Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros











Intervalo de año de publicación
1.
Int J Radiat Oncol Biol Phys ; 94(5): 1163-72, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26883561

RESUMEN

PURPOSE: To determine whether the delivery of recombinant truncated plasminogen activator inhibitor-1 (PAI-1) protein (rPAI-1(23)) would protect from the development of radiation-induced lung injury. METHODS AND MATERIALS: C57Bl/6 mice received intraperitoneal injections of rPAI-1(23) (5.4 µg/kg/d) or vehicle for 18 weeks, beginning 2 days before irradiation (IR) (5 daily fractions of 6 Gy). Cohorts of mice were followed for survival (n=8 per treatment) and tissue collection (n=3 per treatment and time point). Fibrosis in lung was assessed with Masson-Trichrome staining and measurement of hydroxyproline content. Senescence was assessed with staining for ß-galactosidase activity in lung and primary pneumocytes. RESULTS: Hydroxyproline content in irradiated lung was significantly reduced in mice that received rPAI-1(23) compared with mice that received vehicle (IR+vehicle: 84.97 µg/lung; IR+rPAI-1(23): 56.2 µg/lung, P=.001). C57Bl/6 mice exposed to IR+vehicle had dense foci of subpleural fibrosis at 19 weeks, whereas the lungs of mice exposed to IR+rPAI-1(23) were largely devoid of fibrotic foci. Cellular senescence was significantly decreased by rPAI-1(23) treatment in primary pneumocyte cultures and in lung at multiple time points after IR. CONCLUSIONS: These studies identify that rPAI-1(23) is capable of preventing radiation-induced fibrosis in murine lungs. These antifibrotic effects are associated with increased fibrin metabolism, enhanced matrix metalloproteinase-3 expression, and reduced senescence in type 2 pneumocytes. Thus, rPAI-1(23) is a novel therapeutic option for radiation-induced fibrosis.


Asunto(s)
Células Epiteliales Alveolares/efectos de los fármacos , Senescencia Celular/efectos de los fármacos , Inhibidor 1 de Activador Plasminogénico/uso terapéutico , Fibrosis Pulmonar/prevención & control , Neumonitis por Radiación/complicaciones , Proteínas Recombinantes/uso terapéutico , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/efectos de la radiación , Animales , Proliferación Celular , Senescencia Celular/efectos de la radiación , Colágeno/metabolismo , Citocinas/metabolismo , Femenino , Fibrina/metabolismo , Hidroxiprolina/análisis , Hidroxiprolina/metabolismo , Pulmón/metabolismo , Pulmón/efectos de la radiación , Metaloproteinasa 3 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Células 3T3 NIH , Inhibidor 1 de Activador Plasminogénico/metabolismo , Fibrosis Pulmonar/etiología , Fibrosis Pulmonar/metabolismo , Neumonitis por Radiación/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa
2.
J Biol Chem ; 291(12): 6232-44, 2016 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-26801614

RESUMEN

Acyl-CoA:cholesterol acyltransferase 1 (Acat1) converts cellular cholesterol to cholesteryl esters and is considered a drug target for treating atherosclerosis. However, in mouse models for atherosclerosis, global Acat1 knockout (Acat1(-/-)) did not prevent lesion development. Acat1(-/-) increased apoptosis within lesions and led to several additional undesirable phenotypes, including hair loss, dry eye, leukocytosis, xanthomatosis, and a reduced life span. To determine the roles of Acat1 in monocytes/macrophages in atherosclerosis, we produced a myeloid-specific Acat1 knockout (Acat1(-M/-M)) mouse and showed that, in the Apoe knockout (Apoe(-/-)) mouse model for atherosclerosis, Acat1(-M/-M) decreased the plaque area and reduced lesion size without causing leukocytosis, dry eye, hair loss, or a reduced life span. Acat1(-M/-M) enhanced xanthomatosis in apoe(-/-) mice, a skin disease that is not associated with diet-induced atherosclerosis in humans. Analyses of atherosclerotic lesions showed that Acat1(-M/-M) reduced macrophage numbers and diminished the cholesterol and cholesteryl ester load without causing detectable apoptotic cell death. Leukocyte migration analysis in vivo showed that Acat1(-M/-M) caused much fewer leukocytes to appear at the activated endothelium. Studies in inflammatory (Ly6C(hi)-positive) monocytes and in cultured macrophages showed that inhibiting ACAT1 by gene knockout or by pharmacological inhibition caused a significant decrease in integrin ß 1 (CD29) expression in activated monocytes/macrophages. The sparse presence of lesion macrophages without Acat1 can therefore, in part, be attributed to decreased interaction between inflammatory monocytes/macrophages lacking Acat1 and the activated endothelium. We conclude that targeting ACAT1 in a myeloid cell lineage suppresses atherosclerosis progression while avoiding many of the undesirable side effects caused by global Acat1 inhibition.


Asunto(s)
Acetil-CoA C-Acetiltransferasa/genética , Aterosclerosis/inmunología , Macrófagos/inmunología , Acetil-CoA C-Acetiltransferasa/metabolismo , Animales , Apoptosis , Aterosclerosis/genética , Aterosclerosis/patología , Linfocitos B/metabolismo , Médula Ósea/patología , Movimiento Celular , Proliferación Celular , Dieta Alta en Grasa/efectos adversos , Progresión de la Enfermedad , Endotelio Vascular/inmunología , Endotelio Vascular/patología , Femenino , Células Madre Hematopoyéticas/fisiología , Leucocitosis/genética , Leucocitosis/inmunología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Células Mieloides/enzimología
3.
Trends Cardiovasc Med ; 23(4): 114-20, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23313168

RESUMEN

The vasa vasorum are unique networks of vessels that become angiogenic in response to changes in the vessel wall. Structural studies, using various imaging modalities, show that the vasa vasorum form a plexus of microvessels during the atherosclerotic disease process. The events that stimulate vasa vasorum neovascularization remain unclear. Anti-angiogenic molecules have been shown to inhibit/regress the neovascularization; they provide significant insight into vasa vasorum function, structure, and specific requirements for growth and stability. This review discusses evidence for and against potential stimulators of vasa vasorum neovascularization. Anti-angiogenic rPAI-123, a truncated isoform of plasminogen activator inhibitor-1 (PAI-1) stimulates a novel pathway for regulating plasmin activity. This mechanism contributes significantly to vasa vasorum regression/collapse and is discussed as a model of regression.


Asunto(s)
Neovascularización Patológica , Placa Aterosclerótica , Inhibidor 1 de Activador Plasminogénico/metabolismo , Vasa Vasorum , Inhibidores de la Angiogénesis/metabolismo , Animales , Humanos , Modelos Biológicos , Neovascularización Patológica/metabolismo , Neovascularización Patológica/fisiopatología , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/fisiopatología , Vasa Vasorum/metabolismo , Vasa Vasorum/fisiopatología
4.
Arterioscler Thromb Vasc Biol ; 32(11): 2644-51, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22982464

RESUMEN

OBJECTIVE: Vasa vasorum are angiogenic in advanced stages of human atherosclerosis and hypercholesterolemic mouse models. Fibroblast growth factor-2 (FGF-2) is the predominant angiogenic growth factor in the adventitia and plaque of hypercholesterolemic low-density lipoprotein receptor-deficient/apolipoprotein B(100/100) mice (DKO). FGF-2 seems to play a role in the formation of a distinct vasa vasorum network. This study examined the vasa vasorum structure and its relationship to FGF-2. METHODS AND RESULTS: DKO mice treated with saline, antiangiogenic recombinant plasminogen activator inhibitor-1(23) (rPAI-1(23)), or soluble FGF receptor 1 were perfused with fluorescein-labeled Lycopersicon esculentum lectin. Confocal images of FGF-2-probed descending aorta adventitia show that angiogenic vasa vasorum form a plexus-like network in saline-treated DKO similar to the FGF-2 pattern of distribution. Mice treated with rPAI-1(23) and soluble FGF receptor 1 lack a plexus; FGF-2 and vasa vasorum density and area are significantly reduced. A perlecan/FGF-2 complex is critical for plexus stability. Excess plasmin produced in rPAI-1(23)-treated DKO mice degrades perlecan and destabilizes the plexus. Plasmin activity and plaque size measured in DKO and DKO/plasminogen activator inhibitor-1(-)(/-) mice demonstrate that elevated plasmin activity contributes to reduced plaque size. CONCLUSIONS: An FGF-2/perlecan complex is required for vasa vasorum plexus stability. Elevated plasmin activity plays a significant inhibitory role in vasa vasorum plexus and plaque development.


Asunto(s)
Aorta/metabolismo , Enfermedades de la Aorta/metabolismo , Aterosclerosis/metabolismo , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Hipercolesterolemia/metabolismo , Neovascularización Patológica , Vasa Vasorum/metabolismo , Inhibidores de la Angiogénesis/farmacología , Animales , Aorta/efectos de los fármacos , Aorta/patología , Enfermedades de la Aorta/tratamiento farmacológico , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/patología , Apolipoproteína B-100 , Apolipoproteínas B/deficiencia , Apolipoproteínas B/genética , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/genética , Aterosclerosis/patología , Colesterol en la Dieta , Modelos Animales de Enfermedad , Fibrinolisina/metabolismo , Técnicas de Transferencia de Gen , Proteoglicanos de Heparán Sulfato/metabolismo , Hipercolesterolemia/complicaciones , Hipercolesterolemia/genética , Hipercolesterolemia/patología , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal , Placa Aterosclerótica , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Receptores de LDL/deficiencia , Receptores de LDL/genética , Rotura Espontánea , Vasa Vasorum/efectos de los fármacos , Vasa Vasorum/patología
5.
Circulation ; 125(2): 364-74, 2012 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-22144566

RESUMEN

BACKGROUND: Atherosclerotic lesions are believed to grow via the recruitment of bone marrow-derived monocytes. Among the known murine monocyte subsets, Ly-6C(high) monocytes are inflammatory, accumulate in lesions preferentially, and differentiate. Here, we hypothesized that the bone marrow outsources the production of Ly-6C(high) monocytes during atherosclerosis. METHODS AND RESULTS: Using murine models of atherosclerosis and fate-mapping approaches, we show that hematopoietic stem and progenitor cells progressively relocate from the bone marrow to the splenic red pulp, where they encounter granulocyte macrophage colony-stimulating factor and interleukin-3, clonally expand, and differentiate to Ly-6C(high) monocytes. Monocytes born in such extramedullary niches intravasate, circulate, and accumulate abundantly in atheromata. On lesional infiltration, Ly-6C(high) monocytes secrete inflammatory cytokines, reactive oxygen species, and proteases. Eventually, they ingest lipids and become foam cells. CONCLUSIONS: Our findings indicate that extramedullary sites supplement the hematopoietic function of the bone marrow by producing circulating inflammatory cells that infiltrate atherosclerotic lesions.


Asunto(s)
Antígenos Ly , Aterosclerosis/patología , Movimiento Celular/inmunología , Hematopoyesis Extramedular/inmunología , Monocitos/patología , Animales , Antígenos Ly/biosíntesis , Aterosclerosis/inmunología , Médula Ósea , Diferenciación Celular , Factor Estimulante de Colonias de Granulocitos y Macrófagos , Células Madre Hematopoyéticas/patología , Inflamación , Interleucina-3 , Ratones , Monocitos/inmunología
6.
Circ Res ; 108(12): 1419-28, 2011 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-21546607

RESUMEN

RATIONALE: The antiangiogenic activity of rPAI-1(23), a truncated plasminogen activator inhibitor-1 (PAI-1) protein, induces vasa vasorum collapse and significantly reduces plaque area and plaque cholesterol in hypercholesterolemic low-density lipoprotein receptor-deficient/apolipoprotein B48-deficient mice. OBJECTIVE: The objective of this study was to examine rPAI-1(23)-stimulated mechanisms that cause vasa vasorum collapse. METHODS AND RESULTS: The rPAI-1(23) protein opposed PAI-1 antiproteolytic function by stimulating a 1.6-fold increase in plasmin activity compared with the saline-treated counterpart. The increased proteolytic activity corresponded to increased activity of matrix metalloproteinase-3 and degradation of fibrin(ogen), nidogen, and perlecan in the adventitia of descending aortas. PAI-1 activity was reduced by 48% in response to rPAI-1(23); however, PAI-1 protein expression levels were similar in the rPAI-1(23)- and saline-treated hypercholesterolemic mice. Coimmunoprecipitation assays demonstrated a novel PAI-1-plasminogen complex in protein from the descending aorta of rPAI-1(23)- and saline-treated mice, but complexed PAI-1 was 1.6-fold greater in rPAI-1(23)-treated mice. Biochemical analyses demonstrated that rPAI-1(23) and PAI-1 binding interactions with plasminogen increased plasmin activity and reduced PAI-1 antiproteolytic activity. CONCLUSIONS: We conclude that rPAI-1(23) causes regression or collapse of adventitial vasa vasorum in hypercholesterolemic mice by stimulating an increase in plasmin activity. The rPAI-1(23)-enhanced plasmin activity was achieved through a novel mechanism by which rPAI-1(23) and PAI-1 bound plasminogen in a cooperative manner to increase plasmin activity and reduce PAI-1 activity.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Fibrinolisina/metabolismo , Hipercolesterolemia/metabolismo , Plasminógeno/metabolismo , Serpina E2/farmacología , Vasa Vasorum/metabolismo , Animales , Fibrinolisina/genética , Hipercolesterolemia/genética , Hipercolesterolemia/patología , Ratones , Ratones Noqueados , Plasminógeno/genética , Vasa Vasorum/patología
7.
J Clin Invest ; 120(4): 1217-28, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20237411

RESUMEN

Arterial morphogenesis is an important and poorly understood process. In particular, the signaling events controlling arterial formation have not been established. We evaluated whether alterations in the balance between ERK1/2 and PI3K signaling pathways could stimulate arterial formation in the setting of defective arterial morphogenesis in mice and zebrafish. Increased ERK1/2 activity in mouse ECs with reduced VEGF responsiveness was achieved in vitro and in vivo by downregulating PI3K activity, suppressing Akt1 but not Akt2 expression, or introducing a constitutively active ERK1/2 construct. Such restoration of ERK1/2 activation was sufficient to restore impaired arterial development and branching morphogenesis in synectin-deficient mice and synectin-knockdown zebrafish. The same approach effectively stimulated arterial growth in adult mice, restoring arteriogenesis in mice lacking synectin and in atherosclerotic mice lacking both LDL-R and ApoB48. We therefore conclude that PI3K-ERK1/2 crosstalk plays a key role in the regulation of arterial growth and that the augmentation of ERK signaling via suppression of the PI3K signaling pathway can effectively stimulate arteriogenesis.


Asunto(s)
Arterias/crecimiento & desarrollo , Proteína Quinasa 1 Activada por Mitógenos/fisiología , Proteína Quinasa 3 Activada por Mitógenos/fisiología , Morfogénesis , Proteínas Proto-Oncogénicas c-akt/fisiología , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas Portadoras/fisiología , Masculino , Ratones , Neovascularización Fisiológica , Neuropéptidos/fisiología , Fosfatidilinositol 3-Quinasas/fisiología , Transducción de Señal/fisiología , Factor A de Crecimiento Endotelial Vascular/farmacología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/fisiología , Pez Cebra , Proteínas de Pez Cebra
8.
Circ Res ; 104(3): 337-45, 2009 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-19122176

RESUMEN

Plaque vascularity has been implicated in its growth and stability. However, there is a paucity of information regarding the origin of plaque vasculature and the role of vasa vasorum in plaque growth. To inhibit growth of vasa vasorum in atherogenic mice and assess its effect on plaque growth, we used a truncated plasminogen activator inhibitor (PAI)-1 protein, rPAI-1(23), that has significant antiangiogenic activity. Female LDLR(-/-)ApoB-48-deficient mice fed Paigen's diet without cholate for 20 weeks received rPAI-1(23) treatment (n=21) for the last 6 weeks. Plaque size and vasa vasorum density were compared to 2 controls: mice fed Paigen's diet and treated with saline for the last 6 weeks (n=16) and mice fed Paigen's diet until the onset of treatment (n=14). The rPAI-1(23) treatment significantly reduced plaque area and plaque cholesterol in the descending aorta and plaque area in the innominate artery. Measurements of reconstructed confocal microscopy images of vasa vasorum demonstrate that rPAI-1(23) treatment decreased vasa vasorum area and length, which was supported by microCT images. Confocal images provide evidence for vascularized plaque in the saline-treated group but not in rPAI-1(23)-treated mice. The increased vessel density in saline-treated mice is attributable, in part, to upregulated fibroblast growth factor-2 expression, which is inhibited by rPAI-1(23). In conclusion, rPAI-1(23) inhibits growth of vasa vasorum, as well as vessels within the adjacent plaque and vessel wall, through inhibition of fibroblast growth factor-2, leading to reduced plaque growth in atherogenic female LDLR(-/-)ApoB-48-deficient mice.


Asunto(s)
Inhibidores de la Angiogénesis/fisiología , Aterosclerosis/prevención & control , Inhibidor 1 de Activador Plasminogénico/fisiología , Vasa Vasorum/efectos de los fármacos , Inhibidores de la Angiogénesis/farmacología , Animales , Apolipoproteína B-48/genética , Arterias/patología , Aterosclerosis/patología , Femenino , Factor 2 de Crecimiento de Fibroblastos/genética , Ratones , Ratones Endogámicos , Ratones Mutantes , Microscopía Confocal , Fragmentos de Péptidos/farmacología , Fragmentos de Péptidos/fisiología , Inhibidor 1 de Activador Plasminogénico/farmacología , Receptores de LDL/genética , Proteínas Recombinantes/farmacología , Vasa Vasorum/crecimiento & desarrollo , Vasa Vasorum/patología , Factor A de Crecimiento Endotelial Vascular/genética
9.
Rheum Dis Clin North Am ; 34(1): 73-9; vi, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18329533

RESUMEN

Vascular abnormalities are one of the primary pathologic components of scleroderma. An early vascular indicator is aberrant nail fold capillaries that appear to undergo a switch from a pro- to anti-angiogenic process. Later in the disease process, ineffective and aberrant wound healing becomes apparent with frequent and widespread fibrosis. Pulmonary hypertension, largely due to the loss of pulmonary arterial vasculature, is frequently observed in late stages of the disease. The common theme of all these processes is abnormal regeneration of the vasculature and ongoing vascular losses due to defective maintenance of the vasculature. Although most aspects of vascular injury in scleroderma are poorly understood, certain biologic themes are beginning to emerge that are important in understanding scleroderma-related vascular disease.


Asunto(s)
Neovascularización Fisiológica/fisiología , Esclerodermia Sistémica/fisiopatología , Enfermedades Vasculares/fisiopatología , Inhibidores de la Angiogénesis/fisiología , Capilares/patología , Humanos , Esclerodermia Sistémica/complicaciones , Enfermedades Vasculares/etiología , Cicatrización de Heridas/fisiología
10.
Arthritis Rheum ; 56(10): 3448-58, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17907150

RESUMEN

OBJECTIVE: Systemic sclerosis (SSc; scleroderma) is a systemic connective tissue disease with an extensive vascular component that includes aberrant microvasculature and impaired wound healing. The aim of this study was to investigate the presence of antiangiogenic factors in patients with SSc. METHODS: Plasma samples were obtained from 30 patients with SSc and from 10 control patients without SSc. The samples were analyzed for the ability of plasma to affect endothelial cell migration and vascular structure formation and for the presence of antiangiogenic activity. RESULTS: Exposure of normal human microvascular dermal endothelial cells to plasma from patients with SSc resulted in decreased cell migration (mean +/- SEM 52 +/- 5%) and tube formation (34 +/- 6%) compared with that in plasma from control patients (P < 0.001 for both). SSc plasma contained 2.9-fold more plasminogen kringle 1-3 fragments (angiostatin) than that in control plasma. The addition of angiostatin to control plasma resulted in inhibition of endothelial cell migration and proliferation similar to that observed in SSc plasma. In vitro studies demonstrated that granzyme B and other proteases contained in T cell granule content cleave plasminogen and plasmin into angiostatin fragments. CONCLUSION: Plasminogen conformation in patients with SSc enables granzyme B and granule content protease to limit the proangiogenic effects of plasmin and increase the levels of antiangiogenic angiostatin. This increase in angiostatin production may account for some of the vascular defects observed in patients with SSc.


Asunto(s)
Inhibidores de la Angiogénesis/metabolismo , Células Endoteliales/metabolismo , Esclerodermia Sistémica/fisiopatología , Adulto , Inhibidores de la Angiogénesis/sangre , Movimiento Celular/fisiología , Femenino , Granzimas/metabolismo , Humanos , Técnicas In Vitro , Masculino , Persona de Mediana Edad , Esclerodermia Sistémica/sangre , Cicatrización de Heridas/fisiología
11.
J Biol Chem ; 281(44): 33336-44, 2006 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-16950776

RESUMEN

Many angiogenesis inhibitors are breakdown products of endogenous extracellular matrix proteins. Plasmin and matrix metalloproteinase-3 generate breakdown products of matrix-bound plasminogen activator inhibitor-1 (PAI-1). We produced a truncated form of PAI-1, rPAI-1(23), that possesses significant anti-angiogenic activity and stimulates high levels of apoptosis in quiescent arterial endothelial cells. Quiescent endothelial cells are less susceptible to apoptosis than angiogenic endothelial cells. The present study was designed to determine the mechanism of the rPAI-1(23) effects in bovine aortic endothelial cells. Apoptosis was measured in annexin V and caspase 3 assays. Expression of death and survival signaling molecules were examined by Western blot and kinase activity. Fibroblast growth factor 2 (FGF2) functions were analyzed in angiogenesis assays. The early response to rPAI-1(23) was an increase in annexin V-positive cells and phosphorylated (p) JNK isoform expression followed by an increase in p-Akt and p-c-Jun expression. Caspase 3 was activated at 4 h, whereas p-Akt was reduced to control levels. By 6 h of rPAI-1(23) treatment cell number was reduced by 35%, and p-c-Jun and p-JNK were degraded by proteasomes. Confocal microscopic images showed increased amounts of FGF2 in the extracellular matrix. However, rPAI-1(23) blocked FGF2 signaling through FGF receptor 1 and syndecan-4, inhibiting cell migration, tubulogenesis, and proliferation. Exogenous FGF2 stimulation could not reverse these effects. We conclude that rPAI-1(23) stimulation of apoptosis in BAEC triggers a cascade of death versus survival events that includes release of FGF2. The rPAI-1(23) anti-angiogenic activity inhibits FGF2 pro-angiogenic functions by blocking FGF2 signaling through FGF receptor 1 and syndecan-4 and downstream effectors p-Akt, p-JNK, and p-c-Jun.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Fragmentos de Péptidos/farmacología , Inhibidor 1 de Activador Plasminogénico/farmacología , Inhibidores de la Angiogénesis/genética , Inhibidores de la Angiogénesis/metabolismo , Animales , Apoptosis/efectos de los fármacos , Bovinos , Células Cultivadas , Pollos , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Matriz Extracelular/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Fosforilación , Inhibidor 1 de Activador Plasminogénico/genética , Inhibidor 1 de Activador Plasminogénico/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Recombinantes/farmacología , Transducción de Señal
12.
J Biol Chem ; 277(50): 49077-89, 2002 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-12381729

RESUMEN

We have made deletions of the porcine plasminogen activator inhibitor-1 (PAI-1) gene to obtain recombinant truncated PAI-1 proteins to examine functions of the PAI-1 isoforms. We previously reported that one recombinant truncated protein, rPAI-1(23), induces the formation of angiostatin by cleaving plasmin. The rPAI-1(23) protein is also able to bind urokinase plasminogen activator and plasminogen and then reduce the amount of plasmin that is formed. We have now prepared three different truncated rPAI-1 proteins and demonstrate that PAI-1 conformations control the release of heparin-binding vascular endothelial growth factor (VEGF) isoforms. The rPAI-1(23) isoform can regulate the functional activity of heparan sulfate-binding VEGF-A isoforms by blocking the activation of VEGF from heparan sulfate. The rPAI-1(23) conformation induced extensive apoptosis in cultured endothelial cells and thus reduced the number of proliferating cells. The rPAI-1(23) isoform inhibited migration of VEGF-stimulated sprouting from chick aortic rings by 65%, thus displaying a role in anti-angiogenic mechanisms. This insight into anti-angiogenic functions related to PAI-1 conformational changes could provide potential intervention points in angiogenesis associated with atherosclerotic plaques and cancer.


Asunto(s)
Factores de Crecimiento Endotelial/antagonistas & inhibidores , Heparina/metabolismo , Linfocinas/antagonistas & inhibidores , Inhibidor 1 de Activador Plasminogénico/fisiología , Animales , Apoptosis , Secuencia de Bases , Bovinos , Células Cultivadas , Embrión de Pollo , Medios de Cultivo , Cartilla de ADN , Factores de Crecimiento Endotelial/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Linfocinas/metabolismo , Plasminógeno/metabolismo , Inhibidor 1 de Activador Plasminogénico/química , Unión Proteica , Porcinos , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
13.
J Vasc Surg ; 36(1): 164-71, 2002 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12096275

RESUMEN

INTRODUCTION: Plasminogen activator inhibitor-1 (PAI-1), a known inhibitor of plasminogen activators, may regulate smooth muscle cell migration (SMC) through alteration in matrix metalloproteinase (MMP) activity. METHODS: To study the effect of endothelial cell (EC) PAI-1 overexpression on SMC migration, RT-PCR was used to clone the full length PAI-1 gene, which was ligated into the pCMV/myc/ER expression vector. With electroporation, bovine aortic ECs were transfected with either the PAI-1 construct or the empty vector as control. EC PAI-1 overexpression was shown with a specific PAI-1 activity assay and enzyme-linked immunosorbent assay. The effect of EC PAI-1 overexpression on SMC migration was measured with a modified Boyden-chamber assay. SMC MMP expression was measured with zymography. RESULTS: Selected clones (EC9, EC21) had a three-fold to five-fold increase in PAI-1 activity compared with untransfected EC and empty vector EC (ECC). Similarly, enzyme-linked immunosorbent assay results showed a 3.5-fold to 5.5-fold increase in PAI-1 levels in EC9 and EC21 versus ECC. Untransfected EC and ECC had similar effects on SMC migratory patterns. Migration of SMC exposed to PAI-1 overexpressing EC was inhibited by 35% to 57% compared with ECC. This inhibitory effect was reversed with addition of exogenous urokinase-type plasminogen activator (uPA). Zymography showed downregulation of MMP-2 and MMP-9 in SMCs exposed to PAI-1 overexpressing EC. CONCLUSION: PAI-1 overexpression with transfected EC inhibits SMC migration. This effect may be mediated through decreased SMC MMP activity.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Inhibidor 1 de Activador Plasminogénico/biosíntesis , Inhibidor 1 de Activador Plasminogénico/farmacología , Inhibidores de Serina Proteinasa/biosíntesis , Inhibidores de Serina Proteinasa/farmacología , Animales , Aorta Torácica/citología , Aorta Torácica/metabolismo , Bovinos , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Metaloproteinasa 2 de la Matriz/biosíntesis , Metaloproteinasa 2 de la Matriz/efectos de los fármacos , Metaloproteinasa 9 de la Matriz/biosíntesis , Metaloproteinasa 9 de la Matriz/efectos de los fármacos , Modelos Animales , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA