1
|
Das K, Rao LVM. Coagulation protease-induced extracellular vesicles: their potential effects on coagulation and inflammation. J Thromb Haemost 2024; 22:2976-2990. [PMID: 39127325 DOI: 10.1016/j.jtha.2024.07.022] [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] [Received: 04/26/2024] [Revised: 07/09/2024] [Accepted: 07/15/2024] [Indexed: 08/12/2024]
Abstract
Coagulation proteases, in addition to playing an essential role in blood coagulation, often influence diverse cellular functions by inducing specific signaling pathways via the activation of protease-activated receptors (PARs). PAR activation-induced cellular effects are known to be cell-specific as PARs are expressed selectively in specific cell types. However, a growing body of evidence indicates that coagulation protease-induced PAR activation in a specific cell type could affect cellular responses in other cell types via communicating through extracellular vesicles (EVs) as coagulation protease-induced PAR signaling could promote the release of EVs in various cell types. EVs are membrane-enclosed nanosized vesicles that facilitate intercellular communication by transferring bioactive molecules, such as proteins, lipids, messenger RNAs, and microRNAs, etc., from donor cells to recipient cells. Our recent findings established that factor (F)VIIa promotes the release of EVs from vascular endothelium via endothelial cell protein C receptor-dependent activation of PAR1-mediated biased signaling. FVIIa-released EVs exhibit procoagulant activity and cytoprotective responses in both in vitro and in vivo model systems. This review discusses how FVIIa and other coagulation proteases trigger the release of EVs. The review specifically discusses how FVIIa-released EVs are enriched with phosphatidylserine and anti-inflammatory microRNAs and the impact of FVIIa-released EVs on hemostasis in therapeutic settings. The review also briefly highlights the therapeutic potential of FVIIa-released EVs in treating bleeding and inflammatory disorders, such as hemophilic arthropathy.
Collapse
Affiliation(s)
- Kaushik Das
- Biotechnology Research and Innovation Council-National Institute of Biomedical Genomics, Kalyani, West Bengal, India
| | - L Vijaya Mohan Rao
- Department of Cellular and Molecular Biology, The University of Texas at Tyler School of Medicine, The University of Texas at Tyler Health Science Center, Tyler, Texas, USA.
| |
Collapse
|
2
|
Ye SL, Li WD, Li WX, Xiao L, Ran F, Chen MM, Li XQ, Sun LL. The regulatory role of exosomes in venous thromboembolism. Front Cell Dev Biol 2022; 10:956880. [PMID: 36092737 PMCID: PMC9449368 DOI: 10.3389/fcell.2022.956880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 07/26/2022] [Indexed: 11/25/2022] Open
Abstract
Exosomes are nanoscale endocytic vesicles, 30-150 nm in diameter, secreted by most cells. They mainly originate from multivesicular bodies formed by intracellular invagination of lysosomal microparticles, and released into the extracellular matrix after fusion of multivesicular bodies with cell membrane. Studies have shown that exosomes contain a variety of active molecules, such as proteins, lipids and RNAs (such as mRNA, miRNA, lncRNA, circRNA, etc.), which regulate the behavior of recipient cells and serve as circulating biomarkers of diseases, including thrombosis. Therefore, exosome research is important for the diagnosis, treatment, therapeutic monitoring, and prognosis of thrombosis in that it can reveal the counts, surface marker expression, protein, and miRNA cargo involved. Recent studies have shown that exosomes can be used as therapeutic vectors for tissue regeneration and as alternative vectors for drug delivery. In this review, we summarize the physiological and biochemical characteristics, isolation, and identification of exosomes. Moreover, we focus on the role of exosomes in thrombosis, specifically venous thromboembolism, and their potential clinical applications, including as biomarkers and therapeutic vectors for thrombosis.
Collapse
Affiliation(s)
- Sheng-Lin Ye
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Wen-Dong Li
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Wei-Xiao Li
- Department of Vascular Surgery, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Lun Xiao
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Feng Ran
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Meng-Meng Chen
- School of Electronic Engineering, Nanjing Xiaozhuang University, Nanjing, China
| | - Xiao-Qiang Li
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Li-Li Sun
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| |
Collapse
|
3
|
Stępień EŁ, Rząca C, Moskal P. Novel biomarker and drug delivery systems for theranostics – extracellular vesicles. BIO-ALGORITHMS AND MED-SYSTEMS 2021. [DOI: 10.1515/bams-2021-0183] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Abstract
Extracellular vesicles (EVs) are nano- and micro-sized double-layered membrane entities derived from most cell types and released into biological fluids. Biological properties (cell-uptake, biocompatibility), and chemical (composition, structure) or physical (size, density) characteristics make EVs a good candidate for drug delivery systems (DDS). Recent advances in the field of EVs (e.g., scaling-up production, purification) and developments of new imaging methods (total-body positron emission tomography [PET]) revealed benefits of radiolabeled EVs in diagnostic and interventional medicine as a potential DDs in theranostics.
Collapse
Affiliation(s)
- Ewa Ł. Stępień
- M. Smoluchowski Institute of Physics, Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University , Krakow , Poland
- Total-Body Jagiellonian-PET Laboratory, Jagiellonian University , Kraków , Poland
- Theranostics Center, Jagiellonian University , Kraków , Poland
| | - Carina Rząca
- M. Smoluchowski Institute of Physics, Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University , Krakow , Poland
- Theranostics Center, Jagiellonian University , Kraków , Poland
| | - Paweł Moskal
- M. Smoluchowski Institute of Physics, Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University , Krakow , Poland
- Total-Body Jagiellonian-PET Laboratory, Jagiellonian University , Kraków , Poland
- Theranostics Center, Jagiellonian University , Kraków , Poland
| |
Collapse
|
4
|
Mui L, Martin CM, Tschirhart BJ, Feng Q. Therapeutic Potential of Annexins in Sepsis and COVID-19. Front Pharmacol 2021; 12:735472. [PMID: 34566657 PMCID: PMC8458574 DOI: 10.3389/fphar.2021.735472] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 08/23/2021] [Indexed: 12/13/2022] Open
Abstract
Sepsis is a continuing problem in modern healthcare, with a relatively high prevalence, and a significant mortality rate worldwide. Currently, no specific anti-sepsis treatment exists despite decades of research on developing potential therapies. Annexins are molecules that show efficacy in preclinical models of sepsis but have not been investigated as a potential therapy in patients with sepsis. Human annexins play important roles in cell membrane dynamics, as well as mediation of systemic effects. Most notably, annexins are highly involved in anti-inflammatory processes, adaptive immunity, modulation of coagulation and fibrinolysis, as well as protective shielding of cells from phagocytosis. These discoveries led to the development of analogous peptides which mimic their physiological function, and investigation into the potential of using the annexins and their analogous peptides as therapeutic agents in conditions where inflammation and coagulation play a large role in the pathophysiology. In numerous studies, treatment with recombinant human annexins and annexin analogue peptides have consistently found positive outcomes in animal models of sepsis, myocardial infarction, and ischemia reperfusion injury. Annexins A1 and A5 improve organ function and reduce mortality in animal sepsis models, inhibit inflammatory processes, reduce inflammatory mediator release, and protect against ischemic injury. The mechanisms of action and demonstrated efficacy of annexins in animal models support development of annexins and their analogues for the treatment of sepsis. The effects of annexin A5 on inflammation and platelet activation may be particularly beneficial in disease caused by SARS-CoV-2 infection. Safety and efficacy of recombinant human annexin A5 are currently being studied in clinical trials in sepsis and severe COVID-19 patients.
Collapse
Affiliation(s)
- Louise Mui
- Division of Critical Care, Department of Medicine, Schulich School of Dentistry and Medicine, Western University, London, ON, Canada
| | - Claudio M Martin
- Division of Critical Care, Department of Medicine, Schulich School of Dentistry and Medicine, Western University, London, ON, Canada.,Lawson Health Research Institute, London Health Sciences Centre, London, ON, Canada
| | - Brent J Tschirhart
- Department of Physiology and Pharmacology, Schulich School of Dentistry and Medicine, Western University, London, ON, Canada
| | - Qingping Feng
- Lawson Health Research Institute, London Health Sciences Centre, London, ON, Canada.,Department of Physiology and Pharmacology, Schulich School of Dentistry and Medicine, Western University, London, ON, Canada
| |
Collapse
|
5
|
Zifkos K, Dubois C, Schäfer K. Extracellular Vesicles and Thrombosis: Update on the Clinical and Experimental Evidence. Int J Mol Sci 2021; 22:ijms22179317. [PMID: 34502228 PMCID: PMC8431093 DOI: 10.3390/ijms22179317] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 08/24/2021] [Accepted: 08/25/2021] [Indexed: 12/16/2022] Open
Abstract
Extracellular vesicles (EVs) compose a heterogenous group of membrane-derived particles, including exosomes, microvesicles and apoptotic bodies, which are released into the extracellular environment in response to proinflammatory or proapoptotic stimuli. From earlier studies suggesting that EV shedding constitutes a cellular clearance mechanism, it has become evident that EV formation, secretion and uptake represent important mechanisms of intercellular communication and exchange of a wide variety of molecules, with relevance in both physiological and pathological situations. The putative role of EVs in hemostasis and thrombosis is supported by clinical and experimental studies unraveling how these cell-derived structures affect clot formation (and resolution). From those studies, it has become clear that the prothrombotic effects of EVs are not restricted to the exposure of tissue factor (TF) and phosphatidylserines (PS), but also involve multiplication of procoagulant surfaces, cross-linking of different cellular players at the site of injury and transfer of activation signals to other cell types. Here, we summarize the existing and novel clinical and experimental evidence on the role and function of EVs during arterial and venous thrombus formation and how they may be used as biomarkers as well as therapeutic vectors.
