1
|
Ferrat LA, Templeman EL, Steck AK, Parikh HM, You L, Onengut-Gumuscu S, Gottlieb PA, Triolo TM, Rich SS, Krischer J, McQueen RB, Oram RA, Redondo MJ. Type 1 diabetes prediction in autoantibody-positive individuals: performance, time and money matter. Diabetologia 2025:10.1007/s00125-025-06434-2. [PMID: 40347237 DOI: 10.1007/s00125-025-06434-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 02/21/2025] [Indexed: 05/12/2025]
Abstract
AIMS/HYPOTHESIS Efficient prediction of clinical type 1 diabetes is important for risk stratification and monitoring of autoantibody-positive individuals. In this study, we compared type 1 diabetes predictive models for predictive performance, cost and participant time needed for testing. METHODS We developed 1943 predictive models using a Cox model based on a type 1 diabetes genetic risk score (GRS2), autoantibody count and types, BMI, age, self-reported gender and OGTT-derived glucose and C-peptide measures. We trained and validated the models using halves of a dataset comprising autoantibody-positive first-degree relatives of individuals with type 1 diabetes (n=3967, 49% female, 14.9 ± 12.1 years of age) from the TrialNet Pathway to Prevention study. The median duration of follow-up was 4.7 years (IQR 2.0-8.1), and 1311 participants developed clinical type 1 diabetes. Models were compared for predictive performances, estimated cost and participant time. RESULTS Models that included metabolic measures had best performance, with most exhibiting small performance differences (less than 3% and p>0.05). However, the cost and participant time associated with measuring metabolic variables ranged between US$56 and US$293 and 10-165 min, respectively. The predictive model performance had temporal variability, with the highest GRS2 influence and discriminative power being exhibited in the earliest preclinical stages. OGTT-derived metabolic measures had a similar performance to HbA1c- or Index60-derived models, with an important difference in cost and participant time. CONCLUSIONS/INTERPRETATION Cost-performance model analyses identified trade-offs between cost and performance models, and identified cost-minimising options to tailor risk-screening strategies.
Collapse
Affiliation(s)
- Lauric A Ferrat
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK.
- Department of Genetic Medicine and Development, University of Geneva, Geneva, Switzerland.
| | - Erin L Templeman
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK
| | - Andrea K Steck
- Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Hemang M Parikh
- Health Informatics Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Lu You
- Health Informatics Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | | | - Peter A Gottlieb
- Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Taylor M Triolo
- Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Stephen S Rich
- Department of Genome Sciences, University of Virginia, Charlottesville, VA, USA
| | - Jeffrey Krischer
- Health Informatics Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - R Brett McQueen
- Department of Clinical Pharmacy, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Richard A Oram
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK.
| | - Maria J Redondo
- Baylor College of Medicine, Texas Children's Hospital, Houston, TX, USA
| |
Collapse
|
2
|
Sims EK, Cuthbertson D, Ferrat LA, Bosi E, Evans-Molina C, DiMeglio LA, Nathan BM, Ismail HM, Jacobsen LM, Redondo MJ, Oram RA, Sosenko JM. IA-2A positivity increases risk of progression within and across established stages of type 1 diabetes. Diabetologia 2025; 68:993-1004. [PMID: 40016443 PMCID: PMC12021956 DOI: 10.1007/s00125-025-06382-x] [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: 07/22/2024] [Accepted: 01/10/2025] [Indexed: 03/01/2025]
Abstract
AIMS/HYPOTHESIS Accurate understanding of type 1 diabetes risk is critical for optimisation of counselling, monitoring and interventions, yet even within established staging classifications, individual time to clinical disease varies. Previous work has associated IA-2A positivity with increased type 1 diabetes progression but a comprehensive assessment of the impact of screening for IA-2A positivity across the natural history of autoantibody positivity has not been performed. We asked whether IA-2A would consistently be associated with higher risk of progression within and across established stages of type 1 diabetes in a large natural history study. METHODS Genetic, autoantibody and metabolic data from adult and paediatric autoantibody-negative (n=192) and autoantibody-positive (n=4577) relatives of individuals with type 1 diabetes followed longitudinally in the Type 1 Diabetes TrialNet Pathway to Prevention Study were analysed. Cox regression was used to compare cumulative incidences of clinical diabetes by autoantibody profiles and disease stages. RESULTS Compared with IA-2A- individuals, IA-2A+ individuals had higher genetic risk scores and clinical progression risk within single-autoantibody-positive (5.3-fold increased 5 year risk), stage 1 (2.2-fold increased 5 year risk) and stage 2 (1.3-fold increased 5 year risk) type 1 diabetes categories. Individuals with single-autoantibody positivity for IA-2A showed increased metabolic dysfunction and diabetes progression compared with people who were autoantibody negative, those positive for another single autoantibody, and IA-2A- stage 1 individuals. Individuals at highest risk within the single-IA-2A+ category included children (HR 14.2 [95% CI 1.9, 103.1], p=0.009), individuals with IA-2A titres above the median (HR 3.5 [95% CI 1.9, 6.6], p<0.001), individuals with high genetic risk scores (HR 1.4 [95% CI 1.2,1.6], p<0.001) and individuals with HLA DR4-positive status (HR 3.7 [95% CI 1.6, 8.3], p=0.002). When considering all autoantibody-positive individuals, progression risk was similar for euglycaemic IA-2A+ individuals and dysglycaemic IA-2A- individuals. CONCLUSIONS/INTERPRETATION IA-2A positivity is consistently associated with increased progression risk throughout the natural history of type 1 diabetes development. Individuals with single-autoantibody positivity for IA-2A have a greater risk of disease progression than those who meet stage 1 criteria but who are IA-2A-. Approaches to incorporate IA-2A+ status into monitoring strategies for autoantibody-positive individuals should be considered.
Collapse
Affiliation(s)
- Emily K Sims
- Department of Pediatrics, Wells Center for Pediatric Research, Division of Pediatric Endocrinology and Diabetology, Indiana University School of Medicine, Indianapolis, IN, USA.
| | - David Cuthbertson
- Health Informatics Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Lauric A Ferrat
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK
| | - Emanuele Bosi
- Diabetes Research Institute, University Vita-Salute San Raffaele, Milan, Italy
- IRCCS San Raffaele Hospital, Milan, Italy
| | - Carmella Evans-Molina
- Department of Pediatrics, Wells Center for Pediatric Research, Division of Pediatric Endocrinology and Diabetology, Indiana University School of Medicine, Indianapolis, IN, USA
- Richard L. Roudebush VA Medical Center, Indianapolis, IN, USA
| | - Linda A DiMeglio
- Department of Pediatrics, Wells Center for Pediatric Research, Division of Pediatric Endocrinology and Diabetology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Brandon M Nathan
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | - Heba M Ismail
- Department of Pediatrics, Wells Center for Pediatric Research, Division of Pediatric Endocrinology and Diabetology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Laura M Jacobsen
- Departments of Pediatrics and Pathology, Diabetes Institute, University of Florida, Gainesville, FL, USA
| | - Maria J Redondo
- Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
| | - Richard A Oram
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK
- The Academic Renal Unit, Royal Devon University Healthcare NHS Foundation Trust, Exeter, UK
| | | |
Collapse
|
3
|
Lernmark Å, Agardh D, Akolkar B, Gesualdo P, Hagopian WA, Haller MJ, Hyöty H, Johnson SB, Elding Larsson H, Liu E, Lynch KF, McKinney EF, McIndoe R, Melin J, Norris JM, Rewers M, Rich SS, Toppari J, Triplett E, Vehik K, Virtanen SM, Ziegler AG, Schatz DA, Krischer J. Looking back at the TEDDY study: lessons and future directions. Nat Rev Endocrinol 2025; 21:154-165. [PMID: 39496810 PMCID: PMC11825287 DOI: 10.1038/s41574-024-01045-0] [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] [Accepted: 09/26/2024] [Indexed: 11/06/2024]
Abstract
The goal of the TEDDY (The Environmental Determinants of Diabetes in the Young) study is to elucidate factors leading to the initiation of islet autoimmunity (first primary outcome) and those related to progression to type 1 diabetes mellitus (T1DM; second primary outcome). This Review outlines the key findings so far, particularly related to the first primary outcome. The background, history and organization of the study are discussed. Recruitment and follow-up (from age 4 months to 15 years) of 8,667 children showed high retention and compliance. End points of the presence of autoantibodies against insulin, GAD65, IA-2 and ZnT8 revealed the HLA-associated early appearance of insulin autoantibodies (1-3 years of age) and the later appearance of GAD65 autoantibodies. Competing autoantibodies against tissue transglutaminase (marking coeliac disease autoimmunity) also appeared early (2-4 years). Genetic and environmental factors, including enterovirus infection and gastroenteritis, support mechanistic differences underlying one phenotype of autoimmunity against insulin and another against GAD65. Infant growth and both probiotics and high protein intake affect the two phenotypes differently, as do serious life events during pregnancy. As the end of the TEDDY sampling phase is approaching, major omics approaches are in progress to further dissect the mechanisms that might explain the two possible endotypes of T1DM.
Collapse
Affiliation(s)
- Åke Lernmark
- Department of Clinical Sciences, Lund University CRC, Skåne University Hospital, Malmö, Sweden.
| | - Daniel Agardh
- Department of Clinical Sciences, Lund University CRC, Skåne University Hospital, Malmö, Sweden
| | - Beena Akolkar
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Patricia Gesualdo
- Barbara Davis Center for Diabetes, University of Colorado, Aurora, CO, USA
| | - William A Hagopian
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Michael J Haller
- Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - Heikki Hyöty
- Department of Virology, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Suzanne Bennett Johnson
- Department of Behavioral Sciences and Social Medicine, Florida State University College of Medicine, Tallahassee, FL, USA
| | - Helena Elding Larsson
- Department of Clinical Sciences, Lund University CRC, Skåne University Hospital, Malmö, Sweden
| | - Edwin Liu
- Digestive Health Institute, Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kristian F Lynch
- Health Informatics Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Eoin F McKinney
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Richard McIndoe
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Jessica Melin
- Department of Clinical Sciences, Lund University CRC, Skåne University Hospital, Malmö, Sweden
| | - Jill M Norris
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Marian Rewers
- Barbara Davis Center for Diabetes, University of Colorado, Aurora, CO, USA
| | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Jorma Toppari
- Department of Paediatrics, Turku University Hospital, Turku, Finland
- Institute of Biomedicine, Research Centre for Integrated Physiology and Pharmacology, University of Turku, Turku, Finland
| | - Eric Triplett
- University of Florida, Department of Microbiology and Cell Science, Gainesville, FL, USA
| | - Kendra Vehik
- Health Informatics Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Suvi M Virtanen
- Center for Child Health Research, Tampere University and University Hospital and Research, Tampere, Finland
| | - Anette-G Ziegler
- Institute of Diabetes Research, Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt, Munich, Germany
- Forschergruppe Diabetes e.V. at Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt, Munich, Germany
- Forschergruppe Diabetes, Klinikum rechts der Isar, Technische Universität München and e.V., Munich, Germany
| | - Desmond A Schatz
- Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - Jeffrey Krischer
- Health Informatics Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| |
Collapse
|
4
|
Ling EM, Lemos JRN, Hirani K, von Herrath M. Type 1 diabetes: immune pathology and novel therapeutic approaches. Diabetol Int 2024; 15:761-776. [PMID: 39469552 PMCID: PMC11512973 DOI: 10.1007/s13340-024-00748-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: 03/07/2024] [Accepted: 07/17/2024] [Indexed: 10/30/2024]
Abstract
Type 1 diabetes (T1D) is characterized by the progressive destruction of insulin-producing beta cells in the pancreas. Despite improvements in insulin monitoring techniques, there remains no cure for T1D. Individuals with T1D require lifelong insulin therapy and some develop life-threatening complications. T1D is a complex, multifactorial, autoimmune condition. Understanding why people get T1D and how it progresses has advanced our knowledge of the disease and led to the discovery of specific targets that can be therapeutically manipulated to halt or reverse the course of T1D. Scientists investigating the potential of immunotherapy treatment for the treatment have recently had some encouraging results. Teplizumab, an anti-CD3 monoclonal antibody that has been approved by the FDA, delays the onset of clinical T1D in patients ≥ 8 years of age with preclinical T1D and improves beta cell function. Therapies targeting beta cell health, vitality, and function are now thought to be an essential component of successful combination therapy for T1D. The idea that the beta cells themselves may influence their own destruction during the development of T1D is a notion that has recently been gaining acceptance in the field. Researchers have recently made remarkable strides in beta cell replacement therapy and beta cell regeneration techniques. This review offers a detailed exploration of the pathophysiological mechanisms of T1D. It discusses the intricate interplay of factors leading to T1D development and the innovative approaches being explored to discover new treatments and a cure for the millions of people living with T1D worldwide.
Collapse
Affiliation(s)
- Eleanor M. Ling
- Diabetes Research Institute (DRI), University of Miami Miller School of Medicine, Miami, FL USA
| | - Joana R. N. Lemos
- Diabetes Research Institute (DRI), University of Miami Miller School of Medicine, Miami, FL USA
- Division of Endocrine, Diabetes, and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL USA
| | - Khemraj Hirani
- Diabetes Research Institute (DRI), University of Miami Miller School of Medicine, Miami, FL USA
- Division of Endocrine, Diabetes, and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL USA
| | - Matthias von Herrath
- Diabetes Research Institute (DRI), University of Miami Miller School of Medicine, Miami, FL USA
- Division of Endocrine, Diabetes, and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL USA
- Global Chief Medical Office, Novo Nordisk A/S, Søborg, Denmark
| |
Collapse
|
5
|
Zhao LP, Papadopoulos GK, Skyler JS, Parikh HM, Kwok WW, Bondinas GP, Moustakas AK, Wang R, Pyo CW, Nelson WC, Geraghty DE, Lernmark Å. Oral Insulin Delay of Stage 3 Type 1 Diabetes Revisited in HLA DR4-DQ8 Participants in the TrialNet Oral Insulin Prevention Trial (TN07). Diabetes Care 2024; 47:1608-1616. [PMID: 38949847 PMCID: PMC11362107 DOI: 10.2337/dc24-0573] [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: 03/18/2024] [Accepted: 06/04/2024] [Indexed: 07/02/2024]
Abstract
OBJECTIVE To explore if oral insulin could delay onset of stage 3 type 1 diabetes (T1D) among patients with stage 1/2 who carry HLA DR4-DQ8 and/or have elevated levels of IA-2 autoantibodies (IA-2As). RESEARCH AND METHODS Next-generation targeted sequencing technology was used to genotype eight HLA class II genes (DQA1, DQB1, DRB1, DRB3, DRB4, DRB5, DPA1, and DPB1) in 546 participants in the TrialNet oral insulin preventative trial (TN07). Baseline levels of autoantibodies against insulin (IAA), GAD65 (GADA), and IA-2A were determined prior to treatment assignment. Available clinical and demographic covariables from TN07 were used in this post hoc analysis with the Cox regression model to quantify the preventive efficacy of oral insulin. RESULTS Oral insulin reduced the frequency of T1D onset among participants with elevated IA-2A levels (HR 0.62; P = 0.012) but had no preventive effect among those with low IA-2A levels (HR 1.03; P = 0.91). High IA-2A levels were positively associated with the HLA DR4-DQ8 haplotype (OR 1.63; P = 6.37 × 10-6) and negatively associated with the HLA DR7-containing DRB1*07:01-DRB4*01:01-DQA1*02:01-DQB1*02:02 extended haplotype (OR 0.49; P = 0.037). Among DR4-DQ8 carriers, oral insulin delayed the progression toward stage 3 T1D onset (HR 0.59; P = 0.027), especially if participants also had high IA-2A level (HR 0.50; P = 0.028). CONCLUSIONS These results suggest the presence of a T1D endotype characterized by HLA DR4-DQ8 and/or elevated IA-2A levels; for those patients with stage 1/2 disease with such an endotype, oral insulin delays the clinical T1D onset.
Collapse
Affiliation(s)
- Lue Ping Zhao
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- School of Public Health, University of Washington, Seattle, WA
| | - George K. Papadopoulos
- Laboratory of Biophysics, Biochemistry, Biomaterials and Bioprocessing, Faculty of Agricultural Technology, Technological Educational Institute of Epirus, Arta, Greece
| | - Jay S. Skyler
- Diabetes Research Institute and Division of Endocrinology, Diabetes, and Metabolism, University of Miami Miller School of Medicine, Miami, FL
| | - Hemang M. Parikh
- Health Informatics Institute, Morsani College of Medicine, University of South Florida, Tampa, FL
| | | | - George P. Bondinas
- Department of Food Science and Technology, Faculty of Environmental Sciences, Ionian University, Argostoli, Cephalonia, Greece
| | - Antonis K. Moustakas
- Department of Food Science and Technology, Faculty of Environmental Sciences, Ionian University, Argostoli, Cephalonia, Greece
| | - 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
|
6
|
Sims EK, Cuthbertson D, Jacobsen L, Ismail HM, Nathan BM, Herold KC, Redondo MJ, Sosenko J. Comparisons of Metabolic Measures to Predict T1D vs Detect a Preventive Treatment Effect in High-Risk Individuals. J Clin Endocrinol Metab 2024; 109:2116-2123. [PMID: 38267821 PMCID: PMC11244203 DOI: 10.1210/clinem/dgae048] [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: 09/20/2023] [Revised: 12/19/2023] [Accepted: 01/23/2024] [Indexed: 01/26/2024]
Abstract
CONTEXT Metabolic measures are frequently used to predict type 1 diabetes (T1D) and to understand effects of disease-modifying therapies. OBJECTIVE Compare metabolic endpoints for their ability to detect preventive treatment effects and predict T1D. METHODS Six-month changes in metabolic endpoints were assessed for (1) detecting treatment effects by comparing placebo and treatment arms from the randomized controlled teplizumab prevention trial, a multicenter clinical trial investigating 14-day intravenous teplizumab infusion and (2) predicting T1D in the TrialNet Pathway to Prevention natural history study. For each metabolic measure, t-Values from t tests for detecting a treatment effect were compared with chi-square values from proportional hazards regression for predicting T1D. Participants in the teplizumab prevention trial and participants in the Pathway to Prevention study selected with the same inclusion criteria used for the teplizumab trial were studied. RESULTS Six-month changes in glucose-based endpoints predicted diabetes better than C-peptide-based endpoints, yet the latter were better at detecting a teplizumab effect. Combined measures of glucose and C-peptide were more balanced than measures of glucose alone or C-peptide alone for predicting diabetes and detecting a teplizumab effect. CONCLUSION The capacity of a metabolic endpoint to detect a treatment effect does not necessarily correspond to its accuracy for predicting T1D. However, combined glucose and C-peptide endpoints appear to be effective for both predicting diabetes and detecting a response to immunotherapy. These findings suggest that combined glucose and C-peptide endpoints should be incorporated into the design of future T1D prevention trials.
Collapse
Affiliation(s)
- Emily K Sims
- Department of Pediatrics, Wells Center for Pediatric Research, Pediatric Endocrinology and Diabetology, and the Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - David Cuthbertson
- Department of Pediatrics, Pediatrics Epidemiology Center, Morsani College of Medicine, University of South Florida, Tampa, FL 33606, USA
| | - Laura Jacobsen
- Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Heba M Ismail
- Department of Pediatrics, Wells Center for Pediatric Research, Pediatric Endocrinology and Diabetology, and the Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Brandon M Nathan
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Kevan C Herold
- Division of Diabetes and Endocrinology, Yale University, New Haven, CT 06520, USA
- Departments of Immunobiology and Internal Medicine, Yale University, New Haven, CT 06520, USA
| | - Maria J Redondo
- Texas Children's Hospital, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jay Sosenko
- Department of Medicine, Division of Diabetes, Metabolism, and Endocrinology, University of Miami, Miami, FL 33136, USA
- Diabetes Research Institute, University of Miami, Miami, FL 33136, USA
| |
Collapse
|
7
|
Linsley PS, Nakayama M, Balmas E, Chen J, Barahmand-Pour-Whitman F, Bansal S, Bottorff T, Serti E, Speake C, Pugliese A, Cerosaletti K. Germline-like TCR-α chains shared between autoreactive T cells in blood and pancreas. Nat Commun 2024; 15:4971. [PMID: 38871688 PMCID: PMC11176301 DOI: 10.1038/s41467-024-48833-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Accepted: 05/13/2024] [Indexed: 06/15/2024] Open
Abstract
Human type 1 diabetes (T1D) is caused by autoimmune attack on the insulin-producing pancreatic beta cells by islet antigen-reactive T cells. How human islet antigen-reactive (IAR) CD4+ memory T cells from peripheral blood affect T1D progression in the pancreas is poorly understood. Here, we aim to determine if IAR T cells in blood could be detected in pancreas. We identify paired αβ (TRA/TRB) T cell receptors (TCRs) in IAR T cells from the blood of healthy, at-risk, new-onset, and established T1D donors, and measured sequence overlap with TCRs in pancreata from healthy, at risk and T1D organ donors. We report extensive TRA junction sharing between IAR T cells and pancreas-infiltrating T cells (PIT), with perfect-match or single-mismatch TRA junction amino acid sequences comprising ~29% total unique IAR TRA junctions (942/3,264). PIT-matched TRA junctions were largely public and enriched for TRAV41 usage, showing significant nucleotide sequence convergence, increased use of germline-encoded versus non-templated residues in epitope engagement, and a potential for cross-reactivity. Our findings thus link T cells with distinctive germline-like TRA chains in the peripheral blood with T cells in the pancreas.
Collapse
Affiliation(s)
- Peter S Linsley
- Benaroya Research Institute at Virginia Mason, Seattle, WA, USA.
| | - Maki Nakayama
- Barbara Davis Center for Childhood Diabetes, Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Elisa Balmas
- Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| | - Janice Chen
- Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| | | | - Shubham Bansal
- Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| | - Ty Bottorff
- Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| | | | - Cate Speake
- Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| | - Alberto Pugliese
- Department of Diabetes Immunology & The Wanek Family Project for Type 1 Diabetes, Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope, Duarte, CA, USA
| | | |
Collapse
|
8
|
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
|
9
|
Xie QY, Oh S, Wong A, Yau C, Herold KC, Danska JS. Immune responses to gut bacteria associated with time to diagnosis and clinical response to T cell-directed therapy for type 1 diabetes prevention. Sci Transl Med 2023; 15:eadh0353. [PMID: 37878676 DOI: 10.1126/scitranslmed.adh0353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 10/02/2023] [Indexed: 10/27/2023]
Abstract
Immune-targeted therapies have efficacy for treatment of autoinflammatory diseases. For example, treatment with the T cell-specific anti-CD3 antibody teplizumab delayed disease onset in participants at high risk for type 1 diabetes (T1D) in the TrialNet 10 (TN-10) trial. However, heterogeneity in therapeutic responses in TN-10 and other immunotherapy trials identifies gaps in understanding disease progression and treatment responses. The intestinal microbiome is a potential source of biomarkers associated with future T1D diagnosis and responses to immunotherapy. We previously reported that antibody responses to gut commensal bacteria were associated with T1D diagnosis, suggesting that certain antimicrobial immune responses may help predict disease onset. Here, we investigated anticommensal antibody (ACAb) responses against a panel of taxonomically diverse intestinal bacteria species in sera from TN-10 participants before and after teplizumab or placebo treatment. We identified IgG2 responses to three species that were associated with time to T1D diagnosis and with teplizumab treatment responses that delayed disease onset. These antibody responses link human intestinal bacteria with T1D progression, adding predictive value to known T1D risk factors. ACAb analysis provides a new approach to elucidate heterogeneity in responses to immunotherapy and identify individuals who may benefit from teplizumab, recently approved by the U.S. Food and Drug Administration for delaying T1D onset.
Collapse
Affiliation(s)
- Quin Yuhui Xie
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5T2S8, Canada
- Genetics and Genome Biology, Hospital for Sick Children, Toronto, Ontario M5G1X8, Canada
| | - Sean Oh
- Genetics and Genome Biology, Hospital for Sick Children, Toronto, Ontario M5G1X8, Canada
| | - Anthony Wong
- Genetics and Genome Biology, Hospital for Sick Children, Toronto, Ontario M5G1X8, Canada
| | - Christopher Yau
- Genetics and Genome Biology, Hospital for Sick Children, Toronto, Ontario M5G1X8, Canada
- Department of Immunology, University of Toronto, Toronto, Ontario M5T2S8, Canada
| | - Kevan C Herold
- Department of Immunobiology, Yale University, New Haven, CT 06520, USA
- Department of Internal Medicine, Yale University, New Haven, CT 06520, USA
| | - Jayne S Danska
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5T2S8, Canada
- Genetics and Genome Biology, Hospital for Sick Children, Toronto, Ontario M5G1X8, Canada
- Department of Immunology, University of Toronto, Toronto, Ontario M5T2S8, Canada
| |
Collapse
|
10
|
Linsley P, Nakayama M, Balmas E, Chen J, Pour F, Bansal S, Serti E, Speake C, Pugliese A, Cerosaletti K. Self-reactive germline-like TCR alpha chains shared between blood and pancreas. RESEARCH SQUARE 2023:rs.3.rs-3446917. [PMID: 37886513 PMCID: PMC10602137 DOI: 10.21203/rs.3.rs-3446917/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
Human islet antigen reactive CD4 + memory T cells (IAR T cells) from peripheral blood have been studied extensively for their role in the pathogenesis of autoimmune type 1 diabetes (T1D). However, IAR T cells are rare, and it remains poorly understood how they affect T1D progression in the pancreas. Using single cell RNA-sequencing coupled with a multiplexed activation induced marker (AIM) enrichment assay, we identified paired TCR alpha/beta (TRA/TRB) T cell receptors (TCRs) in IAR T cells from the blood of healthy, at-risk, new onset, and established T1D donors. Using TCR sequences as barcodes, we measured infiltration of IAR T cells from blood into pancreas of organ donors with and without T1D. We detected extensive TCR sharing between IAR T cells from peripheral blood and pancreatic infiltrating T cells (PIT), with perfectly matched or single mismatched TRA junctions and J gene regions, comprising ~ 34% of unique IAR TCRs. PIT-matching IAR T cells had public TRA chains that showed increased use of germline-encoded residues in epitope engagement and a propensity for cross-reactivity. The link with T cells in the pancreas implicates autoreactive IAR T cells with shared TRA junctions and increased levels in blood with the prediabetic and new onset phases of T1D progression.
Collapse
|
11
|
Shapiro MR, Dong X, Perry DJ, McNichols JM, Thirawatananond P, Posgai AL, Peters LD, Motwani K, Musca RS, Muir A, Concannon P, Jacobsen LM, Mathews CE, Wasserfall CH, Haller MJ, Schatz DA, Atkinson MA, Brusko MA, Bacher R, Brusko TM. Human immune phenotyping reveals accelerated aging in type 1 diabetes. JCI Insight 2023; 8:e170767. [PMID: 37498686 PMCID: PMC10544250 DOI: 10.1172/jci.insight.170767] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 07/19/2023] [Indexed: 07/29/2023] Open
Abstract
The proportions and phenotypes of immune cell subsets in peripheral blood undergo continual and dramatic remodeling throughout the human life span, which complicates efforts to identify disease-associated immune signatures in type 1 diabetes (T1D). We conducted cross-sectional flow cytometric immune profiling on peripheral blood from 826 individuals (stage 3 T1D, their first-degree relatives, those with ≥2 islet autoantibodies, and autoantibody-negative unaffected controls). We constructed an immune age predictive model in unaffected participants and observed accelerated immune aging in T1D. We used generalized additive models for location, shape, and scale to obtain age-corrected data for flow cytometry and complete blood count readouts, which can be visualized in our interactive portal (ImmScape); 46 parameters were significantly associated with age only, 25 with T1D only, and 23 with both age and T1D. Phenotypes associated with accelerated immunological aging in T1D included increased CXCR3+ and programmed cell death 1-positive (PD-1+) frequencies in naive and memory T cell subsets, despite reduced PD-1 expression levels on memory T cells. Phenotypes associated with T1D after age correction were predictive of T1D status. Our findings demonstrate advanced immune aging in T1D and highlight disease-associated phenotypes for biomarker monitoring and therapeutic interventions.
Collapse
Affiliation(s)
- Melanie R. Shapiro
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, and
- Diabetes Institute and
| | - Xiaoru Dong
- Diabetes Institute and
- Department of Biostatistics, College of Public Health and Health Professions, University of Florida, Gainesville, Florida, USA
| | - Daniel J. Perry
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, and
- Diabetes Institute and
| | - James M. McNichols
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, and
- Diabetes Institute and
| | - Puchong Thirawatananond
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, and
- Diabetes Institute and
| | - Amanda L. Posgai
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, and
- Diabetes Institute and
| | - Leeana D. Peters
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, and
- Diabetes Institute and
| | - Keshav Motwani
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, and
- Diabetes Institute and
| | - Richard S. Musca
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, and
- Diabetes Institute and
| | - Andrew Muir
- Department of Pediatrics, Emory University, Atlanta, Georgia, USA
| | - Patrick Concannon
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, and
- Diabetes Institute and
- Genetics Institute and
| | - Laura M. Jacobsen
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, and
- Diabetes Institute and
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Clayton E. Mathews
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, and
- Diabetes Institute and
| | - Clive H. Wasserfall
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, and
- Diabetes Institute and
| | - Michael J. Haller
- Diabetes Institute and
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Desmond A. Schatz
- Diabetes Institute and
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Mark A. Atkinson
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, and
- Diabetes Institute and
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Maigan A. Brusko
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, and
- Diabetes Institute and
| | - Rhonda Bacher
- Diabetes Institute and
- Department of Biostatistics, College of Public Health and Health Professions, University of Florida, Gainesville, Florida, USA
| | - Todd M. Brusko
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, and
- Diabetes Institute and
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
12
|
Luckett AM, Weedon MN, Hawkes G, Leslie RD, Oram RA, Grant SFA. Utility of genetic risk scores in type 1 diabetes. Diabetologia 2023; 66:1589-1600. [PMID: 37439792 PMCID: PMC10390619 DOI: 10.1007/s00125-023-05955-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 05/23/2023] [Indexed: 07/14/2023]
Abstract
Iterative advances in understanding of the genetics of type 1 diabetes have identified >70 genetic regions associated with risk of the disease, including strong associations across the HLA class II region that account for >50% of heritability. The increased availability of genetic data combined with the decreased costs of generating these data, have facilitated the development of polygenic scores that aggregate risk variants from associated loci into a single number: either a genetic risk score (GRS) or a polygenic risk score (PRS). PRSs incorporate the risk of many possibly correlated variants from across the genome, even if they do not reach genome-wide significance, whereas GRSs estimate the cumulative contribution of a smaller subset of genetic variants that reach genome-wide significance. Type 1 diabetes GRSs have utility in diabetes classification, aiding discrimination between type 1 diabetes, type 2 diabetes and MODY. Type 1 diabetes GRSs are also being used in newborn screening studies to identify infants at risk of future presentation of the disease. Most early studies of type 1 diabetes genetics have been conducted in European ancestry populations, but, to develop accurate GRSs across diverse ancestries, large case-control cohorts from non-European populations are still needed. The current barriers to GRS implementation within healthcare are mainly related to a lack of guidance and knowledge on integration with other biomarkers and clinical variables. Once these limitations are addressed, there is huge potential for 'test and treat' approaches to be used to tailor care for individuals with type 1 diabetes.
Collapse
Affiliation(s)
- Amber M Luckett
- University of Exeter College of Medicine and Health, Exeter, UK
| | | | - Gareth Hawkes
- University of Exeter College of Medicine and Health, Exeter, UK
| | - R David Leslie
- Blizard Institute, Queen Mary University of London, London, UK.
| | - Richard A Oram
- University of Exeter College of Medicine and Health, Exeter, UK.
- Royal Devon University Healthcare NHS Foundation Trust, Exeter, UK.
| | - Struan F A Grant
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Division of Diabetes and Endocrinology, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
13
|
Morales JF, Muse R, Podichetty JT, Burton J, David S, Lang P, Schmidt S, Romero K, O'Doherty I, Martin F, Campbell‐Thompson M, Haller MJ, Atkinson MA, Kim S. Disease progression joint model predicts time to type 1 diabetes onset: Optimizing future type 1 diabetes prevention studies. CPT Pharmacometrics Syst Pharmacol 2023; 12:1016-1028. [PMID: 37186151 PMCID: PMC10349195 DOI: 10.1002/psp4.12973] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 04/07/2023] [Accepted: 04/11/2023] [Indexed: 05/17/2023] Open
Abstract
Clinical trials seeking type 1 diabetes prevention are challenging in terms of identifying patient populations likely to progress to type 1 diabetes within limited (i.e., short-term) trial durations. Hence, we sought to improve such efforts by developing a quantitative disease progression model for type 1 diabetes. Individual-level data obtained from the TrialNet Pathway to Prevention and The Environmental Determinants of Diabetes in the Young natural history studies were used to develop a joint model that links the longitudinal glycemic measure to the timing of type 1 diabetes diagnosis. Baseline covariates were assessed using a stepwise covariate modeling approach. Our study focused on individuals at risk of developing type 1 diabetes with the presence of two or more diabetes-related autoantibodies (AAbs). The developed model successfully quantified how patient features measured at baseline, including HbA1c and the presence of different AAbs, alter the timing of type 1 diabetes diagnosis with reasonable accuracy and precision (<30% RSE). In addition, selected covariates were statistically significant (p < 0.0001 Wald test). The Weibull model best captured the timing to type 1 diabetes diagnosis. The 2-h oral glucose tolerance values assessed at each visit were included as a time-varying biomarker, which was best quantified using the sigmoid maximum effect function. This model provides a framework to quantitatively predict and simulate the time to type 1 diabetes diagnosis in individuals at risk of developing the disease and thus, aligns with the needs of pharmaceutical companies and scientists seeking to advance therapies aimed at interdicting the disease process.
Collapse
Affiliation(s)
- Juan Francisco Morales
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of PharmacyUniversity of FloridaFloridaOrlandoUSA
| | | | | | | | | | | | - Stephan Schmidt
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of PharmacyUniversity of FloridaFloridaOrlandoUSA
| | | | | | | | - Martha Campbell‐Thompson
- Department of Pathology, Immunology, and Laboratory MedicineDiabetes Institute, College of Medicine, University of FloridaFloridaGainesvilleUSA
| | - Michael J. Haller
- Department of PediatricsDiabetes Institute, College of Medicine, University of FloridaFloridaGainesvilleUSA
| | - Mark A. Atkinson
- Department of Pathology, Immunology, and Laboratory MedicineDiabetes Institute, College of Medicine, University of FloridaFloridaGainesvilleUSA
- Department of PediatricsDiabetes Institute, College of Medicine, University of FloridaFloridaGainesvilleUSA
| | - Sarah Kim
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of PharmacyUniversity of FloridaFloridaOrlandoUSA
| |
Collapse
|
14
|
Ghalwash M, Anand V, Lou O, Martin F, Rewers M, Ziegler AG, Toppari J, Hagopian WA, Veijola R. Islet autoantibody screening in at-risk adolescents to predict type 1 diabetes until young adulthood: a prospective cohort study. THE LANCET. CHILD & ADOLESCENT HEALTH 2023; 7:261-268. [PMID: 36681087 PMCID: PMC10038928 DOI: 10.1016/s2352-4642(22)00350-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/21/2022] [Accepted: 11/23/2022] [Indexed: 01/20/2023]
Abstract
BACKGROUND Screening for islet autoantibodies in children and adolescents identifies individuals who will later develop type 1 diabetes, allowing patient and family education to prevent diabetic ketoacidosis at onset and to enable consideration of preventive therapies. We aimed to assess whether islet autoantibody screening is effective for predicting type 1 diabetes in adolescents aged 10-18 years with an increased risk of developing type 1 diabetes. METHODS Data were harmonised from prospective studies from Finland (the Diabetes Prediction and Prevention study), Germany (the BABYDIAB study), and the USA (Diabetes Autoimmunity Study in the Young and the Diabetes Evaluation in Washington study). Autoantibodies against insulin, glutamic acid decarboxylase, and insulinoma-associated protein 2 were measured at each follow-up visit. Children who were lost to follow-up or diagnosed with type 1 diabetes before 10 years of age were excluded. Inverse probability censoring weighting was used to include data from remaining participants. Sensitivity and the positive predictive value of these autoantibodies, tested at one or two ages, to predict type 1 diabetes by the age of 18 years were the main outcomes. FINDINGS Of 20 303 children with an increased type 1 diabetes risk, 8682 were included for the analysis with inverse probability censoring weighting. 1890 were followed up to 18 years of age or developed type 1 diabetes between the ages of 10 years and 18 years, and their median follow-up was 18·3 years (IQR 14·5-20·3). 442 (23·4%) of 1890 adolescents were positive for at least one islet autoantibody, and 262 (13·9%) developed type 1 diabetes. Time from seroconversion to diabetes diagnosis increased by 0·64 years (95% CI 0·34-0·95) for each 1-year increment of diagnosis age (Pearson's correlation coefficient 0·88, 95% CI 0·50-0·97, p=0·0020). The median interval between the last prediagnostic sample and diagnosis was 0·3 years (IQR 0·1-1·3) in the 227 participants who were autoantibody positive and 6·8 years (1·6-9·9) for the 35 who were autoantibody negative. Single screening at the age of 10 years was 90% (95% CI 86-95) sensitive, with a positive predictive value of 66% (60-72) for clinical diabetes. Screening at two ages (10 years and 14 years) increased sensitivity to 93% (95% CI 89-97) but lowered the positive predictive value to 55% (49-60). INTERPRETATION Screening of adolescents at risk for type 1 diabetes only once at 10 years of age for islet autoantibodies was highly effective to detect type 1 diabetes by the age of 18 years, which in turn could enable prevention of diabetic ketoacidosis and participation in secondary prevention trials. FUNDING JDRF International.
Collapse
Affiliation(s)
- Mohamed Ghalwash
- Center for Computational Health, IBM Research, Yorktown Heights, NY, USA; Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Vibha Anand
- Center for Computational Health, IBM Research, Cambridge, MA, USA
| | | | | | - Marian Rewers
- Barbara Davis Center for Diabetes, University of Colorado, Denver, CO, USA
| | - Anette-G Ziegler
- Forschergruppe Diabetes and Institute of Diabetes Research, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany der TU München, Munich, Germany
| | - Jorma Toppari
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, and Centre for Population Health Research, University of Turku, Turku, Finland; Department of Pediatrics, Turku University Hospital, Turku, Finland
| | | | - Riitta Veijola
- Department of Pediatrics, Research Unit of Clinical Medicine, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland.
| |
Collapse
|
15
|
Jia X, Yu L. Effective assay technologies fit for large-scale population screening of type 1 diabetes. FRONTIERS IN CLINICAL DIABETES AND HEALTHCARE 2023; 3:1034698. [PMID: 36992730 PMCID: PMC10012058 DOI: 10.3389/fcdhc.2022.1034698] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 12/30/2022] [Indexed: 01/24/2023]
Abstract
While worldwide prevention efforts for type 1 diabetes (T1D) are underway to abrogate or slow progression to diabetes, mass screening of islet autoantibodies (IAbs) in the general population is urgently needed. IAbs, the most reliable biomarkers, play an essential role in prediction and clinical diagnosis of T1D. Through laboratory proficiency programs and harmonization efforts, a radio-binding assay (RBA) has been well established as the current 'gold' standard assay for all four IAbs. However, in view of the need for large-scale screening in the non-diabetic population, RBA consistently faces two fundamental challenges, cost-efficiency and disease specificity. While all four IAbs are important for disease prediction, the RBA platform, with a separate IAb test format is laborious, inefficient and expensive. Furthermore, the majority of IAb positivity in screening, especially from individuals with single IAb were found to be low risk with low affinity. It is well documented from multiple clinical studies that IAbs with low affinity are low risk with less or no disease relevance. At present, two non-radioactive multiplex assays, a 3-assay ELISA combining three IAbs and a multiplex ECL assay combining all four IAbs, have been successfully used as the primary methods for general population screenings in Germany and the US, respectively. Recently, the TrialNet Pathway to Prevention study has been organizing an IAb workshop which aims to analyze the 5-year T1D predictive values of IAbs. A T1D-specific assay with high efficiency, low cost and requiring low volume of sample will definitely be necessary to benefit general population screening.
Collapse
Affiliation(s)
| | - Liping Yu
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, CO, United States
| |
Collapse
|
16
|
Kero J, Koskenniemi JJ, Karsikas S, Pokka T, Lou O, Toppari J, Veijola R. INnoVative trial design for testing the Efficacy, Safety and Tolerability of 6-month treatment with incretin-based therapy to prevent type 1 DIAbetes in autoantibody positive participants: A protocol for three parallel double-blind, randomised controlled trials (INVESTDIA). Diabet Med 2022; 39:e14913. [PMID: 35797241 PMCID: PMC9540026 DOI: 10.1111/dme.14913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 06/30/2022] [Accepted: 07/06/2022] [Indexed: 11/28/2022]
Abstract
AIMS β-cell stress and dysfunction may contribute to islet autoimmunity and progression to clinical type 1 diabetes. We present a protocol of three randomised controlled trials assessing the effects of glucagon-like peptide 1 (GLP - 1) analogue liraglutide in three early stages of type 1 diabetes. METHODS We will test 10- to 30-year-old people with multiple islet autoantibodies for their glucose metabolism and randomise participants with stage 1 (multiple islet autoantibodies and normoglycaemia), stage 2 (multiple islet autoantibodies and dysglycaemia) and early stage 3 (clinical diagnosis) type 1 diabetes, 10-14 persons in each, to a 6-month intervention with liraglutide or placebo with 6-month follow-up in the stage 2 and stage 3 trials and 18-month follow-up in the stage 1 trial. Primary efficacy outcome in the stage 1 and stage 2 trials is a first-phase insulin response in an intravenous glucose tolerance test and C-peptide area under the curve in a 2-h mixed-meal tolerance test in the stage 3 trial. In addition, safety and tolerability of liraglutide treatment will be assessed. CONCLUSIONS Most prevention trials of type 1 diabetes have targeted the immune system. Treatment with GLP-1 analogue liraglutide supports the pancreatic β-cells, which should likewise attenuate islet autoimmunity. Our innovative study design allows simultaneous investigation of an intervention in three groups of people who represent various early stages of type 1 diabetes and maximises the eligibility to participate. TRIAL REGISTRATION NCT02611232 (stage 1 trial), NCT02898506 (stage 2 trial), NCT02908087 (stage 3 trial).
Collapse
Affiliation(s)
- Jukka Kero
- Research Centre for Integrative Physiology and Pharmacology, Institute of BiomedicineUniversity of TurkuTurkuFinland
- Department of PaediatricsTurku University HospitalTurkuFinland
- Centre for Population Health ResearchUniversity of Turku and Turku University HospitalTurkuFinland
| | - Jaakko J. Koskenniemi
- Research Centre for Integrative Physiology and Pharmacology, Institute of BiomedicineUniversity of TurkuTurkuFinland
- Department of PaediatricsTurku University HospitalTurkuFinland
- Centre for Population Health ResearchUniversity of Turku and Turku University HospitalTurkuFinland
| | - Sara Karsikas
- Department of PaediatricsTurku University HospitalTurkuFinland
| | - Tytti Pokka
- Department for Children and AdolescentsOulu University HospitalOuluFinland
- Department of Paediatrics, PEDEGO Research UnitMRC Oulu, University of OuluOuluFinland
| | | | - Jorma Toppari
- Research Centre for Integrative Physiology and Pharmacology, Institute of BiomedicineUniversity of TurkuTurkuFinland
- Centre for Population Health ResearchUniversity of Turku and Turku University HospitalTurkuFinland
| | - Riitta Veijola
- Department for Children and AdolescentsOulu University HospitalOuluFinland
- Department of Paediatrics, PEDEGO Research UnitMRC Oulu, University of OuluOuluFinland
| |
Collapse
|
17
|
Mastrandrea LD, Quattrin T. Preventing type 1 diabetes development and preserving beta-cell function. Curr Opin Endocrinol Diabetes Obes 2022; 29:386-391. [PMID: 35799459 DOI: 10.1097/med.0000000000000746] [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] [Indexed: 01/27/2023]
Abstract
PURPOSE OF REVIEW Type 1 diabetes (T1D) is the most common chronic disease of childhood presenting a significant burden, both in terms of day-to-day medical management and lifelong care. Studies aligned with diverse strategies to prevent or modify the course of T1D are reviewed. RECENT FINDINGS The diagnosis of T1D precedes the classic clinical presentation when insulin dependence develops. With an increased understanding of the pathophysiology of the autoimmune process leading to T1D, treatment strategies to prevent the development of autoimmunity and/or modify the immune response have been trialed in persons at risk for developing the disease. Interventions prior to insulin dependence or very early after clinical diagnosis show some promise both in preventing disease onset and prolonging beta-cell insulin production. SUMMARY Significant progress has been made in the treatment of T1D. However, suboptimal glycemic control remains a challenge impacting overall health and quality of life for patients with this chronic disease. Although physicians and basic sciences investigators continue to pursue the prevention of the autoimmune process, the advent of disease-modifying agents is a promising strategy. Further studies are needed to ensure that insulin preservation can be achieved longer term.
Collapse
Affiliation(s)
- Lucy D Mastrandrea
- Jacobs School of Medicine and Biomedical Sciences, University at Buffalo
- Diabetes Center, John R. Oishei Children's Hospital, Buffalo, New York, USA
| | - Teresa Quattrin
- Jacobs School of Medicine and Biomedical Sciences, University at Buffalo
- Diabetes Center, John R. Oishei Children's Hospital, Buffalo, New York, USA
| |
Collapse
|
18
|
Dunger DB, Bruggraber SFA, Mander AP, Marcovecchio ML, Tree T, Chmura PJ, Knip M, Schulte AM, Mathieu C. INNODIA Master Protocol for the evaluation of investigational medicinal products in children, adolescents and adults with newly diagnosed type 1 diabetes. Trials 2022; 23:414. [PMID: 35585600 PMCID: PMC9116021 DOI: 10.1186/s13063-022-06259-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 03/30/2022] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND The INNODIA consortium has established a pan-European infrastructure using validated centres to prospectively evaluate clinical data from individuals with newly diagnosed type 1 diabetes combined with centralised collection of clinical samples to determine rates of decline in beta-cell function and identify novel biomarkers, which could be used for future stratification of phase 2 clinical trials. METHODS In this context, we have developed a Master Protocol, based on the "backbone" of the INNODIA natural history study, which we believe could improve the delivery of phase 2 studies exploring the use of single or combinations of Investigational Medicinal Products (IMPs), designed to prevent or reverse declines in beta-cell function in individuals with newly diagnosed type 1 diabetes. Although many IMPs have demonstrated potential efficacy in phase 2 studies, few subsequent phase 3 studies have confirmed these benefits. Currently, phase 2 drug development for this indication is limited by poor evaluation of drug dosage and lack of mechanistic data to understand variable responses to the IMPs. Identification of biomarkers which might permit more robust stratification of participants at baseline has been slow. DISCUSSION The Master Protocol provides (1) standardised assessment of efficacy and safety, (2) comparable collection of mechanistic data, (3) the opportunity to include adaptive designs and the use of shared control groups in the evaluation of combination therapies, and (4) benefits of greater understanding of endpoint variation to ensure more robust sample size calculations and future baseline stratification using existing and novel biomarkers.
Collapse
Affiliation(s)
- David B. Dunger
- Department of Paediatrics, University of Cambridge, Cambridge, UK
- Wellcome Trust-MRC Institute of Metabolic Sciences, University of Cambridge, Cambridge, UK
| | | | - Adrian P. Mander
- Centre for Trials Research, Cardiff University, Cardiff, UK
- NIHR Biomedical Research Centre, Guy’s and St Thomas’ NHS Foundation Trust and King’s College London, London, UK
| | | | - Timothy Tree
- Centre for Trials Research, Cardiff University, Cardiff, UK
- NIHR Biomedical Research Centre, Guy’s and St Thomas’ NHS Foundation Trust and King’s College London, London, UK
| | - Piotr Jaroslaw Chmura
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mikael Knip
- Pediatric Research Centre, Children’s Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | | | | |
Collapse
|
19
|
Madani S, Amanzadi M, Aghayan HR, Setudeh A, Rezaei N, Rouhifard M, Larijani B. Investigating the safety and efficacy of hematopoietic and mesenchymal stem cell transplantation for treatment of T1DM: a systematic review and meta-analysis. Syst Rev 2022; 11:82. [PMID: 35501872 PMCID: PMC9059401 DOI: 10.1186/s13643-022-01950-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 04/05/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Stem cell transplantation (SCT) has paved the way for treatment of autoimmune diseases. SCT has been investigated in type 1 diabetes mellitus (T1DM) as an autoimmune-based disorder, but previous studies have not presented a comprehensive view of its effect on treatment of T1DM. METHODOLOGY After registration of the present systematic review and meta-analysis in the PROSPERO, a search was done according to the Cochrane guidelines for evaluation of clinical trials to find eligible clinical trials that investigated the effect of SCT on T1DM (based on ADA® diagnostic criteria) from PubMed, Web of science, Scopus, etc, as well as registries of clinical trials from January 1, 2000, to September 31, 2019. A search strategy was designed using MeSH and EM-tree terms. Primary outcome included the changes in the insulin total daily dose (TDD) (U/kg) level, and secondary outcomes included the changes in the HbA1c, c-peptide, and adjusted HbA1c levels. The Q Cochrane test and I2 statistic were performed to assess the heterogeneity and its severity in primary clinical trials. The Cochrane ROB was used to determine risk of bias, and Cochrane Handbook for Systematic Reviews of Interventions was used in the full text papers. The meta-analysis was accomplished in the STATA software, and the results were shown on their forest plots. Confounders were evaluated by the meta-regression test. RESULTS A total of 9452 studies were electronically screened, and 35 papers were included for data extraction. The results of this review study showed that 173 (26.5%) diabetic patients experienced insulin-free period (from 1 to 80 months), and 445 (68%) showed reduction in TDD of insulin after the SCT. Combination of hematopoietic stem cell (HSC) with mesenchymal stem cell (MSC) transplantation were significantly associated with improvement of the TDD (SMD: - 0.586, 95% CI: - 1.204/- 0.509, I2: 0%), HbA1c (SMD: - 0.736, 95% CI: - 1.107/- 0.365, I2: 0%), adjusted HbA1c (SMD: - 2.041, 95% CI: - 2.648/- 1.434, I2: 38.4%), and c-peptide (SMD: 1.917, 95% CI: 0.192/3.641, I2: 92.5%) on month 3 of follow-up, while its association had a growing trend from 3 to 12 months after the transplantation. Considering severe adverse events, HSC transplantation accompanied with conditioning could not be suggested as a safe treatment. CONCLUSION Most of the clinical trials of SCT in T1DM were single arm. Although meta-analysis illustrated the SCT is associated with T1DM improvement, well-designed randomized clinical trials are needed to clarify its efficacy. RECOMMENDATION Based on the results of this meta-analysis, the MSC and its combination with HSC could be considered as "Safe Cell" for SCT in T1DM. Furthermore, to evaluate the SCT efficacy, calculation of insulin TDD (U/kg/day), AUC of c-peptide, and adjusted HbA1c are highly recommended.
Collapse
Affiliation(s)
- Sedigheh Madani
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Islamic Republic of Iran
| | - Mahdiyeh Amanzadi
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Islamic Republic of Iran
| | - Hamid Reza Aghayan
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute Tehran University of Medical Sciences, Tehran, Islamic Republic of Iran
| | - Aria Setudeh
- Children's Medical Center, Tehran University of Medical Sciences, Tehran, Islamic Republic of Iran
| | - Negar Rezaei
- Non-Communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Islamic Republic of Iran
| | - Mahtab Rouhifard
- Non-Communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Islamic Republic of Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute Tehran University of Medical Sciences, Tehran, Islamic Republic of Iran.
| |
Collapse
|
20
|
Ng SM, Chandrasekaran S, Nesbitt V, Grey A, Lim PK, Hui YC, Lek N. Age at diagnosis, anthropometry and birthweight in two ethnically different cohorts of children with type 1 diabetes living in Northwest England or Singapore. Acta Paediatr 2022; 111:1070-1074. [PMID: 35090048 DOI: 10.1111/apa.16274] [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: 11/02/2021] [Revised: 01/21/2022] [Accepted: 01/25/2022] [Indexed: 11/30/2022]
Abstract
AIM To compare children with type 1 diabetes (T1D) living in the Northwest England, United Kingdom (UK) or Singapore, and to correlate age at diagnosis with birthweight and anthropometry at T1D diagnosis. METHODS We included 166 T1D children of white ethnicity in England (UK-White) and 185 T1D children of East-Asian ethnicity origin in Singapore (SG-Asian) who were born between 2002 and 2020. RESULTS The cohorts from UK-White and SG-Asian children differed significantly in FH of T1D (p < 0.001), FH of T2D (p < 0.001) and pubertal status at diagnosis (p = 0.01). Median interquartile range (IQR) for age at diagnosis was similar in the two groups. UK-White children had significantly higher birthweight SDS, height SDS, weight SDS and BMI SDS (all p < 0.001). Among the subgroup of 174 children who were prepubertal and diagnosed after age 5 years, the UK-White children were 11 months older than the SG-Asian children (p = 0.02) indicating that SG-Asian children at the time of T1D diagnosis were more likely to be in puberty compared with UK-White children (30% vs. 18%). CONCLUSION These two cohorts have substantially different genetic and environmental backgrounds, yet age at the diagnosis of T1D was similar except for the prepubertal children who were diagnosed after 5 years old. Timing of puberty and other factors may influence how early T1D presents during childhood.
Collapse
Affiliation(s)
- Sze May Ng
- Department of Paediatrics Southport and Ormskirk Hospital NHS Trust Southport UK
- Department of Women’s and Children’s Health University of Liverpool Southport UK
| | | | - Victoria Nesbitt
- Department of Paediatrics Southport and Ormskirk Hospital NHS Trust Southport UK
| | - Alexandra Grey
- Department of Paediatrics Macclesfield District General Hospital Macclesfield UK
| | - Pei Kwee Lim
- Singapore Department of Paediatrics KK Women’s and Children’s Hospital KK Singapore Singapore
| | - Yuen Ching Hui
- Singapore Department of Paediatrics KK Women’s and Children’s Hospital KK Singapore Singapore
| | - Ngee Lek
- Singapore Department of Paediatrics KK Women’s and Children’s Hospital KK Singapore Singapore
| |
Collapse
|
21
|
Sims EK, Besser RE, Dayan C, Geno Rasmussen C, Greenbaum C, Griffin KJ, Hagopian W, Knip M, Long AE, Martin F, Mathieu C, Rewers M, Steck AK, Wentworth JM, Rich SS, Kordonouri O, Ziegler AG, Herold KC. Screening for Type 1 Diabetes in the General Population: A Status Report and Perspective. Diabetes 2022; 71:610-623. [PMID: 35316839 PMCID: PMC9114719 DOI: 10.2337/dbi20-0054] [Citation(s) in RCA: 92] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 01/05/2022] [Indexed: 01/10/2023]
Abstract
Most screening programs to identify individuals at risk for type 1 diabetes have targeted relatives of people living with the disease to improve yield and feasibility. However, ∼90% of those who develop type 1 diabetes do not have a family history. Recent successes in disease-modifying therapies to impact the course of early-stage disease have ignited the consideration of the need for and feasibility of population screening to identify those at increased risk. Existing population screening programs rely on genetic or autoantibody screening, and these have yielded significant information about disease progression and approaches for timing for screening in clinical practice. At the March 2021 Type 1 Diabetes TrialNet Steering Committee meeting, a session was held in which ongoing efforts for screening in the general population were discussed. This report reviews the background of these efforts and the details of those programs. Additionally, we present hurdles that need to be addressed for successful implementation of population screening and provide initial recommendations for individuals with positive screens so that standardized guidelines for monitoring and follow-up can be established.
Collapse
Affiliation(s)
- Emily K. Sims
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN
| | - Rachel E.J. Besser
- Department of Paediatrics, National Institute for Health Research Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, U.K
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, U.K
| | - Colin Dayan
- Cardiff University School of Medicine, Cardiff, U.K
| | - Cristy Geno Rasmussen
- Barbara Davis Center for Diabetes, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | | | | | | | - 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
| | - Anna E. Long
- Bristol Medical School, University of Bristol, Bristol, U.K
| | | | - Chantal Mathieu
- Department of Endocrinology, UZ Gasthuisberg, KU Leuven, Leuven, Belgium
| | - Marian Rewers
- Barbara Davis Center for Diabetes, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Andrea K. Steck
- Barbara Davis Center for Diabetes, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - John M. Wentworth
- Departments of Diabetes and Endocrinology and Population Health and Immunity, Royal Melbourne Hospital and Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Stephen S. Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA
| | - Olga Kordonouri
- Kinder und Jugendkrankenhaus Auf der Bult, Hannover, Germany
| | - Anette-Gabriele Ziegler
- Institute of Diabetes Research, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany
- School of Medicine, Technical University of Munich, Munich, Germany
| | - Kevan C. Herold
- Department of Immunobiology and Department of Internal Medicine, Yale University, New Haven, CT
| | | |
Collapse
|
22
|
Affiliation(s)
- Mark A Sperling
- From the Department of Pediatrics, Division of Endocrinology and Diabetes, Icahn School of Medicine at Mount Sinai, New York (M.A.S.); and the Pediatric, Adolescent, and Young Adult Section, Joslin Diabetes Center, and Harvard Medical School - both in Boston (L.M.L.)
| | - Lori M Laffel
- From the Department of Pediatrics, Division of Endocrinology and Diabetes, Icahn School of Medicine at Mount Sinai, New York (M.A.S.); and the Pediatric, Adolescent, and Young Adult Section, Joslin Diabetes Center, and Harvard Medical School - both in Boston (L.M.L.)
| |
Collapse
|
23
|
It is time for a moonshot to find “Cures” for diabetic retinal disease. Prog Retin Eye Res 2022; 90:101051. [DOI: 10.1016/j.preteyeres.2022.101051] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 01/19/2022] [Accepted: 01/31/2022] [Indexed: 12/13/2022]
|
24
|
Costenbader KH. Vitamin D and fish oil supplements and risk of autoimmune disease. BMJ 2022; 376:o243. [PMID: 35091393 DOI: 10.1136/bmj.o243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- Karen H Costenbader
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
25
|
Russ HA, Davidson HW. Found in Translation: Novel Insights Into Type 1 Diabetes and β-Cell Biology. Diabetes 2021; 70:2185-2186. [PMID: 34593539 PMCID: PMC8576502 DOI: 10.2337/dbi21-0031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Holger A Russ
- Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Howard W Davidson
- Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
26
|
Kwon BC, Anand V, Severson KA, Ghosh S, Sun Z, Frohnert BI, Lundgren M, Ng K. DPVis: Visual Analytics With Hidden Markov Models for Disease Progression Pathways. IEEE TRANSACTIONS ON VISUALIZATION AND COMPUTER GRAPHICS 2021; 27:3685-3700. [PMID: 32275600 DOI: 10.1109/tvcg.2020.2985689] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Clinical researchers use disease progression models to understand patient status and characterize progression patterns from longitudinal health records. One approach for disease progression modeling is to describe patient status using a small number of states that represent distinctive distributions over a set of observed measures. Hidden Markov models (HMMs) and its variants are a class of models that both discover these states and make inferences of health states for patients. Despite the advantages of using the algorithms for discovering interesting patterns, it still remains challenging for medical experts to interpret model outputs, understand complex modeling parameters, and clinically make sense of the patterns. To tackle these problems, we conducted a design study with clinical scientists, statisticians, and visualization experts, with the goal to investigate disease progression pathways of chronic diseases, namely type 1 diabetes (T1D), Huntington's disease, Parkinson's disease, and chronic obstructive pulmonary disease (COPD). As a result, we introduce DPVis which seamlessly integrates model parameters and outcomes of HMMs into interpretable and interactive visualizations. In this article, we demonstrate that DPVis is successful in evaluating disease progression models, visually summarizing disease states, interactively exploring disease progression patterns, and building, analyzing, and comparing clinically relevant patient subgroups.
Collapse
|
27
|
Abstract
As part of the centennial celebration of insulin's discovery, this review summarizes the current understanding of the genetics, pathogenesis, treatment, and outcomes in type 1 diabetes (T1D). T1D results from an autoimmune response that leads to destruction of the β cells in the pancreatic islet and requires lifelong insulin therapy. While much has been learned about T1D, it is now clear that there is considerable heterogeneity in T1D with regard to genetics, pathology, response to immune-based therapies, clinical course, and susceptibility to diabetes-related complications. This Review highlights knowledge gaps and opportunities to improve the understanding of T1D pathogenesis and outlines emerging therapies to treat or prevent T1D and reduce the burden of T1D.
Collapse
|
28
|
Sims EK, Bundy BN, Stier K, Serti E, Lim N, Long SA, Geyer SM, Moran A, Greenbaum CJ, Evans-Molina C, Herold KC. Teplizumab improves and stabilizes beta cell function in antibody-positive high-risk individuals. Sci Transl Med 2021; 13:eabc8980. [PMID: 33658358 PMCID: PMC8610022 DOI: 10.1126/scitranslmed.abc8980] [Citation(s) in RCA: 191] [Impact Index Per Article: 47.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 02/05/2021] [Indexed: 12/30/2022]
Abstract
We analyzed the effects of a single 14-day course of teplizumab treatment on metabolic function and immune cells among participants in a previously reported randomized controlled trial of nondiabetic relatives at high risk for type 1 diabetes (T1D). In an extended follow-up (923-day median) of a previous report of teplizumab treatment, we found that the median times to diagnosis were 59.6 and 27.1 months for teplizumab- and placebo-treated participants, respectively (HR = 0.457, P = 0.01). Fifty percent of teplizumab-treated but only 22% of the placebo-treated remained diabetes-free. Glucose tolerance, C-peptide area under the curve (AUC), and insulin secretory rates were calculated, and relationships to T cell subsets and function were analyzed. Teplizumab treatment improved beta cell function, reflected by average on-study C-peptide AUC (1.94 versus 1.72 pmol/ml; P = 0.006). Drug treatment reversed a decline in insulin secretion before enrollment, followed by stabilization of the declining C-peptide AUC seen with placebo treatment. Proinsulin:C-peptide ratios after drug treatment were similar between the treatment groups. The changes in C-peptide with teplizumab treatment were associated with increases in partially exhausted memory KLRG1+TIGIT+CD8+ T cells (r = 0.44, P = 0.014) that showed reduced secretion of IFNγ and TNFα. A single course of teplizumab had lasting effects on delay of T1D diagnosis and improved beta cell function in high-risk individuals. Changes in CD8+ T cell subsets indicated that partially exhausted effector cells were associated with clinical response. Thus, this trial showed improvement in metabolic responses and delay of diabetes with immune therapy.
Collapse
Affiliation(s)
- Emily K Sims
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Brian N Bundy
- Department of Epidemiology, and Pediatrics University of South Florida, Tampa, FL 33612, USA
| | - Kenneth Stier
- Departments of Immunobiology and Internal Medicine, Yale University, New Haven, CT 06520, USA
| | | | - Noha Lim
- Immune Tolerance Network, Bethesda, MD 20814, USA
| | - S Alice Long
- Benaroya Research Institute, Seattle WA 98101, USA
| | | | - Antoinette Moran
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
| | | | - Carmella Evans-Molina
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Kevan C Herold
- Departments of Immunobiology and Internal Medicine, Yale University, New Haven, CT 06520, USA.
| |
Collapse
|
29
|
Siller AF, Tosur M, Relan S, Astudillo M, McKay S, Dabelea D, Redondo MJ. Challenges in the diagnosis of diabetes type in pediatrics. Pediatr Diabetes 2020; 21:1064-1073. [PMID: 32562358 DOI: 10.1111/pedi.13070] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 05/07/2020] [Accepted: 06/10/2020] [Indexed: 12/11/2022] Open
Abstract
The incidence of diabetes, both type 1 and type 2, is increasing. Health outcomes in pediatric diabetes are currently poor, with trends indicating that they are worsening. Minority racial/ethnic groups are disproportionately affected by suboptimal glucose control and have a higher risk of acute and chronic complications of diabetes. Correct clinical management starts with timely and accurate classification of diabetes, but in children this is becoming increasingly challenging due to high prevalence of obesity and shifting demographic composition. The growing obesity epidemic complicates classification by obesity's effects on diabetes. Since the prevalence and clinical characteristics of diabetes vary among racial/ethnic groups, migration between countries leads to changes in the distribution of diabetes types in a certain geographical area, challenging the clinician's ability to classify diabetes. These challenges must be addressed to correctly classify diabetes and establish an appropriate treatment strategy early in the course of disease for all. This may be the first step in improving diabetes outcomes across racial/ethnic groups. This review will discuss the pitfalls in the current diabetes classification scheme that is leading to increasing overlap between diabetes types and heterogeneity within each type. It will also present proposed alternative classification schemes and approaches to understanding diabetes type that may improve the timely and accurate classification of pediatric diabetes type.
Collapse
Affiliation(s)
- Alejandro F Siller
- Diabetes and Endocrinology Section, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
| | - Mustafa Tosur
- Diabetes and Endocrinology Section, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
| | - Shilpi Relan
- Diabetes and Endocrinology Section, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
| | - Marcela Astudillo
- Diabetes and Endocrinology Section, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
| | - Siripoom McKay
- Diabetes and Endocrinology Section, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
| | - Dana Dabelea
- Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Maria J Redondo
- Diabetes and Endocrinology Section, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
| |
Collapse
|
30
|
Abstract
PURPOSE OF REVIEW Emerging data have suggested that β-cell dysfunction may exacerbate the development and progression of type 1 diabetes (T1D). In this review, we highlight clinical and preclinical studies suggesting a role for β-cell dysfunction during the evolution of T1D and suggest agents that may promote β-cell health in T1D. RECENT FINDINGS Metabolic abnormalities exist years before development of hyperglycemia and exhibit a reproducible pattern reflecting progressive deterioration of β-cell function and increases in β-cell stress and death. Preclinical studies indicate that T1D may be prevented by modification of pathways impacting intrinsic β-cell stress and antigen presentation. Recent findings suggest that differences in metabolic phenotypes and β-cell stress may reflect differing endotypes of T1D. Multiple pathways representing potential drug targets have been identified, but most remain to be tested in human populations with preclinical disease. SUMMARY This cumulative body of work shows clear evidence that β-cell stress, dysfunction, and death are harbingers of impending T1D and likely contribute to progression of disease and insulin deficiency. Treatment with agents targeting β-cell health could augment interventions with immunomodulatory therapies but will need to be tested in intervention studies with endpoints carefully designed to capture changes in β-cell function and health.
Collapse
Affiliation(s)
- Emily K. Sims
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN
- Department of Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN
- Department of Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN
| | - Raghavendra G. Mirmira
- Kovler Diabetes Center and the Department of Medicine, The University of Chicago, Chicago, IL
| | - Carmella Evans-Molina
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN
- Department of Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN
- Department of Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN
- Roudebush VA Medical Center, Indianapolis, IN
| |
Collapse
|
31
|
Rahman AH, Homann D. Mass cytometry and type 1 diabetes research in the age of single-cell data science. Curr Opin Endocrinol Diabetes Obes 2020; 27:231-239. [PMID: 32618635 PMCID: PMC7596883 DOI: 10.1097/med.0000000000000549] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW New single-cell tec. hnologies developed over the past decade have considerably reshaped the biomedical research landscape, and more recently have found their way into studies probing the pathogenesis of type 1 diabetes (T1D). In this context, the emergence of mass cytometry in 2009 revolutionized immunological research in two fundamental ways that also affect the T1D world: first, its ready embrace by the community and rapid dissemination across academic and private science centers alike established a new standard of analytical complexity for the high-dimensional proteomic stratification of single-cell populations; and second, the somewhat unexpected arrival of mass cytometry awoke the flow cytometry field from its seeming sleeping beauty stupor and precipitated substantial technological advances that by now approach a degree of analytical dimensionality comparable to mass cytometry. RECENT FINDINGS Here, we summarize in detail how mass cytometry has thus far been harnessed for the pursuit of discovery studies in T1D science; we provide a succinct overview of other single-cell analysis platforms that already have been or soon will be integrated into various T1D investigations; and we briefly consider how effective adoption of these technologies requires an adjusted model for expense allocation, prioritization of experimental questions, division of labor, and recognition of scientific contributions. SUMMARY The introduction of contemporary single-cell technologies in general, and of mass cytometry, in particular, provides important new opportunities for current and future T1D research; the necessary reconfiguration of research strategies to accommodate implementation of these technologies, however, may both broaden research endeavors by fostering genuine team science, and constrain their actual practice because of the need for considerable investments into infrastructure and technical expertise.
Collapse
Affiliation(s)
| | - Dirk Homann
- Precision Immunology Institute
- Diabetes, Obesity & Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
32
|
Kakleas K, Basatemur E, Karavanaki K. Association Between Severity of Diabetic Ketoacidosis at Diagnosis and Multiple Autoimmunity in Children With Type 1 Diabetes Mellitus: A Study From a Greek Tertiary Centre. Can J Diabetes 2020; 45:33-38.e2. [PMID: 32800761 DOI: 10.1016/j.jcjd.2020.05.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 05/05/2020] [Accepted: 05/06/2020] [Indexed: 11/29/2022]
Abstract
OBJECTIVES Type 1 diabetes mellitus is a chronic disorder associated with development of autoimmunity. In this work, we studied the relationship between severity of acidosis at diagnosis and future risk for autoimmunity development in children with type 1 diabetes. METHODS We investigated the presence of associated autoimmunity in 144 children with type 1 diabetes (mean ± standard deviation: age, 12.44±4.76 years; diabetes duration, 4.41±3.70 years). We identified the presence of thyroid disease, celiac disease, autoimmune gastritis and adrenal autoimmunity, and retrospectively reviewed the files for presence of diabetic ketoacidosis at diagnosis. RESULTS Autoimmunity prevalence was 16.7% for thyroid autoimmunity, 9.5% for celiac disease, 5% for gastric autoimmunity and 8.0% for multiple autoimmunities. There were strong associations between severe acidosis at diabetes diagnosis (pH<7.10) and development of thyroid autoimmunity (odds ratio [OR], 5.34; 95% confidence interval [CI], 1.90‒15.1; p<0.001), celiac disease (OR, 5.83; 95% CI, 1.19‒28.6; p=0.013), gastric autoimmunity (OR, 13.1; 95% CI, 1.22‒140; p=0.006) and multiple autoimmunity (OR, 26.7; 95% CI, 2.36‒301; p<0.01). The associations persisted after adjustment for sex, age at diabetes diagnosis, age at assessment, time since diabetes diagnosis and antiglutamic acid decarboxylase autoantibody status. CONCLUSIONS The severity of acidosis at diagnosis is strongly associated with the development of associated autoimmune diseases in children with type 1 diabetes and could act as a predictive factor for multiple autoimmunity development. This association can be either due to effect of acidosis on immune system or to the presence of a more aggressive diabetes endotype.
Collapse
Affiliation(s)
- Kostas Kakleas
- Paediatric Department, Leicester Royal Infirmary, Leicester, United Kingdom.
| | - Emre Basatemur
- Population, Policy and Practice Programme, Institute of Child Health, University College of London, London, United Kingdom
| | - Kyriaki Karavanaki
- Diabetic Clinic, Second Department of Pediatrics, University of Athens, "P&A Kyriakou" Children's Hospital, Athens, Greece
| |
Collapse
|
33
|
Wang Y, Chen J, Li S, Zhang X, Guo Z, Hu J, Shao X, Song N, Zhao Y, Li H, Yang G, Xu C, Wei C. Exogenous spermine attenuates rat diabetic cardiomyopathy via suppressing ROS-p53 mediated downregulation of calcium-sensitive receptor. Redox Biol 2020; 32:101514. [PMID: 32234613 PMCID: PMC7113441 DOI: 10.1016/j.redox.2020.101514] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 02/23/2020] [Accepted: 03/18/2020] [Indexed: 12/13/2022] Open
Abstract
Diabetic cardiomyopathy (DCM) is a severe complication of type 1 diabetic (T1D) patients, manifested as combined diastolic and systolic dysfunction. DCM is associated with impaired calcium homeostasis secondary to decreased calcium-sensitive receptor (CaSR) expression. Spermine, a direct agonist of CaSR, was found deficient in cardiomyocytes of T1D rats. However, the role of spermine in DCM was unclear. Here, we examined the cardioprotective effect of exogenous spermine on DCM in streptozotocin (STZ) induced-T1D rats and high-glucose (HG)-incubated neonatal rat cardiomyocytes. Exogenous spermine significantly attenuated cardiac dysfunction in T1D rats, characterized by improved echocardiography, less fibrosis, reduced myocardial endoplasmic reticulum (ER) stress and oxidative stress, and increased expression of myocardial membrane CaSR. In cultured neonatal rat cardiomyocytes, exogenous spermine attenuated myocardial injury induced by HG treatment, demonstrated by restored cellular glucose uptake capacity, reduced expression of apoptotic markers, lowered level of oxidative stress, ER stress and unfolded protein response, and upregulated cell membrane CaSR. Mechanistically, the cardioprotective effect of spermine appeared dependent upon effective elimination of reactive oxygen species (ROS) and up-regulation of CaSR expression by suppressing the Nrf2-ROS-p53-MuRF1 axis. Taken together, these results suggest that exogenous spermine protects against DCM in vivo and in vitro, partially via suppressing ROS and p53-mediated downregulation of cell membrane CaSR.
Collapse
Affiliation(s)
- Yuehong Wang
- Department of Pathophysiology, Harbin Medical University, Harbin, 150081, China
| | - Junting Chen
- Department of Anesthesiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, 150000, China
| | - Siwei Li
- Department of Pathophysiology, Harbin Medical University, Harbin, 150081, China
| | - Xinying Zhang
- Department of Pathophysiology, Harbin Medical University, Harbin, 150081, China
| | - Zuoming Guo
- Department of Hepatobiliary and Pancreatic Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Jing Hu
- Department of Pathophysiology, Harbin Medical University, Harbin, 150081, China
| | - Xiaoting Shao
- Department of Pathophysiology, Harbin Medical University, Harbin, 150081, China
| | - Ningyang Song
- Department of Pathophysiology, Harbin Medical University, Harbin, 150081, China
| | - Yajun Zhao
- Department of Pathophysiology, Harbin Medical University, Harbin, 150081, China
| | - Hongzhu Li
- Department of Pathophysiology, Harbin Medical University, Harbin, 150081, China
| | - Guangdong Yang
- Department of Chemistry and Biochemistry, Laurentian University, Sudbury, P3E 2C6, Canada
| | - Changqing Xu
- Department of Pathophysiology, Harbin Medical University, Harbin, 150081, China
| | - Can Wei
- Department of Pathophysiology, Harbin Medical University, Harbin, 150081, China.
| |
Collapse
|
34
|
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
|
35
|
Harmonization of immunoassays for biomarkers in diabetes mellitus. Biotechnol Adv 2020; 39:107359. [DOI: 10.1016/j.biotechadv.2019.02.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 02/07/2019] [Accepted: 02/21/2019] [Indexed: 12/13/2022]
|
36
|
Simeonovic CJ, Popp SK, Brown DJ, Li FJ, Lafferty ARA, Freeman C, Parish CR. Heparanase and Type 1 Diabetes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1221:607-630. [PMID: 32274728 DOI: 10.1007/978-3-030-34521-1_24] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Type 1 diabetes (T1D) results from autoimmune destruction of insulin-producing beta cells in pancreatic islets. The degradation of the glycosaminoglycan heparan sulfate (HS) by the endo-β-D-glycosidase heparanase plays a critical role in multiple stages of the disease process. Heparanase aids (i) migration of inflammatory leukocytes from the vasculature to the islets, (ii) intra-islet invasion by insulitis leukocytes, and (iii) selective destruction of beta cells. These disease stages are marked by the solubilization of HS in the subendothelial basement membrane (BM), HS breakdown in the peri-islet BM, and the degradation of HS inside beta cells, respectively. Significantly, healthy islet beta cells are enriched in highly sulfated HS which is essential for their viability, protection from damage by reactive oxygen species (ROS), beta cell function and differentiation. Consequently, mouse and human beta cells but not glucagon-producing alpha cells (which contain less-sulfated HS) are exquisitely vulnerable to heparanase-mediated damage. In vitro, the death of HS-depleted mouse and human beta cells can be prevented by HS replacement using highly sulfated HS mimetics or analogues. T1D progression in NOD mice and recent-onset T1D in humans correlate with increased expression of heparanase by circulating leukocytes of myeloid origin and heparanase-expressing insulitis leukocytes. Treatment of NOD mice with the heparanase inhibitor and HS replacer, PI-88, significantly reduced T1D incidence by 50%, impaired the development of insulitis and preserved beta cell HS. These outcomes identified heparanase as a novel destructive tool in T1D, distinct from the conventional cytotoxic and apoptosis-inducing mechanisms of autoreactive T cells. In contrast to exogenous catalytically active heparanase, endogenous heparanase may function in HS homeostasis, gene expression and insulin secretion in normal beta cells and immune gene expression in leukocytes. In established diabetes, the interplay between hyperglycemia, local inflammatory cells (e.g. macrophages) and heparanase contributes to secondary micro- and macro-vascular disease. We have identified dual activity heparanase inhibitors/HS replacers as a novel class of therapeutic for preventing T1D progression and potentially for mitigating secondary vascular disease that develops with long-term T1D.
Collapse
Affiliation(s)
- Charmaine J Simeonovic
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia.
| | - Sarah K Popp
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Debra J Brown
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Fei-Ju Li
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Antony R A Lafferty
- Department of Paediatrics, The Canberra Hospital, Woden, ACT, Australia.,The ANU Medical School, The Australian National University, Canberra, ACT, Australia
| | - Craig Freeman
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Christopher R Parish
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| |
Collapse
|
37
|
Battaglia M, Ahmed S, Anderson MS, Atkinson MA, Becker D, Bingley PJ, Bosi E, Brusko TM, DiMeglio LA, Evans-Molina C, Gitelman SE, Greenbaum CJ, Gottlieb PA, Herold KC, Hessner MJ, Knip M, Jacobsen L, Krischer JP, Long SA, Lundgren M, McKinney EF, Morgan NG, Oram RA, Pastinen T, Peters MC, Petrelli A, Qian X, Redondo MJ, Roep BO, Schatz D, Skibinski D, Peakman M. Introducing the Endotype Concept to Address the Challenge of Disease Heterogeneity in Type 1 Diabetes. Diabetes Care 2020; 43:5-12. [PMID: 31753960 PMCID: PMC6925574 DOI: 10.2337/dc19-0880] [Citation(s) in RCA: 230] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 10/14/2019] [Indexed: 02/06/2023]
Abstract
The clinical diagnosis of new-onset type 1 diabetes has, for many years, been considered relatively straightforward. Recently, however, there is increasing awareness that within this single clinical phenotype exists considerable heterogeneity: disease onset spans the complete age range; genetic susceptibility is complex; rates of progression differ markedly, as does insulin secretory capacity; and complication rates, glycemic control, and therapeutic intervention efficacy vary widely. Mechanistic and immunopathological studies typically show considerable patchiness across subjects, undermining conclusions regarding disease pathways. Without better understanding, type 1 diabetes heterogeneity represents a major barrier both to deciphering pathogenesis and to the translational effort of designing, conducting, and interpreting clinical trials of disease-modifying agents. This realization comes during a period of unprecedented change in clinical medicine, with increasing emphasis on greater individualization and precision. For complex disorders such as type 1 diabetes, the option of maintaining the "single disease" approach appears untenable, as does the notion of individualizing each single patient's care, obliging us to conceptualize type 1 diabetes less in terms of phenotypes (observable characteristics) and more in terms of disease endotypes (underlying biological mechanisms). Here, we provide our view on an approach to dissect heterogeneity in type 1 diabetes. Using lessons from other diseases and the data gathered to date, we aim to delineate a roadmap through which the field can incorporate the endotype concept into laboratory and clinical practice. We predict that such an effort will accelerate the implementation of precision medicine and has the potential for impact on our approach to translational research, trial design, and clinical management.
Collapse
Affiliation(s)
- Manuela Battaglia
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Hospital, Milan, Italy
| | | | - Mark S Anderson
- Diabetes Center, University of California, San Francisco, San Francisco, CA
| | - Mark A Atkinson
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL
| | - Dorothy Becker
- Division of Endocrinology and Diabetes, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA
| | - Polly J Bingley
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, U.K
| | - Emanuele Bosi
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Hospital, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy, and Department of Internal Medicine, IRCCS San Raffaele Hospital, Milan, Italy
| | - Todd M Brusko
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL
| | - Linda A DiMeglio
- Division of Pediatric Endocrinology and Diabetology and Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN
| | - Carmella Evans-Molina
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN
| | - Stephen E Gitelman
- Division of Pediatric Endocrinology and Diabetes, University of California, San Francisco, San Francisco, CA
| | | | - Peter A Gottlieb
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, Aurora, CO
| | - Kevan C Herold
- Department of Immunobiology, Yale University, New Haven, CT
| | - Martin J Hessner
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI
| | - Mikael Knip
- Children's Hospital, University of Helsinki and Helsinki University Hospital, Clinical and Molecular Metabolism Research Program, University of Helsinki, Helsinki, Finland
| | - Laura Jacobsen
- Department of Pediatrics, University of Florida, Gainesville, FL
| | - Jeffrey P Krischer
- Health Informatics Institute, Morsani College of Medicine, University of South Florida, Tampa, FL
| | - S Alice Long
- Diabetes Program, Benaroya Research Institute, Seattle, WA
| | - Markus Lundgren
- Department of Clinical Sciences, Clinical Research Centre, Faculty of Medicine, Lund University, and Skåne University Hospital, Malmö, Sweden
| | - Eoin F McKinney
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's Hospital, Cambridge, U.K
| | - Noel G Morgan
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, U.K.,University of Exeter Medical School and Royal Devon and Exeter Hospital, Exeter, U.K
| | - Richard A Oram
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Royal Devon and Exeter Hospital, Exeter, U.K.,NIHR Exeter Clinical Research Facility, University of Exeter Medical School, Exeter, U.K.,Academic Renal Unit, Royal Devon and Exeter NHS Foundation Trust, Exeter, U.K
| | - Tomi Pastinen
- Center for Pediatric Genomic Medicine, Children's Mercy Kansas City, Kansas City, MO
| | - Michael C Peters
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, and Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA
| | - Alessandra Petrelli
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Hospital, Milan, Italy
| | - Xiaoning Qian
- Department of Electrical and Computer Engineering, TEES-AgriLife Center for Bioinformatics and Genomic Systems Engineering, Texas A&M University, College Station, TX
| | - Maria J Redondo
- Baylor College of Medicine, Texas Children's Hospital, Houston, TX
| | - Bart O Roep
- Department of Diabetes Immunology, Diabetes & Metabolism Research Institute, Beckman Research Institute, National Medical Center, City of Hope, Duarte, CA.,Department of Immunohaematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
| | - Desmond Schatz
- Department of Pediatrics, University of Florida, Gainesville, FL
| | | | - Mark Peakman
- Peter Gorer Department of Immunobiology, Faculty of Life Sciences and Medicine, King's College London, London, U.K. .,King's Health Partners Institute of Diabetes, Obesity and Endocrinology, London, U.K
| |
Collapse
|
38
|
Acevedo-Calado MJ, Pietropaolo SL, Morran MP, Schnell S, Vonberg AD, Verge CF, Gianani R, Becker DJ, Huang S, Greenbaum CJ, Yu L, Davidson HW, Michels AW, Rich SS, Pietropaolo M. Autoantibodies Directed Toward a Novel IA-2 Variant Protein Enhance Prediction of Type 1 Diabetes. Diabetes 2019; 68:1819-1829. [PMID: 31167877 PMCID: PMC6702638 DOI: 10.2337/db18-1351] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Accepted: 05/30/2019] [Indexed: 02/06/2023]
Abstract
We identified autoantibodies (AAb) reacting with a variant IA-2 molecule (IA-2var) that has three amino acid substitutions (Cys27, Gly608, and Pro671) within the full-length molecule. We examined IA-2var AAb in first-degree relatives of type 1 diabetes (T1D) probands from the TrialNet Pathway to Prevention Study. The presence of IA-2var-specific AAb in relatives was associated with accelerated progression to T1D in those positive for AAb to GAD65 and/or insulin but negative in the standard test for IA-2 AAb. Furthermore, relatives with single islet AAb (by traditional assays) and carrying both IA-2var AAb and the high-risk HLA-DRB1*04-DQB1*03:02 haplotype progress rapidly to onset of T1D. Molecular modeling of IA-2var predicts that the genomic variation that alters the three amino acids induces changes in the three-dimensional structure of the molecule, which may lead to epitope unmasking in the IA-2 extracellular domain. Our observations suggest that the presence of AAb to IA-2var would identify high-risk subjects who would benefit from participation in prevention trials who have one islet antibody by traditional testing and otherwise would be misclassified as "low risk" relatives.
Collapse
Affiliation(s)
- Maria J. Acevedo-Calado
- Diabetes Research Center, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX
| | - Susan L. Pietropaolo
- Diabetes Research Center, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX
| | - Michael P. Morran
- Department of Surgery, College of Medicine, University of Toledo, Toledo, OH
| | - Santiago Schnell
- Department of Molecular & Integrative Physiology and Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI
| | - Andrew D. Vonberg
- Diabetes Research Center, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX
| | - Charles F. Verge
- School of Women’s and Children’s Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Roberto Gianani
- Diabetes Research Center, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX
| | - Dorothy J. Becker
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Shuai Huang
- Department of Industrial & Systems Engineering, University of Washington, Seattle, WA
| | | | - Liping Yu
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, CO
| | - Howard W. Davidson
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, CO
| | - Aaron W. Michels
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, CO
| | - Stephen S. Rich
- Center for Public Health Genomics, Department of Public Health Sciences, University of Virginia, Charlottesville, VA
| | - Massimo Pietropaolo
- Diabetes Research Center, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX
| |
Collapse
|
39
|
Vecchio F, Messina G, Giovenzana A, Petrelli A. New Evidence of Exocrine Pancreatopathy in Pre-symptomatic and Symptomatic Type 1 Diabetes. Curr Diab Rep 2019; 19:92. [PMID: 31471779 DOI: 10.1007/s11892-019-1223-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW Type 1 diabetes (T1D) is one of the most frequent chronic autoimmune diseases in humans, characterized by the lack of insulin production resulting in high blood glucose levels and lifelong requirement of exogenous insulin administration for survival. It is now recognized that the autoimmune process begins years before the clinical onset, in a stage called pre-symptomatic T1D, in which the presence of β-cell-specific autoantibodies is detectable. Our aim is to review evidence for T1D as a "whole-pancreas disease," featured by both endocrine and exocrine pancreas alterations already at early disease stages. RECENT FINDINGS In this review, we discuss a series of recent observations indicating that in genetically predisposed individuals, structural and functional abnormalities as well as immune cell infiltration of the exocrine pancreas are already present in the pre-symptomatic stages of the disease. Despite T1D being considered a β-cell-specific disease, numerous reports point to the presence of exocrine pancreas subclinical abnormalities occurring during disease development. These observations challenge the long-standing idea that T1D exocrine damage exists as a mere consequence of disease progression and provide further explanation of mechanisms underlying T1D pathogenesis.
Collapse
Affiliation(s)
- Federica Vecchio
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | | | - Anna Giovenzana
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Alessandra Petrelli
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy.
| |
Collapse
|
40
|
Peters L, Posgai A, Brusko TM. Islet-immune interactions in type 1 diabetes: the nexus of beta cell destruction. Clin Exp Immunol 2019; 198:326-340. [PMID: 31309537 DOI: 10.1111/cei.13349] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/17/2019] [Indexed: 12/12/2022] Open
Abstract
Recent studies in Type 1 Diabetes (T1D) support an emerging model of disease pathogenesis that involves intrinsic β-cell fragility combined with defects in both innate and adaptive immune cell regulation. This combination of defects induces systematic changes leading to organ-level atrophy and dysfunction of both the endocrine and exocrine portions of the pancreas, ultimately culminating in insulin deficiency and β-cell destruction. In this review, we discuss the animal model data and human tissue studies that have informed our current understanding of the cross-talk that occurs between β-cells, the resident stroma, and immune cells that potentiate T1D. Specifically, we will review the cellular and molecular signatures emerging from studies on tissues derived from organ procurement programs, focusing on in situ defects occurring within the T1D islet microenvironment, many of which are not yet detectable by standard peripheral blood biomarkers. In addition to improved access to organ donor tissues, various methodological advances, including immune receptor repertoire sequencing and single-cell molecular profiling, are poised to improve our understanding of antigen-specific autoimmunity during disease development. Collectively, the knowledge gains from these studies at the islet-immune interface are enhancing our understanding of T1D heterogeneity, likely to be an essential component for instructing future efforts to develop targeted interventions to restore immune tolerance and preserve β-cell mass and function.
Collapse
Affiliation(s)
- L Peters
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL, USA
| | - A Posgai
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL, USA
| | - T M Brusko
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL, USA
| |
Collapse
|
41
|
Battaglia M, Petrelli A, Vecchio F. Neutrophils and type 1 diabetes: current knowledge and suggested future directions. Curr Opin Endocrinol Diabetes Obes 2019; 26:201-206. [PMID: 31157631 DOI: 10.1097/med.0000000000000485] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE OF REVIEW Purpose of this review is to describe the most recent human studies on neutrophils in type 1 diabetes (T1D) and to focus on the key questions that still need to be addressed. RECENT FINDINGS Recent evidences demonstrate that neutrophils have marked abnormalities in phenotype and function and play a central role in initiation and perpetuation of aberrant immune responses and organ damage in various systemic autoimmune diseases such as lupus erythematosus and rheumatoid arthritis. In T1D, we have recently demonstrated that reduced circulating neutrophil numbers precede and accompany the disease and that neutrophils infiltrate the pancreas and extrude neutrophil extracellular traps already before the onset of clinical symptoms. However, few other evidences of alterations in neutrophil phenotype and function have been reported in humans, especially in the T1D presymptomatic phases. SUMMARY Dissecting the pathogenic role of these cells in human T1D is crucial for a better understanding of the disease and to open new therapeutic opportunities.
Collapse
Affiliation(s)
- Manuela Battaglia
- San Raffaele Diabetes Research Institute, IRCCS Ospedale, San Raffaele, Milan, Italy
| | | | | |
Collapse
|
42
|
Vaitaitis GM, Rihanek M, Alkanani AK, Waid DM, Gottlieb PA, Wagner DH. Biomarker discovery in pre-Type 1 Diabetes; Th40 cells as a predictive risk factor. J Clin Endocrinol Metab 2019; 104:4127-4142. [PMID: 31063181 PMCID: PMC6685715 DOI: 10.1210/jc.2019-00364] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 05/01/2019] [Indexed: 01/31/2023]
Abstract
CONTEXT The incidence of Type 1 Diabetes (T1D) is increasing worldwide. The quest to understand T1D etiology as well as how to predict diabetes is ongoing and, in many ways, those goals intertwine. While genetic components associate with T1D, not all T1D individuals have those components and not all subjects with those components develop disease. OBJECTIVE More robust methods for prediction of T1D are needed. Can high CD4+CD40+ T cell (Th40) levels be used as a biomarker in addition to other markers? METHODS Th40 levels were assessed along with other parameters in blood collected from prediabetic TrialNet subjects. RESULTS Pre-diabetic subjects, stratified according to their Th40 cell levels, demonstrate patterns that parallel those seen between control and T1D subjects. Cytokine patterns are significantly different between Th40-high and -low subjects and a CD4/CD8 double-positive population is more represented in Th40-high groups. Subjects experiencing impaired glucose tolerance present a significantly higher Th40 level than control subjects do. HLA DR4/DR4 and DQ8/DQ8, HLAs associated with T1D, are more likely found among Th40-high subjects. Interestingly, HLA DR4/DR4 subjects were significantly older compared with all other subjects, suggesting that this haplotype together with a high Th40 level may represent someone who will onset after age 30, which is reported for 42% of T1D cases. CONCLUSION Considering the differences found in relation to prediabetic Th40 cell level, it may be possible to devise methods that more accurately predicts who will proceed toward diabetes and, possibly, at what stage of prediabetes a subject is.
Collapse
Affiliation(s)
- Gisela M Vaitaitis
- Webb-Waring Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Marynette Rihanek
- Barbara Davis Center for Childhood Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Aimon K Alkanani
- Barbara Davis Center for Childhood Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Dan M Waid
- Webb-Waring Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Peter A Gottlieb
- Barbara Davis Center for Childhood Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - David H Wagner
- Webb-Waring Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- Correspondence and Reprint Requests: David H. Wagner, Jr., PhD, University of Colorado Anschutz Medical Campus, 12850 East Montview Boulevard, Aurora, Colorado 80045. E-mail:
| | | |
Collapse
|
43
|
Thompson PJ, Shah A, Ntranos V, Van Gool F, Atkinson M, Bhushan A. Targeted Elimination of Senescent Beta Cells Prevents Type 1 Diabetes. Cell Metab 2019; 29:1045-1060.e10. [PMID: 30799288 DOI: 10.1016/j.cmet.2019.01.021] [Citation(s) in RCA: 230] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Revised: 07/08/2018] [Accepted: 01/23/2019] [Indexed: 12/25/2022]
Abstract
Type 1 diabetes (T1D) is an organ-specific autoimmune disease characterized by hyperglycemia due to progressive loss of pancreatic beta cells. Immune-mediated beta cell destruction drives the disease, but whether beta cells actively participate in the pathogenesis remains unclear. Here, we show that during the natural history of T1D in humans and the non-obese diabetic (NOD) mouse model, a subset of beta cells acquires a senescence-associated secretory phenotype (SASP). Senescent beta cells upregulated pro-survival mediator Bcl-2, and treatment of NOD mice with Bcl-2 inhibitors selectively eliminated these cells without altering the abundance of the immune cell types involved in the disease. Significantly, elimination of senescent beta cells halted immune-mediated beta cell destruction and was sufficient to prevent diabetes. Our findings demonstrate that beta cell senescence is a significant component of the pathogenesis of T1D and indicate that clearance of senescent beta cells could be a new therapeutic approach for T1D.
Collapse
Affiliation(s)
- Peter J Thompson
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Ajit Shah
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Vasilis Ntranos
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Electrical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Frederic Van Gool
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Mark Atkinson
- Diabetes Institute, University of Florida, Gainesville, FL 32610-0296, USA
| | - Anil Bhushan
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
44
|
Dufort MJ, Greenbaum CJ, Speake C, Linsley PS. Cell type-specific immune phenotypes predict loss of insulin secretion in new-onset type 1 diabetes. JCI Insight 2019; 4:125556. [PMID: 30830868 DOI: 10.1172/jci.insight.125556] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 01/17/2019] [Indexed: 12/12/2022] Open
Abstract
The rate of decline in insulin secretion after diagnosis with type 1 diabetes (T1D) varies substantially among individuals and with age at diagnosis, but the mechanism(s) behind this heterogeneity are not well understood. We investigated the loss of pancreatic β cell function in new-onset T1D subjects using unbiased whole blood RNA-seq and verified key findings by targeted cell count measurements. We found that patients who lost insulin secretion more rapidly had immune phenotypes ("immunotypes") characterized by higher levels of B cells and lower levels of neutrophils, especially neutrophils expressing primary granule genes. The B cell and neutrophil immunotypes showed strong age dependence, with B cell levels in particular predicting rate of progression in young subjects only. This age relationship suggested that therapy targeting B cells in T1D would be most effective in young subjects with high pretreatment B cell levels, a prediction which was supported by data from a clinical trial of rituximab in new-onset subjects. These findings demonstrate a link between age-related immunotypes and disease outcome in new-onset T1D. Furthermore, our data suggest that greater success could be achieved by targeted use of immunomodulatory therapy in specific T1D populations defined by age and immune characteristics.
Collapse
Affiliation(s)
| | - Carla J Greenbaum
- Diabetes Clinical Research Program, Benaroya Research Institute at Virginia Mason, Seattle, Washington, USA
| | - Cate Speake
- Diabetes Clinical Research Program, Benaroya Research Institute at Virginia Mason, Seattle, Washington, USA
| | | |
Collapse
|
45
|
Campbell-Thompson ML, Filipp SL, Grajo JR, Nambam B, Beegle R, Middlebrooks EH, Gurka MJ, Atkinson MA, Schatz DA, Haller MJ. Relative Pancreas Volume Is Reduced in First-Degree Relatives of Patients With Type 1 Diabetes. Diabetes Care 2019; 42:281-287. [PMID: 30552130 PMCID: PMC6341284 DOI: 10.2337/dc18-1512] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 10/31/2018] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Pancreas size is reduced in patients at type 1 diabetes onset and in autoantibody (AAB)-positive donors without diabetes. We sought to determine whether pancreas volume (PV) imaging could improve understanding of the loss of pancreas size in first-degree relatives (FDRs) of patients with type 1 diabetes. We also examined relationships among PV, AAB status, and endocrine and exocrine functions. RESEARCH DESIGN AND METHODS We conducted a cross-sectional study that included five groups: AAB- control subjects (no diabetes and no first- or second-degree relatives with type 1 diabetes) (N = 49), AAB- FDRs (N = 61), AAB+ FDRs (N = 67 total: n = 31 with a single positive AAB [AAB+ single] and n = 36 with multiple positive AABs [AAB+ multiple]), and patients with recent-onset type 1 diabetes (<1 year) (N = 52). Fasting subjects underwent 1.5T pancreatic MRI, and PV and relative PV (RPV) (PV-to-BMI ratio) were analyzed between groups and for correlations with HbA1c, C-peptide, glucose, and trypsinogen. RESULTS All FDR groups had significantly lower RPV adjusted for BMI (RPVBMI) than control subjects (all P < 0.05). Patients with type 1 diabetes had lower RPVBMI than AAB- FDR (P < 0.0001) and AAB+ multiple (P ≤ 0.013) subjects. Transformed data indicated that trypsinogen levels were lowest in patients with type 1 diabetes. CONCLUSIONS This study demonstrates, for the first time, all FDRs having significantly smaller RPVBMI compared with AAB- control subjects. Furthermore, RPVBMI was significantly lower in patients with recent-onset type 1 diabetes than in the AAB- FDR and AAB+ multiple groups. As such, RPVBMI may be a novel noninvasive biomarker for predicting progression through stages of type 1 diabetes risk. This study highlights the potential paracrine relationships between the exocrine and endocrine pancreas in progression to type 1 diabetes in subjects at risk.
Collapse
Affiliation(s)
- Martha L Campbell-Thompson
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL
| | - Stephanie L Filipp
- Health Outcomes and Biomedical Informatics, Institute for Child Health Policy, College of Medicine, University of Florida, Gainesville, FL
| | - Joseph R Grajo
- Department of Radiology, College of Medicine, University of Florida, Gainesville, FL
| | - Bimota Nambam
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL
| | - Richard Beegle
- Department of Radiology, College of Medicine, University of Florida, Gainesville, FL
| | - Erik H Middlebrooks
- Department of Radiology, College of Medicine, University of Florida, Gainesville, FL
| | - Matthew J Gurka
- Health Outcomes and Biomedical Informatics, Institute for Child Health Policy, College of Medicine, University of Florida, Gainesville, FL
| | - Mark A Atkinson
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL.,Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL
| | - Desmond A Schatz
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL
| | - Michael J Haller
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL
| |
Collapse
|
46
|
Marré ML, Piganelli JD, James EA. Protecting functional β cells with a therapeutic peptide. ANNALS OF TRANSLATIONAL MEDICINE 2018; 6:372. [PMID: 30370299 DOI: 10.21037/atm.2018.07.26] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Meghan L Marré
- Division of Pediatric Surgery, Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jon D Piganelli
- Division of Pediatric Surgery, Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Eddie A James
- Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| |
Collapse
|
47
|
Harms RZ, Lorenzo-Arteaga KM, Ostlund KR, Smith VB, Smith LM, Gottlieb P, Sarvetnick N. Abnormal T Cell Frequencies, Including Cytomegalovirus-Associated Expansions, Distinguish Seroconverted Subjects at Risk for Type 1 Diabetes. Front Immunol 2018; 9:2332. [PMID: 30405601 PMCID: PMC6204396 DOI: 10.3389/fimmu.2018.02332] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 09/19/2018] [Indexed: 12/12/2022] Open
Abstract
We analyzed T cell subsets from cryopreserved PBMC obtained from the TrialNet Pathway to Prevention archives. We compared subjects who had previously seroconverted for one or more autoantibodies with non-seroconverted, autoantibody negative individuals. We observed a reduced frequency of MAIT cells among seroconverted subjects. Seroconverted subjects also possessed decreased frequencies of CCR4-expressing CD4 T cells, including a regulatory-like subset. Interestingly, we found an elevation of CD57+, CD28–, CD127–, CD27– CD8 T cells (SLEC) among seroconverted subjects that was most pronounced among those that progressed to disease. The frequency of these SLEC was strongly correlated with CMV IgG abundance among seroconverted subjects, associated with IA-2 levels, and most elevated among CMV+ seroconverted subjects who progressed to disease. Combined, our data indicate discrete, yet profound T cell alterations are associated with islet autoimmunity among at-risk subjects.
Collapse
Affiliation(s)
- Robert Z Harms
- Surgery-Transplant, University of Nebraska Medical Center, Omaha, NE, United States
| | | | - Katie R Ostlund
- Surgery-Transplant, University of Nebraska Medical Center, Omaha, NE, United States
| | - Victoria B Smith
- Office of the Vice Chancellor of Research, University of Nebraska Medical Center, Omaha, NE, United States
| | - Lynette M Smith
- Biostatistics, University of Nebraska Medical Center, Omaha, NE, United States
| | - Peter Gottlieb
- Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Nora Sarvetnick
- Surgery-Transplant, University of Nebraska Medical Center, Omaha, NE, United States.,Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
48
|
Couper JJ, Haller MJ, Greenbaum CJ, Ziegler AG, Wherrett DK, Knip M, Craig ME. ISPAD Clinical Practice Consensus Guidelines 2018: Stages of type 1 diabetes in children and adolescents. Pediatr Diabetes 2018; 19 Suppl 27:20-27. [PMID: 30051639 DOI: 10.1111/pedi.12734] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 07/16/2018] [Indexed: 12/15/2022] Open
Affiliation(s)
- Jennifer J Couper
- Department of Diabetes and Endocrinology, Womens and Childrens Hospital, North Adelaide, Australia.,Robinson Research Institute, University of Adelaide, Adelaide, Australia
| | - Michael J Haller
- Department of Pediatrics, Division of Endocrinology, University of Florida, Gainesville, Florida
| | | | - Anette-Gabriele Ziegler
- Institute of Diabetes Research, Helmholtz Zentrum München, and Forschergruppe Diabetes, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Diane K Wherrett
- Division of Endocrinology, Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, Canada
| | - Mikael Knip
- Children's Hospital, University of Helsinki, Helsinki, Finland
| | - Maria E Craig
- Department of Diabetes and Endocrinology, The Children's Hospital at Westmead, Sydney, Australia.,Discipline of Pediatrics and Child Health, University of Sydney, Sydney, Australia.,School of Women's and Children's Health, University of New South Wales, Sydney, Australia
| |
Collapse
|
49
|
Leete P, Mallone R, Richardson SJ, Sosenko JM, Redondo MJ, Evans-Molina C. The Effect of Age on the Progression and Severity of Type 1 Diabetes: Potential Effects on Disease Mechanisms. Curr Diab Rep 2018; 18:115. [PMID: 30259209 PMCID: PMC10043737 DOI: 10.1007/s11892-018-1083-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
PURPOSE OF REVIEW To explore the impact of age on type 1 diabetes (T1D) pathogenesis. RECENT FINDINGS Children progress more rapidly from autoantibody positivity to T1D and have lower C-peptide levels compared to adults. In histological analysis of post-mortem pancreata, younger age of diagnosis is associated with reduced numbers of insulin containing islets and a hyper-immune CD20hi infiltrate. Moreover compared to adults, children exhibit decreased immune regulatory function and increased engagement and trafficking of autoreactive CD8+ T cells, and age-related differences in β cell vulnerability may also contribute to the more aggressive immune phenotype observed in children. To account for some of these differences, HLA and non-HLA genetic loci that influence multiple disease characteristics, including age of onset, are being increasingly characterized. The exception of T1D as an autoimmune disease more prevalent in children than adults results from a combination of immune, metabolic, and genetic factors. Age-related differences in T1D pathology have important implications for better tailoring of immunotherapies.
Collapse
Affiliation(s)
- Pia Leete
- Islet Biology Exeter (IBEx), Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Exeter, UK
| | - Roberto Mallone
- INSERM U1016, CNRS UMR8104, Cochin Institute, Sorbonne Paris Cité; Assistance Publique Hôpitaux de Paris, Service de Diabétologie, Cochin Hospital, INSERM and Assistance Publique Hôpitaux de Paris, Paris, France
| | - Sarah J Richardson
- Islet Biology Exeter (IBEx), Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Exeter, UK
| | - Jay M Sosenko
- Department of Medicine and the Diabetes Research Institute, Leonard Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Maria J Redondo
- Department of Pediatrics, Baylor College of Medicine and the Texas Children's Hospital, Houston, TX, USA
| | - Carmella Evans-Molina
- Departments of Medicine and Pediatrics and the Herman B Wells Center for Pediatric Research, Indiana University School of Medicine and the Roudebush VA Medical Center, 635 Barnhill Drive, MS 2031A, Indianapolis, IN, 46202, USA.
| |
Collapse
|
50
|
Abstract
PURPOSE OF REVIEW The immunosuppressive agent cyclosporine was first reported to lower daily insulin dose and improve glycemic control in patients with new-onset type 1 diabetes (T1D) in 1984. While renal toxicity limited cyclosporine's extended use, this observation ignited collaborative efforts to identify immunotherapeutic agents capable of safely preserving β cells in patients with or at risk for T1D. RECENT FINDINGS Advances in T1D prediction and early diagnosis, together with expanded knowledge of the disease mechanisms, have facilitated trials targeting specific immune cell subsets, autoantigens, and pathways. In addition, clinical responder and non-responder subsets have been defined through the use of metabolic and immunological readouts. Herein, we review emerging T1D biomarkers within the context of recent and ongoing T1D immunotherapy trials. We also discuss responder/non-responder analyses in an effort to identify therapeutic mechanisms, define actionable pathways, and guide subject selection, drug dosing, and tailored combination drug therapy for future T1D trials.
Collapse
Affiliation(s)
- Laura M Jacobsen
- Department of Pediatrics, College of Medicine, University of Florida Diabetes Institute, Gainesville, FL, USA
| | - Brittney N Newby
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida Diabetes Institute, 1275 Center Drive, Biomedical Sciences Building J-589, Box 100275, Gainesville, FL, 32610, USA
| | - Daniel J Perry
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida Diabetes Institute, 1275 Center Drive, Biomedical Sciences Building J-589, Box 100275, Gainesville, FL, 32610, USA
| | - Amanda L Posgai
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida Diabetes Institute, 1275 Center Drive, Biomedical Sciences Building J-589, Box 100275, Gainesville, FL, 32610, USA
| | - Michael J Haller
- Department of Pediatrics, College of Medicine, University of Florida Diabetes Institute, Gainesville, FL, USA
| | - Todd M Brusko
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida Diabetes Institute, 1275 Center Drive, Biomedical Sciences Building J-589, Box 100275, Gainesville, FL, 32610, USA.
| |
Collapse
|