1
|
Li R. Another layer of complexity in ectodomain shedding of platelet glycoprotein Ibα. J Thromb Haemost 2023; 21:2051-2053. [PMID: 37468175 DOI: 10.1016/j.jtha.2023.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 04/07/2023] [Indexed: 07/21/2023]
Affiliation(s)
- Renhao Li
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA.
| |
Collapse
|
2
|
Trivigno SMG, Guidetti GF, Barbieri SS, Zarà M. Blood Platelets in Infection: The Multiple Roles of the Platelet Signalling Machinery. Int J Mol Sci 2023; 24:ijms24087462. [PMID: 37108623 PMCID: PMC10138547 DOI: 10.3390/ijms24087462] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/12/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
Platelets are classically recognized for their important role in hemostasis and thrombosis but they are also involved in many other physiological and pathophysiological processes, including infection. Platelets are among the first cells recruited to sites of inflammation and infection and they exert their antimicrobial response actively cooperating with the immune system. This review aims to summarize the current knowledge on platelet receptor interaction with different types of pathogens and the consequent modulations of innate and adaptive immune responses.
Collapse
Affiliation(s)
- Silvia M G Trivigno
- Department of Biology and Biotechnology, University of Pavia, 27100 Pavia, Italy
- University School for Advanced Studies, IUSS, 27100 Pavia, Italy
| | | | - Silvia Stella Barbieri
- Unit of Heart-Brain Axis: Cellular and Molecular Mechanisms, Centro Cardiologico Monzino IRCCS, 20138 Milano, Italy
| | - Marta Zarà
- Unit of Heart-Brain Axis: Cellular and Molecular Mechanisms, Centro Cardiologico Monzino IRCCS, 20138 Milano, Italy
| |
Collapse
|
3
|
Simeone P, Liani R, Tripaldi R, Ciotti S, Recchiuti A, Abbonante V, Porro B, Del Boccio P, Di Castelnuovo A, Lanuti P, Camera M, Pieragostino D, Lee-Sundlov M, Luongo M, Auciello R, Bologna G, Cufaro MC, Tremoli E, Hoffmeister KM, Cipollone F, Balduini A, Santilli F. Reduced platelet glycoprotein Ibα shedding accelerates thrombopoiesis and COX-1 recovery: implications for aspirin dosing regimen. Haematologica 2023; 108:1141-1157. [PMID: 36546455 PMCID: PMC10071111 DOI: 10.3324/haematol.2022.281006] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022] Open
Abstract
Cardiovascular (CV) disease prevention with low-dose aspirin can be less effective in patients with a faster recovery of platelet (PLT) cyclooxygenase (COX)-1 activity during the 24-hour dosing interval. We previously showed that incomplete suppression of TXA2 over 24 hours can be rescued by a twice daily aspirin regimen. Here we show that reduced PLT glycoprotein (GP)Ibα shedding characterizes patients with accelerated COX-1 recovery and may contribute to higher thrombopoietin (TPO) production and higher rates of newly formed PLT, escaping aspirin inhibition over 24 hours. Two hundred aspirin-treated patients with high CV risk (100 with type 2 diabetes mellitus) were stratified according to the kinetics of PLT COX-1 activity recovery during the 10- to 24-hour dosing interval. Whole proteome analysis showed that PLT from patients with accelerated COX-1 recovery were enriched in proteins involved in cell survival, inhibition of apoptosis and cellular protrusion formation. In agreement, we documented increased plasma TPO, megakaryocyte maturation and proplatelet formation, and conversely increased PLT galactose and reduced caspase 3, phosphatidylserine exposure and ADAM17 activation, translating into diminished GPIbα cleavage and glycocalicin (GC) release. Treatment of HepG2 cells with recombinant GC led to a dose-dependent reduction of TPO mRNA in the liver, suggesting that reduced GPIbα ectodomain shedding may unleash thrombopoiesis. A cluster of clinical markers, including younger age, non-alcoholic fatty liver disease, visceral obesity and higher TPO/GC ratio, predicted with significant accuracy the likelihood of faster COX-1 recovery and suboptimal aspirin response. Circulating TPO/GC ratio, reflecting a dysregulation of PLT lifespan and production, may provide a simple tool to identify patients amenable to more frequent aspirin daily dosing.
Collapse
Affiliation(s)
- Paola Simeone
- Department of Medicine and Aging Sciences, Center for Advanced Studies and Technology (CAST), University of Chieti
| | - Rossella Liani
- Department of Medicine and Aging Sciences, Center for Advanced Studies and Technology (CAST), University of Chieti
| | - Romina Tripaldi
- Department of Medicine and Aging Sciences, Center for Advanced Studies and Technology (CAST), University of Chieti
| | - Sonia Ciotti
- Department of Medicine and Aging Sciences, Center for Advanced Studies and Technology (CAST), University of Chieti
| | - Antonio Recchiuti
- Department of Medical, Oral, and Biotechnological Science, Center for Advanced Studies and Technology (CAST), Chieti
| | - Vittorio Abbonante
- Department of Molecular Medicine, University of Pavia, Pavia, Italy; Department of Health Sciences, Magna Graecia University of Catanzaro, Catanzaro
| | | | - Piero Del Boccio
- Department of Pharmacy, Center for Advanced Studies and Technology (CAST), Chieti
| | | | - Paola Lanuti
- Department of Medicine and Aging Sciences, Center for Advanced Studies and Technology (CAST), University of Chieti
| | - Marina Camera
- Centro Cardiologico Monzino IRCCS, Milan; Department of Pharmaceutical Sciences, Università degli Studi di Milano, Milan
| | - Damiana Pieragostino
- Department of Innovative Technologies in Medicine and Dentistry, Center for Advanced Studies and Technology (CAST), Chieti
| | - Melissa Lee-Sundlov
- Versiti Translational Glycomics Center and Versiti Blood Research Institute, Milwaukee, WI
| | - Myriam Luongo
- Immunotransfusion Service, Clinical Haematology of Chieti University Hospital
| | | | - Giuseppina Bologna
- Department of Medicine and Aging Sciences, Center for Advanced Studies and Technology (CAST), University of Chieti
| | - Maria Concetta Cufaro
- Department of Innovative Technologies in Medicine and Dentistry, Center for Advanced Studies and Technology (CAST), Chieti
| | | | - Karin M Hoffmeister
- Versiti Translational Glycomics Center and Versiti Blood Research Institute, Milwaukee, WI, USA; Departments of Biochemistry and Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Francesco Cipollone
- Department of Medicine and Aging Sciences, Center for Advanced Studies and Technology (CAST), University of Chieti
| | | | - Francesca Santilli
- Department of Medicine and Aging Sciences, Center for Advanced Studies and Technology (CAST), University of Chieti.
| |
Collapse
|
4
|
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
|
5
|
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
|
6
|
Chen Z, Tran D, Li T, Arias K, Griffith BP, Wu ZJ. The Role of a Disintegrin and Metalloproteinase Proteolysis and Mechanical Damage in Nonphysiological Shear Stress-Induced Platelet Receptor Shedding. ASAIO J 2020; 66:524-531. [PMID: 31192844 DOI: 10.1097/mat.0000000000001028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
In order to explore the role of a disintegrin and metalloproteinase (ADAM) proteolysis and direct mechanical damage in non-physiologic shear stress (NPSS)-caused platelet receptor shedding, the healthy donor blood treated with/without ADAM inhibitor was exposed to NPSS (150 Pa). The expression of the platelet surface receptors glycoprotein (GP) Ibα and glycoprotein (GP) VI (GPVI) in NPSS-damaged blood was quantified with flow cytometry. The impact of ADAM inhibition on adhesion of NPSS-damaged platelets on von Willibrand factor (VWF) and collagen was explored with fluorescence microscopy. The impact of ADAM inhibition on ristocetin- and collagen-caused aggregation of NPSS-damaged platelets was examined by aggregometry. The results showed that ADAM inhibition could lessen the NPSS-induced loss of platelet surface receptor GPIbα (12%) and GPVI (9%), moderately preserve adhesion of platelets on VWF (7.4%) and collagen (8.4%), and partially restore the aggregation of NPSS-sheared platelets induced by ristocetin (18.6 AU*min) and collagen (48.2 AU*min). These results indicated that ADAM proteolysis played a role in NPSS-induced receptor shedding. However, the ADAM inhibition couldn't completely suppress the NPSS-caused loss of the platelet surface receptors (GPIbα and GPVI), only partially prevented the NPSS-induced reduction of platelet adhesion to VWF and collagen, and the agonist (ristocetin and collagen)-caused platelet aggregation. These results suggested that the direct mechanical damage is partially responsible for NPSS-induced receptor shedding in addition to the ADAM proteolysis. In conclusion, NPSS relevant to blood contacting medical devices can induce ADAM proteolysis and direct mechanical damage on the platelet receptor GPIbα and GPVI, leading to comprised hemostasis.
Collapse
Affiliation(s)
- Zengsheng Chen
- Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, China.,Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Douglas Tran
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland
| | - Tieluo Li
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland
| | - Katherin Arias
- Fischell Department of Bioengineering, A. James Clark School of Engineering, University of Maryland, College Park, Maryland
| | - Bartley P Griffith
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland
| | - Zhongjun J Wu
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland.,Fischell Department of Bioengineering, A. James Clark School of Engineering, University of Maryland, College Park, Maryland
| |
Collapse
|
7
|
Moroi M, Farndale RW, Jung SM. Activation-induced changes in platelet surface receptor expression and the contribution of the large-platelet subpopulation to activation. Res Pract Thromb Haemost 2020; 4:285-297. [PMID: 32110760 PMCID: PMC7040538 DOI: 10.1002/rth2.12303] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 12/02/2019] [Accepted: 12/17/2019] [Indexed: 11/08/2022] Open
Abstract
OBJECTIVE Platelet surface receptors are also present subcellularly in organelle membranes and can be expressed on the surface upon platelet activation. However, some receptors were reported to be decreased after activation. We analyzed the mechanism of activation-dependent expression for different receptors. METHODS Flow cytometry using platelet-rich plasma or washed platelets was used to analyze receptor-expression changes after platelet activation by glycoprotein (GP) VI-specific agonists, crosslinked collagen-related peptide (CRP-XL) and convulxin (Cvx), and thrombin. Platelets prelabeled with fluorescent antibody specific for a receptor were allowed to adhere on immobilized collagen or fibrinogen and post-stained with antibody against the same receptor labeled with another fluorophore, allowing us to differentiate preexisting receptors from newly expressed receptors. RESULTS Surface expression of αIIbβ3 increased in CRP-XL-, Cvx-, or thrombin-stimulated platelets, but GPIb decreased due to shedding and internalization. Both total and dimeric GPVI increased in thrombin-induced platelets, but decreased in platelets stimulated by Cvx, as a result of internalization. The larger platelets showed a greater increase in surface receptor (α2β1, αIIbβ3, GPVI, GPIb) expression upon activation compared to the smaller ones. Pre- and postlabeling with antibody specific for the same receptor, but conjugated with different fluorophores, allowed us to differentiate the receptors expressed on the surface of resting platelets from receptors newly exposed to the surface upon platelet activation. CONCLUSIONS Increased receptor expressions after activation are mainly manifested in the larger platelets. On platelets adhered on fibrinogen, the newly expressed receptors, especially GPVI, are localized in the lamellipodia of the spread platelets.
Collapse
Affiliation(s)
- Masaaki Moroi
- Department of BiochemistryUniversity of CambridgeCambridgeUK
| | | | | |
Collapse
|
8
|
Yakubu A, Azlan A, Loh SP, Md Noor S. Can Yellow Stripe Scad Compete with Salmon on Its Role in Platelet Phospholipid Membrane and Its Cardiovascular Benefits? J Obes 2019; 2019:4929131. [PMID: 31354987 PMCID: PMC6636454 DOI: 10.1155/2019/4929131] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 05/26/2019] [Indexed: 11/17/2022] Open
Abstract
This review article stresses the effective role of dietary fish fillet docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA) on overweight as a risk factor of cardiovascular disease (CVD) via platelet phospholipid modification. Several reports have demonstrated that saturated fat in overweight evokes systemic inflammation and more importantly predisposes it to cardiovascular disorder. Prospective studies have shown that saturated fat is directly proportional to the level of arachidonic acids (AA), precursor of thromboxane in the platelet phospholipid membrane as omega-6 fatty acid in overweight and obese people. Some literature has demonstrated that omega-3 fatty acid from fish fillet ameliorates inflammation, reduces proinflammatory cytokine, inhibits signaling pathway, and regulates the physical composition of inflammatory leukocytes and free radicals (ROS). Yellow stripe scad (YSS) is a local Malaysian fish that has been shown to contain a comparable level of EPA/DHA content as observed in salmon. This review article will focus on the dietary role of fish fillet that will balance the omega-6 fatty acid/omega-3 fatty acid ratio in platelet phospholipid from YSS to manage and prevent healthy overweight/obesity-related risk factor of CVD and to avoid the risk orthodox drug treatment.
Collapse
Affiliation(s)
- Abdulrahman Yakubu
- Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
- Department of Haematology, Faculty of Medical Laboratory Science, Usmanu Danfodiyo University Sokoto, P.M.P 2346, Sokoto, Northern Western, Nigeria
| | - Azrina Azlan
- Department of Nutrition and Dietetics, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Su Peng Loh
- Department of Nutrition and Dietetics, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Sabariah Md Noor
- Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| |
Collapse
|
9
|
Amelirad A, Shamsasenjan K, Akbarzadehlaleh P, Pashoutan Sarvar D. Signaling Pathways of Receptors Involved in Platelet Activation and Shedding of These Receptors in Stored Platelets. Adv Pharm Bull 2019; 9:38-47. [PMID: 31011556 PMCID: PMC6468227 DOI: 10.15171/apb.2019.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 10/25/2018] [Accepted: 11/12/2018] [Indexed: 12/26/2022] Open
Abstract
All cells encounter various signals coming from the surrounding environment and they need to receive and respond to these signals in order to perform their functions. Cell surface receptors are responsible for signal transduction .Platelets are blood cells which perform several functions using diverse receptors. Platelet concentrate is one of the most consumed blood products. However, due to the short lifespan of the platelets and platelets damage during storage, we face shortage of platelet products. One of the damages that platelets undergo during storage is the loss of surface receptors. Since cell surface receptors are responsible for all cell functions, the loss of platelet receptors reduces the quality of platelet products. In this study, we reviewed the important receptors involved in platelet activation and their associated signaling pathways. We also looked at the platelet receptors that shed during storage and the causes of this incident. We found that GPIbα, P-selectin, CD40 and GPVI are platelet receptors that fall during platelet storage at room temperature. Considering that GPVI and GPIbα are the most important receptors which involved in platelet activation, their shedding can cause decrease in platelet activation after transfusion and decrease thrombus consistence. Shear stress and platelet contact with the container wall are among the mechanisms discussed in this process, but studies in this area have to be continued.
Collapse
Affiliation(s)
- Asra Amelirad
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Karim Shamsasenjan
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parvin Akbarzadehlaleh
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | | |
Collapse
|
10
|
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
|
11
|
Pagel O, Walter E, Jurk K, Zahedi RP. Taking the stock of granule cargo: Platelet releasate proteomics. Platelets 2016; 28:119-128. [PMID: 27928935 DOI: 10.1080/09537104.2016.1254762] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Human platelets are key players in a multitude of physiological and pathological processes. Upon activation they release cargo from different types of granules as well as microparticles in an apparently well-regulated and orchestrated manner. The resulting specific platelet releasates create microenvironments of biologically active compounds and proteins during platelet aggregation and thrombus formation, allowing efficient delivery of growth factors and immune modulators to their sites of effect and enhancing the coagulative response in a positive feedback loop. Thus, platelet releasates play a central role in the regulation of platelet homeostasis and heterotypic cell interaction. Additionally, it recently emerged that both the qualitative and quantitative composition of the releasate as well as release dynamics may be stimulus dependent and therefore more complex than expected. Mass spectrometry-based proteomics is an important asset for studying platelet releasates in vitro, as it allows not only (i) identifying released proteins, but moreover (ii) determining their quantities and the dynamics of release as well as (iii) differentially comparing releasates across a variety of conditions. Though owing to the high sensitivity and comprehensiveness of modern proteomic techniques, a thorough experimental design and a standardized and robust sample preparation are essential to obtain highly confident and reliable insights into platelet biology and pathology. Here, we review releasate proteome studies and crucial sample preparation strategies to summarize possible achievements of state-of-the-art technologies and furthermore discuss potential pitfalls and limitations. We provide a future perspective of platelet releasate proteomics including targeted analyses, post-translational modifications and multi-omics approaches that should be adopted by platelet releasate researchers due to their tremendous depth and comprehensiveness.
Collapse
Affiliation(s)
- Oliver Pagel
- a Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V ., Dortmund , Germany
| | - Elena Walter
- b Center for Thrombosis and Hemostasis (CTH) , Universitätsklinikum der Johannes Gutenberg-Universität Mainz , Mainz , Germany
| | - Kerstin Jurk
- b Center for Thrombosis and Hemostasis (CTH) , Universitätsklinikum der Johannes Gutenberg-Universität Mainz , Mainz , Germany
| | - René P Zahedi
- a Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V ., Dortmund , Germany
| |
Collapse
|
12
|
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
|
13
|
Xu XR, Zhang D, Oswald BE, Carrim N, Wang X, Hou Y, Zhang Q, Lavalle C, McKeown T, Marshall AH, Ni H. Platelets are versatile cells: New discoveries in hemostasis, thrombosis, immune responses, tumor metastasis and beyond. Crit Rev Clin Lab Sci 2016; 53:409-30. [PMID: 27282765 DOI: 10.1080/10408363.2016.1200008] [Citation(s) in RCA: 208] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Platelets are small anucleate blood cells generated from megakaryocytes in the bone marrow and cleared in the reticuloendothelial system. At the site of vascular injury, platelet adhesion, activation and aggregation constitute the first wave of hemostasis. Blood coagulation, which is initiated by the intrinsic or extrinsic coagulation cascades, is the second wave of hemostasis. Activated platelets can also provide negatively-charged surfaces that harbor coagulation factors and markedly potentiate cell-based thrombin generation. Recently, deposition of plasma fibronectin, and likely other plasma proteins, onto the injured vessel wall has been identified as a new "protein wave of hemostasis" that may occur even earlier than the first wave of hemostasis, platelet accumulation. Although no experimental evidence currently exists, it is conceivable that platelets may also contribute to this protein wave of hemostasis by releasing their granule fibronectin and other proteins that may facilitate fibronectin self- and non-self-assembly on the vessel wall. Thus, platelets may contribute to all three waves of hemostasis and are central players in this critical physiological process to prevent bleeding. Low platelet counts in blood caused by enhanced platelet clearance and/or impaired platelet production are usually associated with hemorrhage. Auto- and allo-immune thrombocytopenias such as idiopathic thrombocytopenic purpura and fetal and neonatal alloimmune thrombocytopenia may cause life-threatening bleeding such as intracranial hemorrhage. When triggered under pathological conditions such as rupture of an atherosclerotic plaque, excessive platelet activation and aggregation may result in thrombosis and vessel occlusion. This may lead to myocardial infarction or ischemic stroke, the major causes of mortality and morbidity worldwide. Platelets are also involved in deep vein thrombosis and thromboembolism, another leading cause of mortality. Although fibrinogen has been documented for more than half a century as essential for platelet aggregation, recent studies demonstrated that fibrinogen-independent platelet aggregation occurs in both gene deficient animals and human patients under physiological and pathological conditions (non-anti-coagulated blood). This indicates that other unidentified platelet ligands may play important roles in thrombosis and might be novel antithrombotic targets. In addition to their critical roles in hemostasis and thrombosis, emerging evidence indicates that platelets are versatile cells involved in many other pathophysiological processes such as innate and adaptive immune responses, atherosclerosis, angiogenesis, lymphatic vessel development, liver regeneration and tumor metastasis. This review summarizes the current knowledge of platelet biology, highlights recent advances in the understanding of platelet production and clearance, molecular and cellular events of thrombosis and hemostasis, and introduces the emerging roles of platelets in the immune system, vascular biology and tumorigenesis. The clinical implications of these basic science and translational research findings will also be discussed.
Collapse
Affiliation(s)
- Xiaohong Ruby Xu
- a Department of Laboratory Medicine and Pathobiology , University of Toronto , Toronto , ON , Canada .,b Department of Laboratory Medicine , Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Li Ka Shing Knowledge Institute , Toronto , ON , Canada .,c Department of Medicine , Guangzhou University of Chinese Medicine , Guangzhou , Guangdong , P.R. China
| | - Dan Zhang
- b Department of Laboratory Medicine , Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Li Ka Shing Knowledge Institute , Toronto , ON , Canada .,c Department of Medicine , Guangzhou University of Chinese Medicine , Guangzhou , Guangdong , P.R. China
| | - Brigitta Elaine Oswald
- b Department of Laboratory Medicine , Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Li Ka Shing Knowledge Institute , Toronto , ON , Canada .,d Canadian Blood Services , Toronto , ON , Canada .,e Department of Physiology , University of Toronto , Toronto , ON , Canada
| | - Naadiya Carrim
- a Department of Laboratory Medicine and Pathobiology , University of Toronto , Toronto , ON , Canada .,b Department of Laboratory Medicine , Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Li Ka Shing Knowledge Institute , Toronto , ON , Canada .,d Canadian Blood Services , Toronto , ON , Canada
| | - Xiaozhong Wang
- b Department of Laboratory Medicine , Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Li Ka Shing Knowledge Institute , Toronto , ON , Canada .,f The Second Affiliated Hospital of Nanchang University , Nanchang , Jiangxi , P.R. China
| | - Yan Hou
- b Department of Laboratory Medicine , Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Li Ka Shing Knowledge Institute , Toronto , ON , Canada .,g Jilin Provincial Center for Disease Prevention and Control , Changchun , Jilin , P.R. China
| | - Qing Zhang
- b Department of Laboratory Medicine , Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Li Ka Shing Knowledge Institute , Toronto , ON , Canada .,h State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University , Guangzhou , Guangdong , P.R. China , and
| | - Christopher Lavalle
- b Department of Laboratory Medicine , Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Li Ka Shing Knowledge Institute , Toronto , ON , Canada .,e Department of Physiology , University of Toronto , Toronto , ON , Canada
| | - Thomas McKeown
- b Department of Laboratory Medicine , Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Li Ka Shing Knowledge Institute , Toronto , ON , Canada
| | - Alexandra H Marshall
- b Department of Laboratory Medicine , Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Li Ka Shing Knowledge Institute , Toronto , ON , Canada
| | - Heyu Ni
- a Department of Laboratory Medicine and Pathobiology , University of Toronto , Toronto , ON , Canada .,b Department of Laboratory Medicine , Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Li Ka Shing Knowledge Institute , Toronto , ON , Canada .,d Canadian Blood Services , Toronto , ON , Canada .,e Department of Physiology , University of Toronto , Toronto , ON , Canada .,i Department of Medicine , University of Toronto , Toronto , ON , Canada
| |
Collapse
|
14
|
Naitoh K, Hosaka Y, Honda M, Ogawa K, Shirakawa K, Furusako S. Properties of soluble glycoprotein VI, a potential platelet activation biomarker. Platelets 2014; 26:745-50. [PMID: 25549161 DOI: 10.3109/09537104.2014.991707] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Glycoprotein VI (GPVI) plays a critical role in the platelet response to collagen. Clinical studies suggest that the plasma level of soluble GPVI (sGPVI) is a highly specific and useful platelet activation marker. However, many properties of sGPVI have not been fully characterized, such as its sensitivity in detecting platelet activation and its elimination rate from the blood. In this study we established a sandwich enzyme-linked immunosorbent assay for human sGPVI, which cross-reacts to cynomolgus monkey sGPVI, and evaluated the time course of sGPVI production in a cynomolgus monkey model of lipopolysaccharide (LPS)-induced thrombocytopenia. The sGPVI levels in this model were dramatically elevated and returned to baseline by 24 hours after LPS injection, the change was more pronounced than the existing platelet activation biomarker, soluble P-selectin (sP-selectin) levels. The elimination half-life of recombinant human sGPVI was about 2.5 hours following intravenous administration to monkeys. These results suggest that plasma sGPVI closely reflects platelet activation in the bloodstream and has a short half-life. sGPVI would be a useful biomarker for disorders marked by platelet activation and for monitoring anti-platelet therapy.
Collapse
Affiliation(s)
- Katsuki Naitoh
- a Biology Laboratory, Discovery Research, Mochida Pharmaceutical Co., LTD. , Jimba , Gotemba , Shizuoka , Japan
| | - Yoshitaka Hosaka
- a Biology Laboratory, Discovery Research, Mochida Pharmaceutical Co., LTD. , Jimba , Gotemba , Shizuoka , Japan
| | - Motoyasu Honda
- a Biology Laboratory, Discovery Research, Mochida Pharmaceutical Co., LTD. , Jimba , Gotemba , Shizuoka , Japan
| | - Kumiko Ogawa
- a Biology Laboratory, Discovery Research, Mochida Pharmaceutical Co., LTD. , Jimba , Gotemba , Shizuoka , Japan
| | - Kamon Shirakawa
- a Biology Laboratory, Discovery Research, Mochida Pharmaceutical Co., LTD. , Jimba , Gotemba , Shizuoka , Japan
| | - Shoji Furusako
- a Biology Laboratory, Discovery Research, Mochida Pharmaceutical Co., LTD. , Jimba , Gotemba , Shizuoka , Japan
| |
Collapse
|
15
|
Clinically relevant concepts of haemostasis and detection of coagulopathy after trauma and surgery. ACTA ACUST UNITED AC 2014; 60:485-8. [PMID: 25374983 DOI: 10.1016/j.redar.2013.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
16
|
Identification of a calmodulin-binding domain in Sema4D that regulates its exodomain shedding in platelets. Blood 2013; 121:4221-30. [PMID: 23564909 DOI: 10.1182/blood-2012-11-470609] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Semaphorin 4D (Sema4D) is a transmembrane protein that supports contact-dependent amplification of platelet activation by collagen before being gradually cleaved by the metalloprotease ADAM17, as we have previously shown. Cleavage releases a soluble 120-kDa exodomain fragment for which receptors exist on platelets and endothelial cells. Here we have examined the mechanism that regulates Sema4D exodomain cleavage. The results show that the membrane-proximal cytoplasmic domain of Sema4D contains a binding site for calmodulin within the polybasic region Arg762-Lys779. Coprecipitation studies show that Sema4D and calmodulin are associated in resting platelets, forming a complex that dissociates upon platelet activation by the agonists that trigger Sema4D cleavage. Inhibiting calmodulin with W7 or introducing a membrane-permeable peptide corresponding to the calmodulin-binding site is sufficient to trigger the dissociation of Sema4D from calmodulin and initiate cleavage. Conversely, deletion of the calmodulin-binding site causes constitutive shedding of Sema4D. These results show that (1) Sema4D is a calmodulin-binding protein with a site of interaction in its membrane-proximal cytoplasmic domain, (2) platelet agonists cause dissociation of the calmodulin-Sema4D complex, and (3) dissociation of the complex is sufficient to trigger ADAM17-dependent cleavage of Sema4D, releasing a bioactive fragment.
Collapse
|
17
|
Hofmann S, Vögtle T, Bender M, Rose-John S, Nieswandt B. The SLAM family member CD84 is regulated by ADAM10 and calpain in platelets. J Thromb Haemost 2012; 10:2581-92. [PMID: 23025437 DOI: 10.1111/jth.12013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
BACKGROUND AND OBJECTIVE Ectodomain shedding is a major mechanism to modulate platelet receptor signaling and to downregulate platelet reactivity. Proteins of the a disintegrin and metalloproteinase (ADAM) family are implicated in the shedding of various platelet receptors. The signaling lymphocyte activation molecule (SLAM) family receptor CD84 is highly expressed in platelets and immune cells, but its role in platelet physiology is not well explored. Because of its ability to form homodimers, CD84 has been suggested to mediate contact-dependent signaling and contribute to thrombus stability. However, nothing is known about the cellular regulation of CD84. METHODS We studied the regulation of CD84 in murine platelets by biochemical approaches and use of three different genetically modified mouse lines. Regulation of CD84 in human platelets was studied using inhibitors and biochemical approaches. RESULTS We show that CD84 is cleaved from the surface of human and murine platelets in response to different shedding inducing agents and platelet receptor agonists. CD84 downregulation occurs through ectodomain-shedding and intracellular cleavage. Studies in transgenic mice identified ADAM10 as the principal sheddase responsible for CD84 cleavage, whereas ADAM17 was dispensable. Western blot analyses revealed calpain-mediated intracellular cleavage of the CD84 C-terminus, occurring simultaneously with, but independently of, ectodomain shedding. Furthermore, analysis of plasma and serum samples from transgenic mice demonstrated that CD84 is constitutively shed from the platelet surface by ADAM10 in vivo. CONCLUSIONS These results reveal a dual regulation mechanism for platelet CD84 by simultaneous extra- and intracellular cleavage that may modulate platelet-platelet and platelet-immune cell interactions.
Collapse
Affiliation(s)
- S Hofmann
- Chair of Vascular Medicine, University of Würzburg, University Hospital and Rudolf Virchow Center for Experimental Biomedicine, Würzburg, Germany
| | | | | | | | | |
Collapse
|
18
|
Ferroni P, Riondino S, Vazzana N, Santoro N, Guadagni F, Davì G. Biomarkers of platelet activation in acute coronary syndromes. Thromb Haemost 2012; 108:1109-23. [PMID: 23014768 DOI: 10.1160/th12-08-0550] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2012] [Accepted: 09/07/2012] [Indexed: 12/17/2022]
Abstract
The most convincing evidence for the participation of platelets in arterial thrombosis in humans comes from studies of platelet activation in patients with acute coronary syndromes (ACS) and from trials of antiplatelet drugs. Both strongly support the concept that repeated episodes of platelet activation over the thrombogenic surface of a vulnerable plaque may contribute to the risk of death from coronary causes. However, the relation of in vivo platelet activation and adverse clinical events to results of platelet function tests remains largely unknown. A valuable marker of in vivo platelet activation should be specific, unaltered by pre-analytical artefacts and reproducibly measured by easily performed methods. This article describes current biomarkers of platelet activation in ACS, reviews their advantages and disadvantages, discusses their potential pitfalls, and demonstrates emerging data supporting the positive clinical implications of monitoring in vivo platelet activation in the setting of ACS.
Collapse
Affiliation(s)
- Patrizia Ferroni
- Department of Advanced Biotechnologies and Bioimaging, IRCCS San Raffaele Pisana, Rome, Italy
| | | | | | | | | | | |
Collapse
|
19
|
Abstract
Receptor shedding is a mechanism for irreversible removal of transmembrane cell surface receptors by proteolysis of the receptor at a position near the extracellular surface of the plasma membrane. This process generates a soluble ectodomain fragment and a membrane-associated remnant fragment, and is distinct from loss of receptor surface expression by internalization or microparticle release or secretion of alternatively spliced soluble forms of receptors lacking a transmembrane domain. There has been an increased focus on new methods for analyzing shedding of platelet glycoprotein (GP)Ib-IX-V and GPVI because these receptors are platelet specific and are critical for the initiation of platelet adhesion and activation in thrombus formation at arterial shear rates. Platelet receptor shedding provides a mechanism for downregulating surface expression resulting in loss of ligand binding, decreasing the surface density affecting receptor cross linking and signalling and generation of proteolytic fragments that may be functional and/or provide platelet-specific biomarkers.
Collapse
Affiliation(s)
- Elizabeth E Gardiner
- Australian Centre for Blood Diseases, Monash University, Melbourne, VIC, Australia
| | | | | | | |
Collapse
|
20
|
Straub A, Krajewski S, Hohmann JD, Westein E, Jia F, Bassler N, Selan C, Kurz J, Wendel HP, Dezfouli S, Yuan Y, Nandurkar H, Jackson S, Hickey MJ, Peter K. Evidence of platelet activation at medically used hypothermia and mechanistic data indicating ADP as a key mediator and therapeutic target. Arterioscler Thromb Vasc Biol 2011; 31:1607-16. [PMID: 21512161 DOI: 10.1161/atvbaha.111.226373] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Hypothermia is used in various clinical settings to inhibit ischemia-related organ damage. However, prothrombotic effects have been described as potential side effects. This study aimed to elucidate the mechanism of hypothermia-induced platelet activation and subsequent prothrombotic events and to develop preventative pharmacological strategies applicable during clinically used hypothermia. METHODS AND RESULTS Platelet function was investigated ex vivo and in vivo at clinically used hypothermia (28°C/18°C). Hypothermic mice demonstrated increased expression of platelet activation marker P-selectin, platelet-leukocyte aggregate formation, and thrombocytopenia. Intravital microscopy of FeCl(3)-injured murine mesenteric arteries revealed increased platelet thrombus formation with hypothermia. Ex vivo flow chamber experiments indicated increased platelet-fibrinogen adhesion under hypothermia. We show that hypothermia results in reduced ADP hydrolysis via reduction of CD39 (E-NTPDase1) activity, resulting in increased levels of ADP and subsequent augmented primary and secondary platelet activation. In vivo administration of ADP receptor P(2)Y(12) antagonists and recombinant soluble CD39 prevented hypothermia-induced thrombus formation and thrombocytopenia, respectively. CONCLUSIONS The platelet agonist ADP plays a key role in hypothermia-induced platelet activation. Inhibition of receptor binding or hydrolysis of ADP has the potential to protect platelets against hypothermia-induced activation. Our findings provide a rational basis for further evaluation of novel antithrombotic strategies in clinically applied hypothermia.
Collapse
Affiliation(s)
- Andreas Straub
- Atherothrombosis and Vascular Biology, Baker IDI Heart and Diabetes Institute, PO Box 6492 St Kilda Rd Central, Melbourne, Victoria 8008, Australia.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Abstract
Activated platelets shed surface proteins, potentially modifying platelet function as well as providing a source of bioactive fragments. Previous studies have identified several constituents of the platelet sheddome, but the full extent of shedding is unknown. Here we have taken a global approach, analyzing protein fragments in the supernate of activated platelets using mass spectroscopy and looking for proteins originating from platelet membranes. After removing plasma proteins and microparticles, 1048 proteins were identified, including 69 membrane proteins. Nearly all of the membrane proteins had been detected previously, but only 10 had been shown to be shed in platelets. The remaining 59 are candidates subject to confirmation. Based on spectral counts, protein representation in the sheddome varies considerably. As proof of principle, we validated one of the less frequently detected proteins, semaphorin 7A, which had not previously been identified in platelets. Surface expression, cleavage, and shedding of semaphorin 7A were demonstrated, as was its association with α-granules. Finally, cleavage of semaphorin 7A and 12 other proteins was substantially reduced by an inhibitor of ADAM17, a known sheddase. These results define a subset of membrane proteins as sheddome candidates, forming the basis for further studies examining the impact of ectodomain shedding on platelet function.
Collapse
|
22
|
Wijeyewickrema LC, Gardiner EE, Gladigau EL, Berndt MC, Andrews RK. Nerve growth factor inhibits metalloproteinase-disintegrins and blocks ectodomain shedding of platelet glycoprotein VI. J Biol Chem 2010; 285:11793-9. [PMID: 20164177 DOI: 10.1074/jbc.m110.100479] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Nerve growth factor (NGF) plays an important role in regulating mammalian neuronal/embryonic development, angiogenesis, and other physiological processes and has recently been investigated as a potential treatment for the neurodegenerative disorder, Alzheimer disease. In this study, we provide evidence that human NGF may also function as a metalloproteinase inhibitor, based on studies of NGF from snake venom. Originally, our aim was to isolate snake venom metalloproteinases targeting platelet receptors and/or ligands relevant to hemostasis and thrombosis, using Ni(2+)-agarose as a purification step based on the conserved metal ion-coordination motif in venom metalloproteinases. However, subsequent analysis of cobra (Naja kaouthia) venom led to the unexpected discovery that cobra venom NGF bound to Ni(2+)-agarose, eluting at approximately 15 mm imidazole, enabling a one-step purification. The identity of the purified protein was confirmed by mass spectrometry and N-terminal sequence analysis. Partial co-purification of NGF within metalloproteinase-enriched venom fractions led us to test whether NGF affected metalloproteinase activity. Venom NGF potently inhibited metalloproteinases isolated from the same or different venom and specifically bound to purified Nk metalloproteinase immobilized on agarose beads. Human NGF also interacted with human metalloproteinases because it blocked metalloproteinase-mediated shedding of the platelet collagen receptor, glycoprotein (GP)VI, and associated with recombinant ADAM10 by surface plasmon resonance. Together, these results suggest that NGF can function as a metalloproteinase inhibitor.
Collapse
Affiliation(s)
- Lakshmi C Wijeyewickrema
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Victoria 3004, Australia
| | | | | | | | | |
Collapse
|
23
|
Abstract
Although platelets are best known as primary mediators of hemostasis, this function intimately associates them with inflammatory processes, and it has been increasingly recognized that platelets play an active role in both innate and adaptive immunity. For example, platelet adhesive interactions with leukocytes and endothelial cells via P-selectin can lead to several pro-inflammatory events, including leukocyte rolling and activation, production of cytokine cascades, and recruitment of the leukocytes to sites of tissue damage. Superimposed on this, platelets express immunologically-related molecules such as CD40L and Toll-like receptors that have been shown to functionally modulate innate immunity. Furthermore, platelets themselves can interact with microorganisms, and several viruses have been shown to cross-react immunologically with platelet antigens. This review discusses the central role that platelets play in inflammation, linking them with varied pathological conditions, such as atherosclerosis, sepsis, and immune thrombocytopenic purpura, and suggests that platelets also act as primary mediators of our innate defences.
Collapse
Affiliation(s)
- John W Semple
- Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael's Hospital, 30 Bond Street, Toronto, ON M5B 1W8, Canada.
| | | |
Collapse
|
24
|
Brass L. Understanding and evaluating platelet function. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2010; 2010:387-396. [PMID: 21239824 DOI: 10.1182/asheducation-2010.1.387] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
The contribution of platelets to normal hemostasis and vascular disease is well described. However, recent studies make it clear that much remains to be learned about platelet activation at the single cell and the molecular level, and about the contribution of platelets to inflammation, tumor angiogenesis, and embryonic development. This article is divided into two themes. The first is an overview of current knowledge of the mechanisms that drive platelet function in vivo and a brief summary of some of the emerging ideas that are modifying older views. The second theme is a consideration of the strengths and weaknesses of the tools we have as hematologists to assess platelet function in the clinical setting, identify mechanisms, and evaluate the impact of antiplatelet agents.
Collapse
Affiliation(s)
- Lawrence Brass
- Hematology-Oncology Division, Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19004, USA.
| |
Collapse
|
25
|
Fibrinogen is required for maintenance of platelet intracellular and cell-surface P-selectin expression. Blood 2009; 114:425-36. [DOI: 10.1182/blood-2008-03-145821] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Platelet P-selectin plays important roles in inflammation and contributes to thrombosis and hemostasis. Although it has been reported that von Willebrand factor (VWF) affects P-selectin expression on endothelial cells, little information is available regarding regulation of platelet P-selectin expression. Here, we first observed that P-selectin expression was significantly decreased on platelets of fibrinogen and VWF double-deficient mice. Subsequently, we identified this was due to fibrinogen deficiency. Impaired P-selectin expression on fibrinogen-deficient platelets was further confirmed in human hypofibrinogenemic patients. We demonstrated that this impairment is unlikely due to excessive P-selectin shedding, deficient fibrinogen-mediated cell surface P-selectin binding, or impaired platelet granule release, but rather is due to decreased platelet P-selectin content. Fibrinogen transfusion completely recovered this impairment in fibrinogen-deficient (Fg−/−) mice, and engagement of the C-terminus of the fibrinogen γ chain with β3 integrin was required for this process. Furthermore, Fg−/− platelets significantly increased P-selectin expression following transfusion into β3 integrin–deficient mice and when cultured with fibrinogen. These data suggest fibrinogen may play important roles in inflammation, thrombosis, and hemostasis via enhancement of platelet P-selectin expression. Since human fibrinogen levels vary significantly in normal and diseased populations, P-selectin as an activation marker on platelets should be used with caution.
Collapse
|
26
|
Al-Tamimi M, Mu FT, Arthur JF, Shen Y, Moroi M, Berndt MC, Andrews RK, Gardiner EE. Anti-glycoprotein VI monoclonal antibodies directly aggregate platelets independently of FcγRIIa and induce GPVI ectodomain shedding. Platelets 2009; 20:75-82. [DOI: 10.1080/09537100802645029] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
27
|
Bigalke B, Stellos K, Weig HJ, Geisler T, Seizer P, Kremmer E, Pötz O, Joos T, May AE, Lindemann S, Gawaz M. Regulation of platelet glycoprotein VI (GPVI) surface expression and of soluble GPVI in patients with atrial fibrillation (AF) and acute coronary syndrome (ACS). Basic Res Cardiol 2009; 104:352-7. [PMID: 19190951 DOI: 10.1007/s00395-009-0779-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2008] [Accepted: 01/05/2009] [Indexed: 11/25/2022]
Abstract
BACKGROUND The platelet collagen receptor glycoprotein VI (GPVI) mediates platelet adhesion to subendothelial matrix and thrombus formation in acute coronary syndrome (ACS). This study examined patients with both ACS and stable coronary artery disease (CAD), which presented with atrial fibrillation (AF) and sinus rhythm (SR). METHODS AND RESULTS We evaluated 992 patients with acute or stable CAD, and determined platelet surface expression of GPVI using flow cytometry. Seventy-eight patients presented with nonvalvular persistent AF. After 1:1 propensity score matching 156 matched cases with 78 pairs were obtained. Patients with AF and ACS showed a significantly decreased GPVI expression compared to patients with ACS and SR, whereas patients with stable angina pectoris (SA) presented with low level activation and no significant difference between SR and AF [mean fluorescence intensity (MFI) for ACS (SR Vs. AF): 20 +/- 6.3 Vs. 17.7 +/- 4.4; P = 0.023; SA (SR Vs. AF): 18.8 +/- 9.4 Vs. 18.1 +/- 6.1; P = 0.649]. In contrast, soluble GPVI was increased in ACS and AF accordingly [plasma GPVI (ng/ml) for ACS (SR Vs. AF): 1.4 +/- 0.8 Vs. 1.9 +/- 1.1; P = 0.038; SA (SR Vs. AF): 0.9 +/- 0.4 Vs. 1.1 +/- 0.5; P = 0.127]. CONCLUSION Platelet GPVI surface expression is decreased in patients with AF and ACS compared to patients with SR and ACS. Nonvalvular AF is related to indices of chronic platelet activation and might be responsible for a down-regulation of GPVI receptor density on platelets, while soluble GPVI was increased in ACS and AF accordingly.
Collapse
Affiliation(s)
- Boris Bigalke
- Medizinische Klinik III, Klinik für Kardiologie und Kreislauferkrankungen, Eberhard-Karls-Universität Tübingen, Tübingen, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Hsu CC, Wu WB, Huang TF. A snake venom metalloproteinase, kistomin, cleaves platelet glycoprotein VI and impairs platelet functions. J Thromb Haemost 2008; 6:1578-85. [PMID: 18624975 DOI: 10.1111/j.1538-7836.2008.03071.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
BACKGROUND AND OBJECTIVES Injuries to the vessel wall and subsequent exposure of the matrix of the subendothelial layer resulted in thrombus formation. Platelet glycoprotein (GP) Ib and VI play a crucial role in matrix-induced activation and aggregation of platelets. METHODS AND RESULTS In the present study, we reported that the GPIb-cleaving snake venom metalloproteinase (SVMP), kistomin, inhibited collagen-induced platelet aggregation. Moreover, kistomin inhibited platelet aggregation induced by convulxin (CVX, a GPVI agonist) and a GPVI-specific antibody in a concentration and time-dependent manner. Kistomin treatment decreased platelet GPVI but not integrin alpha2beta1 and alphaIIbbeta3, accompanied with the formation of GPVI cleavage fragments, as determined by flow cytometric and Western blot analyses. In addition, intact platelet GPVI and recombinant GPVI were digested by kistomin to release 25- and 35-kDa fragments, suggesting that kistomin cleaved GPVI near the mucin-like region. We designed four synthetic peptides ranging from Leu180 to Asn249 as the substrates for kistomin and found that kistomin cleaved these synthetic peptides at FSE205/A206TA and NKV218/F219TT, as analyzed by MALDI-TOF-MS. In addition, GPVI-specific antibody-induced tyrosine kinase phosphorylation in platelets was reduced after kistomin pretreatment, and platelet adhesion to collagen but not to fibrinogen was attenuated by kistomin. CONCLUSIONS We provided here the first evidence that a P-I snake venom metalloproteinase, kistomin, inhibits the interaction between collagen and platelet GPVI through its proteolytic activity on GPVI, thus providing an alternative strategy for developing new anti-thrombotic agents.
Collapse
Affiliation(s)
- C C Hsu
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | | | | |
Collapse
|
29
|
Abstract
Several recent findings point to an important role for redox regulation of platelet responses to collagen involving the receptor, glycoprotein (GP)VI. First, the antioxidant dietary compound, quercetin, was shown to inhibit GPVI-dependent platelet activation and signaling responses to collagen. Second, collagen increased platelet production of the oxygen radical, superoxide anion (O2-), mediated by the multi-subunit enzyme nicotinamide adenine dinucleotide (phosphate) (NAD(P)H) oxidase. In that case, O2- was implicated in regulating not initial aggregation, but collagen-induced thrombus stabilization involving release of ADP. Third, our laboratory showed that an unpaired thiol in the GPVI cytoplasmic tail undergoes rapid oxidation to form GPVI homodimers following ligand binding, preceding GPVI signaling and ectodomain metalloproteolysis, and indicating formation of an oxidative submembranous environment in activated platelets. This review examines receptor/redox regulation in other cells, and relevance to the pathophysiological function of GPVI and other platelet receptors initiating thrombus formation in haemostasis or thrombotic diseases such as heart attack and stroke.
Collapse
Affiliation(s)
- Jane F Arthur
- Department of Immunology, Monash University, Alfred Medical Research & Education Precinct, Melbourne 3004, Victoria, Australia
| | | | | | | | | |
Collapse
|
30
|
Abstract
Platelet disorders are common bleeding disorders, with a variety of congenital and acquired causes. The diagnostic evaluation of platelet disorders challenges both clinicians and clinical laboratories, as testing for these conditions is complex, not well standardized and time consuming. An understanding of normal platelet function has provided insights on the pathogenesis of many platelet function disorders. Knowledge of the key features of platelet disorders aids their diagnostic assessment. Tests for aggregation, secretion and dense granule defects continue to be the most helpful for the evaluation of suspected platelet function disorders.
Collapse
Affiliation(s)
- Catherine P M Hayward
- Department of Pathology and Molecular Medicine, Room 2N30, McMaster University Medical Center, 1200 Main Street West, Hamilton, Ontario, Canada L8N 3Z5.
| |
Collapse
|
31
|
Braking platelet activation: deactivating the receptors. Blood 2008. [DOI: 10.1182/blood-2007-09-113365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Glycoprotein (GP) VI and Fc receptor FcγRIIa are 2 ITAM (immunoreceptor tyrosine-activation motif)–bearing platelet surface receptors. In this issue of Blood, Gardiner and colleagues show that activation of either receptor results in simultaneous proteolytic cleavage of GPVI ectodomain and FcγRIIa cytoplasmic tail, providing distinct mechanisms for their down-regulation on platelet activation.
Collapse
|
32
|
Wijeyewickrema LC, Gardiner EE, Shen Y, Berndt MC, Andrews RK. Fractionation of snake venom metalloproteinases by metal ion affinity: a purified cobra metalloproteinase, Nk, from Naja kaouthia binds Ni2+-agarose. Toxicon 2007; 50:1064-72. [PMID: 17822731 DOI: 10.1016/j.toxicon.2007.07.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2007] [Revised: 07/20/2007] [Accepted: 07/20/2007] [Indexed: 11/29/2022]
Abstract
Snake venom metalloproteinases represent unique probes for analyzing platelet adhesion receptors regulating hemostasis and thrombosis. Snake venom metalloproteinase-disintegrins consist of a propeptide domain, a catalytic domain containing a metal ion-coordination sequence (HEXXHXXGXXH), a disintegrin domain, and a Cys-rich domain. Here, we investigate whether metal ion-affinity chromatography may be used to fractionate venom metalloproteinases based on the metal ion-coordination motif. First, we showed that a purified cobra metalloproteinase, Nk, from Naja kaouthia bound Ni(2+)-agarose, and was eluted by approximately 10mM imidazole, confirming the validity of the approach. Nk cleaved the platelet von Willebrand factor (VWF) receptor, glycoprotein (GP)Ibalpha, with similar activity to the previously reported cobra metalloproteinase, mocarhagin, as shown by EDTA-inhibitable Nk-dependent proteolysis of a purified GPIbalpha extracellular fragment (glycocalicin), and inhibition of (125)I-VWF binding to GPIbalpha on washed human or canine platelets. Second, crude venom from the viper, Trimeresurus albolabris, was fractionated on Ni(2+)-agarose. Samples of flow-through, wash, and imidazole-eluted (0-30mM gradient) fractions were analyzed by (i) SDS-polyacrylamide gel electrophoresis, (ii) immunoblotting with a rabbit anti-mocarhagin antibody, and (iii) assessing metalloproteinase activity using human fibrinogen as substrate. The combined results support the general concept of using Ni(2+)-agarose to fractionate snake venom metalloproteinases.
Collapse
Affiliation(s)
- Lakshmi C Wijeyewickrema
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, 3008 Victoria, Australia
| | | | | | | | | |
Collapse
|