51
|
Wang L, Tang C. Targeting Platelet in Atherosclerosis Plaque Formation: Current Knowledge and Future Perspectives. Int J Mol Sci 2020; 21:ijms21249760. [PMID: 33371312 PMCID: PMC7767086 DOI: 10.3390/ijms21249760] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/09/2020] [Accepted: 12/16/2020] [Indexed: 12/23/2022] Open
Abstract
Besides their role in hemostasis and thrombosis, it has become increasingly clear that platelets are also involved in many other pathological processes of the vascular system, such as atherosclerotic plaque formation. Atherosclerosis is a chronic vascular inflammatory disease, which preferentially develops at sites under disturbed blood flow with low speeds and chaotic directions. Hyperglycemia, hyperlipidemia, and hypertension are all risk factors for atherosclerosis. When the vascular microenvironment changes, platelets can respond quickly to interact with endothelial cells and leukocytes, participating in atherosclerosis. This review discusses the important roles of platelets in the plaque formation under pro-atherogenic factors. Specifically, we discussed the platelet behaviors under disturbed flow, hyperglycemia, and hyperlipidemia conditions. We also summarized the molecular mechanisms involved in vascular inflammation during atherogenesis based on platelet receptors and secretion of inflammatory factors. Finally, we highlighted the studies of platelet migration in atherogenesis. In general, we elaborated an atherogenic role of platelets and the aspects that should be further studied in the future.
Collapse
Affiliation(s)
- Lei Wang
- Cyrus Tang Hematology Center, Cyrus Tang Medical Institute, Soochow University, Suzhou 215123, China;
| | - Chaojun Tang
- Cyrus Tang Hematology Center, Cyrus Tang Medical Institute, Soochow University, Suzhou 215123, China;
- Collaborative Innovation Center of Hematology of Jiangsu Province, Soochow University, Suzhou 215123, China
- National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou 215123, China
- Correspondence: ; Tel.: +86-512-6588-0899
| |
Collapse
|
52
|
Harbi MH, Smith CW, Nicolson PLR, Watson SP, Thomas MR. Novel antiplatelet strategies targeting GPVI, CLEC-2 and tyrosine kinases. Platelets 2020; 32:29-41. [PMID: 33307909 DOI: 10.1080/09537104.2020.1849600] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Antiplatelet medications comprise the cornerstone of treatment for diseases that involve arterial thrombosis, including acute coronary syndromes (ACS), stroke and peripheral arterial disease. However, antiplatelet medications may cause bleeding and, furthermore, thrombotic events may still recur despite treatment. The interaction of collagen with GPVI receptors on the surface of platelets has been identified as one of the major players in the pathophysiology of arterial thrombosis that occurs following atherosclerotic plaque rupture. Promisingly, GPVI deficiency in humans appears to have a minimal impact on bleeding. These findings together suggest that targeting platelet GPVI may provide a novel treatment strategy that provides additional antithrombotic efficacy with minimal disruption of normal hemostasis compared to conventional antiplatelet medications. CLEC-2 is gaining interest as a therapeutic target for a variety of thrombo-inflammatory disorders including deep vein thrombosis (DVT) with treatment also predicted to cause minimal disruption to hemostasis. GPVI and CLEC-2 signal through Src, Syk and Tec family tyrosine kinases, providing additional strategies for inhibiting both receptors. In this review, we summarize the evidence regarding GPVI and CLEC-2 and strategies for inhibiting these receptors to inhibit platelet recruitment and activation in thrombotic diseases.
Collapse
Affiliation(s)
- Maan H Harbi
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham , Birmingham, UK
| | - Christopher W Smith
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham , Birmingham, UK
| | - Phillip L R Nicolson
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham , Birmingham, UK.,University Hospitals Birmingham NHS Foundation Trust , Birmingham, UK
| | - Steve P Watson
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham , Birmingham, UK
| | - Mark R Thomas
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham , Birmingham, UK.,University Hospitals Birmingham NHS Foundation Trust , Birmingham, UK.,Sandwell and West Birmingham NHS Trust , Birmingham, UK
| |
Collapse
|
53
|
Perrella G, Huang J, Provenzale I, Swieringa F, Heubel-Moenen FCJI, Farndale RW, Roest M, van der Meijden PEJ, Thomas M, Ariëns RAS, Jandrot-Perrus M, Watson SP, Heemskerk JWM. Nonredundant Roles of Platelet Glycoprotein VI and Integrin αIIbβ3 in Fibrin-Mediated Microthrombus Formation. Arterioscler Thromb Vasc Biol 2020; 41:e97-e111. [PMID: 33267658 DOI: 10.1161/atvbaha.120.314641] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Fibrin is considered to strengthen thrombus formation via integrin αIIbβ3, but recent findings indicate that fibrin can also act as ligand for platelet glycoprotein VI. Approach and Results: To investigate the thrombus-forming potential of fibrin and the roles of platelet receptors herein, we generated a range of immobilized fibrin surfaces, some of which were cross-linked with factor XIIIa and contained VWF-BP (von Willebrand factor-binding peptide). Multicolor microfluidics assays with whole-blood flowed at high shear rate (1000 s-1) indicated that the fibrin surfaces, regardless of the presence of factor XIIIa or VWF-BP, supported platelet adhesion and activation (P-selectin expression), but only microthrombi were formed consisting of bilayers of platelets. Fibrinogen surfaces produced similar microthrombi. Markedly, tiggering of coagulation with tissue factor or blocking of thrombin no more than moderately affected the fibrin-induced microthrombus formation. Absence of αIIbβ3 in Glanzmann thrombasthenia annulled platelet adhesion. Blocking of glycoprotein VI with Fab 9O12 substantially, but incompletely reduced platelet secretion, Ca2+ signaling and aggregation, while inhibition of Syk further reduced these responses. In platelet suspension, glycoprotein VI blockage or Syk inhibition prevented fibrin-induced platelet aggregation. Microthrombi on fibrin surfaces triggered only minimal thrombin generation, in spite of thrombin binding to the fibrin fibers. CONCLUSIONS Together, these results indicate that fibrin fibers, regardless of their way of formation, act as a consolidating surface in microthrombus formation via nonredundant roles of platelet glycoprotein VI and integrin αIIbβ3 through signaling via Syk and low-level Ca2+ rises.
Collapse
Affiliation(s)
- Gina Perrella
- Department of Biochemistry, CARIM, Maastricht University, the Netherlands (G.P., J.H., I.P., F.S., P.E.J.v.d.M., R.A.S.A., S.P.W., J.W.M.H.)
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, United Kingdom (G.P., M.T., S.P.W.)
| | - Jingnan Huang
- Department of Biochemistry, CARIM, Maastricht University, the Netherlands (G.P., J.H., I.P., F.S., P.E.J.v.d.M., R.A.S.A., S.P.W., J.W.M.H.)
- ISAS Institute, Dortmund, DE (J.H.)
| | - Isabella Provenzale
- Department of Biochemistry, CARIM, Maastricht University, the Netherlands (G.P., J.H., I.P., F.S., P.E.J.v.d.M., R.A.S.A., S.P.W., J.W.M.H.)
| | - Frauke Swieringa
- Department of Biochemistry, CARIM, Maastricht University, the Netherlands (G.P., J.H., I.P., F.S., P.E.J.v.d.M., R.A.S.A., S.P.W., J.W.M.H.)
| | | | - Richard W Farndale
- Department of Biochemistry, University of Cambridge, United Kingdom (R.W.F.)
| | - Mark Roest
- Synapse Research Institute, Maastricht, the Netherlands (M.R.)
| | - Paola E J van der Meijden
- Department of Biochemistry, CARIM, Maastricht University, the Netherlands (G.P., J.H., I.P., F.S., P.E.J.v.d.M., R.A.S.A., S.P.W., J.W.M.H.)
| | - Mark Thomas
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, United Kingdom (G.P., M.T., S.P.W.)
| | - Robert A S Ariëns
- Department of Biochemistry, CARIM, Maastricht University, the Netherlands (G.P., J.H., I.P., F.S., P.E.J.v.d.M., R.A.S.A., S.P.W., J.W.M.H.)
- Department of Discovery and Translational Science, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (R.A.S.A.)
| | - Martine Jandrot-Perrus
- UMR S1148, Laboratory for Vascular Translational Science, INSERM, University Paris Diderot, France (M.J.-P.)
| | - Steve P Watson
- Department of Biochemistry, CARIM, Maastricht University, the Netherlands (G.P., J.H., I.P., F.S., P.E.J.v.d.M., R.A.S.A., S.P.W., J.W.M.H.)
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, United Kingdom (G.P., M.T., S.P.W.)
- COMPARE, The Universities of Birmingham and Nottingham, the Midlands, United Kingdom (S.P.W.)
| | - Johan W M Heemskerk
- Department of Biochemistry, CARIM, Maastricht University, the Netherlands (G.P., J.H., I.P., F.S., P.E.J.v.d.M., R.A.S.A., S.P.W., J.W.M.H.)
| |
Collapse
|
54
|
Janus-Bell E, Ahmed MU, Receveur N, Mouriaux C, Nieswandt B, Gardiner EE, Gachet C, Jandrot-Perrus M, Mangin PH. Differential Role of Glycoprotein VI in Mouse and Human Thrombus Progression and Stability. Thromb Haemost 2020; 121:543-546. [PMID: 33124026 DOI: 10.1055/s-0040-1718737] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Affiliation(s)
- Emily Janus-Bell
- Université de Strasbourg, INSERM, EFS Grand-Est, BPPS UMR-S1255, FMTS, F-67065 Strasbourg, France
| | - Muhammad Usman Ahmed
- Université de Strasbourg, INSERM, EFS Grand-Est, BPPS UMR-S1255, FMTS, F-67065 Strasbourg, France
| | - Nicolas Receveur
- Université de Strasbourg, INSERM, EFS Grand-Est, BPPS UMR-S1255, FMTS, F-67065 Strasbourg, France
| | - Clarisse Mouriaux
- Université de Strasbourg, INSERM, EFS Grand-Est, BPPS UMR-S1255, FMTS, F-67065 Strasbourg, France
| | - Bernhard Nieswandt
- Institute of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany
| | - Elizabeth E Gardiner
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Christian Gachet
- Université de Strasbourg, INSERM, EFS Grand-Est, BPPS UMR-S1255, FMTS, F-67065 Strasbourg, France
| | - Martine Jandrot-Perrus
- Université de Paris INSERM, Hôpital Bichat, Paris, France.,Acticor Biotech, Paris, France
| | - Pierre H Mangin
- Université de Strasbourg, INSERM, EFS Grand-Est, BPPS UMR-S1255, FMTS, F-67065 Strasbourg, France
| |
Collapse
|
55
|
Gupta S, Brass LF. Glycoprotein VI Blockade: Not Just Targeting Collagen Anymore? Arterioscler Thromb Vasc Biol 2020; 40:1964-1966. [PMID: 32845775 DOI: 10.1161/atvbaha.120.315059] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Shuchi Gupta
- From the Hematology-Oncology Division, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Lawrence F Brass
- From the Hematology-Oncology Division, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia
| |
Collapse
|
56
|
Krueger I, Gremer L, Mangels L, Klier M, Jurk K, Willbold D, Bock HH, Elvers M. Reelin Amplifies Glycoprotein VI Activation and AlphaIIb Beta3 Integrin Outside-In Signaling via PLC Gamma 2 and Rho GTPases. Arterioscler Thromb Vasc Biol 2020; 40:2391-2403. [PMID: 32787521 DOI: 10.1161/atvbaha.120.314902] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Reelin, a secreted glycoprotein, was originally identified in the central nervous system, where it plays an important role in brain development and maintenance. In the cardiovascular system, reelin plays a role in atherosclerosis by enhancing vascular inflammation and in arterial thrombosis by promoting platelet adhesion, activation, and thrombus formation via APP (amyloid precursor protein) and GP (glycoprotein) Ib. However, the role of reelin in hemostasis and arterial thrombosis is not fully understood to date. Approach and Results: In the present study, we analyzed the importance of reelin for cytoskeletal reorganization of platelets and thrombus formation in more detail. Platelets release reelin to amplify alphaIIb beta3 integrin outside-in signaling by promoting platelet adhesion, cytoskeletal reorganization, and clot retraction via activation of Rho GTPases RAC1 (Ras-related C3 botulinum toxin substrate) and RhoA (Ras homolog family member A). Reelin interacts with the collagen receptor GP (glycoprotein) VI with subnanomolar affinity, induces tyrosine phosphorylation in a GPVI-dependent manner, and supports platelet binding to collagen and GPVI-dependent RAC1 activation, PLC gamma 2 (1-phosphatidylinositol-4,5-bisphosphate phosphodiesterase gamma-2) phosphorylation, platelet activation, and aggregation. When GPVI was deleted from the platelet surface by antibody treatment in reelin-deficient mice, thrombus formation was completely abolished after injury of the carotid artery while being only reduced in either GPVI-depleted or reelin-deficient mice. CONCLUSIONS Our study identified a novel signaling pathway that involves reelin-induced GPVI activation and alphaIIb beta3 integrin outside-in signaling in platelets. Loss of both, GPVI and reelin, completely prevents stable arterial thrombus formation in vivo suggesting that inhibiting reelin-platelet-interaction might represent a novel strategy to avoid arterial thrombosis in cardiovascular disease.
Collapse
Affiliation(s)
- Irena Krueger
- Department of Vascular and Endovascular Surgery, Heinrich-Heine-University University Medical Center, Düsseldorf, Germany (I.K., M.K., M.E.)
| | - Lothar Gremer
- Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, Germany (L.G., D.W.).,Institute of Biological Information Processing (IBI-7: Structural Biochemistry) and JuStruct, Forschungszentrum Jülich, Germany (L.G., L.M., D.W.)
| | - Lena Mangels
- Institute of Biological Information Processing (IBI-7: Structural Biochemistry) and JuStruct, Forschungszentrum Jülich, Germany (L.G., L.M., D.W.)
| | - Meike Klier
- Department of Vascular and Endovascular Surgery, Heinrich-Heine-University University Medical Center, Düsseldorf, Germany (I.K., M.K., M.E.)
| | - Kerstin Jurk
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Germany (K.J.)
| | - Dieter Willbold
- Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, Germany (L.G., D.W.).,Institute of Biological Information Processing (IBI-7: Structural Biochemistry) and JuStruct, Forschungszentrum Jülich, Germany (L.G., L.M., D.W.)
| | - Hans H Bock
- Gastroenterology, Hepatology and Infectiology Department, Heinrich-Heine-University, Düsseldorf, Germany (H.H.B.)
| | - Margitta Elvers
- Department of Vascular and Endovascular Surgery, Heinrich-Heine-University University Medical Center, Düsseldorf, Germany (I.K., M.K., M.E.)
| |
Collapse
|
57
|
Donner L, Toska LM, Krüger I, Gröniger S, Barroso R, Burleigh A, Mezzano D, Pfeiler S, Kelm M, Gerdes N, Watson SP, Sun Y, Elvers M. The collagen receptor glycoprotein VI promotes platelet-mediated aggregation of β-amyloid. Sci Signal 2020; 13:13/643/eaba9872. [PMID: 32753479 DOI: 10.1126/scisignal.aba9872] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cerebral amyloid angiopathy (CAA) and β-amyloid (Aβ) deposition in the brain parenchyma are hallmarks of Alzheimer's disease (AD). We previously reported that platelets contribute to Aβ aggregation in cerebral vessels by secreting the factor clusterin upon binding of Aβ40 to the fibrinogen receptor integrin αIIbβ3 Here, we investigated the contribution of the collagen receptor GPVI (glycoprotein VI) in platelet-induced amyloid aggregation. Using platelets isolated from GPVI-wild type and GPVI-deficient human donors and mice, we found that Aβ40 bound to GPVI, which induced the release of ATP and fibrinogen, resulting in platelet aggregation. Binding of Aβ40 to integrin αIIbβ3, fibrinogen, and GPVI collectively contributed to the formation of amyloid clusters at the platelet surface. Consequently, blockade of αIIbβ3 or genetic loss of GPVI reduced amyloid fibril formation in cultured platelets and decreased the adhesion of Aβ-activated platelets to injured carotid arteries in mice. Application of losartan to inhibit collagen binding to GPVI resulted in decreased Aβ40-stimulated platelet activation, factor secretion, and platelet aggregation. Furthermore, the application of GPVI- or integrin-blocking antibodies reduced the formation of platelet-associated amyloid aggregates. Our findings indicate that Aβ40 promotes platelet-mediated amyloid aggregation by binding to both GPVI and integrin αIIbβ3 Blocking these pathways may therapeutically reduce amyloid plaque formation in cerebral vessels and the brain parenchyma of patients.
Collapse
Affiliation(s)
- Lili Donner
- Department of Vascular and Endovascular Surgery, Heinrich-Heine University Medical Center, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Laura Mara Toska
- Department of Vascular and Endovascular Surgery, Heinrich-Heine University Medical Center, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Irena Krüger
- Department of Vascular and Endovascular Surgery, Heinrich-Heine University Medical Center, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Sandra Gröniger
- Department of Vascular and Endovascular Surgery, Heinrich-Heine University Medical Center, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Ruben Barroso
- Institute of Cardiovascular Sciences, IBR Building, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Alice Burleigh
- Institute of Cardiovascular Sciences, IBR Building, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Diego Mezzano
- Department of Hematology and Oncology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330034, Chile
| | - Susanne Pfeiler
- Division of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich-Heine University, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Malte Kelm
- Division of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich-Heine University, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Norbert Gerdes
- Division of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich-Heine University, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Steve P Watson
- Institute of Cardiovascular Sciences, IBR Building, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK.,Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands B12 2TT, UK
| | - Yi Sun
- Institute of Cardiovascular Sciences, IBR Building, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK.,Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands B12 2TT, UK
| | - Margitta Elvers
- Department of Vascular and Endovascular Surgery, Heinrich-Heine University Medical Center, Moorenstrasse 5, 40225 Düsseldorf, Germany.
| |
Collapse
|
58
|
Zhou J, Yang RP, Song W, Xu HM, Wang YH. Antiplatelet Activity of Tussilagone via Inhibition of the GPVI Downstream Signaling Pathway in Platelets. Front Med (Lausanne) 2020; 7:380. [PMID: 32850895 PMCID: PMC7403204 DOI: 10.3389/fmed.2020.00380] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 06/19/2020] [Indexed: 12/22/2022] Open
Abstract
Tussilagone is a sesquiterpenoid extracted from Tussilago farfara and is used as an oriental medicine for asthma and bronchitis. Although previous studies have shown that tussilagone has an inhibitory effect on platelet aggregation, no studies have been performed to investigate its precise effect on platelets, and the underlying mechanism remains unclear. In the present study, we showed that tussilagone inhibited platelet aggregation induced by collagen, thrombin and ADP, as well as platelet release induced by collagen and thrombin, in mice. Tussilagone decreased P-selectin expression and αIIbβ3 activation (JON/A binding) in activated platelets, which indicated that tussilagone inhibited platelet activation. Moreover, tussilagone suppressed platelet spreading on fibrinogen and clot retraction. The levels of phosphorylated Syk, PLCγ2, Akt, GSK3β, and MAPK (ERK1/2 and P38) and molecules associated with GPVI downstream signaling were downregulated in the presence of tussilagone. In addition, tussilagone prolonged the occlusion time in a mouse model of FeCl3-induced carotid artery thrombosis and had no effect on mouse tail bleeding time. These results indicate that tussilagone inhibits platelet function in vitro and in vivo and that the underlying mechanism involves the Syk/PLCγ2-PKC/MAPK and PI3K-Akt-GSK3β signaling pathways downstream of GPVI. This research suggests that tussilagone is a potential candidate antiplatelet drug for the prevention of thrombosis.
Collapse
Affiliation(s)
- Jing Zhou
- Department of Pharmacy, Zhumadian Central Hospital, Zhumadian, China
| | - Ru-Ping Yang
- Department of Pharmacy, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - Wei Song
- Department of Pharmacy, Renmin Hospital, Wuhan University, Wuhan, China
| | - Hui-Min Xu
- Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yong-Hui Wang
- Department of Pharmacy, Zhumadian Central Hospital, Zhumadian, China
| |
Collapse
|
59
|
Ahmed MU, Kaneva V, Loyau S, Nechipurenko D, Receveur N, Le Bris M, Janus-Bell E, Didelot M, Rauch A, Susen S, Chakfé N, Lanza F, Gardiner EE, Andrews RK, Panteleev M, Gachet C, Jandrot-Perrus M, Mangin PH. Pharmacological Blockade of Glycoprotein VI Promotes Thrombus Disaggregation in the Absence of Thrombin. Arterioscler Thromb Vasc Biol 2020; 40:2127-2142. [PMID: 32698684 DOI: 10.1161/atvbaha.120.314301] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Atherothrombosis occurs upon rupture of an atherosclerotic plaque and leads to the formation of a mural thrombus. Computational fluid dynamics and numerical models indicated that the mechanical stress applied to a thrombus increases dramatically as a thrombus grows, and that strong inter-platelet interactions are essential to maintain its stability. We investigated whether GPVI (glycoprotein VI)-mediated platelet activation helps to maintain thrombus stability by using real-time video-microscopy. Approach and Results: We showed that GPVI blockade with 2 distinct Fab fragments promoted efficient disaggregation of human thrombi preformed on collagen or on human atherosclerotic plaque material in the absence of thrombin. ACT017-induced disaggregation was achieved under arterial blood flow conditions, and its effect increased with wall shear rate. GPVI regulated platelet activation within a growing thrombus as evidenced by the loss in thrombus contraction when GPVI was blocked, and the absence of the disaggregating effect of an anti-GPVI agent when the thrombi were fully activated with soluble agonists. The GPVI-dependent thrombus stabilizing effect was further supported by the fact that inhibition of any of the 4 key immunoreceptor tyrosine-based motif signalling molecules, src-kinases, Syk, PI3Kβ, or phospholipase C, resulted in kinetics of thrombus disaggregation similar to ACT017. The absence of ACT017-induced disaggregation of thrombi from 2 afibrinogenemic patients suggests that the role of GPVI requires interaction with fibrinogen. Finally, platelet disaggregation of fibrin-rich thrombi was also promoted by ACT017 in combination with r-tPA (recombinant tissue plasminogen activator). CONCLUSIONS This work identifies an unrecognized role for GPVI in maintaining thrombus stability and suggests that targeting GPVI could dissolve platelet aggregates with a poor fibrin content.
Collapse
Affiliation(s)
- Muhammad Usman Ahmed
- From the Université de Strasbourg, INSERM, EFS Grand-Est, BPPS UMR-S1255, FMTS, F-67065 Strasbourg, France (M.U.A., N.R., M.L.B., E.J.-B., F.L., C.G., P.H.M.)
| | - Valeria Kaneva
- Faculty of Physics, Moscow State University, Russia (V.K., D.N., M.P.).,Federal Research and Clinical Centre of Pediatric Hematology, Oncology and Immunology, Russia (V.K., D.N., M.P.).,Center for Theoretical Problems of Physicochemical Pharmacology, Russia (V.K., D.M., M.P.)
| | - Stéphane Loyau
- Université de Paris, INSERM, Hôpital Bichat, UMR-S1148, France (S.L., M.J.-P.)
| | - Dmitry Nechipurenko
- Faculty of Physics, Moscow State University, Russia (V.K., D.N., M.P.).,Federal Research and Clinical Centre of Pediatric Hematology, Oncology and Immunology, Russia (V.K., D.N., M.P.).,Center for Theoretical Problems of Physicochemical Pharmacology, Russia (V.K., D.M., M.P.)
| | - Nicolas Receveur
- From the Université de Strasbourg, INSERM, EFS Grand-Est, BPPS UMR-S1255, FMTS, F-67065 Strasbourg, France (M.U.A., N.R., M.L.B., E.J.-B., F.L., C.G., P.H.M.)
| | - Marion Le Bris
- From the Université de Strasbourg, INSERM, EFS Grand-Est, BPPS UMR-S1255, FMTS, F-67065 Strasbourg, France (M.U.A., N.R., M.L.B., E.J.-B., F.L., C.G., P.H.M.)
| | - Emily Janus-Bell
- From the Université de Strasbourg, INSERM, EFS Grand-Est, BPPS UMR-S1255, FMTS, F-67065 Strasbourg, France (M.U.A., N.R., M.L.B., E.J.-B., F.L., C.G., P.H.M.)
| | - Mélusine Didelot
- CHU Lille, Université de Lille, INSERM UMR-SU1011-EGID, Institut Pasteur de Lille, France (M.D., A.R., S.S.)
| | - Antoine Rauch
- CHU Lille, Université de Lille, INSERM UMR-SU1011-EGID, Institut Pasteur de Lille, France (M.D., A.R., S.S.)
| | - Sophie Susen
- CHU Lille, Université de Lille, INSERM UMR-SU1011-EGID, Institut Pasteur de Lille, France (M.D., A.R., S.S.)
| | - Nabil Chakfé
- Université de Strasbourg, Department of Vascular Surgery and Kidney Transplantation, France (N.C.)
| | - François Lanza
- From the Université de Strasbourg, INSERM, EFS Grand-Est, BPPS UMR-S1255, FMTS, F-67065 Strasbourg, France (M.U.A., N.R., M.L.B., E.J.-B., F.L., C.G., P.H.M.)
| | - Elizabeth E Gardiner
- The Australian National University, The John Curtin School of Medical Research, ACRF Department of Cancer Biology and Therapeutics, Canberra, Australia (E.E.G.)
| | - Robert K Andrews
- Australian Centre for Blood Diseases, Monash University, Australia (R.K.A.)
| | - Mikhail Panteleev
- Faculty of Physics, Moscow State University, Russia (V.K., D.N., M.P.).,Federal Research and Clinical Centre of Pediatric Hematology, Oncology and Immunology, Russia (V.K., D.N., M.P.).,Center for Theoretical Problems of Physicochemical Pharmacology, Russia (V.K., D.M., M.P.)
| | - Christian Gachet
- From the Université de Strasbourg, INSERM, EFS Grand-Est, BPPS UMR-S1255, FMTS, F-67065 Strasbourg, France (M.U.A., N.R., M.L.B., E.J.-B., F.L., C.G., P.H.M.)
| | - Martine Jandrot-Perrus
- Université de Paris, INSERM, Hôpital Bichat, UMR-S1148, France (S.L., M.J.-P.).,Acticor Biotech, France (M.J.-P.)
| | - Pierre H Mangin
- From the Université de Strasbourg, INSERM, EFS Grand-Est, BPPS UMR-S1255, FMTS, F-67065 Strasbourg, France (M.U.A., N.R., M.L.B., E.J.-B., F.L., C.G., P.H.M.)
| |
Collapse
|
60
|
Grobler C, Maphumulo SC, Grobbelaar LM, Bredenkamp JC, Laubscher GJ, Lourens PJ, Steenkamp J, Kell DB, Pretorius E. Covid-19: The Rollercoaster of Fibrin(Ogen), D-Dimer, Von Willebrand Factor, P-Selectin and Their Interactions with Endothelial Cells, Platelets and Erythrocytes. Int J Mol Sci 2020; 21:ijms21145168. [PMID: 32708334 PMCID: PMC7403995 DOI: 10.3390/ijms21145168] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 07/13/2020] [Accepted: 07/14/2020] [Indexed: 02/07/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-Cov-2), also known as coronavirus disease 2019 (COVID-19)-induced infection, is strongly associated with various coagulopathies that may result in either bleeding and thrombocytopenia or hypercoagulation and thrombosis. Thrombotic and bleeding or thrombotic pathologies are significant accompaniments to acute respiratory syndrome and lung complications in COVID-19. Thrombotic events and bleeding often occur in subjects with weak constitutions, multiple risk factors and comorbidities. Of particular interest are the various circulating inflammatory coagulation biomarkers involved directly in clotting, with specific focus on fibrin(ogen), D-dimer, P-selectin and von Willebrand Factor (VWF). Central to the activity of these biomarkers are their receptors and signalling pathways on endothelial cells, platelets and erythrocytes. In this review, we discuss vascular implications of COVID-19 and relate this to circulating biomarker, endothelial, erythrocyte and platelet dysfunction. During the progression of the disease, these markers may either be within healthy levels, upregulated or eventually depleted. Most significant is that patients need to be treated early in the disease progression, when high levels of VWF, P-selectin and fibrinogen are present, with normal or slightly increased levels of D-dimer (however, D-dimer levels will rapidly increase as the disease progresses). Progression to VWF and fibrinogen depletion with high D-dimer levels and even higher P-selectin levels, followed by the cytokine storm, will be indicative of a poor prognosis. We conclude by looking at point-of-care devices and methodologies in COVID-19 management and suggest that a personalized medicine approach should be considered in the treatment of patients.
Collapse
Affiliation(s)
- Corlia Grobler
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch 7602, South Africa; (C.G.); (S.C.M.); (L.M.G.); (J.C.B.)
| | - Siphosethu C. Maphumulo
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch 7602, South Africa; (C.G.); (S.C.M.); (L.M.G.); (J.C.B.)
| | - L. Mireille Grobbelaar
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch 7602, South Africa; (C.G.); (S.C.M.); (L.M.G.); (J.C.B.)
| | - Jhade C. Bredenkamp
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch 7602, South Africa; (C.G.); (S.C.M.); (L.M.G.); (J.C.B.)
| | - Gert J. Laubscher
- Elsie du Toit Street, Stellenbosch MediClinic, Stellenbosch 7600, South Africa; (G.J.L.); (P.J.L.)
| | - Petrus J. Lourens
- Elsie du Toit Street, Stellenbosch MediClinic, Stellenbosch 7600, South Africa; (G.J.L.); (P.J.L.)
| | - Janami Steenkamp
- PathCare Laboratories, PathCare Business Centre, Neels Bothma Street, N1 City, Cape Town 7460, South Africa;
| | - Douglas B. Kell
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch 7602, South Africa; (C.G.); (S.C.M.); (L.M.G.); (J.C.B.)
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Crown St, Liverpool L69 7ZB, UK
- The Novo Nordisk Foundation Centre for Biosustainability, Building 220, Kemitorve Technical University of Denmark, 2800 Kongens Lyngby, Denmark
- Correspondence: (D.B.K.); (E.P.)
| | - Etheresia Pretorius
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch 7602, South Africa; (C.G.); (S.C.M.); (L.M.G.); (J.C.B.)
- Correspondence: (D.B.K.); (E.P.)
| |
Collapse
|
61
|
Bolliger D, Lancé MD, Siegemund M. Point-of-Care Platelet Function Monitoring: Implications for Patients With Platelet Inhibitors in Cardiac Surgery. J Cardiothorac Vasc Anesth 2020; 35:1049-1059. [PMID: 32807601 DOI: 10.1053/j.jvca.2020.07.050] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 07/13/2020] [Accepted: 07/15/2020] [Indexed: 12/13/2022]
Abstract
Although most physicians are comfortable managing the limited anticoagulant effect of aspirin, the recent administration of potent P2Y12 receptor inhibitors in patients undergoing cardiac surgery remains a dilemma. Guidelines recommend discontinuation of potent P2Y12 inhibitors 5- to- 7 days before surgery to reduce the risk of postoperative hemorrhage. Such a strategy might not be feasible before urgent surgery, due to ongoing myocardial ischemia or in patients at high risk for thromboembolic events. Recently, different point-of-care devices to assess functional platelet quality have become available for clinical use. The aim of this narrative review was to evaluate the implications and potential benefits of platelet function monitoring in guiding perioperative management and therapeutic options in patients treated with antiplatelets, including aspirin or P2Y12 receptor inhibitors, undergoing cardiac surgery. No objective superiority of one point-of-care device over another was found in a large meta-analysis. Their accuracy and reliability are generally limited in the perioperative period. In particular, preoperative platelet function testing has been used to assess platelet contribution to bleeding after cardiac surgery. However, predictive values for postoperative hemorrhage and transfusion requirements are low, and there is a significant variability between and within these tests. Further, platelet function monitoring has been used to optimize the preoperative waiting period after cessation of dual antiplatelet therapy before urgent cardiac surgery. Furthermore, studies assessing their value in therapeutic decisions in bleeding patients after cardiac surgery are scarce. A general and liberal use of perioperative platelet function testing is not yet recommended.
Collapse
Affiliation(s)
- Daniel Bolliger
- Department for Anesthesia, Prehospital Emergency Medicine and Pain Therapy, University Hospital Basel, Basel, Switzerland.
| | - Marcus D Lancé
- Department of Anesthesiology, Intensive Care Unit and Perioperative Medicine, Weill-Cornell Medicine-Qatar, Hamad Medical Corporation, Doha, Qatar
| | - Martin Siegemund
- Intensive Care Medicine, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
62
|
Foster H, Wilson C, Philippou H, Foster R. Progress toward a Glycoprotein VI Modulator for the Treatment of Thrombosis. J Med Chem 2020; 63:12213-12242. [PMID: 32463237 DOI: 10.1021/acs.jmedchem.0c00262] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Pathogenic thrombus formation accounts for the etiology of many serious conditions including myocardial infarction, stroke, deep vein thrombosis, and pulmonary embolism. Despite the development of numerous anticoagulants and antiplatelet agents, the mortality rate associated with these diseases remains high. In recent years, however, significant epidemiological evidence and clinical models have emerged to suggest that modulation of the glycoprotein VI (GPVI) platelet receptor could be harnessed as a novel antiplatelet strategy. As such, many peptidic agents have been described in the past decade, while more recent efforts have focused on the development of small molecule modulators. Herein the rationale for targeting GPVI is summarized and the published GPVI modulators are reviewed, with particular focus on small molecules. A qualitative pharmacophore hypothesis for small molecule ligands at GPVI is also presented.
Collapse
Affiliation(s)
- Holly Foster
- School of Chemistry and Leeds Institute of Cardiovascular and Metabolic Medicine (LICAMM), School of Medicine, University of Leeds, Leeds LS2 9JT, U.K
| | - Clare Wilson
- Leeds Institute of Cardiovascular and Metabolic Medicine (LICAMM), School of Medicine, University of Leeds, Leeds LS2 9JT, U.K
| | - Helen Philippou
- Leeds Institute of Cardiovascular and Metabolic Medicine (LICAMM), School of Medicine, University of Leeds, Leeds LS2 9JT, U.K
| | - Richard Foster
- School of Chemistry and Leeds Institute of Cardiovascular and Metabolic Medicine (LICAMM), School of Medicine, University of Leeds, Leeds LS2 9JT, U.K
| |
Collapse
|
63
|
Abstract
Acute coronary syndromes (ACS) are a global cause of mortality and morbidity that affect millions of lives worldwide. Following atherosclerotic plaque rupture, platelet activation and aggregation are the two major elements that initiate thrombus formation inside a coronary artery, which can obstruct blood flow and cause myocardial ischemia; ergo, antiplatelet therapy forms a major part of the treatment strategy for ACS. Patients with ACS routinely receive dual antiplatelet therapy (DAPT), which consists of aspirin and a platelet P2Y12 inhibitor to both treat and prevent atherothrombosis. Use of platelet glycoprotein (GP) IIb/IIIa inhibitors is now limited due to the risk of severe bleeding and thrombocytopenia. Thus, administration of GPIIb/IIIa inhibitors is generally restricted to bail out thrombotic events associated with PCI. Furthermore, current antiplatelet medications mainly rely on thromboxane A2 and P2Y12 inhibition, which have broad-acting effects on platelets and are known to cause bleeding, which especially limits the long-term use of these agents. In addition, not all ACS patients treated with current antiplatelet treatments are protected from recurrence of arterial thrombosis, since many platelet mechanisms and activation pathways remain uninhibited by current antiplatelet therapy. Pharmacological antagonism of novel targets involved in platelet function could shape future antiplatelet therapies that could ultimately lead to more effective or safer therapeutic approaches. In this article, we focus on inhibitors of promising targets that have not yet been introduced into clinical practice, including inhibitors of GPVI, protease-activated receptor (PAR)-4, GPIb, 5-hydroxytryptamine receptor subtype 2A (5-HT2A), protein disulfide isomerase, P-selectin and phosphoinositide 3-kinase β.
Collapse
Affiliation(s)
- Fawaz O Alenazy
- Institute of Cardiovascular Sciences, University of Birmingham , Birmingham, UK
| | - Mark R Thomas
- Institute of Cardiovascular Sciences, University of Birmingham , Birmingham, UK.,UHB and SWBH NHS Trusts , Birmingham, UK
| |
Collapse
|
64
|
Montague SJ, Hicks SM, Lee CSM, Coupland LA, Parish CR, Lee WM, Andrews RK, Gardiner EE. Fibrin exposure triggers αIIbβ3-independent platelet aggregate formation, ADAM10 activity and glycoprotein VI shedding in a charge-dependent manner. J Thromb Haemost 2020; 18:1447-1458. [PMID: 32198957 DOI: 10.1111/jth.14797] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 03/11/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND Collagen and fibrin engagement and activation of glycoprotein (GP) VI induces proteolytic cleavage of the GPVI ectodomain generating shed soluble GPVI (sGPVI). Collagen-mediated GPVI shedding requires intracellular signalling to release the sGPVI, mediated by A Disintegrin And Metalloproteinase 10 (ADAM10); however, the precise mechanism by which fibrin induces GPVI shedding remains elusive. Plasma sGPVI levels are elevated in patients with coagulopathies, sepsis, or inflammation and can predict onset of sepsis and sepsis-related mortality; therefore, it is clinically important to understand the mechanisms of GPVI shedding under conditions of minimal collagen exposure. OBJECTIVES Our aim was to characterize mechanisms by which fibrin-GPVI interactions trigger GPVI shedding. METHODS Platelet aggregometry, sGPVI ELISA, and an ADAM10 fluorescence resonance energy transfer assay were used to measure fibrin-mediated platelet responses. RESULTS Fibrin induced αIIbβ3-independent washed platelet aggregate formation, GPVI shedding, and increased ADAM10 activity, all of which were insensitive to pre-treatment with inhibitors of Src family kinases but were divalent cation- and metalloproteinase-dependent. In contrast, treatment of washed platelets with other GPVI ligands, collagen, and collagen-related peptide caused αIIbβ3-dependent platelet aggregation and GPVI release but did not increase constitutive ADAM10 activity. CONCLUSIONS Fibrin engages GPVI in a manner that differs from other GPVI ligands. Inclusion of polyanionic molecules disrupted fibrin-induced platelet aggregate formation and sGPVI release, suggesting that electrostatic charge may play a role in fibrin/GPVI engagement. It may be feasible to exploit this property and specifically disrupt GPVI/fibrin interactions whilst sparing GPVI/collagen engagement.Fibrin engages GPVI in a manner that differs from other GPVI ligands. Inclusion of polyanionic molecules disrupted fibrin-induced platelet aggregate formation and sGPVI release, suggesting that electrostatic charge may play a role in fibrin/GPVI engagement. It may be feasible to exploit this property and specifically disrupt GPVI/fibrin interactions whilst sparing GPVI/collagen engagement.
Collapse
Affiliation(s)
- Samantha J Montague
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Sarah M Hicks
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Christine S-M Lee
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Lucy A Coupland
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Christopher R Parish
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Woei M Lee
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
- Research School of Electrical, Energy and Materials Engineering, College of Engineering and Computer Science, The Australian National University, Canberra, ACT, Australia
| | - Robert K Andrews
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
- Australian Centre for Blood Diseases, Monash University, Melbourne, VIC, Australia
| | - Elizabeth E Gardiner
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| |
Collapse
|
65
|
Spiryova DV, Vorobev AY, Klimontov VV, Koroleva EA, Moskalensky AE. Optical uncaging of ADP reveals the early calcium dynamics in single, freely moving platelets. BIOMEDICAL OPTICS EXPRESS 2020; 11:3319-3330. [PMID: 32637257 PMCID: PMC7316007 DOI: 10.1364/boe.392745] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 05/11/2020] [Accepted: 05/11/2020] [Indexed: 06/11/2023]
Abstract
Platelet activation is considered to be a cornerstone in pathogenesis of cardiovascular disease. The assessment of platelet activation at the single-cell level is a promising approach for the research of platelet function in physiological and pathological conditions. Previous studies used the immobilization of platelets on the surface, which significantly alters the activation signaling. Here we show that the use of photolabile "caged" analog of ADP allows one to track the very early stage of platelet activation in single, freely moving cells. In this approach, the diffusion step and ADP receptor ligation are separated in time, and a millisecond-timescale optical pulse may trigger the activation. The technique allows us to measure the delay (lag time) between the stimulus and calcium response in platelets. We also propose a simple model function for calcium peaks, which is in good agreement with the measured data. The proposed technique and model function can be used for in-depth studies of platelet physiology.
Collapse
Affiliation(s)
| | - Alexei Yu. Vorobev
- Novosibirsk State University, Novosibirsk, 630090, Russia
- N.N. Vorozhtsov Novosibirsk Institute of Organic Chemistry SB RAS, Novosibirsk, 630090, Russia
| | - Vadim V. Klimontov
- Novosibirsk State University, Novosibirsk, 630090, Russia
- Research Institute of Clinical and Experimental Lymphology – Branch of the Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Novosibirsk, 630117, Russia
| | - Elena A. Koroleva
- Novosibirsk State University, Novosibirsk, 630090, Russia
- Research Institute of Clinical and Experimental Lymphology – Branch of the Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Novosibirsk, 630117, Russia
| | - Alexander E. Moskalensky
- Novosibirsk State University, Novosibirsk, 630090, Russia
- Voevodsky Institute of Chemical Kinetics and Combustion SB RAS, Novosibirsk, 630090, Russia
| |
Collapse
|
66
|
Horev MB, Zabary Y, Zarka R, Sorrentino S, Medalia O, Zaritsky A, Geiger B. Differential dynamics of early stages of platelet adhesion and spreading on collagen IV- and fibrinogen-coated surfaces. F1000Res 2020; 9. [PMID: 32566134 PMCID: PMC7281675 DOI: 10.12688/f1000research.23598.2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/25/2020] [Indexed: 12/29/2022] Open
Abstract
Background: Upon wound formation, platelets adhere to the neighboring extracellular matrix and spread on it, a process which is critical for physiological wound healing. Multiple external factors, such as the molecular composition of the environment and its mechanical properties, play a key role in this process and direct its speed and outcome. Methods: We combined live cell imaging, quantitative interference reflection microscopy and cryo-electron tomography to characterize, at a single platelet level, the differential spatiotemporal dynamics of the adhesion process to fibrinogen- and collagen IV-functionalized surfaces. Results: Initially, platelets sense both substrates by transient rapid extensions of filopodia. On collagen IV, a short-term phase of filopodial extension is followed by lamellipodia-based spreading. This transition is preceded by the extension of a single or couple of microtubules into the platelet's periphery and their apparent insertion into the core of the filopodia. On fibrinogen surfaces, the filopodia-to-lamellipodia transition was partial and microtubule extension was not observed leading to limited spreading, which could be restored by manganese or thrombin. Conclusions: Based on these results, we propose that interaction with collagen IV stimulate platelets to extend microtubules to peripheral filopodia, which in turn, enhances filopodial-to-lamellipodial transition and overall lamellipodia-based spreading. Fibrinogen, on the other hand, fails to induce these early microtubule extensions, leading to full lamellipodia spreading in only a fraction of the seeded platelets. We further suggest that activation of integrin αIIbβ3 is essential for filopodial-to-lamellipodial transition, based on the capacity of integrin activators to enhance lamellipodia spreading on fibrinogen.
Collapse
Affiliation(s)
- Melanie B Horev
- Department of Immunology, Weizmann Institute of Science, Rehovot, Rehovot, 76100, Israel
| | - Yishaia Zabary
- Department of Software and Information Systems Engineering, Ben-Gurion University of the Negev, Beer-Sheva, 84105, Israel
| | - Revital Zarka
- Department of Immunology, Weizmann Institute of Science, Rehovot, Rehovot, 76100, Israel
| | - Simona Sorrentino
- Department of Biochemistry, University of Zurich, Zurich, CH-8057, Switzerland
| | - Ohad Medalia
- Department of Biochemistry, University of Zurich, Zurich, CH-8057, Switzerland
| | - Assaf Zaritsky
- Department of Software and Information Systems Engineering, Ben-Gurion University of the Negev, Beer-Sheva, 84105, Israel
| | - Benjamin Geiger
- Department of Immunology, Weizmann Institute of Science, Rehovot, Rehovot, 76100, Israel
| |
Collapse
|
67
|
Lakshmanan HHS, Melrose AR, Sepp ALI, Mitrugno A, Ngo ATP, Khader A, Thompson R, Sallee D, Pang J, Mangin PH, Jandrot-Perrus M, Aslan JE, McCarty OJT. The basement membrane protein nidogen-1 supports platelet adhesion and activation. Platelets 2020; 32:424-428. [PMID: 32233694 DOI: 10.1080/09537104.2020.1745170] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The core structure of the extracellular basement membrane is made up of self-assembling networks of collagen and laminin which associate with each other through the bridging adapter proteins including the sulfated monomeric glycoprotein nidogen. While collagen and laminin are known to support platelet adhesion and activation via β1 integrins and glycoprotein (GP) VI, respectively, whether nidogen contributes to platelet activation and hemostasis is unknown. In this study, we demonstrate that recombinant human nidogen-1 supports platelet adhesion and stimulates platelet activation in a phospholipase-C γ-2 (PLCγ2), Src and Syk kinase-dependent manner downstream. Platetet adhesion to nidogen-1 was inhibited by blocking the platelet receptors GPVI and β1 integrins. Platelet adhesion to nidogen-1 activated the IκB kinase (IKK) complex, while pharmacological inhibition of IKK blocked platelet spreading on nidogen. Taken together our results suggest that nidogen may play a redundant role in hemostasis by activating platelets downstream of GPVI.
Collapse
Affiliation(s)
| | - Alexander R Melrose
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, USA
| | - Anna-Liisa I Sepp
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA.,Chemical, Biological and Environmental Engineering, Oregon State University, Corvallis, USA
| | - Annachiara Mitrugno
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA
| | - Anh T P Ngo
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA
| | - Ayesha Khader
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA
| | - Rachel Thompson
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA.,Biomedical Engineering Department, University of Connecticut, Storrs, CT, USA
| | - Daniel Sallee
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA
| | - Jiaqing Pang
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA
| | - Pierre H Mangin
- INSERM, EFS Grand-Est, BPPS UMR-S 1255, FMTS, Université De Strasbourg, Strasbourg, France
| | - Martine Jandrot-Perrus
- Laboratory of Vascular Translational Science, Sorbonne Paris Cité, Institut National de la Santé et de la Recherche Médicale (INSERM), Université Paris Diderot, Paris, France
| | - Joseph E Aslan
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA.,Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, USA
| | - Owen J T McCarty
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
68
|
Dunster JL, Unsworth AJ, Bye AP, Haining EJ, Sowa MA, Di Y, Sage T, Pallini C, Pike JA, Hardy AT, Nieswandt B, García Á, Watson SP, Poulter NS, Gibbins JM, Pollitt AY. Interspecies differences in protein expression do not impact the spatiotemporal regulation of glycoprotein VI mediated activation. J Thromb Haemost 2020; 18:485-496. [PMID: 31680418 PMCID: PMC7027541 DOI: 10.1111/jth.14673] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 10/29/2019] [Accepted: 10/30/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Accurate protein quantification is a vital prerequisite for generating meaningful predictions when using systems biology approaches, a method that is increasingly being used to unravel the complexities of subcellular interactions and as part of the drug discovery process. Quantitative proteomics, flow cytometry, and western blotting have been extensively used to define human platelet protein copy numbers, yet for mouse platelets, a model widely used for platelet research, evidence is largely limited to a single proteomic dataset in which the total amount of proteins was generally comparatively higher than those found in human platelets. OBJECTIVES To investigate the functional implications of discrepancies between levels of mouse and human proteins in the glycoprotein VI (GPVI) signalling pathway using a systems pharmacology model of GPVI. METHODS The protein copy number of mouse platelet receptors was determined using flow cytometry. The Virtual Platelet, a mathematical model of GPVI signalling, was used to determine the consequences of protein copy number differences observed between human and mouse platelets. RESULTS AND CONCLUSION Despite the small size of mouse platelets compared to human platelets they possessed a greater density of surface receptors alongside a higher concentration of intracellular signalling proteins. Surprisingly the predicted temporal profile of Syk activity was similar in both species with predictions supported experimentally. Super resolution microscopy demonstrates that the spatial distribution of Syk is similar between species, suggesting that the spatial distribution of receptors and signalling molecules in activated platelets, rather than their copy number, is important for signalling pathway regulation.
Collapse
Affiliation(s)
- Joanne L. Dunster
- Institute for Cardiovascular and Metabolic Research (ICMR)School of Biological SciencesUniversity of ReadingReadingUK
| | - Amanda J. Unsworth
- Institute for Cardiovascular and Metabolic Research (ICMR)School of Biological SciencesUniversity of ReadingReadingUK
- Department of Life SciencesSchool of Science and EngineeringManchester Metropolitan UniversityManchesterUK
| | - Alexander P. Bye
- Institute for Cardiovascular and Metabolic Research (ICMR)School of Biological SciencesUniversity of ReadingReadingUK
| | - Elizabeth J. Haining
- Institute of Cardiovascular Sciences (ICVS)College of Medical and Dental SciencesUniversity of BirminghamBirminghamUK
| | - Marcin A. Sowa
- Institute for Cardiovascular and Metabolic Research (ICMR)School of Biological SciencesUniversity of ReadingReadingUK
- Platelet Proteomics GroupCenter for Research in Molecular Medicine and Chronic Diseases (CIMUS)Universidade de Santiago de CompostelaSantiago de CompostelaSpain
| | - Ying Di
- Institute of Cardiovascular Sciences (ICVS)College of Medical and Dental SciencesUniversity of BirminghamBirminghamUK
| | - Tanya Sage
- Institute for Cardiovascular and Metabolic Research (ICMR)School of Biological SciencesUniversity of ReadingReadingUK
| | - Chiara Pallini
- Institute of Cardiovascular Sciences (ICVS)College of Medical and Dental SciencesUniversity of BirminghamBirminghamUK
| | - Jeremy A. Pike
- Centre of Membrane Proteins and Receptors (COMPARE)Universities of Birmingham and NottinghamMidlandsBirminghamUK
| | - Alexander T. Hardy
- Institute of Cardiovascular Sciences (ICVS)College of Medical and Dental SciencesUniversity of BirminghamBirminghamUK
| | - Bernhard Nieswandt
- Department of Experimental BiomedicineUniversity HospitalUniversity of WürzburgWürzburgGermany
| | - Ángel García
- Platelet Proteomics GroupCenter for Research in Molecular Medicine and Chronic Diseases (CIMUS)Universidade de Santiago de CompostelaSantiago de CompostelaSpain
| | - Steve P. Watson
- Institute of Cardiovascular Sciences (ICVS)College of Medical and Dental SciencesUniversity of BirminghamBirminghamUK
- Centre of Membrane Proteins and Receptors (COMPARE)Universities of Birmingham and NottinghamMidlandsBirminghamUK
| | - Natalie S. Poulter
- Institute of Cardiovascular Sciences (ICVS)College of Medical and Dental SciencesUniversity of BirminghamBirminghamUK
- Centre of Membrane Proteins and Receptors (COMPARE)Universities of Birmingham and NottinghamMidlandsBirminghamUK
| | - Jonathan M. Gibbins
- Institute for Cardiovascular and Metabolic Research (ICMR)School of Biological SciencesUniversity of ReadingReadingUK
| | - Alice Y. Pollitt
- Institute for Cardiovascular and Metabolic Research (ICMR)School of Biological SciencesUniversity of ReadingReadingUK
| |
Collapse
|
69
|
Gaspar RS, da Silva SA, Stapleton J, Fontelles JLDL, Sousa HR, Chagas VT, Alsufyani S, Trostchansky A, Gibbins JM, Paes AMDA. Myricetin, the Main Flavonoid in Syzygium cumini Leaf, Is a Novel Inhibitor of Platelet Thiol Isomerases PDI and ERp5. Front Pharmacol 2020; 10:1678. [PMID: 32116678 PMCID: PMC7011086 DOI: 10.3389/fphar.2019.01678] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 12/23/2019] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Flavonoids have been characterized as a prominent class of compounds to treat thrombotic diseases through the inhibition of thiol isomerases. Syzygium cumini is a flavonoid-rich medicinal plant that contains myricetin and gallic acid. Little is known about the potential antiplatelet properties of S. cumini and its constituent flavonoids. OBJECTIVE To evaluate the antiplatelet effects and mechanism of action of a polyphenol-rich extract (PESc) from S. cumini leaf and its most prevalent polyphenols, myricetin and gallic acid. METHODS PESc, myricetin, and gallic acid were incubated with platelet-rich plasma and washed platelets to assess platelet aggregation and activation. In vitro platelet adhesion and thrombus formation as well as in vivo bleeding time were performed. Finally, myricetin was incubated with recombinant thiol isomerases to assess its potential to bind and inhibit these, while molecular docking studies predicted possible binding sites. RESULTS PESc decreased platelet activation and aggregation induced by different agonists. Myricetin exerted potent antiplatelet effects, whereas gallic acid did not. Myricetin reduced the ability of platelets to spread on collagen, form thrombi in vitro without affecting hemostasis in vivo. Fluorescence quenching studies suggested myricetin binds to different thiol isomerases with similar affinity, despite inhibiting only protein disulfide isomerase (PDI) and ERp5 reductase activities. Finally, molecular docking studies suggested myricetin formed non-covalent bonds with PDI and ERp5. CONCLUSIONS PESc and its most abundant flavonoid myricetin strongly inhibit platelet function. Additionally, myricetin is a novel inhibitor of ERp5 and PDI, unveiling a new therapeutic perspective for the treatment of thrombotic disorders.
Collapse
Affiliation(s)
- Renato Simões Gaspar
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
- Laboratory of Experimental Physiology, Department of Physiological Sciences, Federal University of Maranhão, São Luís, Brazil
| | - Samira Abdalla da Silva
- Laboratory of Experimental Physiology, Department of Physiological Sciences, Federal University of Maranhão, São Luís, Brazil
| | - Jennifer Stapleton
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
| | - João Lucas de Lima Fontelles
- Laboratory of Experimental Physiology, Department of Physiological Sciences, Federal University of Maranhão, São Luís, Brazil
| | - Hiran Reis Sousa
- Laboratory of Experimental Physiology, Department of Physiological Sciences, Federal University of Maranhão, São Luís, Brazil
| | - Vinicyus Teles Chagas
- Laboratory of Experimental Physiology, Department of Physiological Sciences, Federal University of Maranhão, São Luís, Brazil
| | - Shuruq Alsufyani
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
| | - Andrés Trostchansky
- Departamento de Bioquímica and Centro de Investigaciones Biomédicas, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Jonathan M. Gibbins
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
| | - Antonio Marcus de Andrade Paes
- Laboratory of Experimental Physiology, Department of Physiological Sciences, Federal University of Maranhão, São Luís, Brazil
| |
Collapse
|
70
|
Antiplatelet therapy abrogates platelet-assisted Staphylococcus aureus infectivity of biological heart valve conduits. J Thorac Cardiovasc Surg 2020; 161:e457-e472. [PMID: 31926702 DOI: 10.1016/j.jtcvs.2019.10.188] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 10/25/2019] [Accepted: 10/25/2019] [Indexed: 12/23/2022]
Abstract
OBJECTIVE Although recent advances in pulmonary valve replacement have enabled excellent hemodynamics, infective endocarditis remains a serious complication, particularly for implanted bovine jugular vein (BJV) conduits. METHODS We investigated contributions by platelets and plasma fibrinogen to endocarditis initiation on various grafts used for valve replacement. Thus, adherence of Staphylococcus aureus and platelets to 5 graft tissues was studied quantitatively in perfusion chambers, assisted by microscopic analysis. We also evaluated standard antiplatelet therapy to prevent onset of S aureus endocarditis. RESULTS Of all tissues, bovine pericardium (BP) showed the greatest fibrinogen binding. Perfusion of all plasma-precoated tissues identified BP and BJVwall with the greatest affinity for S aureus. Perfusions of anticoagulated human blood over all tissues also triggered more platelet adhesion to BP and BJVwall as single platelets. Several controls confirmed that both S aureus and platelets were recruited on immobilized fibrinogen. In addition, perfusions (and controls) over plasma-coated tissues with whole blood, spiked with S aureus, revealed that bacteria exclusively bound to adhered platelets. Both the platelet adhesion and platelet-mediated S aureus recruitment required platelet αIIbβ3 and coated or soluble fibrinogen, respectively, interactions abrogated by the αIIbβ3-antagonist eptifibatide. Also, standard antiplatelet therapy (aspirin/ticagrelor) reduced the adherence of S aureus in blood to BJV 3-fold. CONCLUSIONS Binding of plasma fibrinogen to especially BJV grafts enables adhesion of single platelets via αIIbβ3. S aureus then attaches from blood to (activated) bound platelet αIIbβ3 via plasma fibrinogen. Dual antiplatelet therapy appears a realistic approach to prevent endocarditis and its associated mortality.
Collapse
|
71
|
Stegner D, Klaus V, Nieswandt B. Platelets as Modulators of Cerebral Ischemia/Reperfusion Injury. Front Immunol 2019; 10:2505. [PMID: 31736950 PMCID: PMC6838001 DOI: 10.3389/fimmu.2019.02505] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 10/07/2019] [Indexed: 12/29/2022] Open
Abstract
Ischemic stroke is among the leading causes of disability and death worldwide. In acute ischemic stroke, the rapid recanalization of occluded cranial vessels is the primary therapeutic aim. However, experimental data (obtained using mostly the transient middle cerebral artery occlusion model) indicates that progressive stroke can still develop despite successful recanalization, a process termed "reperfusion injury." Mounting experimental evidence suggests that platelets and T cells contribute to cerebral ischemia/reperfusion injury, and ischemic stroke is increasingly considered a thrombo-inflammatory disease. The interaction of von Willebrand factor and its receptor on the platelet surface, glycoprotein Ib, as well as many activatory platelet receptors and platelet degranulation contribute to secondary infarct growth in this setting. In contrast, interference with GPIIb/IIIa-dependent platelet aggregation and thrombus formation does not improve the outcome of acute brain ischemia but dramatically increases the susceptibility to intracranial hemorrhage. Here, we summarize the current understanding of the mechanisms and the potential translational impact of platelet contributions to cerebral ischemia/reperfusion injury.
Collapse
Affiliation(s)
- David Stegner
- Institute of Experimental Biomedicine–Department I, University Hospital Würzburg, Würzburg, Germany
- Rudolf Virchow Center for Experimental Biomedicine, University of Würzburg, Würzburg, Germany
| | - Vanessa Klaus
- Institute of Experimental Biomedicine–Department I, University Hospital Würzburg, Würzburg, Germany
| | - Bernhard Nieswandt
- Institute of Experimental Biomedicine–Department I, University Hospital Würzburg, Würzburg, Germany
- Rudolf Virchow Center for Experimental Biomedicine, University of Würzburg, Würzburg, Germany
| |
Collapse
|
72
|
Hechler B, Dupuis A, Mangin PH, Gachet C. Platelet preparation for function testing in the laboratory and clinic: Historical and practical aspects. Res Pract Thromb Haemost 2019; 3:615-625. [PMID: 31624781 PMCID: PMC6781931 DOI: 10.1002/rth2.12240] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 06/05/2019] [Indexed: 01/04/2023] Open
Abstract
Laboratory tests of platelet function are instrumental in studying platelet physiology and inherited or acquired platelet abnormalities. Light transmission aggregometry, developed in the early 1960s, is still considered the gold standard for the identification and diagnosis of platelet function disorders. Since then, novel techniques have been developed, including flow-based assays and flow cytometry. In this tutorial, we describe the basic methodologies for the preparation of citrated platelet-rich plasma and washed platelet suspensions and discuss their respective advantages and limitations as well as important factors to consider to perform high-quality tests of platelet function. In addition, the methodologies of the main platelet function tests (light transmission aggregation, flow-based assays, and flow cytometric assays) are described, and their respective strengths and limitations are discussed to assess various aspects of platelet biology.
Collapse
Affiliation(s)
- Béatrice Hechler
- Université de StrasbourgINSERMEtablissement Français du Sang (EFS) Grand EstBPPS UMR_S 1255Fédération de Médecine Translationnelle de Strasbourg (FMTS)StrasbourgFrance
| | - Arnaud Dupuis
- Université de StrasbourgINSERMEtablissement Français du Sang (EFS) Grand EstBPPS UMR_S 1255Fédération de Médecine Translationnelle de Strasbourg (FMTS)StrasbourgFrance
| | - Pierre H. Mangin
- Université de StrasbourgINSERMEtablissement Français du Sang (EFS) Grand EstBPPS UMR_S 1255Fédération de Médecine Translationnelle de Strasbourg (FMTS)StrasbourgFrance
| | - Christian Gachet
- Université de StrasbourgINSERMEtablissement Français du Sang (EFS) Grand EstBPPS UMR_S 1255Fédération de Médecine Translationnelle de Strasbourg (FMTS)StrasbourgFrance
| |
Collapse
|
73
|
Abstract
Acute kidney injury (AKI), a major public health problem associated with high mortality and increased risk of progression towards end-stage renal disease, is characterized by the activation of intra-renal haemostatic and inflammatory processes. Platelets, which are present in high numbers in the circulation and can rapidly release a broad spectrum of bioactive mediators, are important acute modulators of inflammation and haemostasis, as they are the first cells to arrive at sites of acute injury, where they interact with endothelial cells and leukocytes. Diminished control of platelet reactivity by endothelial cells and/or an increased release of platelet-activating mediators can lead to uncontrolled platelet activation in AKI. As increased platelet sequestration and increased expression levels of the markers P-selectin, thromboxane A2, CC-chemokine ligand 5 and platelet factor 4 on platelets have been reported in kidneys following AKI, platelet activation likely plays a part in AKI pathology. Results from animal models and some clinical studies highlight the potential of antiplatelet therapies in the preservation of renal function in the context of AKI, but as current strategies also affect other cell types and non-platelet-derived mediators, additional studies are required to further elucidate the extent of platelet contribution to the pathology of AKI and to determine the best therapeutic approach by which to specifically target related pathogenic pathways.
Collapse
Affiliation(s)
- Marcel P B Jansen
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Sandrine Florquin
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Joris J T H Roelofs
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands.
| |
Collapse
|
74
|
Nurden AT. Clinical significance of altered collagen-receptor functioning in platelets with emphasis on glycoprotein VI. Blood Rev 2019; 38:100592. [PMID: 31351674 DOI: 10.1016/j.blre.2019.100592] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 07/05/2019] [Accepted: 07/19/2019] [Indexed: 01/01/2023]
Abstract
Much interest surrounds the receptors α2β1 and glycoprotein VI (GPVI) whose synchronized action mediates the attachment and activation of platelets on collagen, essential for preventing blood loss but also the most thrombogenic component of the vessel wall. Subject to density variations on platelets through natural polymorphisms, the absence of α2β1 or GPVI uniquely leads to a substantial block of hemostasis without causing major bleeding. Specific to the megakaryocyte lineage, GPVI and its signaling pathways are most promising targets for anti-thrombotic therapy. This review looks at the clinical consequences of the loss of collagen receptor function with emphasis on both the inherited and acquired loss of GPVI with brief mention of mouse models when necessary. A detailed survey of rare case reports of patients with inherited disease-causing variants of the GP6 gene is followed by an assessment of the causes and clinical consequences of acquired GPVI deficiency, a more frequent finding most often due to antibody-induced platelet GPVI shedding. Release of soluble GPVI is brought about by platelet metalloproteinases; a process induced by ligand or antibody binding to GPVI or even high shear forces. Also included is an assessment of the clinical importance of GPVI-mediated platelet interactions with fibrin and of the promise shown by the pharmacological inhibition of GPVI in a cardiovascular context. The role for GPVI in platelet function in inflammation and in the evolution and treatment of major illnesses such as rheumatoid arthritis, cancer and sepsis is also discussed.
Collapse
Affiliation(s)
- Alan T Nurden
- Institut de Rhythmologie et de Modélisation Cardiaque, PTIB, Hôpital Xavier Arnozan, 33600 Pessac, France.
| |
Collapse
|
75
|
Obydennyi SI, Artemenko EO, Sveshnikova AN, Ignatova AA, Varlamova TV, Gambaryan S, Lomakina GY, Ugarova NN, Kireev II, Ataullakhanov FI, Novichkova GA, Maschan AA, Shcherbina A, Panteleev M. Mechanisms of increased mitochondria-dependent necrosis in Wiskott-Aldrich syndrome platelets. Haematologica 2019; 105:1095-1106. [PMID: 31278208 PMCID: PMC7109739 DOI: 10.3324/haematol.2018.214460] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 07/04/2019] [Indexed: 11/23/2022] Open
Abstract
Wiskott-Aldrich syndrome (WAS) is associated with thrombocytopenia of unclear origin. We investigated real-time cytosolic calcium dynamics, mitochondrial membrane potential and phoszphatidylserine (PS) exposure in single fibrinogen-bound platelets using confocal microscopy. The WAS platelets had higher resting calcium levels, more frequent spikes, and their mitochondria more frequently lost membrane potential followed by PS exposure (in 22.9% of platelets vs. 3.9% in controls; P<0.001) after the collapse of the last mitochondria. This phenomenon was inhibited by the mitochondrial permeability transition pore inhibitor cyclosporine A, as well by xestospongin C and lack of extracellular calcium. Thapsigargin by itself caused accelerated cell death in the WAS platelets. The number of mitochondria was predictive of PS exposure: 33% of platelets from WAS patients with fewer than five mitochondria exposed PS, while only 12% did among those that had five or more mitochondria. Interestingly, healthy donor platelets with fewer mitochondria also more readily became procoagulant upon PAR1/PAR4 stimulation. Collapse of single mitochondria led to greater cytosolic calcium increase in WAS platelets if they had one to three mitochondria compared with platelets containing higher numbers. A computer systems biology model of platelet calcium homeostasis showed that smaller platelets with fewer mitochondria could have impaired calcium homeostasis because of higher surface-to-volume ratio and greater metabolic load, respectively. There was a correlation (C=0.81, P<0.02) between the mean platelet size and platelet count in the WAS patients. We conclude that WAS platelets readily expose PS via a mitochondria-dependent necrotic mechanism caused by their smaller size, which could contribute to the development of thrombocytopenia.
Collapse
Affiliation(s)
- Sergey I Obydennyi
- National Scientific and Practical Center of Pediatric Hematology, Oncology and Immunology named after Dmitry Rogachev, Moscow.,Center for Theoretical Problems of Physicochemical Pharmacology, Moscow
| | - Elena O Artemenko
- National Scientific and Practical Center of Pediatric Hematology, Oncology and Immunology named after Dmitry Rogachev, Moscow.,Center for Theoretical Problems of Physicochemical Pharmacology, Moscow
| | - Anastasia N Sveshnikova
- National Scientific and Practical Center of Pediatric Hematology, Oncology and Immunology named after Dmitry Rogachev, Moscow.,Center for Theoretical Problems of Physicochemical Pharmacology, Moscow.,Faculty of Physics, Lomonosov Moscow State University, Moscow.,I.M. Sechenov First Moscow State Medical University, Moscow
| | - Anastasia A Ignatova
- National Scientific and Practical Center of Pediatric Hematology, Oncology and Immunology named after Dmitry Rogachev, Moscow.,Center for Theoretical Problems of Physicochemical Pharmacology, Moscow
| | - Tatiana V Varlamova
- National Scientific and Practical Center of Pediatric Hematology, Oncology and Immunology named after Dmitry Rogachev, Moscow
| | - Stepan Gambaryan
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St Petersburg
| | - Galina Y Lomakina
- Department of Chemistry, Lomonosov Moscow State University, Moscow.,Bauman Moscow State Technical University, Moscow
| | | | - Igor I Kireev
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow
| | - Fazoil I Ataullakhanov
- National Scientific and Practical Center of Pediatric Hematology, Oncology and Immunology named after Dmitry Rogachev, Moscow.,Center for Theoretical Problems of Physicochemical Pharmacology, Moscow.,Faculty of Physics, Lomonosov Moscow State University, Moscow.,Faculty of Biological and Medical Physics, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Galina A Novichkova
- National Scientific and Practical Center of Pediatric Hematology, Oncology and Immunology named after Dmitry Rogachev, Moscow
| | - Aleksey A Maschan
- National Scientific and Practical Center of Pediatric Hematology, Oncology and Immunology named after Dmitry Rogachev, Moscow
| | - Anna Shcherbina
- National Scientific and Practical Center of Pediatric Hematology, Oncology and Immunology named after Dmitry Rogachev, Moscow
| | - Mikhail Panteleev
- National Scientific and Practical Center of Pediatric Hematology, Oncology and Immunology named after Dmitry Rogachev, Moscow .,Center for Theoretical Problems of Physicochemical Pharmacology, Moscow.,Faculty of Physics, Lomonosov Moscow State University, Moscow.,Faculty of Biological and Medical Physics, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| |
Collapse
|
76
|
Role of Platelet Glycoprotein VI and Tyrosine Kinase Syk in Thrombus Formation on Collagen-Like Surfaces. Int J Mol Sci 2019; 20:ijms20112788. [PMID: 31181592 PMCID: PMC6600290 DOI: 10.3390/ijms20112788] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 05/29/2019] [Accepted: 06/04/2019] [Indexed: 01/20/2023] Open
Abstract
Platelet interaction with collagens, via von Willebrand factor, is a potent trigger of shear-dependent thrombus formation mediated by subsequent engagement of the signaling collagen receptor glycoprotein (GP)VI, enforced by integrin α2β1. Protein tyrosine kinase Syk is central in the GPVI-induced signaling pathway, leading to elevated cytosolic Ca2+. We aimed to determine the Syk-mediated thrombogenic activity of several collagen peptides and (fibrillar) type I and III collagens. High-shear perfusion of blood over microspots of these substances resulted in thrombus formation, which was assessed by eight parameters and was indicative of platelet adhesion, activation, aggregation, and contraction, which were affected by the Syk inhibitor PRT-060318. In platelet suspensions, only collagen peptides containing the consensus GPVI-activating sequence (GPO)n and Horm-type collagen evoked Syk-dependent Ca2+ rises. In whole blood under flow, Syk inhibition suppressed platelet activation and aggregation parameters for the collagen peptides with or without a (GPO)n sequence and for all of the collagens. Prediction models based on a regression analysis indicated a mixed role of GPVI in thrombus formation on fibrillar collagens, which was abolished by Syk inhibition. Together, these findings indicate that GPVI-dependent signaling through Syk supports platelet activation in thrombus formation on collagen-like structures regardless of the presence of a (GPO)n sequence.
Collapse
|
77
|
Jandrot-Perrus M, Hermans C, Mezzano D. Platelet glycoprotein VI genetic quantitative and qualitative defects. Platelets 2019; 30:708-713. [PMID: 31068042 DOI: 10.1080/09537104.2019.1610166] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Platelet membrane glycoprotein VI (GPVI) is increasingly recognized as an important receptor for thrombus formation and growth. Numerous arguments have been published indicating that GPVI plays a major role in thrombosis without being essential for physiological hemostasis. In humans, GPVI deficiencies are rarely reported. These are most often deficiencies occurring in the context of autoimmunity and, more rarely, genetic deficits. The purpose of this review is to compile data on the quantitative and qualitative genetic abnormalities of GPVI.
Collapse
Affiliation(s)
- Martine Jandrot-Perrus
- a UMR_S1148, Laboratory for Vascular Translational Science , INSERM, University Paris Diderot , Paris , France
| | - Cedric Hermans
- b Haemostasis and Thrombosis Unit, Haemophilia Clinic, Division of Adult Haematology , St-Luc University Hospital , Brussels , Belgium
| | - Diego Mezzano
- c Department of Hematology-Oncology , School of Medicine, Pontificia Universidad Católica de Chile , Santiago , Chile
| |
Collapse
|
78
|
Voors-Pette C, Lebozec K, Dogterom P, Jullien L, Billiald P, Ferlan P, Renaud L, Favre-Bulle O, Avenard G, Machacek M, Plétan Y, Jandrot-Perrus M. Safety and Tolerability, Pharmacokinetics, and Pharmacodynamics of ACT017, an Antiplatelet GPVI (Glycoprotein VI) Fab. Arterioscler Thromb Vasc Biol 2019; 39:956-964. [DOI: 10.1161/atvbaha.118.312314] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Objective—
ACT017 is a novel, first in class, therapeutic antibody to platelet GPVI (glycoprotein VI) with potent and selective antiplatelet effects. This first-in-human, randomized, placebo-controlled phase 1 study was conducted to evaluate the safety, tolerability, pharmacokinetics, and pharmacodynamics of ACT017 in healthy subjects.
Approach and Results—
Six cohorts of 8 healthy male and female subjects each received ascending single doses of ACT017 (n=6) or placebo (n=2) as a 6-hour intravenous infusion, with ¼ of the total dose administered within 15 minutes and the rest of the dose (¾ of the total dose) administered within 5 hours and 45 minutes. The 6 investigated doses ranged from 62.5 to 2000 mg. All doses of ACT017 were well tolerated, and no serious adverse events occurred during the study. None of the subjects reported an infusion site reaction. Template bleeding time was not affected in a clinically significant manner by any of the ACT017 doses. Plasma concentrations, determined by liquid chromatography-tandem mass spectrometry, increased linearly with the dose received as were the established pharmacokinetics values. There was no change in the platelet count, platelet GPVI expression assessed by flow cytometry, or plasma levels of soluble GPVI assessed by ELISA. In contrast, administration of ACT017 inhibited collagen-induced platelet aggregation measured by light transmission aggregometry on platelet-rich plasma, and the extent and duration of the effect were dose-dependent.
Conclusions—
The novel antiplatelet agent ACT017 has consistent pharmacokinetic/pharmacodynamic properties and favorable safety and tolerability profiles warranting further clinical development.
Collapse
Affiliation(s)
| | - Kristell Lebozec
- Acticor-Biotech, Paris, France (K.L., L.J., P.B., P.F., O.F-B., G.A., Y.P., M.J-P.)
- University Paris Sud, School of Pharmacy, Inserm-S 1193, Châtenay Malabry, France (K.L., P.B.)
| | - Peter Dogterom
- From the QPS Holdings LLC, Groningen, the Netherlands (C.V.-P., P.D.)
| | - Laurie Jullien
- Acticor-Biotech, Paris, France (K.L., L.J., P.B., P.F., O.F-B., G.A., Y.P., M.J-P.)
| | - Philippe Billiald
- Acticor-Biotech, Paris, France (K.L., L.J., P.B., P.F., O.F-B., G.A., Y.P., M.J-P.)
- University Paris Sud, School of Pharmacy, Inserm-S 1193, Châtenay Malabry, France (K.L., P.B.)
| | - Pauline Ferlan
- Acticor-Biotech, Paris, France (K.L., L.J., P.B., P.F., O.F-B., G.A., Y.P., M.J-P.)
| | | | - Olivier Favre-Bulle
- Acticor-Biotech, Paris, France (K.L., L.J., P.B., P.F., O.F-B., G.A., Y.P., M.J-P.)
- 3Biotech, Paris, France (O.F-B.)
| | - Gilles Avenard
- Acticor-Biotech, Paris, France (K.L., L.J., P.B., P.F., O.F-B., G.A., Y.P., M.J-P.)
| | | | - Yannick Plétan
- Acticor-Biotech, Paris, France (K.L., L.J., P.B., P.F., O.F-B., G.A., Y.P., M.J-P.)
- ULTRACE, Orsay, France (Y.P.)
| | - Martine Jandrot-Perrus
- Acticor-Biotech, Paris, France (K.L., L.J., P.B., P.F., O.F-B., G.A., Y.P., M.J-P.)
- Inserm, Université Paris Sorbonne Cité, UMR_S1148, Paris, France (M.J-P.)
| |
Collapse
|
79
|
Kaneva VN, Martyanov AA, Morozova DS, Panteleev MA, Sveshnikova AN. Platelet Integrin αIIbβ3: Mechanisms of Activation and Clustering; Involvement into the Formation of the Thrombus Heterogeneous Structure. BIOCHEMISTRY (MOSCOW), SUPPLEMENT SERIES A: MEMBRANE AND CELL BIOLOGY 2019. [DOI: 10.1134/s1990747819010033] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
80
|
Rayes J, Watson SP, Nieswandt B. Functional significance of the platelet immune receptors GPVI and CLEC-2. J Clin Invest 2019; 129:12-23. [PMID: 30601137 DOI: 10.1172/jci122955] [Citation(s) in RCA: 217] [Impact Index Per Article: 36.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Although platelets are best known for their role in hemostasis, they are also crucial in development, host defense, inflammation, and tissue repair. Many of these roles are regulated by the immune-like receptors glycoprotein VI (GPVI) and C-type lectin receptor 2 (CLEC-2), which signal through an immunoreceptor tyrosine-based activation motif (ITAM). GPVI is activated by collagen in the subendothelial matrix, by fibrin and fibrinogen in the thrombus, and by a remarkable number of other ligands. CLEC-2 is activated by the transmembrane protein podoplanin, which is found outside of the vasculature and is upregulated in development, inflammation, and cancer, but there is also evidence for additional ligands. In this Review, we discuss the physiological and pathological roles of CLEC-2 and GPVI and their potential as targets in thrombosis and thrombo-inflammatory disorders (i.e., disorders in which inflammation plays a critical role in the ensuing thrombosis) relative to current antiplatelet drugs.
Collapse
Affiliation(s)
- Julie Rayes
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Steve P Watson
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom.,Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, United Kingdom
| | - Bernhard Nieswandt
- Institute of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany
| |
Collapse
|
81
|
Mutch NJ. Regulation of Fibrinolysis by Platelets. Platelets 2019. [DOI: 10.1016/b978-0-12-813456-6.00023-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
82
|
Mechanisms of receptor shedding in platelets. Blood 2018; 132:2535-2545. [DOI: 10.1182/blood-2018-03-742668] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 10/12/2018] [Indexed: 02/07/2023] Open
Abstract
Abstract
The ability to upregulate and downregulate surface-exposed proteins and receptors is a powerful process that allows a cell to instantly respond to its microenvironment. In particular, mobile cells in the bloodstream must rapidly react to conditions where infection or inflammation are detected, and become proadhesive, phagocytic, and/or procoagulant. Platelets are one such blood cell that must rapidly acquire and manage proadhesive and procoagulant properties in order to execute their primary function in hemostasis. The regulation of platelet membrane properties is achieved via several mechanisms, one of which involves the controlled metalloproteolytic release of adhesion receptors and other proteins from the platelet surface. Proteolysis effectively lowers receptor density and reduces the reactivity of platelets, and is a mechanism to control robust platelet activation. Recent research has also established clear links between levels of platelet receptors and platelet lifespan. In this review, we will discuss the current knowledge of metalloproteolytic receptor regulation in the vasculature with emphasis on the platelet receptor system to highlight how receptor density can influence both platelet function and platelet survival.
Collapse
|
83
|
A mutation of the human EPHB2 gene leads to a major platelet functional defect. Blood 2018; 132:2067-2077. [PMID: 30213874 DOI: 10.1182/blood-2018-04-845644] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 09/08/2018] [Indexed: 12/14/2022] Open
Abstract
The ephrin transmembrane receptor family of tyrosine kinases is involved in platelet function. We report the first EPHB2 variant affecting platelets in 2 siblings (P1 and P2) from a consanguineous family with recurrent bleeding and normal platelet counts. Whole-exome sequencing identified a c.2233C>T variant (missense p.R745C) of the EPHB2 gene. P1 and P2 were homozygous for this variant, while their asymptomatic parents were heterozygous. The p.R745C variant within the tyrosine kinase domain was associated with defects in platelet aggregation, αIIbβ3 activation, and granule secretion induced by G-protein-coupled receptor (GPCR) agonists and convulxin, as well as in thrombus formation on collagen under flow. In contrast, clot retraction, flow-dependent platelet adhesion, and spreading on fibrinogen were only mildly affected, indicating limited effects on αIIbβ3 outside-in signaling. Most importantly, Lyn, Syk, and FcRγ phosphorylation, the initial steps in glycoprotein VI (GPVI) platelet signaling were drastically impaired in the absence of platelet-platelet contact, indicating a positive role for EPHB2 in GPVI activation. Likewise platelet activation by PAR4-AP showed defective Src activation, as opposed to normal protein kinase C activity and Ca2+ mobilization. Overexpression of wild-type and R745C EPHB2 variant in RBL-2H3 (rat basophilic leukemia) cells stably expressing human GPVI confirmed that EPHB2 R745C mutation impaired EPHB2 autophosphorylation but had no effect on ephrin ligand-induced EPHB2 clustering, suggesting it did not interfere with EPHB2-ephrin-mediated cell-to-cell contact. In conclusion, this novel inherited platelet disorder affecting EPHB2 demonstrates this tyrosine kinase receptor plays an important role in platelet function through crosstalk with GPVI and GPCR signaling.
Collapse
|
84
|
Slater A, Perrella G, Onselaer MB, Martin EM, Gauer JS, Xu RG, Heemskerk JWM, Ariëns RAS, Watson SP. Does fibrin(ogen) bind to monomeric or dimeric GPVI, or not at all? Platelets 2018; 30:281-289. [DOI: 10.1080/09537104.2018.1508649] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
- Alexandre Slater
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Gina Perrella
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, The Netherlands
| | - Marie-Blanche Onselaer
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Eleyna M Martin
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Julia S Gauer
- Thrombosis and Tissue Repair Group, Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Rui-Gang Xu
- Thrombosis and Tissue Repair Group, Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Johan WM Heemskerk
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, The Netherlands
| | - Robert A S Ariëns
- Thrombosis and Tissue Repair Group, Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Steve P Watson
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| |
Collapse
|
85
|
Gardiner EE. Proteolytic processing of platelet receptors. Res Pract Thromb Haemost 2018; 2:240-250. [PMID: 30046726 PMCID: PMC6055504 DOI: 10.1002/rth2.12096] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Accepted: 03/01/2018] [Indexed: 12/17/2022] Open
Abstract
Platelets have a major role in hemostasis and an emerging role in biological processes including inflammation and immunity. Many of these processes require platelet adhesion and localization at sites of tissue damage or infection and regulated platelet activation, mediated by platelet adheso-signalling receptors, glycoprotein (GP) Ib-IX-V and GPVI. Work from a number of laboratories has demonstrated that levels of these receptors are closely regulated by metalloproteinases of the A Disintegrin And Metalloproteinase (ADAM) family, primarily ADAM17 and ADAM10. It is becoming increasingly evident that platelets have important roles in innate immunity, inflammation, and in combating infection that extends beyond processes of hemostasis. This overview will examine the molecular events that regulate levels of platelet receptors and then assess ramifications for these events in settings where hemostasis, inflammation, and infection processes are triggered.
Collapse
Affiliation(s)
- Elizabeth E. Gardiner
- ACRF Department of Cancer Biology and TherapeuticsJohn Curtin School of Medical ResearchThe Australian National UniversityCanberraACTAustralia
| |
Collapse
|