Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 10 de 10
1.
Physiol Rep ; 8(19): e14584, 2020 10.
Article En | MEDLINE | ID: mdl-33052618

In mice, both androgens and the gut microbiota modify pulmonary responses to ozone. We hypothesized that androgens affect gut microbiota and thereby impact pulmonary responses to ozone. To address this hypothesis, we transferred cecal microbiota from male castrated or sham castrated C57BL/6J mice into female germ-free recipient C57BL/6J mice. Four weeks later mice were exposed to ozone (2 ppm) or room air for 3 hr. The gut microbiomes of castrated versus sham castrated donors differed, as did those of recipients of microbiota from castrated versus sham castrated donors. In recipients, ozone-induced airway hyperresponsiveness was not affected by donor castration status. However, compared to mice receiving microbiota from sham castrated donors, mice receiving microbiota from castrated donors had elevated numbers of bronchoalveolar lavage (BAL) neutrophils despite evidence of reduced lung injury as measured by BAL protein. Serum concentrations of IL-17A and G-CSF were significantly greater in recipients of castrated versus sham castrated microbiota. Furthermore, BAL neutrophils correlated with both serum IL-17A and serum G-CSF. Our data indicate that androgen-mediated effects on the gut microbiota modulate pulmonary inflammatory responses to ozone and suggest a role for circulating IL-17A and G-CSF in these events.


Androgens/pharmacology , Bronchoalveolar Lavage Fluid/microbiology , Gastrointestinal Microbiome/drug effects , Ozone/adverse effects , Animals , Interleukin-17/metabolism , Lung/drug effects , Lung/metabolism , Mice, Inbred C57BL , Neutrophils/drug effects , Neutrophils/metabolism , Respiratory Hypersensitivity/physiopathology
2.
Am J Respir Cell Mol Biol ; 62(4): 503-512, 2020 04.
Article En | MEDLINE | ID: mdl-31913653

Ozone causes airway hyperresponsiveness, a defining feature of asthma. We have reported that the gut microbiome contributes to sex differences in ozone-induced airway hyperresponsiveness. Altering dietary fiber affects the gut microbiome. The purpose of this study was to determine the effects of dietary fiber on pulmonary responses to ozone and whether these effects differ by sex. We fed male and female mice fiber-free diets or diets enriched in one of two types of dietary fiber, cellulose and pectin, for 3 days before ozone exposure. Compared with control diets or pectin-enriched diets, cellulose-enriched diets attenuated ozone-induced airway hyperresponsiveness in male but not female mice. In contrast, fiber-free diets augmented responses to ozone in female but not male mice. Analysis of 16S rRNA sequencing of fecal DNA also indicated sex differences in the impact of dietary fiber on the gut microbiome and identified bacterial taxa that were associated with ozone-induced airway hyperresponsiveness. Our data suggest that microbiome-based therapies such as prebiotics may provide an alternative therapeutic strategy for air pollution-triggered asthma, but they indicate that such therapeutics may need to be tailored differently for males and females.


Dietary Fiber/metabolism , Lung/drug effects , Ozone/pharmacology , Animals , Asthma/metabolism , Diet/methods , Female , Gastrointestinal Microbiome/drug effects , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , RNA, Ribosomal, 16S/metabolism , Respiratory Hypersensitivity/metabolism , Sex Characteristics
3.
Physiol Rep ; 7(18): e14214, 2019 09.
Article En | MEDLINE | ID: mdl-31544355

Ozone causes airway hyperresponsiveness, a defining feature of asthma, and is an asthma trigger. In mice, ozone-induced airway hyperresponsiveness is greater in males than in females, suggesting a role for sex hormones in the response to ozone. To examine the role of androgens in these sex differences, we castrated 4-week-old mice. Controls underwent sham surgery. At 8 weeks of age, mice were exposed to ozone (2ppm, 3 h) or room air. Twenty-four hours later, mice were anesthetized and measurements of airway responsiveness to inhaled aerosolized methacholine were made. Mice were then euthanized and bronchoalveolar lavage was performed. Castration attenuated ozone-induced airway hyperresponsiveness and reduced bronchoalveolar lavage cells. In intact males, flutamide, an androgen receptor inhibitor, had similar effects to castration. Bronchoalveolar lavage concentrations of several cytokines were reduced by either castration or flutamide treatment, but only IL-1α was reduced by both castration and flutamide. Furthermore, an anti-IL-1α antibody reduced bronchoalveolar lavage neutrophils in intact males, although it did not alter ozone-induced airway hyperresponsiveness. Our data indicate that androgens augment pulmonary responses to ozone and that IL-1α may contribute to the effects of androgens on ozone-induced cellular inflammation but not airway hyperresponsiveness.


Androgens/physiology , Lung/drug effects , Ozone/toxicity , Respiratory Hypersensitivity/chemically induced , Androgen Antagonists/therapeutic use , Androgens/deficiency , Animals , Bronchoalveolar Lavage Fluid/chemistry , Corticosterone/blood , Cytokines/metabolism , Flutamide/therapeutic use , Interleukin-1alpha/metabolism , Interleukin-6/blood , Male , Methacholine Chloride , Mice, Inbred C57BL , Neutrophil Infiltration/drug effects , Neutrophil Infiltration/physiology , Orchiectomy , Oxidative Stress/physiology , Pneumonia/chemically induced , Pneumonia/physiopathology , Pneumonia/prevention & control , Respiratory Hypersensitivity/physiopathology , Respiratory Hypersensitivity/prevention & control , Respiratory Mechanics/drug effects , Sex Characteristics
4.
PLoS One ; 14(8): e0221633, 2019.
Article En | MEDLINE | ID: mdl-31454377

Ozone is an asthma trigger. In mice, the gut microbiome contributes to ozone-induced airway hyperresponsiveness, a defining feature of asthma, but the mechanistic basis for the role of the gut microbiome has not been established. Gut bacteria can affect the function of distal organs by generating metabolites that enter the blood and circulate systemically. We hypothesized that global metabolomic profiling of serum collected from ozone exposed mice could be used to identify metabolites contributing to the role of the microbiome in ozone-induced airway hyperresponsiveness. Mice were treated for two weeks with a cocktail of antibiotics (ampicillin, neomycin, metronidazole, and vancomycin) in the drinking water or with control water and then exposed to air or ozone (2 ppm for 3 hours). Twenty four hours later, blood was harvested and serum analyzed via liquid-chromatography or gas-chromatography coupled to mass spectrometry. Antibiotic treatment significantly affected 228 of the 562 biochemicals identified, including reductions in the known bacterially-derived metabolites, equol, indole propionate, 3-indoxyl sulfate, and 3-(4-hydroxyphenyl)propionate, confirming the efficacy of the antibiotic treatment. Ozone exposure caused significant changes in 334 metabolites. Importantly, ozone-induced changes in many of these metabolites were different in control and antibiotic-treated mice. For example, most medium and long chain fatty acids declined by 20-50% with ozone exposure in antibiotic-treated but not control mice. Most taurine-conjugated bile acids increased with ozone exposure in antibiotic-treated but not control mice. Ozone also caused marked (9-fold and 5-fold) increases in the polyamines, spermine and spermidine, respectively, in control but not antibiotic-treated mice. Each of these metabolites has the capacity to alter airway responsiveness and may account for the role of the microbiome in pulmonary responses to ozone.


Metabolome , Microbiota , Ozone/adverse effects , Serum/metabolism , Air , Amino Acids/blood , Animals , Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Bacteria/metabolism , Bile Acids and Salts/biosynthesis , Corticosterone/blood , Glutathione/blood , Hormones/metabolism , Lipids/blood , Liver/metabolism , Metabolic Networks and Pathways , Metabolome/drug effects , Mice, Inbred C57BL , Microbiota/drug effects , Polyamines/blood , Principal Component Analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Thyroxine/blood
5.
Toxicol Sci ; 169(1): 180-193, 2019 05 01.
Article En | MEDLINE | ID: mdl-30690640

Low molecular weight polycyclic aromatic hydrocarbons (LMW PAHs; < 206.3 g/mol) are under regulated environmental contaminants (eg, secondhand smoke) that lead to gap junction dysregulation, p38 MAPK activation, and increased mRNA production of inflammatory mediators, such as cytokines and cyclooxygenase (COX2), in lung epithelial cells. However, the early mechanisms involving lipid signaling through the arachidonic acid pathway and subsequent eicosanoid production leading to these downstream events are not known. Common human exposures are to mixtures of LMW PAHs, thus C10 cells (a mouse lung epithelial cell line) were exposed to a representative binary PAH mixture, 1-methylanthracene (1-MeA) and fluoranthene (Flthn), for 30 min-24 h with and without p38 and cytosolic phospholipase A2 (cPLA2) inhibitors. Cytosolic phospholipase A2 inhibition reversed PAH-induced phospho-p38 MAPK activation and gap junction dysregulation at 30 min. A significant biphasic increase in cPLA2 protein was observed at 30 min, 2, and 4 h, as well as COX2 protein at 2 and 8 h. Untargeted metabolomics demonstrated a similar trend with significantly changing metabolites at 30 min and 4 h of exposure relative to 1 h; a "cPLA2-like" subset of metabolites within the biphasic response were predominately phospholipids. Targeted metabolomics showed several eicosanoids (eg, prostaglandin D2 (PGD2), PGE2α) were significantly increased at 4, 8, and 12 h following exposure to the binary PAH mixture and this effect was p38-dependent. Finally, PAH metabolism was not observed until after 8 h. These results indicate an early lipid signaling mechanism of LMW PAH toxicity in lung epithelial cells due to parent PAH compounds.


Alveolar Epithelial Cells/drug effects , Anthracenes/toxicity , Eicosanoids/metabolism , Fluorenes/toxicity , Signal Transduction/drug effects , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/pathology , Animals , Anthracenes/chemistry , Cell Line , Cyclooxygenase 2/metabolism , Enzyme Activation , Fluorenes/chemistry , Group IV Phospholipases A2/metabolism , Metabolomics , Mice, Inbred BALB C , Molecular Weight , Phosphorylation , Time Factors , Up-Regulation , p38 Mitogen-Activated Protein Kinases/metabolism
6.
Am J Respir Cell Mol Biol ; 60(2): 198-208, 2019 02.
Article En | MEDLINE | ID: mdl-30240285

We have previously reported that the mouse gut microbiome contributes to pulmonary responses to ozone, a common asthma trigger, and that short-chain fatty acids, end products of bacterial fermentation, likely contribute to this role of the microbiome. A growing body of evidence indicates that there are sex-related differences in gut microbiota and these differences can have important functional consequences. The purpose of this study was to determine whether there are sex-related differences in the impact of the gut microbiota on pulmonary responses to ozone. After acute exposure to ozone, male mice developed greater airway hyperresponsiveness than female mice. This difference was abolished after antibiotic ablation of the gut microbiome. Moreover, weanling female pups housed in cages conditioned by adult male mice developed greater ozone-induced airway hyperresponsiveness than weanling female pups raised in cages conditioned by adult females. Finally, ad libitum oral administration via drinking water of the short-chain fatty acid propionate resulted in augmented ozone-induced airway hyperresponsiveness in male, but not female, mice. Overall, these data are consistent with the hypothesis that the microbiome contributes to sex differences in ozone-induced airway hyperresponsiveness, likely as a result of sex differences in the response to short-chain fatty acids.


Lung/drug effects , Microbiota/drug effects , Microbiota/physiology , Ozone/adverse effects , Respiratory Hypersensitivity/microbiology , Animals , Anti-Bacterial Agents/pharmacology , Bronchoalveolar Lavage Fluid/microbiology , Fatty Acids, Volatile/metabolism , Female , Lung/metabolism , Male , Mice, Inbred C57BL , Propionates/pharmacology , Respiratory Hypersensitivity/chemically induced , Respiratory Hypersensitivity/drug therapy , Sex Factors
7.
Toxicol Sci ; 157(1): 156-171, 2017 05 01.
Article En | MEDLINE | ID: mdl-28329830

Low molecular weight polycyclic aromatic hydrocarbons (LMW PAHs; < 206.3 g/mol) are prevalent and ubiquitous environmental contaminants, presenting a human health concern, and have not been as thoroughly studied as the high MW PAHs. LMW PAHs exert their pulmonary effects, in part, through P38-dependent and -independent mechanisms involving cell-cell communication and the production of pro-inflammatory mediators known to contribute to lung disease. Specifically, we determined the effects of two representative LMW PAHs, 1-methylanthracene (1-MeA) and fluoranthene (Flthn), individually and as a binary PAH mixture on the dysregulation of gap junctional intercellular communication (GJIC) and connexin 43 (Cx43), activation of mitogen activated protein kinases (MAPK), and induction of inflammatory mediators in a mouse non-tumorigenic alveolar type II cell line (C10). Both 1-MeA, Flthn, and the binary PAH mixture of 1-MeA and Flthn dysregulated GJIC in a dose and time-dependent manner, reduced Cx43 protein, and activated the following MAPKs: P38, ERK1/2, and JNK. Inhibition of P38 MAPK prevented PAH-induced dysregulation of GJIC, whereas inhibiting ERK and JNK did not prevent these PAHs from dysregulating GJIC indicating a P38-dependent mechanism. A toxicogenomic approach revealed significant P38-dependent and -independent pathways involved in inflammation, steroid synthesis, metabolism, and oxidative responses. Genes in these pathways were significantly altered by the binary PAH mixture when compared with 1-MeA and Flthn alone suggesting interactive effects. Exposure to the binary PAH mixture induced the production and release of cytokines and metalloproteinases from the C10 cells. Our findings with a binary mixture of PAHs suggest that combinations of LMW PAHs may elicit synergistic or additive inflammatory responses which warrant further investigation and confirmation.


Inflammation/metabolism , Lung/drug effects , Mitogens/metabolism , Polycyclic Aromatic Hydrocarbons/toxicity , Signal Transduction/drug effects , Tobacco Smoke Pollution , Animals , Cell Communication/drug effects , Cell Line , Connexin 43/metabolism , Dose-Response Relationship, Drug , Enzyme Activation , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Gap Junctions/drug effects , Lung/metabolism , Lung/pathology , Mice , Mitogen-Activated Protein Kinases/metabolism , Transcriptome
8.
Exp Lung Res ; 42(3): 154-73, 2016 Apr.
Article En | MEDLINE | ID: mdl-27093379

Tumor promotion is an early and critical stage during lung adenocarcinoma (ADC). We previously demonstrated that Tlr4 mutant mice were more susceptible to butylated hydroxytoluene (BHT)-induced pulmonary inflammation and tumor promotion in comparison to Tlr4-sufficient mice. Our study objective was to elucidate the underlying differences in Tlr4 mutant mice in innate immune cell populations, their functional responses, and the influence of these cellular differences on ADC progenitor (type II) cells following BHT-treatment. BALB (Tlr4-sufficient) and C.C3-Tlr4(Lps-d)/J (BALB(Lpsd); Tlr4 mutant) mice were treated with BHT (promoter) followed by bronchoalveolar lavage (BAL) and flow cytometry processing on the lungs. ELISAs, Club cell enrichment, macrophage function, and RNA isolation were also performed. Bone marrow-derived macrophages (BMDM) co-cultured with a type II cell line were used for wound healing assays. Innate immune cells significantly increased in whole lung in BHT-treated BALB(Lpsd) mice compared to BALB mice. BHT-treated BALB(Lpsd) mice demonstrated enhanced macrophage functionality, increased epithelial wound closure via BMDMs, and increased Club cell number in BALB(Lpsd) mice, all compared to BALB BHT-treated mice. Cytokine/chemokine (Kc, Mcp1) and growth factor (Igf1) levels also significantly differed among the strains and within macrophages, gene expression, and cell surface markers collectively demonstrated a more plastic phenotype in BALB(Lpsd) mice. Therefore, these correlative studies suggest that distinct innate immune cell populations are associated with the differences observed in the Tlr4-mutant model. Future studies will investigate the macrophage origins and the utility of the pathways identified herein as indicators of immune system deficiencies and lung tumorigenesis.


Carcinogenesis/genetics , Carcinogenesis/immunology , Immunity, Innate/immunology , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Toll-Like Receptor 4/immunology , Adenocarcinoma/genetics , Adenocarcinoma/immunology , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Animals , Bronchoalveolar Lavage/methods , Butylated Hydroxytoluene/pharmacology , Carcinogenesis/pathology , Chemokines/immunology , Gene Expression/genetics , Gene Expression/immunology , Immunity, Innate/genetics , Lung/immunology , Lung/pathology , Lung Neoplasms/genetics , Macrophages/immunology , Macrophages/pathology , Male , Mice , Mice, Inbred BALB C , Mutation/genetics , Mutation/immunology , Pneumonia/chemically induced , Pneumonia/genetics , Pneumonia/immunology , Pneumonia/pathology , Toll-Like Receptor 4/genetics
9.
PLoS One ; 8(6): e65150, 2014.
Article En | MEDLINE | ID: mdl-23755184

Polycyclic aromatic hydrocarbons (PAHs) are ubiquitous environmental and occupational toxicants, which are a major human health concern in the U.S. and abroad. Previous research has focused on the genotoxic events caused by high molecular weight PAHs, but not on non-genotoxic events elicited by low molecular weight PAHs. We used an isomeric pair of low molecular weight PAHs, namely 1-Methylanthracene (1-MeA) and 2-Methylanthracene (2-MeA), in which only 1-MeA possessed a bay-like region, and hypothesized that 1-MeA, but not 2-MeA, would affect non-genotoxic endpoints relevant to tumor promotion in murine C10 lung cells, a non-tumorigenic type II alveolar pneumocyte and progenitor cell type of lung adenocarcinoma. The non-genotoxic endpoints assessed were dysregulation of gap junction intercellular communication function and changes in the major pulmonary connexin protein, connexin 43, using fluorescent redistribution and immunoblots, activation of mitogen activated protein kinases (MAPK) using phosphospecific MAPK antibodies for immunoblots, and induction of inflammatory genes using quantitative RT-PCR. 2-MeA had no effect on any of the endpoints, but 1-MeA dysregulated gap junctional communication in a dose and time dependent manner, reduced connexin 43 protein expression, and altered membrane localization. 1-MeA also activated ERK1/2 and p38 MAP kinases. Inflammatory genes, such as cyclooxygenase 2, and chemokine ligand 2 (macrophage chemoattractant 2), were also upregulated in response to 1-MeA only. These results indicate a possible structure-activity relationship of these low molecular weight PAHs relevant to non-genotoxic endpoints of the promoting aspects of cancer. Therefore, our novel findings may improve the ability to predict outcomes for future studies with additional toxicants and mixtures, identify novel targets for biomarkers and chemotherapeutics, and have possible implications for future risk assessment for these PAHs.


Carcinogenesis/pathology , Inflammation/pathology , Lung/pathology , Polycyclic Aromatic Hydrocarbons/chemistry , Polycyclic Aromatic Hydrocarbons/toxicity , Signal Transduction/drug effects , Animals , Anthracenes/chemistry , Anthracenes/toxicity , Carcinogenesis/metabolism , Cell Communication/drug effects , Cell Death/drug effects , Cell Line, Tumor , Connexin 43/metabolism , Enzyme Activation/drug effects , Gap Junctions/metabolism , Humans , Mice , Mitogen-Activated Protein Kinases/metabolism , Molecular Weight , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Time Factors
10.
J Mol Biomark Diagn ; 5(1)2013 Dec 27.
Article En | MEDLINE | ID: mdl-25035812

TLR4 protects against lung tumor promotion and pulmonary inflammation in mice. Connexin 43 (Cx43), a gap junction gene, was increased in Tlr4 wildtype compared to Tlr4-mutant mice in response to promotion, which suggests gap junctional intercellular communication (GJIC) may be compromised. We hypothesized that the early tumor microenvironment, represented by Bronchoalveolar Lavage Fluid (BALF) from Butylated hydroxytoluene (BHT; promoter)-treated mice, would produce TLR4-dependent changes in pulmonary epithelium, including dysregulation of GJIC in the Tlr4-mutant (BALB Lps-d ) compared to the Tlr4-sufficient (BALB; wildtype) mice. BHT (4 weekly doses) was injected ip followed by BALF collection at 24 h. BALF total protein and total macrophages were significantly elevated in BHT-treated BALB Lps-d over BALB mice, similar to previous findings. BALF was then utilized in an ex vivo manner to treat C10 cells, a murine alveolar type II cell line, followed by the scrape-load dye transfer assay (GJIC), Cx43 immunostaining, and quantitative RT-PCR (Mcp-1, monocyte chemotactic protein 1). GJIC was markedly reduced in C10 cells treated with BHT-treated BALB Lps-d BALF for 4 and 24 h compared to BALB and control BALF from the respective mice (p < 0.05). Mcp-1, a chemokine, was also significantly increased in the BHT-treated BALB Lps-d BALF compared to the BALB mice, and Cx43 protein expression in the cell membrane altered. These novel findings suggest signaling from the BALF milieu is involved in GJIC dysregulation associated with promotion and links gap junctions to pulmonary TLR4 protection in a novel ex vivo model that could assist in future potential tumor promoter screening.

...