Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Am J Respir Crit Care Med ; 184(2): 233-42, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21562127

ABSTRACT

RATIONALE: Cigarette smoke is the major cause of chronic obstructive pulmonary disease and lung cancer. The mechanisms by which smoking induces pulmonary dysfunction are complex, involving stress from toxic components and inflammatory responses. Although CCCAAT/enhancer-binding protein (C/EBP)-ß is known as a key intracellular regulator of inflammatory signaling, its role in pulmonary inflammation has not been established. OBJECTIVES: To characterize the role of C/EBPß in the airway epithelial response to cigarette smoke. METHODS: mRNA expression in the airway epithelium of current, former, and never-smokers, and in in vitro cigarette smoke extract-treated primary human airway epithelial cells, was analyzed by microarray and quantitative real-time polymerase chain reaction, respectively. Mice with lung epithelial-specific inactivation of C/EBPß were generated and exposed to cigarette smoke for 4 or 11 days. Lung histology, bronchoalveolar lavage cell differentials, and expression of inflammatory and innate immune mediators in the lungs were assessed. MEASUREMENTS AND MAIN RESULTS: C/EBPß was significantly down-regulated in the airway epithelium of both current and former smokers compared with never-smokers, and in cigarette smoke-treated primary human airway epithelial cells in vitro. Cigarette smoke-exposed mice with a lung epithelial-specific inactivation of C/EBPß displayed blunted respiratory neutrophil influx and compromised induction of neutrophil chemoattractants growth-regulated oncogene-α, macrophage inflammatory protein-1γ, granulocyte colony-stimulating factor, and serum amyloid A 3 and proinflammatory cytokines tumor necrosis factor-α and interleukin-1ß, compared with smoke-exposed controls. Inhibition of C/EBPß in human airway cells in vitro caused a similarly compromised response to smoke. CONCLUSION: Our data suggest a previously unknown role for C/EBPß and the airway epithelium in mediating inflammatory and innate immune responses to cigarette smoke.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/immunology , Epithelial Cells/immunology , Inflammation/immunology , Lung/immunology , Smoking/immunology , Animals , Blotting, Western , Bronchoalveolar Lavage Fluid/immunology , Cell Culture Techniques , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Epithelial Cells/drug effects , Humans , Immunity, Innate/immunology , Lung/drug effects , Mice , Mice, Inbred C57BL , Reverse Transcriptase Polymerase Chain Reaction
2.
J Immunol ; 184(4): 2001-13, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-20065113

ABSTRACT

Influenza virus infection is a leading cause of death and disability throughout the world. Influenza-infected hosts are vulnerable to secondary bacterial infection, however, and an ensuing bacterial pneumonia is actually the predominant cause of influenza-attributed deaths during pandemics. A number of mechanisms have been proposed by which influenza may predispose to superinfection with an unrelated or heterologous pathogen, but the subsequent interaction between the host, virus, and bacteria remains an understudied area. In this study, we develop and examine a novel model of heterologous pulmonary infection in which an otherwise subclinical Bordetella parapertussis infection synergizes with an influenza virus infection to yield a life-threatening secondary pneumonia. Despite a profound pulmonary inflammatory response and unaltered viral clearance, bacterial clearance was significantly impaired in heterologously infected mice. No deficits were observed in pulmonary or systemic adaptive immune responses or the viability or function of infiltrating inflammatory cells to explain this phenomenon, and we provide evidence that the onset of severe pulmonary inflammation actually precedes the increased bacterial burden, suggesting that exacerbated inflammation is independent of bacterial burden. To that end, neutralization of the ELR(+) inflammatory chemokine MIP-2 (CXCL2/GRO-beta) attenuated the inflammation, weight loss, and clinical presentation of heterologously infected mice without impacting bacterial burden. These data suggest that pulmonary inflammation, rather than pathogen burden, is the key threat during bacterial superinfection of influenza and that selective chemokine antagonists may be a novel therapeutic intervention in cases of bacterial superinfection of influenza.


Subject(s)
Bordetella Infections/immunology , Bordetella parapertussis/immunology , Chemokine CXCL2/biosynthesis , Influenza A Virus, H1N1 Subtype/immunology , Orthomyxoviridae Infections/immunology , Superinfection/immunology , Animals , Bordetella Infections/pathology , Bordetella Infections/therapy , Cell Line , Chemokine CXCL2/antagonists & inhibitors , Chemokine CXCL2/physiology , Dogs , Female , Gene Expression Regulation, Bacterial/immunology , Gene Expression Regulation, Viral/immunology , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Inflammation Mediators/physiology , Mice , Mice, Inbred C57BL , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/therapy , Pneumonia, Bacterial/immunology , Pneumonia, Bacterial/pathology , Pneumonia, Bacterial/virology , Pneumonia, Viral/immunology , Pneumonia, Viral/microbiology , Pneumonia, Viral/pathology , Receptors, Interleukin-8B/antagonists & inhibitors , Superinfection/microbiology , Superinfection/virology
3.
Am J Respir Cell Mol Biol ; 42(4): 394-403, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19502389

ABSTRACT

The objective of this study was to characterize the impact of cigarette smoke exposure on lung immune and inflammatory processes. BALB/c and C57BL/6 mice were exposed to cigarette smoke for 4 days (acute) or at least 5 weeks (prolonged). Both mouse strains manifested an inflammatory response after acute smoke exposure, characterized by an influx of neutrophils and mononuclear cells. Multiplex analysis revealed a greater than twofold increase of the cytokines IL-1alpha, -5, -6, and -18, as well as the chemokines monocyte chemotactic protein-1 and -3, macrophage inflammatory protein-1alpha, -beta, and -gamma, -2, -3beta, macrophage defined chemokine, granulocyte chemotactic protein-2, and interferon-gamma-inducible protein-10. In BALB/c mice, neutrophilia persisted after prolonged exposure, whereas C57BL/6 showed evidence of attenuated neutrophilia both in the bronchoalveolar lavage and the lungs. In both mouse strains, cigarette smoke exposure was associated with an expansion of mature (CD11c(hi)/major histocompatibility complex class II(hi)) myeloid dendritic cells; we observed no changes in plasmacytoid dendritic cells. Lymphocytes in the lungs displayed an activated phenotype that persisted for CD4 T cells only after prolonged exposure. In BALB/c mice, T cells acquired T helper (Th) 1 and Th2 effector function after 5 weeks of smoke exposure, whereas, in C57BL/6 mice, neither Th1 nor Th2 cells were detected. In both mouse strains, cigarette smoke exposure led to an accumulation of FoxP3+ T regulatory cells in the lungs. Studies in RAG1 knockout mice suggest that these regulatory cells may participate in controlling smoke-induced inflammation. Acute and prolonged cigarette smoke exposure was associated with inflammation, activation of the adaptive immune system, and expansion of T regulatory cells in the lungs.


Subject(s)
Immunity, Innate/immunology , Pneumonia/immunology , Smoking/adverse effects , Adaptive Immunity/genetics , Adaptive Immunity/immunology , Animals , Bronchoalveolar Lavage , CD11c Antigen/genetics , CD11c Antigen/immunology , Cytokines/genetics , Cytokines/immunology , Dendritic Cells/immunology , Dendritic Cells/physiology , Female , Homeodomain Proteins/genetics , Homeodomain Proteins/immunology , Humans , Immunity, Innate/genetics , Lung/immunology , Lung/pathology , Mice , Mice, Inbred BALB C , Mice, Knockout , Myeloid Cells/immunology , Myeloid Cells/pathology , Neutrophil Infiltration/genetics , Neutrophil Infiltration/immunology , Neutrophils/immunology , Neutrophils/pathology , Pneumonia/etiology , Pneumonia/genetics , Pneumonia/pathology , Smoking/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Th1 Cells/immunology , Th1 Cells/pathology , Th2 Cells/immunology , Th2 Cells/pathology , Time Factors
4.
Am J Respir Crit Care Med ; 179(8): 666-75, 2009 Apr 15.
Article in English | MEDLINE | ID: mdl-19179487

ABSTRACT

RATIONALE: The pathogenesis of chronic obstructive pulmonary disease is associated with acute episodes of bacterial exacerbations. The most commonly isolated bacteria during episodes of exacerbation is nontypeable Haemophilus influenzae (NTHI). OBJECTIVES: In this study, we investigated the in vivo consequences of cigarette smoke exposure on the inflammatory response to an NTHI challenge. METHODS: C57BL/6 and BALB/c mice were exposed to cigarette smoke for 8 weeks and subsequently challenged intranasally with NTHI. MEASUREMENTS AND MAIN RESULTS: We observed increased pulmonary inflammation and lung damage in cigarette smoke-exposed NTHI-challenged mice as compared with control NTHI-challenged mice. Furthermore, although NTHI challenge in control mice was marked by increases in tumor necrosis factor-alpha, IL-6, MIP-2, and KC/GROalpha, NTHI challenge in cigarette smoke-exposed mice led to a prominent up-regulation of a different subset of inflammatory mediators, most notably MCP-1, -3, and -5, IP-10, and MIP-1gamma. This skewed inflammatory mediator expression was also observed after ex vivo NTHI stimulation of alveolar macrophages, signifying their importance to this altered response. Importantly, corticosteroids attenuated inflammation after NTHI challenge in both cigarette smoke-exposed and control mice; however, this was associated with significantly increased bacterial burden. CONCLUSIONS: Collectively, these data suggest that cigarette smoke exacerbates the inflammatory response to a bacterial challenge via skewed inflammatory mediator expression.


Subject(s)
Haemophilus influenzae/immunology , Pulmonary Disease, Chronic Obstructive/immunology , Pulmonary Disease, Chronic Obstructive/microbiology , Tobacco Smoke Pollution/adverse effects , Administration, Intranasal , Animals , Disease Progression , Female , Inflammation/etiology , Inflammation/microbiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
5.
Cell Immunol ; 253(1-2): 38-44, 2008.
Article in English | MEDLINE | ID: mdl-18533139

ABSTRACT

Although its direct effects cannot be discounted, tobacco's effects on the immune system have been proposed to play a key role in mediating its deleterious health impact. Studies in rats using high levels of smoke exposure have suggested that tobacco smoke exhausts cellular signal transduction cascades, making lymphocytes unresponsive to stimulation. In the present study, we show that purified B or T cells, and total lymphocytes from the lungs, lymph nodes and spleens of smoke-exposed mice fluxed calcium, proliferated, and secreted immunoglobulin or IFN-gamma similarly to control mice when stimulated with ligands including anti-IgM, and anti-CD3. Importantly, we recapitulated these findings in PBMCs from human smokers; cells from long-term smokers and never-smokers proliferated equivalently when stimulated ex vivo. Previous reports of lymphocyte unresponsiveness in rats are inconsistent with these findings, and may reflect a phenomenon observed only at levels of smoke exposure well above those seen in actual human smokers.


Subject(s)
B-Lymphocytes/drug effects , Lymphocyte Activation/drug effects , Lymphocyte Subsets/drug effects , Nicotiana/adverse effects , Smoke/adverse effects , T-Lymphocytes/drug effects , Animals , B-Lymphocytes/immunology , Calcium/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Female , Humans , Lung/cytology , Lung/immunology , Lymph Nodes/cytology , Lymph Nodes/immunology , Lymphocyte Activation/immunology , Lymphocyte Subsets/immunology , Mice , Mice, Inbred C57BL , Rats , Smoking/immunology , Spleen/cytology , Spleen/immunology , T-Lymphocytes/immunology , Nicotiana/immunology
6.
Am J Respir Cell Mol Biol ; 38(2): 218-26, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17872497

ABSTRACT

Alveolar macrophages (aMs) play a central role in respiratory host defense by sensing microbial antigens and initiating immune-inflammatory responses early in the course of an infection. The purpose of this study was to investigate the effect of cigarette smoke exposure on aMs after stimulation of innate pattern recognition receptors (PRRs) in a murine model. To accomplish this, C57BL/6 mice were exposed for 8 weeks using two models of cigarette smoke exposure, nose-only or whole-body exposure, and aMs isolated from the bronchoalveolar lavage. After stimulation of aMs with pI:C, a mimic of viral replication, and bacterial cell-wall constituent LPS, aMs from cigarette smoke-exposed mice produced significantly attenuated levels of the inflammatory cytokines TNF-alpha and IL-6, and the chemokine RANTES. This attenuation was specific to the aM compartment, and not related to changes in aM viability or expression of Toll-like receptor (TLR)3 or TLR4 between groups. Furthermore, aMs from smoke-exposed mice had decreased cytokine RNA as compared with aMs from sham-exposed mice. Mechanistically, this was associated with decreased nuclear translocation of the proinflammatory transcription factor NF-kappaB, and increased activator protein-1 nuclear translocation, in aMs from smoke-exposed mice. Attenuated cytokine production was reversible after smoking cessation. Cigarette smoke exposure also attenuated TNF-alpha production after stimulation with nucleotide-oligomerization domain-like receptor agonists, showing that the effect applies more broadly to other PRR pathways. Our data demonstrate that cigarette smoke exposure attenuates aM responses after innate stimulation, including pathways typically associated with bacterial and viral infections.


Subject(s)
Cytokines/biosynthesis , Macrophages, Alveolar/metabolism , Nicotiana , Smoke/adverse effects , Animals , Carboxyhemoglobin/metabolism , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Fluorescent Antibody Technique , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred C57BL , Nitric Oxide/metabolism , Pulmonary Disease, Chronic Obstructive/etiology , Reverse Transcriptase Polymerase Chain Reaction , Toll-Like Receptors/metabolism
7.
Contrib Microbiol ; 14: 126-141, 2007.
Article in English | MEDLINE | ID: mdl-17684337

ABSTRACT

Modeling acute exacerbations of chronic obstructive pulmonary disease (AECOPD) in animals has proven challenging due to the clinical and pathological complexity of the underlying disease. This has hindered the progress in understanding the cellular and molecular mechanisms that lie beneath AECOPD. In this chapter, we will address modeling possibilities of AECOPD that may be drawn from the current knowledge of factors that cause exacerbations. Importantly, since it is widely accepted that the most common causes of AECOPD are viral and bacterial infections, animal models of AECOPD should incorporate both the causative agents of exacerbation: viruses and bacteria. However, other factors that are also believed to determine both progression of COPD, as well as the frequency and severity of AECOPD, such as proteolytic enzymes, cigarette smoke or other noxious stimuli must also be considered. Such animal models will provide mechanistic insight into the etiology of AECOPD and will prove invaluable in furthering our understanding of key events in disease pathogenesis.


Subject(s)
Disease Models, Animal , Pulmonary Disease, Chronic Obstructive/etiology , Animals , Humans , Pulmonary Disease, Chronic Obstructive/immunology , Pulmonary Disease, Chronic Obstructive/microbiology , Pulmonary Disease, Chronic Obstructive/virology
8.
Am J Respir Crit Care Med ; 174(12): 1342-51, 2006 Dec 15.
Article in English | MEDLINE | ID: mdl-17023734

ABSTRACT

RATIONALE: Studies have shown that cigarette smoke impacts respiratory host defense mechanisms; however, it is poorly understood how these smoke-induced changes impact the overall ability of the host to deal with pathogenic agents. OBJECTIVE: The objective of this study was to investigate the impact of mainstream cigarette smoke exposure on immune inflammatory responses and viral burden after respiratory infection with influenza A. METHODS: C57BL/6 mice were sham- or smoke-exposed for 3 to 5 mo and infected with either 2.5 x 10(3) pfu (low dose) or 2.5 x 10(5) pfu (high dose) influenza virus. MEASUREMENTS AND MAIN RESULTS: Although smoke exposure attenuated the airway's inflammatory response to low-dose infection, we observed increased inflammation in smoke-exposed compared with sham-exposed mice after infection with high-dose influenza, despite a similar rate of viral clearance. The heightened inflammatory response was associated with increased expression of tumor necrosis factor-alpha, interleukin-6, and type 1 IFN in the airway, and increased mortality. Importantly, smoke exposure did not interfere with the development of influenza-specific memory responses; sham- and smoke-exposed animals were equally protected upon viral rechallenge. CONCLUSION: Our study suggests that, in mice, cigarette smoke affects primary antiviral immune-inflammatory responses, whereas secondary immune protection remains intact.


Subject(s)
Inflammation/immunology , Influenza A virus , Orthomyxoviridae Infections/immunology , Smoking/adverse effects , Animals , Female , Flow Cytometry , Interleukin-6/analysis , Mice , Mice, Inbred C57BL , Peroxidase/analysis , Tumor Necrosis Factor-alpha/analysis , Viral Load
SELECTION OF CITATIONS
SEARCH DETAIL
...