1
|
Rao C, Cater DT, Roy S, Xu J, De Oliveira AG, Evans-Molina C, Piganelli JD, Eizirik DL, Mirmira RG, Sims EK. Beta cell extracellular vesicle PD-L1 as a novel regulator of CD8 + T cell activity and biomarker during the evolution of type 1 diabetes. Diabetologia 2025; 68:382-396. [PMID: 39508879 DOI: 10.1007/s00125-024-06313-2] [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: 08/02/2024] [Accepted: 09/16/2024] [Indexed: 11/15/2024]
Abstract
AIMS/HYPOTHESIS Surviving beta cells in type 1 diabetes respond to inflammation by upregulating programmed death-ligand 1 (PD-L1) to engage immune cell programmed death protein 1 (PD-1) and limit destruction by self-reactive immune cells. Extracellular vesicles (EVs) and their cargo can serve as biomarkers of beta cell health and contribute to islet intercellular communication. We hypothesised that the inflammatory milieu of type 1 diabetes increases PD-L1 in beta cell EV cargo and that EV PD-L1 may protect beta cells against immune-mediated cell death. METHODS Beta cell lines and human islets were treated with proinflammatory cytokines to model the proinflammatory type 1 diabetes microenvironment. EVs were isolated using ultracentrifugation or size exclusion chromatography and analysed via immunoblot, flow cytometry and ELISA. EV PD-L1 binding to PD-1 was assessed using a competitive binding assay and in vitro functional assays testing the ability of EV PD-L1 to inhibit NOD CD8+ T cells. Plasma EV and soluble PD-L1 were assayed in the plasma of islet autoantibody-positive (Ab+) individuals or individuals with recent-onset type 1 diabetes and compared with levels in non-diabetic control individuals. RESULTS PD-L1 protein co-localised with tetraspanin-associated proteins intracellularly and was detected on the surface of beta cell EVs. Treatment with IFN-α or IFN-γ for 24 h induced a twofold increase in EV PD-L1 cargo without a corresponding increase in the number of EVs. IFN exposure predominantly increased PD-L1 expression on the surface of beta cell EVs and beta cell EV PD-L1 showed a dose-dependent capacity to bind PD-1. Functional experiments demonstrated specific effects of beta cell EV PD-L1 to suppress proliferation and cytotoxicity of murine CD8+ T cells. Plasma EV PD-L1 levels were increased in Ab+individuals, particularly in those positive for a single autoantibody. Additionally, in Ab+ individuals or those who had type 1 diabetes, but not in control individuals, plasma EV PD-L1 positively correlated with circulating C-peptide, suggesting that higher EV PD-L1 could be protective for residual beta cell function. CONCLUSIONS/INTERPRETATION IFN exposure increases PD-L1 on the beta cell EV surface. Beta cell EV PD-L1 binds PD1 and inhibits CD8+ T cell proliferation and cytotoxicity. Circulating EV PD-L1 is higher in Ab+ individuals than in control individuals. Circulating EV PD-L1 levels correlate with residual C-peptide at different stages in type 1 diabetes progression. These findings suggest that EV PD-L1 could contribute to heterogeneity in type 1 diabetes progression and residual beta cell function and raise the possibility that EV PD-L1 could be exploited as a means to inhibit immune-mediated beta cell death.
Collapse
Affiliation(s)
- Chaitra Rao
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Daniel T Cater
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Saptarshi Roy
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jerry Xu
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Andre G De Oliveira
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Carmella Evans-Molina
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jon D Piganelli
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Decio L Eizirik
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Raghavendra G Mirmira
- Department of Medicine and the Kovler Diabetes Center, The University of Chicago, Chicago, IL, USA
| | - Emily K Sims
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA.
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA.
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
2
|
Narendran P, Newland-Jones P, Kanumilli N, Stewart R, Regan F, Randell T. Highlighting the new consensus guidelines for managing people at risk of, and with early-stage type 1 diabetes-Relevance to clinical care in the UK. Diabet Med 2024:e15508. [PMID: 39733443 DOI: 10.1111/dme.15508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 12/15/2024] [Accepted: 12/23/2024] [Indexed: 12/31/2024]
Affiliation(s)
- Parth Narendran
- Diabetes Medicine, College of Medicine and Health, University of Birmingham, Birmingham, UK
- The Queen Elizabeth Hospital, Birmingham, UK
| | | | - Naresh Kanumilli
- Manchester University Foundation Trust and Clinical Network Lead for Diabetes-Greater Manchester, Manchester, UK
| | - Rose Stewart
- Betsi Cadwaladr University Health Board, Bangor, UK
| | - Fiona Regan
- Paediatric Endocrinology and Diabetes, Evelina London Children's Healthcare, London, UK
| | - Tabitha Randell
- Paediatric Endocrinology and Diabetes, Nottingham University Hospital, Nottingham, UK
| |
Collapse
|
3
|
Nostaeva A, Shimansky V, Apalko S, Kuznetsov I, Sushentseva N, Popov O, Asinovskaya A, Mosenko S, Karssen L, Sarana A, Aulchenko Y, Shcherbak S. Case-control association study between polygenic risk score and COVID-19 severity in a Russian population using low-pass genome sequencing. Epidemiol Infect 2024; 153:e13. [PMID: 39721951 DOI: 10.1017/s0950268824001778] [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] [Indexed: 12/28/2024] Open
Abstract
The course of COVID-19 is highly variable, with genetics playing a significant role. Through large-scale genetic association studies, a link between single nucleotide polymorphisms and disease susceptibility and severity was established. However, individual single nucleotide polymorphisms identified thus far have shown modest effects, indicating a polygenic nature of this trait, and individually have limited predictive performance. To address this limitation, we investigated the performance of a polygenic risk score model in the context of COVID-19 severity in a Russian population. A genome-wide polygenic risk score model including information from over a million common single nucleotide polymorphisms was developed using summary statistics from the COVID-19 Host Genetics Initiative consortium. Low-coverage sequencing (5x) was performed for ~1000 participants, and polygenic risk score values were calculated for each individual. A multivariate logistic regression model was used to analyse the association between polygenic risk score and COVID-19 outcomes. We found that individuals in the top 10% of the polygenic risk score distribution had a markedly elevated risk of severe COVID-19, with adjusted odds ratio of 2.9 (95% confidence interval: 1.8-4.6, p-value = 4e-06), and more than four times higher risk of mortality from COVID-19 (adjusted odds ratio = 4.3, p-value = 2e-05). This study highlights the potential of polygenic risk score as a valuable tool for identifying individuals at increased risk of severe COVID-19 based on their genetic profile.
Collapse
Affiliation(s)
- Arina Nostaeva
- City Hospital No. 40 of Kurortny District, St. Petersburg State Budgetary Healthcare Institution, Sestroretsk, Russia
- St. Petersburg State University, St. Petersburg, Russia
| | - Valentin Shimansky
- City Hospital No. 40 of Kurortny District, St. Petersburg State Budgetary Healthcare Institution, Sestroretsk, Russia
- St. Petersburg State University, St. Petersburg, Russia
| | - Svetlana Apalko
- City Hospital No. 40 of Kurortny District, St. Petersburg State Budgetary Healthcare Institution, Sestroretsk, Russia
- St. Petersburg State University, St. Petersburg, Russia
| | - Ivan Kuznetsov
- Center of Life Sciences, Skolkovo Institute of Science and Technology, Moscow, Russia
| | - Natalya Sushentseva
- City Hospital No. 40 of Kurortny District, St. Petersburg State Budgetary Healthcare Institution, Sestroretsk, Russia
| | - Oleg Popov
- City Hospital No. 40 of Kurortny District, St. Petersburg State Budgetary Healthcare Institution, Sestroretsk, Russia
- St. Petersburg State University, St. Petersburg, Russia
| | - Anna Asinovskaya
- City Hospital No. 40 of Kurortny District, St. Petersburg State Budgetary Healthcare Institution, Sestroretsk, Russia
- St. Petersburg State University, St. Petersburg, Russia
| | - Sergei Mosenko
- City Hospital No. 40 of Kurortny District, St. Petersburg State Budgetary Healthcare Institution, Sestroretsk, Russia
- St. Petersburg State University, St. Petersburg, Russia
| | | | - Andrey Sarana
- St. Petersburg State University, St. Petersburg, Russia
| | | | - Sergey Shcherbak
- City Hospital No. 40 of Kurortny District, St. Petersburg State Budgetary Healthcare Institution, Sestroretsk, Russia
- St. Petersburg State University, St. Petersburg, Russia
| |
Collapse
|
4
|
Mallone R, Bismuth E, Thivolet C, Benhamou PY, Hoffmeister N, Collet F, Nicolino M, Reynaud R, Beltrand J. Screening and care for preclinical stage 1-2 type 1 diabetes in first-degree relatives: French expert position statement. DIABETES & METABOLISM 2024; 51:101603. [PMID: 39675522 DOI: 10.1016/j.diabet.2024.101603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 11/29/2024] [Accepted: 12/11/2024] [Indexed: 12/17/2024]
Abstract
The natural history of type 1 diabetes (T1D) evolves from stage 1 (islet autoimmunity with normoglycemia; ICD-10 diagnostic code E10.A1) to stage 2 (autoimmunity with dysglycemia; E10.A2) and subsequent clinical stage 3 (overt hyperglycemia), which is commonly the first time of referral. Autoantibody testing can diagnose T1D at its preclinical stages 1-2 and lead to earlier initiation of care, particularly for first-degree relatives of people living with T1D, who are at higher genetic risk. Preclinical T1D screening and monitoring aims to avoid inaugural ketoacidosis and prolong preservation of endogenous insulin secretion, thereby improving glycemic control and reducing long-term morbidity. Moreover, early management can help coping with T1D and correct modifiable risk factors (obesity, sedentary lifestyle). New treatments currently under clinical deployment or trials also offer the possibility of delaying clinical progression. All these arguments lead to the proposition of a national screening and care pathway open to interested first-degree relatives. This pathway represents a new expertise to acquire for healthcare professionals. By adapting international consensus guidance to the French specificities, the proposed screening strategy involves testing for ≥ 2 autoantibodies (among IAA, anti-GAD, anti-IA-2) in relatives aged 2-45 years. Negative screening (∼95 % of cases) should be repeated every 4 years until the age of 12. A management workflow is proposed for relatives screening positive (∼5 % of cases), with immuno-metabolic monitoring by autoantibody testing, OGTT, glycemia and/or HbA1c of variable frequency, depending on T1D stage, age, patient preference and available resources, as well as the definition of expert centers for preclinical T1D.
Collapse
Affiliation(s)
- Roberto Mallone
- Université Paris Cité, Institut Cochin, CNRS, INSERM, Paris, France; Assistance Publique Hôpitaux de Paris, Université Paris Cité, Service de Diabétologie et Immunologie Clinique, Hôpital Cochin, Paris, France; Indiana Biosciences Research Institute, Indianapolis, IN, USA.
| | - Elise Bismuth
- Assistance Publique Hôpitaux de Paris, Université Paris Cité, Service d'Endocrinologie et Diabétologie Pédiatrique, Hôpital Robert Debré, Paris, France
| | - Charles Thivolet
- Hospices Civils de Lyon, Université de Lyon, Centre du diabète DIAB-eCARE, Lyon, France
| | - Pierre-Yves Benhamou
- Université Grenoble Alpes, INSERM U1055, LBFA, Endocrinologie, CHU Grenoble Alpes, France
| | | | - François Collet
- CHU Lille, Psychiatrie de Liaison et psycho-oncologie, Lille, France
| | - Marc Nicolino
- Hospices Civils de Lyon, Université de Lyon, Service d'Endocrinologie et Diabétologie Pédiatrique, Lyon, France
| | - Rachel Reynaud
- Assistance Publique Hôpitaux de Marseille, Université Aix-Marseille, Service de Pédiatrie Multidisciplinaire, Hôpital de la Timone, Marseille, France
| | - Jacques Beltrand
- Université Paris Cité, Institut Cochin, CNRS, INSERM, Paris, France; Assistance Publique Hôpitaux de Paris, Université Paris Cité, Service d'Endocrinologie, Gynécologie et Diabétologie Pédiatrique, Necker Hospital, Paris, France
| |
Collapse
|
5
|
Mari J, Solidoro S, Braida C, Tamaro G, Faleschini E, Tornese G. Perceptions and understanding of family pediatricians regarding the new Italian Type 1 Diabetes screening program. Diabetes Res Clin Pract 2024; 218:111931. [PMID: 39536974 DOI: 10.1016/j.diabres.2024.111931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 11/04/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
AIMS Italy is the first country to implement a nationwide Type 1 Diabetes (T1D) screening program aimed at reducing cases of diabetic ketoacidosis and potentially delaying disease progression through early interventions. This study assesses the knowledge, perceptions, and willingness of family pediatricians (FPs) to participate in this program. METHODS An anonymous online survey was conducted among 113 FPs in the Friuli Venezia Giulia region, an area not included in the initial pilot study. The survey evaluated their knowledge, readiness, and concerns regarding the screening program. RESULTS Of the 62 respondents (55% response rate), 84% expressed a willingness to participate in the screening program, although most reported limited knowledge about it. The key concerns included family anxiety, identifying individuals at risk of diabetes, and uncertainty regarding follow-up procedures. While FPs acknowledged the potential benefits of reducing ketoacidosis, 75% emphasized the need for more training and support. CONCLUSIONS While FPs are largely willing to participate in the T1D screening program, significant gaps in knowledge and preparedness remain. Addressing these gaps through comprehensive education and clear follow-up protocols is crucial for the successful nationwide implementation of the program in 2025.
Collapse
Affiliation(s)
- Johara Mari
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Sara Solidoro
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Cinzia Braida
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Gianluca Tamaro
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Elena Faleschini
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Gianluca Tornese
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy; Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy.
| |
Collapse
|
6
|
Jacobsen LM, Atkinson MA, Sosenko JM, Gitelman SE. Time to reframe the disease staging system for type 1 diabetes. Lancet Diabetes Endocrinol 2024; 12:924-933. [PMID: 39608963 DOI: 10.1016/s2213-8587(24)00239-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 06/16/2024] [Accepted: 07/25/2024] [Indexed: 11/30/2024]
Abstract
In 2015, introduction of a disease staging system offered a framework for benchmarking progression to clinical type 1 diabetes. This model, based on islet autoantibodies (stage 1) and dysglycaemia (stage 2) before type 1 diabetes diagnosis (stage 3), has facilitated screening and identification of people at risk. Yet, there are many limitations to this model as the stages combine a very heterogeneous group of individuals; do not have high specificity for type 1 diabetes; can occur without persistence (ie, reversion to an earlier risk stage); and exclude age and other influential risk factors. The current staging system also infers that individuals at risk of type 1 diabetes progress linearly from stage 1 to stage 2 and subsequently stage 3, whereas such movements are often more complex. With the approval of teplizumab by the US Food and Drug Administration in 2022 to delay type 1 diabetes in people at stage 2, there is a need to refine the definition and accuracy of type 1 diabetes staging. Theoretically, we propose that a type 1 diabetes risk calculator should incorporate any available demographic, genetic, autoantibody, metabolic, and immune data that could be continuously updated. Additionally, we call to action for the field to increase the breadth of knowledge regarding type 1 diabetes risk in non-relatives, adults, and individuals from minority populations.
