Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Am J Respir Cell Mol Biol ; 58(6): 736-744, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29314863

RESUMO

Loss of secretory IgA is common in the small airways of patients with chronic obstructive pulmonary disease and may contribute to disease pathogenesis. Using mice that lack secretory IgA in the airways due to genetic deficiency of polymeric Ig receptor (pIgR-/- mice), we investigated the role of neutrophils in driving the fibrotic small airway wall remodeling and emphysema that develops spontaneously in these mice. By flow cytometry, we found an increase in the percentage of neutrophils among CD45+ cells in the lungs, as well as an increase in total neutrophils, in pIgR-/- mice compared with wild-type controls. This increase in neutrophils in pIgR-/- mice was associated with elastin degradation in the alveolar compartment and around small airways, along with increased collagen deposition in small airway walls. Neutrophil depletion using anti-Ly6G antibodies or treatment with broad-spectrum antibiotics inhibited development of both emphysema and small airway remodeling, suggesting that airway bacteria provide the stimulus for deleterious neutrophilic inflammation in this model. Exogenous bacterial challenge using lysates prepared from pathogenic and nonpathogenic bacteria worsened neutrophilic inflammation and lung remodeling in pIgR-/- mice. This phenotype was abrogated by antiinflammatory therapy with roflumilast. Together, these studies support the concept that disruption of the mucosal immune barrier in small airways contributes to chronic obstructive pulmonary disease progression by allowing bacteria to stimulate chronic neutrophilic inflammation, which, in turn, drives progressive airway wall fibrosis and emphysematous changes in the lung parenchyma.


Assuntos
Remodelação das Vias Aéreas/fisiologia , Neutrófilos/patologia , Pneumonia Bacteriana/patologia , Doença Pulmonar Obstrutiva Crônica/etiologia , Remodelação das Vias Aéreas/efeitos dos fármacos , Aminopiridinas/farmacologia , Animais , Bacillus/patogenicidade , Benzamidas/farmacologia , Ciclopropanos/farmacologia , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Neutrófilos/microbiologia , Doença Pulmonar Obstrutiva Crônica/tratamento farmacológico , Enfisema Pulmonar/patologia , Receptores de Superfície Celular/genética
2.
Chin J Integr Med ; 2016 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-27933509

RESUMO

OBJECTIVE: To observe the clinical efficacy and safety of Shuanghu Qinggan Granule ( , SQG) plus Yigan Yiqi Jieyu Granule (, YYJG) combined with lamivudine (LAM) on chronic hepatitis B (CHB) patients. METHODS: The study was a multicenter, randomized, double-blinded and parallel controlled trial. A total of 320 patients were randomly allocated into 2 groups equally: 160 patients (treatment group) were given SQG and YYJG combined with LAM; and 160 patients (control group) were given LAM plus Chinese herb placebo, respectively. Liver functions, hepatitis B envelop antigen (HBeAg) titer levels, and hepatitis B virus DNA (HBV-DNA) load were monitored. RESULTS: (1) In the 48th week, the treatment group showed superior HBeAg seroconversion rate than that in the control group (38.0% vs. 24.0%, P<0.05). (2) In the 48th week, the treatment group demonstrated lower HBeAg titer than that in the control group (P<0.05). (3) In the 12th, 24th, 48th week, there was no statistical significance in HBV-DNA response rate between the two groups. (4) In the 12th week, the level of glutamyl transpeptidase (GGT) was significantly decreased in the treatment group compared with the control group (P<0.05); in the 36th week, the levels of alanine aminotransferase and aspartate transaminase were significantly lower in the treatment group than those in the control group (P<0.05). CONCLUSION: The protocol of SQG and YYJG combined with LAM to treat CHB showed superior efficacy than LAM monotherapy.

3.
Oncoimmunology ; 5(6): e1168549, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27471643

RESUMO

Several studies have demonstrated that NF-κB activation is common in lung cancer; however, the mechanistic links between NF-κB signaling and tumorigenesis remain to be fully elucidated. We investigated the function of NF-κB signaling in epidermal growth factor receptor (EGFR)-mutant lung tumors using a transgenic mouse model with doxycycline (dox)-inducible expression of oncogenic EGFR in the lung epithelium with or without a dominant inhibitor of NF-κB signaling. NF-κB inhibition resulted in a significant reduction in tumor burden in both EGFR tyrosine kinase inhibitor (TKI)-sensitive and resistant tumors. However, NF-κB inhibition did not alter epithelial cell survival in vitro or in vivo, and no changes were detected in activation of EGFR downstream signaling pathways. Instead, we observed an influx of inflammatory cells (macrophages and neutrophils) in the lungs of mice with oncogenic EGFR expression that was blocked in the setting of NF-κB inhibition. To investigate whether inflammatory cells play a role in promoting EGFR-mutant lung tumors, we depleted macrophages and neutrophils during tumorigenesis and found that neutrophil depletion had no effect on tumor formation, but macrophage depletion caused a significant reduction in tumor burden. Together, these data suggest that epithelial NF-κB signaling supports carcinogenesis in a non-cell autonomous manner in EGFR-mutant tumors through recruitment of pro-tumorigenic macrophages.

4.
Cell Rep ; 16(1): 120-132, 2016 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-27320908

RESUMO

Although epithelial NF-κB signaling is important for lung carcinogenesis, NF-κB inhibitors are ineffective for cancer treatment. To explain this paradox, we studied mice with genetic deletion of IKKß in myeloid cells and found enhanced tumorigenesis in Kras(G12D) and urethane models of lung cancer. Myeloid-specific inhibition of NF-κB augmented pro-IL-1ß processing by cathepsin G in neutrophils, leading to increased IL-1ß and enhanced epithelial cell proliferation. Combined treatment with bortezomib, a proteasome inhibitor that blocks NF-κB activation, and IL-1 receptor antagonist reduced tumor formation and growth in vivo. In lung cancer patients, plasma IL-1ß levels correlated with poor prognosis, and IL-1ß increased following bortezomib treatment. Together, our studies elucidate an important role for neutrophils and IL-1ß in lung carcinogenesis and resistance to NF-κB inhibitors.


Assuntos
Interleucina-1beta/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , NF-kappa B/antagonistas & inibidores , Neutrófilos/metabolismo , Animais , Bortezomib/farmacologia , Bortezomib/uso terapêutico , Carcinogênese/efeitos dos fármacos , Carcinogênese/patologia , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Proliferação de Células/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Humanos , Quinase I-kappa B/metabolismo , Neoplasias Pulmonares/tratamento farmacológico , Camundongos , Células Mieloides/efeitos dos fármacos , Células Mieloides/metabolismo , NF-kappa B/metabolismo , Neutrófilos/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Análise de Sobrevida
5.
Nat Commun ; 7: 11240, 2016 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-27046438

RESUMO

Mechanisms driving persistent airway inflammation in chronic obstructive pulmonary disease (COPD) are incompletely understood. As secretory immunoglobulin A (SIgA) deficiency in small airways has been reported in COPD patients, we hypothesized that immunobarrier dysfunction resulting from reduced SIgA contributes to chronic airway inflammation and disease progression. Here we show that polymeric immunoglobulin receptor-deficient (pIgR(-/-)) mice, which lack SIgA, spontaneously develop COPD-like pathology as they age. Progressive airway wall remodelling and emphysema in pIgR(-/-) mice are associated with an altered lung microbiome, bacterial invasion of the airway epithelium, NF-κB activation, leukocyte infiltration and increased expression of matrix metalloproteinase-12 and neutrophil elastase. Re-derivation of pIgR(-/-) mice in germ-free conditions or treatment with the anti-inflammatory phosphodiesterase-4 inhibitor roflumilast prevents COPD-like lung inflammation and remodelling. These findings show that pIgR/SIgA deficiency in the airways leads to persistent activation of innate immune responses to resident lung microbiota, driving progressive small airway remodelling and emphysema.


Assuntos
Envelhecimento/imunologia , Aminopiridinas/farmacologia , Benzamidas/farmacologia , Microbiota/imunologia , Inibidores da Fosfodiesterase 4/farmacologia , Doença Pulmonar Obstrutiva Crônica/imunologia , Enfisema Pulmonar/imunologia , Receptores de Imunoglobulina Polimérica/deficiência , Envelhecimento/patologia , Remodelação das Vias Aéreas/imunologia , Animais , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/genética , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/imunologia , Ciclopropanos/farmacologia , Modelos Animais de Doenças , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno , Humanos , Imunidade Inata , Imunoglobulina A Secretora/genética , Elastase de Leucócito/genética , Elastase de Leucócito/imunologia , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Pulmão/microbiologia , Pulmão/patologia , Metaloproteinase 12 da Matriz/genética , Metaloproteinase 12 da Matriz/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NF-kappa B/genética , NF-kappa B/imunologia , Doença Pulmonar Obstrutiva Crônica/tratamento farmacológico , Doença Pulmonar Obstrutiva Crônica/genética , Doença Pulmonar Obstrutiva Crônica/microbiologia , Enfisema Pulmonar/tratamento farmacológico , Enfisema Pulmonar/genética , Enfisema Pulmonar/microbiologia , Receptores de Imunoglobulina Polimérica/genética , Receptores de Imunoglobulina Polimérica/imunologia , Mucosa Respiratória/efeitos dos fármacos , Mucosa Respiratória/imunologia , Mucosa Respiratória/microbiologia , Mucosa Respiratória/patologia
6.
J Immunol ; 196(4): 1891-9, 2016 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-26773153

RESUMO

Although numerous studies have demonstrated a critical role for canonical NF-κB signaling in inflammation and disease, the function of the noncanonical NF-κB pathway remains ill-defined. In lung tissue from patients with acute respiratory distress syndrome, we identified increased expression of the noncanonical pathway component p100/p52. To investigate the effects of p52 expression in vivo, we generated a novel transgenic mouse model with inducible expression of p52 in Clara cell secretory protein-expressing airway epithelial cells. Although p52 overexpression alone did not cause significant inflammation, p52 overexpression caused increased lung inflammation, injury, and mortality following intratracheal delivery of Escherichia coli LPS. No differences in cytokine/chemokine expression were measured between p52-overexpressing mice and controls, but increased apoptosis of Clara cell secretory protein-positive airway epithelial cells was observed in transgenic mice after LPS stimulation. In vitro studies in lung epithelial cells showed that p52 overexpression reduced cell survival and increased the expression of several proapoptotic genes during cellular stress. Collectively, these studies demonstrate a novel role for p52 in cell survival/apoptosis of airway epithelial cells and implicate noncanonical NF-κB signaling in the pathogenesis of acute respiratory distress syndrome.


Assuntos
Apoptose/imunologia , Subunidade p52 de NF-kappa B/imunologia , Síndrome do Desconforto Respiratório/patologia , Mucosa Respiratória/patologia , Animais , Western Blotting , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Humanos , Imuno-Histoquímica , Lipopolissacarídeos/toxicidade , Camundongos , Camundongos Transgênicos , Subunidade p52 de NF-kappa B/biossíntese , Pneumonia/imunologia , Pneumonia/patologia , Reação em Cadeia da Polimerase em Tempo Real , Síndrome do Desconforto Respiratório/imunologia , Mucosa Respiratória/imunologia , Transdução de Sinais/imunologia , Regulação para Cima
8.
Am J Respir Crit Care Med ; 191(6): 646-55, 2015 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-25607374

RESUMO

RATIONALE: Up to 20% of cases of idiopathic interstitial pneumonia cluster in families, comprising the syndrome of familial interstitial pneumonia (FIP); however, the genetic basis of FIP remains uncertain in most families. OBJECTIVES: To determine if new disease-causing rare genetic variants could be identified using whole-exome sequencing of affected members from FIP families, providing additional insights into disease pathogenesis. METHODS: Affected subjects from 25 kindreds were selected from an ongoing FIP registry for whole-exome sequencing from genomic DNA. Candidate rare variants were confirmed by Sanger sequencing, and cosegregation analysis was performed in families, followed by additional sequencing of affected individuals from another 163 kindreds. MEASUREMENTS AND MAIN RESULTS: We identified a potentially damaging rare variant in the gene encoding for regulator of telomere elongation helicase 1 (RTEL1) that segregated with disease and was associated with very short telomeres in peripheral blood mononuclear cells in 1 of 25 families in our original whole-exome sequencing cohort. Evaluation of affected individuals in 163 additional kindreds revealed another eight families (4.7%) with heterozygous rare variants in RTEL1 that segregated with clinical FIP. Probands and unaffected carriers of these rare variants had short telomeres (<10% for age) in peripheral blood mononuclear cells and increased T-circle formation, suggesting impaired RTEL1 function. CONCLUSIONS: Rare loss-of-function variants in RTEL1 represent a newly defined genetic predisposition for FIP, supporting the importance of telomere-related pathways in pulmonary fibrosis.


Assuntos
DNA Helicases/genética , Doenças Pulmonares Intersticiais/genética , Idoso , Idoso de 80 Anos ou mais , Feminino , Variação Genética , Heterozigoto , Humanos , Pulmão/patologia , Doenças Pulmonares Intersticiais/patologia , Masculino , Pessoa de Meia-Idade , Linhagem , Telômero/genética
9.
Am J Respir Crit Care Med ; 191(4): 417-26, 2015 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-25389906

RESUMO

RATIONALE: Asymptomatic relatives of patients with familial interstitial pneumonia (FIP), the inherited form of idiopathic interstitial pneumonia, carry increased risk for developing interstitial lung disease. OBJECTIVES: Studying these at-risk individuals provides a unique opportunity to investigate early stages of FIP pathogenesis and develop predictive models of disease onset. METHODS: Seventy-five asymptomatic first-degree relatives of FIP patients (mean age, 50.8 yr) underwent blood sampling and high-resolution chest computed tomography (HRCT) scanning in an ongoing cohort study; 72 consented to bronchoscopy with bronchoalveolar lavage (BAL) and transbronchial biopsies. Twenty-seven healthy individuals were used as control subjects. MEASUREMENTS AND MAIN RESULTS: Eleven of 75 at-risk subjects (14%) had evidence of interstitial changes by HRCT, whereas 35.2% had abnormalities on transbronchial biopsies. No differences were noted in inflammatory cells in BAL between at-risk individuals and control subjects. At-risk subjects had increased herpesvirus DNA in cell-free BAL and evidence of herpesvirus antigen expression in alveolar epithelial cells (AECs), which correlated with expression of endoplasmic reticulum stress markers in AECs. Peripheral blood mononuclear cell and AEC telomere length were shorter in at-risk individuals than healthy control subjects. The minor allele frequency of the Muc5B rs35705950 promoter polymorphism was increased in at-risk subjects. Levels of several plasma biomarkers differed between at-risk subjects and control subjects, and correlated with abnormal HRCT scans. CONCLUSIONS: Evidence of lung parenchymal remodeling and epithelial dysfunction was identified in asymptomatic individuals at risk for FIP. Together, these findings offer new insights into the early pathogenesis of idiopathic interstitial pneumonia and provide an ongoing opportunity to characterize presymptomatic abnormalities that predict progression to clinical disease.


Assuntos
Doenças Pulmonares Intersticiais/diagnóstico , Fenótipo , Adulto , Idoso , Doenças Assintomáticas , Biomarcadores/metabolismo , Biópsia , Lavagem Broncoalveolar , Broncoscopia , Estudos de Casos e Controles , DNA Viral/análise , Feminino , Frequência do Gene , Marcadores Genéticos , Herpesviridae/genética , Herpesviridae/isolamento & purificação , Humanos , Pulmão/diagnóstico por imagem , Pulmão/metabolismo , Pulmão/patologia , Pulmão/virologia , Doenças Pulmonares Intersticiais/genética , Doenças Pulmonares Intersticiais/metabolismo , Doenças Pulmonares Intersticiais/virologia , Masculino , Pessoa de Meia-Idade , Mucina-5B/genética , Polimorfismo Genético , Estudos Prospectivos , Tomografia Computadorizada por Raios X
10.
Am J Physiol Renal Physiol ; 304(10): F1295-307, 2013 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-23486012

RESUMO

Aquaporin 11 (AQP11) is a newly described member of the protein family of transport channels. AQP11 associates with the endoplasmic reticulum (ER) and is highly expressed in proximal tubular epithelial cells in the kidney. Previously, we identified and characterized a recessive mutation of the highly conserved Cys227 to Ser227 in mouse AQP11 that caused proximal tubule (PT) injury and kidney failure in mutant mice. The current study revealed induction of ER stress, unfolded protein response, and apoptosis as molecular mechanisms of this PT injury. Cys227Ser mutation interfered with maintenance of AQP11 oligomeric structure. AQP11 is abundantly expressed in the S1 PT segment, a site of major renal glucose flux, and Aqp11 mutant mice developed PT-specific mitochondrial injury. Glucose increased AQP11 protein expression in wild-type kidney and upregulation of AQP11 expression by glucose in vitro was prevented by phlorizin, an inhibitor of sodium-dependent glucose transport across PT. Total AQP11 levels in heterozygotes were higher than in wild-type mice but were not further increased in response to glucose. In Aqp11 insufficient PT cells, glucose potentiated increases in reactive oxygen species (ROS) production. ROS production was also elevated in Aqp11 mutation carriers. Phenotypically normal mice heterozygous for the Aqp11 mutation repeatedly treated with glucose showed increased blood urea nitrogen levels that were prevented by the antioxidant sulforaphane or by phlorizin. Our results indicate an important role for AQP11 to prevent glucose-induced oxidative stress in proximal tubules.


Assuntos
Aquaporinas/genética , Retículo Endoplasmático/metabolismo , Rim/metabolismo , Estresse Oxidativo/genética , Insuficiência Renal/genética , Animais , Aquaporinas/metabolismo , Linhagem Celular , Estresse do Retículo Endoplasmático/fisiologia , Camundongos , Mutação , Espécies Reativas de Oxigênio/metabolismo , Insuficiência Renal/metabolismo , Regulação para Cima
11.
Am J Respir Crit Care Med ; 187(6): 630-9, 2013 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-23306543

RESUMO

RATIONALE: Alveolar epithelial cells (AECs) play central roles in the response to lung injury and the pathogenesis of pulmonary fibrosis. OBJECTIVES: We aimed to determine the role of ß-catenin in alveolar epithelium during bleomycin-induced lung fibrosis. METHODS: Genetically modified mice were developed to selectively delete ß-catenin in AECs and were crossed to cell fate reporter mice that express ß-galactosidase (ßgal) in cells of AEC lineage. Mice were given intratracheal bleomycin (0.04 units) and assessed for AEC death, inflammation, lung injury, and fibrotic remodeling. Mouse lung epithelial cells (MLE12) with small interfering RNA knockdown of ß-catenin underwent evaluation for wound closure, proliferation, and bleomycin-induced cytotoxicity. MEASUREMENTS AND MAIN RESULTS: Increased ß-catenin expression was noted in lung parenchyma after bleomycin. Mice with selective deletion of ß-catenin in AECs had greater AEC death at 1 week after bleomycin, followed by increased numbers of fibroblasts and enhanced lung fibrosis as determined by semiquantitative histological scoring and total collagen content. However, no differences in lung inflammation or protein levels in bronchoalveolar lavage were noted. In vitro, ß-catenin-deficient AECs showed increased bleomycin-induced cytotoxicity as well as reduced proliferation and impaired wound closure. Consistent with these findings, mice with AEC ß-catenin deficiency showed delayed recovery after bleomycin. CONCLUSIONS: ß-Catenin in the alveolar epithelium protects against bleomycin-induced fibrosis. Our studies suggest that AEC survival and wound healing are enhanced through ß-catenin-dependent mechanisms. Activation of the developmentally important ß-catenin pathway in AECs appears to contribute to epithelial repair after epithelial injury.


Assuntos
Lesão Pulmonar/patologia , Alvéolos Pulmonares/fisiologia , Fibrose Pulmonar/patologia , beta Catenina/fisiologia , Animais , Bleomicina/efeitos adversos , Modelos Animais de Doenças , Epitélio , Marcação In Situ das Extremidades Cortadas , Lesão Pulmonar/induzido quimicamente , Camundongos , Camundongos Transgênicos , Fibrose Pulmonar/induzido quimicamente , Cicatrização/fisiologia
12.
Carcinogenesis ; 33(4): 859-67, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22287559

RESUMO

Since recent evidence indicates a requirement for epithelial nuclear factor (NF)-κB signaling in lung tumorigenesis, we investigated the impact of the NF-κB inhibitor bortezomib on lung tumor promotion and growth. We used an experimental model in which wild-type mice or mice expressing an NF-κB reporter received intraperitoneal urethane (1 g/kg) followed by twice weekly bortezomib (1 mg/kg) during distinct periods of tumor initiation/progression. Mice were serially assessed for lung NF-κB activation, inflammation and carcinogenesis. Short-term proteasome inhibition with bortezomib did not impact tumor formation but retarded the growth of established lung tumors in mice via effects on cell proliferation. In contrast, long-term treatment with bortezomib resulted in significantly increased lung tumor number and size. This tumor-promoting effect of prolonged bortezomib treatment was associated with perpetuation of urethane-induced inflammation and chronic upregulation of interleukin-1ß and proinflammatory C-X-C motif chemokine ligands (CXCL) 1 and 2 in the lungs. In addition to airway epithelium, bortezomib inhibited NF-κB in pulmonary macrophages in vivo, presenting a possible mechanism of tumor amplification. In this regard, RAW264.7 macrophages exposed to bortezomib showed increased expression of interleukin-1ß, CXCL1 and CXCL2. In conclusion, although short-term bortezomib may exert some beneficial effects, prolonged NF-κB inhibition accelerates chemical lung carcinogenesis by perpetuating carcinogen-induced inflammation. Inhibition of NF-κB in pulmonary macrophages appears to play an important role in this adverse process.


Assuntos
Antineoplásicos/farmacologia , Ácidos Borônicos/farmacologia , Neoplasias Pulmonares/patologia , NF-kappa B/antagonistas & inibidores , Pirazinas/farmacologia , Animais , Bortezomib , Linhagem Celular , Linhagem Celular Tumoral , Humanos , Neoplasias Pulmonares/induzido quimicamente , Neoplasias Pulmonares/metabolismo , Camundongos , Camundongos Endogâmicos BALB C
13.
Am J Respir Cell Mol Biol ; 46(6): 748-56, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22227563

RESUMO

The incidence of idiopathic pulmonary fibrosis (IPF) increases with age. The mechanisms that underlie the age-dependent risk for IPF are unknown. Based on studies that suggest an association of IPF and γherpesvirus infection, we infected young (2-3 mo) and old (≥18 mo) C57BL/6 mice with the murine γherpesvirus 68. Acute murine γherpesvirus 68 infection in aging mice resulted in severe pneumonitis and fibrosis compared with young animals. Progressive clinical deterioration and lung fibrosis in the late chronic phase of infection was observed exclusively in old mice with diminution of tidal volume. Infected aging mice showed higher expression of transforming growth factor-ß during the acute phase of infection. In addition, aging, infected mice showed elevation of proinflammatory cytokines and the fibrocyte recruitment chemokine, CXCL12, in bronchoalveolar lavage. Analyses of lytic virus infection and virus reactivation indicate that old mice were able to control chronic infection and elicit antivirus immune responses. However, old, infected mice showed a significant increase in apoptotic responses determined by in situ terminal deoxynucleotidyl transferase dUTP nick end labeling assay, levels of caspase-3, and expression of the proapoptotitc molecule, Bcl-2 interacting mediator. Apoptosis of type II lung epithelial cells in aging lungs was accompanied by up-regulation of endoplasmic reticulum stress marker, binding immunoglobulin protein, and splicing of X-box-binding protein 1. These results indicate that the aging lung is more susceptible to injury and fibrosis associated with endoplasmic reticulum stress, apoptosis of type II lung epithelial cells, and activation of profibrotic pathways.


Assuntos
Retículo Endoplasmático/metabolismo , Fibrose Pulmonar/metabolismo , Estresse Fisiológico , Animais , Apoptose , Western Blotting , Líquido da Lavagem Broncoalveolar , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase em Tempo Real , Fator de Crescimento Transformador beta/metabolismo
14.
J Biol Chem ; 286(35): 30972-30980, 2011 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-21757695

RESUMO

Expression of mutant surfactant protein C (SFTPC) results in endoplasmic reticulum (ER) stress in type II alveolar epithelial cells (AECs). AECs have been implicated as a source of lung fibroblasts via epithelial-to-mesenchymal transition (EMT); therefore, we investigated whether ER stress contributes to EMT as a possible mechanism for fibrotic remodeling. ER stress was induced by tunicamyin administration or stable expression of mutant (L188Q) SFTPC in type II AEC lines. Both tunicamycin treatment and mutant SFTPC expression induced ER stress and the unfolded protein response. With tunicamycin or mutant SFTPC expression, phase contrast imaging revealed a change to a fibroblast-like appearance. During ER stress, expression of epithelial markers E-cadherin and Zonula occludens-1 decreased while expression of mesenchymal markers S100A4 and α-smooth muscle actin increased. Following induction of ER stress, we found activation of a number of pathways, including MAPK, Smad, ß-catenin, and Src kinase. Using specific inhibitors, the combination of a Smad2/3 inhibitor (SB431542) and a Src kinase inhibitor (PP2) blocked EMT with maintenance of epithelial appearance and epithelial marker expression. Similar results were noted with siRNA targeting Smad2 and Src kinase. Together, these studies reveal that induction of ER stress leads to EMT in lung epithelial cells, suggesting possible cross-talk between Smad and Src kinase pathways. Dissecting pathways involved in ER stress-induced EMT may lead to new treatment strategies to limit fibrosis.


Assuntos
Retículo Endoplasmático/metabolismo , Epitélio/metabolismo , Mesoderma/metabolismo , Alvéolos Pulmonares/citologia , Acetilcisteína/metabolismo , Animais , Caderinas/biossíntese , Fibrose , Humanos , Proteínas de Membrana/biossíntese , Camundongos , Microscopia de Contraste de Fase/métodos , Modelos Biológicos , Mutação , Fosfoproteínas/biossíntese , Alvéolos Pulmonares/metabolismo , Proteína C Associada a Surfactante Pulmonar/metabolismo , Ratos , Tunicamicina/farmacologia , Proteína da Zônula de Oclusão-1
15.
Proc Natl Acad Sci U S A ; 108(26): 10562-7, 2011 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-21670280

RESUMO

Evidence of endoplasmic reticulum (ER) stress has been found in lungs of patients with familial and sporadic idiopathic pulmonary fibrosis. We tested whether ER stress causes or exacerbates lung fibrosis by (i) conditional expression of a mutant form of surfactant protein C (L188Q SFTPC) found in familial interstitial pneumonia and (ii) intratracheal treatment with the protein misfolding agent tunicamycin. We developed transgenic mice expressing L188Q SFTPC exclusively in type II alveolar epithelium by using the Tet-On system. Expression of L188Q SFTPC induced ER stress, as determined by increased expression of heavy-chain Ig binding protein (BiP) and splicing of X-box binding protein 1 (XBP1) mRNA, but no lung fibrosis was identified in the absence of a second profibrotic stimulus. After intratracheal bleomycin, L188Q SFTPC-expressing mice developed exaggerated lung fibrosis and reduced static lung compliance compared with controls. Bleomycin-treated L188Q SFTPC mice also demonstrated increased apoptosis of alveolar epithelial cells and greater numbers of fibroblasts in the lungs. With a complementary model, intratracheal tunicamycin treatment failed to induce lung remodeling yet resulted in augmentation of bleomycin-induced fibrosis. These data support the concept that ER stress produces a dysfunctional epithelial cell phenotype that facilitates fibrotic remodeling. ER stress pathways may serve as important therapeutic targets in idiopathic pulmonary fibrosis.


Assuntos
Retículo Endoplasmático/metabolismo , Pulmão/patologia , Fibrose Pulmonar/patologia , Animais , Apoptose/genética , Bleomicina/toxicidade , Peptídeos e Proteínas de Sinalização Intercelular , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Camundongos , Camundongos Transgênicos , Mutação , Peptídeos/genética , Fibrose Pulmonar/genética , Fibrose Pulmonar/metabolismo , Proteína C Associada a Surfactante Pulmonar , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tunicamicina/toxicidade
16.
J Cell Physiol ; 222(1): 73-81, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19746431

RESUMO

We investigated whether nuclear factor kappa B (NF-kappaB), which exhibits a regulated pattern of activity during murine mammary gland development, plays an important role during lactation and involution, when milk production ceases and the gland undergoes apoptosis and re-modeling. We generated a doxycycline inducible transgenic mouse model to activate NF-kappaB specifically in the mammary epithelium through expression of a constitutively active form of IKK2, the upstream kinase in the classical NF-kappaB signaling cascade. We found that activation of NF-kappaB during involution resulted in a more rapid reduction in milk levels and increased cleavage of caspase-3, an indicator of apoptosis. We also found that activation of NF-kappaB during lactation with no additional involution signals had a similar effect. The observation that NF-kappaB is a key regulator of milk production led us to investigate the role of NF-kappaB during mastitis, an infection of the mammary gland in which milk loss is observed. Mammary gland injection of E. coli LPS resulted in activation of NF-kappaB and milk loss during lactation. This milk loss was decreased by selective inhibition of NF-kappaB in mammary epithelium. Together, our data reveal that activation of NF-kappaB leads to milk clearance in the lactating mammary gland. Therefore, targeting of NF-kappaB signaling may prove therapeutic during mastitis in humans and could be beneficial for the dairy industry, where such infections have a major economic impact.


Assuntos
Epitélio/metabolismo , Epitélio/patologia , Glândulas Mamárias Animais/crescimento & desenvolvimento , Glândulas Mamárias Animais/patologia , Mastite/metabolismo , Leite/metabolismo , NF-kappa B/metabolismo , Animais , Apoptose/efeitos dos fármacos , Modelos Animais de Doenças , Doxiciclina/farmacologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Epitélio/efeitos dos fármacos , Feminino , Lactação/efeitos dos fármacos , Lipopolissacarídeos/farmacologia , Mastite/patologia , Camundongos , Camundongos Transgênicos , Leite/efeitos dos fármacos , Proteínas do Leite/metabolismo , NF-kappa B/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos
17.
Ann N Y Acad Sci ; 1137: 264-72, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18837958

RESUMO

HBV genome replication intermediates blocked at early stages of minus strand synthesis have been identified in a study on circulating DNA and RNA during short-term lamivudine therapy. This suggested that the inhibition of HBV replication processes in the liver are mirrored in the blood. Levels of circulating HBV mRNA remained largely unaffected. Here we followed therapy with two patients (patients 1 and 2) up to stages without apparent replication. As in the earlier study, DNA segments produced successively during replication were used as targets for quantitative PCR: X (early minus strand), C (completed minus strand), and preC (nascent plus strand). Corresponding RNA was quantified by RT/PCR. Polyadenylated viral RNA were assayed as full-length (f) and as truncated (tr) RNAs. Blocked X-region intermediates persisted for about one year. After a period of undetectable HBV DNA viral replication resumed in patient 1 because of the emergence of drug-resistant mutants and in patient 2 because of the discontinuation of therapy. In the former case, X-region intermediates reappeared first, then C- and, finally, preC-region intermediates. Stopping therapy, in contrast, led to a simultaneous reappearance of all three types of intermediates. At low replication levels or its absence, trRNA represented the only polyadenylated viral RNA. Apparently, HBV serum nucleic acid markers allow a study of replication and transcription separately. Specifically, it is concluded (1) that PCR assays for monitoring lamivudine therapy must target the X-gene region and (2) that in the absence of HBV replication, trRNA may constitute a serum marker for HBV expression.


Assuntos
Biomarcadores/sangue , DNA Viral/sangue , Farmacorresistência Viral/genética , Vírus da Hepatite B/genética , Lamivudina/uso terapêutico , Inibidores da Transcriptase Reversa/uso terapêutico , Adulto , DNA Viral/genética , Feminino , Humanos , Masculino , Replicação Viral
18.
Am J Physiol Lung Cell Mol Physiol ; 294(6): L1119-26, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18390830

RESUMO

Recent evidence suggests that dysfunctional type II alveolar epithelial cells (AECs) contribute to the pathogenesis of idiopathic pulmonary fibrosis (IPF). Based on the hypothesis that disease-causing mutations in surfactant protein C (SFTPC) provide an important paradigm for studying IPF, we investigated a potential mechanism of AEC dysfunction suggested to result from mutant SFTPC expression: induction of endoplasmic reticulum (ER) stress and the unfolded protein response (UPR). We evaluated biopsies from 23 IPF patients (including 3 family members with L188Q SFTPC mutations, 10 individuals with familial interstitial pneumonia without SFTPC mutations, and 10 individuals with sporadic IPF) and sections from 10 control lungs. After demonstrating UPR activation in cultured A549 cells expressing mutant SFTPC, we identified prominent expression of UPR markers in AECs in the lungs of patients with SFTPC mutation-associated fibrosis. In individuals with familial interstitial pneumonia without SFTPC mutations and patients with sporadic IPF, we also found UPR activation selectively in AECs lining areas of fibrotic remodeling. Because herpesviruses are found frequently in IPF lungs and can induce ER stress, we investigated expression of viral proteins in lung biopsies. Herpesvirus protein expression was found in AECs from 15/23 IPF patients and colocalized with UPR markers in AECs from these patients. ER stress and UPR activation are found in the alveolar epithelium in patients with IPF and could contribute to disease progression. Activation of these pathways may result from altered surfactant protein processing or chronic herpesvirus infection.


Assuntos
Retículo Endoplasmático/fisiologia , Infecções por Herpesviridae/fisiopatologia , Alvéolos Pulmonares/ultraestrutura , Fibrose Pulmonar/fisiopatologia , Proteína C Associada a Surfactante Pulmonar/fisiologia , Estresse Fisiológico/fisiopatologia , Antígenos Virais/biossíntese , Células Cultivadas , Proteínas de Ligação a DNA/biossíntese , Chaperona BiP do Retículo Endoplasmático , Glicoproteínas/biossíntese , Proteínas de Choque Térmico/biossíntese , Infecções por Herpesviridae/complicações , Humanos , Imuno-Histoquímica , Chaperonas Moleculares/biossíntese , Proteínas Nucleares/biossíntese , Dobramento de Proteína , Fibrose Pulmonar/complicações , Proteína C Associada a Surfactante Pulmonar/genética , Fatores de Transcrição de Fator Regulador X , Fatores de Transcrição , alfa-Manosidase/biossíntese
19.
Proc Natl Acad Sci U S A ; 104(47): 18514-9, 2007 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-18000061

RESUMO

Chronic inflammation is linked to carcinogenesis in several organ systems. In the lungs, NF-kappaB, a central effector of inflammatory responses, is frequently activated in non-small-cell lung cancer, but its role in tumor promotion has not been studied. Several lines of evidence indicate that ethyl carbamate (urethane)-induced lung tumor formation, a prototypical mouse model of multistage lung carcinogenesis, is potentiated by inflammation. We found that mouse strains susceptible to lung tumor formation (FVB, BALB/c) exhibited early NF-kappaB activation and inflammation in the lungs after urethane treatment. However, a resistant strain (C57B6) failed to activate NF-kappaB or induce lung inflammation. In FVB mice, we identified urethane-induced NF-kappaB activation in airway epithelium, as well as type II alveolar epithelial cells and macrophages. Using an inducible transgenic mouse model (FVB strain) to express a dominant inhibitor of NF-kappaB specifically in airway epithelial cells, we found that urethane-induced lung inflammation was blocked and tumor formation was reduced by >50%. Selective NF-kappaB inhibition resulted in increased apoptosis of airway epithelial cells at 2 weeks after urethane treatment in association with a marked reduction of Bcl-2 expression. These studies indicate that NF-kappaB signaling in airway epithelium is integral to tumorigenesis in the urethane model and identify the NF-kappaB pathway as a potential target for chemoprevention of lung cancer.


Assuntos
Transformação Celular Neoplásica/metabolismo , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Neoplasias Pulmonares/metabolismo , NF-kappa B/metabolismo , Uretana/farmacologia , Animais , Transformação Celular Neoplásica/patologia , Suscetibilidade a Doenças , Feminino , Neoplasias Pulmonares/induzido quimicamente , Neoplasias Pulmonares/patologia , Masculino , Camundongos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo
20.
J Immunol ; 178(10): 6504-13, 2007 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-17475880

RESUMO

Although airway epithelial cells provide important barrier and host defense functions, a crucial role for these cells in development of acute lung inflammation and injury has not been elucidated. We investigated whether NF-kappaB pathway signaling in airway epithelium could decisively impact inflammatory phenotypes in the lungs by using a tetracycline-inducible system to achieve selective NF-kappaB activation or inhibition in vivo. In transgenic mice that express a constitutively active form of IkappaB kinase 2 under control of the epithelial-specific CC10 promoter, treatment with doxycycline induced NF-kappaB activation with consequent production of a variety of proinflammatory cytokines, high-protein pulmonary edema, and neutrophilic lung inflammation. Continued treatment with doxycycline caused progressive lung injury and hypoxemia with a high mortality rate. In contrast, inducible expression of a dominant inhibitor of NF-kappaB in airway epithelium prevented lung inflammation and injury resulting from expression of constitutively active form of IkappaB kinase 2 or Escherichia coli LPS delivered directly to the airways or systemically via an osmotic pump implanted in the peritoneal cavity. Our findings indicate that the NF-kappaB pathway in airway epithelial cells is critical for generation of lung inflammation and injury in response to local and systemic stimuli; therefore, targeting inflammatory pathways in airway epithelium could prove to be an effective therapeutic strategy for inflammatory lung diseases.


Assuntos
Pulmão/imunologia , Pulmão/patologia , NF-kappa B/fisiologia , Mucosa Respiratória/imunologia , Mucosa Respiratória/patologia , Transdução de Sinais/imunologia , Doença Aguda , Animais , Células Cultivadas , Feminino , Humanos , Inflamação/genética , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/prevenção & controle , Mediadores da Inflamação/metabolismo , Mediadores da Inflamação/fisiologia , Lipopolissacarídeos/farmacologia , Pulmão/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Mucosa Respiratória/metabolismo , Transdução de Sinais/genética , Traqueia/imunologia , Traqueia/metabolismo , Traqueia/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...