Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
J Immunol ; 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-39033086

ABSTRACT

The aryl hydrocarbon receptor (AHR) is a receptor/transcription factor widely expressed in the lung. The physiological roles of AHR expressed in the alveolar epithelium remain unclear. In this study, we tested the hypothesis that alveolar epithelial AHR activity plays an important role in modulating inflammatory responses and maintaining alveolar integrity during lung injury and repair. AHR is expressed in alveolar epithelial cells (AECs) and is active. AHR activation with the endogenous AHR ligand, FICZ (5,11-dihydroindolo[3,2-b] carbazole-6-carboxaldehyde), significantly suppressed inflammatory cytokine expression in response to inflammatory stimuli in primary murine AECs and in the MLE-15 epithelial cell line. In an LPS model of acute lung injury in mice, coadministration of FICZ with LPS suppressed protein leak, reduced neutrophil accumulation in BAL fluid, and suppressed inflammatory cytokine expression in lung tissue and BAL fluid. Relevant to healing following inflammatory injury, AHR activation suppressed TGF-ß-induced expression of genes associated with epithelial-mesenchymal transition. Knockdown of AHR in primary AECs with shRNA or in CRISPR-Cas-9-induced MLE-15 cells resulted in upregulation of α-smooth muscle actin (αSma), Col1a1, and Fn1 and reduced expression of epithelial genes Col4a1 and Sdc1. MLE-15 clones lacking AHR demonstrated accelerated wound closure in a scratch model. AHR activation with FICZ enhanced barrier function (transepithelial electrical resistance) in primary murine AECs and limited decline of transepithelial electrical resistance following inflammatory injury. AHR activation in AECs preserves alveolar integrity by modulating inflammatory cytokine expression while enhancing barrier function and limiting stress-induced expression of mesenchymal genes.

2.
Article in English | MEDLINE | ID: mdl-35722658

ABSTRACT

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.


Subject(s)
Cytochrome P-450 CYP1A1 , Tumor Necrosis Factor-alpha , Aerosols/chemistry , Coculture Techniques , Cytochrome P-450 CYP1A1/metabolism , Epithelial Cells , Lung , Tumor Necrosis Factor-alpha/metabolism
3.
Physiol Rep ; 9(9): e14854, 2021 05.
Article in English | MEDLINE | ID: mdl-33991451

ABSTRACT

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.


Subject(s)
Acute Lung Injury/enzymology , Alveolar Epithelial Cells/enzymology , Angiotensin-Converting Enzyme 2/biosynthesis , COVID-19/enzymology , Gene Expression Regulation, Enzymologic , Hypoxia/enzymology , Acute Lung Injury/genetics , Angiotensin-Converting Enzyme 2/genetics , Animals , COVID-19/genetics , Cells, Cultured , Female , Humans , Hypoxia/genetics , Male , Mice , Mice, Inbred C57BL
4.
Physiol Rep ; 9(4): e14761, 2021 02.
Article in English | MEDLINE | ID: mdl-33625796

ABSTRACT

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.


Subject(s)
COVID-19/immunology , Immunity, Innate/immunology , Nasopharyngeal Diseases/virology , SARS-CoV-2/immunology , Viral Load/immunology , Adolescent , Adult , Age Factors , Aged , Aged, 80 and over , COVID-19/virology , Child , Cytokines/blood , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Middle Aged , Nasopharyngeal Diseases/immunology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , SARS-CoV-2/genetics , Sex Factors , Young Adult
5.
medRxiv ; 2020 Nov 04.
Article in English | MEDLINE | ID: mdl-33173878

ABSTRACT

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.

6.
Article in English | MEDLINE | ID: mdl-31342848

ABSTRACT

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.


Subject(s)
Air Pollutants/toxicity , Coal Ash/toxicity , Environmental Monitoring/methods , Macrophages/drug effects , Particulate Matter/toxicity , Air Pollutants/chemistry , Coal Ash/chemistry , Humans , Macrophages/metabolism , Oxidation-Reduction , Particle Size , Particulate Matter/chemistry , Reactive Oxygen Species/metabolism , THP-1 Cells
7.
J Immunol ; 201(11): 3411-3420, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30381478

ABSTRACT

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.


Subject(s)
Alveolar Epithelial Cells/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Hypoxia/immunology , Lung/pathology , Macrophages, Alveolar/immunology , Animals , Cells, Cultured , Female , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Immunity, Innate , Male , Mice , Mice, Inbred C57BL , NF-kappa B/genetics , NF-kappa B/metabolism , Phagocytosis , Promoter Regions, Genetic/genetics , Signal Transduction
8.
Article in English | MEDLINE | ID: mdl-29227181

ABSTRACT

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.


Subject(s)
Air Pollutants/toxicity , Biofuels/toxicity , Lung/drug effects , Particulate Matter/toxicity , Polycyclic Aromatic Hydrocarbons/toxicity , Vehicle Emissions/toxicity , Cell Line , Cell Survival/drug effects , Cytochrome P-450 CYP1A1/biosynthesis , Cytochrome P-450 CYP1B1/biosynthesis , Humans , Interleukin-8/metabolism , Lung/immunology , Lung/metabolism , Oxidative Stress/drug effects , Particle Size , TRPA1 Cation Channel/metabolism
9.
Int J Environ Res Public Health ; 12(5): 5061-75, 2015 May 12.
Article in English | MEDLINE | ID: mdl-25985308

ABSTRACT

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.


Subject(s)
Oxidative Stress/drug effects , Ozone/toxicity , Pneumonia/chemically induced , Pulmonary Disease, Chronic Obstructive/pathology , Air Pollution , Biomarkers/metabolism , Breath Tests , Environmental Monitoring , Female , Humans , Inflammation , Lung/drug effects , Male , Middle Aged
10.
Physiol Rep ; 3(3)2015 Mar.
Article in English | MEDLINE | ID: mdl-25747588

ABSTRACT

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.

11.
J Biol Chem ; 289(7): 4095-105, 2014 Feb 14.
Article in English | MEDLINE | ID: mdl-24371146

ABSTRACT

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.


Subject(s)
3' Untranslated Regions/physiology , Gene Expression Regulation/physiology , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , MicroRNAs/metabolism , Oxidative Stress/physiology , Pulmonary Alveoli/metabolism , Animals , Cell Line , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Humans , Mice , MicroRNAs/genetics , Pulmonary Alveoli/cytology , Real-Time Polymerase Chain Reaction
12.
PLoS One ; 7(8): e42748, 2012.
Article in English | MEDLINE | ID: mdl-22916155

ABSTRACT

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.


Subject(s)
Biomarkers/metabolism , Cystic Fibrosis/physiopathology , Sputum/metabolism , Adult , Cohort Studies , Cystic Fibrosis/metabolism , Cystic Fibrosis/surgery , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , HMGB1 Protein/metabolism , Humans , Lung Transplantation , Prognosis , Treatment Outcome
13.
Am J Physiol Lung Cell Mol Physiol ; 302(6): L504-11, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22227205

ABSTRACT

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.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Pulmonary Alveoli/metabolism , Animals , Cell Line , Collagen/metabolism , Culture Media , Epithelial Cells/cytology , Epithelial Cells/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Homeostasis , Hyaluronic Acid/metabolism , Intercellular Signaling Peptides and Proteins , Interleukin-1beta/metabolism , Lung Injury/genetics , Lung Injury/metabolism , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Peptides/genetics , Pulmonary Alveoli/cytology , Pulmonary Surfactant-Associated Protein C , Tumor Necrosis Factor-alpha/metabolism
14.
Am J Physiol Lung Cell Mol Physiol ; 302(3): L343-51, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22140071

ABSTRACT

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.


Subject(s)
Autocrine Communication , Cytoprotection , Epithelial Cells/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Mitochondria/metabolism , Oxidative Stress , Pulmonary Alveoli/cytology , Animals , Cells, Cultured , Epithelial Cells/drug effects , Gene Expression , Gene Knockdown Techniques , Glycogen Synthase Kinases/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Hydrogen Peroxide/pharmacology , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Myeloid Cell Leukemia Sequence 1 Protein , Oxidants/pharmacology , Oxidation-Reduction , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA Interference
15.
Am J Respir Cell Mol Biol ; 42(5): 545-51, 2010 May.
Article in English | MEDLINE | ID: mdl-19541845

ABSTRACT

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.


Subject(s)
Hyperoxia/complications , Lung Injury/etiology , Lung Injury/prevention & control , Receptors, Immunologic/metabolism , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/pathology , Animals , Bronchoalveolar Lavage Fluid/cytology , Cells, Cultured , Female , Hyperoxia/metabolism , Hyperoxia/pathology , Inflammation Mediators/metabolism , Lung Injury/metabolism , Lung Injury/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor for Advanced Glycation End Products , Survival Analysis
16.
Am J Physiol Lung Cell Mol Physiol ; 298(3): L446-53, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20034963

ABSTRACT

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.


Subject(s)
Alveolar Epithelial Cells/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Hyperoxia/metabolism , Hyperoxia/pathology , Oxidative Stress , Alveolar Epithelial Cells/drug effects , Animals , Catalase/pharmacology , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Culture Media, Serum-Free , Gene Expression Regulation/drug effects , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Oxidative Stress/drug effects , RNA Stability/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism
17.
Infect Immun ; 77(3): 1053-60, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19124601

ABSTRACT

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.


Subject(s)
Acute Lung Injury/immunology , Apoptosis/immunology , Hyperoxia/immunology , Inflammation/immunology , Pneumonia, Pneumocystis/immunology , Acute Lung Injury/etiology , Acute Lung Injury/pathology , Animals , Anti-Inflammatory Agents/pharmacology , Apoptosis/drug effects , CD4-Positive T-Lymphocytes/immunology , Dexamethasone/pharmacology , Fas Ligand Protein/drug effects , Fas Ligand Protein/immunology , Fas Ligand Protein/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Hyperoxia/complications , Hyperoxia/pathology , Immunohistochemistry , Inflammation/pathology , Mice , Mice, Inbred C57BL , Pneumonia/complications , Pneumonia/immunology , Pneumonia/pathology , Pneumonia, Pneumocystis/complications , Pneumonia, Pneumocystis/pathology , Pulmonary Alveoli/immunology , Pulmonary Alveoli/metabolism , Signal Transduction/immunology , fas Receptor/drug effects , fas Receptor/immunology , fas Receptor/metabolism
18.
Am J Physiol Lung Cell Mol Physiol ; 296(3): L489-99, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19036873

ABSTRACT

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.


Subject(s)
Cell Hypoxia/physiology , Insulin-Like Growth Factor Binding Proteins/biosynthesis , Myocytes, Smooth Muscle/metabolism , NADPH Oxidases/metabolism , Pulmonary Artery/metabolism , Transforming Growth Factor beta1/biosynthesis , Autocrine Communication , Cell Hypoxia/genetics , Cell Proliferation , Cells, Cultured , Gene Expression , Humans , Insulin-Like Growth Factor Binding Protein 3 , Insulin-Like Growth Factor Binding Proteins/genetics , Models, Biological , Myocytes, Smooth Muscle/cytology , NADPH Oxidase 4 , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Pulmonary Artery/cytology , RNA Interference , Reactive Oxygen Species/metabolism , Signal Transduction
19.
Am J Physiol Lung Cell Mol Physiol ; 294(6): L1094-101, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18390831

ABSTRACT

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.


Subject(s)
Early Growth Response Protein 1/physiology , Lung/drug effects , Nuclear Proteins/physiology , Receptors, Immunologic/biosynthesis , Respiratory Mucosa/metabolism , Smoking , Transcription Factors/physiology , Animals , Cell Line , Feedback, Physiological , Gene Expression Regulation, Developmental , Lung/growth & development , Mice , Rats , Receptor for Advanced Glycation End Products , Respiratory Mucosa/drug effects , Thyroid Nuclear Factor 1
20.
Am J Physiol Lung Cell Mol Physiol ; 294(1): L139-48, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17993584

ABSTRACT

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.


Subject(s)
Lung/physiology , Muscle Relaxation/physiology , Muscle, Smooth/physiology , NADPH Oxidases/deficiency , Protein Subunits/deficiency , Respiratory Physiological Phenomena , Animals , Crosses, Genetic , Mice , Mice, Knockout , Muscle, Smooth/anatomy & histology , Muscle, Smooth/cytology , Muscle, Smooth/pathology , Weight-Bearing
SELECTION OF CITATIONS
SEARCH DETAIL
...