1
|
Jia X, Wenzlau JM, Zhang C, Dong F, Waugh K, Leslie RD, Rewers MJ, Michels AW, Yu L. Strong Association of Autoantibodies Targeting Deamidated Extracellular Epitopes of Insulinoma Antigen-2 With Clinical Onset of Type 1 Diabetes. Diabetes 2025; 74:544-553. [PMID: 39821270 PMCID: PMC11926269 DOI: 10.2337/db24-0571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 01/13/2025] [Indexed: 01/19/2025]
Abstract
ARTICLE HIGHLIGHTS CD4+ T cells from patients with type 1 diabetes (T1D) have a significant response to post-translationally modified (PTM) deamidated IA-2 peptides; autoantibodies to these PTM neoepitopes remain to be identified in T1D. We aimed to identify autoantibodies specifically targeting reported T-cell reactive, deamidated epitopes of IA-2 and explore their relationship with T1D development. Autoantibodies to deamidated IA-2 were specific to deamidated epitopes and were predominantly present during the late stages of T1D development, challenging the hypothesis that the loss of immune tolerance occurs via post-translational modification of islet antigens. Newly identified autoantibodies to deamidated IA-2 are new biomarkers of islet autoimmunity and have the potential to aid in T1D diagnosis.
Collapse
Affiliation(s)
- Xiaofan Jia
- Barbara Davis Center for Diabetes, University of Colorado, Aurora, CO
| | - Janet M. Wenzlau
- Barbara Davis Center for Diabetes, University of Colorado, Aurora, CO
| | - Caiguo Zhang
- Barbara Davis Center for Diabetes, University of Colorado, Aurora, CO
| | - Fran Dong
- Barbara Davis Center for Diabetes, University of Colorado, Aurora, CO
| | - Kathleen Waugh
- Barbara Davis Center for Diabetes, University of Colorado, Aurora, CO
| | - R. David Leslie
- Centre for Immunobiology, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, U.K
| | - Marian J. Rewers
- Barbara Davis Center for Diabetes, University of Colorado, Aurora, CO
| | - Aaron W. Michels
- Barbara Davis Center for Diabetes, University of Colorado, Aurora, CO
| | - Liping Yu
- Barbara Davis Center for Diabetes, University of Colorado, Aurora, CO
| | | |
Collapse
|
2
|
Alhamar G, Vinci C, Franzese V, Tramontana F, Le Goux N, Ludvigsson J, Nissim A, Strollo R. The role of oxidative post-translational modifications in type 1 diabetes pathogenesis. Front Immunol 2025; 16:1537405. [PMID: 40028329 PMCID: PMC11868110 DOI: 10.3389/fimmu.2025.1537405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Accepted: 01/22/2025] [Indexed: 03/05/2025] Open
Abstract
The pathogenesis of type 1 diabetes (T1D) involves a complex interplay of genetic predisposition, immune processes, and environmental factors, leading to the selective destruction of pancreatic beta-cells by the immune system. Emerging evidence suggests that intrinsic beta-cell factors, including oxidative stress and post-translational modifications (PTM) of beta-cell antigens, may also contribute to their immunogenicity, shedding new light on the multifaceted pathogenesis of T1D. Over the past 30 years, neoepitopes generated by PTMs have been hypothesized to play a role in T1D pathogenesis, but their involvement has only been systematically investigated in recent years. In this review, we explored the interplay between oxidative PTMs, neoepitopes, and T1D, highlighting oxidative stress as a pivotal factor in immune system dysfunction, beta-cell vulnerability, and disease onset.
Collapse
Affiliation(s)
- Ghadeer Alhamar
- Department of Immunology and Microbiology, Dasman Diabetes Institute, Dasman, Kuwait
| | - Chiara Vinci
- Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
| | - Valentina Franzese
- Department for the Promotion of Human Science and Quality of Life, San Raffaele Open University, Rome, Italy
- Department of Medicine, Fondazione Policlinico Universitario Campus Bio-Medico di Roma, Rome, Italy
- Department of Medicine, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Flavia Tramontana
- Department of Medicine, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Nelig Le Goux
- Biochemical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Johnny Ludvigsson
- Crown Princess Victoria Children’s Hospital and Division of Pediatrics, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Ahuva Nissim
- Biochemical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Rocky Strollo
- Department for the Promotion of Human Science and Quality of Life, San Raffaele Open University, Rome, Italy
- Department of Medicine, Fondazione Policlinico Universitario Campus Bio-Medico di Roma, Rome, Italy
| |
Collapse
|
3
|
Dabiri H, Sadeghizadeh M, Ziaei V, Moghadasi Z, Maham A, Hajizadeh-Saffar E, Habibi-Anbouhi M. Development of an ostrich-derived single-chain variable fragment (scFv) against PTPRN extracellular domain. Sci Rep 2024; 14:3689. [PMID: 38355744 PMCID: PMC10866909 DOI: 10.1038/s41598-024-53386-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Accepted: 01/31/2024] [Indexed: 02/16/2024] Open
Abstract
In type 1 diabetes, the immune system destroys pancreatic beta cells in an autoimmune condition. To overcome this disease, a specific monoclonal antibody that binds to pancreatic beta cells could be used for targeted immunotherapy. Protein tyrosine phosphatase receptor N (PTPRN) is one of the important surface antigen candidates. Due to its high sequence homology among mammals, so far, no single-chain monoclonal antibody has been produced against this receptor. In this study, we developed a novel single-chain variable fragment (scFv) against the PTPRN extracellular domain. To this aim, ostrich species was used as a host is far phylogenetically birds from mammals to construct a phage display library for the first time. An ostrich-derived scfv phage display library was prepared and biopanning steps were done to enrich and screen for isolating the best anti-PTPRN binders. An scFv with appropriate affinity and specificity to the PTPRN extracellular domain was selected and characterized by ELISA, western blotting, and flow cytometry. The anti-PTPRN scFv developed in this study could be introduced as an effective tool that can pave the way for the creation of antibody-based targeting systems in cooperation with the detection and therapy of type I diabetes.
Collapse
Affiliation(s)
- Hamed Dabiri
- Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Majid Sadeghizadeh
- Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Vahab Ziaei
- National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, Iran
| | - Zahra Moghadasi
- National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, Iran
| | - Ali Maham
- Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Ensiyeh Hajizadeh-Saffar
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
- Advanced Therapy Medicinal Product Technology Development Center (ATMP-TDC), Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | | |
Collapse
|
4
|
Jia X, Yu L. Understanding Islet Autoantibodies in Prediction of Type 1 Diabetes. J Endocr Soc 2023; 8:bvad160. [PMID: 38169963 PMCID: PMC10758755 DOI: 10.1210/jendso/bvad160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Indexed: 01/05/2024] Open
Abstract
As screening studies and preventive interventions for type 1 diabetes (T1D) advance rapidly, the utility of islet autoantibodies (IAbs) in T1D prediction comes with challenges for early and accurate disease progression prediction. Refining features of IAbs can provide more accurate risk assessment. The advances in islet autoantibodies assay techniques help to screen out islet autoantibodies with high efficiency and high disease specificity. Exploring new islet autoantibodies to neoepitopes/neoantigens remains a hot research field for improving prediction and disease pathogenesis. We will review the recent research progresses of islet autoantibodies to better understand the utility of islet autoantibodies in prediction of T1D.
Collapse
Affiliation(s)
- Xiaofan Jia
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Liping Yu
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, CO 80045, USA
| |
Collapse
|
5
|
Kawasaki E. Anti-Islet Autoantibodies in Type 1 Diabetes. Int J Mol Sci 2023; 24:10012. [PMID: 37373160 PMCID: PMC10298549 DOI: 10.3390/ijms241210012] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 06/08/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
Anti-islet autoantibodies serve as key markers in immune-mediated type 1 diabetes (T1D) and slowly progressive T1D (SPIDDM), also known as latent autoimmune diabetes in adults (LADA). Autoantibodies to insulin (IAA), glutamic acid decarboxylase (GADA), tyrosine phosphatase-like protein IA-2 (IA-2A), and zinc transporter 8 (ZnT8A) are currently employed in the diagnosis, pathological analysis, and prediction of T1D. GADA can also be detected in non-diabetic patients with autoimmune diseases other than T1D and may not necessarily reflect insulitis. Conversely, IA-2A and ZnT8A serve as surrogate markers of pancreatic β-cell destruction. A combinatorial analysis of these four anti-islet autoantibodies demonstrated that 93-96% of acute-onset T1D and SPIDDM cases were diagnosed as immune-mediated T1D, while the majority of fulminant T1D cases were autoantibody-negative. Evaluating the epitopes and immunoglobulin subclasses of anti-islet autoantibodies help distinguish between diabetes-associated and non-diabetes-associated autoantibodies and is valuable for predicting future insulin deficiency in SPIDDM (LADA) patients. Additionally, GADA in T1D patients with autoimmune thyroid disease reveals the polyclonal expansion of autoantibody epitopes and immunoglobulin subclasses. Recent advancements in anti-islet autoantibody assays include nonradioactive fluid-phase assays and the simultaneous determination of multiple biochemically defined autoantibodies. Developing a high-throughput assay for detecting epitope-specific or immunoglobulin isotype-specific autoantibodies will facilitate a more accurate diagnosis and prediction of autoimmune disorders. The aim of this review is to summarize what is known about the clinical significance of anti-islet autoantibodies in the pathogenesis and diagnosis of T1D.
Collapse
Affiliation(s)
- Eiji Kawasaki
- Diabetes Center, Shin-Koga Hospital, Kurume 830-8577, Japan
| |
Collapse
|
6
|
Strollo R, Vinci C, Man YKS, Bruzzaniti S, Piemonte E, Alhamar G, Briganti SI, Malandrucco I, Tramontana F, Fanali C, Garnett J, Buccafusca R, Guyer P, Mamula M, James EA, Pozzilli P, Ludvigsson J, Winyard PG, Galgani M, Nissim A. Autoantibody and T cell responses to oxidative post-translationally modified insulin neoantigenic peptides in type 1 diabetes. Diabetologia 2023; 66:132-146. [PMID: 36207582 PMCID: PMC9729141 DOI: 10.1007/s00125-022-05812-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 07/28/2022] [Indexed: 12/14/2022]
Abstract
AIMS/HYPOTHESIS Antibodies specific to oxidative post-translational modifications (oxPTM) of insulin (oxPTM-INS) are present in most individuals with type 1 diabetes, even before the clinical onset. However, the antigenic determinants of such response are still unknown. In this study, we investigated the antibody response to oxPTM-INS neoepitope peptides (oxPTM-INSPs) and evaluated their ability to stimulate humoral and T cell responses in type 1 diabetes. We also assessed the concordance between antibody and T cell responses to the oxPTM-INS neoantigenic peptides. METHODS oxPTM-INS was generated by exposing insulin to various reactive oxidants. The insulin fragments resulting from oxPTM were fractionated by size-exclusion chromatography further to ELISA and LC-MS/MS analysis to identify the oxidised peptide neoepitopes. Immunogenic peptide candidates were produced and then modified in house or designed to incorporate in silico-oxidised amino acids during synthesis. Autoantibodies to the oxPTM-INSPs were tested by ELISA using sera from 63 participants with new-onset type 1 diabetes and 30 control participants. An additional 18 fresh blood samples from participants with recently diagnosed type 1 diabetes, five with established disease, and from 11 control participants were used to evaluate, in parallel, CD4+ and CD8+ T cell activation by oxPTM-INSPs. RESULTS We observed antibody and T cell responses to three out of six LC-MS/MS-identified insulin peptide candidates: A:12-21 (SLYQLENYCN, native insulin peptide 3 [Nt-INSP-3]), B:11-30 (LVEALYLVCGERGFFYTPKT, Nt-INSP-4) and B:21-30 (ERGFFYTPKT, Nt-INSP-6). For Nt-INSP-4 and Nt-INSP-6, serum antibody binding was stronger in type 1 diabetes compared with healthy control participants (p≤0.02), with oxidised forms of ERGFFYTPKT, oxPTM-INSP-6 conferring the highest antibody binding (83% binders to peptide modified in house by hydroxyl radical [●OH] and >88% to in silico-oxidised peptide; p≤0.001 vs control participants). Nt-INSP-4 induced the strongest T cell stimulation in type 1 diabetes compared with control participants for both CD4+ (p<0.001) and CD8+ (p=0.049). CD4+ response to oxPTM-INSP-6 was also commoner in type 1 diabetes than in control participants (66.7% vs 27.3%; p=0.039). Among individuals with type 1 diabetes, the CD4+ response to oxPTM-INSP-6 was more frequent than to Nt-INSP-6 (66.7% vs 27.8%; p=0.045). Overall, 44.4% of patients showed a concordant autoimmune response to oxPTM-INSP involving simultaneously CD4+ and CD8+ T cells and autoantibodies. CONCLUSIONS/INTERPRETATION Our findings support the concept that oxidative stress, and neoantigenic epitopes of insulin, may be involved in the immunopathogenesis of type 1 diabetes.
Collapse
Affiliation(s)
- Rocky Strollo
- Department of Science and Technology for Humans and the Environment, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Chiara Vinci
- Biochemical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Y K Stella Man
- Biochemical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Sara Bruzzaniti
- Institute for Experimental Endocrinology and Oncology 'G. Salvatore', Consiglio Nazionale delle Ricerche, Naples, Italy
- Department of Biology, Università degli Studi di Napoli 'Federico II', Naples, Italy
| | - Erica Piemonte
- Department of Molecular Medicine and Medical Biotechnology, Università degli Studi di Napoli 'Federico II', Naples, Italy
| | - Ghadeer Alhamar
- Department of Medicine, Unit of Endocrinology & Diabetes, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Silvia Irina Briganti
- Department of Medicine, Unit of Endocrinology & Diabetes, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Ilaria Malandrucco
- The UOSD of Endocrinology and Metabolic Diseases, Azienda Sanitaria Locale (ASL) Frosinone, Frosinone, Italy
| | - Flavia Tramontana
- Department of Medicine, Unit of Endocrinology & Diabetes, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Chiara Fanali
- Department of Science and Technology for Humans and the Environment, Università Campus Bio-Medico di Roma, Rome, Italy
| | - James Garnett
- Centre for Host-Microbiome Interactions, Dental Institute, King's College London, London, UK
- School of Biological and Chemical Sciences, Queen Mary University of London, London, UK
| | - Roberto Buccafusca
- School of Biological and Chemical Sciences, Queen Mary University of London, London, UK
| | - Perrin Guyer
- Program for Translational Immunology, Benaroya Research Institute, Seattle, WA, USA
| | - Mark Mamula
- Department of Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Eddie A James
- Program for Translational Immunology, Benaroya Research Institute, Seattle, WA, USA
| | - Paolo Pozzilli
- Department of Medicine, Unit of Endocrinology & Diabetes, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Johnny Ludvigsson
- Division of Pediatrics, Department of Biomedical and Clinical Sciences, Crown Princess Victoria Children's Hospital, Linköping University, Linköping, Sweden
| | - Paul G Winyard
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, St Luke's Campus, Exeter, UK
| | - Mario Galgani
- Institute for Experimental Endocrinology and Oncology 'G. Salvatore', Consiglio Nazionale delle Ricerche, Naples, Italy
- Department of Molecular Medicine and Medical Biotechnology, Università degli Studi di Napoli 'Federico II', Naples, Italy
| | - Ahuva Nissim
- Biochemical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London, UK.
| |
Collapse
|
7
|
Mendis T, Filipova B, Wang JJ, Pietropaolo M, Jackson MW. Affinity purification of serum-derived anti-IA-2 autoantibodies in type 1 diabetes using a novel MBP-IA-2 fusion protein. Biochem Biophys Rep 2022; 33:101413. [PMID: 36578528 PMCID: PMC9791830 DOI: 10.1016/j.bbrep.2022.101413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/05/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Autoantibodies targeting epitopes contained within the intracellular domain (IC) of the protein phosphatase-like islet antigen 2 (IA-2) are a common marker of autoimmune type 1 diabetes (T1D), however the isolation of genuine, serum derived anti-IA-2 autoantibodies has proven challenging due to a lack of suitable bioassays. In the current study, an ELISA format was developed for affinity purification of human anti-IA-2ic autoantibodies utilizing a fusion protein (FP) incorporating maltose binding protein and the full-length IA-2IC domain. Using a T1D patient cohort validated for anti-IA-2ic autoantibodies by commercial ELISA, we demonstrate the MBP-IA-2ic FP ELISA detects serum anti-IA-2IC autoantibodies from 3 of 9 IA-2 positive patients. Further to this, a multi-plate MBP-IA-2ic FP ELISA protocol specifically affinity purifies IgG enriched for anti-IA-2ic autoantibodies. Interestingly, serum derived autoantibodies immobilised on the MBP-IA-2ic FP ELISA demonstrate increased Kappa light chain usage when compared to the respective total IgG derived from donor patients, suggesting a clonally restricted repertoire of anti-IA-2ic autoantigen specific B plasma cells is responsible for autoantibodies detect by the MBP-IA-2ic FP ELISA. This study is the first to demonstrate the generation of specific, genuine human derived anti-IA-2ic autoantibodies, thereby facilitating further investigation into the origin and functional significance of IA-2 autoantibodies in T1D.
Collapse
Affiliation(s)
- Thilini Mendis
- Department of Immunology, Allergy & Arthritis, Flinders Medical Centre and Flinders University, Bedford Park, 5042, South Australia, Australia
| | - Barbora Filipova
- Department of Immunology, Allergy & Arthritis, Flinders Medical Centre and Flinders University, Bedford Park, 5042, South Australia, Australia
| | - Jing Jing Wang
- Department of Immunology, Allergy & Arthritis, Flinders Medical Centre and Flinders University, Bedford Park, 5042, South Australia, Australia
| | - Massimo Pietropaolo
- Dept of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Michael W. Jackson
- Department of Immunology, Allergy & Arthritis, Flinders Medical Centre and Flinders University, Bedford Park, 5042, South Australia, Australia,Corresponding author. Department of Immunology, Allergy and Arthritis, Flinders University, GPO Box 2100, Adelaide, South Australia, 5001, Australia.
| |
Collapse
|
8
|
Tiberti C, D’Onofrio L, Panimolle F, Zampetti S, Maddaloni E, Buzzetti R. Immunoreactivities Against Different Tyrosine-Phosphatase 2 (IA-2)(256-760) Protein Domains Characterize Distinct Phenotypes in Subjects With LADA. Front Endocrinol (Lausanne) 2022; 13:921886. [PMID: 35813648 PMCID: PMC9263087 DOI: 10.3389/fendo.2022.921886] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 05/23/2022] [Indexed: 11/13/2022] Open
Abstract
Antibodies (Abs) against intracellular epitopes of the tyrosine-phosphatase 2 (IA-2) are detected in type 1 diabetes. Abs directed against the IA-2(256-760) portion, with both intra- and extracellular epitopes, are present in people with latent autoimmune diabetes in adults (LADA) and in obese subjects with normal glucose tolerance (NGT). We aim to characterize distribution and clinical features of intra- and extra-cellular IA-2(256-760) immunoreactivities in people with LADA compared to obese people with NGT. The intracellular immunoreactivity represented by immune response against two intracellular IA-2 constructs (IA-2JM(601-630) and IA-2IC(605-979)) was analyzed and related to clinical and biochemical features in 101 people with LADA and in 20 NGT obese subjects, all testing positive for IA-2(256-760) Abs. IA-2 intracellular immunoreactivity showed a frequency of 40.6% in LADA while it was not detected among NGT obese (p<0.001). Amongst LADA, the presence of immunoreactivity against the IA-2 intracellular domains was associated with lower BMI, waist circumference, higher HDL cholesterol and lower triglycerides, lower prevalence of hypertension and higher prevalence of other autoimmune disorders. Immunoreactivity against IA-2 does not involve intracellular domains in the majority of LADA and in obese people with NGT. This study shows that there is heterogeneity in the IA-2 epitopes, associated with different clinical features.
Collapse
|
9
|
Catriona C, Paolo P. SARS-CoV-2 induced post-translational protein modifications: A trigger for developing autoimmune diabetes? Diabetes Metab Res Rev 2022; 38:e3508. [PMID: 34990520 PMCID: PMC9015335 DOI: 10.1002/dmrr.3508] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Emerging evidence indicates a bi-directional relationship between SARS-CoV-2 and diabetes. The possibility exists that SARS-CoV-2 could induce diabetes, but it is not yet clear whether this might be a fulminant-type diabetes, autoimmune diabetes, or a new-onset transient hyperglycaemia. This viewpoint discusses mechanisms by which SARS-CoV-2 might trigger type 1 diabetes mellitus (T1DM). Specifically, we looked at the role of post-translational protein modifications (PTMs) and the generation of neoepitopes as a potential mechanism in the induction of islet autoimmunity, and the pathways via which coronavirus infections might exacerbate the formation of PTMs and, in so doing, provoke the onset of T1DM.
Collapse
Affiliation(s)
| | - Pozzilli Paolo
- Department of MedicineUnit of Endocrinology and DiabetesCampus Bio‐Medico UniversityRomeItaly
- Blizard InstituteBarts and The London School of Medicine and DentistryUniversity of LondonLondonUK
| |
Collapse
|
10
|
So M, Speake C, Steck AK, Lundgren M, Colman PG, Palmer JP, Herold KC, Greenbaum CJ. Advances in Type 1 Diabetes Prediction Using Islet Autoantibodies: Beyond a Simple Count. Endocr Rev 2021; 42:584-604. [PMID: 33881515 DOI: 10.1210/endrev/bnab013] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Indexed: 02/06/2023]
Abstract
Islet autoantibodies are key markers for the diagnosis of type 1 diabetes. Since their discovery, they have also been recognized for their potential to identify at-risk individuals prior to symptoms. To date, risk prediction using autoantibodies has been based on autoantibody number; it has been robustly shown that nearly all multiple-autoantibody-positive individuals will progress to clinical disease. However, longitudinal studies have demonstrated that the rate of progression among multiple-autoantibody-positive individuals is highly heterogenous. Accurate prediction of the most rapidly progressing individuals is crucial for efficient and informative clinical trials and for identification of candidates most likely to benefit from disease modification. This is increasingly relevant with the recent success in delaying clinical disease in presymptomatic subjects using immunotherapy, and as the field moves toward population-based screening. There have been many studies investigating islet autoantibody characteristics for their predictive potential, beyond a simple categorical count. Predictive features that have emerged include molecular specifics, such as epitope targets and affinity; longitudinal patterns, such as changes in titer and autoantibody reversion; and sequence-dependent risk profiles specific to the autoantibody and the subject's age. These insights are the outworking of decades of prospective cohort studies and international assay standardization efforts and will contribute to the granularity needed for more sensitive and specific preclinical staging. The aim of this review is to identify the dynamic and nuanced manifestations of autoantibodies in type 1 diabetes, and to highlight how these autoantibody features have the potential to improve study design of trials aiming to predict and prevent disease.
Collapse
Affiliation(s)
- Michelle So
- Diabetes Clinical Research Program, and Center for Interventional Immunology, Benaroya Research Institute at Virginia Mason, Seattle, WA 98101, USA
| | - Cate Speake
- Diabetes Clinical Research Program, and Center for Interventional Immunology, Benaroya Research Institute at Virginia Mason, Seattle, WA 98101, USA
| | - Andrea K Steck
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Markus Lundgren
- Department of Clinical Sciences Malmö, Lund University, Malmö 22200, Sweden
| | - Peter G Colman
- Department of Diabetes and Endocrinology, Royal Melbourne Hospital, Melbourne, Victoria 3050, Australia
| | - Jerry P Palmer
- VA Puget Sound Health Care System, Department of Medicine, University of Washington, Seattle, WA 98108, USA
| | - Kevan C Herold
- Department of Immunobiology, and Department of Internal Medicine, Yale University, New Haven, CT 06520, USA
| | - Carla J Greenbaum
- Diabetes Clinical Research Program, and Center for Interventional Immunology, Benaroya Research Institute at Virginia Mason, Seattle, WA 98101, USA
| |
Collapse
|
11
|
Ning Z, Song Z, Wang C, Peng S, Wan X, Liu Z, Lu A. How Perturbated Metabolites in Diabetes Mellitus Affect the Pathogenesis of Hypertension? Front Physiol 2021; 12:705588. [PMID: 34483960 PMCID: PMC8416465 DOI: 10.3389/fphys.2021.705588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 07/26/2021] [Indexed: 11/17/2022] Open
Abstract
The presence of hypertension (HTN) in type 2 diabetes mellitus (DM) is a common phenomenon in more than half of the diabetic patients. Since HTN constitutes a predictor of vascular complications and cardiovascular disease in type 2 DM patients, it is of significance to understand the molecular and cellular mechanisms of type 2 DM binding to HTN. This review attempts to understand the mechanism via the perspective of the metabolites. It reviewed the metabolic perturbations, the biological function of perturbated metabolites in two diseases, and the mechanism underlying metabolic perturbation that contributed to the connection of type 2 DM and HTN. DM-associated metabolic perturbations may be involved in the pathogenesis of HTN potentially in insulin, angiotensin II, sympathetic nervous system, and the energy reprogramming to address how perturbated metabolites in type 2 DM affect the pathogenesis of HTN. The recent integration of the metabolism field with microbiology and immunology may provide a wider perspective. Metabolism affects immune function and supports immune cell differentiation by the switch of energy. The diverse metabolites produced by bacteria modified the biological process in the inflammatory response of chronic metabolic diseases either. The rapidly evolving metabolomics has enabled to have a better understanding of the process of diseases, which is an important tool for providing some insight into the investigation of diseases mechanism. Metabolites served as direct modulators of biological processes were believed to assess the pathological mechanisms involved in diseases.
Collapse
Affiliation(s)
- Zhangchi Ning
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhiqian Song
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Chun Wang
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shitao Peng
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaoying Wan
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhenli Liu
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Aiping Lu
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| |
Collapse
|
12
|
Gu Y, Merriman C, Guo Z, Jia X, Wenzlau J, Li H, Li H, Rewers M, Yu L, Fu D. Novel autoantibodies to the β-cell surface epitopes of ZnT8 in patients progressing to type-1 diabetes. J Autoimmun 2021; 122:102677. [PMID: 34130115 PMCID: PMC9029399 DOI: 10.1016/j.jaut.2021.102677] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/26/2021] [Accepted: 05/29/2021] [Indexed: 11/22/2022]
Abstract
Type 1 diabetes (T1D) is a chronic autoimmune disease characterized by autoimmune destruction of insulin-producing β-cells in pancreatic islets. Seroconversions to islet autoantibodies (IAbs) precede the disease onset by many years, but the role of humoral autoimmunity in the disease initiation and progression are unclear. In the present study, we identified a new IAb directed to the extracellular epitopes of ZnT8 (ZnT8ec) in newly diagnosed patients with T1D, and demonstrated immunofluorescence staining of the surface of human β-cells by autoantibodies to ZnT8ec (ZnT8ecA). With the assay specificity set on 99th percentile of 336 healthy controls, the ZnT8ecA positivity rate was 23.6% (74/313) in patients with T1D. Moreover, 30 children in a longitudinal follow up of clinical T1D development were selected for sequential expression of four major IAbs (IAA, GADA, IA-2A and ZnT8icA). Among them, 10 children were ZnT8ecA positive. Remarkably, ZnT8ecA was the earliest IAb to appear in all 10 children. The identification of ZnT8ec as a cell surface target of humoral autoimmunity in the earliest phase of IAb responses opens a new avenue of investigation into the role of IAbs in the development of β-cell autoimmunity.
Collapse
Affiliation(s)
- Yong Gu
- Barbara Davis Center for Diabetes University of Colorado School of Medicine, Aurora, CO, USA
| | - Chengfeng Merriman
- Department of Physiology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Zheng Guo
- Department of Physiology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Xiaofan Jia
- Barbara Davis Center for Diabetes University of Colorado School of Medicine, Aurora, CO, USA
| | - Janet Wenzlau
- Barbara Davis Center for Diabetes University of Colorado School of Medicine, Aurora, CO, USA
| | - Hua Li
- Department of Structural Biology, Van Andel Institute, Grand Rapids, MI, USA
| | - Huilin Li
- Department of Structural Biology, Van Andel Institute, Grand Rapids, MI, USA
| | - Marian Rewers
- Barbara Davis Center for Diabetes University of Colorado School of Medicine, Aurora, CO, USA
| | - Liping Yu
- Barbara Davis Center for Diabetes University of Colorado School of Medicine, Aurora, CO, USA.
| | - Dax Fu
- Department of Physiology, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
13
|
T-cell responses to hybrid insulin peptides prior to type 1 diabetes development. Proc Natl Acad Sci U S A 2021; 118:2019129118. [PMID: 33542101 DOI: 10.1073/pnas.2019129118] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
T-cell responses to posttranslationally modified self-antigens are associated with many autoimmune disorders. In type 1 diabetes, hybrid insulin peptides (HIPs) are implicated in the T-cell-mediated destruction of insulin-producing β-cells within pancreatic islets. The natural history of the disease is such that it allows for the study of T-cell reactivity prior to the onset of clinical symptoms. We hypothesized that CD4 T-cell responses to posttranslationally modified islet peptides precedes diabetes onset. In a cohort of genetically at-risk individuals, we measured longitudinal T-cell responses to native insulin and hybrid insulin peptides. Both proinflammatory (interferon-γ) and antiinflammatory (interluekin-10) cytokine responses to HIPs were more robust than those to native peptides, and the ratio of such responses oscillated between pro- and antiinflammatory over time. However, individuals who developed islet autoantibodies or progressed to clinical type 1 diabetes had predominantly inflammatory T-cell responses to HIPs. Additionally, several HIP T-cell responses correlated to worsening measurements of blood glucose, highlighting the relevance of T-cell responses to posttranslationally modified peptides prior to autoimmune disease development.
Collapse
|
14
|
Buzzetti R, Tuomi T, Mauricio D, Pietropaolo M, Zhou Z, Pozzilli P, Leslie RD. Management of Latent Autoimmune Diabetes in Adults: A Consensus Statement From an International Expert Panel. Diabetes 2020; 69:2037-2047. [PMID: 32847960 PMCID: PMC7809717 DOI: 10.2337/dbi20-0017] [Citation(s) in RCA: 126] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 07/09/2020] [Indexed: 02/07/2023]
Abstract
A substantial proportion of patients with adult-onset diabetes share features of both type 1 diabetes (T1D) and type 2 diabetes (T2D). These individuals, at diagnosis, clinically resemble T2D patients by not requiring insulin treatment, yet they have immunogenetic markers associated with T1D. Such a slowly evolving form of autoimmune diabetes, described as latent autoimmune diabetes of adults (LADA), accounts for 2-12% of all patients with adult-onset diabetes, though they show considerable variability according to their demographics and mode of ascertainment. While therapeutic strategies aim for metabolic control and preservation of residual insulin secretory capacity, endotype heterogeneity within LADA implies a personalized approach to treatment. Faced with a paucity of large-scale clinical trials in LADA, an expert panel reviewed data and delineated one therapeutic approach. Building on the 2020 American Diabetes Association (ADA)/European Association for the Study of Diabetes (EASD) consensus for T2D and heterogeneity within autoimmune diabetes, we propose "deviations" for LADA from those guidelines. Within LADA, C-peptide values, proxy for β-cell function, drive therapeutic decisions. Three broad categories of random C-peptide levels were introduced by the panel: 1) C-peptide levels <0.3 nmol/L: a multiple-insulin regimen recommended as for T1D; 2) C-peptide values ≥0.3 and ≤0.7 nmol/L: defined by the panel as a "gray area" in which a modified ADA/EASD algorithm for T2D is recommended; consider insulin in combination with other therapies to modulate β-cell failure and limit diabetic complications; 3) C-peptide values >0.7 nmol/L: suggests a modified ADA/EASD algorithm as for T2D but allowing for the potentially progressive nature of LADA by monitoring C-peptide to adjust treatment. The panel concluded by advising general screening for LADA in newly diagnosed non-insulin-requiring diabetes and, importantly, that large randomized clinical trials are warranted.
Collapse
Affiliation(s)
- Raffaella Buzzetti
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Tiinamaija Tuomi
- Division of Endocrinology, Abdominal Center, Helsinki University Hospital, Institute for Molecular Medicine Finland FIMM and Research Program for Clinical and Molecular Metabolism, University of Helsinki, and Folkhälsan Research Center, Helsinki, Finland
- Lund University Diabetes Center, University of Lund, Malmo, Sweden
| | - Didac Mauricio
- Department of Endocrinology & Nutrition, CIBERDEM, Hospital de la Santa Creu i Sant Pau & Institut d'Investigació Biomèdica Sant Pau (IIB Sant Pau), Autonomous University of Barcelona, Barcelona, Spain
| | - Massimo Pietropaolo
- Division of Endocrinology, Diabetes and Metabolism, Diabetes Research Center, Baylor College of Medicine, Houston, TX
| | - Zhiguang Zhou
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University and Key Laboratory of Diabetes Immunology, Central South University, Ministry of Education, National Clinical Research Center for Metabolic Diseases, Changsha, Hunan, China
| | - Paolo Pozzilli
- Unit of Endocrinology and Diabetes, Department of Medicine, Campus Bio-Medico University, Rome, Italy
- Blizard Institute, Barts and The London School of Medicine and Dentistry, University of London, London, U.K
| | - Richard David Leslie
- Blizard Institute, Barts and The London School of Medicine and Dentistry, University of London, London, U.K.
| |
Collapse
|
15
|
Simmons KM, Mitchell AM, Alkanani AA, McDaniel KA, Baschal EE, Armstrong T, Pyle L, Yu L, Michels AW. Failed Genetic Protection: Type 1 Diabetes in the Presence of HLA-DQB1*06:02. Diabetes 2020; 69:1763-1769. [PMID: 32439825 PMCID: PMC7372070 DOI: 10.2337/db20-0038] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 05/13/2020] [Indexed: 12/16/2022]
Abstract
Certain HLA class II genes increase the risk for type 1 diabetes (T1D) development while others provide protection from disease development. HLA class II alleles encode MHC proteins on antigen-presenting cells, which function to present peptides and activate CD4 T cells. The DRB1*15:01 (DR15)-DQA1*01:02-DQB1*06:02 (DQ6) haplotype provides dominant protection across all stages of T1D and is a common haplotype found in Caucasians. However, it is present in <1% of people with T1D. Knowing which metabolic, immunologic, and genetic features are unique to individuals who fail genetic protection and develop T1D is important for defining the underlying mechanisms of DQB1*06:02-mediated protection. We describe a T1D cohort with DQB1*06:02 (n = 50) and compare them to individuals with T1D and without DQB1*06:02 (n = 2,759) who were identified over the last 26 years at the Barbara Davis Center for Diabetes. The age at diagnosis was similar between the cohorts and normally distributed throughout childhood and early adulthood. The average hemoglobin A1c was 10.8 ± 2.8% (95 ± 7 mmol/mol) at diagnosis in those DQB1*06:02 positive. The majority of T1D DQB1*06:02 + individuals were positive for one or more islet autoantibodies; however, there was a greater proportion who were islet autoantibody negative compared with those T1D DQB1*06:02 - individuals. Interestingly, DQB1*03:02, which confers significant T1D risk, was present in only those DQB1*06:02 + individuals with islet autoantibodies. This is one of the largest studies examining patients presenting with clinical T1D in the presence of DQB1*06:02, which provides a population to study the mechanisms of failed genetic protection against T1D.
Collapse
Affiliation(s)
- Kimber M Simmons
- Barbara Davis Center for Diabetes, University of Colorado, Aurora, CO
| | - Angela M Mitchell
- Barbara Davis Center for Diabetes, University of Colorado, Aurora, CO
| | - Aimon A Alkanani
- Barbara Davis Center for Diabetes, University of Colorado, Aurora, CO
| | | | - Erin E Baschal
- Barbara Davis Center for Diabetes, University of Colorado, Aurora, CO
| | - Taylor Armstrong
- Barbara Davis Center for Diabetes, University of Colorado, Aurora, CO
| | - Laura Pyle
- Barbara Davis Center for Diabetes, University of Colorado, Aurora, CO
| | - Liping Yu
- Barbara Davis Center for Diabetes, University of Colorado, Aurora, CO
| | - Aaron W Michels
- Barbara Davis Center for Diabetes, University of Colorado, Aurora, CO
| |
Collapse
|
16
|
Maddaloni E, Moretti C, Mignogna C, Buzzetti R. Adult-onset autoimmune diabetes in 2020: An update. Maturitas 2020; 137:37-44. [DOI: 10.1016/j.maturitas.2020.04.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 04/18/2020] [Accepted: 04/21/2020] [Indexed: 12/11/2022]
|
17
|
Balakrishnan S, Kumar P, Prabhakar BS. Post-translational modifications contribute to neoepitopes in Type-1 diabetes: Challenges for inducing antigen-specific tolerance. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2020; 1868:140478. [PMID: 32599298 DOI: 10.1016/j.bbapap.2020.140478] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 05/20/2020] [Accepted: 06/19/2020] [Indexed: 12/11/2022]
Abstract
Type-1 Diabetes (T1D) is the major autoimmune disease affecting the juvenile population in which insulin-producing pancreatic β-cells are destroyed by self-reactive T-cells and B-cells. Emerging studies have identified the presence of autoantibodies and altered T-cell reactivity against several autoantigens in individuals who are at risk of developing T1D even before the clinical onset of diabetes. Whilst these findings could lead to the development of predictive biomarkers for early diagnosis, growing evidence on the generation of neoepitopes, epitope spreading and diverse antigen repertoire in T1D poses a major challenge for developing approaches to induce antigen-specific tolerance. Mechanisms of neoepitope generation include post-translational modifications of existing epitopes, aberrant translational products, peptide fusion, and differences in MHC binding registers. Here, we focus our discussion on how post-translational modifications can give rise to immunogenic neoepitopes in T1D and present our perspective on how it could affect the development of therapeutic approaches to induce antigen-specific tolerance.
Collapse
Affiliation(s)
- Sivasangari Balakrishnan
- Department of Microbiology and Immunology, University of Illinois-College of Medicine, Chicago, IL, United States of America.
| | - Prabhakaran Kumar
- Department of Microbiology and Immunology, University of Illinois-College of Medicine, Chicago, IL, United States of America.
| | - Bellur S Prabhakar
- Department of Microbiology and Immunology, University of Illinois-College of Medicine, Chicago, IL, United States of America.
| |
Collapse
|
18
|
Human Leukocyte Antigen (HLA) and Islet Autoantibodies Are Tools to Characterize Type 1 Diabetes in Arab Countries: Emphasis on Kuwait. DISEASE MARKERS 2019; 2019:9786078. [PMID: 31827651 PMCID: PMC6886320 DOI: 10.1155/2019/9786078] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 07/15/2019] [Accepted: 09/20/2019] [Indexed: 12/11/2022]
Abstract
The incidence rate of type 1 diabetes in Kuwait had been increasing exponentially and has doubled in children ≤ 14 years old within almost two decades. Therefore, there is a dire need for a careful systematic familial cohort study. Several immunogenetic factors affect the pathogenesis of the disease. The human leukocyte antigen (HLA) accounts for the major genetic susceptibility to the disease. The triggering agents initiate disease onset by type 1 destruction of pancreatic β-cells. Both HLA and anti-islet antibodies can be used to characterize, predict susceptibility to the disease, innovate, or delay the β-cell destruction. Evidence from prospective longitudinal studies suggested that the underlying disease process represents a continuum that begins before the symptoms are clinically evident. Autoimmunity of the functional pancreatic β-cells results in symptomatic type 1 diabetes and lifelong insulin dependence. The autoantibodies against glutamic acid decarboxylase (GADA), insulinoma antigen-2 (IA-2A), insulin (IAA), and zinc transporter-8 (ZnT-8A) comprise the most reliable biomarkers for type 1 diabetes in both children and adults. Although Kuwait is the second among the top 10 countries with a high incidence rate of type 1 diabetes, there have been no proper diagnostic and prediction tools as per the World Health Organization. The Kuwaiti Type 1 Diabetes Study (KADS) was initiated to understand the disease pathogenesis as well as the HLA and anti-islet autoantibody profile of type 1 diabetes in Kuwait. Understanding the disease sequela in a homogenous gene pool and highly consanguineous population of Kuwaitis could help solve the challenges and pathogenesis, as well as hasten the prevention, of type 1 diabetes.
Collapse
|
19
|
Acevedo-Calado MJ, Pietropaolo SL, Morran MP, Schnell S, Vonberg AD, Verge CF, Gianani R, Becker DJ, Huang S, Greenbaum CJ, Yu L, Davidson HW, Michels AW, Rich SS, Pietropaolo M. Autoantibodies Directed Toward a Novel IA-2 Variant Protein Enhance Prediction of Type 1 Diabetes. Diabetes 2019; 68:1819-1829. [PMID: 31167877 PMCID: PMC6702638 DOI: 10.2337/db18-1351] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Accepted: 05/30/2019] [Indexed: 02/06/2023]
Abstract
We identified autoantibodies (AAb) reacting with a variant IA-2 molecule (IA-2var) that has three amino acid substitutions (Cys27, Gly608, and Pro671) within the full-length molecule. We examined IA-2var AAb in first-degree relatives of type 1 diabetes (T1D) probands from the TrialNet Pathway to Prevention Study. The presence of IA-2var-specific AAb in relatives was associated with accelerated progression to T1D in those positive for AAb to GAD65 and/or insulin but negative in the standard test for IA-2 AAb. Furthermore, relatives with single islet AAb (by traditional assays) and carrying both IA-2var AAb and the high-risk HLA-DRB1*04-DQB1*03:02 haplotype progress rapidly to onset of T1D. Molecular modeling of IA-2var predicts that the genomic variation that alters the three amino acids induces changes in the three-dimensional structure of the molecule, which may lead to epitope unmasking in the IA-2 extracellular domain. Our observations suggest that the presence of AAb to IA-2var would identify high-risk subjects who would benefit from participation in prevention trials who have one islet antibody by traditional testing and otherwise would be misclassified as "low risk" relatives.
Collapse
Affiliation(s)
- Maria J. Acevedo-Calado
- Diabetes Research Center, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX
| | - Susan L. Pietropaolo
- Diabetes Research Center, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX
| | - Michael P. Morran
- Department of Surgery, College of Medicine, University of Toledo, Toledo, OH
| | - Santiago Schnell
- Department of Molecular & Integrative Physiology and Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI
| | - Andrew D. Vonberg
- Diabetes Research Center, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX
| | - Charles F. Verge
- School of Women’s and Children’s Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Roberto Gianani
- Diabetes Research Center, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX
| | - Dorothy J. Becker
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Shuai Huang
- Department of Industrial & Systems Engineering, University of Washington, Seattle, WA
| | | | - Liping Yu
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, CO
| | - Howard W. Davidson
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, CO
| | - Aaron W. Michels
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, CO
| | - Stephen S. Rich
- Center for Public Health Genomics, Department of Public Health Sciences, University of Virginia, Charlottesville, VA
| | - Massimo Pietropaolo
- Diabetes Research Center, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX
| |
Collapse
|
20
|
Mulukutla SN, Acevedo-Calado M, Hampe CS, Pietropaolo M, Balasubramanyam A. Response to Comment on Mulukutla et al. Autoantibodies to the IA-2 Extracellular Domain Refine the Definition of "A+" Subtypes of Ketosis-Prone Diabetes. Diabetes Care 2018;41:2637-2640. Diabetes Care 2019; 42:e82-e83. [PMID: 31010950 PMCID: PMC6489110 DOI: 10.2337/dci19-0007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Surya N Mulukutla
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX
| | - Maria Acevedo-Calado
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX
| | - Christiane S Hampe
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA
| | - Massimo Pietropaolo
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX
| | - Ashok Balasubramanyam
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX
| |
Collapse
|
21
|
Purcell AW, Sechi S, DiLorenzo TP. The Evolving Landscape of Autoantigen Discovery and Characterization in Type 1 Diabetes. Diabetes 2019; 68:879-886. [PMID: 31010879 PMCID: PMC6477901 DOI: 10.2337/dbi18-0066] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 01/29/2019] [Indexed: 12/20/2022]
Abstract
Type 1 diabetes (T1D) is an autoimmune disease that is caused, in part, by T cell-mediated destruction of insulin-producing β-cells. High risk for disease, in those with genetic susceptibility, is predicted by the presence of two or more autoantibodies against insulin, the 65-kDa form of glutamic acid decarboxylase (GAD65), insulinoma-associated protein 2 (IA-2), and zinc transporter 8 (ZnT8). Despite this knowledge, we still do not know what leads to the breakdown of tolerance to these autoantigens, and we have an incomplete understanding of T1D etiology and pathophysiology. Several new autoantibodies have recently been discovered using innovative technologies, but neither their potential utility in monitoring disease development and treatment nor their role in the pathophysiology and etiology of T1D has been explored. Moreover, neoantigen generation (through posttranslational modification, the formation of hybrid peptides containing two distinct regions of an antigen or antigens, alternative open reading frame usage, and translation of RNA splicing variants) has been reported, and autoreactive T cells that target these neoantigens have been identified. Collectively, these new studies provide a conceptual framework to understand the breakdown of self-tolerance, if such modifications occur in a tissue- or disease-specific context. A recent workshop sponsored by the National Institute of Diabetes and Digestive and Kidney Diseases brought together investigators who are using new methods and technologies to identify autoantigens and characterize immune responses toward these proteins. Researchers with diverse expertise shared ideas and identified resources to accelerate antigen discovery and the detection of autoimmune responses in T1D. The application of this knowledge will direct strategies for the identification of improved biomarkers for disease progression and treatment response monitoring and, ultimately, will form the foundation for novel antigen-specific therapeutics. This Perspective highlights the key issues that were addressed at the workshop and identifies areas for future investigation.
Collapse
Affiliation(s)
- Anthony W Purcell
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Salvatore Sechi
- Division of Diabetes, Endocrinology, and Metabolic Diseases, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD
| | - Teresa P DiLorenzo
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY
- Division of Endocrinology, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY
- Einstein-Mount Sinai Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY
- Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, NY
| |
Collapse
|
22
|
Heneberg P. Comment on Mulukutla et al. Autoantibodies to the IA-2 Extracellular Domain Refine the Definition of "A+" Subtypes of Ketosis-Prone Diabetes. Diabetes Care 2018;41:2637-2640. Diabetes Care 2019; 42:e81. [PMID: 31010949 DOI: 10.2337/dc19-0022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Petr Heneberg
- 2nd Department of Internal Medicine, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| |
Collapse
|
23
|
Mulukutla SN, Acevedo-Calado M, Hampe CS, Pietropaolo M, Balasubramanyam A. Autoantibodies to the IA-2 Extracellular Domain Refine the Definition of "A+" Subtypes of Ketosis-Prone Diabetes. Diabetes Care 2018; 41:2637-2640. [PMID: 30327357 PMCID: PMC6245211 DOI: 10.2337/dc18-0613] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 09/10/2018] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Autoantibodies directed against tyrosine phosphatase IA-2 antibody (IA-2 Ab) are diagnostic for autoimmune type 1 diabetes. Conventional assays target the intracellular domain of IA-2. Among patients with ketosis-prone diabetes (KPD), characterized by presentation with diabetic ketoacidosis (DKA), >60% of adults lack three classic islet autoantibodies-IA-2, GAD65, and ZnT8 Abs-associated with type 1 diabetes. We aimed to determine whether apparently autoantibody-negative ("A-") KPD patients possess occult IA-2 Ab directed against full-length IA-2 (IA-2FL) or its extracellular domain (IA-2EC). RESEARCH DESIGN AND METHODS We developed an assay that targets IA-2FL and IA-2EC and used it to analyze 288 subjects with A- KPD. RESULTS Ten A- KPD patients were positive for IA-2EC Ab (3.5%), and three were also positive for IA-2FL Ab (1.0%), similar to frequencies in type 1 and type 2 diabetes. CONCLUSIONS Measurement of IA-2FL Ab and IA-2EC Ab improves the accuracy of the Aβ classification of KPD patients.
Collapse
Affiliation(s)
- Surya N Mulukutla
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX
| | - Maria Acevedo-Calado
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX
| | - Christiane S Hampe
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA
| | - Massimo Pietropaolo
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX
| | - Ashok Balasubramanyam
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX
| |
Collapse
|
24
|
Marre ML, McGinty JW, Chow IT, DeNicola ME, Beck NW, Kent SC, Powers AC, Bottino R, Harlan DM, Greenbaum CJ, Kwok WW, Piganelli JD, James EA. Modifying Enzymes Are Elicited by ER Stress, Generating Epitopes That Are Selectively Recognized by CD4 + T Cells in Patients With Type 1 Diabetes. Diabetes 2018; 67:1356-1368. [PMID: 29654212 PMCID: PMC6014552 DOI: 10.2337/db17-1166] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 04/04/2018] [Indexed: 12/18/2022]
Abstract
In spite of tolerance mechanisms, some individuals develop T-cell-mediated autoimmunity. Posttranslational modifications that increase the affinity of epitope presentation and/or recognition represent one means through which self-tolerance mechanisms can be circumvented. We investigated T-cell recognition of peptides that correspond to modified β-cell antigens in subjects with type 1 diabetes. Modified peptides elicited enhanced proliferation by autoreactive T-cell clones. Endoplasmic reticulum (ER) stress in insulinoma cells increased cytosolic calcium and the activity of tissue transglutaminase 2 (tTG2). Furthermore, stressed human islets and insulinomas elicited effector responses from T cells specific for modified peptides, suggesting that ER stress-derived tTG2 activity generated deamidated neoepitopes that autoreactive T cells recognized. Patients with type 1 diabetes had large numbers of T cells specific for these epitopes in their peripheral blood. T cells with these specificities were also isolated from the pancreatic draining lymph nodes of cadaveric donors with established diabetes. Together, these results suggest that self-antigens are enzymatically modified in β-cells during ER stress, giving rise to modified epitopes that could serve to initiate autoimmunity or to further broaden the antigenic repertoire, activating potentially pathogenic CD4+ T cells that may not be effectively eliminated by negative selection.
Collapse
Affiliation(s)
- Meghan L Marre
- Division of Pediatric Surgery, Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - John W McGinty
- Benaroya Research Institute at Virginia Mason, Seattle, WA
| | - I-Ting Chow
- Benaroya Research Institute at Virginia Mason, Seattle, WA
| | - Megan E DeNicola
- Department of Medicine, Division of Diabetes, Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, MA
| | - Noah W Beck
- Division of Pediatric Surgery, Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Sally C Kent
- Department of Medicine, Division of Diabetes, Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, MA
| | - Alvin C Powers
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, and Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, TN
- VA Tennessee Valley Healthcare System, Nashville, TN
| | - Rita Bottino
- Islet Isolation Laboratory, Institute of Cellular Therapeutics, Allegheny Health Network, Pittsburgh, PA
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA
| | - David M Harlan
- Department of Medicine, Division of Diabetes, Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, MA
| | - Carla J Greenbaum
- Benaroya Research Institute at Virginia Mason, Seattle, WA
- Department of Medicine, University of Washington, Seattle, WA
| | - William W Kwok
- Benaroya Research Institute at Virginia Mason, Seattle, WA
- Department of Medicine, University of Washington, Seattle, WA
| | - Jon D Piganelli
- Division of Pediatric Surgery, Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Eddie A James
- Benaroya Research Institute at Virginia Mason, Seattle, WA
| |
Collapse
|
25
|
James EA, Pietropaolo M, Mamula MJ. Immune Recognition of β-Cells: Neoepitopes as Key Players in the Loss of Tolerance. Diabetes 2018; 67:1035-1042. [PMID: 29784651 PMCID: PMC5961411 DOI: 10.2337/dbi17-0030] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 02/27/2018] [Indexed: 12/13/2022]
Abstract
Prior to the onset of type 1 diabetes, there is progressive loss of immune self-tolerance, evidenced by the accumulation of islet autoantibodies and emergence of autoreactive T cells. Continued autoimmune activity leads to the destruction of pancreatic β-cells and loss of insulin secretion. Studies of samples from patients with type 1 diabetes and of murine disease models have generated important insights about genetic and environmental factors that contribute to susceptibility and immune pathways that are important for pathogenesis. However, important unanswered questions remain regarding the events that surround the initial loss of tolerance and subsequent failure of regulatory mechanisms to arrest autoimmunity and preserve functional β-cells. In this Perspective, we discuss various processes that lead to the generation of neoepitopes in pancreatic β-cells, their recognition by autoreactive T cells and antibodies, and potential roles for such responses in the pathology of disease. Emerging evidence supports the relevance of neoepitopes generated through processes that are mechanistically linked with β-cell stress. Together, these observations support a paradigm in which neoepitope generation leads to the activation of pathogenic immune cells that initiate a feed-forward loop that can amplify the antigenic repertoire toward pancreatic β-cell proteins.
Collapse
Affiliation(s)
- Eddie A James
- Translational Research Program, Benaroya Research Institute at Virginia Mason, Seattle, WA
| | - Massimo Pietropaolo
- Diabetes Research Center, Department of Medicine, Baylor College of Medicine, Houston, TX
| | - Mark J Mamula
- Section of Rheumatology, Department of Medicine, Yale School of Medicine, New Haven, CT
| |
Collapse
|
26
|
Sanda S. Increasing ICA512 autoantibody titers predict development of abnormal oral glucose tolerance tests. Pediatr Diabetes 2018; 19:271-276. [PMID: 28707353 PMCID: PMC5849060 DOI: 10.1111/pedi.12542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 04/20/2017] [Accepted: 04/25/2017] [Indexed: 10/19/2022] Open
Abstract
OBJECTIVE Determine if autoantibody titer magnitude and variability predict glucose abnormalities in subjects at risk for type 1 diabetes. RESEARCH DESIGNS AND METHODS Demographic information, longitudinal autoantibody titers, and oral glucose tolerance test (OGTT) data were obtained from the TrialNet Pathway to Prevention study. Subjects (first and second degree relatives of individuals with type 1 diabetes) with at least 2 diabetes autoantibodies were selected for analysis. Autoantibody titer means were calculated for each subject for the duration of study participation and the relationship between titer tertiles and glucose value tertiles from OGTTs (normal, impaired, and diabetes) was assessed with a proportional odds ordinal regression model. A matched pairs analysis was used to examine the relationship between changes in individual autoantibody titers and 120-minute glucose values. Titer variability was quantified using cumulative titer standard deviations. RESULTS We studied 778 subjects recruited in the TrialNet Pathway to Prevention study between 2006 and 2014. Increased cumulative mean titer values for both ICA512 and GAD65 (estimated increase in proportional odds = 1.61, 95% CI = 1.39, 1.87, P < 1 × 10-9 and 1.17, 95% CI = 1.03, 1.32, P = .016, respectively) were associated with peak 120-minute glucose values. While fluctuating titer levels were observed in some subjects, no significant relationship between titer standard deviation and glucose values was observed. CONCLUSION ICA512 autoantibody titers associate with progressive abnormalities in glucose metabolism in subjects at risk for type 1 diabetes. Fluctuations in autoantibody titers do not correlate with lower rates of progression to clinical disease.
Collapse
Affiliation(s)
- Srinath Sanda
- UC San Francisco Diabetes Center, UC San Francisco School of Medicine
| | | |
Collapse
|
27
|
Li R, Huang J, Yu Y, Yang Y. Islet Autoantibody Patterns in Patients With Type 2 Diabetes Aged 60 and Higher: A Cross-Sectional Study in a Chinese Hospital. Front Endocrinol (Lausanne) 2018; 9:260. [PMID: 29887833 PMCID: PMC5980972 DOI: 10.3389/fendo.2018.00260] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Accepted: 05/07/2018] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Some elderly citizens with a clinical diagnosis of type 2 diabetes had evidence of positive islet autoantibodies. We aimed to discover their islet autoantibody patterns and independent correlative factors that might lead to a better understanding of significance of islet autoimmunity in the progression of elderly diabetes. METHODS A total of 541 inpatients of clinically diagnosed type 2 diabetes aged 60 and over were recruited. Three islet autoantibodies including insulin autoantibody (IAA), islet cell antibody (ICA), and glutamic acid decarboxylase antibody (GADA) as well as clinical and biochemical characteristics were tested and collected in Huashan Hospital. Associations between these antibodies and clinical features were analyzed by Spearman correlation and binary logistic analyses. RESULTS In our current study, total positive rate of islet autoantibodies (IAA, ICA, and GADA) was 35.67% with 26.62% for individual IAA, 5.55% for ICA, and 5.91% for GADA, in elderly with type 2 diabetes. None of combinations of such autoantibodies were observed, with the exception of IAA + ICA (0.74%, n = 4), IAA + GADA (1.48%, n = 8), and ICA + GADA (0.18%, n = 1). Compared with GADA negative group, patients in positive group tended to have lower level of fasting and postprandial C peptide, fasting blood glucose (FBG), and body mass index (BMI). After adjusted for the BMI, FBG, and postprandial C peptide, fasting C peptide seemed to be an independent factor related to GADA positivity (OR = 0.52, p = 0.02). As for patients with positive IAA, they were more likely to have insulin treatment with longer duration of diabetes, higher level of BMI, and lower level of postprandial C peptide. After adjusted for the duration of diabetes, BMI, and postprandial C peptide, insulin treatment was a significant predictor for IAA positivity (OR = 5.20, p < 0.0001). Furthermore, hs-CRP was positively related to ICA positivity, and hs-CRP appeared to be an independent indicator for ICA (OR = 3.43, p = 0.008). CONCLUSION In elderly with type 2 diabetes, high prevalence rate of IAA was frequently accompanied with insulin treatment, while ICA and GADA were more closely associated with the systemic inflammation and beta-cell failure, respectively.
Collapse
|
28
|
Affiliation(s)
- Eddie A James
- Benaroya Research Institute at Virginia Mason, Seattle, WA
| |
Collapse
|
29
|
Buzzetti R, Zampetti S, Maddaloni E. Adult-onset autoimmune diabetes: current knowledge and implications for management. Nat Rev Endocrinol 2017; 13:674-686. [PMID: 28885622 DOI: 10.1038/nrendo.2017.99] [Citation(s) in RCA: 165] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Adult-onset autoimmune diabetes is a heterogeneous disease that is characterized by a reduced genetic load, a less intensive autoimmune process and a mild metabolic decompensation at onset compared with young-onset type 1 diabetes mellitus (T1DM). The majority of patients with adult-onset autoimmune diabetes do not require insulin treatment for at least 6 months after diagnosis. Such patients are defined as having latent autoimmune diabetes in adults (LADA), which is distinct from classic adult-onset T1DM. The extensive heterogeneity of adult-onset autoimmune diabetes is apparent beyond the distinction between classic adult-onset T1DM and LADA. LADA is characterized by genetic, phenotypic and humoral heterogeneity, encompassing different degrees of insulin resistance and autoimmunity; this heterogeneity is probably a result of different pathological mechanisms, which have implications for treatment. The existence of heterogeneous phenotypes in LADA makes it difficult to establish an a priori treatment algorithm, and therefore, a personalized medicine approach is required. In this Review, we discuss the current understanding and gaps in knowledge regarding the pathophysiology and clinical features of adult-onset autoimmune diabetes and highlight the similarities and differences with classic T1DM and type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Raffaella Buzzetti
- Department of Experimental Medicine, Sapienza University, Viale Regina Elena 324, 00161, Rome, Italy
| | - Simona Zampetti
- Department of Experimental Medicine, Sapienza University, Viale Regina Elena 324, 00161, Rome, Italy
| | - Ernesto Maddaloni
- Department of Medicine, Unit of Endocrinology and Diabetes, University Campus Bio-Medico, Via Álvaro del Portillo 21, 00128, Rome, Italy
| |
Collapse
|