Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
1.
Blood ; 138(25): 2607-2620, 2021 12 23.
Artigo em Inglês | MEDLINE | ID: mdl-34293122

RESUMO

In addition to their hemostatic role, platelets play a significant role in immunity. Once activated, platelets release extracellular vesicles (EVs) formed by the budding of their cytoplasmic membranes. Because of their heterogeneity, platelet EVs (PEVs) are thought to perform diverse functions. It is unknown, however, whether the proteasome is transferred from platelets to PEVs or whether its function is retained. We hypothesized that functional protein processing and antigen presentation machinery are transferred to PEVs by activated platelets. Using molecular and functional assays, we found that the active 20S proteasome was enriched in PEVs, along with major histocompatibility complex class I (MHC-I) and lymphocyte costimulatory molecules (CD40L and OX40L). Proteasome-containing PEVs were identified in healthy donor blood, but did not increase in platelet concentrates that caused adverse transfusion reactions. They were augmented, however, after immune complex injections in mice. The complete biodistribution of murine PEVs after injection into mice revealed that they principally reached lymphoid organs, such as spleen and lymph nodes, in addition to the bone marrow, and to a lesser extent, liver and lungs. The PEV proteasome processed exogenous ovalbumin (OVA) and loaded its antigenic peptide onto MHC-I molecules, which promoted OVA-specific CD8+ T-lymphocyte proliferation. These results suggest that PEVs contribute to adaptive immunity through cross-presentation of antigens and have privileged access to immune cells through the lymphatic system, a tissue location that is inaccessible to platelets.


Assuntos
Plaquetas/imunologia , Vesículas Extracelulares/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Complexo de Endopeptidases do Proteassoma/imunologia , Animais , Apresentação de Antígeno , Plaquetas/química , Vesículas Extracelulares/química , Antígenos de Histocompatibilidade Classe I/análise , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Complexo de Endopeptidases do Proteassoma/análise
2.
Brain Behav Immun ; 114: 242-254, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37648005

RESUMO

A growing body of evidences suggests that suicidal ideation (SI) and suicidal behaviors have biological bases. However, no biological marker is currently available to evaluate the suicide risk in individuals with SI or suicide attempt (SA). Moreover, the current risk assessment techniques poorly predict future suicidal events. The aim of this study was to examine the association of 39 new and already described peripheral cells and proteins (implicated in the immune system, oxidative stress and plasticity) with lifetime SA, past month SA, current SI, and future suicidal events (visit to the Emergency Department for SI or SA) in 266 treatment-seeking individuals with mood disorders. Equal parts of patients with and without past history of SA were recruited. All individuals at inclusion gave blood, were evaluated for SA recency, current SI, and were followed for two years afterwards. The 39 peripheral blood cellular and protein markers were entered separately for each outcome in Elastic Net models with 10-fold cross-validation, followed by single-analyte covariate-adjusted regression analyses for pre-selected analytes. Past month SA was associated with increased plasma levels of thrombospondin-2 and C-reactive protein, whereas current SI was associated with lower plasma serotonin levels. These associations were robust to adjustments for key covariates and corrections for multiple testing. The Cox proportional hazards regression showed that higher levels of thrombospondin-1 and of platelet-derived growth factor-AB predicted a future suicidal event. These two associations remained after adjustment for sex, age, and SA history, and outperformed the predictive value of past SA. Thrombospondins and platelet-derived growth factors have never been investigated in the context of suicide. Altogether, our results highlight the involvement in the suicidal process of platelet biological response and plasticity modifiers and also of inflammatory factors. They also suggest that SI and SA may have different biological correlates and that biomarkers associated with past SA or current SI do not automatically also predict future events.

3.
Transfusion ; 63(8): 1506-1518, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37387566

RESUMO

BACKGROUND: Blood donors at the extremes of the age spectrum (16-19 years vs. ≥75 years) are characterized by increased risks of iron deficiency and anemia, and are often underrepresented in studies evaluating the effects of donor characteristics on red blood cells (RBC) transfusion effectiveness. The aim of this study was to conduct quality assessments of RBC concentrates from these unique age groups. STUDY DESIGN: We characterized 150 leukocyte-reduced (LR)-RBCs units from 75 teenage donors, who were matched by sex, and ethnicity with 75 older donors. LR-RBC units were manufactured at three large blood collection centers in the USA and Canada. Quality assessments included storage hemolysis, osmotic hemolysis, oxidative hemolysis, osmotic gradient ektacytometry, hematological indices, and RBC bioactivity. RESULTS: RBC concentrates from teenage donors had smaller (9%) mean corpuscular volume and higher (5%) RBC concentration compared with older donors counterparts. Stored RBCs from teenage donors exhibited increased susceptibility to oxidative hemolysis (>2-fold) compared with RBCs from older donors. This was observed at all testing centers independent of sex, storage duration, or the type of additive solution. RBCs from teenage male donors had increased cytoplasmatic viscosity and lower hydration compared with older donor RBCs. Evaluations of RBC supernatant bioactivity suggested that donor age was not associated with altered expression of inflammatory markers (CD31, CD54, and IL-6) on endothelial cells. CONCLUSIONS: The reported findings are likely intrinsic to RBCs and reflect age-specific changes in RBC antioxidant capacity and physical characteristics that may impact RBC survival during cold storage and after transfusion.


Assuntos
Doadores de Sangue , Hemólise , Humanos , Masculino , Adolescente , Células Endoteliais , Eritrócitos/metabolismo , Citoplasma , Preservação de Sangue
4.
Transfusion ; 63(1): 217-228, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36453841

RESUMO

BACKGROUND: Structural and biochemical changes in stored platelets are influenced by collection and processing methods. This international study investigates the effects of platelet (PLT) processing and storage conditions on HMGB1, sCD40L, and sCD62P protein levels in platelet concentrate supernatants (PCs). STUDY DESIGN/METHODS: PC supernatants (n = 3748) were collected by each international centre using identical centrifugation methods (n = 9) and tested centrally using the ELISA/Luminex platform. Apheresis versus the buffy coat (BC-PC) method, plasma storage versus PAS and RT storage versus cold (4°C) were investigated. We focused on PC preparation collecting samples during early (RT: day 1-3; cold: day 1-5) and late (RT: day 4-7; cold: day 7-10) storage time points. RESULTS: HMGB1, sCD40L, and sCD62P concentrations were similar during early storage periods, regardless of storage solution (BC-PC plasma and BC-PC PAS-E) or temperature. During storage and without PAS, sCD40L and CD62P in BC-PC supernatants increased significantly (+33% and +41%, respectively) depending on storage temperature (22 vs. 4°C). However, without PAS-E, levels decreased significantly (-31% and -20%, respectively), depending on storage temperature (22 vs. 4°C). Contrastingly, the processing method appeared to have greater impact on HMGB1 release versus storage duration. These data highlight increases in these parameters during storage and differences between preparation methods and storage temperatures. CONCLUSIONS: The HMGB1 release mechanism/intracellular pathways appear to differ from sCD62P and sCD40L. The extent to which these differences affect patient outcomes, particularly post-transfusion platelet increment and adverse events, warrants further investigation in clinical trials with various therapeutic indications.


Assuntos
Remoção de Componentes Sanguíneos , Proteína HMGB1 , Humanos , Remoção de Componentes Sanguíneos/métodos , Plaquetas/metabolismo , Preservação de Sangue/métodos , Ligante de CD40/metabolismo , Proteína HMGB1/metabolismo , Transfusão de Plaquetas
5.
Transfusion ; 62(9): 1721-1726, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35950480

RESUMO

BACKGROUND: Patients with severe thrombocytopenia due to bone marrow failure and after chemotherapy are still treated with platelet transfusions. Platelet concentrates (PC) are associated with a high incidence of adverse reactions (AR). Platelet-derived damage-associated molecular patterns (DAMPS) and complement were proposed to play a role in the pathology of AR. STUDY DESIGN AND METHODS: Single donor apheresis platelet concentrates (SDA PCs) were produced in a regional setting of the French Blood Establishment. After transfusion samples were collected from PC and possible AR in patients were recorded. Platelet activation markers, High mobility group box 1 (HMGB1) and complement activation products (CAP) were measured. The correlation between platelet activation, and HMGB1 and complement activation was analyzed. RESULTS: A total of 56 PC were included in the study. 30 PC induced no AR, and 26 induced AR (Febrile non-hemolytic transfusion reaction n = 16; Atypical Allergic Transfusion Reactions n = 11; hemodynamic instability n = 5) in the patients. The levels of P-selectin, sCD40L, HMGB1, C3b/c, and C4b/c were all significantly increased in PC that induced AR following transfusion in patients. Additionally, HMGB1, C3b/c, and C4b/c were positively correlated with P-selectin and sCD40L. CONCLUSION: In this study, we observed an association between HMGB1 and CAP and the incidence of AR. Furthermore, we demonstrated that both HMGB1 and complement activation were correlated to platelet activation.


Assuntos
Proteína HMGB1 , Reação Transfusional , Alarminas , Plaquetas/fisiologia , Ativação do Complemento , Humanos , Selectina-P , Ativação Plaquetária , Transfusão de Plaquetas/efeitos adversos , Reação Transfusional/etiologia
6.
Int J Mol Sci ; 23(21)2022 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-36361963

RESUMO

Venous thromboembolism (VTE) is the third leading cardiovascular cause of death and is conventionally treated with anticoagulants that directly antagonize coagulation. However, recent data have demonstrated that also platelets play a crucial role in VTE pathophysiology. In the current review, we outline how platelets are involved during all stages of experimental venous thrombosis. Platelets mediate initiation of the disease by attaching to the vessel wall upon which they mediate leukocyte recruitment. This process is referred to as immunothrombosis, and within this novel concept inflammatory cells such as leukocytes and platelets directly drive the progression of VTE. In addition to their involvement in immunothrombosis, activated platelets can directly drive venous thrombosis by supporting coagulation and secreting procoagulant factors. Furthermore, fibrinolysis and vessel resolution are (partly) mediated by platelets. Finally, we summarize how conventional antiplatelet therapy can prevent experimental venous thrombosis and impacts (recurrent) VTE in humans.


Assuntos
Tromboembolia Venosa , Trombose Venosa , Humanos , Plaquetas , Tromboinflamação , Coagulação Sanguínea , Anticoagulantes/farmacologia , Anticoagulantes/uso terapêutico
7.
Virol J ; 18(1): 211, 2021 10 29.
Artigo em Inglês | MEDLINE | ID: mdl-34715884

RESUMO

BACKGROUND: The SARS-CoV-2 virus is the causing agent of the Coronavirus disease 2019 (COVID-19) characterized by a huge pro-inflammatory response and coagulation disorders that may lead to for its severe forms, in organ failure or even death. As major players of thrombo-inflammation, platelets release large amounts of immunomodulatory molecules and regulate leukocyte and endothelial activity, which are both altered in COVID-19. Altogether, this makes platelets a very likely actor of the thrombo-inflammatory complications of COVID-19. Thus, we propose to identify a platelet inflammatory signature of severe COVID-19 specifically modulated throughout the course of the disease. METHODS: Luminex technology and enzyme-linked immunosorbent assay were used to assess plasma levels of platelet inflammatory markers in patients with severe acute respiratory syndrome coronavirus 2 infection on admission and for 14 days afterwards. RESULTS: In accordance with the observations of other teams, we evidence that the plasma levels of the platelet soluble (s)CD40L is significantly elevated in the early stages of the disease. Interestingly we observe that the plasma level of sCD40L decreases overtime while that of sCD62P increases significantly. CONCLUSIONS: Our data suggest that there is a platelet signature of inflammatory response to SARS-COv-2 infection which varies overtime and could serve as monitoring biomarkers of patient inflammatory state. CLINICAL TRIAL REGISTRATION NUMBER: 2020-A01100-39; title: Human Ab Response & immunoMONItoring of COVID-19 Patients, registration date: 05/25/2020; URL of the registry: https://clinicaltrials.gov/ct2/history/NCT04373200?V_5=View .


Assuntos
Biomarcadores/sangue , Plaquetas/imunologia , COVID-19 , Inflamação , Adulto , Idoso , COVID-19/sangue , COVID-19/imunologia , Feminino , Humanos , Inflamação/sangue , Inflamação/imunologia , Masculino , Pessoa de Meia-Idade , Adulto Jovem
8.
Int J Mol Sci ; 22(15)2021 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-34360659

RESUMO

Platelets are hematopoietic cells whose main function has for a long time been considered to be the maintenance of vascular integrity. They have an essential role in the hemostatic response, but they also have functional capabilities that go far beyond it. This review will provide an overview of platelet functions. Indeed, stress signals may induce platelet apoptosis through proapoptotis or hemostasis receptors, necrosis, and even autophagy. Platelets also interact with immune cells and modulate immune responses in terms of activation, maturation, recruitment and cytokine secretion. This review will also show that platelets, thanks to their wide range of innate immune receptors, and in particular toll-like receptors, and can be considered sentinels actively participating in the immuno-surveillance of the body. We will discuss the diversity of platelet responses following the engagement of these receptors as well as the signaling pathways involved. Finally, we will show that while platelets contribute significantly, via their TLRs, to immune response and inflammation, these receptors also participate in the pathophysiological processes associated with various pathogens and diseases, including cancer and atherosclerosis.


Assuntos
Aterosclerose/patologia , Plaquetas/patologia , Imunidade Inata/imunologia , Neoplasias/patologia , Ativação Plaquetária , Receptores Imunológicos/metabolismo , Receptores Toll-Like/metabolismo , Animais , Aterosclerose/imunologia , Aterosclerose/metabolismo , Plaquetas/imunologia , Plaquetas/metabolismo , Humanos , Neoplasias/imunologia , Neoplasias/metabolismo
9.
Transfusion ; 60(4): 713-723, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32108957

RESUMO

BACKGROUND: Transfusion-related acute lung injury (TRALI) is a severe pulmonary reaction due to blood transfusions. The pathophysiology of this complication is still not widely elucidated by the scientific community, especially regarding the direct role of blood platelets within the cellular mechanism responsible for the development of TRALI. STUDY DESIGN AND METHODS: In this study, a mouse model was used to induce the development of antibody-mediated acute lung injury through injections of lipopolysaccharide and an anti-major histocompatibility complex Class I antibody. BALB/c mice were pretreated with an anti-GPIbα antibody, which induces platelet depletion, or ML354, a protease receptor 4 pathway inhibitor, 30 minutes before TRALI induction. RESULTS: Depletion of platelets before TRALI induction appeared to reduce the severity of TRALI without completely inhibiting its development. Also, inhibition of platelet activation by ML354 did not prevent the onset of TRALI. Finally, the stimuli used for TRALI induction also triggered specific platelet activation upon ex vivo stimulation. CONCLUSIONS: This study suggests that blood platelets are not critically required for TRALI induction, although they are to some extent involved in its pathophysiology.


Assuntos
Lesão Pulmonar Aguda Relacionada à Transfusão/prevenção & controle , Animais , Anticorpos/farmacologia , Plaquetas/efeitos dos fármacos , Humanos , Indóis/farmacologia , Camundongos , Ativação Plaquetária/efeitos dos fármacos , Complexo Glicoproteico GPIb-IX de Plaquetas/imunologia
10.
Int J Mol Sci ; 21(17)2020 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-32858930

RESUMO

While platelet function has traditionally been described in the context of maintaining vascular integrity, recent evidence suggests that platelets can modulate inflammation in a much more sophisticated and nuanced manner than previously thought. Some aspects of this expanded repertoire of platelet function are mediated via expression of Toll-like receptors (TLRs). TLRs are a family of pattern recognition receptors that recognize pathogen-associated and damage-associated molecular patterns. Activation of these receptors is crucial for orchestrating and sustaining the inflammatory response to both types of danger signals. The TLR family consists of 10 known receptors, and there is at least some evidence that each of these are expressed on or within human platelets. This review presents the literature on TLR-mediated platelet activation for each of these receptors, and the existing understanding of platelet-TLR immune modulation. This review also highlights unresolved methodological issues that potentially contribute to some of the discrepancies within the literature, and we also suggest several recommendations to overcome these issues. Current understanding of TLR-mediated platelet responses in influenza, sepsis, transfusion-related injury and cardiovascular disease are discussed, and key outstanding research questions are highlighted. In summary, we provide a resource-a "researcher's toolkit"-for undertaking further research in the field of platelet-TLR biology.


Assuntos
Plaquetas/imunologia , Trombose/metabolismo , Receptores Toll-Like/metabolismo , Animais , Regulação da Expressão Gênica , Humanos , Imunomodulação , Ativação Plaquetária
11.
Transfusion ; 59(3): 1090-1101, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30784079

RESUMO

BACKGROUND: Acute lung injury (ALI) is a severe complication of transfusion. In a previous study, we saw that inhibition of the CD40/CD40L complex allowed restoration of ALI lesions in an experimental mouse model. OBJECTIVES: This study focused on pancreas-associated injury development during experimental ALI pathogenesis and its limitation through CD40/CD40L complex inhibition. MATERIALS AND METHODS: An ALI mouse model was established through intraperitoneal lipopolysaccharide and intravenous anti-major histocompatibility complex class I monoclonal antibody injection. Preemption of lesions was achieved with intravenous injection of neutralizing anti-CD40L monoclonal antibody 30 minutes before the trigger, that is, anti-major histocompatibility complex class I monoclonal antibody administration. Histology and immunoassay analyses were used to evaluate pancreatic lesions. RESULTS: ALI development induced significant degradation of the lungs and pancreas and was associated with pancreatic lesions. Different scores were established showing more severe injury to the pancreas in ALI conditions; however, injury was significantly reduced through CD40/CD40L complex inhibition. CONCLUSION: This study supports the idea that several organs are exposed during ALI development, and particularly when such experimental ALI aims at mimicking transfusion-associated ALI; nevertheless, preventive treatment inhibiting CD40/CD40L (sCD40L) complex formation provides protection from lung disease as well as disease of other organs.


Assuntos
Lesão Pulmonar Aguda/metabolismo , Antígenos CD40/metabolismo , Ligante de CD40/metabolismo , Pâncreas/metabolismo , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/imunologia , Animais , Antígenos CD40/imunologia , Ligante de CD40/imunologia , Lipopolissacarídeos/toxicidade , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Pâncreas/imunologia
12.
Transfusion ; 59(7): 2403-2414, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30973972

RESUMO

BACKGROUND: Whereas platelet transfusion is a common medical procedure, inflammation still occurs in a fraction of transfused individuals despite the absence of any apparent infectious agents. Platelets can shed membrane vesicles, called extracellular vesicles (EVs), some of which contain mitochondria (mito+EV). With its content of damage-associated molecular pattern (DAMP), the mitochondrion can stimulate the innate immune system. Mitochondrial DNA (mtDNA) is a recognized DAMP detected in the extracellular milieu in numerous inflammatory conditions and in platelet concentrates. We hypothesized that platelet-derived mitochondria encapsulated in EVs may represent a reservoir of mtDNA. STUDY DESIGN AND METHODS: Herein, we explored the implication of mito+EVs in the occurrence of mtDNA quantified in platelet concentrate supernatants that induced or did not induce transfusion adverse reactions. RESULTS: We observed that EVs were abundant in platelet concentrates, and platelet-derived mito+EVs were more abundant in platelet concentrates that induced adverse reactions. A significant correlation (rs = 0.73; p < 0.0001) between platelet-derived mito+EV levels and mtDNA concentrations was found. However, there was a nonsignificant correlation between the levels of EVs without mitochondria and mtDNA concentrations (rs = -0.11; p = 0.5112). The majority of the mtDNA was encapsulated into EVs. CONCLUSION: This study suggests that platelet-derived EVs, such as those that convey mitochondrial DAMPs, may be a useful biomarker for the prediction of potential risk of adverse transfusion reactions. Moreover, our work implies that investigations are necessary to determine whether there is a causal pathogenic role of mitochondrial DAMP encapsulated in EVs as opposed to mtDNA in solution.


Assuntos
Plaquetas/metabolismo , DNA Mitocondrial/metabolismo , Vesículas Extracelulares/metabolismo , Transfusão de Plaquetas , Reação Transfusional/metabolismo , Humanos , Inflamação/metabolismo
13.
Transfusion ; 59(1): 16-20, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30291753

RESUMO

BACKGROUND: Platelet storage lesions are structural and biochemical changes in platelet concentrates (PCs), and depend on variables in collection and processing, as well as secondary procedures and storage conditions; such lesions can be mitigated by the use of platelet additive solutions (PASs). STUDY DESIGN AND METHODS: This study investigated release of the inflammatory markers sCD40L and sCD62P by single-donor apheresis platelet concentrates (SDA-PCs) and buffy coat-derived pooled platelet concentrates (PPCs) before and after storage. SDA-PC and PPC samples (n = 9089) processed by various methods and stored for different durations were obtained following production in one regional setting, the French National Blood Service. Soluble factors were quantified in PC supernatants immediately after processing and at the time of delivery, using biological testing technology (Luminex). RESULTS: SDA-PCs appeared more activated than PPCs at the end of the production step (i.e., prior to storage); however, proinflammatory soluble factors exhibited greater increases in PPCs than in SDA-PCs during storage. In SDA-PCs, PAS-D (65%) led to reduced secretion of sCD62P, but favored secretion of sCD40L, compared with the alternative PAS-E. CONCLUSION: These data stress the importance of the production (processing) steps of PC manufacture and of storage. The extent to which they affect patient outcomes awaits further investigation in clinical studies.


Assuntos
Buffy Coat/metabolismo , Ligante de CD40/metabolismo , Selectina-P/metabolismo , Plaquetoferese/métodos , Buffy Coat/citologia , Preservação de Sangue , Humanos , Inflamação/metabolismo
14.
Transfusion ; 58(11): 2635-2644, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30325037

RESUMO

BACKGROUND: Platelet transfusions are safe but can nevertheless cause serious adverse reactions (SARs). This study investigated the effects of platelet biological response modifiers (BRMs) that accumulate during storage and are commonly associated with transfusion adverse reactions. STUDY DESIGN AND METHODS: Endothelial cells (ECs), that is, EA.hy926, were exposed in vitro to supernatants of platelet components (PCs) that had been either implicated or not in SARs. The EC Biology RT2 Profiler PCR Array was used at the same time to study 84 genes related to functions of ECs. Soluble cytokines and surface expression of EC markers were determined by Luminex/enzyme-linked immunosorbent assay technology and flow cytometry, respectively. Apoptosis and scratch wound assays were performed using IncuCyte technology. RESULTS: In vitro exposure of EA.hy926 monolayers with Day 0, 1-2, and 3-4 stored PC supernatants resulted in decreases in surface expression of markers of ECs. There was differential production of soluble BRMs in the tested cell line. Exposure to the supernatants of PCs that had been implicated in SARs showed a significant difference in the expression of the EC surface markers. EC mediators also responded differently when exposed to PC supernatants of different storage times and PCs involved in SARs. CONCLUSION: PC supernatants collected at Day 1-2 activate fewer cell lines of ECs compared with supernatants collected at Day 3-4. Moreover, PC supernatants involved in SARs appear to alter EC activation compared with the control and storage length.


Assuntos
Plaquetas/metabolismo , Preservação de Sangue , Células Endoteliais/metabolismo , RNA Mensageiro/metabolismo , Apoptose , Biomarcadores/metabolismo , Linhagem Celular , Citometria de Fluxo , Humanos , Transfusão de Plaquetas , Proteômica , Fatores de Tempo
15.
Transfusion ; 58(2): 439-445, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29238987

RESUMO

BACKGROUND: Platelets (PLTs) are prone to activation and the release of biologic response modifiers (BRMs) under storage conditions. The transfusion inflammatory reaction in the vascular compartment involves endothelial cell activation due to cell-cell interactions and BRMs infused with the blood products. Endocan/ESM-1 is a proteoglycan secreted by endothelial cells under the control of proinflammatory cytokines. We aimed to measure endocan activity in supernatants of PLT components (PCs), implicated in serious adverse reactions (SARs) or not (no.AR), sampled at different stages during storage. STUDY DESIGN AND METHODS: PLT function, by quantification of soluble CD62P, and their ability to produce endocan were assessed. Functional testing of PC supernatants was performed on EA.hy926 endothelial cells in vitro by exposing them to PC supernatants from each group (no.AR or SARs); EA.hy926 activation was evaluated by their production of interleukin (IL)-6 and endocan. RESULTS: PLT endocan secretion was not induced in response to PLT surface molecule agonists, and no significant correlation was observed between sCD62P and endocan concentration after PLT activation. However, we observed a significant increase in the secretion of IL-6 and endocan after EA.hy926 activation by all PC supernatants. IL-6 and endocan secretion were significantly higher for cells stimulated with SAR than those stimulated with no.AR PC supernatants, as well as cell apoptosis. CONCLUSION: The correlation between the secretion of endocan and that of IL-6 by endothelial cells suggests that endocan can be used as a predictive marker of inflammation for the quality assessment of transfusion grade PLTs.


Assuntos
Plaquetas/metabolismo , Células Endoteliais/metabolismo , Modelos Biológicos , Proteínas de Neoplasias/metabolismo , Selectina-P/biossíntese , Proteoglicanas/metabolismo , Biomarcadores/metabolismo , Plaquetas/citologia , Técnicas de Cocultura , Células Endoteliais/citologia , Humanos , Interleucina-6/biossíntese , Transfusão de Plaquetas
16.
Platelets ; 29(6): 533-540, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29533683

RESUMO

In addition to their haemostatic role and function in the repair of damaged vascular epithelium, platelets play a defensive role in innate immunity, having the capacity to produce and secrete various anti-infectious factors, as well as cytokines, chemokines and related products, to interact with other immune cells to modulate immune responses to pathogens. Thus, it is now widely acknowledged that platelets participate in inflammatory processes and infection resolution, most notably by expressing and using receptors to bind infectious pathogen moieties and contributing to pathogen clearance. The ability of platelets to sense external danger signals relates to the expression of certain innate immunity receptors, such as toll-like receptors (TLRs), and the activation of efficient cell signalling machinery. TLR engagement triggers platelet response, which results in adapted degranulation according to: the type of TLR engaged, the nature of the ligand and the milieu; together, the TLR-mediated event and other signalling events may be followed by aggregation. Platelets thus use complex tools to mediate a whole range of functions upon sensing danger. By linking the inflammatory and haemostatic platelet response to infection, TLRs play a central role. The extent of the inflammatory response to pathogen clearance is still a debatable issue and is discussed in this short review.


Assuntos
Plaquetas/metabolismo , Imunidade Inata/imunologia , Receptores Toll-Like/imunologia , Humanos , Transdução de Sinais
17.
Transfusion ; 57(1): 166-177, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27807848

RESUMO

BACKGROUND: Red blood cells (RBCs) contain large amounts of iron, and periodic therapeutic phlebotomy is thus the main treatment for hereditary hemochromatosis (HH). However, the donation of therapeutic phlebotomy products from asymptomatic patients for transfusion purposes remains controversial. In this study, we compared the quality of RBCs obtained from HH patients with those of non-HH RBCs, within the allowed 42-day storage period. STUDY DESIGN AND METHODS: RBCs were obtained from HH patient donors and random regular blood donors by whole blood collection. RBCs were stored for up to 42 days, according to national regulations and standard blood bank conditions in France. The following variables were assessed: hematologic and biochemical results, RBC membrane and soluble inflammatory markers, and the proinflammatory potential of HH RBC supernatant toward endothelial cells in an in vitro model. RESULTS: There were no major differences between the two groups in terms of biophysical, biochemical, or soluble immunomodulatory factors. However, we observed small but significant differences in changes in RBC membrane proteins during storage, including increased phosphatidylserine expression and decreased hemolysis in HH compared with normal RBCs. However, there were no differences in terms of bioactivity of soluble immunomodulatory factors in the RBC supernatant during storage between HH and control donors, as determined by their effects on endothelial cells in vitro. CONCLUSIONS: These in vitro studies suggest that RBCs from HH patients appear, while exhibiting subtle differences, to be suitable for transfusion purposes according to currently accepted criteria.


Assuntos
Doadores de Sangue , Preservação de Sangue , Eritrócitos/metabolismo , Hemocromatose/metabolismo , Adulto , Transfusão de Eritrócitos , Feminino , Regulação da Expressão Gênica , Hemólise , Humanos , Masculino , Pessoa de Meia-Idade , Fosfatidilserinas/biossíntese , Fatores de Tempo
18.
Blood ; 124(14): 2173-83, 2014 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-25082876

RESUMO

Mitochondrial DNA (mtDNA) is a highly potent inflammatory trigger and is reportedly found outside the cells in blood in various pathologies. Platelets are abundant in blood where they promote hemostasis. Although lacking a nucleus, platelets contain functional mitochondria. On activation, platelets produce extracellular vesicles known as microparticles. We hypothesized that activated platelets could also release their mitochondria. We show that activated platelets release respiratory-competent mitochondria, both within membrane-encapsulated microparticles and as free organelles. Extracellular mitochondria are found in platelet concentrates used for transfusion and are present at higher levels in those that induced acute reactions (febrile nonhemolytic reactions, skin manifestations, and cardiovascular events) in transfused patients. We establish that the mitochondrion is an endogenous substrate of secreted phospholipase A2 IIA (sPLA2-IIA), a phospholipase otherwise specific for bacteria, likely reflecting the ancestral proteobacteria origin of mitochondria. The hydrolysis of the mitochondrial membrane by sPLA2-IIA yields inflammatory mediators (ie, lysophospholipids, fatty acids, and mtDNA) that promote leukocyte activation. Two-photon microscopy in live transfused animals revealed that extracellular mitochondria interact with neutrophils in vivo, triggering neutrophil adhesion to the endothelial wall. Our findings identify extracellular mitochondria, produced by platelets, at the midpoint of a potent mechanism leading to inflammatory responses.


Assuntos
Plaquetas/metabolismo , Fosfolipases A2 do Grupo II/metabolismo , Inflamação/metabolismo , Mitocôndrias/metabolismo , Animais , DNA Mitocondrial/metabolismo , Endotélio Vascular/metabolismo , Citometria de Fluxo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ativação Plaquetária , Rickettsia prowazekii/metabolismo
19.
Transfusion ; 56(2): 497-504, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26446055

RESUMO

BACKGROUND: Biological response modifiers (BRMs), secreted by platelets (PLTs) during storage, play a role in adverse events (AEs) associated with transfusion. Moreover, mitochondrial DNA (mtDNA) levels in PLT components (PCs) are associated with AEs. In this study we explore whether there is a correlation between pathogenic BRMs and mtDNA levels and whether these markers can be considered predictors of transfusion pathology. STUDY DESIGN AND METHODS: We investigated a series of reported AEs after PC transfusion, combining clinical observations and mathematical modeling systems. RESULTS: mtDNA was consistently released during the first days of PC storage; however, mtDNA release was earlier in "pathogenic" than in nonpathogenic PCs. PC supernatants with high levels of mtDNA along with soluble CD40 ligand (sCD40L) were significantly associated with occurrences of AEs. The fact that mtDNA did not associate with the 14 BRMs tested suggests the role of mtDNA in PC transfusion-linked inflammation is independent of that of BRMs, known to be associated with AEs. We present evidence that PLTs generate distinct pathogenic secretion profiles of BRMs and mtDNA. The calculated area under the curve for mtDNA was significantly associated with AEs, although less stringently predictive than those of sCD40L or interleukin-13, standard predictors of AE. The established model predicts that distinct subtypes of AEs can be distinguished, dependent on mtDNA levels and PC storage length. CONCLUSIONS: Further work should be considered to test the propensity of mtDNA in PLT concentrates to generate inflammation and cause an AE.


Assuntos
Plaquetas/metabolismo , Preservação de Sangue/efeitos adversos , Ligante de CD40/metabolismo , DNA Mitocondrial/metabolismo , Interleucina-13/metabolismo , Transfusão de Plaquetas/efeitos adversos , Feminino , Humanos , Masculino , Fatores de Tempo
20.
BMC Immunol ; 16: 26, 2015 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-25913718

RESUMO

Blood platelets are first aimed at ensuring primary hemostasis. Beyond this role, they have been acknowledged as having functions in the maintenance of the vascular arborescence and, more recently, as being also innate immune cells, devoted notably to the detection of danger signals, of which infectious ones. Platelets express pathogen recognition receptors that can sense bacterial and viral moieties. Besides, several molecules that bind epithelial or sub-endothelial molecules and, so forth, are involved in hemostasis, happen to be able to ligate viral determinants, making platelets capable of either binding viruses or even to be infected by some of them. Further, as platelets express both Fc-receptors for Ig and complement receptors, they also bind occasionally virus-Ig or virus-Ig-complement immune complexes. Interplays of viruses with platelets are very complex and viral infections often interfere with platelet number and functions. Through a few instances of viral infections, the present review aims at presenting some of the most important interactions from pathophysiological and clinical points of view, which are observed between human viruses and platelets.


Assuntos
Plaquetas/imunologia , Viroses/imunologia , Vírus/imunologia , Animais , Plaquetas/virologia , Interações Hospedeiro-Patógeno , Humanos , Imunidade Inata , Ligação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA