Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Antioxidants (Basel) ; 13(2)2024 Jan 23.
Article in English | MEDLINE | ID: mdl-38397736

ABSTRACT

We previously reported that maternal alcohol use increased the risk of sepsis in premature and term newborns. In the neonatal mouse, fetal ethanol (ETOH) exposure depleted the antioxidant glutathione (GSH), which promoted alveolar macrophage (AM) immunosuppression and respiratory syncytial virus (RSV) infections. In this study, we explored if oral liposomal GSH (LGSH) would attenuate oxidant stress and RSV infections in the ETOH-exposed mouse pups. C57BL/6 female mice were pair-fed a liquid diet with 25% of calories from ethanol or maltose-dextrin. Postnatal day 10 pups were randomized to intranasal saline, LGSH, and RSV. After 48 h, we assessed oxidant stress, AM immunosuppression, pulmonary RSV burden, and acute lung injury. Fetal ETOH exposure increased oxidant stress threefold, lung RSV burden twofold and acute lung injury threefold. AMs were immunosuppressed with decreased RSV clearance. However, LGSH treatments of the ETOH group normalized oxidant stress, AM immune phenotype, the RSV burden, and acute lung injury. These studies suggest that the oxidant stress caused by fetal ETOH exposure impaired AM clearance of infectious agents, thereby increasing the viral infection and acute lung injury. LGSH treatments reversed the oxidative stress and restored AM immune functions, which decreased the RSV infection and subsequent acute lung injury.

2.
Alcohol ; 106: 30-43, 2023 02.
Article in English | MEDLINE | ID: mdl-36328183

ABSTRACT

Alcohol use disorders (AUD) cause alveolar macrophage (AM) immune dysfunction and increase risk of lung infections. Excessive alcohol use causes AM oxidative stress, which impairs AM phagocytosis and pathogen clearance from the alveolar space. Alcohol induces expression of NADPH oxidases (Noxes), primary sources of oxidative stress in AM. In contrast, alcohol decreases AM peroxisome proliferator-activated receptor gamma (PPARγ), a critical regulator of AM immune function. To explore the underlying molecular mechanisms for these effects of alcohol, we hypothesized that ethanol promotes CCAAT/enhancer-binding protein beta (C/EBPß)-mediated suppression of Nox-related microRNAs (miRs), in turn enhancing AM Nox expression, oxidative stress, and phagocytic dysfunction. We also hypothesized that PPARγ activation with pioglitazone (PIO) would reverse alcohol-induced C/EBPß expression and attenuate AM oxidative stress and phagocytic dysfunction. Cells from the mouse AM cell line (MH-S) were exposed to ethanol in vitro or primary AM were isolated from mice fed ethanol in vivo. Ethanol enhanced C/EBPß expression, decreased Nox 1-related miR-1264 and Nox 2-related miR-107 levels, and increased Nox1, Nox2, and Nox 4 expression in MH-S cells in vitro and mouse AM in vivo. These alcohol-induced AM derangements were abrogated by loss of C/EBPß, overexpression of miRs-1264 or -107, or PIO treatment. These findings identify C/EBPß and Nox-related miRs as novel therapeutic targets for PPARγ ligands, which could provide a translatable strategy to mitigate susceptibility to lung infections in people with a history of AUD. These studies further clarify the molecular underpinnings for a previous clinical trial using short-term PIO treatment to improve AM immunity in AUD individuals.


Subject(s)
Ethanol , Macrophages, Alveolar , MicroRNAs , RNA Processing, Post-Transcriptional , Animals , Mice , Alcoholism/drug therapy , Alcoholism/genetics , Ethanol/adverse effects , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/pathology , MicroRNAs/genetics , MicroRNAs/metabolism , PPAR gamma/genetics , PPAR gamma/metabolism
3.
J Immunol ; 207(2): 483-492, 2021 07 15.
Article in English | MEDLINE | ID: mdl-34193599

ABSTRACT

Alcohol use disorders (AUD) increase susceptibility to respiratory infections by 2- to 4-fold in part because of impaired alveolar macrophage (AM) immune function. Alcohol causes AM oxidative stress, diminishing AM phagocytic capacity and clearance of microbes from the alveolar space. Alcohol increases AM NADPH oxidases (Noxes), primary sources of AM oxidative stress, and reduces peroxisome proliferator-activated receptor γ (PPARγ) expression, a critical regulator of AM immune function. To investigate the underlying mechanisms of these alcohol-induced AM derangements, we hypothesized that alcohol stimulates CCAAT/enhancer-binding protein ß (C/EBPß) to suppress Nox-related microRNAs (miRs), thereby enhancing AM Nox expression, oxidative stress, and phagocytic dysfunction. Furthermore, we postulated that pharmacologic PPARγ activation with pioglitazone would inhibit C/EBPß and attenuate alcohol-induced AM dysfunction. AM isolated from human AUD subjects or otherwise healthy control subjects were examined. Compared with control AM, alcohol activated AM C/EBPß, decreased Nox1-related miR-1264 and Nox2-related miR-107, and increased Nox1, Nox2, and Nox4 expression and activity. These alcohol-induced AM derangements were abrogated by inhibition of C/EBPß, overexpression of miR-1264 or miR-107, or pioglitazone treatment. These findings define novel molecular mechanisms of alcohol-induced AM dysfunction mediated by C/EBPß and Nox-related miRs that are amenable to therapeutic targeting with PPARγ ligands. These results demonstrate that PPARγ ligands provide a novel and rapidly translatable strategy to mitigate susceptibility to respiratory infections and related morbidity in individuals with AUD.


Subject(s)
Alcoholism/drug therapy , Alcoholism/metabolism , Ethanol/adverse effects , Macrophages, Alveolar/drug effects , Phagocytes/drug effects , Pioglitazone/pharmacology , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cell Line , Humans , Lung/drug effects , Lung/metabolism , Macrophages, Alveolar/metabolism , Male , NADPH Oxidases/metabolism , Oxidative Stress/drug effects , PPAR gamma/metabolism , Phagocytes/metabolism
4.
Alcohol ; 90: 27-38, 2021 02.
Article in English | MEDLINE | ID: mdl-33278514

ABSTRACT

Excessive alcohol users have increased risk of developing respiratory infections in part due to oxidative stress-induced alveolar macrophage (AM) phagocytic dysfunction. Chronic ethanol exposure increases cellular oxidative stress in AMs via upregulation of NADPH oxidase (Nox) 4, and treatment with the peroxisome proliferator-activated receptor gamma (PPARγ) ligand, rosiglitazone, decreases ethanol-induced Nox4. However, the mechanism by which ethanol induces Nox4 expression and the PPARγ ligand reverses this defect has not been elucidated. Since microRNA (miR)-92a has been predicted to target Nox4 for destabilization, we hypothesized that ethanol exposure decreases miR-92a expression and leads to Nox4 upregulation. Previous studies have implicated mitochondrial-derived oxidative stress in AM dysfunction. We further hypothesized that ethanol increases mitochondrial-derived AM oxidative stress and dysfunction via miR-92a, and that treatment with the PPARγ ligand, pioglitazone, could reverse these derangements. To test these hypotheses, a mouse AM cell line, MH-S cells, was exposed to ethanol in vitro, and primary AMs were isolated from a mouse model of chronic ethanol consumption to measure Nox4, mitochondrial target mRNA (qRT-PCR) and protein levels (confocal microscopy), mitochondria-derived reactive oxygen species (confocal immunofluorescence), mitochondrial fission (electron microscopy), and mitochondrial bioenergetics (extracellular flux analyzer). Ethanol exposure increased Nox4, enhanced mitochondria-derived oxidative stress, augmented mitochondrial fission, and impaired mitochondrial bioenergetics. Transfection with a miR-92a mimic in vitro or pioglitazone treatment in vivo diminished Nox4 levels, resulting in improvements in these ethanol-mediated derangements. These findings demonstrate that pioglitazone may provide a novel therapeutic approach to mitigate ethanol-induced AM mitochondrial derangements.


Subject(s)
Ethanol , Macrophages, Alveolar/pathology , NADPH Oxidase 4/metabolism , Animals , Cell Line , Ethanol/toxicity , Macrophages, Alveolar/metabolism , Mice , NADPH Oxidase 4/genetics , Oxidative Stress , Reactive Oxygen Species/metabolism
5.
Alcohol ; 80: 5-16, 2019 11.
Article in English | MEDLINE | ID: mdl-30580016

ABSTRACT

BACKGROUND: We previously reported that maternal alcohol use significantly increases the risk of sepsis in premature and term newborns. In the mouse, fetal ethanol exposure results in an immunosuppressed phenotype for the alveolar macrophage (AM) and decreases bacterial phagocytosis. In pregnant mice, ethanol decreased AM zinc homeostasis, which contributed to immunosuppression and impaired AM phagocytosis. In this study, we explored whether ethanol-induced zinc insufficiency extended to the pup AMs and contributed to immunosuppression and exacerbated viral lung infections. METHODS: C57BL/6 female mice were fed a liquid diet with 25% ethanol-derived calories or pair-fed a control diet with 25% of calories as maltose-dextrin. Some pup AMs were treated in vitro with zinc acetate before measuring zinc pools or transporter expression and bacteria phagocytosis. Some dams were fed additional zinc supplements in the ethanol or control diets, and then we assessed pup AM zinc pools, zinc transporters, and the immunosuppressant TGFß1. On postnatal day 10, some pups were given intranasal saline or respiratory syncytial virus (RSV), and then AM RSV phagocytosis and the RSV burden in the airway lining fluid were assessed. RESULTS: Fetal ethanol exposure decreased pup AM zinc pools, zinc transporter expression, and bacterial clearance, but in vitro zinc treatments reversed these alterations. In addition, the expected ethanol-induced increase in TGFß1 and immunosuppression were associated with decreased RSV phagocytosis and exacerbated RSV infections. However, additional maternal zinc supplements blocked the ethanol-induced perturbations in the pup AM zinc homeostasis and TGFß1 immunosuppression, thereby improving RSV phagocytosis and attenuating the RSV burden in the lung. CONCLUSION: These studies suggest that, despite normal maternal dietary zinc intake, in utero alcohol exposure results in zinc insufficiency, which contributes to compromised neonatal AM immune functions, thereby increasing the risk of bacterial and viral infections.


Subject(s)
Fetal Alcohol Spectrum Disorders/etiology , Macrophages, Alveolar/drug effects , Respiratory Syncytial Virus Infections/etiology , Zinc/deficiency , Animals , Dietary Supplements , Disease Models, Animal , Female , Fetal Alcohol Spectrum Disorders/immunology , Fetal Alcohol Spectrum Disorders/physiopathology , Immune Tolerance , Macrophages, Alveolar/physiology , Mice , Mice, Inbred C57BL , Pregnancy , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/physiopathology
6.
Physiol Rep ; 5(6)2017 Mar.
Article in English | MEDLINE | ID: mdl-28325787

ABSTRACT

Our understanding of the intrinsic effects of cystic fibrosis (CF) transmembrane conductance regulator (cftr) deletion on resident neonatal alveolar macrophage (AM) remains limited. We previously demonstrated that diminished glutathione (GSH) or excessive AM transforming growth factor beta one (TGFß1) contributes to AM dysfunction in a variety of disease states. In this study, using a gut-corrected cftr neonatal knockout (KO) mouse model and a siRNA-manipulated macrophage-like cell line (THP-1 cell), we hypothesized (1) that cftr mutation alone increases neonatal AM oxidant stress and cellular TGFß1 signaling via altered GSH, thereby impairing cellular function, and (2) that exogenous GSH attenuates AM alterations and dysfunction in the KO AM In neonatal KO mice, the baseline bronchoalveolar lavage fluid demonstrated a near doubling in mixed disulfides (P ≤ 0.05) and oxidized GSSG (P ≤ 0.05) without concurrent inflammation compared to WT littermates. KO AM demonstrated diminished AM thiols (P ≤ 0.05), increased AM mitochondrial ROS (P ≤ 0.05), increased AM TGFß1 (P ≤ 0.05) with increased TGFß1 signaling (P ≤ 0.05), and impaired phagocytosis (P ≤ 0.05). KO AM mitochondrial ROS was modulated by exogenous GSH (P ≤ 0.05). Conversely, TGFß1 was reduced (P ≤ 0.05) and impaired phagocytosis was rescued (P ≤ 0.05) by exogenous GSH in the KO AM These results suggest that an altered neonatal AM phenotype may contribute to the initiation of lung inflammation/infection in the CF lung. Modulation of the AM in the neonatal CF lung may potentially alter progression of disease.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Glutathione/pharmacology , Macrophages, Alveolar/metabolism , Oxidative Stress/physiology , Transforming Growth Factor beta1/metabolism , Animals , Bronchoalveolar Lavage Fluid , Cell Line , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Macrophages, Alveolar/drug effects , Mice , Mice, Inbred CFTR , Mice, Knockout , Oxidative Stress/drug effects , Phagocytosis/drug effects , Phagocytosis/physiology
7.
AIDS Res Hum Retroviruses ; 33(10): 1018-1026, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28314381

ABSTRACT

Despite antiretroviral therapy (ART), respiratory infections increase mortality in individuals living with chronic human immunodeficiency virus (HIV) infection. In experimental and clinical studies of chronic HIV infection, alveolar macrophages (AMs) exhibit impaired phagocytosis and bacterial clearance. Peroxisome proliferator-activated receptor (PPAR)γ, NADPH oxidase (Nox) isoforms Nox1, Nox2, Nox4, and transforming growth factor-beta 1 (TGFß1) are critical mediators of AM oxidative stress and phagocytic dysfunction. Therefore, we hypothesized that HIV alters AM expression of these targets, resulting in chronic lung oxidative stress and subsequent immune dysfunction. A cross-sectional study of HIV-infected (n = 22) and HIV-uninfected (n = 6) subjects was conducted. Bronchoalveolar lavage (BAL) was performed, and AMs were isolated. Lung H2O2 generation was determined by measuring H2O2 in the BAL fluid. In AMs, PPARγ, Nox1, Nox2, Nox4, and TGFß1 mRNA (quantitative real-time polymerase chain reaction) and protein (fluorescent immunomicroscopy) levels were assessed. Compared with HIV-uninfected (control) subjects, HIV-infected subjects were relatively older and the majority were African American; ∼86% were on ART, and their median CD4 count was 445, with a median viral load of 0 log copies/ml. HIV infection was associated with increased H2O2 in the BAL, decreased AM mRNA and protein levels of PPARγ, and increased AM mRNA and protein levels of Nox1, Nox2, Nox4, and TGFß1. PPARγ attenuation and increases in Nox1, Nox2, Nox4, and TGFß1 contribute to AM oxidative stress and immune dysfunction in the AMs of otherwise healthy HIV-infected subjects. These findings provide novel insights into the molecular mechanisms by which HIV increases susceptibility to pulmonary infections.


Subject(s)
HIV Infections/pathology , Macrophages, Alveolar/immunology , NADPH Oxidases/metabolism , Oxidative Stress/immunology , PPAR gamma/metabolism , Transforming Growth Factor beta1/metabolism , Adult , Anti-HIV Agents/therapeutic use , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , CD4 Lymphocyte Count , Cells, Cultured , Cross-Sectional Studies , Female , HIV Infections/drug therapy , HIV Infections/virology , Humans , Hydrogen Peroxide/analysis , Male , Middle Aged , NADPH Oxidases/genetics , PPAR gamma/genetics , Phagocytosis/immunology , RNA, Messenger/genetics , Transforming Growth Factor beta1/genetics , Viral Load
8.
Am J Respir Cell Mol Biol ; 55(1): 35-46, 2016 07.
Article in English | MEDLINE | ID: mdl-26677910

ABSTRACT

Peroxisome proliferator-activated receptor (PPAR) γ is critical for alveolar macrophage (AM) function. Chronic alcohol abuse causes AM phagocytic dysfunction and susceptibility to respiratory infections by stimulating nicotinamide adenine dinucleotide oxidases (Nox), transforming growth factor-ß1, and oxidative stress in the AM. Because PPARγ inhibits Nox expression, we hypothesized that alcohol reduces PPARγ, stimulating AM dysfunction. AMs were examined from: (1) patients with alcoholism or control patients; (2) a mouse model of chronic ethanol consumption; (3) PPARγ knockout mice; or (4) MH-S cells exposed to ethanol in vitro. Alcohol reduced AM PPARγ levels and increased Nox1, -2, and -4, transforming growth factor-ß1, oxidative stress, and phagocytic dysfunction. Genetic loss of PPARγ recapitulated, whereas stimulating PPARγ activity attenuated alcohol-mediated alterations in gene expression and phagocytic function, supporting the importance of PPARγ in alcohol-induced AM derangements. Similarly, PPARγ activation in vivo reduced alcohol-mediated impairments in lung bacterial clearance. Alcohol increased levels of microRNA-130a/-301a, which bind to the PPARγ 3' untranslated region to reduce PPARγ expression. MicroRNA-130a/-301a inhibition attenuated alcohol-mediated PPARγ reductions and derangements in AM gene expression and function. Alcohol-induced Toll-like receptor 4 endocytosis was reversed by PPARγ activation. These findings demonstrate that targeting PPARγ provides a novel therapeutic approach for mitigating alcohol-induced AM derangements and susceptibility to lung infection.


Subject(s)
Ethanol/adverse effects , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/pathology , PPAR gamma/metabolism , Animals , Cell Line , Humans , Klebsiella/drug effects , Ligands , Lung/microbiology , Lung/pathology , Male , Mice, Inbred C57BL , MicroRNAs/metabolism , Oxidative Stress/drug effects , Phagocytes/drug effects , Phagocytes/pathology , Rosiglitazone , Thiazolidinediones/pharmacology
10.
PLoS One ; 10(6): e0129570, 2015.
Article in English | MEDLINE | ID: mdl-26102199

ABSTRACT

An alcohol use disorder (AUD) is associated with an increased susceptibility to respiratory infection and injury and, upon hospitalization, higher mortality rates. Studies in model systems show effects of alcohol on mitochondrial function, lipid metabolism and antioxidant systems. The present study applied high-resolution metabolomics to test for these changes in bronchoalveolar lavage fluid (BALF) of subjects with an AUD. Smokers were excluded to avoid confounding effects and compliance was verified by cotinine measurements. Statistically significant metabolic features, differentially expressed by control and AUD subjects, were identified by statistical and bioinformatic methods. The results show that fatty acid and acylcarnitine concentrations were increased in AUD subjects, consistent with perturbed mitochondrial and lipid metabolism. Decreased concentrations of methyl-donor compounds suggest altered one-carbon metabolism and oxidative stress. An accumulation of peptides suggests proteolytic activity, which could reflect altered epithelial barrier function. Two metabolites of possible microbial origin suggest subclinical bacterial infection. Furthermore, increased diacetylspermine suggests additional metabolic perturbations, which could contribute to dysregulated alveolar macrophage function and vulnerability to infection. Together, the results show an extended metabolic consequence of AUD in the bronchoalveolar space.


Subject(s)
Alcoholism/metabolism , Antioxidants/metabolism , Biomarkers/metabolism , Bronchoalveolar Lavage Fluid , Chronic Disease , Humans , Lipid Metabolism , Mass Spectrometry , Metabolomics , Mitochondria/physiology , Oxidative Stress , Pilot Projects , Smoking
11.
PLoS One ; 10(5): e0126552, 2015.
Article in English | MEDLINE | ID: mdl-25978403

ABSTRACT

The accumulation of fatty acid ethyl esters (FAEEs) in meconium of term newborns has been described as one potential biomarker of maternal alcohol use during pregnancy. FAEEs accumulate in multiple alcohol-exposed fetal tissues and in the placenta. Limited research has focused on the identification of the premature newborn exposed to alcohol in utero. We hypothesized that maternal alcohol use occurs in a significant proportion of premature deliveries and that this exposure can be detected as elevated placental FAEEs. The goals of this study were to 1) determine the prevalence of maternal alcohol use in the premature newborn and 2) investigate whether placental FAEEs could identify those newborns with fetal alcohol exposure. This prospective observational study evaluated 80 placentas from 80 women after premature delivery. Subjects were interviewed for alcohol intake and placental FAEEs were quantified via GC/MS. Receiver Operator Characteristic (ROC) Curves were generated to evaluate the ability of placental FAEEs to predict maternal drinking during pregnancy. Adjusted ROC curves were generated to adjust for gestational age, maternal smoking, and illicit drug use. 30% of the subjects admitted to drinking alcohol during pregnancy and approximately 14% answered questions indicative of problem drinking (designated AUDIT+). The specific FAEEs ethyl stearate and linoleate, as well as combinations of oleate + linoleate + linolenate (OLL) and of OLL + stearate, were significantly (p<0.05) elevated in placentas from AUDIT+ pregnancies. Adjusted ROC Curves generated areas under the curve ranging from 88-93% with negative predictive values of 97% for AUDIT+ pregnancies. We conclude that nearly one third of premature pregnancies were alcohol-exposed, and that elevated placental FAEEs hold great promise to accurately determine maternal alcohol use, particularly heavy use, in pregnancies complicated by premature delivery.


Subject(s)
Alcohol Drinking/adverse effects , Esters/analysis , Esters/metabolism , Fatty Acids/metabolism , Infant, Premature/metabolism , Meconium/metabolism , Adult , Female , Humans , Infant, Newborn , Pregnancy , Prospective Studies , Surveys and Questionnaires , Young Adult
12.
Alcohol Clin Exp Res ; 39(3): 434-44, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25703924

ABSTRACT

BACKGROUND: Chronic alcohol exposure alters the function of alveolar macrophages (AM), impairing immune defenses in both adult and neonatal lungs. Fatty acid ethyl esters (FAEEs) are biological markers of prenatal alcohol exposure in newborns. FAEEs contribute to alcohol-induced mitochondrial (MT) damage in multiple organs. We hypothesized that in utero ethanol exposure would increase FAEEs in the neonatal lung and that direct exposure of neonatal AM to FAEEs would contribute to MT injury and cellular dysfunction. METHODS: FAEEs were measured in neonatal guinea pig lungs after ± in utero ethanol exposure via gas chromatography/mass spectrometry. The NR8383 cell line and freshly isolated neonatal guinea pig AM were exposed to ethyl oleate (EO) in vitro. MT membrane potential, MT reactive oxygen species generation (mROS), phagocytosis, and apoptosis were evaluated after exposure to EO ± the MT-specific antioxidant mito-TEMPO (mitoT) or ± the pan-caspase inhibitor Z-VAD-FMK. Whole lung FAEEs were compared using the Mann-Whitney U-test. Cellular results were analyzed using 1-way analysis of variance, followed by the Student-Newman-Keuls Method for post hoc comparisons. RESULTS: In utero ethanol significantly increased ethyl linoleate and the combinations of ethyl oleate + linoleate + linolenate (OLL), and OLL + stearate in the neonatal lung. In vitro EO caused significant MT dysfunction in both NR8383 and primary neonatal AM, as indicated by increased mROS and loss of MT membrane potential. Impaired phagocytosis and apoptosis were significantly increased in both the cell line and primary AM after EO exposure. MitoT conferred significant but only partial protection against EO-induced MT injury, as did caspase inhibition with Z-VAD-FMK. CONCLUSIONS: In utero ethanol exposure increased FAEEs in the neonatal guinea pig lung. Direct exposure to the FAEE EO significantly contributed to AM dysfunction, in part via oxidant injury to the MT and in part via secondary apoptosis.


Subject(s)
Ethanol/toxicity , Fatty Acids, Nonesterified/toxicity , Lung/cytology , Lung/drug effects , Macrophages, Alveolar/drug effects , Mitochondria/drug effects , Animals , Animals, Newborn , Female , Guinea Pigs , Lung/metabolism , Macrophages, Alveolar/metabolism , Mitochondria/metabolism , Pregnancy , Prenatal Exposure Delayed Effects/metabolism
13.
Alcohol Alcohol ; 50(1): 30-8, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25371044

ABSTRACT

AIMS: (a) Establish the minimum number of weeks of chronic ethanol ingestion needed to perturb zinc homeostasis, (b) Examine intracellular zinc status in the alveolar macrophages (AMs) when ethanol ingestion is combined with pregnancy, (c) Investigate whether in vitro zinc treatment reverses the effects of ethanol ingestion on the AM. METHODS: C57BL/6 female mice were fed a liquid diet (±25% ethanol-derived calories) during preconception and pregnancy. The control group was pair-fed to the ethanol group. In the isolated AMs, we measured intracellular AM zinc levels, zinc transporter expression, alternative activation and phagocytic index. Zinc acetate was added to some cells prior to analysis. RESULTS: Intracellular zinc levels in the AM decreased within 3 weeks of ethanol ingestion. After ethanol ingestion prior to and during pregnancy, zinc transporter expression and intracellular zinc levels were decreased in the AMs when compared with controls. Bacterial clearance was decreased because the AMs were alternatively activated. In vitro additions of zinc reversed these effects of ethanol. CONCLUSION: Ethanol ingestion prior to and during pregnancy perturbed AM zinc balance resulting in impaired bacterial clearance, but these effects were ameliorated by in vitro zinc treatments.


Subject(s)
Ethanol/adverse effects , Macrophages, Alveolar/drug effects , Zinc/deficiency , Animals , Carrier Proteins/analysis , Carrier Proteins/biosynthesis , Female , Macrophages, Alveolar/chemistry , Macrophages, Alveolar/physiology , Mice , Mice, Inbred C57BL , Phagocytosis/drug effects , Pregnancy , Pregnancy Complications/chemically induced , Zinc/analysis
14.
Biomed Res Int ; 2014: 371593, 2014.
Article in English | MEDLINE | ID: mdl-24701574

ABSTRACT

An alcohol use disorder increases the risk of invasive and antimicrobial resistant community-acquired pneumonia and tuberculosis. Since the alveolar macrophage (AM) orchestrates the immune response in the alveolar space, understanding the underlying mechanisms by which alcohol suppresses AM phagocytosis is critical to improving clinical outcomes. In the alveolar space, chronic alcohol ingestion causes severe oxidative stress and depletes antioxidants which are critical for AM function. The mitochondrion is important in maintaining cellular redox balance and providing the ATP critical for phagocytosis. The focus of this study was to understand how alcohol triggers mitochondrial reactive oxygen species (ROS), stimulates cellular oxidative stress, and induces AM dysfunction. The current study also investigated the capacity of the mitochondrial targeted antioxidant, mitoTEMPOL (mitoT), in modulating mitochondrial oxidative stress, and AM dysfunction. Using in vitro ethanol exposure and AMs from ethanol-fed mice, ethanol promoted mitochondrial dysfunction including increased mitochondrial ROS, decreased mitochondrial membrane potential, and decreased ATP. Treatment with mitoT reversed these effects. Ethanol-induced decreases in phagocytosis and cell viability were also attenuated with mitoT. Therefore, antioxidants targeted to the mitochondria have the potential to ameliorate ethanol-induced mitochondrial oxidative stress and subsequent decreases in AM phagocytosis and cell viability.


Subject(s)
Alcoholism/metabolism , Mitochondria/pathology , Oxidative Stress , Pulmonary Alveoli/pathology , Alcoholism/pathology , Animals , Cell Survival/drug effects , Ethanol/toxicity , Humans , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/pathology , Mice , Mitochondria/drug effects , Phagocytosis/drug effects , Pulmonary Alveoli/metabolism , Reactive Oxygen Species
15.
Am J Physiol Lung Cell Mol Physiol ; 306(5): L429-41, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24441868

ABSTRACT

Chronic alcohol abuse increases lung oxidative stress and susceptibility to respiratory infections by impairing alveolar macrophage (AM) function. NADPH oxidases (Nox) are major sources of reactive oxygen species in AMs. We hypothesized that treatment with the critical antioxidant glutathione (GSH) attenuates chronic alcohol-induced oxidative stress by downregulating Noxes and restores AM phagocytic function. Bronchoalveolar lavage (BAL) fluid and AMs were isolated from male C57BL/6J mice (8-10 wk) treated ± ethanol in drinking water (20% wt/vol, 12 wk) ± orally gavaged GSH in methylcellulose vehicle (300 mg x kg(-1) x day(-1), during week 12). MH-S cells, a mouse AM cell line, were treated ± ethanol (0.08%, 3 days) ± GSH (500 µM, 3 days or last 1 day of ethanol). BAL and AMs were also isolated from ethanol-fed and control mice ± inoculated airway Klebsiella pneumoniae (200 colony-forming units, 28 h) ± orally gavaged GSH (300 mg/kg, 24 h). GSH levels (HPLC), Nox mRNA (quantitative RT-PCR) and protein levels (Western blot and immunostaining), oxidative stress (2',7'-dichlorofluorescein-diacetate and Amplex Red), and phagocytosis (Staphylococcus aureus internalization) were measured. Chronic alcohol decreased GSH levels, increased Nox expression and activity, enhanced oxidative stress, impaired phagocytic function in AMs in vivo and in vitro, and exacerbated K. pneumonia-induced oxidative stress. Although how oral GSH restored GSH pools in ethanol-fed mice is unknown, oral GSH treatments abrogated the detrimental effects of chronic alcohol exposure and improved AM function. These studies provide GSH as a novel therapeutic approach for attenuating alcohol-induced derangements in AM Nox expression, oxidative stress, dysfunction, and risk for pneumonia.


Subject(s)
Alcoholism/immunology , Antioxidants/metabolism , Glutathione/metabolism , Macrophages, Alveolar/immunology , NADH, NADPH Oxidoreductases/metabolism , Alcoholism/metabolism , Animals , Antioxidants/pharmacology , Bronchoalveolar Lavage Fluid/immunology , Cell Line , Central Nervous System Depressants/pharmacology , Down-Regulation/drug effects , Down-Regulation/immunology , Enzyme Activation/drug effects , Enzyme Activation/immunology , Ethanol/pharmacology , Glutathione/pharmacology , Klebsiella Infections/immunology , Klebsiella Infections/metabolism , Klebsiella pneumoniae/immunology , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/metabolism , Male , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , NADPH Oxidase 1 , NADPH Oxidase 2 , NADPH Oxidase 4 , NADPH Oxidases/metabolism , Oxidative Stress/drug effects , Oxidative Stress/immunology , Phosphoproteins/metabolism
16.
Free Radic Biol Med ; 65: 1427-1434, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24140864

ABSTRACT

Alcohol abuse suppresses the immune responses of alveolar macrophages (AMs) and increases the risk of a respiratory infection via chronic oxidative stress and depletion of critical antioxidants within alveolar cells and the alveolar lining fluid. Although alcohol-induced mitochondrial oxidative stress has been demonstrated, the oxidation of the mitochondrial thioredoxin redox circuit in response to alcohol has not been examined. In vitro ethanol exposure of a mouse AM cell line and AMs from ethanol-fed mice demonstrated NADPH depletion concomitant with oxidation of mitochondrial glutathione and oxidation of the thioredoxin redox circuit system including thioredoxin 2 (Trx2) and thioredoxin 2 reductase (Trx2R). Mitochondrial peroxiredoxins (Prdx's), which are critical for the reduction of the thioredoxin circuit, were irreversibly hyperoxidized to an inactive form. Ethanol also decreased the mRNAs for Trx2, Trx2R, Prdx3, and Prdx5 plus the mitochondrial thiol-disulfide proteins glutaredoxin 2, glutathione reductase, and glutathione peroxidase 2. Thus, the mitochondrial thioredoxin circuit was highly oxidized by ethanol, thereby compromising the mitochondrial antioxidant capacity and ability to detoxify mitochondrial reactive oxygen species. Oxidation of the mitochondrial thioredoxin redox circuit would further compromise the transient oxidation of thiol groups within specific proteins, the basis of redox signaling, and the processes by which cells respond to oxidants. Impaired mitochondria can then jeopardize cellular function of AMs, such as phagocytosis, which may explain the increased risk of respiratory infection in subjects with an alcohol use disorder.


Subject(s)
Ethanol/pharmacology , Macrophages, Alveolar/immunology , Mitochondria/pathology , Phagocytosis/drug effects , Alcohol Drinking , Animals , Cell Line , Central Nervous System Depressants/administration & dosage , Central Nervous System Depressants/pharmacology , Ethanol/administration & dosage , Glutaredoxins/biosynthesis , Glutathione/metabolism , Glutathione Peroxidase , Glutathione Reductase/biosynthesis , Mice , Mice, Inbred C57BL , NADP/metabolism , Oxidation-Reduction/drug effects , Oxidative Stress , Peroxiredoxins/metabolism , Phagocytosis/immunology , RNA, Messenger/biosynthesis , Reactive Oxygen Species/metabolism , Signal Transduction/immunology , Thioredoxin-Disulfide Reductase/genetics , Thioredoxin-Disulfide Reductase/metabolism , Thioredoxins/metabolism
17.
J Immunol ; 188(8): 3648-57, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22412195

ABSTRACT

Chronic alcohol abuse is a comorbid variable of acute respiratory distress syndrome. Previous studies showed that, in the lung, chronic alcohol consumption increased oxidative stress and impaired alveolar macrophage (AM) function. NADPH oxidases (Noxes) are the main source of reactive oxygen species in AMs. Therefore, we hypothesized that chronic alcohol consumption increases AM oxidant stress through modulation of Nox1, Nox2, and Nox4 expression. AMs were isolated from male C57BL/6J mice, aged 8-10 wk, which were treated with or without ethanol in drinking water (20% w/v, 12 wk). MH-S cells, a mouse AM cell line, were treated with or without ethanol (0.08%, 3 d) for in vitro studies. Selected cells were treated with apocynin (300 µM), a Nox1 and Nox2 complex formation inhibitor, or were transfected with Nox small interfering RNAs (20-35 nM), before ethanol exposure. Human AMs were isolated from alcoholic and control patients' bronchoalveolar lavage fluid. Nox mRNA levels (quantitative RT-PCR), protein levels (Western blot and immunostaining), oxidative stress (2',7'-dichlorofluorescein-diacetate and Amplex Red analysis), and phagocytosis (Staphylococcus aureus internalization) were measured. Chronic alcohol increased Nox expression and oxidative stress in mouse AMs in vivo and in vitro. Experiments using apocynin and Nox small interfering RNAs demonstrated that ethanol-induced Nox4 expression, oxidative stress, and AM dysfunction were modulated through Nox1 and Nox2 upregulation. Further, Nox1, Nox2, and Nox4 protein levels were augmented in human AMs from alcoholic patients compared with control subjects. Ethanol induces AM oxidative stress initially through upregulation of Nox1 and Nox2 with downstream Nox4 upregulation and subsequent impairment of AM function.


Subject(s)
Ethanol/pharmacology , Lung/drug effects , Macrophages, Alveolar/drug effects , NADPH Oxidases/genetics , Acetophenones/pharmacology , Adult , Alcoholism/enzymology , Alcoholism/genetics , Alcoholism/immunology , Animals , Bronchoalveolar Lavage Fluid/cytology , Cell Line , Humans , Isoenzymes/genetics , Isoenzymes/immunology , Lung/enzymology , Lung/immunology , Macrophages, Alveolar/enzymology , Macrophages, Alveolar/immunology , Male , Mice , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/immunology , Oxidative Stress/drug effects , Phagocytosis/drug effects , RNA, Messenger/biosynthesis , RNA, Messenger/immunology , RNA, Small Interfering/genetics , Reactive Oxygen Species/immunology , Reactive Oxygen Species/metabolism , Signal Transduction , Up-Regulation
18.
Alcohol Clin Exp Res ; 33(2): 300-6, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19032578

ABSTRACT

BACKGROUND: The effects of fetal alcohol exposure on the risks of neonatal lung injury and infection remain under investigation. The resident alveolar macrophage (AM) is the first line of immune defense against pulmonary infections. In utero ethanol (ETOH) exposure deranges the function of both premature and term guinea pig AM. We hypothesized that fetal ETOH exposure would increase the risk of pulmonary infection in vivo. METHODS: We developed a novel in vivo model of group B Streptococcus (GBS) pneumonia using our established guinea pig model of fetal ETOH exposure. Timed-pregnant guinea pigs were pair fed +/-ETOH and some were supplemented with the glutathione (GSH) precursor S-adenosyl-methionine (SAM-e). Term pups were given GBS intratracheally while some were pretreated with inhaled GSH prior to the experimental GBS. Neonatal lung and whole blood were evaluated for GBS while isolated AM were evaluated using fluorescent microscopy for GBS phagocytosis. RESULTS: Ethanol-exposed pups demonstrated increased lung infection and sepsis while AM phagocytosis of GBS was deficient compared with control. When SAM-e was added to the maternal diet containing ETOH, neonatal lung and systemic infection from GBS was attenuated and AM phagocytosis was improved. Inhaled GSH therapy prior to GBS similarly protected the ETOH-exposed pup from lung and systemic infection. CONCLUSIONS: In utero ETOH exposure impaired the neonatal lung's defense against experimental GBS, while maintaining GSH availability protected the ETOH-exposed lung. This study suggested that fetal alcohol exposure deranges the neonatal lung's defense against bacterial infection, and support further investigations into the potential therapeutic role for exogenous GSH to augment neonatal AM function.


Subject(s)
Central Nervous System Depressants/toxicity , Ethanol/toxicity , Pneumonia, Bacterial/immunology , Streptococcal Infections/immunology , Streptococcus agalactiae , Animals , Animals, Newborn , Antidotes/pharmacokinetics , Antidotes/pharmacology , Body Weight/drug effects , Central Nervous System Depressants/antagonists & inhibitors , Ethanol/antagonists & inhibitors , Female , Glutathione/pharmacokinetics , Glutathione/pharmacology , Guinea Pigs , Lung/pathology , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/immunology , Microscopy, Confocal , Phagocytosis/drug effects , Pneumonia, Bacterial/pathology , Pregnancy , Respiratory Function Tests , S-Adenosylmethionine/administration & dosage , S-Adenosylmethionine/pharmacology , Sepsis/microbiology , Streptococcal Infections/pathology
19.
Alcohol Clin Exp Res ; 31(2): 308-16, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17250624

ABSTRACT

BACKGROUND: The effects of in utero alcohol exposure on the immune function of the newborn remain under investigation. Fetal ethanol (ETOH) exposure increases oxidative stress in the developing lung, in part due to decreased availability of the antioxidant glutathione (GSH). We have previously shown that in utero ETOH impairs alveolar macrophage phagocytosis and viability in the premature pup, while maintaining GSH availability with maternal supplementation of S-adenosyl-methionine (SAM) during ETOH ingestion improves macrophage function and viability. We hypothesized that dysfunction of the neonatal alveolar macrophage exposed to ETOH in utero would persist at term gestation. METHODS: Using a guinea-pig model of fetal ETOH exposure, timed-pregnant guinea-pigs were pair-fed ETOH+/-the GSH precursor SAM and the diet continued until spontaneous delivery. Term alveolar macrophages were evaluated using fluorescent microscopy for phagocytosis and apoptosis after in vitro incubation with Staphalococcus aureus. Using an in vivo model of intranasal Staph. aureus inoculation, the in vivo function of the term alveolar macrophage was also investigated using confocal fluorescent analysis. RESULTS: In utero ETOH exposure increased oxidant stress in the alveolar macrophage and decreased phagocytosis and viability in vitro and in vivo. Confocal analysis of phagocytosis in vivo demonstrated a marked impairment of internalization of the bacteria by the ETOH-exposed alveolar macrophage. The addition of SAM during maternal ETOH ingestion prevented loss of alveolar macrophage function and viability in vitro and in vivo. CONCLUSIONS: In utero ETOH exposure impairs alveolar macrophage function and viability in vitro and in vivo even at term gestation. The ETOH-induced changes in macrophage function and viability can be ablated with maternal SAM supplementation. Further investigations are required to identify the mechanisms of ETOH-induced derangement of phagocytosis in the neonatal alveolar macrophage and the clinical ramifications of altered immune function after in utero alcohol exposure for the newborn.


Subject(s)
Central Nervous System Depressants/toxicity , Ethanol/toxicity , Macrophages, Alveolar/drug effects , Maternal Exposure/adverse effects , Animals , Animals, Newborn , Apoptosis/drug effects , Apoptosis/physiology , Cell Survival/drug effects , Cell Survival/physiology , Female , Glutathione/metabolism , Guinea Pigs , Macrophages, Alveolar/cytology , Macrophages, Alveolar/physiology , Oxidative Stress/drug effects , Oxidative Stress/physiology , Phagocytosis/drug effects , Phagocytosis/physiology , Pregnancy , Prenatal Exposure Delayed Effects , Staphylococcus aureus/metabolism
20.
Am J Physiol Lung Cell Mol Physiol ; 292(4): L824-32, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17122355

ABSTRACT

We have previously demonstrated that chronic alcohol exposure decreases glutathione in the alveolar space. Although alcohol use is associated with decreased alveolar macrophage function, the mechanism by which alcohol impairs macrophage phagocytosis is unknown. In the current study, we examined the possibility that ethanol-induced alveolar macrophage dysfunction was secondary to decreased glutathione and subsequent chronic oxidative stress in the alveolar space. After 6 wk of ethanol ingestion, oxidant stress in the alveolar macrophages was evidenced by a 30-mV oxidation of the GSH/GSSG redox potential (P

Subject(s)
Alcoholism/physiopathology , Glutathione/physiology , Macrophages, Alveolar/physiology , Animals , Apoptosis/drug effects , Cell Survival/drug effects , Cells, Cultured , Homeostasis , In Situ Nick-End Labeling , Male , Microscopy, Confocal , Oxidative Stress/physiology , Phagocytosis/drug effects , Poly(ADP-ribose) Polymerases/metabolism , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...