Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Nat Immunol ; 19(9): 1035, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29880894

ABSTRACT

In the version of this Article originally published, the asterisks indicating statistical significance were missing from Supplementary Figure 6; the file with the correct figure is now available.

2.
Nat Immunol ; 18(12): 1321-1331, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28991267

ABSTRACT

Type 1 diabetes (T1D) is an autoimmune disease that results from the destruction of pancreatic ß-cells by the immune system that involves innate and adaptive immune cells. Mucosal-associated invariant T cells (MAIT cells) are innate-like T-cells that recognize derivatives of precursors of bacterial riboflavin presented by the major histocompatibility complex (MHC) class I-related molecule MR1. Since T1D is associated with modification of the gut microbiota, we investigated MAIT cells in this pathology. In patients with T1D and mice of the non-obese diabetic (NOD) strain, we detected alterations in MAIT cells, including increased production of granzyme B, which occurred before the onset of diabetes. Analysis of NOD mice that were deficient in MR1, and therefore lacked MAIT cells, revealed a loss of gut integrity and increased anti-islet responses associated with exacerbated diabetes. Together our data highlight the role of MAIT cells in the maintenance of gut integrity and the control of anti-islet autoimmune responses. Monitoring of MAIT cells might represent a new biomarker of T1D, while manipulation of these cells might open new therapeutic strategies.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Histocompatibility Antigens Class I/analysis , Intestinal Mucosa/immunology , Minor Histocompatibility Antigens/analysis , Mucosal-Associated Invariant T Cells/immunology , Pancreas/immunology , Animals , Cells, Cultured , Gastrointestinal Microbiome/immunology , Granzymes/biosynthesis , Humans , Insulin-Secreting Cells/immunology , Intestinal Mucosa/cytology , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Pancreas/cytology
3.
Genes Immun ; 21(1): 71-77, 2020 01.
Article in English | MEDLINE | ID: mdl-31435002

ABSTRACT

Genome-wide association studies have implicated more than 50 genomic regions in type 1 diabetes (T1D). A T1D region at chromosome 16p13.13 includes the candidate genes CLEC16A and DEXI. Conclusive evidence as to which gene is causal for the disease association of this region is missing. We previously reported that Clec16a deficiency modified immune reactivity and protected against autoimmunity in the nonobese diabetic (NOD) mouse model for T1D. However, the diabetes-associated SNPs at 16p13.13 were described to also impact on DEXI expression and others have argued that DEXI is the causal gene in this disease locus. To help resolve whether DEXI affects disease, we generated Dexi knockout (KO) NOD mice. We found that Dexi deficiency had no effect on the frequency of diabetes. To test for possible interactions between Dexi and Clec16a, we intercrossed Dexi KO and Clec16a knockdown (KD) NOD mice. Dexi KO did not modify the disease protection afforded by Clec16a KD. We conclude that Dexi plays no role in autoimmune diabetes in the NOD model. Our data provide strongly suggestive evidence that CLEC16A, not DEXI, is causal for the T1D association of variants in the 16p13.13 region.


Subject(s)
DNA-Binding Proteins/genetics , Diabetes Mellitus, Type 1/genetics , Lectins, C-Type/genetics , Membrane Proteins/genetics , Monosaccharide Transport Proteins/genetics , Animals , Autoimmunity , DNA-Binding Proteins/metabolism , Diabetes Mellitus, Type 1/metabolism , Disease Models, Animal , Female , Genetic Predisposition to Disease , Genome-Wide Association Study , Male , Membrane Proteins/metabolism , Mice , Mice, Inbred NOD , Polymorphism, Single Nucleotide/genetics , Risk Factors
4.
bioRxiv ; 2023 Mar 23.
Article in English | MEDLINE | ID: mdl-36993342

ABSTRACT

Regulatory T cells (Tregs) protect against autoimmunity. In type 1 diabetes (T1D), Tregs slow the progression of beta cell autoimmunity within pancreatic islets. Increasing the potency or frequency of Tregs can prevent diabetes, as evidenced by studies in the nonobese diabetic (NOD) mouse model for T1D. We report herein that a significant proportion of islets Tregs in NOD mice express Gata3. The expression of Gata3 was correlated with the presence of IL-33, a cytokine known to induce and expand Gata3+ Tregs. Despite significantly increasing the frequency of Tregs in the pancreas, exogenous IL-33 was not protective. Based on these data, we hypothesized that Gata3 is deleterious to Treg function in autoimmune diabetes. To test this notion, we generated NOD mice with a Treg-specific deletion of Gata3. We found that deleting Gata3 in Tregs strongly protected against diabetes. Disease protection was associated with a shift of islet Tregs toward a suppressive CXCR3+Foxp3+ population. Our results suggest that islet Gata3+ Tregs are maladaptive and that this Treg subpopulation compromises the regulation of islet autoimmunity, contributing to diabetes onset.

5.
Front Immunol ; 13: 906499, 2022.
Article in English | MEDLINE | ID: mdl-35720357

ABSTRACT

CD5 is constitutively expressed on all T cells and is a negative regulator of lymphocyte function. However, the full extent of CD5 function in immunity remains unclear. CD5 deficiency impacts thymic selection and extra-thymic regulatory T cell generation, yet CD5 knockout was reported to cause no immune pathology. Here we show that CD5 is a key modulator of gut immunity. We generated mice with inducible CD5 knockdown (KD) in the autoimmune-prone nonobese diabetic (NOD) background. CD5 deficiency caused T cell-dependent wasting disease driven by chronic gut immune dysregulation. CD5 inhibition also exacerbated acute experimental colitis. Mechanistically, loss of CD5 increased phospho-Stat3 levels, leading to elevated IL-17A secretion. Our data reveal a new facet of CD5 function in shaping the T cell cytokine profile.


Subject(s)
CD5 Antigens , Animals , Lymphocyte Count , Mice
6.
Nat Metab ; 2(9): 934-945, 2020 09.
Article in English | MEDLINE | ID: mdl-32719542

ABSTRACT

Type 1 diabetes (T1D) is caused by the autoimmune destruction of pancreatic beta cells. Pluripotent stem cells can now be differentiated into beta cells, thus raising the prospect of a cell replacement therapy for T1D. However, autoimmunity would rapidly destroy newly transplanted beta cells. Using a genome-scale CRISPR screen in a mouse model for T1D, we show that deleting RNLS, a genome-wide association study candidate gene for T1D, made beta cells resistant to autoimmune killing. Structure-based modelling identified the U.S. Food and Drug Administration-approved drug pargyline as a potential RNLS inhibitor. Oral pargyline treatment protected transplanted beta cells in diabetic mice, thus leading to disease reversal. Furthermore, pargyline prevented or delayed diabetes onset in several mouse models for T1D. Our results identify RNLS as a modifier of beta cell vulnerability and as a potential therapeutic target to avert beta cell loss in T1D.


Subject(s)
CRISPR-Cas Systems , Diabetes Mellitus, Type 1/drug therapy , Genome-Wide Association Study , Insulin-Secreting Cells/drug effects , Monoamine Oxidase/drug effects , Animals , Autoimmunity/drug effects , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/pathology , Endoplasmic Reticulum Stress , Enzyme Inhibitors/pharmacology , Female , Induced Pluripotent Stem Cells/immunology , Insulin-Secreting Cells/immunology , Insulin-Secreting Cells/pathology , Islets of Langerhans Transplantation , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mutation , Pargyline/pharmacology
7.
Nat Commun ; 11(1): 3755, 2020 07 24.
Article in English | MEDLINE | ID: mdl-32709874

ABSTRACT

Obesity is associated with low-grade chronic inflammation promoting insulin-resistance and diabetes. Gut microbiota dysbiosis is a consequence as well as a driver of obesity and diabetes. Mucosal-associated invariant T cells (MAIT) are innate-like T cells expressing a semi-invariant T cell receptor restricted to the non-classical MHC class I molecule MR1 presenting bacterial ligands. Here we show that during obesity MAIT cells promote inflammation in both adipose tissue and ileum, leading to insulin resistance and impaired glucose and lipid metabolism. MAIT cells act in adipose tissue by inducing M1 macrophage polarization in an MR1-dependent manner and in the gut by inducing microbiota dysbiosis and loss of gut integrity. Both MAIT cell-induced tissue alterations contribute to metabolic dysfunction. Treatment with MAIT cell inhibitory ligand demonstrates its potential as a strategy against inflammation, dysbiosis and metabolic disorders.


Subject(s)
Dysbiosis/immunology , Inflammation/pathology , Intestines/pathology , Mucosal-Associated Invariant T Cells/pathology , Obesity/metabolism , Adipose Tissue/pathology , Animals , Cytokines/genetics , Cytokines/metabolism , Diet, High-Fat , Dysbiosis/complications , Gastrointestinal Microbiome , Glucose Tolerance Test , Ileum/pathology , Inflammation/complications , Intestinal Mucosa/pathology , Intestines/diagnostic imaging , Ligands , Lymphocyte Count , Macrophages/metabolism , Magnetic Resonance Imaging , Mice , Mice, Inbred C57BL , Obesity/complications , Obesity/diagnostic imaging , Phenotype , Pterins/pharmacology , Receptors, Antigen, T-Cell/metabolism
8.
Nat Commun ; 9(1): 2146, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29858567

ABSTRACT

Liver fibrosis is the common response to chronic liver injury, and leads to cirrhosis and its complications. Persistent inflammation is a driving force of liver fibrosis progression. Mucosal-associated invariant T (MAIT) cells are non-conventional T cells that display altered functions during chronic inflammatory diseases. Here, we show that circulating MAIT cells are reduced in patients with alcoholic or non-alcoholic fatty liver disease-related cirrhosis while they accumulate in liver fibrotic septa. Using two models of chronic liver injury, we demonstrate that MAIT cell-enriched mice show increased liver fibrosis and accumulation of hepatic fibrogenic cells, whereas MAIT cell-deficient mice are resistant. Co-culture experiments indicate that MAIT cells enhance the proinflammatory properties of monocyte-derived macrophages, and promote mitogenic and proinflammatory functions of fibrogenic cells, via distinct mechanisms. Our results highlight the profibrogenic functions of MAIT cells and suggest that targeting MAIT cells may constitute an attractive antifibrogenic strategy during chronic liver injury.


Subject(s)
Liver Cirrhosis/immunology , Macrophages/immunology , Mucosal-Associated Invariant T Cells/immunology , Non-alcoholic Fatty Liver Disease/immunology , Adult , Aged , Animals , Cell Count , Cells, Cultured , Coculture Techniques , Female , Humans , Liver/immunology , Liver/pathology , Liver Cirrhosis/blood , Liver Cirrhosis/pathology , Male , Mice , Middle Aged , Non-alcoholic Fatty Liver Disease/blood , Non-alcoholic Fatty Liver Disease/pathology
9.
Front Immunol ; 6: 341, 2015.
Article in English | MEDLINE | ID: mdl-26191063

ABSTRACT

Type 1 diabetes (T1D) and type 2 diabetes (T2D) are multifactorial diseases with different etiologies in which chronic inflammation takes place. Defects in invariant natural killer T (iNKT) cell populations have been reported in both T1D and T2D patients, mouse models and our recent study revealed mucosal-associated invariant T (MAIT) cell defects in T2D and obese patients. Regarding iNKT cells many studies in non-obese diabetic mice demonstrated their protective role against T1D whereas their potential role in human T1D is still under debate. Studies in mouse models and patients suggest that iNKT cells present in adipose tissue (AT) could exert a regulatory role against obesity and associated metabolic disorders, such as T2D. Scarce data are yet available on MAIT cells; however, we recently described MAIT cell abnormalities in the blood and ATs from obese and T2D patients. These data show that a link between MAIT cells and metabolic disorders pave the way for further investigations on MAIT cells in T1D and T2D in humans and mouse models. Furthermore, we hypothesize that the gut microbiota alterations associated with T1D and T2D could modulate iNKT and MAIT cell frequency and functions. The potential role of iNKT and MAIT cells in the regulation of metabolic pathways and their cross-talk with microbiota represent exciting new lines of research.

10.
J Clin Invest ; 125(4): 1752-62, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25751065

ABSTRACT

Obesity and type 2 diabetes (T2D) are associated with low-grade inflammation, activation of immune cells, and alterations of the gut microbiota. Mucosal-associated invariant T (MAIT) cells, which are innate-like T cells that recognize bacterial ligands, are present in blood and enriched in mucosal and inflamed tissues. Here, we analyzed MAIT cells in the blood and adipose tissues of patients with T2D and/or severe obesity. We determined that circulating MAIT cell frequency was dramatically decreased in both patient groups, and this population was even undetectable in some obese patients. Moreover, in both patient groups, circulating MAIT cells displayed an activated phenotype that was associated with elevated Th1 and Th17 cytokine production. In obese patients, MAIT cells were more abundant in adipose tissue than in the blood and exhibited a striking IL-17 profile. Bariatric surgery in obese patients not only improved their metabolic parameters but also increased circulating MAIT cell frequency at 3 months after surgery. Similarly, cytokine production by blood MAIT cells was strongly decreased after surgery. This study reveals profound MAIT cell abnormalities in patients harboring metabolic disorders, suggesting their potential role in these pathologies.


Subject(s)
Adipose Tissue/immunology , Diabetes Mellitus, Type 2/immunology , Natural Killer T-Cells/immunology , Obesity/immunology , T-Lymphocyte Subsets/immunology , Adiponectin/blood , Adult , Bariatric Surgery , Blood Cells/immunology , Cytokines/biosynthesis , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/pathology , Female , Humans , Inflammation , Interleukin-17/biosynthesis , Interleukin-17/blood , Leptin/blood , Lymphocyte Count , Male , Middle Aged , Obesity/blood , Obesity/pathology , Obesity/surgery , Omentum/immunology , Organ Specificity , Postoperative Period , Subcutaneous Tissue/immunology
SELECTION OF CITATIONS
SEARCH DETAIL