Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 298
Filter
1.
Tissue Antigens ; 84(4): 361-9, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25040682

ABSTRACT

Genetic similarities between patients from the United States and South African (SA) Addison's Disease (AD) strengthen evidence for genetic association. SA-AD (n = 73), SA healthy controls (N = 78), and US-AD patients (N = 83) were genotyped for DQA1, DQB1, DRB1, and HLA-B alleles. Serum was tested for the quantity of 21OH-AA and IFNα-AA at the Barbara Davis Center. Although not as profound as in US-AD, in SA-AD 21OH-AA + subjects the predominantly associated risk haplotypes were DRB1*0301-DQB1*0201 (DR3), DRB1*04xx-DQB1*0302 (DR4), and the combined DR3/4 genotype. DQB1*0302 associated DRB1*04xx haplotypes conferred higher risk than those DRB1*04xx haplotypes associated with other DQB1 alleles. We found negative association in 21OH-AA + SA-AD for DQA1*0201-DQB1*0202 and DQA1*0101-DQB1*0501 vs SA controls, and positive association for DQA1*0401-DQB1*0402 vs US-AD. Apart from the class II DR3 haplotype, HLA-B8 did not have an independent effect; however together DR3 and HLA-B8 conferred the highest risk vs 21OH-AA negative SA-AD and SA-controls. HLA-B7 (often with DR4) conferred novel risk in 21OH-AA + SA-AD vs controls. This study represents the first comparison between South African and United States AD populations utilizing genotyping and serology performed at the same center. SA-AD and US-AD 21OH-AA + patients share common HLA risk haplotypes including DR4 (with HLA-B7) and DR3 (with HLA-B8), strengthening previously described HLA associations and implicating similar genetic etiology.


Subject(s)
Addison Disease/genetics , Autoantibodies/immunology , HLA-A2 Antigen/genetics , Steroid 21-Hydroxylase/immunology , Addison Disease/immunology , Adolescent , Adult , Female , HLA-A2 Antigen/immunology , Humans , Male , Middle Aged , Risk Factors , South Africa , United States
2.
J Steroid Biochem Mol Biol ; 133: 51-7, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22960018

ABSTRACT

The present study investigated the association between variants in the vitamin D receptor gene (VDR) and protein tyrosine phosphatase, non-receptor type 2 gene (PTPN2), as well as an interaction between VDR and PTPN2 and the risk of islet autoimmunity (IA) and progression to type 1 diabetes (T1D). The Diabetes Autoimmunity Study in the Young (DAISY) has followed children at increased risk of T1D since 1993. Of the 1692 DAISY children genotyped for VDR rs1544410, VDR rs2228570, VDR rs11568820, PTPN2 rs1893217, and PTPN2 rs478582, 111 developed IA, defined as positivity for GAD, insulin or IA-2 autoantibodies on 2 or more consecutive visits, and 38 IA positive children progressed to T1D. Proportional hazards regression analyses were conducted. There was no association between IA development and any of the gene variants, nor was there evidence of a VDR*PTPN2 interaction. Progression to T1D in IA positive children was associated with the VDR rs2228570 GG genotype (HR: 0.49, 95% CI: 0.26-0.92) and there was an interaction between VDR rs1544410 and PTPN2 rs1893217 (p(interaction)=0.02). In children with the PTPN2 rs1893217 AA genotype, the VDR rs1544410 AA/AG genotype was associated with a decreased risk of T1D (HR: 0.24, 95% CI: 0.11-0.53, p=0.0004), while in children with the PTPN2 rs1893217 GG/GA genotype, the VDR rs1544410 AA/AG genotype was not associated with T1D (HR: 1.32, 95% CI: 0.43-4.06, p=0.62). These findings should be replicated in larger cohorts for confirmation. The interaction between VDR and PTPN2 polymorphisms in the risk of progression to T1D offers insight concerning the role of vitamin D in the etiology of T1D.


Subject(s)
Diabetes Mellitus, Type 1/genetics , Islets of Langerhans/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 2/genetics , Receptors, Calcitriol/genetics , Autoantibodies/blood , Autoimmunity/genetics , Child , Child, Preschool , Cohort Studies , Diabetes Mellitus, Type 1/etiology , Diabetes Mellitus, Type 1/immunology , Disease Progression , Female , Glutamate Decarboxylase/immunology , Humans , Infant , Insulin Antibodies/blood , Male , Polymorphism, Single Nucleotide , Prospective Studies , Risk Factors
3.
Diabetologia ; 55(4): 996-1000, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22278338

ABSTRACT

AIMS/HYPOTHESIS: Over 50 regions of the genome have been associated with type 1 diabetes risk, mainly using large case/control collections. In a recent genome-wide association (GWA) study, 18 novel susceptibility loci were identified and replicated, including replication evidence from 2,319 families. Here, we, the Type 1 Diabetes Genetics Consortium (T1DGC), aimed to exclude the possibility that any of the 18 loci were false-positives due to population stratification by significantly increasing the statistical power of our family study. METHODS: We genotyped the most disease-predicting single-nucleotide polymorphisms at the 18 susceptibility loci in 3,108 families and used existing genotype data for 2,319 families from the original study, providing 7,013 parent-child trios for analysis. We tested for association using the transmission disequilibrium test. RESULTS: Seventeen of the 18 susceptibility loci reached nominal levels of significance (p < 0.05) in the expanded family collection, with 14q24.1 just falling short (p = 0.055). When we allowed for multiple testing, ten of the 17 nominally significant loci reached the required level of significance (p < 2.8 × 10(-3)). All susceptibility loci had consistent direction of effects with the original study. CONCLUSIONS/INTERPRETATION: The results for the novel GWA study-identified loci are genuine and not due to population stratification. The next step, namely correlation of the most disease-associated genotypes with phenotypes, such as RNA and protein expression analyses for the candidate genes within or near each of the susceptibility regions, can now proceed.


Subject(s)
Diabetes Mellitus, Type 1/genetics , Genetic Loci , Genetic Predisposition to Disease , White People/genetics , Genotype , Humans , Polymorphism, Single Nucleotide
4.
Clin Exp Immunol ; 166(3): 317-24, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22059988

ABSTRACT

Highly sensitive and specific radioimmunoassays have been validated for autoantibodies reacting with the four major autoantigens identified so far in autoimmune diabetes. However, the analysis of this large number of autoantigens has increased the costs and time necessary for complete autoantibody screenings. Our aim was to demonstrate that it is possible to detect the immunoreactivity against a combination of four different autoantigens by a single assay, this representing a rapid, low-cost first approach to evaluate humoral autoimmunity in diabetes. By using this novel multi-autoantigen radioimmunoassay (MAA), in subsequent steps we analysed 830 sera, 476 of known and 354 of unknown diabetes-specific immunoreactivity, collected from various groups of individuals including type 1 and type 2 diabetes patients, autoantibody-positive patients with a clinical diagnosis of type 2 diabetes (LADA), prediabetic subjects, individuals at risk to develop autoimmune diabetes, siblings of type 1 diabetic patients, coeliac patients and healthy control subjects. All sera reacting with one or more of the four autoantigens by single assays also resulted positive with MAA, as well as eight of 24 type 1 diabetic patients classified initially as autoantibody-negative at disease onset based on single autoantibody assays. In addition, MAA showed 92% sensitivity and 99% specificity by analysing 140 blinded sera from type 1 diabetic patients and control subjects provided in the 2010 Diabetes Autoantibody Standardization Program. MAA is the first combined method also able to evaluate, in addition to glutamic acid decarboxylase (GAD) and tyrosine phosphatase (IA)-2, insulin and islet beta-cell zinc cation efflux transporter (ZnT8) autoantibodies. It appears to be particularly appropriate as a first-line approach for large-scale population-based screenings of anti-islet autoimmunity.


Subject(s)
Autoantibodies/blood , Diabetes Mellitus, Type 1/diagnosis , Radioimmunoassay/methods , Adult , Aged , Autoantibodies/immunology , Autoantigens/immunology , Cation Transport Proteins/immunology , Diabetes Mellitus, Type 1/immunology , Female , Glutamate Decarboxylase/immunology , High-Throughput Screening Assays/methods , Humans , Insulin/immunology , Male , Middle Aged , Protein Tyrosine Phosphatases/immunology , Sensitivity and Specificity
5.
Diabetologia ; 54(11): 2779-88, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21858504

ABSTRACT

AIMS/HYPOTHESIS: The aim of the study was to investigate the association between vitamin D intake and status and the risk of islet autoimmunity (IA) and subsequent type 1 diabetes in children at increased risk of type 1 diabetes. METHODS: The Diabetes Autoimmunity Study in the Young (DAISY) in Denver, CO, USA, has been following children at increased risk of diabetes since 1993. As of February 2011, 198 children developed IA during follow-up of 2,644 DAISY children. Vitamin D intake and plasma 25-hydroxyvitamin D [25(OH)D] were measured longitudinally. Proportional hazards regression analyses of time to IA, or type 1 diabetes in IA-positive children, were conducted, with vitamin D intake and 25(OH)D as time-varying covariates. HRs were calculated for a standard deviation difference in exposure, with adjustment for confounders. RESULTS: Intake of vitamin D was not associated with the risk of IA (adjusted HR 1.13; 95% CI 0.95, 1.35; p = 0.18) nor progression to diabetes in IA-positive children (adjusted HR 1.30; 95% CI 0.91, 1.86; p = 0.15). Moreover, 25(OH)D level was not associated with the risk of IA (adjusted HR 1.12; 95% CI 0.88, 1.43; p = 0.36), nor progression to diabetes in IA-positive children (adjusted HR 0.91; 95% CI 0.68, 1.22; p = 0.54). In the 128 children in whom we measured 25(OH)D at 9 months of age, 25(OH)D was not associated with risk of IA (n = 30 IA-positive children) (adjusted HR 1.02; 95% CI 0.96, 1.07; p = 0.58). CONCLUSIONS/INTERPRETATION: Neither vitamin D intake nor 25(OH)D levels throughout childhood were associated with the risk of IA or progression to type 1 diabetes in our population.


Subject(s)
Autoimmunity , Calcifediol/blood , Diabetes Mellitus, Type 1/etiology , Diabetes Mellitus, Type 1/immunology , Islets of Langerhans/immunology , Vitamin D Deficiency/physiopathology , Vitamin D/administration & dosage , 25-Hydroxyvitamin D 2/blood , Child , Child, Preschool , Cohort Studies , Colorado/epidemiology , Diet/adverse effects , Dietary Supplements/adverse effects , Female , Humans , Infant , Infant, Newborn , Longitudinal Studies , Male , Prospective Studies , Risk , Surveys and Questionnaires , Vitamin D/adverse effects
6.
Diabetologia ; 54(7): 1702-9, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21533899

ABSTRACT

AIMS/HYPOTHESIS: We investigated the risk associated with HLA-B*39 alleles in the context of specific HLA-DR/DQ haplotypes. METHODS: We studied a readily available dataset from the Type 1 Diabetes Genetics Consortium that consists of 2,300 affected sibling pair families genotyped for both HLA alleles and 2,837 single nucleotide polymorphisms across the major histocompatibility complex region. RESULTS: The B*3906 allele significantly enhanced the risk of type 1 diabetes when present on specific HLA-DR/DQ haplotypes (DRB1 0801-DQB1 0402: p = 1.6 × 10(-6), OR 25.4; DRB1 0101-DQB1 0501: p = 4.9 × 10(-5), OR 10.3) but did not enhance the risk of DRB1 0401-DQB1 0302 haplotypes. In addition, the B 3901 allele enhanced risk on the DRB1 1601-DQB1 0502 haplotype (p = 3.7 × 10(-3), OR 7.2). CONCLUSIONS/INTERPRETATION: These associations indicate that the B 39 alleles significantly increase risk when present on specific HLA-DR/DQ haplotypes, and HLA-B typing in concert with specific HLA-DR/DQ genotypes should facilitate genetic prediction of type 1 diabetes, particularly in a research setting.


Subject(s)
Diabetes Mellitus, Type 1/genetics , HLA-B Antigens/genetics , HLA-DQ Antigens/genetics , HLA-DR Antigens/genetics , Haplotypes/genetics , Alleles , Genetic Predisposition to Disease , Genotype , Humans
7.
Apoptosis ; 16(4): 438-48, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21274633

ABSTRACT

C/EBP homologous protein (CHOP) has been proposed as a key transcription factor for endoplasmic reticulum (ER) stress-mediated ß-cell death induced by inflammatory cytokines in vitro. However, the contribution of CHOP induction to the pathogenesis of type 1 diabetes is not yet clear. To evaluate the relevance of CHOP in the pathogenesis of type 1 diabetes in vivo, we generated CHOP-deficient non-obese diabetic (NOD.Chop (-/-)) mice. CHOP deficiency did not affect the development of insulitis and diabetes and apoptosis in ß-cells. Interestingly, NOD.Chop (-/-) mice exhibited a delayed appearance of insulin autoantibodies compared to wild-type (wt) mice. Adoptive transfer with the diabetogenic, whole or CD8(+)-depleted splenocytes induced ß-cell apoptosis and the rapid onset of diabetes in the irradiated NOD.Chop (-/-) recipients with similar kinetics as in wt mice. Expression of ER stress-associated genes was not significantly up-regulated in the islets from NOD.Chop (-/-) compared to those from wt mice or NOD-scid mice. These findings suggest that CHOP expression is independent of the development of insulitis and diabetes but might affect the early production of insulin autoantibodies in the NOD mouse.


Subject(s)
Autoantibodies/biosynthesis , Gene Deletion , Insulin/immunology , Prediabetic State/immunology , Prediabetic State/pathology , Transcription Factor CHOP/genetics , Adoptive Transfer , Animals , Apoptosis , Autoantibodies/immunology , CD8-Positive T-Lymphocytes/immunology , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/pathology , Gene Expression Regulation , In Situ Nick-End Labeling , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Lymphocyte Depletion , Mice , Mice, Inbred NOD , Peroxidase/metabolism , Spleen/immunology , Stress, Physiological/genetics , Transcription Factor CHOP/metabolism
8.
J Clin Endocrinol Metab ; 95(10): 4712-9, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20610599

ABSTRACT

CONTEXT: Zinc transporter 8 (ZnT8) is a newly discovered islet autoantigen in human type 1A diabetes (T1D). OBJECTIVE: The objective was to document changes in ZnT8 autoantibody (ZnT8A) titer and prevalence after onset of disease in relationship to 65 kDa glutamate decarboxylase antibody (GADA) and islet cell antigen antibody (IA2A). DESIGN/PATIENTS: Autoantibody radioimmunoprecipitation assays were performed on sera from three groups: 21 individuals monitored every 3 months from diagnosis for 2.5 yr; 61 individuals monitored at six monthly intervals for 5-12 yr; and a cross-sectional study of 424 patients with T1D of 20-57 yr duration. Circulating C-peptide was determined as an index of residual ß-cell function. RESULTS: ZnT8A titers declined exponentially from clinical onset of T1D with a t(1/2) ranging from 26 to 530 wk, similar to C-peptide (23-300 wk). Life-table analysis of antibody prevalence to 12 yr indicated that ZnT8A measured with either Arg325 or Trp325 probes persisted for a shorter interval than IA2A. Although prevalence of ZnT8A, IA2A, and GADA were comparable at disease onset (70.4 vs. 73.4 vs. 64%), only 6.7% of individuals remained ZnT8A positive after 25 yr compared with 19.5% for IA2A and 25.9% for GADA. Titers of ZnT8A and IA2A in seropositive individuals decreased progressively, whereas GADA remained elevated consistent with periodic reactivation of GADA humoral autoimmunity. CONCLUSIONS: ZnT8 humoral autoreactivity declines rapidly in the first years after disease onset and is less persistent than IA2A or GADA in the longer term. ZnT8A determination may be a useful measure of therapeutic efficacy in the context of immune-based clinical interventions.


Subject(s)
Autoantibodies/blood , Cation Transport Proteins/immunology , Diabetes Mellitus, Type 1/immunology , Adolescent , Adult , Age of Onset , Autoantibodies/metabolism , Biomarkers/blood , Biomarkers/metabolism , Child , Child, Preschool , Cross-Sectional Studies , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/epidemiology , Down-Regulation/immunology , Female , Humans , Kinetics , Male , Middle Aged , Seroepidemiologic Studies , Time Factors , Young Adult , Zinc Transporter 8
9.
Clin Exp Immunol ; 160(3): 331-9, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20180832

ABSTRACT

Immunotherapy can be used to induce immunological tolerance by a number of different protocols. During the last decade the ability to use tolerogenic dendritic cells (DCs) to prevent autoimmunity has received much attention. Many studies have attempted to use immature or semi-mature DCs to induce tolerance in the non-obese diabetic (NOD) mouse model of human type 1 diabetes. However, most studies to date have used protocols in which generation of DCs involved a culture step in fetal bovine serum (FBS)-supplemented medium which may affect tolerance induction in a non-specific fashion. Indeed, several studies have shown that DCs cultured in the presence of FBS will induce a powerful T helper type 2 (Th2) immune response towards FBS-related antigens which can suppress an ongoing immune response. Hence, this may interfere with diabetes development in the NOD mouse by induction of immune deviation rather than by antigen-specific tolerance. In order to test whether antigen-specific tolerance induction by DC therapy is feasible in the NOD mouse, we therefore generated immature DCs using autologous serum [normal mouse serum (NMS)-supplemented cultures] instead of FBS, and we show that these DCs can protect NOD mice from diabetes, if pulsed with insulin-peptide antigens before adoptive transfer. Our data therefore support that DC therapy is able to prevent diabetes in the NOD mouse in an antigen-specific manner.


Subject(s)
Antigens/immunology , Dendritic Cells , Diabetes Mellitus, Type 1/therapy , Immune Tolerance , Immunotherapy, Adoptive , Th2 Cells/immunology , Animals , Cattle , Dendritic Cells/immunology , Dendritic Cells/transplantation , Diabetes Mellitus, Type 1/immunology , Female , Mice , Mice, Inbred NOD , Transplantation, Autologous
10.
Diabetologia ; 53(4): 690-8, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20062967

ABSTRACT

AIMS/HYPOTHESIS: Childhood diabetes is thought to usually result from autoimmune beta cell destruction (type 1A) with eventual total loss of beta cells. Analysis of C-peptide in children characterised at diabetes onset for autoantibodies shows heterogeneous preservation of insulin secretion in long-standing diabetes. The aim of this study was to characterise the pancreases of childhood-onset diabetes in order to define the pathological basis of this heterogeneity. METHODS: We evaluated 20 cadaveric organ donor pancreases of childhood-onset long-term patients for disease heterogeneity and obtained corresponding C-peptide measurements. RESULTS: Pancreases from the majority of cadaveric donors contained only insulin-deficient islets (14 of 20). The remaining six patients (30%) had numerous insulin-positive cells within at least some islets, with two different histological patterns. Pattern A (which we would associate with type 1A diabetes) had lobular retention of areas with 'abnormal' beta cells producing the apoptosis inhibitor survivin and HLA class I. In pattern B, 100% of all islets contained normal-appearing but quantitatively reduced beta cells without survivin or HLA class I. CONCLUSIONS/INTERPRETATION: Our data demonstrate that C-peptide secretion in long-standing diabetic patients can be explained by two different patterns of beta cell survival,possibly reflecting different subsets of type 1 diabetes.


Subject(s)
Diabetes Mellitus, Type 1/pathology , Insulin-Secreting Cells/pathology , Pancreas/pathology , Sex Characteristics , Adolescent , Adult , Age of Onset , Autoantibodies/blood , C-Peptide/blood , Child , Child, Preschool , Diabetes Mellitus, Type 1/immunology , Female , HLA-DR Antigens , Histocompatibility Testing , Humans , Hyperinsulinism/pathology , Male , Middle Aged , Tissue Donors
11.
Genes Immun ; 10 Suppl 1: S21-6, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19956096

ABSTRACT

Protein tyrosine phosphatase non-receptor type 22 (PTPN22) is the third major locus affecting risk of type I diabetes (T1D), after HLA-DR/DQ and INS. The most associated single-nucleotide polymorphism (SNP), rs2476601, has a C->T variant and results in an arginine (R) to tryptophan (W) amino acid change at position 620. To assess whether this, or other specific variants, are responsible for T1D risk, the Type I Diabetes Genetics Consortium analyzed 28 PTPN22 SNPs in 2295 affected sib-pair (ASP) families. Transmission Disequilibrium Test analyses of haplotypes revealed that all three haplotypes with a T allele at rs2476601 were overtransmitted to affected children, and two of these three haplotypes showed statistically significant overtransmission (P=0.003 to P=5.9E-12). Another haplotype had decreased transmission to affected children (P=3.5E-05). All haplotypes containing the rs2476601 T allele were identical for all SNPs across PTPN22 and only varied at centromeric SNPs. When considering rs2476601 'C' founder chromosomes, a second haplotype (AGGGGC) centromeric of PTPN22 in the C1orf178 region was associated with protection from T1D (odds ratio=0.81, P=0.0005). This novel finding requires replication in independent populations. We conclude the major association of PTPN22 with T1D is likely due to the recognized non-synonymous SNP rs2476601 (R620W).


Subject(s)
Diabetes Mellitus, Type 1/genetics , Genetic Predisposition to Disease , Haplotypes , Polymorphism, Single Nucleotide , Protein Tyrosine Phosphatase, Non-Receptor Type 22/genetics , Adolescent , Adult , Aged , Child , Child, Preschool , Diabetes Mellitus, Type 1/epidemiology , Female , Humans , Infant , Infant, Newborn , Male , Middle Aged , Risk Factors , Young Adult
12.
Scand J Immunol ; 70(5): 439-46, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19874548

ABSTRACT

Insulin is a critical autoantigen for the development of autoimmune diabetes in non-obese diabetic (NOD) mice. About 80% of NOD females and 30-40% of NOD males develop diabetes. However, Insulin2 (Ins2) knockout NOD mice develop autoimmune diabetes with complete penetrance in both sexes, at an earlier age, and have stronger autoimmune responses to insulin. The severe diabetes phenotype observed in NOD-Ins2-/- mice suggests that lack of Ins2 expression in the thymus may compromise immunological tolerance to insulin. Insulin is a prototypical tissue specific antigen (TSA) for which tolerance is dependent on expression in thymus and peripheral lymphoid tissues. TSA are naturally expressed by medullary thymic epithelial cells (mTEC), stromal cells in peripheral lymphoid tissues and bone marrow (BM)-derived cells, mainly CD11c(+) dendritic cells. The natural expression of TSA by mTEC and stromal cells has been shown to contribute to self-tolerance. However, it is unclear whether this also applies to BM-derived cells naturally expressing TSA. To address this question, we created BM chimeras and investigated whether reintroducing Ins2 expression solely by NOD BM-derived cells delays diabetes development in NOD-Ins2-/- mice. On follow-up, NOD-Ins2-/- mice receiving Ins2-expressing NOD BM cells developed diabetes at similar rates of those receiving NOD-Ins2-/- BM cells. Diabetes developed in 64% of NOD recipients transplanted with NOD BM and in 47% of NOD mice transplanted with NOD-Ins2-/- BM (P = ns). Thus, NOD-Ins2-/- BM did not worsen diabetes in NOD recipients and Ins2 expression by NOD BM-derived cells did not delay diabetes development in NOD-Ins2-/- mice.


Subject(s)
Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Immune Tolerance , Insulin/genetics , Insulin/immunology , Animals , Bone Marrow Cells/immunology , Female , Male , Mice , Mice, Inbred NOD , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic
13.
Clin Genet ; 76(5): 441-8, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19807739

ABSTRACT

Autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED) is a rare recessive disorder resulting from mutations in the autoimmune regulator (AIRE) gene. There is no information on AIRE mutations in Indians. In a cross-sectional study, nine patients (eight families), from four referral hospitals in India, were studied for AIRE mutations by direct sequencing. We screened for new mutations in 150 controls by allele-specific PCR. The patients had 1-7 known components of APECED. Three patients had unusual manifestations: presentation with type 1 diabetes; chronic sinusitis and otitis media; and facial dysmorphism. All patients carried homozygous, probably recessive, AIRE mutations. Two unrelated patients from a small in-bred community (Vanika Vaisya) in south India carried an unreported missense mutation, p.V80G, in the N-terminal caspase recruitment domain. Another unique mutation, p.C302X, resulting in a truncated protein with deletion of both zinc-finger domains, was detected in a patient from Gujarat. Neither mutation was detected in controls. Other mutations, previously described in Caucasians, were: 13 base pair deletion (p.C322fsX372) in 4 (38%), and Finn-major (p.R257X) and p.R139X (Sardinian) mutation in one subject each. In conclusion, in this first series of APECED in Indians, we detected AIRE mutations previously reported in Caucasians, as well as unique mutations. Of these, p.V80G is possibly an ancestral mutation in an in-bred community.


Subject(s)
Asian People/genetics , Mutation , Polyendocrinopathies, Autoimmune/ethnology , Polyendocrinopathies, Autoimmune/genetics , Transcription Factors/genetics , Adult , Base Sequence , Child , Genetic Testing , Humans , India , Male , Molecular Sequence Data , Phenotype , Polyendocrinopathies, Autoimmune/pathology , AIRE Protein
14.
J Intern Med ; 265(5): 530-40, 2009 May.
Article in English | MEDLINE | ID: mdl-19382992

ABSTRACT

Autoimmune polyendocrine syndromes type 1 and 2 (APS-1 and APS-2) are diverse in regards to their component diseases and immunologic features of pathogenesis. Animal models and human studies highlight the importance of alleles of HLA (human leukocyte antigen)-like molecules determining tissue specific targeting that with the loss of tolerance leads to organ specific autoimmunity. Knowledge of the syndromes and component diseases allows clinicians to recognize and prevent illness prior to morbidity. With the current understanding of the syndromes, a paradigm for diagnosis, screening and treatment can be established. Once genetically susceptible individuals are identified screening for autoantibodies can be performed. Amongst autoantibody positive individuals, monitoring for physiologic decompensation, with a goal of treating prior to morbidity and in some cases mortality, follows. With continued basic and clinical research, therapies aimed at treating the underlying autoimmunity and disease prevention should become possible.


Subject(s)
Models, Immunological , Polyendocrinopathies, Autoimmune/immunology , Adrenal Insufficiency/immunology , Animals , Autoantibodies/blood , Autoimmunity/genetics , Genetic Predisposition to Disease , Histocompatibility Antigens Class II/genetics , Humans , Mutation , Polyendocrinopathies, Autoimmune/blood , Polyendocrinopathies, Autoimmune/diagnosis , Self Tolerance/genetics , Syndrome , Transcription Factors/genetics , AIRE Protein
15.
Diabetes Obes Metab ; 11 Suppl 1: 25-30, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19143812

ABSTRACT

AIM: The goal of this study was to develop and implement methodology that would aid in the analysis of extended high-density single nucleotide polymorphism (SNP) major histocompatibility complex (MHC) haplotypes combined with human leucocyte antigen (HLA) alleles in relation to type 1 diabetes risk. METHODS: High-density SNP genotype data (2918 SNPs) across the MHC from the Type 1 Diabetes Genetics Consortium (1240 families), in addition to HLA data, were processed into haplotypes using PedCheck and Merlin, and extended DR3 haplotypes were analysed. RESULTS: With this large dense set of SNPs, the conservation of DR3-B8-A1 (8.1) haplotypes spanned the MHC (>/=99% SNP identity). Forty-seven individuals homozygous for the 8.1 haplotype also shared the same homozygous genotype at four 'sentinel' SNPs (rs2157678 'T', rs3130380 'A', rs3094628 'C' and rs3130352 'T'). Conservation extended from HLA-DQB1 to the telomeric end of the SNP panels (3.4 Mb total). In addition, we found that the 8.1 haplotype is associated with lower risk than other DR3 haplotypes by both haplotypic and genotypic analyses [haplotype: p = 0.009, odds ratio (OR) = 0.65; genotype: p = 6.3 x 10(-5), OR = 0.27]. The 8.1 haplotype (from genotypic analyses) is associated with lower risk than the high-risk DR3-B18-A30 haplotype (p = 0.01, OR = 0.23), but the DR3-B18-A30 haplotype did not differ from other non-8.1 DR3 haplotypes relative to diabetes association. CONCLUSION: The 8.1 haplotype demonstrates extreme conservation (>3.4 Mb) and is associated with significantly lower risk for type 1 diabetes than other DR3 haplotypes.


Subject(s)
Diabetes Mellitus, Type 1/genetics , HLA-DR3 Antigen/genetics , Polymorphism, Single Nucleotide/genetics , Conserved Sequence , Genetic Predisposition to Disease/genetics , Genotype , Haplotypes , Heterozygote , Homozygote , Humans , Pedigree , Risk Factors
16.
Ann N Y Acad Sci ; 1150: 245-7, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19120304

ABSTRACT

IA2 and phogrin are important targets of humoral and cell-mediated autoimmunity in type 1 diabetes in man. They belong to a conserved subfamily of transmembrane protein tyrosine phosphatases (PTPs) associated with the regulatory pathway of secretion. To examine potential cross-reactivity between PTP family members we tested sera from T1D patients for reactivity to IA2, and the Drosophila (FLYDA) and C. elegans (IDA) orthologs using radioimmunoprecipitation assays of (35)S Met-labeled in vitro translated products of the cytosolic domains of these proteins. Approximately 80% of sera reacted with at least one probe. Of these, 82.5% showed reactivity to human IA2, 74.1% to FLYDA, and 33.7% to IDA. The majority of sera that bound FLYDA and/or IDA also recognized IA2. This raises the possibility that in some cases reactivity to IA2 may have arisen by molecular mimicry.


Subject(s)
Autoantigens/immunology , Conserved Sequence/immunology , Diabetes Mellitus, Type 1/immunology , Epitopes/immunology , Protein Tyrosine Phosphatases/immunology , Animals , Antibody Specificity , Autoantigens/chemistry , Autoantigens/genetics , Caenorhabditis elegans/enzymology , Caenorhabditis elegans/genetics , Case-Control Studies , Conserved Sequence/genetics , Cross Reactions/immunology , Drosophila/enzymology , Drosophila/genetics , Epitope Mapping , Epitopes/chemistry , Epitopes/genetics , Female , Humans , Male , Multigene Family/immunology , Multigene Family/physiology , Protein Tyrosine Phosphatases/chemistry , Protein Tyrosine Phosphatases/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 8/chemistry , Receptor-Like Protein Tyrosine Phosphatases, Class 8/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 8/immunology , Sequence Homology
17.
Genes Immun ; 8(8): 628-33, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17728790

ABSTRACT

Autoimmune diabetes shows extreme variation in age of onset and clinical presentation, although most studies have been done in children with the most severe subtype. Disease risk is strongly associated with HLA-DRB1*0301-DQA1*0501-DQB1*0201 (DR3-DQ2), but it has not been possible to separate the effects of the DR and DQ alleles. We have identified a large Bedouin kindred in which a high prevalence of islet autoimmunity is associated with two different DR3 haplotypes, one carrying the usual DQ2 and the other carrying DQA1*0102-DQB1*0502 (DQ5). Results of prospective follow-up studies indicate that DR3 is associated with the initial activation of islet autoimmunity whereas DQ2 is associated with early-onset and severe clinical disease. The association signals map to a 350-kb interval, thus implicating primary effects for DR3 and DQ2. Overall, our results emphasize the importance of prospective genetic studies that examine the full range of variation in the initiation, progression and expression of autoimmune disease.


Subject(s)
Autoimmunity/genetics , Diabetes Mellitus, Type 1/genetics , Genetic Predisposition to Disease , HLA-DQ Antigens/genetics , HLA-DR Antigens/genetics , Islets of Langerhans/immunology , Adolescent , Adult , Age of Onset , Aged , Arabs/genetics , Child , Child, Preschool , Female , Gene Frequency , HLA-DQ alpha-Chains , HLA-DQ beta-Chains , HLA-DRB1 Chains , Haplotypes , Humans , Infant , Infant, Newborn , Male , Middle Aged
18.
Cell Mol Life Sci ; 64(7-8): 865-72, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17429581

ABSTRACT

Type 1A diabetes is an organ-specific autoimmune disease resulting from destruction of insulin-producing pancreatic beta-cells. The main susceptibility genes code for polymorphic HLA molecules and in particular alleles of class II MHC genes (DR, DQ and DP). Polymorphisms of individual genes outside the MHC also contribute to diabetes risk but recent evidence suggests that there are additional non-HLA genes determining susceptibility linked to the MHC. It is now possible using genetic and autoantibody assays to predict the development of type 1A diabetes in the majority of individuals, and trials of diabetes prevention are underway.


Subject(s)
Diabetes Mellitus, Type 1/genetics , Genetic Predisposition to Disease , Animals , Autoimmune Diseases , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/prevention & control , Genes, MHC Class II , HLA Antigens/genetics , Humans , Insulin-Secreting Cells/immunology , Polymorphism, Genetic , Risk Factors
19.
Ann N Y Acad Sci ; 1079: 122-9, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17130542

ABSTRACT

We analyzed double native insulin gene knockout NOD mice with a mutated (B16:alanine) proinsulin transgene at multiple ages for the development of insulin autoantibodies, insulitis, and diabetes. In contrast to mice with at least one copy of a native insulin gene that expressed insulin antibodies, only 2 out of 21 (10%) double native insulin gene knockout mice with a mutated insulin transgene developed insulin autoantibodies. Of 21 double insulin knockout mice sacrificed between 10 to 48 weeks of age, only 5 showed minimal insulitis versus 100% of wild-type NOD and more than 90% of insulin 1 knockout mice. Consistent with robust suppression of insulin autoantibodies and insulitis, no double insulin knockout mice developed diabetes. In that the B9-23 peptide with B16A is an altered peptide ligand inducing Th2 responses, we analyzed transfer of splenocytes into NOD.SCID mice. There was no evidence for regulatory T cells able to inhibit transfer of diabetes by diabetogenic NOD splenocytes. Insulin peptide B9-23 is likely a crucial target for initiation of islet autoimmunity and further mutation of the sequence will be tested to attempt to eliminate all anti-islet autoimmunity.


Subject(s)
Autoantibodies/analysis , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/prevention & control , Insulin Antibodies/analysis , Proinsulin/genetics , Alanine/metabolism , Amino Acid Substitution , Animals , Cell Transplantation , Diabetes Mellitus, Type 1/etiology , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/metabolism , Insulin Antibodies/immunology , Islets of Langerhans/pathology , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Proinsulin/chemistry , Proinsulin/immunology , Spleen/cytology , Spleen/immunology , Transgenes
20.
Clin Immunol ; 121(1): 40-6, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16798097

ABSTRACT

The aim of the present study was to evaluate the epitope specific humoral human tissue transglutaminase (tTG) immunoreactivity against 3 different human recombinant tTG constructs [(full-length tTG (a.a. 1-687), tTG (a.a. 227-687); tTG (a.a. 473-687)] before and after the introduction of a gluten-free diet (GFD). To this end, sera from 64 celiac disease (CD) subjects on a gluten-containing diet (44 f, 20 m) and after 0.6 +/- 0.3 years and 2.1 +/- 1.3 years of GFD were studied using a quantitative radioimmunoprecipitation assay. All 64 CD patients at diagnosis were full-length anti-tTG (a.a. 1-687)Ab positive. These Abs significantly decreased in frequency and titer after 6 months and 2 years of GFD. However, at low titers, 64.1% (41/64) of CD patients were still fl-tTG (a.a. 1-687)Ab positive after 2 years of GFD. At disease diagnosis, 70.3% (45/64) of the CD patients had Abs directed against fragments (227-687) and/or (473-687) of the tTG protein. This percentage, after 2 years of GFD, significantly decreased to 18.7%, whereas almost 50% of GFD patients had no tTG (227-687) and tTG (473-687) fragment reactivity, but only persistent, low-titer full-length tTG (1-687)Abs. We suggest that the selective loss of immunoreactivity against tTG (227-687) and tTG (473-687) fragments in CD patients with a GFD, could be due to quantitative decrease of autoreactivity driven by tTG-gliadin interaction underlying celiac disease pathogenesis.


Subject(s)
Autoantigens/immunology , Celiac Disease/enzymology , Celiac Disease/immunology , Epitopes/immunology , Glutens , Transglutaminases/immunology , Adolescent , Adult , Antigen-Antibody Reactions , Autoantibodies/blood , Autoantigens/blood , Celiac Disease/blood , Celiac Disease/diet therapy , Child , Child, Preschool , Epitopes/blood , Female , Glutens/blood , Humans , Infant , Male , Middle Aged , Muscle Fibers, Skeletal/immunology , Peptide Fragments/immunology , Peptide Fragments/metabolism , Transglutaminases/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...