1
|
Caramori G, Ruggeri P, Di Stefano A, Mumby S, Girbino G, Adcock IM, Kirkham P. Autoimmunity and COPD. Chest 2018; 153:1424-1431. [DOI: 10.1016/j.chest.2017.10.033] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 10/21/2017] [Accepted: 10/27/2017] [Indexed: 01/22/2023] Open
|
2
|
Mark NM, Kargl J, Busch SE, Yang GHY, Metz HE, Zhang H, Hubbard JJ, Pipavath SNJ, Madtes DK, Houghton AM. Chronic Obstructive Pulmonary Disease Alters Immune Cell Composition and Immune Checkpoint Inhibitor Efficacy in Non-Small Cell Lung Cancer. Am J Respir Crit Care Med 2018; 197:325-336. [PMID: 28934595 PMCID: PMC5803651 DOI: 10.1164/rccm.201704-0795oc] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 09/20/2017] [Indexed: 12/19/2022] Open
Abstract
RATIONALE Chronic obstructive pulmonary disease (COPD) and non-small cell lung cancer (NSCLC) are interrelated diseases with substantial mortality, and the pathogenesis of both involves aberrant immune functioning. OBJECTIVES To profile immune cell composition and function in patients with NSCLC and describe the effects of COPD on lung and tumor microenvironments. METHODS We profiled resected lung and tumor tissue using flow cytometry and T-cell receptor sequencing in patients with and without COPD from a prospective cohort of patients undergoing resection of NSCLC. A murine cigarette smoke exposure model was used to evaluate the effect on pulmonary immune populations. A separate retrospective cohort of patients who received immune checkpoint inhibitors (ICIs) was analyzed, and their survival was quantified. MEASUREMENTS AND MAIN RESULTS We observed an increased number of IFN-γ-producing CD8+ and CD4+ (T-helper cell type 1 [Th1]) lymphocytes in the lungs of patients with COPD. In both humans and mice, increased Th17 content was seen with smoke exposure, but was not associated with the development or severity of COPD. COPD-affected lung tissue displayed increased Th1 differentiation that was recapitulated in the matching tumor sample. PD-1 (programmed cell death protein 1) expression was increased in tumors of patients with COPD, and the presence of COPD was associated with progression-free survival in patients treated with ICIs. CONCLUSIONS In patients with COPD, Th1 cell populations were expanded in both lung and tumor microenvironments, and the presence of COPD was associated with longer progression-free intervals in patients treated with ICIs. This has implications for understanding the immune mediators of COPD and developing novel therapies for NSCLC.
Collapse
Affiliation(s)
- Nicholas M. Mark
- Division of Pulmonary and Critical Care, and
- Clinical Research Division and
| | - Julia Kargl
- Clinical Research Division and
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Graz, Austria
| | | | | | | | | | | | | | - David K. Madtes
- Division of Pulmonary and Critical Care, and
- Clinical Research Division and
| | - A. McGarry Houghton
- Division of Pulmonary and Critical Care, and
- Clinical Research Division and
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; and
| |
Collapse
|
3
|
Sorigué M, Juncà J, Marcé S, Cabezón M, García O, Zamora L. The role of T-cell phenotype and T-cell receptor rearrangement in the diagnosis of T-cell malignancies. Leuk Lymphoma 2015; 57:244-6. [PMID: 25956041 DOI: 10.3109/10428194.2015.1046865] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Marc Sorigué
- a Department of Hematology , ICO-Hospital Germans Trias i Pujol, Universitat Autònoma de Barcelona , Badalona , Spain
| | - Jordi Juncà
- b Josep Carreras Leukemia Research Institute, ICO-Hospital Germans Trias i Pujol, Universitat Autònoma de Barcelona , Badalona , Spain
| | - Silvia Marcé
- b Josep Carreras Leukemia Research Institute, ICO-Hospital Germans Trias i Pujol, Universitat Autònoma de Barcelona , Badalona , Spain
| | - Marta Cabezón
- b Josep Carreras Leukemia Research Institute, ICO-Hospital Germans Trias i Pujol, Universitat Autònoma de Barcelona , Badalona , Spain
| | - Olga García
- b Josep Carreras Leukemia Research Institute, ICO-Hospital Germans Trias i Pujol, Universitat Autònoma de Barcelona , Badalona , Spain
| | - Lurdes Zamora
- b Josep Carreras Leukemia Research Institute, ICO-Hospital Germans Trias i Pujol, Universitat Autònoma de Barcelona , Badalona , Spain
| |
Collapse
|
4
|
Wei L, Xu D, Qian Y, Huang G, Ma W, Liu F, Shen Y, Wang Z, Li L, Zhang S, Chen Y. Comprehensive analysis of gene-expression profile in chronic obstructive pulmonary disease. Int J Chron Obstruct Pulmon Dis 2015; 10:1103-9. [PMID: 26089660 PMCID: PMC4468932 DOI: 10.2147/copd.s68570] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
OBJECTIVE To investigate the gene-expression profile of chronic obstructive pulmonary disease (COPD) patients and explore the possible therapeutic targets. METHODS The microarray raw dataset GSE29133, including three COPD samples and three normal samples, was obtained from Gene Expression Omnibus. After data preprocessing with the Affy package, Student's t-test was employed to identify the differentially expressed genes (DEGs). The up- and downregulated DEGs were then pooled for gene-ontology and pathway-enrichment analyses using the Database for Annotation, Visualization and Integrated Discovery (DAVID). The upstream regulatory elements of these DEGs were also explored by using Whole-Genome rVISTA. Furthermore, we constructed a protein-protein interaction (PPI) network for DEGs. The surfactant protein D (SP-D) serum level and HLA-A gene frequency in COPD patients and healthy controls were also measured by enzyme-linked immunosorbent assay (ELISA) and real-time polymerase chain reaction, respectively. RESULTS A total of 39 up- and 15 downregulated DEGs were screened. Most of the upregulated genes were involved in the immune response process, while the downregulated genes were involved in the steroid metabolic process. Moreover, we also found that HLA-A has the highest degree in the PPI network. The SP-D serum level and HLA-A gene frequency in COPD patients were significantly higher than those in healthy controls (13.62±2.09 ng/mL vs 10.28±2.86 ng/mL; 62.5% vs 12.5%; P<0.05). CONCLUSION Our results may help further the understanding of the mechanisms of COPD. The identified DEGs, especially HLA-A, may serve as diagnosis markers for COPD.
Collapse
Affiliation(s)
- Lei Wei
- Department of Respiratory Disease, Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai, People's Republic of China
| | - Dong Xu
- Medical College of Soochow University, Suzhou, People's Republic of China
| | - Yechang Qian
- Department of Respiratory Disease, Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai, People's Republic of China
| | - Guoyi Huang
- Department of Respiratory Disease, Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai, People's Republic of China
| | - Wei Ma
- Department of Respiratory Disease, Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai, People's Republic of China
| | - Fangying Liu
- Department of Respiratory Disease, Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai, People's Republic of China
| | - Yanhua Shen
- Department of Respiratory Disease, Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai, People's Republic of China
| | - Zhongfu Wang
- Department of Respiratory Disease, Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai, People's Republic of China
| | - Li Li
- Department of Respiratory Disease, Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai, People's Republic of China
| | - Shanfang Zhang
- Department of Respiratory Disease, Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai, People's Republic of China
| | - Yafang Chen
- Department of Respiratory Disease, Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai, People's Republic of China
| |
Collapse
|
5
|
Faner R, Cruz T, Agusti A. Immune response in chronic obstructive pulmonary disease. Expert Rev Clin Immunol 2014; 9:821-33. [PMID: 24070046 DOI: 10.1586/1744666x.2013.828875] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a major public health problem because of its high prevalence, rising incidence and associated socio-economic cost. The inhalation of toxic particles and gases, mostly tobacco smoke, is the main risk factor for COPD. Yet, not all smokers are equally susceptible to these toxic effects and only a percentage of them develop the disease (so-called 'susceptible smokers'). This, in combination with the observation that COPD shows familial aggregation, suggests that the genetic background of the smoker is a key element in the pathogenesis of the disease. On the other hand, it is well established that 'susceptible' smokers exhibit an enhanced inflammatory response of the lung parenchyma as compared with 'resistant' smokers (i.e., those who manage to maintain lung function within the normal age range despite their habit). Importantly, in COPD patients this inflammatory response does not resolve after quitting smoking, again at variance with resistant smokers. All in all, these observations suggest that the pathogenesis of COPD may involve, in some patients, an autoimmune component which contributes to the enhanced and persistent inflammatory response that characterizes the disease. Here we: i) review briefly the pathobiology of COPD; ii) present the available scientific evidence supporting a potential role for autoimmunity in COPD; iii) propose a three-step pathogenic hypothesis in the transition from smoking to COPD; and iv) discuss potential implications for the diagnosis and treatment of this frequent, growing, devastating and costly disease.
Collapse
Affiliation(s)
- Rosa Faner
- FISIB, CIBER Enfermedades Respiratorias (CIBERES), Mallorca, Spain
| | | | | |
Collapse
|
6
|
Antal M, Braunitzer G, Mattheos N, Gyulai R, Nagy K. Smoking as a permissive factor of periodontal disease in psoriasis. PLoS One 2014; 9:e92333. [PMID: 24651659 PMCID: PMC3961310 DOI: 10.1371/journal.pone.0092333] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 02/21/2014] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Population-based studies have identified smoking as a pathogenetic factor in chronic periodontitis. At the same time, chronic periodontal disease has also been found to occur more often in persons suffering from psoriasis than in controls with no psoriasis. It is known that smoking aggravates both periodontal disease and psoriasis, but so far it has not been investigated how smoking influences the occurrence and severity of periodontal disease in psoriasis. METHODS A hospital-based study was conducted to investigate this question. The study population consisted of 82 psoriasis patients and 89 controls. All patients received a full-mouth periodontal examination, and a published classification based on bleeding on probing, clinical attachment level and probing depth was utilized for staging. Both patients and controls were divided into smoker and non-smoker groups, and the resulting groups were compared in terms of periodontal status. Beyond the descriptive statistics, odds ratios were computed. RESULTS Psoriasis in itself increased the likelihood of severe periodontal disease to 4.373 (OR, as compared to non-smoker controls, p<0.05), while smoking increased it to 24.278 (OR, as compared to non-smoker controls, p<0.001) in the studied population. In other words, the risk of severe periodontal disease in psoriasis turned out to be six times higher in smokers than in non-smokers. CONCLUSIONS The results of this study corroborate those of other studies regarding the link between psoriasis and periodontal disease, but they also seem to reveal a powerful detrimental effect of smoking on the periodontal health of psoriasis patients, whereby the authors propose that smoking may have a permissive effect on the development of severe periodontal disease in psoriasis.
Collapse
Affiliation(s)
- Márk Antal
- University of Szeged, Faculty of Dentistry, Department of Operative and Esthetic Dentistry, Szeged, Hungary
| | - Gábor Braunitzer
- University of Szeged, Faculty of Dentistry, Department of Oral Surgery, Szeged, Hungary
| | - Nikos Mattheos
- The University of Hong Kong, Faculty of Dentistry, Department of Oral Rehabilitation, Hong Kong, SAR PR China
| | - Rolland Gyulai
- University of Szeged, Faculty of Medicine, Department of Dermatology and Allergology, Szeged, Hungary and University of Pécs, Department of Dermatology, Venerology and Oncodermatology, Pécs, Hungary
| | - Katalin Nagy
- University of Szeged, Faculty of Dentistry, Department of Oral Surgery, Szeged, Hungary
| |
Collapse
|
7
|
Podolin PL, Foley JP, Carpenter DC, Bolognese BJ, Logan GA, Long E, Harrison OJ, Walsh PT. T cell depletion protects against alveolar destruction due to chronic cigarette smoke exposure in mice. Am J Physiol Lung Cell Mol Physiol 2013; 304:L312-23. [DOI: 10.1152/ajplung.00152.2012] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The role of T cells in chronic obstructive pulmonary disease (COPD) is not well understood. We have previously demonstrated that chronic cigarette smoke exposure can lead to the accumulation of CD4+ and CD8+ T cells in the alveolar airspaces in a mouse model of COPD, implicating these cells in disease pathogenesis. However, whether specific inhibition of T cell responses represents a therapeutic strategy has not been fully investigated. In this study inhibition of T cell responses through specific depleting antibodies, or the T cell immunosuppressant drug cyclosporin A, prevented airspace enlargement and neutrophil infiltration in a mouse model of chronic cigarette smoke exposure. Furthermore, individual inhibition of either CD4+ T helper or CD8+ T cytotoxic cells prevented airspace enlargement to a similar degree, implicating both T cell subsets as critical mediators of the adaptive immune response induced by cigarette smoke exposure. Importantly, T cell depletion resulted in significantly decreased levels of the Th17-associated cytokine IL-17A, and of caspase 3 and caspase 7 gene expression and activity, induced by cigarette smoke exposure. Finally, inhibition of T cell responses in a therapeutic manner also inhibited cigarette smoke-induced airspace enlargement, IL-17A expression, and neutrophil influx in mice. Together these data demonstrate for the first time that therapeutic inhibition of T cell responses may be efficacious in the treatment of COPD. Given that broad immunosuppression may be undesirable in COPD patients, this study provides proof-of-concept for more targeted approaches to inhibiting the role of T cells in emphysema development.
Collapse
Affiliation(s)
- Patricia L. Podolin
- Respiratory Therapeutic Area, GlaxoSmithKline, King Of Prussia, Pennsylvania; and
| | - Joseph P. Foley
- Respiratory Therapeutic Area, GlaxoSmithKline, King Of Prussia, Pennsylvania; and
| | - Donald C. Carpenter
- Respiratory Therapeutic Area, GlaxoSmithKline, King Of Prussia, Pennsylvania; and
| | - Brian J. Bolognese
- Respiratory Therapeutic Area, GlaxoSmithKline, King Of Prussia, Pennsylvania; and
| | - Gregory A. Logan
- Respiratory Therapeutic Area, GlaxoSmithKline, King Of Prussia, Pennsylvania; and
| | - Edward Long
- Respiratory Therapeutic Area, GlaxoSmithKline, King Of Prussia, Pennsylvania; and
| | - Oliver J. Harrison
- Respiratory Therapeutic Area, GlaxoSmithKline, King Of Prussia, Pennsylvania; and
| | - Patrick T. Walsh
- Respiratory Therapeutic Area, GlaxoSmithKline, King Of Prussia, Pennsylvania; and
- School of Medicine, Trinity College Dublin, National Childrens Research Centre, Our Ladys Childrens Hospital, Crumlin, Dublin, Ireland
| |
Collapse
|
8
|
Fujino N, Ota C, Takahashi T, Suzuki T, Suzuki S, Yamada M, Nagatomi R, Kondo T, Yamaya M, Kubo H. Gene expression profiles of alveolar type II cells of chronic obstructive pulmonary disease: a case-control study. BMJ Open 2012; 2:bmjopen-2012-001553. [PMID: 23117565 PMCID: PMC3532994 DOI: 10.1136/bmjopen-2012-001553] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
OBJECTIVES The aim of this study was to identify the gene expression pattern specific in alveolar epithelial type II cells (ATII cells) isolated from patients with chronic obstructive pulmonary disease (COPD). DESIGN Case control. SETTING Two hospitals in Japan. PARTICIPANTS Three patients without COPD and three patients with COPD in microarray analyses. Five smokers without COPD and nine smokers with COPD in the following analyses. PRIMARY AND SECONDARY OUTCOME MEASURED: Primary outcome included identification of differentially expressed genes and activated or inhibited pathways in ATII cells of the patients with COPD, compared to those of the patients without COPD, using Affymetrix gene expression arrays. Secondary outcome included validation of the results of microarray analyses by quantitative reverse transcription-PCR. RESULTS We isolated ATII cells from COPD and non-COPD lungs using fluorescence-activated cell sorting. We performed Affymetrix gene expression arrays on both types of ATII cells. Gene set enrichment analyses revealed that two major gene sets were enriched in ATII cells from COPD lungs: interferon-responsive gene sets and gene sets associated with cell cycle progression. Gene ontology term enrichment analyses indicated that among the interferon-stimulated genes, ATII cells in COPD expressed genes such as PSMB8, PSMB9, TAP1 and TAP2 associated with the antigen processing and presentation pathway. We validated the results of the microarray analyses using quantitative reverse transcriptase-PCR. In addition, FACS analysis indicated that the percentage of ATII cells to CD45-negative lung cells isolated from COPD lungs were significantly increased more than that from non-COPD lungs. CONCLUSIONS Our study demonstrated that interferon-stimulated genes involved in the antigen processing and presentation pathway and genes involved in cell cycle progression were enriched in ATII cells of the patients with COPD. These pathways might alter phenotypes of ATII cells in COPD lungs.
Collapse
Affiliation(s)
- Naoya Fujino
- Department of Advanced Preventive Medicine for Infectious Disease, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Chiharu Ota
- Department of Advanced Preventive Medicine for Infectious Disease, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Toru Takahashi
- Department of Advanced Preventive Medicine for Infectious Disease, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takaya Suzuki
- Department of Thoracic Surgery, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Satoshi Suzuki
- Department of Thoracic Surgery, Japanese Red Cross Ishinomaki Hospital, Ishinomaki, Japan
| | - Mitsuhiro Yamada
- Department of Infection Control and Laboratory Diagnostics, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Ryouichi Nagatomi
- Department of Biomedical Engineering, Tohoku University Graduate School of Biomedical Engineering, Sendai, Japan
| | - Takashi Kondo
- Department of Thoracic Surgery, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Mutsuo Yamaya
- Department of Advanced Preventive Medicine for Infectious Disease, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hiroshi Kubo
- Department of Advanced Preventive Medicine for Infectious Disease, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
9
|
Taylor JD. COPD and the response of the lung to tobacco smoke exposure. Pulm Pharmacol Ther 2010; 23:376-83. [PMID: 20382250 DOI: 10.1016/j.pupt.2010.04.003] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2009] [Revised: 03/18/2010] [Accepted: 04/03/2010] [Indexed: 11/28/2022]
Abstract
Chronic Obstructive Pulmonary Disease (COPD) is a major cause of death in the western world and increasing in prevalence in developing countries. COPD is characterised by irreversible airflow obstruction, loss of lung tissue, reduced quality of life and high rates of mortality. The major cause of COPD is tobacco smoke. The changes in the innate immune system directed by tobacco smoke exposure lead to a pronounced and chronic inflammation in the lung. This in turn leads to other pathological changes including remodelling and destruction of lung tissue. Tobacco smoke exposure also leads to infection of the lung by bacteria and viruses. These, bacteria, viruses and co-infection are key triggers of acute worsening's of COPD termed exacerbations. COPD exacerbations are an additional major factor in the morbidity and mortality within COPD and are also the major healthcare costs associated with the disease. Within this review we discuss the response of the immune system to cigarette smoke exposure and inappropriate harmful responses. Successful treatment strategies will need to balance the positive effects of reducing inflammatory aspects of the disease whilst retaining some of the needed immune responses triggered by tobacco smoke exposure.
Collapse
Affiliation(s)
- John D Taylor
- Integrative Pharmacology, Biosciences R&D Lung, Respiratory & Inflammation Research Area, AstraZeneca, Lund, Sweden.
| |
Collapse
|
10
|
Motz GT, Eppert BL, Wesselkamper SC, Flury JL, Borchers MT. Chronic cigarette smoke exposure generates pathogenic T cells capable of driving COPD-like disease in Rag2-/- mice. Am J Respir Crit Care Med 2010; 181:1223-33. [PMID: 20133926 DOI: 10.1164/rccm.200910-1485oc] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Pathogenic T cells drive, or sustain, a number of inflammatory diseases. Chronic obstructive pulmonary disease (COPD) is an inflammatory lung disease associated with the accumulation of activated T cells. We previously demonstrated that chronic cigarette smoke (CS) exposure causes oligoclonal expansion of lung CD4(+) T cells and CD8(+) T cells in a mouse model of COPD, thus implicating these cells in disease pathogenesis. OBJECTIVES To determine whether T cells are pathogenic in a CS-induced mouse model of COPD. METHODS We transferred lung CD3(+) T cells from filtered air (FA)- and CS-exposed mice into Rag2(-/-) recipients. Endpoints associated with the COPD phenotype were then measured. MEASUREMENTS AND MAIN RESULTS Here, we demonstrate that chronic CS exposure generates pathogenic T cells. Transfer of CD3(+) T cells from the lungs of CS-exposed mice into Rag2(-/-) recipients led to substantial pulmonary changes pathognomonic of COPD. These changes included monocyte/macrophage and neutrophil accumulation, increased expression of cytokines and chemokines, activation of proteases, apoptosis of alveolar epithelial cells, matrix degradation, and airspace enlargement reminiscent of emphysema. CONCLUSIONS These data formally demonstrate, for the first time, that chronic CS exposure leads to the generation of pathogenic T cells capable of inducing COPD-like disease in Rag2(-/-) mice. This report provides novel insights into COPD pathogenesis.
Collapse
Affiliation(s)
- Gregory T Motz
- Department of Environmental Health, Division of Environmental Genetics and Molecular Toxicology, University of Cincinnati, College of Medicine, Cincinnati, OH 45267-0056, USA
| | | | | | | | | |
Collapse
|
11
|
Stämpfli MR, Anderson GP. How cigarette smoke skews immune responses to promote infection, lung disease and cancer. Nat Rev Immunol 2009; 9:377-84. [PMID: 19330016 DOI: 10.1038/nri2530] [Citation(s) in RCA: 471] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
A complex and multilayered immune defence system protects the host against harmful agents and maintains tissue homeostasis. Cigarette smoke exposure markedly impacts the immune system, compromising the host's ability to mount appropriate immune and inflammatory responses and contributing to smoking-related pathologies. These adverse effects on the immune system not only occur in active smokers, but also in those exposed to smoke passively in contaminated environments, and may persist for decades after exposure has ended.
Collapse
Affiliation(s)
- Martin R Stämpfli
- Department of Pathology and Molecular Medicine, Centre for Gene Therapeutics, McMaster University, Hamilton, Ontario, Canada.
| | | |
Collapse
|
12
|
Zhu X, Gadgil AS, Givelber R, George MP, Stoner MW, Sciurba FC, Duncan SR. Peripheral T cell functions correlate with the severity of chronic obstructive pulmonary disease. THE JOURNAL OF IMMUNOLOGY 2009; 182:3270-7. [PMID: 19234225 DOI: 10.4049/jimmunol.0802622] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Adaptive immune processes have been implicated in the pathogenesis of chronic obstructive pulmonary disease (COPD). We hypothesized that peripheral T cell abnormalities may be present in afflicted patients. We tested this hypothesis by characterizing circulating T cells in COPD patients and correlated these findings with disease severity, smoking status, and use of inhaled glucocorticosteroids (ICS). Compared with normal controls, a lesser proportion of peripheral CD4 T cells from COPD subjects produced IL-10, whereas the CD8 T cells from these patients were more often activated and more frequently produced both IFN-gamma and IL-4. COPD severity was significantly and inversely associated with the proportion of circulating CD4 T cells and directly correlated with CD4 production of IL-2, as well as frequency of CD8 T cell activation and CD8 IFN-gamma production. Adjustments for current smoking status and ICS use by linear regression showed independent, and generally inhibitory, effects of these clinical variables on the abnormal T cell functions of these patients. We conclude that circulating T cells from COPD patients are abnormally activated and elaborate proinflammatory mediators with admixed features of Th1 and Th2 responses. Furthermore, many of these effector processes are significantly correlated with disease severity. These findings further implicate adaptive immune processes in COPD progression and indicate that facile assays of peripheral lymphocytes may provide useful insights into disease mechanisms. Current smoking and ICS use had independent effects on T cell functions among the COPD subjects, illustrating the importance of controlling for clinical parameters as covariates in immunological studies of patients afflicted with this disease.
Collapse
Affiliation(s)
- Xuehai Zhu
- Division of Pulmonary, Allergy and Critical Care, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | | | | | | | | | | | | |
Collapse
|
13
|
Gadgil A, Duncan SR. Role of T-lymphocytes and pro-inflammatory mediators in the pathogenesis of chronic obstructive pulmonary disease. Int J Chron Obstruct Pulmon Dis 2009; 3:531-41. [PMID: 19281072 PMCID: PMC2650590 DOI: 10.2147/copd.s1759] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is the fourth leading cause of death in the US and a major worldwide healthcare problem. The pathophysiologic mechanisms that drive development and progression of this disease are complex and only poorly understood. While tobacco smoking is the primary risk factor, other disease processes also appear to play a role. Components of the innate immune system (eg, macrophages and neutrophils) have long been believed to be important in the development of COPD. More recent evidence also suggests involvement of the adaptive immune system in pathogenesis of this disease. Here we will review the literature supporting the participation of T-cells in the development of COPD, and comment on the potential antigenic stimuli that may account for these responses. We will further explore the prospective contributions of T-cell derived mediators that could contribute to the inflammation, alveolar wall destruction, and small airway fibrosis of advanced COPD. A better understanding of these complex immune processes will lead to new insights that could result in improved preventative and/or treatment strategies.
Collapse
Affiliation(s)
- Aneal Gadgil
- Division of Pulmonary,Allergy and Critical Care Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | | |
Collapse
|
14
|
Motz GT, Eppert BL, Sun G, Wesselkamper SC, Linke MJ, Deka R, Borchers MT. Persistence of lung CD8 T cell oligoclonal expansions upon smoking cessation in a mouse model of cigarette smoke-induced emphysema. THE JOURNAL OF IMMUNOLOGY 2008; 181:8036-43. [PMID: 19017996 DOI: 10.4049/jimmunol.181.11.8036] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The role of adaptive immunity in the development or progression of chronic obstructive pulmonary disease (COPD) remains undefined. Recently, the presence of autoantibodies and autoreactive T cells has been demonstrated in COPD patients. In addition, oligoclonal expansions of lung T cells have been observed in COPD patients, but the overlapping incidence of infections, tumors, and cigarette smoke exposure obscures the antigenic stimulus. We analyzed the TCR Vbeta repertoire of CD4 and CD8 T cells purified from the lungs and spleens of mice chronically exposed to cigarette smoke. In a mouse model of COPD, we demonstrate that chronic cigarette smoke exposure causes oligoclonal expansions of T cells isolated from the lungs, but not spleens. TCR Vbeta repertoire analyses revealed oligoclonal expansions predominantly occurred in lung CD8 T cells, with preferential usage of Vbeta7, Vbeta9, Vbeta13, and Vbeta14. Using nucleotide sequence analysis based on Jbeta analyses, we demonstrate selection of CDR3 amino acid motifs, which strongly suggests Ag-driven oligoclonal T cell expansion. Analysis of the lung TCR Vbeta repertoire of mice with cigarette smoke-induced emphysema, which had undergone smoking cessation for 6 mo, revealed that oligoclonal expansions persisted. This study formally demonstrates that chronic cigarette smoke exposure, alone, causes a persistent adaptive T cell immune response. These findings have important implications for therapeutic approaches in the treatment of COPD, and provide insight into potential mechanisms involved in disease pathogenesis.
Collapse
Affiliation(s)
- Gregory T Motz
- Department of Environmental Health, Division of Environmental Genetics and Molecular Toxicology, University of Cincinnati, College of Medicine, Cincinnati, OH 45267-0056, USA
| | | | | | | | | | | | | |
Collapse
|
15
|
Yoshida T, Tuder RM. Pathobiology of cigarette smoke-induced chronic obstructive pulmonary disease. Physiol Rev 2007; 87:1047-82. [PMID: 17615396 DOI: 10.1152/physrev.00048.2006] [Citation(s) in RCA: 375] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Chronic obstructive pulmonary diseases (COPD), comprised of pulmonary emphysema, chronic bronchitis, and structural and inflammatory changes of small airways, is a leading cause of morbidity and mortality in the world. A better understanding of the pathobiology of COPD is critical for the developing of novel therapies, as the majority of patients with the disease have little therapeutic options at the present time. The pathobiology of COPD encompasses multiple injurious processes including inflammation (excessive or inappropriate innate and adaptive immunity), cellular apoptosis, altered cellular and molecular alveolar maintenance program, abnormal cell repair, extracellular matrix destruction (protease and anti-protease imbalance), and oxidative stress (oxidant and antioxidant imbalance). These processes are triggered by urban and rural air pollutants and active and/or passive cigarette smoke and modified by cellular senescence and infection. A series of receptor-mediated signal transduction pathways are activated by reactive oxygen species and tobacco components, resulting in impairment of a variety of cell signaling and cytokine networks, subsequently leading to chronic airway responses with mucus production, airway remodeling, and alveolar destruction. The authors provide an updated insight into the molecular and cellular pathobiology of COPD based on human and/or animal data.
Collapse
Affiliation(s)
- Toshinori Yoshida
- Division of Cardiopulmonary Pathology, Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, Maryland 21205, USA
| | | |
Collapse
|
16
|
Tuder RM, Yoshida T, Arap W, Pasqualini R, Petrache I. State of the art. Cellular and molecular mechanisms of alveolar destruction in emphysema: an evolutionary perspective. Ann Am Thorac Soc 2006; 3:503-10. [PMID: 16921129 PMCID: PMC2647641 DOI: 10.1513/pats.200603-054ms] [Citation(s) in RCA: 129] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Emphysema consists of a unique pattern of alveolar destruction, resulting in marked airspace enlargement with reduction of alveolar capillary exchange area. Classical concepts of the pathogenesis of emphysema have relied on the paradigm set by the inflammation and protease/antiprotease imbalance. We propose herein that cigarette smoke constitutes an environmental hazard that causes alveolar destruction by the interaction of apoptosis, oxidative stress, and protease/antiprotease imbalance. We draw a parallel between organismal aging, organ structural maintenance, and the damage resulting from chronic cigarette smoke inhalation. The stochastic interaction between environmental hazards and the effort of an organism or a particular organ to fend off these hazards results in the accumulation of cellular damage and features characteristic of aging. Inflammation follows as the result of the multiplication of injuries. We highlight the importance of understanding the biology of the interaction of alveolar cells in homeostasis and in alveolar destruction, and the potential role of novel processes related to senescence and stress response. An evolutionary perspective of emphysema that incorporates mechanisms related to aging may lead to important advances in the understanding and therapeutic targeting of chronic obstructive pulmonary disease.
Collapse
Affiliation(s)
- Rubin M Tuder
- Department of Pathology, Division of Cardiopulmonary Pathology, Ross Research Building, Ross 519, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Baltimore, MD 21205, USA.
| | | | | | | | | |
Collapse
|
17
|
Rosen D, Lee JH, Cuttitta F, Rafiqi F, Degan S, Sunday ME. Accelerated thymic maturation and autoreactive T cells in bronchopulmonary dysplasia. Am J Respir Crit Care Med 2006; 174:75-83. [PMID: 16574933 PMCID: PMC2662921 DOI: 10.1164/rccm.200511-1784oc] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Bronchopulmonary dysplasia (BPD), a chronic lung disease of newborns triggered by oxygen and barotrauma, is characterized by arrested alveolarization. Increased levels of bombesin-like peptides shortly after birth mediate lung injury: anti-bombesin antibody 2A11 protects against BPD in two baboon models. The role of adaptive immunity in BPD has not been explored previously. OBJECTIVES Our goal was to test the hypothesis that thymic architecture and/or T-cell function is altered with BPD, leading to autoimmunity and immunodeficiency. METHODS Thymic structure was analyzed by histopathology of thymic architecture and immunohistochemistry for thymic maturation markers (terminal deoxynucleotidyl transferase, proliferating cell nuclear antigen, CD4, and CD8). Thymic cortical epithelial cells (nurse cells) were studied using HLA-DR and protein gene product 9.5 as markers. Functional analysis was performed with "mixed lymphocyte reaction" of thymocyte or splenocyte responder cells with autologous lung cells as the stimulators. MEASUREMENTS AND MAIN RESULTS 2A11 treatment attenuates thymic cortical involution in BPD animals, sustaining terminal deoxynucleotidyl transferase-positive prothymocytes and thymocyte proliferation. BPD animals have increased CD4(+) cells in thymic cortex and lung interstitium, which are reduced by 2A11. Conversely, cortical protein gene product 9.5/HLA-DR-positive thymic nurse cells are depleted in BPD animals, but are preserved by 2A11-treatment. Whereas fetal thymocytes and splenocytes respond to phythemagglutinin/ionomycin and to a lesser extent, to autologous lung, BPD thymocytes and splenocytes are phythemagglutinin/ionomycin-unresponsive, and yet react strongly to autologous lung. The 2A11 normalizes these responses. CONCLUSIONS These observations suggest that bombesin-like peptides mediate premature thymic maturation and thymic nurse-cell depletion, leading to autoreactive T cells that could contribute to lung injury.
Collapse
Affiliation(s)
- Dennis Rosen
- Division of Pulmonary Medicine, Department of Medicine, Children's and Brigham and Women's Hospitals, and Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | | | | | |
Collapse
|
18
|
Vlahos R, Bozinovski S, Jones JE, Powell J, Gras J, Lilja A, Hansen MJ, Gualano RC, Irving L, Anderson GP. Differential protease, innate immunity, and NF-kappaB induction profiles during lung inflammation induced by subchronic cigarette smoke exposure in mice. Am J Physiol Lung Cell Mol Physiol 2005; 290:L931-45. [PMID: 16361358 DOI: 10.1152/ajplung.00201.2005] [Citation(s) in RCA: 171] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Cigarette smoke exposure is a major determinant of adverse lung health, but the molecular processes underlying its effects on inflammation and immunity remain poorly understood. Therefore, we sought to understand whether inflammatory and host defense determinants are affected during subchronic cigarette smoke exposure. Dose-response and time course studies of lungs from Balb/c mice exposed to smoke generated from 3, 6, and 9 cigarettes/day for 4 days showed macrophage- and S100A8-positive neutrophil-rich inflammation in lung tissue and bronchoalveolar lavage (BAL) fluid, matrix metalloproteinase (MMP) and serine protease induction, sustained NF-kappaB translocation and binding, and mucus cell induction but very small numbers of CD3+CD4+ and CD3+CD8+ lymphocytes. Cigarette smoke had no effect on phospho-Akt but caused a small upregulation of phospho-Erk1/2. Activator protein-1 and phospho-p38 MAPK could not be detected. Quantitative real-time PCR showed upregulation of chemokines (macrophage inflammatory protein-2, monocyte chemoattractant protein-1), inflammatory mediators (TNF-alpha, IL-1beta), leukocyte growth and survival factors [granulocyte-macrophage colony-stimulating factor, colony-stimulating factor (CSF)-1, CSF-1 receptor], transforming growth factor-beta, matrix-degrading MMP-9 and MMP-12, and Toll-like receptor (TLR)2, broadly mirroring NF-kappaB activation. No upregulation was observed for MMP-2, urokinase-type plasminogen activator, tissue-type plasminogen activator, and TLRs 3, 4, and 9. In mouse strain comparisons the rank order of susceptibility was Balb/c > C3H/HeJ > 129SvJ > C57BL6. Partition of responses into BAL macrophages vs. lavaged lung strongly implicated macrophages in the inflammatory responses. Strikingly, except for IL-10 and MMP-12, macrophage and lung gene profiles in Balb/c and C57BL/6 mice were very similar. The response pattern we observed suggests that subchronic cigarette smoke exposure may be useful to understand pathogenic mechanisms triggered by cigarette smoke in the lungs including inflammation and alteration of host defense.
Collapse
Affiliation(s)
- R Vlahos
- Department of Medicine, University of Melbourne, Royal Melbourne Hospital, Parkville, VIC 3050, Australia.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Sullivan AK, Simonian PL, Falta MT, Mitchell JD, Cosgrove GP, Brown KK, Kotzin BL, Voelkel NF, Fontenot AP. Oligoclonal CD4+ T cells in the lungs of patients with severe emphysema. Am J Respir Crit Care Med 2005; 172:590-6. [PMID: 15937291 PMCID: PMC2718531 DOI: 10.1164/rccm.200410-1332oc] [Citation(s) in RCA: 102] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Within the lungs of patients with severe emphysema, inflammation continues despite smoking cessation. Foci of T lymphocytes in the small airways of patients with emphysema have been associated with disease severity. Whether these T cells play an important role in this continued inflammatory response is unknown. OBJECTIVE The aim of this study was to determine if T cells recruited to the lungs of subjects with severe emphysema contain oligoclonal T-cell populations, suggesting their accumulation in response to antigenic stimuli. METHODS Lung T-cell receptor (TCR) Vbeta repertoire from eight patients with severe emphysema and six control subjects was evaluated at the time of tissue procurement (ex vivo) and after 2 weeks of culture with interleukin 2 (in vitro). Junctional region nucleotide sequencing of expanded TCR-Vbeta subsets was performed. RESULTS No significantly expanded TCR-Vbeta subsets were identified in ex vivo samples. However, T cells grew from all emphysema (n = 8) but from only one of the control lung samples (n = 6) when exposed to interleukin 2 (p = 0.0013). Within the cultured cells, seven major CD4-expressing TCR-Vbeta subset expansions were identified from five of the patients with emphysema. These expansions were composed of oligoclonal populations of T cells that had already been expanded in vivo. CONCLUSION Severe emphysema is associated with inflammation involving T lymphocytes that are composed of oligoclonal CD4+ T cells. These T cells are accumulating in the lung secondary to conventional antigenic stimulation and are likely involved in the persistent pulmonary inflammation characteristic of severe emphysema.
Collapse
Affiliation(s)
- Andrew K Sullivan
- Department of Medicine, University of Colorado Health Sciences Center, Denver, Colorado 80262, USA
| | | | | | | | | | | | | | | | | |
Collapse
|