Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Int J Exp Pathol ; 100(2): 72-82, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30887659

RESUMEN

Vitronectin (VN), an extracellular matrix protein, is a promising immune biomarker of non-alcoholic steatohepatitis (NASH); however, its precise function remains unclear. This study investigated how VN deficiency contributes to the development of NASH. Towards this aim, wild-type (WT) and VN-/- mice were fed with a choline-deficient, L-amino acid-defined, high-fat diet (CDAHFD) for 6 and 10 weeks to induce NASH, and the livers were isolated. In WT mice fed with CDAHFD for 6 and 10 weeks, the expression of Vn mRNA and protein was up-regulated compared with that in mice fed with the MF control diet, indicating that VN is regulated in NASH condition. VN-/- mice showed decreased picrosirius red staining in the liver area and Col1a2 mRNA expression levels, compared with WT mice, indicating that the severity of hepatic fibrosis is attenuated in the CDAHFD-fed VN-/- mice. In addition, VN deficiency did not affect the area of lipid droplets in haematoxylin-eosin staining and the mRNA expression levels of fatty acid synthases, Srebp, Acc and Fas in the CDAHFD-fed mice. Moreover, VN deficiency decreased the inflammation score and the mRNA expression levels of Cd11b and F4/80, macrophage markers, as well as Tnf-α and Il-1ß, inflammatory cytokines in the CDAHFD-fed mice. Furthermore, VN deficiency decreased the protein and mRNA expression levels of α-smooth muscle actin in the CDAHFD-fed mice, suggesting that VN deficiency inhibits the activation of hepatic stellate cells (HSCs). Our findings indicate that VN contributes to the development of fibrosis in the NASH model mice via modulation of the inflammatory reaction and activation of HSCs.


Asunto(s)
Cirrosis Hepática/metabolismo , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Vitronectina/fisiología , Animales , Deficiencia de Colina/complicaciones , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Regulación de la Expresión Génica , Células Estrelladas Hepáticas/fisiología , Metabolismo de los Lípidos/fisiología , Hígado/metabolismo , Hígado/patología , Cirrosis Hepática/etiología , Cirrosis Hepática/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Enfermedad del Hígado Graso no Alcohólico/complicaciones , Enfermedad del Hígado Graso no Alcohólico/patología , ARN Mensajero/genética , Vitronectina/deficiencia , Vitronectina/genética
2.
Arterioscler Thromb Vasc Biol ; 35(1): 111-20, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25378411

RESUMEN

OBJECTIVE: Plasminogen activator inhibitor-1 (PAI-1) regulates angiogenesis via effects on extracellular matrix proteolysis and cell adhesion. However, no previous study has implicated PAI-1 in controlling vascular endothelial growth factor (VEGF) signaling. We tested the hypothesis that PAI-1 downregulates VEGF receptor-2 (VEGFR-2) activation by inhibiting a vitronectin-dependent cooperative binding interaction between VEGFR-2 and αVß3. APPROACH AND RESULTS: We studied effects of PAI-1 on VEGF signaling in human umbilical vein endothelial cells. PAI-1 inhibited VEGF-induced phosphorylation of VEGFR-2 in human umbilical vein endothelial cells grown on vitronectin, but not on fibronectin or collagen. PAI-1 inhibited the binding of VEGFR-2 to ß3 integrin, VEGFR-2 endocytosis, and intracellular signaling pathways downstream of VEGFR-2. The anti-VEGF effect of PAI-1 was mediated by 2 distinct pathways, one requiring binding to vitronectin and another requiring binding to very low-density lipoprotein receptor. PAI-1 inhibited VEGF-induced angiogenesis in vitro and in vivo, and pharmacological inhibition of PAI-1 promoted collateral arteriole development and recovery of hindlimb perfusion after femoral artery interruption. CONCLUSIONS: PAI-1 inhibits activation of VEGFR-2 by VEGF by disrupting a vitronectin-dependent proangiogenic binding interaction involving αVß3 and VEGFR-2. These results broaden our understanding of the roles of PAI-1, vitronectin, and endocytic receptors in regulating VEGFR-2 activation and suggest novel therapeutic strategies for regulating VEGF signaling.


Asunto(s)
Células Endoteliales/metabolismo , Integrina alfaVbeta3/metabolismo , Músculo Esquelético/irrigación sanguínea , Neovascularización Fisiológica , Inhibidor 1 de Activador Plasminogénico/metabolismo , Receptor Cross-Talk , Transducción de Señal , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Adhesión Celular , Movimiento Celular , Células Cultivadas , Modelos Animales de Enfermedad , Endocitosis , Células Endoteliales/efectos de los fármacos , Miembro Posterior , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Ácidos Indolacéticos/administración & dosificación , Isquemia/metabolismo , Isquemia/fisiopatología , Isquemia/prevención & control , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Neovascularización Fisiológica/efectos de los fármacos , Fosforilación , Inhibidor 1 de Activador Plasminogénico/genética , Interferencia de ARN , Receptor Cross-Talk/efectos de los fármacos , Receptores de LDL/metabolismo , Proteínas Recombinantes/metabolismo , Inhibidores de Serina Proteinasa/administración & dosificación , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Transfección , Vitronectina/deficiencia , Vitronectina/genética
3.
J Immunol ; 190(5): 2273-81, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23345331

RESUMEN

Effective removal of apoptotic cells, particularly apoptotic neutrophils, is essential for the successful resolution of acute inflammatory conditions. In these experiments, we found that whereas interaction between vitronectin and integrins diminished the ability of macrophages to ingest apoptotic cells, interaction between vitronectin with urokinase-type plasminogen activator receptor (uPAR) on the surface of apoptotic cells also had equally important inhibitory effects on efferocytosis. Preincubation of vitronectin with plasminogen activator inhibitor-1 eliminated its ability to inhibit phagocytosis of apoptotic cells. Similarly, incubation of apoptotic cells with soluble uPAR or Abs to uPAR significantly diminished efferocytosis. In the setting of LPS-induced ALI, enhanced efferocytosis and decreased numbers of neutrophils were found in bronchoalveolar lavage obtained from vitronectin-deficient (vtn(-/-)) mice compared with wild type (vtn(+/+)) mice. Furthermore, there was increased clearance of apoptotic vtn(-/-) as compared with vtn(+/+) neutrophils after introduction into the lungs of vtn(-/-) mice. Incubation of apoptotic vtn(-/-) neutrophils with purified vitronectin before intratracheal instillation decreased efferocytosis in vivo. These findings demonstrate that the inhibitory effects of vitronectin on efferocytosis involve interactions with both the engulfing phagocyte and the apoptotic target cell.


Asunto(s)
Lesión Pulmonar Aguda/inmunología , Apoptosis/efectos de los fármacos , Macrófagos Peritoneales/efectos de los fármacos , Neutrófilos/efectos de los fármacos , Fagocitosis/efectos de los fármacos , Timocitos/efectos de los fármacos , Vitronectina/inmunología , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/metabolismo , Lesión Pulmonar Aguda/patología , Animales , Anticuerpos/farmacología , Apoptosis/inmunología , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/inmunología , Recuento de Células , Técnicas de Cocultivo , Femenino , Lipopolisacáridos , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/inmunología , Neutrófilos/patología , Fagocitosis/inmunología , Inhibidor 1 de Activador Plasminogénico/farmacología , Receptores del Activador de Plasminógeno Tipo Uroquinasa/antagonistas & inhibidores , Receptores del Activador de Plasminógeno Tipo Uroquinasa/genética , Receptores del Activador de Plasminógeno Tipo Uroquinasa/inmunología , Timocitos/inmunología , Timocitos/patología , Vitronectina/deficiencia , Vitronectina/genética
4.
J Vasc Res ; 51(2): 110-7, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24603119

RESUMEN

BACKGROUND: Vascular endothelial growth factor (VEGF) plays a key role in regulating angiogenesis, and this process is largely dependent on the newly formed extracellular matrix (ECM). The levels of vitronectin (VN) are increased in patients with various cardiovascular diseases. A role for VN in regulating VEGF-induced angiogenesis has not been previously reported. We tested the hypothesis that VN regulates VEGFR-2 activation via effects on αvß3, thus contributing to angiogenesis. METHODS: We used a 3-dimensional angiogenesis assay, and examined the effects of VN on VEGF-mediated angiogenesis in aortic endothelial cells (ECs) isolated from wild-type and VN-deficient mice. RESULTS: The addition of multimeric VN significantly enhanced VEGF-induced increases in EC migration and capillary formation. In vitro, Vn(-/-) ECs migrated significantly slower than wild-type ECs. The addition of VN to Vn(-/-) ECs increased EC migration and augmented the promigratory effect of VEGF in a manner that involved VEGFR-2 and Src signaling. Analysis of the mechanisms involved revealed that multimeric VN, but not monomeric VN, binds VEGF and enhances VEGF-induced VEGFR-2/Src activation in ECs. CONCLUSION: These results underscore the importance of VN in the regulation of angiogenesis induced by VEGF.


Asunto(s)
Células Endoteliales/metabolismo , Neovascularización Fisiológica , Factor A de Crecimiento Endotelial Vascular/metabolismo , Vitronectina/metabolismo , Animales , Movimiento Celular , Proliferación Celular , Células Cultivadas , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Integrina alfaVbeta3/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Interferencia de ARN , Proteínas Recombinantes/metabolismo , Transducción de Señal , Transfección , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Vitronectina/deficiencia , Vitronectina/genética , Familia-src Quinasas/genética , Familia-src Quinasas/metabolismo
5.
Arterioscler Thromb Vasc Biol ; 31(8): 1781-7, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21571686

RESUMEN

OBJECTIVE: The role of plasminogen activator inhibitor-1 (PAI-1) in vein graft (VG) remodeling is undefined. We examined the effect of PAI-1 on VG intimal hyperplasia and tested the hypothesis that PAI-1 regulates VG thrombin activity. METHODS AND RESULTS: VGs from wild-type (WT), Pai1(-/-), and PAI-1-transgenic mice were implanted into WT, Pai1(-/-), or PAI-1-transgenic arteries. VG remodeling was assessed 4 weeks later. Intimal hyperplasia was significantly greater in PAI-1-deficient mice than in WT mice. The proliferative effect of PAI-1 deficiency was retained in vitronectin-deficient mice, suggesting that PAI-1's antiproteolytic function plays a key role in regulating intimal hyperplasia. Thrombin-induced proliferation of PAI-1-deficient venous smooth muscle cells (SMC) was significantly greater than that of WT SMC, and thrombin activity was significantly higher in PAI-1-deficient VGs than in WT VGs. Increased PAI-1 expression, which has been associated with obstructive VG disease, did not increase intimal hyperplasia. CONCLUSIONS: Decreased PAI-1 expression (1) promotes intimal hyperplasia by pathways that do not require vitronectin and (2) increases thrombin activity in VG. PAI-1 overexpression, although it promotes SMC migration in vitro, did not increase intimal hyperplasia. These results challenge the concept that PAI-1 drives nonthrombotic obstructive disease in VG and suggest that PAI-1's antiproteolytic function, including its antithrombin activity, inhibits intimal hyperplasia.


Asunto(s)
Serpina E2/fisiología , Vena Cava Inferior/trasplante , Animales , Movimiento Celular , Proliferación Celular , Puente de Arteria Coronaria/efectos adversos , Fibrina/metabolismo , Fibrinógeno/metabolismo , Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Miocitos del Músculo Liso/patología , Miocitos del Músculo Liso/fisiología , Neointima/etiología , Neointima/patología , Neointima/fisiopatología , Serpina E2/deficiencia , Serpina E2/genética , Túnica Íntima/patología , Vena Cava Inferior/patología , Vitronectina/deficiencia
6.
Eur J Neurosci ; 30(8): 1451-60, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19811533

RESUMEN

The mammalian circadian clock in the suprachiasmatic nucleus (SCN) maintains environmental synchrony through light signals transmitted by glutamate released from retinal ganglion terminals. Brain-derived neurotrophic factor (BDNF) is required for light/glutamate to reset the clock. In the hippocampus, BDNF is activated by the extracellular protease, plasmin, which is produced from plasminogen by tissue-type plasminogen activator (tPA). We provide data showing expression of proteins from the plasminogen activation cascade in the SCN and their involvement in circadian clock phase-resetting. Early night glutamate application to SCN-containing brain slices resets the circadian clock. Plasminogen activator inhibitor-1 (PAI-1) blocked these shifts in slices from wild-type mice but not mice lacking its stabilizing protein, vitronectin (VN). Plasmin, but not plasminogen, prevented inhibition by PAI-1. Both plasmin and active BDNF reversed alpha(2)-antiplasmin inhibition of glutamate-induced shifts. alpha(2)-Antiplasmin decreased the conversion of inactive to active BDNF in the SCN. Finally, both tPA and BDNF allowed daytime glutamate-induced phase-resetting. Together, these data are the first to demonstrate expression of these proteases in the SCN, their involvement in modulating photic phase-shifts, and their activation of BDNF in the SCN, a potential 'gating' mechanism for photic phase-resetting. These data also demonstrate a functional interaction between PAI-1 and VN in adult brain. Given the usual association of these proteins with the extracellular matrix, these data suggest new lines of investigation into the locations and processes modulating mammalian circadian clock phase-resetting.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Ritmo Circadiano/efectos de los fármacos , Fibrinolisina/metabolismo , Núcleo Supraquiasmático/efectos de los fármacos , Núcleo Supraquiasmático/fisiología , Activador de Tejido Plasminógeno/metabolismo , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/genética , Potenciales de Acción/fisiología , Análisis de Varianza , Animales , Factor Neurotrófico Derivado del Encéfalo/farmacología , Ritmo Circadiano/fisiología , Interacciones Farmacológicas , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Ácido Glutámico/farmacología , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/efectos de los fármacos , Neuronas/fisiología , Inhibidor 1 de Activador Plasminogénico/metabolismo , Núcleo Supraquiasmático/citología , Activador de Tejido Plasminógeno/farmacología , Vitronectina/deficiencia
7.
J Thromb Haemost ; 15(12): 2451-2460, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29028290

RESUMEN

Essentials Vitronectin (VN) is produced by smooth muscle cells (SMCs) and promotes neointima formation. We studied the regulation of vascular VN expression by plasminogen activator inhibitor-1 (PAI-1). PAI-1 stimulates VN gene expression in SMCs by binding LDL receptor-related protein 1. Stimulation of VN gene expression may be a mechanism by which PAI-1 controls vascular remodeling. SUMMARY: Background Increased expression of vitronectin (VN) by smooth muscle cells (SMCs) promotes neointima formation after vascular injury, and may contribute to chronic vascular diseases, such as atherosclerosis. However, the molecular regulation of vascular VN expression is poorly defined. Given the overlapping expression profiles and functions of VN and plasminogen activator inhibitor (PAI)-1, we hypothesized that PAI-1 regulates vascular VN expression. Objectives To determine whether PAI-1 regulates VN expression in SMCs and in vivo. Methods The effects of genetic alterations in PAI-1 expression, pharmacologic PAI-1 inhibition and recombinant PAI-1 on SMC VN expression were studied, and vascular VN expression in wild-type (WT) and PAI-1-deficient mice was assessed. Results VN expression was significantly lower in PAI-1-deficient SMCs and significantly increased in PAI-1-overexpressing SMCs. PAI-1 small interfering RNA and pharmacologic PAI-1 inhibition significantly decreased SMC VN expression. Recombinant PAI-1 stimulated VN expression by binding LDL receptor-related protein-1 (LRP1), but another LRP1 ligand, α2 -macroglobulin, did not. As compared with WT controls, carotid artery VN expression was significantly lower in PAI-1-deficient mice and significantly higher in PAI-1-transgenic mice. In a vein graft (VG) model of intimal hyperplasia, VN expression was significantly attenuated in PAI-1-deficient VGs as compared with WT controls. The plasma VN concentration was significantly decreased in PAI-1-deficient mice versus WT controls at 4 weeks, but not at 5 days or 8 weeks, after surgery. Conclusions PAI-1 stimulates SMC VN expression by binding LRP1, and controls vascular VN expression in vivo. Autocrine regulation of vascular VN expression by PAI-1 may play important roles in vascular homeostasis and pathologic vascular remodeling.


Asunto(s)
Músculo Liso Vascular/metabolismo , Serpina E2/metabolismo , Vitronectina/metabolismo , Animales , Células Cultivadas , Regulación de la Expresión Génica , Humanos , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Neointima/etiología , Neointima/genética , Neointima/metabolismo , ARN Interferente Pequeño/genética , Receptores de LDL/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Serpina E2/deficiencia , Serpina E2/genética , Proteínas Supresoras de Tumor/metabolismo , Remodelación Vascular , Vitronectina/deficiencia , Vitronectina/genética
8.
J Neurotrauma ; 34(22): 3183-3191, 2017 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-28683586

RESUMEN

Vitronectin (VN), one of the serum proteins, is known to be involved in the regulation of blood coagulation, fibrinolysis, and cell migration. It has been proposed that the regulation of fibrinolysis by VN promotes the blood-brain barrier (BBB) recovery from brain injuries such as traumatic injury and subarachnoid hemorrhage. The effects of VN on fibrinolysis in the injured brain remain unclear, however. We examined the effects of VN on the fibrinolytic system in the stab-wounded cerebral cortex of VN-knockout (KO) mice. First, hemorrhage and recovery from BBB breakdown in the wounded regions were assessed by serum immunoglobulin G (IgG) extravasation. The level of IgG extravasation increased 3-7 days after the stab wound (D3-7) in the cortex of VN-KO mice, compared with that in wild type mice, indicating that VN deficiency inhibited the recovery from BBB breakdown. The VN deficiency decreased fibrin fiber deposition at D1-3, suggesting that VN deficiency tilts the balance between fibrinogenesis and fibrinolysis toward fibrinolysis. Next, the effects of VN deficiency on the fibrinolytic factors were analyzed in the stab-wounded cortex. The VN deficiency impaired the activity of plasminogen activator inhibitor-1, an inhibitor of the fibrinolytic system, at D3-5. Further, VN deficiency up-regulated the mRNA and protein expression levels of tissue-type plasminogen activator, and urokinase-type plasminogen activator. These results demonstrate that VN contributes to the regulation of the fibrinolytic system and recovery from BBB breakdown in the wounded brain.


Asunto(s)
Barrera Hematoencefálica/lesiones , Lesiones Encefálicas/metabolismo , Corteza Cerebral/lesiones , Corteza Cerebral/metabolismo , Fibrina/metabolismo , Fibrinólisis/fisiología , Inhibidor 1 de Activador Plasminogénico/metabolismo , Activador de Tejido Plasminógeno/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Vitronectina/fisiología , Animales , Lesiones Encefálicas/etiología , Modelos Animales de Enfermedad , Traumatismos Penetrantes de la Cabeza/complicaciones , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Mensajero/metabolismo , Factores de Tiempo , Vitronectina/deficiencia
9.
J Thromb Haemost ; 3(10): 2290-5, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16194205

RESUMEN

OBJECTIVE: The role of vitronectin (VN) in thrombosis is not fully understood, primarily because this adhesive glycoprotein not only stabilizes plasminogen activator inhibitor 1 (PAI-1) and thus protects fibrin from premature lysis, but also because it binds to platelet integrins and may influence platelet aggregation. The absence of quantitative approaches to characterize the thrombi formed in animal models under different conditions further complicates this analysis. METHODS: In this report, we describe a more comprehensive approach to assess the stability of thrombi formed in mice deficient in PAI-1 (PAI-1(-/-)), VN (VN(-/-)) or both (PAI-1(-/-)/VN(-/-)). RESULTS: We observed that all deficient mice developed unstable thrombi compared with wild type (WT) mice. Thus, only 31% of the thrombi formed in WT mice were unstable compared with 74% of PAI-1(-/-), 80% of VN(-/-), and 87% of PAI-1(-/-)/VN(-/-) mice. In this regard, the average number of emboli per WT mouse was significantly lower (0.55) compared with VN(-/-) (2.66), PAI-1(-/-) (2.1), and VN(-/-)/PAI-1(-/-) (2.35) mice. Finally, the total duration of complete vascular occlusion was higher and the rate of vascular patency was lower in the WT mice compared with the deficient mice. CONCLUSIONS: Taken together, these observations indicate that the thrombotic phenotype of mice with a combined deficiency in PAI-1 and VN does not differ significantly from the phenotype of mice with deficiencies in only PAI-1 or VN. This observation suggests that PAI-1 and VN may influence thrombus stability by regulating a common pathway.


Asunto(s)
Inhibidor 1 de Activador Plasminogénico/deficiencia , Trombosis/etiología , Vitronectina/deficiencia , Animales , Cloruros , Compuestos Férricos , Ratones , Ratones Noqueados , Fenotipo , Inhibidor 1 de Activador Plasminogénico/fisiología , Trombosis/inducido químicamente , Factores de Tiempo , Grado de Desobstrucción Vascular , Vitronectina/fisiología
10.
Arterioscler Thromb Vasc Biol ; 22(6): 934-9, 2002 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-12067901

RESUMEN

We examined the roles of vitronectin and plasminogen activator inhibitor-1 (PAI-1) in neointima development. Neointima formation after carotid artery ligation or chemical injury was significantly greater in wild-type mice than in vitronectin-deficient (Vn(-/-)) mice. Vascular smooth muscle cell (VSMC) proliferation did not differ between groups, suggesting that vitronectin promoted neointima development by enhancing VSMC migration. Neointima formation was significantly attenuated in PAI-1-deficient (PAI-1(-/-)) mice compared with control mice. Because intravascular fibrin may function as a provisional matrix for invading VSMCs, we examined potential mechanisms by which vitronectin and PAI-1 regulate fibrin stability and fibrin-VSMC interactions. Inhibition of activated protein C by PAI-1 was markedly attenuated in vitronectin-deficient plasma. The capacity of PAI-1 to inhibit clot lysis was significantly attenuated in vitronectin-deficient plasma, and this effect was not explained simply by the PAI-1-stabilizing properties of vitronectin. The adhesion and spreading of VSMCs were significantly greater on wild-type plasma clots and PAI-1-deficient plasma clots than on vitronectin-deficient plasma clots. We conclude that endogenous levels of vitronectin and PAI-1 enhance neointima formation in response to vascular occlusion or injury. Their effects may be mediated to a significant extent by their capacity to promote intravascular fibrin deposition and by the capacity of vitronectin to enhance VSMC-fibrin interactions.


Asunto(s)
Neovascularización Fisiológica/fisiología , Inhibidor 1 de Activador Plasminogénico/fisiología , Túnica Íntima/fisiopatología , Vitronectina/fisiología , Animales , Arterias Carótidas/fisiopatología , Arterias Carótidas/cirugía , Traumatismos de las Arterias Carótidas/inducido químicamente , Línea Celular , Movimiento Celular/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Músculo Liso Vascular/citología , Músculo Liso Vascular/embriología , Músculo Liso Vascular/fisiopatología , Neovascularización Fisiológica/genética , Inhibidor 1 de Activador Plasminogénico/deficiencia , Inhibidor 1 de Activador Plasminogénico/metabolismo , Ratas , Vitronectina/deficiencia , Vitronectina/metabolismo
11.
Arterioscler Thromb Vasc Biol ; 22(12): 1978-83, 2002 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-12482822

RESUMEN

OBJECTIVE: We previously reported that plasminogen activator inhibitor 1 (PAI-1), in the presence of vitronectin (VN), inhibits thrombin activity in vitro. Furthermore, we demonstrated in human atherosclerotic plaques the colocalization of thrombin, PAI-1, and VN, as well as activity of thrombin and PAI-1. Here, we show that PAI-1 is a local thrombin inhibitor in vivo. METHODS AND RESULTS: We used the murine carotid artery ligation model to assess the role of PAI-1 and VN in stenosis by using PAI-1-deficient (PAI-1(-/-)) and VN(-/-) mice. Ligation resulted in a smooth muscle cell (SMC)-rich intima without infiltrating cells. We show that PAI-1(-/-) and VN(-/-) mice generate a larger intima than wild-type mice as the result of more extensive SMC proliferation, as evidenced by cell counting and staining for proliferating cell-nuclear antigen. CONCLUSIONS: In PAI-1(-/-) mice, excessive intima formation is prevented by the thrombin-specific inhibitor hirudin. Finally, immunohistochemical analysis revealed PAI-1, VN, and (pro)thrombin antigen in intimal lesions. Our observations are compatible with inhibition of thrombin-mediated SMC proliferation by PAI-1/VN complexes.


Asunto(s)
Arterias Carótidas , Estenosis Carotídea/metabolismo , Estenosis Carotídea/prevención & control , Modelos Animales de Enfermedad , Inhibidor 1 de Activador Plasminogénico/fisiología , Vitronectina/fisiología , Animales , Arterias Carótidas/cirugía , División Celular/fisiología , Tejido Conectivo/química , Hirudinas/farmacología , Inmunohistoquímica , Ligadura/métodos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/citología , Inhibidor 1 de Activador Plasminogénico/deficiencia , Inhibidor 1 de Activador Plasminogénico/inmunología , Inhibidor 1 de Activador Plasminogénico/metabolismo , Protrombina/inmunología , Protrombina/metabolismo , Túnica Íntima/química , Túnica Íntima/citología , Túnica Íntima/efectos de los fármacos , Túnica Íntima/fisiología , Túnica Media/química , Vitronectina/deficiencia , Vitronectina/inmunología , Vitronectina/metabolismo
12.
Surgery ; 127(6): 696-704, 2000 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10840366

RESUMEN

BACKGROUND: Vitronectin has several putative functions including regulating hemostasis, cell adhesion, and cell migration. However, the targeted deletion of vitronectin in mice results in normal development and normal coagulation parameters. To determine whether vitronectin may be necessary for nondevelopmental processes, we examined the response to tissue injury in vitronectin-null mice. METHODS: We examined wound healing in control and vitronectin-null mice by healing rate, zymography, reverse zymography, and Western blots. RESULTS: We found that dermal wound healing was slightly delayed in mice lacking vitronectin. More importantly, we found extensive areas of delayed hemorrhage near the sprouting tips of microvessels between days 7 and 14, which temporally coincided with increased urokinase-type plasminogen activator and tissue-type plasminogen activator activity by zymography. Though Western blots confirmed the presence of plasminogen activator inhibitor-1 protein throughout wound repair and reverse zymograms showed decreased plasminogen activator inhibitor-1 activity between days 7 and 14. CONCLUSIONS: Loss of vitronectin in mice was associated with changes in the fibrinolytic balance, and this may have led to focal sites of delayed hemorrhage. The mechanism that resulted in decreased angiogenesis and the formation of larger blood vessels in response to tissue injury remains unknown. This study suggests that vitronectin may have several distinct functions that are not required for normal development but are manifested in response to tissue injury.


Asunto(s)
Fibrinólisis/fisiología , Neovascularización Fisiológica , Vitronectina/deficiencia , Heridas y Lesiones/fisiopatología , Animales , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microcirculación/fisiología , Vitronectina/genética , Vitronectina/fisiología , Cicatrización de Heridas/fisiología , Heridas y Lesiones/patología
13.
PLoS One ; 7(5): e37195, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22606350

RESUMEN

BACKGROUND: Cross-talk between integrins and cadherins regulates cell function. We tested the hypothesis that vitronectin (VN), a multi-functional adhesion molecule present in the extracellular matrix and plasma, regulates vascular permeability via effects on VE-cadherin, a critical regulator of endothelial cell (EC) adhesion. METHODOLOGY/PRINCIPAL FINDINGS: Addition of multimeric VN (mult VN) significantly increased VE-cadherin internalization in human umbilical vein EC (HUVEC) monolayers. This effect was blocked by the anti-α(V)ß(3) antibody, pharmacological inhibition and knockdown of Src kinase. In contrast to mult VN, monomeric VN did not trigger VE-cadherin internalization. In a modified Miles assay, VN deficiency impaired vascular endothelial growth factor-induced permeability. Furthermore, ischemia-induced enhancement of vascular permeability, expressed as the ratio of FITC-dextran leakage from the circulation into the ischemic and non-ischemic hindlimb muscle, was significantly greater in the WT mice than in the Vn(-/-) mice. Similarly, ischemia-mediated macrophage infiltration was significantly reduced in the Vn(-/-) mice vs. the WT controls. We evaluated changes in the multimerization of VN in ischemic tissue in a mouse hindlimb ischemia model. VN plays a previously unrecognized role in regulating endothelial permeability via conformational- and integrin-dependent effects on VE-cadherin trafficking. CONCLUSION/SIGNIFICANCE: These results have important implications for the regulation of endothelial function and angiogenesis by VN under normal and pathological conditions.


Asunto(s)
Antígenos CD/metabolismo , Cadherinas/metabolismo , Permeabilidad Capilar/fisiología , Vitronectina/metabolismo , Animales , Permeabilidad Capilar/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Uniones Intercelulares/efectos de los fármacos , Uniones Intercelulares/metabolismo , Isquemia/metabolismo , Isquemia/patología , Macrófagos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Multimerización de Proteína , ARN Interferente Pequeño/genética , Proteínas Recombinantes/farmacología , Factor A de Crecimiento Endotelial Vascular/farmacología , Vitronectina/deficiencia , Vitronectina/genética , Vitronectina/farmacología , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/metabolismo
16.
J Physiol ; 560(Pt 3): 659-75, 2004 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-15319414

RESUMEN

Because the hyperpolarization-activated cation-selective current I(h) makes important contributions to neural excitability, we examined its long-term regulation by vitronectin, an extracellular matrix component commonly elevated at injury sites and detected immunochemically in activated microglia. Focusing on mouse hippocampal pyramidal neurones in organotypic slice cultures established at postnatal day 0 or 1 and examined after 3-4 days in vitro, we observed differences in the amplitude and activation rate of I(h) between neurones in naive and vitronectin-exposed slices (10 microg ml(-1) added to serum-free medium), and between neurones in slices derived from wild-type and vitronectin-deficient mice. The potassium inward rectifier I(K(ir)), activated at similar voltages to I(h), was not affected by vitronectin. In CA1, differences in I(h) amplitude primarily reflected changes in maximum conductance (G(max)): a 23.3% increase to 3.18 +/- 0.64 nS from 2.58 +/- 0.96 nS (P < 0.05) in vitronectin-exposed neurones, and a 17.9% decrease to 2.24 +/- 0.26 nS from 2.73 +/- 0.64 nS (P < 0.05) in neurones from vitronectin-deficient slices. The voltage of one-half maximum activation (V(1/2)) was not significantly affected by vitronectin exposure (-78.1 +/- 2.3 mV versus -80.0 +/- 4.9 mV in naive neurones; P > 0.05) or vitronectin deficiency (-83.8 +/- 3.1 mV versus -82.0 +/- 2.9 mV in wild-type neurones; P > 0.05). In CA3 neurones, changes in I(h) reflected differences in both G(max) and V(1/2): in vitronectin-exposed neurones there was a 35.4% increase in G(max) to 1.30 +/- 0.49 nS from 0.96 +/- 0.26 nS (P < 0.01), and a +3.0 mV shift in V(1/2) to -89.8 mV from -92.8 mV (P < 0.05). The time course of I(h) activation could be fitted by the sum of two exponential functions, fast and slow. In both CA1 and CA3 neurones the fast component amplitude was preferentially sensitive to vitronectin, with its relatively larger contribution to total current in vitronectin-exposed cells contributing to the acceleration of I(h) activation. Further, HCN1 immunoreactivity appeared elevated in vitronectin-exposed slices, while HCN2 levels appeared unaltered. We suggest that vitronectin-stimulated increases in I(h) may potentially affect excitability under pathological conditions.


Asunto(s)
Matriz Extracelular/fisiología , Canales Iónicos/fisiología , Proteínas del Tejido Nervioso/fisiología , Células Piramidales/efectos de los fármacos , Vitronectina/fisiología , Animales , Canales Catiónicos Regulados por Nucleótidos Cíclicos , Hipocampo/efectos de los fármacos , Hipocampo/fisiología , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización , Ratones , Ratones Endogámicos C57BL , Técnicas de Cultivo de Órganos , Canales de Potasio , Células Piramidales/fisiología , Vitronectina/deficiencia , Vitronectina/genética
17.
Blood ; 95(2): 577-80, 2000 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-10627465

RESUMEN

Occlusive thrombosis depends on the net balance between platelets, coagulation, and fibrinolytic factors. Epidemiologic information suggests that plasminogen activator inhibitor-1 (PAI-1), a central regulator of the fibrinolytic system, plays an important role in determining the overall risk for clinically significant vascular thrombosis. Vitronectin (VN), an abundant plasma and matrix glycoprotein, binds PAI-1 and stabilizes its active conformation. This study assessed the role of PAI-1 and VN expression in the formation of occlusive vascular thrombosis following arterial or venous injury. The common carotid arteries of 17 wild-type (WT) mice and 8 mice deficient in PAI-1 were injured photochemically while blood flow was continuously monitored. WT mice developed occlusive thrombi at 52.0 +/- 3.8 minutes (mean +/- SEM) following injury; mice deficient in PAI-1 developed occlusive thrombosis at 127 +/- 15 minutes (P <.0001). Mice deficient in VN (n = 12) developed vascular occlusion 77 +/- 11 minutes after injury, intermediate between the values observed for WT mice (P <.03) and mice deficient in PAI-1 (P <.01). PAI-1 and VN also affected the time to occlusion after injury to the jugular vein. Three WT mice developed occlusive venous thrombosis an average of 39.7 +/- 1 minutes following the onset of injury, whereas the jugular veins of 4 mice deficient in PAI-1 and 4 deficient in VN occluded 56.7 +/- 5 and 58.7 +/- 2 minutes, respectively, following injury (P <.04 and P <.01 compared to WT mice). These results suggest that endogenous fibrinolysis and its regulation by PAI-1 and VN have important roles in the development of occlusive vascular thrombosis after vascular injury. (Blood. 2000;95:577-580)


Asunto(s)
Estenosis Carotídea/fisiopatología , Inhibidor 1 de Activador Plasminogénico/fisiología , Trombosis/fisiopatología , Trombosis de la Vena/fisiopatología , Vitronectina/fisiología , Animales , Estenosis Carotídea/sangre , Estenosis Carotídea/genética , Venas Yugulares , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Inhibidor 1 de Activador Plasminogénico/deficiencia , Inhibidor 1 de Activador Plasminogénico/genética , Trombosis/sangre , Trombosis/genética , Trombosis de la Vena/sangre , Trombosis de la Vena/genética , Vitronectina/deficiencia , Vitronectina/genética
18.
Blood ; 93(6): 1825-30, 1999 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-10068653

RESUMEN

Vitronectin (VN) binds to plasminogen activator inhibitor-1 (PAI-1) and integrins and may play an important role in the vascular response to injury by regulating fibrinolysis and cell migration. However, the role of VN in the earliest response to vascular injury, thrombosis, is not well characterized. The purpose of this study was to test the hypothesis that variation in vitronectin expression alters the thrombotic response to arterial injury in mice. Ferric chloride (FeCl3) injury was used to induce platelet-rich thrombi in mouse carotid arteries. Wild-type (VN +/+, n = 14) and VN-deficient (VN -/-, n = 15) mice, matched for age and gender, were studied. Time to occlusion after FeCl3 injury was determined by application of a Doppler flowprobe to the carotid artery. Occlusion times of VN -/- mice were significantly shorter than those of VN +/+ mice (6.0 +/- 1.2 minutes v 17.8 +/- 2.3 minutes, respectively, P < .001). Histologic analysis of injured arterial segments showed that thrombi from VN +/+ and VN -/- mice consisted of dense platelet aggregates. In vitro studies of murine VN +/+ and VN -/- platelets showed no significant differences in ADP-induced aggregation, but a trend towards increased thrombin-induced aggregation in VN -/- platelets. Purified, denatured VN inhibited thrombin-induced platelet aggregation, whereas native VN did not. Thrombin times of plasma from VN -/- mice (20.5 +/- 2.1 seconds, n = 4) were significantly shorter than those of VN +/+ mice (34.2 +/- 6.7 seconds, n = 4, P < .01), and the addition of purified VN to VN -/- plasma prolonged the thrombin time into the normal range, suggesting that VN inhibits thrombin-fibrinogen interactions. PAI-1-deficient mice (n = 6) did not demonstrate significantly enhanced arterial thrombosis compared with wild-type mice (n = 6), excluding a potential indirect antithrombin function of VN mediated by interactions with PAI-1 as an explanation for the accelerated thrombosis observed in VN -/- mice. These results suggest that vitronectin plays a previously unappreciated antithrombotic role at sites of arterial injury and that this activity may be mediated, at least in part, by inhibiting platelet-platelet interactions and/or thrombin procoagulant activity.


Asunto(s)
Trombosis de las Arterias Carótidas/prevención & control , Vitronectina/fisiología , Adenosina Difosfato/farmacología , Animales , Plaquetas/fisiología , Trombosis de las Arterias Carótidas/inducido químicamente , Trombosis de las Arterias Carótidas/patología , Cloruros , Cruzamientos Genéticos , Compuestos Férricos , Humanos , Ratones , Ratones Endogámicos C57BL , Inhibidor 1 de Activador Plasminogénico/fisiología , Agregación Plaquetaria/efectos de los fármacos , Trombina/antagonistas & inhibidores , Trombina/farmacología , Tiempo de Trombina , Vitronectina/deficiencia , Vitronectina/genética
19.
Am J Respir Cell Mol Biol ; 31(6): 672-8, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15308506

RESUMEN

The pathogenesis of pulmonary fibrosis is thought to involve alveolar epithelial injury that, when successfully repaired, can limit subsequent scarring. The plasminogen system participates in this process with the balance between urokinase-type plasminogen activator (uPA) and plasminogen activator inhibitor-1 (PAI-1) being a critical determinant of the extent of collagen accumulation that follows lung injury. Because the plasminogen system is known to influence the rate of migration of epithelial cells, including keratinocytes and bronchial epithelial cells, we hypothesized that the balance of uPA and PAI-1 would affect the efficiency of alveolar epithelial cell (AEC) wound repair. Using an in vitro model of AEC wounding, we show that the efficiency of repair is adversely affected by a deficiency in uPA or by the exogenous administration of PAI-1. By using PAI-1 variants and AEC from mice transgenically deficient in vitronectin (Vn), we demonstrate that the PAI-1 effect requires its Vn-binding activity. Furthermore, we have found that cell motility is enhanced by the availability of Vn in the matrix and that the AEC-Vn interaction is mediated, in part, by the alpha(v)beta(1) integrin. The significant effect of uPA and PAI-1 on epithelial repair suggests a mechanism by which the plasminogen system may modulate pulmonary fibrosis.


Asunto(s)
Bleomicina/análogos & derivados , Inhibidor 1 de Activador Plasminogénico/farmacología , Alveolos Pulmonares/efectos de los fármacos , Alveolos Pulmonares/patología , Vitronectina/metabolismo , Cicatrización de Heridas/efectos de los fármacos , Animales , Bleomicina/farmacología , Línea Celular Tumoral , Movimiento Celular , Colágeno/metabolismo , Humanos , Ratones , Ratones Noqueados , Unión Proteica , Alveolos Pulmonares/metabolismo , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/farmacología , Vitronectina/deficiencia , Vitronectina/genética
20.
Proc Natl Acad Sci U S A ; 92(26): 12426-30, 1995 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-8618914

RESUMEN

Vitronectin (VN) is an abundant glycoprotein present in plasma and the extracellular matrix of most tissues. Though the precise function of VN in vivo is unknown, it has been implicated as a participant in diverse biological processes, including cell attachment and spreading, complement activation, and regulation of hemostasis. The major site of synthesis appears to be the liver, though VN is also found in the brain at an early stage of mouse organogenesis, suggesting that it may play an important role in mouse development. Genetic deficiency of VN has not been reported in humans or in other higher organisms. To examine the biologic function of VN within the context of the intact animal, we have established a murine model for VN deficiency through targeted disruption of the murine VN gene. Southern blot analysis of DNA obtained from homozygous null mice demonstrates deletion of all VN coding sequences, and immunological analysis confirms the complete absence of VN protein expression in plasma. However, heterozygous mice carrying one normal and one null VN allele and homozygous null mice completely deficient in VN demonstrate normal development, fertility, and survival. Sera obtained from VN-deficient mice are completely deficient in "serum spreading factor" and plasminogen activator inhibitor 1 binding activities. These observations demonstrate that VN is not essential for cell adhesion and migration during normal mouse development and suggest that its role in these processes may partially overlap with other adhesive matrix components.


Asunto(s)
Envejecimiento/fisiología , Fertilidad/fisiología , Hígado/metabolismo , Recombinación Genética , Células Madre/fisiología , Vitronectina/genética , Vitronectina/fisiología , Envejecimiento/genética , Animales , Secuencia de Bases , Blastocisto/fisiología , Línea Celular , Quimera , Cruzamientos Genéticos , Cartilla de ADN , Exones , Femenino , Fertilidad/genética , Eliminación de Gen , Expresión Génica , Biblioteca Genómica , Color del Cabello/genética , Homocigoto , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Ratones Noqueados , Datos de Secuencia Molecular , Inhibidor 1 de Activador Plasminogénico/sangre , Reacción en Cadena de la Polimerasa , Mapeo Restrictivo , Caracteres Sexuales , Vitronectina/deficiencia
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda