Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Rheumatology (Oxford) ; 63(4): 1039-1048, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-37402619

ABSTRACT

OBJECTIVE: FMF is the most common monogenic autoinflammatory disease associated with MEFV mutations. Disease phenotype and response to treatment vary from one patient to another, despite similar genotype, suggesting the role of environmental factors. The objective of this study was to analyse the gut microbiota of a large cohort of FMF patients in relation to disease characteristics. METHODS: The gut microbiotas of 119 FMF patients and 61 healthy controls were analysed using 16 s rRNA gene sequencing. Associations between bacterial taxa, clinical characteristics, and genotypes were evaluated using multivariable association with linear models (MaAslin2), adjusting on age, sex, genotype, presence of AA amyloidosis (n = 17), hepatopathy (n = 5), colchicine intake, colchicine resistance (n = 27), use of biotherapy (n = 10), CRP levels, and number of daily faeces. Bacterial network structures were also analysed. RESULTS: The gut microbiotas of FMF patients differ from those of controls in having increased pro-inflammatory bacteria, such as the Enterobacter, Klebsiella and Ruminococcus gnavus group. Disease characteristics and resistance to colchicine correlated with homozygous mutations and were associated with specific microbiota alteration. Colchicine treatment was associated with the expansion of anti-inflammatory taxa such as Faecalibacterium and Roseburia, while FMF severity was associated with expansion of the Ruminococcus gnavus group and Paracoccus. Colchicine-resistant patients exhibited an alteration of the bacterial network structure, with decreased intertaxa connectivity. CONCLUSION: The gut microbiota of FMF patients correlates with disease characteristics and severity, with an increase in pro-inflammatory taxa in the most severe patients. This suggests a specific role for the gut microbiota in shaping FMF outcomes and response to treatment.


Subject(s)
Clostridiales , Familial Mediterranean Fever , Gastrointestinal Microbiome , Humans , Familial Mediterranean Fever/drug therapy , Familial Mediterranean Fever/genetics , Familial Mediterranean Fever/complications , Gastrointestinal Microbiome/genetics , Genotype , Colchicine/therapeutic use , Phenotype , Mutation , Pyrin/genetics
2.
Front Immunol ; 14: 1224383, 2023.
Article in English | MEDLINE | ID: mdl-38146368

ABSTRACT

Chronic obstructive pulmonary disease (COPD) is a major health issue primarily caused by cigarette smoke (CS) and characterized by breathlessness and repeated airway inflammation. NLRP6 is a cytosolic innate receptor controlling intestinal inflammation and orchestrating the colonic host-microbial interface. However, its roles in the lungs remain largely unexplored. Using CS exposure models, our data show that airway inflammation is strongly impaired in Nlrp6-deficient mice with drastically fewer recruited neutrophils, a key cell subset in inflammation and COPD. We found that NLRP6 expression in lung epithelial cells is important to control airway and lung tissue inflammation in an inflammasome-dependent manner. Since gut-derived metabolites regulate NLRP6 inflammasome activation in intestinal epithelial cells, we investigated the link between NLRP6, CS-driven lung inflammation, and gut microbiota composition. We report that acute CS exposure alters gut microbiota in both wild-type (WT) and Nlrp6-deficient mice and that antibiotic treatment decreases CS-induced lung inflammation. In addition, gut microbiota transfer from dysbiotic Nlrp6-deficient mice to WT mice decreased airway lung inflammation in WT mice, highlighting an NLRP6-dependent gut-to-lung axis controlling pulmonary inflammation.


Subject(s)
Gastrointestinal Microbiome , Pneumonia , Receptors, Cell Surface , Tobacco Smoke Pollution , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Pneumonia/chemically induced , Pneumonia/genetics , Pneumonia/microbiology , Animals , Mice , Mice, Inbred C57BL , Cells, Cultured , Epithelial Cells/cytology , Epithelial Cells/pathology , Feces/microbiology , Bacteria/classification , Bacteria/metabolism , Biodiversity , Gene Expression
3.
Biomed Pharmacother ; 163: 114813, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37150031

ABSTRACT

BACKGROUND: Lipin-1 deficiency is a life-threatening disease that causes severe rhabdomyolysis (RM) and chronic symptoms associated with oxidative stress. In the absence of treatment, Hydroxychloroquine sulfate (HCQ) was administered to patients off label use on a compassionate basis in order to improve their physical conditions. METHODS: Eleven patients with LPIN1 mutations were treated with HCQ. Clinical and biological efficacy and tolerance were assessed, including pain and quality of life, physical capacities, cardiopulmonary parameters, creatine kinase levels and plasma proinflammatory cytokines. To explore a dose-dependent effect of HCQ, primary myoblasts from 4 patients were incubated with various HCQ concentrations in growth medium (GM) or during starvation (EBSS medium) to investigate autophagy and oxidative stress. FINDINGS: Under HCQ treatment, patient physical capacities improved. Abnormal cardiac function and peripheral muscle adaptation to exercise were normalized. However, two patients who had the highest mean blood HCQ concentrations experienced RM. We hypothesized that HCQ exerts deleterious effects at high concentrations by blocking autophagy, and beneficial effects on oxidative stress at low concentrations. We confirmed in primary myoblasts from 4 patients that high in vitro HCQ concentration (10 µM) but not low concentration (1 µM and 0.1 µM) induced autophagy blockage by modifying endolysosomal pH. Low HCQ concentration (1 µM) prevented reactive oxygen species (ROS) and oxidized DNA accumulation in myoblasts during starvation. INTERPRETATION: HCQ improves the condition of patients with lipin-1 deficiency, but at low concentrations. In vitro, 1 µM HCQ decreases oxidative stress in myoblasts whereas higher concentrations have a deleterious effect by blocking autophagy.


Subject(s)
Hydroxychloroquine , Quality of Life , Humans , Hydroxychloroquine/pharmacology , Hydroxychloroquine/therapeutic use , Cytokines , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Phosphatidate Phosphatase/genetics
4.
JCI Insight ; 7(12)2022 06 22.
Article in English | MEDLINE | ID: mdl-35536673

ABSTRACT

Abundance of Faecalibacterium prausnitzii, a dominant bacterium of the human microbiota that exhibits antiinflammatory effects, is decreased in patients with inflammatory bowel diseases (IBD). In humans, colonic lamina propria contains IL-10-secreting, Foxp3- Tregs characterized by a double expression of CD4 and CD8α (DP8α) and a specificity for F. prausnitzii. This Treg subset is decreased in IBD. The in vivo effect of DP8α cells has not been evaluated yet to our knowledge. Here, using a humanized model of a NSG immunodeficient mouse strain that expresses the HLA D-related allele HLA-DR*0401 but not murine class II (NSG-Ab° DR4) molecules, we demonstrated a protective effect of a HLA-DR*0401-restricted DP8α Treg clone combined with F. prausnitzii administration in a colitis model. In a cohort of patients with IBD, we showed an independent association between the frequency of circulating DP8α cells and disease activity. Finally, we pointed out a positive correlation between F. prausnitzii-specific DP8α Tregs and the amount of F. prausnitzii in fecal microbiota in healthy individuals and patients with ileal Crohn's disease.


Subject(s)
Colitis , Faecalibacterium prausnitzii , Inflammatory Bowel Diseases , T-Lymphocytes, Regulatory , Animals , Colitis/immunology , Humans , Inflammation , Inflammatory Bowel Diseases/immunology , Mice , T-Lymphocytes, Regulatory/immunology
5.
Gut Microbes ; 14(1): 2022997, 2022.
Article in English | MEDLINE | ID: mdl-35090380

ABSTRACT

Alterations in brain/gut/microbiota axis are linked to Irritable Bowel Syndrome (IBS) physiopathology. Upon gastrointestinal infection, chronic abdominal pain and anxio-depressive comorbidities may persist despite pathogen clearance leading to Post-Infectious IBS (PI-IBS). This study assesses the influence of tryptophan metabolism, and particularly the microbiota-induced AhR expression, on intestinal homeostasis disturbance following gastroenteritis resolution, and evaluates the efficacy of IL-22 cytokine vectorization on PI-IBS symptoms. The Citrobacter rodentium infection model in C57BL6/J mice was used to mimic Enterobacteria gastroenteritis. Intestinal homeostasis was evaluated as low-grade inflammation, permeability, mucosa-associated microbiota composition, and colonic sensitivity. Cognitive performances and emotional state of animals were assessed using several tests. Tryptophan metabolism was analyzed by targeted metabolomics. AhR activity was evaluated using a luciferase reporter assay method. One Lactococcus lactis strain carrying an eukaryotic expression plasmid for murine IL-22 (L. lactisIL-22) was used to induce IL-22 production in mouse colonic mucosa. C. rodentium-infected mice exhibited persistent colonic hypersensitivity and cognitive impairments and anxiety-like behaviors after pathogen clearance. These post-infectious disorders were associated with low-grade inflammation, increased intestinal permeability, decrease of Lactobacillaceae abundance associated with the colonic layer, and increase of short-chain fatty acids (SCFAs). During post-infection period, the indole pathway and AhR activity were decreased due to a reduction of tryptophol production. Treatment with L. lactisIL-22 restored gut permeability and normalized colonic sensitivity, restored cognitive performances and decreased anxiety-like behaviors. Data from the video-tracking system suggested an upgrade of welfare for mice receiving the L.lactisIL-22 strain. Our findings revealed that AhR/IL-22 signaling pathway is altered in a preclinical PI-IBS model. IL-22 delivering alleviate PI-IBS symptoms as colonic hypersensitivity, cognitive impairments, and anxiety-like behaviors by acting on intestinal mucosa integrity. Thus, therapeutic strategies targeting this pathway could be developed to treat IBS patients suffering from chronic abdominal pain and associated well-being disorders.


Subject(s)
Depression/etiology , Interleukins/metabolism , Irritable Bowel Syndrome/complications , Irritable Bowel Syndrome/psychology , Receptors, Aryl Hydrocarbon/metabolism , Animals , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Bacteria/metabolism , Cognition , Depression/genetics , Depression/metabolism , Depression/psychology , Fatty Acids, Volatile/metabolism , Gastrointestinal Microbiome , Humans , Interleukins/genetics , Intestines/metabolism , Intestines/microbiology , Irritable Bowel Syndrome/metabolism , Irritable Bowel Syndrome/microbiology , Mice , Mice, Inbred C57BL , Receptors, Aryl Hydrocarbon/genetics , Interleukin-22
6.
Gut ; 70(7): 1299-1308, 2021 07.
Article in English | MEDLINE | ID: mdl-33004548

ABSTRACT

OBJECTIVE: Chronic alcohol consumption is an important cause of liver-related deaths. Specific intestinal microbiota profiles are associated with susceptibility or resistance to alcoholic liver disease in both mice and humans. We aimed to identify the mechanisms by which targeting intestinal microbiota can improve alcohol-induced liver lesions. DESIGN: We used human associated mice, a mouse model of alcoholic liver disease transplanted with the intestinal microbiota of alcoholic patients and used the prebiotic, pectin, to modulate the intestinal microbiota. Based on metabolomic analyses, we focused on microbiota tryptophan metabolites, which are ligands of the aryl hydrocarbon receptor (AhR). Involvement of the AhR pathway was assessed using both a pharmacological approach and AhR-deficient mice. RESULTS: Pectin treatment modified the microbiome and metabolome in human microbiota-associated alcohol-fed mice, leading to a specific faecal signature. High production of bacterial tryptophan metabolites was associated with an improvement of liver injury. The AhR agonist Ficz (6-formylindolo (3,2-b) carbazole) reduced liver lesions, similarly to prebiotic treatment. Conversely, inactivation of the ahr gene in alcohol-fed AhR knock-out mice abrogated the beneficial effects of the prebiotic. Importantly, patients with severe alcoholic hepatitis have low levels of bacterial tryptophan derivatives that are AhR agonists. CONCLUSIONS: Improvement of alcoholic liver disease by targeting the intestinal microbiota involves the AhR pathway, which should be considered as a new therapeutic target.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Intestines/microbiology , Liver Diseases, Alcoholic/etiology , Microbiota/physiology , Pectins/pharmacology , Receptors, Aryl Hydrocarbon/metabolism , Tryptophan/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/agonists , Basic Helix-Loop-Helix Transcription Factors/genetics , Carbazoles/pharmacology , Disease Models, Animal , Fecal Microbiota Transplantation , Feces/chemistry , Female , Humans , Intestines/physiopathology , Liver Diseases, Alcoholic/drug therapy , Liver Diseases, Alcoholic/metabolism , Metabolome/drug effects , Mice , Mice, Knockout , Microbiota/drug effects , Pectins/therapeutic use , Prebiotics , Receptors, Aryl Hydrocarbon/agonists , Receptors, Aryl Hydrocarbon/genetics
7.
Front Immunol ; 11: 144, 2020.
Article in English | MEDLINE | ID: mdl-32161582

ABSTRACT

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.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Interleukins/metabolism , Lung Injury/chemically induced , Lung Injury/metabolism , Ozone/adverse effects , Pneumonia/chemically induced , Pneumonia/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Respiratory Hypersensitivity/chemically induced , Respiratory Hypersensitivity/metabolism , Animals , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/therapeutic use , Basic Helix-Loop-Helix Transcription Factors/genetics , CD4-Positive T-Lymphocytes/immunology , Interleukin-17/immunology , Interleukin-17/metabolism , Interleukins/genetics , Interleukins/immunology , Lipoxins/metabolism , Lung Injury/drug therapy , Mice , Mice, Inbred C57BL , Mice, Knockout , Pneumonia/drug therapy , Receptors, Aryl Hydrocarbon/genetics , Receptors, Interleukin-17/genetics , Respiratory Hypersensitivity/drug therapy , Tryptophan/metabolism , Interleukin-22
8.
Front Immunol ; 11: 117, 2020.
Article in English | MEDLINE | ID: mdl-32117268

ABSTRACT

Individuals with impaired immune responses, such as ventilated and cystic fibrosis patients are often infected with Pseudomonas aeruginosa (P.a) bacteria, and a co-infection with the Influenza virus (IAV) is often present. It has been known for many years that infection with IAV predisposes the host to secondary bacterial infections (such as Streptococcus pneumoniae or Staphylococcus aureus), and there is an abundance of mechanistic studies, including those studying the role of desensitization of TLR signaling, type I IFN- mediated impairment of neutrophil chemokines and antimicrobial production, attenuation of IL1ß production etc., showing this. However, little is known about the mechanistic events underlying the potential deleterious synergy between Influenza and P.a co-infections. We demonstrate here in vitro in epithelial cells and in vivo in three independent models (two involving mice given IAV +/- P.a, and one involving mice given IAV +/- IL-1ß) that IAV promotes secondary P.a-mediated lung disease or augmented IL-1ß-mediated inflammation. We show that IAV-P.a-mediated deleterious responses includes increased matrix metalloprotease (MMP) activity, and MMP-9 in particular, and that the use of the MMP inhibitor improves lung resilience. Furthermore, we show that IAV post-transcriptionally inhibits the antimicrobial/anti-protease molecule elafin/trappin-2, which we have shown previously to be anti-inflammatory and to protect the host against maladaptive neutrophilic inflammation in P.a infections. Our work highlights the capacity of IAV to promote further P.a-mediated lung damage, not necessarily through its interference with host resistance to the bacterium, but by down-regulating tissue resilience to lung inflammation instead. Our study therefore suggests that restoring tissue resilience in clinical settings where IAV/P.a co-exists could prove a fruitful strategy.


Subject(s)
Coinfection/immunology , Elafin/metabolism , Influenza A virus/immunology , Matrix Metalloproteinase 9/metabolism , Pseudomonas aeruginosa/immunology , Animals , Cell Line , Coinfection/chemically induced , Coinfection/metabolism , Cystic Fibrosis/immunology , Cytokines/metabolism , Disease Susceptibility/metabolism , Epithelial Cells/metabolism , Humans , Inflammation/chemically induced , Inflammation/immunology , Lung/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Animal , Pneumonia/metabolism , Staphylococcal Infections/immunology
9.
Gut ; 69(1): 92-102, 2020 01.
Article in English | MEDLINE | ID: mdl-31003979

ABSTRACT

OBJECTIVE: Patients with primary sclerosing cholangitis (PSC) were previously shown to display a bacterial gut dysbiosis but fungal microbiota has never been examined in these patients. The aim of this study was to assess the fungal gut microbiota in patients with PSC. DESIGN: We analysed the faecal microbiota of patients with PSC and concomitant IBD (n=27), patients with PSC and no IBD (n=22), patients with IBD and no PSC (n=33) and healthy subjects (n=30). Bacterial and fungal composition of the faecal microbiota was determined using 16S and ITS2 sequencing, respectively. RESULTS: We found that patients with PSC harboured bacterial dysbiosis characterised by a decreased biodiversity, an altered composition and a decreased correlation network density. These alterations of the microbiota were associated with PSC, independently of IBD status. For the first time, we showed that patients with PSC displayed a fungal gut dysbiosis, characterised by a relative increase in biodiversity and an altered composition. Notably, we observed an increased proportion of Exophiala and a decreased proportion of Saccharomyces cerevisiae. Compared with patients with IBD and healthy subjects, the gut microbiota of patients with PSC exhibited a strong disruption in bacteria-fungi correlation network, suggesting an alteration in the interkingdom crosstalk. CONCLUSION: This study demonstrates that bacteria and fungi contribute to gut dysbiosis in PSC.


Subject(s)
Cholangitis, Sclerosing/microbiology , Dysbiosis/microbiology , Fungi/isolation & purification , Gastrointestinal Microbiome , Adult , Aged , Bacteria/classification , Bacteria/isolation & purification , Bacterial Typing Techniques/methods , Biodiversity , Female , Fungi/classification , Humans , Inflammatory Bowel Diseases/microbiology , Male , Middle Aged , Mycological Typing Techniques/methods , Young Adult
10.
Ann Rheum Dis ; 78(10): 1398-1404, 2019 10.
Article in English | MEDLINE | ID: mdl-31377728

ABSTRACT

OBJECTIVES: Familial Mediterranean fever (FMF) can be complicated by AA amyloidosis (AAA), though it remains unclear why only some patients develop amyloidosis. We examined the gut microbiota composition and inflammatory markers in patients with FMF complicated or not by AAA. METHODS: We analysed the gut microbiota of 34 patients with FMF without AAA, 7 patients with FMF with AAA, 19 patients with AAA of another origin, and 26 controls using 16S ribosomal RNA gene sequencing with the Illumina MiSeq platform. Associations between bacterial taxa and clinical phenotypes were evaluated using multivariate association with linear models statistical method. Blood levels of interleukin (IL)-1ß, IL-6, tumour necrosis factor-α and adipokines were assessed by ELISA; indoleamine 2,3-dioxygenase (IDO) activity was determined by high-performance liquid chromatography. RESULTS: Compared with healthy subjects, specific changes in faecal microbiota were observed in FMF and AAA groups. Several operational taxonomic units (OTUs) were associated with FMF. Moreover, two OTUs were over-represented in FMF-related AAA compared with FMF without AAA. Additionally, higher adiponectin levels and IDO activity were observed in FMF-related AAA compared with FMF without AAA (p<0.05). CONCLUSION: The presence of specific changes in faecal microbiota in FMF and in FMF-related AAA suggests that intestinal microorganisms may play a role in the pathogenesis of these diseases. These findings may offer an opportunity to use techniques for gut microbiota manipulation.


Subject(s)
Amyloidosis/microbiology , Familial Mediterranean Fever/microbiology , Feces/microbiology , Gastrointestinal Microbiome , Adiponectin/blood , Adult , Aged , Biomarkers , Cross-Sectional Studies , Cytokines/blood , Enzyme-Linked Immunosorbent Assay , Familial Mediterranean Fever/blood , Female , Humans , Inflammation Mediators/blood , Linear Models , Male , Middle Aged , Phenotype
12.
Microbiome ; 6(1): 152, 2018 09 01.
Article in English | MEDLINE | ID: mdl-30172257

ABSTRACT

BACKGROUND: Host-microbe balance maintains intestinal homeostasis and strongly influences inflammatory conditions such as inflammatory bowel diseases (IBD). Here we focused on bacteria-fungi interactions and their implications on intestinal inflammation, a poorly understood area. METHODS: Dextran sodium sulfate (DSS)-induced colitis was assessed in mice treated with vancomycin (targeting gram-positive bacteria) or colistin (targeting Enterobacteriaceae) and supplemented with either Saccharomyces boulardii CNCM I-745 or Candida albicans. Inflammation severity as well as bacterial and fungal microbiota compositions was monitored. RESULTS: While S. boulardii improved DSS-induced colitis and C. albicans worsened it in untreated settings, antibiotic treatment strongly modified DSS susceptibility and effects of fungi on colitis. Vancomycin-treated mice were fully protected from colitis, while colistin-treated mice retained colitis phenotype but were not affected anymore by administration of fungi. Antibacterial treatments not only influenced bacterial populations but also had indirect effects on fungal microbiota. Correlations between bacterial and fungal relative abundance were dramatically decreased in colistin-treated mice compared to vancomycin-treated and control mice, suggesting that colistin-sensitive bacteria are involved in interactions with fungi. Restoration of the Enterobacteriaceae population by administrating colistin-resistant Escherichia coli reestablished both beneficial effects of S. boulardii and pathogenic effects of C. albicans on colitis severity. This effect was at least partly mediated by an improved gut colonization by fungi. CONCLUSIONS: Fungal colonization of the gut is affected by the Enterobacteriaceae population, indirectly modifying effects of mycobiome on the host. This finding provides new insights into the role of inter-kingdom functional interactions in intestinal physiopathology and potentially in IBD.


Subject(s)
Candida albicans/physiology , Colitis/microbiology , Enterobacteriaceae/physiology , Saccharomyces boulardii/physiology , Animals , Antibiosis , Antibodies/administration & dosage , Candida albicans/genetics , Candida albicans/isolation & purification , Colitis/drug therapy , Disease Models, Animal , Enterobacteriaceae/classification , Enterobacteriaceae/genetics , Enterobacteriaceae/isolation & purification , Female , Gastrointestinal Microbiome , Humans , Mice , Mice, Inbred C57BL , Saccharomyces boulardii/genetics , Saccharomyces boulardii/isolation & purification
13.
Cell Metab ; 28(5): 737-749.e4, 2018 11 06.
Article in English | MEDLINE | ID: mdl-30057068

ABSTRACT

The extent to which microbiota alterations define or influence the outcome of metabolic diseases is still unclear, but the byproducts of microbiota metabolism are known to have an important role in mediating the host-microbiota interaction. Here, we identify that in both pre-clinical and clinical settings, metabolic syndrome is associated with the reduced capacity of the microbiota to metabolize tryptophan into derivatives that are able to activate the aryl hydrocarbon receptor. This alteration is not merely an effect of the disease as supplementation with AhR agonist or a Lactobacillus strain, with a high AhR ligand-production capacity, leads to improvement of both dietary- and genetic-induced metabolic impairments, particularly glucose dysmetabolism and liver steatosis, through improvement of intestinal barrier function and secretion of the incretin hormone GLP-1. These results highlight the role of gut microbiota-derived metabolites as a biomarker and as a basis for novel preventative or therapeutic interventions for metabolic disorders.


Subject(s)
Gastrointestinal Microbiome , Metabolic Syndrome/metabolism , Metabolic Syndrome/microbiology , Receptors, Aryl Hydrocarbon/metabolism , Tryptophan/metabolism , Animals , Limosilactobacillus reuteri/metabolism , Ligands , Male , Metabolic Syndrome/drug therapy , Metabolic Syndrome/therapy , Mice , Mice, Inbred C57BL , Probiotics/therapeutic use , Receptors, Aryl Hydrocarbon/agonists
14.
Cell Metab ; 28(4): 557-572.e6, 2018 10 02.
Article in English | MEDLINE | ID: mdl-30017352

ABSTRACT

The gut microbiota is essential for the normal function of the gut immune system, and microbiota alterations are associated with autoimmune disorders. However, how the gut microbiota prevents autoimmunity in distant organs remains poorly defined. Here we reveal that gut microbiota conditioned innate lymphoid cells (ILCs) induce the expression of mouse ß-defensin 14 (mBD14) by pancreatic endocrine cells, preventing autoimmune diabetes in the non-obese diabetic (NOD) mice. MBD14 stimulates, via Toll-like receptor 2, interleukin-4 (IL-4)-secreting B cells that induce regulatory macrophages, which in turn induce protective regulatory T cells. The gut microbiota-derived molecules, aryl hydrocarbon receptor (AHR) ligands and butyrate, promote IL-22 secretion by pancreatic ILCs, which induce expression of mBD14 by endocrine cells. Dysbiotic microbiota and low-affinity AHR allele explain the defective pancreatic expression of mBD14 observed in NOD mice. Our study reveals a yet unidentified crosstalk between ILCs and endocrine cells in the pancreas that is essential for the prevention of autoimmune diabetes development.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/prevention & control , Gastrointestinal Microbiome/immunology , Insulin-Secreting Cells/metabolism , Lymphocytes/metabolism , Pancreatic Polypeptide-Secreting Cells/metabolism , beta-Defensins/metabolism , Animals , B-Lymphocytes, Regulatory/metabolism , Female , Humans , Immunity, Innate , Interleukins/metabolism , Islets of Langerhans/metabolism , Kaplan-Meier Estimate , Macrophages/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Statistics, Nonparametric , T-Lymphocytes, Regulatory/metabolism , Toll-Like Receptor 2/metabolism , Interleukin-22
15.
ACS Nano ; 12(2): 1188-1202, 2018 02 27.
Article in English | MEDLINE | ID: mdl-29357226

ABSTRACT

Silver nanoparticles (AgNPs) are microbicidal agents which could be potentially used as an alternative to antivirals to treat human infectious diseases, especially influenza virus infections where antivirals have generally proven unsuccessful. However, concerns about the use of AgNPs on humans arise from their potential toxicity, although mechanisms are not well-understood. We show here, in the context of an influenza virus infection of lung epithelial cells, that AgNPs down-regulated influenza induced CCL-5 and -IFN-ß release (two cytokines important in antiviral immunity) through RIG-I inhibition, while enhancing IL-8 production, a cytokine important for mobilizing host antibacterial responses. AgNPs activity was independent of coating and was not observed with gold nanoparticles. Down-stream analysis indicated that AgNPs disorganized the mitochondrial network and prevented the antiviral IRF-7 transcription factor influx into the nucleus. Importantly, we showed that the modulation of RIG-I-IRF-7 pathway was concomitant with inhibition of either classical or alternative autophagy (ATG-5- and Rab-9 dependent, respectively), depending on the epithelial cell type used. Altogether, this demonstration of a AgNPs-mediated functional dichotomy (down-regulation of IFN-dependent antiviral responses and up-regulation of IL-8-dependent antibacterial responses) may have practical implications for their use in the clinic.


Subject(s)
Antiviral Agents/pharmacology , Epithelial Cells/drug effects , Lung/drug effects , Metal Nanoparticles/chemistry , Mitochondria/drug effects , Orthomyxoviridae/drug effects , Silver/pharmacology , Tretinoin/pharmacology , Animals , Antiviral Agents/chemistry , Autophagy/drug effects , Cell Line, Tumor , Dogs , Epithelial Cells/metabolism , Epithelial Cells/virology , Humans , Lung/metabolism , Lung/virology , Madin Darby Canine Kidney Cells/drug effects , Madin Darby Canine Kidney Cells/virology , Microbial Sensitivity Tests , Mitochondria/metabolism , Silver/chemistry , Tretinoin/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...