1
|
Dalla Vestra M, Grolla E, Autiero G, Presotto F. Peripheral artery disease risk factors: A focus on lipoprotein(a). Arch Cardiovasc Dis 2024; 117:584-589. [PMID: 39227282 DOI: 10.1016/j.acvd.2024.07.061] [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: 02/20/2024] [Revised: 07/04/2024] [Accepted: 07/14/2024] [Indexed: 09/05/2024]
Abstract
There is a well-established and strong link between high lipoprotein(a) concentration and coronary heart disease, but the evidence regarding peripheral artery disease and carotid atherosclerosis is not as conclusive. This review aims to summarize the relationships between lipoprotein(a), peripheral artery disease and carotid atherosclerosis, in order to try to understand the weight of lipoprotein(a) in determining the development, progression and any complications of atherosclerotic plaque at the carotid and peripheral artery level. There is currently no effective therapy to reduce lipoprotein(a) concentration, but understanding its significance as a vascular risk factor is the starting point to then explore (when effective therapies become available) if there is the possibility, even in patients with peripheral artery disease and carotid atherosclerosis, to achieve better control of the residual vascular risk that is ultimately induced by lipoprotein(a).
Collapse
Affiliation(s)
- Michele Dalla Vestra
- Department of Internal Medicine, Ospedale dell'Angelo, 30174 Mestre-Venezia, Italy.
| | - Elisabetta Grolla
- Department of Cardiology, Ospedale dell'Angelo, 30174 Mestre-Venezia, Italy
| | - Giuliana Autiero
- Department of Internal Medicine, Ospedale dell'Angelo, 30174 Mestre-Venezia, Italy
| | - Fabio Presotto
- Department of Internal Medicine, Ospedale dell'Angelo, 30174 Mestre-Venezia, Italy
| |
Collapse
|
2
|
Cancro FP, Bellino M, Silverio A, Di Maio M, Esposito L, Palumbo R, Manna ML, Formisano C, Ferruzzi G, Vecchione C, Galasso G. Novel Targets and Strategies Addressing Residual Cardiovascular Risk in Post-acute Coronary Syndromes Patients. Transl Med UniSa 2024; 26:99-110. [PMID: 39385797 PMCID: PMC11460530 DOI: 10.37825/2239-9747.1058] [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: 12/30/2023] [Revised: 07/03/2024] [Accepted: 07/08/2024] [Indexed: 10/12/2024] Open
Abstract
Despite the advancement in secondary cardiovascular prevention strategies for post-acute coronary syndrome (ACS) patients, the development of new drugs addressing dyslipidemia and the personalization of dual antiplatelet therapies (DAPT), these patients continue to suffer a significant incidence of recurrent ischemic events. Therefore, novel targets that can be tackled to reduce cardiovascular risk are needed to improve the outcome of this very high-risk population. The role of chronic inflammation and inflammasome in the development and progression of atherosclerosis has been broadly investigated in patients with established coronary artery disease (CAD) and recent randomized trials have highlighted the possibility to manage these targets with specific drugs such as colchicine and monocolonal antibodies with a significant improvement of cardiovascular outcomes in post-ACS patients. Lipoprotein(a) [Lp(a)] is the most promising non-traditional risk factor and has shown to predict worse outcome in post-ACS patients. Lowering Lp(a) through PCSK9 inhibitors and specific targeted therapies has shown positive results in reducing adverse cardiovascular events in patients with established CAD. The effect of microbiome and its alteration in gut dysbiosis seems to actively participate in residual cardiovascular risk of CAD patients; however, the risk-modifying effect of targeted-microbiome therapies hasn't been yet investigated in large population-based studies. Long-term outcome of post-ACS patients is a complex puzzle of multiple factors. In this minireview, we summarize the emerging risk factors that may interplay in the residual risk of post-ACS patients and their possible prognostic and therapeutic implications.
Collapse
Affiliation(s)
- Francesco P. Cancro
- Department of Medicine Surgery and Dentistry, University of Salerno, Baronissi, SA,
Italy
| | - Michele Bellino
- Department of Medicine Surgery and Dentistry, University of Salerno, Baronissi, SA,
Italy
| | - Angelo Silverio
- Department of Medicine Surgery and Dentistry, University of Salerno, Baronissi, SA,
Italy
| | - Marco Di Maio
- Department of Medicine Surgery and Dentistry, University of Salerno, Baronissi, SA,
Italy
| | - Luca Esposito
- Department of Medicine Surgery and Dentistry, University of Salerno, Baronissi, SA,
Italy
| | - Rossana Palumbo
- Department of Medicine Surgery and Dentistry, University of Salerno, Baronissi, SA,
Italy
| | - Martina L. Manna
- Department of Medicine Surgery and Dentistry, University of Salerno, Baronissi, SA,
Italy
| | - Ciro Formisano
- Department of Medicine Surgery and Dentistry, University of Salerno, Baronissi, SA,
Italy
| | - Germano Ferruzzi
- Department of Medicine Surgery and Dentistry, University of Salerno, Baronissi, SA,
Italy
| | - Carmine Vecchione
- Department of Medicine Surgery and Dentistry, University of Salerno, Baronissi, SA,
Italy
| | - Gennaro Galasso
- Department of Medicine Surgery and Dentistry, University of Salerno, Baronissi, SA,
Italy
| |
Collapse
|
3
|
Min T, Yue Y, Fan X, Yang D, Su S, Wan H. The levels of serum lipoprotein(a) on clinical outcomes in Chinese hospitalized patients with cardiovascular diseases. Eur J Med Res 2024; 29:421. [PMID: 39152471 PMCID: PMC11330145 DOI: 10.1186/s40001-024-01957-7] [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: 09/02/2023] [Accepted: 07/01/2024] [Indexed: 08/19/2024] Open
Abstract
OBJECTIVE Serum lipoprotein(a) [Lp(a)] is a risk factor of cardiovascular diseases. However, the relationship between the serum Lp(a) and clinical outcomes has been seldom studied in Chinese hospitalized patients with cardiovascular diseases. METHODS We retrospectively collected the clinical data of hospitalized patients with cardiovascular diseases in the Cardiovascular Department of Dongguan People's Hospital from 2016 to 2021 through the electronic case system. Patients were divided into 4 groups based on Lp(a) quartiles: Quartile1 (≤ 80.00 mg/L), Quartile 2 (80.01 ~ 160.90 mg/L), Quartile 3 (160.91 ~ 336.41 mg/L), Quartile 4 (> 336.41 mg/L). Cox proportional hazard regression models were constructed to examine the relationship between Lp(a) and cardiovascular events. RESULTS A total of 8382 patients were included in this study. After an average follow-up of 619 (320 to 1061) days, 1361 (16.2%) patients developed major adverse cardiovascular events, and 125 (1.5%) all-cause death were collected. The incidence of MACEs was 7.65, 8.24, 9.73 and 10.75 per 100 person-years in each Lp(a) quartile, respectively; the all-cause mortality was 0.48, 0.69, 0.64 and 1.18 per 100 person-years in each Lp(a) quartile, respectively. The multivariate Cox regression analysis suggested that high Lp(a) level was an independent risk factor for MACEs (HR: 1.189, [95% CI: 1.045 to 1.353], P = 0.030) and all-cause death (HR: 1.573, [95% CI: 1.009 to 2.452], P = 0.046). CONCLUSION In addition to traditional lipid indicators, higher Lp(a) exhibited higher risks of adverse cardiovascular events and death, indicated worse prognosis. Lp(a) may be a new target for the prevention of atherosclerotic diseases.
Collapse
Affiliation(s)
- Tingting Min
- Department of Cardiology, Dongguan People's Hospital, The First School of Clinical Medicine, Southern Medical University, Dongguan, 523059, Guangdong, People's Republic of China
| | - Yilin Yue
- Department of Cardiology, Shenhe People's Hospital, The Fifth Affiliated Hospital of Jinan University, Heyuan, 517000, Guangdong, People's Republic of China
| | - Xin Fan
- Department of Cardiology, Dongguan People's Hospital, The First School of Clinical Medicine, Southern Medical University, Dongguan, 523059, Guangdong, People's Republic of China
| | - Deguang Yang
- Department of Cardiology, Shenhe People's Hospital, The Fifth Affiliated Hospital of Jinan University, Heyuan, 517000, Guangdong, People's Republic of China
| | - Shaohui Su
- Department of Cardiology, Dongguan People's Hospital, The First School of Clinical Medicine, Southern Medical University, Dongguan, 523059, Guangdong, People's Republic of China.
| | - Huaibin Wan
- Department of Cardiology, Shenhe People's Hospital, The Fifth Affiliated Hospital of Jinan University, Heyuan, 517000, Guangdong, People's Republic of China.
| |
Collapse
|
4
|
Wang Z, Yan X, Fang L, Tang J, Zhang J. Association between lipoprotein(a), fibrinogen and their combination with all-cause, cardiovascular disease and cancer-related mortality: findings from the NHANES. BMC Public Health 2024; 24:1927. [PMID: 39026192 PMCID: PMC11256372 DOI: 10.1186/s12889-024-19443-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 07/10/2024] [Indexed: 07/20/2024] Open
Abstract
BACKGROUND There is evidence indicating that both lipoprotein(a) [Lp(a)] and fibrinogen (FIB) are associated with mortality, However, the impact of their combination on mortality has not been determined. Thus, the aim of this study was to examine the association between the combination of Lp(a) and FIB with all-cause and cause-specific mortality. METHODS This prospective cohort study enrolled 4,730 participants from the third National Health and Nutrition Examination Survey. The exposure variables included Lp(a), FIB and their combination, while the outcome variables consisted of all-cause, cardiovascular disease (CVD) and cancer-related mortality. Multivariate COX regression, subgroup analysis, sensitivity analysis and restricted cubic spline (RCS) were used to investigate the association between Lp(a), FIB and their combination with all-cause, CVD and cancer-related mortality. RESULTS Over a median follow-up period of 235 months, 2,668 individuals died, including 1,051 deaths attributed to CVD and 549 deaths due to cancer. Multivariate Cox regression analyses revealed independent associations between both Lp(a) and FIB with all-cause, CVD, and cancer-related mortality. Compared to participants in the 1st to 50th percentiles of both Lp(a) and FIB, those in the 90th to 100th percentiles exhibited multivariable adjusted HRs of 1.813 (95% CI: 1.419-2.317, P < 0.001), 2.147 (95% CI: 1.483-3.109, P < 0.001) and 2.355 (95% CI: 1.396, 3.973, P = 0.001) for all-cause, CVD and cancer-related mortality, respectively. Subgroup and sensitivity analyses did not substantially attenuate the association between the combination of high Lp(a) and high FIB with the risk of all-cause and CVD-related mortality. Additionally, the RCS analysis showed that the relationship between Lp(a) and the risk of all-cause and cancer-related mortality, as well as the relationship between FIB and the risk of cancer-related mortality, were linear (P for nonlinearity > 0.05). Conversely, the relationship between Lp(a) and the risk of CVD-related mortality, as well as the relationship between FIB and the risk of all-cause and CVD-related mortality, were nonlinear (P for nonlinearity < 0.05). CONCLUSIONS High levels of Lp(a) and FIB together conferred a greater risk of mortality from all-cause, CVD and cancer.
Collapse
Grants
- 81900453, 82222007, 82170281, and U2004203 National Natural Science Foundation of China
- 81900453, 82222007, 82170281, and U2004203 National Natural Science Foundation of China
- 81900453, 82222007, 82170281, and U2004203 National Natural Science Foundation of China
- Hohhot Healthcare Medical-2023030 Hohhot Healthcare Science and Technology Programme
- ZYQR201912131 Henan Thousand Talents Program
- 202300410362 Excellent Youth Science Foundation of Henan Province
- 2021-CCA-ACCESS-125 Central Plains Youth Top Talent, Advanced funds
- SBGJ202101012 Henan Province Medical Science and Technology Key Joint Project
- 222102230025 Key Scientific and Technological Research Projects in Henan Province
Collapse
Affiliation(s)
- Zhenwei Wang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, 450018, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, 450052, China
| | - Xuejiao Yan
- Department of Cardiology, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou, 213003, China
| | - Lijuan Fang
- Department of Cardiology, The First Hospital of Hohhot, Hohhot, 010030, China.
| | - Junnan Tang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China.
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, 450018, China.
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, 450052, China.
| | - Jinying Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China.
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, 450018, China.
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, 450052, China.
| |
Collapse
|
5
|
Pavlatos N, Kalra DK. The Role of Lipoprotein(a) in Peripheral Artery Disease. Biomedicines 2024; 12:1229. [PMID: 38927436 PMCID: PMC11200468 DOI: 10.3390/biomedicines12061229] [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: 05/05/2024] [Revised: 05/25/2024] [Accepted: 05/29/2024] [Indexed: 06/28/2024] Open
Abstract
Lipoprotein(a) is a low-density-lipoprotein-like particle that consists of apolipoprotein(a) bound to apolipoprotein(b). It has emerged as an established causal risk factor for atherosclerotic cardiovascular disease, stroke, and aortic valve stenosis through multifactorial pathogenic mechanisms that include inflammation, atherogenesis, and thrombosis. Despite an estimated 20% of the global population having elevated lipoprotein(a) levels, testing remains underutilized due to poor awareness and a historical lack of effective and safe therapies. Although lipoprotein(a) has a strong association with coronary artery disease and cerebrovascular disease, its relationship with peripheral artery disease is less well established. In this article, we review the epidemiology, biology, and pathogenesis of lipoprotein(a) as it relates to peripheral artery disease. We also discuss emerging treatment options to help mitigate major adverse cardiac and limb events in this population.
Collapse
Affiliation(s)
- Nicholas Pavlatos
- Department of Internal Medicine, University of Louisville School of Medicine, Louisville, KY 40202, USA;
| | - Dinesh K. Kalra
- Division of Cardiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| |
Collapse
|
6
|
Wańczura P, Aebisher D, Iwański MA, Myśliwiec A, Dynarowicz K, Bartusik-Aebisher D. The Essence of Lipoproteins in Cardiovascular Health and Diseases Treated by Photodynamic Therapy. Biomedicines 2024; 12:961. [PMID: 38790923 PMCID: PMC11117957 DOI: 10.3390/biomedicines12050961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 04/22/2024] [Accepted: 04/24/2024] [Indexed: 05/26/2024] Open
Abstract
Lipids, together with lipoprotein particles, are the cause of atherosclerosis, which is a pathology of the cardiovascular system. In addition, it affects inflammatory processes and affects the vessels and heart. In pharmaceutical answer to this, statins are considered a first-stage treatment method to block cholesterol synthesis. Many times, additional drugs are also used with this method to lower lipid concentrations in order to achieve certain values of low-density lipoprotein (LDL) cholesterol. Recent advances in photodynamic therapy (PDT) as a new cancer treatment have gained the therapy much attention as a minimally invasive and highly selective method. Photodynamic therapy has been proven more effective than chemotherapy, radiotherapy, and immunotherapy alone in numerous studies. Consequently, photodynamic therapy research has expanded in many fields of medicine due to its increased therapeutic effects and reduced side effects. Currently, PDT is the most commonly used therapy for treating age-related macular degeneration, as well as inflammatory diseases, and skin infections. The effectiveness of photodynamic therapy against a number of pathogens has also been demonstrated in various studies. Also, PDT has been used in the treatment of cardiovascular diseases, such as atherosclerosis and hyperplasia of the arterial intima. This review evaluates the effectiveness and usefulness of photodynamic therapy in cardiovascular diseases. According to the analysis, photodynamic therapy is a promising approach for treating cardiovascular diseases and may lead to new clinical trials and management standards. Our review addresses the used therapeutic strategies and also describes new therapeutic strategies to reduce the cardiovascular burden that is induced by lipids.
Collapse
Affiliation(s)
- Piotr Wańczura
- Department of Cardiology, Medical College of the University of Rzeszów, 35-310 Rzeszów, Poland
| | - David Aebisher
- Department of Photomedicine and Physical Chemistry, Medical College of the University of Rzeszów, 35-310 Rzeszów, Poland
| | - Mateusz A Iwański
- English Division Science Club, Medical College of the University of Rzeszów, 35-310 Rzeszów, Poland
| | - Angelika Myśliwiec
- Center for Innovative Research in Medical and Natural Sciences, Medical College of the University of Rzeszów, 35-310 Rzeszów, Poland
| | - Klaudia Dynarowicz
- Center for Innovative Research in Medical and Natural Sciences, Medical College of the University of Rzeszów, 35-310 Rzeszów, Poland
| | - Dorota Bartusik-Aebisher
- Department of Biochemistry and General Chemistry, Medical College of the University of Rzeszów, 35-310 Rzeszów, Poland
| |
Collapse
|
7
|
Dong W, Zhong X, Yuan K, Miao M, Zhai Y, Che B, Xu T, Xu X, Zhong C. Lipoprotein(a) and functional outcome of acute ischemic stroke when discordant with low-density lipoprotein cholesterol. Postgrad Med J 2023; 99:1160-1166. [PMID: 37624118 DOI: 10.1093/postmj/qgad070] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/23/2023] [Accepted: 07/28/2023] [Indexed: 08/26/2023]
Abstract
BACKGROUND Several studies have indicated that residual cardiovascular risk might be associated with elevated lipoprotein(a) [Lp(a)] even in the setting of controlled low-density lipoprotein cholesterol (LDL-C). We aimed to prospectively examine the association between Lp(a) and unfavorable functional outcome among patients with acute ischemic stroke when Lp(a) and LDL-C were discordant. METHODS Based on samples from the Infectious Factors, Inflammatory Markers and Prognosis of Acute Ischemic Stroke study, 973 patients with baseline plasma Lp(a) levels were included. The primary outcome was the composite outcome of death or major disability (modified Rankin Scale score of 3-6) at 6 months. Logistic regression models were used to estimate the risk for the primary outcome. Discordance analyses were performed, using difference in percentile units (>10 units), to detect the relative risk when Lp(a) and LDL-C were discordant. RESULTS In total, 201 (20.7%) participants experienced major disability or death at 6 months. The multivariable-adjusted odds ratio (OR) for the highest quartile was 1.88 [95% confidence interval (CI): 1.16-3.04] compared with the lowest quartile. Each 1-SD higher log-Lp(a) was associated with a 23% increased risk (95% CI: 2%-47%) for the primary outcome. Compared with the concordant group, the high Lp(a)/low LDL-C discordant group was associated with increased risk for the primary outcome (adjusted OR: 1.59, 95% CI: 1.01-2.52). CONCLUSIONS Elevated plasma Lp(a) levels were associated with increased risk of major disability and death at 6 months. Discordantly high Lp(a)/low LDL-C was associated with an unfavorable functional outcome, supporting the predictive potential of plasma Lp(a) after ischemic stroke, especially when discordant with LDL-C. Key messages What is already known on this topic Previous studies have indicated that a positive association between increased lipoprotein(a) [Lp(a)] and cardiovascular disease risk remained even in patients who achieved controlled low-density lipoprotein cholesterol (LDL-C) levels. The findings of studies exploring the association between Lp(a) and unfavorable clinical outcomes of stroke were inconsistent, and whether Lp(a) can predict the risk of unfavorable functional outcome in stroke patients when Lp(a) and LDL-C levels are discordant remains unknown. What this study adds Elevated plasma Lp(a) levels were associated with increased risk of major disability and death at 6 months beyond LDL-C levels in acute ischemic stroke patients. How this study might affect research, practice, or policy The combination of LDL-C-lowering therapies and Lp(a)-lowering therapies may have better clinical efficacy for patients with ischemic stroke, and it is of great clinical interest to further explore this possibility in dedicated randomized trials.
Collapse
Affiliation(s)
- Wenjing Dong
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, Suzhou, 215123, China
| | - Xiaoyan Zhong
- School of Public Health, Suzhou Medical College of Soochow University, Suzhou, 215123, China
| | - Ke Yuan
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, Suzhou, 215123, China
| | - Mengyuan Miao
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, Suzhou, 215123, China
| | - Yujia Zhai
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, Suzhou, 215123, China
| | - Bizhong Che
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, Suzhou, 215123, China
| | - Tan Xu
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, Suzhou, 215123, China
| | - Xiang Xu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Chongke Zhong
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, Suzhou, 215123, China
| |
Collapse
|
8
|
Vinci P, Di Girolamo FG, Panizon E, Tosoni LM, Cerrato C, Pellicori F, Altamura N, Pirulli A, Zaccari M, Biasinutto C, Roni C, Fiotti N, Schincariol P, Mangogna A, Biolo G. Lipoprotein(a) as a Risk Factor for Cardiovascular Diseases: Pathophysiology and Treatment Perspectives. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:6721. [PMID: 37754581 PMCID: PMC10531345 DOI: 10.3390/ijerph20186721] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 07/31/2023] [Accepted: 08/09/2023] [Indexed: 09/28/2023]
Abstract
Cardiovascular disease (CVD) is still a leading cause of morbidity and mortality, despite all the progress achieved as regards to both prevention and treatment. Having high levels of lipoprotein(a) [Lp(a)] is a risk factor for cardiovascular disease that operates independently. It can increase the risk of developing cardiovascular disease even when LDL cholesterol (LDL-C) levels are within the recommended range, which is referred to as residual cardiovascular risk. Lp(a) is an LDL-like particle present in human plasma, in which a large plasminogen-like glycoprotein, apolipoprotein(a) [Apo(a)], is covalently bound to Apo B100 via one disulfide bridge. Apo(a) contains one plasminogen-like kringle V structure, a variable number of plasminogen-like kringle IV structures (types 1-10), and one inactive protease region. There is a large inter-individual variation of plasma concentrations of Lp(a), mainly ascribable to genetic variants in the Lp(a) gene: in the general po-pulation, Lp(a) levels can range from <1 mg/dL to >1000 mg/dL. Concentrations also vary between different ethnicities. Lp(a) has been established as one of the risk factors that play an important role in the development of atherosclerotic plaque. Indeed, high concentrations of Lp(a) have been related to a greater risk of ischemic CVD, aortic valve stenosis, and heart failure. The threshold value has been set at 50 mg/dL, but the risk may increase already at levels above 30 mg/dL. Although there is a well-established and strong link between high Lp(a) levels and coronary as well as cerebrovascular disease, the evidence regarding incident peripheral arterial disease and carotid atherosclerosis is not as conclusive. Because lifestyle changes and standard lipid-lowering treatments, such as statins, niacin, and cholesteryl ester transfer protein inhibitors, are not highly effective in reducing Lp(a) levels, there is increased interest in developing new drugs that can address this issue. PCSK9 inhibitors seem to be capable of reducing Lp(a) levels by 25-30%. Mipomersen decreases Lp(a) levels by 25-40%, but its use is burdened with important side effects. At the current time, the most effective and tolerated treatment for patients with a high Lp(a) plasma level is apheresis, while antisense oligonucleotides, small interfering RNAs, and microRNAs, which reduce Lp(a) levels by targeting RNA molecules and regulating gene expression as well as protein production levels, are the most widely explored and promising perspectives. The aim of this review is to provide an update on the current state of the art with regard to Lp(a) pathophysiological mechanisms, focusing on the most effective strategies for lowering Lp(a), including new emerging alternative therapies. The purpose of this manuscript is to improve the management of hyperlipoproteinemia(a) in order to achieve better control of the residual cardiovascular risk, which remains unacceptably high.
Collapse
Affiliation(s)
- Pierandrea Vinci
- Clinica Medica, Cattinara Hospital, Department of Medical Surgical and Health Science, University of Trieste, 34149 Trieste, Italy; (F.G.D.G.); (E.P.); (L.M.T.); (C.C.); (F.P.); (N.A.); (A.P.); (M.Z.); (N.F.); (G.B.)
| | - Filippo Giorgio Di Girolamo
- Clinica Medica, Cattinara Hospital, Department of Medical Surgical and Health Science, University of Trieste, 34149 Trieste, Italy; (F.G.D.G.); (E.P.); (L.M.T.); (C.C.); (F.P.); (N.A.); (A.P.); (M.Z.); (N.F.); (G.B.)
- SC Assistenza Farmaceutica, Cattinara Hospital, Azienda Sanitaria Universitaria Integrata di Trieste, 34149 Trieste, Italy; (C.B.); (C.R.); (P.S.)
| | - Emiliano Panizon
- Clinica Medica, Cattinara Hospital, Department of Medical Surgical and Health Science, University of Trieste, 34149 Trieste, Italy; (F.G.D.G.); (E.P.); (L.M.T.); (C.C.); (F.P.); (N.A.); (A.P.); (M.Z.); (N.F.); (G.B.)
| | - Letizia Maria Tosoni
- Clinica Medica, Cattinara Hospital, Department of Medical Surgical and Health Science, University of Trieste, 34149 Trieste, Italy; (F.G.D.G.); (E.P.); (L.M.T.); (C.C.); (F.P.); (N.A.); (A.P.); (M.Z.); (N.F.); (G.B.)
| | - Carla Cerrato
- Clinica Medica, Cattinara Hospital, Department of Medical Surgical and Health Science, University of Trieste, 34149 Trieste, Italy; (F.G.D.G.); (E.P.); (L.M.T.); (C.C.); (F.P.); (N.A.); (A.P.); (M.Z.); (N.F.); (G.B.)
| | - Federica Pellicori
- Clinica Medica, Cattinara Hospital, Department of Medical Surgical and Health Science, University of Trieste, 34149 Trieste, Italy; (F.G.D.G.); (E.P.); (L.M.T.); (C.C.); (F.P.); (N.A.); (A.P.); (M.Z.); (N.F.); (G.B.)
| | - Nicola Altamura
- Clinica Medica, Cattinara Hospital, Department of Medical Surgical and Health Science, University of Trieste, 34149 Trieste, Italy; (F.G.D.G.); (E.P.); (L.M.T.); (C.C.); (F.P.); (N.A.); (A.P.); (M.Z.); (N.F.); (G.B.)
| | - Alessia Pirulli
- Clinica Medica, Cattinara Hospital, Department of Medical Surgical and Health Science, University of Trieste, 34149 Trieste, Italy; (F.G.D.G.); (E.P.); (L.M.T.); (C.C.); (F.P.); (N.A.); (A.P.); (M.Z.); (N.F.); (G.B.)
| | - Michele Zaccari
- Clinica Medica, Cattinara Hospital, Department of Medical Surgical and Health Science, University of Trieste, 34149 Trieste, Italy; (F.G.D.G.); (E.P.); (L.M.T.); (C.C.); (F.P.); (N.A.); (A.P.); (M.Z.); (N.F.); (G.B.)
| | - Chiara Biasinutto
- SC Assistenza Farmaceutica, Cattinara Hospital, Azienda Sanitaria Universitaria Integrata di Trieste, 34149 Trieste, Italy; (C.B.); (C.R.); (P.S.)
| | - Chiara Roni
- SC Assistenza Farmaceutica, Cattinara Hospital, Azienda Sanitaria Universitaria Integrata di Trieste, 34149 Trieste, Italy; (C.B.); (C.R.); (P.S.)
| | - Nicola Fiotti
- Clinica Medica, Cattinara Hospital, Department of Medical Surgical and Health Science, University of Trieste, 34149 Trieste, Italy; (F.G.D.G.); (E.P.); (L.M.T.); (C.C.); (F.P.); (N.A.); (A.P.); (M.Z.); (N.F.); (G.B.)
| | - Paolo Schincariol
- SC Assistenza Farmaceutica, Cattinara Hospital, Azienda Sanitaria Universitaria Integrata di Trieste, 34149 Trieste, Italy; (C.B.); (C.R.); (P.S.)
| | - Alessandro Mangogna
- Institute for Maternal and Child Health, I.R.C.C.S “Burlo Garofolo”, 34137 Trieste, Italy;
| | - Gianni Biolo
- Clinica Medica, Cattinara Hospital, Department of Medical Surgical and Health Science, University of Trieste, 34149 Trieste, Italy; (F.G.D.G.); (E.P.); (L.M.T.); (C.C.); (F.P.); (N.A.); (A.P.); (M.Z.); (N.F.); (G.B.)
| |
Collapse
|
9
|
Boyanovsky BB. Lipoprotein(a) Lowering Medications: Another Step Forward in Atherosclerosis Management. J Cardiovasc Pharmacol 2023; 82:32-36. [PMID: 37192439 DOI: 10.1097/fjc.0000000000001438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
|
10
|
Wang HP, Zhang N, Liu YJ, Xia TL, Chen GC, Yang J, Li FR. Lipoprotein(a), family history of cardiovascular disease, and incidence of heart failure. J Lipid Res 2023; 64:100398. [PMID: 37276941 PMCID: PMC10339055 DOI: 10.1016/j.jlr.2023.100398] [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: 03/28/2023] [Revised: 05/26/2023] [Accepted: 05/27/2023] [Indexed: 06/07/2023] Open
Abstract
Lipoprotein(a) (Lp(a)) is a largely genetically determined biomarker for cardiovascular disease (CVD), while its potential interplay with family history (FHx) of CVD, a measure of both genetic and environmental exposures, remains unclear. We examined the associations of Lp(a) in terms of circulating concentration or polygenetic risk score (PRS), and FHx of CVD with risk of incident heart failure (HF). Included were 299,158 adults from the UK Biobank without known HF and CVD at baseline. Hazards ratios (HRs) and 95% Cls were estimated by Cox regression models adjusted for traditional risk factors defined by the Atherosclerosis Risk in Communities study HF risk score. During the 11.8-year follow-up, 5,502 incidents of HF occurred. Higher levels of circulating Lp(a), Lp(a) PRS, and positive FHx of CVD were associated with higher risks of HF. Compared with individuals who had lower circulating Lp(a) and no FHx, HRs (95% CIs) of HF were 1.36 (1.25, 1.49), 1.31 (1.19, 1.43), and 1.42 (1.22, 1.67) for those with higher Lp(a) and a positive history of CVD for all family members, parents, and siblings, respectively; similar results were observed by using Lp(a) PRS. The risk estimates for HF associated with elevated Lp(a) and positive FHx were attenuated after excluding those with incident myocardial infarction (MI) during follow-up. Lp(a) and FHx of CVD were independent risk factors for incident HF, and the highest risk of HF was observed among individuals with both risk factors. The association may be partly mediated by myocardial infarction.
Collapse
Affiliation(s)
- Hai-Peng Wang
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Na Zhang
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu-Jie Liu
- Department of Nutrition and Food Hygiene, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, China
| | - Tian-Long Xia
- Division of Public Health Emergency, Shenzhen Center for Disease Control and Prevention, Shenzhen, Guangdong, China
| | - Guo-Chong Chen
- Department of Nutrition and Food Hygiene, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, China
| | - Jing Yang
- Department of Nutrition and Food Hygiene, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, China; Department of Clinical Nutrition, The First Affiliated Hospital of Soochow University, Suzhou, China.
| | - Fu-Rong Li
- Shenzhen Key Laboratory of Cardiovascular Health and Precision Medicine, Southern University of Science and Technology, Shenzhen, China; School of Public Health and Emergency Management, Southern University of Science and Technology, Shenzhen, China; Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, China.
| |
Collapse
|
11
|
Amiri M, Raeisi-Dehkordi H, Verkaar AJCF, Wu Y, van Westing AC, Berk KA, Bramer WM, Aune D, Voortman T. Circulating lipoprotein (a) and all-cause and cause-specific mortality: a systematic review and dose-response meta-analysis. Eur J Epidemiol 2023; 38:485-499. [PMID: 36708412 PMCID: PMC10164031 DOI: 10.1007/s10654-022-00956-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 12/16/2022] [Indexed: 01/29/2023]
Abstract
AIMS To investigate the association between circulating lipoprotein(a) (Lp(a)) and risk of all-cause and cause-specific mortality in the general population and in patients with chronic diseases, and to elucidate the dose-response relations. METHODS AND RESULTS We searched literature to find prospective studies reporting adjusted risk estimates on the association of Lp(a) and mortality outcomes. Forty-three publications, reporting on 75 studies (957,253 participants), were included. The hazard ratios (HRs) and 95% confidence intervals (95%CI ) for the top versus bottom tertile of Lp(a) levels and risk of all-cause mortality were 1.09 (95%CI: 1.01-1.18, I2: 75.34%, n = 19) in the general population and 1.18 (95%CI: 1.04-1.34, I2: 52.5%, n = 12) in patients with cardiovascular diseases (CVD). The HRs for CVD mortality were 1.33 (95%CI: 1.11-1.58, I2: 82.8%, n = 31) in the general population, 1.25 (95%CI: 1.10-1.43, I2: 54.3%, n = 17) in patients with CVD and 2.53 (95%CI: 1.13-5.64, I2: 66%, n = 4) in patients with diabetes mellitus. Linear dose-response analyses revealed that each 50 mg/dL increase in Lp(a) levels was associated with 31% and 15% greater risk of CVD death in the general population and in patients with CVD. No non-linear dose-response association was observed between Lp(a) levels and risk of all-cause or CVD mortality in the general population or in patients with CVD (Pnonlinearity > 0.05). CONCLUSION This study provides further evidence that higher Lp(a) levels are associated with higher risk of all-cause mortality and CVD-death in the general population and in patients with CVD. These findings support the ESC/EAS Guidelines that recommend Lp(a) should be measured at least once in each adult person's lifetime, since our study suggests those with higher Lp(a) might also have higher risk of mortality.
Collapse
Affiliation(s)
- Mojgan Amiri
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Hamidreza Raeisi-Dehkordi
- Institute of Social and Preventive Medicine (ISPM), University of Bern, Bern, Switzerland
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Auke J C F Verkaar
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, The Netherlands
| | - Yahong Wu
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Anniek C van Westing
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, The Netherlands
| | - Kirsten A Berk
- Department of Internal Medicine, Division of Pharmacology and Vascular Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Internal Medicine, Division of Dietetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Wichor M Bramer
- Medical Library, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Dagfinn Aune
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- Department of Nutrition, Bjørknes University College, Oslo, Norway
- Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital, Oslo, Norway
| | - Trudy Voortman
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, The Netherlands.
| |
Collapse
|
12
|
Secondary Cardiovascular Prevention after Acute Coronary Syndrome: Emerging Risk Factors and Novel Therapeutic Targets. J Clin Med 2023; 12:jcm12062161. [PMID: 36983163 PMCID: PMC10056379 DOI: 10.3390/jcm12062161] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 03/02/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023] Open
Abstract
The control of cardiovascular risk factors, the promotion of a healthy lifestyle, and antithrombotic therapy are the cornerstones of secondary prevention after acute coronary syndrome (ACS). However, many patients have recurrent ischemic events despite the optimal control of traditional modifiable risk factors and the use of tailored pharmacological therapy, including new-generation antiplatelet and lipid-lowering agents. This evidence emphasizes the importance of identifying novel risk factors and targets to optimize secondary preventive strategies. Lipoprotein(a) (Lp(a)) has emerged as an independent predictor of adverse events after ACS. New molecules such as anti-PCSK9 monoclonal antibodies, small interfering RNAs, and antisense oligonucleotides can reduce plasma Lp(a) levels and are associated with a long-term outcome benefit after the index event. The inflammatory stimulus and the inflammasome, pivotal elements in the development and progression of atherosclerosis, have been widely investigated in patients with coronary artery disease. More recently, randomized clinical trials including post-ACS patients treated with colchicine and monoclonal antibodies targeting cytokines yielded promising results in the reduction in major cardiovascular events after an ACS. Gut dysbiosis has also raised great interest for its potential pathophysiological role in cardiovascular disease. This evidence, albeit preliminary and needing confirmation by larger population-based studies, suggests the possibility of targeting the gut microbiome in particularly high-risk populations. The risk of recurrent ischemic events after ACS is related to the complex interaction between intrinsic predisposing factors and environmental triggers. The identification of novel risk factors and targets is fundamental to customizing patient clinical management with a precision medicine perspective.
Collapse
|
13
|
Gaba P, Gersh BJ, Muller J, Narula J, Stone GW. Evolving concepts of the vulnerable atherosclerotic plaque and the vulnerable patient: implications for patient care and future research. Nat Rev Cardiol 2023; 20:181-196. [PMID: 36151312 DOI: 10.1038/s41569-022-00769-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/12/2022] [Indexed: 11/08/2022]
Abstract
Understanding the natural history of coronary artery atherosclerosis is necessary to determine prognosis and prescribe effective therapies. Traditional management of coronary artery disease has focused on the treatment of flow-limiting anatomical obstructions that lead to ischaemia. In most scenarios, revascularization of these atherosclerotic plaques has not substantially improved freedom from death or myocardial infarction, questioning the utility of contemporary revascularization strategies to improve prognosis. Advances in non-invasive and invasive imaging techniques have helped to identify the characteristics of obstructive and non-obstructive plaques that are precursors for plaque progression and future acute coronary syndromes as well as cardiac death. These 'vulnerable plaques' develop as a consequence of systemic inflammation and are prone to inducing thrombosis. Vulnerable plaques most commonly have a large plaque burden with a well-formed necrotic core and thin fibrous cap and are metabolically active. Perivascular adipose tissue might, in some patients, be used as a surrogate for coronary inflammation and predict future risk of adverse cardiac events. Vulnerable plaques can be identified in their quiescent state, offering the potential for therapeutic passivation. In this Review, we describe the biological and compositional features of vulnerable plaques, the non-invasive and invasive diagnostic modalities to characterize vulnerable plaques, the prognostic utility of identifying vulnerable plaques, and the future studies needed to explore the value of intensified pharmacological and focal treatments of vulnerable plaques.
Collapse
Affiliation(s)
- Prakriti Gaba
- Division of Cardiovascular Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Bernard J Gersh
- Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - James Muller
- Division of Cardiovascular Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Jagat Narula
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Gregg W Stone
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
14
|
Wang Z, Xiao S, Liu N. Association of lipoprotein(a) with coronary severity in patients with new-onset acute myocardial infarction: A large cross-sectional study. Clin Chim Acta 2023; 540:117220. [PMID: 36621598 DOI: 10.1016/j.cca.2023.117220] [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: 11/14/2022] [Revised: 01/03/2023] [Accepted: 01/03/2023] [Indexed: 01/07/2023]
Abstract
BACKGROUND Although current evidence suggests a causal association between lipoprotein(a) [Lp(a)] and cardiovascular disease, there is still no consensus on its association with coronary severity in new-onset acute myocardial infarction (AMI). We explored the association of Lp(a) with coronary severity. METHODS In this large cross-sectional study, we enrolled 2,740 patients with new-onset AMI from the Zhongda Hospital affiliated to Southeast University. Lp(a) was considered as an exposure variable. Gensini score, left main disease and three-vessel disease were used to assess coronary severity. Multivariate logistic regression, restricted cubic spline (RCS) models and threshold effects were used to analyze the association of Lp(a) with coronary severity. RESULTS Multivariate adjusted models showed that Lp(a) was independently associated with Gensini score (≥100), left main disease and three-vessel disease [Q4 vs Q1, OR (95 % CI), P value: 2.301 (1.770, 2.992), P < 0.001; 1.743 (1.174, 2.587), P = 0.006; 1.431 (1.128, 1.816), P = 0.003; respectively], and the associations persisted in sensitivity analyses and most subgroups (P < 0.05). Additionally, the RCS showed that Lp(a) was nonlinearly associated with Gensini score (continuous variable), Gensini score (≥100) and three-vessel disease (P for nonlinearity < 0.05). Threshold effects analysis showed that Lp(a) above the inflection point was positively associated with Gensini score (continuous variable) as well as the risk of Gensini score (≥100) and three-vessel disease. CONCLUSION Lp(a) was closely associated with coronary severity represented by Gensini score, left main disease and three-vessel disease in patients with new-onset AMI.
Collapse
Affiliation(s)
- Zhenwei Wang
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Shengjue Xiao
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Naifeng Liu
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China.
| |
Collapse
|
15
|
Al Hageh C, Chacar S, Ghassibe-Sabbagh M, Platt DE, Henschel A, Hamdan H, Gauguier D, El Murr Y, Alefishat E, Chammas E, O’Sullivan S, Abchee A, Nader M, Zalloua PA. Elevated Lp(a) Levels Correlate with Severe and Multiple Coronary Artery Stenotic Lesions. Vasc Health Risk Manag 2023; 19:31-41. [PMID: 36703868 PMCID: PMC9871050 DOI: 10.2147/vhrm.s394134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 12/21/2022] [Indexed: 01/19/2023] Open
Abstract
Backgrounds and Aims The role of Lipoprotein(a) (Lp(a)) in increasing the risk of cardiovascular diseases is reported in several populations. The aim of this study is to investigate the correlation of high Lp(a) levels with the degree of coronary artery stenosis. Methods Two hundred and sixty-eight patients were enrolled for this study. Patients who underwent coronary artery angiography and who had Lp(a) measurements available were included in this study. Binomial logistic regressions were applied to investigate the association between Lp(a) and stenosis in the four major coronary arteries. The effect of LDL and HDL Cholesterol on modulating the association of Lp(a) with coronary artery disease (CAD) was also evaluated. Multinomial regression analysis was applied to assess the association of Lp(a) with the different degrees of stenosis in the four major coronary arteries. Results Our analyses showed that Lp(a) is a risk factor for CAD and this risk is significantly apparent in patients with HDL-cholesterol ≥35 mg/dL and in non-obese patients. A large proportion of the study patients with elevated Lp(a) levels had CAD even when exhibiting high HDL serum levels. Increased HDL with low Lp(a) serum levels were the least correlated with stenosis. A significantly higher levels of Lp(a) were found in patients with >50% stenosis in at least two major coronary vessels arguing for pronounced and multiple stenotic lesions. Finally, the derived variant (rs1084651) of the LPA gene was significantly associated with CAD. Conclusion Our study highlights the importance of Lp(a) levels as an independent biological marker of severe and multiple coronary artery stenosis.
Collapse
Affiliation(s)
- Cynthia Al Hageh
- Department of Molecular Biology and Genetics, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Stephanie Chacar
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Michella Ghassibe-Sabbagh
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut, Lebanon
| | - Daniel E Platt
- Computational Biology Center, IBM TJ Watson Research Centre, Yorktown Hgts, NY, USA
| | - Andreas Henschel
- Department of Electrical Engineering and Computer, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Hamdan Hamdan
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Dominique Gauguier
- Université Paris Cité, INSERM UMR 1124, Paris, 75006, France,McGill University and Genome Quebec Innovation Centre, Montreal, QC, H3A 0G1, Canada
| | - Yara El Murr
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut, Lebanon
| | - Eman Alefishat
- Department of Pharmacology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates,Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, 127788, United Arab Emirates
| | - Elie Chammas
- School of Medicine, Lebanese University, Beirut, Lebanon
| | - Siobhán O’Sullivan
- Department of Molecular Biology and Genetics, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Antoine Abchee
- Sheikh Shakhbout Medical City, Abu Dhabi, United Arab Emirates
| | - Moni Nader
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates,Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, 127788, United Arab Emirates
| | - Pierre A Zalloua
- Department of Molecular Biology and Genetics, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates,Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, 127788, United Arab Emirates,Harvard T.H. Chan School of Public Health, Boston, MA, USA,Correspondence: Pierre A Zalloua; Moni Nader, College of Medicine and Health Sciences, Khalifa University for Science and Technology, PO Box 127788, Abu Dhabi, United Arab Emirates, Email ;
| |
Collapse
|
16
|
Lampsas S, Oikonomou E, Pantelidis P, Theofilis P, Grammatopoulos K, Marathonitis A, Vavuranakis MA, Siasos G, Tousoulis D, Vavuranakis M. Lipoprotein (a) Levels and Abdominal Aortic Aneurysm. A Systematic Review and Meta-analysis. Curr Pharm Des 2022; 28:3492-3499. [PMID: 36424795 DOI: 10.2174/1381612829666221124110920] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 09/29/2022] [Accepted: 10/14/2022] [Indexed: 11/27/2022]
Abstract
BACKGROUND Several studies have linked high Lipoprotein (a) (Lp(a)) concentrations to cardiovascular events, including the formation of Abdominal Aortic Aneurysms (AAA). We review and meta-analyze existing evidence on the association of Lp(a) levels with AAA. METHODS Studies evaluating the link of Lp(a) with AAA, up to December 27th 2021, were identified by a systematic search of PubMed, SCOPUS, and Web of Science databases. The results were qualitatively and quantitatively synthesized according to PRISMA guidelines. Results are presented as standardized mean differences (SMD) with 95% confidence intervals (CI). RESULTS A total of 5,078 subjects (1,637 patients with AAA vs. 3,441 controls) from 11 studies were included in the meta-analysis, with a mean age of 69.9 years and a male sex prevalence of 85.8%. Based on the qualitative synthesis, high Lp(a) concentrations are linked to abdominal aortic wall degradation and extracellular matrix disarrangement. Moreover, despite the considerable variability among races, high Lp(a) levels are related to increased AAA risk, independently of race differences. Accordingly, patients with AAA displayed significantly higher Lp(a) levels compared to controls (SMD: 0.86, 95% CI: 0.55-1.17, p < 0.001). The outcome was not affected in a sensitivity analysis excluding three outlying studies (SMD: 0.40, 95% CI: 0.22-0.58, p < 0.001). CONCLUSION This meta-analysis indicates the association between high Lp(a) levels and the presence of AAA, although existing literature presents high heterogeneity. Further studies are needed to standardize Lp(a) measurements and to conclude whether Lp(a) can be used as a sensitive biomarker of early presymptomatic AAA diagnosis.
Collapse
Affiliation(s)
- Stamatios Lampsas
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Athens, Greece
| | - Evangelos Oikonomou
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Athens, Greece.,Cardiometabolic Disease Unit, 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Athens, 11527 Greece
| | - Panteleimon Pantelidis
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Athens, Greece
| | - Panagiotis Theofilis
- Cardiometabolic Disease Unit, 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Athens, 11527 Greece
| | - Konstantinos Grammatopoulos
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Athens, Greece
| | - Anastasios Marathonitis
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Athens, Greece
| | - Michael A Vavuranakis
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Athens, Greece
| | - Gerasimos Siasos
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Athens, Greece.,Cardiometabolic Disease Unit, 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Athens, 11527 Greece.,Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Dimitris Tousoulis
- 1st Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Hippokration General Hospital, Athens, Greece
| | - Manolis Vavuranakis
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Athens, Greece
| |
Collapse
|
17
|
Wang ZW, Li M, Li JJ, Liu NF. Association of lipoprotein(a) with all-cause and cause-specific mortality: A prospective cohort study. Eur J Intern Med 2022; 106:63-70. [PMID: 36127218 DOI: 10.1016/j.ejim.2022.09.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 08/27/2022] [Accepted: 09/13/2022] [Indexed: 11/03/2022]
Abstract
BACKGROUND A growing number of studies have demonstrated a causal association between lipoprotein(a) [Lp(a)] and atherosclerotic cardiovascular diseases (ASCVDs), but its association with all-cause and cause-specific mortality remains unclear. Therefore, this study aimed to explore the association of Lp(a) with all-cause and cause-specific mortality. METHODS This prospective cohort study included 8,525 participants from the third National Health and Nutrition Examination Survey. Lp(a) was considered an exposure variable, all-cause and cause-specific mortality were used as outcome variables, and all participants were followed from the interview date until death or December 31, 2015. COX proportional hazards regression models, stratified analysis, sensitivity analysis, restricted cubic spline plots and Kaplan-Meier survival curves were used to analyze the association of Lp(a) with all-cause and cause-specific mortality. RESULTS After adjusting for traditional cardiovascular risk factors, Lp(a) remained strongly associated with all-cause and CVDs-related mortality (P for trend = 0.007 and < 0.001). Subgroup analyses showed that higher Lp(a) remained associated with higher risk of all-cause mortality in those > 60 years of age, with a BMI < 30 kg/m2, and without diabetes, whereas the association between Lp(a) and CVDs-related mortality remained stable in participants ≤ 60 years of age, male, with a BMI < 30 kg/m2, with hypertension, without diabetes, or without CVDs (P < 0.05). In sensitivity analyses, we found that the association of Lp(a) with all-cause and CVDs-related mortality remained robust after excluding individuals who died within one year of follow-up (P for trend = 0.041 and 0.002). CONCLUSIONS Lp(a) was associated with the risk of all-cause and CVDs-related mortality.
Collapse
Affiliation(s)
- Zhen-Wei Wang
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Min Li
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Jing-Jie Li
- Department of Hematology and Oncology, Affiliated Xuchang People's Hospital of Xinxiang Medical College, Xuchang, China
| | - Nai-Feng Liu
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China.
| |
Collapse
|
18
|
Kato A, Kinoshita D, Nagata T, Asakura K, Katamine M, Katsura A, Hashimoto T, Minami Y, Ako J. Lipoprotein (a) levels and vulnerable characteristics in nonculprit plaque in patients with acute coronary syndrome. IJC HEART & VASCULATURE 2022; 43:101120. [PMID: 36118156 PMCID: PMC9474856 DOI: 10.1016/j.ijcha.2022.101120] [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: 06/04/2022] [Revised: 07/31/2022] [Accepted: 09/02/2022] [Indexed: 11/06/2022]
Abstract
High Lp(a) level is associated with more TCFA in nonculprit plaque in ACS patients. The association was significant among patients with plaque erosion. Measurement of Lp(a) may be useful for risk stratification in ACS patients.
Background High plasma levels of Lp(a) are associated with a worse prognosis in patients with coronary artery disease. The aim of the present study is to clarify the association between high lipoprotein a [Lp(a)] levels and vulnerable characteristics of nonculprit plaques in patients with acute coronary syndrome (ACS). Methods A total of 185 consecutive patients with ACS who underwent optical coherence tomography imaging of nonculprit plaques in the culprit vessels were enrolled. Patients were divided into the high Lp(a) group (≥30 mg/dL; 50 nonculprit plaques in 49 patients) or the low Lp(a) group (<30 mg/dL; 139 nonculprit plaques in 136 patients). Results The prevalence of thin-cap fibroatheroma (TCFA) was significantly higher in the high Lp(a) group than in the low Lp(a) group (38.0 vs. 21.6%, p = 0.034). Multivariate logistic analysis demonstrated that a high Lp(a) level was independently associated with the prevalence of TCFA (odds ratio, 1.18; 95% confidence interval, 1.01–1.36; p = 0.033). The prevalence of TCFA was significantly higher in the high Lp(a) group than in the low Lp(a) group among patients with plaque erosion (50.0 vs. 9.4%, respectively; p = 0.027), although the difference was not statistically significant between the two groups in patients with plaque rupture. Conclusions High Lp(a) levels were associated with a high prevalence of TCFA in nonculprit plaques among patients with ACS, particularly in patients with plaque erosion. The present results may partly explain the pathogenesis of worse clinical outcomes in patients with ACS and a high Lp(a) level as shown in clinical studies.
Collapse
|
19
|
Duan Y, Gong K, Xu S, Zhang F, Meng X, Han J. Regulation of cholesterol homeostasis in health and diseases: from mechanisms to targeted therapeutics. Signal Transduct Target Ther 2022; 7:265. [PMID: 35918332 PMCID: PMC9344793 DOI: 10.1038/s41392-022-01125-5] [Citation(s) in RCA: 104] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 07/04/2022] [Accepted: 07/12/2022] [Indexed: 12/13/2022] Open
Abstract
Disturbed cholesterol homeostasis plays critical roles in the development of multiple diseases, such as cardiovascular diseases (CVD), neurodegenerative diseases and cancers, particularly the CVD in which the accumulation of lipids (mainly the cholesteryl esters) within macrophage/foam cells underneath the endothelial layer drives the formation of atherosclerotic lesions eventually. More and more studies have shown that lowering cholesterol level, especially low-density lipoprotein cholesterol level, protects cardiovascular system and prevents cardiovascular events effectively. Maintaining cholesterol homeostasis is determined by cholesterol biosynthesis, uptake, efflux, transport, storage, utilization, and/or excretion. All the processes should be precisely controlled by the multiple regulatory pathways. Based on the regulation of cholesterol homeostasis, many interventions have been developed to lower cholesterol by inhibiting cholesterol biosynthesis and uptake or enhancing cholesterol utilization and excretion. Herein, we summarize the historical review and research events, the current understandings of the molecular pathways playing key roles in regulating cholesterol homeostasis, and the cholesterol-lowering interventions in clinics or in preclinical studies as well as new cholesterol-lowering targets and their clinical advances. More importantly, we review and discuss the benefits of those interventions for the treatment of multiple diseases including atherosclerotic cardiovascular diseases, obesity, diabetes, nonalcoholic fatty liver disease, cancer, neurodegenerative diseases, osteoporosis and virus infection.
Collapse
Affiliation(s)
- Yajun Duan
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Ke Gong
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Suowen Xu
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Feng Zhang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Xianshe Meng
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Jihong Han
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China. .,College of Life Sciences, Key Laboratory of Bioactive Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China.
| |
Collapse
|
20
|
Ugovšek S, Šebeštjen M. Lipoprotein(a)—The Crossroads of Atherosclerosis, Atherothrombosis and Inflammation. Biomolecules 2021; 12:biom12010026. [PMID: 35053174 PMCID: PMC8773759 DOI: 10.3390/biom12010026] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 12/22/2021] [Accepted: 12/23/2021] [Indexed: 12/14/2022] Open
Abstract
Increased lipoprotein(a) (Lp(a)) levels are an independent predictor of coronary artery disease (CAD), degenerative aortic stenosis (DAS), and heart failure independent of CAD and DAS. Lp(a) levels are genetically determinated in an autosomal dominant mode, with great intra- and inter-ethnic diversity. Most variations in Lp(a) levels arise from genetic variations of the gene that encodes the apolipoprotein(a) component of Lp(a), the LPA gene. LPA is located on the long arm of chromosome 6, within region 6q2.6–2.7. Lp(a) levels increase cardiovascular risk through several unrelated mechanisms. Lp(a) quantitatively carries all of the atherogenic risk of low-density lipoprotein cholesterol, although it is even more prone to oxidation and penetration through endothelia to promote the production of foam cells. The thrombogenic properties of Lp(a) result from the homology between apolipoprotein(a) and plasminogen, which compete for the same binding sites on endothelial cells to inhibit fibrinolysis and promote intravascular thrombosis. LPA has up to 70% homology with the human plasminogen gene. Oxidized phospholipids promote differentiation of pro-inflammatory macrophages that secrete pro-inflammatory cytokines (e. g., interleukin (IL)-1β, IL-6, IL-8, tumor necrosis factor-α). The aim of this review is to define which of these mechanisms of Lp(a) is predominant in different groups of patients.
Collapse
Affiliation(s)
- Sabina Ugovšek
- Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia;
| | - Miran Šebeštjen
- Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia;
- Department of Cardiology, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia
- Department of Vascular Diseases, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia
- Correspondence:
| |
Collapse
|
21
|
Liu J, Liu L, Wang B, Chen S, Liu B, Liang J, Huang H, Li Q, Lun Z, Ying M, Chen G, Huang Z, Xu D, Yan X, Zhu T, Tadesse GA, Tan N, Chen J, Liu Y. Coronary Artery Disease: Optimal Lipoprotein(a) for Survival-Lower Is Better? A Large Cohort With 43,647 Patients. Front Cardiovasc Med 2021; 8:670859. [PMID: 34532348 PMCID: PMC8438333 DOI: 10.3389/fcvm.2021.670859] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 06/09/2021] [Indexed: 12/24/2022] Open
Abstract
Background: A high level of lipoprotein(a) can lead to a high risk of cardiovascular events or mortality. However, the association of moderately elevated lipoprotein(a) levels (≥15 mg/dL) with long-term prognosis among patients with coronary artery disease (CAD) is still uncertain. Hence, we aim to systematically analyzed the relevance of baseline plasma lipoprotein(a) levels to long-term mortality in a large cohort of CAD patients. Methods: We obtained data from 43,647 patients who were diagnosed with CAD and had follow-up information from January 2007 to December 2018. The patients were divided into two groups (<15 and ≥15 mg/dL). The primary endpoint was long-term all-cause death. Kaplan–Meier curve analysis and Cox proportional hazards models were used to investigate the association between moderately elevated baseline lipoprotein(a) levels (≥15 mg/dL) and long-term all-cause mortality. Results: During a median follow-up of 5.04 years, 3,941 (18.1%) patients died. We observed a linear association between lipoprotein(a) levels and long-term all-cause mortality. Compared with lipoprotein(a) concentrations <15 mg/dL, lipoprotein(a) ≥15 mg/dL was associated with a significantly higher risk of all-cause mortality [adjusted hazard ratio (aHR) 1.10, 95%CI: 1.04–1.16, P-values = 0.001). Similar results were found for the subgroup analysis of non-acute myocardial infarction, non-percutaneous coronary intervention, chronic heart failure, diabetes mellitus, or non-chronic kidney diseases. Conclusion: Moderately elevated baseline plasma lipoprotein(a) levels (≥15 mg/dL) are significantly associated with higher all-cause mortality in patients with CAD. Our finding provides a rationale for testing the lipoprotein(a)-reducing hypothesis with lower targets (even <15 mg/dL) in CAD outcome trials.
Collapse
Affiliation(s)
- Jin Liu
- Department of Cardiology, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Liwei Liu
- Department of Cardiology, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, IA, United States
| | - Bo Wang
- Department of Cardiology, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Shiqun Chen
- Department of Cardiology, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Buyun Liu
- Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, IA, United States
| | - Jingjing Liang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Haozhang Huang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Qiang Li
- Department of Cardiology, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Zhubin Lun
- Department of Cardiology, Dongguan TCM Hospital, Dongguan, China
| | - Ming Ying
- Department of Cardiology, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Guanzhong Chen
- Guangdong Provincial People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Zhidong Huang
- Department of Cardiology, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Danyuan Xu
- Department of Cardiology, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xiaoming Yan
- Department of Information Technology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Tingting Zhu
- Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Oxford, United Kingdom
| | - Girmaw Abebe Tadesse
- Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Oxford, United Kingdom
| | - Ning Tan
- Department of Cardiology, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Guangdong Provincial People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Jiyan Chen
- Department of Cardiology, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Guangdong Provincial People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yong Liu
- Department of Cardiology, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Guangdong Provincial People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| |
Collapse
|
22
|
Kamstrup PR. Lipoprotein(a) and Cardiovascular Disease. Clin Chem 2021; 67:154-166. [PMID: 33236085 DOI: 10.1093/clinchem/hvaa247] [Citation(s) in RCA: 103] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 09/25/2020] [Indexed: 01/07/2023]
Abstract
BACKGROUND High lipoprotein(a) concentrations present in 10%-20% of the population have long been linked to increased risk of ischemic cardiovascular disease. It is unclear whether high concentrations represent an unmet medical need. Lipoprotein(a) is currently not a target for treatment to prevent cardiovascular disease. CONTENT The present review summarizes evidence of causality for high lipoprotein(a) concentrations gained from large genetic epidemiologic studies and discusses measurements of lipoprotein(a) and future treatment options for high values found in an estimated >1 billion individuals worldwide. SUMMARY Evidence from mechanistic, observational, and genetic studies support a causal role of lipoprotein(a) in the development of cardiovascular disease, including coronary heart disease and peripheral arterial disease, as well as aortic valve stenosis, and likely also ischemic stroke. Effect sizes are most pronounced for myocardial infarction, peripheral arterial disease, and aortic valve stenosis where high lipoprotein(a) concentrations predict 2- to 3-fold increases in risk. Lipoprotein(a) measurements should be performed using well-validated assays with traceability to a recognized calibrator to ensure common cut-offs for high concentrations and risk assessment. Randomized cardiovascular outcome trials are needed to provide final evidence of causality and to assess the potential clinical benefit of novel, potent lipoprotein(a) lowering therapies.
Collapse
Affiliation(s)
- Pia R Kamstrup
- Department of Clinical Biochemistry.,The Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Denmark
| |
Collapse
|
23
|
Sang T, Cheng N, Dang A, Lv N, Zhang W, Li Y, Ji Y, Gu Y. Lipoprotein (a) is associated with poor long-term prognosis in patients aged 80 years and older with acute coronary syndrome. J Clin Lipidol 2021; 15:466-476. [PMID: 34006456 DOI: 10.1016/j.jacl.2021.04.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 03/11/2021] [Accepted: 04/11/2021] [Indexed: 12/20/2022]
Abstract
BACKGROUND Lipoprotein(a) has been suggested as an independent risk factor for cardiovascular events in patients with coronary heart disease (CHD). OBJECTIVE This study aimed to investigate the association of lipoprotein(a) with long-term poor prognosis following acute coronary syndromes (ACS) in advanced-age patients. METHODS We enrolled 536 patients aged ≥80 years hospitalized for ACS and plasma lipoprotein(a) concentrations were measured at admission. The primary outcomes were hard CHD events (a composite of fatal or non-fatal myocardial infarction, and CHD death). The secondary outcomes included major adverse cardiovascular events (MACEs), all-cause death and cardiac death. RESULTS During a median 66-month follow-up, 89 hard CHD events occurred. The optimal cutoff points of lipoprotein(a) levels were obtained from ROC curve analyses. Kaplan-Meier curves showed a significantly higher cumulative incidence of hard CHD events, MACEs, all-cause death and cardiac death in high lipoprotein(a) group than that in low lipoprotein(a) group. Multivariate Cox proportional hazards analyses revealed that elevated lipoprotein(a) levels were independently associated with an increased risk of hard CHD events [hazard ratio (HR): 1.714, 95% confidence interval (95%CI): 1.114-2.638], MACEs (HR 1.354, 95%CI: 1.024-1.790), all-cause death (HR 1.804, 95%CI: 1.286-2.532) and cardiac death (HR 1.891, 95%CI: 1.112-3.217). Furthermore, adding lipoprotein(a) to the prognostic model for hard CHD events improved the C-statistic value (P < 0.05). CONCLUSION Elevated lipoprotein(a) levels were associated with an increased risk of hard CHD events, MACEs, all-cause death and cardiac death in the advanced-age patients with ACS, which indicated that routine screening for lipoprotein(a) might aid prognosis and risk assessment.
Collapse
Affiliation(s)
- Tiantian Sang
- Department of Special Care Center, Fuwai Hospital, National Clinical Research Center for Cardiovascular Diseases, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, No 167 Bei Li Shi Road, Xi Cheng District, Beijing 100037, China.
| | - Nan Cheng
- Department of Special Care Center, Fuwai Hospital, National Clinical Research Center for Cardiovascular Diseases, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, No 167 Bei Li Shi Road, Xi Cheng District, Beijing 100037, China
| | - Aimin Dang
- Department of Special Care Center, Fuwai Hospital, National Clinical Research Center for Cardiovascular Diseases, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, No 167 Bei Li Shi Road, Xi Cheng District, Beijing 100037, China.
| | - Naqiang Lv
- Department of Special Care Center, Fuwai Hospital, National Clinical Research Center for Cardiovascular Diseases, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, No 167 Bei Li Shi Road, Xi Cheng District, Beijing 100037, China
| | - Wei Zhang
- Department of Special Care Center, Fuwai Hospital, National Clinical Research Center for Cardiovascular Diseases, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, No 167 Bei Li Shi Road, Xi Cheng District, Beijing 100037, China
| | - Yifan Li
- Department of Special Care Center, Fuwai Hospital, National Clinical Research Center for Cardiovascular Diseases, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, No 167 Bei Li Shi Road, Xi Cheng District, Beijing 100037, China
| | - Yinze Ji
- Department of Special Care Center, Fuwai Hospital, National Clinical Research Center for Cardiovascular Diseases, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, No 167 Bei Li Shi Road, Xi Cheng District, Beijing 100037, China
| | - Yingzhen Gu
- Department of Special Care Center, Fuwai Hospital, National Clinical Research Center for Cardiovascular Diseases, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, No 167 Bei Li Shi Road, Xi Cheng District, Beijing 100037, China
| |
Collapse
|
24
|
Memtsas VP, Arachchillage DRJ, Gorog DA. Role, Laboratory Assessment and Clinical Relevance of Fibrin, Factor XIII and Endogenous Fibrinolysis in Arterial and Venous Thrombosis. Int J Mol Sci 2021; 22:ijms22031472. [PMID: 33540604 PMCID: PMC7867291 DOI: 10.3390/ijms22031472] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/27/2021] [Accepted: 01/28/2021] [Indexed: 02/06/2023] Open
Abstract
Diseases such as myocardial infarction, ischaemic stroke, peripheral vascular disease and venous thromboembolism are major contributors to morbidity and mortality. Procoagulant, anticoagulant and fibrinolytic pathways are finely regulated in healthy individuals and dysregulated procoagulant, anticoagulant and fibrinolytic pathways lead to arterial and venous thrombosis. In this review article, we discuss the (patho)physiological role and laboratory assessment of fibrin, factor XIII and endogenous fibrinolysis, which are key players in the terminal phase of the coagulation cascade and fibrinolysis. Finally, we present the most up-to-date evidence for their involvement in various disease states and assessment of cardiovascular risk.
Collapse
Affiliation(s)
- Vassilios P. Memtsas
- Cardiology Department, East and North Hertfordshire NHS Trust, Stevenage, Hertfordshire SG1 4AB, UK;
| | - Deepa R. J. Arachchillage
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College London, London SW7 2AZ, UK;
- Department of Haematology, Imperial College Healthcare NHS Trust, London W2 1NY, UK
- Department of Haematology, Royal Brompton Hospital, London SW3 6NP, UK
| | - Diana A. Gorog
- Cardiology Department, East and North Hertfordshire NHS Trust, Stevenage, Hertfordshire SG1 4AB, UK;
- School of Life and Medical Sciences, Postgraduate Medical School, University of Hertfordshire, Hertfordshire AL10 9AB, UK
- Faculty of Medicine, National Heart and Lung Institute, Imperial College, London SW3 6LY, UK
- Correspondence: ; Tel.: +44-207-0348841
| |
Collapse
|
25
|
Soppert J, Lehrke M, Marx N, Jankowski J, Noels H. Lipoproteins and lipids in cardiovascular disease: from mechanistic insights to therapeutic targeting. Adv Drug Deliv Rev 2020; 159:4-33. [PMID: 32730849 DOI: 10.1016/j.addr.2020.07.019] [Citation(s) in RCA: 141] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 07/20/2020] [Accepted: 07/22/2020] [Indexed: 12/12/2022]
Abstract
With cardiovascular disease being the leading cause of morbidity and mortality worldwide, effective and cost-efficient therapies to reduce cardiovascular risk are highly needed. Lipids and lipoprotein particles crucially contribute to atherosclerosis as underlying pathology of cardiovascular disease and influence inflammatory processes as well as function of leukocytes, vascular and cardiac cells, thereby impacting on vessels and heart. Statins form the first-line therapy with the aim to block cholesterol synthesis, but additional lipid-lowering drugs are sometimes needed to achieve low-density lipoprotein (LDL) cholesterol target values. Furthermore, beyond LDL cholesterol, also other lipid mediators contribute to cardiovascular risk. This review comprehensively discusses low- and high-density lipoprotein cholesterol, lipoprotein (a), triglycerides as well as fatty acids and derivatives in the context of cardiovascular disease, providing mechanistic insights into their role in pathological processes impacting on cardiovascular disease. Also, an overview of applied as well as emerging therapeutic strategies to reduce lipid-induced cardiovascular burden is provided.
Collapse
Affiliation(s)
- Josefin Soppert
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital Aachen, Aachen, Germany
| | - Michael Lehrke
- Medical Clinic I, University Hospital Aachen, Aachen, Germany
| | - Nikolaus Marx
- Medical Clinic I, University Hospital Aachen, Aachen, Germany
| | - Joachim Jankowski
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital Aachen, Aachen, Germany; Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, Maastricht University, the Netherlands
| | - Heidi Noels
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital Aachen, Aachen, Germany; Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands.
| |
Collapse
|
26
|
Lipoprotein(a) and long-term recurrent infarction after an episode of ST-segment elevation acute myocardial infarction. Coron Artery Dis 2020; 31:378-384. [PMID: 32040026 DOI: 10.1097/mca.0000000000000852] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND In established ischemic heart disease, the relationship between lipoprotein(a) and new cardiovascular events showed contradictory results. Our aim was to assess the relationship between lipoprotein(a) and very long-term recurrent myocardial infarction (MI) after an index episode of ST-segment elevation acute myocardial infarction (STEMI). METHODS We included 435 consecutive STEMI patients discharged from October 2000 to June 2003 in a single teaching center. The relationship between lipoprotein(a) at discharge and recurrent MI was evaluated through negative binomial regression and Cox regression analysis. RESULTS The mean age was 65 years (55-74 years), 25.5% were women, 34.7% were diabetic, and 66% had a MI of anterior location. Fibrinolysis, rescue, or primary angioplasty was performed in 215 (49.4%), 19 (4.4%), and 18 (4.1%) patients, respectively. The median lipoprotein(a) was 30.4 mg/dL (12-59.4 mg/dL). After a median follow-up of 9.6 years (4.1-15 years), 180 (41.4%) deaths and 187 MI in 133 (30.6%) patients were recorded. After a multivariate adjustment, the risk gradient of lipoprotein(a) showed a neutral effect along most of the continuum and only extreme higher values identified those at higher risk of recurrent MI (P = 0.020). Those with lipoprotein(a) values >95th percentile (≥135 mg/dL) showed a higher risk of recurrent MI (incidence rate ratio, 2.34; 95% confidence interval, 1.37-4.02; P = 0.002). Lipoprotein(a) was not related to the risk of mortality (P = 0.245). CONCLUSIONS After an episode of STEMI, only extreme high values of lipoprotein(a) were associated with an increased risk of long-term recurrent MI.
Collapse
|
27
|
Jawi MM, Frohlich J, Chan SY. Lipoprotein(a) the Insurgent: A New Insight into the Structure, Function, Metabolism, Pathogenicity, and Medications Affecting Lipoprotein(a) Molecule. J Lipids 2020; 2020:3491764. [PMID: 32099678 PMCID: PMC7016456 DOI: 10.1155/2020/3491764] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 08/17/2019] [Indexed: 12/15/2022] Open
Abstract
Lipoprotein(a) [Lp(a)], aka "Lp little a", was discovered in the 1960s in the lab of the Norwegian physician Kåre Berg. Since then, we have greatly improved our knowledge of lipids and cardiovascular disease (CVD). Lp(a) is an enigmatic class of lipoprotein that is exclusively formed in the liver and comprises two main components, a single copy of apolipoprotein (apo) B-100 (apo-B100) tethered to a single copy of a protein denoted as apolipoprotein(a) apo(a). Plasma levels of Lp(a) increase soon after birth to a steady concentration within a few months of life. In adults, Lp(a) levels range widely from <2 to 2500 mg/L. Evidence that elevated Lp(a) levels >300 mg/L contribute to CVD is significant. The improvement of isoform-independent assays, together with the insight from epidemiologic studies, meta-analyses, genome-wide association studies, and Mendelian randomization studies, has established Lp(a) as the single most common independent genetically inherited causal risk factor for CVD. This breakthrough elevated Lp(a) from a biomarker of atherosclerotic risk to a target of therapy. With the emergence of promising second-generation antisense therapy, we hope that we can answer the question of whether Lp(a) is ready for prime-time clinic use. In this review, we present an update on the metabolism, pathophysiology, and current/future medical interventions for high levels of Lp(a).
Collapse
Affiliation(s)
- Motasim M. Jawi
- Healthy Heart Program, St. Paul's Hospital, Vancouver V6Z 1Y6, Canada
- Division of Experimental Medicine, Department of Medicine, Faculty of Medicine, University of British Columbia, Vancouver V5Z 1M9, Canada
- Department of Clinical PhysiologyCorrection: Department of Physiology, University of Jeddah, P.O. Box: 24, Jeddah 21959, Saudi Arabia
| | - Jiri Frohlich
- Healthy Heart Program, St. Paul's Hospital, Vancouver V6Z 1Y6, Canada
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia V6T 2B5, Canada
| | - Sammy Y. Chan
- Healthy Heart Program, St. Paul's Hospital, Vancouver V6Z 1Y6, Canada
- Department of Medicine, Division of Cardiology, University of British Columbia, Vancouver V5Z 1M9, Canada
| |
Collapse
|
28
|
Labudovic D, Kostovska I, Tosheska Trajkovska K, Cekovska S, Brezovska Kavrakova J, Topuzovska S. Lipoprotein(a) – Link between Atherogenesis and Thrombosis. Prague Med Rep 2019; 120:39-51. [DOI: 10.14712/23362936.2019.9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Lipoprotein(a) – Lp(a) – is an independent risk factor for cardiovascular disease (CVD). Indeed, individuals with plasma concentrations of Lp(a) > 200 mg/l carry an increased risk of developing CVD. Circulating levels of Lp(a) are remarkably resistant to common lipid lowering therapies, currently available treatment for reduction of Lp(a) is plasma apheresis, which is costly and labour intensive. The Lp(a) molecule is composed of two parts: LDL/apoB-100 core and glycoprotein, apolipoprotein(a) – Apo(a), both of them can interact with components of the coagulation cascade, inflammatory pathways and blood vessel cells (smooth muscle cells and endothelial cells). Therefore, it is very important to determine the molecular pathways by which Lp(a) affect the vascular system in order to design therapeutics for targeting the Lp(a) cellular effects. This paper summarises the cellular effects and molecular mechanisms by which Lp(a) participate in atherogenesis, thrombogenesis, inflammation and development of cardiovascular diseases.
Collapse
|
29
|
Zhang LL, Kwak H, Yin SJ, Lee BN, Chang YJ, Hahn MJ, Yang JM, Lee JR, Park YD. An OMICS-based study of the role of C3dg in keratinocytes: RNA sequencing, antibody-chip array, and bioinformatics approaches. Int J Biol Macromol 2019; 133:391-411. [DOI: 10.1016/j.ijbiomac.2019.04.039] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 04/06/2019] [Accepted: 04/06/2019] [Indexed: 01/01/2023]
|
30
|
Hiraiwa H, Morimoto R, Okumura T, Arao Y, Oishi H, Kato H, Yamaguchi S, Kuwayama T, Haga T, Yokoi T, Kondo T, Watanabe N, Mitsuda T, Fukaya K, Sawamura A, Tanaka A, Ishii H, Morishima I, Tsuboi H, Murohara T. A survival case of a young adult patient with ST-elevated myocardial infarction with high levels of lipoprotein(a). J Cardiol Cases 2019; 19:207-210. [DOI: 10.1016/j.jccase.2019.02.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 01/16/2019] [Accepted: 02/11/2019] [Indexed: 11/24/2022] Open
|
31
|
Serum Amyloid A, Paraoxonase-1 Activity, and Apolipoprotein Concentrations as Biomarkers of Subclinical Atherosclerosis Risk in Adrenal Incidentaloma Patients. Arch Med Res 2018; 49:182-190. [PMID: 30031631 DOI: 10.1016/j.arcmed.2018.07.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 07/02/2018] [Indexed: 11/21/2022]
Abstract
BACKGROUND Adrenal incidentalomas (AIs), particularly subclinical hypercortisolism (SH), are related to an increased risk of atherosclerosis. The anti-oxidative enzyme paraoxonase-1 (PON1) and the acute phase reactant serum amyloid A (SAA) are transported by highdensity lipoprotein and reciprocally regulated in acute inflammatory response. Our aim was to investigate serum SAA, PON1, and apolipoprotein levels as indicators of subclinical atherosclerosis in patients with nonfunctioning AI (NFAI) and SH. METHODS The study group consisted of 60 controls, 14 SH, and 86 NFAI subjects. Serum amyloid A (SAA), PON1 activity, lipid profiles, apoA and B, lipoprotein A (LpA), hsCRP, and HOMA-IR levels were compared in all groups. RESULTS Serum insulin, triglyceride, SAA, SAA/PON1 ratio, LpA, apoB, hsCRP, and morning cortisol levels were found to be higher while PON1 and apoAI levels were lower in the SH and NFAI groups compared with the controls, and these parameters were found to be more impaired in SH group than NFAI group (p <0.001). HOMA-IR was higher and DHEAS was lower in the SH group than in the other groups. The SAA/PON1 ratio was positively correlated with LpA (r = 0.460; p <0.001), apoB (r = 0.515; p <0.001), insulin (r = 0.275; p = 0.026), triglyceride (r = 0.248; p = 0.002), morning cortisol (r = 0.259; p = 0.045), and UFC (r = 0.274; p <0.001) and negatively correlated with apoAI (r = 0.329; p <0.001), ACTH (r = -0.384; p <0.001), and DHEAS (r = -0.521, p <0.001) levels. The cut-off value of the SAA/PON1 ratio for NFAI was >0.23, and for SH it was >1.33. CONCLUSION The serum SAA/PON1 ratio was high in both the NFAI and SH groups and also exhibited higher levels in SH group. An increased SAA/PON1 ratio and low DHEAS could be attributable to subclinical atherosclerosis risk in SH patients.
Collapse
|
32
|
Abstract
PURPOSE OF REVIEW Genetic dyslipidemias contribute to the prevalence of ischemic heart disease. The field of genetic dyslipidemias and their influence on atherosclerotic heart disease is rapidly developing and accumulating increasing evidence. The purpose of this review is to describe the current state of knowledge in regard to inherited atherogenic dyslipidemias. The disorders of familial hypercholesterolemia (FH) and elevated lipoprotein(a) will be detailed. Genetic technology has made rapid advancements, leading to new discoveries in inherited atherogenic dyslipidemias, which will be explored in this review, as well as a description of possible future developments. Increasing attention has come upon the genetic disorders of familial hypercholesterolemia and elevated lipoprotein(a). RECENT FINDINGS This review includes new knowledge of these disorders including description of these disorders, their method of diagnosis, their prevalence, their genetic underpinnings, and their effect on the development of cardiovascular disease. In addition, it discusses major advances in genetic technology, including the completion of the human genome sequence, next-generation sequencing, and genome-wide association studies. Also discussed are rare variant studies with specific genetic mechanisms involved in inherited dyslipidemias, such as in the proprotein convertase subtilisin/kexin type 9 (PCSK9) enzyme. The field of genetics of dyslipidemia and cardiovascular disease is rapidly growing, which will result in a bright future of novel mechanisms of action and new therapeutics.
Collapse
Affiliation(s)
- Kavita Sharma
- Ohio Health Heart and Vascular Physicians, 765 North Hamilton Road, Suite 120, Gahanna, OH, 43230, USA
| | - Ragavendra R Baliga
- The Ohio State University Wexner Medical Center, Suite 200, 473 West 12th Avenue, Columbus, OH, 43210, USA.
| |
Collapse
|
33
|
Gue YX, Gorog DA. Importance of Endogenous Fibrinolysis in Platelet Thrombus Formation. Int J Mol Sci 2017; 18:E1850. [PMID: 28841147 PMCID: PMC5618499 DOI: 10.3390/ijms18091850] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 08/21/2017] [Accepted: 08/21/2017] [Indexed: 12/13/2022] Open
Abstract
The processes of thrombosis and coagulation are finely regulated by endogenous fibrinolysis maintaining healthy equilibrium. When the balance is altered in favour of platelet activation and/or coagulation, or if endogenous fibrinolysis becomes less efficient, pathological thrombosis can occur. Arterial thrombosis remains a major cause of morbidity and mortality in the world despite advances in medical therapies. The role endogenous fibrinolysis in the pathogenesis of arterial thrombosis has gained increasing attention in recent years as it presents novel ways to prevent and treat existing diseases. In this review article, we discuss the role of endogenous fibrinolysis in platelet thrombus formation, methods of measurement of fibrinolytic activity, its role in predicting cardiovascular diseases and clinical outcomes and future directions.
Collapse
Affiliation(s)
- Ying X Gue
- Department of Cardiology, East and North Hertfordshire NHS Trust, Hertfordshire SG1 4AB, UK.
| | - Diana A Gorog
- Department of Cardiology, East and North Hertfordshire NHS Trust, Hertfordshire SG1 4AB, UK.
- Department of Postgraduate Medicine, University of Hertfordshire, Hertfordshire AL10 9AB, UK.
- National Heart & Lung Institute, Imperial College, London SW3 6LY, UK.
| |
Collapse
|
34
|
Hishikari K, Hikita H, Nakamura S, Nakagama S, Mizusawa M, Yamamoto T, Doi J, Utsugi Y, Sudo Y, Kimura S, Ashikaga T, Takahashi A, Isobe M. Usefulness of Lipoprotein(a) for Predicting Clinical Outcomes After Endovascular Therapy for Aortoiliac Atherosclerotic Lesions. J Endovasc Ther 2017; 24:793-799. [PMID: 28830274 DOI: 10.1177/1526602817728068] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
PURPOSE To evaluate the usefulness of serum lipoprotein(a) as a biomarker of clinical outcomes after endovascular therapy (EVT) for atherosclerotic aortoiliac lesions. METHODS Serum lipoprotein(a) concentrations were measured at admission in 189 consecutive patients (median age 72 years; 160 men) with peripheral artery disease who underwent EVT for aortoiliac occlusive disease. The patients were dichotomized into 2 groups based on serum lipoprotein(a) levels ≤40 mg/dL (LOW; n=135) or >40 mg/dL (HIGH; n=54). After EVT, the incidences of major adverse limb events (MALE) were analyzed. Predictors of MALE were sought with a Cox proportional hazards analysis; results are presented as the hazard ratio (HR) and 95% confidence interval. RESULTS At the median follow-up of 33 months (interquartile range 11, 54), MALE occurred in 44 (23.3%) patients. The MALE-free survival estimate was significantly lower in patients in the HIGH group (55.6% vs 85.2%, p<0.001). Independent predictors of MALE after EVT were hemodialysis (HR 2.23, 95% CI 1.04 to 4.78, p=0.039) and high lipoprotein(a) levels (HR 2.80, 95% CI 1.44 to 5.45, p=0.003). CONCLUSION High lipoprotein(a) levels were associated with a higher incidence of MALE after EVT for patients with aortoiliac lesions.
Collapse
Affiliation(s)
- Keiichi Hishikari
- 1 Cardiovascular Center, Yokosuka Kyosai Hospital, Yokosuka, Japan.,2 Cardiovascular Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hiroyuki Hikita
- 1 Cardiovascular Center, Yokosuka Kyosai Hospital, Yokosuka, Japan
| | - Shun Nakamura
- 1 Cardiovascular Center, Yokosuka Kyosai Hospital, Yokosuka, Japan
| | - Shun Nakagama
- 1 Cardiovascular Center, Yokosuka Kyosai Hospital, Yokosuka, Japan
| | | | - Tasuku Yamamoto
- 1 Cardiovascular Center, Yokosuka Kyosai Hospital, Yokosuka, Japan
| | - Junichi Doi
- 1 Cardiovascular Center, Yokosuka Kyosai Hospital, Yokosuka, Japan
| | - Yuya Utsugi
- 1 Cardiovascular Center, Yokosuka Kyosai Hospital, Yokosuka, Japan
| | - Yuta Sudo
- 1 Cardiovascular Center, Yokosuka Kyosai Hospital, Yokosuka, Japan
| | - Shigeki Kimura
- 1 Cardiovascular Center, Yokosuka Kyosai Hospital, Yokosuka, Japan
| | - Takashi Ashikaga
- 2 Cardiovascular Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | | | - Mitsuaki Isobe
- 2 Cardiovascular Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
35
|
Kamstrup PR, Hung MY, Witztum JL, Tsimikas S, Nordestgaard BG. Oxidized Phospholipids and Risk of Calcific Aortic Valve Disease: The Copenhagen General Population Study. Arterioscler Thromb Vasc Biol 2017; 37:1570-1578. [PMID: 28572160 DOI: 10.1161/atvbaha.116.308761] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 05/17/2017] [Indexed: 12/26/2022]
Abstract
OBJECTIVE Lipoprotein(a) is causally associated with calcific aortic valve disease (CAVD). Lipoprotein(a) carries proinflammatory and procalcific oxidized phospholipids (OxPL). We tested whether the CAVD risk is mediated by the content of OxPL on lipoprotein(a). APPROACH AND RESULTS A case-control study was performed within the Copenhagen General Population Study (n=87 980), including 725 CAVD cases (1977-2013) and 1413 controls free of cardiovascular disease. OxPL carried by apoB (apolipoprotein B-100; OxPL-apoB) or apolipoprotein(a) (OxPL-apo(a)) containing lipoproteins, lipoprotein(a) levels, LPA kringle IV type 2 repeat, and rs10455872 genetic variants were measured. OxPL-apoB and OxPL-apo(a) levels correlated with lipoprotein(a) levels among cases (r=0.75 and r=0.95; both P<0.001) and controls (r=0.65 and r=0.93; both P<0.001). OxPL-apoB levels associated with risk of CAVD with odds ratios of 1.2 (95% confidence interval [CI]:1.0-1.6) for 34th to 66th percentile levels, 1.6 (95% CI, 1.2-2.1) for 67th to 90th percentile levels, 2.0 (95% CI, 1.3-3.0) for 91st to 95th percentile levels, and 3.4 (95% CI, 2.1-5.5) for levels >95th percentile, versus levels <34th percentile (trend, P<0.001). Corresponding odds ratios for OxPL-apo(a) were 1.2 (95% CI, 1.0-1.5), 1.2(95% CI, 0.9-1.6), 2.1(95% CI, 1.4-3.1), and 2.9(95% CI, 1.9-4.5; trend, P<0.001) and were similar for lipoprotein(a). LPA genotypes associated with OxPL-apoB, OxPL-apo(a), and lipoprotein(a) levels and explained 34%, 46%, and 39%, respectively, of the total variation in levels. LPA genotypes associated with risk of CAVD; a doubling in genetically determined OxPL-apoB, OxPL-apo(a), and lipoprotein(a) levels associated with odds ratio of CAVD of 1.18 (95% CI, 1.10-1.27), 1.09 (95% CI, 1.05-1.13), and 1.09 (95% CI, 1.05-1.14), respectively, comparable to the corresponding observational estimates of 1.27 (95% CI, 1.16-1.39), 1.13 (95% CI, 1.08-1.18), and 1.11 (95% CI, 1.06-1.17). CONCLUSIONS OxPL-apoB and OxPL-apo(a) are novel genetic and potentially causal risk factors for CAVD and may explain the association of lipoprotein(a) with CAVD.
Collapse
Affiliation(s)
- Pia R Kamstrup
- From the Department of Clinical Biochemistry (P.R.K., B.G.N.) and the Copenhagen General Population Study (P.R.K., B.G.N.), Herlev and Gentofte Hospital, Copenhagen University Hospital, Denmark; Department of Medicine, University of California San Diego, La Jolla (M.-Y.H., J.L.W., S.T.); Department of Internal Medicine, School of Medicine, College of Medicine (M.-Y.H.) and Division of Cardiology, Department of Internal Medicine, Shuang Ho Hospital (M.-Y.H.), Taipei Medical University, Taiwan; Graduate Institute of Clinical Medical Sciences, Chang Gung University College of Medicine, Taoyuan, Taiwan (M.-Y.H.); and Faculty of Health and Medical Sciences, University of Copenhagen, Denmark (B.G.N.).
| | - Ming-Yow Hung
- From the Department of Clinical Biochemistry (P.R.K., B.G.N.) and the Copenhagen General Population Study (P.R.K., B.G.N.), Herlev and Gentofte Hospital, Copenhagen University Hospital, Denmark; Department of Medicine, University of California San Diego, La Jolla (M.-Y.H., J.L.W., S.T.); Department of Internal Medicine, School of Medicine, College of Medicine (M.-Y.H.) and Division of Cardiology, Department of Internal Medicine, Shuang Ho Hospital (M.-Y.H.), Taipei Medical University, Taiwan; Graduate Institute of Clinical Medical Sciences, Chang Gung University College of Medicine, Taoyuan, Taiwan (M.-Y.H.); and Faculty of Health and Medical Sciences, University of Copenhagen, Denmark (B.G.N.)
| | - Joseph L Witztum
- From the Department of Clinical Biochemistry (P.R.K., B.G.N.) and the Copenhagen General Population Study (P.R.K., B.G.N.), Herlev and Gentofte Hospital, Copenhagen University Hospital, Denmark; Department of Medicine, University of California San Diego, La Jolla (M.-Y.H., J.L.W., S.T.); Department of Internal Medicine, School of Medicine, College of Medicine (M.-Y.H.) and Division of Cardiology, Department of Internal Medicine, Shuang Ho Hospital (M.-Y.H.), Taipei Medical University, Taiwan; Graduate Institute of Clinical Medical Sciences, Chang Gung University College of Medicine, Taoyuan, Taiwan (M.-Y.H.); and Faculty of Health and Medical Sciences, University of Copenhagen, Denmark (B.G.N.)
| | - Sotirios Tsimikas
- From the Department of Clinical Biochemistry (P.R.K., B.G.N.) and the Copenhagen General Population Study (P.R.K., B.G.N.), Herlev and Gentofte Hospital, Copenhagen University Hospital, Denmark; Department of Medicine, University of California San Diego, La Jolla (M.-Y.H., J.L.W., S.T.); Department of Internal Medicine, School of Medicine, College of Medicine (M.-Y.H.) and Division of Cardiology, Department of Internal Medicine, Shuang Ho Hospital (M.-Y.H.), Taipei Medical University, Taiwan; Graduate Institute of Clinical Medical Sciences, Chang Gung University College of Medicine, Taoyuan, Taiwan (M.-Y.H.); and Faculty of Health and Medical Sciences, University of Copenhagen, Denmark (B.G.N.).
| | - Børge G Nordestgaard
- From the Department of Clinical Biochemistry (P.R.K., B.G.N.) and the Copenhagen General Population Study (P.R.K., B.G.N.), Herlev and Gentofte Hospital, Copenhagen University Hospital, Denmark; Department of Medicine, University of California San Diego, La Jolla (M.-Y.H., J.L.W., S.T.); Department of Internal Medicine, School of Medicine, College of Medicine (M.-Y.H.) and Division of Cardiology, Department of Internal Medicine, Shuang Ho Hospital (M.-Y.H.), Taipei Medical University, Taiwan; Graduate Institute of Clinical Medical Sciences, Chang Gung University College of Medicine, Taoyuan, Taiwan (M.-Y.H.); and Faculty of Health and Medical Sciences, University of Copenhagen, Denmark (B.G.N.)
| |
Collapse
|
36
|
Liu C, Xu MX, He YM, Zhao X, Du XJ, Yang XJ. Lipoprotein (a) is not significantly associated with type 2 diabetes mellitus: cross-sectional study of 1604 cases and 7983 controls. Acta Diabetol 2017; 54:443-453. [PMID: 28132096 DOI: 10.1007/s00592-017-0965-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2016] [Accepted: 01/11/2017] [Indexed: 12/19/2022]
Abstract
AIMS Lipoprotein (a) (Lp(a)), a well-established risk factor for coronary artery diseases (CAD), would also be anticipated to be associated in a similar manner with risk of type 2 diabetes mellitus (T2DM) based on the common soil hypothesis of etiology of T2DM and CAD. Unfortunately, there remains considerable uncertainty regarding the association of Lp(a) with the risk of T2DM. We aimed to examine the association of Lp(a) with T2DM. METHODS Cross-sectional study of 1604 cases and 7983 controls was performed for identifying the association of Lp(a) with T2DM, its possible interactions with risk factors and threshold effects on T2DM. The association of Lp(a) with CAD was also examined and compared within the same study. RESULTS On a continuous scale, 10 mg/L higher Lp(a) levels were insignificantly associated with a fully adjusted OR of 1.011, 95% CI 0.961-1.063 for T2DM. On a categorical scale, the fully adjusted ORs for T2DM were 0.733 (0.526-1.022), 0.554 (0.387-0.793), 0.848 (0.612-1.176), 0.727 (0.515-1.026), 0.692 (0.488-0.981), 0.696 (0.492-0.985), 0.719 (0.509-1.016), 0.74 (0.523-1.045), 0.809 (0.571-1.146), and 0.99 (0.962-1.019) for decile 2-10 in reference to decile 1. The magnitude of association did not increase with increasing decile (P for trend test = 0.990). In contrast, higher Lp(a) levels were significantly associated with prevalent CAD on a continuous or categorical scale in a fully adjusted model. No threshold effects were observed in terms of association of Lp(a) with T2DM or with CAD in Lp(a) <50 mg/dL. CONCLUSIONS The current study suggested that there was a lack of association of Lp(a) levels with prevalent T2DM. In contrast, Lp(a) levels were significantly associated with CAD in a dose-responding manner. Our findings provided evidence for differential approaches to higher Lp(a) levels in patients with T2DM or with CAD.
Collapse
Affiliation(s)
- Chang Liu
- Division of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Ming-Xing Xu
- Division of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Yong-Ming He
- Division of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China.
| | - Xin Zhao
- Division of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Xiao-Jiao Du
- Division of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Xiang-Jun Yang
- Division of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| |
Collapse
|
37
|
Saad AAA, Mohsen MA, Kandil SM, Hassan EM, Megahed MAG, Abdel Rahman MM. Predictive values of some atherogenic risk factors in young workers occupationally exposed to vinyl chloride and heavy metals. ARAB J CHEM 2017. [DOI: 10.1016/j.arabjc.2014.01.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
38
|
Relation of Plasma Lipoprotein(a) to Subclinical Coronary Plaque Volumes, Three-Vessel and Left Main Coronary Disease, and Severe Coronary Stenoses in Apparently Healthy African-Americans With a Family History of Early-Onset Coronary Artery Disease. Am J Cardiol 2016; 118:656-61. [PMID: 27530333 DOI: 10.1016/j.amjcard.2016.06.020] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 06/03/2016] [Accepted: 06/03/2016] [Indexed: 11/24/2022]
Abstract
Serum lipoprotein(a) [Lp(a)] is a coronary artery disease (CAD) risk factor in persons of European ancestry. Levels are twofold to threefold higher in African-Americans (AAs), but reported associations with CAD have been inconsistent. The relation of Lp(a) with the extent and severity of subclinical coronary plaque has not been described in AAs. We screened 269 apparently healthy AAs for risk factors and coronary plaque using advanced coronary computed tomographic angiography. Total coronary plaque (TCP), noncalcified coronary plaque, and calcified coronary plaque volumes (mm(3)) were quantified using a validated automated method. Lp(a) was measured by ELISA. Multivariable modeling was performed with adjustment for traditional CAD risk factors and intrafamilial correlations. Mean age was 51 ± 11 years and 64% were female. Plaque was present in 41%. Lp(a) was independently associated with TCP volume [log(TCP + 1)] (p = 0.04), 3-vessel and/or left main involvement (p = 0.04), and at least 1 stenosis >50% (p = 0.006). Best-fit regression analyses showed that subjects with Lp(a) >40 mg/dl were threefold more likely to have 3-vessel and/or left main disease (95% confidence interval 1.4 to 6.8, p = 0.005) and fourfold more likely to have stenosis >50% (95% confidence interval 1.3 to 15.0, p = 0.02). In subjects with plaque (n = 110), multivariable models showed the Lp(a) level was significantly and independently associated with TCP (p = 0.009), noncalcified coronary plaque (p = 0.01), and calcified coronary plaque (p = 0.003) and affected vessel length (p = 0.01). In conclusion, high Lp(a) is strongly associated with coronary plaque volumes, extent, and severity in apparently healthy AAs. High levels of Lp(a) may be particularly important in the pathogenesis of CAD in AAs.
Collapse
|
39
|
Kunutsor SK, Khan H, Nyyssönen K, Laukkanen JA. Lipoprotein(a) and risk of sudden cardiac death in middle-aged Finnish men: A new prospective cohort study. Int J Cardiol 2016; 220:718-25. [PMID: 27393854 DOI: 10.1016/j.ijcard.2016.06.069] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Accepted: 06/21/2016] [Indexed: 10/21/2022]
Abstract
BACKGROUND Lipoprotein(a) [Lp(a)] is an established and independent risk factor for cardiovascular outcomes. However, the relationship of Lp(a) with risk of sudden cardiac death (SCD) is unknown. We aimed to assess the association of Lp(a) with risk of SCD in the Kuopio Ischemic Heart Disease prospective cohort study of 1881 men aged 42-61years at recruitment. METHODS AND RESULTS Plasma Lp(a) concentration was assessed at baseline and repeat measurements made several years apart. After a median follow-up of 24.7years, 141 SCDs were recorded. Hazard ratios (HRs) (95% confidence intervals [CI]) were assessed and were corrected for within-person variability in Lp(a) levels. The regression dilution ratio of loge Lp(a) adjusted for age was 0.84 (95% CI: 0.81-0.88). Lipoprotein(a) levels were log-linearly associated with risk of SCD. In analyses adjusted for established risk factors, the HR (95% CI) for SCD per 1 standard deviation (3.56-fold) higher baseline loge Lp(a) was 1.24 (1.05-1.47; P=0.013). This remained consistent on further adjustment for alcohol consumption, resting heart rate, lipids, and C-reactive protein 1.23 (1.04-1.46; P=0.018). HRs remained unchanged after accounting for incident coronary events and did not vary importantly in several relevant clinical subgroups. Adding Lp(a) to a SCD risk prediction model did not significantly improve risk discrimination beyond established risk factors, but improved the continuous net reclassification 30.2% (1.1 to 59.2%, P=0.042). CONCLUSIONS Available evidence shows a continuous and independent association between Lp(a) levels and risk of SCD. Further research is needed to replicate these findings.
Collapse
Affiliation(s)
- Setor K Kunutsor
- School of Clinical Sciences, University of Bristol, Learning & Research Building (Level 1), Southmead Hospital, Southmead Road, Bristol, UK.
| | - Hassan Khan
- Emory University School of Medicine, Atlanta, GA, USA
| | - Kristiina Nyyssönen
- Eastern Finland Laboratory Center, and Department of Clinical Chemistry, University of Eastern Finland, Kuopio, Finland; Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Jari A Laukkanen
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland; Central Finland Central Hospital, Jyväskylä, Finland
| |
Collapse
|
40
|
Mitsuda T, Uemura Y, Ishii H, Takemoto K, Uchikawa T, Koyasu M, Ishikawa S, Miura A, Imai R, Iwamiya S, Ozaki Y, Kato T, Shibata R, Watarai M, Murohara T. Lipoprotein(a) levels predict adverse vascular events after acute myocardial infarction. Heart Vessels 2016; 31:1923-1929. [DOI: 10.1007/s00380-016-0823-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 02/19/2016] [Indexed: 12/19/2022]
|
41
|
Kamstrup PR, Nordestgaard BG. Elevated Lipoprotein(a) Levels, LPA Risk Genotypes, and Increased Risk of Heart Failure in the General Population. JACC-HEART FAILURE 2015; 4:78-87. [PMID: 26656145 DOI: 10.1016/j.jchf.2015.08.006] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 08/13/2015] [Accepted: 08/23/2015] [Indexed: 10/22/2022]
Abstract
OBJECTIVES This study sough to test whether elevated lipoprotein(a) levels and corresponding LPA risk genotypes (low number of kringle IV type 2 repeats, rs3798220 and rs10455872, minor allele carriers) are associated with an increased risk of heart failure (HF). BACKGROUND Elevated lipoprotein(a) levels represent a genetically determined risk factor for myocardial infarction (MI) and aortic valve stenosis (AVS). It is presently unknown whether elevated lipoprotein(a) levels also cause heart failure (HF). METHODS We combined 2 general population studies, the Copenhagen City Heart Study (n = 10,855) and the Copenhagen General Population Study (n = 87,242), which totaled 98,097 Danish participants, of whom 4,122 were diagnosed with HF (1976 to 2013). We conducted observational and genetic instrumental variable analyses in a Mendelian randomization study design, assessing evidence of causality, and we performed mediation analyses. RESULTS Elevated lipoprotein(a) levels were associated with multivariable adjusted hazard ratios for HF of 1.10 (95% CI: 0.97 to 1.25) for the 34th to 66th percentiles (8 to 19 mg/dl), 1.24 (95% CI: 1.08 to 1.42) for the 67th to 90th percentiles (20 to 67 mg/dl), 1.57 (95% CI: 1.32 to 1.87) for the 91st to 99th percentiles (68 to 153 mg/dl), and 1.79 (95% CI: 1.18 to 2.73) for levels >99th percentile (>153 mg/dl) versus levels <34th percentile (<8 mg/dl) (trend, p < 0.001), corresponding to a population-attributable risk of 9%. By combining all LPA risk genotypes, instrumental variable analysis yielded a genetic relative risk for HF of 1.18 (95% CI: 1.04 to 1.34) per 10-fold higher lipoprotein(a) levels, which was comparable to the corresponding observational hazard ratio of 1.22 (95% CI: 1.11 to 1.35). Upon exclusion of participants diagnosed with MI or AVS, risk estimates were attenuated. Accordingly, 63% (95% CI: 45% to 99%) of HF risk was mediated via MI and AVS combined. CONCLUSIONS Elevated lipoprotein(a) levels and corresponding LPA risk genotypes were associated with an increased risk of HF consistent with a causal association. The association appeared to be partly mediated by MI and AVS.
Collapse
Affiliation(s)
- Pia R Kamstrup
- Department of Clinical Biochemistry, Herlev Hospital, Copenhagen University Hospital, Copenhagen, Denmark; The Copenhagen General Population Study, Herlev Hospital, Copenhagen University Hospital, Copenhagen, Denmark; Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Børge G Nordestgaard
- Department of Clinical Biochemistry, Herlev Hospital, Copenhagen University Hospital, Copenhagen, Denmark; The Copenhagen General Population Study, Herlev Hospital, Copenhagen University Hospital, Copenhagen, Denmark; Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; The Copenhagen City Heart Study, Frederiksberg Hospital, Copenhagen University Hospital, Copenhagen, Denmark
| |
Collapse
|
42
|
Hoerl G, Ledinski G, Kager G, Thalhammer M, Koestenberger M, Juergens G, Gary T, Cvirn G. Virtually same oxidizability of LDL but higher Lp(a) levels in arterial compared to venous plasma. Chem Phys Lipids 2014; 184:38-41. [PMID: 25240239 DOI: 10.1016/j.chemphyslip.2014.09.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Revised: 09/02/2014] [Accepted: 09/15/2014] [Indexed: 10/24/2022]
Abstract
Plaque formation is confined to the arterial trunk. We assumed that due to the higher aeration of arterial compared to venous blood, higher levels of the atherogenic agent oxidized LDL might be present in arteries, contributing to plaque formation. We aimed to compare (i) the basal oxidative status of LDL in arterial and venous blood and (ii) the susceptibility of arterial and venous LDL to oxidation. The basal oxidative status of LDL was determined by measuring lipid hydroperoxide (LPO) concentrations, plasma levels of auto-antibodies against oxidized LDL, and by measuring oxidation-specific epitopes on LDL particles. The oxidizability of arterial vs. venous LDL (catalyzed by copper) was estimated by monitoring the time-course of conjugated dienes formation. Interestingly, we found the same basal oxidative status of LDL in arterial and venous plasma. LPO concentrations and levels of auto-antibodies against oxidized LDL were similar in arterial and venous plasma and amounts of oxidation-specific epitopes were similar on the respective LDL particles. Moreover, we found similar susceptibilities of arterial and venous LDL to (copper-mediated) oxidation. Lag-times until the onset of conjugated diene formation were slightly shorter in arterial compared to venous LDL in the presence of 5 μM, but not in the presence of 1 μM CuCl2. Additionally, we found significantly higher levels of the atherogenic lipoprotein(a) in arterial plasma. We conclude that not higher oxidizability of arterial LDL but higher arterial lipoprotein(a) levels might help to explain why sclerosis is confined to the arterial trunk.
Collapse
Affiliation(s)
- Gerd Hoerl
- Institute of Physiological Chemistry, Medical University of Graz, Harrachgasse 21/II, Graz A-8010, Austria
| | - Gerhard Ledinski
- Institute of Physiological Chemistry, Medical University of Graz, Harrachgasse 21/II, Graz A-8010, Austria
| | - Gerd Kager
- Institute of Physiological Chemistry, Medical University of Graz, Harrachgasse 21/II, Graz A-8010, Austria
| | | | | | - Guenther Juergens
- Institute of Physiological Chemistry, Medical University of Graz, Harrachgasse 21/II, Graz A-8010, Austria
| | - Thomas Gary
- Department of Angiology, Medical University of Graz, Graz, Austria
| | - Gerhard Cvirn
- Institute of Physiological Chemistry, Medical University of Graz, Harrachgasse 21/II, Graz A-8010, Austria.
| |
Collapse
|
43
|
Lipoprotein(a): Fasting and nonfasting levels, inflammation, and cardiovascular risk. Atherosclerosis 2014; 234:95-101. [DOI: 10.1016/j.atherosclerosis.2014.01.049] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 12/30/2013] [Accepted: 01/22/2014] [Indexed: 01/20/2023]
|
44
|
Nozue T, Yamamoto S, Tohyama S, Fukui K, Umezawa S, Onishi Y, Kunishima T, Sato A, Nozato T, Miyake S, Takeyama Y, Morino Y, Yamauchi T, Muramatsu T, Hibi K, Terashima M, Michishita I. Lipoprotein(a) is associated with necrotic core progression of non-culprit coronary lesions in statin-treated patients with angina pectoris. Lipids Health Dis 2014; 13:59. [PMID: 24684829 PMCID: PMC4230650 DOI: 10.1186/1476-511x-13-59] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 03/18/2014] [Indexed: 01/23/2023] Open
Abstract
Background Statin therapy results in regression and stabilization of coronary artery plaques, and reduces the incidence of coronary artery disease. However, statin therapy does not effectively halt the accumulation of necrotic core in all patients. The purpose of the present study was to identify the predictors associated with necrotic core progression during statin therapy. Methods Coronary atherosclerosis in non-culprit lesions was evaluated using virtual histology intravascular ultrasound at baseline and 8 months after statin therapy. One hundred nineteen patients were divided into 2 groups based on necrotic core progression or regression during an 8-month follow-up period. Results Patients with necrotic core progression had higher serum lipoprotein(a) [Lp(a)] levels than patients with regression at baseline (16 mg/dL vs. 12 mg/dL, p = 0.02) and at the 8-month follow-up (17 mg/dL vs. 10 mg/dL, p = 0.006). Patients with necrotic core progression had a higher fibro-fatty plaque volume (1.28 mm3/mm vs. 0.73 mm3/mm, p = 0.002), and less necrotic core (0.56 mm3/mm vs. 1.04 mm3/mm, p < 0.0001) and dense calcium (0.35 mm3/mm vs. 0.56 mm3/mm, p = 0.006) plaque volumes at baseline than patients with regression. Multivariate logistic regression analysis showed that Lp(a) was a significant independent predictor associated with necrotic core progression during statin therapy (odds ratio [OR]: 3.514; 95% confidence interval [CI]: 1.338-9.228; p = 0.01). Conclusions Serum Lp(a) is independently associated with necrotic core progression in statin-treated patients with angina pectoris.
