Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
1.
Respir Res ; 14: 33, 2013 Mar 09.
Article in English | MEDLINE | ID: mdl-23497334

ABSTRACT

BACKGROUND: Cigarette smoking is associated with increased frequency and duration of viral respiratory infections, but the underlying mechanisms are incompletely defined. We investigated whether smoking reduces expression by human lung macrophages (Mø) of receptors for viral nucleic acids and, if so, the effect on CXCL10 production. METHODS: We collected alveolar macrophages (AMø) by bronchoalveolar lavage of radiographically-normal lungs of subjects undergoing bronchoscopies for solitary nodules (n = 16) and of volunteers who were current or former smokers (n = 7) or never-smokers (n = 13). We measured expression of mRNA transcripts for viral nucleic acid receptors by real-time PCR in those AMø and in the human Mø cell line THP-1 following phorbol myristate acetate/vitamin D3 differentiation and exposure to cigarette smoke extract, and determined TLR3 protein expression using flow cytometry and immunohistochemistry. We also used flow cytometry to examine TLR3 expression in total lung Mø from subjects undergoing clinically-indicated lung resections (n = 25). Of these, seven had normal FEV1 and FEV1/FVC ratio (three former smokers, four current smokers); the remaining 18 subjects (14 former smokers; four current smokers) had COPD of GOLD stages I-IV. We measured AMø production of CXCL10 in response to stimulation with the dsRNA analogue poly(I:C) using Luminex assay. RESULTS: Relative to AMø of never-smokers, AMø of smokers demonstrated reduced protein expression of TLR3 and decreased mRNA for TLR3 but not TLR7, TLR8, TLR9, RIG-I, MDA-5 or PKR. Identical changes in TLR3 gene expression were induced in differentiated THP-1 cells exposed to cigarette smoke-extract in vitro for 4 hours. Among total lung Mø, the percentage of TLR3-positive cells correlated inversely with active smoking but not with COPD diagnosis, FEV1% predicted, sex, age or pack-years. Compared to AMø of never-smokers, poly(I:C)-stimulated production of CXCL10 was significantly reduced in AMø of smokers. CONCLUSIONS: Active smoking, independent of COPD stage or smoking duration, reduces both the percent of human lung Mø expressing TLR3, and dsRNA-induced CXCL10 production, without altering other endosomal or cytoplasmic receptors for microbial nucleic acids. This effect provides one possible mechanism for increased frequency and duration of viral lower respiratory tract infections in smokers. TRIAL REGISTRATION: ClinicalTrials.gov NCT00281190, NCT00281203 and NCT00281229.


Subject(s)
Down-Regulation/genetics , Macrophages, Alveolar/metabolism , RNA, Double-Stranded/antagonists & inhibitors , Smoking/metabolism , Toll-Like Receptor 3/antagonists & inhibitors , Adult , Aged , Cell Line , Cells, Cultured , Cohort Studies , Female , Humans , Lung/cytology , Lung/metabolism , Lung/virology , Macrophages, Alveolar/virology , Male , Middle Aged , RNA, Double-Stranded/genetics , Smoking/genetics , Toll-Like Receptor 3/biosynthesis , Toll-Like Receptor 3/genetics , Young Adult
2.
J Immunol ; 189(1): 112-9, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22615206

ABSTRACT

The lung environment actively inhibits apoptotic cell (AC) uptake by alveolar macrophages (AMøs) via lung collectin signaling through signal regulatory protein α (SIRPα). Even brief glucocorticoid (GC) treatment during maturation of human blood monocyte-derived or murine bone marrow-derived macrophages (Møs) increases their AC uptake. Whether GCs similarly impact differentiated tissue Møs and the mechanisms for this rapid response are unknown and important to define, given the widespread therapeutic use of inhaled GCs. We found that the GC fluticasone rapidly and dose-dependently increased AC uptake by murine AMøs without a requirement for protein synthesis. Fluticasone rapidly suppressed AMø expression of SIRPα mRNA and surface protein, and also activated a more delayed, translation-dependent upregulation of AC recognition receptors that was not required for the early increase in AC uptake. Consistent with a role for SIRPα suppression in rapid GC action, murine peritoneal Møs that had not been exposed to lung collectins showed delayed, but not rapid, increase in AC uptake. However, pretreatment of peritoneal Møs with the lung collectin surfactant protein D inhibited AC uptake, and fluticasone treatment rapidly reversed this inhibition. Thus, GCs act not only by upregulating AC recognition receptors during Mø maturation but also via a novel rapid downregulation of SIRPα expression by differentiated tissue Møs. Release of AMøs from inhibition of AC uptake by lung collectins may, in part, explain the beneficial role of inhaled GCs in inflammatory lung diseases, especially emphysema, in which there is both increased lung parenchymal cell apoptosis and defective AC uptake by AMøs.


Subject(s)
Androstadienes/pharmacology , Apoptosis Regulatory Proteins/pharmacology , Apoptosis/immunology , Collectins/physiology , Down-Regulation/immunology , Immune Tolerance/immunology , Macrophages, Alveolar/immunology , Receptors, Immunologic/antagonists & inhibitors , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/physiology , Cells, Cultured , Dose-Response Relationship, Immunologic , Down-Regulation/drug effects , Fluticasone , Immune Tolerance/drug effects , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/pathology , Mice , Mice, Inbred C57BL , Receptors, Immunologic/physiology , Up-Regulation/drug effects , Up-Regulation/immunology
3.
J Immunol ; 184(11): 6504-13, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20427767

ABSTRACT

Lung CD8(+) T cells might contribute to progression of chronic obstructive pulmonary disease (COPD) indirectly via IFN-gamma production or directly via cytolysis, but evidence for either mechanism is largely circumstantial. To gain insights into these potential mechanisms, we analyzed clinically indicated lung resections from three human cohorts, correlating findings with spirometrically defined disease severity. Expression by lung CD8(+) T cells of IL-18R and CD69 correlated with severity, as did mRNA transcripts for perforin and granzyme B, but not Fas ligand. These correlations persisted after correction for age, smoking history, presence of lung cancer, recent respiratory infection, or inhaled corticosteroid use. Analysis of transcripts for killer cell lectin-like receptor G1, IL-7R, and CD57 implied that lung CD8(+) T cells in COPD do not belong to the terminally differentiated effector populations associated with chronic infections or extreme age. In vitro stimulation of lung CD8(+) T cells with IL-18 plus IL-12 markedly increased production of IFN-gamma and TNF-alpha, whereas IL-15 stimulation induced increased intracellular perforin expression. Both IL-15 and IL-18 protein expression could be measured in whole lung tissue homogenates, but neither correlated in concentration with spirometric severity. Although lung CD8(+) T cell expression of mRNA for both T-box transcription factor expressed in T cells and GATA-binding protein 3 (but not retinoic acid receptor-related orphan receptor gamma or alpha) increased with spirometric severity, stimulation of lung CD8(+) T cells via CD3epsilon-induced secretion of IFN-gamma, TNF-alpha, and GM-CSF, but not IL-5, IL-13, and IL-17A. These findings suggest that the production of proinflammatory cytokines and cytotoxic molecules by lung-resident CD8(+) T cells contributes to COPD pathogenesis.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Interleukin-15/immunology , Interleukin-18/immunology , Pulmonary Disease, Chronic Obstructive/immunology , T-Lymphocyte Subsets/immunology , Aged , Antigens, CD/biosynthesis , Antigens, Differentiation, T-Lymphocyte/biosynthesis , CD8-Positive T-Lymphocytes/metabolism , Cell Separation , Cytokines/biosynthesis , Cytokines/immunology , Cytotoxicity, Immunologic , Female , Flow Cytometry , Forced Expiratory Volume , Humans , Lectins, C-Type/biosynthesis , Male , Middle Aged , Pulmonary Disease, Chronic Obstructive/physiopathology , RNA, Messenger/analysis , Respiratory Function Tests , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocyte Subsets/metabolism
4.
Am J Respir Crit Care Med ; 180(12): 1179-88, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-19729666

ABSTRACT

RATIONALE: Dendritic cells (DCs) have not been well studied in chronic obstructive pulmonary disease (COPD), yet their integral role in activating and differentiating T cells makes them potential participants in COPD pathogenesis. OBJECTIVES: To determine the expression of maturation molecules by individual DC subsets in relationship to COPD stage and to expression of the acute activation marker CD69 by lung CD4(+) T cells. METHODS: We nonenzymatically released lung leukocytes from human surgical specimens (n = 42) and used flow cytometry to identify three DC subsets (mDC1, mDC2, and pDC) and to measure their expression of three costimulatory molecules (CD40, CD80 and CD86) and of CD83, the definitive marker of DC maturation. Spearman nonparametric correlation analysis was used to identify significant correlations between expression of DC maturation molecules and COPD severity. MEASUREMENTS AND MAIN RESULTS: Expression of CD40 by mDC1 and mDC2 and of CD86 by mDC2 was high regardless of GOLD stage, but CD80 and CD83 on these two DC subsets increased with disease progression. pDC also showed significant increases in expression of CD40 and CD80. Expression of all but one of the DC molecules that increased with COPD severity also correlated with CD69 expression on lung CD4(+) T cells from the same patients, with the exception of CD83 on mDC2. CONCLUSIONS: This cross-sectional study implies that COPD progression is associated with significant increases in costimulatory molecule expression by multiple lung DC subsets. Interactions with lung DCs may contribute to the immunophenotype of CD4(+) T cells in advanced COPD. Clinical trial registered with www.clinicaltrials.gov (NCT00281229).


Subject(s)
Dendritic Cells/immunology , Lung/immunology , Pulmonary Disease, Chronic Obstructive/immunology , Aged , Antigens, CD/immunology , Antigens, Differentiation, T-Lymphocyte/immunology , CD4-Positive T-Lymphocytes/immunology , Cell Culture Techniques , Cell Differentiation/immunology , Cross-Sectional Studies , Female , Flow Cytometry , Humans , Lectins, C-Type , Male , Middle Aged , Severity of Illness Index , Smoking/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...