1
|
Liani R, Simeone PG, Tripaldi R, D'Ardes D, Creato V, Pepe R, Lessiani G, Bologna G, Cipollone F, Marchisio M, Lanuti P, Santilli F. Kinetics of Circulating Extracellular Vesicles Over the 24-Hour Dosing Interval After Low-Dose Aspirin Administration in Patients at Cardiovascular Risk. Clin Pharmacol Ther 2023; 113:1096-1106. [PMID: 36749026 DOI: 10.1002/cpt.2865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 02/01/2023] [Indexed: 02/08/2023]
Abstract
Extracellular vesicles (EVs) are small vesicles deriving from all cell types during cell activation, involved in transcellular communication, and regarded as predictors of vascular damage and of cardiovascular events. We tested the hypothesis that, in patients on chronic low-dose aspirin treatment for cardiovascular prevention, aspirin may affect the release of EVs within the 24-hour interval. We enrolled 84 patients, mostly at high or very high cardiovascular risk, on chronic low-dose aspirin treatment. The numbers of circulating EVs (cEVs) and annexinV+ cEVs (total, platelet-derived, endothelial-derived, and leucocyte-derived) were assessed immediately before, and after 10 and 24 hours of a witnessed aspirin administration. Platelet cyclooxygenase 1 (COX-1) recovery was characterized by measuring serum thromboxane B2 (sTXB2 ) at the same timepoints. Nine healthy participants were also enrolled. In patients, daily aspirin administration acutely inhibited after 10 hours following aspirin administrations the release of cEVs (total and leukocyte-derived) and annexinV+ cEVs (total, platelet-derived, endothelial-derived, and leukocyte-derived), with a rapid recovery at 24 hours. The inhibition after 10 hours suggests a COX-1-dependent mechanism. Interestingly, the slope of platelet-derived and of annexinV+ platelet-derived cEVs were both directly related to sTXB2 slope and COX-1 messenger RNA, raising the hypothesis that vice versa, cEVs may affect the rate of COX-1 recovery and the subsequent duration of aspirin effect. In healthy participants, no circadian difference was observed, except for leukocyte-derived cEVs. Our findings suggest a previously unappreciated effect of aspirin on the kinetics of a subset of cEVs possibly contributing to the cardioprotective effects of this drug.
Collapse
Affiliation(s)
- Rossella Liani
- Department of Medicine and Aging Sciences, Center for Advanced Studies and Technology, University of Chieti, Chieti, Italy
| | - Paola Giustina Simeone
- Department of Medicine and Aging Sciences, Center for Advanced Studies and Technology, University of Chieti, Chieti, Italy
| | - Romina Tripaldi
- Department of Medicine and Aging Sciences, Center for Advanced Studies and Technology, University of Chieti, Chieti, Italy
| | - Damiano D'Ardes
- Department of Medicine and Aging Sciences, Center for Advanced Studies and Technology, University of Chieti, Chieti, Italy
| | - Valeria Creato
- Internal Medicine, Clinica Medica, SS. Annunziata Hospital, Chieti, Italy
| | - Raffaele Pepe
- Internal Medicine, Clinica Medica, SS. Annunziata Hospital, Chieti, Italy
| | | | - Giuseppina Bologna
- Department of Medicine and Aging Sciences, Center for Advanced Studies and Technology, University of Chieti, Chieti, Italy
| | - Francesco Cipollone
- Department of Medicine and Aging Sciences, Center for Advanced Studies and Technology, University of Chieti, Chieti, Italy
| | - Marco Marchisio
- Department of Medicine and Aging Sciences, Center for Advanced Studies and Technology, University of Chieti, Chieti, Italy
| | - Paola Lanuti
- Department of Medicine and Aging Sciences, Center for Advanced Studies and Technology, University of Chieti, Chieti, Italy
| | - Francesca Santilli
- Department of Medicine and Aging Sciences, Center for Advanced Studies and Technology, University of Chieti, Chieti, Italy
| |
Collapse
|
2
|
Abstract
In addition to the key role in hemostasis and thrombosis, platelets have also been wildly acknowledged as immune regulatory cells and involving in the pathogenesis of inflammation-related diseases. Since purine receptor P2Y12 plays a crucial role in platelet activation, P2Y12 antagonists such as clopidogrel, prasugrel, and ticagrelor have been widely used in cardiovascular diseases worldwide in recent decades due to their potent antiplatelet and antithrombotic effects. Meanwhile, the role of P2Y12 in inflammatory diseases has also been extensively studied. Relatively, there are few studies on the regulation of P2Y12. This review first summarizes the various roles of P2Y12 in the process of platelet activation, as well as downstream effects and signaling pathways; then introduces the effects of P2Y12 in inflammatory diseases such as sepsis, atherosclerosis, cancer, autoimmune diseases, and asthma; and finally reviews the current researches on P2Y12 regulation.
Collapse
Affiliation(s)
- Xiaohua Li
- Department of Infectious Diseases, The First Hospital of Jilin University, Changchun, 130021, Jilin, China
- Department of Pharmacology, School of Pharmacy, Jilin University, Fujin Road, Changchun, 130021, Jilin, China
| | | | - Xia Cao
- Department of Pharmacology, School of Pharmacy, Jilin University, Fujin Road, Changchun, 130021, Jilin, China.
| |
Collapse
|
3
|
Carotti V, Rigalli JP, van Asbeck-van der Wijst J, G J Hoenderop J. Interplay between purinergic signalling and extracellular vesicles in health and disease. Biochem Pharmacol 2022; 203:115192. [PMID: 35905971 DOI: 10.1016/j.bcp.2022.115192] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 07/21/2022] [Accepted: 07/22/2022] [Indexed: 11/25/2022]
Abstract
Purinergic signalling is a receptor-mediated process characterized by the binding of extracellular nucleotides and nucleosides to purinergic receptors, which results in the activation intracellular signalling pathways, and, ultimately, leads to changes in cell physiology. Purinergic signalling has been related to the regulation of important physiological processes (e.g., renal electrolyte reabsorption; platelet aggregation; immune response). In addition, it has been associated with pathophysiological situations such as cancer and inflammation. Extracellular vesicles (EVs) are nanoparticles released by all cells of the organism, which play a key role in cell-cell communication. In this regard, EVs can mediate effects on target cells located at distant locations. Within their cargo, EVs contain molecules with the potential to affect purinergic signalling at the target cells and tissues. Here, we review the studies addressing the regulation of purinergic signalling by EVs based on the cell type or tissue where the regulation takes place. In this regard, EVs are found to play a major role in modulating the extracellular ATP levels and, specially, adenosine. This has a clear impact on, for instance, the inflammatory and immune response against cancer cells. Furthermore, we discuss the data available on the regulation of EV secretion and its cargo by purinergic signalling. Here, a major role of the purinergic receptor P2X7 and again, an impact on processes such as inflammation, immune response and cancer pathogenesis has been established. Finally, we highlight uninvestigated aspects of these two regulatory networks and address their potential as therapeutic targets.
Collapse
Affiliation(s)
- Valentina Carotti
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, the Netherlands
| | - Juan P Rigalli
- Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Jenny van Asbeck-van der Wijst
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, the Netherlands
| | - Joost G J Hoenderop
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, the Netherlands.
| |
Collapse
|
4
|
Different Contribution of Monocyte- and Platelet-Derived Microvesicles to Endothelial Behavior. Int J Mol Sci 2022; 23:ijms23094811. [PMID: 35563201 PMCID: PMC9105732 DOI: 10.3390/ijms23094811] [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: 04/06/2022] [Revised: 04/22/2022] [Accepted: 04/24/2022] [Indexed: 11/17/2022] Open
Abstract
Several contributions of circulating microvesicles (MVs) to the endothelial dysfunction have been reported in the past; a head-to-head comparison of platelet- and monocyte–derived MVs has however never been performed. To this aim, we assessed the involvement of these MVs in vessel damage related processes, i.e., oxidative stress, inflammation, and leukocyte-endothelial adhesion. Platelets and monocytes isolated from healthy subjects (HS, n = 15) were stimulated with TRAP-6 and LPS to release MVs that were added to human vascular endothelial cell (hECV) culture to evaluate superoxide anion production, inflammatory markers (IL-6, TNFα, NF-κB mRNA expression), and hECV adhesiveness. The effects of the MVs-induced from HS were compared to those induced by MVs spontaneously released from cells of patients with ST-segment elevation myocardial infarction (STEMI, n = 7). MVs released by HS-activated cells triggered a threefold increase in oxidative burst in a concentration-dependent manner. Only MVs released from monocytes doubled IL-6, TNFα, and NF-κB mRNA expression and monocyte-endothelial adhesion. Interestingly, the effects of the MVs isolated from STEMI-monocytes were not superimposable to previous ones except for adhesion to hECV. Conversely, MVs released from STEMI-platelets sustained both redox state and inflammatory phenotype. These data provide evidence that MVs released from activated and/or pathologic platelets and monocytes differently affect endothelial behavior, highlighting platelet-MVs as causative factors of impaired endothelial function in the acute phase of STEMI.
Collapse
|
5
|
Mak A, Chan JKY. Endothelial function and endothelial progenitor cells in systemic lupus erythematosus. Nat Rev Rheumatol 2022; 18:286-300. [PMID: 35393604 DOI: 10.1038/s41584-022-00770-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/04/2022] [Indexed: 12/13/2022]
Abstract
The observations that traditional cardiovascular disease (CVD) risk factors fail to fully account for the excessive cardiovascular mortality in patients with systemic lupus erythematosus (SLE) compared with the general population have prompted in-depth investigations of non-traditional, SLE-related risk factors that contribute to cardiovascular complications in patients with SLE. Of the various perturbations of vascular physiology, endothelial dysfunction, which is believed to occur in the earliest step of atherosclerosis, has been extensively investigated for its contribution to CVD risk in SLE. Endothelial progenitor cells (EPCs), which play a crucial part in vascular repair, neovascularization and maintenance of endothelial function, are quantitatively and functionally reduced in patients with SLE. Yet, the lack of a unified definition of EPCs, standardization of the quantity and functional assessment of EPCs as well as endothelial function measurement pose challenges to the translation of endothelial function measurements and EPC levels into prognostic markers for CVD in patients with SLE. This Review discusses factors that contribute to CVD in SLE, with particular focus on how endothelial function and EPCs are evaluated currently, and how EPCs are quantitatively and functionally altered in patients with SLE. Potential strategies for the use of endothelial function measurements and EPC quantification as prognostic markers of CVD in patients with SLE, and the limitations of their prognostication potential, are also discussed.
Collapse
Affiliation(s)
- Anselm Mak
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore. .,Division of Rheumatology, University Medicine Cluster, National University Health System, Singapore, Singapore.
| | - Jerry Kok Yen Chan
- Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore, Singapore.,Academic Clinical Programme in Obstetrics and Gynaecology, Duke-NUS Medical School, Singapore, Singapore.,Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
6
|
Ghaffari F, Rasmi Y, Seyed Mohammadzad MH, Seyedi S, Shirpoor A, Roshani-Asl E, Saboory E. Increased circulating platelet and endothelial-derived microparticles in patients with cardiac syndrome X. ARYA ATHEROSCLEROSIS 2021; 17:1-10. [PMID: 34703482 PMCID: PMC8519618 DOI: 10.22122/arya.v17i0.2094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 06/02/2020] [Indexed: 11/09/2022]
Abstract
BACKGROUND Cardiac syndrome X (CSX) has been associated with endothelial dysfunction and inflammation. We conducted a case-control study to evaluate the association between plateletý and endothelial-derived microparticles (PMPs and EMPs), as specific quantitative plasma markers of endothelial dysfunction, and the presence of CSX. METHODS The present study was conducted on 40 CSX patients and 19 healthy individuals. C-reactive protein (CRP), and hematological and biochemical parameters were evaluated. The MP concentration in platelet-poor plasma (PPP) was quantitatively determined through flow cytometry using specific anti-human CD31, CD41a, CD62E, and CD144 antibodies. RESULTS The mean platelet volume (MPV) and positive CRP rate (≥ 3.8 mg/l) were higher in patients compared to controls (P = 0.020 and P = 0.010, respectively). The CD62E+, CD144+, and CD31+41− EMPs, as well as CD41+ and CD31+CD41+ PMPs showed significant increase in CSX patients compared to controls (P < 0.050). There were direct correlations between the mean percentage of detected EMPs and PMPs as well as between their expression intensity; however, a reverse correlation was seen between the percentage of MPs and CD144 and CD41. Moreover, the MP level was reversely associated with prothrombin time (PT) and partial thromboplastin time (PTT) values. Only CD31+CD41+ PMP was correlated with CRP. CONCLUSION It seems that EMPs and PMPs increase in CSX, which may contribute to various processes involved in the development of this syndrome.
