Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Biol Chem ; 289(8): 4710-22, 2014 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-24398689

RESUMO

Acute lung injury (ALI) is characterized by increased endothelial hyperpermeability. Protein nitration is involved in the endothelial barrier dysfunction in LPS-exposed mice. However, the nitrated proteins involved in this process have not been identified. The activation of the small GTPase RhoA is a critical event in the barrier disruption associated with LPS. Thus, in this study we evaluated the possible role of RhoA nitration in this process. Mass spectroscopy identified a single nitration site, located at Tyr(34) in RhoA. Tyr(34) is located within the switch I region adjacent to the nucleotide-binding site. Utilizing this structure, we developed a peptide designated NipR1 (nitration inhibitory peptide for RhoA 1) to shield Tyr(34) against nitration. TAT-fused NipR1 attenuated RhoA nitration and barrier disruption in LPS-challenged human lung microvascular endothelial cells. Further, treatment of mice with NipR1 attenuated vessel leakage and inflammatory cell infiltration and preserved lung function in a mouse model of ALI. Molecular dynamics simulations suggested that the mechanism by which Tyr(34) nitration stimulates RhoA activity was through a decrease in GDP binding to the protein caused by a conformational change within a region of Switch I, mimicking the conformational shift observed when RhoA is bound to a guanine nucleotide exchange factor. Stopped flow kinetic analysis was used to confirm this prediction. Thus, we have identified a new mechanism of nitration-mediated RhoA activation involved in LPS-mediated endothelial barrier dysfunction and show the potential utility of "shielding" peptides to prevent RhoA nitration in the management of ALI.


Assuntos
Lesão Pulmonar/enzimologia , Lesão Pulmonar/patologia , Proteína rhoA de Ligação ao GTP/metabolismo , Lesão Pulmonar Aguda/enzimologia , Lesão Pulmonar Aguda/patologia , Lesão Pulmonar Aguda/fisiopatologia , Sequência de Aminoácidos , Animais , Líquido da Lavagem Broncoalveolar , Citocinas/metabolismo , Modelos Animais de Doenças , Células Endoteliais/enzimologia , Células Endoteliais/patologia , Ativação Enzimática , Humanos , Lipopolissacarídeos , Lesão Pulmonar/induzido quimicamente , Lesão Pulmonar/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microvasos/patologia , Dados de Sequência Molecular , Óxido Nítrico Sintase Tipo III/metabolismo , Nitrosação , Peptídeos/metabolismo , Substâncias Protetoras/metabolismo , Tirosina/metabolismo , Proteína rhoA de Ligação ao GTP/química
2.
Am J Respir Cell Mol Biol ; 50(1): 170-9, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23972231

RESUMO

Permeability of the endothelial monolayer is increased when exposed to the bacterial endotoxin LPS. Our previous studies have shown that heat shock protein (Hsp) 90 inhibitors protect and restore LPS-mediated hyperpermeability in bovine pulmonary arterial endothelial cells. In this study, we assessed the effect of Hsp90 inhibition against LPS-mediated hyperpermeability in cultured human lung microvascular endothelial cells (HLMVECs) and delineated the underlying molecular mechanisms. We demonstrate that Hsp90 inhibition is critical in the early phase, to prevent LPS-mediated hyperpermeability, and also in the later phase, to restore LPS-mediated hyperpermeability in HLMVECs. Because RhoA is a well known mediator of endothelial hyperpermeability, we investigated the effect of Hsp90 inhibition on LPS-mediated RhoA signaling. RhoA nitration and activity were increased by LPS in HLMVECs and suppressed when pretreated with the Hsp90 inhibitor, 17-allylamino-17 demethoxy-geldanamycin (17-AAG). In addition, inhibition of Rho kinase, a downstream effector of RhoA, protected HLMVECs from LPS-mediated hyperpermeability and abolished LPS-induced myosin light chain (MLC) phosphorylation, a target of Rho kinase. In agreement with these findings, 17-AAG or dominant-negative RhoA attenuated LPS-induced MLC phosphorylation. MLC phosphorylation induced by constitutively active RhoA was also suppressed by 17-AAG, suggesting a role for Hsp90 downstream of RhoA. Inhibition of Src family kinases also suppressed RhoA activity and MLC phosphorylation. Together, these data indicate that Hsp90 inhibition prevents and repairs LPS-induced lung endothelial barrier dysfunction by suppressing Src-mediated RhoA activity and signaling.


Assuntos
Células Endoteliais/efeitos dos fármacos , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Proteínas de Choque Térmico HSP90/metabolismo , Lipopolissacarídeos/efeitos adversos , Quinases Associadas a rho/metabolismo , Animais , Benzoquinonas/farmacologia , Permeabilidade Capilar/efeitos dos fármacos , Células Cultivadas , Células Endoteliais/metabolismo , Humanos , Lactamas Macrocíclicas/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Cadeias Leves de Miosina/metabolismo , Fosforilação/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Proteína rhoA de Ligação ao GTP/metabolismo , Quinases da Família src/metabolismo
3.
Am J Physiol Lung Cell Mol Physiol ; 304(12): L883-93, 2013 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-23585225

RESUMO

Heat shock protein 90 (Hsp90) inhibitors were initially developed as anticancer agents; however, it is becoming increasing clear that they also possess potent anti-inflammatory properties. Posttranslational modifications of Hsp90 have been reported in tumors and have been hypothesized to affect client protein- and inhibitor-binding activities. In the present study we investigated the posttranslational modification of Hsp90 in inflammation. LPS, a prototypical inflammatory agent, induced concentration- and time-dependent tyrosine (Y) phosphorylation of Hsp90α and Hsp90ß in bovine pulmonary arterial and human lung microvascular endothelial cells (HLMVEC). Mass spectrometry identified Y309 as a major site of Y phosphorylation on Hsp90α (Y300 of Hsp90ß). LPS-induced Hsp90 phosphorylation was prevented by the Hsp90 inhibitor 17-allyl-amino-demethoxy-geldanamycin (17-AAG) in vitro as well as in lungs from LPS-treated mice, in vivo. Furthermore, 17-AAG prevented LPS-induced pp60src activation. LPS-induced Hsp90 phosphorylation was also prevented by the pp60src inhibitor PP2. Additionally, Hsp90 phosphorylation was induced by infecting cells with a constitutively active pp60src adenovirus, whereas either a dominant-negative pp60src adenovirus or reduced expression of pp60src by a specific siRNA prevented the LPS-induced Y phosphorylation of Hsp90. Transfection of HLMVEC with the nonphosphorylatable Hsp90ß Y300F mutant prevented LPS-induced Hsp90ß tyrosine phosphorylation but not pp60src activation. Furthermore, the Hsp90ß Y300F mutant showed a reduced ability to bind the Hsp90 client proteins eNOS and pp60src and HLMVEC transfected with the mutant exhibited reduced LPS-induced barrier dysfunction. We conclude that inflammatory stimuli cause posttranslational modifications of Hsp90 that are Hsp90-inhibitor sensitive and may be important to the proinflammatory actions of Hsp90.


Assuntos
Células Endoteliais/efeitos dos fármacos , Proteínas de Choque Térmico HSP90/metabolismo , Lipopolissacarídeos/farmacologia , Pulmão/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Proteínas Proto-Oncogênicas pp60(c-src)/metabolismo , Animais , Benzoquinonas/farmacologia , Bovinos , Células Cultivadas , Células Endoteliais/imunologia , Células Endoteliais/metabolismo , Proteínas de Choque Térmico HSP90/genética , Proteínas de Choque Térmico HSP90/imunologia , Humanos , Lactamas Macrocíclicas/farmacologia , Pulmão/imunologia , Pulmão/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Óxido Nítrico Sintase Tipo III/genética , Óxido Nítrico Sintase Tipo III/imunologia , Óxido Nítrico Sintase Tipo III/metabolismo , Fosforilação , Ligação Proteica/efeitos dos fármacos , Proteínas Proto-Oncogênicas pp60(c-src)/genética , Proteínas Proto-Oncogênicas pp60(c-src)/imunologia , Pirimidinas/farmacologia , RNA Interferente Pequeno/genética , Transdução de Sinais/efeitos dos fármacos , Tirosina/metabolismo
4.
Am J Physiol Cell Physiol ; 302(12): C1786-96, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-22496241

RESUMO

O-linked ß-N-acetylglucosamine (O-GlcNAc) modification of proteins is involved in many important cellular processes. Increased O-GlcNAc has been implicated in major diseases, such as diabetes and its complications and cardiovascular and neurodegenerative diseases. Recently, we reported that O-GlcNAc modification occurs in the proteasome and serves to inhibit proteasome function by blocking the ATPase activity in the 19S regulatory cap, explaining, at least in part, the adverse effects of O-GlcNAc modification and suggesting that downregulating O-GlcNAc might be important in the treatment of human diseases. In this study, we report on a novel mechanism to modulate cellular O-GlcNAc modification, namely through heat shock protein 90 (Hsp90) inhibition. We observed that O-linked ß-N-acetylglucosamine transferase (OGT) interacts with the tetratricopeptide repeat binding site of Hsp90. Inhibition of Hsp90 by its specific inhibitors, radicicol or 17-N-allylamino-17-demethoxygeldanamycin, destabilized OGT in primary endothelial cell cultures and enhanced its degradation by the proteasome. Furthermore, Hsp90 inhibition downregulated O-GlcNAc protein modifications and attenuated the high glucose-induced increase in O-GlcNAc protein modification, including high glucose-induced increase in endothelial or type 3 isoform of nitric oxide synthase (eNOS) O-GlcNAcylation. These results suggest that Hsp90 is involved in the regulation of OGT and O-GlcNAc modification and that Hsp90 inhibitors might be used to modulate O-GlcNAc modification and reverse its adverse effects in human diseases.


Assuntos
Acetilglucosamina/metabolismo , Células Endoteliais/enzimologia , Proteínas de Choque Térmico HSP90/metabolismo , N-Acetilglucosaminiltransferases/metabolismo , Processamento de Proteína Pós-Traducional , Animais , Benzoquinonas/farmacologia , Sítios de Ligação , Bovinos , Células Cultivadas , Células Endoteliais/efeitos dos fármacos , Estabilidade Enzimática , Glucose/metabolismo , Glicosilação , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Meia-Vida , Humanos , Lactamas Macrocíclicas/farmacologia , Macrolídeos/farmacologia , Camundongos , Camundongos Knockout , Óxido Nítrico Sintase Tipo III/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Fatores de Tempo , Ubiquitina-Proteína Ligases/deficiência , Ubiquitina-Proteína Ligases/genética
5.
Vascul Pharmacol ; 52(5-6): 175-81, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20060932

RESUMO

Endothelial barrier dysfunction is an important contributor to the pathogenesis of acute lung injury (ALI) and acute respiratory distress syndrome (ARDS). Even though approaches that target the prevention and repair of endothelial barrier dysfunction are clearly needed, our understanding of the molecular regulation of pulmonary microvascular endothelial permeability remains incomplete. Cultured pulmonary microvascular endothelial cells represent an attractive paradigm for the study of barrier function. Here, we describe a method for the harvest, identification and culture of human lung microvascular endothelial cells (HLMVEC). HLMVEC thus obtained, grow as a monolayer, exhibit contact inhibition and have the typical cobblestone appearance. They express endothelial proteins, such as von Willebrand factor and endothelial nitric oxide synthase and take up an acetylated LDL. Furthermore, HLMVEC respond predictably and with superior sensitivity to the barrier disruptive effects of Gram positive and Gram negative bacterial products, thrombin, vascular endothelial growth factor and microtubule disrupting agents. These HLMVEC present an in-house-derived alternative to commercially available human cells for the study of mechanisms contributing to ALI and ARDS.


Assuntos
Células Endoteliais/metabolismo , Endotélio Vascular/citologia , Pulmão/citologia , Lesão Pulmonar Aguda/fisiopatologia , Técnicas de Cultura de Células/métodos , Células Cultivadas , Endotélio Vascular/metabolismo , Humanos , Microvasos , Óxido Nítrico Sintase Tipo III/metabolismo , Permeabilidade , Síndrome do Desconforto Respiratório/fisiopatologia , Fator de von Willebrand/metabolismo
6.
Vascul Pharmacol ; 52(5-6): 207-13, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20074664

RESUMO

Listeriosis can lead to potentially lethal pulmonary complications in newborns and immune compromised patients, characterized by extensive permeability edema. Listeriolysin (LLO), the main virulence factor of Listeria monocytogenes, induces a dose-dependent hyperpermeability in monolayers of human lung microvascular endothelial cells in vitro. The permeability increasing activity of LLO, which is accompanied by an increased reactive oxygen species (ROS) generation, RhoA activation and myosin light chain (MLC) phosphorylation, can be completely inhibited by the protein kinase C (PKC) alpha/beta inhibitor GO6976, indicating a crucial role for PKC in the induction of barrier dysfunction. The TNF-derived TIP peptide, which mimics the lectin-like domain of the cytokine, blunts LLO-induced hyperpermeability in vitro, upon inhibiting LLO-induced protein kinase C-alpha activation, ROS generation and MLC phosphorylation and upon restoring the RhoA/Rac 1 balance. These results indicate that the lectin-like domain of TNF has a potential therapeutic value in protecting from LLO-induced pulmonary endothelial hyperpermeability.


Assuntos
Toxinas Bacterianas/toxicidade , Endotélio Vascular/metabolismo , Proteínas de Choque Térmico/toxicidade , Proteínas Hemolisinas/toxicidade , Listeria monocytogenes/patogenicidade , Fator de Necrose Tumoral alfa/metabolismo , Animais , Bovinos , Células Cultivadas , Endotélio Vascular/microbiologia , Humanos , Pulmão/citologia , Pulmão/metabolismo , Pulmão/microbiologia , Cadeias Leves de Miosina/metabolismo , Peptídeos/farmacologia , Permeabilidade , Fosforilação , Proteína Quinase C-alfa/antagonistas & inibidores , Artéria Pulmonar/metabolismo , Artéria Pulmonar/microbiologia , Espécies Reativas de Oxigênio/metabolismo , Ovinos , Fator de Necrose Tumoral alfa/química , Proteína rhoA de Ligação ao GTP/metabolismo
7.
Vascul Pharmacol ; 52(5-6): 182-90, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-19962451

RESUMO

Acute lung injury (ALI) is associated with severe alterations in lung structure and function and is characterized by hypoxemia, pulmonary edema, low lung compliance and widespread capillary leakage. Asymmetric dimethylarginine (ADMA), a known cardiovascular risk factor, has been linked to endothelial dysfunction and the pathogenesis of a number of cardiovascular diseases. However, the role of ADMA in the pathogenesis of ALI is less clear. ADMA is metabolized via hydrolytic degradation to l-citrulline and dimethylamine by the enzyme, dimethylarginine dimethylaminohydrolase (DDAH). Recent studies suggest that lipopolysaccharide (LPS) markedly increases the level of ADMA and decreases DDAH activity in endothelial cells. Thus, the purpose of this study was to determine if alterations in the ADMA/DDAH pathway contribute to the development of ALI initiated by LPS-exposure in mice. Our data demonstrate that LPS exposure significantly increases ADMA levels and this correlates with a decrease in DDAH activity but not protein levels of either DDAH I or DDAH II isoforms. Further, we found that the increase in ADMA levels cause an early decrease in nitric oxide (NO(x)) and a significant increase in both NO synthase (NOS)-derived superoxide and total nitrated lung proteins. Finally, we found that decreasing peroxynitrite levels with either uric acid or Manganese (III) tetrakis (1-methyl-4-pyridyl) porphyrin (MnTymPyp) significantly attenuated the lung leak associated with LPS-exposure in mice suggesting a key role for protein nitration in the progression of ALI. In conclusion, this is the first study that suggests a role of the ADMA/DDAH pathway during the development of ALI in mice and that ADMA may be a novel therapeutic biomarker to ascertain the risk for development of ALI.


Assuntos
Lesão Pulmonar Aguda/fisiopatologia , Amidoidrolases/metabolismo , Arginina/análogos & derivados , Óxido Nítrico Sintase/metabolismo , Animais , Arginina/metabolismo , Biomarcadores/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Lipopolissacarídeos/toxicidade , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Óxido Nítrico/metabolismo , Ácido Peroxinitroso/metabolismo
8.
Physiol Genomics ; 40(1): 48-60, 2009 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-19825830

RESUMO

Peroxisome proliferator-activated receptor type gamma (PPARgamma) is a subgroup of the PPAR transcription factor family. Recent studies indicate that loss of PPARgamma is associated with the development of pulmonary hypertension (PH). We hypothesized that the endothelial dysfunction associated with PPARgamma inhibition may play an important role in the disease process by altering cellular gene expression and signaling cascades. We utilized microarray analysis to determine if PPARgamma inhibition induced changes in gene expression in pulmonary arterial endothelial cells (PAEC). We identified 100 genes and expressed sequence tags (ESTs) that were upregulated by >1.5-fold and 21 genes and ESTs that were downregulated by >1.3-fold (P < 0.05) by PPARgamma inhibition. The upregulated genes can be broadly classified into four functional groups: cell cycle, angiogenesis, ubiquitin system, and zinc finger proteins. The genes with the highest fold change in expression: hyaluronan-mediated motility receptor (HMMR), VEGF receptor 2 (Flk-1), endothelial PAS domain protein 1 (EPAS1), basic fibroblast growth factor (FGF-2), and caveolin-1 in PAEC were validated by real time RT-PCR. We further validated the upregulation of HMMR, Flk-1, FGF2, and caveolin-1 by Western blot analysis. In keeping with the microarray results, PPARgamma inhibition led to re-entry of cell cycle at G(1)/S phase and cyclin C upregulation. PPARgamma inhibition also exacerbated VEGF-induced endothelial barrier disruption. Finally we confirmed the downregulation of PPARgamma and the upregulation of HMMR, Flk-1, FGF2, and Cav-1 proteins in the peripheral lung tissues of an ovine model of PH. In conclusion, we have identified an array of endothelial genes modulated by attenuated PPARgamma signaling that may play important roles in the development of PH.


Assuntos
Anilidas/farmacologia , Células Endoteliais/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Hipertensão Pulmonar/genética , Pulmão/patologia , PPAR gama/antagonistas & inibidores , Animais , Artérias/efeitos dos fármacos , Artérias/patologia , Barreira Alveolocapilar/efeitos dos fármacos , Barreira Alveolocapilar/metabolismo , Bovinos , Adesão Celular/efeitos dos fármacos , Adesão Celular/genética , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/genética , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/patologia , Humanos , Hipertensão Pulmonar/patologia , Neovascularização Fisiológica/efeitos dos fármacos , Neovascularização Fisiológica/genética , Reprodutibilidade dos Testes , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ovinos , Ubiquitina/genética , Fator A de Crescimento do Endotélio Vascular/farmacologia , Dedos de Zinco/genética
9.
Lung ; 187(2): 116-27, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19083056

RESUMO

We previously reported that 17beta-estradiol (E2) prevents hyperresponsiveness to carbachol of murine asthmatic tracheal rings in vitro. We now investigated whether E2 is similarly effective in reducing airway hyperreactivity in a murine model of allergic asthma in vivo. Female ovariectomized BALB/c mice were rendered asthmatic by a 25-day protocol of sensitization to ovalbumin. Under positive-pressure ventilation, anesthetized asthmatic mice exhibited a dramatic increase in airway responsiveness to increasing doses of inhaled methacholine compared to PBS-sensitized controls, as reflected in decreased dynamic compliance of the respiratory system and increased tissue damping, tissue elastance, and airway resistance. Furthermore, asthmatic mice exhibited hypercellularity and increased protein concentration in the bronchoalveolar lavage, strong signs of peribronchial cuffing with inflammatory cells and increased goblet cell activity. To test the effects of estrogen, three additional groups of mice were implanted subcutaneously with different amounts of slow-release E2 pellets at the time of ovariectomy and rendered asthmatic as before. E2 dose-dependently inhibited airway hyperresponsiveness to methacholine, reduced bronchoalveolar lavage hypercellularity, and virtually eliminated histologic signs of inflammation and goblet cell hyperactivity. The inflammation and airway hyperactivity in asthmatic mice was associated with an increase in bronchoalveolar lavage levels of TGFbeta1, which was completely abolished in E2-treated asthmatic mice. We conclude that estrogen replacement therapy effectively ameliorates the pathologic profile of murine allergic asthma.


Assuntos
Asma/prevenção & controle , Hiper-Reatividade Brônquica/prevenção & controle , Broncoconstrição/efeitos dos fármacos , Estradiol/administração & dosagem , Terapia de Reposição de Estrogênios , Células Caliciformes/efeitos dos fármacos , Ovariectomia , Alérgenos , Animais , Anticorpos/sangue , Asma/imunologia , Asma/fisiopatologia , Hiper-Reatividade Brônquica/imunologia , Hiper-Reatividade Brônquica/fisiopatologia , Testes de Provocação Brônquica , Líquido da Lavagem Broncoalveolar/imunologia , Broncoconstritores , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Implantes de Medicamento , Estradiol/sangue , Feminino , Células Caliciformes/imunologia , Cloreto de Metacolina , Camundongos , Camundongos Endogâmicos BALB C , Ovalbumina , Mecânica Respiratória/efeitos dos fármacos , Fator de Crescimento Transformador beta1/metabolismo
10.
Am J Respir Cell Mol Biol ; 39(5): 551-9, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18474672

RESUMO

Heat shock protein 90 (hsp90) inhibitors inactivate and/or degrade various client proteins, including many involved in inflammation. Increased vascular permeability is a hallmark of acute lung injury (ALI) and acute respiratory distress syndrome (ARDS). Thus, we tested the hypothesis that hsp90 inhibitors may prevent and/or restore endothelial cell (EC) permeability after injury. Exposure of confluent bovine pulmonary arterial endothelial cell (BPAEC) monolayer to TGF-beta1, thrombin, bacterial lipopolysaccharide (LPS), or vascular endothelial growth factor (VEGF) increased BPAEC permeability, as revealed by decreased transendothelial electrical resistance (TER). Treatment of injured endothelium with hsp90 inhibitors completely restored TER of BPAEC. Similarly, preincubation of BPAEC with hsp90 inhibitors prevented the decline in TER induced by the exposure to thrombin, LPS, VEGF, or TGF-beta1. In addition, hsp90 inhibitors restored the EC barrier function after PMA or nocodazole-induced hyperpermeability. These effects of the hsp90 inhibitors were associated with the restoration of TGF-beta1- or nocodazole-induced decrease in VE-cadherin and beta-catenin expression at EC junctions. The protective effect of hsp90 inhibitors on TGF-beta1-induced hyperpermeability was critically dependent upon preservation of F-actin cytoskeleton and was associated with the inhibition of agonist-induced myosin light chain (MLC) and myosin phosphatase target subunit 1 (MYPT1) phosphorylation, F-actin stress fibers formation, microtubule disassembly, increase in hsp27 phosphorylation, and association of hsp90 with hsp27, but independent of p38MAPK activity. We conclude that hsp90 inhibitors exert barrier protective effects on BPAEC, at least in part, via inhibition of hsp27-mediated, agonist-induced cytoskeletal rearrangement, and therefore may have useful therapeutic value in ALI, ARDS, and other pulmonary inflammatory disease.


Assuntos
Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Proteínas de Choque Térmico HSP90/metabolismo , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Actinas/metabolismo , Animais , Bovinos , Permeabilidade da Membrana Celular , Células Cultivadas , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/metabolismo , Ativação Enzimática/efeitos dos fármacos , Macrolídeos/farmacologia , Fator de Crescimento Transformador beta1/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
11.
Am J Physiol Lung Cell Mol Physiol ; 294(4): L755-63, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18245267

RESUMO

Endothelial hyperperme ability leading to vascular leak is an important consequence of sepsis and sepsis-induced lung injury. We previously reported that heat shock protein (hsp) 90 inhibitor pretreatment improved pulmonary barrier dysfunction in a murine model of sepsis-induced lung injury. We now examine the effects of hsp90 inhibitors on LPS-mediated endothelial hyperpermeability, as reflected in changes in transendothelial electrical resistance (TER) of bovine pulmonary arterial endothelial cells (BPAEC). Vehicle-pretreated cells exposed to endotoxin exhibited a concentration-dependent decrease in TER, activation of pp60(Src), phosphorylation of the focal adhesion protein paxillin, and reduced expression of the adherens junction proteins, vascular endothelial (VE)-cadherin and beta-catenin. Pretreatment with the hsp90 inhibitor, radicicol, prevented the decrease in TER, maintained VE-cadherin and beta-catenin expression, and inhibited activation of pp60(Src) and phosphorylation of paxillin. Similarly, when BPAEC hyperpermeability was induced by endotoxin-activated neutrophils, pretreatment of neutrophils and/or endothelial cells with radicicol protected against the activated neutrophil-induced decrease in TER. Increased paxillin phosphorylation and decreased expression of beta-catenin and VE-cadherin were also observed in mouse lungs 12 h after intraperitoneal endotoxin and attenuated in mice pretreated with radicicol. These results suggest that hsp90 plays an important role in sepsis-associated endothelial barrier dysfunction.


Assuntos
Permeabilidade da Membrana Celular/efeitos dos fármacos , Endotélio Vascular/fisiologia , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Lipopolissacarídeos/farmacologia , Artéria Pulmonar/fisiologia , Animais , Bovinos , Células Cultivadas , Condutividade Elétrica , Impedância Elétrica , Eletrofisiologia , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Lipopolissacarídeos/antagonistas & inibidores , Artéria Pulmonar/citologia , Artéria Pulmonar/efeitos dos fármacos , Sepse/prevenção & controle
12.
Am J Physiol Heart Circ Physiol ; 293(5): H3080-7, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17873020

RESUMO

The nitric oxide receptor soluble guanylyl cyclase (sGC) exists in multimeric protein complexes, including heat shock protein (HSP) 90 and endothelial nitric oxide synthase. Inhibition of HSP90 by geldanamycin causes proteasomal degradation of sGC protein. In this study, we have investigated whether COOH terminus of heat shock protein 70-interacting protein (CHIP), a co-chaperone molecule that is involved in protein folding but is also a chaperone-dependent ubiquitin E3 ligase, could play a role in the process of degradation of sGC. Transient overexpression of CHIP in COS-7 cells degraded heterologous sGC in a concentration-related manner; this downregulation of sGC was abrogated by the proteasome inhibitor MG-132. Transfection of tetratricopeptide repeats and U-box domain CHIP mutants attenuated sGC degradation, suggesting that both domains are indispensable for CHIP function. Results from immunoprecipitation and indirect immunofluorescent microscopy experiments demonstrated that CHIP is associated with sGC, HSP90, and HSP70 in COS-7 cells. Furthermore, CHIP increased the association of HSP70 with sGC. In in vitro ubiquitination assays using purified proteins and ubiquitin enzymes, E3 ligase CHIP directly ubiquitinated sGC; this ubiquitination was potentiated by geldanamycin in COS-7 cells, followed by proteasomal degradation. In rat aortic smooth muscle cells, endogenous sGC was also degraded by adenovirus-infected wild-type CHIP but not by the chaperone interaction-deficient K30A CHIP, whereas CHIP, but not K30A, attenuated sGC expression in, and nitric oxide donor-induced relaxation of, rat aortic rings, suggesting that CHIP plays a regulatory role under physiological conditions. This study reveals a new mechanism for the regulation of sGC, an important mediator of cellular and vascular function.


Assuntos
Guanilato Ciclase/metabolismo , Proteínas de Choque Térmico HSC70/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Transdução de Sinais/fisiologia , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitina/metabolismo , Animais , Células COS , Chlorocebus aethiops , Chaperonas Moleculares/metabolismo , Solubilidade , Guanilil Ciclase Solúvel
13.
Am J Respir Crit Care Med ; 176(7): 667-75, 2007 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-17615388

RESUMO

RATIONALE: Severe sepsis is the leading cause of death for patients in intensive care units. Patients with severe sepsis develop multiple organ failure, including acute lung injury (ALI), resulting from a deregulated inflammatory response. Inhibitors of the ubiquitous chaperone, heat shock protein 90 (Hsp90), block the activity of certain proinflammatory mediators in vitro. We hypothesized that Hsp90 inhibitors may ameliorate the inflammation and ALI associated with severe sepsis. OBJECTIVES: To test the hypothesis that Hsp90 inhibitors prolong survival, attenuate inflammation, and reduce lung injury in a murine model of sepsis. METHODS: Male C57BL/6 mice received either one of two Hsp90 inhibitors, radicicol or 17-allylaminodemethoxygeldanamycin (17-AAG), 24, 12, 6, and 0 hours before receiving a lethal dose of endotoxin (6.75 x 10(4) endotoxin units/g body weight). Outcomes included survival and parameters of systemic inflammation (plasma neutrophil, cytokine, chemokine, and nitrite/nitrate levels), pulmonary inflammation (lung nuclear factor-kappaB and myeloperoxidase activities, inducible nitric oxide synthase expression, inducible nitric oxide synthase-Hsp90 complex formation, and leukocyte infiltration), and lung injury (pulmonary capillary leak and lung function). MEASUREMENTS AND MAIN RESULTS: Mice pretreated with vehicle and receiving endotoxin exhibited 100% 24-hour lethality, a dramatic increase in all parameters of systemic and pulmonary inflammation, increased capillary leak, and reduced lung function. Compared with them, mice receiving either radicicol or 17-AAG before endotoxin exhibited prolonged survival, reduced or abolished increases in systemic and pulmonary inflammatory parameters, attenuated capillary leak, and restored, normal lung function. CONCLUSIONS: Hsp90 inhibitors may offer a new pharmacological tool in the management of severe sepsis and severe sepsis-induced ALI.


Assuntos
Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Mediadores da Inflamação/metabolismo , Síndrome do Desconforto Respiratório/prevenção & controle , Sepse/fisiopatologia , Animais , Benzoquinonas/farmacologia , Quimiocinas/metabolismo , Citocinas/metabolismo , Inflamação , Lactamas Macrocíclicas/farmacologia , Pulmão/metabolismo , Macrolídeos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Nitratos/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Nitritos/metabolismo , Peroxidase/metabolismo , Síndrome do Desconforto Respiratório/etiologia , Síndrome do Desconforto Respiratório/metabolismo , Síndrome do Desconforto Respiratório/mortalidade , Sepse/complicações , Taxa de Sobrevida
14.
Shock ; 28(3): 353-9, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17545942

RESUMO

Epigallocatechin-3-gallate (EGCG) is the main polyphenolic flavonoid found in green tea. Recent in vitro studies have suggested that EGCG inhibits activation of the nuclear factor-kappaB (NF-kappaB) pathway. The NF-kappaB is a transcriptional factor required for gene expression of many inflammatory mediators, including the inducible isoform of nitric oxide synthase (NOS2). Excessive NO production by NOS2 is directly linked to the vasoplegia, shock, and mortality associated with sepsis. Accordingly, we hypothesized that EGCG administration would inhibit NOS2 gene expression and thereby improve survival in a rodent model of polymicrobial sepsis. Polymicrobial sepsis was induced in male Sprague-Dawley rats (hemodynamic study) and C57BL6 mice (mortality study) via cecal ligation and double puncture (CL2P). Rodents were treated with either EGCG (10 mg/kg intraperitoneally) or vehicle at 1 and 6 h after CL2P and every 12 h thereafter. In the hemodynamic study, mean arterial blood pressure was monitored for 18 h, and rats were killed at 3, 6, and 18 h after CL2P. In the mortality study, survival was monitored for 72 h after CL2P in mice. In vehicle-treated rodents, CL2P was associated with profound hypotension and greater than 80% mortality rate. Epigallocatechin-3-gallate treatment significantly improved both the hypotension and survival. In vitro experiments further showed that EGCG inhibited activation of NF-kappaB and subsequent NOS2 gene expression in a primary culture of rat aortic smooth muscle cells. Epigallocatechin-3-gallate may therefore represent a potential nutritional supplement or pharmacologic agent in patients with sepsis.


Assuntos
Catequina/análogos & derivados , Sepse/tratamento farmacológico , Animais , Pressão Sanguínea/efeitos dos fármacos , Catequina/uso terapêutico , Modelos Animais de Doenças , Expressão Gênica/efeitos dos fármacos , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/antagonistas & inibidores , Nitratos/sangue , Óxido Nítrico Sintase Tipo II/antagonistas & inibidores , Óxido Nítrico Sintase Tipo II/metabolismo , Nitritos/sangue , Ratos , Ratos Sprague-Dawley , Sepse/mortalidade , Taxa de Sobrevida
15.
Am J Physiol Heart Circ Physiol ; 292(2): H893-903, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17012359

RESUMO

Large (pathological) amounts of nitric oxide (NO) induce cell injury, whereas low (physiological) NO concentrations often ameliorate cell injury. We tested the hypotheses that pretreatment of endothelial cells with low concentrations of NO (preconditioning) would prevent injury induced by high NO concentrations. Apoptosis, induced in bovine aortic endothelial cells (BAECs) by exposing them to either 4 mM sodium nitroprusside (SNP) or 0.5 mM N-(2-aminoethyl)-N-(2-hydroxy-2-nitrosohydrazino)-1,2-ethylenediamine (spermine NONOate) for 8 h, was abolished by 24-h pretreatment with either 100 microM SNP, 10 microM spermine NONOate, or 100 microM 8-bromo-cGMP (8-Br-cGMP). Repair of BAECs following wounding, measured as the recovery rate of transendothelial electrical resistance, was delayed by 8-h exposure to 4 mM SNP, and this delay was significantly attenuated by 24-h pretreatment with 100 microM SNP. NO preconditioning produced increased association and expression of soluble guanyl cyclase (sGC) and heat shock protein 90 (HSP90). The protective effect of NO preconditioning, but not the injurious effect of 4 mM SNP, was abolished by either a sGC activity inhibitor 1H-[1,2,4]oxadiazolo-[4,3-a]quinoxalin-1-one (ODQ) or a HSP90 binding inhibitor (radicicol) and was mimicked by 8-Br-cGMP. We conclude that preconditioning with a low dose of NO donor accelerates repair and maintains endothelial integrity via a mechanism that includes the HSP90/sGC pathway. HSP90/sGC may thus play a role in the protective effects of NO-generating drugs from injurious stimuli.


Assuntos
Células Endoteliais/metabolismo , Guanilato Ciclase/metabolismo , Proteínas de Choque Térmico HSP90/metabolismo , Doadores de Óxido Nítrico/farmacologia , Óxido Nítrico/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Cicatrização/efeitos dos fármacos , Citoesqueleto de Actina/efeitos dos fármacos , Citoesqueleto de Actina/metabolismo , Animais , Aorta/efeitos dos fármacos , Aorta/metabolismo , Apoptose/efeitos dos fármacos , Bovinos , Células Cultivadas , GMP Cíclico/análogos & derivados , GMP Cíclico/metabolismo , GMP Cíclico/farmacologia , Proteínas Quinases Dependentes de GMP Cíclico/antagonistas & inibidores , Relação Dose-Resposta a Droga , Impedância Elétrica , Células Endoteliais/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Guanilato Ciclase/antagonistas & inibidores , Nitroprussiato/farmacologia , Oxidiazóis/farmacologia , Quinoxalinas/farmacologia , Receptores Citoplasmáticos e Nucleares/antagonistas & inibidores , Guanilil Ciclase Solúvel , Espermina/análogos & derivados , Espermina/farmacologia , Fatores de Tempo
16.
Am J Physiol Heart Circ Physiol ; 285(2): H669-78, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12676772

RESUMO

Soluble guanylate cyclase (sGC) is an important downstream intracellular target of nitric oxide (NO) that is produced by endothelial NO synthase (eNOS) and inducible NO synthase (iNOS). In this study, we demonstrate that sGC exists in a complex with eNOS and heat shock protein 90 (HSP90) in aortic endothelial cells. In addition, we show that in aortic smooth muscle cells, sGC forms a complex with HSP90. Formation of the sGC/eNOS/HSP90 complex is increased in response to eNOS-activating agonists in a manner that depends on HSP90 activity. In vitro binding assays with glutathione S-transferase fusion proteins that contain the alpha- or beta-subunit of sGC show that the sGC beta-subunit interacts directly with HSP90 and indirectly with eNOS. Confocal immunofluorescent studies confirm the subcellular colocalization of sGC and HSP90 in both endothelial and smooth muscle cells. Complex formation of sGC with HSP90 facilitates responses to NO donors in cultured cells (cGMP accumulation) as well as in anesthetized rats (hypotension). These complexes likely function to stabilize sGC as well as to provide directed intracellular transfer of NO from NOS to sGC, thus preventing inactivation of NO by superoxide anion and formation of peroxynitrite, which is a toxic molecule that has been implicated in the pathology of several vascular diseases.


Assuntos
Endotélio Vascular/enzimologia , Guanilato Ciclase/metabolismo , Proteínas de Choque Térmico HSP90/metabolismo , Músculo Liso Vascular/enzimologia , Óxido Nítrico Sintase/metabolismo , Animais , Aorta/citologia , Benzoquinonas , Bovinos , Células Cultivadas , Endotélio Vascular/citologia , Inibidores Enzimáticos/farmacologia , Lactamas Macrocíclicas , Músculo Liso Vascular/citologia , Doadores de Óxido Nítrico/farmacologia , Óxido Nítrico Sintase Tipo II , Óxido Nítrico Sintase Tipo III , Nitroprussiato/farmacologia , Quinonas/farmacologia , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
17.
Cardiovasc Res ; 53(3): 650-61, 2002 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-11861036

RESUMO

OBJECTIVE: Postmenopausal estrogen replacement therapy lowers the incidence of cardiovascular disease, suggesting that estrogens support cardiovascular function. Estrogens dilate coronary arteries; however, little is known about the molecular basis of how estrogen affects the human coronary circulation. The cellular/molecular effects of estrogen action on human coronary smooth muscle were investigated in the present study. METHODS: Patch-clamp and fluorescent microscopy studies were performed on human coronary myocytes in the absence of endothelium. RESULTS: Estrogen increased whole-cell currents over a range of membrane potentials, and further studies indicated that the large-conductance (186.5 +/- 3 pS), calcium- and voltage-activated potassium (BK(Ca)) channel was the target of estrogen action. Channel activity was stimulated approximately 15-fold by nanomolar concentrations of 17 beta-estradiol, and this stimulation was reversed >90% by inhibiting cGMP-dependent protein kinase activity with 300 nM KT5823. 17 beta-Estradiol increased the level of cGMP and nitric oxide in human myocytes, and the stimulatory effect of estrogen on channel activity and NO production was reversed by inhibiting NO synthase with 10 microM N(G)-monomethyl-L-arginine. CONCLUSIONS: Our cellular and molecular studies identify the BK(Ca) channel as a target of estrogen action in human coronary artery smooth muscle. This response to estrogen involves cGMP-dependent phosphorylation of the BK(Ca) channel or a closely associated regulatory molecule, and further evidence suggests involvement of the NO/cGMP signaling system in coronary smooth muscle. These findings are the first to provide direct evidence for a molecular mechanism that can account for endothelium-independent effects of estrogen on human arteries, and may also help explain why estrogens reduce myocardial ischemia and stimulate coronary blood flow in patients with diseased coronary arteries.


Assuntos
Carbazóis , Estradiol/farmacologia , Indóis , Músculo Liso Vascular/metabolismo , Peptídeos/farmacologia , Canais de Potássio Cálcio-Ativados/efeitos dos fármacos , Canais de Potássio de Abertura Dependente da Tensão da Membrana/efeitos dos fármacos , Alcaloides/farmacologia , Análise de Variância , Vasos Coronários , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Humanos , Microscopia de Fluorescência , Músculo Liso Vascular/efeitos dos fármacos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase/antagonistas & inibidores , Técnicas de Patch-Clamp , ômega-N-Metilarginina/farmacologia
18.
Am J Physiol Cell Physiol ; 282(1): C144-52, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11742807

RESUMO

In rat aortic smooth muscle cells (RASMC), interferon (IFN)-gamma enhanced nitrite accumulation and type II nitric oxide synthase (iNOS) protein expression induced by interleukin (IL)-1 beta. IFN-gamma alone had no effect on nitrite accumulation or iNOS protein. IL-1 beta, but not IFN-gamma, induced nuclear factor (NF)-kappa B and CCAAT box/enhancer binding protein (C/EBP) nuclear binding. Conversely, IFN-gamma, but not IL-1 beta, induced signal transducer and activator of transcription (STAT) 1 and interferon regulatory factor (IRF)-1 binding. In a -1.4-kb rat iNOS promoter segment, deletion of an IFN-gamma-activated site (GAS) increased IL-1 beta-induced activity but inhibited IFN-gamma-enhanced activity, suggesting a two-way effect of the GAS site on iNOS induction: enhancing induction through STAT1 activation and inhibiting induction through a non-IFN-gamma-mediated mechanism. Deletion of both an IRF and a C/EBP site reduced the IL-1 beta-induced and the IFN-gamma-enhanced activities. However, IRF site mutations decreased the IFN-gamma-enhanced activity without affecting the IL-1 beta-induced activity. Insertion of two IRF sites increased the IFN-gamma-enhanced, but not the IL-1 beta-induced, activity. Mutations of a reverse NF-kappa B site did not significantly change IFN-gamma-enhanced activity. We conclude that in RASMC, NF-kappa B and C/EBP mediate the IL-1 beta-induced iNOS expression, whereas IRF-1 and STAT1 mediate the IFN-gamma-enhanced iNOS induction.


Assuntos
Antineoplásicos/farmacologia , Regulação Enzimológica da Expressão Gênica/fisiologia , Interferon gama/farmacologia , Interleucina-1/farmacologia , Músculo Liso Vascular/enzimologia , Óxido Nítrico Sintase/genética , Animais , Aorta/citologia , Sequência de Bases , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Primers do DNA , Proteínas de Ligação a DNA/metabolismo , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Fator Regulador 1 de Interferon , Dados de Sequência Molecular , Músculo Liso Vascular/citologia , Mutagênese/fisiologia , NF-kappa B/metabolismo , Óxido Nítrico Sintase Tipo II , Fosfoproteínas/metabolismo , Regiões Promotoras Genéticas/fisiologia , Ratos , Ratos Wistar , Fator de Transcrição STAT1 , Transativadores/metabolismo , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...