Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 699
Filtrar
1.
Angiogenesis ; 24(3): 677-693, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33770321

RESUMO

Endothelial barrier disruption and vascular leak importantly contribute to organ dysfunction and mortality during inflammatory conditions like sepsis and acute respiratory distress syndrome. We identified the kinase Arg/Abl2 as a mediator of endothelial barrier disruption, but the role of Arg in endothelial monolayer regulation and its relevance in vivo remain poorly understood. Here we show that depletion of Arg in endothelial cells results in the activation of both RhoA and Rac1, increased cell spreading and elongation, redistribution of integrin-dependent cell-matrix adhesions to the cell periphery, and improved adhesion to the extracellular matrix. We further show that Arg is activated in the endothelium during inflammation, both in murine lungs exposed to barrier-disruptive agents, and in pulmonary microvessels of septic patients. Importantly, Arg-depleted endothelial cells were less sensitive to barrier-disruptive agents. Despite the formation of F-actin stress fibers and myosin light chain phosphorylation, Arg depletion diminished adherens junction disruption and intercellular gap formation, by reducing the disassembly of cell-matrix adhesions and cell retraction. In vivo, genetic deletion of Arg diminished vascular leak in the skin and lungs, in the presence of a normal immune response. Together, our data indicate that Arg is a central and non-redundant regulator of endothelial barrier integrity, which contributes to cell retraction and gap formation by increasing the dynamics of adherens junctions and cell-matrix adhesions in a Rho GTPase-dependent fashion. Therapeutic inhibition of Arg may provide a suitable strategy for the treatment of a variety of clinical conditions characterized by vascular leak.


Assuntos
Matriz Extracelular/metabolismo , Junções Comunicantes/enzimologia , Células Endoteliais da Veia Umbilical Humana/enzimologia , Proteínas Tirosina Quinases/metabolismo , Alvéolos Pulmonares/enzimologia , Animais , Adesão Celular/genética , Ativação Enzimática , Matriz Extracelular/genética , Junções Comunicantes/genética , Humanos , Inflamação/enzimologia , Inflamação/genética , Camundongos , Camundongos Knockout , Proteínas Tirosina Quinases/genética
2.
Part Fibre Toxicol ; 18(1): 11, 2021 03 11.
Artigo em Inglês | MEDLINE | ID: mdl-33706759

RESUMO

BACKGROUND: Air pollution exposure and idiopathic pulmonary fibrosis (IPF) cause a poor prognosis after SARS-CoV-2 infection, but the underlying mechanisms are not well explored. Angiotensin-converting enzyme 2 (ACE2) and transmembrane serine protease 2 (TMPRSS2) are the keys to the entry of SARS-CoV-2. We therefore hypothesized that air pollution exposure and IPF may increase the expression of ACE2 and TMPRSS2 in the lung alveolar region. We measured their expression levels in lung tissues of control non-IPF and IPF patients, and used murine animal models to study the deterioration of IPF caused by particulate matter (PM) and the molecular pathways involved in the expression of ACE2 and TMPRSS2. RESULTS: In non-IPF patients, cells expressing ACE2 and TMPRSS2 were limited to human alveolar cells. ACE2 and TMPRSS2 were largely upregulated in IPF patients, and were co-expressed by fibroblast specific protein 1 (FSP-1) + lung fibroblasts in human pulmonary fibrotic tissue. In animal models, PM exposure increased the severity of bleomycin-induced pulmonary fibrosis. ACE2 and TMPRSS2 were also expressed in FSP-1+ lung fibroblasts in bleomycin-induced pulmonary fibrosis, and when combined with PM exposure, they were further upregulated. The severity of pulmonary fibrosis and the expression of ACE2 and TMPRSS2 caused by PM exposure were blocked by deletion of KC, a murine homologue of IL-8, or treatment with reparixin, an inhibitor of IL-8 receptors CXCR1/2. CONCLUSIONS: These data suggested that risk of SARS-CoV-2 infection and COVID-19 disease severity increased by air pollution exposure and underlying IPF. It can be mediated through upregulating ACE2 and TMPRSS2 in pulmonary fibroblasts, and prevented by blocking the IL-8/CXCR1/2 pathway.


Assuntos
Enzima de Conversão de Angiotensina 2/genética , COVID-19/etiologia , Fibrose Pulmonar Idiopática/complicações , Material Particulado/toxicidade , SARS-CoV-2 , Serina Endopeptidases/genética , Enzima de Conversão de Angiotensina 2/fisiologia , Animais , Humanos , Interleucina-8/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Alvéolos Pulmonares/enzimologia , Serina Endopeptidases/fisiologia , Regulação para Cima
3.
Lung ; 197(2): 227-233, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30759273

RESUMO

BACKGROUND: Pancreatic digestive enzymes present in meconium might be responsible for meconium-induced lung injury. The local Renin Angiotensin System plays an important role in lung injury and inflammation. Particularly, angiotensin converting enzyme-2 (ACE-2) has been identified as a protective lung enzyme against the insult. ACE-2 converts pro-apoptotic Angiotensin II to anti-apoptotic Angiotensin 1-7. However, the effect of meconium on ACE-2 has never been studied before. OBJECTIVE: To study the effect of meconium on ACE-2, and whether inhibition of proteolytic enzymes present in the meconium reverses its effects on ACE-2. METHODS: Alveolar epithelial A549 cells were exposed to F-12 medium, 2.5% meconium, meconium + a protease inhibitor cocktail (PIc) and PIc alone for 16 h. At the end of incubation, apoptosis was measured with a nuclear fragmentation assay and cell lysates were collected for ACE-2 immunoblotting and enzyme activity. RESULTS: Meconium caused a fourfold increase in apoptotic nuclei (p < 0.001). The pro-apoptotic effect of meconium can be reversed by PIc. Meconium reduced ACE-2 enzyme activity by cleaving ACE-2 into a fragment detected at ~ 37 kDa by immunoblot. PIc prevented the degradation of ACE-2 and restored 50% of ACE-2 activity (p < 0.05). CONCLUSION: These data suggest that meconium causes degradation of lung protective ACE-2 by proteolytic enzymes present in meconium, since the effects of meconium can be reversed by PIc.


Assuntos
Células Epiteliais/enzimologia , Síndrome de Aspiração de Mecônio/enzimologia , Mecônio/enzimologia , Peptídeo Hidrolases/metabolismo , Peptidil Dipeptidase A/metabolismo , Alvéolos Pulmonares/enzimologia , Células A549 , Enzima de Conversão de Angiotensina 2 , Apoptose , Estabilidade Enzimática , Células Epiteliais/patologia , Humanos , Síndrome de Aspiração de Mecônio/patologia , Proteólise , Alvéolos Pulmonares/patologia
4.
Am J Respir Cell Mol Biol ; 55(6): 767-778, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27438994

RESUMO

In premature infants, sepsis is associated with alveolar simplification manifesting as bronchopulmonary dysplasia. The redox-dependent mechanisms underlying sepsis-induced inflammation and alveolar remodeling in the immature lung remain unclear. We developed a neonatal mouse model of sepsis-induced lung injury to investigate whether nicotinamide adenine dinucleotide phosphate oxidase 2 (NOX2) regulates Toll-like receptor (TLR)-mediated inflammation and alveolar remodeling. Six-day-old NOX2+/+ and NOX2-/- mice were injected with intraperitoneal LPS to induce sepsis. Lung inflammation and canonical TLR signaling were assessed 24 hours after LPS. Alveolar development was examined in 15-day-old mice after LPS on Day 6. The in vivo efficacy of a NOX2 inhibitor (NOX2-I) on NOX2 complex assembly and sepsis-induced lung inflammation were examined. Lung cytokine expression and neutrophil influx induced with sepsis in NOX2+/+ mice was decreased by >50% in NOX2-/- mice. LPS-induced TLR4 signaling evident by inhibitor of NF-κB kinase-ß and mitogen-activated protein kinase phosphorylation, and nuclear factor-κB/AP-1 translocation were attenuated in NOX2-/- mice. LPS increased matrix metalloproteinase 9 while decreasing elastin and keratinocyte growth factor levels in NOX2+/+ mice. An LPS-induced increase in matrix metalloproteinase 9 and decrease in fibroblast growth factor 7 and elastin were not evident in NOX2-/- mice. An LPS-induced reduction in radial alveolar counts and increased mean linear intercepts were attenuated in NOX2-/- mice. LPS-induced NOX2 assembly evident by p67phox/gp91phox coimmunoprecipitation was disrupted with NOX2-I. NOX2-I also mitigated LPS-induced cytokine expression, TLR pathway signaling, and alveolar simplification. In a mouse model of neonatal sepsis, NOX2 regulates proinflammatory TLR signaling and alveolar remodeling induced by a single dose of LPS. Our results provide mechanistic insight into the regulation of sepsis-induced alveolar remodeling in the developing lung.


Assuntos
Glicoproteínas de Membrana/metabolismo , NADPH Oxidases/metabolismo , Pneumonia/enzimologia , Pneumonia/patologia , Alvéolos Pulmonares/enzimologia , Alvéolos Pulmonares/crescimento & desenvolvimento , Doença Aguda , Animais , Biomarcadores/metabolismo , Citocinas/metabolismo , Matriz Extracelular/metabolismo , Lipopolissacarídeos , Glicoproteínas de Membrana/deficiência , Camundongos , NADPH Oxidase 2 , NADPH Oxidases/deficiência , NF-kappa B/metabolismo , Pneumonia/metabolismo , Alvéolos Pulmonares/patologia , Sepse/complicações , Sepse/metabolismo , Sepse/patologia , Receptores Toll-Like/metabolismo , Fator de Transcrição AP-1/metabolismo
5.
J Biol Chem ; 290(52): 30813-29, 2015 Dec 25.
Artigo em Inglês | MEDLINE | ID: mdl-26518879

RESUMO

Mutations of human telomerase RNA component (TERC) and telomerase reverse transcriptase (TERT) are associated with a subset of lung aging diseases, but the mechanisms by which TERC and TERT participate in lung diseases remain unclear. In this report, we show that knock-out (KO) of the mouse gene Terc or Tert causes pulmonary alveolar stem cell replicative senescence, epithelial impairment, formation of alveolar sacs, and characteristic inflammatory phenotype. Deficiency in TERC or TERT causes a remarkable elevation in various proinflammatory cytokines, including IL-1, IL-6, CXCL15 (human IL-8 homolog), IL-10, TNF-α, and monocyte chemotactic protein 1 (chemokine ligand 2 (CCL2)); decrease in TGF-ß1 and TGFßRI receptor in the lungs; and spillover of IL-6 and CXCL15 into the bronchoalveolar lavage fluids. In addition to increased gene expressions of α-smooth muscle actin and collagen 1α1, suggesting myofibroblast differentiation, TERC deficiency also leads to marked cellular infiltrations of a mononuclear cell population positive for the leukocyte common antigen CD45, low-affinity Fc receptor CD16/CD32, and pattern recognition receptor CD11b in the lungs. Our data demonstrate for the first time that telomerase deficiency triggers alveolar stem cell replicative senescence-associated low-grade inflammation, thereby driving pulmonary premature aging, alveolar sac formation, and fibrotic lesion.


Assuntos
Pneumopatias/imunologia , Alvéolos Pulmonares/enzimologia , Células-Tronco/citologia , Telomerase/deficiência , Animais , Senescência Celular , Feminino , Humanos , Interleucina-1/genética , Interleucina-1/imunologia , Interleucina-6/genética , Interleucina-6/imunologia , Pulmão/citologia , Pulmão/imunologia , Pneumopatias/enzimologia , Pneumopatias/genética , Pneumopatias/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Alvéolos Pulmonares/citologia , Alvéolos Pulmonares/imunologia , RNA/genética , Células-Tronco/imunologia , Telomerase/genética , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia
6.
Blood ; 123(26): 4077-88, 2014 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-24833351

RESUMO

Inflammation is a key process in various diseases, characterized by leukocyte recruitment to the inflammatory site. This study investigates the role of a disintegrin and a metalloproteinase (ADAM) 10 and ADAM17 for leukocyte migration in vitro and in a murine model of acute pulmonary inflammation. Inhibition experiments or RNA knockdown indicated that monocytic THP-1 cells and primary human neutrophils require ADAM10 but not ADAM17 for efficient chemokine-induced cell migration. Signaling and adhesion events that are linked to cell migration such as p38 and ρ GTPase-family activation, F-actin polymerization, adhesion to fibronectin, and up-regulation of α5 integrin were also dependent on ADAM10 but not ADAM17. This was confirmed with leukocytes isolated from mice lacking either ADAM10 or ADAM17 in all hematopoietic cells (vav 1 guanine nucleotide exchange factor [Vav]-Adam10(-/-) or Vav-Adam17(-/-) mice). In lipopolysaccharide-induced acute pulmonary inflammation, alveolar recruitment of neutrophils and monocytes was transiently increased in Vav-Adam17(-/-) but steadily reduced in Vav-Adam10(-/-) mice. This deficit in alveolar leukocyte recruitment was also observed in LysM-Adam10(-/-) mice lacking ADAM10 in myeloid cells and correlated with protection against edema formation. Thus, with regard to leukocyte migration, leukocyte-expressed ADAM10 but not ADAM17 displays proinflammatory activities and may therefore serve as a target to limit inflammatory cell recruitment.


Assuntos
Proteínas ADAM/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Movimento Celular , Proteínas de Membrana/metabolismo , Infiltração de Neutrófilos , Neutrófilos/enzimologia , Pneumonia/enzimologia , Alvéolos Pulmonares/enzimologia , Edema Pulmonar/enzimologia , Proteínas ADAM/genética , Proteína ADAM10 , Proteína ADAM17 , Doença Aguda , Secretases da Proteína Precursora do Amiloide/genética , Animais , Linhagem Celular Tumoral , Células HEK293 , Humanos , Inflamação/induzido quimicamente , Inflamação/enzimologia , Inflamação/genética , Inflamação/patologia , Lipopolissacarídeos/toxicidade , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Neutrófilos/patologia , Pneumonia/induzido quimicamente , Pneumonia/genética , Pneumonia/patologia , Alvéolos Pulmonares/patologia , Edema Pulmonar/induzido quimicamente , Edema Pulmonar/genética , Edema Pulmonar/patologia
7.
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
8.
J Biol Chem ; 289(9): 6165-76, 2014 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-24429287

RESUMO

Mitochondria-targeted human 8-oxoguanine DNA glycosylase (mt-hOgg1) and aconitase-2 (Aco-2) each reduce oxidant-induced alveolar epithelial cell (AEC) apoptosis, but it is unclear whether protection occurs by preventing AEC mitochondrial DNA (mtDNA) damage. Using quantitative PCR-based measurements of mitochondrial and nuclear DNA damage, mtDNA damage was preferentially noted in AEC after exposure to oxidative stress (e.g. amosite asbestos (5-25 µg/cm(2)) or H2O2 (100-250 µM)) for 24 h. Overexpression of wild-type mt-hOgg1 or mt-long α/ß 317-323 hOgg1 mutant incapable of DNA repair (mt-hOgg1-Mut) each blocked A549 cell oxidant-induced mtDNA damage, mitochondrial p53 translocation, and intrinsic apoptosis as assessed by DNA fragmentation and cleaved caspase-9. In contrast, compared with controls, knockdown of Ogg1 (using Ogg1 shRNA in A549 cells or primary alveolar type 2 cells from ogg1(-/-) mice) augmented mtDNA lesions and intrinsic apoptosis at base line, and these effects were increased further after exposure to oxidative stress. Notably, overexpression of Aco-2 reduced oxidant-induced mtDNA lesions, mitochondrial p53 translocation, and apoptosis, whereas siRNA for Aco-2 (siAco-2) enhanced mtDNA damage, mitochondrial p53 translocation, and apoptosis. Finally, siAco-2 attenuated the protective effects of mt-hOgg1-Mut but not wild-type mt-hOgg1 against oxidant-induced mtDNA damage and apoptosis. Collectively, these data demonstrate a novel role for mt-hOgg1 and Aco-2 in preserving AEC mtDNA integrity, thereby preventing oxidant-induced mitochondrial dysfunction, p53 mitochondrial translocation, and intrinsic apoptosis. Furthermore, mt-hOgg1 chaperoning of Aco-2 in preventing oxidant-mediated mtDNA damage and apoptosis may afford an innovative target for the molecular events underlying oxidant-induced toxicity.


Assuntos
Aconitato Hidratase/metabolismo , Dano ao DNA , DNA Glicosilases/metabolismo , DNA Mitocondrial/metabolismo , Células Epiteliais/enzimologia , Mitocôndrias/enzimologia , Oxidantes/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Alvéolos Pulmonares/enzimologia , Aconitato Hidratase/genética , Animais , Apoptose/efeitos dos fármacos , Amianto Amosita/toxicidade , Linhagem Celular Tumoral , DNA Glicosilases/genética , DNA Mitocondrial/genética , Células Epiteliais/patologia , Humanos , Camundongos , Camundongos Knockout , Mitocôndrias/genética , Mitocôndrias/patologia , Mutação , Oxidantes/efeitos adversos , Alvéolos Pulmonares/patologia , Ratos , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
9.
J Biol Chem ; 289(39): 26804-26816, 2014 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-25096573

RESUMO

Tumors are often greatly dependent on signaling cascades promoting cell growth or survival and may become hypersensitive to inactivation of key components within these signaling pathways. Ras and RAF mutations found in human cancer confer constitutive activity to these signaling molecules thereby converting them into an oncogenic state. RAF dimerization is required for normal Ras-dependent RAF activation and is required for the oncogenic potential of mutant RAFs. Here we describe a new mouse model for lung tumor development to investigate the role of B-RAF in oncogenic C-RAF-mediated adenoma initiation and growth. Conditional elimination of B-RAF in C-RAF BxB-expressing embryonic alveolar epithelial type II cells did not block adenoma formation. However, loss of B-RAF led to significantly reduced tumor growth. The diminished tumor growth upon B-RAF inactivation was due to reduced cell proliferation in absence of senescence and increased apoptosis. Furthermore, B-RAF elimination inhibited C-RAF BxB-mediated activation of the mitogenic cascade. In line with these data, mutation of Ser-621 in C-RAF BxB abrogated in vitro the dimerization with B-RAF and blocked the ability to activate the MAPK cascade. Taken together these data indicate that B-RAF is an important factor in oncogenic C-RAF-mediated tumorigenesis.


Assuntos
Adenoma/enzimologia , Transformação Celular Neoplásica/metabolismo , Células Epiteliais/enzimologia , Neoplasias Pulmonares/enzimologia , Sistema de Sinalização das MAP Quinases , Proteínas Proto-Oncogênicas B-raf/metabolismo , Proteínas Proto-Oncogênicas c-raf/metabolismo , Alvéolos Pulmonares/enzimologia , Mucosa Respiratória/enzimologia , Adenoma/genética , Adenoma/patologia , Animais , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Células Epiteliais/patologia , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Transgênicos , Mutação , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Proto-Oncogênicas c-raf/genética , Alvéolos Pulmonares/patologia , Mucosa Respiratória/patologia
10.
Am J Physiol Lung Cell Mol Physiol ; 309(7): L710-24, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-26232299

RESUMO

The gasotransmitter hydrogen sulfide (H2S) is emerging as a mediator of lung physiology and disease. Recent studies revealed that H2S administration limited perturbations to lung structure in experimental animal models of bronchopulmonary dysplasia (BPD), partially restoring alveolarization, limiting pulmonary hypertension, limiting inflammation, and promoting epithelial repair. No studies have addressed roles for endogenous H2S in lung development. H2S is endogenously generated by cystathionine ß-synthase (Cbs) and cystathionine γ-lyase (Cth). We demonstrate here that the expression of Cbs and Cth in mouse lungs is dynamically regulated during lung alveolarization and that alveolarization is blunted in Cbs(-/-) and Cth(-/-) mouse pups, where a 50% reduction in the total number of alveoli was observed, without any impact on septal thickness. Laser-capture microdissection and immunofluorescence staining indicated that Cbs and Cth were expressed in the airway epithelium and lung vessels. Loss of Cbs and Cth led to a 100-500% increase in the muscularization of small- and medium-sized lung vessels, which was accompanied by increased vessel wall thickness, and an apparent decrease in lung vascular supply. Ablation of Cbs expression using small interfering RNA or pharmacological inhibition of Cth using propargylglycine in lung endothelial cells limited angiogenic capacity, causing a 30-40% decrease in tube length and a 50% decrease in number of tubes formed. In contrast, exogenous administration of H2S with GYY4137 promoted endothelial tube formation. These data confirm a key role for the H2S-generating enzymes Cbs and Cth in pulmonary vascular development and homeostasis and in lung alveolarization.


Assuntos
Cistationina beta-Sintase/biossíntese , Cistationina gama-Liase/biossíntese , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Regulação Enzimológica da Expressão Gênica/fisiologia , Sulfeto de Hidrogênio/metabolismo , Alvéolos Pulmonares , Mucosa Respiratória , Animais , Cistationina beta-Sintase/genética , Cistationina gama-Liase/genética , Camundongos , Camundongos Knockout , Alvéolos Pulmonares/irrigação sanguínea , Alvéolos Pulmonares/embriologia , Alvéolos Pulmonares/enzimologia , Mucosa Respiratória/irrigação sanguínea , Mucosa Respiratória/embriologia , Mucosa Respiratória/enzimologia
11.
Biochem Biophys Res Commun ; 459(4): 699-705, 2015 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-25769947

RESUMO

Occurrence of oxidative stress is common in influenza, and renders the host more susceptible to pathogenic effects including cell death. We previously reported that down-regulation of superoxide anion dismutase 1 (SOD1) by influenza A virus (IAV) resulted in a significant increase in the levels of reactive oxygen species (ROS) and viral PB1 polymerase gene product in the early stage of infection. However, the precise molecular mechanism of IAV-mediated ROS generation is not yet fully understood. In this study, we investigated the possible involvement of the key virulence factor PB1-F2 in ROS generation and its contribution to the viral propagation and cell death. The key virulence factor PB1-F2 was found to be responsible, at least in part, for the ROS generation through lowering the SOD1 level in alveolar epithelial A549 cells. PB1-F2 overexpression resulted in SOD1 diminishment and ROS enhancement, while another virulent factor, NS1, did not show significant changes. Inversely, we examined the effects of the absence of PB1-F2 using mutant IAV lacking PB1-F2 expression (mutantΔF2). Infection with mutantΔF2 virus did not significantly lower the SOD1 level, and thus generated moderately low levels of ROS. In addition, the oxidative activity of PB1-F2 was directly reflected by cell viability and death. Infection with the mutant virus reduced the percentage of apoptotic cells more than two-fold compared to the wild-type IAV in A549 cells. Furthermore, expression of exogenous SOD1 gene abrogated a large portion of the PB1-F2-induced apoptosis of cells infected with wild-type IAV, but affected much less of the mutantΔF2 virus-infected cells. These results suggest that the PB1-F2 is directly implicated in virus-induced oxidative stress, thereby contributing to the early stages of IAV replication cycle and ultimately to disease severity.


Assuntos
Alvéolos Pulmonares/metabolismo , Proteínas Virais/fisiologia , Animais , Linhagem Celular , Cães , Humanos , Células Madin Darby de Rim Canino , Oxirredução , Alvéolos Pulmonares/citologia , Alvéolos Pulmonares/enzimologia , Espécies Reativas de Oxigênio/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Superóxido Dismutase/metabolismo , Superóxido Dismutase-1
12.
Pediatr Res ; 78(6): 634-40, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26322414

RESUMO

BACKGROUND: Pulmonary hypertension (PH) worsens clinical outcomes in former preterm infants with bronchopulmonary dysplasia (BPD). Oxidant stress disrupts alveolar and vascular development in models of BPD. Bleomycin causes oxidative stress and induces BPD and PAH in neonatal rats. Disruption in the vascular endothelial growth factor (VEGF) and nitric oxide signaling pathways contributes to BPD. We hypothesized that loss of EC-SOD would worsen PAH associated with BPD in a neonatal mouse model of bleomycin-induced BPD by disrupting the VEGF/NO signaling pathway. METHODS: Neonatal wild-type mice (WT), and mice lacking EC-SOD (EC-SOD KO) received intraperitoneal bleomycin (2 units/kg) or phosphate-buffered saline (PBS) three times weekly and were evaluated at weeks 3 or 4. RESULTS: Lack of EC-SOD impaired alveolar development and resulted in PH (elevated right ventricular systolic pressures, right ventricular hypertrophy (RVH)), decreased vessel density, and increased small vessel muscularization. Exposure to bleomycin further impaired alveolar development, worsened RVH and vascular remodeling. Lack of EC-SOD and bleomycin treatment decreased lung total and phosphorylated VEGFR2 and eNOS protein expression. CONCLUSION: EC-SOD is critical in preserving normal lung development and loss of EC-SOD results in disrupted alveolar development, PAH and vascular remodeling at baseline, which is further worsened with bleomycin and associated with decreased activation of VEGFR2.


Assuntos
Bleomicina , Displasia Broncopulmonar/enzimologia , Células Endoteliais/enzimologia , Hipertensão Pulmonar/enzimologia , Alvéolos Pulmonares/irrigação sanguínea , Alvéolos Pulmonares/enzimologia , Artéria Pulmonar/enzimologia , Superóxido Dismutase/deficiência , Remodelação Vascular , Animais , Animais Recém-Nascidos , Displasia Broncopulmonar/induzido quimicamente , Displasia Broncopulmonar/genética , Displasia Broncopulmonar/patologia , Displasia Broncopulmonar/fisiopatologia , Células Endoteliais/patologia , Predisposição Genética para Doença , Hipertensão Pulmonar/induzido quimicamente , Hipertensão Pulmonar/genética , Hipertensão Pulmonar/patologia , Hipertensão Pulmonar/fisiopatologia , Hipertrofia Ventricular Direita/induzido quimicamente , Hipertrofia Ventricular Direita/enzimologia , Hipertrofia Ventricular Direita/genética , Hipertrofia Ventricular Direita/fisiopatologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Estresse Oxidativo , Fenótipo , Fosforilação , Alvéolos Pulmonares/patologia , Artéria Pulmonar/patologia , Artéria Pulmonar/fisiopatologia , Transdução de Sinais , Superóxido Dismutase/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Disfunção Ventricular Direita/induzido quimicamente , Disfunção Ventricular Direita/enzimologia , Disfunção Ventricular Direita/genética , Disfunção Ventricular Direita/fisiopatologia , Função Ventricular Direita , Pressão Ventricular
13.
Am J Respir Crit Care Med ; 190(12): 1402-12, 2014 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-25358054

RESUMO

RATIONALE: Idiopathic pulmonary fibrosis (IPF) is a deadly lung disease with few therapeutic options. Apoptosis of alveolar epithelial cells, followed by abnormal tissue repair characterized by hyperplastic epithelial cell formation, is a pathogenic process that contributes to the progression of pulmonary fibrosis. However, the signaling pathways responsible for increased proliferation of epithelial cells remain poorly understood. OBJECTIVES: To investigate the role of deoxycytidine kinase (DCK), an important enzyme for the salvage of deoxynucleotides, in the progression of pulmonary fibrosis. METHODS: DCK expression was examined in the lungs of patients with IPF and mice exposed to bleomycin. The regulation of DCK expression by hypoxia was studied in vitro and the importance of DCK in experimental pulmonary fibrosis was examined using a DCK inhibitor and alveolar epithelial cell-specific knockout mice. MEASUREMENTS AND MAIN RESULTS: DCK was elevated in hyperplastic alveolar epithelial cells of patients with IPF and in mice exposed to bleomycin. Increased DCK was localized to cells associated with hypoxia, and hypoxia directly induced DCK in alveolar epithelial cells in vitro. Hypoxia-induced DCK expression was abolished by silencing hypoxia-inducible factor 1α and treatment of bleomycin-exposed mice with a DCK inhibitor attenuated pulmonary fibrosis in association with decreased epithelial cell proliferation. Furthermore, DCK expression, and proliferation of epithelial cells and pulmonary fibrosis was attenuated in mice with conditional deletion of hypoxia-inducible factor 1α in the alveolar epithelium. CONCLUSIONS: Our findings suggest that the induction of DCK after hypoxia plays a role in the progression of pulmonary fibrosis by contributing to alveolar epithelial cell proliferation.


Assuntos
Desoxicitidina Quinase/fisiologia , Hipóxia/complicações , Fibrose Pulmonar Idiopática/etiologia , Animais , Linhagem Celular , Proliferação de Células/fisiologia , Humanos , Hipóxia/enzimologia , Hipóxia/fisiopatologia , Fibrose Pulmonar Idiopática/enzimologia , Fibrose Pulmonar Idiopática/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Alvéolos Pulmonares/enzimologia , Alvéolos Pulmonares/fisiopatologia , Reação em Cadeia da Polimerase em Tempo Real , Mucosa Respiratória/enzimologia
14.
Am J Respir Cell Mol Biol ; 50(3): 526-37, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24093724

RESUMO

Transepithelial sodium transport via alveolar epithelial Na(+) channels (ENaC) and Na(+),K(+)-ATPase constitutes the driving force for removal of alveolar edema fluid. Alveolar hypoxia associated with pulmonary edema may impair ENaC activity and alveolar Na(+) absorption through a decrease of ENaC subunit expression at the apical membrane of alveolar epithelial cells (AECs). Here, we investigated the mechanism(s) involved in this process in vivo in the ß-Liddle mouse strain mice carrying a truncation of ß-ENaC C-terminus abolishing the interaction between ß-ENaC and the ubiquitin protein-ligase Nedd4-2 that targets the channel for endocytosis and degradation and in vitro in rat AECs. Hypoxia (8% O2 for 24 h) reduced amiloride-sensitive alveolar fluid clearance by 69% in wild-type mice but had no effect in homozygous mutated ß-Liddle littermates. In vitro, acute exposure of AECs to hypoxia (0.5-3% O2 for 1-6 h) rapidly decreased transepithelial Na(+) transport as assessed by equivalent short-circuit current Ieq and the amiloride-sensitive component of Na(+) current across the apical membrane, reflecting ENaC activity. Hypoxia induced a decrease of ENaC subunit expression in the apical membrane of AECs with no change in intracellular expression and induced a 2-fold increase in α-ENaC polyubiquitination. Hypoxic inhibition of amiloride-sensitive Ieq was fully prevented by preincubation with the proteasome inhibitors MG132 and lactacystin or with the antioxidant N-acetyl-cysteine. Our data strongly suggest that Nedd4-2-mediated ubiquitination of ENaC leading to endocytosis and degradation of apical Na(+) channels is a key feature of hypoxia-induced inhibition of transepithelial alveolar Na(+) transport.


Assuntos
Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Células Epiteliais/enzimologia , Canais Epiteliais de Sódio/metabolismo , Hipóxia/enzimologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Alvéolos Pulmonares/enzimologia , Sódio/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitina/metabolismo , Animais , Antioxidantes/farmacologia , Hipóxia Celular , Células Cultivadas , Modelos Animais de Doenças , Endocitose , Células Epiteliais/efeitos dos fármacos , Canais Epiteliais de Sódio/deficiência , Canais Epiteliais de Sódio/efeitos dos fármacos , Canais Epiteliais de Sódio/genética , Hipóxia/genética , Masculino , Potenciais da Membrana , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Depuração Mucociliar , Ubiquitina-Proteína Ligases Nedd4 , Inibidores de Proteassoma/farmacologia , Alvéolos Pulmonares/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Bloqueadores dos Canais de Sódio/farmacologia , Fatores de Tempo
15.
J Biol Chem ; 288(36): 25964-25975, 2013 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-23902766

RESUMO

Idiopathic pulmonary fibrosis is a devastating lung disorder of unknown etiology. Although its pathogenesis is unclear, considerable evidence supports an important role of aberrantly activated alveolar epithelial cells (AECs), which produce a large variety of mediators, including several matrix metalloproteases (MMPs), which participate in fibroblast activation and lung remodeling. MMP-1 has been shown to be highly expressed in AECs from idiopathic pulmonary fibrosis lungs although its role is unknown. In this study, we explored the role of MMP-1 in several AECs functions. Mouse lung epithelial cells (MLE12) transfected with human Mmp-1 showed significantly increased cell growth and proliferation at 36 and 48 h of culture (p < 0.01). Also, MMP-1 promoted MLE12 cell migration through collagen I, accelerated wound closing, and protected cells from staurosporine- and bleomycin-induced apoptosis compared with mock cells (p < 0.01). MLE12 cells expressing human MMP-1 showed a significant repression of oxygen consumption ratio compared with the cells with the empty vector. As under hypoxic conditions hypoxia-inducible factor-1α (HIF-1α) mediates a transition from oxidative to glycolytic metabolism, we analyzed activation of HIF-1α. Ηigher activation of this factor was detected in MMP-1-transfected cells under normoxia and hypoxia. Likewise, a significant decrease of both total and mitochondrial reactive oxygen species was observed in MMP-1-transfected cells. Paralleling these findings, attenuation of MMP-1 expression by shRNA in A549 (human) AECs markedly reduced proliferation and migration (p < 0.01) and increased the oxygen consumption ratio. These findings indicate that epithelial expression of MMP-1 inhibits mitochondrial function, increases HIF-1α expression, decreases reactive oxygen species production, and contributes to a proliferative, migratory, and anti-apoptotic AEC phenotype.


Assuntos
Apoptose/fisiologia , Movimento Celular/fisiologia , Proliferação de Células , Células Epiteliais/enzimologia , Metaloproteinase 13 da Matriz/biossíntese , Metaloproteinase 1 da Matriz/biossíntese , Mitocôndrias/metabolismo , Consumo de Oxigênio/fisiologia , Alvéolos Pulmonares/enzimologia , Mucosa Respiratória/enzimologia , Animais , Antibióticos Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Bleomicina/farmacologia , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/genética , Inibidores Enzimáticos/farmacologia , Células Epiteliais/citologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/genética , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/biossíntese , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Metaloproteinase 1 da Matriz/genética , Metaloproteinase 13 da Matriz/genética , Camundongos , Mitocôndrias/genética , Consumo de Oxigênio/efeitos dos fármacos , Alvéolos Pulmonares/citologia , Mucosa Respiratória/citologia , Estaurosporina/farmacologia
16.
Am J Physiol Lung Cell Mol Physiol ; 307(6): L435-48, 2014 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-25085625

RESUMO

Previous observations made by our laboratory indicate that Bruton's tyrosine kinase (Btk) may play an important role in the pathophysiology of local inflammation in acute lung injury (ALI)/acute respiratory distress syndrome (ARDS). We have shown that there is cross talk between FcγRIIa and TLR4 in alveolar neutrophils from patients with ALI/ARDS and that Btk mediates the molecular cooperation between these two receptors. To study the function of Btk in vivo we have developed a unique two-hit model of ALI: LPS/immune complex (IC)-induced ALI. Furthermore, we conjugated F(ab)2 fragments of anti-neutrophil antibodies (Ly6G1A8) with specific siRNA for Btk to silence Btk specifically in alveolar neutrophils. It should be stressed that we are the first group to perform noninvasive transfections of neutrophils, both in vitro and in vivo. Importantly, our present findings indicate that silencing Btk in alveolar neutrophils has a dramatic protective effect in mice with LPS/IC-induced ALI, and that Btk regulates neutrophil survival and clearance of apoptotic neutrophils in this model. In conclusion, we put forward a hypothesis that Btk-targeted neutrophil specific therapy is a valid goal of research geared toward restoring homeostasis in lungs of patients with ALI/ARDS.


Assuntos
Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/enzimologia , Complexo Antígeno-Anticorpo/toxicidade , Inativação Gênica , Lipopolissacarídeos/toxicidade , Neutrófilos/enzimologia , Proteínas Tirosina Quinases/metabolismo , Alvéolos Pulmonares/enzimologia , Lesão Pulmonar Aguda/genética , Lesão Pulmonar Aguda/patologia , Tirosina Quinase da Agamaglobulinemia , Animais , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Neutrófilos/patologia , Proteínas Tirosina Quinases/genética , Alvéolos Pulmonares/patologia , Receptores de IgG/genética , Receptores de IgG/metabolismo , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo
17.
Am J Pathol ; 183(4): 1169-1182, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23933064

RESUMO

Bronchopulmonary dysplasia of the premature newborn is characterized by lung injury, resulting in alveolar simplification and reduced pulmonary function. Exposure of neonatal mice to hyperoxia enhanced sphingosine-1-phosphate (S1P) levels in lung tissues; however, the role of increased S1P in the pathobiological characteristics of bronchopulmonary dysplasia has not been investigated. We hypothesized that an altered S1P signaling axis, in part, is responsible for neonatal lung injury leading to bronchopulmonary dysplasia. To validate this hypothesis, newborn wild-type, sphingosine kinase1(-/-) (Sphk1(-/-)), sphingosine kinase 2(-/-) (Sphk2(-/-)), and S1P lyase(+/-) (Sgpl1(+/-)) mice were exposed to hyperoxia (75%) from postnatal day 1 to 7. Sphk1(-/-), but not Sphk2(-/-) or Sgpl1(+/-), mice offered protection against hyperoxia-induced lung injury, with improved alveolarization and alveolar integrity compared with wild type. Furthermore, SphK1 deficiency attenuated hyperoxia-induced accumulation of IL-6 in bronchoalveolar lavage fluids and NADPH oxidase (NOX) 2 and NOX4 protein expression in lung tissue. In vitro experiments using human lung microvascular endothelial cells showed that exogenous S1P stimulated intracellular reactive oxygen species (ROS) generation, whereas SphK1 siRNA, or inhibitor against SphK1, attenuated hyperoxia-induced S1P generation. Knockdown of NOX2 and NOX4, using specific siRNA, reduced both basal and S1P-induced ROS formation. These results suggest an important role for SphK1-mediated S1P signaling-regulated ROS in the development of hyperoxia-induced lung injury in a murine neonatal model of bronchopulmonary dysplasia.


Assuntos
Displasia Broncopulmonar/enzimologia , Displasia Broncopulmonar/prevenção & controle , Hiperóxia/complicações , Lisofosfolipídeos/metabolismo , Glicoproteínas de Membrana/metabolismo , NADPH Oxidases/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/deficiência , Esfingosina/análogos & derivados , Aldeído Liases/deficiência , Aldeído Liases/metabolismo , Animais , Animais Recém-Nascidos , Displasia Broncopulmonar/etiologia , Displasia Broncopulmonar/patologia , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/enzimologia , Células Endoteliais/patologia , Humanos , Hiperóxia/enzimologia , Hiperóxia/patologia , Camundongos , Camundongos Endogâmicos C57BL , NADPH Oxidase 2 , NADPH Oxidase 4 , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Pneumonia/complicações , Pneumonia/patologia , Alvéolos Pulmonares/enzimologia , Alvéolos Pulmonares/patologia , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Esfingosina/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo
18.
Respir Res ; 15: 34, 2014 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-24661418

RESUMO

BACKGROUND: The formation of discrete elastin bands at the tips of secondary alveolar septa is important for normal alveolar development, but the mechanisms regulating the lung elastogenic program are incompletely understood. JNK suppress elastin synthesis in the aorta and is important in a host of developmental processes. We sought to determine whether JNK suppresses pulmonary fibroblast elastogenesis during lung development. METHODS: Alveolar size, elastin content, and mRNA of elastin-associated genes were quantitated in wild type and JNK-deficient mouse lungs, and expression profiles were validated in primary lung fibroblasts. Tropoelastin protein was quantitated by Western blot. Changes in lung JNK activity throughout development were quantitated, and pJNK was localized by confocal imaging and lineage tracing. RESULTS: By morphometry, alveolar diameters were increased by 7% and lung elastin content increased 2-fold in JNK-deficient mouse lungs compared to wild type. By Western blot, tropoelastin protein was increased 5-fold in JNK-deficient lungs. Postnatal day 14 (PND14) lung JNK activity was 11-fold higher and pJNK:JNK ratio 6-fold higher compared to PN 8 week lung. Lung tropoelastin, emilin-1, fibrillin-1, fibulin-5, and lysyl oxidase mRNAs inversely correlated with lung JNK activity during alveolar development. Phosphorylated JNK localized to pulmonary lipofibroblasts. PND14 JNK-deficient mouse lungs contained 7-fold more tropoelastin, 2,000-fold more emilin-1, 800-fold more fibrillin-1, and 60-fold more fibulin-5 than PND14 wild type lungs. Primarily lung fibroblasts from wild type and JNK-deficient mice showed similar differences in elastogenic mRNAs. CONCLUSIONS: JNK suppresses fibroblast elastogenesis during the alveolar stage of lung development.


Assuntos
Elasticidade/fisiologia , Fibroblastos/enzimologia , Proteína Quinase 8 Ativada por Mitógeno/fisiologia , Alvéolos Pulmonares/enzimologia , Animais , Animais Recém-Nascidos , Células Cultivadas , Ativação Enzimática/fisiologia , Pulmão/citologia , Pulmão/enzimologia , Pulmão/crescimento & desenvolvimento , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Alvéolos Pulmonares/citologia
19.
Mol Cell Biochem ; 394(1-2): 199-208, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24894820

RESUMO

Streptococcus pneumoniae is an important pathogen of pneumonia in human. Human alveolar epithelium acts as an effective barrier and is an active participant in host defense against invasion of bacterial by production of various mediators. Sirtuin 1 (SIRT1), the prototypic class III histone deacetylase, is involved in the molecular control of lifespans and immune responses. This study aimed at examining the role of SIRT1 in mediating S. pneumoniae-induced human ß-defensin-2 (hBD2) and interleukin-8(IL-8) expression in the alveolar epithelial cell line A549 and the underlying mechanisms involved. A549 cells were infected with S. pneumoniae for indicated times. Exposure of A549 cells to S. pneumoniae increased the expressions of SIRT1 protein, hBD2 and IL-8 mRNA, and protein. The SIRT1 activator resveratrol enhanced S. pneumoniae-induced gene expression of hBD2 but decreased IL-8 mRNA levels. Blockade of SIRT1 activity by the SIRT1 inhibitors nicotinamide reduced S. pneumoniae-induced hBD2 mRNA expression but increased its stimulatory effects on IL-8 mRNA. S. pneumoniae-induced activation of extracellular signal-regulated kinase (ERK) and p38 mitogen-activated protein kinase (MAPK). SIRT1 expression was attenuated by selective inhibitors of ERK and p38 MAPK. The hBD2 mRNA production was decreased by pretreatment with p38 MAPK inhibitor but not with ERK inhibitor, whereas the IL-8 mRNA expression was controlled by phosphorylation of ERK. These results suggest that SIRT1 mediates the induction of hBD2 and IL-8 gene expression levels in A549 cell by S. pneumoniae. SIRT1 may play a key role in host immune and defense response in A549.


Assuntos
Células Epiteliais/enzimologia , Células Epiteliais/microbiologia , Interleucina-8/metabolismo , Alvéolos Pulmonares/enzimologia , Alvéolos Pulmonares/microbiologia , Sirtuína 1/metabolismo , Streptococcus pneumoniae/patogenicidade , beta-Defensinas/metabolismo , Linhagem Celular Tumoral , Ativação Enzimática , Ativadores de Enzimas/farmacologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/imunologia , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Inibidores de Histona Desacetilases/farmacologia , Interações Hospedeiro-Patógeno , Humanos , Interleucina-8/genética , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Alvéolos Pulmonares/efeitos dos fármacos , Alvéolos Pulmonares/imunologia , RNA Mensageiro/metabolismo , Transdução de Sinais , Sirtuína 1/genética , Streptococcus pneumoniae/imunologia , Fatores de Tempo , Regulação para Cima , beta-Defensinas/genética , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
20.
Am J Respir Cell Mol Biol ; 49(5): 721-30, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23590304

RESUMO

Protein phosphatase-2A (PP2A) is a primary serine-threonine phosphatase that modulates inflammatory responses in asthma and chronic obstructive pulmonary disease (COPD). Despite its importance, the mechanisms that regulate lung PP2A activity remain to be determined. The redox-sensitive enzyme protein tyrosine phosphatase-1B (PTP1B) activates PP2A by dephosphorylating the catalytic subunit of the protein at tyrosine 307. This study aimed to identify how the interaction between the intracellular antioxidant glutathione peroxidase-1 (GPx-1) and PTP1B affected lung PP2A activity and airway inflammation. Experiments using gene silencing techniques in mouse lung or human small airway epithelial cells determined that knocking down PTP1B expression blocked GPx-1's activation of PP2A and negated the anti-inflammatory effects of GPx-1 protein in the lung. Similarly, the expression of human GPx-1 in transgenic mice significantly increased PP2A and PTP1B activities and prevented chronic cigarette smoke-induced airway inflammation and alveolar destruction. GPx-1 knockout mice, however, exhibited an exaggerated emphysema phenotype, correlating with a nonresponsive PP2A pathway. Importantly, GPx-1-PTP1B-PP2A signaling becomes inactivated in advanced lung disease. Indeed, PTP1B protein was oxidized in the lungs of subjects with advanced emphysema, and cigarette smoke did not increase GPx-1 or PTP1B activity within epithelial cells isolated from subjects with COPD, unlike samples of healthy lung epithelial cells. In conclusion, these findings establish that the GPx-1-PTP1B-PP2A axis plays a critical role in countering the inflammatory and proteolytic responses that result in lung-tissue destruction in response to cigarette smoke exposure.


Assuntos
Glutationa Peroxidase/metabolismo , Pneumonia/enzimologia , Proteína Fosfatase 2/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 1/metabolismo , Alvéolos Pulmonares/enzimologia , Mucosa Respiratória/enzimologia , Transdução de Sinais , Animais , Estudos de Casos e Controles , Linhagem Celular , Ativação Enzimática , Técnicas de Silenciamento de Genes , Glutationa Peroxidase/deficiência , Glutationa Peroxidase/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Knockout , Oxirredução , Estresse Oxidativo , Fosforilação , Pneumonia/etiologia , Pneumonia/genética , Pneumonia/patologia , Pneumonia/prevenção & controle , Ligação Proteica , Proteína Tirosina Fosfatase não Receptora Tipo 1/genética , Alvéolos Pulmonares/patologia , Doença Pulmonar Obstrutiva Crônica/enzimologia , Doença Pulmonar Obstrutiva Crônica/patologia , Enfisema Pulmonar/enzimologia , Enfisema Pulmonar/patologia , Interferência de RNA , Mucosa Respiratória/patologia , Fumar/efeitos adversos , Transfecção , Glutationa Peroxidase GPX1
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa