Short-term Oral Antibiotics Treatment Promotes Inflammatory Activation of Colonic Invariant Natural Killer T and Conventional CD4+ T Cells

The gut mucosa is continuously exposed to a vast community of microorganisms, collectively defined as microbiota, establishing a mutualistic relationship with the host and contributing to shape the immune system. Gut microbiota is acquired at birth, and its composition is relatively stable during th...

Full description

Bibliographic Details
Main Authors: Claudia Burrello, Federica Garavaglia, Fulvia Milena Cribiù, Giulia Ercoli, Silvano Bosari, Flavio Caprioli, Federica Facciotti
Format: Article
Language:English
Published: Frontiers Media S.A. 2018-02-01
Series:Frontiers in Medicine
Subjects:
Online Access:http://journal.frontiersin.org/article/10.3389/fmed.2018.00021/full
id doaj-aa28cdc59f154fb79b0c777ffe9f5384
record_format Article
spelling doaj-aa28cdc59f154fb79b0c777ffe9f53842020-11-24T22:25:21ZengFrontiers Media S.A.Frontiers in Medicine2296-858X2018-02-01510.3389/fmed.2018.00021334642Short-term Oral Antibiotics Treatment Promotes Inflammatory Activation of Colonic Invariant Natural Killer T and Conventional CD4+ T CellsClaudia Burrello0Claudia Burrello1Federica Garavaglia2Fulvia Milena Cribiù3Giulia Ercoli4Silvano Bosari5Flavio Caprioli6Flavio Caprioli7Federica Facciotti8Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, ItalyDepartment of Experimental Oncology, European Institute of Oncology, Milan, ItalyDepartment of Experimental Oncology, European Institute of Oncology, Milan, ItalyPathology Unit, Fondazione IRCCS Cà Granda, Ospedale Policlinico di Milano, Milan, ItalyPathology Unit, Fondazione IRCCS Cà Granda, Ospedale Policlinico di Milano, Milan, ItalyPathology Unit, Fondazione IRCCS Cà Granda, Ospedale Policlinico di Milano, Milan, ItalyGastroenterology and Endoscopy Unit, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico, Milan, ItalyDepartment of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, ItalyDepartment of Experimental Oncology, European Institute of Oncology, Milan, ItalyThe gut mucosa is continuously exposed to a vast community of microorganisms, collectively defined as microbiota, establishing a mutualistic relationship with the host and contributing to shape the immune system. Gut microbiota is acquired at birth, and its composition is relatively stable during the entire adult life. Intestinal dysbiosis, defined as a microbial imbalance of gut bacterial communities, can be caused by several factors, including bacterial infections and antibiotic use, and has been associated with an increased risk to develop or exacerbate immune-mediated pathologies, such as allergic reactions, asthma, and inflammatory bowel diseases. Still, the mechanisms by which antibiotic-induced gut dysbiosis may lead to development of mucosal inflammation are still matter of debate. To this end, we aimed to evaluate the impact of antibiotic treatment on phenotype and functions of intestinal immune cell populations, including invariant natural killer T (iNKT) cells, a subset of lipid-specific T cells profoundly influenced by alterations on the commensal microbiota. To this aim, a cocktail of broad-spectrum antibiotics was administered for 2 weeks to otherwise healthy mice before re-colonization of the intestinal microbial community with oral gavage of eubiotic or dysbiotic mucosa-associated bacteria and luminal colonic content, followed or not by intestinal inflammation induction. Here. we showed that short-term antibiotic treatment alters frequency and functions of intestinal iNKT cells, even in the absence of intestinal inflammation. The presence of a dysbiotic microbiota after antibiotic treatment imprints colonic iNKT and CD4+ T cells toward a pro-inflammatory phenotype that collectively contributes to aggravate intestinal inflammation. Nonetheless, the inflammatory potential of the dysbiotic microbiota decreases over time opening the possibility to temporally intervene on the microbial composition to re-equilibrate dysbiosis, thus controlling concomitantly mucosal immune T cell activations.http://journal.frontiersin.org/article/10.3389/fmed.2018.00021/fulliNKT cellsantibioticsmicrobiotaT cellsintestinal inflammation
collection DOAJ
language English
format Article
sources DOAJ
author Claudia Burrello
Claudia Burrello
Federica Garavaglia
Fulvia Milena Cribiù
Giulia Ercoli
Silvano Bosari
Flavio Caprioli
Flavio Caprioli
Federica Facciotti
spellingShingle Claudia Burrello
Claudia Burrello
Federica Garavaglia
Fulvia Milena Cribiù
Giulia Ercoli
Silvano Bosari
Flavio Caprioli
Flavio Caprioli
Federica Facciotti
Short-term Oral Antibiotics Treatment Promotes Inflammatory Activation of Colonic Invariant Natural Killer T and Conventional CD4+ T Cells
Frontiers in Medicine
iNKT cells
antibiotics
microbiota
T cells
intestinal inflammation
author_facet Claudia Burrello
Claudia Burrello
Federica Garavaglia
Fulvia Milena Cribiù
Giulia Ercoli
Silvano Bosari
Flavio Caprioli
Flavio Caprioli
Federica Facciotti
author_sort Claudia Burrello
title Short-term Oral Antibiotics Treatment Promotes Inflammatory Activation of Colonic Invariant Natural Killer T and Conventional CD4+ T Cells
title_short Short-term Oral Antibiotics Treatment Promotes Inflammatory Activation of Colonic Invariant Natural Killer T and Conventional CD4+ T Cells
title_full Short-term Oral Antibiotics Treatment Promotes Inflammatory Activation of Colonic Invariant Natural Killer T and Conventional CD4+ T Cells
title_fullStr Short-term Oral Antibiotics Treatment Promotes Inflammatory Activation of Colonic Invariant Natural Killer T and Conventional CD4+ T Cells
title_full_unstemmed Short-term Oral Antibiotics Treatment Promotes Inflammatory Activation of Colonic Invariant Natural Killer T and Conventional CD4+ T Cells
title_sort short-term oral antibiotics treatment promotes inflammatory activation of colonic invariant natural killer t and conventional cd4+ t cells
publisher Frontiers Media S.A.
series Frontiers in Medicine
issn 2296-858X
publishDate 2018-02-01
description The gut mucosa is continuously exposed to a vast community of microorganisms, collectively defined as microbiota, establishing a mutualistic relationship with the host and contributing to shape the immune system. Gut microbiota is acquired at birth, and its composition is relatively stable during the entire adult life. Intestinal dysbiosis, defined as a microbial imbalance of gut bacterial communities, can be caused by several factors, including bacterial infections and antibiotic use, and has been associated with an increased risk to develop or exacerbate immune-mediated pathologies, such as allergic reactions, asthma, and inflammatory bowel diseases. Still, the mechanisms by which antibiotic-induced gut dysbiosis may lead to development of mucosal inflammation are still matter of debate. To this end, we aimed to evaluate the impact of antibiotic treatment on phenotype and functions of intestinal immune cell populations, including invariant natural killer T (iNKT) cells, a subset of lipid-specific T cells profoundly influenced by alterations on the commensal microbiota. To this aim, a cocktail of broad-spectrum antibiotics was administered for 2 weeks to otherwise healthy mice before re-colonization of the intestinal microbial community with oral gavage of eubiotic or dysbiotic mucosa-associated bacteria and luminal colonic content, followed or not by intestinal inflammation induction. Here. we showed that short-term antibiotic treatment alters frequency and functions of intestinal iNKT cells, even in the absence of intestinal inflammation. The presence of a dysbiotic microbiota after antibiotic treatment imprints colonic iNKT and CD4+ T cells toward a pro-inflammatory phenotype that collectively contributes to aggravate intestinal inflammation. Nonetheless, the inflammatory potential of the dysbiotic microbiota decreases over time opening the possibility to temporally intervene on the microbial composition to re-equilibrate dysbiosis, thus controlling concomitantly mucosal immune T cell activations.
topic iNKT cells
antibiotics
microbiota
T cells
intestinal inflammation
url http://journal.frontiersin.org/article/10.3389/fmed.2018.00021/full
work_keys_str_mv AT claudiaburrello shorttermoralantibioticstreatmentpromotesinflammatoryactivationofcolonicinvariantnaturalkillertandconventionalcd4tcells
AT claudiaburrello shorttermoralantibioticstreatmentpromotesinflammatoryactivationofcolonicinvariantnaturalkillertandconventionalcd4tcells
AT federicagaravaglia shorttermoralantibioticstreatmentpromotesinflammatoryactivationofcolonicinvariantnaturalkillertandconventionalcd4tcells
AT fulviamilenacribiu shorttermoralantibioticstreatmentpromotesinflammatoryactivationofcolonicinvariantnaturalkillertandconventionalcd4tcells
AT giuliaercoli shorttermoralantibioticstreatmentpromotesinflammatoryactivationofcolonicinvariantnaturalkillertandconventionalcd4tcells
AT silvanobosari shorttermoralantibioticstreatmentpromotesinflammatoryactivationofcolonicinvariantnaturalkillertandconventionalcd4tcells
AT flaviocaprioli shorttermoralantibioticstreatmentpromotesinflammatoryactivationofcolonicinvariantnaturalkillertandconventionalcd4tcells
AT flaviocaprioli shorttermoralantibioticstreatmentpromotesinflammatoryactivationofcolonicinvariantnaturalkillertandconventionalcd4tcells
AT federicafacciotti shorttermoralantibioticstreatmentpromotesinflammatoryactivationofcolonicinvariantnaturalkillertandconventionalcd4tcells
_version_ 1725758100551499776