Smoking decreases the response of human lung macrophages to double-stranded RNA by reducing TLR3 expression

Jill C Todt, Christine M Freeman, Jeanette P Brown, Joanne Sonstein, Theresa M Ames, Alexandra L McCubbrey, Fernando J Martinez, Stephen W Chensue, James M Beck, Jeffrey L Curtis, Jill C Todt, Christine M Freeman, Jeanette P Brown, Joanne Sonstein, Theresa M Ames, Alexandra L McCubbrey, Fernando J Martinez, Stephen W Chensue, James M Beck, Jeffrey L Curtis

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.

Figures

Figure 1
Figure 1
AMø of current smokers show reduced expression of TLR3 mRNA transcripts and intracellular protein relative to AMø of never-smokers. A. mRNA transcripts. RNA was isolated from AMø, depleted of contaminating genomic DNA, reverse-transcribed and analyzed by quantitative real-time RT-PCR using Taqman chemistry and specific primer-probe sets, normalized to GAPDH transcripts. Data are expressed on the horizontal axis as mean ± SEM for relative quantity (dRn), calculated in comparison to a single never-smoker who was arbitrarily designated the reference sample. Never-smokers (n = 6), red bars; smokers (n = 11), blue bars. *, p < 0.05 by Mann–Whitney test. B. Intracellular TLR3 (top panel) and TLR9 (bottom panel) expression by flow cytometry. AMø were permeabilized, stained for TLR3 and TLR9 expression and analyzed by flow cytometry. Data are expressed as TLR-positive AMø as mean ± SEM, never-smokers (n = 10 for TLR3, n = 5 for TLR9), red circles; smokers (n = 13 for TLR3, n = 6 for TLR9), blue circles. *, p < 0.05 by Mann–Whitney test. C. Immunohistochemical staining. Cytospins from a representative smoker (right) and a never-smoker (left), stained for intracellular TLR3 expression using AEC (red product) and hematoxylin (original 20 X). Top row, isotype control staining, bottom row, specific TLR3 staining. Representative of three experiments with similar results.
Figure 2
Figure 2
AMø TLR3 expression does not correlate with lung function or total duration of smoking exposure. AMø were harvested, processed and analyzed as described in the legend to Figure 1. A-C. TLR3 mRNA transcripts by quantitative real-time PCR (smokers, n = 11; never-smokers, n = 6, except in panel C, which includes only current or former smokers). D-F. Flow cytometric analysis of TLR3 protein expression (smokers, n = 13; never-smokers, n = 10, except in panel F which includes only current or former smokers). A, D: FEV1 % predicted; B, E: participant age (years); C, F: smoking history (pack-years); note the difference in the vertical scale of panel F. Blue circles, smokers; red crosses, never-smokers. p values by Spearman correlation.
Figure 3
Figure 3
In current smokers, the frequency of lung Mø expressing TLR3 protein is reduced relative to lung Mø of former-smokers. To isolate total lung Mø, areas of human lung parenchyma, resected for clinical indications and remote from any evidence of nodules or infection, were processed by mechanical disaggregation without the use of enzymes. Lung Mø were permeabilized, stained for TLR3 expression and analyzed by flow cytometry. A, B. Representative histograms of intracellular TLR3 expression; A, former smoker; B, current smoker. C. Percentage of lung Mo expressing TLR3 relative to smoking status; x’s represent former smokers (n = 17); circles represent current smokers (n = 8). D. Percentage of TLR3-positive by lung Mø relative to COPD status; as in panel C, x’s represent former smokers (n = 17); circles represent current smokers (n = 8); *, p < 0.05; NS, non-significant. The Mann–Whitney test was used to calculate p values.
Figure 4
Figure 4
Exposure to CSE specifically and rapidly reduces TLR3 gene expression in a differentiated human Mø cell line. THP-1 cells differentiated using PMA & vitamin D3 for 48 hours were cultured in complete medium alone or with various concentration of CSE for an additional 4 hr, then analyzed by quantitative real-time RT-PCR. Data are mean ± SEM of four independent experiments, expressed as dRn (relative to differentiated THP-1 cells cultures in complete medium alone). A, TLR3; B, TLR7; C, TLR8; D, TLR9; E, RIG-1; F, MDA-5; G, PKR. Kruskal-Wallis one-way ANOVA was used to compare groups. *, p < 0.05, compared to cells that did not receive CSE exposure.
Figure 5
Figure 5
Production of CXCL10 by AMø of smokers in response to poly(I:C) stimulation is impaired. AMø from never-smokers (n = 4) (white and red columns) and smokers (n = 5) (light and dark blue columns) were cultured for 24 h in either complete medium alone or with poly(I:C). Supernatants were collected and CXCL10 protein was measured by Luminex assay; results are expressed as the fold-increase in CXCL10 over the unstimulated control condition; note the logarithmic scale of the vertical axis. Data are mean ± SEM. One-way ANOVA with Bonferroni’s Multiple Comparison post-hoc testing was used to compare differences among groups, *, p < 0.01.

References

    1. Johnston SL. Overview of virus-induced airway disease. Proc Am Thorac Soc. 2005;2:150–156. doi: 10.1513/pats.200502-018AW.
    1. Proud D, Chow CW. Role of viral infections in asthma and chronic obstructive pulmonary disease. Am J Respir Cell Mol Biol. 2006;35:513–518. doi: 10.1165/rcmb.2006-0199TR.
    1. Arcavi L, Benowitz NL. Cigarette smoking and infection. Arch Intern Med. 2004;164:2206–2216. doi: 10.1001/archinte.164.20.2206.
    1. Aronson MD, Weiss ST, Ben RL, Komaroff AL. Association between cigarette smoking and acute respiratory tract illness in young adults. JAMA. 1982;248:181–183. doi: 10.1001/jama.1982.03330020025023.
    1. Bensenor IM, Cook NR, Lee IM, Chown MJ, Hennekens CH, Buring JE, Manson JE. Active and passive smoking and risk of colds in women. Ann Epidemiol. 2001;11:225–231. doi: 10.1016/S1047-2797(00)00214-3.
    1. Marcy TW, Merrill WW. Cigarette smoking and respiratory tract infection. Clin Chest Med. 1987;8:381–391.
    1. Sopori M. Effects of cigarette smoke on the immune system. Nat Rev Immunol. 2002;2:372–377. doi: 10.1038/nri803.
    1. Mehta H, Nazzal K, Sadikot RT. Cigarette smoking and innate immunity. Inflamm Res. 2008;57:497–503. doi: 10.1007/s00011-008-8078-6.
    1. Matsunaga K, Klein TW, Friedman H, Yamamoto Y. Involvement of nicotinic acetylcholine receptors in suppression of antimicrobial activity and cytokine responses of alveolar macrophages to Legionella pneumophila infection by nicotine. J Immunol. 2001;167:6518–6524.
    1. Ouyang Y, Virasch N, Hao P, Aubrey MT, Mukerjee N, Bierer BE, Freed BM. Suppression of human IL-1beta, IL-2, IFN-gamma, and TNF-alpha production by cigarette smoke extracts. J Allergy Clin Immunol. 2000;106:280–287. doi: 10.1067/mai.2000.107751.
    1. Wewers MD, Diaz PT, Wewers ME, Lowe MP, Nagaraja HN, Clanton TL. Cigarette smoking in HIV infection induces a suppressive inflammatory environment in the lung. Am J Respir Crit Care Med. 1998;158:1543–1549.
    1. Chen H, Cowan MJ, Hasday JD, Vogel SN, Medvedev AE. Tobacco smoking inhibits expression of proinflammatory cytokines and activation of IL-1R-associated kinase, p38, and NF-kappaB in alveolar macrophages stimulated with TLR2 and TLR4 agonists. J Immunol. 2007;179:6097–6106.
    1. Birrell MA, Wong S, Catley MC, Belvisi MG. Impact of tobacco-smoke on key signaling pathways in the innate immune response in lung macrophages. J Cell Physiol. 2008;214:27–37. doi: 10.1002/jcp.21158.
    1. Edwards K, Braun KM, Evans G, Sureka AO, Fan S. Mainstream and sidestream cigarette smoke condensates suppress macrophage responsiveness to interferon gamma. Hum Exp Toxicol. 1999;18:233–240. doi: 10.1191/096032799678839978.
    1. Matsumoto M, Funami K, Tanabe M, Oshiumi H, Shingai M, Seto Y, Yamamoto A, Seya T. Subcellular localization of Toll-like receptor 3 in human dendritic cells. J Immunol. 2003;171:3154–3162.
    1. Orinska Z, Bulanova E, Budagian V, Metz M, Maurer M, Bulfone-Paus S. TLR3-induced activation of mast cells modulates CD8+ T-cell recruitment. Blood. 2005;106:978–987. doi: 10.1182/blood-2004-07-2656.
    1. Meylan E, Tschopp J. Toll-like receptors and RNA helicases: two parallel ways to trigger antiviral responses. Mol Cell. 2006;22:561–569. doi: 10.1016/j.molcel.2006.05.012.
    1. Matsukura S, Kokubu F, Kurokawa M, Kawaguchi M, Ieki K, Kuga H, Odaka M, Suzuki S, Watanabe S, Homma T. Role of RIG-I, MDA-5, and PKR on the expression of inflammatory chemokines induced by synthetic dsRNA in airway epithelial cells. Int Arch Allergy Immunol. 2007;143(Suppl 1):80–83.
    1. Hui KP, Lee SM, Cheung CY, Ng IH, Poon LL, Guan Y, Ip NY, Lau AS, Peiris JS. Induction of proinflammatory cytokines in primary human macrophages by influenza A virus (H5N1) is selectively regulated by IFN regulatory factor 3 and p38 MAPK. J Immunol. 2009;182:1088–1098.
    1. Dhillon N, Zhu X, Peng F, Yao H, Williams R, Qiu J, Callen S, Ladner AO, Buch S. Molecular mechanism(s) involved in the synergistic induction of CXCL10 by human immunodeficiency virus type 1 Tat and interferon-gamma in macrophages. J Neurovirol. 2008;14:196–204. doi: 10.1080/13550280801993648.
    1. Livengood AJ, Wu CC, Carson DA. Opposing roles of RNA receptors TLR3 and RIG-I in the inflammatory response to double-stranded RNA in a Kaposi’s sarcoma cell line. Cell Immunol. 2007;249:55–62. doi: 10.1016/j.cellimm.2007.11.004.
    1. Michalec L, Choudhury BK, Postlethwait E, Wild JS, Alam R, Lett-Brown M, Sur S. CCL7 and CXCL10 orchestrate oxidative stress-induced neutrophilic lung inflammation. J Immunol. 2002;168:846–852.
    1. Agostini C, Calabrese F, Poletti V, Marcer G, Facco M, Miorin M, Cabrelle A, Baesso I, Zambello R, Trentin L, Semenzato G. CXCR3/CXCL10 interactions in the development of hypersensitivity pneumonitis. Respir Res. 2005;6:20. doi: 10.1186/1465-9921-6-20.
    1. Yuan J, Liu Z, Lim T, Zhang H, He J, Walker E, Shier C, Wang Y, Su Y, Sall A. CXCL10 inhibits viral replication through recruitment of natural killer cells in coxsackievirus B3-induced myocarditis. Circ Res. 2009;104:628–638. doi: 10.1161/CIRCRESAHA.108.192179.
    1. Cole AM, Ganz T, Liese AM, Burdick MD, Liu L, Strieter RM. Cutting edge: IFN-inducible ELR- CXC chemokines display defensin-like antimicrobial activity. J Immunol. 2001;167:623–627.
    1. Vasquez RE, Xin L, Soong L. Effects of CXCL10 on dendritic cell and CD4+ T-cell functions during Leishmania amazonensis infection. Infect Immun. 2008;76:161–169. doi: 10.1128/IAI.00825-07.
    1. Freeman CM, Curtis JL, Chensue SW. CC chemokine receptor 5 and CXC chemokine receptor 6 expression by lung CD8+ cells correlates with chronic obstructive pulmonary disease severity. Am J Pathol. 2007;171:767–776. doi: 10.2353/ajpath.2007.061177.
    1. Freeman CM, Han MK, Martinez FJ, Murray S, Liu LX, Chensue SW, Polak TJ, Sonstein J, Todt JC, Ames TM. Cytotoxic potential of lung CD8(+) T cells increases with chronic obstructive pulmonary disease severity and with in vitro stimulation by IL-18 or IL-15. J Immunol. 2010;184:6504–6513. doi: 10.4049/jimmunol.1000006.
    1. McCubbrey AL, Sonstein J, Ames TM, Freeman CM, Curtis JL. Glucocorticoids relieve collectin-driven suppression of apoptotic cell uptake in murine alveolar macrophages through downregulation of SIRPalpha. J Immunol. 2012;189:112–119. doi: 10.4049/jimmunol.1200984.
    1. Koarai A, Yanagisawa S, Sugiura H, Ichikawa T, Akamatsu K, Hirano T, Nakanishi M, Matsunaga K, Minakata Y, Ichinose M. Cigarette smoke augments the expression and responses of toll-like receptor 3 in human macrophages. Respirology. 2012;17:1018–1025. doi: 10.1111/j.1440-1843.2012.02198.x.
    1. Seemungal T, Harper-Owen R, Bhowmic A, Moric I, Sanderson G, Message S, MacCallum P, Meade T, Jeffries D, Johnston S, Wedzicha J. Respiratory viruses, symptoms, and inflammatory markers in acute exacerbations and stable chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2001;164:1618–1623.
    1. Papi A, Bellettato C, Braccioni F, Romagnoli M, Casolari P, Caramori G, Fabbri L, Johnston S. Infections and airway inflammation in chronic obstructive pulmonary disease severe exacerbations. Am J Respir Crit Care Med. 2006;173:1114–1121. doi: 10.1164/rccm.200506-859OC.
    1. Nicholson KG, Kent J, Ireland DC. Respiratory viruses and exacerbations of asthma in adults. BMJ. 1993;307:982–986. doi: 10.1136/bmj.307.6910.982.
    1. Johnston SL, Pattemore PK, Sanderson G, Smith S, Lampe F, Josephs L, Symington P, O’Toole S, Myint SH, Tyrrell DA. Community study of role of viral infections in exacerbations of asthma in 9–11 year old children. BMJ. 1995;310:1225–1229. doi: 10.1136/bmj.310.6989.1225.
    1. Johnston SL, Pattemore PK, Sanderson G, Smith S, Campbell MJ, Josephs LK, Cunningham A, Robinson BS, Myint SH, Ward ME. The relationship between upper respiratory infections and hospital admissions for asthma: a time-trend analysis. Am J Respir Crit Care Med. 1996;154:654–660.
    1. Kanner RE, Anthonisen NR, Connett JE. Lower respiratory illnesses promote FEV(1) decline in current smokers but not ex-smokers with mild chronic obstructive pulmonary disease: results from the lung health study. Am J Respir Crit Care Med. 2001;164:358–364.
    1. Thomson NC, Chaudhuri R. Asthma in smokers: challenges and opportunities. Curr Opin Pulm Med. 2009;15:39–45. doi: 10.1097/MCP.0b013e32831da894.
    1. Eisner MD, Iribarren C. The influence of cigarette smoking on adult asthma outcomes. Nicotine Tob Res. 2007;9:53–56. doi: 10.1080/14622200601078293.
    1. Sippel JM, Pedula KL, Vollmer WM, Buist AS, Osborne ML. Associations of smoking with hospital-based care and quality of life in patients with obstructive airway disease. Chest. 1999;115:691–696. doi: 10.1378/chest.115.3.691.
    1. Siroux V, Pin I, Oryszczyn MP, LeMoual N, Kauffmann F. Relationships of active smoking to asthma and asthma severity in the EGEA study. Epidemiological study on the Genetics and Environment of Asthma. Eur Respir J. 2000;15:470–477. doi: 10.1034/j.1399-3003.2000.15.08.x.
    1. Lazarus SC, Chinchilli VM, Rollings NJ, Boushey HA, Cherniack R, Craig TJ, Deykin A, DiMango E, Fish JE, Ford JG. Smoking affects response to inhaled corticosteroids or leukotriene receptor antagonists in asthma. Am J Respir Crit Care Med. 2007;175:783–790. doi: 10.1164/rccm.200511-1746OC.
    1. Kohlmeier JE, Woodland DL. Immunity to respiratory viruses. Annu Rev Immunol. 2009;27:61–82. doi: 10.1146/annurev.immunol.021908.132625.
    1. Khaitov MR, Laza-Stanca V, Edwards MR, Walton RP, Rohde G, Contoli M, Papi A, Stanciu LA, Kotenko SV, Johnston SL. Respiratory virus induction of alpha-, beta- and lambda-interferons in bronchial epithelial cells and peripheral blood mononuclear cells. Allergy. 2009;64:375–386. doi: 10.1111/j.1398-9995.2008.01826.x.
    1. Contoli M, Message SD, Laza-Stanca V, Edwards MR, Wark PA, Bartlett NW, Kebadze T, Mallia P, Stanciu LA, Parker HL. Role of deficient type III interferon-lambda production in asthma exacerbations. Nat Med. 2006;12:1023–1026. doi: 10.1038/nm1462.
    1. Cakebread JA, Xu Y, Grainge C, Kehagia V, Howarth PH, Holgate ST, Davies DE. Exogenous IFN-beta has antiviral and anti-inflammatory properties in primary bronchial epithelial cells from asthmatic subjects exposed to rhinovirus. J Allergy Clin Immunol. 2011;127:1148–1154. doi: 10.1016/j.jaci.2011.01.023. e1149.
    1. Laza-Stanca V, Stanciu LA, Message SD, Edwards MR, Gern JE, Johnston SL. Rhinovirus replication in human macrophages induces NF-kappaB-dependent tumor necrosis factor alpha production. J Virol. 2006;80:8248–8258. doi: 10.1128/JVI.00162-06.
    1. Wang J, Oberley-Deegan R, Wang S, Nikrad M, Funk CJ, Hartshorn KL, Mason RJ. Differentiated human alveolar type II cells secrete antiviral IL-29 (IFN-lambda 1) in response to influenza A infection. J Immunol. 2009;182:1296–1304.
    1. Siren J, Pirhonen J, Julkunen I, Matikainen S. IFN-alpha regulates TLR-dependent gene expression of IFN-alpha, IFN-beta, IL-28, and IL-29. J Immunol. 2005;174:1932–1937.
    1. Sheppard P, Kindsvogel W, Xu W, Henderson K, Schlutsmeyer S, Whitmore TE, Kuestner R, Garrigues U, Birks C, Roraback J. IL-28, IL-29 and their class II cytokine receptor IL-28R. Nat Immunol. 2003;4:63–68. doi: 10.1038/ni873.
    1. Kim HM, Lee YW, Lee KJ, Kim HS, Cho SW, Van Rooijen N, Guan Y, Seo SH. Alveolar macrophages are indispensable for controlling influenza viruses in lungs of pigs. J Virol. 2008;82:4265–4274. doi: 10.1128/JVI.02602-07.
    1. Kumagai Y, Takeuchi O, Kato H, Kumar H, Matsui K, Morii E, Aozasa K, Kawai T, Akira S. Alveolar macrophages are the primary interferon-alpha producer in pulmonary infection with RNA viruses. Immunity. 2007;27:240–252. doi: 10.1016/j.immuni.2007.07.013.
    1. Pribul PK, Harker J, Wang B, Wang H, Tregoning JS, Schwarze J, Openshaw PJ. Alveolar macrophages are a major determinant of early responses to viral lung infection but do not influence subsequent disease development. J Virol. 2008;82:4441–4448. doi: 10.1128/JVI.02541-07.
    1. Woodruff PG, Koth LL, Yang YH, Rodriguez MW, Favoreto S, Dolganov GM, Paquet AC, Erle DJ. A distinctive alveolar macrophage activation state induced by cigarette smoking. Am J Respir Crit Care Med. 2005;172:1383–1392. doi: 10.1164/rccm.200505-686OC.
    1. Shaykhiev R, Krause A, Salit J, Strulovici-Barel Y, Harvey BG, O’Connor TP, Crystal RG. Smoking-dependent reprogramming of alveolar macrophage polarization: implication for pathogenesis of chronic obstructive pulmonary disease. J Immunol. 2009;183:2867–2883. doi: 10.4049/jimmunol.0900473.
    1. Hunninghake GW, Gadek JE, Kawanami O, Ferrans VJ, Crysta l RG. Inflammatory and immune processes in the human lung in health and disease. Evaluation by bronchoalveolar lavage. Am J Pathol. 1979;97:149–206.
    1. Richards SW, Peterson PK, Verbrugh HA, Nelson RD, Hammerschmidt DE, Hoidal JR. Chemotactic and phagocytic responses of human alveolar macrophages to activated complement components. Infect Immun. 1984;43:775–778.
    1. Holt PG. Immune and inflammatory function in cigarette smokers. Thorax. 1987;42:241–249. doi: 10.1136/thx.42.4.241.
    1. Marti-Lliteras P, Regueiro V, Morey P, Hood DW, Saus C, Sauleda J, Agusti AG, Bengoechea JA, Garmendia J. Nontypeable Haemophilus influenzae clearance by alveolar macrophages is impaired by exposure to cigarette smoke. Infect Immun. 2009;77:4232–4242. doi: 10.1128/IAI.00305-09.
    1. King TE Jr, Savici D, Campbell PA. Phagocytosis and killing of Listeria monocytogenes by alveolar macrophages: smokers versus nonsmokers. J Infect Dis. 1988;158:1309–1316. doi: 10.1093/infdis/158.6.1309.
    1. Cosio BG, Tsaprouni L, Ito K, Jazrawi E, Adcock IM, Barnes PJ. Theophylline restores histone deacetylase activity and steroid responses in COPD macrophages. J Exp Med. 2004;200:689–695. doi: 10.1084/jem.20040416.
    1. Cosio MG, Saetta M, Agusti A. Immunologic aspects of chronic obstructive pulmonary disease. N Engl J Med. 2009;360:2445–2454. doi: 10.1056/NEJMra0804752.
    1. McCrea KA, Ensor JE, Nall K, Bleecker ER, Hasday JD. Altered cytokine regulation in the lungs of cigarette smokers. Am J Respir Crit Care Med. 1994;150:696–703.
    1. Morrison D, Strieter RM, Donnelly SC, Burdick MD, Kunkel SL, MacNee W. Neutrophil chemokines in bronchoalveolar lavage fluid and leukocyte-conditioned medium from nonsmokers and smokers. Eur Respir J. 1998;12:1067–1072. doi: 10.1183/09031936.98.12051067.
    1. Ohta T, Yamashita N, Maruyama M, Sugiyama E, Kobayashi M. Cigarette smoking decreases interleukin-8 secretion by human alveolar macrophages. Respir Med. 1998;92:922–927. doi: 10.1016/S0954-6111(98)90191-3.
    1. Hutchens M, Luker KE, Sottile P, Sonstein J, Lukacs NW, Nunez G, Curtis JL, Luker GD. TLR3 increases disease morbidity and mortality from vaccinia infection. J Immunol. 2008;180:483–491.
    1. Bunting RA, Duffy KE, Lamb RJ, San Mateo LR, Smalley K, Raymond H, Liu X, Petley T, Fisher J, Beck H. Novel antagonist antibody to TLR3 blocks poly(I:C)-induced inflammation in vivo and in vitro. Cell Immunol. 2011;267:9–16. doi: 10.1016/j.cellimm.2010.10.008.
    1. Cavassani KA, Ishii M, Wen H, Schaller MA, Lincoln PM, Lukacs NW, Hogaboam CM, Kunkel SL. TLR3 is an endogenous sensor of tissue necrosis during acute inflammatory events. J Exp Med. 2008;205:2609–2621. doi: 10.1084/jem.20081370.
    1. Henson PM, Tuder RM. Apoptosis in the lung: induction, clearance and detection. Am J Physiol Lung Cell Mol Physiol. 2008;294:L601–L611. doi: 10.1152/ajplung.00320.2007.
    1. Hodge S, Hodge G, Brozyna S, Jersmann H, Holmes M, Reynolds PN. Azithromycin increases phagocytosis of apoptotic bronchial epithelial cells by alveolar macrophages. Eur Respir J. 2006;28:486–495. doi: 10.1183/09031936.06.00001506.
    1. Hodge S, Hodge G, Scicchitano R, Reynolds PN, Holmes M. Alveolar macrophages from subjects with chronic obstructive pulmonary disease are deficient in their ability to phagocytose apoptotic airway epithelial cells. Immunol Cell Biol. 2003;81:289–296. doi: 10.1046/j.1440-1711.2003.t01-1-01170.x.
    1. Richens TR, Linderman DJ, Horstmann SA, Lambert C, Xiao YQ, Keith RL, Boe DM, Morimoto K, Bowler RP, Day BJ. Cigarette smoke impairs clearance of apoptotic cells through oxidant-dependent activation of RhoA. Am J Respir Crit Care Med. 2009;179:1011–1021. doi: 10.1164/rccm.200807-1148OC.
    1. Vandivier RW, Henson PM, Douglas IS. Burying the dead: the impact of failed apoptotic cell removal (efferocytosis) on chronic inflammatory lung disease. Chest. 2006;129:1673–1682. doi: 10.1378/chest.129.6.1673.
    1. Hodge S, Hodge G, Ahern J, Jersmann H, Holmes M, Reynolds PN. Smoking alters alveolar macrophage recognition and phagocytic ability: implications in chronic obstructive pulmonary disease. Am J Respir Cell Mol Biol. 2007;37:748–755. doi: 10.1165/rcmb.2007-0025OC.
    1. Drömann D, Goldmann T, Tiedje T, Zabel P, Dalhoff K, Schaaf B. Toll-like receptor 2 expression is decreased on alveolar macrophages in cigarette smokers and COPD patients. Respir Res. 2005;6:68. doi: 10.1186/1465-9921-6-68.
    1. Doyle I, Ratcliffe M, Walding A, Vanden Bon E, Dymond M, Tomlinson W, Tilley D, Shelton P, Dougall I. Differential gene expression analysis in human monocyte-derived macrophages: impact of cigarette smoke on host defence. Mol Immunol. 2010;47:1058–1065. doi: 10.1016/j.molimm.2009.11.008.
    1. Gaschler GJ, Zavitz CC, Bauer CM, Skrtic M, Lindahl M, Robbins CS, Chen B, Stampfli MR. Cigarette smoke exposure attenuates cytokine production by mouse alveolar macrophages. Am J Respir Cell Mol Biol. 2008;38:218–226.
    1. Kang MJ, Lee CG, Lee JY, Dela Cruz CS, Chen ZJ, Enelow R, Elias JA. Cigarette smoke selectively enhances viral PAMP- and virus-induced pulmonary innate immune and remodeling responses in mice. J Clin Invest. 2008;118:2771–2784.
    1. Bonecchi R, Galliera E, Borroni EM, Corsi MM, Locati M, Mantovani A. Chemokines and chemokine receptors: an overview. Front Biosci. 2009;14:540–551.
    1. Jiang D, Liang J, Campanella GS, Guo R, Yu S, Xie T, Liu N, Jung Y, Homer R, Meltzer EB. Inhibition of pulmonary fibrosis in mice by CXCL10 requires glycosaminoglycan binding and syndecan-4. J Clin Invest. 2010;120:2049–2057. doi: 10.1172/JCI38644.
    1. Pekarek V, Srinivas S, Eskdale J, Gallagher G. Interferon lambda-1 (IFN-lambda1/IL-29) induces ELR(−) CXC chemokine mRNA in human peripheral blood mononuclear cells, in an IFN-gamma-independent manner. Genes Immun. 2007;8:177–180. doi: 10.1038/sj.gene.6364372.
    1. Korpi-Steiner NL, Bates ME, Lee WM, Hall DJ, Bertics PJ. Human rhinovirus induces robust IP-10 release by monocytic cells, which is independent of viral replication but linked to type I interferon receptor ligation and STAT1 activation. J Leukoc Biol. 2006;80:1364–1374. doi: 10.1189/jlb.0606412.
    1. Miller AL, Bowlin TL, Lukacs NW. Respiratory syncytial virus-induced chemokine production: linking viral replication to chemokine production in vitro and in vivo. J Infect Dis. 2004;189:1419–1430. doi: 10.1086/382958.
    1. Proost P, Vynckier AK, Mahieu F, Put W, Grillet B, Struyf S, Wuyts A, Opdenakker G, Van Damme J. Microbial Toll-like receptor ligands differentially regulate CXCL10/IP-10 expression in fibroblasts and mononuclear leukocytes in synergy with IFN-gamma and provide a mechanism for enhanced synovial chemokine levels in septic arthritis. Eur J Immunol. 2003;33:3146–3153. doi: 10.1002/eji.200324136.
    1. D’Ambrosio D, Mariani M, Panina-Bordignon P, Sinigaglia F. Chemokines and their receptors guiding T lymphocyte recruitment in lung inflammation. Am J Respir Crit Care Med. 2001;164:1266–1275.
    1. Kohlmeier JE, Cookenham T, Miller SC, Roberts AD, Christensen JP, Thomsen AR, Woodland DL. CXCR3 directs antigen-specific effector CD4+ T cell migration to the lung during parainfluenza virus infection. J Immunol. 2009;183:4378–4384. doi: 10.4049/jimmunol.0902022.
    1. Costa C, Rufino R, Traves SL, Lapa ESJR, Barnes PJ, Donnelly LE. CXCR3 and CCR5 chemokines in induced sputum from patients with COPD. Chest. 2008;133:26–33. doi: 10.1378/chest.07-0393.
    1. Han MK, Postma D, Mannino DM, Giardino ND, Buist S, Curtis JL, Martinez FJ. Gender and chronic obstructive pulmonary disease: why it matters. Am J Respir Crit Care Med. 2007;176:1179–1184. doi: 10.1164/rccm.200704-553CC.
    1. Antonelli A, Rotondi M, Fallahi P, Ferrari SM, Paolicchi A, Romagnani P, Serio M, Ferrannini E. Increase of CXC chemokine CXCL10 and CC chemokine CCL2 serum levels in normal ageing. Cytokine. 2006;34:32–38. doi: 10.1016/j.cyto.2006.03.012.
    1. Antonelli A, Rotondi M, Fallahi P, Romagnani P, Ferrari SM, Ferrannini E, Serio M. Age-dependent changes in CXC chemokine ligand 10 serum levels in euthyroid subjects. J Interferon Cytokine Res. 2005;25:547–552. doi: 10.1089/jir.2005.25.547.

Source: PubMed

3
Prenumerera