Collapse
Affiliation(s)
- Konstantinos Zifkos
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, D-55131 Mainz, Germany;
| | - Christophe Dubois
- Aix Marseille University, INSERM 1263, Institut National de la Recherche pour l’Agriculture, l’alimentation et l’Environnement (INRAE) 1260, Center for CardioVascular and Nutrition Research (C2VN), F-13380 Marseille, France;
| | - Katrin Schäfer
- Department of Cardiology, Cardiology I, University Medical Center Mainz, D-55131 Mainz, Germany
- Correspondence:
| |
Collapse
|
6
|
Platelets Extracellular Vesicles as Regulators of Cancer Progression-An Updated Perspective. Int J Mol Sci 2020; 21:ijms21155195. [PMID: 32707975 PMCID: PMC7432409 DOI: 10.3390/ijms21155195] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 07/19/2020] [Accepted: 07/20/2020] [Indexed: 12/14/2022] Open
Abstract
Extracellular vesicles (EVs) are a diverse group of membrane-bound structures secreted in physiological and pathological conditions by prokaryotic and eukaryotic cells. Their role in cell-to-cell communications has been discussed for more than two decades. More attention is paid to assess the impact of EVs in cancer. Numerous papers showed EVs as tumorigenesis regulators, by transferring their cargo molecules (miRNA, DNA, protein, cytokines, receptors, etc.) among cancer cells and cells in the tumor microenvironment. During platelet activation or apoptosis, platelet extracellular vesicles (PEVs) are formed. PEVs present a highly heterogeneous EVs population and are the most abundant EVs group in the circulatory system. The reason for the PEVs heterogeneity are their maternal activators, which is reflected on PEVs size and cargo. As PLTs role in cancer development is well-known, and PEVs are the most numerous EVs in blood, their feasible impact on cancer growth is strongly discussed. PEVs crosstalk could promote proliferation, change tumor microenvironment, favor metastasis formation. In many cases these functions were linked to the transfer into recipient cells specific cargo molecules from PEVs. The article reviews the PEVs biogenesis, cargo molecules, and their impact on the cancer progression.
Collapse
|
7
|
Kerris EWJ, Hoptay C, Calderon T, Freishtat RJ. Platelets and platelet extracellular vesicles in hemostasis and sepsis. J Investig Med 2019; 68:813-820. [PMID: 31843956 DOI: 10.1136/jim-2019-001195] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/23/2019] [Indexed: 01/09/2023]
Abstract
Platelets, cell fragments traditionally thought of as important only for hemostasis, substantially and dynamically contribute to the immune system's response to infection. In addition, there is increasing evidence that externally active platelet entities, including platelet granules and platelet extracellular vesicles (PEVs), play a role not only in hemostasis, but also in inflammatory actions previously ascribed to platelets themselves. Given the functions of platelets and PEVs during inflammation and infection, their role in sepsis is being investigated. Sepsis is a condition marked by the dysregulation of the body's normal activation of the immune system in response to a pathogen. The mechanisms for controlling infection locally become detrimental to the host if they are applied systemically. Similar to cells traditionally ascribed to the immune system, including neutrophils, lymphocytes, and macrophages, platelets are instrumental in helping a host clear an infection, but are also implicated in the uncontrolled amplification of the immune response that leads to sepsis. Clearly, the function of platelets is more complicated than its simple structure and primary role in hemostasis initially suggest. This review provides an overview of platelet and platelet extracellular vesicle structure and function, highlighting the complex role platelets and PEVs play in the body in the context of infection and sepsis.
Collapse
Affiliation(s)
- Elizabeth W J Kerris
- Division of Critical Care Medicine, Children's National Hospital, Washington, DC, USA.,Center for Genetic Medicine Research, Children's National Hospital, Washington, DC, USA
| | - Claire Hoptay
- Center for Genetic Medicine Research, Children's National Hospital, Washington, DC, USA
| | - Thais Calderon
- Department of Medical Education, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Robert J Freishtat
- Center for Genetic Medicine Research, Children's National Hospital, Washington, DC, USA.,Division of Emergency Medicine, Children's National Hospital, Washington, DC, USA
| |
Collapse
|
8
|
Krajewska-Włodarczyk M, Owczarczyk-Saczonek A, Żuber Z, Wojtkiewicz M, Wojtkiewicz J. Role of Microparticles in the Pathogenesis of Inflammatory Joint Diseases. Int J Mol Sci 2019; 20:E5453. [PMID: 31683793 PMCID: PMC6862866 DOI: 10.3390/ijms20215453] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 10/29/2019] [Accepted: 10/30/2019] [Indexed: 12/13/2022] Open
Abstract
Rheumatoid arthritis (RA), juvenile idiopathic arthritis (JIA), ankylosing spondylitis (AS), and psoriatic arthritis (PsA) make up a group of chronic immune-mediated inflammatory diseases (IMIDs). The course of these diseases involves chronic inflammation of joints and enthesopathies, which can result in joint damage and disability. Microparticles (MPs) are a group of small spherical membranous vesicles. The structure and cellular origin of MPs, mechanisms that stimulate their secretion and the place of their production, determine their biological properties, which could become manifest in the pathogenesis of immune-mediated inflammatory diseases. Microparticles can stimulate synovitis with proinflammatory cytokines and chemokines. MPs may also contribute to the pathogenesis of rheumatic diseases by the formation of immune complexes and complement activation, pro-coagulation activity, activation of vascular endothelium cells, and stimulation of metalloproteinase production. It seems that in the future, microparticles can become a modern marker of disease activity, a response to treatment, and, possibly, they can be used in the prognosis of the course of arthritis. The knowledge of the complexity of MPs biology remains incomplete and it requires further comprehensive studies to explain how they affect the development of rheumatic diseases. This review focuses on the immunopathogenic and therapeutic role of MPs in chronic immune-mediated inflammatory joint diseases.
Collapse
Affiliation(s)
- Magdalena Krajewska-Włodarczyk
- Department of Rheumatology, Municipal Hospital in Olsztyn, 10-900 Olsztyn, Poland.
- Department of Internal Medicine, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-900 Olsztyn, Poland.
| | - Agnieszka Owczarczyk-Saczonek
- Department of Dermatology, Sexually Transmitted Diseases and Clinical Immunology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-900 Olsztyn, Poland.
| | - Zbigniew Żuber
- Department of Pediatrics, Faculty of Medicine and Health Sciences, Andrzej Frycz Modrzewski Kraków University, 30-705 Kraków, Poland.
| | - Maja Wojtkiewicz
- Faculty of Earth Sciences, Department of Geomatics and Cartography Nicolaus Copernicus University, 87-100 Torun, Poland.
| | - Joanna Wojtkiewicz
- Department of Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-900 Olsztyn, Poland.
| |
Collapse
|
9
|
Jing J, Sun Y. An α IIbβ 3- and phosphatidylserine (PS)-binding recombinant fusion protein promotes PS-dependent anticoagulation and integrin-dependent antithrombosis. J Biol Chem 2019; 294:6670-6684. [PMID: 30803987 DOI: 10.1074/jbc.ra118.006044] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 02/10/2019] [Indexed: 11/06/2022] Open
Abstract
Blood platelets are required for normal wound healing, but they are also involved in thrombotic diseases, which are usually managed with anticoagulant drugs. Here, using genetic engineering, we coupled the disintegrin protein echistatin, which specifically binds to the platelet integrin αIIbβ3 receptor, to annexin V, which binds platelet membrane-associated phosphatidylserine (PS), to create the bifunctional antithrombotic molecule recombinant echistatin-annexin V fusion protein (r-EchAV). Lipid binding and plasma coagulation studies revealed that r-EchAV dose-dependently binds PS and delays plasma clotting time. Moreover, r-EchAV inhibited ADP-induced platelet aggregation in a dose-dependent manner and exhibited potent antiplatelet aggregation effects. r-EchAV significantly prolonged activated partial thromboplastin time, suggesting that it primarily affects the in vivo coagulation pathway. Flow cytometry results indicated that r-EchAV could effectively bind to the platelet αIIbβ3 receptor, indicating that r-EchAV retains echistatin's receptor-recognition region. In vivo experiments in mice disclosed that r-EchAV significantly prolongs bleeding time, indicating a significant anticoagulant effect in vivo resulting from the joint binding of r-EchAV to both PS and the αIIbβ3 receptor. We also report optimization of the r-EchAV production steps and its purification for high purity and yield. Our findings indicate that r-EchAV retains the active structural regions of echistatin and annexin V and that the whole molecule exhibits multitarget-binding ability arising from the dual functions of echistatin and annexin V. Therefore, r-EchAV represents a new class of anticoagulant that specifically targets the anionic membrane-associated coagulation enzyme complexes at thrombogenesis sites and may be a potentially useful antithrombotic agent.
Collapse
Affiliation(s)
- Jian Jing
- From the Beijing Key Laboratory of Genetic Engineering and Biotechnology, College of Life Sciences, Beijing Normal University, Xinwai St. 19, Haidian District, Beijing 100875, China
| | - Yanna Sun
- From the Beijing Key Laboratory of Genetic Engineering and Biotechnology, College of Life Sciences, Beijing Normal University, Xinwai St. 19, Haidian District, Beijing 100875, China
| |
Collapse
|
10
|
Didelot M, Docq C, Wahl D, Lacolley P, Regnault V, Lagrange J. Platelet aggregation impacts thrombin generation assessed by calibrated automated thrombography. Platelets 2017; 29:156-161. [PMID: 29022492 DOI: 10.1080/09537104.2017.1356452] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
A calibrated automated thrombogram (CAT) is performed usually with human platelet-free plasma (PFP) but may be more relevant with platelet-rich plasma (PRP). In this case, platelets are not stimulated by subendothelial molecules like collagen. Our aim was to assess the consequence of strong (collagen) or weak (ADP) induction of platelet release and aggregation on thrombin generation. Platelet aggregation in PRP was triggered with 10 µg/mL collagen or 10 µM ADP using a lumi-aggregometer. Thrombin generation curves were monitored by CAT in different conditions: PRP, PRP with activated platelets (actPRP), aggregated PRP (agPRP), aggregated platelets resuspended in autologous PFP (resPRP), PFP and PFP obtained after aggregation (agPFP). We found a 3-fold shortening of the lag time and time to peak and a marked increase in velocity and thrombin peak without changes in endogenous thrombin potential (ETP) in agPRP with both agonists compared with PRP. The same holds true in agPFP but with a marked increase in ETP compared with PFP. Similar changes in the kinetics of thrombin generation were observed with actPRP-collagen and to a lesser extent in resPRP-collagen compared with PRP. By contrast, there were no modifications of the thrombin generation curves in actPRP-ADP. Alpha-2-macroglobin-thrombin complexes were unchanged in the different PRP conditions but were increased in PFP prepared from agPFP compared to control PFP. Platelet aggregation during activation by agonists other than thrombin did not increase thrombin generation but accelerated its kinetics mainly via platelet content release and platelet-derived extracellular vesicules formation. In diseases characterized by altered platelet granule content or release as well as altered platelet activation, a platelet aggregation step prior to CAT analysis may be clinically relevant to improve laboratory estimation of the bleeding/thrombotic balance.
Collapse
Affiliation(s)
- Mélusine Didelot
- a INSERM, U1116 , Faculté de Médecine , Vandoeuvre-les-Nancy , France.,b Université de Lorraine , Nancy , France
| | - Clémence Docq
- a INSERM, U1116 , Faculté de Médecine , Vandoeuvre-les-Nancy , France.,b Université de Lorraine , Nancy , France
| | - Denis Wahl
- a INSERM, U1116 , Faculté de Médecine , Vandoeuvre-les-Nancy , France.,b Université de Lorraine , Nancy , France.,c CHRU Nancy , Vandœuvre-lès-Nancy , France
| | - Patrick Lacolley
- a INSERM, U1116 , Faculté de Médecine , Vandoeuvre-les-Nancy , France.,b Université de Lorraine , Nancy , France.,c CHRU Nancy , Vandœuvre-lès-Nancy , France
| | - Véronique Regnault
- a INSERM, U1116 , Faculté de Médecine , Vandoeuvre-les-Nancy , France.,b Université de Lorraine , Nancy , France.,c CHRU Nancy , Vandœuvre-lès-Nancy , France
| | - Jérémy Lagrange
- a INSERM, U1116 , Faculté de Médecine , Vandoeuvre-les-Nancy , France.,b Université de Lorraine , Nancy , France.,d Center for Thrombosis and Hemostasis , University Medical Center Mainz , Mainz , Germany
| |
Collapse
|
11
|
Brisson AR, Tan S, Linares R, Gounou C, Arraud N. Extracellular vesicles from activated platelets: a semiquantitative cryo-electron microscopy and immuno-gold labeling study. Platelets 2017; 28:263-271. [DOI: 10.1080/09537104.2016.1268255] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Alain R. Brisson
- Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS-University of Bordeaux-IPB, Allée Geoffroy Saint-Hilaire, Pessac, France
| | - Sisareuth Tan
- Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS-University of Bordeaux-IPB, Allée Geoffroy Saint-Hilaire, Pessac, France
| | - Romain Linares
- Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS-University of Bordeaux-IPB, Allée Geoffroy Saint-Hilaire, Pessac, France
| | - Céline Gounou
- Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS-University of Bordeaux-IPB, Allée Geoffroy Saint-Hilaire, Pessac, France
| | - Nicolas Arraud
- Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS-University of Bordeaux-IPB, Allée Geoffroy Saint-Hilaire, Pessac, France
| |
Collapse
|
12
|
Katsura K, Nomura S, Gui Lan Xie, Ohtani T, Ishida T, Kagawa H, Kitada C, Yamanaka Y, Kitajima H, Fukuhara S. Platelet Procoagulant Activity During,Peripheral Blood Stem Cell Harvest. Clin Appl Thromb Hemost 2016. [DOI: 10.1177/107602969700300210] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
We used flow cytometry to measure platelet-derived microparticle levels in plasma obtained from 16 patients during peripheral blood stem cell harvest (PBSC) and in platelet concentrates prepared by apheresis from 10 normal controls. We also studied the binding of an anti-P-selectin antibody and annexin-V to platelets. When all 60 harvests were assessed, we noted a significant difference in microparticle levels between patients with a platelet count >10 x 104/μl and those with a platelet count <10 X 104/μl (12.3 ± 4.4 vs. 75 ± 3.9%). In both the first and total harvests, the percentage of platelets and microparticles positive for anti-P-selectin and annexin-V were significantly higher than the normal control levels. These results suggest that patients undergoing mobilization by granulocyte colony-stimulating factor (G-CSF) who have a platelet count >10 X 104/μl are at risk of increased procoagulant activity after retransfusion following PBSC harvest. Key Words: Platelet-derived microparticle— Peripheral blood stem cell harvest—Granulocyte colony-stimulating factor.
Collapse
Affiliation(s)
- Kaoruko Katsura
- First Department of Internal Medicine, Kansai Medical University, Osaka, Japan
| | - Shosaku Nomura
- First Department of Internal Medicine, Kansai Medical University, Osaka, Japan, Department of Blood Transfusion, Kansai Medical University, Osaka, Japan
| | - Gui Lan Xie
- First Department of Internal Medicine, Kansai Medical University, Osaka, Japan
| | - Tetsuji Ohtani
- Department of Blood Transfusion, Kansai Medical University, Osaka, Japan
| | - Tomoko Ishida
- Department of Blood Transfusion, Kansai Medical University, Osaka, Japan
| | - Hideo Kagawa
- First Department of Internal Medicine, Kansai Medical University, Osaka, Japan
| | - Chikaho Kitada
- First Department of Internal Medicine, Kansai Medical University, Osaka, Japan
| | - Yoshitaka Yamanaka
- First Department of Internal Medicine, Kansai Medical University, Osaka, Japan
| | - Hiroyuki Kitajima
- First Department of Internal Medicine, Kansai Medical University, Osaka, Japan
| | - Shirou Fukuhara
- First Department of Internal Medicine, Kansai Medical University, Osaka, Japan, Department of Blood Transfusion, Kansai Medical University, Osaka, Japan
| |
Collapse
|
13
|
Krailadsiri P, Seghatchian J, Bode AP. State-of-the-Art-Review: Microvesicles in Blood Components: Laboratory and Clinical Aspects. Clin Appl Thromb Hemost 2016. [DOI: 10.1177/107602969700300203] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
There is ample evidence for the presence of microvesicles (MV) of different sizes and functions in various blood components. A variety of mechanisms have been proposed for the formation of MV. These include mechanical injury, shear stress, cell activation, activation of complements, hypoxia, and the cell aging process. While MV share many biological properties and surface receptors of their parental cells, they demonstrate significant differences in membrane asymmetry of the inner membrane phospholipid, in particular phosphatidylserine (PS). This provides high-affinity binding sites for the components of the prothrombinase complex. To what extent these MV contribute to hemostatic effectiveness, immudomodulation, and some untoward effects of the transfused blood components remains to be fully elucidated. Several methods for qualitative and semiquantitative characterization of MV are now available. Although in most cases it is necessary to separate MV from the intact cells for improved characterization, recent advances in flow cytometry make it possible to accurately differentiate MV in the presence of their parental cells on the basis of light scattering and fluorescent intensity. This review focuses on four main areas of MV in blood components: (1) the proposed mechanisms of platelet vesiculation, (2) factors influencing the formation of MV, (3) laboratory analysis of MV, and (4) the clinical impact of the presence of MV in blood components. Key Words: Microvesicte—Vesicutation—Biood component—Ptatelets—Transfusion.
Collapse
Affiliation(s)
| | | | - Arthur P. Bode
- Department of Pathology and Laboratory Medicine East Carolina University School of Medicine, Greenville, North Carolina, U.S.A
| |
Collapse
|
14
|
Chandler WL. Measurement of microvesicle levels in human blood using flow cytometry. CYTOMETRY PART B-CLINICAL CYTOMETRY 2016; 90:326-36. [PMID: 26606416 DOI: 10.1002/cyto.b.21343] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Revised: 11/04/2015] [Accepted: 11/19/2015] [Indexed: 11/08/2022]
Abstract
Microvesicles are fragments of cells released when the cells are activated, injured, or apoptotic. Analysis of microvesicle levels in blood has the potential to shed new light on the pathophysiology of many diseases. Flow cytometry is currently the only method that can simultaneously separate true lipid microvesicles from other microparticles in blood, determine the cell of origin and other microvesicle characteristics, and handle large numbers of clinical samples with a reasonable effort, but expanded use of flow cytometric measurement of microvesicle levels as a clinical and research tool requires improved, standardized assays. The goal of this review is to aid investigators in applying current best practices to microvesicle measurements. First pre-analytical factors are evaluated and data summarized for anticoagulant effects, sample transport and centrifugation. Next flow cytometer optimization is reviewed including interference from background in buffers and reagents, accurate microvesicle counting, swarm interference, and other types of coincidence errors, size calibration, and detection limits using light scattering, impedance and fluorescence. Finally current progress on method standardization is discussed and a summary of current best practices provided. © 2016 Clinical Cytometry Society.
Collapse
Affiliation(s)
- Wayne L Chandler
- Department of Laboratories, Seattle Children's Hospital, and Department of Laboratory Medicine, University of Washington, Seattle, Washington
| |
Collapse
|
15
|
Pichler Hefti J, Leichtle A, Stutz M, Hefti U, Geiser T, Huber AR, Merz TM. Increased endothelial microparticles and oxidative stress at extreme altitude. Eur J Appl Physiol 2016; 116:739-48. [DOI: 10.1007/s00421-015-3309-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 12/08/2015] [Indexed: 02/04/2023]
|
16
|
Immuno-analysis of microparticles: probing at the limits of detection. Sci Rep 2015; 5:16314. [PMID: 26553743 PMCID: PMC4639787 DOI: 10.1038/srep16314] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 10/07/2015] [Indexed: 12/20/2022] Open
Abstract
Microparticle (MP) research is clouded by debate regarding the accuracy and validity of flow cytometry (FCM) as an analytical methodology, as it is influenced by many variables including the pre-analytical conditions, instruments physical capabilities and detection parameters. This study utilises a simplistic in vitro system for generating MP, and through comparative analysis with immuno-electron microscopy (Immuno-EM) assesses the strengths and limitations of probe selection and high-sensitivity FCM. Of the markers examined, MP were most specifically labelled with phosphatidylserine ligands, annexin V and lactadherin, although only ~60% MP are PS positive. Whilst these two ligands detect comparable absolute MP numbers, they interact with the same population in distinct manners; annexin V binding is enhanced on TNF induced MP. CD105 and CD54 expression were, as expected, consistent and enhanced following TNF activation respectively. Their labelling however accounted for as few as 30-40% of MP. The greatest discrepancies between FCM and I-EM were observed in the population solely labelled for the surface antigen. These findings demonstrate that despite significant improvements in resolution, high-sensitivity FCM remains limited in detecting small-size MP expressing low antigen levels. This study highlights factors to consider when selecting endothelial MP probes, as well as interpreting and representing data.
Collapse
|
17
|
Banz Y, Item GM, Vogt A, Rieben R, Candinas D, Beldi G. Endothelial- and Platelet-Derived Microparticles Are Generated During Liver Resection in Humans. J INVEST SURG 2015; 29:20-31. [DOI: 10.3109/08941939.2015.1047540] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
18
|
Abstract
PURPOSE OF REVIEW Several decades of work by many investigators have elucidated the major signaling pathways responsible for platelet activation. Still to be fully understood is how these pathways are integrated into a single network and how changing conditions within a growing thrombus affect that network. In this review we will consider some of the recent studies that address these issues and describe a model that provides insights into platelet activation as it occurs in vivo. RECENT FINDINGS Genetic and pharmacologic studies performed in vivo have demonstrated that platelet activation during hemostasis and thrombosis is heterogeneous. Those studies indicate that distinct platelet activation pathways are not merely redundant, but are coordinated in time and space to achieve an optimal response. This coordination is achieved at least in part by the evolving distribution of platelet agonists and changes in solute transport within a hemostatic plug. SUMMARY Studies examining the coordination of platelet signaling in time and space continue to increase our understanding of hemostasis and thrombosis. In addition to helping to decipher platelet biology, the results have implications for the understanding of new and existing antiplatelet agents and their potential risks.
Collapse
|
19
|
Attenuating a sickle cell crisis with annexin V. Med Hypotheses 2015; 84:434-6. [PMID: 25665862 DOI: 10.1016/j.mehy.2015.01.037] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 01/22/2015] [Accepted: 01/23/2015] [Indexed: 01/29/2023]
Abstract
A sickle cell crisis is a painful and dangerous condition that defies effective treatment but fortunately it usually terminates spontaneously and patients spend far more time crisis free than in its painful throes. This suggests that an unstable physiologic balance exists between steady state sickle cell disease (SCD) and the crisis state and if this is so a therapeutic nudge during a crisis may help to terminate it. Annexin V may be able to provide this push. The phosphatidylserine (PS) molecules normally appear on the surface of senescent erythrocytes where they are recognized by macrophages and rapidly removed so that normally only about 1% are present in the circulation but in SCD 30-40% are prematurely senescent and their removal is delayed. The PS+ sickle erythrocytes remaining in the circulation adhere to the endothelium and their exposed PS acts as a platform for the initiation of the coagulation cascade that is responsible for clot propagation. Annexin V's great affinity for PS allows it to bond to it forming a shield that blocks both of these actions suggesting that its therapeutic administration during a sickle crisis may be able to hasten its termination.
Collapse
|
20
|
Murate M, Abe M, Kasahara K, Iwabuchi K, Umeda M, Kobayashi T. Transbilayer lipid distribution in nano scale. J Cell Sci 2015; 128:1627-38. [DOI: 10.1242/jcs.163105] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 02/02/2015] [Indexed: 01/19/2023] Open
Abstract
There are a limited number of methods to examine transbilayer lipid distribution in biomembranes. We employed freeze-fracture replica labelling immunoelectron microscopy in combination with multiple lipid-binding peptide/proteins to examine both transbilayer and lateral distribution of various phospholipids in mammalian cells. Our results indicate that phospholipids are exclusively distributed either in the outer or inner leaflet of human red blood cell (RBC) membranes. In contrast, in nucleated cells such as human skin fibroblasts and neutrophils, sphingomyelin was distributed in both leaflets while exhibiting characteristic lipid domains in the inner leaflet. Similar to RBC, lipid asymmetry was maintained both in resting and thrombin-activated platelets. However, the microparticles released from thrombin-activated platelets lost membrane asymmetry. Our results suggest that the microparticles were shed from platelet plasma membrane domains enriched with phosphatidylserine/phosphatidylinositol at the outer leaflet. These findings underscore the strict regulation and cell-type specificity of lipid asymmetry in the plasma membrane.
Collapse
|
21
|
Agonist-induced platelet procoagulant activity requires shear and a Rac1-dependent signaling mechanism. Blood 2014; 124:1957-67. [PMID: 25079357 DOI: 10.1182/blood-2014-03-560821] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Activated platelets facilitate blood coagulation by exposing phosphatidylserine (PS) and releasing microvesicles (MVs). However, the potent physiological agonists thrombin and collagen poorly induce PS exposure when a single agonist is used. To obtain a greater procoagulant response, thrombin is commonly used in combination with glycoprotein VI agonists. However, even under these conditions, only a percentage of platelets express procoagulant activity. To date, it remains unclear why platelets poorly expose PS even when stimulated with multiple agonists and what the signaling pathways are of soluble agonist-induced platelet procoagulant activity. Here we show that physiological levels of shear present in blood significantly enhance agonist-induced platelet PS exposure and MV release, enabling low doses of a single agonist to induce full-scale platelet procoagulant activity. PS exposed on the platelet surface was immediately released as MVs, revealing a tight coupling between the 2 processes under shear. Using platelet-specific Rac1(-/-) mice, we discovered that Rac1 plays a common role in mediating the low-dose agonist-induced procoagulant response independent of platelet aggregation, secretion, and the apoptosis pathway. Platelet-specific Rac1 function was not only important for coagulation in vitro but also for fibrin accumulation in vivo following laser-induced arteriolar injury.
Collapse
|
22
|
Gourvas V, Soulitzis N, Konstantinidou A, Dalpa E, Koukoura O, Koutroulakis D, Spandidos DA, Sifakis S. Reduced ANXA5 mRNA and protein expression in pregnancies complicated by preeclampsia. Thromb Res 2013; 133:495-500. [PMID: 24393658 DOI: 10.1016/j.thromres.2013.12.027] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Revised: 12/16/2013] [Accepted: 12/19/2013] [Indexed: 12/20/2022]
Abstract
INTRODUCTION The placental anticoagulant protein Annexin A5 (ANXA5) is a multifunctional protein that is highly expressed on the apical surfaces of syncytiotrophoblasts, and plays an important role in haemostatic regulations, maintaining blood fluidity of the placenta. The aim of this study was to investigate the expression of ANXA5 in pregnancies complicated by preeclampsia (PE). MATERIALS AND METHODS Placental tissue samples were collected from 23 pregnancies with PE and 34 normal pregnancies. ANXA5 mRNA levels were measured by quantitative Real-Time PCR (qPCR), while ANXA5 protein expression was measured by Western Blot (WB) and immunohistochemistry. RESULTS ANXA5 mRNA expression in PE samples was lower than 1% of its expression in normal samples (mean ± SD: 0.002 ± 0.004 vs. 0.55 ± 0.38, p < 0.001), while ANXA5 protein levels in PE samples were approximately at 65% of the average normal expression (mean ± SD: 0.53 ± 0.30 vs. 0.81 ± 0.25, p=0.001). Immunohistochemical analysis also verified the above results, since PE placentas tended to have low labelling indexes (LIs), in contrast to controls which demonstrated high LIs (p=0.020). Statistical analysis of the WB data revealed that ANXA5 protein expression was increased in PE smokers vs. PE non-smokers (mean ± SD: 0.64 ± 0.23 vs. 0.41 ± 0.33, p=0.027). CONCLUSIONS These results suggest that ANXA5 downregulation could be part of the pathophysiology of PE and the possible impairment in coagulation processes, which are seen in pregnancies that demonstrate PE. Further studies may investigate whether ANXA5 could be used as a biomarker for the early detection of PE and for the prediction of its severity.
Collapse
Affiliation(s)
- Victor Gourvas
- Laboratory of Clinical Virology, Medical School, University of Crete, Heraklion, Crete, Greece
| | - Nikolaos Soulitzis
- Laboratory of Clinical Virology, Medical School, University of Crete, Heraklion, Crete, Greece
| | | | - Efterpi Dalpa
- Laboratory of Clinical Virology, Medical School, University of Crete, Heraklion, Crete, Greece
| | - Ourania Koukoura
- Department of Obstetrics and Gynaecology, University Hospital of Heraklion, Crete, Greece
| | - Demetrios Koutroulakis
- Department of Obstetrics and Gynaecology, University Hospital of Heraklion, Crete, Greece
| | - Demetrios A Spandidos
- Laboratory of Clinical Virology, Medical School, University of Crete, Heraklion, Crete, Greece
| | - Stavros Sifakis
- Department of Obstetrics and Gynaecology, University Hospital of Heraklion, Crete, Greece.
| |
Collapse
|
23
|
Rho associated coiled-coil kinase-1 regulates collagen-induced phosphatidylserine exposure in platelets. PLoS One 2013; 8:e84649. [PMID: 24358370 PMCID: PMC3865301 DOI: 10.1371/journal.pone.0084649] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Accepted: 11/26/2013] [Indexed: 01/08/2023] Open
Abstract
Background The transbilayer movement of phosphatidylserine mediates the platelet procoagulant activity during collagen stimulation. The Rho-associated coiled-coil kinase (ROCK) inhibitor Y-27632 inhibits senescence induced but not activation induced phosphatidylserine exposure. To investigate further the specific mechanisms, we now utilized mice with genetic deletion of the ROCK1 isoform. Methods and Results ROCK1-deficient mouse platelets expose significantly more phosphatidylserine and generate more thrombin upon activation with collagen compared to wild-type platelets. There were no significant defects in platelet shape change, aggregation, or calcium response compared to wild-type platelets. Collagen-stimulated ROCK1-deficient platelets also displayed decreased phosphorylation levels of Lim Kinase-1 and cofilin-1. However, there was no reduction in phosphorylation levels of myosin phosphatase subunit-1 (MYPT1) or myosin light chain (MLC). In an invivo light/dye-induced endothelial injury/thrombosis model, ROCK1-deficient mice presented a shorter occlusion time in cremasteric venules when compared to wild-type littermates (3.16 ± 1.33 min versus 6.6 ± 2.6 min; p = 0.01). Conclusions These studies define ROCK1 as a new regulator for collagen-induced phosphatidylserine exposure in platelets with functional consequences on thrombosis. This effect was downstream of calcium signaling and was mediated by Lim Kinase-1 / cofilin-1-induced cytoskeletal changes.
Collapse
|
24
|
Cloutier N, Tan S, Boudreau LH, Cramb C, Subbaiah R, Lahey L, Albert A, Shnayder R, Gobezie R, Nigrovic PA, Farndale RW, Robinson WH, Brisson A, Lee DM, Boilard E. The exposure of autoantigens by microparticles underlies the formation of potent inflammatory components: the microparticle-associated immune complexes. EMBO Mol Med 2012; 5:235-49. [PMID: 23165896 PMCID: PMC3569640 DOI: 10.1002/emmm.201201846] [Citation(s) in RCA: 198] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Revised: 10/17/2012] [Accepted: 11/05/2012] [Indexed: 01/27/2023] Open
Abstract
Immunoglobulins, antigens and complement can assemble to form immune complexes (IC). ICs can be detrimental as they propagate inflammation in autoimmune diseases. Like ICs, submicron extracellular vesicles termed microparticles (MP) are present in the synovial fluid from patients affected with autoimmune arthritis. We examined MPs in rheumatoid arthritis (RA) using high sensitivity flow cytometry and electron microscopy. We find that the MPs in RA synovial fluid are highly heterogeneous in size. The observed larger MPs were in fact MP-containing ICs (mpICs) and account for the majority of the detectable ICs. These mpICs frequently express the integrin CD41, consistent with platelet origin. Despite expression of the Fc receptor FcγRIIa by platelet-derived MPs, we find that the mpICs form independently of this receptor. Rather, mpICs display autoantigens vimentin and fibrinogen, and recognition of these targets by anti-citrullinated peptide antibodies contributes to the production of mpICs. Functionally, platelet mpICs are highly pro-inflammatory, eliciting leukotriene production by neutrophils. Taken together, our data suggest a unique role for platelet MPs as autoantigen-expressing elements capable of perpetuating formation of inflammatory ICs.
Collapse
Affiliation(s)
- Nathalie Cloutier
- Faculté de Médecine de l'Université Laval, Centre de Recherche en Rhumatologie et Immunologie, Centre de Recherche du Centre Hospitalier Universitaire de Québec, Québec, Québec, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
|
26
|
Farag YMK, Keithy-Reddy SR, Mittal BV, Bansal V, Fareed J, Singh AK. Modulation of platelet activation in chronic kidney disease patients on erythropoiesis-stimulating agents. Clin Appl Thromb Hemost 2012; 18:453-61. [PMID: 22496088 DOI: 10.1177/1076029611431954] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Clinical trials demonstrate either no benefit or increased risk of cardiovascular events and mortality in patients with chronic kidney disease (CKD) targeted for higher hemoglobin levels, who are treated with erythropoiesis-stimulating agents (ESAs). The mechanism underlying this observation remains unexplained. METHODS AND RESULTS We assessed platelet activation by measuring soluble P-selectin (sPsel), CD40 ligand (CD40L), and circulating microparticles (CMP) in patients with CKD. Higher hemoglobin levels were associated with increased Psel levels in patients on ESAs but not in ESA-naïve anemic and nonanemic patients. Psel positively correlated with CMP and CD40L in both anemic and nonanemic patients. Multivariate linear regression analysis revealed an association between increased Psel levels and hemoglobin concentration in patients receiving ESAs. CONCLUSIONS Anemic CKD patients on ESAs demonstrate increased levels of markers of platelet activation. These observations suggest a potentially complex interplay between platelet activation, impaired kidney function, and treatment of CKD anemia with ESAs.
Collapse
Affiliation(s)
- Youssef M K Farag
- Renal Division, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | |
Collapse
|
27
|
Rank A, Nieuwland R, Delker R, Pihusch V, Wilkowski R, Toth B, Kolb HJ, Pihusch R. Surveillance of megakaryocytic function by measurement of CD61-exposing microparticles in allogeneic hematopoietic stem cell recipients. Clin Transplant 2011; 25:E233-42. [PMID: 21303416 DOI: 10.1111/j.1399-0012.2011.01406.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Increasing evidence suggests that circulating microparticles (MP) exposing CD61 originate predominantly from megakaryocytes. Dramatic changes in megakaryocytic homeostasis are regularly observed following allogeneic hematopoietic stem cell transplantation (HSCT) and associated with transplantation-associated complications. We studied MP plasma levels prospectively in healthy subjects (n = 10) and allogeneic HSCT recipients (n = 19) twice weekly from the start of conditioning therapy up to day 30. A total of 224 measurement points were evaluated. MP were isolated, double-stained with annexin V and anti-CD61, and analyzed by flow cytometry. In uncomplicated HSCT, we found a correlation between platelet and CD61-exposing MP count, which resulted in a constant ratio of MP per platelet. The ratio was increased in patients with active hematological malignancies before transplantation and normalized during conditioning therapy. After take, the MP ratio increased, whereas infections and microangiopathic hemolytic anemia did not affect the ratio. In patients with GvHD, a decreased MP ratio was observed depending on the grade of GvHD, possibly indicating megakaryocytic damage. The MP ratio was able to discriminate between toxic, septic, and GvHD-induced hyperbilirubinemia. We first describe CD61+ MP levels during allogeneic HSCT and postulate that the MP ratio might be a useful biomarker for the surveillance of megakaryocytes during HSCT.
Collapse
Affiliation(s)
- Andreas Rank
- Medizinische Klinik III - Großhadern, Klinikum der Ludwig Maximilians-Universität München, München, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Kim JE, Han M, Hanl KS, Kim HK. Vitamin E inhibition on platelet procoagulant activity: involvement of aminophospholipid translocase activity. Thromb Res 2011; 127:435-42. [PMID: 21296386 DOI: 10.1016/j.thromres.2011.01.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2010] [Revised: 12/22/2010] [Accepted: 01/13/2011] [Indexed: 10/18/2022]
Abstract
BACKGROUND Activated platelets provide an important procoagulant surface, because exposed negatively charged phosphatidylserine (PS) is an important cofactor of the coagulation cascade. Aminophospholipid translocase (APLT) can transport PS from the outer to the inner membrane leaflet. Although vitamin E has been investigated for its anti-aggregating effect on platelets, its effect on platelet procoagulant activity has not been reported. METHODS Phorbol 12-myristate 13-acetate (PMA), a well-known PKC activator, and thrombin were used to induce PS exposure on platelet surface. The expression of PS was measured by annexin A5 binding with flow cytometry. Platelet procoagulant activity was measured by a prothrombinase assay. APLT activity was measured by flow cytometry by determining the percent of 1-palmitoyl-2-[6-[(7-nitro-2-1,3-benzoxadiazol-4-yl)amino]caproyl]-sn-glycero-3-phosphatidylserine (NBD-PS) translocated from the outer to the inner membrane leaflet. Inhibition effects of vitamin E on platelet aggregation were simultaneously measured by a Multiplate aggregometer, a Chrono-log aggregometer, and a PFA-100 system. RESULTS Vitamin E significantly attenuated PMA-induced conformational change of glycoprotein IIb/IIIa and P-selectin expression. Vitamin E significantly inhibited PMA and thrombin-induced PS externalization and reduced prothrombinase activity on platelet surfaces both in vitro and ex vivo. APLT activity was increased by vitamin E in a dose-dependent manner, indicating that reduced procoagulant activity may be attributed, at least in part, to this increased APLT activity. Vitamin E inhibited platelet aggregation induced by combined chemokine SDF-1 and low-dose ADP as well as by usual doses of ADP or collagen when measured by the Multiplate and Chrono-log aggregometers but not when measured by PFA-100. CONCLUSIONS These in vitro and ex vivo results showed that vitamin E inhibited platelet PS exposure and procoagulant activity partly by increasing APLT activity. These actions of vitamin E on platelet function provide new insights into the anticoagulation properties of vitamin E.
Collapse
Affiliation(s)
- Ji-Eun Kim
- Department of Laboratory Medicine and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | | | | | | |
Collapse
|
29
|
Rank A, Nieuwland R, Liebhardt S, Iberer M, Grützner S, Toth B, Pihusch R. Apheresis platelet concentrates contain platelet-derived and endothelial cell-derived microparticles. Vox Sang 2011; 100:179-86. [DOI: 10.1111/j.1423-0410.2010.01385.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
30
|
Rank A, Nieuwland R, Delker R, Köhler A, Toth B, Pihusch V, Wilkowski R, Pihusch R. Cellular origin of platelet-derived microparticles in vivo. Thromb Res 2010; 126:e255-9. [DOI: 10.1016/j.thromres.2010.07.012] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2010] [Revised: 07/15/2010] [Accepted: 07/16/2010] [Indexed: 12/20/2022]
|
31
|
Dasgupta SK, Argaiz ER, Mercado JEC, Maul HOE, Garza J, Enriquez AB, Abdel-Monem H, Prakasam A, Andreeff M, Thiagarajan P. Platelet senescence and phosphatidylserine exposure. Transfusion 2010; 50:2167-75. [PMID: 20456701 PMCID: PMC2921562 DOI: 10.1111/j.1537-2995.2010.02676.x] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND The exposure of phosphatidylserine occurs during platelet (PLT) activation and during in vitro storage. Phosphatidylserine exposure also occurs during apoptosis after the release of mitochondrial cytochrome c. We have examined the role of cytochrome c release, mitochondrial membrane potential (ΔΨm), and cyclophilin D (CypD) in phosphatidylserine exposure due to activation and storage. STUDY DESIGN AND METHODS The exposure of phosphatidylserine and the loss of ΔΨm were determined in a flow cytometer using fluorescein isothiocyanate-lactadherin and JC-1, a lipophilic cationic reporter dye. The role of CypD was determined with cyclosporin A and CypD-deficient murine PLTs. Cytochrome c-induced caspase-3 and Rho-associated kinase I (ROCK1) activation were determined by immunoblotting and using their inhibitors. RESULTS Collagen- and thrombin-induced exposure of phosphatidylserine was accompanied by a decrease in ΔΨm. Cyclosporin A inhibited the phosphatidylserine exposure and the loss of ΔΨm. CypD(-/-) mice had decreased loss of ΔΨm and impaired phosphatidylserine exposure. Collagen and thrombin did not induce the release of cytochrome c nor the activation of caspase-3 and ROCK1. In contrast, in PLTs stored for more than 5 days, the phosphatidylserine exposure was associated with cytochrome c-induced caspase-3 and ROCK1 activation. ABT737, a BH3 mimetic that induces mitochondrial pathway of apoptosis, induced cytochrome c release and activation of caspase-3 and ROCK1 and phosphatidylserine exposure independent of CypD. CONCLUSION These results show that in stored PLTs cytochrome c release and the subsequent activation of caspase-3 and ROCK1 mediate phosphatidylserine exposure and it is distinct from activation-induced phosphatidylserine exposure.
Collapse
Affiliation(s)
- Swapan Kumar Dasgupta
- Michael E. DeBakey Veterans Affairs Medical Center, Department of Pathology, Baylor College of Medicine
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Gilio K, van Kruchten R, Braun A, Berna-Erro A, Feijge MAH, Stegner D, van der Meijden PEJ, Kuijpers MJE, Varga-Szabo D, Heemskerk JWM, Nieswandt B. Roles of platelet STIM1 and Orai1 in glycoprotein VI- and thrombin-dependent procoagulant activity and thrombus formation. J Biol Chem 2010; 285:23629-38. [PMID: 20519511 DOI: 10.1074/jbc.m110.108696] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
In platelets, STIM1 has been recognized as the key regulatory protein in store-operated Ca(2+) entry (SOCE) with Orai1 as principal Ca(2+) entry channel. Both proteins contribute to collagen-dependent arterial thrombosis in mice in vivo. It is unclear whether STIM2 is involved. A key platelet response relying on Ca(2+) entry is the surface exposure of phosphatidylserine (PS), which accomplishes platelet procoagulant activity. We studied this response in mouse platelets deficient in STIM1, STIM2, or Orai1. Upon high shear flow of blood over collagen, Stim1(-/-) and Orai1(-/-) platelets had greatly impaired glycoprotein (GP) VI-dependent Ca(2+) signals, and they were deficient in PS exposure and thrombus formation. In contrast, Stim2(-/-) platelets reacted normally. Upon blood flow in the presence of thrombin generation and coagulation, Ca(2+) signals of Stim1(-/-) and Orai1(-/-) platelets were partly reduced, whereas the PS exposure and formation of fibrin-rich thrombi were normalized. Washed Stim1(-/-) and Orai1(-/-) platelets were deficient in GPVI-induced PS exposure and prothrombinase activity, but not when thrombin was present as co-agonist. Markedly, SKF96365, a blocker of (receptor-operated) Ca(2+) entry, inhibited Ca(2+) and procoagulant responses even in Stim1(-/-) and Orai1(-/-) platelets. These data show for the first time that: (i) STIM1 and Orai1 jointly contribute to GPVI-induced SOCE, procoagulant activity, and thrombus formation; (ii) a compensating Ca(2+) entry pathway is effective in the additional presence of thrombin; (iii) platelets contain two mechanisms of Ca(2+) entry and PS exposure, only one relying on STIM1-Orai1 interaction.
Collapse
Affiliation(s)
- Karen Gilio
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6200 MD Maastricht, The Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Boilard E, Nigrovic PA, Larabee K, Watts GFM, Coblyn JS, Weinblatt ME, Massarotti EM, Remold-O'Donnell E, Farndale RW, Ware J, Lee DM. Platelets amplify inflammation in arthritis via collagen-dependent microparticle production. Science 2010; 327:580-3. [PMID: 20110505 DOI: 10.1126/science.1181928] [Citation(s) in RCA: 815] [Impact Index Per Article: 58.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In addition to their pivotal role in thrombosis and wound repair, platelets participate in inflammatory responses. We investigated the role of platelets in the autoimmune disease rheumatoid arthritis. We identified platelet microparticles--submicrometer vesicles elaborated by activated platelets--in joint fluid from patients with rheumatoid arthritis and other forms of inflammatory arthritis, but not in joint fluid from patients with osteoarthritis. Platelet microparticles were proinflammatory, eliciting cytokine responses from synovial fibroblasts via interleukin-1. Consistent with these findings, depletion of platelets attenuated murine inflammatory arthritis. Using both pharmacologic and genetic approaches, we identified the collagen receptor glycoprotein VI as a key trigger for platelet microparticle generation in arthritis pathophysiology. Thus, these findings demonstrate a previously unappreciated role for platelets and their activation-induced microparticles in inflammatory joint diseases.
Collapse
Affiliation(s)
- Eric Boilard
- Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Elevated platelet angiostatin and circulating endothelial microfragments in idiopathic pulmonary arterial hypertension: A preliminary study. Thromb Res 2010; 125:53-60. [DOI: 10.1016/j.thromres.2009.04.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2008] [Revised: 04/01/2009] [Accepted: 04/02/2009] [Indexed: 12/13/2022]
|
35
|
Sakalihasan N, Michel J. Functional Imaging of Atherosclerosis to Advance Vascular Biology. Eur J Vasc Endovasc Surg 2009; 37:728-34. [DOI: 10.1016/j.ejvs.2008.12.024] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2008] [Accepted: 12/30/2008] [Indexed: 12/20/2022]
|
36
|
Rouzet F, Sarda-Mantel L, Michel JB, Le Guludec D. Molecular imaging of platelet activation in thrombus. J Nucl Cardiol 2009; 16:277-86. [PMID: 19224152 DOI: 10.1007/s12350-009-9053-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2008] [Accepted: 01/06/2009] [Indexed: 11/24/2022]
Affiliation(s)
- François Rouzet
- Department of Nuclear Medicine, Bichat-Claude Bernard Hospital, AP-HP, Paris, France.
| | | | | | | |
Collapse
|
37
|
Mezzano D, Matus V, Sáez CG, Pereira J, Panes O. Tissue factor storage, synthesis and function in normal and activated human platelets. Thromb Res 2008; 122 Suppl 1:S31-6. [PMID: 18691497 DOI: 10.1016/s0049-3848(08)70016-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The source and significance of blood-borne tissue factor (TF) are controversial. The presence of TF in platelets was initially attributed to transfer of the protein from other cells (e.g., monocytes) and/or TF-bearing microparticles. Recently, TF-mRNA, neo-synthesis of the protein and TF-dependent procoagulant activity (PCA) have been reported in human platelets. The storage of "encrypted", potentially active TF in circulating, non-stimulated platelets remains debatable. One report strongly suggests that the starting of platelet PCA depends on de novo TF synthesis induced by platelet activation, whereas others provide persuasive evidence that platelets circulate with preformed TF, readily functional upon demand. These findings may have an impact on our current ideas of physiological hemostasis and thrombus formation. In fact, platelets would lead not only the formation of the primary plug, but in this microenvironment they would also contribute to the triggering of thrombin generation, fibrin deposition, clot consolidation and initial protection from fibrinolysis. Much research is needed to validate this platelet-based hemostasis model.
Collapse
Affiliation(s)
- Diego Mezzano
- Department of Hematology-Oncology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | | | | | | | | |
Collapse
|
38
|
Vassiliou AG, Fragoulis EG, Vassilacopoulou D. Detection, purification and identification of an endogenous inhibitor of L-Dopa decarboxylase activity from human placenta. Neurochem Res 2008; 34:1089-100. [PMID: 19005753 DOI: 10.1007/s11064-008-9879-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/23/2008] [Indexed: 01/01/2023]
Abstract
An endogenous inhibitor of L-Dopa decarboxylase activity was identified and purified from human placenta. The endogenous inhibitor of L-Dopa decarboxylase (Ddc) was localized in the membrane fraction of placental tissue. Treatment of membranes with phosphatidylinositol-specific phospholipase C or proteinase K did not affect membrane-associated Ddc inhibitory activity, suggesting that a population of the inhibitor is embedded within membranes. Purification was achieved by extraction from a nondenaturing polyacrylamide gel. The purification scheme resulted in the isolation of a single 35 kDa band, bearing L-Dopa decarboxylase inhibitory activity. The purified inhibitor was identified as Annexin V. The elucidation of the biological importance of the presence of an L-Dopa decarboxylase activity inhibitor in normal human tissues could provide us with new information leading to the better understanding of the biological pathways that Ddc is involved in.
Collapse
|
39
|
Hayashi T, Mogami H, Murakami Y, Nakamura T, Kanayama N, Konno H, Urano T. Real-time analysis of platelet aggregation and procoagulant activity during thrombus formation in vivo. Pflugers Arch 2008; 456:1239-51. [PMID: 18553102 DOI: 10.1007/s00424-008-0466-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2007] [Revised: 12/30/2007] [Accepted: 01/22/2008] [Indexed: 11/27/2022]
Abstract
The exact mechanism of blood vessel thrombus formation remains to be defined. Here, we introduce a new approach to probe thrombus formation in blood vessels of living animals using intravital microscopy in green fluorescent protein (GFP)-transgenic mice to simultaneously monitor platelet aggregation and procoagulant activity. To this end, GFP-expressing platelets and annexin A5 labeled with a fluorescent dye were employed to visualize and analyze platelet aggregation and markers of procoagulant activity (platelet surface phosphatidylserine (PS)). Laser-induced thrombi increased and then decreased in size with time in vessels of living animals, whereas platelet surface PS initiated at the site of injury and then penetrated into the thrombus. PS-positive platelets were predominantly localized in the center of the thrombus, as was fibrin generation. The experimental system proposed here is a valuable tool not only for investigating mechanisms of thrombus formation but also to assess the efficacy of antithrombotic drugs within the vasculature.
Collapse
Affiliation(s)
- Tadataka Hayashi
- Second Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | | | | | | | | | | | | |
Collapse
|
40
|
van Genderen HO, Kenis H, Hofstra L, Narula J, Reutelingsperger CPM. Extracellular annexin A5: functions of phosphatidylserine-binding and two-dimensional crystallization. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2008; 1783:953-63. [PMID: 18334229 DOI: 10.1016/j.bbamcr.2008.01.030] [Citation(s) in RCA: 153] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2007] [Revised: 01/22/2008] [Accepted: 01/23/2008] [Indexed: 02/06/2023]
Abstract
In normal healthy cells phosphatidylserine is located in the inner leaflet of the plasma membrane. However, on activated platelets, dying cells and under specific circumstances also on various types of viable leukocytes phosphatidylserine is actively externalized to the outer leaflet of the plasma membrane. Annexin A5 has the ability to bind in a calcium-dependent manner to phosphatidylserine and to form a membrane-bound two-dimensional crystal lattice. Based on these abilities various functions for extracellular annexin A5 on the phosphatidylserine-expressing plasma membrane have been proposed. In this review we describe possible mechanisms for externalization of annexin A5 and various processes in which extracellular annexin A5 may play a role such as blood coagulation, apoptosis, phagocytosis and formation of plasma membrane-derived microparticles. We further highlight the recent discovery of internalization of extracellular annexin A5 by phosphatidylserine-expressing cells.
Collapse
Affiliation(s)
- Hugo O van Genderen
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, University Maastricht, Maastricht, The Netherlands
| | | | | | | | | |
Collapse
|
41
|
Technetium 99m–Labeled Annexin V Scintigraphy of Platelet Activation in Vegetations of Experimental Endocarditis. Circulation 2008; 117:781-9. [DOI: 10.1161/circulationaha.107.718114] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background—
The pathophysiology of infective endocarditis involves a pathogen/host tissue interaction, leading to formation of infected thrombotic vegetations. Annexin V is a ligand of phosphatidylserines exposed by activated platelets and apoptotic cells. Because vegetations are platelet-fibrin clots in which platelet proaggregant activity is enhanced by bacterial colonization, we investigated the ability of annexin V labeled with technetium Tc 99m (
99m
Tc-ANX) to provide functional imaging of these vegetations in experimental models of infective endocarditis. This ability was assessed in rabbits and rats because of the different interest of these 2 species in preclinical analysis.
Methods and Results—
Nonbacterial thrombotic endocarditis was induced with the use of a catheter left indwelling through the aortic or tricuspid valve, and animals were injected with either a bacterial inoculum or saline. Scintigraphic investigations were performed 5 days later and showed a higher
99m
Tc-ANX uptake by vegetations in infected versus noninfected animals (ratio, 1.3 for in vivo acquisitions and 2 for autoradiography;
P
<0.0001 for all), whereas no significant uptake was present in controls. Right-sided endocarditis was associated with pulmonary uptake foci corresponding to emboli. Histological analysis of vegetations showed a specific uptake of
99m
Tc-ANX at the interface between circulating blood and vegetation. In parallel, underlying myocardial tissue showed myocyte apoptosis and mucoid degeneration, without extracellular matrix degradation at this stage.
Conclusions—
99m
Tc-ANX is suitable for functional imaging of platelet-fibrin vegetations in endocarditis, as well as embolic events.
99m
Tc-ANX uptake reflects mainly platelet activation in the luminal layer of vegetations. This uptake is enhanced by bacterial colonization.
Collapse
|
42
|
Nomura S, Ozaki Y, Ikeda Y. Function and role of microparticles in various clinical settings. Thromb Res 2008; 123:8-23. [DOI: 10.1016/j.thromres.2008.06.006] [Citation(s) in RCA: 154] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2008] [Revised: 05/16/2008] [Accepted: 06/09/2008] [Indexed: 12/18/2022]
|
43
|
Sahu SK, Gummadi SN, Manoj N, Aradhyam GK. Phospholipid scramblases: An overview. Arch Biochem Biophys 2007; 462:103-14. [PMID: 17481571 DOI: 10.1016/j.abb.2007.04.002] [Citation(s) in RCA: 140] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2007] [Revised: 03/30/2007] [Accepted: 04/01/2007] [Indexed: 12/23/2022]
Abstract
Phospholipid scramblases are a group of homologous proteins that are conserved in all eukaryotic organisms. They are believed to be involved in destroying plasma membrane phospholipid asymmetry at critical cellular events like cell activation, injury and apoptosis. However, a detailed mechanism of phospholipid scrambling still awaits a proper understanding. The most studied member of this family, phospholipid scramblase 1 (PLSCR1) (a 37kDa protein), is involved in rapid Ca2+ dependent transbilayer redistribution of plasma membrane phospholipids. Recently the function of PLSCR1 as a phospholipids translocator has been challenged and evidences suggest that PLSCR1 acts as signaling molecule. It has been shown to be involved in protein phosphorylation and as a potential activator of genes in response to interferon and other cytokines. Interferon induced rapid biosynthesis of PLSCR1 targets some of the protein into the nucleus, where it binds to the promoter region of inositol 1,4,5-triphosphate (IP3) receptor type 1 (IP3R1) gene and induces its expression. Palmitoylation of PLSCR1 acts as a switch, controlling its localization either to the PM or inside the nucleus. In the present review, we discuss the current understanding of PLSCR1 in relation to its trafficking, localization and signaling functions.
Collapse
Affiliation(s)
- Santosh Kumar Sahu
- Department of Biotechnology, Indian Institute of Technology Madras, Chennai, India
| | | | | | | |
Collapse
|
44
|
Chiou JF, Woon MD, Cheng SN, Hsu CH, Cherng SC, Hsieh FK, Lin SM, Shiau CY. Staphylokinase-annexin XI chimera exhibited efficient in vitro thrombolytic activities. Biosci Biotechnol Biochem 2007; 71:1122-9. [PMID: 17485856 DOI: 10.1271/bbb.60279] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Annexins (ANXs) are a family of calcium dependent phospholipid binding proteins. Phospholipids such as phosphatidylserine are rapidly exposed on the surfaces of injured endothelial cells, activated platelets, and apoptotic cells in a large number of disorders. In this study, annexin V and XI (ANXV and ANXXI) were individually fused to the C-terminal of staphylokinase (SAK), a fibrin-selective thrombolytic protein, to form chimeras for evaluation of their in-vitro thrombolytic activities. The two chimeras were found to have plasminogen activation activity of comparable efficiency. When the chimeras were challenged under higher concentrations of plasmin for 1 h, hydrolysis of them into moieties was not seen on SDS-PAGE. In two thrombolytic assays, SAK-ANXXI was found to resolve both platelet rich plasma (PRP) clots and platelet poor plasma (PPP) clots with an efficiency similar to that of SAK. However, SAK-ANXV showed significantly reduced efficiency. With regard to anticoagulation ability, SAK-ANXXI was also found to have a stronger effect on dose-dependent extension of clotting time among the four tested proteins. The unique long N-terminal tail of ANXXI, composed of 202 residues, in contrast to the 16 residues of ANXV, probably served successfully to dispatch two moieties to function properly in a complicated microenvironment. Hence, a new option other than the most committed ANXV for the ANX based chimera without elaboration of linker construction is presented.
Collapse
Affiliation(s)
- Jeng-Fong Chiou
- Cancer Center and Department of Radiation Oncology, Taipei Medical University Hospital, Taipei, Taiwan, ROC
| | | | | | | | | | | | | | | |
Collapse
|
45
|
|
46
|
Abstract
Microparticles (MP) derived from vascular endothelium or circulating blood cells circulate in the peripheral blood. They originate from blebbing and shedding from cell membrane surfaces in physiological and pathological conditions and are present in low concentrations in normal plasma. Increased levels are generated by a number of mechanisms including platelet activation, direct vascular endothelial damage, thrombin activity on the cell surface, C5b-9 activation, and PF4-heparin-antibody interaction. Several techniques are currently used to study the generation and nature of circulating microparticles. In particular, the genesis and role of microparticles, derived from platelets, endothelial cells and monocytes, in sepsis (especially meningococcal-induced), heparin-induced thrombocytopenia (HIT), thrombotic thrombocytopenic purpura (TTP), aplastic anaemia, paroxysmal nocturnal haemoglobinuria (PNH) and sickle cell disease (SCD) have been well studied, and provide important insights into the underlying diseases. A defect in the ability to form microparticles leads to the severe bleeding disorder of Scott syndrome, which in turn provides a revealing insight into the physiology of coagulation. In addition the complex role of microparticles in vascular and cardiovascular diseases is an area of immense interest, that promises to yield important advances into diagnosis and therapy.
Collapse
Affiliation(s)
- Andrea Piccin
- Irish Blood Transfusion Service, James's Street, Dublin 8, Ireland.
| | | | | |
Collapse
|
47
|
Schulz-Heik K, Ramachandran J, Bluestein D, Jesty J. The extent of platelet activation under shear depends on platelet count: differential expression of anionic phospholipid and factor Va. PATHOPHYSIOLOGY OF HAEMOSTASIS AND THROMBOSIS 2006; 34:255-62. [PMID: 16772736 DOI: 10.1159/000093104] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2005] [Accepted: 09/02/2005] [Indexed: 11/19/2022]
Abstract
It is widely accepted that shear stress activates platelets. However, this may have two linked but separate causes: a direct effect of shear stress on individual platelets, and secondary inter-platelet activation dependent on the release of agonists caused by shear. Gel-filtered platelets were exposed to intermittent low shear at 20,000 and 200,000 platelets/microl and their activation was measured with a prothrombinase-based assay. At the lower count, activation was slow and essentially linear, but at the higher count, it rose exponentially with time, leading to 3-fold more prothrombinase activity. Inclusion of apyrase and/or prostaglandin I(2) slightly reduced activation at high platelet counts, but did not abolish the nonlinear kinetics, and antibodies against von Willebrand factor had no significant effect. The contributions of anionic phospholipid and factor Va to the prothrombinase activity were assessed by measurements in the presence of exogenous factor Va. The results strongly suggest that anionic phospholipid appearance is caused directly by shear exposure, but that factor Va release from the alpha-granules is a secondary event and largely the result of platelet-platelet signaling.
Collapse
|
48
|
Dasgupta SK, Guchhait P, Thiagarajan P. Lactadherin binding and phosphatidylserine expression on cell surface-comparison with annexin A5. Transl Res 2006; 148:19-25. [PMID: 16887494 DOI: 10.1016/j.lab.2006.03.006] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2005] [Revised: 02/20/2006] [Accepted: 03/06/2006] [Indexed: 11/25/2022]
Abstract
BACKGROUND Transbilayer movement of anionic phospholipids from the inner to the outer leaflet of the plasma membrane occurs during platelet activation, red cell senescence, and apoptosis. The anionic phospholipid-binding protein, annexin A5, has been used to detect the presence of phosphatidylserine on the outer leaflet of the cell membrane. Lactadherin, a glycoprotein secreted by macrophages, binds to phosphatidylserine on apoptotic cells and promote their clearance by macrophages. METHODS The authors isolated and labeled lactadherin and annexin A5 with FITC and compared their ability to detect phosphatidylserine expression by flow cytometry. RESULTS FITC-lactadherin induced greater shift in the histogram and a higher mean fluorescence intensity than FITC-annexin A5 when platelets were activated with thrombin (0.1 unit/mL) or Ca(2+) ionophore A23187 (1 microM). Similarly, lactadherin was more sensitive in detecting phosphatidylserine in red cells induced to express phosphatidylserine. Also, in HL 60 cells undergoing apoptosis, lactadherin detected phosphatidylserine expression earlier than annexin A5. In patients with disseminated intravascular coagulation, lactadherin detected phosphatidylserine-expressing platelets in most patients, whereas under similar conditions, FITC-annexin A5 could not. CONCLUSIONS The authors' studies show that FITC-lactadherin is a better probe than annexin A5 in detecting phosphatidylserine-expressing activated platelets, red cells, and apoptotic cells.
Collapse
Affiliation(s)
- Swapan K Dasgupta
- Department of Pathology and Medicine (Thrombosis Research), Baylor College of Medicine, Houston, Texas, USA
| | | | | |
Collapse
|
49
|
Furie B, Furie BC. Cancer-associated thrombosis. Blood Cells Mol Dis 2006; 36:177-81. [PMID: 16490369 DOI: 10.1016/j.bcmd.2005.12.018] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2005] [Accepted: 12/07/2005] [Indexed: 12/22/2022]
Abstract
Thrombosis is one of the major complications of malignant disease, but the underlying molecular and cellular basis remains elusive. A number of hypotheses have been put forth, including the expression of tissue factor by tumor cells, release of tissue factor during cell death following chemotherapy, intrinsic unique tumor procoagulants and microparticles. Exploration of a potential role of microparticles in cancer-associated thrombosis indicates that tissue factor microparticles are present in a spectrum of cancer patients known to have a high incidence of thromboembolic complications.
Collapse
Affiliation(s)
- Bruce Furie
- Division of Hemostasis and Thrombosis, Center for Vascular Biology Research, Beth Israel Deaconess Medical Center and the Department of Medicine, Harvard Medical School, Boston, MA 02215, USA.
| | | |
Collapse
|
50
|
Abstract
Tissue factor (TF) encryption is the post-translational suppression of TF procoagulant activity (PCA) on the cell surface. There is emerging evidence of encrypted TF in normal blood associated with monocytes and platelets. Expression of this latent TF PCA during the propagation phase of blood coagulation may contribute to hemostasis. One pathway leading to the decryption of TF PCA begins with an increase in cytosolic calcium. A large calcium influx triggers both the exposure of phosphatidylserine and the expression of TF PCA on cell surfaces. The connections between these events are reviewed along with evidence that lipid raft association may also contribute to TF encryption. The last step in the decryption of TF PCA is the proteolytic activation of zymogen factor VII. This event may be a key to understanding the different roles of intravascular and extravascular TF in the process of blood coagulation.
Collapse
Affiliation(s)
- Ronald R Bach
- Minneapolis Veterans Affairs Medical Center, Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN, USA.
| |
Collapse
|