Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Am J Respir Crit Care Med ; 189(4): 463-74, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24325366

RESUMEN

RATIONALE: The death receptor Fas is critical for bacterial clearance and survival of mice after Pseudomonas aeruginosa infection. OBJECTIVES: Fas ligand (FasL)-induced apoptosis is augmented by S-glutathionylation of Fas (Fas-SSG), which can be reversed by glutaredoxin-1 (Grx1). Therefore, the objective of this study was to investigate the interplay between Grx1 and Fas in regulating the clearance of P. aeruginosa infection. METHODS: Lung samples from patients with bronchopneumonia were analyzed by immunofluorescence. Primary tracheal epithelial cells, mice lacking the gene for Grx1 (Glrx1(-/-)), Glrx1(-/-) mice treated with caspase inhibitor, or transgenic mice overexpressing Grx1 in the airway epithelium were analyzed after infection with P. aeruginosa. MEASUREMENTS AND MAIN RESULTS: Patient lung samples positive for P. aeruginosa infection demonstrated increased Fas-SSG compared with normal lung samples. Compared with wild-type primary lung epithelial cells, infection of Glrx1(-/-) cells with P. aeruginosa showed enhanced caspase 8 and 3 activities and cell death in association with increases in Fas-SSG. Infection of Glrx1(-/-) mice with P. aeruginosa resulted in enhanced caspase activity and increased Fas-SSG as compared with wild-type littermates. Absence of Glrx1 significantly enhanced bacterial clearance, and decreased mortality postinfection with P. aeruginosa. Inhibition of caspases significantly decreased bacterial clearance postinfection with P. aeruginosa, in association with decreased Fas-SSG. In contrast, transgenic mice that overexpress Grx1 in lung epithelial cells had significantly higher lung bacterial loads, enhanced mortality, decreased caspase activation, and Fas-SSG in the lung after infection with P. aeruginosa, compared with wild-type control animals. CONCLUSIONS: These results suggest that S-glutathionylation of Fas within the lung epithelium enhances epithelial apoptosis and promotes clearance of P. aeruginosa and that glutaredoxin-1 impairs bacterial clearance and increases the severity of pneumonia in association with deglutathionylation of Fas.


Asunto(s)
Bronconeumonía/metabolismo , Glutarredoxinas/metabolismo , Infecciones por Pseudomonas/metabolismo , Pseudomonas aeruginosa , Receptor fas/metabolismo , Animales , Apoptosis , Carga Bacteriana , Biomarcadores/metabolismo , Bronconeumonía/microbiología , Caspasas/metabolismo , Citocinas/metabolismo , Glutatión/metabolismo , Humanos , Pulmón/metabolismo , Pulmón/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oxidación-Reducción , Infecciones por Pseudomonas/microbiología , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/microbiología , Índice de Severidad de la Enfermedad
2.
Am J Physiol Lung Cell Mol Physiol ; 306(9): L866-75, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24610935

RESUMEN

Chronic allergic asthma leads to airway remodeling and subepithelial fibrosis via mechanisms not fully understood. Airway remodeling is amplified by profibrotic mediators, such as transforming growth factor-ß1 (TGF-ß1), which plays a cardinal role in various models of fibrosis. We recently have identified a critical role for c-Jun-NH2-terminal-kinase (JNK) 1 in augmenting the profibrotic effects of TGF-ß1, linked to epithelial-to-mesenchymal transition of airway epithelial cells. To examine the role of JNK1 in house dust mite (HDM)-induced airway remodeling, we induced allergic airway inflammation in wild-type (WT) and JNK1-/- mice by intranasal administration of HDM extract. WT and JNK1-/- mice were sensitized with intranasal aspirations of HDM extract for 15 days over 3 wk. HDM caused similar increases in airway hyperresponsiveness, mucus metaplasia, and airway inflammation in WT and JNK1-/- mice. In addition, the profibrotic cytokine TGF-ß1 and phosphorylation of Smad3 were equally increased in WT and JNK1-/- mice. In contrast, increases in collagen content in lung tissue induced by HDM were significantly attenuated in JNK1-/- mice compared with WT controls. Furthermore HDM-induced increases of α-smooth muscle actin (α-SMA) protein and mRNA expression as well as the mesenchymal markers high-mobility group AT-hook 2 and collagen1A1 in WT mice were attenuated in JNK1-/- mice. The let-7 family of microRNAs has previously been linked to fibrosis. HDM exposure in WT mice and primary lung epithelial cells resulted in striking decreases in let-7g miRNA that were not observed in mice or primary lung epithelial cells lacking JNK1-/- mice. Overexpression of let-7g in lung epithelial cells reversed the HDM-induced increases in α-SMA. Collectively, these findings demonstrate an important requirement for JNK1 in promoting HDM-induced fibrotic airway remodeling.


Asunto(s)
Remodelación de las Vías Aéreas (Respiratorias) , Hiperreactividad Bronquial/patología , Dermatophagoides pteronyssinus/patogenicidad , Proteínas Quinasas JNK Activadas por Mitógenos/fisiología , Neumonía/patología , Sistema Respiratorio/patología , Animales , Western Blotting , Hiperreactividad Bronquial/etiología , Hiperreactividad Bronquial/metabolismo , Citocinas/genética , Citocinas/metabolismo , Técnicas para Inmunoenzimas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neumonía/etiología , Neumonía/metabolismo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Sistema Respiratorio/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo
3.
Am J Respir Cell Mol Biol ; 46(5): 573-81, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-21799120

RESUMEN

Influenza A virus (IAV) infection is known to induce endoplasmic reticulum (ER) stress, Fas-dependent apoptosis, and TGF-ß production in a variety of cells. However, the relationship between these events in murine primary tracheal epithelial cells (MTECS), which are considered one of the primary sites of IAV infection and replication, is unclear. We show that IAV infection induced ER stress marker activating transcription factor-6 and endoplasmic reticulum protein 57-kD (ERp57), but not C/EBP homologous protein (CHOP). In contrast, the ER stress inducer thapsigargin (THP) increased CHOP. IAV infection activated caspases and apoptosis, independently of Fas and caspase-8, in MTECs. Instead, apoptosis was mediated by caspase-12. A decrease in ERp57 attenuated the IAV burden and decreased caspase-12 activation and apoptosis in epithelial cells. TGF-ß production was enhanced in IAV-infected MTECs, compared with THP or staurosporine. IAV infection caused the activation of c-Jun N-terminal kinase (JNK). Furthermore, IAV-induced TGF-ß production required the presence of JNK1, a finding that suggests a role for JNK1 in IAV-induced epithelial injury and subsequent TGF-ß production. These novel findings suggest a potential mechanistic role for a distinct ER stress response induced by IAV, and a profibrogenic/repair response in contrast to other pharmacological inducers of ER stress. These responses may also have a potential role in acute lung injury, fibroproliferative acute respiratory distress syndrome, and the recently identified H1N1 influenza-induced exacerbations of chronic obstructive pulmonary disease (Wedzicha JA. Proc Am Thorac Soc 2004;1:115-120) and idiopathic pulmonary fibrosis (Umeda Y, et al. Int Med 2010;49:2333-2336).


Asunto(s)
Apoptosis , Estrés del Retículo Endoplásmico , Retículo Endoplásmico/metabolismo , Subtipo H1N1 del Virus de la Influenza A/fisiología , Pulmón/metabolismo , Infecciones por Orthomyxoviridae/metabolismo , Animales , Caspasa 12/metabolismo , Células Cultivadas , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/virología , Activación Enzimática , Ensayo de Inmunoadsorción Enzimática , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Pulmón/patología , Pulmón/virología , Ratones , Ratones Endogámicos C57BL , Infecciones por Orthomyxoviridae/patología , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/patología , Mucosa Respiratoria/virología , Estaurosporina/farmacología , Tapsigargina/farmacología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Carga Viral
4.
Am J Respir Cell Mol Biol ; 47(4): 497-508, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22652196

RESUMEN

The transcription factor NF-κB has been causally linked to inflammatory lung diseases. Recent studies have unraveled the complexity of NF-κB activation by identifying two parallel activation pathways: the classical NF-κB pathway, which is controlled by IκB kinase complex-ß (IKKß) and RelA/p50, and the alternative pathway, which is controlled by IKKα and RelB/p52. The alternative pathway regulates adaptive immune responses and lymphoid development, yet its role in the regulation of innate immune responses remains largely unknown. In this study, we determined the relevance of the alternative NF-κB pathway in proinflammatory responses in lung epithelial cells. The exposure of C10 murine alveolar lung epithelial cells to diverse stimuli, or primary murine tracheal epithelial cells to LPS, resulted in the activation of both NF-κB pathways, based on the nuclear translocation of RelA, p50, RelB, and p52. Increases in the nuclear content of RelA occurred rapidly, but transiently, whereas increases in nuclear RelB content were protracted. The small interfering (si) RNA-mediated knockdown of IKKα, RelA, or RelB resulted in decreases of multiple LPS-induced proinflammatory cytokines. Surprisingly, the siRNA ablation of IKKα or RelB led to marked increases in the production of IL-6 in response to LPS. The simultaneous expression of constitutively active (CA)-IKKα and CA-IKKß caused synergistic increases in proinflammatory mediators. Lastly, the disruption of the IKK signalsome inhibited the activation of both NF-κB pathways. These results demonstrate that the coordinated activation of both NF-κB pathways regulates the magnitude and nature of proinflammatory responses in lung epithelial cells.


Asunto(s)
Mediadores de Inflamación/metabolismo , FN-kappa B/metabolismo , Mucosa Respiratoria/metabolismo , Transducción de Señal , Animales , Anoctaminas , Células Cultivadas , Canales de Cloruro , Citocinas/genética , Citocinas/metabolismo , Citocinas/fisiología , Expresión Génica , Técnicas de Silenciamiento del Gen , Histonas/metabolismo , Proteínas I-kappa B/genética , Proteínas I-kappa B/metabolismo , Lipopolisacáridos/farmacología , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , FN-kappa B/genética , Cultivo Primario de Células , Interferencia de ARN , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/patología , Tráquea/patología
5.
Am J Respir Cell Mol Biol ; 47(3): 306-14, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22461429

RESUMEN

Recent studies suggest the importance of the transition of airway epithelial cells (EMT) in pulmonary fibrosis, and also indicate a role for Wingless protein (Wnt)/ß-catenin signaling in idiopathic pulmonary fibrosis. We investigated the possible role of the Wnt signaling pathway in inducing EMT in lung epithelial cells, and sought to unravel the role of c-Jun-N-terminal-kinase-1 (JNK1). The exposure of C10 lung epithelial cells or primary mouse tracheal epithelial cells (MTECs) to Wnt3a resulted in increases in JNK phosphorylation and nuclear ß-catenin content. Because the role of ß-catenin as a transcriptional coactivator is well established, we investigated T-cell factor/lymphocyte-enhancement factor (TCF/LEF) transcriptional activity in C10 lung epithelial cells after the activation of Wnt. TCF/LEF transcriptional activity was enhanced after the activation of Wnt, and this increase in TCF/LEF transcriptional activity was diminished after the small interfering (si)RNA-mediated ablation of JNK. The activation of the Wnt pathway by Wnt3a, or the expression of either wild-type or constitutively active ß-catenin (S37A), led to the activation of an EMT transcriptome, manifested by the increased mRNA expression of CArG box-binding factor-A, fibroblast-specific protein (FSP)-1, α-smooth muscle actin (α-SMA), and vimentin, increases in the content of α-SMA and FSP1, and the concomitant loss of zona occludens-1. The siRNA-mediated ablation of ß-catenin substantially decreased Wnt3a-induced EMT. The siRNA ablation of JNK1 largely abolished Wnt3a, ß-catenin, and ß-catenin S37a-induced EMT. In MTECs lacking Jnk1, Wnt3a-induced increases in nuclear ß-catenin, EMT transcriptome, and the content of α-SMA or FSP1 were substantially diminished. These data show that the activation of the Wnt signaling pathway is capable of inducing an EMT program in lung epithelial cells through ß-catenin, and that this process is controlled by JNK1.


Asunto(s)
Pulmón/metabolismo , Mesodermo/metabolismo , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Proteínas Wnt/fisiología , beta Catenina/fisiología , Animales , Secuencia de Bases , Western Blotting , Células Cultivadas , Cartilla de ADN , Células Epiteliales/metabolismo , Técnica del Anticuerpo Fluorescente , Pulmón/citología , Ratones , Fosforilación , ARN Interferente Pequeño , Transcripción Genética
6.
Am J Physiol Lung Cell Mol Physiol ; 303(6): L528-38, 2012 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-22752969

RESUMEN

Protein-S-glutathionylation (PSSG) is an oxidative modification of reactive cysteines that has emerged as an important player in pathophysiological processes. Under physiological conditions, the thiol transferase, glutaredoxin-1 (Glrx1) catalyses deglutathionylation. Although we previously demonstrated that Glrx1 expression is increased in mice with allergic inflammation, the impact of Glrx1/PSSG in the development of allergic airways disease remains unknown. In the present study we examined the impact of genetic ablation of Glrx1 in the pathogenesis of allergic inflammation and airway hyperresponsiveness (AHR) in mice. Glrx1(-/-) or WT mice were subjected to the antigen, ovalbumin (OVA), and parameters of allergic airways disease were evaluated 48 h after three challenges, and 48 h or 7 days after six challenges with aerosolized antigen. Although no clear increases in PSSG were observed in WT mice in response to OVA, marked increases were detected in lung tissue of mice lacking Glrx1 48 h following six antigen challenges. Inflammation and expression of proinflammatory mediators were decreased in Glrx1(-/-) mice, dependent on the time of analysis. WT and Glrx1(-/-) mice demonstrated comparable increases in AHR 48 h after three or six challenges with OVA. However, 7 days postcessation of six challenges, parameters of AHR in Glrx1(-/-) mice were resolved to control levels, accompanied by marked decreases in mucus metaplasia and expression of Muc5AC and GOB5. These results demonstrate that the Glrx1/S-glutathionylation redox status in mice is a critical regulator of AHR, suggesting that avenues to increase S-glutathionylation of specific target proteins may be beneficial to attenuate AHR.


Asunto(s)
Hiperreactividad Bronquial/inmunología , Glutarredoxinas/genética , Pulmón/patología , Moco , Animales , Glutarredoxinas/deficiencia , Glutatión/metabolismo , Enfermedades Pulmonares/patología , Metaplasia/patología , Ratones , Ovalbúmina/inmunología , Neumonía/etiología , Proteínas/metabolismo
7.
Am J Respir Cell Mol Biol ; 44(4): 571-81, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20581097

RESUMEN

Transforming growth factor (TGF)-ß1 is a key mediator of lung remodeling and fibrosis. Epithelial cells are both a source of and can respond to TGF-ß1 with epithelial-to-mesenchymal transition (EMT). We recently determined that TGF-ß1-induced EMT in lung epithelial cells requires the presence of c-Jun N-terminal kinase (JNK) 1. Because TGF-ß1 signals via Smad complexes, the goal of the present study was to determine the impact of JNK1 on phosphorylation of Smad3 and Smad3-dependent transcriptional responses in lung epithelial cells. Evaluation of JNK1-deficient lung epithelial cells demonstrated that TGF-ß1-induced terminal phosphorylation of Smad3 was similar, whereas phosphorylation of mitogen-activated protein kinase sites in the linker regions of Smad3 was diminished, in JNK1-deficient cells compared with wild-type cells. In comparison to wild-type Smad3, expression of a mutant Smad3 in which linker mitogen-activated protein kinase sites were ablated caused a marked attenuation in JNK1 or TGF-ß1-induced Smad-binding element transcriptional activity, and expression of plasminogen activator inhibitor-1, fibronectin-1, high-mobility group A2, CArG box-binding factor-A, and fibroblast-specific protein-1, genes critical in the process of EMT. JNK1 enhanced the interaction between Smad3 and Smad4, which depended on linker phosphorylation of Smad3. Conversely, Smad3 with phosphomimetic mutations in the linker domain further enhanced EMT-related genes and proteins, even in the absence of JNK1. Finally, we demonstrated a TGF-ß1-induced interaction between Smad3 and JNK1. Collectively, these results demonstrate that Smad3 phosphorylation in the linker region and Smad transcriptional activity are directly or indirectly controlled by JNK1, and provide a putative mechanism whereby JNK1 promotes TGF-ß1-induced EMT.


Asunto(s)
Transición Epitelial-Mesenquimal/efectos de los fármacos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Proteína smad3/genética , Activación Transcripcional/efectos de los fármacos , Factor de Crecimiento Transformador beta1/farmacología , Animales , Sitios de Unión , Células Epiteliales/efectos de los fármacos , Células Epiteliales/enzimología , Transición Epitelial-Mesenquimal/genética , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Pulmón/citología , Ratones , Proteínas Mutantes/metabolismo , Fosforilación/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Proteínas Recombinantes de Fusión/metabolismo , Proteína smad3/metabolismo , Proteína Smad4/metabolismo
8.
Am J Respir Cell Mol Biol ; 44(4): 491-9, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20539014

RESUMEN

Protein S-glutathionylation (PSSG), a reversible posttranslational modification of reactive cysteines, recently emerged as a regulatory mechanism that affects diverse cell-signaling cascades. The extent of cellular PSSG is controlled by the oxidoreductase glutaredoxin-1 (Grx1), a cytosolic enzyme that specifically de-glutathionylates proteins. Here, we sought to evaluate the impact of the genetic ablation of Grx1 on PSSG and on LPS-induced lung inflammation. In response to LPS, Grx1 activity increased in lung tissue and bronchoalveolar lavage (BAL) fluid in WT (WT) mice compared with PBS control mice. Glrx1(-/-) mice consistently showed slight but statistically insignificant decreases in total numbers of inflammatory cells recovered by BAL. However, LPS-induced concentrations of IL-1ß, TNF-α, IL-6, and Granulocyte/Monocyte Colony-Stimulating Factor (GM-CSF) in BAL were significantly decreased in Glrx1(-/-) mice compared with WT mice. An in situ assessment of PSSG reactivity and a biochemical evaluation of PSSG content demonstrated increases in the lung tissue of Glrx1(-/-) animals in response to LPS, compared with WT mice or PBS control mice. We also demonstrated that PSSG reactivity was prominent in alveolar macrophages (AMs). Comparative BAL analyses from WT and Glrx1(-/-) mice revealed fewer and smaller AMs in Glrx1(-/-) mice, which showed a significantly decreased expression of NF-κB family members, impaired nuclear translocation of RelA, and lower levels of NF-κB-dependent cytokines after exposure to LPS, compared with WT cells. Taken together, these results indicate that Grx1 regulates the production of inflammatory mediators through control of S-glutathionylation-sensitive signaling pathways such as NF-κB, and that Grx1 expression is critical to the activation of AMs.


Asunto(s)
Eliminación de Gen , Glutarredoxinas/deficiencia , Activación de Macrófagos/inmunología , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/patología , Neumonía/metabolismo , Neumonía/prevención & control , Animales , Líquido del Lavado Bronquioalveolar , Recuento de Células , Núcleo Celular/metabolismo , Forma de la Célula , Citocinas/metabolismo , Disulfuros/metabolismo , Glutarredoxinas/metabolismo , Glutatión/análogos & derivados , Glutatión/metabolismo , Lipopolisacáridos/administración & dosificación , Pulmón/metabolismo , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Penicilamina/metabolismo , Neumonía/patología , Transporte de Proteínas , Factor de Transcripción ReIA/metabolismo
9.
Am J Respir Cell Mol Biol ; 45(5): 931-7, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21454804

RESUMEN

It is established that cigarette smoke (CS) causes irreversible oxidations in lung epithelial cells, and can lead to their death. However, its impact on reversible and physiologically relevant redox-dependent protein modifications remains to be investigated. Glutathione is an important antioxidant against inhaled reactive oxygen species as a direct scavenger, but it can also covalently bind protein thiols upon mild oxidative stress to protect them against irreversible oxidation. This posttranslational modification, known as S-glutathionylation, can be reversed under physiological conditions by the enzyme, glutaredoxin 1 (Grx1). The aim of this study was to investigate if CS modifies Grx1, and if this impacts on protein S-glutathionylation and epithelial cell death. Upon exposure of alveolar epithelial cells to CS extract (CSE), a decrease in Grx1 mRNA and protein expression was observed, in conjunction with decreased activity and increased protein S-glutathionylation. Using mass spectrometry, irreversible oxidation of recombinant Grx1 by CSE and acrolein was demonstrated, which was associated with attenuated enzyme activity. Furthermore, carbonylation of Grx1 in epithelial cells after exposure to CSE was shown. Overexpression of Grx1 attenuated CSE-induced increases in protein S-glutathionylation and increased survival. Conversely, primary tracheal epithelial cells of mice lacking Grx1 were more sensitive to CS-induced cell death, with corresponding increases in protein S-glutathionylation. These results show that CS can modulate Grx1, not only at the expression level, but can also directly modify Grx1 itself, decreasing its activity. These findings demonstrate a role for the Grx1/S-glutathionylation redox system in CS-induced lung epithelial cell death.


Asunto(s)
Apoptosis , Glutarredoxinas/metabolismo , Glutatión/metabolismo , Fumar/metabolismo , Acroleína/toxicidad , Animales , Línea Celular , Humanos , Ratones , Alveolos Pulmonares/efectos de los fármacos , Alveolos Pulmonares/metabolismo , Fumar/efectos adversos
10.
Am J Respir Cell Mol Biol ; 43(4): 443-51, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19901348

RESUMEN

Reactive oxidants such as nitrogen dioxide (NO(2)) injure the pulmonary epithelium, causing airway damage and inflammation. We previously demonstrated that nuclear factor-κ B (NF-κB) activation within airway epithelial cells occurs in response to NO(2) inhalation, and is critical for lipopolysaccharide-induced or antigen-induced inflammatory responses. Here, we investigated whether manipulation of NF-κB activity in lung epithelium affected severe lung injuries induced by NO(2) inhalation. Wild-type C57BL/6J, CC10-IκBα(SR) transgenic mice with repressed airway epithelial NF-κB function, or transgenic mice expressing a doxycycline-inducible, constitutively active I κ B kinase ß (CC10-rTet-(CA)IKKß) with augmented NF-κB function in airway epithelium, were exposed to toxic levels of 25 ppm or 50 ppm NO(2) for 6 hours a day for 1 or 3 days. In wild-type mice, NO(2) caused the activation of NF-κB in airway epithelium after 6 hours, and after 3 days resulted in severe acute lung injury, characterized by neutrophilia, peribronchiolar lesions, and increased protein, lactate dehydrogenase, and inflammatory cytokines. Compared with wild-type mice, neutrophilic inflammation and elastase activity, lung injury, and several proinflammatory cytokines were significantly suppressed in CC10-IκBα(SR) mice exposed to 25 or 50 ppm NO(2). Paradoxically, CC10-rTet-(CA)IKKß mice that received doxycycline showed no further increase in NO(2)-induced lung injury compared with wild-type mice exposed to NO(2), instead displaying significant reductions in histologic parameters of lung injury, despite elevations in several proinflammatory cytokines. These intriguing findings demonstrate distinct functions of airway epithelial NF-κB activities in oxidant-induced severe acute lung injury, and suggest that although airway epithelial NF-κB activities modulate NO(2)-induced pulmonary inflammation, additional NF-κB-regulated functions confer partial protection from lung injury.


Asunto(s)
Lesión Pulmonar Aguda/metabolismo , Lesión Pulmonar Aguda/patología , Células Epiteliales/metabolismo , Células Epiteliales/patología , Pulmón/metabolismo , Pulmón/patología , FN-kappa B/metabolismo , Animales , Líquido del Lavado Bronquioalveolar , Núcleo Celular/metabolismo , Quimiocinas/biosíntesis , Células Epiteliales/enzimología , Proteínas I-kappa B/metabolismo , Mediadores de Inflamación/metabolismo , L-Lactato Deshidrogenasa/metabolismo , Ratones , Ratones Endogámicos C57BL , Inhibidor NF-kappaB alfa , Neutrófilos/metabolismo , Dióxido de Nitrógeno , Neumonía/metabolismo , Transporte de Proteínas , Factor de Transcripción ReIA/metabolismo , Uteroglobina/metabolismo
11.
Am J Physiol Lung Cell Mol Physiol ; 298(1): L57-66, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19897746

RESUMEN

NF-kappaB activation in the airway epithelium has been established as a critical pathway in ovalbumin (Ova)-induced airway inflammation in BALB/c mice (Poynter ME, Cloots R, van Woerkom T, Butnor KJ, Vacek P, Taatjes DJ, Irvin CG, Janssen-Heininger YM. J Immunol 173: 7003-7009, 2004). BALB/c mice are susceptible to the development of allergic airway disease, whereas other strains of mice, such as C57BL/6, are considered more resistant. The goal of the present study was to determine the proximal signals required for NF-kappaB activation in the airway epithelium in allergic airway disease and to unravel whether these signals are strain-dependent. Our previous studies, conducted in the BALB/c mouse background, demonstrated that transgenic mice expressing a dominant-negative version of IkappaBalpha in the airway epithelium (CC10-IkappaBalpha(SR)) were protected from Ova-induced inflammation. In contrast to these earlier observations, we demonstrate here that CC10-IkappaBalpha(SR) transgenic mice on the C57BL/6 background were not protected from Ova-induced allergic airway inflammation. Consistent with this finding, Ova-induced nuclear localization of the RelA subunit of NF-kappaB was not observed in C57BL/6 mice, in contrast to the marked nuclear presence of RelA in BALB/c mice. Evaluation of cytokine profiles in bronchoalveolar lavage demonstrated elevated expression of TNF-alpha in BALB/c mice compared with C57BL/6 mice after an acute challenge with Ova. Finally, neutralization of TNF-alpha by a blocking antibody prevented nuclear localization of RelA in BALB/c mice after Ova challenge. These data suggest that the mechanism of response of the airway epithelium of immunized C57BL/6 mice to antigen challenge is fundamentally different from that of immunized BALB/c mice and highlight the potential importance of TNF-alpha in regulating epithelial NF-kappaB activation in allergic airway disease.


Asunto(s)
Epitelio/metabolismo , Epitelio/patología , FN-kappa B/metabolismo , Neumonía/metabolismo , Neumonía/patología , Hipersensibilidad Respiratoria/metabolismo , Hipersensibilidad Respiratoria/patología , Animales , Antígenos/inmunología , Activación Enzimática , Células Epiteliales/enzimología , Células Epiteliales/patología , Femenino , Quinasa I-kappa B/metabolismo , Proteínas I-kappa B/metabolismo , Metaplasia , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Moco/metabolismo , Inhibidor NF-kappaB alfa , Pruebas de Neutralización , Ovalbúmina/inmunología , Neumonía/inmunología , Hipersensibilidad Respiratoria/inmunología , Especificidad de la Especie , Tráquea/patología , Transgenes/genética , Factor de Necrosis Tumoral alfa/metabolismo , Uteroglobina/metabolismo
12.
Am J Pathol ; 175(1): 36-45, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19556513

RESUMEN

Protein S-glutathionylation (PSSG) is a posttranslational modification that involves the conjugation of the small antioxidant molecule glutathione to cysteine residues and is emerging as a critical mechanism of redox-based signaling. PSSG levels increase under conditions of oxidative stress and are controlled by glutaredoxins (Grx) that, under physiological conditions, preferentially deglutathionylate cysteines and restore sulfhydryls. Both the occurrence and distribution of PSSG in tissues is unknown because of the labile nature of this oxidative event and the lack of specific reagents. The goal of this study was to establish and validate a protocol that enables detection of PSSG in situ, using the property of Grx to deglutathionylate cysteines. Using Grx1-catalyzed cysteine derivatization, we evaluated PSSG content in mice subjected to various models of lung injury and fibrosis. In control mice, PSSG was detectable primarily in the airway epithelium and alveolar macrophages. Exposure of mice to NO(2) resulted in enhanced PSSG levels in parenchymal regions, while exposure to O(2) resulted in minor detectable changes. Finally, bleomycin exposure resulted in marked increases in PSSG reactivity both in the bronchial epithelium as well as in parenchymal regions. Taken together, these findings demonstrate that Grx1-based cysteine derivatization is a powerful technique to specifically detect patterns of PSSG expression in lungs, and will enable investigations into regional changes in PSSG content in a variety of diseases.


Asunto(s)
Glutarredoxinas/metabolismo , Glutatión/análisis , Histocitoquímica/métodos , Pulmón/metabolismo , Proteína S/análisis , Animales , Biocatálisis , Cisteína/metabolismo , Glutatión/metabolismo , Pulmón/química , Pulmón/efectos de los fármacos , Enfermedades Pulmonares/inducido químicamente , Enfermedades Pulmonares/metabolismo , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Microscopía Fluorescente/métodos , Dióxido de Nitrógeno/farmacología , Oxidantes Fotoquímicos/farmacología , Oxidación-Reducción , Oxígeno/farmacología , Adhesión en Parafina , Proteína S/química , Proteína S/metabolismo
13.
Am J Respir Crit Care Med ; 177(9): 959-69, 2008 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-18263801

RESUMEN

RATIONALE: Nuclear factor (NF)-kappaB is a prominent proinflammatory transcription factor that plays a critical role in allergic airway disease. Previous studies demonstrated that inhibition of NF-kappaB in airway epithelium causes attenuation of allergic inflammation. OBJECTIVES: We sought to determine if selective activation of NF-kappaB within the airway epithelium in the absence of other agonists is sufficient to cause allergic airway disease. METHODS: A transgenic mouse expressing a doxycycline (Dox)-inducible, constitutively active (CA) version of inhibitor of kappaB (IkappaB) kinase-beta (IKKbeta) under transcriptional control of the rat CC10 promoter, was generated. MEASUREMENTS AND MAIN RESULTS: After administration of Dox, expression of the CA-IKKbeta transgene induced the nuclear translocation of RelA in airway epithelium. IKKbeta-triggered activation of NF-kappaB led to an increased content of neutrophils and lymphocytes, and concomitant production of proinflammatory mediators, responses that were not observed in transgenic mice not receiving Dox, or in transgene-negative littermate control animals fed Dox. Unexpectedly, expression of the IKKbeta transgene in airway epithelium was sufficient to cause airway hyperresponsiveness and smooth muscle thickening in absence of an antigen sensitization and challenge regimen, the presence of eosinophils, or the induction of mucus metaplasia. CONCLUSIONS: These findings demonstrate that selective activation NF-kappaB in airway epithelium is sufficient to induce airway hyperresponsiveness and smooth muscle thickening, which are both critical features of allergic airway disease.


Asunto(s)
Hiperreactividad Bronquial/metabolismo , Bronquitis/metabolismo , Doxiciclina/farmacología , FN-kappa B/metabolismo , Mucosa Respiratoria/metabolismo , Animales , Antibacterianos/farmacología , Hiperreactividad Bronquial/patología , Bronquitis/patología , Células Cultivadas , Modelos Animales de Enfermedad , Furanos , Expresión Génica , Quinasa I-kappa B/efectos de los fármacos , Quinasa I-kappa B/genética , Quinasa I-kappa B/metabolismo , Linfocitos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Músculo Liso/patología , FN-kappa B/efectos de los fármacos , Neutrófilos/patología , ARN/genética , Mucosa Respiratoria/patología , Tiofenos , Transgenes
14.
Biochim Biophys Acta ; 1760(3): 380-7, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16515838

RESUMEN

S-glutathionylation is rapidly emerging as an important post-translational modification, responsible for transducing oxidant signals. However, few approaches are available that allow visualization of glutathione mixed disulfides in intact cells. We describe here a glutaredoxin1-dependent cysteine derivatization and labeling approach, in order to visualize S-glutathionylation patterns in situ. Using this new method, marked S-glutathionylation was observed in epithelial cells, which was predominant at membrane ruffles. As expected, the labeling intensity was further enhanced in response to bolus oxidant treatments, or in cells overexpressing Nox1 plus its coactivators. In addition, manipulation of endogenous levels of glutaredoxin-1 via RNAi, or overexpression resulted in altered sensitivity to H2O2 induced formation of glutathione mixed disulfides. Overall, the derivatization approach described here preferentially detects S-glutathionylation and provides an important means to visualize this post-translational modification in sub-cellular compartments and to investigate its relation to normal physiology as well as pathology.


Asunto(s)
Cisteína/análogos & derivados , Glutatión/metabolismo , Oxidorreductasas/metabolismo , Animales , Línea Celular , Glutarredoxinas , Ratones , Ratones Endogámicos C57BL , Oxidación-Reducción , Procesamiento Proteico-Postraduccional , Albúmina Sérica Bovina/metabolismo
15.
Free Radic Biol Med ; 73: 143-53, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24816292

RESUMEN

Interleukin-17A (IL-17A) is a newly emerging player in the pathogenesis of chronic lung diseases that amplifies inflammatory responses and promotes tissue remodeling. Stimulation of lung epithelial cells with IL-17A leads to activation of the transcription factor nuclear factor κB (NF-κB), a key player in the orchestration of lung inflammation. We have previously demonstrated the importance of the redox-dependent posttranslational modification S-glutathionylation in limiting activation of NF-κB and downstream gene induction. Under physiological conditions, the enzyme glutaredoxin 1 (Grx1) acts to deglutathionylate NF-κB proteins, which restores functional activity. In this study, we sought to determine the impact of S-glutathionylation on IL-17A-induced NF-κB activation and expression of proinflammatory mediators. C10 mouse lung alveolar epithelial cells or primary mouse tracheal epithelial cells exposed to IL-17A show rapid activation of NF-κB and the induction of proinflammatory genes. Upon IL-17A exposure, sulfenic acid formation and S-glutathionylated proteins increased. Assessment of S-glutathionylation of NF-κB pathway components revealed S-glutathionylation of RelA (RelA-SSG) and inhibitory κB kinase α (IKKα-SSG) after stimulation with IL-17A. SiRNA-mediated ablation of Grx1 increased both RelA-SSG and IKKα-SSG and acutely increased nuclear content of RelA and tended to decrease nuclear RelB. SiRNA-mediated ablation or genetic ablation of Glrx1 decreased the expression of the NF-κB-regulated genes KC and CCL20 in response to IL-17A, but conversely increased the expression of IL-6. Last, siRNA-mediated ablation of IKKα attenuated nuclear RelA and RelB content and decreased expression of KC and CCL20 in response to IL-17A. Together, these data demonstrate a critical role for the S-glutathionylation/Grx1 redox axis in regulating IKKα and RelA S-glutathionylation and the responsiveness of epithelial cells to IL-17A.


Asunto(s)
Glutarredoxinas/genética , Quinasa I-kappa B/metabolismo , Interleucina-17/metabolismo , Factor de Transcripción ReIA/metabolismo , Factor de Transcripción ReIB/metabolismo , Animales , Células Cultivadas , Quimiocina CCL20/biosíntesis , Células Epiteliales/metabolismo , Regulación de la Expresión Génica , Glutatión/química , Quinasa I-kappa B/genética , Inflamación/inmunología , Inflamación/patología , Interleucina-6/biosíntesis , Pulmón/citología , Enfermedades Pulmonares/patología , Ratones , Ratones Noqueados , Oxidación-Reducción , Procesamiento Proteico-Postraduccional , Interferencia de ARN , ARN Interferente Pequeño , Mucosa Respiratoria/citología , Ácidos Sulfénicos/metabolismo , Tráquea/citología
16.
Antioxid Redox Signal ; 16(6): 496-505, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-21929356

RESUMEN

SIGNIFICANCE: Redox-based signaling governs a number of important pathways in tissue homeostasis. Consequently, deregulation of redox-controlled processes has been linked to a number of human diseases. Among the biological processes regulated by redox signaling, apoptosis or programmed cell death is a highly conserved process important for tissue homeostasis. Apoptosis can be triggered by a wide variety of stimuli, including death receptor ligands, environmental agents, and cytotoxic drugs. Apoptosis has also been implicated in the etiology of many human diseases. RECENT ADVANCES: Recent discoveries demonstrate that redox-based changes are required for efficient activation of apoptosis. Among these redox changes, alterations in the abundant thiol, glutathione (GSH), and the oxidative post-translational modification, protein S-glutathionylation (PSSG) have come to the forefront as critical regulators of apoptosis. CRITICAL ISSUES: Although redox-based changes have been documented in apoptosis and disease pathogenesis, the mechanistic details, whereby redox perturbations intersect with pathogenic processes, remain obscure. FUTURE DIRECTIONS: Further research will be needed to understand the context in which of the members of the death receptor pathways undergo ligand dependent oxidative modifications. Additional investigation into the interplay between oxidative modifications, redox enzymes, and apoptosis pathway members are also critically needed to improve our understanding how redox-based control is achieved. Such analyses will be important in understanding the diverse chronic diseases. In this review we will discuss the emerging paradigms in our current understanding of redox-based regulation of apoptosis with an emphasis on S-glutathionylation of proteins and the enzymes involved in this important post-translational modification.


Asunto(s)
Apoptosis , Glutatión/metabolismo , Animales , Humanos , Oxidación-Reducción
17.
Mol Cell Biol ; 32(17): 3464-78, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22751926

RESUMEN

We recently demonstrated that S-glutathionylation of the death receptor Fas (Fas-SSG) amplifies apoptosis (V. Anathy et al., J. Cell Biol. 184:241-252, 2009). In the present study, we demonstrate that distinct pools of Fas exist in cells. Upon ligation of surface Fas, a separate pool of latent Fas in the endoplasmic reticulum (ER) underwent rapid oxidative processing characterized by the loss of free sulfhydryl content (Fas-SH) and resultant increases in S-glutathionylation of Cys294, leading to increases of surface Fas. Stimulation with FasL rapidly induced associations of Fas with ERp57 and glutathione S-transferase π (GSTP), a protein disulfide isomerase and catalyst of S-glutathionylation, respectively, in the ER. Knockdown or inhibition of ERp57 and GSTP1 substantially decreased FasL-induced oxidative processing and S-glutathionylation of Fas, resulting in decreased death-inducing signaling complex formation and caspase activity and enhanced survival. Bleomycin-induced pulmonary fibrosis was accompanied by increased interactions between Fas-ERp57-GSTP1 and S-glutathionylation of Fas. Importantly, fibrosis was largely prevented following short interfering RNA-mediated ablation of ERp57 and GSTP. Collectively, these findings illuminate a regulatory switch, a ligand-initiated oxidative processing of latent Fas, that controls the strength of apoptosis.


Asunto(s)
Apoptosis , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/metabolismo , Retículo Endoplásmico/metabolismo , Proteína Ligando Fas/metabolismo , Gutatión-S-Transferasa pi/metabolismo , Receptor fas/metabolismo , Secuencia de Aminoácidos , Animales , Bleomicina , Caspasas/metabolismo , Línea Celular , Células Cultivadas , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/antagonistas & inhibidores , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/genética , Técnicas de Silenciamiento del Gen , Glutatión/metabolismo , Gutatión-S-Transferasa pi/antagonistas & inhibidores , Gutatión-S-Transferasa pi/genética , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Oxidación-Reducción , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/patología , Regulación hacia Arriba , Receptor fas/química
18.
Free Radic Biol Med ; 51(6): 1249-57, 2011 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-21762778

RESUMEN

The transcription factor nuclear factor κB (NF-κB) is a critical regulator of inflammation and immunity and is negatively regulated via S-glutathionylation. The inhibitory effect of S-glutathionylation is overcome by glutaredoxin-1 (Grx1), which under physiological conditions catalyzes deglutathionylation and enhances NF-κB activation. The mechanisms whereby expression of the Glrx1 gene is regulated remain unknown. Here we examined the role of NF-κB in regulating activation of Glrx1. Transgenic mice that express a doxycycline-inducible constitutively active version of inhibitory κB kinase-ß (CA-IKKß) demonstrate elevated expression of Grx1. Transient transfection of CA-IKKß also resulted in significant induction of Grx1. A 2-kb region of the Glrx1 promoter that contains two putative NF-κB binding sites was activated by CA-IKKß, RelA/p50, and lipopolysaccharide (LPS). Chromatin immunoprecipitation experiments confirmed binding of RelA to the promoter of Glrx1 in response to LPS. Stimulation of C10 lung epithelial cells with LPS caused transient increases in Grx1 mRNA expression and time-dependent increases in S-glutathionylation of IKKß. Overexpression of Grx1 decreased S-glutathionylation of IKKß, prolonged NF-κB activation, and increased levels of proinflammatory mediators. Collectively, this study demonstrates that the Glrx1 gene is positively regulated by NF-κB and suggests a feed-forward mechanism to promote NF-κB signaling by decreasing S-glutathionylation.


Asunto(s)
Células Epiteliales/metabolismo , Glutarredoxinas/metabolismo , FN-kappa B/metabolismo , Animales , Línea Celular , Células Epiteliales/inmunología , Células Epiteliales/patología , Regulación de la Expresión Génica/inmunología , Glutarredoxinas/genética , Glutarredoxinas/inmunología , Quinasa I-kappa B/genética , Inmunización , Mediadores de Inflamación/metabolismo , Lipopolisacáridos/inmunología , Lipopolisacáridos/metabolismo , Pulmón/patología , Ratones , Ratones Transgénicos , FN-kappa B/genética , Regiones Promotoras Genéticas/genética , Transducción de Señal/genética , Factor de Transcripción ReIA/metabolismo , Activación Transcripcional/genética , Transgenes/genética
19.
Ann N Y Acad Sci ; 1203: 23-8, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20716279

RESUMEN

Tissue fibrosis is believed to be a manifestation of dysregulated repair following injury, in association with impaired reepithelialization, and aberrant myofibroblast activation and proliferation. Numerous pathways have been linked to the pathogenesis of fibrotic lung disease, including the death receptor Fas, which contributes to apoptosis of lung epithelial cells. A redox imbalance also has been implicated in disease pathogenesis, although mechanistic details whereby oxidative changes intersect with profibrotic signaling pathways remain elusive. Oxidation of cysteines in proteins, such as S-glutathionylation (PSSG), is known to act as a regulatory event that affects protein function. This manuscript will discuss evidence that S-glutathionylation regulates death receptor induced apoptosis, and the potential implications for cysteine oxidations in the pathogenesis of in fibrotic lung disease.


Asunto(s)
Apoptosis/fisiología , Cisteína/metabolismo , Enfermedades Pulmonares/metabolismo , Enfermedades Pulmonares/patología , Animales , Dominio Catalítico/fisiología , Cisteína/química , Fibrosis , Glutatión/química , Glutatión/metabolismo , Humanos , Oxidación-Reducción , Transducción de Señal/fisiología
20.
J Cell Biol ; 184(2): 241-52, 2009 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-19171757

RESUMEN

Reactive oxygen species (ROS) increase ligation of Fas (CD95), a receptor important for regulation of programmed cell death. Glutathionylation of reactive cysteines represents an oxidative modification that can be reversed by glutaredoxins (Grxs). The goal of this study was to determine whether Fas is redox regulated under physiological conditions. In this study, we demonstrate that stimulation with Fas ligand (FasL) induces S-glutathionylation of Fas at cysteine 294 independently of nicotinamide adenine dinucleotide phosphate reduced oxidase-induced ROS. Instead, Fas is S-glutathionylated after caspase-dependent degradation of Grx1, increasing subsequent caspase activation and apoptosis. Conversely, overexpression of Grx1 attenuates S-glutathionylation of Fas and partially protects against FasL-induced apoptosis. Redox-mediated Fas modification promotes its aggregation and recruitment into lipid rafts and enhances binding of FasL. As a result, death-inducing signaling complex formation is also increased, and subsequent activation of caspase-8 and -3 is augmented. These results define a novel redox-based mechanism to propagate Fas-dependent apoptosis.


Asunto(s)
Apoptosis , Caspasas/metabolismo , Glutarredoxinas/metabolismo , Glutatión/metabolismo , Receptor fas/metabolismo , Animales , Muerte Celular , Células Epiteliales/metabolismo , Proteína Ligando Fas/metabolismo , Ratones , Ratones Endogámicos , Peso Molecular , Células 3T3 NIH , Oxidación-Reducción , Especies Reactivas de Oxígeno/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA