Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Angiogenesis ; 16(1): 15-28, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22903372

RESUMEN

The 5-hydroxytryptamine type 4 receptor (5-HT(4)R) regulates many physiological processes, including learning and memory, cognition, and gastrointestinal motility. Little is known about its role in angiogenesis. Using mouse hindlimb ischemia model of angiogenesis, we observed a significant reduction of limb blood flow recovery 14 days after ischemia and a decrease in density of CD31-positive vessels in adductor muscles in 5-HT(4)R(-/-) mice compared to wild type littermates. Our in vitro data indicated that 5-HT(4)R endogenously expressed in endothelial cells (ECs) may promote angiogenesis. Inhibition of the receptor with 5-HT(4)R antagonist RS 39604 reduced EC capillary tube formation in the reconstituted basement membrane. Using Boyden chamber migration assay and wound healing "scratch" assay, we demonstrated that RS 39604 treatment significantly suppressed EC migration. Transendothelial resistance measurement and immunofluorescence analysis showed that a 5-HT(4)R agonist RS 67333 led to an increase in endothelial permeability, actin stress fiber and interendothelial gap formation. Importantly, we provided the evidence that 5-HT(4)R-regulated EC migration may be mediated by Gα13 and RhoA. Our results suggest a prominent role of 5-HT(4)R in promoting angiogenesis and identify 5-HT(4)R as a potential therapeutic target for modulating angiogenesis under pathological conditions.


Asunto(s)
Células Endoteliales de la Vena Umbilical Humana/metabolismo , Neovascularización Fisiológica , Receptores de Serotonina 5-HT4/metabolismo , Compuestos de Anilina/farmacología , Animales , Capilares/efectos de los fármacos , Capilares/crecimiento & desarrollo , Adhesión Celular/efectos de los fármacos , Permeabilidad de la Membrana Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Activación Enzimática/efectos de los fármacos , Femenino , Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/enzimología , Células Endoteliales de la Vena Umbilical Humana/patología , Isquemia/metabolismo , Isquemia/patología , Ratones , Músculos/irrigación sanguínea , Músculos/patología , Neovascularización Fisiológica/efectos de los fármacos , Piperidinas/farmacología , Propano/análogos & derivados , Propano/farmacología , Flujo Sanguíneo Regional/efectos de los fármacos , Agonistas del Receptor de Serotonina 5-HT4/farmacología , Antagonistas del Receptor de Serotonina 5-HT4/farmacología , Cicatrización de Heridas/efectos de los fármacos , Proteína de Unión al GTP rhoA/metabolismo
2.
J Cell Physiol ; 226(5): 1255-64, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-20945373

RESUMEN

Vasodilator-stimulated phosphoprotein (VASP) is implicated in the protection of the endothelial barrier in vitro and in vivo. The function of VASP in thrombin signaling in the endothelial cells (ECs) is not known. For the first time we studied the effects of VASP deficiency on EC permeability and pulmonary vascular permeability in response to thrombin receptor stimulation. We provided the evidence that VASP deficiency potentiates the increase in endothelial permeability induced by activation of thrombin receptor in cultured human umbilical vein endothelial cells (HUVECs) and isolated mouse lungs. Using transendothelial resistance measurement, we showed that siRNA-mediated VASP downregulation in HUVECs leads to a potentiation of thrombin- and protease-activated receptor 1 (PAR-1) agonist-induced increase in endothelial permeability. Compared to control cells, VASP-deficient HUVECs had delayed endothelial junctional reassembly and abrogated VE-cadherin cytoskeletal anchoring in the recovery phase after thrombin stimulation, as demonstrated by immunofluorescence studies and cell fractionation analysis, respectively. Measurement of the capillary filtration coefficient in isolated mouse lungs demonstrated that VASP(-/-) mice have increased microvascular permeability in response to infusion with PAR-1 agonist compared to wild type mice. Lack of VASP led to decreased Rac1 activation both in VASP-deficient HUVECs after thrombin stimulation and VASP(-/-) mouse lungs after PAR-1 agonist infusion, indicating that VASP effects on thrombin signaling may be correlated with changes in Rac1 activity. This study demonstrates that VASP may play critical and complex role in the regulation of thrombin-dependent disruption of the endothelial barrier function.


Asunto(s)
Permeabilidad Capilar , Moléculas de Adhesión Celular/deficiencia , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Pulmón/irrigación sanguínea , Proteínas de Microfilamentos/deficiencia , Fosfoproteínas/deficiencia , Receptor PAR-1/metabolismo , Trombina/metabolismo , Animales , Antígenos CD/metabolismo , Cadherinas/metabolismo , Moléculas de Adhesión Celular/genética , Células Cultivadas , Impedancia Eléctrica , Humanos , Uniones Intercelulares/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Microfilamentos/genética , Neuropéptidos/metabolismo , Fosfoproteínas/genética , Interferencia de ARN , Factores de Tiempo , Transfección , Regulación hacia Arriba , Proteínas de Unión al GTP rac/metabolismo , Proteína de Unión al GTP rac1/metabolismo
3.
Circ Res ; 105(6): 549-56, 2009 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-19679840

RESUMEN

RATIONALE: Disruption of endothelial barrier function and neutrophil-mediated injury are two major mechanisms underlying the pathophysiology of sepsis-induced acute lung injury (ALI). Recently we reported that endotoxin induced activation of RhoA in mice lungs that led to the disruption of endothelial barrier and lung edema formation; however, the molecular mechanism of this phenomenon remained unknown. OBJECTIVE: We reasoned that LIMK1, which participates in the regulation of endothelial cell contractility and is activated by RhoA/Rho kinase pathway, could mediate RhoA-dependent disruption of endothelial barrier function in mouse lungs during ALI. And if that is the case, then attenuation of endothelial cell contractility by downregulating LIMK1 may lead to the enhancement of endothelial barrier function, which could protect mice from endotoxin-induced ALI. METHODS AND RESULTS: Here we report that LIMK1 deficiency in mice significantly reduced mortality induced by endotoxin. Data showed that lung edema formation, lung microvascular permeability, and neutrophil infiltration into the lungs were suppressed in limk1(-/-) mice. CONCLUSIONS: We identified that improvement of endothelial barrier function along with impaired neutrophil chemotaxis were the underlying mechanisms that reduced severity of ALI in limk1(-/-) mice, pointing to a new therapeutic target for diseases associated with acute inflammation of the lungs.


Asunto(s)
Lesión Pulmonar Aguda/enzimología , Endotelio/enzimología , Quinasas Lim/metabolismo , Infiltración Neutrófila , Neutrófilos/enzimología , Sepsis/enzimología , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/genética , Animales , Quimiotaxis/efectos de los fármacos , Endotelio/patología , Humanos , Quinasas Lim/genética , Lipopolisacáridos/toxicidad , Pulmón/enzimología , Pulmón/patología , Ratones , Ratones Noqueados , Neutrófilos/patología , Edema Pulmonar/inducido químicamente , Edema Pulmonar/enzimología , Edema Pulmonar/genética , Sepsis/inducido químicamente , Sepsis/genética , Proteínas de Unión al GTP rho/genética , Proteínas de Unión al GTP rho/metabolismo , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA
4.
J Immunol ; 182(12): 7997-8004, 2009 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-19494325

RESUMEN

Bacterial LPS induces rapid thrombocytopenia, hypotension, and sepsis. Although growing evidence suggests that platelet activation plays a critical role in LPS-induced thrombocytopenia and tissue damage, the mechanism of LPS-mediated platelet activation is unclear. In this study, we show that LPS stimulates platelet secretion of dense and alpha granules as indicated by ATP release and P-selectin expression, and thus enhances platelet activation induced by low concentrations of platelet agonists. Platelets express components of the LPS receptor-signaling complex, including TLR (TLR4), CD14, MD2, and MyD88, and the effect of LPS on platelet activation was abolished by an anti-TLR4-blocking Ab or TLR4 knockout, suggesting that the effect of LPS on platelet aggregation requires the TLR4 pathway. Furthermore, LPS-potentiated thrombin- and collagen-induced platelet aggregation and FeCl(3)-induced thrombus formation were abolished in MyD88 knockout mice. LPS also induced cGMP elevation and the stimulatory effect of LPS on platelet aggregation was abolished by inhibitors of NO synthase and the cGMP-dependent protein kinase (PKG). LPS-induced cGMP elevation was inhibited by an anti-TLR4 Ab or by TLR4 deficiency, suggesting that activation of the cGMP/protein kinase G pathway by LPS involves the TLR4 pathway. Taken together, our data indicate that LPS stimulates platelet secretion and potentiates platelet aggregation through a TLR4/MyD88- and cGMP/PKG-dependent pathway.


Asunto(s)
Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Lipopolisacáridos/farmacología , Factor 88 de Diferenciación Mieloide/metabolismo , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 4/metabolismo , Animales , GMP Cíclico/metabolismo , Humanos , Receptores de Lipopolisacáridos/metabolismo , Antígeno 96 de los Linfocitos/metabolismo , Ratones , Agregación Plaquetaria/efectos de los fármacos , Unión Proteica , Trombina/metabolismo
5.
J Cell Physiol ; 223(1): 94-102, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20039275

RESUMEN

T-cadherin is an atypical member of the cadherin family, which lacks the transmembrane and intracellular domains and is attached to the plasma membrane via a glycosylphosphatidylinositol anchor. Unlike canonical cadherins, it is believed to function primarily as a signaling molecule. T-cadherin is highly expressed in endothelium. Using transendothelial electrical resistance measurements and siRNA-mediated depletion of T-cadherin in human umbilical vein endothelial cells, we examined its involvement in regulation of endothelial barrier. We found that in resting confluent monolayers adjusted either to 1% or 10% serum, T-cadherin depletion modestly, but consistently reduced transendothelial resistance. This was accompanied by increased phosphorylation of Akt and LIM kinase, reduced phosphorylation of p38 MAP kinase, but no difference in tubulin acetylation and in phosphorylation of an actin filament severing protein cofilin and myosin light chain kinase. Serum stimulation elicited a biphasic increase in resistance with peaks at 0.5 and 4-5 h, which was suppressed by a PI3 kinase/Akt inhibitor wortmannin and a p38 inhibitor SB 239063. T-cadherin depletion increased transendothelial resistance between the two peaks and reduced the amplitude of the second peak. T-cadherin depletion abrogated serum-induced Akt phosphorylation at Thr308 and reduced phosphorylation at Ser473, reduced phosphorylation of cofilin, and accelerated tubulin deacetylation. Adiponectin slightly improved transendothelial resistance irrespectively of T-cadherin depletion. T-cadherin depletion also resulted in a reduced sensitivity and delayed responses to thrombin. These data implicate T-cadherin in regulation of endothelial barrier function, and suggest a complex signaling network that links T-cadherin and regulation of barrier function.


Asunto(s)
Cadherinas/metabolismo , Permeabilidad Capilar , Células Endoteliales/metabolismo , Acetilación , Factores Despolimerizantes de la Actina/metabolismo , Adiponectina/metabolismo , Cadherinas/genética , Permeabilidad Capilar/efectos de los fármacos , Miosinas Cardíacas/metabolismo , Células Cultivadas , Impedancia Eléctrica , Células Endoteliales/efectos de los fármacos , Humanos , Quinasas Lim/metabolismo , Cadenas Ligeras de Miosina/metabolismo , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , Transducción de Señal , Trombina/metabolismo , Factores de Tiempo , Tubulina (Proteína)/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
6.
FASEB J ; 23(12): 4193-206, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19690217

RESUMEN

Protease-activated receptor 1 (PAR-1) mediates thrombin signaling in human endothelial cells. As a G-protein-coupled receptor, PAR-1 transmits thrombin signal through activation of the heterotrimeric G proteins, Gi, Gq, and G12/13. In this study, we demonstrated that zyxin, a LIM-domain-containing protein, is involved in thrombin-mediated actin cytoskeleton remodeling and serum response element (SRE)-dependent gene transcription. We determined that zyxin binds to the C-terminal domain of PAR-1, providing a possible mechanism of involvement of zyxin as a signal transducer in PAR-1 signaling. Data showing that disruption of PAR-1-zyxin interaction inhibited thrombin-induced stress fiber formation and SRE activation supports this hypothesis. Similarly, depletion of zyxin using siRNA inhibited thrombin-induced actin stress fiber formation and SRE-dependent gene transcription. In addition, depletion of zyxin resulted in delay of endothelial barrier restoration after thrombin treatment. Notably, down-regulation of zyxin did not affect thrombin-induced activation of RhoA or Gi, Gq, and G12/13 heterotrimeric G proteins, implicating a novel signaling pathway regulated by PAR-1 that is not mediated by G-proteins. The observation that zyxin targets VASP, a partner of zyxin in regulation of actin assembly and dynamics, to focal adhesions and along stress fibers on thrombin stimulation suggests that zyxin may participate in thrombin-induced cytoskeletal remodeling through recruitment of VASP. In summary, this study establishes a crucial role of zyxin in thrombin signaling in endothelial cells and provides evidence for a novel PAR-1 signaling pathway mediated by zyxin.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Células Endoteliales/metabolismo , Glicoproteínas/metabolismo , Receptor PAR-1/metabolismo , Transducción de Señal/fisiología , Trombina/metabolismo , Animales , Células COS , Moléculas de Adhesión Celular/metabolismo , Chlorocebus aethiops , Proteínas del Citoesqueleto/genética , Regulación de la Expresión Génica/fisiología , Glicoproteínas/genética , Proteínas de Unión al GTP Heterotriméricas/metabolismo , Humanos , Proteínas de Microfilamentos/metabolismo , Fosfoproteínas/metabolismo , Transporte de Proteínas , Receptor PAR-1/genética , Trombina/genética , Zixina
7.
Am J Physiol Cell Physiol ; 297(5): C1168-77, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19726744

RESUMEN

T-cadherin (H-cadherin, cadherin 13) is upregulated in vascular proliferative disorders and in tumor-associated neovascularization and is deregulated in many cancers. Unlike canonical cadherins, it lacks transmembrane and intracellular domains and is attached to the plasma membrane via a glycosylphosphatidylinositol anchor. T-cadherin is thought to function in signaling rather than as an adhesion molecule. Some interactive partners of T-cadherin at the plasma membrane have recently been identified. We examined T-cadherin location in human endothelial cells using confocal microscopy and subcellular fractionation. We found that a considerable proportion of T-cadherin is located in the nucleus and in the centrosomes. T-cadherin colocalized with a centrosomal marker gamma-tubulin uniformly throughout the cell cycle at least in human umbilical vein endothelial cells. In the telophase, T-cadherin transiently concentrated in the midbody and was apparently degraded. Its overexpression resulted in an increase in the number of multinuclear cells, whereas its downregulation by small interfering RNA led to an increase in the number of cells with multiple centrosomes. These findings indicate that deregulation of T-cadherin in endothelial cells may lead to disturbances in cytokinesis or centrosomal replication.


Asunto(s)
Cadherinas/metabolismo , Núcleo Celular/metabolismo , Centrosoma/metabolismo , Citocinesis/fisiología , Células Endoteliales/metabolismo , Western Blotting , Técnica del Anticuerpo Fluorescente , Humanos , Inmunoprecipitación , Microscopía Confocal , Transporte de Proteínas/fisiología , ARN Interferente Pequeño , Tubulina (Proteína)/metabolismo
8.
Angiogenesis ; 12(1): 1-15, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19093215

RESUMEN

The alpha subunit of heterotrimeric G13 protein is required for the embryonic angiogenesis (Offermanns et al., Science 275:533-536, 1997). However, the molecular mechanism of Galpha13-dependent angiogenesis is not understood. Here, we show that myocyte-specific enhancer factor-2 (MEF2) mediates Galpha13-dependent angiogenesis. Our data showed that constitutively activated Galpha13Q226L stimulated MEF2-dependent gene transcription. In addition, downregulation of endogenous Galpha13 inhibited thrombin-stimulated MEF2-dependent gene transcription in endothelial cells. Both Ca(2+)/calmodulin-dependent kinase IV (CaMKIV) and histone deacetylase 5 (HDAC5) were involved in Galpha13-mediated MEF2-dependent gene transcription. Galpha13Q226L also increased Ca(2+)/calmodulin-independent CaMKIV activity, while dominant negative mutant of CaMKIV inhibited MEF2-dependent gene transcription induced by Galpha13Q226L. Furthermore, Galpha13Q226L was able to derepress HDAC5-mediated repression of gene transcription and induce the translocation of HDAC5 from nucleus to cytoplasm. Finally, downregulation of endogenous Galpha13 and MEF2 proteins in endothelial cells reduced cell proliferation and capillary tube formation. Decrease of endothelial cell proliferation that was caused by the Galpha13 downregulation was partially restored by the constitutively active MEF2-VP16. Our studies suggest that MEF2 proteins are an important component in Galpha13-mediated angiogenesis.


Asunto(s)
Células Endoteliales/metabolismo , Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Factores Reguladores Miogénicos/metabolismo , Neovascularización Fisiológica , Transcripción Genética , Animales , Proteína Quinasa Tipo 4 Dependiente de Calcio Calmodulina , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/enzimología , Histona Desacetilasas/metabolismo , Humanos , Factores de Transcripción MEF2 , Ratones , Proteínas Mutantes/metabolismo , Células 3T3 NIH , Neovascularización Fisiológica/efectos de los fármacos , Proteína Fosfatasa 2/metabolismo , Transporte de Proteínas/efectos de los fármacos , Proteínas Represoras/metabolismo , Trombina/farmacología , Transcripción Genética/efectos de los fármacos , Venas Umbilicales/citología
9.
FASEB J ; 22(8): 2821-31, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18367648

RESUMEN

G alpha12 constitutes, along with G alpha13, one of the four families of alpha subunits of heterotrimeric G proteins. We found that the N terminus of G alpha12, but not those of other G alpha subunits, contains a predicted mitochondrial targeting sequence. Using confocal microscopy and cell fractionation, we demonstrated that up to 40% of endogenous G alpha12 in human umbilical vein endothelial cells colocalize with mitochondrial markers. N-terminal sequence of G alpha12 fused to GFP efficiently targeted the fusion protein to mitochondria. G alpha12 with mutated mitochondrial targeting sequence was still located in mitochondria, suggesting the existence of additional mechanisms for mitochondrial localization. Lysophosphatidic acid, one of the known stimuli transduced by G alpha12/13, inhibited mitochondrial motility, while depletion of endogenous G alpha12 increased mitochondrial motility. G alpha12Q229L variants uncoupled from RhoGEFs (but not fully functional activated G alpha12Q229L) induced transformation of the mitochondrial network into punctate mitochondria and resulted in a loss of mitochondrial membrane potential. All examined G alpha12Q229L variants reduced phosphorylation of Bcl-2 at Ser-70, while only mutants unable to bind RhoGEFs also decreased cellular levels of Bcl-2. These G alpha12 mutants were also more efficient Hsp90 interactors. These findings are the first demonstration of a heterotrimeric G protein alpha subunit specifically targeted to mitochondria and involved in the control of mitochondrial morphology and dynamics.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Mitocondrias/metabolismo , Animales , Células COS , Células Cultivadas , Chlorocebus aethiops , Células Endoteliales/metabolismo , Células Endoteliales/ultraestructura , Subunidades alfa de la Proteína de Unión al GTP G12-G13/antagonistas & inhibidores , Subunidades alfa de la Proteína de Unión al GTP G12-G13/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Potencial de la Membrana Mitocondrial , Movimiento , Mutagénesis Sitio-Dirigida , Fosforilación , Unión Proteica , Proteínas Proto-Oncogénicas c-bcl-2/química , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , ARN Interferente Pequeño/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Factores de Intercambio de Guanina Nucleótido Rho
10.
Circ Res ; 101(1): 50-8, 2007 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-17525371

RESUMEN

Rho family GTPases have been implicated in the regulation of endothelial permeability via their actions on actin cytoskeletal organization and integrity of interendothelial junctions. In cell culture studies, activation of RhoA disrupts interendothelial junctions and increases endothelial permeability, whereas activation of Rac1 and Cdc42 enhances endothelial barrier function by promoting the formation of restrictive junctions. The primary regulators of Rho proteins, guanine nucleotide dissociation inhibitors (GDIs), form a complex with the GDP-bound form of the Rho family of monomeric G proteins, and thus may serve as a nodal point regulating the activation state of RhoGTPases. In the present study, we addressed the in vivo role of RhoGDI-1 in regulating pulmonary microvascular permeability using RhoGDI-1(-/-) mice. We observed that basal endothelial permeability in lungs of RhoGDI-1(-/-) mice was 2-fold greater than wild-type mice. This was the result of opening of interendothelial junctions in lung microvessels which are normally sealed. The activity of RhoA (but not of Rac1 or Cdc42) was significantly increased in RhoGDI-1(-/-) lungs as well as in cultured endothelial cells on downregulation of RhoGDI-1 with siRNA, consistent with RhoGDI-1-mediated modulation RhoA activity. Thus, RhoGDI-1 by repressing RhoA activity regulates lung microvessel endothelial barrier function in vivo. In this regard, therapies augmenting endothelial RhoGDI-1 function may be beneficial in reestablishing the endothelial barrier and lung fluid balance in lung inflammatory diseases such as acute respiratory distress syndrome.


Asunto(s)
Barrera Alveolocapilar/enzimología , Permeabilidad Capilar , Endotelio Vascular/citología , Endotelio Vascular/enzimología , Inhibidores de Disociación de Guanina Nucleótido/fisiología , Pulmón/enzimología , Proteínas de Unión al GTP rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Animales , Barrera Alveolocapilar/fisiopatología , Permeabilidad Capilar/genética , Células Endoteliales/enzimología , Células Endoteliales/fisiología , Endotelio Vascular/fisiología , Inhibidores de Disociación de Guanina Nucleótido/deficiencia , Inhibidores de Disociación de Guanina Nucleótido/genética , Pulmón/patología , Pulmón/fisiopatología , Enfermedades Pulmonares/enzimología , Enfermedades Pulmonares/genética , Enfermedades Pulmonares/fisiopatología , Ratones , Ratones Noqueados , Inhibidores de la Disociación del Nucleótido Guanina rho-Específico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA