Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Nat Rev Cancer ; 23(4): 258-269, 2023 04.
Article in English | MEDLINE | ID: mdl-36807417

ABSTRACT

The theory that cancer-associated fibroblasts (CAFs) are immunosuppressive cells has prevailed throughout the past decade. However, recent high-throughput, high-resolution mesenchyme-directed single-cell studies have harnessed computational advances to functionally characterize cell states, highlighting the existence of immunostimulatory CAFs. Our group and others have uncovered and experimentally substantiated key functions of cancer antigen-presenting CAFs in T cell immunity, both in vitro and in vivo, refuting the conventional assumption that CAFs impede adaptive immune rejection of tumours. In this Perspective, I unify the follicular and non-follicular, non-endothelial stroma of tumours under the 'peripheral adaptive immune mesenchyme' framework and position subsets of CAFs as direct positive regulators of the adaptive immune system. Building on the understanding of cancer antigen presentation by CAFs and the second touch hypothesis, which postulates that full T cell polarization requires interaction with antigen-presenting cells in the non-lymphoid tissue where the antigen resides, I re-design the 'cancer-immunity cycle' to incorporate intratumoural activation of cancer-specific CD4+ T cells. Lastly, a road map to therapeutic harnessing of immunostimulatory CAF states is proposed.


Subject(s)
Cancer-Associated Fibroblasts , Neoplasms , Humans , Cancer-Associated Fibroblasts/pathology , Neoplasms/pathology , T-Lymphocytes , Antigen Presentation , Tumor Microenvironment
2.
J Exp Med ; 219(2)2022 02 07.
Article in English | MEDLINE | ID: mdl-35029648

ABSTRACT

A key unknown of the functional space in tumor immunity is whether CD4 T cells depend on intratumoral MHCII cancer antigen recognition. MHCII-expressing, antigen-presenting cancer-associated fibroblasts (apCAFs) have been found in breast and pancreatic tumors and are considered to be immunosuppressive. This analysis shows that antigen-presenting fibroblasts are frequent in human lung non-small cell carcinomas, where they seem to actively promote rather than suppress MHCII immunity. Lung apCAFs directly activated the TCRs of effector CD4 T cells and at the same time produced C1q, which acted on T cell C1qbp to rescue them from apoptosis. Fibroblast-specific MHCII or C1q deletion impaired CD4 T cell immunity and accelerated tumor growth, while inducing C1qbp in adoptively transferred CD4 T cells expanded their numbers and reduced tumors. Collectively, we have characterized in the lungs a subset of antigen-presenting fibroblasts with tumor-suppressive properties and propose that cancer immunotherapies might be strongly dependent on in situ MHCII antigen presentation.


Subject(s)
Antigen Presentation/immunology , Antigens, Neoplasm/immunology , Cancer-Associated Fibroblasts/immunology , Histocompatibility Antigens Class II/immunology , Lung Neoplasms/immunology , Animals , Apoptosis , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Carrier Proteins/metabolism , Disease Models, Animal , Humans , Interferon-gamma/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lymphocyte Activation , Lymphocyte Count , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Mice , Mitochondrial Proteins/metabolism , Single-Cell Analysis , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Transcriptome , Tumor Microenvironment/immunology
4.
Nat Commun ; 10(1): 1405, 2019 03 29.
Article in English | MEDLINE | ID: mdl-30926812

ABSTRACT

Lung adenocarcinoma (LUAD)-derived Wnts increase cancer cell proliferative/stemness potential, but whether they impact the immune microenvironment is unknown. Here we show that LUAD cells use paracrine Wnt1 signaling to induce immune resistance. In TCGA, Wnt1 correlates strongly with tolerogenic genes. In another LUAD cohort, Wnt1 inversely associates with T cell abundance. Altering Wnt1 expression profoundly affects growth of murine lung adenocarcinomas and this is dependent on conventional dendritic cells (cDCs) and T cells. Mechanistically, Wnt1 leads to transcriptional silencing of CC/CXC chemokines in cDCs, T cell exclusion and cross-tolerance. Wnt-target genes are up-regulated in human intratumoral cDCs and decrease upon silencing Wnt1, accompanied by enhanced T cell cytotoxicity. siWnt1-nanoparticles given as single therapy or part of combinatorial immunotherapies act at both arms of the cancer-immune ecosystem to halt tumor growth. Collectively, our studies show that Wnt1 induces immunologically cold tumors through cDCs and highlight its immunotherapeutic targeting.


Subject(s)
Adaptive Immunity , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/immunology , Chemokines/genetics , Dendritic Cells/metabolism , Gene Silencing , Wnt1 Protein/metabolism , Adoptive Transfer , Animals , Cell Line, Tumor , Cell Proliferation , Chemokines/metabolism , Humans , Immune Evasion , Mice, Inbred C57BL , RNA Interference , Signal Transduction , T-Lymphocytes/metabolism , Up-Regulation
5.
J Immunol ; 201(5): 1558-1569, 2018 09 01.
Article in English | MEDLINE | ID: mdl-30037849

ABSTRACT

Chronic obstructive pulmonary disease is a chronic inflammatory disorder with an increased incidence of lung cancer. The emphysema component of chronic obstructive pulmonary disease confers the greatest proportion to lung cancer risk. Although tumors create inflammatory conditions to escape immunity, the immunological responses that control growth of nascent cancer cells in pre-established inflammatory microenvironments are unknown. In this study, we addressed this issue by implanting OVA-expressing cancer cells in the lungs of mice with cigarette smoke-induced emphysema. Emphysema augmented the growth of cancer cells, an effect that was dependent on T cytotoxic cells. OVA-specific OTI T cells showed early signs of exhaustion upon transfer in emphysema tumor hosts that was largely irreversible because sorting, expansion, and adoptive transfer failed to restore their antitumor activity. Increased numbers of PD-L1- and IDO-positive CD11c+ myeloid dendritic cells (DCs) infiltrated emphysema tumors, whereas sorted emphysema tumor DCs poorly stimulated OTI T cells. Upon adoptive transfer in immunocompetent hosts, T cells primed by emphysema tumor DCs were unable to halt tumor growth. DCs exposed to the emphysema tumor microenvironment downregulated MHC class II and costimulatory molecules, whereas they upregulated PD-L1/IDO via oxidative stress-dependent mechanisms. T cell activation increased upon PD-L1 blockade in emphysema DC-T cell cocultures and in emphysema tumor hosts in vivo. Analysis of the transcriptome of primary human lung tumors showed a strong association between computed tomography-based emphysema scoring and downregulation of immunogenic processes. Thus, suppression of adaptive immunity against lung cancer cells links a chronic inflammatory disorder, emphysema, to cancer, with clinical implications for emphysema patients to be considered optimal candidates for cancer immunotherapies.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/transplantation , Cigarette Smoking/immunology , Lung Neoplasms/immunology , Pulmonary Emphysema/immunology , Adoptive Transfer , Animals , Cigarette Smoking/pathology , Lung Neoplasms/pathology , Mice , Mice, Transgenic , Pulmonary Emphysema/physiopathology
6.
J Allergy Clin Immunol ; 134(4): 944-954.e8, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25051954

ABSTRACT

BACKGROUND: Increased mortality rates in patients with chronic obstructive pulmonary disease (COPD) are largely due to severe infectious exacerbations. Impaired respiratory immunity is linked to the enhanced susceptibility to infections. Dendritic cells (DCs) direct host immune responses toward immunity or tolerance. Pulmonary CD1c(+) DCs elicit robust antiviral immune responses in healthy subjects. Nevertheless, their functional specialization in patients with COPD remains unexplored. OBJECTIVE: We sought to better understand the mechanisms that suppress respiratory immunity in patients with COPD by examining the immunostimulatory and tolerogenic properties of pulmonary CD1c(+) DCs. METHODS: We analyzed the expression of costimulatory and tolerogenic molecules by pulmonary CD1c(+) DCs from patients with COPD (CD1c(+)DCCOPD) and former smokers without COPD. We isolated lung CD1c(+) DCs and determined their ability to stimulate allogeneic T-cell responses. The suppressive effects of lung CD1c(+) DCs and CD1c(+) DC-primed T cells on mixed leukocyte reactions were examined. An experimental human model of COPD exacerbation was used to investigate the levels of critical immunosuppressive molecules in vivo. RESULTS: CD1c(+) DCs from patients with COPD hinder T-cell effector functions and favor the generation of suppressive IL-10-secreting CD4(+) T cells that function through IL-10 and TGF-ß. IL-27, IL-10, and inducible T-cell costimulator ligand signaling are essential for CD1c(+)DCCOPD-mediated differentiation of IL-10-producing suppressive T cells. Exposure of lung CD1c(+) DCs from nonobstructed subjects to lungs of patients with COPD confers tolerogenic properties. IL-27 and IL-10 levels are increased in the lung microenvironment on rhinovirus-induced COPD exacerbation in vivo. CONCLUSION: We identify a novel tolerogenic circuit encompassing suppressive CD1c(+) DCs and regulatory T cells in patients with COPD that might be implicated in impaired respiratory immunity and further highlight IL-10 and IL-27 as potent therapeutic targets.


Subject(s)
Dendritic Cells/immunology , Interleukin-10/metabolism , Interleukin-27/metabolism , Lung/immunology , Pulmonary Disease, Chronic Obstructive/immunology , Rhinovirus/immunology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Regulatory/immunology , Aged , Antigens, CD1/metabolism , Bystander Effect , Cell Differentiation , Cells, Cultured , Dendritic Cells/virology , Disease Progression , Female , Glycoproteins/metabolism , Humans , Immune Tolerance , Inducible T-Cell Co-Stimulator Protein/metabolism , Interleukin-10/genetics , Interleukin-27/genetics , Isoantigens/immunology , Lymphocyte Activation , Lymphocyte Culture Test, Mixed , Male , Middle Aged , Pulmonary Disease, Chronic Obstructive/complications , Pulmonary Disease, Chronic Obstructive/virology , Signal Transduction/immunology
9.
Am J Respir Crit Care Med ; 187(11): 1194-202, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23525932

ABSTRACT

RATIONALE: Little is known about what drives the appearance of lymphoid follicles (LFs), which may function as lymphoid organs in chronic obstructive pulmonary disease (COPD). In animal infection models, pulmonary LF formation requires expression of homeostatic chemokines by stromal cells and dendritic cells, partly via lymphotoxin. OBJECTIVES: To study the role of homeostatic chemokines in LF formation in COPD and to identify mechanism(s) responsible for their production. METHODS: Peripheral lung homeostatic chemokine and lymphotoxin expression were visualized by immunostainings and quantified by ELISA/quantitative reverse transcriptase-polymerase chain reaction in patients with COPD with and without LFs. Expression of lymphotoxin and homeostatic chemokine receptors was investigated by flow cytometry. Primary lung cell cultures, followed by ELISA/quantitative reverse transcriptase-polymerase chain reaction/flow cytometry, were performed to identify mechanisms of chemokine expression. Polycarbonate membrane filters were used to assess primary lung cell migration toward lung homogenates. MEASUREMENTS AND MAIN RESULTS: LFs expressed the homeostatic chemokine CXCL13. Total CXCL13 levels correlated with LF density. Lung B cells of patients with COPD were important sources of CXCL13 and lymphotoxin and also expressed their receptors. Cigarette smoke extract, H2O2, and LPS exposure up-regulated B cell-derived CXCL13. The LPS-induced increase in CXCL13 was partly mediated via lymphotoxin. Notably, CXCL13 was required for efficient lung B-cell migration toward COPD lung homogenates and induced lung B cells to up-regulate lymphotoxin, which further promoted CXCL13 production, establishing a positive feedback loop. CONCLUSIONS: LF formation in COPD may be driven by lung B cells via a CXCL13-dependent mechanism that involves toll-like receptor and lymphotoxin receptor signaling.


Subject(s)
B-Lymphocytes/metabolism , Chemokine CXCL13/biosynthesis , Lymphoid Tissue/pathology , Lymphotoxin-alpha/metabolism , Neovascularization, Pathologic/immunology , Toll-Like Receptors/metabolism , Aged , B-Lymphocytes/immunology , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Female , Humans , Lymphoid Tissue/immunology , Lymphoid Tissue/metabolism , Lymphotoxin-alpha/immunology , Male , Middle Aged , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Pulmonary Disease, Chronic Obstructive/immunology , Pulmonary Disease, Chronic Obstructive/metabolism , Signal Transduction/immunology , Sputum/chemistry , Sputum/cytology , Toll-Like Receptors/immunology
10.
BMC Pulm Med ; 11: 35, 2011 Jun 02.
Article in English | MEDLINE | ID: mdl-21635782

ABSTRACT

BACKGROUND: Little is known about the effect of smoking cessation on airway inflammation. Secretory Leukocyte Protease Inhibitor (SLPI), Clara Cell protein 16 (CC16), elafin and human defensin beta-2 (HBD-2) protect human airways against inflammation and oxidative stress. In this longitudinal study we aimed to investigate changes in sputum and nasal lavage SLPI, CC16, elafin and HBD-2 levels in healthy smokers after 6 and 12 months of smoking cessation. METHODS: Induced sputum and nasal lavage was obtained from healthy current smokers (n = 76) before smoking cessation, after 6 months of smoking cessation (n = 29), after 1 year of smoking cessation (n = 22) and from 10 healthy never smokers. SLPI, CC16, elafin and HBD-2 levels were measured in sputum and nasal lavage supernatants by commercially available ELISA kits. RESULTS: Sputum SLPI and CC-16 levels were increased in healthy smokers before smoking cessation versus never-smokers (p = 0.005 and p = 0.08 respectively). SLPI and CC16 levels did not differ before and 6 months after smoking cessation (p = 0.118 and p = 0.543 respectively), neither before and 1 year after smoking cessation (p = 0.363 and p = 0.470 respectively). Nasal lavage SLPI was decreased 12 months after smoking cessation (p = 0.033). Nasal lavage elafin levels were increased in healthy smokers before smoking cessation versus never-smokers (p = 0.007), but there were no changes 6 months and 1 year after smoking cessation. CONCLUSIONS: Only nasal lavage SLPI decrease after 1 year after smoking cessation. We may speculate that there is an ongoing inflammatory process stimulating the production of counter-regulating proteins in the airways of healthy ex-smokers.


Subject(s)
Elafin/metabolism , Nasal Lavage Fluid , Secretory Leukocyte Peptidase Inhibitor/metabolism , Smoking Cessation , Smoking/metabolism , Sputum/metabolism , Uteroglobin/metabolism , beta-Defensins/metabolism , Adult , Biomarkers/metabolism , Case-Control Studies , Female , Humans , Longitudinal Studies , Male , Middle Aged , Time Factors
11.
Exp Lung Res ; 36(6): 373-80, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20653472

ABSTRACT

Chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF) may be caused by epithelial cell injury. Epithelial cells respond to injury by secreting innate immunity proteins. To investigate whether altered levels of innate immunity proteins are observed in COPD and IPF, the authors assessed secretory leukocyte protease inhibitor (SLPI), elafin, CC16, and beta-defensin-2 levels by enzyme-linked immunosorbent assay (ELISA) in sputum supernatants from COPD patients (n = 19), smokers without COPD (n = 21), and never-smokers (n = 10) and in BALF supernatants from patients with IPF (n = 11) and subjects without IPF (n = 11). CC16 levels were decreased, whereas SLPI and elafin levels were increased in COPD patients (0.8 [0-4.2] microg/mL, 2.5 [0.3-10.5] microg/mL, 213 [152-318] pg/mL, respectively) compared to smokers without COPD (1.8 [0.1-21.2] microg/mL, 0.8 [0.2-2.6] microg/mL, 172 [71-473] pg/mL, respectively) and never-smokers (0.5 [0-4.8] microg/mL, 0.1 [0.05-0.6] microg/mL, 188 [129-218] pg/mL, respectively) (CC16: P = .001; SLPI: P <.001; elafin: P = .041). beta-Defensin-2 was detected in smokers without COPD (98 [10-729] pg/mL) and never-smokers (74 [35-410] pg/mL), but not in COPD. SLPI and elafin levels did not differ between IPF patients and controls, but CC16 levels were increased in IPF (0.5 [0-2.3] versus 0.2 [0-0.3] microg/mL; P = .019). beta-Defensin-2 was not detected in BALF. In conclusion, in COPD, secretion of CC16 and beta-defensin-2 might be suppressed, whereas SLPI and elafin secretion is up-regulated. In IPF, only CC16 secretion is up-regulated.


Subject(s)
Idiopathic Pulmonary Fibrosis/immunology , Immunity, Innate , Pulmonary Disease, Chronic Obstructive/immunology , Respiratory Mucosa/immunology , Adult , Aged , Biomarkers/analysis , Bronchoalveolar Lavage Fluid/immunology , Elafin/analysis , Enzyme-Linked Immunosorbent Assay , Female , Greece , Humans , Male , Middle Aged , Secretory Leukocyte Peptidase Inhibitor/analysis , Smoking/immunology , Smoking Cessation , Sputum/immunology , Uteroglobin/analysis , Young Adult , beta-Defensins/analysis
12.
Int Arch Allergy Immunol ; 150(4): 389-97, 2009.
Article in English | MEDLINE | ID: mdl-19571571

ABSTRACT

BACKGROUND: Asthmatics who smoke have decreased pulmonary mature dendritic cells (DCs). Chronic obstructive pulmonary disease (COPD) patients have an increased amount of pulmonary immature DCs. We hypothesized that healthy smokers and patients with COPD have decreased pulmonary mature DCs. METHODS: We identified sputum DCs expressing the maturation markers CD83 and DC-lysosome associated membrane protein (DC-LAMP) and DC subpopulations (i.e. myeloid and plasmacytoid DCs) by flow cytometry in healthy smokers before they entered a smoking cessation trial (n = 30), in the same smokers after 6 months of smoking cessation (n = 11) and in COPD patients (n = 28, 14 current and 14 ex-smokers). 12 healthy never-smokers served as controls. DC numbers were expressed as percentage of total sputum CD45(+) leukocytes. RESULTS: CD83(+) and DC-LAMP(+) mature DCs were decreased in healthy smokers before they ceased smoking compared to after (p = 0.003 and p = 0.049, respectively) and in smokers before smoking cessation compared to never-smokers (p = 0.027 and p = 0.028, respectively). COPD patients, both current and ex-smokers, showed decreased CD83(+) mature DCs compared to never-smokers and smokers after cessation (p = 0.042 and p = 0.004, respectively). CONCLUSIONS: Cigarette smoking and COPD per se are associated with a decrease in pulmonary mature DCs. We speculate that this reduction is involved in the immunopathogenesis of smoking-related respiratory disorders, such as COPD.


Subject(s)
Antigens, Differentiation/metabolism , Dendritic Cells/metabolism , Pulmonary Disease, Chronic Obstructive/immunology , Smoking/immunology , Sputum/immunology , Adult , Aged , Antigens, CD/biosynthesis , Antigens, Differentiation/immunology , Cell Count , Dendritic Cells/immunology , Dendritic Cells/pathology , Female , Humans , Immunoglobulins/biosynthesis , Immunomodulation , Leukocyte Common Antigens/biosynthesis , Lysosomal Membrane Proteins/biosynthesis , Male , Membrane Glycoproteins/biosynthesis , Middle Aged , Pulmonary Disease, Chronic Obstructive/etiology , Pulmonary Disease, Chronic Obstructive/pathology , Pulmonary Disease, Chronic Obstructive/physiopathology , Pulmonary Disease, Chronic Obstructive/prevention & control , Smoking/adverse effects , Smoking/pathology , Smoking/physiopathology , Smoking/therapy , Smoking Cessation , Sputum/cytology , CD83 Antigen
13.
Chest ; 136(3): 726-733, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19465512

ABSTRACT

BACKGROUND: Dendritic cells (DCs) have been reported to be increased in the small airways of patients with COPD, but the maturity status of these cells is unclear. We have quantified the numbers of cells expressing markers associated with DC maturation. METHODS: Lung tissue was obtained at resection for lung cancer from 41 patients with COPD (30 current smokers and 11 ex-smokers; 32 steroid-treated patients and 9 steroid-naïve patients), 19 ex-smokers without COPD and 9 never-smokers without COPD. Tissue sections were immunostained for CD1a to mark immature DCs, and for CD83, fascin, and DC-lysosome-associated membrane protein (DC-LAMP) to delineate mature DCs. RESULTS: The volume density (ie, the volume of DCs as the percentage volume of the airway wall) comprising CD83+ DCs was significantly reduced in patients with COPD (median, 0; range, 0 to 5.1%) vs smokers (median, 2.8%; range, 0 to 10.2%) and never-smokers (median, 1.9%; range, 0.8 to 5.1%) without COPD (p = 0.000 and 0.012, respectively). Using a semiquantitative score for the alveolar wall, CD83+ DCs also were decreased in patients with COPD (median, 0; range, 0 to 2%) vs smokers (median, 1%; range, 0 to 2%) and never-smokers (median, 1%; range, 0.7 to 2%) without COPD (p = 0.004 and 0.04, respectively). No differences were detected in CD83+ DCs between current smokers and ex-smokers with COPD or between steroid-treated and steroid-naive patients. No differences were detected in CD1a+ DCs. Fascin and DC-LAMP were found to have poor specificity for mature DCs. CONCLUSIONS: COPD is associated with decreased numbers of (mature) CD83+ DCs in small airways and alveoli. The relevance of such a reduction on pulmonary immune responses requires further investigation.


Subject(s)
Dendritic Cells/immunology , Pulmonary Disease, Chronic Obstructive/immunology , Aged , Antigens, CD/metabolism , Carrier Proteins/metabolism , Dendritic Cells/pathology , Female , Humans , Immunoglobulins/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/surgery , Lysosomal Membrane Proteins/metabolism , Male , Membrane Glycoproteins/metabolism , Microfilament Proteins/metabolism , Middle Aged , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Disease, Chronic Obstructive/pathology , Smoking/immunology , Smoking/pathology , Statistics, Nonparametric , CD83 Antigen
14.
Respir Med ; 103(6): 935-8, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19328670

ABSTRACT

BACKGROUND: Myeloid and plasmacytoid dendritic cell (DC) subsets have been recently identified in the human lung based on their differential expression of Blood DC Antigens 1-3 (BDCAs). We investigated the expression of these antigens by isolated human pulmonary CD1a(+) DCs, namely Langerhan's cells. METHODS: Using an in vitro cell culture system we successfully isolated a population of relatively pure (>70%) CD1a(+) cells from human lung tissue (n=5 subject samples) and stained these with antibodies against the myeloid DC markers BDCA1 (CD1c) and BDCA3 (CD303), the plasmacytoid DC marker BDCA2 (CD141), the Langerhan's cell marker Langerin and the maturation marker CD83. RESULTS: Among different subject samples, the isolated CD1a(+) cells showed variable expression of Langerin, BDCAs and CD83. Interestingly, in two subject samples, which contained >70% CD83(+) mature CD1a(+) cells, >50% of the cells were positive for all of the BDCAs. CONCLUSIONS: We conclude that isolated pulmonary CD1a(+) DCs in vitro have the capacity to express both myeloid and plasmacytoid BDCA markers and that rather than subset restriction in pulmonary DCs, a significant degree of flexibility/plasticity can be induced, albeit experimentally.


Subject(s)
Antigens, Surface/metabolism , Langerhans Cells/immunology , Aged , Aged, 80 and over , Antigens, CD1/immunology , Cells, Cultured , Female , Glycoproteins , Humans , In Vitro Techniques , Langerhans Cells/cytology , Lung/cytology , Male , Middle Aged , Phenotype
15.
Mediators Inflamm ; 2009: 812102, 2009.
Article in English | MEDLINE | ID: mdl-20182552

ABSTRACT

Little is known about the longitudinal effects of smoking cessation on sputum inflammatory cells. We aimed to investigate the changes in sputum inflammatory cells and T-lymphocyte subpopulations after 6 and 12 months smoking cessation. Induced sputum was obtained from 68 healthy smokers before and after 6 months (n = 21) and 1 year (n = 14) smoking cessation and from ten healthy never-smokers. Inflammatory cells were identified by morphology and T-lymphocyte subpopulations by flow cytometry. Sputum macrophages were decreased after 12 months of smoking cessation in comparison to baseline, while neutrophils increased. Moreover, CD8+ T-cells were decreased in smokers before smoking cessation compared to never-smokers and increased in smokers after 6 months of smoking cessation in comparison to baseline; result that was maintained after 1 year of smoking cessation. These novel findings indicate that smoking cessation can equilibrate certain inflammatory cells of smokers with those of nonsmokers, within 6 months of smoking cessation.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Smoking Cessation , Sputum , T-Lymphocyte Subsets/immunology , Adult , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Female , Humans , Male , Middle Aged , Risk Factors , Sputum/cytology , Sputum/immunology
16.
Am J Respir Crit Care Med ; 177(11): 1180-6, 2008 Jun 01.
Article in English | MEDLINE | ID: mdl-18337593

ABSTRACT

Dendritic cells (DCs) are professional antigen-presenting cells responsible for immune homeostasis. In the lung's responses to tissue damage or infection, they initiate and orchestrate innate and adaptive immunity. There are immature and mature states and at least three phenotypic and functional subsets. DCs circulate in the blood and localize to mucosal surfaces in immature form where they act as sentinels, sampling constituents of the external environment that breach the epithelium. With internalization of antigen, they are activated, mature, and migrate to draining lymph nodes to induce the proliferation and regulate the balance of Th1/Th2 T cells or to induce a state of tolerance, the last dependent on maturation status, extent of cell surface costimulatory molecule expression, and cytokine release. Cigarette smoke has modulatory effects varying with species, dose, the location examined within the lung, and the marker or technique used to identify DCs. Healthy smokers (and smokers with asthma) have reduced numbers of large airway mature DCs. In chronic obstructive pulmonary disease, the number of immature DCs is increased in small airways, whereas in smokers with chronic obstructive pulmonary disease, the total number of DCs appears to be reduced in large airways. We hypothesize that the long-term effects of cigarette smoke include reduction of DC maturation and function, changes that favor repeated infection, increased exacerbation frequency, and the altered (CD8(+) T-cell predominant) pattern of inflammation associated with this progressive chronic disease.


Subject(s)
Dendritic Cells/physiology , Pulmonary Disease, Chronic Obstructive/etiology , Smoking/adverse effects , Antigens, CD/physiology , Dendritic Cells/drug effects , Ganglionic Stimulants/adverse effects , Humans , Nicotine/adverse effects , Pulmonary Disease, Chronic Obstructive/pathology , Nicotiana/adverse effects
18.
Am J Respir Crit Care Med ; 175(9): 919-25, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17303795

ABSTRACT

RATIONALE: Cigarette smoking worsens asthma and is associated with reduced response to corticosteroid therapy. As cigarette smoke is known to have immunomodulatory effects, we hypothesized that one mechanism by which smoking mediates its adverse effect is by reduction of the numbers of bronchial mucosal dendritic cells (DCs), which control B-cell growth and T-cell responses. OBJECTIVES: We set out to sample the bronchial mucosa in smoking and never-smoking patients with asthma and to count DCs, B cells, and cells expressing genes for two key T-lymphocyte regulatory cytokines. METHODS: Twenty-one never-smoker patients with asthma (6 steroid naive), 24 smoker patients with asthma (9 steroid naive), and 10 healthy never-smokers (control subjects) were recruited and their endobronchial biopsy samples were immunostained for detection of mature DCs (CD83(+)), Langerhans cells (CD1a(+)), B lymphocytes (CD20(+)), and helper T-cell type 1 (IFN-gamma) and helper T-cell type 2 (IL-4) cytokine-expressing cells. MEASUREMENTS AND MAIN RESULTS: The number (per square millimeter) of CD83(+) mature DCs was significantly lower in smoker patients with asthma (median [range]: 37 [0, 131]) in comparison with never-smoker steroid-naive and steroid-treated patients with asthma (76 [24, 464]; p = 0.006) or control subjects (85 [40, 294]; p = 0.004). Moreover, B cells were fewer in smoker (26 [4, 234]) versus never-smoker steroid-naive and steroid-treated patients with asthma (45 [10, 447]; p = 0.01) and in smoker steroid-naive patients with asthma (23 [4, 111]) versus control subjects (34 [10, 130]; p = 0.05). The number of cells expressing IFN-gamma showed a trend toward fewer in smoker (70 [6, 24]) versus never-smoker steroid-naive patients with asthma (144 [44, 323]; p = 0.10). CONCLUSIONS: There are important and statistically significant differences in the number of CD83(+) mature DCs and B cells in the large airways of smokers with asthma. We speculate that their reductions may render patients with asthma less responsive to corticosteroids and more susceptible to infection.


Subject(s)
Asthma/immunology , Bronchi/immunology , Immunity, Mucosal/physiology , Smoking/immunology , Adolescent , Adult , Aged , Antigens, CD/metabolism , Asthma/metabolism , Asthma/pathology , Bronchi/metabolism , Bronchi/pathology , Case-Control Studies , Dendritic Cells/physiology , Female , Humans , Interferon-gamma/metabolism , Interleukin-4/metabolism , Male , Middle Aged , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Smoking/metabolism , Smoking/pathology
19.
Chest ; 129(5): 1194-202, 2006 May.
Article in English | MEDLINE | ID: mdl-16685009

ABSTRACT

STUDY OBJECTIVES: It has been suggested that patients with noneosinophilic asthma (NEA) show increased numbers of sputum neutrophils and a lack of response to therapy with corticosteroids, which are features that are commonly related to COPD. The aim of our study was to test the hypothesis that airway inflammation in NEA patients is different from that seen in patients with eosinophilic asthma (EA) and is similar to COPD. DESIGN: Sputum cellular stress markers and neutrophilic and eosinophilic fluid-phase mediators were analyzed in asthma and COPD patients. NEA patients were identified based on a sputum eosinophil count of < or = 2.2% of the total nonsquamous cell count, and were compared to EA and COPD patients. SETTING: University Hospital of Heraklion, Department of Thoracic Medicine. PATIENTS: A total of 37 atopic asthmatic patients and 25 patients with COPD. MEASUREMENTS: Sputum cell counts, cellular expression of heme oxygenase-1, inducible nitric oxide synthase, and nitrotyrosine, and sputum levels of eosinophilic cationic protein (ECP), myeloperoxidase (MPO), interleukin-8, and granulocyte macrophage colony-stimulating factor. RESULTS: A total of 17 asthmatic patients (46%) belonged to the NEA group and 20 patients (54%) to the EA group. Patients with NEA showed no difference in neutrophil counts, fluid-phase mediators, or cellular stress markers compared to patients with EA. Compared to COPD patients, NEA patients showed the following significant differences: lower total cell counts (p < 0.03); lower neutrophil counts (p < 0.01); lower nitrotyrosine positive cell counts (p < 0.003); lower ECP levels (p < 0.005); lower MPO levels (p < 0.000); higher lymphocyte counts (p < 0.01); and higher macrophage counts (p < 0.03). CONCLUSIONS: Despite low eosinophil counts, airway inflammation in NEA patients may share common features with that in EA patients but is distinct from COPD. Larger studies are needed to investigate further the clinical and inflammatory characteristics of NEA before we are able to categorize asthma patients into those with or without eosinophilic inflammation.


Subject(s)
Asthma/pathology , Eosinophil Cationic Protein/metabolism , Eosinophils/pathology , Interleukin-8/metabolism , Oxidative Stress/physiology , Peroxidase/metabolism , Sputum/cytology , Aged , Asthma/metabolism , Asthma/physiopathology , Biomarkers/metabolism , Cell Count , Eosinophils/metabolism , Female , Forced Expiratory Volume/physiology , Humans , Immunohistochemistry , Inflammation/metabolism , Inflammation/pathology , Male , Middle Aged , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Disease, Chronic Obstructive/pathology , Pulmonary Disease, Chronic Obstructive/physiopathology , Severity of Illness Index
20.
Respir Res ; 7: 80, 2006 May 22.
Article in English | MEDLINE | ID: mdl-16716229

ABSTRACT

Airway inflammation increases during acute exacerbations of COPD. Extrinsic factors, such as airway infections, increased air pollution, and intrinsic factors, such as increased oxidative stress and altered immunity may contribute to this increase. The evidence for this and the potential mechanisms by which various aetiological agents increase inflammation during COPD exacerbations is reviewed. The pathophysiologic consequences of increased airway inflammation during COPD exacerbations are also discussed. This review aims to establish a cause and effect relationship between etiological factors of increased airway inflammation and COPD exacerbations based on recently published data. Although it can be speculated that reducing inflammation may prevent and/or treat COPD exacerbations, the existing anti-inflammatory treatments are modestly effective.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Inflammation/etiology , Pulmonary Disease, Chronic Obstructive/etiology , Air Pollutants/adverse effects , Animals , Antioxidants/metabolism , Bacterial Infections/complications , Humans , Inflammation/drug therapy , Inflammation/metabolism , Neutrophils/metabolism , Oxidative Stress , Pulmonary Disease, Chronic Obstructive/drug therapy , Pulmonary Disease, Chronic Obstructive/physiopathology , Reactive Oxygen Species/metabolism , T-Lymphocytes/metabolism , Virus Diseases/complications
SELECTION OF CITATIONS
SEARCH DETAIL
...