Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 13 de 13
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Sci Rep ; 12(1): 17591, 2022 10 20.
Article de Anglais | MEDLINE | ID: mdl-36266398

RÉSUMÉ

Live biotherapeutic products constitute an emerging therapeutic approach to prevent or treat inflammatory bowel diseases. Lactobacillus acidophilus is a constituent of the human microbiota with probiotic potential, that is illustrated by improvement of intestinal inflammation and antimicrobial activity against several pathogens. In this study, we evaluated the immunomodulatory properties of the L. acidophilus strain BIO5768 at steady state and upon acute inflammation. Supplementation of naïve mice with BIO5768 heightened the transcript level of some IL-17 target genes encoding for protein with microbicidal activity independently of NOD2 signaling. Of these, the BIO5768-induced expression of Angiogenin-4 was blunted in monocolonized mice that are deficient for the receptor of IL-17 (but not for NOD2). Interestingly, priming of bone marrow derived dendritic cells by BIO5768 enhanced their ability to support the secretion of IL-17 by CD4+ T cells. Equally of importance, the production of IL-22 by type 3 innate lymphoid cells is concomitantly heightened in response to BIO5768. When administered alone or in combination with Bifidobacterium animalis spp. lactis BIO5764 and Limosilactobacillus reuteri, BIO5768 was able to alleviate at least partially intestinal inflammation induced by Citrobacter rodentium infection. Furthermore, BIO5768 was also able to improve colitis induced by 2,4,6-trinitrobenzene sulfonic acid (TNBS). In conclusion, we identify a new potential probiotic strain for the management of inflammatory bowel diseases, and provide some insights into its IL-17-dependent and independent mode of action.


Sujet(s)
Colite , Immunité innée , Maladies inflammatoires intestinales , Lactobacillus acidophilus , Probiotiques , Animaux , Souris , Bifidobacterium animalis , Colite/induit chimiquement , Colite/thérapie , Colite/microbiologie , Infections à Enterobacteriaceae/thérapie , Inflammation , Maladies inflammatoires intestinales/thérapie , Interleukine-17 , Lymphocytes , Probiotiques/pharmacologie , Probiotiques/usage thérapeutique , Acide 2,4,6-trinitro-benzènesulfonique/effets indésirables
2.
Sci Rep ; 10(1): 5345, 2020 03 24.
Article de Anglais | MEDLINE | ID: mdl-32210304

RÉSUMÉ

Crohn's disease is linked to a decreased diversity in gut microbiota composition as a potential consequence of an impaired anti-microbial response and an altered polarization of T helper cells. Here, we evaluated the immunomodulatory properties of two potential probiotic strains, namely a Bifidobacterium animalis spp. lactis Bl 5764 and a Lactobacillus reuteri Lr 5454 strains. Both strains improved colitis triggered by either 2,4,6-trinitrobenzenesulfonic acid (TNBS) or Citrobacter rodentium infection in mice. Training of dendritic cells (DC) with Lr 5454 efficiently triggered IL-22 secretion and regulatory T cells induction in vitro, while IL-17A production by CD4+ T lymphocytes was stronger when cultured with DCs that were primed with Bl 5764. This strain was sufficient for significantly inducing expression of antimicrobial peptides in vivo through the Crohn's disease predisposing gene encoding for the nucleotide-binding oligomerization domain, containing protein 2 (NOD2). In contrast, NOD2 was dispensable for the impact on antimicrobial peptide expression in mice that were monocolonized with Lr 5454. In conclusion, our work highlights a differential mode of action of two potential probiotic strains that protect mice against colitis, providing the rational for a personalized supportive preventive therapy by probiotics for individuals that are genetically predisposed to Crohn's disease.


Sujet(s)
Bifidobacterium animalis , Colite/microbiologie , Colite/thérapie , Cellules dendritiques/physiologie , Limosilactobacillus reuteri , Probiotiques/pharmacologie , Animaux , Anti-inflammatoires non stéroïdiens/pharmacologie , Citrobacter rodentium/pathogénicité , Colite/induit chimiquement , Colite/anatomopathologie , Modèles animaux de maladie humaine , Infections à Enterobacteriaceae/microbiologie , Femelle , Microbiome gastro-intestinal , Axénie , Souris de lignée BALB C , Souris de lignée C57BL , Souris knockout , Protéines associées à la pancréatite/génétique , Lymphocytes T auxiliaires/physiologie , Lymphocytes T régulateurs/physiologie , Acide 2,4,6-trinitro-benzènesulfonique/toxicité
3.
Front Immunol ; 11: 144, 2020.
Article de Anglais | MEDLINE | ID: mdl-32161582

RÉSUMÉ

Airborne ozone exposure causes severe lung injury and inflammation. The aryl hydrocarbon Receptor (AhR) (1), activated in pollutant-induced inflammation, is critical for cytokine production, especially IL-22 and IL-17A. The role of AhR in ozone-induced lung inflammation is unknown. We report here that chronic ozone exposure activates AhR with increased tryptophan and lipoxin A4 production in mice. AhR-/- mice show increased lung inflammation, airway hyperresponsiveness, and tissue remodeling with an increased recruitment of IL-17A and IL-22-expressing cells in comparison to control mice. IL-17A- and IL-22-neutralizing antibodies attenuate lung inflammation in AhR-/- and control mice. Enhanced lung inflammation and recruitment of ILC3, ILC2, and T cells were observed after T cell-specific AhR depletion using the AhRCD4cre-deficient mice. Together, the data demonstrate that ozone exposure activates AhR, which controls lung inflammation, airway hyperresponsiveness, and tissue remodeling via the reduction of IL-22 expression.


Sujet(s)
Facteurs de transcription à motif basique hélice-boucle-hélice/métabolisme , Interleukines/métabolisme , Lésion pulmonaire/induit chimiquement , Lésion pulmonaire/métabolisme , Ozone/effets indésirables , Pneumopathie infectieuse/induit chimiquement , Pneumopathie infectieuse/métabolisme , Récepteurs à hydrocarbure aromatique/métabolisme , Hypersensibilité respiratoire/induit chimiquement , Hypersensibilité respiratoire/métabolisme , Animaux , Anticorps neutralisants/immunologie , Anticorps neutralisants/usage thérapeutique , Facteurs de transcription à motif basique hélice-boucle-hélice/génétique , Lymphocytes T CD4+/immunologie , Interleukine-17/immunologie , Interleukine-17/métabolisme , Interleukines/génétique , Interleukines/immunologie , Lipoxines/métabolisme , Lésion pulmonaire/traitement médicamenteux , Souris , Souris de lignée C57BL , Souris knockout , Pneumopathie infectieuse/traitement médicamenteux , Récepteurs à hydrocarbure aromatique/génétique , Récepteurs à l'interleukine-17/génétique , Hypersensibilité respiratoire/traitement médicamenteux , Tryptophane/métabolisme ,
4.
Nat Commun ; 9(1): 5338, 2018 12 17.
Article de Anglais | MEDLINE | ID: mdl-30559449

RÉSUMÉ

Mutations in the nucleotide-binding oligomerization domain protein 12 (NLRP12) cause recurrent episodes of serosal inflammation. Here we show that NLRP12 efficiently sequesters HSP90 and promotes K48-linked ubiquitination and degradation of NOD2 in response to bacterial muramyl dipeptide (MDP). This interaction is mediated by the linker-region proximal to the nucleotide-binding domain of NLRP12. Consequently, the disease-causing NLRP12 R284X mutation fails to repress MDP-induced NF-κB and subsequent activity of the JAK/STAT signaling pathway. While NLRP12 deficiency renders septic mice highly susceptible towards MDP, a sustained sensing of MDP through NOD2 is observed among monocytes lacking NLRP12. This loss of tolerance in monocytes results in greater colonization resistance towards Citrobacter rodentium. Our data show that this is a consequence of NOD2-dependent accumulation of inflammatory mononuclear cells that correlates with induction of interferon-stimulated genes. Our study unveils a relevant process of tolerance towards the gut microbiota that is exploited by an attaching/effacing enteric pathogen.


Sujet(s)
Acétylmuramyl alanyl isoglutamine/métabolisme , Capsules bactériennes/métabolisme , Citrobacter rodentium/immunologie , Infections à Enterobacteriaceae/immunologie , Protéines du choc thermique HSP90/métabolisme , Tolérance immunitaire/immunologie , Protéines et peptides de signalisation intracellulaire/métabolisme , Protéine adaptatrice de signalisation NOD2/métabolisme , Animaux , Lignée cellulaire , Infections à Enterobacteriaceae/microbiologie , Microbiome gastro-intestinal/immunologie , Cellules HEK293 , Humains , Inflammation/immunologie , Inflammation/microbiologie , Protéines et peptides de signalisation intracellulaire/génétique , Mâle , Souris , Souris knockout , Facteur de transcription NF-kappa B/métabolisme , Ubiquitination
5.
Sci Rep ; 8(1): 11245, 2018 07 26.
Article de Anglais | MEDLINE | ID: mdl-30050168

RÉSUMÉ

Allergic asthma is characterized by a strong Th2 and Th17 response with inflammatory cell recruitment, airways hyperreactivity and structural changes in the lung. The protease allergen papain disrupts the airway epithelium triggering a rapid eosinophilic inflammation by innate lymphoid cell type 2 (ILC2) activation, leading to a Th2 immune response. Here we asked whether the daily oral administrations of the probiotic Escherichia coli strain Nissle 1917 (ECN) might affect the outcome of the papain protease induced allergic lung inflammation in BL6 mice. We find that ECN gavage significantly prevented the severe allergic response induced by repeated papain challenges and reduced lung inflammatory cell recruitment, Th2 and Th17 response and respiratory epithelial barrier disruption with emphysema and airway hyperreactivity. In conclusion, ECN administration attenuated severe protease induced allergic inflammation, which may be beneficial to prevent allergic asthma.


Sujet(s)
Allergènes/administration et posologie , Asthme/prévention et contrôle , Escherichia coli/croissance et développement , Facteurs immunologiques/administration et posologie , Papaïne/administration et posologie , Probiotiques/administration et posologie , Administration par voie orale , Animaux , Asthme/induit chimiquement , Asthme/anatomopathologie , Modèles animaux de maladie humaine , Souris , Souris de lignée C57BL , Muqueuse respiratoire/anatomopathologie , Cellules Th17/immunologie , Lymphocytes auxiliaires Th2/immunologie , Résultat thérapeutique
6.
Eur J Immunol ; 46(11): 2531-2541, 2016 11.
Article de Anglais | MEDLINE | ID: mdl-27569535

RÉSUMÉ

Allergic asthma is characterized by a strong Th2 response with inflammatory cell recruitment and structural changes in the lung. Papain is a protease allergen disrupting the airway epithelium triggering a rapid inflammation with eosinophilia mediated by innate lymphoid cell activation (ILC2) and leading to a Th2 immune response. In this study, we focused on inflammatory responses to a single exposure to papain and showed that intranasal administration of papain results in the recruitment of inflammatory cells, including neutrophils and eosinophils with a rapid production of IL-1α, IL-1ß, and IL-33. The inflammatory response is abrogated in the absence of IL-1R1 and MyD88. To decipher the cell type(s) involved in MyD88-dependent IL-1R1/MyD88 signaling, we used new cell-specific MyD88-deficient mice and found that the deletion of MyD88 signaling in single cell types such as T cells, epithelial cells, CD11c-positive or myeloid cells leads to only a partial inhibition compared to complete absence of MyD88, suggesting that several cell types contribute to the response. Importantly, the inflammatory response is largely ST2 and IL-36R independent. In conclusion, IL-1R1 signaling via MyD88 is critical for the first step of inflammatory response to papain.


Sujet(s)
Allergènes/immunologie , Immunité innée , Poumon/immunologie , Facteur de différenciation myéloïde-88/métabolisme , Papaïne/immunologie , Pneumopathie infectieuse/immunologie , Récepteur à l'interleukine-1 de type I/métabolisme , Allergènes/administration et posologie , Animaux , Granulocytes éosinophiles/immunologie , Interleukine-1 alpha/métabolisme , Interleukine-1 bêta/métabolisme , Interleukine-33/métabolisme , Poumon/physiopathologie , Souris , Facteur de différenciation myéloïde-88/déficit , Facteur de différenciation myéloïde-88/génétique , Facteur de différenciation myéloïde-88/immunologie , Granulocytes neutrophiles/immunologie , Papaïne/administration et posologie , Récepteurs à l'interleukine-1/immunologie , Récepteurs à l'interleukine-1/métabolisme , Récepteur à l'interleukine-1 de type I/immunologie , Transduction du signal , Lymphocytes auxiliaires Th2/immunologie
7.
Am J Clin Exp Immunol ; 5(1): 33-40, 2016.
Article de Anglais | MEDLINE | ID: mdl-27168953

RÉSUMÉ

Exposure to ambient ozone causes airway hyperreactivity and lung inflammation, which represent an important health concern in humans. Recent clinical and experimental studies contributed to the understanding of the mechanisms of epithelial injury, inflammation and airway hyperreactivity, which is reviewed here. The present data suggest that ozone induced oxidative stress causes inflammasome activation with the release of IL-1, other cytokines and proteases driving lung inflammation leading to the destruction of alveolar epithelia with emphysema and respiratory failure. Insights in the pathogenic pathway may allow to identify novel biomarkers of ozone-induced lung disease and therapeutic targets.

8.
Nat Med ; 22(6): 598-605, 2016 06.
Article de Anglais | MEDLINE | ID: mdl-27158904

RÉSUMÉ

Complex interactions between the host and the gut microbiota govern intestinal homeostasis but remain poorly understood. Here we reveal a relationship between gut microbiota and caspase recruitment domain family member 9 (CARD9), a susceptibility gene for inflammatory bowel disease (IBD) that functions in the immune response against microorganisms. CARD9 promotes recovery from colitis by promoting interleukin (IL)-22 production, and Card9(-/-) mice are more susceptible to colitis. The microbiota is altered in Card9(-/-) mice, and transfer of the microbiota from Card9(-/-) to wild-type, germ-free recipients increases their susceptibility to colitis. The microbiota from Card9(-/-) mice fails to metabolize tryptophan into metabolites that act as aryl hydrocarbon receptor (AHR) ligands. Intestinal inflammation is attenuated after inoculation of mice with three Lactobacillus strains capable of metabolizing tryptophan or by treatment with an AHR agonist. Reduced production of AHR ligands is also observed in the microbiota from individuals with IBD, particularly in those with CARD9 risk alleles associated with IBD. Our findings reveal that host genes affect the composition and function of the gut microbiota, altering the production of microbial metabolites and intestinal inflammation.


Sujet(s)
Protéines adaptatrices de signalisation CARD/immunologie , Colite/immunologie , Microbiome gastro-intestinal/immunologie , Interleukines/immunologie , Lactobacillus/métabolisme , Récepteurs à hydrocarbure aromatique/immunologie , Tryptophane/métabolisme , Adolescent , Adulte , Animaux , Protéines adaptatrices de signalisation CARD/génétique , Chromatographie en phase liquide à haute performance , Colite/induit chimiquement , Colite/anatomopathologie , Côlon/immunologie , Côlon/microbiologie , Côlon/anatomopathologie , Cytokines/immunologie , Sulfate dextran/toxicité , Transplantation de microbiote fécal , Femelle , Microbiome gastro-intestinal/génétique , Analyse de profil d'expression de gènes , Humains , Maladies inflammatoires intestinales/génétique , Maladies inflammatoires intestinales/immunologie , Mâle , Souris , Souris knockout , Adulte d'âge moyen , ARN ribosomique 16S/génétique , RT-PCR , Tryptophane/immunologie , Jeune adulte ,
9.
Mol Med ; 21(1): 1025-1037, 2016 Mar.
Article de Anglais | MEDLINE | ID: mdl-26623925

RÉSUMÉ

Although the molecular links underlying the causative relationship between chronic low-grade inflammation and insulin resistance are not completely understood, compelling evidence suggests a pivotal role of the nucleotide-binding oligomerization domain (NOD)-like receptor pyrin domain containing 3 (NLRP3) inflammasome. Here we tested the hypothesis that either a selective pharmacological inhibition or a genetic downregulation of the NLRP3 inflammasome results in reduction of the diet-induced metabolic alterations. Male C57/BL6 wild-type mice and NLRP3-/- littermates were fed control diet or high-fat, high-fructose diet (HD). A subgroup of HD-fed wild-type mice was treated with the NLRP3 inflammasome inhibitor BAY 11-7082 (3 mg/kg intraperitoneally [IP]). HD feeding increased plasma and hepatic lipids and impaired glucose homeostasis and renal function. Renal and hepatic injury was associated with robust increases in profibrogenic markers, while only minimal fibrosis was recorded. None of these metabolic abnormalities were detected in HD-fed NLRP3-/- mice, and they were dramatically reduced in HD-mice treated with the NLRP3 inflammasome inhibitor. BAY 11-7082 also attenuated the diet-induced increase in NLRP3 inflammasome expression, resulting in inhibition of caspase-1 activation and interleukin (IL)-1ß and IL-18 production (in liver and kidney). Interestingly, BAY 11-7082, but not gene silencing, inhibited nuclear factor (NF)-κB nuclear translocation. Overall, these results demonstrate that the selective pharmacological modulation of the NLRP3 inflammasome attenuates the metabolic abnormalities and the related organ injury/dysfunction caused by chronic exposure to HD, with effects similar to those obtained by NLRP3 gene silencing.

10.
Eur J Immunol ; 45(5): 1354-65, 2015 May.
Article de Anglais | MEDLINE | ID: mdl-25682948

RÉSUMÉ

Cerebral malaria, a severe complication of Plasmodium falciparum infection, can be modeled in murine Plasmodium berghei ANKA (PbA) infection. PbA-induced experimental cerebral malaria (ECM) is CD8(+) T-cell mediated, and influenced by TH 1/TH 2 balance. Here, we show that IL-33 expression is increased in brain undergoing ECM and we address the role of the IL-33/ST2 pathway in ECM development. ST2-deficient mice were resistant to PbA-induced neuropathology. They survived >20 days with no ECM neurological sign and a preserved cerebral microcirculation, while WT mice succumbed within 10 days with ECM, brain vascular leakage, distinct microvascular pathology obstruction, and hemorrhages. Parasitemia and brain parasite load were similar in ST2-deficient and WT mice. Protection was accompanied by reduced brain sequestration of activated CD4(+) T cells and perforin(+) CD8(+) T cells. While IFN-γ and T-cell-attracting chemokines CXCL9 and CXCL10 were not affected in the absence of functional ST2 pathway, the local expression of ICAM-1, CXCR3, and LT-α, crucial for ECM development, was strongly reduced, and this may explain the diminished pathogenic T-cell recruitment and resistance to ECM. Therefore, IL-33 is induced in PbA sporozoite infection, and the pathogenic T-cell responses with local microvascular pathology are dependent on IL-33/ST2 signaling, identifying IL-33 as a new actor in ECM development.


Sujet(s)
Paludisme cérébral/étiologie , Plasmodium berghei , Récepteurs aux interleukines/métabolisme , Animaux , Encéphale/immunologie , Encéphale/parasitologie , Encéphale/anatomopathologie , Lymphocytes T CD4+/immunologie , Lymphocytes T CD4+/anatomopathologie , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/anatomopathologie , Modèles animaux de maladie humaine , Femelle , Inflammation/étiologie , Inflammation/immunologie , Inflammation/anatomopathologie , Protéine-1 analogue au récepteur de l'interleukin-1 , Interleukine-33 , Interleukines/métabolisme , Activation des lymphocytes , Paludisme cérébral/immunologie , Paludisme cérébral/parasitologie , Souris , Souris de lignée C57BL , Souris knockout , Plasmodium berghei/immunologie , Plasmodium berghei/pathogénicité , Récepteurs aux interleukines/déficit , Récepteurs aux interleukines/génétique
11.
Am J Respir Crit Care Med ; 189(7): 799-811, 2014 Apr 01.
Article de Anglais | MEDLINE | ID: mdl-24555512

RÉSUMÉ

RATIONALE: Pseudomonas aeruginosa, a major problem pathogen responsible for severe infections in critically ill patients, triggers, through a functional type-3 secretion system (T3SS), the activation of an intracellular cytosolic sensor of innate immunity, NLRC4. Although the NLRC4-inflammasome-dependent response contributes to increased clearance of intracellular pathogens, it seems that NLRC4 inflammasome activation decreases the clearance of P. aeruginosa, a mainly extracellular pathogen. OBJECTIVES: We sought to determine the underlying mechanisms of this effect of the activation of NLRC4 by P. aeruginosa. METHODS: We established acute lung injury in wild-type and Nlrc4(-/-) mice using sublethal intranasal inocula of P. aeruginosa strain CHA expressing or not a functional T3SS. We studied 96-hour survival, lung injury, bacterial clearance from the lungs, cytokine secretion in bronchoalveolar lavage, lung antimicrobial peptide expression by quantitative polymerase chain reaction, and flow cytometry analysis of lung cells. MEASUREMENTS AND MAIN RESULTS: Nlrc4(-/-) mice showed enhanced bacterial clearance and decreased lung injury contributing to increased survival against extracellular P. aeruginosa strain expressing a functional T3SS. The mechanism involved decreased NLRC4-inflammasome-driven IL-18 secretion attenuating lung injury caused by excessive neutrophil recruitment. Additionally, in the lungs of Nlrc4(-/-) mice secretion of IL-17 by innate immune cells was increased and responsible for increased expression of lung epithelial antimicrobial peptides. Furthermore, IL-18 secretion was found to repress IL-17 and IL-17-driven lung antimicrobial peptide expression. CONCLUSIONS: We report a new role of the T3SS apparatus itself, independently of exotoxin translocation. Through NLRC4 inflammasome activation, the T3SS promotes IL-18 secretion, which dampens a beneficial IL-17-mediated antimicrobial host response.


Sujet(s)
Lésion pulmonaire aigüe/microbiologie , Protéines régulatrices de l'apoptose/métabolisme , Protéines de liaison au calcium/métabolisme , Inflammasomes/métabolisme , Infections à Pseudomonas/microbiologie , Pseudomonas aeruginosa/pathogénicité , Lésion pulmonaire aigüe/immunologie , Lésion pulmonaire aigüe/métabolisme , Animaux , Marqueurs biologiques/métabolisme , Liquide de lavage bronchoalvéolaire/composition chimique , Caspase-1/métabolisme , Cellules cultivées , Femelle , Cytométrie en flux , Immunité innée , Interleukine-17/métabolisme , Interleukine-18/métabolisme , Macrophages/immunologie , Macrophages/métabolisme , Macrophages/microbiologie , Mâle , Souris , Souris de lignée C57BL , Souris knockout , Infections à Pseudomonas/immunologie , Infections à Pseudomonas/métabolisme , Pseudomonas aeruginosa/métabolisme , RT-PCR
12.
Am J Clin Exp Immunol ; 2(1): 107-16, 2013.
Article de Anglais | MEDLINE | ID: mdl-23885328

RÉSUMÉ

Interleukin 1 is a critical inflammatory mediator and involved in host defense to several pathogens. Oral T. gondii infection causes lethal ileitis in C57BL/6 (BL6) mice and serves to investigate the mechanisms of acute intestinal inflammation. Here we show that IL-1 is expressed upon oral T. gondii (76K strain) infection in the small intestine and mediates ileitis as IL-1R1 deficient mice have reduced neutrophil recruitment in the lamina propria, parasite invasion, inflammatory lesions and enhanced survival as compared to BL6 infected control mice. Protection in the absence of IL-1R1 signaling was associated with reduced IFN-γ expression and preserved Paneth cells, while these cells were eliminated in infected BL6 mice. Furthermore, blockade of IL-1 by IL-1ß antibody attenuated inflammation in BL6 mice. In conclusion, IL-1 signaling contributes to the inflammatory response with increase IFN-γ expression and Paneth cell depletion upon oral T. gondii infection.

13.
J Clin Invest ; 123(2): 700-11, 2013 Feb.
Article de Anglais | MEDLINE | ID: mdl-23281400

RÉSUMÉ

Instability in the composition of gut bacterial communities (dysbiosis) has been linked to common human intestinal disorders, such as Crohn's disease and colorectal cancer. Here, we show that dysbiosis caused by Nod2 deficiency gives rise to a reversible, communicable risk of colitis and colitis-associated carcinogenesis in mice. Loss of either Nod2 or RIP2 resulted in a proinflammatory microenvironment that enhanced epithelial dysplasia following chemically induced injury. The condition could be improved by treatment with antibiotics or an anti-interleukin-6 receptor-neutralizing antibody. Genotype-dependent disease risk was communicable via maternally transmitted microbiota in both Nod2-deficient and WT hosts. Furthermore, reciprocal microbiota transplantation reduced disease risk in Nod2-deficient mice and led to long-term changes in intestinal microbial communities. Conversely, disease risk was enhanced in WT hosts that were recolonized with dysbiotic fecal microbiota from Nod2-deficient mice. Thus, we demonstrated that licensing of dysbiotic microbiota is a critical component of disease risk. Our results demonstrate that NOD2 has an unexpected role in shaping a protective assembly of gut bacterial communities and suggest that manipulation of dysbiosis is a potential therapeutic approach in the treatment of human intestinal disorders.


Sujet(s)
Colite/étiologie , Tumeurs colorectales/étiologie , Protéine adaptatrice de signalisation NOD2/déficit , Animaux , Colite/métabolisme , Colite/microbiologie , Colite/anatomopathologie , Tumeurs colorectales/métabolisme , Tumeurs colorectales/microbiologie , Tumeurs colorectales/anatomopathologie , Système digestif/métabolisme , Système digestif/microbiologie , Système digestif/anatomopathologie , Modèles animaux de maladie humaine , Femelle , Humains , Mâle , Métagénome , Souris , Souris knockout , Protéine adaptatrice de signalisation NOD2/génétique , Grossesse , Receptor-Interacting Protein Serine-Threonine Kinase 2 , Receptor-Interacting Protein Serine-Threonine Kinases/déficit , Receptor-Interacting Protein Serine-Threonine Kinases/génétique , Facteurs de risque
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE