1
|
Katsiki N, Filippatos T, Vlachopoulos C, Panagiotakos D, Milionis H, Tselepis A, Garoufi A, Rallidis L, Richter D, Nomikos T, Kolovou G, Kypreos K, Chrysohoou C, Tziomalos K, Skoumas I, Koutagiar I, Attilakos A, Papagianni M, Boutari C, Kotsis V, Pitsavos C, Elisaf M, Tsioufis K, Liberopoulos E. Executive summary of the Hellenic Atherosclerosis Society guidelines for the diagnosis and treatment of dyslipidemias - 2023. ATHEROSCLEROSIS PLUS 2024; 55:74-92. [PMID: 38425675 PMCID: PMC10901915 DOI: 10.1016/j.athplu.2024.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 12/20/2023] [Accepted: 01/29/2024] [Indexed: 03/02/2024]
Abstract
Atherosclerotic cardiovascular disease (ASCVD) remains the main cause of death worldwide, and thus its prevention, early diagnosis and treatment is of paramount importance. Dyslipidemia represents a major ASCVD risk factor that should be adequately managed at different clinical settings. 2023 guidelines of the Hellenic Atherosclerosis Society focus on the assessment of ASCVD risk, laboratory evaluation of dyslipidemias, new and emerging lipid-lowering drugs, as well as diagnosis and treatment of lipid disorders in women, the elderly and in patients with familial hypercholesterolemia, acute coronary syndromes, heart failure, stroke, chronic kidney disease, diabetes, autoimmune diseases, and non-alcoholic fatty liver disease. Statin intolerance is also discussed.
Collapse
Affiliation(s)
- N Katsiki
- Department of Nutritional Sciences and Dietetics, International Hellenic University, Thessaloniki, Greece
- School of Medicine, European University Cyprus, Nicosia, Cyprus
| | - Td Filippatos
- Department of Internal Medicine, School of Medicine, University of Crete, Crete, Greece
| | - C Vlachopoulos
- Cardiology Department, First Cardiology Clinic, Athens Medical School, Hippokration Hospital, Athens, Greece
| | - D Panagiotakos
- Department of Nutrition and Dietetics, School of Health Sciences & Education, Harokopio University, Athens, Greece
| | - H Milionis
- Department of Internal Medicine, School of Health Sciences, Faculty of Medicine, University of Ioannina, Ioannina, Greece
| | - A Tselepis
- Atherothrombosis Research Centre, University of Ioannina, Ioannina, Greece
| | - A Garoufi
- 2nd Department of Pediatrics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - L Rallidis
- 2nd Department of Cardiology, Medical School, National and Kapodistrian University of Athens, University General Hospital ATTIKON, Athens, Greece
| | - D Richter
- Head of Cardiac Department, Euroclinic Hospital, Athens, Greece
| | - T Nomikos
- Department of Nutrition and Dietetics, School of Health Sciences & Education, Harokopio University, Athens, Greece
| | - G Kolovou
- Metropolitan Hospital, Cardiometabolic Center, Lipoprotein Apheresis and Lipid Disorders Clinic, Athens, Greece
| | - K Kypreos
- School of Medicine, European University Cyprus, Nicosia, Cyprus
- University of Patras, School of Health Science, Department of Medicine, Pharmacology Laboratory, Patras, 26500, Greece
| | - C Chrysohoou
- 1st Cardiology Clinic National and Kapodistrian University of Athens, Hippokration Hospital, Athens, Greece
| | - K Tziomalos
- First Propedeutic Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, AHEPA Hospital, Thessaloniki, Greece
| | - I Skoumas
- 1st Department of Cardiology, National & Kapodistrian University of Athens, Athens, Greece
| | - I Koutagiar
- 1st Cardiology Department, Hygeia Hospital, Athens, Greece
| | - A Attilakos
- 3rd Department of Pediatrics, School of Medicine, National and Kapodistrian University of Athens, Attikon General Hospital, Athens, Greece
| | - M Papagianni
- Third Department of Pediatrics, Aristotle University of Thessaloniki, School of Medicine, “Hippokrateion" General Hospital of Thessaloniki, Thessaloniki, Greece
| | - C Boutari
- Second Propedeutic Department of Internal Medicine, Hippocration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - V Kotsis
- 3rd Department of Internal Medicine, Papageorgiou Hospital, Aristotle University Thessaloniki, Greece
| | - C Pitsavos
- First Cardiology Clinic, School of Medicine, University of Athens, Greece
| | - M Elisaf
- Department of Internal Medicine, Faculty of Medicine, University Hospital of Ioannina, 45110, Ioannina, Greece
| | - K Tsioufis
- 1st Department of Cardiology, National and Kapodistrian University of Athens, Hippocration Hospital, Greece
| | - E Liberopoulos
- 1st Department of Propedeutic Medicine, School of Medicine, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
| |
Collapse
|
2
|
Hober A, Rekanovic M, Forsström B, Hansson S, Kotol D, Percy AJ, Uhlén M, Oscarsson J, Edfors F, Miliotis T. Targeted proteomics using stable isotope labeled protein fragments enables precise and robust determination of total apolipoprotein(a) in human plasma. PLoS One 2023; 18:e0281772. [PMID: 36791076 PMCID: PMC9931122 DOI: 10.1371/journal.pone.0281772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 01/31/2023] [Indexed: 02/16/2023] Open
Abstract
Lipoprotein(a), also known as Lp(a), is an LDL-like particle composed of apolipoprotein(a) (apo(a)) bound covalently to apolipoprotein B100. Plasma concentrations of Lp(a) are highly heritable and vary widely between individuals. Elevated plasma concentration of Lp(a) is considered as an independent, causal risk factor of cardiovascular disease (CVD). Targeted mass spectrometry (LC-SRM/MS) combined with stable isotope-labeled recombinant proteins provides robust and precise quantification of proteins in the blood, making LC-SRM/MS assays appealing for monitoring plasma proteins for clinical implications. This study presents a novel quantitative approach, based on proteotypic peptides, to determine the absolute concentration of apo(a) from two microliters of plasma and qualified according to guideline requirements for targeted proteomics assays. After optimization, assay parameters such as linearity, lower limits of quantification (LLOQ), intra-assay variability (CV: 4.7%) and inter-assay repeatability (CV: 7.8%) were determined and the LC-SRM/MS results were benchmarked against a commercially available immunoassay. In summary, the measurements of an apo(a) single copy specific peptide and a kringle 4 specific peptide allow for the determination of molar concentration and relative size of apo(a) in individuals.
Collapse
Affiliation(s)
- Andreas Hober
- Science for Life Laboratory, Solna, Sweden
- Division of Systems Biology, Department of Protein Science, School of Chemistry, Biotechnology and Health, The Royal Institute of Technology (KTH), Stockholm, Sweden
| | - Mirela Rekanovic
- Translational Science and Experimental Medicine, Cardiovascular, Renal and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Björn Forsström
- Science for Life Laboratory, Solna, Sweden
- Division of Systems Biology, Department of Protein Science, School of Chemistry, Biotechnology and Health, The Royal Institute of Technology (KTH), Stockholm, Sweden
| | - Sara Hansson
- Translational Science and Experimental Medicine, Cardiovascular, Renal and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - David Kotol
- Science for Life Laboratory, Solna, Sweden
- Division of Systems Biology, Department of Protein Science, School of Chemistry, Biotechnology and Health, The Royal Institute of Technology (KTH), Stockholm, Sweden
| | - Andrew J. Percy
- Department of Applications Development, Cambridge Isotope Laboratories, Inc., Tewksbury, Massachusetts, United States of America
| | - Mathias Uhlén
- Science for Life Laboratory, Solna, Sweden
- Division of Systems Biology, Department of Protein Science, School of Chemistry, Biotechnology and Health, The Royal Institute of Technology (KTH), Stockholm, Sweden
| | - Jan Oscarsson
- Late-stage Development, Cardiovascular, Renal and Metabolism, Biopharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Fredrik Edfors
- Science for Life Laboratory, Solna, Sweden
- Division of Systems Biology, Department of Protein Science, School of Chemistry, Biotechnology and Health, The Royal Institute of Technology (KTH), Stockholm, Sweden
| | - Tasso Miliotis
- Translational Science and Experimental Medicine, Cardiovascular, Renal and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
- * E-mail:
| |
Collapse
|
3
|
Familial Hypercholesterolemia and Lipoprotein(a): A Gordian Knot in Cardiovascular Prevention. Metabolites 2022; 12:metabo12111065. [PMID: 36355148 PMCID: PMC9693181 DOI: 10.3390/metabo12111065] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/01/2022] [Accepted: 11/02/2022] [Indexed: 11/06/2022] Open
Abstract
Familial hypercholesterolemia (FH) is the most frequent genetic disorder resulting in increased low-density lipoprotein cholesterol (LDL-C) levels from childhood, leading to premature atherosclerotic cardiovascular disease (ASCVD) if left untreated. FH diagnosis is based on clinical criteria and/or genetic testing and its prevalence is estimated as being up to 1:300,000−400,000 for the homozygous and ~1:200−300 for the heterozygous form. Apart from its late diagnosis, FH is also undertreated, despite the available lipid-lowering therapies. In addition, elevated lipoprotein(a) (Lp(a)) (>50 mg/dL; 120 nmol/L), mostly genetically determined, has been identified as an important cardiovascular risk factor with prevalence rate of ~20% in the general population. Novel Lp(a)-lowering therapies have been recently developed and their cardiovascular efficacy is currently investigated. Although a considerable proportion of FH patients is also diagnosed with high Lp(a) levels, there is a debate whether these two entities are associated. Nevertheless, Lp(a), particularly among patients with FH, has been established as a significant cardiovascular risk factor. In this narrative review, we present up-to-date evidence on the pathophysiology, diagnosis, and treatment of both FH and elevated Lp(a) with a special focus on their association and joint effect on ASCVD risk.
Collapse
|
4
|
Yang Y, Hong Y, Yang W, Zheng Z. Association of lipoprotein(a) with aortic dissection. Clin Cardiol 2022; 45:908-912. [PMID: 35925003 PMCID: PMC9451660 DOI: 10.1002/clc.23834] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 03/28/2022] [Accepted: 04/07/2022] [Indexed: 11/10/2022] Open
Abstract
Background Lipoprotein(a) [Lp(a)] is associated with coronary atherosclerotic heart disease, aortic stenosis, stroke, and heart failure. We aimed to determine the relationship between Lp(a) and aortic dissection (AD). Methods Two hundred patients with AD were included in our case group. The control group consisted of 200 non‐AD people who were age‐ (±5 years) and gender‐matched to the case group. Data were collected retrospectively, including hypertension, smoking, coronary artery disease, diabetes mellitus, Lp(a), total cholesterol, triglyceride, low‐density lipoprotein cholesterol, and high‐density lipoprotein cholesterol. The association between Lp(a) and AD was studied using univariate and multivariate logistic regression analysis. Results Patients with AD had greater median Lp(a) concentrations than non‐AD people (152.50 vs. 81.75 mg/L). Lp(a) was associated with AD in a multivariate logistic regression analysis (odds ratio, 8.03; 95% confidence interval, 2.85–22.62), comparing those with Lp(a) quartile 4 with those with Lp(a) quartile 1. Stratified analysis showed that this relationship was observed in both men and women, as well as in older and younger individuals. Conclusions High levels of Lp(a) are strongly associated with AD, independent of other cardiovascular risk factors.
Collapse
Affiliation(s)
- Yiheng Yang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yuting Hong
- Department of Renal Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Weihua Yang
- Department of Cardiovascular Medicine, Affiliated Renhe Hospital of China Three Gorges University, Yichang, Hubei, China
| | - Zhenzhong Zheng
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
5
|
Joseph J, Menon JC, Sebastien PK, Sudhakar A, John D, Menon GR. Association of lipoprotein (a) with coronary artery disease in a South Asian population: A case-control study. PLoS One 2022; 17:e0267807. [PMID: 35503788 PMCID: PMC9064091 DOI: 10.1371/journal.pone.0267807] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 04/16/2022] [Indexed: 11/19/2022] Open
Abstract
Introduction
Coronary artery disease (CAD), the leading cause of mortality worldwide, is characterised by an earlier onset and more severe disease in South Asians as compared to Western populations.
Methods
This is an observational study on 928 individuals who attended three tertiary care centres in Kerala, India from 2014-to 2017. The demographic, anthropometric, behavioural factors and the lipoprotein (Lp(a)) and cholesterol values were compared between the two groups and across disease severity. The Chi-square test was used to compare the categorical variables and independent sample t-test for the continuous variables. Multivariable logistic regression was performed to investigate the association of demographic, clinical and behavioural factors with the disease. Odds ratios are presented with a 95% confidence interval. In individuals below 50 years, two logistic regression models were compared to investigate the improvement in modelling the association of the independent factors and Lp(a) with the occurrence of the disease.
Results
We included 682 patients in the diseased group and 246 patients treated for non-coronary conditions in the control group. Those in the control group were significantly younger than in the diseased group(p<0.002). Significantly more patients were diabetic, hypertensive, tobacco users and consumers of alcohol in the diseased group. Multivariable logistic regression on data from all age groups showed that age (OR = 2.55, 95% CI 1.51–4.33, p = 0.01), diabetes (OR = 3.71, 95% CI 2.42–5.70, p = 0.01), hypertension (OR = 3.03, 95% CI 2.12–4.34, p = 0.01) and tobacco use (OR = 5.44, 95% CI 3.39–8.75, p = 0.01) are significantly associated with the disease. Lp(a) (OR = 1.22, 95% CI 0.87–1.72) increased the odds of the disease by 22% but was not statistically significant. In individuals below 50 years, Lp(a) significantly increased the likelihood of CAD (OR = 3.52, 95% CI 1.63–7.57, p = 0.01). Those with diabetes were seven times more likely to be diseased (OR = 7.06, 95% CI 2.59–19.21, p = 0.01) and the tobacco users had six times the likelihood of disease occurrence (OR = 6.38, 95% CI 2.62–15.54, p = 0.01). The median Lp(a) values showed a statistically significant increasing trend with the extent/severity of the disease in those below 50 years.
Conclusion
Age, diabetes, hypertension and tobacco use appear to be associated more with the occurrence of coronary artery disease in adults of all ages. Lipoprotein(a), cholesterol and BMI categories do not seem to be related to disease status in all ages. However, in individuals below 50 years, diabetes, tobacco use and lipoprotein (a) are significantly associated with the occurrence of the disease.
Collapse
Affiliation(s)
- Jacob Joseph
- Department of Cardiology, Lisie Hospital, Ernakulam, Kerala
| | | | | | - Abish Sudhakar
- Department of Pediatric Cardiology, Amrita Institute of Medical Sciences & Research Centre, Kochi, Kerala
| | - Denny John
- Adjunct Faculty, Public Health, Amrita Institute of Medical Sciences & Research Centre, Kochi, Kerala
| | - Geetha R Menon
- ICMR-National Institute of Medical Statistics, New Delhi, India
| |
Collapse
|
6
|
Hu J, Lei H, Liu L, Xu D. Lipoprotein(a), a Lethal Player in Calcific Aortic Valve Disease. Front Cell Dev Biol 2022; 10:812368. [PMID: 35155427 PMCID: PMC8830536 DOI: 10.3389/fcell.2022.812368] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 01/07/2022] [Indexed: 12/11/2022] Open
Abstract
Calcified aortic valve disease (CAVD) is the most common valvular cardiovascular disease with increasing incidence and mortality. The primary treatment for CAVD is surgical or transcatheter aortic valve replacement and there remains a lack of effective drug treatment. Recently, lipoprotein (a) (Lp(a)) has been considered to play a crucial role in CAVD pathophysiology. Multiple studies have shown that Lp(a) represents an independent risk factor for CAVD. Moreover, Lp(a) mediates the occurrence and development of CAVD by affecting aortic valve endothelial dysfunction, indirectly promoting foam cell formation through oxidized phospholipids (OxPL), inflammation, oxidative stress, and directly promotes valve calcification. However, there is a lack of clinical trials with Lp(a) reduction as a primary endpoint. This review aims to explore the relationship and mechanism between Lp(a) and CAVD, and focuses on the current drugs that can be used as potential therapeutic targets for CAVD.
Collapse
Affiliation(s)
- Jiahui Hu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, China
- Cardiovascular Disease Research Center of Hunan Province, Changsha, China
| | - Hao Lei
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, China
- Cardiovascular Disease Research Center of Hunan Province, Changsha, China
| | - Leiling Liu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, China
- Cardiovascular Disease Research Center of Hunan Province, Changsha, China
| | - Danyan Xu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, China
- Cardiovascular Disease Research Center of Hunan Province, Changsha, China
- *Correspondence: Danyan Xu,
| |
Collapse
|
7
|
Diet and Lp(a): Does Dietary Change Modify Residual Cardiovascular Risk Conferred by Lp(a)? Nutrients 2020; 12:nu12072024. [PMID: 32646066 PMCID: PMC7400957 DOI: 10.3390/nu12072024] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/01/2020] [Accepted: 07/03/2020] [Indexed: 02/06/2023] Open
Abstract
Lipoprotein(a) [Lp(a)] is an independent, causal, genetically determined risk factor for cardiovascular disease (CVD). We provide an overview of current knowledge on Lp(a) and CVD risk, and the effect of pharmacological agents on Lp(a). Since evidence is accumulating that diet modulates Lp(a), the focus of this paper is on the effect of dietary intervention on Lp(a). We identified seven trials with 15 comparisons of the effect of saturated fat (SFA) replacement on Lp(a). While replacement of SFA with carbohydrate, monounsaturated fat (MUFA), or polyunsaturated fat (PUFA) consistently lowered low-density lipoprotein cholesterol (LDL-C), heterogeneity in the Lp(a) response was observed. In two trials, Lp(a) increased with carbohydrate replacement; one trial showed no effect and another showed Lp(a) lowering. MUFA replacement increased Lp(a) in three trials; three trials showed no effect and one showed lowering. PUFA or PUFA + MUFA inconsistently affected Lp(a) in four trials. Seven trials of diets with differing macronutrient compositions showed similar divergence in the effect on LDL-C and Lp(a). The identified clinical trials show diet modestly affects Lp(a) and often in the opposing direction to LDL-C. Further research is needed to understand how diet affects Lp(a) and its properties, and the lack of concordance between diet-induced LDL-C and Lp(a) changes.
Collapse
|
8
|
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: 9.8] [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
|
9
|
Tada H, Yamagami K, Nishikawa T, Yoshida T, Teramoto R, Sakata K, Takamura M, Kawashiri MA. Lipoprotein (a) and the Risk of Chronic Kidney Disease in Hospitalized Japanese Patients. Intern Med 2020; 59:1705-1710. [PMID: 32669515 PMCID: PMC7434553 DOI: 10.2169/internalmedicine.4503-20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Objective Lipoprotein (a), or Lp (a), has been shown to be associated with the development of chronic kidney disease (CKD) in populations of various ethnicities. This study aimed to investigate the association between serum Lp (a) and CKD in Japanese patients. Methods A total of 6,130 subjects who underwent a serum Lp (a) level assessment for any reason (e.g. any type of surgery requiring prolonged bed rest or risk factors for atherosclerosis, such as hypertension or diabetes) were retrospectively investigated at Kanazawa University Hospital from April 2004 to March 2014. Of these, 1,895 subjects were excluded because of the lack of clinical data. Subjects were assessed for Lp (a), low-density lipoprotein (LDL) cholesterol, high-density lipoprotein (HDL) cholesterol, triglycerides, hypertension, diabetes, smoking, body mass index (BMI), coronary artery disease (CAD), and CKD (stage ≥3). Results When the study subjects were divided into quartiles of Lp (a) levels, significant trends were observed with regard to the presence of CKD (p = 2.7×10-13). A multiple regression analysis showed that Lp (a) was significantly associated with CKD [odds ratio (OR), 1.12; 95% confidence interval (CI), 1.08-1.17; p = 1.3×10-7, per 10 mg/dL], independent of other classical risk factors, including age, gender, BMI, hypertension, diabetes, smoking, LDL cholesterol, and triglycerides. Under these conditions, Lp (a) was significantly associated with CAD (OR = 1.11, 95% CI = 1.06-1.16; p = 1.7×10-6, per 10 mg/dL), independent of other risk factors. Conclusion Serum Lp (a) was associated with CKD, independent of other classical risk factors in a Japanese population.
Collapse
Affiliation(s)
- Hayato Tada
- Department of Cardiovascular Medicine, Kanazawa University Graduate School of Medical Sciences, Japan
| | - Kan Yamagami
- Department of Cardiovascular Medicine, Kanazawa University Graduate School of Medical Sciences, Japan
| | - Tetsuo Nishikawa
- Department of Cardiovascular Medicine, Kanazawa University Graduate School of Medical Sciences, Japan
| | - Taiji Yoshida
- Department of Cardiovascular Medicine, Kanazawa University Graduate School of Medical Sciences, Japan
| | - Ryota Teramoto
- Department of Cardiovascular Medicine, Kanazawa University Graduate School of Medical Sciences, Japan
| | - Kenji Sakata
- Department of Cardiovascular Medicine, Kanazawa University Graduate School of Medical Sciences, Japan
| | - Masayuki Takamura
- Department of Cardiovascular Medicine, Kanazawa University Graduate School of Medical Sciences, Japan
| | - Masa-Aki Kawashiri
- Department of Cardiovascular Medicine, Kanazawa University Graduate School of Medical Sciences, Japan
| |
Collapse
|
10
|
Tuten A, Gungor Z, Ekmekci H, Ekmekci OB, Kucur M, Yilmaz N, Donma O, Sonmez H, Acıkgoz A, Madazlı R. Relationship between LPA SNPs and inflammatory burden in patients with preeclampsia to address future cardiovascular risk. J Matern Fetal Neonatal Med 2019; 34:898-906. [PMID: 31113255 DOI: 10.1080/14767058.2019.1622667] [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] [Indexed: 12/12/2022]
Abstract
OBJECTIVE The study tested whether cardiovascular corresponding LPA risk genotypes improve pre-eclampsia and coronary heart disease (CHD) risk prediction beyond conventional risk factors. BACKGROUND Studies have shown that women specific risk factors for cardiovascular disease (CVD) have taken an attention recently. It might be possible to identify women who have the highest risk in developing CVD in their further lives. It is well-known that Lp(a) levels have an impact on increased risk of CVD which is affected by LPA gene. Further, LPA risk genotypes are not considered in cardiovascular risk prediction. METHODS We have included 200 pregnant Turkish women into the study. We stratified the preeclamptic (PE) group: early (EOP) (28.7 ± 3.0 weeks) and late onset (LOP) (36.0 ± 1.4 weeks). 14 LPA SNPs were evaluated in the study. Rs9355296 and rs3798220 were found as independent risk factors for preeclampsia by logistic regression analysis. A positive correlation was found between rs9355296 and the diagnostic criteria of preeclampsia. Further rs9355296 G/* carriers have higher vascular inflammation rather than AA carriers. CONCLUSIONS The findings reveal that LPA genetic variability with high inflammatory response might be an indication of future cardiovascular events.
Collapse
Affiliation(s)
- Abdullah Tuten
- Faculty of Medicine, University of Istanbul, Cerrahpasa Medical School, Istanbul, Turkey
| | - Zeynep Gungor
- Faculty of Medicine, University of Istanbul, Cerrahpasa Medical School, Istanbul, Turkey
| | - Hakan Ekmekci
- Faculty of Medicine, University of Istanbul, Cerrahpasa Medical School, Istanbul, Turkey
| | - Ozlem Balci Ekmekci
- Faculty of Medicine, University of Istanbul, Cerrahpasa Medical School, Istanbul, Turkey
| | - Mine Kucur
- Faculty of Medicine, University of Istanbul, Cerrahpasa Medical School, Istanbul, Turkey
| | - Nevin Yilmaz
- Faculty of Medicine, University of Istanbul, Cerrahpasa Medical School, Istanbul, Turkey
| | - Orkide Donma
- Faculty of Medicine, University of Istanbul, Cerrahpasa Medical School, Istanbul, Turkey
| | - Huseyin Sonmez
- Faculty of Medicine, University of Istanbul, Cerrahpasa Medical School, Istanbul, Turkey
| | - Abdullah Acıkgoz
- Faculty of Medicine, University of Istanbul, Cerrahpasa Medical School, Istanbul, Turkey
| | - Rıza Madazlı
- Faculty of Medicine, University of Istanbul, Cerrahpasa Medical School, Istanbul, Turkey
| |
Collapse
|
11
|
Lipoprotein(a). Clin Res Cardiol Suppl 2019; 14:1-4. [PMID: 30945119 DOI: 10.1007/s11789-019-00100-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
12
|
A Meta-Analysis of the Effect of PCSK9-Monoclonal Antibodies on Circulating Lipoprotein (a) Levels. Am J Cardiovasc Drugs 2019; 19:87-97. [PMID: 30229525 DOI: 10.1007/s40256-018-0303-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Lipoprotein (a) [Lp(a)] is an atherogenic lipoprotein. While no effective therapy for Lp(a) is currently available, recently, several pooled analyses with small sample sizes have suggested that proprotein convertase subtilisin/kexin type 9 monoclonal antibodies (PCSK9-mAbs) could reduce circulating Lp(a) levels. This meta-analysis was performed to comprehensively investigate the efficacy of PCSK9-mAbs with respect to serum Lp(a) concentrations. METHODS PubMed, MEDLINE, Embase, ClinicalTrials.gov, Cochrane CENTRAL, Web of Science and recent conferences up to July 2018 were searched. Randomized clinical trials evaluating the effect of PCSK9-mAbs and control treatment on plasma Lp(a) concentrations were included. Mean differences and odds ratios with 95% confidence intervals (CIs) were used. RESULTS Twenty-seven randomized clinical trials with a total of 11,864 participants were included. PCSK9-mAbs showed a significant efficacy in reducing Lp(a) (- 21.9%, 95% CI - 24.3 to - 19.5), irrespective of PCSK9-mAb types, treatment duration, participant characteristics, treatment methods, differences of control treatment, baseline Lp(a) levels, and test methods. The greatest reduction was achieved with 150 mg alirocumab biweekly (- 24.6%, 95% CI - 28.0 to - 21.2) and 140 mg evolocumab monthly (- 26.8%, 95% CI - 31.6 to - 21.9). Meta-regression analyses found that the more intense low-density lipoprotein cholesterol levels declined during PCSK9-mAb treatment, the greater the reduction in Lp(a) levels. Safety was in accordance with previous reports. CONCLUSIONS The results of this analysis suggested that PCSK9-mAbs could significantly reduce circulating Lp(a) levels. Long-term studies may be needed to confirm the effect of PCSK9-mAbs on Lp(a) in the future.
Collapse
|
13
|
Kostner KM, Kostner GM, Wierzbicki AS. Is Lp(a) ready for prime time use in the clinic? A pros-and-cons debate. Atherosclerosis 2018; 274:16-22. [PMID: 29747086 DOI: 10.1016/j.atherosclerosis.2018.04.032] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 04/16/2018] [Accepted: 04/25/2018] [Indexed: 12/11/2022]
Abstract
Lipoprotein (a) (Lp(a)) is a cholesterol-rich lipoprotein known since 1963. In spite of extensive research on Lp(a), there are still numerous gaps in our knowledge relating to its function, biosynthesis and catabolism. One reason for this might be that apo(a), the characteristic glycoprotein of Lp(a), is expressed only in primates. Results from experiments using transgenic animals therefore may need verification in humans. Studies on Lp(a) are also handicapped by the great number of isoforms of apo(a) and the heterogeneity of apo(a)-containing fractions in plasma. Quantification of Lp(a) in the clinical laboratory for a long time has not been standardized. Starting from its discovery, reports accumulated that Lp(a) contributed to the risk of cardiovascular disease (CVD), myocardial infarction (MI) and stroke. Early reports were based on case control studies but in the last decades a great deal of prospective studies have been published that highlight the increased risk for CVD and MI in patients with elevated Lp(a). Final answers to the question of whether Lp(a) is ready for wider clinical use will come from intervention studies with novel selective Lp(a) lowering medications that are currently underway. This article expounds arguments for and against this proposition from currently available data.
Collapse
Affiliation(s)
- Karam M Kostner
- Department of Cardiology, Mater Hospital and University of Queensland, Brisbane, Australia
| | - Gert M Kostner
- Department of Molecular Biology and Biochemistry, Gottfried Schatz Research Center for Cell Signaling, Medical University of Graz, Austria
| | - Anthony S Wierzbicki
- Department of Metabolic Medicine/Chemical Pathology, Guy's & St Thomas' Hospitals, London, UK.
| |
Collapse
|
14
|
Tselepis AD. Oxidized phospholipids and lipoprotein-associated phospholipase A 2 as important determinants of Lp(a) functionality and pathophysiological role. J Biomed Res 2018; 31. [PMID: 27346583 PMCID: PMC5956253 DOI: 10.7555/jbr.31.20160009] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 01/29/2016] [Accepted: 02/12/2016] [Indexed: 12/23/2022] Open
Abstract
Lipoprotein(a) [Lp(a)] is composed of a low density lipoprotein (LDL)-like particle to which apolipoprotein (a) [apo(a)] is linked by a single disulfide bridge. Lp(a) is considered a causal risk factor for ischemic cardiovascular disease (CVD) and calcific aortic valve stenosis (CAVS). The evidence for a causal role of Lp(a) in CVD and CAVS is based on data from large epidemiological databases, mendelian randomization studies, and genome-wide association studies. Despite the well-established role of Lp(a) as a causal risk factor for CVD and CAVS, the underlying mechanisms are not well understood. A key role in the Lp(a) functionality may be played by its oxidized phospholipids (OxPL) content. Importantly, most of circulating OxPL are associated with Lp(a); however, the underlying mechanisms leading to this preferential sequestration of OxPL on Lp(a) over the other lipoproteins, are mostly unknown. Several studies support the hypothesis that the risk of Lp(a) is primarily driven by its OxPL content. An important role in Lp(a) functionality may be played by the lipoprotein-associated phospholipase A2 (Lp-PLA2), an enzyme that catalyzes the degradation of OxPL and is bound to plasma lipoproteins including Lp(a). The present review article discusses new data on the pathophysiological role of Lp(a) and particularly focuses on the functional role of OxPL and Lp-PLA2 associated with Lp(a).
Collapse
Affiliation(s)
- Alexandros D Tselepis
- Atherothrombosis Research Centre / Laboratory of Biochemistry, Department of Chemistry, University of Ioannina, 45110 Ioannina, Greece.
| |
Collapse
|
15
|
Yu B, Hafiane A, Thanassoulis G, Ott L, Filwood N, Cerruti M, Gourgas O, Shum-Tim D, Al Kindi H, de Varennes B, Alsheikh-Ali A, Genest J, Schwertani A. Lipoprotein(a) Induces Human Aortic Valve Interstitial Cell Calcification. JACC Basic Transl Sci 2017; 2:358-371. [PMID: 30062157 PMCID: PMC6034440 DOI: 10.1016/j.jacbts.2017.03.015] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 03/30/2017] [Accepted: 03/30/2017] [Indexed: 12/22/2022]
Abstract
Lp(a) significantly increased alkaline phosphatase activity, phosphate and calcium content, and matrix vesicle formation and induced apoptosis and calcification of normal human aortic valve interstitial cells. The type of minerals induced by Lp(a) resembles that seen in calcified human aortic valves as shown by Raman spectroscopy. Lp(a)-induced calcification of human aortic valve interstitial cells is mediated by activation of MAPK38, GSK3β, and Wnt signaling. Inhibition of GSK3β and MAPK38 significantly reduced lipoprotein(a)-induced aortic valve interstitial cell calcification. Lp(a)is abundant in calcified aortic valves, and lipoprotein(a) immunoreactivity colocalized with that of oxidized phospholipids.
Lipoprotein(a), or Lp(a), significantly increased alkaline phosphatase activity, release of phosphate, calcium deposition, hydroxyapatite, cell apoptosis, matrix vesicle formation, and phosphorylation of signal transduction proteins; increased expression of chondro-osteogenic mediators; and decreased SOX9 and matrix Gla protein (p < 0.001). Inhibition of MAPK38 and GSK3β significantly reduced Lp(a)-induced calcification of human aortic valve interstitial cells (p < 0.001). There was abundant presence of Lp(a) and E06 immunoreactivity in diseased human aortic valves. The present study demonstrates a causal effect for Lp(a) in aortic valve calcification and suggests that interfering with the Lp(a)pathway could provide a novel therapeutic approach in the management of this debilitating disease.
Collapse
Key Words
- ALP, alkaline phosphatase
- BMP, bone morphogenetic protein
- FWHM, full width half maximum
- HAVIC, human aortic valve interstitial cell
- LDL, low-density lipoprotein
- LOX-1, oxidized LDL receptor 1
- Lp(a), lipoprotein(a)
- MAPK, mitogen-activated protein kinase
- MGP, matrix Gla protein
- OxPL, oxidized phospholipid
- Raman spectroscopy
- apo(a), apolipoprotein(a)
- mRNA, messenger ribonucleic acid
- oxidized phospholipids
- real-time PCR
- stenosis
Collapse
Affiliation(s)
- Bin Yu
- Divisions of Cardiology and Cardiac Surgery, Department of Medicine, Surgery and Pathology, McGill University, Montreal, Quebec, Canada
| | - Anouar Hafiane
- Divisions of Cardiology and Cardiac Surgery, Department of Medicine, Surgery and Pathology, McGill University, Montreal, Quebec, Canada
| | - George Thanassoulis
- Divisions of Cardiology and Cardiac Surgery, Department of Medicine, Surgery and Pathology, McGill University, Montreal, Quebec, Canada
| | - Leah Ott
- Divisions of Cardiology and Cardiac Surgery, Department of Medicine, Surgery and Pathology, McGill University, Montreal, Quebec, Canada
| | - Nial Filwood
- Divisions of Cardiology and Cardiac Surgery, Department of Medicine, Surgery and Pathology, McGill University, Montreal, Quebec, Canada
| | - Marta Cerruti
- Department of Materials Engineering, McGill University, Montreal, Quebec, Canada
| | - Ophélie Gourgas
- Department of Materials Engineering, McGill University, Montreal, Quebec, Canada
| | - Dominique Shum-Tim
- Divisions of Cardiology and Cardiac Surgery, Department of Medicine, Surgery and Pathology, McGill University, Montreal, Quebec, Canada
| | - Hamood Al Kindi
- Divisions of Cardiology and Cardiac Surgery, Department of Medicine, Surgery and Pathology, McGill University, Montreal, Quebec, Canada
| | - Benoit de Varennes
- Divisions of Cardiology and Cardiac Surgery, Department of Medicine, Surgery and Pathology, McGill University, Montreal, Quebec, Canada
| | - Alawi Alsheikh-Ali
- College of Medicine, Mohammed Bin Rashid University of Medical and Health Sciences, Dubai, United Arab Emirates
| | - Jacques Genest
- Divisions of Cardiology and Cardiac Surgery, Department of Medicine, Surgery and Pathology, McGill University, Montreal, Quebec, Canada
| | - Adel Schwertani
- Divisions of Cardiology and Cardiac Surgery, Department of Medicine, Surgery and Pathology, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
16
|
Effect of Two Lipoprotein (a)-Associated Genetic Variants on Plasminogen Levels and Fibrinolysis. G3-GENES GENOMES GENETICS 2016; 6:3525-3532. [PMID: 27605514 PMCID: PMC5100851 DOI: 10.1534/g3.116.034702] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Two genetic variants (rs3798220 and rs10455872) in the apolipoprotein (a) gene (LPA) have been implicated in cardiovascular disease (CVD), presumably through their association with lipoprotein (a) [Lp(a)] levels. While Lp(a) is recognized as a lipoprotein with atherogenic and thrombogenic characteristics, it is unclear whether or not the two Lp(a)-associated genetic variants are also associated with markers of thrombosis (i.e., plasminogen levels and fibrinolysis). In the present study, we genotyped the two genetic variants in 2919 subjects of the Old Order Amish (OOA) and recruited 146 subjects according to the carrier and noncarrier status for rs3798220 and rs10455872, and also matched for gender and age. We measured plasma Lp(a) and plasminogen levels in these subjects, and found that the concentrations of plasma Lp(a) were 2.62- and 1.73-fold higher in minor allele carriers of rs3798220 and rs10455872, respectively, compared with noncarriers (P = 2.04 × 10−17 and P = 1.64 × 10−6, respectively). By contrast, there was no difference in plasminogen concentrations between carriers and noncarriers of rs3798220 and rs10455872. Furthermore, we observed no association between carrier status of rs3798220 or rs10455872 with clot lysis time. Finally, plasminogen mRNA expression in liver samples derived from 76 Caucasian subjects was not significantly different between carriers and noncarriers of these two genetic variants. Our results provide further insight into the mechanism of action behind two genetic variants previously implicated in CVD risk and show that these polymorphisms are not major modulating factors for plasma plasminogen levels and fibrinolysis.
Collapse
|
17
|
Rather RA, Dhawan V. Genetic markers: Potential candidates for cardiovascular disease. Int J Cardiol 2016; 220:914-923. [PMID: 27416153 DOI: 10.1016/j.ijcard.2016.06.251] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Revised: 06/22/2016] [Accepted: 06/26/2016] [Indexed: 02/07/2023]
Abstract
The effective prevention of cardiovascular disease depends upon the ability to recognize the high-risk individuals at an early stage of the disease or long before the development of adverse events. Evolving technologies in the fields of proteomics, metabolomics, and genomics have played a significant role in the discovery of cardiovascular biomarkers, but so far these methods have achieved the modest success. Hence, there is a crucial need for more reliable, suitable, and lasting diagnostic and therapeutic markers to screen the disease well in time to start the clinical aid to the patients. Gene polymorphisms associated with the cardiovascular disease play a decisive role in the disease onset. Therefore, the genetic marker evaluation to classify high-risk patients from low-risk patients trends an effective approach to patient management and care. Currently, there are no genetic markers available for extensive adoption as risk factors for coronary vascular disease, yet, there are numerous promising, biologically acceptable candidates. Many of these gene biomarkers, alone or in combination, can play an essential role in the prediction of cardiovascular risk. The present review highlights some putative emerging genetic biomarkers that could facilitate more authentic and fast diagnosis of CVD. This review also briefly describes few technological approaches employed in the biomarker search.
Collapse
Affiliation(s)
- Riyaz Ahmad Rather
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Veena Dhawan
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh, India.
| |
Collapse
|
18
|
Franchini M, Capuzzo E, Liumbruno GM. Lipoprotein apheresis for the treatment of elevated circulating levels of lipoprotein(a): a critical literature review. BLOOD TRANSFUSION = TRASFUSIONE DEL SANGUE 2016; 14:413-8. [PMID: 26710351 PMCID: PMC5016300 DOI: 10.2450/2015.0163-15] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 07/28/2015] [Indexed: 11/21/2022]
Abstract
Lipoprotein(a), which consists of a low-density lipoprotein (LDL) particle linked to an apolipoprotein(a) moiety, is currently considered an independent risk factor for cardiovascular disease due to its atherogenic (LDL-like) and prothrombotic (plasminogen-like) properties. The aim of this review is to provide an overview of the current and newer therapies for lowering increased lipoprotein(a) levels, focusing on lipoprotein apheresis. After a systematic literature search, we identified ten studies which, overall, documented that lipoprotein apheresis is effective in reducing increased lipoprotein(a) levels and cardiovascular events.
Collapse
Affiliation(s)
- Massimo Franchini
- Department of Haematology and Transfusion Medicine, “Carlo Poma” Hospital, Mantua, Italy
| | - Enrico Capuzzo
- Department of Haematology and Transfusion Medicine, “Carlo Poma” Hospital, Mantua, Italy
| | | |
Collapse
|
19
|
Is lipoprotein (a) protective of dementia? Eur J Epidemiol 2016; 31:1149-1152. [DOI: 10.1007/s10654-016-0184-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 07/08/2016] [Indexed: 01/07/2023]
|
20
|
Diffenderfer MR, Lamon-Fava S, Marcovina SM, Barrett PHR, Lel J, Dolnikowski GG, Berglund L, Schaefer EJ. Distinct metabolism of apolipoproteins (a) and B-100 within plasma lipoprotein(a). Metabolism 2016; 65:381-90. [PMID: 26975530 PMCID: PMC4795479 DOI: 10.1016/j.metabol.2015.10.031] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 10/28/2015] [Accepted: 10/31/2015] [Indexed: 11/30/2022]
Abstract
OBJECTIVES Lipoprotein(a) [Lp(a)] is mainly similar in composition to LDL, but differs in having apolipoprotein (apo) (a) covalently linked to apoB-100. Our purpose was to examine the individual metabolism of apo(a) and apoB-100 within plasma Lp(a). MATERIALS AND METHODS The kinetics of apo(a) and apoB-100 in plasma Lp(a) were assessed in four men with dyslipidemia [Lp(a) concentration: 8.9-124.7nmol/L]. All subjects received a primed constant infusion of [5,5,5-(2)H3] L-leucine while in the constantly fed state. Lp(a) was immunoprecipitated directly from whole plasma; apo(a) and apoB-100 were separated by gel electrophoresis; and isotopic enrichment was determined by gas chromatography/mass spectrometry. RESULTS Multicompartmental modeling analysis indicated that the median fractional catabolic rates of apo(a) and apoB-100 within Lp(a) were significantly different at 0.104 and 0.263 pools/day, respectively (P=0.04). The median Lp(a) apo(a) production rate at 0.248nmol/kg·day(-1) was significantly lower than that of Lp(a) apoB-100 at 0.514nmol/kg·day(-1) (P=0.03). CONCLUSION Our data indicate that apo(a) has a plasma residence time (11days) that is more than twice as long as that of apoB-100 (4days) within Lp(a), supporting the concept that apo(a) and apoB-100 within plasma Lp(a) are not catabolized from the bloodstream as a unit in humans in the fed state.
Collapse
Affiliation(s)
- Margaret R Diffenderfer
- Cardiovascular Nutrition Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, 711 Washington Street, Boston, MA 02111, USA.
| | - Stefania Lamon-Fava
- Cardiovascular Nutrition Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, 711 Washington Street, Boston, MA 02111, USA.
| | - Santica M Marcovina
- Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, 401 Queen Anne Avenue North, Seattle, WA 98109, USA.
| | - P Hugh R Barrett
- School of Medicine and Pharmacology and Faculty of Engineering, Computing and Mathematics, The University of Western Australia, 35 Stirling Highway, Crawley, WA 6009, Australia.
| | - Julian Lel
- Cardiovascular Nutrition Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, 711 Washington Street, Boston, MA 02111, USA.
| | - Gregory G Dolnikowski
- Mass Spectrometry Unit, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, 711 Washington Street, Boston, MA 02111, USA.
| | - Lars Berglund
- Clinical and Translational Science Center, University of California, Davis, 2921 Stockton Boulevard, Suite 1400, Sacramento, CA 95817, USA.
| | - Ernst J Schaefer
- Cardiovascular Nutrition Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, 711 Washington Street, Boston, MA 02111, USA.
| |
Collapse
|
21
|
Kettunen J, Demirkan A, Würtz P, Draisma HHM, Haller T, Rawal R, Vaarhorst A, Kangas AJ, Lyytikäinen LP, Pirinen M, Pool R, Sarin AP, Soininen P, Tukiainen T, Wang Q, Tiainen M, Tynkkynen T, Amin N, Zeller T, Beekman M, Deelen J, van Dijk KW, Esko T, Hottenga JJ, van Leeuwen EM, Lehtimäki T, Mihailov E, Rose RJ, de Craen AJM, Gieger C, Kähönen M, Perola M, Blankenberg S, Savolainen MJ, Verhoeven A, Viikari J, Willemsen G, Boomsma DI, van Duijn CM, Eriksson J, Jula A, Järvelin MR, Kaprio J, Metspalu A, Raitakari O, Salomaa V, Slagboom PE, Waldenberger M, Ripatti S, Ala-Korpela M. Genome-wide study for circulating metabolites identifies 62 loci and reveals novel systemic effects of LPA. Nat Commun 2016; 7:11122. [PMID: 27005778 PMCID: PMC4814583 DOI: 10.1038/ncomms11122] [Citation(s) in RCA: 523] [Impact Index Per Article: 58.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 02/24/2016] [Indexed: 01/20/2023] Open
Abstract
Genome-wide association studies have identified numerous loci linked with complex
diseases, for which the molecular mechanisms remain largely unclear. Comprehensive
molecular profiling of circulating metabolites captures highly heritable traits,
which can help to uncover metabolic pathophysiology underlying established disease
variants. We conduct an extended genome-wide association study of genetic influences
on 123 circulating metabolic traits quantified by nuclear magnetic resonance
metabolomics from up to 24,925 individuals and identify eight novel loci for amino
acids, pyruvate and fatty acids. The LPA locus link with cardiovascular risk
exemplifies how detailed metabolic profiling may inform underlying aetiology via
extensive associations with very-low-density lipoprotein and triglyceride
metabolism. Genetic fine mapping and Mendelian randomization uncover wide-spread
causal effects of lipoprotein(a) on overall lipoprotein metabolism and we assess
potential pleiotropic consequences of genetically elevated lipoprotein(a) on diverse
morbidities via electronic health-care records. Our findings strengthen the argument
for safe LPA-targeted intervention to reduce cardiovascular risk. Circulating metabolites reflect human health and disease. Here,
Kettunen et al. perform a genome-wide association study on 123 circulating
metabolic traits and identify novel genetic loci influencing systemic metabolism. They
also link new molecular pathways with a known cardiovascular risk factor
Lp(a).
Collapse
Affiliation(s)
- Johannes Kettunen
- Computational Medicine, Faculty of Medicine, University of Oulu, PO Box 5000, 90014 Oulu, Finland.,National Institute for Health and Welfare, PO Box 30, FI-00271 Helsinki, Finland.,NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Yliopistonranta 1C, Kuopio 70210, Finland.,Biocenter Oulu, University of Oulu, PO Box 5000, FI-90014 Oulu, Finland
| | - Ayşe Demirkan
- Department of Human Genetics, Leiden University Medical Center, PO Box 9600, 2300 RC Leiden, The Netherlands.,Department of Epidemiology, Erasmus Medical Center, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Peter Würtz
- Computational Medicine, Faculty of Medicine, University of Oulu, PO Box 5000, 90014 Oulu, Finland
| | - Harmen H M Draisma
- Department of Biological Psychology, VU University Amsterdam, Van der Boechorststraat 1, Room 2B-29, 1081 BT Amsterdam, The Netherlands.,EMGO Institute for Health and Care Research, Van der Boechorststraat 7, 1081BT Amsterdam, The Netherlands.,Neuroscience Campus Amsterdam, De Boelelaan 1085, 1081HV Amsterdam, The Netherlands
| | - Toomas Haller
- Estonian Genome Center, University of Tartu, Riia 23b, 51010 Tartu, Estonia
| | - Rajesh Rawal
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany.,Institute of Epidemiology II, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Anika Vaarhorst
- Department of Molecular Epidemiology, Leiden University Medical Center, PO Box 9600, 2300 RC Leiden, The Netherlands
| | - Antti J Kangas
- Computational Medicine, Faculty of Medicine, University of Oulu, PO Box 5000, 90014 Oulu, Finland
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Fimlab Laboratories, University of Tampere School of Medicine, Tampere University, Kalevantie 4, Tampere 33014, Finland
| | - Matti Pirinen
- Institute for Molecular Medicine (FIMM), University of Helsinki, Biomedicum 2, Tukholmankatu 8, Helsinki 00290, Finland
| | - René Pool
- Department of Biological Psychology, VU University Amsterdam, Van der Boechorststraat 1, Room 2B-29, 1081 BT Amsterdam, The Netherlands.,EMGO Institute for Health and Care Research, Van der Boechorststraat 7, 1081BT Amsterdam, The Netherlands
| | - Antti-Pekka Sarin
- National Institute for Health and Welfare, PO Box 30, FI-00271 Helsinki, Finland.,Institute for Molecular Medicine (FIMM), University of Helsinki, Biomedicum 2, Tukholmankatu 8, Helsinki 00290, Finland
| | - Pasi Soininen
- Computational Medicine, Faculty of Medicine, University of Oulu, PO Box 5000, 90014 Oulu, Finland.,NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Yliopistonranta 1C, Kuopio 70210, Finland
| | - Taru Tukiainen
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, 55 Fruit Street, Boston, Massachusetts 02114, USA.,Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, NRB 0330, Boston, Massachusetts 02115, USA
| | - Qin Wang
- Computational Medicine, Faculty of Medicine, University of Oulu, PO Box 5000, 90014 Oulu, Finland.,NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Yliopistonranta 1C, Kuopio 70210, Finland
| | - Mika Tiainen
- Computational Medicine, Faculty of Medicine, University of Oulu, PO Box 5000, 90014 Oulu, Finland.,NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Yliopistonranta 1C, Kuopio 70210, Finland
| | - Tuulia Tynkkynen
- Computational Medicine, Faculty of Medicine, University of Oulu, PO Box 5000, 90014 Oulu, Finland.,NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Yliopistonranta 1C, Kuopio 70210, Finland
| | - Najaf Amin
- Department of Epidemiology, Erasmus Medical Center, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Tanja Zeller
- German Center for Cardiovascular Research (DZHK e.V.), Partner Site Hamburg/Lübeck/Kiel, Martinistraße 52, 20246 Hamburg, Germany.,University Heart Center Hamburg, Clinic of general and interventional Cardiology, Martinistraße 52, 20246 Hamburg, Germany
| | - Marian Beekman
- Department of Molecular Epidemiology, Leiden University Medical Center, PO Box 9600, 2300 RC Leiden, The Netherlands
| | - Joris Deelen
- Department of Molecular Epidemiology, Leiden University Medical Center, PO Box 9600, 2300 RC Leiden, The Netherlands
| | - Ko Willems van Dijk
- Department of Human Genetics, Leiden University Medical Center, PO Box 9600, 2300 RC Leiden, The Netherlands.,Department of Endocrinology, Leiden University Medical Center, PO Box 9600, 2300 RC Leiden, The Netherlands
| | - Tõnu Esko
- Estonian Genome Center, University of Tartu, Riia 23b, 51010 Tartu, Estonia
| | - Jouke-Jan Hottenga
- Department of Biological Psychology, VU University Amsterdam, Van der Boechorststraat 1, Room 2B-29, 1081 BT Amsterdam, The Netherlands.,EMGO Institute for Health and Care Research, Van der Boechorststraat 7, 1081BT Amsterdam, The Netherlands
| | - Elisabeth M van Leeuwen
- Department of Epidemiology, Erasmus Medical Center, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, University of Tampere School of Medicine, Tampere University, Kalevantie 4, Tampere 33014, Finland
| | - Evelin Mihailov
- Estonian Genome Center, University of Tartu, Riia 23b, 51010 Tartu, Estonia
| | - Richard J Rose
- Department of Public Health, Hjelt Institute, University of Helsinki, PO Box 41 Mannerheimintie 172, Helsinki 00014, Finland.,Department of Psychological and Brain Sciences, Indiana University, 1101 E 10th Street, Bloomington, Indiana 47405, USA
| | - Anton J M de Craen
- Department of Geriatrics and Gerontology, Leiden University Medical Center, Postzone C7-Q, PO Box 9600, 2300RC Leiden, The Netherlands
| | - Christian Gieger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany.,Institute of Epidemiology II, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Mika Kähönen
- Department of Clinical Physiology, University of Tampere and Tampere, University Hospital, PO Box 2000, FIN-33521 Tampere, Finland
| | - Markus Perola
- National Institute for Health and Welfare, PO Box 30, FI-00271 Helsinki, Finland.,Estonian Genome Center, University of Tartu, Riia 23b, 51010 Tartu, Estonia.,Institute for Molecular Medicine (FIMM), University of Helsinki, Biomedicum 2, Tukholmankatu 8, Helsinki 00290, Finland
| | - Stefan Blankenberg
- German Center for Cardiovascular Research (DZHK e.V.), Partner Site Hamburg/Lübeck/Kiel, Martinistraße 52, 20246 Hamburg, Germany.,University Heart Center Hamburg, Clinic of general and interventional Cardiology, Martinistraße 52, 20246 Hamburg, Germany
| | - Markku J Savolainen
- Biocenter Oulu, University of Oulu, PO Box 5000, FI-90014 Oulu, Finland.,Medical Research Center, Internal Medicine, Oulu University Hospital, University of Oulu, Aapistie 5A, Oulu FI-90220, Finland
| | - Aswin Verhoeven
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Jorma Viikari
- Department of Medicine, University of Turku and Turku University Hospital, PB 52, 20521 Turku, Finland
| | - Gonneke Willemsen
- Department of Biological Psychology, VU University Amsterdam, Van der Boechorststraat 1, Room 2B-29, 1081 BT Amsterdam, The Netherlands.,EMGO Institute for Health and Care Research, Van der Boechorststraat 7, 1081BT Amsterdam, The Netherlands
| | - Dorret I Boomsma
- Department of Biological Psychology, VU University Amsterdam, Van der Boechorststraat 1, Room 2B-29, 1081 BT Amsterdam, The Netherlands.,EMGO Institute for Health and Care Research, Van der Boechorststraat 7, 1081BT Amsterdam, The Netherlands
| | - Cornelia M van Duijn
- Department of Epidemiology, Erasmus Medical Center, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Johan Eriksson
- National Institute for Health and Welfare, PO Box 30, FI-00271 Helsinki, Finland.,Department of General Practice and Primary Health Care, University of Helsinki, PL 20, Tukholmankatu 8B, Helsinki 00029, Finland.,Folkhälsan Research Centre, Helsingfors Universitet, PB 63, Helsinki 00014, Finland
| | - Antti Jula
- National Institute for Health and Welfare, PO Box 30, FI-00271 Helsinki, Finland
| | - Marjo-Riitta Järvelin
- Biocenter Oulu, University of Oulu, PO Box 5000, FI-90014 Oulu, Finland.,Department of Epidemiology and Biostatistics, MRC-PHE Centre for Environment and Health, School of Public Health, Imperial College London, London SW7 2AZ, UK.,Center for Life Course and Systems Epidemiology, Faculty of Medicine, University of Oulu, PL 5000, 90014 Oulu, Finland.,Unit of Primary Care, Oulu University Hospital, P.O. Box 20, OYS, Oulu 90029, Finland
| | - Jaakko Kaprio
- Institute for Molecular Medicine (FIMM), University of Helsinki, Biomedicum 2, Tukholmankatu 8, Helsinki 00290, Finland.,Department of Public Health, Hjelt Institute, University of Helsinki, PO Box 41 Mannerheimintie 172, Helsinki 00014, Finland.,Department of Mental Health and Substance Abuse Services, National Institute for Health and Welfare, PO Box 30 (Mannerheimintie 166), Helsinki 00300, Finland
| | - Andres Metspalu
- Estonian Genome Center, University of Tartu, Riia 23b, 51010 Tartu, Estonia
| | - Olli Raitakari
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Kiinamyllynkatu 4-8, Turku 20521, Finland.,Department of Clinical Physiology, Turku University Hospital, Kiinamyllynkatu 4-8, Turku 20521, Finland
| | - Veikko Salomaa
- National Institute for Health and Welfare, PO Box 30, FI-00271 Helsinki, Finland
| | - P Eline Slagboom
- Department of Molecular Epidemiology, Leiden University Medical Center, PO Box 9600, 2300 RC Leiden, The Netherlands
| | - Melanie Waldenberger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany.,Institute of Epidemiology II, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Samuli Ripatti
- National Institute for Health and Welfare, PO Box 30, FI-00271 Helsinki, Finland.,Institute for Molecular Medicine (FIMM), University of Helsinki, Biomedicum 2, Tukholmankatu 8, Helsinki 00290, Finland.,Department of Public Health, Hjelt Institute, University of Helsinki, PO Box 41 Mannerheimintie 172, Helsinki 00014, Finland.,Human Genetics, Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton CB10 1SA, UK
| | - Mika Ala-Korpela
- Computational Medicine, Faculty of Medicine, University of Oulu, PO Box 5000, 90014 Oulu, Finland.,NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Yliopistonranta 1C, Kuopio 70210, Finland.,Biocenter Oulu, University of Oulu, PO Box 5000, FI-90014 Oulu, Finland.,Oulu University Hospital, Kajaanintie 50, Oulu 90220, Finland.,Computational Medicine, School of Social and Community Medicine, University of Bristol, Senate House, Tyndall Avenue, Bristol, Bristol BS8 1TH, UK.,Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, Bristol BS8 1TH, UK
| |
Collapse
|
22
|
Tada H, Kawashiri MA, Yoshida T, Teramoto R, Nohara A, Konno T, Inazu A, Mabuchi H, Yamagishi M, Hayashi K. Lipoprotein(a) in Familial Hypercholesterolemia With Proprotein Convertase Subtilisin/Kexin Type 9 (PCSK9) Gain-of-Function Mutations. Circ J 2015; 80:512-8. [PMID: 26632531 DOI: 10.1253/circj.cj-15-0999] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND It has been shown that serum lipoprotein(a) [Lp(a)] is elevated in familial hypercholesterolemia (FH) with mutation(s) of the LDL receptor (LDLR) gene. However, few data exist regarding Lp(a) levels in FH with gain-of-function mutations of the PCSK9 gene. METHODS AND RESULTS We evaluated 42 mutation-determined heterozygous FH patients with aPCSK9gain-of-function mutation (FH-PCSK9, mean age 52, mean LDL-C 235 mg/dl), 198 mutation-determined heterozygous FH patients with aLDLRmutation (FH-LDLR, mean age 44, mean LDL-C 217 mg/dl), and 4,015 controls (CONTROL, mean age 56, mean LDL-C 109 mg/dl). We assessed their Lp(a), total cholesterol, triglycerides, HDL-C, LDL-C, use of statins, presence of hypertension, diabetes, chronic kidney disease, smoking, body mass index (BMI) and coronary artery disease (CAD). Multiple regression analysis showed that HDL-C, use of statins, presence of hypertension, smoking, BMI, and Lp(a) were independently associated with the presence of CAD. Under these conditions, the serum levels of Lp(a) in patients with FH were significantly higher than those of the CONTROL group regardless of their causative genes, among the groups propensity score-matched (median Lp(a) 12.6 mg/dl [IQR:9.4-33.9], 21.1 mg/dl [IQR:11.7-34.9], and 5.0 mg/dl [IQR:2.7-8.1] in the FH-LDLR, FH-PCSK9, and CONTROL groups, respectively, P=0.002 for FH-LDLR vs. CONTROL, P=0.002 for FH-PCSK9 vs. CONTROL). CONCLUSIONS These data demonstrate that serum Lp(a) is elevated in patients with FH caused by PCSK9 gain-of-function mutations to the same level as that in FH caused by LDLR mutations.
Collapse
Affiliation(s)
- Hayato Tada
- Division of Cardiovascular Medicine, Kanazawa University Graduate School of Medicine
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Abstract
Cardiovascular disease (CVD) is the most common cause of death and disability worldwide. Therefore, great importance has been placed on the discovery of novel risk factors and metabolic pathways relevant in the prevention and management of CVD. Such research is ongoing and may continue to lead to better risk stratification of individuals and/or the development of new intervention targets and treatment options. This review highlights emerging biomarkers related to lipid metabolism, glycemia, inflammation, and cardiac damage, some of which show promising associations with CVD risk and provide further understanding of the underlying pathophysiology. However, their measurement methodology and assays will require validation and standardization, and it will take time to accumulate evidence of their role in CVD in various population settings in order to fully assess their clinical utility. Several of the novel biomarkers represent intriguing, potentially game-changing targets for therapy.
Collapse
Affiliation(s)
- Leah E Cahill
- Department of Medicine, Dalhousie University, 5790 University Ave, Halifax, NS, B3H 1V7, Canada.
- Department of Nutrition, Harvard T. H. Chan School of Public Health, 665 Huntington Avenue, Boston, MA, 02115, USA.
| | - Monica L Bertoia
- Department of Nutrition, Harvard T. H. Chan School of Public Health, 665 Huntington Avenue, Boston, MA, 02115, USA.
| | - Sarah A Aroner
- Department of Nutrition, Harvard T. H. Chan School of Public Health, 665 Huntington Avenue, Boston, MA, 02115, USA.
| | - Kenneth J Mukamal
- Beth Israel Deaconess Medical Center, 1309 Beacon Street, 2nd Floor, Brookline, Boston, MA, USA.
| | - Majken K Jensen
- Department of Nutrition, Harvard T. H. Chan School of Public Health, 665 Huntington Avenue, Boston, MA, 02115, USA.
| |
Collapse
|
24
|
Lipoprotein (a) as a risk factor for ischemic stroke: a meta-analysis. Atherosclerosis 2015; 242:496-503. [PMID: 26298741 DOI: 10.1016/j.atherosclerosis.2015.08.021] [Citation(s) in RCA: 134] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 07/11/2015] [Accepted: 08/13/2015] [Indexed: 01/01/2023]
Abstract
OBJECTIVE Lipoprotein (a) [Lp(a)] harbors atherogenic potential but its role as a risk factor for ischemic stroke remains controversial. We conducted a meta-analysis to determine the relative strength of the association between Lp(a) and ischemic stroke and identify potential subgroup-specific risk differences. METHODS A systematic search using the MeSH terms "lipoproteins" OR "lipoprotein a" AND "stroke" was performed in PubMed and ScienceDirect for case-control studies from June 2006 and prospective cohort studies from April 2009 until December 20th 2014. Data from eligible papers published before these dates were reviewed and extracted from previous meta-analyses. Studies that assessed the relationship between Lp(a) levels and ischemic stroke and reported generic data-i.e. odds ratio [OR], hazard ratio, or risk ratio [RR]-were eligible for inclusion. Studies that not distinguish between ischemic and hemorrhagic stroke and transient ischemic attack were excluded. Random effects meta-analyses with mixed-effects meta-regression were performed by pooling adjusted OR or RR. RESULTS A total of 20 articles comprising 90,904 subjects and 5029 stroke events were eligible for the meta-analysis. Comparing high with low Lp(a) levels, the pooled estimated OR was 1.41 (95% CI, 1.26-1.57) for case-control studies (n = 11) and the pooled estimated RR was 1.29 (95% CI, 1.06-1.58) for prospective studies (n = 9). Sex-specific differences in RR were inconsistent between case-control and prospective studies. Study populations with a mean age of ≤55 years had an increased RR compared to older study populations. Reported Lp(a) contrast levels and ischemic stroke subtype significantly contributed to the heterogeneity observed in the analyses. CONCLUSION Elevated Lp(a) is an independent risk factor for ischemic stroke and may be especially relevant for young stroke patients. Sex-specific risk differences remain conflicting. Further studies in these subgroups may be warranted.
Collapse
|
25
|
Manocha A, Srivastava LM. Lipoprotein (a): a Unique Independent Risk Factor for Coronary Artery Disease. Indian J Clin Biochem 2015; 31:13-20. [PMID: 26855483 DOI: 10.1007/s12291-015-0483-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2015] [Accepted: 02/06/2015] [Indexed: 12/22/2022]
Abstract
The current epidemic affecting Indians is coronary artery disease (CAD), and is currently one of the most common causes of mortality and morbidity in developed and developing countries. The higher rate of CAD in Indians, as compared to people of other ethnic origin, may indicate a possible genetic susceptibility. Hence, Lp(a), an independent genetic risk marker for atherosclerosis and cardiovascular disease assumes great importance. Lp(a), an atherogenic lipoprotein, contains a cholesterol rich LDL particle, one molecule of apolipoprotein B-100 and a unique protein, apolipoprotein (a) which distinguishes it from LDL. Apo(a) is highly polymorphic and an inverse relationship between Lp(a) concentration and apo(a) isoform size has been observed. This is genetically controlled suggesting a functional diversity among the apo(a) isoforms. The LPA gene codes for apo(a) whose genetic heterogeneity is due to variations in its number of kringles. The exact pathogenic mechanism of Lp(a) is still not completely elucidated, but the structural homology of Lp(a) with LDL and plasmin is possibly responsible for its acting as a link between atherosclerosis and thrombosis. Upper limits of normal Lp(a) levels have not been defined for the Indian population. A cut off limit of 20 mg/dL has been suggested while for the Caucasian population it is 30 mg/dL. Though a variety of assays are available for its measurement, standardization of the analytical method is highly complicated as a majority of the methods are affected by the heterogeneity in apo(a) size. No therapeutic drug selectively targets Lp(a) but recently, new modifiers of apo(a) synthesis are being considered.
Collapse
Affiliation(s)
- Anjali Manocha
- Department of Biochemistry, Sir Ganga Ram Hospital, Rajender Nagar, New Delhi, 110060 India
| | - L M Srivastava
- Department of Biochemistry, Sir Ganga Ram Hospital, Rajender Nagar, New Delhi, 110060 India
| |
Collapse
|
26
|
|
27
|
Jensen MK, Bertoia ML, Cahill LE, Agarwal I, Rimm EB, Mukamal KJ. Novel metabolic biomarkers of cardiovascular disease. Nat Rev Endocrinol 2014; 10:659-72. [PMID: 25178732 DOI: 10.1038/nrendo.2014.155] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Coronary heart disease (CHD) accounts for one in every six deaths in US individuals. Great advances have been made in identifying important risk factors for CHD, such as hypertension, diabetes mellitus, smoking and hypercholesterolaemia, which have led to major developments in therapy. In particular, statins represent one of the greatest successes in the prevention of CHD. While these standard risk factors are important, an obvious opportunity exists to take advantage of ongoing scientific research to better risk-stratify individuals and to identify new treatment targets. In this Review, we summarize ongoing scientific research in a number of metabolic molecules or features, including lipoproteins, homocysteine, calcium metabolism and glycaemic markers. We evaluate the current state of the research and the strength of evidence supporting each emerging biomarker. We also discuss whether the associations with CHD are strong and consistent enough to improve current risk stratification metrics, and whether these markers enhance our understanding of the underlying biology of CHD and thus point towards new treatment options.
Collapse
Affiliation(s)
- Majken K Jensen
- Department of Nutrition, Harvard School of Public Health, 665 Huntington Avenue, 02115 Boston, MA, USA
| | - Monica L Bertoia
- Department of Nutrition, Harvard School of Public Health, 665 Huntington Avenue, 02115 Boston, MA, USA
| | - Leah E Cahill
- Department of Nutrition, Harvard School of Public Health, 665 Huntington Avenue, 02115 Boston, MA, USA
| | - Isha Agarwal
- Department of Nutrition, Harvard School of Public Health, 665 Huntington Avenue, 02115 Boston, MA, USA
| | - Eric B Rimm
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, 181 Longwood Avenue, 02115 Boston, MA, USA
| | - Kenneth J Mukamal
- Department of Medicine, Beth Israel Deaconess Medical Centre, 1309 Beacon Street, 02446 Brookline, MA, USA
| |
Collapse
|
28
|
Xue Y, Iqbal N, Chan J, Maisel A. Biomarkers in Hypertension and Their Relationship with Myocardial Target-Organ Damage. Curr Hypertens Rep 2014; 16:502. [DOI: 10.1007/s11906-014-0502-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
29
|
New therapies targeting apoB metabolism for high-risk patients with inherited dyslipidaemias: what can the clinician expect? Cardiovasc Drugs Ther 2014; 27:559-67. [PMID: 23913122 DOI: 10.1007/s10557-013-6479-4] [Citation(s) in RCA: 134] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Apolipoprotein B (apoB) has a key role in the assembly and secretion of very low-density lipoprotein (VLDL) from the liver. Plasma apoB concentration affects the number of circulating atherogenic particles, and serves as an independent predictor of the risk of atherosclerotic cardiovascular disease. While statins are the most potent apoB-lowering agents currently prescribed, their efficacy in achieving therapeutic targets for low-density lipoprotein cholesterol (LDL-C) in high-risk patients, such as those with familial hypercholesterolaemia (FH), is limited. Resistance and intolerance to statins also occurs in a significant number of patients, necessitating new types of lipid-lowering therapies. Monoclonal antibodies against proprotein convertase subtilisin/kexin type 9 (PCSK9; AMG 145 and REGN727), a sequence-specific antisense oligonucleotide against apoB mRNA (mipomersen) and a synthetic inhibitor of microsomal triglyceride transfer protein (MTTP; lomitapide) have been tested in phase III clinical trials, particularly in patients with FH. The trials demonstrated the efficacy of these agents in lowering apoB, LDL-C, non-high-density lipoprotein cholesterol and lipoprotein(a) by 32-55 %, 37-66 %, 38-61 % and 22-50 % (AMG 145), 21-68 %, 29-72 %, 16-60 % and 8-36 % (REGN727), 16-71 %, 15-71 %, 12-66 % and 23-49 % (mipomersen) and 24-55 %, 25-51 %, 27-50 % and 15-19 % (lomitapide), respectively. Monoclonal antibodies against PCSK9 have an excellent safety profile and may be indicated not only in heterozygous FH, but also in statin-intolerant patients and those with other inherited dyslipidemias, such as familial combined hyperlipidaemia and familial elevation in Lp(a). Mipomersen and lomitapide increase hepatic fat content and are at present indicated for treating adult patients with homozygous FH alone.
Collapse
|
30
|
Yusuf J, Yadav N, Mukhopadhyay S, Goyal A, Mehta V, Trehan V, Tyagi S. Relook at lipoprotein (A): independent risk factor of coronary artery disease in north Indian population. Indian Heart J 2014; 66:272-9. [PMID: 24973831 DOI: 10.1016/j.ihj.2014.03.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Revised: 12/28/2013] [Accepted: 03/23/2014] [Indexed: 02/03/2023] Open
Abstract
AIMS Lipoprotein (a) [Lp(a)] levels have shown wide ethnic variations. Sparse data on mean Lp(a) levels, its link with clinical variables and severity of coronary artery disease (CAD) in North Indian population needed further studies. METHODS 150 patients, each of single vessel disease (SVD), double vessel disease (DVD) and triple vessel disease (TVD) with 150 healthy controls were drawn for the study. Serum Lp(a) estimation was performed by immunoturbidimetric method. RESULTS Lp(a) had a skewed distribution. Median Lp(a) level was significantly raised in cases as compared to controls (median 30.30 vs. 20 mg/dl, p < 0.001). Cases with acute coronary syndrome (ACS, 55.8%) had significantly higher median Lp(a) levels as compared to those with chronic stable angina (35.4 mg/dl vs. 23 mg/dl, p < 0.001). Significant difference in median Lp(a) levels were observed in patients with DVD or TVD versus control (30, 39.05 vs 20 mg/dl, p < 0.008). Lp(a) level was found to be an independent risk factor for CAD (AOR{adjusted odds ratio} 1.018, 95% CI 1.010-1.027; p < 0.001). Analysis using Lp(a) as categorical variable showed that progressive increase in Lp(a) concentration was associated with increased risk of CAD [AOR from lowest to highest quartile (1, 1.04, 1.43 and 2.65, p value for trend = 0.00026)]. Multivariably AOR of CAD for subjects with Lp(a) in the highest quartile (above 40 mg/dl) compared to those with Lp(a) ≤40 mg/dl was 2.308 (95% CI 1.465-3.636, p < 0.001). CONCLUSION Lp(a) above 40 mg/dl (corresponding to 75th percentile)assessed by an isoform insensitive assay is an independent risk factor for CAD. Raised Lp(a) level is also associated with increased risk of ACS and multivessel CAD.
Collapse
Affiliation(s)
- Jamal Yusuf
- Professor, Department of Cardiology, G.B. Pant Hospital, New Delhi, India
| | - Neeraj Yadav
- Consultant Cardiologist, Sterling Hospital, Ahmedabad, India
| | | | - Abhishek Goyal
- Assistant Professor, Dayanand Medical College, Ludhiana, India.
| | - Vimal Mehta
- Professor, Department of Cardiology, G.B. Pant Hospital, New Delhi, India
| | - Vijay Trehan
- Professor, Department of Cardiology, G.B. Pant Hospital, New Delhi, India
| | - Sanjay Tyagi
- Professor & Head, Department of Cardiology, G.B. Pant Hospital, New Delhi, India
| |
Collapse
|
31
|
The association between the LPA gene polymorphism and coronary artery disease in Chinese Han population. BIOMED RESEARCH INTERNATIONAL 2014; 2014:370670. [PMID: 24790998 PMCID: PMC3984839 DOI: 10.1155/2014/370670] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 02/26/2014] [Indexed: 01/13/2023]
Abstract
Lp(a) has been well known as an independent risk factor for coronary artery disease (CAD). The LPA gene, as it encodes apo(a) of the Lp(a) lipoprotein particle, was associated with increased risk of CAD. The purpose of this study was to analyze the relationship between the polymorphisms of LPA gene and CAD in Chinese Han population. Five SNPs (rs1367211, rs3127596, rs6415085, rs9347438, and rs9364559) in the LPA gene were genotyped using Sequenom MassARRAY time-of-flight mass spectrometer (TOF) in 560 CAD patients as case group and 531 non-CAD subjects as control group. The numbers of these two groups were from Chinese Han ancestry. The results showed that allele (P = 0.046) and genotype (P = 0.026) of rs9364559 in the LPA gene was associated with CAD. The frequency of rs9364559 minor allele (G) in case group was obviously higher than that in control group. Results of haplotype analysis showed that 4 haplotypes which contained rs9364559-G were associated with increased risk of CAD in this population. This study explored rs9364559 in the LPA gene may be associated with the pathogenesis of CAD; and the risk of CAD might be higher in the population carrying 4 haplotypes of different blocks in the LPA gene.
Collapse
|
32
|
Rogenhofer N, Engels L, Bogdanova N, Tüttelmann F, Thaler CJ, Markoff A. Lower Incidence of M2/ANXA5 Carriage in Recurrent Pregnancy Loss Patients With Elevated Lipoprotein(a) Levels. Clin Appl Thromb Hemost 2013; 20:706-9. [DOI: 10.1177/1076029613516189] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
This study compared the incidence of M2/ANXA5 haplotype carriage, a documented repeated miscarriage risk factor, in patient groups with normal and elevated lipoprotein(a) (Lp(a)) levels. A total of 138 women with ≥2 consecutive, idiopathic recurrent miscarriages, categorized in patients with elevated (≥30 mg/dL, n = 44) and normal Lp(a) level (<30 mg/dL, n = 94) were recruited at the recurrent pregnancy loss (RPL) clinic of Munich Großhadern University Hospital. A total of 500 fertile women served as controls. All patients were genotyped for ANXA5 promoter haplotypes, genetic frequencies were compared, and odds ratios (ORs) and relative risks of M2 carriers were calculated. Women with M2 haplotype had an almost 2 times higher relative risk of RPL (OR 2.6, 95% confidence interval 1.5-4.6, P = .001) than fertile controls. Furthermore, risk rises to 2.47 in patients having normal Lp(a) levels (OR 3.2, 95% confidence interval 1.7-5.9, P = .001), whereas women with high Lp(a) levels exhibit notably lower apparent RPL risk of 1.39 (OR 1.4, 95% confidence interval 0.5-4.1, P = .659).
Collapse
Affiliation(s)
- Nina Rogenhofer
- Department of Gynecology and Obstetrics, Division of Gynecological Endocrinology and Reproductive Medicine, Klinikum der Ludwig-Maximilians-University, Munich, Germany
| | - Laura Engels
- Department of Gynecology and Obstetrics, Division of Gynecological Endocrinology and Reproductive Medicine, Klinikum der Ludwig-Maximilians-University, Munich, Germany
| | - Nadja Bogdanova
- Insititute of Human Genetics, UKM and WWU Muenster, Muenster, Germany
| | - Frank Tüttelmann
- Insititute of Human Genetics, UKM and WWU Muenster, Muenster, Germany
| | - Christian J Thaler
- Department of Gynecology and Obstetrics, Division of Gynecological Endocrinology and Reproductive Medicine, Klinikum der Ludwig-Maximilians-University, Munich, Germany
| | - Arseni Markoff
- Institute of Medical Biochemistry, ZMBE and IZKF, WWU Muenster, Muenster, Germany
| |
Collapse
|
33
|
Lipoprotein(a): a promising marker for residual cardiovascular risk assessment. DISEASE MARKERS 2013; 35:551-9. [PMID: 24249942 PMCID: PMC3819768 DOI: 10.1155/2013/563717] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Revised: 09/03/2013] [Accepted: 09/04/2013] [Indexed: 01/14/2023]
Abstract
Atherosclerotic cardiovascular diseases (CVD) are still the leading cause of morbidity and mortality worldwide, although optimal medical therapy has been prescribed for primary and secondary preventions. Residual cardiovascular risk for some population groups is still considerably high although target low density lipoprotein-cholesterol (LDL-C) level has been achieved. During the past few decades, compelling pieces of evidence from clinical trials and meta-analyses consistently illustrate that lipoprotein(a) (Lp(a)) is a significant risk factor for atherosclerosis and CVD due to its proatherogenic and prothrombotic features. However, the lack of effective medication for Lp(a) reduction significantly hampers randomized, prospective, and controlled trials conducting. Based on previous findings, for patients with LDL-C in normal range, Lp(a) may be a useful marker for identifying and evaluating the residual cardiovascular risk, and aggressively lowering LDL-C level than current guidelines' recommendation may be reasonable for patients with particularly high Lp(a) level.
Collapse
|
34
|
Abstract
Recent epidemiologic and Mendelian randomization studies together have provided evidence that lipoprotein(a) (Lp(a)) plays a causal role in the pathogenesis of atherosclerosis and cardiovascular disease (CVD). The risk association with CVD is weak but seems continuous in shape and without an obvious threshold for Lp(a) levels. A plasma concentration of 60 mg/dl compared to usual levels is associated with an odds ratio for coronary heart disease of about 1.5 after adjustment for other cardiovascular risk factors. Niacin (nicotinic acid) is the pharmacologic means of choice for decreasing elevated Lp(a) levels but the drug is often poorly tolerated due to adverse reactions. Dietary measures, exercise and other lipid-lowering drugs, especially statins, fibrates and ezetimibe, are without effect. In patients with severe progressive cardiovascular disease and very high Lp(a) levels, lipoprotein apheresis may be used to effectively decrease Lp(a) concentrations. The method is expensive and impractical for most patients and its feasibility depends by and large on the healthcare reimbursement system of the respective country. No established treatment, however, selectively reduces Lp(a) without influencing other lipoproteins. Moreover, despite the clear association of hyperlipoproteinemia(a) with cardiovascular risk, no rigorously designed study to date has demonstrated that lowering Lp(a) concentrations has beneficial effects on cardiovascular endpoints. Randomized trials to this effect are urgently needed.
Collapse
Affiliation(s)
- Heiner K Berthold
- Charité University Medicine Berlin, Evangelical Geriatrics Center Berlin, Berlin, Germany.
| | | |
Collapse
|
35
|
Abstract
Plasma lipoprotein(a) [Lp(a)] is a quantitative genetic trait with a very broad and skewed distribution, which is largely controlled by genetic variants at the LPA locus on chromosome 6q27. Based on genetic evidence provided by studies conducted over the last two decades, Lp(a) is currently considered to be the strongest genetic risk factor for coronary heart disease (CHD). The copy number variation of kringle IV in the LPA gene has been strongly associated with both Lp(a) levels in plasma and risk of CHD, thereby fulfilling the main criterion for causality in a Mendelian randomization approach. Alleles with a low kringle IV copy number that together have a population frequency of 25-35% are associated with a doubling of the relative risk for outcomes, which is exceptional in the field of complex genetic phenotypes. The recently identified binding of oxidized phospholipids to Lp(a) is considered as one of the possible mechanisms that may explain the pathogenicity of Lp(a). Drugs that have been shown to lower Lp(a) have pleiotropic effects on other CHD risk factors, and an improvement of cardiovascular endpoints is up to now lacking. However, it has been established in a proof of principle study that lowering of very high Lp(a) by apheresis in high-risk patients with already maximally reduced low-density lipoprotein cholesterol levels can dramatically reduce major coronary events.
Collapse
Affiliation(s)
- F Kronenberg
- Division of Genetic Epidemiology, Innsbruck Medical University, Innsbruck, Austria
| | | |
Collapse
|
36
|
Tsimikas S, Hall JL. Lipoprotein(a) as a Potential Causal Genetic Risk Factor of Cardiovascular Disease. J Am Coll Cardiol 2012; 60:716-21. [DOI: 10.1016/j.jacc.2012.04.038] [Citation(s) in RCA: 161] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2012] [Revised: 04/12/2012] [Accepted: 04/16/2012] [Indexed: 11/27/2022]
|
37
|
Significant reduction of elevated serum lipoprotein(a) concentrations during levo-thyroxine–replacement therapy in a hypothyroid patient. J Clin Lipidol 2012; 6:388-91. [DOI: 10.1016/j.jacl.2012.01.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Revised: 01/16/2012] [Accepted: 01/20/2012] [Indexed: 11/20/2022]
|
38
|
Fazio S, Linton MF. Inhibition of Apolipoprotein(a) Synthesis by Farnesoid X Receptor and Fibroblast Growth Factor 15/19. Arterioscler Thromb Vasc Biol 2012; 32:1060-2. [DOI: 10.1161/atvbaha.112.245571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Sergio Fazio
- From the Division of Cardiovascular Medicine, Atherosclerosis Research Unit, Vanderbilt University Medical Center, Nashville, TN
| | - MacRae F. Linton
- From the Division of Cardiovascular Medicine, Atherosclerosis Research Unit, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
39
|
Chennamsetty I, Claudel T, Kostner KM, Trauner M, Kostner GM. FGF19 signaling cascade suppresses APOA gene expression. Arterioscler Thromb Vasc Biol 2012; 32:1220-7. [PMID: 22267484 DOI: 10.1161/atvbaha.111.243055] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Lipoprotein(a) is a highly atherogenic lipoprotein, whose metabolism is poorly understood. Currently no safe drugs exists that lower elevated plasma lipoprotein(a) concentrations. We therefore focused on molecular mechanisms that influence apolipoprotein(a) (APOA) biosynthesis. METHODS AND RESULTS Transgenic human APOA mice (tg-APO mice) were injected with 1 mg/kg of recombinant human fibroblast growth factor 19 (FGF19). This led to a significant reduction of plasma APOA and hepatic expression of APOA. Incubation of primary hepatocytes of tg-APOA mice with FGF19 induced ERK1/2 phosphorylation and, in turn, downregulated APOA expression. Repression of APOA by FGF19 was abrogated by specific ERK1/2 phosphorylation inhibitors. The FGF19 effect on APOA was attenuated by transfection of primary hepatocytes with siRNA against the FGF19 receptor 4 (FGFR4). Using promoter reporter assays, mutation analysis, gel shift, and chromatin immune-precipitation assays, an Ets-1 binding element was identified at -1630/-1615bp region in the human APOA promoter. This element functions as an Elk-1 binding site that mediates repression of APOA transcription by FGF19. CONCLUSIONS These findings provide mechanistic insights into the transcriptional regulation of human APOA by FGF19. Further studies in the human system are required to substantiate our findings and to design therapeutics for hyper lipoprotein(a).
Collapse
Affiliation(s)
- Indumathi Chennamsetty
- Institute of Molecular Biology and Biochemistry, Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Austria
| | | | | | | | | |
Collapse
|
40
|
Lankin VZ, Afanasieva OI, Konovalova GG, Utkina EA, Dmitrieva OA, Tikhaze AK, Kumskova EM, Pokrovsky SN. Modification of lipoprotein(a) by natural dicarbonyls induced their following free radical peroxidation. DOKL BIOCHEM BIOPHYS 2012; 441:287-9. [PMID: 22218957 DOI: 10.1134/s1607672911060159] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2011] [Indexed: 11/23/2022]
Affiliation(s)
- V Z Lankin
- Russian Cardiology Research and Production Association, Ministry of Public Health of the Russian Federation, 3-ya Cherepkovskaya ul. 15a, Moscow, 121552, Russia
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Konerman M, Kulkarni K, Toth PP, Jones SR. Evidence of dependence of lipoprotein(a) on triglyceride and high-density lipoprotein metabolism. J Clin Lipidol 2012; 6:27-32. [DOI: 10.1016/j.jacl.2011.08.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2011] [Revised: 07/22/2011] [Accepted: 08/26/2011] [Indexed: 10/17/2022]
|
42
|
Qi Q, Workalemahu T, Zhang C, Hu FB, Qi L. Genetic variants, plasma lipoprotein(a) levels, and risk of cardiovascular morbidity and mortality among two prospective cohorts of type 2 diabetes. Eur Heart J 2011; 33:325-34. [PMID: 21900290 DOI: 10.1093/eurheartj/ehr350] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
AIMS To examine the relations between genetic loci, plasma lipoprotein(a) [Lp(a)] levels, and cardiovascular disease (CVD) risk among diabetic patients and compare with the observations in the general population. METHODS AND RESULTS In two prospective cohorts of patients with type 2 diabetes (n= 2308) from the Nurses' Health Study and the Health Professional Follow-Up Study, we performed (i) genome-wide association (GWA) scans for plasma Lp(a); (ii) prospective analysis of plasma Lp(a) for CVD risk and mortality; and (iii) genetic association analysis for CVD risk and mortality. Meta-analysis of the two GWA scans yielded 71 single-nucleotide polymorphisms (SNPs) on chromosome 6q associated with plasma Lp(a) levels at a genome-wide significance level (P< 5 × 10(-8)). The SNP rs10455872 in LPA was most strongly associated with Lp(a) (P= 4.60 × 10(-39)). Forward-selection analysis indicated that rs10455872 and other five SNPs in a region encompassing LPA, PLG, SLC22A3, and LPAL2 genes were independently associated with Lp(a) levels and jointly explained ∼20% of variation in diabetic patients. In prospective analysis, we did not find any significant association between plasma levels and CVD incidence; the relative risk for coronary heart disease (CHD), CVD, and CVD death was 1.05 [95% confidence interval (CI): 0.95-1.15], 1.05 (0.96-1.15), and 1.21 (0.99-1.47) per 1-SD higher log-transformed Lp(a) levels, respectively. Consistently, none of the Lp(a) SNPs were associated with CVD risk or mortality (all P> 0.09). For the best SNP rs10455872 for plasma Lp(a) levels, the OR for CHD, CVD, and CVD death was 0.94 (95% CI: 0.69-1.28), 0.97 (0.72-1.29), and 1.23 (0.79-1.92), respectively. The genetic effect on CHD risk showed a significant heterogeneity between the diabetic and the general populations (P= 0.006). CONCLUSION Our data indicate that the effect of Lp(a) on CVD risk among diabetic patients might be different from that in the general population. Diabetes status may attenuate the relation between Lp(a) and cardiovascular risk.
Collapse
Affiliation(s)
- Qibin Qi
- Department of Nutrition, Harvard School of Public Health, 665 Huntington Ave., Boston, MA 02115, USA
| | | | | | | | | |
Collapse
|
43
|
Chennamsetty I, Claudel T, Kostner KM, Baghdasaryan A, Kratky D, Levak-Frank S, Frank S, Gonzalez FJ, Trauner M, Kostner GM. Farnesoid X receptor represses hepatic human APOA gene expression. J Clin Invest 2011; 121:3724-34. [PMID: 21804189 DOI: 10.1172/jci45277] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2011] [Accepted: 06/01/2011] [Indexed: 12/31/2022] Open
Abstract
High plasma concentrations of lipoprotein(a) [Lp(a), which is encoded by the APOA gene] increase an individual's risk of developing diseases, such as coronary artery diseases, restenosis, and stroke. Unfortunately, increased Lp(a) levels are minimally influenced by dietary changes or drug treatment. Further, the development of Lp(a)-specific medications has been hampered by limited knowledge of Lp(a) metabolism. In this study, we identified patients suffering from biliary obstructions with very low plasma Lp(a) concentrations that rise substantially after surgical intervention. Consistent with this, common bile duct ligation in mice transgenic for human APOA (tg-APOA mice) lowered plasma concentrations and hepatic expression of APOA. To test whether farnesoid X receptor (FXR), which is activated by bile acids, was responsible for the low plasma Lp(a) levels in cholestatic patients and mice, we treated tg-APOA and tg-APOA/Fxr-/- mice with cholic acid. FXR activation markedly reduced plasma concentrations and hepatic expression of human APOA in tg-APOA mice but not in tg-APOA/Fxr-/- mice. Incubation of primary hepatocytes from tg-APOA mice with bile acids dose dependently downregulated APOA expression. Further analysis determined that the direct repeat 1 element between nucleotides -826 and -814 of the APOA promoter functioned as a negative FXR response element. This motif is also bound by hepatocyte nuclear factor 4α (HNF4α), which promotes APOA transcription, and FXR was shown to compete with HNF4α for binding to this motif. These findings may have important implications in the development of Lp(a)-lowering medications.
Collapse
Affiliation(s)
- Indumathi Chennamsetty
- Institute of Molecular Biology and Biochemistry, Center of Molecular Medicine, Medical University of Graz, Graz, Austria
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Guardamagna O, Abello F, Anfossi G, Pirro M. Lipoprotein(a) and family history of cardiovascular disease in children with familial dyslipidemias. J Pediatr 2011; 159:314-9. [PMID: 21392785 DOI: 10.1016/j.jpeds.2011.01.038] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2010] [Revised: 11/30/2010] [Accepted: 01/19/2011] [Indexed: 10/18/2022]
Abstract
OBJECTIVE To investigate in children and adolescents with familial dyslipidemias the association between lipoprotein(a) [Lp(a)] level and family history of cardiovascular disease (CVD), and whether this association is independent of the disturbed lipid profile. STUDY DESIGN Lp(a) level, lipid profile, and a 2-generation genealogic tree to detect cardiovascular events were evaluated in 231 patients with familial dyslipidemias. Lp(a) levels were stratified according to presence, age of occurrence, and number and type of cardiovascular events in the patient's kindreds. RESULTS Lp(a) and other plasma lipid fractions did not differ between patients with and those without a family history of cardiovascular events. However, the percentage of patients with elevated Lp(a) level (≥85th percentile) was higher in those with a positive family history for early cardiovascular events (P = .01). Lp(a) level was a significant independent predictor of the number of premature cardiovascular events (β = 0.17; P = .01) and of cerebrovascular events in kindreds (OR, 2.5; 95% CI, 1.05-6.03; P = .039), independent of plasma lipid fractions and other cardiovascular risk factors. CONCLUSIONS In children and adolescents with familial dyslipidemias, the overall association between Lp(a) level and family history of early CVD may be due to a threshold effect in those with the highest Lp(a) levels. However, multiple cardiovascular events and cerebrovascular events are predicted by any increase in plasma Lp(a) level, independent of other cardiovascular risk factors.
Collapse
|
45
|
Song P, Seok JM, Kim WS, Lee YT, Kim DK, Kim GM. Increased lipoprotein(a) is associated with polyvascular disease in patients undergoing coronary artery bypass graft. Atherosclerosis 2011; 219:285-90. [PMID: 21708382 DOI: 10.1016/j.atherosclerosis.2011.05.039] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2011] [Revised: 05/16/2011] [Accepted: 05/31/2011] [Indexed: 11/29/2022]
Abstract
OBJECTIVE We sought to identify clinical and biochemical predictors of disease in multiple vascular territories, in patients with established coronary heart disease. METHODS A total of 470 patients (329 men, 141 female) who had undergone coronary artery bypass grafting (CABG) were enrolled in this prospective study. Polyvascular disease was defined on the presence of existing symptomatic or asymptomatic carotid artery stenosis and/or peripheral artery disease, which is present in 32.1% of patients (n=151). RESULTS Clinical and laboratory features independently associated with the presence of polyvascular disease included age ≥65 years, male sex, hypertension, former or current smoker, low BMI, and high Lp(a). Lp(a) was the only biochemical marker that had an independent association with polyvascular disease (OR=1.01 per 1mg/dl increase; 95% CI, 1.00-1.01). The fourth quartile of Lp(a) has significant associations with the risk of two or more vascular territories involvement (OR=1.866; 95% CI, 1.056-3.297), and three vascular territories involvement (OR=4.240; 95% CI, 1.405-12.798). There was a significant trend towards patients with the highest quartile of Lp(a) that has association with more advanced polyvascular disease (test for trend: p=0.008 for involvement of three vascular territories). CONCLUSION High Lp(a) was independently associated with polyvascular disease in patients who undergo CABG, which is suggestive of an indirect evidence of the pathophysiologic function of Lp(a) in polyvascular disease.
Collapse
Affiliation(s)
- Pamela Song
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | | | | | | | | | | |
Collapse
|
46
|
Antisense oligonucleotide lowers plasma levels of apolipoprotein (a) and lipoprotein (a) in transgenic mice. J Am Coll Cardiol 2011; 57:1611-21. [PMID: 21474042 DOI: 10.1016/j.jacc.2010.10.052] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2010] [Revised: 10/13/2010] [Accepted: 10/18/2010] [Indexed: 12/21/2022]
Abstract
OBJECTIVES This study sought to assess whether an antisense oligonucleotide (ASO) directed to apolipoprotein (a) [apo(a)] reduces apo(a) and lipoprotein (a) [Lp(a)] levels in transgenic mouse models. BACKGROUND Elevated Lp(a) is a causal, independent, genetic risk factor for cardiovascular disease and myocardial infarction. Effective therapies to specifically lower plasma Lp(a) levels are lacking. METHODS Three transgenic mouse models were utilized: 8K-apo(a) mice expressing 8 kringle IV (KIV) repeats with a single copy of KIV-2; 8K-Lp(a) mice expressing both the 8K apo(a) plus human apolipoprotein B-100; and 12K-apo(a) mice expressing a 12K apo(a) with 3 KIV-2 repeats. The mice were treated intraperitoneally with saline, a control ASO, or ASO 144367 directed to KIV-2 for 4 to 6 weeks. Apo(a), Lp(a), and oxidized phospholipids present on human apoB (OxPL/h-apoB) or apo(a) [OxPL/apo(a)] were measured at baseline and on and off therapy. RESULTS ASO 144367 significantly reduced Lp(a) by 24.8% in 8K-Lp(a) mice, and reduced apo(a) levels by 19.2% in 8K-Lp(a) mice, 30.0% in 8K-apo(a) mice, and 86% in 12K-apo(a) mice; ASO 144367 also significantly reduced OxPL/apoB 22.4% in 8K-Lp(a) mice, and OxPL/apo(a) levels by 19.9% in 8K-Lp(a) mice, 22.1% in 8K-apo(a) mice, and 92.5% in 12K-apo(a) mice (p < 0.004, or less, for all). No significant changes occurred in Lp(a), apo(a), OxPL/apoB, or OxPL/apo(a) levels with control ASO or saline. CONCLUSIONS This study documents the first specific therapy, to our knowledge, for lowering apo(a)/Lp(a) levels and their associated OxPL. A more potent effect was documented in mice expressing apo(a) with multiple KIV-2 repeats. Targeting liver expression of apo(a) with ASOs directed to KIV-2 repeats may provide an effective approach to lower elevated Lp(a) levels in humans.
Collapse
|
47
|
Anuurad E, Enkhmaa B, Berglund L. Enigmatic role of lipoprotein(a) in cardiovascular disease. Clin Transl Sci 2011; 3:327-32. [PMID: 21167011 DOI: 10.1111/j.1752-8062.2010.00238.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Lipoprotein (a), [Lp(a)] has many properties in common with low-density lipoprotein, (LDL) but contains a unique protein apolipoprotein(a), linked to apolipoprotein B-100 by a single disulfide bond. There is a substantial size heterogeneity of apo(a), and generally smaller apo(a) sizes tend to correspond to higher plasma Lp(a) levels, but this relation is far from linear, underscoring the importance to assess allele-specific apo(a) levels. The presence of apo(a), a highly charged, carbohydrate-rich, hydrophilic protein may obscure key features of the LDL moiety and offer opportunities for binding to vessel wall elements. Recently, interest in Lp(a) has increased because studies over the past decade have confirmed and more robustly demonstrated a risk factor role of Lp(a) for cardiovascular disease. In particular, levels of Lp(a) carried in particles with smaller size apo(a) isoforms are associated with coronary artery disease (CAD). Other studies suggest that proinflammatory conditions may modulate risk factor properties of Lp(a). Further, Lp(a) may act as a preferential acceptor for proinflammatory oxidized phospholipids transferred from tissues or from other lipoproteins. However, at present only a limited number of agents (e.g., nicotinic acid and estrogen) has proven efficacy in lowering Lp(a) levels. Although Lp(a) has not been definitely established as a cardiovascular risk factor and no guidelines presently recommend intervention, Lp(a)-lowering therapy might offer benefits in subgroups of patients with high Lp(a) levels.
Collapse
|
48
|
Bhattacharjee PS, Huq TS, Mandal TK, Graves RA, Muniruzzaman S, Clement C, McFerrin HE, Hill JM. A novel peptide derived from human apolipoprotein E is an inhibitor of tumor growth and ocular angiogenesis. PLoS One 2011; 6:e15905. [PMID: 21253017 PMCID: PMC3017052 DOI: 10.1371/journal.pone.0015905] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2010] [Accepted: 11/25/2010] [Indexed: 12/21/2022] Open
Abstract
Angiogenesis is a hallmark of tumor development and metastasis and now a validated target for cancer treatment. We previously reported that a novel dimer peptide (apoEdp) derived from the receptor binding region of human apolipoprotein E (apoE) inhibits virus-induced angiogenesis. However, its role in tumor anti-angiogenesis is unknown. This study demonstrates that apoEdp has anti-angiogenic property in vivo through reduction of tumor growth in a mouse model and ocular angiogenesis in a rabbit eye model. Our in vitro studies show that apoEdp inhibits human umbilical vein endothelial cell proliferation, migration, invasion and capillary tube formation. We document that apoEdp inhibits vascular endothelial growth factor-induced Flk-1 activation as well as downstream signaling pathways that involve c-Src, Akt, eNOS, FAK, and ERK1/2. These in vitro data suggest potential sites of the apoE dipeptide inhibition that could occur in vivo. This is the first evidence that a synthetic dimer peptide mimicking human apoE has anti-angiogenesis functions and could be an anti-tumor drug candidate.
Collapse
Affiliation(s)
- Partha S. Bhattacharjee
- Department of Biology, Xavier University of Louisiana, New Orleans, Louisiana, United States of America
- Department of Ophthalmology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, United States of America
| | - Tashfin S. Huq
- Department of Biology, Xavier University of Louisiana, New Orleans, Louisiana, United States of America
| | - Tarun K. Mandal
- College of Pharmacy, Xavier University of Louisiana, New Orleans, Louisiana, United States of America
| | - Richard A. Graves
- College of Pharmacy, Xavier University of Louisiana, New Orleans, Louisiana, United States of America
| | - Syed Muniruzzaman
- Department of Biology, Xavier University of Louisiana, New Orleans, Louisiana, United States of America
| | - Christian Clement
- Department of Ophthalmology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, United States of America
| | - Harris E. McFerrin
- Department of Biology, Xavier University of Louisiana, New Orleans, Louisiana, United States of America
| | - James M. Hill
- Department of Ophthalmology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, United States of America
- Neuroscience Center, Louisiana State University Health Sciences Center, New Orleans, Louisiana, United States of America
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, United States of America
- Department of Pharmacology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, United States of America
- * E-mail:
| |
Collapse
|
49
|
Packard CJ. Optimizing lipid-lowering therapy in the prevention of coronary heart disease. Expert Rev Clin Pharmacol 2010; 3:649-61. [PMID: 22111747 DOI: 10.1586/ecp.10.51] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Optimized lipid-lowering therapy is laid out in guidelines from national and international bodies. Statins are first-line treatment and instituted early in secondary prevention. The challenge in primary prevention is identification of the person at risk. This can be achieved by using scoring systems that assess classical risk factors, and then by adding information from predictive panels of biomarkers related to atherogenic pathways and by noninvasive imaging of vascular beds. At present, outcome trials validate the widespread use of statins in the population but studies of other agents have not generated proof of efficacy. Levels of high-density lipoprotein are related inversely to coronary heart disease risk but, so far, it is unclear if increasing high-density lipoprotein leads to a reduction in risk. Clinical trials on the utility of high-density lipoprotein raising on a background of statin therapy are underway.
Collapse
Affiliation(s)
- Chris J Packard
- NHS GG&C Health Board, Western Infirmary Glasgow, Tennent Blg, 38 Church Street, Glasgow, G11 6NT, UK.
| |
Collapse
|
50
|
Apolipoprotein(a) Isoforms and the Risk of Vascular Disease. J Am Coll Cardiol 2010; 55:2160-7. [DOI: 10.1016/j.jacc.2009.10.080] [Citation(s) in RCA: 234] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2009] [Revised: 10/26/2009] [Accepted: 10/26/2009] [Indexed: 11/23/2022]
|