Allergy Asthma Immunol Res.  2014 May;6(3):201-207. 10.4168/aair.2014.6.3.201.

Bacillus Calmette-Guerin Suppresses Asthmatic Responses via CD4+CD25+ Regulatory T Cells and Dendritic Cells

Affiliations
  • 1Asan Institute for Life Sciences, University of Ulsan College of Medicine, Seoul, Korea.
  • 2Department of Pediatrics, Childhood Asthma Atopy Center, Research Center for Standardization of Allergic Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea. sjhong@amc.seoul.kr
  • 3Department of Internal Medicine, Research Center for Pulmonary Disorders, Chonbuk National University Medical School, Jeonju, Korea. leeyc@chonbuk.ac.kr

Abstract

PURPOSE
Bacillus Calmette-Guerin (BCG) is known to suppress the asthmatic responses in a murine model of asthma and to induce dendritic cells (DCs) maturation. Mature DCs play a crucial role in the differentiation of regulatory T cells (Tregs), which are known to regulate allergic inflammatory responses. To investigate whether BCG regulates Tregs in a DCs-mediated manner, we analyzed in a murine model of asthma.
METHODS
BALB/c mice were injected intraperitoneally with BCG or intravenously with BCG-stimulated DCs and then sensitized and challenged with ovalbumin (OVA). Mice were analysed for bronchial hyperresponsiveness (BHR), the influx of inflammatory cells in the bronchoalveolar lavage (BAL) fluid, and histopathological changes in the lung. To identify the mechanisms, IgE, IgG1 and IgG2a in the serum were analysed and the CD25+ Tregs in the mice were depleted with anti-CD25 monoclonal antibody (mAb).
RESULTS
BCG and the transfer of BCG-stimulated DCs both suppressed all aspects of the asthmatic responses, namely, BHR, the production of total IgE and OVA-specific IgE and IgGs, and pulmonary eosinophilic inflammation. Anti-CD25mAb treatment reversed these effects.
CONCLUSIONS
BCG can attenuate the allergic inflammation in a mouse model of asthma by a Tregs-related mechanism that is mediated by DCs.

Keyword

BCG; dendritic cells; T-lymphocytes, regulatory; asthma; allergic inflammation; mouse

MeSH Terms

Animals
Asthma
Bacillus*
Bronchoalveolar Lavage
Dendritic Cells*
Eosinophils
Immunoglobulin E
Immunoglobulin G
Inflammation
Lung
Mice
Mycobacterium bovis
Ovalbumin
T-Lymphocytes, Regulatory*
Immunoglobulin E
Immunoglobulin G
Ovalbumin

Figure

  • Fig. 1 Effect of treatment with Bacillus Calmette-Guerin (BCG) vaccine-stimulated DCs on the ovalbumin (OVA)-induced allergic asthma of mouse model. OVA: mice sensitized with intraperitoneal injections of OVA on days 0 and 7 and then challenged with aerosolized OVA on days 14-16; Saline: mice sensitized and challenged with saline following the same time schedule described for OVA mice; BGC-DC: mice that were injected intravenously with BCG vaccine-stimulated DCs on day -5 and then sensitized and challenged with OVA; Non-DC: mice that were injected intravenously with unstimulated DCs on day -5 and then sensitized and challenged with OVA; BCG: mice injected intraperitoneally with BCG vaccine emulsified in alum and then sensitized and challenged with OVA (A) Brochial hyperresponsiveness (BHR), *P<0.05, **P<0.01 compared to the OVA mice; †P<0.01 compared to the BCG-DC-treated mice. (B) Differential cell counts in the Bronchoalvoelar lavage (BAL) fluid. (C) Lung pathology. (D) Peribronchial and perivascular lung inflammation scores. The values are the means±SD of the results from 5 mice per group. *P<0.05, **P<0.01.

  • Fig. 2 Effect of treatment with BCG vaccine-stimulated DCs on the serum levels of immunoglobulins in mouse model of allergic asthma. The plot legends are as described in Fig. 1. BCG vaccine-stimulated DC transfer inhibited total IgE, OVA-specific IgE and OVA- specific IgG1 levels compared to OVA mice. The values are the means±SD of the results from 5 mice per group. *P<0.05, **P<0.01.

  • Fig. 3 Effect of anti-CD25 monoclonal antibody (mAb) treatment on the allergic asthma of mice injected with BCG vaccine-stimulated DCs and then sensitized and challenged with OVA. The mice were injected intraperitoneally with anti-CD25 mAb before challenge with OVA. The plot legends are as described in Fig. 1 except that BCG-DC+ anti-CD25 indicate mice that received BCG-DCs and the anti-CD25 mAb. (A) BHR, *P<0.05, **P<0.01 compared to the OVA mice; †P<0.05 compared to the BCG-DC-treated mice. (B) Differential cell counts in the BAL fluid. (C) Lung pathology. (D) Peribronchial and perivascular lung inflammation scores. The values shown are the means±SD of the results from 5 mice per group. *P<0.05, **P<0.01.


Reference

1. Walker C, Bode E, Boer L, Hansel TT, Blaser K, Virchow JC Jr. Allergic and nonallergic asthmatics have distinct patterns of T-cell activation and cytokine production in peripheral blood and bronchoalveolar lavage. Am Rev Respir Dis. 1992; 146:109–115.
2. Lee SH, Park JS, Park CS. The search for genetic variants and epigenetics related to asthma. Allergy Asthma Immunol Res. 2011; 3:236–244.
3. Wang JM, Rambaldi A, Biondi A, Chen ZG, Sanderson CJ, Mantovani A. Recombinant human interleukin 5 is a selective eosinophil chemoattractant. Eur J Immunol. 1989; 19:701–705.
4. Strachan DP. Hay fever, hygiene, and household size. BMJ. 1989; 299:1259–1260.
5. Riedler J, Braun-Fahrlnder C, Eder W, Schreuer M, Waser M, Maisch S, Carr D, Schierl R, Nowak D, von Mutius E. ALEX Study Team. Exposure to farming in early life and development of asthma and allergy: a cross-sectional survey. Lancet. 2001; 358:1129–1133.
6. Ege MJ, Mayer M, Normand AC, Genuneit J, Cookson WO, Braun-Fahrlnder C, Heederik D, Piarroux R, von Mutius E. GABRIELA Transregio 22 Study Group. Exposure to environmental microorganisms and childhood asthma. N Engl J Med. 2011; 364:701–709.
7. Eder W, Ege MJ, von Mutius E. The asthma epidemic. N Engl J Med. 2006; 355:2226–2235.
8. Hong SJ, Ahn KM, Lee SY, Kim KE. The prevalences of asthma and allergic diseases in Korean children. Korean J Pediatr. 2008; 51:343–350.
9. Lee SI. Prevalence of childhood asthma in Korea: international study of asthma and allergies in childhood. Allergy Asthma Immunol Res. 2010; 2:61–64.
10. Bach JF. The effect of infections on susceptibility to autoimmune and allergic diseases. N Engl J Med. 2002; 347:911–920.
11. Seroogy CM, Gern JE. The role of T regulatory cells in asthma. J Allergy Clin Immunol. 2005; 116:996–999.
12. Curotto de Lafaille MA, Kutchukhidze N, Shen S, Ding Y, Yee H, Lafaille JJ. Adaptive Foxp3+ regulatory T cell-dependent and -independent control of allergic inflammation. Immunity. 2008; 29:114–126.
13. Robinson DS, Larch M, Durham SR. Tregs and allergic disease. J Clin Invest. 2004; 114:1389–1397.
14. Kwon HK, Lee CG, So JS, Chae CS, Hwang JS, Sahoo A, Nam JH, Rhee JH, Hwang KC, Im SH. Generation of regulatory dendritic cells and CD4+Foxp3+ T cells by probiotics administration suppresses immune disorders. Proc Natl Acad Sci U S A. 2010; 107:2159–2164.
15. Read S, Malmstrm V, Powrie F. Cytotoxic T lymphocyte-associated antigen 4 plays an essential role in the function of CD25(+)CD4(+) regulatory cells that control intestinal inflammation. J Exp Med. 2000; 192:295–302.
16. Hopfenspirger MT, Agrawal DK. Airway hyperresponsiveness, late allergic response, and eosinophilia are reversed with mycobacterial antigens in ovalbumin-presensitized mice. J Immunol. 2002; 168:2516–2522.
17. Erb KJ, Holloway JW, Sobeck A, Moll H, Le Gros G. Infection of mice with Mycobacterium bovis-Bacillus Calmette-Gu&eacute;rin (BCG) suppresses allergen-induced airway eosinophilia. J Exp Med. 1998; 187:561–569.
18. Jason J, Archibald LK, Nwanyanwu OC, Kazembe PN, Chatt JA, Norton E, Dobbie H, Jarvis WR. Clinical and immune impact of Mycobacterium bovis BCG vaccination scarring. Infect Immun. 2002; 70:6188–6195.
19. Li Q, Shen HH. Neonatal bacillus Calmette-Gu&eacute;rin vaccination inhibits de novo allergic inflammatory response in mice via alteration of CD4+CD25+ T-regulatory cells. Acta Pharmacol Sin. 2009; 30:125–133.
20. Hammad H, Lambrecht BN. Dendritic cells and epithelial cells: linking innate and adaptive immunity in asthma. Nat Rev Immunol. 2008; 8:193–204.
21. Geijtenbeek TB, Torensma R, van Vliet SJ, van Duijnhoven GC, Adema GJ, van Kooyk Y, Figdor CG. Identification of DC-SIGN, a novel dendritic cell-specific ICAM-3 receptor that supports primary immune responses. Cell. 2000; 100:575–585.
22. Steinman RM, Hawiger D, Nussenzweig MC. Tolerogenic dendritic cells. Annu Rev Immunol. 2003; 21:685–711.
23. Banchereau J, Steinman RM. Dendritic cells and the control of immunity. Nature. 1998; 392:245–252.
24. Brinster C, Shevach EM. Bone marrow-derived dendritic cells reverse the anergic state of CD4+CD25+ T cells without reversing their suppressive function. J Immunol. 2005; 175:7332–7340.
25. Watanabe N, Wang YH, Lee HK, Ito T, Wang YH, Cao W, Liu YJ. Hassall's corpuscles instruct dendritic cells to induce CD4+CD25+ regulatory T cells in human thymus. Nature. 2005; 436:1181–1185.
26. Chung DJ, Rossi M, Romano E, Ghith J, Yuan J, Munn DH, Young JW. Indoleamine 2,3-dioxygenase-expressing mature human monocyte-derived dendritic cells expand potent autologous regulatory T cells. Blood. 2009; 114:555–563.
27. Kim YJ, Kim HJ, Kang MJ, Kwon JW, Seo JH, Kim BJ, Yu J, Hong SJ. Effects of Bacillus Calmette-Gu&eacute;rin (BCG) vaccination according to time point of administration on asthma in a murine model. Korean J Asthma Allergy Clin Immunol. 2010; 30:307–313.
28. Choi MS, Park S, Choi T, Lee G, Haam KK, Hong MC, Min BI, Bae H. Bee venom ameliorates ovalbumin induced allergic asthma via modulating CD4+CD25+ regulatory T cells in mice. Cytokine. 2013; 61:256–265.
29. Yu J, Jang SO, Kim BJ, Song YH, Kwon JW, Kang MJ, Choi WA, Jung HD, Hong SJ. The effects of Lactobacillus rhamnosus on the prevention of asthma in a murine model. Allergy Asthma Immunol Res. 2010; 2:199–205.
30. Tournoy KG, Kips JC, Schou C, Pauwels RA. Airway eosinophilia is not a requirement for allergen-induced airway hyperresponsiveness. Clin Exp Allergy. 2000; 30:79–85.
31. Soualhine H, Deghmane AE, Sun J, Mak K, Talal A, Av-Gay Y, Hmama Z. Mycobacterium bovis bacillus Calmette-Gu&eacute;rin secreting active cathepsin S stimulates expression of mature MHC class II molecules and antigen presentation in human macrophages. J Immunol. 2007; 179:5137–5145.
32. Liu E, Law HK, Lau YL. BCG promotes cord blood monocyte-derived dendritic cell maturation with nuclear Rel-B up-regulation and cytosolic I kappa B alpha and beta degradation. Pediatr Res. 2003; 54:105–112.
33. Demangel C, Bean AG, Martin E, Feng CG, Kamath AT, Britton WJ. Protection against aerosol Mycobacterium tuberculosis infection using Mycobacterium bovis Bacillus Calmette Gu&eacute;rin-infected dendritic cells. Eur J Immunol. 1999; 29:1972–1979.
34. Han Y, Guo Q, Zhang M, Chen Z, Cao X. CD69+ CD4+ CD25- T cells, a new subset of regulatory T cells, suppress T cell proliferation through membrane-bound TGF-beta 1. J Immunol. 2009; 182:111–120.
35. Ahrens B, Gruber C, Rha RD, Freund T, Quarcoo D, Awagyan A, Hutloff A, Dittrich AM, Wahn U, Hamelmann E. BCG priming of dendritic cells enhances T regulatory and Th1 function and suppresses allergen-induced Th2 function in vitro and in vivo. Int Arch Allergy Immunol. 2009; 150:210–220.
36. Lagranderie M, Abolhassani M, Vanoirbeek JA, Lima C, Balazuc AM, Vargaftig BB, Marchal G. Mycobacterium bovis bacillus Calmette-Gu&eacute;rin killed by extended freeze-drying targets plasmacytoid dendritic cells to regulate lung inflammation. J Immunol. 2010; 184:1062–1070.
37. Lewkowich IP, Herman NS, Schleifer KW, Dance MP, Chen BL, Dienger KM, Sproles AA, Shah JS, Khl 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.
38. Lagranderie MR, Balazuc AM, Deriaud E, Leclerc CD, Gheorghiu M. Comparison of immune responses of mice immunized with five different Mycobacterium bovis BCG vaccine strains. Infect Immun. 1996; 64:1–9.
39. Fehérvari Z, Sakaguchi S. Development and function of CD25+CD4+ regulatory T cells. Curr Opin Immunol. 2004; 16:203–208.
40. Gao X, Bai H, Cheng J, Fan Y, Wang S, Jiao L, Xiu N, Yang X. CD8α+ and CD8α- DC subsets from BCG-infected mice inhibit allergic Th2-cell responses by enhancing Th1-cell and Treg-cell activity respectively. Eur J Immunol. 2012; 42:165–175.
41. Orme IM, Andersen P, Boom WH. T cell response to Mycobacterium tuberculosis. J Infect Dis. 1993; 167:1481–1497.
42. Christ AP, Rodriguez D, Bortolatto J, Borducchi E, Keller A, Mucida D, Silva JS, Leite LC, Russo M. Enhancement of Th1 lung immunity induced by recombinant Mycobacterium bovis Bacillus Calmette-Gu&eacute;rin attenuates airway allergic disease. Am J Respir Cell Mol Biol. 2010; 43:243–252.
43. Barlan I, Bahceciler NN, Akdis M, Akdis CA. Bacillus Calmette-Gu&eacute;rin, Mycobacterium bovis, as an immunomodulator in atopic diseases. Immunol Allergy Clin North Am. 2006; 26:365–377.
44. Fonseca DM, Paula MO, Wowk PF, Campos LW, Gembre AF, Turato WM, Ramos SG, Dias-Baruffi M, Barboza R, Gomes E, Horn C, Marchal G, Arruda LK, Russo M, Bonato VL. IFN-γ-mediated efficacy of allergen-free immunotherapy using mycobacterial antigens and CpG-ODN. Immunol Cell Biol. 2011; 89:777–785.
Full Text Links
  • AAIR
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr