1
|
Amini AM, Zhou R, Austermann K, Králová D, Serra G, Ibrahim IS, Corona G, Bergillos-Meca T, Aboufarrag H, Kroon PA, Spencer JP, Yaqoob P. Acute effects of an anthocyanin-rich blackcurrant beverage on markers of cardiovascular disease risk in healthy adults: a randomized, double-blind, placebo-controlled, crossover trial. J Nutr 2025:S0022-3166(25)00299-8. [PMID: 40414296 DOI: 10.1016/j.tjnut.2025.05.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 05/08/2025] [Accepted: 05/14/2025] [Indexed: 05/27/2025] Open
Abstract
BACKGROUND Epidemiological evidence suggests an inverse association between anthocyanin consumption and cardiovascular disease (CVD) risk. Modulation of vascular function and hemostasis may contribute to this, but there is limited clinical evidence. OBJECTIVE The present study investigated the acute effects of an anthocyanin-rich blackcurrant beverage, compared with a matched placebo, on selected markers of CVD risk in healthy middle-aged subjects in response to a high-fat meal. METHODS Twenty-three volunteers aged 39.9 ± 8.1 years (BMI 22.9 ± 2.3 kg/m2) completed a double-blind, randomized, placebo-controlled, crossover trial. Volunteers consumed either 200 mL blackcurrant beverage (744 mg polyphenols comprising 711 mg anthocyanins and 32 mg procyanidins) or a placebo, together with a high-fat breakfast (52.3 g fat) followed by a lunch (30 g fat) at 3 h, and the postprandial vascular response was compared. The primary endpoints were the assessment of vascular function by flow-mediated dilation (FMD) and the inhibition of collagen- and adenosine diphosphate-induced platelet aggregation. Secondary outcomes included blood pressure (BP), digital volume pulse waveforms, circulating numbers of endothelium- and platelet-derived extracellular vesicles (EVs), plasma concentrations of interleukin (IL)-8, and plasma and urinary concentrations of polyphenols and their metabolites were also evaluated. RESULTS There was a significant cumulative improvement in FMD following consumption of an anthocyanin-rich blackcurrant beverage compared with a matched placebo in conjunction with a high-fat meal over a 6 h postprandial period. There was a trend for an inhibitory effect of the blackcurrant beverage on agonist-induced platelet aggregation and significant effects on the secondary outcomes, SBP and IL-8, although these were exploratory and not adjusted for multiple testing. Plasma concentrations of hippuric acid and isovanillic acid were strong independent predictors of FMD and 4-hydroxybenzaldehyde and isoferulic acid glucuronide were predictors of SBP and DBP. CONCLUSIONS An anthocyanin-rich blackcurrant beverage mitigated the effects of a high fat meal on vascular function and markers of CVD risk, and this is associated with the appearance of specific plasma anthocyanin phenolic metabolites. REGISTRATION URL: https://classic. CLINICALTRIALS gov/ct2/show/NCT02459756; Unique identifier: NCT02459756.
Collapse
Affiliation(s)
- Anna M Amini
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences, University of Reading, Reading, United Kingdom
| | - Ruihan Zhou
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences, University of Reading, Reading, United Kingdom; Current address: Division of Pediatric Oncology and Pediatric Surgery, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Katharina Austermann
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences, University of Reading, Reading, United Kingdom
| | - Dominika Králová
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences, University of Reading, Reading, United Kingdom
| | - Gessica Serra
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences, University of Reading, Reading, United Kingdom
| | - Ibrahim S Ibrahim
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences, University of Reading, Reading, United Kingdom
| | - Giulia Corona
- School of Life and Health Sciences, Whitelands College, University of Roehampton, Holybourne Avenue, London, United Kingdom
| | - Triana Bergillos-Meca
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences, University of Reading, Reading, United Kingdom
| | - Hassan Aboufarrag
- Quadram Institute Bioscience, Norwich Research Park, Norwich, United Kingdom; Current address: Department of Food Science and Technology, Faculty of Agriculture, Alexandria University, Alexandria 21545, Egypt
| | - Paul A Kroon
- Quadram Institute Bioscience, Norwich Research Park, Norwich, United Kingdom
| | - Jeremy Pe Spencer
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences, University of Reading, Reading, United Kingdom
| | - Parveen Yaqoob
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences, University of Reading, Reading, United Kingdom.
| |
Collapse
|
2
|
Pennings GJ, Reddel CJ, Chen VM, Gnanenthiran SR, Kritharides L. Perspective: Collagen induced platelet activation via the GPVI receptor as a primary target of colchicine in cardiovascular disease. Front Cardiovasc Med 2023; 9:1104744. [PMID: 36741844 PMCID: PMC9892722 DOI: 10.3389/fcvm.2022.1104744] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 12/30/2022] [Indexed: 01/20/2023] Open
Abstract
Colchicine has been demonstrated to reduce cardiovascular death, myocardial infarction (MI), ischemic stroke, and ischemia-driven coronary revascularization in people with coronary artery disease (CAD). These reductions were observed even in patients already taking antiplatelet therapy. As well as having anti-inflammatory effects, colchicine demonstrates antiplatelet effects. We propose that colchicine's antiplatelet effects primarily target collagen-induced platelet activation via the collagen receptor, glycoprotein (GP)VI, which is critical for arterial thrombosis formation. In settings such as stroke and MI, GPVI signaling is upregulated. We have demonstrated in vitro that therapeutic concentrations of colchicine lead to a decrease in collagen-induced platelet aggregation and alter GPVI signaling. Clinical studies of colchicine given for 6 months lead to a significant reduction in serum GPVI levels in CAD patients, which may ameliorate thrombotic risk. Future evaluation of the effects of colchicine in clinical trials should include assessment of its effects on collagen-mediated platelet activation, and consideration be given to quantifying the contribution of such antiplatelet effects additional to the known anti-inflammatory effects of colchicine.
Collapse
Affiliation(s)
- Gabrielle J. Pennings
- Vascular Biology Group, ANZAC Research Institute, The University of Sydney, Concord, NSW, Australia,Department of Cardiology, Concord Repatriation General Hospital, Concord, NSW, Australia
| | - Caroline J. Reddel
- Vascular Biology Group, ANZAC Research Institute, The University of Sydney, Concord, NSW, Australia
| | - Vivien M. Chen
- Department of Haematology, Concord Repatriation General Hospital, Concord, NSW, Australia,Platelet, Thrombosis Research Laboratory, ANZAC Research Institute, The University of Sydney, Concord, NSW, Australia
| | - Sonali R. Gnanenthiran
- Vascular Biology Group, ANZAC Research Institute, The University of Sydney, Concord, NSW, Australia,Department of Cardiology, Concord Repatriation General Hospital, Concord, NSW, Australia,The George Institute for Global Health, University of New South Wales, Newtown, NSW, Australia
| | - Leonard Kritharides
- Vascular Biology Group, ANZAC Research Institute, The University of Sydney, Concord, NSW, Australia,Department of Cardiology, Concord Repatriation General Hospital, Concord, NSW, Australia,*Correspondence: Leonard Kritharides ✉
| |
Collapse
|
3
|
Cofer LB, Barrett TJ, Berger JS. Aspirin for the Primary Prevention of Cardiovascular Disease: Time for a Platelet-Guided Approach. Arterioscler Thromb Vasc Biol 2022; 42:1207-1216. [PMID: 36047408 PMCID: PMC9484763 DOI: 10.1161/atvbaha.122.318020] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 08/10/2022] [Indexed: 11/17/2022]
Abstract
Aspirin protects against atherothrombosis while increasing the risk of major bleeding. Although it is widely used to prevent cardiovascular disease (CVD), its benefit does not outweigh its risk for primary CVD prevention in large population settings. The recent United States Preventive Services Task Force guidelines on aspirin use to prevent CVD reflect this clinical tradeoff as well as the persistent struggle to define a population that would benefit from prophylactic aspirin therapy. Past clinical trials of primary CVD prevention with aspirin have not included consideration of a biomarker relevant to aspirin's mechanism of action, platelet inhibition. This approach is at odds with the paradigm used in other key areas of pharmacological CVD prevention, including antihypertensive and statin therapy, which combine cardiovascular risk assessment with the measurement of mechanistic biomarkers (eg, blood pressure and LDL [low-density lipoprotein]-cholesterol). Reliable methods for quantifying platelet activity, including light transmission aggregometry and platelet transcriptomics, exist and should be considered to identify individuals at elevated cardiovascular risk due to a hyperreactive platelet phenotype. Therefore, we propose a new, platelet-guided approach to the study of prophylactic aspirin therapy. We think that this new approach will reveal a population with hyperreactive platelets who will benefit most from primary CVD prevention with aspirin and usher in a new era of precision-guided antiplatelet therapy.
Collapse
|
4
|
Li XZ, Liu Q, Zhang SN, Yang WD, Zhou Y. Biolabel-led research pattern reveals serum profile in rats after treatment with Herba Lysimachiae: Combined analysis of metabonomics and proteomics. Biomed Chromatogr 2022; 36:e5385. [PMID: 35445417 DOI: 10.1002/bmc.5385] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 04/08/2022] [Accepted: 04/18/2022] [Indexed: 11/07/2022]
Abstract
In traditional Chinese medicine, Herba Lysimachiae (HL) is mainly used to treat rheumatic arthralgia. Current pharmacological studies also showed that HL has therapeutic potential for synovial diseases. HL is an oral drug, whose compounds need to enter the blood circulation before reaching the injured tissue, thus potentially causing activity or toxicity to the blood system. In this study, the biolabel-led research pattern was used to analyze the serum profile after HL intervention, based on which the safety and efficacy of HL were explored. Metabonomics and proteomics were combined to analyze the biolabels responsible for the interventions of HL on serum. Bioinformatics databases were used to screen for the material basis that may interfere with biolabels. Omics analysis showed that differentially expressed 19 proteins and 5 metabolites were identified and considered as the potential biolabels, which were involved in 8 biochemical processes (platelet activation and aggregation, blood glucose release, immune and inflammatory regulation, oxidative stress, endoplasmic reticulum stress, tumor progression, blood pressure regulation, and uric acid synthesis). Thirty-one compounds may be the material basis to interfere with eleven biolabels. The present research reveals that the potential activities and toxicities of HL can be explored based on the biolabel-led research pattern.
Collapse
Affiliation(s)
- Xu-Zhao Li
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guian New Area, China
| | - Qi Liu
- The Research Institute of Medicine and Pharmacy, Qiqihar Medical University, Qiqihar, China
| | - Shuai-Nan Zhang
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guian New Area, China
| | - Wu-de Yang
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guian New Area, China
| | - Ying Zhou
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guian New Area, China
| |
Collapse
|
5
|
Myers RA, Ortel TL, Waldrop A, Dave S, Ginsburg GS, Voora D. Aspirin effects on platelet gene expression are associated with a paradoxical, increase in platelet function. Br J Clin Pharmacol 2021; 88:2074-2083. [PMID: 34705291 PMCID: PMC9007832 DOI: 10.1111/bcp.15127] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 10/10/2021] [Accepted: 10/18/2021] [Indexed: 01/04/2023] Open
Abstract
Aspirin has known effects beyond inhibiting platelet cyclooxygenase-1 (COX-1) that have been incompletely characterized. Transcriptomics can comprehensively characterize the on- and off-target effects of medications. We used a systems pharmacogenomics approach of aspirin exposure in volunteers coupled with serial platelet function and purified platelet mRNA sequencing to test the hypothesis that aspirin's effects on the platelet transcriptome are associated with platelet function. We prospectively recruited 74 adult volunteers for a randomized crossover study of 81- vs. 325 mg/day, each for 4 weeks. Using mRNA sequencing of purified platelets collected before and after each 4-week exposure, we identified 208 aspirin-responsive genes with no evidence for dosage effects. In independent cohorts of healthy volunteers and patients with diabetes, we validated aspirin's effects on five genes: EIF2S3, CHRNB1, EPAS1, SLC9A3R2 and HLA-DRA. Functional characterization of the effects of aspirin on mRNA as well as platelet ribosomal RNA demonstrated that aspirin may act as an inhibitor of protein synthesis. Database searches for small molecules that mimicked the effects of aspirin on platelet gene expression in vitro identified aspirin but no other molecules that share aspirin's known mechanisms of action. The effects of aspirin on platelet mRNA were correlated with higher levels of platelet function both at baseline and after aspirin exposure-an effect that counteracts aspirin's known antiplatelet effect. In summary, this work collectively demonstrates a dose-independent effect of aspirin on the platelet transcriptome that counteracts the well-known antiplatelet effects of aspirin.
Collapse
Affiliation(s)
- Rachel A Myers
- Center for Applied Genomics & Precision Medicine, Duke University School of Medicine, Durham, NC, United States
| | - Thomas L Ortel
- Division of Hematology, Department of Medicine, Duke University Medical Center, Durham, NC, United States
| | - Alexander Waldrop
- Center for Genomics and Computational Biology, Duke University, Durham, NC, United States
| | - Sandeep Dave
- Center for Genomics and Computational Biology, Duke University, Durham, NC, United States
| | - Geoffrey S Ginsburg
- Center for Applied Genomics & Precision Medicine, Duke University School of Medicine, Durham, NC, United States
| | - Deepak Voora
- Center for Applied Genomics & Precision Medicine, Duke University School of Medicine, Durham, NC, United States
| |
Collapse
|
6
|
Keramati AR, Chen MH, Rodriguez BAT, Yanek LR, Bhan A, Gaynor BJ, Ryan K, Brody JA, Zhong X, Wei Q, Kammers K, Kanchan K, Iyer K, Kowalski MH, Pitsillides AN, Cupples LA, Li B, Schlaeger TM, Shuldiner AR, O'Connell JR, Ruczinski I, Mitchell BD, Faraday N, Taub MA, Becker LC, Lewis JP, Mathias RA, Johnson AD. Genome sequencing unveils a regulatory landscape of platelet reactivity. Nat Commun 2021; 12:3626. [PMID: 34131117 PMCID: PMC8206369 DOI: 10.1038/s41467-021-23470-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 04/13/2021] [Indexed: 12/16/2022] Open
Abstract
Platelet aggregation at the site of atherosclerotic vascular injury is the underlying pathophysiology of myocardial infarction and stroke. To build upon prior GWAS, here we report on 16 loci identified through a whole genome sequencing (WGS) approach in 3,855 NHLBI Trans-Omics for Precision Medicine (TOPMed) participants deeply phenotyped for platelet aggregation. We identify the RGS18 locus, which encodes a myeloerythroid lineage-specific regulator of G-protein signaling that co-localizes with expression quantitative trait loci (eQTL) signatures for RGS18 expression in platelets. Gene-based approaches implicate the SVEP1 gene, a known contributor of coronary artery disease risk. Sentinel variants at RGS18 and PEAR1 are associated with thrombosis risk and increased gastrointestinal bleeding risk, respectively. Our WGS findings add to previously identified GWAS loci, provide insights regarding the mechanism(s) by which genetics may influence cardiovascular disease risk, and underscore the importance of rare variant and regulatory approaches to identifying loci contributing to complex phenotypes.
Collapse
Affiliation(s)
- Ali R Keramati
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- GeneSTAR Research Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ming-Huei Chen
- Division of Intramural Research, Population Sciences Branch, National Heart, Lung and Blood Institute, Bethesda, MD, USA
- The Framingham Heart Study, Framingham, MA, USA
| | - Benjamin A T Rodriguez
- Division of Intramural Research, Population Sciences Branch, National Heart, Lung and Blood Institute, Bethesda, MD, USA
- The Framingham Heart Study, Framingham, MA, USA
- Valo Health, Boston, MA, USA
| | - Lisa R Yanek
- GeneSTAR Research Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Division of General Internal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Brady J Gaynor
- Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, MD, USA
- Program in Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, Baltimore, MD, USA
| | - Kathleen Ryan
- Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, MD, USA
- Program in Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, Baltimore, MD, USA
| | - Jennifer A Brody
- Cardiovascular Health Research Unit, University of Washington School of Medicine, Seattle, WA, USA
| | - Xue Zhong
- Vanderbilt Genetics Institute, Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Qiang Wei
- Vanderbilt Genetics Institute, Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Kai Kammers
- Biostatistics and Bioinformatics, Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kanika Kanchan
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kruthika Iyer
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Madeline H Kowalski
- Department of Biostatistics, University of North Carolina, Chapel Hill, NC, USA
| | - Achilleas N Pitsillides
- The Framingham Heart Study, Framingham, MA, USA
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA, USA
| | - L Adrienne Cupples
- The Framingham Heart Study, Framingham, MA, USA
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA, USA
| | - Bingshan Li
- Vanderbilt Genetics Institute, Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | | | - Alan R Shuldiner
- Program in Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, Baltimore, MD, USA
| | - Jeffrey R O'Connell
- Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, MD, USA
- Program in Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, Baltimore, MD, USA
| | - Ingo Ruczinski
- Bloomberg School of Public Health, Biostatistics, Johns Hopkins University, Baltimore, MD, USA
| | - Braxton D Mitchell
- Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, MD, USA
- Program in Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, Baltimore, MD, USA
| | - Nauder Faraday
- GeneSTAR Research Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Margaret A Taub
- Bloomberg School of Public Health, Biostatistics, Johns Hopkins University, Baltimore, MD, USA
| | - Lewis C Becker
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- GeneSTAR Research Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Joshua P Lewis
- Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, MD, USA.
- Program in Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, Baltimore, MD, USA.
| | - Rasika A Mathias
- GeneSTAR Research Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Andrew D Johnson
- Division of Intramural Research, Population Sciences Branch, National Heart, Lung and Blood Institute, Bethesda, MD, USA.
- The Framingham Heart Study, Framingham, MA, USA.
| |
Collapse
|
7
|
Kontos A, Willoughby S, Lushington K, Martin J, Wabnitz D, Dorrian J, Kennedy D. Increased Platelet Aggregation in Children and Adolescents with Sleep-disordered Breathing. Am J Respir Crit Care Med 2020; 202:1560-1566. [PMID: 32628860 DOI: 10.1164/rccm.201911-2229oc] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Rationale: Sleep-disordered breathing (SDB) is associated with increased vascular resistance in children and adults. Persistent increased vascular resistance damages vascular endothelial cells-a marker of which is increased platelet activation.Objectives: This study compared whole-blood impedance platelet aggregation in children with clinically diagnosed SDB warranting adenotonsillectomy and healthy control subjects.Methods: Thirty children who had SDB warranting intervention clinically diagnosed by experienced pediatric otolaryngologists were recruited from adenotonsillectomy waitlists, and 20 healthy children from the community underwent overnight polysomnography to determine SDB severity (obstructive apnea-hypopnea index). Snoring frequency was collected from parents. In the morning, a fasting blood sample was taken, and whole-blood platelet aggregation was measured.Measurements and Main Results: Children with SDB exhibited increased platelet aggregation to TRAP (thrombin receptor-activating peptide) (children with SDB = 114.8 aggregation units [AU] vs. control subjects = 98.0 AU; P < 0.05) and COL antibody (96.7 vs. 82.2 AU; P < 0.05) and an increased trend in ADP antibody (82.3 vs. 69.2 AU; P < 0.07) but not aspirin dialuminate (82.1 vs. 79.5 AU; P > 0.05). No significant association was observed between either the obstructive apnea-hypopnea index and any aggregation parameter, but parental report of snoring was positively associated with TRAP aggregation (Kendall's τ-c = 0.23; P < 0.05).Conclusions: The finding of increased platelet aggregation is consistent with endothelial damage. This suggests that the profile of cardiovascular changes noted in adults with SDB may also occur in children with SDB.
Collapse
Affiliation(s)
- Anna Kontos
- Department of Respiratory and Sleep Medicine and.,Robinson Research Institute.,Discipline of Paediatrics, School of Medicine, and
| | - Scott Willoughby
- School of Medicine, University of Adelaide, Adelaide, South Australia, Australia; and
| | - Kurt Lushington
- Robinson Research Institute.,Centre for Behaviour, Brain and Body, Justice and Society Unit, University of South Australia, Adelaide, South Australia, Australia
| | - James Martin
- Department of Respiratory and Sleep Medicine and.,Robinson Research Institute.,Discipline of Paediatrics, School of Medicine, and
| | - David Wabnitz
- Department of Otolaryngology, Head and Neck Surgery, Women's and Children's Hospital, Adelaide, South Australia, Australia
| | - Jill Dorrian
- Centre for Behaviour, Brain and Body, Justice and Society Unit, University of South Australia, Adelaide, South Australia, Australia
| | - Declan Kennedy
- Department of Respiratory and Sleep Medicine and.,Robinson Research Institute.,Discipline of Paediatrics, School of Medicine, and
| |
Collapse
|
8
|
Friede KA, Infeld MM, Tan RS, Knickerbocker HJ, Myers RA, Dubois LG, Thompson JW, Kaddurah‐Daouk R, Ginsburg GS, Ortel TL, Voora D. Influence of Sex on Platelet Reactivity in Response to Aspirin. J Am Heart Assoc 2020; 9:e014726. [PMID: 32654613 PMCID: PMC7660714 DOI: 10.1161/jaha.119.014726] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 05/06/2020] [Indexed: 01/29/2023]
Abstract
Background There are sex differences in the efficacy and safety of aspirin for the prevention of myocardial infarction and stroke. Whether this is explained by underlying differences in platelet reactivity and aspirin response remains poorly understood. Methods and Results Healthy volunteers (n=378 208 women) and patients with coronary artery disease or coronary artery disease risk factors (n=217 112 women) took aspirin for 4 weeks. Light transmittance aggregometry using platelet-rich plasma was used to measure platelet reactivity in response to epinephrine, collagen, and ADP at baseline, 3 hours after the first aspirin dose, and after 4 weeks of daily aspirin therapy. A subset of patients underwent pharmacokinetic and pharmacodynamic assessment with levels of salicylate and cyclooxygenase-1-derived prostaglandin metabolites and light transmittance aggregometry in response to arachidonic acid and after ex vivo exposure to aspirin. At baseline, women had increased platelet aggregation in response to ADP and collagen. Innate platelet response to aspirin, assessed with ex vivo aspirin exposure of baseline platelets, did not differ by sex. Three hours after the first oral aspirin dose, platelet aggregation was inhibited in women to a greater degree in response to epinephrine and to a lesser degree with collagen. After 4 weeks of daily therapy, despite higher salicylate concentrations and greater cyclooxygenase-1 inhibition, women exhibited an attenuation of platelet inhibition in response to epinephrine and ADP. Conclusions We observed agonist-dependent sex differences in platelet responses to aspirin. Despite higher cyclooxygenase-1 inhibition, daily aspirin exposure resulted in a paradoxical attenuation of platelet inhibition in response to epinephrine and ADP over time in women but not in men.
Collapse
Affiliation(s)
- Kevin A. Friede
- Division of CardiologyDuke UniversityDurhamNC
- Center for Applied Genomics & Precision MedicineDuke UniversityDurhamNC
| | - Margaret M. Infeld
- Division of CardiologyLarner College of Medicine at the University of VermontBurlingtonVT
| | - Ru San Tan
- Department of CardiologyNational Heart CentreSingapore
| | | | - Rachel A. Myers
- Center for Applied Genomics & Precision MedicineDuke UniversityDurhamNC
| | - Laura G. Dubois
- Center for Genomic and Computational BiologyDuke UniversityDurhamNC
| | - J. Will Thompson
- Center for Genomic and Computational BiologyDuke UniversityDurhamNC
| | | | - Geoffrey S. Ginsburg
- Division of CardiologyDuke UniversityDurhamNC
- Center for Applied Genomics & Precision MedicineDuke UniversityDurhamNC
| | | | - Deepak Voora
- Division of CardiologyDuke UniversityDurhamNC
- Center for Applied Genomics & Precision MedicineDuke UniversityDurhamNC
| |
Collapse
|
9
|
Keramati AR, Yanek LR, Iyer K, Taub MA, Ruczinski I, Becker DM, Becker LC, Faraday N, Mathias RA. Targeted deep sequencing of the PEAR1 locus for platelet aggregation in European and African American families. Platelets 2018; 30:380-386. [PMID: 29553866 DOI: 10.1080/09537104.2018.1447659] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Coronary artery disease (CAD) remains a major cause of mortality and morbidity worldwide. The aggregation of activated platelets on a ruptured atherosclerotic plaque is a critical step in most acute cardiovascular events like myocardial infarction. Platelet aggregation both at baseline and after aspirin is highly heritable. Genome-wide association studies (GWAS) have identified a common variant within the first intron of the platelet endothelial aggregation receptor1 (PEAR1), to be robustly associated with platelet aggregation. In this study, we used targeted deep sequencing to fine-map the prior GWAS peak and identify additional rare variants of PEAR1 that account for missing heritability in platelet aggregation within the GeneSTAR families. In this study, 1709 subjects (1043 European Americans, EA and 666 African Americans, AA) from families in the GeneSTAR study were included. In vitro platelet aggregation in response to collagen, ADP and epinephrine was measured at baseline and 14 days after aspirin therapy (81 mg/day). Targeted deep sequencing of PEAR1 in addition to 2kb of upstream and downstream of the gene was performed. Under an additive genetic model, the association of single variants of PEAR1 with platelet aggregation phenotypes were examined. Additionally, we examined the association between the burden of PEAR1 rare non-synonymous variants and platelet aggregation phenotypes. Of 532 variants identified through sequencing, the intron 1 variant, rs12041331, was significantly associated with all platelet aggregation phenotypes at baseline and after platelet inhibition with aspirin therapy. rs12566888, which is in linkage disequilibrium with rs12041331, was associated with platelet aggregation phenotypes but to a lesser extent. In the EA families, the burden of PEAR1 missense variants was associated with platelet aggregation after aspirin therapy when the platelets were stimulated with epinephrine (p = 0.0009) and collagen (p = 0.03). In AAs, the burden of PEAR1 missense variants was associated, to a lesser degree, with platelet aggregation in response to epinephrine (p = 0.02) and ADP (p = 0.04). Our study confirmed that the GWAS-identified variant, rs12041331, is the strongest variant associated with platelet aggregation both at baseline and after aspirin therapy in our GeneSTAR families in both races. We identified additional association of rare missense variants in PEAR1 with platelet aggregation following aspirin therapy. However, we observed a racial difference in the contribution of these rare variants to the platelet aggregation, most likely due to higher residual missing heritability of platelet aggregation after accounting for rs12041331 in the EAs compared to AAs.
Collapse
Affiliation(s)
- Ali R Keramati
- a GeneSTAR Research Program Department of Medicine, Division of General Internal Medicine , Johns Hopkins University School of Medicine , Baltimore , MD , USA.,b Department of Medicine, Division of Cardiology , Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Lisa R Yanek
- a GeneSTAR Research Program Department of Medicine, Division of General Internal Medicine , Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Kruthika Iyer
- a GeneSTAR Research Program Department of Medicine, Division of General Internal Medicine , Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Margaret A Taub
- c Department of Biostatistics , Johns Hopkins University Bloomberg School of Public Health , Baltimore , MD , USA
| | - Ingo Ruczinski
- c Department of Biostatistics , Johns Hopkins University Bloomberg School of Public Health , Baltimore , MD , USA
| | - Diane M Becker
- a GeneSTAR Research Program Department of Medicine, Division of General Internal Medicine , Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Lewis C Becker
- a GeneSTAR Research Program Department of Medicine, Division of General Internal Medicine , Johns Hopkins University School of Medicine , Baltimore , MD , USA.,b Department of Medicine, Division of Cardiology , Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Nauder Faraday
- a GeneSTAR Research Program Department of Medicine, Division of General Internal Medicine , Johns Hopkins University School of Medicine , Baltimore , MD , USA.,d Department of Anesthesiology and Critical Care Medicine , Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Rasika A Mathias
- a GeneSTAR Research Program Department of Medicine, Division of General Internal Medicine , Johns Hopkins University School of Medicine , Baltimore , MD , USA.,e Department of Medicine, Division of Allergy and Clinical Immunology , Johns Hopkins University School of Medicine , Baltimore , MD , USA
| |
Collapse
|
10
|
Puurunen MK, Hwang SJ, Larson MG, Vasan RS, O'Donnell CJ, Tofler G, Johnson AD. ADP Platelet Hyperreactivity Predicts Cardiovascular Disease in the FHS (Framingham Heart Study). J Am Heart Assoc 2018; 7:JAHA.118.008522. [PMID: 29502103 PMCID: PMC5866343 DOI: 10.1161/jaha.118.008522] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Platelet function is associated with adverse events in patients with cardiovascular disease (CVD). METHODS AND RESULTS We examined associations of baseline platelet function with incident CVD events in the community-based FHS (Framingham Heart Study). Participants free of prevalent CVD and without recent aspirin treatment with available data in the Framingham Offspring cohort (1991-1995) and Omni cohort (1994-1998) were included. Platelet function was measured with light transmission aggregometry using collagen (1.9 μg/mL), ADP (0.05-15 μmol/L), and epinephrine (0.01-15 μmol/L). We used proportional hazards models to analyze incident outcomes (myocardial infarction/stroke, CVD, and CVD mortality) with respect to platelet measures. The study sample included 2831 participants (average age, 54.3 years; 57% women). During follow-up (median, 20.4 years), we observed 191 composite incident myocardial infarction or stroke events, 432 incident CVD cases, and 117 CVD deaths. Hyperreactivity to ADP and platelet aggregation at ADP concentration of 1.0 μmol/L were significantly associated with incident myocardial infarction/stroke in a multivariable model (hazard ratio, 1.68 [95% confidence interval, 1.13-2.50] [P=0.011] for hyperreactivity across ADP doses; and hazard ratio, 1.16 [95% confidence interval, 1.02-1.33] [P=0.029] for highest quartile of ADP response at 1.0 μmol/L versus others). No association was observed for collagen lag time or any epinephrine measures with incident myocardial infarction or stroke. CONCLUSIONS Intrinsic hyperreactivity to low-dose ADP in our community-based sample, who were free of CVD and any antiplatelet therapy, is associated with future arterial thrombosis during a 20-year follow-up. These findings reinforce ADP activation inhibition as a critical treatment paradigm and encourage further study of ADP inhibitor-refractive populations.
Collapse
Affiliation(s)
- Marja K Puurunen
- National Heart, Lung, and Blood Institute's and Boston University's The Framingham Heart Study, Framingham, MA.,Schools of Medicine and Public Health, Boston University, Boston, MA
| | - Shih-Jen Hwang
- National Heart, Lung, and Blood Institute's and Boston University's The Framingham Heart Study, Framingham, MA.,Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Framingham, MA
| | - Martin G Larson
- National Heart, Lung, and Blood Institute's and Boston University's The Framingham Heart Study, Framingham, MA.,Biostatistics Department, Boston University School of Public Health, Boston, MA
| | - Ramachandran S Vasan
- National Heart, Lung, and Blood Institute's and Boston University's The Framingham Heart Study, Framingham, MA.,Schools of Medicine and Public Health, Boston University, Boston, MA
| | - Christopher J O'Donnell
- National Heart, Lung, and Blood Institute's and Boston University's The Framingham Heart Study, Framingham, MA.,Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Framingham, MA
| | - Geoffrey Tofler
- Royal North Shore Hospital, Sydney, New South Wales, Australia.,University of Sydney, New South Wales, Australia
| | - Andrew D Johnson
- National Heart, Lung, and Blood Institute's and Boston University's The Framingham Heart Study, Framingham, MA .,Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Framingham, MA
| |
Collapse
|
11
|
Wingrove JA, Fitch K, Rhees B, Rosenberg S, Voora D. Peripheral blood gene expression signatures which reflect smoking and aspirin exposure are associated with cardiovascular events. BMC Med Genomics 2018; 11:1. [PMID: 29329538 PMCID: PMC5767057 DOI: 10.1186/s12920-017-0318-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 12/21/2017] [Indexed: 11/10/2022] Open
Abstract
Background Cardiovascular disease and its sequelae are major causes of global mortality, and better methods are needed to identify patients at risk for future cardiovascular events. Gene expression analysis can inform on the molecular underpinnings of risk factors for cardiovascular events. Smoking and aspirin have known opposing effects on platelet reactivity and MACE, however their effects on each other and on MACE are not well described. Methods We measured peripheral blood gene expression levels of ITGA2B, which is upregulated by aspirin and correlates with platelet reactivity on aspirin, and a 5 gene validated smoking gene expression score (sGES) where higher expression correlates with smoking status, in participants from the previously reported PREDICT trial (NCT 00500617). The primary outcome was a composite of death, myocardial infarction, and stroke/TIA (MACE). We tested whether selected genes were associated with MACE risk using logistic regression. Results Gene expression levels were determined in 1581 subjects (50.5% female, mean age 60.66 +/−11.46, 18% self-reported smokers); 3.5% of subjects experienced MACE over 12 months follow-up. Elevated sGES and ITGA2B expression were each associated with MACE (odds ratios [OR] =1.16 [95% CI 1.10–1.31] and 1.42 [95% CI 1.00–1.97], respectively; p < 0.05). ITGA2B expression was inversely associated with self-reported smoking status and the sGES (p < 0.001). A logistic regression model combining sGES and ITGA2B showed better performance (AIC = 474.9) in classifying MACE subjects than either alone (AIC = 479.1, 478.2 respectively). Conclusion Gene expression levels associated with smoking and aspirin are independently predictive of an increased risk of cardiovascular events. Electronic supplementary material The online version of this article (10.1186/s12920-017-0318-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | - Karen Fitch
- CardioDx, Inc, 600 Saginaw Dr., Redwood City, CA, 94063, USA
| | - Brian Rhees
- CardioDx, Inc, 600 Saginaw Dr., Redwood City, CA, 94063, USA
| | | | - Deepak Voora
- Center for Applied Genomics & Precision Medicine, Department of Medicine, Duke University, 101 Science Drive, 2187 CIEMAS, Durham, NC, 27708, UK.
| |
Collapse
|
12
|
Chen MH, Yanek LR, Backman JD, Eicher JD, Huffman JE, Ben-Shlomo Y, Beswick AD, Yerges-Armstrong LM, Shuldiner AR, O'Connell JR, Mathias RA, Becker DM, Becker LC, Lewis JP, Johnson AD, Faraday N. Exome-chip meta-analysis identifies association between variation in ANKRD26 and platelet aggregation. Platelets 2017; 30:164-173. [PMID: 29185836 DOI: 10.1080/09537104.2017.1384538] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Previous genome-wide association studies (GWAS) have identified several variants associated with platelet function phenotypes; however, the proportion of variance explained by the identified variants is mostly small. Rare coding variants, particularly those with high potential for impact on protein structure/function, may have substantial impact on phenotype but are difficult to detect by GWAS. The main purpose of this study was to identify low frequency or rare variants associated with platelet function using genotype data from the Illumina HumanExome Bead Chip. Three family-based cohorts of European ancestry, including ~4,000 total subjects, comprised the discovery cohort and two independent cohorts, one of European and one of African American ancestry, were used for replication. Optical aggregometry in platelet-rich plasma was performed in all the discovery cohorts in response to adenosine diphosphate (ADP), epinephrine, and collagen. Meta-analyses were performed using both gene-based and single nucleotide variant association methods. The gene-based meta-analysis identified a significant association (P = 7.13 × 10-7) between rare genetic variants in ANKRD26 and ADP-induced platelet aggregation. One of the ANKRD26 SNVs - rs191015656, encoding a threonine to isoleucine substitution predicted to alter protein structure/function, was replicated in Europeans. Aggregation increases of ~20-50% were observed in heterozygotes in all cohorts. Novel genetic signals in ABCG1 and HCP5 were also associated with platelet aggregation to ADP in meta-analyses, although only results for HCP5 could be replicated. The SNV in HCP5 intersects epigenetic signatures in CD41+ megakaryocytes suggesting a new functional role in platelet biology for HCP5. This is the first study to use gene-based association methods from SNV array genotypes to identify rare variants related to platelet function. The molecular mechanisms and pathophysiological relevance for the identified genetic associations requires further study.
Collapse
Affiliation(s)
- Ming-Huei Chen
- a National Heart, Lung and Blood Institute's The Framingham Heart Study, Population Sciences Branch, Division of Intramural Research , National Heart, Lung and Blood Institute , Framingham , MA , USA
| | - Lisa R Yanek
- b GeneSTAR Research Program, Department of Medicine, Division of General Internal Medicine , Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Joshua D Backman
- c School of Medicine, Division of Endocrinology, Diabetes and Nutrition, and Program for Personalized and Genomic Medicine , University of Maryland School of Medicine , Baltimore , MD , USA
| | - John D Eicher
- a National Heart, Lung and Blood Institute's The Framingham Heart Study, Population Sciences Branch, Division of Intramural Research , National Heart, Lung and Blood Institute , Framingham , MA , USA
| | - Jennifer E Huffman
- a National Heart, Lung and Blood Institute's The Framingham Heart Study, Population Sciences Branch, Division of Intramural Research , National Heart, Lung and Blood Institute , Framingham , MA , USA
| | - Yoav Ben-Shlomo
- d School of Social and Community Medicine , University of Bristol , Bristol , UK
| | - Andrew D Beswick
- e School of Clinical Sciences , University of Bristol , Bristol , UK
| | - Laura M Yerges-Armstrong
- c School of Medicine, Division of Endocrinology, Diabetes and Nutrition, and Program for Personalized and Genomic Medicine , University of Maryland School of Medicine , Baltimore , MD , USA
| | - Alan R Shuldiner
- c School of Medicine, Division of Endocrinology, Diabetes and Nutrition, and Program for Personalized and Genomic Medicine , University of Maryland School of Medicine , Baltimore , MD , USA
| | - Jeffrey R O'Connell
- c School of Medicine, Division of Endocrinology, Diabetes and Nutrition, and Program for Personalized and Genomic Medicine , University of Maryland School of Medicine , Baltimore , MD , USA
| | - Rasika A Mathias
- f GeneSTAR Research Program, Department of Medicine, Divisions of Allergy and Clinical Immunology and General Internal Medicine , Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Diane M Becker
- b GeneSTAR Research Program, Department of Medicine, Division of General Internal Medicine , Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Lewis C Becker
- g GeneSTAR Research Program, Department of Medicine, Divisions of Cardiology and General Internal Medicine , Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Joshua P Lewis
- c School of Medicine, Division of Endocrinology, Diabetes and Nutrition, and Program for Personalized and Genomic Medicine , University of Maryland School of Medicine , Baltimore , MD , USA
| | - Andrew D Johnson
- a National Heart, Lung and Blood Institute's The Framingham Heart Study, Population Sciences Branch, Division of Intramural Research , National Heart, Lung and Blood Institute , Framingham , MA , USA
| | - Nauder Faraday
- h GeneSTAR Research Program, Department of Anesthesiology & Critical Care Medicine , Johns Hopkins University School of Medicine , Baltimore , MD , USA
| |
Collapse
|
13
|
Kontos A, Lushington K, Martin J, Schwarz Q, Green R, Wabnitz D, Xu X, M Sokoya E, Willoughby S, Baumert M, Ferrante A, La Forgia M, Kennedy D. Relationship between Vascular Resistance and Sympathetic Nerve Fiber Density in Arterial Vessels in Children With Sleep Disordered Breathing. J Am Heart Assoc 2017; 6:JAHA.117.006137. [PMID: 28716800 PMCID: PMC5586314 DOI: 10.1161/jaha.117.006137] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Background Sleep disordered breathing in children is associated with increased blood flow velocity and sympathetic overactivity. Sympathetic overactivity results in peripheral vasoconstriction and reduced systemic vascular compliance, which increases blood flow velocity during systole. Augmented blood flow velocity is recognized to promote vascular remodeling. Importantly, increased vascular sympathetic nerve fiber density and innervation in early life plays a key role in the development of early‐onset hypertension in animal models. Examination of sympathetic nerve fiber density of the tonsillar arteries in children undergoing adenotonsillectomy for Sleep disordered breathing will address this question in humans. Methods and Results Thirteen children scheduled for adenotonsillectomy to treat sleep disordered breathing underwent pupillometry, polysomnography, flow‐mediated dilation, resting brachial artery blood flow velocity (velocity time integral), and platelet aggregation. The dorsal lingual artery (tonsil) was stained and immunofluorescence techniques used to determine sympathetic nerve fiber density. Sympathetic nerve fiber density was correlated with increased resting velocity time integral (r=0.63; P<0.05) and a lower Neuronal Pupillary Index (r=−0.71, P<0.01), as well as a slower mean pupillary constriction velocity (mean, r=−0.64; P<0.05). A faster resting velocity time integral was associated with a slower peak pupillary constriction velocity (r=−0.77; P<0.01) and higher platelet aggregation to collagen antigen (r=0.64; P<0.05). Slower mean and peak pupillary constriction velocity were associated with higher platelet aggregation scores (P<0.05; P<0.01, respectively). Conclusions These results indicate that sympathetic activity is associated with change in both the function and structure of systemic vasculature in children with sleep disordered breathing.
Collapse
Affiliation(s)
- Anna Kontos
- Robinson's Research Institute, School of Medicine, Discipline of Paediatrics and Reproductive Health, University of Adelaide, Adelaide, Australia
| | - Kurt Lushington
- School of Psychology, Social Work and Social Policy, University of South Australia, Adelaide, Australia
| | - James Martin
- Robinson's Research Institute, School of Medicine, Discipline of Paediatrics and Reproductive Health, University of Adelaide, Adelaide, Australia.,Department of Respiratory and Sleep Medicine, Women's and Children's Hospital, Adelaide, Australia
| | - Quenten Schwarz
- Neurovascular Research Laboratory, Centre for Cancer Biology, University of South Australia, Adelaide, Australia
| | - Ryan Green
- Department of Information Technology, Engineering and the Environment, University of South Australia, Adelaide, Australia
| | - David Wabnitz
- Department of Otolaryngology-Head and Neck Surgery, Women's and Children's Hospital, Adelaide, Australia
| | - Xiangjun Xu
- Neurovascular Research Laboratory, Centre for Cancer Biology, University of South Australia, Adelaide, Australia
| | - Elke M Sokoya
- Department of Human Physiology, Flinders University, Adelaide, South Australia, Australia
| | - Scott Willoughby
- Centre for Heart Rhythm Disorders, South Australian Health and Medical Research Institute, University of Adelaide and Royal Adelaide Hospital, Adelaide, Australia
| | - Mathias Baumert
- School of Electrical and Electronic Engineering, University of Adelaide, Adelaide, Australia
| | - Antonio Ferrante
- Robinson's Research Institute, School of Medicine, Discipline of Paediatrics and Reproductive Health, University of Adelaide, Adelaide, Australia.,Department of Immunology SA Pathology, Schools of Medicine and Biological Science, University of Adelaide, Adelaide, Australia
| | - Melissa La Forgia
- Department of Medical Imaging, Women's and Children's Hospital, Adelaide, Australia
| | - Declan Kennedy
- Robinson's Research Institute, School of Medicine, Discipline of Paediatrics and Reproductive Health, University of Adelaide, Adelaide, Australia.,Department of Respiratory and Sleep Medicine, Women's and Children's Hospital, Adelaide, Australia
| |
Collapse
|
14
|
Gurbel PA, Jeong YH, Navarese EP, Tantry US. Platelet-Mediated Thrombosis. Circ Res 2016; 118:1380-91. [DOI: 10.1161/circresaha.115.307016] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 02/17/2016] [Indexed: 11/16/2022]
Abstract
The pivotal role that platelets play in thrombosis and resultant ischemic event occurrences in patients with high-risk coronary artery disease is well established. This role provides the fundamental basis for the current wide implementation of dual antiplatelet therapy with aspirin and a P2Y
12
receptor inhibitor. The development of user friendly point-of-care methods to assess platelet reactivity to adenosine diphosphate has increased the frequency of platelet function testing in clinical practice. Recent large observational studies have established an independent relation between the results of point-of-care platelet function testing and clinical event occurrence in patients undergoing coronary artery stenting. However, prospective, randomized trials have failed to demonstrate that personalized antiplatelet therapy based on point-of-care assessment of platelet function is effective in reducing ischemic event occurrences. Important limitations were associated with these trials. In addition, the concept of a therapeutic window of P2Y
12
receptor reactivity with an upper threshold associated with ischemic event occurrence and a lower threshold associated with bleeding has also been proposed. In the absence of strong prospective evidence to support personalized antiplatelet therapy, clinical decision making about antiplatelet therapy rests on the large body of observational data and the fundamental importance of platelet physiology in catastrophic event occurrence in patients with high-risk coronary artery disease.
Collapse
Affiliation(s)
- Paul A. Gurbel
- From the Inova Center for Thrombosis Research and Drug Development, Inova Heart and Vascular Institute, Fairfax, VA (P.A.G., E.P.N., U.S.T.); and Clinical Trial Center, Gyeongsang National University Hospital, Gyeongsangnam-do, Korea (Y.-H.J.)
| | - Young-Hoon Jeong
- From the Inova Center for Thrombosis Research and Drug Development, Inova Heart and Vascular Institute, Fairfax, VA (P.A.G., E.P.N., U.S.T.); and Clinical Trial Center, Gyeongsang National University Hospital, Gyeongsangnam-do, Korea (Y.-H.J.)
| | - Eliano P. Navarese
- From the Inova Center for Thrombosis Research and Drug Development, Inova Heart and Vascular Institute, Fairfax, VA (P.A.G., E.P.N., U.S.T.); and Clinical Trial Center, Gyeongsang National University Hospital, Gyeongsangnam-do, Korea (Y.-H.J.)
| | - Udaya S. Tantry
- From the Inova Center for Thrombosis Research and Drug Development, Inova Heart and Vascular Institute, Fairfax, VA (P.A.G., E.P.N., U.S.T.); and Clinical Trial Center, Gyeongsang National University Hospital, Gyeongsangnam-do, Korea (Y.-H.J.)
| |
Collapse
|