1
|
Liu S, Wang X, Hu J, Zhao C, Qin X. Comprehensive evaluation of siRNA therapeutics on Lp(a): A network meta-analysis. Diabetes Obes Metab 2025; 27:3367-3378. [PMID: 40116213 DOI: 10.1111/dom.16355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 03/02/2025] [Accepted: 03/09/2025] [Indexed: 03/23/2025]
Abstract
AIMS To evaluate the efficacy and safety of siRNA drugs that lower Lp(a) in patients with dyslipidaemia. MATERIALS AND METHODS A network meta-analysis and systematic review were conducted to compare siRNA drugs targeting Lp(a), based on relevant randomized controlled trials (RCTs). A comprehensive search was performed in PubMed, Embase, Web of Science and the Cochrane Library (up to October 24, 2024). RCTs with an intervention duration of at least 12 weeks were included. Eligible studies compared siRNA drugs that reduce Lp(a), including both Lp(a)-targeted and non-targeted agents, with placebo or other siRNA drugs that reduce Lp(a). The primary outcomes were the percentage reduction and absolute reduction in Lp(a), percentage reduction in low-density lipoprotein cholesterol (LDL-C), percentage reduction in apolipoprotein B (apo(B)), adverse events and serious adverse events, including injection-site reactions. The risk of bias was assessed using the Cochrane Risk of Bias Tool (ROB2), and a random-effects network meta-analysis was performed using the frequentist approach. Confidence in effect estimates was evaluated using the Confidence In Network Meta-Analysis (CINeMA) framework. RESULTS A total of 14 trials involving 5646 participants were included. Lp(a)-targeted siRNA agents, particularly Olpasiran, demonstrated strong efficacy in significantly reducing Lp(a) levels, with the greatest percentage reduction in Lp(a) (mean difference [MD]: -92.06%; 95% CI: -102.43% to -81.69%; P-score: 0.98). Olpasiran also showed the greatest absolute reduction in Lp(a) (MD: -250.70 nmol/L; 95% confidence interval [CI]: -279.89 to -221.50; P-score: 0.99). Certain non-Lp(a)-targeted siRNA agents, such as inclisiran and zodasiran, also showed modest reductions in Lp(a) levels, reducing Lp(a) by approximately 15%. Lp(a)-targeted siRNA agents reduced LDL-C by more than 20% and decreased apo(B) by approximately 15%. In terms of safety, most drugs exhibited favourable safety profiles with no significant differences compared to placebo. However, zerlasiran raised concerns regarding injection-site reactions and other adverse events when compared to placebo. CONCLUSIONS Lp(a)-targeted siRNA agents have shown robust effectiveness in substantially reducing Lp(a) levels, including both percentage and absolute reductions, with moderate improvements in LDL-C and apo(B) concentrations. Non-Lp(a)-targeted siRNA agents also demonstrate modest reductions in Lp(a) levels. The safety profile is generally favourable, but zerlasiran and inclisiran may increase the incidence of injection-site reactions.
Collapse
Affiliation(s)
- Song Liu
- Department of Pharmacy, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Xingjin Wang
- Medical Faculty, Heidelberg University, Heidelberg, Germany
| | - Jiaqiang Hu
- Department of Pharmacy, Guang'an People's Hospital, Guang'an, Sichuan, China
| | - Chen Zhao
- Department of Pharmacy, Beijing Anzhen Nanchong Hospital, Capital Medical University & Nanchong Central Hospital, Nanchong, Sichuan, China
- Nanchong Key Laboratory of Individualized Drug Therapy, Nanchong, China
| | - Xiaoli Qin
- Department of Pharmacy, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, Sichuan, China
| |
Collapse
|
2
|
Abomoawad A, Sedhom R, Golwala H, Abdelazeem M, Mamas M, Jneid H, Bavry AA, Kumbhani DJ, Kapadia S, Elbadawi A. Transcatheter Versus Surgical Aortic Valve Replacement in Patients with Polyvascular Disease. Cardiol Ther 2025:10.1007/s40119-025-00415-7. [PMID: 40382742 DOI: 10.1007/s40119-025-00415-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 04/17/2025] [Indexed: 05/20/2025] Open
Abstract
INTRODUCTION There is a paucity of data regarding the trends and comparative outcomes of transcatheter aortic valve replacement (TAVR) versus surgical aortic valve replacement (SAVR) among patients with polyvascular disease (PVD). METHODS The Nationwide Readmissions Database (2016-2020) was queried for patients undergoing AVR. Propensity score matching was used to compare the outcomes of TAVR versus SAVR among patients with PVD, and for comparing TAVR among those with versus without PVD. The primary outcome was in-hospital mortality. RESULTS The final cohort included 545,409 hospitalizations for AVR. During the study years, there was an increase in the utilization of TAVR versus SAVR among patients with PVD. Patients with PVD undergoing TAVR were older and more likely to be women compared with patients with PVD undergoing SAVR. Compared with SAVR, patients with PVD undergoing TAVR had lower odds of in-hospital mortality (adjusted odds ratio (aOR) 0.26; 95% confidence interval (CI) 0.19-0.35), acute myocardial infarction (AMI), ischemic stroke, hemorrhagic stroke, and major bleeding, but higher odds of pacemaker and non-elective 90-day readmissions (aOR 1.13; 95% CI 1.01-1.26). TAVR among patients with versus without PVD showed similar in-hospital mortality (aOR 1.10; 95% CI 0.94-1.20), while there were higher odds of AMI, ischemic stroke, and vascular complications after TAVR in patients with PVD. A higher burden of atherosclerotic vascular beds conferred higher mortality with SAVR more than with TAVR, while a higher burden of atherosclerotic vascular beds conferred a higher risk of ischemic stroke and readmissions after both TAVR and SAVR. CONCLUSIONS Nationwide data demonstrated that patients with PVD who undergo TAVR were associated with lower in-hospital mortality and major cardiovascular complications compared with those who undergo SAVR. Patients with PVD have similar mortality to those with no PVD undergoing TAVR, but were associated with a higher risk for complications and readmission.
Collapse
Affiliation(s)
- Abdelrhman Abomoawad
- Division of Cardiovascular Medicine, Boston University School of Medicine, Boston Medical Center, Boston, MA, USA
| | - Ramy Sedhom
- Department of Cardiology, Loma Linda University, Loma Linda, CA, USA
| | - Harsh Golwala
- Division of Cardiology, Oregon Health and Science University, Portland, OR, USA
| | - Mohamed Abdelazeem
- Division of Cardiology, Christus Good Shepherd Medical Center, 707 East Marshall Avenue, Longview, TX, 75604, USA
- Texas A&M School of Medicine, Bryan, TX, USA
| | - Mamas Mamas
- Keele Cardiovascular Research Group, Centre for Prognosis Research, Keele University, Keele, UK
| | - Hani Jneid
- Division of Cardiovascular Medicine, University of Texas Medical Branch, Galveston, TX, USA
| | - Anthony A Bavry
- Division of Cardiovascular Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Dharam J Kumbhani
- Division of Cardiovascular Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Samir Kapadia
- Division of Cardiovascular Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Ayman Elbadawi
- Division of Cardiology, Christus Good Shepherd Medical Center, 707 East Marshall Avenue, Longview, TX, 75604, USA.
- Texas A&M School of Medicine, Bryan, TX, USA.
| |
Collapse
|
3
|
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
|
4
|
Ong JYS, Tan SML, Koh AS, Kong W, Sia CH, Yeo TC, Quek SC, Poh KK. Novel Circulating Biomarkers in Aortic Valve Stenosis. Int J Mol Sci 2025; 26:1902. [PMID: 40076529 PMCID: PMC11899762 DOI: 10.3390/ijms26051902] [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: 12/14/2024] [Revised: 02/14/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025] Open
Abstract
The underlying pathophysiology of aortic stenosis and factors affecting its clinical progression remain poorly understood. Apart from B-type natriuretic peptide (BNP), novel and emerging biomarkers have been described in association with aortic stenosis, emphasising the potential for these biomarkers to illuminate on yet unknown mechanisms of its pathogenesis. In this review, we aimed to summarise what is known about aortic stenosis biomarkers, highlight the emerging ones, and provide a roadmap for translating these insights into clinical applications. Among the biomarkers studied, lipoprotein(a) [Lp(a)] has emerged as the most promising for risk stratification. Elevated Lp(a) levels are often associated with more rapid aortic stenosis progression. This detrimental effect is attributed to its role in promoting valve calcification. While other emerging biomarkers such as matrix metalloproteinases, monocytes, and metabolites show promises, their specific roles in aortic stenosis pathophysiology remain less clear. This may be due to their relatively recent discovery. Ongoing research aims to elucidate their mechanisms of action.
Collapse
Affiliation(s)
- Joy Yi-Shan Ong
- Department of Cardiology, National University Heart Centre Singapore, Singapore 119074, Singapore; (J.Y.-S.O.); (S.M.L.T.)
| | - Sarah Ming Li Tan
- Department of Cardiology, National University Heart Centre Singapore, Singapore 119074, Singapore; (J.Y.-S.O.); (S.M.L.T.)
| | - Angela S. Koh
- National Heart Centre Singapore, Singapore 169609, Singapore
- DUKE-NUS Medical School, Singapore 169857, Singapore
| | - William Kong
- Department of Cardiology, National University Heart Centre Singapore, Singapore 119074, Singapore; (J.Y.-S.O.); (S.M.L.T.)
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Ching Hui Sia
- Department of Cardiology, National University Heart Centre Singapore, Singapore 119074, Singapore; (J.Y.-S.O.); (S.M.L.T.)
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Tiong Cheng Yeo
- Department of Cardiology, National University Heart Centre Singapore, Singapore 119074, Singapore; (J.Y.-S.O.); (S.M.L.T.)
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Swee Chye Quek
- Department of Cardiology, National University Heart Centre Singapore, Singapore 119074, Singapore; (J.Y.-S.O.); (S.M.L.T.)
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Kian Keong Poh
- Department of Cardiology, National University Heart Centre Singapore, Singapore 119074, Singapore; (J.Y.-S.O.); (S.M.L.T.)
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| |
Collapse
|
5
|
Blaser MC, Bäck M, Lüscher TF, Aikawa E. Calcific aortic stenosis: omics-based target discovery and therapy development. Eur Heart J 2025; 46:620-634. [PMID: 39656785 PMCID: PMC11825147 DOI: 10.1093/eurheartj/ehae829] [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: 07/19/2024] [Revised: 10/01/2024] [Accepted: 11/14/2024] [Indexed: 12/17/2024] Open
Abstract
Calcific aortic valve disease (CAVD) resulting in aortic stenosis (AS) is the most common form of valvular heart disease, affecting 2% of those over age 65. Those who develop symptomatic severe AS have an average further lifespan of <2 years without valve replacement, and three-quarters of these patients will develop heart failure, undergo valve replacement, or die within 5 years. There are no approved pharmaceutical therapies for AS, due primarily to a limited understanding of the molecular mechanisms that direct CAVD progression in the complex haemodynamic environment. Here, advances in efforts to understand the pathogenesis of CAVD and to identify putative drug targets derived from recent multi-omics studies [including (epi)genomics, transcriptomics, proteomics, and metabolomics] of blood and valvular tissues are reviewed. The recent explosion of single-cell omics-based studies in CAVD and the pathobiological and potential drug discovery insights gained from the application of omics to this disease area are a primary focus. Lastly, the translation of knowledge gained in valvular pathobiology into clinical therapies is addressed, with a particular emphasis on treatment regimens that consider sex-specific, renal, and lipid-mediated contributors to CAVD, and ongoing Phase I/II/III trials aimed at the prevention/treatment of AS are described.
Collapse
Affiliation(s)
- Mark C Blaser
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, 3 Blackfan Street, 17th Floor, Boston, MA 02115, USA
| | - Magnus Bäck
- Department of Medicine, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Division of Valvular and Coronary Disease, Heart and Vascular Theme, Karolinska University Hospital, Stockholm, Sweden
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
- Heart Division, Royal Brompton and Harefield Hospitals, London, UK
- National Heart and Lung Institute, Imperial College, London, UK
| | - Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, 3 Blackfan Street, 17th Floor, Boston, MA 02115, USA
- Center for Excellence in Vascular Biology, Brigham and Women's Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, NRB 741, Boston, MA 02115, USA
| |
Collapse
|
6
|
Blaser MC, Aikawa E. Disease Drivers in Aortic Stenosis vs Atherosclerosis. JAMA Cardiol 2025; 10:109-111. [PMID: 39504008 DOI: 10.1001/jamacardio.2024.3749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/13/2025]
Affiliation(s)
- Mark C Blaser
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
- Center for Excellence in Vascular Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
7
|
Trenkwalder T, Maj C, Al-Kassou B, Debiec R, Doppler SA, Musameh MD, Nelson CP, Dasmeh P, Grover S, Knoll K, Naamanka J, Mordi IR, Braund PS, Dreßen M, Lahm H, Wirth F, Baldus S, Kelm M, von Scheidt M, Krefting J, Ellinghaus D, Small AM, Peloso GM, Natarajan P, Thanassoulis G, Engert JC, Dufresne L, Franke A, Görg S, Laudes M, Nowak-Göttl U, Vaht M, Metspalu A, Stoll M, Berger K, Pellegrini C, Kastrati A, Hengstenberg C, Lang CC, Kessler T, Hovatta I, Nickenig G, Nöthen MM, Krane M, Schunkert H, Samani NJ, Schumacher J. Distinct Genetic Risk Profile in Aortic Stenosis Compared With Coronary Artery Disease. JAMA Cardiol 2025; 10:145-154. [PMID: 39504041 PMCID: PMC11541746 DOI: 10.1001/jamacardio.2024.3738] [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: 10/24/2023] [Accepted: 08/11/2024] [Indexed: 11/09/2024]
Abstract
Importance Aortic stenosis (AS) and coronary artery disease (CAD) frequently coexist. However, it is unknown which genetic and cardiovascular risk factors might be AS-specific and which could be shared between AS and CAD. Objective To identify genetic risk loci and cardiovascular risk factors with AS-specific associations. Design, Setting, and Participants This was a genomewide association study (GWAS) of AS adjusted for CAD with participants from the European Consortium for the Genetics of Aortic Stenosis (EGAS) (recruited 2000-2020), UK Biobank (recruited 2006-2010), Estonian Biobank (recruited 1997-2019), and FinnGen (recruited 1964-2019). EGAS participants were collected from 7 sites across Europe. All participants were of European ancestry, and information on comorbid CAD was available for all participants. Follow-up analyses with GWAS data on cardiovascular traits and tissue transcriptome data were also performed. Data were analyzed from October 2022 to July 2023. Exposures Genetic variants. Main Outcomes and Measures Cardiovascular traits associated with AS adjusted for CAD. Replication was performed in 2 independent AS GWAS cohorts. Results A total of 18 792 participants with AS and 434 249 control participants were included in this GWAS adjusted for CAD. The analysis found 17 AS risk loci, including 5 loci with novel and independently replicated associations (RNF114A, AFAP1, PDGFRA, ADAMTS7, HAO1). Of all 17 associated loci, 11 were associated with risk specifically for AS and were not associated with CAD (ALPL, PALMD, PRRX1, RNF144A, MECOM, AFAP1, PDGFRA, IL6, TPCN2, NLRP6, HAO1). Concordantly, this study revealed only a moderate genetic correlation of 0.15 (SE, 0.05) between AS and CAD (P = 1.60 × 10-3). Mendelian randomization revealed that serum phosphate was an AS-specific risk factor that was absent in CAD (AS: odds ratio [OR], 1.20; 95% CI, 1.11-1.31; P = 1.27 × 10-5; CAD: OR, 0.97; 95% CI 0.94-1.00; P = .04). Mendelian randomization also found that blood pressure, body mass index, and cholesterol metabolism had substantially lesser associations with AS compared with CAD. Pathway and transcriptome enrichment analyses revealed biological processes and tissues relevant for AS development. Conclusions and Relevance This GWAS adjusted for CAD found a distinct genetic risk profile for AS at the single-marker and polygenic level. These findings provide new targets for future AS research.
Collapse
Affiliation(s)
- Teresa Trenkwalder
- Technical University of Munich, School of Medicine and Health, Department of Cardiovascular Diseases, German Heart Centre Munich, TUM University Hospital, Munich, Germany
- German Center for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - Carlo Maj
- Institute of Human Genetics, Philipps University of Marburg, Marburg, Germany
| | - Baravan Al-Kassou
- Department of Medicine II, Heart Center Bonn, University of Bonn and University Hospital Bonn, Bonn, Germany
| | - Radoslaw Debiec
- Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom
- National Institute for Health and Care Research Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, United Kingdom
| | - Stefanie A. Doppler
- Department of Cardiovascular Surgery, German Heart Center Munich, School of Medicine and Health, TUM University Hospital, Technical University of Munich, Munich, Germany
- Institute Insure, German Heart Center Munich, School of Medicine and Health, TUM University Hospital, Technical University of Munich, Munich, Germany
| | - Muntaser D. Musameh
- Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom
- National Institute for Health and Care Research Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, United Kingdom
| | - Christopher P. Nelson
- Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom
- National Institute for Health and Care Research Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, United Kingdom
| | - Pouria Dasmeh
- Institute of Human Genetics, Philipps University of Marburg, Marburg, Germany
| | - Sandeep Grover
- Institute of Human Genetics, Philipps University of Marburg, Marburg, Germany
| | - Katharina Knoll
- Technical University of Munich, School of Medicine and Health, Department of Cardiovascular Diseases, German Heart Centre Munich, TUM University Hospital, Munich, Germany
- German Center for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - Joonas Naamanka
- SleepWell Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Ify R. Mordi
- Division of Molecular and Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, United Kingdom
| | - Peter S. Braund
- Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom
- National Institute for Health and Care Research Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, United Kingdom
| | - Martina Dreßen
- Department of Cardiovascular Surgery, German Heart Center Munich, School of Medicine and Health, TUM University Hospital, Technical University of Munich, Munich, Germany
- Institute Insure, German Heart Center Munich, School of Medicine and Health, TUM University Hospital, Technical University of Munich, Munich, Germany
| | - Harald Lahm
- Department of Cardiovascular Surgery, German Heart Center Munich, School of Medicine and Health, TUM University Hospital, Technical University of Munich, Munich, Germany
- Institute Insure, German Heart Center Munich, School of Medicine and Health, TUM University Hospital, Technical University of Munich, Munich, Germany
| | - Felix Wirth
- Department of Cardiovascular Surgery, German Heart Center Munich, School of Medicine and Health, TUM University Hospital, Technical University of Munich, Munich, Germany
- Institute Insure, German Heart Center Munich, School of Medicine and Health, TUM University Hospital, Technical University of Munich, Munich, Germany
| | - Stephan Baldus
- Department of Cardiology, Faculty of Medicine, Heart Center, University of Cologne, Cologne, Germany
| | - Malte Kelm
- Department of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty of the Heinrich Heine University, Düsseldorf, Germany
| | - Moritz von Scheidt
- Technical University of Munich, School of Medicine and Health, Department of Cardiovascular Diseases, German Heart Centre Munich, TUM University Hospital, Munich, Germany
- German Center for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - Johannes Krefting
- Technical University of Munich, School of Medicine and Health, Department of Cardiovascular Diseases, German Heart Centre Munich, TUM University Hospital, Munich, Germany
- German Center for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - David Ellinghaus
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Aeron M. Small
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
- Center for Genomic Medicine and Cardiovascular Research Center, Massachusetts General Hospital, Boston
- Program in Medical and Population Genetics and the Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Gina M. Peloso
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts
| | - Pradeep Natarajan
- Center for Genomic Medicine and Cardiovascular Research Center, Massachusetts General Hospital, Boston
- Program in Medical and Population Genetics and the Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - George Thanassoulis
- Division of Experimental Medicine, McGill University, Montreal, Canada
- Preventive and Genomic Cardiology, McGill University Health Centre and Research Institute, Montreal, Canada
| | - James C. Engert
- Division of Experimental Medicine, McGill University, Montreal, Canada
- Preventive and Genomic Cardiology, McGill University Health Centre and Research Institute, Montreal, Canada
| | - Line Dufresne
- Preventive and Genomic Cardiology, McGill University Health Centre and Research Institute, Montreal, Canada
| | - Andre Franke
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Siegfried Görg
- Institute of Transfusion Medicine, University Hospital of Schleswig-Holstein, Kiel, Germany
| | - Matthias Laudes
- Institute for Diabetes and Clinical Metabolic Research, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Ulrike Nowak-Göttl
- Thrombosis and Hemostasis Unit, Institute of Clinical Chemistry, University Hospital Kiel, Kiel, Germany
| | - Mariliis Vaht
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Andres Metspalu
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Monika Stoll
- Institute of Human Genetics, Division of Genetic Epidemiology, University of Muenster, Muenster, Germany
| | - Klaus Berger
- Institute of Epidemiology and Social Medicine, University of Münster, Munster, Germany
| | - Costanza Pellegrini
- Technical University of Munich, School of Medicine and Health, Department of Cardiovascular Diseases, German Heart Centre Munich, TUM University Hospital, Munich, Germany
- German Center for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - Adnan Kastrati
- Technical University of Munich, School of Medicine and Health, Department of Cardiovascular Diseases, German Heart Centre Munich, TUM University Hospital, Munich, Germany
- German Center for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - Christian Hengstenberg
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Chim C. Lang
- Division of Molecular and Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, United Kingdom
| | - Thorsten Kessler
- Technical University of Munich, School of Medicine and Health, Department of Cardiovascular Diseases, German Heart Centre Munich, TUM University Hospital, Munich, Germany
- German Center for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - Iiris Hovatta
- SleepWell Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Georg Nickenig
- Department of Medicine II, Heart Center Bonn, University of Bonn and University Hospital Bonn, Bonn, Germany
| | - Markus M. Nöthen
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
| | - Markus Krane
- German Center for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
- Department of Cardiovascular Surgery, German Heart Center Munich, School of Medicine and Health, TUM University Hospital, Technical University of Munich, Munich, Germany
- Institute Insure, German Heart Center Munich, School of Medicine and Health, TUM University Hospital, Technical University of Munich, Munich, Germany
- Yale School of Medicine, Division of Cardiac Surgery, Department of Surgery, New Haven, Connecticut
| | - Heribert Schunkert
- Technical University of Munich, School of Medicine and Health, Department of Cardiovascular Diseases, German Heart Centre Munich, TUM University Hospital, Munich, Germany
- German Center for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - Nilesh J. Samani
- Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom
- National Institute for Health and Care Research Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, United Kingdom
| | - Johannes Schumacher
- Institute of Human Genetics, Philipps University of Marburg, Marburg, Germany
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
| |
Collapse
|
8
|
Zang K, Brossard M, Wilson T, Ali SA, Espin-Garcia O. A scoping review of statistical methods to investigate colocalization between genetic associations and microRNA expression in osteoarthritis. OSTEOARTHRITIS AND CARTILAGE OPEN 2024; 6:100540. [PMID: 39640910 PMCID: PMC11617925 DOI: 10.1016/j.ocarto.2024.100540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 10/31/2024] [Indexed: 12/07/2024] Open
Abstract
Background Genetic colocalization analysis is a statistical method that evaluates whether two traits (e.g., osteoarthritis [OA] risk and microRNA [miRNA] expression levels) share the same or distinct genetic association signals in a locus typically identified in genome-wide association studies (GWAS). This method is useful for providing insights into the biological relevance of genetic association signals, particularly in intergenic regions, which can help to elucidate disease mechanisms in OA and other complex traits. Objectives To review the existing literature on genetic colocalization methods, assess their suitability for studying OA, and investigate their capacity to integrate miRNA data, while bearing in view their statistical assumptions. Design We followed scoping review methodology and used Covidence software for data management. Search terms for colocalization, GWAS, and genetic or statistical models were used in the databases MEDLINE and EMBASE, searched till March 4, 2024. Results Our search returned 546 peer-reviewed papers, of which 96 were included following title/abstract and full-text screening. Based on both cumulative and annual publication counts, the most cited method for colocalization analysis was coloc. Four papers examined OA-related phenotypes, and none examined miRNA. An approach to colocalization analysis using miRNA was postulated based on further hand-searching. Conclusions Colocalization analysis is a largely unexplored method in OA. Many of the approaches to colocalization analysis identified in this review, including the integration of GWAS and miRNA data, may help to elucidate genetic and epigenetic factors implicated in OA and other complex traits.
Collapse
Affiliation(s)
- Kathleen Zang
- Department of Epidemiology and Biostatistics, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, ON, Canada
| | - Myriam Brossard
- Department of Epidemiology and Biostatistics, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, ON, Canada
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON, Canada
| | - Thomas Wilson
- Henry Ford Health + Michigan State University Health Sciences, Detroit, MI, USA
| | - Shabana Amanda Ali
- Henry Ford Health + Michigan State University Health Sciences, Detroit, MI, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA
| | - Osvaldo Espin-Garcia
- Department of Epidemiology and Biostatistics, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, ON, Canada
- Department of Biostatistics, Krembil Research Institute and Schroeder Arthritis Institute, University Health Network, Toronto, ON, Canada
| |
Collapse
|
9
|
Rämö JT, Jurgens SJ, Kany S, Choi SH, Wang X, Smirnov AN, Friedman SF, Maddah M, Khurshid S, Ellinor PT, Pirruccello JP. Rare Genetic Variants in LDLR, APOB, and PCSK9 Are Associated With Aortic Stenosis. Circulation 2024; 150:1767-1780. [PMID: 39222019 PMCID: PMC11915876 DOI: 10.1161/circulationaha.124.070982] [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: 06/16/2024] [Accepted: 08/25/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Despite a proposed causal role for LDL-C (low-density lipoprotein cholesterol) in aortic stenosis (AS), randomized controlled trials of lipid-lowering therapy failed to prevent severe AS. We aimed to assess the impact on AS and peak velocity across the aortic valve conferred by lifelong alterations in LDL-C levels mediated by protein-disrupting variants in 3 clinically significant genes for LDL (low-density lipoprotein) metabolism (LDLR, APOB, and PCSK9). METHODS We used sequencing data and electronic health records from UK Biobank (UKB) and All of Us and magnetic resonance imaging data from UKB. We identified predicted protein-disrupting variants with the Loss Of Function Transcript Effect Estimator (LOFTEE) and AlphaMissense algorithms and evaluated their associations with LDL-C and peak velocity across the aortic valve (UK Biobank), as well as diagnosed AS and aortic valve replacement (UK Biobank and All of Us). RESULTS We included 421 049 unrelated participants (5621 with AS) in UKB and 195 519 unrelated participants (1087 with AS) in All of Us. Carriers of protein-disrupting variants in LDLR had higher mean LDL-C (UKB: +42.6 mg/dL; P=4.4e-237) and greater risk of AS (meta-analysis: odds ratio, 3.52 [95% CI, 2.39-5.20]; P=2.3e-10) and aortic valve replacement (meta-analysis: odds ratio, 3.78 [95% CI, 2.26-6.32]; P=4.0e-7). Carriers of protein-disrupting variants in APOB or PCSK9 had lower mean LDL-C (UKB: -32.3 mg/dL; P<5e-324) and lower risk of AS (meta-analysis: odds ratio, 0.49 [95% CI, 0.31-0.75]; P=0.001) and aortic valve replacement (meta-analysis: odds ratio, 0.54 [95% CI, 0.30-0.97]; P=0.04). Among 57 371 UKB imaging substudy participants, peak velocities across the aortic valve were greater in carriers of protein-disrupting variants in LDLR (+12.2 cm/s; P=1.6e-5) and lower in carriers of protein-disrupting variants in PCSK9 (-6.9 cm/s; P=0.022). CONCLUSIONS Rare genetic variants that confer lifelong higher or lower LDL-C levels are associated with substantially increased and decreased risk of AS, respectively. Early and sustained lipid-lowering therapy may slow or prevent AS development.
Collapse
Affiliation(s)
- Joel T Rämö
- Cardiovascular Disease Initiative (J.T.R., S.J.J., S. Kany, X.W., S. Khurshid, P.T.E., J.P.P.), Cambridge, MA
- Broad Institute of MIT and Harvard (J.T.R., S.J.J., S. Kany, X.W., S. Khurshid, P.T.E., J.P.P, A.N.S., S.F.F., M.M.), Cambridge, MA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA (J.T.R.)
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki (J.T.R.)
| | - Sean J Jurgens
- Cardiovascular Disease Initiative (J.T.R., S.J.J., S. Kany, X.W., S. Khurshid, P.T.E., J.P.P.), Cambridge, MA
- Broad Institute of MIT and Harvard (J.T.R., S.J.J., S. Kany, X.W., S. Khurshid, P.T.E., J.P.P, A.N.S., S.F.F., M.M.), Cambridge, MA
- Cardiovascular Research Center (J.T.R., S.J.J., S. Kany, S. Khurshid, P.T.E.), Massachusetts General Hospital, Boston
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam University Medical Center, University of Amsterdam, The Netherlands (S.J.J.)
| | - Shinwan Kany
- Cardiovascular Disease Initiative (J.T.R., S.J.J., S. Kany, X.W., S. Khurshid, P.T.E., J.P.P.), Cambridge, MA
- Broad Institute of MIT and Harvard (J.T.R., S.J.J., S. Kany, X.W., S. Khurshid, P.T.E., J.P.P, A.N.S., S.F.F., M.M.), Cambridge, MA
- Cardiovascular Research Center (J.T.R., S.J.J., S. Kany, S. Khurshid, P.T.E.), Massachusetts General Hospital, Boston
- Department of Cardiology, University Heart and Vascular Center Hamburg-Eppendorf, Hamburg, Germany (S. Kany)
- German Center for Cardiovascular Research, Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany (S. Kany)
| | | | - Xin Wang
- Cardiovascular Disease Initiative (J.T.R., S.J.J., S. Kany, X.W., S. Khurshid, P.T.E., J.P.P.), Cambridge, MA
- Broad Institute of MIT and Harvard (J.T.R., S.J.J., S. Kany, X.W., S. Khurshid, P.T.E., J.P.P, A.N.S., S.F.F., M.M.), Cambridge, MA
- Wellcome Trust Sanger Institute, Hinxton, United Kingdom (X.W.)
- University of Cambridge, United Kingdom (X.W.)
| | - Andrey N Smirnov
- Broad Institute of MIT and Harvard (J.T.R., S.J.J., S. Kany, X.W., S. Khurshid, P.T.E., J.P.P, A.N.S., S.F.F., M.M.), Cambridge, MA
| | - Samuel F Friedman
- Cardiovascular Disease Initiative (J.T.R., S.J.J., S. Kany, X.W., S. Khurshid, P.T.E., J.P.P.), Cambridge, MA
- Broad Institute of MIT and Harvard (J.T.R., S.J.J., S. Kany, X.W., S. Khurshid, P.T.E., J.P.P, A.N.S., S.F.F., M.M.), Cambridge, MA
| | - Mahnaz Maddah
- Broad Institute of MIT and Harvard (J.T.R., S.J.J., S. Kany, X.W., S. Khurshid, P.T.E., J.P.P, A.N.S., S.F.F., M.M.), Cambridge, MA
| | - Shaan Khurshid
- Broad Institute of MIT and Harvard (J.T.R., S.J.J., S. Kany, X.W., S. Khurshid, P.T.E., J.P.P, A.N.S., S.F.F., M.M.), Cambridge, MA
- Cardiovascular Research Center (J.T.R., S.J.J., S. Kany, S. Khurshid, P.T.E.), Massachusetts General Hospital, Boston
- Telemachus and Irene Demoulas Family Foundation Center for Cardiac Arrhythmias (S. Khurshid, P.T.E.), Massachusetts General Hospital, Boston
| | - Patrick T Ellinor
- Broad Institute of MIT and Harvard (J.T.R., S.J.J., S. Kany, X.W., S. Khurshid, P.T.E., J.P.P, A.N.S., S.F.F., M.M.), Cambridge, MA
- Cardiovascular Research Center (J.T.R., S.J.J., S. Kany, S. Khurshid, P.T.E.), Massachusetts General Hospital, Boston
- Telemachus and Irene Demoulas Family Foundation Center for Cardiac Arrhythmias (S. Khurshid, P.T.E.), Massachusetts General Hospital, Boston
- Cardiology Division (P.T.E.), Massachusetts General Hospital, Boston
- Harvard Medical School, Boston, MA (P.T.E.)
| | - James P Pirruccello
- Cardiovascular Disease Initiative (J.T.R., S.J.J., S. Kany, X.W., S. Khurshid, P.T.E., J.P.P.), Cambridge, MA
- Broad Institute of MIT and Harvard (J.T.R., S.J.J., S. Kany, X.W., S. Khurshid, P.T.E., J.P.P, A.N.S., S.F.F., M.M.), Cambridge, MA
- Division of Cardiology (J.P.P.), University of California San Francisco
- Bakar Computational Health Sciences Institute (J.P.P.), University of California San Francisco
- Institute for Human Genetics (J.P.P.), University of California San Francisco
| |
Collapse
|
10
|
Ma XR, Conley SD, Kosicki M, Bredikhin D, Cui R, Tran S, Sheth MU, Qiu WL, Chen S, Kundu S, Kang HY, Amgalan D, Munger CJ, Duan L, Dang K, Rubio OM, Kany S, Zamirpour S, DePaolo J, Padmanabhan A, Olgin J, Damrauer S, Andersson R, Gu M, Priest JR, Quertermous T, Qiu X, Rabinovitch M, Visel A, Pennacchio L, Kundaje A, Glass IA, Gifford CA, Pirruccello JP, Goodyer WR, Engreitz JM. Molecular convergence of risk variants for congenital heart defects leveraging a regulatory map of the human fetal heart. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.11.20.24317557. [PMID: 39606363 PMCID: PMC11601760 DOI: 10.1101/2024.11.20.24317557] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Congenital heart defects (CHD) arise in part due to inherited genetic variants that alter genes and noncoding regulatory elements in the human genome. These variants are thought to act during fetal development to influence the formation of different heart structures. However, identifying the genes, pathways, and cell types that mediate these effects has been challenging due to the immense diversity of cell types involved in heart development as well as the superimposed complexities of interpreting noncoding sequences. As such, understanding the molecular functions of both noncoding and coding variants remains paramount to our fundamental understanding of cardiac development and CHD. Here, we created a gene regulation map of the healthy human fetal heart across developmental time, and applied it to interpret the functions of variants associated with CHD and quantitative cardiac traits. We collected single-cell multiomic data from 734,000 single cells sampled from 41 fetal hearts spanning post-conception weeks 6 to 22, enabling the construction of gene regulation maps in 90 cardiac cell types and states, including rare populations of cardiac conduction cells. Through an unbiased analysis of all 90 cell types, we find that both rare coding variants associated with CHD and common noncoding variants associated with valve traits converge to affect valvular interstitial cells (VICs). VICs are enriched for high expression of known CHD genes previously identified through mapping of rare coding variants. Eight CHD genes, as well as other genes in similar molecular pathways, are linked to common noncoding variants associated with other valve diseases or traits via enhancers in VICs. In addition, certain common noncoding variants impact enhancers with activities highly specific to particular subanatomic structures in the heart, illuminating how such variants can impact specific aspects of heart structure and function. Together, these results implicate new enhancers, genes, and cell types in the genetic etiology of CHD, identify molecular convergence of common noncoding and rare coding variants on VICs, and suggest a more expansive view of the cell types instrumental in genetic risk for CHD, beyond the working cardiomyocyte. This regulatory map of the human fetal heart will provide a foundational resource for understanding cardiac development, interpreting genetic variants associated with heart disease, and discovering targets for cell-type specific therapies.
Collapse
Affiliation(s)
- X Rosa Ma
- Basic Science and Engineering (BASE) Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Stephanie D Conley
- Basic Science and Engineering (BASE) Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Michael Kosicki
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Danila Bredikhin
- Basic Science and Engineering (BASE) Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Ran Cui
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Steven Tran
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Maya U Sheth
- Basic Science and Engineering (BASE) Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Wei-Lin Qiu
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Sijie Chen
- Basic Science and Engineering (BASE) Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Soumya Kundu
- Department of Genetics, Stanford University, Stanford, CA, USA
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Helen Y Kang
- Basic Science and Engineering (BASE) Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
- Current address: PhD Program in Computational and Systems Biology, MIT, Cambridge, MA, USA
| | - Dulguun Amgalan
- Basic Science and Engineering (BASE) Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
- Maternal and Child Health Research Institute, Stanford University, Stanford, CA, USA
| | - Chad J Munger
- Basic Science and Engineering (BASE) Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Lauren Duan
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Katherine Dang
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Oriane Matthys Rubio
- Basic Science and Engineering (BASE) Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA, USA
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Shinwan Kany
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Department of Cardiology, University Heart and Vascular Center Hamburg-Eppendorf, Hamburg, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Siavash Zamirpour
- School of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - John DePaolo
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Arun Padmanabhan
- Gladstone Institutes, San Francisco, CA, USA
- Department of Medicine, University of California San Francisco School of Medicine, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Jeffrey Olgin
- Division of Cardiology, Department of Medicine and Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
| | - Scott Damrauer
- Division of Vascular Surgery and Endovascular Therapy, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Corporal Michael Crescenz VA Medical Center, Philadelphia, PA, USA
- Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Robin Andersson
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Mingxia Gu
- Center for Stem Cell and Organoid Medicine, Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- University of Cincinnati School of Medicine, Cincinnati, OH, USA
| | - James R Priest
- Department of Pediatrics, Stanford University, Stanford, CA, USA
- Tenaya Therapeutics, South San Francisco, CA, USA
| | - Thomas Quertermous
- Department of Genetics, Stanford University, Stanford, CA, USA
- Department of Medicine, Stanford University, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | - Xiaojie Qiu
- Basic Science and Engineering (BASE) Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
- Department of Computer Science, Stanford University, Stanford, CA, USA
- Maternal and Child Health Research Institute, Stanford University, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
- Stanford Bio-X, Stanford University, Stanford, CA, USA
| | - Marlene Rabinovitch
- Basic Science and Engineering (BASE) Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA, USA
- Department of Pediatrics, Stanford University, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
- Vera Moulton Wall Center for Pulmonary Vascular Diseases, Stanford University, Stanford, CA, USA
| | - Axel Visel
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- US Department of Energy Joint Genome Institute, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- School of Natural Sciences, University of California, Merced, Merced, CA, USA
| | - Len Pennacchio
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- US Department of Energy Joint Genome Institute, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- Comparative Biochemistry Program, University of California, Berkeley, CA, 94720, USA
| | - Anshul Kundaje
- Department of Genetics, Stanford University, Stanford, CA, USA
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Ian A Glass
- Maternal and Child Health Research Institute, Stanford University, Stanford, CA, USA
- Department of Pediatrics and Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Casey A Gifford
- Basic Science and Engineering (BASE) Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
- Department of Pediatrics, Stanford University, Stanford, CA, USA
- Maternal and Child Health Research Institute, Stanford University, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA
| | - James P Pirruccello
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of Cardiology, Department of Medicine and Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
- Institute for Human Genetics, University of California, San Francisco, CA, USA
- Bakar Computation Health Sciences Institute, University of California, San Francisco, CA, USA
| | - William R Goodyer
- Department of Pediatrics, Stanford University, Stanford, CA, USA
- Maternal and Child Health Research Institute, Stanford University, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | - Jesse M Engreitz
- Basic Science and Engineering (BASE) Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Maternal and Child Health Research Institute, Stanford University, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
- Gene Regulation Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
11
|
Vassiliou VS, Johnson N, Langlands K, Tsampasian V. Genetics of Calcific Aortic Stenosis: A Systematic Review. Genes (Basel) 2024; 15:1309. [PMID: 39457433 PMCID: PMC11508093 DOI: 10.3390/genes15101309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 09/27/2024] [Accepted: 10/03/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Calcific aortic stenosis is the most prevalent valvular abnormality in the Western world. Factors commonly associated with calcific aortic stenosis include advanced age, male sex, hypertension, diabetes and impaired renal function. This review synthesises the existing literature on genetic associations with calcific aortic stenosis. Methods: A systematic search was conducted in the PubMed, Ovid and Cochrane libraries from inception to 21 July 2024 to identify human studies investigating the genetic factors involved in calcific aortic stenosis. From an initial pool of 1392 articles, 78 were selected for full-text review and 31 were included in the final qualitative synthesis. The risk of bias in these studies was assessed using the Newcastle Ottawa Scale. Results: Multiple genes have been associated with calcific aortic stenosis. These genes are involved in different biological pathways, including the lipid metabolism pathway (PLA, LDL, APO, PCSK9, Lp-PLA2, PONS1), the inflammatory pathway (IL-6, IL-10), the calcification pathway (PALMD, TEX41) and the endocrine pathway (PTH, VIT D, RUNX2, CACNA1C, ALPL). Additional genes such as NOTCH1, NAV1 and FADS1/2 influence different pathways. Mechanistically, these genes may promote a pro-inflammatory and pro-calcific environment in the aortic valve itself, leading to increased osteoblastic activity and subsequent calcific degeneration of the valve. Conclusions: Numerous genetic associations contribute to calcific aortic stenosis. Recognition of these associations can enhance risk stratification for individuals and their first-degree relatives, facilitate family screening, and importantly, pave the way for targeted therapeutic interventions focusing on the identified genetic factors. Understanding these genetic factors can also lead to gene therapy to prevent calcific aortic stenosis in the future.
Collapse
Affiliation(s)
- Vassilios S. Vassiliou
- Norwich Medical School, University of East Anglia, Norwich NR4 7TJ, UK; (N.J.); (V.T.)
- Fitzwilliam College, University of Cambridge, Cambridge CB3 0DG, UK
| | - Nicholas Johnson
- Norwich Medical School, University of East Anglia, Norwich NR4 7TJ, UK; (N.J.); (V.T.)
| | - Kenneth Langlands
- Institute of Continuing Education, University of Cambridge, Cambridge CB23 8AQ, UK;
| | - Vasiliki Tsampasian
- Norwich Medical School, University of East Anglia, Norwich NR4 7TJ, UK; (N.J.); (V.T.)
| |
Collapse
|
12
|
Takeji Y, Tada H, Taniguchi T, Sakata K, Kitai T, Shirai S, Takamura M. Current Management and Therapy of Severe Aortic Stenosis and Future Perspective. J Atheroscler Thromb 2024; 31:1353-1364. [PMID: 39111841 PMCID: PMC11456350 DOI: 10.5551/jat.rv22023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 06/12/2024] [Indexed: 10/04/2024] Open
Abstract
Intervention for severe aortic stenosis (AS) has dramatically progressed since the introduction of transcatheter aortic valve replacement (TAVR). Decades ago, controversies existed regarding comparing clinical outcomes between TAVR and surgical aortic valve replacement (SAVR) in various risk profiles. Recently, we discussed the durability of transcatheter heart valves and their lifetime management after aortic valve replacement (AVR). Regarding the management of AS, we discuss the appropriate timing of intervention for severe aortic stenosis, especially in asymptomatic patients. In spite of dramatic progression of intervention for AS, there are no established medications available to prevent or slow the progression of AS at present. Basic research and genome studies have suggested several targets associated with the progression of aortic valve calcification. Randomized controlled trials evaluating the efficacy of medications to prevent AS progression are ongoing, which might lead to new strategies for AS management. In this review, we summarize the current management of AS and the drugs expected to prevent the progression of AS.
Collapse
Affiliation(s)
- Yasuaki Takeji
- Department of Cardiovascular Medicine, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Hayato Tada
- Department of Cardiovascular Medicine, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Tomohiko Taniguchi
- Department of Cardiovascular Medicine, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Kenji Sakata
- Department of Cardiovascular Medicine, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Takeshi Kitai
- Department of Heart Failure and Transplantation, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Shinichi Shirai
- Division of Cardiology, Kokura Memorial Hospital, Fukuoka, Japan
| | - Masayuki Takamura
- Department of Cardiovascular Medicine, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| |
Collapse
|
13
|
Zhu L, Li N, Shi H, Shao G, Sun L. Genetic causal association between lipidomic profiles, inflammatory proteomics, and aortic stenosis: a Mendelian randomization investigation. Eur J Med Res 2024; 29:446. [PMID: 39217396 PMCID: PMC11365128 DOI: 10.1186/s40001-024-02014-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 08/06/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Aortic stenosis (AS) is a prevalent and serious valvular heart disease with a complex etiology involving genetic predispositions, lipid dysregulation, and inflammation. The specific roles of lipid and protein biomarkers in AS development are not fully elucidated. This study aimed to elucidate the causal relationships between lipidome, inflammatory proteins, and AS using Mendelian randomization (MR), identifying potential therapeutic targets. METHODS Utilizing data from large-scale genome-wide association studies (GWAS) and genome-wide protein quantitative trait loci (pQTL) studies, we conducted MR analyses on 179 plasma lipidome and 91 inflammatory proteins to assess their causal associations with AS. Our approach included Inverse Variance Weighting (IVW), Wald ratio, and robust adjusted profile score (RAPS) analyses to refine these associations. MR-Egger regression was used to address directional horizontal pleiotropy. RESULTS Our MR analysis showed that genetically predicted 50 lipids were associated with AS, including 38 as risk factors [(9 Sterol ester, 18 Phosphatidylcholine, 4 Phosphatidylethanolamine, 1 Phosphatidylinositol and 6 Triacylglycerol)] and 12 as protective. Sterol ester (27:1/17:1) emerged as the most significant risk factor with an odds ratio (OR) of 3.11. Additionally, two inflammatory proteins, fibroblast growth factor 19 (FGF19) (OR = 0.830, P = 0.015), and interleukin 6 (IL-6) (OR = 0.729, P = 1.79E-04) were significantly associated with reduced AS risk. However, a two-step MR analysis showed no significant mediated correlations between these proteins and the lipid-AS pathway. CONCLUSION This study reveals complex lipid and protein interactions in AS, identifying potential molecular targets for therapy. These results go beyond traditional lipid profiling and significantly advance our genetic and molecular understanding of AS, highlighting potential pathways for intervention and prevention.
Collapse
Affiliation(s)
- Linwen Zhu
- Department of Cardiovascular Surgery, Lihuili Hospital Affiliated to Ningbo University, Ningbo, 315041, Zhejiang, China
| | - Ni Li
- Department of Cardiovascular Surgery, Lihuili Hospital Affiliated to Ningbo University, Ningbo, 315041, Zhejiang, China
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Huoshun Shi
- Department of Cardiovascular Surgery, Lihuili Hospital Affiliated to Ningbo University, Ningbo, 315041, Zhejiang, China
| | - Guofeng Shao
- Department of Cardiovascular Surgery, Lihuili Hospital Affiliated to Ningbo University, Ningbo, 315041, Zhejiang, China.
| | - Lebo Sun
- Department of Cardiovascular Surgery, Lihuili Hospital Affiliated to Ningbo University, Ningbo, 315041, Zhejiang, China.
| |
Collapse
|
14
|
Small AM, Yutzey KE, Binstadt BA, Voigts Key K, Bouatia-Naji N, Milan D, Aikawa E, Otto CM, St Hilaire C. Unraveling the Mechanisms of Valvular Heart Disease to Identify Medical Therapy Targets: A Scientific Statement From the American Heart Association. Circulation 2024; 150:e109-e128. [PMID: 38881493 PMCID: PMC11542557 DOI: 10.1161/cir.0000000000001254] [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: 06/18/2024]
Abstract
Valvular heart disease is a common cause of morbidity and mortality worldwide and has no effective medical therapy. Severe disease is managed with valve replacement procedures, which entail high health care-related costs and postprocedural morbidity and mortality. Robust ongoing research programs have elucidated many important molecular pathways contributing to primary valvular heart disease. However, there remain several key challenges inherent in translating research on valvular heart disease to viable molecular targets that can progress through the clinical trials pathway and effectively prevent or modify the course of these common conditions. In this scientific statement, we review the basic cellular structures of the human heart valves and discuss how these structures change in primary valvular heart disease. We focus on the most common primary valvular heart diseases, including calcific aortic stenosis, bicuspid aortic valves, mitral valve prolapse, and rheumatic heart disease, and outline the fundamental molecular discoveries contributing to each. We further outline potential therapeutic molecular targets for primary valvular heart disease and discuss key knowledge gaps that might serve as future research priorities.
Collapse
|
15
|
Yang TY, Small AM, Dufresne L, Peloso GM, Natarajan P, Engert JC, Thanassoulis G. Plasma Proteomic Biomarkers of Aortic Stenosis: A Mendelian Randomization Study. J Am Coll Cardiol 2024; 84:592-594. [PMID: 39084832 DOI: 10.1016/j.jacc.2024.05.046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 04/03/2024] [Accepted: 05/10/2024] [Indexed: 08/02/2024]
Affiliation(s)
- Ta-Yu Yang
- McGill University Health Centre and Research Institute, Montréal, Québec, Canada; McGill University, Montréal, Québec, Canada; Kyoto-McGill International Collaborative School in Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Aeron M Small
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA; Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Line Dufresne
- McGill University Health Centre and Research Institute, Montréal, Québec, Canada
| | - Gina M Peloso
- Boston University School of Public Health, Boston, Massachusetts, USA
| | - Pradeep Natarajan
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA; Massachusetts General Hospital, Boston, Massachusetts, USA; Harvard Medical School, Boston, Massachusetts, USA
| | - James C Engert
- McGill University Health Centre and Research Institute, Montréal, Québec, Canada; McGill University, Montréal, Québec, Canada
| | - George Thanassoulis
- McGill University Health Centre and Research Institute, Montréal, Québec, Canada; McGill University, Montréal, Québec, Canada.
| |
Collapse
|
16
|
Irtyuga O, Skitchenko R, Babakekhyan M, Usoltsev D, Tarnovskaya S, Malashicheva A, Fomicheva Y, Rotar O, Moiseeva O, Shadrina U, Artomov M, Kostareva A, Shlyakhto E. The Role of NOTCH Pathway Genes in the Inherited Susceptibility to Aortic Stenosis. J Cardiovasc Dev Dis 2024; 11:226. [PMID: 39057646 PMCID: PMC11277067 DOI: 10.3390/jcdd11070226] [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: 05/15/2024] [Revised: 06/26/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
The NOTCH-signaling pathway is responsible for intercellular interactions and cell fate commitment. Recently, NOTCH pathway genes were demonstrated to play an important role in aortic valve development, leading to an increased calcified aortic valve disease (CAVD) later in life. Here, we further investigate the association between genetic variants in the NOTCH pathway genes and aortic stenosis in a case-control study of 90 CAVD cases and 4723 controls using target panel sequencing of full-length 20 genes from a NOTCH-related pathway (DVL2, DTX2, MFNG, NUMBL, LFNG, DVL1, DTX4, APH1A, DTX1, APH1B, NOTCH1, ADAM17, DVL3, NCSTN, DTX3L, ILK, RFNG, DTX3, NOTCH4, PSENEN). We identified a common intronic variant in NOTCH1, protecting against CAVD development (rs3812603), as well as several rare and unique new variants in NOTCH-pathway genes (DTX4, NOTCH1, DTX1, DVL2, NOTCH1, DTX3L, DVL3), with a prominent effect of the protein structure and function.
Collapse
Affiliation(s)
- Olga Irtyuga
- Almazov National Medical Research Centre, 197341 Saint-Petersburg, Russia; (R.S.); (M.B.); (D.U.); (S.T.); (A.M.); (Y.F.); (O.R.); (O.M.); (U.S.); (M.A.); (A.K.); (E.S.)
| | - Rostislav Skitchenko
- Almazov National Medical Research Centre, 197341 Saint-Petersburg, Russia; (R.S.); (M.B.); (D.U.); (S.T.); (A.M.); (Y.F.); (O.R.); (O.M.); (U.S.); (M.A.); (A.K.); (E.S.)
| | - Mary Babakekhyan
- Almazov National Medical Research Centre, 197341 Saint-Petersburg, Russia; (R.S.); (M.B.); (D.U.); (S.T.); (A.M.); (Y.F.); (O.R.); (O.M.); (U.S.); (M.A.); (A.K.); (E.S.)
| | - Dmitrii Usoltsev
- Almazov National Medical Research Centre, 197341 Saint-Petersburg, Russia; (R.S.); (M.B.); (D.U.); (S.T.); (A.M.); (Y.F.); (O.R.); (O.M.); (U.S.); (M.A.); (A.K.); (E.S.)
- Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Svetlana Tarnovskaya
- Almazov National Medical Research Centre, 197341 Saint-Petersburg, Russia; (R.S.); (M.B.); (D.U.); (S.T.); (A.M.); (Y.F.); (O.R.); (O.M.); (U.S.); (M.A.); (A.K.); (E.S.)
| | - Anna Malashicheva
- Almazov National Medical Research Centre, 197341 Saint-Petersburg, Russia; (R.S.); (M.B.); (D.U.); (S.T.); (A.M.); (Y.F.); (O.R.); (O.M.); (U.S.); (M.A.); (A.K.); (E.S.)
| | - Yulya Fomicheva
- Almazov National Medical Research Centre, 197341 Saint-Petersburg, Russia; (R.S.); (M.B.); (D.U.); (S.T.); (A.M.); (Y.F.); (O.R.); (O.M.); (U.S.); (M.A.); (A.K.); (E.S.)
| | - Oksana Rotar
- Almazov National Medical Research Centre, 197341 Saint-Petersburg, Russia; (R.S.); (M.B.); (D.U.); (S.T.); (A.M.); (Y.F.); (O.R.); (O.M.); (U.S.); (M.A.); (A.K.); (E.S.)
| | - Olga Moiseeva
- Almazov National Medical Research Centre, 197341 Saint-Petersburg, Russia; (R.S.); (M.B.); (D.U.); (S.T.); (A.M.); (Y.F.); (O.R.); (O.M.); (U.S.); (M.A.); (A.K.); (E.S.)
| | - Ulyana Shadrina
- Almazov National Medical Research Centre, 197341 Saint-Petersburg, Russia; (R.S.); (M.B.); (D.U.); (S.T.); (A.M.); (Y.F.); (O.R.); (O.M.); (U.S.); (M.A.); (A.K.); (E.S.)
| | - Mykyta Artomov
- Almazov National Medical Research Centre, 197341 Saint-Petersburg, Russia; (R.S.); (M.B.); (D.U.); (S.T.); (A.M.); (Y.F.); (O.R.); (O.M.); (U.S.); (M.A.); (A.K.); (E.S.)
- Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children’s Hospital, Columbus, OH 43205, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH 43205, USA
| | - Anna Kostareva
- Almazov National Medical Research Centre, 197341 Saint-Petersburg, Russia; (R.S.); (M.B.); (D.U.); (S.T.); (A.M.); (Y.F.); (O.R.); (O.M.); (U.S.); (M.A.); (A.K.); (E.S.)
- Department of Women’s and Children’s Health and Centre for Molecular Medicine, Karolinska Institute, 17176 Stockholm, Sweden
| | - Evgeny Shlyakhto
- Almazov National Medical Research Centre, 197341 Saint-Petersburg, Russia; (R.S.); (M.B.); (D.U.); (S.T.); (A.M.); (Y.F.); (O.R.); (O.M.); (U.S.); (M.A.); (A.K.); (E.S.)
| |
Collapse
|
17
|
Xie L, Gan W, Cai G. The causal relationship between gut microbiota and diabetic neuropathy: a bi-directional two-sample Mendelian randomization study. Front Endocrinol (Lausanne) 2024; 15:1402014. [PMID: 39050567 PMCID: PMC11266094 DOI: 10.3389/fendo.2024.1402014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 06/26/2024] [Indexed: 07/27/2024] Open
Abstract
Background Many studies suggest a strong correlation between gut microbiota (GM) and diabetic neuropathy (DN). However, the precise causal relationship between GM and DN has yet to be fully elucidated. Hence, a bi-directional Mendelian randomization (MR) analysis was used to examine the association between GM and DN. Methods Widely known genome-wide association study (GWAS) of GM was collected from the MiBio Gen project. Summary-level datasets for DN were taken from the FinnGen project. Inverse variance weighted approach was used for evaluating the causal relationship between GM and DN. Subsequently, pleiotropy and heterogeneity tests were performed to verify the reliability of the data. Furthermore, a bidirectional two-sample MR analysis was done to investigate the directionality of the causal relationships. Gene Ontology analysis was conducted to identify the associations that could indicate biological functions. Results We identified potential causal associations between GM and DN (p< 0.05 in all three MR methods). Among them, we found increased levels of Christensenellaceae R-7 (Odds ratio, OR= 1.52; 95% confidence interval, CI = 1.03-2.23; p = 0.03), Ruminococcaceae UCG013 (OR =1.35; 95% CI = 1.00-1.85; p = 0.04), and Eggerthella groups (OR = 1.27; 95% CI = 1.05-1.55; p = 0.01), which may be associated with a higher risk of DN, while increased levels of Peptococcaceae (OR = 0.69; 95% CI = 0.54-0.90; p< 0.01) and Eubacterium coprostanoligenes groups (OR = 0.68; 95% CI = 0.49-0.93; p = 0.01) could be associated with a lower risk. Gene Ontology pathway analysis revealed enrichment of genes regulated by the associated single-nucleotide polymorphisms (SNPs) in the apical plasma membrane, glycosyltransferase activity, hexosyltransferase activity and membrane raft. Reverse MR analyses indicated that DN was associated with five microbial taxa in all three MR methods. Conclusion The results of our study validate the possible causative relationship between GM and DN. This discovery gives new perspectives into the mechanism on how GM influences DN, and establishes a theoretical foundation for future investigations into targeted preventive measures.
Collapse
Affiliation(s)
- Long Xie
- Department of Orthopedics, The Fourth Hospital of Changsha (The Changsha Affiliated Hospital of Hunan Normal University), Hunan Normal University, Changsha, China
| | - Wen Gan
- Department of Thoracic Surgery, Yuebei People’s Hospital, Shaoguan, Guangdong, China
| | - GuangRong Cai
- Trauma Department of Orthopaedics, Yuebei People’s Hospital, Shaoguan, Guangdong, China
| |
Collapse
|
18
|
Zheng S, Tsao PS, Pan C. Abdominal aortic aneurysm and cardiometabolic traits share strong genetic susceptibility to lipid metabolism and inflammation. Nat Commun 2024; 15:5652. [PMID: 38969659 PMCID: PMC11226445 DOI: 10.1038/s41467-024-49921-7] [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: 12/05/2023] [Accepted: 06/25/2024] [Indexed: 07/07/2024] Open
Abstract
Abdominal aortic aneurysm has a high heritability and often co-occurs with other cardiometabolic disorders, suggesting shared genetic susceptibility. We investigate this commonality leveraging recent GWAS studies of abdominal aortic aneurysm and 32 cardiometabolic traits. We find significant genetic correlations between abdominal aortic aneurysm and 21 of the cardiometabolic traits investigated, including causal relationships with coronary artery disease, hypertension, lipid traits, and blood pressure. For each trait pair, we identify shared causal variants, genes, and pathways, revealing that cholesterol metabolism and inflammation are shared most prominently. Additionally, we show the tissue and cell type specificity in the shared signals, with strong enrichment across traits in the liver, arteries, adipose tissues, macrophages, adipocytes, and fibroblasts. Finally, we leverage drug-gene databases to identify several lipid-lowering drugs and antioxidants with high potential to treat abdominal aortic aneurysm with comorbidities. Our study provides insight into the shared genetic mechanism between abdominal aortic aneurysm and cardiometabolic traits, and identifies potential targets for pharmacological intervention.
Collapse
Affiliation(s)
- Shufen Zheng
- Center for Intelligent Medicine Research, Greater Bay Area Institute of Precision Medicine (Guangzhou), Guangzhou, China
- Center for Evolutionary Biology, Intelligent Medicine Institute, School of Life Sciences, Fudan University, Shanghai, China
| | - Philip S Tsao
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California, USA.
- Stanford Cardiovascular Institute, Stanford University, California, USA.
- VA Palo Alto Health Care System, Palo Alto, California, USA.
| | - Cuiping Pan
- Center for Intelligent Medicine Research, Greater Bay Area Institute of Precision Medicine (Guangzhou), Guangzhou, China.
- Center for Evolutionary Biology, Intelligent Medicine Institute, School of Life Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
19
|
Diaz N, Perez C, Escribano AM, Sanz G, Priego J, Lafuente C, Barberis M, Calle L, Espinosa JF, Priest BT, Zhang HY, Nosie AK, Haas JV, Cannady E, Borel A, Schultze AE, Sauder JM, Hendle J, Weichert K, Nicholls SJ, Michael LF. Discovery of potent small-molecule inhibitors of lipoprotein(a) formation. Nature 2024; 629:945-950. [PMID: 38720069 PMCID: PMC11111404 DOI: 10.1038/s41586-024-07387-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 04/04/2024] [Indexed: 05/24/2024]
Abstract
Lipoprotein(a) (Lp(a)), an independent, causal cardiovascular risk factor, is a lipoprotein particle that is formed by the interaction of a low-density lipoprotein (LDL) particle and apolipoprotein(a) (apo(a))1,2. Apo(a) first binds to lysine residues of apolipoprotein B-100 (apoB-100) on LDL through the Kringle IV (KIV) 7 and 8 domains, before a disulfide bond forms between apo(a) and apoB-100 to create Lp(a) (refs. 3-7). Here we show that the first step of Lp(a) formation can be inhibited through small-molecule interactions with apo(a) KIV7-8. We identify compounds that bind to apo(a) KIV7-8, and, through chemical optimization and further application of multivalency, we create compounds with subnanomolar potency that inhibit the formation of Lp(a). Oral doses of prototype compounds and a potent, multivalent disruptor, LY3473329 (muvalaplin), reduced the levels of Lp(a) in transgenic mice and in cynomolgus monkeys. Although multivalent molecules bind to the Kringle domains of rat plasminogen and reduce plasmin activity, species-selective differences in plasminogen sequences suggest that inhibitor molecules will reduce the levels of Lp(a), but not those of plasminogen, in humans. These data support the clinical development of LY3473329-which is already in phase 2 studies-as a potent and specific orally administered agent for reducing the levels of Lp(a).
Collapse
Affiliation(s)
- Nuria Diaz
- Lilly Research Laboratories, Alcobendas, Spain
| | | | | | - Gema Sanz
- Lilly Research Laboratories, Alcobendas, Spain
| | | | | | | | - Luis Calle
- Lilly Research Laboratories, Alcobendas, Spain
| | | | | | - Hong Y Zhang
- Lilly Research Laboratories, Indianapolis, IN, USA
| | | | | | | | | | | | | | - Jörg Hendle
- Lilly Research Laboratories, San Diego, CA, USA
| | | | - Stephen J Nicholls
- Victorian Heart Institute, Monash University, Clayton, Victoria, Australia
| | | |
Collapse
|
20
|
Yu M, Bouatia-Naji N. Insights into the Inherited Basis of Valvular Heart Disease. Curr Cardiol Rep 2024; 26:381-392. [PMID: 38581562 DOI: 10.1007/s11886-024-02041-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/07/2024] [Indexed: 04/08/2024]
Abstract
PURPOSE OF REVIEW: Increases in the availability of genetic data and advances in the tools and methods for their analyses have enabled well-powered genetic association studies that have significantly enhanced our understanding of the genetic factors underlying both rare and common valve diseases. Valvular heart diseases, such as congenital valve malformations and degenerative valve lesions, increase the risk of heart failure, arrhythmias, and sudden death. In this review, we provide an updated overview of our current understanding of the genetic mechanisms underlying valvular heart diseases. With a focus on discoveries from the past 5 years, we describe recent insights into genetic risk and underlying biological pathways. RECENT FINDINGS: Recently acquired knowledge around valvular heart disease genetics has provided important insights into novel mechanisms related to disease pathogenesis. Newly identified risk loci associated valvular heart disease mainly regulate the composition of the extracellular matrix, accelerate the endothelial-to-mesenchymal transition, contribute to cilia formation processes, and play roles in lipid metabolism. Large-scale genomic analyses have identified numerous risk loci, genes, and biological pathways associated with degenerative valve disease and congenital valve malformations. Shared risk genes suggest common mechanistic pathways for various valve pathologies. More recent studies have combined cardiac magnetic resonance imaging and machine learning to offer a novel approach for exploring genotype-phenotype relationships regarding valve disease. Progress in the field holds promise for targeted prevention, particularly through the application of polygenic risk scores, and innovative therapies based on the biological mechanisms for predominant forms of valvular heart diseases.
Collapse
Affiliation(s)
- Mengyao Yu
- Shanghai Pudong Hospital, Human Phenome Institute, Fudan University Pudong Medical Center, Zhangjiang Fudan International Innovation Center, Fundan University, 825 Zhangheng Road, Pudong District, Shanghai, 201203, China.
| | | |
Collapse
|
21
|
Sengupta PP, Kluin J, Lee SP, Oh JK, Smits AIPM. The future of valvular heart disease assessment and therapy. Lancet 2024; 403:1590-1602. [PMID: 38554727 DOI: 10.1016/s0140-6736(23)02754-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 08/15/2023] [Accepted: 12/06/2023] [Indexed: 04/02/2024]
Abstract
Valvular heart disease (VHD) is becoming more prevalent in an ageing population, leading to challenges in diagnosis and management. This two-part Series offers a comprehensive review of changing concepts in VHD, covering diagnosis, intervention timing, novel management strategies, and the current state of research. The first paper highlights the remarkable progress made in imaging and transcatheter techniques, effectively addressing the treatment paradox wherein populations at the highest risk of VHD often receive the least treatment. These advances have attracted the attention of clinicians, researchers, engineers, device manufacturers, and investors, leading to the exploration and proposal of treatment approaches grounded in pathophysiology and multidisciplinary strategies for VHD management. This Series paper focuses on innovations involving computational, pharmacological, and bioengineering approaches that are transforming the diagnosis and management of patients with VHD. Artificial intelligence and digital methods are enhancing screening, diagnosis, and planning procedures, and the integration of imaging and clinical data is improving the classification of VHD severity. The emergence of artificial intelligence techniques, including so-called digital twins-eg, computer-generated replicas of the heart-is aiding the development of new strategies for enhanced risk stratification, prognostication, and individualised therapeutic targeting. Various new molecular targets and novel pharmacological strategies are being developed, including multiomics-ie, analytical methods used to integrate complex biological big data to find novel pathways to halt the progression of VHD. In addition, efforts have been undertaken to engineer heart valve tissue and provide a living valve conduit capable of growth and biological integration. Overall, these advances emphasise the importance of early detection, personalised management, and cutting-edge interventions to optimise outcomes amid the evolving landscape of VHD. Although several challenges must be overcome, these breakthroughs represent opportunities to advance patient-centred investigations.
Collapse
Affiliation(s)
- Partho P Sengupta
- Division of Cardiovascular Diseases and Hypertension, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA; Cardiovascular Services, Robert Wood Johnson University Hospital, New Brunswick, NJ, USA.
| | - Jolanda Kluin
- Department of Cardiothoracic Surgery, Erasmus MC Rotterdam, Thorax Center, Rotterdam, Netherlands
| | - Seung-Pyo Lee
- Department of Internal Medicine, Seoul National University Hospital and Seoul National University College of Medicine, Seoul, South Korea
| | - Jae K Oh
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Anthal I P M Smits
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands; Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| |
Collapse
|
22
|
Small AM, Melloni GEM, Kamanu FK, Bergmark BA, Bonaca MP, O’Donoghue ML, Giugliano RP, Scirica BM, Bhatt D, Antman EM, Raz I, Wiviott SD, Truong B, Wilson PWF, Cho K, O’Donnell CJ, Braunwald E, Lubitz SA, Ellinor P, Peloso GM, Ruff CT, Sabatine MS, Natarajan P, Marston NA. Novel Polygenic Risk Score and Established Clinical Risk Factors for Risk Estimation of Aortic Stenosis. JAMA Cardiol 2024; 9:357-366. [PMID: 38416462 PMCID: PMC10902779 DOI: 10.1001/jamacardio.2024.0011] [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: 09/06/2023] [Accepted: 12/28/2023] [Indexed: 02/29/2024]
Abstract
Importance Polygenic risk scores (PRSs) have proven to be as strong as or stronger than established clinical risk factors for many cardiovascular phenotypes. Whether this is true for aortic stenosis remains unknown. Objective To develop a novel aortic stenosis PRS and compare its aortic stenosis risk estimation to established clinical risk factors. Design, Setting, and Participants This was a longitudinal cohort study using data from the Million Veteran Program (MVP; 2011-2020), UK Biobank (2006-2010), and 6 Thrombolysis in Myocardial Infarction (TIMI) trials, including DECLARE-TIMI 58 (2013-2018), FOURIER (TIMI 59; 2013-2017), PEGASUS-TIMI 54 (2010-2014), SAVOR-TIMI 53 (2010-2013), SOLID-TIMI 52 (2009-2014), and ENGAGE AF-TIMI 48 (2008-2013), which were a mix of population-based and randomized clinical trials. Individuals from UK Biobank and the MVP meeting a previously validated case/control definition for aortic stenosis were included. All individuals from TIMI trials were included unless they had a documented preexisting aortic valve replacement. Analysis took place from January 2022 to December 2023. Exposures PRS for aortic stenosis (developed using data from MVP and validated in UK Biobank) and other previously validated cardiovascular PRSs, defined either as a continuous variable or as low (bottom 20%), intermediate, and high (top 20%), and clinical risk factors. Main Outcomes Aortic stenosis (defined using International Classification of Diseases or Current Procedural Terminology codes in UK Biobank and MVP or safety event data in the TIMI trials). Results The median (IQR) age in MVP was 67 (57-73) years, and 135 140 of 147 104 participants (92%) were male. The median (IQR) age in the TIMI trials was 66 (54-78) years, and 45 524 of 59 866 participants (71%) were male. The best aortic stenosis PRS incorporated 5 170 041 single-nucleotide variants and was associated with aortic stenosis in both the MVP testing sample (odds ratio, 1.41; 95% CI, 1.37-1.45 per 1 SD PRS; P = 4.6 × 10-116) and TIMI trials (hazard ratio, 1.44; 95% CI, 1.27-1.62 per 1 SD PRS; P = 3.2 × 10-9). Among genetic and clinical risk factors, the aortic stenosis PRS performed comparably to most risk factors besides age, and within a given age range, the combination of clinical and genetic risk factors was additive, providing a 3- to 4-fold increased gradient of risk of aortic stenosis. However, the addition of the aortic stenosis PRS to a model including clinical risk factors only improved risk discrimination of aortic stenosis by 0.01 to 0.02 (C index in MVP: 0.78 with clinical risk factors, 0.79 with risk factors and aortic stenosis PRS; C index in TIMI: 0.71 with clinical risk factors, 0.73 with risk factors and aortic stenosis PRS). Conclusions This study developed and validated 1 of the first aortic stenosis PRSs. While aortic stenosis genetic risk was independent from clinical risk factors and performed comparably to all other risk factors besides age, genetic risk resulted in only a small improvement in overall aortic stenosis risk discrimination beyond age and clinical risk factors. This work sets the stage for further development of an aortic stenosis PRS.
Collapse
Affiliation(s)
- Aeron M. Small
- Cardiovascular Medicine Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
- Department of Cardiology, Boston Veterans Affairs Healthcare System, West Roxbury, Massachusetts
| | - Giorgio E. M. Melloni
- TIMI Study Group, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Frederick K. Kamanu
- TIMI Study Group, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Brian A. Bergmark
- Cardiovascular Medicine Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
- TIMI Study Group, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Marc P. Bonaca
- Department of Medicine, Cardiology and Vascular Medicine, University of Colorado School of Medicine, Aurora
| | - Michelle L. O’Donoghue
- Cardiovascular Medicine Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
- TIMI Study Group, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Robert P. Giugliano
- Cardiovascular Medicine Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
- TIMI Study Group, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Benjamin M. Scirica
- Cardiovascular Medicine Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
- TIMI Study Group, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Deepak Bhatt
- Mount Sinai Fuster Heart Hospital, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Elliott M. Antman
- Cardiovascular Medicine Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
- TIMI Study Group, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Itamar Raz
- Department of Endocrinology and Metabolism, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Stephen D. Wiviott
- Cardiovascular Medicine Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
- TIMI Study Group, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Buu Truong
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Peter W. F. Wilson
- Atlanta Veterans Affairs Medical Center, Decatur, Georgia
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Kelly Cho
- Veterans Affairs Healthcare System, Boston, Massachusetts
- Division of Aging, Mass General Brigham and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Christopher J. O’Donnell
- Cardiovascular Medicine Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
- Department of Cardiology, Boston Veterans Affairs Healthcare System, West Roxbury, Massachusetts
| | - Eugene Braunwald
- Cardiovascular Medicine Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
- TIMI Study Group, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Steve A. Lubitz
- Cardiovascular Medicine Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Cardiovascular Research Center, Massachusetts General Hospital, Boston
| | - Patrick Ellinor
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Cardiovascular Research Center, Massachusetts General Hospital, Boston
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston
| | - Gina M. Peloso
- Veterans Affairs Healthcare System, Boston, Massachusetts
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts
| | - Christian T. Ruff
- Cardiovascular Medicine Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
- TIMI Study Group, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Marc S. Sabatine
- Cardiovascular Medicine Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
- TIMI Study Group, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Pradeep Natarajan
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Cardiovascular Research Center, Massachusetts General Hospital, Boston
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston
- Center for Genomic Medicine, Massachusetts General Hospital, Boston
| | - Nicholas A. Marston
- Cardiovascular Medicine Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
- TIMI Study Group, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
23
|
Aikawa E, Blaser MC, Singh SA, Levine RA, Yacoub MH. Challenges and Opportunities in Valvular Heart Disease: From Molecular Mechanisms to the Community. Arterioscler Thromb Vasc Biol 2024; 44:763-767. [PMID: 38536897 PMCID: PMC10977651 DOI: 10.1161/atvbaha.123.319563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2024]
Affiliation(s)
- Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Center for Excellence in Vascular Biology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Mark C. Blaser
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Sasha A. Singh
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Robert A. Levine
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | |
Collapse
|
24
|
Thériault S, Li Z, Abner E, Luan J, Manikpurage HD, Houessou U, Zamani P, Briend M, Boudreau DK, Gaudreault N, Frenette L, Argaud D, Dahmene M, Dagenais F, Clavel MA, Pibarot P, Arsenault BJ, Boekholdt SM, Wareham NJ, Esko T, Mathieu P, Bossé Y. Integrative genomic analyses identify candidate causal genes for calcific aortic valve stenosis involving tissue-specific regulation. Nat Commun 2024; 15:2407. [PMID: 38494474 PMCID: PMC10944835 DOI: 10.1038/s41467-024-46639-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 03/05/2024] [Indexed: 03/19/2024] Open
Abstract
There is currently no medical therapy to prevent calcific aortic valve stenosis (CAVS). Multi-omics approaches could lead to the identification of novel molecular targets. Here, we perform a genome-wide association study (GWAS) meta-analysis including 14,819 cases among 941,863 participants of European ancestry. We report 32 genomic loci, among which 20 are novel. RNA sequencing of 500 human aortic valves highlights an enrichment in expression regulation at these loci and prioritizes candidate causal genes. Homozygous genotype for a risk variant near TWIST1, a gene involved in endothelial-mesenchymal transition, has a profound impact on aortic valve transcriptomics. We identify five genes outside of GWAS loci by combining a transcriptome-wide association study, colocalization, and Mendelian randomization analyses. Using cross-phenotype and phenome-wide approaches, we highlight the role of circulating lipoproteins, blood pressure and inflammation in the disease process. Our findings pave the way for the development of novel therapies for CAVS.
Collapse
Affiliation(s)
- Sébastien Thériault
- Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Quebec City, QC, Canada.
- Department of Molecular Biology, Medical Biochemistry and Pathology, Université Laval, Quebec City, QC, Canada.
| | - Zhonglin Li
- Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Quebec City, QC, Canada
| | - Erik Abner
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Jian'an Luan
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Hasanga D Manikpurage
- Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Quebec City, QC, Canada
| | - Ursula Houessou
- Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Quebec City, QC, Canada
| | - Pardis Zamani
- Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Quebec City, QC, Canada
| | - Mewen Briend
- Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Quebec City, QC, Canada
| | - Dominique K Boudreau
- Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Quebec City, QC, Canada
| | - Nathalie Gaudreault
- Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Quebec City, QC, Canada
| | - Lily Frenette
- Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Quebec City, QC, Canada
| | - Déborah Argaud
- Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Quebec City, QC, Canada
| | - Manel Dahmene
- Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Quebec City, QC, Canada
| | - François Dagenais
- Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Quebec City, QC, Canada
- Department of Surgery, Université Laval, Quebec City, QC, Canada
| | - Marie-Annick Clavel
- Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Quebec City, QC, Canada
- Department of Medicine, Université Laval, Quebec City, QC, Canada
| | - Philippe Pibarot
- Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Quebec City, QC, Canada
- Department of Medicine, Université Laval, Quebec City, QC, Canada
| | - Benoit J Arsenault
- Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Quebec City, QC, Canada
- Department of Medicine, Université Laval, Quebec City, QC, Canada
| | - S Matthijs Boekholdt
- Department of Cardiology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Nicholas J Wareham
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Tõnu Esko
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Patrick Mathieu
- Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Quebec City, QC, Canada
- Department of Surgery, Université Laval, Quebec City, QC, Canada
| | - Yohan Bossé
- Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Quebec City, QC, Canada
- Department of Molecular Medicine, Université Laval, Quebec City, QC, Canada
| |
Collapse
|
25
|
Abstract
PURPOSE OF REVIEW Lp(a) is one of the most atherogenic lipoproteins, and significant progress has been made to understand its pathophysiology over the last 20 years. There are now selective therapies in late-stage clinical trials to lower Lp(a). Yet there are many outstanding questions about Lp(a). This review outlines 10 of the most burning questions and tries to answer some of them. RECENT FINDINGS Antisense oligonucleotide (ASO) treatment is currently the most advanced therapy to lower plasma Lp(a) by 60-80%. There are, however, also two small molecule medications in early stage of development with similar efficacy. SUMMARY This review aims to answer important preclinical and clinical questions about the metabolism and physiological role of Lp(a) and also outlines possible therapeutic approaches with nutraceuticals, currently available lipid-lowering therapies and new medications. In addition, ways are illustrated to use Lp(a) as a marker to better predict cardiovascular risk.
Collapse
Affiliation(s)
- Karam M Kostner
- Department of Cardiology Mater Hospital and University of Queensland, Brisbane, Australia
| | - Gerhard M Kostner
- Institute of Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| |
Collapse
|
26
|
Martin SS, Aday AW, Almarzooq ZI, Anderson CAM, Arora P, Avery CL, Baker-Smith CM, Barone Gibbs B, Beaton AZ, Boehme AK, Commodore-Mensah Y, Currie ME, Elkind MSV, Evenson KR, Generoso G, Heard DG, Hiremath S, Johansen MC, Kalani R, Kazi DS, Ko D, Liu J, Magnani JW, Michos ED, Mussolino ME, Navaneethan SD, Parikh NI, Perman SM, Poudel R, Rezk-Hanna M, Roth GA, Shah NS, St-Onge MP, Thacker EL, Tsao CW, Urbut SM, Van Spall HGC, Voeks JH, Wang NY, Wong ND, Wong SS, Yaffe K, Palaniappan LP. 2024 Heart Disease and Stroke Statistics: A Report of US and Global Data From the American Heart Association. Circulation 2024; 149:e347-e913. [PMID: 38264914 DOI: 10.1161/cir.0000000000001209] [Citation(s) in RCA: 804] [Impact Index Per Article: 804.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2024]
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 2024 AHA Statistical Update is the product of a full year's worth of effort in 2023 by dedicated volunteer clinicians and scientists, committed government professionals, and AHA staff members. The AHA strives to further understand and help heal health problems inflicted by structural racism, a public health crisis that can significantly damage physical and mental health and perpetuate disparities in access to health care, education, income, housing, and several other factors vital to healthy lives. This year's edition includes additional global data, as well as data on the monitoring and benefits of cardiovascular health in the population, with an enhanced focus on health equity across several key domains. 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
|
Lindman BR. Innovative Methods to Tackle Longstanding Goals in Aortic Stenosis. J Am Coll Cardiol 2024; 83:592-594. [PMID: 38296403 DOI: 10.1016/j.jacc.2023.11.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 11/28/2023] [Indexed: 02/08/2024]
Affiliation(s)
- Brian R Lindman
- Structural Heart and Valve Center, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA.
| |
Collapse
|
28
|
Notenboom ML, Van Hoof L, Schuermans A, Takkenberg JJM, Rega FR, Taverne YJHJ. Aortic Valve Embryology, Mechanobiology, and Second Messenger Pathways: Implications for Clinical Practice. J Cardiovasc Dev Dis 2024; 11:49. [PMID: 38392263 PMCID: PMC10888685 DOI: 10.3390/jcdd11020049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 01/22/2024] [Accepted: 01/29/2024] [Indexed: 02/24/2024] Open
Abstract
During the Renaissance, Leonardo Da Vinci was the first person to successfully detail the anatomy of the aortic root and its adjacent structures. Ever since, novel insights into morphology, function, and their interplay have accumulated, resulting in advanced knowledge on the complex functional characteristics of the aortic valve (AV) and root. This has shifted our vision from the AV as being a static structure towards that of a dynamic interconnected apparatus within the aortic root as a functional unit, exhibiting a complex interplay with adjacent structures via both humoral and mechanical stimuli. This paradigm shift has stimulated surgical treatment strategies of valvular disease that seek to recapitulate healthy AV function, whereby AV disease can no longer be seen as an isolated morphological pathology which needs to be replaced. As prostheses still cannot reproduce the complexity of human nature, treatment of diseased AVs, whether stenotic or insufficient, has tremendously evolved, with a similar shift towards treatments options that are more hemodynamically centered, such as the Ross procedure and valve-conserving surgery. Native AV and root components allow for an efficient Venturi effect over the valve to allow for optimal opening during the cardiac cycle, while also alleviating the left ventricle. Next to that, several receptors are present on native AV leaflets, enabling messenger pathways based on their interaction with blood and other shear-stress-related stimuli. Many of these physiological and hemodynamical processes are under-acknowledged but may hold important clues for innovative treatment strategies, or as potential novel targets for therapeutic agents that halt or reverse the process of valve degeneration. A structured overview of these pathways and their implications for cardiothoracic surgeons and cardiologists is lacking. As such, we provide an overview on embryology, hemodynamics, and messenger pathways of the healthy and diseased AV and its implications for clinical practice, by relating this knowledge to current treatment alternatives and clinical decision making.
Collapse
Affiliation(s)
- Maximiliaan L. Notenboom
- Department of Cardiothoracic Surgery, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands; (M.L.N.)
| | - Lucas Van Hoof
- Department of Cardiac Surgery, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Art Schuermans
- Department of Cardiac Surgery, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Johanna J. M. Takkenberg
- Department of Cardiothoracic Surgery, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands; (M.L.N.)
| | - Filip R. Rega
- Department of Cardiac Surgery, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Yannick J. H. J. Taverne
- Department of Cardiothoracic Surgery, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands; (M.L.N.)
| |
Collapse
|
29
|
Lan NSR, Khan Z, Watts GF. Lipoprotein(a) and calcific aortic valve disease: current evidence and future directions. Curr Opin Clin Nutr Metab Care 2024; 27:77-86. [PMID: 37650693 DOI: 10.1097/mco.0000000000000976] [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] [Indexed: 09/01/2023]
Abstract
PURPOSE OF REVIEW Calcific aortic valve disease (CAVD), the most common cause of aortic stenosis (AS), is characterized by slowly progressive fibrocalcific remodelling of the valve cusps. Once symptomatic, severe AS is associated with poor survival unless surgical or transcatheter valve replacement is performed. Unfortunately, no pharmacological interventions have been demonstrated to alter the natural history of CAVD. Lipoprotein(a) [Lp(a)], a low-density lipoprotein-like particle, has been implicated in the pathophysiology of CAVD. RECENT FINDINGS The mechanisms by which Lp(a) results in CAVD are not well understood. However, the oxidized phospholipids carried by Lp(a) are considered a crucial mediator of the disease process. An increasing number of studies demonstrate a causal association between plasma Lp(a) levels and frequency of AS and need for aortic valve replacement, which is independent of inflammation, as measured by plasma C-reactive protein levels. However, not all studies show an association between Lp(a) and increased progression of calcification in individuals with established CAVD. SUMMARY Epidemiologic, genetic, and Mendelian randomization studies have collectively suggested that Lp(a) is a causal risk factor for CAVD. Whether Lp(a)-lowering can prevent initiation or slow progression of CAVD remains to be demonstrated.
Collapse
Affiliation(s)
- Nick S R Lan
- Departments of Cardiology and Internal Medicine, Royal Perth Hospital
- School of Medicine, The University of Western Australia, Perth, Western Australia, Australia
| | - Zahid Khan
- Department of Cardiology, Barts Heart Centre, London
- University of South Wales, Cardiff, UK
| | - Gerald F Watts
- Departments of Cardiology and Internal Medicine, Royal Perth Hospital
- School of Medicine, The University of Western Australia, Perth, Western Australia, Australia
| |
Collapse
|
30
|
Thomas PE, Vedel-Krogh S, Nordestgaard BG. Measuring lipoprotein(a) for cardiovascular disease prevention - in whom and when? Curr Opin Cardiol 2024; 39:39-48. [PMID: 38078600 DOI: 10.1097/hco.0000000000001104] [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] [Indexed: 12/18/2023]
Abstract
PURPOSE OF REVIEW The aim of this study is to summarize major cardiovascular guideline recommendations on lipoprotein(a) and highlighting recent findings that emphasize how measuring lipoprotein(a) once in all adults is meaningful regardless of age, sex, comorbidities, or ethnicity. RECENT FINDINGS Many international guidelines now recommend once in a lifetime measurement of lipoprotein(a) in all adult individuals to facilitate accurate risk prediction. Lipoprotein(a)-lowering therapy to reduce cardiovascular disease is on the horizon, with results from the first phase 3 trial expected in 2025. SUMMARY Elevated lipoprotein(a) is an independent causal risk factor for atherosclerotic cardiovascular disease and aortic valve stenosis and measuring lipoprotein(a) once in all individuals regardless of age, sex, comorbidities, or ethnicity is meaningful to aid in risk stratification.
Collapse
Affiliation(s)
- Peter E Thomas
- Department of Clinical Biochemistry
- The Copenhagen General Population Study, Copenhagen University Hospital - Herlev and Gentofte
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Signe Vedel-Krogh
- Department of Clinical Biochemistry
- The Copenhagen General Population Study, Copenhagen University Hospital - Herlev and Gentofte
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Børge G Nordestgaard
- Department of Clinical Biochemistry
- The Copenhagen General Population Study, Copenhagen University Hospital - Herlev and Gentofte
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
31
|
Ostrominski JW, Powell-Wiley TM. Risk Stratification and Treatment of Obesity for Primary and Secondary Prevention of Cardiovascular Disease. Curr Atheroscler Rep 2024; 26:11-23. [PMID: 38159162 DOI: 10.1007/s11883-023-01182-3] [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: 12/04/2023] [Indexed: 01/03/2024]
Abstract
PURPOSE OF REVIEW In this review, we discuss contemporary and emerging approaches for risk stratification and management of excess adiposity for the primary and secondary prevention of cardiovascular disease. RECENT FINDINGS Obesity is simultaneously a pandemic-scale disease and major risk factor for the incidence and progression of a wide range of cardiometabolic conditions, but risk stratification and treatment remain clinically challenging. However, sex-, race-, and ethnicity-sensitive anthropometric measures, body composition-focused imaging, and health burden-centric staging systems have emerged as important facilitators of holistic risk prediction. Further, expanding therapeutic approaches, including comprehensive lifestyle programs, anti-obesity pharmacotherapies, device/endoscopy-based interventions, metabolic surgery, and novel healthcare delivery resources offer new empowerment for cardiovascular risk reduction in individuals with obesity. Personalized risk stratification and weight management are central to reducing the lifetime prevalence and impact of cardiovascular disease. Further evidence informing long-term safety, efficacy, and cost-effectiveness of novel approaches targeting obesity are critically needed.
Collapse
Affiliation(s)
- John W Ostrominski
- Division of Cardiovascular Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Tiffany M Powell-Wiley
- Social Determinants of Obesity and Cardiovascular Risk Laboratory, Cardiovascular Branch, Division of Intramural Research, National Heart, Lung and Blood Institute, National Institutes of Health, Building 10, Room 5-5332, 10 Center Dr., Bethesda, MD, 20892, USA.
- Intramural Research Program, National Institute on Minority Health and Health Disparities, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
32
|
Schuermans A, Pournamdari AB, Lee J, Bhukar R, Ganesh S, Darosa N, Small AM, Yu Z, Hornsby W, Koyama S, Januzzi JL, Honigberg MC, Natarajan P. Integrative proteomic analyses across common cardiac diseases yield new mechanistic insights and enhanced prediction. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.12.19.23300218. [PMID: 38196601 PMCID: PMC10775327 DOI: 10.1101/2023.12.19.23300218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2024]
Abstract
Cardiac diseases represent common highly morbid conditions for which underlying molecular mechanisms remain incompletely understood. Here, we leveraged 1,459 protein measurements in 44,313 UK Biobank participants to characterize the circulating proteome associated with incident coronary artery disease, heart failure, atrial fibrillation, and aortic stenosis. Multivariable-adjusted Cox regression identified 820 protein-disease associations-including 441 proteins-at Bonferroni-adjusted P <8.6×10 -6 . Cis -Mendelian randomization suggested causal roles that aligned with epidemiological findings for 6% of proteins identified in primary analyses, prioritizing novel therapeutic targets for different cardiac diseases (e.g., interleukin-4 receptor for heart failure and spondin-1 for atrial fibrillation). Interaction analyses identified seven protein-disease associations that differed Bonferroni-significantly by sex. Models incorporating proteomic data (vs. clinical risk factors alone) improved prediction for coronary artery disease, heart failure, and atrial fibrillation. These results lay a foundation for future investigations to uncover novel disease mechanisms and assess the clinical utility of protein-based prevention strategies for cardiac diseases.
Collapse
|
33
|
Massera D, Bartz TM, Biggs ML, Sotoodehnia N, Reiner AP, Semba RD, Gottdiener JS, Psaty BM, Owens DS, Kizer JR. Traditional and novel risk factors for incident aortic stenosis in community-dwelling older adults. Heart 2023; 110:57-64. [PMID: 37463733 PMCID: PMC10794538 DOI: 10.1136/heartjnl-2023-322709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 06/28/2023] [Indexed: 07/20/2023] Open
Abstract
OBJECTIVES Calcific aortic stenosis (AS) is the most common valvular disease in older adults, yet its risk factors remain insufficiently studied in this population. Such studies are necessary to enhance understanding of mechanisms, disease management and therapeutics. METHODS The Cardiovascular Health Study is a population-based investigation of older adults that completed adjudication of incident AS over long-term follow-up. We evaluated traditional cardiovascular risk factors or disease, as well as novel risk factors from lipid, inflammatory and mineral metabolism pathways, in relation to incident moderate or severe AS (including AS procedures) and clinically significant AS (severe AS, including procedures). RESULTS Of 5390 participants (age 72.9±5.6 years, 57.6% female, 12.5% black), 287 developed moderate or severe AS, and 175 clinically significant AS, during median follow-up of 13.1 years. After full adjustment, age (HR=1.66 per SD (95% CI=1.45, 1.91)), male sex (HR=1.41 (1.06, 1.87)), diabetes (HR=1.53 (1.10, 2.13)), coronary heart disease (CHD, HR=1.36 (1.01, 1.84)), lipoprotein-associated phospholipase-A2 (LpPLA2) activity (HR=1.21 per SD (1.07, 1.37)) and sCD14 (HR=1.16 per SD (1.01, 1.34)) were associated with incident moderate/severe AS, while black race demonstrated an inverse association (HR=0.40 (0.24, 0.65)), and creatinine-based estimated glomerular filtration rate (eGFRcr) showed a U-shaped relationship. Findings were similar for clinically significant AS, although CHD and sCD14 fell short of significance, but interleukin-(IL) 6 showed a positive association. CONCLUSION This comprehensive evaluation of risk factors for long-term incidence of AS identified associations for diabetes and prevalent CHD, LpPLA2 activity, sCD14 and IL-6, and eGFRcr. These factors may hold clues to biology, preventive efforts and potential therapeutics for those at highest risk.
Collapse
Affiliation(s)
- Daniele Massera
- Leon H. Charney Division of Cardiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Traci M Bartz
- Department of Biostatistics, University of Washington, Seattle, Washington, USA
| | - Mary L Biggs
- Department of Biostatistics, University of Washington, Seattle, Washington, USA
| | - Nona Sotoodehnia
- Division of Cardiology, University of Washington, Seattle, Washington, USA
- Cardiovascular Health Research Unit, Departments of Medicine, Epidemiology and Health Services, University of Washington, Seattle, Washington, USA
| | - Alexander P Reiner
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
| | - Richard D Semba
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - John S Gottdiener
- Division of Cardiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Departments of Medicine, Epidemiology and Health Services, University of Washington, Seattle, Washington, USA
| | - David S Owens
- Division of Cardiology, University of Washington, Seattle, Washington, USA
| | - Jorge R Kizer
- Cardiology Section, San Francisco VA Health Care System, San Francisco, California, USA
- Departments of Medicine, Epidemiology and Biostatistics, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
34
|
Larsson SC, Butterworth AS, Burgess S. Mendelian randomization for cardiovascular diseases: principles and applications. Eur Heart J 2023; 44:4913-4924. [PMID: 37935836 PMCID: PMC10719501 DOI: 10.1093/eurheartj/ehad736] [Citation(s) in RCA: 276] [Impact Index Per Article: 138.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 09/13/2023] [Accepted: 10/17/2023] [Indexed: 11/09/2023] Open
Abstract
Large-scale genome-wide association studies conducted over the last decade have uncovered numerous genetic variants associated with cardiometabolic traits and risk factors. These discoveries have enabled the Mendelian randomization (MR) design, which uses genetic variation as a natural experiment to improve causal inferences from observational data. By analogy with the random assignment of treatment in randomized controlled trials, the random segregation of genetic alleles when DNA is transmitted from parents to offspring at gamete formation is expected to reduce confounding in genetic associations. Mendelian randomization analyses make a set of assumptions that must hold for valid results. Provided that the assumptions are well justified for the genetic variants that are employed as instrumental variables, MR studies can inform on whether a putative risk factor likely has a causal effect on the disease or not. Mendelian randomization has been increasingly applied over recent years to predict the efficacy and safety of existing and novel drugs targeting cardiovascular risk factors and to explore the repurposing potential of available drugs. This review article describes the principles of the MR design and some applications in cardiovascular epidemiology.
Collapse
Affiliation(s)
- Susanna C Larsson
- Unit of Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Adam S Butterworth
- 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, Papworth Road, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, School of Clinical Medicine, Addenbrooke’s Hospital, University of Cambridge, Cambridge, UK
- Health Data Research UK, Wellcome Genome Campus and University of Cambridge, Hinxton, UK
- NIHR Blood and Transplant Research Unit in Donor Health and Behaviour, University of Cambridge, Cambridge, UK
| | - Stephen Burgess
- 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, Papworth Road, Cambridge, UK
- MRC Biostatistics Unit, University of Cambridge, Cambridge, UK
| |
Collapse
|
35
|
Chai C, Li S, Chen L, Song X. Causal association of calcific aortic valve stenosis and atrial fibrillation: a Mendelian randomization study. Sci Rep 2023; 13:20284. [PMID: 37985719 PMCID: PMC10662195 DOI: 10.1038/s41598-023-47770-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: 04/03/2023] [Accepted: 11/18/2023] [Indexed: 11/22/2023] Open
Abstract
Calcific aortic valve stenosis (CAVS) is associated with an increased risk of atrial fibrillation (AF) in observational studies, but whether these associations are causal has not been determined. This study aimed to explore the potential causal relationship between CAVS and AF via Mendelian randomization (MR). Genetic variants from the genome-wide association study (GWAS) summary data of the European population for CAVS were used to investigate the association with AF. The inverse variance weighted (IVW) approach was used to obtain the primary causal inference, and several sensitivity analysis approaches, such as the MR‒Egger and weighted median (WM), were performed to assess the robustness of the results. A total of nineteen valid and independent genetic SNPs associated with CAVS were obtained from the GWAS database. Genetically predicted CAVS (OR: 1.105; 95% CI: 1.072-1.139; p = 8.60E-11) was associated with an increased risk of AF. Similar results were discovered in the sensitivity analyses by using MR Egger and weighted median approaches. An MR design was used to reduce confounding variables and the potential for reverse causality bias. The results provide genetic evidence that CAVS considerably increased the risk of AF.
Collapse
Affiliation(s)
- Chen Chai
- Emergency Center, Hubei Clinical Research Center for Emergency and Resuscitation, Zhongnan Hospital of Wuhan University, No. 169, Donghu Road, Wuchang District, Wuhan, 430071, China
| | - Shoupeng Li
- Emergency Department, Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan, China
| | - Lin Chen
- Emergency Department, Xiantao First People's Hospital Affiliated to Changjiang University, Xiantao, China
| | - Xiaobing Song
- Emergency Center, Hubei Clinical Research Center for Emergency and Resuscitation, Zhongnan Hospital of Wuhan University, No. 169, Donghu Road, Wuchang District, Wuhan, 430071, China.
| |
Collapse
|
36
|
Lee YH, Park S. Genetic and Lifestyle-Related Factors Influencing Serum Hyper-Propionylcarnitine Concentrations and Their Association with Metabolic Syndrome and Cardiovascular Disease Risk. Int J Mol Sci 2023; 24:15810. [PMID: 37958793 PMCID: PMC10647558 DOI: 10.3390/ijms242115810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 10/27/2023] [Accepted: 10/27/2023] [Indexed: 11/15/2023] Open
Abstract
The genetic and environmental determinants of serum propionylcarnitine concentrations (PC) remain largely unexplored. This study investigated the impact of genetic and environmental factors on serum propionylcarnitine levels in middle-aged and elderly participants from the Ansan/Ansung cohort of the Korean Genome and Epidemiology Study. Our goal was to understand the role of PC on the risk of metabolic syndrome (MetS) leading to cardiovascular disease, particularly concerning branched-chain amino acid (BCAA) metabolism. We analyzed participants' demographic, lifestyle, and biochemical data with and without MetS. Serum metabolite concentrations, including carnitine, acylcarnitine, and amino acid concentrations, were measured, and the components of MetS were evaluated. Genetic variants associated with low and high PC were selected using genome-wide association studies after adjusting for MetS-related parameters. Further, genetic variants and lifestyle factors that interacted with the polygenic risk score (PRS) were analyzed. Participants with MetS were older and less educated, and their alcohol intake was higher than non-MetS participants. PC was significantly associated with the MetS risk and increased the serum levels of BCAAs and other amino acids. Higher PC positively correlated with MetS components, insulin resistance, and cardiovascular risk factors. Intake of calcium, sodium, and vitamin D were inversely associated with PC, but coffee consumption was positively linked to PC. Multiple C2 And Transmembrane Domain Containing-1 (MCTP1)_rs4290997, Kinesin Family Member-7 (KIF7)_rs2350480, Coagulation Factor-II (F2)_rs2070850, Peroxisomal Biogenesis Factor-3 (PEX3)_rs223231, TBC1 Domain Family Member-22A (TBC1D22A)_rs910543, and Phospholipase A2 Group-IV-C (PLA2G4C)_rs7252136 interact with each other to have a threefold influence on PC. The PRS for the six-genetic variant model also interacted with age; the diet rich in beans, potato, and kimchi; and smoking status, influencing PC. In conclusion, elevated PC was associated with MetS and cardiovascular disease risk, suggesting their potential as disease biomarkers.
Collapse
Affiliation(s)
- Yong-Hwa Lee
- Department of Cosmetic Biotechnology, Hoseo University, Asan 31499, Republic of Korea;
| | - Sunmin Park
- Department of Food and Nutrition, Institute of Basic Science, Obesity/Diabetes Research Center, Hoseo University, Asan 31499, Republic of Korea
| |
Collapse
|
37
|
Di Costanzo A, Indolfi C, Franzone A, Esposito G, Spaccarotella CAM. Lp(a) in the Pathogenesis of Aortic Stenosis and Approach to Therapy with Antisense Oligonucleotides or Short Interfering RNA. Int J Mol Sci 2023; 24:14939. [PMID: 37834387 PMCID: PMC10573862 DOI: 10.3390/ijms241914939] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 09/28/2023] [Accepted: 10/02/2023] [Indexed: 10/15/2023] Open
Abstract
To date, no medical therapy can slow the progression of aortic stenosis. Fibrocalcific stenosis is the most frequent form in the general population and affects about 6% of the elderly population. Over the years, diagnosis has evolved thanks to echocardiography and computed tomography assessments. The application of artificial intelligence to electrocardiography could further implement early diagnosis. Patients with severe aortic stenosis, especially symptomatic patients, have valve repair as their only therapeutic option by surgical or percutaneous technique (TAVI). The discovery that the pathogenetic mechanism of aortic stenosis is similar to the atherosclerosis process has made it possible to evaluate the hypothesis of medical therapy for aortic stenosis. Several drugs have been tested to reduce low-density lipoprotein (LDL) and lipoprotein(a) (Lp(a)) levels, inflammation, and calcification. The Proprotein Convertase Subtilisin/Kexin type 9 inhibitors (PCSK9-i) could decrease the progression of aortic stenosis and the requirement for valve implantation. Great interest is related to circulating Lp(a) levels as causally linked to degenerative aortic stenosis. New therapies with ASO (antisense oligonucleotides) and siRNA (small interfering RNA) are currently being tested. Olpasiran and pelacarsen reduce circulating Lp(a) levels by 85-90%. Phase 3 studies are underway to evaluate the effect of these drugs on cardiovascular events (cardiovascular death, non-fatal myocardial injury, and non-fatal stroke) in patients with elevated Lp(a) and CVD (cardiovascular diseases). For instance, if a reduction in Lp(a) levels is associated with aortic stenosis prevention or progression, further prospective clinical trials are warranted to confirm this observation in this high-risk population.
Collapse
Affiliation(s)
- Assunta Di Costanzo
- Division of Cardiology, Cardiovascular Research Center, University Magna Graecia Catanzaro, 88100 Catanzaro, Italy;
| | - Ciro Indolfi
- Division of Cardiology, Cardiovascular Research Center, University Magna Graecia Catanzaro, 88100 Catanzaro, Italy;
| | - Anna Franzone
- Division of Cardiology, Department of Advanced Biomedical Sciences, University of Naples Federico II, 80131 Naples, Italy; (A.F.); (G.E.); (C.A.M.S.)
| | - Giovanni Esposito
- Division of Cardiology, Department of Advanced Biomedical Sciences, University of Naples Federico II, 80131 Naples, Italy; (A.F.); (G.E.); (C.A.M.S.)
| | - Carmen Anna Maria Spaccarotella
- Division of Cardiology, Department of Advanced Biomedical Sciences, University of Naples Federico II, 80131 Naples, Italy; (A.F.); (G.E.); (C.A.M.S.)
| |
Collapse
|
38
|
Koschinsky ML, Stroes ESG, Kronenberg F. Daring to dream: Targeting lipoprotein(a) as a causal and risk-enhancing factor. Pharmacol Res 2023; 194:106843. [PMID: 37406784 DOI: 10.1016/j.phrs.2023.106843] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 06/15/2023] [Accepted: 06/27/2023] [Indexed: 07/07/2023]
Abstract
Lipoprotein(a) [Lp(a)], a distinct lipoprotein class, has become a major focus for cardiovascular research. This review is written in light of the recent guideline and consensus statements on Lp(a) and focuses on 1) the causal association between Lp(a) and cardiovascular outcomes, 2) the potential mechanisms by which elevated Lp(a) contributes to cardiovascular diseases, 3) the metabolic insights on the production and clearance of Lp(a) and 4) the current and future therapeutic approaches to lower Lp(a) concentrations. The concentrations of Lp(a) are under strict genetic control. There exists a continuous relationship between the Lp(a) concentrations and risk for various endpoints of atherosclerotic cardiovascular disease (ASCVD). One in five people in the Caucasian population is considered to have increased Lp(a) concentrations; the prevalence of elevated Lp(a) is even higher in black populations. This makes Lp(a) a cardiovascular risk factor of major public health relevance. Besides the association between Lp(a) and myocardial infarction, the relationship with aortic valve stenosis has become a major focus of research during the last decade. Genetic studies provided strong support for a causal association between Lp(a) and cardiovascular outcomes: carriers of genetic variants associated with lifelong increased Lp(a) concentration are significantly more frequent in patients with ASCVD. This has triggered the development of drugs that can specifically lower Lp(a) concentrations: mRNA-targeting therapies such as anti-sense oligonucleotide (ASO) therapies and short interfering RNA (siRNA) therapies have opened new avenues to lower Lp(a) concentrations more than 95%. Ongoing Phase II and III clinical trials of these compounds are discussed in this review.
Collapse
Affiliation(s)
- Marlys L Koschinsky
- Robarts Research Institute, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada; Department of Physiology & Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Erik S G Stroes
- Department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Florian Kronenberg
- Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
39
|
Xu ZY, Jing X, Xiong XD. Emerging Role and Mechanism of the FTO Gene in Cardiovascular Diseases. Biomolecules 2023; 13:biom13050850. [PMID: 37238719 DOI: 10.3390/biom13050850] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 05/10/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
The fat mass and obesity-associated (FTO) gene was the first obesity-susceptibility gene identified through a genome-wide association study (GWAS). A growing number of studies have suggested that genetic variants of FTO are strongly associated with the risk of cardiovascular diseases, including hypertension and acute coronary syndrome. In addition, FTO was also the first N6-methyladenosine (m6A) demethylase, suggesting the reversible nature of m6A modification. m6A is dynamically deposited, removed, and recognized by m6A methylases, demethylases, and m6A binding proteins, respectively. By catalyzing m6A demethylation on mRNA, FTO may participate in various biological processes by modulating RNA function. Recent studies demonstrated that FTO plays a pivotal role in the initiation and progression of cardiovascular diseases such as myocardial fibrosis, heart failure, and atherosclerosis and may hold promise as a potential therapeutic target for treating or preventing a variety of cardiovascular diseases. Here, we review the association between FTO genetic variants and cardiovascular disease risk, summarize the role of FTO as an m6A demethylase in cardiovascular disorders, and discuss future research directions and possible clinical implications.
Collapse
Affiliation(s)
- Zi-Yang Xu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523808, China
| | - Xia Jing
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523808, China
| | - Xing-Dong Xiong
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523808, China
| |
Collapse
|
40
|
Bäck M. Icosapent ethyl in cardiovascular prevention: Resolution of inflammation through the eicosapentaenoic acid - resolvin E1 - ChemR23 axis. Pharmacol Ther 2023:108439. [PMID: 37201735 DOI: 10.1016/j.pharmthera.2023.108439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 05/03/2023] [Accepted: 05/15/2023] [Indexed: 05/20/2023]
Abstract
Cardiovascular outcome trials on omega-3 fatty acids have generated contradictory results but indicate a dose-dependent beneficial effect of eicosapentaenoic acid (EPA). Beneficial cardiovascular effects of EPA may in addition to triglyceride lowering be mediated through alternative mechanisms of action. In this review, the link between EPA and a resolution of atherosclerotic inflammation is addressed. EPA is a substrate for the enzymatic metabolism into the lipid mediator resolvin E1 (RvE1), which activates the receptor ChemR23 to transduce an active resolution of inflammation. This has been shown to dampen the immune response and provide atheroprotective responses in different models. The intermediate EPA metabolite 18-HEPE emerges as a biomarker of EPA metabolism towards proresolving mediators in observational studies. Genetic variations within the EPA-RvE1-ChemR23 axis affecting the response to EPA may open up for precision medicine to identify responders and non-responders to EPA and fish oil supplementation. In conclusion, activation of the EPA-RvE1-ChemR23 axis towards a resolution of inflammation may contribute to beneficial effects in cardiovascular prevention.
Collapse
Affiliation(s)
- Magnus Bäck
- Department of Cardiology, Heart and Vascular Center, Karolinska University Hospital, Stockholm, Sweden; Translational Cardiology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden; Université de Lorraine, Inserm, DCAC, Nancy, France; CHRU Nancy, Vandœuvre-lès-Nancy, France.
| |
Collapse
|
41
|
Kany S, Rämö JT, Hou C, Jurgens SJ, Nauffal V, Cunningham J, Lau ES, Butte AJ, Ho JE, Olgin JE, Elmariah S, Lindsay ME, Ellinor PT, Pirruccello JP. Assessment of valvular function in over 47,000 people using deep learning-based flow measurements. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.04.29.23289299. [PMID: 37205587 PMCID: PMC10187336 DOI: 10.1101/2023.04.29.23289299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Valvular heart disease is associated with a high global burden of disease. Even mild aortic stenosis confers increased morbidity and mortality, prompting interest in understanding normal variation in valvular function at scale. We developed a deep learning model to study velocity-encoded magnetic resonance imaging in 47,223 UK Biobank participants. We calculated eight traits, including peak velocity, mean gradient, aortic valve area, forward stroke volume, mitral and aortic regurgitant volume, greatest average velocity, and ascending aortic diameter. We then computed sex-stratified reference ranges for these phenotypes in up to 31,909 healthy individuals. In healthy individuals, we found an annual decrement of 0.03cm 2 in the aortic valve area. Participants with mitral valve prolapse had a 1 standard deviation [SD] higher mitral regurgitant volume (P=9.6 × 10 -12 ), and those with aortic stenosis had a 4.5 SD-higher mean gradient (P=1.5 × 10 -431 ), validating the derived phenotypes' associations with clinical disease. Greater levels of ApoB, triglycerides, and Lp(a) assayed nearly 10 years prior to imaging were associated with higher gradients across the aortic valve. Metabolomic profiles revealed that increased glycoprotein acetyls were also associated with an increased aortic valve mean gradient (0.92 SD, P=2.1 x 10 -22 ). Finally, velocity-derived phenotypes were risk markers for aortic and mitral valve surgery even at thresholds below what is considered relevant disease currently. Using machine learning to quantify the rich phenotypic data of the UK Biobank, we report the largest assessment of valvular function and cardiovascular disease in the general population.
Collapse
|