51
|
Dewitte A, Tanga A, Villeneuve J, Lepreux S, Ouattara A, Desmoulière A, Combe C, Ripoche J. New frontiers for platelet CD154. Exp Hematol Oncol 2015; 4:6. [PMID: 25763299 PMCID: PMC4355125 DOI: 10.1186/s40164-015-0001-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 02/03/2015] [Indexed: 02/07/2023] Open
Abstract
The role of platelets extends beyond hemostasis. The pivotal role of platelets in inflammation has shed new light on the natural history of conditions associated with acute or chronic inflammation. Beyond the preservation of vascular integrity, platelets are essential to tissue homeostasis and platelet-derived products are already used in the clinics. Unanticipated was the role of platelets in the adaptative immune response, allowing a renewed conceptual approach of auto-immune diseases. Platelets are also important players in cancer growth and dissemination. Platelets fulfill most of their functions through the expression of still incompletely characterized membrane-bound or soluble mediators. Among them, CD154 holds a peculiar position, as platelets represent a major source of CD154 and as CD154 contributes to most of these new platelet attributes. Here, we provide an overview of some of the new frontiers that the study of platelet CD154 is opening, in inflammation, tissue homeostasis, immune response, hematopoiesis and cancer.
Collapse
Affiliation(s)
- Antoine Dewitte
- INSERM U1026, and Université de Bordeaux, F-33000 Bordeaux, France ; Service d'Anesthésie-Réanimation II, CHU de Bordeaux, F-33600 Pessac, France
| | - Annabelle Tanga
- INSERM U1026, and Université de Bordeaux, F-33000 Bordeaux, France
| | - Julien Villeneuve
- Cell and Developmental Biology Programme, Centre for Genomic Regulation, 08003 Barcelona, Spain ; Department of Molecular and Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, CA 94720-3200 USA
| | | | - Alexandre Ouattara
- Service d'Anesthésie-Réanimation II, CHU de Bordeaux, F-33600 Pessac, France
| | | | - Christian Combe
- INSERM U1026, and Université de Bordeaux, F-33000 Bordeaux, France ; Service de Néphrologie Transplantation Dialyse, CHU de Bordeaux, F-33076 Bordeaux, France
| | - Jean Ripoche
- INSERM U1026, and Université de Bordeaux, F-33000 Bordeaux, France
| |
Collapse
|
52
|
Gros A, Ollivier V, Ho-Tin-Noé B. Platelets in inflammation: regulation of leukocyte activities and vascular repair. Front Immunol 2015; 5:678. [PMID: 25610439 PMCID: PMC4285099 DOI: 10.3389/fimmu.2014.00678] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 12/16/2014] [Indexed: 12/29/2022] Open
Abstract
There is now a large body of evidence that platelets are central actors of inflammatory reactions. Indeed, platelets play a significant role in a variety of inflammatory diseases. These diseases include conditions as varied as atherosclerosis, arthritis, dermatitis, glomerulonephritis, or acute lung injury. In this context, one can note that inflammation is a convenient but imprecise catch-all term that is used to cover a wide range of situations. Therefore, when discussing the role of platelets in inflammation, it is important to clearly define the pathophysiological context and the exact stage of the reaction. Inflammatory reactions are indeed multistep processes that can be either acute or chronic, and their sequence can vary greatly depending on the situation and organ concerned. Here, we focus on how platelets contribute to inflammatory reactions involving recruitment of neutrophils and/or macrophages. Specifically, we review past and recent data showing that platelets intervene at various stages of these reactions to regulate parameters such as endothelial permeability, the recruitment of neutrophils and macrophages and their effector functions, as well as inflammatory bleeding. The mechanisms underlying these various modulating effect of platelets are also discussed.
Collapse
Affiliation(s)
- Angèle Gros
- Université Paris Diderot, Sorbonne Paris Cité , Paris , France ; Unit 1148, Laboratory for Vascular Translational Science, INSERM , Paris , France
| | - Véronique Ollivier
- Université Paris Diderot, Sorbonne Paris Cité , Paris , France ; Unit 1148, Laboratory for Vascular Translational Science, INSERM , Paris , France
| | - Benoît Ho-Tin-Noé
- Université Paris Diderot, Sorbonne Paris Cité , Paris , France ; Unit 1148, Laboratory for Vascular Translational Science, INSERM , Paris , France
| |
Collapse
|
53
|
Abstract
Pathogenic Leptospira has the capacity to infect a broad range of mammalian hosts. Leptospirosis may appear as an acute, potentially fatal infection in accidental hosts, or progress into a chronic, largely asymptomatic infection in natural maintenance hosts. The course that Leptospira infection follows is dependent upon poorly understood factors, but is heavily influenced by both the host species and bacterial serovar involved in infection. Recognition of pathogen-associated molecular patterns (PAMPs) by a variety of host pattern recognition receptors (PRRs) activates the host immune system. The outcome of this response may result in bacterial clearance, limited bacterial colonization of a few target organs, principally the kidney, or induction of sepsis as the host succumbs to infection and dies. This chapter describes current knowledge of how the host recognizes Leptospira and responds to infection using innate and acquired immune responses. Aspects of immune-mediated pathology and pathogen strategies to evade the host immune response are also addressed.
Collapse
Affiliation(s)
- Richard L Zuerner
- Department of Biomedical Sciences and Veterinary Public Health, Swedish University for Agricultural Sciences, 75007, Uppsala, Sweden,
| |
Collapse
|
54
|
Wang YQ, Wang B, Liang Y, Cao SH, Liu L, Xu XN. Role of platelet TLR4 expression in pathogensis of septic thrombocytopenia. World J Emerg Med 2014; 2:13-7. [PMID: 25214976 DOI: 10.5847/wjem.j.1920-8642.2011.01.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2010] [Accepted: 12/15/2010] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Infection-induced thrombocytopenia (TCP) is an independent risk factor for death of patients with sepsis, but its mechanism is unknown. This study aimed to explore the underlying mechanism of TCP based on the relationship between TLR4 expression and platelet activation in septic patients. METHODS A total of 64 patients with sepsis were prospectively studied. Platelet count (PC), mean platelet volume (MPV), platelet distribution width (PDW), platelet TLR4 expression, platelet PAC-1 expression, sCD40L and TNF-α concentrations were compared between the healthy control group (15 volunteers) and sepsis group (64 patients) at admission and on the 3, 5, and 9 days after admission. The changes of MPV and PDW in the TCP and non-TCP subgroups of sepsis before and after treatment were recorded. Prognostic index was analyzed. RESULTS PC was lower in the sepsis group (P=0.006), and MPV and PDW were higher in the sepsis group than those in the healthy control group (P=0.046, P=0.001). Platelet TLR4 and PAC-1 expressions, and sCD40L and TNF-α levels increased more significantly in the sepsis group (P<0.001). PAC-1 expression and TNF-α level were higher in the TCP group than in the non-TCP group before and after treatment (P=0.023, P=0.011). sCD40L concentration and platelet TLR4 expression were significantly higher in the treated TCP group than in the non-TCP group (P=0.047, P=0.001). Compared to the non-TCP group, the rate of bleeding was higher (P=0.024) and the length of ICU stay was longer (P=0.013). The APACHE II score and the 28-day mortality were higher in the TCP group (P<0.01, P=0.048). CONCLUSIONS The elevation of platelet TLR4 expression in sepsis along with platelet activation is closely related to the incidence of thrombocytopenia. The occurrence of TCP is a sign of poor prognosis in sepsis patients.
Collapse
Affiliation(s)
- Yong-Qiang Wang
- ICU, Tianjin First Center Hospitial, Tianjin 300192, China (WangYQ, Wang B, Cao SH, Xu XN); Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China (Liang Y); General Hospital of Tianjin Medical University, Tianjin 300052, China (Liu L)
| | - Bing Wang
- ICU, Tianjin First Center Hospitial, Tianjin 300192, China (WangYQ, Wang B, Cao SH, Xu XN); Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China (Liang Y); General Hospital of Tianjin Medical University, Tianjin 300052, China (Liu L)
| | - Yong Liang
- ICU, Tianjin First Center Hospitial, Tianjin 300192, China (WangYQ, Wang B, Cao SH, Xu XN); Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China (Liang Y); General Hospital of Tianjin Medical University, Tianjin 300052, China (Liu L)
| | - Shu-Hua Cao
- ICU, Tianjin First Center Hospitial, Tianjin 300192, China (WangYQ, Wang B, Cao SH, Xu XN); Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China (Liang Y); General Hospital of Tianjin Medical University, Tianjin 300052, China (Liu L)
| | - Li Liu
- ICU, Tianjin First Center Hospitial, Tianjin 300192, China (WangYQ, Wang B, Cao SH, Xu XN); Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China (Liang Y); General Hospital of Tianjin Medical University, Tianjin 300052, China (Liu L)
| | - Xin-Nv Xu
- ICU, Tianjin First Center Hospitial, Tianjin 300192, China (WangYQ, Wang B, Cao SH, Xu XN); Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China (Liang Y); General Hospital of Tianjin Medical University, Tianjin 300052, China (Liu L)
| |
Collapse
|
55
|
Calpain activity and Toll-like receptor 4 expression in platelet regulate haemostatic situation in patients undergoing cardiac surgery and coagulation in mice. Mediators Inflamm 2014; 2014:484510. [PMID: 25258477 PMCID: PMC4167458 DOI: 10.1155/2014/484510] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 08/07/2014] [Accepted: 08/07/2014] [Indexed: 01/02/2023] Open
Abstract
Human platelets express Toll-like receptors (TLR) 4. However, the mechanism by which TLR4 directly affects platelet aggregation and blood coagulation remains to be explored. Therefore, in this study, we evaluated the platelet TLR4 expression in patients who underwent CABG surgery; we explored the correlation between platelet TLR4 expression and the early outcomes in hospital of patients. Additionally, C57BL/6 and C57BL/6-TlrLPS−/− mice were used to explore the roles of platelet TLR4 in coagulation by platelet aggregometry and rotation thromboelastometry. In conclusion, our results highlight the important roles of TLR4 in blood coagulation and platelet function. Of clinical relevance, we also explored novel roles for platelet TLR4 that are associated with early outcomes in cardiac surgery.
Collapse
|
56
|
Collagen can selectively trigger a platelet secretory phenotype via glycoprotein VI. PLoS One 2014; 9:e104712. [PMID: 25116206 PMCID: PMC4130581 DOI: 10.1371/journal.pone.0104712] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 07/11/2014] [Indexed: 11/22/2022] Open
Abstract
Platelets are not only central actors of hemostasis and thrombosis but also of other processes including inflammation, angiogenesis, and tissue regeneration. Accumulating evidence indicates that these “non classical” functions of platelets do not necessarily rely on their well-known ability to form thrombi upon activation. This suggests the existence of non-thrombotic alternative states of platelets activation. We investigated this possibility through dose-response analysis of thrombin- and collagen-induced changes in platelet phenotype, with regards to morphological and functional markers of platelet activation including shape change, aggregation, P-selectin and phosphatidylserine surface expression, integrin activation, and release of soluble factors. We show that collagen at low dose (0.25 µg/mL) selectively triggers a platelet secretory phenotype characterized by the release of dense- and alpha granule-derived soluble factors without causing any of the other major platelet changes that usually accompany thrombus formation. Using a blocking antibody to glycoprotein VI (GPVI), we further show that this response is mediated by GPVI. Taken together, our results show that platelet activation goes beyond the mechanisms leading to platelet aggregation and also includes alternative platelet phenotypes that might contribute to their thrombus-independent functions.
Collapse
|
57
|
Israels SJ, McNicol A, Dean HJ, Cognasse F, Sellers EAC. Markers of platelet activation are increased in adolescents with type 2 diabetes. Diabetes Care 2014; 37:2400-3. [PMID: 24879840 DOI: 10.2337/dc13-2718] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE In adults with diabetes, in vivo platelet activation is a marker for atherosclerosis and cardiovascular disease (CVD). This pilot study investigated whether adolescents with diabetes had evidence of increased in vivo platelet activation. RESEARCH DESIGN AND METHODS In vivo platelet activation was compared in four groups of age-matched adolescents: type 1 diabetes (T1D, n = 15), type 2 diabetes (T2D; n = 15), control subjects with normal BMI (n = 14), and overweight/obese control subjects (n = 13). Platelet surface activation markers and plasma levels of soluble activation markers were measured and compared among groups. RESULTS Increased expression of all activation markers was observed in T2D compared with either control group (P < 0.05); levels of soluble markers were also higher in T2D than in T1D (P < 0.05). There were no differences in marker expression between the nondiabetic control groups. CONCLUSIONS Platelet activation in adolescents with T2D may be a marker for the risk of CVD development in early adulthood.
Collapse
Affiliation(s)
- Sara J Israels
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Archibald McNicol
- Department of Oral Biology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Heather J Dean
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Fabrice Cognasse
- Groupe Immunité des Muqueuses et Agents Pathogènes-EA 3064, Faculty of Medicine, University of Lyon/Saint-Etienne, Saint-Etienne, FranceEtablissement Français du Sang Auvergne-Loire, Saint-Etienne, France
| | - Elizabeth A C Sellers
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
58
|
Ding N, Chen G, Hoffman R, Loughran PA, Sodhi CP, Hackam DJ, Billiar TR, Neal MD. Toll-like receptor 4 regulates platelet function and contributes to coagulation abnormality and organ injury in hemorrhagic shock and resuscitation. ACTA ACUST UNITED AC 2014; 7:615-24. [PMID: 25049041 DOI: 10.1161/circgenetics.113.000398] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Growing evidence indicates that the presence of toll-like receptor 4 (TLR4) on platelets is a key regulator of platelet number and function. Platelets exposed to TLR4 agonists may serve to activate other cells such as neutrophils and endothelial cells in sepsis and other inflammatory conditions. The functional significance of platelet TLR4 in hemorrhagic shock (HS), however, remains unexplored. METHODS AND RESULTS Using thromboelastography and platelet aggregometry, we demonstrate that platelet function is impaired in a mouse model of HS with resuscitation. Further analysis using cellular-specific TLR4 deletion in mice revealed that platelet TLR4 is essential for platelet activation and function in HS with resuscitation and that platelet TLR4 regulates the development of coagulopathy after hemorrhage and resuscitation. Transfusion of TLR4-negative platelets into mice resulted in protection from coagulopathy and restored platelet function. Additionally, platelet-specific TLR4 knockout mice were protected from lung and liver injury and exhibited a marked reduction in systemic inflammation as measured by circulating interleukin-6 after HS with resuscitation. CONCLUSIONS We demonstrate for the first time that platelet TLR4 is an essential mediator of the inflammatory response as well as platelet activation and function in HS and resuscitation.
Collapse
Affiliation(s)
- Ning Ding
- From the Department of Surgery, University of Pittsburgh, PA (N.D., G.C., R.H., P.A.L., T.R.B., M.D.N.); Department of Anesthesiology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China (N.D.); Department of Anesthesiology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai, People's Republic of China (G.C.); and Division of Pediatric Surgery, Department of Surgery, Children's Hospital of Pittsburgh of UPMC, PA (C.P.S., D.J.H.)
| | - Guoqiang Chen
- From the Department of Surgery, University of Pittsburgh, PA (N.D., G.C., R.H., P.A.L., T.R.B., M.D.N.); Department of Anesthesiology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China (N.D.); Department of Anesthesiology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai, People's Republic of China (G.C.); and Division of Pediatric Surgery, Department of Surgery, Children's Hospital of Pittsburgh of UPMC, PA (C.P.S., D.J.H.)
| | - Rosemary Hoffman
- From the Department of Surgery, University of Pittsburgh, PA (N.D., G.C., R.H., P.A.L., T.R.B., M.D.N.); Department of Anesthesiology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China (N.D.); Department of Anesthesiology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai, People's Republic of China (G.C.); and Division of Pediatric Surgery, Department of Surgery, Children's Hospital of Pittsburgh of UPMC, PA (C.P.S., D.J.H.)
| | - Patricia A Loughran
- From the Department of Surgery, University of Pittsburgh, PA (N.D., G.C., R.H., P.A.L., T.R.B., M.D.N.); Department of Anesthesiology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China (N.D.); Department of Anesthesiology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai, People's Republic of China (G.C.); and Division of Pediatric Surgery, Department of Surgery, Children's Hospital of Pittsburgh of UPMC, PA (C.P.S., D.J.H.)
| | - Chhinder P Sodhi
- From the Department of Surgery, University of Pittsburgh, PA (N.D., G.C., R.H., P.A.L., T.R.B., M.D.N.); Department of Anesthesiology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China (N.D.); Department of Anesthesiology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai, People's Republic of China (G.C.); and Division of Pediatric Surgery, Department of Surgery, Children's Hospital of Pittsburgh of UPMC, PA (C.P.S., D.J.H.)
| | - David J Hackam
- From the Department of Surgery, University of Pittsburgh, PA (N.D., G.C., R.H., P.A.L., T.R.B., M.D.N.); Department of Anesthesiology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China (N.D.); Department of Anesthesiology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai, People's Republic of China (G.C.); and Division of Pediatric Surgery, Department of Surgery, Children's Hospital of Pittsburgh of UPMC, PA (C.P.S., D.J.H.)
| | - Timothy R Billiar
- From the Department of Surgery, University of Pittsburgh, PA (N.D., G.C., R.H., P.A.L., T.R.B., M.D.N.); Department of Anesthesiology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China (N.D.); Department of Anesthesiology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai, People's Republic of China (G.C.); and Division of Pediatric Surgery, Department of Surgery, Children's Hospital of Pittsburgh of UPMC, PA (C.P.S., D.J.H.).
| | - Matthew D Neal
- From the Department of Surgery, University of Pittsburgh, PA (N.D., G.C., R.H., P.A.L., T.R.B., M.D.N.); Department of Anesthesiology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China (N.D.); Department of Anesthesiology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai, People's Republic of China (G.C.); and Division of Pediatric Surgery, Department of Surgery, Children's Hospital of Pittsburgh of UPMC, PA (C.P.S., D.J.H.).
| |
Collapse
|
59
|
|
60
|
Platelets protect from septic shock by inhibiting macrophage-dependent inflammation via the cyclooxygenase 1 signalling pathway. Nat Commun 2014; 4:2657. [PMID: 24150174 DOI: 10.1038/ncomms3657] [Citation(s) in RCA: 145] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Accepted: 09/23/2013] [Indexed: 12/31/2022] Open
Abstract
Although it has long been known that patients with sepsis often have thrombocytopenia and that septic patients with severe thrombocytopenia have a poor prognosis and higher mortality, the role of platelets in the pathogenesis of sepsis is poorly understood. Here we report a protective role of platelets in septic shock. We show that experimental thrombocytopenia induced by intraperitoneal injection of an anti-glycoprotein Ibα monoclonal antibody increases mortality and aggravates organ failure, whereas transfusion of platelets reduces mortality in lipopolysaccharide-induced endotoxemia and a bacterial infusion mouse sepsis model. Plasma concentrations of proinflammatory cytokines TNF-α and IL-6 are elevated by thrombocytopenia and decreased by platelet transfusion in septic mice. Furthermore, we identify that platelets protect from septic shock by inhibiting macrophage-dependent inflammation via the COX1/PGE₂/EP4-dependent pathway. Thus, these findings demonstrate a previously unappreciated role for platelets in septic shock and suggest that platelet transfusion may be effective in treating severely septic patients.
Collapse
|
61
|
Abstract
Unlike other blood cells, platelets are small anucleate structures derived from marrow megakaryocytes. Thought for almost a century to possess solely hemostatic potentials, platelets, however, play a much wider role in tissue regeneration and repair and interact intimately with tumor cells. On one hand, tumor cells induce platelet aggregation (TCIPA), known to act as the trigger of cancer-associated thrombosis. On the other hand, platelets recruited to the tumor microenvironment interact, directly, with tumor cells, favoring their proliferation, and, indirectly, through the release of a wide palette of growth factors, including angiogenic and mitogenic proteins. In addition, the role of platelets is not solely confined to the primary tumor site. Indeed, they escort tumor cells, helping their intravasation, vascular migration, arrest, and extravasation to the tissues to form distant metastasis. As expected, nonspecific or specific inhibition of platelets and their content represents an attractive novel approach in the fight against cancer. This review illustrates the role played by platelets at primary tumor sites and in the various stages of the metastatic process.
Collapse
Affiliation(s)
- Hadi A Goubran
- Saskatoon Cancer Centre and Division of Oncology, Department of Medicine, College of Medicine, University of Saskatchewan, SK, Canada.
| | - Julie Stakiw
- Saskatoon Cancer Centre and Division of Oncology, Department of Medicine, College of Medicine, University of Saskatchewan, SK, Canada
| | | | - Thierry Burnouf
- Institute of Biomedical Materials and Tissue Engineering, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
62
|
Cognasse F, Hamzeh-Cognasse H, Chabert A, Jackson E, Arthaud CA, Garraud O, McNicol A. Streptococcus sanguinis-induced cytokine and matrix metalloproteinase-1 release from platelets. BMC Immunol 2014; 15:15. [PMID: 24755160 PMCID: PMC3998947 DOI: 10.1186/1471-2172-15-15] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Accepted: 04/07/2014] [Indexed: 01/01/2023] Open
Abstract
Background Streptococcus sanguinis (S.sanguinis), a predominant bacterium in the human oral cavity, has been widely associated with the development of infective endocarditis. Platelets play both a haemostatic function and can influence both innate and adaptive immune responses. Previous studies have shown that S.sanguinis can interact with, and activate, platelets. Results The aim of this study was to determine whether S.sanguinis stimulates the release of matrix metalloproteinases (MMPs) 1, 2 and 9 and the pro-inflammatory mediators SDF-1, VEGF and sCD40L, from platelets and to subsequently pharmacologically address the release mechanism (s). S.sanguinis stimulated the release of MMP-1, SDF-1, VEGF and sCD40L from platelets and inhibitors of cyclooxygenase and phosphatidylinositol 3-kinase, and antagonists of the αIIbβ3 integrin and glycoprotein Ib, each inhibited the secretion of all factors. Conclusions Therefore the release of MMP-1, SDF-1, VEGF and sCD40L occurs late in the platelet response to S.sanguinis and highlights the complex intracellular signalling pathways stimulated in response to S.sanguinis which lead to haemostasis, MMP and pro-inflammatory mediator secretion.
Collapse
Affiliation(s)
- Fabrice Cognasse
- Etablissement Français du Sang (EFS) Auvergne-Loire, Saint-Etienne, France.
| | | | | | | | | | | | | |
Collapse
|
63
|
Abstract
Platelets play crucial functions in hemostasis and the prevention of bleeding. During H1N1 influenza A virus infection, platelets display activation markers. The platelet activation triggers during H1N1 infection remain elusive. We observed that H1N1 induces surface receptor activation, lipid mediator synthesis, and release of microparticles from platelets. These activation processes require the presence of serum/plasma, pointing to the contribution of soluble factor(s). Considering that immune complexes in the H1N1 pandemic were reported to play a pathogenic role, we assessed their contribution in H1N1-induced platelet activation. In influenza-immunized subjects, we observed that the virus scaffolds with immunoglobulin G (IgG) to form immune complexes that promote platelet activation. Mechanistically, this activation occurs through stimulation of low-affinity type 2 receptor for Fc portion of IgG (FcγRIIA), a receptor for immune complexes, independently of thrombin. Using a combination of in vitro and in vivo approaches, we found that the antibodies from H3N2-immunized mice activate transgenic mouse platelets that express FcγRIIA when put in the presence of H1N1, suggesting that cross-reacting influenza antibodies suffice. Alternatively, H1N1 can activate platelets via thrombin formation, independently of complement and FcγRIIA. These observations identify both the adaptive immune response and the innate response against pathogens as 2 intertwined processes that activate platelets during influenza infections.
Collapse
|
64
|
Secor D, Swarbreck S, Ellis CG, Sharpe MD, Tyml K. Ascorbate reduces mouse platelet aggregation and surface P-selectin expression in an ex vivo model of sepsis. Microcirculation 2014; 20:502-10. [PMID: 23402318 DOI: 10.1111/micc.12047] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Accepted: 02/05/2013] [Indexed: 12/23/2022]
Abstract
OBJECTIVE Compromised perfusion of the capillary bed can lead to organ failure and mortality in sepsis. We have reported that intravenous injection of ascorbate inhibits platelet adhesion and plugging in septic capillaries. In this study, we hypothesized that ascorbate reduces aggregation of platelets and their surface expression of P-selectin (a key adhesion molecule) in mice. METHODS Platelets were isolated from control mice and subjected to agents known to be released into the bloodstream during sepsis (thrombin, ADP or U46619, thromboxane A2 analog). Platelet aggregation was analyzed by aggregometry and P-selectin expression by flow cytometry. RESULTS Platelet-activating agents increased aggregation and P-selectin expression. Ascorbate inhibited these increases. This inhibitory effect was NOS-independent (LNAME had no effect). In contrast to the platelet-activating agents, direct stimuli lipopolysaccharide, TNFα, or plasma from septic mice did not increase aggregation/expression, a finding consistent with the literature. The results suggest a complex mechanism of platelet aggregation and P-selectin expression in sepsis, where generation of platelet-activating stimuli is required first, before platelet aggregation and adhesion in capillaries occur. CONCLUSION The ability of ascorbate to reduce platelet aggregation and P-selectin expression could be an important mechanism by which ascorbate inhibits capillary plugging in sepsis.
Collapse
Affiliation(s)
- Dan Secor
- Critical Illness Research, Lawson Health Research Institute, London, Ontario, Canada
| | | | | | | | | |
Collapse
|
65
|
Corken A, Russell S, Dent J, Post SR, Ware J. Platelet glycoprotein Ib-IX as a regulator of systemic inflammation. Arterioscler Thromb Vasc Biol 2014; 34:996-1001. [PMID: 24504734 DOI: 10.1161/atvbaha.113.303113] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVE The platelet glycoprotein Ib-IX (GP Ib-IX) receptor is a well-characterized adhesion receptor supporting hemostasis and thrombosis via interactions with von Willebrand factor. We examine the GP Ib-IX/von Willebrand factor axis in murine polymicrobial sepsis, as modeled by cecal ligation and puncture (CLP). APPROACH AND RESULTS Genetic absence of the GP Ib-IX ligand, von Willebrand factor, prolongs survival after CLP, but absence of the receptor, GP Ib-IX, does not. Because absence of either von Willebrand factor or GP Ib-IX significantly impairs hemostasis and thrombosis, we sought to define additional GP Ib-IX-dependent pathways impacting survival in the CLP model. We document that the absence of GP Ib-IX leads to reduced platelet-neutrophil and platelet-monocyte interactions. Twenty-four hours after CLP, absence of GP Ib-IX coincides with an alteration in cytokine levels, such as tumor necrosis factor-α secreted by monocytes, and increased macrophage-1 antigen expression by neutrophils. CONCLUSIONS In contrast to the well-characterized proinflammatory properties of platelets, we describe in the CLP model an anti-inflammatory property associated with platelet GP Ib-IX. Thus, a single platelet receptor displays a dual modulatory role in both the thrombotic and inflammatory pathways associated with polymicrobial sepsis. In sharing leucine-rich motifs with toll-like receptors, platelet GP Ib-IX can be considered a multifunctional participant in hemostasis, thrombosis, and the inflammatory cascade. The results highlight a dynamic role for platelets in systemic inflammation and add to the complex pathophysiologic events that occur during the dysregulated coagulation and inflammation associated with sepsis.
Collapse
Affiliation(s)
- Adam Corken
- From the Departments of Physiology and Biophysics (A.C., S.R., J.D., J.W.) and Pathology (S.R.P.), University of Arkansas for Medical Sciences, Little Rock
| | | | | | | | | |
Collapse
|
66
|
Tsai JC, Lin YW, Huang CY, Lin CY, Tsai YT, Shih CM, Lee CY, Chen YH, Li CY, Chang NC, Lin FY, Tsai CS. The role of calpain-myosin 9-Rab7b pathway in mediating the expression of Toll-like receptor 4 in platelets: a novel mechanism involved in α-granules trafficking. PLoS One 2014; 9:e85833. [PMID: 24489676 PMCID: PMC3904858 DOI: 10.1371/journal.pone.0085833] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Accepted: 12/02/2013] [Indexed: 01/26/2023] Open
Abstract
Toll-like receptors (TLRs) plays a critical role in innate immunity. In 2004, Aslam R. and Shiraki R. first determined that murine and human platelets express functional TLRs. Additionally, Andonegui G. demonstrated that platelets express TLR4, which contributes to thrombocytopenia. However, the underlying mechanisms of TLR4 expression by platelets have been rarely explored until now. The aim of this study was to identify the mechanism of TLR4 expression underlying thrombin treatment. The human washed platelets were used in this study. According to flowcytometry and western blot analysis, the surface levels of TLR4 were significantly enhanced in thrombin-activated human platelets and decreased by TMB-8, calpeptin, and U73122, but not Y27632 (a Rho-associated protein kinase ROCK inhibitor) indicating that thrombin-mediated TLR4 expression was modulated by PAR/PLC pathway, calcium and calpain. Co-immunoprecipitation (co-IP) assay demonstrated that the interaction between TLR4 and myosin-9 (a substrate of calpain) was regulated by calpain; cleavage of myosin-9 enhanced TLR4 expression in thrombin treated platelets. Transmission electron microscope data indicated that human platelets used α-granules to control TLR4 expression; the co-IP experiment suggested that myosin-9 did not coordinate with Rab7b to negatively regulate TLR4 trafficking in thrombin treated platelets. In summary, phospholipase Cγ-calpain-myosin 9-Rab7b axis was responsible for the mechanism underlying the regulation of TLR4 containing α-granules trafficking in thrombin-stimulated platelets, which was involved in coagulation.
Collapse
Affiliation(s)
- Jui-Chi Tsai
- Graduate Institute of Medical Sciences, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- Departments of Internal Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yi-Wen Lin
- Division of Cardiovascular Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chun-Yao Huang
- Departments of Internal Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Cardiology and Cardiovascular Research Center, Taipei Medical University Hospital, Taipei, Taiwan
| | - Chih-Yuan Lin
- Division of Cardiovascular Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Yi-Ting Tsai
- Division of Cardiovascular Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chun-Min Shih
- Departments of Internal Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Cardiology and Cardiovascular Research Center, Taipei Medical University Hospital, Taipei, Taiwan
| | - Chung-Yi Lee
- Division of Cardiovascular Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Yung-Hsiang Chen
- Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan
| | - Chi-Yuan Li
- Graduate Institute of Clinical Medical Sciences, China Medical University, Taichung, Taiwan
- Department of Anesthesiology, China Medical University Hospital, Taichung, Taiwan
| | - Nen-Chung Chang
- Departments of Internal Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Cardiology and Cardiovascular Research Center, Taipei Medical University Hospital, Taipei, Taiwan
| | - Feng-Yen Lin
- Departments of Internal Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Cardiology and Cardiovascular Research Center, Taipei Medical University Hospital, Taipei, Taiwan
- * E-mail: (FYL); (CST)
| | - Chien-Sung Tsai
- Division of Cardiovascular Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- * E-mail: (FYL); (CST)
| |
Collapse
|
67
|
Abstract
PURPOSE OF REVIEW The platelet paradigm that is well established in hemostasis and thrombosis can be extended to other disease states. A consideration for some major health issues, such as inflammation, cancer, infection, and neuroscience, and how platelet function impacts the pathophysiology of each clinical situation is provided. RECENT FINDINGS Decades of research and knowledge of platelet function exist and the same is true for inflammation and cancer. The literature is full of platelet biology overlapping into other, nonthrombotic disease states. However, major gaps exist that prevent a complete mechanistic understanding of platelet function in these other diseases. Although much of the overlap provides antidotal relationships, future studies will likely uncover novel pathophysiological pathways that are highly relevant to human diseases. SUMMARY Recent findings in four major disease areas, inflammation, cancer, infection, and neuroscience, are described, with current literature linking the disease to platelet function. The availability of antiplatelet therapies, such as aspirin, exists and future consideration can be given as to whether antiplatelet therapy is potentially beneficial or harmful as the mechanisms of platelet involvement are better defined.
Collapse
|
68
|
Kappelmayer J, Beke Debreceni I, Vida A, Antal-Szalmás P, Clemetson KJ, Nagy B. Distinct effects of Re- and S-forms of LPS on modulating platelet activation. J Thromb Haemost 2013; 11:775-8. [PMID: 23347166 DOI: 10.1111/jth.12151] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2012] [Accepted: 01/17/2013] [Indexed: 11/30/2022]
|
69
|
Berlacher MD, Vieth JA, Heflin BC, Gay SR, Antczak AJ, Tasma BE, Boardman HJ, Singh N, Montel AH, Kahaleh MB, Worth RG. FcγRIIa Ligation Induces Platelet Hypersensitivity to Thrombotic Stimuli. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 182:244-54. [DOI: 10.1016/j.ajpath.2012.09.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Revised: 09/07/2012] [Accepted: 09/13/2012] [Indexed: 01/18/2023]
|
70
|
|
71
|
Berthet J, Damien P, Hamzeh-Cognasse H, Arthaud CA, Eyraud MA, Zéni F, Pozzetto B, McNicol A, Garraud O, Cognasse F. Human platelets can discriminate between various bacterial LPS isoforms via TLR4 signaling and differential cytokine secretion. Clin Immunol 2012; 145:189-200. [PMID: 23108090 DOI: 10.1016/j.clim.2012.09.004] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Revised: 09/07/2012] [Accepted: 09/12/2012] [Indexed: 12/15/2022]
Abstract
Platelets are currently acknowledged as cells of innate immunity and inflammation and play a complex role in sepsis. We examined whether different types of LPS have different effects on the release of soluble signaling/effective molecules from platelets. We used platelet-rich plasma from healthy volunteers and LPS from two strains of gram-negative bacteria with disparate LPS structures. We combined LPS-stimulated platelet supernatants with reporter cells and measured the PBMC cytokine secretion profiles. Upon stimulation of platelets with both Escherichia coli O111 and Salmonella minnesota LPS, the platelet LPS::TLR4 interaction activated pathways to trigger the production of a large number of molecules. The different platelet supernatants caused differential PBMC secretion of IL-6, TNFα, and IL-8. Our data demonstrate that platelets have the capacity to sense external signals differentially through a single type of pathogen recognition receptor and adjust the innate immune response appropriately for pathogens exhibiting different types of 'danger' signals.
Collapse
Affiliation(s)
- Julien Berthet
- Université de Lyon, F-42023, GIMAP, EA3064, Saint-Etienne, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
72
|
Abstract
AbstractPlatelets survey blood vessels, searching for endothelial damage and preventing loss of vascular integrity. However, there are circumstances where vascular permeability increases, suggesting that platelets sometimes fail to fulfill their expected function. Human inflammatory arthritis is associated with tissue edema attributed to enhanced permeability of the synovial microvasculature. Murine studies have suggested that such vascular leak facilitates entry of autoantibodies and may thereby promote joint inflammation. Whereas platelets typically help to promote microvascular integrity, we examined the role of platelets in synovial vascular permeability in murine experimental arthritis. Using an in vivo model of autoimmune arthritis, we confirmed the presence of endothelial gaps in inflamed synovium. Surprisingly, permeability in the inflamed joints was abrogated if the platelets were absent. This effect was mediated by platelet serotonin accumulated via the serotonin transporter and could be antagonized using serotonin-specific reuptake inhibitor antidepressants. As opposed to the conventional role of platelets to microvascular leakage, this demonstration that platelets are capable of amplifying and maintaining permeability adds to the rapidly growing list of unexpected functions for platelets.
Collapse
|
73
|
Garraud O, Hamzeh-Cognasse H, Cognasse F. Platelets and cytokines: How and why? Transfus Clin Biol 2012; 19:104-8. [PMID: 22682309 DOI: 10.1016/j.tracli.2012.02.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Accepted: 02/29/2012] [Indexed: 12/11/2022]
Abstract
For patients with platelet deficiencies, platelet components are therapeutic products for which there is no substitute. However, transfusion complications are more frequent with this labile blood product than with others. This is attributable to products secreted by the platelets themselves, including a variety of cytokines, chemokines, and biological response modifiers, some of which are secreted in large quantities following platelet activation. Why platelets are activated and prone to releasing these molecules during certain inflammatory and innate immune responses is not yet fully understood, but it could be due to several parameters including incompatibilities between blood donors and recipients, the process of platelet preparation and preservation, and the ability of the donor's immune system to sense danger presented by external stimuli during the blood donation process. This review presents our current knowledge of how the platelets that constitute the platelet component for transfusion are sources of cytokines and biological response modifiers and discusses methods to improve the quality of blood transfusion products and safety for patients.
