Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
J Scleroderma Relat Disord ; 9(1): 67-78, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38333523

ABSTRACT

Objective: Systemic sclerosis is an autoimmune disease characterized by fibrosis of the skin and internal organs including the lung. Mucosal-associated invariant T cells are innate-like T lymphocytes able to produce various cytokines and cytotoxic mediators such as granzyme B. A large body of evidence supports a role of mucosal-associated invariant T cells in autoimmune disease but more recent reports suggest also a potential role in fibrotic conditions. Therefore, we herein addressed the question as whether mucosal-associated invariant T cells may have an altered profile in systemic sclerosis. Methods: Mucosal-associated invariant T cell frequency was analyzed by flow cytometry, using fresh peripheral blood from 74 consecutive systemic sclerosis patients who were compared to 44 healthy donors. In addition, in-depth mucosal-associated invariant T cell phenotype and function were analyzed in unselected 29 women with systemic sclerosis who were compared to 23 healthy women donors. Results: Proportion of circulating mucosal-associated invariant T cells was significantly reduced by 68% in systemic sclerosis compared to healthy donors (0.78% in systemic sclerosis vs 2.5%, p < 0.0001). Within systemic sclerosis subsets, mucosal-associated invariant T cells were reduced in patients with interstitial lung disease (systemic sclerosis-interstitial lung disease) (0.56% vs 0.96% in patients without interstitial lung disease, p = 0.04). Moreover, in systemic sclerosis patients, mucosal-associated invariant T cells displayed an activated phenotype indicated by markedly increased CD69+ mucosal-associated invariant T cell frequency (20% mucosal-associated invariant T cell CD69+ compared to 9.4% in healthy donors, p = 0.0014). Interestingly, mucosal-associated invariant T cells from systemic sclerosis-interstitial lung disease patients had a more pronounced altered phenotype compared to systemic sclerosis without interstitial lung disease with a correlation between mucosal-associated invariant T cells expressing CCR6+ and mucosal-associated invariant T cell frequency (r = 0.8, p = 0.006). Conclusion: Circulating mucosal-associated invariant T cells were reduced and exhibited an activated phenotype in systemic sclerosis patients. This peripheral mucosal-associated invariant T cell deficiency may be related to enhanced apoptosis and/or homing in inflamed tissue, particularly in systemic sclerosis-interstitial lung disease patients.

2.
Front Immunol ; 14: 1205405, 2023.
Article in English | MEDLINE | ID: mdl-37885889

ABSTRACT

Introduction: Systemic lupus erythematosus (SLE) is an autoimmune disease in which circulating immune complexes can cause different types of glomerulonephritis, according to immune deposits and to the type of glomerular cell injury. Proliferative lesions represent the most severe form of lupus nephritis (LN) and often lead to kidney failure and death. Mucosal-associated invariant T (MAIT) cells are a subset of innate-like T cells that recognize microbial-derived ligands from the riboflavin synthesis pathway. Although abundant in peripheral blood, MAIT cells are enriched in mucosal and inflamed tissues. While previous studies have reported concordant results concerning lower MAIT cell frequencies in the blood of SLE patients, no information is known about MAIT cell function and LN severity and outcome. Methods: In the current study, we analyzed the baseline phenotype and function of peripheral blood MAIT cells by flow cytometry in 26 patients with LN and in a control group of 16 healthy individuals. Results: We observe that MAIT cell frequencies are markedly reduced in blood of LN patients. MAIT cells from patients have an altered phenotype in terms of migration, proliferation and differentiation markers, notably in most severe forms of LN. Frequencies of PMA/ionomycin stimulated MAIT cells secreting effector molecules, such as proinflammatory IL-17 and cytotoxic protein granzyme B, are higher in LN patients. Patients undergoing a complete renal remission after immunosuppressive therapy had higher MAIT cell frequency, lower expression of proliferation marker Ki-67 and granzyme B (GzB) at inclusion. Remarkably, GzB production defines a predictive model for complete remission. Discussion: We report here that blood MAIT cells display proinflammatory and cytotoxic function in severe lupus nephritis which may play a pathogenesis role, but without association with systemic lupus activity. Finally, low cytotoxic profile of MAIT cells may represent a promising prognostic factor of lupus nephritis remission one year after induction therapy.


Subject(s)
Lupus Erythematosus, Systemic , Lupus Nephritis , Mucosal-Associated Invariant T Cells , Humans , Granzymes , Phenotype , Patient Acuity
3.
Mol Metab ; 57: 101438, 2022 03.
Article in English | MEDLINE | ID: mdl-35007789

ABSTRACT

OBJECTIVE: A common feature of metabolic diseases is their association with chronic low-grade inflammation. While enhanced gut permeability and systemic bacterial endotoxin translocation have been suggested as key players of this metaflammation, the mechanistic bases underlying these features upon the diabesity cascade remain partly understood. METHODS: Here, we show in mice that, independently of obesity, the induction of acute and global insulin resistance and associated hyperglycemia, upon treatment with an insulin receptor (IR) antagonist (S961), elicits gut hyperpermeability without triggering systemic inflammatory response. RESULTS: Of note, S961-treated diabetic mice display major defects of gut barrier epithelial functions, such as increased epithelial paracellular permeability and impaired cell-cell junction integrity. We also observed in these mice the early onset of a severe gut dysbiosis, as characterized by the bloom of pro-inflammatory Proteobacteria, and the later collapse of Paneth cells antimicrobial defense. Interestingly, S961 treatment discontinuation is sufficient to promptly restore both the gut microbial balance and the intestinal barrier integrity. Moreover, fecal transplant approaches further confirm that S961-mediated dybiosis contributes at least partly to the disruption of the gut selective epithelial permeability upon diabetic states. CONCLUSIONS: Together, our results highlight that insulin signaling is an indispensable gatekeeper of intestinal barrier integrity, acting as a safeguard against microbial imbalance and acute infections by enteropathogens.


Subject(s)
Diabetes Mellitus, Experimental , Gastrointestinal Microbiome , Insulin Resistance , Animals , Dysbiosis/metabolism , Dysbiosis/microbiology , Gastrointestinal Microbiome/physiology , Inflammation/metabolism , Mice
4.
Gut ; 71(2): 296-308, 2022 02.
Article in English | MEDLINE | ID: mdl-33593807

ABSTRACT

OBJECTIVE: Type 1 diabetes (T1D) is an autoimmune disease caused by the destruction of pancreatic ß-cells producing insulin. Both T1D patients and animal models exhibit gut microbiota and mucosa alterations, although the exact cause for these remains poorly understood. We investigated the production of key cytokines controlling gut integrity, the abundance of segmented filamentous bacteria (SFB) involved in the production of these cytokines, and the respective role of autoimmune inflammation and hyperglycaemia. DESIGN: We used several mouse models of autoimmune T1D as well as mice rendered hyperglycaemic without inflammation to study gut mucosa and microbiota dysbiosis. We analysed cytokine expression in immune cells, epithelial cell function, SFB abundance and microbiota composition by 16S sequencing. We assessed the role of anti-tumour necrosis factor α on gut mucosa inflammation and T1D onset. RESULTS: We show in models of autoimmune T1D a conserved loss of interleukin (IL)-17A, IL-22 and IL-23A in gut mucosa. Intestinal epithelial cell function was altered and gut integrity was impaired. These defects were associated with dysbiosis including progressive loss of SFB. Transfer of diabetogenic T-cells recapitulated these gut alterations, whereas induction of hyperglycaemia with no inflammation failed to do so. Moreover, anti-inflammatory treatment restored gut mucosa and immune cell function and dampened diabetes incidence. CONCLUSION: Our results demonstrate that gut mucosa alterations and dysbiosis in T1D are primarily linked to inflammation rather than hyperglycaemia. Anti-inflammatory treatment preserves gut homeostasis and protective commensal flora reducing T1D incidence.


Subject(s)
Bacteria/isolation & purification , Diabetes Mellitus, Type 1/complications , Dysbiosis/etiology , Gastrointestinal Microbiome , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Animals , Cytokines/metabolism , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/microbiology , Disease Models, Animal , Epithelial Cells/metabolism , Epithelial Cells/pathology , Hyperglycemia/etiology , Inflammation/etiology , Intestinal Mucosa/metabolism , Mice
5.
Diabetologia ; 64(10): 2306-2321, 2021 10.
Article in English | MEDLINE | ID: mdl-34350463

ABSTRACT

AIMS/HYPOTHESIS: Mucosal-associated invariant T (MAIT) cells are innate-like T lymphocytes expressing an αß T cell antigen receptor that recognises the MHC-related 1 molecule. MAIT cells are altered in children at risk for and with type 1 diabetes, and mouse model studies have shown MAIT cell involvement in type 1 diabetes development. Since several studies support heterogeneity in type 1 diabetes physiopathology according to the age of individuals, we investigated whether MAIT cells were altered in adults with type 1 diabetes. METHODS: MAIT cell frequency, phenotype and function were analysed by flow cytometry, using fresh peripheral blood from 21 adults with recent-onset type 1 diabetes (2-14 days after disease onset) and 47 adults with long-term disease (>2 years after diagnosis) compared with 55 healthy blood donors. We also separately analysed 17 women with long-term type 1 diabetes and an associated autoimmune disease, compared with 30 healthy women and 27 women with long-term type 1 diabetes. RESULTS: MAIT cells from adults with recent-onset type 1 diabetes, compared with healthy adult donors, harboured a strongly activated phenotype indicated by an elevated CD25+ MAIT cell frequency. In adults with long-term type 1 diabetes, MAIT cells displayed an activated and exhausted phenotype characterised by high CD25 and programmed cell death 1 (PD1) expression and a decreased production of proinflammatory cytokines, IL-2, IFN-γ and TNF-α. Even though MAIT cells from these patients showed upregulated IL-17 and IL-4 production, the polyfunctionality of MAIT cells was decreased (median 4.8 vs 13.14% of MAIT cells, p < 0.001) and the frequency of MAIT cells producing none of the effector molecules analysed increased (median 34.40 vs 19.30% of MAIT cells, p < 0.01). Several MAIT cell variables correlated with HbA1c level and more particularly in patients with recent-onset type 1 diabetes. In women with long-term type 1 diabetes, MAIT cell alterations were more pronounced in those with an associated autoimmune disease than in those without another autoimmune disease. In women with long-term type 1 diabetes and an associated autoimmune disease, there was an increase in CD69 expression and a decrease in the survival B-cell lymphoma 2 (BCL-2) (p < 0.05) and CD127 (IL-7R) (p < 0.01) marker expression compared with women without a concomitant autoimmune disorder. Concerning effector molecules, TNF-α and granzyme B production by MAIT cells was decreased. CONCLUSIONS/INTERPRETATION: Alterations in MAIT cell frequency, phenotype and function were more pronounced in adults with long-term type 1 diabetes compared with adults with recent-onset type 1 diabetes. There were several correlations between MAIT cell variables and clinical characteristics. Moreover, the presence of another autoimmune disease in women with long-term type 1 diabetes further exacerbated MAIT cell alterations. Our results suggest that MAIT cell alterations in adults with type 1 diabetes could be associated with two aspects of the disease: impaired glucose homeostasis; and autoimmunity.


Subject(s)
Diabetes Mellitus, Type 1/pathology , Mucosal-Associated Invariant T Cells/pathology , Adult , Aged , Antigens, CD , Antigens, Differentiation, T-Lymphocyte , Biomarkers/metabolism , Blood Donors , Cytokines/metabolism , Diabetes Mellitus, Type 1/metabolism , Female , Flow Cytometry , Humans , Interleukin-2 Receptor alpha Subunit/metabolism , Lectins, C-Type , Male , Middle Aged , Mucosal-Associated Invariant T Cells/metabolism , Phenotype , Programmed Cell Death 1 Receptor/metabolism , Proto-Oncogene Proteins c-bcl-2 , Young Adult
6.
Nat Immunol ; 22(3): 322-335, 2021 03.
Article in English | MEDLINE | ID: mdl-33531712

ABSTRACT

Immune system dysfunction is paramount in coronavirus disease 2019 (COVID-19) severity and fatality rate. Mucosal-associated invariant T (MAIT) cells are innate-like T cells involved in mucosal immunity and protection against viral infections. Here, we studied the immune cell landscape, with emphasis on MAIT cells, in cohorts totaling 208 patients with various stages of disease. MAIT cell frequency is strongly reduced in blood. They display a strong activated and cytotoxic phenotype that is more pronounced in lungs. Blood MAIT cell alterations positively correlate with the activation of other innate cells, proinflammatory cytokines, notably interleukin (IL)-18, and with the severity and mortality of severe acute respiratory syndrome coronavirus 2 infection. We also identified a monocyte/macrophage interferon (IFN)-α-IL-18 cytokine shift and the ability of infected macrophages to induce the cytotoxicity of MAIT cells in an MR1-dependent manner. Together, our results suggest that altered MAIT cell functions due to IFN-α-IL-18 imbalance contribute to disease severity, and their therapeutic manipulation may prevent deleterious inflammation in COVID-19 aggravation.


Subject(s)
COVID-19/immunology , Interferon-alpha/immunology , Interleukin-18/immunology , Macrophages/immunology , Monocytes/immunology , Mucosal-Associated Invariant T Cells/immunology , Adult , Aged , Aged, 80 and over , Animals , Bronchoalveolar Lavage , Case-Control Studies , Chlorocebus aethiops , Cohort Studies , Female , France , Humans , Immunophenotyping , Interleukin-10/immunology , Interleukin-15/immunology , Interleukin-1beta/immunology , Interleukin-6/immunology , Interleukin-8/immunology , Male , Middle Aged , RNA-Seq , SARS-CoV-2 , Severity of Illness Index , Single-Cell Analysis , Vero Cells , Young Adult
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.
Methods Mol Biol ; 2098: 283-297, 2020.
Article in English | MEDLINE | ID: mdl-31792829

ABSTRACT

MAIT cells are unconventional T cells expressing a semi-invariant αß TCR, and they recognize bacterial metabolites via the highly conserved MR1 protein. MAIT cells interact with gut microbiota and literature reports alterations of gut homeostasis in type 1 diabetes (T1D), suggesting the involvement of MAIT cells in T1D. Since NOD mice is a well-established mouse model of T1D, MAIT cells were studied in these mice to evaluate their potential involvement in disease development. This chapter describes the material and methods required to characterize MAIT cells and to determine their function in T1D mouse models.


Subject(s)
Diabetes Mellitus, Type 1/metabolism , Mucosal-Associated Invariant T Cells/immunology , Mucosal-Associated Invariant T Cells/metabolism , Animals , Biomarkers , Cell Separation/methods , Diabetes Mellitus, Type 1/etiology , Disease Models, Animal , Disease Susceptibility , Flow Cytometry , Gastrointestinal Microbiome , Humans , Immunophenotyping , Lymphocyte Activation/immunology , Mice , Mice, Inbred NOD , Organ Specificity/immunology
9.
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.

10.
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
11.
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
12.
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
13.
Eur J Immunol ; 44(5): 1454-66, 2014 May.
Article in English | MEDLINE | ID: mdl-24481989

ABSTRACT

Invariant NKT (iNKT)-cell stimulation with exogenous specific ligands prevents the development of type 1 diabetes (T1D) in NOD mice. Studies based on anti-islet T-cell transfer showed that iNKT cells prevent the differentiation of these T cells into effector T cells in the pancreatic lymph nodes (PLNs). We hypothesize that this defective priming could be explained by the ability of iNKT cells to induce tolerogenic dendritic cells (DCs) in the PLNs. We evaluated the effect of iNKT-cell stimulation on T1D development by transferring naïve diabetogenic BDC2.5 T cells into proinsulin 2(-/-) NOD mice treated with a long-lasting α-galactosylceramide regimen. In this context, iNKT cells induce the conversion of BDC2.5 T cells into Foxp3(+) Treg cells in the PLNs accumulating in the pancreatic islets. Furthermore, tolerogenic plasmacytoid DCs (pDCs) characterized by low MHC class II molecule expression and TGF-ß production are critical in the PLNs for the recruitment of Treg cells into the pancreatic islets by inducing CXCR3 expression. Accordingly, pDC depletion in α-galactosylceramide-treated proinsulin 2(-/-) NOD mice abrogates the protection against T1D. These findings reveal that upon repetitive iNKT-cell stimulation, pDCs are critical for the recruitment of Treg cells in the pancreatic islets and the prevention of T1D development.


Subject(s)
Dendritic Cells/immunology , Diabetes Mellitus, Type 1/immunology , Islets of Langerhans/immunology , Natural Killer T-Cells/immunology , Plasma Cells/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Dendritic Cells/pathology , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 1/prevention & control , Islets of Langerhans/pathology , Lymph Nodes/immunology , Lymph Nodes/pathology , Mice , Mice, Inbred NOD , Mice, Knockout , Natural Killer T-Cells/pathology , Plasma Cells/pathology , Proinsulin/genetics , Proinsulin/immunology , T-Lymphocytes, Regulatory/pathology
14.
PLoS One ; 9(1): e87717, 2014.
Article in English | MEDLINE | ID: mdl-24498175

ABSTRACT

Invariant NKT cells are innate lymphocytes with a broad tissue distribution. Here we demonstrate that iNKT cells reside in the central nervous system (CNS) in the absence of inflammation. Their presence in the CNS dramatically augments following inoculation of C57Bl/6 mice with the neurotropic Theiler's murine encephalomyelitis virus (TMEV). At the peak of inflammation the cellular infiltrate comprises 45,000 iNKT cells for 1250 CD8 T cells specific for the immunodominant TMEV epitope. To study the interaction between these two T cell subsets, we infected both iNKT cell deficient Jα18(-/-) mice and iNKT cell enriched Vα14 transgenic mice with TMEV. The CD8 T cell response readily cleared TMEV infection in the iNKT cell deficient mice. However, in the iNKT cell enriched mice TMEV infection persisted and was associated with significant mortality. This was caused by the inhibition of the CD8 T cell response in the cervical lymph nodes and spleen after T cell priming. Taken together we demonstrate that iNKT cells reside in the CNS in the absence of inflammation and that their enrichment is associated with the inhibition of the anti-viral CD8 T cell response and an augmented mortality during acute encephalomyelitis.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Natural Killer T-Cells/immunology , Poliomyelitis/immunology , Theilovirus/immunology , Animals , CD8-Positive T-Lymphocytes/virology , Central Nervous System/immunology , Central Nervous System/virology , Encephalomyelitis/immunology , Encephalomyelitis/virology , Epitopes/immunology , Inflammation/immunology , Inflammation/virology , Lymph Nodes/immunology , Lymph Nodes/virology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Poliomyelitis/virology , Spleen/immunology , Spleen/virology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/virology
15.
Med Sci (Paris) ; 29(8-9): 722-8, 2013.
Article in French | MEDLINE | ID: mdl-24005626

ABSTRACT

Type 1 diabetes is an autoimmune disease resulting from the destruction of pancreatic ß cells by the immune system. NKT cells are innate-like T cells that can exert potent immuno-regulatory functions. The regulatory role of NKT cells was initially proposed after the observed decreased frequency of this subset in mouse models of type 1 diabetes, as well as in patients developing various autoimmune pathologies. Increasing NKT cell frequency and function prevent the development of type 1 diabetes in mouse models. Several mechanisms including IL-4 and IL-10 production by NKT cells and the accumulation of tolerogenic dendritic cells are critical for the dampening of pathogenic anti-islet T cell responses by NKT cells. Importantly, these cells can at the same time prevent diabetes and promote efficient immune responses against infectious agents. These results strengthen the potential role of NKT cells as a key target for the development of therapeutic strategies against type 1 diabetes.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/prevention & control , Killer Cells, Natural/immunology , Animals , Antigen Presentation/immunology , Dendritic Cells/immunology , Galactosylceramides/therapeutic use , Homeostasis , Humans , Immune Tolerance/immunology , Interleukin-10/physiology , Interleukin-4/physiology , Mice , Virus Diseases/immunology
16.
J Immunol ; 191(5): 2335-43, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23926325

ABSTRACT

IgA plays ambivalent roles in the immune system. The balance between inhibitory and activating responses relies on the multimerization status of IgA and interaction with their cognate receptors. In mucosal sites, secretory IgA (SIgA) protects the host through immune-exclusion mechanisms, but its function in the bloodstream remains unknown. Using bone marrow-derived dendritic cells, we found that both human and mouse SIgA induce tolerogenic dendritic cells (DCs) following binding to specific ICAM-3 grabbing nonintegrin receptor 1. This interaction was dependent on Ca(2+) and mannose residues. SIgA-primed DCs (SIgA-DCs) are resistant to TLR-dependent maturation. Although SIgA-DCs fail to induce efficient proliferation and Th1 differentiation of naive responder T cells, they generate the expansion of regulatory T cells through IL-10 production. SIgA-DCs are highly potent in inhibiting autoimmune responses in mouse models of type 1 diabetes and multiple sclerosis. This discovery may offer new insights about mucosal-derived DC immunoregulation through SIgA opening new therapeutic approaches to autoimmune diseases.


Subject(s)
Autoimmunity/immunology , Cell Adhesion Molecules/immunology , Dendritic Cells/immunology , Immunoglobulin A, Secretory/immunology , Lectins, C-Type/immunology , Receptors, Cell Surface/immunology , Animals , Bone Marrow Cells/immunology , Cell Differentiation/immunology , Dendritic Cells/cytology , Flow Cytometry , Gene Knockdown Techniques , Humans , Immune Tolerance/immunology , Mice , Mice, Inbred C57BL
17.
Diabetes ; 62(11): 3785-96, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23894189

ABSTRACT

Invariant natural killer T (iNKT) cells belong to the innate immune system and exercise a dual role as potent regulators of autoimmunity and participate in responses against different pathogens. They have been shown to prevent type 1 diabetes development and to promote antiviral responses. Many studies in the implication of environmental factors on the etiology of type 1 diabetes have suggested a link between enteroviral infections and the development of this disease. This study of the pancreatropic enterovirus Coxsackievirus B4 (CVB4) shows that although infection accelerated type 1 diabetes development in a subset of proinsulin 2-deficient NOD mice, the activation of iNKT cells by a specific agonist, α-galactosylceramide, at the time of infection inhibited the disease. Diabetes development was associated with the infiltration of pancreatic islets by inflammatory macrophages, producing high levels of interleukin (IL)-1ß, IL-6, and tumor necrosis factor-α and activation of anti-islet T cells. On the contrary, macrophages infiltrating the islets after CVB4 infection and iNKT-cell stimulation expressed a number of suppressive enzymes, among which indoleamine 2,3-dioxygenase was sufficient to inhibit anti-islet T-cell response and to prevent diabetes. This study highlights the critical interaction between virus and the immune system in the acceleration or prevention of type 1 diabetes.


Subject(s)
Islets of Langerhans/immunology , Natural Killer T-Cells/immunology , Animals , Coxsackievirus Infections/immunology , Diabetes Mellitus, Type 1/prevention & control , Female , Galactosylceramides/pharmacology , Indoleamine-Pyrrole 2,3,-Dioxygenase/biosynthesis , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Interferon-gamma/physiology , Interleukin-13/physiology , Islets of Langerhans/drug effects , Macrophages/immunology , Mice , Mice, Inbred NOD , Mice, Transgenic , Natural Killer T-Cells/drug effects
18.
Kidney Int ; 84(2): 317-26, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23515052

ABSTRACT

Mast cell release of chymase is important in tissue remodeling and may participate in inflammation leading to fibrosis and organ failure. Here we analyzed the function of chymase in unilateral ureteral obstruction, an established accelerated model of renal tubulointerstitial fibrosis. Mice deficient in mouse mast cell protease 4 (mMCP4), the functional counterpart of human chymase, had increased obstruction-induced fibrosis when compared to wild-type mice indicating a protective effect of mMCP4. Engraftment of mast cell-deficient Kit(Wsh/Wsh) mice with wild type, but not mMCP4-deficient mast cells, restored protection confirming the role of mMCP4. Kidneys of mMCP4-deficient mice had higher levels of renal tubular damage, interstitial fibrosis, collagen deposition, increased α-smooth muscle actin, and decreased E-cadherin expression compared to the kidneys of wild-type mice. Further analysis showed an elevated inflammatory response in mMCP4-deficient mice with increased levels of kidney-infiltrating macrophages and T cells and local profibrotic TGF-ß1 and CCL2. Granulated and degranulated mast cells and mMCP4 were mainly found in the kidney capsule, respectively, before and after ureteral obstruction. Analysis of mMCP4 substrates showed that it mediates its anti-fibrotic actions by degrading interstitial deposits of fibronectin, a known promoter of inflammatory cell infiltration and adhesion. Thus, mast cell released mMCP4 has anti-fibrotic potential in acutely induced obstructive nephropathy.


Subject(s)
Chymases/metabolism , Kidney Diseases/prevention & control , Kidney/enzymology , Mast Cells/enzymology , Serine Endopeptidases/metabolism , Ureteral Obstruction/complications , Actins/metabolism , Animals , Cadherins/metabolism , Cell Degranulation , Chemokine CCL2/metabolism , Chemotaxis , Chymases/deficiency , Chymases/genetics , Collagen/metabolism , Disease Models, Animal , Fibronectins/metabolism , Fibrosis , Kidney/immunology , Kidney/pathology , Kidney Diseases/enzymology , Kidney Diseases/etiology , Kidney Diseases/immunology , Kidney Diseases/pathology , Macrophages/immunology , Macrophages/metabolism , Male , Mast Cells/immunology , Mast Cells/transplantation , Mice , Mice, Inbred C57BL , Mice, Knockout , Myofibroblasts/metabolism , Serine Endopeptidases/deficiency , Serine Endopeptidases/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transforming Growth Factor beta1/metabolism , Ureteral Obstruction/enzymology , Ureteral Obstruction/immunology , Ureteral Obstruction/pathology
19.
J Immunol ; 190(7): 3299-308, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23427253

ABSTRACT

Invariant NKT (iNKT) cells play key roles in host defense by recognizing lipid Ags presented by CD1d. iNKT cells are activated by bacterial-derived lipids and are also strongly autoreactive toward self-lipids. iNKT cell responsiveness must be regulated to maintain effective host defense while preventing uncontrolled stimulation and potential autoimmunity. CD1d-expressing thymocytes support iNKT cell development, but thymocyte-restricted expression of CD1d gives rise to Ag hyperresponsive iNKT cells. We hypothesized that iNKT cells require functional education by CD1d(+) cells other than thymocytes to set their correct responsiveness. In mice that expressed CD1d only on thymocytes, hyperresponsive iNKT cells in the periphery expressed significantly reduced levels of tyrosine phosphatase SHP-1, a negative regulator of TCR signaling. Accordingly, heterozygous SHP-1 mutant mice displaying reduced SHP-1 expression developed a comparable population of Ag hyperresponsive iNKT cells. Restoring nonthymocyte CD1d expression in transgenic mice normalized SHP-1 expression and iNKT cell reactivity. Radiation chimeras revealed that CD1d(+) dendritic cells supported iNKT cell upregulation of SHP-1 and decreased responsiveness after thymic emigration. Hence, dendritic cells functionally educate iNKT cells by tuning SHP-1 expression to limit reactivity.


Subject(s)
Dendritic Cells/metabolism , Natural Killer T-Cells/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Animals , Antigens, CD1d/genetics , Antigens, CD1d/metabolism , Cluster Analysis , Dendritic Cells/immunology , Gene Expression Profiling , Gene Expression Regulation , Genomic Imprinting , Humans , Hypersensitivity/genetics , Hypersensitivity/immunology , Mice , Mice, Knockout , Natural Killer T-Cells/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Thymus Gland/immunology , Thymus Gland/metabolism
20.
Cell Mol Life Sci ; 70(2): 239-55, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22766971

ABSTRACT

Type 1 diabetes is an autoimmune disease characterized by the destruction of insulin-producing pancreatic ß-cells. Even though extensive scientific research has yielded important insights into the immune mechanisms involved in pancreatic ß-cell destruction, little is known about the events that trigger the autoimmune process. Recent epidemiological and experimental data suggest that environmental factors are involved in this process. In this review, we discuss the role of viruses as an environmental factor on the development of type 1 diabetes, and the immune mechanisms by which they can trigger or protect against this pathology.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/virology , Enterovirus Infections/immunology , Enterovirus Infections/virology , Enterovirus/pathogenicity , Insulin-Secreting Cells/immunology , Animals , B-Lymphocytes/immunology , Diabetes Mellitus, Type 1/etiology , Diabetes Mellitus, Type 1/genetics , Humans , Hygiene Hypothesis , Insulin-Secreting Cells/virology , Mice , Risk Factors , Socioeconomic Factors , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...