Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 110
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Diabetologia ; 67(6): 995-1008, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38517484

ABSTRACT

AIMS/HYPOTHESIS: Type 1 diabetes is an heterogenous condition. Characterising factors explaining differences in an individual's clinical course and treatment response will have important clinical and research implications. Our aim was to explore type 1 diabetes heterogeneity, as assessed by clinical characteristics, autoantibodies, beta cell function and glycaemic outcomes, during the first 12 months from diagnosis, and how it relates to age at diagnosis. METHODS: Data were collected from the large INNODIA cohort of individuals (aged 1.0-45.0 years) newly diagnosed with type 1 diabetes, followed 3 monthly, to assess clinical characteristics, C-peptide, HbA1c and diabetes-associated antibodies, and their changes, during the first 12 months from diagnosis, across three age groups: <10 years; 10-17 years; and ≥18 years. RESULTS: The study population included 649 individuals (57.3% male; age 12.1±8.3 years), 96.9% of whom were positive for one or more diabetes-related antibodies. Baseline (IQR) fasting C-peptide was 242.0 (139.0-382.0) pmol/l (AUC 749.3 [466.2-1106.1] pmol/l × min), with levels increasing with age (p<0.001). Over time, C-peptide remained lower in participants aged <10 years but it declined in all age groups. In parallel, glucose levels progressively increased. Lower baseline fasting C-peptide, BMI SD score and presence of diabetic ketoacidosis at diagnosis were associated with lower stimulated C-peptide over time. HbA1c decreased during the first 3 months (p<0.001), whereas insulin requirement increased from 3 months post diagnosis (p<0.001). CONCLUSIONS/INTERPRETATION: In this large cohort with newly diagnosed type 1 diabetes, we identified age-related differences in clinical and biochemical variables. Of note, C-peptide was lower in younger children but there were no main age differences in its rate of decline.


Subject(s)
Autoantibodies , C-Peptide , Diabetes Mellitus, Type 1 , Glycated Hemoglobin , Humans , Diabetes Mellitus, Type 1/diagnosis , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/epidemiology , Adolescent , Child , Male , Female , C-Peptide/blood , Adult , Young Adult , Child, Preschool , Autoantibodies/blood , Glycated Hemoglobin/metabolism , Blood Glucose/metabolism , Cohort Studies , Infant , Europe/epidemiology , Middle Aged , Insulin-Secreting Cells/metabolism
2.
PLoS Genet ; 15(6): e1008178, 2019 06.
Article in English | MEDLINE | ID: mdl-31199784

ABSTRACT

Type 1 diabetes (T1D) is a chronic multi-factorial disorder characterized by the immune-mediated destruction of insulin-producing pancreatic beta cells. Variations at a large number of genes influence susceptibility to spontaneous autoimmune T1D in non-obese diabetic (NOD) mice, one of the most frequently studied animal models for human disease. The genetic analysis of these mice allowed the identification of many insulin-dependent diabetes (Idd) loci and candidate genes, one of them being Cd101. CD101 is a heavily glycosylated transmembrane molecule which exhibits negative-costimulatory functions and promotes regulatory T (Treg) function. It is abundantly expressed on subsets of lymphoid and myeloid cells, particularly within the gastrointestinal tract. We have recently reported that the genotype-dependent expression of CD101 correlates with a decreased susceptibility to T1D in NOD.B6 Idd10 congenic mice compared to parental NOD controls. Here we show that the knockout of CD101 within the introgressed B6-derived Idd10 region increased T1D frequency to that of the NOD strain. This loss of protection from T1D was paralleled by decreased Gr1-expressing myeloid cells and FoxP3+ Tregs and an enhanced accumulation of CD4-positive over CD8-positive T lymphocytes in pancreatic tissues. As compared to CD101+/+ NOD.B6 Idd10 donors, adoptive T cell transfers from CD101-/- NOD.B6 Idd10 mice increased T1D frequency in lymphopenic NOD scid and NOD.B6 Idd10 scid recipients. Increased T1D frequency correlated with a more rapid expansion of the transferred CD101-/- T cells and a lower proportion of recipient Gr1-expressing myeloid cells in the pancreatic lymph nodes. Fewer of the Gr1+ cells in the recipients receiving CD101-/- T cells expressed CD101 and the cells had lower levels of IL-10 and TGF-ß mRNA. Thus, our results connect the Cd101 haplotype-dependent protection from T1D to an anti-diabetogenic function of CD101-expressing Tregs and Gr1-positive myeloid cells and confirm the identity of Cd101 as Idd10.


Subject(s)
Antigens, CD/genetics , Antigens, Ly/genetics , Diabetes Mellitus, Type 1/genetics , Pancreas/metabolism , Animals , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Diabetes Mellitus, Type 1/pathology , Gene Expression Regulation/genetics , Genetic Predisposition to Disease , Haplotypes/genetics , Humans , Lymph Nodes/metabolism , Lymph Nodes/pathology , Mice , Mice, Inbred NOD , Mice, Knockout , Myeloid Cells/immunology , Myeloid Cells/metabolism , Pancreas/pathology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
3.
J Immunol ; 200(1): 147-162, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29158418

ABSTRACT

We previously reported that NOD.c3c4 mice develop spontaneous autoimmune biliary disease (ABD) with anti-mitochondrial Abs, histopathological lesions, and autoimmune T lymphocytes similar to human primary biliary cholangitis. In this article, we demonstrate that ABD in NOD.c3c4 and related NOD ABD strains is caused by a chromosome 1 region that includes a novel mutation in polycystic kidney and hepatic disease 1 (Pkhd1). We show that a long terminal repeat element inserted into intron 35 exposes an alternative polyadenylation site, resulting in a truncated Pkhd1 transcript. A novel NOD congenic mouse expressing aberrant Pkhd1, but lacking the c3 and c4 chromosomal regions (NOD.Abd3), reproduces the immunopathological features of NOD ABD. RNA sequencing of NOD.Abd3 common bile duct early in disease demonstrates upregulation of genes involved in cholangiocyte injury/morphology and downregulation of immunoregulatory genes. Consistent with this, bone marrow chimera studies show that aberrant Pkhd1 must be expressed in the target tissue (cholangiocytes) and the immune system (bone marrow). Mutations of Pkhd1 produce biliary abnormalities in mice but have not been previously associated with autoimmunity. In this study, we eliminate clinical biliary disease by backcrossing this Pkhd1 mutation onto the C57BL/6 genetic background; thus, the NOD genetic background (which promotes autoimmunity) is essential for disease. We propose that loss of functional Pkhd1 on the NOD background produces early bile duct abnormalities, initiating a break in tolerance that leads to autoimmune cholangitis in NOD.Abd3 congenic mice. This model is important for understanding loss of tolerance to cholangiocytes and is relevant to the pathogenesis of several human cholangiopathies.


Subject(s)
Autoimmune Diseases/genetics , Cholangitis/genetics , Diabetes Mellitus/genetics , Liver Cirrhosis, Biliary/genetics , Mutation/genetics , Receptors, Cell Surface/genetics , Animals , Chimera , Disease Models, Animal , Genetic Background , Humans , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Terminal Repeat Sequences/genetics
4.
J Autoimmun ; 95: 1-14, 2018 12.
Article in English | MEDLINE | ID: mdl-30446251

ABSTRACT

Susceptibility to multiple autoimmune diseases is associated with common gene polymorphisms influencing IL-2 signaling and Treg function, making Treg-specific expansion by IL-2 a compelling therapeutic approach to treatment. As an in vivo IL-2 half-life enhancer we used a non-targeted, effector-function-silent human IgG1 as a fusion protein. An IL-2 mutein (N88D) with reduced binding to the intermediate affinity IL-2Rßγ receptor was engineered with a stoichiometry of two IL-2N88D molecules per IgG, i.e. IgG-(IL-2N88D)2. The reduced affinity of IgG-(IL-2N88D)2 for the IL-2Rßγ receptor resulted in a Treg-selective molecule in human whole blood pSTAT5 assays. Treatment of cynomolgus monkeys with single low doses of IgG-(IL-2N88D)2 induced sustained preferential activation of Tregs accompanied by a corresponding 10-14-fold increase in CD4+ and CD8+ CD25+FOXP3+ Tregs; conditions that had no effect on CD4+ or CD8+ memory effector T cells. The expanded cynomolgus Tregs had demethylated FOXP3 and CTLA4 epigenetic signatures characteristic of functionally suppressive cells. Humanized mice had similar selective in vivo responses; IgG-(IL-2N88D)2 increased Tregs while wild-type IgG-IL-2 increased NK cells in addition to Tregs. The expanded human Tregs had demethylated FOXP3 and CTLA4 signatures and were immunosuppressive. These results describe a next-generation immunotherapy using a long-lived and Treg-selective IL-2 that activates and expands functional Tregsin vivo. Patients should benefit from restored immune homeostasis in a personalized fashion to the extent that their autoimmune disease condition dictates opening up the possibility for remissions and cures.


Subject(s)
Autoimmune Diseases/therapy , Immunoglobulin G/immunology , Immunotherapy/methods , Interleukin-2/immunology , Lymphotoxin-alpha/immunology , Recombinant Fusion Proteins/immunology , T-Lymphocytes, Regulatory/drug effects , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Binding Sites , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , CTLA-4 Antigen/genetics , CTLA-4 Antigen/immunology , Cell Proliferation , DNA Methylation/drug effects , Disease Models, Animal , Epigenesis, Genetic , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Humans , Immunoglobulin G/administration & dosage , Immunoglobulin G/chemistry , Immunoglobulin G/genetics , Interleukin-2/administration & dosage , Interleukin-2/chemistry , Interleukin-2/genetics , Interleukin-2 Receptor beta Subunit/genetics , Interleukin-2 Receptor beta Subunit/immunology , Lymphocyte Activation/drug effects , Lymphotoxin-alpha/administration & dosage , Lymphotoxin-alpha/chemistry , Lymphotoxin-alpha/genetics , Macaca fascicularis , Male , Mice , Mice, Transgenic , Models, Molecular , Protein Binding , Protein Isoforms/genetics , Protein Isoforms/immunology , Protein Structure, Secondary , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/immunology , Signal Transduction , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology
5.
PLoS Genet ; 11(6): e1005272, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26106896

ABSTRACT

Identification of candidate causal variants in regions associated with risk of common diseases is complicated by linkage disequilibrium (LD) and multiple association signals. Nonetheless, accurate maps of these variants are needed, both to fully exploit detailed cell specific chromatin annotation data to highlight disease causal mechanisms and cells, and for design of the functional studies that will ultimately be required to confirm causal mechanisms. We adapted a Bayesian evolutionary stochastic search algorithm to the fine mapping problem, and demonstrated its improved performance over conventional stepwise and regularised regression through simulation studies. We then applied it to fine map the established multiple sclerosis (MS) and type 1 diabetes (T1D) associations in the IL-2RA (CD25) gene region. For T1D, both stepwise and stochastic search approaches identified four T1D association signals, with the major effect tagged by the single nucleotide polymorphism, rs12722496. In contrast, for MS, the stochastic search found two distinct competing models: a single candidate causal variant, tagged by rs2104286 and reported previously using stepwise analysis; and a more complex model with two association signals, one of which was tagged by the major T1D associated rs12722496 and the other by rs56382813. There is low to moderate LD between rs2104286 and both rs12722496 and rs56382813 (r2 ≃ 0:3) and our two SNP model could not be recovered through a forward stepwise search after conditioning on rs2104286. Both signals in the two variant model for MS affect CD25 expression on distinct subpopulations of CD4+ T cells, which are key cells in the autoimmune process. The results support a shared causal variant for T1D and MS. Our study illustrates the benefit of using a purposely designed model search strategy for fine mapping and the advantage of combining disease and protein expression data.


Subject(s)
Bayes Theorem , Chromosome Mapping/methods , Diabetes Mellitus, Type 1/genetics , Genetic Predisposition to Disease , Multiple Sclerosis/genetics , Algorithms , Chromosome Mapping/statistics & numerical data , Haplotypes , Humans , Interleukin-2 Receptor alpha Subunit/genetics , Linkage Disequilibrium , Polymorphism, Single Nucleotide , Stochastic Processes
6.
Clin Immunol ; 179: 25-39, 2017 06.
Article in English | MEDLINE | ID: mdl-28284938

ABSTRACT

To date many clinical studies aim to increase the number and/or fitness of CD4+CD127lowCD25+ regulatory T cells (Tregs) in vivo to harness their regulatory potential in the context of treating autoimmune disease. Here, we sought to define the phenotype and function of Tregs expressing the highest levels of IL-6 receptor (IL-6R). We have identified a population of CD4+CD127lowCD25+ TIGIT- T cells distinguished by their elevated IL-6R expression that lacked expression of HELIOS, showed higher CTLA-4 expression, and displayed increased suppressive capacity compared to IL-6RhiTIGIT+ Tregs. IL-6RhiTIGIT- CD127lowCD25+ T cells contained a majority of cells demethylated at FOXP3 and displayed a Th17 transcriptional signature, including RORC (RORγt) and the capacity of producing both pro- and anti-inflammatory cytokines, such as IL-17, IL-22 and IL-10. We propose that in vivo, in the presence of IL-6-associated inflammation, the suppressive function of CD4+CD127lowCD25+ FOXP3+IL-6RhiTIGIT- T cells is temporarily disarmed allowing further activation of the effector functions and potential pathogenic tissue damage.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , T-Lymphocyte Subsets/immunology , Adolescent , Adult , Cytokines/immunology , Diabetes Mellitus, Type 1/immunology , Humans , Male , Middle Aged , Phenotype , Receptors, Immunologic/immunology , STAT3 Transcription Factor/immunology , Young Adult
7.
J Autoimmun ; 84: 75-86, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28747257

ABSTRACT

Identification of alterations in the cellular composition of the human immune system is key to understanding the autoimmune process. Recently, a subset of FOXP3+ cells with low CD25 expression was found to be increased in peripheral blood from systemic lupus erythematosus (SLE) patients, although its functional significance remains controversial. Here we find in comparisons with healthy donors that the frequency of FOXP3+ cells within CD127lowCD25low CD4+ T cells (here defined as CD25lowFOXP3+ T cells) is increased in patients affected by autoimmune disease of varying severity, from combined immunodeficiency with active autoimmunity, SLE to type 1 diabetes. We show that CD25lowFOXP3+ T cells share phenotypic features resembling conventional CD127lowCD25highFOXP3+ Tregs, including demethylation of the Treg-specific epigenetic control region in FOXP3, HELIOS expression, and lack of IL-2 production. As compared to conventional Tregs, more CD25lowFOXP3+HELIOS+ T cells are in cell cycle (33.0% vs 20.7% Ki-67+; P = 1.3 × 10-9) and express the late-stage inhibitory receptor PD-1 (67.2% vs 35.5%; P = 4.0 × 10-18), while having reduced expression of the early-stage inhibitory receptor CTLA-4, as well as other Treg markers, such as FOXP3 and CD15s. The number of CD25lowFOXP3+ T cells is correlated (P = 3.1 × 10-7) with the proportion of CD25highFOXP3+ T cells in cell cycle (Ki-67+). These findings suggest that CD25lowFOXP3+ T cells represent a subset of Tregs that are derived from CD25highFOXP3+ T cells, and are a peripheral marker of recent Treg expansion in response to an autoimmune reaction in tissues.


Subject(s)
Forkhead Transcription Factors/metabolism , Ikaros Transcription Factor/metabolism , Lupus Erythematosus, Systemic/immunology , T-Lymphocyte Subsets/physiology , T-Lymphocytes, Regulatory/physiology , Adolescent , Adult , Aged , Autoimmunity , Cells, Cultured , Child , Demethylation , Epigenetic Repression , Female , Forkhead Transcription Factors/genetics , High-Throughput Nucleotide Sequencing , Humans , Ikaros Transcription Factor/genetics , Interleukin-2 Receptor alpha Subunit/metabolism , Male , Middle Aged , Young Adult
8.
J Immunol ; 194(6): 2654-63, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25672752

ABSTRACT

Type 1 diabetes (T1D) is a polygenic disease with multiple insulin-dependent diabetes (Idd) loci predisposing humans and NOD mice to disease. NOD.B10 Idd9 congenic mice, in which the NOD Idd9 chromosomal region is replaced by the Idd9 from T1D-resistant C57BL/10 mice, are significantly protected from T1D development. However, the genes and pathways conferring T1D development or protection by Idd9 remain to be fully elucidated. We have developed novel NOD.B10-Idd9 (line 905) congenic mice that predominantly harbor islet-reactive CD4(+) T cells expressing the BDC2.5 TCR (BDC-Idd9.905 mice). To establish functional links between the Idd9 genotype and its phenotype, we used microarray analyses to investigate the gene expression profiles of ex vivo and Ag-activated CD4(+) T cells from these mice and BDC2.5 (BDC) NOD controls. Among the differentially expressed genes, those located within the Idd9 region were greatly enriched in islet-specific CD4(+) T cells. Bioinformatics analyses of differentially expressed genes between BDC-Idd9.905 and BDC CD4(+) T cells identified Eno1, Rbbp4, and Mtor, all of which are encoded by Idd9 and part of gene networks involved in cellular growth and development. As predicted, proliferation and Th1/Th17 responses of islet-specific CD4(+) T cells from BDC-Idd9.905 mice following Ag stimulation in vitro were reduced compared with BDC mice. Furthermore, proliferative responses to endogenous autoantigen and diabetogenic function were impaired in BDC-Idd9.905 CD4(+) T cells. These findings suggest that differential expression of the identified Idd9 genes contributed to Idd9-dependent T1D susceptibility by controlling the diabetogenic function of islet-specific CD4(+) T cells.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Diabetes Mellitus, Type 1/genetics , Gene Expression Profiling/methods , Genetic Predisposition to Disease/genetics , Genome-Wide Association Study/methods , Islets of Langerhans/metabolism , Phosphopyruvate Hydratase/genetics , Retinoblastoma-Binding Protein 4/genetics , Animals , CD4-Positive T-Lymphocytes/immunology , Cell Proliferation/genetics , Chromosome Mapping , Chromosomes, Mammalian/genetics , Cluster Analysis , Diabetes Mellitus, Type 1/immunology , Gene Regulatory Networks , Mice , Mice, Congenic , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
9.
J Immunol ; 195(10): 4841-52, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26438525

ABSTRACT

By congenic strain mapping using autoimmune NOD.C57BL/6J congenic mice, we demonstrated previously that the type 1 diabetes (T1D) protection associated with the insulin-dependent diabetes (Idd)10 locus on chromosome 3, originally identified by linkage analysis, was in fact due to three closely linked Idd loci: Idd10, Idd18.1, and Idd18.3. In this study, we define two additional Idd loci--Idd18.2 and Idd18.4--within the boundaries of this cluster of disease-associated genes. Idd18.2 is 1.31 Mb and contains 18 genes, including Ptpn22, which encodes a phosphatase that negatively regulates T and B cell signaling. The human ortholog of Ptpn22, PTPN22, is associated with numerous autoimmune diseases, including T1D. We, therefore, assessed Ptpn22 as a candidate for Idd18.2; resequencing of the NOD Ptpn22 allele revealed 183 single nucleotide polymorphisms with the C57BL/6J (B6) allele--6 exonic and 177 intronic. Functional studies showed higher expression of full-length Ptpn22 RNA and protein, and decreased TCR signaling in congenic strains with B6-derived Idd18.2 susceptibility alleles. The 953-kb Idd18.4 locus contains eight genes, including the candidate Cd2. The CD2 pathway is associated with the human autoimmune disease, multiple sclerosis, and mice with NOD-derived susceptibility alleles at Idd18.4 have lower CD2 expression on B cells. Furthermore, we observed that susceptibility alleles at Idd18.2 can mask the protection provided by Idd10/Cd101 or Idd18.1/Vav3 and Idd18.3. In summary, we describe two new T1D loci, Idd18.2 and Idd18.4, candidate genes within each region, and demonstrate the complex nature of genetic interactions underlying the development of T1D in the NOD mouse model.


Subject(s)
CD2 Antigens/genetics , Chromosomes, Mammalian/genetics , Diabetes Mellitus, Type 1/genetics , Genetic Predisposition to Disease , Polymorphism, Single Nucleotide , Protein Tyrosine Phosphatase, Non-Receptor Type 22/genetics , Alleles , Animals , B-Lymphocytes/immunology , B-Lymphocytes/pathology , CD2 Antigens/immunology , Chromosomes, Mammalian/immunology , Diabetes Mellitus, Type 1/immunology , Gene Expression Regulation/immunology , Genetic Loci/immunology , Humans , Mice , Mice, Inbred NOD , Mice, Transgenic , Molecular Sequence Data , Protein Tyrosine Phosphatase, Non-Receptor Type 22/immunology , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocytes/immunology , T-Lymphocytes/pathology
10.
PLoS Med ; 13(10): e1002139, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27727279

ABSTRACT

BACKGROUND: Interleukin-2 (IL-2) has an essential role in the expansion and function of CD4+ regulatory T cells (Tregs). Tregs reduce tissue damage by limiting the immune response following infection and regulate autoreactive CD4+ effector T cells (Teffs) to prevent autoimmune diseases, such as type 1 diabetes (T1D). Genetic susceptibility to T1D causes alterations in the IL-2 pathway, a finding that supports Tregs as a cellular therapeutic target. Aldesleukin (Proleukin; recombinant human IL-2), which is administered at high doses to activate the immune system in cancer immunotherapy, is now being repositioned to treat inflammatory and autoimmune disorders at lower doses by targeting Tregs. METHODS AND FINDINGS: To define the aldesleukin dose response for Tregs and to find doses that increase Tregs physiologically for treatment of T1D, a statistical and systematic approach was taken by analysing the pharmacokinetics and pharmacodynamics of single doses of subcutaneous aldesleukin in the Adaptive Study of IL-2 Dose on Regulatory T Cells in Type 1 Diabetes (DILT1D), a single centre, non-randomised, open label, adaptive dose-finding trial with 40 adult participants with recently diagnosed T1D. The primary endpoint was the maximum percentage increase in Tregs (defined as CD3+CD4+CD25highCD127low) from the baseline frequency in each participant measured over the 7 d following treatment. There was an initial learning phase with five pairs of participants, each pair receiving one of five pre-assigned single doses from 0.04 × 106 to 1.5 × 106 IU/m2, in order to model the dose-response curve. Results from each participant were then incorporated into interim statistical modelling to target the two doses most likely to induce 10% and 20% increases in Treg frequencies. Primary analysis of the evaluable population (n = 39) found that the optimal doses of aldesleukin to induce 10% and 20% increases in Tregs were 0.101 × 106 IU/m2 (standard error [SE] = 0.078, 95% CI = -0.052, 0.254) and 0.497 × 106 IU/m2 (SE = 0.092, 95% CI = 0.316, 0.678), respectively. On analysis of secondary outcomes, using a highly sensitive IL-2 assay, the observed plasma concentrations of the drug at 90 min exceeded the hypothetical Treg-specific therapeutic window determined in vitro (0.015-0.24 IU/ml), even at the lowest doses (0.040 × 106 and 0.045 × 106 IU/m2) administered. A rapid decrease in Treg frequency in the circulation was observed at 90 min and at day 1, which was dose dependent (mean decrease 11.6%, SE = 2.3%, range 10.0%-48.2%, n = 37), rebounding at day 2 and increasing to frequencies above baseline over 7 d. Teffs, natural killer cells, and eosinophils also responded, with their frequencies rapidly and dose-dependently decreased in the blood, then returning to, or exceeding, pretreatment levels. Furthermore, there was a dose-dependent down modulation of one of the two signalling subunits of the IL-2 receptor, the ß chain (CD122) (mean decrease = 58.0%, SE = 2.8%, range 9.8%-85.5%, n = 33), on Tregs and a reduction in their sensitivity to aldesleukin at 90 min and day 1 and 2 post-treatment. Due to blood volume requirements as well as ethical and practical considerations, the study was limited to adults and to analysis of peripheral blood only. CONCLUSIONS: The DILT1D trial results, most notably the early altered trafficking and desensitisation of Tregs induced by a single ultra-low dose of aldesleukin that resolves within 2-3 d, inform the design of the next trial to determine a repeat dosing regimen aimed at establishing a steady-state Treg frequency increase of 20%-50%, with the eventual goal of preventing T1D. TRIAL REGISTRATION: ISRCTN Registry ISRCTN27852285; ClinicalTrials.gov NCT01827735.


Subject(s)
Diabetes Mellitus, Type 1/prevention & control , Interleukin-2/analogs & derivatives , T-Lymphocytes, Regulatory/drug effects , Adolescent , Adult , Biomarkers , Chemokines/biosynthesis , Dose-Response Relationship, Drug , Eosinophils/drug effects , Female , Humans , Immunophenotyping , Inflammation Mediators/metabolism , Interleukin-2/adverse effects , Interleukin-2/pharmacology , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Lymphocyte Count , Male , Middle Aged , Recombinant Proteins/adverse effects , Recombinant Proteins/pharmacology , Young Adult
11.
J Immunol ; 193(2): 889-900, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24928993

ABSTRACT

Expression of the CTLA-4 gene is absolutely required for immune homeostasis, but aspects of its molecular nature remain undefined. In particular, the characterization of the soluble CTLA-4 (sCTLA-4) protein isoform generated by an alternatively spliced mRNA of CTLA4 lacking transmembrane-encoding exon 3 has been hindered by the difficulty in distinguishing it from the transmembrane isoform of CTLA-4, Tm-CTLA-4. In the current study, sCTLA-4 has been analyzed using novel mAbs and polyclonal Abs specific for its unique C-terminal amino acid sequence. We demonstrate that the sCTLA-4 protein is secreted at low levels following the activation of primary human CD4(+) T cells and is increased only rarely in the serum of autoimmune patients. Unexpectedly, during our studies aimed to define the kinetics of sCTLA-4 produced by activated human CD4(+) T cells, we discovered that Tm-CTLA-4 is associated with microvesicles produced by the activated cells. The functional roles of sCTLA-4 and microvesicle-associated Tm-CTLA-4 warrant further investigation, especially as they relate to the multiple mechanisms of action described for the more commonly studied cell-associated Tm-CTLA-4.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , CTLA-4 Antigen/metabolism , Cytoplasmic Vesicles/metabolism , Membrane Glycoproteins/metabolism , Adult , Animals , Antibodies, Monoclonal/immunology , Antibody Specificity/immunology , Blotting, Western , CTLA-4 Antigen/blood , CTLA-4 Antigen/genetics , Cells, Cultured , Cytoplasmic Vesicles/ultrastructure , Diabetes Mellitus, Type 1/blood , Female , Graves Disease/blood , HeLa Cells , Humans , Immunoassay , Male , Membrane Glycoproteins/blood , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Microscopy, Immunoelectron , Middle Aged , Protein Isoforms/genetics , Protein Isoforms/immunology , Protein Isoforms/metabolism , Solubility , Young Adult
12.
Nat Genet ; 39(9): 1074-82, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17676041

ABSTRACT

Genome-wide association studies are now identifying disease-associated chromosome regions. However, even after convincing replication, the localization of the causal variant(s) requires comprehensive resequencing, extensive genotyping and statistical analyses in large sample sets leading to targeted functional studies. Here, we have localized the type 1 diabetes (T1D) association in the interleukin 2 receptor alpha (IL2RA) gene region to two independent groups of SNPs, spanning overlapping regions of 14 and 40 kb, encompassing IL2RA intron 1 and the 5' regions of IL2RA and RBM17 (odds ratio = 2.04, 95% confidence interval = 1.70-2.45; P = 1.92 x 10(-28); control frequency = 0.635). Furthermore, we have associated IL2RA T1D susceptibility genotypes with lower circulating levels of the biomarker, soluble IL-2RA (P = 6.28 x 10(-28)), suggesting that an inherited lower immune responsiveness predisposes to T1D.


Subject(s)
Chromosome Mapping/methods , Diabetes Mellitus, Type 1/genetics , Interleukin-2 Receptor alpha Subunit/genetics , Polymorphism, Single Nucleotide , Diabetes Mellitus, Type 1/blood , Family Health , Female , Gene Frequency , Genetic Predisposition to Disease , Genotype , Haplotypes , Humans , Interleukin-2 Receptor alpha Subunit/blood , Linkage Disequilibrium , Logistic Models , Male , Odds Ratio , Phenotype
13.
Nat Genet ; 39(3): 329-37, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17277778

ABSTRACT

Autoimmune diseases are thought to result from imbalances in normal immune physiology and regulation. Here, we show that autoimmune disease susceptibility and resistance alleles on mouse chromosome 3 (Idd3) correlate with differential expression of the key immunoregulatory cytokine interleukin-2 (IL-2). In order to test directly that an approximately twofold reduction in IL-2 underpins the Idd3-linked destabilization of immune homeostasis, we show that engineered haplodeficiency of Il2 gene expression not only reduces T cell IL-2 production by twofold but also mimics the autoimmune dysregulatory effects of the naturally occurring susceptibility alleles of Il2. Reduced IL-2 production achieved by either genetic mechanism correlates with reduced function of CD4(+) CD25(+) regulatory T cells, which are critical for maintaining immune homeostasis.


Subject(s)
Autoimmunity/genetics , Diabetes Mellitus, Type 1/immunology , Interleukin-2/genetics , T-Lymphocytes, Regulatory/immunology , Alleles , Animals , Autoimmunity/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/metabolism , Homeostasis/immunology , Interleukin-2/biosynthesis , Interleukin-2/immunology , Mice , Mice, Congenic , Mice, Inbred NOD , T-Lymphocytes, Regulatory/metabolism , Transcription, Genetic
14.
Nat Genet ; 39(7): 857-64, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17554260

ABSTRACT

The Wellcome Trust Case Control Consortium (WTCCC) primary genome-wide association (GWA) scan on seven diseases, including the multifactorial autoimmune disease type 1 diabetes (T1D), shows associations at P < 5 x 10(-7) between T1D and six chromosome regions: 12q24, 12q13, 16p13, 18p11, 12p13 and 4q27. Here, we attempted to validate these and six other top findings in 4,000 individuals with T1D, 5,000 controls and 2,997 family trios independent of the WTCCC study. We confirmed unequivocally the associations of 12q24, 12q13, 16p13 and 18p11 (P(follow-up)

Subject(s)
Chromosome Mapping , Diabetes Mellitus, Type 1/genetics , Genetic Predisposition to Disease , Genome, Human , Adolescent , Case-Control Studies , Humans , Polymorphism, Single Nucleotide
15.
Diabetologia ; 58(4): 781-90, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25652388

ABSTRACT

AIMS/HYPOTHESIS: Type 1 diabetes results from the autoimmune destruction of insulin-secreting pancreatic beta cells by T cells. Despite the established role of T cells in the pathogenesis of the disease, to date, with the exception of the identification of islet-specific T effector (Teff) cells, studies have mostly failed to identify reproducible alterations in the frequency or function of T cell subsets in peripheral blood from patients with type 1 diabetes. METHODS: We assessed the production of the proinflammatory cytokines IL-21, IFN-γ and IL-17 in peripheral blood mononuclear cells from 69 patients with type 1 diabetes and 61 healthy donors. In an additional cohort of 30 patients with type 1 diabetes and 32 healthy donors, we assessed the frequency of circulating T follicular helper (Tfh) cells in whole blood. IL-21 and IL-17 production was also measured in peripheral blood mononuclear cells (PBMCs) from a subset of 46 of the 62 donors immunophenotyped for Tfh. RESULTS: We found a 21.9% (95% CI 5.8, 40.2; p = 3.9 × 10(-3)) higher frequency of IL-21(+) CD45RA(-) memory CD4(+) Teffs in patients with type 1 diabetes (geometric mean 5.92% [95% CI 5.44, 6.44]) compared with healthy donors (geometric mean 4.88% [95% CI 4.33, 5.50]). Consistent with this finding, we found a 14.9% increase in circulating Tfh cells in the patients (95% CI 2.9, 26.9; p = 0.016). CONCLUSIONS/INTERPRETATION: These results indicate that increased IL-21 production is likely to be an aetiological factor in the pathogenesis of type 1 diabetes that could be considered as a potential therapeutic target.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Immunologic Memory , Interleukins/immunology , T-Lymphocytes, Helper-Inducer/immunology , Adolescent , Adult , Biomarkers/metabolism , Case-Control Studies , Cells, Cultured , Child , Diabetes Mellitus, Type 1/diagnosis , Diabetes Mellitus, Type 1/metabolism , Female , Humans , Immunophenotyping , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-17/immunology , Interleukin-17/metabolism , Interleukins/metabolism , Lymphocyte Count , Male , Middle Aged , T-Lymphocytes, Helper-Inducer/metabolism , Up-Regulation , Young Adult
16.
J Autoimmun ; 56: 66-80, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25457307

ABSTRACT

Regulatory T cells (Tregs) expressing FOXP3 are essential for the maintenance of self-tolerance and are deficient in many common autoimmune diseases. Immune tolerance is maintained in part by IL-2 and deficiencies in the IL-2 pathway cause reduced Treg function and an increased risk of autoimmunity. Recent studies expanding Tregs in vivo with low-dose IL-2 achieved major clinical successes highlighting the potential to optimize this pleiotropic cytokine for inflammatory and autoimmune disease indications. Here we compare the clinically approved IL-2 molecule, Proleukin, with two engineered IL-2 molecules with long half-lives owing to their fusion in monovalent and bivalent stoichiometry to a non-FcRγ binding human IgG1. Using nonhuman primates, we demonstrate that single ultra-low doses of IL-2 fusion proteins induce a prolonged state of in vivo activation that increases Tregs for an extended period of time similar to multiple-dose Proleukin. One of the common pleiotropic effects of high dose IL-2 treatment, eosinophilia, is eliminated at doses of the IL-2 fusion proteins that greatly expand Tregs. The long half-lives of the IL-2 fusion proteins facilitated a detailed characterization of an IL-2 dose response driving Treg expansion that correlates with increasingly sustained, suprathreshold pSTAT5a induction and subsequent sustained increases in the expression of CD25, FOXP3 and Ki-67 with retention of Treg-specific epigenetic signatures at FOXP3 and CTLA4.


Subject(s)
Interleukin-2/metabolism , Signal Transduction , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Animals , Biomarkers/metabolism , CTLA-4 Antigen/metabolism , Dose-Response Relationship, Drug , Eosinophilia/chemically induced , Female , Forkhead Transcription Factors/metabolism , Humans , Interleukin-2/analogs & derivatives , Interleukin-2/pharmacology , Interleukin-2 Receptor alpha Subunit/deficiency , Interleukin-2 Receptor alpha Subunit/metabolism , Lymphocyte Count , Macaca fascicularis , Male , Mice , Mice, Knockout , Phenotype , Phosphorylation/drug effects , Protein Binding , Recombinant Fusion Proteins/pharmacology , Recombinant Proteins/pharmacology , STAT5 Transcription Factor/metabolism , Signal Transduction/drug effects , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/drug effects
17.
J Immunol ; 190(7): 3109-20, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23427248

ABSTRACT

In the NOD mouse model of type 1 diabetes, insulin-dependent diabetes (Idd) loci control the development of insulitis and diabetes. Independently, protective alleles of Idd3/Il2 or Idd5 are able to partially protect congenic NOD mice from insulitis and diabetes, and to partially tolerize islet-specific CD8(+) T cells. However, when the two regions are combined, mice are almost completely protected, strongly suggesting the existence of genetic interactions between the two loci. Idd5 contains at least three protective subregions/causative gene candidates, Idd5.1/Ctla4, Idd5.2/Slc11a1, and Idd5.3/Acadl, yet it is unknown which of them interacts with Idd3/Il2. Through the use of a series of novel congenic strains containing the Idd3/Il2 region and different combinations of Idd5 subregion(s), we defined these genetic interactions. The combination of Idd3/Il2 and Idd5.3/Acadl was able to provide nearly complete protection from type 1 diabetes, but all three Idd5 subregions were required to protect from insulitis and fully restore self-tolerance. By backcrossing a Slc11a1 knockout allele onto the NOD genetic background, we have demonstrated that Slc11a1 is responsible for the diabetes protection resulting from Idd5.2. We also used Slc11a1 knockout-SCID and Idd5.2-SCID mice to show that both loss-of-function alleles provide protection from insulitis when expressed on the SCID host alone. These results lend further support to the hypothesis that Slc11a1 is Idd5.2.


Subject(s)
Diabetes Mellitus, Type 1/genetics , Epistasis, Genetic , Quantitative Trait Loci , Alleles , Animals , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Diabetes Mellitus, Type 1/immunology , Female , Genetic Predisposition to Disease , Glucose-6-Phosphatase/immunology , Immune Tolerance/genetics , Islets of Langerhans/immunology , Mice , Mice, Inbred NOD , Mice, SCID , Proteins/immunology
18.
J Immunol ; 190(6): 2554-66, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23418630

ABSTRACT

As the thymus involutes with age, the maintenance of peripheral naive T cells in humans becomes strongly dependent on peripheral cell division. However, mechanisms that orchestrate homeostatic division remain unclear. In this study we present evidence that the frequency of naive CD4 T cells that express CD25 (IL-2 receptor α-chain) increases with age on subsets of both CD31(+) and CD31(-) naive CD4 T cells. Analyses of TCR excision circles from sorted subsets indicate that CD25(+) naive CD4 T cells have undergone more rounds of homeostatic proliferation than their CD25(-) counterparts in both the CD31(+) and CD31(-) subsets, indicating that CD25 is a marker of naive CD4 T cells that have preferentially responded to survival signals from self-Ags or cytokines. CD25 expression on CD25(-) naive CD4 T cells can be induced by IL-7 in vitro in the absence of TCR activation. Although CD25(+) naive T cells respond to lower concentrations of IL-2 as compared with their CD25(-) counterparts, IL-2 responsiveness is further increased in CD31(-) naive T cells by their expression of the signaling IL-2 receptor ß-chain CD122, forming with common γ-chain functional high-affinity IL-2 receptors. CD25 plays a role during activation: CD25(+) naive T cells stimulated in an APC-dependent manner were shown to produce increased levels of IL-2 as compared with their CD25(-) counterparts. This study establishes CD25(+) naive CD4 T cells, which are further delineated by CD31 expression, as a major functionally distinct immune cell subset in humans that warrants further characterization in health and disease.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/immunology , Cellular Senescence/immunology , Receptors, Interleukin-2/metabolism , Thymus Gland/immunology , Thymus Gland/metabolism , Adult , Age Factors , CD4-Positive T-Lymphocytes/cytology , Cell Death/genetics , Cell Death/immunology , Cell Differentiation/genetics , Cells, Cultured , Cellular Senescence/genetics , Child , Humans , Immunophenotyping , Interleukin-2 Receptor alpha Subunit/biosynthesis , Interleukin-2 Receptor alpha Subunit/blood , Interleukin-2 Receptor alpha Subunit/genetics , Protein Binding/genetics , Protein Binding/immunology , Receptors, Interleukin-2/biosynthesis , Receptors, Interleukin-2/physiology , Thymus Gland/cytology , Young Adult
19.
Nat Genet ; 38(4): 479-83, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16550170

ABSTRACT

Type 1 diabetes is an autoimmune disease influenced by multiple genetic loci. Although more than 20 insulin-dependent diabetes (Idd) loci have been implicated in the nonobese diabetic (NOD) mouse model, few causal gene variants have been identified. Here we show that RNA interference (RNAi) can be used to probe candidate genes in this disease model. Slc11a1 encodes a phagosomal ion transporter, Nramp1, that affects resistance to intracellular pathogens and influences antigen presentation. This gene is the strongest candidate among the 42 genes in the Idd5.2 region; a naturally occurring mutation in the protective Idd5.2 haplotype results in loss of function of the Nramp1 protein. Using lentiviral transgenesis, we generated NOD mice in which Slc11a1 is silenced by RNAi. Silencing reduced the frequency of type 1 diabetes, mimicking the protective Idd5.2 region. Our results demonstrate a role for Slc11a1 in modifying susceptibility to type 1 diabetes and illustrate that RNAi can be used to study causal genes in a mammalian model organism.


Subject(s)
Cation Transport Proteins/genetics , Diabetes Mellitus, Type 1/genetics , Genetic Predisposition to Disease , RNA Interference , Animals , Female , Male , Mice , Mice, Inbred NOD
20.
J Immunol ; 188(11): 5267-75, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22539785

ABSTRACT

PTPN22 encodes a tyrosine phosphatase that inhibits Src-family kinases responsible for Ag receptor signaling in lymphocytes and is strongly linked with susceptibility to a number of autoimmune diseases. As strength of TCR signal is critical to the thymic selection of regulatory T cells (Tregs), we examined the effect of murine PTPN22 deficiency on Treg development and function. In the thymus, numbers of pre-Tregs and Tregs increased inversely with the level of PTPN22. This increase in Tregs persisted in the periphery and could play a key part in the reduced severity observed in the PTPN22-deficient mice of experimental autoimmune encephalomyelitis, a mouse model of multiple sclerosis. This could explain the lack of association of certain autoimmune conditions with PTPN22 risk alleles.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/enzymology , Encephalomyelitis, Autoimmune, Experimental/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 22/physiology , T-Lymphocytes, Regulatory/enzymology , T-Lymphocytes, Regulatory/immunology , Thymus Gland/enzymology , Thymus Gland/immunology , Animals , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Protein Tyrosine Phosphatase, Non-Receptor Type 22/biosynthesis , Protein Tyrosine Phosphatase, Non-Receptor Type 22/deficiency , T-Lymphocytes, Regulatory/pathology , Thymus Gland/pathology , Up-Regulation/immunology
SELECTION OF CITATIONS
SEARCH DETAIL