Cigarette smoke enhances Th-2 driven airway inflammation and delays inhalational tolerance

Chris L Van Hove, Katrien Moerloose, Tania Maes, Guy F Joos, Kurt G Tournoy, Chris L Van Hove, Katrien Moerloose, Tania Maes, Guy F Joos, Kurt G Tournoy

Abstract

Background: Active smoking increases asthma severity and is related to diminished treatment efficacy. Animal models in which inhalation of both allergen and mainstream cigarette smoke are combined can help us to understand the complex interaction between both agents. We have recently shown that, in allergic mice, the airway inflammation can be cleared by repeated allergen challenge, resulting in the establishment of a state of inhalational tolerance.

Methods: In this study, we assessed in vivo the impact of cigarette smoke on the efficacy and time course of this form of tolerance induction. We exposed sensitized mice to concurrent mainstream cigarette smoke and allergen (Ovalbumin- OVA) and measured the airway inflammation at different time points.

Results: We first confirmed that aerosolized OVA administered for a prolonged time period (4-8 weeks) resulted in the establishment of tolerance. Concurrent OVA and smoke exposure for 2 weeks showed that tobacco smoke enhanced the Th-2 driven airway inflammation in the acute phase. In addition, the induction of the tolerance by repeated inhalational OVA challenge was delayed significantly by the tobacco smoke, since 4 weeks of concurrent exposure resulted in a more persistent eosinophilic airway inflammation, paralleled by a more mature dendritic cell phenotype. However, smoke exposure could not prevent the establishment of tolerance after 8 weeks of antigen exposure as shown by both histopathology (disappearance of the Th-2 driven inflammation) and by in vivo functional experiments. In these tolerized mice, some of the inflammatory responses to the smoke were even attenuated.

Conclusion: Cigarette smoke enhances acute allergic inflammation and delays, but does not abrogate the development of tolerance due to prolonged challenge with inhaled antigen in experimental asthma.

Figures

Figure 1
Figure 1
Cigarette smoke and allergen exposure protocols. Allergen exposure protocols (Allergen Protocols): Allergen Protocol 1A: Exposure of sensitized C57BL/6 mice to PBS or OVA aerosols for 2 weeks. (n = 8 mice/group). Allergen Protocol 1B: Prolonged PBS or OVA aerosol exposure in sensitized mice (8 weeks). (n = 8 mice/group). Allergen Protocol 2: OVA sensitized mice were exposed to OVA for 8 weeks ("chronic"), followed by HEL/alum immunisation and short-term ("acute") HEL aerosol challenge (OVA/HEL group). Control mice were exposed to PBS for 8 weeks, immunized with HEL/alum and challenged with either PBS (PBS/PBS group) or with HEL (PBS/HEL group). (n = 8–12 mice/group). Smoke exposure protocol (Smoke Protocol): C57BL/6 mice are exposed to Smoke or Air for 8 weeks. (n = 6 mice/group). Combined protocols (Combination Protocols): Combination Protocol 1: similar to Allergen Protocol 1B, combined with Smoke exposures or Air. (n = 8 mice/group). Combination Protocol 2: similar to Allergen Protocol 2, combined with or without Smoke. (n = 8 mice/group). Combination Protocol 3A: similar to Allergen Protocol 1A, combined with Smoke exposures or Air. (n = 8 mice/group). Combination Protocol 3B: 4 weeks of OVA aerosol or PBS exposures in sensitized mice, combined with Smoke exposures or Air. (n = 8 mice/group). Vertical arrows = aerosol challenges (OVA, HEL or PBS). Stripped horizontal bars = period of Smoke or Air exposures.
Figure 2
Figure 2
BALF cell differentiation (A) and BALF cytokines (pg/ml) MCP-1 and KC (B) after 8 week exposure to Smoke or Air in C57BL/6 mice (Smoke Protocol). n = 6 mice/group. * p ≤ 0.05: Smoke vs Air (Mann-Whitney U test); Bars indicate mean ± SEM. ND = Not Detectable (detection limit = 1–2 pg/ml).
Figure 3
Figure 3
Histology (Congo Red) of the airways of the four groups of mice after 8 week exposure to OVA or PBS aerosols combined with Smoke or Air in sensitized C57BL/6 mice (Combination Protocol 1) (Magnification = 400×; A = PBS/Air, B = PBS/Smoke, C = OVA/Air, D = OVA/Smoke).
Figure 4
Figure 4
Results obtained from combining 8 weeks OVA or PBS aerosol exposure with Smoke or Air in sensitized C57BL/6 mice (Combination Protocol 1). (A.) BALF analysis of total cell counts and differentiation after prolonged exposure (8 weeks) to a combination of OVA or PBS aerosols and Smoke or Air. (B) Evaluation of the peribronchial inflammation via a semi-quantitative method (Mean score/airway). (C) Percentage of T – lymphocytes in lung that are CD4+CD69+ (as a percentage within the T cell population). (D) Percentage of dendritic cells in digested lung tissue (percentage of total lung cells). (E) Th-2 cytokine (IL-13 and TARC) profiles in BALF (pg/ml). (F) Cytokines MCP-1 and KC in BALF (pg/ml). n = 8 mice/group; bars indicate mean ± SEM. * p ≤ 0.05: all groups vs PBS/Air (Mann-Whitney U test); + p ≤ 0.05: PBS/Smoke vs OVA/Smoke (Mann-Whitney U test); # p ≤ 0.05: OVA/Smoke vs OVA/Air (Mann-Whitney U test); ND = Not Detectable (detection limit = 1–2 pg/ml)
Figure 5
Figure 5
Results obtained 8 weeks of OVA aerosols followed by re-sensitisation (i. p.) and re-challenge to HEL aerosols (Allergen Protocol 2) combined with concurrent Smoke or Air-exposures (Combination Protocol 2). (A) BALF cell differentiation. (B) Peribronchial inflammation (Mean score/airway). n = 8 mice/group; bars indicate mean ± SEM. * p ≤ 0.05: OVA/HEL/Smoke vs OVA/HEL/Air (Mann-Whitney U test);
Figure 6
Figure 6
BALF cell differentiation and cytokines (pg/ml) IL-13 and TARC after respectively 2 (Fig. 6A and 6B) and 4 (Fig. 6C and 6D) weeks of concurrent exposure to OVA/PBS and Smoke or Air in C57BL/6 mice (Combination Protocol 3A and 3B). n = 8 mice/group; bars indicate mean ± SEM. * p ≤ 0.05: all groups vs PBS/Air (Mann-Whitney U test); + p ≤ 0.05: OVA/Smoke vs. PBS/Smoke (Mann-Whitney U test); # p ≤ 0.05: OVA/Smoke vs. OVA/Air (Mann-Whitney U test); ND = Not Detectable (detection limit = 1–2 pg/ml)
Figure 7
Figure 7
The evolution in the levels of TARC in BALF (pg/ml) and the percentage of eosinophils in BALF over time after 2, 4 and 8 weeks of concurrent exposure to OVA/PBS and to Smoke or Air in C57BL/6 mice (Combination Protocol 1, 3A and 3B). # p ≤ 0.05: OVA/Smoke vs. OVA/Air (Mann-Whitney U test);

References

    1. O'Garra A, Vieira P. Regulatory T cells and mechanisms of immune system control. Nat Med. 2004;10:801–805. doi: 10.1038/nm0804-801.
    1. Jutel M, Akdis M, Budak F, Aebischer-Casaulta C, Wrzyszcz M, Blaser K, Akdis CA. IL-10 and TGF-beta cooperate in the regulatory T cell response to mucosal allergens in normal immunity and specific immunotherapy. Eur J Immunol. 2003;33:1205–1214. doi: 10.1002/eji.200322919.
    1. Van Hove CL, Maes T, Joos GF, Tournoy KG. Prolonged Inhaled Allergen Exposure Can Induce Persistent Tolerance. Am J Respir Cell Mol Biol. 2007;36:573–584. doi: 10.1165/rcmb.2006-0385OC.
    1. Swirski FK, Sajic D, Robbins CS, Gajewska BU, Jordana M, Stampfli MR. Chronic exposure to innocuous antigen in sensitized mice leads to suppressed airway eosinophilia that is reversed by granulocyte macrophage colony-stimulating factor. J Immunol. 2002;169:3499–3506.
    1. Swirski FK, D'Sa A, Kianpour S, Inman MD, Stampfli MR. Prolonged ovalbumin exposure attenuates airway hyperresponsiveness and T cell function in mice. Int Arch Allergy Immunol. 2006;141:130–140. doi: 10.1159/000094715.
    1. Schramm CM, Puddington L, Wu C, Guernsey L, Gharaee-Kermani M, Phan SH, Thrall RS. Chronic inhaled ovalbumin exposure induces antigen-dependent but not antigen-specific inhalational tolerance in a murine model of allergic airway disease. Am J Pathol. 2004;164:295–304.
    1. Johnson JR, Wiley RE, Fattouh R, Swirski FK, Gajewska BU, Coyle AJ, Gutierrez-Ramos JC, Ellis R, Inman MD, Jordana M. Continuous exposure to house dust mite elicits chronic airway inflammation and structural remodeling. Am J Respir Crit Care Med. 2004;169:378–385. doi: 10.1164/rccm.200308-1094OC.
    1. Wegmann M, Fehrenbach H, Fehrenbach A, Held T, Schramm C, Garn H, Renz H. Involvement of distal airways in a chronic model of experimental asthma. Clin Exp Allergy. 2005;35:1263–1271. doi: 10.1111/j.1365-2222.2005.02306.x.
    1. Temelkovski J, Hogan SP, Shepherd DP, Foster PS, Kumar RK. An improved murine model of asthma: selective airway inflammation, epithelial lesions and increased methacholine responsiveness following chronic exposure to aerosolised allergen. Thorax. 1998;53:849–856.
    1. Siegle JS, Hansbro N, Herbert C, Yang M, Foster PS, Kumar RK. Airway hyperreactivity in exacerbation of chronic asthma is independent of eosinophilic inflammation. Am J Respir Cell Mol Biol. 2006;35:565–570. doi: 10.1165/rcmb.2006-0135OC.
    1. D'Amato G, Liccardi G, D'Amato M, Holgate S. Environmental risk factors and allergic bronchial asthma. Clin Exp Allergy. 2005;35:1113–1124. doi: 10.1111/j.1365-2222.2005.02328.x.
    1. Eder W, Ege MJ, von Mutius E. The asthma epidemic. N Engl J Med. 2006;355:2226–2235. doi: 10.1056/NEJMra054308.
    1. Moerloose KB, Robays LJ, Maes T, Brusselle GG, Tournoy KG, Joos GF. Cigarette smoke exposure facilitates allergic sensitization in mice. Respir Res. 2006;7:49. doi: 10.1186/1465-9921-7-49.
    1. Robbins CS, Pouladi MA, Fattouh R, Dawe DE, Vujicic N, Richards CD, Jordana M, Inman MD, Stampfli MR. Mainstream cigarette smoke exposure attenuates airway immune inflammatory responses to surrogate and common environmental allergens in mice, despite evidence of increased systemic sensitization. J Immunol. 2005;175:2834–2842.
    1. Rumold R, Jyrala M, az-Sanchez D. Secondhand smoke induces allergic sensitization in mice. J Immunol. 2001;167:4765–4770.
    1. Seymour BW, Pinkerton KE, Friebertshauser KE, Coffman RL, Gershwin LJ. Second-hand smoke is an adjuvant for T helper-2 responses in a murine model of allergy. J Immunol. 1997;159:6169–6175.
    1. Moerloose KB, Pauwels RA, Joos GF. Short-term cigarette smoke exposure enhances allergic airway inflammation in mice. Am J Respir Crit Care Med. 2005;172(2):168–172. doi: 10.1164/rccm.200409-1174OC.
    1. Melgert BN, Postma DS, Geerlings M, Luinge MA, Klok PA, van der Strate BW, Kerstjens HA, Timens W, Hylkema MN. Short-term smoke exposure attenuates ovalbumin-induced airway inflammation in allergic mice. Am J Respir Cell Mol Biol. 2004;30:880–885. doi: 10.1165/rcmb.2003-0178OC.
    1. Lazarus SC, Chinchilli VM, Rollings NJ, Boushey HA, Cherniack R, Craig TJ, Deykin A, Dimango E, Fish JE, Ford JG, Israel E, Kiley J, Kraft M, Lemanske RF, Jr., Leone FT, Martin RJ, Pesola GR, Peters SP, Sorkness CA, Szefler SJ, Wechsler ME, Fahy JV. 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. D'hulst AI, Vermaelen KY, Brusselle GG, Joos GF, Pauwels RA. Time course of cigarette smoke-induced pulmonary inflammation in mice. Eur Respir J. 2005;26:204–213. doi: 10.1183/09031936.05.00095204.
    1. Curtis JL, Byrd PK, Warnock ML, Kaltreider HB. Requirement of CD4-positive T cells for cellular recruitment to the lungs of mice in response to a particulate intratracheal antigen. J Clin Invest. 1991;88:1244–1254. doi: 10.1172/JCI115428.
    1. Ostroukhova M, Seguin-Devaux C, Oriss TB, Dixon-McCarthy B, Yang L, Ameredes BT, Corcoran TE, Ray A. Tolerance induced by inhaled antigen involves CD4(+) T cells expressing membrane-bound TGF-beta and FOXP3. J Clin Invest. 2004;114:28–38.
    1. Mucida D, Kutchukhidze N, Erazo A, Russo M, Lafaille JJ, Curotto de Lafaille MA. Oral tolerance in the absence of naturally occurring Tregs. J Clin Invest. 2005:1923–1933. doi: 10.1172/JCI24487.
    1. Melgert BN, Timens W, Kerstjens HA, Geerlings M, Luinge MA, Schouten JP, Postma DS, Hylkema MN. Effects of 4 months of smoking in mice with ovalbumin-induced airway inflammation. Clin Exp Allergy. 2007;37(12):1798–1808. doi: 10.1111/j.1365-2222.2007.02843.x.
    1. Cottrez F, Hurst SD, Coffman RL, Groux H. T regulatory cells 1 inhibit a Th2-specific response in vivo. J Immunol. 2000;165:4848–4853.
    1. Lewkowich IP, Herman NS, Schleifer KW, Dance MP, Chen BL, Dienger KM, Sproles AA, Shah JS, Kohl J, Belkaid Y, Wills-Karp M. CD4+CD25+ T cells protect against experimentally induced asthma and alter pulmonary dendritic cell phenotype and function. J Exp Med. 2005;202:1549–1561. doi: 10.1084/jem.20051506.
    1. Niu N, Le Goff MK, Li F, Rahman M, Homer RJ, Cohn L. A novel pathway that regulates inflammatory disease in the respiratory tract. J Immunol. 2007;178:3846–3855.
    1. Kohl J, Baelder R, Lewkowich IP, Pandey MK, Hawlisch H, Wang L, Best J, Herman NS, Sproles AA, Zwirner J, Whitsett JA, Gerard C, Sfyroera G, Lambris JD, Wills-Karp M. A regulatory role for the C5a anaphylatoxin in type 2 immunity in asthma. J Clin Invest. 2006;116:783–796. doi: 10.1172/JCI26582.
    1. Drouin SM, Sinha M, Sfyroera G, Lambris JD, Wetsel RA. A protective role for the fifth complement component (c5) in allergic airway disease. Am J Respir Crit Care Med. 2006;173:852–857. doi: 10.1164/rccm.200503-334OC.
    1. McKinley L, Kim J, Bolgos GL, Siddiqui J, Remick DG. Allergens induce enhanced bronchoconstriction and leukotriene production in C5 deficient mice. Respir Res. 2006;7:129. doi: 10.1186/1465-9921-7-129.
    1. Hamelmann E, Tadeda K, Oshiba A, Gelfand EW. Role of IgE in the development of allergic airway inflammation and airway hyperresponsiveness--a murine model. Allergy. 1999;54:297–305. doi: 10.1034/j.1398-9995.1999.00085.x.
    1. Nouri-Shirazi M, Tinajero R, Guinet E. Nicotine alters the biological activities of developing mouse bone marrow-derived dendritic cells (DCs) Immunol Lett. 2007;109:155–164. doi: 10.1016/j.imlet.2007.02.005.
    1. Nouri-Shirazi M, Guinet E. A possible mechanism linking cigarette smoke to higher incidence of respiratory infection and asthma. Immunol Lett. 2006;103:167–176. doi: 10.1016/j.imlet.2005.10.024.
    1. Holgate ST. The epithelium takes centre stage in asthma and atopic dermatitis. Trends Immunol. 2007;28:248–251. doi: 10.1016/j.it.2007.04.007.
    1. Zhou B, Comeau MR, De ST, Liggitt HD, Dahl ME, Lewis DB, Gyarmati D, Aye T, Campbell DJ, Ziegler SF. Thymic stromal lymphopoietin as a key initiator of allergic airway inflammation in mice. Nat Immunol. 2005;6:1047–1053. doi: 10.1038/ni1247.
    1. Swirski FK, Gajewska BU, Robbins CS, D'Sa A, Johnson JR, Pouladi MA, Inman MD, Stampfli MR. Concomitant airway expression of granulocyte-macrophage colony-stimulating factor and decorin, a natural inhibitor of transforming growth factor-beta, breaks established inhalation tolerance. Eur J Immunol. 2004;34:2375–2386. doi: 10.1002/eji.200425012.
    1. Bratke K, Klug M, Bier A, Julius P, Kuepper M, Virchow JC, Lommatzsch M. Function-associated Surface Molecules on Airway Dendritic Cells in Cigarette Smokers. Am J Respir Cell Mol Biol. 2008;175(8):783–790.
    1. Robbins CS, Dawe DE, Goncharova SI, Pouladi MA, Drannik AG, Swirski FK, Cox G, Stampfli MR. Cigarette smoke decreases pulmonary dendritic cells and impacts antiviral immune responsiveness. Am J Respir Cell Mol Biol. 2004;30:202–211. doi: 10.1165/rcmb.2003-0259OC.
    1. Vassallo R, Tamada K, Lau JS, Kroening PR, Chen L. Cigarette smoke extract suppresses human dendritic cell function leading to preferential induction of th-2 priming. J Immunol. 2005;175:2684–2691.
    1. Tsoumakidou M, Elston W, Zhu J, Wang Z, Gamble E, Siafakas NM, Barnes NC, Jeffery PK. Cigarette Smoking Alters Bronchial Mucosal Immunity in Asthma. Am J Respir Crit Care Med. 2007;175(9):919–925. doi: 10.1164/rccm.200607-908OC.
    1. Alexis NE, Lay JC, Almond M, Peden DB. Inhalation of low-dose endotoxin favors local T(H)2 response and primes airway phagocytes in vivo. J Allergy Clin Immunol. 2004;114:1325–1331. doi: 10.1016/j.jaci.2004.09.002.
    1. Alexis NE, Lay JC, Almond M, Bromberg PA, Patel DD, Peden DB. Acute LPS inhalation in healthy volunteers induces dendritic cell maturation in vivo. J Allergy Clin Immunol. 2005;115:345–350. doi: 10.1016/j.jaci.2004.11.040.
    1. Braun-Fahrlander C, Riedler J, Herz U, Eder W, Waser M, Grize L, Maisch S, Carr D, Gerlach F, Bufe A, Lauener RP, Schierl R, Renz H, Nowak D, von Mutius E. Environmental exposure to endotoxin and its relation to asthma in school-age children. N Engl J Med. 2002;347:869–877. doi: 10.1056/NEJMoa020057.
    1. Kim YK, Oh SY, Jeon SG, Park HW, Lee SY, Chun EY, Bang B, Lee HS, Oh MH, Kim YS, Kim JH, Gho YS, Cho SH, Min KU, Kim YY, Zhu Z. Airway exposure levels of lipopolysaccharide determine type 1 versus type 2 experimental asthma. J Immunol. 2007;178:5375–5382.
    1. Lundy SK, Berlin AA, Lukacs NW. Interleukin-12-independent down-modulation of cockroach antigen-induced asthma in mice by intranasal exposure to bacterial lipopolysaccharide. Am J Pathol. 2003;163:1961–1968.
    1. Holt PG, Strickland DH, Wikstrom ME, Jahnsen FL. Regulation of immunological homeostasis in the respiratory tract. Nat Rev Immunol. 2008;8:142–152. doi: 10.1038/nri2236.

Source: PubMed

3
購読する