) age was 56.4 (16.8) years. A total of 104 947 participants (61.3%) were female. A total of 35 590 participants (20.8%) were most genetically similar to an African reference population, 29 801 (17.4%) to an admixed American reference population, 100 493 (58.7%) to a European reference population, and the remaining to Central/South Asian, East Asian, and Middle Eastern reference populations. There were 17 589 participants (10.3%) with and 153 506 participants without (89.7%) CHD. When included in a model of prevalent CHD, 46 scores had practically equivalent Brier scores and area under the receiver operator curves (region of practical equivalence ±0.02). Twenty percent of participants had at least 1 score in both the top and bottom 5% of risk. Continuous agreement of individual predictions was poor (ICC, 0.373 [95% CI, 0.372-0.375]). Light κ, used to evaluate consistency of risk assignment, did not exceed 0.56. Analysis among 41 193 PMBB and 53 092 ATLAS participants yielded different sets of equivalent scores, which also lacked individual-level agreement. Conclusions and Relevance CHD PRSs that performed similarly at the population level demonstrated highly variable individual-level estimates of risk. Recognizing that CHD PRSs may generate incongruent individual-level risk estimates, effective clinical implementation will require refined statistical methods to quantify uncertainty and new strategies to communicate this uncertainty to patients and clinicians.
Collapse
Affiliation(s)
- Sarah A. Abramowitz
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York
| | - Kristin Boulier
- Department of Computational Medicine, University of California, Los Angeles
| | - Karl Keat
- Genomics and Computational Biology Graduate Program, University of Pennsylvania, Philadelphia
| | - Katie M. Cardone
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Manu Shivakumar
- Genomics and Computational Biology Graduate Program, University of Pennsylvania, Philadelphia
| | - John DePaolo
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Renae Judy
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Francisca Bermudez
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Nour Mimouni
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Christopher Neylan
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Dokyoon Kim
- Institute for Biomedical Informatics, University of Pennsylvania, Philadelphia
| | - Daniel J. Rader
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia
- Institute of Translational Medicine and Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Marylyn D. Ritchie
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia
- Institute for Biomedical Informatics, University of Pennsylvania, Philadelphia
| | - Benjamin F. Voight
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia
- Institute for Biomedical Informatics, University of Pennsylvania, Philadelphia
- Institute of Translational Medicine and Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Bogdan Pasaniuc
- Department of Computational Medicine, University of California, Los Angeles
| | - Michael G. Levin
- Institute of Translational Medicine and Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia
- Cardiovascular Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, Pennsylvania
- Division of Cardiovascular Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Scott M. Damrauer
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia
- Institute for Biomedical Informatics, University of Pennsylvania, Philadelphia
- Institute of Translational Medicine and Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia
- Cardiovascular Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, Pennsylvania
| |
Collapse
|
43
|
Cerezo M, Sollis E, Ji Y, Lewis E, Abid A, Bircan K, Hall P, Hayhurst J, John S, Mosaku A, Ramachandran S, Foreman A, Ibrahim A, McLaughlin J, Pendlington Z, Stefancsik R, Lambert SA, McMahon A, Morales J, Keane T, Inouye M, Parkinson H, Harris LW. The NHGRI-EBI GWAS Catalog: standards for reusability, sustainability and diversity. Nucleic Acids Res 2025; 53:D998-D1005. [PMID: 39530240 PMCID: PMC11701593 DOI: 10.1093/nar/gkae1070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 10/15/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
The NHGRI-EBI GWAS Catalog serves as a vital resource for the genetic research community, providing access to the most comprehensive database of human GWAS results. Currently, it contains close to 7 000 publications for >15 000 traits, from which more than 625 000 lead associations have been curated. Additionally, 85 000 full genome-wide summary statistics datasets-containing association data for all variants in the analysis-are available for downstream analyses such as meta-analysis, fine-mapping, Mendelian randomisation or development of polygenic risk scores. As a centralised repository for GWAS results, the GWAS Catalog sets and implements standards for data submission and harmonisation, and encourages the use of consistent descriptors for traits, samples and methodologies. We share processes and vocabulary with the PGS Catalog, improving interoperability for a growing user group. Here, we describe the latest changes in data content, improvements in our user interface, and the implementation of the GWAS-SSF standard format for summary statistics. We address the challenges of handling the rapid increase in large-scale molecular quantitative trait GWAS and the need for sensitivity in the use of population and cohort descriptors while maintaining data interoperability and reusability.
Collapse
Affiliation(s)
- Maria Cerezo
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Elliot Sollis
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Yue Ji
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Elizabeth Lewis
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Ala Abid
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Karatuğ Ozan Bircan
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Peggy Hall
- Division of Genomic Medicine, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - James Hayhurst
- Open Targets, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Sajo John
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Abayomi Mosaku
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Santhi Ramachandran
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Amy Foreman
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Arwa Ibrahim
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - James McLaughlin
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Zoë Pendlington
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Ray Stefancsik
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Samuel A Lambert
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Aoife McMahon
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Joannella Morales
- Division of Genomic Medicine, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Thomas Keane
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Michael Inouye
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, 75 Commercial Rd, Melbourne, 3004 Victoria, Australia
| | - Helen Parkinson
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Laura W Harris
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| |
Collapse
|
44
|
Biswas S, Hilser JR, Woodward NC, Wang Z, Gukasyan J, Nemet I, Schwartzman WS, Huang P, Han Y, Fouladian Z, Charugundla S, Spencer NJ, Pan C, Tang WHW, Lusis AJ, Hazen SL, Hartiala JA, Allayee H. Exploring the Role of Glycine Metabolism in Coronary Artery Disease: Insights from Human Genetics and Mouse Models. Nutrients 2025; 17:198. [PMID: 39796632 PMCID: PMC11723402 DOI: 10.3390/nu17010198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/19/2024] [Accepted: 12/31/2024] [Indexed: 01/13/2025] Open
Abstract
Background: Circulating glycine levels have been associated with reduced risk of coronary artery disease (CAD) in humans but these associations have not been observed in all studies. We evaluated whether the relationship between glycine levels and atherosclerosis was causal using genetic analyses in humans and feeding studies in mice. Methods: Serum glycine levels were evaluated for association with risk of CAD in the UK Biobank. Genetic determinants of glycine levels were identified through a genome-wide association study (GWAS) and used to evaluate the causal relationship between glycine and risk of CAD by Mendelian randomization (MR). A dietary supplementation study was carried out with atherosclerosis-prone apolipoprotein E deficient (ApoE-/-) mice to determine the effects of increased circulating glycine levels on cardiometabolic traits and aortic lesion formation. Results: Among 105,718 UK Biobank subjects, elevated serum glycine levels were associated with significantly reduced risk of prevalent CAD (Quintile 5 vs. Quintile 1 OR = 0.76, 95% CI 0.67-0.87; p < 0.0001) and incident CAD (Quintile 5 vs. Quintile 1 HR = 0.70, 95% CI 0.65-0.77; p < 0.0001) after adjustment for age, sex, ethnicity, anti-hypertensive and lipid-lowering medications, blood pressure, kidney function, and diabetes. A GWAS meta-analysis with 230,947 subjects identified 61 loci for glycine levels, of which 26 were novel. MR analyses provided modest evidence that genetically elevated glycine levels were causally associated with reduced systolic blood pressure and risk of type 2 diabetes, but did not provide significant evidence for an association with decreased risk of CAD. Glycine supplementation in mice had no effects on cardiometabolic traits or atherosclerotic lesion development. Conclusions: While expanding the genetic architecture of glycine metabolism, MR analyses and in vivo feeding studies did not provide evidence that the clinical association of this amino acid with atherosclerosis represents a causal relationship.
Collapse
Affiliation(s)
- Subarna Biswas
- Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - James R. Hilser
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Nicholas C. Woodward
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Zeneng Wang
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Microbiome and Human Health, Cleveland Clinic, Cleveland, OH 44195, USA
- Department of Cardiovascular Medicine, Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Janet Gukasyan
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Ina Nemet
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Microbiome and Human Health, Cleveland Clinic, Cleveland, OH 44195, USA
| | - William S. Schwartzman
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Pin Huang
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Yi Han
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Zachary Fouladian
- Department of Medicine, David Geffen School of Medicine of UCLA, Los Angeles, CA 90095, USA
| | - Sarada Charugundla
- Department of Medicine, David Geffen School of Medicine of UCLA, Los Angeles, CA 90095, USA
| | - Neal J. Spencer
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Calvin Pan
- Department of Human Genetics, David Geffen School of Medicine of UCLA, Los Angeles, CA 90095, USA
| | - W. H. Wilson Tang
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Microbiome and Human Health, Cleveland Clinic, Cleveland, OH 44195, USA
- Department of Cardiovascular Medicine, Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Aldons J. Lusis
- Department of Medicine, David Geffen School of Medicine of UCLA, Los Angeles, CA 90095, USA
- Department of Human Genetics, David Geffen School of Medicine of UCLA, Los Angeles, CA 90095, USA
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine of UCLA, Los Angeles, CA 90095, USA
| | - Stanley L. Hazen
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Microbiome and Human Health, Cleveland Clinic, Cleveland, OH 44195, USA
- Department of Cardiovascular Medicine, Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Jaana A. Hartiala
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Hooman Allayee
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
45
|
Wayne N, Singamneni VS, Venkatesh R, Cherlin T, Verma SS, Guerraty MA. Genetic Insights Into Coronary Microvascular Disease. Microcirculation 2025; 32:e12896. [PMID: 39755372 DOI: 10.1111/micc.12896] [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: 09/18/2024] [Revised: 10/29/2024] [Accepted: 11/26/2024] [Indexed: 01/06/2025]
Abstract
Coronary microvascular disease (CMVD) affects the coronary pre-arterioles, arterioles, and capillaries and can lead to blood supply-demand mismatch and cardiac ischemia. CMVD can present clinically as ischemia or myocardial infarction with no obstructive coronary arteries (INOCA or MINOCA, respectively). Currently, therapeutic options for CMVD are limited, and there are no targeted therapies. Genetic studies have emerged as an important tool to gain rapid insights into the molecular mechanisms of human diseases. For example, coronary artery disease (CAD) genome-wide association studies (GWAS) have enrolled hundreds of thousands of patients and have identified > 320 loci, elucidating CAD pathogenic pathways and helping to identify therapeutic targets. Here, we review the current landscape of genetic studies of CMVD, consisting mostly of genotype-first approaches. We then present the hypothesis that CAD GWAS have enrolled heterogenous populations and may be better characterized as ischemic heart disease (IHD) GWAS. We discuss how several of the genetic loci currently associated with CAD may be involved in the pathogenesis of CMVD. Genetic studies could help accelerate progress in understanding CMVD pathophysiology and identifying putative therapeutic targets. Larger phenotype-first genomic studies into CMVD with adequate sex and ancestry representation are needed. Given the extensive CAD genetic and functional validation data, future research should leverage these loci as springboards for CMVD genomic research.
Collapse
Affiliation(s)
- Nicole Wayne
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Venkata S Singamneni
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Rasika Venkatesh
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Tess Cherlin
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Shefali S Verma
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Marie A Guerraty
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
46
|
Ding K, Qin X, Wang H, Wang K, Kang X, Yu Y, Liu Y, Gong H, Wu T, Chen D, Hu Y, Wang T, Wu Y. Identification of shared genetic etiology of cardiovascular and cerebrovascular diseases through common cardiometabolic risk factors. Commun Biol 2024; 7:1703. [PMID: 39730871 DOI: 10.1038/s42003-024-07417-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 12/18/2024] [Indexed: 12/29/2024] Open
Abstract
Cardiovascular diseases (CVDs) and cerebrovascular diseases (CeVDs) are closely related vascular diseases, sharing common cardiometabolic risk factors (RFs). Although pleiotropic genetic variants of these two diseases have been reported, their underlying pathological mechanisms are still unclear. Leveraging GWAS summary data and using genetic correlation, pleiotropic variants identification, and colocalization analyses, we identified 11 colocalized loci for CVDs-CeVDs-BP (blood pressure), CVDs-CeVDs-LIP (lipid traits), and CVDs-CeVDs-cIMT (carotid intima-media thickness) triplets. No shared causal loci were found for CVDs-CeVDs-T2D (type 2 diabetes) or CVDs-CeVDs-BMI (body mass index) triplets. The 11 loci were mapped to 12 genes, namely CASZ1, CDKN1A, TWIST1, CDKN2B, ABO, SWAP70, SH2B3, LRCH1, FES, GOSR2, RPRML, and LDLR, where both GOSR2 and RPRML were mapped to one locus. They were enriched in pathways related to cellular response to external stimulus and regulation of the phosphate metabolic process and were highly expressed in endothelial cells, epithelial cells, and smooth muscle cells. Multi-omics analysis revealed methylation of two genes (CASZ1 and LRCH1) may play a causal role in the genetic pleiotropy. Notably, these pleiotropic loci are highly enriched in the targets of antihypertensive drugs, which further emphasizes the role of the blood pressure regulation pathway in the shared etiology of CVDs and CeVDs.
Collapse
Affiliation(s)
- Kexin Ding
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University; Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Xueying Qin
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University; Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China.
| | - Huairong Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University; Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Kun Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University; Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Xiaoying Kang
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Neurology, Brigham and Women's Hospital & Harvard Medical School, Boston, MA, USA
| | - Yao Yu
- Department of Neurology, Peking University People's Hospital, Beijing, China
| | - Yang Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University; Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Haiying Gong
- Fangshan District Center for Disease Control and Prevention, Beijing, China
| | - Tao Wu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University; Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Dafang Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University; Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Yonghua Hu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University; Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Tao Wang
- Department of Epidemiology and Population Health, Yeshiva University Albert Einstein College of Medicine, Bronx, NY, USA
| | - Yiqun Wu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University; Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China.
| |
Collapse
|
47
|
Noto D, Gagliardo CM, Spina R, Giammanco A, Ciaccio M, Cefalù AB, Averna M. Six genetic variants are associated with cardiovascular disease independently from canonical risk factors: a new method to refine GWAS results based on the UKBiobank phenotype database. Mol Genet Genomics 2024; 300:4. [PMID: 39704901 DOI: 10.1007/s00438-024-02202-w] [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: 04/05/2024] [Accepted: 10/28/2024] [Indexed: 12/21/2024]
Abstract
This paper describes a novel methodology based on GWAS filtering, aimed to find novel phenotypes associated to genetic loci independently of canonical risk factors using the large database of UK Biobank. Genome wide association studies (GWAS) is an untargeted methodology able to identify novel gene variants associated with diseases. Novel gene-phenotype associations might be discovered by this method. UKBiobank was interrogated by an automated routine to search associations between hundreds of phenotypes and single nucleotide polymorphisms (SNPs) resulting from GWAS, using Cardiovascular Disease as investigated trait. Six gene variants associated with CVD, independently of canonical risk factors, were identified using a variants database of more than 400k genotyped subjects (rs9349379 PHACTR1;intragenic_variant, rs74617384 LPA; intron_variant, rs4977574 CDKN2B-AS1;intron_variant, rs11191846 STN1;intron_variant, rs3184504, SH2B3;missense_variant, rs2929155 ADAMTS7;synonymous_variant). Novel clinical and biochemical phenotypes have been associated to the variants. The phenotypical characterization of the loci helped to propose mechanistic links that could explain their connection to CVD.
Collapse
Affiliation(s)
- Davide Noto
- Department of Health Promotion, Maternal and Child Care, Internal Medicine and Medical Specialities "G. D'Alessandro" (PROMISE), University of Palermo, Via del Vespro 129, Palermo, 90127, Italy.
| | - Carola Maria Gagliardo
- Department of Health Promotion, Maternal and Child Care, Internal Medicine and Medical Specialities "G. D'Alessandro" (PROMISE), University of Palermo, Via del Vespro 129, Palermo, 90127, Italy
| | - Rossella Spina
- Department of Health Promotion, Maternal and Child Care, Internal Medicine and Medical Specialities "G. D'Alessandro" (PROMISE), University of Palermo, Via del Vespro 129, Palermo, 90127, Italy
| | - Antonina Giammanco
- Department of Health Promotion, Maternal and Child Care, Internal Medicine and Medical Specialities "G. D'Alessandro" (PROMISE), University of Palermo, Via del Vespro 129, Palermo, 90127, Italy
| | - Marcello Ciaccio
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), Section of Clinical Biochemistry, Clinical Molecular Medicine and Laboratory Medicine, University of Palermo, Palermo, Italy
| | - Angelo B Cefalù
- Department of Health Promotion, Maternal and Child Care, Internal Medicine and Medical Specialities "G. D'Alessandro" (PROMISE), University of Palermo, Via del Vespro 129, Palermo, 90127, Italy
| | - Maurizio Averna
- Department of Health Promotion, Maternal and Child Care, Internal Medicine and Medical Specialities "G. D'Alessandro" (PROMISE), University of Palermo, Via del Vespro 129, Palermo, 90127, Italy
- Institute of Biophysics (IBF), National Research Council (CNR), Palermo, Italy
| |
Collapse
|
48
|
Li F, Zhang Y, Wang Y, Cai X, Fan X. Cytokine Gene Variants as Predisposing Factors for the Development and Progression of Coronary Artery Disease: A Systematic Review. Biomolecules 2024; 14:1631. [PMID: 39766338 PMCID: PMC11726869 DOI: 10.3390/biom14121631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 12/12/2024] [Accepted: 12/16/2024] [Indexed: 01/04/2025] Open
Abstract
Coronary artery disease (CAD) is the most prevalent form of cardiovascular disease. A growing body of research shows that interleukins (ILs), such as IL-8, IL-18 and IL-16, elicit pro-inflammatory responses and may play critical roles in the pathologic process of CAD. Single nucleotide polymorphisms (SNPs), capable of generating functional modifications in IL genes, appear to be associated with CAD risk. This study aims to evaluate the associations of ten previously identified SNPs of the three cytokines with susceptibility to or protection of CAD. A systematic review and meta-analysis were conducted using Pubmed, EMBASE, WOS, CENTRAL, CNKI, CBM, Weipu, WANFANG Data and Google Scholar databases for relevant literature published up to September 2024. Odds ratios (ORs) with 95% confidence intervals (CIs) were calculated for the four genetic models of the investigated SNPs in overall and subgroups analyses. Thirty-eight articles from 16 countries involving 14574 cases and 13001 controls were included. The present meta-analysis revealed no significant association between CAD and IL-8-rs2227306 or five IL-16 SNPs (rs8034928, rs3848180, rs1131445, rs4778889 and rs11556218). However, IL-8-rs4073 was significantly associated with an increased risk of CAD across all genetic models. In contrast, three IL-18 (rs187238, rs1946518 and rs1946519) variants containing minor alleles were associated with decreased risks of CAD under all models. Subgroups analyses by ethnicity indicated that IL-8-rs4073 conferred a significantly higher risk of CAD among Asians, including East, South and West Asians (allelic OR = 1.46, homozygous OR = 1.96, heterozygous OR = 1.47, dominant OR = 1.65), while it showed an inversely significant association with CAD risk in Caucasians (homozygous OR = 0.82, dominant OR = 0.85). Additionally, IL-18-rs187238 and IL-18-rs1946518 were significantly associated with reduced CAD risks in East Asians (for rs187238: allelic OR = 0.72, homozygous OR = 0.33, heterozygous OR = 0.73, dominant OR = 0.71; for rs1946518: allelic OR = 0.62, homozygous OR = 0.38, heterozygous OR = 0.49, dominant OR = 0.45). IL-18-rs187238 also demonstrated protective effects in Middle Eastern populations (allelic OR = 0.76, homozygous OR = 0.63, heterozygous OR = 0.72, dominant OR = 0.71). No significant associations were observed in South Asians or Caucasians for these IL-18 SNPs. Consistent with the overall analysis results, subgroups analyses further highlighted a significant association between IL-8-rs4073 and increased risk of acute coronary syndrome (heterozygous OR = 0.72). IL-18-rs187238 was significantly associated with decreased risks of myocardial infarction (MI) (allelic OR = 0.81, homozygous OR = 0.55, dominant OR = 0.80) and multiple vessel stenosis (allelic OR = 0.54, heterozygous OR = 0.45, dominant OR = 0.45). Similarly, IL-18-rs1946518 was significantly associated with reduced MI risk (allelic OR = 0.75, heterozygous OR = 0.68). These findings support the role of cytokine gene IL-8 and IL-18 variants as predisposing factors for the development and progression of CAD.
Collapse
Affiliation(s)
- Fang Li
- Correspondence: (F.L.); (X.F.); Tel.: +86-731-88872780 (F.L. & X.F.)
| | | | | | | | - Xiongwei Fan
- The Laboratory of Heart Development Research, College of Life Sciences, Hunan Normal University, Changsha 410081, China; (Y.Z.); (Y.W.); (X.C.)
| |
Collapse
|
49
|
Jain V, Dabbs‐Brown A, Liu C, Hui Q, Mehta A, Wilson PW, Quyyumi AA, Sun YV. Genome-Wide European Ancestry Study Identifies Coronary Artery Disease-Associated Loci Through Gene-Sex Hormone Interaction. J Am Heart Assoc 2024; 13:e034132. [PMID: 39673284 PMCID: PMC11935546 DOI: 10.1161/jaha.123.034132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 09/20/2024] [Indexed: 12/16/2024]
Abstract
BACKGROUND Although sex differences in coronary artery disease (CAD) risk have been observed, little is known about the role of sex hormones in CAD genetics. Accounting for sex hormone levels may help identify CAD-risk loci and extend our knowledge of its genetic architecture. METHODS AND RESULTS A total of 365 662 individuals of European ancestry enrolled in the UK Biobank were considered. Genetic interaction of total testosterone, bioavailable testosterone, and SHBG (sex hormone-binding globulin) were evaluated. Gene-environment interactions in millions of samples software was used to conduct sex-stratified genome-wide interaction analysis with prevalent CAD as the outcome. Participant age at enrollment and principal components 1 to 10 were adjusted as covariates. We identified 45 loci in men and 8 loci in women that reached genome-wide significance (P < 5 × 10-8) for CAD. Ten of the loci identified (5 loci in both men and women) were through joint effects and would not have been picked up using a traditional genome-wide association study. Two of the joint effect loci in women were independently identified with significant single nucleotide polymorphism-total testosterone interactions. CONCLUSIONS This genome-wide gene-sex hormone interaction study identified genomic-risk loci that may contribute to the differential CAD risk between men and women, which otherwise would not have been discovered in a traditional genome-wide association study solely including marginal genetic effects.
Collapse
Affiliation(s)
- Vardhmaan Jain
- Division of CardiologyEmory University School of MedicineAtlantaGAUSA
| | - Amonae Dabbs‐Brown
- Department of EpidemiologyEmory University Rollins School of Public HealthAtlantaGAUSA
| | - Chang Liu
- Division of CardiologyEmory University School of MedicineAtlantaGAUSA
- Department of EpidemiologyEmory University Rollins School of Public HealthAtlantaGAUSA
| | - Qin Hui
- Department of EpidemiologyEmory University Rollins School of Public HealthAtlantaGAUSA
| | - Anurag Mehta
- Virginia Commonwealth University Health, Pauley Heart CenterRichmondVAUSA
| | - Peter W.F. Wilson
- Division of CardiologyEmory University School of MedicineAtlantaGAUSA
- Atlanta VA Healthcare SystemDecaturGAUSA
| | - Arshed A. Quyyumi
- Division of CardiologyEmory University School of MedicineAtlantaGAUSA
| | - Yan V. Sun
- Department of EpidemiologyEmory University Rollins School of Public HealthAtlantaGAUSA
- Atlanta VA Healthcare SystemDecaturGAUSA
| |
Collapse
|
50
|
Kim Y, Landstrom AP, Shah SH, Wu JC, Seidman CE. Gene Therapy in Cardiovascular Disease: Recent Advances and Future Directions in Science: A Science Advisory From the American Heart Association. Circulation 2024; 150:e471-e480. [PMID: 39523949 DOI: 10.1161/cir.0000000000001296] [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: 11/16/2024]
Abstract
Cardiovascular disease remains the foremost cause of morbidity and mortality globally, affecting millions of individuals. Recent discoveries illuminate the substantial role of genetics in cardiovascular disease pathogenesis, encompassing both monogenic and polygenic mechanisms and identifying tangible targets for gene therapies. Innovative strategies have emerged to rectify pathogenic variants that cause monogenic disorders such as hypertrophic, dilated, and arrhythmogenic cardiomyopathies and hypercholesterolemia. These include delivery of exogenous genes to supplement insufficient protein levels caused by pathogenic variants or genome editing to correct, delete, or modify mutant sequences to restore protein function. However, effective delivery of gene therapy to specified cells presents formidable challenges. Viral vectors, notably adeno-associated viruses and nonviral vectors such as lipid and engineered nanoparticles, offer distinct advantages and limitations. Additional risks and obstacles remain, including treatment durability, tissue-specific targeting, vector-associated adverse events, and off-target effects. Addressing these challenges is an ongoing imperative; several clinical gene therapy trials are underway, and many more first-in-human studies are anticipated. This science advisory reviews core concepts of gene therapy, key obstacles, patient risks, and ongoing research endeavors to enable clinicians to understand the complex landscape of this emerging therapy and its remarkable therapeutic potential to benefit cardiovascular disease.
Collapse
|