Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
PLoS One ; 13(7): e0200691, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30016341

RESUMEN

BACKGROUND: As an effort to reduce hospital readmissions, early follow-up visits were recommended by the Society of Hospital Medicine. However, published literature on the effect of follow-up visits is limited with mixed conclusions. Our goal here is to fully explore the relationship between follow-up visits and the all-cause non-elective 30-day readmission rate (RR) after adjusting for confounders. METHODS AND RESULTS: To conduct this retrospective observational study, we extracted data for 55,378 adult inpatients from Advocate Health Care, a large, multi-hospital system serving a diverse population in a major metropolitan area. These patients were discharged to Home or Home with Home Health services between June 1, 2013 and April 30, 2015. Our findings from time-dependent Cox proportional hazard models showed that follow-up visits were significantly associated with a reduced RR (adjusted hazard ratio: 0.86; 95% CI: 0.82-0.91), but in a complicated way because the interaction between follow-up visits and a readmission risk score was significant with p-value < 0.001. Our analysis using logistic models on an adjusted data set confirmed the above findings with the following additional results. First, time matter. Follow-up visits within 2 days were associated with the greatest reduction in RR (adjusted odds ratio: 0.72; 95% CI: 0.63-0.83). Visits beyond 2 days were also associated with a reduction in RR, but the strength of the effect decreased as the time between discharge and follow-up visit increased. Second, the strength of such association varied for patients with different readmission risk scores. Patients with a risk score of 0.113, high but not extremely high risk, had the greatest reduction in RR from follow-up visits. Patients with an extremely high risk score (> 0.334) saw no RR reduction from follow-up visits. Third, a patient was much more likely to have a 2-day follow-up visit if that visit was scheduled before the patient was discharged from the hospital (30% versus < 5%). CONCLUSIONS: Follow-up visits are associated with a reduction in readmission risk. The timing of follow-up visits can be important: beyond two days, the earlier, the better. The effect of follow-up visits is more significant for patients with a high but not extremely high risk of readmission.


Asunto(s)
Modelos Biológicos , Readmisión del Paciente , Adulto , Femenino , Estudios de Seguimiento , Humanos , Masculino , Modelos de Riesgos Proporcionales , Estudios Retrospectivos , Medición de Riesgo
2.
Cell Signal ; 29: 1-11, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27667566

RESUMEN

Rapid changes in microtubule (MT) polymerization dynamics affect regional activity of small GTPases RhoA and Rac1, which play a key role in the regulation of actin cytoskeleton and endothelial cell (EC) permeability. This study tested the role of End Binding Protein-1 (EB1) in the mechanisms of increased and decreased EC permeability caused by thrombin and hepatocyte growth factor (HGF) and mediated by RhoA and Rac1 GTPases, respectively. Stimulation of human lung EC with thrombin inhibited peripheral MT growth, which was monitored by morphological and biochemical evaluation of peripheral MT and the levels of stabilized MT. In contrast, stimulation of EC with HGF promoted peripheral MT growth and protrusion of EB1-positive MT plus ends to the EC peripheral submembrane area. EB1 knockdown by small interfering RNA did not affect partial MT depolymerization, activation of Rho signaling, and permeability response to thrombin, but suppressed the HGF-induced endothelial barrier enhancement. EB1 knockdown suppressed HGF-induced activation of Rac1 and Rac1 cytoskeletal effectors cortactin and PAK1, impaired HGF-induced assembly of cortical cytoskeleton regulatory complex (WAVE-p21Arc-IQGAP1), and blocked HGF-induced enhancement of peripheral actin cytoskeleton and VE-cadherin-positive adherens junctions. Altogether, these data demonstrate a role for EB1 in coordination of MT-dependent barrier enhancement response to HGF, but show no involvement of EB1 in acute increase of EC permeability caused by the barrier disruptive agonist. The results suggest that increased peripheral EB1 distribution is a critical component of the Rac1-mediated pathway and peripheral cytoskeletal remodeling essential for agonist-induced EC barrier enhancement.


Asunto(s)
Permeabilidad de la Membrana Celular , Células Endoteliales/citología , Células Endoteliales/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Citoesqueleto de Actina/efectos de los fármacos , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Uniones Adherentes/efectos de los fármacos , Uniones Adherentes/metabolismo , Línea Celular , Permeabilidad de la Membrana Celular/efectos de los fármacos , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Técnicas de Silenciamiento del Gen , Factor de Crecimiento de Hepatocito/farmacología , Humanos , Microtúbulos/efectos de los fármacos , Microtúbulos/metabolismo , Transducción de Señal/efectos de los fármacos , Trombina/farmacología , Familia de Proteínas del Síndrome de Wiskott-Aldrich/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Proteínas Activadoras de ras GTPasa/metabolismo , Proteínas de Unión al GTP rho/metabolismo
3.
J Biol Chem ; 291(45): 23681-23692, 2016 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-27590342

RESUMEN

Agonist-induced activation of Rho GTPase signaling leads to endothelial cell (EC) permeability and may culminate in pulmonary edema, a devastating complication of acute lung injury. Cingulin is an adaptor protein first discovered in epithelium and is involved in the organization of the tight junctions. This study investigated the role of cingulin in control of agonist-induced lung EC permeability via interaction with RhoA-specific activator GEF-H1. The siRNA-induced cingulin knockdown augmented thrombin-induced EC permeability monitored by measurements of transendothelial electrical resistance and endothelial cell permeability for macromolecules. Increased thrombin-induced permeability in ECs with depleted cingulin was associated with increased activation of GEF-H1 and RhoA detected in pulldown activation assays. Increased GEF-H1 association with cingulin was essential for down-regulation of thrombin-induced RhoA barrier disruptive signaling. Using cingulin-truncated mutants, we determined that GEF-H1 interaction with the rod + tail domain of cingulin was required for inactivation of GEF-H1 and endothelial cell barrier preservation. The results demonstrate the role for association of GEF-H1 with cingulin as the mechanism of RhoA pathway inactivation and rescue of EC barrier after agonist challenge.


Asunto(s)
Permeabilidad Capilar , Células Endoteliales/metabolismo , Pulmón/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Microfilamentos/metabolismo , Trombina/metabolismo , Lesión Pulmonar Aguda/genética , Lesión Pulmonar Aguda/metabolismo , Lesión Pulmonar Aguda/fisiopatología , Línea Celular , Células Endoteliales/patología , Endotelio Vascular/metabolismo , Endotelio Vascular/fisiopatología , Humanos , Pulmón/fisiopatología , Proteínas de la Membrana/genética , Proteínas de Microfilamentos/genética , Mutación , Mapas de Interacción de Proteínas , Interferencia de ARN , ARN Interferente Pequeño/genética , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Transducción de Señal , Proteína de Unión al GTP rhoA/metabolismo
4.
Am J Physiol Lung Cell Mol Physiol ; 311(4): L800-L809, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27566003

RESUMEN

Oxidized 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphatidylcholine (OxPAPC) attenuates agonist-induced endothelial cell (EC) permeability and increases pulmonary endothelial barrier function via enhancement of both the peripheral actin cytoskeleton and cell junctions mediated by Rac1 and Cdc42 GTPases. This study evaluated the role for the multifunctional Rac1/Cdc42 effector and regulator, IQGAP1, as a molecular transducer of the OxPAPC-mediated EC barrier enhancing signal. IQGAP1 knockdown in endothelial cells by gene-specific siRNA abolished OxPAPC-induced enlargement of VE-cadherin-positive adherens junctions, suppressed peripheral accumulation of actin polymerization regulators, namely cortactin, N-WASP and Arp3, and attenuated remodeling of the peripheral actin cytoskeleton. Inhibition of OxPAPC-induced barrier enhancement by IQGAP1 knockdown was due to suppressed Rac1 and Cdc42 activation. Expression of an IQGAP1 truncated mutant showed that the GTPase regulatory domain (GRD) of IQGAP1 was essential for the OxPAPC-induced membrane localization of cortactin, adherens junction proteins VE-cadherin and p120-catenin as well as for EC permeability response. IQGAP1knockdown attenuated the protective effect of OxPAPC against thrombin-induced cell contraction, cell junction disruption and EC permeability. These results demonstrate for the first time the role of IQGAP1 as a critical transducer of OxPAPC-induced Rac1/Cdc42 signaling to the actin cytoskeleton and adherens junctions which promotes cortical cytoskeletal remodeling and EC barrier protective effects of oxidized phospholipids.

5.
J Biol Chem ; 290(7): 4097-109, 2015 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-25492863

RESUMEN

Hepatocyte growth factor (HGF) attenuates agonist-induced endothelial cell (EC) permeability and increases pulmonary endothelial barrier function via Rac-dependent enhancement of the peripheral actin cytoskeleton. However, the precise mechanisms of HGF effects on the peripheral cytoskeleton are not well understood. This study evaluated a role for Rac/Cdc42-specific guanine nucleotide exchange factor Asef and the multifunctional Rac effector, IQGAP1, in the mechanism of HGF-induced EC barrier enhancement. HGF induced Asef and IQGAP1 co-localization at the cell cortical area and stimulated formation of an Asef-IQGAP1 functional protein complex. siRNA-induced knockdown of Asef or IQGAP1 attenuated HGF-induced EC barrier enhancement. Asef knockdown attenuated HGF-induced Rac activation and Rac association with IQGAP1, and it abolished both IQGAP1 accumulation at the cell cortical layer and IQGAP1 interaction with actin cytoskeletal regulators cortactin and Arp3. Asef activation state was essential for Asef interaction with IQGAP1 and protein complex accumulation at the cell periphery. In addition to the previously reported role of the IQGAP1 RasGAP-related domain in the Rac-dependent IQGAP1 activation and interaction with its targets, we show that the IQGAP1 C-terminal domain is essential for HGF-induced IQGAP1/Asef interaction and Asef-Rac-dependent activation leading to IQGAP1 interaction with Arp3 and cortactin as a positive feedback mechanism of IQGAP1 activation. These results demonstrate a novel feedback mechanism of HGF-induced endothelial barrier enhancement via Asef/IQGAP1 interactions, which regulate the level of HGF-induced Rac activation and promote cortical cytoskeletal remodeling via IQGAP1-Arp3/cortactin interactions.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Permeabilidad de la Membrana Celular , Endotelio Vascular/metabolismo , Factor de Crecimiento de Hepatocito/farmacología , Arteria Pulmonar/metabolismo , Proteínas Activadoras de ras GTPasa/metabolismo , Western Blotting , Células Cultivadas , Endotelio Vascular/citología , Técnica del Anticuerpo Fluorescente , Humanos , Inmunoprecipitación , Arteria Pulmonar/citología , ARN Interferente Pequeño/genética , Factores de Intercambio de Guanina Nucleótido Rho/antagonistas & inhibidores , Factores de Intercambio de Guanina Nucleótido Rho/genética , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Proteínas Activadoras de ras GTPasa/antagonistas & inhibidores , Proteínas Activadoras de ras GTPasa/genética
6.
Mol Biol Cell ; 26(4): 636-50, 2015 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-25518936

RESUMEN

Increased levels of hepatocyte growth factor (HGF) in injured lungs may reflect a compensatory response to diminish acute lung injury (ALI). HGF-induced activation of Rac1 GTPase stimulates endothelial barrier protective mechanisms. This study tested the involvement of Rac-specific guanine nucleotide exchange factor Asef in HGF-induced endothelial cell (EC) cytoskeletal dynamics and barrier protection in vitro and in a two-hit model of ALI. HGF induced membrane translocation of Asef and stimulated Asef Rac1-specific nucleotide exchange activity. Expression of constitutively activated Asef mutant mimicked HGF-induced peripheral actin cytoskeleton enhancement. In contrast, siRNA-induced Asef knockdown or expression of dominant-negative Asef attenuated HGF-induced Rac1 activation evaluated by Rac-GTP pull down and FRET assay with Rac1 biosensor. Molecular inhibition of Asef attenuated HGF-induced peripheral accumulation of cortactin, formation of lamellipodia-like structures, and enhancement of VE-cadherin adherens junctions and compromised HGF-protective effect against thrombin-induced RhoA GTPase activation, Rho-dependent cytoskeleton remodeling, and EC permeability. Intravenous HGF injection attenuated lung inflammation and vascular leak in the two-hit model of ALI induced by excessive mechanical ventilation and thrombin signaling peptide TRAP6. This effect was lost in Asef(-/-) mice. This study shows for the first time the role of Asef in HGF-mediated protection against endothelial hyperpermeability and lung injury.


Asunto(s)
Permeabilidad de la Membrana Celular , Factores de Intercambio de Guanina Nucleótido/fisiología , Pulmón/metabolismo , Animales , Técnicas de Silenciamiento del Gen , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Factor de Crecimiento de Hepatocito/farmacología , Ratones , Interferencia de ARN , Factores de Intercambio de Guanina Nucleótido Rho , Transducción de Señal , Proteína de Unión al GTP rac1/metabolismo
7.
PLoS One ; 9(9): e105912, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25198505

RESUMEN

Microtubules (MT) play a vital role in many cellular functions, but their role in peripheral actin cytoskeletal dynamics which is essential for control of endothelial barrier and monolayer integrity is less understood. We have previously described the enhancement of lung endothelial cell (EC) barrier by hepatocyte growth factor (HGF) which was associated with Rac1-mediated remodeling of actin cytoskeleton. This study investigated involvement of MT-dependent mechanisms in the HGF-induced enhancement of EC barrier. HGF-induced Rac1 activation was accompanied by phosphorylation of stathmin, a regulator of MT dynamics. HGF also stimulated MT peripheral growth monitored by time lapse imaging and tracking analysis of EB-1-decorated MT growing tips, and increased the pool of acetylated tubulin. These effects were abolished by EC pretreatment with HGF receptor inhibitor, downregulation of Rac1 pathway, or by expression of a stathmin-S63A phosphorylation deficient mutant. Expression of stathmin-S63A abolished the HGF protective effects against thrombin-induced activation of RhoA cascade, permeability increase, and EC barrier dysfunction. These results demonstrate a novel MT-dependent mechanism of HGF-induced EC barrier regulation via Rac1/PAK1/stathmin-dependent control of MT dynamics.


Asunto(s)
Endotelio Vascular/fisiología , Factor de Crecimiento de Hepatocito/fisiología , Pulmón/fisiología , Microtúbulos/fisiología , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Humanos , Pulmón/metabolismo , Fosforilación , Estatmina/metabolismo , Fracciones Subcelulares/metabolismo , Proteína de Unión al GTP rac1/fisiología
8.
Mol Cell Biol ; 34(18): 3546-58, 2014 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-25022754

RESUMEN

Cross talk between the actin cytoskeleton and microtubules (MT) has been implicated in the amplification of agonist-induced Rho signaling, leading to increased vascular endothelial permeability. This study tested the involvement of actin-MT cross talk in the mechanisms of barrier enhancement induced by hepatocyte growth factor (HGF) and evaluated the role of the adaptor protein IQGAP1 in integrating the MT- and actin-dependent pathways of barrier enhancement. IQGAP1 knockdown by small interfering RNA attenuated the HGF-induced increase in endothelial barrier properties and abolished HGF-activated cortical actin dynamics. IQGAP1 reduction abolished HGF-induced peripheral accumulation of Rac cytoskeletal effector cortactin and cortical actin remodeling. In addition, HGF stimulated peripheral MT growth in an IQGAP1-dependent fashion. HGF also induced Rac1-dependent IQGAP1 association with the MT fraction and the formation of a protein complex containing end-binding protein 1 (EB1), IQGAP1, and cortactin. Decreasing endogenous IQGAP1 abolished HGF-induced EB1-cortactin colocalization at the cell periphery. In turn, expression of IQGAP1ΔC (IQGAP1 lacking the C-terminal domain) attenuated the cortactin association with EB1 and suppressed HGF-induced endothelial cell peripheral actin cytoskeleton enhancement. These results demonstrate for the first time the MT-actin cross talk mechanism of HGF-induced endothelial barrier enhancement and suggest that IQGAP1 functions as a hub linking HGF-induced signaling to MT and actin remodeling via EB1-IQGAP1-cortactin interactions.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Cortactina/metabolismo , Células Endoteliales/metabolismo , Factor de Crecimiento de Hepatocito/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Microtúbulos/metabolismo , Proteínas Activadoras de ras GTPasa/metabolismo , Cortactina/genética , Técnicas de Silenciamiento del Gen , Humanos , Proteínas Asociadas a Microtúbulos/genética , ARN Interferente Pequeño , Transducción de Señal
9.
Mol Biol Cell ; 25(13): 2006-16, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24829380

RESUMEN

Vascular integrity and the maintenance of blood vessel continuity are fundamental features of the circulatory system maintained through endothelial cell-cell junctions. Defects in the endothelial barrier become an initiating factor in several pathologies, including ischemia/reperfusion, tumor angiogenesis, pulmonary edema, sepsis, and acute lung injury. Better understanding of mechanisms stimulating endothelial barrier enhancement may provide novel therapeutic strategies. We previously reported that oxidized phospholipids (oxidized 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphocholine [OxPAPC]) promote endothelial cell (EC) barrier enhancement both in vitro and in vivo. This study examines the initiating mechanistic events triggered by OxPAPC to increase vascular integrity. Our data demonstrate that OxPAPC directly binds the cell membrane-localized chaperone protein, GRP78, associated with its cofactor, HTJ-1. OxPAPC binding to plasma membrane-localized GRP78 leads to GRP78 trafficking to caveolin-enriched microdomains (CEMs) on the cell surface and consequent activation of sphingosine 1-phosphate receptor 1, Src and Fyn tyrosine kinases, and Rac1 GTPase, processes essential for cytoskeletal reorganization and EC barrier enhancement. Using animal models of acute lung injury with vascular hyperpermeability, we observed that HTJ-1 knockdown blocked OxPAPC protection from interleukin-6 and ventilator-induced lung injury. Our data indicate for the first time an essential role of GRP78 and HTJ-1 in OxPAPC-mediated CEM dynamics and enhancement of vascular integrity.


Asunto(s)
Células Endoteliales/metabolismo , Proteínas de Choque Térmico/fisiología , Fosfatidilcolinas/fisiología , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Animales , Caveolinas/metabolismo , Células Cultivadas , Impedancia Eléctrica , Chaperón BiP del Retículo Endoplásmico , Endotelio Vascular/citología , Endotelio Vascular/fisiología , Proteínas del Choque Térmico HSP40/metabolismo , Humanos , Masculino , Microdominios de Membrana/metabolismo , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Oxidación-Reducción , Transporte de Proteínas , Arteria Pulmonar/citología , Receptores de Lisoesfingolípidos/metabolismo
10.
FASEB J ; 28(7): 3249-60, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24706358

RESUMEN

Suboptimal ventilator support or regional ventilation heterogeneity in inflamed lungs causes excessive tissue distension, which triggers stretch-induced pathological signaling and may lead to vascular leak and lung dysfunction. Focal adhesions (FAs) are cell-substrate adhesive complexes participating in cellular mechanotransduction and regulation of the Rho GTPase pathway. Stretch-induced Rho regulation remains poorly understood. We used human lung endothelial cells (ECs) exposed to pathological cyclic stretch (CS) at 18% distension to test the hypothesis that FA protein paxillin participates in CS-induced Rho activation by recruiting the Rho-specific guanine nucleotide exchange factor GEF-H1. CS induced phosphorylation of paxillin and activated p42/44-MAP kinase, Rho GTPase, and paxillin/GEF-H1/p42/44-MAPK association. CS caused nearly 2-fold increase in EC permeability, which was attenuated by paxillin knockdown. Expression of the paxillin-Y31/118F phosphorylation mutant decreased the CS-induced paxillin/GEF-H1 association (16.3 ± 4.1%), GEF-H1 activation (28.9 ± 9.2%), and EC permeability (28.7 ± 8.1%) but not CS-induced p42/44-MAPK activation. Inhibition of p42/44-MAPK suppressed CS-induced paxillin/GEF-H1 interactions (15.9 ± 7.9%), GEF-H1 activation (11.7 ± 4.3%), and disruption of EC monolayer. Expression of GEF-H1T678A lacking p42/44-MAPK phosphorylation site attenuated Rho activation (31.2±11.6%). We conclude that MAPK-dependent targeting of GEF-H1 to paxillin is involved in the regulation of CS-induced Rho signaling and EC permeability. This study proposes a novel concept of paxillin-GEF-H1-p42/44-MAPK module as a regulator of pathological mechanotransduction.-Gawlak, G., Tian, Y., O'Donnell, J. J., III, Tian, X., Birukova, A. A., Birukov, K. G. Paxillin mediates stretch-induced Rho signaling and endothelial permeability via assembly of paxillin-p42/44MAPK-GEF-H1 complex.


Asunto(s)
Permeabilidad Capilar/fisiología , Células Endoteliales/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Paxillin/metabolismo , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Células Cultivadas , Humanos , Pulmón/metabolismo , Pulmón/fisiología , Fosforilación/fisiología , Transducción de Señal/fisiología
11.
J Biol Chem ; 289(8): 5168-83, 2014 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-24352660

RESUMEN

Microtubule (MT) dynamics is involved in a variety of cell functions, including control of the endothelial cell (EC) barrier. Release of Rho-specific nucleotide exchange factor GEF-H1 from microtubules activates the Rho pathway of EC permeability. In turn, pathologic vascular leak can be prevented by treatment with atrial natriuretic peptide (ANP). This study investigated a novel mechanism of vascular barrier protection by ANP via modulation of GEF-H1 function. In pulmonary ECs, ANP suppressed thrombin-induced disassembly of peripheral MT and attenuated Rho signaling and cell retraction. ANP effects were mediated by the Rac1 GTPase effector PAK1. Activation of Rac1-PAK1 promoted PAK1 interaction with the Rho activator GEF-H1, inducing phosphorylation of total and MT-bound GEF-H1 and leading to attenuation of Rho-dependent actin remodeling. In vivo, ANP attenuated lung injury caused by excessive mechanical ventilation and TRAP peptide (TRAP/HTV), which was further exacerbated in ANP(-/-) mice. The protective effects of ANP against TRAP/HTV-induced lung injury were linked to the increased pool of stabilized MT and inactivation of Rho signaling via ANP-induced, PAK1-dependent inhibitory phosphorylation of GEF-H1. This study demonstrates a novel protective mechanism of ANP against pathologic hyperpermeability and suggests a novel pharmacological intervention for the prevention of increased vascular leak via PAK1-dependent modulation of GEF-H1 activity.


Asunto(s)
Factor Natriurético Atrial/metabolismo , Permeabilidad Capilar , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Actinas/metabolismo , Lesión Pulmonar Aguda/enzimología , Lesión Pulmonar Aguda/patología , Animales , Factor Natriurético Atrial/farmacología , Permeabilidad Capilar/efectos de los fármacos , Citoprotección/efectos de los fármacos , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Genes Dominantes , Células HEK293 , Humanos , Pulmón/irrigación sanguínea , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Microtúbulos/efectos de los fármacos , Microtúbulos/metabolismo , Modelos Biológicos , Cadenas Ligeras de Miosina/metabolismo , Fosforilación/efectos de los fármacos , Neumonía/enzimología , Neumonía/patología , Unión Proteica/efectos de los fármacos , Trombina/farmacología , Quinasas p21 Activadas/metabolismo , Proteína de Unión al GTP rac1/metabolismo
12.
Mol Biol Cell ; 24(17): 2678-88, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23864716

RESUMEN

Activation of the Rho GTPase pathway determines endothelial cell (EC) hyperpermeability after injurious stimuli. To date, feedback mechanisms of Rho down-regulation critical for barrier restoration remain poorly understood. We tested a hypothesis that Rho down-regulation and barrier recovery of agonist-stimulated ECs is mediated by the Ras family GTPase Rap1. Thrombin-induced EC permeability driven by rapid activation of the Rho GTPase pathway was followed by Src kinase-dependent phosphorylation of the Rap1-specific guanine nucleotide exchange factor (GEF) C3G, activation of Rap1, and initiation of EC barrier recovery. Knockdown experiments showed that Rap1 activation was essential for down-regulation of Rho signaling and actin stress fiber dissolution. Rap1 activation also enhanced interaction between adherens junction (AJ) proteins VE-cadherin and p120-catenin and stimulated AJ reannealing mediated by the Rap1 effector afadin. This mechanism also included Rap1-dependent membrane translocation of the Rac1-specific GEF Tiam1 and activation of Rac1-dependent peripheral cytoskeletal dynamics, leading to resealing of intercellular gaps. These data demonstrate that activation of the Rap1-afadin axis is a physiological mechanism driving restoration of barrier integrity in agonist-stimulated EC monolayers via negative-feedback regulation of Rho signaling, stimulation of actin peripheral dynamics, and reestablishment of cell-cell adhesive complexes.


Asunto(s)
Células Endoteliales/citología , Células Endoteliales/metabolismo , Proteínas de Microfilamentos/metabolismo , Trombina/farmacología , Proteínas de Unión al GTP rap1/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Citoesqueleto/genética , Citoesqueleto/metabolismo , Regulación hacia Abajo , Técnicas de Silenciamiento del Gen , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Fosforilación , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Proteínas de Unión al GTP rho/genética
13.
J Biol Chem ; 288(25): 18290-9, 2013 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-23653363

RESUMEN

p120-catenin is a multidomain intracellular protein, which mediates a number of cellular functions, including stabilization of cell-cell transmembrane cadherin complexes as well as regulation of actin dynamics associated with barrier function, lamellipodia formation, and cell migration via modulation of the activities of small GTPAses. One mechanism involves p120 catenin interaction with Rho GTPase activating protein (p190RhoGAP), leading to p190RhoGAP recruitment to cell periphery and local inhibition of Rho activity. In this study, we have identified a stretch of 23 amino acids within the C-terminal domain of p120 catenin as the minimal sequence responsible for the recruitment of p190RhoGAP (herein referred to as CRAD; catenin-RhoGAP association domain). Expression of the p120-catenin truncated mutant lacking the CRAD in endothelial cells attenuated effects of barrier protective oxidized phospholipid, OxPAPC. This effect was accompanied by inhibition of membrane translocation of p190RhoGAP, increased Rho signaling, as well as suppressed activation of Rac1 and its cytoskeletal effectors PAK1 (p21-activated kinase 1) and cortactin. Expression of p120 catenin-truncated mutant lacking CRAD also delayed the recovery process after thrombin-induced endothelial barrier disruption. Concomitantly, RhoA activation and downstream signaling were sustained for a longer period of time, whereas Rac signaling was inhibited. These data demonstrate a critical role for p120-catenin (amino acids 820-843) domain in the p120-catenin·p190RhoGAP signaling complex assembly, membrane targeting, and stimulation of p190RhoGAP activity toward inhibition of the Rho pathway and reciprocal up-regulation of Rac signaling critical for endothelial barrier regulation.


Asunto(s)
Cateninas/metabolismo , Permeabilidad de la Membrana Celular , Citoesqueleto/metabolismo , Células Endoteliales/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Uniones Adherentes/metabolismo , Antígenos CD/metabolismo , Sitios de Unión/genética , Western Blotting , Cadherinas/metabolismo , Cateninas/genética , Membrana Celular/metabolismo , Células Cultivadas , Células Endoteliales/efectos de los fármacos , Técnica del Anticuerpo Fluorescente , Proteínas Activadoras de GTPasa/genética , Factores de Intercambio de Guanina Nucleótido , Células HEK293 , Humanos , Mutación , Fosfatidilcolinas/metabolismo , Unión Proteica , Mapeo de Interacción de Proteínas , Proteínas Represoras , Trombina/farmacología , Quinasas p21 Activadas/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Catenina delta
14.
Am J Physiol Lung Cell Mol Physiol ; 304(11): L757-64, 2013 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-23585227

RESUMEN

Myofibroblast differentiation induced by transforming growth factor-ß (TGF-ß) is characterized by the expression of smooth muscle α-actin (SMA) and extracellular matrix proteins. We and others have previously shown that these changes are regulated by protein kinase A (PKA). Adrenomedullin (ADM) is a vasodilator peptide that activates cAMP/PKA signaling through the calcitonin-receptor-like receptor (CRLR) and receptor-activity-modifying proteins (RAMP). In this study, we found that recombinant ADM had little effect on cAMP/PKA in quiescent human pulmonary fibroblasts, whereas it induced a profound activation of cAMP/PKA signaling in differentiated (by TGF-ß) myofibroblasts. In contrast, the prostacyclin agonist iloprost was equally effective at activating PKA in both quiescent fibroblasts and differentiated myofibroblasts. TGF-ß stimulated a profound expression of CRLR with a time course that mirrored the increased PKA responses to ADM. The TGF-ß receptor kinase inhibitor SB431542 abolished expression of CRLR and attenuated the PKA responses of cells to ADM but not to iloprost. CRLR expression was also dramatically increased in lungs from bleomycin-treated mice. Functionally, ADM did not affect initial differentiation of quiescent fibroblasts in response to TGF-ß but significantly attenuated the expression of SMA, collagen-1, and fibronectin in pre-differentiated myofibroblasts, which was accompanied by decreased contractility of myofibroblasts. Finally, sensitization of ADM signaling by transgenic overexpression of RAMP2 in myofibroblasts resulted in enhanced survival and reduced pulmonary fibrosis in the bleomycin model of the disease. In conclusion, differentiated pulmonary myofibroblasts gain responsiveness to ADM via increased CRLR expression, suggesting the possibility of using ADM for targeting pathological myofibroblasts without affecting normal fibroblasts.


Asunto(s)
Adrenomedulina/farmacología , Diferenciación Celular/efectos de los fármacos , Miofibroblastos/citología , Fibrosis Pulmonar/fisiopatología , Actinas/metabolismo , Adrenomedulina/uso terapéutico , Animales , Bleomicina , Proteína Similar al Receptor de Calcitonina/biosíntesis , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Humanos , Iloprost/farmacología , Ratones , Miofibroblastos/efectos de los fármacos , Miofibroblastos/fisiología , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/tratamiento farmacológico , Proteína 2 Modificadora de la Actividad de Receptores/genética , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta/farmacología
15.
Transl Res ; 161(6): 495-504, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23305708

RESUMEN

Excessive concentrations of oxidized phospholipids (OxPL), the products of 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphatidylcholine (PAPC) oxidation have been detected in atherosclerosis, septic inflammation, and acute lung injury (ALI) and have been shown to induce vascular barrier dysfunction. In contrast, oxidized PAPC (OxPAPC) at low concentrations exhibit potent barrier protective effects. The nature of such biphasic effects remains unclear. We tested the hypothesis that barrier-disruptive effects of high OxPAPC doses on endothelial cell (EC) monolayer are defined by fragmented products of PAPC oxidation (lysophosphatidyl choline [lyso-PC], 1-palmitoyl-2-(5-oxovaleroyl)-sn-glycero-phosphatidylcholine [POVPC], 1-palmitoyl-2-glutaroyl-sn-glycero-phosphatidylcholine [PGPC]), whereas barrier enhancing effects are mediated by full length oxidated PAPC products and may be reproduced by single compounds contained in the OxPAPC such as 1-palmitoyl-2-(5,6-epoxyisoprostane E2)-sn-glycero-3-phosphatidyl choline (PEIPC). All 3 fragmented OxPAPC products increased EC permeability in a dose-dependent manner, whereas PEIPC decreased it and reversed barrier disruptive effects of lyso-PC, POVPC, and PGPC monitored by measurements of transendothelial electrical resistance. Immunofluorescence staining and western blot analysis showed that PGPC mimicked the cytoskeletal remodeling and tyrosine phosphorylation of adherens junction (AJ) protein vascular endothelial (VE)-cadherin leading to EC barrier dysfunction induced by high OxPAPC concentrations. Barrier-disruptive effects of PGPC were abrogated by reactive oxygen species (ROS) inhibitor, N-acetyl cysteine, or Src kinase inhibitor, PP-2. The results of this study show that barrier disruptive effects of fragmented OxPAPC constituents (lyso-PC, POVPC, PGPC) are balanced by barrier enhancing effects of full length oxygenated products (PEIPC). These data strongly suggest that barrier disruptive effects of OxPAPC at higher concentrations are dictated by predominant effects of fragmented phospholipids such as PGPC, which promote ROS-dependent activation of Src kinase and VE-cadherin phosphorylation at Tyr(658) and Tyr(731) leading to disruption of endothelial cell AJs.


Asunto(s)
Barrera Alveolocapilar/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Fosfatidilcolinas/farmacología , Barrera Alveolocapilar/metabolismo , Permeabilidad Capilar/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Impedancia Eléctrica , Técnicas Electroquímicas , Endotelio Vascular/metabolismo , Humanos , Oxidación-Reducción
16.
Microvasc Res ; 87: 50-7, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23296034

RESUMEN

Circulating barrier disruptive agonists bind specific cell membrane receptors and trigger signal transduction pathways leading to the activation of cell contractility and endothelial cell (EC) permeability. Although all cells in tissues including vascular EC are surrounded by compliant extracellular matrix, the impact of matrix stiffness on agonist-induced signaling, cytoskeletal remodeling and EC barrier regulation is not well understood. This study examined agonist-induced cytoskeletal and signaling changes associated with EC barrier disruption and recovery using pulmonary EC grown on compliant substrates of physiologically relevant (8.6 kPa) stiffness, very low (0.55 kPa) and very high (42 kPa) stiffness. Human pulmonary microvascular and macrovascular EC grown on 0.55 kPa substrate contained a few actin stress fibers, while stress fiber amount increased with increasing matrix stiffness. Thrombin-induced stress fiber formation was maximal in EC grown on 42 kPa substrate, diminished on 8.6 kPa substrate, and was minimal on 0.55 kPa substrate. These effects were linked to a stiffness-dependent increase in thrombin-induced phosphorylation of the Rho kinase target, myosin light chain phosphatase (MYPT1), and regulatory myosin light chains (MLC). Surprisingly, EC barrier recovery and activation of Rac GTPase-dependent barrier protective signaling reached maximal levels in EC grown on 8.6 kPa, but not on 0.55 kPa substrate. In conclusion, these data show a critical role of extracellular matrix stiffness in the regulation of the Rac/Rho signaling balance during onset and resolution of agonist-induced EC permeability. The optimal conditions for the Rho/Rac signaling switch, which provides an effective and reversible EC cytoskeletal and permeability response to agonist, are reached in cells grown on the matrix of physiologically relevant stiffness.


Asunto(s)
Resinas Acrílicas/química , Permeabilidad Capilar , Células Endoteliales/metabolismo , Pulmón/irrigación sanguínea , Permeabilidad Capilar/efectos de los fármacos , Técnicas de Cultivo de Célula , Células Cultivadas , Elasticidad , Células Endoteliales/efectos de los fármacos , Humanos , Hidrogeles , Cadenas Ligeras de Miosina/metabolismo , Fosfatasa de Miosina de Cadena Ligera/metabolismo , Fosforilación , Transducción de Señal , Fibras de Estrés/metabolismo , Trombina/farmacología , Factores de Tiempo , Proteínas de Unión al GTP rac/metabolismo , Proteínas de Unión al GTP rho/metabolismo
17.
Eur Respir J ; 41(1): 165-76, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22790920

RESUMEN

The protective effects of prostacyclin and its stable analogue iloprost are mediated by elevation of intracellular cyclic AMP (cAMP) leading to enhancement of the peripheral actin cytoskeleton and cell-cell adhesive structures. This study tested the hypothesis that iloprost may exhibit protective effects against lung injury and endothelial barrier dysfunction induced by bacterial wall lipopolysaccharide (LPS). Endothelial barrier dysfunction was assessed by measurements of transendothelial permeability, morphologically and by analysis of LPS-activated inflammatory signalling. In vivo, C57BL/6J mice were challenged with LPS with or without iloprost or 8-bromoadenosine-3',5'-cyclic monophosphate (Br-cAMP) treatment. Lung injury was monitored by measurements of bronchoalveolar lavage protein content, cell count and Evans blue extravasation. Iloprost and Br-cAMP attenuated the disruption of the endothelial monolayer, and suppressed the activation of p38 mitogen-activated protein kinase (MAPK), the nuclear factor (NF)-κB pathway, Rho signalling, intercellular adhesion molecular (ICAM)-1 expression and neutrophil migration after LPS challenge. In vivo, iloprost was effective against LPS-induced protein and neutrophil accumulation in bronchoalveolar lavage fluid, and reduced myeloperoxidase activation, ICAM-1 expression and Evans blue extravasation in the lungs. Inhibition of Rac activity abolished the barrier-protective and anti-inflammatory effects of iloprost and Br-cAMP. Iloprost-induced elevation of intracellular cAMP triggers Rac signalling, which attenuates LPS-induced NF-κB and p38 MAPK inflammatory pathways and the Rho-dependent mechanism of endothelial permeability.


Asunto(s)
Iloprost/uso terapéutico , Lesión Pulmonar/tratamiento farmacológico , Pulmón/efectos de los fármacos , Pulmón/fisiopatología , Animales , Células Cultivadas , Endotelio/efectos de los fármacos , Endotelio/fisiología , Lipopolisacáridos/administración & dosificación , Lesión Pulmonar/inducido químicamente , Ratones , Ratones Endogámicos C57BL , Neuropéptidos/efectos de los fármacos , Neuropéptidos/fisiología , Proteínas de Unión al GTP rac/efectos de los fármacos , Proteínas de Unión al GTP rac/fisiología , Proteína de Unión al GTP rac1
18.
FASEB J ; 26(9): 3862-74, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22700873

RESUMEN

Microtubule (MT) dynamics in vascular endothelium are modulated by vasoactive mediators and are critically involved in the control of endothelial cell (EC) permeability via Rho GTPase-dependent crosstalk with the actin cytoskeleton. However, the role of regulators in MT stability in these mechanisms remains unclear. This study investigated the involvement of the MT-associated protein stathmin in the mediation of agonist-induced permeability in EC cultures and vascular leak in vivo. Thrombin treatment of human pulmonary ECs induced rapid dephosphorylation and activation of stathmin. Inhibition of stathmin activity by small interfering RNA-based knockdown or cAMP-mediated phosphorylation abrogated thrombin-induced F-actin remodeling and Rho-dependent EC hyperpermeability, while expression of a phosphorylation-deficient stathmin mutant exacerbated thrombin-induced EC barrier disruption. Stathmin suppression preserved the MT network against thrombin-induced MT disassembly and release of Rho-specific guanine nucleotide exchange factor, GEF-H1. The protective effects of stathmin knockdown were observed in vivo in the mouse 2-hit model of ventilator-induced lung injury and were linked to MT stabilization and down-regulation of Rho signaling in the lung. These results demonstrate the mechanism of stathmin-dependent control of MT dynamics, Rho signaling, and permeability and suggest novel potential pharmacological interventions in the prevention of increased vascular leak via modulation of stathmin activity.


Asunto(s)
Permeabilidad de la Membrana Celular , Pulmón/citología , Microtúbulos/fisiología , Estatmina/fisiología , Animales , Endotelio/citología , Endotelio/efectos de los fármacos , Técnica del Anticuerpo Fluorescente , Técnicas de Silenciamiento del Gen , Pulmón/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Fosforilación , ARN Interferente Pequeño/genética , Respiración Artificial/efectos adversos , Estatmina/genética , Estatmina/metabolismo , Trombina/farmacología
19.
FEBS Lett ; 586(16): 2294-9, 2012 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-22728435

RESUMEN

Barrier-protective agonists induce association of focal adhesions (FA) and adherens junctions (AJ) in endothelial cells. Here we identified specific domains of FA protein paxillin interacting with AJ protein and examined regulation of paxillin domain interactions with ß-catenin by Rac GTPase. Co-expression of paxillin LD-1,2; LD-3,4; LIM-1,2; and LIM-3,4 domains with ß-catenin showed exclusive interaction of LIM-1,2 and LIM-3,4 with ß-catenin, which was enhanced by agonist-induced Rac activation or expression of activated Rac mutant. These results demonstrate a novel function of paxillin LIM domains in targeting ß-catenin in a Rac-dependent manner, which may play a role in Rac-dependent control of FA-AJ interactions and monolayer integrity.


Asunto(s)
Regulación de la Expresión Génica , Paxillin/química , beta Catenina/química , Uniones Adherentes/química , Sitio Alostérico , Células Endoteliales/citología , Escherichia coli/metabolismo , Adhesiones Focales/química , Glutatión Transferasa/metabolismo , Células HEK293 , Humanos , Plásmidos/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-akt/química , Proteínas Recombinantes/química , Transducción de Señal
20.
J Cell Physiol ; 227(10): 3405-16, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22213015

RESUMEN

Small GTPase Rac is important regulator of endothelial cell (EC) barrier enhancement by prostacyclin characterized by increased peripheral actin cytoskeleton and increased interactions between VE-cadherin and other adherens junction (AJ) proteins. This study utilized complementary approaches including siRNA knockdown, culturing in Ca(2+) -free medium, and VE-cadherin blocking antibody to alter VE-cadherin extracellular interactions to investigate the role of VE-cadherin outside-in signaling in modulation of Rac activation and EC barrier regulation by prostacyclin analog iloprost. Spatial analysis of Rac activation in pulmonary EC by FRET revealed additional spike in iloprost-induced Rac activity at the sites of newly formed cell-cell junctions. In contrast, disruption of VE-cadherin extracellular trans-interactions suppressed iloprost-activated Rac signaling and attenuated EC barrier enhancement and cytoskeletal remodeling. These inhibitory effects were associated with decreased membrane accumulation and activation of Rac-specific guanine nucleotide exchange factors (GEFs) Tiam1 and Vav2. Conversely, plating of pulmonary EC on surfaces coated with extracellular VE-cadherin domain further promoted iloprost-induced Rac signaling. In the model of thrombin-induced EC barrier recovery, blocking of VE-cadherin trans-interactions attenuated activation of Rac pathway during recovery phase and delayed suppression of Rho signaling and restoration of EC barrier properties. These results suggest that VE-cadherin outside-in signaling controls locally Rac activity stimulated by barrier protective agonists. This control is essential for maximal EC barrier enhancement and accelerated barrier recovery.


Asunto(s)
Antígenos CD/metabolismo , Cadherinas/metabolismo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Iloprost/farmacología , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Uniones Adherentes/efectos de los fármacos , Uniones Adherentes/metabolismo , Anticuerpos/farmacología , Cadherinas/antagonistas & inhibidores , Permeabilidad Capilar/efectos de los fármacos , Células Cultivadas , Citoesqueleto/efectos de los fármacos , Citoesqueleto/metabolismo , Células Endoteliales/citología , Epoprostenol/farmacología , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Uniones Intercelulares/efectos de los fármacos , Uniones Intercelulares/metabolismo , Pulmón/citología , Proteínas Proto-Oncogénicas c-vav/metabolismo , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Proteína 1 de Invasión e Inducción de Metástasis del Linfoma-T
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...