Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Gastroenterology ; 162(4): 1288-1302.e16, 2022 04.
Article in English | MEDLINE | ID: mdl-34973295

ABSTRACT

BACKGROUND & AIMS: Alteration of the gut microbiota is implicated in the development of autoimmune type 1 diabetes (T1D), as shown in humans and the nonobese diabetic (NOD) mouse model. However, how gut dysbiosis arises and promotes the autoimmune response remains an open question. We investigated whether early events affecting the intestinal homeostasis in newborn NOD mice may explain the development of the autoimmune response in the adult pancreas. METHODS: We profiled the transcriptome and the microbiota in the colon between newborn NOD mice and nonautoimmune strains. We identified a seminal defect in the intestinal homeostasis of newborn NOD mice and deciphered the mechanism linking this defect to the diabetogenic response in the adult. RESULTS: We determined that the cathelicidin-related antimicrobial peptide (CRAMP) expression was defective in the colon of newborn NOD mice, allowing inducing dysbiosis. Dysbiosis stimulated the colonic epithelial cells to produce type I interferons that pathologically imprinted the local neonatal immune system. This pathological immune imprinting later promoted the pancreatic autoimmune response in the adult and the development of diabetes. Increasing colonic CRAMP expression in newborn NOD mice by means of local CRAMP treatment or CRAMP-expressing probiotic restored colonic homeostasis and halted the diabetogenic response, preventing autoimmune diabetes. CONCLUSIONS: We identified whether a defective colonic expression in the CRAMP antimicrobial peptide induces dysbiosis, contributing to autoimmunity in the pancreas. Hence, the manipulation of intestinal antimicrobial peptides may be considered a relevant therapeutic approach to prevent autoimmune diabetes in at-risk children.


Subject(s)
Diabetes Mellitus, Type 1 , Gastrointestinal Microbiome , Animals , Antimicrobial Cationic Peptides , Antimicrobial Peptides , Autoimmunity , Diabetes Mellitus, Type 1/prevention & control , Dysbiosis/pathology , Gastrointestinal Microbiome/physiology , Humans , Mice , Mice, Inbred NOD , Pancreas/pathology , Cathelicidins
2.
Anal Bioanal Chem ; 415(20): 4961-4971, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37338567

ABSTRACT

Bile acids (BAs) are a complex class of metabolites that have been described as specific biomarkers of gut microbiota activity. The development of analytical methods allowing the quantification of an ample spectrum of BAs in different biological matrices is needed to enable a wider implementation of BAs as complementary measures in studies investigating the functional role of the gut microbiota. This work presents results from the validation of a targeted ultra-high performance liquid chromatography-tandem mass spectrometry (UHPLC-MS/MS) method for the determination of 28 BAs and six sulfated BAs, covering primary, secondary, and conjugated BAs. The analysis of 73 urine and 20 feces samples was used to test the applicability of the method. Concentrations of BAs in human urine and murine feces were reported, ranging from 0.5 to 50 nmol/g creatinine and from 0.012 to 332 nmol/g, respectively. Seventy-nine percent of BAs present in human urine samples corresponded to secondary conjugated BAs, while 69% of BAs present in murine feces corresponded to primary conjugated BAs. Glycocholic acid sulfate (GCA-S) was the most abundant BA in human urine samples, while taurolithocholic acid was the lowest concentrated compound detected. In murine feces, the most abundant BAs were α-murocholic, deoxycholic, dehydrocholic, and ß-murocholic acids, while GCA-S was the lowest concentrated BA. The presented method is a non-invasive approach for the simultaneous assessment of BAs and sulfated BAs in urine and feces samples, and the results will serve as a knowledge base for future translational studies focusing on the role of the microbiota in health.


Subject(s)
Bile Acids and Salts , Tandem Mass Spectrometry , Humans , Mice , Animals , Bile Acids and Salts/analysis , Tandem Mass Spectrometry/methods , Sulfates/analysis , Chromatography, High Pressure Liquid/methods , Feces/chemistry
3.
Proc Natl Acad Sci U S A ; 116(30): 15140-15149, 2019 07 23.
Article in English | MEDLINE | ID: mdl-31182588

ABSTRACT

Low-grade intestinal inflammation and alterations of gut barrier integrity are found in patients affected by extraintestinal autoimmune diseases such as type 1 diabetes (T1D), but a direct causal link between enteropathy and triggering of autoimmunity is yet to be established. Here, we found that onset of autoimmunity in preclinical models of T1D is associated with alterations of the mucus layer structure and loss of gut barrier integrity. Importantly, we showed that breakage of the gut barrier integrity in BDC2.5XNOD mice carrying a transgenic T cell receptor (TCR) specific for a beta cell autoantigen leads to activation of islet-reactive T cells within the gut mucosa and onset of T1D. The intestinal activation of islet-reactive T cells requires the presence of gut microbiota and is abolished when mice are depleted of endogenous commensal microbiota by antibiotic treatment. Our results indicate that loss of gut barrier continuity can lead to activation of islet-specific T cells within the intestinal mucosa and to autoimmune diabetes and provide a strong rationale to design innovative therapeutic interventions in "at-risk" individuals aimed at restoring gut barrier integrity to prevent T1D occurrence.


Subject(s)
Colitis/immunology , Diabetes Mellitus, Type 1/genetics , Gastrointestinal Microbiome/immunology , Intestinal Mucosa/immunology , Islets of Langerhans/immunology , T-Lymphocytes/immunology , Animals , Bacteria/classification , Bacteria/genetics , Bacteria/immunology , Blood Glucose/immunology , Blood Glucose/metabolism , Colitis/chemically induced , Colitis/pathology , Diabetes Mellitus, Type 1/pathology , Disease Models, Animal , Female , Gene Expression , Humans , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Islets of Langerhans/pathology , Mice , Mice, Inbred NOD , Mice, Transgenic , Permeability , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Sodium Dodecyl Sulfate/administration & dosage , Survival Analysis , T-Lymphocytes/pathology , Transgenes
4.
Immunity ; 34(4): 602-15, 2011 Apr 22.
Article in English | MEDLINE | ID: mdl-21511186

ABSTRACT

This study describes a CD4+ T helper (Th) cell subset marked by coexpression of the cytokine interleukin 21 (IL-21) and the gut-homing chemokine receptor CCR9. Although CCR9+ Th cells were observed in healthy mice and humans, they were enriched in the inflamed pancreas and salivary glands of NOD mice and in the circulation of Sjögren's syndrome patients. CCR9+ Th cells expressed large amounts of IL-21, inducible T cell costimulator (ICOS), and the transcription factors Bcl6 and Maf, and also supported antibody production from B cells, thereby resembling T follicular B helper (Tfh) cells. However, in contrast to Tfh cells, CCR9+ Th cells displayed limited expression of CXCR5 and the targets of CCR9+ Th cells were CD8+ T cells whose responsiveness to IL-21 was necessary for the development of diabetes. Thus, CCR9+ Th cells are a subset of IL-21-producing T helper cells that influence regional specification of autoimmune diseases that affect accessory organs of the digestive system.


Subject(s)
Autoimmunity , Interleukins/immunology , Islets of Langerhans/immunology , Receptors, CCR/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , B-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Movement , Cell Survival , Diabetes Mellitus/immunology , Female , Humans , Interleukins/biosynthesis , Mice , Mice, Knockout , Receptors, Interleukin-21/deficiency , Receptors, Interleukin-21/immunology , Sjogren's Syndrome/immunology , T-Lymphocytes, Helper-Inducer/cytology
5.
J Immunol ; 197(8): 3271-3280, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27605013

ABSTRACT

NKT17 cells represent a functional subset of Vα14 invariant NKT (iNKT) cells with important effector functions in infections and autoimmune diseases. The mechanisms that drive NKT17 cell differentiation in the thymus are still largely unknown. The percentage of NKT17 cells has a high variability between murine strains due to differential thymic differentiation. For example, the NOD strain carries a high percentage and absolute numbers of NKT17 cells compared with other strains. In this study, we used the NOD mouse model to analyze what regulates NKT17 cell frequency in the thymus and peripheral lymphoid organs. In accordance with previous studies showing that the zinc finger transcription factor Th-POK is a key negative regulator of thymic NKT17 cell differentiation in the thymus, our data indicate that excessive NKT17 cell frequency in NOD mice correlates with defective Th-POK expression by thymic Vα14iNKT cells. Moreover, we found that Th-POK expression is under epigenetic regulation mediated by microRNA-133b whose expression is reduced in Vα14iNKT cells of NOD mice. We also demonstrated in a conditional knockout model of dendritic cell (DC) depletion (CD11cCreXDTA.B6 and CD11cCreRosa26DTA.NOD mice) that DCs play a crucial role in regulating Vα14iNKT cell maturation and their acquisition of an NKT17 cytokine secretion phenotype in the thymus. Overall, our data show that mechanisms regulating NKT17 cell differentiation are unique and completely different from those of Vα14iNKT cells. Specifically, we found that epigenetic regulation through microRNA-133b-regulated Th-POK expression and signals provided by DCs are fundamental for thymic NKT17 cell differentiation.


Subject(s)
Cell Differentiation/genetics , Dendritic Cells/cytology , MicroRNAs/genetics , Natural Killer T-Cells/cytology , Signal Transduction/genetics , Thymus Gland/cytology , Transcription Factors/genetics , Animals , Cell Line , Dendritic Cells/immunology , Female , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Natural Killer T-Cells/metabolism , Thymus Gland/metabolism , Transcription Factors/biosynthesis , Transcription Factors/metabolism
6.
Clin Immunol ; 178: 29-38, 2017 05.
Article in English | MEDLINE | ID: mdl-26732858

ABSTRACT

Mast cells (MCs) are innate immune cells that exert positive and negative immune modulatory functions capable to enhance or limit the intensity and/or duration of adaptive immune responses. Although MCs are crucial to regulate T cell immunity, their action in the pathogenesis of autoimmune diseases is still debated. Here we demonstrate that MCs play a crucial role in T1D pathogenesis so that their selective depletion in conditional MC knockout NOD mice protects them from the disease. MCs of diabetic NOD mice are overly inflammatory and secrete large amounts of IL-6 that favors differentiation of IL-17-secreting T cells at the site of autoimmunity. Moreover, while MCs of control mice acquire an IL-10+ phenotype upon interaction with FoxP3+ Treg cells, MCs of NOD mice do not undergo this tolerogenic differentiation. Our data indicate that overly inflammatory MCs unable to acquire a tolerogenic IL-10+ phenotype contribute to the pathogenesis of autoimmune T1D.


Subject(s)
Autoimmunity/immunology , Diabetes Mellitus, Type 1/immunology , Immune Tolerance/immunology , Islets of Langerhans/immunology , Mast Cells/immunology , Animals , Blood Glucose/metabolism , Chymases/genetics , Diabetes Mellitus, Type 1/metabolism , Flow Cytometry , Forkhead Transcription Factors/metabolism , Immunohistochemistry , Inflammation , Interleukin-10/immunology , Interleukin-17/immunology , Interleukin-6/immunology , Laser Capture Microdissection , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, Transgenic , Real-Time Polymerase Chain Reaction , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Th17 Cells/immunology
7.
Microorganisms ; 12(5)2024 May 03.
Article in English | MEDLINE | ID: mdl-38792760

ABSTRACT

The role of Bifidobacterium species and microbial metabolites such as short-chain fatty acids (SCFAs) and human milk oligosaccharides in controlling intestinal inflammation and the pathogenesis of obesity and type 1 diabetes (T1D) has been largely studied in recent years. This paper discusses the discovery of signature biomarkers for obesity and T1D based on data from a novel test for profiling several Bifidobacterium species, combined with metabolomic analysis. Through the NUTRISHIELD clinical study, a total of 98 children were recruited: 40 healthy controls, 40 type 1 diabetics, and 18 obese children. Bifidobacterium profiles were assessed in stool samples through an innovative test allowing high taxonomic resolution and precise quantification, while SCFAs and branched amino acids were measured in urine samples through gas chromatography-mass spectrometry (GC-MS). KIDMED questionnaires were used to evaluate the children's dietary habits and correlate them with the Bifidobacterium and metabolomic profiles. We found that B. longum subs. infantis and B. breve were higher in individuals with obesity, while B. bifidum and B. longum subs. longum were lower compared to healthy individuals. In individuals with T1D, alterations were found at the metabolic level, with an overall increase in the level of the most measured metabolites. The high taxonomic resolution of the Bifidobacterium test used meant strong correlations between the concentrations of valine and isoleucine, and the relative abundance of some Bifidobacterium species such as B. longum subs. infantis, B. breve, and B. bifidum could be observed.

8.
EBioMedicine ; 91: 104567, 2023 May.
Article in English | MEDLINE | ID: mdl-37062177

ABSTRACT

BACKGROUND: In preclinical models of Type 1 Diabetes (T1D) the integrity of the gut barrier (GB) is instrumental to avoid dysregulated crosstalk between the commensal microbiota and immune cells and to prevent autoimmunity. The GB is composed of the intestinal epithelial barrier (IEB) and of the mucus layer containing mucins and antimicrobial peptides (AMPs) that are crucial to maintain immune tolerance. In preclinical models of T1D the alterations of the GB primarily affect the mucus layer. In human T1D increased gut permeability and IEB damage have been demonstrated but the integrity of the mucus layer was never assessed. METHODS: We evaluated GB integrity by measuring serological markers of IEB damage (serological levels of zonulin) and bacterial translocation such as lipopolysaccharide binding protein (LBP) and myeloid differentiation protein 2 (MD2), and mRNA expression of tight junction proteins, mucins and AMPs in intestinal tissue of T1D patients and healthy controls (HC). Simultaneously, we performed immunological profiling on intestinal tissue and 16S rRNA analysis on the mucus-associated gut microbiota (MAGM). FINDINGS: Our data show a GB damage with mucus layer alterations and reduced mRNA expression of several mucins (MUC2, MUC12, MUC13, MUC15, MUC20, MUC21) and AMPs (HD4 and HD5) in T1D patients. Mucus layer alterations correlated with reduced relative abundance of short chain fatty acids (SCFA)-producing bacteria such as Bifidobacterium dentium, Clostridium butyricum and Roseburia intestinalis that regulate mucin expression and intestinal immune homeostasis. In T1D patients we also found intestinal immune dysregulation with higher percentages of effector T cells such as T helper (Th) 1, Th17 and TNF-α+ T cells. INTERPRETATION: Our data show that mucus layer alterations are present in T1D subjects and associated with dysbiosis and immune dysregulation. FUNDING: Research Grants from the Juvenile Diabetes Foundation (Grant 1-INO-2018-640-A-N to MF and 2-SRA-2019-680-S-B to JD) and from the Italian Ministry of Health (Grant RF19-12370721 to MF).


Subject(s)
Diabetes Mellitus, Type 1 , Humans , Intestinal Mucosa/metabolism , Dysbiosis/metabolism , RNA, Ribosomal, 16S/metabolism , Mucins/metabolism , Mucus/metabolism , RNA, Messenger/metabolism
9.
J Immunol ; 185(12): 7317-29, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21078913

ABSTRACT

Invariant NKT (iNKT) cells play an effector/adjuvant function during antimicrobial and antitumoral immunity and a regulatory role to induce immune tolerance and prevent autoimmunity. iNKT cells that differentially modulate adaptive immunity do not bear a unique phenotype and/or specific cytokine secretion profile, thus opening questions on how a single T cell subset can exert opposite immunological tasks. In this study, we show that iNKT cells perform their dual roles through a single mechanism of action relying on the cognate interaction with myeloid dendritic cells (DCs) and leading to opposite effects depending on the presence of other maturation stimuli simultaneously acting on DCs. The contact of murine purified iNKT cells with immature autologous DCs directly triggers the tolerogenic maturation of DCs, rendering them able to induce regulatory T cell differentiation and prevent autoimmune diabetes in vivo. Conversely, the interaction of the same purified iNKT cells with DCs, in the presence of simultaneous TLR4 stimulation, significantly enhances proinflammatory DC maturation and IL-12 secretion. The different iNKT cell effects are mediated through distinct mechanisms and activation of different molecular pathways within the DC: CD1d signaling and activation of the ERK1/2 pathway for the tolerogenic action, and CD40-CD40L interaction and NF-κB activation for the adjuvant effect. Our data suggest that the DC decision to undergo proinflammatory or tolerogenic maturation results from the integration of different signals received at the time of iNKT cell contact and could have important therapeutic implications for exploiting iNKT cell adjuvant/regulatory properties in autoimmune diseases, infections, and cancer.


Subject(s)
Antigens, CD1d/immunology , Cell Communication/immunology , Dendritic Cells/immunology , Immune Tolerance , Natural Killer T-Cells/immunology , Signal Transduction/immunology , Toll-Like Receptor 4/immunology , Animals , Antigens, CD1d/genetics , CD40 Antigens/genetics , CD40 Antigens/immunology , CD40 Ligand/genetics , CD40 Ligand/immunology , Cell Communication/genetics , Cell Differentiation/genetics , Cell Differentiation/immunology , Dendritic Cells/cytology , Interleukin-12/genetics , Interleukin-12/immunology , Mice , Mice, Inbred NOD , Mice, Knockout , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/immunology , Myeloid Cells/immunology , NF-kappa B/genetics , NF-kappa B/immunology , Signal Transduction/genetics , Toll-Like Receptor 4/genetics
10.
Front Immunol ; 13: 1089987, 2022.
Article in English | MEDLINE | ID: mdl-36713378

ABSTRACT

Introduction: The integrity of the gut barrier (GB) is fundamental to regulate the crosstalk between the microbiota and the immune system and to prevent inflammation and autoimmunity at the intestinal level but also in organs distal from the gut such as the pancreatic islets. In support to this idea, we recently demonstrated that breakage of GB integrity leads to activation of islet-reactive T cells and triggers autoimmune Type 1 Diabetes (T1D). In T1D patients as in the NOD mice, the spontaneous model of autoimmune diabetes, there are alterations of the GB that specifically affect structure and composition of the mucus layer; however, it is yet to be determined whether a causal link between breakage of the GB integrity and occurrence of autoimmune T1D exists. Methods: Here we restored GB integrity in the NOD mice through administration of an anti-inflammatory diet (AID- enriched in soluble fiber inulin and omega 3-PUFA) and tested the effect on T1D pathogenesis. Results: We found that the AID prevented T1D in NOD mice by restoring GB integrity with increased mucus layer thickness and higher mRNA transcripts of structural (Muc2) and immunoregulatory mucins (Muc1 and Muc3) as well as of tight junction proteins (claudin1). Restoration of GB integrity was linked to reduction of intestinal inflammation (i.e., reduced expression of IL-1ß, IL-23 and IL-17 transcripts) and expansion of regulatory T cells (FoxP3+ Treg cells and IL-10+ Tr1 cells) at the expenses of effector Th1/Th17 cells in the intestine, pancreatic lymph nodes (PLN) and intra-islet lymphocytes (IIL) of AID-fed NOD mice. Importantly, the restoration of GB integrity and immune homeostasis were associated with enhanced concentrations of anti-inflammatory metabolites of the ω3/ω6 polyunsaturated fatty acids (PUFA) and arachidonic pathways and modifications of the microbiome profile with increased relative abundance of mucus-modulating bacterial species such as Akkermansia muciniphila and Akkermansia glycaniphila. Discussion: Our data provide evidence that the restoration of GB integrity and intestinal immune homeostasis through administration of a tolerogenic AID that changed the gut microbial and metabolic profiles prevents autoimmune T1D in preclinical models.


Subject(s)
Diabetes Mellitus, Type 1 , Mice , Animals , Mice, Inbred NOD , Inulin/pharmacology , Diet , Inflammation , Homeostasis , Anti-Inflammatory Agents
11.
J Clin Invest ; 118(5): 1806-14, 2008 May.
Article in English | MEDLINE | ID: mdl-18431516

ABSTRACT

Successful transplantation requires the prevention of allograft rejection and, in the case of transplantation to treat autoimmune disease, the suppression of autoimmune responses. The standard immunosuppressive treatment regimen given to patients with autoimmune type 1 diabetes who have received an islet transplant results in the loss of T cells. In many other situations, the immune system responds to T cell loss through cytokine-dependant homeostatic proliferation of any remaining T cells. Here we show that T cell loss after islet transplantation in patients with autoimmune type 1 diabetes was associated with both increased serum concentrations of IL-7 and IL-15 and in vivo proliferation of memory CD45RO(+) T cells, highly enriched in autoreactive glutamic acid decarboxylase 65-specific T cell clones. Immunosuppression with FK506 and rapamycin after transplantation resulted in a chronic homeostatic expansion of T cells, which acquired effector function after immunosuppression was removed. In contrast, the cytostatic drug mycophenolate mofetil efficiently blocked homeostatic T cell expansion. We propose that the increased production of cytokines that induce homeostatic expansion could contribute to recurrent autoimmunity in transplanted patients with autoimmune disease and that therapy that prevents the expansion of autoreactive T cells will improve the outcome of islet transplantation.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/therapy , Interleukin-15/immunology , Interleukin-7/immunology , Islets of Langerhans Transplantation , T-Lymphocyte Subsets/immunology , T-Lymphocytes/immunology , Adult , Animals , Diabetes Mellitus, Type 1/blood , Homeostasis , Humans , Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/therapeutic use , Interleukin-15/blood , Interleukin-7/blood , Male , Mice , Mycophenolic Acid/analogs & derivatives , Mycophenolic Acid/pharmacology , Mycophenolic Acid/therapeutic use , Sirolimus/pharmacology , Sirolimus/therapeutic use , T-Lymphocytes/drug effects , Tacrolimus/pharmacology , Tacrolimus/therapeutic use
12.
J Immunol ; 181(2): 869-77, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18606638

ABSTRACT

The regulatory function of invariant NKT (iNKT) cells for tolerance induction and prevention of autoimmunity is linked to a specific cytokine profile that comprises the secretion of type 2 cytokines like IL-4 and IL-10 (NKT2 cytokine profile). The mechanism responsible for iNKT cell differentiation toward a type 2 phenotype is unknown. Herein we show that costimulatory signals provided by the surface receptor signaling lymphocytic activation molecule (SLAM) on myeloid dendritic cells (mDC) to iNKT cells is crucial for NKT2 orientation. Additionally, we demonstrate that the impaired acquisition of an NKT2 cytokine phenotype in nonobese diabetic (NOD) mice that spontaneously develop autoimmune diabetes is due to defective SLAM-induced signals generated by NOD mDC. Mature mDC of C57BL/6 mice express SLAM and induce C57BL/6 or NOD iNKT cells to acquire a predominant NKT2 cytokine phenotype in response to antigenic stimulation with the iNKT cell-specific Ag, the alpha-galactosylceramide. In contrast, mature NOD mDC express significantly lower levels of SLAM and are unable to promote GATA-3 (the SLAM-induced intracellular signal) up-regulation and IL-4/IL-10 production in iNKT cells from NOD or C57BL/6 mice. NOD mice carry a genetic defect of the Slamf1 gene that is associated with reduced SLAM expression on double-positive thymocytes and altered iNKT cell development in the thymus. Our data suggest that the genetic Slamf1 defect in NOD mice also affects SLAM expression on other immune cells such as the mDC, thus critically impairing the peripheral differentiation of iNKT cells toward a regulatory NKT2 type.


Subject(s)
Antigens, CD/metabolism , Dendritic Cells/immunology , GATA3 Transcription Factor/metabolism , Killer Cells, Natural/immunology , Receptors, Cell Surface/metabolism , T-Lymphocyte Subsets/immunology , Animals , Antigens, CD/immunology , Cell Differentiation , Dendritic Cells/metabolism , GATA3 Transcription Factor/immunology , Interleukin-10/immunology , Interleukin-10/metabolism , Interleukin-4/immunology , Interleukin-4/metabolism , Killer Cells, Natural/cytology , Killer Cells, Natural/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Receptors, Cell Surface/immunology , Signaling Lymphocytic Activation Molecule Family Member 1 , T-Lymphocyte Subsets/metabolism
13.
Article in English | MEDLINE | ID: mdl-32973682

ABSTRACT

The human pancreas, like almost all organs in the human body, is immunologically tolerated despite the presence of innate and adaptive immune cells that promptly mediate protective immune responses against pathogens in situ. The PD-1/PD-L1 inhibitory pathway seems to play a key role in the maintenance of immune tolerance systemically and within the pancreatic tissue. Tissue resident memory T cells (TRM), T regulatory cells (Treg), macrophages and even ß cells exhibit PD-1 or PD-L1 expression that contributes in controlling pancreatic immune homeostasis and tolerance. Dysregulation of the PD-1/PD-L1 axis as shown by animal studies and our recent experience with checkpoint inhibitory blockade in humans can lead to immune dysfunctions leading to chronic inflammatory disease and to type 1 diabetes (T1D) in genetically susceptible individuals. In this review, we discuss the role of the PD-1/PD-L1 axis in pancreatic tissue homeostasis and tolerance, speculate how genetic and environmental factors can regulate the PD-1/PD-L1 pathway, and discuss PD-1/PD-L1-based therapeutic approaches for pancreatic islet transplantation and T1D treatment.


Subject(s)
B7-H1 Antigen/metabolism , Diabetes Mellitus, Type 1/immunology , Immune Tolerance/physiology , Pancreas/immunology , Programmed Cell Death 1 Receptor/metabolism , Diabetes Mellitus, Type 1/metabolism , Humans , Pancreas/metabolism
15.
Front Immunol ; 10: 1937, 2019.
Article in English | MEDLINE | ID: mdl-31475000

ABSTRACT

The intestinal barrier provides the host with a strong defense line against the external environment playing also a pivotal role in the crosstalk between the gut microbiota and the immune system. Notably, increasing lines of evidence concerning autoimmune disorders such as Multiple Sclerosis (MS) report an imbalance in both intestinal microbiota composition and mucosal immunity activation, along with an alteration of gut barrier permeability, suggesting this complex network plays a crucial role in modulating the course of autoimmune responses occurring in tissues outside the gut such as the central nervous system (CNS). Here, we review current knowledge on how gut inflammation and breakage of gut barrier integrity modulates the interplay between the commensal gut microbiota and the immune system and its role in shaping brain immunity.


Subject(s)
Autoimmune Diseases/immunology , Brain/immunology , Gastrointestinal Microbiome/immunology , Tight Junctions/immunology , Autoimmunity/immunology , Bacteria/immunology , Dysbiosis/immunology , Humans , Immunity, Mucosal/immunology , Inflammation/immunology , Th17 Cells/immunology
16.
Front Biosci ; 13: 4827-37, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18508548

ABSTRACT

Invariant NKT cells are important regulators of T cell immunity and autoimmunity. In this review we describe evidence that supports their regulatory role in the prevention of autoimmune disease. Moreover, we will discuss the current knowledge on iNKT cell biology, antigen recognition, acquisition of a specific cytokine profile, and mechanism of action that suggest a key role for iNKT cells as negative regulators of autoimmune diseases.


Subject(s)
Autoimmunity , Killer Cells, Natural/immunology , T-Lymphocytes/immunology , Animals , Autoimmune Diseases/immunology , Cytokines/immunology , Diabetes Mellitus, Type 1/immunology , Disease Models, Animal , Glycolipids/immunology , Humans , Immune Tolerance , Immunity, Innate , Lupus Erythematosus, Systemic/immunology , Multiple Sclerosis/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology
17.
Front Immunol ; 9: 1752, 2018.
Article in English | MEDLINE | ID: mdl-30105027

ABSTRACT

iNKT cells play different immune function depending on their cytokine-secretion phenotype. iNKT17 cells predominantly secrete IL-17 and have an effector and pathogenic role in the pathogenesis of autoimmune diseases such as type 1 diabetes (T1D). In line with this notion, non-obese diabetic (NOD) mice that spontaneously develop T1D have an increased percentage of iNKT17 cells compared to non-autoimmune strains of mice. The factors that regulate iNKT cell expansion and acquisition of a specific iNKT17 cell phenotype are unclear. Here, we demonstrate that the percentage of iNKT17 cells is increased in the gut more than peripheral lymphoid organs of NOD mice, thus suggesting that the intestinal environment promotes iNKT17 cell differentiation in these mice. Increased intestinal iNKT17 cell differentiation in NOD mice is associated with the presence of pro-inflammatory IL-6-secreting dendritic cells that could contribute to iNKT cell expansion and iNKT17 cell differentiation. In addition, we found that increased iNKT17 cell differentiation in the large intestine of NOD mice is associated with a specific gut microbiota profile. We demonstrated a positive correlation between percentage of intestinal iNKT17 cells and bacterial strain richness (α-diversity) and relative abundance of Bacterioidales strains. On the contrary, the relative abundance of the anti-inflammatory Clostridiales strains negatively correlates with the intestinal iNKT17 cell frequency. Considering that iNKT17 cells play a key pathogenic role in T1D, our data support the notion that modulation of iNKT17 cell differentiation through gut microbiota changes could have a beneficial effect in T1D.


Subject(s)
Clostridiales , Gastrointestinal Microbiome , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Lymphocyte Count , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Animals , Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/metabolism , Female , Intestinal Mucosa/pathology , Mice , Mice, Inbred NOD
18.
Sci Adv ; 3(7): e1700492, 2017 07.
Article in English | MEDLINE | ID: mdl-28706993

ABSTRACT

T helper 17 (TH17) cells are key players in multiple sclerosis (MS), and studies in animal models demonstrated that effector TH17 cells that trigger brain autoimmunity originate in the intestine. We validate in humans the crucial role of the intestinal environment in promoting TH17 cell expansion in MS patients. We found that increased frequency of TH17 cells correlates with high disease activity and with specific alterations of the gut mucosa-associated microbiota in MS patients. By using 16S ribosomal RNA sequencing, we analyzed the microbiota isolated from small intestinal tissues and found that MS patients with high disease activity and increased intestinal TH17 cell frequency showed a higher Firmicutes/Bacteroidetes ratio, increased relative abundance of Streptococcus, and decreased Prevotella strains compared to healthy controls and MS patients with no disease activity. We demonstrated that the intestinal TH17 cell frequency is inversely related to the relative abundance of Prevotella strains in the human small intestine. Our data demonstrate that brain autoimmunity is associated with specific microbiota modifications and excessive TH17 cell expansion in the human intestine.


Subject(s)
Gastrointestinal Microbiome , Lymphocyte Count , Multiple Sclerosis/diagnosis , Multiple Sclerosis/etiology , Peyer's Patches , Th17 Cells/immunology , Th17 Cells/metabolism , Adult , Biomarkers , Biopsy , Female , Humans , Immunity, Mucosal , Magnetic Resonance Imaging , Male , Middle Aged , Severity of Illness Index , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
20.
J Diabetes Res ; 2016: 7569431, 2016.
Article in English | MEDLINE | ID: mdl-26779542

ABSTRACT

The gut microbiota modulates the autoimmune pathogenesis of type 1 diabetes (T1D) via mechanisms that remain largely unknown. The inflammasome components are innate immune sensors that are highly influenced by the gut environment and play pivotal roles in maintaining intestinal immune homeostasis. In this study we show that modifications of the gut microbiota induced by oral treatment with Lactobacillaceae-enriched probiotic VSL#3, alone or in combination with retinoic acid (RA), protect NOD mice from T1D by affecting inflammasome at the intestinal level. In particular, we show that VSL#3 treatment inhibits IL-1ß expression while enhancing release of protolerogenic components of the inflammasome, such as indoleamine 2,3-dioxygenase (IDO) and IL-33. Those modifications of the intestinal microenvironment in VSL#3-treated NOD mice modulate gut immunity by promoting differentiation of tolerogenic CD103(+) DCs and reducing differentiation/expansion of Th1 and Th17 cells in the intestinal mucosa and at the sites of autoimmunity, that is, within the pancreatic lymph nodes (PLN) of VSL#3-treated NOD mice. Our data provide a link between dietary factors, microbiota composition, intestinal inflammation, and immune homeostasis in autoimmune diabetes and could pave the way for new therapeutic approaches aimed at changing the intestinal microenvironment with probiotics to counterregulate autoimmunity and prevent T1D.


Subject(s)
Autoimmunity , Diabetes Mellitus, Type 1/prevention & control , Gastrointestinal Microbiome , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Inflammasomes/metabolism , Intestines/microbiology , Lactobacillaceae/growth & development , Probiotics/administration & dosage , Administration, Oral , Age Factors , Animals , Cellular Microenvironment , Diabetes Mellitus, Type 1/enzymology , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/microbiology , Disease Models, Animal , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Inflammasomes/immunology , Interleukin-1beta/metabolism , Interleukin-33/metabolism , Intestines/enzymology , Intestines/immunology , Lactobacillaceae/immunology , Mice, Inbred NOD , Th1 Cells/immunology , Th1 Cells/metabolism , Th1 Cells/microbiology , Th17 Cells/immunology , Th17 Cells/metabolism , Th17 Cells/microbiology , Tretinoin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL