1
|
Annink ME, Janssen ES, Reeskamp LF. Effectiveness of cascade screening for elevated lipoprotein(a), an underdiagnosed family disorder. Curr Opin Lipidol 2024; 35:290-296. [PMID: 39259684 PMCID: PMC11540276 DOI: 10.1097/mol.0000000000000951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
PURPOSE OF REVIEW Elevated lipoprotein(a) [Lp(a)] is a prevalent, independent, genetic risk factor for cardiovascular disease. Though crucial for adequate risk assessment, detection of individuals at increased risk because of elevated Lp(a) is severely lacking in practice. In this light, several consensus statements have recommended familial cascade screening strategies to increase detection of elevated Lp(a). This review aims to synthesize findings from recent research into the effectiveness of cascade screening for elevated Lp(a). RECENT FINDINGS Cascade screening is an effective method for identifying individuals with elevated Lp(a) and is superior to opportunistic screening. Cascade screening identifies approximately one new case of elevated Lp(a) ≥ 125 nmol/L for every two first-degree relatives screened. The number needed to screen (NNS) ranged from 1.3 to 2.9, depending on Lp(a) threshold values and selected population. SUMMARY Cascade screening appears to be a promising strategy for identifying individuals with elevated Lp(a). However, several challenges persist regarding the implementation of this strategy in clinical practice. Deciding on threshold values for initiating cascade screening, considering the implications of ethnicity-related variability of Lp(a) levels, and further research into the clinical relevance of cascade screening are crucial steps. Understanding these factors will be essential for optimizing cascade screening protocols and enhancing its effectiveness in clinical practice.
Collapse
Affiliation(s)
- Maxim E Annink
- Department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | | | | |
Collapse
|
2
|
Annink ME, Boekholdt SM, Stroes ESG. Calcific Aortic Valve Disease: Lp(a) Takes the Heat, But Is OxPL Really Fanning the Flames? J Am Coll Cardiol 2024:S0735-1097(24)10048-4. [PMID: 39545904 DOI: 10.1016/j.jacc.2024.09.1240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 09/03/2024] [Indexed: 11/17/2024]
Affiliation(s)
- Maxim E Annink
- Department of Vascular Medicine, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - S Matthijs Boekholdt
- Department of Cardiology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Erik S G Stroes
- Department of Vascular Medicine, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
3
|
Björnson E, Adiels M, Borén J, Packard CJ. Lipoprotein(a) is a highly atherogenic lipoprotein: pathophysiological basis and clinical implications. Curr Opin Cardiol 2024; 39:503-510. [PMID: 39360655 DOI: 10.1097/hco.0000000000001170] [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: 10/04/2024]
Abstract
PURPOSE OF REVIEW Lipoprotein(a) has been identified as a causal risk factor for atherosclerotic cardiovascular disease (ASCVD) and aortic valve stenosis. However, as reviewed here, there is ongoing debate as to the key pathogenic features of Lp(a) particles and the degree of Lp(a) atherogenicity relative to low-density lipoprotein (LDL). RECENT FINDINGS Genetic analyses have revealed that Lp(a) on a per-particle basis is markedly (about six-fold) more atherogenic than LDL. Oxidized phospholipids carried on Lp(a) have been found to have substantial pro-inflammatory properties triggering pathways that may contribute to atherogenesis. Whether the strength of association of Lp(a) with ASCVD risk is dependent on inflammatory status is a matter of current debate and is critical to implementing intervention strategies. Contradictory reports continue to appear, but most recent studies in large cohorts indicate that the relationship of Lp(a) to risk is independent of C-reactive protein level. SUMMARY Lp(a) is a highly atherogenic lipoprotein and a viable target for intervention in a significant proportion of the general population. Better understanding the basis of its enhanced atherogenicity is important for risk assessment and interpreting intervention trials.
Collapse
Affiliation(s)
| | - Martin Adiels
- Department of Molecular and Clinical Medicine
- School of Public Health and Community Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Jan Borén
- Department of Molecular and Clinical Medicine
| | - Chris J Packard
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
4
|
Bhatia HS, Dweck MR, Craig N, Capoulade R, Pibarot P, Trainor PJ, Whelton SP, Rikhi R, Lidani KCF, Post WS, Tsai MY, Criqui MH, Shapiro MD, Budoff MJ, DeFilippis AP, Thanassoulis G, Tsimikas S. Oxidized Phospholipids and Calcific Aortic Valvular Disease. J Am Coll Cardiol 2024:S0735-1097(24)08460-2. [PMID: 39545902 DOI: 10.1016/j.jacc.2024.08.070] [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: 04/30/2024] [Revised: 08/12/2024] [Accepted: 08/15/2024] [Indexed: 11/17/2024]
Abstract
BACKGROUND Oxidized phospholipids (OxPLs) are carried by apolipoprotein B-100-containing lipoproteins (OxPL-apoB) including lipoprotein(a) (Lp[a]). Both OxPL-apoB and Lp(a) have been associated with calcific aortic valve disease (CAVD). OBJECTIVES This study aimed to evaluate the associations between OxPL-apoB, Lp(a) and the prevalence, incidence, and progression of CAVD. METHODS OxPL-apoB and Lp(a) were evaluated in MESA (Multi-Ethnic Study of Atherosclerosis) and a participant-level meta-analysis of 4 randomized trials of participants with established aortic stenosis (AS). In MESA, the association of OxPL-apoB and Lp(a) with aortic valve calcium (AVC) at baseline and 9.5 years was evaluated using multivariable ordinal regression models. In the meta-analysis, the association between OxPL-apoB and Lp(a) with AS progression (annualized change in peak aortic valve jet velocity) was evaluated using multivariable linear regression models. RESULTS In MESA, both OxPL-apoB and Lp(a) were associated with prevalent AVC (OR per SD: 1.19 [95% CI: 1.07-1.32] and 1.13 [95% CI: 1.01-1.27], respectively) with a significant interaction between the two (P < 0.01). Both OxPL-apoB and Lp(a) were associated with incident AVC at 9.5 years when evaluated individually (interaction P < 0.01). The OxPL-apoB∗Lp(a) interaction demonstrated higher odds of prevalent and incident AVC for OxPL-apoB with increasing Lp(a) levels. In the meta-analysis, when analyzed separately, both OxPL-apoB and Lp(a) were associated with faster increase in peak aortic valve jet velocity, but when evaluated together, only OxPL-apoB remained significant (ß: 0.07; 95% CI: 0.01-0.12). CONCLUSIONS OxPL-apoB is a predictor of the presence, incidence, and progression of AVC and established AS, particularly in the setting of elevated Lp(a) levels, and may represent a novel therapeutic target for CAVD.
Collapse
Affiliation(s)
- Harpreet S Bhatia
- Division of Cardiology, Department of Medicine, University of California-San Diego, La Jolla, California, USA
| | - Marc R Dweck
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Neil Craig
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Romain Capoulade
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes, France
| | - Philippe Pibarot
- Department of Cardiology, Institut universitaire de cardiologie et de pneumologie de Québec, Laval University, Québec, Québec, Canada
| | - Patrick J Trainor
- Department of Chemistry and Biochemistry, New Mexico State University, Las Cruces, New Mexico, USA
| | - Seamus P Whelton
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Rishi Rikhi
- Section of Cardiovascular Medicine, Department of Internal Medicine, Wake Forest University School of Medicine, Winston Salem, North Carolina, USA
| | - Karita C F Lidani
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Wendy S Post
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Michael Y Tsai
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Michael H Criqui
- Division of Cardiology, Department of Medicine, University of California-San Diego, La Jolla, California, USA; Division of Preventive Medicine, Department of Family Medicine, University of California-San Diego, La Jolla, California, USA
| | - Michael D Shapiro
- Section of Cardiovascular Medicine, Department of Internal Medicine, Wake Forest University School of Medicine, Winston Salem, North Carolina, USA
| | - Matthew J Budoff
- Division of Cardiology, Lundquist Institute at Harbor-UCLA Medical Center, Torrance, California, USA
| | - Andrew P DeFilippis
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - George Thanassoulis
- Department of Medicine, Division of Experimental Medicine, McGill University Health Center, Montreal, Québec, Canada
| | - Sotirios Tsimikas
- Division of Cardiology, Department of Medicine, University of California-San Diego, La Jolla, California, USA.
| |
Collapse
|
5
|
Boffa MB, Koschinsky ML. Lipoprotein(a) and cardiovascular disease. Biochem J 2024; 481:1277-1296. [PMID: 39302109 PMCID: PMC11555715 DOI: 10.1042/bcj20240037] [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: 07/03/2024] [Revised: 08/28/2024] [Accepted: 08/28/2024] [Indexed: 09/22/2024]
Abstract
Elevated plasma levels of lipoprotein(a) (Lp(a)) are a prevalent, independent, and causal risk factor for atherosclerotic cardiovascular disease and calcific aortic valve disease. Lp(a) consists of a lipoprotein particle resembling low density lipoprotein and the covalently-attached glycoprotein apolipoprotein(a) (apo(a)). Novel therapeutics that specifically and potently lower Lp(a) levels are currently in advanced stages of clinical development, including in large, phase 3 cardiovascular outcomes trials. However, fundamental unanswered questions remain concerning some key aspects of Lp(a) biosynthesis and catabolism as well as the true pathogenic mechanisms of the particle. In this review, we describe the salient biochemical features of Lp(a) and apo(a) and how they underlie the disease-causing potential of Lp(a), the factors that determine plasma Lp(a) concentrations, and the mechanism of action of Lp(a)-lowering drugs.
Collapse
Affiliation(s)
- Michael B. Boffa
- Department of Biochemistry, University of Western Ontario, London, Ontario, Canada
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
| | - Marlys L. Koschinsky
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
6
|
Lian PA, Zhu WQ, Zhao WX, Huang PP, Ran JL, Tang YX, Huang XS, Li R. Lipoprotein(a) in atherosclerotic cardiovascular disease and proprotein convertase subtilisin/kexin-type 9 inhibitors. Clin Chim Acta 2024; 565:119982. [PMID: 39366516 DOI: 10.1016/j.cca.2024.119982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 09/28/2024] [Accepted: 09/29/2024] [Indexed: 10/06/2024]
Abstract
High plasma lipoprotein(a) (Lp(a)) levels increase the cardiovascular risk in populations with atherosclerotic cardiovascular disease (ASCVD). Apolipoprotein (a) [apo(a)], a unique protein component of Lp(a), plays an important role in the pathogenesis of atherosclerosis. Statins, the primary medication in managing ASCVD, lower low-density lipoprotein cholesterol (LDL-C) but concurrently elevate plasma Lp(a) levels, contributing to an increased residual cardiovascular risk. In turn, proprotein convertase subtilisin/kexin-type 9 (PCSK9) inhibitors, a novel class of LDL-C lowering drugs, effectively reduce plasma Lp(a) levels, which is believed to decrease residual cardiovascular risk. However, the mechanism by which PCSK9 inhibitors reduce Lp(a) levels remains unknown. In addition, there are some clinical limitations of PCSK9 inhibitors. Here, we systematically review the past, present, and prospects of studies pertaining to Lp(a), PCSK9 inhibitors, and ASCVD.
Collapse
Affiliation(s)
- Ping-An Lian
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wen-Qiang Zhu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wei-Xin Zhao
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Piao-Piao Huang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Juan-Li Ran
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ya-Xin Tang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xian-Sheng Huang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Cardiovascular Medicine, Guilin Hospital of The Second Xiangya Hospital, Central South University, Guilin, China
| | - Rong Li
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Stomatology, Guilin Hospital of The Second Xiangya Hospital, Central South University, Guilin, China.
| |
Collapse
|
7
|
Baragetti A, Da Dalt L, Norata GD. New insights into the therapeutic options to lower lipoprotein(a). Eur J Clin Invest 2024; 54:e14254. [PMID: 38778431 DOI: 10.1111/eci.14254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 05/04/2024] [Accepted: 05/05/2024] [Indexed: 05/25/2024]
Abstract
BACKGROUND Elevated levels of lipoprotein(a) [Lp(a)] represent a risk factor for cardiovascular disease including aortic valve stenosis, myocardial infarction and stroke. While the patho-physiological mechanisms linking Lp(a) with atherosclerosis are not fully understood, from genetic studies that lower Lp(a) levels protect from CVD independently of other risk factors including lipids and lipoproteins. Hereby, Lp(a) has been considered an appealing pharmacological target. RESULTS However, approved lipid lowering therapies such as statins, ezetimibe or PCSK9 inhibitors have a neutral to modest effect on Lp(a) levels, thus prompting the development of new strategies selectively targeting Lp(a). These include antisense oligonucleotides and small interfering RNAs (siRNAs) directed towards apolipoprotein(a) [Apo(a)], which are in advanced phase of clinical development. More recently, additional approaches including inhibitors of Apo(a) and gene editing approaches via CRISPR-Cas9 technology entered early clinical development. CONCLUSION If the results from the cardiovascular outcome trials, designed to demonstrate whether the reduction of Lp(a) of more than 80% as observed with pelacarsen, olpasiran or lepodisiran translates into the decrease of cardiovascular mortality and major adverse cardiovascular events, will be positive, lowering Lp(a) will become a new additional target in the management of patients with elevated cardiovascular risk.
Collapse
Grants
- RF-2019-12370896 Ministero Della Salute, Ricerca Finalizzata
- Ministero Dell'Università e Della Ricerca, CARDINNOV, ERA4 Health, GAN°101095426, the EU Horizon Europe Research and Innovation Programe
- PRIN-PNRRR2022P202294PHK Ministero Dell'Università e Della Ricerca, Progetti di Rilevante Interesse Nazionale
- PRIN2022KTSAT Ministero Dell'Università e Della Ricerca, Progetti di Rilevante Interesse Nazionale
- NANOKOSEUROPEAID/173691/DD/ACT/XK European Commission
- Ministero Dell'Università e Della Ricerca, Progetti di Rilevante Interesse Nazionale PNRR Missione 4, Progetto CN3-National Center for Gene Therpay and Drugs based on RNA Technology
- Ministero Dell'Università e Della Ricerca, Progetti di Rilevante Interesse Nazionale, MUSA-Multilayered Urban Sustainabiliy Action
- PNRR-MAD-2022-12375913 Ministero Dell'Università e Della Ricerca, Progetti di Rilevante Interesse Nazionale
Collapse
Affiliation(s)
- A Baragetti
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università Degli Studi di Milano, Milano, Italy
| | - L Da Dalt
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università Degli Studi di Milano, Milano, Italy
| | - G D Norata
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università Degli Studi di Milano, Milano, Italy
| |
Collapse
|
8
|
Qin T, Ma TY, Huang K, Lu SJ, Zhong JH, Li JJ. Lipoprotein (a)-Related Inflammatory Imbalance: A Novel Horizon for the Development of Atherosclerosis. Curr Atheroscler Rep 2024; 26:383-394. [PMID: 38878139 PMCID: PMC11236888 DOI: 10.1007/s11883-024-01215-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/15/2024] [Indexed: 07/11/2024]
Abstract
PURPOSE OF REVIEW The primary objective of this review is to explore the pathophysiological roles and clinical implications of lipoprotein(a) [Lp(a)] in the context of atherosclerotic cardiovascular disease (ASCVD). We seek to understand how Lp(a) contributes to inflammation and arteriosclerosis, aiming to provide new insights into the mechanisms of ASCVD progression. RECENT FINDINGS Recent research highlights Lp(a) as an independent risk factor for ASCVD. Studies show that Lp(a) not only promotes the inflammatory processes but also interacts with various cellular components, leading to endothelial dysfunction and smooth muscle cell proliferation. The dual role of Lp(a) in both instigating and, under certain conditions, mitigating inflammation is particularly noteworthy. This review finds that Lp(a) plays a complex role in the development of ASCVD through its involvement in inflammatory pathways. The interplay between Lp(a) levels and inflammatory responses highlights its potential as a target for therapeutic intervention. These insights could pave the way for novel approaches in managing and preventing ASCVD, urging further investigation into Lp(a) as a therapeutic target.
Collapse
Affiliation(s)
- Ting Qin
- Department of Cardiology, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Hainan, 570208, China
| | - Tian-Yi Ma
- Department of Cardiology, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Hainan, 570208, China
| | - Kang Huang
- Department of Cardiology, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Hainan, 570208, China
| | - Shi-Juan Lu
- Department of Cardiology, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Hainan, 570208, China.
| | - Jiang-Hua Zhong
- Department of Cardiology, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Hainan, 570208, China.
| | - Jian-Jun Li
- Cadiometabolic Center, State Key Laboratory of Cardiovascular Diseases, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China.
| |
Collapse
|
9
|
Tamehri Zadeh SS, Shapiro MD. Therapeutic Gene Editing in Dyslipidemias. Rev Cardiovasc Med 2024; 25:286. [PMID: 39228490 PMCID: PMC11367006 DOI: 10.31083/j.rcm2508286] [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: 02/06/2024] [Revised: 04/21/2024] [Accepted: 04/26/2024] [Indexed: 09/05/2024] Open
Abstract
Dyslipidemia, characterized by abnormal lipid levels in the blood, significantly escalates the risk of atherosclerotic cardiovascular disease and requires effective treatment strategies. While existing therapies can be effective, long-term adherence is often challenging. There has been an interest in developing enduring and more efficient solutions. In this context, gene editing, particularly clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) technology, emerges as a groundbreaking approach, offering potential long-term control of dyslipidemia by directly modifying gene expression. This review delves into the mechanistic insights of various gene-editing tools. We comprehensively analyze various pre-clinical and clinical studies, evaluating the safety, efficacy, and therapeutic implications of gene editing in dyslipidemia management. Key genetic targets, such as low-density lipoprotein receptor (LDLR), proprotein convertase subtilisin/kexin type 9 (PCSK9), angiopoietin-like protein 3 (ANGPTL3), apolipoprotein C3 (APOC3), and lipoprotein (a) (Lp(a)), known for their pivotal roles in lipid metabolism, are scrutinized. The paper highlights the promising outcomes of gene editing in achieving sustained lipid homeostasis, discusses the challenges and ethical considerations in genome editing, and envisions the future of gene therapy in revolutionizing dyslipidemia treatment and cardiovascular risk reduction.
Collapse
Affiliation(s)
- Seyed Saeed Tamehri Zadeh
- Prevention of Metabolic Disorders Research Center, Research Institute for
Endocrine Sciences, Shahid Beheshti University of Medical Sciences, 19395-4763
Tehran, Iran
| | - Michael D. Shapiro
- Center for Prevention of Cardiovascular Disease, Section on Cardiovascular
Medicine, Wake Forest University School of Medicine, Winston Salem, NC 25157, USA
| |
Collapse
|
10
|
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
|
11
|
Gać P, Jaworski A, Grajnert F, Kicman K, Trejtowicz-Sutor A, Witkowski K, Poręba M, Poręba R. Aortic Valve Calcium Score: Applications in Clinical Practice and Scientific Research-A Narrative Review. J Clin Med 2024; 13:4064. [PMID: 39064103 PMCID: PMC11277735 DOI: 10.3390/jcm13144064] [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: 05/25/2024] [Revised: 06/29/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
In this narrative review, we investigate the essential role played by the computed tomography Aortic Valve Calcium Score (AVCS) in the cardiovascular diagnostic landscape, with a special focus on its implications for clinical practice and scientific research. Calcific aortic valve stenosis is the most prevalent type of aortic stenosis (AS) in industrialized countries, and due to the aging population, its prevalence is increasing. While transthoracic echocardiography (TTE) remains the gold standard, AVCS stands out as an essential complementary tool in evaluating patients with AS. The advantage of AVCS is its independence from flow; this allows for a more precise evaluation of patients with discordant findings in TTE. Further clinical applications of AVCS include in the assessment of patients before transcatheter aortic valve replacement (TAVR), as it helps in predicting outcomes and provides prognostic information post-TAVR. Additionally, we describe different AVCS thresholds regarding gender and the anatomical variations of the aortic valve. Finally, we discuss various scientific studies where AVCS was applied. As AVCS has some limitations, due to the pathophysiologies of AS extending beyond calcification and gender differences, scientists strive to validate contrast-enhanced AVCS. Furthermore, research on developing radiation-free methods of measuring calcium content is ongoing.
Collapse
Affiliation(s)
- Paweł Gać
- Centre of Diagnostic Imaging, 4th Military Hospital, Rudolfa Weigla 5, 50-981 Wrocław, Poland; (P.G.); (A.T.-S.); (K.W.)
- Department of Population Health, Division of Environmental Health and Occupational Medicine, Wroclaw Medical University, J. Mikulicza-Radeckiego 7, 50-345 Wrocław, Poland
| | - Arkadiusz Jaworski
- Healthcare Team “County Hospital” in Sochaczew, Batalionow Chlopskich 3/7, 96-500 Sochaczew, Poland
| | - Filip Grajnert
- 4th Military Hospital, Rudolfa Weigla 5, 50-981 Wrocław, Poland;
| | - Katarzyna Kicman
- Healthcare Team “County Hospital” in Sochaczew, Batalionow Chlopskich 3/7, 96-500 Sochaczew, Poland
| | - Agnieszka Trejtowicz-Sutor
- Centre of Diagnostic Imaging, 4th Military Hospital, Rudolfa Weigla 5, 50-981 Wrocław, Poland; (P.G.); (A.T.-S.); (K.W.)
| | - Konrad Witkowski
- Centre of Diagnostic Imaging, 4th Military Hospital, Rudolfa Weigla 5, 50-981 Wrocław, Poland; (P.G.); (A.T.-S.); (K.W.)
| | - Małgorzata Poręba
- Department of Paralympic Sports, Wroclaw University of Health and Sport Sciences, Witelona 25a, 51-617 Wrocław, Poland
| | - Rafał Poręba
- Department of Internal and Occupational Diseases, Hypertension and Clinical Oncology, Wroclaw Medical University, Borowska 213, 50-556 Wrocław, Poland;
| |
Collapse
|
12
|
Tsimikas S. Lipoprotein(a) in the Year 2024: A Look Back and a Look Ahead. Arterioscler Thromb Vasc Biol 2024; 44:1485-1490. [PMID: 38924439 PMCID: PMC11210685 DOI: 10.1161/atvbaha.124.319483] [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: 06/28/2024]
Abstract
In fitting with the American Heart Association’s 100th anniversary of its founding and Arteriosclerosis, Thrombosis and Vascular Biology organizing a Centennial Collection to celebrate this event, lipoprotein(a) [Lp(a)] celebrates its 61st birthday in November 2024. There has been substantial progress in understanding the biology and pathophysiology of Lp(a) in the last 6 decades, including its discovery as a unique β-lipoprotein containing the pathognomonic apolipoprotein(a) moiety covalently bound to apolipoprotein B-100, its independent monogenetic association with cardiovascular disease and calcific aortic valve disease, its increased content of pro-atherogenic and pro-inflammatory of oxidized phospholipids relative to other lipoproteins and the development of RNA therapeutics to lower plasma Lp(a) levels. The validation or refutation of the “Lp(a) hypothesis”, namely that lowering plasma Lp(a) will lead to clinical benefit, is ongoing in 3 clinical outcomes trials. This essay reviews the discovery of Lp(a), summarizes the seminal pathophysiological findings since its discovery, discusses ongoing clinical trials with novel drugs and approaches, and provides a look ahead to unanswered questions.
Collapse
|
13
|
Yazdanpanah M, Yazdanpanah N, Chardoli M, Dehghan A. Role of interleukin 6 signaling pathway in the anti-inflammatory effects of statins on coronary artery disease: Evidence from Mendelian randomization analysis. Int J Cardiol 2024; 406:131964. [PMID: 38513732 DOI: 10.1016/j.ijcard.2024.131964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 12/08/2023] [Accepted: 03/10/2024] [Indexed: 03/23/2024]
Abstract
BACKGROUND Statins are currently widely used in the prevention of coronary artery disease (CAD) primarily for lipid-lowering with a potential anti-inflammatory effect. However, it is not clear if their potential anti-inflammatory effects are mediated through the interleukin 6 (IL-6) signaling pathway. METHODS Using the Mendelian randomization (MR) approach followed by multivariable MR analyses, we examined the extent to which the effects of statins on CAD might be mediated through the IL-6 signaling pathway. RESULTS Our observations showed that HMG-CoA reductase, using LDL levels as a proxy, had a significant effect on upstream IL-6 (βMR = 0.47, P-IVW = 0.01) and nominally significant effects on IL-6RA (βMR = 0.22, P-IVW = 0.047) and APOB (βMR = 0.82, P-IVW = 1.8 × 10-33). While the IL-6 signaling cascade (IL-6RA βMR = -0.06, P-IVW = 3.45 × 10-20 and IL-6 βMR = -0.03, P-IVW = 0.09) and the anti-inflammatory effect of HMG-CoA reductase (βMR = -0.31, P-IVW = 0.01) was found to influence the risk of CAD, the multivariable MR (MVMR) model indicated that the anti-inflammatory effect of HMG-CoA reductase is not likely to be mediated through the IL-6 signaling cascade, including APOB and IL-6RA (MVMRβ = 0.23, P = 0.688). CONCLUSIONS Our findings suggest that statins may use inflammatory mechanisms independent of the IL-6 signaling pathway to prevent CAD. This result could potentially affect the definition of the target population for statin use.
Collapse
Affiliation(s)
- Mojgan Yazdanpanah
- Department of Emergency Medicine, Firoozgar Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Nahid Yazdanpanah
- Department of Emergency Medicine, Firoozgar Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Mojtaba Chardoli
- Department of Emergency Medicine, Firoozgar Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Abbas Dehghan
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK.
| |
Collapse
|
14
|
Bhatia HS, Becker RC, Leibundgut G, Patel M, Lacaze P, Tonkin A, Narula J, Tsimikas S. Lipoprotein(a), platelet function and cardiovascular disease. Nat Rev Cardiol 2024; 21:299-311. [PMID: 37938756 PMCID: PMC11216952 DOI: 10.1038/s41569-023-00947-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/10/2023] [Indexed: 11/09/2023]
Abstract
Lipoprotein(a) (Lp(a)) is associated with atherothrombosis through several mechanisms, including putative antifibrinolytic properties. However, genetic association studies have not demonstrated an association between high plasma levels of Lp(a) and the risk of venous thromboembolism, and studies in patients with highly elevated Lp(a) levels have shown that Lp(a) lowering does not modify the clotting properties of plasma ex vivo. Lp(a) can interact with several platelet receptors, providing biological plausibility for a pro-aggregatory effect. Observational clinical studies suggest that elevated plasma Lp(a) concentrations are associated with worse long-term outcomes in patients undergoing revascularization. Furthermore, in these patients, those with elevated plasma Lp(a) levels derive more benefit from prolonged dual antiplatelet therapy than those with normal Lp(a) levels. The ASPREE trial in healthy older individuals treated with aspirin showed a reduction in ischaemic events in those who had a single-nucleotide polymorphism in LPA that is associated with elevated Lp(a) levels in plasma, without an increase in bleeding events. In this Review, we re-examine the role of Lp(a) in the regulation of platelet function and suggest areas of research to define further the clinical relevance to cardiovascular disease of the observed associations between Lp(a) and platelet function.
Collapse
Affiliation(s)
- Harpreet S Bhatia
- Division of Cardiovascular Medicine, Sulpizio Cardiovascular Center, University of California San Diego, La Jolla, CA, USA
| | - Richard C Becker
- Heart, Lung and Vascular Institute, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Gregor Leibundgut
- Division of Cardiology, University Hospital of Basel, Basel, Switzerland
| | - Mitul Patel
- Division of Cardiovascular Medicine, Sulpizio Cardiovascular Center, University of California San Diego, La Jolla, CA, USA
| | - Paul Lacaze
- Department of Epidemiology and Preventive Medicine, School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Andrew Tonkin
- Department of Epidemiology and Preventive Medicine, School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Jagat Narula
- Mount Sinai Heart, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sotirios Tsimikas
- Division of Cardiovascular Medicine, Sulpizio Cardiovascular Center, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
15
|
Thazhe Poyil NJ, Vadakkekuttical RJ, Radhakrishnan C. Correlation of periodontal inflamed surface area with glycated hemoglobin, interleukin-6 and lipoprotein(a) in type 2 diabetes with retinopathy. World J Diabetes 2024; 15:686-696. [PMID: 38680698 PMCID: PMC11045419 DOI: 10.4239/wjd.v15.i4.686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/19/2024] [Accepted: 03/01/2024] [Indexed: 04/11/2024] Open
Abstract
BACKGROUND The two-way relationship between periodontitis and type 2 diabetes mellitus (T2DM) is well established. Prolonged hyperglycemia contributes to increased periodontal destruction and severe periodontitis, accentuating diabetic complications. An inflammatory link exists between diabetic retinopathy (DR) and periodontitis, but the studies regarding this association and the role of lipoprotein(a) [Lp(a)] and interleukin-6 (IL-6) in these conditions are scarce in the literature. AIM To determine the correlation of periodontal inflamed surface area (PISA) with glycated Hb (HbA1c), serum IL-6 and Lp(a) in T2DM subjects with retinopathy. METHODS This cross-sectional study comprised 40 T2DM subjects with DR and 40 T2DM subjects without DR. All subjects were assessed for periodontal parameters [bleeding on probing (BOP), probing pocket depth, clinical attachment loss (CAL), oral hygiene index-simplified, plaque index (PI) and PISA], and systemic parameters [HbA1c, fasting plasma glucose and postprandial plasma glucose, fasting lipid profile, serum IL-6 and serum Lp(a)]. RESULTS The proportion of periodontitis in T2DM with and without DR was 47.5% and 27.5% respectively. Severity of periodontitis, CAL, PISA, IL-6 and Lp(a) were higher in T2DM with DR group compared to T2DM without DR group. Sig-nificant difference was observed in the mean percentage of sites with BOP between T2DM with DR (69%) and T2DM without DR (41%), but there was no significant difference in PI (P > 0.05). HbA1c was positively correlated with CAL (r = 0.351, P = 0.001), and PISA (r = 0.393, P ≤ 0.001) in study subjects. A positive correlation was found between PISA and IL-6 (r = 0.651, P < 0.0001); PISA and Lp(a) (r = 0.59, P < 0.001); CAL and IL-6 (r = 0.527, P < 0.0001) and CAL and Lp(a) (r = 0.631, P < 0.001) among study subjects. CONCLUSION Despite both groups having poor glycemic control and comparable plaque scores, the periodontal parameters were higher in DR as compared to T2DM without DR. Since a bidirectional link exists between periodontitis and DM, the presence of DR may have contributed to the severity of periodontal destruction and periodontitis may have influenced the progression of DR.
Collapse
Affiliation(s)
- Nusreen Jamal Thazhe Poyil
- Department of Periodontics, Government Dental College (Affiliated to Kerala University of Health Sciences), Calicut 673008, Kerala, India
| | - Rosamma Joseph Vadakkekuttical
- Department of Periodontics, Government Dental College (Affiliated to Kerala University of Health Sciences), Calicut 673008, Kerala, India
| | - Chandni Radhakrishnan
- Department of Medicine, Government Medical College (Affiliated to Kerala University of Health Sciences), Calicut 673008, Kerala, India
| |
Collapse
|
16
|
Groenen AG, Matveyenko A, Matienzo N, Halmos B, Zhang H, Westerterp M, Reyes-Soffer G. Apolipoprotein(a) production and clearance are associated with plasma IL-6 and IL-18 levels, dependent on ethnicity. Atherosclerosis 2024; 391:117474. [PMID: 38428286 DOI: 10.1016/j.atherosclerosis.2024.117474] [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: 09/19/2023] [Revised: 01/31/2024] [Accepted: 02/01/2024] [Indexed: 03/03/2024]
Abstract
BACKGROUND AND AIMS High plasma lipoprotein (a) [Lp(a)] levels are associated with increased atherosclerotic cardiovascular disease (ASCVD), in part attributed to elevated inflammation. High plasma Lp(a) levels inversely correlate with apolipoprotein (a) [(APO(a)] isoform size. APO(a) isoform size is negatively associated with APO(a) production rate (PR) and positively associated with APO(a) fractional catabolic rate (FCR). We asked whether APO(a) PR and FCR (kinetics) are associated with plasma levels of interleukin (IL)-6 and IL-18, pro-inflammatory interleukins that promote ASCVD. METHODS We used samples from existing data of APO(a) kinetic studies from an ethnically diverse cohort (n = 25: 10 Black, 9 Hispanic, and 6 White subjects) and assessed IL-6 and IL-18 plasma levels. We performed multivariate linear regression analyses to examine the relationships between predictors APO(a) PR or APO(a) FCR, and outcome variables IL-6 or IL-18. In these analyses, we adjusted for parameters known to affect Lp(a) levels and APO(a) PR and FCR, including race/ethnicity and APO(a) isoform size. RESULTS APO(a) PR and FCR were positively associated with plasma IL-6, independent of isoform size, and dependent on race/ethnicity. APO(a) PR was positively associated with plasma IL-18, independent of isoform size and race/ethnicity. APO(a) FCR was not associated with plasma IL-18. CONCLUSIONS Our studies demonstrate a relationship between APO(a) PR and FCR and plasma IL-6 or IL-18, interleukins that promote ASCVD. These studies provide new insights into Lp(a) pro-inflammatory properties and are especially relevant in view of therapies targeting APO(a) to decrease cardiovascular risk.
Collapse
Affiliation(s)
- Anouk G Groenen
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Anastasiya Matveyenko
- Columbia University Irving Medical Center, College of Physicians and Surgeons, Department of Medicine, Division of Preventive Medicine and Nutrition, New York, NY, USA
| | - Nelsa Matienzo
- Columbia University Irving Medical Center, College of Physicians and Surgeons, Department of Medicine, Division of Preventive Medicine and Nutrition, New York, NY, USA
| | - Benedek Halmos
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Hanrui Zhang
- Columbia University Irving Medical Center, Division of Cardiology, New York, NY, USA
| | - Marit Westerterp
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| | - Gissette Reyes-Soffer
- Columbia University Irving Medical Center, College of Physicians and Surgeons, Department of Medicine, Division of Preventive Medicine and Nutrition, New York, NY, USA.
| |
Collapse
|
17
|
Lu J, Wang Z, Zhang J, Jiao F, Zou C, Han L, Jiang G. Causal association of blood lipids with all-cause and cause-specific mortality risk: a Mendelian randomization study. J Lipid Res 2024; 65:100528. [PMID: 38458338 PMCID: PMC10993189 DOI: 10.1016/j.jlr.2024.100528] [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: 07/28/2023] [Revised: 02/03/2024] [Accepted: 03/02/2024] [Indexed: 03/10/2024] Open
Abstract
Dyslipidemia has long been implicated in elevating mortality risk; yet, the precise associations between lipid traits and mortality remained undisclosed. Our study aimed to explore the causal effects of lipid traits on both all-cause and cause-specific mortality. One-sample Mendelian randomization (MR) with linear and nonlinear assumptions was conducted in a cohort of 407,951 European participants from the UK Biobank. Six lipid traits, consisting of low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C), triglycerides, apolipoprotein A1 (ApoA1), apolipoprotein B (ApoB), and lipoprotein(a), were included to investigate the causal associations with mortality. Two-sample MR was performed to replicate the association between each lipid trait and all-cause mortality. Univariable MR results showed that genetically predicted higher ApoA1 was significantly associated with a decreased all-cause mortality risk (HR[95% CI]:0.93 [0.89-0.97], P value = 0.001), which was validated by the two-sample MR analysis. Higher lipoprotein(a) was associated with an increased risk of all-cause mortality (1.03 [1.01-1.04], P value = 0.002). Multivariable MR confirmed the direct causal effects of ApoA1 and lipoprotein(a) on all-cause mortality. Meanwhile, nonlinear MR found no evidence for nonlinearity between lipids and all-cause mortality. Our examination into cause-specific mortality revealed a suggestive inverse association between ApoA1 and cancer mortality, a significant positive association between lipoprotein(a) and cardiovascular disease mortality, and a suggestive positive association between lipoprotein(a) and digestive disease mortality. High LDL-C was associated with an increased risk of cardiovascular disease mortality but a decreased risk of neurodegenerative disease mortality. The findings suggest that implementing interventions to raise ApoA1 and decrease lipoprotein(a) levels may improve overall health outcomes and mitigate cancer and digestive disease mortality.
Collapse
Affiliation(s)
- Jiawen Lu
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhenqian Wang
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jiaying Zhang
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Feng Jiao
- Guangzhou Centre for Applied Mathematics, Guangzhou University, Guangzhou, Guangdong, China
| | - Chenfeng Zou
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Liyuan Han
- Department of Global Health, Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, China
| | - Guozhi Jiang
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
18
|
Laffin LJ, Nissen SE. Lp(a) - an overlooked risk factor. Trends Cardiovasc Med 2024; 34:193-199. [PMID: 36681362 DOI: 10.1016/j.tcm.2023.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 12/09/2022] [Accepted: 01/14/2023] [Indexed: 01/20/2023]
Abstract
Lipoprotein(a) (Lp(a)) is an increasingly discussed and studied risk factor for atherosclerotic cardiovascular disease and aortic valve stenosis. Many genetic and epidemiological studies support the important causal role that Lp(a) plays in the incidence of cardiovascular disease. Although dependent upon the threshold and unit of measurement of Lp(a), most estimates suggest between 20 and 30% of the world's population have elevated serum levels of Lp(a). Lp(a) levels are predominantly mediated by genetics and are not significantly modified by lifestyle interventions. Efforts are ongoing to develop effective pharmacotherapies to lower Lp(a) and to determine if lowering Lp(a) with these medications ultimately decreases the incidence of adverse cardiovascular events. In this review, the genetics and pathophysiological properties of Lp(a) will be discussed as well as the epidemiological data demonstrating its impact on the incidence of cardiovascular disease. Recommendations for screening and how to currently approach patients with elevated Lp(a) are also noted. Finally, the spectrum of pharmacotherapies under development for Lp(a) lowering is detailed.
Collapse
|
19
|
Abstract
Prolonged or excessive exposure to oxidized phospholipids (OxPLs) generates chronic inflammation. OxPLs are present in atherosclerotic lesions and can be detected in plasma on apolipoprotein B (apoB)-containing lipoproteins. When initially conceptualized, OxPL-apoB measurement in plasma was expected to reflect the concentration of minimally oxidized LDL, but, surprisingly, it correlated more strongly with plasma lipoprotein(a) (Lp(a)) levels. Indeed, experimental and clinical studies show that Lp(a) particles carry the largest fraction of OxPLs among apoB-containing lipoproteins. Plasma OxPL-apoB levels provide diagnostic information on the presence and extent of atherosclerosis and improve the prognostication of peripheral artery disease and first and recurrent myocardial infarction and stroke. The addition of OxPL-apoB measurements to traditional cardiovascular risk factors improves risk reclassification, particularly in patients in intermediate risk categories, for whom improving decision-making is most impactful. Moreover, plasma OxPL-apoB levels predict cardiovascular events with similar or greater accuracy than plasma Lp(a) levels, probably because this measurement reflects both the genetics of elevated Lp(a) levels and the generalized or localized oxidation that modifies apoB-containing lipoproteins and leads to inflammation. Plasma OxPL-apoB levels are reduced by Lp(a)-lowering therapy with antisense oligonucleotides and by lipoprotein apheresis, niacin therapy and bariatric surgery. In this Review, we discuss the role of role OxPLs in the pathophysiology of atherosclerosis and Lp(a) atherogenicity, and the use of OxPL-apoB measurement for improving prognosis, risk reclassification and therapeutic interventions.
Collapse
Affiliation(s)
- Sotirios Tsimikas
- Division of Cardiovascular Medicine, University of California San Diego, La Jolla, CA, USA.
| | - Joseph L Witztum
- Division of Endocrinology and Metabolism, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
20
|
Nakamura Y, Kulkarni NN, Takahashi T, Alimohamadi H, Dokoshi T, Liu E, Shia M, Numata T, Luo EW, Gombart AF, Yang X, Secrest P, Gordts PL, Tsimikas S, Wong GC, Gallo RL. Increased LL37 in psoriasis and other inflammatory disorders promotes LDL uptake and atherosclerosis. J Clin Invest 2024; 134:e172578. [PMID: 38194294 PMCID: PMC10904043 DOI: 10.1172/jci172578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 01/05/2024] [Indexed: 01/10/2024] Open
Abstract
Patients with chronic inflammatory disorders such as psoriasis have an increased risk of cardiovascular disease and elevated levels of LL37, a cathelicidin host defense peptide that has both antimicrobial and proinflammatory properties. To explore whether LL37 could contribute to the risk of heart disease, we examined its effects on lipoprotein metabolism and show that LL37 enhanced LDL uptake in macrophages through the LDL receptor (LDLR), scavenger receptor class B member 1 (SR-B1), and CD36. This interaction led to increased cytosolic cholesterol in macrophages and changes in expression of lipid metabolism genes consistent with increased cholesterol uptake. Structure-function analysis and synchrotron small-angle x-ray scattering showed structural determinants of the LL37-LDL complex that underlie its ability to bind its receptors and promote uptake. This function of LDL uptake is unique to cathelicidins from humans and some primates and was not observed with cathelicidins from mice or rabbits. Notably, Apoe-/- mice expressing LL37 developed larger atheroma plaques than did control mice, and a positive correlation between plasma LL37 and oxidized phospholipid on apolipoprotein B (OxPL-apoB) levels was observed in individuals with cardiovascular disease. These findings provide evidence that LDL uptake can be increased via interaction with LL37 and may explain the increased risk of cardiovascular disease associated with chronic inflammatory disorders.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Adrian F. Gombart
- Linus Pauling Institute, Department of Biochemistry and Biophysics, Oregon State University, Corvallis, Oregon, USA
| | | | - Patrick Secrest
- Department of Medicine, Division of Endocrinology and Metabolism, and
| | - Philip L.S.M. Gordts
- Department of Medicine, Division of Endocrinology and Metabolism, and
- Glycobiology Research and Training Center, UCSD, La Jolla, California, USA
| | | | - Gerard C.L. Wong
- Department of Bioengineering, UCLA, Los Angeles, California, USA
| | | |
Collapse
|
21
|
Bhatia HS, Trainor P, Carlisle S, Tsai MY, Criqui MH, DeFilippis A, Tsimikas S. Aspirin and Cardiovascular Risk in Individuals With Elevated Lipoprotein(a): The Multi-Ethnic Study of Atherosclerosis. J Am Heart Assoc 2024; 13:e033562. [PMID: 38293935 PMCID: PMC11056170 DOI: 10.1161/jaha.123.033562] [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: 11/15/2023] [Accepted: 12/15/2023] [Indexed: 02/01/2024]
Abstract
BACKGROUND Effective therapies for reducing cardiovascular disease (CVD) risk in people with elevated lipoprotein(a) are lacking, especially for primary prevention. Because of the potential association of lipoprotein(a) with thrombosis, we evaluated the relationship between aspirin use and CVD events in people with elevated lipoprotein(a). METHODS AND RESULTS We used data from the MESA (Multi-Ethnic Study of Atherosclerosis), a prospective cohort study of individuals free of baseline cardiovascular disease. Due to potential confounding by indication, we matched aspirin users to nonusers using a propensity score based on CVD risk factors. We then evaluated the association between aspirin use and coronary heart disease (CHD) events (CHD death, nonfatal myocardial infarction) stratified by baseline lipoprotein(a) level (threshold of 50 mg/dL) using Cox proportional hazards models with adjustment for CVD risk factors. After propensity matching, the study cohort included 2183 participants, including 1234 (57%) with baseline aspirin use and 423 (19%) with lipoprotein(a) >50 mg/dL. Participants with lipoprotein(a) >50 mg/dL had a higher burden of CVD risk factors, more frequent aspirin use (61.7% versus 55.3%, P=0.02), and higher rate of incident CHD events (13.7% versus 8.9%, P<0.01). Aspirin was associated with a significant reduction in CHD events among those with elevated lipoprotein(a) (hazard ratio, 0.54 [95% CI, 0.32-0.94]; P=0.03). Those with lipoprotein(a) >50 mg/dL and aspirin use had similar CHD risk as those with lipoprotein(a) ≤50 mg/dL regardless of aspirin use. CONCLUSIONS Aspirin use was associated with a significantly lower risk for CHD events in participants with lipoprotein(a) >50 mg/dL without baseline CVD. The results of this observational propensity-matched study require confirmation in studies with randomization of aspirin use.
Collapse
Affiliation(s)
- Harpreet S. Bhatia
- Division of Cardiovascular Medicine, Department of MedicineUniversity of California, San DiegoLa JollaCA
| | - Patrick Trainor
- Department of Chemistry and BiochemistryNew Mexico State UniversityLas CrucesNM
| | - Samantha Carlisle
- Department of Chemistry and BiochemistryNew Mexico State UniversityLas CrucesNM
| | - Michael Y. Tsai
- Department of Laboratory Medicine and PathologyUniversity of MinnesotaMinneapolisMN
| | - Michael H. Criqui
- Division of Cardiovascular Medicine, Department of MedicineUniversity of California, San DiegoLa JollaCA
- Division of Preventive Medicine, Department of Family MedicineUniversity of California, San DiegoLa JollaCA
| | - Andrew DeFilippis
- Division of Cardiovascular Medicine, Department of MedicineVanderbilt University Medical CenterNashvilleTN
| | - Sotirios Tsimikas
- Division of Cardiovascular Medicine, Department of MedicineUniversity of California, San DiegoLa JollaCA
| |
Collapse
|
22
|
Sosnowska B, Stepinska J, Mitkowski P, Bielecka-Dabrowa A, Bobrowska B, Budzianowski J, Burchardt P, Chlebus K, Dobrowolski P, Gasior M, Jankowski P, Kubica J, Mickiewicz A, Mysliwiec M, Osadnik T, Prejbisz A, Rajtar-Salwa R, Wita K, Witkowski A, Gil R, Banach M. Recommendations of the Experts of the Polish Cardiac Society (PCS) and the Polish Lipid Association (PoLA) on the diagnosis and management of elevated lipoprotein(a) levels. Arch Med Sci 2024; 20:8-27. [PMID: 38414479 PMCID: PMC10895977 DOI: 10.5114/aoms/183522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 01/31/2024] [Indexed: 02/29/2024] Open
Abstract
Lipoprotein(a) [Lp(a)] is made up of a low-density lipoprotein (LDL) particle and a specific apolipoprotein(a). The blood concentration of Lp(a) is approximately 90% genetically determined, and the main genetic factor determining Lp(a) levels is the size of the apo(a) isoform, which is determined by the number of KIV2 domain repeats. The size of the apo(a) isoform is inversely proportional to the blood concentration of Lp(a). Lp(a) is a strong and independent cardiovascular risk factor. Elevated Lp(a) levels ≥ 50 mg/dl (≥ 125 nmol/l) are estimated to occur in more than 1.5 billion people worldwide. However, determination of Lp(a) levels is performed far too rarely, including Poland, where, in fact, it is only since the 2021 guidelines of the Polish Lipid Association (PoLA) and five other scientific societies that Lp(a) measurements have begun to be performed. Determination of Lp(a) concentrations is not easy due to, among other things, the different sizes of the apo(a) isoforms; however, the currently available certified tests make it possible to distinguish between people with low and high cardiovascular risk with a high degree of precision. In 2022, the first guidelines for the management of patients with elevated lipoprotein(a) levels were published by the European Atherosclerosis Society (EAS) and the American Heart Association (AHA). The first Polish guidelines are the result of the work of experts from the two scientific societies and their aim is to provide clear, practical recommendations for the determination and management of elevated Lp(a) levels.
Collapse
Affiliation(s)
- Bożena Sosnowska
- Department of Preventive Cardiology and Lipidology, Medical University of Lodz (MUL), Lodz, Poland
| | | | - Przemyslaw Mitkowski
- 1 Department of Cardiology, Poznan University of Medical Sciences, Poznan, Poland
| | - Agata Bielecka-Dabrowa
- Department of Preventive Cardiology and Lipidology, Medical University of Lodz (MUL), Lodz, Poland
- Department of Cardiology and Adult Congenital Defects, Polish Mother’s Memorial Hospital Research Institute (PMMHRI), Lodz, Poland
| | - Beata Bobrowska
- Department of Clinical Cardiology and Cardiovascular Interventions, University Hospital in Krakow, Krakow, Poland
| | - Jan Budzianowski
- Department of Interventional Cardiology and Cardiac Surgery, University of Zielona Gora, Collegium Medicum, Zielona Gora, Poland
- Multidisciplinary Hospital, Nowa Sol, Poland
| | - Pawel Burchardt
- Department of Cardiology, J. Strus Hospital, Poznan, Poland
- Department of Hypertension, Angiology and Internal Medicine, Poznan University of Medical Sciences, Poznan, Poland
| | - Krzysztof Chlebus
- National Center for Familial Hypercholesterolemia, 1 Chair and Department of Cardiology, Medical University of Gdansk, Gdansk, Poland
| | - Piotr Dobrowolski
- Department of Epidemiology, Cardiovascular Disease Prevention and Health Promotion, National Institute of Cardiology, Warsaw, Poland
| | - Mariusz Gasior
- 3 Department of Cardiology, Silesian Centre for Heart Diseases, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Zabrze, Poland
| | - Piotr Jankowski
- Department of Internal Medicine and Geriatric Cardiology, Medical Centre for Postgraduate Education, Warsaw, Poland
| | - Jacek Kubica
- Department of Cardiology and Internal Medicine, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Agnieszka Mickiewicz
- Lipoprotein Apheresis Laboratory, 1 Department of Cardiology, Medical University of Gdansk, Gdansk, Poland
| | - Malgorzata Mysliwiec
- Department of Paediatrics, Diabetology and Endocrinology, Medical University of Gdansk, Gdansk, Poland
| | - Tadeusz Osadnik
- Department of Pharmacology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Katowice, Poland
| | - Aleksander Prejbisz
- Department of Epidemiology, Cardiovascular Disease Prevention and Health Promotion, National Institute of Cardiology, Warsaw, Poland
| | - Renata Rajtar-Salwa
- Department of Clinical Cardiology and Cardiovascular Interventions, University Hospital in Krakow, Krakow, Poland
| | - Kristian Wita
- 1 Department of Cardiology, School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland
| | - Adam Witkowski
- Department of Interventional Cardiology and Angiology, National Institute of Cardiology, Warsaw, Poland
| | - Robert Gil
- Department of Cardiology, National Medical Institute of the Ministry of Internal Affairs and Administration, Warsaw, Poland
| | - Maciej Banach
- Department of Preventive Cardiology and Lipidology, Medical University of Lodz (MUL), Lodz, Poland
- Department of Cardiology and Adult Congenital Defects, Polish Mother’s Memorial Hospital Research Institute (PMMHRI), Lodz, Poland
- Ciccarone Center for the Prevention of Cardiovascular Disease, Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
23
|
Noothi SK, Ahmed MR, Agrawal DK. Residual risks and evolving atherosclerotic plaques. Mol Cell Biochem 2023; 478:2629-2643. [PMID: 36897542 PMCID: PMC10627922 DOI: 10.1007/s11010-023-04689-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 02/23/2023] [Indexed: 03/11/2023]
Abstract
Atherosclerotic disease of the coronary and carotid arteries is the primary global cause of significant mortality and morbidity. The chronic occlusive diseases have changed the epidemiological landscape of health problems both in developed and the developing countries. Despite the enormous benefit of advanced revascularization techniques, use of statins, and successful attempts of targeting modifiable risk factors, like smoking and exercise in the last four decades, there is still a definite "residual risk" in the population, as evidenced by many prevalent and new cases every year. Here, we highlight the burden of the atherosclerotic diseases and provide substantial clinical evidence of the residual risks in these diseases despite advanced management settings, with emphasis on strokes and cardiovascular risks. We critically discussed the concepts and potential underlying mechanisms of the evolving atherosclerotic plaques in the coronary and carotid arteries. This has changed our understanding of the plaque biology, the progression of unstable vs stable plaques, and the evolution of plaque prior to the occurrence of a major adverse atherothrombotic event. This has been facilitated using intravascular ultrasound, optical coherence tomography, and near-infrared spectroscopy in the clinical settings to achieve surrogate end points. These techniques are now providing exquisite information on plaque size, composition, lipid volume, fibrous cap thickness and other features that were previously not possible with conventional angiography.
Collapse
Affiliation(s)
- Sunil K Noothi
- Department of Translational Research, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, USA
| | - Mohamed Radwan Ahmed
- Department of Translational Research, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, USA
| | - Devendra K Agrawal
- Department of Translational Research, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, USA.
| |
Collapse
|
24
|
Assini JM, Clark JR, Youssef A, Xing C, Doerfler AM, Park SH, Saxena L, Yaseen AB, Børen J, Gros R, Bao G, Lagor WR, Boffa MB, Koschinsky ML. High levels of lipoprotein(a) in transgenic mice exacerbate atherosclerosis and promote vulnerable plaque features in a sex-specific manner. Atherosclerosis 2023; 384:117150. [PMID: 37290980 DOI: 10.1016/j.atherosclerosis.2023.05.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/18/2023] [Accepted: 05/23/2023] [Indexed: 06/10/2023]
Abstract
BACKGROUND AND AIMS Despite increased clinical interest in lipoprotein(a) (Lp(a)), many questions remain about the molecular mechanisms by which it contributes to atherosclerotic cardiovascular disease. Existing murine transgenic (Tg) Lp(a) models are limited by low plasma levels of Lp(a) and have not consistently shown a pro-atherosclerotic effect of Lp(a). METHODS We generated Tg mice expressing both human apolipoprotein(a) (apo(a)) and human apoB-100, with pathogenic levels of plasma Lp(a) (range 87-250 mg/dL). Female and male Lp(a) Tg mice (Tg(LPA+/0;APOB+/0)) and human apoB-100-only controls (Tg(APOB+/0)) (n = 10-13/group) were fed a high-fat, high-cholesterol diet for 12 weeks, with Ldlr knocked down using an antisense oligonucleotide. FPLC was used to characterize plasma lipoprotein profiles. Plaque area and necrotic core size were quantified and immunohistochemical assessment of lesions using a variety of cellular and protein markers was performed. RESULTS Male and female Tg(LPA+/0;APOB+/0) and Tg(APOB+/0) mice exhibited proatherogenic lipoprotein profiles with increased cholesterol-rich VLDL and LDL-sized particles and no difference in plasma total cholesterol between genotypes. Complex lesions developed in the aortic sinus of all mice. Plaque area (+22%), necrotic core size (+25%), and calcified area (+65%) were all significantly increased in female Tg(LPA+/0;APOB+/0) mice compared to female Tg(APOB+/0) mice. Immunohistochemistry of lesions demonstrated that apo(a) deposited in a similar pattern as apoB-100 in Tg(LPA+/0;APOB+/0) mice. Furthermore, female Tg(LPA+/0;APOB+/0) mice exhibited less organized collagen deposition as well as 42% higher staining for oxidized phospholipids (OxPL) compared to female Tg(APOB+/0) mice. Tg(LPA+/0;APOB+/0) mice had dramatically higher levels of plasma OxPL-apo(a) and OxPL-apoB compared to Tg(APOB+/0) mice, and female Tg(LPA+/0;APOB+/0) mice had higher plasma levels of the proinflammatory cytokine MCP-1 (+3.1-fold) compared to female Tg(APOB+/0) mice. CONCLUSIONS These data suggest a pro-inflammatory phenotype exhibited by female Tg mice expressing Lp(a) that appears to contribute to the development of more severe lesions with greater vulnerable features.
Collapse
Affiliation(s)
- Julia M Assini
- Department of Biochemistry, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, N6A 5B7, Canada; Robarts Research Institute, Schulich School of Medicine & Dentistry, London, Ontario, Canada
| | - Justin R Clark
- Robarts Research Institute, Schulich School of Medicine & Dentistry, London, Ontario, Canada; Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, N6A 5B7, Canada
| | - Amer Youssef
- Robarts Research Institute, Schulich School of Medicine & Dentistry, London, Ontario, Canada
| | - Chuce Xing
- Robarts Research Institute, Schulich School of Medicine & Dentistry, London, Ontario, Canada
| | - Alexandria M Doerfler
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - So Hyun Park
- Department of Bioengineering, Rice University, Houston, USA
| | - Lavanya Saxena
- Department of Bioengineering, Rice University, Houston, USA
| | - Adam B Yaseen
- Department of Bioengineering, Rice University, Houston, USA
| | - Jan Børen
- Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Robert Gros
- Robarts Research Institute, Schulich School of Medicine & Dentistry, London, Ontario, Canada; Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, N6A 5B7, Canada
| | - Gang Bao
- Robarts Research Institute, Schulich School of Medicine & Dentistry, London, Ontario, Canada
| | - William R Lagor
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA; Department of Bioengineering, Rice University, Houston, USA
| | - Michael B Boffa
- Department of Biochemistry, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, N6A 5B7, Canada; Robarts Research Institute, Schulich School of Medicine & Dentistry, London, Ontario, Canada.
| | - Marlys L Koschinsky
- Robarts Research Institute, Schulich School of Medicine & Dentistry, London, Ontario, Canada; Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, N6A 5B7, Canada.
| |
Collapse
|
25
|
Afanasieva OI, Tyurina AV, Ezhov MV, Razova OA, Klesareva EA, Pokrovsky SN. Lipoprotein(a) and Low-Molecular-Weight Apo(a) Phenotype as Determinants of New Cardiovascular Events in Patients with Premature Coronary Heart Disease. Diseases 2023; 11:145. [PMID: 37873789 PMCID: PMC10594435 DOI: 10.3390/diseases11040145] [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: 07/01/2023] [Revised: 09/29/2023] [Accepted: 10/12/2023] [Indexed: 10/25/2023] Open
Abstract
BACKGROUND Lipoprotein(a) (Lp(a)) is a genetic risk factor of atherosclerotic cardiovascular diseases (ASCVDs). Proprotein convertase subtilisin/kexin type 9 (PCSK9) is related to vascular inflammation and detected in atherosclerotic plaques. A temporary increase in the circulating concentration of PCSK9 and Lp(a) was shown in patients with myocardial infarction (MI). The aim of this study was to evaluate the role of the apo(a) phenotype and the Lp(a) concentration as well as its complex with PCSK9 in the development of cardiac events and MI in patients with a premature manifestation of coronary heart disease (CHD). METHODS In a prospective study with retrospective data collection, we included 116 patients with premature CHD who were followed for a median of 14 years. The medical history and information on cardiovascular events after an initial exam as well as data on the levels of lipids, Lp(a), PCSK9, PCSK9-Lp(a) complex, and apo(a) phenotype were obtained. RESULTS The patients were divided into two groups depending on the presence of a low- (LMW, n = 52) or high-molecular weight (HMW, n = 64) apo(a) phenotype. LMW apo(a) phenotype (odds ratio 2.3 (1.1 to 4.8), p = 0.03), but not elevated Lp(a) (1.9 (0.8-4.6), p = 0.13), was an independent predictor for the development of MI after adjustment for sex, age of CHD debut, initial lipids levels, and lipid-lowering treatment. The apo(a) phenotype also determined the relationship between Lp(a) and PCSK9 concentrations. The level of the PCSK9-Lp(a) complex was higher in LMW apo(a) patients. CONCLUSION The LMW apo(a) phenotype is a risk factor for non-fatal MI in a long-term prospective follow-up of patients with premature CHD, and this link could be mediated via PCSK9.
Collapse
Affiliation(s)
- Olga I. Afanasieva
- Institute of Experimental Cardiology, National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Ministry of Health of the Russian Federation, Academician Chazov str., 15a, 121552 Moscow, Russia; (O.I.A.); (O.A.R.); (E.A.K.); (S.N.P.)
| | - Alexandra V. Tyurina
- A.L. Myasnikov Institute of Clinical Cardiology, National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Ministry of Health of the Russian Federation, Academician Chazov str., 15a, 121552 Moscow, Russia;
| | - Marat V. Ezhov
- A.L. Myasnikov Institute of Clinical Cardiology, National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Ministry of Health of the Russian Federation, Academician Chazov str., 15a, 121552 Moscow, Russia;
| | - Oxana A. Razova
- Institute of Experimental Cardiology, National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Ministry of Health of the Russian Federation, Academician Chazov str., 15a, 121552 Moscow, Russia; (O.I.A.); (O.A.R.); (E.A.K.); (S.N.P.)
| | - Elena A. Klesareva
- Institute of Experimental Cardiology, National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Ministry of Health of the Russian Federation, Academician Chazov str., 15a, 121552 Moscow, Russia; (O.I.A.); (O.A.R.); (E.A.K.); (S.N.P.)
| | - Sergei N. Pokrovsky
- Institute of Experimental Cardiology, National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Ministry of Health of the Russian Federation, Academician Chazov str., 15a, 121552 Moscow, Russia; (O.I.A.); (O.A.R.); (E.A.K.); (S.N.P.)
| |
Collapse
|
26
|
Chiesa G, Zenti MG, Baragetti A, Barbagallo CM, Borghi C, Colivicchi F, Maggioni AP, Noto D, Pirro M, Rivellese AA, Sampietro T, Sbrana F, Arca M, Averna M, Catapano AL. Consensus document on Lipoprotein(a) from the Italian Society for the Study of Atherosclerosis (SISA). Nutr Metab Cardiovasc Dis 2023; 33:1866-1877. [PMID: 37586921 DOI: 10.1016/j.numecd.2023.07.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/07/2023] [Accepted: 07/13/2023] [Indexed: 08/18/2023]
Abstract
AIMS In view of the consolidating evidence on the causal role of Lp(a) in cardiovascular disease, the Italian Society for the Study of Atherosclerosis (SISA) has assembled a consensus on Lp(a) genetics and epidemiology, together with recommendations for its measurement and current and emerging therapeutic approaches to reduce its plasma levels. Data on the Italian population are also provided. DATA SYNTHESIS Lp(a) is constituted by one apo(a) molecule and a lipoprotein closely resembling to a low-density lipoprotein (LDL). Its similarity with an LDL, together with its ability to carry oxidized phospholipids are considered the two main features making Lp(a) harmful for cardiovascular health. Plasma Lp(a) concentrations vary over about 1000 folds in humans and are genetically determined, thus they are quite stable in any individual. Mendelian Randomization studies have suggested a causal role of Lp(a) in atherosclerotic cardiovascular disease (ASCVD) and aortic valve stenosis and observational studies indicate a linear direct correlation between cardiovascular disease and Lp(a) plasma levels. Lp(a) measurement is strongly recommended once in a patient's lifetime, particularly in FH subjects, but also as part of the initial lipid screening to assess cardiovascular risk. The apo(a) size polymorphism represents a challenge for Lp(a) measurement in plasma, but new strategies are overcoming these difficulties. A reduction of Lp(a) levels can be currently attained only by plasma apheresis and, moderately, with PCSK9 inhibitor treatment. CONCLUSIONS Awaiting the approval of selective Lp(a)-lowering drugs, an intensive management of the other risk factors for individuals with elevated Lp(a) levels is strongly recommended.
Collapse
Affiliation(s)
- Giulia Chiesa
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università Degli Studi di Milano, Milan, Italy.
| | - Maria Grazia Zenti
- Section of Diabetes and Metabolism, Pederzoli Hospital, Peschiera Del Garda, Verona, Italy.
| | - Andrea Baragetti
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università Degli Studi di Milano, Milan, Italy; IRCCS MultiMedica, Sesto San Giovanni, Milan, Italy
| | - Carlo M Barbagallo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Claudio Borghi
- Department of Cardiovascular Medicine, IRCCS AOU S. Orsola, Bologna, Italy
| | - Furio Colivicchi
- Division of Clinical Cardiology, San Filippo Neri Hospital, Rome, Italy
| | - Aldo P Maggioni
- ANMCO Research Center, Heart Care Foundation, Firenze, Italy
| | - Davide Noto
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Matteo Pirro
- Unit of Internal Medicine, Angiology and Arteriosclerosis Diseases, Department of Medicine and Surgery, University of Perugia, Italy
| | - Angela A Rivellese
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Tiziana Sampietro
- Lipoapheresis Unit, Reference Center for Diagnosis and Treatment of Inherited Dyslipidemias, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Francesco Sbrana
- Lipoapheresis Unit, Reference Center for Diagnosis and Treatment of Inherited Dyslipidemias, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Marcello Arca
- Department of Translational and Precision Medicine (DTPM), Sapienza University of Rome, Policlinico Umberto I, Rome, Italy
| | - Maurizio Averna
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy; Institute of Biophysics, National Council of Researches, Palermo, Italy
| | - Alberico L Catapano
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università Degli Studi di Milano, Milan, Italy; IRCCS MultiMedica, Sesto San Giovanni, Milan, Italy
| |
Collapse
|
27
|
Chemello K, Gallo A, Guedon AF, Techer R, Croyal M, Swietek MJ, Meilhac O, Amarenco P, Lambert G. Lipoprotein(a): A Residual Cardiovascular Risk Factor in Statin-Treated Stroke Survivors: Insights From the SPARCL Trial. JACC. ADVANCES 2023; 2:100557. [PMID: 38939496 PMCID: PMC11198425 DOI: 10.1016/j.jacadv.2023.100557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 06/05/2023] [Accepted: 06/24/2023] [Indexed: 06/29/2024]
Abstract
Background In the SPARCL (Stroke Prevention by Aggressive Reduction in Cholesterol levels) trial, atorvastatin (80 mg/d) was compared to placebo in patients with recent stroke or transient ischemic attack (TIA) and no known coronary artery disease. Objectives This study aimed to assess the contribution of lipoprotein(a) [Lp(a)] to subsequent cerebrovascular and cardiovascular events in stroke/TIA survivors. Methods Lp(a) levels and apolipoprotein(a) [apo(a)] isoform size were determined by liquid-chromatography mass spectrometry in samples collected at baseline from 2,814 SPARCL participants (1,418 randomized to atorvastatin and 1,396 to placebo). Within each treatment arm, patients in the highest quartile (≥84.0 nmol/L) were compared with those in the lowest quartiles of Lp(a) concentrations. Patients in the lowest quartile (≤25.9 Kringle IV domains) of apo(a) isoform sizes were compared with those in the highest quartiles. Multivariable-adjusted HRs were calculated using Cox proportional regression models. Results There was no significant association between Lp(a) concentrations or apo(a) isoform sizes and the risk of recurrent stroke, the primary outcome of SPARCL, or cerebrovascular events in patients randomized to atorvastatin or placebo. In contrast, in patients randomized to atorvastatin, elevated Lp(a) concentrations and short apo(a) isoforms were positively and independently associated with an increased risk of coronary events (HR: 1.607 [95% CI: 1.007-2.563] and HR: 2.052 [95% CI: 1.303-3.232]). No such association was found in patients randomized to placebo (HR: 1.025 [95% CI: 0.675-1.555] and HR: 1.097 [95% CI: 0.735-1.637]). Conclusions Lp(a) contributes to the residual coronary artery disease risk of statin-treated stroke/TIA survivors, paving the way for use of therapies targeting Lp(a) in this population with stroke. (Lipitor In The Prevention Of Stroke, For Patients Who Have Had A Previous Stroke [SPARCL]; NCT00147602).
Collapse
Affiliation(s)
- Kévin Chemello
- Inserm, UMR1188 Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, Sainte-Pierre, France
| | - Antonio Gallo
- Inserm, UMR1188 Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, Sainte-Pierre, France
- Department of Endocrinology and Prevention of Cardiovascular Disease, Institute of Cardio Metabolism and Nutrition (ICAN), Sorbonne Université Pitié-Salpêtrière Hospital, SU-APHP, Paris, France
| | - Alexis F. Guedon
- APHP, Service de Médecine Interne, Département Hospitalo-Universitaire Inflammation Immunopathologie Biothérapie (DMUi3), Sorbonne Université, Paris, France
| | - Romuald Techer
- Inserm, UMR1188 Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, Sainte-Pierre, France
| | - Mikael Croyal
- CHU Nantes, CNRS, Inserm, BioCore, US16, SFR Bonamy, Nantes Université, Nantes, France
- CHU Nantes, CNRS, Inserm, Institut du Thorax, Nantes Université, Nantes, France
- CRNH-Ouest Mass Spectrometry Core Facility, Nantes, France
| | | | - Olivier Meilhac
- Inserm, UMR1188 Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, Sainte-Pierre, France
- CHU de La Réunion, Saint-Denis, France
| | - Pierre Amarenco
- INSERM 1148, Bichat Stroke Centre, Paris Université, Paris, France
| | - Gilles Lambert
- Inserm, UMR1188 Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, Sainte-Pierre, France
| |
Collapse
|
28
|
Kuno T, Arce J, Fattouh M, Sarkar S, Skendelas JP, Daich J, Schenone AL, Zhang L, Rodriguez CJ, Virani SS, Slomka PJ, Shaw LJ, Williamson EE, Berman DS, Garcia MJ, Dey D, Slipczuk L. Cardiometabolic predictors of high-risk CCTA phenotype in a diverse patient population. Am J Prev Cardiol 2023; 15:100578. [PMID: 37675408 PMCID: PMC10477443 DOI: 10.1016/j.ajpc.2023.100578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 08/06/2023] [Accepted: 08/13/2023] [Indexed: 09/08/2023] Open
Abstract
Introduction Low-attenuation non-calcified plaque (LAP) burden and vascular inflammation by pericoronary adipose tissue (PCAT) measured from coronary CT angiography (CCTA) have shown to be predictors of cardiovascular outcomes. We aimed to investigate the relationships of cardiometabolic risk factors including lipoprotein(a) and epicardial adipose tissue (EAT) with CCTA high-risk imaging biomarkers, LAP and vascular inflammation. Methods The patient population consisted of consecutive patients who underwent CCTA for stable chest pain and had a complete cardiometabolic panel including lipoprotein(a). Plaque, PCAT and EAT were measured from CT using semiautomated software. Elevated LAP burden and PCAT attenuation were defined as ≥4% and ≥70.5 HU, respectively. The primary clinical end-point was a composite of myocardial infarction, revascularization or cardiovascular death. Results A total of 364 consecutive patients were included (median age 56 years, 64% female); the majority of patients were of Hispanic (60%), and the rest were of non-Hispanic Black (21%), non-Hispanic White (6%) and non-Hispanic Asian (4%) race/ethnicity. The prevalence of elevated LAP burden and PCAT attenuation was 31 and 18%, respectively, while only 8% had obstructive stenosis. There were significant differences in plaque characteristics among different racial/ethnic groups (p<0.001). Lipoprotein(a) correlated with LAP burden in Hispanic patients. Patients with elevated LAP were older, more likely to be have diabetes, hypertension, hyperlipidemia and smoke with higher CAC and EAT volume (all P<0.05). Patients with elevated LAP were more likely to develop the primary clinical outcome (p<0.001) but those with elevated PCAT were not (p=0.797). Conclusion The prevalence of LAP and PCAT attenuation were 31 and 18%, respectively. Lipoprotein(a) levels correlated with LAP burden in Hispanic patients. Age, male sex, hypertension and hyperlipidemia increased the odds of elevated LAP, which showed prognostic significance.
Collapse
Affiliation(s)
- Toshiki Kuno
- Cardiology Division, Montefiore Medical Center, Montefiore Medical Center/Albert Einstein Colalege of Medicine, Cardiology Division. 111 E210th, Bronx, NY 10467, United States
| | - Javier Arce
- Cardiology Division, Montefiore Medical Center, Montefiore Medical Center/Albert Einstein Colalege of Medicine, Cardiology Division. 111 E210th, Bronx, NY 10467, United States
| | - Michael Fattouh
- Cardiology Division, Montefiore Medical Center, Montefiore Medical Center/Albert Einstein Colalege of Medicine, Cardiology Division. 111 E210th, Bronx, NY 10467, United States
| | - Sharmila Sarkar
- Cardiology Division, Montefiore Medical Center, Montefiore Medical Center/Albert Einstein Colalege of Medicine, Cardiology Division. 111 E210th, Bronx, NY 10467, United States
| | - John P Skendelas
- Cardiothoracic and Vascular Surgery Department, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, United States
| | - Jonathan Daich
- Cardiology Division, Montefiore Medical Center, Montefiore Medical Center/Albert Einstein Colalege of Medicine, Cardiology Division. 111 E210th, Bronx, NY 10467, United States
| | - Aldo L Schenone
- Cardiology Division, Montefiore Medical Center, Montefiore Medical Center/Albert Einstein Colalege of Medicine, Cardiology Division. 111 E210th, Bronx, NY 10467, United States
| | - Lili Zhang
- Cardiology Division, Montefiore Medical Center, Montefiore Medical Center/Albert Einstein Colalege of Medicine, Cardiology Division. 111 E210th, Bronx, NY 10467, United States
| | - Carlos J Rodriguez
- Cardiology Division, Montefiore Medical Center, Montefiore Medical Center/Albert Einstein Colalege of Medicine, Cardiology Division. 111 E210th, Bronx, NY 10467, United States
| | - Salim S Virani
- Office of the Vice Provost (Research), The Aga Khan University, Karachi, Pakistan
- Division of Cardiology, The Texas Heart Institute/Baylor College of Medicine, Houston, TX, United States
| | - Piotr J Slomka
- Division of Cardiology, The Texas Heart Institute/Baylor College of Medicine, Houston, TX, United States
| | - Leslee J Shaw
- Departments of Medicine (Cardiology) and Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | | | - Daniel S Berman
- Division of Cardiology, The Texas Heart Institute/Baylor College of Medicine, Houston, TX, United States
| | - Mario J Garcia
- Cardiology Division, Montefiore Medical Center, Montefiore Medical Center/Albert Einstein Colalege of Medicine, Cardiology Division. 111 E210th, Bronx, NY 10467, United States
| | - Damini Dey
- Department of Imaging, Cedars-Sinai Medical Center, Biomedical Imaging Research Institute, Los Angeles, CA, United States
| | - Leandro Slipczuk
- Cardiology Division, Montefiore Medical Center, Montefiore Medical Center/Albert Einstein Colalege of Medicine, Cardiology Division. 111 E210th, Bronx, NY 10467, United States
| |
Collapse
|
29
|
Brosolo G, Da Porto A, Marcante S, Picci A, Capilupi F, Capilupi P, Bulfone L, Vacca A, Bertin N, Vivarelli C, Comand J, Catena C, Sechi LA. Lipoprotein(a): Just an Innocent Bystander in Arterial Hypertension? Int J Mol Sci 2023; 24:13363. [PMID: 37686169 PMCID: PMC10487946 DOI: 10.3390/ijms241713363] [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: 08/07/2023] [Revised: 08/25/2023] [Accepted: 08/25/2023] [Indexed: 09/10/2023] Open
Abstract
Elevated plasma lipoprotein(a) [Lp(a)] is a relatively common and highly heritable trait conferring individuals time-dependent risk of developing atherosclerotic cardiovascular disease (CVD). Following its first description, Lp(a) triggered enormous scientific interest in the late 1980s, subsequently dampened in the mid-1990s by controversial findings of some prospective studies. It was only in the last decade that a large body of evidence has provided strong arguments for a causal and independent association between elevated Lp(a) levels and CVD, causing renewed interest in this lipoprotein as an emerging risk factor with a likely contribution to cardiovascular residual risk. Accordingly, the 2022 consensus statement of the European Atherosclerosis Society has suggested inclusion of Lp(a) measurement in global risk estimation. The development of highly effective Lp(a)-lowering drugs (e.g., antisense oligonucleotides and small interfering RNA, both blocking LPA gene expression) which are still under assessment in phase 3 trials, will provide a unique opportunity to reduce "residual cardiovascular risk" in high-risk populations, including patients with arterial hypertension. The current evidence in support of a specific role of Lp(a) in hypertension is somehow controversial and this narrative review aims to overview the general mechanisms relating Lp(a) to blood pressure regulation and hypertension-related cardiovascular and renal damage.
Collapse
Affiliation(s)
- Gabriele Brosolo
- Department of Medicine, University of Udine, 33100 Udine, Italy; (A.D.P.); (S.M.); (A.P.); (F.C.); (P.C.); (L.B.); (A.V.); (N.B.); (C.V.); (J.C.); (C.C.)
- European Hypertension Excellence Center, Clinica Medica, University of Udine, 33100 Udine, Italy
| | - Andrea Da Porto
- Department of Medicine, University of Udine, 33100 Udine, Italy; (A.D.P.); (S.M.); (A.P.); (F.C.); (P.C.); (L.B.); (A.V.); (N.B.); (C.V.); (J.C.); (C.C.)
- Diabetes and Metabolism Unit, Clinica Medica, University of Udine, 33100 Udine, Italy
| | - Stefano Marcante
- Department of Medicine, University of Udine, 33100 Udine, Italy; (A.D.P.); (S.M.); (A.P.); (F.C.); (P.C.); (L.B.); (A.V.); (N.B.); (C.V.); (J.C.); (C.C.)
- European Hypertension Excellence Center, Clinica Medica, University of Udine, 33100 Udine, Italy
| | - Alessandro Picci
- Department of Medicine, University of Udine, 33100 Udine, Italy; (A.D.P.); (S.M.); (A.P.); (F.C.); (P.C.); (L.B.); (A.V.); (N.B.); (C.V.); (J.C.); (C.C.)
- European Hypertension Excellence Center, Clinica Medica, University of Udine, 33100 Udine, Italy
| | - Filippo Capilupi
- Department of Medicine, University of Udine, 33100 Udine, Italy; (A.D.P.); (S.M.); (A.P.); (F.C.); (P.C.); (L.B.); (A.V.); (N.B.); (C.V.); (J.C.); (C.C.)
- European Hypertension Excellence Center, Clinica Medica, University of Udine, 33100 Udine, Italy
| | - Patrizio Capilupi
- Department of Medicine, University of Udine, 33100 Udine, Italy; (A.D.P.); (S.M.); (A.P.); (F.C.); (P.C.); (L.B.); (A.V.); (N.B.); (C.V.); (J.C.); (C.C.)
- European Hypertension Excellence Center, Clinica Medica, University of Udine, 33100 Udine, Italy
| | - Luca Bulfone
- Department of Medicine, University of Udine, 33100 Udine, Italy; (A.D.P.); (S.M.); (A.P.); (F.C.); (P.C.); (L.B.); (A.V.); (N.B.); (C.V.); (J.C.); (C.C.)
- European Hypertension Excellence Center, Clinica Medica, University of Udine, 33100 Udine, Italy
| | - Antonio Vacca
- Department of Medicine, University of Udine, 33100 Udine, Italy; (A.D.P.); (S.M.); (A.P.); (F.C.); (P.C.); (L.B.); (A.V.); (N.B.); (C.V.); (J.C.); (C.C.)
- European Hypertension Excellence Center, Clinica Medica, University of Udine, 33100 Udine, Italy
| | - Nicole Bertin
- Department of Medicine, University of Udine, 33100 Udine, Italy; (A.D.P.); (S.M.); (A.P.); (F.C.); (P.C.); (L.B.); (A.V.); (N.B.); (C.V.); (J.C.); (C.C.)
- Thrombosis and Hemostasis Unit, Clinica Medica, University of Udine, 33100 Udine, Italy
| | - Cinzia Vivarelli
- Department of Medicine, University of Udine, 33100 Udine, Italy; (A.D.P.); (S.M.); (A.P.); (F.C.); (P.C.); (L.B.); (A.V.); (N.B.); (C.V.); (J.C.); (C.C.)
| | - Jacopo Comand
- Department of Medicine, University of Udine, 33100 Udine, Italy; (A.D.P.); (S.M.); (A.P.); (F.C.); (P.C.); (L.B.); (A.V.); (N.B.); (C.V.); (J.C.); (C.C.)
- European Hypertension Excellence Center, Clinica Medica, University of Udine, 33100 Udine, Italy
| | - Cristiana Catena
- Department of Medicine, University of Udine, 33100 Udine, Italy; (A.D.P.); (S.M.); (A.P.); (F.C.); (P.C.); (L.B.); (A.V.); (N.B.); (C.V.); (J.C.); (C.C.)
- European Hypertension Excellence Center, Clinica Medica, University of Udine, 33100 Udine, Italy
| | - Leonardo A. Sechi
- Department of Medicine, University of Udine, 33100 Udine, Italy; (A.D.P.); (S.M.); (A.P.); (F.C.); (P.C.); (L.B.); (A.V.); (N.B.); (C.V.); (J.C.); (C.C.)
- European Hypertension Excellence Center, Clinica Medica, University of Udine, 33100 Udine, Italy
- Diabetes and Metabolism Unit, Clinica Medica, University of Udine, 33100 Udine, Italy
- Thrombosis and Hemostasis Unit, Clinica Medica, University of Udine, 33100 Udine, Italy
| |
Collapse
|
30
|
Koschinsky ML, Stroes ESG, Kronenberg F. Daring to dream: Targeting lipoprotein(a) as a causal and risk-enhancing factor. Pharmacol Res 2023; 194:106843. [PMID: 37406784 DOI: 10.1016/j.phrs.2023.106843] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 06/15/2023] [Accepted: 06/27/2023] [Indexed: 07/07/2023]
Abstract
Lipoprotein(a) [Lp(a)], a distinct lipoprotein class, has become a major focus for cardiovascular research. This review is written in light of the recent guideline and consensus statements on Lp(a) and focuses on 1) the causal association between Lp(a) and cardiovascular outcomes, 2) the potential mechanisms by which elevated Lp(a) contributes to cardiovascular diseases, 3) the metabolic insights on the production and clearance of Lp(a) and 4) the current and future therapeutic approaches to lower Lp(a) concentrations. The concentrations of Lp(a) are under strict genetic control. There exists a continuous relationship between the Lp(a) concentrations and risk for various endpoints of atherosclerotic cardiovascular disease (ASCVD). One in five people in the Caucasian population is considered to have increased Lp(a) concentrations; the prevalence of elevated Lp(a) is even higher in black populations. This makes Lp(a) a cardiovascular risk factor of major public health relevance. Besides the association between Lp(a) and myocardial infarction, the relationship with aortic valve stenosis has become a major focus of research during the last decade. Genetic studies provided strong support for a causal association between Lp(a) and cardiovascular outcomes: carriers of genetic variants associated with lifelong increased Lp(a) concentration are significantly more frequent in patients with ASCVD. This has triggered the development of drugs that can specifically lower Lp(a) concentrations: mRNA-targeting therapies such as anti-sense oligonucleotide (ASO) therapies and short interfering RNA (siRNA) therapies have opened new avenues to lower Lp(a) concentrations more than 95%. Ongoing Phase II and III clinical trials of these compounds are discussed in this review.
Collapse
Affiliation(s)
- Marlys L Koschinsky
- Robarts Research Institute, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada; Department of Physiology & Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Erik S G Stroes
- Department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Florian Kronenberg
- Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
31
|
Genovesi S, Giussani M, Lieti G, Orlando A, Patti I, Parati G. Evidence and Uncertainties on Lipoprotein(a) as a Marker of Cardiovascular Health Risk in Children and Adolescents. Biomedicines 2023; 11:1661. [PMID: 37371756 PMCID: PMC10295837 DOI: 10.3390/biomedicines11061661] [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: 05/10/2023] [Revised: 06/05/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
Lipoprotein(a) (Lp(a)) is made up of apoprotein(a) (apo(a)) and an LDL-like particle. The LPA gene encodes apo(a) and thus determines the characteristics and amount of apo(a) and Lp(a). The proportion of Lp(a) in each individual is genetically determined and is only minimally modifiable by the environment or diet. Lp(a) has important pro-atherosclerotic and pro-inflammatory effects. It has been hypothesized that Lp(a) also has pro-coagulant and antifibrinolytic actions. For these reasons, high Lp(a) values are an important independent risk factor for cardiovascular disease and calcific aortic valve stenosis. Numerous studies have been performed in adults about the pathophysiology and epidemiology of Lp(a) and research is under way for the development of drugs capable of reducing Lp(a) plasma values. Much less information is available regarding Lp(a) in children and adolescents. The present article reviews the evidence on this topic. The review addresses the issues of Lp(a) changes during growth, the correlation between Lp(a) values in children and those in their parents, and between Lp(a) levels in children, and the presence of cardiovascular disease in the family. Gaining information on these points is particularly important for deciding whether Lp(a) assay may be useful for defining the cardiovascular risk in children, in order to plan a prevention program early.
Collapse
Affiliation(s)
- Simonetta Genovesi
- School of Medicine and Surgery, Milano-Bicocca University, 20126 Milan, Italy; (G.L.); (I.P.); (G.P.)
- Istituto Auxologico Italiano, Istituto Ricovero Cura Carattere Scientifico (IRCCS), 20135 Milan, Italy; (M.G.); (A.O.)
| | - Marco Giussani
- Istituto Auxologico Italiano, Istituto Ricovero Cura Carattere Scientifico (IRCCS), 20135 Milan, Italy; (M.G.); (A.O.)
| | - Giulia Lieti
- School of Medicine and Surgery, Milano-Bicocca University, 20126 Milan, Italy; (G.L.); (I.P.); (G.P.)
| | - Antonina Orlando
- Istituto Auxologico Italiano, Istituto Ricovero Cura Carattere Scientifico (IRCCS), 20135 Milan, Italy; (M.G.); (A.O.)
| | - Ilenia Patti
- School of Medicine and Surgery, Milano-Bicocca University, 20126 Milan, Italy; (G.L.); (I.P.); (G.P.)
| | - Gianfranco Parati
- School of Medicine and Surgery, Milano-Bicocca University, 20126 Milan, Italy; (G.L.); (I.P.); (G.P.)
- Istituto Auxologico Italiano, Istituto Ricovero Cura Carattere Scientifico (IRCCS), 20135 Milan, Italy; (M.G.); (A.O.)
| |
Collapse
|
32
|
Lorey MB, Youssef A, Äikäs L, Borrelli M, Hermansson M, Assini JM, Kemppainen A, Ruhanen H, Ruuth M, Matikainen S, Kovanen PT, Käkelä R, Boffa MB, Koschinsky ML, Öörni K. Lipoprotein(a) induces caspase-1 activation and IL-1 signaling in human macrophages. Front Cardiovasc Med 2023; 10:1130162. [PMID: 37293282 PMCID: PMC10244518 DOI: 10.3389/fcvm.2023.1130162] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 05/02/2023] [Indexed: 06/10/2023] Open
Abstract
Introduction Lipoprotein(a) (Lp(a)) is an LDL-like particle with an additional apolipoprotein (apo)(a) covalently attached. Elevated levels of circulating Lp(a) are a risk factor for atherosclerosis. A proinflammatory role for Lp(a) has been proposed, but its molecular details are incompletely defined. Methods and results To explore the effect of Lp(a) on human macrophages we performed RNA sequencing on THP-1 macrophages treated with Lp(a) or recombinant apo(a), which showed that especially Lp(a) induces potent inflammatory responses. Thus, we stimulated THP-1 macrophages with serum containing various Lp(a) levels to investigate their correlations with cytokines highlighted by the RNAseq, showing significant correlations with caspase-1 activity and secretion of IL-1β and IL-18. We further isolated both Lp(a) and LDL particles from three donors and then compared their atheroinflammatory potentials together with recombinant apo(a) in primary and THP-1 derived macrophages. Compared with LDL, Lp(a) induced a robust and dose-dependent caspase-1 activation and release of IL-1β and IL-18 in both macrophage types. Recombinant apo(a) strongly induced caspase-1 activation and IL-1β release in THP-1 macrophages but yielded weak responses in primary macrophages. Structural analysis of these particles revealed that the Lp(a) proteome was enriched in proteins associated with complement activation and coagulation, and its lipidome was relatively deficient in polyunsaturated fatty acids and had a high n-6/n-3 ratio promoting inflammation. Discussion Our data show that Lp(a) particles induce the expression of inflammatory genes, and Lp(a) and to a lesser extent apo(a) induce caspase-1 activation and IL-1 signaling. Major differences in the molecular profiles between Lp(a) and LDL contribute to Lp(a) being more atheroinflammatory.
Collapse
Affiliation(s)
- Martina B. Lorey
- Atherosclerosis Research Laboratory, Wihuri Research Institute, Helsinki, Finland
- Molecular and Integrative Biosciences, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Amer Youssef
- Robarts Research Institute, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada
| | - Lauri Äikäs
- Atherosclerosis Research Laboratory, Wihuri Research Institute, Helsinki, Finland
| | - Matthew Borrelli
- Department of Physiology & Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada
| | - Martin Hermansson
- Atherosclerosis Research Laboratory, Wihuri Research Institute, Helsinki, Finland
| | - Julia M. Assini
- Robarts Research Institute, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada
- Department of Biochemistry, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada
| | - Aapeli Kemppainen
- Atherosclerosis Research Laboratory, Wihuri Research Institute, Helsinki, Finland
| | - Hanna Ruhanen
- Molecular and Integrative Biosciences, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
- Helsinki University Lipidomics Unit (HiLIPID), Helsinki Institute of Life Science (HiLIFE) and Biocenter Finland, Helsinki, Finland
| | - Maija Ruuth
- Atherosclerosis Research Laboratory, Wihuri Research Institute, Helsinki, Finland
| | - Sampsa Matikainen
- Helsinki Rheumatic Disease and Inflammation Research Group, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Petri T. Kovanen
- Atherosclerosis Research Laboratory, Wihuri Research Institute, Helsinki, Finland
| | - Reijo Käkelä
- Molecular and Integrative Biosciences, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
- Helsinki University Lipidomics Unit (HiLIPID), Helsinki Institute of Life Science (HiLIFE) and Biocenter Finland, Helsinki, Finland
| | - Michael B. Boffa
- Robarts Research Institute, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada
- Department of Biochemistry, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada
| | - Marlys L. Koschinsky
- Robarts Research Institute, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada
- Department of Physiology & Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada
| | - Katariina Öörni
- Atherosclerosis Research Laboratory, Wihuri Research Institute, Helsinki, Finland
- Molecular and Integrative Biosciences, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| |
Collapse
|
33
|
Gilliland TC, Liu Y, Mohebi R, Miksenas H, Haidermota S, Wong M, Hu X, Cristino JR, Browne A, Plutzky J, Tsimikas S, Januzzi JL, Natarajan P. Lipoprotein(a), Oxidized Phospholipids, and Coronary Artery Disease Severity and Outcomes. J Am Coll Cardiol 2023; 81:1780-1792. [PMID: 37137588 PMCID: PMC10824318 DOI: 10.1016/j.jacc.2023.02.050] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 02/08/2023] [Accepted: 02/15/2023] [Indexed: 05/05/2023]
Abstract
BACKGROUND Lipoprotein(a) (Lp[a]) and oxidized phospholipids (OxPLs) are each independent risk factors for atherosclerotic cardiovascular disease. The extent to which Lp(a) and OxPLs predict coronary artery disease (CAD) severity and outcomes in a contemporary, statin-treated cohort is not well established. OBJECTIVES This study sought to evaluate the relationships between Lp(a) particle concentration and OxPLs associated with apolipoprotein B (OxPL-apoB) or apolipoprotein(a) (OxPL-apo[a]) with angiographic CAD and cardiovascular outcomes. METHODS Among 1,098 participants referred for coronary angiography in the CASABLANCA (Catheter Sampled Blood Archive in Cardiovascular Diseases) study, Lp(a), OxPL-apoB, and OxPL-apo(a) were measured. Logistic regression estimated the risk of multivessel coronary stenoses by Lp(a)-related biomarker level. Cox proportional hazards regression estimated the risk of major adverse cardiovascular events (MACEs) (coronary revascularization, nonfatal myocardial infarction, nonfatal stroke, and cardiovascular death) in follow-up. RESULTS Median Lp(a) was 26.45 nmol/L (IQR: 11.39-89.49 nmol/L). Lp(a), OxPL-apoB, and OxPL-apo(a) were highly correlated (Spearman R ≥0.91 for all pairwise combinations). Lp(a) and OxPL-apoB were associated with multivessel CAD. Odds of multivessel CAD per doubling of Lp(a), OxPL-apoB, and OxPL-apo(a) were 1.10 (95% CI: 1.03-1.18; P = 0.006), 1.18 (95% CI: 1.03-1.34; P = 0.01), and 1.07 (95% CI: 0.99-1.16; P = 0.07), respectively. All biomarkers were associated with cardiovascular events. HRs for MACE per doubling of Lp(a), OxPL-apoB, and OxPL-apo(a) were 1.08 (95% CI: 1.03-1.14; P = 0.001), 1.15 (95% CI: 1.05-1.26; P = 0.004), and 1.07 (95% CI: 1.01-1.14; P = 0.02), respectively. CONCLUSIONS In patients undergoing coronary angiography, Lp(a) and OxPL-apoB are associated with multivessel CAD. Lp(a), OxPL-apoB, and OxPL-apo(a) are associated with incident cardiovascular events. (Catheter Sampled Blood Archive in Cardiovascular Diseases [CASABLANCA]; NCT00842868).
Collapse
Affiliation(s)
- Thomas C Gilliland
- Division of Cardiology, Massachusetts General Hospital, Boston, Massachusetts, USA; Program in Medical and Population Genetics and the Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA; Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Yuxi Liu
- Division of Cardiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Reza Mohebi
- Division of Cardiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Hannah Miksenas
- Division of Cardiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Sara Haidermota
- Division of Cardiology, Massachusetts General Hospital, Boston, Massachusetts, USA; Program in Medical and Population Genetics and the Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
| | - Megan Wong
- Division of Cardiology, Massachusetts General Hospital, Boston, Massachusetts, USA; Program in Medical and Population Genetics and the Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
| | - Xingdi Hu
- Novartis Pharmaceuticals Corporation, East Hanover, New Jersey, USA
| | | | - Auris Browne
- Novartis Pharmaceuticals Corporation, East Hanover, New Jersey, USA
| | - Jorge Plutzky
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA; Division of Cardiology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Sotirios Tsimikas
- Sulpizio Cardiovascular Center, University of California San Diego, La Jolla, California, USA
| | - James L Januzzi
- Division of Cardiology, Massachusetts General Hospital, Boston, Massachusetts, USA; Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA; Baim Institute for Clinical Research, Boston, Massachusetts, USA
| | - Pradeep Natarajan
- Division of Cardiology, Massachusetts General Hospital, Boston, Massachusetts, USA; Program in Medical and Population Genetics and the Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA; Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
34
|
Matveyenko A, Pavlyha M, Reyes-Soffer G. Supporting evidence for lipoprotein(a) measurements in clinical practice. Best Pract Res Clin Endocrinol Metab 2023; 37:101746. [PMID: 36828715 PMCID: PMC11014458 DOI: 10.1016/j.beem.2023.101746] [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: 02/13/2023]
Abstract
High levels of lipoprotein(a) [Lp(a)] are causal for development of atherosclerotic cardiovascular disease and highly regulated by genetics. Levels are higher in Blacks compared to Whites, and in women compared to men. Lp(a)'s main protein components are apolipoprotein (apo) (a) and apoB100, the latter being the main component of Low-Density Lipoprotein (LDL) particles. Studies have identified Lp(a) to be associated with inflammatory, coagulation and wound healing pathways. Lack of validated and accepted assays to measure Lp(a), risk cutoff values, guidelines for diagnosis, and targeted therapies have added challenges to the field. Scientific efforts are ongoing to address these, including studies evaluating the cardiovascular benefits of decreasing Lp(a) levels with targeted apo(a) lowering treatments. This review will provide a synopsis of evidence-based effects of high Lp(a) on disease presentation, highlight available guidelines and discuss promising therapies in development. We will conclude with current clinical information and future research needs in the field.
Collapse
Affiliation(s)
- Anastasiya Matveyenko
- Columbia University College of Physicians and Surgeons, Columbia University Irving Medical Center, 622 West 168th Street, P&S 10-501, New York, NY 10032, USA.
| | - Marianna Pavlyha
- Columbia University College of Physicians and Surgeons, Columbia University Irving Medical Center, 622 West 168th Street, P&S 10-501, New York, NY 10032, USA.
| | - Gissette Reyes-Soffer
- Columbia University College of Physicians and Surgeons, Columbia University Irving Medical Center, 622 West 168th Street, P&S 10-501, New York, NY 10032, USA.
| |
Collapse
|
35
|
Hussain Z, Iqbal J, Liu H, Zhou HD. Exploring the role of lipoprotein(a) in cardiovascular diseases and diabetes in Chinese population. Int J Biol Macromol 2023; 233:123586. [PMID: 36758756 DOI: 10.1016/j.ijbiomac.2023.123586] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/31/2023] [Accepted: 02/04/2023] [Indexed: 02/10/2023]
Abstract
A high level of lipoprotein (a) in the plasma has been associated with a variety of cardiovascular diseases and is considered to be an independent predictor of some other diseases. Based on recent studies, the concentration levels of Lp(a) in the Chinese population exhibit a distinctive variation from other populations. In the Chinese population, a high level of Lp(a) indicates a higher incidence of revascularization, platelet aggregation, and thrombogenicity following PCI. Increased risk of atherosclerotic cardiovascular disease (ASCVD) in Chinese population has been linked to higher levels of Lp(a), according to studies. More specifically, it has been found that in Chinese populations, higher levels of Lp(a) were linked to an increased risk of coronary heart disease, severe aortic valve stenosis, deep vein thrombosis in patients with spinal cord injuries, central vein thrombosis in patients receiving hemodialysis, and stroke. Furthermore, new and consistent data retrieved from several clinical trials also suggest that Lp (a) might also play an essential role in some other conditions, including metabolic syndrome, type 2 diabetes and cancers. This review explores the clinical and epidemiological relationships among Lp(a), cardiovascular diseases and diabetes in the Chinese population as well as potential Lp(a) underlying mechanisms in these diseases. However, further research is needed to better understand the role of Lp(a) in cardiovascular diseases and especially diabetes in the Chinese population.
Collapse
Affiliation(s)
- Zubair Hussain
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, 450000, Zhengzhou, China; Department of Pathophysiology, Academy of Medical Science, College of Medicine, Zhengzhou University, 450000, Zhengzhou, China; China-US (Henan) Hormel Cancer Institute, 450000, Zhengzhou, China
| | - Junaid Iqbal
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Hongcai Liu
- Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China; Suzhou Institute of Systems Medicine, Suzhou 215123, China
| | - Hou-De Zhou
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China.
| |
Collapse
|
36
|
Small AM, Peloso G, Linefsky J, Aragam J, Galloway A, Tanukonda V, Wang LC, Yu Z, Selvaraj MS, Farber-Eger EH, Baker MT, Setia-Verma S, Lee SSK, Preuss M, Ritchie M, Damrauer SM, Rader DJ, Wells QS, Loos RJF, Lubitz S, Thanassoulis G, Cho K, Wilson PWF, Natarajan P, O’Donnell CJ. Multiancestry Genome-Wide Association Study of Aortic Stenosis Identifies Multiple Novel Loci in the Million Veteran Program. Circulation 2023; 147:942-955. [PMID: 36802703 PMCID: PMC10806851 DOI: 10.1161/circulationaha.122.061451] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 12/15/2022] [Indexed: 02/22/2023]
Abstract
BACKGROUND Calcific aortic stenosis (CAS) is the most common valvular heart disease in older adults and has no effective preventive therapies. Genome-wide association studies (GWAS) can identify genes influencing disease and may help prioritize therapeutic targets for CAS. METHODS We performed a GWAS and gene association study of 14 451 patients with CAS and 398 544 controls in the Million Veteran Program. Replication was performed in the Million Veteran Program, Penn Medicine Biobank, Mass General Brigham Biobank, BioVU, and BioMe, totaling 12 889 cases and 348 094 controls. Causal genes were prioritized from genome-wide significant variants using polygenic priority score gene localization, expression quantitative trait locus colocalization, and nearest gene methods. CAS genetic architecture was compared with that of atherosclerotic cardiovascular disease. Causal inference for cardiometabolic biomarkers in CAS was performed using Mendelian randomization and genome-wide significant loci were characterized further through phenome-wide association study. RESULTS We identified 23 genome-wide significant lead variants in our GWAS representing 17 unique genomic regions. Of the 23 lead variants, 14 were significant in replication, representing 11 unique genomic regions. Five replicated genomic regions were previously known risk loci for CAS (PALMD, TEX41, IL6, LPA, FADS) and 6 were novel (CEP85L, FTO, SLMAP, CELSR2, MECOM, CDAN1). Two novel lead variants were associated in non-White individuals (P<0.05): rs12740374 (CELSR2) in Black and Hispanic individuals and rs1522387 (SLMAP) in Black individuals. Of the 14 replicated lead variants, only 2 (rs10455872 [LPA], rs12740374 [CELSR2]) were also significant in atherosclerotic cardiovascular disease GWAS. In Mendelian randomization, lipoprotein(a) and low-density lipoprotein cholesterol were both associated with CAS, but the association between low-density lipoprotein cholesterol and CAS was attenuated when adjusting for lipoprotein(a). Phenome-wide association study highlighted varying degrees of pleiotropy, including between CAS and obesity at the FTO locus. However, the FTO locus remained associated with CAS after adjusting for body mass index and maintained a significant independent effect on CAS in mediation analysis. CONCLUSIONS We performed a multiancestry GWAS in CAS and identified 6 novel genomic regions in the disease. Secondary analyses highlighted the roles of lipid metabolism, inflammation, cellular senescence, and adiposity in the pathobiology of CAS and clarified the shared and differential genetic architectures of CAS with atherosclerotic cardiovascular diseases.
Collapse
Affiliation(s)
- Aeron M Small
- Department of Cardiology, Boston Veterans Affairs Healthcare System, West Roxbury, MA, USA
- Cardiovascular Medicine Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, MA, USA
| | - Gina Peloso
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), Veterans Affairs, Boston Healthcare System, Boston, Massachusetts
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, United States
| | - Jason Linefsky
- Atlanta Veterans Affairs Medical Center, Decatur, Georgia
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Jayashri Aragam
- Department of Cardiology, Boston Veterans Affairs Healthcare System, West Roxbury, MA, USA
| | - Ashley Galloway
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), Veterans Affairs, Boston Healthcare System, Boston, Massachusetts
| | | | - Lu-Chen Wang
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA, 02114
- Cardiovascular Disease Initiative, Broad Institute, Cambridge, MA, USA, 02142
| | - Zhi Yu
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA, 02114
- Cardiovascular Disease Initiative, Broad Institute, Cambridge, MA, USA, 02142
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Margaret Sunitha Selvaraj
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA, 02114
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Eric H Farber-Eger
- Vanderbilt Institute for Clinical and Translational Research, Vanderbilt University Medical Center, Nashville, TN, United States, 37232
| | - Michael T Baker
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Shefali Setia-Verma
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Simon SK Lee
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029
| | - Michael Preuss
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029
| | - Marylyn Ritchie
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Scott M Damrauer
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA, 19104
- Corporal Michael Crescenz VA Medical Center, Philadelphia, PA, USA, 19104
| | - Daniel J Rader
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Quinn S Wells
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN 37232
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Ruth J F Loos
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA, 10029
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark
| | - Steven Lubitz
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA, 02114
| | - George Thanassoulis
- Department of Medicine, Division of Experimental Medicine, McGill University Health Center, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada
| | - Kelly Cho
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), Veterans Affairs, Boston Healthcare System, Boston, Massachusetts
| | - Peter WF Wilson
- Atlanta Veterans Affairs Medical Center, Decatur, Georgia
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | | | - Pradeep Natarajan
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA, 02114
- Cardiovascular Disease Initiative, Broad Institute, Cambridge, MA, USA, 02142
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston
- Center for Genomic Medicine, Department of Medicine, Massachusetts General Hospital, Boston
| | - Christopher J O’Donnell
- Department of Cardiology, Boston Veterans Affairs Healthcare System, West Roxbury, MA, USA
- Cardiovascular Medicine Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, MA, USA
| |
Collapse
|
37
|
Itabe H, Obama T. The Oxidized Lipoproteins In Vivo: Its Diversity and Behavior in the Human Circulation. Int J Mol Sci 2023; 24:ijms24065747. [PMID: 36982815 PMCID: PMC10053446 DOI: 10.3390/ijms24065747] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 03/07/2023] [Accepted: 03/15/2023] [Indexed: 03/19/2023] Open
Abstract
A high concentration of low-density lipoproteins (LDLs) in circulation has been well-known as a major risk factor for cardiovascular diseases. The presence of oxidized LDLs (oxLDLs) in atherosclerotic lesions and circulation was demonstrated using anti-oxLDL monoclonal antibodies. The so-called “oxLDL hypothesis”, as a mechanism for atherosclerosis development, has been attracting attention for decades. However, the oxLDL has been considered a hypothetical particle since the oxLDL present in vivo has not been fully characterized. Several chemically modified LDLs have been proposed to mimic oxLDLs. Some of the subfractions of LDL, especially Lp(a) and electronegative LDL, have been characterized as oxLDL candidates as oxidized phospholipids that stimulate vascular cells. Oxidized high-density lipoprotein (oxHDL) and oxLDL were discovered immunologically in vivo. Recently, an oxLDL-oxHDL complex was found in human plasma, suggesting the involvement of HDLs in the oxidative modification of lipoproteins in vivo. In this review, we summarize our understanding of oxidized lipoproteins and propose a novel standpoint to understand the oxidized lipoproteins present in vivo.
Collapse
|
38
|
Li J. Lipoprotein(a) and Atherosclerotic Cardiovascular Diseases: Evidence from Chinese Population. CARDIOLOGY DISCOVERY 2023; 3:40-47. [DOI: 10.1097/cd9.0000000000000059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Cardiovascular disease (CVD) is the leading cause of mortality worldwide. Multiple factors are involved in CVD, and emerging data indicate that lipoprotein(a) (Lp(a)) may be associated with atherosclerotic cardiovascular disease (ASCVD) independent of other traditional risk factors. Lp(a) has been identified as a novel therapeutic target. Previous studies on the influence of Lp(a) in CVD have mainly used in western populations. In this review, the association of plasma Lp(a) concentration with ASCVD was summarized, with regards to epidemiological, population-based observational, and pathological studies in Chinese populations. Lp(a) mutations and copy number variations in Chinese populations are also explored. Finally, the impact of plasma Lp(a) levels on patients with type 2 diabetes mellitus, cancer, and familial hypercholesterolemia are discussed.
Collapse
Affiliation(s)
- Jianjun Li
- Cardiometabolic Center, Fuwai Hospital, Chinese Academy of Medical Sciences, Beijing 100037, China
| |
Collapse
|
39
|
Girard A, Gaillard E, Puri R, Capoulade R, Chan KL, Paulin A, Manikpurage HD, Dumesnil J, Tam JW, Teo KK, Couture C, Wareham NJ, Clavel MA, Stroes ESG, Mathieu P, Thériault S, Tsimikas S, Pibarot P, Boekholdt SM, Arsenault BJ. Impact of C-reactive protein levels on lipoprotein(a)-associated aortic stenosis incidence and progression. EUROPEAN HEART JOURNAL OPEN 2023; 3:oead032. [PMID: 37077580 PMCID: PMC10108885 DOI: 10.1093/ehjopen/oead032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 02/14/2023] [Accepted: 03/22/2023] [Indexed: 04/01/2023]
Abstract
Aims Elevated lipoprotein(a) [Lp(a)] levels are associated with the risk of coronary artery disease (CAD) and calcific aortic valve stenosis (CAVS). Observational studies revealed that Lp(a) and C-reactive protein (CRP) levels, a biomarker of systemic inflammation, may jointly predict CAD risk. Whether Lp(a) and CRP levels also jointly predict CAVS incidence and progression is unknown. Methods and results We investigated the association of Lp(a) with CAVS according to CRP levels in the European Prospective Investigation into Cancer and Nutrition (EPIC)-Norfolk study (n = 18 226, 406 incident cases) and the UK Biobank (n = 438 260, 4582 incident cases), as well as in the ASTRONOMER study (n = 220), which assessed the haemodynamic progression rate of pre-existing mild-to-moderate aortic stenosis. In EPIC-Norfolk, in comparison to individuals with low Lp(a) levels (<50 mg/dL) and low CRP levels (<2.0 mg/L), those with elevated Lp(a) (>50 mg/dL) and low CRP levels (<2.0 mg/L) and those with elevated Lp(a) (>50 mg/dL) and elevated CRP levels (>2.0 mg/L) had a higher CAVS risk [hazard ratio (HR) = 1.86 (95% confidence intervals, 1.30-2.67) and 2.08 (1.44-2.99), respectively]. A comparable predictive value of Lp(a) in patients with vs. without elevated CRP levels was also noted in the UK Biobank. In ASTRONOMER, CAVS progression was comparable in patients with elevated Lp(a) levels with or without elevated CRP levels. Conclusion Lp(a) predicts the incidence and possibly progression of CAVS regardless of plasma CRP levels. Lowering Lp(a) levels may warrant further investigation in the prevention and treatment of CAVS, regardless of systemic inflammation.
Collapse
Affiliation(s)
- Arnaud Girard
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Québec, QC, G1V 4G5, Canada
| | - Emilie Gaillard
- Department of Cardiology, Amsterdam UMC, University of Amsterdam, Amsterdam, 1105 AZ, The Netherlands
| | - Rishi Puri
- Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Romain Capoulade
- Nantes Université, CNRS, INSERM, l’institut du thorax, F-44000 Nantes, 44007, France
| | - Kwan L Chan
- Department of Medicine, University of Ottawa Heart Institute, Ottawa, ON, K1Y 4W7, Canada
| | - Audrey Paulin
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Québec, QC, G1V 4G5, Canada
| | - Hasanga D Manikpurage
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Québec, QC, G1V 4G5, Canada
| | - Jean Dumesnil
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Québec, QC, G1V 4G5, Canada
| | - James W Tam
- Department of Medicine, St. Boniface General Hospital, Winnipeg, MB, R2H 2A6, Canada
| | - Koon K Teo
- Department of Medicine (Cardiology), McMaster University, Hamilton, ON, L8S 4L8, Canada
| | - Christian Couture
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Québec, QC, G1V 4G5, Canada
| | - Nicholas J Wareham
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB2 1TN, UK
| | - Marie-Annick Clavel
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Québec, QC, G1V 4G5, Canada
- Department of Medicine, Faculty of Medicine, Université Laval, Québec, QC, G1V 0A6, Canada
| | - Erik S G Stroes
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, 1105 AZ, The Netherlands
| | - Patrick Mathieu
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Québec, QC, G1V 4G5, Canada
- Department of Surgery, Faculty of Medicine, Université Laval, Québec, QC, G1V 0A6, Canada
| | - Sébastien Thériault
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Québec, QC, G1V 4G5, Canada
- Department of Molecular Biology, Medical Biochemistry and Pathology, Faculty of Medicine, Université Laval, Québec, QC, G1V 0A6, Canada
| | - Sotirios Tsimikas
- Division of Cardiovascular Diseases, Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Philippe Pibarot
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Québec, QC, G1V 4G5, Canada
- Department of Medicine, Faculty of Medicine, Université Laval, Québec, QC, G1V 0A6, Canada
| | - S Matthijs Boekholdt
- Department of Cardiology, Amsterdam UMC, University of Amsterdam, Amsterdam, 1105 AZ, The Netherlands
| | - Benoit J Arsenault
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Québec, QC, G1V 4G5, Canada
- Department of Medicine, Faculty of Medicine, Université Laval, Québec, QC, G1V 0A6, Canada
| |
Collapse
|
40
|
Lipoprotein(a) in Atherosclerotic Diseases: From Pathophysiology to Diagnosis and Treatment. Molecules 2023; 28:molecules28030969. [PMID: 36770634 PMCID: PMC9918959 DOI: 10.3390/molecules28030969] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 01/12/2023] [Accepted: 01/17/2023] [Indexed: 01/21/2023] Open
Abstract
Lipoprotein(a) (Lp(a)) is a low-density lipoprotein (LDL) cholesterol-like particle bound to apolipoprotein(a). Increased Lp(a) levels are an independent, heritable causal risk factor for atherosclerotic cardiovascular disease (ASCVD) as they are largely determined by variations in the Lp(a) gene (LPA) locus encoding apo(a). Lp(a) is the preferential lipoprotein carrier for oxidized phospholipids (OxPL), and its role adversely affects vascular inflammation, atherosclerotic lesions, endothelial function and thrombogenicity, which pathophysiologically leads to cardiovascular (CV) events. Despite this crucial role of Lp(a), its measurement lacks a globally unified method, and, between different laboratories, results need standardization. Standard antilipidemic therapies, such as statins, fibrates and ezetimibe, have a mediocre effect on Lp(a) levels, although it is not yet clear whether such treatments can affect CV events and prognosis. This narrative review aims to summarize knowledge regarding the mechanisms mediating the effect of Lp(a) on inflammation, atherosclerosis and thrombosis and discuss current diagnostic and therapeutic potentials.
Collapse
|
41
|
Chen Z, Lehertshuber C, Schunkert H. Genome Editing in Dyslipidemia and Atherosclerosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1396:139-156. [DOI: 10.1007/978-981-19-5642-3_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
|
42
|
Simantiris S, Antonopoulos AS, Papastamos C, Benetos G, Koumallos N, Tsioufis K, Tousoulis D. Lipoprotein(a) and inflammation- pathophysiological links and clinical implications for cardiovascular disease. J Clin Lipidol 2023; 17:55-63. [PMID: 36333256 DOI: 10.1016/j.jacl.2022.10.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 10/11/2022] [Accepted: 10/12/2022] [Indexed: 11/05/2022]
Abstract
The role of lipoprotein(a) (Lp[a]) as a significant and possibly causal cardiovascular disease (CVD) risk factor has been well established. Many studies, mostly experimental, have supported inflammation as a mediator of Lp(a)-induced increase in CVD risk. Lp(a), mainly through oxidized phospholipids bound to its apolipoprotein(a) part, leads to monocyte activation and endothelial dysfunction. The relationship between Lp(a) and inflammation is bidirectional as Lp(a) levels, besides being associated with inflammatory properties, are regulated by inflammatory stimuli or anti-inflammatory treatment. Reduction of Lp(a) concentration, especially by potent siRNA agents, contributes to partial reversion of the Lp(a) related inflammatory profile. This review aims to present the current pathophysiological and clinical evidence of the relationship between Lp(a) and inflammation.
Collapse
Affiliation(s)
- Spyridon Simantiris
- 1st Cardiology Department, Hippokration Hospital, National and Kapodistrian University of Athens, Vas. Sofias Avenue 114, Athens 11527, Greece (Drs Simantiris, Antonopoulos, Papastamos, Benetos, Tsioufis, and Tousoulis)
| | - Alexios S Antonopoulos
- 1st Cardiology Department, Hippokration Hospital, National and Kapodistrian University of Athens, Vas. Sofias Avenue 114, Athens 11527, Greece (Drs Simantiris, Antonopoulos, Papastamos, Benetos, Tsioufis, and Tousoulis)
| | - Charalampos Papastamos
- 1st Cardiology Department, Hippokration Hospital, National and Kapodistrian University of Athens, Vas. Sofias Avenue 114, Athens 11527, Greece (Drs Simantiris, Antonopoulos, Papastamos, Benetos, Tsioufis, and Tousoulis)
| | - Georgios Benetos
- 1st Cardiology Department, Hippokration Hospital, National and Kapodistrian University of Athens, Vas. Sofias Avenue 114, Athens 11527, Greece (Drs Simantiris, Antonopoulos, Papastamos, Benetos, Tsioufis, and Tousoulis)
| | - Nikolaos Koumallos
- Department of Cardiothoracic Surgery, Hippokration Hospital, Athens, Greece (Dr Koumallos)
| | - Konstantinos Tsioufis
- 1st Cardiology Department, Hippokration Hospital, National and Kapodistrian University of Athens, Vas. Sofias Avenue 114, Athens 11527, Greece (Drs Simantiris, Antonopoulos, Papastamos, Benetos, Tsioufis, and Tousoulis)
| | - Dimitris Tousoulis
- 1st Cardiology Department, Hippokration Hospital, National and Kapodistrian University of Athens, Vas. Sofias Avenue 114, Athens 11527, Greece (Drs Simantiris, Antonopoulos, Papastamos, Benetos, Tsioufis, and Tousoulis).
| |
Collapse
|
43
|
Maloberti A, Fabbri S, Colombo V, Gualini E, Monticelli M, Daus F, Busti A, Galasso M, De Censi L, Algeri M, Merlini PA, Giannattasio C. Lipoprotein(a): Cardiovascular Disease, Aortic Stenosis and New Therapeutic Option. Int J Mol Sci 2022; 24:ijms24010170. [PMID: 36613613 PMCID: PMC9820656 DOI: 10.3390/ijms24010170] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/23/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
Atherosclerosis is a chronic and progressive inflammatory process beginning early in life with late clinical manifestation. This slow pathological trend underlines the importance to early identify high-risk patients and to treat intensively risk factors to prevent the onset and/or the progression of atherosclerotic lesions. In addition to the common Cardiovascular (CV) risk factors, new markers able to increase the risk of CV disease have been identified. Among them, high levels of Lipoprotein(a)-Lp(a)-lead to very high risk of future CV diseases; this relationship has been well demonstrated in epidemiological, mendelian randomization and genome-wide association studies as well as in meta-analyses. Recently, new aspects have been identified, such as its association with aortic stenosis. Although till recent years it has been considered an unmodifiable risk factor, specific drugs have been developed with a strong efficacy in reducing the circulating levels of Lp(a) and their capacity to reduce subsequent CV events is under testing in ongoing trials. In this paper we will review all these aspects: from the synthesis, clearance and measurement of Lp(a), through the findings that examine its association with CV diseases and aortic stenosis to the new therapeutic options that will be available in the next years.
Collapse
Affiliation(s)
- Alessandro Maloberti
- Cardiology 4, Cardio Center A. De Gasperis, ASST GOM Niguarda, 20162 Milan, Italy
- School of Medicine and Surgery, Milano-Bicocca University, 20126 Milan, Italy
- Correspondence: ; Tel.: +39-02-644-478-55; Fax: +39-02-644-425-66
| | - Saverio Fabbri
- School of Medicine and Surgery, Milano-Bicocca University, 20126 Milan, Italy
| | - Valentina Colombo
- School of Medicine and Surgery, Milano-Bicocca University, 20126 Milan, Italy
| | - Elena Gualini
- School of Medicine and Surgery, Milano-Bicocca University, 20126 Milan, Italy
| | | | - Francesca Daus
- School of Medicine and Surgery, Milano-Bicocca University, 20126 Milan, Italy
| | - Andrea Busti
- School of Medicine and Surgery, Milano-Bicocca University, 20126 Milan, Italy
| | - Michele Galasso
- School of Medicine and Surgery, Milano-Bicocca University, 20126 Milan, Italy
| | - Lorenzo De Censi
- School of Medicine and Surgery, Milano-Bicocca University, 20126 Milan, Italy
| | - Michela Algeri
- Cardiology 4, Cardio Center A. De Gasperis, ASST GOM Niguarda, 20162 Milan, Italy
| | | | - Cristina Giannattasio
- Cardiology 4, Cardio Center A. De Gasperis, ASST GOM Niguarda, 20162 Milan, Italy
- School of Medicine and Surgery, Milano-Bicocca University, 20126 Milan, Italy
| |
Collapse
|
44
|
Nestel P, Loh WJ, Ward NC, Watts GF. New Horizons: Revival of Lipoprotein (a) as a Risk Factor for Cardiovascular Disease. J Clin Endocrinol Metab 2022; 107:e4281-e4294. [PMID: 36108076 DOI: 10.1210/clinem/dgac541] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Indexed: 02/13/2023]
Abstract
The status of lipoprotein (a) [Lp(a)] as a cardiovascular risk factor has been resurrected by advances in genetics. Mendelian randomization studies show a causal link of Lp(a) with coronary artery disease (CAD), peripheral artery disease (PAD), and calcific aortic valve stenosis (CAVS). The genetics of Lp(a) is complex and extends beyond the kringle-IV type 2, as it is also dependent on ancestry. The plasma concentration of Lp(a) is determined by the hepatic production of apolipoprotein(a) [apo(a)] component of Lp(a), supporting the use of nucleic acids that inhibit the messenger RNA (mRNA) gene transcript for apo(a). Analytical barriers to measurement of Lp(a) are being addressed using isoform independent assays and a traceable standard. The association of Lp(a) and atherosclerotic cardiovascular disease is higher for myocardial infarction than PAD and CAVS. Increased risk of type 2 diabetes mellitus associated with low Lp(a) levels is perplexing and requires further investigation. The greatest advancement in Lp(a)-lowering therapies is based on using RNA therapeutics that are now being investigated in clinical trials. Proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibition lowers Lp(a) modestly, but whether cardiovascular benefit is independent of low-density lipoprotein lowering remains unclear. Opportunistic and selective testing for Lp(a) is supported by moderate evidence, with the case for universal screening premature. Modification of behavioral and clinical risk factors may be targeted to mitigate Lp(a)-mediated risk of cardiovascular disease. Clinical practice guidelines have been developed to address gaps in care of high Lp(a), but full implementation awaits the findings of clinical outcome trials using RNA-directed therapies currently underway.
Collapse
Affiliation(s)
- Paul Nestel
- Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Wann Jia Loh
- School of Medicine, University of Western Australia, Perth, Australia
- Department of Cardiology and Internal Medicine, Royal Perth Hospital, Perth, Australia
- Department of Endocrinology, Changi General Hospital, Singapore
- Duke-NUS Medical School, Singapore
| | - Natalie C Ward
- School of Medicine, University of Western Australia, Perth, Australia
| | - Gerald F Watts
- School of Medicine, University of Western Australia, Perth, Australia
- Department of Cardiology and Internal Medicine, Royal Perth Hospital, Perth, Australia
| |
Collapse
|
45
|
Pantazi D, Tellis C, Tselepis AD. Oxidized phospholipids and lipoprotein-associated phospholipase A 2 (Lp-PLA 2 ) in atherosclerotic cardiovascular disease: An update. Biofactors 2022; 48:1257-1270. [PMID: 36192834 DOI: 10.1002/biof.1890] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 09/06/2022] [Indexed: 12/24/2022]
Abstract
Inflammation and oxidative stress conditions lead to a variety of oxidative modifications of lipoprotein phospholipids implicated in the occurrence and development of atherosclerotic lesions. Lipoprotein-associated phospholipase A2 (Lp-PLA2 ) is established as an independent risk biomarker of atherosclerosis-related cardiovascular disease (ASCVD) and mediates vascular inflammation through the regulation of lipid metabolism in the blood and in atherosclerotic lesions. Lp-PLA2 is associated with low- and high-density lipoproteins and Lipoprotein (a) in human plasma and specifically hydrolyzes oxidized phospholipids involved in oxidative stress modification. Several oxidized phospholipids (OxPLs) subspecies can be detoxified through enzymatic degradation by Lp-PLA2 activation, forming lysophospholipids and oxidized non-esterified fatty acids (OxNEFAs). Lysophospholipids promote the expression of adhesion molecules, stimulate cytokines production (TNF-α, IL-6), and attract macrophages to the arterial intima. The present review article discusses new data on the functional roles of OxPLs and Lp-PLA2 associated with lipoproteins.
Collapse
Affiliation(s)
- Despoina Pantazi
- Atherothrombosis Research Centre/Laboratory of Biochemistry, Department of Chemistry, University of Ioannina, Ioannina, Greece
| | - Constantinos Tellis
- Atherothrombosis Research Centre/Laboratory of Biochemistry, Department of Chemistry, University of Ioannina, Ioannina, Greece
| | - Alexandros D Tselepis
- Atherothrombosis Research Centre/Laboratory of Biochemistry, Department of Chemistry, University of Ioannina, Ioannina, Greece
| |
Collapse
|
46
|
Elevated Lipoprotein(a) prevalence and association with family history of premature cardiovascular disease in general population with moderate cardiovascular risk and increased LDL cholesterol. IJC HEART & VASCULATURE 2022; 42:101100. [PMID: 35937950 PMCID: PMC9352902 DOI: 10.1016/j.ijcha.2022.101100] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 07/20/2022] [Accepted: 07/27/2022] [Indexed: 02/05/2023]
Abstract
Background Elevated Lipoprotein(a) [Lp(a)] is independently associated with increased cardiovascular disease (CVD) risk. There are discrepancies regarding its epidemiology due to great variability in different populations. This study aimed to evaluate the prevalence of elevated Lp(a) in people with moderate CVD risk and increased LDL-c and to determine the association between family history of premature CVD and elevated Lp(a). Methods Random subjects from the CESCAS population-based study of people with moderate CVD risk (Framingham score 10–20 %) and LDL-c ≥ 130 mg/dL, were selected to evaluate Lp(a) by immunoturbidimetry independent of the Isoforms variability. The association between family history of premature CVD and elevated Lp(a) was evaluated using multivariate logistic regression models. Elevated Lp(a) was defined as Lp(a) ≥ 125 nmol/L. Results Lp(a) was evaluated in 484 samples; men = 39.5 %, median age = 57 years (Q1-Q3: 50–63), mean CVD risk = 14.4 % (SE: 0.2), family history of premature CVD = 11.2 %, Lp(a) median of 21 nmol/L (Q1-Q3: 9–42 nmol/L), high Lp(a) = 6.1 % (95 % CI = 3.8–9.6). Association between family history of premature CVD and elevated Lp(a) in total population: OR 1.31 (95 % CI = 0.4, 4.2) p = 0.642; in subgroup of people with LDL-c ≥ 160 mg%, OR 4.24 (95 % CI = 1.2, 15.1) p = 0.026. Conclusions In general population with moderate CVD risk and elevated LDL-c from the Southern Cone of Latin America, less than one over ten people had elevated Lp(a). Family history of premature CVD was significantly associated with the presence of elevated Lp(a) in people with LDL-c ≥ 160 mg/dL.
Collapse
|
47
|
Tsimikas S, Marcovina SM. Ancestry, Lipoprotein(a), and Cardiovascular Risk Thresholds: JACC Review Topic of the Week. J Am Coll Cardiol 2022; 80:934-946. [PMID: 36007992 DOI: 10.1016/j.jacc.2022.06.019] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 06/28/2022] [Accepted: 06/30/2022] [Indexed: 12/24/2022]
Abstract
This study reviews ancestral differences in the genetics of the LPA gene, risk categories of elevated lipoprotein(a) [Lp(a)] as defined by guidelines, ancestry-specific Lp(a) risk, absolute and proportional risk, predictive value of risk thresholds among different ancestries, and differences between laboratory vs clinical accuracy in Lp(a) assays. For clinical decision-making, the preponderance of evidence suggests that the predictive value of Lp(a) does not vary sufficiently to mandate the use of ancestry-specific risk thresholds. This paper interprets the literature on Lp(a) and ancestral risk to support: 1) clinicians on understanding cardiovascular disease risk in different ancestral groups; 2) trialists for the design of clinical trials to ensure adequate ancestral diversity to support broad conclusions of drug effects; 3) regulators in the evaluation of the design and interpretation of results of Lp(a)-lowering trials with different Lp(a) inclusion thresholds; and 4) clinical laboratories to measure Lp(a) by assays that discriminate risk thresholds appropriately.
Collapse
Affiliation(s)
- Sotirios Tsimikas
- Division of Cardiovascular Medicine, Sulpizio Cardiovascular Center, University of California San Diego, La Jolla, California, USA.
| | | |
Collapse
|
48
|
Kronenberg F, Mora S, Stroes ESG, Ference BA, Arsenault BJ, Berglund L, Dweck MR, Koschinsky M, Lambert G, Mach F, McNeal CJ, Moriarty PM, Natarajan P, Nordestgaard BG, Parhofer KG, Virani SS, von Eckardstein A, Watts GF, Stock JK, Ray KK, Tokgözoğlu LS, Catapano AL. Lipoprotein(a) in atherosclerotic cardiovascular disease and aortic stenosis: a European Atherosclerosis Society consensus statement. Eur Heart J 2022; 43:3925-3946. [PMID: 36036785 PMCID: PMC9639807 DOI: 10.1093/eurheartj/ehac361] [Citation(s) in RCA: 394] [Impact Index Per Article: 197.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/10/2022] [Accepted: 06/21/2022] [Indexed: 12/20/2022] Open
Abstract
This 2022 European Atherosclerosis Society lipoprotein(a) [Lp(a)] consensus statement updates evidence for the role of Lp(a) in atherosclerotic cardiovascular disease (ASCVD) and aortic valve stenosis, provides clinical guidance for testing and treating elevated Lp(a) levels, and considers its inclusion in global risk estimation. Epidemiologic and genetic studies involving hundreds of thousands of individuals strongly support a causal and continuous association between Lp(a) concentration and cardiovascular outcomes in different ethnicities; elevated Lp(a) is a risk factor even at very low levels of low-density lipoprotein cholesterol. High Lp(a) is associated with both microcalcification and macrocalcification of the aortic valve. Current findings do not support Lp(a) as a risk factor for venous thrombotic events and impaired fibrinolysis. Very low Lp(a) levels may associate with increased risk of diabetes mellitus meriting further study. Lp(a) has pro-inflammatory and pro-atherosclerotic properties, which may partly relate to the oxidized phospholipids carried by Lp(a). This panel recommends testing Lp(a) concentration at least once in adults; cascade testing has potential value in familial hypercholesterolaemia, or with family or personal history of (very) high Lp(a) or premature ASCVD. Without specific Lp(a)-lowering therapies, early intensive risk factor management is recommended, targeted according to global cardiovascular risk and Lp(a) level. Lipoprotein apheresis is an option for very high Lp(a) with progressive cardiovascular disease despite optimal management of risk factors. In conclusion, this statement reinforces evidence for Lp(a) as a causal risk factor for cardiovascular outcomes. Trials of specific Lp(a)-lowering treatments are critical to confirm clinical benefit for cardiovascular disease and aortic valve stenosis.
Collapse
Affiliation(s)
- Florian Kronenberg
- Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Samia Mora
- Center for Lipid Metabolomics, Division of Preventive Medicine, and Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Erik S G Stroes
- Department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Brian A Ference
- Centre for Naturally Randomized Trials, University of Cambridge, Cambridge, UK
| | - Benoit J Arsenault
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, and Department of Medicine, Faculty of Medicine, Université Laval, Québec City, QC, Canada
| | - Lars Berglund
- Department of Internal Medicine, School of Medicine, University of California-Davis, Davis, Sacramento, CA, USA
| | - Marc R Dweck
- British Heart Foundation Centre for Cardiovascular Science, Edinburgh Heart Centre, University of Edinburgh, Chancellors Building, Little France Crescent, Edinburgh EH16 4SB, UK
| | - Marlys Koschinsky
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Gilles Lambert
- Inserm, UMR 1188 Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, 97400 Saint-Denis de La Reunion, France
| | - François Mach
- Department of Cardiology, Geneva University Hospital, Geneva, Switzerland
| | - Catherine J McNeal
- Division of Cardiology, Department of Internal Medicine, Baylor Scott & White Health, 2301 S. 31st St., USA
| | | | - Pradeep Natarajan
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, and Program in Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Børge G Nordestgaard
- Department of Clinical Biochemistry and the Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Klaus G Parhofer
- Medizinische Klinik und Poliklinik IV, Ludwigs- Maximilians University Klinikum, Munich, Germany
| | - Salim S Virani
- Section of Cardiovascular Research, Baylor College of Medicine & Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, USA
| | - Arnold von Eckardstein
- Institute of Clinical Chemistry, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Gerald F Watts
- Medical School, University of Western Australia, and Department of Cardiology, Lipid Disorders Clinic, Royal Perth Hospital, Perth, Australia
| | - Jane K Stock
- European Atherosclerosis Society, Mässans Gata 10, SE-412 51 Gothenburg, Sweden
| | - Kausik K Ray
- Imperial Centre for Cardiovascular Disease Prevention, Department of Primary Care and Public Health, School of Public Health, Imperial College London, London, UK
| | - Lale S Tokgözoğlu
- Department of Cardiology, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Alberico L Catapano
- Department of Pharmacological and Biomolecular Sciences, University of Milano, Milano, Italy.,IRCCS Multimedica, Milano, Italy
| |
Collapse
|
49
|
Song J, Zhang X, Wei M, Bo Y, Zhou X, Tang B. Association between lipoprotein(a) and thromboembolism in patients with non-valvular atrial fibrillation: a cross-sectional study. Lipids Health Dis 2022; 21:78. [PMID: 36002888 PMCID: PMC9404645 DOI: 10.1186/s12944-022-01682-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 07/29/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Lipoprotein(a) [Lp(a)] is a recognized risk factor for ischemic stroke (IS); however, its role in thromboembolism in patients with non-valvular atrial fibrillation (NVAF) remains controversial. We aimed to assess the association of Lp(a) and IS and systemic embolism (SEE) in NVAF patients. METHODS In total, 16,357 patients with NVAF were recruited from the First Affiliated Hospital of Xinjiang Medical University from January 1, 2009, to December 31, 2021, and were divided into groups based on Lp(a) quartiles. Logistic regression models analyzed the association between Lp(a), IS, and SEE. The restriction cubic spline was used to assess the potential nonlinear relationship between Lp(a), IS, and SEE. We conducted subgroup analyses and estimated the multiplicative interaction between the stratified variables and Lp(a) to investigate whether the association between Lp(a) and IS and SEE was affected by age, sex, anticoagulants, and CHA2DS2-VASc score. RESULTS We identified 1319 IS and 133 SEE events. After correcting for CHA2DS2-VASc score and other potential confounders, each 1-standard deviation (SD) increase in log-Lp(a) was related to a 23% increased risk of IS (odds ratios [OR], 1.23; 95% confidence intervals [CI], 1.07-1.41). NVAF patients in the highest Lp(a) quartile were 1.23-fold more likely to have IS than those in the lowest quartile (OR, 1.23; 95% CI, 1.04-1.45). A positive linear relationship between Lp(a) and IS risk was observed (P for nonlinear = 0.341). In the fully adjusted model, subjects had a 1.78-fold increased risk of SEE for each 1-SD increase in log-Lp(a) (OR, 2.78; 95% CI, 1.78-4.36). Subjects in the highest Lp(a) quartile had a 2.38-fold elevated risk of SEE (OR, 3.38; 95% CI, 1.85-6.19) compared with the lowest quartile. Furthermore, Lp(a) had a nonlinear relationship with the risk of SEE (P for nonlinear = 0.005). CONCLUSIONS Elevated Lp(a) concentration was significantly associated with IS and SEE, suggesting that Lp(a) may be an emerging biomarker that can help clinicians identify patients at high risk of thromboembolism in this population.
Collapse
Affiliation(s)
- Jie Song
- Department of Cardiac Pacing and Electrophysiology, The First Affiliated Hospital of Xinjiang Medical University, No. 137, Liyushan Road, Urumqi, 830054, PR China.,Xinjiang Key Laboratory of Cardiac Electrophysiology and Remodeling, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, PR China
| | - Xiaoxue Zhang
- Department of Cardiac Pacing and Electrophysiology, The First Affiliated Hospital of Xinjiang Medical University, No. 137, Liyushan Road, Urumqi, 830054, PR China.,Xinjiang Key Laboratory of Cardiac Electrophysiology and Remodeling, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, PR China
| | - Meng Wei
- Department of outpatient, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, PR China
| | - Yakun Bo
- Department of Cardiac Pacing and Electrophysiology, The First Affiliated Hospital of Xinjiang Medical University, No. 137, Liyushan Road, Urumqi, 830054, PR China.,Xinjiang Key Laboratory of Cardiac Electrophysiology and Remodeling, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, PR China
| | - Xianhui Zhou
- Department of Cardiac Pacing and Electrophysiology, The First Affiliated Hospital of Xinjiang Medical University, No. 137, Liyushan Road, Urumqi, 830054, PR China. .,Xinjiang Key Laboratory of Cardiac Electrophysiology and Remodeling, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, PR China.
| | - Baopeng Tang
- Department of Cardiac Pacing and Electrophysiology, The First Affiliated Hospital of Xinjiang Medical University, No. 137, Liyushan Road, Urumqi, 830054, PR China. .,Xinjiang Key Laboratory of Cardiac Electrophysiology and Remodeling, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, PR China.
| |
Collapse
|
50
|
Clarke R, Hammami I, Sherliker P, Valdes-Marquez E, Watkins H, Hill M, Yang X, Tsimikas S, Hopewell JC. Oxidized phospholipids on apolipoprotein B-100 versus plasminogen and risk of coronary heart disease in the PROCARDIS study. Atherosclerosis 2022; 354:15-22. [PMID: 35803063 DOI: 10.1016/j.atherosclerosis.2022.06.1020] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 06/16/2022] [Accepted: 06/22/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND AND AIMS Oxidized phospholipids carried on the apolipoprotein B-100 (OxPL-apoB) component of Lp(a) are predictive of coronary heart disease (CHD), but the role of oxidized phospholipids carried on plasminogen (OxPL-PLG) is unknown. We examined the independent effects of OxPL-apoB and OxPL-PLG for risk of CHD before and after adjustment for Lp(a). METHODS Plasma levels of OxPL-apoB, OxPL-PLG, plasminogen and Lp(a) were measured in the PROCARDIS study of early-onset CHD (906 cases/858 controls). Multivariable logistic regression was used to estimate the odds ratios (OR) for each biomarker with CHD after adjustment for established risk factors. RESULTS Mean levels of OxPL-apoB were higher in cases than controls, but levels of OxPL-PLG and plasminogen were similar. For OxPL-apoB, individuals in the top vs bottom fifth had 2-fold higher age and sex-adjusted OR of CHD (OR = 2.61 [95%CI: 1.91, 3.55]), which were partially attenuated after adjustment for established risk factors. The findings for OxPL-apoB and CHD in PROCARDIS were comparable with those of a meta-analysis of all such studies. However, the associations of OxPL-apoB with CHD were fully attenuated by additional adjustment for Lp(a) (OR = 0.93 [0.54,1.60]). Neither OxPL-PLG nor plasminogen were associated with CHD. Overall, there were no differences in the predictive value for CHD of high vs normal levels (<20th or >80th percentile) of OxPL-apoB, OxPL-PLG, plasminogen or Lp(a) after stratifying for each other. CONCLUSIONS These results highlight the context-dependency of OxPL in plasma and suggest that their associated risk of CHD is chiefly mediated by their carriage on Lp(a).
Collapse
Affiliation(s)
- Robert Clarke
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom.
| | - Imen Hammami
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Paul Sherliker
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Elsa Valdes-Marquez
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Hugh Watkins
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Michael Hill
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Xiaohong Yang
- Division of Cardiovascular Diseases, University of California, San Diego, USA
| | - Sotirios Tsimikas
- Division of Cardiovascular Diseases, University of California, San Diego, USA
| | - Jemma C Hopewell
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom.
| | | |
Collapse
|