Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Blood ; 126(2): 242-6, 2015 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-25979951

RESUMEN

Neutrophil extracellular traps (NETs) can be released in the vasculature. In addition to trapping microbes, they promote inflammatory and thrombotic diseases. Considering that P-selectin induces prothrombotic and proinflammatory signaling, we studied the role of this selectin in NET formation. NET formation (NETosis) was induced by thrombin-activated platelets rosetting with neutrophils and was inhibited by anti-P-selectin aptamer or anti-P-selectin glycoprotein ligand-1 (PSGL-1) inhibitory antibody but was not induced by platelets from P-selectin(-/-) mice. Moreover, NETosis was also promoted by P-selectin-immunoglobulin fusion protein but not by control immunoglobulin. We isolated neutrophils from mice engineered to overproduce soluble P-selectin (P-selectin(ΔCT/ΔCT) mice). Although the levels of circulating DNA and nucleosomes (indicative of spontaneous NETosis) were normal in these mice, basal neutrophil histone citrullination and presence of P-selectin on circulating neutrophils were elevated. NET formation after stimulation with platelet activating factor, ionomycin, or phorbol 12-myristate 13-acetate was significantly enhanced, indicating that the P-selectin(ΔCT/ΔCT) neutrophils were primed for NETosis. In summary, P-selectin, cellular or soluble, through binding to PSGL-1, promotes NETosis, suggesting that this pathway is a potential therapeutic target for NET-related diseases.


Asunto(s)
Trampas Extracelulares/genética , Selectina-P/fisiología , Trombosis/genética , Vasculitis/genética , Animales , Plaquetas/fisiología , Trampas Extracelulares/efectos de los fármacos , Trampas Extracelulares/metabolismo , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Selectina-P/genética , Selectina-P/farmacología , Activación Plaquetaria/genética , Proteínas Recombinantes de Fusión/farmacología , Trombosis/patología , Vasculitis/patología
2.
J Am Soc Nephrol ; 27(1): 120-31, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26038528

RESUMEN

Thrombotic microangiopathy (TMA) is a life-threatening condition that affects some, but not all, recipients of vascular endothelial growth factor (VEGF) inhibitors given as part of chemotherapy. TMA is also a complication of preeclampsia, a disease characterized by excess production of the VEGF-scavenging soluble VEGF receptor 1 (soluble fms-like tyrosine kinase 1; sFlt-1). Risk factors for VEGF inhibitor-related TMA remain unknown. We hypothesized that deficiency of the VWF-cleaving ADAMTS13 endopeptidase contributes to the development of VEGF inhibitor-related TMA. ADAMTS13(-/-) mice overexpressing sFlt-1 presented all hallmarks of TMA, including thrombocytopenia, schistocytosis, anemia, and VWF-positive microthrombi in multiple organs. Similar to VEGF inhibitor-related TMA in humans, these mice exhibited severely impaired kidney function and hypertension. In contrast, wild-type mice overexpressing sFlt-1 developed modest hypertension but no other features of TMA. Recombinant ADAMTS13 therapy ameliorated all symptoms of TMA in ADAMTS13(-/-) mice overexpressing sFlt-1 and normalized BP in wild-type mice. ADAMTS13 activity may thus be a critical determinant for the development of TMA secondary to VEGF inhibition. Administration of recombinant ADAMTS13 may serve as a therapeutic approach to treat or prevent thrombotic complications of VEGF inhibition.


Asunto(s)
Metaloendopeptidasas/fisiología , Microangiopatías Trombóticas/etiología , Microangiopatías Trombóticas/prevención & control , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Proteína ADAMTS13 , Animales , Ratones , Ratones Endogámicos C57BL , Receptor 1 de Factores de Crecimiento Endotelial Vascular/fisiología
3.
Blood ; 121(6): 1008-15, 2013 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-23243271

RESUMEN

The majority of peripheral serotonin is stored in platelets, which secrete it on activation. Serotonin releases Weibel-Palade bodies (WPBs) and we asked whether absence of platelet serotonin affects neutrophil recruitment in inflammatory responses. Tryptophan hydroxylase (Tph)1­deficient mice, lacking non-neuronal serotonin, showed mild leukocytosis compared with wild-type (WT), primarily driven by an elevated neutrophil count. Despite this, 50% fewer leukocytes rolled on unstimulated mesenteric venous endothelium of Tph1(-/-) mice. The velocity of rolling leukocytes was higher in Tph1(-/-) mice, indicating fewer selectin-mediated interactions with endothelium. Stimulation of endothelium with histamine, a secretagogue of WPBs, or injection of serotonin normalized the rolling in Tph1(-/-) mice. Diminished rolling in Tph1(-/-) mice resulted in reduced firm adhesion of leukocytes after lipopolysaccharide treatment. Blocking platelet serotonin uptake with fluoxetine in WT mice reduced serum serotonin by > 80% and similarly reduced leukocyte rolling and adhesion. Four hours after inflammatory stimulation, neutrophil extravasation into lung, peritoneum, and skin wounds was reduced in Tph1(-/-) mice, whereas in vitro neutrophil chemotaxis was independent of serotonin. Survival of lipopolysaccharide-induced endotoxic shock was improved in Tph1(-/-) mice. In conclusion, platelet serotonin promotes the recruitment of neutrophils in acute inflammation, supporting an important role for platelet serotonin in innate immunity.


Asunto(s)
Plaquetas/inmunología , Inflamación/inmunología , Neutrófilos/inmunología , Serotonina/inmunología , Enfermedad Aguda , Animales , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Quimiotaxis/efectos de los fármacos , Quimiotaxis/inmunología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/inmunología , Endotelio Vascular/metabolismo , Citometría de Flujo , Fluoxetina/inmunología , Fluoxetina/farmacología , Histamina/inmunología , Histamina/farmacología , Inflamación/genética , Inflamación/metabolismo , Estimación de Kaplan-Meier , Selectina L/inmunología , Selectina L/metabolismo , Rodamiento de Leucocito/efectos de los fármacos , Rodamiento de Leucocito/genética , Rodamiento de Leucocito/inmunología , Leucocitosis/genética , Leucocitosis/inmunología , Leucocitosis/metabolismo , Lipopolisacáridos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infiltración Neutrófila/inmunología , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Serotonina/sangre , Serotonina/metabolismo , Inhibidores Selectivos de la Recaptación de Serotonina/inmunología , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Choque Séptico/inducido químicamente , Choque Séptico/genética , Choque Séptico/inmunología , Triptófano Hidroxilasa/deficiencia , Triptófano Hidroxilasa/genética , Cuerpos de Weibel-Palade/efectos de los fármacos , Cuerpos de Weibel-Palade/inmunología , Cuerpos de Weibel-Palade/metabolismo
4.
Blood ; 119(5): 1263-73, 2012 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-22101895

RESUMEN

When refrigerated platelets are rewarmed, they secrete active sialidases, including the lysosomal sialidase Neu1, and express surface Neu3 that remove sialic acid from platelet von Willebrand factor receptor (VWFR), specifically the GPIbα subunit. The recovery and circulation of refrigerated platelets is greatly improved by storage in the presence of inhibitors of sialidases. Desialylated VWFR is also a target for metalloproteinases (MPs), because GPIbα and GPV are cleaved from the surface of refrigerated platelets. Receptor shedding is inhibited by the MP inhibitor GM6001 and does not occur in Adam17(ΔZn/ΔZn) platelets expressing inactive ADAM17. Critically, desialylation in the absence of MP-mediated receptor shedding is sufficient to cause the rapid clearance of platelets from circulation. Desialylation of platelet VWFR therefore triggers platelet clearance and primes GPIbα and GPV for MP-dependent cleavage.


Asunto(s)
Plaquetas/fisiología , Glicoproteínas de Membrana/metabolismo , Metaloproteasas/metabolismo , Neuraminidasa/metabolismo , Refrigeración , Proteínas ADAM/metabolismo , Proteínas ADAM/fisiología , Proteína ADAM17 , Animales , Plaquetas/metabolismo , Conservación de la Sangre/métodos , Activación Enzimática , Glicosilación , Humanos , Masculino , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/fisiología , Metaloproteasas/fisiología , Ratones , Ratones Endogámicos C57BL , Neuraminidasa/fisiología , Complejo GPIb-IX de Glicoproteína Plaquetaria , Procesamiento Proteico-Postraduccional/fisiología , Proteolisis , Refrigeración/métodos , Factor de von Willebrand/metabolismo
5.
Blood ; 119(26): 6335-43, 2012 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-22596262

RESUMEN

Transfusion-related acute lung injury (TRALI) is the leading cause of transfusion-related death. The biologic processes contributing to TRALI are poorly understood. All blood products can cause TRALI, and no specific treatment is available. A "2-event model" has been proposed as the trigger. The first event may include surgery, trauma, or infection; the second involves the transfusion of antileukocyte antibodies or bioactive lipids within the blood product. Together, these events induce neutrophil activation in the lungs, causing endothelial damage and capillary leakage. Neutrophils, in response to pathogens or under stress, can release their chromatin coated with granule contents, thus forming neutrophil extracellular traps (NETs). Although protective against infection, these NETs are injurious to tissue. Here we show that NET biomarkers are present in TRALI patients' blood and that NETs are produced in vitro by primed human neutrophils when challenged with anti-HNA-3a antibodies previously implicated in TRALI. NETs are found in alveoli of mice experiencing antibody-mediated TRALI. DNase 1 inhalation prevents their alveolar accumulation and improves arterial oxygen saturation even when administered 90 minutes after TRALI onset. We suggest that NETs form in the lungs during TRALI, contribute to the disease process, and thus could be targeted to prevent or treat TRALI.


Asunto(s)
Lesión Pulmonar Aguda/etiología , ADN/inmunología , ADN/metabolismo , Neutrófilos/inmunología , Neutrófilos/metabolismo , Reacción a la Transfusión , Lesión Pulmonar Aguda/inmunología , Animales , Donantes de Sangre , Células Cultivadas , Espacio Extracelular/genética , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Activación Neutrófila/inmunología , Neutrófilos/patología , Inmunología del Trasplante , Trasplante Homólogo/inmunología
6.
Arterioscler Thromb Vasc Biol ; 33(9): 2112-20, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23825365

RESUMEN

OBJECTIVE: Aberrant blood-brain barrier (BBB) permeability is a hallmark pathology of many central nervous system diseases. von Willebrand factor (VWF) is stored in endothelial Weibel-Palade bodies from where it is released on activation into plasma and basement membrane. The role of VWF in endothelial homeostasis is unclear. The goal of this study was to assess the role of VWF in disease models associated with increased BBB permeability. APPROACH AND RESULTS: We did not find any differences in BBB permeability to Evans blue dye at baseline between wild-type and VWF(-/-) animals. We next used 2 models presenting with increased BBB permeability, hypoxia/reoxygenation and pilocarpine-induced status epilepticus, to assess the response of VWF(-/-) animals. In both models, VWF(-/-) mice maintained a tighter BBB than wild-type mice. VWF(-/-) mice fared worse in both conditions, with ≈ 100% of VWF(-/-) mice dying within 120 minutes after pilocarpine administration, whereas >80% of wild-type animals survived. Investigation into the status of tight junction proteins revealed that VWF(-/-) mice expressed more claudin-5 at baseline. In vitro work confirmed that the presence of subendothelial VWF is inhibitory to claudin-5 expression. CONCLUSIONS: VWF deficiency confers partial preservation of BBB integrity after hypoxia/reoxygenation and seizures. Surprisingly, this decrease in BBB permeability did not result in protection of animals because they demonstrated more severe pathology in both models compared with wild-type animals. These data suggest that a rigid BBB is detrimental (to the organism) during certain disease states and that VWF release may provide desired flexibility under stress.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Permeabilidad Capilar , Células Endoteliales/metabolismo , Hipoxia-Isquemia Encefálica/prevención & control , Estado Epiléptico/prevención & control , Enfermedades de von Willebrand/metabolismo , Factor de von Willebrand/metabolismo , Animales , Células Cultivadas , Claudina-5/metabolismo , Modelos Animales de Enfermedad , Femenino , Genotipo , Hipoxia-Isquemia Encefálica/genética , Hipoxia-Isquemia Encefálica/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Pilocarpina , Proteínas Recombinantes/administración & dosificación , Estado Epiléptico/inducido químicamente , Estado Epiléptico/genética , Estado Epiléptico/metabolismo , Trombocitopenia/metabolismo , Uniones Estrechas/metabolismo , Enfermedades de von Willebrand/genética , Factor de von Willebrand/administración & dosificación , Factor de von Willebrand/genética
7.
Blood ; 115(9): 1835-42, 2010 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-19965619

