Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Redox Biol ; 38: 101794, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33248422

RESUMO

Acute lung injury (ALI), a devastating illness induced by systemic inflammation e.g., sepsis or local lung inflammation e.g., COVID-19 mediated severe pneumonia, has an unacceptably high mortality and has no effective therapy. ALI is associated with increased pulmonary microvascular hyperpermeability and alveolar flooding. The small Rho GTPases, RhoA and Rac1 are central regulators of vascular permeability through cytoskeleton rearrangements. RhoA and Rac1 have opposing functional outcome: RhoA induces an endothelial contractile phenotype and barrier disruption, while Rac1 stabilizes endothelial junctions and increases barrier integrity. In ALI, RhoA activity is increased while Rac1 activity is reduced. We have shown that the activation of RhoA in lipopolysaccharide (LPS)-mediated ALI, is dependent, at least in part, on a single nitration event at tyrosine (Y)34. Thus, the purpose of this study was to determine if the inhibition of Rac1 is also dependent on its nitration. Our data show that Rac1 inhibition by LPS is associated with its nitration that mass spectrometry identified as Y32, within the switch I region adjacent to the nucleotide-binding site. Using a molecular modeling approach, we designed a nitration shielding peptide for Rac1, designated NipR2 (nitration inhibitor peptide for the Rho GTPases 2), which attenuated the LPS-induced nitration of Rac1 at Y32, preserves Rac1 activity and attenuates the LPS-mediated disruption of the endothelial barrier in human lung microvascular endothelial cells (HLMVEC). Using a murine model of ALI induced by intratracheal installation of LPS we found that NipR2 successfully prevented Rac1 nitration and Rac1 inhibition, and more importantly attenuated pulmonary inflammation, reduced lung injury and prevented the loss of lung function. Together, our data identify a new post-translational mechanism of Rac1 inhibition through its nitration at Y32. As NipR2 also reduces sepsis induced ALI in the mouse lung, we conclude that Rac1 nitration is a therapeutic target in ALI.


Assuntos
Lesão Pulmonar Aguda , Barreira Alveolocapilar , COVID-19 , Células Endoteliais , Lipopolissacarídeos/toxicidade , Neuropeptídeos/metabolismo , SARS-CoV-2/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/enzimologia , Lesão Pulmonar Aguda/patologia , Lesão Pulmonar Aguda/virologia , Animais , Barreira Alveolocapilar/enzimologia , Barreira Alveolocapilar/patologia , Barreira Alveolocapilar/virologia , COVID-19/induzido quimicamente , COVID-19/enzimologia , COVID-19/patologia , Linhagem Celular , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Células Endoteliais/virologia , Humanos , Masculino , Camundongos , Neuropeptídeos/genética , Proteínas rac1 de Ligação ao GTP/genética
2.
Am J Physiol Lung Cell Mol Physiol ; 315(1): L66-L77, 2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-29597831

RESUMO

Compromised pulmonary endothelial cell (PEC) barrier function characterizes acute respiratory distress syndrome (ARDS), a cause of substantial morbidity and mortality. Survival from ARDS is greater in children compared with adults. Whether developmental differences intrinsic to PEC barrier function contribute to this survival advantage remains unknown. To test the hypothesis that PEC barrier function is more well-preserved in neonatal lungs compared with adult lungs in response to inflammation, we induced lung injury in neonatal and adult mice with systemic lipopolysaccharide (LPS). We assessed PEC barrier function in vivo and in vitro, evaluated changes in the expression of focal adhesion kinase 1 (FAK1) and phosphorylation in response to LPS, and determined the effect of FAK silencing and overexpression on PEC barrier function. We found that LPS induced a greater increase in lung permeability and PEC barrier disruption in the adult mice, despite similar degrees of inflammation and apoptosis. Although baseline expression was similar, LPS increased FAK1 expression in neonatal PEC but increased FAK1 phosphorylation and decreased FAK1 expression in adult PEC. Pharmacologic inhibition of FAK1 accentuated LPS-induced barrier disruption most in adult PEC. Finally, in response to LPS, FAK silencing markedly impaired neonatal PEC barrier function, whereas FAK overexpression preserved adult PEC barrier function. Thus, developmental differences in FAK expression during inflammatory injury serve to preserve neonatal pulmonary endothelial barrier function compared with that of adults and suggest that intrinsic differences in the immature versus pulmonary endothelium, especially relative to FAK1 phosphorylation, may contribute to the improved outcomes of children with ARDS.


Assuntos
Apoptose , Barreira Alveolocapilar/enzimologia , Células Endoteliais/enzimologia , Endotélio/enzimologia , Quinase 1 de Adesão Focal/metabolismo , Transdução de Sinais , Animais , Barreira Alveolocapilar/crescimento & desenvolvimento , Barreira Alveolocapilar/patologia , Células Endoteliais/patologia , Endotélio/patologia , Quinase 1 de Adesão Focal/antagonistas & inibidores , Inflamação/induzido quimicamente , Inflamação/enzimologia , Inflamação/patologia , Lipopolissacarídeos/toxicidade , Camundongos
3.
Respir Res ; 17(1): 71, 2016 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-27301375

RESUMO

BACKGROUND: Gastric contents aspiration in humans is a risk factor for severe respiratory failure with elevated mortality. Although aspiration-induced local lung inflammation has been studied in animal models, little is known about extrapulmonary effects of aspiration. We investigated whether a single orotracheal instillation of whole gastric fluid elicits a liver acute phase response and if this response contributes to enrich the alveolar spaces with proteins having antiprotease activity. METHODS: In anesthetized Sprague-Dawley rats receiving whole gastric fluid, we studied at different times after instillation (4 h -7 days): changes in blood cytokines and acute phase proteins (fibrinogen and the antiproteases alpha1-antitrypsin and alpha2-macroglobulin) as well as liver mRNA expression of the two antiproteases. The impact of the systemic changes on lung antiprotease defense was evaluated by measuring levels and bioactivity of antiproteases in broncho-alveolar lavage fluid (BALF). Markers of alveolar-capillary barrier derangement were also studied. Non-parametric ANOVA (Kruskall-Wallis) and linear regression analysis were used. RESULTS: Severe peribronchiolar injury involving edema, intra-alveolar proteinaceous debris, hemorrhage and PMNn cell infiltration was seen in the first 24 h and later resolved. Despite a large increase in several lung cytokines, only IL-6 was found elevated in blood, preceding increased liver expression and blood concentration of both antiproteases. These changes, with an acute phase response profile, were significantly larger for alpha2-macroglobulin (40-fold increment in expression with 12-fold elevation in blood protein concentration) than for alpha1-antitrypsin (2-3 fold increment in expression with 0.5-fold elevation in blood protein concentration). Both the increment in capillary-alveolar antiprotease concentration gradient due to increased antiprotease liver synthesis and a timely-associated derangement of the alveolar-capillary barrier induced by aspiration, contributed a 58-fold and a 190-fold increase in BALF alpha1-antitrypsin and alpha2-macroglobulin levels respectively (p < 0.001). CONCLUSIONS: Gastric contents-induced acute lung injury elicits a liver acute phase response characterized by increased mRNA expression of antiproteases and elevation of blood antiprotease concentrations. Hepatic changes act in concert with derangement of the alveolar capillary barrier to enrich alveolar spaces with antiproteases. These findings may have significant implications decreasing protease burden, limiting injury in this and other models of acute lung injury and likely, in recurrent aspiration.


Assuntos
Lesão Pulmonar Aguda/enzimologia , Reação de Fase Aguda/enzimologia , Fígado/metabolismo , alfa 2-Macroglobulinas Associadas à Gravidez/biossíntese , Alvéolos Pulmonares/enzimologia , Aspiração Respiratória de Conteúdos Gástricos/complicações , alfa 1-Antitripsina/biossíntese , Lesão Pulmonar Aguda/sangue , Lesão Pulmonar Aguda/etiologia , Lesão Pulmonar Aguda/patologia , Reação de Fase Aguda/sangue , Reação de Fase Aguda/etiologia , Reação de Fase Aguda/patologia , Animais , Barreira Alveolocapilar/enzimologia , Barreira Alveolocapilar/patologia , Modelos Animais de Doenças , Indução Enzimática , Mediadores da Inflamação/sangue , Interleucina-6/sangue , Masculino , alfa 2-Macroglobulinas Associadas à Gravidez/genética , Alvéolos Pulmonares/patologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ratos Sprague-Dawley , Fatores de Tempo , alfa 1-Antitripsina/sangue , alfa 1-Antitripsina/genética
4.
Am J Physiol Lung Cell Mol Physiol ; 305(2): L185-92, 2013 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-23686854

RESUMO

It is becoming increasingly apparent that cAMP signals within the pulmonary endothelium are highly compartmentalized, and this compartmentalization is critical to maintaining endothelial barrier integrity. Studies demonstrate that the exogenous soluble bacterial toxin, ExoY, and heterologous expression of the forskolin-stimulated soluble mammalian adenylyl cyclase (AC) chimera, sACI/II, elevate cytosolic cAMP and disrupt the pulmonary microvascular endothelial barrier. The barrier-disruptive effects of cytosolic cAMP generated by exogenous soluble ACs are in contrast to the barrier-protective effects of subplasma membrane cAMP generated by transmembrane AC, which strengthens endothelial barrier integrity. Endogenous soluble AC isoform 10 (AC10 or commonly known as sAC) lacks transmembrane domains and localizes within the cytosolic compartment. AC10 is uniquely activated by bicarbonate to generate cytosolic cAMP, yet its role in regulation of endothelial barrier integrity has not been addressed. Here we demonstrate that, within the pulmonary circulation, AC10 is expressed in pulmonary microvascular endothelial cells (PMVECs) and pulmonary artery endothelial cells (PAECs), yet expression in PAECs is lower. Furthermore, pulmonary endothelial cells selectively express bicarbonate cotransporters. While extracellular bicarbonate generates a phosphodiesterase 4-sensitive cAMP pool in PMVECs, no such cAMP response is detected in PAECs. Finally, addition of extracellular bicarbonate decreases resistance across the PMVEC monolayer and increases the filtration coefficient in the isolated perfused lung above osmolality controls. Collectively, these findings suggest that PMVECs have a bicarbonate-sensitive cytosolic cAMP pool that disrupts endothelial barrier integrity. These studies could provide an alternative mechanism for the controversial effects of bicarbonate correction of acidosis of acute respiratory distress syndrome patients.


Assuntos
Adenilil Ciclases/biossíntese , Bicarbonatos/metabolismo , Barreira Alveolocapilar/enzimologia , Endotélio/enzimologia , Regulação Enzimológica da Expressão Gênica , Acidose/enzimologia , Animais , Barreira Alveolocapilar/patologia , Células Cultivadas , AMP Cíclico/metabolismo , Endotélio/patologia , Humanos , Ratos , Síndrome do Desconforto Respiratório/enzimologia
5.
Am J Respir Cell Mol Biol ; 48(4): 477-88, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23306835

RESUMO

Earlier studies indicated a role for reactive oxygen species (ROS) in host defense against Pseudomonas aeruginosa infection. However, the role of nicotinamide adenine dinucleotide phosphate-reduced (NADPH) oxidase (NOX) proteins and the mechanism of activation for NADPH oxidase in P. aeruginosa infection are not well-defined. Here, we investigated the role of NOX2 and NOX4 proteins in P. aeruginosa infection, ROS generation, and endothelial barrier function in murine lungs and in human lung microvascular endothelial cells (HLMVECs). Airway instillation of P. aeruginosa strain 103 (PA103) significantly increased ROS concentrations in bronchial alveolar lavage (BAL) fluid, along with the expression of NOX2 and NOX4, but not NOX1 and NOX3, in lung tissue. In addition, PA103-infected HLMVECs revealed elevated concentrations of ROS, NOX2, and NOX4. In murine lungs and HLMVECs, PA103 induced the NF-κB pathway, and its inhibition blocked PA103-dependent NOX2 and NOX4 expression. Barrier function analysis showed that heat-killed PA103 induced endothelial permeability in a dose-dependent manner, which was attenuated by treatment with small interfering (si)RNA specific for NOX4, but not NOX2. Furthermore, the knockdown of NOX4, but not NOX2, with siRNA reduced PA103-mediated apoptosis in HLMVECs. In vivo, the down-regulation of NOX4 with NOX4 siRNA attenuated PA103-induced lung vascular permeability. The deletion of NOX2 in mice exerted no effect on permeability, but offered significant resistance to P. aeruginosa-induced lung inflammation. These data show that P. aeruginosa lung infection up-regulates NOX2 and NOX4 expression and ROS generation, which play distinct roles in regulating lung inflammation, apoptosis, and permeability.


Assuntos
Permeabilidade Capilar , Glicoproteínas de Membrana/biossíntese , NADPH Oxidases/biossíntese , NADP/metabolismo , Pneumonia Bacteriana/enzimologia , Infecções por Pseudomonas/enzimologia , Pseudomonas aeruginosa , Animais , Barreira Alveolocapilar/enzimologia , Barreira Alveolocapilar/patologia , Células Cultivadas , Regulação Enzimológica da Expressão Gênica/genética , Humanos , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Knockout , NADP/genética , NADPH Oxidase 2 , NADPH Oxidase 4 , NADPH Oxidases/genética , Pneumonia Bacteriana/genética , Pneumonia Bacteriana/patologia , Infecções por Pseudomonas/genética , Infecções por Pseudomonas/patologia , Espécies Reativas de Oxigênio/metabolismo
6.
Am J Physiol Lung Cell Mol Physiol ; 303(10): L880-8, 2012 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-22983354

RESUMO

The pathogenesis of acute lung injury and acute respiratory distress syndrome is characterized by sequestration of leukocytes in lung tissue, disruption of capillary integrity, and pulmonary edema. PKCδ plays a critical role in RhoA-mediated endothelial barrier function and inflammatory responses. We used mice with genetic deletion of PKCδ (PKCδ(-/-)) to assess the role of PKCδ in susceptibility to LPS-induced lung injury and pulmonary edema. Under baseline conditions or in settings of increased capillary hydrostatic pressures, no differences were noted in the filtration coefficients (k(f)) or wet-to-dry weight ratios between PKCδ(+/+) and PKCδ(-/-) mice. However, at 24 h after exposure to LPS, the k(f) values were significantly higher in lungs isolated from PKCδ(+/+) than PKCδ(-/-) mice. In addition, bronchoalveolar lavage fluid obtained from LPS-exposed PKCδ(+/+) mice displayed increased protein and cell content compared with LPS-exposed PKCδ(-/-) mice, but similar changes in inflammatory cytokines were measured. Histology indicated elevated LPS-induced cellularity and inflammation within PKCδ(+/+) mouse lung parenchyma relative to PKCδ(-/-) mouse lungs. Transient overexpression of catalytically inactive PKCδ cDNA in the endothelium significantly attenuated LPS-induced endothelial barrier dysfunction in vitro and increased k(f) lung values in PKCδ(+/+) mice. However, transient overexpression of wild-type PKCδ cDNA in PKCδ(-/-) mouse lung vasculature did not alter the protective effects of PKCδ deficiency against LPS-induced acute lung injury. We conclude that PKCδ plays a role in the pathological progression of endotoxin-induced lung injury, likely mediated through modulation of inflammatory signaling and pulmonary vascular barrier function.


Assuntos
Lesão Pulmonar Aguda/enzimologia , Barreira Alveolocapilar/enzimologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Lipopolissacarídeos/toxicidade , Proteína Quinase C-delta/biossíntese , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/genética , Lesão Pulmonar Aguda/patologia , Animais , Barreira Alveolocapilar/patologia , Citocinas/genética , Citocinas/metabolismo , Mediadores da Inflamação/metabolismo , Camundongos , Camundongos Knockout , Proteína Quinase C-delta/genética , Edema Pulmonar/induzido quimicamente , Edema Pulmonar/enzimologia , Edema Pulmonar/genética , Edema Pulmonar/patologia , Síndrome do Desconforto Respiratório
7.
Am J Physiol Lung Cell Mol Physiol ; 298(3): L361-70, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20023173

RESUMO

Pulmonary edema is mediated in part by disruption of interendothelial cell contacts. Protein tyrosine phosphatases (PTP) have been shown to affect both cell-extracellular matrix and cell-cell junctions. The SH2 domain-containing nonreceptor PTP, SHP2, is involved in intercellular signaling through direct interaction with adherens junction proteins. In this study, we examined the role of SHP2 in pulmonary endothelial barrier function. Inhibition of SHP2 promoted edema formation in rat lungs and increased monolayer permeability in cultured lung endothelial cells. In addition, pulmonary endothelial cells demonstrated a decreased level of p190RhoGAP activity following inhibition of SHP2, events that were accompanied by a concomitant increase in RhoA activity. Furthermore, immunofluorescence microscopy confirmed enhanced actin stress fiber formation and diminished interendothelial staining of adherens junction complex-associated proteins upon SHP2 inhibition. Finally, immunoprecipitation and immunoblot analyses demonstrated increased tyrosine phosphorylation of VE-cadherin, beta-catenin, and p190RhoGAP proteins, as well as decreased association between p120-catenin and VE-cadherin proteins. Our findings suggest that SHP2 supports basal pulmonary endothelial barrier function by coordinating the tyrosine phosphorylation profile of VE-cadherin, beta-catenin, and p190RhoGAP and the activity of RhoA, signaling molecules important in adherens junction complex integrity.


Assuntos
Barreira Alveolocapilar/enzimologia , Endotélio/enzimologia , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Junções Aderentes/efeitos dos fármacos , Junções Aderentes/metabolismo , Animais , Antígenos CD/metabolismo , Biocatálise/efeitos dos fármacos , Barreira Alveolocapilar/efeitos dos fármacos , Barreira Alveolocapilar/patologia , Caderinas/metabolismo , Cateninas/metabolismo , Bovinos , Endotélio/efeitos dos fármacos , Endotélio/patologia , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Fosfotirosina/metabolismo , Ligação Proteica/efeitos dos fármacos , Proteína Tirosina Fosfatase não Receptora Tipo 11/antagonistas & inibidores , Edema Pulmonar/enzimologia , Edema Pulmonar/patologia , Ratos , Proteínas Repressoras/metabolismo , Fibras de Estresse/efeitos dos fármacos , Fibras de Estresse/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , delta Catenina
8.
Am J Physiol Lung Cell Mol Physiol ; 295(2): L293-302, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18539681

RESUMO

As part of the innate immune defense, the polarized conducting lung epithelium acts as a barrier to keep particulates carried in respiration from underlying tissue. Arsenic is a metalloid toxicant that can affect the lung via inhalation or ingestion. We have recently shown that chronic exposure of mice or humans to arsenic (10-50 ppb) in drinking water alters bronchiolar lavage or sputum proteins consistent with reduced epithelial cell migration and wound repair in the airway. In this report, we used an in vitro model to examine effects of acute exposure of arsenic (15-290 ppb) on conducting airway lung epithelium. We found that arsenic at concentrations as low as 30 ppb inhibits reformation of the epithelial monolayer following scrape wounds of monolayer cultures. In an effort to understand functional contributions to epithelial wound repair altered by arsenic, we showed that acute arsenic exposure increases activity and expression of matrix metalloproteinase (MMP)-9, an important protease in lung function. Furthermore, inhibition of MMP-9 in arsenic-treated cells improved wound repair. We propose that arsenic in the airway can alter the airway epithelial barrier by restricting proper wound repair in part through the upregulation of MMP-9 by lung epithelial cells.


Assuntos
Arsênio/toxicidade , Células Epiteliais/enzimologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Metaloproteinase 9 da Matriz/biossíntese , Mucosa Respiratória/enzimologia , Poluentes Químicos da Água/toxicidade , Cicatrização/efeitos dos fármacos , Animais , Barreira Alveolocapilar/enzimologia , Barreira Alveolocapilar/patologia , Linhagem Celular Transformada , Relação Dose-Resposta a Droga , Exposição Ambiental/efeitos adversos , Células Epiteliais/patologia , Humanos , Imunidade Inata/efeitos dos fármacos , Camundongos , Consumo de Oxigênio/efeitos dos fármacos , Mucosa Respiratória/patologia , Regulação para Cima/efeitos dos fármacos
9.
Circ Res ; 101(1): 50-8, 2007 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-17525371

RESUMO

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.


Assuntos
Barreira Alveolocapilar/enzimologia , Permeabilidade Capilar , Endotélio Vascular/citologia , Endotélio Vascular/enzimologia , Inibidores de Dissociação do Nucleotídeo Guanina/fisiologia , Pulmão/enzimologia , Proteínas rho de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Animais , Barreira Alveolocapilar/fisiopatologia , Permeabilidade Capilar/genética , Células Endoteliais/enzimologia , Células Endoteliais/fisiologia , Endotélio Vascular/fisiologia , Inibidores de Dissociação do Nucleotídeo Guanina/deficiência , Inibidores de Dissociação do Nucleotídeo Guanina/genética , Pulmão/patologia , Pulmão/fisiopatologia , Pneumopatias/enzimologia , Pneumopatias/genética , Pneumopatias/fisiopatologia , Camundongos , Camundongos Knockout , Inibidores da Dissociação do Nucleotídeo Guanina rho-Específico
10.
Am J Physiol Lung Cell Mol Physiol ; 292(2): L487-99, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17012370

RESUMO

2-Methoxyestradiol (2ME), a promising anti-tumor agent, is currently tested in phase I/II clinical trial to assess drug tolerance and clinical effects. 2ME is known to affect microtubule (MT) polymerization rather than act through estrogen receptors. We hypothesized that 2ME, similar to other MT inhibitors, disrupts endothelial barrier properties. We show that 2ME decreases transendothelial electrical resistance and increases FITC-dextran leakage across human pulmonary artery endothelial monolayer, which correlates with 2ME-induced MT depolymerization. Pretreatment of endothelium with MT stabilizer taxol significantly attenuates the decrease in transendothelial resistance. 2ME treatment results in the induction of F-actin stress fibers, accompanied by the increase in myosin light chain (MLC) phosphorylation. The experiments with Rho kinase (ROCK) and MLC kinase inhibitors and ROCK small interfering RNA (siRNA) revealed that increase in MLC phosphorylation is attributed to the ROCK activation rather than MLC kinase activation. 2ME induces significant ERK1/2, p38, and JNK phosphorylation and activation; however, only p38 activation is relevant to the 2ME-induced endothelial hyperpermeability. p38 activation is accompanied by a marked increase in MAPKAP2 and 27-kDa heat shock protein (HSP27) phosphorylation level. Taxol significantly decreases p38 phosphorylation and activation in response to 2ME stimulation. Vice versa, p38 inhibitor SB203580 attenuates MT rearrangement in 2ME-challenged cells. Together, these results indicate that 2ME-induced barrier disruption is governed by MT depolymerization and p38- and ROCK-dependent mechanisms. The fact that certain concentrations of 2ME induce endothelial hyperpermeability suggests that the issue of the maximum-tolerated dose of 2ME for cancer treatment should be addressed with caution.


Assuntos
Barreira Alveolocapilar/enzimologia , Barreira Alveolocapilar/fisiopatologia , Estradiol/análogos & derivados , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Microtúbulos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , 2-Metoxiestradiol , Actomiosina/metabolismo , Amidas/farmacologia , Barreira Alveolocapilar/efeitos dos fármacos , Permeabilidade da Membrana Celular/efeitos dos fármacos , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/enzimologia , Estradiol/farmacologia , Humanos , Imidazóis/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/deficiência , Microeletrodos , Modelos Biológicos , Quinase de Cadeia Leve de Miosina/metabolismo , Paclitaxel/farmacologia , Fosforilação/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/deficiência , Artéria Pulmonar/citologia , Artéria Pulmonar/efeitos dos fármacos , Artéria Pulmonar/enzimologia , Piridinas/farmacologia , Tubulina (Proteína)/metabolismo , Quinases Associadas a rho
11.
J Cell Biochem ; 98(4): 931-53, 2006 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-16475161

RESUMO

Our recently published data suggested the involvement of protein phosphatase 2A (PP2A) in endothelial cell (EC) barrier regulation (Tar et al. [2004] J Cell Biochem 92:534-546). In order to further elucidate the role of PP2A in the regulation of EC cytoskeleton and permeability, PP2A catalytic (PP2Ac) and A regulatory (PP2Aa) subunits were cloned and human pulmonary arterial EC (HPAEC) were transfected with PP2A mammalian expression constructs or infected with PP2A recombinant adenoviruses. Immunostaining of PP2Ac or of PP2Aa + c overexpressing HPAEC indicated actin cytoskeleton rearrangement. PP2A overexpression hindered or at least dramatically reduced thrombin- or nocodazole-induced F-actin stress fiber formation and microtubule (MT) dissolution. Accordingly, it also attenuated thrombin- or nocodazole-induced decrease in transendothelial electrical resistance indicative of barrier protection. Inhibition of PP2A by okadaic acid abolished its effect on agonist-induced changes in EC cytoskeleton; this indicates a critical role of PP2A activity in EC cytoskeletal maintenance. The overexpression of PP2A significantly attenuated thrombin- or nocodazole-induced phosphorylation of HSP27 and tau, two cytoskeletal proteins, which potentially could be involved in agonist-induced cytoskeletal rearrangement and in the increase of permeability. PP2A-mediated dephosphorylation of HSP27 and tau correlated with PP2A-induced preservation of EC cytoskeleton and barrier maintenance. Collectively, our observations clearly demonstrate the crucial role of PP2A in EC barrier protection.


Assuntos
Barreira Alveolocapilar/enzimologia , Proteínas do Citoesqueleto/metabolismo , Citoesqueleto/metabolismo , Células Endoteliais/enzimologia , Fosfoproteínas Fosfatases/metabolismo , Artéria Pulmonar/enzimologia , Barreira Alveolocapilar/citologia , Células Cultivadas , Células Endoteliais/citologia , Expressão Gênica , Humanos , Fosfoproteínas Fosfatases/genética , Fosforilação , Proteína Fosfatase 2 , Processamento de Proteína Pós-Traducional/fisiologia , Artéria Pulmonar/citologia , Transfecção
12.
J Physiol Pharmacol ; 56 Suppl 4: 47-64, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16204776

RESUMO

NOS-2-derived NO is involved in hypotension, vasoplegia, metabolic disorders and lung injury in endotoxic shock. On the other hand, NOS-3-derived NO protects against LPS-induced lung injury. We have previously shown that NO limits lung injury in the isolated blood-perfused rat lung. Here we characterize the ultrastructure of microvascular lung injury induced by LPS in the absence of endogenous NO and summarize our data on the mechanisms of immediate lung response to LPS in the presence and absence of endogenous NO. Injection of LPS (from E.Coli, 300 microg/ml) into the isolated blood-perfused rat lung induced an immediate transient constriction of airways and vessels that was not associated with lung edema and pulmonary microcirculation injury. In contrast, in the presence of the NOS inhibitor L-NAME (300 microg/ml), LPS produced an enhanced constriction of airways and vessels, which was accompanied by profound lung edema and capillary-alveolar barrier injury, as evidenced by optic and electron microscopy. Microvascular lung injury was confirmed by the following findings: edema of pulmonary endothelium with low electronic density of endothelial cytoplasm, presence of protein-rich fluid and numerous erythrocytes in alveolar space, concentric figures of damaged tubular myelin of surfactant (myelin-like bodies), edema of epithelium type I cells with low electronic density of their cytoplasm and alterations in ultrastructure of basal membrane of vascular-alveolar barrier. Interestingly, epithelial type II cells did not show signs of injury. It is worth noting that capillary-alveolar barrier injury induced by L-NAME+LPS was associated with sequestration of platelets and neutrophils in pulmonary microcirculation and internalization of LPS by neutrophils. In conclusion, in the absence of endogenous nitric oxide LPS induces injury of microvascular endothelium and vascular-alveolar barrier that leads to fatal pulmonary edema. Mechanisms of immediate lung response to LPS in presence of NO and those leading to acute microvascular lung injury in response to LPS in absence of NO are summarized. In our view, immediate lung response to bacterial endotoxin represents a phylogenetically ancient host defence response involving complement-dependent activation of platelets and neutrophils and subsequent production of lipid mediators. This response is designed for a quick elimination of bacterial endotoxin from the circulation and is safeguarded by endothelial NO.


Assuntos
Infecções Bacterianas/patologia , Barreira Alveolocapilar/ultraestrutura , Endotélio Vascular/ultraestrutura , Pulmão/irrigação sanguínea , Óxido Nítrico Sintase/antagonistas & inibidores , Óxido Nítrico/metabolismo , Edema Pulmonar/etiologia , Animais , Infecções Bacterianas/induzido quimicamente , Infecções Bacterianas/complicações , Infecções Bacterianas/metabolismo , Infecções Bacterianas/fisiopatologia , Pressão Sanguínea , Barreira Alveolocapilar/efeitos dos fármacos , Barreira Alveolocapilar/enzimologia , Barreira Alveolocapilar/metabolismo , Barreira Alveolocapilar/fisiopatologia , Modelos Animais de Doenças , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/enzimologia , Endotélio Vascular/metabolismo , Endotélio Vascular/fisiopatologia , Inibidores Enzimáticos/farmacologia , Hipóxia/metabolismo , Hipóxia/patologia , Hipóxia/fisiopatologia , Técnicas In Vitro , Lipopolissacarídeos , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/fisiopatologia , Pulmão/ultraestrutura , Lisina/análogos & derivados , Lisina/farmacologia , Microcirculação/metabolismo , Microcirculação/ultraestrutura , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico Sintase/metabolismo , Tamanho do Órgão , Perfusão , Circulação Pulmonar , Edema Pulmonar/metabolismo , Edema Pulmonar/patologia , Edema Pulmonar/fisiopatologia , Ratos , Ratos Wistar , Volume de Ventilação Pulmonar , Vasoconstrição , Pressão Venosa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA