1
|
Li CX, Sun LC, Wang YQ, Liu TT, Cai JR, Liu H, Ren Z, Yi Z. The associations of candidate gene polymorphisms with aspirin resistance in patients with ischemic disease: a meta-analysis. Hum Genomics 2024; 18:135. [PMID: 39617913 PMCID: PMC11610159 DOI: 10.1186/s40246-024-00699-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 11/18/2024] [Indexed: 12/13/2024] Open
Abstract
BACKGROUND Recently, extensive research has been conducted on the relationship between aspirin gene polymorphisms and aspirin resistance (AR) in patients with ischemic diseases. Among the numerous candidate genes, it remains unclear which ones are significantly associated with AR and could potentially serve as potential biomarkers for genetic testing before aspirin use. METHODS Eligible articles were searched in PubMed, Embase, Cochrane Library, WanFang, CNKI and Sinomed. A cohort study examining the efficacy of aspirin in secondary prevention for patients with ischemic diseases, along with a discussion on genetic polymorphisms and their association with AR, has been included. The Newcastle-Ottawa Scale for assessing the quality of included studies. Odds ratios (OR) with 95% confidence intervals (CI) were used as measures of effect. Subgroup analyses were conducted based on different genotypes with the same genetic polymorphisms, different research regions and types of ischemic diseases. RESULTS From 75 eligible articles, 94 candidate gene polymorphisms were analyzed. In the overall analysis, 25 genes were subjected to meta-analysis and 69 genes were systematically described. 23 gene polymorphisms were observed to be significantly associated with AR, including PTGS2(rs20417) (OR = 0.57, 95% CI: 0.44-0.73), ITGA2(rs1126643) (OR = 0.52, 95% CI: 0.29-0.93), and TbXA2R(rs1131882) (OR = 1.54, 95% CI: 1.09-2.18) were obtained from the combined analysis of this study, and 20 genes were systematically described in this study. Further subgroup analyses demonstrated that AA genotype for PTGS1(rs1330344) (OR = 0.56, 95%CI:0.43-0.74), C allele for PTGS1(rs5788) (OR = 0.51, 95%CI: 0.30-0.87) polymorphisms were significantly associated with AR. The polymorphisms of 13 genes, including PTGS1(rs1236913), have been studied only in Asia, GP6(rs1613662) has been studied only in Europe, and the polymorphisms of 5 genes, including ABCB1(rs1045642), showed different correlations with AR in various regions. The individuals with the PTGS1 (rs5788) variant who experienced an ischemic stroke (OR = 0.98, 95%CI: 0.54-1.67) may exhibit an elevated risk of AR compared to those with coronary artery disease (OR = 0.51, 95%CI: 0.3-0.87). CONCLUSIONS Our meta-analysis indicates that PTGS2(rs20417), ITGA2(rs1126643), and TbXA2R(rs1131882) could be potential genetic biomarkers for AR. Among these, PTGS2 (rs20417) is particularly suggested for individuals in Asia with ischemic diseases before aspirin use, as the GC/CC genotype raises AR risk by 42% compared to GG. ITGA2 (rs1126643) increases AR risk by 48% in Asia with the TC/TC genotype versus CC. However, results for ABCB1(rs1045642) and GP1BA(rs2243093) vary by regions, requiring further research.
Collapse
Affiliation(s)
- Chun-Xing Li
- Department of Pharmacy, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, No.15, Yuquan Road, Haidian District, Beijing, 100049, China.
| | - Li-Chaoyue Sun
- Department of Pharmacy, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, No.15, Yuquan Road, Haidian District, Beijing, 100049, China
| | - Yu-Qiao Wang
- Department of Pharmacy, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, No.15, Yuquan Road, Haidian District, Beijing, 100049, China
| | - Tian-Tian Liu
- Department of Pharmacy, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, No.15, Yuquan Road, Haidian District, Beijing, 100049, China
| | - Jin-Rui Cai
- Beijing Haidian District WanShouLu Community Health Service Center, Beijing, China
| | - Hua Liu
- Department of Pharmacy, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, No.15, Yuquan Road, Haidian District, Beijing, 100049, China
| | - Zhao Ren
- Department of Pharmacy, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, No.15, Yuquan Road, Haidian District, Beijing, 100049, China
| | - Zhanmiao Yi
- Department of Pharmacy, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, China.
- Institute for Drug Evaluation, Peking University Health Science Center, Beijing, China.
| |
Collapse
|
2
|
De Luca G, Verburg A, Hof AV, ten Berg J, Kereiakes DJ, Coller BS, Gibson CM. Current and Future Roles of Glycoprotein IIb-IIIa Inhibitors in Primary Angioplasty for ST-Segment Elevation Myocardial Infarction. Biomedicines 2024; 12:2023. [PMID: 39335537 PMCID: PMC11428685 DOI: 10.3390/biomedicines12092023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/21/2024] [Accepted: 08/26/2024] [Indexed: 09/30/2024] Open
Abstract
Acute myocardial infarction still represents the major cause of mortality in high-income countries. Therefore, considerable efforts have been focused on the treatment of myocardial infarctions in the acute and long-term phase, with special attention being paid to reperfusion strategies and adjunctive antithrombotic therapies. In fact, despite the successful mechanical recanalization of the epicardial conduit, a substantial percentage of patients still experience poor myocardial reperfusion or acute/subacute in-stent thrombosis. Due the delayed onset of action of currently available oral antiplatelet therapies, glycoprotein (GP) IIb-IIIa inhibitors could be expected to improve clinical outcomes, especially when administrated in the early phase of the infarction, due to the larger platelet composition of fresh thrombi, the dynamic nature of early thrombi, and the larger amount of viable myocardium existing in the early, as compared to a delayed, phase. Considerable evidence has accumulated regarding the benefits from GP IIb-IIIa inhibitors on mortality, especially among high-risk patients and when administered as an upstream strategy. Therefore, based on currently available data, GP IIb-IIIa inhibitors can be considered when the drug can be administered within the first 3 h of symptom onset and among high-risk patients (e.g., those with advanced Killip class or an anterior myocardial infarction). Even though it is not universally accepted, in our opinion, this strategy should be implemented in a pre-hospital setting (in an ambulance) or as soon as possible when arriving at the hospital (at the Emergency Room or Coronary Care Unit, irrespective of whether they are in spoke or hub hospitals). A new, second-generation GP IIb-IIIa inhibitor (zalunfiban) appears to be highly suitable as a pre-hospital pharmacological facilitation strategy at the time of first medical contact due to its favourable features, including its simple subcutaneous administration, rapid onset of action (15 min), and limited time of action (with a half-life of ~1 h), which is likely to minimize the risk of bleeding. The ongoing CELEBRATE trial, including 2499 STEMI patients, may potentially provide compelling data to support the upstream treatment of STEMI patients undergoing mechanical reperfusion. In fact, although the current therapeutic target of increased rates of timely reperfusion has been achieved, the future goal in myocardial infarction treatment should be to achieve the most rapid reperfusion prior to primary percutaneous coronary intervention, thus further minimizing myocardial damage, or, in some cases, even preventing it completely, and improving survival.
Collapse
Affiliation(s)
- Giuseppe De Luca
- Division of Cardiology, Polyclinic G. Martino, University of Messina, 98122 Messina, Italy
- Division of Cardiology, IRCSS Hospital Nuovo-Galeazzi Sant’Ambrogio, 20157 Milan, Italy
| | - Ashley Verburg
- Department of Cardiology, St. Antonius Hospital, 3435 CM Nieuwegein, The Netherlands; (A.V.)
| | - Arnoud van’t Hof
- Department of Cardiology, Maastricht University Medical Centre, 6229 HX Maastricht, The Netherlands;
- Cardiovascular Research Institute Maastricht, 6229 ER Maastricht, The Netherlands
| | - Jurrien ten Berg
- Department of Cardiology, St. Antonius Hospital, 3435 CM Nieuwegein, The Netherlands; (A.V.)
| | - Dean J. Kereiakes
- The Carl and Edyth Lindner Research Center, The Christ Hospital, Cincinnati, OH 45219, USA
| | - Barry S. Coller
- Laboratory of Blood and Vascular Biology, Rockefeller University, New York, NY 10065, USA;
| | - Charles Michael Gibson
- Perfuse Study Group, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
3
|
Yazbeck A, Akika R, Awada Z, Zgheib NK. Pharmacogenetic considerations in therapy with novel antiplatelet and anticoagulant agents. Pharmacogenet Genomics 2024; 34:61-72. [PMID: 38372412 DOI: 10.1097/fpc.0000000000000520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Antiplatelets and anticoagulants are extensively used in cardiovascular medicine for the prevention and treatment of thrombosis in the venous and arterial circulations. Wide inter-individual variability has been observed in response to antiplatelets and anticoagulants, which triggered researchers to investigate the genetic basis of this variability. Data from extensive pharmacogenetic studies pointed to strong evidence of association between polymorphisms in candidate genes and the pharmacokinetics and pharmacodynamic action and clinical response of the antiplatelets clopidogrel and the anticoagulant warfarin. In this review, we conducted an extensive search on Medline for the time period of 2009-2023. We also searched the PharmGKB website for levels of evidence of variant-drug combinations and for drug labels and clinical guidelines. We focus on the pharmacogenetics of novel antiplatelets and anticoagulants while excluding acetylsalicylic acid, warfarin and heparins, and discuss the current knowledge with emphasis on the level of evidence.
Collapse
Affiliation(s)
| | - Reem Akika
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Zainab Awada
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Nathalie K Zgheib
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
4
|
Yu Q, Yang J, Wang J, Yu R, Li J, Cheng J, Hu Y, Li Z, Zheng N, Zhang Z, Li X, Wang Y, Du W, Zhu K, Chen X, Su J. DNA methylation profile in the whole blood of acute coronary syndrome patients with aspirin resistance. J Clin Lab Anal 2022; 37:e24821. [PMID: 36550638 PMCID: PMC9833987 DOI: 10.1002/jcla.24821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/07/2022] [Accepted: 12/10/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Aspirin resistance (AR) results in major adverse cardiovascular events, and DNA methylation might participate in the regulation of this pathological process. METHODS In present study, a sum of 35 patients with AR and 35 non-AR (NAR) controls were enrolled. Samples from 5 AR and 5 NAR were evaluated in an 850 BeadChip DNA methylation assay, and another 30 AR versus 30 NAR were evaluated to validate the differentially methylated CpG loci (DML). Then, qRT-PCR was used to investigate the target mRNA expression of genes at CpG loci. Finally, Gene Ontology (GO) as well as Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were performed to reveal the enriched pathways. RESULTS The AR and NAR groups displayed significant differences in DNA methylation at 7707 positions, with 270 hypermethylated sites (e.g., cg09555818 located in APOC2) and 7437 sites hypomethylated sites (e.g., cg26828689 located in SLC12A5). Six DML were validated by pyrosequencing, and it was confirmed that DNA methylation (cg16391727, cg21008208, cg21293749, and cg13945576) was related to the increasing risk of AR. The relative mRNA expression of the ROR1 gene was also associated with AR (p = 0.007), suggesting that the change of cg21293749 in DNA methylation might lead to differential ROR1 mRNA expression, ultimately resulting in AR. Furthermore, the identified differentially methylated sites were associated with the molecular pathways such as circadian rhythms and insulin secretion. CONCLUSION Hence, the distinct DNA methylation might play a vital role in the biological regulation of AR through the pathways such as circadian rhythms.
Collapse
Affiliation(s)
- Qinglin Yu
- Department of Traditional Chinese Internal MedicineNingbo No. 1 HospitalNingboChina
| | - Jin Yang
- Department of GeriatricsNingbo No. 1 HospitalNingboChina
| | - Jiang Wang
- Department of CardiologyNingbo No.1 HospitalNingboChina,Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang ProvinceNingboChina
| | - Ruoyan Yu
- Department of CardiologyNingbo No.1 HospitalNingboChina,Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang ProvinceNingboChina
| | - Jiyi Li
- Department of CardiologyYuyao People's Hospital of Zhejiang ProvinceYuyaoChina
| | - Ji Cheng
- Department of Emergency, HwaMei HospitalUniversity of Chinese Academy of SciencesNingboChina
| | - Yingchu Hu
- Department of CardiologyNingbo No.1 HospitalNingboChina,Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang ProvinceNingboChina
| | - Zhenwei Li
- Department of CardiologyNingbo No.1 HospitalNingboChina,Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang ProvinceNingboChina
| | - Nan Zheng
- Department of Cardiology, HwaMei HospitalUniversity of Chinese Academy of SciencesNingboChina
| | - Zhaoxia Zhang
- Department of CardiologyNingbo No.1 HospitalNingboChina,Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang ProvinceNingboChina
| | - Xiaojing Li
- Department of GeriatricsNingbo No. 1 HospitalNingboChina
| | - Yong Wang
- Department of CardiologyNingbo No.1 HospitalNingboChina,Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang ProvinceNingboChina
| | - Weiping Du
- Department of CardiologyNingbo No.1 HospitalNingboChina,Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang ProvinceNingboChina
| | - Keqi Zhu
- Department of Traditional Chinese Internal MedicineNingbo No. 1 HospitalNingboChina
| | - Xiaomin Chen
- Department of CardiologyNingbo No.1 HospitalNingboChina,Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang ProvinceNingboChina
| | - Jia Su
- Department of CardiologyNingbo No.1 HospitalNingboChina,Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang ProvinceNingboChina
| |
Collapse
|
5
|
García-Alfaro MD, Pérez-Nuñez MI, Amigo MT, Arbona C, Ballesteros MÁ, González-Lamuño D. PlA2 Polymorphism of Platelet Glycoprotein IIb/IIIa and C677T Polymorphism of Methylenetetrahydrofolate Reductase ( MTHFR), but Not Factor V Leiden and Prothrombin G20210A Polymorphisms, Are Associated with More Severe Forms of Legg-Calvé-Perthes Disease. CHILDREN-BASEL 2021; 8:children8070614. [PMID: 34356593 PMCID: PMC8307051 DOI: 10.3390/children8070614] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/14/2021] [Accepted: 07/17/2021] [Indexed: 11/16/2022]
Abstract
The possible association of common polymorphic variants related to thrombophilia (the rs6025(A) allele encoding the Leiden mutation, rs1799963(A), i.e., the G20210A mutation of the prothrombin F2 gene, the rs1801133(T) variant of the methylenetetrahydrofolate reductase (MTHFR) gene that encodes an enzyme involved in folate metabolism, and rs5918(C), i.e., the ‘A2’ allele of the platelet-specific alloantigen system that increases platelet aggregation induced by agonists), with the risk of Legg–Calvé–Perthes disease (LCPD) and the degree of hip involvement (Catterall stages I to IV) was analyzed in a cohort study, including 41 children of ages 2 to 10.9 (mean 5.4, SD 2.2), on the basis of clinical and radiological criteria of LCPD. In 10 of the cases, hip involvement was bilateral; thus, a total of 51 hips were followed-up for a mean of 75.5 months. The distribution of genotypes among patients and 118 controls showed no significant differences, with a slightly increased risk for LCPD in rs6025(A) carriers (OR: 2.9, CI: 0.2–47.8). Regarding the severity of LCPD based on Catterall classification, the rs1801133(T) variant of the MTHFR gene and the rs5918(C) variant of the platelet glycoprotein IIb/IIIa were associated with more severe forms of Perthes disease (Catterall III–IV) (p < 0.05). The four children homozygous for mutated MTHFR had a severe form of the disease (Stage IV of Catterall) and a higher risk of non-favorable outcome (Stulberg IV–V).
Collapse
Affiliation(s)
- María Dolores García-Alfaro
- Department of Orthopaedic Surgery, Hospital Universitario Marqués de Valdecilla, Avda Valdecilla s/n, 39008 Santander, Spain; (M.D.G.-A.); (M.I.P.-N.); (C.A.)
| | - María Isabel Pérez-Nuñez
- Department of Orthopaedic Surgery, Hospital Universitario Marqués de Valdecilla, Avda Valdecilla s/n, 39008 Santander, Spain; (M.D.G.-A.); (M.I.P.-N.); (C.A.)
| | - María Teresa Amigo
- Laboratorio de Pediatría, Departamento de Ciencias Médicas y Quirúrgicas, Universidad de Cantabria, Cardenal Herrera Oria s/n, 39011 Santander, Spain;
| | - Carmelo Arbona
- Department of Orthopaedic Surgery, Hospital Universitario Marqués de Valdecilla, Avda Valdecilla s/n, 39008 Santander, Spain; (M.D.G.-A.); (M.I.P.-N.); (C.A.)
| | - María Ángeles Ballesteros
- Department of Critical Care Medicine, Hospital Marqués de Valdecilla-IDIVAL, Avda Valdecilla s/n, 39008 Santander, Spain;
| | - Domingo González-Lamuño
- Laboratorio de Pediatría, Departamento de Ciencias Médicas y Quirúrgicas, Universidad de Cantabria, Cardenal Herrera Oria s/n, 39011 Santander, Spain;
- Division of Pediatrics, Hospital Universitario Marqués de Valdecilla-IDIVAL, Avda Valdecilla s/n, 39008 Santander, Spain
- Correspondence: ; Tel.: +34-942-202-604
| |
Collapse
|
6
|
MacKeigan DT, Ni T, Shen C, Stratton TW, Ma W, Zhu G, Bhoria P, Ni H. Updated Understanding of Platelets in Thrombosis and Hemostasis: The Roles of Integrin PSI Domains and their Potential as Therapeutic Targets. Cardiovasc Hematol Disord Drug Targets 2021; 20:260-273. [PMID: 33001021 DOI: 10.2174/1871529x20666201001144541] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 07/20/2020] [Accepted: 07/26/2020] [Indexed: 11/22/2022]
Abstract
Platelets are small blood cells known primarily for their ability to adhere and aggregate at injured vessels to arrest bleeding. However, when triggered under pathological conditions, the same adaptive mechanism of platelet adhesion and aggregation may cause thrombosis, a primary cause of heart attack and stroke. Over recent decades, research has made considerable progress in uncovering the intricate and dynamic interactions that regulate these processes. Integrins are heterodimeric cell surface receptors expressed on all metazoan cells that facilitate cell adhesion, movement, and signaling, to drive biological and pathological processes such as thrombosis and hemostasis. Recently, our group discovered that the plexin-semaphorin-integrin (PSI) domains of the integrin β subunits exert endogenous thiol isomerase activity derived from their two highly conserved CXXC active site motifs. Given the importance of redox reactions in integrin activation and its location in the knee region, this PSI domain activity may be critically involved in facilitating the interconversions between integrin conformations. Our monoclonal antibodies against the β3 PSI domain inhibited its thiol isomerase activity and proportionally attenuated fibrinogen binding and platelet aggregation. Notably, these antibodies inhibited thrombosis without significantly impairing hemostasis or causing platelet clearance. In this review, we will update mechanisms of thrombosis and hemostasis, including platelet versatilities and immune-mediated thrombocytopenia, discuss critical contributions of the newly discovered PSI domain thiol isomerase activity, and its potential as a novel target for anti-thrombotic therapies and beyond.
Collapse
Affiliation(s)
- Daniel T MacKeigan
- Department of Physiology, University of Toronto, Toronto, ON M5S, Canada
| | - Tiffany Ni
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Canada
| | - Chuanbin Shen
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Canada
| | - Tyler W Stratton
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Canada
| | - Wenjing Ma
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Canada
| | - Guangheng Zhu
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Canada
| | - Preeti Bhoria
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Canada
| | - Heyu Ni
- Department of Physiology, University of Toronto, Toronto, ON M5S, Canada
| |
Collapse
|
7
|
Rezaei Dezaki Z, Bagheri R, Pourgheysari B. Association between fibrinogen receptor (Glycoprotein IIb) polymorphism and the risk of venous thromboembolism: a systematic review. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2019. [DOI: 10.1186/s43042-019-0042-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
The fibrinogen receptor is an integrin on the platelet surface and is shaped from two types of glycoprotein (GP) subunits, GPIIb and GPIIIa. Membrane glycoprotein IIb/IIIa plays an important role in platelet function. The gene encoding the glycoprotein IIIa shows a common polymorphism, PLA2 that increases the binding of the receptor to fibrinogen and enhances the platelet aggregation. The clinical impact of PLA2 polymorphism has been studied in some diseases, but the definition of its exact role on venous thromboembolism complications has been challenging. The present systematic review aimed to clarify the association of PLA2 polymorphism and venous thromboembolism.
Main text
In this study, Electronic databases including PubMed, Embase, Scopus, Web of Science, and Cochrane Library were searched. All the assessed studies focused on the relationship between PLA2 polymorphism and venous thromboembolism. Five studies were eligible for systematic review. One study revealed a significant correlation between PLA2 polymorphism and venous thromboembolism. PLA2 polymorphism was associated with deep vein thrombosis in one study and pulmonary thromboembolism in another one.
Conclusion
The published data supported the hypothesis that having the PLA2 polymorphism of GPIIIa may be a risk factor for venous thromboembolism, but the association cannot be concluded; it needs more clinical investigation.
Collapse
|
8
|
Association among PlA1/A2 gene polymorphism, laboratory aspirin resistance and clinical outcomes in patients with coronary artery disease: An updated meta-analysis. Sci Rep 2019; 9:13177. [PMID: 31511539 PMCID: PMC6739359 DOI: 10.1038/s41598-019-49123-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 08/20/2019] [Indexed: 11/09/2022] Open
Abstract
The aim of this study was to investigate the association among the PlA1/A2 gene polymorphism, laboratory aspirin resistance and adverse clinical outcomes in coronary artery disease (CAD) patients who were on aspirin maintainance therapy. A comprehensive literature search was performed and 35 eligible clinical trials including 19025 CAD patients were recruited. Adverse clinical outcomes involving all-cause death, non-fatal myocardial infarction (MI), ischemic stroke and target vessel revascularization (TVR) were analyzed. The definition of aspirin resistance in each study was accepted. Meta-analysis was performed using the Review Manager 5.3.5 System. In CAD patients, the PlA2 gene carriers had similar incidence of laboratory aspirin resistance compared to those with PlA1/A1 genotype [29.7% vs 28.3%, OR = 0.94 (95% CI 0.63 to 1.40, P = 0.74)], and there were no significant differences in the adverse clinical outcomes between the PlA2 carriers and the PlA1/A1 genotype patients. However, the laboratory aspirin non-responders had higher risks of death [7.9% vs. 2.5%, OR = 2.42 (95% CI 1.86 to 3.15, P < 0.00001)] and TVR [4.5% vs. 1.7%, OR = 2.20 (95% CI 1.19 to 4.08, P = 0.01)] compared to the responders. In aspirin-treated CAD patients, the laboratory aspirin resistance predicts all-cause death and TVR. However, the PlA1/A2 gene polymorphism predicts neither the laboratory aspirin response nor the clinical outcomes.
Collapse
|
9
|
|
10
|
Coller BS. Foreword: A Brief History of Ideas About Platelets in Health and Disease. Platelets 2019. [DOI: 10.1016/b978-0-12-813456-6.09988-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
11
|
Leu33Pro (PlA) polymorphism of integrin beta3 modulates platelet Src pY418 and focal adhesion kinase pY397 phosphorylation in response to abnormally high shear stress. Blood Coagul Fibrinolysis 2018; 29:488-495. [PMID: 29965811 DOI: 10.1097/mbc.0000000000000744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVES Shear stress can activate platelet integrin-mediated signaling that leads to shear-induced platelet aggregation (SIPA) and eventually contribute to acute myocardial infarction. The major platelet integrin αIIbβ3 is polymorphic at residue 33 [Leu33Pro (PlA) polymorphism]. The Pro33 isoform has been shown to have a prothrombotic phenotype. In this work, we studied the impact of Leu33/Pro33 polymorphism on the shear-induced integrin-mediated Src and FAK activation in platelets. METHODS Platelets of both genotypes were placed on immobilized fibrinogen or heat activated BSA and were exposed to physiological (500/s) or abnormally high (5000/s) shear rates for 2-10 min. Platelets after exposure to shear were analysed for Src pY418 and FAK pY397 activities. RESULTS Whereas physiological shear stress does not affect platelet signaling, abnormally high-shear stress considerably elevates Src and FAK phosphorylation in both Pro33 and Leu33 platelets. Both under static and flow conditions, Pro33 platelets exhibited a significantly higher Src and FAK activities than Leu33 platelets. Interestingly, even in the absence of the αIIbβ3-fibrinogen interaction, we could detect a shear-induced integrin-mediated signaling of Src and FAK in platelets. In parallel experiments in which platelets were pretreated with abciximab, an integrin αIIbβ3 antagonist, activation of both kinases by shear was inhibited. CONCLUSION Taken together, our data indicates an important role of αIIbβ3 and shows that Leu33Pro polymorphism modulates the integrin-mediated Src and FAK signaling in platelets in response to shear stress.
Collapse
|
12
|
Affiliation(s)
- Andrew L. Frelinger
- Center for Platelet Research Studies, Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
13
|
A novel ultrasonic method for evaluation of blood clotting parameters. J Med Ultrason (2001) 2018. [PMID: 29536280 DOI: 10.1007/s10396-018-0874-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
PURPOSE For long time, blood clot retraction was measured only by thromboelastographic or platelet contractile force measurement techniques. The purpose of the present study was development of a novel ultrasonic method based on simultaneous monitoring of variations in the ultrasound velocity and the frequency spectrum of the signal propagating in clotting blood and its application for automatic evaluation of blood clotting parameters. METHODS Simultaneous measurement of ultrasound velocity and variations in the frequency spectrum of wideband ultrasonic signals in clotting blood samples was performed. All measurements were performed in pulse-echo mode. Standard clinical data were obtained using routine clinical laboratory methods. RESULTS The amplitudes of ultrasonic signals during native blood coagulation varied up to ten times for different frequencies. The measurement results of the start and duration of blood clot retraction differed between patient samples: different components of the blood coagulation system had significant impact on the blood clot retraction process. CONCLUSIONS Our results showed that during blood clotting, the ultrasound velocity and variations in frequency spectrum should be used simultaneously to determine the beginning and duration of blood clot retraction. Our results also showed that blood clot retraction is controlled by the activity of factor XIII.
Collapse
|
14
|
Papp E, Havasi V, Bene J, Komlosi K, Czopf L, Magyar E, Feher C, Feher G, Horvath B, Marton Z, Alexy T, Habon T, Szabo L, Toth K, Melegh B. Glycoprotein IIIA Gene (PIA) Polymorphism and Aspirin Resistance: Is There Any Correlation? Ann Pharmacother 2017; 39:1013-8. [PMID: 15840736 DOI: 10.1345/aph.1e227] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND:Platelet glycoprotein (GP) IIb/IIIa receptors play an inevitable role in platelet aggregation. The GP IIIa gene is polymorphic ( PIA1/PIA2) and the presence of a PIA2 allele might be associated with an increased risk for acute coronary syndrome (ACS).OBJECTIVE:To examine the prevalence of the PIA2 allele in patients with ACS and in subjects with or without aspirin resistance.METHODS:The prevalence of the PIA2 allele was assessed in 158 patients with ACS and PIA2 compared with its prevalence in 199 healthy volunteers. The antiplatelet efficacy of aspirin was examined in all patients with ACS, as well as in 69 individuals who had suffered ischemic stroke and in 58 high-risk subjects without any known ischemic vascular events.RESULTS:PIA2 prevalence was significantly higher in patients with ACS (59/158) than in the control group (51/199; p < 0.05). Carriers of the PIA2 allele had a significantly higher risk of developing ACS, even after an adjustment to the risk factors (OR 5.74; 95% CI 1.75 to 18.8; p = 0.004). The occurrence of the PIA2 allele was significantly higher among patients with aspirin resistance than in subjects who demonstrated an appropriate response to the drug (allele frequencies, 0.21 vs 0.14; p < 0.05). All patients homozygous for the PIA2 allele had an inadequate platelet response to aspirin.CONCLUSIONS:Our results support the hypothesis that carriers of the PIA2 allele might have an increased risk for ACS. PIA2 homozygosity was associated with an inadequate response to aspirin therapy. Our data further suggest that patients with PIA2 allele homozygosity might benefit from antiplatelet therapy based on adenosine diphosphate antagonists throughout secondary treatment for prevention of ACS.
Collapse
Affiliation(s)
- Elod Papp
- First Department of Medicine, School of Medicine, University of Pecs, Pecs, Hungary.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Abstract
Taken together, there is ample evidence of the association of cardiovascular disease, cerebrovascular, and inflammatory disease with single nucleotide variants (SNV) due to their impact on platelet size, number, and function. With the use of electronic medical record (EMR) or other phenotypic-linked bioinformatics sources, the more important "functional" variants are emerging and provide valuable information on their specific role in promoting early onset of disease or poor response to therapeutic measures. This review will focus upon the recognized common polymorphisms or gene variants with small, but functional effects, as it is becoming clear that these contribute to hyper- or hypo-responsive platelet phenotypes. The impact of these gene variants is distinguishable among normal individuals, and they are suspected contributors to increased risk of adverse outcomes in patients with underlying disease. There are thousands of gene variants and environmental factors that may mitigate risk or amplify the potential for disease within each of us. When combined with the environment and epigenetic influences, it is clear that whole-genome sequencing and bioinformatics alone will not be enough to truly predict "risk" or probability, but awareness of their potential influence may be a starting point in selective screening and generating prevention strategies to promote a healthy lifestyle or fine-tune therapeutic choices in the future.
Collapse
Affiliation(s)
- Diane Nugent
- a Hematology Advanced Diagnostic Laboratory , CHOC Children's Hospital , Orange , CA , USA.,b Center for Inherited Blood Disorders , Orange , CA.,c UC Irvine Medical School , Irvine , CA , USA
| | - Thomas Kunicki
- a Hematology Advanced Diagnostic Laboratory , CHOC Children's Hospital , Orange , CA , USA.,b Center for Inherited Blood Disorders , Orange , CA.,c UC Irvine Medical School , Irvine , CA , USA
| |
Collapse
|
16
|
Huynh KC, Nguyen TH, Pham DC, Nguyen HTT, Van Vo T, Gyenes M, Stoldt VR. Integrin αIIbβ3-Dependent ERK Signaling Is Regulated by Src and Rho Kinases in Both Leu33 and Pro33 Polymorphic Isoforms. Acta Haematol 2016; 137:44-50. [PMID: 27923225 DOI: 10.1159/000450783] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 09/08/2016] [Indexed: 12/29/2022]
Abstract
Platelet integrin αIIbβ3 possesses a Leu/Pro polymorphism at residue 33 (Leu33/HPA-1a or Pro33/HPA-1b). The Pro33 isoform has been suggested to exhibit prothrombotic features. αIIbβ3-expressing CHO (Chinese hamster ovary) cells on immobilized fibrinogen show activation of the MAP kinase family member ERK2, with an enhanced ERK2 activity in Pro33 cells compared to Leu33 cells. In our present work, we examined how the Leu/Pro polymorphism modulates the ERK2 activation stimulated by 2 differently triggered outside-in signalings. We either treated the CHO cells with Mn2+ or allowed them to adhere to fibrinogen. Moreover, we studied which signaling cascades are involved in ERK2 activation. In contrast to immobilized fibrinogen, Mn2+ did not significantly increase ERK2 activation. However, Mn2+ had a synergistic effect on ERK2 phosphorylation when combined with immobilized fibrinogen. Pro33 cells adherent to fibrinogen exhibited a significantly greater ERK2 activity than Leu33 cells in the presence of Mn2+, which peaked after 10 min of adhesion. Our data showed that Src family and rho kinases play a crucial role in the integrin αIIbβ3-dependent outside-in signaling to ERK2.
Collapse
Affiliation(s)
- Khon C Huynh
- Biomedical Engineering Department, International University, Vietnam National University, Ho Chi Minh City, Vietnam
| | | | | | | | | | | | | |
Collapse
|
17
|
Vijayan KV, Bray PF. Molecular Mechanisms of Prothrombotic Risk Due to Genetic Variations in Platelet Genes: Enhanced Outside-In Signaling Through the Pro33 Variant of Integrin β3. Exp Biol Med (Maywood) 2016; 231:505-13. [PMID: 16636298 DOI: 10.1177/153537020623100504] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
In recent years inherited variations in platelet proteins have emerged as potential risk factors that could predispose individuals to arterial thrombosis. Although many studies have examined the association of platelet gene polymorphisms with particular disease states, the underlying mechanisms by which most of these polymorphisms contribute to the pathophysiology of thrombosis have remained largely unexplored. This review will focus on the cellular and molecular features by which these genetic changes affect platelet physiology. Although many genes have been investigated in this regard, only the genes encoding integrins β3 and α2, and the platelet Fc receptor, FcγRIIA, have been studied in any depth. In some cases (such as integrin α2), evidence supports a quantitative trait locus. For other genes, nonsynonymous nucleotide substitutions lead to structural and functional consequences. A large portion of this review will focus on the widely studied Leu33Pro (PIA) polymorphism of integrin β3, and will consider the potential mechanisms by which the Pro33 polymorphism could induce a prothrombotic risk. A detailed understanding of how polymorphisms modulate platelet physiology will be important for understanding individual differences in response to antiplatelet therapy.
Collapse
Affiliation(s)
- K Vinod Vijayan
- Department of Medicine, Baylor College of Medicine, One Baylor Plaza, BCM 286, N1319, Houston, TX 77030, USA
| | | |
Collapse
|
18
|
Liu H, Wang Y, Zheng J, Li G, Chen T, Lei J, Mao Y, Wang J, Liu W, Zhao G, Tacey M, Yan B. Platelet glycoprotein gene Ia C807T, HPA-3, and Ibα VNTR polymorphisms are associated with increased ischemic stroke risk: Evidence from a comprehensive meta-analysis. Int J Stroke 2016; 12:46-70. [PMID: 28004990 DOI: 10.1177/1747493016672085] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Background/aims Platelet glycoproteins play a crucial role in the initial stage of thrombus formation and may contribute to the pathophysiology of atherosclerosis. Polymorphisms in glycoprotein genes alter the function of the protein, possibly leading to increased risk of ischemic stroke. However, previous genetic association studies that examined the relationship between glycoprotein genes polymorphisms and ischemic stroke have yielded inconsistent results. This study aimed to evaluate the association between glycoprotein genes and ischemic stroke by the application of meta-analysis. Methods Relevant studies were identified by an extensive search through databases. The quality of included studies was assessed independently using the Newcastle–Ottawa Scale. Allele and genotype frequencies for each included study were extracted. The odds ratio (OR) with 95% confidence interval (95%CI) was calculated using a random-effects or fixed-effects model. Q statistic was used to evaluate homogeneity, and a meta-regression model was used to explore the study-level variables and to describe the heterogeneity in included studies. Egger’s test and funnel plot were used to assess publication bias. Results A total of 60 studies (9 polymorphisms) were included and identified in the current meta-analysis. The Newcastle–Ottawa Scale scores ranged from 7 to 9 except for two studies with Newcastle–Ottawa Scale scores of 6. The T allele or TT genotype of the glycoprotein Ia C807T polymorphism were associated with an increased susceptibility to ischemic stroke in combined population (807T allele: OR, 95%CI: 1.24, 1.03–1.50, p = 0.02) or Asian populations (807T allele: OR, 95%CI: 1.31, 1.10–1.54, p = 0.002 and 807TT genotype: OR, 95%CI: 1.53, 1.13–2.08, p = 0.006, respectively), and the Ser allele of HPA-3 was associated with increased risk of ischemic stroke in combined population or in Asians (OR, 95%CI: 1.21, 1.04–1.40, p = 0.01 or 1.54, 1.18–2.01, p = 0.001). Of note, the Ser/Ser genotype was more common in Asians (OR, 95%CI: 2.09, 1.40–3.13, p < 0.001). For glycoprotein Ibα variable number tandem repeat, only B allele showed a mild significant association with ischemic stroke risk in combined population or in Caucasians (OR, 95%CI: 2.17, 1.04–4.55, p = 0.04 or 1.79, 1.02–3.13, p = 0.04). There was no significant association between HPA-1, HPA-2, HPA-4, HPA-5, glycoprotein Ibα-5 T/C as well as Ia G873A polymorphisms and increased risk of ischemic stroke. Conclusions We found that glycoprotein Ia C807T T allele or the TT genotype, the Ser-allele of HPA-3 and B allele of glycoprotein Ibα variable number tandem repeat polymorphisms were associated with increased risk for ischemic stroke. Future studies with larger sample sizes will be necessary to confirm the results. In addition, analyses of ischemic stroke subtypes and gene–gene and gene–environment interactions are warranted.
Collapse
Affiliation(s)
- Hua Liu
- Department of Neurology, the Second Clinical Medical College of North Sichuan Medical College & Nanchong Central Hospital, Nanchong, P R China
- Department of Neurology, The First Affiliated Hospital, Kunming Medical University, Kunming, PR China
- Melbourne Brain Centre, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, Australia
| | - Yi Wang
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, P R China
| | - Jian Zheng
- Department of Neurology, Xinqiao Hospital, the Third Military Medical University, Chongqing, PR China
| | - Guangming Li
- Department of Neurology, the Second Clinical Medical College of North Sichuan Medical College & Nanchong Central Hospital, Nanchong, P R China
| | - Tao Chen
- Department of Neurology, The First Affiliated Hospital, Kunming Medical University, Kunming, PR China
| | - Jianguo Lei
- Department of Neurology, the Fifth people's Hospital of Chengdu, North Sichuan Medical College, Chengdu, P R China
| | - Yiting Mao
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, P R China
| | - Jun Wang
- Department of Neurology, the Fifth people's Hospital of Chengdu, North Sichuan Medical College, Chengdu, P R China
| | - Wei Liu
- Department of Neurology, the Second Clinical Medical College of North Sichuan Medical College & Nanchong Central Hospital, Nanchong, P R China
| | - Ge Zhao
- Department of Neurology, the Fifth people's Hospital of Chengdu, North Sichuan Medical College, Chengdu, P R China
| | - Mark Tacey
- Melbourne EpiCentre, Royal Melbourne Hospital and Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia
| | - Bernard Yan
- Melbourne Brain Centre, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
19
|
Doly JS, Lorian E, Desormais I, Constans J, Bura Rivière A, Lacroix P. [Prevalence and prognosis of aspirin resistance in critical limb ischemia patients]. ACTA ACUST UNITED AC 2016; 41:358-364. [PMID: 27745943 DOI: 10.1016/j.jmv.2016.08.004] [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: 10/06/2015] [Accepted: 07/29/2016] [Indexed: 11/16/2022]
Abstract
OBJECTIVES To assess the prevalence and the association between aspirin resistance in critical limb ischemia patients using the VerifyNow® bed-side platelet test, and occurrence of cardiovascular morbidity and/or death at one year. MATERIALS AND METHODS National multicenter prospective observational study related to COPART II centers. From 2010 through 2014, 64 subjects hospitalized for critical limb ischemia and already treated by aspirin before the VerifyNow® test were included. A VerifyNow® test>550 ARU was defined as aspirin resistance. Critical limb ischemia was defined according to the TASC I criteria. The primary outcome was a composite including death, acute coronary syndrome, stroke and major amputation during the one-year follow-up period. RESULTS In all, 9/64 patients were aspirin resistant, the status was confirmed in one case. The prevalence of aspirin resistance was 14.06%. There was no significant difference between aspirin resistant and aspirin non-resistant groups in terms of cardiovascular history and glycemia status. Neither was there significant difference between the two groups in terms of survival. CONCLUSION Aspirin resistance was not predictive of poorer survival in critical limb ischemia patients. However, our population was limited. Considering that a clear definition of aspirin resistance and standardized diagnostic tests are lacking, complementary studies might be useful.
Collapse
Affiliation(s)
- J-S Doly
- Service chirurgie thoracique et cardiovasculaire et angiologie, unité de médecine vasculaire, CHU de Limoges, 2, avenue Martin-Luther-King, 87000 Limoges, France.
| | - E Lorian
- Service chirurgie thoracique et cardiovasculaire et angiologie, unité de médecine vasculaire, CHU de Limoges, 2, avenue Martin-Luther-King, 87000 Limoges, France
| | - I Desormais
- Service chirurgie thoracique et cardiovasculaire et angiologie, unité de médecine vasculaire, CHU de Limoges, 2, avenue Martin-Luther-King, 87000 Limoges, France
| | - J Constans
- Service chirurgie thoracique et cardiovasculaire et angiologie, unité de médecine vasculaire, CHU de Limoges, 2, avenue Martin-Luther-King, 87000 Limoges, France
| | - A Bura Rivière
- Service chirurgie thoracique et cardiovasculaire et angiologie, unité de médecine vasculaire, CHU de Limoges, 2, avenue Martin-Luther-King, 87000 Limoges, France
| | - P Lacroix
- Service chirurgie thoracique et cardiovasculaire et angiologie, unité de médecine vasculaire, CHU de Limoges, 2, avenue Martin-Luther-King, 87000 Limoges, France
| |
Collapse
|
20
|
Rather RA, Dhawan V. Genetic markers: Potential candidates for cardiovascular disease. Int J Cardiol 2016; 220:914-923. [PMID: 27416153 DOI: 10.1016/j.ijcard.2016.06.251] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Revised: 06/22/2016] [Accepted: 06/26/2016] [Indexed: 02/07/2023]
Abstract
The effective prevention of cardiovascular disease depends upon the ability to recognize the high-risk individuals at an early stage of the disease or long before the development of adverse events. Evolving technologies in the fields of proteomics, metabolomics, and genomics have played a significant role in the discovery of cardiovascular biomarkers, but so far these methods have achieved the modest success. Hence, there is a crucial need for more reliable, suitable, and lasting diagnostic and therapeutic markers to screen the disease well in time to start the clinical aid to the patients. Gene polymorphisms associated with the cardiovascular disease play a decisive role in the disease onset. Therefore, the genetic marker evaluation to classify high-risk patients from low-risk patients trends an effective approach to patient management and care. Currently, there are no genetic markers available for extensive adoption as risk factors for coronary vascular disease, yet, there are numerous promising, biologically acceptable candidates. Many of these gene biomarkers, alone or in combination, can play an essential role in the prediction of cardiovascular risk. The present review highlights some putative emerging genetic biomarkers that could facilitate more authentic and fast diagnosis of CVD. This review also briefly describes few technological approaches employed in the biomarker search.
Collapse
Affiliation(s)
- Riyaz Ahmad Rather
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Veena Dhawan
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh, India.
| |
Collapse
|
21
|
Kubisz P, Ivankov J, Holly P, Stasko JN, Musiał J. The Glycoprotein IIIa PLA1/A2 Polymorphism—A Defect Responsible for the Sticky Platelet Syndrome? Clin Appl Thromb Hemost 2016; 12:117-9. [PMID: 16444447 DOI: 10.1177/107602960601200121] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
22
|
Abstract
Understanding how platelet activation is regulated is important in the context of cardiovascular disorders and their management with antiplatelet therapy. Recent evidence points to different platelet subpopulations performing different functions. In particular, procoagulant and aggregating subpopulations have been reported in the literature in platelets treated with the GPVI agonists. How the formation of platelet subpopulations upon activation is regulated remains unclear. Here, it is shown that procoagulant and aggregating platelet subpopulations arise spontaneously upon adhesion of purified platelets on clean glass surfaces. Calcium ionophore treatment of the adhering platelets resulted in one platelet population expressing both the procoagulant and the adherent population markers phosphatidylserine and the activated form of GPIIb/IIIa, while all of the platelets expressed CD62P independently of the ionophore treatment. Therefore, all platelets have the capacity to express all three activation markers. It is concluded that platelet subpopulations observed in various studies reflect the dynamics of the platelet activation process.
Collapse
|
23
|
Milanowski L, Pordzik J, Janicki PK, Postula M. Common genetic variants in platelet surface receptors and its association with ischemic stroke. Pharmacogenomics 2016; 17:953-71. [PMID: 27269246 DOI: 10.2217/pgs.16.21] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Ischemic stroke has been named one of the leading causes of death worldwide. Whereas numerous biological mechanisms and molecules were found to be associated with stroke, platelets are particularly contributive to its pathogenesis. Recent data indicate considerable variability in platelet phenotype which accounts for differences in platelet surface receptor function, count and reactivity. These features collectively influence both the events leading to a disease and effectiveness of antiplatelet therapies. Consequently, genetic variants predisposing to cerebrovascular diseases can be sequenced using a wide array of techniques and become a useful tool in clinical setting. In this review, we provide an outline of common platelet polymorphisms that impose risk on ischemic stroke development and should be evaluated as targets to improve treatment. As study results are often inconsistent, partly due to differences in demographic characteristics between study populations and the fact that the functional impact of these variants has been relatively small, we conclude that both rare, low-frequency and common variants might account for genetic contribution on abnormal platelet response to antiplatelet drugs.
Collapse
Affiliation(s)
- Lukasz Milanowski
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Center for Preclinical Research and Technology CEPT, Warsaw, Poland
| | - Justyna Pordzik
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Center for Preclinical Research and Technology CEPT, Warsaw, Poland
| | - Piotr K Janicki
- Perioperative Genomics Laboratory, Penn State College of Medicine, Hershey, PA 17033, USA
| | - Marek Postula
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Center for Preclinical Research and Technology CEPT, Warsaw, Poland.,Perioperative Genomics Laboratory, Penn State College of Medicine, Hershey, PA 17033, USA
| |
Collapse
|
24
|
Floyd CN, Ferro A, Warner TD. Expression of the PlA2 allele of glycoprotein IIIa and its impact on platelet function. JRSM Cardiovasc Dis 2016; 4:2048004015610252. [PMID: 26858830 PMCID: PMC4734162 DOI: 10.1177/2048004015610252] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 08/04/2015] [Accepted: 08/06/2015] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND The platelet fibrinogen receptor represents the final common pathway of platelet activation, and is formed from two glycoprotein (GP) subunits (GPIIb/IIIa). Carriage of the mutant PlA2 allele of GPIIIa has been shown to confer an increased risk of cardiovascular events, but published studies have disagreed as to the mechanism for this association. OBJECTIVES To assess whether carriage of the PlA2 allele conforms to Mendelian patterns of expression and to identify whether carriage of the mutant allele modulates platelet function. METHODS Expression of the PlA2 allele was assessed in both healthy subjects (n = 25) and patients with known coronary artery disease (n = 90) through the development and validation of a liquid chromatography, tandem mass spectrometry (LC-MS/MS) assay. Platelet function was assessed in the patient cohort in response to multiple agonists, and these data were analysed in the context of the proteomic data. RESULTS Expression of the wild-type PlA1 allele and mutant PlA2 alleles was readily quantifiable and conformed to Mendelian patterns in both healthy and patient cohorts. Patients who were homozygous for the mutant PlA2 allele had an increased aggregatory response to adenosine diphosphate, collagen, adrenaline, ristocetin, thrombin receptor-activating peptide 6 and U46619, when assessed using agonist-concentration response curves. CONCLUSIONS These findings support the hypothesis that carriage of the mutant PlA2 allele mediates an increased risk of cardiovascular events through the modulation of platelet reactivity.
Collapse
Affiliation(s)
- Christopher N Floyd
- Department of Clinical Pharmacology, Cardiovascular Division, British Heart Foundation Centre of Research Excellence, King's College London, London, UK
| | - Albert Ferro
- Department of Clinical Pharmacology, Cardiovascular Division, British Heart Foundation Centre of Research Excellence, King's College London, London, UK
| | - Timothy D Warner
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, London, UK
| |
Collapse
|
25
|
Kubisz P, Stanciakova L, Stasko J, Dobrotova M, Skerenova M, Ivankova J, Holly P. Sticky platelet syndrome: an important cause of life-threatening thrombotic complications. Expert Rev Hematol 2015; 9:21-35. [PMID: 26567442 DOI: 10.1586/17474086.2016.1121095] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Sticky platelet syndrome (SPS) is a prothrombotic thrombocytopathy with familial occurrence, characterized by hyperaggregability of platelets in response to adenosine diphosphate (ADP), epinephrine (EPI) or both. The syndrome has been identified in approximately 21% of unexplained arterial thrombotic episodes, regarded to be the most common thrombophilia in arterial thrombosis and 13.2% of unexplained venous thromboembolism (VTE). The relatively young age at the first manifestation, relation to fertility and pregnancy, seriousness of the symptoms, easy and effective management of the disorder indicate to the necessity to take it into account in the differential diagnosis of the underlying cause of the thrombotic event. As the various localizations of the thrombosis in SPS have been reported, its management often requires a multidisciplinary approach. This review deals with the clinical aspects of thrombophilia, its etiopathogenesis, diagnosis as well as novel advances in the treatment and outlines the challenges for the further research.
Collapse
Affiliation(s)
- Peter Kubisz
- a National Center of Hemostasis and Thrombosis, Clinic of Hematology and Transfusiology, Jessenius Faculty of Medicine in Martin , Comenius University in Bratislava, Martin University Hospital , Martin , Slovak Republic
| | - Lucia Stanciakova
- a National Center of Hemostasis and Thrombosis, Clinic of Hematology and Transfusiology, Jessenius Faculty of Medicine in Martin , Comenius University in Bratislava, Martin University Hospital , Martin , Slovak Republic
| | - Jan Stasko
- a National Center of Hemostasis and Thrombosis, Clinic of Hematology and Transfusiology, Jessenius Faculty of Medicine in Martin , Comenius University in Bratislava, Martin University Hospital , Martin , Slovak Republic
| | - Miroslava Dobrotova
- a National Center of Hemostasis and Thrombosis, Clinic of Hematology and Transfusiology, Jessenius Faculty of Medicine in Martin , Comenius University in Bratislava, Martin University Hospital , Martin , Slovak Republic
| | - Maria Skerenova
- b Department of Clinical Biochemistry, Jessenius Faculty of Medicine in Martin , Comenius University in Bratislava, Martin University Hospital , Martin , Slovak Republic
| | - Jela Ivankova
- a National Center of Hemostasis and Thrombosis, Clinic of Hematology and Transfusiology, Jessenius Faculty of Medicine in Martin , Comenius University in Bratislava, Martin University Hospital , Martin , Slovak Republic
| | - Pavol Holly
- a National Center of Hemostasis and Thrombosis, Clinic of Hematology and Transfusiology, Jessenius Faculty of Medicine in Martin , Comenius University in Bratislava, Martin University Hospital , Martin , Slovak Republic
| |
Collapse
|
26
|
Multiple thrombophilic single nucleotide polymorphisms lack a significant effect on outcomes in fresh IVF cycles: an analysis of 1717 patients. J Assist Reprod Genet 2015; 33:67-73. [PMID: 26545911 DOI: 10.1007/s10815-015-0606-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 10/26/2015] [Indexed: 10/22/2022] Open
Abstract
PURPOSE The aim of the study is to determine if thrombophilic single nucleotide polymorphisms (SNPs) affect outcomes in fresh in vitro fertilization (IVF) cycles in a large general infertility population. METHODS A prospective cohort analysis was performed at a university-affiliated private IVF center of female patients undergoing fresh non-donor IVF cycles. The effect of the following thrombophilic SNPs on IVF outcomes were explored: factor V (Leiden and H1299R), prothrombin (G20210A), factor XIII (V34L), β-fibrinogen (-455G → A), plasminogen activator inhibitor-1 (4G/5G), human platelet antigen-1 (a/b9L33P), and methylenetetrahydrofolate reductase (C677T and A1298C). The main outcome measures included positive pregnancy test, clinical pregnancy, embryo implantation, live birth, and pregnancy loss. RESULTS Patients (1717) were enrolled in the study, and a total of 4169 embryos were transferred. There were no statistically significant differences in positive pregnancy test, clinical pregnancy, embryo implantation, live birth, or pregnancy loss in the analysis of 1717 patients attempting their first cycle of IVF. Receiver operator characteristics and logistic regression analyses showed that outcomes cannot be predicted by the cumulative number of thrombophilic mutations present in the patient. CONCLUSIONS Individual and cumulative thrombophilic SNPs do not affect IVF outcomes. Therefore, initial screening for these SNPs is not indicated.
Collapse
|
27
|
Effect of acetylsalicylic acid on platelet activation and oxidative profile in a set of Brazilian patients with type 2 diabetes mellitus. Blood Coagul Fibrinolysis 2015; 26:123-30. [PMID: 25325344 DOI: 10.1097/mbc.0000000000000199] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Type 2 diabetes mellitus (DM2) is a metabolic disorder associated with hyperactivation of platelets, increased formation of platelet microparticles (PMPs) and oxidative stress that are related to cardiovascular complications. Acetylsalicylic acid (ASA) is an antiplatelet agent used in the prevention of atherothrombosis. The aim of this study was to evaluate the effect of ASA by means of platelet activation and oxidative profile. We collected blood samples of 81 patients with DM2 before and during ASA treatment. These samples were analyzed to determine the levels of 2,3-dinor thromboxane-B2 (2,3-dinor-TXB2), PMPs, thiobarbituric acid reactive species (TBARS) and 3-(4,5-dimethylthiazol-2yl)-2,5-diphenyltetrazolium bromide (MTT). Moreover, the relationship between the levels of 2,3-dinor-TXB2 with some clinical and laboratory variables such as glycated hemoglobin, platelet count, D dimer, low-density lipoprotein cholesterol and glycoprotein IIb/IIIa and cyclooxygenase-1 polymorphisms was evaluated. ASA intake did not change the levels of PMP, TBARS and MTT. Although a significant decrease in the levels of 2,3 dinorTXB2 (P < 0.001) in patients under ASA has been observed, an equal and satisfactory response to this drug was not found. However, the presence of PIA2 allele in GPIIIa gene may be associated with a better response to ASA intake in these patients, whereas other clinical and laboratory variables showed no association with this drug use. These findings are consistent with previous reports in the literature that patients with DM2 do not benefit in an equal way from the use of ASA for primary prevention of atherothrombotic events.
Collapse
|
28
|
Gabbasov Z, Sabo J, Petrovic D, Martell-Claros N, Zagatina A, Mrdovic I, Ciccocioppo R, Cangemi GC, Klimas J, Kruzliak P. Impact of platelet phenotype on myocardial infarction. Biomarkers 2014; 20:17-25. [PMID: 25510672 DOI: 10.3109/1354750x.2014.993707] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
In acute myocardial infarction patients the injured vascular wall triggers thrombus formation in the damage site. Fibrin fibers and blood cellular elements are the major components of thrombus formed in acute occlusion of coronary arteries. It has been established that the initial thrombus is primarily composed of activated platelets rapidly stabilized by fibrin fibers. This review highlights the role of platelet membrane phenotype in pathophysiology of myocardial infarction. Here, we regard platelet phenotype as quantitative and qualitative parameters of the plasma membrane outer surface, which are crucial for platelet participation in blood coagulation, development of local inflammation and tissue repair.
Collapse
Affiliation(s)
- Zufar Gabbasov
- Institute of Experimental Cardiology, Cardiology Research Center , Moscow , Russia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Floyd CN, Ferro A. The PlA1/A2 polymorphism of glycoprotein IIIa in relation to efficacy of antiplatelet drugs: a systematic review and meta-analysis. Br J Clin Pharmacol 2014; 77:446-57. [PMID: 23834376 DOI: 10.1111/bcp.12204] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Accepted: 06/24/2013] [Indexed: 11/29/2022] Open
Abstract
AIM The PlA1/A2 polymorphism of glycoprotein IIIa (GPIIIa) has been associated with both antiplatelet drug resistance and increased cardiovascular events. The aim of this study was to conduct the first meta-analysis investigating the association between carriage of the PlA2 allele and resistance to currently licensed antiplatelet drugs. METHODS Electronic databases (MEDLINE and EMBASE) were searched for all articles evaluating genetic polymorphisms of GPIIIa. For studies where antiplatelet resistance was measured using validated techniques, pooled odds ratios (ORs) were calculated using fixed effects and random effects models. RESULTS Sixteen studies were eligible for statistical analysis and included 1650 PlA1 homozygous subjects and 668 carriers of the PlA2 allele. For carriers of the PlA2 allele, OR 0.924 (n = 2318; 95% CI 0.743, 1.151; P = 0.481) was observed for resistance to any antiplatelet drug, OR 0.862 (n = 2085; 95% CI 0.685, 1.086; P = 0.208) for resistance to aspirin and OR 1.429 (n = 233; 95% CI 0.791, 2.582; P = 0.237) for resistance to clopidogrel. In the aspirin cohort, sub-group analysis revealed no statistical association in either healthy subjects or those with cardiovascular disease. PlA2 carriage was marginally associated with aspirin sensitivity using the fixed effects model when identified by the PFA-100 assay (n = 1151; OR 0.743, 95% CI 0.558, 0.989; P = 0.041) but with significant heterogeneity (I(2) = 55%; P = 0.002). Significance was lost with analysis using a random effects model. CONCLUSIONS The totality of published data does not support an association between carriage of the PlA2 allele and antiplatelet drug resistance. Significant heterogeneity indicates the need for larger studies using validated and standardized assays.
Collapse
Affiliation(s)
- Christopher N Floyd
- Department of Clinical Pharmacology, Cardiovascular Division, British Heart Foundation Centre of Research Excellence, King's College London, London, UK
| | | |
Collapse
|
30
|
Gong JY, Deng DT, Sun YH. Association of platelet glycoprotein receptor alpha2beta1 integrin and glycoprotein IIIa gene polymorphisms with diabetic retinopathy: evidence from 3007 subjects. Curr Eye Res 2014; 40:476-83. [PMID: 24979111 DOI: 10.3109/02713683.2014.932386] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
PURPOSE Many epidemiological studies have evaluated associations of platelet glycoprotein receptor alpha2beta1 integrin (ITGA2) and glycoprotein IIIa (ITGB3) gene polymorphisms with diabetic retinopathy (DR), but the published data are inconclusive. The aim of the present study was to assess the associations by using meta-analysis. METHODS A comprehensive electronic search (PubMed, EMBASE, Elsevier Science Direct, CNKI and Wanfang) was carried out until 31 August 2013. Odds ratios (ORs) and its 95% confidence intervals (CIs) were used to assess the strength of the associations. RESULTS Nine studies including 1678 cases and 1329 controls were included in the meta-analysis. Meta-analysis was performed for ITGA2 gene BgI II polymorphism (7 studies including 758 cases and 570 controls) and ITGB3 gene PlA1/A2 polymorphism (4 studies including 1047 cases and 861 controls). Significant associations were found for BgI II (+ versus -: OR = 1.42, 95% CI = 1.06-1.90, p = 0.02; +/- + +/+ versus -/-: OR = 1.46, 95% CI = 0.99-2.15, p = 0.06; +/+ versus -/- + +/-: OR = 1.90, 95% CI = 1.35-2.67, p = 0.0003) and PlA1/A2 (A2 versus A1: OR = 0.74, 95% CI = 0.52-1.07, p = 0.11; A1A2 + A2A2 versus A1A1: OR = 0.80, 95% CI = 0.64-0.99, p = 0.04; A2A2 versus A1A1 + A1A2: OR = 0.45, 95% CI = 0.27-0.75, p = 0.002) polymorphisms. CONCLUSION This meta-analysis demonstrates that DR is associated with ITGA2 BgI II and ITGB3 PlA1/A2 polymorphisms.
Collapse
Affiliation(s)
- Jian-Yang Gong
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University , Hefei, Anhui , China
| | | | | |
Collapse
|
31
|
Khaspekova SG, Zyuryaev IT, Yakushkin VV, Naimushin YA, Sirotkina OV, Zaytseva NO, Ruda MY, Mazurov AV. Mean platelet volume: Interrelation with platelet aggregation activity and glycoprotein IIb-IIIa and Ib expression levels. BIOCHEMISTRY MOSCOW-SUPPLEMENT SERIES B-BIOMEDICAL CHEMISTRY 2014. [DOI: 10.1134/s199075081402005x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
32
|
Oliver KH, Jessen T, Crawford EL, Chung CY, Sutcliffe JS, Carneiro AM. Pro32Pro33 mutations in the integrin β3 PSI domain result in αIIbβ3 priming and enhanced adhesion: reversal of the hypercoagulability phenotype by the Src inhibitor SKI-606. Mol Pharmacol 2014; 85:921-31. [PMID: 24695082 PMCID: PMC4014669 DOI: 10.1124/mol.114.091736] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 04/02/2014] [Indexed: 11/22/2022] Open
Abstract
The plasma-membrane integrin αIIbβ3 (CD41/CD61, GPIIbIIIa) is a major functional receptor in platelets during clotting. A common isoform of integrin β3, Leu33Pro is associated with enhanced platelet function and increased risk for coronary thrombosis and stroke, although these findings remain controversial. To better understand the molecular mechanisms by which this sequence variation modifies platelet function, we produced transgenic knockin mice expressing a Pro32Pro33 integrin β3. Consistent with reports utilizing human platelets, we found significantly reduced bleeding and clotting times, as well as increased in vivo thrombosis, in Pro32Pro33 homozygous mice. These alterations paralleled increases in platelet attachment and spreading onto fibrinogen resulting from enhanced integrin αIIbβ3 function. Activation with protease-activated receptor 4- activating peptide, the main thrombin signaling receptor in mice, showed no significant difference in activation of Pro32Pro33 mice as compared with controls, suggesting that inside-out signaling remains intact. However, under unstimulated conditions, the Pro32Pro33 mutation led to elevated Src phosphorylation, facilitated by increased talin interactions with the β3 cytoplasmic domain, indicating that the αIIbβ3 intracellular domains are primed for activation while the ligand-binding domain remains unchanged. Acute dosing of animals with a Src inhibitor was sufficient to rescue the clotting phenotype in knockin mice to wild-type levels. Together, our data establish that the Pro32Pro33 structural alteration modifies the function of integrin αIIbβ3, priming the integrin for outside-in signaling, ultimately leading to hypercoagulability. Furthermore, our data may support a novel approach to antiplatelet therapy by Src inhibition where hemostasis is maintained while reducing risk for cardiovascular disease.
Collapse
Affiliation(s)
- Kendra H Oliver
- Departments of Pharmacology (K.H.O., T.J., C.Y.C., A.M.C.) and Psychiatry, Molecular Physiology, and Biophysics (E.L.C., J.S.S.), Vanderbilt University Medical Center, Nashville, Tennessee
| | | | | | | | | | | |
Collapse
|
33
|
Spiliopoulos S, Kassimis G, Hatzidakis A, Krokidis M. High on-treatment platelet reactivity in peripheral endovascular procedures. Cardiovasc Intervent Radiol 2014; 37:559-571. [PMID: 23897511 DOI: 10.1007/s00270-013-0707-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Accepted: 07/10/2013] [Indexed: 02/05/2023]
Abstract
The use of aspirin is considered the "gold standard" for the decrease of major adverse cardiovascular events in patients with atherosclerosis, including peripheral arterial disease (PAD), whereas a dual-antiplatelet regimen with aspirin and clopidogrel is usually indicated for such patients after angioplasty and stent deployment. However, a substantial number of subsequent adverse events still occur, even in patients who receive double-antiplatelet therapy. The "high on-treatment platelet reactivity" (HTPR) phenomenon has been lately recognized and plays a major role in the management of patients with PAD. Greater and more rapid inhibition of platelet aggregation has become the goal for new antiplatelet agents with the expectation of further improving outcomes for percutaneous intervention for PAD. The purpose of this review article is to highlight current evidence regarding the prevalence, aetiology, and clinical implications of HTPR in PAD as well as to discuss the possibilities of novel alternative antiplatelet regiments.
Collapse
|
34
|
Khaspekova SG, Ziuriaev IT, Iakushkin VV, Naĭmushin IA, Sirotkina OV, Zaĭtseva NO, Ruda MI, Mazurov AV. [Mean platelet volume: interactions with platelet aggregation activity and glycoprotein IIb-IIIa and Ib expression levels]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2014; 60:94-108. [PMID: 24749250 DOI: 10.18097/pbmc20146001094] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Increased mean platelet volume (MPV) is an independent risk factor of thrombotic events in patients with cardiovascular diseases. Interactions of MPV with platelet aggregation activity and contents of glycoprotein (GP) IIb-IIIa (alphaIIb/beta3 integrin, fibrinogen receptor) and GP Ib (von Willebrand factor receptor) were investigated in this study. Investigation was performed in a group of healthy volunteers (n = 38) and in a group of patients with acute coronary syndrome (ACS). In patients blood was collected at days 1, 3-5 and 8-12 after ACS development. As an antiaggregant therapy all patients received acetylsalicylic acid (ASA, inhibitor of thromboxane A2 synthesis) and most of them--clopidogrel (ADP receptor antagonist) with the exception of part of the patients (n = 44) at day 1 who had not taken clopidogrel before first blood collection. In volunteers platelet aggregation was stimulated by 1.25, 2.5, 5 and 20 M ADP, and in patients--by 5 and 20 M ADP. GP IIb-IIIa and GP Ib content on platelet surface was measured using 125I-labelled monoclonal antibodies. GP IIb-IIIa and GP Ib genetic polymorphisms were determined in ACS patients. In healthy donors significant correlations between MPV and aggregation levels were revealed at 1.25 and 2.5 M ADP (coefficients of correlation (r)--0.396 and 0.373, p < 0.05) and at 5 and 20 those interactions did not reach significant level (r--0.279 and 0.205, p > 0.05). Correlations between MPV and aggregation levels were observed at day 1 of ACS in a subgroup of patients who received ASA but had not started clopidogrel treatment (r--0.526, p < 0.01 and 0.368, p < 0.05 for 5 and 20 M ADP respectively). Interactions between these parameters were not registered upon combined treatment with ASA and clopidogrel. Strong direct correlations between MPV and GP IIb-IIIa and GP Ib contents were detected in healthy donors and ACS patients (at all time points) -r from 0.439 to 0.647 (p < or = 0.001 for all correlations). Genetic polymorphisms of GP IIb-IIIa (GP IIIa Leu33Pro) and GP Ib ((-5)T/C (Kozak) and Thr145Met) identified in ACS patients did not affect expression levels of corresponding glycoproteins. The data obtained indicated that increased MPV values correlate with increased platelet aggregation activity and enhanced GP IIb-IIIa and GP Ib expression.
Collapse
|
35
|
Stratmann B, Xu T, Meisinger C, Menart B, Roden M, Herder C, Grallert H, Peters A, Koenig W, Illig T, Wichmann HE, Wang-Sattler R, Rathmann W, Tschoepe D. PLA1A2 platelet polymorphism predicts mortality in prediabetic subjects of the population based KORA S4-Cohort. Cardiovasc Diabetol 2014; 13:90. [PMID: 24886443 PMCID: PMC4022397 DOI: 10.1186/1475-2840-13-90] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 04/28/2014] [Indexed: 01/15/2023] Open
Abstract
OBJECTIVE The genetic polymorphism concerning the ß3-subunit of platelet integrin receptor glycoprotein IIIa is held responsible for enhanced binding of adhesive proteins resulting in increased thrombogenic potential. Whether it is associated with mortality, HbA1c or platelet volume is tested prospectively in an epidemiological cohort. RESEARCH DESIGN AND METHODS Population-based Cooperative Health Research in the Region of Augsburg (KORA) S4-Survey (N = 4,028) was investigated for prognostic value of PLA1A2-polymorphism regarding all-cause mortality, correlation with HbA1c, and mean platelet volume. Multivariate analysis was performed to investigate association between genotype and key variables. RESULTS Prevalence of thrombogenic allele variant PLA2 was 15.0%. Multivariate analysis revealed no association between PLA1A2 polymorphism and mortality in the KORA-cohort. HbA1c was a prognostic marker of mortality in non-diabetic persons resulting in J-shaped risk curve with dip at HbA1c = 5.5% (37 mmol/mol), confirming previous findings regarding aged KORA-S4 participants (55-75 years). PLA1A2 was significantly associated with elevated HbA1c levels in diabetic patients (N = 209) and reduced mean platelet volume in general population. In non-diabetic participants (N = 3,819), carriers of PLA2 allele variant presenting with HbA1c > 5.5% (37 mmol/mol) showed higher relative risk of mortality with increasing HbA1c. CONCLUSION PLA1A2 polymorphism is associated with mortality in participants with HbA1c ranging from 5.5% (37 mmol/mol) to 6.5% (48 mmol/mol). Maintenance of euglycemic control and antiplatelet therapy are therefore regarded as effective primary prevention in this group.
Collapse
Affiliation(s)
- Bernd Stratmann
- Heart and Diabetes Center NRW, Ruhr University Bochum, Georgstr. 11, D-32545 Bad Oeynhausen, Germany
| | - Tao Xu
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum Muenchen, German Research Center of Environmental Health, Neuherberg, Germany
| | - Christa Meisinger
- Institute of Epidemiology II, Helmholtz Zentrum Muenchen, German Research Center of Environmental Health, Neuherberg, Germany
| | - Barbara Menart
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Duesseldorf, Germany
| | - Michael Roden
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Duesseldorf, Germany
| | - Christian Herder
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Duesseldorf, Germany
| | - Harald Grallert
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum Muenchen, German Research Center of Environmental Health, Neuherberg, Germany
| | - Annette Peters
- Institute of Epidemiology II, Helmholtz Zentrum Muenchen, German Research Center of Environmental Health, Neuherberg, Germany
| | - Wolfgang Koenig
- Department Internal Medicine II, University Clinic Ulm, Ulm, Germany
| | - Thomas Illig
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum Muenchen, German Research Center of Environmental Health, Neuherberg, Germany
- Hannover Unified Biobank, Hannover Medical School, Hannover, Germany
| | - Heinz-Erich Wichmann
- Institute of Epidemiology I, Helmholtz Zentrum Muenchen, German Research Center of Environmental Health, Neuherberg, Germany
- Institute of Medical Informatics, Biometry and Epidemiology, Ludwig-Maximilians-Universität, Munich, Germany
| | - Rui Wang-Sattler
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum Muenchen, German Research Center of Environmental Health, Neuherberg, Germany
| | - Wolfgang Rathmann
- Institute of Biometrics and Epidemiology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Duesseldorf, Germany
| | - Diethelm Tschoepe
- Heart and Diabetes Center NRW, Ruhr University Bochum, Georgstr. 11, D-32545 Bad Oeynhausen, Germany
| |
Collapse
|
36
|
Platelet PIA1/PIA2 polymorphism and the risk of periprocedural myocardial infarction in patients with acute coronary syndromes undergoing coronary angioplasty. Blood Coagul Fibrinolysis 2014; 25:107-13. [DOI: 10.1097/mbc.0b013e3283650717] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
37
|
Relationships of glycoproteins IIb-IIIa and Ib content with mean platelet volume and their genetic polymorphisms. Blood Coagul Fibrinolysis 2014; 25:128-34. [DOI: 10.1097/mbc.0b013e328364b025] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
38
|
Li MP, Xiong Y, Xu A, Zhou JP, Tang J, Zhang ZL, Zhou HH, Zhang W, Chen XP. Association of platelet ITGA2B and ITGB3 polymorphisms with ex vivo antiplatelet effect of ticagrelor in healthy Chinese male subjects. Int J Hematol 2014; 99:263-71. [PMID: 24474638 DOI: 10.1007/s12185-014-1502-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Revised: 01/09/2014] [Accepted: 01/14/2014] [Indexed: 01/15/2023]
Abstract
Ticagrelor (TIC) is the first reversible P2Y12 receptor antagonist that exhibits rapid antiplatelet effect by indirect inhibition of the GPIIb/IIIa complex. Polymorphisms in genes coding GPIIb/IIIa, namely ITGA2B and ITGB3, are associated with aspirin resistance and risk for thrombotic diseases. We assessed whether ITGA2B and ITGB3 polymorphisms can influence the ex vivo antiplatelet activity of ticagrelor in Chinese population. A total of 196 healthy Chinese male individuals were recruited. ADP-induced platelet aggregation was determined using optical aggregometry at baseline and after incubation of the platelet-rich plasma with 15 and 50 μM ticagrelor, respectively. Single nucleotide polymorphisms in ITGA2B (rs5911 G>T) and ITGB3 (rs4642 A>G and rs4634 G>A) were genotyped by sequencing. TIC at both concentrations of 15 and 50 μM decreased ADP-induced platelet aggregation significantly (P < 0.05, respectively). As compared to ITGA2B rs5911 GG homozygotes, individuals with the rs5911 TG genotype showed significantly increased inhibition of platelet aggregation (IPA) by both 15 and 50 μM ticagrelor incubation (P < 0.05, respectively). Neither rs4642 nor rs4634 polymorphism affected ticagrelor-induced IPA. We suggest that the ITGA2B rs5911 GG genotype is associated with decreased ex vivo antiplatelet activity of ticagrelor in healthy Chinese male subjects.
Collapse
Affiliation(s)
- Mu-Peng Li
- Institute of Clinical Pharmacology, Central South University, Changsha, 410078, Hunan, China
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Thomas MR, Storey RF. Genetics of Response to Antiplatelet Therapy. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2014; 124:123-53. [DOI: 10.1016/b978-0-12-386930-2.00006-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
40
|
Verdoia M, Secco GG, Barbieri L, Cassetti E, Schaffer A, Sinigaglia F, Marino P, Suryapranata H, De Luca G. Platelet HPA-1 a/HPA-1 b polymorphism and the risk of periprocedural myocardial infarction in patients undergoing elective PCI. Platelets 2013; 25:367-72. [PMID: 24283589 DOI: 10.3109/09537104.2013.821602] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Periprocedural myocardial infarction (PMI) represents a relatively common complication of percutaneous coronary intervention (PCI) and large interests have been focused on platelets in order to prevent such a complication. The single nucleotide polymorphism Leu33Pro of platelet glycoprotein IIIa has been related to an increased platelet reactivity, a lower response to antiplatelet agents and higher risk of stent restenosis. Therefore, aim of our study was to evaluate the impact of this polymorphism on PMI in elective patients undergoing PCI. Our population is represented by 422 consecutive patients with cardiac biomarkers within normality undergoing elective PCI. We measured cardiac biomarkers (CK-MB and Troponin I) at baseline, and 8, 24 and 48 hours after the procedure. For all subjects, we performed genetic analysis to assess the presence of Leu33Pro polymorphism. A total of 136 patients (32.2%) were polymorphic. Those patients were younger (p = 0.03) and more often dislypidemic (p = 0.01). Angiographic features did not differ according to genetic status. Pharmacological treatment pre and during angioplasty was similar. PCI-related complications did not differ according to genotype, with the only exception of higher rate of distal embolization in polymorphic patients. However, Leu33Pro polymorphism was not associated with increased risk of periprocedural myonecrosis and PMI even after correction for baseline differences, (respectively OR = 1.22 [0.81-1.84], p = 0.34 for myonecrosis and OR = 1.66 [0.85-3.23]; p = 0.14 for PMI). At subgroup analysis, the Leu33Pro substitution was associated with higher risk of PMI only among diabetics (adjusted OR = 4.46 [1.12-17.76], p = 0.03). Among patients undergoing elective PCI, the polymorphism Leu33Pro of platelet glycoprotein IIIa is associated with increased risk of PMI only in diabetic patients.
Collapse
Affiliation(s)
- Monica Verdoia
- Division of Cardiology, Azienda Ospedaliera-Universitaria "Maggiore della Carità", Eastern Piedmont University , Novara , Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Geisler T, Schaeffeler E, Gawaz M, Schwab M. Genetic variation of platelet function and pharmacology: an update of current knowledge. Thromb Haemost 2013; 110:876-87. [PMID: 23702580 DOI: 10.1160/th13-02-0145] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Accepted: 04/25/2013] [Indexed: 11/05/2022]
Abstract
Platelets are critically involved in atherosclerosis and acute thrombosis. The platelet phenotype shows a wide variability documented by the inherited difference of platelet reactivity, platelet volume and count and function of platelet surface receptors. Several candidate genes have been put into focus and investigated for their functional and prognostic role in healthy individuals and patients with cardiovascular (CV) disease treated with antiplatelet agents. In addition to genetic variation, other clinical, disease-related and demographic factors are important so-called non-genetic factors. Due to the small effect sizes of single nucleotide polymorphisms (SNP) in candidate genes and due to the low allele frequencies of functional relevant candidate SNPs, the identification of genetic risk factors with high predictive values generally depends on the sample size of study cohorts. In the post-genome era new array and bioinformatic technologies facilitate high throughput genome-wide association studies (GWAS) for the identification of novel candidate genes in large cardiovascular cohorts. One of the crucial aspects of platelet genomic studies is the precise definition of a specific clinical phenotype (e.g. stent thrombosis) as this will impact importantly the findings of genomic studies like GWAS. Here, we provide an update on genetic variation of platelet receptors and drug metabolising enzymes under specific consideration of data derived by GWAS. The potential impact of this information and the role in personalised therapeutic concepts will be discussed.
Collapse
Affiliation(s)
- T Geisler
- Tobias Geisler, Medizinische Klinik, Innere Medizin III, Kardiologie und Kreislauferkrankungen, Otfried-Müller-Str. 10, 72076 Tübingen, Germany, Tel: +49 7071 29 83688, E-mail:
| | | | | | | |
Collapse
|
42
|
Abstract
From the initial description of platelets in 1882, their propensity to aggregate and to contribute to thrombosis was apparent. Indeed, excessive platelet aggregation is associated with myocardial infarction and other thrombotic diseases whereas Glanzmann thrombasthenia, in which platelet aggregation is reduced, is a bleeding syndrome. Over the last half of the 20th century, many investigators have provided insights into the cellular and molecular basis for platelet aggregation. The major membrane protein on platelets, integrin αIIbβ3, mediates this response by rapidly transiting from its resting to an activated state in which it serves as a receptor for ligands that can bridge platelets together. Monoclonal antibodies, natural products, and small peptides were all shown to inhibit αIIbβ3 dependent platelet aggregation, and these inhibitors became the forerunners of antagonists that proceeded through preclinical testing and into large patient trials to treat acute coronary syndromes, particularly in the context of percutaneous coronary interventions. Three such αIIbβ3 antagonists, abciximab, eptifibatide, and tirofiban, received Food and Drug Administration approval. Over the past 15 years, millions of patients have been treated with these αIIbβ3 antagonists and many lives have been saved by their administration. With the side effect of increased bleeding and the development of new antithrombotic drugs, the use of αIIbβ3 antagonists is waning. Nevertheless, they are still widely used for the prevention of periprocedural thrombosis during percutaneous coronary interventions. This review focuses on the biology of αIIbβ3, the development of its antagonists, and some of the triumphs and shortcomings of αIIbβ3 antagonism.
Collapse
Affiliation(s)
- Kamila Bledzka
- Department of Molecular Cardiology, Joseph J. Jacobs Center for Thrombosis and Vascular Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | | | | |
Collapse
|
43
|
Stewart A, Katznelson R, Kraeva N, Carroll J, Pickworth T, Rao V, Djaiani G. Genetic variation and cognitive dysfunction one year after cardiac surgery. Anaesthesia 2013; 68:571-5. [DOI: 10.1111/anae.12170] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/27/2012] [Indexed: 11/28/2022]
Affiliation(s)
| | - R. Katznelson
- Department of Anesthesia and Pain Management; Toronto General Hospital; University Health Network; University of Toronto; Toronto; Canada
| | - N. Kraeva
- Department of Anesthesia and Pain Management; Toronto General Hospital; University Health Network; University of Toronto; Toronto; Canada
| | - J. Carroll
- Department of Anesthesia and Pain Management; Toronto General Hospital; University Health Network; University of Toronto; Toronto; Canada
| | - T. Pickworth
- Department of Anesthesia and Pain Management; Toronto General Hospital; University Health Network; University of Toronto; Toronto; Canada
| | - V. Rao
- Division of Cardiovascular Surgery; Toronto General Hospital; University Health Network; University of Toronto; Toronto; Canada
| | - G. Djaiani
- Department of Anesthesia and Pain Management; Toronto General Hospital; University Health Network; University of Toronto; Toronto; Canada
| |
Collapse
|
44
|
|
45
|
Williams MS. Platelets and depression in cardiovascular disease: A brief review of the current literature. World J Psychiatry 2012; 2:114-23. [PMID: 24175177 PMCID: PMC3782186 DOI: 10.5498/wjp.v2.i6.114] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2011] [Revised: 09/19/2012] [Accepted: 11/17/2012] [Indexed: 02/05/2023] Open
Abstract
Major depression is an independent risk factor for cardiovascular mortality and morbidity. The exact mechanisms linking depression and increased cardiovascular risk remain poorly understood. Several mechanisms have been proposed including increased platelet reactivity. This review focuses on the current literature that examines the platelet hypothesis of depression. To date studies show increased serotonin response, increased platelet serotonin receptor density, decreased serotonin transporter binding, and decreased platelet serotonin levels in individuals with depression. However other studies have shown no change in serotonin uptake. In addition to platelet serotonin specific pathways, other platelet pathways that have shown significant changes in depressed individuals include blunting of the platelet adenosine response, increased platelet thrombin response, increased glycoprotein Ib expression, increased P-selectin, β thromboglobulin, and platelet factor four, as well as decreased platelet brain derived neurotrophic factor. However there are other studies that show conflicting evidence of increased platelet activation as measured by integrin receptor α2bβ3. Other conflicting data include α adrenergic density and platelet response to augmented serotonin. The direction of future research in platelet functional changes in depression and coronary artery disease should continue to focus on serotonin specific pathways with emphasis on potential mechanisms of specific pathway changes.
Collapse
Affiliation(s)
- Marlene S Williams
- Marlene S Williams, Division of Cardiology, Johns Hopkins Bayview Medical Center, The Johns Hopkins University, Baltimore, MD 21224, United States
| |
Collapse
|
46
|
The a1/a2 polymorphism of the glycoprotein IIIa gene and myocardial infarction in Caucasians with type 2 diabetes. Mol Biol Rep 2012. [PMID: 23184042 DOI: 10.1007/s11033-012-2265-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
A PlA1/A2 polymorphism of glycoprotein IIIa is known to be involved in the pathogenesis of coronary artery disease (CAD) and myocardial infarction (MI). The aim of this study was to investigate an association between the PlA1/A2 polymorphism of the glycoprotein IIIa gene and the risk of MI in Caucasians with type 2 diabetes. 549 Caucasians with type 2 diabetes were enrolled in the cross sectional retrospective case-control study: 224 patients with MI and 325 diabetic subjects without CAD. Blood biochemical analysis was performed. The polymerase chain reaction with restriction fragment length polymorphism was used for genetic analysis. Patients with MI were older (62 ± 11.8 vs. 58.5 ± 11.6 years; P < 0.001), and had a longer duration of type 2 diabetes (17.6 ± 8.9 vs. 15.1 ± 9.2; P = 0.01) compared to the diabetics without CAD. A significant difference in distribution of the A2A2 genotype of glycoprotein IIIa was not found between 224 diabetic patients with MI in comparison to 325 diabetics without CAD (11.6 vs. 14,1 %; n.s.). The diabetes duration and male sex were independent factors for the development of MI, whereas the PlA1/A2 polymorphism of glycoprotein IIIa was not. To conclude, The A2A2 genotype of the glycoprotein IIIa polymorphism was not associated with MI risk in Caucasians with type 2 diabetes.
Collapse
|
47
|
Abstract
PURPOSE OF REVIEW This review summarizes our current knowledge of common gene variants (polymorphisms) that have small individual effects on platelet function in humans, but can cumulatively lead to hyperreactive platelets and increase risk for negative outcomes in thrombotic disorders. RECENT FINDINGS Candidate gene association and genome-wide association studies (GWAS) have identified loci that include single nucleotide polymorphisms, which exert a cumulative effect on platelet function by modifying basic platelet parameters, such as mean platelet volume (MPV) or platelet count, by altering the expression or activity of key platelet receptors, or by influencing downstream effector pathways utilized by these receptors. SUMMARY Variation in MPV between normal individuals is responsible for roughly a two-fold range in platelet protein content, including key surface receptors and reactive granule constituents, the association of ADRA2, GP1BA, GP6, ITGA2 and P2Y12 variants with platelet reactivity, initially identified by candidate gene analyses, has now been validated by genome-wide approaches in much larger individual cohorts, and GWAS have identified novel gene variants, most notably PEAR1, that participate in variation in platelet reactivity among normal individuals, all of which contribute to a genetic basis for differences in platelet reactivty among normal individuals.
Collapse
|
48
|
Clinical Impact of Genetically Determined Platelet Reactivity. J Cardiovasc Transl Res 2012; 6:398-403. [DOI: 10.1007/s12265-012-9421-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Accepted: 10/30/2012] [Indexed: 01/04/2023]
|
49
|
Depta JP, Bhatt DL. Aspirin and platelet adenosine diphosphate receptor antagonists in acute coronary syndromes and percutaneous coronary intervention: role in therapy and strategies to overcome resistance. Am J Cardiovasc Drugs 2012; 8:91-112. [PMID: 18422393 DOI: 10.1007/bf03256587] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Platelet activation and aggregation are key components in the cascade of events causing thrombosis following plaque rupture. Antiplatelet therapy is essential in the treatment of patients with acute coronary syndromes (ACS) and for those requiring percutaneous coronary intervention (PCI). Aspirin (acetylsalicylic acid) is a well established antiplatelet therapy and is mandated for secondary prevention of cardiovascular events following ACS. In patients with ACS, the addition of clopidogrel to aspirin is more effective than aspirin alone. For patients undergoing PCI, dual antiplatelet therapy with aspirin and clopidogrel is warranted. Aspirin should be continued indefinitely after PCI. Pretreatment of patients with clopidogrel prior to PCI lowers the incidence of cardiovascular events, yet the optimum timing of drug administration and dose are still being investigated, as is the duration of therapy following PCI. Late-stent thrombosis with drug-eluting stents has pushed the recommendation for duration of clopidogrel therapy up to 1 year and perhaps beyond, in patients without risks for bleeding. The concepts of aspirin and clopidogrel resistance are important clinical questions. No uniform definition exists for aspirin or clopidogrel resistance. Measurements of resistance are often highly variable and do not necessarily correlate with clinical resistance. Noncompliance remains the most prominent mode of resistance. Screening of selected patient populations for resistance or pharmacologic intervention of those patients termed 'resistant' warrants further study.
Collapse
Affiliation(s)
- Jeremiah P Depta
- Department of Internal Medicine, Cleveland Clinic, Cleveland, Ohio, USA.
| | | |
Collapse
|
50
|
Williams PJ, Morgan L. The role of genetics in pre-eclampsia and potential pharmacogenomic interventions. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2012; 5:37-51. [PMID: 23226061 PMCID: PMC3513227 DOI: 10.2147/pgpm.s23141] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Indexed: 01/23/2023]
Abstract
The pregnancy-specific condition pre-eclampsia not only affects the health of mother and baby during pregnancy but also has long-term consequences, increasing the chances of cardiovascular disease in later life. It is accepted that pre-eclampsia has a placental origin, but the pathogenic mechanisms leading to the systemic endothelial dysfunction characteristic of the disorder remain to be determined. In this review we discuss some key factors regarded as important in the development of pre-eclampsia, including immune maladaptation, inadequate placentation, oxidative stress, and thrombosis. Genetic factors influence all of these proposed pathophysiological mechanisms. The inherited nature of pre-eclampsia has been known for many years, and extensive genetic studies have been undertaken in this area. Genetic research offers an attractive strategy for studying the pathogenesis of pre-eclampsia as it avoids the ethical and practical difficulties of conducting basic science research during the preclinical phase of pre-eclampsia when the underlying pathological changes occur. Although pharmacogenomic studies have not yet been conducted in pre-eclampsia, a number of studies investigating treatment for essential hypertension are of relevance to therapies used in pre-eclampsia. The pharmacogenomics of antiplatelet agents, alpha and beta blockers, calcium channel blockers, and magnesium sulfate are discussed in relation to the treatment and prevention of pre-eclampsia. Pharmacogenomics offers the prospect of individualized patient treatment, ensuring swift introduction of optimal treatment whilst minimizing the use of inappropriate or ineffective drugs, thereby reducing the risk of harmful effects to both mother and baby.
Collapse
|