1
|
Chen J, Zhang H, Li L, Zhang X, Zhao D, Wang L, Wang J, Yang P, Sun H, Liu K, Chen W, Li L, Lin F, Li Z, Chen YE, Zhang J, Pang D, Ouyang H, He Y, Fan J, Tang X. Lp-PLA 2 (Lipoprotein-Associated Phospholipase A 2) Deficiency Lowers Cholesterol Levels and Protects Against Atherosclerosis in Rabbits. Arterioscler Thromb Vasc Biol 2023; 43:e11-e28. [PMID: 36412196 DOI: 10.1161/atvbaha.122.317898] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
BACKGROUND Elevated plasma Lp-PLA2 (lipoprotein-associated phospholipase A2) activity is closely associated with an increased risk of cardiovascular events. However, whether and how Lp-PLA2 is directly involved in the pathogenesis of atherosclerosis is still unclear. To examine the hypothesis that Lp-PLA2 could be a potential preventative target of atherosclerosis, we generated Lp-PLA2 knockout rabbits and investigated the pathophysiological functions of Lp-PLA2. METHODS Lp-PLA2 knockout rabbits were generated using CRISPR/Cas9 system to assess the role of Lp-PLA2 in plasma lipids regulation and identify its underlying molecular mechanisms. Homozygous knockout rabbits along with wild-type rabbits were fed a cholesterol-rich diet for up to 14 weeks and their atherosclerotic lesions were compared. Moreover, the effects of Lp-PLA2 deficiency on the key cellular behaviors in atherosclerosis were assessed in vitro. RESULTS When rabbits were fed a standard diet, Lp-PLA2 deficiency led to a significant reduction in plasma lipids. The decreased protein levels of SREBP2 (sterol regulatory element-binding protein 2) and HMGCR (3-hydroxy-3-methylglutaryl coenzyme A reductase) in livers of homozygous knockout rabbits indicated that the cholesterol biosynthetic pathway was impaired with Lp-PLA2 deficiency. In vitro experiments further demonstrated that intracellular Lp-PLA2 efficiently enhanced SREBP2-related cholesterol biosynthesis signaling independently of INSIGs (insulin-induced genes). When fed a cholesterol-rich diet, homozygous knockout rabbits exhibited consistently lower level of hypercholesterolemia, and their aortic atherosclerosis lesions were significantly reduced by 60.2% compared with those of wild-type rabbits. The lesions of homozygous knockout rabbits were characterized by reduced macrophages and the expression of inflammatory cytokines. Macrophages of homozygous knockout rabbits were insensitive to M1 polarization and showed reduced DiI-labeled lipoprotein uptake capacity compared with wild-type macrophages. Lp-PLA2 deficiency also inhibited the adhesion between monocytes and endothelial cells. CONCLUSIONS These results demonstrate that Lp-PLA2 plays a causal role in regulating blood lipid homeostasis and Lp-PLA2 deficiency protects against dietary cholesterol-induced atherosclerosis in rabbits. Lp-PLA2 could be a potential target for the prevention of atherosclerosis.
Collapse
Affiliation(s)
- Jiahuan Chen
- College of Animal Sciences, Jilin University, Changchun, Jilin Province, China (J.C., H.Z., Linquan Li, X.Z., D.Z., L.W., J.W., Lin Li, F.L., Z.L., D.P., H.O., X.T.)
| | - Huanyu Zhang
- College of Animal Sciences, Jilin University, Changchun, Jilin Province, China (J.C., H.Z., Linquan Li, X.Z., D.Z., L.W., J.W., Lin Li, F.L., Z.L., D.P., H.O., X.T.)
| | - Linquan Li
- College of Animal Sciences, Jilin University, Changchun, Jilin Province, China (J.C., H.Z., Linquan Li, X.Z., D.Z., L.W., J.W., Lin Li, F.L., Z.L., D.P., H.O., X.T.)
| | - Xinwei Zhang
- College of Animal Sciences, Jilin University, Changchun, Jilin Province, China (J.C., H.Z., Linquan Li, X.Z., D.Z., L.W., J.W., Lin Li, F.L., Z.L., D.P., H.O., X.T.)
| | - Dazhong Zhao
- College of Animal Sciences, Jilin University, Changchun, Jilin Province, China (J.C., H.Z., Linquan Li, X.Z., D.Z., L.W., J.W., Lin Li, F.L., Z.L., D.P., H.O., X.T.)
| | - Lingyu Wang
- College of Animal Sciences, Jilin University, Changchun, Jilin Province, China (J.C., H.Z., Linquan Li, X.Z., D.Z., L.W., J.W., Lin Li, F.L., Z.L., D.P., H.O., X.T.)
| | - Jiaqi Wang
- College of Animal Sciences, Jilin University, Changchun, Jilin Province, China (J.C., H.Z., Linquan Li, X.Z., D.Z., L.W., J.W., Lin Li, F.L., Z.L., D.P., H.O., X.T.)
| | - Ping Yang
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China (P.Y., H.S., K.L., W.C., Y.H.)
| | - Huan Sun
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China (P.Y., H.S., K.L., W.C., Y.H.)
| | - Kun Liu
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China (P.Y., H.S., K.L., W.C., Y.H.)
| | - Weiwei Chen
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China (P.Y., H.S., K.L., W.C., Y.H.)
| | - Lin Li
- College of Animal Sciences, Jilin University, Changchun, Jilin Province, China (J.C., H.Z., Linquan Li, X.Z., D.Z., L.W., J.W., Lin Li, F.L., Z.L., D.P., H.O., X.T.)
| | - Feng Lin
- College of Animal Sciences, Jilin University, Changchun, Jilin Province, China (J.C., H.Z., Linquan Li, X.Z., D.Z., L.W., J.W., Lin Li, F.L., Z.L., D.P., H.O., X.T.)
| | - Zhanjun Li
- College of Animal Sciences, Jilin University, Changchun, Jilin Province, China (J.C., H.Z., Linquan Li, X.Z., D.Z., L.W., J.W., Lin Li, F.L., Z.L., D.P., H.O., X.T.)
| | - Y Eugene Chen
- Department of Internal Medicine, Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical Center, Ann Arbor (Y.E.C., J.Z.)
| | - Jifeng Zhang
- Department of Internal Medicine, Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical Center, Ann Arbor (Y.E.C., J.Z.)
| | - Daxin Pang
- College of Animal Sciences, Jilin University, Changchun, Jilin Province, China (J.C., H.Z., Linquan Li, X.Z., D.Z., L.W., J.W., Lin Li, F.L., Z.L., D.P., H.O., X.T.).,Chongqing Research Institute, Jilin University, Chongqing, China (D.P., H.O., X.T.)
| | - Hongsheng Ouyang
- College of Animal Sciences, Jilin University, Changchun, Jilin Province, China (J.C., H.Z., Linquan Li, X.Z., D.Z., L.W., J.W., Lin Li, F.L., Z.L., D.P., H.O., X.T.).,Chongqing Research Institute, Jilin University, Chongqing, China (D.P., H.O., X.T.)
| | - Yuquan He
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China (P.Y., H.S., K.L., W.C., Y.H.)
| | - Jianglin Fan
- Department of Molecular Pathology, Faculty of Medicine, Graduate School of Medical Sciences, University of Yamanashi, Japan (J.F.)
| | - Xiaochun Tang
- College of Animal Sciences, Jilin University, Changchun, Jilin Province, China (J.C., H.Z., Linquan Li, X.Z., D.Z., L.W., J.W., Lin Li, F.L., Z.L., D.P., H.O., X.T.).,Chongqing Research Institute, Jilin University, Chongqing, China (D.P., H.O., X.T.)
| |
Collapse
|
2
|
Khan MI, Hariprasad G. Human Secretary Phospholipase A2 Mutations and Their Clinical Implications. J Inflamm Res 2020; 13:551-561. [PMID: 32982370 PMCID: PMC7502393 DOI: 10.2147/jir.s269557] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 08/13/2020] [Indexed: 01/05/2023] Open
Abstract
Phospholipases A2 (PLA2s) belong to a superfamily of enzymes responsible for hydrolysis of the sn-2 fatty acids of membrane phospholipids to release arachidonic acid. PLA2s are the rate limiting enzyme for the downstream synthesis of prostaglandins and leukotrienes that are the main mediators of inflammation. The extracellular forms of this enzyme are also called the secretary phospholipase A2 (sPLA2) and are distributed extensively in most of the tissues in the human body. Their integral role in inflammatory pathways has been the primary reason for the extensive research on this molecule. The catalytic mechanism of sPLA2 is initiated by a histidine/aspartic acid/calcium complex within the active site. Though they are known to have certain housekeeping functions, certain mutations of sPLA2 are known to be implicated in causation of certain pathologies leading to diseases such as atherosclerosis, cardiovascular diseases, benign fleck retina, neurodegeneration, and asthma. We present an overview of human sPLA2 and a comprehensive compilation of the mutations that result in various disease phenotypes. The study not only helps to have a holistic understanding of human sPLA2 mutations and their clinical implications, but is also a useful platform to initiate research pertaining to structure–function relationship of the mutations to develop effective therapies for management of these diseases.
Collapse
Affiliation(s)
- Mohd Imran Khan
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Gururao Hariprasad
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India
| |
Collapse
|
3
|
Santoso A, Heriansyah T, Rohman MS. Phospholipase A2 is an Inflammatory Predictor in Cardiovascular Diseases: Is there any Spacious Room to Prove the Causation? Curr Cardiol Rev 2020; 16:3-10. [PMID: 31146670 PMCID: PMC7393598 DOI: 10.2174/1573403x15666190531111932] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 05/01/2019] [Accepted: 05/05/2019] [Indexed: 12/17/2022] Open
Abstract
Lipoprotein-associated phospholipase A2 (Lp-PLA2) is an enzyme family of phospholipase A2 produced by the inflammatory cell in atherosclerotic plaque. It is transported in the circulation, attached mainly to low-density lipoprotein-cholesterol (LDL-C). It hydrolyzes glycerophospholipids particularly fatty acids at the sn-2 position and produces numerous bioactive lipids; and leads to endothelial dysfunction, atherosclerotic plaque inflammation, and development of the necrotic core in plaques. There are two kinds of phospholipase A2, namely: secretory phospholipase A2 (sPLA2) and Lp- PLA2. They are deemed as evolving predictors of cardiovascular disease (CVD) risk in hospitaland population-based studies, including healthy subjects, acute coronary syndromes (ACS) and patients with CVD. Unfortunately, Lp-PLA2 inhibitor (darapladib) and s-PLA2 inhibitor (varespladib methyl) failed to prove to lower the risk of composite CVD mortality, myocardial infarction and stroke in those with stable CVD and ACS. Herein, we describe the explanation based on the existing data why there is still a discrepancy among them. So, it highlights the opinion that phospholipase A2 is merely the inflammatory biomarkers of CVD and playing an important role in atherosclerosis. Further, there is more spacious room to prove the causation.
Collapse
Affiliation(s)
- Anwar Santoso
- Address correspondence to this author at the Department of Cardiology and Vascular Medicine, National Cardiovascular Centre, Harapan Kita Hospital, Universitas Indonesia, Jakarta, Indonesia; Tel: +62 21 5684093;
E-mail:
| | | | | |
Collapse
|
4
|
Huang F, Wang K, Shen J. Lipoprotein-associated phospholipase A2: The story continues. Med Res Rev 2019; 40:79-134. [PMID: 31140638 PMCID: PMC6973114 DOI: 10.1002/med.21597] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 04/20/2019] [Accepted: 04/30/2019] [Indexed: 12/15/2022]
Abstract
Inflammation is thought to play an important role in the pathogenesis of vascular diseases. Lipoprotein-associated phospholipase A2 (Lp-PLA2) mediates vascular inflammation through the regulation of lipid metabolism in blood, thus, it has been extensively investigated to identify its role in vascular inflammation-related diseases, mainly atherosclerosis. Although darapladib, the most advanced Lp-PLA2 inhibitor, failed to meet the primary endpoints of two large phase III trials in atherosclerosis patients cotreated with standard medical care, the research on Lp-PLA2 has not been terminated. Novel pathogenic, epidemiologic, genetic, and crystallographic studies regarding Lp-PLA2 have been reported recently, while novel inhibitors were identified through a fragment-based lead discovery strategy. More strikingly, recent clinical and preclinical studies revealed that Lp-PLA2 inhibition showed promising therapeutic effects in diabetic macular edema and Alzheimer's disease. In this review, we not only summarized the knowledge of Lp-PLA2 established in the past decades but also emphasized new findings in recent years. We hope this review could be valuable for helping researchers acquire a much deeper insight into the nature of Lp-PLA2, identify more potent and selective Lp-PLA2 inhibitors, and discover the potential indications of Lp-PLA2 inhibitors.
Collapse
Affiliation(s)
- Fubao Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, China.,School of Pharmacy, University of Chinese Academy of Sciences, Beijing, China
| | - Kai Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, China
| | - Jianhua Shen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
5
|
Hu Q, Hao P, Liu Q, Dong M, Gong Y, Zhang C, Zhang Y. Mendelian randomization studies on atherosclerotic cardiovascular disease: evidence and limitations. SCIENCE CHINA-LIFE SCIENCES 2019; 62:758-770. [DOI: 10.1007/s11427-019-9537-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 03/26/2019] [Indexed: 12/26/2022]
|
6
|
Santoso A, Maulana R, Alzahra F, Maghfirah I, Putrinarita AD, Heriansyah T. Associations between four types of single-nucleotide polymorphisms in PLA2G7 gene and clinical atherosclerosis: a meta-analysis. AMERICAN JOURNAL OF CARDIOVASCULAR DISEASE 2017; 7:122-133. [PMID: 29348973 PMCID: PMC5768870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 12/13/2017] [Indexed: 06/07/2023]
Abstract
BACKGROUND Previous studies suggested that some types of single nucleotide polymorphisms (SNPs) in PLA2G7 genes, encoding Lp-PLA2 have been reported to yield an antiatherogenic effect, but other studies mentioned otherwise. Thus, a comprehensive study to explore the effect of SNPs in PLA2G7 genes (V279F, A379V, R92H, I198T) toward clinical atherosclerosis is needed. METHODS We searched eligible studies from PubMed, EBSCO, ProQuest, Science Direct, Springer, and Cochrane databases for case-control studies to assess the between four types of SNPs in PLA2G7 gene with risk of clinical atherosclerosis (CVD = cardiovascular disease, CAD = coronary artery disease, PAD = peripheral artery disease, ischemic stroke). All studies were assessed under Hardy-Weinberg Equilibrium, an additive model. This meta-analysis was performed by RevMan 5.3 to provide pooled estimate for odds ratio (ORs) with 95% confidence intervals (95% CIs). RESULTS Fourteen clinical studies met our inclusion criteria. Those included 12,432 patients with clinical atherosclerosis and 10,171 were controls. We found that ORs of two variants SNPs (V279F, R92H) were associated with clinical atherosclerosis {V279F, OR = 0.88 (95% CI, 0.81-0.95); p = 0.0007, I2 = 40%}, {R92H, OR = 1.29 (95% CI, 1.09-1.53); p = 0.003, I2 = 73%}. Meanwhile, there was no significant associations between the other two, A379V {OR = 1.08 (95% CI, 0.93-1.26); p = 0.31, I2 = 78%} and I198T {OR = 1.12 (95% CI = 0.79-1.59); p = 0.53, I2 = 81%}. CONCLUSIONS These results suggested that V279F polymorphism in PLA2G7 gene has a protective effect for clinical atherosclerosis, whereas R92H polymorphism might contribute toward increased risk of clinical atherosclerosis.
Collapse
Affiliation(s)
- Anwar Santoso
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Universitas Indonesia, Harapan Kita Hospital, National Cardiovascular CentreJakarta, Indonesia
| | - Rido Maulana
- Faculty of Medicine, Muhammadiyah Jakarta UniversityJakarta, Indonesia
| | - Fatimah Alzahra
- Faculty of Medicine, Gadjah Mada UniversityYogyakarta, Indonesia
| | - Irma Maghfirah
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Airlangga UniversitySurabaya, Indonesia
| | - Agnes Dinar Putrinarita
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Universitas Indonesia, Harapan Kita Hospital, National Cardiovascular CentreJakarta, Indonesia
| | - Teuku Heriansyah
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Syiah Kuala UniversityAceh, Indonesia
| |
Collapse
|
7
|
Abstract
The importance of inflammation and inflammatory pathways in atherosclerotic disease and acute coronary syndromes (ACS) is well established. The success of statin therapy rests not only on potently reducing levels of low-density lipoprotein cholesterol, but also on the many beneficial, pleiotropic effects statin therapy has on various inflammatory mechanisms in atherosclerotic disease, from reducing endothelial dysfunction to attenuating levels of serum C-reactive protein. Due to the growing awareness of the importance of inflammation in ACS, investigators have attempted to develop novel therapies against known markers of inflammation for several decades. Targeted pathways have ranged from inhibiting C5 cleavage with a high-affinity monoclonal antibody against C5 to inhibiting the activation of the p38 mitogen-activated protein kinase signaling cascades. In each of these instances, despite promising early preclinical and mechanistic studies and phase 2 trials suggesting a potential benefit in reducing post-MI complications or restenosis, these novel therapies have failed to show benefits during large, phase 3 clinical outcomes trials. This review discusses several examples of novel anti-inflammatory therapies that failed to show significant improvement on clinical outcomes when tested in large, randomized trials and highlights potential explanations for why targeted therapies against known markers of inflammation in ACS have failed to launch.
Collapse
Key Words
- ACS, acute coronary syndromes
- CABG, coronary artery bypass graft
- CAD, coronary artery disease
- HDL-C, high-density lipoprotein cholesterol
- IL, interleukin
- LDL-C, low-density lipoprotein cholesterol
- Lp-PLA2, lipoprotein-associated phospholipase A2
- MAPK, mitogen-activated protein kinase
- MI, myocardial infarction
- NSTEMI, non–ST-segment myocardial infarction
- PCI, percutaneous coronary intervention
- PSGL, P-selectin glycoprotein ligand
- STEMI, ST-segment elevation myocardial infarction
- SVG, saphenous vein grafts
- TBR, tissue-to-background ratio
- acute coronary syndrome
- anti-inflammatory
- drug targets
- hsCRP, high-sensitivity C-reactive protein
- sPLA2, secretory phospholipase A2
Collapse
|
8
|
Yeo A, Li L, Warren L, Aponte J, Fraser D, King K, Johansson K, Barnes A, MacPhee C, Davies R, Chissoe S, Tarka E, O’Donoghue ML, White HD, Wallentin L, Waterworth D. Pharmacogenetic meta-analysis of baseline risk factors, pharmacodynamic, efficacy and tolerability endpoints from two large global cardiovascular outcomes trials for darapladib. PLoS One 2017; 12:e0182115. [PMID: 28753643 PMCID: PMC5533343 DOI: 10.1371/journal.pone.0182115] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 07/11/2017] [Indexed: 01/27/2023] Open
Abstract
Darapladib, a lipoprotein-associated phospholipase A2 (Lp-PLA2) inhibitor, failed to demonstrate efficacy for the primary endpoints in two large phase III cardiovascular outcomes trials, one in stable coronary heart disease patients (STABILITY) and one in acute coronary syndrome (SOLID-TIMI 52). No major safety signals were observed but tolerability issues of diarrhea and odor were common (up to 13%). We hypothesized that genetic variants associated with Lp-PLA2 activity may influence efficacy and tolerability and therefore performed a comprehensive pharmacogenetic analysis of both trials. We genotyped patients within the STABILITY and SOLID-TIMI 52 trials who provided a DNA sample and consent (n = 13,577 and 10,404 respectively, representing 86% and 82% of the trial participants) using genome-wide arrays with exome content and performed imputation using a 1000 Genomes reference panel. We investigated baseline and change from baseline in Lp-PLA2 activity, two efficacy endpoints (major coronary events and myocardial infarction) as well as tolerability parameters at genome-wide and candidate gene level using a meta-analytic approach. We replicated associations of published loci on baseline Lp-PLA2 activity (APOE, CELSR2, LPA, PLA2G7, LDLR and SCARB1) and identified three novel loci (TOMM5, FRMD5 and LPL) using the GWAS-significance threshold P≤5E-08. Review of the PLA2G7 gene (encoding Lp-PLA2) within these datasets identified V279F null allele carriers as well as three other rare exonic null alleles within various ethnic groups, however none of these variants nor any other loci associated with Lp-PLA2 activity at baseline were associated with any of the drug response endpoints. The analysis of darapladib efficacy endpoints, despite low power, identified six low frequency loci with main genotype effect (though with borderline imputation scores) and one common locus (minor allele frequency 0.24) with genotype by treatment interaction effect passing the GWAS-significance threshold. This locus conferred risk in placebo subjects, hazard ratio (HR) 1.22 with 95% confidence interval (CI) 1.11–1.33, but was protective in darapladib subjects, HR 0.79 (95% CI 0.71–0.88). No major loci for tolerability were found. Thus, genetic analysis confirmed and extended the influence of lipoprotein loci on Lp-PLA2 levels, identified some novel null alleles in the PLA2G7 gene, and only identified one potentially efficacious subgroup within these two large clinical trials.
Collapse
Affiliation(s)
- Astrid Yeo
- Department of Genetics, GlaxoSmithKline Medicines Research Centre, Stevenage, Hertfordshire, United Kingdom
| | - Li Li
- Department of Genetics, GlaxoSmithKline Medicines Research Centre, Research Triangle Park, North Carolina, United States of America
| | - Liling Warren
- Department of Genetics, GlaxoSmithKline Medicines Research Centre, Research Triangle Park, North Carolina, United States of America
| | - Jennifer Aponte
- Department of Genetics, GlaxoSmithKline Medicines Research Centre, Research Triangle Park, North Carolina, United States of America
| | - Dana Fraser
- Department of Genetics, GlaxoSmithKline Medicines Research Centre, Research Triangle Park, North Carolina, United States of America
| | - Karen King
- Department of Genetics, GlaxoSmithKline Medicines Research Centre, Research Triangle Park, North Carolina, United States of America
| | - Kelley Johansson
- Department of Genetics, GlaxoSmithKline Medicines Research Centre, Research Triangle Park, North Carolina, United States of America
| | - Allison Barnes
- Clinical Statistics, GlaxoSmithKline Medicines Research Centre, Research Triangle Park, North Carolina, United States of America
| | - Colin MacPhee
- Department of Vascular Biology & Thrombosis, GlaxoSmithKline Medicines Research Centre, King of Prussia, Pennsylvania, United States of America
| | - Richard Davies
- Metabolic Pathways and Cardiovascular Therapeutic Area, GlaxoSmithKline Medicines Research Centre, King of Prussia, Pennsylvania, United States of America
| | - Stephanie Chissoe
- Department of Genetics, GlaxoSmithKline Medicines Research Centre, Research Triangle Park, North Carolina, United States of America
| | - Elizabeth Tarka
- Metabolic Pathways and Cardiovascular Therapeutic Area, GlaxoSmithKline Medicines Research Centre, King of Prussia, Pennsylvania, United States of America
| | - Michelle L. O’Donoghue
- TIMI Study Group, Cardiovascular Division, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
| | - Harvey D. White
- Green Lane Cardiovascular Service, Auckland City Hospital and University of Auckland, Auckland, New Zealand
| | - Lars Wallentin
- Department of Medical Sciences, Cardiology & Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
| | - Dawn Waterworth
- Department of Genetics, GlaxoSmithKline Medicines Research Centre, Upper Merion, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
9
|
Kim M, Kim M, Yoo HJ, Jang HY, Lee SH, Lee JH. Effects of overweight and the PLA2G7 V279F polymorphism on the association of age with systolic blood pressure. PLoS One 2017; 12:e0173611. [PMID: 28334001 PMCID: PMC5363925 DOI: 10.1371/journal.pone.0173611] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 02/22/2017] [Indexed: 12/20/2022] Open
Abstract
This prospective study aimed to determine the effects of the persistence of overweight for three years and the PLA2G7 V279F polymorphism, as well as the interaction between these factors, on the association of age with blood pressure (BP). Healthy middle-aged subjects with normotensive BP were divided into the normal-weight and overweight groups. The PLA2G7 V279F genotype, BP, lipoprotein-associated phospholipase A2 (Lp-PLA2) activity, and oxidized low-density lipoprotein (ox-LDL) were determined. Lp-PLA2 activity was lower in the F allele subjects (n = 111) than in those with the VV genotype (n = 389). The overweight individuals with the F allele had lower Lp-PLA2 activity and ox-LDL at both baseline and after three years and lower systolic and diastolic BP and LDL cholesterol after three years compared with those with the VV phenotype. After three years, the overweight subjects with the VV phenotype exhibited greater increases in Lp-PLA2 activity, systolic BP, and ox-LDL than those with the F allele and normal-weight subjects with the VV phenotype. A multivariate analysis revealed that the PLA2G7 V279F genotype, baseline BMI, changes in Lp-PLA2 activity and ox-LDL remained independently and positively associated with changes in systolic BP. The simultaneous presence of the PLA2G7 279VV genotype and persistence of overweight synergistically increases the risk for hypertension, whereas lower Lp-PLA2 activity in PLA2G7 279F allele carriers might offer certain protection against hypertension, even in individuals who have been overweight for over three years.
Collapse
Affiliation(s)
- Minjoo Kim
- Research Center for Silver Science, Institute of Symbiotic Life-TECH, Yonsei University, Seoul, Korea
| | - Minkyung Kim
- Research Center for Silver Science, Institute of Symbiotic Life-TECH, Yonsei University, Seoul, Korea
| | - Hye Jin Yoo
- National Leading Research Laboratory of Clinical Nutrigenetics/Nutrigenomics, Department of Food and Nutrition, College of Human Ecology, Yonsei University, Seoul, Korea
- Department of Food and Nutrition, Brain Korea 21 PLUS Project, College of Human Ecology, Yonsei University, Seoul, Korea
| | - Hye Young Jang
- National Leading Research Laboratory of Clinical Nutrigenetics/Nutrigenomics, Department of Food and Nutrition, College of Human Ecology, Yonsei University, Seoul, Korea
- Department of Food and Nutrition, Brain Korea 21 PLUS Project, College of Human Ecology, Yonsei University, Seoul, Korea
| | - Sang-Hyun Lee
- Department of Family Practice, National Health Insurance Corporation, Ilsan Hospital, Goyang, Korea
| | - Jong Ho Lee
- Research Center for Silver Science, Institute of Symbiotic Life-TECH, Yonsei University, Seoul, Korea
- National Leading Research Laboratory of Clinical Nutrigenetics/Nutrigenomics, Department of Food and Nutrition, College of Human Ecology, Yonsei University, Seoul, Korea
- Department of Food and Nutrition, Brain Korea 21 PLUS Project, College of Human Ecology, Yonsei University, Seoul, Korea
- * E-mail:
| |
Collapse
|
10
|
Gregson JM, Freitag DF, Surendran P, Stitziel NO, Chowdhury R, Burgess S, Kaptoge S, Gao P, Staley JR, Willeit P, Nielsen SF, Caslake M, Trompet S, Polfus LM, Kuulasmaa K, Kontto J, Perola M, Blankenberg S, Veronesi G, Gianfagna F, Männistö S, Kimura A, Lin H, Reilly DF, Gorski M, Mijatovic V, Munroe PB, Ehret GB, Thompson A, Uria-Nickelsen M, Malarstig A, Dehghan A, Vogt TF, Sasaoka T, Takeuchi F, Kato N, Yamada Y, Kee F, Müller-Nurasyid M, Ferrières J, Arveiler D, Amouyel P, Salomaa V, Boerwinkle E, Thompson SG, Ford I, Wouter Jukema J, Sattar N, Packard CJ, Shafi Majumder AA, Alam DS, Deloukas P, Schunkert H, Samani NJ, Kathiresan S, Nordestgaard BG, Saleheen D, Howson JMM, Di Angelantonio E, Butterworth AS, Danesh J. Genetic invalidation of Lp-PLA 2 as a therapeutic target: Large-scale study of five functional Lp-PLA 2-lowering alleles. Eur J Prev Cardiol 2017; 24:492-504. [PMID: 27940953 PMCID: PMC5460752 DOI: 10.1177/2047487316682186] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 10/24/2016] [Indexed: 01/12/2023]
Abstract
Aims Darapladib, a potent inhibitor of lipoprotein-associated phospholipase A2 (Lp-PLA2), has not reduced risk of cardiovascular disease outcomes in recent randomized trials. We aimed to test whether Lp-PLA2 enzyme activity is causally relevant to coronary heart disease. Methods In 72,657 patients with coronary heart disease and 110,218 controls in 23 epidemiological studies, we genotyped five functional variants: four rare loss-of-function mutations (c.109+2T > C (rs142974898), Arg82His (rs144983904), Val279Phe (rs76863441), Gln287Ter (rs140020965)) and one common modest-impact variant (Val379Ala (rs1051931)) in PLA2G7, the gene encoding Lp-PLA2. We supplemented de-novo genotyping with information on a further 45,823 coronary heart disease patients and 88,680 controls in publicly available databases and other previous studies. We conducted a systematic review of randomized trials to compare effects of darapladib treatment on soluble Lp-PLA2 activity, conventional cardiovascular risk factors, and coronary heart disease risk with corresponding effects of Lp-PLA2-lowering alleles. Results Lp-PLA2 activity was decreased by 64% ( p = 2.4 × 10-25) with carriage of any of the four loss-of-function variants, by 45% ( p < 10-300) for every allele inherited at Val279Phe, and by 2.7% ( p = 1.9 × 10-12) for every allele inherited at Val379Ala. Darapladib 160 mg once-daily reduced Lp-PLA2 activity by 65% ( p < 10-300). Causal risk ratios for coronary heart disease per 65% lower Lp-PLA2 activity were: 0.95 (0.88-1.03) with Val279Phe; 0.92 (0.74-1.16) with carriage of any loss-of-function variant; 1.01 (0.68-1.51) with Val379Ala; and 0.95 (0.89-1.02) with darapladib treatment. Conclusions In a large-scale human genetic study, none of a series of Lp-PLA2-lowering alleles was related to coronary heart disease risk, suggesting that Lp-PLA2 is unlikely to be a causal risk factor.
Collapse
Affiliation(s)
- John M Gregson
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, UK
| | - Daniel F Freitag
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, UK
| | - Praveen Surendran
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, UK
| | - Nathan O Stitziel
- Departments of Medicine and Genetics, Washington University School of Medicine, St Louis, USA
| | - Rajiv Chowdhury
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, UK
| | - Stephen Burgess
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, UK
| | - Stephen Kaptoge
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, UK
| | - Pei Gao
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, UK
| | - James R Staley
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, UK
| | - Peter Willeit
- Department of Public Health and Primary Care, University of Cambridge, UK
- Department of Neurology, Innsbruck Medical University, Austria
| | - Sune F Nielsen
- Copenhagen University Hospital, University of Copenhagen, Denmark
| | | | | | | | - Kari Kuulasmaa
- THL-National Institute for Health and Welfare, Helsinki, Finland
| | - Jukka Kontto
- THL-National Institute for Health and Welfare, Helsinki, Finland
| | - Markus Perola
- Institute of Molecular Medicine FIMM, University of Helsinki, Finland
- Department of Health, National Institute for Health and Welfare, Helsinki, Finland
| | - Stefan Blankenberg
- Department of General and Interventional Cardiology, University Heart Centre Hamburg, Germany
- University Medical Centre Hamburg Eppendorf, Hamburg, Germany
| | - Giovanni Veronesi
- Research Centre, Department of Clinical and Experimental Medicine, University of Insubria, Varese, Italy
| | - Francesco Gianfagna
- Research Centre, Department of Clinical and Experimental Medicine, University of Insubria, Varese, Italy
- Department of Epidemiology and Prevention, IRCCS Istituto Neurologico Mediterraneo Neuromed, Pozzilli, Italy
| | - Satu Männistö
- THL-National Institute for Health and Welfare, Helsinki, Finland
| | - Akinori Kimura
- Department of Molecular Pathogenesis, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Japan
| | - Honghuang Lin
- Section of Computational Biomedicine, Department of Medicine, Boston University School of Medicine, USA
- The NHLBI’s Framingham Heart Study, Framingham, USA
| | - Dermot F Reilly
- Merck Research Laboratories, Genetics and Pharmacogenomics, Boston, USA
| | - Mathias Gorski
- Department of Genetic Epidemiology, University of Regensburg, Germany
- Department of Nephrology, University Hospital Regensburg, Germany
| | - Vladan Mijatovic
- Department of Life and Reproduction Sciences, University of Verona, Italy
| | | | - Patricia B Munroe
- Clinical Pharmacology and The Genome Centre, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, UK
- NIHR Barts Cardiovascular Biomedical Research Unit, Queen Mary University of London, UK
| | - Georg B Ehret
- Center for Complex Disease Genomics, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, USA
- Cardiology, Department of Medicine, Geneva University Hospital, Switzerland
- Institute of Social and Preventive Medicine (IUMSP), Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | | | | | | | | | - Abbas Dehghan
- Department of Epidemiology, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | | | - Thomas F Vogt
- Merck Research Laboratories, Cardiometabolic Disease, Kenilworth, USA
- CHDI Management/CHDI Foundation, Princeton, USA
| | - Taishi Sasaoka
- Department of Molecular Pathogenesis, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Japan
| | - Fumihiko Takeuchi
- Department of Gene Diagnostics and Therapeutics, Research Institute, National Centre for Global Health and Medicine, Tokyo, Japan
| | - Norihiro Kato
- Department of Gene Diagnostics and Therapeutics, Research Institute, National Centre for Global Health and Medicine, Tokyo, Japan
| | - Yoshiji Yamada
- Department of Human Functional Genomics, Life Science Research Centre, Mie University, Japan
| | - Frank Kee
- UKCRC Centre of Excellence for Public Health, Queens University, Belfast, Ireland
| | - Martina Müller-Nurasyid
- Institute of Genetic Epidemiology, Helmholtz Zentrum München - German Research Centre for Environmental Health, Neuherberg, Germany
- Institute of Medical Informatics, Biometry and Epidemiology, Ludwig-Maximilians-Universität, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
- Department of Medicine I, Ludwig-Maximilians-University Munich, Germany
| | - Jean Ferrières
- Department of Epidemiology, UMR 1027-INSERM, Toulouse University-CHU Toulouse, France
| | - Dominique Arveiler
- Department of Epidemiology and Public Health, EA 3430, University of Strasbourg and Strasbourg University Hospital, France
| | - Philippe Amouyel
- Department of Epidemiology and Public Health, Institut Pasteur de Lille, France
| | - Veikko Salomaa
- THL-National Institute for Health and Welfare, Helsinki, Finland
| | - Eric Boerwinkle
- Human Genetics Center, University of Texas Health Science Center at Houston, USA
| | - Simon G Thompson
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, UK
| | | | | | | | | | | | - Dewan S Alam
- Centre for Global Health Research, St Michael Hospital, Toronto, Canada
| | - Panos Deloukas
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, UK
| | - Heribert Schunkert
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
- Deutsches Herzzentrum München, Technische Universität München, Germany
| | - Nilesh J Samani
- Department of Cardiovascular Sciences, University of Leicester and National Institute for Health Research Leicester Cardiovascular Biomedical Research Unit, UK
| | - Sekar Kathiresan
- Broad Institute, Cambridge and Massachusetts General Hospital, Boston, USA
| | | | | | | | - Joanna MM Howson
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, UK
| | - Emanuele Di Angelantonio
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, UK
| | - Adam S Butterworth
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, UK
| | - John Danesh
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, UK
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK
- British Heart Foundation Cambridge Centre of Excellence, University of Cambridge, Cambridge, UK
- National Institute of Health Research Blood and Transplant Research Unit in Donor Health and Genomics, University of Cambridge, Cambridge, UK
| |
Collapse
|
11
|
Measom ND, Down KD, Hirst DJ, Jamieson C, Manas ES, Patel VK, Somers DO. Investigation of a Bicyclo[1.1.1]pentane as a Phenyl Replacement within an LpPLA 2 Inhibitor. ACS Med Chem Lett 2017; 8:43-48. [PMID: 28105273 DOI: 10.1021/acsmedchemlett.6b00281] [Citation(s) in RCA: 141] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 11/15/2016] [Indexed: 02/07/2023] Open
Abstract
We describe the incorporation of a bicyclo[1.1.1]pentane moiety within two known LpPLA2 inhibitors to act as bioisosteric phenyl replacements. An efficient synthesis to the target compounds was enabled with a dichlorocarbene insertion into a bicyclo[1.1.0]butane system being the key transformation. Potency, physicochemical, and X-ray crystallographic data were obtained to compare the known inhibitors to their bioisosteric counterparts, which showed the isostere was well tolerated and positively impacted on the physicochemical profile.
Collapse
Affiliation(s)
- Nicholas D. Measom
- GlaxoSmithKline, Medicines Research Centre, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, U.K
- Department
of Pure and Applied Chemistry, University of Strathclyde, Thomas
Graham Building, 295 Cathedral Street, Glasgow, G1 1XL, U.K
| | - Kenneth D. Down
- GlaxoSmithKline, Medicines Research Centre, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, U.K
| | - David J. Hirst
- GlaxoSmithKline, Medicines Research Centre, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, U.K
| | - Craig Jamieson
- Department
of Pure and Applied Chemistry, University of Strathclyde, Thomas
Graham Building, 295 Cathedral Street, Glasgow, G1 1XL, U.K
| | - Eric S. Manas
- GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, Pennsylvania 19426-0989, United States
| | - Vipulkumar K. Patel
- GlaxoSmithKline, Medicines Research Centre, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, U.K
| | - Don O. Somers
- GlaxoSmithKline, Medicines Research Centre, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, U.K
| |
Collapse
|
12
|
Val279Phe variant of Lp-PLA2 is a risk factor for a subpopulation of Indonesia patients with acute myocardial infarction. Genes Dis 2016; 3:289-293. [PMID: 30258899 PMCID: PMC6147166 DOI: 10.1016/j.gendis.2016.08.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 08/29/2016] [Indexed: 11/20/2022] Open
Abstract
Lipoprotein-associated phospholipase A2 (Lp-PLA2), a member of the phospholipase A2 superfamily, is an enzyme that hydrolyses phospholipids, is found in blood circulation as a sign of inflammation, and takes a role in atherogenesis. There is an epidemiologic relation between increased Lp-PLA2 levels and coronary heart disease. Lp-PLA2 is an enzyme that is produced by macrophages and takes a role as an independent predictor of a coronary event. A genetic variant of Val279Phe on the Lp-PLA2 gene has been reported with various results in Japan, China, Korea, and Caucasian populations. This study aims to analyse the influence of the Val279Phe genetic variant on acute myocardial infarction (AMI) at Saiful Anwar Hospital, Indonesia. This study was conducted on 151 patients (111 AMI patients and 40 non-AMI patients). The genetic variant of Val279Phe was identified through a genotyping method. There were no significant differences in age, total cholesterol level, LDL-C (low-density lipoprotein cholesterol) level, and family history data between AMI and non-AMI patients. However, AMI patients had low HDL-C (high-density lipoprotein cholesterol), triglyceride levels, dyslipidaemia, and hypertension risk factors compared to non-AMI patients. The frequency of the GG genotype (279Val) was dominant in both AMI and non-AMI groups. Further analysis suggested that the GG genotype has a 2.9 times greater risk of AMI compared to the GT/TT genotype (279Phe). This study concluded that the Val279Phe genetic variant undoubtedly influenced AMI risk, which is a warrant for further development of early detection and improving strategy to prevent AMI in patients.
Collapse
|
13
|
Wallentin L, Held C, Armstrong PW, Cannon CP, Davies RY, Granger CB, Hagström E, Harrington RA, Hochman JS, Koenig W, Krug-Gourley S, Mohler ER, Siegbahn A, Tarka E, Steg PG, Stewart RAH, Weiss R, Östlund O, White HD. Lipoprotein-Associated Phospholipase A2 Activity Is a Marker of Risk But Not a Useful Target for Treatment in Patients With Stable Coronary Heart Disease. J Am Heart Assoc 2016; 5:e003407. [PMID: 27329448 PMCID: PMC4937279 DOI: 10.1161/jaha.116.003407] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 05/19/2016] [Indexed: 01/20/2023]
Abstract
BACKGROUND We evaluated lipoprotein-associated phospholipase A2 (Lp-PLA2) activity in patients with stable coronary heart disease before and during treatment with darapladib, a selective Lp-PLA2 inhibitor, in relation to outcomes and the effects of darapladib in the STABILITY trial. METHODS AND RESULTS Plasma Lp-PLA2 activity was determined at baseline (n=14 500); at 1 month (n=13 709); serially (n=100) at 3, 6, and 18 months; and at the end of treatment. Adjusted Cox regression models evaluated associations between Lp-PLA2 activity levels and outcomes. At baseline, the median Lp-PLA2 level was 172.4 μmol/min per liter (interquartile range 143.1-204.2 μmol/min per liter). Comparing the highest and lowest Lp-PLA2 quartile groups, the hazard ratios were 1.50 (95% CI 1.23-1.82) for the primary composite end point (cardiovascular death, myocardial infarction, or stroke), 1.95 (95% CI 1.29-2.93) for hospitalization for heart failure, 1.42 (1.07-1.89) for cardiovascular death, and 1.37 (1.03-1.81) for myocardial infarction after adjustment for baseline characteristics, standard laboratory variables, and other prognostic biomarkers. Treatment with darapladib led to a ≈65% persistent reduction in median Lp-PLA2 activity. There were no associations between on-treatment Lp-PLA2 activity or changes of Lp-PLA2 activity and outcomes, and there were no significant interactions between baseline and on-treatment Lp-PLA2 activity or changes in Lp-PLA2 activity levels and the effects of darapladib on outcomes. CONCLUSIONS Although high Lp-PLA2 activity was associated with increased risk of cardiovascular events, pharmacological lowering of Lp-PLA2 activity by ≈65% did not significantly reduce cardiovascular events in patients with stable coronary heart disease, regardless of the baseline level or the magnitude of change of Lp-PLA2 activity. CLINICAL TRIAL REGISTRATION URL: https://www.clinicaltrials.gov. Unique identifier: NCT00799903.
Collapse
Affiliation(s)
- Lars Wallentin
- Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden Uppsala Clinical Research Center (UCR), Uppsala University, Uppsala, Sweden
| | - Claes Held
- Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden Uppsala Clinical Research Center (UCR), Uppsala University, Uppsala, Sweden
| | | | - Christopher P Cannon
- Cardiovascular Division, Brigham and Women's Hospital, Boston, MA Harvard Clinical Research Institute, Boston, MA
| | - Richard Y Davies
- Metabolic Pathways and Cardiovascular Therapeutic Area, GlaxoSmithKline, King of Prussia, PA
| | | | - Emil Hagström
- Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden Uppsala Clinical Research Center (UCR), Uppsala University, Uppsala, Sweden
| | | | - Judith S Hochman
- Department of Medicine, NYU Langone Medical Center, New York, NY
| | - Wolfgang Koenig
- Department of Internal Medicine II-Cardiology, University of Ulm Medical Center, Ulm, Germany Deutsches Herzzentrum München, Technische Universität München, Munich, Germany DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Sue Krug-Gourley
- Metabolic Pathways and Cardiovascular Therapeutic Area, GlaxoSmithKline, King of Prussia, PA
| | - Emile R Mohler
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Agneta Siegbahn
- Uppsala Clinical Research Center (UCR), Uppsala University, Uppsala, Sweden Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden
| | - Elizabeth Tarka
- Former Employee of Metabolic Pathways and Cardiovascular Therapeutic Area, GlaxoSmithKline, King of Prussia, PA
| | - Philippe Gabriel Steg
- FACT (French Alliance for Cardiovascular Trials), Paris, France DHU FIRE, Université Paris-Diderot, Sorbonne Paris-Cité, Paris, France Hôpital Bichat, INSERUM U-1148, Paris, France NHLI, ICMS, Imperial College, Royal Brompton Hospital, London, UK
| | - Ralph A H Stewart
- Green Lane Cardiovascular Service, Auckland City Hospital, Auckland, New Zealand University of Auckland, New Zealand
| | | | - Ollie Östlund
- Uppsala Clinical Research Center (UCR), Uppsala University, Uppsala, Sweden
| | - Harvey D White
- Green Lane Cardiovascular Service, Auckland City Hospital, Auckland, New Zealand University of Auckland, New Zealand
| |
Collapse
|
14
|
Millwood IY, Bennett DA, Walters RG, Clarke R, Waterworth D, Johnson T, Chen Y, Yang L, Guo Y, Bian Z, Hacker A, Yeo A, Parish S, Hill MR, Chissoe S, Peto R, Cardon L, Collins R, Li L, Chen Z. A phenome-wide association study of a lipoprotein-associated phospholipase A2 loss-of-function variant in 90 000 Chinese adults. Int J Epidemiol 2016; 45:1588-1599. [PMID: 27301456 PMCID: PMC5100610 DOI: 10.1093/ije/dyw087] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/24/2016] [Indexed: 12/20/2022] Open
Abstract
Background: Lipoprotein-associated phospholipase A2 (Lp-PLA2) has been implicated in development of atherosclerosis; however, recent randomized trials of Lp-PLA2 inhibition reported no beneficial effects on vascular diseases. In East Asians, a loss-of-function variant in the PLA2G7 gene can be used to assess the effects of genetically determined lower Lp-PLA2. Methods:PLA2G7 V279F (rs76863441) was genotyped in 91 428 individuals randomly selected from the China Kadoorie Biobank of 0.5 M participants recruited in 2004–08 from 10 regions of China, with 7 years’ follow-up. Linear regression was used to assess effects of V279F on baseline traits. Logistic regression was conducted for a range of vascular and non-vascular diseases, including 41 ICD-10 coded disease categories. Results:PLA2G7 V279F frequency was 5% overall (range 3–7% by region), and 9691 (11%) participants had at least one loss-of-function variant. V279F was not associated with baseline blood pressure, adiposity, blood glucose or lung function. V279F was not associated with major vascular events [7141 events; odds ratio (OR) = 0.98 per F variant, 95% confidence interval (CI) 0.90-1.06] or other vascular outcomes, including major coronary events (922 events; 0.96, 0.79-1.18) and stroke (5967 events; 1.00, 0.92-1.09). Individuals with V279F had lower risks of diabetes (7031 events; 0.91, 0.84-0.98) and asthma (182 events; 0.53, 0.28-0.98), but there was no association after adjustment for multiple testing. Conclusions: Lifelong lower Lp-PLA2 activity was not associated with major risks of vascular or non-vascular diseases in Chinese adults. Using functional genetic variants in large-scale prospective studies with linkage to a range of health outcomes is a valuable approach to inform drug development and repositioning.
Collapse
Affiliation(s)
- Iona Y Millwood
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, UK
| | - Derrick A Bennett
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, UK
| | - Robin G Walters
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, UK
| | - Robert Clarke
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, UK
| | - Dawn Waterworth
- GlaxoSmithKline (GSK) Medicines Research Centre, GSK, Stevenage, UK, Research Triangle Park, NC, USA and King of Prussia, PA, USA
| | - Toby Johnson
- GlaxoSmithKline (GSK) Medicines Research Centre, GSK, Stevenage, UK, Research Triangle Park, NC, USA and King of Prussia, PA, USA
| | - Yiping Chen
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, UK
| | - Ling Yang
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, UK
| | - Yu Guo
- Chinese Academy of Medical Sciences, Dong Cheng District, Beijing, China and
| | - Zheng Bian
- Chinese Academy of Medical Sciences, Dong Cheng District, Beijing, China and
| | - Alex Hacker
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, UK
| | - Astrid Yeo
- GlaxoSmithKline (GSK) Medicines Research Centre, GSK, Stevenage, UK, Research Triangle Park, NC, USA and King of Prussia, PA, USA
| | - Sarah Parish
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, UK
| | - Michael R Hill
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, UK
| | - Stephanie Chissoe
- GlaxoSmithKline (GSK) Medicines Research Centre, GSK, Stevenage, UK, Research Triangle Park, NC, USA and King of Prussia, PA, USA
| | - Richard Peto
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, UK
| | - Lon Cardon
- GlaxoSmithKline (GSK) Medicines Research Centre, GSK, Stevenage, UK, Research Triangle Park, NC, USA and King of Prussia, PA, USA
| | - Rory Collins
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, UK
| | - Liming Li
- Chinese Academy of Medical Sciences, Dong Cheng District, Beijing, China and.,Department of Epidemiology & Biostatistics, Peking University Health Science Centre, Beijing, China
| | - Zhengming Chen
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, UK
| | | |
Collapse
|
15
|
Masbuchin AN, Rohman MS, Putri JF, Cahyaningtyas M, Widodo. 279(Val→Phe) Polymorphism of lipoprotein-associated phospholipase A(2) resulted in changes of folding kinetics and recognition to substrate. Comput Biol Chem 2015; 59 Pt A:199-207. [PMID: 26595893 DOI: 10.1016/j.compbiolchem.2015.10.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 10/12/2015] [Accepted: 10/14/2015] [Indexed: 10/22/2022]
Abstract
INTRODUCTION PLA2G7 encodes Lp-PLA2 having role in the formation of atherosclerotic plaques by catalyzing its substrate, phosphatydilcholine (PC), to be pro-inflammatory substances. The increased risk for coronary artery disease (CAD) in Asian population has been related with this enzyme. 279(Val→Phe) variant was reported to have a protective role against CAD due to, in part, secretion defect or loss of enzymatic function. Therefore, We study folding kinetics and enzyme-substrate interaction in 279(Val→Phe) by using clinical and computational biology approach. METHODS Polymorphisms were detected by genotyping among 103 acute myocardial infarction patients and 37 controls. Folding Lp-PLA2 was simulated using GROMACS software by assessing helicity, hydrogen bond formation and stability. The interactions of Lp-PLA2 and its substrate were simulated using Pyrx software followed by molecular dynamics simulation using YASARA software. RESULT Polymorphism of 279(Val→Phe) was represented by the change of nucleotide from G to T of 994th PLA2G7 gene. The folding simulation suggested a decreased percentage of α-helix, hydrogen bond formation, hydrogen bond stability and hydrophobicity in 279(Val→Phe). The PC did not interact with active site of 279(Val→Phe) as paradoxically observed in 279 valine. 279(Val→Phe) polymorphism is likely to cause unstable binding to the substrate and decrease the enzymatic activity as observed in molecular dynamics simulations. The results of our computational biology study supported a protected effect of 279(Val→Phe) Polymorphism showed by the odd ratio for MI of 0.22 (CI 95% 0.035-1.37) in this study. CONCLUSION 279(Val→Phe) Polymorphism of Lp-PLA2 may lead to decrease the enzymatic activity via changes of folding kinetics and recognition to its substrate.
Collapse
Affiliation(s)
- Ainun Nizar Masbuchin
- Department of Biomedical Science, Faculty of Medicine, Brawijaya University, Malang, Indonesia.
| | - Mohammad Saifur Rohman
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Brawijaya University, Malang, Indonesia
| | - Jayarani Fatimah Putri
- Department of Biology, Faculty of Mathematics and Natural Sciences, Brawijaya University, Malang, Indonesia
| | - Miryanti Cahyaningtyas
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Brawijaya University, Malang, Indonesia
| | - Widodo
- Department of Biology, Faculty of Mathematics and Natural Sciences, Brawijaya University, Malang, Indonesia
| |
Collapse
|
16
|
[Lp-PLA2, a biomarker of vascular inflammation and vulnerability of atherosclerosis plaques]. ANNALES PHARMACEUTIQUES FRANÇAISES 2015; 74:190-7. [PMID: 26499399 DOI: 10.1016/j.pharma.2015.09.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2015] [Accepted: 09/09/2015] [Indexed: 01/21/2023]
Abstract
A chronic inflammation is involved in various stages of development of the atherosclerotic plaques. Among the emerging biomarkers of atherogenesis, the lipoprotein-associated phospholipase A2 (Lp-PLA2), formerly known as PAF-acetylhydrolase (McIntyre et al., 2009), hydrolyses the oxidized short chain phospholipids of low-density lipoproteins (LDL), thereby releasing pro-inflammatory mediators (lysophospholipids and oxidized fatty acids). Lp-PLA2, produced by monocytes/macrophages and T-lymphocytes, and mainly associated with LDL (Gazi et al., 2005), is predominantly expressed in the necrotic center of the atherosclerotic plaques and in the macrophage-rich areas (Kolodgie et al., 2006). It would have a predictive role of cardiovascular (CV) events in relation to the vulnerability of atherosclerotic plaques. Determination of Lp-PLA2 has been proposed in the assessment of the CV risk, to ensure a better stratification of populations at intermediate risk for targeted therapy (Davidson et al., 2008). Its proatherogenic role suggested that inhibition of its activity could ensure a better vascular protection in combination with cholesterol-lowering agents. Nevertheless, Lp-PLA2 is not yet a fully validated marker for use in daily clinical practice, especially since the studies using an inhibitor of Lp-PLA2 (darapladib) (STABILITY Investigators et al., 2014; O'Donoghue et al., 2014) did not show any reduction in coronary events. Lp-PLA2 could have a site-specific role in plaque inflammation and development (Fenning et al., 2015). High Lp-PLA2 activity could reflect a response to pro-inflammatory stress characteristic of atherosclerosis (Marathe et al., 2014). This presentation aims at clarifying the involvement of Lp-PLA2 in the pathophysiology of atherosclerosis, and at assessing its interest both as a biomarker for the onset of CV events and as a therapeutic target.
Collapse
|
17
|
Campos CM, Suwannasom P, Koenig W, Serruys PW, Garcia-Garcia HM. Darapladib for the treatment of cardiovascular disease. Expert Rev Cardiovasc Ther 2015; 13:33-48. [PMID: 25521799 DOI: 10.1586/14779072.2015.986466] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Elevated levels of phospholipase A2 have been linked to atherosclerotic plaque progression, instability via promoting inflammation and subsequent acute coronary events. Epidemiological studies have demonstrated the correlation between elevated levels associated phospholipase A2 and cardiovascular events. Therefore, specific inhibition of lipoprotein-associated phospholipase A2 with darapladib has been tested as a therapeutic option for atherosclerosis. The aim of this profile is to review the physiologic aspects of lipoprotein-associated phospholipase A2 and to revisit the clinical evidence of darapladib as therapeutic option for atherosclerosis.
Collapse
Affiliation(s)
- Carlos M Campos
- Department of Interventional Cardiology, Erasmus University Medical Centre, Thoraxcenter, s-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands
| | | | | | | | | |
Collapse
|
18
|
Talmud PJ, Holmes MV. Deciphering the Causal Role of sPLA2s and Lp-PLA2 in Coronary Heart Disease. Arterioscler Thromb Vasc Biol 2015; 35:2281-9. [PMID: 26338298 DOI: 10.1161/atvbaha.115.305234] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 08/19/2015] [Indexed: 11/16/2022]
Abstract
Over the last 10 to 15 years, animal and human observational studies have identified elevated levels of both proinflammatory secretory phospholipase A2-IIA and lipoprotein-associated phospholipase A2 as potential risk factors for coronary heart disease. However, Mendelian randomization, a genetic tool to test causality of a biomarker, and phase III randomized controlled trials of inhibitors of theses enzymes (varespladib and darapladib) converged to indicate that elevated levels are unlikely to be themselves causal of coronary heart disease and that inhibition had little or no clinical utility. The concordance of findings from Mendelian randomization and clinical trials suggests that for these 2 drugs, and for other novel biomarkers in future, validation of potential therapeutic targets by genetic studies (such as Mendelian randomization) before embarking on costly phase III randomized controlled trials could increase efficiency and offset the high risk of drug development, thereby facilitating discovery of new therapeutics and mitigating against the exuberant costs of drug development.
Collapse
Affiliation(s)
- Philippa J Talmud
- From the Center for Cardiovascular Genetics, Institute of Cardiovascular Science, University College London, London, UK (P.J.T.); and Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK (M.V.H.).
| | - Michael V Holmes
- From the Center for Cardiovascular Genetics, Institute of Cardiovascular Science, University College London, London, UK (P.J.T.); and Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK (M.V.H.)
| |
Collapse
|
19
|
Maiolino G, Lenzini L, Pedon L, Cesari M, Seccia TM, Frigo AC, Rossitto G, Caroccia B, Rossi GP. Lipoprotein-associated phospholipase A2 single-nucleotide polymorphisms and cardiovascular events in patients with coronary artery disease. J Cardiovasc Med (Hagerstown) 2015; 16:29-36. [PMID: 24732951 DOI: 10.2459/jcm.0000000000000057] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
AIMS We tested the hypothesis that variations in the PLA2G7 gene encoding the lipoprotein-associated phospholipase A2 (Lp-PLA2), an enzyme deemed to have proatherogenic activity, affect the Lp-PLA2 levels and predicts cardiovascular events. METHODS Using a prospective cohort study design, we investigated incident cardiovascular events as a function of the PLA2G7 gene for rs1805017, rs1805018, and rs1051931 single-nucleotide polymorphisms (SNPs) in 643 randomly selected white patients from the GENICA Study, who at baseline underwent coronary angiography, measurement of Lp-PLA2 mass and activity. Cardiovascular event-free survival was compared across the genotypes by Cox regression, propensity score matching, and haplotype analysis. RESULTS The rs1805018 SNP did not follow the Hardy-Weinberg equilibrium and was not further explored. The rs1805017 GG genotype had a lower Lp-PLA2 mass and a higher Lp-PLA2 activity, thus suggesting that this SNP is functional. Long-term follow-up (median 7.8 years) was obtained in 75% of the cohort and allowed recording of incident cardiovascular events in 25.8% of the patients. On Cox regression analysis, the common rs1805017 GG genotype predicted acute myocardial infarction (AMI) [hazard ratio 1.75, 95% confidence interval (CI) 1.03-2.99, P = 0.041]; this finding was confirmed on propensity score matching (82.6% AMI-free survival in GG vs. 94.4% in GA + AA, P = 0.003). The rs1805017 and rs1051931 G/G haplotype was also associated with AMI (52.7 vs. 42.2%, P = 0.026) and cardiovascular event incidence (49.5 vs. 41.7%, P = 0.025). CONCLUSION In high-risk coronary artery disease patients of European ancestry, the PLA2G7 rs1805017 GG genotype is associated with increased Lp-PLA2 plasma activity and AMI.
Collapse
Affiliation(s)
- Giuseppe Maiolino
- aDepartment of Medicine - DIMED - Internal Medicine 4 bDivisione di Cardiologia, Ospedale di Cittadella, Cittadella cDepartment of Cardiac, Thoracic and Vascular Sciences, University of Padua, Padua, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Garg PK, McClelland RL, Jenny NS, Criqui MH, Greenland P, Rosenson RS, Siscovick DS, Jorgensen N, Cushman M. Lipoprotein-associated phospholipase A2 and risk of incident cardiovascular disease in a multi-ethnic cohort: The multi ethnic study of atherosclerosis. Atherosclerosis 2015; 241:176-82. [PMID: 26004387 PMCID: PMC4504012 DOI: 10.1016/j.atherosclerosis.2015.05.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 04/10/2015] [Accepted: 05/13/2015] [Indexed: 12/23/2022]
Abstract
OBJECTIVE Prospective studies reporting a positive association of lipoprotein-associated phospholipase A2 (Lp-PLA2) mass and activity with incident cardiovascular disease (CVD) have included primarily white individuals. We evaluated associations of Lp-PLA2 and first-time cardiovascular events in a healthy multi-ethnic cohort characterized by presence or absence of baseline subclinical atherosclerosis. METHODS Lp-PLA2 mass and activity were measured at baseline in 5456 participants in the Multi-Ethnic Study of Atherosclerosis. Individuals were characterized for presence of baseline subclinical disease (coronary artery calcium score > 0 or carotid intima-media thickness value > 80th percentile) and followed prospectively for development of CVD events (coronary heart disease, ischemic stroke, and cardiovascular death). RESULTS 516 incident CVD events occurred over median follow-up of 10.2 years. In adjusted Cox proportional hazards models, each higher standard deviation of both Lp-PLA2 activity and mass was associated with an increased risk of cardiovascular events; hazard ratios (HR; 95% confidence intervals (CI)) 1.12 (1.01-1.26) for Lp-PLA2 activity and 1.10 (1.01-1.21) for mass. Associations did not differ by subclinical disease status (p-value for interaction 0.99 for Lp-PLA2 activity and 0.32 for Lp-PLA2 mass) and there was no confounding by subclinical atherosclerosis measures. Associations of Lp-PLA2 activity but not mass were weaker in Chinese participants but there were relatively few events among Chinese in race-stratified analysis. CONCLUSION In this multi-ethnic cohort, Lp-PLA2 was positively associated with CVD risk, regardless of the presence of coronary artery calcium or a thickened carotid-intimal media.
Collapse
Affiliation(s)
- Parveen K Garg
- Division of Cardiovascular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | | | - Nancy S Jenny
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT, USA
| | - Michael H Criqui
- Department of Family & Preventive Medicine, University of California in San Diego, La Jolla, CA, USA
| | - Philip Greenland
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA; Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Robert S Rosenson
- Mount Sinai Heart, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Neal Jorgensen
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Mary Cushman
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT, USA; Department of Medicine, University of Vermont College of Medicine, Burlington, VT, USA.
| |
Collapse
|
21
|
Nonsynonymous polymorphisms in PLA2G7 gene are associated with the risk of coronary heart disease in a southern Chinese population. Mamm Genome 2015; 26:191-9. [PMID: 25690150 DOI: 10.1007/s00335-015-9559-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 01/27/2015] [Indexed: 12/26/2022]
Abstract
Lipoprotein-associated phospholipase A2 (Lp-PLA2) plays an important role in coronary heart disease (CHD). This study was aimed to investigate the associations of polymorphisms (R92H, V279F, I198T, and A379V) in PLA2G7 with CHD. A total of 322 patients with CHD and 414 CHD-free controls were included in the study. Polymorphisms in PLA2G7 were sequenced by DNA Sequencer and statistical analyses were performed to study the associations between polymorphisms and CHD. RH + HH genotype, RH genotype, and H allele of R92H were significantly associated with an increased risk of CHD (P = 0.005, P = 0.009, and P = 0.003, respectively), while no associations were observed between V279F and I198T and CHD (A379V was not analyzed because of deviation from Hardy-Weinberg equilibrium). Correlations between R92H and CHD still existed after adjustment for confounding risk factors of CHD (P = 0.001). Furthermore, stratified analyses showed subgroups of the senior, hypertension, non-smoking, non-diabetics, and male subjects brought a higher risk for CHD (P = 0.015, P = 0.001, P = 0.001, P = 0.002, and P = 0.004, respectively). We also observed a lower level of protective factor HDL-C in CHD patients carrying genotype RH + HH than patients with RR (P = 0.047). Furthermore, we conducted haplotype analysis and detected more harmful effects of haplotypes HVI and RVT as compared with other haplotypes (P = 2.538 × 10(-3) and P = 0.031). These findings indicated that R92H variant in PLA2G7 gene might contribute to CHD susceptibility in a southern Chinese population.
Collapse
|
22
|
Karabina S, Ninio E. Plasma PAFAH/PLA2G7 Genetic Variability, Cardiovascular Disease, and Clinical Trials. ACTA ACUST UNITED AC 2015; 38:145-55. [DOI: 10.1016/bs.enz.2015.09.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
23
|
Jung S, Kim M, Chae JS, Lee SH, Joo J, Lee JH. Carriage of the V279F homozygous genotype, a rare allele, within the gene encoding Lp-PLA2 leads to changes in circulating intermediate metabolites in individuals without metabolic syndrome. J Atheroscler Thromb 2014; 21:1243-52. [PMID: 25078067 DOI: 10.5551/jat.23267] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
AIM Identifying differences in plasma metabolic profiling between Lp-PLA2 279VV and 279FF in individuals without metabolic syndrome (MetS) can be used to elucidate the roles of novel Lp-PLA2 activities in normal physiological processes. METHODS Non-MetS individuals with 279FF (n=36) and age-, sex- and BMI-matched VV subjects (n=36) were included in this analysis. RESULTS The FF subjects exhibited no appreciable enzyme activity. No significant differences were observed between the VV and FF subjects in the serum lipid profiles or hs-CRP, plasma ox-LDL, MDA or urinary 8-epi-PGF2α levels. The FF subjects also showed lower activities of lyso-phosphatidylcholine (lysoPC) (16:0) (p=0.003) and oleamide (p<0.001) and a higher activity of L-tryptophan (p=0.016) than the VV subjects. In addition, the Lp-PLA2 activity positively correlated with the lysoPC (16:0) and lysoPC (18:0) activities and negatively correlated with the PC (16:0/22:6) and L-tryptophan activities in the VV subjects. Furthermore, in the VV subjects, the lysoPC (16:0) and lysoPC (18:0) activities negatively correlated with the presence of PCs containing 14:0/20:2, 14:0/22:4 and 16:0/22:6, while the oleamide activity exhibited a strong positive correlation with lysoPCs and a negative correlation with PCs, whereas the relationship between oleamide and lysoPCs and PCs was weaker in the FF subjects. CONCLUSIONS The present results indicate that the natural absence of the plasma Lp-PLA2 activity due to carriage of the Lp-PLA2 279FF genotype may reduce the generation of lysoPC (16:0) and oleamide and thereby enhance the activity of plasma tryptophan in normal physiological processes.
Collapse
Affiliation(s)
- Saem Jung
- National Leading Research Laboratory of Clinical Nutrigenetics/Nutrigenomics, Department of Food and Nutrition, College of Human Ecology, Yonsei University
| | | | | | | | | | | |
Collapse
|
24
|
Zheng GH, Xiong SQ, Chen HY, Mei LJ, Wang T. Associations of platelet-activating factor acetylhydrolase (PAF-AH) gene polymorphisms with circulating PAF-AH levels and risk of coronary heart disease or blood stasis syndrome in the Chinese Han population. Mol Biol Rep 2014; 41:7141-51. [PMID: 25034894 DOI: 10.1007/s11033-014-3597-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2013] [Accepted: 07/07/2014] [Indexed: 02/07/2023]
Abstract
The circulating level of platelet-activating factor acetylhydrolase (PAF-AH) is a novel biomarker to predict the presence of coronary heart disease. PAF-AH gene polymorphisms may be responsible for the variance of circulating PAF-AH levels in individuals. However, the association of PAF-AH gene polymorphisms with circulating PAF-AH levels and the susceptibility to coronary heart disease (CHD) remains unsolved. Blood stasis syndrome (BSS) of CHD is the most common type of TCM syndromes, and a previous study discovered its relationship with the elevated circulating PAF-AH levels. However, the association of gene polymorphisms and CHD with BSS is unclear at present. In this study, four polymorphisms (R92H, I198T, A379V, V279F) of the PAF-AH gene were genotyped in 570 CHD patients, of which 299 had BSS. In addition, 317 unaffected individuals from the same hospitals served as controls. Plasma PAF-AH levels were measured in 155 controls and 271 CHD patients selected randomly, including 139 CHD patients with BSS. In the Chinese Han population, plasma PAF-AH levels in CHD patients with BSS or without BSS were significantly higher (12.9 ± 6.5 and 11.1 ± 5.0 μM, respectively) than in controls (9.3 ± 5.2 μM); this difference still remained significant after adjustment for traditional risk factors or the inflammatory factors. The R92H polymorphism was highly related to the plasma PAF-AH levels and the risk of CHD, especially among patients with BSS, even with the adjustment for the effects of traditional factors. The I198T polymorphism was highly associated with risk of CHD with BSS, but was associated with neither the risk of CHD with no BSS nor with elevated plasma PAF-AH levels.
Collapse
Affiliation(s)
- Guo-Hua Zheng
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350108, Fujian, China,
| | | | | | | | | |
Collapse
|
25
|
Maeda T, Takeuchi K, Xiaoling P, P Zankov D, Takashima N, Fujiyoshi A, Kadowaki T, Miura K, Ueshima H, Ogita H. Lipoprotein-associated phospholipase A2 regulates macrophage apoptosis via the Akt and caspase-7 pathways. J Atheroscler Thromb 2014; 21:839-53. [PMID: 24717759 DOI: 10.5551/jat.21386] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
AIM Mutations in lipoprotein-associated phospholipase A2 (Lp-PLA2) are related to atherosclerosis. However, the molecular effects of Lp-PLA2 on atherosclerosis have not been fully investigated. Therefore, this study attempted to elucidate this issue. METHODS Monocytes were isolated from randomly selected healthy male volunteers according to each Lp-PLA2 genotype (wild-type Lp-PLA2 [Lp-PLA2 (V/V)], the heterozygous V279F mutation [LpPLA2 (V/F)] and the homozygous V279F mutation [Lp-PLA2 (F/F)]) and differentiated into macrophages. The level of apoptosis in the macrophages following incubation without serum was measured using the annexin V/propidium iodide double staining method, and the underlying mechanisms were further examined using a culture cell line. RESULTS The average plasma Lp-PLA2 concentration [Lp-PLA2 (V/V): 129.4 ng/mL, Lp-PLA2 (V/F): 70.7 ng/mL, Lp-PLA2 (F/F): 0.4 ng/mL] and activity [Lp-PLA2 (V/V): 164.3 nmol/min/mL, LpPLA2 (V/F): 100.9 nmol/min/mL, Lp-PLA2 (F/F): 11.6 nmol/min/mL] were significantly different between each genotype, although the basic clinical characteristics were similar. The percentage of apoptotic cells was significantly higher among the Lp-PLA2 (F/F) macrophages compared with that observed in the Lp-PLA2 (V/V) macrophages. This induction of apoptosis was independent of the actions of acetylated low-density lipoproteins. In addition, the transfection of the expression plasmid of V279F mutant Lp-PLA2 into Cos-7 cells or monocyte/macrophage-like U937 cells promoted apoptosis. The knockdown of Lp-PLA2 also increased the number of apoptotic cells. Among the cells expressing mutant Lp-PLA2, the caspase-7 activity was increased, while the activated Akt level was decreased. CONCLUSIONS The V279F mutation of Lp-PLA2 positively regulates the induction of apoptosis in macrophages and Cos-7 cells. An increase in the caspase-7 activity and a reduction in the activated Akt level are likely to be involved in this phenomenon.
Collapse
Affiliation(s)
- Toshinaga Maeda
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Steen DL, O'Donoghue ML. Lp-PLA2 Inhibitors for the Reduction of Cardiovascular Events. Cardiol Ther 2013; 2:125-34. [PMID: 25135391 PMCID: PMC4107429 DOI: 10.1007/s40119-013-0022-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Indexed: 12/14/2022] Open
Abstract
Evidence suggests that inflammation plays a central role in the pathogenesis of atherosclerosis (Libby, Nature 420:868–874, 2002). Inflammation is a physiologic process with highly regulated and often redundant mechanisms to balance pro-inflammatory and anti-inflammatory responses. The complexity of these networks has made it challenging to identify those specific pathways or key enzymes that contribute directly to atherogenesis and could act as a valuable therapeutic target. Lipoprotein-associated phospholipase A2 (Lp-PLA2) is a member of the phospholipase A2 family of enzymes and is believed to contribute to atherosclerotic plaque progression and instability by promoting inflammation. A large number of epidemiologic studies have demonstrated that elevated levels of Lp-PLA2 are associated with an increased risk of cardiovascular events across diverse patient populations, independent of established risk factors including low-density lipoprotein cholesterol. Further, a growing number of preclinical and genetic studies support a causal role for Lp-PLA2 in atherosclerosis. The development of a novel therapeutic agent that directly inhibits the Lp-PLA2 enzyme has provided a unique opportunity to directly test the hypothesis that inhibition of this inflammatory enzyme will translate into improved clinical outcomes. In this article, we will review the evidence to support the notion that Lp-PLA2 is causally implicated in the pathobiology of atherogenesis and discuss the potential utility of inhibiting this enzyme as a therapeutic target.
Collapse
Affiliation(s)
- Dylan L Steen
- TIMI Study Group, Cardiovascular Division, Brigham and Women's Hospital, Boston, MA, USA
| | | |
Collapse
|
27
|
Xu L, Zhou J, Huang S, Huang Y, LE Y, Jiang D, Wang F, Yang X, Xu W, Huang X, Dong C, Zhang L, Ye M, Lian J, Duan S. An association study between genetic polymorphisms related to lipoprotein-associated phospholipase A(2) and coronary heart disease. Exp Ther Med 2013; 5:742-750. [PMID: 23404648 PMCID: PMC3570076 DOI: 10.3892/etm.2013.911] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2012] [Accepted: 01/14/2013] [Indexed: 01/06/2023] Open
Abstract
Previous genome-wide association studies (GWAS) have revealed seven single nucleotide polymorphisms (SNPs) that affect lipoprotein-associated phospholipase A2 (Lp-PLA2) activity or levels in American and European individuals. A total of 290 coronary heart disease (CHD) patients, 198 non-CHD patients and 331 unrelated healthy volunteers were recruited for the present case-control study of Han Chinese. Four SNPs (rs964184 of ZNF259, rs7528419 of CELSR2 and rs7756935 and rs1805017 of PLA2G7) were shown to be significantly associated with CHD. The rs964184-G allele of the ZNF259 gene was identified as a risk factor of CHD in females (odds ratio (OR) =1.49, 95% confidence interval (CI) =1.00–2.22, P=0.05). The rs7528419-G allele of the CELSR2 gene was protective against CHD in males (OR=0.48, 95% CI=0.25–0.93, P=0.04). The other two alleles (rs7756935-C and rs1805017-A) of the PLA2G7 gene acted as protective factors against CHD in females (rs7756935-C: OR=0.59, 95% CI=0.35–1.00, P=0.05; rs1805017-A: OR=0.51, 95% CI=0.28–0.93, P=0.03). Moreover, rs1805017 of the PLA2G7 gene was associated with the severity of CHD only in females (r2=0.02, P=0.04). We identified four Lp-PLA2-associated SNPs significantly associated with CHD in a Han Chinese population. Specifically, rs7528419 was protective factor against CHD in males, while the other two SNPs (rs7756935 and rs1805017 of the PLA2G7 gene) were protective factors against CHD in females and rs964184 of the ZNF259 gene was regarded as a risk factor for CHD in females.
Collapse
Affiliation(s)
- Limin Xu
- School of Medicine, The Affiliated Hospital, Ningbo University, Ningbo, Zhejiang 315211
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Nagano JMG, Hsu KL, Whitby LR, Niphakis MJ, Speers AE, Brown SJ, Spicer T, Fernandez-Vega V, Ferguson J, Hodder P, Srinivasan P, Gonzalez TD, Rosen H, Bahnson BJ, Cravatt BF. Selective inhibitors and tailored activity probes for lipoprotein-associated phospholipase A(2). Bioorg Med Chem Lett 2012; 23:839-43. [PMID: 23260346 DOI: 10.1016/j.bmcl.2012.11.061] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Accepted: 11/14/2012] [Indexed: 01/09/2023]
Abstract
Lipoprotein-associated phospholipase A(2) (Lp-PLA(2) or PLA(2)G7) binds to low-density lipoprotein (LDL) particles, where it is thought to hydrolyze oxidatively truncated phospholipids. Lp-PLA(2) has also been implicated as a pro-tumorigenic enzyme in human prostate cancer. Several inhibitors of Lp-PLA(2) have been described, including darapladib, which is currently in phase 3 clinical development for the treatment of atherosclerosis. The selectivity that darapladib and other Lp-PLA(2) inhibitors display across the larger serine hydrolase family has not, however, been reported. Here, we describe the use of both general and tailored activity-based probes for profiling Lp-PLA(2) and inhibitors of this enzyme in native biological systems. We show that both darapladib and a novel class of structurally distinct carbamate inhibitors inactivate Lp-PLA(2) in mouse tissues and human cell lines with high selectivity. Our findings thus identify both inhibitors and chemoproteomic probes that are suitable for investigating Lp-PLA(2) function in biological systems.
Collapse
Affiliation(s)
- Joseph M G Nagano
- The Department of Chemical Physiology, The Scripps Research Institute, 10550 N. Torrey Pines Rd. La Jolla, CA 92037, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Chu AY, Guilianini F, Grallert H, Dupuis J, Ballantyne CM, Barratt BJ, Nyberg F, Chasman DI, Ridker PM. Genome-Wide Association Study Evaluating Lipoprotein-Associated Phospholipase A
2
Mass and Activity at Baseline and After Rosuvastatin Therapy. ACTA ACUST UNITED AC 2012; 5:676-85. [DOI: 10.1161/circgenetics.112.963314] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Background—
Lipoprotein-associated phospholipase A
2
(Lp-PLA
2
) is a proinflammatory enzyme bound to low-density lipoprotein cholesterol and other circulating lipoproteins. Two measures of Lp-PLA
2
, mass and activity, are associated with increased cardiovascular risk. Data are sparse regarding genetic determinants of Lp-PLA
2
mass and activity, and no prior data are available addressing genetic determinants of statin-induced changes for this proinflammatory biomarker.
Methods and Results
—
We performed a genome-wide association study of Lp-PLA
2
mass and activity at baseline and after 12 months of rosuvastatin therapy (20 mg/d) among 6851 participants of European ancestry from the Justification for Use of Statins in Prevention: an Intervention Trial Evaluating Rosuvastatin (JUPITER) and performed replication in a meta-analysis of 13 664 participants from the Cohorts for Heart and Aging Research in Genomic Epidemiology (CHARGE) Consortium. Novel associations were identified and replicated at
MS4A4E
and
TMEM49
for baseline Lp-PLA
2
activity with genome-wide significant joint
P
values (
P
=2.0×10
−11
and
P
=2.9×10
−9
, respectively). In addition, genome-wide associations (
P
<5×10
−8
) were identified and replicated for baseline Lp-PLA
2
mass at
CETP
and for Lp-PLA
2
activity at the
APOC1-APOE
and
PLA2G7
loci. Among 2673 statin-allocated participants, both Lp-PLA
2
mass and activity were reduced by >30% and low-density lipoprotein cholesterol by 50% after 12 months of statin therapy (
P
<0.001 for both). Variants in
ABCG2
and
LPA
were associated with change in statin-induced Lp-PLA
2
activity at genome-wide significance but were substantially attenuated after adjustment for statin-induced changes in lipid levels.
Conclusions—
Genome-wide significant associations at
MS4A4E
and
TMEM49
may reflect novel influences on circulating levels of Lp-PLA
2
activity. In addition, genome-wide significant associations with rosuvastatin-induced change in Lp-PLA
2
activity were observed in
ABCG2
and
LPA
, likely because of their impact on statin-induced low-density lipoprotein cholesterol lowering.
Collapse
Affiliation(s)
- Audrey Y. Chu
- From the Center for Cardiovascular Disease Prevention, the Division of Preventive Medicine (A.Y.C., F.G., D.I.C., P.M.R.), Division of Genetics (D.I.C.), and Division of Cardiology (P.M.R.), Brigham and Women’s Hospital, Boston, MA; Helmholtz ZentrumMünchen, German Research Center for Environmental Health, Research Unit of Molecular Epidemiology, Neuherberg, Germany (H.G.); National Heart, Lung, and Blood Institute’s and Boston University’s Framingham Heart Study, Framingham, MA (J.D.); Department
| | - Franco Guilianini
- From the Center for Cardiovascular Disease Prevention, the Division of Preventive Medicine (A.Y.C., F.G., D.I.C., P.M.R.), Division of Genetics (D.I.C.), and Division of Cardiology (P.M.R.), Brigham and Women’s Hospital, Boston, MA; Helmholtz ZentrumMünchen, German Research Center for Environmental Health, Research Unit of Molecular Epidemiology, Neuherberg, Germany (H.G.); National Heart, Lung, and Blood Institute’s and Boston University’s Framingham Heart Study, Framingham, MA (J.D.); Department
| | - Harald Grallert
- From the Center for Cardiovascular Disease Prevention, the Division of Preventive Medicine (A.Y.C., F.G., D.I.C., P.M.R.), Division of Genetics (D.I.C.), and Division of Cardiology (P.M.R.), Brigham and Women’s Hospital, Boston, MA; Helmholtz ZentrumMünchen, German Research Center for Environmental Health, Research Unit of Molecular Epidemiology, Neuherberg, Germany (H.G.); National Heart, Lung, and Blood Institute’s and Boston University’s Framingham Heart Study, Framingham, MA (J.D.); Department
| | - Josée Dupuis
- From the Center for Cardiovascular Disease Prevention, the Division of Preventive Medicine (A.Y.C., F.G., D.I.C., P.M.R.), Division of Genetics (D.I.C.), and Division of Cardiology (P.M.R.), Brigham and Women’s Hospital, Boston, MA; Helmholtz ZentrumMünchen, German Research Center for Environmental Health, Research Unit of Molecular Epidemiology, Neuherberg, Germany (H.G.); National Heart, Lung, and Blood Institute’s and Boston University’s Framingham Heart Study, Framingham, MA (J.D.); Department
| | - Christie M. Ballantyne
- From the Center for Cardiovascular Disease Prevention, the Division of Preventive Medicine (A.Y.C., F.G., D.I.C., P.M.R.), Division of Genetics (D.I.C.), and Division of Cardiology (P.M.R.), Brigham and Women’s Hospital, Boston, MA; Helmholtz ZentrumMünchen, German Research Center for Environmental Health, Research Unit of Molecular Epidemiology, Neuherberg, Germany (H.G.); National Heart, Lung, and Blood Institute’s and Boston University’s Framingham Heart Study, Framingham, MA (J.D.); Department
| | - Bryan J. Barratt
- From the Center for Cardiovascular Disease Prevention, the Division of Preventive Medicine (A.Y.C., F.G., D.I.C., P.M.R.), Division of Genetics (D.I.C.), and Division of Cardiology (P.M.R.), Brigham and Women’s Hospital, Boston, MA; Helmholtz ZentrumMünchen, German Research Center for Environmental Health, Research Unit of Molecular Epidemiology, Neuherberg, Germany (H.G.); National Heart, Lung, and Blood Institute’s and Boston University’s Framingham Heart Study, Framingham, MA (J.D.); Department
| | - Fredrik Nyberg
- From the Center for Cardiovascular Disease Prevention, the Division of Preventive Medicine (A.Y.C., F.G., D.I.C., P.M.R.), Division of Genetics (D.I.C.), and Division of Cardiology (P.M.R.), Brigham and Women’s Hospital, Boston, MA; Helmholtz ZentrumMünchen, German Research Center for Environmental Health, Research Unit of Molecular Epidemiology, Neuherberg, Germany (H.G.); National Heart, Lung, and Blood Institute’s and Boston University’s Framingham Heart Study, Framingham, MA (J.D.); Department
| | - Daniel I. Chasman
- From the Center for Cardiovascular Disease Prevention, the Division of Preventive Medicine (A.Y.C., F.G., D.I.C., P.M.R.), Division of Genetics (D.I.C.), and Division of Cardiology (P.M.R.), Brigham and Women’s Hospital, Boston, MA; Helmholtz ZentrumMünchen, German Research Center for Environmental Health, Research Unit of Molecular Epidemiology, Neuherberg, Germany (H.G.); National Heart, Lung, and Blood Institute’s and Boston University’s Framingham Heart Study, Framingham, MA (J.D.); Department
| | - Paul M. Ridker
- From the Center for Cardiovascular Disease Prevention, the Division of Preventive Medicine (A.Y.C., F.G., D.I.C., P.M.R.), Division of Genetics (D.I.C.), and Division of Cardiology (P.M.R.), Brigham and Women’s Hospital, Boston, MA; Helmholtz ZentrumMünchen, German Research Center for Environmental Health, Research Unit of Molecular Epidemiology, Neuherberg, Germany (H.G.); National Heart, Lung, and Blood Institute’s and Boston University’s Framingham Heart Study, Framingham, MA (J.D.); Department
| |
Collapse
|
30
|
|
31
|
Gregson J, Stirnadel-Farrant HA, Doobaree IU, Koro C. Variation of lipoprotein associated phospholipase A2 across demographic characteristics and cardiovascular risk factors: a systematic review of the literature. Atherosclerosis 2012; 225:11-21. [PMID: 22784637 DOI: 10.1016/j.atherosclerosis.2012.06.020] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2012] [Revised: 06/11/2012] [Accepted: 06/13/2012] [Indexed: 12/23/2022]
Abstract
BACKGROUND Lipoprotein association phospholipase A2 (Lp-PLA(2)), an enzyme which has been found in atherosclerotic plaque is currently under investigation in large Phase III clinical trials of vascular disease prevention. We assessed in a variety of different population settings variation of Lp-PLA(2) mass and activity across gender, ethnicity, diabetes, kidney disease and metabolic syndrome. We also assessed correlations with measures of circulating lipids, systemic inflammation and adiposity. METHODS Systematic review of studies measuring Lp-PLA(2) and at least one of the relevant characteristics in >50 participants. RESULTS We identified a total of 77 studies involving 102,499 participants meeting the inclusion criteria. Lp-PLA(2) mass and activity were consistently approximately 10% higher in males than females and 15% higher in Caucasians than African Americans or Hispanics. There were no clear associations of Lp-PLA(2) mass or activity with type II diabetes, markers of systemic inflammation (C-reactive protein, fibrinogen) or with body mass index. Correlations of Lp-PLA(2) mass or activity with low density lipoprotein cholesterol and apolipoprotein B were moderate and positive, whilst correlations with high density lipoprotein cholesterol were negative and moderate to weak. There was no clear differences in associations with any of the above characteristics in groups defined based upon prevalent cardiovascular disease or its risk factors. CONCLUSIONS Despite considerable variability in absolute levels of Lp-PLA(2) across studies, the variability of Lp-PLA(2) across gender, ethnicity, and levels of circulating lipids and markers of systemic inflammation are more consistent and appear not to vary importantly across categories defined by CVD or its risk factors.
Collapse
Affiliation(s)
- John Gregson
- Department of Public Healthy and Primary Care, University of Cambridge, Cambridge CB1 8RN, United Kingdom.
| | | | | | | |
Collapse
|
32
|
Rosenson RS, Stafforini DM. Modulation of oxidative stress, inflammation, and atherosclerosis by lipoprotein-associated phospholipase A2. J Lipid Res 2012; 53:1767-82. [PMID: 22665167 DOI: 10.1194/jlr.r024190] [Citation(s) in RCA: 115] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Lipoprotein-associated phospholipase A(2) (Lp-PLA(2)), also known as platelet-activating factor acetylhydrolase (PAF-AH), is a unique member of the phospholipase A(2) superfamily. This enzyme is characterized by its ability to specifically hydrolyze PAF as well as glycerophospholipids containing short, truncated, and/or oxidized fatty acyl groups at the sn-2 position of the glycerol backbone. In humans, Lp-PLA(2) circulates in active form as a complex with low- and high-density lipoproteins. Clinical studies have reported that plasma Lp-PLA(2) activity and mass are strongly associated with atherogenic lipids and vascular risk. These observations led to the hypothesis that Lp-PLA(2) activity and/or mass levels could be used as biomarkers of cardiovascular disease and that inhibition of the activity could offer an attractive therapeutic strategy. Darapladib, a compound that inhibits Lp-PLA(2) activity, is anti-atherogenic in mice and other animals, and it decreases atherosclerotic plaque expansion in humans. However, disagreement continues to exist regarding the validity of Lp-PLA(2) as an independent marker of atherosclerosis and a scientifically justified target for intervention. Circulating Lp-PLA(2) mass and activity are associated with vascular risk, but the strength of the association is reduced after adjustment for basal concentrations of the lipoprotein carriers with which the enzyme associates. Genetic studies in humans harboring an inactivating mutation at this locus indicate that loss of Lp-PLA(2) function is a risk factor for inflammatory and vascular conditions in Japanese cohorts. Consistently, overexpression of Lp-PLA(2) has anti-inflammatory and anti-atherogenic properties in animal models. This thematic review critically discusses results from laboratory and animal studies, analyzes genetic evidence, reviews clinical work demonstrating associations between Lp-PLA(2) and vascular disease, and summarizes results from animal and human clinical trials in which administration of darapladib was tested as a strategy for the management of atherosclerosis.
Collapse
|
33
|
Ferguson JF, Hinkle CC, Mehta NN, Bagheri R, Derohannessian SL, Shah R, Mucksavage MI, Bradfield JP, Hakonarson H, Wang X, Master SR, Rader DJ, Li M, Reilly MP. Translational studies of lipoprotein-associated phospholipase A₂ in inflammation and atherosclerosis. J Am Coll Cardiol 2012; 59:764-72. [PMID: 22340269 DOI: 10.1016/j.jacc.2011.11.019] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2011] [Revised: 10/31/2011] [Accepted: 11/16/2011] [Indexed: 01/14/2023]
Abstract
OBJECTIVES This study sought to examine the role of lipoprotein-associated phospholipase A₂ (Lp-PLA₂/PLA2G7) in human inflammation and coronary atherosclerosis. BACKGROUND Lp-PLA₂ has emerged as a potential therapeutic target in coronary heart disease. Data supporting Lp-PLA₂ are indirect and confounded by species differences; whether Lp-PLA₂ is causal in coronary heart disease remains in question. METHODS We examined inflammatory regulation of Lp-PLA₂ during experimental endotoxemia in humans, probed the source of Lp-PLA₂ in human leukocytes under inflammatory conditions, and assessed the relationship of variation in PLA2G7, the gene encoding Lp-PLA₂, with coronary artery calcification. RESULTS In contrast to circulating tumor necrosis factor-alpha and C-reactive protein, blood and monocyte Lp-PLA₂ messenger ribonucleic acid decreased transiently, and plasma Lp-PLA₂ mass declined modestly during endotoxemia. In vitro, Lp-PLA₂ expression increased dramatically during human monocyte to macrophage differentiation and further in inflammatory macrophages and foamlike cells. Despite only a marginal association of single nucleotide polymorphisms in PLA2G7 with Lp-PLA₂ activity or mass, numerous PLA2G7 single nucleotide polymorphisms were associated with coronary artery calcification. In contrast, several single nucleotide polymorphisms in CRP were significantly associated with plasma C-reactive protein levels but had no relation with coronary artery calcification. CONCLUSIONS Circulating Lp-PLA₂ did not increase during acute phase response in humans, whereas inflammatory macrophages and foam cells, but not circulating monocytes, are major leukocyte sources of Lp-PLA₂. Common genetic variation in PLA2G7 is associated with subclinical coronary atherosclerosis. These data link Lp-PLA₂ to atherosclerosis in humans while highlighting the challenge in using circulating Lp-PLA₂ as a biomarker of Lp-PLA₂ actions in the vasculature.
Collapse
Affiliation(s)
- Jane F Ferguson
- Cardiovascular Institute, Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104-6160, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Grallert H, Dupuis J, Bis JC, Dehghan A, Barbalic M, Baumert J, Lu C, Smith NL, Uitterlinden AG, Roberts R, Khuseyinova N, Schnabel RB, Rice KM, Rivadeneira F, Hoogeveen RC, Fontes JD, Meisinger C, Keaney JF, Lemaitre R, Aulchenko YS, Vasan RS, Ellis S, Hazen SL, van Duijn CM, Nelson JJ, März W, Schunkert H, McPherson RM, Stirnadel-Farrant HA, Psaty BM, Gieger C, Siscovick D, Hofman A, Illig T, Cushman M, Yamamoto JF, Rotter JI, Larson MG, Stewart AF, Boerwinkle E, Witteman JC, Tracy RP, Koenig W, Benjamin EJ, Ballantyne CM. Eight genetic loci associated with variation in lipoprotein-associated phospholipase A2 mass and activity and coronary heart disease: meta-analysis of genome-wide association studies from five community-based studies. Eur Heart J 2012; 33:238-51. [PMID: 22003152 PMCID: PMC3258449 DOI: 10.1093/eurheartj/ehr372] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2010] [Revised: 08/16/2011] [Accepted: 09/09/2011] [Indexed: 12/20/2022] Open
Abstract
AIMS Lipoprotein-associated phospholipase A2 (Lp-PLA2) generates proinflammatory and proatherogenic compounds in the arterial vascular wall and is a potential therapeutic target in coronary heart disease (CHD). We searched for genetic loci related to Lp-PLA2 mass or activity by a genome-wide association study as part of the Cohorts for Heart and Aging Research in Genomic Epidemiology (CHARGE) Consortium. METHODS AND RESULTS In meta-analyses of findings from five population-based studies, comprising 13 664 subjects, variants at two loci (PLA2G7, CETP) were associated with Lp-PLA2 mass. The strongest signal was at rs1805017 in PLA2G7 [P = 2.4 × 10(-23), log Lp-PLA2 difference per allele (beta): 0.043]. Variants at six loci were associated with Lp-PLA2 activity (PLA2G7, APOC1, CELSR2, LDL, ZNF259, SCARB1), among which the strongest signals were at rs4420638, near the APOE-APOC1-APOC4-APOC2 cluster [P = 4.9 × 10(-30); log Lp-PLA2 difference per allele (beta): -0.054]. There were no significant gene-environment interactions between these eight polymorphisms associated with Lp-PLA2 mass or activity and age, sex, body mass index, or smoking status. Four of the polymorphisms (in APOC1, CELSR2, SCARB1, ZNF259), but not PLA2G7, were significantly associated with CHD in a second study. CONCLUSION Levels of Lp-PLA2 mass and activity were associated with PLA2G7, the gene coding for this protein. Lipoprotein-associated phospholipase A2 activity was also strongly associated with genetic variants related to low-density lipoprotein cholesterol levels.
Collapse
Affiliation(s)
- Harald Grallert
- Helmholtz Zentrum München, German Research Center for Environmental Health, Institute of Epidemiology, Neuherberg, Germany
| | - Josée Dupuis
- National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, 73 Mount Wayte Ave. Suite 2, Framingham, MA, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Joshua C. Bis
- Cardiovascular Health Research Unit and Department of Medicine, University of Washington, Seattle, WA, USA
| | - Abbas Dehghan
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Member of the Netherlands Consortium on Healthy Aging (NCHA), Leiden, The Netherlands
| | - Maja Barbalic
- Human Genetics Center, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Jens Baumert
- Helmholtz Zentrum München, German Research Center for Environmental Health, Institute of Epidemiology, Neuherberg, Germany
| | - Chen Lu
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Nicholas L. Smith
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Seattle Epidemiologic Research and Information Center of the Department of Veterans Affairs Office of Research and Development, Seattle, WA, USA
- Group Health Research Institute, Group Health Cooperative, Seattle, WA, USA
| | - André G. Uitterlinden
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Member of the Netherlands Consortium on Healthy Aging (NCHA), Leiden, The Netherlands
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Robert Roberts
- John & Jennifer Ruddy Canadian Cardiovascular Genetics Centre, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Natalie Khuseyinova
- Department of Internal Medicine II–Cardiology, University of Ulm Medical Center, Albert-Einstein-Allee 23, Ulm D-89081, Germany
| | - Renate B. Schnabel
- National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, 73 Mount Wayte Ave. Suite 2, Framingham, MA, USA
| | - Kenneth M. Rice
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Fernando Rivadeneira
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Member of the Netherlands Consortium on Healthy Aging (NCHA), Leiden, The Netherlands
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Ron C. Hoogeveen
- Division of Atherosclerosis and Vascular Medicine, Department of Medicine, Baylor College of Medicine, Methodist DeBakey Heart and Vascular Center, 6565 Fannin, MS A-601, Houston, TX, USA
| | - João Daniel Fontes
- National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, 73 Mount Wayte Ave. Suite 2, Framingham, MA, USA
- Section of Preventive Medicine and Cardiology, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Christa Meisinger
- Helmholtz Zentrum München, German Research Center for Environmental Health, Institute of Epidemiology, Neuherberg, Germany
| | - John F. Keaney
- Division of Cardiovascular Medicine, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Rozenn Lemaitre
- Cardiovascular Health Research Unit and Department of Medicine, University of Washington, Seattle, WA, USA
| | - Yurii S. Aulchenko
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Ramachandran S. Vasan
- National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, 73 Mount Wayte Ave. Suite 2, Framingham, MA, USA
| | | | | | - Cornelia M. van Duijn
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Member of the Netherlands Consortium on Healthy Aging (NCHA), Leiden, The Netherlands
| | - Jeanenne J. Nelson
- Worldwide Epidemiology, GlaxoSmithKline, Research Triangle Park, NC, USA
| | - Winfried März
- Synlab Center of Laboratory Diagnostics Heidelberg, Heidelberg, Germany
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
- Institute of Public Health, Social and Preventive Medicine, Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany
| | | | - Ruth M. McPherson
- John & Jennifer Ruddy Canadian Cardiovascular Genetics Centre, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | | | - Bruce M. Psaty
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Group Health Research Institute, Group Health Cooperative, Seattle, WA, USA
- Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Health Services, University of Washington, Seattle, WA, USA
| | - Christian Gieger
- Helmholtz Zentrum München, German Research Center for Environmental Health, Institute of Epidemiology, Neuherberg, Germany
| | - David Siscovick
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Albert Hofman
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Member of the Netherlands Consortium on Healthy Aging (NCHA), Leiden, The Netherlands
| | - Thomas Illig
- Helmholtz Zentrum München, German Research Center for Environmental Health, Institute of Epidemiology, Neuherberg, Germany
- Ludwig-Maximilians University Munich, Institute of Medical Data Management, Biometrics and Epidemiology, Chair of Epidemiology, Munich, Germany
| | - Mary Cushman
- Department of Medicine, University of Vermont, Burlington, VT, USA
| | - Jennifer F. Yamamoto
- National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, 73 Mount Wayte Ave. Suite 2, Framingham, MA, USA
| | - Jerome I. Rotter
- Medical Genetics Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Martin G. Larson
- National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, 73 Mount Wayte Ave. Suite 2, Framingham, MA, USA
| | - Alexandre F.R. Stewart
- John & Jennifer Ruddy Canadian Cardiovascular Genetics Centre, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Eric Boerwinkle
- Human Genetics Center, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Jacqueline C.M. Witteman
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Member of the Netherlands Consortium on Healthy Aging (NCHA), Leiden, The Netherlands
| | - Russell P. Tracy
- Department of Pathology, University of Vermont College of Medicine, Burlington, VT, USA
- Department of Biochemistry, University of Vermont College of Medicine, Burlington, VT, USA
| | - Wolfgang Koenig
- Department of Internal Medicine II–Cardiology, University of Ulm Medical Center, Albert-Einstein-Allee 23, Ulm D-89081, Germany
| | - Emelia J. Benjamin
- National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, 73 Mount Wayte Ave. Suite 2, Framingham, MA, USA
- Section of Preventive Medicine and Cardiology, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Christie M. Ballantyne
- Division of Atherosclerosis and Vascular Medicine, Department of Medicine, Baylor College of Medicine, Methodist DeBakey Heart and Vascular Center, 6565 Fannin, MS A-601, Houston, TX, USA
| |
Collapse
|
35
|
Sergouniotis P, Davidson A, Mackay D, Lenassi E, Li Z, Robson A, Yang X, Kam J, Isaacs T, Holder G, Jeffery G, Beck J, Moore A, Plagnol V, Webster A. Biallelic mutations in PLA2G5, encoding group V phospholipase A2, cause benign fleck retina. Am J Hum Genet 2011; 89:782-91. [PMID: 22137173 DOI: 10.1016/j.ajhg.2011.11.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Revised: 11/02/2011] [Accepted: 11/07/2011] [Indexed: 10/14/2022] Open
Abstract
Flecked-retina syndromes, including fundus flavimaculatus, fundus albipunctatus, and benign fleck retina, comprise a group of disorders with widespread or limited distribution of yellow-white retinal lesions of various sizes and configurations. Three siblings who have benign fleck retina and were born to consanguineous parents are the basis of this report. A combination of homozygosity mapping and exome sequencing helped to identify a homozygous missense mutation, c.133G>T (p.Gly45Cys), in PLA2G5, a gene encoding a secreted phospholipase (group V phospholipase A(2)). A screen of a further four unrelated individuals with benign fleck retina detected biallelic variants in the same gene in three patients. In contrast, no loss of function or common (minor-allele frequency>0.05%) nonsynonymous PLA2G5 variants have been previously reported (EVS, dbSNP, 1000 Genomes Project) or were detected in an internal database of 224 exomes (from subjects with adult onset neurodegenerative disease and without a diagnosis of ophthalmic disease). All seven affected individuals had fundoscopic features compatible with those previously described in benign fleck retina and no visual or electrophysiological deficits. No medical history of major illness was reported. Levels of low-density lipoprotein were mildly elevated in two patients. Optical coherence tomography and fundus autofluorescence findings suggest that group V phospholipase A(2) plays a role in the phagocytosis of photoreceptor outer-segment discs by the retinal pigment epithelium. Surprisingly, immunohistochemical staining of human retinal tissue revealed localization of the protein predominantly in the inner and outer plexiform layers.
Collapse
|
36
|
Dennis EA, Cao J, Hsu YH, Magrioti V, Kokotos G. Phospholipase A2 enzymes: physical structure, biological function, disease implication, chemical inhibition, and therapeutic intervention. Chem Rev 2011; 111:6130-85. [PMID: 21910409 PMCID: PMC3196595 DOI: 10.1021/cr200085w] [Citation(s) in RCA: 856] [Impact Index Per Article: 61.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Edward A. Dennis
- Department of Chemistry and Biochemistry and Pharmacology, School of Medicine, University of California, San Diego, La Jolla, California 92093-0601
| | - Jian Cao
- Department of Chemistry and Biochemistry and Pharmacology, School of Medicine, University of California, San Diego, La Jolla, California 92093-0601
| | - Yuan-Hao Hsu
- Department of Chemistry and Biochemistry and Pharmacology, School of Medicine, University of California, San Diego, La Jolla, California 92093-0601
| | - Victoria Magrioti
- Laboratory of Organic Chemistry, Department of Chemistry, University of Athens, Panepistimiopolis, Athens 15771, Greece
| | - George Kokotos
- Laboratory of Organic Chemistry, Department of Chemistry, University of Athens, Panepistimiopolis, Athens 15771, Greece
| |
Collapse
|
37
|
O'Donoghue ML, Braunwald E, White HD, Serruys P, Steg PG, Hochman J, Maggioni AP, Bode C, Weaver D, Johnson JL, Cicconetti G, Lukas MA, Tarka E, Cannon CP. Study design and rationale for the Stabilization of pLaques usIng Darapladib-Thrombolysis in Myocardial Infarction (SOLID-TIMI 52) trial in patients after an acute coronary syndrome. Am Heart J 2011; 162:613-619.e1. [PMID: 21982651 DOI: 10.1016/j.ahj.2011.07.018] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2011] [Accepted: 07/26/2011] [Indexed: 10/17/2022]
Abstract
BACKGROUND Higher levels of lipoprotein-associated phospholipase A(2) (Lp-PLA(2)) are associated with a higher risk of cardiovascular events and may play a causal role in atherogenesis. Darapladib inhibits Lp-PLA(2) activity in plasma and in arterial plaques and may confer clinical benefit in preventing cardiovascular events. STUDY DESIGN The SOLID-TIMI 52 trial is a randomized, double-blind, placebo-controlled, multicenter, event-driven trial. Approximately 13,000 subjects are being randomized to darapladib (160 mg enteric-coated tablet daily) or matching placebo within 30 days of hospitalization with an acute coronary syndrome. The primary end point is the composite of cardiovascular death, nonfatal myocardial infarction, or nonfatal stroke. Secondary end points include major and total coronary events, individual components of the primary end point, and all-cause mortality. The study will continue until approximately 1,500 primary end point events have occurred to achieve 90% power to detect a 15.5% reduction in the primary end point. The median treatment duration is anticipated to be approximately 3 years, with a total study duration of approximately 4.1 years. CONCLUSIONS The SOLID-TIMI 52 trial will determine the clinical benefit of direct inhibition of Lp-PLA(2) activity with darapladib in patients after an acute coronary syndrome.
Collapse
|
38
|
Effects of Dietary Factors on Lipoprotein-Associated Phospholipase A2 (Lp-PLA2). Curr Atheroscler Rep 2011; 13:461-6. [DOI: 10.1007/s11883-011-0201-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|