1
|
Hoffmann L, Kohls M, Arnolds S, Achenbach P, Bergholdt R, Bonifacio E, Bosi E, Gündert M, Hoefelschweiger BK, Hummel S, Jarosz-Chobot P, Kordonouri O, Lampasona V, Narendran P, Overbergh L, Pociot F, Raposo JF, Šumník Z, Szypowska A, Vercauteren J, Winkler C, Mathieu C, Ziegler AG. EDENT1FI Master Protocol for screening of presymptomatic early-stage type 1 diabetes in children and adolescents. BMJ Open 2025; 15:e088522. [PMID: 39753267 PMCID: PMC11749223 DOI: 10.1136/bmjopen-2024-088522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 12/11/2024] [Indexed: 01/23/2025] Open
Abstract
INTRODUCTION The identification of type 1 diabetes at an early presymptomatic stage has clinical benefits. These include a reduced risk of diabetic ketoacidosis (DKA) at the clinical manifestation of the disease and a significant reduction in clinical symptoms. The European action for the Diagnosis of Early Non-clinical Type 1 diabetes For disease Interception (EDENT1FI) represents a pioneering effort to advance early detection of type 1 diabetes through public health screening. With the EDENT1FI Master Protocol, the project aims to harmonise and standardise screening for early-stage type 1 diabetes and care. METHODS AND ANALYSIS Public health islet autoantibody screening is conducted in the Czech Republic, Denmark, Germany, Italy, Poland, Portugal, Sweden and the UK. Between November 2023 (start date) and October 2028 (planned end date), an estimated number of 200 000 children and adolescents aged 1-17 years are expected to be screened. Screening is performed in capillary blood, examining different islet autoantibodies (autoantibodies against insulin, glutamic acid decarboxylase-65, insulinoma-associated antigen-2 and/or zinc transporter-8). Positive screening results undergo confirmation through a second antibody method. A second (venous) blood sample is requested if at least two autoantibodies are detected, to confirm the autoantibody status. Children and adolescents with confirmed two or more autoantibodies are invited to metabolic staging (oral glucose tolerance test, haemoglobin A1c (HbA1c), random glucose, optionally continuous glucose monitoring); an educational programme and recommendations for monitoring are provided. The feasibility and acceptability of screening are evaluated by feedback questionnaires. Pseudonymised data is collated in the EDENT1FI Registry. Study outcomes include country-specific screening rates, prevalences of stage 1 and stage 2 type 1 diabetes, number in EDENT1FI Registry, proportion with DKA and symptoms at clinical diagnosis and median HbA1c. ETHICS AND DISSEMINATION Following the EDENT1FI Master Protocol, site-specific protocols are developed and approved by local ethics committees (Technical University of Munich, Medical Faculty, Nr. 70/14; Medizinische Hochschule Hannover, Nr. 9588_BO_S_2021; Technische Universität Dresden, Nr. BO-EK-356082020; Center for Sundhed Region Hovedstaden, Nr. H-22053116; Swedish Ethical Review Authority, Nr. 2023-00312-01; National Health Service Health Research Authority and Health Care Research Wales, IRAS (Integrated Research Application System) project ID 309252; Italian National Institute of Health, National ethics committee for clinical trials of public research bodies (EPR) and other national public institutions, Prot. PRE BIO CE Nr. 0059835; Charles University in Prague, Ethics Committee for Multi-Centric Clinical Trials of the University Hopital Motol and 2nd Faculty of Medicine, Nr. 1271/23; Bioethics Committee at the Medical University of Warsaw, Nr. 21/2024 and KB/6/R/2024; Associação Protectora dos Diabéticos de Portugal, Nr. 211/2024). Results are disseminated through peer-reviewed journals and conference presentations and will be shared openly.
Collapse
Affiliation(s)
- Luisa Hoffmann
- Institute of Diabetes Research, Helmholtz Munich German Research Center for Environmental Health, Munich, Germany
| | - Mirjam Kohls
- Institute of Diabetes Research, Helmholtz Munich German Research Center for Environmental Health, Munich, Germany
| | - Stefanie Arnolds
- Institute of Diabetes Research, Helmholtz Munich German Research Center for Environmental Health, Munich, Germany
| | - Peter Achenbach
- Institute of Diabetes Research, Helmholtz Munich German Research Center for Environmental Health, Munich, Germany
- Forschergruppe Diabetes at Klinikum Rechts der Isar, Technical University of Munich School of Medicine, Munich, Germany
| | | | - Ezio Bonifacio
- Center for Regenerative Therapies Dresden, Faculty of Medicine, TU Dresden, Dresden, Germany
| | - Emanuele Bosi
- Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Melanie Gündert
- Institute of Diabetes Research, Helmholtz Munich German Research Center for Environmental Health, Munich, Germany
| | - Bianca K Hoefelschweiger
- Institute of Diabetes Research, Helmholtz Munich German Research Center for Environmental Health, Munich, Germany
| | - Sandra Hummel
- Institute of Diabetes Research, Helmholtz Munich German Research Center for Environmental Health, Munich, Germany
| | - Przemysława Jarosz-Chobot
- Department of Children's Diabetology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | | | - Vito Lampasona
- Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | | | - Lut Overbergh
- Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Flemming Pociot
- Department of Clinical Research, Translational Type 1 Diabetes Research, Steno Diabetes Center Copenhagen, Herlev, Denmark
| | - João Filipe Raposo
- NOVA Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon, Portugal
- Education and Research Center (APDP-ERC), APDP-Diabetes Portugal, Lisbon, Portugal
| | - Zdeněk Šumník
- Department of Pediatrics, Second Faculty of Medicine, Charles University in Prague and Motol University Hospital, Prague, Czech Republic
| | | | - Jurgen Vercauteren
- Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Christiane Winkler
- Institute of Diabetes Research, Helmholtz Munich German Research Center for Environmental Health, Munich, Germany
| | - Chantal Mathieu
- Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
- Department of Endocrinology, University Hospitals Leuven, Leuven, Belgium
| | - Anette-Gabriele Ziegler
- Institute of Diabetes Research, Helmholtz Munich German Research Center for Environmental Health, Munich, Germany
- Forschergruppe Diabetes at Klinikum Rechts der Isar, Technical University of Munich School of Medicine, Munich, Germany
| |
Collapse
|
2
|
Mallone R, Sims E, Achenbach P, Mathieu C, Pugliese A, Atkinson M, Dutta S, Evans-Molina C, Klatzmann D, Koralova A, Long SA, Overbergh L, Rodriguez-Calvo T, Ziegler AG, You S. Emerging Concepts and Success Stories in Type 1 Diabetes Research: A Road Map for a Bright Future. Diabetes 2025; 74:12-21. [PMID: 39446565 DOI: 10.2337/db24-0439] [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: 05/27/2024] [Accepted: 10/20/2024] [Indexed: 10/26/2024]
Abstract
Type 1 diabetes treatment stands at a crucial and exciting crossroad since the 2022 U.S. Food and Drug Administration approval of teplizumab to delay disease development. In this article, we discuss four major conceptual and practical issues that emerged as key to further advancement in type 1 diabetes research and therapies. First, collaborative networks leveraging the synergy between the type 1 diabetes research and care community members are key to fostering innovation, know-how, and translation into the clinical arena worldwide. Second, recent clinical trials in presymptomatic stage 2 and recent-onset stage 3 disease have shown the promise, and potential pitfalls, of using immunomodulatory and/or β-cell protective agents to achieve sustained remission or prevention. Third, the increasingly appreciated heterogeneity of clinical, immunological, and metabolic phenotypes and disease trajectories is of critical importance to advance the decision-making process for tailored type 1 diabetes care and therapy. Fourth, the clinical benefits of early diagnosis of β-cell autoimmunity warrant consideration of general population screening for islet autoantibodies, which requires further efforts to address the technical, organizational, and ethical challenges inherent to a sustainable program. Efforts are underway to integrate these four concepts into the future directions of type 1 diabetes research and therapy. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Roberto Mallone
- Institut Cochin, CNRS, INSERM, Université Paris Cité, Paris, France
- Service de Diabétologie et Immunologie Clinique, Hôpital Cochin, Assistance Publique Hôpitaux de Paris, Paris, France
- Indiana Biosciences Research Institute, Indianapolis, IN
| | - Emily Sims
- Division of Pediatric Endocrinology and Diabetology, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN
| | - Peter Achenbach
- Institute of Diabetes Research, Helmholtz Munich, German Center for Environmental Health, Munich, Germany
| | - Chantal Mathieu
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven, Belgium
| | - Alberto Pugliese
- Department of Diabetes Immunology, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope, Duarte, CA
| | - Mark Atkinson
- Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL
- Department of Pediatrics, University of Florida, Gainesville, FL
| | | | - Carmella Evans-Molina
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Department of Medicine, and Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN
- Richard L. Roudebush Veterans' Administration Medical Center, Indianapolis, IN
| | - David Klatzmann
- Clinical Investigation Center for Biotherapies and Inflammation-Immunopathology-Biotherapy Department (i2B), Sorbonne Université, Pitié-Salpêtrière Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
- Immunology-Immunopathology-Immunotherapy (i3), INSERM UMRS 959, Sorbonne UniversitéParis, France
| | - Anne Koralova
- The Leona M. and Harry B. Helmsley Charitable Trust, New York, NY
| | - S Alice Long
- Translational Immunology, Benaroya Research Institute, Seattle, WA
| | - Lut Overbergh
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven, Belgium
| | - Teresa Rodriguez-Calvo
- Institute of Diabetes Research, Helmholtz Munich, German Center for Environmental Health, Munich, Germany
| | - Anette-Gabriele Ziegler
- Institute of Diabetes Research, Helmholtz Munich, German Center for Environmental Health, Munich, Germany
| | - Sylvaine You
- Institut Cochin, CNRS, INSERM, Université Paris Cité, Paris, France
- Indiana Biosciences Research Institute, Indianapolis, IN
| |
Collapse
|
3
|
Achenbach P, Berner R, Bonifacio E, Brämswig S, Braig S, Dunstheimer D, Ermer U, Ewald D, Gemulla G, Hauer J, Haupt F, Haus G, Hubmann M, Hummel S, Kandler M, Kordonouri O, Lange K, Laub O, Lorrmann A, Nellen-Hellmuth N, Sindichakis M, von dem Berge T, Warncke K, Weber L, Winkler C, Wintermeyer P, Ziegler AG. [Early Detection Of Type 1 Diabetes By Islet Autoantibody Screening: A Position Paper Of The Fr1daplex Project Leaders And Training Centres, Bvkj Bavaria And Paednetz (Registered) Bavaria]. DAS GESUNDHEITSWESEN 2025; 87:27-37. [PMID: 38710228 PMCID: PMC11740224 DOI: 10.1055/a-2320-2859] [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] [Indexed: 05/08/2024]
Abstract
This position paper is based on the authors' many years of clinical experience and basic science research on the diagnosis and treatment of children and adolescents with a presymptomatic early stage of type 1 diabetes. The benefits as well as potential disadvantages of early detection of type 1 diabetes by islet autoantibody screening are critically discussed. In addition, the perspectives of delaying the onset of the clinical metabolic disease through treatment with teplizumab are addressed. Today, we see the chance for a relevant improvement in therapeutic options and life perspectives of affected children and adolescents. Important next steps for the implementation of islet autoantibody screening in Germany are the training of pediatricians who should inform families about the screening, establishment of a few transregional laboratories that carry out the test, and expansion of regional capacities for the training and care of children with an early stage of type 1 diabetes.
Collapse
Affiliation(s)
- Peter Achenbach
- Institut für Diabetesforschung, Helmholtz Zentrum München Deutsches
Forschungszentrum für Gesundheit und Umwelt, Neuherberg, Germany
- Forschergruppe Diabetes, Klinikum rechts der Isar, Technische
Universität München Fakultät für Medizin, Munchen, Germany
| | - Reinhard Berner
- Universitätsklinikum Carl Gustav Carus, Klinik und Poliklinik für
Kinder- und Jugendmedizin, Technische Universität Dresden, Dresden,
Germany
| | - Ezio Bonifacio
- Center for Regenerative Therapies Dresden, Technische Universität
Dresden, Dresden, Germany
| | - Susanne Brämswig
- Klinik für Kinder- und Jugendmedizin, RoMed Klinikum Rosenheim,
Rosenheim, Germany
| | - Sonja Braig
- Klinik für Kinder und Jugendliche, Klinikum Bayreuth GmbH, Bayreuth,
Germany
| | - Desiree Dunstheimer
- Klinik für Kinder- und Jugendmedizin, Universitätsklinikum Augsburg,
Augsburg, Germany
| | - Uwe Ermer
- Kinder- und Jugendmedizin, Ameos Klinikum St. Elisabeth Neuburg,
Neuburg an der Donau, Germany
| | - Dominik Ewald
- Bahnhofstr. 24, Kinderarztpraxis, Regensburg, Germany
| | - Gita Gemulla
- Universitätsklinikum Carl Gustav Carus, Klinik und Poliklinik für
Kinder- und Jugendmedizin, Technische Universität Dresden, Dresden,
Germany
- Center for Regenerative Therapies Dresden, Technische Universität
Dresden, Dresden, Germany
| | - Julia Hauer
- Zentrum für Kinder und Jugendmedizin, München Klinik und Klinikum
rechts der Isar, Technische Universität München Fakultät für Medizin, Munchen,
Germany
| | - Florian Haupt
- Institut für Diabetesforschung, Helmholtz Zentrum München Deutsches
Forschungszentrum für Gesundheit und Umwelt, Neuherberg, Germany
- Forschergruppe Diabetes, Klinikum rechts der Isar, Technische
Universität München Fakultät für Medizin, Munchen, Germany
| | - Gabi Haus
- Hans-Mielich-Str. 35, Kinderarztpraxis, München, Germany
| | | | - Sandra Hummel
- Institut für Diabetesforschung, Helmholtz Zentrum München Deutsches
Forschungszentrum für Gesundheit und Umwelt, Neuherberg, Germany
- Forschergruppe Diabetes, Klinikum rechts der Isar, Technische
Universität München Fakultät für Medizin, Munchen, Germany
| | | | - Olga Kordonouri
- Diabetologie, Endokrinologie und Allgemeine Pädiatrie, Kinder- und
Jugendkrankenhaus AUF DER BULT, Hannover, Germany
| | - Karin Lange
- Medizinische Psychologie, Medizinische Hochschule Hannover, Hannover,
Germany
| | - Otto Laub
- Happinger Str. 98, Kinderarztpraxis, Rosenheim, Germany
| | - Anja Lorrmann
- Kinder und Jugendmedizin, KJF Klinik Josefinum GmbH, Augsburg,
Germany
| | | | - Marina Sindichakis
- Klinik für Kinder- und Jugendmedizin, Kinderdiabetologie, Klinikum
Traunstein, Traunstein, Germany
| | - Thekla von dem Berge
- Diabetologie, Endokrinologie und Allgemeine Pädiatrie, Kinder- und
Jugendkrankenhaus AUF DER BULT, Hannover, Germany
| | - Katharina Warncke
- Zentrum für Kinder und Jugendmedizin, München Klinik und Klinikum
rechts der Isar, Technische Universität München Fakultät für Medizin, Munchen,
Germany
| | - Leonie Weber
- Klinik für Kinderheilkunde und Jugendmedizin, Kinderdiabetologie,
Klinikum Kempten-Oberallgau GmbH, Kempten, Germany
| | - Christiane Winkler
- Institut für Diabetesforschung, Helmholtz Zentrum München Deutsches
Forschungszentrum für Gesundheit und Umwelt, Neuherberg, Germany
- Forschergruppe Diabetes, Klinikum rechts der Isar, Technische
Universität München Fakultät für Medizin, Munchen, Germany
| | | | - Anette-Gabriele Ziegler
- Forschergruppe Diabetes, Klinikum rechts der Isar, Technische
Universität München Fakultät für Medizin, Munchen, Germany
- Institute of Diabetes Research, Helmholtz Center Munich German Research
Center for Environmental Health, Neuherberg, Germany
| |
Collapse
|
4
|
Mallone R, Bismuth E, Thivolet C, Benhamou PY, Hoffmeister N, Collet F, Nicolino M, Reynaud R, Beltrand J. Screening and care for preclinical stage 1-2 type 1 diabetes in first-degree relatives: French expert position statement. DIABETES & METABOLISM 2025; 51:101603. [PMID: 39675522 DOI: 10.1016/j.diabet.2024.101603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 11/29/2024] [Accepted: 12/11/2024] [Indexed: 12/17/2024]
Abstract
The natural history of type 1 diabetes (T1D) evolves from stage 1 (islet autoimmunity with normoglycemia; ICD-10 diagnostic code E10.A1) to stage 2 (autoimmunity with dysglycemia; E10.A2) and subsequent clinical stage 3 (overt hyperglycemia), which is commonly the first time of referral. Autoantibody testing can diagnose T1D at its preclinical stages 1-2 and lead to earlier initiation of care, particularly for first-degree relatives of people living with T1D, who are at higher genetic risk. Preclinical T1D screening and monitoring aims to avoid inaugural ketoacidosis and prolong preservation of endogenous insulin secretion, thereby improving glycemic control and reducing long-term morbidity. Moreover, early management can help coping with T1D and correct modifiable risk factors (obesity, sedentary lifestyle). New treatments currently under clinical deployment or trials also offer the possibility of delaying clinical progression. All these arguments lead to the proposition of a national screening and care pathway open to interested first-degree relatives. This pathway represents a new expertise to acquire for healthcare professionals. By adapting international consensus guidance to the French specificities, the proposed screening strategy involves testing for ≥ 2 autoantibodies (among IAA, anti-GAD, anti-IA-2) in relatives aged 2-45 years. Negative screening (∼95 % of cases) should be repeated every 4 years until the age of 12. A management workflow is proposed for relatives screening positive (∼5 % of cases), with immuno-metabolic monitoring by autoantibody testing, OGTT, glycemia and/or HbA1c of variable frequency, depending on T1D stage, age, patient preference and available resources, as well as the definition of expert centers for preclinical T1D.
Collapse
Affiliation(s)
- Roberto Mallone
- Université Paris Cité, Institut Cochin, CNRS, INSERM, Paris, France; Assistance Publique Hôpitaux de Paris, Université Paris Cité, Service de Diabétologie et Immunologie Clinique, Hôpital Cochin, Paris, France; Indiana Biosciences Research Institute, Indianapolis, IN, USA.
| | - Elise Bismuth
- Assistance Publique Hôpitaux de Paris, Université Paris Cité, Service d'Endocrinologie et Diabétologie Pédiatrique, Hôpital Robert Debré, Paris, France
| | - Charles Thivolet
- Hospices Civils de Lyon, Université de Lyon, Centre du diabète DIAB-eCARE, Lyon, France
| | - Pierre-Yves Benhamou
- Université Grenoble Alpes, INSERM U1055, LBFA, Endocrinologie, CHU Grenoble Alpes, France
| | | | - François Collet
- CHU Lille, Psychiatrie de Liaison et psycho-oncologie, Lille, France
| | - Marc Nicolino
- Hospices Civils de Lyon, Université de Lyon, Service d'Endocrinologie et Diabétologie Pédiatrique, Lyon, France
| | - Rachel Reynaud
- Assistance Publique Hôpitaux de Marseille, Université Aix-Marseille, Service de Pédiatrie Multidisciplinaire, Hôpital de la Timone, Marseille, France
| | - Jacques Beltrand
- Université Paris Cité, Institut Cochin, CNRS, INSERM, Paris, France; Assistance Publique Hôpitaux de Paris, Université Paris Cité, Service d'Endocrinologie, Gynécologie et Diabétologie Pédiatrique, Necker Hospital, Paris, France
| |
Collapse
|
5
|
ElSayed NA, McCoy RG, Aleppo G, Balapattabi K, Beverly EA, Briggs Early K, Bruemmer D, Ebekozien O, Echouffo-Tcheugui JB, Ekhlaspour L, Gaglia JL, Garg R, Khunti K, Lal R, Lingvay I, Matfin G, Pandya N, Pekas EJ, Pilla SJ, Polsky S, Segal AR, Seley JJ, Selvin E, Stanton RC, Bannuru RR. 2. Diagnosis and Classification of Diabetes: Standards of Care in Diabetes-2025. Diabetes Care 2025; 48:S27-S49. [PMID: 39651986 PMCID: PMC11635041 DOI: 10.2337/dc25-s002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 09/12/2024] [Indexed: 12/14/2024]
Abstract
The American Diabetes Association (ADA) "Standards of Care in Diabetes" includes the ADA's current clinical practice recommendations and is intended to provide the components of diabetes care, general treatment goals and guidelines, and tools to evaluate quality of care. Members of the ADA Professional Practice Committee, an interprofessional expert committee, are responsible for updating the Standards of Care annually, or more frequently as warranted. For a detailed description of ADA standards, statements, and reports, as well as the evidence-grading system for ADA's clinical practice recommendations and a full list of Professional Practice Committee members, please refer to Introduction and Methodology. Readers who wish to comment on the Standards of Care are invited to do so at professional.diabetes.org/SOC.
Collapse
|
6
|
Jacobsen LM, Atkinson MA, Sosenko JM, Gitelman SE. Time to reframe the disease staging system for type 1 diabetes. Lancet Diabetes Endocrinol 2024; 12:924-933. [PMID: 39608963 DOI: 10.1016/s2213-8587(24)00239-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 06/16/2024] [Accepted: 07/25/2024] [Indexed: 11/30/2024]
Abstract
In 2015, introduction of a disease staging system offered a framework for benchmarking progression to clinical type 1 diabetes. This model, based on islet autoantibodies (stage 1) and dysglycaemia (stage 2) before type 1 diabetes diagnosis (stage 3), has facilitated screening and identification of people at risk. Yet, there are many limitations to this model as the stages combine a very heterogeneous group of individuals; do not have high specificity for type 1 diabetes; can occur without persistence (ie, reversion to an earlier risk stage); and exclude age and other influential risk factors. The current staging system also infers that individuals at risk of type 1 diabetes progress linearly from stage 1 to stage 2 and subsequently stage 3, whereas such movements are often more complex. With the approval of teplizumab by the US Food and Drug Administration in 2022 to delay type 1 diabetes in people at stage 2, there is a need to refine the definition and accuracy of type 1 diabetes staging. Theoretically, we propose that a type 1 diabetes risk calculator should incorporate any available demographic, genetic, autoantibody, metabolic, and immune data that could be continuously updated. Additionally, we call to action for the field to increase the breadth of knowledge regarding type 1 diabetes risk in non-relatives, adults, and individuals from minority populations.
Collapse
Affiliation(s)
- Laura M Jacobsen
- Department of Paediatrics and Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, USA.
| | - Mark A Atkinson
- Department of Paediatrics and Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Jay M Sosenko
- Division of Endocrinology, University of Miami, Miami, FL, USA
| | - Stephen E Gitelman
- Department of Paediatrics, Diabetes Center, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
7
|
Kelly CS, Wolf WA, Cornelius EM, Peter ME, Chapman KS, Dunne JL. Insights into Knowledge and Attitudes About Autoantibody Screening from People Affected by Type 1 Diabetes: A Brief Report. Diabetes Ther 2024; 15:2249-2261. [PMID: 39190273 PMCID: PMC11410738 DOI: 10.1007/s13300-024-01637-z] [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: 05/08/2024] [Accepted: 07/31/2024] [Indexed: 08/28/2024] Open
Abstract
INTRODUCTION Screening for islet-specific autoantibodies can identify individuals at risk for type 1 diabetes (T1D). Despite calls for increased nationwide autoantibody screening efforts, it is unclear how many individuals have participated in screening among people who may benefit from it. Moreover, knowledge and perceptions of autoantibody screening in real-world samples are not well understood. METHODS We surveyed a sample of individuals (aged 18+ years old) from T1D Exchange Registry with a personal or family history of T1D to assess their self-reported T1D autoantibody knowledge, experiences, and attitudes. Participants belonged to one of three groups: adults with T1D who had a biological child without T1D or future plans for a child (PWD); parents without T1D who had a biological child with T1D and one or more biological children without T1D (Caregivers); and first-degree adult children or siblings to a person with T1D (Relatives). Descriptive analyses (means, standard deviations, frequencies) are presented by participant groups. RESULTS A total of 510 participants enrolled in the study. Across groups, participants reported feeling a little to somewhat knowledgeable about autoantibody screening and positive perceptions of autoantibody screening in general. However, few participants had screened their child without T1D (PWDs, 21.94%; Caregivers, 46.30%) or themselves (Relatives, 19.23%). Among those who had screened, participants reported generally positive experiences. Among those who had not screened, many participants were "undecided" about autoantibody screening (PWD, 38.46%; Caregivers, 40.52%; Relatives, 44.44%). Influences reported for participants' decisions to screen, not screen, or their current indecision differed by group: PWDs (21.70%) and Caregivers (26.87%) most often reported self-initiated research as an influence and Relatives reported they had not previously considered screening (48.28%). CONCLUSION Results highlight the need for more accessible information about screening, including real experiences from those who have screened.
Collapse
Affiliation(s)
- Caitlin S Kelly
- T1D Exchange, 101 Federal St Suite 440, Boston, MA, 02110, USA.
| | - Wendy A Wolf
- T1D Exchange, 101 Federal St Suite 440, Boston, MA, 02110, USA
| | | | - Megan E Peter
- T1D Exchange, 101 Federal St Suite 440, Boston, MA, 02110, USA
| | | | - Jessica L Dunne
- Janssen R&D, 1000 U.S. Route 202 South, Raritan, NJ, 08869, USA
- General Medicine, US Medical Bridgewater, Sanofi, NJ, 08807, USA
| |
Collapse
|
8
|
Phillip M, Achenbach P, Addala A, Albanese-O'Neill A, Battelino T, Bell KJ, Besser REJ, Bonifacio E, Colhoun HM, Couper JJ, Craig ME, Danne T, de Beaufort C, Dovc K, Driscoll KA, Dutta S, Ebekozien O, Larsson HE, Feiten DJ, Frohnert BI, Gabbay RA, Gallagher MP, Greenbaum CJ, Griffin KJ, Hagopian W, Haller MJ, Hendrieckx C, Hendriks E, Holt RIG, Hughes L, Ismail HM, Jacobsen LM, Johnson SB, Kolb LE, Kordonouri O, Lange K, Lash RW, Lernmark Å, Libman I, Lundgren M, Maahs DM, Marcovecchio ML, Mathieu C, Miller KM, O'Donnell HK, Oron T, Patil SP, Pop-Busui R, Rewers MJ, Rich SS, Schatz DA, Schulman-Rosenbaum R, Simmons KM, Sims EK, Skyler JS, Smith LB, Speake C, Steck AK, Thomas NPB, Tonyushkina KN, Veijola R, Wentworth JM, Wherrett DK, Wood JR, Ziegler AG, DiMeglio LA. Consensus guidance for monitoring individuals with islet autoantibody-positive pre-stage 3 type 1 diabetes. Diabetologia 2024; 67:1731-1759. [PMID: 38910151 PMCID: PMC11410955 DOI: 10.1007/s00125-024-06205-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/25/2024]
Abstract
Given the proven benefits of screening to reduce diabetic ketoacidosis (DKA) likelihood at the time of stage 3 type 1 diabetes diagnosis, and emerging availability of therapy to delay disease progression, type 1 diabetes screening programmes are being increasingly emphasised. Once broadly implemented, screening initiatives will identify significant numbers of islet autoantibody-positive (IAb+) children and adults who are at risk of (confirmed single IAb+) or living with (multiple IAb+) early-stage (stage 1 and stage 2) type 1 diabetes. These individuals will need monitoring for disease progression; much of this care will happen in non-specialised settings. To inform this monitoring, JDRF in conjunction with international experts and societies developed consensus guidance. Broad advice from this guidance includes the following: (1) partnerships should be fostered between endocrinologists and primary-care providers to care for people who are IAb+; (2) when people who are IAb+ are initially identified there is a need for confirmation using a second sample; (3) single IAb+ individuals are at lower risk of progression than multiple IAb+ individuals; (4) individuals with early-stage type 1 diabetes should have periodic medical monitoring, including regular assessments of glucose levels, regular education about symptoms of diabetes and DKA, and psychosocial support; (5) interested people with stage 2 type 1 diabetes should be offered trial participation or approved therapies; and (6) all health professionals involved in monitoring and care of individuals with type 1 diabetes have a responsibility to provide education. The guidance also emphasises significant unmet needs for further research on early-stage type 1 diabetes to increase the rigour of future recommendations and inform clinical care.
Collapse
Affiliation(s)
- Moshe Phillip
- Institute for Endocrinology and Diabetes, National Center for Childhood Diabetes, Schneider Children's Medical Center of Israel, Petah Tikva, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Peter Achenbach
- Institute of Diabetes Research, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany
- Forschergruppe Diabetes, Technical University Munich, Klinikum Rechts Der Isar, Munich, Germany
| | - Ananta Addala
- Division of Endocrinology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Tadej Battelino
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Department of Endocrinology, Diabetes and Metabolism, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Kirstine J Bell
- Charles Perkins Centre and Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Rachel E J Besser
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Wellcome Centre Human Genetics, Nuffield Department of Medicine Oxford NIHR Biomedical Research Centre, University of Oxford, Oxford, UK
- Department of Paediatrics, University of Oxford, Oxford, UK
| | - Ezio Bonifacio
- Center for Regenerative Therapies Dresden, Faculty of Medicine, Technical University of Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden, Helmholtz Centre Munich at the University Clinic Carl Gustav Carus of TU Dresden and Faculty of Medicine, Dresden, Germany
| | - Helen M Colhoun
- The Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
- Department of Public Health, NHS Fife, Kirkcaldy, UK
| | - Jennifer J Couper
- Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
- Division of Paediatrics, Women's and Children's Hospital, Adelaide, SA, Australia
| | - Maria E Craig
- Charles Perkins Centre and Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
- Discipline of Paediatrics & Child Health, School of Clinical Medicine, UNSW Medicine & Health, Sydney, NSW, Australia
| | | | - Carine de Beaufort
- International Society for Pediatric and Adolescent Diabetes (ISPAD), Berlin, Germany
- Diabetes & Endocrine Care Clinique Pédiatrique (DECCP), Clinique Pédiatrique/Centre Hospitalier (CH) de Luxembourg, Luxembourg City, Luxembourg
- Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-Belval, Luxembourg
| | - Klemen Dovc
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Department of Endocrinology, Diabetes and Metabolism, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Kimberly A Driscoll
- Department of Pediatrics, Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Clinical and Health Psychology, University of Florida, Gainesville, FL, USA
- Department of Pediatrics, University of Florida Diabetes Institute, Gainesville, FL, USA
| | | | | | - Helena Elding Larsson
- Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Department of Pediatrics, Skåne University Hospital, Malmö and Lund, Sweden
| | | | - Brigitte I Frohnert
- Department of Pediatrics, Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | | | | | - Carla J Greenbaum
- Center for Interventional Immunology and Diabetes Program, Benaroya Research Institute, Seattle, WA, USA
| | - Kurt J Griffin
- Sanford Research, Sioux Falls, SD, USA
- Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD, USA
| | - William Hagopian
- Pacific Northwest Diabetes Research Institute, University of Washington, Seattle, WA, USA
| | - Michael J Haller
- Department of Pediatrics, University of Florida Diabetes Institute, Gainesville, FL, USA
- Division of Endocrinology, University of Florida College of Medicine, Gainesville, FL, USA
| | - Christel Hendrieckx
- School of Psychology, Deakin University, Geelong, VIC, Australia
- The Australian Centre for Behavioural Research in Diabetes, Diabetes Victoria, Carlton, VIC, Australia
- Institute for Health Transformation, Deakin University, Geelong, VIC, Australia
| | - Emile Hendriks
- Department of Paediatrics, University of Cambridge and Cambridge University Hospitals NHS Foundation Trust, Cambridge Biomedical Campus, Cambridge, UK
| | - Richard I G Holt
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
- National Institute for Health and Care Research Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | | | - Heba M Ismail
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Laura M Jacobsen
- Division of Endocrinology, University of Florida College of Medicine, Gainesville, FL, USA
| | - Suzanne B Johnson
- Department of Behavioral Sciences and Social Medicine, Florida State University College of Medicine, Tallahassee, FL, USA
| | - Leslie E Kolb
- Association of Diabetes Care & Education Specialists, Chicago, IL, USA
| | | | - Karin Lange
- Medical Psychology, Hannover Medical School, Hannover, Germany
| | | | - Åke Lernmark
- Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
| | - Ingrid Libman
- Division of Pediatric Endocrinology and Diabetes, University of Pittsburgh, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Markus Lundgren
- Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Department of Pediatrics, Kristianstad Hospital, Kristianstad, Sweden
| | - David M Maahs
- Division of Endocrinology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - M Loredana Marcovecchio
- Department of Pediatrics, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Chantal Mathieu
- Department of Endocrinology, UZ Gasthuisberg, KU Leuven, Leuven, Belgium
| | | | - Holly K O'Donnell
- Department of Pediatrics, Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Tal Oron
- Institute for Endocrinology and Diabetes, National Center for Childhood Diabetes, Schneider Children's Medical Center of Israel, Petah Tikva, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Shivajirao P Patil
- Department of Family Medicine, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - Rodica Pop-Busui
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, MI, USA
| | - Marian J Rewers
- Department of Pediatrics, Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Desmond A Schatz
- Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - Rifka Schulman-Rosenbaum
- Division of Endocrinology, Long Island Jewish Medical Center, Northwell Health, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, New Hyde Park, NY, USA
| | - Kimber M Simmons
- Department of Pediatrics, Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Emily K Sims
- Division of Pediatric Endocrinology and Diabetology, Herman B Wells Center for Pediatric Research, Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jay S Skyler
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Laura B Smith
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Cate Speake
- Center for Interventional Immunology and Diabetes Program, Benaroya Research Institute, Seattle, WA, USA
| | - Andrea K Steck
- Department of Pediatrics, Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | | | - Ksenia N Tonyushkina
- Division of Endocrinology and Diabetes, Baystate Children's Hospital and University of Massachusetts Chan Medical School - Baystate, Springfield, MA, USA
| | - Riitta Veijola
- Research Unit of Clinical Medicine, Department of Pediatrics, Medical Research Center, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - John M Wentworth
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Diabetes and Endocrinology, Royal Melbourne Hospital, Parkville, VIC, Australia
| | - Diane K Wherrett
- Department of Paediatrics, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Jamie R Wood
- Department of Pediatric Endocrinology, Rainbow Babies and Children's Hospital, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Anette-Gabriele Ziegler
- Institute of Diabetes Research, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany
- Forschergruppe Diabetes, Technical University Munich, Klinikum Rechts Der Isar, Munich, Germany
| | - Linda A DiMeglio
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
9
|
Robertson CC, Elgamal RM, Henry-Kanarek BA, Arvan P, Chen S, Dhawan S, Eizirik DL, Kaddis JS, Vahedi G, Parker SCJ, Gaulton KJ, Soleimanpour SA. Untangling the genetics of beta cell dysfunction and death in type 1 diabetes. Mol Metab 2024; 86:101973. [PMID: 38914291 PMCID: PMC11283044 DOI: 10.1016/j.molmet.2024.101973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 06/18/2024] [Accepted: 06/19/2024] [Indexed: 06/26/2024] Open
Abstract
BACKGROUND Type 1 diabetes (T1D) is a complex multi-system disease which arises from both environmental and genetic factors, resulting in the destruction of insulin-producing pancreatic beta cells. Over the past two decades, human genetic studies have provided new insight into the etiology of T1D, including an appreciation for the role of beta cells in their own demise. SCOPE OF REVIEW Here, we outline models supported by human genetic data for the role of beta cell dysfunction and death in T1D. We highlight the importance of strong evidence linking T1D genetic associations to bona fide candidate genes for mechanistic and therapeutic consideration. To guide rigorous interpretation of genetic associations, we describe molecular profiling approaches, genomic resources, and disease models that may be used to construct variant-to-gene links and to investigate candidate genes and their role in T1D. MAJOR CONCLUSIONS We profile advances in understanding the genetic causes of beta cell dysfunction and death at individual T1D risk loci. We discuss how genetic risk prediction models can be used to address disease heterogeneity. Further, we present areas where investment will be critical for the future use of genetics to address open questions in the development of new treatment and prevention strategies for T1D.
Collapse
Affiliation(s)
- Catherine C Robertson
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA; Center for Precision Health Research, National Human Genome Research Institute, NIH, Bethesda, MD 20892, USA
| | - Ruth M Elgamal
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
| | - Belle A Henry-Kanarek
- Department of Internal Medicine and Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, USA
| | - Peter Arvan
- Department of Internal Medicine and Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, USA
| | - Shuibing Chen
- Department of Surgery, Weill Cornell Medicine, New York, NY, USA; Center for Genomic Health, Weill Cornell Medicine, New York, NY, USA
| | - Sangeeta Dhawan
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA, USA
| | - Decio L Eizirik
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
| | - John S Kaddis
- Department of Diabetes and Cancer Discovery Science, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Golnaz Vahedi
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Stephen C J Parker
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA; Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA; Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA.
| | - Kyle J Gaulton
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA.
| | - Scott A Soleimanpour
- Department of Internal Medicine and Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
10
|
Phillip M, Achenbach P, Addala A, Albanese-O’Neill A, Battelino T, Bell KJ, Besser RE, Bonifacio E, Colhoun HM, Couper JJ, Craig ME, Danne T, de Beaufort C, Dovc K, Driscoll KA, Dutta S, Ebekozien O, Elding Larsson H, Feiten DJ, Frohnert BI, Gabbay RA, Gallagher MP, Greenbaum CJ, Griffin KJ, Hagopian W, Haller MJ, Hendrieckx C, Hendriks E, Holt RI, Hughes L, Ismail HM, Jacobsen LM, Johnson SB, Kolb LE, Kordonouri O, Lange K, Lash RW, Lernmark Å, Libman I, Lundgren M, Maahs DM, Marcovecchio ML, Mathieu C, Miller KM, O’Donnell HK, Oron T, Patil SP, Pop-Busui R, Rewers MJ, Rich SS, Schatz DA, Schulman-Rosenbaum R, Simmons KM, Sims EK, Skyler JS, Smith LB, Speake C, Steck AK, Thomas NP, Tonyushkina KN, Veijola R, Wentworth JM, Wherrett DK, Wood JR, Ziegler AG, DiMeglio LA. Consensus Guidance for Monitoring Individuals With Islet Autoantibody-Positive Pre-Stage 3 Type 1 Diabetes. Diabetes Care 2024; 47:1276-1298. [PMID: 38912694 PMCID: PMC11381572 DOI: 10.2337/dci24-0042] [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: 05/07/2024] [Accepted: 05/09/2024] [Indexed: 06/25/2024]
Abstract
Given the proven benefits of screening to reduce diabetic ketoacidosis (DKA) likelihood at the time of stage 3 type 1 diabetes diagnosis, and emerging availability of therapy to delay disease progression, type 1 diabetes screening programs are being increasingly emphasized. Once broadly implemented, screening initiatives will identify significant numbers of islet autoantibody-positive (IAb+) children and adults who are at risk for (confirmed single IAb+) or living with (multiple IAb+) early-stage (stage 1 and stage 2) type 1 diabetes. These individuals will need monitoring for disease progression; much of this care will happen in nonspecialized settings. To inform this monitoring, JDRF, in conjunction with international experts and societies, developed consensus guidance. Broad advice from this guidance includes the following: 1) partnerships should be fostered between endocrinologists and primary care providers to care for people who are IAb+; 2) when people who are IAb+ are initially identified, there is a need for confirmation using a second sample; 3) single IAb+ individuals are at lower risk of progression than multiple IAb+ individuals; 4) individuals with early-stage type 1 diabetes should have periodic medical monitoring, including regular assessments of glucose levels, regular education about symptoms of diabetes and DKA, and psychosocial support; 5) interested people with stage 2 type 1 diabetes should be offered trial participation or approved therapies; and 6) all health professionals involved in monitoring and care of individuals with type 1 diabetes have a responsibility to provide education. The guidance also emphasizes significant unmet needs for further research on early-stage type 1 diabetes to increase the rigor of future recommendations and inform clinical care.
Collapse
Affiliation(s)
- Moshe Phillip
- Institute for Endocrinology and Diabetes, National Center for Childhood Diabetes, Schneider Children's Medical Center of Israel, Petah Tikva, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Peter Achenbach
- Institute of Diabetes Research, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany
- Forschergruppe Diabetes, Technical University Munich, Klinikum Rechts Der Isar, Munich, Germany
| | - Ananta Addala
- Division of Endocrinology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA
- Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA
| | | | - Tadej Battelino
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Department of Endocrinology, Diabetes and Metabolism, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Kirstine J. Bell
- Charles Perkins Centre and Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Rachel E.J. Besser
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Wellcome Centre Human Genetics, Nuffield Department of Medicine Oxford National Institute for Health and Care Research Biomedical Research Centre, University of Oxford, Oxford, U.K
- Department of Paediatrics, University of Oxford, Oxford, U.K
| | - Ezio Bonifacio
- Center for Regenerative Therapies Dresden, Faculty of Medicine, Technical University of Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden, Helmholtz Centre Munich at the University Clinic Carl Gustav Carus of Technical University of Dresden, and Faculty of Medicine, Technical University of Dresden, Dresden, Germany
| | - Helen M. Colhoun
- The Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, U.K
- Department of Public Health, NHS Fife, Kirkcaldy, U.K
| | - Jennifer J. Couper
- Robinson Research Institute, The University of Adelaide, Adelaide, South Australia, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
- Division of Paediatrics, Women’s and Children’s Hospital, Adelaide, South Australia, Australia
| | - Maria E. Craig
- Charles Perkins Centre and Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Discipline of Paediatrics & Child Health, School of Clinical Medicine, UNSW Medicine & Health, Sydney, New South Wales, Australia
| | | | - Carine de Beaufort
- International Society for Pediatric and Adolescent Diabetes (ISPAD), Berlin, Germany
- Diabetes & Endocrine Care Clinique Pédiatrique (DECCP), Clinique Pédiatrique/Centre Hospitalier (CH) de Luxembourg, Luxembourg City, Luxembourg
- Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-Belval, Luxembourg
| | - Klemen Dovc
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Department of Endocrinology, Diabetes and Metabolism, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Kimberly A. Driscoll
- Department of Pediatrics, Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO
- Department of Clinical and Health Psychology, University of Florida, Gainesville, FL
- Department of Pediatrics, University of Florida Diabetes Institute, Gainesville, FL
| | | | | | - Helena Elding Larsson
- Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Department of Pediatrics, Skåne University Hospital, Malmö and Lund, Sweden
| | | | - Brigitte I. Frohnert
- Department of Pediatrics, Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO
| | | | | | - Carla J. Greenbaum
- Center for Interventional Immunology and Diabetes Program, Benaroya Research Institute, Seattle, WA
| | - Kurt J. Griffin
- Sanford Research, Sioux Falls, SD
- Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD
| | - William Hagopian
- Pacific Northwest Diabetes Research Institute, University of Washington, Seattle, WA
| | - Michael J. Haller
- Department of Pediatrics, University of Florida Diabetes Institute, Gainesville, FL
- Division of Endocrinology, University of Florida College of Medicine, Gainesville, FL
| | - Christel Hendrieckx
- School of Psychology, Deakin University, Geelong, Victoria, Australia
- The Australian Centre for Behavioural Research in Diabetes, Diabetes Victoria, Carlton, Victoria, Australia
- Institute for Health Transformation, Deakin University, Geelong, Victoria, Australia
| | - Emile Hendriks
- Department of Paediatrics, University of Cambridge and Cambridge University Hospitals NHS Foundation Trust, Cambridge, U.K
| | - Richard I.G. Holt
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, U.K
- National Institute for Health and Care Research Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, U.K
| | | | - Heba M. Ismail
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN
| | - Laura M. Jacobsen
- Division of Endocrinology, University of Florida College of Medicine, Gainesville, FL
| | - Suzanne B. Johnson
- Department of Behavioral Sciences and Social Medicine, Florida State University College of Medicine, Tallahassee, FL
| | - Leslie E. Kolb
- Association of Diabetes Care & Education Specialists, Chicago, IL
| | | | - Karin Lange
- Medical Psychology, Hannover Medical School, Hannover, Germany
| | | | - Åke Lernmark
- Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
| | - Ingrid Libman
- Division of Pediatric Endocrinology and Diabetes, University of Pittsburgh, University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, PA
| | - Markus Lundgren
- Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Department of Pediatrics, Kristianstad Hospital, Kristianstad, Sweden
| | - David M. Maahs
- Division of Endocrinology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA
| | | | - Chantal Mathieu
- Department of Endocrinology, UZ Gasthuisberg, KU Leuven, Leuven, Belgium
| | | | - Holly K. O’Donnell
- Department of Pediatrics, Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Tal Oron
- Institute for Endocrinology and Diabetes, National Center for Childhood Diabetes, Schneider Children's Medical Center of Israel, Petah Tikva, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Shivajirao P. Patil
- Department of Family Medicine, Brody School of Medicine, East Carolina University, Greenville, NC
| | - Rodica Pop-Busui
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, MI
| | - Marian J. Rewers
- Department of Pediatrics, Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Stephen S. Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA
| | | | - Rifka Schulman-Rosenbaum
- Division of Endocrinology, Long Island Jewish Medical Center, Northwell Health, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, New Hyde Park, NY
| | - Kimber M. Simmons
- Department of Pediatrics, Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Emily K. Sims
- Division of Pediatric Endocrinology and Diabetology, Herman B Wells Center for Pediatric Research, Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN
| | - Jay S. Skyler
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL
| | - Laura B. Smith
- Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Cate Speake
- Center for Interventional Immunology and Diabetes Program, Benaroya Research Institute, Seattle, WA
| | - Andrea K. Steck
- Department of Pediatrics, Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Nicholas P.B. Thomas
- National Institute of Health and Care Research Clinical Research Network Thames Valley and South Midlands, Oxford, U.K
| | - Ksenia N. Tonyushkina
- Division of Endocrinology and Diabetes, Baystate Children’s Hospital and University of Massachusetts Chan Medical School–Baystate, Springfield, MA
| | - Riitta Veijola
- Research Unit of Clinical Medicine, Department of Pediatrics, Medical Research Center, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - John M. Wentworth
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Diabetes and Endocrinology, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Diane K. Wherrett
- Department of Paediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Jamie R. Wood
- Department of Pediatric Endocrinology, Rainbow Babies and Children's Hospital, University Hospitals Cleveland Medical Center, Cleveland, OH
| | - Anette-Gabriele Ziegler
- Institute of Diabetes Research, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany
- Forschergruppe Diabetes, Technical University Munich, Klinikum Rechts Der Isar, Munich, Germany
| | - Linda A. DiMeglio
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN
| |
Collapse
|
11
|
Ghalwash M, Anand V, Ng K, Dunne JL, Lou O, Lundgren M, Hagopian WA, Rewers M, Ziegler AG, Veijola R. Data-Driven Phenotyping of Presymptomatic Type 1 Diabetes Using Longitudinal Autoantibody Profiles. Diabetes Care 2024; 47:1424-1431. [PMID: 38861550 PMCID: PMC11272969 DOI: 10.2337/dc24-0198] [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: 01/30/2024] [Accepted: 05/16/2024] [Indexed: 06/13/2024]
Abstract
OBJECTIVE To characterize distinct islet autoantibody profiles preceding stage 3 type 1 diabetes. RESEARCH DESIGN AND METHODS The T1DI (Type 1 Diabetes Intelligence) study combined data from 1,845 genetically susceptible prospectively observed children who were positive for at least one islet autoantibody: insulin autoantibody (IAA), GAD antibody (GADA), or islet antigen 2 antibody (IA-2A). Using a novel similarity algorithm that considers an individual's temporal autoantibody profile, age at autoantibody appearance, and variation in the positivity of autoantibody types, we performed an unsupervised hierarchical clustering analysis. Progression rates to diabetes were analyzed via survival analysis. RESULTS We identified five main clusters of individuals with distinct autoantibody profiles characterized by seroconversion age and sequence of appearance of the three autoantibodies. The highest 5-year risk from first positive autoantibody to type 1 diabetes (69.9%; 95% CI 60.0-79.2) was observed in children who first developed IAA in early life (median age 1.6 years) followed by GADA (1.9 years) and then IA-2A (2.1 years). Their 10-year risk was 89.9% (95% CI 81.9-95.4). A high 5-year risk was also found in children with persistent IAA and GADA (39.1%) and children with persistent GADA and IA-2A (30.9%). A lower 5-year risk (10.5%) was observed in children with a late appearance of persistent GADA (6.1 years). The lowest 5-year diabetes risk (1.6%) was associated with positivity for a single, often reverting, autoantibody. CONCLUSIONS The novel clustering algorithm identified children with distinct islet autoantibody profiles and progression rates to diabetes. These results are useful for prediction, selection of individuals for prevention trials, and studies investigating various pathways to type 1 diabetes.
Collapse
Affiliation(s)
- Mohamed Ghalwash
- T.J. Watson Research Center, IBM, Yorktown Heights, NY
- Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Vibha Anand
- T.J. Watson Research Center, IBM, Cambridge, MA
| | - Kenney Ng
- T.J. Watson Research Center, IBM, Yorktown Heights, NY
| | | | | | - Markus Lundgren
- Department of Clinical Sciences, Lund University/Clinical Research Centre, Skåne University Hospital, Malmö, Sweden
| | | | - Marian Rewers
- Department of Pediatrics, Barbara Davis Center for Diabetes, Denver, CO
| | - Anette-G. Ziegler
- Institute of Diabetes Research, German Research Center for Environmental Health, Helmholtz Zentrum München, Munich-Neuherberg, Germany
| | - Riitta Veijola
- Research Unit of Clinical Medicine, Medical Research Center, Department of Pediatrics, University of Oulu and Oulu University Hospital, Oulu, Finland
| |
Collapse
|
12
|
Carry PM, Vanderlinden LA, Johnson RK, Buckner T, Steck AK, Kechris K, Yang IV, Fingerlin TE, Fiehn O, Rewers M, Norris JM. Longitudinal changes in DNA methylation during the onset of islet autoimmunity differentiate between reversion versus progression of islet autoimmunity. Front Immunol 2024; 15:1345494. [PMID: 38915393 PMCID: PMC11194352 DOI: 10.3389/fimmu.2024.1345494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 05/21/2024] [Indexed: 06/26/2024] Open
Abstract
Background Type 1 diabetes (T1D) is preceded by a heterogenous pre-clinical phase, islet autoimmunity (IA). We aimed to identify pre vs. post-IA seroconversion (SV) changes in DNAm that differed across three IA progression phenotypes, those who lose autoantibodies (reverters), progress to clinical T1D (progressors), or maintain autoantibody levels (maintainers). Methods This epigenome-wide association study (EWAS) included longitudinal DNAm measurements in blood (Illumina 450K and EPIC) from participants in Diabetes Autoimmunity Study in the Young (DAISY) who developed IA, one or more islet autoantibodies on at least two consecutive visits. We compared reverters - individuals who sero-reverted, negative for all autoantibodies on at least two consecutive visits and did not develop T1D (n=41); maintainers - continued to test positive for autoantibodies but did not develop T1D (n=60); progressors - developed clinical T1D (n=42). DNAm data were measured before (pre-SV visit) and after IA (post-SV visit). Linear mixed models were used to test for differences in pre- vs post-SV changes in DNAm across the three groups. Linear mixed models were also used to test for group differences in average DNAm. Cell proportions, age, and sex were adjusted for in all models. Median follow-up across all participants was 15.5 yrs. (interquartile range (IQR): 10.8-18.7). Results The median age at the pre-SV visit was 2.2 yrs. (IQR: 0.8-5.3) in progressors, compared to 6.0 yrs. (IQR: 1.3-8.4) in reverters, and 5.7 yrs. (IQR: 1.4-9.7) in maintainers. Median time between the visits was similar in reverters 1.4 yrs. (IQR: 1-1.9), maintainers 1.3 yrs. (IQR: 1.0-2.0), and progressors 1.8 yrs. (IQR: 1.0-2.0). Changes in DNAm, pre- vs post-SV, differed across the groups at one site (cg16066195) and 11 regions. Average DNAm (mean of pre- and post-SV) differed across 22 regions. Conclusion Differentially changing DNAm regions were located in genomic areas related to beta cell function, immune cell differentiation, and immune cell function.
Collapse
Affiliation(s)
- Patrick M. Carry
- Colorado Program for Musculoskeletal Research, Department of Orthopedics, University of Colorado, Aurora, CO, United States
- Department of Epidemiology, Colorado School of Public Health, Aurora, CO, United States
- Department of Biomedical Informatics, School of Medicine, University of Colorado, Aurora, CO, United States
| | | | - Randi K. Johnson
- Department of Epidemiology, Colorado School of Public Health, Aurora, CO, United States
- Department of Biomedical Informatics, School of Medicine, University of Colorado, Aurora, CO, United States
| | - Teresa Buckner
- Department of Epidemiology, Colorado School of Public Health, Aurora, CO, United States
- Department of Kinesiology, Nutrition, and Dietetics, University of Northern Colorado, Greeley, CO, United States
| | - Andrea K. Steck
- Barbara Davis Center, Department of Pediatrics, University of Colorado, Aurora, CO, United States
| | - Katerina Kechris
- Department of Epidemiology, Colorado School of Public Health, Aurora, CO, United States
- Department of Biomedical Informatics, School of Medicine, University of Colorado, Aurora, CO, United States
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, CO, United States
| | - Ivana V. Yang
- Department of Biomedical Informatics, School of Medicine, University of Colorado, Aurora, CO, United States
- Department of Medicine, University of Colorado, Aurora, CO, United States
| | - Tasha E. Fingerlin
- Department of Epidemiology, Colorado School of Public Health, Aurora, CO, United States
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, CO, United States
- Department of Immunology and Genomic Medicine, National Jewish Health, Aurora, CO, United States
| | - Oliver Fiehn
- University of California Davis West Coast Metabolomics Center, Davis, CA, United States
| | - Marian Rewers
- Barbara Davis Center, Department of Pediatrics, University of Colorado, Aurora, CO, United States
| | - Jill M. Norris
- Department of Epidemiology, Colorado School of Public Health, Aurora, CO, United States
| |
Collapse
|
13
|
Lind A, Freyhult E, de Jesus Cortez F, Ramelius A, Bennet R, Robinson PV, Seftel D, Gebhart D, Tandel D, Maziarz M, Larsson HE, Lundgren M, Carlsson A, Nilsson AL, Fex M, Törn C, Agardh D, Tsai CT, Lernmark Å. Childhood screening for type 1 diabetes comparing automated multiplex Antibody Detection by Agglutination-PCR (ADAP) with single plex islet autoantibody radiobinding assays. EBioMedicine 2024; 104:105144. [PMID: 38723553 PMCID: PMC11090024 DOI: 10.1016/j.ebiom.2024.105144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 04/16/2024] [Accepted: 04/18/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND Two or more autoantibodies against either insulin (IAA), glutamic acid decarboxylase (GADA), islet antigen-2 (IA-2A) or zinc transporter 8 (ZnT8A) denote stage 1 (normoglycemia) or stage 2 (dysglycemia) type 1 diabetes prior to stage 3 type 1 diabetes. Automated multiplex Antibody Detection by Agglutination-PCR (ADAP) assays in two laboratories were compared to single plex radiobinding assays (RBA) to define threshold levels for diagnostic specificity and sensitivity. METHODS IAA, GADA, IA-2A and ZnT8A were analysed in 1504 (54% females) population based controls (PBC), 456 (55% females) doctor's office controls (DOC) and 535 (41% females) blood donor controls (BDC) as well as in 2300 (48% females) patients newly diagnosed (1-10 years of age) with stage 3 type 1 diabetes. The thresholds for autoantibody positivity were computed in 100 10-fold cross-validations to separate patients from controls either by maximizing the χ2-statistics (chisq) or using the 98th percentile of specificity (Spec98). Mean and 95% CI for threshold, sensitivity and specificity are presented. FINDINGS The ADAP ROC curves of the four autoantibodies showed comparable AUC in the two ADAP laboratories and were higher than RBA. Detection of two or more autoantibodies using chisq showed 0.97 (0.95, 0.99) sensitivity and 0.94 (0.91, 0.97) specificity in ADAP compared to 0.90 (0.88, 0.95) sensitivity and 0.97 (0.94, 0.98) specificity in RBA. Using Spec98, ADAP showed 0.92 (0.89, 0.95) sensitivity and 0.99 (0.98, 1.00) specificity compared to 0.89 (0.77, 0.86) sensitivity and 1.00 (0.99, 1.00) specificity in the RBA. The diagnostic sensitivity and specificity were higher in PBC compared to DOC and BDC. INTERPRETATION ADAP was comparable in two laboratories, both comparable to or better than RBA, to define threshold levels for two or more autoantibodies to stage type 1 diabetes. FUNDING Supported by The Leona M. and Harry B. Helmsley Charitable Trust (grant number 2009-04078), the Swedish Foundation for Strategic Research (Dnr IRC15-0067) and the Swedish Research Council, Strategic Research Area (Dnr 2009-1039). AL was supported by the DiaUnion collaborative study, co-financed by EU Interreg ÖKS, Capital Region of Denmark, Region Skåne and the Novo Nordisk Foundation.
Collapse
Affiliation(s)
- Alexander Lind
- Department of Clinical Sciences, Lund University CRC, Malmö, Sweden
| | - Eva Freyhult
- Department of Cell and Molecular Biology, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | | | - Anita Ramelius
- Department of Clinical Sciences, Lund University CRC, Malmö, Sweden
| | - Rasmus Bennet
- Department of Clinical Sciences, Lund University CRC, Malmö, Sweden
| | | | - David Seftel
- Enable Biosciences Inc., South San Francisco, CA, USA
| | - David Gebhart
- Enable Biosciences Inc., South San Francisco, CA, USA
| | | | - Marlena Maziarz
- Department of Clinical Sciences, Lund University CRC, Malmö, Sweden
| | | | - Markus Lundgren
- Department of Clinical Sciences, Lund University CRC, Malmö, Sweden
| | | | | | - Malin Fex
- Department of Clinical Sciences, Lund University CRC, Malmö, Sweden
| | - Carina Törn
- Department of Clinical Sciences, Lund University CRC, Malmö, Sweden
| | - Daniel Agardh
- Department of Clinical Sciences, Lund University CRC, Malmö, Sweden
| | | | - Åke Lernmark
- Department of Clinical Sciences, Lund University CRC, Malmö, Sweden.
| |
Collapse
|
14
|
Felton JL, Redondo MJ, Oram RA, Speake C, Long SA, Onengut-Gumuscu S, Rich SS, Monaco GSF, Harris-Kawano A, Perez D, Saeed Z, Hoag B, Jain R, Evans-Molina C, DiMeglio LA, Ismail HM, Dabelea D, Johnson RK, Urazbayeva M, Wentworth JM, Griffin KJ, Sims EK. Islet autoantibodies as precision diagnostic tools to characterize heterogeneity in type 1 diabetes: a systematic review. COMMUNICATIONS MEDICINE 2024; 4:66. [PMID: 38582818 PMCID: PMC10998887 DOI: 10.1038/s43856-024-00478-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 03/05/2024] [Indexed: 04/08/2024] Open
Abstract
BACKGROUND Islet autoantibodies form the foundation for type 1 diabetes (T1D) diagnosis and staging, but heterogeneity exists in T1D development and presentation. We hypothesized that autoantibodies can identify heterogeneity before, at, and after T1D diagnosis, and in response to disease-modifying therapies. METHODS We systematically reviewed PubMed and EMBASE databases (6/14/2022) assessing 10 years of original research examining relationships between autoantibodies and heterogeneity before, at, after diagnosis, and in response to disease-modifying therapies in individuals at-risk or within 1 year of T1D diagnosis. A critical appraisal checklist tool for cohort studies was modified and used for risk of bias assessment. RESULTS Here we show that 152 studies that met extraction criteria most commonly characterized heterogeneity before diagnosis (91/152). Autoantibody type/target was most frequently examined, followed by autoantibody number. Recurring themes included correlations of autoantibody number, type, and titers with progression, differing phenotypes based on order of autoantibody seroconversion, and interactions with age and genetics. Only 44% specifically described autoantibody assay standardization program participation. CONCLUSIONS Current evidence most strongly supports the application of autoantibody features to more precisely define T1D before diagnosis. Our findings support continued use of pre-clinical staging paradigms based on autoantibody number and suggest that additional autoantibody features, particularly in relation to age and genetic risk, could offer more precise stratification. To improve reproducibility and applicability of autoantibody-based precision medicine in T1D, we propose a methods checklist for islet autoantibody-based manuscripts which includes use of precision medicine MeSH terms and participation in autoantibody standardization workshops.
Collapse
Affiliation(s)
- Jamie L Felton
- Department of Pediatrics, Center for Diabetes and Metabolic Diseases, Indianapolis, IN, USA
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Maria J Redondo
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Division of Pediatric Diabetes and Endocrinology, Texas Children's Hospital, Houston, TX, USA
| | - Richard A Oram
- NIHR Exeter Biomedical Research Centre (BRC), Academic Kidney Unit, University of Exeter, Exeter, UK
- Department of Clinical and Biomedical Sciences, University of Exeter Medical School, Exeter, UK
- Royal Devon University Healthcare NHS Foundation Trust, Exeter, UK
| | - Cate Speake
- Center for Interventional Immunology, Benaroya Research Institute, Seattle, WA, USA
| | - S Alice Long
- Center for Translational Immunology, Benaroya Research Institute, Seattle, WA, USA
| | - Suna Onengut-Gumuscu
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Gabriela S F Monaco
- Department of Pediatrics, Center for Diabetes and Metabolic Diseases, Indianapolis, IN, USA
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Arianna Harris-Kawano
- Department of Pediatrics, Center for Diabetes and Metabolic Diseases, Indianapolis, IN, USA
| | - Dianna Perez
- Department of Pediatrics, Center for Diabetes and Metabolic Diseases, Indianapolis, IN, USA
| | - Zeb Saeed
- Department of Endocrinology, Diabetes and Metabolism, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Benjamin Hoag
- Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD, USA
| | - Rashmi Jain
- Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD, USA
| | - Carmella Evans-Molina
- Department of Pediatrics, Center for Diabetes and Metabolic Diseases, Indianapolis, IN, USA
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Endocrinology, Diabetes and Metabolism, Indiana University School of Medicine, Indianapolis, IN, USA
- Richard L. Roudebush VAMC, Indianapolis, IN, USA
| | - Linda A DiMeglio
- Department of Pediatrics, Center for Diabetes and Metabolic Diseases, Indianapolis, IN, USA
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Heba M Ismail
- Department of Pediatrics, Center for Diabetes and Metabolic Diseases, Indianapolis, IN, USA
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Dana Dabelea
- Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, Aurora, CO, USA
| | - Randi K Johnson
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Epidemiology, Colorado School of Public Health, Aurora, CO, USA
| | | | - John M Wentworth
- Royal Melbourne Hospital Department of Diabetes and Endocrinology, Parkville, VIC, Australia
- Walter and Eliza Hall Institute, Parkville, VIC, Australia
- University of Melbourne Department of Medicine, Parkville, VIC, Australia
| | - Kurt J Griffin
- Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD, USA
- Sanford Research, Sioux Falls, SD, USA
| | - Emily K Sims
- Department of Pediatrics, Center for Diabetes and Metabolic Diseases, Indianapolis, IN, USA.
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
15
|
Bruzzaniti S, Piemonte E, Bruzzese D, Lepore MT, Strollo R, Izzo L, Di Candia F, Franzese A, Bifulco M, Mozzillo E, Ludvigsson J, Matarese G, Galgani M. Progression of type 1 diabetes is associated with high levels of soluble PD-1 in islet autoantibody-positive children. Diabetologia 2024; 67:714-723. [PMID: 38214712 PMCID: PMC10904438 DOI: 10.1007/s00125-023-06075-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 11/27/2023] [Indexed: 01/13/2024]
Abstract
AIMS/HYPOTHESIS Type 1 diabetes is an autoimmune disorder that is characterised by destruction of pancreatic beta cells by autoreactive T lymphocytes. Although islet autoantibodies (AAb) are an indicator of disease progression, specific immune biomarkers that can be used as target molecules to halt development of type 1 diabetes have not been discovered. Soluble immune checkpoint molecules (sICM) play a pivotal role in counteracting excessive lymphocyte responses, but their role in type 1 diabetes is unexplored. In this longitudinal study, we measured sICM levels in AAb-positive (AAb+) children to identify molecules related to type 1 diabetes progression. METHODS We measured the levels of 14 sICM in the sera of AAb+ children (n=57) compared to those with recent-onset type 1 diabetes (n=79) and healthy children (n=44), obtained from two cohorts. AAb+ children were followed up and divided based on their progression to type 1 diabetes (AAbP) or not (AAbNP) (if they lost islet autoimmunity and did not develop disease in subsequent years). sICM were also measured in the sample taken at the visit closest to disease onset in AAbP children. RESULTS We found that AAb+ children had a distinct sICM profile compared with healthy children and those with recent-onset type 1 diabetes. In addition, AAb+ children who progressed to type 1 diabetes (AAbP) had higher sICM concentrations than non-progressors (AAbNP). Further, sICM levels decreased in AAbP children close to disease onset. Application of Cox regression models highlighted that high concentrations of soluble programmed cell death protein 1 (sPD-1) are associated with type 1 diabetes progression (HR 1.71; 95% CI 1.16, 2.51; p=0.007). CONCLUSIONS/INTERPRETATION This study reveals an sICM profile that is dysregulated during the preclinical stage of type 1 diabetes, and identifies sPD-1 as a pathophysiologically-relevant molecule that is associated with disease progression, offering a potential target for early interventions in autoimmune diabetes.
Collapse
Affiliation(s)
- Sara Bruzzaniti
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale 'G. Salvatore', Consiglio Nazionale delle Ricerche, Naples, Italy
| | - Erica Piemonte
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli 'Federico II', Naples, Italy
| | - Dario Bruzzese
- Dipartimento di Sanità Pubblica, Università degli Studi di Napoli 'Federico II', Naples, Italy
| | - Maria Teresa Lepore
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale 'G. Salvatore', Consiglio Nazionale delle Ricerche, Naples, Italy
| | - Rocky Strollo
- Dipartimento di Scienze Umane e Promozione della Qualità della Vita, Università Telematica San Raffaele Roma, Rome, Italy
| | - Lavinia Izzo
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli 'Federico II', Naples, Italy
| | - Francesca Di Candia
- Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli 'Federico II', Naples, Italy
| | - Adriana Franzese
- Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli 'Federico II', Naples, Italy
| | - Maurizio Bifulco
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli 'Federico II', Naples, Italy
| | - Enza Mozzillo
- Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli 'Federico II', Naples, Italy
| | - Johnny Ludvigsson
- Crown Princess Victoria's Children's Hospital and Division of Pediatrics, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden.
| | - Giuseppe Matarese
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale 'G. Salvatore', Consiglio Nazionale delle Ricerche, Naples, Italy
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli 'Federico II', Naples, Italy
| | - Mario Galgani
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale 'G. Salvatore', Consiglio Nazionale delle Ricerche, Naples, Italy.
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli 'Federico II', Naples, Italy.
| |
Collapse
|
16
|
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
|
17
|
Frohnert BI, Ghalwash M, Li Y, Ng K, Dunne JL, Lundgren M, Hagopian W, Lou O, Winkler C, Toppari J, Veijola R, Anand V. Refining the Definition of Stage 1 Type 1 Diabetes: An Ontology-Driven Analysis of the Heterogeneity of Multiple Islet Autoimmunity. Diabetes Care 2023; 46:1753-1761. [PMID: 36862942 PMCID: PMC10516254 DOI: 10.2337/dc22-1960] [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: 10/07/2022] [Accepted: 01/30/2023] [Indexed: 03/04/2023]
Abstract
OBJECTIVE To estimate the risk of progression to stage 3 type 1 diabetes based on varying definitions of multiple islet autoantibody positivity (mIA). RESEARCH DESIGN AND METHODS Type 1 Diabetes Intelligence (T1DI) is a combined prospective data set of children from Finland, Germany, Sweden, and the U.S. who have an increased genetic risk for type 1 diabetes. Analysis included 16,709 infants-toddlers enrolled by age 2.5 years and comparison between groups using Kaplan-Meier survival analysis. RESULTS Of 865 (5%) children with mIA, 537 (62%) progressed to type 1 diabetes. The 15-year cumulative incidence of diabetes varied from the most stringent definition (mIA/Persistent/2: two or more islet autoantibodies positive at the same visit with two or more antibodies persistent at next visit; 88% [95% CI 85-92%]) to the least stringent (mIA/Any: positivity for two islet autoantibodies without co-occurring positivity or persistence; 18% [5-40%]). Progression in mIA/Persistent/2 was significantly higher than all other groups (P < 0.0001). Intermediate stringency definitions showed intermediate risk and were significantly different than mIA/Any (P < 0.05); however, differences waned over the 2-year follow-up among those who did not subsequently reach higher stringency. Among mIA/Persistent/2 individuals with three autoantibodies, loss of one autoantibody by the 2-year follow-up was associated with accelerated progression. Age was significantly associated with time from seroconversion to mIA/Persistent/2 status and mIA to stage 3 type 1 diabetes. CONCLUSIONS The 15-year risk of progression to type 1 diabetes risk varies markedly from 18 to 88% based on the stringency of mIA definition. While initial categorization identifies highest-risk individuals, short-term follow-up over 2 years may help stratify evolving risk, especially for those with less stringent definitions of mIA.
Collapse
Affiliation(s)
| | - Mohamed Ghalwash
- Center for Computational Health at IBM Research at IBM T.J. Watson Research Center, Yorktown Heights, NY
- Ain Shams University, Cairo, Egypt
| | - Ying Li
- Center for Computational Health at IBM Research at IBM T.J. Watson Research Center, Yorktown Heights, NY
| | - Kenney Ng
- Center for Computational Health at IBM Research at IBM T.J. Watson Research Center, Cambridge, MA
| | | | - Markus Lundgren
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
- Department of Pediatrics, Kristianstad Hospital, Kristianstad, Sweden
| | | | | | - Christiane Winkler
- Institute of Diabetes Research, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Munich, Germany
- Forschergruppe Diabetes e.V. at Helmholtz Zentrum, Munich, Germany
| | - Jorma Toppari
- Institute of Biomedicine and Population Research Centre, University of Turku and Department of Pediatrics, Turku University Hospital, Turku, Finland
| | - Riitta Veijola
- Department of Pediatrics, PEDEGO Research Unit, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Vibha Anand
- Center for Computational Health at IBM Research at IBM T.J. Watson Research Center, Cambridge, MA
| |
Collapse
|
18
|
Limbert C, von dem Berge T, Danne T. Personalizing Early-Stage Type 1 Diabetes in Children. Diabetes Care 2023; 46:1747-1749. [PMID: 37729506 DOI: 10.2337/dci23-0025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 07/13/2023] [Indexed: 09/22/2023]
Affiliation(s)
- Catarina Limbert
- Unit of Paediatric Endocrinology and Diabetes, Hospital Dona Estefânia, Lisbon, Portugal
- Comprehensive Health Research Centre, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
| | | | - Thomas Danne
- Children's Hospital AUF DER BULT, Hannover, Germany
- Hannover Medical School, Hannover, Germany
| |
Collapse
|
19
|
Papadimitriou DT, Dermitzaki E, Christopoulos P, Papagianni M, Kleanthous K, Marakaki C, Papadimitriou A, Mastorakos G. Secondary Prevention of Diabetes Type 1 with Oral Calcitriol and Analogs, the PRECAL Study. CHILDREN (BASEL, SWITZERLAND) 2023; 10:children10050862. [PMID: 37238410 DOI: 10.3390/children10050862] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 05/04/2023] [Accepted: 05/10/2023] [Indexed: 05/28/2023]
Abstract
Screening for Type 1 Diabetes (T1D, incidence 1:300) with T1D autoantibodies (T1Ab) at ages 2 and 6, while sensitive, lacks a preventive strategy. Cholecalciferol 2000 IU daily since birth reduced T1D by 80% at 1 year. T1D-associated T1Ab negativized within 0.6 years with oral calcitriol in 12 children. To further investigate secondary prevention of T1D with calcitriol and its less calcemic analog, paricalcitol, we initiated a prospective interventional non-randomized clinical trial, the PRECAL study (ISRCTN17354692). In total, 50 high-risk children were included: 44 were positive for T1Ab, and 6 had predisposing for T1D HLA genotypes. Nine T1Ab+ patients had variable impaired glucose tolerance (IGT), four had pre-T1D (3 T1Ab+, 1 HLA+), nine had T1Ab+ new-onset T1D not requiring insulin at diagnosis. T1Ab, thyroid/anti-transglutaminase Abs, glucose/calcium metabolism were determined prior and q3-6 months on calcitriol, 0.05 mcg/Kg/day, or paricalcitol 1-4 mcg × 1-3 times/day p.o. while on cholecalciferol repletion. Available data on 42 (7 dropouts, 1 follow-up < 3 months) patients included: all 26 without pre-T1D/T1D followed for 3.06 (0.5-10) years negativized T1Ab (15 +IAA, 3 IA2, 4 ICA, 2 +GAD, 1 +IAA/+GAD, 1 +ICA/+GAD) within 0.57 (0.32-1.3) years or did not develop to T1D (5 +HLA, follow-up 3 (1-4) years). From four pre-T1D cases, one negativized T1Ab (follow-up 1 year), one +HLA did not progress to T1D (follow-up 3.3 years) and two +T1Ab patients developed T1D in 6 months/3 years. Three out of nine T1D cases progressed immediately to overt disease, six underwent complete remission for 1 year (1 month-2 years). Five +T1Ab patients relapsed and negativized again after resuming therapy. Four (aged <3 years) negativized anti-TPO/TG, and two anti-transglutaminase-IgA. Eight presented mild hypercalciuria/hypercalcemia, resolving with dose titration/discontinuation. Secondary prevention of T1D with calcitriol and paricalcitol seems possible and reasonably safe, if started soon enough after seroconversion.
Collapse
Affiliation(s)
- Dimitrios T Papadimitriou
- Second Department of Obstetrics and Gynecology, Aretaieion University Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Department of Pediatric-Adolescent Endocrinology and Diabetes, Athens Medical Center, 15125 Marousi, Greece
| | - Eleni Dermitzaki
- Department of Pediatric-Adolescent Endocrinology and Diabetes, Athens Medical Center, 15125 Marousi, Greece
| | - Panagiotis Christopoulos
- Second Department of Obstetrics and Gynecology, Aretaieion University Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Maria Papagianni
- Department of Nutrition and Dietetics, University of Thessaly, 42132 Trikala, Greece
- Unit of Endocrinology, Diabetes and Metabolism, Third Department of Pediatrics, Aristotle University of Thessaloniki, Hippokrateion Hospital of Thessaloniki, 54642 Thessaloniki, Greece
| | - Kleanthis Kleanthous
- Department of Pediatric-Adolescent Endocrinology and Diabetes, Athens Medical Center, 15125 Marousi, Greece
| | - Chrysanthi Marakaki
- Department of Pediatric-Adolescent Endocrinology and Diabetes, Athens Medical Center, 15125 Marousi, Greece
| | - Anastasios Papadimitriou
- Pediatric Endocrinology Unit, Attikon University Hospital, National and Kapodistrian University of Athens, 12462 Haidari, Greece
| | - George Mastorakos
- Second Department of Obstetrics and Gynecology, Aretaieion University Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
20
|
Elhassan S, Dong F, Buckner T, Johnson RK, Seifert JA, Carry PM, Vanderlinden L, Waugh K, Rewers M, Norris JM. Investigating iron intake in risk of progression from islet autoimmunity to type 1 diabetes: The diabetes autoimmunity study in the young. Front Immunol 2023; 14:1124370. [PMID: 37056761 PMCID: PMC10086157 DOI: 10.3389/fimmu.2023.1124370] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 03/15/2023] [Indexed: 03/30/2023] Open
Abstract
Background Studies of the role of iron in the risk of type 1 diabetes (T1D) have been inconsistent. Given that iron generates reactive oxygen radicals, which can lead to oxidative damage and apoptosis in the beta cells of the pancreas, we examined whether iron intake was associated with the risk of progressing to T1D in individuals with islet autoimmunity (IA), the pre-clinical phase of T1D. Methods DAISY is a prospective cohort following 2,547 children at increased risk for IA and progression to T1D. IA is defined as at least two consecutive serum samples positive for at least one autoantibody (insulin, GAD, IA-2, or ZnT8). We measured dietary intake at the time of IA seroconversion in 175 children with IA, and of these, 64 progressed to T1D. We used Cox regression to examine the association between energy-adjusted iron intake and progression to T1D, adjusting for HLA-DR3/4 genotype, race/ethnicity, age at seroconversion, presence of multiple autoantibodies at seroconversion, and multiple vitamin use. In addition, we tested whether this association was modified by vitamin C or calcium intake. Results In children with IA, high iron intake (as defined as above the 75th percentile, > 20.3 mg/day) was associated with decreased risk of progression to T1D compared to moderate iron intake (as defined by the middle 25-75th percentiles, 12.7-20.3 mg/day) (adjusted hazard ratio (HR): 0.35; 95% confidence interval (CI): 0.15, 0.79). The association between iron intake and T1D was not modified by vitamin C nor calcium intake. In a sensitivity analysis, the removal of six children who had been diagnosed with celiac disease prior to IA seroconversion did not affect this association. Conclusion Higher iron intake at the time of IA seroconversion is associated with a lower risk of progression to T1D, independent of multivitamin supplement use. Further research that includes plasma biomarkers of iron status is needed to investigate the relationship between iron and the risk of T1D.
Collapse
Affiliation(s)
- Sulafa Elhassan
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Fran Dong
- Barbara Davis Center for Diabetes, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Teresa Buckner
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Department of Kinesiology, Nutrition, and Dietetics, University of Northern Colorado, Greeley, CO, United States
| | - Randi K. Johnson
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Department of Biomedical Informatics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Jennifer A. Seifert
- Department of Medicine, Division of Rheumatology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Patrick M. Carry
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Colorado Program for Musculoskeletal Research, Department of Orthopedics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Lauren Vanderlinden
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Kathleen Waugh
- Barbara Davis Center for Diabetes, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Marian Rewers
- Barbara Davis Center for Diabetes, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Jill M. Norris
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
21
|
Kwon BC, Achenbach P, Anand V, Frohnert BI, Hagopian W, Hu J, Koski E, Lernmark Å, Lou O, Martin F, Ng K, Toppari J, Veijola R. Islet Autoantibody Levels Differentiate Progression Trajectories in Individuals With Presymptomatic Type 1 Diabetes. Diabetes 2022; 71:2632-2641. [PMID: 36112006 PMCID: PMC9750947 DOI: 10.2337/db22-0360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 08/29/2022] [Indexed: 01/24/2023]
Abstract
In our previous data-driven analysis of evolving patterns of islet autoantibodies (IAb) against insulin (IAA), GAD (GADA), and islet antigen 2 (IA-2A), we discovered three trajectories, characterized according to multiple IAb (TR1), IAA (TR2), or GADA (TR3) as the first appearing autoantibodies. Here we examined the evolution of IAb levels within these trajectories in 2,145 IAb-positive participants followed from early life and compared those who progressed to type 1 diabetes (n = 643) with those remaining undiagnosed (n = 1,502). With use of thresholds determined by 5-year diabetes risk, four levels were defined for each IAb and overlaid onto each visit. In diagnosed participants, high IAA levels were seen in TR1 and TR2 at ages <3 years, whereas IAA remained at lower levels in the undiagnosed. Proportions of dwell times (total duration of follow-up at a given level) at the four IAb levels differed between the diagnosed and undiagnosed for GADA and IA-2A in all three trajectories (P < 0.001), but for IAA dwell times differed only within TR2 (P < 0.05). Overall, undiagnosed participants more frequently had low IAb levels and later appearance of IAb than diagnosed participants. In conclusion, while it has long been appreciated that the number of autoantibodies is an important predictor of type 1 diabetes, consideration of autoantibody levels within the three autoimmune trajectories improved differentiation of IAb-positive children who progressed to type 1 diabetes from those who did not.
Collapse
Affiliation(s)
- Bum Chul Kwon
- Center for Computational Health, IBM Research, Cambridge, MA
- Corresponding author: Bum Chul Kwon,
| | - Peter Achenbach
- Institute of Diabetes Research, Helmholtz Zentrum München—German Research Center for Environmental Health, Munich-Neuherberg, Germany
| | - Vibha Anand
- Center for Computational Health, IBM Research, Cambridge, MA
| | | | | | - Jianying Hu
- Center for Computational Health, IBM Research, Yorktown Heights, NY
| | - Eileen Koski
- Center for Computational Health, IBM Research, Yorktown Heights, NY
| | - Åke Lernmark
- Department of Clinical Sciences Malmö, Lund University CRC, Skåne University Hospital, Malmö, Sweden
| | | | | | - Kenney Ng
- Center for Computational Health, IBM Research, Cambridge, MA
| | - Jorma Toppari
- Institute of Biomedicine and Centre for Population Health Research, University of Turku, and Department of Pediatrics, Turku University Hospital, Turku, Finland
| | - Riitta Veijola
- Medical Research Center, PEDEGO Research Unit, Department of Pediatrics, University of Oulu and Oulu University Hospital, Oulu, Finland
| |
Collapse
|
22
|
Helminen O, Pokka T, Aspholm S, Ilonen J, Simell O, Knip M, Veijola R. Early glucose metabolism in children at risk for type 1 diabetes based on islet autoantibodies compared to low-risk control groups. Front Endocrinol (Lausanne) 2022; 13:972714. [PMID: 36171903 PMCID: PMC9511031 DOI: 10.3389/fendo.2022.972714] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Accepted: 08/19/2022] [Indexed: 11/16/2022] Open
Abstract
Background Anatomic variation or early differences in glucose metabolism have been linked to the development of type 1 diabetes. We aimed to describe early glucose metabolism based on HbA1c, oral glucose tolerance test (OGTT), and random plasma glucose years before the presentation of type 1 diabetes in five risk groups based on autoantibody combinations. For the first time, we were able to include for comparison children with very low risk of progression to type 1 diabetes. Methods The Finnish Diabetes Prediction and Prevention birth cohort study screened newborn infants for HLA susceptibility to type 1 diabetes since 1994. Those carrying a risk genotype were prospectively followed up with islet autoantibody testing. Glucose parameters were obtained starting from the time of seroconversion. By 31 August 2014, 1162 children had developed at least one islet autoantibody and were included in the current study. Type 1 diabetes was diagnosed in 335 children (progressors). In the non-progressor groups, 207 developed multiple (≥2) biochemical islet autoantibodies, 229 a single biochemical autoantibody, 370 ICA only, and 64 transient autoantibodies. Children were divided into five risk groups. Glucose metabolism was evaluated. Results We observed lower HbA1c values in early follow-up 4.5 to 6.0 years before diagnosis in the progressors when compared to the same time in children with a single biochemical autoantibody or low-risk (ICA only and transient) participants, who did not progress to clinical type 1 diabetes. However, no such differences were observed in OGTTs or random plasma glucose. The variation was minimal in glucose values in the low-risk groups. Conclusion We report the possibility of early alteration in glucose metabolism in future progressors. This could suggest early defects in multiple glucose-regulating hormones.
Collapse
Affiliation(s)
- Olli Helminen
- Department of Pediatrics, PEDEGO Research Group, Medical Research Center, Oulu University, Hospital and University of Oulu, Oulu, Finland
- Surgery Research Unit, Cancer and Translational Medicine Research Unit, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Tytti Pokka
- Department of Pediatrics, PEDEGO Research Group, Medical Research Center, Oulu University, Hospital and University of Oulu, Oulu, Finland
| | - Susanna Aspholm
- Tampere Centre for Child Health Research, Tampere University Hospital, Tampere, Finland
| | - Jorma Ilonen
- Immunogenetics Laboratory, University of Turku, Turku, Finland
- Department of Pediatrics, University of Turku and Turku University Hospital, Turku, Finland
| | - Olli Simell
- Department of Pediatrics, University of Turku and Turku University Hospital, Turku, Finland
| | - Mikael Knip
- Tampere Centre for Child Health Research, Tampere University Hospital, Tampere, Finland
- Pediatric Research Center, New Children’s Hospital, Helsinki University Hospital, Helsinki, Finland
- Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Riitta Veijola
- Department of Pediatrics, PEDEGO Research Group, Medical Research Center, Oulu University, Hospital and University of Oulu, Oulu, Finland
| |
Collapse
|
23
|
Zhang X, Dong Y, Liu D, Yang L, Xu J, Wang Q. Antigen-specific immunotherapies in type 1 diabetes. J Trace Elem Med Biol 2022; 73:127040. [PMID: 35868165 DOI: 10.1016/j.jtemb.2022.127040] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 06/18/2022] [Accepted: 07/14/2022] [Indexed: 11/16/2022]
Abstract
Type 1 diabetes mellitus (T1DM) is an autoimmune disease caused by the destruction of pancreatic beta cells, in which immune system disorder plays an important role. Finding a cure for T1DM and restoring beta cell function has been a long-standing goal. Research has shown that immune regulation with pancreatic islet auto-antigens may be the most specific and safe treatment for T1DM. Immunological intervention using diabetogenic auto-antigens as a target can help identify T1DM in high-risk individuals by early screening of autoantibodies (AAbs) before the loss of pancreatic islet function and thus achieve primary prevention of T1DM. However, induction of self-tolerance in patients with pre-diabetes can also slow down the attack of autoimmunity, and achieve secondary prevention. Antigen-based immune therapy opens up new avenues for the prevention and treatment of T1DM. The zinc transporter 8 (ZnT8) protein, presents in the serum of pre-diabetic and diabetic patients, is immunogenic and can cause T1D autoimmune responses. ZnT8 has become a potential target of humoral autoimmunity; it is of great significance for the early diagnosis of T1D. ZnT8-specific CD8+ T cells can be detected in most T1DM patients, and play a key role in the progression of T1D. As an immunotherapy target, it can improve the dysfunction of beta cells in T1DM and provide new ideas for the treatment of T1D. In this review, we summarize research surrounding antigen-specific immunotherapies (ASI) over the past 10 years and the ZnT8 antigen as an autoimmune target to induce self-tolerance for T1DM.
Collapse
Affiliation(s)
- Xuejiao Zhang
- Department of Endocrinology, China-Japan Union Hospital of Jilin University, Changchun 130000, China
| | - Ying Dong
- Department of Radiation Oncology, Jilin Cancer Hospital, Changchun 130000, China
| | - Dianyuan Liu
- Department of Endocrinology, China-Japan Union Hospital of Jilin University, Changchun 130000, China
| | - Liu Yang
- Department of Endocrinology, China-Japan Union Hospital of Jilin University, Changchun 130000, China
| | - Jiayi Xu
- School of Public Health, Jilin University, Changchun 130000, China
| | - Qing Wang
- Department of Endocrinology, China-Japan Union Hospital of Jilin University, Changchun 130000, China.
| |
Collapse
|
24
|
Choi R, Park W, Chun G, Lee J, Lee SG, Lee EH. Recent information on test utilization and intraindividual change in anti-glutamic acid decarboxylase antibody in Korea: a retrospective study. BMJ Open Diabetes Res Care 2022; 10:10/3/e002739. [PMID: 35750359 PMCID: PMC9234797 DOI: 10.1136/bmjdrc-2021-002739] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 05/24/2022] [Indexed: 11/04/2022] Open
Abstract
INTRODUCTION This study aimed to evaluate the test utilization and intraindividual changes of anti-glutamic acid decarboxylase antibody (GADA), a biomarker for autoimmune diabetes in Korean adults. RESEARCH DESIGN AND METHODS We retrospectively investigated longitudinally measured GADA test results to assess test utilization and intraindividual changes through a laboratory information system. RESULTS During the 3-year study period, 11 668 GADA tests were performed in 11 184 Korean adults. The overall rate of GADA positivity at initial measurement was 7.8%. Among the 11 668 test results, 871 GADA test results from 401 Korean patients (228 men and 173 women) requested by 54 hospitals were analyzed for intraindividual changes. Among these 401 patients, 80 (20.0%) had positive (≥2.0 U/mL) and 35 (8.7%) had gray zone GADA (1.0-1.9 U/mL) level at initial measurement. The prevalence of GADA-positive patients based on initial measurement was significantly different by type of medical institution. Among 80 patients with initial positive results, 5 (6.3%) experienced qualitative GADA changes during follow-up. Among the 321 patients with initially negative or gray zone GADA, 9 (2.8%) changed to GADA positive at least once during follow-up. CONCLUSIONS Although most patients had stable GADA results, some exhibited qualitative changes during follow-up. This study can help to understand the variation in GADA positivity in the monitored patients.
Collapse
Affiliation(s)
- Rihwa Choi
- Department of Laboratory Medicine, Green Cross Laboratories, Yongin-si, Republic of Korea
- Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Gangnam-gu, Republic of Korea
| | - Wonseo Park
- Infectious Disease Research Center, Green Cross Laboratories, Yongin-si, Republic of Korea
| | - Gayoung Chun
- Infectious Disease Research Center, Green Cross Laboratories, Yongin-si, Republic of Korea
| | - Jiwon Lee
- Department of Laboratory Medicine, Green Cross Laboratories, Yongin-si, Republic of Korea
| | - Sang Gon Lee
- Department of Laboratory Medicine, Green Cross Laboratories, Yongin-si, Republic of Korea
| | - Eun Hee Lee
- Green Cross Laboratories, Yongin-si, Republic of Korea
| |
Collapse
|
25
|
Progression of type 1 diabetes from latency to symptomatic disease is predicted by distinct autoimmune trajectories. Nat Commun 2022; 13:1514. [PMID: 35314671 PMCID: PMC8938551 DOI: 10.1038/s41467-022-28909-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 02/16/2022] [Indexed: 12/13/2022] Open
Abstract
Development of islet autoimmunity precedes the onset of type 1 diabetes in children, however, the presence of autoantibodies does not necessarily lead to manifest disease and the onset of clinical symptoms is hard to predict. Here we show, by longitudinal sampling of islet autoantibodies (IAb) to insulin, glutamic acid decarboxylase and islet antigen-2 that disease progression follows distinct trajectories. Of the combined Type 1 Data Intelligence cohort of 24662 participants, 2172 individuals fulfill the criteria of two or more follow-up visits and IAb positivity at least once, with 652 progressing to type 1 diabetes during the 15 years course of the study. Our Continuous-Time Hidden Markov Models, that are developed to discover and visualize latent states based on the collected data and clinical characteristics of the patients, show that the health state of participants progresses from 11 distinct latent states as per three trajectories (TR1, TR2 and TR3), with associated 5-year cumulative diabetes-free survival of 40% (95% confidence interval [CI], 35% to 47%), 62% (95% CI, 57% to 67%), and 88% (95% CI, 85% to 91%), respectively (p < 0.0001). Age, sex, and HLA-DR status further refine the progression rates within trajectories, enabling clinically useful prediction of disease onset. Presence of islet autoantibodies precedes the onset of type 1 diabetes but it does not predict whether and how fast symptomatic disease appears. Here authors present a model to predict and visualize progression to diabetes by using a large longitudinal data set on autoantibodies and clinical parameters as input.
Collapse
|
26
|
Korneva KG, Strongin LG, Kolbasina EV, Budylina MV, Makeeva NV, Zagainov VE. Diagnostic Capabilities of Islet Autoantibodies in Children with New-Onset Type 1 Diabetes Mellitus and Healthy Siblings. Sovrem Tekhnologii Med 2021; 12:29-34. [PMID: 34796016 PMCID: PMC8596233 DOI: 10.17691/stm2020.12.6.04] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Indexed: 12/16/2022] Open
Abstract
The aim of the study is to determine the diagnostic utility of several islet autoantibodies and their combinations in order to identify individuals susceptible to type 1 diabetes mellitus (T1DM) among healthy siblings in the pediatric population within the scope of the development of a screening program.
Collapse
Affiliation(s)
- K G Korneva
- Associate Professor, Department of Endocrinology and Internal Medicine; Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia
| | - L G Strongin
- Professor, Head of the Department of Endocrinology and Internal Medicine; Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia
| | - E V Kolbasina
- Pediatric Endocrinologist, Head of the Department of Endocrinology; Nizhny Novgorod Regional Children's Clinical Hospital, 211 Vaneeva St., Nizhny Novgorod, 603136, Russia
| | - M V Budylina
- Head of the Department of Pediatric Endocrinology and Gastroenterology; Republican Children's Clinical Hospital of the Ministry of Health of the Chuvash Republic, 27 Fedora Gladkova St., Cheboksary, 428020, Russia
| | - N V Makeeva
- Pediatric Endocrinologist, Chief Non-Staff Pediatric Endocrinologist; Children's Republican Clinical Hospital, 10 Meditsinskaya St., Yoshkar-Ola, the Republic of Mari El, 424005, Russia
| | - V E Zagainov
- Associate Professor, Head of the Department of Faculty Surgery and Transplantology Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia
| |
Collapse
|
27
|
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: 33] [Impact Index Per Article: 8.3] [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
|
28
|
Kahn SE, Chen YC, Esser N, Taylor AJ, van Raalte DH, Zraika S, Verchere CB. The β Cell in Diabetes: Integrating Biomarkers With Functional Measures. Endocr Rev 2021; 42:528-583. [PMID: 34180979 PMCID: PMC9115372 DOI: 10.1210/endrev/bnab021] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Indexed: 02/08/2023]
Abstract
The pathogenesis of hyperglycemia observed in most forms of diabetes is intimately tied to the islet β cell. Impairments in propeptide processing and secretory function, along with the loss of these vital cells, is demonstrable not only in those in whom the diagnosis is established but typically also in individuals who are at increased risk of developing the disease. Biomarkers are used to inform on the state of a biological process, pathological condition, or response to an intervention and are increasingly being used for predicting, diagnosing, and prognosticating disease. They are also proving to be of use in the different forms of diabetes in both research and clinical settings. This review focuses on the β cell, addressing the potential utility of genetic markers, circulating molecules, immune cell phenotyping, and imaging approaches as biomarkers of cellular function and loss of this critical cell. Further, we consider how these biomarkers complement the more long-established, dynamic, and often complex measurements of β-cell secretory function that themselves could be considered biomarkers.
Collapse
Affiliation(s)
- Steven E Kahn
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, VA Puget Sound Health Care System and University of Washington, Seattle, 98108 WA, USA
| | - Yi-Chun Chen
- BC Children's Hospital Research Institute and Centre for Molecular Medicine and Therapeutics, Vancouver, BC, V5Z 4H4, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada.,Department of Surgery, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada
| | - Nathalie Esser
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, VA Puget Sound Health Care System and University of Washington, Seattle, 98108 WA, USA
| | - Austin J Taylor
- BC Children's Hospital Research Institute and Centre for Molecular Medicine and Therapeutics, Vancouver, BC, V5Z 4H4, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada.,Department of Surgery, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada
| | - Daniël H van Raalte
- Department of Internal Medicine, Amsterdam University Medical Center (UMC), Vrije Universiteit (VU) University Medical Center, 1007 MB Amsterdam, The Netherlands.,Department of Experimental Vascular Medicine, Amsterdam University Medical Center (UMC), Academic Medical Center, 1007 MB Amsterdam, The Netherlands
| | - Sakeneh Zraika
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, VA Puget Sound Health Care System and University of Washington, Seattle, 98108 WA, USA
| | - C Bruce Verchere
- BC Children's Hospital Research Institute and Centre for Molecular Medicine and Therapeutics, Vancouver, BC, V5Z 4H4, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada.,Department of Surgery, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada
| |
Collapse
|
29
|
Carry PM, Vanderlinden LA, Johnson RK, Buckner T, Fiehn O, Steck AK, Kechris K, Yang I, Fingerlin TE, Rewers M, Norris JM. Phospholipid Levels at Seroconversion Are Associated With Resolution of Persistent Islet Autoimmunity: The Diabetes Autoimmunity Study in the Young. Diabetes 2021; 70:1592-1601. [PMID: 33863802 PMCID: PMC8336007 DOI: 10.2337/db20-1251] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 04/11/2021] [Indexed: 12/14/2022]
Abstract
Reversion of islet autoimmunity (IA) may point to mechanisms that prevent IA progression. We followed 199 individuals who developed IA during the Diabetes Autoimmunity Study in the Young. Untargeted metabolomics was performed in serum samples following IA. Cox proportional hazards models were used to test whether the metabolites (2,487) predicted IA reversion: two or more consecutive visits negative for all autoantibodies. We conducted a principal components analysis (PCA) of the top metabolites; |hazard ratio (HR) >1.25| and nominal P < 0.01. Phosphatidylcholine (16:0_18:1(9Z)) was the strongest individual metabolite (HR per 1 SD 2.16, false discovery rate (FDR)-adjusted P = 0.0037). Enrichment analysis identified four clusters (FDR P < 0.10) characterized by an overabundance of sphingomyelin (d40:0), phosphatidylcholine (16:0_18:1(9Z)), phosphatidylcholine (30:0), and l-decanoylcarnitine. Overall, 63 metabolites met the criteria for inclusion in the PCA. PC1 (HR 1.4, P < 0.0001), PC2 (HR 0.85, P = 0.0185), and PC4 (HR 1.28, P = 0.0103) were associated with IA reversion. Given the potential influence of diet on the metabolome, we investigated whether nutrients were correlated with PCs. We identified 20 nutrients that were correlated with the PCs (P < 0.05). Total sugar intake was the top nutrient. Overall, we identified an association between phosphatidylcholine, sphingomyelin, and carnitine levels and reversion of IA.
Collapse
Affiliation(s)
- Patrick M Carry
- Department of Epidemiology, Colorado School of Public Health, Aurora, CO
| | | | - Randi K Johnson
- Colorado Center for Personalized Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Teresa Buckner
- Department of Epidemiology, Colorado School of Public Health, Aurora, CO
| | | | - Andrea K Steck
- Barbara Davis Center for Diabetes, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Katerina Kechris
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, CO
| | - Ivana Yang
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Tasha E Fingerlin
- Department of Epidemiology, Colorado School of Public Health, Aurora, CO
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, CO
- Department of Immunology and Genomic Medicine, National Jewish Health, Denver, CO
| | - Marian Rewers
- Barbara Davis Center for Diabetes, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Jill M Norris
- Department of Epidemiology, Colorado School of Public Health, Aurora, CO
- Barbara Davis Center for Diabetes, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
30
|
Abstract
Life is about timing. -Carl LewisThe understanding of autoimmune type 1 diabetes is increasing, and examining etiology separate from pathogenesis has become crucial. The components to explain type 1 diabetes development have been known for some time. The strong association with HLA has been researched for nearly 50 years. Genome-wide association studies added another 60+ non-HLA genetic factors with minor contribution to risk. Insulitis has long been known to be present close to clinical diagnosis. T and B cells recognizing β-cell autoantigens are detectable prior to diagnosis and in newly diagnosed patients. Islet autoantibody tests against four major autoantigens have been standardized and used as biomarkers of islet autoimmunity. However, to clarify the etiology would require attention to time. Etiology may be defined as the cause of a disease (i.e., type 1 diabetes) or abnormal condition (i.e., islet autoimmunity). Timing is everything, as neither the prodrome of islet autoimmunity nor the clinical onset of type 1 diabetes tells us much about the etiology. Rather, the islet autoantibody that appears first and persists would mark the diagnosis of an autoimmune islet disease (AID). Events after the diagnosis of AID would represent the pathogenesis. Several islet autoantibodies without (stage 1) or with impaired glucose tolerance (stage 2) or with symptoms (stage 3) would define the pathogenesis culminating in clinical type 1 diabetes. Etiology would be about the timing of events that take place before the first-appearing islet autoantibody.
Collapse
Affiliation(s)
- Åke Lernmark
- Department of Clinical Sciences, Lund University Clinical Research Centre, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
31
|
Salami F, N.Tamura R, Elding Larsson H, Lernmark Å, Törn C. Complete blood counts with red blood cell determinants associate with reduced beta-cell function in seroconverted Swedish TEDDY children. Endocrinol Diabetes Metab 2021; 4:e00251. [PMID: 34277975 PMCID: PMC8279594 DOI: 10.1002/edm2.251] [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] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 03/01/2021] [Accepted: 03/22/2021] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVES To investigate whether changes in complete blood count (CBC) in islet autoantibody positive children with increased genetic risk for type 1 diabetes are associated with oral glucose tolerance tests (OGTT) and HbA1c over time. METHODS The Environmental Determinants of Diabetes in the Young (TEDDY) study follows children with increased risk for type 1 diabetes in the United States, Germany, Sweden and Finland. In the current study, 89 Swedish TEDDY children (median age 8.8 years) positive for one or multiple islet autoantibodies were followed up to 5 (median 2.3) years for CBC, OGTT and HbA1c. A statistical mixed effect model was used to investigate the association between CBC and OGTT or HbA1c. RESULTS HbA1c over time increased by the number of autoantibodies (p < .001). Reduction in mean corpuscular haemoglobin (MCH) and mean cell volume (MCV) was both associated with an increase in HbA1c (p < .001). A reduction in red blood cell (RBC) counts (p = .003), haemoglobin (p = .002) and haematocrit (p = .006) levels was associated with increased fasting glucose. Increased red blood cells, haemoglobin, haematocrit and MCH but decreased levels of red blood cell distribution widths (RDW) were all associated with increased fasting insulin. CONCLUSIONS The decrease in RBC indices with increasing HbA1c and the decrease in RBC and its parameters with increasing fasting glucose in seroconverted children may reflect an insidious deterioration in glucose metabolism associated with islet beta-cell autoimmunity.
Collapse
Affiliation(s)
- Falastin Salami
- Department of Clinical SciencesClinical Research CentreLund UniversitySkåne University HospitalMalmöSweden
| | - Roy N.Tamura
- Health Informatics InstituteDepartment of PediatricsUniversity of South FloridaTampaFloridaUSA
| | - Helena Elding Larsson
- Department of Clinical SciencesClinical Research CentreLund UniversitySkåne University HospitalMalmöSweden
| | - Åke Lernmark
- Department of Clinical SciencesClinical Research CentreLund UniversitySkåne University HospitalMalmöSweden
| | - Carina Törn
- Department of Clinical SciencesClinical Research CentreLund UniversitySkåne University HospitalMalmöSweden
| | | |
Collapse
|
32
|
Anand V, Li Y, Liu B, Ghalwash M, Koski E, Ng K, Dunne JL, Jönsson J, Winkler C, Knip M, Toppari J, Ilonen J, Killian MB, Frohnert BI, Lundgren M, Ziegler AG, Hagopian W, Veijola R, Rewers M. Islet Autoimmunity and HLA Markers of Presymptomatic and Clinical Type 1 Diabetes: Joint Analyses of Prospective Cohort Studies in Finland, Germany, Sweden, and the U.S. Diabetes Care 2021; 44:dc201836. [PMID: 34162665 PMCID: PMC8929180 DOI: 10.2337/dc20-1836] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 04/07/2021] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To combine prospective cohort studies, by including HLA harmonization, and estimate risk of islet autoimmunity and progression to clinical diabetes. RESEARCH DESIGN AND METHODS For prospective cohorts in Finland, Germany, Sweden, and the U.S., 24,662 children at increased genetic risk for development of islet autoantibodies and type 1 diabetes have been followed. Following harmonization, the outcomes were analyzed in 16,709 infants-toddlers enrolled by age 2.5 years. RESULTS In the infant-toddler cohort, 1,413 (8.5%) developed at least one autoantibody confirmed at two or more consecutive visits (seroconversion), 865 (5%) developed multiple autoantibodies, and 655 (4%) progressed to diabetes. The 15-year cumulative incidence of diabetes varied in children with one, two, or three autoantibodies at seroconversion: 45% (95% CI 40-52), 85% (78-90), and 92% (85-97), respectively. Among those with a single autoantibody, status 2 years after seroconversion predicted diabetes risk: 12% (10-25) if reverting to autoantibody negative, 30% (20-40) if retaining a single autoantibody, and 82% (80-95) if developing multiple autoantibodies. HLA-DR-DQ affected the risk of confirmed seroconversion and progression to diabetes in children with stable single-autoantibody status. Their 15-year diabetes incidence for higher- versus lower-risk genotypes was 40% (28-50) vs. 12% (5-38). The rate of progression to diabetes was inversely related to age at development of multiple autoantibodies, ranging from 20% per year to 6% per year in children developing multipositivity in ≤2 years or >7.4 years, respectively. CONCLUSIONS The number of islet autoantibodies at seroconversion reliably predicts 15-year type 1 diabetes risk. In children retaining a single autoantibody, HLA-DR-DQ genotypes can further refine risk of progression.
Collapse
Affiliation(s)
- Vibha Anand
- Center for Computational Health, IBM T.J. Watson Research Center, Cambridge, MA, and Yorktown Heights, NY
| | - Ying Li
- Center for Computational Health, IBM T.J. Watson Research Center, Cambridge, MA, and Yorktown Heights, NY
| | - Bin Liu
- Center for Computational Health, IBM T.J. Watson Research Center, Cambridge, MA, and Yorktown Heights, NY
| | - Mohamed Ghalwash
- Center for Computational Health, IBM T.J. Watson Research Center, Cambridge, MA, and Yorktown Heights, NY
- Ain Shams University, Cairo, Egypt
| | - Eileen Koski
- Center for Computational Health, IBM T.J. Watson Research Center, Cambridge, MA, and Yorktown Heights, NY
| | - Kenney Ng
- Center for Computational Health, IBM T.J. Watson Research Center, Cambridge, MA, and Yorktown Heights, NY
| | | | - Josefine Jönsson
- Department of Clinical Sciences Malmö, Lund University/CRC, Skåne University Hospital, Malmö
| | - Christiane Winkler
- Institute of Diabetes Research, Helmholtz Zentrum München German Research Center for Environmental Health, Munich-Neuherberg, Germany
- Forschergruppe Diabetes e.V. am Helmholtz Zentrum München, Munich, Germany
- Forschergruppe Diabetes, Technical University Munich, Germany
| | - Mikael Knip
- Tampere Center for Child Health Research, Tampere University Hospital, Tampere, Finland
- Pediatric Research Center, Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland
| | - Jorma Toppari
- Institute of Biomedicine and Population Research Centre, University of Turku, and Department of Pediatrics, Turku University Hospital, Turku, Finland
| | - Jorma Ilonen
- Immunogenetics Laboratory, Institute of Biomedicine, University of Turku, and Clinical Microbiology, Turku University Hospital, Turku, Finland
| | | | | | - Markus Lundgren
- Institute of Diabetes Research, Helmholtz Zentrum München German Research Center for Environmental Health, Munich-Neuherberg, Germany
| | - Anette-Gabriele Ziegler
- Institute of Diabetes Research, Helmholtz Zentrum München German Research Center for Environmental Health, Munich-Neuherberg, Germany
- Forschergruppe Diabetes e.V. am Helmholtz Zentrum München, Munich, Germany
- Forschergruppe Diabetes, Technical University Munich, Germany
| | | | - Riitta Veijola
- PEDEGO Research Unit, Department of Pediatrics, University of Oulu and Oulu University Hospital, Oulu, Finland
| | | |
Collapse
|
33
|
Gougourelas D, Tsentidis C, Koufadaki AM, Koutsovasilis A, Gougourela E, Karanasios S, Sotiropoulos A, Bousboulas S, Karavanaki KA. Associated autoimmunity in Type 1 Diabetes and latent autoimmune diabetes of adults: The role of glutamic-acid decarboxylase autoantibodies. Diabetes Res Clin Pract 2021; 175:108847. [PMID: 33945840 DOI: 10.1016/j.diabres.2021.108847] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 04/19/2021] [Accepted: 04/28/2021] [Indexed: 11/17/2022]
Abstract
AIMS To determine the prevalence of Associated Autoimmune Diseases (AADs) in Latent Autoimmune Diabetes of Adults (LADA) versus autoimmune Type 1 Diabetes (T1D) and the role of glutamic-acid decarboxylase antibodies (GADA) and other factors. METHODS Adults with autoimmune diabetes mellitus (DM) were recruited from the Diabetes Center of Nikaia-Piraeus Hospital. Demographic and clinical parameters were recorded and anti-pancreatic and organ-specific antibodies were measured. RESULTS Of 160 patients, 33.75% had one AAD and 24.37% had two or more. Patients with LADA had higher overall prevalence of AADs, mainly autoimmune thyroiditis and gastritis. Celiac disease was present only in T1D. GADA positive patients had higher prevalence of AADs and multiple autoimmunity, especially thyroiditis and gastritis. Patients with LADA had higher rates of positive GADA or islet-cell antibodies (ICA). After controlling for LADA, GADA remained a significant predictor of AADs. Female gender and chronological age were also significant predictors of AADs. CONCLUSIONS AADs were present in 58.13% of patients. Patients with LADA were more prone to a generalized autoimmune disorder than those with T1D. AADs development was significantly associated with female sex, older age and positive GADA, which proved an independent marker of associated autoimmunity.
Collapse
Affiliation(s)
- Dimitrios Gougourelas
- Diabetes Center, General Hospital of Nikaia - Piraeus "Agios Panteleimon", Athens, Greece.
| | - Charalampos Tsentidis
- Department of Endocrinology, Metabolism & Diabetes Mellitus, General Hospital of Nikaia - Piraeus "Agios Panteleimon", Athens, Greece
| | - Athina Maria Koufadaki
- Diabetes and Metabolism Clinic, 2(nd) Department of Pediatrics, National and Kapodistrian University of Athens, "P&A Kyriakou" Children's Hospital, Athens, Greece
| | | | - Eupraxia Gougourela
- Diabetes Center, General Hospital of Nikaia - Piraeus "Agios Panteleimon", Athens, Greece
| | - Spyridon Karanasios
- Diabetes and Metabolism Clinic, 2(nd) Department of Pediatrics, National and Kapodistrian University of Athens, "P&A Kyriakou" Children's Hospital, Athens, Greece
| | - Alexios Sotiropoulos
- Diabetes Center, General Hospital of Nikaia - Piraeus "Agios Panteleimon", Athens, Greece
| | - Stavros Bousboulas
- Diabetes Center, General Hospital of Nikaia - Piraeus "Agios Panteleimon", Athens, Greece
| | - Kyriaki Athina Karavanaki
- Diabetes and Metabolism Clinic, 2(nd) Department of Pediatrics, National and Kapodistrian University of Athens, "P&A Kyriakou" Children's Hospital, Athens, Greece
| |
Collapse
|
34
|
Li Q, Liu X, Yang J, Erlund I, Lernmark Å, Hagopian W, Rewers M, She JX, Toppari J, Ziegler AG, Akolkar B, Krischer JP. Plasma Metabolome and Circulating Vitamins Stratified Onset Age of an Initial Islet Autoantibody and Progression to Type 1 Diabetes: The TEDDY Study. Diabetes 2021; 70:282-292. [PMID: 33106256 PMCID: PMC7876562 DOI: 10.2337/db20-0696] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 10/20/2020] [Indexed: 12/11/2022]
Abstract
Children's plasma metabolome, especially lipidome, reflects gene regulation and dietary exposures, heralding the development of islet autoantibodies (IA) and type 1 diabetes (T1D). The Environmental Determinants of Diabetes in the Young (TEDDY) study enrolled 8,676 newborns by screening of HLA-DR-DQ genotypes at six clinical centers in four countries, profiled metabolome, and measured concentrations of ascorbic acid, 25-hydroxyvitamin D [25(OH)D], and erythrocyte membrane fatty acids following birth until IA seroconversion under a nested case-control design. We grouped children having an initial autoantibody only against insulin (IAA-first) or GAD (GADA-first) by unsupervised clustering of temporal lipidome, identifying a subgroup of children having early onset of each initial autoantibody, i.e., IAA-first by 12 months and GADA-first by 21 months, consistent with population-wide early seroconversion age. Differential analysis showed that infants having reduced plasma ascorbic acid and cholesterol experienced IAA-first earlier, while early onset of GADA-first was preceded by reduced sphingomyelins at infancy. Plasma 25(OH)D prior to either autoantibody was lower in T1D progressors compared with nonprogressors, with simultaneous lower diglycerides, lysophosphatidylcholines, triglycerides, and alanine before GADA-first. Plasma ascorbic acid and 25(OH)D at infancy were lower in HLA-DR3/DR4 children among IA case subjects but not in matched control subjects, implying gene expression dysregulation of circulating vitamins as latent signals for IA or T1D progression.
Collapse
Affiliation(s)
- Qian Li
- Health Informatics Institute, University of South Florida, Tampa, FL
| | - Xiang Liu
- Health Informatics Institute, University of South Florida, Tampa, FL
| | - Jimin Yang
- Health Informatics Institute, University of South Florida, Tampa, FL
| | - Iris Erlund
- Department of Government Services, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Åke Lernmark
- Department of Clinical Sciences, Clinical Research Centre, Skåne University Hospital, Lund University, Malmö, Sweden
| | | | - Marian Rewers
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, CO
| | - Jin-Xiong She
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia, Augusta University, Augusta, GA
| | - Jorma Toppari
- Department of Pediatrics, Turku University Hospital, Turku, Finland
- Department of Physiology, University of Turku, Turku, Finland
| | - Anette-G Ziegler
- Institute of Diabetes Research, Helmholtz Zentrum München, Munich, Germany
- Forschergruppe Diabetes, Technical University of Munich, Klinikum Rechts der Isar, Munich, Germany
- Forschergruppe Diabetes e.V. at Helmholtz Zentrum München, Munich, Germany
| | - Beena Akolkar
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD
| | | |
Collapse
|
35
|
Pöllänen PM, Ryhänen SJ, Toppari J, Ilonen J, Vähäsalo P, Veijola R, Siljander H, Knip M. Dynamics of Islet Autoantibodies During Prospective Follow-Up From Birth to Age 15 Years. J Clin Endocrinol Metab 2020; 105:5901133. [PMID: 32882033 PMCID: PMC7686032 DOI: 10.1210/clinem/dgaa624] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 08/31/2020] [Indexed: 01/23/2023]
Abstract
CONTEXT We set out to characterize the dynamics of islet autoantibodies over the first 15 years of life in children carrying genetic susceptibility to type 1 diabetes (T1D). We also assessed systematically the role of zinc transporter 8 autoantibodies (ZnT8A) in this context. DESIGN HLA-predisposed children (N = 1006, 53.0% boys) recruited from the general population during 1994 to 1997 were observed from birth over a median time of 14.9 years (range, 1.9-15.5 years) for ZnT8A, islet cell (ICA), insulin (IAA), glutamate decarboxylase (GADA), and islet antigen-2 (IA-2A) antibodies, and for T1D. RESULTS By age 15.5 years, 35 (3.5%) children had progressed to T1D. Islet autoimmunity developed in 275 (27.3%) children at a median age of 7.4 years (range, 0.3-15.1 years). The ICA seroconversion rate increased toward puberty, but the biochemically defined autoantibodies peaked at a young age. Before age 2 years, ZnT8A and IAA appeared commonly as the first autoantibody, but in the preschool years IA-2A- and especially GADA-initiated autoimmunity increased. Thereafter, GADA-positive seroconversions continued to appear steadily until ages 10 to 15 years. Inverse IAA seroconversions occurred frequently (49.3% turned negative) and marked a prolonged delay from seroconversion to diagnosis compared to persistent IAA (8.2 vs 3.4 years; P = .01). CONCLUSIONS In HLA-predisposed children, the primary autoantibody is characteristic of age and might reflect the events driving the disease process toward clinical T1D. Autoantibody persistence affects the risk of T1D. These findings provide a framework for identifying disease subpopulations and for personalizing the efforts to predict and prevent T1D.
Collapse
Affiliation(s)
- Petra M Pöllänen
- Pediatric Research Center, Children’s Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Samppa J Ryhänen
- Pediatric Research Center, Children’s Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Jorma Toppari
- Department of Pediatrics, Turku University Hospital, and Institute of Biomedicine and Centre for Population Health Research, University of Turku, Turku, Finland
| | - Jorma Ilonen
- Immunogenetics Laboratory, Institute of Biomedicine, University of Turku and Clinical Microbiology, Turku University Hospital, Turku, Finland
| | - Paula Vähäsalo
- Department of Pediatrics, PEDEGO Research Group, Medical Research Center, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Riitta Veijola
- Department of Pediatrics, PEDEGO Research Group, Medical Research Center, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Heli Siljander
- Pediatric Research Center, Children’s Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Mikael Knip
- Pediatric Research Center, Children’s Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Tampere Center for Child Health Research, Tampere University Hospital, Tampere, Finland
- Folkhälsan Research Center, Helsinki, Finland
- Correspondence and Reprint Requests: Mikael Knip, MD, PhD, Children’s Hospital, University of Helsinki, P.O. Box 22 (Stenbäckinkatu 11), FI-00014 Helsinki, Finland. E-mail:
| |
Collapse
|
36
|
So M, O'Rourke C, Bahnson HT, Greenbaum CJ, Speake C. Response to Comment on So et al. Autoantibody Reversion: Changing Risk Categories in Multiple-Autoantibody-Positive Individuals. Diabetes Care 2020;43:913-917. Diabetes Care 2020; 43:e103-e104. [PMID: 32669415 PMCID: PMC7372046 DOI: 10.2337/dci20-0016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Michelle So
- Diabetes Clinical Research Program, Benaroya Research Institute at Virginia Mason, Seattle, WA
| | - Colin O'Rourke
- Diabetes Clinical Research Program, Benaroya Research Institute at Virginia Mason, Seattle, WA
| | - Henry T Bahnson
- Diabetes Clinical Research Program, Benaroya Research Institute at Virginia Mason, Seattle, WA
| | - Carla J Greenbaum
- Diabetes Clinical Research Program, Benaroya Research Institute at Virginia Mason, Seattle, WA
| | - Cate Speake
- Diabetes Clinical Research Program, Benaroya Research Institute at Virginia Mason, Seattle, WA
| |
Collapse
|
37
|
Zhao LP, Papadopoulos GK, Kwok WW, Moustakas AK, Bondinas GP, Larsson HE, Ludvigsson J, Marcus C, Samuelsson U, Wang R, Pyo CW, Nelson WC, Geraghty DE, Lernmark Å. Motifs of Three HLA-DQ Amino Acid Residues (α44, β57, β135) Capture Full Association With the Risk of Type 1 Diabetes in DQ2 and DQ8 Children. Diabetes 2020; 69:1573-1587. [PMID: 32245799 PMCID: PMC7306123 DOI: 10.2337/db20-0075] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 03/30/2020] [Indexed: 12/25/2022]
Abstract
HLA-DQA1 and -DQB1 are strongly associated with type 1 diabetes (T1D), and DQ8.1 and DQ2.5 are major risk haplotypes. Next-generation targeted sequencing of HLA-DQA1 and -DQB1 in Swedish newly diagnosed 1- to 18 year-old patients (n = 962) and control subjects (n = 636) was used to construct abbreviated DQ haplotypes, converted into amino acid (AA) residues, and assessed for their associations with T1D. A hierarchically organized haplotype (HOH) association analysis allowed 45 unique DQ haplotypes to be categorized into seven clusters. The DQ8/9 cluster included two DQ8.1 risk and the DQ9 resistant haplotypes, and the DQ2 cluster included the DQ2.5 risk and DQ2.2 resistant haplotypes. Within each cluster, HOH found residues α44Q (odds ratio [OR] 3.29, P = 2.38 * 10-85) and β57A (OR 3.44, P = 3.80 * 10-84) to be associated with T1D in the DQ8/9 cluster representing all ten residues (α22, α23, α44, α49, α51, α53, α54, α73, α184, β57) due to complete linkage disequilibrium (LD) of α44 with eight such residues. Within the DQ2 cluster and due to LD, HOH analysis found α44C and β135D to share the risk for T1D (OR 2.10, P = 1.96 * 10-20). The motif "QAD" of α44, β57, and β135 captured the T1D risk association of DQ8.1 (OR 3.44, P = 3.80 * 10-84), and the corresponding motif "CAD" captured the risk association of DQ2.5 (OR 2.10, P = 1.96 * 10-20). Two risk associations were related to GAD65 autoantibody (GADA) and IA-2 autoantibody (IA-2A) but in opposite directions. CAD was positively associated with GADA (OR 1.56, P = 6.35 * 10-8) but negatively with IA-2A (OR 0.59, P = 6.55 * 10-11). QAD was negatively associated with GADA (OR 0.88; P = 3.70 * 10-3) but positively with IA-2A (OR 1.64; P = 2.40 * 10-14), despite a single difference at α44. The residues are found in and around anchor pockets 1 and 9, as potential T-cell receptor contacts, in the areas for CD4 binding and putative homodimer formation. The identification of three HLA-DQ AAs (α44, β57, β135) conferring T1D risk should sharpen functional and translational studies.
Collapse
Affiliation(s)
- Lue Ping Zhao
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - George K Papadopoulos
- Laboratory of Biophysics, Biochemistry, Biomaterials and Bioprocessing, Faculty of Agricultural Technology, Technological Educational Institute of Epirus, Arta, Greece
| | - William W Kwok
- Benaroya Research Institute at Virginia Mason, Seattle, WA
| | - Antonis K Moustakas
- Department of Food Science and Technology, Faculty of Environmental Sciences, Ionian University, Argostoli, Cephalonia, Greece
| | - George P Bondinas
- Laboratory of Biophysics, Biochemistry, Biomaterials and Bioprocessing, Faculty of Agricultural Technology, Technological Educational Institute of Epirus, Arta, Greece
| | - Helena Elding Larsson
- Department of Clinical Sciences, Lund University CRC, Skåne University Hospital, Malmö, Sweden
| | - Johnny Ludvigsson
- Crown Princess Victoria Children's Hospital, Region Östergötland, and Division of Pediatrics, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Claude Marcus
- Division of Pediatrics, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Ulf Samuelsson
- Crown Princess Victoria Children's Hospital, Region Östergötland, and Division of Pediatrics, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Ruihan Wang
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Chul-Woo Pyo
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Wyatt C Nelson
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Daniel E Geraghty
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Åke Lernmark
- Department of Clinical Sciences, Lund University CRC, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
38
|
Naseri R, Navabi SJ, Samimi Z, Mishra AP, Nigam M, Chandra H, Olatunde A, Tijjani H, Morais-Urano RP, Farzaei MH. Targeting Glycoproteins as a therapeutic strategy for diabetes mellitus and its complications. Daru 2020; 28:333-358. [PMID: 32006343 PMCID: PMC7095136 DOI: 10.1007/s40199-020-00327-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 01/10/2020] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVES Glycoproteins are organic compounds formed from proteins and carbohydrates, which are found in many parts of the living systems including the cell membranes. Furthermore, impaired metabolism of glycoprotein components plays the main role in the pathogenesis of diabetes mellitus. The aim of this study is to investigate the influence of glycoprotein levels in the treatment of diabetes mellitus. METHODS All relevant papers in the English language were compiled by searching electronic databases, including Scopus, PubMed and Cochrane library. The keywords of glycoprotein, diabetes mellitus, glycan, glycosylation, and inhibitor were searched until January 2019. RESULTS Glycoproteins are pivotal elements in the regulation of cell proliferation, growth, maturation and signaling pathways. Moreover, they are involved in drug binding, drug transportation, efflux of chemicals and stability of therapeutic proteins. These functions, structure, composition, linkages, biosynthesis, significance and biological effects are discussed as related to their use as a therapeutic strategy for the treatment of diabetes mellitus and its complications. CONCLUSIONS The findings revealed several chemical and natural compounds have significant beneficial effects on glycoprotein metabolism. The comprehension of glycoprotein structure and functions are very essential and inevitable to enhance the knowledge of glycoengineering for glycoprotein-based therapeutics as may be required for the treatment of diabetes mellitus and its associated complications. Graphical abstract.
Collapse
Affiliation(s)
- Rozita Naseri
- Internal Medicine Department, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Seyed Jafar Navabi
- Internal Medicine Department, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Zeinab Samimi
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Abhay Prakash Mishra
- Department of Pharmaceutical Chemistry, Hemwati Nandan Bahuguna Garhwal (A Central) University, Srinagar Garhwal, Uttarakhand, 246174, India.
| | - Manisha Nigam
- Department of Biochemistry, Hemwati Nandan Bahuguna Garhwal University, Srinagar Garhwal, Uttarakhand, 246174, India
| | - Harish Chandra
- Department of Microbiology, Gurukul Kangri Vishwavidhyalya, Haridwar, Uttarakhand, 249404, India
| | - Ahmed Olatunde
- Department of Biochemistry, Abubakar Tafawa Balewa University, Bauchi, Nigeria
| | - Habibu Tijjani
- Natural Product Research Laboratory, Department of Biochemistry, Bauchi State University, Gadau, Nigeria
| | - Raquel P Morais-Urano
- Instituto de Química de São Carlos, Universidade de São Paulo, 13560-970, São Carlos, SP, Brasil
| | - Mohammad Hosein Farzaei
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
39
|
Paun A, Yau C, Meshkibaf S, Daigneault MC, Marandi L, Mortin-Toth S, Bar-Or A, Allen-Vercoe E, Poussier P, Danska JS. Association of HLA-dependent islet autoimmunity with systemic antibody responses to intestinal commensal bacteria in children. Sci Immunol 2020; 4:4/32/eaau8125. [PMID: 30709843 DOI: 10.1126/sciimmunol.aau8125] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 12/13/2018] [Indexed: 12/11/2022]
Abstract
Microbiome sequence analyses have suggested that changes in gut bacterial composition are associated with autoimmune disease in humans and animal models. However, little is known of the mechanisms through which the gut microbiota influences autoimmune responses to distant tissues. Here, we evaluated systemic antibody responses against cultured human gut bacterial strains to determine whether observed patterns of anticommensal antibody (ACAb) responses are associated with type 1 diabetes (T1D) in two cohorts of pediatric study participants. In the first cohort, ACAb responses in sera collected from participants within 6 months of T1D diagnosis were compared with age-matched healthy controls and also with patients with recent onset Crohn's disease. ACAb responses against multiple bacterial species discriminated among these three groups. In the second cohort, we asked whether ACAb responses present before diagnosis were associated with later T1D development and with HLA genotype in participants who were discordant for subsequent progression to diabetes. Serum IgG2 antibodies against Roseburia faecis and against a bacterial consortium were associated with future T1D diagnosis in an HLA DR3/DR4 haplotype-dependent manner. These analyses reveal associations between antibody responses to intestinal microbes and HLA-DR genotype and islet autoantibody specificity and with a future diagnosis of T1D. Further, we present a platform to investigate antibacterial antibodies in biological fluids that is applicable to studies of autoimmune diseases and responses to therapeutic interventions.
Collapse
Affiliation(s)
- Alexandra Paun
- Hospital for Sick Children, Toronto, ON, Canada. .,Department of Immunology, University of Toronto, Faculty of Medicine, Toronto, ON, Canada
| | - Christopher Yau
- Hospital for Sick Children, Toronto, ON, Canada. .,Department of Immunology, University of Toronto, Faculty of Medicine, Toronto, ON, Canada
| | | | - Michelle C Daigneault
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, Canada
| | | | | | - Amit Bar-Or
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada.,Department of Neurology, Perelman Center for Advanced Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, U.S.A
| | - Emma Allen-Vercoe
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, Canada
| | - Philippe Poussier
- Department of Immunology, University of Toronto, Faculty of Medicine, Toronto, ON, Canada.,Sunnybrook Research Institute, Toronto, ON Canada
| | - Jayne S Danska
- Hospital for Sick Children, Toronto, ON, Canada. .,Department of Immunology, University of Toronto, Faculty of Medicine, Toronto, ON, Canada.,Department of Medical Biophysics, University of Toronto, Faculty of Medicine, Toronto, ON, Canada
| |
Collapse
|
40
|
So M, O'Rourke C, Bahnson HT, Greenbaum CJ, Speake C. Autoantibody Reversion: Changing Risk Categories in Multiple-Autoantibody-Positive Individuals. Diabetes Care 2020; 43:913-917. [PMID: 32019856 PMCID: PMC7085807 DOI: 10.2337/dc19-1731] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 01/11/2020] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Most individuals with two or more islet autoantibodies progress to clinical type 1 diabetes. However, in some individuals, autoantibodies are subsequently lost. Here, our objectives were to determine the frequency of autoantibody loss (reversion) in multiple-autoantibody-positive individuals and to determine the association between reversion and progression to clinical disease. RESEARCH DESIGN AND METHODS We analyzed multiple-autoantibody-positive individuals participating in TrialNet's Pathway to Prevention Study for reversion and determined the effect of reversion on progression to clinical disease using a Cox regression analysis. RESULTS Of 3,284 multiple-autoantibody-positive subjects, reversion occurred in 134 (4.1%) and was associated with reduced incidence of clinical disease. Reversion occurred more frequently with older age, lower autoantibody titers, and fewer positive autoantibodies. CONCLUSIONS Although reversion of multiple-autoantibody positivity is rare, when it occurs, the risk of progressing to clinical disease is reduced. This suggests unknown mechanisms promoting immune remission in some individuals.
Collapse
Affiliation(s)
- Michelle So
- Diabetes Clinical Research Program, Benaroya Research Institute at Virginia Mason, Seattle, WA
| | - Colin O'Rourke
- Diabetes Clinical Research Program, Benaroya Research Institute at Virginia Mason, Seattle, WA
| | - Henry T Bahnson
- Diabetes Clinical Research Program, Benaroya Research Institute at Virginia Mason, Seattle, WA
| | - Carla J Greenbaum
- Diabetes Clinical Research Program, Benaroya Research Institute at Virginia Mason, Seattle, WA
| | - Cate Speake
- Diabetes Clinical Research Program, Benaroya Research Institute at Virginia Mason, Seattle, WA
| |
Collapse
|
41
|
Jacobsen LM, Bocchino L, Evans-Molina C, DiMeglio L, Goland R, Wilson DM, Atkinson MA, Aye T, Russell WE, Wentworth JM, Boulware D, Geyer S, Sosenko JM. The risk of progression to type 1 diabetes is highly variable in individuals with multiple autoantibodies following screening. Diabetologia 2020; 63:588-596. [PMID: 31768570 PMCID: PMC7229995 DOI: 10.1007/s00125-019-05047-w] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Accepted: 10/11/2019] [Indexed: 12/30/2022]
Abstract
AIMS/HYPOTHESIS Young children who develop multiple autoantibodies (mAbs) are at very high risk for type 1 diabetes. We assessed whether a population with mAbs detected by screening is also at very high risk, and how risk varies according to age, type of autoantibodies and metabolic status. METHODS Type 1 Diabetes TrialNet Pathway to Prevention participants with mAbs (n = 1815; age, 12.35 ± 9.39 years; range, 1-49 years) were analysed. Type 1 diabetes risk was assessed according to age, autoantibody type/number (insulin autoantibodies [IAA], glutamic acid decarboxylase autoantibodies [GADA], insulinoma-associated antigen-2 autoantibodies [IA-2A] or zinc transporter 8 autoantibodies [ZnT8A]) and Index60 (composite measure of fasting C-peptide, 60 min glucose and 60 min C-peptide). Cox regression and cumulative incidence curves were utilised in this cohort study. RESULTS Age was inversely related to type 1 diabetes risk in those with mAbs (HR 0.97 [95% CI 0.96, 0.99]). Among participants with 2 autoantibodies, those with GADA had less risk (HR 0.35 [95% CI 0.22, 0.57]) and those with IA-2A had higher risk (HR 2.82 [95% CI 1.76, 4.51]) of type 1 diabetes. Those with IAA and GADA had only a 17% 5 year risk of type 1 diabetes. The risk was significantly lower for those with Index60 <1.0 (HR 0.23 [95% CI 0.19, 0.30]) vs those with Index60 values ≥1.0. Among the 12% (225/1815) ≥12.0 years of age with GADA positivity, IA-2A negativity and Index60 <1.0, the 5 year risk of type 1 diabetes was 8%. CONCLUSIONS/INTERPRETATION Type 1 diabetes risk varies substantially according to age, autoantibody type and metabolic status in individuals screened for mAbs. An appreciable proportion of older children and adults with mAbs appear to have a low risk of progressing to type 1 diabetes at 5 years. With this knowledge, clinical trials of type 1 diabetes prevention can better target those most likely to progress.
Collapse
Affiliation(s)
- Laura M Jacobsen
- Division of Pediatric Endocrinology, Department of Pediatrics, College of Medicine, University of Florida, 1275 Center Drive, Gainesville, FL, 32610, USA.
| | - Laura Bocchino
- Health Informatics Institute, University of South Florida, Tampa, FL, USA
| | - Carmella Evans-Molina
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Linda DiMeglio
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Robin Goland
- Division of Pediatric Endocrinology, Diabetes, and Metabolism, Columbia University Medical Center, New York, NY, USA
| | - Darrell M Wilson
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Mark A Atkinson
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL, USA
| | - Tandy Aye
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - William E Russell
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - John M Wentworth
- Walter and Eliza Hall Institute, Parkville, VIC, Australia
- Department of Diabetes and Endocrinology, Royal Melbourne Hospital, Parkville, VIC, Australia
| | - David Boulware
- Health Informatics Institute, University of South Florida, Tampa, FL, USA
| | - Susan Geyer
- Health Informatics Institute, University of South Florida, Tampa, FL, USA
| | - Jay M Sosenko
- Division of Endocrinology, University of Miami, Miami, FL, USA
| |
Collapse
|
42
|
Le Bagge S, Fotheringham AK, Leung SS, Forbes JM. Targeting the receptor for advanced glycation end products (RAGE) in type 1 diabetes. Med Res Rev 2020; 40:1200-1219. [PMID: 32112452 DOI: 10.1002/med.21654] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 11/09/2019] [Accepted: 11/12/2019] [Indexed: 12/18/2022]
Abstract
Type 1 diabetes (T1D) is one of the most common chronic diseases manifesting in early life, with the prevalence increasing worldwide at a rate of approximately 3% per annum. The prolonged hyperglycaemia characteristic of T1D upregulates the receptor for advanced glycation end products (RAGE) and accelerates the formation of RAGE ligands, including advanced glycation end products, high-mobility group protein B1, S100 calcium-binding proteins, and amyloid-beta. Interestingly, changes in the expression of RAGE and these ligands are evident in patients before the onset of T1D. RAGE signals via various proinflammatory cascades, resulting in the production of reactive oxygen species and cytokines. A large number of proinflammatory ligands that can signal via RAGE have been implicated in several chronic diseases, including T1D. Therefore, it is unsurprising that RAGE has become a potential therapeutic target for the treatment and prevention of disease. In this review, we will explore how RAGE might be targeted to prevent the development of T1D.
Collapse
Affiliation(s)
- Selena Le Bagge
- Glycation and Diabetes, Translational Research Institute (TRI), Mater Research Institute-The University of Queensland (MRI-UQ), Brisbane, Queensland, Australia.,School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Amelia K Fotheringham
- Glycation and Diabetes, Translational Research Institute (TRI), Mater Research Institute-The University of Queensland (MRI-UQ), Brisbane, Queensland, Australia.,School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Sherman S Leung
- Glycation and Diabetes, Translational Research Institute (TRI), Mater Research Institute-The University of Queensland (MRI-UQ), Brisbane, Queensland, Australia.,School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Josephine M Forbes
- Glycation and Diabetes, Translational Research Institute (TRI), Mater Research Institute-The University of Queensland (MRI-UQ), Brisbane, Queensland, Australia.,Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia.,Mater Clinical School, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
43
|
Frohnert BI, Webb-Robertson BJ, Bramer LM, Reehl SM, Waugh K, Steck AK, Norris JM, Rewers M. Predictive Modeling of Type 1 Diabetes Stages Using Disparate Data Sources. Diabetes 2020; 69:238-248. [PMID: 31740441 PMCID: PMC6971485 DOI: 10.2337/db18-1263] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 11/11/2019] [Indexed: 12/18/2022]
Abstract
This study aims to model genetic, immunologic, metabolomics, and proteomic biomarkers for development of islet autoimmunity (IA) and progression to type 1 diabetes in a prospective high-risk cohort. We studied 67 children: 42 who developed IA (20 of 42 progressed to diabetes) and 25 control subjects matched for sex and age. Biomarkers were assessed at four time points: earliest available sample, just prior to IA, just after IA, and just prior to diabetes onset. Predictors of IA and progression to diabetes were identified across disparate sources using an integrative machine learning algorithm and optimization-based feature selection. Our integrative approach was predictive of IA (area under the receiver operating characteristic curve [AUC] 0.91) and progression to diabetes (AUC 0.92) based on standard cross-validation (CV). Among the strongest predictors of IA were change in serum ascorbate, 3-methyl-oxobutyrate, and the PTPN22 (rs2476601) polymorphism. Serum glucose, ADP fibrinogen, and mannose were among the strongest predictors of progression to diabetes. This proof-of-principle analysis is the first study to integrate large, diverse biomarker data sets into a limited number of features, highlighting differences in pathways leading to IA from those predicting progression to diabetes. Integrated models, if validated in independent populations, could provide novel clues concerning the pathways leading to IA and type 1 diabetes.
Collapse
Affiliation(s)
- Brigitte I Frohnert
- Barbara Davis Center for Diabetes, School of Medicine, University of Colorado, Aurora, CO
| | - Bobbie-Jo Webb-Robertson
- Computational and Statistical Analytics Division, Pacific Northwest National Laboratory, Richland, WA
| | - Lisa M Bramer
- Computational and Statistical Analytics Division, Pacific Northwest National Laboratory, Richland, WA
| | - Sara M Reehl
- Computational and Statistical Analytics Division, Pacific Northwest National Laboratory, Richland, WA
| | - Kathy Waugh
- Barbara Davis Center for Diabetes, School of Medicine, University of Colorado, Aurora, CO
| | - Andrea K Steck
- Barbara Davis Center for Diabetes, School of Medicine, University of Colorado, Aurora, CO
| | - Jill M Norris
- Department of Epidemiology, Colorado School of Public Health, University of Colorado, Aurora, CO
| | - Marian Rewers
- Barbara Davis Center for Diabetes, School of Medicine, University of Colorado, Aurora, CO
| |
Collapse
|
44
|
Ilonen J, Lempainen J, Veijola R. The heterogeneous pathogenesis of type 1 diabetes mellitus. Nat Rev Endocrinol 2019; 15:635-650. [PMID: 31534209 DOI: 10.1038/s41574-019-0254-y] [Citation(s) in RCA: 251] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/13/2019] [Indexed: 12/14/2022]
Abstract
Type 1 diabetes mellitus (T1DM) results from the destruction of pancreatic β-cells that is mediated by the immune system. Multiple genetic and environmental factors found in variable combinations in individual patients are involved in the development of T1DM. Genetic risk is defined by the presence of particular allele combinations, which in the major susceptibility locus (the HLA region) affect T cell recognition and tolerance to foreign and autologous molecules. Multiple other loci also regulate and affect features of specific immune responses and modify the vulnerability of β-cells to inflammatory mediators. Compared with the genetic factors, environmental factors that affect the development of T1DM are less well characterized but contact with particular microorganisms is emerging as an important factor. Certain infections might affect immune regulation, and the role of commensal microorganisms, such as the gut microbiota, are important in the education of the developing immune system. Some evidence also suggests that nutritional factors are important. Multiple islet-specific autoantibodies are found in the circulation from a few weeks to up to 20 years before the onset of clinical disease and this prediabetic phase provides a potential opportunity to manipulate the islet-specific immune response to prevent or postpone β-cell loss. The latest developments in understanding the heterogeneity of T1DM and characterization of major disease subtypes might help in the development of preventive treatments.
Collapse
Affiliation(s)
- Jorma Ilonen
- Institue of Biomedicine, University of Turku and Clinical Microbiology, Turku University Hospital, Turku, Finland.
| | - Johanna Lempainen
- Institue of Biomedicine, University of Turku and Clinical Microbiology, Turku University Hospital, Turku, Finland
- Department of Paediatrics, University of Turku and Turku University Hospital, Turku, Finland
| | - Riitta Veijola
- Department of Paediatrics, University of Oulu and Oulu University Hospital, Oulu, Finland
| |
Collapse
|
45
|
Sørgjerd EP. Type 1 Diabetes-related Autoantibodies in Different Forms of Diabetes. Curr Diabetes Rev 2019; 15:199-204. [PMID: 30058495 DOI: 10.2174/1573399814666180730105351] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 04/29/2018] [Accepted: 07/16/2018] [Indexed: 12/11/2022]
Abstract
Autoantibodies against Glutamic Acid Decarboxylase (GADA), insulinoma antigen-2 (IA- 2A), insulin (IAA) and the most recently Zinc Transporter 8 (ZnT8A) are one of the most reliable biomarkers for autoimmune diabetes in both children and adults. They are today the only biomarkers that can distinguish Latent Autoimmune Diabetes in Adults (LADA) from phenotypically type 2 diabetes. As the frequency of autoantibodies at diagnosis in childhood type 1 diabetes depends on age, GADA is by far the most common in adult onset autoimmune diabetes, especially LADA. Being multiple autoantibody positive have also shown to be more common in childhood diabetes compared to adult onset diabetes, and multiple autoantibody positivity have a high predictive value of childhood type 1 diabetes. Autoantibodies have shown inconsistent results to predict diabetes in adults. Levels of autoantibodies are reported to cause heterogeneity in LADA. Reports indicate that individuals with high levels of autoantibodies have a more type 1 diabetes like phenotype and individuals with low levels of autoantibody positivity have a more type 2 diabetes like phenotype. It is also well known that autoantibody levels can fluctuate and transient autoantibody positivity in adult onset autoimmune diabetes have been reported to affect the phenotype.
Collapse
Affiliation(s)
- Elin Pettersen Sørgjerd
- HUNT Research Centre, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
46
|
Endesfelder D, Zu Castell W, Bonifacio E, Rewers M, Hagopian WA, She JX, Lernmark Å, Toppari J, Vehik K, Williams AJK, Yu L, Akolkar B, Krischer JP, Ziegler AG, Achenbach P. Time-Resolved Autoantibody Profiling Facilitates Stratification of Preclinical Type 1 Diabetes in Children. Diabetes 2019; 68:119-130. [PMID: 30305370 PMCID: PMC6302536 DOI: 10.2337/db18-0594] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2018] [Accepted: 10/03/2018] [Indexed: 12/13/2022]
Abstract
Progression to clinical type 1 diabetes varies among children who develop β-cell autoantibodies. Differences in autoantibody patterns could relate to disease progression and etiology. Here we modeled complex longitudinal autoantibody profiles by using a novel wavelet-based algorithm. We identified clusters of similar profiles associated with various types of progression among 600 children from The Environmental Determinants of Diabetes in the Young (TEDDY) birth cohort study; these children developed persistent insulin autoantibodies (IAA), GAD autoantibodies (GADA), insulinoma-associated antigen 2 autoantibodies (IA-2A), or a combination of these, and they were followed up prospectively at 3- to 6-month intervals (median follow-up 6.5 years). Children who developed multiple autoantibody types (n = 370) were clustered, and progression from seroconversion to clinical diabetes within 5 years ranged between clusters from 6% (95% CI 0, 17.4) to 84% (59.2, 93.6). Children who seroconverted early in life (median age <2 years) and developed IAA and IA-2A that were stable-positive on follow-up had the highest risk of diabetes, and this risk was unaffected by GADA status. Clusters of children who lacked stable-positive GADA responses contained more boys and lower frequencies of the HLA-DR3 allele. Our novel algorithm allows refined grouping of β-cell autoantibody-positive children who distinctly progressed to clinical type 1 diabetes, and it provides new opportunities in searching for etiological factors and elucidating complex disease mechanisms.
Collapse
Affiliation(s)
- David Endesfelder
- Scientific Computing Research Unit, Helmholtz Zentrum München, Munich, Germany
| | - Wolfgang Zu Castell
- Scientific Computing Research Unit, Helmholtz Zentrum München, Munich, Germany
- Department of Mathematics, Technische Universität München, Munich, Germany
| | - Ezio Bonifacio
- Center for Regenerative Therapies, Dresden, and Paul Langerhans Institute Dresden, Technische Universität Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Marian Rewers
- Barbara Davis Center for Childhood Diabetes, University of Colorado, Aurora, CO
| | | | - Jin-Xiong She
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia, Augusta University, Augusta, GA
| | - Åke Lernmark
- Department of Clinical Sciences, Lund University Clinical Research Center, Skåne University Hospital, Malmo, Sweden
| | - Jorma Toppari
- Department of Pediatrics, Turku University Hospital, Turku, Finland
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Turku, Finland
| | - Kendra Vehik
- Health Informatics Institute, Morsani College of Medicine, University of South Florida, Tampa, FL
| | - Alistair J K Williams
- Diabetes and Metabolism, Translational Health Sciences, Southmead Hospital, University of Bristol, Bristol, U.K
| | - Liping Yu
- Barbara Davis Center for Childhood Diabetes, University of Colorado, Aurora, CO
| | - Beena Akolkar
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD
| | - Jeffrey P Krischer
- Health Informatics Institute, Morsani College of Medicine, University of South Florida, Tampa, FL
| | - Anette-G Ziegler
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute of Diabetes Research, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany
- Forschergruppe Diabetes, Technische Universität München at Klinikum rechts der Isar, Munich, Germany
- Forschergruppe Diabetes e.V. at Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany
| | - Peter Achenbach
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute of Diabetes Research, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany
- Forschergruppe Diabetes, Technische Universität München at Klinikum rechts der Isar, Munich, Germany
- Forschergruppe Diabetes e.V. at Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany
| |
Collapse
|
47
|
Yi L, Swensen AC, Qian WJ. Serum biomarkers for diagnosis and prediction of type 1 diabetes. Transl Res 2018; 201:13-25. [PMID: 30144424 PMCID: PMC6177288 DOI: 10.1016/j.trsl.2018.07.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 07/02/2018] [Accepted: 07/24/2018] [Indexed: 12/25/2022]
Abstract
Type 1 diabetes (T1D) culminates in the autoimmune destruction of the pancreatic βcells, leading to insufficient production of insulin and development of hyperglycemia. Serum biomarkers including a combination of glucose, glycated molecules, C-peptide, and autoantibodies have been well established for the diagnosis of T1D. However, these molecules often mark a late stage of the disease when ∼90% of the pancreatic insulin-producing β-cells have already been lost. With the prevalence of T1D increasing worldwide and because of the physical and psychological burden induced by this disease, there is a great need for prognostic biomarkers to predict T1D development or progression. This would allow us to identify individuals at high risk for early prevention and intervention. Therefore, considerable efforts have been dedicated to the understanding of disease etiology and the discovery of novel biomarkers in the last few decades. The advent of high-throughput and sensitive "-omics" technologies for the study of proteins, nucleic acids, and metabolites have allowed large scale profiling of protein expression and gene changes in T1D patients relative to disease-free controls. In this review, we briefly discuss the classical diagnostic biomarkers of T1D but mainly focus on the novel biomarkers that are identified as markers of β-cell destruction and screened with the use of state-of-the-art "-omics" technologies.
Collapse
Affiliation(s)
- Lian Yi
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington
| | - Adam C Swensen
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington
| | - Wei-Jun Qian
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington.
| |
Collapse
|
48
|
Salami F, Lee HS, Freyhult E, Elding Larsson H, Lernmark Å, Törn C. Reduction in White Blood Cell, Neutrophil, and Red Blood Cell Counts Related to Sex, HLA, and Islet Autoantibodies in Swedish TEDDY Children at Increased Risk for Type 1 Diabetes. Diabetes 2018; 67:2329-2336. [PMID: 30104249 PMCID: PMC6198343 DOI: 10.2337/db18-0355] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 07/29/2018] [Indexed: 12/19/2022]
Abstract
Islet autoantibodies (IAs) precede the clinical onset of type 1 diabetes (T1D); however, the knowledge is limited about whether the prodrome affects complete blood counts (CBCs) in 4- to 12-year-old children with increased genetic risk for T1D. This study tested whether CBCs were altered in 4- to 12-year-old children without (n = 376) or with one or several IAs against insulin, GAD65, or IA-2 (n = 72). CBC was analyzed during longitudinal follow-up in 448 Swedish children enrolled in The Environmental Determinants of Diabetes in the Young (TEDDY) study. A linear mixed-effects model was used to assess potential association between IA and CBC measurements over time. The white blood cell and neutrophil counts were reduced in children with IAs, primarily in boys. In contrast, girls had lower levels of hemoglobin and hematocrit. Positivity for multiple IAs showed the lowest counts in white blood cells and neutrophils in boys and red blood cells, hemoglobin, and hematocrit in girls. These associations were primarily observed in children with the HLA-DR3-DQ2/DR4-DQ8 genotype. We conclude that the reduction in neutrophils and red blood cells in children with multiple IAs and HLA-DR3-DQ2/DR4-DQ8 genotype may signal a sex-dependent islet autoimmunity detected in longitudinal CBCs.
Collapse
Affiliation(s)
- Falastin Salami
- Department of Clinical Sciences, Lund University/Clinical Research Centre, Skåne University Hospital, Malmö, Sweden
| | - Hye-Seung Lee
- Health Informatics Institute, Department of Pediatrics, University of South Florida, Tampa, FL
| | - Eva Freyhult
- Department of Medical Sciences, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Helena Elding Larsson
- Department of Clinical Sciences, Lund University/Clinical Research Centre, Skåne University Hospital, Malmö, Sweden
| | - Åke Lernmark
- Department of Clinical Sciences, Lund University/Clinical Research Centre, Skåne University Hospital, Malmö, Sweden
| | - Carina Törn
- Department of Clinical Sciences, Lund University/Clinical Research Centre, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
49
|
Rewers M, Hyöty H, Lernmark Å, Hagopian W, She JX, Schatz D, Ziegler AG, Toppari J, Akolkar B, Krischer J. The Environmental Determinants of Diabetes in the Young (TEDDY) Study: 2018 Update. Curr Diab Rep 2018; 18:136. [PMID: 30353256 PMCID: PMC6415767 DOI: 10.1007/s11892-018-1113-2] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE OF REVIEW The environmental triggers of islet autoimmunity leading to type 1 diabetes (T1D) need to be elucidated to inform primary prevention. The Environmental Determinants of Diabetes in the Young (TEDDY) Study follows from birth 8676 children with T1D risk HLA-DR-DQ genotypes in the USA, Finland, Germany, and Sweden. Most study participants (89%) have no first-degree relative with T1D. The primary outcomes include the appearance of one or more persistent islet autoantibodies (islet autoimmunity, IA) and clinical T1D. RECENT FINDINGS As of February 28, 2018, 769 children had developed IA and 310 have progressed to T1D. Secondary outcomes include celiac disease and autoimmune thyroid disease. While the follow-up continues, TEDDY has already evaluated a number of candidate environmental triggers, including infections, probiotics, micronutrient, and microbiome. TEDDY results suggest that there are multiple pathways leading to the destruction of pancreatic beta-cells. Ongoing measurements of further specific exposures, gene variants, and gene-environment interactions and detailed "omics" studies will provide novel information on the pathogenesis of T1D.
Collapse
Affiliation(s)
- Marian Rewers
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, 1775 Aurora Ct, Aurora, CO, 80045, USA.
| | - Heikki Hyöty
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
- Fimlab Laboratories, Pirkanmaa Hospital District, Tampere, Finland
| | - Åke Lernmark
- Department of Clinical Sciences, Lund University/CRC, Skane University, Malmö, Sweden
| | | | - Jin-Xiong She
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | | | - Anette-G Ziegler
- Forschergruppe Diabetes e.V. and Institute of Diabetes Research, Helmholtz Zentrum, Munich, Germany
| | - Jorma Toppari
- Institute of Biomedicine, Centre for Integrative Physiology and Pharmacology, University of Turku, Turku, Finland
| | - Beena Akolkar
- Department of Pediatrics, Turku University Hospital, Turku, Finland
- National Institutes of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA
| | - Jeffrey Krischer
- Health Informatics Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| |
Collapse
|
50
|
Koskinen MK, Lempainen J, Löyttyniemi E, Helminen O, Hekkala A, Härkönen T, Kiviniemi M, Simell O, Knip M, Ilonen J, Toppari J, Veijola R. Class II HLA Genotype Association With First-Phase Insulin Response Is Explained by Islet Autoantibodies. J Clin Endocrinol Metab 2018; 103:2870-2878. [PMID: 29300921 PMCID: PMC6097602 DOI: 10.1210/jc.2017-02040] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 12/21/2017] [Indexed: 12/17/2022]
Abstract
CONTEXT A declining first-phase insulin response (FPIR) is characteristic of the disease process leading to clinical type 1 diabetes. It is not known whether reduced FPIR depends on class II human leukocyte antigen (HLA) genotype, islet autoimmunity, or both. OBJECTIVE To dissect the role of class II HLA DR-DQ genotypes and biochemical islet autoantibodies in the compromised FPIR. DESIGN, SETTING, PARTICIPANTS A total of 438 children with defined HLA DR-DQ genotype in the prospective Finnish Type 1 Diabetes Prediction and Prevention Study were analyzed for FPIR in a total of 1149 intravenous glucose tolerance tests and were categorized by their HLA DR-DQ genotype and the number of biochemical islet autoantibodies at the time of the first FPIR. Age-adjusted hierarchical linear mixed models were used to analyze repeated measurements of FPIR. MAIN OUTCOME MEASURE The associations between class II HLA DR-DQ genotype, islet autoantibody status, and FPIR. RESULTS A strong association between the degree of risk conferred by HLA DR-DQ genotype and positivity for islet autoantibodies existed (P < 0.0001). FPIR was inversely associated with the number of biochemical autoantibodies (P < 0.0001) irrespective of HLA DR-DQ risk group. FPIR decreased over time in children with multiple autoantibodies and increased in children with no biochemical autoantibodies (P < 0.0001 and P = 0.0013, respectively). CONCLUSIONS The class II HLA DR-DQ genotype association with FPIR was secondary to the association between HLA and islet autoimmunity. Declining FPIR was associated with positivity for multiple islet autoantibodies irrespective of class II HLA DR-DQ genotype.
Collapse
Affiliation(s)
- Maarit K Koskinen
- Department of Pediatrics, University of Turku and Turku University Hospital, Turku, Finland
- Correspondence and Reprint Requests: Maarit K. Koskinen, MD, University of Turku, Lemminkäisenkatu 3, 20520 Turku, Finland. E-mail:
| | - Johanna Lempainen
- Department of Pediatrics, University of Turku and Turku University Hospital, Turku, Finland
- Immunogenetics Laboratory, Institute of Biomedicine, University of Turku and Turku University Hospital, Turku, Finland
| | | | - Olli Helminen
- Department of Pediatrics, PEDEGO Research Unit, Medical Research Center, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Anne Hekkala
- Department of Pediatrics, PEDEGO Research Unit, Medical Research Center, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Taina Härkönen
- Children’s Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Programs Unit, Diabetes and Obesity, University of Helsinki, Helsinki, Finland
| | - Minna Kiviniemi
- Immunogenetics Laboratory, Institute of Biomedicine, University of Turku and Turku University Hospital, Turku, Finland
| | - Olli Simell
- Department of Pediatrics, University of Turku and Turku University Hospital, Turku, Finland
| | - Mikael Knip
- Children’s Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Programs Unit, Diabetes and Obesity, University of Helsinki, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland
- Tampere Center for Child Health Research, Tampere University Hospital, Tampere, Finland
| | - Jorma Ilonen
- Immunogenetics Laboratory, Institute of Biomedicine, University of Turku and Turku University Hospital, Turku, Finland
| | - Jorma Toppari
- Department of Pediatrics, University of Turku and Turku University Hospital, Turku, Finland
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Turku, Finland
| | - Riitta Veijola
- Department of Pediatrics, PEDEGO Research Unit, Medical Research Center, Oulu University Hospital and University of Oulu, Oulu, Finland
| |
Collapse
|