RESUMEN

Platelets undergo several modifications during storage that reduce their posttransfusion survival and functionality. One important feature of these changes, which are known as platelet storage lesion, is the shedding of the surface glycoproteins GPIb-alpha and GPV. We recently demonstrated that tumor necrosis factor-alpha converting enzyme (TACE/ADAM17) mediates mitochondrial injury-induced shedding of adhesion receptors and that TACE activity correlates with reduced posttransfusion survival of these cells. We now confirm that TACE mediates receptor shedding and clearance of platelets stored for 16 hours at 37 degrees C or 22 degrees C. We further demonstrate that both storage and mitochondrial injury lead to the phosphorylation of p38 mitogen-activated kinase (MAPK) in platelets and that TACE-mediated receptor shedding from mouse and human platelets requires p38 MAP kinase signaling. Protein kinase C, extracellular regulated-signal kinase MAPK, and caspases were not involved in TACE activation. Both inhibition of p38 MAPK and inactivation of TACE during platelet storage led to a markedly improved posttransfusion recovery and hemostatic function of platelets in mice. p38 MAPK inhibitors had only minor effects on the aggregation of fresh platelets under static or flow conditions in vitro. In summary, our data suggest that inhibition of p38 MAPK or TACE during storage may significantly improve the quality of stored platelets.


Asunto(s)
Plaquetas/enzimología , Proteínas Quinasas p38 Activadas por Mitógenos/sangre , Proteínas ADAM/antagonistas & inhibidores , Proteínas ADAM/sangre , Proteínas ADAM/deficiencia , Proteínas ADAM/genética , Proteína ADAM17 , Animales , Plaquetas/fisiología , Conservación de la Sangre , Activación Enzimática , Hemostasis/fisiología , Humanos , Técnicas In Vitro , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Ratones Transgénicos , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Transfusión de Plaquetas , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores
8.
J Clin Invest ; 117(6): 1699-707, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17492052

RESUMEN

Single gene mutations in beta integrins can account for functional defects of individual cells of the hematopoietic system. In humans, mutations in beta(2) integrin lead to leukocyte adhesion deficiency (LAD) syndrome and mutations in beta(3) integrin cause the bleeding disorder Glanzmann thrombasthenia. However, multiple defects in blood cells involving various beta integrins (beta(1), beta(2), and beta(3)) occur simultaneously in patients with the recently described LAD type III (LAD-III). Here we show that the product of a single gene, Ca(2+) and diacylglycerol-regulated guanine nucleotide exchange factor I (CalDAG-GEFI), controlled the activation of all 3 integrins in the hematopoietic system. Neutrophils from CalDAG-GEFI(-/-) mice exhibited strong defects in Rap1 and beta(1) and beta(2) integrin activation while maintaining normal calcium flux, degranulation, and ROS generation. Neutrophils from CalDAG-GEFI-deficient mice failed to adhere firmly to stimulated venules and to migrate into sites of inflammation. Furthermore, CalDAG-GEFI regulated the activation of beta(1) and beta(3) integrins in platelets, and CalDAG-GEFI deficiency caused complete inhibition of arterial thrombus formation in mice. Thus, mice engineered to lack CalDAG-GEFI have a combination of defects in leukocyte and platelet functions similar to that of LAD-III patients.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/deficiencia , Síndrome de Deficiencia de Adhesión del Leucocito/etiología , Animales , Plaquetas/metabolismo , Antígenos CD18/metabolismo , Modelos Animales de Enfermedad , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Técnicas In Vitro , Integrina beta1/metabolismo , Integrina beta3/metabolismo , Síndrome de Deficiencia de Adhesión del Leucocito/genética , Síndrome de Deficiencia de Adhesión del Leucocito/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Neutrófilos/metabolismo , Activación Plaquetaria , Trombosis/etiología , Trombosis/prevención & control , Proteínas de Unión al GTP rap1/metabolismo
9.
Am J Pathol ; 175(4): 1699-708, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19729481

RESUMEN

Platelets are crucial regulators of tumor vascular homeostasis and continuously prevent tumor hemorrhage through secretion of their granules. However, the reason for tumor bleeding in the absence of platelets remains unknown. Tumors are associated with inflammation, a cause of hemorrhage in thrombocytopenia. Here, we investigated the role of the inflamed tumor microenvironment in the induction of tumor vessel injury in thrombocytopenic mice. Using s.c. injections of vascular endothelial growth factor or tumor necrosis factor-alpha combined with depletion of neutrophils, we demonstrate that enhancing the opening of endothelial cell junctions was not sufficient to cause bleeding in the absence of platelets; instead, induction of tissue hemorrhage in thrombocytopenia required recruitment of leukocytes. Immunohistology revealed that thrombocytopenia-induced tumor hemorrhage occurs at sites of macrophage and neutrophil accumulation. Mice deficient in beta2 or beta3 integrins, which have decreased neutrophil and/or macrophage infiltration in their tumor stroma, were protected from thrombocytopenia-induced tumor hemorrhage, indicating that, in the absence of platelets, stroma-infiltrating leukocytes induced tumor vessel injury. This injury was independent of reactive oxygen species generation and of complement activation, as suggested by the persistence of tumor hemorrhage in C3- and nicotinamide adenine dinucleotide phosphate oxidase-deficient thrombocytopenic mice. Our results show that platelets counteract tumor-associated inflammation and that the absence of this platelet function elicits vascular injuries by tumor-infiltrating innate immune cells.


Asunto(s)
Hemorragia/complicaciones , Hemorragia/inmunología , Inmunidad Innata/inmunología , Neoplasias/complicaciones , Trombocitopenia/complicaciones , Animales , Permeabilidad Capilar , Recuento de Células , Muerte Celular , Línea Celular Tumoral , Movimiento Celular , Activación de Complemento/inmunología , Femenino , Hemorragia/patología , Integrinas/metabolismo , Macrófagos/patología , Ratones , Neoplasias/irrigación sanguínea , Neoplasias/patología , Neutrófilos/patología , Especies Reactivas de Oxígeno/metabolismo , Piel/irrigación sanguínea , Piel/patología , Trombocitopenia/patología
10.
Blood ; 112(5): 1696-703, 2008 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-18544684

RESUMEN

Second messenger-mediated inside-out activation of integrin alphaIIbbeta3 is a key step in platelet aggregation. We recently showed strongly impaired but not absent alphaIIbbeta3-mediated aggregation of CalDAG-GEFI-deficient platelets activated with various agonists. Here we further evaluated the roles of CalDAG-GEFI and protein kinase C (PKC) for alphaIIbbeta3 activation in platelets activated with a PAR4 receptor-specific agonist, GYPGKF (PAR4p). Compared with wild-type controls, platelets treated with the PKC inhibitor Ro31-8220 or CalDAG-GEFI-deficient platelets showed a marked defect in aggregation at low (< 1mM PAR4p) but not high PAR4p concentrations. Blocking of PKC function in CalDAG-GEFI-deficient platelets, how-ever, strongly decreased aggregation at all PAR4p concentrations, demonstrating that CalDAG-GEFI and PKC represent separate, but synergizing, pathways important for alphaIIbbeta3 activation. PAR4p-induced aggregation in the absence of CalDAG-GEFI required cosignaling through the Galphai-coupled receptor for ADP, P2Y12. Independent roles for CalDAG-GEFI and PKC/Galphai signaling were also observed for PAR4p-induced activation of the small GTPase Rap1, with CalDAG-GEFI mediating the rapid but reversible activation of this small GTPase. In summary, our study identifies CalDAG-GEFI and PKC as independent pathways leading to Rap1 and alphaIIbbeta3 activation in mouse platelets activated through the PAR4 receptor.


Asunto(s)
Plaquetas/metabolismo , Factores de Intercambio de Guanina Nucleótido/sangre , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Proteína Quinasa C/sangre , Adenosina Difosfato/sangre , Animales , Plaquetas/efectos de los fármacos , Plaquetas/fisiología , Degranulación de la Célula/efectos de los fármacos , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/sangre , Factores de Intercambio de Guanina Nucleótido/deficiencia , Factores de Intercambio de Guanina Nucleótido/genética , Cinética , Ratones , Ratones Noqueados , Modelos Biológicos , Oligopéptidos/farmacología , Agregación Plaquetaria/efectos de los fármacos , Agregación Plaquetaria/fisiología , Proteína Quinasa C/antagonistas & inhibidores , Receptores de Trombina/sangre , Receptores de Trombina/efectos de los fármacos , Transducción de Señal , Proteínas de Unión al GTP rap1/sangre
11.
Blood ; 111(10): 4958-64, 2008 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-18256319

RESUMEN

The role of platelets in hemostasis is to produce a plug to arrest bleeding. During thrombocytopenia, spontaneous bleeding is seen in some patients but not in others; the reason for this is unknown. Here, we subjected thrombocytopenic mice to models of dermatitis, stroke, and lung inflammation. The mice showed massive hemorrhage that was limited to the area of inflammation and was not observed in uninflamed thrombocytopenic mice. Endotoxin-induced lung inflammation during thrombocytopenia triggered substantial intra-alveolar hemorrhage leading to profound anemia and respiratory distress. By imaging the cutaneous Arthus reaction through a skin window, we observed in real time the loss of vascular integrity and the kinetics of skin hemorrhage in thrombocytopenic mice. Bleeding-observed mostly from venules-occurred as early as 20 minutes after challenge, pointing to a continuous need for platelets to maintain vascular integrity in inflamed microcirculation. Inflammatory hemorrhage was not seen in genetically engineered mice lacking major platelet adhesion receptors or their activators (alphaIIbbeta3, glycoprotein Ibalpha [GPIbalpha], GPVI, and calcium and diacylglycerol-regulated guanine nucleotide exchange factor I [CalDAG-GEFI]), thus indicating that firm platelet adhesion was not necessary for their supporting role. While platelets were previously shown to promote endothelial activation and recruitment of inflammatory cells, they also appear indispensable to maintain vascular integrity in inflamed tissue. Based on our observations, we propose that inflammation may cause life-threatening hemorrhage during thrombocytopenia.


Asunto(s)
Hemorragia/etiología , Inflamación/complicaciones , Trombocitopenia/complicaciones , Animales , Plaquetas/fisiología , Permeabilidad Capilar , Ratones , Adhesividad Plaquetaria , Trombocitopenia/patología
12.
PLoS One ; 12(12): e0188341, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29236713

RESUMEN

Inflammation is a common denominator in chronic diseases of aging. Yet, how inflammation fuels these diseases remains unknown. Neutrophils are the primary leukocytes involved in the early phase of innate immunity and inflammation. As part of their anti-microbial defense, neutrophils form extracellular traps (NETs) by releasing decondensed chromatin lined with cytotoxic proteins. NETs have been shown to induce tissue injury and thrombosis. Here, we demonstrated that Sirt3, a nicotinamide adenine dinucleotide (NAD+)-dependent protein deacetylase, an enzyme linked to human longevity, was expressed in mouse neutrophils and platelets. Using Sirt3-/- mice as a model of accelerated aging, we investigated the effects of Sirt3 deficiency on NETosis and platelet function, aiming to detect enhancement of thrombosis. More mitochondrial reactive oxygen species (ROS) were generated in neutrophils and platelets of Sirt3-/- mice compared to WT, when stimulated with a low concentration of phorbol 12-myristate 13-acetate (PMA) and a high concentration of thrombin, respectively. There were no differences in in vitro NETosis, with or without stimulation. Platelet aggregation was mildly augmented in Sirt3-/- mice compared to WT mice, when stimulated with a low concentration of collagen. The effect of Sirt3 deficiency on platelet and neutrophil activation in vivo was examined by the venous thrombosis model of inferior vena cava stenosis. Elevation of plasma DNA concentration was observed after stenosis in both genotypes, but no difference was shown between the two genotypes. The systemic response to thrombosis was enhanced in Sirt3-/- mice with significantly elevated neutrophil count and reduced platelet count. However, no differences were observed in incidence of thrombus formation, thrombus weight and thrombin-antithrombin complex generation between WT and Sirt3-/- mice. We conclude that Sirt3 does not considerably impact NET formation, platelet function, or venous thrombosis in healthy young mice.


Asunto(s)
Plaquetas/metabolismo , Neutrófilos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Sirtuina 3/fisiología , Trombosis de la Vena/genética , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Sirtuina 3/genética
13.
TH Open ; 1(1): e11-e23, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29376146

RESUMEN

BACKGROUND: Inflammatory bowel disease (IBD) affects 1.6 million people in the United States. IBD is associated with an increased risk of thrombosis, which rises with disease activity. The pathogenesis of IBD and its increased thrombotic risk is not completely understood. Ultra large von Willebrand factor (ULVWF) multimers are secreted from activated endothelium, leading to recruitment of platelets and leukocytes. A disintegrin and metalloproteinase with thrombospondin type I repeats motif 13 (ADAMTS13) cleaves highly adhesive ULVWF into smaller, less bioactive, multimers, releasing them into circulation. Mice deficient in ADAMTS13 (ADAMTS13-/-) have heightened inflammatory and thrombotic responses. OBJECTIVES: We hypothesized that upon colitis induction, ADAMTS13-/- mice would have more severe symptoms compared with wild-type (WT) mice, and rhADAMTS13 administration to mice with colitis would improve their condition. RESULTS: Dextran sodium sulfate-induced colitis was worse in ADAMTS13-/- mice than WT. ADAMTS13-/- showed increased weight loss, worse anemia, and increased clinical and histologic colitis severity, compared with WT mice. ADAMTS13-/- mice had increased VWF release, with accumulation at inflamed colonic sites. Also, the majority of mice showed one or more submucosal colonic thrombi. ADAMTS13 deficiency worsened colitis and propagated intestinal inflammation, most likely through increased platelet-leukocyte recruitment by VWF. Treatment of WT mice with rhA-DAMTS13 decreased colitis severity without worsening anemia. Additionally, several immune-mediated chronic murine colitis models, and inflamed colon tissue specimens from IBD patients, showed increased VWF release at inflamed sites, suggesting a generalizability of our findings. CONCLUSION: Measuring VWF/ADAMTS13 levels could have clinical utility. When applicable, the administration of ADAMTS13, in addition to primary treatment, may improve outcomes for IBD patients.

14.
J Exp Med ; 214(2): 439-458, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28031479

RESUMEN

Aging promotes inflammation, a process contributing to fibrosis and decline in organ function. The release of neutrophil extracellular traps (NETs [NETosis]), orchestrated by peptidylarginine deiminase 4 (PAD4), damages organs in acute inflammatory models. We determined that NETosis is more prevalent in aged mice and investigated the role of PAD4/NETs in age-related organ fibrosis. Reduction in fibrosis was seen in the hearts and lungs of aged PAD4-/- mice compared with wild-type (WT) mice. An increase in left ventricular interstitial collagen deposition and a decline in systolic and diastolic function were present only in WT mice, and not in PAD4-/- mice. In an experimental model of cardiac fibrosis, cardiac pressure overload induced NETosis and significant platelet recruitment in WT but not PAD4-/- myocardium. DNase 1 was given to assess the effects of extracellular chromatin. PAD4 deficiency or DNase 1 similarly protected hearts from fibrosis. We propose a role for NETs in cardiac fibrosis and conclude that PAD4 regulates age-related organ fibrosis and dysfunction.


Asunto(s)
Hidrolasas/fisiología , Miocardio/patología , Factores de Edad , Animales , Colágeno/metabolismo , Trampas Extracelulares/fisiología , Fibrosis , Hidrolasas/genética , Ratones , Ratones Endogámicos C57BL , Arginina Deiminasa Proteína-Tipo 4 , Fibrosis Pulmonar/etiología , Especies Reactivas de Oxígeno/metabolismo , Función Ventricular Izquierda
15.
J Leukoc Biol ; 88(2): 313-9, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20413728

RESUMEN

Chemotaxis and integrin activation are essential processes for neutrophil transmigration in response to injury. CalDAG-GEFI plays a key role in the activation of beta1, beta2, and beta3 integrins in platelets and neutrophils by exchanging a GDP for a GTP on Rap1. Here, we explored the role of CalDAG-GEFI and Rap1b in integrin-independent neutrophil chemotaxis. In a transwell assay, CalDAG-GEFI-/- neutrophils had a 46% reduction in transmigration compared with WT in response to a low concentration of LTB4. Visualization of migrating neutrophils in the presence of 10 mM EDTA revealed that CalDAG-GEFI-/- neutrophils had abnormal chemotactic behavior compared with WT neutrophils, including reduced speed and directionality. Interestingly, Rap1b-/- neutrophils had a similar phenotype in this assay, suggesting that CalDAG-GEFI may be acting through Rap1b. We investigated whether the deficit in integrin-independent chemotaxis in CalDAG-GEFI-/- neutrophils could be explained by defective cytoskeleton rearrangement. Indeed, we found that CalDAG-GEFI-/- neutrophils had reduced formation of F-actin pseudopodia after LTB4 stimulation, suggesting that they have a defect in polarization. Overall, our studies show that CalDAG-GEFI helps regulate neutrophil chemotaxis, independent of its established role in integrin activation, through a mechanism that involves actin cytoskeleton and cellular polarization.


Asunto(s)
Quimiotaxis de Leucocito , Factores de Intercambio de Guanina Nucleótido/fisiología , Integrinas/metabolismo , Neutrófilos/fisiología , Actinas , Animales , Polaridad Celular , Citoesqueleto/patología , Leucotrieno B4/farmacología , Ratones , Ratones Endogámicos C57BL , Proteínas de Unión al GTP rap1/fisiología
16.
Cancer Res ; 68(16): 6851-8, 2008 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-18701510

RESUMEN

Cancer is associated with a prothrombogenic state capable of platelet activation. Platelets, on the other hand, can support angiogenesis, a process involved in the progression of tumor growth and metastasis. However, it is unclear whether platelet/tumor interactions substantially contribute to tumor physiology. We investigated whether platelets stabilize tumor vessels and studied the underlying mechanisms. We induced severe acute thrombocytopenia in mice bearing s.c. Lewis lung carcinoma or B16F10 melanoma. Intravital microscopy revealed that platelet depletion led to a rapid destabilization of tumor vessels with intratumor hemorrhage starting as soon as 30 min after induction of thrombocytopenia. Using an inhibitor of glycoprotein Ibalpha (GPIbalpha) and genetically engineered mice with platelet adhesion defects, we investigated the role of platelet adhesion receptors in stabilizing tumor vessels. We found that a single defect in either GPIbalpha, von Willebrand factor, P-selectin, or platelet integrin activation did not lead to intratumor hemorrhage. We then compared the ability of transfused resting and degranulated platelets to prevent intratumor hemorrhage. Whereas resting platelets prevented thrombocytopenia-induced tumor bleeding, circulating degranulated platelets did not. This suggests that the prevention of intratumor hemorrhage by platelets relies on the secretion of the content of platelet granules. Supporting this hypothesis, we further found that thrombocytopenia dramatically impairs the balance between propermeability and antipermeability factors in tumor-bearing animals, in particular depleting blood of angiopoietin-1 and serotonin. Our results show a crucial contribution of platelets to tumor homeostasis through continuous prevention of severe intratumor hemorrhage and consequent cell death. The study also suggests platelet function as a reasonable target for specific destabilization of tumor vessels.


Asunto(s)
Plaquetas/metabolismo , Carcinoma Pulmonar de Lewis/prevención & control , Hemorragia/prevención & control , Melanoma Experimental/prevención & control , Adhesividad Plaquetaria , Trombocitopenia/prevención & control , Angiopoyetina 1/metabolismo , Animales , Carcinoma Pulmonar de Lewis/patología , Proliferación Celular , Femenino , Factores de Intercambio de Guanina Nucleótido/fisiología , Hemoglobinas/metabolismo , Técnicas para Inmunoenzimas , Neoplasias Pulmonares/secundario , Melanoma Experimental/patología , Metaloendopeptidasas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Necrosis , Selectina-P/fisiología , Activación Plaquetaria , Complejo GPIb-IX de Glicoproteína Plaquetaria/fisiología , Serotonina/metabolismo , Trombocitopenia/etiología , Trombocitopenia/patología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor de von Willebrand/fisiología
17.
Proc Natl Acad Sci U S A ; 103(45): 16900-5, 2006 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-17075060

RESUMEN

GPIbalpha binding to von Willebrand factor (VWF) exposed at a site of vascular injury is thought to be the first step in the formation of a hemostatic plug. However, our previous studies in VWF-deficient mice demonstrated delayed but not absent arterial thrombus formation, suggesting that, under these conditions, GPIbalpha may bind other ligands or that a receptor other than GPIbalpha can mediate platelet adhesion. Here, we studied thrombus formation in transgenic mice expressing GPIbalpha in which the extracellular domain was replaced by that of the human IL-4 receptor (IL4Ralpha/GPIbalpha-tg mice). Platelet adhesion to ferric chloride-treated mesenteric arterioles in IL4Ralpha/GPIbalpha-tg mice was virtually absent in contrast to avid adhesion in WT mice. As a consequence, arterial thrombus formation was inhibited completely in the mutant mice. Our studies further show that, when infused into WT recipient mice, IL4Ralpha/GPIbalpha-tg platelets or WT platelets lacking the 45-kDa N-terminal domain of GPIbalpha failed to incorporate into growing arterial thrombi, even if the platelets were activated before infusion. Surprisingly, platelets lacking beta3 integrins, which are unable to form thrombi on their own, incorporated efficiently into WT thrombi. Our studies provide in vivo evidence that GPIbalpha absolutely is required for recruitment of platelets to both exposed subendothelium and thrombi under arterial flow conditions. Thus, GPIbalpha contributes to arterial thrombosis by important adhesion mechanisms independent of the binding to VWF.


Asunto(s)
Complejo GPIb-IX de Glicoproteína Plaquetaria/fisiología , Trombosis/sangre , Factor de von Willebrand/fisiología , Animales , Arteriolas/patología , Arteriolas/fisiopatología , Plaquetas/patología , Plaquetas/fisiología , Modelos Animales de Enfermedad , Humanos , Subunidad alfa del Receptor de Interleucina-4/genética , Subunidad alfa del Receptor de Interleucina-4/fisiología , Ligandos , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Adhesividad Plaquetaria/fisiología , Complejo GPIb-IX de Glicoproteína Plaquetaria/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Trombosis/etiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA