Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
JCI Insight ; 5(14)2020 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-32573489

RESUMO

Cigarette smoking (CS) and genetic susceptibility determine the risk for development, progression, and severity of chronic obstructive pulmonary diseases (COPD). We posited that an incidental balanced reciprocal chromosomal translocation was linked to a patient's risk of severe COPD. We determined that 46,XX,t(1;4)(p13.1;q34.3) caused a breakpoint in the immunoglobulin superfamily member 3 (IGSF3) gene, with markedly decreased expression. Examination of COPDGene cohort identified 14 IGSF3 SNPs, of which rs1414272 and rs12066192 were directly and rs6703791 inversely associated with COPD severity, including COPD exacerbations. We confirmed that IGSF3 is a tetraspanin-interacting protein that colocalized with CD9 and integrin B1 in tetraspanin-enriched domains. IGSF3-deficient patient-derived lymphoblastoids exhibited multiple alterations in gene expression, especially in the unfolded protein response and ceramide pathways. IGSF3-deficient lymphoblastoids had high ceramide and sphingosine-1 phosphate but low glycosphingolipids and ganglioside levels, and they were less apoptotic and more adherent, with marked changes in multiple TNFRSF molecules. Similarly, IGSF3 knockdown increased ceramide in lung structural cells, rendering them more adherent, with impaired wound repair and weakened barrier function. These findings suggest that, by maintaining sphingolipid and membrane receptor homeostasis, IGSF3 is required for cell mobility-mediated lung injury repair. IGSF3 deficiency may increase susceptibility to CS-induced lung injury in COPD.


Assuntos
Fumar Cigarros/genética , Predisposição Genética para Doença , Imunoglobulinas/genética , Proteínas de Membrana/genética , Doença Pulmonar Obstrutiva Crônica/genética , Translocação Genética/genética , Apoptose/genética , Adesão Celular/genética , Movimento Celular/genética , Cromossomos Humanos Par 1/genética , Cromossomos Humanos Par 4/genética , Fumar Cigarros/efeitos adversos , Feminino , Regulação da Expressão Gênica/genética , Humanos , Integrina beta1/genética , Masculino , Pessoa de Meia-Idade , Mutação/genética , Polimorfismo de Nucleotídeo Único/genética , Doença Pulmonar Obstrutiva Crônica/induzido quimicamente , Doença Pulmonar Obstrutiva Crônica/patologia , Índice de Gravidade de Doença , Tetraspanina 29/genética
2.
Am J Physiol Lung Cell Mol Physiol ; 312(1): L143-L153, 2017 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-27913425

RESUMO

Elevated levels of reactive oxygen species and intracellular Ca2+ play a key role in endothelial barrier dysfunction in acute lung injury. We previously showed that H2O2-induced increases in intracellular calcium concentrations ([Ca2+]i) in lung microvascular endothelial cells (LMVECs) involve the membrane Ca2+ channel, transient receptor potential vanilloid-4 (TRPV4) and that inhibiting this channel attenuated H2O2-induced barrier disruption in vitro. We also showed that phosphorylation of TRPV4 by the Src family kinase, Fyn, contributes to H2O2-induced Ca2+ influx in LMVEC. In endothelial cells, Fyn is tethered to the cell membrane by CD36, a fatty acid transporter. In this study, we assessed the effect of genetic loss or pharmacological inhibition of CD36 on Ca2+ responses to H2O2 H2O2-induced Ca2+ influx was attenuated in LMVEC isolated from mice lacking CD36 (CD36-/-). TRPV4 expression and function was unchanged in LMVEC isolated from wild-type (WT) and CD36-/- mice, as well as mice with deficiency for Fyn (Fyn-/-). TRPV4 immunoprecipitated with Fyn, but this interaction was decreased in CD36-/- LMVEC. The amount of phosphorylated TRPV4 was decreased in LMVEC from CD36-/- mice compared with WT controls. Loss of CD36 altered subcellular localization of Fyn, while inhibition of CD36 fatty acid transport with succinimidyl oleate did not attenuate H2O2-induced Ca2+ influx. Lastly, we found that CD36-/- mice were protected from ischemia-reperfusion injury in vivo. In conclusion, our data suggest that CD36 plays an important role in H2O2-mediated lung injury and that the mechanism may involve CD36-dependent scaffolding of Fyn to the cell membrane to facilitate TRPV4 phosphorylation.


Assuntos
Antígenos CD36/metabolismo , Cálcio/metabolismo , Células Endoteliais/metabolismo , Peróxido de Hidrogênio/farmacologia , Pulmão/irrigação sanguínea , Microvasos/citologia , Animais , Células Endoteliais/efeitos dos fármacos , Ácidos Graxos/metabolismo , Deleção de Genes , Lipoproteínas LDL/farmacologia , Camundongos Endogâmicos C57BL , Ácido Oleico/farmacologia , Fosforilação/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-fyn/metabolismo , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia , Frações Subcelulares/efeitos dos fármacos , Frações Subcelulares/metabolismo , Canais de Cátion TRPV/metabolismo
3.
Am J Physiol Lung Cell Mol Physiol ; 309(12): L1467-77, 2015 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-26453519

RESUMO

In acute respiratory distress syndrome, both reactive oxygen species (ROS) and increased intracellular calcium ([Ca(2+)]i) are thought to play important roles in promoting endothelial paracellular permeability, but the mechanisms linking ROS and [Ca(2+)]i in microvascular endothelial cells are not known. In this study, we assessed the effect of hydrogen peroxide (H2O2) on [Ca(2+)]i in mouse and human lung microvascular endothelial cells (MLMVEC and HLMVEC, respectively). We found that in both MLMVECs and HLMVECs, exogenously applied H2O2 increased [Ca(2+)]i through Ca(2+) influx and that pharmacologic inhibition of the calcium channel transient receptor potential vanilloid 4 (TRPV4) attenuated the H2O2-induced Ca(2+) influx. Additionally, knockdown of TRPV4 in HLMVEC also attenuated calcium influx following H2O2 challenge. Administration of H2O2 or TRPV4 agonists decreased transmembrane electrical resistance (TER), suggesting increased barrier permeability. To explore the regulatory mechanisms underlying TRPV4 activation by ROS, we examined H2O2-induced Ca(2+) influx in MLMVECs and HLMVECs with either genetic deletion, silencing, or pharmacologic inhibition of Fyn, a Src family kinase. In both MLMVECs derived from mice deficient for Fyn and HLMVECs treated with either siRNA targeted to Fyn or the Src family kinase inhibitor SU-6656 for 24 or 48 h, the H2O2-induced Ca(2+) influx was attenuated. Treatment with SU-6656 decreased the levels of phosphorylated, but not total, TRPV4 protein and had no effect on TRPV4 response to the external agonist, GSK1016790A. In conclusion, our data suggest that application of exogenous H2O2 increases [Ca(2+)]i and decreases TER in microvascular endothelial cells via activation of TRPV4 through a mechanism that requires the Src kinase Fyn.


Assuntos
Cálcio/metabolismo , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Peróxido de Hidrogênio/farmacologia , Pulmão/metabolismo , Microvasos/metabolismo , Canais de Cátion TRPV/metabolismo , Animais , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Células Cultivadas , Células Endoteliais/efeitos dos fármacos , Endotélio Vascular/efeitos dos fármacos , Humanos , Leucina/análogos & derivados , Leucina/farmacologia , Pulmão/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microvasos/efeitos dos fármacos , Permeabilidade/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-fyn/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Sulfonamidas/farmacologia , Quinases da Família src/metabolismo
4.
J Leukoc Biol ; 96(5): 907-15, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25063878

RESUMO

PDE2A is a dual-function PDE that is stimulated by cGMP to hydrolyze cAMP preferentially. In a two-hit model of ALI, we found previously that PDE2A decreased lung cAMP, up-regulated lung iNOS, and exacerbated ALI. Recent data suggest that macrophage iNOS expression contributes to ALI but later, promotes lung-injury resolution. However, macrophage iNOS is increased by cAMP, suggesting that PDE2A could negatively regulate macrophage iNOS expression. To test this, we examined the effects of manipulating PDE2A expression and function on LPS-induced iNOS expression in a mouse AM cell line (MH-S) and primary mouse AMs. In MH-S cells, LPS (100 ng/ml) increased PDE2A expression by 15% at 15 min and 50% at 6 h before decreasing at 24 h and 48 h. iNOS expression appeared at 6 h and remained increased 48 h post-LPS. Compared with control Ad, Ad.PDE2A-shRNA enhanced LPS-induced iNOS expression further by fourfold, an effect mimicked by the PDE2A inhibitor BAY 60-7550. Adenoviral PDE2A overexpression or treatment with ANP decreased LPS-induced iNOS expression. ANP-induced inhibition of iNOS was lost by knocking down PDE2A and was not mimicked by 8-pCPT-cGMP, a cGMP analog that does not stimulate PDE2A activity. Finally, we found that in primary AMs from LPS-treated mice, PDE2A knockdown also increased iNOS expression, consistent with the MH-S cell data. We conclude that increased AM PDE2A is an important negative regulator of macrophage iNOS expression.


Assuntos
Nucleotídeo Cíclico Fosfodiesterase do Tipo 2/metabolismo , Regulação da Expressão Gênica , Macrófagos Alveolares/metabolismo , Óxido Nítrico Sintase Tipo II/genética , Animais , GMP Cíclico/análogos & derivados , GMP Cíclico/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 2/antagonistas & inibidores , Nucleotídeo Cíclico Fosfodiesterase do Tipo 2/genética , Regulação para Baixo , Regulação da Expressão Gênica/efeitos dos fármacos , Lipopolissacarídeos/farmacologia , Macrófagos Alveolares/imunologia , Masculino , Camundongos , Óxido Nítrico Sintase Tipo II/metabolismo , Tionucleotídeos/metabolismo
5.
Am J Physiol Cell Physiol ; 306(6): C559-69, 2014 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-24401847

RESUMO

Oxidant injury contributes to acute lung injury (ALI). We previously reported that activation of protein kinase GI (PKGI) posttranscriptionally increased the key antioxidant enzymes catalase and glutathione peroxidase 1 (Gpx-1) and attenuated oxidant-induced cytotoxicity in mouse lung microvascular endothelial cells (MLMVEC). The present studies tested the hypothesis that the antioxidant effect of PKGI is mediated via inhibition of the c-Abl tyrosine kinase. We found that activation of PKGI with the cGMP analog 8pCPT-cGMP inhibited c-Abl activity and decreased c-Abl expression in wild-type but not PKGI(-/-) MLMVEC. Treatment of wild-type MLMVEC with atrial natriuretic peptide also inhibited c-Abl activation. Moreover, treatment of MLMVEC with the c-Abl inhibitor imatinib increased catalase and GPx-1 protein in a posttranscriptional fashion. In imatinib-treated MLMVEC, there was no additional effect of 8pCPT-cGMP on catalase or GPx-1. The imatinib-induced increase in antioxidant proteins was associated with an increase in extracellular H2O2 scavenging by MLMVEC, attenuation of oxidant-induced endothelial barrier dysfunction, and prevention of oxidant-induced endothelial cell death. Finally, in the isolated perfused lung, imatinib prevented oxidant-induced endothelial toxicity. We conclude that cGMP, through activation of PKGI, inhibits c-Abl, leading to increased key antioxidant enzymes and resistance to lung endothelial oxidant injury. Inhibition of c-Abl by active PKGI may be the downstream mechanism underlying PKGI-mediated antioxidant signaling. Tyrosine kinase inhibitors may represent a novel therapeutic approach in oxidant-induced ALI.


Assuntos
Lesão Pulmonar Aguda/metabolismo , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Pulmão/metabolismo , Proteínas Proto-Oncogênicas c-abl/antagonistas & inibidores , Transporte Ativo do Núcleo Celular/fisiologia , Animais , Apoptose/efeitos dos fármacos , Fator Natriurético Atrial/metabolismo , Benzamidas/farmacologia , Catalase/metabolismo , Células Cultivadas , GMP Cíclico/análogos & derivados , GMP Cíclico/farmacologia , Proteínas Quinases Dependentes de GMP Cíclico/efeitos dos fármacos , Proteínas Quinases Dependentes de GMP Cíclico/genética , Células Endoteliais/metabolismo , Ativação Enzimática , Glutationa Peroxidase/metabolismo , Peróxido de Hidrogênio/metabolismo , Mesilato de Imatinib , Pulmão/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oxirredução/efeitos dos fármacos , Piperazinas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-abl/metabolismo , Pirimidinas/farmacologia , RNA Mensageiro/biossíntese , Transdução de Sinais/efeitos dos fármacos , Glutationa Peroxidase GPX1
7.
PLoS One ; 8(8): e71010, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23967147

RESUMO

Severe malaria can trigger acute lung injury characterized by pulmonary edema resulting from increased endothelial permeability. However, the mechanism through which lung fluid conductance is altered during malaria remains unclear. To define the role that the scavenger receptor CD36 may play in mediating this response, C57BL/6J (WT) and CD36-/- mice were infected with P. berghei ANKA and monitored for changes in pulmonary endothelial barrier function employing an isolated perfused lung system. WT lungs demonstrated a >10-fold increase in two measures of paracellular fluid conductance and a decrease in the albumin reflection coefficient (σalb) compared to control lungs indicating a loss of barrier function. In contrast, malaria-infected CD36-/- mice had near normal fluid conductance but a similar reduction in σalb. In WT mice, lung sequestered iRBCs demonstrated production of reactive oxygen species (ROS). To determine whether knockout of CD36 could protect against ROS-induced endothelial barrier dysfunction, mouse lung microvascular endothelial monolayers (MLMVEC) from WT and CD36-/- mice were exposed to H2O2. Unlike WT monolayers, which showed dose-dependent decreases in transendothelial electrical resistance (TER) from H2O2 indicating loss of barrier function, CD36-/- MLMVEC demonstrated dose-dependent increases in TER. The differences between responses in WT and CD36-/- endothelial cells correlated with important differences in the intracellular compartmentalization of the CD36-associated Fyn kinase. Malaria infection increased total lung Fyn levels in CD36-/- lungs compared to WT, but this increase was due to elevated production of the inactive form of Fyn further suggesting a dysregulation of Fyn-mediated signaling. The importance of Fyn in CD36-dependent endothelial signaling was confirmed using in vitro Fyn knockdown as well as Fyn-/- mice, which were also protected from H2O2- and malaria-induced lung endothelial leak, respectively. Our results demonstrate that CD36 and Fyn kinase are critical mediators of the increased lung endothelial fluid conductance caused by malaria infection.


Assuntos
Células Epiteliais Alveolares/metabolismo , Barreira Alveolocapilar/metabolismo , Antígenos CD36/metabolismo , Malária/metabolismo , Plasmodium berghei , Proteínas Proto-Oncogênicas c-fyn/metabolismo , Células Epiteliais Alveolares/patologia , Animais , Barreira Alveolocapilar/fisiopatologia , Antígenos CD36/genética , Eritrócitos/metabolismo , Eritrócitos/parasitologia , Técnicas de Silenciamento de Genes , Pulmão/metabolismo , Pulmão/patologia , Malária/patologia , Camundongos , Camundongos Knockout , Permeabilidade , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais
8.
Nat Med ; 18(8): 1217-23, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22820644

RESUMO

Sepsis, a systemic inflammatory response to infection, commonly progresses to acute lung injury (ALI), an inflammatory lung disease with high morbidity. We postulated that sepsis-associated ALI is initiated by degradation of the pulmonary endothelial glycocalyx, leading to neutrophil adherence and inflammation. Using intravital microscopy, we found that endotoxemia in mice rapidly induced pulmonary microvascular glycocalyx degradation via tumor necrosis factor-α (TNF-α)-dependent mechanisms. Glycocalyx degradation involved the specific loss of heparan sulfate and coincided with activation of endothelial heparanase, a TNF-α-responsive, heparan sulfate-specific glucuronidase. Glycocalyx degradation increased the availability of endothelial surface adhesion molecules to circulating microspheres and contributed to neutrophil adhesion. Heparanase inhibition prevented endotoxemia-associated glycocalyx loss and neutrophil adhesion and, accordingly, attenuated sepsis-induced ALI and mortality in mice. These findings are potentially relevant to human disease, as sepsis-associated respiratory failure in humans was associated with higher plasma heparan sulfate degradation activity; moreover, heparanase content was higher in human lung biopsies showing diffuse alveolar damage than in normal human lung tissue.


Assuntos
Lesão Pulmonar Aguda/fisiopatologia , Endotoxemia/complicações , Glicocálix/fisiologia , Pulmão/fisiopatologia , Neutrófilos/fisiologia , Lesão Pulmonar Aguda/etiologia , Lesão Pulmonar Aguda/prevenção & controle , Transferência Adotiva , Animais , Adesão Celular/fisiologia , Modelos Animais de Doenças , Endotélio/enzimologia , Endotélio/fisiologia , Endotoxemia/fisiopatologia , Ativação Enzimática , Regulação da Expressão Gênica/efeitos dos fármacos , Glucuronidase/análise , Glucuronidase/deficiência , Glucuronidase/fisiologia , Heparitina Sulfato/antagonistas & inibidores , Heparitina Sulfato/metabolismo , Humanos , Molécula 1 de Adesão Intercelular/biossíntese , Molécula 1 de Adesão Intercelular/genética , Perfuração Intestinal/complicações , Perfuração Intestinal/microbiologia , Lipopolissacarídeos/toxicidade , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Alvéolos Pulmonares/enzimologia , Alvéolos Pulmonares/patologia , Receptores Tipo I de Fatores de Necrose Tumoral/deficiência , Receptores Tipo I de Fatores de Necrose Tumoral/fisiologia , Insuficiência Respiratória/enzimologia , Insuficiência Respiratória/patologia , Fator de Necrose Tumoral alfa/fisiologia , Lesão Pulmonar Induzida por Ventilação Mecânica/enzimologia , Lesão Pulmonar Induzida por Ventilação Mecânica/patologia
9.
J Immunol ; 189(5): 2234-45, 2012 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-22844117

RESUMO

Although early events in the pathogenesis of acute lung injury (ALI) have been defined, little is known about the mechanisms mediating resolution. To search for determinants of resolution, we exposed wild type (WT) mice to intratracheal LPS and assessed the response at intervals to day 10, when injury had resolved. Inducible NO synthase (iNOS) was significantly upregulated in the lung at day 4 after LPS. When iNOS-/- mice were exposed to intratracheal LPS, early lung injury was attenuated; however, recovery was markedly impaired compared with WT mice. iNOS-/- mice had increased mortality and sustained increases in markers of lung injury. Adoptive transfer of WT (iNOS+/+) bone marrow-derived monocytes or direct adenoviral gene delivery of iNOS into injured iNOS-/- mice restored resolution of ALI. Irradiated bone marrow chimeras confirmed the protective effects of myeloid-derived iNOS but not of epithelial iNOS. Alveolar macrophages exhibited sustained expression of cosignaling molecule CD86 in iNOS-/- mice compared with WT mice. Ab-mediated blockade of CD86 in iNOS-/- mice improved survival and enhanced resolution of lung inflammation. Our findings show that monocyte-derived iNOS plays a pivotal role in mediating resolution of ALI by modulating lung immune responses, thus facilitating clearance of alveolar inflammation and promoting lung repair.


Assuntos
Lesão Pulmonar Aguda/enzimologia , Lesão Pulmonar Aguda/terapia , Monócitos/enzimologia , Monócitos/imunologia , Óxido Nítrico Sintase Tipo II/uso terapêutico , Lesão Pulmonar Aguda/imunologia , Animais , Antígeno B7-2/biossíntese , Linhagem Celular , Linhagem Celular Transformada , Modelos Animais de Doenças , Mediadores da Inflamação/metabolismo , Mediadores da Inflamação/uso terapêutico , Macrófagos Peritoneais/enzimologia , Macrófagos Peritoneais/imunologia , Masculino , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/patologia , Óxido Nítrico Sintase Tipo II/deficiência
10.
Am J Physiol Lung Cell Mol Physiol ; 301(2): L161-70, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21571906

RESUMO

Phosphodiesterase 2A (PDE2A) is stimulated by cGMP to hydrolyze cAMP, a potent endothelial barrier-protective molecule. We previously found that lung PDE2A contributed to a mouse model of ventilator-induced lung injury (VILI). The purpose of the present study was to determine the contribution of PDE2A in a two-hit mouse model of 1-day intratracheal (IT) LPS followed by 4 h of 20 ml/kg tidal volume ventilation. Compared with IT water controls, LPS alone (3.75 µg/g body wt) increased lung PDE2A mRNA and protein expression by 6 h with a persistent increase in protein through day 4 before decreasing to control levels on days 6 and 10. Similar to the PDE2A time course, the peak in bronchoalveolar lavage (BAL) neutrophils, lactate dehydrogenase (LDH), and protein concentration also occurred on day 4 post-LPS. IT LPS (1 day) and VILI caused a threefold increase in lung PDE2A and inducible nitric oxide synthase (iNOS) and a 24-fold increase in BAL neutrophilia. Compared with a control adenovirus, PDE2A knockdown with an adenovirus expressing a short hairpin RNA administered IT 3 days before LPS/VILI effectively decreased lung PDE2A expression and significantly attenuated BAL neutrophilia, LDH, protein, and chemokine levels. PDE2A knockdown also reduced lung iNOS expression by 53%, increased lung cAMP by nearly twofold, and improved survival from 47 to 100%. We conclude that in a mouse model of LPS/VILI, a synergistic increase in lung PDE2A expression increased lung iNOS and alveolar inflammation and contributed significantly to the ensuing acute lung injury.


Assuntos
Lesão Pulmonar Aguda/etiologia , Nucleotídeo Cíclico Fosfodiesterase do Tipo 2/deficiência , Pulmão/metabolismo , Alvéolos Pulmonares/metabolismo , Alvéolos Pulmonares/patologia , Adenoviridae/enzimologia , Adenoviridae/genética , Animais , Líquido da Lavagem Broncoalveolar/citologia , AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 2/genética , Nucleotídeo Cíclico Fosfodiesterase do Tipo 2/metabolismo , Inflamação/metabolismo , Inflamação/patologia , Lipopolissacarídeos/administração & dosagem , Pulmão/virologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neutrófilos/patologia , Óxido Nítrico Sintase Tipo II/metabolismo , Proteínas/metabolismo , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Volume de Ventilação Pulmonar , Fatores de Tempo , Traqueia , Lesão Pulmonar Induzida por Ventilação Mecânica/metabolismo , Lesão Pulmonar Induzida por Ventilação Mecânica/patologia
11.
Am J Physiol Lung Cell Mol Physiol ; 295(6): L1056-65, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18849438

RESUMO

High tidal volume (HV(T)) ventilation causes pulmonary endothelial barrier dysfunction. HV(T) ventilation also increases lung nitric oxide (NO) and cGMP. NO contributes to HV(T) lung injury, but the role of cGMP is unknown. In the current study, ventilation of isolated C57BL/6 mouse lungs increased perfusate cGMP as a function of V(T). Ventilation with 20 ml/kg V(T) for 80 min increased the filtration coefficient (K(f)), an index of vascular permeability. The increased cGMP and K(f) caused by HV(T) were attenuated by nitric oxide synthase (NOS) inhibition and in lungs from endothelial NOS knockout mice. Inhibition of soluble guanylyl cyclase (sGC) in wild-type lungs (10 muM ODQ) also blocked cGMP generation and inhibited the increase in K(f), suggesting an injurious role for sGC-derived cGMP. sGC inhibition also attenuated lung Evans blue dye albumin extravasation and wet-to-dry weight ratio in an anesthetized mouse model of HV(T) injury. Additional activation of sGC (1.5 muM BAY 41-2272) in isolated lungs at 40 min increased cGMP production and K(f) in lungs ventilated with 15 ml/kg V(T). HV(T) endothelial barrier dysfunction was attenuated with a nonspecific phosphodiesterase (PDE) inhibitor (100 muM IBMX) as well as an inhibitor (10 muM BAY 60-7550) specific for the cGMP-stimulated PDE2A. Concordantly, we found a V(T)-dependent increase in lung cAMP hydrolytic activity and PDE2A protein expression with a decrease in lung cAMP concentration that was blocked by BAY 60-7550. We conclude that HV(T)-induced endothelial barrier dysfunction resulted from a simultaneous increase in NO/sGC-derived cGMP and PDE2A expression causing decreased cAMP.


Assuntos
Nucleotídeo Cíclico Fosfodiesterase do Tipo 2/biossíntese , Guanilato Ciclase/metabolismo , Lesão Pulmonar Induzida por Ventilação Mecânica/enzimologia , 1-Metil-3-Isobutilxantina/farmacologia , Animais , Barreira Alveolocapilar , AMP Cíclico/genética , AMP Cíclico/metabolismo , GMP Cíclico/genética , GMP Cíclico/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 2/genética , Modelos Animais de Doenças , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/genética , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/genética , Guanilato Ciclase/genética , Camundongos , Camundongos Knockout , Óxido Nítrico/genética , Óxido Nítrico/metabolismo , Inibidores de Fosfodiesterase/farmacologia , Lesão Pulmonar Induzida por Ventilação Mecânica/genética
12.
Am J Physiol Lung Cell Mol Physiol ; 294(4): L686-97, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18281604

RESUMO

Increased pulmonary endothelial cGMP was shown to prevent endothelial barrier dysfunction through activation of protein kinase G (PKG(I)). Vasodilator-stimulated phosphoprotein (VASP) has been hypothesized to mediate PKG(I) barrier protection because VASP is a cytoskeletal phosphorylation target of PKG(I) expressed in cell-cell junctions. Unphosphorylated VASP was proposed to increase paracellular permeability through actin polymerization and stress fiber bundling, a process inhibited by PKG(I)-mediated phosphorylation of Ser(157) and Ser(239). To test this hypothesis, we examined the role of VASP in the transient barrier dysfunction caused by H(2)O(2) in human pulmonary artery endothelial cell (HPAEC) monolayers studied without and with PKG(I) expression introduced by adenoviral infection (Ad.PKG). In the absence of PKG(I) expression, H(2)O(2) (100-250 microM) caused a transient increased permeability and pSer(157)-VASP formation that were both attenuated by protein kinase C inhibition. Potentiation of VASP Ser(157) phosphorylation by either phosphatase 2B inhibition with cyclosporin or protein kinase A activation with forskolin prolonged, rather than inhibited, the increased permeability caused by H(2)O(2). With Ad.PKG infection, inhibition of VASP expression with small interfering RNA exacerbated H(2)O(2)-induced barrier dysfunction but had no effect on cGMP-mediated barrier protection. In addition, expression of a Ser-double phosphomimetic mutant VASP failed to reproduce the protective effects of activated PKG(I). Finally, expression of a Ser-double phosphorylation-resistant VASP failed to interfere with the ability of cGMP/PKG(I) to attenuate H(2)O(2)-induced disruption of VE-cadherin homotypic binding. Our results suggest that VASP phosphorylation does not explain the protective effect of cGMP/PKG(I) on H(2)O(2)-induced endothelial barrier dysfunction in HPAEC.


Assuntos
GMP Cíclico/farmacologia , Endotélio Vascular/fisiologia , Fosfoproteínas/metabolismo , Artéria Pulmonar/fisiologia , Vasodilatadores/farmacologia , Colforsina/farmacologia , Ciclosporina/farmacologia , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Humanos , Peróxido de Hidrogênio/farmacologia , Cinética , Fosfosserina/metabolismo , Plasmídeos , Artéria Pulmonar/citologia , Artéria Pulmonar/efeitos dos fármacos , RNA Interferente Pequeno/genética , Transfecção
13.
Am J Physiol Lung Cell Mol Physiol ; 294(4): L714-23, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18223163

RESUMO

Ischemia-reperfusion (IR) causes human lung injury in association with the release of atrial and brain natriuretic peptides (ANP and BNP), but the role of ANP/BNP in IR lung injury is unknown. ANP and BNP bind to natriuretic peptide receptor-A (NPR-A) generating cGMP and to NPR-C, a clearance receptor that can decrease intracellular cAMP. To determine the role of NPR-A signaling in IR lung injury, we administered the NPR-A blocker anantin in an in vivo SWR mouse preparation of unilateral lung IR. With uninterrupted ventilation, the left pulmonary artery was occluded for 30 min and then reperfused for 60 or 150 min. Anantin administration decreased IR-induced Evans blue dye extravasation and wet weight in the reperfused left lung, suggesting an injurious role for NPR-A signaling in lung IR. In isolated mouse lungs, exogenous ANP (2.5 nM) added to the perfusate significantly increased the filtration coefficient sevenfold only if lungs were subjected to IR. This effect of ANP was also blocked by anantin. Unilateral in vivo IR increased endogenous plasma ANP, lung cGMP concentration, and lung protein kinase G (PKG(I)) activation. Anantin enhanced plasma ANP concentrations and attenuated the increase in cGMP and PKG(I) activation but had no effect on lung cAMP. These data suggest that lung IR triggered ANP release and altered endothelial signaling so that NPR-A activation caused increased pulmonary endothelial permeability.


Assuntos
Guanilato Ciclase/fisiologia , Receptores do Fator Natriurético Atrial/fisiologia , Traumatismo por Reperfusão/fisiopatologia , Síndrome do Desconforto Respiratório/fisiopatologia , Animais , Fator Natriurético Atrial/metabolismo , Fator Natriurético Atrial/farmacologia , Moléculas de Adesão Celular/efeitos dos fármacos , Moléculas de Adesão Celular/fisiologia , AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Pulmão/efeitos dos fármacos , Pulmão/fisiologia , Pulmão/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos , Proteínas dos Microfilamentos/efeitos dos fármacos , Proteínas dos Microfilamentos/fisiologia , Perfusão , Fosfoproteínas/efeitos dos fármacos , Fosfoproteínas/fisiologia , Transdução de Sinais
14.
Am J Physiol Lung Cell Mol Physiol ; 290(5): L919-30, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16339778

RESUMO

Increases in endothelial cGMP prevent oxidant-mediated endothelial barrier dysfunction, but the downstream mechanisms remain unclear. To determine the role of cGMP-dependent protein kinase (PKG)(I), human pulmonary artery endothelial cells (HPAEC) lacking PKG(I) expression were infected with a recombinant adenovirus encoding PKG(Ibeta) (Ad.PKG) and compared with uninfected and control-infected (Ad.betagal) HPAEC. Transendothelial electrical resistance (TER), an index of permeability, was measured after H(2)O(2) (250 microM) exposure with or without pretreatment with 8-(4-chlorophenylthio)guanosine 3',5'-cyclic monophosphate (CPT-cGMP). HPAEC infected with Ad.PKG, but not Ad.betagal, expressed PKG(I) protein and demonstrated Ser(239) and Ser(157) phosphorylation of vasodilator-stimulated phosphoprotein after treatment with CPT-cGMP. Adenoviral infection decreased basal permeability equally in Ad.PKG- and Ad.betagal-infected HPAEC compared with uninfected cells. Treatment with CPT-cGMP (100 microM) caused a PKG(I)-independent decrease in permeability (8.2 +/- 0.6%). In all three groups, H(2)O(2) (250 microM) caused a similar approximately 35% increase in permeability associated with increased actin stress fiber formation, intercellular gaps, loss of membrane VE-cadherin, and increased intracellular Ca(2+) concentration ([Ca(2+)](i)). In uninfected and Ad.betagal-infected HPAEC, pretreatment with CPT-cGMP (100 microM) partially blocked the increased permeability induced by H(2)O(2). In Ad.PKG-infected HPAEC, CPT-cGMP (50 microM) prevented the H(2)O(2)-induced TER decrease, cytoskeletal rearrangement, and loss of junctional VE-cadherin. CPT-cGMP attenuated the peak [Ca(2+)](i) caused by H(2)O(2) similarly (23%) in Ad.betagal- and Ad.PKG-infected HPAEC, indicating a PKG(I)-independent effect. These data suggest that cGMP decreased HPAEC basal permeability by a PKG(I)-independent process, whereas the ability of cGMP to prevent H(2)O(2)-induced barrier dysfunction was predominantly mediated by PKG(I) through a Ca(2+)-independent mechanism.


Assuntos
Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , GMP Cíclico/fisiologia , Endotélio Vascular/fisiologia , Artéria Pulmonar/fisiologia , Adenoviridae , Permeabilidade da Membrana Celular , Células Cultivadas , Proteínas Quinases Dependentes de GMP Cíclico/genética , Humanos , Proteínas Recombinantes/metabolismo , Transfecção
15.
Circ Res ; 97(2): 115-24, 2005 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-15994434

RESUMO

Endothelial barrier dysfunction caused by inflammatory agonists is a frequent underlying cause of vascular leak and edema. Novel strategies to preserve barrier integrity could have profound clinical impact. Adenosine triphosphate (ATP) released from endothelial cells by shear stress and injury has been shown to protect the endothelial barrier in some settings. We have demonstrated that ATP and its nonhydrolyzed analogues enhanced barrier properties of cultured endothelial cell monolayers and caused remodeling of cell-cell junctions. Increases in cytosolic Ca2+ and Erk activation caused by ATP were irrelevant to barrier enhancement. Experiments using biochemical inhibitors or siRNA indicated that G proteins (specifically Galphaq and Galphai2), protein kinase A (PKA), and the PKA substrate vasodilator-stimulated phosphoprotein were involved in ATP-induced barrier enhancement. ATP treatment decreased phosphorylation of myosin light chain and specifically activated myosin-associated phosphatase. Depletion of Galphaq with siRNA prevented ATP-induced activation of myosin phosphatase. We conclude that the mechanisms of ATP-induced barrier enhancement are independent of intracellular Ca2+, but involve activation of myosin phosphatase via a novel G-protein-coupled mechanism and PKA.


Assuntos
Trifosfato de Adenosina/farmacologia , Células Endoteliais/efeitos dos fármacos , Transdução de Sinais , Animais , Cálcio/metabolismo , Bovinos , Moléculas de Adesão Celular/fisiologia , Células Cultivadas , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Impedância Elétrica , Células Endoteliais/metabolismo , Ativação Enzimática , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/fisiologia , Humanos , Junções Intercelulares/efeitos dos fármacos , Proteínas dos Microfilamentos , Cadeias Leves de Miosina/metabolismo , Fosfatase de Miosina-de-Cadeia-Leve/fisiologia , Fosfoproteínas/fisiologia , Fosforilação
16.
Am J Physiol Heart Circ Physiol ; 287(2): H927-36, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15277207

RESUMO

Cardiopulmonary bypass (CPB) causes acute lung injury. Reactive oxygen species (ROS) from NADPH oxidase may contribute to this injury. To determine the role of NADPH oxidase, we pretreated pigs with structurally dissimilar NADPH oxidase inhibitors. Low-dose apocynin (4-hydroxy-3-methoxy-acetophenone; 200 mg/kg, n = 6), high-dose apocynin (400 mg/kg, n = 6), or diphenyleneiodonium (DPI; 8 mg/kg) was compared with diluent (n = 8). An additional group was treated with indomethacin (10 mg/kg, n = 3). CPB was performed for 2 h with deflated lungs, complete pulmonary artery occlusion, and bronchial artery ligation to maximize lung injury. Parameters of pulmonary function were evaluated for 25 min following CPB. Blood chemiluminescence indicated neutrophil ROS production. Electron paramagnetic resonance determined the effect of apocynin and DPI on in vitro pulmonary endothelial ROS production following hypoxia-reoxygenation. Both apocynin and DPI attenuated blood chemiluminescence and post-CPB hypoxemia. At 25 min post-CPB with Fi(O(2)) = 1, arterial Po(2) (Pa(o(2))) averaged 52 +/- 5, 162 +/- 54, 335 +/- 88, and 329 +/- 119 mmHg in control, low-dose apocynin, high-dose apocynin, and DPI-treated groups, respectively (P < 0.01). Indomethacin had no effect. Pa(O(2)) correlated with blood chemiluminescence measured after drug administration before CPB (R = -0.60, P < 0.005). Neither apocynin nor DPI prevented the increased tracheal pressure, plasma cytokine concentrations (tumor necrosis factor-alpha and IL-6), extravascular lung water, and pulmonary vascular protein permeability observed in control pigs. NADPH oxidase inhibition, but not xanthine oxidase inhibition, significantly blocked endothelial ROS generation following hypoxia-reoxygenation (P < 0.05). NADPH oxidase-derived ROS contribute to the severe hypoxemia but not to the increased cytokine generation and pulmonary vascular protein permeability, which occur following CPB.


Assuntos
Ponte Cardiopulmonar/efeitos adversos , Pneumopatias/etiologia , Pneumopatias/fisiopatologia , NADPH Oxidases/metabolismo , Animais , Contagem de Células Sanguíneas , Pressão Sanguínea , Hipóxia Celular , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Inibidores Enzimáticos/farmacologia , Hematócrito , Medições Luminescentes , Pulmão/patologia , Pneumopatias/sangue , NADPH Oxidases/antagonistas & inibidores , Tamanho do Órgão , Oxigênio/farmacologia , Pressão , Artéria Pulmonar/efeitos dos fármacos , Artéria Pulmonar/metabolismo , Troca Gasosa Pulmonar , Espécies Reativas de Oxigênio/metabolismo , Suínos , Traqueia/fisiopatologia
17.
Am J Physiol Heart Circ Physiol ; 286(2): H693-700, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14563666

RESUMO

Cardiovascular surgery requiring cardiopulmonary bypass (CPB) is frequently complicated by postoperative lung injury. Bronchial artery (BA) blood flow has been hypothesized to attenuate this injury. The purpose of the present study was to determine the effect of BA blood flow on CPB-induced lung injury in anesthetized pigs. In eight pigs (BA ligated) the BA was ligated, whereas in six pigs (BA patent) the BA was identified but left intact. Warm (37 degrees C) CPB was then performed in all pigs with complete occlusion of the pulmonary artery and deflated lungs to maximize lung injury. BA ligation significantly exacerbated nearly all aspects of pulmonary function beginning at 5 min post-CPB. At 25 min, BA-ligated pigs had a lower arterial Po(2) at a fraction of inspired oxygen of 1.0 (52 +/- 5 vs. 312 +/- 58 mmHg) and greater peak tracheal pressure (39 +/- 6 vs. 15 +/- 4 mmHg), pulmonary vascular resistance (11 +/- 1 vs. 6 +/- 1 mmHg x l(-1) x min), plasma TNF-alpha (1.2 +/- 0.60 vs. 0.59 +/- 0.092 ng/ml), extravascular lung water (11.7 +/- 1.2 vs. 7.7 +/- 0.5 ml/g blood-free dry weight), and pulmonary vascular protein permeability, as assessed by a decreased reflection coefficient for albumin (sigma(alb); 0.53 +/- 0.1 vs. 0.82 +/- 0.05). There was a negative correlation (R = 0.95, P < 0.001) between sigma(alb) and the 25-min plasma TNF-alpha concentration. These results suggest that a severe decrease in BA blood flow during and after warm CPB causes increased pulmonary vascular permeability, edema formation, cytokine production, and severe arterial hypoxemia secondary to intrapulmonary shunt.


Assuntos
Artérias Brônquicas/fisiologia , Lesão Pulmonar , Análise de Variância , Animais , Peso Corporal , Ponte Cardiopulmonar/efeitos adversos , Pulmão/irrigação sanguínea , Modelos Animais , Músculo Liso Vascular/fisiologia , Fluxo Sanguíneo Regional , Suínos , Fatores de Tempo
18.
J Appl Physiol (1985) ; 95(5): 1971-8, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12897031

RESUMO

Ischemia-reperfusion (I/R) lung injury causes increased vascular permeability and edema. We developed an in vivo murine model of I/R allowing measurement of pulmonary vascular barrier function without airway occlusion. The left pulmonary artery (PA) was occluded with an exteriorized, slipknotted suture in anesthetized C57BL/6J mice. The effect of ischemic time was determined by subjecting mice to 5, 10, or 30 min of left lung ischemia followed by 150 min of reperfusion. The effect of reperfusion time was determined by subjecting mice to 30 min of left lung ischemia followed by 30 or 150 min of reperfusion. Changes in pulmonary vascular barrier function were measured with the Evans blue dye (EBD) technique, dual-isotope radiolabeled albumin (RA), bronchoalveolar lavage (BAL) protein concentration, and wet weight-to-dry weight ratio (WW/DW). Increasing left lung ischemia with constant reperfusion time or increasing left lung reperfusion time after constant ischemic time resulted in significant increases in left lung EBD content at all times compared with both right lung values and sham surgery mice. The effects of left lung ischemia on lung EBD were corroborated by RA but the effects of increasing reperfusion time differed, suggesting binding of EBD to lung tissue. An increase in WW/DW was only detected after 30 min of reperfusion, suggesting edema clearance. BAL protein concentrations were unaffected. We conclude that short periods of I/R, without airway occlusion, increase pulmonary vascular permeability in the in vivo mouse, providing a useful model to study molecular mechanisms of I/R lung injury.


Assuntos
Circulação Pulmonar/fisiologia , Edema Pulmonar/metabolismo , Edema Pulmonar/fisiopatologia , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/fisiopatologia , Albuminas/farmacocinética , Animais , Brônquios/irrigação sanguínea , Líquido da Lavagem Broncoalveolar , Permeabilidade Capilar/fisiologia , Corantes/farmacocinética , Azul Evans/farmacocinética , Feminino , Camundongos , Camundongos Endogâmicos C57BL
19.
Chest ; 121(3): 806-14, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11888964

RESUMO

BACKGROUND: The presence of right heart thromboemboli complicating pulmonary thromboemboli carries with it an increased mortality rate compared to pulmonary thromboemboli alone, but little is known about the optimal management of this difficult clinical situation. This fact is highlighted in the case study of a patient with a 19-cm right atrial thrombus complicating bilateral pulmonary thromboemboli. STUDY OBJECTIVES: We sought to determine the effects of anticoagulation therapy, thrombolysis, and surgical embolectomy on mortality rate in patients with right heart thromboemboli. DESIGN: Retrospective analysis of all reported cases in the English language literature (1966 to 2000) of right heart thromboembolism in which age, sex, therapy, and outcome were reported. MEASUREMENTS AND RESULTS: We analyzed 177 cases of right heart thromboembolism. Pulmonary thromboembolism was present in 98% of the cases. The patients were evenly divided by gender with an average age of 59.8 years (SD, 16.6 years) years. Dyspnea (54.2%), chest pain (22.6%), and syncope (17.5%) were the most common presenting symptoms. The treatments administered were none (9%), anticoagulation therapy (35.0%), surgical procedure (35.6%), or thrombolytic therapy (19.8%). The overall mortality rate was 27.1%. The mortality rate associated with no therapy, anticoagulation therapy, surgical embolectomy, and thrombolysis was 100.0%, 28.6%, 23.8%, and 11.3%, respectively. Using multivariate modeling with survival as the primary outcome, age and gender were not associated with mortality rate, but thrombolytic therapy was associated with an improved survival rate (p < 0.05) when compared either to anticoagulation therapy or surgery. CONCLUSION: The presence of right heart thromboemboli may have diagnostic and therapeutic implications in pulmonary thromboembolism patients. A well-designed prospective, randomized trial is needed to determine the optimal treatment of right heart thromboemboli.


Assuntos
Cardiopatias/terapia , Trombose/terapia , Idoso , Átrios do Coração , Cardiopatias/complicações , Cardiopatias/mortalidade , Humanos , Modelos Logísticos , Masculino , Pessoa de Meia-Idade , Embolia Pulmonar/complicações , Estudos Retrospectivos , Análise de Sobrevida , Terapia Trombolítica , Trombose/complicações , Trombose/mortalidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA