Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Artículo en Inglés | MEDLINE | ID: mdl-35722658

RESUMEN

Air liquid interface (ALI) exposure systems are gaining interest, and studies suggest enhanced response of lung cells exposed to particles at ALI as compared to submerged exposure, although the results have been somewhat inconsistent. Previous studies have used monocultures and measured particle deposition using assumptions including consistent particle deposition, particle density, and shape. This study exposed co-cultures of A549 and differentiated THP-1 cells to flame-generated particles using three exposure methods: ALI, pseudo-ALI, and submerged. The dose at ALI was measured directly, reducing the need for assumptions about particle properties and deposition. For all exposure methods an enhanced pro-inflammatory response (TNFα) and Cytochrome P450 (CYP1A1) gene expression, compared to their corresponding negative controls, was observed. ALI exposure induced a significantly greater TNFα response compared to submerged exposure. The submerged exposures exhibited greater induction of CYP1A1 than other exposure methods, although not statistically significant. Some of the factors behind the observed difference in responses for the three exposure methods include differences in physicochemical properties of particles in suspending media, delivered dose, and potential contribution of gas-phase species to cellular response in ALI exposure. However, given the difficulty and expense of ALI exposures, submerged exposure may still provide relevant information for particulate exposures.


Asunto(s)
Citocromo P-450 CYP1A1 , Factor de Necrosis Tumoral alfa , Aerosoles/química , Técnicas de Cocultivo , Citocromo P-450 CYP1A1/metabolismo , Células Epiteliales , Pulmón , Factor de Necrosis Tumoral alfa/metabolismo
2.
Physiol Rep ; 9(9): e14854, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33991451

RESUMEN

SARS-CoV-2 uptake by lung epithelial cells is a critical step in the pathogenesis of COVID-19. Viral entry is dependent on the binding of the viral spike protein to the angiotensin converting enzyme II protein (ACE2) on the host cell surface, followed by proteolytic cleavage by a host serine protease such as TMPRSS2. Infection of alveolar epithelial cells (AEC) in the distal lung is a key feature in progression to the acute respiratory distress syndrome (ARDS). We hypothesized that AEC expression of ACE2 is induced by hypoxia. In a murine model of hypoxic stress (12% FiO2), the total lung Ace2 mRNA and protein expression was significantly increased after 24 hours in hypoxia compared to normoxia (21% FiO2). In experiments with primary murine type II AEC, we found that exposure to hypoxia either in vivo (prior to isolation) or in vitro resulted in greatly increased AEC expression of both Ace2 (mRNA and protein) and of Tmprss2. However, when isolated type II AEC were maintained in culture over 5 days, with loss of type II cell characteristics and induction of type I cell features, Ace2 expression was greatly reduced, suggesting that this expression was a feature of only this subset of AEC. Finally, in primary human small airway epithelial cells (SAEC), ACE2 mRNA and protein expression were also induced by hypoxia, as was binding to purified spike protein. Hypoxia-induced increase in ACE2 expression in type II AEC may provide an explanation of the extended temporal course of human patients who develop ARDS in COVID-19.


Asunto(s)
Lesión Pulmonar Aguda/enzimología , Células Epiteliales Alveolares/enzimología , Enzima Convertidora de Angiotensina 2/biosíntesis , COVID-19/enzimología , Regulación Enzimológica de la Expresión Génica , Hipoxia/enzimología , Lesión Pulmonar Aguda/genética , Enzima Convertidora de Angiotensina 2/genética , Animales , COVID-19/genética , Células Cultivadas , Femenino , Humanos , Hipoxia/genética , Masculino , Ratones , Ratones Endogámicos C57BL
3.
Physiol Rep ; 9(4): e14761, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33625796

RESUMEN

COVID-19 causes severe disease with poor outcomes. We tested the hypothesis that early SARS-CoV-2 viral infection disrupts innate immune responses. These changes may be important for understanding subsequent clinical outcomes. We obtained residual nasopharyngeal swab samples from individuals who requested COVID-19 testing for symptoms at drive-through COVID-19 clinical testing sites operated by the University of Utah. We applied multiplex immunoassays, real-time polymerase chain reaction assays and quantitative proteomics to 20 virus-positive and 20 virus-negative samples. ACE-2 transcripts increased with infection (OR =17.4, 95% CI [CI] =4.78-63.8) and increasing viral N1 protein transcript load (OR =1.16, CI =1.10-1.23). Transcripts for two interferons (IFN) were elevated, IFN-λ1 (OR =71, CI =7.07-713) and IFN-λ2 (OR =40.2, CI =3.86-419), and closely associated with viral N1 transcripts (OR =1.35, CI =1.23-1.49 and OR =1.33 CI =1.20-1.47, respectively). Only transcripts for IP-10 were increased among systemic inflammatory cytokines that we examined (OR =131, CI =1.01-2620). We found widespread discrepancies between transcription and translation. IFN proteins were unchanged or decreased in infected samples (IFN-γ OR =0.90 CI =0.33-0.79, IFN-λ2,3 OR =0.60 CI =0.48-0.74) suggesting viral-induced shut-off of host antiviral protein responses. However, proteins for IP-10 (OR =3.74 CI =2.07-6.77) and several interferon-stimulated genes (ISG) increased with viral load (BST-1 OR =25.1, CI =3.33-188; IFIT1 OR =19.5, CI =4.25-89.2; IFIT3 OR =245, CI =15-4020; MX-1 OR =3.33, CI =1.44-7.70). Older age was associated with substantial modifications of some effects. Ambulatory symptomatic patients had an innate immune response with SARS-CoV-2 infection characterized by elevated IFN, proinflammatory cytokine and ISG transcripts, but there is evidence of a viral-induced host shut-off of antiviral responses. Our findings may characterize the disrupted immune landscape common in patients with early disease.


Asunto(s)
COVID-19/inmunología , Inmunidad Innata/inmunología , Enfermedades Nasofaríngeas/virología , SARS-CoV-2/inmunología , Carga Viral/inmunología , Adolescente , Adulto , Factores de Edad , Anciano , Anciano de 80 o más Años , COVID-19/virología , Niño , Citocinas/sangre , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Masculino , Persona de Mediana Edad , Enfermedades Nasofaríngeas/inmunología , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , SARS-CoV-2/genética , Factores Sexuales , Adulto Joven
4.
medRxiv ; 2020 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-33173878

RESUMEN

To examine innate immune responses in early SARS-CoV-2 infection that may change clinical outcomes, we compared nasopharyngeal swab data from 20 virus-positive and 20 virus-negative individuals. Multiple innate immune-related and ACE-2 transcripts increased with infection and were strongly associated with increasing viral load. We found widespread discrepancies between transcription and translation. Interferon proteins were unchanged or decreased in infected samples suggesting virally-induced shut-off of host anti-viral protein responses. However, IP-10 and several interferon-stimulated gene proteins increased with viral load. Older age was associated with modifications of some effects. Our findings may characterize the disrupted immune landscape of early disease.

5.
Artículo en Inglés | MEDLINE | ID: mdl-31342848

RESUMEN

In vitro studies are a first step toward understanding the biological effects of combustion-derived particulate matter (cdPM). A vast majority of studies expose cells to cdPM suspensions, which requires a method to collect cdPM and suspend it in an aqueous media. The consequences of different particle collection methods on particle physiochemical properties and resulting biological responses are not fully understood. This study investigated the effect of two common approaches (collection on a filter and a cold plate) and one relatively new (direct bubbling in DI water) approach to particle collection. The three approaches yielded cdPM with differences in particle size distribution, surface area, composition, and oxidative potential. The directly bubbled sample retained the smallest sized particles and the bimodal distribution observed in the gas-phase. The bubbled sample contained ∼50% of its mass as dissolved species and lower molecular weight compounds, not found in the other two samples. These differences in the cdPM properties affected the biological responses in THP-1 cells. The bubbled sample showed greater oxidative potential and cellular reactive oxygen species. The scraped sample induced the greatest TNFα secretion. These findings have implications for in vitro studies of air pollution and for efforts to better understand the underlying mechanisms.


Asunto(s)
Contaminantes Atmosféricos/toxicidad , Ceniza del Carbón/toxicidad , Monitoreo del Ambiente/métodos , Macrófagos/efectos de los fármacos , Material Particulado/toxicidad , Contaminantes Atmosféricos/química , Ceniza del Carbón/química , Humanos , Macrófagos/metabolismo , Oxidación-Reducción , Tamaño de la Partícula , Material Particulado/química , Especies Reactivas de Oxígeno/metabolismo , Células THP-1
6.
J Immunol ; 201(11): 3411-3420, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30381478

RESUMEN

Pulmonary innate immune responses involve a highly regulated multicellular network to defend the enormous surface area of the lung. Disruption of these responses renders the host susceptible to pneumonia. Alveolar epithelial cells (AEC) are a critical source of innate immune molecules such as GM-CSF, which determine the functional maturation of alveolar macrophages. In many pulmonary diseases, heterogeneous ventilation leads to regional hypoxia in the lung. The effect of hypoxia on AEC innate immune function is unknown. We now report that exposure of primary murine AEC to hypoxia (1% oxygen) for 24 h results in significant suppression of key innate immune molecules, including GM-CSF, CCL2, and IL-6. This exposure did not cause toxicity but did induce stabilization of hypoxia-inducible factor 1α protein (HIF-1α) and shift to glycolytic metabolism. Focusing on GM-CSF, we found that hypoxia greatly decreased the rate of GM-CSF transcription. Hypoxia both decreased NF-κB signaling in AEC and induced chromosomal changes, resulting in decreased accessibility in the GM-CSF proximal promoter of target sequences for NF-κB binding. In mice exposed to hypoxia in vivo (12% oxygen for 2 d), lung GM-CSF protein expression was reduced. In vivo phagocytosis of fluorescent beads by alveolar macrophages was also suppressed, but this effect was reversed by treatment with GM-CSF. These studies suggest that in critically ill patients, local hypoxia may contribute to the susceptibility of poorly ventilated lung units to infection through complementary effects on several pathways, reducing AEC expression of GM-CSF and other key innate immune molecules.


Asunto(s)
Células Epiteliales Alveolares/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Hipoxia/inmunología , Pulmón/patología , Macrófagos Alveolares/inmunología , Animales , Células Cultivadas , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Inmunidad Innata , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/genética , FN-kappa B/metabolismo , Fagocitosis , Regiones Promotoras Genéticas/genética , Transducción de Señal
7.
Artículo en Inglés | MEDLINE | ID: mdl-29227181

RESUMEN

The physicochemical properties of combustion particles that promote lung toxicity are not fully understood, hindered by the fact that combustion particles vary based on the fuel and combustion conditions. Real-world combustion-particle properties also continually change as new fuels are implemented, engines age, and engine technologies evolve. This work used laboratory-generated particles produced under controlled combustion conditions in an effort to understand the relationship between different particle properties and the activation of established toxicological outcomes in human lung cells (H441 and THP-1). Particles were generated from controlled combustion of two simple biofuel/diesel surrogates (methyl decanoate and dodecane/biofuel-blended diesel (BD), and butanol and dodecane/alcohol-blended diesel (AD)) and compared to a widely studied reference diesel (RD) particle (NIST SRM2975/RD). BD, AD, and RD particles exhibited differences in size, surface area, extractable chemical mass, and the content of individual polycyclic aromatic hydrocarbons (PAHs). Some of these differences were directly associated with different effects on biological responses. BD particles had the greatest surface area, amount of extractable material, and oxidizing potential. These particles and extracts induced cytochrome P450 1A1 and 1B1 enzyme mRNA in lung cells. AD particles and extracts had the greatest total PAH content and also caused CYP1A1 and 1B1 mRNA induction. The RD extract contained the highest relative concentration of 2-ring PAHs and stimulated the greatest level of interleukin-8 (IL-8) and tumor necrosis factor-alpha (TNFα) cytokine secretion. Finally, AD and RD were more potent activators of TRPA1 than BD, and while neither the TRPA1 antagonist HC-030031 nor the antioxidant N-acetylcysteine (NAC) affected CYP1A1 or 1B1 mRNA induction, both inhibitors reduced IL-8 secretion and mRNA induction. These results highlight that differences in fuel and combustion conditions affect the physicochemical properties of particles, and these differences, in turn, affect commonly studied biological/toxicological responses.


Asunto(s)
Contaminantes Atmosféricos/toxicidad , Biocombustibles/toxicidad , Pulmón/efectos de los fármacos , Material Particulado/toxicidad , Hidrocarburos Policíclicos Aromáticos/toxicidad , Emisiones de Vehículos/toxicidad , Línea Celular , Supervivencia Celular/efectos de los fármacos , Citocromo P-450 CYP1A1/biosíntesis , Citocromo P-450 CYP1B1/biosíntesis , Humanos , Interleucina-8/metabolismo , Pulmón/inmunología , Pulmón/metabolismo , Estrés Oxidativo/efectos de los fármacos , Tamaño de la Partícula , Canal Catiónico TRPA1/metabolismo
8.
Int J Environ Res Public Health ; 12(5): 5061-75, 2015 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-25985308

RESUMEN

This study aimed to determine if naturally occurring episodes of ozone air pollution in the Salt Lake Valley in Utah, USA, during the summer are associated with increased pulmonary inflammation and oxidative stress, increased respiratory symptoms, and decreased lung function in individuals with chronic obstructive pulmonary disease (COPD) compared to controls. We measured biomarkers (nitrite/nitrate (NOx), 8-isoprostane) in exhaled breath condensate (EBC), spirometry, and respiratory symptoms in 11 former smokers with moderate-to-severe COPD and nine former smokers without airflow obstruction during periods of low and high ozone air pollution. High ozone levels were associated with increased NOx in EBC in both COPD (8.7 (±8.5) vs. 28.6 (±17.6) µmol/L on clean air vs. pollution days, respectively, p < 0.01) and control participants (7.6 (±16.5) vs. 28.5 (±15.6) µmol/L on clean air vs. pollution days, respectively, p = 0.02). There was no difference in pollution effect between COPD and control groups, and no difference in EBC 8-isoprostane, pulmonary function, or respiratory symptoms between clean air and pollution days in either group. Former smokers both with and without airflow obstruction developed airway oxidative stress and inflammation in association with ozone air pollution episodes.


Asunto(s)
Estrés Oxidativo/efectos de los fármacos , Ozono/toxicidad , Neumonía/inducido químicamente , Enfermedad Pulmonar Obstructiva Crónica/patología , Contaminación del Aire , Biomarcadores/metabolismo , Pruebas Respiratorias , Monitoreo del Ambiente , Femenino , Humanos , Inflamación , Pulmón/efectos de los fármacos , Masculino , Persona de Mediana Edad
9.
Physiol Rep ; 3(3)2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25747588

RESUMEN

Granulocyte/macrophage colony-stimulating factor (GM-CSF) is critically important for normal pulmonary innate immunity and for functional maturation of alveolar macrophages. Alveolar epithelial cells (AEC) are a major source of GM-CSF in the lung and express this growth factor constitutively, whereas most other cells, including T cells, express GM-CSF following inflammatory stimulation. AEC expression of GM-CSF is suppressed by oxidative stress, at least in part through induction of microRNA leading to increased mRNA turnover. In this report, we compare and contrast the effect of hyperoxia on transcriptional aspects of gene regulation of GM-CSF in lung epithelia and T cells of human and mouse origin. Similar to primary murine AEC, human H820 cells that express multiple characteristics of normal alveolar epithelial cells express GM-CSF constitutively, with decreased expression and increased mRNA turnover following exposure to hyperoxia. In contrast, hyperoxia induces augmented GM-CSF expression in human and murine activated T cells, in association with enhanced GM-CSF mRNA stability. Alveolar epithelial cells demonstrate constitutive transcription, with the proximal promoter in an open configuration in normoxia, without change in hyperoxia. Conversely, in both human and murine T cells, hyperoxia increased GM-CSF gene transcription. The proximal promoter was in a closed configuration in unstimulated T cells but became accessible upon activation and still more accessible in activated T cells exposed to hyperoxia. These fundamental differences in molecular regulation of GM-CSF expression highlight the distinctive niche of alveolar epithelial cell expression of GM-CSF and offer insights into the biology of GM-CSF in the setting of acute lung injury.

10.
J Biol Chem ; 289(7): 4095-105, 2014 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-24371146

RESUMEN

GM-CSF is an endogenous pulmonary cytokine produced by normal alveolar epithelial cells (AEC) that is a key defender of the alveolar space. AEC GM-CSF expression is suppressed by oxidative stress through alternations in mRNA turnover, an effect that is reversed by treatment with recombinant GM-CSF. We hypothesized that specific microRNA (miRNA) would play a key role in AEC GM-CSF regulation. A genome-wide miRNA microarray identified 19 candidate miRNA altered in primary AEC during oxidative stress with reversal by treatment with GM-CSF. Three of these miRNA (miR 133a, miR 133a*, and miR 133b) are also predicted to bind the GM-CSF 3'-untranslated region (UTR). PCR for the mature miRNA confirmed induction during oxidative stress that was reversed by treatment with GM-CSF. Experiments using a GM-CSF 3'-UTR reporter construct demonstrated that miR133a and miR133b effects on GM-CSF expression are through interactions with the GM-CSF 3'-UTR. Using lentiviral transduction of specific mimics and inhibitors in primary murine AEC, we determined that miR133a and miR133b suppress GM-CSF expression and that their inhibition both reverses oxidant-induced suppression of GM-CSF expression and increases basal expression of GM-CSF in cells in normoxia. In contrast, these miRNAs are not active in regulation of GM-CSF expression in murine EL4 T cells. Thus, members of the miR133 family play key roles in regulation of GM-CSF expression through effects on mRNA turnover in AEC during oxidative stress. Increased understanding of GM-CSF gene regulation may provide novel miRNA-based interventions to augment pulmonary innate immune defense in lung injury.


Asunto(s)
Regiones no Traducidas 3'/fisiología , Regulación de la Expresión Génica/fisiología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/biosíntesis , MicroARNs/metabolismo , Estrés Oxidativo/fisiología , Alveolos Pulmonares/metabolismo , Animales , Línea Celular , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Humanos , Ratones , MicroARNs/genética , Alveolos Pulmonares/citología , Reacción en Cadena en Tiempo Real de la Polimerasa
11.
PLoS One ; 7(8): e42748, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22916155

RESUMEN

Lung function, acute pulmonary exacerbations (APE), and weight are the best clinical predictors of survival in cystic fibrosis (CF); however, underlying mechanisms are incompletely understood. Biomarkers of current disease state predictive of future outcomes might identify mechanisms and provide treatment targets, trial endpoints and objective clinical monitoring tools. Such CF-specific biomarkers have previously been elusive. Using observational and validation cohorts comprising 97 non-transplanted consecutively-recruited adult CF patients at the Intermountain Adult CF Center, University of Utah, we identified biomarkers informative of current disease and predictive of future clinical outcomes. Patients represented the majority of sputum producers. They were recruited March 2004-April 2007 and followed through May 2011. Sputum biomarker concentrations were measured and clinical outcomes meticulously recorded for a median 5.9 (interquartile range 5.0 to 6.6) years to study associations between biomarkers and future APE and time-to-lung transplantation or death. After multivariate modeling, only high mobility group box-1 protein (HMGB-1, mean=5.84 [log ng/ml], standard deviation [SD] =1.75) predicted time-to-first APE (hazard ratio [HR] per log-unit HMGB-1=1.56, p-value=0.005), number of future APE within 5 years (0.338 APE per log-unit HMGB-1, p<0.001 by quasi-Poisson regression) and time-to-lung transplantation or death (HR=1.59, p=0.02). At APE onset, sputum granulocyte macrophage colony stimulating factor (GM-CSF, mean 4.8 [log pg/ml], SD=1.26) was significantly associated with APE-associated declines in lung function (-10.8 FEV(1)% points per log-unit GM-CSF, p<0.001 by linear regression). Evaluation of validation cohorts produced similar results that passed tests of mutual consistency. In CF sputum, high HMGB-1 predicts incidence and recurrence of APE and survival, plausibly because it mediates long-term airway inflammation. High APE-associated GM-CSF identifies patients with large acute declines in FEV(1)%, possibly providing a laboratory-based objective decision-support tool for determination of an APE diagnosis. These biomarkers are potential CF reporting tools and treatment targets for slowing long-term progression and reducing short-term severity.


Asunto(s)
Biomarcadores/metabolismo , Fibrosis Quística/fisiopatología , Esputo/metabolismo , Adulto , Estudios de Cohortes , Fibrosis Quística/metabolismo , Fibrosis Quística/cirugía , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Proteína HMGB1/metabolismo , Humanos , Trasplante de Pulmón , Pronóstico , Resultado del Tratamiento
12.
Am J Physiol Lung Cell Mol Physiol ; 302(6): L504-11, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22227205

RESUMEN

Local pulmonary expression of granulocyte-macrophage colony-stimulating factor (GM-CSF) is critically important for defense of the pulmonary alveolar space. It is required for surfactant homeostasis and pulmonary innate immune responses and is protective against lung injury and aberrant repair. Alveolar epithelial cells (AEC) are a major source of GM-CSF; however, the control of homeostatic expression of GM-CSF is incompletely characterized. Increasing evidence suggests considerable plasticity of expression of AEC phenotypic characteristics. We tested the hypothesis that this plasticity extends to regulation of expression of GM-CSF using 1) MLE-12 cells (a commonly used murine cell line expressing some features of normal type II AEC, 2) primary murine AEC incubated under standard conditions [resulting in rapid spreading and loss of surfactant protein C (SP-C) expression with induction of the putative type I cell marker (T1α)], or 3) primary murine AEC on a hyaluronic acid/collagen matrix in defined medium, resulting in preservation of SP-C expression. AEC in standard cultures constitutively express abundant GM-CSF, with further induction in response to IL-1ß but little response to TNF-α. In contrast, primary cells cultured to preserve SP-C expression and MLE-12 cells both express little GM-CSF constitutively, with significant induction in response to TNF-α and limited response to IL-1ß. We conclude that constitutive and cytokine-induced expression of GM-CSF by AEC varies in concert with other cellular phenotypic characteristics. These changes may have important implications both for the maintenance of normal pulmonary homeostasis and for the process of repair following lung injury.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos y Macrófagos/biosíntesis , Alveolos Pulmonares/metabolismo , Animales , Línea Celular , Colágeno/metabolismo , Medios de Cultivo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Homeostasis , Ácido Hialurónico/metabolismo , Péptidos y Proteínas de Señalización Intercelular , Interleucina-1beta/metabolismo , Lesión Pulmonar/genética , Lesión Pulmonar/metabolismo , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Péptidos/genética , Alveolos Pulmonares/citología , Proteína C Asociada a Surfactante Pulmonar , Factor de Necrosis Tumoral alfa/metabolismo
13.
Am J Physiol Lung Cell Mol Physiol ; 302(3): L343-51, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22140071

RESUMEN

Exposure of mice to hyperoxia induces alveolar epithelial cell (AEC) injury, acute lung injury and death. Overexpression of granulocyte-macrophage colony-stimulating factor (GM-CSF) in the lung protects against these effects, although the mechanisms are not yet clear. Hyperoxia induces cellular injury via effects on mitochondrial integrity, associated with induction of proapoptotic members of the Bcl-2 family. We hypothesized that GM-CSF protects AEC through effects on mitochondrial integrity. MLE-12 cells (a murine type II cell line) and primary murine type II AEC were subjected to oxidative stress by exposure to 80% oxygen and by exposure to H(2)O(2). Exposure to H(2)O(2) induced cytochrome c release and decreased mitochondrial reductase activity in MLE-12 cells. Incubation with GM-CSF significantly attenuated these effects. Protection induced by GM-CSF was associated with Akt activation. GM-CSF treatment also resulted in increased expression of the antiapoptotic Bcl-2 family member, Mcl-1. Primary murine AEC were significantly more tolerant of oxidative stress than MLE-12 cells. In contrast to MLE-12 cells, primary AEC expressed significant GM-CSF at baseline and demonstrated constitutive activation of Akt and increased baseline expression of Mcl-1. Treatment with exogenous GM-CSF further increased Akt activation and Mcl-1 expression in primary AEC. Conversely, suppression of AEC GM-CSF expression by use of GM-CSF-specific small interfering RNA resulted in decreased tolerance of oxidative stress, Furthermore, silencing of Mcl-1 prevented GM-CSF-induced protection. We conclude that GM-CSF protects alveolar epithelial cells against oxidative stress-induced mitochondrial injury via the Akt pathway and its downstream components, including Mcl-1. Epithelial cell-derived GM-CSF may contribute to intrinsic defense mechanisms limiting lung injury.


Asunto(s)
Comunicación Autocrina , Citoprotección , Células Epiteliales/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/fisiología , Mitocondrias/metabolismo , Estrés Oxidativo , Alveolos Pulmonares/citología , Animales , Células Cultivadas , Células Epiteliales/efectos de los fármacos , Expresión Génica , Técnicas de Silenciamiento del Gen , Glucógeno Sintasa Quinasas/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Peróxido de Hidrógeno/farmacología , Ratones , Ratones Endogámicos C57BL , Mitocondrias/efectos de los fármacos , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Oxidantes/farmacología , Oxidación-Reducción , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Interferencia de ARN
14.
Am J Respir Cell Mol Biol ; 42(5): 545-51, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-19541845

RESUMEN

Patients with acute lung injury almost always require supplemental oxygen during treatment; however, elevated oxygen itself is toxic. Receptors for advanced glycation end-products (RAGE) are multi-ligand cell surface receptors predominantly localized to alveolar type I cells that influence development and cigarette smoke-induced inflammation, but studies that address the role of RAGE in acute lung injury are insufficient. In the present investigation, we test the hypothesis that RAGE signaling functions in hyperoxia-induced inflammation. RAGE-null mice exposed to hyperoxia survived 3 days longer than age-matched wild-type mice. After 4 days in hyperoxia, RAGE-null mice had less total cell infiltration into the airway, decreased total protein leak, diminished alveolar damage in hematoxylin and eosin-stained lung sections, and a lower lung wet-to-dry weight ratio. An inflammatory cytokine antibody array revealed decreased secretion of several proinflammatory molecules in lavage fluid obtained from RAGE knockout mice when compared with wild-type control animals. Real-time RT-PCR and immunoblotting revealed that hyperoxia induced RAGE expression in primary alveolar epithelial cells, and immunohistochemistry identified increased RAGE expression in the lungs of mice after exposure to hyperoxia. These data reveal that RAGE targeting leads to a diminished hyperoxia-induced pulmonary inflammatory response. Further research into the role of RAGE signaling in the lung should identify novel targets likely to be important in the therapeutic alleviation of lung injury and associated persistent inflammation.


Asunto(s)
Hiperoxia/complicaciones , Lesión Pulmonar/etiología , Lesión Pulmonar/prevención & control , Receptores Inmunológicos/metabolismo , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/patología , Animales , Líquido del Lavado Bronquioalveolar/citología , Células Cultivadas , Femenino , Hiperoxia/metabolismo , Hiperoxia/patología , Mediadores de Inflamación/metabolismo , Lesión Pulmonar/metabolismo , Lesión Pulmonar/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor para Productos Finales de Glicación Avanzada , Análisis de Supervivencia
15.
Am J Physiol Lung Cell Mol Physiol ; 298(3): L446-53, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20034963

RESUMEN

Pulmonary expression of granulocyte/macrophage colony-stimulating factor (GM-CSF) is critically important for normal functional maturation of alveolar macrophages. We found previously that lung GM-CSF is dramatically suppressed in mice exposed to hyperoxia. Alveolar epithelial cells (AEC) are a major source of GM-CSF in the peripheral lung, and in vivo hyperoxia resulted in greatly reduced expression of GM-CSF protein by AEC ex vivo. We now explore the mechanisms responsible for this effect, using primary cultures of murine AEC exposed to hyperoxia in vitro. Exposure of AEC to 80% oxygen/5% CO(2) for 48 h did not induce overt toxicity, but resulted in significantly decreased GM-CSF protein and mRNA expression compared with cells in normoxia. Similar effects were seen when AEC were stressed with serum deprivation, an alternative inducer of oxidative stress. The effects in AEC were opposite those in a murine lung epithelial cell line (MLE-12 cells), in which hyperoxia induced GM-CSF expression. Both hyperoxia and serum deprivation resulted in increased intracellular reactive oxygen species (ROS) in AEC. Hyperoxia and serum deprivation induced significantly accelerated turnover of GM-CSF mRNA. Treatment of AEC with catalase during oxidative stress preserved GM-CSF protein and mRNA and was associated with stabilization of GM-CSF mRNA. We conclude that hyperoxia-induced suppression of AEC GM-CSF expression is a function of ROS-induced destabilization of GM-CSF mRNA. We speculate that AEC oxidative stress results in significantly impaired pulmonary innate immune defense due to effects on local GM-CSF expression in the lung.


Asunto(s)
Células Epiteliales Alveolares/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Hiperoxia/metabolismo , Hiperoxia/patología , Estrés Oxidativo , Células Epiteliales Alveolares/efectos de los fármacos , Animales , Catalasa/farmacología , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Medio de Cultivo Libre de Suero , Regulación de la Expresión Génica/efectos de los fármacos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Ratones , Ratones Endogámicos C57BL , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/patología , Estrés Oxidativo/efectos de los fármacos , Estabilidad del ARN/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo
16.
Infect Immun ; 77(3): 1053-60, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19124601

RESUMEN

Pneumocystis infections increase host susceptibility to additional insults that would be tolerated in the absence of infection, such as hyperoxia. In an in vivo model using CD4-depleted mice, we previously demonstrated that Pneumocystis murina pneumonia causes significant mortality following an otherwise nonlethal hyperoxic insult. Infected mice demonstrated increased pulmonary inflammation and alveolar epithelial cell apoptosis compared to controls. To test the mechanisms underlying these observations, we examined expression of components of the Fas-Fas ligand pathway in P. murina-infected mice exposed to hyperoxia. Hyperoxia alone increased expression of Fas on the surface of type II alveolar epithelial cells; conversely, infection with P. murina led to increased lung expression of Fas ligand. We hypothesized that inhibition of inflammatory responses or direct inhibition of alveolar epithelial cell apoptosis would improve survival in P. murina-infected mice exposed to hyperoxia. Mice were depleted of CD4(+) T cells and infected with P. murina and then were exposed to >95% oxygen for 4 days, followed by return to normoxia. Experimental groups received vehicle, dexamethasone, or granulocyte-macrophage colony-stimulating factor (GM-CSF). Compared with the vehicle-treated group, treatment with dexamethasone reduced Fas ligand expression and significantly improved survival. Similarly, treatment with GM-CSF, an agent we have shown protects alveolar epithelial cells against apoptosis, decreased Fas ligand expression and also improved survival. Our results suggest that the dual stresses of P. murina infection and hyperoxia induce lung injury via activation of the Fas-Fas ligand pathway and that corticosteroids and GM-CSF reduce mortality in P. murina-infected mice exposed to hyperoxic stress by inhibition of inflammation and apoptosis.


Asunto(s)
Lesión Pulmonar Aguda/inmunología , Apoptosis/inmunología , Hiperoxia/inmunología , Inflamación/inmunología , Neumonía por Pneumocystis/inmunología , Lesión Pulmonar Aguda/etiología , Lesión Pulmonar Aguda/patología , Animales , Antiinflamatorios/farmacología , Apoptosis/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Dexametasona/farmacología , Proteína Ligando Fas/efectos de los fármacos , Proteína Ligando Fas/inmunología , Proteína Ligando Fas/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Hiperoxia/complicaciones , Hiperoxia/patología , Inmunohistoquímica , Inflamación/patología , Ratones , Ratones Endogámicos C57BL , Neumonía/complicaciones , Neumonía/inmunología , Neumonía/patología , Neumonía por Pneumocystis/complicaciones , Neumonía por Pneumocystis/patología , Alveolos Pulmonares/inmunología , Alveolos Pulmonares/metabolismo , Transducción de Señal/inmunología , Receptor fas/efectos de los fármacos , Receptor fas/inmunología , Receptor fas/metabolismo
17.
Am J Physiol Lung Cell Mol Physiol ; 296(3): L489-99, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19036873

RESUMEN

Persistent hypoxia can cause pulmonary arterial hypertension that may be associated with significant remodeling of the pulmonary arteries, including smooth muscle cell proliferation and hypertrophy. We previously demonstrated that the NADPH oxidase homolog NOX4 mediates human pulmonary artery smooth muscle cell (HPASMC) proliferation by transforming growth factor-beta1 (TGF-beta1). We now show that hypoxia increases HPASMC proliferation in vitro, accompanied by increased reactive oxygen species generation and NOX4 gene expression, and is inhibited by antioxidants, the flavoenzyme inhibitor diphenyleneiodonium (DPI), and NOX4 gene silencing. HPASMC proliferation and NOX4 expression are also observed when media from hypoxic HPASMC are added to HPASMC grown in normoxic conditions, suggesting autocrine stimulation. TGF-beta1 and insulin-like growth factor binding protein-3 (IGFBP-3) are both increased in the media of hypoxic HPASMC, and increased IGFBP-3 gene expression is noted in hypoxic HPASMC. Treatment with anti-TGF-beta1 antibody attenuates NOX4 and IGFBP-3 gene expression, accumulation of IGFBP-3 protein in media, and proliferation. Inhibition of IGFBP-3 expression with small interfering RNA (siRNA) decreases NOX4 gene expression and hypoxic proliferation. Conversely, NOX4 silencing does not decrease hypoxic IGFBP-3 gene expression or secreted protein. Smad inhibition does not but the phosphatidylinositol 3-kinase (PI3K) signaling pathway inhibitor LY-294002 does inhibit NOX4 and IGFBP-3 gene expression, IGFBP-3 secretion, and cellular proliferation resulting from hypoxia. Immunoblots from hypoxic HPASMC reveal increased TGF-beta1-mediated phosphorylation of the serine/threonine kinase (Akt), consistent with hypoxia-induced activation of PI3K/Akt signaling pathways to promote proliferation. We conclude that hypoxic HPASMC produce TGF-beta1 that acts in an autocrine fashion to induce IGFBP-3 through PI3K/Akt. IGFBP-3 increases NOX4 gene expression, resulting in HPASMC proliferation. These observations add to our understanding hypoxic pulmonary vascular remodeling.


Asunto(s)
Hipoxia de la Célula/fisiología , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/biosíntesis , Miocitos del Músculo Liso/metabolismo , NADPH Oxidasas/metabolismo , Arteria Pulmonar/metabolismo , Factor de Crecimiento Transformador beta1/biosíntesis , Comunicación Autocrina , Hipoxia de la Célula/genética , Proliferación Celular , Células Cultivadas , Expresión Génica , Humanos , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/genética , Modelos Biológicos , Miocitos del Músculo Liso/citología , NADPH Oxidasa 4 , NADPH Oxidasas/antagonistas & inhibidores , NADPH Oxidasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Arteria Pulmonar/citología , Interferencia de ARN , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal
18.
Am J Physiol Lung Cell Mol Physiol ; 294(6): L1094-101, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18390831

RESUMEN

The receptor for advanced glycation end-products (RAGE) is a member of the immunoglobin superfamily of multiligand receptors. Following ligand binding, mechanisms associated with host defense, tissue remodeling, and inflammation are activated. RAGE is highly expressed in pulmonary epithelium transitioning from alveolar type (AT) II to ATI cells and is upregulated in the presence of ligand; however, the regulation and function of RAGE during development are less clear. Herein, immunohistochemistry demonstrated a temporal-spatial pattern of RAGE expression in pulmonary epithelial cells from embryonic day 17.5 to postnatal day 10. Cotransfection experiments revealed that the mouse RAGE promoter was activated by early growth response gene 1 (Egr-1) and inhibited by thyroid transcription factor-1 (TTF-1) via interaction with specific regulatory elements. A rat ATI cell line (R3/1) with endogenous RAGE expression also differentially regulated RAGE when transfected with TTF-1 or Egr-1. Because Egr-1 is markedly induced in pulmonary epithelial cells exposed to cigarette smoke extract (CSE; Reynolds PR, Hoidal JR. Am J Respir Cell Mol Biol 35: 314-319, 2006.), we sought to investigate RAGE induction by CSE. Employing RT-PCR and Western blotting, RAGE and common ligands (amphoterin and S100A12) were upregulated in epithelial (R3/1 and A549) and macrophage (RAW) cell lines following exposure to CSE. Immunostaining for RAGE in cells similarly exposed and in lungs from mice exposed to cigarette smoke for 6 mo revealed elevated RAGE expression in pulmonary epithelium. After the addition of glyoxylated BSA, an advanced glycation end-product that binds RAGE, real-time RT-PCR detected a 200-fold increase in Egr-1. These results indicate that Egr-1 regulates RAGE expression during development and the likelihood of positive feedback involving Egr-1 and RAGE in cigarette smoke-related disease.


Asunto(s)
Proteína 1 de la Respuesta de Crecimiento Precoz/fisiología , Pulmón/efectos de los fármacos , Proteínas Nucleares/fisiología , Receptores Inmunológicos/biosíntesis , Mucosa Respiratoria/metabolismo , Fumar , Factores de Transcripción/fisiología , Animales , Línea Celular , Retroalimentación Fisiológica , Regulación del Desarrollo de la Expresión Génica , Pulmón/crecimiento & desarrollo , Ratones , Ratas , Receptor para Productos Finales de Glicación Avanzada , Mucosa Respiratoria/efectos de los fármacos , Factor Nuclear Tiroideo 1
19.
Am J Physiol Lung Cell Mol Physiol ; 294(1): L139-48, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17993584

RESUMEN

NAD(P)H oxidase is one of the critical enzymes mediating cellular production of reactive oxygen species and has a central role in airway smooth muscle (ASM) cell proliferation. Since reactive oxygen species also affect ASM contractile response, we hypothesized a regulatory role of NAD(P)H oxidase in ASM contractility. We therefore studied ASM function in wild-type mice (C57BL/6J) and mice deficient in a component (p47phox) of NAD(P)H oxidase. In histological sections of the trachea, we found that the area occupied by ASM was 17% more in p47(phox-/-) than in wild-type mice. After correcting for the difference in ASM content, we found that force generation did not vary between the two genotypes. Similarly, their ASM shortening velocity, maximal power, and sensitivity to acetylcholine, as well as airway responsiveness to methacholine in vivo, were not significantly different. The main finding of this study was a significantly reduced ASM relaxation in p47phox-/- compared with wild-type mice both during the stimulus and after the end of stimulation. The tension relaxation attained at the 20th second of electric field stimulation was, respectively, 17.6 +/- 2.4 and 9.2 +/- 2.3% in null and wild-type mice (P <0.01 by t-test). Similar significant differences were found in the rate of tension relaxation and the time required to reduce tension by one-half. Our data suggest that NAD(P)H oxidase may have a role in the structural arrangement and mechanical properties of the airway tissue. Most importantly, we report the first evidence that the p47phox subunit of NAD(P)H oxidase plays a role in ASM relaxation.


Asunto(s)
Pulmón/fisiología , Relajación Muscular/fisiología , Músculo Liso/fisiología , NADPH Oxidasas/deficiencia , Subunidades de Proteína/deficiencia , Fenómenos Fisiológicos Respiratorios , Animales , Cruzamientos Genéticos , Ratones , Ratones Noqueados , Músculo Liso/anatomía & histología , Músculo Liso/citología , Músculo Liso/patología , Soporte de Peso
20.
Crit Care Med ; 35(5): 1369-75, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17414728

RESUMEN

OBJECTIVE: Both persistent accumulation and activation of neutrophils may contribute to the most severe form of acute lung injury, acute respiratory distress syndrome. We analyzed the expression of neutrophil-derived S100A12 and the proinflammatory receptor for advanced glycation end products (RAGE) in patients with acute respiratory distress syndrome. Additional in vivo and in vitro experiments were performed to further analyze the contribution of S100A12 to pulmonary inflammation. SUBJECTS: We included 14 patients with acute respiratory distress syndrome and eight controls. In addition, 16 healthy subjects were included in an experimental lipopolysaccharide challenge model. INTERVENTIONS: Concentrations of S100A12 and soluble RAGE were analyzed in bronchoalveolar lavage fluid. The expression of S100A12 and RAGE in lung biopsies from patients was analyzed by immunohistochemistry. S100A12 was also analyzed in bronchoalveolar lavage fluid from eight healthy subjects after challenge with lipopolysaccharide and compared with eight controls who received placebo inhalation. Effects of S100A12 on endothelial cells were analyzed in vitro. MAIN RESULTS: Patients with acute respiratory distress syndrome had significantly enhanced pulmonary S100A12 expression and higher S100A12 protein concentrations in bronchoalveolar lavage fluid than controls. Levels of soluble RAGE were not significantly elevated in acute respiratory distress syndrome. S100A12 concentrations decreased with time from disease onset. In healthy volunteers, S100A12 was elevated in bronchoalveolar lavage fluid after lipopolysaccharide inhalation. In vitro experiments confirmed strong proinflammatory effects of human S100A12. CONCLUSIONS: S100A12 and its receptor RAGE are found at high concentrations in pulmonary tissue and bronchoalveolar lavage fluid in acute lung injury. S100A12 expression may reflect neutrophil activation during lung inflammation and contribute to pulmonary inflammation and endothelial activation via binding to RAGE.


Asunto(s)
Líquido del Lavado Bronquioalveolar/química , Neutrófilos/metabolismo , Receptores Inmunológicos/metabolismo , Síndrome de Dificultad Respiratoria/metabolismo , Proteínas S100/biosíntesis , Adulto , Anciano , Anciano de 80 o más Años , Células Endoteliales/efectos de los fármacos , Femenino , Humanos , Inmunohistoquímica , Lipopolisacáridos , Pulmón/patología , Masculino , Persona de Mediana Edad , Receptor para Productos Finales de Glicación Avanzada , Proteínas S100/análisis , Proteína S100A12 , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...