Collapse
Affiliation(s)
- Tsuyoshi Nozue
- Division of Cardiology, Department of Internal Medicine, Yokohama Sakae Kyosai Hospital, 132 Katsura-cho, Sakae-ku, Yokohama 247-8581, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Nestel PJ, Barnes EH, Tonkin AM, Simes J, Fournier M, White HD, Colquhoun DM, Blankenberg S, Sullivan DR. Plasma Lipoprotein(a) Concentration Predicts Future Coronary and Cardiovascular Events in Patients With Stable Coronary Heart Disease. Arterioscler Thromb Vasc Biol 2013; 33:2902-8. [DOI: 10.1161/atvbaha.113.302479] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
- Paul J. Nestel
- From the Lipoprotein and Atherosclerosis Laboratory, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (P.J.N.); NHMRC Clinical Trials Centre, University of Sydney, Sydney, Australia (E.H.B., J.S., M.F.); Department of Epidemiology & Preventative Medicine, Monash University, Melbourne, Australia (A.M.T.); Green Lane Cardiovascular Research Unit, Auckland City Hospital, Auckland, New Zealand (H.D.W.); Department of Medicine, University of Queensland, Brisbane, Australia (D.M.C.)
| | - Elizabeth H. Barnes
- From the Lipoprotein and Atherosclerosis Laboratory, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (P.J.N.); NHMRC Clinical Trials Centre, University of Sydney, Sydney, Australia (E.H.B., J.S., M.F.); Department of Epidemiology & Preventative Medicine, Monash University, Melbourne, Australia (A.M.T.); Green Lane Cardiovascular Research Unit, Auckland City Hospital, Auckland, New Zealand (H.D.W.); Department of Medicine, University of Queensland, Brisbane, Australia (D.M.C.)
| | - Andrew M. Tonkin
- From the Lipoprotein and Atherosclerosis Laboratory, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (P.J.N.); NHMRC Clinical Trials Centre, University of Sydney, Sydney, Australia (E.H.B., J.S., M.F.); Department of Epidemiology & Preventative Medicine, Monash University, Melbourne, Australia (A.M.T.); Green Lane Cardiovascular Research Unit, Auckland City Hospital, Auckland, New Zealand (H.D.W.); Department of Medicine, University of Queensland, Brisbane, Australia (D.M.C.)
| | - John Simes
- From the Lipoprotein and Atherosclerosis Laboratory, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (P.J.N.); NHMRC Clinical Trials Centre, University of Sydney, Sydney, Australia (E.H.B., J.S., M.F.); Department of Epidemiology & Preventative Medicine, Monash University, Melbourne, Australia (A.M.T.); Green Lane Cardiovascular Research Unit, Auckland City Hospital, Auckland, New Zealand (H.D.W.); Department of Medicine, University of Queensland, Brisbane, Australia (D.M.C.)
| | - Marion Fournier
- From the Lipoprotein and Atherosclerosis Laboratory, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (P.J.N.); NHMRC Clinical Trials Centre, University of Sydney, Sydney, Australia (E.H.B., J.S., M.F.); Department of Epidemiology & Preventative Medicine, Monash University, Melbourne, Australia (A.M.T.); Green Lane Cardiovascular Research Unit, Auckland City Hospital, Auckland, New Zealand (H.D.W.); Department of Medicine, University of Queensland, Brisbane, Australia (D.M.C.)
| | - Harvey D. White
- From the Lipoprotein and Atherosclerosis Laboratory, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (P.J.N.); NHMRC Clinical Trials Centre, University of Sydney, Sydney, Australia (E.H.B., J.S., M.F.); Department of Epidemiology & Preventative Medicine, Monash University, Melbourne, Australia (A.M.T.); Green Lane Cardiovascular Research Unit, Auckland City Hospital, Auckland, New Zealand (H.D.W.); Department of Medicine, University of Queensland, Brisbane, Australia (D.M.C.)
| | - David M. Colquhoun
- From the Lipoprotein and Atherosclerosis Laboratory, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (P.J.N.); NHMRC Clinical Trials Centre, University of Sydney, Sydney, Australia (E.H.B., J.S., M.F.); Department of Epidemiology & Preventative Medicine, Monash University, Melbourne, Australia (A.M.T.); Green Lane Cardiovascular Research Unit, Auckland City Hospital, Auckland, New Zealand (H.D.W.); Department of Medicine, University of Queensland, Brisbane, Australia (D.M.C.)
| | - Stefan Blankenberg
- From the Lipoprotein and Atherosclerosis Laboratory, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (P.J.N.); NHMRC Clinical Trials Centre, University of Sydney, Sydney, Australia (E.H.B., J.S., M.F.); Department of Epidemiology & Preventative Medicine, Monash University, Melbourne, Australia (A.M.T.); Green Lane Cardiovascular Research Unit, Auckland City Hospital, Auckland, New Zealand (H.D.W.); Department of Medicine, University of Queensland, Brisbane, Australia (D.M.C.)
| | - David R. Sullivan
- From the Lipoprotein and Atherosclerosis Laboratory, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (P.J.N.); NHMRC Clinical Trials Centre, University of Sydney, Sydney, Australia (E.H.B., J.S., M.F.); Department of Epidemiology & Preventative Medicine, Monash University, Melbourne, Australia (A.M.T.); Green Lane Cardiovascular Research Unit, Auckland City Hospital, Auckland, New Zealand (H.D.W.); Department of Medicine, University of Queensland, Brisbane, Australia (D.M.C.)
| |
Collapse
|
46
|
Alihanoglu YI, Kilic ID, Yildiz BS. Non-Atherosclerotic Causes of Acute Coronary Syndrome
and Management of The Patients. ELECTRONIC JOURNAL OF GENERAL MEDICINE 2013. [DOI: 10.29333/ejgm/82300] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
47
|
Whayne TF. Coronary atherosclerosis, low-density lipoproteins and markers of thrombosis, inflammation and endothelial dysfunction. Int J Angiol 2012; 16:12-6. [PMID: 22477242 DOI: 10.1055/s-0031-1278237] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Available information regarding the relation among atherosclerosis, low-density lipoproteins, markers of thrombosis, inflammation and endothelial dysfunction has accumulated, but is still very limited, making only minimal contributions to clinical decision-making. Many more clinical trials are needed, but unless there is a relationship between atherosclerosis prevention, specific markers and a pharmaceutical product, financial support for such trials will be difficult to obtain. The anti-inflammatory effect of statins is well established. Angiotensin-converting enzyme inhibitors are generally not thought of as having anti-inflammatory effects, but the European Trial on Reduction of Cardiac Events with Perindopril in Stable Coronary Artery Disease (EUROPA) study observed extensive RR reduction with perindopril. It was explained not simply by control of hypertension, but by reduced activity of multiple factors, supported by specific substudies. The 'cardiovascular continuum' is an excellent unifying term to explain atherosclerosis mechanisms, relate mechanisms to clinical understanding, and assist the clinician in selecting the appropriate prevention and control therapies. This so-called continuum actually describes a relationship among different biochemical, enzymatic and hormonal factors that affect the cardiovascular system. It can be seen in the downregulation of the angiotensin II receptor type 1 by statins, which contributes to hypertension control while lowering low-density lipoproteins. Peroxisome proliferator activator receptor-gamma also demonstrates the cardiovascular continuum with activation of the receptor by glitazones. The glitazones increase insulin sensitivity for diabetes control. Activation of the peroxisome proliferator activator receptor-gamma inhibits inflammation, which is possibly related to atherosclerosis, normalization of endothelial function, suppression of metalloproteinases and a decrease in smooth muscle cell migration. All of these effects may decrease atherosclerosis production while improving control of diabetes mellitus, a key disease in the cardiovascular continuum for development of atherosclerosis. Consideration of such interrelationships is just scratching the surface. Nevertheless, it can be seen that the complicated process of atherosclerosis development has a multifaceted explanation that has been minimally defined, but holds the key to prevention and control of this major medical problem faced in modern society.
Collapse
Affiliation(s)
- Thomas F Whayne
- Division of Cardiovascular Medicine, Gill Heart Institute, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
48
|
Kamstrup PR, Tybjærg-Hansen A, Nordestgaard BG. Genetic Evidence That Lipoprotein(a) Associates With Atherosclerotic Stenosis Rather Than Venous Thrombosis. Arterioscler Thromb Vasc Biol 2012; 32:1732-41. [DOI: 10.1161/atvbaha.112.248765] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Objective—
The aim of the present study was to determine whether lipoprotein(a) [Lp(a)], considered a causal risk factor for cardiovascular disease, primarily promotes thrombosis or atherosclerosis.
Methods and Results—
Using a Mendelian randomization study design, we measured plasma Lp(a) and genetically elevated Lp(a) levels through the
LPA
kringle IV type 2 repeat genotype in 41231 individuals. We included 2 general population studies of both venous thrombosis and combined thrombosis and atherosclerosis in coronary arteries (=myocardial infarction), and 3 case–control studies of atherosclerotic stenosis. Neither Lp(a) tertiles nor
LPA
kringle IV type 2 tertiles associated with the risk of venous thrombosis in general population studies (trend:
P
=0.12–0.76), but did each associate with risk of coronary, carotid, and femoral atherosclerotic stenosis in case–control studies (trend:
P
<0.001 to 0.04). Lp(a) and
LPA
kringle IV type 2 tertiles also associated with the risk of myocardial infarction in general population studies (trend:
P
<0.001 to 0.003). For doubling of Lp(a) levels, instrumental variable estimates of hazard/odds ratios were 1.02 (95% CI 0.90–1.15) and 1.04 (0.93–1.16) for venous thrombosis in the 2 general population studies, 1.12 (1.01–1.25), 1.17 (1.05–1.32), and 1.16 (1.01–1.35), respectively, for coronary, carotid, and femoral atherosclerotic stenosis in case–control studies, and 1.21 (1.10–1.33) and 1.17 (1.05–1.29) for myocardial infarction in general population studies.
Conclusion—
This supports that Lp(a) primarily promotes atherosclerotic stenosis rather than venous thrombosis.
Collapse
Affiliation(s)
- Pia R. Kamstrup
- From the Department of Clinical Biochemistry, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark (P.R.K., A.T-H.); The Copenhagen General Population Study (P.R.K., A.T-H., B.G.N.) and Department of Clinical Biochemistry (P.R.K., B.G.N.), Herlev Hospital, Copenhagen University Hospital, Herlev, Denmark; The Copenhagen City Heart Study, Bispebjerg Hospital, Copenhagen University Hospital, Copenhagen, Denmark (A.T-H., B.G.N.); and Faculty of Health Sciences, University of Copenhagen,
| | - Anne Tybjærg-Hansen
- From the Department of Clinical Biochemistry, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark (P.R.K., A.T-H.); The Copenhagen General Population Study (P.R.K., A.T-H., B.G.N.) and Department of Clinical Biochemistry (P.R.K., B.G.N.), Herlev Hospital, Copenhagen University Hospital, Herlev, Denmark; The Copenhagen City Heart Study, Bispebjerg Hospital, Copenhagen University Hospital, Copenhagen, Denmark (A.T-H., B.G.N.); and Faculty of Health Sciences, University of Copenhagen,
| | - Børge G. Nordestgaard
- From the Department of Clinical Biochemistry, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark (P.R.K., A.T-H.); The Copenhagen General Population Study (P.R.K., A.T-H., B.G.N.) and Department of Clinical Biochemistry (P.R.K., B.G.N.), Herlev Hospital, Copenhagen University Hospital, Herlev, Denmark; The Copenhagen City Heart Study, Bispebjerg Hospital, Copenhagen University Hospital, Copenhagen, Denmark (A.T-H., B.G.N.); and Faculty of Health Sciences, University of Copenhagen,
| |
Collapse
|
49
|
Nenseter MS, Lindvig HW, Ueland T, Langslet G, Ose L, Holven KB, Retterstøl K. Lipoprotein(a) levels in coronary heart disease-susceptible and -resistant patients with familial hypercholesterolemia. Atherosclerosis 2011; 216:426-32. [PMID: 21376325 DOI: 10.1016/j.atherosclerosis.2011.02.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2010] [Revised: 02/02/2011] [Accepted: 02/03/2011] [Indexed: 10/18/2022]
|
50
|
Kamstrup PR, Tybjærg-Hansen A, Nordestgaard BG. Lipoprotein(a) and risk of myocardial infarction--genetic epidemiologic evidence of causality. Scandinavian Journal of Clinical and Laboratory Investigation 2011; 71:87-93. [PMID: 21231777 DOI: 10.3109/00365513.2010.550311] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Elevated levels of lipoprotein(a) are associated with an increased risk of myocardial infarction. Our study aimed to test whether genetic data are consistent with this association being causal. Accordingly, we developed a high-throughput realtime PCR assay to genotype for the lipoprotein(a) kringle IV type 2 (KIV-2) repeat polymorphism in the LPA gene in > 40,000 individuals. The LPA KIV-2 genotype associated with plasma levels of lipoprotein(a) (trend p < 0.001), and the LPA KIV-2 genotype associated with risk of myocardial infarction (trend p < 0.001 to 0.03) in a manner consistent with its effect on plasma levels of lipoprotein(a). The association of LPA KIV-2 genotypes raising plasma levels of lipoprotein(a) with increased risk of myocardial infarction strongly supports a causal association of lipoprotein(a) with risk of myocardial infarction.
Collapse
Affiliation(s)
- Pia R Kamstrup
- Department of Clinical Biochemistry, Herlev Hospital, Copenhagen University Hospital, Copenhagen, Denmark.
| | | | | |
Collapse
|