Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.382
Filter
1.
Nat Commun ; 15(1): 3810, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38714671

ABSTRACT

Previous studies have revealed heterogeneity in the progression to clinical type 1 diabetes in children who develop islet-specific antibodies either to insulin (IAA) or glutamic acid decarboxylase (GADA) as the first autoantibodies. Here, we test the hypothesis that children who later develop clinical disease have different early immune responses, depending on the type of the first autoantibody to appear (GADA-first or IAA-first). We use mass cytometry for deep immune profiling of peripheral blood mononuclear cell samples longitudinally collected from children who later progressed to clinical disease (IAA-first, GADA-first, ≥2 autoantibodies first groups) and matched for age, sex, and HLA controls who did not, as part of the Type 1 Diabetes Prediction and Prevention study. We identify differences in immune cell composition of children who later develop disease depending on the type of autoantibodies that appear first. Notably, we observe an increase in CD161 expression in natural killer cells of children with ≥2 autoantibodies and validate this in an independent cohort. The results highlight the importance of endotype-specific analyses and are likely to contribute to our understanding of pathogenic mechanisms underlying type 1 diabetes development.


Subject(s)
Autoantibodies , Diabetes Mellitus, Type 1 , Glutamate Decarboxylase , Immunity, Cellular , Humans , Diabetes Mellitus, Type 1/immunology , Autoantibodies/immunology , Autoantibodies/blood , Child , Female , Male , Glutamate Decarboxylase/immunology , Child, Preschool , Adolescent , Killer Cells, Natural/immunology , Leukocytes, Mononuclear/immunology , Insulin/immunology , Islets of Langerhans/immunology , Disease Progression
2.
Pharmacol Res ; 203: 107157, 2024 May.
Article in English | MEDLINE | ID: mdl-38531504

ABSTRACT

There are multiple disease-modifying immunotherapies showing the potential of preventing or delaying the progression of type 1 diabetes (T1D). We designed and performed this systematic review and meta-analysis to gain an overview of what a role immunotherapy plays in the treatment of T1D. We searched PubMed, Embase and Cochrane Central Register of Controlled Trials (CENTRAL) from inception to December 2023. We included clinical trials of immunotherapy conducted in patients with T1D that reported the incidence of hypoglycemia or changes from baseline in at least one of following outcomes: 2 h and 4 h mixed-meal-stimulated C-peptide area under the curve (AUC), fasting C-peptide, daily insulin dosage, glycated hemoglobin (HbA1c) and fasting plasma glucose (FPG). The results were computed as the weighted mean differences (WMDs) or odds ratios (ORs) and 95% confidence intervals (CIs) in random-effect model. In all, 34 clinical trials were included. When compared with control groups, 2 h C-peptide AUC was marginally higher in patient treated with nonantigen-based immunotherapies (WMD, 0.04nmol/L, 95% CI, 0.00-0.09 nmol/L, P=0.05), which was mainly driven by the effects of T cell-targeted therapy. A greater preservation in 4 h C-peptide AUC was observed in patients with nonantigen-based immunotherapies (WMD, 0.10nmol/L, 95% CI, 0.04-0.16 nmol/L, P=0.0007), which was mainly driven by the effects of tumor necrosis factor α (TNF-α) inhibitor and T cell-targeted therapy. After excluding small-sample trials, less daily insulin dosage was observed in patient treated with nonantigen-based immunotherapies when compared with control groups (WMD, -0.07units/kg/day, 95% CI, -0.11 to -0.03units/kg/day, P=0.0004). The use of antigen-based immunotherapies was also associated with a lower daily insulin dosage versus control groups (WMD, -0.11units/kg/day, 95% CI, -0.23 to -0.00units/kg/day, P=0.05). However, changes of HbA1c or FPG were comparable between nonantigen-based immunotherapies or antigen-based immunotherapies and control groups. The risk of hypoglycemia was not increased in patients treated with nonantigen-based immunotherapies or patients treated with antigen-based immunotherapies when compared with control groups. In conclusion, nonantigen-based immunotherapies were associated with a preservation of 2 h and 4 h C-peptide AUC in patients with T1D when compared with the controls, which was mainly driven by the effects of TNF-a inhibitor and T cell-targeted therapy. Both nonantigen-based immunotherapies and antigen-based immunotherapies tended to reduce the daily insulin dosage in patients with T1D when compared with the controls. However, they did not contribute to a substantial improvement in HbA1c or FPG. Both nonantigen-based immunotherapies and antigen-based immunotherapies were well tolerated with not increased risk of hypoglycemia in patients with T1D.


Subject(s)
Diabetes Mellitus, Type 1 , Immunotherapy , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/therapy , Diabetes Mellitus, Type 1/drug therapy , Humans , Immunotherapy/methods , Hypoglycemic Agents/therapeutic use , Blood Glucose/drug effects , Insulin/therapeutic use , Insulin/immunology , Glycated Hemoglobin/metabolism
3.
Acta Pharmacol Sin ; 45(4): 674-685, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38097717

ABSTRACT

Autoimmune diseases (AIDs) arise from a breakdown in immunological self-tolerance, wherein the adaptive immune system mistakenly attacks healthy cells, tissues and organs. AIDs impose excessive treatment costs and currently rely on non-specific and universal immunosuppression, which only offer symptomatic relief without addressing the underlying causes. AIDs are driven by autoantigens, targeting the autoantigens holds great promise in transforming the treatment of these diseases. To achieve this goal, a comprehensive understanding of the pathogenic mechanisms underlying different AIDs and the identification of specific autoantigens are critical. In this review, we categorize AIDs based on their underlying causes and compile information on autoantigens implicated in each disease, providing a roadmap for the development of novel immunotherapy regimens. We will focus on type 1 diabetes (T1D), which is an autoimmune disease characterized by irreversible destruction of insulin-producing ß cells in the Langerhans islets of the pancreas. We will discuss insulin as possible autoantigen of T1D and its role in T1D pathogenesis. Finally, we will review current treatments of TID and propose a potentially effective immunotherapy targeting autoantigens.


Subject(s)
Autoantigens , Autoimmune Diseases , Diabetes Mellitus, Type 1 , Drug Discovery , Insulin , Humans , Autoantigens/immunology , Autoimmune Diseases/drug therapy , Autoimmune Diseases/immunology , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/immunology , Insulin/immunology
5.
J Trace Elem Med Biol ; 75: 127100, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36410305

ABSTRACT

BACKGROUND: Type 1 diabetes (T1D) exhibited sex-specific metabolic status including oxidative stress with dynamic change of trace elements, which emphasized the importance of the evaluation of trace elements according to sex. Besides, the most significant characteristic, insulin auto-antibodies, could not be found in all T1D patients, which needed the auxiliary prediction of clinical parameters. And it would benefit the early detection and treatment if some high-risk groups of T1D could predict and prevent the occurrence of disease through common clinical parameters. Hence, there was an urgent need to construct more effective and scientific statistical prediction models to serve clinic better. This study aimed to evaluate the sex-specific levels of trace elements and the relationship between trace elements and clinical parameters in T1D, and construct sex-specific auxiliary prediction model combined with trace elements and clinical parameters. METHODS: A total of 105 T1D patients with negative insulin auto-antibodies and 105 age/sex-matched healthy individuals were enrolled in First Hospital of Jilin University. Inductively Coupled Plasma Mass Spectrometry was performed for the measurement of calcium (Ca), magnesium (Mg), zinc (Zn), copper (Cu), iron (Fe), selenium (Se) in the serum, and the data of clinical parameters were received from medical record system. The lambda-mu-sigma method was used to evaluate the relationship between abnormal clinical parameters and trace elements. Training set and validation set were divided for the construction of predictable models in males and females: clinical parameters model, trace element model and the combined model (clinical parameters and trace elements). Goodness fit test, decision curve analysis and other related statistical methods were used to perform data analysis. RESULTS: Lower levels of Mg, Ca, Fe in the serum were found in T1D population in females compared with healthy population, while levels of Fe, Zn and Cu of serum in T1D individuals were higher than those of healthy population in males. Levels of serum Mg, Fe and Cu in T1D group were found with significant sex difference for (P < 0.05), and the levels of Fe and Cu in serum of males were higher than those of females, level of serum Mg in males was lower than those of females. Levels of serum Mg and Zn showed fluctuation trend with increased numbers of abnormal clinical parameters (NACP) in males. Serum Zn in females showed consistent elevated trend with NACP; serum Se increased first and then decreased with NACP in males and females. The auxiliary prediction model (Triglyceride, Total protein, serum Mg) was found with the highest predicted efficiency in males (AUC=0.993), while the model in females (Apolipoprotein A, Creatinine, Fe, Se, Zn/Cu ratio) showed the best predicted efficiency (AUC=0.951). The models had passed the verification in validation set, and Chi-square goodness-of-fit test, DCA results both confirmed their satisfactory clinical applicability. CONCLUSION: Sex-specific difference were found in serum Mg, Fe and Cu in T1D. The combination of triglyceride, total protein and serum Mg for males, and apolipoprotein A, creatinine, Fe, Se, Zn/Cu ratio for females could effectively predict T1D in patients with negative anti-bodies, which would provide alarm for the population with high-risk of T1D and serve the T1D prediction in patients with negative anti-bodies.


Subject(s)
Diabetes Mellitus, Type 1 , Insulin Antibodies , Insulin , Trace Elements , Female , Humans , Male , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/diagnosis , Insulin/immunology , Insulin Antibodies/blood , Insulin Antibodies/immunology , Trace Elements/blood , Sex Factors , Apolipoproteins A/blood
6.
BMC Endocr Disord ; 22(1): 35, 2022 Feb 05.
Article in English | MEDLINE | ID: mdl-35123455

ABSTRACT

BACKGROUND: To ascertain the efficacy, safety, and immunogenicity from existing evidence via conducting a meta-analysis of randomized controlled trials between biosimilar and originator insulins. METHODS: The PubMed, Cochrane Library, EMBASE, and ClinicalTrails.gov were searched to identify head-to-head randomized controlled trials (RCTs) that directly compare the efficacy and safety of biosimilar insulin and its originator. Efficacy was assessed by change of HbA1C, fasting plasma glucose (laboratory or self-monitoring of blood glucose (SMBG)), and change all mean of 7 points- or 8 points- SMBG. Safety was assessed by change in proportion hypoglycemia and serious hypoglycemia. The occurrence of anti-insulin antibodies (AIAs) was also evaluated. RESULTS: Fourteen RCTs with 6188 patients from different countries were included. Data were pooled using a random-effects model and were expressed as the mean difference (MD), odds ratio (OR), and 95% confidence interval (CI). In efficacy, Insulin biosimilar products showed similar in change of HbA1C at weeks 26 and 52, the MD were 0.03 (95% CI - 0.02 to 0.07, p = 0.28), and 0.05 (95% CI - 0.05 to 0.15, p = 0.36), respectively. The proportion of HbA1C less than 7% at endpoint, the OR were 1.04 (95% CI 0.89 to 1.20, p = 0.64). The change of fasting plasma glucose (laboratory or SMBG) mmol/L in 24-52 weeks and change all mean of 7 points-/8 points- SMBG mmol/L in 24-52 weeks, the MD were 0.02 (95% CI - 0.20 to 0.24, p = 0.87) and - 0.34 (95% CI - 1.35 to 0.67, p = 0.51), respectively. In occurrence of hypoglycemia (≥ 1 events) and severe hypoglycemia, the OR were 0.96 (95% CI 0.85 to 1.09, p = 0.52) and 1.06 (95% CI 0.85 to 1.31, p = 0.62). The AIA was 1.02 (95% CI 0.90 to 1.16, p = 0.76). Analysis stratified by type of diabetes and duration of insulin. There was no significant difference between the biosimilar and their reference group in a different type of diabetes and different duration of insulin. CONCLUSIONS: Insulin biosimilar showed comparable characteristics with the reference drug in terms of efficacy, safety, immunogenicity, through comprehensive and specific conventional meta-analysis.


Subject(s)
Biosimilar Pharmaceuticals/pharmacology , Diabetes Mellitus/drug therapy , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Humans , Hypoglycemic Agents/immunology , Insulin/immunology
7.
Proc Natl Acad Sci U S A ; 119(6)2022 02 08.
Article in English | MEDLINE | ID: mdl-35131852

ABSTRACT

Homeostasis of metabolism by hormone production is crucial for maintaining physiological integrity, as disbalance can cause severe metabolic disorders such as diabetes mellitus. Here, we show that antibody-deficient mice and immunodeficiency patients have subphysiological blood glucose concentrations. Restoring blood glucose physiology required total IgG injections and insulin-specific IgG antibodies detected in total IgG preparations and in the serum of healthy individuals. In addition to the insulin-neutralizing anti-insulin IgG, we identified two fractions of anti-insulin IgM in the serum of healthy individuals. These autoreactive IgM fractions differ in their affinity to insulin. Interestingly, the low-affinity IgM fraction (anti-insulin IgMlow) neutralizes insulin and leads to increased blood glucose, whereas the high-affinity IgM fraction (anti-insulin IgMhigh) protects insulin from neutralization by anti-insulin IgG, thereby preventing blood glucose dysregulation. To demonstrate that anti-insulin IgMhigh acts as a protector of insulin and counteracts insulin neutralization by anti-insulin IgG, we expressed the variable regions of a high-affinity anti-insulin antibody as IgG and IgM. Remarkably, the recombinant anti-insulin IgMhigh normalized insulin function and prevented IgG-mediated insulin neutralization. These results suggest that autoreactive antibodies recognizing insulin are key regulators of blood glucose and metabolism, as they control the concentration of insulin in the blood. Moreover, our data suggest that preventing autoimmune damage and maintaining physiological homeostasis requires adaptive tolerance mechanisms generating high-affinity autoreactive IgM antibodies during memory responses.


Subject(s)
Autoantibodies/immunology , Blood Glucose/immunology , Homeostasis/immunology , Insulin/immunology , Animals , Antibody Affinity/immunology , Autoimmune Diseases/immunology , Female , Humans , Immune Tolerance/immunology , Immunoglobulin G/immunology , Immunoglobulin M/immunology , Mice , Mice, Inbred C57BL
8.
Front Endocrinol (Lausanne) ; 12: 764138, 2021.
Article in English | MEDLINE | ID: mdl-34803927

ABSTRACT

Immune checkpoint inhibitors have transformed the landscape of oncological therapy, but at the price of a new array of immune related adverse events. Among these is ß-cell failure, leading to checkpoint inhibitor-related autoimmune diabetes (CIADM) which entails substantial long-term morbidity. As our understanding of this novel disease grows, parallels and differences between CIADM and classic type 1 diabetes (T1D) may provide insights into the development of diabetes and identify novel potential therapeutic strategies. In this review, we outline the knowledge across the disciplines of endocrinology, oncology and immunology regarding the pathogenesis of CIADM and identify possible management strategies.


Subject(s)
Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/chemically induced , Immune Checkpoint Inhibitors/adverse effects , Antineoplastic Agents, Immunological/adverse effects , Antineoplastic Agents, Immunological/immunology , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/immunology , Humans , Hyperglycemia/blood , Hyperglycemia/chemically induced , Hyperglycemia/drug therapy , Hyperglycemia/immunology , Immune Checkpoint Inhibitors/immunology , Insulin/blood , Insulin/immunology , Insulin/therapeutic use , Risk Factors
9.
Proc Natl Acad Sci U S A ; 118(41)2021 10 12.
Article in English | MEDLINE | ID: mdl-34611019

ABSTRACT

Cytotoxic CD8 T lymphocytes play a central role in the tissue destruction of many autoimmune disorders. In type 1 diabetes (T1D), insulin and its precursor preproinsulin are major self-antigens targeted by T cells. We comprehensively examined preproinsulin specificity of CD8 T cells obtained from pancreatic islets of organ donors with and without T1D and identified epitopes throughout the entire preproinsulin protein and defective ribosomal products derived from preproinsulin messenger RNA. The frequency of preproinsulin-reactive T cells was significantly higher in T1D donors than nondiabetic donors and also differed by individual T1D donor, ranging from 3 to over 40%, with higher frequencies in T1D organ donors with HLA-A*02:01. Only T cells reactive to preproinsulin-related peptides isolated from T1D donors demonstrated potent autoreactivity. Reactivity to similar regions of preproinsulin was also observed in peripheral blood of a separate cohort of new-onset T1D patients. These findings have important implications for designing antigen-specific immunotherapies and identifying individuals that may benefit from such interventions.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Insulin/immunology , Islets of Langerhans/immunology , Protein Precursors/immunology , Adolescent , Adult , Autoantigens/immunology , Autoimmunity/immunology , Child , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 1/therapy , Female , HLA-A2 Antigen , Humans , Immunotherapy/methods , Islets of Langerhans/cytology , Male , Young Adult
11.
J Immunol ; 207(8): 2051-2059, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34526376

ABSTRACT

Ag-specific immunotherapy to restore immune tolerance to self-antigens, without global immune suppression, is a long-standing goal in the treatment of autoimmune disorders such as type 1 diabetes (T1D). However, vaccination with autoantigens such as insulin or glutamic acid decarboxylase have largely failed in human T1D trials. Induction and maintenance of peripheral tolerance by vaccination requires efficient autoantigen presentation by APCs. In this study, we show that a lipophilic modification at the N-terminal end of CD4+ epitopes (lipo-peptides) dramatically improves peptide Ag presentation. We designed amphiphilic lipo-peptides to efficiently target APCs in the lymph nodes by binding and trafficking with endogenous albumin. Additionally, we show that lipophilic modification anchors the peptide into the membranes of APCs, enabling a bivalent cell-surface Ag presentation. The s.c. injected lipo-peptide accumulates in the APCs in the lymph node, enhances the potency and duration of peptide Ag presentation by APCs, and induces Ag-specific immune tolerance that controls both T cell- and B cell-mediated immunity. Immunization with an amphiphilic insulin B chain 9-23 peptide, an immunodominant CD4+ T cell epitope in NOD mice, significantly suppresses the activation of T cells, increases inhibitory cytokine production, induces regulatory T cells, and delays the onset and lowers the incidence of T1D. Importantly, treatment with a lipophilic ß-cell peptide mixture delays progression to end-stage diabetes in acutely diabetic NOD mice, whereas the same doses of standard soluble peptides were not effective. Amphiphilic modification effectively enhances Ag presentation for peptide-based immune regulation of autoimmune diseases.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Epitopes, T-Lymphocyte/metabolism , Insulin/metabolism , Peptide Fragments/metabolism , Surface-Active Agents/metabolism , Albumins , Animals , Antigen Presentation , Female , Humans , Immune Tolerance , Immunization , Immunomodulation , Insulin/immunology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Peptide Fragments/immunology
12.
Front Immunol ; 12: 713276, 2021.
Article in English | MEDLINE | ID: mdl-34526989

ABSTRACT

Autoreactive CD8+ T cells play an indispensable key role in the destruction of pancreatic islet ß-cells and the initiation of type 1 diabetes (T1D). Insulin is an essential ß-cell autoantigen in T1D. An HLA-A*0201-restricted epitope of insulin A chain (mInsA2-10) is an immunodominant ligand for autoreactive CD8+ T cells in NOD.ß2mnull .HHD mice. Altered peptide ligands (APLs) carrying amino acid substitutions at T cell receptor (TCR) contact positions within an epitope are potential to modulate autoimmune responses via triggering altered TCR signaling. Here, we used a molecular simulation strategy to guide the generation of APL candidates by substitution of L-amino acids with D-amino acids at potential TCR contact residues (positions 4 and 6) of mInsA2-10, named mInsA2-10DQ4 and mInsA2-10DC6, respectively. We found that administration of mInsA2-10DQ4, but not DC6, significantly suppressed the development of T1D in NOD.ß2mnull .HHD mice. Mechanistically, treatment with mInsA2-10DQ4 not only notably eliminated mInsA2-10 autoreactive CD8+ T cell responses but also prevented the infiltration of CD4+ T and CD8+ T cells, as well as the inflammatory responses in the pancreas of NOD.ß2mnull.HHD mice. This study provides a new strategy for the development of APL vaccines for T1D prevention.


Subject(s)
Amino Acid Substitution , Diabetes Mellitus, Type 1/etiology , Epitopes/genetics , Epitopes/immunology , Insulin/genetics , Insulin/immunology , Animals , Diabetes Mellitus, Type 1/diagnosis , Diabetes Mellitus, Type 1/metabolism , Disease Models, Animal , Disease Susceptibility , Epitopes/chemistry , HLA-A2 Antigen/genetics , HLA-A2 Antigen/immunology , Humans , Insulin/chemistry , Insulin-Secreting Cells/immunology , Insulin-Secreting Cells/metabolism , Mice , Mice, Inbred NOD , Mice, Transgenic , Structure-Activity Relationship , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
13.
Front Immunol ; 12: 737428, 2021.
Article in English | MEDLINE | ID: mdl-34527002

ABSTRACT

The mechanisms underlying the major histocompatibility complex class II (MHCII) type 1 diabetes (T1D) association remain incompletely understood. We have previously shown that thymocytes expressing the highly diabetogenic, I-Ag7-restricted 4.1-T-cell receptor (TCR) are MHCII-promiscuous, and that, in MHCII-heterozygous mice, they sequentially undergo positive and negative selection/Treg deviation by recognizing pro- and anti-diabetogenic MHCII molecules on cortical thymic epithelial cells and medullary hematopoietic antigen-presenting cells (APCs), respectively. Here, we use a novel autoantigen discovery approach to define the antigenic specificity of this TCR in the context of I-Ag7. This was done by screening the ability of random epitope-GS linker-I- Aßg7 chain fusion pools to form agonistic peptide-MHCII complexes on the surface of I- Aαd chain-transgenic artificial APCs. Pool deconvolution, I-Ag7-binding register-fixing, TCR contact residue mapping, and alanine scanning mutagenesis resulted in the identification of a 4.1-TCR recognition motif XL(G/A)XEXE(D/E)X that was shared by seven agonistic hybrid insulin peptides (HIPs) resulting from the fusion of several different chromogranin A and/or insulin C fragments, including post-translationally modified variants. These data validate a novel, highly sensitive MHCII-restricted epitope discovery approach for orphan TCRs and suggest thymic selection of autoantigen-promiscuous TCRs as a mechanism for the murine T1D-I-Ag7-association.


Subject(s)
Autoantigens/immunology , CD4-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Insulin/immunology , Peptide Fragments/immunology , Receptors, Antigen, T-Cell/immunology , Animals , Autoantigens/genetics , Autoantigens/metabolism , CD4-Positive T-Lymphocytes/metabolism , CHO Cells , Coculture Techniques , Cricetulus , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/metabolism , Epitopes , HEK293 Cells , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/metabolism , Humans , Insulin/genetics , Insulin/metabolism , Jurkat Cells , Mice, Inbred NOD , Mice, Knockout , Peptide Fragments/genetics , Peptide Fragments/metabolism , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism
14.
Inflamm Res ; 70(10-12): 1043-1061, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34476533

ABSTRACT

BACKGROUND: The insulin/IGF-1 signaling pathway has a major role in the regulation of longevity both in Caenorhabditis elegans and mammalian species, i.e., reduced activity of this pathway extends lifespan, whereas increased activity accelerates the aging process. The insulin/IGF-1 pathway controls protein and energy metabolism as well as the proliferation and differentiation of insulin/IGF-1-responsive cells. Insulin/IGF-1 signaling also regulates the functions of the innate and adaptive immune systems. The purpose of this review was to elucidate whether insulin/IGF-1 signaling is linked to immunosuppressive STAT3 signaling which is known to promote the aging process. METHODS: Original and review articles encompassing the connections between insulin/IGF-1 and STAT3 signaling were examined from major databases including Pubmed, Scopus, and Google Scholar. RESULTS: The activation of insulin/IGF-1 receptors stimulates STAT3 signaling through the JAK and AKT-driven signaling pathways. STAT3 signaling is a major activator of immunosuppressive cells which are able to counteract the chronic low-grade inflammation associated with the aging process. However, the activation of STAT3 signaling stimulates a negative feedback response through the induction of SOCS factors which not only inhibit the activity of insulin/IGF-1 receptors but also that of many cytokine receptors. The inhibition of insulin/IGF-1 signaling evokes insulin resistance, a condition known to be increased with aging. STAT3 signaling also triggers the senescence of both non-immune and immune cells, especially through the activation of p53 signaling. CONCLUSIONS: Given that cellular senescence, inflammaging, and counteracting immune suppression increase with aging, this might explain why excessive insulin/IGF-1 signaling promotes the aging process.


Subject(s)
Aging/immunology , Immune Tolerance , Insulin-Like Growth Factor I/immunology , Insulin/immunology , STAT3 Transcription Factor/immunology , Animals , Cellular Senescence , Humans , Janus Kinases/immunology , Signal Transduction
15.
EMBO J ; 40(17): e107621, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34369608

ABSTRACT

The enormous diversity of antibody specificities is generated by random rearrangement of immunoglobulin gene segments and is important for general protection against pathogens. Since random rearrangement harbors the risk of producing self-destructive antibodies, it is assumed that autoreactive antibody specificities are removed during early B-cell development leading to a peripheral compartment devoid of autoreactivity. Here, we immunized wild-type mice with insulin as a common self-antigen and monitored diabetes symptoms as a measure for autoimmune disease. Our results show that autoreactive anti-insulin IgM and IgG antibodies associated with autoimmune diabetes can readily be generated in wild-type animals. Surprisingly, recall immunizations induced increased titers of high-affinity insulin-specific IgM, which prevented autoimmune diabetes. We refer to this phenomenon as adaptive tolerance, in which high-affinity memory IgM prevents autoimmune destruction by competing with self-destructive antibodies. Together, this study suggests that B-cell tolerance is not defined by the absolute elimination of autoreactive specificities, as harmful autoantibody responses can be generated in wild-type animals. In contrast, inducible generation of autoantigen-specific affinity-matured IgM acts as a protective mechanism preventing self-destruction.


Subject(s)
Antibodies, Neutralizing/immunology , Autoantibodies/immunology , Diabetes Mellitus, Type 1/immunology , Immunoglobulin M/immunology , Immunologic Memory , Insulin/immunology , Animals , B-Lymphocytes/immunology , Female , Immune Tolerance , Mice , Mice, Inbred C57BL
16.
Diabetologia ; 64(11): 2511-2516, 2021 11.
Article in English | MEDLINE | ID: mdl-34448034

ABSTRACT

AIMS/HYPOTHESIS: We examined whether the non-HLA susceptibility locus ERBB3/IKZF4 influences progression of type 1 diabetes stage specifically according to sex. METHODS: SNPs of ERBB3 (rs2292239 T/G) and IKZF4 (rs1701704 G/T) were screened by allelic discrimination quantitative PCR assay in first-degree relatives of type 1 diabetes patients who had developed at least one circulating autoantibody. The effect of ERBB3/IKZF4 genotypes and sex, on the progression of single autoantibody positivity to multiple autoantibody positivity and from multiple autoantibody positivity to diabetes, was studied by Kaplan-Meier analysis and multivariate Cox regression. RESULTS: In the cohort of autoantibody-positive first-degree relatives, the risk allele frequencies for ERBB3 rs2292239 (T) and IKZF4 rs1701704 (G) were increased. There was a significant male excess at the stage of multiple autoantibody positivity (p = 0.021). In Kaplan-Meier survival analysis, progression from single to multiple antibody positivity was delayed in female participants with genotype ERBB3 GG (p = 0.018, vs ERBB3 TG+TT) or IKZF4 TT (p = 0.023, vs IKZF4 GT+GG), but not in male participants. In multivariate Cox regression models, the interaction effects between female sex and ERBB3 GG (p = 0.012; HR = 0.305 [95% CI 0.120, 0.773]) or between female sex and IKZF4 TT (p = 0.011; HR = 0.329 [95% CI 0.140, 0.777]) emerged as potential determinants of delayed progression to multiple autoantibodies. The progression from multiple autoantibody positivity to type 1 diabetes appeared not to be influenced by ERBB3/IKZF4. CONCLUSIONS/INTERPRETATION: In siblings and offspring of type 1 diabetes patients, polymorphism in region ERBB3/IKZF4 may affect disease progression at the level of epitope spreading in female individuals. Our findings suggest that interaction between sex and ERBB3/IKZF4 may contribute to the post-pubertal male excess in type 1 diabetes.


Subject(s)
Autoantibodies/blood , Autoantigens/immunology , Diabetes Mellitus, Type 1/immunology , Epitopes/immunology , Ikaros Transcription Factor/genetics , Receptor, ErbB-3/genetics , Sex Characteristics , Adolescent , Adult , Child , Diabetes Mellitus, Type 1/genetics , Disease Progression , Female , Genetic Predisposition to Disease , Humans , Insulin/immunology , Male , Polymorphism, Single Nucleotide/genetics , Proportional Hazards Models , Real-Time Polymerase Chain Reaction , Receptor-Like Protein Tyrosine Phosphatases, Class 8/immunology , Zinc Transporter 8/immunology
17.
Genes (Basel) ; 12(6)2021 06 01.
Article in English | MEDLINE | ID: mdl-34205929

ABSTRACT

Genetic analyses of human type 1 diabetes (T1D) have yet to reveal a complete pathophysiologic mechanism. Inbred rats with a high-risk class II major histocompatibility complex (MHC) haplotype (RT1B/Du) can illuminate such mechanisms. Using T1D-susceptible LEW.1WR1 rats that express RT1B/Du and a susceptible allele of the Ubd promoter, we demonstrate that germline knockout of Tcrb-V13S1A1, which encodes the Vß13a T cell receptor ß chain, completely prevents diabetes. Using the RT1B/Du-identical LEW.1W rat, which does not develop T1D despite also having the same Tcrb-V13S1A1 ß chain gene but a different allele at the Ubd locus, we show that knockout of the Ubash3a regulatory gene renders these resistant rats relatively susceptible to diabetes. In silico structural modeling of the susceptible allele of the Vß13a TCR and its class II RT1u ligand suggests a mechanism by which a germline TCR ß chain gene could promote susceptibility to T1D in the absence of downstream immunoregulation like that provided by UBASH3A. Together these data demonstrate the critical contribution of the Vß13a TCR to the autoimmune synapse in T1D and the regulation of the response by UBASH3A. These experiments dissect the mechanisms by which MHC class II heterodimers, TCR and regulatory element interact to induce autoimmunity.


Subject(s)
Autoimmunity/genetics , Diabetes Mellitus, Type 1/genetics , Receptors, Antigen, T-Cell, alpha-beta/genetics , Animals , Diabetes Mellitus, Type 1/immunology , Genotype , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Insulin/chemistry , Insulin/immunology , Peptide Fragments/chemistry , Peptide Fragments/immunology , Protein Binding , Rats , Rats, Inbred Lew , Receptors, Antigen, T-Cell, alpha-beta/chemistry , Receptors, Antigen, T-Cell, alpha-beta/immunology
18.
Mol Metab ; 52: 101288, 2021 10.
Article in English | MEDLINE | ID: mdl-34242821

ABSTRACT

BACKGROUND: Since its discovery 100 years ago, insulin, as the 'cure' for type 1 diabetes, has rescued the lives of countless individuals. As the century unfolded and the autoimmune nature of type 1 diabetes was recognised, a darker side of insulin emerged. Autoimmunity to insulin was found to be an early marker of risk for type 1 diabetes in young children. In humans, it remains unclear if autoimmunity to insulin is primarily due to a defect in the beta cell itself or to dysregulated immune activation. Conversely, it may be secondary to beta-cell damage from an environmental agent (e.g., virus). Nevertheless, direct, interventional studies in non-obese diabetic (NOD) mouse models of type 1 diabetes point to a critical role for (pro)insulin as a primary autoantigen that drives beta cell pathology. SCOPE OF REVIEW: Modelled on Koch's postulates for the pathogenicity of an infectious agent, evidence for a pathogenic role of (pro)insulin as an autoantigen in type 1 diabetes, particularly applicable to the NOD mouse model, is reviewed. Evidence in humans remains circumstantial. Additionally, as (pro)insulin is a target of autoimmunity in type 1 diabetes, its application as a therapeutic tool to elicit antigen-specific immune tolerance is assessed. MAJOR CONCLUSIONS: Paradoxically, insulin is both a 'cure' and a potential 'cause' of type 1 diabetes, actively participating as an autoantigen to drive autoimmune destruction of beta cells - the instrument of its own destruction.


Subject(s)
Autoantibodies/immunology , Autoantigens/immunology , Diabetes Mellitus, Type 1/immunology , Insulin-Secreting Cells/pathology , Insulin/immunology , Animals , Autoantibodies/metabolism , Autoantigens/metabolism , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Disease Models, Animal , Humans , Insulin/metabolism , Insulin-Secreting Cells/immunology , Insulin-Secreting Cells/metabolism , Mice , Mice, Inbred NOD
19.
Front Immunol ; 12: 668680, 2021.
Article in English | MEDLINE | ID: mdl-34113344

ABSTRACT

Hybrid Insulin Peptides (HIPs), which consist of insulin fragments fused to other peptides from ß-cell secretory granule proteins, are CD4 T cell autoantigens in type 1 diabetes (T1D). We have studied HIPs and HIP-reactive CD4 T cells extensively in the context of the non-obese diabetic (NOD) mouse model of autoimmune diabetes and have shown that CD4 T cells specific for HIPs are major contributors to disease pathogenesis. Additionally, in the human context, HIP-reactive CD4 T cells can be found in the islets and peripheral blood of T1D patients. Here, we performed an in-depth characterization of the CD4 T cell response to a C-peptide/C-peptide HIP (HIP11) in human T1D. We identified the TCR expressed by the previously-reported HIP11-reactive CD4 T cell clone E2, which was isolated from the peripheral blood of a T1D patient, and determined that it recognizes HIP11 in the context of HLA-DQ2. We also identified a HIP11-specific TCR directly in the islets of a T1D donor and demonstrated that this TCR recognizes a different minimal epitope of HIP11 presented by HLA-DQ8. We generated and tested an HLA-DQ2 tetramer loaded with HIP11 that will enable direct ex vivo interrogation of CD4 T cell responses to HIP11 in human patients and control subjects. Using mass spectrometric analysis, we confirmed that HIP11 is present in human islets. This work represents an important step in characterizing the role of CD4 T cell responses to HIPs in human T1D.


Subject(s)
Autoantigens/immunology , C-Peptide/immunology , CD4-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Insulin/immunology , Islets of Langerhans/immunology , Receptors, Antigen, T-Cell/immunology , Autoantigens/metabolism , C-Peptide/metabolism , CD4-Positive T-Lymphocytes/metabolism , Diabetes Mellitus, Type 1/blood , Epitopes , Female , HLA-DQ Antigens/immunology , Humans , Insulin/metabolism , Islets of Langerhans/metabolism , K562 Cells , Male , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism
20.
Curr Opin Endocrinol Diabetes Obes ; 28(4): 390-396, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34091488

ABSTRACT

PURPOSE OF REVIEW: Loss of tolerance to insulin likely contributes to the immunopathogenesis of type 1 diabetes (T1D). Several large clinical trials and smaller mechanistic studies have failed to demonstrate the efficacy of insulin antigen therapy. The growing awareness of the heterogeneity of T1D likely affects the response to various immune therapies including insulin. Identification of biomarkers of clinical response will provide further insight into mechanisms leading to the disease and classify responders in the quest for personalized therapy. RECENT FINDINGS: Several biomarkers have identified subpopulations in posthoc analyses that showed benefit from oral insulin even though the placebo-controlled study was as a whole unsuccessful. High insulin autoantibody titer, low first phase insulin response, and high Diabetes Prevention Trial-Type 1 Risk Score identify at-risk relatives more likely to benefit from oral insulin. Future incorporation of human leukocyte antigen and the variable number of tandem repeats polymorphism located in the insulin gene promoter (INS VNTR) is of interest for both primary and secondary prevention studies. SUMMARY: Although primary and secondary prevention trials using oral insulin are ongoing, those completed have been largely unsuccessful. However, we believe that oral insulin should be considered in future trials as part of combination therapies as prerandomization biomarker testing is refined.


Subject(s)
Diabetes Mellitus, Type 1 , Hypoglycemic Agents , Immunotherapy , Insulin , Prediabetic State , Administration, Oral , Autoantibodies/immunology , Biomarkers/analysis , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Humans , Hypoglycemic Agents/immunology , Hypoglycemic Agents/therapeutic use , Immune Tolerance , Immunotherapy/methods , Insulin/immunology , Insulin/therapeutic use , Polymorphism, Genetic , Prediabetic State/drug therapy , Prediabetic State/genetics , Prediabetic State/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...