Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 175
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Allergy ; 69(2): 237-45, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24127643

RESUMEN

BACKGROUND: Edema represents a key feature of nasal polyp (NP) disease. Members of the vascular endothelial growth factor (VEGF) family may be involved, but the precise role of VEGF-A, VEGF-B, placental growth factor (PlGF), and their receptors VEGFR1 and VEGFR2 in NP edema formation remains elusive. OBJECTIVE: Exploring the expression of VEGF family members and their receptors and their correlation with clinical, radiological, and edema markers in NP. METHODS: The expression of VEGF-A, VEGF-B, PlGF, VEGFR1, and VEGFR2 was measured in NP (n = 23) and control tissue (n = 22) at mRNA and protein level. Edema was evaluated by measuring albumin levels and wet/dry ratios. Computed tomography (CT) scans were scored using the Lund-Mackay scoring system. IL-5 mRNA expression was determined by real-time RT-PCR. Cell suspensions from NP (n = 10) and control tissue (n = 12) were stimulated in vitro with IL-1ß or TNFα. RESULTS: mRNA expression of VEGFR1 and VEGF-B was significantly higher in NP compared with control tissue. Expression levels of VEGF-B and VEGFR1 significantly correlated with NP albumin content (VEGF-B: P = 0.0208; VEGFR1: P = 0.0293), CT scan scores (VEGF-B: P = 0.0075; VEGFR1: P = 0.0068), and IL-5 mRNA (VEGF-B: P = 0.0027; VEGFR1: P = 0.0001). In vitro stimulation of control and NP tissue cell suspensions with IL-1ß or TNFα significantly reduced the expression of VEGFR2 in control tissue, without altering VEGFR1 and VEGF-B expression. hVEGF-B induced nitric oxide production in NP macrophages (P < 0.05). CONCLUSION: Expression levels of VEGFR1 and VEGF-B correlate with edema and clinical markers of NP disease and therefore represent potential therapeutic targets.


Asunto(s)
Pólipos Nasales/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/biosíntesis , Femenino , Humanos , Inmunohistoquímica , Masculino , Proteínas de la Membrana/análisis , Proteínas de la Membrana/biosíntesis , Pólipos Nasales/patología , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor A de Crecimiento Endotelial Vascular/análisis , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Factor B de Crecimiento Endotelial Vascular/análisis , Factor B de Crecimiento Endotelial Vascular/biosíntesis , Receptor 1 de Factores de Crecimiento Endotelial Vascular/análisis , Receptor 2 de Factores de Crecimiento Endotelial Vascular/análisis , Receptor 2 de Factores de Crecimiento Endotelial Vascular/biosíntesis
4.
Nat Genet ; 17(4): 439-44, 1997 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-9398846

RESUMEN

The molecular mechanisms predisposing to atherosclerotic aneurysm formation remain undefined. Nevertheless, rupture of aortic aneurysms is a major cause of death in Western societies, with few available treatments and poor long-term prognosis. Indirect evidence suggests that matrix metalloproteinases (MMPs) and plasminogen activators (PAs) are involved in its pathogenesis. MMPs are secreted as inactive zymogens (pro-MMPs), requiring activation in the extracellular compartment. Plasmin, generated from the zymogen plasminogen by tissue-type plasminogen activator (t-PA) or urokinase-type plasminogen activator (u-PA; refs 14,15), has been proposed as a possible activator in vitro, but evidence for such a role in vivo is lacking. Analysis of atherosclerotic aorta in mice with a deficiency of apoliprotein E (Apoe-/-; ref. 18), singly or combined with a deficiency of t-PA (Apoe-/-:Plat-/-) or of u-PA (Apoe-/-:Plau-/-; ref. 19), indicated that deficiency of u-PA protected against media destruction and aneurysm formation, probably by means of reduced plasmin-dependent activation of pro-MMPs. This genetic evidence suggests that plasmin is a pathophysiologically significant activator of pro-MMPs in vivo and may have implications for the design of therapeutic strategies to prevent aortic-wall destruction by controlling Plau gene function.


Asunto(s)
Aneurisma de la Aorta Abdominal/enzimología , Aneurisma de la Aorta Torácica/enzimología , Fibrinolisina/metabolismo , Metaloendopeptidasas/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Animales , Aneurisma de la Aorta Abdominal/etiología , Aneurisma de la Aorta Abdominal/patología , Aneurisma de la Aorta Torácica/etiología , Aneurisma de la Aorta Torácica/patología , Arteriosclerosis/enzimología , Arteriosclerosis/patología , Colágeno/metabolismo , Dieta Aterogénica , Elastina/metabolismo , Activación Enzimática , Femenino , Macrófagos/enzimología , Masculino , Ratones , Ratones Noqueados , Túnica Media/enzimología , Túnica Media/patología
5.
Nat Genet ; 17(1): 49-57, 1997 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-9288097

RESUMEN

The cerebro-hepato-renal syndrome of Zellweger is a fatal inherited disease caused by deficient import of peroxisomal matrix proteins. The pathogenic mechanisms leading to extreme hypotonia, severe mental retardation and early death are unknown. We generated a Zellweger animal model through inactivation of the murine Pxr1 gene (formally known as Pex5) that encodes the import receptor for most peroxisomal matrix proteins. Pxr1-/- mice lacked morphologically identifiable peroxisomes and exhibited the typical biochemical abnormalities of Zellweger patients. They displayed intrauterine growth retardation, were severely hypotonic at birth and died within 72 hours. Analysis of the neocortex revealed impaired neuronal migration and maturation and extensive apoptotic death of neurons.


Asunto(s)
Receptores Citoplasmáticos y Nucleares/deficiencia , Receptores Citoplasmáticos y Nucleares/genética , Síndrome de Zellweger/genética , Animales , Animales Recién Nacidos , Apoptosis , Secuencia de Bases , Encéfalo/metabolismo , Encéfalo/patología , Corteza Cerebral/patología , ADN/biosíntesis , Cartilla de ADN , Muerte , Modelos Animales de Enfermedad , Femenino , Retardo del Crecimiento Fetal , Fibroblastos/metabolismo , Humanos , Hígado/metabolismo , Hígado/patología , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Neuronas/patología , Neuronas/fisiología , Receptor de la Señal 1 de Direccionamiento al Peroxisoma , Reacción en Cadena de la Polimerasa , Embarazo , Receptores Citoplasmáticos y Nucleares/metabolismo , Recombinación Genética , Síndrome de Zellweger/patología , Síndrome de Zellweger/fisiopatología
6.
Nat Genet ; 28(2): 131-8, 2001 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-11381259

RESUMEN

Hypoxia stimulates angiogenesis through the binding of hypoxia-inducible factors to the hypoxia-response element in the vascular endothelial growth factor (Vegf) promotor. Here, we report that deletion of the hypoxia-response element in the Vegf promotor reduced hypoxic Vegf expression in the spinal cord and caused adult-onset progressive motor neuron degeneration, reminiscent of amyotrophic lateral sclerosis. The neurodegeneration seemed to be due to reduced neural vascular perfusion. In addition, Vegf165 promoted survival of motor neurons during hypoxia through binding to Vegf receptor 2 and neuropilin 1. Acute ischemia is known to cause nonselective neuronal death. Our results indicate that chronic vascular insufficiency and, possibly, insufficient Vegf-dependent neuroprotection lead to the select degeneration of motor neurons.


Asunto(s)
Hipoxia de la Célula/genética , Factores de Crecimiento Endotelial/genética , Linfocinas/genética , Neuronas Motoras/patología , Degeneración Nerviosa/genética , Elementos de Respuesta/genética , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/patología , Animales , Axones/fisiología , Sitios de Unión , Electrofisiología , Factores de Crecimiento Endotelial/metabolismo , Humanos , Linfocinas/metabolismo , Ratones , Ratones Noqueados , Neuronas Motoras/fisiología , Contracción Muscular , Fibras Musculares Esqueléticas/patología , Atrofia Muscular/genética , Atrofia Muscular/patología , Degeneración Nerviosa/patología , Degeneración Nerviosa/fisiopatología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuropilina-1 , Nervios Periféricos/patología , Regiones Promotoras Genéticas , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores de Factores de Crecimiento/genética , Receptores de Factores de Crecimiento/metabolismo , Receptores de Factores de Crecimiento Endotelial Vascular , Eliminación de Secuencia , Médula Espinal/fisiología , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
7.
Nat Med ; 6(4): 389-95, 2000 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10742145

RESUMEN

Endothelial and smooth muscle cells interact with each other to form new blood vessels. In this review, the cellular and molecular mechanisms underlying the formation of endothelium-lined channels (angiogenesis) and their maturation via recruitment of smooth muscle cells (arteriogenesis) during physiological and pathological conditions are summarized, alongside with possible therapeutic applications.


Asunto(s)
Arteriolas/fisiopatología , Neovascularización Patológica , Animales , Endotelio Vascular/citología , Endotelio Vascular/crecimiento & desarrollo , Humanos , Músculo Liso Vascular/fisiopatología , Neovascularización Patológica/metabolismo , Neovascularización Patológica/fisiopatología , Neovascularización Fisiológica/fisiología
8.
Nat Med ; 7(1): 59-64, 2001 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11135617

RESUMEN

Tissue-plasminogen activator (t-PA) is now available for the treatment of thrombo-embolic stroke but adverse effects have been reported in some patients, particularly hemorrhaging. In contrast, the results of animal studies have indicated that t-PA could increase neuronal damage after focal cerebral ischemia. Here we report for the first time that t-PA potentiates signaling mediated by glutamatergic receptors by modifying the properties of the N-methyl-D-aspartate (NMDA) receptor. When depolarized, cortical neurons release bio-active t-PA that interacts with and cleaves the NR1 subunit of the NMDA receptor. Moreover, the treatment with recombinant t-PA leads to a 37% increase in NMDA-stimulated fura-2 fluorescence, which may reflect an increased NMDA-receptor function. These results were confirmed in vivo by the intrastriatal injection of recombinant-PA, which potentiated the excitotoxic lesions induced by NMDA. These data provide insight into the regulation of NMDA-receptor-mediated signaling and could initiate therapeutic strategies to improve the efficacy of t-PA treatment in man.


Asunto(s)
Receptores de N-Metil-D-Aspartato/metabolismo , Transducción de Señal , Activador de Tejido Plasminógeno/metabolismo , Animales , Calcio/metabolismo , Muerte Celular , Hidrólisis , Transporte Iónico , Potenciales de la Membrana , Neuronas/metabolismo , Neuronas/fisiología
9.
Nat Med ; 7(7): 864-8, 2001 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-11433354

RESUMEN

Intravital microscopy coupled with chronic animal window models has provided stunning insight into tumor pathophysiology, including gene expression, angiogenesis, cell adhesion and migration, vascular, interstitial and lymphatic transport, metabolic microenvironment and drug delivery. However, the findings to date have been limited to the tumor surface (< 150 microm). Here, we show that the multiphoton laser-scanning microscope can provide high three-dimensional resolution of gene expression and function in deeper regions of tumors. These insights could be critical to the development of novel therapeutics that target not only the tumor surface, but also internal regions.


Asunto(s)
Expresión Génica , Microscopía/métodos , Neoplasias Experimentales/irrigación sanguínea , Neovascularización Patológica , Adhesión Celular , Hemodinámica , Rayos Láser , Leucocitos/citología , Fotones
10.
Nat Med ; 7(2): 215-21, 2001 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11175853

RESUMEN

The growth arrest-specific gene 6 product (Gas6) is a secreted protein related to the anticoagulant protein S but its role in hemostasis is unknown. Here we show that inactivation of the Gas6 gene prevented venous and arterial thrombosis in mice, and protected against fatal collagen/epinephrine-induced thrombo embolism. Gas6-/- mice did not, however, suffer spontaneous bleeding and had normal bleeding after tail clipping. In addition, we found that Gas6 antibodies inhibited platelet aggregation in vitro and protected mice against fatal thrombo embolism without causing bleeding in vivo. Gas6 amplified platelet aggregation and secretion in response to known agonists. Platelet dysfunction in Gas6-/- mice resembled that of patients with platelet signaling transduction defects. Thus, Gas6 is a platelet-response amplifier that plays a significant role in thrombosis. These findings warrant further evaluation of the possible therapeutic use of Gas6 inhibition for prevention of thrombosis.


Asunto(s)
Plaquetas/fisiología , Péptidos y Proteínas de Señalización Intercelular , Proteínas/fisiología , Trombosis/prevención & control , Animales , Plaquetas/metabolismo , Línea Celular , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Hemostasis , Humanos , Masculino , Ratones , Ratones Noqueados , Fenotipo , Agregación Plaquetaria , Proteínas/genética , Proteínas/inmunología , Proteínas/farmacología , Receptores de Superficie Celular/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología , Trombosis/etiología
11.
Nat Med ; 6(1): 49-55, 2000 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-10613823

RESUMEN

Although tissue injury and inflammation are considered essential for the induction of angiogenesis, the molecular controls of this cascade are mostly unknown. Here we show that a macrophage-derived peptide, PR39, inhibited the ubiquitin-proteasome-dependent degradation of hypoxia-inducible factor-1alpha protein, resulting in accelerated formation of vascular structures in vitro and increased myocardial vasculature in mice. For the latter, coronary flow studies demonstrated that PR39-induced angiogenesis resulted in the production of functional blood vessels. These findings show that PR39 and related compounds can be used as potent inductors of angiogenesis, and that selective inhibition of hypoxia-inducible factor-1alpha degradation may underlie the mechanism of inflammation-induced angiogenesis.


Asunto(s)
Péptidos Catiónicos Antimicrobianos , Proteínas de Unión al ADN/metabolismo , Endotelio Vascular/fisiología , Isquemia Miocárdica/fisiopatología , Neovascularización Fisiológica/fisiología , Proteínas Nucleares/metabolismo , Péptidos/fisiología , Animales , Aorta , Capilares/efectos de los fármacos , Capilares/fisiología , Bovinos , Hipoxia de la Célula , Células Cultivadas , Vasos Coronarios/efectos de los fármacos , Cisteína Endopeptidasas/metabolismo , Endotelio Vascular/citología , Corazón/fisiología , Humanos , Factor 1 Inducible por Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia , Técnicas In Vitro , Macrófagos/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Complejos Multienzimáticos/metabolismo , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Isquemia Miocárdica/patología , Péptidos/genética , Péptidos/farmacología , Complejo de la Endopetidasa Proteasomal , Proteínas Recombinantes/metabolismo , Porcinos , Factores de Transcripción/metabolismo , Ubiquitinas/metabolismo , Venas Umbilicales , Factor de von Willebrand/genética
12.
Nat Med ; 4(8): 923-8, 1998 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-9701244

RESUMEN

Acquisition of invasive/metastatic potential through protease expression is an essential event in tumor progression. High levels of components of the plasminogen activation system, including urokinase, but paradoxically also its inhibitor, plasminogen activator inhibitor 1 (PAI1), have been correlated with a poor prognosis for some cancers. We report here that deficient PAI1 expression in host mice prevented local invasion and tumor vascularization of transplanted malignant keratinocytes. When this PAI1 deficiency was circumvented by intravenous injection of a replication-defective adenoviral vector expressing human PAI1, invasion and associated angiogenesis were restored. This experimental evidence demonstrates that host-produced PAI is essential for cancer cell invasion and angiogenesis.


Asunto(s)
Invasividad Neoplásica/prevención & control , Neovascularización Patológica/prevención & control , Inhibidor 1 de Activador Plasminogénico/biosíntesis , Inhibidor 1 de Activador Plasminogénico/deficiencia , Neoplasias Cutáneas/patología , Adenoviridae , Animales , Transformación Celular Neoplásica , Células Cultivadas , Progresión de la Enfermedad , Femenino , Vectores Genéticos , Genotipo , Humanos , Queratinocitos/patología , Masculino , Mesodermo/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Metástasis de la Neoplasia , Inhibidor 1 de Activador Plasminogénico/genética , Neoplasias Cutáneas/irrigación sanguínea , Transfección
13.
Nat Med ; 7(9): 1021-7, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11533705

RESUMEN

Deletion of amino-acid residues 1505-1507 (KPQ) in the cardiac SCN5A Na(+) channel causes autosomal dominant prolongation of the electrocardiographic QT interval (long-QT syndrome type 3 or LQT3). Excessive prolongation of the action potential at low heart rates predisposes individuals with LQT3 to fatal arrhythmias, typically at rest or during sleep. Here we report that mice heterozygous for a knock-in KPQ-deletion (SCN5A(Delta/+)) show the essential LQT3 features and spontaneously develop life-threatening polymorphous ventricular arrhythmias. Unexpectedly, sudden accelerations in heart rate or premature beats caused lengthening of the action potential with early afterdepolarization and triggered arrhythmias in Scn5a(Delta/+) mice. Adrenergic agonists normalized the response to rate acceleration in vitro and suppressed arrhythmias upon premature stimulation in vivo. These results show the possible risk of sudden heart-rate accelerations. The Scn5a(Delta/+) mouse with its predisposition for pacing-induced arrhythmia might be useful for the development of new treatments for the LQT3 syndrome.


Asunto(s)
Arritmias Cardíacas/genética , Arritmias Cardíacas/fisiopatología , Canales de Sodio/genética , Agonistas Adrenérgicos beta/farmacología , Animales , Arritmias Cardíacas/tratamiento farmacológico , Estimulación Cardíaca Artificial , Electrocardiografía , Humanos , Isoproterenol/farmacología , Síndrome de QT Prolongado/genética , Potenciales de la Membrana , Ratones , Ratones Mutantes , Miocardio/citología , Miocardio/metabolismo , Canal de Sodio Activado por Voltaje NAV1.5 , Eliminación de Secuencia , Sodio/metabolismo
14.
Nat Med ; 5(10): 1135-42, 1999 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-10502816

RESUMEN

Cardiac rupture is a fatal complication of acute myocardial infarction lacking treatment. Here, acute myocardial infarction resulted in rupture in wild-type mice and in mice lacking tissue-type plasminogen activator, urokinase receptor, matrix metalloproteinase stromelysin-1 or metalloelastase. Instead, deficiency of urokinase-type plasminogen activator (u-PA-/-) completely protected against rupture, whereas lack of gelatinase-B partially protected against rupture. However, u-PA-/- mice showed impaired scar formation and infarct revascularization, even after treatment with vascular endothelial growth factor, and died of cardiac failure due to depressed contractility, arrhythmias and ischemia. Temporary administration of PA inhibitor-1 or the matrix metalloproteinase-inhibitor TIMP-1 completely protected wild-type mice against rupture but did not abort infarct healing, thus constituting a new approach to prevent cardiac rupture after acute myocardial infarction.


Asunto(s)
Gasto Cardíaco Bajo/etiología , Rotura Cardíaca/etiología , Metaloendopeptidasas/antagonistas & inhibidores , Infarto del Miocardio/complicaciones , Infarto del Miocardio/tratamiento farmacológico , Inactivadores Plasminogénicos/uso terapéutico , Inhibidores de Proteasas/uso terapéutico , Animales , Arritmias Cardíacas , Trasplante de Médula Ósea , Movimiento Celular , Colagenasas/metabolismo , Técnicas de Transferencia de Gen , Leucocitos/citología , Leucocitos/metabolismo , Metaloproteinasa 3 de la Matriz/genética , Metaloproteinasa 9 de la Matriz , Ratones , Ratones Mutantes , Neovascularización Fisiológica/efectos de los fármacos , Inhibidor 1 de Activador Plasminogénico/genética , Inhibidor 1 de Activador Plasminogénico/metabolismo , Activadores Plasminogénicos/genética , Inhibidor Tisular de Metaloproteinasa-1/genética , Inhibidor Tisular de Metaloproteinasa-1/metabolismo
15.
Nat Med ; 7(5): 575-83, 2001 May.
Artículo en Inglés | MEDLINE | ID: mdl-11329059

RESUMEN

Vascular endothelial growth factor (VEGF) stimulates angiogenesis by activating VEGF receptor-2 (VEGFR-2). The role of its homolog, placental growth factor (PlGF), remains unknown. Both VEGF and PlGF bind to VEGF receptor-1 (VEGFR-1), but it is unknown whether VEGFR-1, which exists as a soluble or a membrane-bound type, is an inert decoy or a signaling receptor for PlGF during angiogenesis. Here, we report that embryonic angiogenesis in mice was not affected by deficiency of PlGF (Pgf-/-). VEGF-B, another ligand of VEGFR-1, did not rescue development in Pgf-/- mice. However, loss of PlGF impaired angiogenesis, plasma extravasation and collateral growth during ischemia, inflammation, wound healing and cancer. Transplantation of wild-type bone marrow rescued the impaired angiogenesis and collateral growth in Pgf-/- mice, indicating that PlGF might have contributed to vessel growth in the adult by mobilizing bone-marrow-derived cells. The synergism between PlGF and VEGF was specific, as PlGF deficiency impaired the response to VEGF, but not to bFGF or histamine. VEGFR-1 was activated by PlGF, given that anti-VEGFR-1 antibodies and a Src-kinase inhibitor blocked the endothelial response to PlGF or VEGF/PlGF. By upregulating PlGF and the signaling subtype of VEGFR-1, endothelial cells amplify their responsiveness to VEGF during the 'angiogenic switch' in many pathological disorders.


Asunto(s)
Permeabilidad Capilar , Factores de Crecimiento Endotelial/fisiología , Linfocinas/fisiología , Neoplasias Experimentales/irrigación sanguínea , Neovascularización Patológica , Proteínas Gestacionales/fisiología , Animales , Secuencia de Bases , Cartilla de ADN , Desarrollo Embrionario y Fetal , Ratones , Factor de Crecimiento Placentario , Plasma , Proteínas Gestacionales/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular , Cicatrización de Heridas/fisiología
16.
J Exp Med ; 185(5): 963-8, 1997 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-9120402

RESUMEN

The plasminogen/plasmin system has the potential to affect the outcome of inflammatory diseases by regulating accumulation of fibrin and other matrix proteins. In human and experimental crescentic glomerulonephritis (GN), fibrin is an important mediator of glomerular injury and renal impairment. Glomerular deposition of matrix proteins is a feature of progressive disease. To study the role of plasminogen and plasminogen activators in the development of inflammatory glomerular injury, GN was induced in mice in which the genes for these proteins had been disrupted by homologous recombination. Deficiency of plasminogen or combined deficiency of tissue type plasminogen activator (tPA) and urokinase type plasminogen activator (uPA) was associated with severe functional and histological exacerbation of glomerular injury. Deficiency of tPA, the predominant plasminogen activator expressed in glomeruli, also exacerbated disease. uPA deficiency reduced glomerular macrophage infiltration and did not significantly exacerbate disease. uPA receptor deficiency did not effect the expression of GN. These studies demonstrate that plasminogen plays an important role in protecting the glomerulus from acute inflammatory injury and that tPA is the major protective plasminogen activator.


Asunto(s)
Glomerulonefritis/etiología , Riñón/patología , Plasminógeno/metabolismo , Activador de Tejido Plasminógeno/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Animales , Fibrina/metabolismo , Glomerulonefritis/inmunología , Glomérulos Renales/patología , Ratones , Ratones Mutantes , Plasminógeno/genética , Insuficiencia Renal/etiología , Activador de Tejido Plasminógeno/genética , Activador de Plasminógeno de Tipo Uroquinasa/genética
17.
J Med Genet ; 46(12): 840-6, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18413368

RESUMEN

BACKGROUND: Targeted delivery of the angiogenic factor, vascular endothelial growth factor (VEGF), to motor neurons prolongs survival in rodent models of amyotrophic lateral sclerosis (ALS), while mice expressing reduced VEGF concentrations develop motor neuron degeneration reminiscent of ALS, raising the question whether VEGF contributes to the pathogenesis of ALS. An initial association study reported that VEGF haplotypes conferred increased susceptibility to ALS in humans, but later studies challenged this initial finding. METHODS AND FINDINGS: A meta-analysis was undertaken to critically reappraise whether any of the three common VEGF gene variations (-2578C/A, -1154G/A and -634G/C) increase the risk of ALS. Over 7000 subjects from eight European and three American populations were included in the analysis. Pooled odds ratios were calculated using fixed-effects and random-effects models, and four potential sources of heterogeneity (location of disease onset, gender, age at disease onset and disease duration) were assessed. After correction, none of the genotypes or haplotypes was significantly associated with ALS. Subgroup analysis by gender revealed, however, that the -2578AA genotype, which lowers VEGF expression, increased the risk of ALS in males (OR = 1.46 males vs females; 95% CI = 1.19 to 1.80; p = 7.8 10E-5), even after correction for publication bias and multiple testing. CONCLUSIONS: This meta-analysis does not support the original conclusion that VEGF haplotypes increase the risk of ALS in humans, but the significant association of the low-VEGF -2578AA genotype with increased susceptibility to ALS in males reappraises the link between reduced VEGF concentrations and ALS, as originally revealed by the fortuitous mouse genetic studies.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Factor A de Crecimiento Endotelial Vascular/genética , Esclerosis Amiotrófica Lateral/patología , Animales , Modelos Animales de Enfermedad , Predisposición Genética a la Enfermedad , Heterocigoto , Humanos , Masculino , Ratones , Neuronas Motoras/patología , Polimorfismo de Nucleótido Simple , Factores Sexuales
18.
J Cell Biol ; 140(1): 233-45, 1998 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-9425170

RESUMEN

It has been proposed that the urokinase receptor (u-PAR) is essential for the various biological roles of urokinase-type plasminogen activator (u-PA) in vivo, and that smooth muscle cells require u-PA for migration during arterial neointima formation. The present study was undertaken to evaluate the role of u-PAR during this process in mice with targeted disruption of the u-PAR gene (u-PAR-/-). Surprisingly, u-PAR deficiency did not affect arterial neointima formation, neointimal cell accumulation, or migration of smooth muscle cells. Indeed, topographic analysis of arterial wound healing after electric injury revealed that u-PAR-/- smooth muscle cells, originating from the uninjured borders, migrated over a similar distance and at a similar rate into the necrotic center of the wound as wild-type (u-PAR+/+) smooth muscle cells. In addition, u-PAR deficiency did not impair migration of wounded cultured smooth muscle cells in vitro. There were no genotypic differences in reendothelialization of the vascular wound. The minimal role of u-PAR in smooth muscle cell migration was not because of absent expression, since wild-type smooth muscle cells expressed u-PAR mRNA and functional receptor in vitro and in vivo. Pericellular plasmin proteolysis, evaluated by degradation of 125I-labeled fibrin and activation of zymogen matrix metalloproteinases, was similar for u-PAR-/- and u-PAR+/+ cells. Immunoelectron microscopy of injured arteries in vivo revealed that u-PA was bound on the cell surface of u-PAR+/+ cells, whereas it was present in the pericellular space around u-PAR-/- cells. Taken together, these results suggest that binding of u-PA to u-PAR is not required to provide sufficient pericellular u-PA-mediated plasmin proteolysis to allow cellular migration into a vascular wound.


Asunto(s)
Arteria Femoral/fisiología , Fibrinolisina/metabolismo , Metaloendopeptidasas/metabolismo , Músculo Liso Vascular/fisiología , Receptores de Superficie Celular/fisiología , Túnica Íntima/fisiología , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Cicatrización de Heridas/fisiología , Animales , Células Cultivadas , Quimiotaxis/genética , Quimiotaxis/fisiología , Femenino , Arteria Femoral/citología , Arteria Femoral/lesiones , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Ratones Noqueados , Microscopía Inmunoelectrónica , Músculo Liso Vascular/citología , Músculo Liso Vascular/lesiones , Receptores de Superficie Celular/deficiencia , Receptores de Superficie Celular/genética , Receptores del Activador de Plasminógeno Tipo Uroquinasa , Regeneración , Túnica Íntima/citología , Túnica Íntima/lesiones , Cicatrización de Heridas/genética
19.
J Cell Biol ; 137(4): 953-63, 1997 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-9151696

RESUMEN

The middle T antigen of murine Polyomavirus (PymT) rapidly transforms endothelial cells, leading to the formation of vascular tumors in newborn mice. Transformed endothelial (End.) cell lines established from such tumors exhibit altered proteolytic activity as a result of increased expression of urokinase-type plasminogen activator (uPA) and are capable of inducing vascular tumors efficiently when injected into adult mice. In this study we have used mice lacking components of the PA/plasmin system to analyze the role of this system in the transformation process and in tumor growth. We found that the proteolytic status of the host is not a critical determinant for PymT-induced vascular tumor formation. In addition, the lack of either uPA or tissue-type PA (tPA) activity is not limiting for the establishment and proliferation of End. cells in vitro, although the combined loss of both PA activities leads to a marked reduction in proliferation rates. Furthermore, the in vitro morphogenetic properties of mutant End. cells in fibrin gels could only be correlated with an altered proteolytic status in cells lacking both uPA and tPA. However, in contrast with tumors induced by PymT itself, the tumorigenic potential of mutant and wild-type End. cell lines was found to be highly dependent on the proteolytic status of both the tumor cells and the host. Thus, genetic alterations in the PA/plasmin system affect vascular tumor development, indicating that this system is a causal component in PymTmediated oncogenesis.


Asunto(s)
Antígenos Transformadores de Poliomavirus , Transformación Celular Viral , Endotelio Vascular/citología , Fibrinolisina/fisiología , Activadores Plasminogénicos/fisiología , Neoplasias Vasculares/etiología , Animales , Animales Recién Nacidos , Línea Celular , Endotelio Vascular/enzimología , Fibrina , Geles , Expresión Génica , Ratones , Ratones Noqueados , Morfogénesis , Inhibidor 1 de Activador Plasminogénico/deficiencia , ARN Mensajero/genética , Activador de Tejido Plasminógeno/deficiencia , Activador de Plasminógeno de Tipo Uroquinasa/deficiencia
20.
J Cell Biol ; 152(4): 777-84, 2001 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-11266468

RESUMEN

The plasminogen (Plg)/plasminogen activator (PA) system plays a key role in cancer progression, presumably via mediating extracellular matrix degradation and tumor cell migration. Consequently, urokinase-type PA (uPA)/plasmin antagonists are currently being developed for suppression of tumor growth and angiogenesis. Paradoxically, however, high levels of PA inhibitor 1 (PAI-1) are predictive of a poor prognosis for survival of patients with cancer. We demonstrated previously that PAI-1 promoted tumor angiogenesis, but by an unresolved mechanism. We anticipated that PAI-1 facilitated endothelial cell migration via its known interaction with vitronectin (VN) and integrins. However, using adenoviral gene transfer of PAI-1 mutants, we observed that PAI-1 promoted tumor angiogenesis, not by interacting with VN, but rather by inhibiting proteolytic activity, suggesting that excessive plasmin proteolysis prevents assembly of tumor vessels. Single deficiency of uPA, tissue-type PA (tPA), uPA receptor, or VN, as well as combined deficiencies of uPA and tPA did not impair tumor angiogenesis, whereas lack of Plg reduced it. Overall, these data indicate that plasmin proteolysis, even though essential, must be tightly controlled during tumor angiogenesis, probably to allow vessel stabilization and maturation. These data provide insights into the clinical paradox whereby PAI-1 promotes tumor progression and warrant against the uncontrolled use of uPA/plasmin antagonists as tumor angiogenesis inhibitors.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Endopeptidasas/metabolismo , Neoplasias Experimentales/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Inhibidor 1 de Activador Plasminogénico/farmacología , Vitronectina/metabolismo , Animales , Endotelio Vascular/efectos de los fármacos , Fibrinolisina/metabolismo , Queratinocitos/patología , Ratones , Ratones Mutantes , Neoplasias de los Músculos/irrigación sanguínea , Invasividad Neoplásica , Neoplasias Experimentales/irrigación sanguínea , Neovascularización Patológica/etiología , Inhibidor 1 de Activador Plasminogénico/metabolismo , Unión Proteica , Vitronectina/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA