1
|
Debaene C, Feys HB, Six KR. Shedding light on GPIbα shedding. Curr Opin Hematol 2024; 31:224-229. [PMID: 38728102 DOI: 10.1097/moh.0000000000000826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2024]
Abstract
PURPOSE OF REVIEW Ectodomain shedding has been investigated since the late 1980s. The abundant and platelet specific GPIbα receptor is cleaved by ADAM17 resulting in the release of its ectodomain called glycocalicin. This review will address the role of glycocalicin as an end-stage marker of platelet turnover and storage lesion and will consider a potential function as effector in processes beyond hemostasis. RECENT FINDINGS Glycocalicin has been described as a marker for platelet senescence, turnover and storage lesion but is not routinely used in a clinical setting because its diagnostic value is nondiscriminatory. Inhibition of glycocalicin shedding improves posttransfusion recovery but little is known (yet) about potential hemostatic improvements. In physiological settings, GPIbα shedding is restricted to the intracellular GPIbα receptor subpopulation suggesting a role for shedding or glycocalicin beyond hemostasis. SUMMARY So far, all evidence represents glycocalicin as an end-stage biomarker of platelet senescence and a potential trigger for platelet clearance. The extensive list of interaction partners of GPIbα in fields beyond hemostasis opens new possibilities to investigate specific effector functions of glycocalicin.
Collapse
Affiliation(s)
- Caitlin Debaene
- Transfusion Research Center, Belgian Red Cross Flanders
- Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Hendrik B Feys
- Transfusion Research Center, Belgian Red Cross Flanders
- Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Katrijn R Six
- Transfusion Research Center, Belgian Red Cross Flanders
| |
Collapse
|
2
|
Wu S, Meena D, Yarmolinsky J, Gill D, Smith A, Dib M, Chauhan G, Rohatgi A, Dehghan A, Tzoulaki I. Mendelian Randomization and Bayesian Colocalization Analysis Implicate Glycoprotein VI as a Potential Drug Target for Cardioembolic Stroke in South Asian Populations. J Am Heart Assoc 2024; 13:e035008. [PMID: 39119976 PMCID: PMC11963915 DOI: 10.1161/jaha.124.035008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 06/20/2024] [Indexed: 08/10/2024]
Abstract
BACKGROUND Circulating plasma proteins are clinically useful biomarkers for stroke risk. We examined the causal links between plasma proteins and stroke risk in individuals of South Asian ancestry. METHODS AND RESULTS We applied proteome-wide Mendelian randomization and colocalization approaches to understand causality of 2922 plasma proteins on stroke risk in individuals of South Asian ancestry. We obtained genetic instruments (proxies) for plasma proteins from the UK Biobank (N=920). Genome-wide association studies summary data for strokes (N≤11 312) were sourced from GIGASTROKE consortium. Our primary approach involved the Wald ratio or inverse-variance-weighted methods, with statistical significance set at false discovery rate <0.1. Additionally, a Bayesian colocalization approach assessed shared causal variants among proteome, transcriptome, and stroke phenotypes to minimize bias from linkage disequilibrium. We found evidence of a potential causal effect of plasma GP6 (glycoprotein VI) levels on cardioembolic stroke (odds ratio [OR]Wald ratio=2.53 [95% CI, 1.59-4.03]; P=9.2×10-5, false discovery rate=0.059). Generalized Mendelian randomization accounting for correlated single nucleotide polymorphisms (SNPs), with the P value threshold at P<5×10-8 and clumped at r2=0.3, showed consistent direction of effect of GP6 on cardioembolic stroke (ORgeneralized inverse-variance-weighted=2.21 [95% CI, 1.46-3.33]; P=1.6×10-4). Colocalization analysis indicated that plasma GP6 levels colocalize with cardioembolic stroke (posterior probability=91.4%). Multitrait colocalization combining transcriptome, proteome, and cardioembolic stroke showed moderate to strong evidence that these 2 traits colocalize with GP6 expression in the coronary artery and brain tissues (multitrait posterior probability>50%). The potential causal effect of GP6 on cardioembolic stroke was not significant in European populations (ORinverse-variance-weighted=1.08 [95% CI, 0.93-1.26]; P=0.29). CONCLUSIONS Our joint Mendelian randomization and colocalization analyses suggest that genetically predicted GP6 is potentially causally associated with cardioembolic stroke risk in individuals of South Asian ancestry. As genetic data on individuals of South Asian ancestry increase, future Mendelian randomization studies with larger sample size for plasma GP6 levels should be implemented to further validate our findings. Additionally, clinical studies will be necessary to verify GP6 as a therapeutic target for cardioembolic stroke in South Asians.
Collapse
Affiliation(s)
- Siwei Wu
- Department of Epidemiology and BiostatisticsSchool of Public Health, Imperial College LondonLondonUnited Kingdom
| | - Devendra Meena
- Department of Epidemiology and BiostatisticsSchool of Public Health, Imperial College LondonLondonUnited Kingdom
| | - James Yarmolinsky
- Department of Epidemiology and BiostatisticsSchool of Public Health, Imperial College LondonLondonUnited Kingdom
| | - Dipender Gill
- Department of Epidemiology and BiostatisticsSchool of Public Health, Imperial College LondonLondonUnited Kingdom
| | - Alexander Smith
- Department of Epidemiology and BiostatisticsSchool of Public Health, Imperial College LondonLondonUnited Kingdom
| | - Marie‐Joe Dib
- Division of Cardiovascular MedicineHospital of the University of PennsylvaniaPhiladelphiaPAUSA
| | - Ganesh Chauhan
- Department of Genetics & GenomicsRajendra Institute of Medical Sciences (RIMS)RanchiIndia
| | - Anand Rohatgi
- Department of Medicine, Division of CardiologyUniversity of Texas Southwestern Medical CenterDallasTXUSA
| | - Abbas Dehghan
- Department of Epidemiology and BiostatisticsSchool of Public Health, Imperial College LondonLondonUnited Kingdom
- Dementia Research Institute, Imperial College LondonLondonUnited Kingdom
| | - Ioanna Tzoulaki
- Department of Epidemiology and BiostatisticsSchool of Public Health, Imperial College LondonLondonUnited Kingdom
- Dementia Research Institute, Imperial College LondonLondonUnited Kingdom
- Biomedical Research FoundationAcademy of AthensAthensGreece
| |
Collapse
|
3
|
Lahu S, Adler K, Mayer K, Hein-Rothweiler R, Bernlochner I, Ndrepepa G, Schüpke S, Holdenrieder S, Bongiovanni D, Laugwitz KL, Schunkert H, Gawaz M, Massberg S, Kastrati A, Münch G. Plasma Soluble Glycoprotein VI, Platelet Function, Bleeding, and Ischemic Events in Patients Undergoing Elective Percutaneous Coronary Intervention. Thromb Haemost 2024; 124:297-306. [PMID: 37591289 DOI: 10.1055/s-0043-1772221] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/19/2023]
Abstract
BACKGROUND AND AIMS Glycoprotein VI (GPVI) is the major platelet-specific collagen receptor. GPVI shedding with generation of soluble GPVI (sGPVI) is an endogenous feedback mechanism preventing platelet overstimulation. sGPVI has not been investigated in patients with chronic coronary syndrome (CCS) undergoing percutaneous coronary intervention (PCI), especially regarding its potential value as a predictor of ischemic and bleeding risk. METHODS Baseline plasma sGPVI levels were available in 318 patients with CCS undergoing PCI. Platelet function was assessed by measuring both adenosine diphosphate (ADP) and collagen-induced platelet aggregation. Co-primary endpoints were a composite of death or myocardial injury at 48 hours after PCI, and Bleeding Academic Research Consortium (BARC) type 1 to 5 bleeding at 30 days. RESULTS There was no significant correlation between sGPVI and platelet function at baseline or at 48 hours after PCI and loading with antiplatelet drugs. Baseline plasma sGPVI levels were not associated with the ischemic risk: the incidence of the ischemic endpoint was 25.0% in the lower, 22.9% in the middle, and 26.7% in the upper sGPVI tertile (p = 0.82). There was a significant nonlinear relationship between sGPVI and the risk of bleeding: the incidence of the bleeding endpoint was 11.8% in the lower, 12.6% in the middle, and 26.4% in the upper sGPVI tertile (p = 0.006). CONCLUSION In patients with CCS undergoing PCI, plasma levels of sGPVI did not correlate with ADP- or collagen-induced platelet aggregation. Patients with higher baseline levels of sGPVI may carry an increased risk of bleeding at 30 days after PCI but no excess risk of ischemic events.
Collapse
Affiliation(s)
- Shqipdona Lahu
- Department of Cardiology, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany
| | | | - Katharina Mayer
- Department of Cardiology, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Ralph Hein-Rothweiler
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
- Department of Cardiology, Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians-Universität, Munich, Germany
| | - Isabell Bernlochner
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
- Klinik und Poliklinik für Innere Medizin I, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Gjin Ndrepepa
- Department of Cardiology, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany
| | - Stefanie Schüpke
- Department of Cardiology, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
- Privatpraxis für Kardiologie, Kaiserstr. 10, 60311 Frankfurt am Main, Germany
| | - Stefan Holdenrieder
- Institut für Laboratoriumsmedizin, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany
| | - Dario Bongiovanni
- Klinik für Kardiologie, Pneumologie, Endokrinologie, Intensivmedizin, Universitätsklinikum Augsburg, Augsburg, Germany
| | - Karl-Ludwig Laugwitz
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
- Klinik und Poliklinik für Innere Medizin I, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Heribert Schunkert
- Department of Cardiology, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Meinrad Gawaz
- Department of Cardiology and Angiology, University of Tübingen, Tübingen, Germany
| | - Steffen Massberg
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
- Department of Cardiology, Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians-Universität, Munich, Germany
| | - Adnan Kastrati
- Department of Cardiology, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | | |
Collapse
|
4
|
Aggarwal A, Singh TK, Pham M, Godwin M, Chen R, McIntyre TM, Scalise A, Chung MK, Jennings C, Ali M, Park H, Englund K, Khorana AA, Svensson LG, Kapadia S, McCrae KR, Cameron SJ. Dysregulated platelet function in patients with postacute sequelae of COVID-19. Vasc Med 2024; 29:125-134. [PMID: 38334067 PMCID: PMC11164201 DOI: 10.1177/1358863x231224383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2024]
Abstract
BACKGROUND Postacute sequelae of COVID-19 (PASC), also referred to as "Long COVID", sometimes follows COVID-19, a disease caused by SARS-CoV-2. Although SARS-CoV-2 is well known to promote a prothrombotic state, less is known about the thrombosis risk in PASC. Our objective was to evaluate platelet function and thrombotic potential in patients following recovery from SARS-CoV-2, but with clear symptoms of patients with PASC. METHODS patients with PASC and matched healthy controls were enrolled in the study on average 15 months after documented SARS-CoV-2 infection. Platelet activation was evaluated by light transmission aggregometry (LTA) and flow cytometry in response to platelet surface receptor agonists. Thrombosis in platelet-deplete plasma was evaluated by Factor Xa activity. A microfluidics system assessed thrombosis in whole blood under shear stress conditions. RESULTS A mild increase in platelet aggregation in patients with PASC through the thromboxane receptor was observed, and platelet activation through the glycoprotein VI (GPVI) receptor was decreased in patients with PASC compared to age- and sex-matched healthy controls. Thrombosis under shear conditions as well as Factor Xa activity were reduced in patients with PASC. Plasma from patients with PASC was an extremely potent activator of washed, healthy platelets - a phenomenon not observed when stimulating healthy platelets after incubation with plasma from healthy individuals. CONCLUSIONS patients with PASC show dysregulated responses in platelets and coagulation in plasma, likely caused by a circulating molecule that promotes thrombosis. A hitherto undescribed protective response appears to exist in patients with PASC to counterbalance ongoing thrombosis that is common to SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Anu Aggarwal
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Tamanna K Singh
- Section of Vascular Medicine, Department of Cardiovascular Medicine, Heart Vascular and Thoracic Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Michael Pham
- Section of Vascular Medicine, Department of Cardiovascular Medicine, Heart Vascular and Thoracic Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Matthew Godwin
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Rui Chen
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Thomas M McIntyre
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Alliefair Scalise
- Section of Vascular Medicine, Department of Cardiovascular Medicine, Heart Vascular and Thoracic Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Mina K Chung
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
- Section of Vascular Medicine, Department of Cardiovascular Medicine, Heart Vascular and Thoracic Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Courtney Jennings
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Mariya Ali
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Hiijun Park
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Kristin Englund
- Department of Infectious Disease, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Alok A Khorana
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
- Department of Hematology and Oncology, Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Lars G Svensson
- Department of Cardiac Surgery, Heart Vascular and Thoracic Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Samir Kapadia
- Section of Vascular Medicine, Department of Cardiovascular Medicine, Heart Vascular and Thoracic Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Keith R McCrae
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
- Section of Vascular Medicine, Department of Cardiovascular Medicine, Heart Vascular and Thoracic Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
- Department of Hematology and Oncology, Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Scott J Cameron
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
- Section of Vascular Medicine, Department of Cardiovascular Medicine, Heart Vascular and Thoracic Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
- Department of Hematology and Oncology, Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| |
Collapse
|
5
|
Aggarwal A, Singh TK, Pham M, Godwin M, Chen R, McIntyre TM, Scalise A, Chung MK, Jennings C, Ali M, Park H, Englund K, Khorana AA, Svensson LG, Kapadia S, McCrae KR, Cameron SJ. Dysregulated Platelet Function in Patients with Post-Acute Sequelae of COVID-19. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.18.545507. [PMID: 38045316 PMCID: PMC10690211 DOI: 10.1101/2023.06.18.545507] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Background Post-acute sequelae of COVID-19 (PASC), also referred as Long-COVID, sometimes follows COVID-19, a disease caused by SARS-CoV-2. While SARS-CoV-2 is well-known to promote a prothrombotic state, less is known about the thrombosis risk in PASC. Aim Our objective was to evaluate the platelet function and thrombotic potential in patients following recovery from SARS-CoV-2 with clear symptoms of PASC. Methods PASC patients and matched healthy controls were enrolled in the study on average 15 months after documented SARS-CoV-2 infection. Platelet activation was evaluated by Light Transmission Aggregometry (LTA) and flow cytometry in response to platelet surface receptor agonists. Thrombosis in platelet-deplete plasma was evaluated by Factor Xa activity. A microfluidics system assessed thrombosis in whole blood under shear stress conditions. Results A mild increase in platelet aggregation in PASC patients through the thromboxane receptor was observed and platelet activation through the glycoprotein VI (GPVI) receptor was decreased in PASC patients compared to age- and sex-matched healthy controls. Thrombosis under shear conditions as well as Factor Xa activity were reduced in PASC patients. Plasma from PASC patients was an extremely potent activator of washed, healthy platelets - a phenomenon not observed when stimulating healthy platelets after incubation with plasma from healthy individuals. Conclusions PASC patients show dysregulated responses in platelets and coagulation in plasma, likely caused by a circulating molecule that promotes thrombosis. A hitherto undescribed protective response appears to exists in PASC patients to counterbalance ongoing thrombosis that is common to SARS-CoV-2 infection.
Collapse
|
6
|
Hearn JI, Gardiner EE. Research and Clinical Approaches to Assess Platelet Function in Flowing Blood. Arterioscler Thromb Vasc Biol 2023; 43:1775-1783. [PMID: 37615110 DOI: 10.1161/atvbaha.123.317048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/25/2023]
Abstract
Platelet adhesion and activation is fundamental to the formation of a hemostatic response to limit loss of blood and instigate wound repair to seal a site of vascular injury. The process of platelet aggregate formation is supported by the coagulation system driving injury-proximal formation of thrombin, which converts fibrinogen to insoluble fibrin. This highly coordinated series of molecular and membranous events must be routinely achieved in flowing blood, at vascular fluid shear rates that place significant strain on molecular and cellular interactions. Platelets have long been recognized to be able to slow down and adhere to sites of vascular injury and then activate and recruit more platelets that forge and strengthen adhesive ties with the vascular wall under these conditions. It has been a major challenge for the Platelet Research Community to construct experimental conditions that allow precise definition of the molecular steps occurring under flow. This brief review will discuss work to date from our group, as well as others that has furthered our understanding of platelet function in flowing blood.
Collapse
Affiliation(s)
- James I Hearn
- Division of Genome Science and Cancer, John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Elizabeth E Gardiner
- Division of Genome Science and Cancer, John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| |
Collapse
|
7
|
Abdulrahman B, Jabbour RJ, Curzen N. Is It Really Safe to Discontinue Antiplatelet Therapy 12 Months After Percutaneous Coronary Intervention in Patients with Atrial Fibrillation? Interv Cardiol 2023; 18:e22. [PMID: 37435601 PMCID: PMC10331563 DOI: 10.15420/icr.2022.40] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 03/29/2023] [Indexed: 07/13/2023] Open
Abstract
The prevalence of AF in patients with coronary artery disease is high. The guidelines from many professional groups, including the European Society of Cardiology, American College of Cardiology/American Heart Association and Heart Rhythm Society, recommend a maximum duration of 12 months of combination single antiplatelet and anticoagulation therapy in patients who undergo percutaneous coronary intervention and who have concurrent AF, followed by anticoagulation alone beyond 1 year. However, the evidence that anticoagulation alone without antiplatelet therapy adequately reduces the well-documented attritional risk of stent thrombosis after coronary stent implantation is relatively sparse, particularly given that very late stent thrombosis (>1 year from stent implantation) is the commonest type. By contrast, the elevated risk of bleeding from combined anticoagulation and antiplatelet therapy is clinically important. The aim of this review is to assess the evidence for long-term anticoagulation alone without antiplatelet therapy 1 year post-percutaneous coronary intervention in patients with AF.
Collapse
Affiliation(s)
- Balen Abdulrahman
- Coronary Research Group, University Hospital Southampton NHS Foundation TrustSouthampton, UK
| | - Richard J Jabbour
- Coronary Research Group, University Hospital Southampton NHS Foundation TrustSouthampton, UK
| | - Nick Curzen
- Coronary Research Group, University Hospital Southampton NHS Foundation TrustSouthampton, UK
- Faculty of Medicine, University of SouthamptonSouthampton, UK
| |
Collapse
|
8
|
De Simone I, Baaten CCFMJ, Jandrot-Perrus M, Gibbins JM, ten Cate H, Heemskerk JWM, Jones CI, van der Meijden PEJ. Coagulation Factor XIIIa and Activated Protein C Activate Platelets via GPVI and PAR1. Int J Mol Sci 2022; 23:ijms231810203. [PMID: 36142125 PMCID: PMC9499330 DOI: 10.3390/ijms231810203] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 08/24/2022] [Accepted: 08/30/2022] [Indexed: 11/16/2022] Open
Abstract
Platelet and coagulation activation are highly reciprocal processes driven by multi-molecular interactions. Activated platelets secrete several coagulation factors and expose phosphatidylserine, which supports the activation of coagulation factor proteins. On the other hand, the coagulation cascade generates known ligands for platelet receptors, such as thrombin and fibrin. Coagulation factor (F)Xa, (F)XIIIa and activated protein C (APC) can also bind to platelets, but the functional consequences are unclear. Here, we investigated the effects of the activated (anti)coagulation factors on platelets, other than thrombin. Multicolor flow cytometry and aggregation experiments revealed that the ‘supernatant of (hirudin-treated) coagulated plasma’ (SCP) enhanced CRP-XL-induced platelet responses, i.e., integrin αIIbβ3 activation, P-selectin exposure and aggregate formation. We demonstrated that FXIIIa in combination with APC enhanced platelet activation in solution, and separately immobilized FXIIIa and APC resulted in platelet spreading. Platelet activation by FXIIIa was inhibited by molecular blockade of glycoprotein VI (GPVI) or Syk kinase. In contrast, platelet spreading on immobilized APC was inhibited by PAR1 blockade. Immobilized, but not soluble, FXIIIa and APC also enhanced in vitro adhesion and aggregation under flow. In conclusion, in coagulation, factors other than thrombin or fibrin can induce platelet activation via GPVI and PAR receptors.
Collapse
Affiliation(s)
- Ilaria De Simone
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6200 MD Maastricht, The Netherlands
- School of Biological Sciences, Institute for Metabolic and Cardiovascular Research, University of Reading, Reading RG6 6AS, UK
| | - Constance C. F. M. J. Baaten
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6200 MD Maastricht, The Netherlands
- Institute for Molecular Cardiovascular Research, University Hospital Aachen, RWTH Aachen University, 52074 Aachen, Germany
| | - Martine Jandrot-Perrus
- UMR_S1148, Laboratory for Vascular Translational Science, INSERM, University Paris Cité, F-75018 Paris, France
| | - Jonathan M. Gibbins
- School of Biological Sciences, Institute for Metabolic and Cardiovascular Research, University of Reading, Reading RG6 6AS, UK
| | - Hugo ten Cate
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6200 MD Maastricht, The Netherlands
- Thrombosis Expertise Center, Heart and Vascular Center, Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands
| | - Johan W. M. Heemskerk
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6200 MD Maastricht, The Netherlands
- Synapse Research Institute, 6217 KD Maastricht, The Netherlands
| | - Chris I. Jones
- School of Biological Sciences, Institute for Metabolic and Cardiovascular Research, University of Reading, Reading RG6 6AS, UK
- Correspondence: (C.I.J.); (P.E.J.v.d.M.); Tel.: +44-(0)-118-378-7047 (C.I.J.); +31-43-388-1684 (P.E.J.v.d.M.)
| | - Paola E. J. van der Meijden
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6200 MD Maastricht, The Netherlands
- Thrombosis Expertise Center, Heart and Vascular Center, Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands
- Correspondence: (C.I.J.); (P.E.J.v.d.M.); Tel.: +44-(0)-118-378-7047 (C.I.J.); +31-43-388-1684 (P.E.J.v.d.M.)
| |
Collapse
|
9
|
Platelet activation and partial desensitization are associated with viral xenophagy in patients with severe COVID-19. Blood Adv 2022; 6:3884-3898. [PMID: 35789374 PMCID: PMC9068266 DOI: 10.1182/bloodadvances.2022007143] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 04/19/2022] [Indexed: 12/17/2022] Open
Abstract
During severe COVID-19, platelets get activated and become partly desensitized through mechanisms involving glycoprotein shedding. Platelets from patients with severe COVID-19 internalize SARS-CoV-2 and develop viral xenophagy.
Mild thrombocytopenia, changes in platelet gene expression, enhanced platelet functionality, and presence of platelet-rich thrombi in the lung have been associated with thromboinflammatory complications of patients with COVID-19. However, whether severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) gets internalized by platelets and directly alters their behavior and function in infected patients remains elusive. Here, we investigated platelet parameters and the presence of viral material in platelets from a prospective cohort of 29 patients with severe COVID-19 admitted to an intensive care unit. A combination of specific assays, tandem mass spectrometry, and flow cytometry indicated high levels of protein and lipid platelet activation markers in the plasma from patients with severe COVID-19 associated with an increase of proinflammatory cytokines and leukocyte-platelets interactions. Platelets were partly desensitized, as shown by a significant reduction of αIIbβ3 activation and granule secretion in response to stimulation and a decrease of surface GPVI, whereas plasma from patients with severe COVID-19 potentiated washed healthy platelet aggregation response. Transmission electron microscopy indicated the presence of SARS-CoV-2 particles in a significant fraction of platelets as confirmed by immunogold labeling and immunofluorescence imaging of Spike and nucleocapsid proteins. Compared with platelets from healthy donors or patients with bacterial sepsis, platelets from patients with severe COVID-19 exhibited enlarged intracellular vesicles and autophagolysosomes. They had large LC3-positive structures and increased levels of LC3II with a co-localization of LC3 and Spike, suggesting that platelets can digest SARS-CoV-2 material by xenophagy in critically ill patients. Altogether, these data show that during severe COVID-19, platelets get activated, become partly desensitized, and develop a selective autophagy response.
Collapse
|
10
|
Koo CZ, Matthews AL, Harrison N, Szyroka J, Nieswandt B, Gardiner EE, Poulter NS, Tomlinson MG. The Platelet Collagen Receptor GPVI Is Cleaved by Tspan15/ADAM10 and Tspan33/ADAM10 Molecular Scissors. Int J Mol Sci 2022; 23:2440. [PMID: 35269584 PMCID: PMC8910667 DOI: 10.3390/ijms23052440] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 02/13/2022] [Accepted: 02/14/2022] [Indexed: 02/01/2023] Open
Abstract
The platelet-activating collagen receptor GPVI represents the focus of clinical trials as an antiplatelet target for arterial thrombosis, and soluble GPVI is a plasma biomarker for several human diseases. A disintegrin and metalloproteinase 10 (ADAM10) acts as a 'molecular scissor' that cleaves the extracellular region from GPVI and many other substrates. ADAM10 interacts with six regulatory tetraspanin membrane proteins, Tspan5, Tspan10, Tspan14, Tspan15, Tspan17 and Tspan33, which are collectively termed the TspanC8s. These are emerging as regulators of ADAM10 substrate specificity. Human platelets express Tspan14, Tspan15 and Tspan33, but which of these regulates GPVI cleavage remains unknown. To address this, CRISPR/Cas9 knockout human cell lines were generated to show that Tspan15 and Tspan33 enact compensatory roles in GPVI cleavage, with Tspan15 bearing the more important role. To investigate this mechanism, a series of Tspan15 and GPVI mutant expression constructs were designed. The Tspan15 extracellular region was found to be critical in promoting GPVI cleavage, and appeared to achieve this by enabling ADAM10 to access the cleavage site at a particular distance above the membrane. These findings bear implications for the regulation of cleavage of other ADAM10 substrates, and provide new insights into post-translational regulation of the clinically relevant GPVI protein.
Collapse
Affiliation(s)
- Chek Ziu Koo
- School of Biosciences, University of Birmingham, Birmingham B15 2TT, UK; (C.Z.K.); (A.L.M.); (N.H.); (J.S.)
- Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, Midlands B15 2TT, UK;
| | - Alexandra L. Matthews
- School of Biosciences, University of Birmingham, Birmingham B15 2TT, UK; (C.Z.K.); (A.L.M.); (N.H.); (J.S.)
- Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, Midlands B15 2TT, UK;
| | - Neale Harrison
- School of Biosciences, University of Birmingham, Birmingham B15 2TT, UK; (C.Z.K.); (A.L.M.); (N.H.); (J.S.)
| | - Justyna Szyroka
- School of Biosciences, University of Birmingham, Birmingham B15 2TT, UK; (C.Z.K.); (A.L.M.); (N.H.); (J.S.)
| | - Bernhard Nieswandt
- Institute of Experimental Biomedicine I, University Hospital and Rudolf Virchow Center Würzburg, University of Würzburg, D-97080 Würzburg, Germany;
| | - Elizabeth E. Gardiner
- Division of Genome Science and Cancer, John Curtin School of Medical Research, Australian National University, Canberra ACT 2601, Australia;
| | - Natalie S. Poulter
- Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, Midlands B15 2TT, UK;
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Michael G. Tomlinson
- School of Biosciences, University of Birmingham, Birmingham B15 2TT, UK; (C.Z.K.); (A.L.M.); (N.H.); (J.S.)
- Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, Midlands B15 2TT, UK;
| |
Collapse
|
11
|
Ishikura H, Irie Y, Kawamura M, Hoshino K, Nakamura Y, Mizunuma M, Maruyama J, Nakashio M, Suzuki-Inoue K, Kitamura T. Early recognition of sepsis-induced coagulopathy using the C2PAC index: a ratio of soluble type C lectin-like receptor 2 (sCLEC-2) level and platelet count. Platelets 2022; 33:935-944. [PMID: 35073814 DOI: 10.1080/09537104.2021.2019694] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
C-type lectin-like receptor 2 (CLEC-2) is a platelet-activated receptor expressed on the surface of platelet membranes. Soluble CLEC-2 (sCLEC-2) has been receiving attention as a predictive marker for thrombotic predisposition. The present study examined the relationship between sCLEC-2 level and degree of coagulation disorder in septic patients. Seventy septic patients were divided into the sepsis-induced disseminated intravascular coagulation (DIC) (SID) group (n = 44) and non-SID group (n = 26). The sCLEC-2 levels were compared between the two groups. Because we suspected that the sCLEC-2 level was affected by the platelet count, we calculated the sCLEC-2/platelet count ratio (C2PAC index). We further divided septic patients into four groups using the Japanese Association for Acute Medicine (JAAM) DIC scoring system (DIC scores: 0-1, 2-3, 4-5, and 6-8). The C2PAC index was significantly higher in the SID group (2.6 ± 1.7) compared with the non-SID group (1.2 ± 0.5) (P < .001). The C2PAC indexes in the four JAAM DIC score groups were 0.9 ± 0.3, 1.1 ± 0.3, 1.7 ± 0.7, and 3.6 ± 1.0, respectively, and this index increased significantly as the DIC score increased (P < .001). According to the receiver-operating curve analysis, the area under the curve (AUC) and optimal cutoff value for the diagnosis of SID were 0.8051 and 1.4 (sensitivity, 75.0%; specificity, 76.9%), respectively. When the C2PAC index and D-dimer level, one of the main fibrinolytic markers, were selected as predictive markers for SID diagnosis in stepwise multiple logistic regression analysis, it was possible to diagnose SID with a high probability (AUC, 0.9528; sensitivity, 0.9545; specificity, 0.8846). The C2PAC index is a useful predictor of SID progression and diagnosis in septic patients.
Collapse
Affiliation(s)
- Hiroyasu Ishikura
- Department of Emergency and Critical Care Medicine, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Yuhei Irie
- Department of Emergency and Critical Care Medicine, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Masahide Kawamura
- Department of Research and Development, IVD Business Segment, LSI Medience Corporation, Tokyo, Japan
| | - Kota Hoshino
- Department of Emergency and Critical Care Medicine, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Yoshihiko Nakamura
- Department of Emergency and Critical Care Medicine, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Mariko Mizunuma
- Department of Emergency and Critical Care Medicine, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Junichi Maruyama
- Department of Emergency and Critical Care Medicine, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Maiko Nakashio
- Department of Emergency and Critical Care Medicine, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Katsue Suzuki-Inoue
- Department of Clinical and Laboratory Medicine, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Taisuke Kitamura
- Department of Emergency and Critical Care Medicine, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| |
Collapse
|
12
|
Perrella G, Nagy M, Watson SP, Heemskerk JWM. Platelet GPVI (Glycoprotein VI) and Thrombotic Complications in the Venous System. Arterioscler Thromb Vasc Biol 2021; 41:2681-2692. [PMID: 34496636 PMCID: PMC9653110 DOI: 10.1161/atvbaha.121.316108] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
The immunoglobulin receptor GPVI (glycoprotein VI) is selectively expressed on megakaryocytes and platelets and is currently recognized as a receptor for not only collagen but also a variety of plasma and vascular proteins, including fibrin, fibrinogen, laminin, fibronectin, and galectin-3. Deficiency of GPVI is protective in mouse models of experimental thrombosis, pulmonary thromboembolism as well as in thromboinflammation, suggesting a role of GPVI in arterial and venous thrombus formation. In humans, platelet GPVI deficiency is associated with a mild bleeding phenotype, whereas a common variant rs1613662 in the GP6 gene is considered a risk factor for venous thromboembolism. However, preclinical studies on the inhibition of GPVI-ligand interactions are focused on arterial thrombotic complications. In this review we discuss the emerging evidence for GPVI in venous thrombus formation and leukocyte-dependent thromboinflammation, extending to venous thromboembolism, pulmonary thromboembolism, and cancer metastasis. We also recapitulate indications for circulating soluble GPVI as a biomarker of thrombosis-related complications. Collectively, we conclude that the current evidence suggests that platelet GPVI is also a suitable cotarget in the prevention of venous thrombosis due to its role in thrombus consolidation and platelet-leukocyte complex formation.
Collapse
Affiliation(s)
- Gina Perrella
- Department of Biochemistry, CARIM, Maastricht University, The Netherlands (G.P., M.N., J.W.M.H.).,Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, United Kingdom (G.P., S.P.W.)
| | - Magdolna Nagy
- Department of Biochemistry, CARIM, Maastricht University, The Netherlands (G.P., M.N., J.W.M.H.)
| | - Steve P Watson
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, United Kingdom (G.P., S.P.W.).,COMPARE, 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., M.N., J.W.M.H.).,Now with Synapse Research Institute, Maastricht, the Netherlands (J.W.M.H.)
| |
Collapse
|
13
|
Yamamoto A, Wada H, Ichkawa Y, Tanaka M, Tashiro H, Shiraki K, Shimpo H, Yamashita Y, Mastumoto T, Shimaoka M, Iba T, Suzuki-Inoue K. Soluble C-Type Lectin-Like Receptor 2 Is a Biomarker for Disseminated Intravascular Coagulation. J Clin Med 2021; 10:jcm10132860. [PMID: 34203210 PMCID: PMC8269023 DOI: 10.3390/jcm10132860] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/23/2021] [Accepted: 06/24/2021] [Indexed: 12/24/2022] Open
Abstract
Disseminated intravascular coagulation (DIC) is induced by excess activation coagulation, and activated platelets are also involved in pathogenesis. Therefore, plasma levels of soluble C-type lectin-like receptor 2 (sCLEC-2), a new marker for platelet activation, can be expected as a marker of DIC in critically ill patients. Plasma levels of sCLEC-2 and D-dimer were measured using the STACIA system. Plasma sCLEC-2 and D-dimer levels were significantly higher in patients with underlying diseases of DIC than in those with unidentified clinical syndrome (UCS). Plasma sCLEC-2 levels were significantly higher in the patients with DIC and Pre-DIC than in those without DIC or Pre-DIC. Similarly, plasma D-dimer levels were also significantly higher in patients with DIC and Pre-DIC than in those without DIC or Pre-DIC. The plasma sCLEC-2 levels in all patients and those with a DIC score ≤ 4 were significantly higher in non-survivors than survivors. The plasma D-dimer levels in all patients, those with a DIC score ≥ 5 and those with a DIC score ≤ 4, were significantly higher in non-survivors than in survivors. The plasma sCLEC-2 is expected as a marker for DIC/Pre-DIC as well as the prognostic marker in critically ill patients.
Collapse
Affiliation(s)
- Akitaka Yamamoto
- Department of Emergency and Critical Care Center, Mie Prefectural General Medical Center, Yokkaichi 510-0885, Japan; (A.Y.); (H.T.)
| | - Hideo Wada
- Department of Central Laboratory, Mie Prefectural General Medical Center, Yokkaichi 510-0885, Japan; (Y.I.); (M.T.); (K.S.)
- Department of General Medicine, Mie Prefectural General Medical Center, Yokkaichi 510-0885, Japan
- Correspondence: ; Tel.: +81-59-345-2321
| | - Yuhuko Ichkawa
- Department of Central Laboratory, Mie Prefectural General Medical Center, Yokkaichi 510-0885, Japan; (Y.I.); (M.T.); (K.S.)
| | - Motoko Tanaka
- Department of Central Laboratory, Mie Prefectural General Medical Center, Yokkaichi 510-0885, Japan; (Y.I.); (M.T.); (K.S.)
| | - Haruhiko Tashiro
- Department of Emergency and Critical Care Center, Mie Prefectural General Medical Center, Yokkaichi 510-0885, Japan; (A.Y.); (H.T.)
| | - Katsuya Shiraki
- Department of Central Laboratory, Mie Prefectural General Medical Center, Yokkaichi 510-0885, Japan; (Y.I.); (M.T.); (K.S.)
- Department of General Medicine, Mie Prefectural General Medical Center, Yokkaichi 510-0885, Japan
| | - Hideto Shimpo
- Mie Prefectural General Medical Center, Yokkaichi 510-0885, Japan;
| | - Yoshiki Yamashita
- Department of Hematology and Oncology, Mie University Hospital, Mie University Graduate School of Medicine, Tsu 514-8507, Japan;
| | - Takeshi Mastumoto
- Department of Blood Transfusion and Cell Therapy, Mie University Hospital, Mie University Graduate School of Medicine, Tsu 514-8507, Japan;
| | - Motomu Shimaoka
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, Tsu 514-8507, Japan;
| | - Toshiaki Iba
- Department of Emergency and Disaster Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan;
| | - Katsue Suzuki-Inoue
- Department of Clinical and Laboratory Medicine, Yamanashi Medical University, Yamanashi 400-8510, Japan;
| |
Collapse
|
14
|
Loss of GPVI and GPIbα contributes to trauma-induced platelet dysfunction in severely injured patients. Blood Adv 2021; 4:2623-2630. [PMID: 32556282 DOI: 10.1182/bloodadvances.2020001776] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 05/17/2020] [Indexed: 11/20/2022] Open
Abstract
Trauma-induced coagulopathy (TIC) is a complex, multifactorial failure of hemostasis that occurs in 25% of severely injured patients and results in a fourfold higher mortality. However, the role of platelets in this state remains poorly understood. We set out to identify molecular changes that may underpin platelet dysfunction after major injury and to determine how they relate to coagulopathy and outcome. We performed a range of hemostatic and platelet-specific studies in blood samples obtained from critically injured patients within 2 hours of injury and collected prospective data on patient characteristics and clinical outcomes. We observed that, although platelet counts were preserved above critical levels, circulating platelets sampled from trauma patients exhibited a profoundly reduced response to both collagen and the selective glycoprotein VI (GPVI) agonist collagen-related peptide, compared with those from healthy volunteers. These responses correlated closely with overall clot strength and mortality. Surface expression of the collagen receptors GPIbα and GPVI was reduced on circulating platelets in trauma patients, with increased levels of the shed ectodomain fragment of GPVI detectable in plasma. Levels of shed GPVI were highest in patients with more severe injuries and TIC. Collectively, these observations demonstrate that platelets experience a loss of GPVI and GPIbα after severe injury and translate into a reduction in the responsiveness of platelets during active hemorrhage. In turn, they are associated with reduced hemostatic competence and increased mortality. Targeting proteolytic shedding of platelet receptors is a potential therapeutic strategy for maintaining hemostatic competence in bleeding and improving the efficacy of platelet transfusions.
Collapse
|
15
|
Dannenberg L, M'Pembele R, Mourikis P, Helten C, Zako S, Ahlbrecht S, Richter H, Zikeli D, Benkhoff M, Huhn-Wientgen R, Thienel M, Levkau B, Kelm M, Petzold T, Polzin A. Rivaroxaban reduces thromboxane induced platelet aggregation - the forgotten Compass Arm? Platelets 2021; 32:1126-1128. [PMID: 33788669 DOI: 10.1080/09537104.2021.1905159] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Background: Recent guidelines only recommend 'vascular dose' rivaroxaban in combination with aspirin in chronic coronary syndrome (CCS) patients with high risk of ischemic events However, in the COMPASS trial, a reduction of MACCE appeared for low-dose rivaroxaban alone compared to aspirin as well. It was recently shown that FXa induces platelet aggregation via protease activated receptor 1 (PAR-1) which is in turn attenuated by rivaroxaban. However, a potential impact of rivaroxaban on TX B2 formation is unclear.Methods and Results: TX B2 levels were measured in supernatant from washed platelets after FXa (52 µg/ml) induced platelet aggregation. TX B2 levels were significantly higher in supernatant from FXa-stimulated platelets compared to unstimulated control (Control 23.53 ± 14.15 ng/ml vs. FXa stimulated 77.4 ± 64.14 ng/ml; p = .0025). This effect was abolished in the presence of 100pM rivaroxaban (Control 23.53 ± 14.15 ng/ml vs. FXa stimulated and rivaroxaban 22.15 ± 24.74 ng/ml; p = .5142). Next, we investigated the effects of 100pM rivaroxaban on platelet aggregation induced by U46619 (TX receptor agonist) using light transmission aggregometry. Platelet aggregation quantified by maximum of aggregation (MoA%) was significantly lower in presence of rivaroxaban (U46619 40.18 ± 20.51% vs. U46619+ rivaroxaban 19.26 ± 15.46%; p = .0274).Conclusion: Our results indicate direct effects of rivaroxaban on the cyclooxygenase-1- TX axis during platelet aggregation. Hence, it seems reasonable that the 'forgotten compass arm' (rivaroxaban alone) might be an alternative to the rivaroxaban plus aspirin combination in CCS patients.
Collapse
Affiliation(s)
- Lisa Dannenberg
- Cardiovascular Research Institute Düsseldorf (CARID), Division of Cardiology, Pulmonology, and Vascular Medicine, University Duesseldorf, Duesseldorf, Germany
| | - René M'Pembele
- Department of Anesthesiology, University Hospital Duesseldorf, Duesseldorf, Germany
| | - Philipp Mourikis
- Cardiovascular Research Institute Düsseldorf (CARID), Division of Cardiology, Pulmonology, and Vascular Medicine, University Duesseldorf, Duesseldorf, Germany
| | - Carolin Helten
- Cardiovascular Research Institute Düsseldorf (CARID), Division of Cardiology, Pulmonology, and Vascular Medicine, University Duesseldorf, Duesseldorf, Germany
| | - Saif Zako
- Cardiovascular Research Institute Düsseldorf (CARID), Division of Cardiology, Pulmonology, and Vascular Medicine, University Duesseldorf, Duesseldorf, Germany
| | - Samantha Ahlbrecht
- Cardiovascular Research Institute Düsseldorf (CARID), Division of Cardiology, Pulmonology, and Vascular Medicine, University Duesseldorf, Duesseldorf, Germany
| | - Hannah Richter
- Cardiovascular Research Institute Düsseldorf (CARID), Division of Cardiology, Pulmonology, and Vascular Medicine, University Duesseldorf, Duesseldorf, Germany
| | - Dorothee Zikeli
- Cardiovascular Research Institute Düsseldorf (CARID), Division of Cardiology, Pulmonology, and Vascular Medicine, University Duesseldorf, Duesseldorf, Germany
| | - Marcel Benkhoff
- Cardiovascular Research Institute Düsseldorf (CARID), Division of Cardiology, Pulmonology, and Vascular Medicine, University Duesseldorf, Duesseldorf, Germany
| | - Ragnar Huhn-Wientgen
- Department of Anesthesiology, University Hospital Duesseldorf, Duesseldorf, Germany
| | - Manuela Thienel
- Medizinische Klinik Und Poliklinik I, Klinikum Der Universität München, Ludwig-Maximilians- University Munich, Germany
| | - Bodo Levkau
- Institute of Molecular Medicine III, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Malte Kelm
- Cardiovascular Research Institute Düsseldorf (CARID), Division of Cardiology, Pulmonology, and Vascular Medicine, University Duesseldorf, Duesseldorf, Germany
| | - Tobias Petzold
- Medizinische Klinik Und Poliklinik I, Klinikum Der Universität München, Ludwig-Maximilians- University Munich, Germany
| | - Amin Polzin
- Cardiovascular Research Institute Düsseldorf (CARID), Division of Cardiology, Pulmonology, and Vascular Medicine, University Duesseldorf, Duesseldorf, Germany
| |
Collapse
|
16
|
Pastori D, Menichelli D, Del Sole F, Pignatelli P, Violi F. Long-Term Risk of Major Adverse Cardiac Events in Atrial Fibrillation Patients on Direct Oral Anticoagulants. Mayo Clin Proc 2021; 96:658-665. [PMID: 33308867 DOI: 10.1016/j.mayocp.2020.06.057] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 06/22/2020] [Indexed: 12/20/2022]
Abstract
OBJECTIVE To determine the association between direct oral anticoagulant (DOAC) use and risk of major adverse cardiac events (MACEs) in patients with atrial fibrillation (AF). PATIENTS AND METHODS This study is a single-center prospective observational cohort study including 2366 outpatients with non-valvular AF on treatment with DOACs or vitamin K antagonists (VKAs) from February 2008 for patients on VKA and September 2013 for patients on novel oral anticoagulants. The primary endpoint was the incidence of MACE including fatal and non-fatal myocardial infarction (MI), cardiac revascularization, and cardiovascular death. RESULTS The mean age was 75.1±9.0 years; 44.7% were women. During a mean follow-up of 33.3±21.9 months (6567 patients/years) 133 MACEs occurred (2.03%/year): 79 MI/cardiac revascularization and 54 cardiovascular deaths. Of these, 101 were on VKAs (2.42%/year) and 32 on DOACs (1.34%/year; log-rank test P=.040). This difference was evident also considering MI alone (1.53%/year and 0.63%/year in the VKA and DOAC group, respectively, log-rank test P=.009). At multivariable Cox proportional hazard regression analysis, use of DOACs was associated with a lower risk of MACE (hazard ratio, 0.636; 95% CI, 0.417 to 0.970; P=.036) and MI (hazard ratio, 0.497; 95% CI, 0.276 to 0.896; p=.020). Sensitivity analysis showed that this association was consistent in younger patients (<75 years), in patients with anemia, and in those without chronic obstructive pulmonary disease and heart failure. We also found that both dabigatran and apixaban/rivaroxaban were associated with a lower rate of MACE, with similar efficacy between full and low doses. CONCLUSION DOACs are associated with a lower risk of MACE in patients with AF independently from dosage.
Collapse
Affiliation(s)
- Daniele Pastori
- I Clinica Medica, Atherothrombosis Center, Department of Clinical, Internal, Anesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Danilo Menichelli
- I Clinica Medica, Atherothrombosis Center, Department of Clinical, Internal, Anesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Francesco Del Sole
- I Clinica Medica, Atherothrombosis Center, Department of Clinical, Internal, Anesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Pasquale Pignatelli
- I Clinica Medica, Atherothrombosis Center, Department of Clinical, Internal, Anesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy; Mediterranea Cardiocentro, Naples, Italy.
| | - Francesco Violi
- I Clinica Medica, Atherothrombosis Center, Department of Clinical, Internal, Anesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy; Mediterranea Cardiocentro, Naples, Italy
| | | |
Collapse
|
17
|
Sang Y, Roest M, de Laat B, de Groot PG, Huskens D. Interplay between platelets and coagulation. Blood Rev 2021; 46:100733. [PMID: 32682574 PMCID: PMC7354275 DOI: 10.1016/j.blre.2020.100733] [Citation(s) in RCA: 203] [Impact Index Per Article: 50.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 06/12/2020] [Accepted: 07/06/2020] [Indexed: 12/15/2022]
Abstract
Haemostasis stops bleeding at the site of vascular injury and maintains the integrity of blood vessels through clot formation. This regulated physiological process consists of complex interactions between endothelial cells, platelets, von Willebrand factor and coagulation factors. Haemostasis is initiated by a damaged vessel wall, followed with a rapid adhesion, activation and aggregation of platelets to the exposed subendothelial extracellular matrix. At the same time, coagulation factors aggregate on the procoagulant surface of activated platelets to consolidate the platelet plug by forming a mesh of cross-linked fibrin. Platelets and coagulation mutually influence each other and there are strong indications that, thanks to the interplay between platelets and coagulation, haemostasis is far more effective than the two processes separately. Clinically this is relevant because impaired interaction between platelets and coagulation may result in bleeding complications, while excessive platelet-coagulation interaction induces a high thrombotic risk. In this review, platelets, coagulation factors and the complex interaction between them will be discussed in detail.
Collapse
Affiliation(s)
- Yaqiu Sang
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands; Synapse Research Institute, Maastricht, the Netherlands
| | - Mark Roest
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands; Synapse Research Institute, Maastricht, the Netherlands
| | - Bas de Laat
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands; Synapse Research Institute, Maastricht, the Netherlands
| | | | - Dana Huskens
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands; Synapse Research Institute, Maastricht, the Netherlands.
| |
Collapse
|
18
|
Lei J, Zhou Y, Zhao H, Chen Y, Yan G, Wu L, Xu Y, Zhang J, Zhang X, Wang J, Li D, Li Y. Dabigatran activates inflammation resolution by promoting fibrinogen-like protein 2 shedding and RvD5 n-3 DPA production. Theranostics 2021; 11:4251-4261. [PMID: 33754059 PMCID: PMC7977467 DOI: 10.7150/thno.50182] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 01/29/2021] [Indexed: 02/06/2023] Open
Abstract
Rationale: The interaction between coagulation and inflammation resolution remains elusive. We recently highlighted a link between fibrinogen-like protein 2 (Fgl2) and a specialized pro-resolving mediator (SPM)-n-3 docosapentaenoic acid-derived resolvin D5 (RvD5n-3 DPA) in sepsis. This study aimed to investigate the functions of commonly used anticoagulants warfarin, dabigatran and heparin in regulating inflammation resolution. Methods: Peripheral blood was collected from clinical sepsis patients and healthy control for the determination of indicated indexes. Mouse sepsis models of zymosan-induced peritonitis and cecal ligation and puncture (CLP) were employed for the measurement of inflammation- and coagulation-related indexes. Western-blotting, ELISA and flow cytometry were applied to assess proteins. UPLC-MS/MS was used to evaluate lipid metabolites. Results: Here we report that the transmembrane Fgl2 (mFgl2) was positively associated with coagulation, while soluble Fgl2 (sFgl2) level correlated with the enhanced number of peripheral blood mononuclear cells in the sepsis patients. The anticoagulants dabigatran and warfarin attenuated zymosan-induced peritonitis, which was not shared by heparin, while only dabigatran significantly improved sepsis survival in the CLP sepsis mouse model. Although these anticoagulants consistently inhibited pro-inflammatory mediators including prostaglandin E2 and leukotriene B4, only dabigatran increased sFgl2 at both the initiation and resolution phases of inflammation. Mechanistically, dabigatran elicited the shedding of sFgl2 via prothrombin-related metalloproteases, thereby enhanced the subsequent biosynthesis of RvD5n-3 DPAvia STAT6-ALOX15 axis. Blocking metalloproteases or ALOX15 significantly impaired dabigatran-enhanced macrophage efferocytosis in vitro, as well as delayed the dabigatran-accelerated inflammation resolution in vivo. Conclusions: Our findings identify the dual anti-inflammatory and pro-resolving actions of dabigatran, through promoting sFgl2-triggered RvD5n-3 DPA production, which has important implications for promoting tissue homeostasis of sepsis.
Collapse
|
19
|
Wu J, Heemskerk JWM, Baaten CCFMJ. Platelet Membrane Receptor Proteolysis: Implications for Platelet Function. Front Cardiovasc Med 2021; 7:608391. [PMID: 33490118 PMCID: PMC7820117 DOI: 10.3389/fcvm.2020.608391] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 11/24/2020] [Indexed: 12/16/2022] Open
Abstract
The activities of adhesion and signaling receptors in platelets are controlled by several mechanisms. An important way of regulation is provided by proteolytic cleavage of several of these receptors, leading to either a gain or a loss of platelet function. The proteases involved are of different origins and types: (i) present as precursor in plasma, (ii) secreted into the plasma by activated platelets or other blood cells, or (iii) intracellularly activated and cleaving cytosolic receptor domains. We provide a comprehensive overview of the proteases acting on the platelet membrane. We describe how these are activated, which are their target proteins, and how their proteolytic activity modulates platelet functions. The review focuses on coagulation-related proteases, plasmin, matrix metalloproteinases, ADAM(TS) isoforms, cathepsins, caspases, and calpains. We also describe how the proteolytic activities are determined by different platelet populations in a thrombus and conversely how proteolysis contributes to the formation of such populations.
Collapse
Affiliation(s)
- Jiayu Wu
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, Netherlands
| | - Johan W. M. Heemskerk
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, Netherlands
| | - Constance C. F. M. J. Baaten
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, Netherlands
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen, Aachen, Germany
| |
Collapse
|
20
|
Polzin A, Dannenberg L, Thienel M, Orban M, Wolff G, Hohlfeld T, Zeus T, Kelm M, Petzold T. Noncanonical Effects of Oral Thrombin and Factor Xa Inhibitors in Platelet Activation and Arterial Thrombosis. Thromb Haemost 2020; 121:122-130. [PMID: 32942315 DOI: 10.1055/s-0040-1716750] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Nonvitamin K oral anticoagulants (NOACs) or direct oral anticoagulants comprise inhibitors of factor Xa (rivaroxaban, apixaban, edoxaban) or factor IIa (dabigatran). Both classes efficiently interfere with the final or penultimate step of the coagulation cascade and showed superior net clinical benefit compared with vitamin K antagonists for prevention of thromboembolic events in patients with AF and for prevention and therapy of deep vein thrombosis and pulmonary embolism. None the less, accumulating data suggested, that there may be differences regarding the frequency of atherothrombotic cardiovascular events between NOACs. Thus, the optimal individualized NOAC for each patient remains a matter of debate. Against this background, some basic and translational analyses emphasized NOAC effects that impact on platelet activity and arterial thrombus formation beyond inhibition of plasmatic coagulation. In this review, we will provide an overview of the available clinical and translational evidence for so-called noncanonical NOAC effects on platelet activation and arterial thrombosis.
Collapse
Affiliation(s)
- Amin Polzin
- Department of Cardiology, Pulmonology, and Vascular Medicine, Cardiovascular Research Institute Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Lisa Dannenberg
- Department of Cardiology, Pulmonology, and Vascular Medicine, Cardiovascular Research Institute Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Manuela Thienel
- Department of Cardiology, LMU München, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), Munich Heart Alliance, Munich, Germany
| | - Martin Orban
- Department of Cardiology, LMU München, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), Munich Heart Alliance, Munich, Germany
| | - Georg Wolff
- Department of Cardiology, Pulmonology, and Vascular Medicine, Cardiovascular Research Institute Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Thomas Hohlfeld
- Instituton of Pharmacology and Clinical Pharmacology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Tobias Zeus
- Department of Cardiology, Pulmonology, and Vascular Medicine, Cardiovascular Research Institute Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Malte Kelm
- Department of Cardiology, Pulmonology, and Vascular Medicine, Cardiovascular Research Institute Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Tobias Petzold
- Department of Cardiology, LMU München, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), Munich Heart Alliance, Munich, Germany
| |
Collapse
|
21
|
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
|
22
|
Violi F, Carnevale R, Pignatelli P, Cammisotto V. Letter by Violi et al Regarding Article, "Rivaroxaban Reduces Arterial Thrombosis by Inhibition of Fxa-Driven Platelet Activation via Protease Activated Receptor-1". Circ Res 2020; 126:e114-e115. [PMID: 32379576 DOI: 10.1161/circresaha.120.316895] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Francesco Violi
- From the Department of Clinical, Internal Medicine, Anesthesiological and Cardiovascular Sciences (F.V., P.P.), Sapienza University of Rome, Italy.,Mediterranea, Cardiocentro, Napoli, Italy (F.V., R.C., P.P.)
| | - Roberto Carnevale
- Department of Medical-Surgical Sciences and Biotechnologies (R.C.), Sapienza University of Rome, Italy.,Mediterranea, Cardiocentro, Napoli, Italy (F.V., R.C., P.P.)
| | - Pasquale Pignatelli
- From the Department of Clinical, Internal Medicine, Anesthesiological and Cardiovascular Sciences (F.V., P.P.), Sapienza University of Rome, Italy.,Mediterranea, Cardiocentro, Napoli, Italy (F.V., R.C., P.P.)
| | - Vittoria Cammisotto
- Department of General Surgery and Surgical Speciality Paride Stefanini (V.C.), Sapienza University of Rome, Italy
| |
Collapse
|
23
|
Petzold T, Dannenberg L, Thienel M, Ahlbrecht S, Mourikis P, Helten C, M'Pembele R, Achilles A, Zikeli D, Zhang Z, Lüsebrink E, Nicolai L, Saleh I, Jung C, Gerdes N, Hoffmann T, Levkau B, Hohlfeld T, Zeus T, Schulz C, Kelm M, Polzin A. Response by Petzold et al to Letter Regarding Article, "Rivaroxaban Reduces Arterial Thrombosis by Inhibition of Fxa-Driven Platelet Activation via Protease Activated Receptor-1". Circ Res 2020; 126:e116-e117. [PMID: 32379578 DOI: 10.1161/circresaha.120.316905] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Tobias Petzold
- From the Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians University Munich (T.P., M.T., Z.Z., E.L., L.N., I.S., C.S.).,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Germany (T.P., M.T., E.L., L.N., C.S.)
| | - Lisa Dannenberg
- Department of Cardiology, Pulmonology and Vascular Medicine, Cardiovascular Research Institute Düsseldorf (CARID) (L.D., S.A., P.M., C.H., R.M., A.A., D.Z., C.J., N.G., T.Z., M.K., A.P.), Medical Faculty of the Heinrich Heine University Düsseldorf, Germany
| | - Manuela Thienel
- From the Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians University Munich (T.P., M.T., Z.Z., E.L., L.N., I.S., C.S.).,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Germany (T.P., M.T., E.L., L.N., C.S.)
| | - Samantha Ahlbrecht
- Department of Cardiology, Pulmonology and Vascular Medicine, Cardiovascular Research Institute Düsseldorf (CARID) (L.D., S.A., P.M., C.H., R.M., A.A., D.Z., C.J., N.G., T.Z., M.K., A.P.), Medical Faculty of the Heinrich Heine University Düsseldorf, Germany
| | - Philipp Mourikis
- Department of Cardiology, Pulmonology and Vascular Medicine, Cardiovascular Research Institute Düsseldorf (CARID) (L.D., S.A., P.M., C.H., R.M., A.A., D.Z., C.J., N.G., T.Z., M.K., A.P.), Medical Faculty of the Heinrich Heine University Düsseldorf, Germany
| | - Carolin Helten
- Department of Cardiology, Pulmonology and Vascular Medicine, Cardiovascular Research Institute Düsseldorf (CARID) (L.D., S.A., P.M., C.H., R.M., A.A., D.Z., C.J., N.G., T.Z., M.K., A.P.), Medical Faculty of the Heinrich Heine University Düsseldorf, Germany
| | - René M'Pembele
- Department of Cardiology, Pulmonology and Vascular Medicine, Cardiovascular Research Institute Düsseldorf (CARID) (L.D., S.A., P.M., C.H., R.M., A.A., D.Z., C.J., N.G., T.Z., M.K., A.P.), Medical Faculty of the Heinrich Heine University Düsseldorf, Germany
| | - Alina Achilles
- Department of Cardiology, Pulmonology and Vascular Medicine, Cardiovascular Research Institute Düsseldorf (CARID) (L.D., S.A., P.M., C.H., R.M., A.A., D.Z., C.J., N.G., T.Z., M.K., A.P.), Medical Faculty of the Heinrich Heine University Düsseldorf, Germany
| | - Dorothee Zikeli
- Department of Cardiology, Pulmonology and Vascular Medicine, Cardiovascular Research Institute Düsseldorf (CARID) (L.D., S.A., P.M., C.H., R.M., A.A., D.Z., C.J., N.G., T.Z., M.K., A.P.), Medical Faculty of the Heinrich Heine University Düsseldorf, Germany
| | - Zhe Zhang
- From the Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians University Munich (T.P., M.T., Z.Z., E.L., L.N., I.S., C.S.)
| | - Enzo Lüsebrink
- From the Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians University Munich (T.P., M.T., Z.Z., E.L., L.N., I.S., C.S.).,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Germany (T.P., M.T., E.L., L.N., C.S.)
| | - Leo Nicolai
- From the Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians University Munich (T.P., M.T., Z.Z., E.L., L.N., I.S., C.S.).,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Germany (T.P., M.T., E.L., L.N., C.S.)
| | - Inas Saleh
- From the Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians University Munich (T.P., M.T., Z.Z., E.L., L.N., I.S., C.S.)
| | - Christian Jung
- Department of Cardiology, Pulmonology and Vascular Medicine, Cardiovascular Research Institute Düsseldorf (CARID) (L.D., S.A., P.M., C.H., R.M., A.A., D.Z., C.J., N.G., T.Z., M.K., A.P.), Medical Faculty of the Heinrich Heine University Düsseldorf, Germany
| | - Norbert Gerdes
- Department of Cardiology, Pulmonology and Vascular Medicine, Cardiovascular Research Institute Düsseldorf (CARID) (L.D., S.A., P.M., C.H., R.M., A.A., D.Z., C.J., N.G., T.Z., M.K., A.P.), Medical Faculty of the Heinrich Heine University Düsseldorf, Germany
| | - Till Hoffmann
- Institute of Transplantation Diagnostics and Cell Therapeutics, Heinrich Heine University Medical Center Düsseldorf, Germany (T. Hoffmann)
| | - Bodo Levkau
- Institute of Pathophysiology, West German Heart and Vascular Center, University Hospital Essen, University of Duisburg-Essen, Germany (B.L.)
| | - Thomas Hohlfeld
- Institute of Pharmacology and Clinical Pharmacology (T. Hohlfeld), Medical Faculty of the Heinrich Heine University Düsseldorf, Germany
| | - Tobias Zeus
- Department of Cardiology, Pulmonology and Vascular Medicine, Cardiovascular Research Institute Düsseldorf (CARID) (L.D., S.A., P.M., C.H., R.M., A.A., D.Z., C.J., N.G., T.Z., M.K., A.P.), Medical Faculty of the Heinrich Heine University Düsseldorf, Germany
| | - Christian Schulz
- From the Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians University Munich (T.P., M.T., Z.Z., E.L., L.N., I.S., C.S.).,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Germany (T.P., M.T., E.L., L.N., C.S.)
| | - Malte Kelm
- Department of Cardiology, Pulmonology and Vascular Medicine, Cardiovascular Research Institute Düsseldorf (CARID) (L.D., S.A., P.M., C.H., R.M., A.A., D.Z., C.J., N.G., T.Z., M.K., A.P.), Medical Faculty of the Heinrich Heine University Düsseldorf, Germany
| | - Amin Polzin
- Department of Cardiology, Pulmonology and Vascular Medicine, Cardiovascular Research Institute Düsseldorf (CARID) (L.D., S.A., P.M., C.H., R.M., A.A., D.Z., C.J., N.G., T.Z., M.K., A.P.), Medical Faculty of the Heinrich Heine University Düsseldorf, Germany
| |
Collapse
|
24
|
Platelet heterogeneity in activation-induced glycoprotein shedding: functional effects. Blood Adv 2019; 2:2320-2331. [PMID: 30232085 DOI: 10.1182/bloodadvances.2017011544] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 08/15/2018] [Indexed: 12/20/2022] Open
Abstract
The platelet receptors glycoprotein Ibα (GPIbα) and GPVI are known to be cleaved by members of a disintegrin and metalloprotease (ADAM) family (ADAM10 and ADAM17), but the mechanisms and consequences of this shedding are not well understood. Our results revealed that (1) glycoprotein shedding is confined to distinct platelet populations showing near-complete shedding, (2) the heterogeneity between (non)shed platelets is independent of agonist type but coincides with exposure of phosphatidylserine (PS), and (3) distinct pathways of shedding are induced by elevated Ca2+, low Ca2+ protein kinase C (PKC), or apoptotic activation. Furthermore, we found that receptor shedding reduces binding of von Willebrand factor, enhances binding of coagulation factors, and augments fibrin formation. In response to Ca2+-increasing agents, shedding of GPIbα was abolished by ADAM10/17 inhibition but not by blockage of calpain. Stimulation of PKC induced shedding of only GPIbα, which was annulled by kinase inhibition. The proapoptotic agent ABT-737 induced shedding, which was caspase dependent. In Scott syndrome platelets that are deficient in Ca2+-dependent PS exposure, shedding occurred normally, indicating that PS exposure is not a prerequisite for ADAM activity. In whole-blood thrombus formation, ADAM-dependent glycoprotein shedding enhanced thrombin generation and fibrin formation. Together, these findings indicate that 2 major activation pathways can evoke ADAM-mediated glycoprotein shedding in distinct platelet populations and that shedding modulates platelet function from less adhesive to more procoagulant.
Collapse
|
25
|
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
|
26
|
Weller CD, Gardiner EE, Arthur JF, Southey M, Andrews RK. Autologous platelet-rich plasma for healing chronic venous leg ulcers: Clinical efficacy and potential mechanisms. Int Wound J 2019; 16:788-792. [PMID: 30864220 PMCID: PMC7949463 DOI: 10.1111/iwj.13098] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 01/26/2019] [Indexed: 12/13/2022] Open
Abstract
The overall quality of evidence of autologous platelet-rich plasma (PRP) for treating chronic wounds remains low. While further well-designed clinical studies are clearly required to convincingly demonstrate the efficacy of autologous PRP in improved healing of venous leg ulcers (VLUs) and other chronic wounds, there is also an increasing need to better define the underlying mechanisms of action and whether positive outcomes can be predicted based on the analysis of PRP. This brief review will discuss the current understanding of autologous PRP in VLUs and whether molecular evaluation of PRP at the time of collection could potentially be informative to clinical outcomes. Benefits of the autologous PRP treatment strategy include that PRP is easily accessible and is relatively inexpensive and safe. Better understanding of the mechanisms involved could improve treatment, enable supplementation, and/or lead to gains in product development. Analysis of PRP could also add value to future clinical trials on efficacy and potentially personalised treatment regimens.
Collapse
Affiliation(s)
- Carolina D. Weller
- School of Nursing and MidwiferyMonash UniversityMelbourneVictoriaAustralia
| | - Elizabeth E. Gardiner
- Department of Cancer Biology and Therapeutics, John Curtin School of Medical ResearchAustralian National UniversityCanberraAustralian Capital TerritoryAustralia
| | - Jane F. Arthur
- Australian Centre for Blood DiseasesMonash UniversityMelbourneVictoriaAustralia
| | - Melissa Southey
- Precision MedicineMonash UniversityMelbourneVictoriaAustralia
- Cancer Epidemiology and Intelligence DivisionCancer Council VictoriaMelbourneVictoriaAustralia
- Department of Clinical PathologyThe University of MelbourneMelbourneVictoriaAustralia
| | - Robert K. Andrews
- Australian Centre for Blood DiseasesMonash UniversityMelbourneVictoriaAustralia
| |
Collapse
|
27
|
Cammisotto V, Carnevale R, Nocella C, Stefanini L, Bartimoccia S, Coluccia A, Silvestri R, Pignatelli P, Pastori D, Violi F. Nox2-mediated platelet activation by glycoprotein (GP) VI: Effect of rivaroxaban alone and in combination with aspirin. Biochem Pharmacol 2019; 163:111-118. [DOI: 10.1016/j.bcp.2019.02.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 02/12/2019] [Indexed: 01/04/2023]
|
28
|
Yamashita Y, Suzuki K, Mastumoto T, Ikejiri M, Ohishi K, Katayama N, Suzuki-Inoue K, Wada H. Elevated plasma levels of soluble C-type lectin-like receptor 2 (CLEC2) in patients with thrombotic microangiopathy. Thromb Res 2019; 178:54-58. [PMID: 30978634 DOI: 10.1016/j.thromres.2019.03.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 03/25/2019] [Accepted: 03/27/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND Thrombotic microangiopathy (TMA) is caused by activated platelets. The plasma C-type lectin-like receptor 2 (CLEC2) levels in 58 patients with TMA were examined and compared with those in healthy volunteers and other diseases. MATERIALS AND METHODS The plasma levels of soluble platelet surface glycoprotein VI (GPVI) and CLEC2 were measured in patients with TMA. RESULTS Plasma CLEC2 levels in patients with DIC and TMA were significantly higher (p < 0.001) than those in thrombocytopenic patients with other hematological diseases, but no significant differences in the plasma CLEC2 levels were observed among patients with thrombotic thrombocytopenic purpura, hemolytic uremic syndrome (HUS), atypical HUS and other TMA. The plasma CLEC2 levels after the remission were significantly lower than those before treatment (p < 0.001). The plasma CLEC2 levels were poorly correlated with the levels of soluble GPVI in the plasma of patients with TMA. The plasma CLEC2 levels were not significantly differ between survivor and non-survivor in TMA patients, but were significantly higher in non-survivor in overall population (p < 0.001). CONCLUSION The measurement of the plasma CLEC2 level is considered to be important for the diagnosis and evaluation of TMA.
Collapse
Affiliation(s)
- Yoshiki Yamashita
- Department of Hematology and Oncology, Mie University Hospital and Mie University Graduate School of Medicine, Tsu, Japan
| | - Kei Suzuki
- Department of Emergency Critical Care Center, Mie University Hospital and Mie University Graduate School of Medicine, Tsu, Japan
| | - Takeshi Mastumoto
- Department of Blood Transfusion and Cell Therapy, Mie University Hospital and Mie University Graduate School of Medicine, Tsu, Japan
| | - Makoto Ikejiri
- Department of Central Laboratory, Mie University Hospital and Mie University Graduate School of Medicine, Tsu, Japan
| | - Koji Ohishi
- Department of Blood Transfusion and Cell Therapy, Mie University Hospital and Mie University Graduate School of Medicine, Tsu, Japan
| | - Naoyuki Katayama
- Department of Hematology and Oncology, Mie University Hospital and Mie University Graduate School of Medicine, Tsu, Japan
| | - Katsue Suzuki-Inoue
- Department of Clinical and Laboratory Medicine, Yamanashi Medical University, Yamanashi, Japan
| | - Hideo Wada
- Deaprtment of Molecular and Laboratory Medicine, Mie University Hospital and Mie University Graduate School of Medicine, Tsu, Japan.
| |
Collapse
|
29
|
Soluble GPVI is elevated in injured patients: shedding is mediated by fibrin activation of GPVI. Blood Adv 2019; 2:240-251. [PMID: 29437639 DOI: 10.1182/bloodadvances.2017011171] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 12/13/2017] [Indexed: 12/18/2022] Open
Abstract
Soluble glycoprotein VI (sGPVI) is shed from the platelet surface and is a marker of platelet activation in thrombotic conditions. We assessed sGPVI levels together with patient and clinical parameters in acute and chronic inflammatory conditions, including patients with thermal injury and inflammatory bowel disease and patients admitted to the intensive care unit (ICU) for elective cardiac surgery, trauma, acute brain injury, or prolonged ventilation. Plasma sGPVI was measured by enzyme-linked immunosorbent assay and was elevated on day 14 after thermal injury, and was higher in patients who developed sepsis. sGPVI levels were associated with sepsis, and the value for predicting sepsis was increased in combination with platelet count and Abbreviated Burn Severity Index. sGPVI levels positively correlated with levels of D-dimer (a fibrin degradation product) in ICU patients and patients with thermal injury. sGPVI levels in ICU patients at admission were significantly associated with 28- and 90-day mortality independent of platelet count. sGPVI levels in patients with thermal injury were associated with 28-day mortality at days 1, 14, and 21 when adjusting for platelet count. In both cohorts, sGPVI associations with mortality were stronger than D-dimer levels. Mechanistically, release of GPVI was triggered by exposure of platelets to polymerized fibrin, but not by engagement of G protein-coupled receptors by thrombin, adenosine 5'-diphosphate, or thromboxane mimetics. Enhanced fibrin production in these patients may therefore contribute to the observed elevated sGPVI levels. sGPVI is an important platelet-specific marker for platelet activation that predicts sepsis progression and mortality in injured patients.
Collapse
|
30
|
|
31
|
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
|
32
|
Affiliation(s)
- Gary E Raskob
- University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | | | | |
Collapse
|
33
|
Swieringa F, Spronk HM, Heemskerk JW, van der Meijden PE. Integrating platelet and coagulation activation in fibrin clot formation. Res Pract Thromb Haemost 2018; 2:450-460. [PMID: 30046749 PMCID: PMC6046596 DOI: 10.1002/rth2.12107] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 03/31/2018] [Indexed: 12/21/2022] Open
Abstract
Platelets interact with the coagulation system in a multitude of ways, not only during the phases of thrombus formation, but also in specific areas within a formed thrombus. This review discusses current concepts of platelet control of thrombin generation, fibrin formation and structure, and anticoagulation. Indicated are how combined signalling via the platelet receptors for collagen (glycoprotein VI) and thrombin induces the secretion of (anti)coagulation factors, as well as surface exposure of phosphatidylserine, thereby catalysing thrombin generation. This procoagulant platelet response is also facilitated by the adhesive complexes glycoprotein Ib-V-IX and integrin αIIbβ3. In the buildup of a platelet-fibrin thrombus, the extrinsic, tissue factor-driven coagulation pathway is predominant in early stages, while the intrinsic, factor XII pathway seems to promote at later time points. Already early generation of thrombin enforces platelet responses and stimulates intra-thrombus heterogeneity with patches of loosely aggregated, contracted, and phosphatidylserine-exposing platelets. Fibrin actively formed on the surface of activated platelets supports thrombus growth, but also captures thrombin. The fibrin distribution in a thrombus appears to rely on the local procoagulant trigger and the blood flow rate. Clinical studies support the importance of the platelet-coagulation interplay, by showing beneficial effects of combination therapy in the secondary prevention of cardiovascular disease.
Collapse
Affiliation(s)
- Frauke Swieringa
- Department of BiochemistryCardiovascular Research Institute Maastricht (CARIM)Maastricht UniversityMaastrichtThe Netherlands
- Leibniz Institute for Analytical SciencesISASDortmundGermany
| | - Henri M.H. Spronk
- Department of BiochemistryCardiovascular Research Institute Maastricht (CARIM)Maastricht UniversityMaastrichtThe Netherlands
| | - Johan W.M. Heemskerk
- Department of BiochemistryCardiovascular Research Institute Maastricht (CARIM)Maastricht UniversityMaastrichtThe Netherlands
| | - Paola E.J. van der Meijden
- Department of BiochemistryCardiovascular Research Institute Maastricht (CARIM)Maastricht UniversityMaastrichtThe Netherlands
| |
Collapse
|
34
|
Switching from abacavir to tenofovir disoproxil fumarate is associated with rises in soluble glycoprotein VI, suggesting changes in platelet-collagen interactions. AIDS 2018; 32:861-866. [PMID: 29438200 DOI: 10.1097/qad.0000000000001783] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVES Altered platelet function has been proposed as an underlying mechanism to explain increased risk of myocardial infarction in people living with HIV associated with use of the nucleoside reverse transcriptase inhibitor abacavir (ABC). We aimed to examine changes in platelet biomarkers in people living with HIV switching from ABC. METHODS In a prospective, 48-week substudy of virally suppressed HIV-1-positive subjects randomized to remain on ABC/lamivudine (ABC/3TC) or switch to tenofovir disoproxil fumarate/emtricitabine, we measured soluble glycoprotein VI (sGPVI), soluble P-selectin, soluble CD40 ligand and von Willebrand factor in plasma collected over time and assessed differences using mixed effect models. RESULTS Of 312 randomized participants, 310 were included in the analysis. Mean (SD) age 46.4 (9.3) years, 262 (85%) men and 201 (65%) white. At baseline, there was no significant between-group difference in sGPVI [tenofovir disoproxil fumarate/emtricitabine 3.75 (0.25) versus ABC/3TC 3.61 (0.22) ng/ml, P = 0.69]. Greater increases in sGPVI from baseline to week 48 occurred in those switched from ABC/3TC (effect size +0.57 ng/ml; 95% confidence interval, 0.2-0.94; P = 0.003). There was no significant baseline difference or change overtime in soluble P-selectin, soluble CD40 ligand or von Willebrand factor between groups. CONCLUSION The significant increases in sGPVI that occur with a switch from ABC/3TC are suggestive of changes in platelet function centred on platelet/collagen interactions and potentially represent an underlying mechanism to explain increased risk of myocardial infarction with ABC.
Collapse
|
35
|
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
|
36
|
Vögtle T, Cherpokova D, Bender M, Nieswandt B. Targeting platelet receptors in thrombotic and thrombo-inflammatory disorders. Hamostaseologie 2017; 35:235-43. [DOI: 10.5482/hamo-14-10-0049] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 01/21/2015] [Indexed: 12/20/2022] Open
Abstract
SummaryPlatelet activation at sites of vascular injury is critical for the formation of a hemostatic plug which limits excessive blood loss, but also represents a major pathomechanism of ischemic cardio- and cerebrovascular diseases. Although currently available antiplatelet therapies have proved beneficial in preventing the recurrence of vascular events, their adverse effects on primary hemostasis emphasize the necessity to identify and characterize novel pharmacological targets for platelet inhibition. Increasing experimental evidence has suggested that several major platelet surface receptors which regulate initial steps of platelet adhesion and activation may become promising new targets for anti-platelet drugs due to their involvement in thrombotic and thrombo-inflammatory signaling cascades.This review summarizes recent developments in understanding the function of glycoprotein (GP)Ib, GPVI and the C-type lectin-like receptor 2 (CLEC-2) in hemostasis, arterial thrombosis and thrombo-inflammation and will discuss the suitability of the receptors as novel targets to treat these diseases in humans.
Collapse
|
37
|
Lee MY, Verni CC, Herbig BA, Diamond SL. Soluble fibrin causes an acquired platelet glycoprotein VI signaling defect: implications for coagulopathy. J Thromb Haemost 2017; 15:2396-2407. [PMID: 28981200 PMCID: PMC5716900 DOI: 10.1111/jth.13863] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2017] [Indexed: 11/27/2022]
Abstract
Essentials Collagen and thrombin when used simultaneously generate highly activated platelets. The effect of thrombin stimulation on subsequent glycoprotein VI (GPVI) function was observed. Soluble fibrin, but not protease activated receptor (PAR) activation, prevented GPVI activation. Circulating soluble fibrin in coagulopathic blood may cause an acquired GPVI signaling defect. SUMMARY Background In coagulopathic blood, circulating thrombin may drive platelet dysfunction. Methods/Results Using calcium dye-loaded platelets, the effect of thrombin exposure and soluble fibrin generation on subsequent platelet GPVI function was investigated. Exposure of apixaban-treated platelet-rich plasma (12% PRP) to thrombin (1-10 nm), but not ADP or thromboxane mimetic U46619 exposure, dramatically blocked subsequent GPVI activation by convulxin, collagen-related peptide or fibrillar collagen. Consistent with soluble fibrin multimerizing and binding GPVI, the onset of convulxin insensitivity required 200-500 s of thrombin exposure, was not mimicked by exposure to PAR-1/4 activating peptides, was not observed with washed platelets, and was blocked by fibrin polymerization inhibitor (GPRP) or factor XIIIa inhibitor (T101). PAR-1 signaling through Gαq was not required because vorapaxar blocked thrombin-induced calcium mobilization but had no effect on the ability of thrombin to impair GPVI-signaling. Convulxin insensitivity was unaffected by the metalloprotease inhibitor GM6001 or the αIIb β3 antagonist GR144053, indicating negligible roles for GPVI shedding or αIIb β3 binding of fibrin. Thrombin treatment of washed platelets resuspended in purified fibrinogen also produced convulxin insensitivity that was prevented by GPRP. Exposure of apixaban/PPACK-treated whole blood to thrombin-treated fibrinogen resulted in > 50% decrease in platelet deposition in a collagen microfluidic assay that required soluble fibrin assembly. Conclusions Conversion of only 1% plasma fibrinogen in coagulopathic blood would generate 90 nm soluble fibrin, far exceeding ~1 nmGPVI in blood. Soluble fibrin, rather than thrombin-induced platelet activation throuh PAR-1 and PAR-4, downregulated GPVI-signaling in response to stimuli, and may lead to subsequent hypofunction of endogenous or transfused platelets.
Collapse
Affiliation(s)
- Mei Yan Lee
- Department of Chemical and Biomolecular Engineering, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Christopher C. Verni
- Department of Chemical and Biomolecular Engineering, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Bradley A. Herbig
- Department of Chemical and Biomolecular Engineering, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Scott L. Diamond
- Department of Chemical and Biomolecular Engineering, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
38
|
Arthur JF, Gardiner EE, Andrews RK, Al-Tamimi M. Focusing on plasma glycoprotein VI. Thromb Haemost 2017; 107:648-55. [DOI: 10.1160/th11-10-0745] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2011] [Accepted: 12/10/2011] [Indexed: 12/18/2022]
Abstract
SummaryNew methods for analysing both platelet and plasma forms of the platelet-specific collagen receptor, glycoprotein VI (GPVI) in experimental models or human clinical samples, and the development of the first therapeutic compounds based on dimeric soluble GPVI-Fc or anti-GPVI antibody-based constructs, coincide with increased understanding of the potential pathophysiological role of GPVI ligand binding and shedding. Platelet GPVI not only mediates platelet activation at the site of vascular injury where collagen is exposed, but is also implicated in the pathogenesis of other diseases, such as atherosclerosis and coagulopathy, rheumatoid arthritis and tumour metastasis. Here, we describe some of the critical mechanisms for generating soluble GPVI from platelets, and future avenues for exploiting this unique platelet-specific receptor for diagnosis and/or disease prevention.
Collapse
|
39
|
Matsui T, Usui M, Wada H, Iizawa Y, Kato H, Tanemura A, Murata Y, Kuriyama N, Kishiwada M, Mizuno S, Sakurai H, Isaji S. Platelet Activation Assessed by Glycoprotein VI/Platelet Ratio Is Associated With Portal Vein Thrombosis After Hepatectomy and Splenectomy in Patients With Liver Cirrhosis. Clin Appl Thromb Hemost 2017; 24:254-262. [PMID: 29050501 DOI: 10.1177/1076029617725600] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Portal vein thrombosis (PVT) is a serious complication after hepatobiliary-pancreatic surgery. Portal vein thrombosis often develops in patients with liver cirrhosis (LC) postoperatively, although they have low platelet counts. Platelet activation is one of the causes of thrombosis formation, and soluble form of glycoprotein VI (sGPVI) has received attention as a platelet activation marker. We had prospectively enrolled the 81 consecutive patients who underwent splenectomy (Sx) and/or hepatectomy: these patients were divided as Sx (n = 38) and hepatectomy (Hx, n = 46) groups. The 3 patients who underwent both procedures were added to both groups. Each group was subdivided into patients with non-LC and LC: non-LC-Sx (n = 22) and LC-Sx (n = 16), non-LC-Hx (n = 40) and LC-Hx (n = 6). The presence of PVT was diagnosed by using enhanced computed tomography (CT) scan. Platelet counts were significantly lower in LC-Sx than in non-LC-Sx, and incidence of PVT was significantly higher in LC-Sx than in non-LC-Sx (68.8% vs 31.8%, P = .024). Soluble form of glycoprotein VI /platelet ratios on preoperative day and postoperative day 1 were significantly higher in LC-Sx than in non-LC-Sx. Incidence of PVT was significantly higher in LC-Hx than in non-LC-Hx (50.0% vs 7.5%, P < .01). Soluble form of glycoprotein VI /platelet ratios were significantly higher in LC-Hx before and after Hx, compared to non-LC-Hx. Patients with LC stay in hypercoagulable state together with platelet activation before and after surgery. Under this circumstance, alteration of portal venous blood flow after Sx or Hx is likely to cause PVT in patients with LC.
Collapse
Affiliation(s)
- Toshiki Matsui
- 1 Department of Hepatobiliary Pancreatic and Transplant Surgery, Mie University Graduate School of Medicine, Tsu, Japan
| | - Masanobu Usui
- 1 Department of Hepatobiliary Pancreatic and Transplant Surgery, Mie University Graduate School of Medicine, Tsu, Japan
| | - Hideo Wada
- 2 Molecular and Laboratory Medicine, Mie University Graduate School of Medicine, Tsu, Japan
| | - Yusuke Iizawa
- 1 Department of Hepatobiliary Pancreatic and Transplant Surgery, Mie University Graduate School of Medicine, Tsu, Japan
| | - Hiroyuki Kato
- 1 Department of Hepatobiliary Pancreatic and Transplant Surgery, Mie University Graduate School of Medicine, Tsu, Japan
| | - Akihiro Tanemura
- 1 Department of Hepatobiliary Pancreatic and Transplant Surgery, Mie University Graduate School of Medicine, Tsu, Japan
| | - Yasuhiro Murata
- 1 Department of Hepatobiliary Pancreatic and Transplant Surgery, Mie University Graduate School of Medicine, Tsu, Japan
| | - Naohisa Kuriyama
- 1 Department of Hepatobiliary Pancreatic and Transplant Surgery, Mie University Graduate School of Medicine, Tsu, Japan
| | - Masashi Kishiwada
- 1 Department of Hepatobiliary Pancreatic and Transplant Surgery, Mie University Graduate School of Medicine, Tsu, Japan
| | - Shugo Mizuno
- 1 Department of Hepatobiliary Pancreatic and Transplant Surgery, Mie University Graduate School of Medicine, Tsu, Japan
| | - Hiroyuki Sakurai
- 1 Department of Hepatobiliary Pancreatic and Transplant Surgery, Mie University Graduate School of Medicine, Tsu, Japan
| | - Shuji Isaji
- 1 Department of Hepatobiliary Pancreatic and Transplant Surgery, Mie University Graduate School of Medicine, Tsu, Japan
| |
Collapse
|
40
|
Layios N, Delierneux C, Hego A, Huart J, Gosset C, Lecut C, Maes N, Geurts P, Joly A, Lancellotti P, Albert A, Damas P, Gothot A, Oury C. Sepsis prediction in critically ill patients by platelet activation markers on ICU admission: a prospective pilot study. Intensive Care Med Exp 2017; 5:32. [PMID: 28699088 PMCID: PMC5505890 DOI: 10.1186/s40635-017-0145-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 07/03/2017] [Indexed: 01/17/2023] Open
Abstract
Background Platelets have been involved in both immune surveillance and host defense against severe infection. To date, whether platelet phenotype or other hemostasis components could be associated with predisposition to sepsis in critical illness remains unknown. The aim of this work was to identify platelet markers that could predict sepsis occurrence in critically ill injured patients. Methods This single-center, prospective, observational, 7-month study was based on a cohort of 99 non-infected adult patients admitted to ICUs for elective cardiac surgery, trauma, acute brain injury, and post-operative prolonged ventilation and followed up during ICU stay. Clinical characteristics and severity score (SOFA) were recorded on admission. Platelet activation markers, including fibrinogen binding to platelets, platelet membrane P-selectin expression, plasma soluble CD40L, and platelet-leukocytes aggregates were assayed by flow cytometry at admission and 48 h later, and then at the time of sepsis diagnosis (Sepsis-3 criteria) and 7 days later for sepsis patients. Hospitalization data and outcomes were also recorded. Methods Of the 99 patients, 19 developed sepsis after a median time of 5 days. These patients had a higher SOFA score at admission; levels of fibrinogen binding to platelets (platelet-Fg) and of D-dimers were also significantly increased compared to the other patients. Levels 48 h after ICU admission no longer differed between the two patient groups. Platelet-Fg % was an independent predictor of sepsis (P = 0.0031). By ROC curve analysis, cutoff point for Platelet-Fg (AUC = 0.75) was 50%. In patients with a SOFA cutoff of 8, the risk of sepsis reached 87% when Platelet-Fg levels were above 50%. Patients with sepsis had longer ICU and hospital stays and higher death rate. Conclusions Platelet-bound fibrinogen levels assayed by flow cytometry within 24 h of ICU admission help identifying critically ill patients at risk of developing sepsis. Electronic supplementary material The online version of this article (doi:10.1186/s40635-017-0145-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Nathalie Layios
- Department of General Intensive Care, University Hospital of Liège, Liège, Belgium.,Laboratory of Thrombosis and Hemostasis, GIGA-Cardiovascular Sciences, University of Liège, Department of Cardiology, University Hospital of Liège, Liège, Belgium
| | - Céline Delierneux
- Laboratory of Thrombosis and Hemostasis, GIGA-Cardiovascular Sciences, University of Liège, Department of Cardiology, University Hospital of Liège, Liège, Belgium
| | - Alexandre Hego
- Laboratory of Thrombosis and Hemostasis, GIGA-Cardiovascular Sciences, University of Liège, Department of Cardiology, University Hospital of Liège, Liège, Belgium
| | - Justine Huart
- Laboratory of Thrombosis and Hemostasis, GIGA-Cardiovascular Sciences, University of Liège, Department of Cardiology, University Hospital of Liège, Liège, Belgium
| | - Christian Gosset
- Laboratory of Hematology, University Hospital of Liège, Liège, Belgium
| | - Christelle Lecut
- Laboratory of Hematology, University Hospital of Liège, Liège, Belgium
| | - Nathalie Maes
- Department of Biostatistics and Medico-Economic Information, University Hospital of Liège, Liège, Belgium
| | - Pierre Geurts
- Systems and Modeling, Department of Electrical Engineering and Computer Science, University of Liège, Liège, Belgium
| | - Arnaud Joly
- Systems and Modeling, Department of Electrical Engineering and Computer Science, University of Liège, Liège, Belgium
| | - Patrizio Lancellotti
- Laboratory of Thrombosis and Hemostasis, GIGA-Cardiovascular Sciences, University of Liège, Department of Cardiology, University Hospital of Liège, Liège, Belgium.,Gruppo Villa Maria Care and Research, Anthea Hospital, Bari, Italy
| | - Adelin Albert
- Department of Biostatistics and Medico-Economic Information, University Hospital of Liège, Liège, Belgium
| | - Pierre Damas
- Department of General Intensive Care, University Hospital of Liège, Liège, Belgium
| | - André Gothot
- Laboratory of Hematology, University Hospital of Liège, Liège, Belgium
| | - Cécile Oury
- Laboratory of Thrombosis and Hemostasis, GIGA-Cardiovascular Sciences, University of Liège, Department of Cardiology, University Hospital of Liège, Liège, Belgium.
| |
Collapse
|
41
|
Egan K, Dillon A, Dunne E, Kevane B, Galvin Z, Maguire P, Kenny D, Stewart S, Ainle FN. Increased soluble GPVI levels in cirrhosis: evidence for early in vivo platelet activation. J Thromb Thrombolysis 2017; 43:54-59. [PMID: 27416950 DOI: 10.1007/s11239-016-1401-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Cirrhosis is a consequence of prolonged liver injury and is characterised by extensive tissue fibrosis: the deposition of collagen-rich extracellular matrix. The haemostatic balance is disordered in cirrhosis and coagulation activation appears to promote fibrosis. In spite of recent studies demonstrating a role for anticoagulant therapy in preventing cirrhosis progression, there has not been a change in clinical practice, suggesting that physicians are reluctant to anticoagulate patients with cirrhosis due to bleeding risks. Platelets play an important role in facilitating coagulation. Glycoprotein VI (GPVI) is a platelet-specific collagen receptor that is shed from the platelet surface in a metalloproteinase-dependent manner in response to GPVI ligation and coagulation activation. Our aim was to use soluble GPVI levels to determine whether there was evidence for collagen and coagulation-induced platelet activation in early, well-compensated cirrhosis. Plasma soluble GPVI levels were quantified in 46 patients with mixed aetiology cirrhosis and 55 healthy controls using an immunoassay. In the cirrhosis group, soluble GPVI levels were significantly increased (5.8 ± 4.4 ng/ml, n = 46) compared to healthy controls (3.3 ± 3.4 ng/ml, n = 55, p < 0.05). This increase in soluble GPVI levels was still evident when levels were adjusted for platelet count (Healthy controls; 0.015 ± 0.018 ng/106 platelets/ml vs. cirrhosis; 0.048 ± 0.04 ng/106 platelets/ml, p < 0.0001). This study provides evidence for early platelet activation in patients with well-compensated cirrhosis. This may have translational implications for prognosis, treatment, and risk stratification.
Collapse
Affiliation(s)
- Karl Egan
- School of Medicine and Medical Sciences, University College Dublin, Dublin 4, Ireland.,SPHERE Research Group, Conway Institute, University College Dublin, Dublin 4, Ireland
| | - Audrey Dillon
- Department of Hepatology, Mater Misericordiae University Hospital, Dublin 7, Ireland
| | - Eimear Dunne
- Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Barry Kevane
- School of Medicine and Medical Sciences, University College Dublin, Dublin 4, Ireland.,SPHERE Research Group, Conway Institute, University College Dublin, Dublin 4, Ireland.,Department of Haematology, Mater Misericordiae University Hospital, Dublin 7, Ireland
| | - Zita Galvin
- Department of Hepatology, Mater Misericordiae University Hospital, Dublin 7, Ireland
| | - Patricia Maguire
- SPHERE Research Group, Conway Institute, University College Dublin, Dublin 4, Ireland
| | - Dermot Kenny
- Department of Hepatology, Mater Misericordiae University Hospital, Dublin 7, Ireland
| | - Stephen Stewart
- Department of Hepatology, Mater Misericordiae University Hospital, Dublin 7, Ireland
| | - Fionnuala Ni Ainle
- School of Medicine and Medical Sciences, University College Dublin, Dublin 4, Ireland. .,SPHERE Research Group, Conway Institute, University College Dublin, Dublin 4, Ireland. .,Department of Haematology, Mater Misericordiae University Hospital, Dublin 7, Ireland.
| |
Collapse
|
42
|
Lukito P, Wong A, Jing J, Arthur JF, Marasco SF, Murphy DA, Bergin PJ, Shaw JA, Collecutt M, Andrews RK, Gardiner EE, Davis AK. Mechanical circulatory support is associated with loss of platelet receptors glycoprotein Ibα and glycoprotein VI. J Thromb Haemost 2016; 14:2253-2260. [PMID: 27601054 DOI: 10.1111/jth.13497] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 08/22/2016] [Indexed: 11/25/2022]
Abstract
Essentials Relationship of acquired von Willebrand disease (VWD) and platelet dysfunction is explored. Patients with ventricular assist devices and on extracorporeal membrane oxygenation are investigated. Acquired VWD and platelet receptor shedding is demonstrated in the majority of patients. Loss of platelet adhesion receptors glycoprotein (GP) Ibα and GPVI may increase bleeding risk. SUMMARY Background Ventricular assist devices (VADs) and extracorporeal membrane oxygenation (ECMO) are associated with bleeding that is not fully explained by anticoagulant or antiplatelet use. Exposure of platelets to elevated shear in vitro leads to increased shedding. Objectives To investigate whether loss of platelet receptors occurs in vivo, and the relationship with acquired von Willebrand syndrome (AVWS). Methods Platelet counts, coagulation tests and von Willebrand factor (VWF) analyses were performed on samples from 21 continuous flow VAD (CF-VAD), 20 ECMO, 12 heart failure and seven aortic stenosis patients. Levels of platelet receptors were measured by flow cytometry or ELISA. Results The loss of high molecular weight VWF multimers was observed in 18 of 19 CF-VAD and 14 of 20 ECMO patients, consistent with AVWS. Platelet receptor shedding was demonstrated by elevated soluble glycoprotein (GP) VI levels in plasma and significantly reduced surface GPIbα and GPVI levels in CF-VAD and ECMO patients as compared with healthy donors. Platelet receptor levels were also significantly reduced in heart failure patients. Conclusions These data link AVWS and increased platelet receptor shedding in patients with CF-VADs or ECMO for the first time. Loss of the platelet surface receptors GPIbα and GPVI in heart failure, CF-VAD and ECMO patients may contribute to ablated platelet adhesion/activation, and limit thrombus formation under high/pathologic shear conditions.
Collapse
Affiliation(s)
- P Lukito
- Haematology Unit, Alfred Hospital, Melbourne, Australia
| | - A Wong
- Haematology Unit, Alfred Hospital, Melbourne, Australia
| | - J Jing
- Australian Centre for Blood Diseases, Monash University, Melbourne, Australia
| | - J F Arthur
- Australian Centre for Blood Diseases, Monash University, Melbourne, Australia
| | - S F Marasco
- Cardiothoracic Unit, Alfred Hospital, Melbourne, Australia
| | - D A Murphy
- Intensive Care Unit, Alfred Hospital, Melbourne, Australia
| | - P J Bergin
- Cardiology Unit, Alfred Hospital, Melbourne, Australia
| | - J A Shaw
- Cardiology Unit, Alfred Hospital, Melbourne, Australia
| | - M Collecutt
- Haematology Laboratory, Alfred Hospital, Melbourne, Australia
| | - R K Andrews
- Australian Centre for Blood Diseases, Monash University, Melbourne, Australia
| | - E E Gardiner
- Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - A K Davis
- Haematology Unit, Alfred Hospital, Melbourne, Australia
| |
Collapse
|
43
|
Abstract
Proteolytic shedding of the extracellular ectodomain of platelet receptors provides a key mechanism for irreversible loss of ligand-binding capacity, and for regulating platelet function in health and disease. Platelets derived from megakaryocytes are small anucleate cells in peripheral blood, with the ability to rapidly adhere, become activated, and secrete an array of procoagulant and proinflammatory factors at sites of vascular injury or disease, and to form a platelet aggregate (thrombus) which is not only critical in normal hemostasis and wound healing, but in atherothrombotic diseases including myocardial infarction and ischemic stroke. Basic mechanisms of receptor shedding on platelets have important distinctions from how receptors on other cell types might be shed, in that shedding is rapidly initiated (within seconds to minutes) and occurs under altered shear conditions encountered in flowing blood or experimentally ex vivo. This review will consider the key components of platelet receptor shedding, that is, the receptor with relevant cleavage site, the (metallo)proteinase or sheddase and how its activity is regulated, and the range of known regulatory factors that control platelet receptor shedding including receptor-associated molecules such as calmodulin, factors controlling sheddase surface expression and activity, and other elements such as shear stress, plasma membrane properties, cellular activation status or age. Understanding these basic mechanisms of platelet receptor shedding is significant in terms of utilizing receptor surface expression or soluble proteolytic fragments as platelet-specific biomarkers and/or ultimately therapeutic targeting of these mechanisms to control platelet reactivity and function.
Collapse
Affiliation(s)
- Robert K Andrews
- a Australian Centre for Blood Diseases , Monash University , Melbourne , Australia 3004.,b Department of Cancer Biology and Therapeutics, the John Curtin School of Medical Research , Australian National University , Canberra , Australia 2600
| | - Elizabeth E Gardiner
- a Australian Centre for Blood Diseases , Monash University , Melbourne , Australia 3004.,b Department of Cancer Biology and Therapeutics, the John Curtin School of Medical Research , Australian National University , Canberra , Australia 2600
| |
Collapse
|
44
|
Chatterjee M, Gawaz M. Clinical significance of receptor shedding-platelet GPVI as an emerging diagnostic and therapeutic tool. Platelets 2016; 28:362-371. [PMID: 27753514 DOI: 10.1080/09537104.2016.1227062] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Platelet membrane bedecked with a wide array of receptors offers a platform to regulate platelet responsiveness, thrombotic propensity, inflammatory disposition, and immune reactivity under diverse pathophysiological conditions. Ectopic proteolytic cleavage of such receptors irreversibly inactivates receptor-mediated intracellular signaling governing cellular functions, further releases soluble fragments into circulation which might modulate functions of target cells. Glycoprotein VI-(GPVI) is a membrane glycoprotein expressed in platelets and megakaryocytes. Platelet GPVI surface expression is enhanced following acute ischemic events like myocardial infarction and cerebral stroke, serves as an imminent diagnostic tool independent of markers of tissue necrosis, and is associated with poor prognosis. Platelets undergo GPVI shedding and thereby contribute to soluble plasma levels of sGPVI, with distinct diagnostic and prognostic attributes. This review summarizes the functional significance and mechanistic basis whereby GPVI surface availability is up- or downregulated on platelets and the impact of GPVI in diagnostic, prognostic, and therapeutic strategies in diseases where platelets play a regulatory role. Further, we also highlight how novel non-invasive platelet-based diagnostic and therapeutic strategies have evolved utilizing GPVI for lesion-directed antithrombotic therapy or to counteract atherosclerotic disposition to ameliorate care of patients particularly in the context of cardio-cerebro-vascular medicine.
Collapse
Affiliation(s)
- Madhumita Chatterjee
- a Innere Medizin III, Kardiologie und Kreislauferkrankungen , Eberhard Karls Universität , Tübingen , Germany
| | - Meinrad Gawaz
- a Innere Medizin III, Kardiologie und Kreislauferkrankungen , Eberhard Karls Universität , Tübingen , Germany
| |
Collapse
|
45
|
Pignatelli P, Pastori D, Bartimoccia S, Menichelli D, Vicario T, Nocella C, Carnevale R, Violi F. Anti Xa oral anticoagulants inhibit in vivo platelet activation by modulating glycoprotein VI shedding. Pharmacol Res 2016; 113:484-489. [PMID: 27693274 DOI: 10.1016/j.phrs.2016.09.035] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 09/25/2016] [Accepted: 09/26/2016] [Indexed: 01/26/2023]
Abstract
Anti Xa non-vitamin K oral anticoagulants (anti Xa NOACs) seem to possess antiplatelet effect in vitro, but it is unclear if this occurs also in vivo. Aim of the study was to compare the effect on platelet activation of two anti Xa NOACs, namely apixaban and rivaroxaban, to warfarin, and to investigate the potential underlying mechanism by evaluating soluble glycoprotein GPVI (sGPVI), a protein involved in platelet activation. We performed a cross-sectional including AF patients treated with warfarin (n=30), or apixaban 10mg/day (n=40), or rivaroxaban 20mg/day (n=40). Patients were balanced for sex, age and cardiovascular risk factors. Platelet activation by urinary excretion of 11-dehydro-thromboxane (Tx) B2 and soluble GPVI (sGPVI) were analysed at baseline and after 3 months of treatment. Baseline TxB2 value was 155.2±42.7ng/mg creatinine. The 3 months-variation of urinary excretion of TxB2 was -6.5% with warfarin (p=0.197), -29% with apixaban (p<0.001) and -31% with rivaroxaban (p<0.001). Use of anti Xa NOACs was independently associated to the variation of urinary TxB2 (B: -0.469, p<0.001), after adjustment for clinical characteristics; sGPVI was significantly lower in patients treated with NOACs at 3 months (p<0.001), while only a trend for the warfarin group (p=0.116) was observed. The variation of sGPVI was correlated with that of TxB2 in the NOACs group (Rs: 0.527, p<0.001). In 15 patients (5 per each group) platelet recruitment was significantly lowered at 3 months by NOACs (p<0.001), but not by warfarin. The study provides evidence that anti Xa NOACs significantly inhibit urinary TxB2 excretion compared to warfarin, suggesting that NOACs possess antiplatelet property.
Collapse
Affiliation(s)
- Pasquale Pignatelli
- IClinica Medica, Atherothrombosis Center, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Italy.
| | - Daniele Pastori
- IClinica Medica, Atherothrombosis Center, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Italy
| | - Simona Bartimoccia
- IClinica Medica, Atherothrombosis Center, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Italy
| | - Danilo Menichelli
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Tommasa Vicario
- IClinica Medica, Atherothrombosis Center, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Italy
| | - Cristina Nocella
- IClinica Medica, Atherothrombosis Center, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Italy
| | - Roberto Carnevale
- IClinica Medica, Atherothrombosis Center, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Italy; Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Francesco Violi
- IClinica Medica, Atherothrombosis Center, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Italy
| |
Collapse
|
46
|
Au AE, Josefsson EC. Regulation of platelet membrane protein shedding in health and disease. Platelets 2016; 28:342-353. [PMID: 27494300 DOI: 10.1080/09537104.2016.1203401] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Extracellular proteolysis of platelet plasma membrane proteins is an event that ensues platelet activation. Shedding of surface receptors such as glycoprotein (GP) Ibα, GPV and GPVI as well as externalized proteins P-selectin and CD40L releases soluble ectodomain fragments that are subsequently detectable in plasma. This results in the irreversible functional downregulation of platelet receptor-mediated adhesive interactions and the generation of biologically active fragments. In this review, we describe molecular insights into the regulation of platelet receptor and ligand shedding in health and disease. The scope of this review is specially focused on GPIbα, GPV, GPVI, P-selectin and CD40L where we: (1) describe the basic physiological regulation of expression and shedding of these proteins in hemostasis illustrate alterations in receptor expression during (2) apoptosis and (3) ex vivo storage relevant for blood banking purposes; (4) discuss considerations to be made when analyzing and interpreting shedding of platelet membrane proteins and finally; (5) collate clinical evidence that quantify these platelet proteins during disease.
Collapse
Affiliation(s)
- Amanda E Au
- a The Walter and Eliza Hall Institute of Medical Research, Cancer & Haematology Division , 1G Royal Parade, Melbourne , Australia.,b Department of Medical Biology , The University of Melbourne , Melbourne , Australia
| | - Emma C Josefsson
- a The Walter and Eliza Hall Institute of Medical Research, Cancer & Haematology Division , 1G Royal Parade, Melbourne , Australia.,b Department of Medical Biology , The University of Melbourne , Melbourne , Australia
| |
Collapse
|
47
|
Takahashi N, Usui M, Naitoh K, Wada H, Mastsui T, Kobayashi T, Matsumoto T, Uemoto S, Isaji S. Elevated Soluble Platelet Glycoprotein VI Levels in Patients After Living Donor Liver Transplantation. Clin Appl Thromb Hemost 2016; 23:274-281. [PMID: 26346441 DOI: 10.1177/1076029615604047] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Plasma-soluble platelet glycoprotein VI (sGPVI) levels were examined in patients undergoing living donor liver transplantation (LDLT), and the relationship between platelet activation and thrombocytopenia was evaluated to understand the mechanism of thrombocytopenia in LDLT. Platelet counts were significantly higher in the donors compared to the recipient, and the plasma sGPVI levels increased in both groups after the operation. Regarding the relationship between the platelet counts and the sGPVI levels, the slope varied on different days, and it became negative on day 3, suggesting that the plasma sGPVI levels are related to platelet activation in LDLT. The frequency of complications was high in the nonsurvivors. The platelet counts were higher in the survivors than in the nonsurvivors on days 14 and 28. Although the plasma levels of sGPVI in the survivors increased after the operation, those in the nonsurvivors were high only on day 3. Although the ADAMTS13 (a disintegrin and metalloproteinase with a thrombospondin type 1 domain, member 13) levels were markedly reduced, von Willebrand factor (VWF) and VWF propeptide (VWFpp) were markedly elevated during LDLT. The antithrombin activity was significantly lower (day 14) and VWFpp (day 28) was significantly higher in the nonsurvivors than in the survivors. These findings suggest that platelet activation first occurs after LDLT, and it is high in the nonsurvivors on day 3. Thereafter, the hemostatic abnormality and vascular endothelial cell injuries may appear on days 14 and 28.
Collapse
Affiliation(s)
- Naoki Takahashi
- 1 Hepatobiliary Pancreatic and Transplant Surgery, Mie University Graduate School of Medicine, Mie, Japan
| | - Masanobu Usui
- 1 Hepatobiliary Pancreatic and Transplant Surgery, Mie University Graduate School of Medicine, Mie, Japan
| | - Katsuki Naitoh
- 2 Biology Laboratory, Discovery Research, Mochida Pharmaceutical Co, Ltd, Shizuoka, Japan
| | - Hideo Wada
- 3 Department of Molecular and Laboratory Medicine, Mie University Graduate School of Medicine, Mie, Japan
| | - Toshiki Mastsui
- 1 Hepatobiliary Pancreatic and Transplant Surgery, Mie University Graduate School of Medicine, Mie, Japan
| | - Toshihiko Kobayashi
- 3 Department of Molecular and Laboratory Medicine, Mie University Graduate School of Medicine, Mie, Japan
| | - Takeshi Matsumoto
- 4 Department of Blood Transfusion, Mie University Graduate School of Medicine, Mie, Japan
| | - Shinji Uemoto
- 5 Hepatobiliary Pancreatic and Transplantation Surgery, Kyoto University Graduate School, Kyoto, Japan
| | - Shuji Isaji
- 1 Hepatobiliary Pancreatic and Transplant Surgery, Mie University Graduate School of Medicine, Mie, Japan
| |
Collapse
|
48
|
Affiliation(s)
- Markus Bender
- Department of Experimental Biomedicine, University of Würzburg, University Hospital and Rudolf Virchow Center, Würzburg, Germany
| | - David Stegner
- Department of Experimental Biomedicine, University of Würzburg, University Hospital and Rudolf Virchow Center, Würzburg, Germany
| | - Bernhard Nieswandt
- Department of Experimental Biomedicine, University of Würzburg, University Hospital and Rudolf Virchow Center, Würzburg, Germany
| |
Collapse
|
49
|
Facey A, Pinar I, Arthur JF, Qiao J, Jing J, Mado B, Carberry J, Andrews RK, Gardiner EE. A-Disintegrin-And-Metalloproteinase (ADAM) 10 Activity on Resting and Activated Platelets. Biochemistry 2016; 55:1187-94. [DOI: 10.1021/acs.biochem.5b01102] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Adam Facey
- Australian
Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia 3004
| | - Isaac Pinar
- Department
of Mechanical and Aerospace Engineering, Monash University, Clayton, Victoria, Australia 3168
| | - Jane F. Arthur
- Australian
Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia 3004
| | - Jianlin Qiao
- Australian
Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia 3004
| | - Jing Jing
- Australian
Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia 3004
| | - Belden Mado
- Australian
Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia 3004
| | - Josie Carberry
- Department
of Mechanical and Aerospace Engineering, Monash University, Clayton, Victoria, Australia 3168
| | - Robert K. Andrews
- Australian
Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia 3004
| | - Elizabeth E. Gardiner
- Australian
Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia 3004
| |
Collapse
|
50
|
Sánchez Guiu IM, Martínez-Martinez I, Martínez C, Navarro-Fernandez J, García-Candel F, Ferrer-Marín F, Vicente V, Watson SP, Andrews RK, Gardiner EE, Lozano ML, Rivera J. An atypical IgM class platelet cold agglutinin induces GPVI-dependent aggregation of human platelets. Thromb Haemost 2015; 114:313-24. [PMID: 25994029 DOI: 10.1160/th14-11-0945] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 03/15/2015] [Indexed: 12/12/2022]
Abstract
Platelet cold agglutinins (PCA) cause pseudothrombocytopenia, spurious thrombocytopenia due to ex vivo platelet clumping, complicating clinical diagnosis, but mechanisms and consequences of PCA are not well defined. Here, we characterised an atypical immunoglobulin (Ig)M PCA in a 37-year-old woman with lifelong bleeding and chronic moderate thrombocytopenia, that induces activation and aggregation of autologous or allogeneic platelets via interaction with platelet glycoprotein (GP)VI. Patient temperature-dependent pseudothrombocytopenia was EDTA-independent, but was prevented by integrin αIIbβ3 blockade. Unstimulated patient platelets revealed elevated levels of bound IgM, increased expression of activation markers (P-selectin and CD63), low GPVI levels and abnormally high thromboxane (TX)A2 production. Patient serum induced temperature- and αIIbβ3-dependent decrease of platelet count in allogeneic donor citrated platelet-rich plasma (PRP), but not in PRP from Glanzmann's thrombasthenia or afibrinogenaemia patients. In allogeneic platelets, patient plasma induced shape change, P-selectin and CD63 expression, (14)C-serotonin release, and TXA2 production. Activation was not inhibited by aspirin, cangrelor or blocking anti-Fc receptor (FcγRIIA) antibody, but was abrogated by inhibitors of Src and Syk, and by a soluble GPVI-Fc fusion protein. GPVI-deficient platelets were not activated by patient plasma. These data provide the first evidence for an IgM PCA causing platelet activation/aggregation via GPVI. The PCA activity persisted over a five-year follow-up period, supporting a causative role in patient chronic thrombocytopenia and bleeding.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - M L Lozano
- María Luisa Lozano, MD, PhD, Centro Regional de Hemodonación, C/ Ronda de Garay s/n, Murcia, 30003, Spain, Tel.: +34 968341990, Fax: +34 96826191, E-mail:
| | | |
Collapse
|