Collapse
Affiliation(s)
- O Garraud
- Établissement français du sang Auvergne-Loire, 25, boulevard Pasteur, 42023 Saint-Étienne cedex 02, France.
| | | | | |
Collapse
|
74
|
Fuentes Q. E, Fuentes Q. F, Andrés V, Pello OM, de Mora JF, Palomo G. I. Role of platelets as mediators that link inflammation and thrombosis in atherosclerosis. Platelets 2012; 24:255-62. [DOI: 10.3109/09537104.2012.690113] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
75
|
Abstract
Observational and experimental studies continue to support the association of infection and infection-stimulated inflammation with development of cardiovascular disease (CVD) including atherosclerosis and thrombosis. Microvesicles (MV) are heterogeneous populations of sealed membrane-derived vesicles shed into circulation by activated mammalian cells and/or pathogenic microbes that may represent an interface between bacterial/microbial infection and increased risk of CVD. This review evaluates how MV act to modulate and intersect immunological and inflammatory responses to infection with particular attention to progression of CVD. Although infection-related stimuli provoke release of MV from blood and vascular cells, MV express phosphatidylserine and other procoagulant factors on their surface, which initiate and amplify blood coagulation. In addition, MV mediate cell-cell adhesion, which may stimulate production of pro-inflammatory cytokines in vascular cells, which in turn aggravate progression of CVD and propagate atherothrombosis. MV transfer membrane receptors, RNA and proteins among cells, and present auto-antigens from their cells of origin to proximal or remote target cells. Because MV harbor cell surface proteins and contain cytoplasmic components of the parent cell, they mediate biological messages and play a pivotal role in the crossroad between infection-stimulated inflammation and CVDs.
Collapse
Affiliation(s)
- Jing Xiong
- Department of Physiology, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
- Department of Surgery, Mayo Clinic, Rochester, Minnesota 55905
| | - Virginia M. Miller
- Department of Surgery, Mayo Clinic, Rochester, Minnesota 55905
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota 55905
| | - Yunman Li
- Department of Physiology, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Muthuvel Jayachandran
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota 55905
| |
Collapse
|
76
|
McNicol A, Agpalza A, Jackson ECG, Hamzeh-Cognasse H, Garraud O, Cognasse F. Streptococcus sanguinis-induced cytokine release from platelets. J Thromb Haemost 2011; 9:2038-49. [PMID: 21824285 DOI: 10.1111/j.1538-7836.2011.04462.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
BACKGROUND There is increasing evidence that both chronic and acute infections play a role in the development and progression of atherothrombotic disorders. One potential mechanism is the direct activation of platelets by bacteria. A wide range of bacterial species activate platelets through heterogeneous mechanisms. The oral micro-organism S. sanguinis stimulates platelet aggregation in vitro in a strain-dependent manner, although there are no reports of associated cytokine production. OBJECTIVE The aim of the present study was to determine whether platelet activation by S. sanguinis involved the release of pro-inflammatory and immune modulating factors, and whether activation was enhanced by epinephrine. METHODS AND RESULTS Four strains of S. sanguinis and one of S. gordonii stimulated the release of RANTES, PF4, sCD40L and PDGF-AB, whereas only one S. sanguinis strain caused the release of sCD62p. Epinephrine enhanced S. sanguinis-induced platelet aggregation and phosphorylation of phospholipase Cγ2 and Erk, but inhibited RANTES, PF4, sCD40L and PDGF-AB release. Wortmannin inhibited S. sanguinis-induced aggregation and release; however, only aggregation was partially reversed by epinephrine. CONCLUSIONS The present study demonstrates that platelets respond to S. sanguinis with both prothrombotic and pro-inflammatory/immune-modulating responses. Epinephrine, potentially released in response to infection and/or stress, can significantly enhance the prothrombotic response, thereby providing a putative link between bacteraemia and acute coronary events during stress. In contrast, epinephrine inhibited the pro-inflammatory/immune-modulating response by an undetermined mechanism.
Collapse
Affiliation(s)
- A McNicol
- Department of Oral Biology, University of Manitoba, Winnipeg, MB, Canada.
| | | | | | | | | | | |
Collapse
|
77
|
Vieira-de-Abreu A, Campbell RA, Weyrich AS, Zimmerman GA. Platelets: versatile effector cells in hemostasis, inflammation, and the immune continuum. Semin Immunopathol 2011; 34:5-30. [PMID: 21818701 DOI: 10.1007/s00281-011-0286-4] [Citation(s) in RCA: 230] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2011] [Accepted: 07/20/2011] [Indexed: 12/28/2022]
Abstract
Platelets are chief effector cells in hemostasis. In addition, however, their specializations include activities and intercellular interactions that make them key effectors in inflammation and in the continuum of innate and adaptive immunity. This review focuses on the immune features of human platelets and platelets from experimental animals and on interactions between inflammatory, immune, and hemostatic activities of these anucleate but complex and versatile cells. The experimental findings and evidence for physiologic immune functions include previously unrecognized biologic characteristics of platelets and are paralleled by new evidence for unique roles of platelets in inflammatory, immune, and thrombotic diseases.
Collapse
Affiliation(s)
- Adriana Vieira-de-Abreu
- Department of Medicine, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | | | | | | |
Collapse
|
78
|
Abstract
The haemostatic role of platelets was established in the 1880s by Bizzozero who observed their ability to adhere and aggregate at sites of vascular injury. It was only some 80 years later that the function of platelets in maintaining the structural integrity of intact blood vessels was reported by Danielli. Danielli noted that platelets help preserve the barrier function of endothelium during organ perfusion. Subsequent studies have demonstrated further that platelets are continuously needed to support intact mature blood vessels. More recently, platelets were shown to safeguard developing vessels, lymphatics, as well as the microvasculature at sites of leukocyte infiltration, including inflamed organs and tumours. Interestingly, from a mechanistic point of view, the supporting role of platelets in these various vessels does not necessarily involve the well-understood process of platelet plug formation but, rather, may rely on secretion of the various platelet granules and their many active components. The present review focuses on these nonconventional aspects of platelet biology and function by presenting situations in which platelets intervene to maintain vascular integrity and discusses possible mechanisms of their actions. We propose that modulating these newly described platelet functions may help treat haemorrhage as well as treat cancer by increasing the efficacy of drug delivery to tumours.
Collapse
|
79
|
Cox D, Kerrigan SW, Watson SP. Platelets and the innate immune system: mechanisms of bacterial-induced platelet activation. J Thromb Haemost 2011; 9:1097-107. [PMID: 21435167 DOI: 10.1111/j.1538-7836.2011.04264.x] [Citation(s) in RCA: 214] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
It has become clear that platelets are not simply cell fragments that plug the leak in a damaged blood vessel; they are, in fact, also key components in the innate immune system, which is supported by the presence of Toll-like receptors (TLRs) on platelets. As the cells that respond first to a site of injury, they are well placed to direct the immune response to deal with any resulting exposure to pathogens. The response is triggered by bacteria binding to platelets, which usually triggers platelet activation and the secretion of antimicrobial peptides. The main platelet receptors that mediate these interactions are glycoprotein (GP)IIb-IIIa, GPIbα, FcγRIIa, complement receptors, and TLRs. This process may involve direct interactions between bacterial proteins and the receptors, or can be mediated by plasma proteins such as fibrinogen, von Willebrand factor, complement, and IgG. Here, we review the variety of interactions between platelets and bacteria, and look at the potential for inhibiting these interactions in diseases such as infective endocarditis and sepsis.
Collapse
Affiliation(s)
- D Cox
- Molecular and Cellular Therapeutics School of Pharmacy, Royal College of Surgeons in Ireland, Dublin, Ireland.
| | | | | |
Collapse
|
80
|
Rondina MT, Schwertz H, Harris ES, Kraemer BF, Campbell RA, Mackman N, Grissom CK, Weyrich AS, Zimmerman GA. The septic milieu triggers expression of spliced tissue factor mRNA in human platelets. J Thromb Haemost 2011; 9:748-58. [PMID: 21255247 PMCID: PMC3071458 DOI: 10.1111/j.1538-7836.2011.04208.x] [Citation(s) in RCA: 109] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
BACKGROUND Activated platelets have previously-unrecognized mechanisms of post-transcriptional gene expression that may influence hemostasis and inflammation. A novel pathway involves splicing of pre-mRNAs in resting platelets to mature, translatable mRNAs in response to cellular activation. OBJECTIVES We asked if bacterial products and host agonists present in the septic milieu induce tissue factor pre-mRNA splicing in platelets from healthy subjects. In parallel, we asked if spliced tissue factor (TF) mRNA is present in platelets from septic patients in a proof-of-principle analysis. PATIENTS/METHODS TF pre-mRNA and mRNA expression patterns were characterized in platelets from septic patients and in platelets isolated from healthy subjects activated with bacteria, toxins and inflammatory agonists. Procoagulant activity was also measured. RESULTS AND CONCLUSIONS Live bacteria, staphylococcal α-toxin and lipopolysaccharide (LPS) induced TF pre-mRNA splicing in platelets isolated from healthy subjects. Toxin-stimulated platelets accelerated plasma clotting, a response that was blocked by a previously-characterized splicing inhibitor and by an anti-tissue factor antibody. Platelets from septic patients expressed spliced TF mRNA, whereas it was absent from unselected and age-matched control subjects. Tissue factor-dependent procoagulant activity was elevated in platelets from a subset of septic patients. Thus, bacterial and host factors induce splicing of TF pre-mRNA, expression of TF mRNA and tissue factor-dependent clotting activity in human platelets. TF mRNA is present in platelets from some septic patients, indicating that it may be a marker of altered platelet phenotype and function in sepsis and that splicing pathways are induced in this syndrome.
Collapse
Affiliation(s)
- M T Rondina
- Division of General Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT 84132, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
81
|
Garraud O, Damien P, Berthet J, Arthaud CA, Hamzeh-Cognasse H, Cognasse F. [Blood platelets and biological response to 'danger' signals and subsequent inflammation: towards a new paradigm?]. Transfus Clin Biol 2011; 18:165-73. [PMID: 21444230 DOI: 10.1016/j.tracli.2011.02.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2011] [Accepted: 02/12/2011] [Indexed: 12/22/2022]
Abstract
Blood platelets are cellular elements of primary haemostasis. During the last decade research on platelets has been subsequently based on this paradigm, with separate observations on issues such as the ability for platelets to bind infectious agents or even engulf them, to drop in counts in case of evolving infectious processes, etc. More recently, novel work has set up bases for novel functions for platelets, as members of functional immune cells, principally in innate immunity but capable of influencing adaptive immunity. Platelets are thus essential to haemostasis and to inflammation, questioning their essential functionality and the set up of a novel paradigm: could platelets be tissue-repairing cells? Such an assumption would open an entire new field of investigations. The present "State of the Art" essay attempts to discuss the main arguments on this.
Collapse
Affiliation(s)
- O Garraud
- EFS Auvergne-Loire Saint-Étienne, 25, boulevard Pasteur, 42023 Saint-Étienne cedex 2, France.
| | | | | | | | | | | |
Collapse
|
82
|
Li Z, Yang F, Dunn S, Gross AK, Smyth SS. Platelets as immune mediators: their role in host defense responses and sepsis. Thromb Res 2011; 127:184-8. [PMID: 21075430 PMCID: PMC3042496 DOI: 10.1016/j.thromres.2010.10.010] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2010] [Revised: 10/15/2010] [Accepted: 10/17/2010] [Indexed: 12/13/2022]
Abstract
Platelets occupy a central role at the interface between thrombosis and inflammation. At sites of vascular damage, adherent platelets physically and functionally interact with circulating leukocytes. Activated platelets release soluble factors into circulation that may have local and systemic effects on blood and vascular cells. Platelets can also interact with a wide variety of microbial pathogens. Emerging evidence from animal models suggests that platelets may participate in a wide variety of processes involving tissue injury, immune responses and repair that underlie diverse diseases such as atherosclerosis, autoimmune disorders, inflammatory lung and bowel disorders, host-defense responses and sepsis. In this review, we summarize the general mechanisms by which platelets may contribute to immune function, and then discuss evidence for their role in host defense responses and sepsis from preclinical and clinical studies.
Collapse
Affiliation(s)
- Zhenyu Li
- The Division of Cardiovascular Medicine, The Gill Heart Institute
| | - Fanmuyi Yang
- The Division of Cardiovascular Medicine, The Gill Heart Institute
| | - Steve Dunn
- College of Pharmacy, University of Kentucky
| | | | | |
Collapse
|
83
|
Internalization of IgG-coated targets results in activation and secretion of soluble CD40 ligand and RANTES by human platelets. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2010; 18:210-6. [PMID: 21177916 DOI: 10.1128/cvi.00296-10] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Platelets are crucial elements for maintenance of hemostasis. Other functions attributable to platelets are now being appreciated, such as their role in inflammatory reactions and host defense. Platelets have been reported to bind immunological stimuli like IgG complexes, and for nearly 50 years it has been speculated that platelets may participate in immunological reactions. Platelets have been reported to bind and internalize various substances, similar to other leukocytes, such as macrophages and dendritic cells. In the present study, we tested the hypothesis that human platelets can bind and internalize IgG-coated particles, similar to leukocytes. To this end, we observed that interaction with IgG-coated beads resulted in platelet activation (as measured by CD62P expression), internalization of targets, and significant soluble CD40 ligand (sCD40L) and RANTES (regulated upon activation, normal T cell expresses and secreted) secretion. Blocking FcγRIIA with monoclonal antibody (MAb) IV.3 or inhibiting actin remodeling with cytochalasin D inhibited platelet activation, internalization, and cytokine production. These data suggest that platelets are capable of mediating internalization of IgG-coated particles, resulting in platelet activation and release of both sCD40L and RANTES.
Collapse
|
84
|
Garraud O, Berthet J, Hamzeh-Cognasse H, Cognasse F. Pathogen sensing, subsequent signalling, and signalosome in human platelets. Thromb Res 2010; 127:283-6. [PMID: 21071069 DOI: 10.1016/j.thromres.2010.10.015] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2010] [Revised: 10/15/2010] [Accepted: 10/17/2010] [Indexed: 12/17/2022]
Abstract
Beyond haemostasis, platelets exert a potent role in innate immunity and particularly in its inflammatory arm. The extent of this action remains however debatable, despite clear - and old - evidence of a link between platelets and infection. Platelets can sense infectious pathogens by pathogen recognition receptors and they can even discriminate between various types of infectious signatures. In reply, they can shape their capacity to respond by activating a signalosome and by producing different profiles of pro-inflammatory cytokines and related products. The links between pathogen sensing, signalosome activation and protein production, and their finely tuned regulation are still under investigation since platelets lack a nucleus and thus, canonical molecular biology and genomics apparati.
Collapse
Affiliation(s)
- Olivier Garraud
- Etablissement Français du Sang Auvergne-Loire & EA 3064, Faculty of Medicine, University of Saint-Etienne, a Member of the University of Lyon, France.
| | | | | | | |
Collapse
|
85
|
de Mast Q, de Groot PG, van Heerde WL, Roestenberg M, van Velzen JF, Verbruggen B, Roest M, McCall M, Nieman AE, Westendorp J, Syafruddin D, Fijnheer R, van Dongen-Lases EC, Sauerwein RW, van der Ven AJ. Thrombocytopenia in early malaria is associated with GP1b shedding in absence of systemic platelet activation and consumptive coagulopathy. Br J Haematol 2010; 151:495-503. [PMID: 20955404 DOI: 10.1111/j.1365-2141.2010.08399.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Thrombocytopenia develops early in malaria, but the underlying mechanisms remain incompletely understood. We studied the aetiology of malaria-associated thrombocytopenia in volunteers experimentally infected with Plasmodium falciparum malaria, in Indonesian malaria patients and in ex vivo studies. In experimental human malaria, the decrease in platelet counts was associated with a concurrent rise in young platelets (immature platelet fraction) and thrombopoietin. D-dimer concentrations were moderately elevated without a prolongation in the activated partial thromboplastin time or decrease in fibrinogen. There was no increase in expression of the platelet surface markers CD62P, PAC-1 and CD63 and in plasma concentrations of the platelet factors P-selectin, CXCR4, CXCL7, RANTES and CD40L. In contrast, concentrations of soluble glycoprotein-1b (sGP1b), the external domain of the platelet receptor for von Willebrand factor (VWF), increased early. Indonesian malaria patients also had elevated concentrations of sGP1b, which correlated with VWF concentrations. Finally, incubation of platelets with parasitized erythrocytes in vitro failed to induce platelet aggregation or activation. We concluded that neither compromised platelet production nor platelet activation or consumptive coagulopathy were responsible for the early thrombocytopenia in malaria. We hypothesize that the increase in sGP1b concentrations results from VWF-mediated GP1b shedding; a process that may prevent excessive adhesion of platelets and parasitized erythrocytes.
Collapse
Affiliation(s)
- Quirijn de Mast
- Department of General Internal Medicine, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
86
|
Qu Z, Chaikof EL. Interface between hemostasis and adaptive immunity. Curr Opin Immunol 2010; 22:634-42. [PMID: 20932735 DOI: 10.1016/j.coi.2010.08.017] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2010] [Accepted: 08/31/2010] [Indexed: 12/15/2022]
Abstract
Stress induced activation or denudation of the endothelium elicits arrest and activation of platelets with attendant triggering of coagulation, culminating in a physical barrier to limit blood loss. Recently, coagulation-activated osteopontin, chemerin, and protease-activated receptor signaling, as well as platelet-derived molecules including platelet factor 4, serotonin, P-selectin, and CD154 (CD40L) have been revealed as new links between hemostasis and adaptive immunity. The initiation of hemostasis establishes a local state of inflammation that serves as an adjuvant system for antigen presentation, consequently influencing the onset and functional characteristics of an evolving adaptive immune response. In this context, the hemostatic system and its associated signaling pathways warrant further study as novel therapeutic targets that may enhance, abrogate, or otherwise selectively direct the adaptive immune response.
Collapse
Affiliation(s)
- Zheng Qu
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | | |
Collapse
|
87
|
Berthet J, Damien P, Hamzeh-Cognasse H, Pozzetto B, Garraud O, Cognasse F. Toll-like receptor 4 signal transduction in platelets: novel pathways. Br J Haematol 2010; 151:89-92. [PMID: 20618335 DOI: 10.1111/j.1365-2141.2010.08292.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
88
|
Yeaman MR. Bacterial-platelet interactions: virulence meets host defense. Future Microbiol 2010; 5:471-506. [PMID: 20210555 DOI: 10.2217/fmb.09.112] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Platelets have historically been viewed as cell fragments that only mediate blood coagulation. Yet, platelets have as - or perhaps even more - important roles in tissue remodeling, modulation of inflammation and antimicrobial host defense. It is evident that platelets interact with prokaryotes directly and indirectly through multiple molecular and cellular mechanisms. The important roles of platelets in antibacterial host defense can be exemplified through contemporary themes in platelet immunobiology. Platelets have unambiguous structures and functions of host defense effector cells. Recent discoveries reveal platelet expression of toll-like and purinonergic receptors, which enable detection and response to bacterial infection, degranulation of an array of microbicidal peptides and coordination of other molecular and cellular host defenses. From multiple perspectives, platelets are now increasingly recognized as critical innate immune effector cells that also bridge and facilitate optimization of adaptive immunity. It follows that clinical deficiencies in platelet quantity or quality are now recognized correlates of increased risk and severity of bacterial and other infections. Along these lines, new evidence suggests that certain prokaryotic organisms may be capable of exploiting platelet interactions to gain a virulence advantage. Indeed, certain bacterial pathogens appear to have evolved highly coordinated means by which to seize opportunities to bind to surfaces of activated platelets, and exploit them to establish or propagate infection. Hence, it is conceivable that certain bacterial pathogens subvert platelet functions. From these perspectives, the net consequences of bacterial virulence versus platelet host defenses likely decide initial steps towards the ultimate result of infection versus immunity.
Collapse
Affiliation(s)
- Michael R Yeaman
- Department of Medicine, David Geffen School of Medicine at UCLA, Harbor-UCLA Medical Center, 1124 West Carson Street, RB-2, Torrance, CA 90502, USA.
| |
Collapse
|
89
|
McNicol A, Israels SJ. Mechanisms of oral bacteria-induced platelet activation. Can J Physiol Pharmacol 2010; 88:510-24. [DOI: 10.1139/y10-029] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The oral cavity is inhabited by over 500 different bacterial species that normally exist in ecological balance both with each other and with the host. When this equilibrium is disturbed, an overgrowth of individual organisms can occur, which, in turn, can lead to the onset of pathological processes, notably dental caries and periodontitis. Generally, bacteraemias occur more frequently in individuals with periodontal disease, and these bacteraemias have been implicated in the development of a range of systemic diseases, including atherothrombotic disorders. The mechanism underlying this relationship remains to be precisely defined, although studies have shown a link between bacteria of oral origin and platelet activation. Several orally derived species of bacteria interact with platelets, including those of the Streptococcus ( Streptococcus sanguinis , Streptococcus mutans , Streptococcus agalactiae , Streptococcus pyogenes , Streptococcus gordonii , Streptococcus pneumoniae , Streptococcus mitis ) and Staphylococcus ( Staphylococcus epidermidis , Staphylococcus capitis ) genera, as well as Pseudomonas aeruginosa and Porphyromonas gingivalis . In addition, some members of both the Streptococcus and the Staphylococcus genera, as well as Porphyromonas gingivalis , can activate platelets in vitro. The current review describes the heterogeneous mechanisms of platelet activation employed by individual bacterial species. The pathological and clinical implications of platelet activation by orally derived bacteria are discussed.
Collapse
Affiliation(s)
- Archibald McNicol
- Department of Oral Biology, University of Manitoba, Winnipeg, MB R3E 0W2, Canada
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, MB R3E 0W2, Canada
- International Centre for Oral Systemic Health, University of Manitoba, Winnipeg, MB R3E 0W2, Canada
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB R3E 0W2, Canada
- Manitoba Institute of Cell Biology, University of Manitoba, Winnipeg, MB R3E 0W2, Canada
| | - Sara J. Israels
- Department of Oral Biology, University of Manitoba, Winnipeg, MB R3E 0W2, Canada
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, MB R3E 0W2, Canada
- International Centre for Oral Systemic Health, University of Manitoba, Winnipeg, MB R3E 0W2, Canada
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB R3E 0W2, Canada
- Manitoba Institute of Cell Biology, University of Manitoba, Winnipeg, MB R3E 0W2, Canada
| |
Collapse
|
90
|
Horstman LL, Jy W, Ahn YS, Zivadinov R, Maghzi AH, Etemadifar M, Steven Alexander J, Minagar A. Role of platelets in neuroinflammation: a wide-angle perspective. J Neuroinflammation 2010; 7:10. [PMID: 20128908 PMCID: PMC2829540 DOI: 10.1186/1742-2094-7-10] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2009] [Accepted: 02/03/2010] [Indexed: 01/03/2023] Open
Abstract
OBJECTIVES This review summarizes recent developments in platelet biology relevant to neuroinflammatory disorders. Multiple sclerosis (MS) is taken as the "Poster Child" of these disorders but the implications are wide. The role of platelets in inflammation is well appreciated in the cardiovascular and cancer research communities but appears to be relatively neglected in neurological research. ORGANIZATION After a brief introduction to platelets, topics covered include the matrix metalloproteinases, platelet chemokines, cytokines and growth factors, the recent finding of platelet PPAR receptors and Toll-like receptors, complement, bioactive lipids, and other agents/functions likely to be relevant in neuroinflammatory diseases. Each section cites literature linking the topic to areas of active research in MS or other disorders, including especially Alzheimer's disease. CONCLUSION The final section summarizes evidence of platelet involvement in MS. The general conclusion is that platelets may be key players in MS and related disorders, and warrant more attention in neurological research.
Collapse
Affiliation(s)
- Lawrence L Horstman
- Wallace Coulter Platelet Laboratory, Division of Hematology and Oncology, Department of Medicine, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Wenche Jy
- Wallace Coulter Platelet Laboratory, Division of Hematology and Oncology, Department of Medicine, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Yeon S Ahn
- Wallace Coulter Platelet Laboratory, Division of Hematology and Oncology, Department of Medicine, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Robert Zivadinov
- Buffalo Neuroimaging Analysis Center, The Jacobs Neurological Institute, Department of Neurology, School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo NY, USA
| | - Amir H Maghzi
- Department of Neurology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Masoud Etemadifar
- Department of Neurology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - J Steven Alexander
- Department of Cellular and Molecular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA
| | - Alireza Minagar
- Department of Neurology, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA
| |
Collapse
|
91
|
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
|
92
|
Abstract
Many bacteria are capable of interacting with platelets and inducing platelet aggregation. This interaction may be a direct interaction between a bacterial surface protein and a platelet receptor or may be an indirect interaction where plasma proteins bind to the bacterial surface and subsequently bind to a platelet receptor. However, these interactions usually do not trigger platelet activation as a secondary co-signal is also required. This is usually due to specific antibody bound to the bacteria interacting with FcgammaRIIa on the platelet surface. Secreted bacterial products such as gingipains and lipopolysaccharide may also be capable of triggering platelet activation.
Collapse
Affiliation(s)
- Steven W. Kerrigan
- School of Pharmacy, Royal College of Surgeons in Ireland, 123 St Stephens Green, Dublin 2, Ireland
- Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, 123 St Stephens Green, Dublin 2, Ireland
| | - Dermot Cox
- Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, 123 St Stephens Green, Dublin 2, Ireland
| |
Collapse
|
93
|
Abstract
Platelets interact with bacterial pathogens through a wide array of cellular and molecular mechanisms. The consequences of this interaction may significantly influence the balance between infection and immunity. On the one hand, recent data indicate that certain bacteria may be capable of exploiting these interactions to gain a virulence advantage. Indeed, certain bacterial pathogens appear to have evolved specific ways in which to subvert activated platelets. Hence, it is conceivable that some bacterial pathogens exploit platelet responses. On the other hand, platelets are now known to possess unambiguous structures and functions of host defense effector cells. Recent discoveries emphasize critical features enabling such functions, including expression of toll-like receptors that detect hallmark signals of bacterial infection, an array of microbicidal peptides, as well as other host defense molecules and functions. These concepts are consistent with increased risk and severity of bacterial infection as correlates of clinical abnormalities in platelet quantity and quality. In these respects, the molecular and cellular roles of platelets in host defense against bacterial pathogens are explored with attention on advances in platelet immunobiology.
Collapse
Affiliation(s)
- Michael R Yeaman
- Division of Infectious Diseases, St. John's Cardiovascular Research Center, Harbor-UCLA Medical Center, David Geffen School of Medicine at UCLA, Torrance, CA 90502, USA.
| |
Collapse
|
94
|
IgG-complex stimulated platelets: A source of sCD40L and RANTES in initiation of inflammatory cascade. Cell Immunol 2010; 263:129-33. [DOI: 10.1016/j.cellimm.2010.03.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2010] [Accepted: 03/16/2010] [Indexed: 12/15/2022]
|
95
|
The role of inflammation in regulating platelet production and function: Toll-like receptors in platelets and megakaryocytes. Thromb Res 2009; 125:205-9. [PMID: 19945154 DOI: 10.1016/j.thromres.2009.11.004] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2009] [Revised: 10/27/2009] [Accepted: 11/02/2009] [Indexed: 12/17/2022]
Abstract
Platelets have been extensively studied as hemostatic regulators, stopping uncontrolled flow of blood from an injured vessel and allowing for repair. However, multiple studies have shown that platelets can interact with bacterial proteins, particularly seen during sepsis and inflammation. Immune cells recognize pathogens through Toll-like Receptors (TLRs). These same receptors allow platelets to recognize bacterial proteins and regulate platelet immunity and function. This review examines the TLRs expressed on platelets and megakaryocytes and how these receptors affect the function of these cells. Through TLRs, platelets go beyond hemostatic regulation and play a pivotal role in inflammation and infection.
Collapse
|
96
|
Cognasse F, Hamzeh-Cognasse H, Berthet J, Damien P, Lucht F, Pozzetto B, Garraud O. Altered release of regulated upon activation, normal T-cell expressed and secreted protein from human, normal platelets: contribution of distinct HIV-1MN gp41 peptides. AIDS 2009; 23:2057-2059. [PMID: 19654498 DOI: 10.1097/qad.0b013e328330da65] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Platelets can bind HIV and, in turn, be altered in with respect to and function during HIV progression. This study examines the secretion of normal platelets after exposure to recombinant HIV-1MN gp120 or gp41 peptides. There was a modest but significant decrease in regulated upon activation, normal T-cell expressed and secreted protein production in the presence of two out of 10 peptides, which was restored by monoclonal antibodies to gp41. Our data provide novel information on possible primary interactions between platelets and HIV env proteins.
Collapse
Affiliation(s)
- Fabrice Cognasse
- aEFS Auvergne-Loire, and GIMAP-EA3064, Université de Saint-Etienne--membre de l'Université de Lyon, Faculté de Médecine, Saint-Etienne, France
| | | | | | | | | | | | | |
Collapse
|
97
|
Ho-Tin-Noé B, Carbo C, Demers M, Cifuni SM, Goerge T, Wagner DD. Innate immune cells induce hemorrhage in tumors during thrombocytopenia. THE AMERICAN JOURNAL OF PATHOLOGY 2009; 175:1699-708. [PMID: 19729481 DOI: 10.2353/ajpath.2009.090460] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Platelets are crucial regulators of tumor vascular homeostasis and continuously prevent tumor hemorrhage through secretion of their granules. However, the reason for tumor bleeding in the absence of platelets remains unknown. Tumors are associated with inflammation, a cause of hemorrhage in thrombocytopenia. Here, we investigated the role of the inflamed tumor microenvironment in the induction of tumor vessel injury in thrombocytopenic mice. Using s.c. injections of vascular endothelial growth factor or tumor necrosis factor-alpha combined with depletion of neutrophils, we demonstrate that enhancing the opening of endothelial cell junctions was not sufficient to cause bleeding in the absence of platelets; instead, induction of tissue hemorrhage in thrombocytopenia required recruitment of leukocytes. Immunohistology revealed that thrombocytopenia-induced tumor hemorrhage occurs at sites of macrophage and neutrophil accumulation. Mice deficient in beta2 or beta3 integrins, which have decreased neutrophil and/or macrophage infiltration in their tumor stroma, were protected from thrombocytopenia-induced tumor hemorrhage, indicating that, in the absence of platelets, stroma-infiltrating leukocytes induced tumor vessel injury. This injury was independent of reactive oxygen species generation and of complement activation, as suggested by the persistence of tumor hemorrhage in C3- and nicotinamide adenine dinucleotide phosphate oxidase-deficient thrombocytopenic mice. Our results show that platelets counteract tumor-associated inflammation and that the absence of this platelet function elicits vascular injuries by tumor-infiltrating innate immune cells.
Collapse
|
98
|
Kulka M, Fukuishi N, Metcalfe DD. Human mast cells synthesize and release angiogenin, a member of the ribonuclease A (RNase A) superfamily. J Leukoc Biol 2009; 86:1217-26. [PMID: 19625371 DOI: 10.1189/jlb.0908517] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
ANG is a plasma protein with angiogenic and ribonucleolytic activity implicated in tumor growth, heart failure, wound healing, asthma, and the composition of the adult gut microflora. Human mast cells (HuMC) are similarly associated with modulation of vascular permeability, angiogenic processes, wound healing, and asthma. We hypothesized that HuMC express and secrete ANG in response to divergent stimuli. ANG expression was evaluated in the LAD2 HMC, the HMC-1, and CD34+-derived HuMC, following exposure to live Escherichia coli, TLR ligands, or neuropeptides and following FcepsilonRI aggregation. Expression and production of ANG were determined by microarray analysis, qRT-PCR, confocal microscopy, and ELISA. Microarray analysis showed that ANG is up-regulated by LAD2 cells exposed to live E. coli. qRT-PCR analysis revealed that LAD2, HMC-1, and HuMC constitutively expressed ANG mRNA and that it was up-regulated by exposure to E. coli. Activation of HuMC by FcepsilonRI aggregation resulted in release of small amounts of ANG (<100 pg/mL), whereas compound 48/80, NGF, LPS, PGN, and flagellin activated HuMC to secrete >160 pg/mL ANG. These observations demonstrate that HuMC store and secrete ANG to a variety of stimuli and suggest that MC-derived ANG is available in the subsequent inflammatory response.
Collapse
Affiliation(s)
- Marianna Kulka
- National Research Council, 550 University Ave., Charlottetown, PE, Canada.
| | | | | |
Collapse
|
99
|
Abstract
Solid tumors generate a prothrombotic environment capable of platelet activation. Recent findings indicate that the activated platelets are crucial regulators of tumor vascular homeostasis in that they prevent tumor hemorrhage. Surprisingly, this effect is independent of platelets' capacity to form thrombi and instead relies on the secretion of their granule content. Thus, targeting platelet secretory activities may represent a new approach to specifically destabilize tumor vasculature.
Collapse
Affiliation(s)
- Benoit Ho-Tin-Noé
- Immune Disease Institute, Harvard Medical School, Boston, Massachusetts
- Department of Pathology, Harvard Medical School, Boston, Massachusetts
- Inserm Unit 698, Bichat Hospital, Paris, France
| | - Tobias Goerge
- Immune Disease Institute, Harvard Medical School, Boston, Massachusetts
- Department of Pathology, Harvard Medical School, Boston, Massachusetts
- University of Münster, Department of Dermatology, Münster, Germany
| | - Denisa D. Wagner
- Immune Disease Institute, Harvard Medical School, Boston, Massachusetts
- Department of Pathology, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
100
|
Hashimoto K, Jayachandran M, Owen WG, Miller VM. Aggregation and microparticle production through toll-like receptor 4 activation in platelets from recently menopausal women. J Cardiovasc Pharmacol 2009; 54:57-62. [PMID: 19528814 PMCID: PMC3021258 DOI: 10.1097/fjc.0b013e3181ab373d] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Bacterial infection may increase risk for thrombosis and atherosclerosis. Human platelets express toll-like receptor 4 (TLR4), the receptor for gram-negative bacterial lipopolysaccharide (LPS). Experiments were designed to evaluate direct, acute effects of TLR4 activation on aggregation, secretion, and generation of prothrombogenic microparticles in vitro on platelets derived from healthy women at risk for development of cardiovascular disease because of their hormonal status. Platelet-rich plasma from recently menopausal women was incubated with ultrapure Escherichia coli LPS in the absence or presence of antibodies that neutralize the human TLR4. Incubating platelets with LPS (100 ng/mL) for 5 minutes decreased aggregation and dense granule adenosine triphosphate secretion induced by thrombin receptor agonist peptide (TRAP) but not by adenosine diphosphate or collagen. The antibody to TLR4 blocked this effect of LPS. TLR4 activation increased phosphorylation of p38 mitogen-activated protein kinase and decreased production of prothrombotic phosphatidylserine and P-selectin-positive microparticles in response to TRAP. Therefore, acute, direct activation of TLR4 reduces platelet reactivity to TRAP stimulation in vitro. Increased thrombotic and cardiovascular risk with bacterial infection most likely reflects the sum of TLR4 activation on other blood and vascular cells to release proinflammatory cytokines/chemokines, which indirectly affect platelet reactivity.
Collapse
Affiliation(s)
| | - Muthuvel Jayachandran
- Department of Surgery, College of Medicine, Mayo Clinic, Rochester, MN
- Department of Physiology and Biomedical Engineering, College of Medicine, Mayo Clinic, Rochester, MN
| | - Whyte G. Owen
- Department of Biochemistry and Molecular Biology, College of Medicine, Mayo Clinic, Rochester, MN
| | - Virginia M. Miller
- Department of Surgery, College of Medicine, Mayo Clinic, Rochester, MN
- Department of Physiology and Biomedical Engineering, College of Medicine, Mayo Clinic, Rochester, MN
| |
Collapse
|