Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Am J Physiol Lung Cell Mol Physiol ; 325(5): L647-L661, 2023 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-37786945

RESUMO

Alcohol use disorder (AUD) is a significant public health concern and people with AUD are more likely to develop severe acute respiratory distress syndrome (ARDS) in response to respiratory infections. To examine whether AUD was a risk factor for more severe outcome in response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, we examined early responses to infection using cultured differentiated bronchial epithelial cells derived from brushings obtained from people with AUD or without AUD. RNA-seq analysis of uninfected cells determined that AUD cells were enriched for expression of epidermal genes as compared with non-AUD cells. Bronchial epithelial cells from patients with AUD showed a significant decrease in barrier function 72 h postinfection, as determined by transepithelial electrical resistance. In contrast, barrier function of non-AUD cells was enhanced 72 h after SARS-CoV-2 infection. AUD cells showed claudin-7 that did not colocalize with zonula occludens-1 (ZO-1), indicative of disorganized tight junctions. However, both AUD and non-AUD cells showed decreased ß-catenin expression following SARS-CoV-2 infection. To determine the impact of AUD on the inflammatory response to SARS-CoV-2 infection, cytokine secretion was measured by multiplex analysis. SARS-CoV-2-infected AUD bronchial cells had enhanced secretion of multiple proinflammatory cytokines including TNFα, IL-1ß, and IFNγ as opposed to non-AUD cells. In contrast, secretion of the barrier-protective cytokines epidermal growth factor (EGF) and granulocyte macrophage-colony stimulating factor (GM-CSF) was enhanced for non-AUD bronchial cells. Taken together, these data support the hypothesis that AUD is a risk factor for COVID-19, where alcohol primes airway epithelial cells for increased inflammation and increased barrier dysfunction and increased inflammation in response to infection by SARS-CoV-2.NEW & NOTEWORTHY Alcohol use disorder (AUD) is a significant risk factor for severe acute respiratory distress syndrome. We found that AUD causes a phenotypic shift in gene expression in human bronchial epithelial cells, enhancing expression of epidermal genes. AUD cells infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) had higher levels of proinflammatory cytokine secretion and barrier dysfunction not present in infected non-AUD cells, consistent with increased early COVID-19 severity due to AUD.


Assuntos
Alcoolismo , COVID-19 , Síndrome do Desconforto Respiratório , Humanos , SARS-CoV-2/metabolismo , Citocinas/metabolismo , Inflamação
2.
J Immunol ; 207(2): 483-492, 2021 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-34193599

RESUMO

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.


Assuntos
Alcoolismo/tratamento farmacológico , Alcoolismo/metabolismo , Etanol/efeitos adversos , Macrófagos Alveolares/efeitos dos fármacos , Fagócitos/efeitos dos fármacos , Pioglitazona/farmacologia , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Linhagem Celular , Humanos , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Macrófagos Alveolares/metabolismo , Masculino , NADPH Oxidases/metabolismo , Estresse Oxidativo/efeitos dos fármacos , PPAR gama/metabolismo , Fagócitos/metabolismo
3.
Am J Respir Cell Mol Biol ; 55(1): 35-46, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26677910

RESUMO

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.


Assuntos
Etanol/efeitos adversos , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/patologia , PPAR gama/metabolismo , Animais , Linhagem Celular , Humanos , Klebsiella/efeitos dos fármacos , Ligantes , Pulmão/microbiologia , Pulmão/patologia , Masculino , Camundongos Endogâmicos C57BL , MicroRNAs/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Fagócitos/efeitos dos fármacos , Fagócitos/patologia , Rosiglitazona , Tiazolidinedionas/farmacologia
4.
Am J Respir Crit Care Med ; 188(6): 716-23, 2013 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-23805851

RESUMO

RATIONALE: Alcohol use disorders cause oxidative stress in the lower airways and increase susceptibility to pneumonia and lung injury. Currently, no therapeutic options exist to mitigate the pulmonary consequences of alcoholism. OBJECTIVES: We recently determined in an animal model that alcohol ingestion impairs pulmonary zinc metabolism and causes alveolar macrophage immune dysfunction. The objective of this research is to determine the effects of alcoholism on zinc bioavailability and alveolar macrophage function in human subjects. METHODS: We recruited otherwise healthy alcoholics (n = 17) and matched control subjects (n = 17) who underwent bronchoscopy for isolation of alveolar macrophages, which were analyzed for intracellular zinc, phagocytic function, and surface expression of granulocyte-macrophage colony-stimulating factor receptor; all three of these indices are decreased in experimental models. MEASUREMENTS AND MAIN RESULTS: Alcoholic subjects had normal serum zinc, but significantly decreased alveolar macrophage intracellular zinc levels (adjusted means [SE], 718 [41] vs. 948 [25] RFU/cell; P < 0.0001); bacterial phagocytosis (adjusted means [SE], 1,027 [48] vs. 1,509 [76] RFU/cell; P < 0.0001); and expression of granulocyte-macrophage colony-stimulating factor receptor ß subunit (adjusted means [SE], 1,471 [42] vs. 2,114 [35] RFU/cell; P < 0.0001]. Treating alveolar macrophages with zinc acetate and glutathione in vitro increased intracellular zinc levels and improved their phagocytic function. CONCLUSIONS: These novel clinical findings provide evidence that alcohol abuse is associated with significant zinc deficiency and immune dysfunction within the alveolar space and suggest that dietary supplementation with zinc and glutathione precursors could enhance airway innate immunity and decrease the risk for pneumonia or lung injury in these vulnerable individuals.


Assuntos
Alcoolismo/complicações , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Doenças do Sistema Imunitário/induzido quimicamente , Macrófagos Alveolares/metabolismo , Zinco/deficiência , Adolescente , Adulto , Alcoolismo/imunologia , Alcoolismo/metabolismo , Líquido da Lavagem Broncoalveolar/imunologia , Broncoscopia/métodos , Etanol/efeitos adversos , Etanol/imunologia , Etanol/metabolismo , Feminino , Humanos , Doenças do Sistema Imunitário/imunologia , Doenças do Sistema Imunitário/metabolismo , Imunidade Inata/efeitos dos fármacos , Imunidade Inata/imunologia , Macrófagos Alveolares/imunologia , Masculino , Pessoa de Meia-Idade , Fagocitose/efeitos dos fármacos , Fagocitose/imunologia , Adulto Jovem , Zinco/imunologia , Zinco/metabolismo
5.
Alcohol Clin Exp Res (Hoboken) ; 48(5): 810-826, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38499395

RESUMO

BACKGROUND: People with alcohol use disorder (AUD) have an increased risk of developing pneumonia and pulmonary diseases. Alveolar macrophages (AMs) are immune cells of the lower respiratory tract that are necessary for clearance of pathogens. However, alcohol causes AM oxidative stress, mitochondrial damage and dysfunction, and diminished phagocytic capacity, leading to lung injury and immune suppression. METHODS: AMs were isolated by bronchoalveolar lavage from people with AUD and male and female C57BL/6J mice given chronic ethanol (20% w/v, 12 weeks) in drinking water. The peroxisome proliferator-activated receptor γ ligand, pioglitazone, was used to treat human AMs ex vivo (10 µM, 24 h) and mice in vivo by oral gavage (10 mg/kg/day). Levels of AM mitochondrial superoxide and hypoxia-inducible factor-1 alpha (HIF-1α) mRNA, a marker of oxidative stress, were measured by fluorescence microscopy and RT-qPCR, respectively. Mouse AM phagocytic ability was determined by internalized Staphylococcus aureus, and mitochondrial capacity, dependency, and flexibility for glucose, long-chain fatty acid, and glutamine oxidation were measured using an extracellular flux analyzer. In vitro studies used a murine AM cell line, MH-S (±0.08% ethanol, 72 h) to investigate mitochondrial fuel oxidation and ATP-linked respiration. RESULTS: Pioglitazone treatment decreased mitochondrial superoxide in AMs from people with AUD and ethanol-fed mice and HIF-1α mRNA in ethanol-fed mouse lungs. Pioglitazone also reversed mouse AM glutamine oxidation and glucose or long-chain fatty acid flexibility to meet basal oxidation needs. In vitro, ethanol decreased the rate of AM mitochondrial and total ATP production, and pioglitazone improved changes in glucose and glutamine oxidation. CONCLUSIONS: Pioglitazone reversed chronic alcohol-induced oxidative stress in human AM and mitochondrial substrate oxidation flexibility and superoxide levels in mouse AM. Decreased ethanol-induced AM HIF-1α mRNA with pioglitazone suggests that this pathway may be a focus for metabolic-targeted therapeutics to improve morbidity and mortality in people with AUD.

6.
Clin Chest Med ; 44(3): 489-499, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37517829

RESUMO

Climate change will alter environmental risks that influence pulmonary health, including heat, air pollution, and pollen. These exposures disproportionately burden populations already at risk of ill health, including those at vulnerable life stages, with low socioeconomic status, and systematically targeted by oppressive policies. Climate change can exacerbate existing environmental injustices by affecting future exposure, as well as through differentials in the ability to adapt; this is compounded by disparities in rates of underlying disease and access to health care. Climate change is therefore a dire threat not only to individual and population health but also to health equity.


Assuntos
Poluição do Ar , Mudança Climática , Humanos , Poluição do Ar/efeitos adversos
7.
Sci Rep ; 13(1): 1760, 2023 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-36720997

RESUMO

Cohesive sediment forms flocs of various sizes and structures in the natural turbulent environment. Understanding flocculation is critical in accurately predicting sediment transport and biogeochemical cycles. In addition to aggregation and breakup, turbulence also reshapes flocs toward more stable structures. An Eulerian-Lagrangian framework has been implemented to investigate the effect of turbulence on flocculation by capturing the time-evolution of individual flocs. We have identified two floc reshaping mechanisms, namely breakage-regrowth and restructuring by hydrodynamic drag. Surface erosion is found to be the primary breakup mechanism for strong flocs, while fragile flocs tend to split into fragments of similar sizes. Aggregation of flocs of sizes comparable to or greater than the Kolmogorov scale is modulated by turbulence with lower aggregation efficiency. Our findings highlight the limiting effects of turbulence on both floc size and structure.

8.
Alcohol Clin Exp Res ; 35(8): 1519-28, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21447000

RESUMO

BACKGROUND: Chronic alcohol abuse causes oxidative stress, impairs alveolar macrophage immune function, and increases the risk of pneumonia and acute lung injury. Recently we determined that chronic alcohol ingestion in rats decreases zinc levels and macrophage function in the alveolar space; provocative findings in that zinc is essential for normal immune and antioxidant defenses. Alveolar macrophage immune function depends on stimulation by granulocyte/monocyte colony-stimulating factor, which signals via the transcription factor PU.1. In parallel, the antioxidant response element signals via the transcription factor Nrf2. However, the role of zinc bioavailability on these signaling pathways within the alveolar space is unknown. METHODS: To determine the efficacy of dietary zinc supplementation on lung bacterial clearance and oxidative stress, we tested 3 different groups of rats: control-fed, alcohol-fed, and alcohol-fed with zinc supplementation. Rats were then inoculated with intratracheal Klebsiella pneumoniae, and lung bacterial clearance was determined 24 hours later. Isolated alveolar macrophages were isolated from uninfected animals and evaluated for oxidative stress and signaling through PU.1 and Nrf2. RESULTS: Alcohol-fed rats had a 5-fold decrease in lung bacterial clearance compared to control-fed rats. Dietary zinc supplementation of alcohol-fed rats normalized bacterial clearance and mitigated oxidative stress in the alveolar space, as reflected by the relative balance of the thiol redox pair cysteine and cystine, and increased nuclear binding of both PU.1 and Nrf2 in alveolar macrophages from alcohol-fed rats. CONCLUSIONS: Dietary zinc supplementation prevents alcohol-induced alveolar macrophage immune dysfunction and oxidative stress in a relevant experimental model, suggesting that such a strategy could decrease the risk of pneumonia and lung injury in individuals with alcohol use disorders.


Assuntos
Macrófagos Alveolares , Fator 2 Relacionado a NF-E2 , Proteínas Proto-Oncogênicas , Oligoelementos , Transativadores , Zinco , Animais , Masculino , Ratos , Alcoolismo/metabolismo , Alcoolismo/fisiopatologia , Modelos Animais de Doenças , Etanol , Fator Estimulador de Colônias de Granulócitos e Macrófagos/efeitos dos fármacos , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Infecções por Klebsiella/metabolismo , Infecções por Klebsiella/microbiologia , Klebsiella pneumoniae/efeitos dos fármacos , Klebsiella pneumoniae/crescimento & desenvolvimento , Pulmão/imunologia , Pulmão/fisiopatologia , Lesão Pulmonar/tratamento farmacológico , Lesão Pulmonar/metabolismo , Macrófagos Alveolares/efeitos dos fármacos , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/metabolismo , Oxirredução/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Proteínas Proto-Oncogênicas/metabolismo , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Fatores de Tempo , Oligoelementos/farmacologia , Oligoelementos/uso terapêutico , Transativadores/metabolismo , Zinco/farmacologia , Zinco/uso terapêutico , Fator 2 Relacionado a NF-E2/metabolismo
9.
Alcohol Res ; 38(2): 243-254, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28988576

RESUMO

Among the many organ systems affected by harmful alcohol use, the lungs are particularly susceptible to infections and injury. The mechanisms responsible for rendering people with alcohol use disorder (AUD) vulnerable to lung damage include alterations in host defenses of the upper and lower airways, disruption of alveolar epithelial barrier integrity, and alveolar macrophage immune dysfunction. Collectively, these derangements encompass what has been termed the "alcoholic lung" phenotype. Alcohol-related reductions in antioxidant levels also may contribute to lung disease in people with underlying AUD. In addition, researchers have identified several regulatory molecules that may play crucial roles in the alcohol-induced disease processes. Although there currently are no approved therapies to combat the detrimental effects of chronic alcohol consumption on the respiratory system, these molecules may be potential therapeutic targets to guide future investigation.


Assuntos
Transtornos Relacionados ao Uso de Álcool/complicações , Pneumopatias/etiologia , Animais , Humanos , Pneumopatias/tratamento farmacológico , Pneumopatias/metabolismo
10.
OA Alcohol ; 1(2)2013 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-37886715

RESUMO

Alcohol use and abuse are prevailing practices in people throughout the world. Unfortunately, alcohol use disorders pose tremendous costs to both society and the individual. While alcoholism has many well-known medical consequences such as liver injury and pancreatitis, the effects of chronic alcohol exposure on the respiratory system are often overlooked. Specifically, studies have shown that alcohol abuse causes significant derangements in the lung and predisposes individuals to the development of pneumonia and acute lung injury. Several important processes are responsible for this increased susceptibility to pulmonary pathology, including alterations in nonimmunological defense systems, impairment of lung immunity, and alveolar epithelial barrier dysfunction. These crucial defects comprise what has been referred to as the "alcohol lung phenotype". Importantly, these abnormalities not only increase the risk of lung infections and injury, they cause worse morbidity and mortality in alcoholics compared to non-alcoholics. While there are no current therapies to combat these alcohol-induced pulmonary abnormalities, current research has revealed several important mechanisms that may be exploited to develop new treatment options for this vulnerable population.

11.
Am J Med Sci ; 343(3): 244-7, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22173040

RESUMO

Alcohol use, and misuse, has been a part of human culture for thousands of years. In the modern medical era, a great deal of attention has been justifiably focused on elucidating the mechanisms underlying the psychological and biological addiction to alcohol. However, a significant percentage, if not the majority, of alcohol-related morbidity and mortality occurs in individuals who do not meet the formal diagnostic criteria for alcohol use disorders. For example, many serious medical consequences of chronic alcohol ingestion can occur in individuals who do not have signs or symptoms of alcohol dependence. There is now clear evidence that even in otherwise healthy-appearing individuals who chronically consume excessive amounts of alcohol, alveolar macrophage immune capacity is impaired and, as a consequence, these individuals are at significantly increased risk of pneumonia. This brief review summarizes some of the key mechanisms underlying this phenomenon and proposes a hypothetical scheme by which alcohol interferes with zinc bioavailability within the alveolar space and thereby dampens macrophage function.


Assuntos
Alcoolismo/complicações , Macrófagos Alveolares/efeitos dos fármacos , Pneumonia/etiologia , Alcoolismo/epidemiologia , Animais , Fator Estimulador de Colônias de Granulócitos e Macrófagos/fisiologia , Humanos , Macrófagos Alveolares/imunologia , Camundongos , Transdução de Sinais , Zinco/deficiência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA