Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Nat Commun ; 14(1): 7770, 2023 Nov 27.
Article in English | MEDLINE | ID: mdl-38012160

ABSTRACT

The transgenic 116C-NOD mouse strain exhibits a prevalent Th17 phenotype, and reduced type 1 diabetes (T1D) compared to non-obese diabetic (NOD) mice. A cohousing experiment between both models revealed lower T1D incidence in NOD mice cohoused with 116C-NOD, associated with gut microbiota changes, reduced intestinal permeability, shifts in T and B cell subsets, and a transition from Th1 to Th17 responses. Distinct gut bacterial signatures were linked to T1D in each group. Using a RAG-2-/- genetic background, we found that T cell alterations promoted segmented filamentous bacteria proliferation in young NOD and 116C-NOD, as well as in immunodeficient NOD.RAG-2-/- and 116C-NOD.RAG-2-/- mice across all ages. Bifidobacterium colonization depended on lymphocytes and thrived in a non-diabetogenic environment. Additionally, 116C-NOD B cells in 116C-NOD.RAG-2-/- mice enriched the gut microbiota in Adlercreutzia and reduced intestinal permeability. Collectively, these results indicate reciprocal modulation between gut microbiota and the immune system in rodent T1D models.


Subject(s)
B-Lymphocyte Subsets , Diabetes Mellitus, Type 1 , Gastrointestinal Microbiome , Mice , Animals , Diabetes Mellitus, Type 1/genetics , Mice, Inbred NOD , Gastrointestinal Microbiome/genetics , B-Lymphocytes
2.
Front Endocrinol (Lausanne) ; 14: 1176566, 2023.
Article in English | MEDLINE | ID: mdl-37334284

ABSTRACT

Introduction: During the development of Autoimmune Diabetes (AD) an autoimmune attack against the Peripheral Nervous System occurs. To gain insight into this topic, analyses of Dorsal Root Ganglia (DRG) from Non-Obese Diabetic (NOD) mice were carried out. Methods: Histopathological analysis by electron and optical microscopy in DRG samples, and mRNA expression analyzes by the microarray technique in DRG and blood leukocyte samples from NOD and C57BL/6 mice were performed. Results: The results showed the formation of cytoplasmic vacuoles in DRG cells early in life that could be related to a neurodegenerative process. In view of these results, mRNA expression analyses were conducted to determine the cause and/or the molecules involved in this suspected disorder. The results showed that DRG cells from NOD mice have alterations in the transcription of a wide range of genes, which explain the previously observed alterations. In addition, differences in the transcription genes in white blood cells were also detected. Discussion: Taken together, these results indicate that functional defects are not only seen in beta cells but also in DRG in NOD mice. These results also indicate that these defects are not a consequence of the autoimmune process that takes place in NOD mice and suggest that they may be involved as triggers for its development.


Subject(s)
Diabetes Mellitus, Type 1 , Mice , Animals , Mice, Inbred NOD , Diabetes Mellitus, Type 1/metabolism , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Mice, Inbred C57BL , Gene Expression , RNA, Messenger/metabolism
3.
Nutrients ; 15(10)2023 May 22.
Article in English | MEDLINE | ID: mdl-37242297

ABSTRACT

Adipocyte dysfunction is the driver of obesity and correlates with insulin resistance and the onset of type 2 diabetes. Protein kinase N1 (PKN1) is a serine/threonine kinase that has been shown to contribute to Glut4 translocation to the membrane and glucose transport. Here, we evaluated the role of PKN1 in glucose metabolism under insulin-resistant conditions in primary visceral adipose tissue (VAT) from 31 patients with obesity and in murine 3T3-L1 adipocytes. In addition, in vitro studies in human VAT samples and mouse adipocytes were conducted to investigate the role of PKN1 in the adipogenic maturation process and glucose homeostasis control. We show that insulin-resistant adipocytes present a decrease in PKN1 activation levels compared to nondiabetic control counterparts. We further show that PKN1 controls the adipogenesis process and glucose metabolism. PKN1-silenced adipocytes present a decrease in both differentiation process and glucose uptake, with a concomitant decrease in the expression levels of adipogenic markers, such as PPARγ, FABP4, adiponectin and CEBPα. Altogether, these results point to PKN1 as a regulator of key signaling pathways involved in adipocyte differentiation and as an emerging player of adipocyte insulin responsiveness. These findings may provide new therapeutic approaches for the management of insulin resistance in type 2 diabetes.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin Resistance , Mice , Humans , Animals , Diabetes Mellitus, Type 2/metabolism , Adipogenesis , Adipocytes/metabolism , Obesity/metabolism , Insulin/metabolism , PPAR gamma/metabolism , Glucose/metabolism , 3T3-L1 Cells , Cell Differentiation
4.
ACS Omega ; 6(44): 29882-29892, 2021 Nov 09.
Article in English | MEDLINE | ID: mdl-34778661

ABSTRACT

Halloysite clay nanotubes (HNTs) have been proposed as highly biocompatible for several biomedical applications. Various polymers have been used to functionalize HNTs, but scarce information exists about polystyrene for this purpose. This work evaluated polystyrene-functionalized HNTs (FHNTs) by comparing its effects with non-FHNTs and innocuous talc powder on in vitro and in vivo models. Monocyte-derived human or murine macrophages and the RAW 264.7 cell line were treated with 0.01, 0.1, 1, and 100 µg mL-1 FHNTs, HNTs, or talc to evaluate the cytotoxic and cytokine response. Our results show that nanoclays did not cause cytotoxic damage to macrophages. Only the 100 µg mL-1 concentration induced slight proinflammatory cytokine production at short exposure, followed by an anti-inflammatory response that increases over time. CD1 mice treated with a single dose of 1, 2.5, or 5 mg Kg-1 of FHNTs or HNTs by oral and inhalation routes caused aluminum accumulation in the kidneys and lungs, without bodily signs of distress or histopathological changes in any treated mice, evaluated at 48 h and 30 days post-treatment. Nanoclay administration simultaneously by four different parenteral routes (20 mg Kg-1) or the combination of administration routes (parenteral + oral or parenteral + inhalation; 25 mg Kg-1) showed accumulation on the injection site and slight surrounding inflammation 30 days post-treatment. CD1 mice chronically exposed to HNTs or FHNTs in the bedding material (ca 1 mg) throughout the parental generation and two successive inbred generations for 8 months did not cause any inflammatory process or damage to the abdominal organs and the reproductive system of the mice of any of the generations, did not affect the number of newborn mice and their survival, and did not induce congenital malformations in the offspring. FHNTs showed a slightly less effect than HNTs in all experiments, suggesting that functionalization makes them less cytotoxic. Doses of up to 25 mg Kg-1 by different administration routes and permanent exposure to 1 mg of HNTs or FHNTs for 8 months seem safe for CD1 mice. Our in vivo and in vitro results indicate that nanoclays are highly biocompatible, supporting their possible safe use for future biomedical and general-purpose applications.

5.
Front Immunol ; 12: 634797, 2021.
Article in English | MEDLINE | ID: mdl-33664748

ABSTRACT

Background: Pancreatic islets are exposed to strong pro-apoptotic stimuli: inflammation and hyperglycemia, during the progression of the autoimmune diabetes (T1D). We found that the Cdk11(Cyclin Dependent Kinase 11) is downregulated by inflammation in the T1D prone NOD (non-obese diabetic) mouse model. The aim of this study is to determine the role of CDK11 in the pathogenesis of T1D and to assess the hierarchical relationship between CDK11 and Cyclin D3 in beta cell viability, since Cyclin D3, a natural ligand for CDK11, promotes beta cell viability and fitness in front of glucose. Methods: We studied T1D pathogenesis in NOD mice hemideficient for CDK11 (N-HTZ), and, in N-HTZ deficient for Cyclin D3 (K11HTZ-D3KO), in comparison to their respective controls (N-WT and K11WT-D3KO). Moreover, we exposed pancreatic islets to either pro-inflammatory cytokines in the presence of increasing glucose concentrations, or Thapsigargin, an Endoplasmic Reticulum (ER)-stress inducing agent, and assessed apoptotic events. The expression of key ER-stress markers (Chop, Atf4 and Bip) was also determined. Results: N-HTZ mice were significantly protected against T1D, and NS-HTZ pancreatic islets exhibited an impaired sensitivity to cytokine-induced apoptosis, regardless of glucose concentration. However, thapsigargin-induced apoptosis was not altered. Furthermore, CDK11 hemideficiency did not attenuate the exacerbation of T1D caused by Cyclin D3 deficiency. Conclusions: This study is the first to report that CDK11 is repressed in T1D as a protection mechanism against inflammation-induced apoptosis and suggests that CDK11 lies upstream Cyclin D3 signaling. We unveil the CDK11/Cyclin D3 tandem as a new potential intervention target in T1D.


Subject(s)
Apoptosis/drug effects , Blood Glucose/metabolism , Cyclin-Dependent Kinases/metabolism , Cytokines/pharmacology , Diabetes Mellitus, Type 1/enzymology , Inflammation/enzymology , Insulin-Secreting Cells/drug effects , Protein Serine-Threonine Kinases/physiology , Activating Transcription Factor 4/metabolism , Animals , Autoimmunity/drug effects , Cyclin D3/genetics , Cyclin D3/metabolism , Cyclin-Dependent Kinases/genetics , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Disease Models, Animal , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/drug effects , Heat-Shock Proteins/metabolism , Inflammation/blood , Inflammation/genetics , Inflammation/pathology , Insulin-Secreting Cells/enzymology , Insulin-Secreting Cells/pathology , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Protein Serine-Threonine Kinases/genetics , Thapsigargin/pharmacology , Tissue Culture Techniques , Transcription Factor CHOP/metabolism
6.
Front Immunol ; 10: 1732, 2019.
Article in English | MEDLINE | ID: mdl-31428087

ABSTRACT

Previous studies indicate that B-lymphocytes play a key role activating diabetogenic T-lymphocytes during the development of autoimmune diabetes. Recently, two transgenic NOD mouse models were generated: the NOD-PerIg and the 116C-NOD mice. In NOD-PerIg mice, B-lymphocytes acquire an activated proliferative phenotype and support accelerated autoimmune diabetes development. In contrast, in 116C-NOD mice, B-lymphocytes display an anergic-like phenotype delaying autoimmune diabetes onset and decreasing disease incidence. The present study further evaluates the T- and B-lymphocyte phenotype in both models. In islet-infiltrating B-lymphocytes (IIBLs) from 116C-NOD mice, the expression of H2-Kd and H2-Ag7 is decreased, whereas that of BAFF, BAFF-R, and TACI is increased. In contrast, IIBLs from NOD-PerIg show an increase in CD86 and FAS expression. In addition, islet-infiltrating T-lymphocytes (IITLs) from NOD-PerIg mice exhibit an increase in PD-1 expression. Moreover, proliferation assays indicate a high capacity of B-lymphocytes from NOD-PerIg mice to secrete high amounts of cytokines and induce T-lymphocyte activation compared to 116C B-lymphocytes. This functional variability between 116C and PerIg B-lymphocytes ultimately results in differences in the ability to shape T-lymphocyte phenotype. These results support the role of B-lymphocytes as key regulators of T-lymphocytes in autoimmune diabetes and provide essential information on the phenotypic characteristics of the T- and B-lymphocytes involved in the autoimmune response in autoimmune diabetes.


Subject(s)
B-Lymphocyte Subsets/immunology , Diabetes Mellitus, Type 1/immunology , T-Lymphocyte Subsets/immunology , Animals , Antigen Presentation , Autoimmunity , Clonal Anergy , Cytokines/blood , Diabetes Mellitus, Type 1/pathology , Disease Models, Animal , Female , Gene Expression Profiling , Genes, Immunoglobulin , Immunophenotyping , Intercellular Signaling Peptides and Proteins/biosynthesis , Intercellular Signaling Peptides and Proteins/genetics , Islets of Langerhans/immunology , Islets of Langerhans/pathology , Lymphocyte Activation , Lymphopoiesis , Male , Mice , Mice, Inbred NOD , Mice, Transgenic , Specific Pathogen-Free Organisms , Spleen/immunology , Spleen/pathology
7.
Sci Rep ; 8(1): 8106, 2018 05 25.
Article in English | MEDLINE | ID: mdl-29802270

ABSTRACT

Type 1 diabetes can be overcome by regulatory T cells (Treg) in NOD mice yet an efficient method to generate and maintain antigen-specific Treg is difficult to come by. Here, we devised a combination therapy of peptide/MHC tetramers and IL-2/anti-IL-2 monoclonal antibody complexes to generate antigen-specific Treg and maintain them over extended time periods. We first optimized treatment protocols conceived to obtain an improved islet-specific Treg/effector T cell ratio that led to the in vivo expansion and activation of these Treg as well as to an improved suppressor function. Optimized protocols were applied to treatment for testing diabetes prevention in NOD mice as well as in an accelerated T cell transfer model of T1D. The combined treatment led to robust protection against diabetes, and in the NOD model, to a close to complete prevention of insulitis. Treatment was accompanied with increased secretion of IL-10, detectable in total splenocytes and in Foxp3- CD4 T cells. Our data suggest that a dual protection mechanism takes place by the collaboration of Foxp3+ and Foxp3- regulatory cells. We conclude that antigen-specific Treg are an important target to improve current clinical interventions against this disease.


Subject(s)
Antibodies, Monoclonal/immunology , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/therapy , Histocompatibility Antigens/chemistry , Interleukin-2/immunology , Peptides/chemistry , T-Lymphocytes, Regulatory/immunology , Amino Acid Sequence , Animals , Diabetes Mellitus, Type 1/drug therapy , Female , Mice , Peptides/pharmacology , Phenotype , Protein Multimerization , Protein Structure, Quaternary , T-Lymphocytes, Regulatory/drug effects
8.
Mol Carcinog ; 55(5): 411-9, 2016 May.
Article in English | MEDLINE | ID: mdl-25647613

ABSTRACT

Oxaliplatin-resistant LoVo colon cancer cells overexpressing c-MET and VEGFR-1 were selected to study several signaling pathways involved in chemoresistance, as well as the effect of increasing amounts of VEGF in the regulation of c-MET. In comparison with chemosensitive LoVo colon cancer cells, oxaliplatin-resistant cells (LoVoR) overexpress and phosphorylate c-MET, upregulate the expression of transmembrane and soluble VEGFR-1 and, unexpectedly, downregulate VEGF. In addition, LoVoR cells activate other transduction pathways involved in chemoresistance such as Akt, ß-catenin-TCF4 and E-cadherin. While c-MET is phosphorylated in LoVoR cells expressing low levels of VEGF, c-MET phosphorylation decreases when recombinant VEGF is added into the culture medium. Inhibition of c-MET by VEGF is mediated by VEGFR-1, since phosphorylation of c-MET in the presence of VEGF is restored after silencing VEGFR-1. Dephosphorylation of c-MET by VEGF suggests that tumors coexpressing VEGFR-1 and c-MET may activate c-MET as a result of anti-VEGF therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Colonic Neoplasms/drug therapy , Drug Resistance, Neoplasm , Organoplatinum Compounds/pharmacology , Proto-Oncogene Proteins c-met/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Drug Resistance, Neoplasm/drug effects , Gene Expression Regulation, Neoplastic , HT29 Cells , Humans , Oxaliplatin , Phosphorylation/drug effects , Proto-Oncogene Proteins c-met/genetics , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor Receptor-1/genetics
9.
Eur J Immunol ; 46(3): 593-608, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26639224

ABSTRACT

Autoreactive B lymphocytes play a key role as APCs in diaebetogenesis. However, it remains unclear whether B-cell tolerance is compromised in NOD mice. Here, we describe a new B lymphocyte transgenic NOD mouse model, the 116C-NOD mouse, where the transgenes derive from an islet-infiltrating B lymphocyte of a (8.3-NODxNOR) F1 mouse. The 116C-NOD mouse produces clonal B lymphocytes with pancreatic islet beta cell specificity. The incidence of T1D in 116C-NOD mice is decreased in both genders when compared with NOD mice. Moreover, several immune selection mechanisms (including clonal deletion and anergy) acting on the development, phenotype, and function of autoreactive B lymphocytes during T1D development have been identified in the 116C-NOD mouse. Surprisingly, a more accurate analysis revealed that, despite their anergic phenotype, 116C B cells express some costimulatory molecules after activation, and induce a T-cell shift toward a Th17 phenotype. Furthermore, this shift on T lymphocytes seems to occur not only when both T and B cells contact, but also when helper T (Th) lineage is established. The 116C-NOD mouse model could be useful to elucidate the mechanisms involved in the generation of Th-cell lineages.


Subject(s)
B-Lymphocytes/immunology , Clonal Anergy , Diabetes Mellitus, Type 1/immunology , Disease Models, Animal , Immune Tolerance/genetics , Lymphocyte Activation , Th17 Cells/immunology , Animals , Clonal Deletion , Cytokines/genetics , Cytokines/immunology , Immune Tolerance/immunology , Mice, Inbred NOD , Mice, Transgenic , Phenotype , Spleen/anatomy & histology , Spleen/cytology , Spleen/immunology , Transgenes
10.
Proc Natl Acad Sci U S A ; 111(33): E3405-14, 2014 Aug 19.
Article in English | MEDLINE | ID: mdl-25092329

ABSTRACT

Type 1 diabetes is an autoimmune condition caused by the lymphocyte-mediated destruction of the insulin-producing ß cells in pancreatic islets. We aimed to identify final molecular entities targeted by the autoimmune assault on pancreatic ß cells that are causally related to ß cell viability. Here, we show that cyclin D3 is targeted by the autoimmune attack on pancreatic ß cells in vivo. Cyclin D3 is down-regulated in a dose-dependent manner in ß cells by leukocyte infiltration into the islets of the nonobese diabetic (NOD) type 1 diabetes-prone mouse model. Furthermore, we established a direct in vivo causal link between cyclin D3 expression levels and ß-cell fitness and viability in the NOD mice. We found that changes in cyclin D3 expression levels in vivo altered the ß-cell apoptosis rates, ß-cell area homeostasis, and ß-cell sensitivity to glucose without affecting ß-cell proliferation in the NOD mice. Cyclin D3-deficient NOD mice exhibited exacerbated diabetes and impaired glucose responsiveness; conversely, transgenic NOD mice overexpressing cyclin D3 in ß cells exhibited mild diabetes and improved glucose responsiveness. Overexpression of cyclin D3 in ß cells of cyclin D3-deficient mice rescued them from the exacerbated diabetes observed in transgene-negative littermates. Moreover, cyclin D3 overexpression protected the NOD-derived insulinoma NIT-1 cell line from cytokine-induced apoptosis. Here, for the first time to our knowledge, cyclin D3 is identified as a key molecule targeted by autoimmunity that plays a nonredundant, protective, and cell cycle-independent role in ß cells against inflammation-induced apoptosis and confers metabolic fitness to these cells.


Subject(s)
Cell Cycle , Cyclin D3/physiology , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/metabolism , Islets of Langerhans/pathology , Animals , Apoptosis , Cytokines/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/pathology , Mice , Mice, Inbred NOD
11.
J Immunol ; 192(7): 3080-90, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24610011

ABSTRACT

Autoreactive B cells are essential for the pathogenesis of type 1 diabetes. The genesis and dynamics of autoreactive B cells remain unknown. In this study, we analyzed the immune response in the NOD mouse model to the neuronal protein peripherin (PRPH), a target Ag of islet-infiltrating B cells. PRPH autoreactive B cells recognized a single linear epitope of this protein, in contrast to the multiple epitope recognition commonly observed during autoreactive B cell responses. Autoantibodies to this epitope were also detected in the disease-resistant NOR and C57BL/6 strains. To specifically detect the accumulation of these B cells, we developed a novel approach, octameric peptide display, to follow the dynamics and localization of anti-PRPH B cells during disease progression. Before extended insulitis was established, anti-PRPH B cells preferentially accumulated in the peritoneum. Anti-PRPH B cells were likewise detected in C57BL/6 mice, albeit at lower frequencies. As disease unfolded in NOD mice, anti-PRPH B cells invaded the islets and increased in number at the peritoneum of diabetic but not prediabetic mice. Isotype-switched B cells were only detected in the peritoneum. Anti-PRPH B cells represent a heterogeneous population composed of both B1 and B2 subsets. In the spleen, anti-PRPH B cell were predominantly in the follicular subset. Therefore, anti-PRPH B cells represent a heterogeneous population that is generated early in life but proliferates as diabetes is established. These findings on the temporal and spatial progression of autoreactive B cells should be relevant for our understanding of B cell function in diabetes pathogenesis.


Subject(s)
B-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Islets of Langerhans/immunology , Peripherins/immunology , Amino Acid Sequence , Animals , Autoantibodies/immunology , Autoantibodies/metabolism , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Blotting, Western , Cell Line, Tumor , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Disease Progression , Epitope Mapping/methods , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte/metabolism , Female , Islets of Langerhans/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Microscopy, Fluorescence , Molecular Sequence Data , Peripherins/genetics , Peripherins/metabolism , Peritoneum/immunology , Peritoneum/metabolism , Protein Isoforms/immunology , Spleen/immunology , Spleen/metabolism
12.
Eur J Immunol ; 43(11): 2969-79, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23925934

ABSTRACT

The cholera toxin B subunit (CTB) has been used as adjuvant to improve oral vaccine delivery in type 1 diabetes. The effect of CTB/peptide formulations on Ag-specific CD4(+) T cells has remained largely unexplored. Here, using tetramer analysis, we investigated how oral delivery of CTB fused to two CD4(+) T-cell epitopes, the BDC-2.5 T-cell 2.5 mi mimotope and glutamic acid decarboxylase (GAD) 286-300, affected diabetogenic CD4(+) T cells in nonobese diabetic (NOD) mice. When administered i.p., CTB-2.5 mi activated 2.5 mi(+) T cells and following intragastric delivery generated Ag-specific Foxp3(+) Treg and Th2 cells. While 2.5 mi(+) and GAD-specific T cells were tolerized in diabetes-resistant NODxB6.Foxp3(EGFP) F1 and nonobese resistant (NOR) mice, this did not occur in NOD mice. This indicated that NOD mice had a recessive genetic resistance to induce oral tolerance to both CTB-fused epitopes. In contrast to NODxB6.Foxp3(EGFP) F1 mice, oral treatment in NOD mice lead to strong 2.5 mi(+) T-cell activation and the sequestration of these cells to the effector-memory pool. Oral treatment of NOD mice with CTB-2.5 mi failed to prevent diabetes. These findings underline the importance of investigating the effect of oral vaccine formulations on diabetogenic T cells as in selected cases they may have counterproductive consequences in human patients.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Cholera Toxin/immunology , Diabetes Mellitus, Type 1/immunology , Epitopes, T-Lymphocyte/immunology , Peptide Fragments/immunology , Recombinant Fusion Proteins/immunology , Administration, Oral , Animals , CD4-Positive T-Lymphocytes/immunology , Cholera Toxin/administration & dosage , Cholera Vaccines/immunology , Glutamate Decarboxylase/immunology , Immune Tolerance/immunology , Interferon-gamma/biosynthesis , Interleukin-4/biosynthesis , Lymphocyte Activation/immunology , Mice , Mice, Inbred NOD , Peptide Fragments/administration & dosage
14.
Eur J Immunol ; 41(5): 1344-51, 2011 May.
Article in English | MEDLINE | ID: mdl-21469125

ABSTRACT

CD4(+) T lymphocytes are required to induce spontaneous autoimmune diabetes in the NOD mouse. Since pancreatic ß cells upregulate Fas expression upon exposure to pro-inflammatory cytokines, we studied whether the diabetogenic action of CD4(+) T lymphocytes depends on Fas expression on target cells. We assayed the diabetogenic capacity of NOD spleen CD4(+) T lymphocytes when adoptively transferred into a NOD mouse model combining: (i) Fas-deficiency, (ii) FasL-deficiency, and (iii) SCID mutation. We found that CD4(+) T lymphocytes require Fas expression in the recipients' target cells to induce diabetes. IL-1ß has been described as a key cytokine involved in Fas upregulation on mouse ß cells. We addressed whether CD4(+) T cells require IL-1ß to induce diabetes. We also studied spontaneous diabetes onset in NOD/IL-1 converting enzyme-deficient mice, in NOD/IL-1ß-deficient mice, and CD4(+) T-cell adoptively transferred diabetes into NOD/SCID IL-1ß-deficient mice. Neither IL-1ß nor IL-18 are required for either spontaneous or CD4(+) T-cell adoptively transferred diabetes. We conclude that CD4(+) T-cell-mediated ß-cell damage in autoimmune diabetes depends on Fas expression, but not on IL-1ß unveiling the existing redundancy regarding the cytokines involved in Fas upregulation on NOD ß cells in vivo.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , fas Receptor/metabolism , Adoptive Transfer , Animals , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Fas Ligand Protein/deficiency , Fas Ligand Protein/metabolism , Genotype , Insulin-Secreting Cells/immunology , Insulin-Secreting Cells/pathology , Interleukin-18/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , fas Receptor/deficiency , fas Receptor/genetics
15.
J Immunol ; 186(7): 4078-87, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21346228

ABSTRACT

CD4 T cells are crucial effectors in the pathology of type 1 diabetes (T1D). Successful therapeutic interventions for prevention and cure of T1D in humans are still elusive. Recent research efforts have focused on the manipulation of T cells by treatment with DNA. In this paper, we studied the effects of a DNA treatment strategy designed to target antigenic peptides to the lysosomal compartment on a monospecific T cell population termed 2.5mi(+) T cells that shares reactivity with the diabetogenic T cell clone BDC-2.5 in the NOD mouse. MHC class II tetramer analysis showed that repeated administrations were necessary to expand 2.5mi(+) T cells in vivo. This expansion was independent of Ag presentation by B cells. A single peptide epitope was sufficient to induce protection against T1D, which was not due to Ag-specific T cell anergy. Typical Th2 cytokines such as IL-10 or IL-4 were undetectable in 2.5mi(+) T cells, arguing against a mechanism of immune deviation. Instead, the expanded 2.5mi(+) T cell population produced IFN-γ similar to 2.5mi(+) T cells from naive mice. Protection against T1D by DNA treatment was completely lost in NOD.CD28(-/-) mice which are largely deficient of natural regulatory T cells (Treg). Although Ag-specific Foxp3(+) Treg did not expand in response to DNA treatment, diabetes onset was delayed in Treg-reconstituted and DNA-treated NOD.SCID mice. These observations provide evidence for a Treg-mediated protective mechanism that is independent of the expansion or de novo generation of Ag-specific Treg.


Subject(s)
Diabetes Mellitus, Type 1/prevention & control , Drug Delivery Systems/methods , Immune Tolerance , Lysosomes/immunology , Peptide Fragments/agonists , Peptide Fragments/metabolism , Vaccines, DNA/agonists , Vaccines, DNA/immunology , Adoptive Transfer/methods , Animals , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Dose-Response Relationship, Immunologic , Female , Immune Tolerance/genetics , Lysosomes/genetics , Lysosomes/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Peptide Fragments/administration & dosage , Spleen/cytology , Spleen/immunology , Spleen/transplantation , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/transplantation , Vaccines, DNA/administration & dosage , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
16.
J Immunol ; 180(2): 1189-98, 2008 Jan 15.
Article in English | MEDLINE | ID: mdl-18178859

ABSTRACT

Cyclin-dependent kinase 4 (Cdk4) plays a central role in perinatal pancreatic beta cell replication, thus becoming a potential target for therapeutics in autoimmune diabetes. Its hyperactive form, Cdk4R24C, causes beta cell hyperplasia without promoting hypoglycemia in a nonautoimmune-prone mouse strain. In this study, we explore whether beta cell hyperproliferation induced by the Cdk4R24C mutation balances the autoimmune attack against beta cells inherent to the NOD genetic background. To this end, we backcrossed the Cdk4R24C knockin mice, which have the Cdk4 gene replaced by the Cdk4R24C mutated form, onto the NOD genetic background. In this study, we show that NOD/Cdk4R24C knockin mice exhibit exacerbated diabetes and insulitis, and that this exacerbated diabetic phenotype is solely due to the hyperactivity of the NOD/Cdk4R24C immune repertoire. Thus, NOD/Cdk4R24C splenocytes confer exacerbated diabetes when adoptively transferred into NOD/SCID recipients, compared with NOD/wild-type (WT) donor splenocytes. Accordingly, NOD/Cdk4R24C splenocytes show increased basal proliferation and higher activation markers expression compared with NOD/WT splenocytes. However, to eliminate the effect of the Cdk4R24C mutation specifically in the lymphocyte compartment, we introduced this mutation into NOD/SCID mice. NOD/SCID/Cdk4R24C knockin mice develop beta cell hyperplasia spontaneously. Furthermore, NOD/SCID/Cdk4R24C knockin females that have been adoptively transferred with NOD/WT splenocytes are more resistant to autoimmunity than NOD/SCID WT female. Thus, the Cdk4R24C mutation opens two avenues in the NOD model: when expressed specifically in beta cells, it provides a new potential strategy for beta cell regeneration in autoimmune diabetes, but its expression in the immune repertoire exacerbates autoimmunity.


Subject(s)
Cyclin-Dependent Kinase 4/metabolism , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/pathology , Insulin-Secreting Cells/immunology , Insulin-Secreting Cells/pathology , Animals , Autoimmunity/genetics , Cyclin-Dependent Kinase 4/genetics , Diabetes Mellitus, Type 1/genetics , Female , Hyperplasia/genetics , Hyperplasia/immunology , Hyperplasia/pathology , Immune System/enzymology , Immune System/immunology , Insulin-Secreting Cells/enzymology , Lymph Nodes/immunology , Male , Mice , Mice, Inbred NOD , Mice, SCID , Mutation , Regeneration/genetics , Sex Factors , Spleen/enzymology , Spleen/immunology , T-Lymphocytes/immunology
17.
J Autoimmun ; 24(1): 11-23, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15725572

ABSTRACT

Type 1 diabetes results from destruction of pancreatic beta cells by beta cell-specific autoreactive T cells in the nonobese diabetic (NOD) mouse. Defects in thymic negative selection are thought to result in failure to delete potential beta cell-reactive T cells, contributing to the development of autoimmune diabetes. We investigated this possibility by comparing the deletion profile of double-positive (DP) thymocytes in NOD mice with diabetes-resistant strains of mice after anti-CD3 Ab treatment to trigger the TCR-mediated signaling pathway. We found that immature NOD CD4+CD8+ DP thymocytes have a lower activation threshold than C57BL/6 and Balb/c thymocytes. This was confirmed by showing that NOD DP thymocytes have a higher level of ERK and JNK phosphorylation. The low activation threshold of immature thymocytes resulted in rapid deletion of strongly activated immature DP thymocytes by negative selection, whereas weakly activated immature thymocytes differentiated more efficiently into CD69+CD3high DP thymocytes by positive selection. SP thymocytes, particularly CD4-CD8+ T cells that were efficiently generated from activated DP thymocytes, could induce severe insulitis and diabetes in NOD.scid mice. We conclude that the development of autoreactive diabetogenic T cells results from inordinate positive selection due to the low activation threshold of DP thymocytes in NOD mice.


Subject(s)
Diabetes Mellitus, Type 1/immunology , T-Lymphocytes/immunology , Thymus Gland/cytology , Thymus Gland/immunology , Animals , Apoptosis , Cell Differentiation , Diabetes Mellitus, Type 1/metabolism , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , JNK Mitogen-Activated Protein Kinases/metabolism , Lymphocyte Activation , Mice , Mice, Inbred NOD , T-Lymphocytes/metabolism , Thymus Gland/metabolism , fas Receptor/metabolism
18.
Int Immunol ; 16(2): 257-64, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14734611

ABSTRACT

Autoimmune diabetes is characterized by an early mononuclear infiltration of pancreatic islets and later selective autoimmune destruction of insulin-producing beta cells. Lymphocyte homing receptors have been considered candidate targets to prevent autoimmune diabetes. L-selectin (CD62L) is an adhesion molecule highly expressed in naive T and B cells. It has been reported that blocking L-selectin in vivo with a specific antibody (Mel-14) partially impairs insulitis and diabetes in autoimmune diabetes-prone non-obese diabetic (NOD) mice. In the present study we aimed to elucidate whether genetic blockade of leukocyte homing into peripheral lymph nodes would prevent the development of diabetes. We backcrossed L-selectin-deficient mice onto the NOD genetic background. Surprisingly NOD/L-selectin-deficient mice exhibited unaltered islet mononuclear infiltration, timing of diabetes onset and cumulative incidence of spontaneous diabetes when compared to L-selectin-sufficient animals. CD4, CD8 T cells and B cells were present in islet infiltrates from 9-week-old L-selectin-sufficient and -deficient littermates. Moreover, total splenocytes from wild-type, heterozygous or NOD/L-selectin-deficient donor mice showed similar capability to adoptively transfer diabetes into NOD/SCID recipients. On the other hand, homing of activated, cloned insulin-specific autoaggressive CD8 T cells (TGNFC8 clone) is not affected in NOD/L-selectin-deficient recipients. We conclude that L-selectin plays a small role in the homing of autoreactive lymphocytes to regional (pancreatic) lymph nodes in NOD mice.


Subject(s)
Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , L-Selectin/genetics , L-Selectin/immunology , Adoptive Transfer , Animals , CD4 Antigens/immunology , CD8 Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/etiology , Inflammation/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Pancreas/immunology , Pancreas/pathology , Receptors, Lymphocyte Homing/immunology
19.
Int Immunol ; 15(3): 351-7, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12618479

ABSTRACT

Although the critical requirement of CD4 T cells in type I (insulin-dependent) diabetes mellitus (T1DM) has been well documented, information on the exact role(s) of CD4 T cells in T1DM development is still limited. Here, utilizing non-obese diabetic (NOD) mice deficient for CD154 (CD154-KO/NOD), we have identified a mandatory role of CD4 T cells as the functional source of CD154 in the initiation of T1DM. Without CD154, CD4 T cells were not capable of mediating help in disease development in NOD mice. In fact, full expression of CD154 on the CD4 T cells seems to be essential in the normal spontaneous development of T1DM, since no diabetes was observed in CD154(+/-) mice in which around half of CD4 T cells do not express CD154 at all, at least by the time they were 40 weeks old. It was also shown that transgenic expression of CD80 on beta cells of pancreatic islets, which is believed to provide beta cells with the ability to prime cytotoxic T lymphocytes specific for islet antigens, did not restore insulitis in CD154-KO/NOD mice. Taken collectively, these results indicated that CD4 T cells play a crucial role in T1DM as a source of CD154, and that the role of CD154 on CD4 T cells in insulitis may not be just to facilitate priming and expanding of auto-reactive CD8 T cells by activating antigen-presenting cells bearing islet antigens.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , CD40 Ligand/metabolism , Diabetes Mellitus, Type 1/etiology , Animals , B7-1 Antigen/biosynthesis , B7-1 Antigen/genetics , CD40 Ligand/genetics , CD8-Positive T-Lymphocytes/metabolism , Genotype , Mice , Mice, Inbred NOD , Mice, Transgenic , Phenotype
20.
J Immunol ; 168(7): 3635-40, 2002 Apr 01.
Article in English | MEDLINE | ID: mdl-11907129

ABSTRACT

The strong association of type 1 diabetes with specific MHC class II genes, such as I-A(g7) in nonobese diabetic mice and HLA-DQ8 in humans, suggests that MHC class II molecules play an important role in the development of the disease. To test whether human DQ8 molecules could cross the species barrier and functionally replace their murine homolog I-A(g7), we generated DQ8/BDC2.5 transgenic mice. We have shown that BDC2.5 transgenic T cells are selected on DQ8 in the thymus and cause diabetes in a manner similar to that seen when the T cells are selected on H2(g7). Splenocytes from DQ8/BDC2.5 mice also showed reactivity toward islets in vitro as seen in H-2(g7)/BDC2.5 mice. We conclude that DQ8 molecules not only share structural similarity with the murine homolog I-A(g7), but also can cross the species barrier and functionally replace I-A(g7) molecules to stimulate diabetogenic T cells and produce diabetes.


Subject(s)
Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , HLA-DQ Antigens/genetics , Histocompatibility Antigens Class II/genetics , T-Lymphocyte Subsets/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Movement/genetics , Cell Movement/immunology , Crosses, Genetic , Diabetes Mellitus, Type 1/etiology , Diabetes Mellitus, Type 1/pathology , Epitopes, T-Lymphocyte/genetics , Female , HLA-DQ Antigens/chemistry , HLA-DQ Antigens/immunology , Histocompatibility Antigens Class II/chemistry , Histocompatibility Antigens Class II/immunology , Humans , Islets of Langerhans/immunology , Islets of Langerhans/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Transgenic , Sequence Homology, Amino Acid , T-Lymphocyte Subsets/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...