51
|
Lasek-Bal A, Kula D, Urbanek T, Puz P, Szymszal J, Jarzab M, Halczok M, Cyplinska R, Bal W, Łabuz-Roszak B, Cieślik A, Jasnos I, Jarzab B, Ziaja D. The Association of SNPs Located in the CDKN2B-AS1 and LPA Genes With Carotid Artery Stenosis and Atherogenic Stroke. Front Neurol 2019; 10:1170. [PMID: 31824394 PMCID: PMC6883000 DOI: 10.3389/fneur.2019.01170] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 10/21/2019] [Indexed: 11/13/2022] Open
Abstract
Introduction: The aim of this project was to assess the prevalence of four selected SNPs rs4977574 and rs7857345 (CDKN2B-AS1 gene) and rs3798220 and rs10455872 polymorphisms (the LPA gene) in the subpopulation of patients with symptomatic and asymptomatic carotid stenosis. Material and Methods: This study included 623 individuals (244 patients with symptomatic carotid artery stenosis, 176 patients with asymptomatic carotid artery stenosis and 203 healthy people. All the participants underwent neurological examination, duplex Doppler ultrasound examination and molecular procedures. Results: In the first part of the analysis the assiociation of SNPs with stroke/TIA was investigated. The association was seen in symptomatic vs. control group for two SNPs: rs4977574 and rs7857345 (CDKN2B-AS1 gene); genotype distributions for rs4977574 and rs7857345 showed the statistically significant differences between patients and controls (p = 0.043 and 0.017, respectively). No association was observed for rs3798220 and rs10455872 located in the LPA gene. There were statistically significant differences between asymptomatic patients vs. control group in genotype distribution for the SNPs located in CDKN2B-AS1: rs4977574 and rs7857345 (p = 0.031 and 0.0099, respectively); and for the rs3798220 (LPA gene; p = 0.003); however, statistically significant differences did not occur for the rs10455872 polymorphism located in the LPA gene. In the next part of the evaluation, a comparison between symptomatic and asymptomatic patients was performed. Significant differences in genotype distribution were seen only for the rs3798220 polymorphism located in the LPA gene (p = 0.0015). The analysis of the prevalence of the polymorphisms in the total group (symptomatic and asymptomatic) patients in comparison with the control group showed significant differences for three polymorphisms: rs4977574 and rs7857345 (CDKN2B-AS1 gene; p = 0.015 and 0.0046, respectively) and rs3798220 (LPA gene, p = 0.044). Conclusions: The present research on the carotid artery stenosis patient cohort suggests the significant association between the rs4977574, rs7857345 and rs3798220 polymorphisms and carotid artery stenosis as well as between the rs4977574 and rs7857345 polymorphisms and atherogenic stroke. The rs4977574 and rs7857345 polymorphisms in patients with carotid artery stenosis appear to affect a person's susceptibility to atherogenic brain ischemia. Our results need to be replicated in future studies.
Collapse
Affiliation(s)
- Anetta Lasek-Bal
- Department of Neurology, School of Health Sciences, Medical University of Silesia, Katowice, Poland
| | - Dorota Kula
- Maria Skłodowska-Curie, Memorial Cancer Center and Institute of Oncology, Gliwice, Poland
| | - Tomasz Urbanek
- Department of General Surgery, Vascular Surgery, Angiology and Phlebology, Medical University of Silesia, Katowice, Poland
| | - Przemysław Puz
- Department of Neurology, School of Health Sciences, Medical University of Silesia, Katowice, Poland
| | - Jan Szymszal
- Faculty of Technical Sciences, University of Occupational Safety Management in Katowice, Katowice, Poland
| | - Michał Jarzab
- 3rd Department of Radiotherapy and Chemotherapy, Maria Sklodowska-Curie Institute-Oncology Center, Gliwice, Poland
| | - Monika Halczok
- Maria Skłodowska-Curie, Memorial Cancer Center and Institute of Oncology, Gliwice, Poland
| | - Renata Cyplinska
- Maria Skłodowska-Curie, Memorial Cancer Center and Institute of Oncology, Gliwice, Poland
| | - Wiesław Bal
- Department of Outpatient Chemotherapy, Maria Skłodowska-Curie, Memorial Cancer Center and Institute of Oncology, Gliwice, Poland
| | - Beata Łabuz-Roszak
- Department of Basic Medical Sciences, Faculty of Public Health, Medical University of Silesia, Katowice, Poland
| | - Aleksandra Cieślik
- Department of Neurology, School of Health Sciences, Medical University of Silesia, Katowice, Poland
| | - Ilona Jasnos
- Department of Neurology, School of Health Sciences, Medical University of Silesia, Katowice, Poland
| | - Barbara Jarzab
- Maria Skłodowska-Curie, Memorial Cancer Center and Institute of Oncology, Gliwice, Poland
| | - Damian Ziaja
- Department of General Surgery, Vascular Surgery, Angiology and Phlebology, Medical University of Silesia, Katowice, Poland
| |
Collapse
|
52
|
Pan Y, Li H, Wang Y, Meng X, Wang Y. Causal Effect of Lp(a) [Lipoprotein(a)] Level on Ischemic Stroke and Alzheimer Disease: A Mendelian Randomization Study. Stroke 2019; 50:3532-3539. [PMID: 31597550 DOI: 10.1161/strokeaha.119.026872] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Background and Purpose- Stroke and Alzheimer disease are 2 major causes of neurological disability in aged people and shared overlapping predictors. In recent prospective studies, high Lp(a) [lipoprotein(a)] level is associated with high risk of stroke but low risk of Alzheimer disease. Whether this reflects a causal association remains to be established. The aim of this study is to examine the causal associations of Lp(a) concentrations on ischemic stroke, ischemic stroke subtypes, and Alzheimer disease. Methods- We used 9 single-nucleotide polymorphisms associated with Lp(a) concentrations as instrumental variables. Summary-level data on ischemic stroke and its subtypes were obtained from the Multiancestry Genome-Wide Association Study of Stroke consortium with European individuals ≤446 696 individuals. Summary-level data on Alzheimer disease were obtained from the International Genomics of Alzheimer Project With European individuals ≤54 162 individuals. Two-sample Mendelian randomization (MR) estimates were calculated with inverse-variance weighted, penalized inverse-variance weighted, simple median, weighted median, and MR Pleiotropy Residual Sum and Outlier approaches, and MR-Egger regression was used to explore pleiotropy. Results- Genetically predicted 1-SD log-transformed increase in Lp(a) concentrations was associated with a substantial increase in risk of large artery stroke (odds ratio, 1.20; 95% CI, 1.11-1.30; P<0.001) and a reduce in risk of small vessel stroke (odds ratio, 0.92; 95% CI, 0.88-0.97; P=0.001) and Alzheimer disease (odds ratio, 0.94; 95% CI, 0.91-0.97; P<0.001) using inverse-variance weighted method. No significant association was observed for total ischemic stroke or cardioembolic stroke. MR-Egger indicated no evidence of pleiotropic bias. Results were broadly consistent in sensitivity analyses using penalized inverse-variance weighted, simple median, weighted median, and MR Pleiotropy Residual Sum and Outlier approaches accounting for potential genetic pleiotropy or outliers. Conclusions- This study provides evidence to support that high Lp(a) concentrations was causally associated with an increased risk of large artery stroke but a decreased risk of small vessel stroke and Alzheimer disease. The mechanism underlying the double-edged sword effect of Lp(a) concentrations on neurological system requires further investigation.
Collapse
Affiliation(s)
- Yuesong Pan
- From the Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, China; and China National Clinical Research Center for Neurological Diseases, Beijing
| | - Hao Li
- From the Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, China; and China National Clinical Research Center for Neurological Diseases, Beijing
| | - Yilong Wang
- From the Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, China; and China National Clinical Research Center for Neurological Diseases, Beijing
| | - Xia Meng
- From the Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, China; and China National Clinical Research Center for Neurological Diseases, Beijing
| | - Yongjun Wang
- From the Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, China; and China National Clinical Research Center for Neurological Diseases, Beijing
| |
Collapse
|
53
|
Boffa MB, Marar TT, Yeang C, Viney NJ, Xia S, Witztum JL, Koschinsky ML, Tsimikas S. Potent reduction of plasma lipoprotein (a) with an antisense oligonucleotide in human subjects does not affect ex vivo fibrinolysis. J Lipid Res 2019; 60:2082-2089. [PMID: 31551368 DOI: 10.1194/jlr.p094763] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 08/29/2019] [Indexed: 12/17/2022] Open
Abstract
It is postulated that lipoprotein (a) [Lp(a)] inhibits fibrinolysis, but this hypothesis has not been tested in humans due to the lack of specific Lp(a) lowering agents. Patients with elevated Lp(a) were randomized to antisense oligonucleotide [IONIS-APO(a)Rx] directed to apo(a) (n = 7) or placebo (n = 10). Ex vivo plasma lysis times and antigen concentrations of plasminogen, factor XI, plasminogen activator inhibitor 1, thrombin activatable fibrinolysis inhibitor, and fibrinogen at baseline, day 85/92/99 (peak drug effect), and day 190 (3 months off drug) were measured. The mean ± SD baseline Lp(a) levels were 477.3 ± 55.9 nmol/l in IONIS-APO(a)Rx and 362.1 ± 89.9 nmol/l in placebo. The mean± SD percentage change in Lp(a) for IONIS-APO(a)Rx was -69.3 ± 12.2% versus -5.4 ± 6.9% placebo (P < 0.0010) at day 85/92/99 and -15.6 ± 8.9% versus 3.2 ± 12.2% (P = 0.003) at day 190. Clot lysis times and coagulation/fibrinolysis-related biomarkers showed no significant differences between IONIS-APO(a)Rx and placebo at all time points. Clot lysis times were not affected by exogenously added Lp(a) at concentrations up to 200 nmol/l to plasma with very low (12.5 nmol/l) Lp(a) levels, whereas recombinant apo(a) had a potent antifibrinolytic effect. In conclusion, potent reductions of Lp(a) in patients with highly elevated Lp(a) levels do not affect ex vivo measures of fibrinolysis; the relevance of any putative antifibrinolytic effects of Lp(a) in vivo needs further study.
Collapse
Affiliation(s)
- Michael B Boffa
- Department of Biochemistry Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada
| | - Tanya T Marar
- Department of Biochemistry Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada
| | - Calvin Yeang
- Division of Endocrinology and Metabolism, University of California San Diego, La Jolla, CA
| | | | | | - Joseph L Witztum
- Division of Endocrinology and Metabolism, University of California San Diego, La Jolla, CA
| | - Marlys L Koschinsky
- Robarts Research Institute, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada
| | - Sotirios Tsimikas
- Division of Endocrinology and Metabolism, University of California San Diego, La Jolla, CA .,Ionis Pharmaceuticals, Carlsbad, CA
| |
Collapse
|
54
|
|
55
|
Chong M, Sjaarda J, Pigeyre M, Mohammadi-Shemirani P, Lali R, Shoamanesh A, Gerstein HC, Paré G. Novel Drug Targets for Ischemic Stroke Identified Through Mendelian Randomization Analysis of the Blood Proteome. Circulation 2019; 140:819-830. [PMID: 31208196 DOI: 10.1161/circulationaha.119.040180] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Novel, effective, and safe drugs are warranted for treatment of ischemic stroke. Circulating protein biomarkers with causal genetic evidence represent promising drug targets, but no systematic screen of the proteome has been performed. METHODS First, using Mendelian randomization (MR) analyses, we assessed 653 circulating proteins as possible causal mediators for 3 different subtypes of ischemic stroke: large artery atherosclerosis, cardioembolic stroke, and small artery occlusion. Second, we used MR to assess whether identified biomarkers also affect risk for intracranial bleeding, specifically intracerebral and subarachnoid hemorrhages. Third, we expanded this analysis to 679 diseases to test a broad spectrum of side effects associated with hypothetical therapeutic agents for ischemic stroke that target the identified biomarkers. For all MR analyses, summary-level data from genome-wide association studies (GWAS) were used to ascertain genetic effects on circulating biomarker levels versus disease risk. Biomarker effects were derived by meta-analysis of 5 GWAS (N≤20 509). Disease effects were derived from large GWAS analyses, including MEGASTROKE (N≤322 150) and UK Biobank (N≤408 961) studies. RESULTS Several biomarkers emerged as causal mediators for ischemic stroke. Causal mediators for cardioembolic stroke included histo-blood group ABO system transferase, coagulation factor XI, scavenger receptor class A5 (SCARA5), and tumor necrosis factor-like weak inducer of apoptosis (TNFSF12). Causal mediators for large artery atherosclerosis included ABO, cluster of differentiation 40, apolipoprotein(a), and matrix metalloproteinase-12. SCARA5 (odds ratio [OR]=0.78; 95% CI, 0.70-0.88; P=1.46×10-5) and TNFSF12 (OR=0.86; 95% CI, 0.81-0.91; P=7.69×10-7) represent novel protective mediators of cardioembolic stroke. TNFSF12 also increased the risk of subarachnoid (OR=1.53; 95% CI, 1.31-1.78; P=3.32×10-8) and intracerebral (OR=1.34; 95% CI, 1.14-1.58; P=4.05×10-4) hemorrhages, whereas SCARA5 decreased the risk of subarachnoid hemorrhage (OR=0.61; 95% CI, 0.47-0.81; P=5.20×10-4). Multiple side effects beyond stroke were identified for 6 of 7 biomarkers, most (75%) of which were beneficial. No adverse side effects were found for coagulation factor XI, apolipoprotein(a), and SCARA5. CONCLUSIONS Through a systematic MR screen of the circulating proteome, causal roles for 5 established and 2 novel biomarkers for ischemic stroke were identified. Side-effect profiles were characterized to help inform drug target prioritization. In particular, SCARA5 represents a promising target for treatment of cardioembolic stroke, with no predicted adverse side effects.
Collapse
Affiliation(s)
- Michael Chong
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Thrombosis and Atherosclerosis Research Institute, Hamilton Health Sciences, Ontario, Canada (M.C., J.S., M.P., P.M.-S., R.L., A.S., H.C.G., G.P.).,Departments of Biochemistry (M.C., G.P.)
| | - Jennifer Sjaarda
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Thrombosis and Atherosclerosis Research Institute, Hamilton Health Sciences, Ontario, Canada (M.C., J.S., M.P., P.M.-S., R.L., A.S., H.C.G., G.P.)
| | - Marie Pigeyre
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Thrombosis and Atherosclerosis Research Institute, Hamilton Health Sciences, Ontario, Canada (M.C., J.S., M.P., P.M.-S., R.L., A.S., H.C.G., G.P.)
| | - Pedrum Mohammadi-Shemirani
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Thrombosis and Atherosclerosis Research Institute, Hamilton Health Sciences, Ontario, Canada (M.C., J.S., M.P., P.M.-S., R.L., A.S., H.C.G., G.P.).,Medical Sciences (P.M.-S.)
| | - Ricky Lali
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Thrombosis and Atherosclerosis Research Institute, Hamilton Health Sciences, Ontario, Canada (M.C., J.S., M.P., P.M.-S., R.L., A.S., H.C.G., G.P.)
| | - Ashkan Shoamanesh
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Thrombosis and Atherosclerosis Research Institute, Hamilton Health Sciences, Ontario, Canada (M.C., J.S., M.P., P.M.-S., R.L., A.S., H.C.G., G.P.).,Medicine, Division of Neurology, McMaster University, Hamilton, Ontario, Canada (A.S.)
| | - Hertzel Chaim Gerstein
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Thrombosis and Atherosclerosis Research Institute, Hamilton Health Sciences, Ontario, Canada (M.C., J.S., M.P., P.M.-S., R.L., A.S., H.C.G., G.P.).,Clinical Epidemiology and Biostatistics (H.C.G., G.P.)
| | - Guillaume Paré
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Thrombosis and Atherosclerosis Research Institute, Hamilton Health Sciences, Ontario, Canada (M.C., J.S., M.P., P.M.-S., R.L., A.S., H.C.G., G.P.).,Departments of Biochemistry (M.C., G.P.).,Clinical Epidemiology and Biostatistics (H.C.G., G.P.).,Pathology and Molecular Medicine (G.P.)
| |
Collapse
|
56
|
Abstract
Stroke remains a leading cause of disability and death worldwide despite significant scientific and therapeutic advances. Therefore, there is a critical need to improve stroke prevention and treatment. In this review, we describe several examples that leverage nucleic acid therapeutics to improve stroke care through prevention, acute treatment, and recovery. Aptamer systems are under development to increase the safety and efficacy of antithrombotic and thrombolytic treatment, which represent the mainstay of medical stroke therapy. Antisense oligonucleotide therapy has shown some promise in treating stroke causes that are genetically determined and resistant to classic prevention approaches such as elevated lipoprotein (a) and cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL). Targeting microRNAs may be attractive because they regulate factors involved in neuronal cell death and reperfusion-associated injury, as well as neurorestorative pathways. Lastly, microRNAs may aid reliable etiologic classification of stroke subtypes, which is important for effective secondary stroke prevention.
Collapse
Affiliation(s)
- Nils Henninger
- Department of Neurology, University of Massachusetts Medical School, 55 Lake Ave, North, Worcester, MA, 01655, USA.
- Department of Psychiatry, University of Massachusetts Medical School, 55 Lake Ave, North, Worcester, MA, 01655, USA.
| | - Yunis Mayasi
- Division of Neurocritical Care, Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, 600 N. Wolfe Street, Baltimore, MD, 21287, USA
| |
Collapse
|
57
|
Enas EA, Varkey B, Dharmarajan TS, Pare G, Bahl VK. Lipoprotein(a): An independent, genetic, and causal factor for cardiovascular disease and acute myocardial infarction. Indian Heart J 2019; 71:99-112. [PMID: 31280836 PMCID: PMC6620428 DOI: 10.1016/j.ihj.2019.03.004] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Revised: 02/10/2019] [Accepted: 03/13/2019] [Indexed: 12/24/2022] Open
Abstract
Lipoprotein(a) [Lp(a)] is a circulating lipoprotein, and its level is largely determined by variation in the Lp(a) gene (LPA) locus encoding apo(a). Genetic variation in the LPA gene that increases Lp(a) level also increases coronary artery disease (CAD) risk, suggesting that Lp(a) is a causal factor for CAD risk. Lp(a) is the preferential lipoprotein carrier for oxidized phospholipids (OxPL), a proatherogenic and proinflammatory biomarker. Lp(a) adversely affects endothelial function, inflammation, oxidative stress, fibrinolysis, and plaque stability, leading to accelerated atherothrombosis and premature CAD. The INTER-HEART Study has established the usefulness of Lp(a) in assessing the risk of acute myocardial infarction in ethnically diverse populations with South Asians having the highest risk and population attributable risk. The 2018 Cholesterol Clinical Practice Guideline have recognized elevated Lp(a) as an atherosclerotic cardiovascular disease risk enhancer for initiating or intensifying statin therapy.
Collapse
Affiliation(s)
- Enas A Enas
- Coronary Artery Disease in Indians (CADI) Research Foundation, Lisle, IL USA.
| | - Basil Varkey
- Emeritus of Medicine, Medical College of Wisconsin, USA
| | - T S Dharmarajan
- Medicine, Albert Einstein College of Medicine, Bronx, NY, USA; Department of Medicine, Montefiore Medical Center (Wakefield Campus), Bronx, NY, USA
| | | | - Vinay K Bahl
- Department of Cardiology, All India Institute of Medical Sciences New Delhi, India
| |
Collapse
|
58
|
Borrelli MJ, Youssef A, Boffa MB, Koschinsky ML. New Frontiers in Lp(a)-Targeted Therapies. Trends Pharmacol Sci 2019; 40:212-225. [PMID: 30732864 DOI: 10.1016/j.tips.2019.01.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 01/07/2019] [Accepted: 01/08/2019] [Indexed: 12/13/2022]
Abstract
Interest in lipoprotein (a) [Lp(a)] has exploded over the past decade with the emergence of genetic and epidemiological studies pinpointing elevated levels of this unique lipoprotein as a causal risk factor for atherosclerotic cardiovascular disease (ASCVD) and calcific aortic valve disease (CAVD). This review summarizes the most recent discoveries regarding therapeutic approaches to lower Lp(a) and presents these findings in the context of an emerging, although far from complete, understanding of the biosynthesis and catabolism of Lp(a). Application of Lp(a)-specific lowering agents to outcome trials will be the key to opening this new frontier in the battle against CVD.
Collapse
Affiliation(s)
- Matthew J Borrelli
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Amer Youssef
- Robarts Research Institute, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Michael B Boffa
- Robarts Research Institute, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada; Department of Biochemistry, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Marlys L Koschinsky
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada; Robarts Research Institute, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada.
| |
Collapse
|
59
|
Boffa MB, Koschinsky ML. Oxidized phospholipids as a unifying theory for lipoprotein(a) and cardiovascular disease. Nat Rev Cardiol 2019; 16:305-318. [DOI: 10.1038/s41569-018-0153-2] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
60
|
Momtazi-Borojeni AA, Katsiki N, Pirro M, Banach M, Rasadi KA, Sahebkar A. Dietary natural products as emerging lipoprotein(a)-lowering agents. J Cell Physiol 2019; 234:12581-12594. [PMID: 30637725 DOI: 10.1002/jcp.28134] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 12/07/2018] [Indexed: 12/13/2022]
Abstract
Elevated plasma lipoprotein(a) (Lp(a)) levels are associated with an increased risk of cardiovascular disease (CVD). Hitherto, niacin has been the drug of choice to reduce elevated Lp(a) levels in hyperlipidemic patients but its efficacy in reducing CVD outcomes has been seriously questioned by recent clinical trials. Additional drugs may reduce to some extent plasma Lp(a) levels but the lack of a specific therapeutic indication for Lp(a)-lowering limits profoundly reduce their use. An attractive therapeutic option is natural products. In several preclinical and clinical studies as well as meta-analyses, natural products, including l-carnitine, coenzyme Q 10 , and xuezhikang were shown to significantly decrease Lp(a) levels in patients with Lp(a) hyperlipoproteinemia. Other natural products, such as pectin, Ginkgo biloba, flaxseed, red wine, resveratrol and curcuminoids can also reduce elevated Lp(a) concentrations but to a lesser degree. In conclusion, aforementioned natural products may represent promising therapeutic agents for Lp(a) lowering.
Collapse
Affiliation(s)
- Amir Abbas Momtazi-Borojeni
- Department of Medical Biotechnology, Nanotechnology Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Niki Katsiki
- Second Propedeutic Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, Hippocration Hospital, Thessaloniki, Greece
| | - Matteo Pirro
- Unit of Internal Medicine, Angiology and Arteriosclerosis Diseases, Department of Medicine, University of Perugia, Perugia, Italy
| | - Maciej Banach
- Department of Hypertension, WAM University Hospital in Lodz, Medical University of Lodz, Lodz, Poland.,Polish Mother's Memorial Hospital Research Institute, Lodz, Poland
| | - Khalid Al Rasadi
- Department of Clinical Biochemistry, Sultan Qaboos University Hospital, Muscat, Oman
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
61
|
Salvagno GL, Pavan C, Lippi G. Rare thrombophilic conditions. ANNALS OF TRANSLATIONAL MEDICINE 2018; 6:342. [PMID: 30306081 DOI: 10.21037/atm.2018.08.12] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Thrombophilia, either acquired or inherited, can be defined as a predisposition to developing thromboembolic complications. Since the discovery of antithrombin deficiency in the 1965, many other conditions have been described so far, which have then allowed to currently detect an inherited or acquired predisposition in approximately 60-70% of patients with thromboembolic disorders. These prothrombotic risk factors mainly include qualitative or quantitative defects of endogenous coagulation factor inhibitors, increased concentration or function of clotting proteins, defects in the fibrinolytic system, impaired platelet function, and hyperhomocysteinemia. In this review article, we aim to provide an overview on epidemiologic, clinic and laboratory aspects of both acquired and inherited rare thrombophilic risk factors, especially including dysfibrinogenemia, heparin cofactor II, thrombomodulin, lipoprotein(a), sticky platelet syndrome, plasminogen activator inhibitor-1 apolipoprotein E, tissue factor pathway inhibitor, paroxysmal nocturnal haemoglobinuria and heparin-induced thrombocytopenia.
Collapse
Affiliation(s)
| | - Chiara Pavan
- Division of Geriatric Medicine, Mater Salutis Hospital, Legnago, Verona, Italy
| | - Giuseppe Lippi
- Section of Clinical Biochemistry, University of Verona, Verona, Italy
| |
Collapse
|
62
|
Abstract
PURPOSE OF REVIEW DNA copy number variations (CNVs) are quantitative structural rearrangements that include deletions, duplications, and higher order amplifications. Because of technical limitations, the contribution of this common form of genetic variation to regulation of lipid metabolism and dyslipidemia has been underestimated. RECENT FINDINGS Recent literature involving CNVs and dyslipidemias has focused mainly on rare CNVs causing familial hypercholesterolemia, and a common CNV polymorphism as the major determinant of lipoprotein(a) plasma concentrations. Additionally, there is tantalizing evidence of largely uninvestigated but plausible presence of CNVs underlying other dyslipidemias. We also discuss the future role of improved technologies in facilitating more economic, routine CNV assessment in dyslipidemias. SUMMARY CNVs account for large proportion of human genetic variation and are already known to contribute to susceptibility of dyslipidemias, particularly in about 10% of familial hypercholesterolemia patients. Increasing availability of clinical next-generation sequencing and bioinformatics presents a cost-effective opportunity for novel CNV discoveries in dyslipidemias.
Collapse
|
63
|
Gencer B, Kronenberg F, Stroes ES, Mach F. Lipoprotein(a): the revenant. Eur Heart J 2018; 38:1553-1560. [PMID: 28329241 DOI: 10.1093/eurheartj/ehx033] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 01/16/2017] [Indexed: 11/12/2022] Open
Abstract
In the mid-1990s, the days of lipoprotein(a) [Lp(a)] were numbered and many people would not have placed a bet on this lipid particle making it to the next century. However, genetic studies brought Lp(a) back to the front-stage after a Mendelian randomization approach used for the first time provided strong support for a causal role of high Lp(a) concentrations in cardiovascular disease and later also for aortic valve stenosis. This encouraged the use of therapeutic interventions to lower Lp(a) as well numerous drug developments, although these approaches mainly targeted LDL cholesterol, while the Lp(a)-lowering effect was only a 'side-effect'. Several drug developments did show a potent Lp(a)-lowering effect but did not make it to endpoint studies, mainly for safety reasons. Currently, three therapeutic approaches are either already in place or look highly promising: (i) lipid apheresis (specific or unspecific for Lp(a)) markedly decreases Lp(a) concentrations as well as cardiovascular endpoints; (ii) PCSK9 inhibitors which, besides lowering LDL cholesterol also decrease Lp(a) by roughly 30%; and (iii) antisense therapy targeting apolipoprotein(a) which has shown to specifically lower Lp(a) concentrations by up to 90% in phase 1 and 2 trials without influencing other lipids. Until the results of phase 3 outcome studies are available for antisense therapy, we will have to exercise patience, but with optimism since never before have we had the tools we have now to prove Koch's extrapolated postulate that lowering high Lp(a) concentrations might be protective against cardiovascular disease.
Collapse
Affiliation(s)
- Baris Gencer
- Cardiology Division, Geneva University Hospitals, Switzerland
| | - Florian Kronenberg
- Department of Medical Genetics, Division of Genetic Epidemiology, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Austria
| | - Erik S Stroes
- Academic Medical Center, Amsterdam, AZ 1100, The Netherlands
| | - François Mach
- Cardiology Division, Geneva University Hospitals, Switzerland
| |
Collapse
|
64
|
Helgadottir A, Thorleifsson G, Gretarsdottir S, Stefansson OA, Tragante V, Thorolfsdottir RB, Jonsdottir I, Bjornsson T, Steinthorsdottir V, Verweij N, Nielsen JB, Zhou W, Folkersen L, Martinsson A, Heydarpour M, Prakash S, Oskarsson G, Gudbjartsson T, Geirsson A, Olafsson I, Sigurdsson EL, Almgren P, Melander O, Franco-Cereceda A, Hamsten A, Fritsche L, Lin M, Yang B, Hornsby W, Guo D, Brummett CM, Abecasis G, Mathis M, Milewicz D, Body SC, Eriksson P, Willer CJ, Hveem K, Newton-Cheh C, Smith JG, Danielsen R, Thorgeirsson G, Thorsteinsdottir U, Gudbjartsson DF, Holm H, Stefansson K. Genome-wide analysis yields new loci associating with aortic valve stenosis. Nat Commun 2018; 9:987. [PMID: 29511194 PMCID: PMC5840367 DOI: 10.1038/s41467-018-03252-6] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 01/31/2018] [Indexed: 01/09/2023] Open
Abstract
Aortic valve stenosis (AS) is the most common valvular heart disease, and valve replacement is the only definitive treatment. Here we report a large genome-wide association (GWA) study of 2,457 Icelandic AS cases and 349,342 controls with a follow-up in up to 4,850 cases and 451,731 controls of European ancestry. We identify two new AS loci, on chromosome 1p21 near PALMD (rs7543130; odds ratio (OR) = 1.20, P = 1.2 × 10−22) and on chromosome 2q22 in TEX41 (rs1830321; OR = 1.15, P = 1.8 × 10−13). Rs7543130 also associates with bicuspid aortic valve (BAV) (OR = 1.28, P = 6.6 × 10−10) and aortic root diameter (P = 1.30 × 10−8), and rs1830321 associates with BAV (OR = 1.12, P = 5.3 × 10−3) and coronary artery disease (OR = 1.05, P = 9.3 × 10−5). The results implicate both cardiac developmental abnormalities and atherosclerosis-like processes in the pathogenesis of AS. We show that several pathways are shared by CAD and AS. Causal analysis suggests that the shared risk factors of Lp(a) and non-high-density lipoprotein cholesterol contribute substantially to the frequent co-occurence of these diseases. Aortic valve stenosis (AS) is the most common valvular heart disease. Here the authors identify two new AS loci that also associate with bicuspid aortic valve, aortic root diameter and/or coronary artery disease implicating both developmental abnormalities and atherosclerosis-like processes in AS.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ingileif Jonsdottir
- deCODE genetics/Amgen Inc., Reykjavik, 101, Iceland.,Faculty of Medicine, University of Iceland, Reykjavik, 101, Iceland
| | | | | | - Niek Verweij
- Department of Cardiology, University of Groningen, University Medical Center Groningen, 9700 RB, Groningen, The Netherlands.,Medical and Population Genetics Program, Broad Institute of MIT and Harvard, Cambridge, 02142, MA, USA
| | - Jonas B Nielsen
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, 48109, MI, USA
| | - Wei Zhou
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, 48109, MI, USA
| | - Lasse Folkersen
- Cardiovascular Medicine Unit, Department of Medicine, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, 17176, Sweden.,Department of Bioinformatics, Technical University of Denmark, Copenhagen, 2800, Denmark
| | - Andreas Martinsson
- Department of Cardiology, Clinical Sciences, Lund University and Skåne University Hospital, Lund, 22185, Sweden
| | - Mahyar Heydarpour
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, 02115, USA
| | - Siddharth Prakash
- Department of Internal Medicine, Division of Medical Genetics, University of Texas Health Science Center at Houston, Houston, 77030, TX, USA
| | - Gylfi Oskarsson
- Childrens Hospital, Landspitali National University Hospital of Iceland, Reykjavik, 101, Iceland
| | - Tomas Gudbjartsson
- Department of Surgery and Cardiothoracic Surgery, Landspitali National University Hospital, Reykjavik, 101, Iceland
| | - Arnar Geirsson
- Section of Cardiac Surgery, Department of Surgery, Yale University School of Medicine, New Haven, 06510, CT, USA
| | - Isleifur Olafsson
- Department of Clinical Biochemistry, Landspitali National University Hospital, Reykjavik, 101, Iceland
| | - Emil L Sigurdsson
- Heilsugaeslan Solvangi, Hafnarfjördur, 220, Iceland.,Department of Family Medicine, University of Iceland, Reykjavik, 101, Iceland
| | - Peter Almgren
- Department of Clinical Sciences, Lund University, Malmö, 22185, Sweden.,Department of Internal Medicine, Skåne University Hospital, Malmö, 22185, Sweden
| | - Olle Melander
- Department of Clinical Sciences, Lund University, Malmö, 22185, Sweden.,Department of Internal Medicine, Skåne University Hospital, Malmö, 22185, Sweden
| | - Anders Franco-Cereceda
- Cardiothoracic Surgery Unit, Department of Molecular Medicine and Surgery, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, 17176, Sweden
| | - Anders Hamsten
- Cardiovascular Medicine Unit, Department of Medicine, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, 17176, Sweden
| | - Lars Fritsche
- HUNT Research Centre, Department of Public Health and General Practice, Norwegian University of Science and Technology, Levanger, 7491, Norway.,K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health, Norwegian University of Science and Technology, Trondheim, 7491, Norway
| | - Maoxuan Lin
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, 48109, MI, USA
| | - Bo Yang
- Department of Cardiac Surgery, University of Michigan, Ann Arbor, MI, 48105, USA.,Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Whitney Hornsby
- Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Dongchuan Guo
- Department of Internal Medicine, Division of Medical Genetics, University of Texas Health Science Center at Houston, Houston, 77030, TX, USA
| | - Chad M Brummett
- Department of Anesthesiology, University of Michigan, Ann Arbor, MI, 48105, USA
| | - Gonçalo Abecasis
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Michael Mathis
- Department of Anesthesiology, University of Michigan, Ann Arbor, MI, 48105, USA
| | - Dianna Milewicz
- Department of Internal Medicine, Division of Medical Genetics, University of Texas Health Science Center at Houston, Houston, 77030, TX, USA.,Medicine Services, Texas Heart Institute, St. Luke's Episcopal Hospital, Houston, TX, 77030, USA
| | - Simon C Body
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, 02115, USA
| | - Per Eriksson
- Cardiovascular Medicine Unit, Department of Medicine, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, 17176, Sweden
| | - Cristen J Willer
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, 48109, MI, USA.,Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, 48109, MI, USA.,Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI, 48109, USA.,Department of Human Genetics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Kristian Hveem
- HUNT Research Centre, Department of Public Health and General Practice, Norwegian University of Science and Technology, Levanger, 7491, Norway.,K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health, Norwegian University of Science and Technology, Trondheim, 7491, Norway
| | - Christopher Newton-Cheh
- Medical and Population Genetics Program, Broad Institute of MIT and Harvard, Cambridge, 02142, MA, USA.,Massachusetts General Hospital, Harvard Medical School, Broad Institute of Harvard and MIT, Boston, MA, 02114, USA.,Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - J Gustav Smith
- Department of Cardiology, Clinical Sciences, Lund University and Skåne University Hospital, Lund, 22185, Sweden
| | - Ragnar Danielsen
- Faculty of Medicine, University of Iceland, Reykjavik, 101, Iceland.,Department of Internal Medicine, Division of Cardiology, Landspitali National University Hospital of Iceland, Reykjavik, 101, Iceland
| | - Gudmundur Thorgeirsson
- deCODE genetics/Amgen Inc., Reykjavik, 101, Iceland.,Faculty of Medicine, University of Iceland, Reykjavik, 101, Iceland.,Department of Internal Medicine, Division of Cardiology, Landspitali National University Hospital of Iceland, Reykjavik, 101, Iceland
| | - Unnur Thorsteinsdottir
- deCODE genetics/Amgen Inc., Reykjavik, 101, Iceland.,Faculty of Medicine, University of Iceland, Reykjavik, 101, Iceland
| | - Daniel F Gudbjartsson
- deCODE genetics/Amgen Inc., Reykjavik, 101, Iceland.,School of Engineering and Natural Sciences, University of Iceland, Reykjavik, 101, Iceland
| | - Hilma Holm
- deCODE genetics/Amgen Inc., Reykjavik, 101, Iceland
| | - Kari Stefansson
- deCODE genetics/Amgen Inc., Reykjavik, 101, Iceland. .,Faculty of Medicine, University of Iceland, Reykjavik, 101, Iceland.
| |
Collapse
|
65
|
Abstract
PURPOSE The Emory Cardiovascular Biobank (EmCAB) is an ongoing prospective registry of patients undergoing cardiac catheterisation, which was established to identify novel factors associated with the pathobiological process and treatment of cardiovascular disease. PARTICIPANTS Individuals aged 18 years and older undergoing cardiac catheterisation at three Emory Healthcare sites in Atlanta are asked to participate in this prospective registry. Around 95% agree to participate. Around 7000 unique patients have been enrolled. The current data set contains detailed phenotyping, patient outcomes, genomics, protein biomarkers, regenerative markers, transcriptomic analysis, metabolomics profiling and longitudinal follow-up for adverse cardiovascular outcomes. FINDINGS TO DATE Thus far, the EmCAB has approximately 3000 major cardiovascular events. About 48% of the EmCAB participants have more than 5 years of follow-up. It is a great resource for discovery of novel predictive factors for cardiovascular disease outcomes, including genomics, transcriptomics, protein biomarkers, oxidative stress markers and circulating progenitor cells. Several circulating inflammatory markers have shown to improve risk prediction metrics beyond standard risk factors. FUTURE PLANS Future integrative -omics analyses will provide the cardiovascular research community opportunities for subsequent mechanistic confirmation studies, which will promote the development of effective personalised therapy that leads to clinical care tailored to the individual patient.
Collapse
Affiliation(s)
- Yi-An Ko
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Salim Hayek
- Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Pratik Sandesara
- Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Ayman Samman Tahhan
- Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Arshed Quyyumi
- Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
66
|
Scipione CA, Koschinsky ML, Boffa MB. Lipoprotein(a) in clinical practice: New perspectives from basic and translational science. Crit Rev Clin Lab Sci 2017; 55:33-54. [PMID: 29262744 DOI: 10.1080/10408363.2017.1415866] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Elevated plasma concentrations of lipoprotein(a) (Lp(a)) are a causal risk factor for coronary heart disease (CHD) and calcific aortic valve stenosis (CAVS). Genetic, epidemiological and in vitro data provide strong evidence for a pathogenic role for Lp(a) in the progression of atherothrombotic disease. Despite these advancements and a race to develop new Lp(a) lowering therapies, there are still many unanswered and emerging questions about the metabolism and pathophysiology of Lp(a). New studies have drawn attention to Lp(a) as a contributor to novel pathogenic processes, yet the mechanisms underlying the contribution of Lp(a) to CVD remain enigmatic. New therapeutics show promise in lowering plasma Lp(a) levels, although the complete mechanisms of Lp(a) lowering are not fully understood. Specific agents targeted to apolipoprotein(a) (apo(a)), namely antisense oligonucleotide therapy, demonstrate potential to decrease Lp(a) to levels below the 30-50 mg/dL (75-150 nmol/L) CVD risk threshold. This therapeutic approach should aid in assessing the benefit of lowering Lp(a) in a clinical setting.
Collapse
Affiliation(s)
- Corey A Scipione
- a Department of Advanced Diagnostics , Toronto General Hospital Research Institute, UHN , Toronto , Canada
| | - Marlys L Koschinsky
- b Robarts Research Institute , Western University , London , Canada.,c Department of Physiology & Pharmacology , Schulich School of Medicine & Dentistry, Western University , London , Canada
| | - Michael B Boffa
- d Department of Biochemistry , Western University , London , Canada
| |
Collapse
|
67
|
Ho JE. Harnessing the Power of Pharmacometabolomics: The Metabolic Footprint of Statins. ACTA ACUST UNITED AC 2017; 10:CIRCGENETICS.117.002014. [PMID: 29237684 DOI: 10.1161/circgenetics.117.002014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Jennifer E Ho
- From the Cardiovascular Research Center and the Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston.
| |
Collapse
|
68
|
Franchini M, Martinelli I, Mannucci PM. Uncertain thrombophilia markers. Thromb Haemost 2017; 115:25-30. [DOI: 10.1160/th15-06-0478] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 07/03/2015] [Indexed: 11/05/2022]
Abstract
SummaryThe development of venous thromboembolism (VTE), which includes deep-vein thrombosis and pulmonary embolism, may be associated with inherited or acquired risk factors that can be measured in plasma or DNA testing. The main inherited thrombophilias include the plasma deficiencies of the natural anticoagulants antithrombin, protein C and S; the gain-of-function mutations factor V Leiden and prothrombin G20210A; some dysfibrinogenaemias and high plasma levels of coagulation factor VIII. Besides these established biomarkers, which usually represent the first-level laboratory tests for thrombophilia screening, a number of additional abnormalities have been less consistently associated with an increased VTE risk. These uncertain causes of thrombophilias will be discussed in this narrative review, focusing on their clinical impact and the underlying pathogenetic mechanisms. Currently, there is insufficient ground to recommend their inclusion within the framework of conventional thrombophilia testing.
Collapse
|
69
|
Ellis KL, Boffa MB, Sahebkar A, Koschinsky ML, Watts GF. The renaissance of lipoprotein(a): Brave new world for preventive cardiology? Prog Lipid Res 2017; 68:57-82. [DOI: 10.1016/j.plipres.2017.09.001] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 09/01/2017] [Accepted: 09/05/2017] [Indexed: 12/24/2022]
|
70
|
Scipione CA, McAiney JT, Simard DJ, Bazzi ZA, Gemin M, Romagnuolo R, Macrae FL, Ariëns RA, Hegele RA, Auld J, Gauld JW, Boffa MB, Koschinsky ML. Characterization of the I4399M variant of apolipoprotein(a): implications for altered prothrombotic properties of lipoprotein(a). J Thromb Haemost 2017; 15:1834-1844. [PMID: 28632940 DOI: 10.1111/jth.13759] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Indexed: 11/30/2022]
Abstract
Essentials Elevated lipoproteinp(a) is an independent and causal risk factor for atherothrombotic diseases. rs3798220 (Ile/Met substitution in apo(a) protease-like domain) is associated with disease risk. Recombinant I4399M apo(a) altered clot structure to accelerate coagulation/delay fibrinolysis. Evidence was found for increased solvent exposure and oxidation of Met residue. SUMMARY Background Lipoprotein(a) (Lp[a]) is a causal risk factor for a variety of cardiovascular diseases. Apolipoprotein(a) (apo[a]), the distinguishing component of Lp(a), is homologous with plasminogen, suggesting that Lp(a) can interfere with the normal fibrinolytic functions of plasminogen. This has implications for the persistence of fibrin clots in the vasculature and hence for atherothrombotic diseases. A single-nucleotide polymorphism (SNP) (rs3798220) in the gene encoding apo(a) has been reported that results in an Ile→Met substitution in the protease-like domain (I4399M variant). In population studies, the I4399M variant has been correlated with elevated plasma Lp(a) levels and higher coronary heart disease risk, and carriers of the SNP had increased cardiovascular benefit from aspirin therapy. In vitro studies suggested an antifibrinolytic role for Lp(a) containing this variant. Objectives We performed a series of experiments to assess the effect of the Ile→Met substitution on fibrin clot formation and lysis, and on the architecture of the clots. Results We found that the Met variant decreased coagulation time and increased fibrin clot lysis time as compared with wild-type apo(a). Furthermore, we observed that the presence of the Met variant significantly increased fibrin fiber width in plasma clots formed ex vivo, while having no effect on fiber density. Mass spectrometry analysis of a recombinant apo(a) species containing the Met variant revealed sulfoxide modification of the Met residue. Conclusions Our data suggest that the I4399M variant differs structurally from wild-type apo(a), which may underlie key differences related to its effects on fibrin clot architecture and fibrinolysis.
Collapse
Affiliation(s)
- C A Scipione
- Robarts Research Institute, London, Ontario, Canada
- Department of Physiology & Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - J T McAiney
- Department of Chemistry & Biochemistry, University of Windsor, Windsor, Ontario, Canada
| | - D J Simard
- Department of Chemistry & Biochemistry, University of Windsor, Windsor, Ontario, Canada
| | - Z A Bazzi
- Department of Biochemistry, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - M Gemin
- Department of Chemistry & Biochemistry, University of Windsor, Windsor, Ontario, Canada
| | - R Romagnuolo
- University Health Network, Toronto, Ontario, Canada
| | - F L Macrae
- Thrombosis and Tissue Repair Group, Division of Cardiovascular and Diabetes Research, Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - R A Ariëns
- Thrombosis and Tissue Repair Group, Division of Cardiovascular and Diabetes Research, Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - R A Hegele
- Robarts Research Institute, London, Ontario, Canada
- Department of Biochemistry, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada
- Department of Medicine, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - J Auld
- Department of Chemistry & Biochemistry, University of Windsor, Windsor, Ontario, Canada
| | - J W Gauld
- Department of Chemistry & Biochemistry, University of Windsor, Windsor, Ontario, Canada
| | - M B Boffa
- Department of Biochemistry, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - M L Koschinsky
- Robarts Research Institute, London, Ontario, Canada
- Department of Physiology & Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
71
|
Boffa MB. Emerging Therapeutic Options for Lowering of Lipoprotein(a): Implications for Prevention of Cardiovascular Disease. Curr Atheroscler Rep 2017; 18:69. [PMID: 27761705 DOI: 10.1007/s11883-016-0622-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW Elevated plasma concentrations of lipoprotein(a) (Lp(a)) are an independent and causal risk factor for cardiovascular diseases including coronary artery disease, ischemic stroke, and calcific aortic valve stenosis. This review summarizes the rationale for Lp(a) lowering and surveys relevant clinical trial data using a variety of agents capable of lowering Lp(a). RECENT FINDINGS Contemporary guidelines and recommendations outline populations of patients who should be screened for elevated Lp(a) and who might benefit from Lp(a) lowering. Therapies including drugs and apheresis have been described that lower Lp(a) levels modestly (∼20 %) to dramatically (∼80 %). Existing therapies that lower Lp(a) also have beneficial effects on other aspects of the lipid profile, with the exception of Lp(a)-specific apheresis and an antisense oligonucleotide that targets the mRNA encoding apolipoprotein(a). No clinical trials conducted to date have managed to answer the key question of whether Lp(a) lowering confers a benefit in terms of ameliorating cardiovascular risk, although additional outcome trials of therapies that lower Lp(a) are ongoing. It is more likely, however, that Lp(a)-specific agents will provide the most appropriate approach for addressing this question.
Collapse
Affiliation(s)
- Michael B Boffa
- Department of Biochemistry, Room 4245A Robarts Research Institute, University of Western Ontario, 1151 Richmond Street North, London, ON, Canada, N6A 5B7.
| |
Collapse
|
72
|
Tsimikas S. A Test in Context: Lipoprotein(a): Diagnosis, Prognosis, Controversies, and Emerging Therapies. J Am Coll Cardiol 2017; 69:692-711. [PMID: 28183512 DOI: 10.1016/j.jacc.2016.11.042] [Citation(s) in RCA: 653] [Impact Index Per Article: 81.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 11/10/2016] [Accepted: 11/21/2016] [Indexed: 12/14/2022]
Abstract
Evidence that elevated lipoprotein(a) (Lp[a]) levels contribute to cardiovascular disease (CVD) and calcific aortic valve stenosis (CAVS) is substantial. Development of isoform-independent assays, in concert with genetic, epidemiological, translational, and pathophysiological insights, have established Lp(a) as an independent, genetic, and likely causal risk factor for CVD and CAVS. These observations are consistent across a broad spectrum of patients, risk factors, and concomitant therapies, including patients with low-density lipoprotein cholesterol <70 mg/dl. Statins tend to increase Lp(a) levels, possibly contributing to the "residual risk" noted in outcomes trials and at the bedside. Recently approved proprotein convertase subtilisin/kexin-type 9 inhibitors and mipomersen lower Lp(a) 20% to 30%, and emerging RNA-targeted therapies lower Lp(a) >80%. These approaches will allow testing of the "Lp(a) hypothesis" in clinical trials. This review summarizes the current landscape of Lp(a), discusses controversies, and reviews emerging therapies to reduce plasma Lp(a) levels to decrease risk of CVD and CAVS.
Collapse
Affiliation(s)
- Sotirios Tsimikas
- Division of Cardiovascular Medicine, Sulpizio Cardiovascular Center, University of California San Diego, La Jolla, California.
| |
Collapse
|
73
|
Zewinger S, Kleber ME, Tragante V, McCubrey RO, Schmidt AF, Direk K, Laufs U, Werner C, Koenig W, Rothenbacher D, Mons U, Breitling LP, Brenner H, Jennings RT, Petrakis I, Triem S, Klug M, Filips A, Blankenberg S, Waldeyer C, Sinning C, Schnabel RB, Lackner KJ, Vlachopoulou E, Nygård O, Svingen GFT, Pedersen ER, Tell GS, Sinisalo J, Nieminen MS, Laaksonen R, Trompet S, Smit RAJ, Sattar N, Jukema JW, Groesdonk HV, Delgado G, Stojakovic T, Pilbrow AP, Cameron VA, Richards AM, Doughty RN, Gong Y, Cooper-DeHoff R, Johnson J, Scholz M, Beutner F, Thiery J, Smith JG, Vilmundarson RO, McPherson R, Stewart AFR, Cresci S, Lenzini PA, Spertus JA, Olivieri O, Girelli D, Martinelli NI, Leiherer A, Saely CH, Drexel H, Mündlein A, Braund PS, Nelson CP, Samani NJ, Kofink D, Hoefer IE, Pasterkamp G, Quyyumi AA, Ko YA, Hartiala JA, Allayee H, Tang WHW, Hazen SL, Eriksson N, Held C, Hagström E, Wallentin L, Åkerblom A, Siegbahn A, Karp I, Labos C, Pilote L, Engert JC, Brophy JM, Thanassoulis G, Bogaty P, Szczeklik W, Kaczor M, Sanak M, Virani SS, Ballantyne CM, Lee VV, Boerwinkle E, Holmes MV, Horne BD, Hingorani A, Asselbergs FW, Patel RS, Krämer BK, Scharnagl H, Fliser D, März W, Speer T. Relations between lipoprotein(a) concentrations, LPA genetic variants, and the risk of mortality in patients with established coronary heart disease: a molecular and genetic association study. Lancet Diabetes Endocrinol 2017; 5:534-543. [PMID: 28566218 PMCID: PMC5651679 DOI: 10.1016/s2213-8587(17)30096-7] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 01/25/2017] [Accepted: 02/14/2017] [Indexed: 01/02/2023]
Abstract
BACKGROUND Lipoprotein(a) concentrations in plasma are associated with cardiovascular risk in the general population. Whether lipoprotein(a) concentrations or LPA genetic variants predict long-term mortality in patients with established coronary heart disease remains less clear. METHODS We obtained data from 3313 patients with established coronary heart disease in the Ludwigshafen Risk and Cardiovascular Health (LURIC) study. We tested associations of tertiles of lipoprotein(a) concentration in plasma and two LPA single-nucleotide polymorphisms ([SNPs] rs10455872 and rs3798220) with all-cause mortality and cardiovascular mortality by Cox regression analysis and with severity of disease by generalised linear modelling, with and without adjustment for age, sex, diabetes diagnosis, systolic blood pressure, BMI, smoking status, estimated glomerular filtration rate, LDL-cholesterol concentration, and use of lipid-lowering therapy. Results for plasma lipoprotein(a) concentrations were validated in five independent studies involving 10 195 patients with established coronary heart disease. Results for genetic associations were replicated through large-scale collaborative analysis in the GENIUS-CHD consortium, comprising 106 353 patients with established coronary heart disease and 19 332 deaths in 22 studies or cohorts. FINDINGS The median follow-up was 9·9 years. Increased severity of coronary heart disease was associated with lipoprotein(a) concentrations in plasma in the highest tertile (adjusted hazard radio [HR] 1·44, 95% CI 1·14-1·83) and the presence of either LPA SNP (1·88, 1·40-2·53). No associations were found in LURIC with all-cause mortality (highest tertile of lipoprotein(a) concentration in plasma 0·95, 0·81-1·11 and either LPA SNP 1·10, 0·92-1·31) or cardiovascular mortality (0·99, 0·81-1·2 and 1·13, 0·90-1·40, respectively) or in the validation studies. INTERPRETATION In patients with prevalent coronary heart disease, lipoprotein(a) concentrations and genetic variants showed no associations with mortality. We conclude that these variables are not useful risk factors to measure to predict progression to death after coronary heart disease is established. FUNDING Seventh Framework Programme for Research and Technical Development (AtheroRemo and RiskyCAD), INTERREG IV Oberrhein Programme, Deutsche Nierenstiftung, Else-Kroener Fresenius Foundation, Deutsche Stiftung für Herzforschung, Deutsche Forschungsgemeinschaft, Saarland University, German Federal Ministry of Education and Research, Willy Robert Pitzer Foundation, and Waldburg-Zeil Clinics Isny.
Collapse
Affiliation(s)
- Stephen Zewinger
- Department of Internal Medicine IV, Saarland University Hospital, Homburg/Saar, Germany
| | - Marcus E Kleber
- Fifth Department of Medicine, University Heidelberg, Mannheim, Germany; Institute of Nutrition, Friedrich-Schiller University, Jena, Germany
| | - Vinicius Tragante
- Department of Cardiology, Heart and Lungs Division, UMC Utrecht, Utrecht, Netherlands
| | - Raymond O McCubrey
- Intermountain Heart Institute, Intermountain Medical Center, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Amand F Schmidt
- Institute of Cardiovascular Science Facultyof Population Health Science, University College London, London, UK
| | - Kenan Direk
- Institute of Cardiovascular Science Facultyof Population Health Science, University College London, London, UK
| | - Ulrich Laufs
- Department of Internal Medicine III, Saarland University Hospital, Homburg/Saar, Germany
| | - Christian Werner
- Department of Internal Medicine III, Saarland University Hospital, Homburg/Saar, Germany
| | - Wolfgang Koenig
- Department of Internal Medicine II-Cardiology, University of Ulm Medical Centre, Ulm, Germany; Deutsches Herzzentrum München, Technische Universität München, Munich, Germany; German Centre of Cardiovascular Research (DZHK), Partner site Munich Heart Alliance, Munich, Germany
| | - Dietrich Rothenbacher
- Division of Clinical Epidemiology and Ageing Research, German Cancer Centre (DKFZ), Heidelberg, Germany; Institute of Epidemiology and Medical Biometry, Ulm University, Ulm, Germany
| | - Ute Mons
- Division of Clinical Epidemiology and Ageing Research, German Cancer Centre (DKFZ), Heidelberg, Germany
| | - Lutz P Breitling
- Division of Clinical Epidemiology and Ageing Research, German Cancer Centre (DKFZ), Heidelberg, Germany
| | - Herrmann Brenner
- Network Ageing Research, University Heidelberg, Mannheim, Germany; Division of Clinical Epidemiology and Ageing Research, German Cancer Centre (DKFZ), Heidelberg, Germany
| | - Richard T Jennings
- Department of Internal Medicine IV, Saarland University Hospital, Homburg/Saar, Germany
| | - Ioannis Petrakis
- Department of Internal Medicine IV, Saarland University Hospital, Homburg/Saar, Germany
| | - Sarah Triem
- Department of Internal Medicine IV, Saarland University Hospital, Homburg/Saar, Germany
| | - Mira Klug
- Department of Internal Medicine IV, Saarland University Hospital, Homburg/Saar, Germany
| | - Alexandra Filips
- Department of Internal Medicine IV, Saarland University Hospital, Homburg/Saar, Germany
| | - Stefan Blankenberg
- University Heart Centre Hamburg, Clinic for General and Interventional Cardiology, Hamburg, Germany; German Centre for Cardiovascular Research (DZHK e.V.), partner site Hamburg/Kiel/Lübeck, Germany
| | - Christoph Waldeyer
- University Heart Centre Hamburg, Clinic for General and Interventional Cardiology, Hamburg, Germany; German Centre for Cardiovascular Research (DZHK e.V.), partner site Hamburg/Kiel/Lübeck, Germany
| | - Christoph Sinning
- University Heart Centre Hamburg, Clinic for General and Interventional Cardiology, Hamburg, Germany; German Centre for Cardiovascular Research (DZHK e.V.), partner site Hamburg/Kiel/Lübeck, Germany
| | - Renate B Schnabel
- University Heart Centre Hamburg, Clinic for General and Interventional Cardiology, Hamburg, Germany; German Centre for Cardiovascular Research (DZHK e.V.), partner site Hamburg/Kiel/Lübeck, Germany
| | - Karl J Lackner
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Centre Mainz, Germany
| | | | - Ottar Nygård
- Department of Clinical Science, University of Bergen, Bergen, Norway; Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| | | | | | - Grethe S Tell
- Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway
| | - Juha Sinisalo
- Heart and Lung Centre, Helsinki University Hospital, Helsinki, Finland
| | - Markku S Nieminen
- Heart and Lung Centre, Helsinki University Hospital, Helsinki, Finland
| | - Reijo Laaksonen
- Medical School, Tampere University, Tampere, Finland; Finnish Clinical Biobank Tampere, University Hospital of Tampere, Tampere, Finland
| | - Stella Trompet
- Department of Geriatics and Gerontology, Leiden University Medical Centre, Leiden, Netherlands; Department of Cardiology, Leiden University Medical Centre, Leiden, Netherlands
| | - Roelof A J Smit
- Department of Geriatics and Gerontology, Leiden University Medical Centre, Leiden, Netherlands; Department of Cardiology, Leiden University Medical Centre, Leiden, Netherlands
| | - Naveed Sattar
- Institute of Cardiovascular and Medical Science, BHF Glasgow, Cardiovascular Research Centre, University of Glasgow, Glasgow, UK
| | - J Wouter Jukema
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Centre, Leiden, Netherlands; Interuniversity Cardiology Institute of the Netherlands, Utrecht, Netherlands
| | - Heinrich V Groesdonk
- Department of Anesthesiology, Intensive Care Medicine, and Pain Medicine, Saarland University Hospital, Homburg/Saar, Germany
| | - Graciela Delgado
- Fifth Department of Medicine, University Heidelberg, Mannheim, Germany
| | - Tatjana Stojakovic
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University Graz, Graz, Austria
| | - Anna P Pilbrow
- Christchurch Heart Institute, University of Otago, Christchurch, New Zealand
| | - Vicky A Cameron
- Christchurch Heart Institute, University of Otago, Christchurch, New Zealand
| | - A Mark Richards
- Christchurch Heart Institute, University of Otago, Christchurch, New Zealand; Cardiovascular Research Institute, National University of Singapore, Singapore
| | - Robert N Doughty
- Heart Health Research Group, University of Auckland, New Zealand
| | - Yan Gong
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics, Colleges of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Rhonda Cooper-DeHoff
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics, Colleges of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Julie Johnson
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics, Colleges of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Markus Scholz
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany; LIFE Research Centre for Civilisation Diseases, University of Leipzig, Leipzig, Germany
| | | | - Joachim Thiery
- LIFE Research Centre for Civilisation Diseases, University of Leipzig, Leipzig, Germany; Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital, Leipzig, Germany
| | - J Gustav Smith
- Department of Cardiology, Clinical Sciences, Lund University, Lund, Sweden; Skåne University Hospital, Lund, Sweden
| | - Ragnar O Vilmundarson
- Ruddy Canadian Cardiovascular Genetics Centre, University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Ruth McPherson
- Ruddy Canadian Cardiovascular Genetics Centre, University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Alexandre F R Stewart
- Ruddy Canadian Cardiovascular Genetics Centre, University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Sharon Cresci
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA; Department of Genetics, Washington University School of Medicine, Saint Louis, MO, USA
| | - Petra A Lenzini
- Statistical Genomics Division, Department of Genetics, Washington University School of Medicine, Saint Louis, MO, USA
| | - John A Spertus
- Saint Luke's Mid America Heart Institute, Kansas City, MO, USA; Department of Biomedical and Health Informatics, University of Missouri-Kansas City, Kansas City, MO, USA
| | | | | | | | - Andreas Leiherer
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria; Private University of the Principality of Liechtenstein, Triesen, Liechtenstein
| | - Christoph H Saely
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria; Private University of the Principality of Liechtenstein, Triesen, Liechtenstein
| | - Heinz Drexel
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria; Private University of the Principality of Liechtenstein, Triesen, Liechtenstein; Department of Medicine and Cardiology, Academic Teaching Hospital Feldkirch, Feldkirch, Austria; Drexel University College of Medicine, Philadelphia, PA, USA
| | - Axel Mündlein
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria
| | - Peter S Braund
- Department of Cardiovascular Sciences, University of Leicester, BHF Cardiovascular Research Centre, Glenfield Hospital, Leicester, UK; Leicester NIHR Biomedical Research Unit in Cardiovascular Disease, Glenfield Hospital, Leicester, UK
| | - Christopher P Nelson
- Department of Cardiovascular Sciences, University of Leicester, BHF Cardiovascular Research Centre, Glenfield Hospital, Leicester, UK; Leicester NIHR Biomedical Research Unit in Cardiovascular Disease, Glenfield Hospital, Leicester, UK
| | - Nilesh J Samani
- Department of Cardiovascular Sciences, University of Leicester, BHF Cardiovascular Research Centre, Glenfield Hospital, Leicester, UK; Leicester NIHR Biomedical Research Unit in Cardiovascular Disease, Glenfield Hospital, Leicester, UK
| | - Daniel Kofink
- Department of Cardiology, Heart and Lungs Division, UMC Utrecht, Utrecht, Netherlands
| | - Imo E Hoefer
- Laboratory of Experimental Cardiology, UMC Utrecht, Utrecht, Netherlands
| | - Gerard Pasterkamp
- Laboratory of Experimental Cardiology, UMC Utrecht, Utrecht, Netherlands
| | - Arshed A Quyyumi
- Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Yi-An Ko
- Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, GA, USA
| | | | | | | | | | - Niclas Eriksson
- Uppsala Clinical Research Centre, Uppsala University, Uppsala, Sweden; Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden
| | - Claes Held
- Uppsala Clinical Research Centre, Uppsala University, Uppsala, Sweden; Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden
| | - Emil Hagström
- Uppsala Clinical Research Centre, Uppsala University, Uppsala, Sweden; Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden
| | - Lars Wallentin
- Uppsala Clinical Research Centre, Uppsala University, Uppsala, Sweden; Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden
| | - Axel Åkerblom
- Uppsala Clinical Research Centre, Uppsala University, Uppsala, Sweden; Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden
| | - Agneta Siegbahn
- Uppsala Clinical Research Centre, Uppsala University, Uppsala, Sweden; Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden
| | - Igor Karp
- University of Montreal Hospital Research Centre (CRCHUM), University of Montreal, Montreal, QC, Canada; Department of Social and Preventive Medicine, University of Montreal, Montreal, QC, Canada; Department of Epidemiology and Biostatistics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | | | - Louise Pilote
- Department of Medicine, McGill University, Montreal, QC, Canada; Division of General Internal Medicine, McGill University Health Centre, Montreal, QC, Canada; Division of Clinical Epidemiology, McGill University Health Centre, Montreal, QC, Canada
| | - James C Engert
- Department of Medicine, McGill University, Montreal, QC, Canada
| | - James M Brophy
- Department of Medicine, McGill University, Montreal, QC, Canada
| | | | - Peter Bogaty
- Department of Medicine, Université Laval, QC, Canada
| | | | - Marcin Kaczor
- Jagielonian University Medical College, Kraków, Poland
| | - Marek Sanak
- Jagielonian University Medical College, Kraków, Poland
| | - Salim S Virani
- Section of Cardiology, Michael E DeBakey Veterans Affairs Medical Center, Baylor College of Medicine, Houston, TX, USA
| | - Christie M Ballantyne
- Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Vei-Vei Lee
- Department of Biostatistics 7, Epidemiology, Texas Heart Institute, Houston, TX, USA
| | - Eric Boerwinkle
- School of Public Health, University of Texas, Houston, TX, USA
| | - Michael V Holmes
- Medical Research Council Population Health Research Unit at the University of Oxford, University of Oxford, Oxford, UK; Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK; National Institute for Health Research Oxford Biomedical Research Centre, Oxford University Hospital, Oxford, UK
| | - Benjamin D Horne
- Intermountain Heart Institute, Intermountain Medical Center, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Aroon Hingorani
- Institute of Cardiovascular Science Facultyof Population Health Science, University College London, London, UK
| | - Folkert W Asselbergs
- Department of Cardiology, Heart and Lungs Division, UMC Utrecht, Utrecht, Netherlands; Institute of Cardiovascular Science Facultyof Population Health Science, University College London, London, UK; Durrer Centre of Cardiogenetic Research, ICIN-Netherlands Heart Institute, Utrecht, Netherlands
| | - Riyaz S Patel
- Institute of Cardiovascular Science Facultyof Population Health Science, University College London, London, UK
| | | | - Bernhard K Krämer
- Fifth Department of Medicine, University Heidelberg, Mannheim, Germany
| | - Hubert Scharnagl
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University Graz, Graz, Austria
| | - Danilo Fliser
- Department of Internal Medicine IV, Saarland University Hospital, Homburg/Saar, Germany
| | - Winfried März
- Fifth Department of Medicine, University Heidelberg, Mannheim, Germany; Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University Graz, Graz, Austria; Synlab Academy, Synlab Holding, Mannheim, Germany.
| | - Thimoteus Speer
- Department of Internal Medicine IV, Saarland University Hospital, Homburg/Saar, Germany
| |
Collapse
|
74
|
Maillet A, Desormais I, Rivière AB, Aboyans V, Lacroix P, Mirault T, Messas E, Bataille V, Constans J, Boulon C. Peripheral Atheromatous Arterial Disease in the Young: Risk Factors, Clinical Features, and Prognosis From the COPART Cohort. Angiology 2017; 68:893-898. [DOI: 10.1177/0003319717699501] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We describe the cardiovascular risk factors, clinical presentation, and prognosis in a comparative study of patients with peripheral artery disease (PAD) from the Cohorte des Patients ARTériopathes (COPART) cohort, which includes patients hospitalized for PAD in France. Among the 2514 patients included in the cohort, 189 had PAD before or at the age of 50 years and 2325 had it after. Young patients with PAD had diabetes less frequently (34% vs 46%, P < .001), were more frequent active smokers (58% vs 23%, P < 0.001), had lower high-density lipoprotein cholesterol (HDL-C; 41 ± 14 vs 44 ± 15 mg/dL, P = .026), and had a less frequent family and personal history of coronary heart disease. In a subset of 59 patients whose lipoprotein (a) (Lp(a)) was measured, the Lp(a) levels were higher in the young patients than in the older ones (89.7 mg/dL [9.7-151.3] vs 19.9 mg/dL [3.0-207.9], P = .004). Survival and amputation-free survival was 2.2 times higher (1.5-3.2, P < .001) in the young after 1 year. The onset of PAD before 50 years was associated with active smoking, low HDL-C, high Lp(a), and lower mortality.
Collapse
Affiliation(s)
- Alexandre Maillet
- Médecine vasculaire, Hôpital St Andre, Bordeaux, France
- Bordeaux University, Bordeaux, France
| | | | - Alessandra Bura Rivière
- Service de Médecine Vasculaire, CHU Toulouse, Toulouse, France
- StromaLab UMR 5273/INSERM U1031, Toulouse, France
- Université de Toulouse III, Toulouse, France
| | - Victor Aboyans
- Service de Cardiologie, Hôpital Universitaire Dupuytren, Limoges, France
| | | | - Tristan Mirault
- Department of Vascular Medicine, Hôpital Européen Georges-Pompidou, Assistance Publique Hôpitaux de Paris, APHP, Paris Descartes University, Sorbonne Paris Cite, Paris, France
| | - Emmanuel Messas
- Department of Vascular Medicine, Hôpital Européen Georges-Pompidou, Assistance Publique Hôpitaux de Paris, APHP, Paris Descartes University, Sorbonne Paris Cite, Paris, France
| | | | - Joël Constans
- Médecine vasculaire, Hôpital St Andre, Bordeaux, France
- Bordeaux University, Bordeaux, France
| | - Carine Boulon
- Médecine vasculaire, Hôpital St Andre, Bordeaux, France
- Bordeaux University, Bordeaux, France
| |
Collapse
|
75
|
Shan Y, Tromp G, Kuivaniemi H, Smelser DT, Verma SS, Ritchie MD, Elmore JR, Carey DJ, Conley YP, Gorin MB, Weeks DE. Genetic risk models: Influence of model size on risk estimates and precision. Genet Epidemiol 2017; 41:282-296. [PMID: 28198095 DOI: 10.1002/gepi.22035] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 11/08/2016] [Accepted: 12/01/2016] [Indexed: 12/11/2022]
Abstract
Disease risk estimation plays an important role in disease prevention. Many studies have found that the ability to predict risk improves as the number of risk single-nucleotide polymorphisms (SNPs) in the risk model increases. However, the width of the confidence interval of the risk estimate is often not considered in the evaluation of the risk model. Here, we explore how the risk and the confidence interval width change as more SNPs are added to the model in the order of decreasing effect size, using both simulated data and real data from studies of abdominal aortic aneurysms and age-related macular degeneration. Our results show that confidence interval width is positively correlated with model size and the majority of the bigger models have wider confidence interval widths than smaller models. Once the model size is bigger than a certain level, the risk does not shift markedly, as 100% of the risk estimates of the one-SNP-bigger models lie inside the confidence interval of the one-SNP-smaller models. We also created a confidence interval-augmented reclassification table. It shows that both more effective SNPs with larger odds ratios and less effective SNPs with smaller odds ratios contribute to the correct decision of whom to screen. The best screening strategy is selected and evaluated by the net benefit quantity and the reclassification rate. We suggest that individuals whose upper bound of their risk confidence interval is above the screening threshold, which corresponds to the population prevalence of the disease, should be screened.
Collapse
Affiliation(s)
- Ying Shan
- Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Gerard Tromp
- The Sigfried and Janet Weis Center for Research, Geisinger Health System, Danville, Pennsylvania, United States of America.,Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | - Helena Kuivaniemi
- The Sigfried and Janet Weis Center for Research, Geisinger Health System, Danville, Pennsylvania, United States of America.,Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | - Diane T Smelser
- The Sigfried and Janet Weis Center for Research, Geisinger Health System, Danville, Pennsylvania, United States of America
| | - Shefali S Verma
- Department of Biomedical and Translational Informatics, Geisinger Health System, Danville, Pennsylvania, United States of America
| | - Marylyn D Ritchie
- Department of Biomedical and Translational Informatics, Geisinger Health System, Danville, Pennsylvania, United States of America
| | - James R Elmore
- Department of Vascular and Endovascular Surgery, Geisinger Health System, Danville, PA
| | - David J Carey
- The Sigfried and Janet Weis Center for Research, Geisinger Health System, Danville, Pennsylvania, United States of America
| | - Yvette P Conley
- Department of Health Promotion and Development, School of Nursing, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America.,Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Michael B Gorin
- Departments of Ophthalmology and Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America.,Stein Eye Institute, Los Angeles, California, United States of America
| | - Daniel E Weeks
- Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America.,Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| |
Collapse
|
76
|
Emdin CA, Khera AV, Natarajan P, Klarin D, Won HH, Peloso GM, Stitziel NO, Nomura A, Zekavat SM, Bick AG, Gupta N, Asselta R, Duga S, Merlini PA, Correa A, Kessler T, Wilson JG, Bown MJ, Hall AS, Braund PS, Samani NJ, Schunkert H, Marrugat J, Elosua R, McPherson R, Farrall M, Watkins H, Willer C, Abecasis GR, Felix JF, Vasan RS, Lander E, Rader DJ, Danesh J, Ardissino D, Gabriel S, Saleheen D, Kathiresan S. Phenotypic Characterization of Genetically Lowered Human Lipoprotein(a) Levels. J Am Coll Cardiol 2016; 68:2761-2772. [PMID: 28007139 PMCID: PMC5328146 DOI: 10.1016/j.jacc.2016.10.033] [Citation(s) in RCA: 170] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 09/21/2016] [Accepted: 10/04/2016] [Indexed: 02/02/2023]
Abstract
BACKGROUND Genomic analyses have suggested that the LPA gene and its associated plasma biomarker, lipoprotein(a) (Lp[a]), represent a causal risk factor for coronary heart disease (CHD). As such, lowering Lp(a) levels has emerged as a therapeutic strategy. Beyond target identification, human genetics may contribute to the development of new therapies by defining the full spectrum of beneficial and adverse consequences and by developing a dose-response curve of target perturbation. OBJECTIVES The goal of this study was to establish the full phenotypic impact of LPA gene variation and to estimate a dose-response curve between genetically altered plasma Lp(a) and risk for CHD. METHODS We leveraged genetic variants at the LPA gene from 3 data sources: individual-level data from 112,338 participants in the U.K. Biobank; summary association results from large-scale genome-wide association studies; and LPA gene sequencing results from case subjects with CHD and control subjects free of CHD. RESULTS One SD genetically lowered Lp(a) level was associated with a 29% lower risk of CHD (odds ratio [OR]: 0.71; 95% confidence interval [CI]: 0.69 to 0.73), a 31% lower risk of peripheral vascular disease (OR: 0.69; 95% CI: 0.59 to 0.80), a 13% lower risk of stroke (OR: 0.87; 95% CI: 0.79 to 0.96), a 17% lower risk of heart failure (OR: 0.83; 95% CI: 0.73 to 0.94), and a 37% lower risk of aortic stenosis (OR: 0.63; 95% CI: 0.47 to 0.83). We observed no association with 31 other disorders, including type 2 diabetes and cancer. Variants that led to gain of LPA gene function increased the risk for CHD, whereas those that led to loss of gene function reduced the CHD risk. CONCLUSIONS Beyond CHD, genetically lowered Lp(a) levels are associated with a lower risk of peripheral vascular disease, stroke, heart failure, and aortic stenosis. As such, pharmacological lowering of plasma Lp(a) may influence a range of atherosclerosis-related diseases.
Collapse
Affiliation(s)
- Connor A Emdin
- Center for Human Genetic Research, Cardiovascular Research Center and Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts
| | - Amit V Khera
- Center for Human Genetic Research, Cardiovascular Research Center and Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts
| | - Pradeep Natarajan
- Center for Human Genetic Research, Cardiovascular Research Center and Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts
| | - Derek Klarin
- Center for Human Genetic Research, Cardiovascular Research Center and Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts
| | - Hong-Hee Won
- Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Samsung Medical Center, Seoul, Republic of Korea
| | - Gina M Peloso
- Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts; Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts
| | - Nathan O Stitziel
- Departments of Medicine, Genetics, and the McDonnell Genome Institute, Washington University School of Medicine, St. Louis, Missouri
| | - Akihiro Nomura
- Center for Human Genetic Research, Cardiovascular Research Center and Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts
| | - Seyedeh M Zekavat
- Center for Human Genetic Research, Cardiovascular Research Center and Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts
| | - Alexander G Bick
- Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts
| | - Namrata Gupta
- Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts
| | - Rosanna Asselta
- Department of Biomedical Sciences, Humanitas University and Humanitas Clinical and Research Center, Milan, Italy
| | - Stefano Duga
- Department of Biomedical Sciences, Humanitas University and Humanitas Clinical and Research Center, Milan, Italy
| | | | - Adolfo Correa
- Jackson Heart Study, Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| | - Thorsten Kessler
- Deutsches Herzzentrum München, Technische Universität München, Deutsches Zentrum für Herz-Kreislauf-Forschung, München, Germany; Munich Heart Alliance, München, Germany
| | - James G Wilson
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi
| | - Matthew J Bown
- Departments of Cardiovascular Sciences and NIHR Leicester Cardiovascular Biomedical Research Unit, University of Leicester, Leicester, United Kingdom
| | - Alistair S Hall
- Leeds Institute of Cardiovascular and Metabolic Medicine, Leeds University, Leeds, United Kingdom
| | - Peter S Braund
- Departments of Cardiovascular Sciences and NIHR Leicester Cardiovascular Biomedical Research Unit, University of Leicester, Leicester, United Kingdom
| | - Nilesh J Samani
- Departments of Cardiovascular Sciences and NIHR Leicester Cardiovascular Biomedical Research Unit, University of Leicester, Leicester, United Kingdom
| | - Heribert Schunkert
- Deutsches Herzzentrum München, Technische Universität München, Deutsches Zentrum für Herz-Kreislauf-Forschung, München, Germany
| | - Jaume Marrugat
- Cardiovascular Epidemiology & Genetics, IMIM (Hospital del Mar Research Institute), Barcelona, Spain
| | - Roberto Elosua
- Cardiovascular Epidemiology & Genetics, IMIM (Hospital del Mar Research Institute), Barcelona, Spain
| | - Ruth McPherson
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Martin Farrall
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and the Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Hugh Watkins
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and the Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Cristen Willer
- Department of Computational Medicine and Bioinformatics, Department of Human Genetics, and Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Gonçalo R Abecasis
- Center for Statistical Genetics, Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, Michigan
| | - Janine F Felix
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Ramachandran S Vasan
- National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, Framingham, Massachusetts; Sections of Cardiology, Preventive Medicine and Epidemiology, Department of Medicine, Boston University Schools of Medicine and Public Health, Boston, Massachusetts
| | - Eric Lander
- Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts
| | - Daniel J Rader
- Department of Genetics, University of Pennsylvania, Philadelphia, Pennsylvania
| | - John Danesh
- Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom; Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, United Kingdom; National Institute for Health Research Blood and Transplant Research Unit in Donor Health and Genomics, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Diego Ardissino
- Division of Cardiology, Azienda Ospedaliero-Universitaria di Parma, University of Parma, Parma, Italy; ASTC: Associazione per lo Studio Della Trombosi in Cardiologia, Pavia, Italy
| | - Stacey Gabriel
- Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts
| | - Danish Saleheen
- Biostatistics and Epidemiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sekar Kathiresan
- Center for Human Genetic Research, Cardiovascular Research Center and Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts.
| |
Collapse
|
77
|
Tybjærg-Hansen A. Using Human Genetics to Predict the Effects and Side Effects of Lipoprotein(a) Lowering Drugs. J Am Coll Cardiol 2016; 68:2773-2775. [PMID: 28007140 DOI: 10.1016/j.jacc.2016.10.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 10/25/2016] [Indexed: 10/20/2022]
Affiliation(s)
- Anne Tybjærg-Hansen
- Department of Clinical Biochemistry, Section for Molecular Genetics, Rigshospitalet, Copenhagen University Hospital, and Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
78
|
Abstract
Lipoprotein(a) [Lp(a)] is a highly atherogenic lipoprotein that is under strong genetic control by the LPA gene locus. Genetic variants including a highly polymorphic copy number variation of the so called kringle IV repeats at this locus have a pronounced influence on Lp(a) concentrations. High concentrations of Lp(a) as well as genetic variants which are associated with high Lp(a) concentrations are both associated with cardiovascular disease which very strongly supports causality between Lp(a) concetrations and cardiovascular disease. This method of using a genetic variant that has a pronounced influence on a biomarker to support causality with an outcome is called Mendelian randomization approach and was applied for the first time two decades ago with data from Lp(a) and cardiovascular disease. This approach was also used to demonstrate a causal association between high Lp(a) concentrations and aortic valve stenosis, between low concentrations and type-2 diabetes mellitus and to exclude a causal association between Lp(a) concentrations and venous thrombosis. Considering the high frequency of these genetic variants in the population makes Lp(a) the strongest genetic risk factor for cardiovascular disease identified so far. Promising drugs that lower Lp(a) are on the horizon but their efficacy in terms of reducing clinical outcomes still has to be shown.
Collapse
|
79
|
Beheshtian A, Shitole SG, Segal AZ, Leifer D, Tracy RP, Rader DJ, Devereux RB, Kizer JR. Lipoprotein (a) level, apolipoprotein (a) size, and risk of unexplained ischemic stroke in young and middle-aged adults. Atherosclerosis 2016; 253:47-53. [PMID: 27575936 PMCID: PMC5181117 DOI: 10.1016/j.atherosclerosis.2016.08.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 08/02/2016] [Accepted: 08/18/2016] [Indexed: 12/21/2022]
Abstract
BACKGROUND AND AIMS Circulating lipoprotein (a) [Lp(a)] level relates inversely to apolipoprotein (a) [apo(a)] size. Both smaller apo(a) isoforms and higher Lp(a) levels have been linked to coronary heart disease and stroke, but their independent contributions are less well defined. We examined the role of Lp(a) in younger adults with cryptogenic stroke. METHODS Lp(a) and apo(a) isoforms were evaluated in a prospectively designed case-control study of patients with unexplained ischemic stroke and stroke-free controls, ages 18 to 64. Serum Lp(a) was measured among 255 cases and 390 controls with both apo(a)-size independent and dependent assays. Apo(a) size was determined by agarose gel electrophoresis. RESULTS Cases and controls were similar in socio-demographic characteristics, but cases had more hypertension, diabetes, smoking, and migraine with aura. In race-specific analyses, Lp(a) levels showed positive associations with cryptogenic stroke in whites, but not in the smaller subgroups of blacks and Hispanics. After full adjustment, comparison of the highest versus lowest quartile in whites was significant for apo(a)-size-independent (OR = 2.10 [95% CI = 1.04, 4.27], p = 0.040), and near-significant for apo(a)-size-dependent Lp(a) (OR = 1.81 [95% CI = 0.95, 3.47], p = 0.073). Apo(a) size was not associated with cryptogenic stroke in any race-ethnic subgroup. CONCLUSIONS This study underscores the importance of Lp(a) level, but not apo(a) size, as an independent risk factor for unexplained ischemic stroke in young and middle-aged white adults. Given the emergence of effective Lp(a)-lowering therapies, these findings support routine testing for Lp(a) in this setting, along with further research to assess the extent to which such therapies improve outcomes in this population.
Collapse
Affiliation(s)
- Azadeh Beheshtian
- Department of Medicine, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
| | - Sanyog G Shitole
- Department of Medicine, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
| | - Alan Z Segal
- Department of Neurology, Weill Cornell Medicine, New York, NY, USA
| | - Dana Leifer
- Department of Neurology, Weill Cornell Medicine, New York, NY, USA
| | - Russell P Tracy
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT, USA
| | - Daniel J Rader
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | | | - Jorge R Kizer
- Department of Medicine, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA; Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
80
|
Kotani K, Sahebkar A, Serban MC, Ursoniu S, Mikhailidis DP, Mariscalco G, Jones SR, Martin S, Blaha MJ, Toth PP, Rizzo M, Kostner K, Rysz J, Banach M. Lipoprotein(a) Levels in Patients With Abdominal Aortic Aneurysm. Angiology 2016; 68:99-108. [PMID: 26980774 DOI: 10.1177/0003319716637792] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Circulating markers relevant to the development of abdominal aortic aneurysm (AAA) are currently required. Lipoprotein(a), Lp(a), is considered a candidate marker associated with the presence of AAA. The present meta-analysis aimed to evaluate the association between circulating Lp(a) levels and the presence of AAA. The PubMed-based search was conducted up to April 30, 2015, to identify the studies focusing on Lp(a) levels in patients with AAA and controls. Quantitative data synthesis was performed using a random effects model, with standardized mean difference (SMD) and 95% confidence interval (CI) as summary statistics. Overall, 9 studies were identified. After a combined analysis, patients with AAA were found to have a significantly higher level of Lp(a) compared to the controls (SMD: 0.87, 95% CI: 0.41-1.33, P < .001). This result remained robust in the sensitivity analysis, and its significance was not influenced after omitting each of the included studies from the meta-analysis. The present meta-analysis confirmed a higher level of circulating Lp(a) in patients with AAA compared to controls. High Lp(a) levels can be associated with the presence of AAA, and Lp(a) may be a marker in screening for AAA. Further studies are needed to establish the clinical utility of measuring Lp(a) in the prevention and management of AAA.
Collapse
Affiliation(s)
- Kazuhiko Kotani
- 1 Division of Community and Family Medicine, Jichi Medical University, Shimotsuke-City, Japan
| | - Amirhossein Sahebkar
- 2 Biotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,3 Metabolic Research Centre, Royal Perth Hospital, School of Medicine and Pharmacology, University of Western Australia, Perth, Australia
| | - Maria-Corina Serban
- 4 Discipline of Pathophysiology, Department of Functional Sciences, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania
| | - Sorin Ursoniu
- 5 Discipline of Public Health, Department of Functional Sciences, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania
| | - Dimitri P Mikhailidis
- 6 Department of Clinical Biochemistry, Royal Free Campus, University College London Medical School, University College London, London, United Kingdom
| | - Giovanni Mariscalco
- 7 Department of Cardiovascular Sciences, University of Leicester Glenfield Hospital, Leicester, United Kingdom
| | - Steven R Jones
- 8 The Johns Hopkins Ciccarone Center for the Prevention of Heart Disease, Baltimore, MD, USA
| | - Seth Martin
- 8 The Johns Hopkins Ciccarone Center for the Prevention of Heart Disease, Baltimore, MD, USA
| | - Michael J Blaha
- 8 The Johns Hopkins Ciccarone Center for the Prevention of Heart Disease, Baltimore, MD, USA
| | - Peter P Toth
- 8 The Johns Hopkins Ciccarone Center for the Prevention of Heart Disease, Baltimore, MD, USA.,9 Preventive Cardiology, CGH Medical Center, Sterling, IL, USA
| | - Manfredi Rizzo
- 10 Biomedical Department of Internal Medicine and Medical Specialties, University of Palermo, Italy
| | - Karam Kostner
- 11 Mater Hospital, University of Queensland, St Lucia, Australia
| | - Jacek Rysz
- 12 Department of Hypertension, Nephrology and Hypertension, WAM University Hospital in Lodz, Medical University of Lodz, Poland
| | - Maciej Banach
- 12 Department of Hypertension, Nephrology and Hypertension, WAM University Hospital in Lodz, Medical University of Lodz, Poland
| | | |
Collapse
|
81
|
Nordestgaard BG, Langsted A. Lipoprotein (a) as a cause of cardiovascular disease: insights from epidemiology, genetics, and biology. J Lipid Res 2016; 57:1953-1975. [PMID: 27677946 DOI: 10.1194/jlr.r071233] [Citation(s) in RCA: 366] [Impact Index Per Article: 40.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Indexed: 12/24/2022] Open
Abstract
Human epidemiologic and genetic evidence using the Mendelian randomization approach in large-scale studies now strongly supports that elevated lipoprotein (a) [Lp(a)] is a causal risk factor for cardiovascular disease, that is, for myocardial infarction, atherosclerotic stenosis, and aortic valve stenosis. The Mendelian randomization approach used to infer causality is generally not affected by confounding and reverse causation, the major problems of observational epidemiology. This approach is particularly valuable to study causality of Lp(a), as single genetic variants exist that explain 27-28% of all variation in plasma Lp(a). The most important genetic variant likely is the kringle IV type 2 (KIV-2) copy number variant, as the apo(a) product of this variant influences fibrinolysis and thereby thrombosis, as opposed to the Lp(a) particle per se. We speculate that the physiological role of KIV-2 in Lp(a) could be through wound healing during childbirth, infections, and injury, a role that, in addition, could lead to more blood clots promoting stenosis of arteries and the aortic valve, and myocardial infarction. Randomized placebo-controlled trials of Lp(a) reduction in individuals with very high concentrations to reduce cardiovascular disease are awaited. Recent genetic evidence documents elevated Lp(a) as a cause of myocardial infarction, atherosclerotic stenosis, and aortic valve stenosis.
Collapse
Affiliation(s)
- Børge G Nordestgaard
- Department of Clinical Biochemistry and Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark; and Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anne Langsted
- Department of Clinical Biochemistry and Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark; and Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
82
|
Arnar DO, Andersen K, Thorgeirsson G. Genetics of cardiovascular diseases: lessons learned from a decade of genomics research in Iceland. SCAND CARDIOVASC J 2016; 50:260-265. [DOI: 10.1080/14017431.2016.1230679] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- David O. Arnar
- Department of Medicine, Division of Cardiology, Landspitali – The National University Hospital of Iceland, Reykjavik, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
- deCODE Genetics, Reykjavik, Iceland
| | - Karl Andersen
- Department of Medicine, Division of Cardiology, Landspitali – The National University Hospital of Iceland, Reykjavik, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Gudmundur Thorgeirsson
- Department of Medicine, Division of Cardiology, Landspitali – The National University Hospital of Iceland, Reykjavik, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| |
Collapse
|
83
|
Relation of Plasma Lipoprotein(a) to Subclinical Coronary Plaque Volumes, Three-Vessel and Left Main Coronary Disease, and Severe Coronary Stenoses in Apparently Healthy African-Americans With a Family History of Early-Onset Coronary Artery Disease. Am J Cardiol 2016; 118:656-61. [PMID: 27530333 DOI: 10.1016/j.amjcard.2016.06.020] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 06/03/2016] [Accepted: 06/03/2016] [Indexed: 11/24/2022]
Abstract
Serum lipoprotein(a) [Lp(a)] is a coronary artery disease (CAD) risk factor in persons of European ancestry. Levels are twofold to threefold higher in African-Americans (AAs), but reported associations with CAD have been inconsistent. The relation of Lp(a) with the extent and severity of subclinical coronary plaque has not been described in AAs. We screened 269 apparently healthy AAs for risk factors and coronary plaque using advanced coronary computed tomographic angiography. Total coronary plaque (TCP), noncalcified coronary plaque, and calcified coronary plaque volumes (mm(3)) were quantified using a validated automated method. Lp(a) was measured by ELISA. Multivariable modeling was performed with adjustment for traditional CAD risk factors and intrafamilial correlations. Mean age was 51 ± 11 years and 64% were female. Plaque was present in 41%. Lp(a) was independently associated with TCP volume [log(TCP + 1)] (p = 0.04), 3-vessel and/or left main involvement (p = 0.04), and at least 1 stenosis >50% (p = 0.006). Best-fit regression analyses showed that subjects with Lp(a) >40 mg/dl were threefold more likely to have 3-vessel and/or left main disease (95% confidence interval 1.4 to 6.8, p = 0.005) and fourfold more likely to have stenosis >50% (95% confidence interval 1.3 to 15.0, p = 0.02). In subjects with plaque (n = 110), multivariable models showed the Lp(a) level was significantly and independently associated with TCP (p = 0.009), noncalcified coronary plaque (p = 0.01), and calcified coronary plaque (p = 0.003) and affected vessel length (p = 0.01). In conclusion, high Lp(a) is strongly associated with coronary plaque volumes, extent, and severity in apparently healthy AAs. High levels of Lp(a) may be particularly important in the pathogenesis of CAD in AAs.
Collapse
|
84
|
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
|
85
|
Wang L, Chen J, Zeng Y, Wei J, Jing J, Li G, Su L, Tang X, Wu T, Zhou L. Functional Variant in the SLC22A3-LPAL2-LPA Gene Cluster Contributes to the Severity of Coronary Artery Disease. Arterioscler Thromb Vasc Biol 2016; 36:1989-96. [PMID: 27417586 DOI: 10.1161/atvbaha.116.307311] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 06/29/2016] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Recent genome-wide association studies have identified that genetic variants in the SLC22A3-LPAL2-LPA gene cluster influence plasma lipoprotein(a) [Lp(a)] concentration. However, the association between this gene cluster and the severity of coronary artery disease (CAD), especially the potential underlying mechanism, remains unclear. The purpose of this study was to investigate the association between variation in the SLC22A3-LPAL2-LPA gene cluster and CAD. APPROACH AND RESULTS We performed 2-stage case-control studies in a Chinese Han population. The variant genotypes were examined for their association with both Lp(a) level and severity of CAD. Putative mechanisms were also evaluated. One single nucleotide polymorphism, rs3088442, in the SLC22A3-LPAL2-LPA gene cluster was significantly associated with both plasma Lp(a) levels and CAD severity. The gene dosage of the risk allele at rs3088442 indicated a robust association with left main trunk disease (P=0.046), number of vascular lesions (P=4.5×10(-3)), and Gensini scores (P=0.012) in patients with CAD. Reporter gene analysis indicated that the rs3088442 G allele might suppress miR-147a binding to the 3' untranslated region of SLC22A3, resulting in altered SLC22A3 and LPA gene expression (P=0.015 and 9.2×10(-6), respectively), possibly explaining the increased plasma Lp(a) levels and risk of CAD. CONCLUSIONS The genotype of rs3088442 within the SLC22A3-LPAL2-LPA gene cluster may contribute to regulation of plasma Lp(a) levels and possibly to the severity of CAD in a Chinese Han population.
Collapse
Affiliation(s)
- Long Wang
- From the Department of Epidemiology, Research Center for Medicine and Social Development, School of Public Health and Management (L.W., J.W., G.L., X.T., L.Z.); the Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital (J.C.) and the Department of Cardiology, the Second Affiliated Hospital and the Chongqing Cardiac Arrhythmias Service Center (J.J., L.S.), Chongqing Medical University, Chongqing, China; the Institute of Cardiovascular Diseases of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing, China (Y.Z.); and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei, China (T.W.)
| | - Juan Chen
- From the Department of Epidemiology, Research Center for Medicine and Social Development, School of Public Health and Management (L.W., J.W., G.L., X.T., L.Z.); the Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital (J.C.) and the Department of Cardiology, the Second Affiliated Hospital and the Chongqing Cardiac Arrhythmias Service Center (J.J., L.S.), Chongqing Medical University, Chongqing, China; the Institute of Cardiovascular Diseases of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing, China (Y.Z.); and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei, China (T.W.)
| | - Ying Zeng
- From the Department of Epidemiology, Research Center for Medicine and Social Development, School of Public Health and Management (L.W., J.W., G.L., X.T., L.Z.); the Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital (J.C.) and the Department of Cardiology, the Second Affiliated Hospital and the Chongqing Cardiac Arrhythmias Service Center (J.J., L.S.), Chongqing Medical University, Chongqing, China; the Institute of Cardiovascular Diseases of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing, China (Y.Z.); and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei, China (T.W.)
| | - Jie Wei
- From the Department of Epidemiology, Research Center for Medicine and Social Development, School of Public Health and Management (L.W., J.W., G.L., X.T., L.Z.); the Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital (J.C.) and the Department of Cardiology, the Second Affiliated Hospital and the Chongqing Cardiac Arrhythmias Service Center (J.J., L.S.), Chongqing Medical University, Chongqing, China; the Institute of Cardiovascular Diseases of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing, China (Y.Z.); and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei, China (T.W.)
| | - Jinjin Jing
- From the Department of Epidemiology, Research Center for Medicine and Social Development, School of Public Health and Management (L.W., J.W., G.L., X.T., L.Z.); the Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital (J.C.) and the Department of Cardiology, the Second Affiliated Hospital and the Chongqing Cardiac Arrhythmias Service Center (J.J., L.S.), Chongqing Medical University, Chongqing, China; the Institute of Cardiovascular Diseases of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing, China (Y.Z.); and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei, China (T.W.)
| | - Ge Li
- From the Department of Epidemiology, Research Center for Medicine and Social Development, School of Public Health and Management (L.W., J.W., G.L., X.T., L.Z.); the Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital (J.C.) and the Department of Cardiology, the Second Affiliated Hospital and the Chongqing Cardiac Arrhythmias Service Center (J.J., L.S.), Chongqing Medical University, Chongqing, China; the Institute of Cardiovascular Diseases of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing, China (Y.Z.); and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei, China (T.W.)
| | - Li Su
- From the Department of Epidemiology, Research Center for Medicine and Social Development, School of Public Health and Management (L.W., J.W., G.L., X.T., L.Z.); the Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital (J.C.) and the Department of Cardiology, the Second Affiliated Hospital and the Chongqing Cardiac Arrhythmias Service Center (J.J., L.S.), Chongqing Medical University, Chongqing, China; the Institute of Cardiovascular Diseases of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing, China (Y.Z.); and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei, China (T.W.)
| | - Xiaojun Tang
- From the Department of Epidemiology, Research Center for Medicine and Social Development, School of Public Health and Management (L.W., J.W., G.L., X.T., L.Z.); the Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital (J.C.) and the Department of Cardiology, the Second Affiliated Hospital and the Chongqing Cardiac Arrhythmias Service Center (J.J., L.S.), Chongqing Medical University, Chongqing, China; the Institute of Cardiovascular Diseases of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing, China (Y.Z.); and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei, China (T.W.)
| | - Tangchun Wu
- From the Department of Epidemiology, Research Center for Medicine and Social Development, School of Public Health and Management (L.W., J.W., G.L., X.T., L.Z.); the Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital (J.C.) and the Department of Cardiology, the Second Affiliated Hospital and the Chongqing Cardiac Arrhythmias Service Center (J.J., L.S.), Chongqing Medical University, Chongqing, China; the Institute of Cardiovascular Diseases of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing, China (Y.Z.); and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei, China (T.W.)
| | - Li Zhou
- From the Department of Epidemiology, Research Center for Medicine and Social Development, School of Public Health and Management (L.W., J.W., G.L., X.T., L.Z.); the Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital (J.C.) and the Department of Cardiology, the Second Affiliated Hospital and the Chongqing Cardiac Arrhythmias Service Center (J.J., L.S.), Chongqing Medical University, Chongqing, China; the Institute of Cardiovascular Diseases of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing, China (Y.Z.); and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei, China (T.W.).
| |
Collapse
|
86
|
Affiliation(s)
- Aris T. Alexandrou
- Division of Cardiovascular Medicine, Sulpizio Cardiovascular Center, University of California, San Diego, La Jolla, CA, USA
| | - Sotirios Tsimikas
- Division of Cardiovascular Medicine, Sulpizio Cardiovascular Center, University of California, San Diego, La Jolla, CA, USA
- Vascular Medicine Program, Department of Medicine, Sulpizio Cardiovascular Center, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
87
|
Kotani K, Serban MC, Penson P, Lippi G, Banach M. Evidence-based assessment of lipoprotein(a) as a risk biomarker for cardiovascular diseases - Some answers and still many questions. Crit Rev Clin Lab Sci 2016; 53:370-8. [PMID: 27173621 DOI: 10.1080/10408363.2016.1188055] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The present article is aimed at outlining the current state of knowledge regarding the clinical value of lipoprotein(a) (Lp(a)) as a marker of cardiovascular disease (CVD) risk by summarizing the results of recent clinical studies, meta-analyses and systematic reviews. The literature supports the predictive value of Lp(a) on CVD outcomes, although the effect size is modest. Lp(a) would also appear to have an effect on cerebrovascular outcomes, however the effect appears even smaller than that for CVD outcomes. Consideration of apolipoprotein(a) (apo(a)) isoforms and LPA genetics in relation to the simple assessment of Lp(a) concentration may enhance clinical practice in vascular medicine. We also describe recent advances in Lp(a) research (including therapies) and highlight areas where further research is needed such as the measurement of Lp(a) and its involvement in additional pathophysiological processes.
Collapse
Affiliation(s)
- Kazuhiko Kotani
- a Division of Community and Family MedicinevJichi Medical University , Shimotsuke-City , Japan .,b Department of Clinical Laboratory Medicine , Jichi Medical University , Shimotsuke-City , Japan
| | - Maria-Corina Serban
- c Department of Epidemiology , University of Alabama at Birmingham , Birmingham , AL , USA .,d Department of Functional Sciences , Discipline of Pathophysiology, "Victor Babes" University of Medicine and Pharmacy , Timisoara , Romania
| | - Peter Penson
- e Section of Clinical Biochemistry , School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University , Liverpool , UK
| | - Giuseppe Lippi
- f Section of Clinical Biochemistry , University of Verona , Verona , Italy , and
| | - Maciej Banach
- g Department of Hypertension , Chair of Nephrology and Hypertension, Medical University of Lodz , Lodz , Poland
| |
Collapse
|
88
|
Andriolo RB, Ponte Jr JR, Gomes Gutierrez E, Andriolo BNG, Ramos LR. Glycaemic control for patients with acute coronary syndrome. Hippokratia 2016. [DOI: 10.1002/14651858.cd010811.pub2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Regis B Andriolo
- Universidade do Estado do Pará; Department of Public Health; Travessa Perebebuí, 2623 Belém Pará Brazil 66087-670
| | | | - Erickson Gomes Gutierrez
- Universidade Federal do Para; Department of Medicine; Avenida Governador Jose Malchar 1192 Belem Para Brazil 660055260
| | - Brenda NG Andriolo
- Centro de Estudos de Saúde Baseada em Evidências e Avaliação Tecnológica em Saúde; Brazilian Cochrane Centre; Rua Borges Lagoa, 564 cj 63 São Paulo São Paulo Brazil 04038-000
| | - Luiz Roberto Ramos
- Escola Paulista de Medicina, Universidade Federal de São Paulo; Department of Preventive Medicine; Rua dos Otonis, 731 Vila Clementino São Paulo São Paulo Brazil 04025-002
| |
Collapse
|
89
|
Freudenberg-Hua Y, Li W, Abhyankar A, Vacic V, Cortes V, Ben-Avraham D, Koppel J, Greenwald B, Germer S, Darnell RB, Barzilai N, Freudenberg J, Atzmon G, Davies P. Differential burden of rare protein truncating variants in Alzheimer's disease patients compared to centenarians. Hum Mol Genet 2016; 25:3096-3105. [PMID: 27260402 DOI: 10.1093/hmg/ddw150] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 05/06/2016] [Accepted: 05/13/2016] [Indexed: 12/31/2022] Open
Abstract
We compared coding region variants of 53 cognitively healthy centenarians and 45 patients with Alzheimer's disease (AD), all of Ashkenazi Jewish (AJ) ancestry. Despite the small sample size, the known AD risk variant APOE4 reached genome-wide significance, indicating the advantage of utilizing 'super-controls'. We restricted our subsequent analysis to rare variants observed at most once in the 1000 Genomes database and having a minor allele frequency below 2% in our AJ sample. We compared the burden of predicted protein altering variants between cases and controls as normalized by the level of rare synonymous variants. We observed an increased burden among AD subjects for predicted loss-of-function (LoFs) variants defined as stop-gain, frame shift, initiation codon (INIT) and splice site mutations (n = 930, OR = 1.3, P = 1.5×E-5). There was no enrichment across all rare protein altering variants defined as missense plus LoFs, in frame indels and stop-loss variants (n = 13 014, OR = 0.97, P = 0.47). Among LoFs, the strongest burden was observed for INIT (OR = 2.16, P = 0.0097) and premature stop variants predicted to cause non-sense-mediated decay in the majority of transcripts (NMD) (OR = 1.98, P = 0.02). Notably, this increased burden of NMD, INIT and splice variants was more pronounced in a set of 1397 innate immune genes (OR = 4.55, P = 0.0043). Further comparison to additional exomes indicates that the difference in LoF burden originated both from the AD and centenarian sample. In summary, we observed an overall increased burden of rare LoFs in AD subjects as compared to centenarians, and this enrichment is more pronounced for innate immune genes.
Collapse
Affiliation(s)
- Yun Freudenberg-Hua
- Litwin-Zucker Research Center for the Study of Alzheimer's Disease, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY 11030, USA .,Division of Geriatric Psychiatry, Zucker Hillside Hospital, Northwell Health, Glen Oaks, NY 11004, USA
| | - Wentian Li
- Robert S Boas Center for Genomics and Human Genetics, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY 11030, USA
| | | | | | - Vanessa Cortes
- Litwin-Zucker Research Center for the Study of Alzheimer's Disease, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY 11030, USA
| | - Danny Ben-Avraham
- Institute for Aging Research and the Diabetes Research Center, Department of Medicine and Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Jeremy Koppel
- Litwin-Zucker Research Center for the Study of Alzheimer's Disease, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY 11030, USA
| | - Blaine Greenwald
- Division of Geriatric Psychiatry, Zucker Hillside Hospital, Northwell Health, Glen Oaks, NY 11004, USA
| | | | | | | | - Nir Barzilai
- Institute for Aging Research and the Diabetes Research Center, Department of Medicine and Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Jan Freudenberg
- Robert S Boas Center for Genomics and Human Genetics, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY 11030, USA.,The Regeneron Genetics Center, Tarrytown, NY 10591, USA and
| | - Gil Atzmon
- Institute for Aging Research and the Diabetes Research Center, Department of Medicine and Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA.,Faculty of Natural Sciences, University of Haifa, Haifa 31905, Israel
| | - Peter Davies
- Litwin-Zucker Research Center for the Study of Alzheimer's Disease, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY 11030, USA
| |
Collapse
|
90
|
Hopewell JC, Clarke R. Emerging Risk Factors for Stroke. Stroke 2016; 47:1673-8. [DOI: 10.1161/strokeaha.115.010646] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 03/14/2016] [Indexed: 12/18/2022]
Affiliation(s)
- Jemma C. Hopewell
- From the Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Robert Clarke
- From the Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
91
|
Yoshino S, Cilluffo R, Prasad M, Best PJM, Atkinson EJ, Aoki T, Cunningham JM, de Andrade M, Lerman LO, Lerman A. Sex-Specific Genetic Variants are Associated With Coronary Endothelial Dysfunction. J Am Heart Assoc 2016; 5:e002544. [PMID: 27091178 PMCID: PMC4859270 DOI: 10.1161/jaha.115.002544] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Background Endothelial dysfunction is an early stage of atherosclerosis. Single‐nucleotide polymorphisms (SNPs) have been associated with vascular dysfunction, cardiac events, and coronary artery remodeling. We aimed to detect SNPs associated with endothelial dysfunction and determine whether these associations are sex specific. Methods and Results Six hundred forty‐three subjects without significant obstructive coronary artery disease underwent invasive coronary endothelial function assessment. We collected data from 1536 SNPs that had previously been associated with vasoreactivity, angiogenesis, inflammation, artery calcification, atherosclerotic risk factors, insulin resistance, hormone levels, blood coagulability, or with coronary heart disease. Coronary vascular reactivity was assessed by the percent change in coronary artery diameter ≤ −20% after an intracoronary bolus injection of acetylcholine on invasive coronary physiology study. SNPs significantly associated with coronary epicardial endothelial dysfunction were ADORA1,KCNQ1, and DNAJC4 in the whole cohort, LPA, MYBPH, ADORA3, and PON1 in women and KIF6 and NFKB1 in men (P<0.01). Conclusions We have identified several significant SNPs that are associated with an increased risk of coronary endothelial dysfunction. These associations appear to be sex specific and may explain gender‐related differences in development of atherosclerosis.
Collapse
Affiliation(s)
- Satoshi Yoshino
- Cardiovascular Medicine and Hypertension, Kagoshima University Hospital, Sakuragaoka, Kagoshima, Japan
| | - Rebecca Cilluffo
- Cardiovascular Diseases and Internal Medicine, Mayo Clinic and College of Medicine, Rochester, MN
| | - Megha Prasad
- Cardiovascular Diseases and Internal Medicine, Mayo Clinic and College of Medicine, Rochester, MN
| | - Patricia J M Best
- Cardiovascular Diseases and Internal Medicine, Mayo Clinic and College of Medicine, Rochester, MN
| | - Elizabeth J Atkinson
- Biomedical Statistics and Informatics, Mayo Clinic and College of Medicine, Rochester, MN
| | - Tatsuo Aoki
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Aoba-ku, Sendai, Japan
| | - Julie M Cunningham
- Genomics Shared Resource, Mayo Clinic and College of Medicine, Rochester, MN
| | - Mariza de Andrade
- Biomedical Statistics and Informatics, Mayo Clinic and College of Medicine, Rochester, MN
| | - Lilach O Lerman
- Nephrology and Internal Medicine, Mayo Clinic and College of Medicine, Rochester, MN
| | - Amir Lerman
- Cardiovascular Diseases and Internal Medicine, Mayo Clinic and College of Medicine, Rochester, MN
| |
Collapse
|
92
|
Schmidt K, Noureen A, Kronenberg F, Utermann G. Structure, function, and genetics of lipoprotein (a). J Lipid Res 2016; 57:1339-59. [PMID: 27074913 DOI: 10.1194/jlr.r067314] [Citation(s) in RCA: 333] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Indexed: 12/29/2022] Open
Abstract
Lipoprotein (a) [Lp(a)] has attracted the interest of researchers and physicians due to its intriguing properties, including an intragenic multiallelic copy number variation in the LPA gene and the strong association with coronary heart disease (CHD). This review summarizes present knowledge of the structure, function, and genetics of Lp(a) with emphasis on the molecular and population genetics of the Lp(a)/LPA trait, as well as aspects of genetic epidemiology. It highlights the role of genetics in establishing Lp(a) as a risk factor for CHD, but also discusses uncertainties, controversies, and lack of knowledge on several aspects of the genetic Lp(a) trait, not least its function.
Collapse
Affiliation(s)
- Konrad Schmidt
- Divisions of Human Genetics Medical University of Innsbruck, Innsbruck, Austria Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Asma Noureen
- Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Florian Kronenberg
- Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Gerd Utermann
- Divisions of Human Genetics Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
93
|
Kuivaniemi H, Ryer EJ, Elmore JR, Tromp G. Understanding the pathogenesis of abdominal aortic aneurysms. Expert Rev Cardiovasc Ther 2016; 13:975-87. [PMID: 26308600 DOI: 10.1586/14779072.2015.1074861] [Citation(s) in RCA: 246] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
An aortic aneurysm is a dilatation in which the aortic diameter is ≥3.0 cm. If left untreated, the aortic wall continues to weaken and becomes unable to withstand the forces of the luminal blood pressure resulting in progressive dilatation and rupture, a catastrophic event associated with a mortality of 50-80%. Smoking and positive family history are important risk factors for the development of abdominal aortic aneurysms (AAA). Several genetic risk factors have also been identified. On the histological level, visible hallmarks of AAA pathogenesis include inflammation, smooth muscle cell apoptosis, extracellular matrix degradation and oxidative stress. We expect that large genetic, genomic, epigenetic, proteomic and metabolomic studies will be undertaken by international consortia to identify additional risk factors and biomarkers, and to enhance our understanding of the pathobiology of AAA. Collaboration between different research groups will be important in overcoming the challenges to develop pharmacological treatments for AAA.
Collapse
Affiliation(s)
- Helena Kuivaniemi
- a 1 Sigfried and Janet Weis Center for Research, Geisinger Health System, Danville, PA 17822, USA
| | | | | | | |
Collapse
|
94
|
Sticchi E, Magi A, Kamstrup PR, Marcucci R, Prisco D, Martinelli I, Mannucci PM, Abbate R, Giusti B. Apolipoprotein(a) Kringle-IV Type 2 Copy Number Variation Is Associated with Venous Thromboembolism. PLoS One 2016; 11:e0149427. [PMID: 26900838 PMCID: PMC4762703 DOI: 10.1371/journal.pone.0149427] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 01/02/2016] [Indexed: 11/18/2022] Open
Abstract
In addition to the established association between high lipoprotein(a) [Lp(a)] concentrations and coronary artery disease, an association between Lp(a) and venous thromboembolism (VTE) has also been described. Lp(a) is controlled by genetic variants in LPA gene, coding for apolipoprotein(a), including the kringle-IV type 2 (KIV-2) size polymorphism. Aim of the study was to investigate the role of LPA gene KIV-2 size polymorphism and single nucleotide polymorphisms (SNPs) (rs1853021, rs1800769, rs3798220, rs10455872) in modulating VTE susceptibility. Five hundred and sixteen patients with VTE without hereditary and acquired thrombophilia and 1117 healthy control subjects, comparable for age and sex, were investigated. LPA KIV-2 polymorphism, rs3798220 and rs10455872 SNPs were genotyped by TaqMan technology. Concerning rs1853021 and rs1800769 SNPs, PCR-RFLP assay was used. LPA KIV-2 repeat number was significantly lower in patients than in controls [median (interquartile range) 11(6–17) vs 15(9–25), p<0.0001]. A significantly higher prevalence of KIV-2 repeat number ≤7 was observed in patients than in controls (33.5% vs 15.5%, p<0.0001). KIV-2 repeat number was independently associated with VTE (p = 4.36 x10-9), as evidenced by the general linear model analysis adjusted for transient risk factors. No significant difference in allele frequency for all SNPs investigated was observed. Haplotype analysis showed that LPA haplotypes rather than individual SNPs influenced disease susceptibility. Receiver operating characteristic curves analysis showed that a combined risk prediction model, including KIV-2 size polymorphism and clinical variables, had a higher performance in identifying subjects at VTE risk than a clinical-only model, also separately in men and women.
Collapse
Affiliation(s)
- Elena Sticchi
- Department of Experimental and Clinical Medicine, University of Florence—Atherothrombotic Disease Center, Careggi Hospital, Florence, Italy
| | - Alberto Magi
- Department of Experimental and Clinical Medicine, University of Florence—Atherothrombotic Disease Center, Careggi Hospital, Florence, Italy
| | - Pia R. Kamstrup
- Department of Clinical Biochemistry, Copenhagen University Hospital—Herlev, Herlev, Denmark
| | - Rossella Marcucci
- Department of Experimental and Clinical Medicine, University of Florence—Atherothrombotic Disease Center, Careggi Hospital, Florence, Italy
| | - Domenico Prisco
- Department of Experimental and Clinical Medicine, University of Florence—SOD Patologia Medica, Center for Autoimmune Systemic Diseases, Behçet Center and Lupus Clinic, Careggi Hospital, Florence, Italy
| | - Ida Martinelli
- A. Bianchi Bonomi Hemophilia and Thrombosis Center—Ospedale Maggiore Policlinico, Milan, Italy
| | - Pier Mannuccio Mannucci
- A. Bianchi Bonomi Hemophilia and Thrombosis Center—Ospedale Maggiore Policlinico, Milan, Italy
- Scientific Direction, Fondazione Ca’ Granda–Ospedale Maggiore Policlinico, Milan, Italy
| | - Rosanna Abbate
- Department of Experimental and Clinical Medicine, University of Florence—Atherothrombotic Disease Center, Careggi Hospital, Florence, Italy
| | - Betti Giusti
- Department of Experimental and Clinical Medicine, University of Florence—Atherothrombotic Disease Center, Careggi Hospital, Florence, Italy
- * E-mail:
| |
Collapse
|
95
|
Boffa MB, Koschinsky ML. Lipoprotein (a): truly a direct prothrombotic factor in cardiovascular disease? J Lipid Res 2015; 57:745-57. [PMID: 26647358 DOI: 10.1194/jlr.r060582] [Citation(s) in RCA: 172] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Indexed: 01/13/2023] Open
Abstract
Elevated plasma concentrations of lipoprotein (a) [Lp(a)] have been determined to be a causal risk factor for coronary heart disease, and may similarly play a role in other atherothrombotic disorders. Lp(a) consists of a lipoprotein moiety indistinguishable from LDL, as well as the plasminogen-related glycoprotein, apo(a). Therefore, the pathogenic role for Lp(a) has traditionally been considered to reflect a dual function of its similarity to LDL, causing atherosclerosis, and its similarity to plasminogen, causing thrombosis through inhibition of fibrinolysis. This postulate remains highly speculative, however, because it has been difficult to separate the prothrombotic/antifibrinolytic functions of Lp(a) from its proatherosclerotic functions. This review surveys the current landscape surrounding these issues: the biochemical basis for procoagulant and antifibrinolytic effects of Lp(a) is summarized and the evidence addressing the role of Lp(a) in both arterial and venous thrombosis is discussed. While elevated Lp(a) appears to be primarily predisposing to thrombotic events in the arterial tree, the fact that most of these are precipitated by underlying atherosclerosis continues to confound our understanding of the true pathogenic roles of Lp(a) and, therefore, the most appropriate therapeutic target through which to mitigate the harmful effects of this lipoprotein.
Collapse
Affiliation(s)
- Michael B Boffa
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, ON, Canada
| | - Marlys L Koschinsky
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, ON, Canada Robarts Research Institute, Western University, London, ON, Canada
| |
Collapse
|
96
|
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
|
97
|
Bradley DT, Badger SA, McFarland M, Hughes AE. Abdominal Aortic Aneurysm Genetic Associations: Mostly False? A Systematic Review and Meta-analysis. Eur J Vasc Endovasc Surg 2015; 51:64-75. [PMID: 26460285 DOI: 10.1016/j.ejvs.2015.09.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 09/07/2015] [Indexed: 01/27/2023]
Abstract
OBJECTIVE/BACKGROUND Many associations between abdominal aortic aneurysm (AAA) and genetic polymorphisms have been reported. It is unclear which are genuine and which may be caused by type 1 errors, biases, and flexible study design. The objectives of the study were to identify associations supported by current evidence and to investigate the effect of study design on reporting associations. METHODS Data sources were MEDLINE, Embase, and Web of Science. Reports were dual-reviewed for relevance and inclusion against predefined criteria (studies of genetic polymorphisms and AAA risk). Study characteristics and data were extracted using an agreed tool and reports assessed for quality. Heterogeneity was assessed using I(2) and fixed- and random-effects meta-analyses were conducted for variants that were reported at least twice, if any had reported an association. Strength of evidence was assessed using a standard guideline. RESULTS Searches identified 467 unique articles, of which 97 were included. Of 97 studies, 63 reported at least one association. Of 92 studies that conducted multiple tests, only 27% corrected their analyses. In total, 263 genes were investigated, and associations were reported in polymorphisms in 87 genes. Associations in CDKN2BAS, SORT1, LRP1, IL6R, MMP3, AGTR1, ACE, and APOA1 were supported by meta-analyses. CONCLUSION Uncorrected multiple testing and flexible study design (particularly testing many inheritance models and subgroups, and failure to check for Hardy-Weinberg equilibrium) contributed to apparently false associations being reported. Heterogeneity, possibly due to the case mix, geographical, temporal, and environmental variation between different studies, was evident. Polymorphisms in nine genes had strong or moderate support on the basis of the literature at this time. Suggestions are made for improving AAA genetics study design and conduct.
Collapse
Affiliation(s)
- D T Bradley
- Centre for Public Health, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Institute of Clinical Sciences, Block B, Royal Victoria Hospital, Belfast BT12 6BA, UK.
| | - S A Badger
- Mater Misericordiae University Hospital, Eccles Street, Dublin, Ireland
| | - M McFarland
- Department of Pathology, Institute of Pathology Building, Royal Victoria Hospital, Belfast Health and Social Care Trust, Grosvenor Road, Belfast BT12 6BL, UK
| | - A E Hughes
- Centre for Public Health, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Institute of Clinical Sciences, Block B, Royal Victoria Hospital, Belfast BT12 6BA, UK
| |
Collapse
|
98
|
Asociación entre variantes genéticas de enfermedad coronaria y aterosclerosis subclínica: estudio de asociación y metanálisis. Rev Esp Cardiol 2015. [DOI: 10.1016/j.recesp.2014.10.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
99
|
Miner GH, Faries PL, Costa KD, Hanss BG, Marin ML. An update on the etiology of abdominal aortic aneurysms: implications for future diagnostic testing. Expert Rev Cardiovasc Ther 2015; 13:1079-90. [PMID: 26401919 DOI: 10.1586/14779072.2015.1082906] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Abdominal aortic aneurysm (AAA) disease is multifactorial with both environmental and genetic risk factors. The current research in AAA revolves around genetic profiles and expression studies in both human and animal models. Variants in genes involved in extracellular matrix degradation, inflammation, the renin-angiotensin system, cell growth and proliferation and lipid metabolism have been associated with AAA using a variety of study designs. However, the results have been inconsistent and without a standard animal model for validation. Thus, despite the growing body of knowledge, the specific variants responsible for AAA development, progression and rupture have yet to be determined. This review explores some of the more significant genetic studies to provide an overview of past studies that have influenced the current understanding of AAA etiology. Expanding our understanding of disease pathogenesis will inform research into novel diagnostics and therapeutics and ultimately to improve outcomes for patients with AAA.
Collapse
Affiliation(s)
- Grace H Miner
- a Icahn school of Medicine at Mount Sinai, New York, USA
| | - Peter L Faries
- a Icahn school of Medicine at Mount Sinai, New York, USA
| | - Kevin D Costa
- a Icahn school of Medicine at Mount Sinai, New York, USA
| | - Basil G Hanss
- a Icahn school of Medicine at Mount Sinai, New York, USA
| | | |
Collapse
|
100
|
Affiliation(s)
- Ilse Luyckx
- Laboratory for aneurysmal disease, Center for Medical Genetics, Antwerp University Hospital/University of Antwerp, Antwerp, Belgium
| | - Bart L Loeys
- Laboratory for aneurysmal disease, Center for Medical Genetics, Antwerp University Hospital/University of Antwerp, Antwerp, Belgium
| | | |
Collapse
|