The incidence of type-1 diabetes in NOD mice is modulated by restricted flora not germ-free conditions

Cecile King, Nora Sarvetnick, Cecile King, Nora Sarvetnick

Abstract

In the NOD mouse, the incidence of type-1 diabetes is thought to be influenced by the degree of cleanliness of the mouse colony. Studies collectively demonstrate that exposure to bacterial antigen or infection in the neonatal period prevents diabetes [1], [2], [3], [4], [5], [6], [7], [8], [9], [10], supporting the notion that immunostimulation can benefit the maturation of the postnatal immune system [11]. A widely accepted extrapolation from this data has been the notion that NOD mice maintained under germ-free conditions have an increased incidence of diabetes. However, evidence supporting this influential concept is surprisingly limited [12]. In this study, we demonstrate that the incidence of diabetes in female NOD mice remained unchanged under germ-free conditions. By contrast, a spontaneous monoculture with a gram-positive aerobic spore-forming rod delayed the onset and reduced the incidence of diabetes. These findings challenge the view that germ-free NOD mice have increased diabetes incidence and demonstrate that modulation of intestinal microbiota can prevent the development of type-1 diabetes.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. The incidence of type-1 diabetes…
Figure 1. The incidence of type-1 diabetes in female NOD mice remains unchanged under germ-free conditions but is reduced by restricted flora.
Cumulative diabetes incidence of germ-free female NOD mice (NOD germ-free n = 22), female NOD mice monocolonized with an aerobic spore-forming bacteria (Bacillus cereus) detected at week 16 (NOD AerSFB n = 22) and female NOD mice housed under specific pathogen-free conditions (NOD SPF Scripps n = 22, NOD SPF Taconic n = 40). Urine glucose was measured every one or two weeks as shown and mice that had reached 4+ (55 mmol/L) were considered diabetic. There was a significant inhibition (p<0.001) in the incidence of diabetes in monocolonized NOD mice compared with SPF NOD mice, but no significant difference between germ free and SPF NOD mice (p>0.05), Anova.

References

    1. Wilberz S, Partke HJ, Dagnaes-Hansen F, Herberg L. Persistent MHV (mouse hepatitis virus) infection reduces the incidence of diabetes mellitus in non-obese diabetic mice. Diabetologia. 1991;34:2–5.
    1. Oldstone MB. Prevention of type I diabetes in nonobese diabetic mice by virus infection. Science. 1988;239:500–502.
    1. Martins TC, Aguas AP. Changes in B and T lymphocytes associated with mycobacteria-induced protection of NOD mice from diabetes. J Autoimmun. 1996;9:501–507.
    1. Satoh J, Shintani S, Oya K, Tanaka S, Nobunaga T, et al. Treatment with streptococcal preparation (OK-432) suppresses anti-islet autoimmunity and prevents diabetes in BB rats. Diabetes. 1988;37:1188–1194.
    1. Qin HY, Singh B. BCG vaccination prevents insulin-dependent diabetes mellitus (IDDM) in NOD mice after disease acceleration with cyclophosphamide. J Autoimmun. 1997;10:271–278.
    1. Elias D, Markovits D, Reshef T, van der Zee R, Cohen IR. Induction and therapy of autoimmune diabetes in the non-obese diabetic (NOD/Lt) mouse by a 65-kDa heat shock protein. Proc Natl Acad Sci U S A. 1990;87:1576–1580.
    1. Sai P, Rivereau AS. Prevention of diabetes in the nonobese diabetic mouse by oral immunological treatments. Comparative efficiency of human insulin and two bacterial antigens, lipopolysacharide from Escherichia coli and glycoprotein extract from Klebsiella pneumoniae. Diabetes Metab. 1996;22:341–348.
    1. Ramiya VK, Shang XZ, Pharis PG, Wasserfall CH, Stabler TV, et al. Antigen based therapies to prevent diabetes in NOD mice. J Autoimmun. 1996;9:349–356.
    1. Sadelain MW, Qin HY, Lauzon J, Singh B. Prevention of type I diabetes in NOD mice by adjuvant immunotherapy. Diabetes. 1990;39:583–589.
    1. McInerney MF, Pek SB, Thomas DW. Prevention of insulitis and diabetes onset by treatment with complete Freund's adjuvant in NOD mice. Diabetes. 1991;40:715–725.
    1. Holt PG. Postnatal maturation of immune competence during infancy and childhood. Pediatr Allergy Immunol. 1995;6:59–70.
    1. Maclaren N, Schatz D, Drash A, Grave G. Initial pathogenic events in IDDM. Diabetes. 1989;38:534–538.
    1. Hudson JA, Luckey TD. Bacteria Induced Morphologic Changes. Proc Soc Exp Biol Med. 1964;116:628–631.
    1. Glaister JR. Factors affecting the lymphoid cells in the small intestinal epithelium of the mouse. Int Arch Allergy Appl Immunol. 1973;45:719–730.
    1. Hooijkaas H, Benner R, Pleasants JR, Wostmann BS. Isotypes and specificities of immunoglobulins produced by germ-free mice fed chemically defined ultrafiltered "antigen-free" diet. Eur J Immunol. 1984;14:1127–1130.
    1. Cebra JJ, Periwal SB, Lee G, Lee F, Shroff KE. Development and maintenance of the gut-associated lymphoid tissue (GALT): the roles of enteric bacteria and viruses. Dev Immunol. 1998;6:13–18.
    1. Crabbe PA, Nash DR, Bazin H, Eyssen H, Heremans JF. Immunohistochemical observations on lymphoid tissues from conventional and germ-free mice. Lab Invest. 1970;22:448–457.
    1. Macpherson AJ, Gatto D, Sainsbury E, Harriman GR, Hengartner H, et al. A primitive T cell-independent mechanism of intestinal mucosal IgA responses to commensal bacteria. Science. 2000;288:2222–2226.
    1. Hapfelmeier S, Lawson MA, Slack E, Kirundi JK, Stoel M, et al. Reversible microbial colonization of germ-free mice reveals the dynamics of IgA immune responses. Science. 2010;328:1705–1709.
    1. Bouskra D, Brezillon C, Berard M, Werts C, Varona R, et al. Lymphoid tissue genesis induced by commensals through NOD1 regulates intestinal homeostasis. Nature. 2008;456:507–510.
    1. Mazmanian SK, Liu CH, Tzianabos AO, Kasper DL. An immunomodulatory molecule of symbiotic bacteria directs maturation of the host immune system. Cell. 2005;122:107–118.
    1. King C, Ilic A, Koelsch K, Sarvetnick N. homeostatic Expansion of T cells during immune Insufficiency Generates Autoimmunity. Cell. 2004;117:265–277.
    1. Shehadeh NN, LaRosa F, Lafferty KJ. Altered cytokine activity in adjuvant inhibition of autoimmune diabetes. J Autoimmun. 1993;6:291–300.
    1. Gombert JM, Herbelin A, Tancrede-Bohin E, Dy M, Chatenoud L, et al. Early defect of immunoregulatory T cells in autoimmune diabetes. C R Acad Sci III. 1996;319:125–129.
    1. Baxter AG, Kinder SJ, Hammond KJ, Scollay R, Godfrey DI. Association between alphabetaTCR+CD4-CD8- T-cell deficiency and IDDM in NOD/Lt mice. Diabetes. 1997;46:572–582.
    1. Martins TC, Aguas AP. Mechanisms of Mycobacterium avium-induced resistance against insulin-dependent diabetes mellitus (IDDM) in non-obese diabetic (NOD) mice: role of Fas and Th1 cells. Clin Exp Immunol. 1999;115:248–254.
    1. Elliott RB, Reddy SN, Bibby NJ, Kida K. Dietary prevention of diabetes in the non-obese diabetic mouse. Diabetologia. 1988;31:62–64.
    1. Mueller DB, Koczwara K, Mueller AS, Pallauf J, Ziegler AG, et al. Influence of early nutritional components on the development of murine autoimmune diabetes. Ann Nutr Metab. 2009;54:208–217.
    1. Alam C, Valkonen S, Palagani V, Jalava J, Eerola E, et al. Inflammatory tendencies and overproduction of IL-17 in the colon of young NOD mice are counteracted with diet change. Diabetes. 2010;59:2237–2246.
    1. Wen L, Ley RE, Volchkov PY, Stranges PB, Avanesyan L, et al. Innate immunity and intestinal microbiota in the development of Type 1 diabetes. Nature. 2008;455:1109–1113.

Source: PubMed

3
Sottoscrivi