Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Cell Rep ; 32(8): 108062, 2020 08 25.
Article in English | MEDLINE | ID: mdl-32846120

ABSTRACT

Pneumonias caused by influenza A virus (IAV) co- and secondary bacterial infections are characterized by their severity and high mortality rate. Previously, we have shown that bacterial pore-forming toxin (PFT)-mediated necroptosis is a key driver of acute lung injury during bacterial pneumonia. Here, we evaluate the impact of IAV on PFT-induced acute lung injury during co- and secondary Streptococcus pneumoniae (Spn) infection. We observe that IAV synergistically sensitizes lung epithelial cells for PFT-mediated necroptosis in vitro and in murine models of Spn co-infection and secondary infection. Pharmacoelogical induction of oxidative stress without virus sensitizes cells for PFT-mediated necroptosis. Antioxidant treatment or inhibition of necroptosis reduces disease severity during secondary bacterial infection. Our results advance our understanding on the molecular basis of co- and secondary bacterial infection to influenza and identify necroptosis inhibition and antioxidant therapy as potential intervention strategies.


Subject(s)
Influenza, Human/complications , Lung/microbiology , Necroptosis/genetics , Oxidative Stress/genetics , Animals , Humans , Mice
2.
J Biol Chem ; 295(6): 1704-1715, 2020 02 07.
Article in English | MEDLINE | ID: mdl-31882535

ABSTRACT

The influenza A (H1N1)pdm09 outbreak in 2009 exemplified the problems accompanying the emergence of novel influenza A virus (IAV) strains and their unanticipated virulence in populations with no pre-existing immunity. Neuraminidase inhibitors (NAIs) are currently the drugs of choice for intervention against IAV outbreaks, but there are concerns that NAI-resistant viruses can transmit to high-risk populations. These issues highlight the need for new approaches that address the annual influenza burden. In this study, we examined whether palmitoyl-oleoyl-phosphatidylglycerol (POPG) and phosphatidylinositol (PI) effectively antagonize (H1N1)pdm09 infection. POPG and PI markedly suppressed cytopathic effects and attenuated viral gene expression in (H1N1)pdm09-infected Madin-Darby canine kidney cells. POPG and PI bound to (H1N1)pdm09 with high affinity and disrupted viral spread from infected to noninfected cells in tissue culture and also reduced (H1N1)pdm09 propagation by a factor of 102 after viral infection was established in vitro In a mouse infection model of (H1N1)pdm09, POPG and PI significantly reduced lung inflammation and viral burden. Of note, when mice were challenged with a typically lethal dose of 1000 plaque-forming units of (H1N1)pdm09, survival after 10 days was 100% (14 of 14 mice) with the POPG treatment compared with 0% (0 of 14 mice) without this treatment. POPG also significantly reduced inflammatory infiltrates and the viral burden induced by (H1N1)pdm09 infection in a ferret model. These findings indicate that anionic phospholipids potently and efficiently disrupt influenza infections in animal models.


Subject(s)
Antiviral Agents/therapeutic use , Influenza A Virus, H1N1 Subtype/drug effects , Orthomyxoviridae Infections/drug therapy , Phosphatidylglycerols/therapeutic use , Phosphatidylinositols/therapeutic use , Animals , Antiviral Agents/pharmacology , Disease Models, Animal , Dogs , Female , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/pathology , Phosphatidylglycerols/pharmacology , Phosphatidylinositols/pharmacology , Pulmonary Surfactants/pharmacology , Pulmonary Surfactants/therapeutic use
3.
Am J Respir Cell Mol Biol ; 61(6): 702-712, 2019 12.
Article in English | MEDLINE | ID: mdl-31144984

ABSTRACT

Obesity is a risk factor for asthma, especially nonatopic asthma, and attenuates the efficacy of standard asthma therapeutics. Obesity also augments pulmonary responses to ozone, a nonatopic asthma trigger. The purpose of this study was to determine whether obesity-related alterations in gut microbiota contribute to these augmented responses to ozone. Ozone-induced increases in airway responsiveness, a canonical feature of asthma, were greater in obese db/db mice than in lean wild-type control mice. Depletion of gut microbiota with a cocktail of antibiotics attenuated obesity-related increases in the response to ozone, indicating a role for microbiota. Moreover, ozone-induced airway hyperresponsiveness was greater in germ-free mice that had been reconstituted with colonic contents of db/db than in wild-type mice. In addition, compared with dietary supplementation with the nonfermentable fiber cellulose, dietary supplementation with the fermentable fiber pectin attenuated obesity-related increases in the pulmonary response to ozone, likely by reducing ozone-induced release of IL-17A. Our data indicate a role for microbiota in obesity-related increases in the response to an asthma trigger and suggest that microbiome-based therapies such as prebiotics may provide an alternative therapeutic strategy for obese patients with asthma.


Subject(s)
Gastrointestinal Microbiome/physiology , Obesity/complications , Ozone/toxicity , Respiratory Hypersensitivity/etiology , Airway Resistance , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Asthma/etiology , Asthma/therapy , Cellulose/administration & dosage , Dietary Fiber/administration & dosage , Fecal Microbiota Transplantation , Female , Fermentation , Gastrointestinal Microbiome/drug effects , Germ-Free Life , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/genetics , Obesity/microbiology , Obesity/physiopathology , Pectins/administration & dosage , Pectins/therapeutic use , Receptors, Leptin/deficiency , Respiratory Hypersensitivity/chemically induced , Respiratory Hypersensitivity/diet therapy , Respiratory Hypersensitivity/microbiology
4.
JCI Insight ; 3(20)2018 10 18.
Article in English | MEDLINE | ID: mdl-30333319

ABSTRACT

Severe influenza (IAV) infection can develop into bronchopneumonia and edema, leading to acquired respiratory distress syndrome (ARDS) and pathophysiology. Underlying causes for pulmonary edema and aberrant fluid regulation largely remain unknown, particularly regarding the role of viral-mediated mechanisms. Herein, we show that distinct IAV strains reduced the functions of the epithelial sodium channel (ENaC) and the cystic fibrosis transmembrane regulator (CFTR) in murine respiratory and alveolar epithelia in vivo, as assessed by measurements of nasal potential differences and single-cell electrophysiology. Reduced ion channel activity was distinctly limited to virally infected cells in vivo and not bystander uninfected lung epithelium. Multiple lines of evidence indicated ENaC and CFTR dysfunction during the acute infection period; however, only CFTR dysfunction persisted beyond the infection period. ENaC, CFTR, and Na,K-ATPase activities and protein levels were also reduced in virally infected human airway epithelial cells. Reduced ENaC and CFTR led to changes in airway surface liquid morphology of human tracheobronchial cultures and airways of IAV-infected mice. Pharmacologic correction of CFTR function ameliorated IAV-induced physiologic changes. These changes are consistent with mucous stasis and pulmonary edema; furthermore, they indicate that repurposing therapeutic interventions correcting CFTR dysfunction may be efficacious for treatment of IAV lung pathophysiology.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Epithelial Sodium Channels/metabolism , Influenza A virus/pathogenicity , Influenza, Human/pathology , Respiratory Mucosa/pathology , Aminopyridines/pharmacology , Animals , Benzodioxoles/pharmacology , Bronchi/metabolism , Bronchi/pathology , Bronchi/virology , Cells, Cultured , Disease Models, Animal , Dogs , Epithelial Cells , Female , Humans , Influenza, Human/complications , Influenza, Human/virology , Madin Darby Canine Kidney Cells , Male , Mice , Primary Cell Culture , Pulmonary Edema/pathology , Pulmonary Edema/virology , Respiratory Distress Syndrome/pathology , Respiratory Distress Syndrome/virology , Respiratory Mucosa/metabolism , Respiratory Mucosa/virology , Sodium-Potassium-Exchanging ATPase/metabolism , Water-Electrolyte Balance/drug effects
5.
Am J Respir Cell Mol Biol ; 59(3): 346-354, 2018 09.
Article in English | MEDLINE | ID: mdl-29529379

ABSTRACT

Previous reports demonstrate that the microbiome impacts allergic airway responses, including airway hyperresponsiveness, a characteristic feature of asthma. Here we examined the role of the microbiome in pulmonary responses to a nonallergic asthma trigger, ozone. We depleted the microbiota of conventional mice with either a single antibiotic (ampicillin, metronidazole, neomycin, or vancomycin) or a cocktail of all four antibiotics given via the drinking water. Mice were then exposed to room air or ozone. In air-exposed mice, airway responsiveness did not differ between antibiotic- and control water-treated mice. Ozone caused airway hyperresponsiveness, the magnitude of which was decreased in antibiotic cocktail-treated mice versus water-treated mice. Except for neomycin, single antibiotics had effects similar to those observed with the cocktail. Compared with conventional mice, germ-free mice also had attenuated airway responsiveness after ozone. 16S ribosomal RNA gene sequencing of fecal DNA to characterize the gut microbiome indicated that bacterial genera that were decreased in mice with reduced ozone-induced airway hyperresponsiveness after antibiotic treatment were short-chain fatty acid producers. Serum analysis indicated reduced concentrations of the short-chain fatty acid propionate in cocktail-treated mice but not in neomycin-treated mice. Dietary enrichment with pectin, which increased serum short-chain fatty acids, also augmented ozone-induced airway hyperresponsiveness. Furthermore, propionate supplementation of the drinking water augmented ozone-induced airway hyperresponsiveness in conventional mice. Our data indicate that the microbiome contributes to ozone-induced airway hyperresponsiveness, likely via its ability to produce short-chain fatty acids.


Subject(s)
Anti-Bacterial Agents/pharmacology , Microbiota/drug effects , Neutrophils/drug effects , Ozone/adverse effects , Animals , Bronchoalveolar Lavage Fluid/cytology , Mice , Microbiota/physiology , Respiratory Hypersensitivity/chemically induced , Respiratory Hypersensitivity/drug therapy , Tumor Necrosis Factor-alpha/antagonists & inhibitors
6.
Am J Respir Cell Mol Biol ; 58(3): 341-351, 2018 03.
Article in English | MEDLINE | ID: mdl-28957638

ABSTRACT

Ozone and obesity both increase IL-17A in the lungs. In mice, obesity augments the airway hyperresponsiveness and neutrophil recruitment induced by acute ozone exposure. Therefore, we examined the role of IL-17A in obesity-related increases in the response to ozone observed in obese mice. Lean wild-type and obese db/db mice were pretreated with IL-17A-blocking or isotype antibodies, exposed to air or ozone (2 ppm for 3 h), and evaluated 24 hours later. Microarray analysis of lung tissue gene expression was used to examine the mechanistic basis for effects of anti-IL-17A. Compared with lean mice, ozone-exposed obese mice had greater concentrations of BAL IL-17A and greater numbers of pulmonary IL-17A+ cells. Ozone-induced increases in BAL IL-23 and CCL20, cytokines important for IL-17A+ cell recruitment and activation, were also greater in obese mice. Anti-IL-17A treatment reduced ozone-induced airway hyperresponsiveness toward levels observed in lean mice. Anti-IL-17A treatment also reduced BAL neutrophils in both lean and obese mice, possibly because of reductions in CXCL1. Microarray analysis identified gastrin-releasing peptide (GRP) receptor (Grpr) among those genes that were both elevated in the lungs of obese mice after ozone exposure and reduced after anti-IL-17A treatment. Furthermore, ozone exposure increased BAL GRP to a greater extent in obese than in lean mice, and GRP-neutralizing antibody treatment reduced obesity-related increases in ozone-induced airway hyperresponsiveness and neutrophil recruitment. Our data indicate that IL-17A contributes to augmented responses to ozone in db/db mice. Furthermore, IL-17A appears to act at least in part by inducing expression of Grpr.


Subject(s)
Gastrin-Releasing Peptide/immunology , Interleukin-17/immunology , Obesity/pathology , Ozone/toxicity , Receptors, Bombesin/metabolism , Respiratory Hypersensitivity/immunology , Animals , Antibodies, Blocking/pharmacology , Chemokine CCL20/immunology , Chemokine CXCL1/immunology , Female , Interleukin-23 Subunit p19/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration/immunology , Neutrophils/immunology , Receptors, Bombesin/genetics
7.
J Immunotoxicol ; 13(3): 428-38, 2016 05.
Article in English | MEDLINE | ID: mdl-27043160

ABSTRACT

Exposure to subacute ozone (O3) causes pulmonary neutrophil recruitment. In mice, this recruitment requires IL-17A. Ozone also causes expression of IL-23 and IL-1, which can induce IL-17A. The purpose of this study was to examine the hypothesis that IL-23 and IL-1 contribute to IL-17A expression and subsequent neutrophil recruitment after subacute O3 exposure. Wild-type, IL-23(-/-), and Flt3l(-/-) mice were exposed to air or 0.3 ppm O3 for 72 h. Flt3l(-/-) mice lack conventional dendritic cells (cDC) that can express IL-23 and IL-1. Other wild-type mice were pre-treated with saline or the IL-1R1 antagonist anakinra prior to O3 exposure. After exposure, bronchoalveolar lavage (BAL) was performed and lung tissue harvested. The results indicated that pulmonary Il17a mRNA abundance and IL-17A(+) F4/80(+) cells were significantly reduced in O3-exposed IL-23(-/-) vs in wild-type mice. In contrast, anakinra had no effect on Il23a or Il17a pulmonary mRNA abundance or on BAL concentrations of the neutrophil survival factor G-CSF, but anakinra did reduce BAL neutrophil numbers, likely because anakinra also reduced BAL IL-6. Compared to air, O3 caused a significant increase in DC numbers in wild-type, but not in Flt3(-/-) mice. However, there was no significant difference in Il23a or Il17a mRNA abundance or in BAL neutrophil count in O3-exposed Flt3(-/-) vs in wild-type mice. From these results, it was concluded that IL-23 but not IL-1 contributes to the IL-17A expression induced by subacute O3 exposure. Induction of IL-23 by O3 does not appear to require cDC.


Subject(s)
Dendritic Cells/immunology , Interleukin-17/metabolism , Interleukin-23/metabolism , Lung/immunology , Ozone/immunology , Administration, Inhalation , Animals , Antigens, Differentiation/metabolism , Cells, Cultured , Gene Expression Regulation , Humans , Interleukin-1/metabolism , Interleukin-17/genetics , Interleukin-23/genetics , Interleukin-6/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration , Ozone/toxicity , fms-Like Tyrosine Kinase 3/genetics
8.
PLoS One ; 9(5): e97707, 2014.
Article in English | MEDLINE | ID: mdl-24823369

ABSTRACT

Ozone is an air pollutant that causes pulmonary symptoms. In mice, ozone exposure causes pulmonary injury and increases bronchoalveolar lavage macrophages and neutrophils. We have shown that IL-17A is important in the recruitment of neutrophils after subacute ozone exposure (0.3 ppm for 24-72 h). We hypothesized that γδ T cells are the main producers of IL-17A after subacute ozone. To explore this hypothesis we exposed wildtype mice and mice deficient in γδ T cells (TCRδ-/-) to ozone or room air. Ozone-induced increases in BAL macrophages and neutrophils were attenuated in TCRδ-/- mice. Ozone increased the number of γδ T cells in the lungs and increased pulmonary Il17a mRNA expression and the number of IL-17A+ CD45+ cells in the lungs and these effects were abolished in TCRδ-/- mice. Ozone-induced increases in factors downstream of IL-17A signaling, including G-CSF, IL-6, IP-10 and KC were also decreased in TCRδ-/- versus wildtype mice. Neutralization of IL-17A during ozone exposure in wildtype mice mimicked the effects of γδ T cell deficiency. TNFR2 deficiency and etanercept, a TNFα antagonist, also reduced ozone-induced increases in Il17a mRNA, IL-17A+ CD45+ cells and BAL G-CSF as well as BAL neutrophils. TNFR2 deficient mice also had decreased ozone-induced increases in Ccl20, a chemoattractant for IL-17A+ γδ T cells. Il17a mRNA and IL-17A+ γδ T cells were also lower in obese Cpefat versus lean WT mice exposed to subacute ozone, consistent with the reduced neutrophil recruitment observed in the obese mice. Taken together, our data indicate that pulmonary inflammation induced by subacute ozone requires γδ T cells and TNFα-dependent recruitment of IL-17A+ γδ T cells to the lung.


Subject(s)
Interleukin-17/metabolism , Lung/drug effects , Ozone/toxicity , Pneumonia/chemically induced , Pneumonia/immunology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , T-Lymphocytes/immunology , Analysis of Variance , Animals , Bronchoalveolar Lavage , DNA Primers/genetics , Etanercept , Flow Cytometry , Immunoglobulin G , Lung/immunology , Macrophages/immunology , Mice , Mice, Knockout , Neutrophils/immunology , Real-Time Polymerase Chain Reaction , Receptors, Antigen, T-Cell, gamma-delta/genetics , Receptors, Tumor Necrosis Factor , Receptors, Tumor Necrosis Factor, Type II , T-Lymphocytes/metabolism
9.
Am J Physiol Lung Cell Mol Physiol ; 303(12): L1079-86, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23087018

ABSTRACT

Pulmonary dendritic cells (DCs) are among the first responders to inhaled environmental stimuli such as ozone (O(3)), which has been shown to activate these cells. O(3) reacts with epithelial lining fluid (ELF) components in an anatomically site-specific manner dictated by O(3) concentration, airway flow patterns, and ELF substrate concentration. Accordingly, the anatomical distribution of ELF reaction products and airway injury are hypothesized to produce selective DC maturation differentially within the airways. To investigate how O(3) affects regional airway DC populations, we utilized a model of O(3)-induced pulmonary inflammation, wherein C57BL/6 mice were exposed to 0.8 ppm O(3) 8 h/day for 1, 3, and 5 days. This model induced mild inflammation and no remarkable epithelial injury. Tracheal, but not more distant airway sites, and mediastinal lymph node (MLN) DC numbers were increased significantly after the third exposure day. The largest increase in each tissue was of the CD103(+) DC phenotype. After 3 days of exposure, fewer DCs expressed CD80, CD40, and CCR7, and, at this same time point, total MLN T cell numbers increased. Together, these data demonstrate that O(3) exposure induced site-specific and phenotype changes in the pulmonary and regional lymph node DC populations. Possibly contributing to ozone-mediated asthma perturbation, the phenotypic changes to DCs within pulmonary regions may alter responses to antigenic stimuli. Decreased costimulatory molecule expression within the MLN suggests induction of tolerance mechanisms; increased tracheal DC number may raise the potential for allergic sensitization and asthmatic exacerbation, thus overcoming O(3)-induced decrements in costimulatory molecule expression.


Subject(s)
Antigens, CD/metabolism , CD11b Antigen/metabolism , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Environmental Exposure , Integrin alpha Chains/metabolism , Oxidants, Photochemical/pharmacology , Ozone/toxicity , Animals , Antigens, CD/analysis , B7-1 Antigen/analysis , CD11b Antigen/analysis , CD40 Antigens/analysis , Integrin alpha Chains/analysis , Lung/drug effects , Lymph Nodes/drug effects , Lymphocyte Count , Male , Mice , Mice, Inbred C57BL , Pneumonia/chemically induced , Receptors, CCR7/analysis , Trachea/drug effects
10.
J Biol Chem ; 285(21): 16239-47, 2010 May 21.
Article in English | MEDLINE | ID: mdl-20228065

ABSTRACT

Transforming growth factor beta (TGF-beta) stimulates reactive oxygen species (ROS) production in various cell types, which mediates many of the effects of TGF-beta. The molecular mechanisms whereby TGF-beta increases ROS production and ROS modulate the signaling processes of TGF-beta, however, remain poorly defined. In this study, we show that TGF-beta1 stimulates NADPH oxidase 4 (Nox4) expression and ROS generation in the nucleus of murine embryo fibroblasts (NIH3T3 cells). This is associated with an increase in protein thiol modification and inactivation of MAPK phosphatase 1 (MKP-1), a nuclear phosphatase. Furthermore, knockdown of MKP-1 using small interfering RNA enhances TGF-beta1-induced phosphorylation of JNK and p38 as well as the expression of plasminogen activator inhibitor 1 (PAI-1), a TGF-beta-responsive gene involved in the pathogenesis of many diseases. Knockdown of Nox4 with Nox4 small interfering RNA, on the other hand, reduces TGF-beta1-stimulated ROS production, p38 phosphorylation, and PAI-1 expression. TGF-beta also increased the nuclear level of Nox4 protein as well as PAI-1 expression in human lung fibroblasts (CCL-210 cells), suggesting that TGF-beta may induce PAI-1 expression by a similar mechanism in human lung fibroblasts. In summary, in this study we have identified nuclear MAPK phosphatase MKP-1 as a novel molecular target of ROS in TGF-beta signaling pathways. Our data suggest that increased generation of ROS by Nox4 mediates TGF-beta1-induced PAI-1 gene expression at least in part through oxidative modification and inhibition of MKP-1 leading to a sustained activation of JNK and p38 MAPKs.


Subject(s)
Dual Specificity Phosphatase 1/metabolism , Fibroblasts/metabolism , Gene Expression Regulation/physiology , MAP Kinase Signaling System/physiology , Nuclear Proteins/metabolism , Plasminogen Activator Inhibitor 1/biosynthesis , Transforming Growth Factor beta1/metabolism , Animals , Dual Specificity Phosphatase 1/genetics , Fibroblasts/cytology , Gene Expression Regulation/drug effects , Humans , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/metabolism , MAP Kinase Signaling System/drug effects , Mice , NADPH Oxidase 4 , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , NIH 3T3 Cells , Nuclear Proteins/genetics , Oxidation-Reduction/drug effects , Phosphorylation/drug effects , Phosphorylation/physiology , Plasminogen Activator Inhibitor 1/genetics , RNA, Small Interfering/genetics , Reactive Oxygen Species/metabolism , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/pharmacology , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...