Collapse
Affiliation(s)
- Fereshteh Ghaffari
- Department of Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Yousef Rasmi
- Professor, Cellular and Molecular Research Center AND Department of Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Mir Hossein Seyed Mohammadzad
- Associate Professor, Department of Cardiology, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Shahram Seyedi
- Assistant Professor, Department of Immunology, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Alireza Shirpoor
- Professor, Department of Physiology, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Elmira Roshani-Asl
- Department of Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Ehsan Saboory
- Professor, Zanjan Metabolic Diseases Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
7
|
Zifkos K, Dubois C, Schäfer K. Extracellular Vesicles and Thrombosis: Update on the Clinical and Experimental Evidence. Int J Mol Sci 2021; 22:ijms22179317. [PMID: 34502228 PMCID: PMC8431093 DOI: 10.3390/ijms22179317] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 08/24/2021] [Accepted: 08/25/2021] [Indexed: 12/16/2022] Open
Abstract
Extracellular vesicles (EVs) compose a heterogenous group of membrane-derived particles, including exosomes, microvesicles and apoptotic bodies, which are released into the extracellular environment in response to proinflammatory or proapoptotic stimuli. From earlier studies suggesting that EV shedding constitutes a cellular clearance mechanism, it has become evident that EV formation, secretion and uptake represent important mechanisms of intercellular communication and exchange of a wide variety of molecules, with relevance in both physiological and pathological situations. The putative role of EVs in hemostasis and thrombosis is supported by clinical and experimental studies unraveling how these cell-derived structures affect clot formation (and resolution). From those studies, it has become clear that the prothrombotic effects of EVs are not restricted to the exposure of tissue factor (TF) and phosphatidylserines (PS), but also involve multiplication of procoagulant surfaces, cross-linking of different cellular players at the site of injury and transfer of activation signals to other cell types. Here, we summarize the existing and novel clinical and experimental evidence on the role and function of EVs during arterial and venous thrombus formation and how they may be used as biomarkers as well as therapeutic vectors.
Collapse
Affiliation(s)
- Konstantinos Zifkos
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, D-55131 Mainz, Germany;
| | - Christophe Dubois
- Aix Marseille University, INSERM 1263, Institut National de la Recherche pour l’Agriculture, l’alimentation et l’Environnement (INRAE) 1260, Center for CardioVascular and Nutrition Research (C2VN), F-13380 Marseille, France;
| | - Katrin Schäfer
- Department of Cardiology, Cardiology I, University Medical Center Mainz, D-55131 Mainz, Germany
- Correspondence:
| |
Collapse
|
8
|
Lugo-Gavidia LM, Burger D, Matthews VB, Nolde JM, Galindo Kiuchi M, Carnagarin R, Kannenkeril D, Chan J, Joyson A, Herat LY, Azzam O, Schlaich MP. Role of Microparticles in Cardiovascular Disease: Implications for Endothelial Dysfunction, Thrombosis, and Inflammation. HYPERTENSION (DALLAS, TEX. : 1979) 2021; 77:1825-1844. [PMID: 33979187 DOI: 10.1161/hypertensionaha.121.16975] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Leslie Marisol Lugo-Gavidia
- Dobney Hypertension Centre, School of Medicine, Royal Perth Hospital Unit, Medical Research Foundation, Faculty of Medicine, Dentistry and Health Sciences, The University of Western Australia (L.M.L.-G., V.B.M., J.M.N., M.G.K., R.C., D.K., J.C., A.J., L.Y.H., O.A., M.P.S.)
| | - Dylan Burger
- Kidney Research Centre, The Ottawa Hospital Research Institute, Department of Cellular and Molecular Medicine, University of Ottawa (D.B.)
| | - Vance B Matthews
- Dobney Hypertension Centre, School of Medicine, Royal Perth Hospital Unit, Medical Research Foundation, Faculty of Medicine, Dentistry and Health Sciences, The University of Western Australia (L.M.L.-G., V.B.M., J.M.N., M.G.K., R.C., D.K., J.C., A.J., L.Y.H., O.A., M.P.S.)
| | - Janis M Nolde
- Dobney Hypertension Centre, School of Medicine, Royal Perth Hospital Unit, Medical Research Foundation, Faculty of Medicine, Dentistry and Health Sciences, The University of Western Australia (L.M.L.-G., V.B.M., J.M.N., M.G.K., R.C., D.K., J.C., A.J., L.Y.H., O.A., M.P.S.)
| | - Márcio Galindo Kiuchi
- Dobney Hypertension Centre, School of Medicine, Royal Perth Hospital Unit, Medical Research Foundation, Faculty of Medicine, Dentistry and Health Sciences, The University of Western Australia (L.M.L.-G., V.B.M., J.M.N., M.G.K., R.C., D.K., J.C., A.J., L.Y.H., O.A., M.P.S.)
| | - Revathy Carnagarin
- Dobney Hypertension Centre, School of Medicine, Royal Perth Hospital Unit, Medical Research Foundation, Faculty of Medicine, Dentistry and Health Sciences, The University of Western Australia (L.M.L.-G., V.B.M., J.M.N., M.G.K., R.C., D.K., J.C., A.J., L.Y.H., O.A., M.P.S.)
| | - Dennis Kannenkeril
- Dobney Hypertension Centre, School of Medicine, Royal Perth Hospital Unit, Medical Research Foundation, Faculty of Medicine, Dentistry and Health Sciences, The University of Western Australia (L.M.L.-G., V.B.M., J.M.N., M.G.K., R.C., D.K., J.C., A.J., L.Y.H., O.A., M.P.S.).,Department of Nephrology and Hypertension, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Germany (D.K.)
| | - Justine Chan
- Dobney Hypertension Centre, School of Medicine, Royal Perth Hospital Unit, Medical Research Foundation, Faculty of Medicine, Dentistry and Health Sciences, The University of Western Australia (L.M.L.-G., V.B.M., J.M.N., M.G.K., R.C., D.K., J.C., A.J., L.Y.H., O.A., M.P.S.)
| | - Anu Joyson
- Dobney Hypertension Centre, School of Medicine, Royal Perth Hospital Unit, Medical Research Foundation, Faculty of Medicine, Dentistry and Health Sciences, The University of Western Australia (L.M.L.-G., V.B.M., J.M.N., M.G.K., R.C., D.K., J.C., A.J., L.Y.H., O.A., M.P.S.)
| | - Lakshini Y Herat
- Dobney Hypertension Centre, School of Medicine, Royal Perth Hospital Unit, Medical Research Foundation, Faculty of Medicine, Dentistry and Health Sciences, The University of Western Australia (L.M.L.-G., V.B.M., J.M.N., M.G.K., R.C., D.K., J.C., A.J., L.Y.H., O.A., M.P.S.)
| | - Omar Azzam
- Dobney Hypertension Centre, School of Medicine, Royal Perth Hospital Unit, Medical Research Foundation, Faculty of Medicine, Dentistry and Health Sciences, The University of Western Australia (L.M.L.-G., V.B.M., J.M.N., M.G.K., R.C., D.K., J.C., A.J., L.Y.H., O.A., M.P.S.).,Department of Internal Medicine (O.A.), Royal Perth Hospital, Western Australia
| | - Markus P Schlaich
- Dobney Hypertension Centre, School of Medicine, Royal Perth Hospital Unit, Medical Research Foundation, Faculty of Medicine, Dentistry and Health Sciences, The University of Western Australia (L.M.L.-G., V.B.M., J.M.N., M.G.K., R.C., D.K., J.C., A.J., L.Y.H., O.A., M.P.S.).,Departments of Cardiology and Nephrology (M.P.S.), Royal Perth Hospital, Western Australia.,Neurovascular Hypertension and Kidney Disease Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia (M.P.S.)
| |
Collapse
|
9
|
CD36+/CD61+ Microparticles Correlate with the Risk of Percutaneous Cardiac Interventions in Coronary Artery Disease Patients and the Effects of Ticagrelor. Cardiovasc Drugs Ther 2021; 36:455-465. [PMID: 33893936 DOI: 10.1007/s10557-021-07184-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/05/2021] [Indexed: 10/21/2022]
Abstract
PURPOSE The CD36 scavenger receptor is a mediator of both atherogenesis and thrombosis. We aimed to investigate the prognostic value of CD36+ microparticles (MPs) released from platelets for cardiovascular event presentation in coronary artery disease (CAD) patients and the effects of different antiplatelet drugs on MPs. METHODS A total of 101 aspirin-treated CAD patients, who were planned to undergo coronary angiography (CAG), were randomized to either a standard clopidogrel regimen or ticagrelor treatment. Total Annexin V-(AV)+ MPs, CD61+/AV+ MPs, and CD36+/CD61+/AV+ MPs were quantified by flow cytometry at baseline, before and immediately after the operation. The ADP-induced platelet inhibition rate was measured by thromboelastogram (TEG) examination 1 h before the operation. RESULTS The baseline levels of CD36+/CD61+/AV+ MPs were significantly increased in percutaneous coronary intervention (PCI) patients (n = 52) compared to no-PCI patients (n = 49) (p < 0.05). A ROC-curve clustered model for CD36+/CD61+/AV+ MPs at baseline predicted an increased risk of PCI [p = 0.009, AUC = 0.761 (95%CI: 0.601 to 0.922)]. Moreover, TEG examination showed that the preoperative proportion of CD36+/CD61+/AV+ MPs was significantly negatively correlated with R time and K time (r = - 0.236, p = 00.026; r = - 0.288, p = 0.006), and positively correlated with MAADP (r = 0.226, p = 0.045). Subgroup analysis of PCI group showed that the platelet inhibition rate of ticagrelor was significantly higher (66.05% ± 28.76% vs.31.01% ± 27.33%, p < 0.001), and the number of AV+ MPs, CD61+/AV+ MPs, and CD36+/CD61+/AV+ MPs before the operation was significantly lower than clopidogrel (p < 0.05, all). CONCLUSION The high levels of CD36+ MPs derived from activated platelets are related to an increased risk of PCI in CAD patients. Ticagrelor significantly reduced the number of CD61+/AV+ MPs and CD36+/CD61+/AV+ MPs. This trial registration number is ChiCTR1800014908 and the date of registration is 2018.05.01.
Collapse
|
10
|
Pelliccia F, Pasceri V, Moretti A, Tanzilli G, Speciale G, Gaudio C. Endothelial progenitor cells predict long-term outcome in patients with coronary artery disease: Ten-year follow-up of the PROCREATION extended study. Int J Cardiol 2020; 318:123-125. [DOI: 10.1016/j.ijcard.2020.06.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 05/13/2020] [Accepted: 06/01/2020] [Indexed: 01/26/2023]
|
11
|
Masuyama T, Sakuma M, Waku R, Hirose S, Kitahara K, Naganuma J, Yazawa H, Toyoda S, Abe S, Nakajima T, Inoue T. Effects of switching from clopidogrel to prasugrel at the chronic phase after coronary stenting on antiplatelet action and vascular endothelial function: Switch-Pras study. Heart Vessels 2020; 36:442-451. [PMID: 33113567 PMCID: PMC7940291 DOI: 10.1007/s00380-020-01714-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 10/09/2020] [Indexed: 01/12/2023]
Abstract
Compared to clopidogrel, prasugrel has a lower incidence of ischemic events following percutaneous coronary intervention (PCI) because of an early reduction during the acute phase in P2Y12 reaction units (PRU). The objective of this study was to compare the antiplatelet effect and vascular endothelial function of both drugs during the chronic phase after PCI. Patients who had undergone PCI and were confirmed to have no restenosis by follow-up coronary angiography under dual anti-platelet therapy with clopidogrel (75 mg/day) and aspirin (100 mg/day) were randomized to either continue clopidogrel or switch to prasugrel (3.75 mg/day). At baseline, prior to randomization we determined the CYP2C19 genotype. At the baseline and 24 weeks after randomization, the P2Y12 reactivity unit (PRU) was measured using the VerifyNow™ P2Y12 assay. Endothelial function was evaluated by flow-mediated vasodilation (FMD) and reactive hyperemia peripheral arterial tonometry (RH-PAT), while and circulating CD34+/CD133+/CD45low progenitor cells were measured by flow cytometric analysis. Serum high-sensitivity C-reactive protein (hsCRP) level was also measured. The PRU was reduced significantly in the prasugrel group (P = 0.0008), especially in patients who were intermediate or poor metabolizers based on the CYP2C19 genotype (P < 0.0001). This reduction was not observed in the clopidogrel group. The number of CD34+/CD133+/CD45low cells increased in the clopidogrel group (P = 0.008), but not in the prasugrel group. The hsCRP, FMD and reactive hyperemia index measured by RH-PAT did not change in either group. Prasugrel is potentially better than clopidogrel for preventing thrombotic events, although clopidogrel may have an advantage over prasugrel in terms of preventing atherosclerotic events. Proper use of thienopyridine drugs based on the CYP2C19 genotype has promising clinical potential.
Collapse
Affiliation(s)
- Taiki Masuyama
- Department of Cardiovascular Medicine, School of Medicine, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| | - Masashi Sakuma
- Department of Cardiovascular Medicine, School of Medicine, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan.
| | - Ryutaro Waku
- Department of Cardiovascular Medicine, School of Medicine, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| | - Suguru Hirose
- Department of Cardiovascular Medicine, School of Medicine, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| | - Keijiro Kitahara
- Department of Cardiovascular Medicine, School of Medicine, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| | - Jin Naganuma
- Department of Cardiovascular Medicine, School of Medicine, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| | - Hiroko Yazawa
- Department of Cardiovascular Medicine, School of Medicine, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| | - Shigeru Toyoda
- Department of Cardiovascular Medicine, School of Medicine, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| | - Shichiro Abe
- Department of Cardiovascular Medicine, School of Medicine, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| | - Toshiaki Nakajima
- Department of Cardiovascular Medicine, School of Medicine, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| | - Teruo Inoue
- Department of Cardiovascular Medicine, School of Medicine, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| |
Collapse
|
12
|
Gąsecka A, Rogula S, Eyileten C, Postuła M, Jaguszewski MJ, Kochman J, Mazurek T, Nieuwland R, Filipiak KJ. Role of P2Y Receptors in Platelet Extracellular Vesicle Release. Int J Mol Sci 2020; 21:ijms21176065. [PMID: 32842470 PMCID: PMC7504123 DOI: 10.3390/ijms21176065] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 08/18/2020] [Accepted: 08/19/2020] [Indexed: 12/23/2022] Open
Abstract
Platelet extracellular vesicles (PEVs) are potential new biomarkers of platelet activation which may allow us to predict and/or diagnose developing coronary thrombosis before myocardial necrosis occurs. The P2Y1 and P2Y12 receptors play a key role in platelet activation and aggregation. Whereas the P2Y1 antagonists are at the preclinical stage, at present, the P2Y12 antagonists are the most effective treatment strategy to prevent stent thrombosis after percutaneous coronary intervention. Despite an increasing number of publications on PEVs, the mechanisms underlying their formation, including the role of purinergic receptors in this process, remain an active research field. Here, we outline the clinical relevance of PEVs in cardiovascular disease, summarize the role and downstream signalling of P2Y receptors in platelet activation, and discuss the available evidence regarding their role in PEV formation.
Collapse
Affiliation(s)
- Aleksandra Gąsecka
- 1st Chair and Department of Cardiology, Medical University of Warsaw, 02-106 Warsaw, Poland; (S.R.); (J.K.); (T.M.); (K.J.F.)
- Laboratory Experimental Clinical Chemistry, and Vesicle Observation Center, Amsterdam UMC, University of Amsterdam, 1012 WX Amsterdam, The Netherlands
- Correspondence: ; Tel.:+48-22-599-19-51
| | - Sylwester Rogula
- 1st Chair and Department of Cardiology, Medical University of Warsaw, 02-106 Warsaw, Poland; (S.R.); (J.K.); (T.M.); (K.J.F.)
| | - Ceren Eyileten
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology, Medical University of Warsaw, 02-097 Warsaw, Poland; (C.E.); (M.P.); (R.N.)
| | - Marek Postuła
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology, Medical University of Warsaw, 02-097 Warsaw, Poland; (C.E.); (M.P.); (R.N.)
| | | | - Janusz Kochman
- 1st Chair and Department of Cardiology, Medical University of Warsaw, 02-106 Warsaw, Poland; (S.R.); (J.K.); (T.M.); (K.J.F.)
| | - Tomasz Mazurek
- 1st Chair and Department of Cardiology, Medical University of Warsaw, 02-106 Warsaw, Poland; (S.R.); (J.K.); (T.M.); (K.J.F.)
| | - Rienk Nieuwland
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology, Medical University of Warsaw, 02-097 Warsaw, Poland; (C.E.); (M.P.); (R.N.)
| | - Krzysztof J. Filipiak
- 1st Chair and Department of Cardiology, Medical University of Warsaw, 02-106 Warsaw, Poland; (S.R.); (J.K.); (T.M.); (K.J.F.)
| |
Collapse
|
13
|
Sun R, Huang J, Sun B. Mobilization of endothelial progenitor cells in sepsis. Inflamm Res 2019; 69:1-9. [DOI: 10.1007/s00011-019-01299-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 11/07/2019] [Accepted: 11/08/2019] [Indexed: 12/17/2022] Open
|
14
|
Taus F, Meneguzzi A, Castelli M, Minuz P. Platelet-Derived Extracellular Vesicles as Target of Antiplatelet Agents. What Is the Evidence? Front Pharmacol 2019; 10:1256. [PMID: 31780927 PMCID: PMC6857039 DOI: 10.3389/fphar.2019.01256] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 09/30/2019] [Indexed: 12/14/2022] Open
Abstract
Platelet-derived large extracellular vesicles (often referred to as microparticles in the field of cardiovascular disease) have been identified as effector in the atherothrombotic process, therefore representing a target of pharmacological intervention of potential interest. Despite that, limited evidence is so far available concerning the effects of antiplatelet agents on the release of platelet-derived extracellular vesicles. In the present narrative review, the mechanisms leading to vesiculation in platelets and the pathophysiological processes implicated will be discussed. This will be followed by a summary of the present evidence concerning the effects of antiplatelet agents under experimental conditions and in clinical settings.
Collapse
Affiliation(s)
- Francesco Taus
- Department of Medicine, Section of Internal Medicine C, University of Verona, Verona, Italy
| | - Alessandra Meneguzzi
- Department of Medicine, Section of Internal Medicine C, University of Verona, Verona, Italy
| | - Marco Castelli
- Department of Medicine, Section of Internal Medicine C, University of Verona, Verona, Italy
| | - Pietro Minuz
- Department of Medicine, Section of Internal Medicine C, University of Verona, Verona, Italy
| |
Collapse
|
15
|
Zarà M, Guidetti GF, Camera M, Canobbio I, Amadio P, Torti M, Tremoli E, Barbieri SS. Biology and Role of Extracellular Vesicles (EVs) in the Pathogenesis of Thrombosis. Int J Mol Sci 2019; 20:ijms20112840. [PMID: 31212641 PMCID: PMC6600675 DOI: 10.3390/ijms20112840] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 06/07/2019] [Indexed: 02/06/2023] Open
Abstract
Extracellular vesicles (EVs) are well-established mediators of cell-to-cell communication. EVs can be released by every cell type and they can be classified into three major groups according to their biogenesis, dimension, density, and predominant protein markers: exosomes, microvesicles, and apoptotic bodies. During their formation, EVs associate with specific cargo from their parental cell that can include RNAs, free fatty acids, surface receptors, and proteins. The biological function of EVs is to maintain cellular and tissue homeostasis by transferring critical biological cargos to distal or neighboring recipient cells. On the other hand, their role in intercellular communication may also contribute to the pathogenesis of several diseases, including thrombosis. More recently, their physiological and biochemical properties have suggested their use as a therapeutic tool in tissue regeneration as well as a novel option for drug delivery. In this review, we will summarize the impact of EVs released from blood and vascular cells in arterial and venous thrombosis, describing the mechanisms by which EVs affect thrombosis and their potential clinical applications.
Collapse
Affiliation(s)
- Marta Zarà
- Unit of Heart-Brain Axis: Cellular and Molecular Mechanisms, Centro Cardiologico Monzino IRCCS, 20138 Milano, Italy.
| | | | - Marina Camera
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milano, Italy.
- Unit of Cell and Molecular Biology in Cardiovascular Diseases, Centro Cardiologico Monzino IRCCS, 20138 Milano, Italy.
| | - Ilaria Canobbio
- Department of Biology and Biotechnology, University of Pavia, 27100 Pavia, Italy.
| | - Patrizia Amadio
- Unit of Heart-Brain Axis: Cellular and Molecular Mechanisms, Centro Cardiologico Monzino IRCCS, 20138 Milano, Italy.
| | - Mauro Torti
- Department of Biology and Biotechnology, University of Pavia, 27100 Pavia, Italy.
| | - Elena Tremoli
- Scientific Direction, Centro Cardiologico Monzino IRCCS, 20138 Milano, Italy.
| | - Silvia Stella Barbieri
- Unit of Heart-Brain Axis: Cellular and Molecular Mechanisms, Centro Cardiologico Monzino IRCCS, 20138 Milano, Italy.
| |
Collapse
|
16
|
Massunaga ND, França CN, Bianco HT, Ferreira CE, Kato JT, Póvoa RM, Figueiredo Neto AM, Izar MCO, Fonseca FAH. Circulating microparticles and central blood pressure according to antihypertensive strategy. Clinics (Sao Paulo) 2019; 74:e1234. [PMID: 31721907 PMCID: PMC6827330 DOI: 10.6061/clinics/2019/e1234] [Citation(s) in RCA: 3] [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: 04/02/2019] [Accepted: 08/18/2019] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVES This prospective, randomized, open-label study aimed to compare the effects of antihypertensive treatment based on amlodipine or hydrochlorothiazide on the circulating microparticles and central blood pressure values of hypertensive patients. METHODS The effects of treatments on circulating microparticles were assessed during monotherapy and after the consecutive addition of valsartan and rosuvastatin followed by the withdrawal of rosuvastatin. Each treatment period lasted for 30 days. Central blood pressure and pulse wave velocity were measured at the end of each period. Endothelial, monocyte, and platelet circulating microparticles were determined by flow cytometry. Central blood pressure values and pulse wave velocity were recorded at the end of each treatment period. RESULTS No differences in brachial blood pressure were observed between the treatment groups throughout the study. Although similar central blood pressure values were observed during monotherapy, lower systolic and diastolic central blood pressure values and early and late blood pressure peaks were observed in the amlodipine arm after the addition of valsartan alone or combined with rosuvastatin. Hydrochlorothiazide-based therapy was associated with a lower number of endothelial microparticles throughout the study, whereas a higher number of platelet microparticles was observed after rosuvastatin withdrawal in the amlodipine arm. CONCLUSIONS Despite similar brachial blood pressure values between groups throughout the study, exposure to amlodipine was associated with lower central blood pressure values after combination with valsartan, indicating a beneficial interaction. Differences between circulating microparticles were modest and were mainly influenced by rosuvastatin withdrawal in the amlodipine arm.
Collapse
Affiliation(s)
- Nayara D. Massunaga
- Departamento de Medicina, Universidade Federal de Sao Paulo (UNIFESP), Sao Paulo, SP, BR
- Corresponding author. E-mail:
| | - Carolina N. França
- Departamento de Medicina, Universidade Federal de Sao Paulo (UNIFESP), Sao Paulo, SP, BR
- Universidade Santo Amaro (UNISA), Sao Paulo, SP, BR
- Corresponding author. E-mail:
| | - Henrique T. Bianco
- Departamento de Medicina, Universidade Federal de Sao Paulo (UNIFESP), Sao Paulo, SP, BR
| | - Carlos E.S. Ferreira
- Departamento de Medicina, Universidade Federal de Sao Paulo (UNIFESP), Sao Paulo, SP, BR
- Hospital Israelita Albert Einstein, Sao Paulo, SP, BR
| | - Juliana T. Kato
- Departamento de Medicina, Universidade Federal de Sao Paulo (UNIFESP), Sao Paulo, SP, BR
| | - Rui M.S. Póvoa
- Departamento de Medicina, Universidade Federal de Sao Paulo (UNIFESP), Sao Paulo, SP, BR
| | - Antonio M. Figueiredo Neto
- Instituto Nacional de Ciencia e Tecnologia de Fluidos Complexos, Universidade Sao Paulo, Sao Paulo, SP, BR
| | - Maria Cristina O. Izar
- Departamento de Medicina, Universidade Federal de Sao Paulo (UNIFESP), Sao Paulo, SP, BR
| | | |
Collapse
|
17
|
Chyrchel B, Drożdż A, Długosz D, Stępień EŁ, Surdacki A. Platelet Reactivity And Circulating Platelet-Derived Microvesicles Are Differently Affected By P2Y 12 Receptor Antagonists. Int J Med Sci 2019; 16:264-275. [PMID: 30745807 PMCID: PMC6367525 DOI: 10.7150/ijms.28580] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 11/29/2018] [Indexed: 12/14/2022] Open
Abstract
Background: Platelet-derived microvesicles (PMVs), shed from platelet surface membranes, constitute the majority of circulating microvesicles and have been implicated in procoagulant, pro-inflammatory and pro-atherosclerotic effects. Our aim was to compare plasma PMVs numbers in relation to platelet reactivity during dual antiplatelet therapy (DAPT) with various P2Y12 adenosine diphosphate (ADP) receptor antagonists. Methods: In pre-discharge men treated with DAPT for an acute coronary syndrome, plasma PMVs were quantified by flow cytometry on the basis of CD62P (P-selectin) and CD42 (glycoprotein Ib) positivity, putative indices of PMVs release from activated and all platelets, respectively. ADP-induced platelet aggregation was measured by multiple-electrode aggregometry. Results: Clinical characteristics were similar in patients on clopidogrel (n=16), prasugrel (n=10) and ticagrelor (n=12). Platelet reactivity was comparably reduced on ticagrelor or prasugrel versus clopidogrel (p<0.01). Compared to clopidogrel-treated patients, CD42+/CD62P+ PMVs counts were 3-4-fold lower in subjects receiving ticagrelor (p=0.001) or prasugrel (p<0.05), while CD42+ PMVs were significantly reduced on ticagrelor (by about 6-fold, p<0.001), but not prasugrel (p=0.3). CD42+/CD62P+ PMVs numbers correlated positively to the ADP-induced aggregation on clopidogrel (p<0.01) or prasugrel (p<0.05), which was absent in ticagrelor users (p=0.8). CD42+ PMVs counts were unrelated to platelet reactivity (p>0.5). Conclusions: Higher antiplatelet potency of prasugrel and ticagrelor versus clopidogrel is associated with decreased plasma CD42+/CD62P+ PMVs numbers. However, in contrast to thienopyridines, the association of reduced CD42+/CD62P+ PMVs counts with ticagrelor use appears independent of its anti-aggregatory effect. Despite similar platelet-inhibitory activity of ticagrelor and prasugrel, only the treatment with ticagrelor seems associated with lower total PMVs release. Our preliminary findings may suggest a novel pleiotropic effect of ticagrelor extending beyond pure anti-aggregatory properties of the drug.
Collapse
Affiliation(s)
- Bernadeta Chyrchel
- Second Department of Cardiology, Jagiellonian University Medical College, Cracow, Poland
| | - Anna Drożdż
- Małopolska Center of Biotechnology, Jagiellonian University, Cracow, Poland
| | - Dorota Długosz
- Students' Scientific Group at the Second Department of Cardiology, Jagiellonian University Medical College, Cracow, Poland
| | - Ewa Ł Stępień
- Department of Medical Physics, Institute of Physics, Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Cracow, Poland
| | - Andrzej Surdacki
- Second Department of Cardiology, Jagiellonian University Medical College, Cracow, Poland
| |
Collapse
|
18
|
Abstract
Platelet-derived microvesicles (pMVs) are small, heterogeneous vesicles released from platelet membranes as a result of activation. These microvesicles possess a wide range of properties, including prothrombotic, proatherogenic, proinflammatory, immunomodulatory, and even anticoagulant activity. The elevated release of these microvesicles has been observed in various metabolic, inflammatory, thrombotic, and vascular diseases, including ischemic heart disease, stroke, hypertension, diabetes, and connective tissue disease. Modulation of both pMV generation and the expression of their surface molecules may have beneficial clinical implications and could become a novel therapeutic target. However, mechanisms by which pharmacological agents can modify pMV formation are elusive. The purpose of this review is to discuss the effects of drugs routinely used in primary and secondary prevention of vascular disease on the release of pMV and expression of their surface procoagulant and proinflammatory molecules.
Collapse
Affiliation(s)
- Justyna Rosińska
- Department of Neurology, Poznan University of Medical Sciences, ul. Przybyszewskiego 49, 60-355, Poznan, Poland.
| | - Maria Łukasik
- Department of Neurology, Poznan University of Medical Sciences, ul. Przybyszewskiego 49, 60-355, Poznan, Poland
| | - Wojciech Kozubski
- Department of Neurology, Poznan University of Medical Sciences, ul. Przybyszewskiego 49, 60-355, Poznan, Poland
| |
Collapse
|
19
|
Chen Y, Li G, Liu ML. Microvesicles as Emerging Biomarkers and Therapeutic Targets in Cardiometabolic Diseases. GENOMICS PROTEOMICS & BIOINFORMATICS 2018; 16:50-62. [PMID: 29462670 PMCID: PMC6000161 DOI: 10.1016/j.gpb.2017.03.006] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 03/03/2017] [Accepted: 03/23/2017] [Indexed: 12/20/2022]
Abstract
Microvesicles (MVs, also known as microparticles) are small vesicles that originate from plasma membrane of almost all eukaryotic cells during apoptosis or activation. MVs can serve as extracellular vehicles to transport bioactive molecules from their parental cells to recipient target cells, thereby serving as novel mediators for intercellular communication. Importantly, more and more evidence indicates that MVs could play important roles in early pathogenesis and subsequent progression of cardiovascular and metabolic diseases. Elevated plasma concentrations of MVs, originating from red blood cells, leukocytes, platelets, or other organs and tissues, have been reported in various cardiometabolic diseases. Circulating MVs could serve as potential biomarkers for disease diagnosis or therapeutic monitoring. In this review, we summarized recently-published studies in the field and discussed the role of MVs in the pathogenesis of cardiometabolic diseases. The emerging values of MVs that serve as biomarker for non-invasive diagnosis and prognosis, as well as their roles as novel therapeutic targets in cardiometabolic diseases, were also described.
Collapse
Affiliation(s)
- Yan Chen
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Guangping Li
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China.
| | - Ming-Lin Liu
- Section of Endocrinology, Diabetes and Metabolism, Department of Medicine, Temple University School of Medicine, Philadelphia, PA 19140, USA; Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19140, USA; Philadelphia VA Medical Center, Philadelphia, PA 19140, USA.
| |
Collapse
|
20
|
Fonseca FAH, Izar MC, Maugeri IML, Berwanger O, Damiani LP, Pinto IM, Szarf G, França CN, Bianco HT, Moreira FT, Caixeta A, Alves CMR, Soriano Lopes A, Klassen A, Tavares MFM, Fonseca HA, Carvalho ACC. Effects of four antiplatelet/statin combined strategies on immune and inflammatory responses in patients with acute myocardial infarction undergoing pharmacoinvasive strategy: Design and rationale of the B and T Types of Lymphocytes Evaluation in Acute Myocardial Infarction (BATTLE-AMI) study: study protocol for a randomized controlled trial. Trials 2017; 18:601. [PMID: 29258572 PMCID: PMC5735810 DOI: 10.1186/s13063-017-2361-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2017] [Accepted: 11/17/2017] [Indexed: 02/07/2023] Open
Abstract
Background Early reperfusion of the occluded coronary artery during acute myocardial infarction is considered crucial for reduction of infarcted mass and recovery of ventricular function. Effective microcirculation and the balance between protective and harmful lymphocytes may have roles in reperfusion injury and may affect final ventricular remodeling. Methods/design BATTLE-AMI is an open-label, randomized trial comparing the effects of four therapeutic strategies (rosuvastatin/ticagrelor, rosuvastatin/clopidogrel, simvastatin plus ezetimibe/ticagrelor, or simvastatin plus ezetimibe/clopidogrel) on infarcted mass and left ventricular ejection fraction (LVEF) (blinded endpoints) in patients with ST-segment elevation myocardial infarction submitted to fibrinolytic therapy before coronary angiogram (pharmacoinvasive strategy). All patients (n = 300, 75 per arm) will be followed up for six months. The effects of treatment on subsets of B and T lymphocytes will be determined by flow-cytometry/ELISPOT and will be correlated with the infarcted mass, LVEF, and microcirculation perfusion obtained by cardiac magnetic resonance imaging. The primary hypothesis is that the combined rosuvastatin/ticagrelor therapy will be superior to other therapies (particularly for the comparison with simvastatin plus ezetimibe/clopidogrel) for the achievement of better LVEF at 30 days (primary endpoint) and smaller infarcted mass (secondary endpoint) at 30 days and six months. The trial will also evaluate the improvement in the immune/inflammatory responses mediated by B and T lymphocytes. Omics field (metabolomics and proteomics) will help to understand these responses by molecular events. Discussion BATTLE-AMI is aimed to (1) evaluate the role of subsets of lymphocytes on microcirculation improvement and (2) show how the choice of statin/antiplatelet therapy may affect cardiac remodeling after acute myocardial infarction with ST elevation. Trial registration ClinicalTrials.gov, NCT02428374. Registered on 28 September 2014. Electronic supplementary material The online version of this article (doi:10.1186/s13063-017-2361-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Francisco A H Fonseca
- Universidade Federal de São Paulo, Rua Loefgren 1350, 04040-001, São Paulo, SP, Brazil.
| | - Maria Cristina Izar
- Universidade Federal de São Paulo, Rua Loefgren 1350, 04040-001, São Paulo, SP, Brazil
| | - Ieda M L Maugeri
- Universidade Federal de São Paulo, Rua Loefgren 1350, 04040-001, São Paulo, SP, Brazil
| | - Otavio Berwanger
- Hospital do Coração, Rua Desembargador Eliseu Guilherme, 147, São Paulo, Brazil
| | - Lucas P Damiani
- Hospital do Coração, Rua Desembargador Eliseu Guilherme, 147, São Paulo, Brazil
| | - Ibraim M Pinto
- Instituto Dante Pazzanese de Cardiologia, Avenida Dante Pazzanese 500, São Paulo, Brazil
| | - Gilberto Szarf
- Universidade Federal de São Paulo, Rua Loefgren 1350, 04040-001, São Paulo, SP, Brazil
| | - Carolina N França
- Universidade Santo Amaro, Rua Professor Enéas de Siqueira 340, São Paulo, Brazil
| | - Henrique T Bianco
- Universidade Federal de São Paulo, Rua Loefgren 1350, 04040-001, São Paulo, SP, Brazil
| | - Flavio T Moreira
- Universidade Federal de São Paulo, Rua Loefgren 1350, 04040-001, São Paulo, SP, Brazil
| | - Adriano Caixeta
- Universidade Federal de São Paulo, Rua Loefgren 1350, 04040-001, São Paulo, SP, Brazil
| | - Claudia M R Alves
- Universidade Federal de São Paulo, Rua Loefgren 1350, 04040-001, São Paulo, SP, Brazil
| | - Aline Soriano Lopes
- Universidade Federal de São Paulo, Rua Loefgren 1350, 04040-001, São Paulo, SP, Brazil
| | - Aline Klassen
- Universidade Federal de São Paulo, Rua Loefgren 1350, 04040-001, São Paulo, SP, Brazil
| | - Marina F M Tavares
- Universidade de São Paulo, Avenida Professor Lineu Prestes, 748, São Paulo, Brazil
| | - Henrique A Fonseca
- Universidade Federal de São Paulo, Rua Loefgren 1350, 04040-001, São Paulo, SP, Brazil
| | - Antonio C C Carvalho
- Universidade Federal de São Paulo, Rua Loefgren 1350, 04040-001, São Paulo, SP, Brazil
| | | |
Collapse
|
21
|
Badimon L, Suades R, Arderiu G, Peña E, Chiva-Blanch G, Padró T. Microvesicles in Atherosclerosis and Angiogenesis: From Bench to Bedside and Reverse. Front Cardiovasc Med 2017; 4:77. [PMID: 29326946 PMCID: PMC5741657 DOI: 10.3389/fcvm.2017.00077] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 11/22/2017] [Indexed: 12/28/2022] Open
Abstract
Atherosclerosis (AT) is a progressive chronic disease involving lipid accumulation, fibrosis, and inflammation in medium and large-sized arteries, and it is the main cause of cardiovascular disease (CVD). AT is caused by dyslipidemia and mediated by both innate and adaptive immune responses. Despite lipid-lowering drugs have shown to decrease the risk of cardiovascular events (CVEs), there is a significant burden of AT-related morbidity and mortality. Identification of subjects at increased risk for CVE as well as discovery of novel therapeutic targets for improved treatment strategies are still unmet clinical needs in CVD. Microvesicles (MVs), small extracellular plasma membrane particles shed by activated and apoptotic cells have been widely linked to the development of CVD. MVs from vascular and resident cells by facilitating exchange of biological information between neighboring cells serve as cellular effectors in the bloodstream and play a key role in all stages of disease progression. This article reviews the current knowledge on the role of MVs in AT and CVD. Attention is focused on novel aspects of MV-mediated regulatory mechanisms from endothelial dysfunction, vascular wall inflammation, oxidative stress, and apoptosis to coagulation and thrombosis in the progression and development of atherothrombosis. MV contribution to vascular remodeling is also discussed, with a particular emphasis on the effect of MVs on the crosstalk between endothelial cells and smooth muscle cells, and their role regulating the active process of AT-driven angiogenesis and neovascularization. This review also highlights the latest findings and main challenges on the potential prognostic, diagnostic, and therapeutic value of cell-derived MVs in CVD. In summary, MVs have emerged as new regulators of biological functions in atherothrombosis and might be instrumental in cardiovascular precision medicine; however, significant efforts are still needed to translate into clinics the latest findings on MV regulation and function.
Collapse
Affiliation(s)
- Lina Badimon
- Cardiovascular Research Center (ICCC) and CiberCV, Sant Pau Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain
- Cardiovascular Research Chair, UAB, Barcelona, Spain
| | - Rosa Suades
- Cardiovascular Research Center (ICCC) and CiberCV, Sant Pau Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain
| | - Gemma Arderiu
- Cardiovascular Research Center (ICCC) and CiberCV, Sant Pau Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain
| | - Esther Peña
- Cardiovascular Research Center (ICCC) and CiberCV, Sant Pau Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain
| | - Gemma Chiva-Blanch
- Cardiovascular Research Center (ICCC) and CiberCV, Sant Pau Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain
| | - Teresa Padró
- Cardiovascular Research Center (ICCC) and CiberCV, Sant Pau Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain
| |
Collapse
|
22
|
Shantsila E, Montoro-García S, Gallego P, Lip GYH. Circulating microparticles: challenges and perspectives of flow cytometric assessment. Thromb Haemost 2017; 111:1009-14. [DOI: 10.1160/th13-11-0937] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Accepted: 01/07/2014] [Indexed: 12/18/2022]
Abstract
SummaryCirculating blood microparticles are likely to play a significant role as messengers of biological information. Their accurate quantification and characterisation is challenging and needs to be carefully designed with preferable usage of fresh minimally-processed blood samples. Utilisation of flow cytometers specifically designed for analysis of small-size particles is likely to provide considerable methodological advantages and should be the preferable option. This viewpoint manuscript provides a critical summary of the key methodological aspects of microparticle analysis.Note: The review process for this viewpoint article was fully handled by Christian Weber, Editor in Chief.
Collapse
|
23
|
Wang Z, Cai W, Hu S, Xia Y, Wang Y, Zhang Q, Chen L. A Meta-Analysis of Circulating Microvesicles in Patients with Myocardial Infarction. Arq Bras Cardiol 2017; 109:0. [PMID: 28700020 PMCID: PMC5576120 DOI: 10.5935/abc.20170102] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 03/15/2017] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND: Cell-derived microvesicles (MVs) are vesicles released from activated or apoptotic cells. However, the levels of MVs in myocardial infarction have been found inconsistent in researches. OBJECTIVE: To assess the association between MVs and myocardial infarction by conducting a meta-analysis. METHODS: A systematic literature search on PubMed, Embase, Cochran, Google Scholar electronic database was conducted. Comparison of the MVs levels between myocardial infarction patients and healthy persons were included in our study. Standard Mean Difference (SMD) and 95% confidence interval (CI) in groups were calculated and meta-analyzed. RESULTS: 11 studies with a total of 436 participants were included. Compared with the health persons, AMVs [SMD = 3.65, 95% CI (1.03, 6.27)], PMVs [SMD = 2.88, 95% CI (1.82, 3.93),] and EMVs [SMD = 2.73, 95% CI (1.13, 4.34)], levels were higher in patients with myocardial infarction. However, LMVs levels [SMD = 0.73, 95% CI (-0.57, 2.03)] were not changed significantly in patients with myocardial infarction. CONCLUSIONS: AMVs, PMVs and EMVs might be potential biomarkers for myocardial infarction. FUNDAMENTOS: As microvesículas derivadas de células (MVs) são vesículas liberadas de células ativadas ou apoptóticas. No entanto, os níveis de MVs no infarto do miocárdio foram encontrados inconsistentes nas pesquisas. OBJETIVO: Avaliar a associação entre MV e infarto do miocárdio por meio de uma meta-análise. MÉTODOS: Foi realizada uma pesquisa sistemática na literatura em PubMed, Embase, Cochran e no banco de dados eletrônico do Google Scholar. Uma comparação dos níveis de MV entre pacientes com infarto do miocárdio e pessoas saudáveis foi incluída no nosso estudo. A Diferença Média Padrão (DMP) e o intervalo de confiança (IC) de 95% nos grupos foram calculadas e meta-analisadas. RESULTADOS: Foram incluídos 11 estudos com um total de 436 participantes. Em comparação com as pessoas saudáveis, as MVA [DMP = 3,65, IC 95% (1,03, 6,27)], MVPs [DMP = 2,88, IC 95% (1,82, 3,93)] e MVEs [DMP = 2,73, IC 95% (1,13, 4.34)], foram maiores em pacientes com infarto do miocárdio. No entanto, os níveis de MVL [DMP = 0,73, IC 95% (-0,57, 2,03)] não foram alterados significativamente em pacientes com infarto do miocárdio. CONCLUSÕES: MVAs, MVPs e MVEs podem ser biomarcadores potenciais para o infarto do miocárdio.
Collapse
Affiliation(s)
- Zhida Wang
- Key Laboratory of Hormones and Development (Ministry of Health) -
Tianjin Key Laboratory of Metabolic Diseases - Tianjin Metabolic Diseases Hospital
& Tianjin Institute of Endocrinology - Tianjin Medical University, Tianjin -
China
| | - Wang Cai
- Department of Surgery - Tianjin Nankai Hospital - Tianjin Medical
University, Tianjin, China
| | - Shaolan Hu
- Key Laboratory of Hormones and Development (Ministry of Health) -
Tianjin Key Laboratory of Metabolic Diseases - Tianjin Metabolic Diseases Hospital
& Tianjin Institute of Endocrinology - Tianjin Medical University, Tianjin -
China
| | - Yufei Xia
- School of Nursing - Tianjin Medical University, Tianjin - China
| | - Yao Wang
- Department of Pharmacology - School of Basic Medical Science -
Tianjin Medical University, Tianjin - China
| | - Qi Zhang
- Institute of Integrative Medicines for Acute Abdominal Diseases -
Nankai Hospital, Tianjin - China
| | - Liming Chen
- Key Laboratory of Hormones and Development (Ministry of Health) -
Tianjin Key Laboratory of Metabolic Diseases - Tianjin Metabolic Diseases Hospital
& Tianjin Institute of Endocrinology - Tianjin Medical University, Tianjin -
China
| |
Collapse
|
24
|
Tomaniak M, Gąsecka A, Filipiak KJ. Cell-derived microvesicles in cardiovascular diseases and antiplatelet therapy monitoring - A lesson for future trials? Current evidence, recent progresses and perspectives of clinical application. Int J Cardiol 2016; 226:93-102. [PMID: 27792994 DOI: 10.1016/j.ijcard.2016.10.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2016] [Revised: 10/03/2016] [Accepted: 10/04/2016] [Indexed: 12/28/2022]
Abstract
Circulating cell-derived microvesicles (MV) represent a subject of increasing interest in recent years as potential effectors in thrombosis, inflammation and vascular injury. Although several studies demonstrated an association between MV plasma concentrations and clinical manifestations of atherosclerosis as well as a clear effect of cardiovascular pharmacotherapy on MV formation pattern, the application of this promising biomarkers in clinical cardiology has been hindered so far due to heterogeneity of the hitherto studies employing non-standardized methodologies. Recently great progresses have been done and international initiatives were started to unify the pre-analytical and analytical procedures, improve the comparison of measurements between the laboratories and increase detector sensitivity of flow cytometry - a golden standard for MV assessment. Likewise, the concept of a "therapeutic window" of P2Y12 inhibitor therapy was introduced, as the prognostic significance of bleeding consequences is equally important with that of ischemic events, particularly with the expanding use of more potent P2Y12 inhibitors. In this review we summarize currently available studies on circulating MV in terms of cardiovascular diagnosis, risk stratification and influence of antiplatelet agents on the MV release to postulate possible future role of MV as supplementary biomarker in monitoring of individual response to antiplatelet therapy. Methodological pitfalls faced in the previous studies and obstacles that need to be addressed before further trials and translation of MV-based assays into clinical practice were defined.
Collapse
Affiliation(s)
- Mariusz Tomaniak
- Medical University of Warsaw, 1st Department of Cardiology, Warsaw, Poland.
| | - Aleksandra Gąsecka
- Medical University of Warsaw, 1st Department of Cardiology, Warsaw, Poland
| | | |
Collapse
|
25
|
Badimon L, Suades R, Fuentes E, Palomo I, Padró T. Role of Platelet-Derived Microvesicles As Crosstalk Mediators in Atherothrombosis and Future Pharmacology Targets: A Link between Inflammation, Atherosclerosis, and Thrombosis. Front Pharmacol 2016; 7:293. [PMID: 27630570 PMCID: PMC5005978 DOI: 10.3389/fphar.2016.00293] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 08/22/2016] [Indexed: 12/24/2022] Open
Abstract
Reports in the last decade have suggested that the role of platelets in atherosclerosis and its thrombotic complications may be mediated, in part, by local secretion of platelet-derived microvesicles (pMVs), small cell blebs released during the platelet activation process. MVs are the most abundant cell-derived microvesicle subtype in the circulation. High concentrations of circulating MVs have been reported in patients with atherosclerosis, acute vascular syndromes, and/or diabetes mellitus, suggesting a potential correlation between the quantity of microvesicles and the clinical severity of the atherosclerotic disease. pMVs are considered to be biomarkers of disease but new information indicates that pMVs are also involved in signaling functions. pMVs evoke or promote haemostatic and inflammatory responses, neovascularization, cell survival, and apoptosis, processes involved in the pathophysiology of cardiovascular disease. This review is focused on the complex cross-talk between platelet-derived microvesicles, inflammatory cells and vascular elements and their relevance in the development of the atherosclerotic disease and its clinical outcomes, providing an updated state-of-the art of pMV involvement in atherothrombosis and pMV potential use as therapeutic agent influencing cardiovascular biomedicine in the future.
Collapse
Affiliation(s)
- Lina Badimon
- Cardiovascular Research Center, Consejo Superior de Investigaciones Científicas - Institut Català de Ciències Cardiovasculars, Institut d'Investigació Biomèdica Sant Pau, Hospital Santa Creu i Sant PauBarcelona, Spain; Cardiovascular Research Chair, Universitat Autònoma de BarcelonaBarcelona, Spain
| | - Rosa Suades
- Cardiovascular Research Center, Consejo Superior de Investigaciones Científicas - Institut Català de Ciències Cardiovasculars, Institut d'Investigació Biomèdica Sant Pau, Hospital Santa Creu i Sant Pau Barcelona, Spain
| | - Eduardo Fuentes
- Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, Interdisciplinary Excellence Research Program on Healthy Aging, Universidad de TalcaTalca, Chile; Centro de Estudios en Alimentos Procesados, Conicyt-RegionalGore-Maule, Talca, Chile
| | - Iván Palomo
- Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, Interdisciplinary Excellence Research Program on Healthy Aging, Universidad de TalcaTalca, Chile; Centro de Estudios en Alimentos Procesados, Conicyt-RegionalGore-Maule, Talca, Chile
| | - Teresa Padró
- Cardiovascular Research Center, Consejo Superior de Investigaciones Científicas - Institut Català de Ciències Cardiovasculars, Institut d'Investigació Biomèdica Sant Pau, Hospital Santa Creu i Sant Pau Barcelona, Spain
| |
Collapse
|
26
|
Jung C, Lichtenauer M, Figulla HR, Wernly B, Goebel B, Foerster M, Edlinger C, Lauten A. Microparticles in patients undergoing transcatheter aortic valve implantation (TAVI). Heart Vessels 2016; 32:458-466. [PMID: 27488119 PMCID: PMC5371631 DOI: 10.1007/s00380-016-0885-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Accepted: 07/29/2016] [Indexed: 12/13/2022]
Abstract
Degenerative aortic stenosis (AS) is the most frequent form of acquired valvular heart disease. AS is known to entail endothelial dysfunction caused by increased mechanical shear stress leading to elevated circulatory levels of microparticles. Endothelial and platelet microparticles (EMP and PMP) are small vesicles that originate from activated cells and thrombocytes. We sought to evaluate whether transcatheter aortic valve implantation (TAVI) procedure would elicit effects on circulating EMP and PMP. 92 patients undergoing TAVI procedure for severe AS were included in this study. Samples were obtained at each visit before TAVI, 1 week post-procedure and at 1, 3 and after 6 months after TAVI and were evaluated using flow cytometry. A 12 month clinical follow-up was also performed. CD62E+ EMP concentration before TAVI was 21.11 % (±6.6 % SD) and declined to 20.99 % (±6.8 % SD) after 1 week, to 16.63 % (±5.4 % SD, p < 0.0001) after 1 month, to 17.08 % (±4.6 % SD, p < 0.0001) after 3 months and to 15.94 % (±5.4 % SD, p < 0.0001) after 6 months. CD31+/CD42b-, CD31+/Annexin+/- EMP remained unchanged. CD31+/CD41b+ PMP evidenced a slight, but statistically significant increase after TAVI and remained elevated during the entire follow-up. Apart from a procedure-related improvement in echocardiographic parameters, TAVI procedure led also to a decline in CD62E+ EMP. The reduction in pressure gradients with less hemodynamic shear stress seems also to have beneficially affected endothelial homeostasis.
Collapse
Affiliation(s)
- Christian Jung
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Hospital Düsseldorf, University Duesseldorf, Moorenstrasse 5, 40225, Düsseldorf, Germany.
- Department of Cardiology, Clinic of Internal Medicine I, Universitätsherzzentrum Thüringen, Friedrich Schiller University Jena, Jena, Germany.
| | - Michael Lichtenauer
- Department of Cardiology, Clinic of Internal Medicine II, Paracelsus Medical University of Salzburg, Salzburg, Austria
- Department of Cardiology, Clinic of Internal Medicine I, Universitätsherzzentrum Thüringen, Friedrich Schiller University Jena, Jena, Germany
| | - Hans-Reiner Figulla
- Department of Cardiology, Clinic of Internal Medicine I, Universitätsherzzentrum Thüringen, Friedrich Schiller University Jena, Jena, Germany
| | - Bernhard Wernly
- Department of Cardiology, Clinic of Internal Medicine II, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Bjoern Goebel
- Department of Cardiology, Clinic of Internal Medicine I, Universitätsherzzentrum Thüringen, Friedrich Schiller University Jena, Jena, Germany
| | - Martin Foerster
- Department of Cardiology, Clinic of Internal Medicine I, Universitätsherzzentrum Thüringen, Friedrich Schiller University Jena, Jena, Germany
| | - Christoph Edlinger
- Department of Cardiology, Clinic of Internal Medicine II, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Alexander Lauten
- Department of Cardiology, Clinic of Internal Medicine I, Universitätsherzzentrum Thüringen, Friedrich Schiller University Jena, Jena, Germany
- Department of Cardiology, Charité-Universitaetsmedizin Berlin, Berlin, Germany
| |
Collapse
|
27
|
Hartopo AB, Puspitawati I, Gharini PPR, Setianto BY. Platelet microparticle number is associated with the extent of myocardial damage in acute myocardial infarction. Arch Med Sci 2016; 12:529-37. [PMID: 27279844 PMCID: PMC4889687 DOI: 10.5114/aoms.2016.59926] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 09/07/2014] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION Activated platelets generate microparticles. Increased platelet microparticles occur in acute myocardial infarction (AMI) and contribute to intracoronary thrombosis and subsequent myocardial injury. This study aimed to investigate the impact of platelet microparticles on intracoronary thrombosis by assessing the relationship between platelet microparticles and the extent of myocardial damage in AMI. MATERIAL AND METHODS This was a cross sectional study. The subjects were patients with acute coronary syndrome (ACS). Forty-one consecutive subjects with ACS admitted to intensive cardiovascular care unit were enrolled. The clinical spectrum of ACS comprised AMI (n = 26), both ST-elevation AMI (STEMI) and non-ST-elevation AMI (NSTEMI), and unstable angina (n = 15). Platelet microparticles were isolated from peripheral venous blood and detected with anti-CD42b-PE by the flow cytometry method. The extent of myocardial damage was determined by measuring the peak level of serial cardiac enzymes within 24 h of admission. RESULTS Subjects with AMI had a significantly higher number of platelet microparticles than those with unstable angina (4855 ±4509/µl vs. 2181 ±1923/µl respectively; p = 0.036). Subjects with STEMI had the highest number of platelet microparticles, but no significant difference was detected as compared to those with NSTEMI (5775 ±5680/µl vs. 3601 ±1632/µl). The number of platelet microparticles in AMI was positively associated with the extent of myocardial damage (peak CK-MB: r = 0.408, p = 0.019 and peak GOT: r = 0.384, p = 0.026). CONCLUSIONS The number of platelet microparticles was increased in AMI as compared to unstable angina and associated with the extent of myocardial damage.
Collapse
Affiliation(s)
- Anggoro Budi Hartopo
- Department of Cardiology and Vascular Medicine, Faculty of Medicine Universitas Gadjah Mada – Dr. Sardjito Hospital, Yogyakarta, Indonesia
| | - Ira Puspitawati
- Department of Clinical Pathology, Faculty of Medicine Universitas Gadjah Mada – Dr. Sardjito Hospital, Yogyakarta, Indonesia
| | - Putrika Prastuti Ratna Gharini
- Department of Cardiology and Vascular Medicine, Faculty of Medicine Universitas Gadjah Mada – Dr. Sardjito Hospital, Yogyakarta, Indonesia
| | - Budi Yuli Setianto
- Department of Cardiology and Vascular Medicine, Faculty of Medicine Universitas Gadjah Mada – Dr. Sardjito Hospital, Yogyakarta, Indonesia
| |
Collapse
|
28
|
Tempo JA, Englyst NA, Holloway JA, Smith DC. Platelet Microvesicles (Microparticles) in Cardiac Surgery. J Cardiothorac Vasc Anesth 2016; 30:222-8. [DOI: 10.1053/j.jvca.2015.08.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2015] [Indexed: 11/11/2022]
|
29
|
Zhou BD, Guo G, Zheng LM, Zu LY, Gao W. Microparticles as novel biomarkers and therapeutic targets in coronary heart disease. Chin Med J (Engl) 2015; 128:267-72. [PMID: 25591573 PMCID: PMC4837849 DOI: 10.4103/0366-6999.149231] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Affiliation(s)
| | | | | | - Ling-Yun Zu
- Department of Cardiology, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Peking University Third Hospital, Beijing 100191, China
| | | |
Collapse
|
30
|
Extracellular vesicles as new pharmacological targets to treat atherosclerosis. Eur J Pharmacol 2015; 763:90-103. [PMID: 26142082 DOI: 10.1016/j.ejphar.2015.06.047] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 05/13/2015] [Accepted: 06/25/2015] [Indexed: 01/09/2023]
Abstract
Extracellular vesicles released by most cell types, include apoptotic bodies (ABs), microvesicles (MVs) and exosomes. They play a crucial role in physiology and pathology, contributing to "cell-to-cell" communication by modifying the phenotype and the function of target cells. Thus, extracellular vesicles participate in the key processes of atherosclerosis from endothelial dysfunction, vascular wall inflammation to vascular remodeling. The purpose of this review is to summarize recent findings on extracellular vesicle formation, structure, release and clearance. We focus on the deleterious and beneficial effects of extracellular vesicles in the development of atherosclerosis. The potential role of extracellular vesicles as biomarkers and pharmacological targets, their innate therapeutic capacity, or their use for novel drug delivery devices in atherosclerotic cardiovascular diseases will also be discussed.
Collapse
|
31
|
Horn P, Baars T, Kahlert P, Heiss C, Westenfeld R, Kelm M, Erbel R, Heusch G, Kleinbongard P. Release of Intracoronary Microparticles during Stent Implantation into Stable Atherosclerotic Lesions under Protection with an Aspiration Device. PLoS One 2015; 10:e0124904. [PMID: 25915510 PMCID: PMC4411166 DOI: 10.1371/journal.pone.0124904] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 03/06/2015] [Indexed: 01/21/2023] Open
Abstract
OBJECTIVE Stent implantation into atherosclerotic coronary vessels impacts on downstream microvascular function and induces the release of particulate debris and soluble substances, which differs qualitatively and quantitatively between native right coronary arteries (RCAs) and saphenous vein grafts on right coronary arteries (SVG-RCAs). We have now quantified the release of microparticles (MPs) during stent implantation into stable atherosclerotic lesions and compared the release between RCAs and SVG-RCAs. METHODS In symptomatic, male patients with stable angina and a stenosis in their RCA or SVG-RCA, respectively (n = 14/14), plaque volume and composition were analyzed using intravascular ultrasound before stent implantation. Coronary aspirate was retrieved during stent implantation with a distal occlusion/aspiration device and divided into particulate debris and plasma. Particulate debris was weighed. Platelet-derived MPs (PMPs) were distinguished by flow cytometry as CD41+, endothelium-derived MPs (EMPs) as CD144+, CD62E+ and CD31+/CD41-, leukocyte-derived MPs as CD45+, and erythrocyte-derived MPs as CD235+. RESULTS In patients with comparable plaque volume and composition in RCAs and SVG-RCAs, intracoronary PMPs and EMPs were increased after stent implantation into their RCAs and SVG-RCAs (CD41+: 2729.6 ± 645.6 vs. 4208.7 ± 679.4 and 2355.9 ± 503.9 vs. 3285.8 ± 733.2 nr/µL; CD144+: 451.5 ± 87.9 vs. 861.7 ± 147.0 and 444.6 ± 74.8 vs. 726.5 ± 136.4 nr/µL; CD62E+: 1404.1 ± 247.7 vs. 1844.3 ± 378.6 and 1084.6 ± 211.0 vs. 1783.8 ± 384.3 nr/µL, P < 0.05), but not different between RCAs and SVG-RCAs. CONCLUSION Stenting in stable atherosclerotic lesions is associated with a substantial release not only of PMPs, but also of EMPs in RCAs and SVG-RCAs. Their release does not differ between RCAs and SVG-RCAs. TRIAL REGISTRATION ClinicalTrials.gov NCT01430884.
Collapse
Affiliation(s)
- Patrick Horn
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Duesseldorf, Duesseldorf, Germany
| | - Theodor Baars
- Institute for Pathophysiology, West German Heart and Vascular Centre Essen, University of Essen Medical School, Essen, Germany
- Clinic for Cardiology, West German Heart and Vascular Centre Essen, University of Essen Medical School, Essen, Germany
| | - Philipp Kahlert
- Clinic for Cardiology, West German Heart and Vascular Centre Essen, University of Essen Medical School, Essen, Germany
| | - Christian Heiss
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Duesseldorf, Duesseldorf, Germany
| | - Ralf Westenfeld
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Duesseldorf, Duesseldorf, Germany
| | - Malte Kelm
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Duesseldorf, Duesseldorf, Germany
| | - Raimund Erbel
- Clinic for Cardiology, West German Heart and Vascular Centre Essen, University of Essen Medical School, Essen, Germany
| | - Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Centre Essen, University of Essen Medical School, Essen, Germany
| | - Petra Kleinbongard
- Institute for Pathophysiology, West German Heart and Vascular Centre Essen, University of Essen Medical School, Essen, Germany
- * E-mail:
| |
Collapse
|
32
|
Ohuchi M, Fujino K, Kishimoto T, Yamane T, Hamamoto T, Tabata T, Tsujita Y, Matsushita M, Takahashi K, Matsumura K, Eguchi Y. Association of the Plasma Platelet-Derived Microparticles to Platelet Count Ratio with Hospital Mortality and Disseminated Intravascular Coagulopathy in Critically Ill Patients. J Atheroscler Thromb 2015; 22:773-82. [PMID: 25864886 DOI: 10.5551/jat.29439] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
AIM The role of platelet-derived microparticles (PDMPs) in the crosstalk between coagulopathy and inflammation in critically ill patients remains unclear. The aim of this cohort observational study was to investigate the associations between the PDMP levels and hospital mortality or disseminated intravascular coagulopathy (DIC). METHODS This study included 119 patients who were admitted to the ICU. The PDMP levels were measured using an enzyme-linked immunosorbent assay three times a week, for a total of 372 samples. We calculated the maximum (max) PDMP value, max PDMP/platelet (PDMP/Plts) ratio (converted to the PDMP levels per 10(4) platelets) and nadir platelet count during the ICU stay. Baseline patient data and scores, including the Japanese Association for Acute Medicine (JAAM) DIC score, were collected, and potential predictors were analyzed for possible associations with hospital mortality. RESULTS The max PDMP/Plts ratio was significantly different comparing the survivors (n=98: median, 2.54) and non-survivors (n=21: median 17.59; p<0.001). There was a weak but statistically significant negative correlation between the max PDMP level and nadir platelet count (r=-0.332, p<0.001). The max PDMP level and max PDMP/Plts ratio were higher in the DIC group (81.48 and 9.27, respectively) than in the non-DIC group (34.88 and 2.35, p=0.001 and p<0.001, respectively). The max PDMP/Plts ratio was the only variable found to be independently associated with hospital mortality according to a multivariate logistic regression analysis. CONCLUSIONS PDMPs are involved in the development of DIC but are not related to hospital mortality. There is a good association between the PDMP/Plts ratio and hospital mortality and/or DIC in critically ill patients.
Collapse
Affiliation(s)
- Masatsugu Ohuchi
- Department of Critical and Intensive Medicine, Shiga University of Medical Science
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Horn P, Stern D, Veulemans V, Heiss C, Zeus T, Merx MW, Kelm M, Westenfeld R. Improved endothelial function and decreased levels of endothelium-derived microparticles after transcatheter aortic valve implantation. EUROINTERVENTION 2015; 10:1456-63. [DOI: 10.4244/eijy14m10_02] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
34
|
França CN, Izar MCDO, Amaral JBD, Tegani DM, Fonseca FAH. Microparticles as potential biomarkers of cardiovascular disease. Arq Bras Cardiol 2015; 104:169-74. [PMID: 25626759 PMCID: PMC4375661 DOI: 10.5935/abc.20140210] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 10/21/2014] [Indexed: 12/26/2022] Open
Abstract
Primary prevention of cardiovascular disease is a choice of great relevance because
of its impact on health. Some biomarkers, such as microparticles derived from
different cell populations, have been considered useful in the assessment of
cardiovascular disease. Microparticles are released by the membrane structures of
different cell types upon activation or apoptosis, and are present in the plasma of
healthy individuals (in levels considered physiological) and in patients with
different pathologies. Many studies have suggested an association between
microparticles and different pathological conditions, mainly the relationship with
the development of cardiovascular diseases. Moreover, the effects of different
lipid-lowering therapies have been described in regard to measurement of
microparticles. The studies are still controversial regarding the levels of
microparticles that can be considered pathological. In addition, the methodologies
used still vary, suggesting the need for standardization of the different protocols
applied, aiming at using microparticles as biomarkers in clinical practice.
Collapse
|
35
|
Ferreira CES, França CN, Izar MCO, Camargo LM, Roman RM, Fonseca FAH. High-intensity statin monotherapy versus moderate-intensity statin plus ezetimibe therapy: effects on vascular biomarkers. Int J Cardiol 2014; 180:78-9. [PMID: 25438220 DOI: 10.1016/j.ijcard.2014.11.177] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2014] [Accepted: 11/23/2014] [Indexed: 01/05/2023]
Affiliation(s)
- C E S Ferreira
- Department of Medicine, Cardiology Division, Federal University of Sao Paulo, Sao Paulo, Brazil; Albert Einstein Israeli Hospital, Sao Paulo, Brazil
| | - C N França
- Department of Medicine, Cardiology Division, Federal University of Sao Paulo, Sao Paulo, Brazil
| | - M C O Izar
- Department of Medicine, Cardiology Division, Federal University of Sao Paulo, Sao Paulo, Brazil
| | - L M Camargo
- Department of Medicine, Cardiology Division, Federal University of Sao Paulo, Sao Paulo, Brazil
| | - R M Roman
- Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - F A H Fonseca
- Department of Medicine, Cardiology Division, Federal University of Sao Paulo, Sao Paulo, Brazil.
| |
Collapse
|
36
|
Wekesa AL, Cross KS, O'Donovan O, Dowdall JF, O'Brien O, Doyle M, Byrne L, Phelan JP, Ross MD, Landers R, Harrison M. Predicting carotid artery disease and plaque instability from cell-derived microparticles. Eur J Vasc Endovasc Surg 2014; 48:489-95. [PMID: 25218652 DOI: 10.1016/j.ejvs.2014.08.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Accepted: 08/01/2014] [Indexed: 11/18/2022]
Abstract
OBJECTIVES Cell-derived microparticles (MPs) are small plasma membrane-derived vesicles shed from circulating blood cells and may act as novel biomarkers of vascular disease. We investigated the potential of circulating MPs to predict (a) carotid plaque instability and (b) the presence of advanced carotid disease. METHODS This pilot study recruited carotid disease patients (aged 69.3 ± 1.2 years [mean ± SD], 69% male, 90% symptomatic) undergoing endarterectomy (n = 42) and age- and sex-matched controls (n = 73). Plaques were classified as stable (n = 25) or unstable (n = 16) post surgery using immunohistochemistry. Blood samples were analysed for MP subsets and molecular biomarkers. Odds ratios (OR) are expressed per standard deviation biomarker increase. RESULTS Endothelial MP (EMP) subsets, but not any vascular, inflammatory, or proteolytic molecular biomarker, were higher (p < .05) in the unstable than the stable plaque patients. The area under the receiver operator characteristic curve for CD31(+)41(-) EMP in discriminating an unstable plaque was 0.73 (0.56-0.90, p < .05). CD31(+)41(-) EMP predicted plaque instability (OR = 2.19, 1.08-4.46, p < .05) and remained significant in a multivariable model that included transient ischaemic attack symptom status. Annexin V(+) MP, platelet MP (PMP) subsets, and C-reactive protein were higher (p < .05) in cases than controls. Annexin V(+) MP (OR = 3.15, 1.49-6.68), soluble vascular cell adhesion molecule-1 (OR = 1.64, 1.03-2.59), and previous smoking history (OR = 3.82, 1.38-10.60) independently (p < .05) predicted the presence of carotid disease in a multivariable model. CONCLUSIONS EMP may have utility in predicting plaque instability in carotid patients and annexin V(+) MPs may predict the presence of advanced carotid disease in aging populations, independent of established biomarkers.
Collapse
Affiliation(s)
- A L Wekesa
- Biomedical Research Group, Schools of Health Science and Science, Waterford Institute of Technology, Waterford, Ireland
| | - K S Cross
- Departments of Vascular Surgery and Histopathology, Waterford Regional Hospital, Waterford, Ireland; Royal College of Surgeons in Ireland, Dublin, Ireland
| | - O O'Donovan
- Biomedical Research Group, Schools of Health Science and Science, Waterford Institute of Technology, Waterford, Ireland
| | - J F Dowdall
- Departments of Vascular Surgery and Histopathology, Waterford Regional Hospital, Waterford, Ireland
| | - O O'Brien
- Departments of Vascular Surgery and Histopathology, Waterford Regional Hospital, Waterford, Ireland
| | - M Doyle
- Departments of Vascular Surgery and Histopathology, Waterford Regional Hospital, Waterford, Ireland
| | - L Byrne
- Departments of Vascular Surgery and Histopathology, Waterford Regional Hospital, Waterford, Ireland
| | - J P Phelan
- Biomedical Research Group, Schools of Health Science and Science, Waterford Institute of Technology, Waterford, Ireland
| | - M D Ross
- Biomedical Research Group, Schools of Health Science and Science, Waterford Institute of Technology, Waterford, Ireland
| | - R Landers
- Departments of Vascular Surgery and Histopathology, Waterford Regional Hospital, Waterford, Ireland
| | - M Harrison
- Biomedical Research Group, Schools of Health Science and Science, Waterford Institute of Technology, Waterford, Ireland.
| |
Collapse
|
37
|
Choi JL, Li S, Han JY. Platelet function tests: a review of progresses in clinical application. BIOMED RESEARCH INTERNATIONAL 2014; 2014:456569. [PMID: 24895576 PMCID: PMC4034486 DOI: 10.1155/2014/456569] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/02/2014] [Accepted: 04/25/2014] [Indexed: 01/03/2023]
Abstract
The major goal of traditional platelet function tests has been to screen and diagnose patients who present with bleeding problems. However, as the central role of platelets implicated in the etiology of arterial thrombotic diseases such as myocardial infarction and stroke became widely known, platelet function tests are now being promoted to monitor the efficacy of antiplatelet drugs and also to potentially identify patients at increased risk of thrombosis. Beyond hemostasis and thrombosis, an increasing number of studies indicate that platelets play an integral role in intercellular communication, are mediators of inflammation, and have immunomodulatory activity. As new potential biomarkers and technologies arrive at the horizon, platelet functions testing appears to take on a new aspect. This review article discusses currently available clinical application of platelet function tests, placing emphasis on essential characteristics.
Collapse
Affiliation(s)
- Jae-Lim Choi
- Department of Laboratory Medicine, Dong-A University College of Medicine, 1,3-Ga, Dongdaesin-dong, Seo-gu, Busan 602-715, Republic of Korea
| | - Shuhua Li
- Department of Laboratory Medicine, Dong-A University College of Medicine, 1,3-Ga, Dongdaesin-dong, Seo-gu, Busan 602-715, Republic of Korea
| | - Jin-Yeong Han
- Department of Laboratory Medicine, Dong-A University College of Medicine, 1,3-Ga, Dongdaesin-dong, Seo-gu, Busan 602-715, Republic of Korea
| |
Collapse
|
38
|
Camargo LM, França CN, Izar MC, Bianco HT, Lins LS, Barbosa SP, Pinheiro LF, Fonseca FAH. Effects of simvastatin/ezetimibe on microparticles, endothelial progenitor cells and platelet aggregation in subjects with coronary heart disease under antiplatelet therapy. ACTA ACUST UNITED AC 2014; 47:432-7. [PMID: 24760119 PMCID: PMC4075313 DOI: 10.1590/1414-431x20143628] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Accepted: 02/20/2014] [Indexed: 01/14/2023]
Abstract
It is not known whether the addition of ezetimibe to statins adds cardiovascular
protection beyond the expected changes in lipid levels. Subjects with coronary heart
disease were treated with four consecutive 1-week courses of therapy (T) and
evaluations. The courses were: T1, 100 mg aspirin alone; T2, 100 mg aspirin and 40 mg
simvastatin/10 mg ezetimibe; T3, 40 mg simvastatin/10 mg ezetimibe, and 75 mg
clopidogrel (300 mg initial loading dose); T4, 75 mg clopidogrel alone. Platelet
aggregation was examined in whole blood. Endothelial microparticles (CD51), platelet
microparticles (CD42/CD31), and endothelial progenitor cells (CD34/CD133;
CDKDR/CD133, or CD34/KDR) were quantified by flow cytometry. Endothelial function was
examined by flow-mediated dilation. Comparisons between therapies revealed
differences in lipids (T2 and T3<T1 and T4 for total cholesterol, LDL-C, and
triglycerides; P<0.002 for all), as well as for endothelial function (T2>T1 and
T4, P=0.001). Decreased platelet aggregation was observed after aspirin (arachidonic
acid, T1<T3 and T4, P=0.034) and clopidogrel (adenosine, T3 and T4<T1 and T2,
P<0.0001) therapy. Simvastatin/ezetimibe diphosphate did not change platelet
aggregation, the amount of circulating endothelial and platelet microparticles, or
endothelial progenitor cells. Cardiovascular protection following therapy with
simvastatin/ezetimibe seems restricted to lipid changes and improvement of
endothelial function not affecting the release of microparticles, mobilization of
endothelial progenitor cells or decreased platelet aggregation.
Collapse
Affiliation(s)
- L M Camargo
- Departamento de Medicina, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brasil
| | - C N França
- Departamento de Medicina, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brasil
| | - M C Izar
- Departamento de Medicina, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brasil
| | - H T Bianco
- Departamento de Medicina, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brasil
| | - L S Lins
- Departamento de Medicina, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brasil
| | - S P Barbosa
- Departamento de Medicina, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brasil
| | - L F Pinheiro
- Departamento de Medicina, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brasil
| | - F A H Fonseca
- Departamento de Medicina, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brasil
| |
Collapse
|
39
|
Effects of Ezetimibe on Endothelial Progenitor Cells and Microparticles in High-Risk Patients. Cell Biochem Biophys 2014; 70:687-96. [DOI: 10.1007/s12013-014-9973-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
40
|
Schiro A, Wilkinson FL, Weston R, Smyth JV, Serracino-Inglott F, Alexander MY. Endothelial microparticles as conveyors of information in atherosclerotic disease. Atherosclerosis 2014; 234:295-302. [PMID: 24721189 DOI: 10.1016/j.atherosclerosis.2014.03.019] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Revised: 03/18/2014] [Accepted: 03/19/2014] [Indexed: 12/19/2022]
Abstract
Endothelial microparticles (EMPs) are complex submicron membrane-shed vesicles released into the circulation following endothelium cell activation or apoptosis. They are classified as either physiological or pathological, with anticoagulant or pro-inflammatory effects respectively. Endothelial dysfunction caused by inflammation is a key initiating event in atherosclerotic plaque formation. Athero-emboli, resulting from ruptured carotid plaques are a major cause of stroke. Current clinical techniques for arterial assessment, angiography and carotid ultrasound, give accurate information about stenosis but limited evidence on plaque composition, inflammation or vulnerability; as a result, patients with asymptomatic, or fragile carotid lesions, may not be identified and treated effectively. There is a need to discover novel biomarkers and develop more efficient diagnostic approaches in order to stratify patients at most risk of stroke, who would benefit from interventional surgery. Increasing evidence suggests that EMPs play an important role in the pathogenesis of cardiovascular disease, acting as a marker of damage, either exacerbating disease progression or triggering a repair response. In this regard, it has been suggested that EMPs have the potential to act as biomarkers of disease status. In this review, we will present the evidence to support this hypothesis and propose a novel concept for the development of a diagnostic device that could be implemented in the clinic.
Collapse
Affiliation(s)
- A Schiro
- Regional Vascular and Endovascular Unit, Manchester Royal Infirmary, Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Oxford Road, Manchester M13 9WL, UK; Cardiovascular Research Institute, Manchester Academic Health Science Centre, University of Manchester, Core Technology Facility, 46 Grafton Street, Manchester M13 9MT, UK.
| | - F L Wilkinson
- Translational Science, Healthcare Science Research Institute, Faculty of Science and Engineering, Manchester Metropolitan University, John Dalton Building, Chester Street, Manchester M1 5GD, UK
| | - R Weston
- Translational Science, Healthcare Science Research Institute, Faculty of Science and Engineering, Manchester Metropolitan University, John Dalton Building, Chester Street, Manchester M1 5GD, UK
| | - J V Smyth
- Regional Vascular and Endovascular Unit, Manchester Royal Infirmary, Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Oxford Road, Manchester M13 9WL, UK
| | - F Serracino-Inglott
- Regional Vascular and Endovascular Unit, Manchester Royal Infirmary, Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Oxford Road, Manchester M13 9WL, UK; Cardiovascular Research Institute, Manchester Academic Health Science Centre, University of Manchester, Core Technology Facility, 46 Grafton Street, Manchester M13 9MT, UK
| | - M Y Alexander
- Cardiovascular Research Institute, Manchester Academic Health Science Centre, University of Manchester, Core Technology Facility, 46 Grafton Street, Manchester M13 9MT, UK; Translational Science, Healthcare Science Research Institute, Faculty of Science and Engineering, Manchester Metropolitan University, John Dalton Building, Chester Street, Manchester M1 5GD, UK
| |
Collapse
|
41
|
Kuzniatsova N, Balakrishnan B, Lip GYH, Blann AD. No effect of clopidogrel activity or cessation on vascular function or markers of inflammation. Int J Angiol 2013; 21:195-200. [PMID: 24293976 DOI: 10.1055/s-0032-1328777] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
The platelet adenosine diphosphate (ADP)-receptor blocker clopidogrel is effective in reducing the rate of thrombosis in cardiovascular disease, but may also have nonplatelet activity. However, there is variability in the suppression of platelet function in individuals, leading to the concept of clopidogrel resistance, that is, reduced platelet-suppressing activity. We tested the hypothesis that some of the beneficial effect of clopidogrel may be due to the variable activity of this drug on the vascular system (assessed by plasma markers von Willebrand factor and soluble E-selectin, and functional arterial pulse wave velocity) and inflammation (C-reactive protein and interleukin-6) while 32 patients with coronary artery disease taking 75 mg clopidogrel daily, and again 2 weeks after cessation of clopidogrel therapy. Platelet responsiveness to clopidogrel was assessed by the phosphorylation of intracellular regulatory protein-vasodilator-stimulated phosphoprotein method and aggregometry to ADP. Response to aspirin was assessed using arachidonic acid (AA), and soluble P-selectin and PAC-1 were also measured. While on clopidogrel, there were no relationships between any vascular or inflammatory index and the response to clopidogrel. After stopping clopidogrel, there were no differences in platelet aggregation to AA, or the expression of P-selectin or PAC-1 at rest, or after stimulation by AA, but platelet responses to ADP all increased (p < 0.01). Although soluble P-selectin increased when clopidogrel was stopped (p = 0.006), there were no changes in plasma markers or vascular function. We conclude that 75 mg/day clopidogrel has no effect of markers of vascular function or inflammation.
Collapse
Affiliation(s)
- Nadja Kuzniatsova
- University of Birmingham Centre for Cardiovascular Sciences, City Hospital, Birmingham, United Kingdom
| | | | | | | |
Collapse
|
42
|
Hsu CY, Huang PH, Chiang CH, Leu HB, Huang CC, Chen JW, Lin SJ. Increased circulating endothelial apoptotic microparticle to endothelial progenitor cell ratio is associated with subsequent decline in glomerular filtration rate in hypertensive patients. PLoS One 2013; 8:e68644. [PMID: 23874701 PMCID: PMC3709900 DOI: 10.1371/journal.pone.0068644] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Accepted: 05/31/2013] [Indexed: 02/07/2023] Open
Abstract
Background Recent research indicates hypertensive patients with microalbuminuria have decreased endothelial progenitor cells (EPCs) and increased levels of endothelial apoptotic microparticles (EMP). However, whether these changes are related to a subsequent decline in glomerular filtration rate (GFR) remains unclear. Methods and Results We enrolled totally 100 hypertensive out-patients with eGFR ≥30 mL/min/1.73 m2. The mean annual rate of GFR decline (△GFR/y) was −1.49±3.26 mL/min/1.73 m2 per year during the follow-up period (34±6 months). Flow cytometry was used to assess circulating EPC (CD34+/KDR+) and EMP levels (CD31+/annexin V+) in peripheral blood. The △GFR/y was correlated with the EMP to EPC ratio (r = −0.465, p<0.001), microalbuminuria (r = −0.329, p = 0.001), and the Framingham risk score (r = −0.245, p = 0.013). When we divided the patients into 4 groups according to the EMP to EPC ratio, there was an association between the EMP to EPC ratio and the ΔGFR/y (mean ΔGFR/y: 0.08±3.04 vs. −0.50±2.84 vs. −1.25±2.49 vs. −4.42±2.82, p<0.001). Multivariate analysis indicated that increased EMP to EPC ratio is an independent predictor of ΔeGFR/y. Conclusions An increased circulating EMP to EPC ratio is associated with subsequent decline in GFR in hypertensive patients, which suggests endothelial damage with reduced vascular repair capacity may contribute to further deterioration of renal function in patients with hypertension.
Collapse
Affiliation(s)
- Chien-Yi Hsu
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Po-Hsun Huang
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
- Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan
- * E-mail:
| | - Chia-Hung Chiang
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
- Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Hsin-Bang Leu
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Healthcare and Management Center, Taipei Veterans General Hospital, Taipei, Taiwan
- Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Chin-Chou Huang
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Medical Research and Education, Taipei Veterans General Hospital, Taipei, Taiwan
- Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan
- Institute and Department of Pharmacology, National Yang-Ming University, Taipei, Taiwan
| | - Jaw-Wen Chen
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Medical Research and Education, Taipei Veterans General Hospital, Taipei, Taiwan
- Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan
- Institute and Department of Pharmacology, National Yang-Ming University, Taipei, Taiwan
| | - Shing-Jong Lin
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Medical Research and Education, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
- Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
43
|
Luo JC, Huo TI, Hou MC, Lin HY, Li CP, Lin HC, Chang FY, Lee FY. Clopidogrel delays gastric ulcer healing in rats. Eur J Pharmacol 2012; 695:112-9. [PMID: 22975710 DOI: 10.1016/j.ejphar.2012.07.054] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2012] [Revised: 07/10/2012] [Accepted: 07/27/2012] [Indexed: 01/05/2023]
Abstract
Clopidogrel is not safe enough for the gastric mucosa in patients with high risk of peptic ulcer. This study aimed to explore if clopidogrel delays gastric ulcer healing and elucidate the involved mechanisms. Gastric ulcer was induced in rats and the ulcer size, mucosal epithelial cell proliferation of the ulcer margin, expression of growth factors [epidermal growth factor (EGF), basic fibroblast growth factor] and their receptors, and signal transduction pathways for cell proliferation were measured and compared between the clopidogrel-treated group and untreated controls. For the in vitro part, rat gastric mucosal epithelial cell line (RGM-1 cells) was used to establish EGF receptor over-expressed cells. Cell proliferation and molecular change under EGF treatment (10ng/ml) with and without clopidogrel (10(-6)M) were demonstrated. Ulcer size was significantly larger in the clopidogrel-treated group compared to the control and mucosal epithelial cell proliferation of the ulcer margin was significantly decreased in the clopidogrel-treated group (P<0.05). Clopidogrel (2mg and 10mg/kg/day) significantly decreased ulcer-induced gastric epithelial cell proliferation and ulcer-stimulated expressions of EGF receptor and phosphorylated extracellular signal-regulated kinase (PERK) at the ulcer margin (P<0.05). Clopidogrel (10(-6)M) also inhibited EGF-stimulated EGF receptor, PERK expression, and cell proliferation in RGM-1 cells (P<0.05), and caused much less inhibition of EGF-stimulated cell proliferation in EGF receptor over-expressed RGM-1 cells than in RGM-1 cells (22% vs. 32% reduction). In conclusion, clopidogrel delays gastric ulcer healing in rats via inhibiting gastric epithelial cell proliferation, at least by inhibition of the EGF receptor-ERK signal transduction pathway.
Collapse
Affiliation(s)
- Jiing-Chyuan Luo
- Department of Medicine, National Yang-Ming University, School of Medicine, No. 155 Section 2 Linong Street, Taipei, Taiwan.
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Sheu JJ, Lin KC, Tsai CY, Tsai TH, Leu S, Yen CH, Chen YL, Chang HW, Sun CK, Chua S, Yang JL, Yip HK. Combination of cilostazol and clopidogrel attenuates rat critical limb ischemia. J Transl Med 2012; 10:164. [PMID: 22897925 PMCID: PMC3479044 DOI: 10.1186/1479-5876-10-164] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2012] [Accepted: 07/25/2012] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND AND AIM Procedural failure and untoward clinical outcomes after surgery remain problematic in critical limb ischemia (CLI) patients. This study tested a clopidogrel-cilostazol combination treatment compared with either treatment alone in attenuating CLI and improving CLI-region blood flow in rats. METHODS Male Sprague-Dawley rats (n = 40) were equally divided into five groups: control, CLI induction only, CL I + cilostazol (12.0 mg/day/kg), CLI + clopidogrel (8.0 mg/kg/day) and CLI + combined cilostazol-clopidogrel. After treatment for 21 days, Laser Doppler imaging was performed. RESULTS On day 21, the untreated CLI group had the lowest ratio of ischemic/normal blood flow (p < 0.001). Inflammation measured by VCAM-1 protein expression; oxidative stress; PAI-1, MMP-9 and TNF-α mRNA expressions; and immunofluorescence staining (IF) of CD68+ cells was lower with combined treatment than with the other treatments, and lower in the two single-treatment groups than the untreated CLI group (all p < 0.01). Anti-inflammatory mRNA expression of interleukin-10, and eNOS showed a reverse pattern among these groups. Apoptosis measured by Bax, caspase-3 and PARP; and muscle damage measured by cytosolic cytochrome-C, and serum and muscle micro-RNA-206 were all lowest with combination treatment, and the two single-treatment groups showed lower values than the untreated group (all p < 0.001). Angiogenesis measured by eNOS, IF staining of CD31+ and vWF + cells; and number of vessels in CLI region were highest with combination treatment and higher in the single-treatment groups than the untreated group (all p < 0.001). CONCLUSION Combined cilostazol-clopidogrel therapy is superior to either agent alone in improving ischemia in rodent CLI.
Collapse
Affiliation(s)
- Jiunn-Jye Sheu
- Division of Thoracic and Cardiovascular Surgery, Chang Gung Memorial Hospital- Kaohsiung Medical Center, Chang Gung University College of Medicine, Gueishan, Taiwan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|