Targeting gut microbiota and barrier function with prebiotics to alleviate autoimmune manifestations in NOD mice

Research output: Contribution to journalJournal articleResearchpeer-review

Standard

Targeting gut microbiota and barrier function with prebiotics to alleviate autoimmune manifestations in NOD mice. / Hansen, Camilla H. F.; Larsen, Christian S.; Petersson, Henriette O.; Zachariassen, Line F.; Vegge, Andreas; Lauridsen, Charlotte; Kot, Witold; Krych, Lukasz; Nielsen, Dennis S.; Hansen, Axel K.

In: Diabetologia, Vol. 62, No. 9, 2019, p. 1689-1700.

Research output: Contribution to journalJournal articleResearchpeer-review

Harvard

Hansen, CHF, Larsen, CS, Petersson, HO, Zachariassen, LF, Vegge, A, Lauridsen, C, Kot, W, Krych, L, Nielsen, DS & Hansen, AK 2019, 'Targeting gut microbiota and barrier function with prebiotics to alleviate autoimmune manifestations in NOD mice', Diabetologia, vol. 62, no. 9, pp. 1689-1700. https://doi.org/10.1007/s00125-019-4910-5

APA

Hansen, C. H. F., Larsen, C. S., Petersson, H. O., Zachariassen, L. F., Vegge, A., Lauridsen, C., Kot, W., Krych, L., Nielsen, D. S., & Hansen, A. K. (2019). Targeting gut microbiota and barrier function with prebiotics to alleviate autoimmune manifestations in NOD mice. Diabetologia, 62(9), 1689-1700. https://doi.org/10.1007/s00125-019-4910-5

Vancouver

Hansen CHF, Larsen CS, Petersson HO, Zachariassen LF, Vegge A, Lauridsen C et al. Targeting gut microbiota and barrier function with prebiotics to alleviate autoimmune manifestations in NOD mice. Diabetologia. 2019;62(9):1689-1700. https://doi.org/10.1007/s00125-019-4910-5

Author

Hansen, Camilla H. F. ; Larsen, Christian S. ; Petersson, Henriette O. ; Zachariassen, Line F. ; Vegge, Andreas ; Lauridsen, Charlotte ; Kot, Witold ; Krych, Lukasz ; Nielsen, Dennis S. ; Hansen, Axel K. / Targeting gut microbiota and barrier function with prebiotics to alleviate autoimmune manifestations in NOD mice. In: Diabetologia. 2019 ; Vol. 62, No. 9. pp. 1689-1700.

Bibtex

@article{e102d820f4d245c788e3e8d5d0a81b07,
title = "Targeting gut microbiota and barrier function with prebiotics to alleviate autoimmune manifestations in NOD mice",
abstract = "Aims/hypothesisAdopting a diet containing indigestible fibre compounds such as prebiotics to fuel advantageous bacteria has proven beneficial for alleviating inflammation. The effect of the microbial changes on autoimmunity, however, remains unknown. We studied the effects of prebiotic xylooligosaccharides (XOS) on pancreatic islet and salivary gland inflammation in NOD mice and tested whether these were mediated by the gut microbiota.MethodsMother and offspring mice were fed an XOS-supplemented diet until diabetes onset or weaning and were compared with a control-fed group. Diabetes incidence was monitored, insulitis and sialadenitis were scored in histological sections from adult mice, and several metabolic and immune variables were analysed in mice before the development of diabetes. Gut barrier function was assessed using an in vivo FITC-dextran permeability test. The importance of XOS-mediated gut microbial changes were evaluated in antibiotic-treated mice fed either XOS or control diet or given a faecal microbiota transplant from test animals.ResultsDiabetes onset was delayed in the XOS-fed mice, which also had fewer cellular infiltrations in their pancreatic islets and salivary glands. Interestingly, insulitis was most reduced in the XOS-fed groups when the mice were also treated with an antibiotic cocktail. There was no difference in sialadenitis between the dietary groups treated with antibiotics; the mice were protected by microbiota depletion regardless of diet. Faecal microbiota transplantation was not able to transfer protection. No major differences in glucose–insulin regulation, glucagon-like peptide-1, or short-chain fatty acid production were related to the XOS diet. The XOS diet did, however, reduce gut permeability markers in the small and large intestine. This was accompanied by a more anti-inflammatory environment locally and systemically, dominated by a shift from M1 to M2 macrophages, a higher abundance of activated regulatory T cells, and lower levels of induction of natural killer T cells and cytotoxic T cells.Conclusions/interpretationPrebiotic XOS have microbiota-dependent effects on salivary gland inflammation and microbiota-independent effects on pancreatic islet pathology that are accompanied by an improved gut barrier that seems able to heighten control of intestinal diabetogenic antigens that have the potential to penetrate the mucosa to activate autoreactive immune responses.",
keywords = "Gut barrier, Gut microbiota, Insulitis, Mucosal immunology, Prebiotics, Sialadenitis, XOS",
author = "Hansen, {Camilla H. F.} and Larsen, {Christian S.} and Petersson, {Henriette O.} and Zachariassen, {Line F.} and Andreas Vegge and Charlotte Lauridsen and Witold Kot and Lukasz Krych and Nielsen, {Dennis S.} and Hansen, {Axel K.}",
year = "2019",
doi = "10.1007/s00125-019-4910-5",
language = "English",
volume = "62",
pages = "1689--1700",
journal = "Diabetologia",
issn = "0012-186X",
publisher = "Springer",
number = "9",

}

RIS

TY - JOUR

T1 - Targeting gut microbiota and barrier function with prebiotics to alleviate autoimmune manifestations in NOD mice

AU - Hansen, Camilla H. F.

AU - Larsen, Christian S.

AU - Petersson, Henriette O.

AU - Zachariassen, Line F.

AU - Vegge, Andreas

AU - Lauridsen, Charlotte

AU - Kot, Witold

AU - Krych, Lukasz

AU - Nielsen, Dennis S.

AU - Hansen, Axel K.

PY - 2019

Y1 - 2019

N2 - Aims/hypothesisAdopting a diet containing indigestible fibre compounds such as prebiotics to fuel advantageous bacteria has proven beneficial for alleviating inflammation. The effect of the microbial changes on autoimmunity, however, remains unknown. We studied the effects of prebiotic xylooligosaccharides (XOS) on pancreatic islet and salivary gland inflammation in NOD mice and tested whether these were mediated by the gut microbiota.MethodsMother and offspring mice were fed an XOS-supplemented diet until diabetes onset or weaning and were compared with a control-fed group. Diabetes incidence was monitored, insulitis and sialadenitis were scored in histological sections from adult mice, and several metabolic and immune variables were analysed in mice before the development of diabetes. Gut barrier function was assessed using an in vivo FITC-dextran permeability test. The importance of XOS-mediated gut microbial changes were evaluated in antibiotic-treated mice fed either XOS or control diet or given a faecal microbiota transplant from test animals.ResultsDiabetes onset was delayed in the XOS-fed mice, which also had fewer cellular infiltrations in their pancreatic islets and salivary glands. Interestingly, insulitis was most reduced in the XOS-fed groups when the mice were also treated with an antibiotic cocktail. There was no difference in sialadenitis between the dietary groups treated with antibiotics; the mice were protected by microbiota depletion regardless of diet. Faecal microbiota transplantation was not able to transfer protection. No major differences in glucose–insulin regulation, glucagon-like peptide-1, or short-chain fatty acid production were related to the XOS diet. The XOS diet did, however, reduce gut permeability markers in the small and large intestine. This was accompanied by a more anti-inflammatory environment locally and systemically, dominated by a shift from M1 to M2 macrophages, a higher abundance of activated regulatory T cells, and lower levels of induction of natural killer T cells and cytotoxic T cells.Conclusions/interpretationPrebiotic XOS have microbiota-dependent effects on salivary gland inflammation and microbiota-independent effects on pancreatic islet pathology that are accompanied by an improved gut barrier that seems able to heighten control of intestinal diabetogenic antigens that have the potential to penetrate the mucosa to activate autoreactive immune responses.

AB - Aims/hypothesisAdopting a diet containing indigestible fibre compounds such as prebiotics to fuel advantageous bacteria has proven beneficial for alleviating inflammation. The effect of the microbial changes on autoimmunity, however, remains unknown. We studied the effects of prebiotic xylooligosaccharides (XOS) on pancreatic islet and salivary gland inflammation in NOD mice and tested whether these were mediated by the gut microbiota.MethodsMother and offspring mice were fed an XOS-supplemented diet until diabetes onset or weaning and were compared with a control-fed group. Diabetes incidence was monitored, insulitis and sialadenitis were scored in histological sections from adult mice, and several metabolic and immune variables were analysed in mice before the development of diabetes. Gut barrier function was assessed using an in vivo FITC-dextran permeability test. The importance of XOS-mediated gut microbial changes were evaluated in antibiotic-treated mice fed either XOS or control diet or given a faecal microbiota transplant from test animals.ResultsDiabetes onset was delayed in the XOS-fed mice, which also had fewer cellular infiltrations in their pancreatic islets and salivary glands. Interestingly, insulitis was most reduced in the XOS-fed groups when the mice were also treated with an antibiotic cocktail. There was no difference in sialadenitis between the dietary groups treated with antibiotics; the mice were protected by microbiota depletion regardless of diet. Faecal microbiota transplantation was not able to transfer protection. No major differences in glucose–insulin regulation, glucagon-like peptide-1, or short-chain fatty acid production were related to the XOS diet. The XOS diet did, however, reduce gut permeability markers in the small and large intestine. This was accompanied by a more anti-inflammatory environment locally and systemically, dominated by a shift from M1 to M2 macrophages, a higher abundance of activated regulatory T cells, and lower levels of induction of natural killer T cells and cytotoxic T cells.Conclusions/interpretationPrebiotic XOS have microbiota-dependent effects on salivary gland inflammation and microbiota-independent effects on pancreatic islet pathology that are accompanied by an improved gut barrier that seems able to heighten control of intestinal diabetogenic antigens that have the potential to penetrate the mucosa to activate autoreactive immune responses.

KW - Gut barrier

KW - Gut microbiota

KW - Insulitis

KW - Mucosal immunology

KW - Prebiotics

KW - Sialadenitis

KW - XOS

U2 - 10.1007/s00125-019-4910-5

DO - 10.1007/s00125-019-4910-5

M3 - Journal article

C2 - 31139852

VL - 62

SP - 1689

EP - 1700

JO - Diabetologia

JF - Diabetologia

SN - 0012-186X

IS - 9

ER -

ID: 225956324