Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
PLoS One ; 9(9): e108342, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25244293

RESUMEN

BACKGROUND/OBJECTIVE: The use of electronic cigarettes (e-cigarettes) is rapidly increasing in the United States, especially among young people since e-cigarettes have been perceived as a safer alternative to conventional tobacco cigarettes. However, the scientific evidence regarding the human health effects of e-cigarettes on the lung is extremely limited. The major goal of our current study is to determine if e-cigarette use alters human young subject airway epithelial functions such as inflammatory response and innate immune defense against respiratory viral (i.e., human rhinovirus, HRV) infection. METHODOLOGY/MAIN RESULTS: We examined the effects of e-cigarette liquid (e-liquid) on pro-inflammatory cytokine (e.g., IL-6) production, HRV infection and host defense molecules (e.g., short palate, lung, and nasal epithelium clone 1, SPLUNC1) in primary human airway epithelial cells from young healthy non-smokers. Additionally, we examined the role of SPLUNC1 in lung defense against HRV infection using a SPLUNC1 knockout mouse model. We found that nicotine-free e-liquid promoted IL-6 production and HRV infection. Addition of nicotine into e-liquid further amplified the effects of nicotine-free e-liquid. Moreover, SPLUNC1 deficiency in mice significantly increased lung HRV loads. E-liquid inhibited SPLUNC1 expression in primary human airway epithelial cells. These findings strongly suggest the deleterious health effects of e-cigarettes in the airways of young people. Our data will guide future studies to evaluate the impact of e-cigarettes on lung health in human populations, and help inform the public about potential health risks of e-cigarettes.


Asunto(s)
Bronquios/patología , Sistemas Electrónicos de Liberación de Nicotina/efectos adversos , Inflamación/etiología , Virosis/patología , Animales , Bronquios/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/patología , Humanos , Interleucina-6/biosíntesis , Ratones , Ratones Noqueados , Virosis/metabolismo
2.
Am J Pathol ; 182(3): 819-27, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23256918

RESUMEN

Bacterial infection plays a critical role in exacerbations of various lung diseases, including chronic pulmonary obstructive disease (COPD) and asthma. Excessive lung inflammation is a prominent feature in disease exacerbations, but the underlying mechanisms remain poorly understood. Cell surface glycoprotein MUC18 (alias CD146 or melanoma cell adhesion molecule) has been shown to promote metastasis in several tumors, including melanoma. We explored the function of MUC18 in lung inflammatory responses to bacteria (eg, Mycoplasma pneumoniae) involved in lung disease exacerbations. MUC18 expression was increased in alveolar macrophages from lungs of COPD and asthma patients, compared with normal healthy human subjects. Mouse alveolar macrophages also express MUC18. After M. pneumoniae lung infection, Muc18(-/-) mice exhibited lower levels of the lung proinflammatory cytokines KC and TNF-α and less neutrophil recruitment than Muc18(+/+) mice. Alveolar macrophages from Muc18(-/-) mice produced less KC than those from Muc18(+/+) mice. In Muc18(-/-) mouse alveolar macrophages, adenovirus-mediated MUC18 gene transfer increased KC production. MUC18 amplified proinflammatory responses in alveolar macrophages, in part through enhancing the activation of nuclear factor-κB (NF-κB). Our results demonstrate, for the first time, that MUC18 exerts a proinflammatory function during lung bacterial infection. Up-regulated MUC18 expression in lungs (eg, in alveolar macrophages) of COPD and asthma patients may contribute to excessive inflammation during disease exacerbations.


Asunto(s)
Infecciones Bacterianas/complicaciones , Infecciones Bacterianas/patología , Neumonía/complicaciones , Neumonía/microbiología , Adenoviridae/metabolismo , Animales , Carga Bacteriana , Lavado Broncoalveolar , Antígeno CD146/genética , Antígeno CD146/metabolismo , Citocinas/biosíntesis , Femenino , Regulación de la Expresión Génica , Técnicas de Transferencia de Gen , Humanos , Mediadores de Inflamación/metabolismo , Pulmón/microbiología , Pulmón/patología , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/patología , Masculino , Ratones , Persona de Mediana Edad , Mycoplasma pneumoniae , FN-kappa B/metabolismo , Neutrófilos/metabolismo , Neutrófilos/patología , Neumonía/patología , Neumonía por Mycoplasma/microbiología , Neumonía por Mycoplasma/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptor Toll-Like 2/agonistas , Receptor Toll-Like 2/metabolismo
3.
Am J Respir Cell Mol Biol ; 47(2): 253-60, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22499853

RESUMEN

Short palate, lung and nasal epithelium clone 1 (SPLUNC1) is enriched in normal airway lining fluid, but is significantly reduced in airway epithelium exposed to a Th2 cytokine milieu. The role of SPLUNC1 in modulating airway allergic inflammation (e.g., eosinophils) remains unknown. We used SPLUNC1 knockout (KO) and littermate wild-type (C57BL/6 background) mice and recombinant SPLUNC1 protein to determine the impact of SPLUNC1 on airway allergic/eosinophilic inflammation, and to investigate the underlying mechanisms. An acute ovalbumin (OVA) sensitization and challenge protocol was used to induce murine airway allergic inflammation (e.g., eosinophils, eotaxin-2, and Th2 cytokines). Our results showed that SPLUNC1 in the bronchoalveolar lavage fluid of OVA-challenged wild-type mice was significantly reduced (P < 0.05), which was negatively correlated with levels of lung eosinophilic inflammation. Moreover, SPLUNC1 KO mice demonstrated significantly higher numbers of eosinophils in the lung after OVA challenges than did wild-type mice. Alveolar macrophages isolated from OVA-challenged SPLUNC1 KO versus wild-type mice had higher concentrations of baseline eotaxin-2 that was amplified by LPS (a known risk factor for exacerbating asthma). Human recombinant SPLUNC1 protein was applied to alveolar macrophages to study the regulation of eotaxin-2 in the context of Th2 cytokine and LPS stimulation. Recombinant SPLUNC1 protein attenuated LPS-induced eotaxin-2 production in Th2 cytokine-pretreated murine macrophages. These findings demonstrate that SPLUNC1 inhibits airway eosinophilic inflammation in allergic mice, in part by reducing eotaxin-2 production in alveolar macrophages.


Asunto(s)
Eosinófilos/inmunología , Glicoproteínas/deficiencia , Fosfoproteínas/deficiencia , Neumonía/inmunología , Animales , Líquido del Lavado Bronquioalveolar/inmunología , Quimiocina CCL24/inmunología , Quimiocina CCL24/metabolismo , Eosinófilos/metabolismo , Glicoproteínas/inmunología , Glicoproteínas/metabolismo , Células HEK293 , Humanos , Interleucina-13/inmunología , Interleucina-13/metabolismo , Interleucina-4/inmunología , Interleucina-4/metabolismo , Lipopolisacáridos/inmunología , Pulmón/inmunología , Pulmón/metabolismo , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ovalbúmina/inmunología , Fosfoproteínas/inmunología , Fosfoproteínas/metabolismo , Neumonía/metabolismo , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/metabolismo , Células Th2/inmunología , Células Th2/metabolismo
4.
PLoS One ; 7(12): e52969, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23285237

RESUMEN

BACKGROUND/OBJECTIVE: Respiratory infections including atypical bacteria Mycoplasma pneumoniae (Mp) contribute to the pathobiology of asthma and chronic obstructive pulmonary disease (COPD). Mp infection mainly targets airway epithelium and activates various signaling pathways such as nuclear factor κB (NF-κB). We have shown that short palate, lung, and nasal epithelium clone 1 (SPLUNC1) serves as a novel host defense protein and is up-regulated upon Mp infection through NF-κB activation in cultured human and mouse primary airway epithelial cells. However, the in vivo role of airway epithelial NF-κB activation in host defense against Mp infection has not been investigated. In the current study, we investigated the effects of in vivo airway epithelial NF-κB activation on lung Mp clearance and its association with airway epithelial SPLUNC1 expression. METHODOLOGY/MAIN RESULTS: Non-antimicrobial tetracycline analog 9-t-butyl doxycycline (9-TB) was initially optimized in mouse primary tracheal epithelial cell culture, and then utilized to induce in vivo airway epithelial specific NF-κB activation in conditional NF-κB transgenic mice (CC10-(CA)IKKß) with or without Mp infection. Lung Mp load and inflammation were evaluated, and airway epithelial SPLUNC1 protein was examined by immunohistochemistry. We found that 9-TB treatment in NF-κB transgene positive (Tg+), but not transgene negative (Tg-) mice significantly reduced lung Mp load. Moreover, 9-TB increased airway epithelial SPLUNC1 protein expression in NF-κB Tg+ mice. CONCLUSION: By using the non-antimicrobial 9-TB, our study demonstrates that in vivo airway epithelial NF-κB activation promotes lung bacterial clearance, which is accompanied by increased epithelial SPLUNC1 expression.


Asunto(s)
Depuración Mucociliar , Mycoplasma pneumoniae/inmunología , FN-kappa B/metabolismo , Neumonía por Mycoplasma/fisiopatología , Mucosa Respiratoria/fisiopatología , Animales , Células Cultivadas , Doxiciclina/farmacología , Humanos , Inmunidad Innata/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Depuración Mucociliar/efectos de los fármacos , Depuración Mucociliar/inmunología , Mycoplasma pneumoniae/efectos de los fármacos , Mycoplasma pneumoniae/crecimiento & desarrollo , Mycoplasma pneumoniae/metabolismo , FN-kappa B/fisiología , Neumonía por Mycoplasma/inmunología , Neumonía por Mycoplasma/metabolismo , Neumonía por Mycoplasma/microbiología , Mucosa Respiratoria/efectos de los fármacos , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/metabolismo
5.
J Innate Immun ; 4(1): 59-68, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22042134

RESUMEN

Heat shock factor 1 (HSF1) is a transcriptional factor that controls the induction of heat shock proteins (e.g. HSP70) in response to stress. Bacterial infections contribute to the pathobiology of chronic lung diseases such as chronic obstructive pulmonary disease and asthma. Whether HSF1 is critical to lung bacterial infection remains unknown. This study is aimed at investigating the impact of HSF1 deficiency on lung Mycoplasma pneumoniae (Mp) infection and elucidating the underlying molecular mechanisms, such as Toll-like receptor 2 (TLR2) signaling. HSF1(-/-) and HSF1(+/+) mice were intranasally infected with Mp or saline and sacrificed 4, 24 and 72 h after treatment. HSF1(-/-) mice had a higher lung Mp load than HSF1(+/+) mice. Mp-induced lung TLR2, nuclear factor-κB and associated inflammation [e.g. keratinocyte-derived chemokine (KC), neutrophils and histopathology] were delayed in HSF1(-/-) mice as compared to HSF1(+/+) mice. HSP70 protein levels in bronchoalveolar lavage fluid of HSF1(-/-) mice were decreased. Furthermore, in response to Mp infection, HSF1(-/-) alveolar macrophages had less TLR2 mRNA expression and KC production than HSF1(+/+) counterparts. Nuclear factor-κB activity and KC production in HSF1(-/-) macrophages could be rescued by addition of exogenous HSP70 protein. These data suggest that HSF1 is necessary to initiate host defense against bacterial infection partly through promoting early TLR2 signaling activation.


Asunto(s)
Proteínas de Unión al ADN/inmunología , Inmunidad Innata , Pulmón/inmunología , Macrófagos Alveolares/inmunología , Mycoplasma pneumoniae/inmunología , Neumonía por Mycoplasma/inmunología , Factores de Transcripción/inmunología , Animales , Lavado Broncoalveolar , Proteínas de Unión al ADN/genética , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP70 de Choque Térmico/inmunología , Proteínas HSP70 de Choque Térmico/farmacología , Factores de Transcripción del Choque Térmico , Pulmón/microbiología , Pulmón/patología , Macrófagos Alveolares/microbiología , Macrófagos Alveolares/patología , Ratones , Ratones Noqueados , FN-kappa B/genética , FN-kappa B/inmunología , Neumonía por Mycoplasma/genética , Neumonía por Mycoplasma/microbiología , Neumonía por Mycoplasma/patología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transducción de Señal/inmunología , Receptor Toll-Like 2/genética , Receptor Toll-Like 2/inmunología , Factores de Transcripción/genética
6.
Exp Lung Res ; 37(8): 500-8, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21892915

RESUMEN

Impaired airway innate immunity (e.g., suppressed Toll-like receptor 2 [TLR2] signaling) has been reported in allergic lungs with bacterial infection. Recently, an allergic mouse lung milieu including the T-helper type 2 (Th2) cytokine interleukin-13 (IL-13) has been shown to up-regulate lung microRNA-21 (miR-21) expression. Whether miR-21 modulates in vivo TLR2 signaling is unknown. The goal of this study was to determine if in vivo, miR-21 regulates a TLR2 agonist-induced lung inflammatory response. Balb/c mice were intranasally pretreated with a locked nucleic acid (LNA) in vivo inhibitor probe for mouse miR-21 or a control probe, followed by intranasal instillation of a TLR2 agonist Pam3CSK4, or saline (control). Four and/or 24 hours later, mice treated with the miR-21 inhibitor probe, as compared to the control probe, significantly increased lung leukocytes, including neutrophils and the keratinocyte-derived chemokine (KC). IL-13 treatment for 72 hours increased (P < .05) miR-21 in cultured primary normal human airway epithelial cells. These results, for the first time, suggest an in vivo role of miR-21 in suppressing TLR2 signaling, and further support that IL-13 can up-regulate miR-21 in human airway epithelial cells. Functional studies on miR-21 likely provide novel approaches to modulate TLR2 signaling in Th2 cytokine-exposed airways.


Asunto(s)
MicroARNs/inmunología , Neumonía/prevención & control , Receptor Toll-Like 2/agonistas , Animales , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Inmunidad Innata , Interleucina-13/farmacología , Lipopéptidos/toxicidad , Ratones , Ratones Endogámicos BALB C , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , MicroARNs/metabolismo , Neumonía/etiología , Neumonía/inmunología , Transducción de Señal/inmunología , Regulación hacia Arriba/efectos de los fármacos
7.
Am J Pathol ; 178(5): 2159-67, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21514430

RESUMEN

Short palate, lung, and nasal epithelium clone 1 (SPLUNC1) protein is highly expressed in normal airways, but is dramatically decreased in allergic and cigarette smoke exposure settings. We have previously demonstrated SPLUNC1 in vitro antibacterial property against Mycoplasma pneumoniae (Mp). However, its in vivo biological functions remain unclear. The objectives of this study were to determine the in vivo functions of SPLUNC1 following bacterial (eg, Mp) infection, and to examine the underlying mechanisms. We generated SPLUNC1-deficient mice and utilized transgenic mice overexpressing human SPLUNC1 exclusively within the airway epithelium. These mice were infected with Mp and, twenty-four hours post infection, their host defense responses were compared to littermate controls. Mp levels and inflammatory cells increased in the lungs of SPLUNC1(-/-) mice as compared to wild type controls. SPLUNC1 deficiency was shown to contribute to impaired neutrophil activation. In contrast, mice overexpressing hSPLUNC1 exclusively in airway epithelial cells demonstrated lower Mp levels. Furthermore, neutrophil elastase activity was significantly increased in mice overexpressing hSPLUNC1. Lastly, we demonstrated that SPLUNC1 enhanced Mp-induced human neutrophil elastase (HNE) activity, and HNE directly inhibited the growth of Mp. Our findings demonstrate a critical in vivo role of SPLUNC1 in host defense against bacterial infection, and likely provide a novel therapeutic approach to restore impaired lung innate immune responses to bacteria in patients with chronic lung diseases.


Asunto(s)
Glicoproteínas/inmunología , Inmunidad Innata/inmunología , Mycoplasma pneumoniae/inmunología , Fosfoproteínas/inmunología , Neumonía por Mycoplasma/inmunología , Animales , Western Blotting , Glicoproteínas/metabolismo , Humanos , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mycoplasma pneumoniae/metabolismo , Fosfoproteínas/metabolismo , Neumonía por Mycoplasma/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
8.
J Infect Dis ; 203(9): 1240-8, 2011 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-21357942

RESUMEN

BACKGROUND: Cigarette smoke (CS) exposure is an epidemiological risk factor for tuberculosis, although the biological basis has not been elucidated. METHODS: We exposed C57BL/6 mice to CS for 14 weeks and examined their ability to control an aerosol infection of Mycobacterium tuberculosis Erdman. RESULTS: CS-exposed mice had more M. tuberculosis isolated from the lungs and spleens after 14 and 30 d, compared with control mice. The CS-exposed mice had worse lung lesions and less lung and splenic macrophages and dendritic cells (DCs) producing interleukin12 and tumor necrosis factor α (TNF-α). There were significantly more interleukin 10-producing macrophages and DCs in the spleens of infected CS-exposed mice than in non-CS-exposed controls. CS-exposed mice also showed a diminished influx of interferon γ-producing and TNF-α-producing CD4(+) and CD8(+) effector and memory T cells into the lungs and spleens. There was a trend toward an increased number of viable intracellular M. tuberculosis in macrophages isolated from humans who smoke compared with nonsmokers. THP-1 human macrophages and primary human alveolar macrophages exposed to CS extract, nicotine, or acrolein showed an increased burden of intracellular M. tuberculosis. CONCLUSION: CS suppresses the protective immune response to M. tuberculosis in mice, human THP-1 cells, and primary human alveolar macrophages.


Asunto(s)
Susceptibilidad a Enfermedades , Mycobacterium tuberculosis/inmunología , Fumar/efectos adversos , Tuberculosis/inmunología , Animales , Modelos Animales de Enfermedad , Femenino , Ratones , Ratones Endogámicos C57BL
9.
Am J Physiol Lung Cell Mol Physiol ; 300(4): L579-86, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21239529

RESUMEN

Decreased Toll-like receptor 2 (TLR2) expression has been reported in patients with chronic obstructive pulmonary disease and in a murine asthma model, which may predispose the hosts to bacterial infections, leading to disease exacerbations. Since airway epithelial cells serve as the first line of respiratory mucosal defense, the present study aimed to reveal the role of airway epithelial TLR2 signaling to lung bacterial [i.e., Mycoplasma pneumoniae (Mp)] clearance. In vivo TLR2 gene transfer via intranasal inoculation of adenoviral vector was performed to reconstitute TLR2 expression in airway epithelium of TLR2(-/-) BALB/c mice, with or without ensuing Mp infection. TLR2 and lactotransferrin (LTF) expression in airway epithelial cells and lung Mp load were assessed. Adenovirus-mediated TLR2 gene transfer to airway epithelial cells of TLR2(-/-) mice reconstituted 30-40% TLR2 expression compared with TLR2(+/+) cells. Such airway epithelial TLR2 reconstitution in TLR2(-/-) mice significantly reduced lung Mp load (an appropriate 45% reduction), coupled with elevated LTF expression. LTF expression in mice was shown to be mainly dependent on TLR2 signaling in response to Mp infection. Exogenous human LTF protein dose-dependently decreased lung bacterial load in Mp-infected TLR2(-/-) mice. In addition, human LTF protein directly dose-dependently decreased Mp levels in vitro. These data indicate that reconstitution of airway epithelial TLR2 signaling in TLR2(-/-) mice significantly restores lung defense against bacteria (e.g., Mp) via increased lung antimicrobial protein LTF production. Our findings may offer a deliverable approach to attenuate bacterial infections in airways of asthma or chronic obstructive pulmonary disease patients with impaired TLR2 function.


Asunto(s)
Infecciones Bacterianas/inmunología , Infecciones Bacterianas/fisiopatología , Células Epiteliales/microbiología , Células Epiteliales/patología , Pulmón/patología , Neumonía por Mycoplasma/inmunología , Receptor Toll-Like 2/inmunología , Adenoviridae/genética , Animales , Infecciones Bacterianas/microbiología , Infecciones Bacterianas/patología , Carga Bacteriana/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas de Transferencia de Gen , Humanos , Lactoferrina/metabolismo , Lactoferrina/farmacología , Ratones , Ratones Endogámicos BALB C , Mycoplasma pneumoniae/efectos de los fármacos , Mycoplasma pneumoniae/crecimiento & desarrollo , Neumonía por Mycoplasma/microbiología , Neumonía por Mycoplasma/patología , Neumonía por Mycoplasma/fisiopatología , Receptor Toll-Like 2/deficiencia , Receptor Toll-Like 2/genética
10.
Respir Res ; 11: 155, 2010 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-21054862

RESUMEN

BACKGROUND: Respiratory infections including Mycoplasma pneumoniae (Mp) contribute to various chronic lung diseases. We have shown that mouse short palate, lung, and nasal epithelium clone 1 (SPLUNC1) protein was able to inhibit Mp growth. Further, airway epithelial cells increased SPLUNC1 expression upon Mp infection. However, the mechanisms underlying SPLUNC1 regulation remain unknown. In the current study, we investigated if SPLUNC1 production following Mp infection is regulated through Toll-like receptor 2 (TLR2) signaling. METHODS: Airway epithelial cell cultures were utilized to reveal the contribution of TLR2 signaling including NF-κB to SPLUNC1 production upon bacterial infection and TLR2 agonist stimulation. RESULTS: Mp and TLR2 agonist Pam3CSK4 increased SPLUNC1 expression in tracheal epithelial cells from wild type, but not TLR2(-/-) BALB/c mice. RNA interference (short-hairpin RNA) of TLR2 in normal human bronchial epithelial cells under air-liquid interface cultures significantly reduced SPLUNC1 levels in Mp-infected or Pam3CSK4-treated cells. Inhibition and activation of NF-κB pathway decreased and increased SPLUNC1 production in airway epithelial cells, respectively. CONCLUSIONS: Our data for the first time suggest that airway epithelial TLR2 signaling is pivotal in mycoplasma-induced SPLUNC1 production, thus improving our understanding of the aberrant SPLUNC1 expression in airways of patients suffering from chronic lung diseases with bacterial infections.


Asunto(s)
Glicoproteínas/metabolismo , Fosfoproteínas/metabolismo , Neumonía por Mycoplasma/metabolismo , Mucosa Respiratoria/metabolismo , Receptor Toll-Like 2/metabolismo , Animales , Células Cultivadas , Células Epiteliales , Regulación de la Expresión Génica , Humanos , Ratones
11.
COPD ; 6(3): 185-91, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19811374

RESUMEN

Decreased Clara cell secretory protein (CCSP) levels have been found in smokers and chronic obstructive pulmonary disease (COPD) patients, which may be related to the development of COPD. A phosphodiesterase-4 (PDE4) inhibitor, roflumilast, appears to have therapeutic value for COPD. However, its effect on CCSP in cigarette smoke (CS)-exposed lungs has not been investigated. AKR/J mice were treated as follows: air control, CS, roflumilast plus CS, and roflumilast. Mice underwent four weeks of air or CS exposure. Roflumilast was administrated at 5mg/kg via gavage once daily for the duration of the study. CCSP levels in bronchoalveolar lavage (BAL) fluid and ERK1/2 activation in lungs were examined. CS exposure tended to decrease CCSP levels in BAL fluid compared to air controls. Treatment with roflumilast significantly reversed CS-induced downward trend of CCSP in BAL fluid. Roflumilast significantly inhibited CS-induced upward trend of ERK1/2 activation in lungs, and the levels of activated ERK1/2 in lungs negatively correlated with CCSP levels of BAL fluid in CS, and CS plus roflumilast groups. Our results demonstrate that one of the therapeutic mechanisms of roflumilast is to reverse CS-induced downward trend in CCSP levels of BAL fluid, which may be mediated by down-regulating ERK1/2 activity.


Asunto(s)
Aminopiridinas/farmacología , Benzamidas/farmacología , Inhibidores de Fosfodiesterasa/farmacología , Fumar/efectos adversos , Uteroglobina/efectos de los fármacos , Animales , Western Blotting , Líquido del Lavado Bronquioalveolar/citología , Ciclopropanos/farmacología , Regulación hacia Abajo , Técnicas para Inmunoenzimas , Masculino , Ratones , Ratones Endogámicos AKR , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Estadísticas no Paramétricas , Uteroglobina/metabolismo
12.
PLoS One ; 4(8): e6860, 2009 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-19718433

RESUMEN

BACKGROUND: Cigarette smoking is the major cause of chronic obstructive pulmonary disease (COPD) and lung cancer. Respiratory bacterial infections have been shown to be involved in the development of COPD along with impaired airway innate immunity. METHODOLOGY/PRINCIPAL FINDINGS: To address the in vivo impact of cigarette smoke (CS) exclusively on host innate defense mechanisms, we took advantage of Caenorhabditis elegans (C. elegans), which has an innate immune system but lacks adaptive immune function. Pseudomonas aeruginosa (PA) clearance from intestines of C. elegans was dampened by CS. Microarray analysis identified 6 candidate genes with a 2-fold or greater reduction after CS exposure, that have a human orthologue, and that may participate in innate immunity. To confirm a role of CS-down-regulated genes in the innate immune response to PA, RNA interference (RNAi) by feeding was carried out in C. elegans to inhibit the gene of interest, followed by PA infection to determine if the gene affected innate immunity. Inhibition of lbp-7, which encodes a lipid binding protein, resulted in increased levels of intestinal PA. Primary human bronchial epithelial cells were shown to express mRNA of human Fatty Acid Binding Protein 5 (FABP-5), the human orthologue of lpb-7. Interestingly, FABP-5 mRNA levels from human smokers with COPD were significantly lower (p = 0.036) than those from smokers without COPD. Furthermore, FABP-5 mRNA levels were up-regulated (7-fold) after bacterial (i.e., Mycoplasma pneumoniae) infection in primary human bronchial epithelial cell culture (air-liquid interface culture). CONCLUSIONS: Our results suggest that the C. elegans model offers a novel in vivo approach to specifically study innate immune deficiencies resulting from exposure to cigarette smoke, and that results from the nematode may provide insight into human airway epithelial cell biology and cigarette smoke exposure.


Asunto(s)
Caenorhabditis elegans/inmunología , Inmunidad Innata , Modelos Animales , Nicotiana , Humo , Animales , Caenorhabditis elegans/microbiología , Cotinina/metabolismo , Nicotina/metabolismo , Pseudomonas aeruginosa/aislamiento & purificación , Interferencia de ARN
13.
Respir Med ; 102(11): 1604-10, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18590957

RESUMEN

Bacterial infections including Mycoplasma pneumoniae (Mp) are a major cause of exacerbations in chronic obstructive pulmonary disease (COPD). Cigarette smoke (CS) is the leading cause of COPD, and affects the function of alveolar macrophages that act as the first line of defense against the invading respiratory pathogens. Macrophages express a transmembrane receptor called macrophage receptor with collagenous structure (MARCO) that is involved in the clearance of microorganisms. Whether CS down-regulates MARCO and eventually decreases the clearance of Mp has not been investigated. We utilized human monocytic cell line (THP-1)-derived macrophages to examine the effects of CS extract (CSE) on MARCO expression and Mp growth. Specifically, macrophages were pre-exposed to CSE for 6 h, and then infected with or without Mp for 2 h. MARCO was examined at both mRNA and protein levels by using real-time PCR and immunofluorescent staining, respectively. Mp in the supernatants was quantified by quantitative culture. In addition, a neutralizing MARCO antibody was added to macrophages to test if blockade of MARCO impaired Mp clearance. We found that CSE significantly decreased MARCO expression in a dose-dependant manner at 6 h post-CSE. Mp levels in CSE-treated cells were higher than those in non-CSE-treated cells, indicating a decreased pathogen clearance. Additionally, neutralizing MARCO in macrophages markedly increased Mp levels. Our results indicate that cigarette smoke exposure down-regulates MARCO expression in macrophages, which may be in part responsible for impaired bacterial (e.g., Mp) clearance.


Asunto(s)
Macrófagos/metabolismo , Mycoplasma pneumoniae/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Receptores Inmunológicos/metabolismo , Mucosa Respiratoria/metabolismo , Animales , Líquido del Lavado Bronquioalveolar/inmunología , Células Cultivadas , Humanos , Macrófagos/inmunología , Ratones , Mycoplasma pneumoniae/inmunología , Reacción en Cadena de la Polimerasa/métodos , Enfermedad Pulmonar Obstructiva Crónica/inmunología , Enfermedad Pulmonar Obstructiva Crónica/patología , Receptores Inmunológicos/inmunología , Humo/efectos adversos , Nicotiana/efectos adversos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...