J Vet Sci.  2016 Dec;17(4):505-513. 10.4142/jvs.2016.17.4.505.

Mycobacterium vaccae induces a strong Th1 response that subsequently declines in C57BL/6 mice

Affiliations
  • 1College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China. wangchunfeng@jlau.edu.cn qianaidong0115@163.com
  • 2College of Biological Science, Changchun Teacher University, Changchun 130032, China.

Abstract

Mycobacterium (M.) vaccae is a fast-growing species of saprophytic bacteria that is widely distributed. To understand the host immune responses induced by M. vaccae isolated from bovine submaxillary lymph nodes, C57BL/6 mice were infected with reference strain M. vaccae Bacillus Calmette-Guérin (BCG) and isolated M. vaccae using intraperitoneal injections. Comparison of the bacterial replication and organ pathology between M. vaccae and M. vaccae BCG revealed that M. vaccae was more malignant than M. vaccae in mice. We also demonstrated that serum from the M. vaccae-infected mice contained a higher expression level of gamma-interferon (IFN-γ), tumor necrosis factor alpha, monocyte chemoattractant protein-1, interleukin (IL)-4, IL-12, IL-10 and transforming growth factor beta than did the other groups, especially after week 4. Furthermore, when the numbers of CD3⁺CD4⁺IFN-γ⁺ and CD3⁺CD4⁺IL4⁺ cells in the infected mice were observed by flow cytometry, we found that a powerful T helper 1 (Th1) response was induced by M. vaccae infection, which was associated with the emergence of CD3⁺CD4⁺IFN-γ⁺ cells. However, the Th1 response declined over time, which was associated with appearance of the CD4⁺CD25⁺FoxP3⁺ and CD4⁺CD25⁺CD152⁺Treg cell reaction. In addition, a strong Th2 response was found. Finally, we found that M. vaccae infection increased the production of type I IFNs, which was associated with a reduced Th1 response.

Keyword

Mycobacterium bovis Bacillus Calmette-Guérin; Mycobacterium vaccae; Th1 cells response; regulatory T cell

MeSH Terms

Animals
Cattle
Cytokines/*genetics/metabolism
Disease Models, Animal
Female
Mice
Mice, Inbred C57BL
Mycobacterium/*immunology
Mycobacterium Infections/*immunology
Mycobacterium bovis/physiology
Specific Pathogen-Free Organisms
Th1 Cells/*immunology
Tuberculosis, Bovine/immunology/microbiology
Cytokines

Figure

  • Fig. 1 Lung histology of Mycobacterium (M.) vaccae and M. bovis Bacillus Calmette-Guérin (BCG)-infected mice. (A) Bacterial counts in the lungs at weeks 2, 4, 6 and 8 after infection of mice infected with M. vaccae or M. bovis BCG. Student's t-test vs. the M. bovis BCG group, **p < 0.01, ***p < 0.001. (B) Representative lung acid fast staining bacilli two weeks after M. vaccae or M. bovis BCG infection. (C) Representative lung histopathology in infected animals on weeks 2, 4, 6 and 8 of the infection. H&E stain. 800× (B), 40× (C).

  • Fig. 2 Early infiltration of CD4+, CD8+ T cells, CD3+CD4+ IFN-γ+ cells and CD3+ CD4+ IL4+ cells in the lungs of mice infected with M. vaccae and M. bovis BCG. (A) Total number of cells expressing CD3+CD4+ surface markers. (B) CD3+CD8+ surface markers in the lungs of mice infected with various strains. (C) Number of CD3+ and CD4+ IFN-γ+ cells in M. vaccae-infected mice. (D) Number of CD3+ and CD4+ IL4+ cells in M. vaccae-infected mice. The results are expressed as the average ± SEM (n = 5 mice) of CD3+CD4+, CD3+CD8+ T cells, CD3+CD4+ IFN-γ+ cells or CD3+ and CD4+ IL4+ cells in the lungs. Student's t-test vs. the normal group, *p < 0.05, **p < 0.01, ***p < 0.001.

  • Fig. 3 Differential production of IFN-γ, TNF-α, MCP-1, IL-12p40, IL-10 and TGF-β cytokines in the serum of C57BL/6 mice infected with various bacterial strains (M. vaccae and M. bovis BCG). The levels of IFN-γ, TNF-α, MCP-1, IL-12p40, IL-10 and TGF-β were measured by ELISA. The results are expressed as the means ± SEM (n = 5 mice) in the serum. Student's t-test vs. the normal group, *p < 0.05, **p < 0.01, ***p < 0.001.

  • Fig. 4 Early infiltration of DCs and pDCs in the lungs of mice infected with the M. vaccae isolate and M. bovis BCG. Lung cells obtained from C57BL/6 mice infected with M. vaccae or M. bovis BCG and normal mice were assayed for the total number of pDCs (A) and DCs (B) using flow cytometry. The results are expressed as the average ± SEM of (n = 5 mice) pDCs or DCs in the lungs. Student's t-test vs. the normal group, *p < 0.05, **p < 0.01, ***p < 0.001.

  • Fig. 5 CD4+CD25+Foxp3+ cells in infected mice. Mice infected with M. vaccae showed a substantial increase in CD4+CD25+Foxp3+ regulatory T cells in the lungs relative to the M. bovis BCG and normal groups (A). Mice infected with M. vaccae showed a significant increase in CD4+CD25+CD152+T cells in the lungs relative to the M. bovis BCG and normal groups of mice at 3 weeks, whereas a larger number was observed in M. bovis BCG-infected mice beginning at 3 weeks (B). The results are expressed as the mean number ± SEM of CD4+CD25+Foxp3+ or CD4+CD25+CD152+ T cells (n = 5 mice) in the lungs. Student's t-test vs. the normal group, *p < 0.05, **p < 0.01, ***p < 0.001.


Reference

1. Aksamit TR, Philley JV, Griffith DE. Nontuberculous mycobacterial (NTM) lung disease: the top ten essentials. Respir Med. 2014; 108:417–425.
Article
2. Altare F, Durandy A, Lammas D, Emile JF, Lamhamedi S, Le Deist F, Drysdale P, Jouanguy E, Döffinger R, Bernaudin F, Jeppsson O, Gollob JA, Meinl E, Segal AW, Fischer A, Kumararatne D, Casanova JL. Impairment of mycobacterial immunity in human interleukin-12 receptor deficiency. Science. 1998; 280:1432–1435.
Article
3. Appelberg R, Castro AG, Pedrosa J, Silva RA, Orme IM, Minóprio P. Role of gamma interferon and tumor necrosis factor alpha during T-cell-independent and -dependent phases of Mycobacterium avium infection. Infect Immun. 1994; 62:3962–3971.
Article
4. Arlotta A, Cefalo MG, Maurizi P, Ruggiero A, Dodi I, Riccardi R. Critical pulmonary infection due to nontuberculous mycobacterium in pediatric leukemia: report of a difficult diagnosis and review of pediatric series. J Pediatr Hematol Oncol. 2014; 36:66–70.
Article
5. Asselin-Paturel C, Trinchieri G. Production of type I interferons: plasmacytoid dendritic cells and beyond. J Exp Med. 2005; 202:461–465.
6. Bönicke R, Juhasz SE. Description of new species Mycobacterium vaccae no. sp. Zentralbl Bakteriol Parasitenkd Infekt Hyg. 1964; 192:133–135.
7. Brown-Elliott BA, Wallace RJ Jr. Clinical and taxonomic status of pathogenic nonpigmented or late-pigmenting rapidly growing mycobacteria. Clin Microbiol Rev. 2002; 15:716–746.
Article
8. Daley CL, Griffith DE. Pulmonary disease caused by rapidly growing mycobacteria. Clin Chest Med. 2002; 23:623–632. vii
Article
9. Dheda K, Booth H, Huggett JF, Johnson MA, Zumla A, Rook GAW. Lung remodeling in pulmonary tuberculosis. J Infect Dis. 2005; 192:1201–1209.
Article
10. Dorman SE, Picard C, Lammas D, Heyne K, van Dissel JT, Baretto R, Rosenzweig SD, Newport M, Levin M, Roesler J, Kumararatne D, Casanova JL, Holland SM. Clinical features of dominant and recessive interferon gamma receptor 1 deficiencies. Lancet. 2004; 364:2113–2121.
Article
11. Egelund EF, Fennelly KP, Peloquin CA. Medications and monitoring in nontuberculous mycobacteria infections. Clin Chest Med. 2015; 36:55–66.
Article
12. Feng CG, Jankovic D, Kullberg M, Cheever A, Scanga CA, Hieny S, Caspar P, Yap GS, Sher A. Maintenance of pulmonary Th1 effector function in chronic tuberculosis requires persistent IL-12 production. J Immunol. 2005; 174:4185–4192.
Article
13. Flynn JL, Chan J. Immunology of tuberculosis. Annu Rev Immunol. 2001; 19:93–129.
Article
14. Gonzalez-Santiago TM, Drage LA. Nontuberculous mycobacteria: skin and soft tissue infections. Dermatol Clin. 2015; 33:563–577.
15. Hambleton S, Salem S, Bustamante J, Bigley V, Boisson-Dupuis S, Azevedo J, Fortin A, Haniffa M, Ceron-Gutierrez L, Bacon CM, Menon G, Trouillet C, McDonald D, Carey P, Ginhoux F, Alsina L, Zumwalt TJ, Kong XF, Kumararatne D, Butler K, Hubeau M, Feinberg J, Al-Muhsen S, Cant A, Abel L, Chaussabel D, Doffinger R, Talesnik E, Grumach A, Duarte A, Abarca K, Moraes-Vasconcelos D, Burk D, Berghuis A, Geissmann F, Collin M, Casanova JL, Gros P. IRF8 mutations and human dendritic-cell immunodeficiency. N Engl J Med. 2011; 365:127–138.
Article
16. Haug M, Awuh JA, Steigedal M, Frengen Kojen J, Marstad A, Nordrum IS, Halaas Ø, Flo TH. Dynamics of immune effector mechanisms during infection with Mycobacterium avium in C57BL/6 mice. Immunology. 2013; 140:232–243.
Article
17. Henkle E, Winthrop KL. Nontuberculous mycobacteria infections in immunosuppressed hosts. Clin Chest Med. 2015; 36:91–99.
Article
18. Hernandez-Pando R, Pavon L, Orozco EH, Rangel J, Rook GA. Interactions between hormone-mediated and vaccine-mediated immunotherapy for pulmonary tuberculosis in BALB/c mice. Immunology. 2000; 100:391–398.
Article
19. Jönsson B, Ridell M, Wold AE. Non-tuberculous mycobacteria and their surface lipids efficiently induced IL-17 production in human T cells. Microbes Infect. 2012; 14:1186–1195.
Article
20. Kobayashi T, Morino E, Takasaki J, Nagahara Y, Sugiyama H. Nontuberculous mycobacterial osteomyelitis in human immunodeficiency virus-negative patients: a case series. Jpn J Infect Dis. 2016; 69:149–150.
Article
21. Koehne G, Maddux R, Britt J. Rapidly growing mycobacteria associated with bovine mastitis. Am J Vet Res. 1981; 42:1238–1239.
22. Lienhardt C, Azzurri A, Amedei A, Fielding K, Sillah J, Sow OY, Bah B, Benagiano M, Diallo A, Manetti R, Manneh K, Gustafson P, Bennett S, D'Elios MM, McAdam K, Del Prete G. Active tuberculosis in Africa is associated with reduced Th1 and increased Th2 activity in vivo. Eur J Immunol. 2002; 32:1605–1613.
Article
23. Monack DM, Mueller A, Falkow S. Persistent bacterial infections: the interface of the pathogen and the host immune system. Nat Rev Microbiol. 2004; 2:747–765.
Article
24. Ordway D, Henao-Tamayo M, Harton M, Palanisamy G, Troudt J, Shanley C, Basaraba RJ, Orme IM. The hypervirulent Mycobacterium tuberculosis strain HN878 induces a potent Th1 response followed by rapid down-regulation. J Immunol. 2007; 179:522–531.
Article
25. Ottenhoff TH, Verreck FAW, Hoeve MA, van de Vosse E. Control of human host immunity to mycobacteria. Tuberculosis (Edinb). 2005; 85:53–64.
Article
26. Quinn KM, McHugh RS, Rich FJ, Goldsack LM, de Lisle GW, Buddle BM, Delahunt B, Kirman JR. Inactivation of CD4+ CD25+ regulatory T cells during early mycobacterial infection increases cytokine production but does not affect pathogen load. Immunol Cell Biol. 2006; 84:467–474.
Article
27. Ramsdell F. Foxp3 and natural regulatory T cells: key to a cell lineage? Immunity. 2003; 19:165–168.
28. Hachem R, Raad I, Rolston KVI, Whimbey E, Katz R, Tarrand J, Libshitz H. Cutaneous and pulmonary infections caused by Mycobacterium vaccae. Clin Infect Dis. 1996; 23:173–175.
29. Rottman M, Catherinot E, Hochedez P, Emile JF, Casanova JL, Gaillard JL, Soudais C. Importance of T cells, gamma interferon, and tumor necrosis factor in immune control of the rapid grower Mycobacterium abscessus in C57BL/6 mice. Infect Immun. 2007; 75:5898–5907.
Article
30. Seddiki N, Sasson SC, Santner-Nanan B, Munier M, van Bockel D, Ip S, Marriott D, Pett S, Nanan R, Cooper DA, Zaunders JJ, Kelleher AD. Proliferation of weakly suppressive regulatory CD4+ T cells is associated with over-active CD4+ T-cell responses in HIV-positive patients with mycobacterial immune restoration disease. Eur J Immunol. 2009; 39:391–403.
Article
31. Shang S, Gibbs S, Henao-Tamayo M, Shanley CA, McDonnell G, Duarte RS, Ordway DJ, Jackson M. Increased virulence of an epidemic strain of Mycobacterium massiliense in mice. PLoS One. 2011; 6:e24726.
32. Shimizu K, Hirose T, Sato M, Tsukamura M. Isolation of acid-fast organisms resembling Mycobacterium vaccae from a lesion of bovine nodular thelitis. Microbiol Immunol. 1977; 21:469–472.
Article
33. Starke JR. Committee on Infectious Diseases. Interferon-γ release assays for diagnosis of tuberculosis infection and disease in children. Pediatrics. 2014; 134:e1763–e1773.
34. Urdahl KB, Shafiani S, Ernst JD. Initiation and regulation of T-cell responses in tuberculosis. Mucosal Immunol. 2011; 4:288–293.
Article
35. Velayati AA, Farnia P, Mozafari M, Mirsaeidi M. Nontuberculous mycobacteria isolation from clinical and environmental samples in Iran: twenty years of surveillance. Biomed Res Int. 2015; 2015:254285.
Article
36. von Reyn CF, Mtei L, Arbeit RD, Waddell R, Cole B, Mackenzie T, Matee M, Bakari M, Tvaroha S, Adams LV, Horsburgh CR, Pallangyo K. DarDar Study Group. Prevention of tuberculosis in Bacille Calmette-Guerin-primed, HIV-infected adults boosted with an inactivated whole-cell mycobacterial vaccine. AIDS. 2010; 24:675–685.
Article
37. Wang C, Qi H, Jiang X, Chen FF, Ma H, Wang C, Qian A. Molecular differentiation of nontuberculous Mycobacterium isolated from different animals. Appl Mech Mater. 2013; 421:300–303.
Article
38. Wang CC, Rook GA. Inhibition of an established allergic response to ovalbumin in BALB/c mice by killed Mycobacterium vaccae. Immunology. 1998; 93:307–313.
Article
39. Xu LJ, Wang Y, Zheng X, Gui X, Tao L, Wei H. Immunotherapeutical potential of Mycobacterium vaccae on M. tuberculosis infection in mice. Cell Mol Immunol. 2009; 6:67–72.
Article
40. Zuany-Amorim C, Manlius C, Trifilieff A, Brunet LR, Rook G, Bowen G, Pay G, Walker C. Long-term protective and antigen-specific effect of heat-killed Mycobacterium vaccae in a murine model of allergic pulmonary inflammation. J Immunol. 2002; 169:1492–1499.
Article
Full Text Links
  • JVS
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