Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
1.
Diabetes Care ; 38(1): 43-50, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25414154

ABSTRACT

OBJECTIVE: Disease-associated T-cell autoreactivities are seen in most type 1 diabetic patients and are thought to emerge before islet autoantibodies, but host factors that impact autoimmune elements remain uncertain. We assessed if adiposity and measures of insulin sensitivity impact T- and B-cell autoimmunity in children with insulin-requiring diabetes. RESEARCH DESIGN AND METHODS: Insulin-requiring children and adolescents diagnosed between January 2004 and June 2008 were studied (n = 261): age 9.7 ± 4 years, 92% white, and 60% male. T-cell responses to 10 diabetes-associated antigens, ß-cell autoantibodies (GADA, IA-2A, IAA, and ICA), BMI z score (BMIz), and waist percentile were measured at onset and 3 months later. RESULTS: All but one subject had either T- or B-cell autoimmunity. Diabetes-associated T-cell autoreactivities were found in 92% of subjects. Higher amplitude T-cell autoreactivities to neuronal diabetes-associated autoantigens were seen in those with the highest BMIz quintile, BMI ≥85th percentile (P < 0.05), and waist circumference ≥85th percentile (P < 0.05). There were no relationships between the number of T-cell reactivities or T-cell diversity with adiposity measures or autoantibody number or type. Patients with positive T-cell reactivities but without autoantibodies had the highest BMIz (P = 0.006). CONCLUSIONS: Our observations link obesity and diabetes-related autoimmunity, suggesting an amplification of neuronal T-cell autoimmunity associated with adiposity and/or insulin resistance, with obesity-related inflammation possibly enhancing islet autoimmunity.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Insulin/therapeutic use , Pediatric Obesity/immunology , T-Lymphocytes/immunology , Adolescent , Alleles , Autoantibodies/immunology , Autoantigens/immunology , Autoimmunity/immunology , B-Lymphocytes/immunology , Body Mass Index , C-Peptide/blood , Cell Proliferation , Child , Cross-Sectional Studies , Diabetes Mellitus, Type 1/genetics , Female , Follow-Up Studies , Humans , Insulin-Secreting Cells/immunology , Islets of Langerhans/immunology , Islets of Langerhans/metabolism , Male , Waist Circumference
2.
Clin Vaccine Immunol ; 21(2): 203-11, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24334687

ABSTRACT

Broad consensus assigns T lymphocytes fundamental roles in inflammatory, infectious, and autoimmune diseases. However, clinical investigations have lacked fully characterized and validated procedures, equivalent to those of widely practiced biochemical tests with established clinical roles, for measuring core T cell functions. The Trial to Reduce Insulin-dependent diabetes mellitus in the Genetically at Risk (TRIGR) type 1 diabetes prevention trial used consecutive measurements of T cell proliferative responses in prospectively collected fresh heparinized blood samples shipped by courier within North America. In this article, we report on the quality control implications of this simple and pragmatic shipping practice and the interpretation of positive- and negative-control analytes in our assay. We used polyclonal and postvaccination responses in 4,919 samples to analyze the development of T cell immunocompetence. We have found that the vast majority of the samples were viable up to 3 days from the blood draw, yet meaningful responses were found in a proportion of those with longer travel times. Furthermore, the shipping time of uncooled samples significantly decreased both the viabilities of the samples and the unstimulated cell counts in the viable samples. Also, subject age was significantly associated with the number of unstimulated cells and T cell proliferation to positive activators. Finally, we observed a pattern of statistically significant increases in T cell responses to tetanus toxin around the timing of infant vaccinations. This assay platform and shipping protocol satisfy the criteria for robust and reproducible long-term measurements of human T cell function, comparable to those of established blood biochemical tests. We present a stable technology for prospective disease-relevant T cell analysis in immunological diseases, vaccination medicine, and measurement of herd immunity.


Subject(s)
Blood/immunology , Specimen Handling/methods , T-Lymphocytes/immunology , T-Lymphocytes/physiology , Cell Proliferation , Cell Survival , Child , Child, Preschool , Cohort Studies , Female , Humans , Infant , Male , North America , Prospective Studies , Time Factors
3.
Diabetes ; 57(4): 918-28, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18198358

ABSTRACT

OBJECTIVE: Type 1 diabetes reflects autoimmune destruction of beta-cells and peri-islet Schwann cells (pSCs), but the mechanisms of pSC death and the T-cell epitopes involved remain unclear. RESEARCH DESIGN AND METHODS: Primary pSC cultures were generated and used as targets in cytotoxic T-lymphocyte (CTL) assays in NOD mice. Cognate interaction between pSC and CD8(+) T-cells was assessed by transgenic restoration of beta2-microglobulin (beta2m) to pSC in NOD.beta2m(-/-) congenics. I-A(g7) and K(d) epitopes in the pSC antigen glial fibrillary acidic protein (GFAP) were identified by peptide mapping or algorithms, respectively, and the latter tested by immunotherapy. RESULTS: pSC cultures did not express major histocompatibility complex (MHC) class II and were lysed by ex vivo CTLs from diabetic NOD mice. In vivo, restoration of MHC class I in GFAP-beta2m transgenics significantly accelerated adoptively transferred diabetes. Target epitopes in the pSC autoantigen GFAP were mapped to residues 79-87 and 253-261 for K(d) and 96-110, 116-130, and 216-230 for I-A(g7). These peptides were recognized spontaneously in NOD spleens as early as 2.5 weeks of age, with proliferative responses peaking around weaning and detectable lifelong. Several were also recognized by T-cells from new-onset type 1 diabetic patients. NOD mouse immunotherapy at 8 weeks with the CD8(+) T-cell epitope, GFAP 79-87 but not 253-261, significantly inhibited type 1 diabetes and was associated with reduced gamma-interferon production to whole protein GFAP. CONCLUSIONS: Collectively, these findings elucidate a role for pSC-specific CD8(+) T-cells in islet inflammation and type 1 diabetes pathogenesis, further supporting neuronal involvement in beta-cell demise.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Animals , Diabetes Mellitus, Type 1/pathology , Female , Glial Fibrillary Acidic Protein/genetics , Insulin/genetics , Insulin-Secreting Cells/immunology , Insulin-Secreting Cells/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Transgenic , Ovulation , Schwann Cells/pathology , T-Lymphocytes, Cytotoxic/immunology , beta 2-Microglobulin/deficiency , beta 2-Microglobulin/genetics
4.
J Clin Immunol ; 27(4): 388-96, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17436063

ABSTRACT

INTRODUCTION: Although myelin autoimmunity is known to be a major factor in the pathogenesis of multiple sclerosis (MS), the role of nonmyelin antigens is less clear. Given the complexity of this disease, it is possible that autoimmunity against nonmyelin antigens also has a pathogenic role. Autoantibodies against enolase and arrestin have previously been reported in MS patients. The T-cell response to these antigens, however, has not been established. METHODS: Thirty-five patients with MS were recruited, along with thirty-five healthy controls. T-cell proliferative responses against non-neuronal enolase, neuron-specific enolase (NSE), retinal arrestin, beta-arrestin, and myelin basic protein were determined. RESULTS: MS patients had a greater prevalence of positive T-cell proliferative responses to NSE, retinal arrestin, and beta-arrestin than healthy controls (p<0.0001). The proliferative response against NSE, retinal arrestin, and beta-arrestin correlated with the response against myelin basic protein (p < or = 0.004). Furthermore, the proliferative response against retinal arrestin was correlated to beta-arrestin (p<0.0001), whereas there was no such correlation between non-neuronal enolase and NSE (p = 0.23). DISCUSSION: There is accumulating evidence to suggest that the pathogenesis of MS involves more than just myelin autoimmunity/destruction. Autoimmunity against nonmyelin antigens may be a component of this myriad of immunopathological events. NSE, retinal arrestin, and beta-arrestin are novel nonmyelin autoantigens that deserve further investigation in this respect. Autoimmunity against these antigens may be linked to neurodegeneration, defective remyelination, and predisposition to uveitis in multiple sclerosis. Further investigation of the role of these antigens in MS is warranted.


Subject(s)
Arrestin/immunology , Autoantigens/immunology , Multiple Sclerosis/immunology , Phosphopyruvate Hydratase/immunology , Adult , Amino Acid Sequence , Arrestin/pharmacology , Autoantigens/pharmacology , Cell Proliferation/drug effects , Cells, Cultured , Dose-Response Relationship, Immunologic , Female , Humans , Immunodominant Epitopes , Lymphocyte Activation/drug effects , Male , Molecular Sequence Data , Phosphopyruvate Hydratase/pharmacology , Reference Values , Sequence Alignment , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
5.
Nat Med ; 9(2): 198-205, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12539039

ABSTRACT

Pancreatic islets of Langerhans are enveloped by peri-islet Schwann cells (pSC), which express glial fibrillary acidic protein (GFAP) and S100beta. pSC-autoreactive T- and B-cell responses arise in 3- to 4-week-old diabetes-prone non-obese diabetic (NOD) mice, followed by progressive pSC destruction before detectable beta-cell death. Humans with probable prediabetes generate similar autoreactivities, and autoantibodies in islet-cell autoantibody (lCA) -positive sera co-localize to pSC. Moreover, GFAP-specific NOD T-cell lines transferred pathogenic peri-insulitis to NOD/severe combined immunodeficient (NOD/SCID) mice, and immunotherapy with GFAP or S100beta prevented diabetes. pSC survived in rat insulin promoter Iymphocytic choriomeningitis virus (rip-LCMV) glycoprotein/CD8+ T-cell receptor(gp) double-transgenic mice with virus-induced diabetes, suggesting that pSC death is not an obligate consequence of local inflammation and beta-cell destruction. However, pSC were deleted in spontaneously diabetic NOD mice carrying the CD8+/8.3 T-cell receptor transgene, a T cell receptor commonly expressed in earliest islet infiltrates. Autoimmune targeting of pancreatic nervous system tissue elements seems to be an integral, early part of natural type 1 diabetes.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Islets of Langerhans/immunology , Animals , Autoantibodies/analysis , Autoantibodies/immunology , Base Sequence , DNA Primers , Female , Fluorescent Antibody Technique , Glial Fibrillary Acidic Protein/metabolism , Islets of Langerhans/pathology , Male , Mice , Mice, Inbred Strains , Nerve Growth Factors/metabolism , S100 Calcium Binding Protein beta Subunit , S100 Proteins/metabolism , Schwann Cells/immunology , Schwann Cells/metabolism , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...