Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Blood ; 140(24): 2626-2643, 2022 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-36026606

RESUMEN

S100A8/A9, also known as "calprotectin" or "MRP8/14," is an alarmin primarily secreted by activated myeloid cells with antimicrobial, proinflammatory, and prothrombotic properties. Increased plasma levels of S100A8/A9 in thrombo-inflammatory diseases are associated with thrombotic complications. We assessed the presence of S100A8/A9 in the plasma and lung autopsies from patients with COVID-19 and investigated the molecular mechanism by which S100A8/A9 affects platelet function and thrombosis. S100A8/A9 plasma levels were increased in patients with COVID-19 and sustained high levels during hospitalization correlated with poor outcomes. Heterodimeric S100A8/A9 was mainly detected in neutrophils and deposited on the vessel wall in COVID-19 lung autopsies. Immobilization of S100A8/A9 with collagen accelerated the formation of a fibrin-rich network after perfusion of recalcified blood at venous shear. In vitro, platelets adhered and partially spread on S100A8/A9, leading to the formation of distinct populations of either P-selectin or phosphatidylserine (PS)-positive platelets. By using washed platelets, soluble S100A8/A9 induced PS exposure but failed to induce platelet aggregation, despite GPIIb/IIIa activation and alpha-granule secretion. We identified GPIbα as the receptor for S100A8/A9 on platelets inducing the formation of procoagulant platelets with a supporting role for CD36. The effect of S100A8/A9 on platelets was abolished by recombinant GPIbα ectodomain, platelets from a patient with Bernard-Soulier syndrome with GPIb-IX-V deficiency, and platelets from mice deficient in the extracellular domain of GPIbα. We identified the S100A8/A9-GPIbα axis as a novel targetable prothrombotic pathway inducing procoagulant platelets and fibrin formation, in particular in diseases associated with high levels of S100A8/A9, such as COVID-19.


Asunto(s)
Plaquetas , COVID-19 , Calgranulina A , Calgranulina B , Complejo GPIb-IX de Glicoproteína Plaquetaria , Animales , Ratones , Plaquetas/metabolismo , Calgranulina A/metabolismo , COVID-19/metabolismo , Fibrina/metabolismo , Fosfatidilserinas/metabolismo , Agregación Plaquetaria , Humanos , Calgranulina B/metabolismo , Autopsia , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo
3.
Front Immunol ; 15: 1372996, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38817606

RESUMEN

Tissue microenvironments during physiology and pathology are highly complex, meaning dynamic cellular activities and their interactions cannot be accurately modelled ex vivo or in vitro. In particular, tissue-specific resident cells which may function and behave differently after isolation and the heterogenous vascular beds in various organs highlight the importance of observing such processes in real-time in vivo. This challenge gave rise to intravital microscopy (IVM), which was discovered over two centuries ago. From the very early techniques of low-optical resolution brightfield microscopy, limited to transparent tissues, IVM techniques have significantly evolved in recent years. Combined with improved animal surgical preparations, modern IVM technologies have achieved significantly higher speed of image acquisition and enhanced image resolution which allow for the visualisation of biological activities within a wider variety of tissue beds. These advancements have dramatically expanded our understanding in cell migration and function, especially in organs which are not easily accessible, such as the brain. In this review, we will discuss the application of rodent IVM in neurobiology in health and disease. In particular, we will outline the capability and limitations of emerging technologies, including photoacoustic, two- and three-photon imaging for brain IVM. In addition, we will discuss the use of these technologies in the context of neuroinflammation.


Asunto(s)
Encéfalo , Microscopía Intravital , Animales , Microscopía Intravital/métodos , Humanos , Técnicas Fotoacústicas/métodos
4.
J Thromb Haemost ; 22(7): 2018-2026, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38670315

RESUMEN

BACKGROUND: Free labile hemin acts as a damage-associated molecular pattern during acute and chronic hemolysis and muscle injury, supporting platelet activation and thrombosis. OBJECTIVES: To investigate the anti-thrombotic potential of hydroxychloroquine on hemolysis-induced platelet activation and arterial thrombosis. METHODS: The effect of hydroxychloroquine on hemin-induced platelet activation and hemolysis-induced platelet recruitment and aggregation was measured in washed platelets and hemolyzed blood, respectively. Its effect on ferric-chloride (FeCl3)-induced arterial thrombosis and lung perfusion following hemin injection was assessed in wild-type mice. RESULTS: Erythrocyte lysis and endothelial cell activation cooperatively supported platelet aggregation and thrombosis at arterial shear stress. This thrombotic effect was reversed by hydroxychloroquine. In a purified system, hydroxychloroquine inhibited platelet build-up on immobilized von Willebrand factor in hemolyzed blood without altering initial platelet recruitment. Hydroxychloroquine inhibited hemin-induced platelet activation and phosphatidylserine exposure independently of reactive oxygen species generation. In the presence of hemin, hydroxychloroquine did not alter glycoprotein VI shedding but reduced C-type-lectin-like-2 expression on platelets. In vivo, hydroxychloroquine reversed pulmonary perfusion decline induced by exogenous administration of hemin. In arterial thrombosis models, hydroxychloroquine inhibited ferric-chloride-induced thrombosis in the carotid artery and reduced von Willebrand factor accumulation in the thrombi. CONCLUSION: Hydroxychloroquine inhibited hemolysis-induced arterial thrombosis ex vivo and improved pulmonary perfusion in hemin-treated mice, supporting a potential benefit of its use as an adjuvant therapy in hemolytic diseases to limit arterial thrombosis and to improve organ perfusion.


Asunto(s)
Hemina , Hemólisis , Hidroxicloroquina , Pulmón , Activación Plaquetaria , Trombosis , Animales , Hidroxicloroquina/farmacología , Hemólisis/efectos de los fármacos , Hemina/farmacología , Trombosis/tratamiento farmacológico , Trombosis/sangre , Pulmón/efectos de los fármacos , Pulmón/irrigación sanguínea , Activación Plaquetaria/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Agregación Plaquetaria/efectos de los fármacos , Compuestos Férricos , Humanos , Masculino , Cloruros , Modelos Animales de Enfermedad , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Eritrocitos/efectos de los fármacos , Eritrocitos/metabolismo , Factor de von Willebrand/metabolismo
5.
Front Immunol ; 14: 1226196, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37622117

RESUMEN

Background: The NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3) inflammasome has been described in both immune cells and platelets, but its role in the megakaryocyte (MK) lineage remains elusive. Objective: The aim of this study was to explore the role of NLRP3 inflammasome in megakaryocytes and platelets. Methods: We generated Nlrp3 A350V/+/Gp1ba-CreKI/+ mice carrying a mutation genetically similar to the one observed in human Muckle-Wells syndrome, which leads to hyperactivity of NLRP3 specifically in MK and platelets. Results: Platelets from the mutant mice expressed elevated levels of both precursor and active form of caspase-1, suggesting hyperactivity of NLRP3 inflammasome. Nlrp3 A350V/+/Gp1ba-CreKI/+ mice developed normally and had normal platelet counts. Expression of major platelet receptors, platelet aggregation, platelet deposition on collagen under shear, and deep vein thrombosis were unchanged. Nlrp3 A350V/+/Gp1ba-CreKI/+ mice had mild anemia, reduced Ter119+ cells in the bone marrow, and splenomegaly. A mild increase in MK TGF-ß1 might be involved in the anemic phenotype. Intraperitoneal injection of zymosan in Nlrp3 A350V/+/Gp1ba-CreKI/+ mice induced increased neutrophil egression and elevated levels of a set of proinflammatory cytokines, alongside IL-10 and G-CSF, in the peritoneal fluid as compared with control animals. Conclusion: MK/platelet NLRP3 inflammasome promotes the acute inflammatory response and its hyperactivation in mice leads to mild anemia and increased extramedullary erythropoiesis.


Asunto(s)
Anemia , Megacariocitos , Proteína con Dominio Pirina 3 de la Familia NLR , Animales , Ratones , Inflamasomas , Ratones Endogámicos NOD , Proteína con Dominio Pirina 3 de la Familia NLR/genética
6.
Methods Mol Biol ; 2441: 287-295, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35099745

RESUMEN

Upon injury, stable thrombi formation requires the recruitment of platelets, leukocytes, and various clotting factors, to provide sufficient inhibition of hemostasis. Classical models of thrombosis involve either ex vivo isolation of platelets and subsequent quantification of aggregation through light transmission aggregometry or in vivo murine intravital thrombosis models (laser injury, ferric chloride, or rose Bengal). Flow adhesion models allow for accurate quantification of the contribution of cell-types to thrombi formation. Here, we describe the use of flow chambers to flow human blood over activated endothelial cells to observe leukocyte-endothelial adhesion at arterial and venous shear rates.


Asunto(s)
Células Endoteliales , Trombosis , Animales , Plaquetas/metabolismo , Comunicación Celular , Humanos , Leucocitos/fisiología , Ratones , Trombosis/metabolismo
7.
Thromb Haemost ; 122(12): 1988-2000, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35817083

RESUMEN

C-type lectin-like receptor 2 (CLEC-2) is highly expressed on platelets and a subpopulation of myeloid cells, and is critical in lymphatic development. CLEC-2 has been shown to support thrombus formation at sites of inflammation, but to have a minor/negligible role in hemostasis. This identifies CLEC-2 as a promising therapeutic target in thromboinflammatory disorders, without hemostatic detriment. We utilized a GPIbα-Cre recombinase mouse for more restricted deletion of platelet-CLEC-2 than the previously used PF4-Cre mouse. clec1bfl/flGPIbα-Cre+ mice are born at a Mendelian ratio, with a mild reduction in platelet count, and present with reduced thrombus size post-FeCl3-induced thrombosis, compared to littermates. Antibody-mediated depletion of platelet count in C57BL/6 mice, to match clec1bfl/flGPIbα-Cre+ mice, revealed that the reduced thrombus size post-FeCl3-injury was due to the loss of CLEC-2, and not mild thrombocytopenia. Similarly, clec1bfl/flGPIbα-Cre+ mouse blood replenished with CLEC-2-deficient platelets ex vivo to match littermates had reduced aggregate formation when perfused over collagen at arterial flow rates. In contrast, platelet-rich thrombi formed following perfusion of human blood under flow conditions over collagen types I or III, atherosclerotic plaque, or inflammatory endothelial cells were unaltered in the presence of CLEC-2-blocking antibody, AYP1, or recombinant CLEC-2-Fc. The reduction in platelet aggregation observed in clec1bfl/flGPIbα-Cre+ mice during arterial thrombosis is mediated by the loss of CLEC-2 on mouse platelets. In contrast, CLEC-2 does not support thrombus generation on collagen, atherosclerotic plaque, or inflamed endothelial cells in human at arterial shear.


Asunto(s)
Placa Aterosclerótica , Trombosis , Ratones , Humanos , Animales , Agregación Plaquetaria , Activación Plaquetaria , Células Endoteliales , Ratones Endogámicos C57BL , Plaquetas , Lectinas Tipo C/genética
8.
Cardiovasc Res ; 118(15): 3085-3096, 2022 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-35709328

RESUMEN

AIMS: Thrombotic complications and vasculopathy have been extensively associated with severe COVID-19 infection; however, the mechanisms inducing endotheliitis and the disruption of endothelial integrity in the microcirculation are poorly understood. We hypothesized that within the vessel wall, pericytes preferentially take up viral particles and mediate the subsequent loss of vascular integrity. METHODS AND RESULTS: Immunofluorescence of post-mortem patient sections was used to assess pathophysiological aspects of COVID-19 infection. The effects of COVID-19 on the microvasculature were assessed using a vascular organoid model exposed to live viral particles or recombinant viral antigens. We find increased expression of the viral entry receptor angiotensin-converting enzyme 2 on pericytes when compared to vascular endothelium and a reduction in the expression of the junctional protein CD144, as well as increased cell death, upon treatment with both live virus and/or viral antigens. We observe a dysregulation of genes implicated in vascular permeability, including Notch receptor 3, angiopoietin-2, and TEK. Activation of vascular organoids with interleukin-1ß did not have an additive effect on vascular permeability. Spike antigen was detected in some patients' lung pericytes, which was associated with a decrease in CD144 expression and increased platelet recruitment and von Willebrand factor (VWF) deposition in the capillaries of these patients, with thrombi in large vessels rich in VWF and fibrin. CONCLUSION: Together, our data indicate that direct viral exposure to the microvasculature modelled by organoid infection and viral antigen treatment results in pericyte infection, detachment, damage, and cell death, disrupting pericyte-endothelial cell crosstalk and increasing microvascular endothelial permeability, which can promote thrombotic and bleeding complications in the microcirculation.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , Antígenos Virales
9.
Front Immunol ; 13: 968981, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36225927

RESUMEN

Background: The systemic inflammatory response post-SARS-CoV-2 infection increases pro-inflammatory cytokine production, multi-organ damage, and mortality rates. Mast cells (MC) modulate thrombo-inflammatory disease progression (e.g., deep vein thrombosis) and the inflammatory response post-infection. Objective: To enhance our understanding of the contribution of MC and their proteases in SARS-CoV-2 infection and the pathogenesis of the disease, which might help to identify novel therapeutic targets. Methods: MC proteases chymase (CMA1), carboxypeptidase A3 (CPA3), and tryptase beta 2 (TPSB2), as well as cytokine levels, were measured in the serum of 60 patients with SARS-CoV-2 infection (30 moderate and 30 severe; severity of the disease assessed by chest CT) and 17 healthy controls by ELISA. MC number and degranulation were quantified by immunofluorescent staining for tryptase in lung autopsies of patients deceased from either SARS-CoV-2 infection or unrelated reasons (control). Immortalized human FcεR1+c-Kit+ LUVA MC were infected with SARS-CoV-2, or treated with its viral proteins, to assess direct MC activation by flow cytometry. Results: The levels of all three proteases were increased in the serum of patients with COVID-19, and strongly correlated with clinical severity. The density of degranulated MC in COVID-19 lung autopsies was increased compared to control lungs. The total number of released granules and the number of granules per each MC were elevated and positively correlated with von Willebrand factor levels in the lung. SARS-CoV-2 or its viral proteins spike and nucleocapsid did not induce activation or degranulation of LUVA MC in vitro. Conclusion: In this study, we demonstrate that SARS-CoV-2 is strongly associated with activation of MC, which likely occurs indirectly, driven by the inflammatory response. The results suggest that plasma MC protease levels could predict the disease course, and that severe COVID-19 patients might benefit from including MC-stabilizing drugs in the treatment scheme.


Asunto(s)
COVID-19 , Carboxipeptidasas , Quimasas/metabolismo , Citocinas , Humanos , Mastocitos/metabolismo , SARS-CoV-2 , Triptasas/metabolismo , Proteínas Virales , Factor de von Willebrand
10.
Front Immunol ; 12: 693974, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34163489

RESUMEN

Platelets play a key role in the development, progression and resolution of the inflammatory response during sterile inflammation and infection, although the mechanism is not well understood. Here we show that platelet CLEC-2 reduces tissue inflammation by regulating inflammatory macrophage activation and trafficking from the inflamed tissues. The immune regulatory function of CLEC-2 depends on the expression of its ligand, podoplanin, upregulated on inflammatory macrophages and is independent of platelet activation and secretion. Mechanistically, platelet CLEC-2 and also recombinant CLEC-2-Fc accelerates actin rearrangement and macrophage migration by increasing the expression of podoplanin and CD44, and their interaction with the ERM proteins. During ongoing inflammation, induced by lipopolysaccharide, treatment with rCLEC-2-Fc induces the rapid emigration of peritoneal inflammatory macrophages to mesenteric lymph nodes, thus reducing the accumulation of inflammatory macrophages in the inflamed peritoneum. This is associated with a significant decrease in pro-inflammatory cytokine, TNF-α and an increase in levels of immunosuppressive, IL-10 in the peritoneum. Increased podoplanin expression and actin remodelling favour macrophage migration towards CCL21, a soluble ligand for podoplanin and chemoattractant secreted by lymph node lymphatic endothelial cells. Macrophage efflux to draining lymph nodes induces T cell priming. In conclusion, we show that platelet CLEC-2 reduces the inflammatory phenotype of macrophages and their accumulation, leading to diminished tissue inflammation. These immunomodulatory functions of CLEC-2 are a novel strategy to reduce tissue inflammation and could be therapeutically exploited through rCLEC-2-Fc, to limit the progression to chronic inflammation.


Asunto(s)
Plaquetas/metabolismo , Movimiento Celular , Lectinas Tipo C/metabolismo , Activación de Macrófagos , Macrófagos Peritoneales/metabolismo , Peritonitis/metabolismo , Animales , Plaquetas/inmunología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Mediadores de Inflamación/metabolismo , Lectinas Tipo C/genética , Lipopolisacáridos , Macrófagos Peritoneales/inmunología , Masculino , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Peritonitis/genética , Peritonitis/inmunología , Fagocitosis , Fenotipo , Células RAW 264.7 , Transducción de Señal , Linfocitos T/inmunología , Linfocitos T/metabolismo
11.
Res Pract Thromb Haemost ; 4(1): 23-35, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31989082

RESUMEN

Beyond their role in hemostasis and thrombosis, platelets are increasingly recognized as key regulators of the inflammatory response under sterile and infectious conditions. Both platelet receptors and secretion are critical for these functions and contribute to their interaction with the endothelium and innate immune system. Platelet-leukocyte interactions are increased in thrombo-inflammatory diseases and are sensitive biomarkers for platelet activation and targets for the development of new therapies. The crosstalk between platelets and innate immune cells promotes thrombosis, inflammation, and tissue damage. However, recent studies have shown that these interactions also regulate the resolution of inflammation, tissue repair, and wound healing. Many of the platelet and leukocyte receptors involved in these bidirectional interactions are not selective for a subset of immune cells. However, specific heterotypic interactions occur in different vascular beds and inflammatory conditions, raising the possibility of disease- and organ-specific pathways of intervention. In this review, we highlight and discuss prominent and emerging interrelationships between platelets and innate immune cells and their dual role in the regulation of the inflammatory response in sterile and infectious thrombo-inflammatory diseases. A better understanding of the functional relevance of these interactions in different vascular beds may provide opportunities for successful therapeutic interventions to regulate the development, progression, and chronicity of various pathological processes.

12.
Thromb Haemost ; 120(4): 538-564, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32289858

RESUMEN

Thrombo-inflammation describes the complex interplay between blood coagulation and inflammation that plays a critical role in cardiovascular diseases. The third Maastricht Consensus Conference on Thrombosis assembled basic, translational, and clinical scientists to discuss the origin and potential consequences of thrombo-inflammation in the etiology, diagnostics, and management of patients with cardiovascular disease, including myocardial infarction, stroke, and peripheral artery disease. This article presents a state-of-the-art reflection of expert opinions and consensus recommendations regarding the following topics: (1) challenges of the endothelial cell barrier; (2) circulating cells and thrombo-inflammation, focused on platelets, neutrophils, and neutrophil extracellular traps; (3) procoagulant mechanisms; (4) arterial vascular changes in atherogenesis; attenuating atherosclerosis and ischemia/reperfusion injury; (5) management of patients with arterial vascular disease; and (6) pathogenesis of venous thrombosis and late consequences of venous thromboembolism.


Asunto(s)
Aterosclerosis/inmunología , Enfermedades Cardiovasculares/inmunología , Endotelio Vascular/fisiología , Inflamación/inmunología , Neutrófilos/inmunología , Tromboembolia Venosa/inmunología , Animales , Aterosclerosis/diagnóstico , Aterosclerosis/terapia , Coagulación Sanguínea , Enfermedades Cardiovasculares/diagnóstico , Enfermedades Cardiovasculares/terapia , Testimonio de Experto , Humanos , Inmunidad Innata , Trombosis , Tromboembolia Venosa/diagnóstico , Tromboembolia Venosa/terapia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA