Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 152
Filtrar
1.
Blood ; 137(11): 1538-1549, 2021 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-33512489

RESUMEN

Neutrophils play a crucial role in the intertwined processes of thrombosis and inflammation. An altered neutrophil phenotype may contribute to inadequate resolution, which is known to be a major pathophysiological contributor of thromboinflammatory conditions such as sickle cell disease (SCD). The endogenous protein annexin A1 (AnxA1) facilitates inflammation resolution via formyl peptide receptors (FPRs). We sought to comprehensively elucidate the functional significance of targeting the neutrophil-dependent AnxA1/FPR2/ALX pathway in SCD. Administration of AnxA1 mimetic peptide AnxA1Ac2-26 ameliorated cerebral thrombotic responses in Sickle transgenic mice via regulation of the FPR2/ALX (a fundamental receptor involved in resolution) pathway. We found direct evidence that neutrophils with SCD phenotype play a key role in contributing to thromboinflammation. In addition, AnxA1Ac2-26 regulated activated SCD neutrophils through protein kinase B (Akt) and extracellular signal-regulated kinases (ERK1/2) to enable resolution. We present compelling conceptual evidence that targeting the AnxA1/FPR2/ALX pathway may provide new therapeutic possibilities against thromboinflammatory conditions such as SCD.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Anemia de Células Falciformes/metabolismo , Anexina A1/metabolismo , Receptores de Formil Péptido/metabolismo , Receptores de Lipoxina/metabolismo , Transducción de Señal , Trombosis/metabolismo , Adulto , Anemia de Células Falciformes/complicaciones , Anemia de Células Falciformes/patología , Animales , Femenino , Humanos , Inflamación/etiología , Inflamación/metabolismo , Inflamación/patología , Masculino , Ratones , Persona de Mediana Edad , Neutrófilos/metabolismo , Neutrófilos/patología , Trombosis/etiología , Trombosis/patología , Adulto Joven
2.
Circulation ; 140(4): 319-335, 2019 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-31154815

RESUMEN

BACKGROUND: Ischemia reperfusion injury (I/RI) is a common complication of cardiovascular diseases. Resolution of detrimental I/RI-generated prothrombotic and proinflammatory responses is essential to restore homeostasis. Platelets play a crucial part in the integration of thrombosis and inflammation. Their role as participants in the resolution of thromboinflammation is underappreciated; therefore we used pharmacological and genetic approaches, coupled with murine and clinical samples, to uncover key concepts underlying this role. METHODS: Middle cerebral artery occlusion with reperfusion was performed in wild-type or annexin A1 (AnxA1) knockout (AnxA1-/-) mice. Fluorescence intravital microscopy was used to visualize cellular trafficking and to monitor light/dye-induced thrombosis. The mice were treated with vehicle, AnxA1 (3.3 mg/kg), WRW4 (1.8 mg/kg), or all 3, and the effect of AnxA1 was determined in vivo and in vitro. RESULTS: Intravital microscopy revealed heightened platelet adherence and aggregate formation post I/RI, which were further exacerbated in AnxA1-/- mice. AnxA1 administration regulated platelet function directly (eg, via reducing thromboxane B2 and modulating phosphatidylserine expression) to promote cerebral protection post-I/RI and act as an effective preventative strategy for stroke by reducing platelet activation, aggregate formation, and cerebral thrombosis, a prerequisite for ischemic stroke. To translate these findings into a clinical setting, we show that AnxA1 plasma levels are reduced in human and murine stroke and that AnxA1 is able to act on human platelets, suppressing classic thrombin-induced inside-out signaling events (eg, Akt activation, intracellular calcium release, and Ras-associated protein 1 [Rap1] expression) to decrease αIIbß3 activation without altering its surface expression. AnxA1 also selectively modifies cell surface determinants (eg, phosphatidylserine) to promote platelet phagocytosis by neutrophils, thereby driving active resolution. (n=5-13 mice/group or 7-10 humans/group.) Conclusions: AnxA1 affords protection by altering the platelet phenotype in cerebral I/RI from propathogenic to regulatory and reducing the propensity for platelets to aggregate and cause thrombosis by affecting integrin (αIIbß3) activation, a previously unknown phenomenon. Thus, our data reveal a novel multifaceted role for AnxA1 to act both as a therapeutic and a prophylactic drug via its ability to promote endogenous proresolving, antithromboinflammatory circuits in cerebral I/RI. Collectively, these results further advance our knowledge and understanding in the field of platelet and resolution biology.


Asunto(s)
Anexina A1/genética , Plaquetas/metabolismo , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Inflamación/genética , Anciano , Anciano de 80 o más Años , Animales , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Transducción de Señal
3.
FASEB J ; 32(6): 3448-3456, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29452567

RESUMEN

Angiotensin II (Ang-II)-induced hypertension is associated with accelerated thrombus formation in arterioles and leukocyte recruitment in venules. The mechanisms that underlie the prothrombotic and proinflammatory responses to chronic Ang-II administration remain poorly understood. We evaluated the role of CD40/CD40 ligand (CD40L) signaling in Ang-II-mediated microvascular responses and assessed whether and how soluble CD40L (sCD40L) contributes to this response. Intravital video microscopy was performed to analyze leukocyte recruitment and dihydrorhodamine-123 oxidation in postcapillary venules. Thrombus formation in cremaster muscle arterioles was induced by using the light/dye endothelial cell injury model. Wild-type (WT), CD40-/-, and CD40L-/- mice received Ang-II for 14 d via osmotic minipumps. Some mice were treated with either recombinant sCD40L or the VLA5 (very late antigen 5; α5ß1) antagonist, ATN-161. Our results demonstrate that CD40-/-, CD40L-/-, and WT mice that were treated with ATN-161 were protected against the thrombotic and inflammatory effects of Ang-II infusion. Infusion of sCD40L into CD40-/- or CD40L-/- mice restored the prothrombotic effect of Ang-II infusion. Mice that were treated with ATN-161 and infused with sCD40L were protected against accelerated thrombosis. Collectively, these novel findings suggest that the mechanisms that underlie Ang-II-dependent thrombotic and inflammatory responses link to the signaling of CD40L via both CD40 and VLA5.-Senchenkova, E. Y., Russell, J., Vital, S. A., Yildirim, A., Orr, A. W., Granger, D. N., Gavins, F. N. E. A critical role for both CD40 and VLA5 in angiotensin II-mediated thrombosis and inflammation.


Asunto(s)
Angiotensina II/metabolismo , Antígenos CD40/metabolismo , Integrina alfa5beta1/metabolismo , Transducción de Señal , Trombosis/metabolismo , Angiotensina II/genética , Animales , Antígenos CD40/genética , Ligando de CD40/genética , Ligando de CD40/metabolismo , Humanos , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Integrina alfa5beta1/genética , Masculino , Ratones , Ratones Noqueados , Trombosis/genética , Trombosis/patología
4.
Circulation ; 133(22): 2169-79, 2016 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-27154726

RESUMEN

BACKGROUND: Platelet activation at sites of vascular injury is essential for hemostasis, but it is also a major pathomechanism underlying ischemic injury. Because anti-inflammatory therapies limit thrombosis and antithrombotic therapies reduce vascular inflammation, we tested the therapeutic potential of 2 proresolving endogenous mediators, annexin A1 N-terminal derived peptide (AnxA1Ac2-26) and aspirin-triggered lipoxin A4 (15-epi-lipoxin A4), on the cerebral microcirculation after ischemia/reperfusion injury. Furthermore, we tested whether the lipoxin A4 receptor formyl-peptide receptor 2/3 (Fpr2/3; ortholog to human FPR2/lipoxin A4 receptor) evoked neuroprotective functions after cerebral ischemia/reperfusion injury. METHODS AND RESULTS: Using intravital microscopy, we found that cerebral ischemia/reperfusion injury was accompanied by neutrophil and platelet activation and neutrophil-platelet aggregate formation within cerebral microvessels. Moreover, aspirin-triggered lipoxin A4 activation of neutrophil Fpr2/3 regulated neutrophil-platelet aggregate formation in the brain and inhibited the reactivity of the cerebral microvasculature. The same results were obtained with AnxA1Ac2-26 administration. Blocking Fpr2/lipoxin A4 receptor with the antagonist Boc2 reversed this effect, and treatments were ineffective in Fpr2/3 knockout mice, which displayed an exacerbated disease severity, evidenced by increased infarct area, blood-brain barrier dysfunction, increased neurological score, and elevated levels of cytokines. Furthermore, aspirin treatment significantly reduced cerebral leukocyte recruitment and increased endogenous levels of aspirin-triggered lipoxin A4, effects again mediated by Fpr2/3. CONCLUSION: Fpr2/lipoxin A4 receptor is a therapeutic target for initiating endogenous proresolving, anti-inflammatory pathways after cerebral ischemia/reperfusion injury.


Asunto(s)
Enfermedades Cardiovasculares/terapia , Infarto Cerebral/patología , Neutrófilos/fisiología , Receptores de Formil Péptido/fisiología , Secuencia de Aminoácidos , Animales , Anexina A1/genética , Anexina A1/farmacología , Anexina A1/uso terapéutico , Enfermedades Cardiovasculares/patología , Infarto Cerebral/prevención & control , Inflamación/patología , Inflamación/prevención & control , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Formil Péptido/agonistas
5.
Stroke ; 48(12): 3356-3365, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29114092

RESUMEN

BACKGROUND AND PURPOSE: Inflammation and thrombosis currently are recognized as critical contributors to the pathogenesis of ischemic stroke. CD147 (cluster of differentiation 147), also known as extracellular matrix metalloproteinase inducer, can function as a key mediator of inflammatory and immune responses. CD147 expression is increased in the brain after cerebral ischemia, but its role in the pathogenesis of ischemic stroke remains unknown. In this study, we show that CD147 acts as a key player in ischemic stroke by driving thrombotic and inflammatory responses. METHODS: Focal cerebral ischemia was induced in C57BL/6 mice by a 60-minute transient middle cerebral artery occlusion. Animals were treated with anti-CD147 function-blocking antibody (αCD147) or isotype control antibody. Blood-brain barrier permeability, thrombus formation, and microvascular patency were assessed 24 hours after ischemia. Infarct size, neurological deficits, and inflammatory cells invaded in the brain were assessed 72 hours after ischemia. RESULTS: CD147 expression was rapidly increased in ischemic brain endothelium after transient middle cerebral artery occlusion. Inhibition of CD147 reduced infarct size and improved functional outcome on day 3 after transient middle cerebral artery occlusion. The neuroprotective effects were associated with (1) prevented blood-brain barrier damage, (2) decreased intravascular fibrin and platelet deposition, which in turn reduced thrombosis and increased cerebral perfusion, and (3) reduced brain inflammatory cell infiltration. The underlying mechanism may include reduced NF-κB (nuclear factor κB) activation, MMP-9 (matrix metalloproteinase-9) activity, and PAI-1 (plasminogen activator inhibitor-1) expression in brain microvascular endothelial cells. CONCLUSIONS: Inhibition of CD147 ameliorates acute ischemic stroke by reducing thromboinflammation. CD147 might represent a novel and promising therapeutic target for ischemic stroke and possibly other thromboinflammatory disorders.


Asunto(s)
Anticuerpos Bloqueadores/uso terapéutico , Basigina/antagonistas & inhibidores , Isquemia Encefálica/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Trombosis Intracraneal/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Accidente Cerebrovascular/tratamiento farmacológico , Animales , Basigina/biosíntesis , Plaquetas/efectos de los fármacos , Barrera Hematoencefálica/efectos de los fármacos , Fibrina/metabolismo , Infarto de la Arteria Cerebral Media/complicaciones , Infarto de la Arteria Cerebral Media/patología , Inflamación/patología , Trombosis Intracraneal/patología , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Resultado del Tratamiento
6.
Pathophysiology ; 24(4): 213-228, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29102280

RESUMEN

Microvascular dysfunction is well established as an early and rate-determining factor in the injury response of tissues to ischemia and reperfusion (I/R). Severe endothelial cell dysfunction, which can develop without obvious morphological cell injury, is a major underlying cause of the microvascular abnormalities that accompany I/R. While I/R-induced microvascular dysfunction is manifested in different ways, two responses that have received much attention in both the experimental and clinical setting are impaired capillary perfusion (no-reflow) and endothelial barrier failure with a transition to hemorrhage. These responses are emerging as potentially important determinants of the severity of the tissue injury response, and there is growing clinical evidence that they are predictive of clinical outcome following reperfusion therapy. This review provides a summary of animal studies that have focused on the mechanisms that may underlie the genesis of no-reflow and hemorrhage following reperfusion of ischemic tissues, and addresses the clinical evidence that implicates these vascular events in the responses of the ischemic brain (stroke) and heart (myocardial infarction) to reperfusion therapy. Inasmuch as reactive oxygen species (ROS) and matrix metalloproteinases (MMP) are frequently invoked as triggers of the microvascular dysfunction elicited by I/R, the potential roles and sources of these mediators are also discussed. The available evidence in the literature justifies the increased interest in the development of no-reflow and hemorrhage in heart and brain following reperfusion therapy, and suggests that these vascular events may be predictive of poor clinical outcome and warrant the development of targeted treatment strategies.

7.
Microvasc Res ; 105: 54-60, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26775070

RESUMEN

OBJECTIVE: Hypertension and hypercholesterolemia elicit inflammatory and thrombogenic responses in the microvasculature. However, little is known about whether and how risk factor combinations alter microvascular function. We examined how the actions of HTN+HCh on the microvasculature differ from the responses elicited by either risk factor alone. METHODS: Intravital microscopy was used to monitor the adhesion and emigration of leukocytes and dihydrorhodamine oxidation in cremaster muscle venules of wild type mice that were infused with angiotensin II for 2 weeks (HTN), placed on a high cholesterol diet (HCD), or both. RESULTS: Either HTN or HCh alone enhanced the production of reactive oxygen species and promoted the recruitment of leukocytes in venules. However, the combination of HTN and HCh produced changes in ROS production and leukocyte recruitment that were greatly attenuated compared to HTN alone. The inhibitory effects of HCh on the AngII mediated responses were also observed in genetically-induced HCh (ApoE-deficient mice). Treating HCh+HTN mice with an antagonist to AT2r reversed the HCh-dependent protection against oxidative stress and inflammation during HTN. CONCLUSIONS: These findings indicate that HCh blunts the oxidative stress and inflammatory cell recruitment elicited by hypertension in venules through a mechanism that involves AT2 receptor activation.


Asunto(s)
Hipercolesterolemia/metabolismo , Hipertensión/metabolismo , Estrés Oxidativo , Receptor de Angiotensina Tipo 2/metabolismo , Vénulas/metabolismo , Angiotensina II , Bloqueadores del Receptor Tipo 2 de Angiotensina II/farmacología , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Adhesión Celular , Quimiotaxis de Leucocito , Colesterol en la Dieta , Modelos Animales de Enfermedad , Hipercolesterolemia/etiología , Hipercolesterolemia/genética , Hipercolesterolemia/fisiopatología , Hipertensión/inducido químicamente , Hipertensión/fisiopatología , Leucocitos/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor de Angiotensina Tipo 2/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Vénulas/efectos de los fármacos , Vénulas/fisiopatología
8.
Semin Thromb Hemost ; 41(6): 582-9, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26270113

RESUMEN

Patients with inflammatory bowel disease (IBD) exhibit a threefold higher risk for development of systemic thrombosis than the general population. Although the underlying causes of the increased risk for thrombus development remain poorly understood, there is a large body of evidence suggesting that abnormalities in coagulation, fibrinolysis, and platelet function may contribute to this response. Changes in hemostatic biomarkers are consistent with subclinical activation of coagulation system, including tissue factor activation, impaired protein C pathway, enhanced thrombin generation, and diminished fibrinolytic capacity. There is also evidence for an increased production and reactivity of platelets, with an enhanced formation of platelet-platelet and platelet-leukocyte aggregates. The altered coagulation and platelet function, and the predisposition to thrombus formation have also been demonstrated in animal models of IBD. The animal studies have revealed a major role for inflammatory cytokines, including tumor necrosis factor-α, interleukin (IL)-1ß, and IL-6, as mediators of the platelet abnormalities and enhanced thrombus development in experimental IBD. These findings in animal models raise hope for the development of novel therapeutic strategies to reduce thrombosis-related mortality in IBD patients.


Asunto(s)
Plaquetas/patología , Enfermedades Inflamatorias del Intestino/sangre , Trombofilia/sangre , Animales , Factores de Coagulación Sanguínea/metabolismo , Colitis/sangre , Colon/irrigación sanguínea , Citocinas/fisiología , Modelos Animales de Enfermedad , Fibrinólisis , Humanos , Enfermedades Inflamatorias del Intestino/complicaciones , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Leucocitos/patología , Microcirculación , Modelos Biológicos , Óxido Nítrico/metabolismo , Activación Plaquetaria , Riesgo , Trombofilia/etiología , Trombofilia/prevención & control , Trombosis/epidemiología , Trombosis/etiología
9.
Microcirculation ; 21(5): 401-7, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24495184

RESUMEN

OBJECTIVE: AngII-induced HTN is associated with accelerated thrombus development in arterioles. This study assessed the contributions of different components of the coagulation cascade and fibrinolysis to AngII-mediated microvascular thrombosis. METHODS: Light/dye-induced thrombus formation (the time of onset and flow cessation) was quantified in cremaster muscle arterioles of AngII infused (two weeks) WT/AngII mice, EPCR-TgN, and mice deficient in PAI-1. WT/AngII mice were also treated with either tissue factor antibody, antithrombin III, heparin, hirudin, or murine APC. RESULTS: TF immunoblockade or hirudin treatment did not prevent the AngII-induced acceleration of thrombosis. While antithrombin III treatment prevented the acceleration in both thrombus onset and flow cessation, heparin only improved the time for blood flow cessation. Neither WT mice treated with murine APC nor EPCR-TgN were protected against AngII-induced thrombus development. A similar lack of protection was noted in PAI-1deficient mice. CONCLUSION: These findings implicate a role for thrombin generation pathway in the accelerated thrombosis induced by AngII and suggest that an impaired protein C pathway and increased PAI-1 do not make a significant contribution to this model of microvascular thrombosis.


Asunto(s)
Angiotensina II/farmacología , Fibrinólisis/efectos de los fármacos , Microvasos/metabolismo , Trombosis/metabolismo , Vasoconstrictores/farmacología , Animales , Antitrombina III/farmacología , Antitrombinas/farmacología , Hemostáticos/farmacología , Masculino , Ratones , Ratones Noqueados , Microvasos/patología , Proteína C/genética , Proteína C/metabolismo , Serpina E2/genética , Serpina E2/metabolismo , Trombina/metabolismo , Tromboplastina/farmacología , Trombosis/genética , Trombosis/patología
10.
Am J Pathol ; 183(1): 173-81, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23673000

RESUMEN

Clinical studies and animal experimentation have shown that colonic inflammation is associated with an increased number and reactivity of platelets, coagulation abnormalities, and enhanced thrombus formation. The objective of this study was to define the contribution of IL-6 to the thrombocytosis, exaggerated agonist-induced platelet aggregation, and enhanced extra-intestinal thrombosis that occur during experimental colitis. The number of mature and immature platelets, platelet life span, thrombin-induced platelet aggregation response, and light/dye-induced thrombus formation in cremaster muscle arterioles were measured in wild-type (WT) and IL-6-deficient (IL-6(-/-)) mice with dextran sodium sulfate (DSS)-induced colitis. DSS colitis in WT mice was associated with thrombocytosis with an elevated number of both mature and immature platelets and no change in platelet life span. The thrombocytosis response was absent in IL-6(-/-) mice. DSS treatment also enhanced the platelet aggregation response to thrombin and accelerated thrombus development in WT mice, but not in IL-6(-/-) mice. Exogenous IL-6 administered to WT mice elicited a dose-dependent enhancement of thrombus formation. These findings indicate that IL-6 mediates the thrombocytosis, platelet hyperreactivity, and accelerated thrombus development associated with experimental colitis. The IL-6-dependent colitis-induced thrombocytosis appears to result from an enhancement of thrombopoiesis because platelet life span is unchanged.


Asunto(s)
Trastornos de las Plaquetas Sanguíneas/etiología , Colitis/complicaciones , Interleucina-6/metabolismo , Trombosis/etiología , Animales , Biomarcadores/metabolismo , Trastornos de las Plaquetas Sanguíneas/metabolismo , Colitis/sangre , Colitis/inducido químicamente , Colitis/metabolismo , Sulfato de Dextran , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Agregación Plaquetaria , Recuento de Plaquetas , Trombina/metabolismo , Trombocitosis/etiología , Trombocitosis/metabolismo , Trombosis/metabolismo
11.
Stroke ; 44(4): 1135-43, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23391769

RESUMEN

BACKGROUND AND PURPOSE: Statins are widely used in the primary and secondary prevention of ischemic stroke, but their effects on stroke-induced immunodepression and poststroke infections are elusive. We investigated the effects of simvastatin treatment on stroke-induced splenic atrophy and lung susceptibility to bacterial infection in acute experimental stroke in mice. METHODS: Ischemic stroke was induced by transient middle cerebral artery occlusion, followed by reperfusion. In some experiments, splenectomies were performed 2 weeks before middle cerebral artery occlusion. Animals were randomly assigned to sham and middle cerebral artery occlusion groups treated subcutaneously with vehicle or simvastatin (20 mg/kg per day). Brain infarction, neurological function, brain interferon-γ expression, splenic atrophy and apoptosis, and lung infection were examined. RESULTS: Simvastatin reduced stroke-induced spleen atrophy and splenic apoptosis via increased mitochrondrial antiapoptotic Bcl-2 expression and decreased proapoptotic Bax translocation from cytosol into mitochondria. Splenectomy reduced brain interferon-γ (3 days) and infarct size (5 days) after stroke, and these effects were reversed by adoptive transfer of splenocytes. Simvastatin inhibited brain interferon-γ (3 days) and reduced infarct volume and neurological deficits (5 days) after stroke, and these protective effects were observed not only in naive stroke mice but also in splenectomied stroke mice adoptively transferred with splenocytes. Simvastatin also decreased the stroke-associated lung susceptibility to spontaneous bacterial infection. CONCLUSIONS: Results provide the first direct experimental evidence that simvastatin ameliorates stroke-induced peripheral immunodepression by attenuating spleen atrophy and lung bacterial infection. These findings contribute to a better understanding of the beneficial effects of statins in the treatment of stroke.


Asunto(s)
Atrofia/terapia , Infecciones Bacterianas/metabolismo , Pulmón/microbiología , Simvastatina/farmacología , Bazo/patología , Accidente Cerebrovascular/tratamiento farmacológico , Animales , Anticolesterolemiantes/farmacología , Apoptosis , Infecciones Bacterianas/tratamiento farmacológico , Infarto Encefálico , Modelos Animales de Enfermedad , Humanos , Infarto de la Arteria Cerebral Media , Infecciones , Interferón gamma/metabolismo , Pulmón/efectos de los fármacos , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Bazo/citología , Bazo/efectos de los fármacos , Esplenectomía , Accidente Cerebrovascular/complicaciones , Accidente Cerebrovascular/fisiopatología , Proteína X Asociada a bcl-2/metabolismo
12.
J Exp Med ; 204(7): 1595-601, 2007 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-17562818

RESUMEN

There is growing evidence for an interplay between inflammatory and coagulation pathways in acute and chronic inflammatory diseases. However, it remains unclear whether components of the coagulation pathway, such as tissue factor (TF), contribute to intestinal inflammation, and whether targeting TF will blunt the inflammatory cell recruitment, tissue injury, and enhanced thrombus formation that occur in experimental colitis. Mice were fed 3% dextran sodium sulfate (DSS) to induce colonic inflammation, with some mice receiving a mouse TF-blocking antibody (muTF-Ab). The adhesion of leukocytes and platelets in colonic venules, light/dye-induced thrombus formation in cremaster muscle microvessels, as well as disease activity index, thrombin-antithrombin (TAT) complexes in plasma, and histopathologic changes in the colonic mucosa were monitored in untreated and muTF-Ab-treated colitic mice. In untreated mice, DSS elicited the recruitment of adherent leukocytes and platelets in colonic venules, caused gross and histologic injury, increased plasma TAT complexes, and enhanced thrombus formation in muscle arterioles. muTF-Ab prevented elevation in TAT complexes, reduced blood cell recruitment and tissue injury, and blunted thrombus formation in DSS colitic mice. These findings implicate TF in intestinal inflammation and support an interaction between inflammation and coagulation in experimental colitis.


Asunto(s)
Colitis/patología , Colitis/fisiopatología , Inflamación/fisiopatología , Tromboplastina/fisiología , Trombosis/fisiopatología , Animales , Antitrombinas/fisiología , Coagulación Sanguínea/fisiología , Plaquetas/fisiología , Colon/irrigación sanguínea , Modelos Animales de Enfermedad , Leucocitos/fisiología , Ratones , Microcirculación/fisiología
13.
Blood ; 117(15): 4125-33, 2011 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-21304105

RESUMEN

The microvasculature assumes an inflammatory and procoagulant state in a variety of different diseases, including sickle cell disease (SCD), which may contribute to the high incidence of ischemic stroke in these patients. This study provides evidence for accelerated thrombus formation in arterioles and venules in the cerebral vasculature of mice that express hemoglobin-S (ß(s) mice). Enhanced microvascular thrombosis in ß(s) mice was blunted by immunologic or genetic interventions that target tissue factor, endothelial protein C receptor, activated protein C, or thrombin. Platelets from ß(s) mice also exhibited enhanced aggregation velocity after stimulation with thrombin but not ADP. Neutropenia also protected against the enhanced thrombosis response in ß(s) mice. These results indicate that the cerebral microvasculature is rendered vulnerable to thrombus formation in ß(s) mice via a neutrophil-dependent mechanism that is associated with an increased formation of and enhanced platelet sensitivity to thrombin.


Asunto(s)
Anemia de Células Falciformes , Arterias Cerebrales/metabolismo , Hemoglobina Falciforme/metabolismo , Trombosis Intracraneal , Microcirculación/fisiología , Anemia de Células Falciformes/complicaciones , Anemia de Células Falciformes/genética , Anemia de Células Falciformes/metabolismo , Animales , Plaquetas/metabolismo , Trasplante de Médula Ósea , Modelos Animales de Enfermedad , Hemoglobina Falciforme/genética , Trombosis Intracraneal/etiología , Trombosis Intracraneal/genética , Trombosis Intracraneal/metabolismo , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Mutantes , Neutrófilos/metabolismo , Agregación Plaquetaria/fisiología , Proteína C/metabolismo , Trombina/metabolismo , Tromboplastina/metabolismo
14.
Arterioscler Thromb Vasc Biol ; 32(1): 50-64, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21998133

RESUMEN

OBJECTIVE: It has been shown that CD40-TRAF6 axis in leukocytes plays a significant role in neointimal formation after carotid ligation. Because CD40 and TRAF6 are expressed not only in leukocytes but also in vascular cells, we examined the role of CD40 contributed by vascular wall cells in neointimal formation after carotid ligation in an atherogenic environment. METHODS AND RESULTS: Both CD40 and TRAF6 in medial smooth muscle cells (SMCs) was upregulated significantly at 3 days and more prominently at 7 days after injury in wildtype mice, but the TRAF6 upregulation was abolished in CD40(-/-) mice. In vitro, TRAF6 expression was induced by cytokines (tumor necrosis factor -α, interleukin-1ß) via a NF-κB-dependent manner in wildtype SMCs, but this induction was blocked in CD40-deficient SMCs. Bone marrow chimeras revealed a comparable reduction in neointimal formation and lumen stenosis in mice lacking either vascular wall- or bone marrow-associated CD40. Lacking vascular wall-associated CD40 resulted in a significant reduction in monocyte/macrophage accumulation, NF-κB activation, and multiple proinflammatory mediators (ICAM-1, VCAM-1, MCP-1, MMP-9, tissue factor). In vitro data confirmed that CD40 deficiency or TRAF6 knockdown suppressed CD40L-induced proinflammatory phenotype of SMCs by inhibition of NF-κB activation. Moreover, both in vivo and in vitro data showed that CD40 deficiency prevented injury-induced SMC apoptosis but did not affect SMC proliferation and migration. CONCLUSIONS: CD40 signaling through TRAF6 in vascular SMCs seems to be centrally involved in neointimal formation in a NF-κB-dependent manner. Modulating CD40 signaling on local vascular wall may become a new therapeutic target against vascular restenosis.


Asunto(s)
Antígenos CD40/fisiología , Neointima/etiología , Animales , Apoptosis , Antígenos CD40/deficiencia , Antígenos CD40/genética , Traumatismos de las Arterias Carótidas/patología , Traumatismos de las Arterias Carótidas/fisiopatología , Movimiento Celular , Proliferación Celular , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocitos del Músculo Liso/patología , Miocitos del Músculo Liso/fisiología , Neointima/patología , Neointima/fisiopatología , ARN Interferente Pequeño/genética , Transducción de Señal , Factor 6 Asociado a Receptor de TNF/antagonistas & inhibidores , Factor 6 Asociado a Receptor de TNF/genética , Factor 6 Asociado a Receptor de TNF/fisiología , Regulación hacia Arriba
15.
Nat Med ; 12(6): 621-3, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16680150

RESUMEN

Mice transgenic for antisense Notch and normal mice treated with inhibitors of the Notch-activating enzyme gamma-secretase showed reduced damage to brain cells and improved functional outcome in a model of focal ischemic stroke. Notch endangers neurons by modulating pathways that increase their vulnerability to apoptosis, and by activating microglial cells and stimulating the infiltration of proinflammatory leukocytes. These findings suggest that Notch signaling may be a therapeutic target for treatment of stroke and related neurodegenerative conditions.


Asunto(s)
Isquemia Encefálica/patología , Encéfalo/patología , Endopeptidasas/metabolismo , Receptor Notch1/metabolismo , Transducción de Señal/fisiología , Accidente Cerebrovascular/patología , Secretasas de la Proteína Precursora del Amiloide , Animales , Apoptosis , Ácido Aspártico Endopeptidasas/antagonistas & inhibidores , Ácido Aspártico Endopeptidasas/genética , Ácido Aspártico Endopeptidasas/metabolismo , Encéfalo/citología , Encéfalo/metabolismo , Isquemia Encefálica/metabolismo , Isquemia Encefálica/terapia , Células Cultivadas , Endopeptidasas/genética , Inhibidores Enzimáticos/metabolismo , Humanos , Leucocitos/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Microglía/metabolismo , Neuronas/citología , Neuronas/metabolismo , Péptidos/genética , Péptidos/metabolismo , Ratas , Receptor Notch1/genética , Daño por Reperfusión , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/terapia , Resultado del Tratamiento
16.
J Physiol ; 590(5): 1023-34, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22183721

RESUMEN

Inflammation is an underlying feature of a variety of human diseases. An important manifestation of this pathophysiological response is microvascular dysfunction, which includes the activation of vascular endothelial cells, and circulating leucocytes and platelets. While endothelial cells and leucocytes are widely accepted as critical players in the microvascular alterations induced by inflammation, recent attention has focused on the modulatory role of platelets, which act both as effector and target cells in inflamed microvessels. Evidence is presented to demonstrate the capacity for 'cross-talk' between platelets and other cells (endothelial cells, leucocytes) that contribute to an inflammatory response, and to illustrate the pathophysiological consequences of these interactions of platelets with other cells within the microvasculature.


Asunto(s)
Plaquetas/fisiología , Endotelio Vascular/fisiopatología , Inflamación/fisiopatología , Microvasos/fisiopatología , Animales , Células Endoteliales/fisiología , Humanos , Leucocitos/fisiología
17.
J Exp Med ; 203(11): 2441-50, 2006 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-17043144

RESUMEN

Inflammatory cells can either promote or inhibit tumor growth. Here we studied whether CD40, a key molecule for adaptive immune response, has any role in mammary carcinogenesis of BALB/NeuT transgenic tumor-prone mice. We transferred the HER2/neu oncogene into CD40-null background to obtain the CD40-KO/NeuT strain. CD40-KO/NeuT mice showed delayed tumor onset and reduced tumor multiplicity. BM (BM) transplantation experiments excluded a role of BM-derived cells in the reduced tumorigenicity associated with CD40 deficiency. Rather, CD40 expressed by endothelial cells (ECs) takes part to the angiogenic process. Accordingly, large vessels, well organized around the tumor lobular structures, characterize BALB/NeuT tumors, whereas tiny numerous vessels with scarce extracellular matrix are dispersed in the parenchyma of poorly organized CD40-KO/NeuT tumors. Activated platelets, which may interact with and activate ECs, are a possible source of CD40L. Their localization within tumor vessels prompted the idea of treating BALB/NeuT and CD40-KO/NeuT mice chronically with the anti-platelet drug clopidogrel, known to inhibit platelet CD40L expression. Treatment of BALB/NeuT mice reduced tumor growth to a level similar to CD40-deficient mice, whereas CD40-KO/NeuT mice treated or not showed the same attenuated tumor outgrowth, indicating that activated platelets are the likely source of CD40L in this model. Collectively, these data point to a participation of CD40/CD40L in the angiogenic processes associated with mammary carcinogenesis of BALB/NeuT mice.


Asunto(s)
Antígenos CD40/fisiología , Proliferación Celular , Endotelio Vascular/inmunología , Endotelio Vascular/metabolismo , Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/patología , Animales , Antígenos CD40/genética , Endotelio Vascular/patología , Femenino , Neoplasias Mamarias Experimentales/irrigación sanguínea , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones Transgénicos , Ratas
18.
Stroke ; 42(7): 2033-44, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21546487

RESUMEN

BACKGROUND AND PURPOSE: Phosphoinositide 3-kinase (PI3K)-γ is linked to inflammation and oxidative stress. This study was conducted to investigate the role of the PI3Kγ in the blood-brain barrier dysfunction and brain damage induced by focal cerebral ischemia/reperfusion. METHODS: Wild-type and PI3Kγ knockout mice were subjected to middle cerebral artery occlusion (60 minutes) followed by reperfusion. Evans blue leakage, brain edema, infarct volumes, and neurological deficits were examined. Oxidative stress, neutrophil infiltration, and matrix metallopeptidase-9 were assessed. Activation of nuclear factor-κB and expression of proinflammatory and pro-oxidative genes were studied. RESULTS: PI3Kγ deficiency significantly reduced blood-brain barrier permeability and brain edema formation, which were time-dependently correlated with preventing the degradation of the tight junction protein, claudin-5, and the basal lamina protein, collagen IV, and the phosphorylation of myosin light chain in brain microvessels. PI3Kγ deficiency suppressed ischemia/reperfusion-induced nuclear factor-κB p65 (Ser536) phosphorylation and the expression of the pro-oxidant enzyme NADPH oxidase (Nox1, Nox2, and Nox4) and proinflammatory adhesion molecules (E- and P-selectin, intercellular adhesion molecule-1) at different time points. These molecular changes were associated with significant inhibition of oxidative stress (superoxide production and malondialdehyde content), neutrophil infiltration, and matrix metallopeptidase-9 expression/activity in PI3Kγ knockout mice. Eventually, PI3Kγ deficiency significantly reduced infarct volumes and neurological scores at 24 hours after ischemia/reperfusion. CONCLUSIONS: Our results provide the first direct demonstration that PI3Kγ plays a significant role in ischemia/reperfusion-induced blood-brain barrier disruption and brain damage. Future studies need to explore PI3Kγ as a potential target for stroke therapy.


Asunto(s)
Barrera Hematoencefálica/fisiopatología , Fosfatidilinositol 3-Quinasa Clase Ib/metabolismo , Accidente Cerebrovascular/enzimología , Animales , Encéfalo/patología , Infarto de la Arteria Cerebral Media/patología , Inflamación , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/metabolismo , Neutrófilos/metabolismo , Estrés Oxidativo , Daño por Reperfusión
19.
Stroke ; 42(3): 806-11, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21257828

RESUMEN

BACKGROUND AND PURPOSE: The therapeutic potential of bone marrow stromal cells (BMSCs) has been demonstrated in different models of stroke. Although it is well established that BMSCs selectively migrate to the site of brain injury, the mechanisms underlying this process are poorly understood. This study addresses the hypothesis that selectins mediate the recruitment of BMSCs into the postischemic cerebral microvasculature. METHODS: Focal ischemic stroke was induced by middle cerebral artery occlusion and reperfusion. Cell recruitment was monitored using either fluorescent- or radiolabeled BMSCs detected by intravital microscopy or tissue radioactivity. Mice were treated with either a blocking antibody against P- or E-selectin or with the nonselective selectin antagonist, fucoidin. The role of CD44 in cell recruitment was evaluated using BMSCs from CD44 knockout mice. RESULTS: Middle cerebral artery occlusion and reperfusion was associated with a significantly increased adhesion of BMSCs in cerebral venules compared with sham mice. Immunoneutralization of either E- or P-selectin blocked the middle cerebral artery occlusion and reperfusion-induced recruitment of adherent BMSCs. An attenuated recruitment response in the postischemic hemisphere was also noted after fucoidin treatment or administration of CD44-deficient BMSCs. CONCLUSIONS: Cerebral vascular endothelium assume a proadhesive phenotype after ischemic stroke that favors the recruitment of BMSCs, which use both P- and E-selectin to home into the infarct site. CD44 may serve as the critical ligand for selectin-mediated BMSC recruitment.


Asunto(s)
Células de la Médula Ósea/metabolismo , Isquemia Encefálica/metabolismo , Movimiento Celular/fisiología , Selectina E/fisiología , Células Madre Hematopoyéticas/metabolismo , Microvasos/metabolismo , Selectina-P/fisiología , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/patología , Isquemia Encefálica/patología , Adhesión Celular/genética , Adhesión Celular/fisiología , Movimiento Celular/genética , Células Cultivadas , Selectina E/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/patología , Humanos , Receptores de Hialuranos/genética , Receptores de Hialuranos/metabolismo , Receptores de Hialuranos/fisiología , Inmunofenotipificación , Ligandos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Noqueados , Ratones Transgénicos , Microvasos/citología , Microvasos/patología , Selectina-P/metabolismo , Distribución Aleatoria , Células del Estroma/citología , Células del Estroma/metabolismo , Células del Estroma/patología
20.
Am J Pathol ; 177(6): 2774-81, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20971730

RESUMEN

Inflammatory bowel diseases (IBDs) are associated with an increased risk for thromboembolism, which is often manifested as deep vein thrombosis or pulmonary embolism, at extra-intestinal sites. Although some of the cytokines that contribute to IBD pathogenesis are also known to alter the coagulation pathway, it remains unclear whether these mediators also contribute to the extra-intestinal thrombosis often associated with IBD. The objective of this study is to evaluate the role of interleukin (IL)-1ß in enhanced extra-intestinal thrombosis observed in mice with dextran sodium sulfate (DSS)-induced colitis. IL-1ß concentrations were measured in plasma, colon, and skeletal muscle of wild-type (WT) control and colitic mice. Microvascular thrombosis was induced in cremaster muscle microvessels by using a light/dye injury model. The effects of exogenous IL-1ß on thrombus formation were determined in control WT mice. DSS-induced thrombogenesis was evaluated in WT mice treated with an IL-1ß antibody and in IL-1 receptor-deficient (IL-1r(-/-)) mice. DSS-induced colonic inflammation in WT mice was associated with enhanced thrombus formation in arterioles. IL-1ß concentrations were elevated in inflamed colon and skeletal muscle. Exogenous IL-1ß enhanced thrombosis in control mice in a dose-dependent manner. DSS colitic mice treated with the IL-1ß antibody as well as IL-1r(-/-) mice exhibited significantly blunted thrombogenic responses. These findings implicate IL-1ß as a mediator of enhanced microvascular thromboses that occur in extra-intestinal tissues during colonic inflammation.


Asunto(s)
Enterocolitis/complicaciones , Interleucina-1beta/fisiología , Trombosis/etiología , Animales , Colon/química , Colon/metabolismo , Sulfato de Dextran , Modelos Animales de Enfermedad , Enterocolitis/inducido químicamente , Enterocolitis/genética , Enterocolitis/metabolismo , Genes RAG-1 , Interleucina-1beta/análisis , Interleucina-1beta/metabolismo , Mucosa Intestinal/metabolismo , Intestinos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Esquelético/química , Músculo Esquelético/metabolismo , Factor 88 de Diferenciación Mieloide/genética , Concentración Osmolar , Receptores de Interleucina-1/genética , Trombosis/genética , Trombosis/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda