Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
1.
Transfusion ; 63(10): 1937-1950, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37615493

RESUMEN

BACKGROUND: Pathogen reduction of platelet concentrates (PCs) using amotosalen and broad-spectrum UVA illumination contributes to the safety of platelet transfusion by reducing the risk of transfusion-transmitted infections. We evaluated the in vitro quality of stored buffy-coat (BC) PCs treated with amotosalen and a prototype light-emitting diode (LED) illuminator. METHODS: Double-dose BC-PCs collected into PAS-III/plasma or SSP+ /plasma (55/45%) were treated with amotosalen in combination with either conventional UVA lamps (INT100 Illuminator 320-400 nm) or LED illuminators at 350 nm. Platelet quality and function were evaluated over 7 days. RESULTS: Platelet counts were conserved during storage in all groups, as was platelet swirling without appearance of macroscopic aggregates. Integrin αIIbß3 and glycoprotein (GP) VI expression remained stable, whereas GPIbα and GPV declined similarly in all groups. UV lamp- and LED-treated PCs displayed similar glucose consumption, lactate generation, and pH variation. Comparable spontaneous and residual P-selectin and phosphatidylserine exposure, activated αIIbß3 exposure, mitochondrial membrane potential, lactate dehydrogenase release, and adhesive properties under flow conditions were observed during storage. The use of SSP+ /plasma compared with PAS-III/plasma better preserved most of these parameters, especially during late storage, irrespective of the type of illuminator. CONCLUSION: Replacing the UVA lamp for photochemical treatment by LED illuminators had no impact on platelet metabolism, spontaneous activation, apoptosis or viability, or on the in vitro function of BC-PCs stored for 7 days in SSP+ or PAS-III/plasma. These findings support improved procedures for the pathogen reduction and storage of PCs, to ensure transfusion safety and retention of platelet functional properties.


Asunto(s)
Furocumarinas , Rayos Ultravioleta , Humanos , Furocumarinas/farmacología , Plaquetas/metabolismo , Transfusión de Plaquetas , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Conservación de la Sangre/métodos
2.
Basic Res Cardiol ; 117(1): 16, 2022 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-35353230

RESUMEN

Emergency hematopoiesis is the driving force of the inflammatory response to myocardial infarction (MI). Increased proliferation of hematopoietic stem and progenitor cells (LSK) after MI enhances cell production in the bone marrow (BM) and replenishes leukocyte supply for local cell recruitment to the infarct. Decoding the regulation of the inflammatory cascade after MI may provide new avenues to improve post-MI remodeling. In this study, we describe the influence of adenosine diphosphate (ADP)-dependent P2Y12-mediated signaling on emergency hematopoiesis and cardiac remodeling after MI. Permanent coronary ligation was performed to induce MI in a murine model. BM activation, inflammatory cell composition and cardiac function were assessed using global and platelet-specific gene knockout and pharmacological inhibition models for P2Y12. Complementary in vitro studies allowed for investigation of ADP-dependent effects on LSK cells. We found that ADP acts as a danger signal for the hematopoietic BM and fosters emergency hematopoiesis by promoting Akt phosphorylation and cell cycle progression. We were able to detect P2Y12 in LSK, implicating a direct effect of ADP on LSK via P2Y12 signaling. P2Y12 knockout and P2Y12 inhibitor treatment with prasugrel reduced emergency hematopoiesis and the excessive inflammatory response to MI, translating to lower numbers of downstream progeny and inflammatory cells in the blood and infarct. Ultimately, P2Y12 inhibition preserved cardiac function and reduced chronic adverse cardiac remodeling after MI. P2Y12-dependent signaling is involved in emergency hematopoiesis after MI and fuels post-ischemic inflammation, proposing a novel, non-canonical value for P2Y12 antagonists beyond inhibition of platelet-mediated atherothrombosis.


Asunto(s)
Infarto del Miocardio , Animales , Hematopoyesis , Leucocitos , Ratones , Ratones Endogámicos C57BL , Infarto del Miocardio/metabolismo , Células Madre/metabolismo
3.
Transfusion ; 61(3): 919-930, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33527430

RESUMEN

BACKGROUND: Deterioration in quality of platelet concentrates (PCs) during storage results from the appearance of storage lesions affecting the hemostatic functions and posttransfusion survival of platelets. These lesions depend on the preparation and pathogen inactivation methods used, duration of storage, and platelet additive solutions (PASs) present in storage bags. METHODS: We investigated the effects of citrate contained in third-generation PAS (PAS-III) on storage lesions in buffy-coat PCs with or without photochemical (amotosalen-ultraviolet A) treatment over 7 days. RESULTS: Platelet counts were conserved in all groups during storage, as was platelet swirling without appearance of macroscopic aggregates. Glycoprotein (GP) IIbIIIa and GPVI expression remained stable, whereas GPIbα declined similarly in all groups during storage. Removal of citrate from PAS-III, resulting in global reduction of citrate from 11 to 5 mM, led to a significant decrease in glucose consumption, which largely countered a modest deleterious effect of photochemical treatment. Citrate reduction also resulted in decreased lactate generation and better maintenance of pH during storage, while photochemical treatment had no impact on these parameters. Moreover, citrate-free storage significantly reduced exposure of P-selectin and the apoptosis signal phosphatidylserine, thereby abolishing the activating effect of photochemical treatment on both parameters. Citrate reduction benefited platelet aggregation to various agonists up to Day 7, whereas PCT had no impact on these responses. CONCLUSION: Removal of citrate from PAS-III has a beneficial impact on platelet metabolism, spontaneous activation, and apoptosis, and improves platelet aggregation, irrespective of photochemical treatment, which should allow transfusion of platelets with better and longer-lasting functional properties.


Asunto(s)
Plaquetas/metabolismo , Conservación de la Sangre/métodos , Ácido Cítrico/farmacología , Agregación Plaquetaria/efectos de los fármacos , Apoptosis/efectos de los fármacos , Furocumarinas/farmacología , Hemostasis/efectos de los fármacos , Humanos , Concentración de Iones de Hidrógeno , Ácido Láctico/metabolismo , Selectina-P/metabolismo , Fosfatidilserinas , Recuento de Plaquetas , Pruebas de Función Plaquetaria , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Glicoproteínas de Membrana Plaquetaria/metabolismo
4.
Arterioscler Thromb Vasc Biol ; 38(9): 2041-2053, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30354258

RESUMEN

Objective- PI3Kα (phosphoinositide 3-kinase alpha) is a therapeutic target in oncology, but its role in platelets and thrombosis remains ill characterized. In this study, we have analyzed the role of PI3Kα in vitro, ex vivo, and in vivo in 2 models of arterial thrombosis. Approach and Results- Using mice selectively deficient in p110α in the megakaryocyte lineage and isoform-selective inhibitors, we confirm that PI3Kα is not mandatory but participates to thrombus growth over a collagen matrix at arterial shear rate. Our data uncover a role for PI3Kα in low-level activation of the GP (glycoprotein) VI-collagen receptor by contributing to ADP secretion and in turn full activation of PI3Kß and Akt/PKB (protein kinase B). This effect was no longer observed at high level of GP VI agonist concentration. Our study also reveals that over a vWF (von Willebrand factor) matrix, PI3Kα regulates platelet stationary adhesion contacts under arterial flow through its involvement in the outside-in signaling of vWF-engaged αIIbß3 integrin. In vivo, absence or inhibition of PI3Kα resulted in a modest but significant decrease in thrombus size after superficial injuries of mouse mesenteric arteries and an increased time to arterial occlusion after carotid lesion, without modification in the tail bleeding time. Considering the more discrete and nonredundant role of PI3Kα compared with PI3Kß, selective PI3Kα inhibitors are unlikely to increase the bleeding risk at least in the absence of combination with antiplatelet drugs or thrombopenia. Conclusions- This study provides mechanistic insight into the role of PI3Kα in platelet activation and arterial thrombosis.


Asunto(s)
Hemostasis , Fosfatidilinositol 3-Quinasa/fisiología , Adhesividad Plaquetaria , Agregación Plaquetaria , Trombosis/fisiopatología , Animales , Activación Enzimática , Femenino , Humanos , Masculino , Ratones , Inhibidores de las Quinasa Fosfoinosítidos-3 , Glicoproteínas de Membrana Plaquetaria/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Factor de von Willebrand/metabolismo
6.
Blood ; 125(5): 881-8, 2015 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-25398937

RESUMEN

Class IA phosphoinositide 3-kinase ß (PI3Kß) is considered a potential drug target in arterial thrombosis, which is a major cause of death worldwide. Here we show that a striking phenotype of mice with selective p110ß deletion in the megakaryocyte lineage is thrombus instability at a high shear rate, which is an effect that is not detected in the absence of p110α in platelets. The high shear rate-dependent thrombus instability in the absence of p110ß is observed both ex vivo and in vivo with the formation of platelet emboli. Moreover, PI3Kß is required for the recruitment of new platelets to a growing thrombus when a pathological high shear is applied. Treatment of human blood with AZD6482, a selective PI3Kß inhibitor, phenocopies p110ß deletion in mouse platelets, which highlights the role of the kinase activity of p110ß. Within the growing platelet thrombus, p110ß inactivation impairs the activating phosphorylations of Akt and the inhibitory phosphorylation of GSK3. In accord with these data, pharmacologic inhibition of GSK3 restores thrombus stability. Thus, platelet PI3Kß is not essential for thrombus growth and stability at normal arterial shear but has a specific and critical role in maintaining the integrity of the formed thrombus on elevation of shear rate, suggesting a potential risk of embolization on treatment with PI3Kß inhibitors.


Asunto(s)
Plaquetas/metabolismo , Fosfatidilinositol 3-Quinasa Clase I/genética , Glucógeno Sintasa Quinasa 3/genética , Trombosis/genética , Animales , Plaquetas/efectos de los fármacos , Plaquetas/patología , Fosfatidilinositol 3-Quinasa Clase I/antagonistas & inhibidores , Fosfatidilinositol 3-Quinasa Clase I/deficiencia , Regulación de la Expresión Génica , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/metabolismo , Humanos , Mecanotransducción Celular , Ratones , Ratones Transgénicos , Fosforilación/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Piridinas/farmacología , Pirimidinas/farmacología , Pirimidinonas/farmacología , Estrés Mecánico , Trombosis/enzimología , Trombosis/patología , ortoaminobenzoatos/farmacología
7.
Blood ; 126(6): 817-24, 2015 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-25931587

RESUMEN

The mechanism of action of the widely used in vivo ferric chloride (FeCl3) thrombosis model remains poorly understood; although endothelial cell denudation is historically cited, a recent study refutes this and implicates a role for erythrocytes. Given the complexity of the in vivo environment, an in vitro reductionist approach is required to systematically isolate and analyze the biochemical, mass transfer, and biological phenomena that govern the system. To this end, we designed an "endothelial-ized" microfluidic device to introduce controlled FeCl3 concentrations to the molecular and cellular components of blood and vasculature. FeCl3 induces aggregation of all plasma proteins and blood cells, independent of endothelial cells, by colloidal chemistry principles: initial aggregation is due to binding of negatively charged blood components to positively charged iron, independent of biological receptor/ligand interactions. Full occlusion of the microchannel proceeds by conventional pathways, and can be attenuated by antithrombotic agents and loss-of-function proteins (as in IL4-R/Iba mice). As elevated FeCl3 concentrations overcome protective effects, the overlap between charge-based aggregation and clotting is a function of mass transfer. Our physiologically relevant in vitro system allows us to discern the multifaceted mechanism of FeCl3-induced thrombosis, thereby reconciling literature findings and cautioning researchers in using the FeCl3 model.


Asunto(s)
Plaquetas/efectos de los fármacos , Cloruros/farmacología , Eritrocitos/efectos de los fármacos , Compuestos Férricos/farmacología , Agregado de Proteínas/efectos de los fármacos , Aspirina/farmacología , Fenómenos Biomecánicos , Plaquetas/química , Plaquetas/citología , Agregación Celular/efectos de los fármacos , Cloruros/antagonistas & inhibidores , Cloruros/química , Eritrocitos/química , Eritrocitos/citología , Compuestos Férricos/antagonistas & inhibidores , Compuestos Férricos/química , Fibrinolíticos/farmacología , Heparina/farmacología , Humanos , Técnicas Analíticas Microfluídicas , Modelos Biológicos , Plasma Rico en Plaquetas/química , Cultivo Primario de Células , Unión Proteica , Electricidad Estática , Trombosis/metabolismo , Trombosis/patología
8.
J Immunol ; 194(2): 739-49, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25480563

RESUMEN

Extracellular ATP is becoming increasingly recognized as an important regulator of inflammation. However, the known repertoire of P2 receptor subtypes responsible for the proinflammatory effects of ATP is sparse. We looked at whether the P2X1 receptor, an ATP-gated cation channel present on platelets, neutrophils, and macrophages, participates in the acute systemic inflammation provoked by LPS. Compared with wild-type (WT) mice, P2X1(-/-) mice displayed strongly diminished pathological responses, with dampened neutrophil accumulation in the lungs, less tissue damage, reduced activation of coagulation, and resistance to LPS-induced death. P2X1 receptor deficiency also was associated with a marked reduction in plasma levels of the main proinflammatory cytokines and chemokines induced by LPS. Interestingly, macrophages and neutrophils isolated from WT and P2X1(-/-) mice produced similar levels of proinflammatory cytokines when stimulated with LPS in vitro. Intravital microscopy revealed a defect in LPS-induced neutrophil emigration from cremaster venules into the tissues of P2X1(-/-) mice. Using adoptive transfer of immunofluorescently labeled neutrophils from WT and P2X1(-/-) mice into WT mice, we demonstrate that the absence of the P2X1 receptor on neutrophils was responsible for this defect. This study reveals a major role for the P2X1 receptor in LPS-induced lethal endotoxemia through its critical involvement in neutrophil emigration from venules.


Asunto(s)
Endotoxemia/inmunología , Lipopolisacáridos/toxicidad , Pulmón/inmunología , Infiltración Neutrófila/inmunología , Neutrófilos/inmunología , Receptores Purinérgicos P2X1/inmunología , Animales , Coagulación Sanguínea/efectos de los fármacos , Coagulación Sanguínea/genética , Coagulación Sanguínea/inmunología , Endotoxemia/inducido químicamente , Endotoxemia/genética , Endotoxemia/patología , Pulmón/patología , Macrófagos/inmunología , Macrófagos/patología , Ratones , Ratones Noqueados , Infiltración Neutrófila/efectos de los fármacos , Infiltración Neutrófila/genética , Neutrófilos/patología , Receptores Purinérgicos P2X1/genética
10.
Transfusion ; 56(9): 2239-47, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27250038

RESUMEN

BACKGROUND: The INTERCEPT Blood System (IBS) using amotosalen-HCl and ultraviolet (UV)A inactivates a large spectrum of microbial pathogens and white blood cells in therapeutic plasma. Our aim was to evaluate to what extent IBS modifies the capacity of plasma to generate thrombin and induces qualitative or quantitative modifications of plasma proteins. STUDY DESIGN AND METHODS: Plasma units from four donors were collected by apheresis. Samples were taken before (control [CTRL]) and after IBS treatment and stored at -80°C until use. The activities of plasma coagulation factors and inhibitors and the thrombin generation potential were determined using assays measuring clotting times and the calibrated automated thrombogram (CAT), respectively. The proteomic profile of plasma proteins was examined using a two-dimensional differential in-gel electrophoresis (2D-DIGE) method. RESULTS: Nearly all of the procoagulant and antithrombotic factors tested retained at least 78% of their initial pre-IBS activity. Only FVII and FVIII displayed a lower level of conservation (67%), which nevertheless remained within the reference range for conventional plasma coagulation factors. The thrombin generation profile of plasma was conserved after IBS treatment. Among the 1331 protein spots revealed by 2D-DIGE analysis, only four were differentially expressed in IBS plasma compared to CTRL plasma and two were identified by mass spectrometric analysis as transthyretin and apolipoprotein A1. CONCLUSION: The IBS technique for plasma moderately decreases the activities of plasma coagulation factors and antithrombotic proteins, with no impact on the thrombin generation potential of plasma and very limited modifications of the proteomic profile.


Asunto(s)
Conservación de la Sangre/métodos , Furocumarinas/farmacología , Plasma/química , Factores de Coagulación Sanguínea/análisis , Proteínas Sanguíneas/química , Proteínas Sanguíneas/efectos de los fármacos , Humanos , Proteómica/métodos , Rayos Ultravioleta
11.
Arterioscler Thromb Vasc Biol ; 35(11): 2307-15, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26359511

RESUMEN

Under various pathological conditions, including thrombosis and inflammation, extracellular nucleotide levels may increase because of both active release and passive leakage from damaged or dying cells. Once in the extracellular compartment, nucleotides interact with plasma membrane receptors belonging to the P2 purinergic family, which are expressed by virtually all circulating blood cells and in most blood vessels. In this review, we focus on the specific role of the 3 platelet P2 receptors P2Y1, P2Y12, and P2X1 in hemostasis and arterial thrombosis. Beyond platelets, these 3 receptors, along with the P2Y2, P2Y6, and P2X7 receptors, constitute the main P2 receptors mediating the proinflammatory effects of nucleotides, which play important roles in various functions of circulating blood cells and cells of the vessel wall. Each of these P2 receptor subtypes specifically contributes to chronic or acute vascular inflammation and related diseases, such as atherosclerosis, restenosis, endotoxemia, and sepsis. The potential for therapeutic targeting of these P2 receptor subtypes is also discussed.


Asunto(s)
Inflamación/metabolismo , Purinas/metabolismo , Receptores Purinérgicos P2/metabolismo , Trombosis/metabolismo , Animales , Antiinflamatorios/uso terapéutico , Coagulación Sanguínea , Fibrinolíticos/uso terapéutico , Humanos , Inflamación/sangre , Inflamación/tratamiento farmacológico , Mediadores de Inflamación/sangre , Agonistas del Receptor Purinérgico P2/uso terapéutico , Antagonistas del Receptor Purinérgico P2/uso terapéutico , Receptores Purinérgicos P2/efectos de los fármacos , Receptores Purinérgicos P2X7/metabolismo , Receptores Purinérgicos P2Y2/metabolismo , Transducción de Señal , Trombosis/sangre , Trombosis/tratamiento farmacológico
12.
Circulation ; 128(5): 541-52, 2013 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-23797810

RESUMEN

BACKGROUND: Laminins are major components of basement membranes, well located to interact with platelets upon vascular injury. Laminin-111 (α1ß1γ1) is known to support platelet adhesion but is absent from most blood vessels, which contain isoforms with the α2, α4, or α5 chain. Whether vascular laminins support platelet adhesion and activation and the significance of these interactions in hemostasis and thrombosis remain unknown. METHODS AND RESULTS: Using an in vitro flow assay, we show that laminin-411 (α4ß1γ1), laminin-511 (α5ß1γ1), and laminin-521 (α5ß2γ1), but not laminin-211 (α2ß1γ1), allow efficient platelet adhesion and activation across a wide range of arterial wall shear rates. Adhesion was critically dependent on integrin α6ß1 and the glycoprotein Ib-IX complex, which binds to plasmatic von Willebrand factor adsorbed on laminins. Glycoprotein VI did not participate in the adhesive process but mediated platelet activation induced by α5-containing laminins. To address the significance of platelet/laminin interactions in vivo, we developed a platelet-specific knockout of integrin α6. Platelets from these mice failed to adhere to laminin-411, laminin-511, and laminin-521 but responded normally to a series of agonists. α6ß1-Deficient mice presented a marked decrease in arterial thrombosis in 3 models of injury of the carotid, aorta, and mesenteric arterioles. The tail bleeding time and blood loss remained unaltered, indicating normal hemostasis. CONCLUSIONS: This study reveals an unsuspected important contribution of laminins to thrombus formation in vivo and suggests that targeting their main receptor, integrin α6ß1, could represent an alternative antithrombotic strategy with a potentially low bleeding risk.


Asunto(s)
Adhesión Celular/fisiología , Integrina alfa6beta1/metabolismo , Activación Plaquetaria/fisiología , Adhesividad Plaquetaria/fisiología , Trombosis/metabolismo , Animales , Aorta/metabolismo , Aorta/patología , Arterias Carótidas/metabolismo , Arterias Carótidas/patología , Humanos , Integrina alfa6beta1/fisiología , Laminina/fisiología , Arterias Mesentéricas/metabolismo , Arterias Mesentéricas/patología , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Factores de Riesgo , Trombosis/patología
13.
Arterioscler Thromb Vasc Biol ; 33(6): 1221-9, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23559635

RESUMEN

OBJECTIVE: The glycoprotein (GP) Ib-V-IX complex regulates the adhesion, activation, and procoagulant activity of platelets. We previously reported that RAM.1, a rat monoclonal antibody directed against the extracellular domain of mouse GPIbß, diminished adhesion of platelets and chinese hamster ovary cells transfected with the human GPIb-IX complex to von Willebrand factor under flow conditions. Here, we further evaluated the functional importance of GPIbß by studying the impact of RAM.1 on GPIb-mediated platelet responses and in vitro and in vivo thrombus formation. APPROACH AND RESULTS: We show that RAM.1 dramatically reduced GPIb-mediated filopodia extension of chinese hamster ovary GPIb-IX cells after adhesion to von Willebrand factor. RAM.1 also reduced filopodia extension and GPIb-mediated Ca(2+) signaling after adhesion of mouse platelets to von Willebrand factor. RAM.1 inhibited thrombin generation in platelet-rich plasma without impairing phosphatidylserine exposure. In addition, RAM.1 reduced thrombus formation after perfusion of mouse whole blood over collagen in a shear-dependent manner. This effect was confirmed in vivo, because injection of F(ab)'2 fragments of RAM.1 diminished thrombus formation induced by laser beam injury of mesenteric arterioles and forceps injury of the abdominal aorta. In contrast, RAM.1 F(ab)'2 did not prolong the tail-bleeding time or increase the volume of blood lost. CONCLUSIONS: These findings are the first evidence that targeting a subunit other than GPIbα can lead to an antithrombotic effect via the GPIb-V-IX complex. This could represent an alternative way to reduce thrombus formation with a minor impact on hemostasis.


Asunto(s)
Arteriopatías Oclusivas/prevención & control , Adhesividad Plaquetaria/fisiología , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo III/metabolismo , Trombosis/prevención & control , Factor de von Willebrand/metabolismo , Animales , Arteriopatías Oclusivas/fisiopatología , Tiempo de Sangría , Adhesión Celular/fisiología , Cricetinae , Modelos Animales de Enfermedad , Humanos , Ratones , Adhesividad Plaquetaria/genética , Complejo GPIb-IX de Glicoproteína Plaquetaria/genética , Distribución Aleatoria , Ratas , Sensibilidad y Especificidad , Transducción de Señal , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo III/genética , Trombosis/fisiopatología
14.
Transfusion ; 53(6): 1187-200, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23067365

RESUMEN

BACKGROUND: Platelet concentrate (PC) functionality decreases during storage. This is referred to as the storage lesion. Pathogen inactivation may accelerate or induce lesions, potentially accounting for reduced viability. Our aim was to characterize functional and biochemical properties of platelets (PLTs) from photochemically treated buffy-coat PCs (PCT-PCs) compared to those from conventional PCs. STUDY DESIGN AND METHODS: Four PCT-PCs and four conventional PCs were stored for 6.5 days and PLT function and proteomic profiles were examined at various time points during storage. To evaluate their intrinsic properties, samples of stored PLTs were taken, washed, and suspended in Tyrode's buffer before testing. RESULTS: PLT counts and morphology were conserved although a slight increase in the PLT volume was observed after PCT. Glycoprotein (GP) IIbIIIa, IaIIa, and VI expression remained stable while GPIbα declined similarly in both types of PCs. A steep decrease (50%) in GPV occurred on Day 1.5 in PCT-PCs and Day 2.5 in control PCs. For both PCT- and control PCs, P-selectin expression and activated GPIIbIIIa remained low during storage. PCT- and control PCs were fully responsive to aggregation agonists up to Day 4.5 and exhibited similar perfusion functionality. Mitochondrial membrane potential and annexin A5 binding of PCT-PCs and control PCs were comparable. Two-dimensional differential in-gel electrophoresis and mass spectrometry profiles for 1882 protein spots revealed only three proteins selectively changed in PCT-PCs compared to control-PCs. CONCLUSION: Washed treated and untreated PCs have similar functional, morphologic, and proteomic characteristics provided that PLTs are suspended in an appropriate medium during testing.


Asunto(s)
Plaquetas/citología , Conservación de la Sangre/métodos , Seguridad de la Sangre/métodos , Patógenos Transmitidos por la Sangre/efectos de la radiación , Furocumarinas/farmacología , Rayos Ultravioleta , Anexina A5/metabolismo , Almacenamiento de Sangre/métodos , Capa Leucocitaria de la Sangre/citología , Capa Leucocitaria de la Sangre/microbiología , Plaquetas/metabolismo , Plaquetas/microbiología , Criopreservación/métodos , Humanos , Potencial de la Membrana Mitocondrial , Selectina-P/metabolismo , Fármacos Fotosensibilizantes/farmacología , Adhesividad Plaquetaria , Agregación Plaquetaria , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Transfusión de Plaquetas , Proteómica
15.
Thromb Res ; 221: 137-148, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36376109

RESUMEN

Severe COVID-19 has been associated with a high rate of thrombotic events but also of bleeding events, particularly when the level of prophylactic anticoagulation was increased. Data on the contribution of platelets to these thrombotic events are discordant between reports, while the involvement of platelets in bleeding events has never been investigated. The objective of the present study was to assess platelet function during the first week of ICU hospitalization in patients with severe COVID-19 pneumonia. A total of 35 patients were prospectively included and blood samples were drawn on day (D) 0, D2 and D7. COVID-19 pneumonia was severe with a median PaO2/FiO2 ratio of 91 [68-119] on D0. Platelets from these patients showed evidence of pre-activation and exhaustion with a significant reduction in the surface expression of GPVI, GPIb and GPIIbIIIa, together with a decrease in serotonin content. Platelets from patients with severe COVID-19 were hyporesponsive with a reduced maximal aggregation response to several platelet agonists and decreased adhesion to immobilized fibrinogen. Aggregation of washed platelets and plasma substitution experiments indicated that a plasma factor was at least partially responsible for this hyporeactivity of platelets. Blood flow experiments showed that severe COVID-19 platelets formed smaller, less stable aggregates on a collagen-coated surface, which could explain why some patients develop bleeding events. These findings should prompt us to carefully evaluate the risks and benefits of high-dose prophylactic anticoagulation, and to decrease the level of anticoagulation once the initial phase of the disease has resolved. TRIAL REGISTRATION: ClinicalTrials.gov identifier: NCT04359992.


Asunto(s)
COVID-19 , Trombosis , Humanos , Anticoagulantes/metabolismo , Coagulación Sanguínea , Plaquetas/metabolismo , COVID-19/complicaciones , Hemorragia/metabolismo , Agregación Plaquetaria , Estudios Prospectivos
16.
Res Pract Thromb Haemost ; 7(1): 100004, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36970741

RESUMEN

Background: Blood platelet Ca2+ stores are regulated by 2 Ca2+-ATPases (SERCA2b and SERCA3). On thrombin stimulation, nicotinic acid adenosine dinucleotide phosphate mobilizes SERCA3-dependent stores, inducing early adenosine 5'-diphosphate (ADP) secretion, potentiating later SERCA2b-dependent secretion. Objectives: The aim of this study was to identify which ADP P2 purinergic receptor (P2Y1 and/or P2Y12) is(are) involved in the amplification of platelet secretion dependent on the SERCA3-dependent Ca2+ mobilization pathway (SERCA3 stores mobilization) as triggered by low concentration of thrombin. Methods: The study used the pharmacologic antagonists MRS2719 and AR-C69931MX, of the P2Y1 and P2Y12, respectively, as well as Serca3 -/- mice and mice exhibiting platelet lineage-specific inactivation of the P2Y1 or P2Y12 genes. Results: We found that in mouse platelets, pharmacological blockade or gene inactivation of P2Y12 but not of P2Y1 led to a marked inhibition of ADP secretion after platelet stimulation with low concentration of thrombin. Likewise, in human platelets, pharmacological inhibition of P2Y12 but not of P2Y1 alters amplification of thrombin-elicited secretion through SERCA2b stores mobilization. Finally, we show that early SERCA3 stores secretion of ADP is a dense granule secretion, based on parallel adenosine triphosphate and serotonin early secretion. Furthermore, early secretion involves a single granule, based on the amount of adenosine triphosphate released. Conclusion: Altogether, these results show that at low concentrations of thrombin, SERCA3- and SERCA2b-dependent Ca2+ mobilization pathways cross-talk via ADP and activation of the P2Y12, and not the P2Y1 ADP receptor. The relevance in hemostasis of the coupling of the SERCA3 and the SERCA2b pathways is reviewed.

17.
Circulation ; 123(21): 2404-13, 2011 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-21576651

RESUMEN

BACKGROUND: Atherosclerosis is an inflammatory disease, and extracellular nucleotides are one of the factors possibly involved in vascular inflammation. The P2Y(1) receptor for adenosine 5'-diphosphate has been shown to be involved in the development of atherosclerosis in apolipoprotein E--deficient mice. Our aim is to determine whether the endothelial P2Y(1) receptor plays a role in leukocyte recruitment during vascular inflammation and characterize underlying mechanisms. METHODS AND RESULTS: We show here that the P2Y(1) receptor plays a role in leukocyte recruitment in inflamed mouse femoral arteries. Moreover, in wild-type bone marrow--transplanted chimeric P2Y(1)-deficient mice with an apolipoprotein E--deficient background, a strong reduction of adhesion molecule--dependent leukocyte recruitment was observed after local injection of tumor necrosis factor and interleukin 1ß, excluding a role for the platelet or other hematopoietic cell type P2Y(1) in these events. Similarly, the in vitro adhesion of isolated mouse monocytes to tumor necrosis factor α--stimulated murine endothelial cell monolayers and their migration across the cell layers were strongly reduced in P2Y(1)-deficient compared with wild-type endothelial cells, as was the expression of the adhesion molecules P-selectin, Vascular cell adhesion molecule 1, and intercellular adhesion molecule 1. Pharmacological inhibition using the selective antagonist MRS2500 also resulted in decreased expression of adhesion molecules. These events are related to the p38 mitogen-activated protein kinase and activating transcription factor 2 pathway. Finally, in vivo administration of MRS2500 resulted in strong reduction of leukocyte recruitment in inflamed femoral arteries of apolipoprotein E--deficient mice. CONCLUSIONS: The data highlight a key role of the endothelial P2Y(1) receptor in acute vascular inflammation. Pharmacological targeting the P2Y(1) receptor could represent a promising approach for the treatment of vascular inflammation.


Asunto(s)
Arteritis/patología , Aterosclerosis/patología , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Mediadores de Inflamación/fisiología , Receptores Purinérgicos P2Y1/fisiología , Factor de Necrosis Tumoral alfa/administración & dosificación , Enfermedad Aguda , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Arteritis/metabolismo , Aterosclerosis/genética , Aterosclerosis/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Movimiento Celular/genética , Células Cultivadas , Endotelio Vascular/citología , Arteria Femoral/metabolismo , Arteria Femoral/patología , Masculino , Venas Mesentéricas/metabolismo , Venas Mesentéricas/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Purinérgicos P2Y1/genética , Quimera por Trasplante , Factor de Necrosis Tumoral alfa/fisiología
18.
J Pharmacol Exp Ther ; 341(1): 156-63, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22238212

RESUMEN

Glycoprotein VI (GPVI) has been proposed as a promising antiplatelet target, because its blockade prevents experimental thrombosis without impairing hemostasis. The objective of this study was to develop a preclinical tool to evaluate the role of human GPVI (hGPVI) in various models of thrombosis and to screen anti-GPVI compounds. A genetically modified mouse strain expressing hGPVI has been developed using a knockin strategy. The mice were viable and fertile and did not present any hematological defects. Approximately 3700 copies of human GPVI were detected at the platelet surface. Platelet aggregation, fibrinogen binding, and P-selectin exposure were normal in response to various agonists. The 9O12.2 Fab fragment directed against human GPVI bound to hGPVI platelets in vitro and ex vivo and markedly reduced collagen- and collagen-related peptide-induced responses. Injection of 9O12.2 into hGPVI animals did not prolong the tail bleeding time but provided protection against lethal thromboembolism induced by a collagen/adrenaline mixture. In addition, 9O12.2 reduced arterial thrombus growth by 44% after superficial laser injury, 43% after deep laser injury in mice pretreated with hirudin, and 48% after mechanical injury. In conclusion, we have developed a humanized mouse model that could be used in preclinical studies to evaluate the effects of anti-GPVI compounds.


Asunto(s)
Modelos Animales de Enfermedad , Fibrinolíticos/uso terapéutico , Glicoproteínas de Membrana Plaquetaria/antagonistas & inhibidores , Glicoproteínas de Membrana Plaquetaria/genética , Trombosis/genética , Trombosis/metabolismo , Animales , Fibrinolíticos/administración & dosificación , Técnicas de Sustitución del Gen , Humanos , Ratones , Ratones Transgénicos , Glicoproteínas de Membrana Plaquetaria/fisiología , Trombosis/tratamiento farmacológico , Resultado del Tratamiento
19.
Blood ; 116(7): 1157-64, 2010 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-20457869

RESUMEN

Activated platelets become procoagulant and efficiently promote the conversion of prothrombin to thrombin. A role of the GPIb-V-IX complex has long been postulated in view of the decreased prothrombin consumption in Bernard-Soulier patients. We evaluated the impact of GPIb-V-IX deficiency and the requirement for the GPIbalpha extracellular domain. In GPIbbeta(-/-) mice, thrombin generation was profoundly decreased in tissue factor- or collagen-related peptide (CRP)-activated platelet-rich plasma and in washed platelets supplemented with normal plasma or with FVa, FXa, and prothrombin. Phosphatidylserine (PS) exposure was similarly decreased in response to thrombin, CRP, or CRP + PAR4 peptide despite a normal platelet phospholipid composition. The hypothesis that these defects originate from lack of the GPIbalpha N-terminal domain was evaluated after its removal from normal mouse and human platelets with Nk protease or O-sialoglycoprotein endopeptidase. Unexpectedly, the treated platelets exhibited normal thrombin generation and PS exposure, indicating that GPIb-V-IX regulates procoagulant activity independently of its GPIbalpha-binding region. These results suggested a more general structuring role through intracellular cytoskeleton-anchoring portions regulating responses leading to PS exposure. This hypothesis was supported by the decreased calcium mobilization observed in GPIbbeta(-/-) platelets in response to several agonists, some acting independently of GPIb, in contrast to the normal calcium responses in Nk protease-treated platelets.


Asunto(s)
Plaquetas/metabolismo , Glicoproteínas de Membrana/metabolismo , Complejo GPIb-IX de Glicoproteína Plaquetaria/fisiología , Animales , Calcio/metabolismo , Citometría de Flujo , Granzimas/metabolismo , Humanos , Ratones , Ratones Noqueados , Fosfatidilserinas/metabolismo , Estructura Terciaria de Proteína , Trombina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA