1
|
Anchouche K, Baass A, Thanassoulis G. Lp(a): A Clinical Review. Clin Biochem 2025; 137:110929. [PMID: 40258460 DOI: 10.1016/j.clinbiochem.2025.110929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 04/10/2025] [Accepted: 04/14/2025] [Indexed: 04/23/2025]
Abstract
Elevated lipoprotein(a) (Lp[a]) is a genetically determined cardiovascular risk factor, linked to both atherosclerotic cardiovascular disease and aortic stenosis. Elevated Lp(a) is widely prevalent, and consequently, several cardiovascular societies now recommend performing Lp(a) screening at least once in all adults. While there are presently no approved drugs specifically aimed at lowering Lp(a), several promising candidates are currently in the drug development pipeline, and many of these are now undergoing late phase clinical trials. In this comprehensive review, we outline Lp(a) biology and genetics, describe Lp(a)'s relationship to various cardiovascular clinical phenotypes of interest, highlight novel Lp(a)-lowering therapies, and outline what role these may have in future clinical practice.
Collapse
Affiliation(s)
- Khalil Anchouche
- McGill University Health Centre and Research Institute, Montreal, QC, Canada; McGill University, Montreal, QC, Canada
| | - Alexis Baass
- Genetic Dyslipidemias Clinic of the Montreal Clinical Research Institute, Montreal, QC, Canada; Department of Medicine, Divisions of Experimental Medicine and Medical Biochemistry, McGill University, Montreal, QC, Canada
| | - George Thanassoulis
- McGill University Health Centre and Research Institute, Montreal, QC, Canada; McGill University, Montreal, QC, Canada.
| |
Collapse
|
2
|
Le NQ, He W, Law MH, Medland SE, Mackey DA, Hewitt AW, Gharahkhani P, MacGregor S. Evaluating Practical Approaches for Including MYOC Variants Alongside Common Variants for Genetics-Based Risk Stratification for Glaucoma. Am J Ophthalmol 2025; 274:232-240. [PMID: 40064388 DOI: 10.1016/j.ajo.2025.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 03/03/2025] [Accepted: 03/04/2025] [Indexed: 04/06/2025]
Abstract
OBJECTIVE Rare variants in the MYOC gene are associated with glaucoma risk, with p.Gln368Ter the most common pathogenic variant in Europeans. Genetics-based risk stratification may aid with early diagnosis for glaucoma but it is unclear how best to combine the p.Gln368Ter status with polygenic risk scores (PRS). Our study aimed to examine approaches for identifying p. Gln368Ter carriers using genotyping array data and the utility of integrating p.Gln368Ter status into glaucoma PRS. DESIGN Retrospective cohort study. METHODS We identified p.Gln368Ter carriers using directly genotyped and imputed data. Results were confirmed in a subset with sequencing data. We evaluated the combined effects of p.Gln368Ter status and PRS in stratified analyses by considering them as two separate factors and as an aggregate score. PARTICIPANTS A total of 58,452 participants from the Genetics of Glaucoma, the QSkin Sun and Health Study (QSKIN), and CARTaGENE projects, including 6015 with sequencing data. MAIN OUTCOMES AND MEASURES The concordance of direct genotyping, compared with imputation and sequencing for p.Gln368Ter identification. RESULTS Without appropriate quality control, substantial mis-calling may occur. Nevertheless, the p.Gln368Ter variant could be accurately genotyped in most cases by filtering individuals for call rate and heterozygosity. In 6015 individuals with sequencing data, direct genotyping exhibited perfect concordance with sequencing results. Filtered direct genotyping results showed high agreement with imputed results, with only 16 discrepancies among 57,468 individuals. When quality control is not possible (eg, heterozygosity filtering for an individual), we recommend comparing genotyped and imputed results to ensure accuracy. Incorporating p.Gln368Ter into PRS had additional effects on stratifying high-risk individuals, but did not improve risk prediction for the general population given the variant's rarity. The MYOC-enhanced PRS increased the proportion of p.Gln368Ter carriers classified as high risk from 32.31% to 75.38% in QSKIN and from 38.24% to 79.41% in CARTaGENE. CONCLUSIONS The p.Gln368Ter variant can be genotyped with high accuracy using array data, provided careful quality control measures are implemented. Incorporating p.Gln368Ter into glaucoma PRS improved risk stratification for carriers.
Collapse
Affiliation(s)
- Ngoc-Quynh Le
- From the Statistical Genetics Lab (N.Q.L., W.H., M.H.L., P.G., S.M.), QIMR Berghofer Medical Research Institute, Herston, Brisbane, Queensland, Australia; Faculty of Medicine (N.Q.L., W.H., M.H.L.), University of Queensland, Herston, Queensland, Australia.
| | - Weixiong He
- From the Statistical Genetics Lab (N.Q.L., W.H., M.H.L., P.G., S.M.), QIMR Berghofer Medical Research Institute, Herston, Brisbane, Queensland, Australia; Faculty of Medicine (N.Q.L., W.H., M.H.L.), University of Queensland, Herston, Queensland, Australia
| | - Matthew H Law
- From the Statistical Genetics Lab (N.Q.L., W.H., M.H.L., P.G., S.M.), QIMR Berghofer Medical Research Institute, Herston, Brisbane, Queensland, Australia; Faculty of Medicine (N.Q.L., W.H., M.H.L.), University of Queensland, Herston, Queensland, Australia; Faculty of Health (M.H.L.), School of Biomedical Sciences, Queensland University of Technology, St Lucia, Queensland, Australia
| | - Sarah E Medland
- Psychiatric Genetics (S.E.M.), QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - David A Mackey
- The University of Western Australia (D.A.M.), Centre for Ophthalmology and Visual Science (Incorporating the Lions Eye Institute), Perth, Western Australia, Australia
| | - Alex W Hewitt
- Menzies Institute for Medical Research (A.W.H.), University of Tasmania, Hobart, Tasmania, Australia; Centre for Eye Research Australia (A.W.H.), University of Melbourne, Melbourne, Victoria, Australia
| | - Puya Gharahkhani
- From the Statistical Genetics Lab (N.Q.L., W.H., M.H.L., P.G., S.M.), QIMR Berghofer Medical Research Institute, Herston, Brisbane, Queensland, Australia
| | - Stuart MacGregor
- From the Statistical Genetics Lab (N.Q.L., W.H., M.H.L., P.G., S.M.), QIMR Berghofer Medical Research Institute, Herston, Brisbane, Queensland, Australia.
| |
Collapse
|
3
|
Barkas F, Brandts J, De Bacquer D, Jennings C, De Backer GG, Kotseva K, Ryden L, Lip GY, Santos RD, Libby P, Erlund I, Ganly S, Vihervaara T, Adamska A, Abreu A, Almahmeed W, Ambari AM, Ge J, Hasan-Ali H, Huo Y, Jankowski P, Jimenez RM, Li Y, Mahmood Zuhdi AS, Makubi A, Mbakwem AC, Mbau L, Navarro Estrada JL, Ogah OS, Ogola EN, Quintero–Baiz A, Sani MU, Sosa Liprandi MI, Tan JWC, Urina Triana MA, Yeo TJ, Wood D, McEvoy JW, Ray KK, McEvoy J, Wood D, Adamska A, De Backer G, De Bacquer D, Erlund I, Ganly S, Jennings C, Kotseva K, Lip G, Ray K, Rydén L, Abreu A, Almahmeed W, Ambari A, Ge J, Hasan-Ali H, Huo Y, Jankowski P, Jimenez R, Li Y, Mahmood Zuhdi A, Makubi A, Mbakwem A, Mbau L, Navarro Estrada J, Ogah O, Ogola E, Quintero-Baiz A, Sani M, Sosa Liprandi M, Tan J, Urina Triana M, Yeo T, Wood D, Adamska A, Jennings C, Kotseva K, Ganly S, Zekic A, Rydén L, Salako D, Toulouse C, Chhabra G, Spragg R, Sullivan M, De Bacquer D, Erlund I, Jarvensivu E, Vihervaara T, Deschle H, Estrada JLN, Liprandi MIS, Bellia S, Villarreal R, Elfman M, Perea J, Lombardi F, Masson W, Castaño B, Gamarra A, et alBarkas F, Brandts J, De Bacquer D, Jennings C, De Backer GG, Kotseva K, Ryden L, Lip GY, Santos RD, Libby P, Erlund I, Ganly S, Vihervaara T, Adamska A, Abreu A, Almahmeed W, Ambari AM, Ge J, Hasan-Ali H, Huo Y, Jankowski P, Jimenez RM, Li Y, Mahmood Zuhdi AS, Makubi A, Mbakwem AC, Mbau L, Navarro Estrada JL, Ogah OS, Ogola EN, Quintero–Baiz A, Sani MU, Sosa Liprandi MI, Tan JWC, Urina Triana MA, Yeo TJ, Wood D, McEvoy JW, Ray KK, McEvoy J, Wood D, Adamska A, De Backer G, De Bacquer D, Erlund I, Ganly S, Jennings C, Kotseva K, Lip G, Ray K, Rydén L, Abreu A, Almahmeed W, Ambari A, Ge J, Hasan-Ali H, Huo Y, Jankowski P, Jimenez R, Li Y, Mahmood Zuhdi A, Makubi A, Mbakwem A, Mbau L, Navarro Estrada J, Ogah O, Ogola E, Quintero-Baiz A, Sani M, Sosa Liprandi M, Tan J, Urina Triana M, Yeo T, Wood D, Adamska A, Jennings C, Kotseva K, Ganly S, Zekic A, Rydén L, Salako D, Toulouse C, Chhabra G, Spragg R, Sullivan M, De Bacquer D, Erlund I, Jarvensivu E, Vihervaara T, Deschle H, Estrada JLN, Liprandi MIS, Bellia S, Villarreal R, Elfman M, Perea J, Lombardi F, Masson W, Castaño B, Gamarra A, Llamedo M, Santander P, Barco A, Flores V, Leyes C, Rodríguez F, Marturano M, Alustiza W, Duronto E, Procopio G, Higa C, Corigliano O, Sanchez M, del Cristo Mendoza Beltrán F, Luna P, Urina Triana M, Quintero-Baiz A, Renowitsky C, Pulgar M, Lievano J, Hernandez A, Cordoba G, Duque M, Duque L, Mendoza F, Rojas J, Jaramillo N, Zuluaga S, Restrepo A, Munos J, Buitrago A, Ramirez H, Figueroa F, Gonzales C, Cadena A, Bermudez S, Gomez L, Firdaus I, Dwiputra B, Arityanti D, Susilowati E, Hamdani R, Syaoqi M, Ridwan M, Fitra ND M, Ridwan M, Hartopo A, Arso I, Anggraeni V, Raynaldo A, Hasan H, Siregar Y, Tjahjono C, Sihotang F, Erickatulistiawan G, Tiksnadi B, Febrianora M, Tarsidin N, Fong A, Ahmad WAW, Zuhdi ASM, Zuhdi ASM, Ahmad WAW, Hadi M, Kassim ZA, Shaharudin N, Mohamad Sithik M, Moussa IM, Fong A, Ong T, Foo D, Jong R, Lee C, Kim H, Mahadevan G, Abdul Kader M, Aziz NA, Liew H, Jam E, Regaibalan D, Bugarin O, Leus A, Baniqued A, Arellano J, Mendoza L, Jimenez R, Mendoza R, Reyes M, Locnen S, Luna D, Tiongco R, Alad P, Besa J, Tirador L, Estoce E, Sian A, Roque R, Montejo A, Discípulo A, Lastimosa H, Ybanez E, Bernan A, Coronel R, Samson M, Tamayo T, Tan H, Tan J, Yeo T, Dalakoti M, Fadzillah NFM, Lim S, Low T, Ong J, Ting J, Wang L, Wong C, Ho J, Chee F, Cheng C, Koh C, Yip D, Huang Z, Chin C, Chua K, Fam J, Idu M, Keh Y, Lim C, Rizwan M, Tan J, Tsang C, Wong N, Yap J, Tan M, Tan J, Goh L, Tan K, Wong Y, Yee Y, Yuslane M, Weng Y, Wong C, Imran S, Lim Z, Lu T, Su J, Hoon V, Ho Y, Quek S, Shawki I, Hasan-Ali H, Abdelmegid M, Marghany A, Mosad E, Shafik E, Ibrahim A, Ghaleb R, Kholef E, Shamandy B, Moustafa T, Sobeih M, Mortada M, Elbarbary M, Elawady M, Al Ali J, Almahmeed W, Hashmani S, Manla Y, Green C, Garrod R, Nour S, Alhammadi AF, Mohamed Y, Isaac R, Bazargani N, Almulla A, Praveen J, Al Ali J, Bashir OAO, Faraj A, Hashmath S, Hamadi RL, Ahmad R, Prabakaran M, JI X, Ge J, Huo Y, Li Y, Ge J, Wang C, Zhao G, Mi S, Peng Y, Qi Z, Li Y, Bao L, Gao W, Huo Y, Zheng B, Fang J, Gu X, Su L, Wang Q, Tong S, Chen X, Fu Y, Li Y, Shi J, Jing J, Sheng L, He M, Wang S, Pan W, Kong Y, Cong H, Zhang Y, Chen J, Shi B, Zhao R, Zhang W, Ye J, Hou C, Huang H, Yang L, Chang L, Yang Z, Gao C, Zhang J, Zhang Y, Samia B, Ogola E, Mbau L, Mwazo K, Bajaber A, Nduati B, Mkilo J, Mwasha M, Sood M, Barasa F, Biwott P, Gitura B, Kinuthia P, Ogah O, Mbakwem A, Sani M, Orimolade O, Omoruyi E, Makinde O, Ojo V, Otesanya F, Amadi C, Ogedegbe S, Ejim E, Odom A, Udora N, Maduka C, Onyemachi J, Edafe E, Dodiyi-Manuel S, Bamigbowu O, Ossai G, Umuerri E, Oghojamoni-Ogefere P, Oyovwevotu M, Ojji D, Ajanya O, Alo-Joseph C, Daniel O, Ibrahim A, Ngada E, Ripiye N, Taiwo F, Ubah I, Ogunmodede J, Adeniyi S, Olugbola O, Sani M, Alfa I, Abdussalam T, Abdullahi U, Isezuo S, Zagga M, Umar H, Okocha H, Aloja N, Buba F, Talle M, Ahmadu N, Galtimari I, Tukur A, Zakariyya Z, Ayoola Y, Oyekunle R, Adamu S, Mvungi R, Makubi A, Kisenge P, Msigwa S, Meda J, Magitta N, Chandika A, Mitkowski P, Jankowski P, Kamiński K, Łapińska M, Sobkowicz B, Knapp M, Mickiewicz K, Dobrzycki S, Hirnle T, Charkiewicz-Szeremeta K, Bychowski J, Guzowska – Suchowolec A, Kubica A, Michalski P, Kosobucka-Ozdoba A, Pietrzykowski Ł, Rzepka-Cholasińska A, Ratajczak J, Siedlaczek M, Skonieczny G, Kostrzewa P, Gąsior Z, Maciejowski Ł, Matla-Hajzyk M, Hapeta-Zeman B, Szóstak-Janiak K, Kosior D, Kulak P, Rajska E, Kosior D, Rak A, Jankowski P, Koczwarska-Maciejek D, Wolfshaut-Wolak R, Gonçalves L, Pereira H, Abreu A, Carvalho M, Amador A, Calvão J, Costa C, Moreira H, Palma P, Pinto R, Proença T, Rocha M, Moreira N, Almeida J, Batista G, Borges-Rosa J, Campos G, Fernandes C, Guimarães J, Martinho S, Santos T, Silva A, Simões M, Morais J, Carvalho M, Gonçalves C, Martins A, Vazão A, Abreu A, Alves da Silva P, Brito J, Pires ML, Lima Martins A, Pinto F, Pinto R, Aguiar C, Amador R, Bello R, Lima R, Pais J, Almeida A, Caria M, Carrington M, Conde D, Francisco C, Kongo K, Patricio L, Peralta M, Rebola E, Rocha R, Silva F, Trinca M. Global Variation in Lipoprotein(a) Levels Among Patients With Coronary Heart Disease. J Am Coll Cardiol 2025; 85:2028-2042. [DOI: 10.1016/j.jacc.2025.04.010] [Show More Authors] [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: 06/04/2025]
|
4
|
Xu R, Wang Z, Dong J, Yu M, Zhou Y. Lipoprotein(a) and panvascular disease. Lipids Health Dis 2025; 24:186. [PMID: 40413492 DOI: 10.1186/s12944-025-02600-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Accepted: 05/08/2025] [Indexed: 05/27/2025] Open
Abstract
Panvascular disease (PVD) is an emerging clinical concept that encompasses a spectrum of atherosclerotic conditions involving multiple major vascular beds, including the coronary, cerebral, peripheral, and valvular arteries. Although not formally recognized as a nosological entity, in this review, PVD is adopted as a conceptual framework to reflect the systemic nature of atherosclerosis affecting vascular territories supplying the heart, brain, and peripheral circulation. This perspective enables a more integrated understanding of disease processes across organ systems that are often studied in isolation. Lipoprotein(a) [Lp(a)] is a genetically regulated, low-density lipoprotein (LDL)-like particle that has garnered increasing attention as an independent pathogenic risk factor for PVD. Accumulating evidence from epidemiological, genetic, and mechanistic studies has confirmed the multifaceted role of Lp(a) in promoting atherogenesis, vascular calcification, inflammation, and thrombogenesis across multiple vascular beds. Elevated Lp(a) levels are associated with increased cardiovascular and cerebrovascular event risk, even after controlling for traditional risk factors. This review systematically outlines the structure, genetic determinants, and pathogenic mechanisms of Lp(a), and synthesizes current clinical evidence regarding its role in various PVD subtypes. The interactions between Lp(a) and traditional cardiovascular risk factors such as hypercholesterolemia, diabetes, and hypertension are explored in depth, highlighting their synergistic contributions to vascular injury and disease progression. Furthermore, sex-based differences in Lp(a)-associated risk, response to therapy, and biological behavior are discussed, providing insights into personalized cardiovascular risk stratification. In addition, the review summarizes current and emerging therapeutic strategies targeting Lp(a), including niacin, antisense oligonucleotides (ASOs), small interfering RNAs (siRNAs), and gene-editing technologies. These advances offer promising new avenues for reducing residual cardiovascular risk attributable to elevated Lp(a). In conclusion, viewing Lp(a)-associated pathology through the lens of PVD provides a comprehensive and unifying approach to understanding its systemic impact. This framework supports the development of integrated risk assessment tools and multi-targeted interventions, ultimately aiming to improve outcomes for patients with complex, multisite vascular involvement.
Collapse
Affiliation(s)
- Ruiyan Xu
- Department of Clinical Medicine, Queen Mary School of Nanchang University, Nanchang, 330031, China
| | - Zhenwei Wang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
| | - Jiayu Dong
- Department of Clinical Medicine, Queen Mary School of Nanchang University, Nanchang, 330031, China
| | - Miao Yu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330008, China.
| | - Yue Zhou
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330008, China.
| |
Collapse
|
5
|
Kim JA, Kim NH. Lipoprotein(a) and Cardiovascular Risk in Asian Populations: A Comprehensive Review. J Lipid Atheroscler 2025; 14:174-187. [PMID: 40492182 PMCID: PMC12145969 DOI: 10.12997/jla.2025.14.2.174] [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: 04/08/2025] [Revised: 04/23/2025] [Accepted: 04/29/2025] [Indexed: 06/11/2025] Open
Abstract
Lipoprotein(a) [Lp(a)] is a genetically determined lipoprotein particle that plays a causal role in atherosclerotic cardiovascular disease (ASCVD), ischemic stroke, and calcific aortic valve stenosis. Structurally similar to low-density lipoprotein, Lp(a) contains apolipoprotein(a) [apo(a)], which imparts unique atherogenic properties. Although Lp(a) levels vary significantly by ethnicity, East Asians generally have lower median concentrations, attributed to a higher frequency of large apo(a) isoforms and fewer high-risk LPA gene variants. However, even modest elevations in Lp(a) are associated with increased ASCVD risk in Asians, especially among high-risk populations. Observational studies from Asian populations have shown that elevated Lp(a) levels are linked to coronary artery calcification, myocardial infarction, stroke, and recurrent cardiovascular events. Novel therapeutic agents, including proprotein convertase subtilisin/kexin type 9 inhibitors, inclisiran, and antisense oligonucleotides such as pelacarsen, have demonstrated promising effects in lowering Lp(a). These therapies are currently under investigation in outcome trials, including Asian subgroups. Given the high burden of cardiovascular disease and ethnic variability in Lp(a) distribution and genetic determinants, routine measurement of Lp(a) could improve risk stratification and therapeutic decision-making. This review summarizes current evidence regarding the epidemiology, genetic background, clinical relevance, and emerging therapeutic strategies targeting Lp(a) in Asian populations, highlighting the need for population-specific thresholds and further research to guide clinical practice.
Collapse
Affiliation(s)
- Jung A Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Nam Hoon Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| |
Collapse
|
6
|
Nazir A, Nazir A, Afzaal U, Aman S, Sadiq SUR, Akah OZ, Jamal MSW, Hassan SZ. Advancements in Biomarkers for Early Detection and Risk Stratification of Cardiovascular Diseases-A Literature Review. Health Sci Rep 2025; 8:e70878. [PMID: 40432692 PMCID: PMC12106349 DOI: 10.1002/hsr2.70878] [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: 06/24/2024] [Revised: 03/16/2025] [Accepted: 05/02/2025] [Indexed: 05/29/2025] Open
Abstract
Introduction CVDs is a leading cause of morbidity, mortality, and healthcare expenditure worldwide. Identifying individuals at risk or in the incipient stages of disease is instrumental in enabling timely interventions, preventive measures, and tailored treatment regimens. The landscape of CVDs is complicated by their heterogeneity, encompassing a spectrum of conditions such as coronary artery disease, heart failure, arrhythmias, and valvular disorders. In recent years, the integration of biomarkers into cardiovascular medicine has emerged as a paradigm-shifting approach with the potential to revolutionize early detection and risk stratification. By synthesizing a multitude of studies, we aim to provide a comprehensive resource that illuminates the transformative potential of biomarkers in ushering in a new era of precision cardiovascular medicine. Aim To identify the biomarkers for the detection and diagnosis of CVDs. Materials and Methods This review examines key studies from 2015 to the present that investigate the impact of cardiac biomarkers on cardiovascular outcomes. Data were gathered from PubMed, Cochrane Library, and Embase to ensure a comprehensive analysis. The review focuses on various cardiac biomarkers, assessing their levels and changes in relation to cardiovascular health, with special emphasis on advanced biomarkers such as proteomic and metabolomic markers in cardiovascular disease (CVD) diagnosis. Peer-reviewed studies published in English that evaluated the diagnostic, prognostic, or therapeutic role of cardiac biomarkers were included, with priority given to clinical trials, cohort studies, systematic reviews, and meta-analyses providing quantitative biomarker data. Studies unrelated to cardiac biomarkers, case reports, editorials, conference abstracts, and those with small sample sizes or insufficient methodological rigor were excluded. The review also accounts for potential confounding factors and research limitations, ensuring a balanced assessment of the literature. By synthesizing data from academic papers, clinical reports, and research articles, this study provides a comprehensive evaluation of the evolving role of cardiac biomarkers in CVD diagnosis and risk stratification. Results Biomarkers play a pivotal role in cardiovascular disease risk prediction, diagnosis, and treatment by providing dynamic biological insights. High-sensitivity cardiac troponins (hs-cTn) enhance myocardial injury detection, while circulating microRNAs (miR-208, miR-499) serve as early indicators of myocardial infarction and heart failure. Lipoprotein(a) [Lp(a)] predicts long-term cardiovascular risk, and inflammatory biomarkers such as C-reactive protein (CRP) and interleukin-6 (IL-6) are linked to adverse outcomes. Multi-biomarker panels, such as hs-cTn with B-type natriuretic peptide (BNP), improve heart failure prognosis, while metabolomic profiling enables precision medicine. Additionally, biomarkers like BNP and NT-proBNP facilitate real-time therapeutic monitoring. These findings underscore the critical role of biomarkers in refining risk stratification, improving diagnostic accuracy, and enabling personalized treatment strategies in cardiovascular medicine. Conclusion The advancement of cardiovascular biomarkers has significantly enhanced early detection, risk stratification, and personalized treatment. Emerging biomarkers, including genetic variants, metabolomics, microRNAs, and imaging-based markers, provide deeper insights into disease mechanisms. Integrating multi-omic approaches with artificial intelligence may further refine predictive accuracy and therapeutic decision-making. However, clinical translation requires rigorous validation through large-scale, multicenter studies to ensure reliability and applicability across diverse populations. Standardization, cost-effectiveness assessments, and the development of biomarker panels are essential for clinical adoption. Future research should focus on bridging discovery and implementation, advancing precision medicine to improve cardiovascular outcomes.
Collapse
Affiliation(s)
- Abubakar Nazir
- Oli Health Magazine Organization, Research and EducationKigaliRwanda
- Department of MedicineKing Edward Medical UniversityLahorePakistan
| | - Awais Nazir
- Oli Health Magazine Organization, Research and EducationKigaliRwanda
- Department of MedicineKing Edward Medical UniversityLahorePakistan
| | - Usama Afzaal
- Oli Health Magazine Organization, Research and EducationKigaliRwanda
- Department of MedicineKing Edward Medical UniversityLahorePakistan
| | - Shafaq Aman
- Department of MedicineKing Edward Medical UniversityLahorePakistan
- St John of God Midland HospitalsAustralia
| | | | | | | | - Syed Zawahir Hassan
- Division of Cardiovascular PreventionHouston Methodist DeBakey Heart & Vascular CenterHoustonUSA
| |
Collapse
|
7
|
Liu W, Chen S, Yang C, Lei F, Huang X, Zhang X, Sun T, Lin L, Wang C, Cao Y, She ZG, Xiao X, Li H. Elevated high-density lipoprotein triglycerides increase atherosclerotic risk. J Lipid Res 2025; 66:100791. [PMID: 40164335 PMCID: PMC12088758 DOI: 10.1016/j.jlr.2025.100791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 03/25/2025] [Accepted: 03/27/2025] [Indexed: 04/02/2025] Open
Abstract
The relationship between high-density lipoprotein (HDL) and atherosclerotic risk remains incompletely elucidated, potentially due to the inherent heterogeneity of HDL particles. Hypertriglyceridemia is associated with alterations in HDL composition. This study investigated the impact of elevated triglycerides (TG) on HDL and its association with coronary artery disease (CAD) risk using a large prospective cohort study and Mendelian randomization (MR). We found that elevated TG was associated with reduced HDL particle size, decreased concentrations of HDL components, and increased triglycerides in HDL (HDL-TG) (all P for trend < 0.001). The protective effects of HDL particle concentration and HDL cholesterol on CAD are attenuated with increasing serum TG levels. An independent and positive association between HDL-TG levels and incident CAD events (hazard ratio [HR] per 1 standard deviation increase: 1.066, 95% CI: 1.052-1.080, P < 0.001) was confirmed even after adjustment for established cardiovascular disease risk factors. MR analyses supported a causal role for HDL-TG in CAD development (inverse-variance weighted [IVW] method: odds ratios [ORs] of 1.120 (95% CI: 1.053-1.192, P < 0.001) and 1.141 (95% CI: 1.032-1.263, P = 0.010) for dataset groups 1 and 2, respectively). Drug-target MR analyses suggested a potential association between omega-3 fatty acids (OM3-FA) and lower HDL-TG levels, with LPL and DGAT2 as key pharmacological targets. Our findings suggest that elevated TG contributes to adverse alterations in HDL, elevating CAD risk. HDL-TG is an independent positive risk factor for CAD and a potential causal contributor to CAD development. OM3-FA supplementation may offer a therapeutic strategy for mitigating the CAD risk associated with elevated HDL-TG.
Collapse
Affiliation(s)
- Weifang Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China; State Key Laboratory of New Drug Discovery and Development for Major Diseases, Gannan Medical University, Ganzhou, China; Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China
| | - Shaoze Chen
- Department of Cardiology, Huanggang Central Hospital of Yangtze University, Huanggang, China
| | - Chengzhang Yang
- Department of Cardiology, Huanggang Central Hospital of Yangtze University, Huanggang, China
| | - Fang Lei
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xuewei Huang
- Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Xingyuan Zhang
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Tao Sun
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Lijin Lin
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Chuansen Wang
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yuanyuan Cao
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zhi-Gang She
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China; State Key Laboratory of New Drug Discovery and Development for Major Diseases, Gannan Medical University, Ganzhou, China; Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China.
| | - Xuan Xiao
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.
| | - Hongliang Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China; State Key Laboratory of New Drug Discovery and Development for Major Diseases, Gannan Medical University, Ganzhou, China; Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China.
| |
Collapse
|
8
|
de Vries P, Hasbani N, Heath A, Hodonsky C, Hahn J, Meena D, Lu H, Dehghan AA, Kavousi M, Voight B, Peyser P, Morrison A, Assimes T, Damrauer S, Miller C. A multi-trait genome-wide association study of coronary artery disease and subclinical atherosclerosis traits. RESEARCH SQUARE 2025:rs.3.rs-6456056. [PMID: 40313769 PMCID: PMC12045367 DOI: 10.21203/rs.3.rs-6456056/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
Measures of subclinical atherosclerosis, such as coronary artery calcification (CAC) and carotid intima-media thickness (CIMT), reflect the underlying pathophysiology of coronary artery disease (CAD) and are genetically correlated with CAD and related risk factors. Leveraging summary statistics from genome-wide association studies of CAD, CIMT, CAC, type 2 diabetes, low-density lipoprotein cholesterol, and systolic blood pressure, we performed 15 separate multi-trait GWAS to identify shared susceptibility loci and elucidate the pleiotropic architecture underlying atherosclerosis. We identified 442 shared risk loci across all analyses that met an experiment-wide Bonferroni threshold of 3.3 × 10-9, uncovering 195 novel atherosclerosis loci. Multi-trait colocalization confirmed a shared causal signal in 25 shared novel loci for atherosclerosis. Trait-eQTL colocalization identified evidence of a shared causal signal in arterial, subcutaneous adipose, and cardiac tissues, implicating genes such as PRRX2, BNC2, CLIC4, SCAI, and PPP6C, and pathways related to vascular remodeling, inflammation, and metabolic regulation.
Collapse
Affiliation(s)
| | | | - Adam Heath
- The University of Texas Health Science Center at Houston
| | | | - Julie Hahn
- The University of Texas Health Science Center at Houston
| | | | | | | | | | | | - Patricia Peyser
- Department of Epidemiology, School of Public Health, University of Michigan
| | | | | | | | | |
Collapse
|
9
|
Mekhael M, Bidaoui G, Falloon A, Pandey AC. Personalization of primary prevention: Exploring the role of coronary artery calcium and polygenic risk score in cardiovascular diseases. Trends Cardiovasc Med 2025; 35:154-163. [PMID: 39442739 DOI: 10.1016/j.tcm.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 10/14/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024]
Abstract
Personalized healthcare is becoming increasingly popular given the vast heterogeneity in disease manifestation between individuals. Many commonly encountered diseases within cardiology are multifactorial in nature and disease progression and response is often variable due to environmental and genetic factors influencing disease states. This makes accurate early identification and primary prevention difficult in certain populations, especially young patients with limited Atherosclerotic Cardiovascular Disease (ASCVD) risk factors. Newer strategies, such as coronary artery calcium (CAC) scans and polygenic risk scores (PRS), are being implemented to aid in the detection of subclinical disease and heritable risk, respectively. Data surrounding CAC scans have shown promising results in their ability to detect subclinical atherosclerosis and predict the risk of future coronary events, especially at the extremes; however, predictive variability exists among different patient populations, limiting the test's specificity. Furthermore, relying only on CAC scores and ASCVD risk scores may fail to identify a large group of patients needing primary prevention who lack subclinical disease and traditional risk factors, but harbor genetic variabilities strongly associated with certain cardiovascular diseases. PRS can overcome these limitations. These scores can be measured in individuals as early as birth to identify genetic variants placing them at elevated risk for developing cardiovascular disease, irrespective of their current cardiovascular health status. By applying PRS alongside CAC scores, previously overlooked patient populations can be identified and begin primary prevention strategies early to achieve optimal outcomes. In this review, we expand on the current knowledge surrounding CAC scores and PRS and highlight the future possibilities of these technologies for preventive cardiology.
Collapse
Affiliation(s)
- Mario Mekhael
- Section of Cardiology, Deming Dept of Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| | - Ghassan Bidaoui
- Section of Cardiology, Deming Dept of Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| | - Austin Falloon
- Section of Cardiology, Deming Dept of Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| | - Amitabh C Pandey
- Section of Cardiology, Deming Dept of Medicine, Tulane University School of Medicine, New Orleans, LA, United States; Southeast Louisiana Veterans Health Care System, New Orleans, LA, United States.
| |
Collapse
|
10
|
Alexander MR, Edwards TL, Harrison DG. GWAS for Defining the Pathogenesis of Hypertension: Have They Delivered? Hypertension 2025; 82:573-582. [PMID: 39936322 PMCID: PMC11922662 DOI: 10.1161/hypertensionaha.124.23451] [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] [Indexed: 02/13/2025]
Abstract
Genome-wide association studies have identified >3500 associated single nucleotide polymorphisms and over 1000 independent loci associated with hypertension. These individually have small effect sizes, and few associated loci have been experimentally tested for causal roles in hypertension using animal models or in humans. Thus, methods to prioritize and maximize the relevance of identified single nucleotide polymorphisms and associated loci are critical to determine their importance in hypertension. We propose several approaches to aid in these efforts, including: (1) integration of genome-wide association study data with multiomic data sets, including proteomics, transcriptomics, and epigenomics, (2) utilizing linked clinical and genetic data sets to determine genetic contributions to hypertension subphenotypes with distinct drivers, and (3) performing whole exome/genome sequencing on cohorts of individuals with severe hypertension to enrich for rare variants with larger effect sizes. Rather than creating longer lists of hypertension-associated single nucleotide polymorphisms, these approaches are needed to identify key mediators of hypertension pathophysiology.
Collapse
Affiliation(s)
- Matthew R Alexander
- Department of Medicine, Division of Clinical Pharmacology (M.R.A., D.G.H.), Vanderbilt University Medical Center, Nashville, TN
- Division of Cardiovascular Medicine (M.R.A., D.G.H.), Vanderbilt University Medical Center, Nashville, TN
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN (M.R.A., D.G.H.)
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Nashville, TN (M.R.A., D.G.H.)
| | - Todd L Edwards
- Division of Epidemiology, Department of Medicine (T.L.E.), Vanderbilt University Medical Center, Nashville, TN
| | - David G Harrison
- Department of Medicine, Division of Clinical Pharmacology (M.R.A., D.G.H.), Vanderbilt University Medical Center, Nashville, TN
- Division of Cardiovascular Medicine (M.R.A., D.G.H.), Vanderbilt University Medical Center, Nashville, TN
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN (M.R.A., D.G.H.)
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Nashville, TN (M.R.A., D.G.H.)
| |
Collapse
|
11
|
Aminbakhsh AP, Théberge ET, Burden E, Adejumo CK, Gravely AK, Lehman A, Sedlak TL. Exploring associations between estrogen and gene candidates identified by coronary artery disease genome-wide association studies. Front Cardiovasc Med 2025; 12:1502985. [PMID: 40182431 PMCID: PMC11965610 DOI: 10.3389/fcvm.2025.1502985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 03/04/2025] [Indexed: 04/05/2025] Open
Abstract
Introduction Coronary artery disease (CAD) is the leading cause of death around the world, with epidemiological sex and gender differences in prevalence, pathophysiology and outcomes. It has been hypothesized that sex steroids, like estrogen, may contribute to these sex differences. There is a relatively large genetic component to developing CAD, with heritability estimates ranging between 40%-60%. In the last two decades, genome-wide association studies (GWAS) have contributed substantially to advancing the understanding of genetic candidates contributing to CAD. The aim of this study was to determine if genes discovered in CAD GWASs are affected by estrogen via direct modulation or indirect down-stream targets. Methods A scoping review was conducted using MEDLINE and EMBASE for studies of atherosclerotic coronary artery disease and a genome-wide association study (GWAS) design. Analysis was limited to candidate genes with corresponding single nucleotide polymorphisms (SNPs) surpassing genome-wide significance and had been mapped to genes by study authors. The number of studies that conducted sex-stratified analyses with significant genes were quantified. A literature search of the final gene lists was done to examine any evidence suggesting estrogen may modulate the genes and/or gene products. Results There were 60 eligible CAD GWASs meeting inclusion criteria for data extraction. Of these 60, only 36 had genome-wide significant SNPs reported, and only 3 of these had significant SNPs from sex-stratified analyses mapped to genes. From these 36 studies, a total of 61 genes were curated, of which 26 genes (43%) were found to have modulation by estrogen. All 26 were discovered in studies that adjusted for sex. 12/26 genes were also discovered in studies that conducted sex-stratified analyses. 12/26 genes were classified as having a role in lipid synthesis, metabolism and/or lipoprotein mechanisms, while 11/26 were classified as having a role in vascular integrity, and 3/26 were classified as having a role in thrombosis. Discussion This study provides further evidence of the relationship between estrogen, genetic risk and the development of CAD. More sex-stratified research will need to be conducted to further characterize estrogen's relation to sex differences in the pathology and progression of CAD.
Collapse
Affiliation(s)
- Ava P. Aminbakhsh
- Department of Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Emilie T. Théberge
- Department of Medical Genetics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Elizabeth Burden
- Division of Internal Medicine, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
- Vancouver Coastal Health, Vancouver, BC, Canada
| | - Cindy Kalenga Adejumo
- Division of Internal Medicine, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
- Vancouver Coastal Health, Vancouver, BC, Canada
| | - Annabel K. Gravely
- Department of Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Anna Lehman
- Department of Medical Genetics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- Vancouver Coastal Health, Vancouver, BC, Canada
| | - Tara L. Sedlak
- Vancouver Coastal Health, Vancouver, BC, Canada
- Division of Cardiology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
12
|
Hu Y, Cui X, Lu M, Guan X, Li Y, Zhang L, Lin L, Zhang Z, Zhang M, Hao J, Wang X, Huan J, Li Y, Li C. Body Fat Distribution and Ectopic Fat Accumulation as Mediator of Diabetogenic Action of Lipid-Modifying Drugs: A Mediation Mendelian Randomization Study. Mayo Clin Proc 2025; 100:424-439. [PMID: 39918451 DOI: 10.1016/j.mayocp.2024.10.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 10/06/2024] [Accepted: 10/25/2024] [Indexed: 05/08/2025]
Abstract
OBJECTIVE To investigate the causal relationship between various lipid-modifying drugs and new-onset diabetes, as well as the mediators contributing to this relationship. METHODS Mediation Mendelian randomization was performed to investigate the causal effect of lipid-modifying drug targets on type 2 diabetes (T2D) outcomes and the proportion of this association that is mediated through ectopic fat accumulation traits. Specific sets of variants in or near genes that encode 11 lipid-modifying drug targets (LDLR, HMGCR, NPC1L1, PCSK9, APOB, ABCG5/ABCG8, LPL, PPARA, ANGPTL3, APOC3, and CETP; for expansion of gene symbols, use search tool at www.genenames.org) were extracted. Random effects inverse variance weighted were performed to evaluate the causal effects among outcomes. Mediation analyses were performed to identify the mediators of the association between lipid-modifying drugs and T2D. The study was conducted from November 10, 2023, to April 2, 2024 RESULTS: The genetic mimicry of HMGCR and APOB inhibition was associated with an increased T2D risk, whereas the genetic mimicry of LPL enhancement was linked to a lower T2D risk. Gluteofemoral adipose tissue volume was a mediator for explaining 9.52% (P=.002), 16.90% (P=.03), and 10.50% (P=.003) of the total effect of HMGCR, APOB, and LPL on T2D susceptibility, respectively. Liver fat was a mediator for explaining 21.12% (P=.005), 12.28% (P=.03), and 9.84% (P=.005) of the total effect of HMGCR, APOB, and LPL on T2D susceptibility, respectively. CONCLUSION Our findings support the hypothesis that liver fat and gluteofemoral adipose tissue play a mediating role in the prodiabetic effects of HMGCR and APOB inhibition, as well as in the antidiabetic effects of LPL enhancement.
Collapse
Affiliation(s)
- Yuanlong Hu
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Xinhai Cui
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Mengkai Lu
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Xiuya Guan
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yuan Li
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Lei Zhang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Lin Lin
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Zhiyuan Zhang
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Muxin Zhang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Jiaqi Hao
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Xiaojie Wang
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macau, China
| | - Jiaming Huan
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yunlun Li
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China; Department of Cardiovascular, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China.
| | - Chao Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China.
| |
Collapse
|
13
|
Phair IR, Sovakova M, Alqurashi N, Nisr RB, McNeilly AD, Lamont D, Rena G. In-depth proteomic profiling identifies potentiation of the LPS response by 7-ketocholesterol. JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY PLUS 2025; 11:100285. [PMID: 39991505 PMCID: PMC11847031 DOI: 10.1016/j.jmccpl.2025.100285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/23/2025] [Accepted: 01/28/2025] [Indexed: 02/25/2025]
Abstract
In patients with stable coronary artery disease, plasma levels of 7-ketocholesterol (7-KC), found at high levels in atherosclerotic lesions, predict risk of incident heart failure dose dependently, potentially contributing to disease aetiology. Previous studies demonstrated that 7-KC can elicit effects on macrophage function; however, effects of 7-KC on the macrophage proteome have not been studied systematically. Here we used quantitative mass spectrometry to establish the effect of 7-KC on the mouse macrophage proteome. 7-KC independently mediated dynamic changes, including on atherogenic/M1 markers, cholesterol metabolism, biosynthesis and transport, as well as nutrient transport more broadly. These changes were however insufficient alone to drive changes in cytokine and chemokine secretion. Rather, they prime the macrophage, potentiating LPS-stimulated TNF alpha secretion and key pro-inflammatory enzymes. Our results indicate that 7-KC has independent metabolic effects on the macrophage; however, effects on the immune system are primarily due to the changes in metabolism priming the response to an inflammatory stimulus. Earlier findings from CANTOS and the recent FDA approval of colchicine highlight that inflammation is a viable target for cardiovascular disease; however, it is currrently unclear which will be the best anti-inflammatory targets to pursue in the future. In this context, our findings suggest that drugs targeting atherogenic markers induced by 7-KC might be well tolerated, as they will not necessarily be expected to be immunosuppressive.
Collapse
Affiliation(s)
- Iain R. Phair
- Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee DD1 9SY, Scotland, United Kingdom
| | - Magdalena Sovakova
- Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee DD1 9SY, Scotland, United Kingdom
| | - Noor Alqurashi
- Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee DD1 9SY, Scotland, United Kingdom
| | - Raid B. Nisr
- Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee DD1 9SY, Scotland, United Kingdom
| | - Alison D. McNeilly
- Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee DD1 9SY, Scotland, United Kingdom
| | - Douglas Lamont
- Centre for Advanced Scientific Technologies, School of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland, United Kingdom
| | - Graham Rena
- Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee DD1 9SY, Scotland, United Kingdom
| |
Collapse
|
14
|
Wei X, Wang M, Yu S, Han Z, Li C, Zhong Y, Zhang M, Yang T. Mapping the knowledge of omics in myocardial infarction: A scientometric analysis in R Studio, VOSviewer, Citespace, and SciMAT. Medicine (Baltimore) 2025; 104:e41368. [PMID: 39960900 PMCID: PMC11835070 DOI: 10.1097/md.0000000000041368] [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: 11/20/2023] [Accepted: 01/09/2025] [Indexed: 02/20/2025] Open
Abstract
Many researchers nowadays choose multi-omics techniques for myocardial infarction studies. However, there's yet to be a review article integrating myocardial infarction multi-omics. Hence, this study adopts the popular bibliometrics. Based on its principles, we use software like R Studio, Vosviewer, Citespace, and SciMAT to analyze literature data of myocardial infarction omics research (1991-2022) from Web of Science. By extracting key information and calculating weights, we conduct analyses from 4 aspects: Collaboration Network Analysis, Co-word Analysis, Citing and Cited Journal Analysis, and Co-citation and Clustering Analysis, aiming to understand the field's cooperation, research topic evolution, and knowledge flow. The results show that myocardial infarction omics research is still in its early stage with limited international cooperation. In terms of knowledge flow, there's no significant difference within the discipline, but non-biomedical disciplines have joined, indicating an interdisciplinary integration trend. In the overall research field, genomics remains the main topic with many breakthroughs identifying susceptibility sites. Meanwhile, other omics fields like lipidomics and proteomics are also progressing, clarifying the pathogenesis. The cooperation details in this article enable researchers to connect with others, facilitating their research. The evolution trend of subject terms helps them set goals and directions, quickly grasp the development context, and read relevant literature. Journal analysis offers submission suggestions, and the analysis of research base and frontier provides references for the research's future development.
Collapse
Affiliation(s)
- Xuan Wei
- Key Laboratory of Evidence Science, China University of Political Science and Law, Ministry of Education, Beijing, China
- Institute of Evidence Law and Forensic Science, China University of Political Science and Law, Beijing, China
| | - Min Wang
- Key Laboratory of Evidence Science, China University of Political Science and Law, Ministry of Education, Beijing, China
- Institute of Evidence Law and Forensic Science, China University of Political Science and Law, Beijing, China
| | - Shengnan Yu
- Key Laboratory of Evidence Science, China University of Political Science and Law, Ministry of Education, Beijing, China
- Institute of Evidence Law and Forensic Science, China University of Political Science and Law, Beijing, China
| | - Zhengqi Han
- Institute for Digital Technology and Law (IDTL), China University of Political Science and Law, Beijing, China
- CUPL Scientometrics and Evaluation Center of Rule of Law, China University of Political Science and Law, Beijing, China
| | - Chang Li
- Key Laboratory of Evidence Science, China University of Political Science and Law, Ministry of Education, Beijing, China
- Institute of Evidence Law and Forensic Science, China University of Political Science and Law, Beijing, China
| | - Yue Zhong
- Key Laboratory of Evidence Science, China University of Political Science and Law, Ministry of Education, Beijing, China
- Institute of Evidence Law and Forensic Science, China University of Political Science and Law, Beijing, China
| | - Mengzhou Zhang
- Key Laboratory of Evidence Science, China University of Political Science and Law, Ministry of Education, Beijing, China
- Institute of Evidence Law and Forensic Science, China University of Political Science and Law, Beijing, China
| | - Tiantong Yang
- Key Laboratory of Evidence Science, China University of Political Science and Law, Ministry of Education, Beijing, China
- Institute of Evidence Law and Forensic Science, China University of Political Science and Law, Beijing, China
| |
Collapse
|
15
|
Palanisamy S, Burka S, Blaha MJ. Coronary Artery Calcium Scoring in the Context of Widespread Lipoprotein(a) Testing: Clinical Considerations and Implications for Lipid-Lowering Therapies. Curr Cardiol Rep 2025; 27:52. [PMID: 39932648 DOI: 10.1007/s11886-025-02210-1] [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] [Accepted: 01/30/2025] [Indexed: 05/08/2025]
Abstract
PURPOSE OF REVIEW This review evaluates the interplay between lipoprotein(a) [Lp(a)] and coronary artery calcium (CAC) for risk prediction and preventive therapy selection, with a special emphasis on scenarios where these measures are discordant, particularly in otherwise intermediate-risk, primary prevention patients. RECENT FINDINGS Observational studies and meta-analyses indicate a nuanced relationship between elevated Lp(a) levels and CAC burden and progression. Elevated Lp(a) is associated with an increased risk of CAC presence and progression; although, there is notable variability across studies. CAC predicts a similarly elevated risk in patients with low and high Lp(a). Joint elevation of Lp(a) and CAC is associated with a very high-risk patient subset. Elevated Lp(a) should prompt consideration of CAC testing for further risk stratification. In the future, we anticipate that an elevated CAC score could prompt consideration of testing for Lp(a) in select patients, as identifying or confirming elevated Lp(a) may help guide the use of dedicated Lp(a)-lowering therapies in very high-risk primary prevention populations.
Collapse
Affiliation(s)
- Srikanth Palanisamy
- Johns Hopkins Ciccarone Center for the Prevention of Heart Disease, Blalock 524D1, 600 N. Wolfe St., Baltimore, 2287, MD, USA
| | - Semenawit Burka
- Johns Hopkins Ciccarone Center for the Prevention of Heart Disease, Blalock 524D1, 600 N. Wolfe St., Baltimore, 2287, MD, USA
| | - Michael J Blaha
- Johns Hopkins Ciccarone Center for the Prevention of Heart Disease, Blalock 524D1, 600 N. Wolfe St., Baltimore, 2287, MD, USA.
| |
Collapse
|
16
|
Fan W, Wu C, Wong ND. Lipoprotein(a) Atherosclerotic Cardiovascular Disease Risk Score Development and Prediction in Primary Prevention From Real-World Data. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2025; 18:e004631. [PMID: 39851061 PMCID: PMC11849056 DOI: 10.1161/circgen.124.004631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 11/19/2024] [Indexed: 01/25/2025]
Abstract
BACKGROUND Lipoprotein(a) [Lp(a)] is a predictor of atherosclerotic cardiovascular disease (ASCVD); however, there are few algorithms incorporating Lp(a), especially from real-world settings. We developed an electronic health record (EHR)-based risk prediction algorithm including Lp(a). METHODS Utilizing a large EHR database, we categorized Lp(a) cut points at 25, 50, and 75 mg/dL and constructed 10-year ASCVD risk prediction models incorporating Lp(a), with external validation in a pooled cohort of 4 US prospective studies. Net reclassification improvement was determined among borderline-intermediate risk patients. RESULTS We included 5902 patients aged ≥18 years (mean age 48.7±16.7 years, 51.2% women, and 7.7% Black). Our EHR model included Lp(a), age, sex, Black race/ethnicity, systolic blood pressure, total and high-density lipoprotein cholesterol, diabetes, smoking, and hypertension medication. Over a mean follow-up of 6.8 years, ASCVD event rates (per 1000 person-years) ranged from 8.7 to 16.7 across Lp(a) groups. A 25 mg/dL increment in Lp(a) was associated with an adjusted hazard ratio of 1.23 (95% CI, 1.10-1.37) for composite ASCVD. Those with Lp(a) ≥75 mg/dL had an 88% higher risk of ASCVD (hazard ratio, 1.88 [95% CI, 1.30-2.70]) and more than double the risk of incident stroke (hazard ratio, 2.55 [95% CI, 1.54-4.23]). C-statistics for our EHR and EHR+Lp(a) models in our EHR training data set were 0.7475 and 0.7556, respectively, with external validation in our pooled cohort (n=21 864) of 0.7350 and 0.7368, respectively. Among those at borderline/intermediate risk, the net reclassification improvement was 21.3%. CONCLUSIONS We show the feasibility of developing an improved ASCVD risk prediction model incorporating Lp(a) based on a real-world adult clinic population. The inclusion of Lp(a) in ASCVD prediction models can reclassify risk in patients who may benefit from more intensified ASCVD prevention efforts.
Collapse
Affiliation(s)
- Wenjun Fan
- Mary and Steve Wen Cardiovascular Division, Department of Medicine, University of California, Irvine
- Department of Epidemiology and Biostatistics, University of California, Irvine
| | - Chuyue Wu
- Department of Epidemiology, University of California, Los Angeles, CA
| | - Nathan D. Wong
- Mary and Steve Wen Cardiovascular Division, Department of Medicine, University of California, Irvine
- Department of Epidemiology and Biostatistics, University of California, Irvine
| |
Collapse
|
17
|
Kohutek ZA, Caslin HL, Fehrenbach DJ, Heimlich JB, Brown JD, Madhur MS, Ferrell PB, Doran AC. Bone Marrow Niche in Cardiometabolic Disease: Mechanisms and Therapeutic Potential. Circ Res 2025; 136:325-353. [PMID: 39883790 PMCID: PMC11790260 DOI: 10.1161/circresaha.124.323778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2025]
Abstract
Cardiovascular and cardiometabolic diseases are leading causes of morbidity and mortality worldwide, driven in part by chronic inflammation. Emerging research suggests that the bone marrow microenvironment, or marrow niche, plays a critical role in both immune system regulation and disease progression. The bone marrow niche is essential for maintaining hematopoietic stem cells (HSCs) and orchestrating hematopoiesis. Under normal conditions, this niche ensures a return to immune homeostasis after acute stress. However, in the setting of inflammatory conditions such as those seen in cardiometabolic diseases, it becomes dysregulated, leading to enhanced myelopoiesis and immune activation. This review explores the reciprocal relationship between the bone marrow niche and cardiometabolic diseases, highlighting how alterations in the niche contribute to disease development and progression. The niche regulates HSCs through complex interactions with stromal cells, endothelial cells, and signaling molecules. However, in the setting of chronic diseases such as hypertension, atherosclerosis, and diabetes, inflammatory signals disrupt the balance between HSC self-renewal and differentiation, promoting the excessive production of proinflammatory myeloid cells that exacerbate the disease. Key mechanisms discussed include the effects of hyperlipidemia, hyperglycemia, and sympathetic nervous system activation on HSC proliferation and differentiation. Furthermore, the review emphasizes the role of epigenetic modifications and metabolic reprogramming in creating trained immunity, a phenomenon whereby HSCs acquire long-term proinflammatory characteristics that sustain disease states. Finally, we explore therapeutic strategies aimed at targeting the bone marrow niche to mitigate chronic inflammation and its sequelae. Novel interventions that modulate hematopoiesis and restore niche homeostasis hold promise for the treatment of cardiometabolic diseases. By interrupting the vicious cycle of inflammation and marrow dysregulation, such therapies may offer new avenues for reducing cardiovascular risk and improving patient outcomes.
Collapse
Affiliation(s)
- Zachary A. Kohutek
- Department of Radiation Oncology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Heather L. Caslin
- Department of Health and Human Performance, University of Houston, Houston, TX 77204, USA
| | - Daniel J. Fehrenbach
- Department of Medicine, Division of Clinical Pharmacology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - J. Brett Heimlich
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Jonathan D. Brown
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Meena S. Madhur
- Department of Medicine, Division of Clinical Pharmacology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - P. Brent Ferrell
- Department of Medicine, Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University, Nashville, TN 37212, USA
| | - Amanda C. Doran
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University, Nashville, TN 37212, USA
| |
Collapse
|
18
|
Narayanan S, Vuckovic S, Bergman O, Wirka R, Verdezoto Mosquera J, Chen QS, Baldassarre D, Tremoli E, Veglia F, Lengquist M, Aherrahrou R, Razuvaev A, Gigante B, Björck HM, Miller CL, Quertermous T, Hedin U, Matic L. Atheroma transcriptomics identifies ARNTL as a smooth muscle cell regulator and with clinical and genetic data improves risk stratification. Eur Heart J 2025; 46:308-322. [PMID: 39552248 PMCID: PMC11735083 DOI: 10.1093/eurheartj/ehae768] [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: 09/13/2023] [Revised: 02/10/2024] [Accepted: 10/23/2024] [Indexed: 11/19/2024] Open
Abstract
BACKGROUND AND AIMS The role of vascular smooth muscle cells (SMCs) in atherosclerosis has evolved to indicate causal genetic links with the disease. Single cell RNA sequencing (scRNAseq) studies have identified multiple cell populations of mesenchymal origin within atherosclerotic lesions, including various SMC sub-phenotypes, but it is unknown how they relate to patient clinical parameters and genetics. Here, mesenchymal cell populations in atherosclerotic plaques were correlated with major coronary artery disease (CAD) genetic variants and functional analyses performed to identify SMC markers involved in the disease. METHODS Bioinformatic deconvolution was done on bulk microarrays from carotid plaques in the Biobank of Karolinska Endarterectomies (BiKE, n = 125) using public plaque scRNAseq data and associated with patient clinical data and follow-up information. BiKE patients were clustered based on the deconvoluted cell fractions. Quantitative trait loci (QTLs) analyses were performed to predict the effect of CAD associated genetic variants on mesenchymal cell fractions (cfQTLs) and gene expression (eQTLs) in plaques. RESULTS Lesions from symptomatic patients had higher fractions of Type 1 macrophages and pericytes, but lower fractions of classical and modulated SMCs compared with asymptomatic ones, particularly females. Presence of diabetes or statin treatment did not affect the cell fraction distribution. Clustering based on plaque cell fractions, revealed three patient groups, with relative differences in their stability profiles and associations to stroke, even during long-term follow-up. Several single nucleotide polymorphisms associated with plaque mesenchymal cell fractions, upstream of the circadian rhythm gene ARNTL were identified. In vitro silencing of ARNTL in human carotid SMCs increased the expression of contractile markers and attenuated cell proliferation. CONCLUSIONS This study shows the potential of combining scRNAseq data with vertically integrated clinical, genetic, and transcriptomic data from a large biobank of human plaques, for refinement of patient vulnerability and risk prediction stratification. The study revealed novel CAD-associated variants that may be functionally linked to SMCs in atherosclerotic plaques. Specifically, variants in the ARNTL gene may influence SMC ratios and function, and its role as a regulator of SMC proliferation should be further investigated.
Collapse
Affiliation(s)
- Sampath Narayanan
- Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska University Hospital and Karolinska Institutet, BioClinicum J8:20, Visionsgatan 4, SE-171 76 Stockholm, Sweden
| | - Sofija Vuckovic
- Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska University Hospital and Karolinska Institutet, BioClinicum J8:20, Visionsgatan 4, SE-171 76 Stockholm, Sweden
| | - Otto Bergman
- Division of Cardiovascular Medicine, Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Karolinska University Hospital, Solna, Sweden
| | - Robert Wirka
- Department of Medicine and Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | | | - Qiao Sen Chen
- Division of Cardiovascular Medicine, Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Karolinska University Hospital, Solna, Sweden
| | - Damiano Baldassarre
- Centro Cardiologico Monzino, IRCCS, Milan, Italy
- Department of Medical Biotechnology and Translational Medicine, Università di Milano, Milan, Italy
| | - Elena Tremoli
- Maria Cecilia Hospital, GVM Care & Research, Cotignola, Italy
| | - Fabrizio Veglia
- Maria Cecilia Hospital, GVM Care & Research, Cotignola, Italy
| | - Mariette Lengquist
- Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska University Hospital and Karolinska Institutet, BioClinicum J8:20, Visionsgatan 4, SE-171 76 Stockholm, Sweden
| | - Redouane Aherrahrou
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
- Institute for Cardiogenetics, Universität zu Lübeck; DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Germany
- University Heart Centre Lübeck, Lübeck, Germany
| | - Anton Razuvaev
- Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska University Hospital and Karolinska Institutet, BioClinicum J8:20, Visionsgatan 4, SE-171 76 Stockholm, Sweden
| | - Bruna Gigante
- Division of Cardiovascular Medicine, Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Karolinska University Hospital, Solna, Sweden
| | - Hanna M Björck
- Division of Cardiovascular Medicine, Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Karolinska University Hospital, Solna, Sweden
| | - Clint L Miller
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Thomas Quertermous
- Division of Cardiovascular Medicine, Stanford University, Stanford, CA, USA
| | - Ulf Hedin
- Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska University Hospital and Karolinska Institutet, BioClinicum J8:20, Visionsgatan 4, SE-171 76 Stockholm, Sweden
| | - Ljubica Matic
- Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska University Hospital and Karolinska Institutet, BioClinicum J8:20, Visionsgatan 4, SE-171 76 Stockholm, Sweden
| |
Collapse
|
19
|
Regan JA, Kwee LC, Nafissi NA, Bick AG, Kraus WE, Natarajan P, Jaiswal S, Shah SH. Clonal Hematopoiesis Associates with Prevalent and Incident Cardiometabolic Disease in High-Risk Individuals. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.01.14.25320566. [PMID: 39867361 PMCID: PMC11759844 DOI: 10.1101/2025.01.14.25320566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Background Clonal hematopoiesis of indeterminate potential (CHIP) is the age-related presence of expanded somatic clones secondary to leukemogenic driver mutations and is associated with cardiovascular (CV) disease and mortality. We sought to evaluate relationships between CHIP with cardiometabolic diseases and incident outcomes in high-risk individuals. Methods CHIP genotyping was performed in 8469 individuals referred for cardiac catheterization at Duke University (CATHGEN study) to identify variants present at a variant allele fraction (VAF) ≥2%. Associations were tested among any CHIP variant, large CHIP clones (VAF ≥10%) and individual CHIP genes with prevalent cardiometabolic traits. Cox proportional hazard models tested CHIP associations with time-to-overall mortality and Fine-Gray analyses tested CHIP associations with incident cardiovascular outcomes. Results We identified 463 CHIP variants in 427 individuals (5.0%) of which 268 (3.2%) harbored large CHIP clones. CHIP and large CHIP were associated with lower odds of obesity (OR 0.79 [95% CI 0.65-0.98], p=0.03; OR 0.76 [95% CI 0.57-0.99], p=0.04, respectively). CHIP was associated with prevalent HF (OR 1.25 [95% CI 1.01 - 1.55], p=0.04; especially for non-DNMT3A CHIP (OR 1.38 [95% CI 1.04-1.82], p=0.02). CHIP was also associated with incident events: Non-DNMT3A CHIP was associated with increased risk of time-to-HF hospitalization (HR 1.29 [95% CI 1.02-1.63], p=0.03). Conclusions In high-risk individuals referred for cardiac catheterization, large CHIP and non-DNTM3A CHIP were associated with obesity, prevalent HF, incident CV events. These findings strengthen the importance of CHIP as a biomarker for CV disease and highlight the contributing risk of large CHIP clones and non-DNMT3A CHIP variants.
Collapse
Affiliation(s)
- Jessica A Regan
- Division of Cardiology, Department of Medicine, Duke University, Durham, NC, USA
- Duke Molecular Physiology Institute, Durham, NC, USA
| | | | - Navid A Nafissi
- Division of Cardiology, Department of Medicine, Duke University, Durham, NC, USA
- Duke Molecular Physiology Institute, Durham, NC, USA
| | - Alexander G Bick
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - William E Kraus
- Division of Cardiology, Department of Medicine, Duke University, Durham, NC, USA
- Duke Molecular Physiology Institute, Durham, NC, USA
| | - Pradeep Natarajan
- Center for Genomic Medicine and Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA
- Program in Medical & Population Genetics, Broad Institute of Harvard & MIT, Cambridge, MA
- Department of Medicine, Harvard Medical School, Boston, MA
| | - Sidd Jaiswal
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Svati H Shah
- Division of Cardiology, Department of Medicine, Duke University, Durham, NC, USA
- Duke Molecular Physiology Institute, Durham, NC, USA
| |
Collapse
|
20
|
Playford E, Stewart S, Hoyne G, Strange G, Dwivedi G, Hamilton-Craig C, Figtree G, Playford D. Comparing predictive risk to actual presence of coronary atherosclerosis on coronary computed tomography angiography. AMERICAN HEART JOURNAL PLUS : CARDIOLOGY RESEARCH AND PRACTICE 2025; 49:100493. [PMID: 39760104 PMCID: PMC11698937 DOI: 10.1016/j.ahjo.2024.100493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Accepted: 11/25/2024] [Indexed: 01/07/2025]
Abstract
Background There is limited data showing the predictive accuracy of traditional cardiovascular risk scores (CVRS) to predict asymptomatic coronary artery disease (CAD) determined by coronary computed tomography angiography (CCTA). Methods Asymptomatic individuals without known CAD undergoing a screening CCTA and sufficient data to calculate their CVRS, were extracted retrospectively. Atherosclerosis was extracted using natural language processing of the CCTA report, including the coronary artery calcium score (CACS) and the extent and severity of CAD. Absence of atherosclerosis was defined as both zero plaque and zero CACS, and atherosclerosis was defined as low, moderate, or extensive by location and extent of plaque-burden. CVRS was categorized as high (>15 %), moderate (10-15 %), low (1-9 %) and "zero" (<1 %) risk. Results 828 individuals (median age 58.6, IQR = 52.0, 65.3 years, 57 % male) met inclusion criteria, and a zero, low, moderate, and high CVRS was identified in 13, 483, 113 and 219 individuals (8 %, 49 %, 74 %, 66 % male), respectively. Predominantly low plaque-burden atherosclerosis was detected in 548 scans (67 % male). However, of the 137 males and 68 females with extensive atherosclerosis, 47 (34 %) and 38 (56 %) respectively had low CVRS classification. Overall, 23 % of males and 31 % of females had CAD predicted by CVRS (Monte Carlo: females, p = 0.024; males, p < 0.001), but there was little to no agreement between CVRS and atherosclerosis burden (Cohen's kappa: males, κ = 0.149; females, κ = 0.096). Conclusions In asymptomatic individuals without known CAD, a low CVRS does not exclude extensive CAD. Newer tools incorporating additional markers may be helpful in risk prediction in such individuals.
Collapse
Affiliation(s)
- Emma Playford
- The University of Notre Dame Australia, Fremantle, WA, Australia
| | - Simon Stewart
- The University of Notre Dame Australia, Fremantle, WA, Australia
| | - Gerard Hoyne
- The University of Notre Dame Australia, Fremantle, WA, Australia
- Institute for Respiratory Health, QEII Medical Centre, Nedlands, WA, Australia
| | - Geoff Strange
- The University of Notre Dame Australia, Fremantle, WA, Australia
| | - Girish Dwivedi
- Harry Perkins Institute for Medical Research, University of Western Australia, Australia
- Fiona Stanley Hospital, Perth, Australia
| | - Christian Hamilton-Craig
- The Prince Charles Hospital, Brisbane, Australia
- Royal North Shore Hospital, Sydney, New South Wales, Australia
| | - Gemma Figtree
- Kolling Institute, University of Sydney, Sydney, New South Wales, Australia
| | - David Playford
- The University of Notre Dame Australia, Fremantle, WA, Australia
| |
Collapse
|
21
|
Benn M, Emanuelsson F, Tybjærg-Hansen A, Nordestgaard BG. Low LDL cholesterol and risk of bacterial and viral infections: observational and Mendelian randomization studies. EUROPEAN HEART JOURNAL OPEN 2025; 5:oeaf009. [PMID: 39991120 PMCID: PMC11843444 DOI: 10.1093/ehjopen/oeaf009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/13/2025] [Accepted: 01/31/2025] [Indexed: 02/25/2025]
Abstract
Aims Low levels of LDL cholesterol may be associated with risk of infectious disease. We tested the hypothesis that low LDL cholesterol due to genetic variation in the LDLR, PCSK9, and HMGCR genes and a polygenic LDL cholesterol score is associated with risk of infectious diseases in the general population. Methods and results Using observational and Mendelian randomization designs, we examined associations of low plasma LDL cholesterol with risk of bacterial and viral infections in 119 805 individuals from the Copenhagen General Population Study/Copenhagen City Heart Study, 468 701 from the UK Biobank, and up to 376 773 from the FinnGen Research Project. Observationally, low LDL cholesterol concentrations were associated with risk of hospitalization for both bacterial and viral infections. In genetic analyses, a 1 mmol/L lower LDL cholesterol was associated with lower plasma PCSK9 {-0.55 nmol/L [95% confidence interval (CI): -1.06 to -0.05]; P = 0.03}, leucocyte count [-0.42 × 109/L (-0.61 to -0.24); P < 0.001], and high-sensitivity C-reactive protein [-0.44 mg/L (-0.79 to -0.09); P = 0.014]. Using an LDLR, HMGCR, and PCSK9 score, a 1 mmol/L lower LDL cholesterol was associated with risk ratios of 0.91 (95% CI: 0.86-0.97; P = 0.002) for unspecified bacterial infection, of 0.92 (0.87-0.97; P = 0.004) for diarrhoeal disease, and of 1.15 (1.03-1.29; P = 0.012) for unspecified viral infections and 1.64 (1.13-2.39; P = 0.009) for HIV/AIDS. Using a polygenic LDL cholesterol score largely showed similar results and in addition a lower risk of 0.85 (0.76-0.96; P = 0.006) for bacterial pneumonia and 0.91 (0.82-0.99; P = 0.035) for sepsis. Conclusion Genetically low LDL cholesterol concentrations were associated with lower concentration of markers of inflammation; lower risk of hospitalization for unspecified bacterial infections, infectious diarrhoeal diseases, bacterial pneumonia, and sepsis; and higher risk of viral infections and HIV/AIDS.
Collapse
Affiliation(s)
- Marianne Benn
- Department of Clinical Biochemistry, Copenhagen University Hospital—Rigshospitalet, Blegdamsvej 9, DK-2100 Copenhagen, Denmark
- The Copenhagen General Population Study, Copenhagen University Hospital—Herlev and Gentofte, Borgmester Ib Juuls vej 1, DK-2730 Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen, Denmark
| | - Frida Emanuelsson
- Department of Clinical Biochemistry, Copenhagen University Hospital—Rigshospitalet, Blegdamsvej 9, DK-2100 Copenhagen, Denmark
| | - Anne Tybjærg-Hansen
- Department of Clinical Biochemistry, Copenhagen University Hospital—Rigshospitalet, Blegdamsvej 9, DK-2100 Copenhagen, Denmark
- The Copenhagen General Population Study, Copenhagen University Hospital—Herlev and Gentofte, Borgmester Ib Juuls vej 1, DK-2730 Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen, Denmark
- The Copenhagen City Heart Study, Copenhagen University Hospital—Bispebjerg and Frederiksberg, Bispebjerg Bakke 23, DK-2400 Copenhagen, Denmark
| | - Børge G Nordestgaard
- The Copenhagen General Population Study, Copenhagen University Hospital—Herlev and Gentofte, Borgmester Ib Juuls vej 1, DK-2730 Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen, Denmark
- The Copenhagen City Heart Study, Copenhagen University Hospital—Bispebjerg and Frederiksberg, Bispebjerg Bakke 23, DK-2400 Copenhagen, Denmark
- Department of Clinical Biochemistry, Copenhagen University Hospital—Herlev and Gentofte, Borgmester Ib Juuls vej 1, DK-2730 Herlev, Denmark
| |
Collapse
|
22
|
Yao L, Chen T. A combined association of alanine aminotransferase, aspartate transaminase and bilirubin with sleep duration in aged 16-85 years (2005-2010). Medicine (Baltimore) 2024; 103:e40915. [PMID: 39654161 PMCID: PMC11630931 DOI: 10.1097/md.0000000000040915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 11/13/2024] [Accepted: 11/22/2024] [Indexed: 12/12/2024] Open
Abstract
Sleep is a vital restorative process that plays a pivotal role in maintaining the delicate equilibrium of mental and physical well-being. Both short and long sleep duration are associated with a range of adverse health outcomes. Numerous studies have consistently demonstrated a robust association between sleep duration and liver disease. In this study, we conducted statistical tests and performed subgroup analyses to explore potential variations in this association across different contexts, aiming to elucidate the correlation between ALT, AST, and TB with sleep duration. This cross-sectional investigation utilized datasets from the National Health and Nutrition Examination Survey 2005 to 2010. Multivariate linear regression models were used to examine the linear association between ALT, AST, and TB with sleep duration. Test for interaction is commonly conducted using multivariabte models to assess statistically significant subgroup disparities. Fitted smoothied curves and threshold effect analyses were employed to depict nonlinear relationships. The study enrolled 17,491 participants aged 16 to 85 years who met the inclusion and exclusion criteria, with a mean age of the participants was 45.58 ± 19.94 years. Multivariate linear regression analysis showed a significant positive association between sleep duration and ALT [-0.23 (-0.45, -0.00) 0.0455] and AST[-0.20 (-0.38, -0.01) 0.0338] in Model 3. Using a two-segment linear regression model, we found an U-shaped relationship and significant inflection point between between ALT and AST with sleep duration. The present study unveiled a significant inverse correlation between sleep duration and levels of ALT and AST, while no significant association was observed with TB levels. Furthermore, variations in the optimal sleep duration for liver function recovery were identified across diverse populations, thereby offering valuable healthcare recommendations to public.
Collapse
Affiliation(s)
- Lishuai Yao
- Department of Cardiothoracic Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Guangzhou, China
| | - Tiantian Chen
- Department of Cardiothoracic Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
23
|
Nicholls SJ, Nelson AJ, Michael LF. Oral agents for lowering lipoprotein(a). Curr Opin Lipidol 2024; 35:275-280. [PMID: 39329200 DOI: 10.1097/mol.0000000000000953] [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: 09/28/2024]
Abstract
PURPOSE OF REVIEW To review the development of oral agents to lower Lp(a) levels as an approach to reducing cardiovascular risk, with a focus on recent advances in the field. RECENT FINDINGS Extensive evidence implicates Lp(a) in the causal pathway of atherosclerotic cardiovascular disease and calcific aortic stenosis. There are currently no therapies approved for lowering of Lp(a). The majority of recent therapeutic advances have focused on development of injectable agents that target RNA and inhibit synthesis of apo(a). Muvalaplin is the first, orally administered, small molecule inhibitor of Lp(a), which acts by disrupting binding of apo(a) and apoB, in clinical development. Nonhuman primate and early human studies have demonstrated the ability of muvalaplin to produce dose-dependent lowering of Lp(a). Ongoing clinical trials will evaluate the impact of muvalaplin in high cardiovascular risk and will ultimately need to determine whether this strategy lowers the rate of cardiovascular events. SUMMARY Muvalaplin is the first oral agent, developed to lower Lp(a) levels. The ability of muvalaplin to reduce cardiovascular risk remains to be investigated, in order to determine whether it will be a useful agent for the prevention of cardiovascular disease.
Collapse
Affiliation(s)
- Stephen J Nicholls
- From the Victorian Heart Institute, Monash University, Melbourne, Australia
| | - Adam J Nelson
- From the Victorian Heart Institute, Monash University, Melbourne, Australia
| | | |
Collapse
|
24
|
Iribarren C, Lu M, Gulati M, Wong ND, Elosua R, Rana JS. Interplay between lifestyle factors and polygenic risk for incident coronary heart disease in a large multiethnic cohort. INTERNATIONAL JOURNAL OF CARDIOLOGY. CARDIOVASCULAR RISK AND PREVENTION 2024; 23:200350. [PMID: 39582945 PMCID: PMC11584587 DOI: 10.1016/j.ijcrp.2024.200350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 08/16/2024] [Accepted: 10/31/2024] [Indexed: 11/26/2024]
Abstract
Introduction The objective of this study was to examine the interplay of polygenic risk and individual lifestyle factors (and a composite score of lifestyle) as antecedents of CHD in a large multiethnic cohort. Methods We used Genetic Epidemiology Resource in Adult Health and Aging (GERA) cohort participants free of CHD at baseline (n = 60,568; 67 % female; 18 % non-European). The individual and joint associations of smoking, Mediterranean diet pattern, level of physical activity and polygenic risk with incident CHD were assessed using Cox regression adjusting for genetic ancestry and non-mediating risk factors. Hazard ratios (HRs) and number needed to treat (NNT) were estimated according to these lifestyle factors and polygenic risk categories. Strengths included large sample size, long-follow-up, ethnic diversity, a clinically-validated polygenic risk score (PRS), and rich phenotype information. Results After 14 years of follow-up, there were 3159 incident CHD events. We observed no statistically significant interactions between individual lifestyle factors and polygenic risk (all p > 0.23). For individuals with a high genetic risk, moving from the worse lifestyle combination (no favorable lifestyle factors) to the best lifestyle combination (all three) is associated with 52 % lower rate of CHD. The NNT was highest in the low polygenic risk group (34), lowest in the high polygenic risk group [19] and in-between (Jin et al., 2011) [24] in the intermediate polygenic risk group. Conclusions Lifestyle and polygenic risk together influence the risk of incident CHD. Our results support consideration of polygenic risk in lifestyle interventions because those with high polygenic risk are likely to derive the most benefit.
Collapse
Affiliation(s)
- Carlos Iribarren
- Kaiser Permanente Northern California Division of Research, Oakland, CA, USA
| | - Meng Lu
- Kaiser Permanente Northern California Division of Research, Oakland, CA, USA
| | - Martha Gulati
- Barbra Streisand Women's Heart Center, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Nathan D. Wong
- Heart Disease Prevention Program, Division of Cardiology, Department of Medicine, University of California Irvine, Irvine, CA, USA
| | - Roberto Elosua
- Cardiovascular Epidemiology and Genetics, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), CIBER Cardiovascular Diseases (CIBERCV), Barcelona, Faculty of Medicine, University of Vic-Central University of Catalonia (UVic-UCC), Vic, Spain
| | - Jamal S. Rana
- Kaiser Permanente Northern California Division of Research, Oakland, CA, USA
- Department of Cardiology, The Permanente Medical Group, Kaiser Permanente Oakland Medical Center, Oakland, CA, USA
| |
Collapse
|
25
|
Lin A, Miano JM, Fisher EA, Misra A. Chronic inflammation and vascular cell plasticity in atherosclerosis. NATURE CARDIOVASCULAR RESEARCH 2024; 3:1408-1423. [PMID: 39653823 DOI: 10.1038/s44161-024-00569-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 10/10/2024] [Indexed: 12/13/2024]
Abstract
Vascular smooth muscle cells, endothelial cells and macrophages undergo phenotypic conversions throughout atherosclerosis progression, both as a consequence of chronic inflammation and as subsequent drivers of it. The inflammatory hypothesis of atherosclerosis has been catapulted to the forefront of cardiovascular research as clinical trials have shown that anti-inflammatory therapy reduces adverse cardiovascular events. However, no current therapies have been specifically designed to target the phenotype of plaque cells. Fate mapping has revealed that plaque cells convert to detrimental and beneficial cell phenotypes during atherosclerosis, with cumulative evidence highlighting that vascular cell plasticity is intimately linked with plaque inflammation, ultimately impacting lesion stability. Here we review vascular cell plasticity during atherosclerosis in the context of the chronic inflammatory plaque microenvironment. We highlight the need to better understand how plaque cells behave during therapeutic intervention. We then propose modulating plaque cell phenotype as an unexplored therapeutic paradigm in the clinical setting.
Collapse
Affiliation(s)
- Alexander Lin
- Atherosclerosis and Vascular Remodelling Group, Heart Research Institute, Sydney, New South Wales, Australia
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Sydney, New South Wales, Australia
| | - Joseph M Miano
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Edward A Fisher
- Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
- Cardiovascular Research Center, New York University Grossman School of Medicine, New York, NY, USA
| | - Ashish Misra
- Atherosclerosis and Vascular Remodelling Group, Heart Research Institute, Sydney, New South Wales, Australia.
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.
| |
Collapse
|
26
|
Jiang JC, Singh K, Nitin R, Davis LK, Wray NR, Shah S. Sex-Specific Association Between Genetic Risk of Psychiatric Disorders and Cardiovascular Diseases. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2024; 17:e004685. [PMID: 39611256 PMCID: PMC11651350 DOI: 10.1161/circgen.124.004685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 10/15/2024] [Indexed: 11/30/2024]
Abstract
BACKGROUND Though epidemiological studies show increased cardiovascular disease (CVD) risks among individuals with psychiatric disorders, findings on sex differences in comorbidity have been inconsistent. METHODS This genetic epidemiology study examined the sex-specific association between the genetic risk of 3 psychiatric disorders (major depression [MD], schizophrenia, and bipolar disorder), estimated using polygenic scores (PGSs), and risks of 3 CVDs (atrial fibrillation [AF], coronary artery disease [CAD], and heart failure [HF]) in 345 169 European-ancestry individuals (UK Biobank), with analyses replicated in an independent BioVU cohort (n=49 057). Mediation analysis was conducted to determine whether traditional CVD risk factors could explain any observed sex difference. RESULTS In the UK Biobank, a 1-SD increase in PGSMD was significantly associated with the incident risks of all 3 CVDs in females after multiple testing corrections (hazard ratio [HR]AF-female=1.04 [95% CI, 1.02-1.06]; P=1.5×10-4; HRCAD-female=1.07 [95% CI, 1.04-1.11]; P=2.6×10-6; and HRHF-female=1.09 [95% CI, 1.06-1.13]; P=9.7×10-10), but not in males. These female-specific associations remained even in the absence of any psychiatric disorder diagnosis or psychiatric medication use. Although mediation analysis demonstrated that the association between PGSMD and CVDs in females was partly mediated by baseline body mass index, hypercholesterolemia, hypertension, and smoking, these risk factors did not explain the higher risk compared with males. The association between PGSMD and CAD was consistent between females who were premenopausal and postmenopausal at baseline, while the association with AF and HF was only observed in the baseline postmenopausal cohort. No significant association with CVD risks was observed for the PGS of schizophrenia or bipolar disorder. The female-specific positive association of PGSMD with CAD risk was replicated in BioVU. CONCLUSIONS Genetic predisposition to MD confers a greater risk of CVDs in females versus males, even in the absence of any depression diagnosis. This study warrants further investigation into whether genetic predisposition to depression could be useful for improving cardiovascular risk prediction, especially in women.
Collapse
Affiliation(s)
- Jiayue-Clara Jiang
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Australia (J.-C.J., N.R.W., S.S.)
| | - Kritika Singh
- Division of Genetic Medicine, Department of Medicine (K.S., R.N., L.K.D.), Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Genetics Institute (K.S., R.N., L.K.D.), Vanderbilt University Medical Center, Nashville, TN
| | - Rachana Nitin
- Division of Genetic Medicine, Department of Medicine (K.S., R.N., L.K.D.), Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Genetics Institute (K.S., R.N., L.K.D.), Vanderbilt University Medical Center, Nashville, TN
| | - Lea K. Davis
- Division of Genetic Medicine, Department of Medicine (K.S., R.N., L.K.D.), Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Genetics Institute (K.S., R.N., L.K.D.), Vanderbilt University Medical Center, Nashville, TN
- Department of Molecular Physiology and Biophysics (L.K.D.), Vanderbilt University Medical Center, Nashville, TN
- Department of Psychiatry and Behavioral Sciences (L.K.D.), Vanderbilt University Medical Center, Nashville, TN
- Departments of Medicine and Biomedical Informatics (L.K.D.), Vanderbilt University Medical Center, Nashville, TN
| | - Naomi R. Wray
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Australia (J.-C.J., N.R.W., S.S.)
- Department of Psychiatry, University of Oxford, Warneford Hospital, United Kingdom (N.R.W.)
| | - Sonia Shah
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Australia (J.-C.J., N.R.W., S.S.)
| |
Collapse
|
27
|
Rudman-Melnick V, Vanhoutte D, Stowers K, Sargent M, Adam M, Ma Q, Perl AKT, Miethke AG, Burg A, Shi T, Hildeman DA, Woodle ESS, Kofron JM, Devarajan P. Gucy1α1 specifically marks kidney, heart, lung and liver fibroblasts. Sci Rep 2024; 14:29307. [PMID: 39592775 PMCID: PMC11599588 DOI: 10.1038/s41598-024-80930-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 11/22/2024] [Indexed: 11/28/2024] Open
Abstract
Fibrosis is a common outcome of numerous pathologies, including chronic kidney disease (CKD), a progressive renal function deterioration. Current approaches to target activated fibroblasts, key effector contributors to fibrotic tissue remodeling, lack specificity. Here, we report Gucy1α1 as a specific kidney fibroblast marker. Gucy1α1 levels significantly increased over the course of two clinically relevant murine CKD models and directly correlated with established fibrosis markers. Immunofluorescent (IF) imaging showed that Gucy1α1 comprehensively labelled cortical and medullary quiescent and activated fibroblasts in the control kidney and throughout injury progression, respectively. Unlike traditionally used markers platelet derived growth factor receptor beta (Pdgfrβ) and vimentin (Vim), Gucy1α1 did not overlap with off-target populations such as podocytes. Notably, Gucy1α1 labelled kidney fibroblasts in both male and female mice. Furthermore, we observed elevated GUCY1α1 expression in the human fibrotic kidney and lung. Studies in the murine models of cardiac and liver fibrosis revealed Gucy1α1 elevation in activated Pdgfrβ-, Vim- and alpha smooth muscle actin (αSma)-expressing fibroblasts paralleling injury progression and resolution. Overall, we demonstrate Gucy1α1 as an exclusive fibroblast marker in both sexes. Due to its multiorgan translational potential, GUCY1α1 might provide a novel promising strategy to specifically target and mechanistically examine fibroblasts.
Collapse
Affiliation(s)
- Valeria Rudman-Melnick
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, 268-280 Albert Sabin Way, location T, floor 6, suite 272, Cincinnati, OH, 45229, USA
| | - Davy Vanhoutte
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Kaitlynn Stowers
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, 268-280 Albert Sabin Way, location T, floor 6, suite 272, Cincinnati, OH, 45229, USA
| | - Michelle Sargent
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Mike Adam
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Qing Ma
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, 268-280 Albert Sabin Way, location T, floor 6, suite 272, Cincinnati, OH, 45229, USA
| | - Anne Karina T Perl
- Division of Neonatology and Pulmonary biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Alexander G Miethke
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Ashley Burg
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Tiffany Shi
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - David A Hildeman
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - E Steve S Woodle
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - J Matthew Kofron
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Prasad Devarajan
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, 268-280 Albert Sabin Way, location T, floor 6, suite 272, Cincinnati, OH, 45229, USA.
| |
Collapse
|
28
|
Mahmoud AK, Farina JM, Awad K, Ali NB, Pereyra M, Scalia IG, Abbas MT, Allam MN, Kamel MA, Abu Rmilah AA, Chao CJ, Barry T, Alsidawi S, Lester SJ, Pollak PM, Alkhouli MA, Lee KS, Yang EH, Lee RW, Sweeney JP, Fortuin DF, Ayoub C, Arsanjani R. Lipoprotein(a) and long-term in-stent restenosis after percutaneous coronary intervention. Eur J Prev Cardiol 2024; 31:1878-1887. [PMID: 38916491 DOI: 10.1093/eurjpc/zwae212] [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: 04/03/2024] [Revised: 05/15/2024] [Accepted: 06/18/2024] [Indexed: 06/26/2024]
Abstract
AIMS Lipoprotein(a) [Lp(a)] has demonstrated its association with atherosclerosis and myocardial infarction. However, its role in the development of in-stent restenosis (ISR) after percutaneous coronary intervention (PCI) is not clearly established. The aim of this study is to investigate the association between Lp(a) and ISR. METHODS AND RESULTS A retrospective study of adult patients who underwent successful PCI between January 2006 and December 2017 at the three Mayo Clinic sites and had a preprocedural Lp(a) measurement was conducted. Patients were divided into two groups according to the serum Lp(a) concentration [high Lp(a) ≥ 50 mg/dL and low Lp(a) < 50 mg/dL]. Univariable and multivariable analyses were performed to compare risk of ISR between patients with high Lp(a) vs. those with low Lp(a). A total of 1209 patients were included, with mean age 65.9 ± 11.7 years and 71.8% were male. Median follow-up after baseline PCI was 8.8 [interquartile range (IQR) 7.4] years. Restenosis was observed in 162 (13.4%) patients. Median serum levels of Lp(a) were significantly higher in patients affected by ISR vs. non-affected cases: 27 (IQR 73.8) vs. 20 (IQR 57.5) mg/dL, P = 0.008. The rate of ISR was significantly higher among patients with high Lp(a) vs. patients with low Lp(a) values (17.0% vs. 11.6%, P = 0.010). High Lp(a) values were independently associated with ISR events (hazard ratio 1.67, 95% confidence interval 1.18-2.37, P = 0.004), and this association was more prominent after the first year following the PCI. CONCLUSION Lipoprotein(a) is an independent predictor for long-term ISR and should be considered in the evaluation of patients undergoing PCI.
Collapse
Affiliation(s)
- Ahmed K Mahmoud
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 E Mayo Blvd, Phoenix, AZ 85054, USA
| | - Juan M Farina
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 E Mayo Blvd, Phoenix, AZ 85054, USA
| | - Kamal Awad
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 E Mayo Blvd, Phoenix, AZ 85054, USA
| | - Nima Baba Ali
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 E Mayo Blvd, Phoenix, AZ 85054, USA
| | - Milagros Pereyra
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 E Mayo Blvd, Phoenix, AZ 85054, USA
| | - Isabel G Scalia
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 E Mayo Blvd, Phoenix, AZ 85054, USA
| | - Mohammed Tiseer Abbas
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 E Mayo Blvd, Phoenix, AZ 85054, USA
| | - Mohamed N Allam
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 E Mayo Blvd, Phoenix, AZ 85054, USA
| | - Moaz A Kamel
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 E Mayo Blvd, Phoenix, AZ 85054, USA
| | - Anan A Abu Rmilah
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Chieh-Ju Chao
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Timothy Barry
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 E Mayo Blvd, Phoenix, AZ 85054, USA
| | - Said Alsidawi
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 E Mayo Blvd, Phoenix, AZ 85054, USA
| | - Steven J Lester
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 E Mayo Blvd, Phoenix, AZ 85054, USA
| | - Peter M Pollak
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
| | | | - Kwan S Lee
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 E Mayo Blvd, Phoenix, AZ 85054, USA
| | - Eric H Yang
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 E Mayo Blvd, Phoenix, AZ 85054, USA
| | - Richard W Lee
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 E Mayo Blvd, Phoenix, AZ 85054, USA
| | - John P Sweeney
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 E Mayo Blvd, Phoenix, AZ 85054, USA
| | - David F Fortuin
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 E Mayo Blvd, Phoenix, AZ 85054, USA
| | - Chadi Ayoub
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 E Mayo Blvd, Phoenix, AZ 85054, USA
| | - Reza Arsanjani
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 E Mayo Blvd, Phoenix, AZ 85054, USA
| |
Collapse
|
29
|
Nikolić S, Ignatov DI, Khvorykh GV, Limborska SA, Khrunin AV. Genome-wide association studies of ischemic stroke based on interpretable machine learning. PeerJ Comput Sci 2024; 10:e2454. [PMID: 39650478 PMCID: PMC11623107 DOI: 10.7717/peerj-cs.2454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 10/04/2024] [Indexed: 12/11/2024]
Abstract
Despite the identification of several dozen genetic loci associated with ischemic stroke (IS), the genetic bases of this disease remain largely unexplored. In this research we present the results of genome-wide association studies (GWAS) based on classical statistical testing and machine learning algorithms (logistic regression, gradient boosting on decision trees, and tabular deep learning model TabNet). To build a consensus on the results obtained by different techniques, the Pareto-Optimal solution was proposed and applied. These methods were applied to real genotypic data of sick and healthy individuals of European ancestry obtained from the Database of Genotypes and Phenotypes (5,581 individuals, 883,749 single nucleotide polymorphisms). Finally, 131 genes were identified as candidates for association with the onset of IS. UBQLN1, TRPS1, and MUSK were previously described as associated with the course of IS in model animals. ACOT11 taking part in metabolism of fatty acids was shown for the first time to be associated with IS. The identified genes were compared with genes from the Illuminating Druggable Genome project. The product of GPR26 representing the G-coupled protein receptor can be considered as a therapeutic target for stroke prevention. The approaches presented in this research can be used to reprocess GWAS datasets from other diseases.
Collapse
Affiliation(s)
- Stefan Nikolić
- Laboratory for Models and Methods of Computational Pragmatics; Department of Data Analysis and Artificial Intelligence, HSE University, Moscow, Russia
| | - Dmitry I. Ignatov
- Laboratory for Models and Methods of Computational Pragmatics; Department of Data Analysis and Artificial Intelligence, HSE University, Moscow, Russia
| | | | | | | |
Collapse
|
30
|
Aherrahrou R, Reinberger T, Hashmi S, Erdmann J. GWAS breakthroughs: mapping the journey from one locus to 393 significant coronary artery disease associations. Cardiovasc Res 2024; 120:1508-1530. [PMID: 39073758 DOI: 10.1093/cvr/cvae161] [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: 01/27/2024] [Revised: 03/20/2024] [Accepted: 06/12/2024] [Indexed: 07/30/2024] Open
Abstract
Coronary artery disease (CAD) poses a substantial threat to global health, leading to significant morbidity and mortality worldwide. It has a significant genetic component that has been studied through genome-wide association studies (GWAS) over the past 17 years. These studies have made progress with larger sample sizes, diverse ancestral backgrounds, and the discovery of multiple genomic regions related to CAD risk. In this review, we provide a comprehensive overview of CAD GWAS, including information about the genetic makeup of the disease and the importance of ethnic diversity in these studies. We also discuss challenges of identifying causal genes and variants within GWAS loci with a focus on non-coding regions. Additionally, we highlight tissues and cell types relevant to CAD, and discuss clinical implications of GWAS findings including polygenic risk scores, sex-specific differences in CAD genetics, ethnical aspects of personalized interventions, and GWAS guided drug development.
Collapse
Affiliation(s)
- Rédouane Aherrahrou
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211 Kuopio, Finland
- Institute for Cardiogenetics, University of Lübeck, Marie-Curie-Str. Haus 67/BMF, 23562 Lübeck, Germany
- DZHK (German Centre for Cardiovascular Research), Institute for Cardiogenetics, Universität zu Lübeck, Partner Site Hamburg/Kiel/Lübeck, Germany
- University Heart Centre Lübeck, University Hospital Schleswig-Holstein, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Tobias Reinberger
- Institute for Cardiogenetics, University of Lübeck, Marie-Curie-Str. Haus 67/BMF, 23562 Lübeck, Germany
- DZHK (German Centre for Cardiovascular Research), Institute for Cardiogenetics, Universität zu Lübeck, Partner Site Hamburg/Kiel/Lübeck, Germany
- University Heart Centre Lübeck, University Hospital Schleswig-Holstein, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Satwat Hashmi
- Department of Biological and Biomedical Sciences, Aga Khan University, Stadium Road, 74800 Karachi, Pakistan
| | - Jeanette Erdmann
- Institute for Cardiogenetics, University of Lübeck, Marie-Curie-Str. Haus 67/BMF, 23562 Lübeck, Germany
- DZHK (German Centre for Cardiovascular Research), Institute for Cardiogenetics, Universität zu Lübeck, Partner Site Hamburg/Kiel/Lübeck, Germany
- University Heart Centre Lübeck, University Hospital Schleswig-Holstein, Ratzeburger Allee 160, 23562 Lübeck, Germany
| |
Collapse
|
31
|
Li J, Wei J, Fu P, Gu J. Identification of novel proteins for coronary artery disease by integrating GWAS data and human plasma proteomes. Heliyon 2024; 10:e38036. [PMID: 39386869 PMCID: PMC11462259 DOI: 10.1016/j.heliyon.2024.e38036] [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: 01/04/2024] [Revised: 09/06/2024] [Accepted: 09/16/2024] [Indexed: 10/12/2024] Open
Abstract
Background Most coronary artery disease (CAD) risk loci identified by genome-wide association studies (GWAS) are located in non-coding regions, hampering the interpretation of how they confer CAD risk. It is essential to integrate GWAS with molecular traits data to further explore the genetic basis of CAD. Methods We used the probabilistic Mendelian randomization (PMR) method to identify potential proteins involved in CAD by integrating CAD GWAS data (∼76,014 cases and ∼264,785 controls) and human plasma proteomes (N = 35,559). Then, Bayesian co-localization analysis, confirmatory PMR analysis using independent plasma proteome data (N = 7752), and gene expression data (N1 = 213, N2 = 670) were performed to validate candidate proteins. We further investigated the associations between candidate proteins and CAD-related traits and explored the rationality and biological functions of candidate proteins through disease enrichment, cell type-specific, GO, and KEGG enrichment analysis. Results This study inferred that the abundance of 30 proteins in the plasma was causally associated with CAD (P < 0.05/4408, Bonferroni correction), such as PLG, IL15RA, and CSNK2A1. PLG, PSCK9, COLEC11, ZNF180, ERP29, TCP1, FN1, CDH5, IL15RA, MGAT4B, TNFRSF6B, DNM2, and TGF1R were replicated in the confirmatory PMR (P < 0.05). PCSK9 (PP.H4 = 0.99), APOB (PP.H4 = 0.89), FN1 (PP.H4 = 0.87), and APOC1 (PP.H4 = 0.78) coding proteins shared one common variant with CAD. MTAP, TCP1, APOC2, ERP29, MORF4L1, C19orf80, PCSK9, APOC1, EPOR, DNM2, TNFRSF6B, CDKN2B, and LDLR were supported by PMR at the transcriptome level in whole blood and/or coronary arteries (P < 0.05). Enrichment analysis identified multiple pathways involved in cholesterol metabolism, regulation of lipoprotein levels and telomerase, such as cholesterol metabolism (hsa04979, P = 2.25E-7), plasma lipoprotein particle clearance (GO:0034381, P = 5.47E-5), and regulation of telomerase activity (GO:0051972, P = 2.34E-3). Conclusions Our integration analysis has identified 30 candidate proteins for CAD, which may provide important leads to design future functional studies and potential drug targets for CAD.
Collapse
Affiliation(s)
- Jiqing Li
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Shandong University, Jinan, 250012, Shandong, China
| | - Jiate Wei
- Office of Hospital Management Research, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Ping Fu
- Jinan Center for Disease Control and Prevention, Jinan, 250012, Shandong, China
| | - Jianhua Gu
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Shandong University, Jinan, 250012, Shandong, China
| |
Collapse
|
32
|
Al Hageh C, O'Sullivan S, Henschel A, Abchee A, Hantouche M, Iakovidou N, Issa T, Chacar S, Nader M, Zalloua PA. PHACTR1 and APOC1 genetic variants are associated with multi-vessel coronary artery disease. Lipids Health Dis 2024; 23:332. [PMID: 39395990 PMCID: PMC11471027 DOI: 10.1186/s12944-024-02327-2] [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: 07/09/2024] [Accepted: 10/07/2024] [Indexed: 10/14/2024] Open
Abstract
BACKGROUND Severe coronary artery disease (CAD) represents an advanced arterial narrowing, often associated with critical complications like myocardial infarction and angina. This study aimed to comprehensively investigate determinants of severe and multi-vessel CAD manifestations. METHODS One thousand nine hundred patients with severe and multivessel CAD (stenosis > 70%) were recruited along with 1,056 controls without stenosis. Associations using a genotyping panel comprising 159 Single Nucleotide Polymorphisms (SNPs) previously implicated in CAD pathogenesis were examined and these associations were replicated using the UK Biobank cohort (N = 29,970). RESULTS The investigation identified 14 genetic associations with severe CAD, of which 7 were also associated with multivessel disease. Notably, PHACTR1 SNP (rs9349379*G) showed a higher association with severe and multivessel CAD in individuals aged ≤ 65, indicating a higher risk of early disease onset. Conversely, the APOC1/APOE SNP (rs445925*T) is associated with reduced susceptibility to severe CAD and multivessel disease in individuals aged over 65, indicating a persistent negative association. CONCLUSIONS Following replication of the associations in the large UK Biobank dataset, it was found that patients carrying the rs9349379*G variant in the PHACTR1 gene are at risk of developing severe or multivessel disease. Conversely, the rs445925*T variant in APOC1/APOE is associated with reduced susceptibility to severe CAD and multivessel disease, highlighting the significance of this genetic variant in these specific CAD presentations. This study contributes to a better understanding of CAD heterogeneity, paving the way for tailored management strategies based on genetic profiles.
Collapse
Affiliation(s)
- Cynthia Al Hageh
- Department of Public Health and Epidemiology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Siobhán O'Sullivan
- Department of Biological Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Andreas Henschel
- Department of Computer Science, College of Computing and Mathematical Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Antoine Abchee
- Faculty of Medicine, University of Balamand, Balamand, Lebanon
| | - Mireille Hantouche
- Department of Public Health and Epidemiology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Nantia Iakovidou
- Department of Informatics, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Taly Issa
- University of Nicosia Medical School, Egkomi, Cyprus
| | - Stephanie Chacar
- Department of Medical Sciences, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, PO Box 127788, United Arab Emirates
| | - Moni Nader
- Department of Medical Sciences, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, PO Box 127788, United Arab Emirates.
| | - Pierre A Zalloua
- Department of Public Health and Epidemiology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates.
- Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
33
|
Tariq Z, Abusnana S, Mussa BM, Zakaria H. New insights on genetic background of major diabetic vascular complications. Diabetol Metab Syndr 2024; 16:243. [PMID: 39375805 PMCID: PMC11457557 DOI: 10.1186/s13098-024-01473-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 09/21/2024] [Indexed: 10/09/2024] Open
Abstract
BACKGROUND By 2045, it is expected that 693 million individuals worldwide will have diabetes and with greater risk of morbidity, mortality, loss of vision, renal failure, and a decreased quality of life due to the devastating effects of macro- and microvascular complications. As such, clinical variables and glycemic control alone cannot predict the onset of vascular problems. An increasing body of research points to the importance of genetic predisposition in the onset of both diabetes and diabetic vascular complications. OBJECTIVES Purpose of this article is to review these approaches and narrow down genetic findings for Diabetic Mellitus and its consequences, highlighting the gaps in the literature necessary to further genomic discovery. MATERIAL AND METHODS In the past, studies looking for genetic risk factors for diabetes complications relied on methods such as candidate gene studies, which were rife with false positives, and underpowered genome-wide association studies, which were constrained by small sample sizes. RESULTS The number of genetic findings for diabetes and diabetic complications has over doubled due to the discovery of novel genomics data, including bioinformatics and the aggregation of global cohort studies. Using genetic analysis to determine whether diabetes individuals are at the most risk for developing diabetic vascular complications (DVC) might lead to the development of more accurate early diagnostic biomarkers and the customization of care plans. CONCLUSIONS A newer method that uses extensive evaluation of single nucleotide polymorphisms (SNP) in big datasets is Genome-Wide Association Studies (GWAS).
Collapse
Affiliation(s)
- Zuira Tariq
- Diabetes and Endocrinology Department, University Hospital Sharjah, P.O. Box: 27272, Sharjah, United Arab Emirates
| | - Salah Abusnana
- Diabetes and Endocrinology Department, University Hospital Sharjah, P.O. Box: 27272, Sharjah, United Arab Emirates.
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates.
| | - Bashair M Mussa
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Hala Zakaria
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
34
|
Zeng M, Yang X, Chen Y, Fan J, Cao L, Wang M, Xiao P, Ling Z, Yin Y, Chen Y. A Network and Pathway Analysis of Genes Associated With Atrial Fibrillation. Cardiovasc Ther 2024; 2024:7054039. [PMID: 39742001 PMCID: PMC11470814 DOI: 10.1155/2024/7054039] [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: 06/19/2024] [Accepted: 08/09/2024] [Indexed: 01/03/2025] Open
Abstract
Background: Atrial fibrillation (AF) is affected by both environmental and genetic factors. Previous genetic association studies, especially genome-wide association studies, revealed a large group of AF-associated genes. However, little is known about the functions and interactions of these genes. Moreover, established genetic variants of AF contribute modestly to AF variance, implying that numerous additional AF-associated genetic variations need to be identified. Hence, a systematic network and pathway analysis is needed. Methods: We retrieved all AF-associated genes from genetic association studies in various databases and performed integrative analyses including pathway enrichment analysis, pathway crosstalk analysis, network analysis, and microarray meta-analysis. Results: We collected 254 AF-associated genes from genetic association studies in various databases. Pathway enrichment analysis revealed the top biological pathways that were enriched in the AF-associated genes related to cardiac electromechanical activity. Pathway crosstalk analysis showed that numerous neuro-endocrine-immune pathways connected AF with various diseases including cancers, inflammatory diseases, and cardiovascular diseases. Furthermore, an AF-specific subnetwork was constructed with the prize-collecting Steiner forest algorithm based on the AF-associated genes, and 24 novel genes that were potentially associated with AF were inferred by the subnetwork. In the microarray meta-analysis, six of the 24 novel genes (APLP1, CREB1, CREBBP, PRMT1, IRAK1, and PLXND1) were expressed differentially in patients with AF and sinus rhythm. Conclusions: AF is not only an isolated disease with abnormal electrophysiological activity but might also share a common genetic basis and biological process with tumors and inflammatory diseases as well as cardiovascular diseases. Moreover, the six novel genes inferred from network analysis might help detect the missing AF risk loci.
Collapse
Affiliation(s)
- Mengying Zeng
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Cardiac Electrophysiology, Chongqing, China
- Cardiac Arrhythmia Intervention Center of Chongqing Medical Quality Control Center, Chongqing, China
- Chongqing Atrial Fibrillation Center Alliance, Chongqing, China
| | - Xian Yang
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Cardiac Electrophysiology, Chongqing, China
- Cardiac Arrhythmia Intervention Center of Chongqing Medical Quality Control Center, Chongqing, China
- Chongqing Atrial Fibrillation Center Alliance, Chongqing, China
| | | | - Jinqi Fan
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Cardiac Electrophysiology, Chongqing, China
- Cardiac Arrhythmia Intervention Center of Chongqing Medical Quality Control Center, Chongqing, China
- Chongqing Atrial Fibrillation Center Alliance, Chongqing, China
| | - Li Cao
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Cardiac Electrophysiology, Chongqing, China
- Cardiac Arrhythmia Intervention Center of Chongqing Medical Quality Control Center, Chongqing, China
- Chongqing Atrial Fibrillation Center Alliance, Chongqing, China
| | - Menghao Wang
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Peilin Xiao
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Cardiac Electrophysiology, Chongqing, China
- Cardiac Arrhythmia Intervention Center of Chongqing Medical Quality Control Center, Chongqing, China
- Chongqing Atrial Fibrillation Center Alliance, Chongqing, China
| | - Zhiyu Ling
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Cardiac Electrophysiology, Chongqing, China
- Cardiac Arrhythmia Intervention Center of Chongqing Medical Quality Control Center, Chongqing, China
- Chongqing Atrial Fibrillation Center Alliance, Chongqing, China
| | - Yuehui Yin
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Cardiac Electrophysiology, Chongqing, China
- Cardiac Arrhythmia Intervention Center of Chongqing Medical Quality Control Center, Chongqing, China
- Chongqing Atrial Fibrillation Center Alliance, Chongqing, China
| | - Yunlin Chen
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Cardiac Electrophysiology, Chongqing, China
- Cardiac Arrhythmia Intervention Center of Chongqing Medical Quality Control Center, Chongqing, China
- Chongqing Atrial Fibrillation Center Alliance, Chongqing, China
| |
Collapse
|
35
|
Gong W, Yan Y, Liu J, Wang X, Zheng W, Que B, Ai H, Smith SC, Fonarow GC, Morgan L, Zhao D, Ma C, Han Y, Nie S, the CCC‐ACS Investigators. In-Hospital Mortality and Treatment in Patients With Acute Coronary Syndrome With and Without Standard Modifiable Cardiovascular Risk Factors: Findings From the CCC-ACS Project. J Am Heart Assoc 2024; 13:e029252. [PMID: 39291502 PMCID: PMC11681477 DOI: 10.1161/jaha.122.029252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 06/28/2024] [Indexed: 09/19/2024]
Abstract
BACKGROUND Patients with acute coronary syndrome without standard modifiable cardiovascular risk factors (SMuRFs; hypertension, smoking, dyslipidemia, diabetes) have not been well studied, with little known about their characteristics, quality of care, or outcomes. We sought to systematically analyze patients with ACS without SMuRFs, especially to evaluate the effectiveness of guideline-directed medical therapy for these patients. METHODS AND RESULTS In the CCC-ACS (Improving Care for Cardiovascular Disease in China-Acute Coronary Syndrome) project (2014-2019), we examined the presence and absence of SMuRFs and features among 89 462 patients with initial acute coronary syndrome. The main outcome was in-hospital all-cause mortality. Among eligible patients, 11.0% had none of the SMuRFs (SMuRF-less). SMuRF-less patients had higher in-hospital mortality (unadjusted hazard ratio [HR], 1.49 [95% CI, 1.19-1.87]). After adjustment for clinical characteristics and treatments, the associations between SMuRF status and in-hospital mortality persisted (adjusted HR, 1.35 [95% CI, 1.07-1.70]). Guideline-directed optimal medical therapy (receiving angiotensin-converting enzyme inhibitors/angiotensin receptor blockers, β-blockers, and statins) was not associated with lower mortality (adjusted HR, 0.98 [95% CI, 0.58-1.67]) in SMuRF-less patients, unlike the association in patients with SMuRFs (adjusted HR, 0.80 [95% CI, 0.66-0.98]). Sensitivity analyses were consistent with these results. CONCLUSIONS SMuRF-less patients were associated with an increased risk of in-hospital mortality. Guideline-directed medical therapy was less effective in SMuRF-less patients than in patients with SMuRFs. Dedicated studies are needed to confirm the optimal therapy for SMuRF-less patients. REGISTRATION URL: https://www.clinicaltrials.gov; Unique identifier: NCT02306616.
Collapse
Affiliation(s)
- Wei Gong
- Center for Coronary Artery Disease, Division of Cardiology, Beijing Anzhen HospitalCapital Medical UniversityBeijingChina
- National Clinical Research Center for Cardiovascular DiseasesBeijingChina
- Beijing Institute of HeartLung, and Blood Vessel DiseasesBeijingChina
| | - Yan Yan
- Center for Coronary Artery Disease, Division of Cardiology, Beijing Anzhen HospitalCapital Medical UniversityBeijingChina
- National Clinical Research Center for Cardiovascular DiseasesBeijingChina
- Beijing Institute of HeartLung, and Blood Vessel DiseasesBeijingChina
| | - Jing Liu
- Department of Epidemiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of HeartLung, and Blood Vessel DiseasesBeijingChina
| | - Xiao Wang
- Center for Coronary Artery Disease, Division of Cardiology, Beijing Anzhen HospitalCapital Medical UniversityBeijingChina
- National Clinical Research Center for Cardiovascular DiseasesBeijingChina
- Beijing Institute of HeartLung, and Blood Vessel DiseasesBeijingChina
| | - Wen Zheng
- Center for Coronary Artery Disease, Division of Cardiology, Beijing Anzhen HospitalCapital Medical UniversityBeijingChina
- National Clinical Research Center for Cardiovascular DiseasesBeijingChina
- Beijing Institute of HeartLung, and Blood Vessel DiseasesBeijingChina
| | - Bin Que
- Center for Coronary Artery Disease, Division of Cardiology, Beijing Anzhen HospitalCapital Medical UniversityBeijingChina
- National Clinical Research Center for Cardiovascular DiseasesBeijingChina
- Beijing Institute of HeartLung, and Blood Vessel DiseasesBeijingChina
| | - Hui Ai
- Center for Coronary Artery Disease, Division of Cardiology, Beijing Anzhen HospitalCapital Medical UniversityBeijingChina
- National Clinical Research Center for Cardiovascular DiseasesBeijingChina
- Beijing Institute of HeartLung, and Blood Vessel DiseasesBeijingChina
| | - Sidney C. Smith
- Division of CardiologyUniversity of North CarolinaChapel HillNCUSA
| | - Gregg C. Fonarow
- Division of CardiologyGeffen School of Medicine at University of CaliforniaLos AngelesCAUSA
| | - Louise Morgan
- International Quality Improvement DepartmentAmerican Heart AssociationDallasTXUSA
| | - Dong Zhao
- Department of Epidemiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of HeartLung, and Blood Vessel DiseasesBeijingChina
| | - Changsheng Ma
- National Clinical Research Center for Cardiovascular DiseasesBeijingChina
- Beijing Institute of HeartLung, and Blood Vessel DiseasesBeijingChina
| | - Yaling Han
- Department of CardiologyGeneral Hospital of Northern Theater CommandShenyangChina
| | - Shaoping Nie
- Center for Coronary Artery Disease, Division of Cardiology, Beijing Anzhen HospitalCapital Medical UniversityBeijingChina
- National Clinical Research Center for Cardiovascular DiseasesBeijingChina
- Beijing Institute of HeartLung, and Blood Vessel DiseasesBeijingChina
| | | |
Collapse
|
36
|
Takaoka M, Zhao X, Lim HY, Magnussen CG, Ang O, Suffee N, Schrank PR, Ong WS, Tsiantoulas D, Sommer F, Mohanta SK, Harrison J, Meng Y, Laurans L, Wu F, Lu Y, Masters L, Newland SA, Denti L, Hong M, Chajadine M, Juonala M, Koskinen JS, Kähönen M, Pahkala K, Rovio SP, Mykkänen J, Thomson R, Kaisho T, Habenicht AJR, Clement M, Tedgui A, Ait-Oufella H, Zhao TX, Nus M, Ruhrberg C, Taleb S, Williams JW, Raitakari OT, Angeli V, Mallat Z. Early intermittent hyperlipidaemia alters tissue macrophages to fuel atherosclerosis. Nature 2024; 634:457-465. [PMID: 39231480 PMCID: PMC11464399 DOI: 10.1038/s41586-024-07993-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 08/27/2024] [Indexed: 09/06/2024]
Abstract
Hyperlipidaemia is a major risk factor of atherosclerotic cardiovascular disease (ASCVD). Risk of cardiovascular events depends on cumulative lifetime exposure to low-density lipoprotein cholesterol (LDL-C) and, independently, on the time course of exposure to LDL-C, with early exposure being associated with a higher risk1. Furthermore, LDL-C fluctuations are associated with ASCVD outcomes2-4. However, the precise mechanisms behind this increased ASCVD risk are not understood. Here we find that early intermittent feeding of mice on a high-cholesterol Western-type diet (WD) accelerates atherosclerosis compared with late continuous exposure to the WD, despite similar cumulative circulating LDL-C levels. We find that early intermittent hyperlipidaemia alters the number and homeostatic phenotype of resident-like arterial macrophages. Macrophage genes with altered expression are enriched for genes linked to human ASCVD in genome-wide association studies. We show that LYVE1+ resident macrophages are atheroprotective, and identify biological pathways related to actin filament organization, of which alteration accelerates atherosclerosis. Using the Young Finns Study, we show that exposure to cholesterol early in life is significantly associated with the incidence and size of carotid atherosclerotic plaques in mid-adulthood. In summary, our results identify early intermittent exposure to cholesterol as a strong determinant of accelerated atherosclerosis, highlighting the importance of optimal control of hyperlipidaemia early in life, and providing insights into the underlying biological mechanisms. This knowledge will be essential to designing effective therapeutic strategies to combat ASCVD.
Collapse
MESH Headings
- Adolescent
- Adult
- Animals
- Child
- Child, Preschool
- Female
- Humans
- Male
- Mice
- Middle Aged
- Young Adult
- Atherosclerosis/epidemiology
- Atherosclerosis/etiology
- Atherosclerosis/genetics
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Cholesterol, LDL/blood
- Cholesterol, LDL/metabolism
- Diet, Western/adverse effects
- Diet, Western/statistics & numerical data
- Finland/epidemiology
- Genome-Wide Association Study
- Hyperlipidemias/complications
- Hyperlipidemias/epidemiology
- Hyperlipidemias/genetics
- Hyperlipidemias/metabolism
- Hyperlipidemias/pathology
- Incidence
- Macrophages/metabolism
- Macrophages/pathology
- Mice, Inbred C57BL
- Phenotype
- Plaque, Atherosclerotic/epidemiology
- Plaque, Atherosclerotic/etiology
- Plaque, Atherosclerotic/genetics
- Plaque, Atherosclerotic/metabolism
- Plaque, Atherosclerotic/pathology
- Time Factors
Collapse
Affiliation(s)
- Minoru Takaoka
- Department of Medicine, Section of CardioRespiratory Medicine, University of Cambridge, Heart and Lung Research Institute, Cambridge, UK
| | - Xiaohui Zhao
- Department of Medicine, Section of CardioRespiratory Medicine, University of Cambridge, Heart and Lung Research Institute, Cambridge, UK
| | - Hwee Ying Lim
- Immunology Translational Research Programme, Yong Loo Lin School of Medicine, Department of Microbiology and Immunology, National University of Singapore, Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Costan G Magnussen
- Research Centre of Applied and Preventive Cardiovascular Medicine; University of Turku, Turku, Finland
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Owen Ang
- Immunology Translational Research Programme, Yong Loo Lin School of Medicine, Department of Microbiology and Immunology, National University of Singapore, Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Nadine Suffee
- Université Paris Cité, Institut National de la Santé et de la Recherche Médicale, U970, PARCC, Paris, France
| | - Patricia R Schrank
- Department of Integrative Biology & Physiology, Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Wei Siong Ong
- Immunology Translational Research Programme, Yong Loo Lin School of Medicine, Department of Microbiology and Immunology, National University of Singapore, Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Dimitrios Tsiantoulas
- Department of Medicine, Section of CardioRespiratory Medicine, University of Cambridge, Heart and Lung Research Institute, Cambridge, UK
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Felix Sommer
- Institute of Clinical Molecular Biology, University of Kiel and University Hospital Schleswig Holstein (UKSH), Kiel, Germany
| | - Sarajo K Mohanta
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München (LMU), Munich, Germany
| | - James Harrison
- Department of Medicine, Section of CardioRespiratory Medicine, University of Cambridge, Heart and Lung Research Institute, Cambridge, UK
| | - Yaxing Meng
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Ludivine Laurans
- Université Paris Cité, Institut National de la Santé et de la Recherche Médicale, U970, PARCC, Paris, France
| | - Feitong Wu
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Yuning Lu
- Department of Medicine, Section of CardioRespiratory Medicine, University of Cambridge, Heart and Lung Research Institute, Cambridge, UK
| | - Leanne Masters
- Department of Medicine, Section of CardioRespiratory Medicine, University of Cambridge, Heart and Lung Research Institute, Cambridge, UK
| | - Stephen A Newland
- Department of Medicine, Section of CardioRespiratory Medicine, University of Cambridge, Heart and Lung Research Institute, Cambridge, UK
| | - Laura Denti
- Institute of Ophthalmology, University College London, London, UK
| | - Mingyang Hong
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München (LMU), Munich, Germany
| | - Mouna Chajadine
- Université Paris Cité, Institut National de la Santé et de la Recherche Médicale, U970, PARCC, Paris, France
| | - Markus Juonala
- Department of Medicine, University of Turku, Turku, Finland
- Division of Medicine, Turku University Hospital, Turku, Finland
| | - Juhani S Koskinen
- Research Centre of Applied and Preventive Cardiovascular Medicine; University of Turku, Turku, Finland
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
- Division of Medicine, Turku University Hospital, Turku, Finland
- Department of Medicine, Satakunta Central Hospital, Pori, Finland
| | - Mika Kähönen
- Department of Clinical Physiology, University of Tampere, Tampere, Finland
- Faculty of Medicine and Health Technology, University of Tampere, Tampere, Finland
- Finnish Cardiovascular Research Center Tampere, University of Tampere, Tampere, Finland
| | - Katja Pahkala
- Research Centre of Applied and Preventive Cardiovascular Medicine; University of Turku, Turku, Finland
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
| | - Suvi P Rovio
- Research Centre of Applied and Preventive Cardiovascular Medicine; University of Turku, Turku, Finland
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
| | - Juha Mykkänen
- Research Centre of Applied and Preventive Cardiovascular Medicine; University of Turku, Turku, Finland
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
| | - Russell Thomson
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Analytical Edge, Hobart, Tasmania, Australia
| | - Tsuneyasu Kaisho
- Department of Immunology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - Andreas J R Habenicht
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München (LMU), Munich, Germany
| | - Marc Clement
- Department of Medicine, Section of CardioRespiratory Medicine, University of Cambridge, Heart and Lung Research Institute, Cambridge, UK
| | - Alain Tedgui
- Université Paris Cité, Institut National de la Santé et de la Recherche Médicale, U970, PARCC, Paris, France
| | - Hafid Ait-Oufella
- Université Paris Cité, Institut National de la Santé et de la Recherche Médicale, U970, PARCC, Paris, France
| | - Tian X Zhao
- Department of Medicine, Section of CardioRespiratory Medicine, University of Cambridge, Heart and Lung Research Institute, Cambridge, UK
| | - Meritxell Nus
- Department of Medicine, Section of CardioRespiratory Medicine, University of Cambridge, Heart and Lung Research Institute, Cambridge, UK
| | | | - Soraya Taleb
- Université Paris Cité, Institut National de la Santé et de la Recherche Médicale, U970, PARCC, Paris, France
| | - Jesse W Williams
- Department of Integrative Biology & Physiology, Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Olli T Raitakari
- Research Centre of Applied and Preventive Cardiovascular Medicine; University of Turku, Turku, Finland
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland
| | - Véronique Angeli
- Immunology Translational Research Programme, Yong Loo Lin School of Medicine, Department of Microbiology and Immunology, National University of Singapore, Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Ziad Mallat
- Department of Medicine, Section of CardioRespiratory Medicine, University of Cambridge, Heart and Lung Research Institute, Cambridge, UK.
- Université Paris Cité, Institut National de la Santé et de la Recherche Médicale, U970, PARCC, Paris, France.
| |
Collapse
|
37
|
Liu HH, Li S, Zhang Y, Guo YL, Zhu CG, Wu NQ, Gao Y, Xu RX, Dong Q, Li JJ. Joint Association of Lipoprotein(a) and a Family History of Coronary Artery Disease with the Cardiovascular Outcomes in Patients with Chronic Coronary Syndrome. J Atheroscler Thromb 2024; 31:1319-1332. [PMID: 38616111 PMCID: PMC11374575 DOI: 10.5551/jat.64693] [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: 10/19/2023] [Accepted: 02/15/2024] [Indexed: 04/16/2024] Open
Abstract
AIM No data are currently available regarding the association between Lp(a) and the cardiovascular outcomes in patients with coronary artery disease (CAD) according to their family history (FHx) of CAD. This study aimed to evaluate the significance of Lp(a) in predicting major adverse cardiovascular events (MACEs) in patients with chronic coronary syndrome (CCS) with or without FHx. METHODS A total of 6056 patients with CCS were enrolled. Information on FHx was collected, and the plasma Lp(a) levels were measured. All patients were followed up regularly. The independent and joint associations of Lp(a) and FHx with the risk of MACEs, including cardiovascular death, nonfatal myocardial infarction, and stroke, were analyzed. RESULTS With over an average of 50.35±18.58 months follow-up, 378 MACEs were recorded. A Cox regression analysis showed an elevated Lp(a) level to be an independent predictor for MACEs in patients with [hazard ratio (HR): 2.77, 95% confidence interval (CI): 1.38-5.54] or without FHx (HR: 1.35, 95% CI: 1.02-1.77). In comparison to subjects with non-elevated Lp(a) and negative FHx, patients with elevated Lp(a) alone were at a nominally higher risk of MACEs (HR: 1.26, 95% CI: 0.96-1.67), while those with both had the highest risk (HR: 1.93, 95% CI: 1.14-3.28). Moreover, adding Lp(a) to the original model increased the C-statistic by 0.048 in subjects with FHx (p=0.004) and by 0.004 in those without FHx (p=0.391). CONCLUSIONS The present study is the first to suggest that Lp(a) could be used to predict MACEs in CCS patients with or without FHx; however, its prognostic significance was more noteworthy in patients with FHx.
Collapse
Affiliation(s)
- Hui-Hui Liu
- Cardiometabolic Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, National Clinical Research, Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Heart Failure Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, National Clinical Research, Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Sha Li
- Cardiometabolic Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, National Clinical Research, Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yan Zhang
- Cardiometabolic Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, National Clinical Research, Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuan-Lin Guo
- Cardiometabolic Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, National Clinical Research, Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Cheng-Gang Zhu
- Cardiometabolic Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, National Clinical Research, Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Na-Qiong Wu
- Cardiometabolic Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, National Clinical Research, Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ying Gao
- Cardiometabolic Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, National Clinical Research, Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Rui-Xia Xu
- Cardiometabolic Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, National Clinical Research, Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qian Dong
- Cardiometabolic Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, National Clinical Research, Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jian-Jun Li
- Cardiometabolic Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, National Clinical Research, Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
38
|
Zhang L, Feng Q, Kong W. ECM Microenvironment in Vascular Homeostasis: New Targets for Atherosclerosis. Physiology (Bethesda) 2024; 39:0. [PMID: 38984789 DOI: 10.1152/physiol.00028.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 03/05/2024] [Accepted: 03/23/2024] [Indexed: 07/11/2024] Open
Abstract
Alterations in vascular extracellular matrix (ECM) components, interactions, and mechanical properties influence both the formation and stability of atherosclerotic plaques. This review discusses the contribution of the ECM microenvironment in vascular homeostasis and remodeling in atherosclerosis, highlighting Cartilage oligomeric matrix protein (COMP) and its degrading enzyme ADAMTS7 as examples, and proposes potential avenues for future research aimed at identifying novel therapeutic targets for atherosclerosis based on the ECM microenvironment.
Collapse
Affiliation(s)
- Lu Zhang
- Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qianqian Feng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Wei Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| |
Collapse
|
39
|
Cañadas-Garre M, Maqueda JJ, Baños-Jaime B, Hill C, Skelly R, Cappa R, Brennan E, Doyle R, Godson C, Maxwell AP, McKnight AJ. Mitochondrial related variants associated with cardiovascular traits. Front Physiol 2024; 15:1395371. [PMID: 39258111 PMCID: PMC11385366 DOI: 10.3389/fphys.2024.1395371] [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: 03/03/2024] [Accepted: 08/05/2024] [Indexed: 09/12/2024] Open
Abstract
Introduction Cardiovascular disease (CVD) is responsible for over 30% of mortality worldwide. CVD arises from the complex influence of molecular, clinical, social, and environmental factors. Despite the growing number of autosomal genetic variants contributing to CVD, the cause of most CVDs is still unclear. Mitochondria are crucial in the pathophysiology, development and progression of CVDs; the impact of mitochondrial DNA (mtDNA) variants and mitochondrial haplogroups in the context of CVD has recently been highlighted. Aims We investigated the role of genetic variants in both mtDNA and nuclear-encoded mitochondrial genes (NEMG) in CVD, including coronary artery disease (CAD), hypertension, and serum lipids in the UK Biobank, with sub-group analysis for diabetes. Methods We investigated 371,542 variants in 2,527 NEMG, along with 192 variants in 32 mitochondrial genes in 381,994 participants of the UK Biobank, stratifying by presence of diabetes. Results Mitochondrial variants showed associations with CVD, hypertension, and serum lipids. Mitochondrial haplogroup J was associated with CAD and serum lipids, whereas mitochondrial haplogroups T and U were associated with CVD. Among NEMG, variants within Nitric Oxide Synthase 3 (NOS3) showed associations with CVD, CAD, hypertension, as well as diastolic and systolic blood pressure. We also identified Translocase Of Outer Mitochondrial Membrane 40 (TOMM40) variants associated with CAD; Solute carrier family 22 member 2 (SLC22A2) variants associated with CAD and CVD; and HLA-DQA1 variants associated with hypertension. Variants within these three genes were also associated with serum lipids. Conclusion Our study demonstrates the relevance of mitochondrial related variants in the context of CVD. We have linked mitochondrial haplogroup U to CVD, confirmed association of mitochondrial haplogroups J and T with CVD and proposed new markers of hypertension and serum lipids in the context of diabetes. We have also evidenced connections between the etiological pathways underlying CVDs, blood pressure and serum lipids, placing NOS3, SLC22A2, TOMM40 and HLA-DQA1 genes as common nexuses.
Collapse
Affiliation(s)
- Marisa Cañadas-Garre
- Molecular Epidemiology and Public Health Research Group, Centre for Public Health, Queen's University Belfast, Institute for Clinical Sciences A, Royal Victoria Hospital, Belfast, United Kingdom
- MRC Integrative Epidemiology Unit, Bristol Medical School (Population Health Sciences), University of Bristol Oakfield House, Belfast, United Kingdom
| | - Joaquín J Maqueda
- Molecular Epidemiology and Public Health Research Group, Centre for Public Health, Queen's University Belfast, Institute for Clinical Sciences A, Royal Victoria Hospital, Belfast, United Kingdom
- Laboratory of Experimental Oncology, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Blanca Baños-Jaime
- Molecular Epidemiology and Public Health Research Group, Centre for Public Health, Queen's University Belfast, Institute for Clinical Sciences A, Royal Victoria Hospital, Belfast, United Kingdom
- Instituto de Investigaciones Químicas (IIQ), Centro de Investigaciones Científicas Isla de la Cartuja (cicCartuja), Universidad de Sevilla, Consejo Superior de Investigaciones Científicas (CSIC), Sevilla, Spain
| | - Claire Hill
- Molecular Epidemiology and Public Health Research Group, Centre for Public Health, Queen's University Belfast, Institute for Clinical Sciences A, Royal Victoria Hospital, Belfast, United Kingdom
| | - Ryan Skelly
- Molecular Epidemiology and Public Health Research Group, Centre for Public Health, Queen's University Belfast, Institute for Clinical Sciences A, Royal Victoria Hospital, Belfast, United Kingdom
| | - Ruaidhri Cappa
- Molecular Epidemiology and Public Health Research Group, Centre for Public Health, Queen's University Belfast, Institute for Clinical Sciences A, Royal Victoria Hospital, Belfast, United Kingdom
| | - Eoin Brennan
- UCD Diabetes Complications Research Centre, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
- School of Medicine, University College Dublin, Dublin, Ireland
| | - Ross Doyle
- UCD Diabetes Complications Research Centre, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
- School of Medicine, University College Dublin, Dublin, Ireland
- Mater Misericordiae University Hospital, Dublin, Ireland
| | - Catherine Godson
- UCD Diabetes Complications Research Centre, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
- School of Medicine, University College Dublin, Dublin, Ireland
| | - Alexander P Maxwell
- Molecular Epidemiology and Public Health Research Group, Centre for Public Health, Queen's University Belfast, Institute for Clinical Sciences A, Royal Victoria Hospital, Belfast, United Kingdom
- Regional Nephrology Unit, Belfast City Hospital Belfast, Belfast, United Kingdom
| | - Amy Jayne McKnight
- Molecular Epidemiology and Public Health Research Group, Centre for Public Health, Queen's University Belfast, Institute for Clinical Sciences A, Royal Victoria Hospital, Belfast, United Kingdom
| |
Collapse
|
40
|
Fry H, Mazidi M, Kartsonaki C, Clarke R, Walters RG, Chen Z, Millwood IY. The Role of Furin and Its Therapeutic Potential in Cardiovascular Disease Risk. Int J Mol Sci 2024; 25:9237. [PMID: 39273186 PMCID: PMC11394739 DOI: 10.3390/ijms25179237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/07/2024] [Accepted: 08/08/2024] [Indexed: 09/15/2024] Open
Abstract
Furin is an important proteolytic enzyme, converting several proteins from inactive precursors to their active forms. Recently, proteo-genomic analyses in European and East Asian populations suggested a causal association of furin with ischaemic heart disease, and there is growing interest in its role in cardiovascular disease (CVD) aetiology. In this narrative review, we present a critical appraisal of evidence from population studies to assess furin's role in CVD risk and potential as a drug target for CVD. Whilst most observational studies report positive associations between furin expression and CVD risk, some studies report opposing effects, which may reflect the complex biological roles of furin and its substrates. Genetic variation in FURIN is also associated with CVD and its risk factors. We found no evidence of current clinical development of furin as a drug target for CVD, although several phase 1 and 2 clinical trials of furin inhibitors as a type of cancer immunotherapy have been completed. The growing field of proteo-genomics in large-scale population studies may inform the future development of furin and other potential drug targets to improve the treatment and prevention of CVD.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Iona Y. Millwood
- Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK; (H.F.); (M.M.); (C.K.); (R.C.); (R.G.W.); (Z.C.)
| |
Collapse
|
41
|
Fu C, Wang L, Cai W. IL6 receptor inhibitors: exploring the therapeutic potential across multiple diseases through drug target Mendelian randomization. Front Immunol 2024; 15:1452849. [PMID: 39229261 PMCID: PMC11368790 DOI: 10.3389/fimmu.2024.1452849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 08/05/2024] [Indexed: 09/05/2024] Open
Abstract
Background High interleukin-6 levels correlate with diseases like cancer, autoimmune disorders, and infections. IL-6 receptor inhibitors (IL-6Ri), used for rheumatoid arthritis and COVID-19, may have wider uses. We apply drug-target Mendelian Randomization (MR) to study IL-6Ri's effects. Method To simulate the effects of genetically blocking the IL-6R, we selected single nucleotide polymorphisms (SNPs) within or near the IL6R gene that show significant genome-wide associations with C-reactive protein. Using rheumatoid arthritis and COVID-19 as positive controls, our primary research outcomes included the risk of asthma, asthmatic pneumonia, cor pulmonale, non-small cell lung cancer, small cell lung cancer, Parkinson's disease, Alzheimer's disease, ulcerative colitis, Crohn's disease, systemic lupus erythematosus, type 1 diabetes, and type 2 diabetes. The Inverse Variance Weighted (IVW) method served as our principal analytical approach, with the hypotheses of MR being evaluated through sensitivity and colocalization analyses. Additionally, we conducted Bayesian Mendelian Randomization analyses to minimize confounding and reverse causation biases to the greatest extent possible. Results IL-6 inhibitors significantly reduced the risk of idiopathic pulmonary fibrosis (OR= 0.278, 95% [CI], 0.138-0.558; P <0.001), Parkinson's disease (OR = 0.354, 95% CI, 0.215-0.582; P <0.001), and positively influenced the causal relationship with Type 2 diabetes (OR = 0.759, 95% CI, 0.637-0.905; P = 0.002). However, these inhibitors increased the risk for asthma (OR = 1.327, 95% CI, 1.118-1.576; P = 0.001) and asthmatic pneumonia (OR = 1.823, 95% CI, 1.246-2.666; P = 0.002). The causal effect estimates obtained via the BWMR method are consistent with those based on the IVW approach. Similarly, sIL-6R also exerts a significant influence on these diseases.Diseases such as Alzheimer's disease, Crohn's disease, pulmonary heart disease, systemic lupus erythematosus, Type 1 diabetes, Non-small cell lung cancer and ulcerative colitis showed non-significant associations (p > 0.05) and were excluded from further analysis. Similarly, Small cell lung cancer were excluded due to inconsistent results. Notably, the colocalization evidence for asthmatic pneumonia (coloc.abf-PPH4 = 0.811) robustly supports its association with CRP. The colocalization evidence for Parkinson's disease (coloc.abf-PPH4 = 0.725) moderately supports its association with CRP. Conclusion IL-6Ri may represent a promising therapeutic avenue for idiopathic pulmonary fibrosis, Parkinson's disease, and Type 2 diabetes.
Collapse
Affiliation(s)
- Chong Fu
- Department of Gastroenterology, Anqing Municipal Hospital, Anqing, Anhui, China
| | - Longquan Wang
- Department of Geriatric Medicine, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui, China
| | - Wenjiao Cai
- Department of Nephrology, Anqing Municipal Hospital, Anqing, Anhui, China
| |
Collapse
|
42
|
Rudman-Melnick V, Vanhoutte D, Stowers K, Sargent M, Adam M, Ma Q, Perl AKT, Miethke AG, Burg A, Shi T, Hildeman DA, Woodle ESS, Kofron JM, Devarajan P. Gucy1α1 specifically marks kidney, heart, lung and liver fibroblasts. RESEARCH SQUARE 2024:rs.3.rs-4746078. [PMID: 39184103 PMCID: PMC11343171 DOI: 10.21203/rs.3.rs-4746078/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Fibrosis is a common outcome of numerous pathologies, including chronic kidney disease (CKD), a progressive renal function deterioration. Current approaches to target activated fibroblasts, key effector contributors to fibrotic tissue remodeling, lack specificity. Here, we report Gucy1α1 as a specific kidney fibroblast marker. Gucy1α1 levels significantly increased over the course of two clinically relevant murine CKD models and directly correlated with established fibrosis markers. Immunofluorescent (IF) imaging showed that Gucy1α1 comprehensively labelled cortical and medullary quiescent and activated fibroblasts in the control kidney and throughout injury progression, respectively. Unlike traditionally used markers platelet derived growth factor receptor beta (Pdgfrβ) and vimentin (Vim), Gucy1α1 did not overlap with off-target populations such as podocytes. Notably, Gucy1α1 labelled kidney fibroblasts in both male and female mice. Furthermore, we observed elevated GUCY1α1 expression in the human fibrotic kidney and lung. Studies in the murine models of cardiac and liver fibrosis revealed Gucy1α1 elevation in activated Pdgfrβ-, Vim- and alpha smooth muscle actin (αSma)-expressing fibroblasts paralleling injury progression and resolution. Overall, we demonstrate Gucy1α1 as an exclusive fibroblast marker in both sexes. Due to its multiorgan translational potential, GUCY1α1 might provide a novel promising strategy to specifically target and mechanistically examine fibroblasts.
Collapse
|
43
|
Afzal Z, Cao H, Chaudhary M, Chigurupati HD, Neppala S, Alruwaili W, Awad M, Sandesara D, Siddique M, Farman A, Zafrullah F, Gonuguntla K, Sattar Y. Elevated lipoprotein(a) levels: A crucial determinant of cardiovascular disease risk and target for emerging therapies. Curr Probl Cardiol 2024; 49:102586. [PMID: 38653440 DOI: 10.1016/j.cpcardiol.2024.102586] [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: 04/15/2024] [Accepted: 04/20/2024] [Indexed: 04/25/2024]
Abstract
Cardiovascular disease (CVD) remains a significant global health challenge despite advancements in prevention and treatment. Elevated Lipoprotein(a) [Lp(a)] levels have emerged as a crucial risk factor for CVD and aortic stenosis, affecting approximately 20 of the global population. Research over the last decade has established Lp(a) as an independent genetic contributor to CVD and aortic stenosis, beginning with Kare Berg's discovery in 1963. This has led to extensive exploration of its molecular structure and pathogenic roles. Despite the unknown physiological function of Lp(a), studies have shed light on its metabolism, genetics, and involvement in atherosclerosis, inflammation, and thrombosis. Epidemiological evidence highlights the link between high Lp(a) levels and increased cardiovascular morbidity and mortality. Newly emerging therapies, including pelacarsen, zerlasiran, olpasiran, muvalaplin, and lepodisiran, show promise in significantly lowering Lp(a) levels, potentially transforming the management of cardiovascular disease. However, further research is essential to assess these novel therapies' long-term efficacy and safety, heralding a new era in cardiovascular disease prevention and treatment and providing hope for at-risk patients.
Collapse
Affiliation(s)
- Zeeshan Afzal
- Department of Medicine, Shanxi Medical University, China
| | - Huili Cao
- Department of Cardiology, Second Hospital of Shanxi Medical University, China
| | | | - Himaja Dutt Chigurupati
- Department of Internal Medicine, New York Medical College at Saint Michael's Medical Center, NJ, USA
| | - Sivaram Neppala
- Department of Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | - Waleed Alruwaili
- Department of Internal Medicine, West Virginia University, Morgantown, WV, USA
| | - Maan Awad
- Department of Internal Medicine, West Virginia University, Morgantown, WV, USA
| | | | | | - Ali Farman
- Department of Medicine, Corewell Health Dearborn Hospital, Dearborn, MI, USA
| | - Fnu Zafrullah
- Department of Cardiology, Ascension Borgess Hospital, MI, USA
| | | | - Yasar Sattar
- Department of Cardiology, West Virginia University, Morgantown, WV, USA.
| |
Collapse
|
44
|
Han X, Zeng Y, Shang Y, Hu Y, Hou C, Yang H, Chen W, Ying Z, Sun Y, Qu Y, Wang J, Zhang W, Fang F, Valdimarsdóttir U, Song H. Risk of Cardiovascular Disease Hospitalization After Common Psychiatric Disorders: Analyses of Disease Susceptibility and Progression Trajectory in the UK Biobank. PHENOMICS (CHAM, SWITZERLAND) 2024; 4:327-338. [PMID: 39583312 PMCID: PMC11584824 DOI: 10.1007/s43657-023-00134-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 10/01/2023] [Accepted: 10/11/2023] [Indexed: 11/26/2024]
Abstract
Whether associations between psychiatric disorders and hospitalization for cardiovascular diseases (CVDs) can be modified by disease susceptibility and the temporal pattern of these associated CVDs remain unknown. In our study, we conducted a matched cohort study of the UK Biobank including 44,430 patients with common psychiatric disorders (anxiety, depression, and stress-related disorders) between 1997 and 2019, together with 222,150 sex-, Townsend deprivation index-, and birth year- individually matched unexposed individuals. The hazard ratios (HRs) for CVD hospitalization associated with a prior psychiatric disorder were derived from Cox models, adjusted for multiple confounders. We then stratified the analyses by self-reported family history of CVD and CVD polygenic risk score (PRS) calculated based on summary statistics of independent genome-wide association studies. We further conducted disease trajectory analysis and visualized the temporal pattern of CVDs after common psychiatric disorders. During a mean follow-up of 12.28 years, we observed an elevated risk of CVD hospitalization among patients with psychiatric disorders, compared with matched unexposed individuals (hazard ratios [HRs] = 1.20, 95% confidence interval [CI]: 1.18-1.23), especially during the first six months of follow-up (1.72 [1.55-1.91]). The stratification analyses by family history of CVD and by CVD PRS obtained similar estimates between subgroups with different susceptibilities to CVD. We conducted trajectory analysis to visualize the temporal pattern of CVDs after common psychiatric disorders, identifying primary hypertension, acute myocardial infarction, and stroke as three main intermediate steps leading to further increased risk of other CVDs. In conclusion, the association between common psychiatric disorders and subsequent CVD hospitalization is not modified by predisposition to CVD. Hypertension, acute myocardial infarction, and stroke are three initial CVDs linking psychiatric disorders to other CVD sequelae, highlighting a need of timely intervention on these targets to prevent further CVD sequelae among all individuals with common psychiatric disorders. Supplementary Information The online version contains supplementary material available at 10.1007/s43657-023-00134-w.
Collapse
Affiliation(s)
- Xin Han
- Mental Health Center and West China Biomedical Big Data Center, West China Hospital, Sichuan University, Guo Xue Lane 37, Chengdu, 610000 China
- Med-X Center for Informatics, Sichuan University, Chengdu, 610000 China
| | - Yu Zeng
- Mental Health Center and West China Biomedical Big Data Center, West China Hospital, Sichuan University, Guo Xue Lane 37, Chengdu, 610000 China
- Med-X Center for Informatics, Sichuan University, Chengdu, 610000 China
| | - Yanan Shang
- Mental Health Center and West China Biomedical Big Data Center, West China Hospital, Sichuan University, Guo Xue Lane 37, Chengdu, 610000 China
- Med-X Center for Informatics, Sichuan University, Chengdu, 610000 China
| | - Yao Hu
- Mental Health Center and West China Biomedical Big Data Center, West China Hospital, Sichuan University, Guo Xue Lane 37, Chengdu, 610000 China
- Med-X Center for Informatics, Sichuan University, Chengdu, 610000 China
| | - Can Hou
- Mental Health Center and West China Biomedical Big Data Center, West China Hospital, Sichuan University, Guo Xue Lane 37, Chengdu, 610000 China
- Med-X Center for Informatics, Sichuan University, Chengdu, 610000 China
| | - Huazhen Yang
- Mental Health Center and West China Biomedical Big Data Center, West China Hospital, Sichuan University, Guo Xue Lane 37, Chengdu, 610000 China
- Med-X Center for Informatics, Sichuan University, Chengdu, 610000 China
| | - Wenwen Chen
- Division of Nephrology, Kidney Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610000 China
| | - Zhiye Ying
- Mental Health Center and West China Biomedical Big Data Center, West China Hospital, Sichuan University, Guo Xue Lane 37, Chengdu, 610000 China
- Med-X Center for Informatics, Sichuan University, Chengdu, 610000 China
| | - Yajing Sun
- Mental Health Center and West China Biomedical Big Data Center, West China Hospital, Sichuan University, Guo Xue Lane 37, Chengdu, 610000 China
- Med-X Center for Informatics, Sichuan University, Chengdu, 610000 China
| | - Yuanyuan Qu
- Mental Health Center and West China Biomedical Big Data Center, West China Hospital, Sichuan University, Guo Xue Lane 37, Chengdu, 610000 China
- Med-X Center for Informatics, Sichuan University, Chengdu, 610000 China
| | - Junren Wang
- Mental Health Center and West China Biomedical Big Data Center, West China Hospital, Sichuan University, Guo Xue Lane 37, Chengdu, 610000 China
- Med-X Center for Informatics, Sichuan University, Chengdu, 610000 China
| | - Wei Zhang
- Mental Health Center and West China Biomedical Big Data Center, West China Hospital, Sichuan University, Guo Xue Lane 37, Chengdu, 610000 China
- Med-X Center for Informatics, Sichuan University, Chengdu, 610000 China
- Mental Health Center, West China Hospital of Sichuan University, Chengdu, 610000 China
| | - Fang Fang
- Institute of Environmental Medicine, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Unnur Valdimarsdóttir
- Center of Public Health Sciences, Faculty of Medicine, University of Iceland, 102 Reykjavík, Iceland
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 17177 Stockholm, Sweden
- Department of Epidemiology, Harvard T H Chan School of Public Health, Boston, MA 02115 USA
| | - Huan Song
- Mental Health Center and West China Biomedical Big Data Center, West China Hospital, Sichuan University, Guo Xue Lane 37, Chengdu, 610000 China
- Med-X Center for Informatics, Sichuan University, Chengdu, 610000 China
- Center of Public Health Sciences, Faculty of Medicine, University of Iceland, 102 Reykjavík, Iceland
| |
Collapse
|
45
|
Baker S, Biroli P, van Kippersluis H, von Hinke S. Advantageous early-life environments cushion the genetic risk for ischemic heart disease. Proc Natl Acad Sci U S A 2024; 121:e2314056121. [PMID: 38917008 PMCID: PMC11228495 DOI: 10.1073/pnas.2314056121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 04/18/2024] [Indexed: 06/27/2024] Open
Abstract
In one of the first papers on the impact of early-life conditions on individuals' health in older age, Barker and Osmond [Lancet, 327, 1077-1081 (1986)] show a strong positive relationship between infant mortality rates in the 1920s and ischemic heart disease in the 1970s. We merge historical data on infant mortality rates to 370,000 individual records in the UK Biobank using information on local area and year of birth. We replicate the association between the early-life infant mortality rate and later-life ischemic heart disease in our sample. We then go "beyond Barker," by showing considerable genetic heterogeneity in this association that is robust to within-area as well as within-family analyses. We find no association between the polygenic index and heart disease in areas with the lowest infant mortality rates, but a strong positive relationship in areas characterized by high infant mortality. These findings suggest that advantageous environments can cushion one's genetic disease risk.
Collapse
Affiliation(s)
- Samuel Baker
- School of Economics, University of Bristol, BristolBS8 1TU, United Kingdom
| | - Pietro Biroli
- Department of Economic Sciences, University of Bologna, Bologna, Italy
| | - Hans van Kippersluis
- Erasmus School of Economics, Erasmus University Rotterdam, 3062 PARotterdam, The Netherlands
| | - Stephanie von Hinke
- School of Economics, University of Bristol, BristolBS8 1TU, United Kingdom
- Institute for Fiscal Studies, LondonWC1E 7AE, United Kingdom
| |
Collapse
|
46
|
Trastulla L, Dolgalev G, Moser S, Jiménez-Barrón LT, Andlauer TFM, von Scheidt M, Budde M, Heilbronner U, Papiol S, Teumer A, Homuth G, Völzke H, Dörr M, Falkai P, Schulze TG, Gagneur J, Iorio F, Müller-Myhsok B, Schunkert H, Ziller MJ. Distinct genetic liability profiles define clinically relevant patient strata across common diseases. Nat Commun 2024; 15:5534. [PMID: 38951512 PMCID: PMC11217418 DOI: 10.1038/s41467-024-49338-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 05/31/2024] [Indexed: 07/03/2024] Open
Abstract
Stratified medicine holds great promise to tailor treatment to the needs of individual patients. While genetics holds great potential to aid patient stratification, it remains a major challenge to operationalize complex genetic risk factor profiles to deconstruct clinical heterogeneity. Contemporary approaches to this problem rely on polygenic risk scores (PRS), which provide only limited clinical utility and lack a clear biological foundation. To overcome these limitations, we develop the CASTom-iGEx approach to stratify individuals based on the aggregated impact of their genetic risk factor profiles on tissue specific gene expression levels. The paradigmatic application of this approach to coronary artery disease or schizophrenia patient cohorts identified diverse strata or biotypes. These biotypes are characterized by distinct endophenotype profiles as well as clinical parameters and are fundamentally distinct from PRS based groupings. In stark contrast to the latter, the CASTom-iGEx strategy discovers biologically meaningful and clinically actionable patient subgroups, where complex genetic liabilities are not randomly distributed across individuals but rather converge onto distinct disease relevant biological processes. These results support the notion of different patient biotypes characterized by partially distinct pathomechanisms. Thus, the universally applicable approach presented here has the potential to constitute an important component of future personalized medicine paradigms.
Collapse
Affiliation(s)
- Lucia Trastulla
- Max Planck Institute of Psychiatry, Munich, Germany
- Technische Universität München Medical Graduate Center Experimental Medicine, Munich, Germany
- Human Technopole, Milan, Italy
- Department of Psychiatry, University of Münster, Münster, Germany
| | - Georgii Dolgalev
- Department of Psychiatry, University of Münster, Münster, Germany
| | - Sylvain Moser
- Max Planck Institute of Psychiatry, Munich, Germany
- Technische Universität München Medical Graduate Center Experimental Medicine, Munich, Germany
- International Max Planck Research School for Translational Psychiatry (IMPRS-TP), Munich, Germany
| | - Laura T Jiménez-Barrón
- Max Planck Institute of Psychiatry, Munich, Germany
- International Max Planck Research School for Translational Psychiatry (IMPRS-TP), Munich, Germany
| | - Till F M Andlauer
- Max Planck Institute of Psychiatry, Munich, Germany
- Department of Neurology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Moritz von Scheidt
- Klinik für Herz-und Kreislauferkrankungen, Deutsches Herzzentrum München, Technical University Munich, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Monika Budde
- Institute of Psychiatric Phenomics and Genomics (IPPG), LMU University Hospital, LMU Munich, Munich, 80336, Germany
| | - Urs Heilbronner
- Institute of Psychiatric Phenomics and Genomics (IPPG), LMU University Hospital, LMU Munich, Munich, 80336, Germany
| | - Sergi Papiol
- Max Planck Institute of Psychiatry, Munich, Germany
- Institute of Psychiatric Phenomics and Genomics (IPPG), LMU University Hospital, LMU Munich, Munich, 80336, Germany
| | - Alexander Teumer
- German Center for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, Germany
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | - Georg Homuth
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Henry Völzke
- German Center for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, Germany
- Institute of Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Marcus Dörr
- German Center for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, Germany
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany
| | - Peter Falkai
- Max Planck Institute of Psychiatry, Munich, Germany
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, 80336, Germany
| | - Thomas G Schulze
- Institute of Psychiatric Phenomics and Genomics (IPPG), LMU University Hospital, LMU Munich, Munich, 80336, Germany
- Department of Psychiatry and Behavioral Sciences, SUNY Upstate Medical University, Syracuse, NY, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Julien Gagneur
- School of Computation, Information and Technology, Technical University of Munich, Garching, Germany
- Institute of Human Genetics, School of Medicine and Health, Technical University of Munich, Munich, Germany
- Computational Health Center, Helmholtz Center Munich, Neuherberg, Germany
| | | | - Bertram Müller-Myhsok
- Max Planck Institute of Psychiatry, Munich, Germany
- Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Heribert Schunkert
- Klinik für Herz-und Kreislauferkrankungen, Deutsches Herzzentrum München, Technical University Munich, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Michael J Ziller
- Max Planck Institute of Psychiatry, Munich, Germany.
- Department of Psychiatry, University of Münster, Münster, Germany.
- Center for Soft Nanoscience, University of Münster, Münster, Germany.
| |
Collapse
|
47
|
Shi W, Song J, Weiner JM, Chopra A, Dommisch H, Beule D, Schaefer AS. lncRNA CDKN2B-AS1 regulates collagen expression. Hum Genet 2024; 143:907-919. [PMID: 38833008 PMCID: PMC11294485 DOI: 10.1007/s00439-024-02674-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 04/27/2024] [Indexed: 06/06/2024]
Abstract
The long noncoding RNA CDKN2B-AS1 harbors a major coronary artery disease risk haplotype, which is also associated with progressive forms of the oral inflammatory disease periodontitis as well as myocardial infarction (MI). Despite extensive research, there is currently no broad consensus on the function of CDKN2B-AS1 that would explain a common molecular role of this lncRNA in these diseases. Our aim was to investigate the role of CDKN2B-AS1 in gingival cells to better understand the molecular mechanisms underlying the increased risk of progressive periodontitis. We downregulated CDKN2B-AS1 transcript levels in primary gingival fibroblasts with LNA GapmeRs. Following RNA-sequencing, we performed differential expression, gene set enrichment analyses and Western Blotting. Putative causal alleles were searched by analyzing associated DNA sequence variants for changes of predicted transcription factor binding sites. We functionally characterized putative functional alleles using luciferase-reporter and antibody electrophoretic mobility shift assays in gingival fibroblasts and HeLa cells. Of all gene sets analysed, collagen biosynthesis was most significantly upregulated (Padj=9.7 × 10- 5 (AUC > 0.65) with the CAD and MI risk gene COL4A1 showing strongest upregulation of the enriched gene sets (Fold change = 12.13, Padj = 4.9 × 10- 25). The inflammatory "TNFA signaling via NFKB" gene set was downregulated the most (Padj=1 × 10- 5 (AUC = 0.60). On the single gene level, CAPNS2, involved in extracellular matrix organization, was the top upregulated protein coding gene (Fold change = 48.5, P < 9 × 10- 24). The risk variant rs10757278 altered a binding site of the pathogen responsive transcription factor STAT1 (P = 5.8 × 10- 6). rs10757278-G allele reduced STAT1 binding 14.4% and rs10757278-A decreased luciferase activity in gingival fibroblasts 41.2% (P = 0.0056), corresponding with GTEx data. CDKN2B-AS1 represses collagen gene expression in gingival fibroblasts. Dysregulated collagen biosynthesis through allele-specific CDKN2B-AS1 expression in response to inflammatory factors may affect collagen synthesis, and in consequence tissue barrier and atherosclerotic plaque stability.
Collapse
Affiliation(s)
- Weiwei Shi
- Dept. of Periodontology, Oral Medicine and Oral Surgery, Institute for Dental and Craniofacial Sciences, Charité - University Medicine Berlin, Berlin, Germany
| | - Jiahui Song
- Dept. of Periodontology, Oral Medicine and Oral Surgery, Institute for Dental and Craniofacial Sciences, Charité - University Medicine Berlin, Berlin, Germany
| | - January Mikolaj Weiner
- Dept. of Periodontology, Oral Medicine and Oral Surgery, Institute for Dental and Craniofacial Sciences, Charité - University Medicine Berlin, Berlin, Germany
| | - Avneesh Chopra
- Dept. of Periodontology, Oral Medicine and Oral Surgery, Institute for Dental and Craniofacial Sciences, Charité - University Medicine Berlin, Berlin, Germany
| | - Henrik Dommisch
- Dept. of Periodontology, Oral Medicine and Oral Surgery, Institute for Dental and Craniofacial Sciences, Charité - University Medicine Berlin, Berlin, Germany
| | - Dieter Beule
- Core Unit Bioinformatics, Berlin Institute of Health at Charité, Berlin, Germany
| | - Arne S Schaefer
- Dept. of Periodontology, Oral Medicine and Oral Surgery, Institute for Dental and Craniofacial Sciences, Charité - University Medicine Berlin, Berlin, Germany.
| |
Collapse
|
48
|
Voorhies K, Young K, Hsu FC, Palmer ND, McDonald MLN, Lee S, Hahn G, Hecker J, Prokopenko D, Wu AC, Regan EA, DeMeo D, Kinney GL, Crapo JD, Cho MH, Silverman EK, Lange C, Budoff MJ, Hokanson JE, Lutz SM. Association of PHACTR1 with Coronary Artery Calcium Differs by Sex and Cigarette Smoking. J Cardiovasc Dev Dis 2024; 11:194. [PMID: 39057616 PMCID: PMC11276683 DOI: 10.3390/jcdd11070194] [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: 05/29/2024] [Revised: 06/22/2024] [Accepted: 06/24/2024] [Indexed: 07/28/2024] Open
Abstract
Background: Coronary artery calcium (CAC) is a marker of subclinical atherosclerosis and is a complex heritable trait with both genetic and environmental risk factors, including sex and smoking. Methods: We performed genome-wide association (GWA) analyses for CAC among all participants and stratified by sex in the COPDGene study (n = 6144 participants of European ancestry and n = 2589 participants of African ancestry) with replication in the Diabetes Heart Study (DHS). We adjusted for age, sex, current smoking status, BMI, diabetes, self-reported high blood pressure, self-reported high cholesterol, and genetic ancestry (as summarized by principal components computed within each racial group). For the significant signals from the GWA analyses, we examined the single nucleotide polymorphism (SNP) by sex interactions, stratified by smoking status (current vs. former), and tested for a SNP by smoking status interaction on CAC. Results: We identified genome-wide significant associations for CAC in the chromosome 9p21 region [CDKN2B-AS1] among all COPDGene participants (p = 7.1 × 10-14) and among males (p = 1.0 × 10-9), but the signal was not genome-wide significant among females (p = 6.4 × 10-6). For the sex stratified GWA analyses among females, the chromosome 6p24 region [PHACTR1] had a genome-wide significant association (p = 4.4 × 10-8) with CAC, but this signal was not genome-wide significant among all COPDGene participants (p = 1.7 × 10-7) or males (p = 0.03). There was a significant interaction for the SNP rs9349379 in PHACTR1 with sex (p = 0.02), but the interaction was not significant for the SNP rs10757272 in CDKN2B-AS1 with sex (p = 0.21). In addition, PHACTR1 had a stronger association with CAC among current smokers (p = 6.2 × 10-7) than former smokers (p = 7.5 × 10-3) and the SNP by smoking status interaction was marginally significant (p = 0.03). CDKN2B-AS1 had a strong association with CAC among both former (p = 7.7 × 10-8) and current smokers (p = 1.7 × 10-7) and the SNP by smoking status interaction was not significant (p = 0.40). Conclusions: Among current and former smokers of European ancestry in the COPDGene study, we identified a genome-wide significant association in the chromosome 6p24 region [PHACTR1] with CAC among females, but not among males. This region had a significant SNP by sex and SNP by smoking interaction on CAC.
Collapse
Affiliation(s)
- Kirsten Voorhies
- Department of Population Medicine, Harvard Pilgrim Health Care Institute, Boston, MA 02115, USA
| | - Kendra Young
- Department of Epidemiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Fang-Chi Hsu
- Department of Biostatistics and Data Science, Division of Public Health Sciences, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA
| | - Nicholette D. Palmer
- Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA
| | - Merry-Lynn N. McDonald
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35212, USA
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Sanghun Lee
- Division of Medicine, Department of Medical Consilience, Graduate School, Dankook University, Yongin 16890, Republic of Korea
| | - Georg Hahn
- Brigham and Women’s Hospital, Division of Pharmacoepidemiology and Pharmacoeconomics, and Department of Medicine, Harvard Medical School, Boston, MA 02120, USA
| | - Julian Hecker
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Dmitry Prokopenko
- Genetics and Aging Research Unit and the McCance Center for Brain Health, Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Ann Chen Wu
- Department of Population Medicine, Harvard Pilgrim Health Care Institute, Boston, MA 02115, USA
| | | | - Dawn DeMeo
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Greg L. Kinney
- Department of Epidemiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - James D. Crapo
- Department of Medicine, National Jewish Health, Denver, CO 80206, USA
| | - Michael H. Cho
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Edwin K. Silverman
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Christoph Lange
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Matthew J. Budoff
- Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - John E. Hokanson
- Department of Epidemiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Sharon M. Lutz
- Department of Population Medicine, Harvard Pilgrim Health Care Institute, Boston, MA 02115, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| |
Collapse
|
49
|
Pietruszyńska-Reszetarska A, Pietruszyński R, Irzmański R. The Significance of Genetically Determined Methylation and Folate Metabolism Disorders in the Pathogenesis of Coronary Artery Disease: A Target for New Therapies? Int J Mol Sci 2024; 25:6924. [PMID: 39000032 PMCID: PMC11241586 DOI: 10.3390/ijms25136924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 06/18/2024] [Accepted: 06/21/2024] [Indexed: 07/14/2024] Open
Abstract
Methylation is a biochemical process involving the addition of a methyl group (-CH3) to various chemical compounds. It plays a crucial role in maintaining the homeostasis of the endothelium, which lines the interior surface of blood vessels, and has been linked, among other conditions, to coronary artery disease (CAD). Despite significant progress in CAD diagnosis and treatment, intensive research continues into genotypic and phenotypic CAD biomarkers. This review explores the significance of the methylation pathway and folate metabolism in CAD pathogenesis, with a focus on endothelial dysfunction resulting from deficiency in the active form of folate (5-MTHF). We discuss emerging areas of research into CAD biomarkers and factors influencing the methylation process. By highlighting genetically determined methylation disorders, particularly the MTHFR polymorphism, we propose the potential use of the active form of folate (5-MTHF) as a novel CAD biomarker and personalized pharmaceutical for selected patient groups. Our aim is to improve the identification of individuals at high risk of CAD and enhance their prognosis.
Collapse
Affiliation(s)
| | - Robert Pietruszyński
- Cardiology Outpatient Clinic, Military Medical Academy Memorial Teaching Hospital of the Medical University of Lodz—Central Veterans’ Hospital, 90-549 Lodz, Poland;
| | - Robert Irzmański
- Department of Internal Medicine, Rehabilitation and Physical Medicine, Medical University of Lodz, 90-645 Lodz, Poland;
| |
Collapse
|
50
|
Mahsoub N, Almenshawy A, Taki Eldin AM, Abdel Hay NM, Youssef AR, El-Farahaty RM, El-Sayed K, Osman AM, Elhennawy ES. Association between Apo B, LDL-R and PCSK9 gene polymorphisms with coronary artery diseases in Egyptians. Mol Biol Rep 2024; 51:752. [PMID: 38874786 DOI: 10.1007/s11033-024-09607-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 05/02/2024] [Indexed: 06/15/2024]
Abstract
BACKGROUND Many studies have focused on the significance of lipid regulatory genes in the pathophysiology of Coronary artery disease (CAD). ApoB XbaI (rs693) and EcoRI (rs1042031) single nucleoid polymorphisms (SNPs) were investigated to detect whether they are risk factors for CAD. Till now, this association remains uncertain. SMARCA4 (rs1122608) SNP has directly related to dyslipidemia. Loss of function mutations (LOF) in PCSK9 result in a reduction in LDL cholesterol and are associated with protection from the development of CAD. METHODS This study was conducted on 54 CAD patients who were admitted at Internal Medicine Specialized Hospital (Cardiology Department) and 47 healthy controls. Peripheral blood samples were taken from both groups. DNA was extracted from EDTA-blood samples, then PCR- RFLP for ApoB XbaI (rs693) and EcoRI (rs1042031), SMARCA4 (rs1122608) and PCSK9 (rs505151) SNPs was done. RESULTS No statistically significant difference was found between patients and controls as regard EcoRI SNP. XbaI (rs693) X + X + genotype was significantly higher in control group (P = 0.0355). SMARCA4 (TT, GT + TT) genotypes, and T allele (P < 0.001); PCSK9 AG genotype and G allele (P = 0.027 and 0.032 respectively) were more frequent in CAD patients than controls. CONCLUSION SMARCA4 (rs1122608) and PCSK9 (rs505151) SNPs are significantly accompanying with the risk of CAD development in the Egyptian population. X + X + genotype appeared to have a protective effect against CAD. However, no observed association between EcoRI (rs1042031) and the risk of CAD development was found.
Collapse
Affiliation(s)
- Nancy Mahsoub
- Department of Clinical Pathology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Ahmed Almenshawy
- Department of Clinical Pathology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | | | - Nehal M Abdel Hay
- Department of Clinical Pathology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Amany R Youssef
- Department of Clinical Pathology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Reham M El-Farahaty
- Department of Clinical Pathology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Kefaya El-Sayed
- Department of Clinical Pathology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Adel Mohamad Osman
- Department of Cardiology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Eman S Elhennawy
- Department of Clinical Pathology, Faculty of Medicine, Mansoura University, Mansoura, Egypt.
| |
Collapse
|