Collapse
Affiliation(s)
- Laura M Jacobsen
- Department of Paediatrics and Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, USA.
| | - Mark A Atkinson
- Department of Paediatrics and Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Jay M Sosenko
- Division of Endocrinology, University of Miami, Miami, FL, USA
| | - Stephen E Gitelman
- Department of Paediatrics, Diabetes Center, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
7
|
Minerba E, Maines E, Quaglia N, Fedi L, Fanti S, Fierro A, Mozzillo E. Diabetes Awareness Campaigns to Prevent Ketoacidosis at the Diagnosis of Type 1 Diabetes: Efficacy on Multiple Outcomes and Predictors of Success: A Systematic Review. J Pers Med 2024; 14:1115. [PMID: 39728028 DOI: 10.3390/jpm14121115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/14/2024] [Accepted: 11/17/2024] [Indexed: 12/28/2024] Open
Abstract
Background/Objectives: In Italy, the incidence of diabetic ketoacidosis (DKA) at diagnosis of type 1 diabetes (T1D) is still very high (35.7-39.6%), especially in youths. We aimed to determine the efficacy of awareness campaigns to prevent DKA on multiple outcomes and identify success predictors. Methods: We searched electronic databases (Pubmed, Cochrane, and Web of Science) for studies published between 1 August 1990 and 1 August 2024. The review included studies that focused on children under 18 years old, and outcomes were measured by comparing before and after implementing the campaigns in the same area and between areas where interventions took place or not. Results: Of 236 records identified, 15 were eligible for analysis. After campaign implementation, the pooled DKA reduction resulted between 1% and 65.5%, based on the characteristics of the campaigns. A decrease in the rate of acute complications, such as cerebral edema, was reported. Hemoglobin A1c (HbA1c) at onset showed a mean reduction of 0.7-5.1%; C-peptide increased in patients without DKA at diagnosis, and length of hospitalization decreased. Campaign costs were lower than the costs of treating subjects with DKA. Conclusions: This review demonstrated that DKA awareness campaigns effectively reduce DKA incidence and improve other parameters, such as acute complications, HbA1c and C-peptide levels, length of hospitalization, and costs, among youths with T1D. To be effective, campaigns must follow specific principles of target population, modality, and minimal duration, as reported in this review.
Collapse
Affiliation(s)
- Elisa Minerba
- Pediatric Diabetology Unit, Pediatric Department, S.Chiara General Hospital of Trento, Azienda Provinciale per i Servizi Sanitari, Largo Medaglie d'Oro 9, 38122 Trento, Italy
| | - Evelina Maines
- Pediatric Diabetology Unit, Pediatric Department, S.Chiara General Hospital of Trento, Azienda Provinciale per i Servizi Sanitari, Largo Medaglie d'Oro 9, 38122 Trento, Italy
| | - Nadia Quaglia
- Pediatric Diabetology Unit, Pediatric Department, S.Chiara General Hospital of Trento, Azienda Provinciale per i Servizi Sanitari, Largo Medaglie d'Oro 9, 38122 Trento, Italy
| | - Ludovica Fedi
- Section of Pediatrics, Regional Center of Pediatric Diabetes, Department of Translational Medical Science, Federico II University of Naples, 80138 Naples, Italy
| | - Stefania Fanti
- Pediatric Diabetology Unit, Pediatric Department, S.Chiara General Hospital of Trento, Azienda Provinciale per i Servizi Sanitari, Largo Medaglie d'Oro 9, 38122 Trento, Italy
| | - Alessandro Fierro
- Section of Pediatrics, Regional Center of Pediatric Diabetes, Department of Translational Medical Science, Federico II University of Naples, 80138 Naples, Italy
| | - Enza Mozzillo
- Section of Pediatrics, Regional Center of Pediatric Diabetes, Department of Translational Medical Science, Federico II University of Naples, 80138 Naples, Italy
| |
Collapse
|
8
|
Roberts AG, Tully AS, Binkowski SK, Bebbington KR, Penno MAS, Anderson AJ, Craig ME, Colman PG, Huynh T, McGorm KJ, Soldatos G, Vuillermin PJ, Wentworth JM, Davis EA, Couper JJ, Haynes A. Parental experiences of using continuous glucose monitoring in their young children with early-stage type 1 diabetes: a qualitative interview study. FRONTIERS IN CLINICAL DIABETES AND HEALTHCARE 2024; 5:1479948. [PMID: 39611061 PMCID: PMC11602481 DOI: 10.3389/fcdhc.2024.1479948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 10/29/2024] [Indexed: 11/30/2024]
Abstract
Aim To explore parents' experiences of using continuous glucose monitoring (CGM) in their young children with early-stage type 1 diabetes, being followed in the Australian Environmental Determinants of Islet Autoimmunity (ENDIA) study. Methods Parents of children with persistent islet autoimmunity who enrolled in the ENDIA CGM sub-study were invited to participate in an optional interview. Semi-structured phone interviews were conducted by a single researcher using an interview guide developed by a multi-disciplinary team. Interviews were conducted following a single CGM monitoring period and prior to parents receiving feedback on their child's glycemic status. Following transcription, thematic analysis was conducted to determine common themes. Results Nine parents (8 mothers, 1 father) were interviewed corresponding to ten children, with a mean (SD) age of 5.6 (2.2) years, who wore CGM for 97 (0.1)% of the time during their monitoring period. Three main themes were identified: (1) Information empowers and helps to reduce uncertainty; (2) Families' acceptance of using CGM; and (3) Involvement in research provides support and preparation for the unknown. Conclusions Parents reported a positive experience of their young child wearing blinded CGM, and the children tolerated wearing CGM very well. Parents were empowered by knowing they would receive information on their child's glucose levels and patterns and felt well supported. This study provides novel insights into parents' experiences of using CGM in very young children with early-stage type 1 diabetes.
Collapse
Affiliation(s)
- Alison G. Roberts
- Department of Endocrinology and Diabetes, Perth Children’s Hospital, Perth, WA, Australia
- Children’s Diabetes Centre, Telethon Kids Institute, The University of Western Australia, Perth, WA, Australia
| | - Alexandra S. Tully
- Children’s Diabetes Centre, Telethon Kids Institute, The University of Western Australia, Perth, WA, Australia
| | - Sabrina K. Binkowski
- Children’s Diabetes Centre, Telethon Kids Institute, The University of Western Australia, Perth, WA, Australia
| | - Keely R. Bebbington
- Children’s Diabetes Centre, Telethon Kids Institute, The University of Western Australia, Perth, WA, Australia
| | - Megan A. S. Penno
- The University of Adelaide, Robinson Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Amanda J. Anderson
- The University of Adelaide, Robinson Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Maria E. Craig
- School of Clinical Medicine, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
- Department of Diabetes & Endocrinology, The Children’s Hospital at Westmead, Sydney, NSW, Australia
| | - Peter G. Colman
- Melbourne Health Pathology, Department of Diabetes and Endocrinology, The Royal Melbourne Hospital, Parkville, VIC, Australia
| | - Tony Huynh
- Department of Endocrinology and Diabetes, Queensland Children’s Hospital, South Brisbane, QLD, Australia
- Children’s Health Research Centre, Faculty of Medicine, The University of Queensland, South Brisbane, QLD, Australia
- Department of Chemical Pathology, Mater Health Services, South Brisbane, QLD, Australia
| | - Kelly J. McGorm
- The University of Adelaide, Robinson Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Georgia Soldatos
- Monash Centre for Health Research and Implementation, School of Public Health and Preventive Medicine, Monash University, Melbourne and Diabetes and Vascular Medicine Unit, Monash Health, Melbourne, VIC, Australia
| | - Peter J. Vuillermin
- Faculty of School of Medicine, Deakin University and Child Health Research Unit, Barwon Health, Geelong, VIC, Australia
| | - John M. Wentworth
- Melbourne Health Pathology, Department of Diabetes and Endocrinology, The Royal Melbourne Hospital, Parkville, VIC, Australia
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
| | - Elizabeth A. Davis
- Department of Endocrinology and Diabetes, Perth Children’s Hospital, Perth, WA, Australia
- Children’s Diabetes Centre, Telethon Kids Institute, The University of Western Australia, Perth, WA, Australia
| | - Jennifer J. Couper
- The University of Adelaide, Robinson Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
- Department of Diabetes & Endocrinology, Women’s and Children’s Hospital, Adelaide, SA, Australia
| | - Aveni Haynes
- Children’s Diabetes Centre, Telethon Kids Institute, The University of Western Australia, Perth, WA, Australia
- The University of Western Australia (UWA) Medical School, Pediatrics, Perth, WA, Australia
| |
Collapse
|
9
|
O’Donovan AJ, Gorelik S, Nally LM. Shifting the paradigm of type 1 diabetes: a narrative review of disease modifying therapies. Front Endocrinol (Lausanne) 2024; 15:1477101. [PMID: 39568817 PMCID: PMC11576206 DOI: 10.3389/fendo.2024.1477101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 09/26/2024] [Indexed: 11/22/2024] Open
Abstract
A new diagnosis of type 1 diabetes (T1D) may be accompanied by numerous lifelong financial, emotional, and physical challenges, thus advancements in therapies that can delay the onset of clinical disease are crucial. T1D is an autoimmune condition involving destruction of pancreatic beta cells leading to insulin deficiency, hyperglycemia, and long-term insulin dependence. The pathogenesis of T1D is classified into stages, with the first signal being the detection of autoantibodies without any glycemic changes. In the second stage, dysglycemia develops without symptoms, and in stage 3, symptoms of hyperglycemia become apparent, and at this time a clinical diagnosis of T1D is made. As a greater understanding of these stages of T1D have evolved, research efforts have been devoted to delaying the onset of clinical disease. To date, only one medication, teplizumab, has been approved by the Food and Drug Administration (FDA) for the treatment of stage 2 T1D. This narrative review present published trials and ongoing research on disease modifying therapies (DMT) in T1D, the mechanisms of action for each therapy, and the stages of T1D that these interventions are being studied.
Collapse
Affiliation(s)
- Alexander J. O’Donovan
- Yale University School of Medicine, Department of Pediatrics, New Haven, CT, United States
| | - Seth Gorelik
- Yale University School of Medicine, Department of Pediatrics, New Haven, CT, United States
- Bowdoin College, Brunswick, ME, United States
| | - Laura M. Nally
- Yale University School of Medicine, Department of Pediatrics, New Haven, CT, United States
| |
Collapse
|
10
|
Greenbaum CJ, Nepom GT, Wood-Heickman LK, Wherrett DK, DiMeglio LA, Herold KC, Krischer JP. Evolving Concepts in Pathophysiology, Screening, and Prevention of Type 1 Diabetes: Report of Diabetes Mellitus Interagency Coordinating Committee Workshop. Diabetes 2024; 73:1780-1790. [PMID: 39167668 PMCID: PMC11493760 DOI: 10.2337/dbi24-0020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 08/01/2024] [Indexed: 08/23/2024]
Abstract
The approval of teplizumab to delay the onset of type 1 diabetes is an important inflection point in the decades-long pursuit to treat the cause of the disease rather than its symptoms. The National Institute of Diabetes and Digestive and Kidney Diseases convened a workshop of the Diabetes Mellitus Interagency Coordinating Committee titled "Evolving Concepts in Pathophysiology, Screening, and Prevention of Type 1 Diabetes" to review this accomplishment and identify future goals. Speakers representing Type 1 Diabetes TrialNet (TrialNet) and the Immune Tolerance Network emphasized that the ability to robustly identify individuals destined to develop type 1 diabetes was essential for clinical trials. The presenter from the U.S. Food and Drug Administration described how regulatory approval relied on data from the single clinical trial of TrialNet with testing of teplizumab for delay of clinical diagnosis, along with confirmatory evidence from studies in patients after diagnosis. The workshop reviewed the etiology of type 1 diabetes as a disease involving multiple immune pathways, highlighting the current understanding of prognostic markers and proposing potential strategies to improve the therapeutic response of disease-modifying therapies based on the mechanism of action. While celebrating these achievements funded by the congressionally appropriated Special Diabetes Program, panelists from professional organizations, nonprofit advocacy/funding groups, and industry also identified significant hurdles in translating this research into clinical care.
Collapse
Affiliation(s)
- Carla J. Greenbaum
- Center for Interventional Immunology, Benaroya Research Institute, Seattle, WA
| | - Gerald T. Nepom
- Immune Tolerance Network, Benaroya Research Institute, Seattle, WA
| | - Lauren K. Wood-Heickman
- Division of Diabetes, Lipid Disorders and Obesity in the Office of New Drugs, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD
| | - Diane K. Wherrett
- Paediatric Endocrinology, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Linda A. DiMeglio
- Department of Pediatrics, Riley Hospital for Children, Indiana University School of Medicine, Indianapolis, IN
| | - Kevan C. Herold
- Departments of Immunobiology and Internal Medicine, Yale University, New Haven, CT
| | - Jeffrey P. Krischer
- Departments of Pediatrics and Internal Medicine, Health Informatics Institute, Morsani College of Medicine, University of South Florida, Tampa, FL
| |
Collapse
|
11
|
Ray S, Palui R. Immunotherapy in type 1 diabetes: Novel pathway to the future ahead. World J Diabetes 2024; 15:2022-2035. [PMID: 39493558 PMCID: PMC11525730 DOI: 10.4239/wjd.v15.i10.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 07/23/2024] [Accepted: 07/26/2024] [Indexed: 09/26/2024] Open
Abstract
Since the discovery of insulin over 100 years ago, the focus of research in the management of type 1 diabetes (T1D) has centered around glycemic control and management of complications rather than the prevention of autoimmune destruction of pancreatic β cells. Fortunately, in recent years, there has been significant advancement in immune-targeted pharmacotherapy to halt the natural progression of T1D. The immune-targeted intervention aims to alter the underlying pathogenesis of T1D by targeting different aspects of the immune system. The immunotherapy can either antagonize the immune mediators like T cells, B cells or cytokines (antibody-based therapy), or reinduce self-tolerance to pancreatic β cells (antigen-based therapy) or stem-cell treatment. Recently, the US Food and Drug Administration approved the first immunotherapy teplizumab to be used only in stage 2 of T1D. However, the window of opportunity to practically implement this approved molecule in the selected target population is limited. In this Editorial, we briefly discuss the various promising recent developments in the field of immunotherapy research in T1D. However, further studies of these newer therapeutic agents are needed to explore their true potential for prevention or cure of T1D.
Collapse
Affiliation(s)
- Sayantan Ray
- Department of Endocrinology, All India Institute of Medical Sciences, Bhubaneswar 751019, India
| | - Rajan Palui
- Department of Endocrinology, The Mission Hospital, Durgapur 713212, India
| |
Collapse
|
12
|
Roy S, Pokharel P, Piganelli JD. Decoding the immune dance: Unraveling the interplay between beta cells and type 1 diabetes. Mol Metab 2024; 88:101998. [PMID: 39069156 PMCID: PMC11342121 DOI: 10.1016/j.molmet.2024.101998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 07/12/2024] [Accepted: 07/23/2024] [Indexed: 07/30/2024] Open
Abstract
BACKGROUND Type 1 diabetes (T1D) is an autoimmune disease characterized by the specific destruction of insulin-producing beta cells in the pancreas by the immune system, including CD4 cells which orchestrate the attack and CD8 cells which directly destroy the beta cells, resulting in the loss of glucose homeostasis. SCOPE OF REVIEW This comprehensive document delves into the complex interplay between the immune system and beta cells, aiming to shed light on the mechanisms driving their destruction in T1D. Insights into the genetic predisposition, environmental triggers, and autoimmune responses provide a foundation for understanding the autoimmune attack on beta cells. From the role of viral infections as potential triggers to the inflammatory response of beta cells, an intricate puzzle starts to unfold. This exploration highlights the importance of beta cells in breaking immune tolerance and the factors contributing to their targeted destruction. Furthermore, it examines the potential role of autophagy and the impact of cytokine signaling on beta cell function and survival. MAJOR CONCLUSIONS This review collectively represents current research findings on T1D which offers valuable perspectives on novel therapeutic approaches for preserving beta cell mass, restoring immune tolerance, and ultimately preventing or halting the progression of T1D. By unraveling the complex dynamics between the immune system and beta cells, we inch closer to a comprehensive understanding of T1D pathogenesis, paving the way for more effective treatments and ultimately a cure.
Collapse
Affiliation(s)
- Saptarshi Roy
- Department of Endocrinology, Indiana University School of Medicine, Indianapolis, IN, 46202, United States
| | - Pravil Pokharel
- Department of Endocrinology, Indiana University School of Medicine, Indianapolis, IN, 46202, United States
| | - Jon D Piganelli
- Department of Endocrinology, Indiana University School of Medicine, Indianapolis, IN, 46202, United States.
| |
Collapse
|
13
|
Haynes A, Tully A, Smith GJ, Penno MA, Craig ME, Wentworth JM, Huynh T, Colman PG, Soldatos G, Anderson AJ, McGorm KJ, Oakey H, Couper JJ, Davis EA. Early Dysglycemia Is Detectable Using Continuous Glucose Monitoring in Very Young Children at Risk of Type 1 Diabetes. Diabetes Care 2024; 47:1750-1756. [PMID: 39159241 PMCID: PMC11417303 DOI: 10.2337/dc24-0540] [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: 03/15/2024] [Accepted: 06/28/2024] [Indexed: 08/21/2024]
Abstract
OBJECTIVE Continuous glucose monitoring (CGM) can detect early dysglycemia in older children and adults with presymptomatic type 1 diabetes (T1D) and predict risk of progression to clinical onset. However, CGM data for very young children at greatest risk of disease progression are lacking. This study aimed to investigate the use of CGM data measured in children being longitudinally observed in the Australian Environmental Determinants of Islet Autoimmunity (ENDIA) study from birth to age 10 years. RESEARCH DESIGN AND METHODS Between January 2021 and June 2023, 31 ENDIA children with persistent multiple islet autoimmunity (PM Ab+) and 24 age-matched control children underwent CGM assessment alongside standard clinical monitoring. The CGM metrics of glucose SD (SDSGL), coefficient of variation (CEV), mean sensor glucose (SGL), and percentage of time >7.8 mmol/L (>140 mg/dL) were determined and examined for between-group differences. RESULTS The mean (SD) ages of PM Ab+ and Ab- children were 4.4 (1.8) and 4.7 (1.9) years, respectively. Eighty-six percent of eligible PM Ab+ children consented to CGM wear, achieving a median (quartile 1 [Q1], Q3) sensor wear period of 12.5 (9.0, 15.0) days. PM Ab+ children had higher median (Q1, Q3) SDSGL (1.1 [0.9, 1.3] vs. 0.9 [0.8, 1.0] mmol/L; P < 0.001) and CEV (17.3% [16.0, 20.9] vs. 14.7% [12.9, 16.6]; P < 0.001). Percentage of time >7.8 mmol/L was greater in PM Ab+ children (median [Q1, Q3] 8.0% [4.4, 13.0] compared with 3.3% [1.4, 5.3] in Ab- children; P = 0.005). Mean SGL did not differ significantly between groups (P = 0.10). CONCLUSIONS CGM is feasible and well tolerated in very young children at risk of T1D. Very young PM Ab+ children have increased SDSGL, CEV, and percentage of time >7.8 mmol/L, consistent with prior studies involving older participants.
Collapse
Affiliation(s)
- Aveni Haynes
- Children’s Diabetes Centre, Telethon Kids Institute, University of Western Australia, Nedlands, Western Australia, Australia
- Paediatrics, UWA Medical School, University of Western Australia, Nedlands, Western Australia, Australia
| | - Alexandra Tully
- Children’s Diabetes Centre, Telethon Kids Institute, University of Western Australia, Nedlands, Western Australia, Australia
| | - Grant J. Smith
- Children’s Diabetes Centre, Telethon Kids Institute, University of Western Australia, Nedlands, Western Australia, Australia
| | - Megan A.S. Penno
- Faculty of Health and Medical Sciences and Robinson Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Maria E. Craig
- Faculty of Medicine, School of Women’s and Children’s Health, University of New South Wales, Sydney, New South Wales, Australia
- Institute of Endocrinology and Diabetes, Children’s Hospital at Westmead, Sydney, New South Wales, Australia
| | - John M. Wentworth
- Department of Diabetes and Endocrinology, Royal Melbourne Hospital, Melbourne, Victoria, Australia
- Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Tony Huynh
- Department of Endocrinology and Diabetes, Queensland Children’s Hospital, South Brisbane, Queensland, Australia
- Faculty of Medicine, Children’s Health Research Centre, University of Queensland, South Brisbane, Queensland, Australia
| | - Peter G. Colman
- Department of Diabetes and Endocrinology, Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Georgia Soldatos
- Monash Centre for Health Research and Implementation, School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
- Diabetes and Vascular Medicine Unit, Monash Health, Melbourne, Victoria, Australia
| | - Amanda J. Anderson
- Faculty of Health and Medical Sciences and Robinson Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Kelly J. McGorm
- Faculty of Health and Medical Sciences and Robinson Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Helena Oakey
- Faculty of Health and Medical Sciences and Robinson Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Jennifer J. Couper
- Department of Diabetes and Endocrinology, Women’s and Children’s Hospital, Adelaide, South Australia, Australia
| | - Elizabeth A. Davis
- Children’s Diabetes Centre, Telethon Kids Institute, University of Western Australia, Nedlands, Western Australia, Australia
- Department of Diabetes and Endocrinology, Perth Children’s Hospital, Nedlands, Western Australia, Australia
- School of Paediatrics, University of Western Australia, Nedlands, Western Australia, Australia
| |
Collapse
|
14
|
Kelly CS, Wolf WA, Cornelius EM, Peter ME, Chapman KS, Dunne JL. Insights into Knowledge and Attitudes About Autoantibody Screening from People Affected by Type 1 Diabetes: A Brief Report. Diabetes Ther 2024; 15:2249-2261. [PMID: 39190273 PMCID: PMC11410738 DOI: 10.1007/s13300-024-01637-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 07/31/2024] [Indexed: 08/28/2024] Open
Abstract
INTRODUCTION Screening for islet-specific autoantibodies can identify individuals at risk for type 1 diabetes (T1D). Despite calls for increased nationwide autoantibody screening efforts, it is unclear how many individuals have participated in screening among people who may benefit from it. Moreover, knowledge and perceptions of autoantibody screening in real-world samples are not well understood. METHODS We surveyed a sample of individuals (aged 18+ years old) from T1D Exchange Registry with a personal or family history of T1D to assess their self-reported T1D autoantibody knowledge, experiences, and attitudes. Participants belonged to one of three groups: adults with T1D who had a biological child without T1D or future plans for a child (PWD); parents without T1D who had a biological child with T1D and one or more biological children without T1D (Caregivers); and first-degree adult children or siblings to a person with T1D (Relatives). Descriptive analyses (means, standard deviations, frequencies) are presented by participant groups. RESULTS A total of 510 participants enrolled in the study. Across groups, participants reported feeling a little to somewhat knowledgeable about autoantibody screening and positive perceptions of autoantibody screening in general. However, few participants had screened their child without T1D (PWDs, 21.94%; Caregivers, 46.30%) or themselves (Relatives, 19.23%). Among those who had screened, participants reported generally positive experiences. Among those who had not screened, many participants were "undecided" about autoantibody screening (PWD, 38.46%; Caregivers, 40.52%; Relatives, 44.44%). Influences reported for participants' decisions to screen, not screen, or their current indecision differed by group: PWDs (21.70%) and Caregivers (26.87%) most often reported self-initiated research as an influence and Relatives reported they had not previously considered screening (48.28%). CONCLUSION Results highlight the need for more accessible information about screening, including real experiences from those who have screened.
Collapse
Affiliation(s)
- Caitlin S Kelly
- T1D Exchange, 101 Federal St Suite 440, Boston, MA, 02110, USA.
| | - Wendy A Wolf
- T1D Exchange, 101 Federal St Suite 440, Boston, MA, 02110, USA
| | | | - Megan E Peter
- T1D Exchange, 101 Federal St Suite 440, Boston, MA, 02110, USA
| | | | - Jessica L Dunne
- Janssen R&D, 1000 U.S. Route 202 South, Raritan, NJ, 08869, USA
- General Medicine, US Medical Bridgewater, Sanofi, NJ, 08807, USA
| |
Collapse
|
15
|
Quinn LM, Dias RP, Bidder C, Bhowmik S, Bumke K, Ganapathi J, Gorman S, Hind E, Karandikar S, Kumar K, Lipscomb N, McGovern S, Puthi VR, Randell T, Watts G, Narendran P. Presentation and characteristics of children with screen-detected type 1 diabetes: learnings from the ELSA general population pediatric screening study. BMJ Open Diabetes Res Care 2024; 12:e004480. [PMID: 39327068 PMCID: PMC11429353 DOI: 10.1136/bmjdrc-2024-004480] [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: 09/05/2024] [Indexed: 09/28/2024] Open
Abstract
INTRODUCTION We describe the identification and management of general population screen-detected type 1 diabetes (T1D) and share learnings for best practice. RESEARCH DESIGN AND METHODS Children diagnosed with T1D through a general population screening initiative, the EarLy Surveillance for Autoimmune diabetes (ELSA) study, were reviewed and described.Parents provided written, informed consent for inclusion in the case series. RESULTS 14 children with insulin requiring (stage 3) T1D are described. These cases offer unique insights into the features of screen-detected T1D. T1D is identified sooner through screening programs, characterized by absent/short symptom duration, median presenting glycated hemoglobin 6.6% (49 mmol/mol) and insulin requirements<0.5 units/kg/day. ELSA identified four children at stage 3 and another 4 progressed within 4 months of ELSA completion, including two single seropositive children. Six children developed stage 3 T1D prior to ELSA completion, including two children (14%, n=2/14) with diabetic ketoacidosis prior to confirmed antibody status. CONCLUSIONS There are three main learnings from this case series. First, T1D identified through screening is at an earlier stage of its natural history and requires personalized insulin regimens with lower total daily insulin doses. Second, single autoantibody seropositivity can rapidly progress to stage 3. Finally, insulin requirement can manifest at any stage of the T1D screening pathway, and therefore early education around symptom recognition is essential for families participating in screening programs.
Collapse
Affiliation(s)
- Lauren M Quinn
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Renuka P Dias
- Institute of Applied Health Research, University of Birmingham, Birmingham, UK
- Department of Paediatric Endocrinology, Birmingham Women’s and Children’s Hospitals NHS Foundation Trust, Birmingham, UK
| | - Christopher Bidder
- Department of Child health, Swansea Bay University Health Board, Morriston Hospital, Swansea, UK
| | | | - Kerstin Bumke
- Paediatric Department, University Hospital Wishaw, Wishaw, UK
| | | | - Shaun Gorman
- Bradford Teaching Hospitals NHS Foundation Trust, Bradford, UK
| | - Edward Hind
- North Hampshire Hospital, Basingstoke, Hampshire, UK
| | | | - Kiran Kumar
- Burton Hospitals NHS Foundation Trust, Derby, UK
| | - Nicholas Lipscomb
- Department of Paediatrics, South West Acute Hospital, Enniskillen, UK
| | | | - Vijith R Puthi
- Department of Paediatrics, Peterborough City Hospital, Peterborough, UK
| | | | | | - Parth Narendran
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
- Department of Diabetes, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| |
Collapse
|
16
|
Rao C, Cater DT, Roy S, Xu J, Olivera ADG, Evans-Molina C, Piganelli JD, Eizirik DL, Mirmira RG, Sims EK. Beta cell extracellular vesicle PD-L1 as a novel regulator of CD8+ T cell activity and biomarker during the evolution of Type 1 Diabetes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.18.613649. [PMID: 39345410 PMCID: PMC11429676 DOI: 10.1101/2024.09.18.613649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Aims/hypothesis Surviving beta cells in type 1 diabetes respond to inflammation by upregulating programmed death-ligand 1 (PD-L1) to engage immune cell programmed death-1 (PD-1) and limit destruction by self-reactive immune cells. Extracellular vesicles (EVs) and their cargo can serve as biomarkers of beta cell health and contribute to islet intercellular communication. We hypothesized that the inflammatory milieu of type 1 diabetes increases PD-L1 in beta cell EV cargo and that EV PD-L1 may protect beta cells against immune-mediated cell death. Methods Beta cell lines and human islets were treated with proinflammatory cytokines to model the proinflammatory type 1 diabetes microenvironment. EVs were isolated using ultracentrifugation or size exclusion chromatography and analysed via immunoblot, flow cytometry, and ELISA. EV PD-L1: PD-1 binding was assessed using a competitive binding assay and in vitro functional assays testing the ability of EV PD-L1 to inhibit NOD CD8 T cells. Plasma EV and soluble PD-L1 were assayed in plasma of individuals with islet autoantibody positivity (Ab+) or recent-onset type 1 diabetes and compared to non-diabetic controls. Results PD-L1 protein colocalized with tetraspanin-associated proteins intracellularly and was detected on the surface of beta cell EVs. 24-h IFN-α or IFN-γ treatment induced a two-fold increase in EV PD-L1 cargo without a corresponding increase in number of EVs. IFN exposure predominantly increased PD-L1 expression on the surface of beta cell EVs and beta cell EV PD-L1 showed a dose-dependent capacity to bind PD-1. Functional experiments demonstrated specific effects of beta cell EV PD-L1 to suppress proliferation and cytotoxicity of murine CD8 T cells. Plasma EV PD-L1 levels were increased in islet Ab+ individuals, particularly in those with single Ab+, Additionally, in from individuals with either Ab+ or type 1 diabetes, but not in controls, plasma EV PD-L1 positively correlated with circulating C-peptide, suggesting that higher EV-PD-L1 could be protective for residual beta cell function. Conclusions/interpretation IFN exposure increases PD-L1 on the beta cell EV surface. Beta cell EV PD-L1 binds PD1 and inhibits CD8 T cell proliferation and cytotoxicity. Circulating EV PD-L1 is higher in islet autoantibody positive patients compared to controls. Circulating EV PD-L1 levels correlate with residual C-peptide at different stages in type 1 diabetes progression. These findings suggest that EV PD-L1 could contribute to heterogeneity in type 1 diabetes progression and residual beta cell function and raise the possibility that EV PD-L1 could be exploited as a means to inhibit immune-mediated beta cell death.
Collapse
Affiliation(s)
- Chaitra Rao
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
- Authors contributed equally to this work
| | - Daniel T Cater
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
- Authors contributed equally to this work
| | - Saptarshi Roy
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jerry Xu
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Andre De G Olivera
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Carmella Evans-Molina
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jon D. Piganelli
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Decio L. Eizirik
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Raghavendra G. Mirmira
- Department of Medicine and the Kovler Diabetes Center, The University of Chicago, Chicago, IL, USA
| | - Emily K. Sims
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
17
|
Phillip M, Achenbach P, Addala A, Albanese-O'Neill A, Battelino T, Bell KJ, Besser REJ, Bonifacio E, Colhoun HM, Couper JJ, Craig ME, Danne T, de Beaufort C, Dovc K, Driscoll KA, Dutta S, Ebekozien O, Larsson HE, Feiten DJ, Frohnert BI, Gabbay RA, Gallagher MP, Greenbaum CJ, Griffin KJ, Hagopian W, Haller MJ, Hendrieckx C, Hendriks E, Holt RIG, Hughes L, Ismail HM, Jacobsen LM, Johnson SB, Kolb LE, Kordonouri O, Lange K, Lash RW, Lernmark Å, Libman I, Lundgren M, Maahs DM, Marcovecchio ML, Mathieu C, Miller KM, O'Donnell HK, Oron T, Patil SP, Pop-Busui R, Rewers MJ, Rich SS, Schatz DA, Schulman-Rosenbaum R, Simmons KM, Sims EK, Skyler JS, Smith LB, Speake C, Steck AK, Thomas NPB, Tonyushkina KN, Veijola R, Wentworth JM, Wherrett DK, Wood JR, Ziegler AG, DiMeglio LA. Consensus guidance for monitoring individuals with islet autoantibody-positive pre-stage 3 type 1 diabetes. Diabetologia 2024; 67:1731-1759. [PMID: 38910151 PMCID: PMC11410955 DOI: 10.1007/s00125-024-06205-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/25/2024]
Abstract
Given the proven benefits of screening to reduce diabetic ketoacidosis (DKA) likelihood at the time of stage 3 type 1 diabetes diagnosis, and emerging availability of therapy to delay disease progression, type 1 diabetes screening programmes are being increasingly emphasised. Once broadly implemented, screening initiatives will identify significant numbers of islet autoantibody-positive (IAb+) children and adults who are at risk of (confirmed single IAb+) or living with (multiple IAb+) early-stage (stage 1 and stage 2) type 1 diabetes. These individuals will need monitoring for disease progression; much of this care will happen in non-specialised settings. To inform this monitoring, JDRF in conjunction with international experts and societies developed consensus guidance. Broad advice from this guidance includes the following: (1) partnerships should be fostered between endocrinologists and primary-care providers to care for people who are IAb+; (2) when people who are IAb+ are initially identified there is a need for confirmation using a second sample; (3) single IAb+ individuals are at lower risk of progression than multiple IAb+ individuals; (4) individuals with early-stage type 1 diabetes should have periodic medical monitoring, including regular assessments of glucose levels, regular education about symptoms of diabetes and DKA, and psychosocial support; (5) interested people with stage 2 type 1 diabetes should be offered trial participation or approved therapies; and (6) all health professionals involved in monitoring and care of individuals with type 1 diabetes have a responsibility to provide education. The guidance also emphasises significant unmet needs for further research on early-stage type 1 diabetes to increase the rigour of future recommendations and inform clinical care.
Collapse
Affiliation(s)
- Moshe Phillip
- Institute for Endocrinology and Diabetes, National Center for Childhood Diabetes, Schneider Children's Medical Center of Israel, Petah Tikva, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Peter Achenbach
- Institute of Diabetes Research, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany
- Forschergruppe Diabetes, Technical University Munich, Klinikum Rechts Der Isar, Munich, Germany
| | - Ananta Addala
- Division of Endocrinology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Tadej Battelino
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Department of Endocrinology, Diabetes and Metabolism, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Kirstine J Bell
- Charles Perkins Centre and Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Rachel E J Besser
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Wellcome Centre Human Genetics, Nuffield Department of Medicine Oxford NIHR Biomedical Research Centre, University of Oxford, Oxford, UK
- Department of Paediatrics, University of Oxford, Oxford, UK
| | - Ezio Bonifacio
- Center for Regenerative Therapies Dresden, Faculty of Medicine, Technical University of Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden, Helmholtz Centre Munich at the University Clinic Carl Gustav Carus of TU Dresden and Faculty of Medicine, Dresden, Germany
| | - Helen M Colhoun
- The Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
- Department of Public Health, NHS Fife, Kirkcaldy, UK
| | - Jennifer J Couper
- Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
- Division of Paediatrics, Women's and Children's Hospital, Adelaide, SA, Australia
| | - Maria E Craig
- Charles Perkins Centre and Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
- Discipline of Paediatrics & Child Health, School of Clinical Medicine, UNSW Medicine & Health, Sydney, NSW, Australia
| | | | - Carine de Beaufort
- International Society for Pediatric and Adolescent Diabetes (ISPAD), Berlin, Germany
- Diabetes & Endocrine Care Clinique Pédiatrique (DECCP), Clinique Pédiatrique/Centre Hospitalier (CH) de Luxembourg, Luxembourg City, Luxembourg
- Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-Belval, Luxembourg
| | - Klemen Dovc
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Department of Endocrinology, Diabetes and Metabolism, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Kimberly A Driscoll
- Department of Pediatrics, Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Clinical and Health Psychology, University of Florida, Gainesville, FL, USA
- Department of Pediatrics, University of Florida Diabetes Institute, Gainesville, FL, USA
| | | | | | - Helena Elding Larsson
- Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Department of Pediatrics, Skåne University Hospital, Malmö and Lund, Sweden
| | | | - Brigitte I Frohnert
- Department of Pediatrics, Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | | | | | - Carla J Greenbaum
- Center for Interventional Immunology and Diabetes Program, Benaroya Research Institute, Seattle, WA, USA
| | - Kurt J Griffin
- Sanford Research, Sioux Falls, SD, USA
- Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD, USA
| | - William Hagopian
- Pacific Northwest Diabetes Research Institute, University of Washington, Seattle, WA, USA
| | - Michael J Haller
- Department of Pediatrics, University of Florida Diabetes Institute, Gainesville, FL, USA
- Division of Endocrinology, University of Florida College of Medicine, Gainesville, FL, USA
| | - Christel Hendrieckx
- School of Psychology, Deakin University, Geelong, VIC, Australia
- The Australian Centre for Behavioural Research in Diabetes, Diabetes Victoria, Carlton, VIC, Australia
- Institute for Health Transformation, Deakin University, Geelong, VIC, Australia
| | - Emile Hendriks
- Department of Paediatrics, University of Cambridge and Cambridge University Hospitals NHS Foundation Trust, Cambridge Biomedical Campus, Cambridge, UK
| | - Richard I G Holt
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
- National Institute for Health and Care Research Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | | | - Heba M Ismail
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Laura M Jacobsen
- Division of Endocrinology, University of Florida College of Medicine, Gainesville, FL, USA
| | - Suzanne B Johnson
- Department of Behavioral Sciences and Social Medicine, Florida State University College of Medicine, Tallahassee, FL, USA
| | - Leslie E Kolb
- Association of Diabetes Care & Education Specialists, Chicago, IL, USA
| | | | - Karin Lange
- Medical Psychology, Hannover Medical School, Hannover, Germany
| | | | - Åke Lernmark
- Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
| | - Ingrid Libman
- Division of Pediatric Endocrinology and Diabetes, University of Pittsburgh, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Markus Lundgren
- Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Department of Pediatrics, Kristianstad Hospital, Kristianstad, Sweden
| | - David M Maahs
- Division of Endocrinology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - M Loredana Marcovecchio
- Department of Pediatrics, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Chantal Mathieu
- Department of Endocrinology, UZ Gasthuisberg, KU Leuven, Leuven, Belgium
| | | | - Holly K O'Donnell
- Department of Pediatrics, Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Tal Oron
- Institute for Endocrinology and Diabetes, National Center for Childhood Diabetes, Schneider Children's Medical Center of Israel, Petah Tikva, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Shivajirao P Patil
- Department of Family Medicine, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - Rodica Pop-Busui
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, MI, USA
| | - Marian J Rewers
- Department of Pediatrics, Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Desmond A Schatz
- Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - Rifka Schulman-Rosenbaum
- Division of Endocrinology, Long Island Jewish Medical Center, Northwell Health, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, New Hyde Park, NY, USA
| | - Kimber M Simmons
- Department of Pediatrics, Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Emily K Sims
- Division of Pediatric Endocrinology and Diabetology, Herman B Wells Center for Pediatric Research, Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jay S Skyler
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Laura B Smith
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Cate Speake
- Center for Interventional Immunology and Diabetes Program, Benaroya Research Institute, Seattle, WA, USA
| | - Andrea K Steck
- Department of Pediatrics, Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | | | - Ksenia N Tonyushkina
- Division of Endocrinology and Diabetes, Baystate Children's Hospital and University of Massachusetts Chan Medical School - Baystate, Springfield, MA, USA
| | - Riitta Veijola
- Research Unit of Clinical Medicine, Department of Pediatrics, Medical Research Center, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - John M Wentworth
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Diabetes and Endocrinology, Royal Melbourne Hospital, Parkville, VIC, Australia
| | - Diane K Wherrett
- Department of Paediatrics, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Jamie R Wood
- Department of Pediatric Endocrinology, Rainbow Babies and Children's Hospital, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Anette-Gabriele Ziegler
- Institute of Diabetes Research, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany
- Forschergruppe Diabetes, Technical University Munich, Klinikum Rechts Der Isar, Munich, Germany
| | - Linda A DiMeglio
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
18
|
Strati M, Moustaki M, Psaltopoulou T, Vryonidou A, Paschou SA. Early onset type 2 diabetes mellitus: an update. Endocrine 2024; 85:965-978. [PMID: 38472622 DOI: 10.1007/s12020-024-03772-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 03/02/2024] [Indexed: 03/14/2024]
Abstract
The incidence and prevalence of type 2 diabetes mellitus (T2DM) in young individuals (aged <40 years) have significantly increased in recent years, approximating two to threefold increase in the respective rates. Numerous risk factors including severe obesity, family history, ethnicity, maternal diabetes or gestational diabetes, and female sex contribute to a younger age of onset. In terms of pathogenesis, impaired insulin secretion is the key operating mechanism, alongside with ectopic adiposity-related insulin resistance. T2DM diagnosis in a young adult requires the exclusion of type 1 diabetes mellitus (T1DM), latent autoimmune diabetes of adults (LADA) and maturity-onset diabetes of the young (MODY). The establishment of such diagnosis is critical for prognosis, because early-onset T2DM is associated with rapid deterioration in pancreatic β-cell secretory function leading to earlier initiation of insulin therapy. Furthermore, mortality and lifetime risk of developing complications, especially microvascular, is increased in these patients compared to both later-onset T2DM and T1DM patients; also, the latter are often developed earlier in the course of disease. The management of early-onset T2DM follows the same guidelines as in later-onset T2DM; yet patients aged 18-39 years are underrepresented in the big clinical trials on which the development of guidelines is based. Finally, young people with T2DM face significant challenges associated with social determinants, which compromise their adherence to therapy and induce diabetes distress. Future research focusing on the pathogenesis of β-cell decline and complications, as well as on specific treatment shall lead to better understanding and management of early-onset T2DM.
Collapse
Affiliation(s)
- Myrsini Strati
- School of Medicine, University of Patras, Patras, Greece
| | - Melpomeni Moustaki
- Department of Endocrinology and Diabetes Center, Hellenic Red Cross Hospital, Athens, Greece
| | - Theodora Psaltopoulou
- Endocrine Unit and Diabetes Center, Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Andromachi Vryonidou
- Department of Endocrinology and Diabetes Center, Hellenic Red Cross Hospital, Athens, Greece
| | - Stavroula A Paschou
- Endocrine Unit and Diabetes Center, Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
19
|
Spilseth B, Fogel EL, Toledo FG, Campbell-Thompson M. Imaging abnormalities of the pancreas in diabetes: implications for diagnosis and treatment. Curr Opin Gastroenterol 2024; 40:381-388. [PMID: 38967933 PMCID: PMC11305921 DOI: 10.1097/mog.0000000000001054] [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] [Indexed: 07/06/2024]
Abstract
PURPOSE OF REVIEW Radiographic imaging of the pancreas has drawn recent interest as pancreas volume may serve as a biomarker in identifying the likelihood of diabetes development, subtyping diabetes, and identifying prognostic indicators of poor ultimate outcomes. In this review, the role of pancreas imaging is discussed in various forms of diabetes including type 1 diabetes (T1D), type 2 diabetes (T2D), and diabetes of the exocrine pancreas, particularly diabetes following acute or chronic pancreatitis. RECENT FINDINGS Recent literature of quantitative pancreatic imaging correlating with various forms of diabetes was reviewed. Imaging-derived pancreas volumes are lower in individuals with diabetes, in particular those with T1D. Additionally, morphologic changes, enhancement characteristics, fat content, and MRI signal changes have been observed in different diabetes subtypes. These characteristics, as well as potential confounding variables, are reviewed. Additionally, future areas of research in MRI, CT radiomics, and pancreatitis-related imaging predictors of diabetes are discussed. SUMMARY Increased understanding of pancreas imaging features which predict diabetes and gauge prognosis has the potential to identify at-risk individuals and will become increasingly important in diabetes care. This article reviews the current knowledge of common pancreas imaging features as well as future directions of ongoing research in diabetes imaging.
Collapse
Affiliation(s)
| | - Evan L Fogel
- Digestive and Liver Disorders, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
| | | | - Martha Campbell-Thompson
- Department of Pathology immunology and Laboratory Medicine, University of Florida College of Medicine
| |
Collapse
|
20
|
Urrutia I, Martinez R, Calvo B, Marcelo I, Saso-Jimenez L, Martinez de Lapiscina I, Bilbao JR, Castano L, Rica I. Risk for progression to type 1 diabetes in first-degree relatives under 50 years of age. Front Endocrinol (Lausanne) 2024; 15:1411686. [PMID: 39188918 PMCID: PMC11345149 DOI: 10.3389/fendo.2024.1411686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 07/24/2024] [Indexed: 08/28/2024] Open
Abstract
Introduction The detection of pancreatic autoantibodies in first-degree relatives of patients with type 1 diabetes (T1D) is considered a risk factor for disease. Novel available immunotherapies to delay T1D progression highlight the importance of identifying individuals at risk who might benefit from emerging treatments. The objective was to assess the autoimmunity in first-degree relatives of patients with T1D, estimate the time from autoimmunity detection to the onset of clinical diabetes, and identify the associated risk factors. Methods Retrospective multicenter study of 3,015 first-degree relatives of patients with T1D recruited between 1992 and 2018. Pancreatic autoantibodies (IAA, GADA, IA2A, and ZnT8A) were determined by radioimmunoassay, starting the analyses at diagnosis of the proband. All those with positive autoimmunity and normal fasting blood glucose without clinical symptoms of diabetes were followed up in the study. The progression rate to T1D was assessed according to sex, relationship with the proband, age at autoimmunity detection, type/number of autoantibodies, and HLA-DRB1 genotype. Cox proportional-hazard models and Kaplan-Meier survival plots were used for statistical analyses. Results Among the relatives, 21 progenitors [43.7 years (IQR: 38.1-47.7)] and 27 siblings [7.6 years (IQR: 5.8-16.1)] had positive autoantibodies. Of these, 54.2% (95% CI: 39.2%-68.6%) developed T1D (age at autoimmunity detection 11 months to 39 years) in a median of 5 years (IQR: 3.6-8.7; ranged from 0.9 to 22.6 years). Risk factors associated with faster progression to T1D were multiple autoimmunity and <20 years at autoimmunity detection. Younger relatives (<20 years) with multiple autoantibodies had a 5-year cumulative risk of developing diabetes of 52.9% (95% CI: 22.1%-71.6%) and a 20-year risk of 91.2% (95% CI: 50.5%-98.4%). The 20-year risk decreased to 59.9% (95% CI: 21.9%-79.5%) if only one risk factor was met and to 35.7% (95% CI: 0.0%-66.2%) if the relative was older than 20 years with one autoantibody. Conclusions In first-degree relatives with autoimmunity, the time to progression to T1D is faster in children and adolescents with multiple autoantibodies. Young adults are also at risk, which supports their consideration in screening strategies for people at risk of developing T1D.
Collapse
Affiliation(s)
- Ines Urrutia
- Biobizkaia Health Research Institute, Barakaldo, Spain
- UPV/EHU, CIBERDEM, CIBERER, Endo-ERN, Barakaldo, Spain
| | - Rosa Martinez
- Biobizkaia Health Research Institute, Barakaldo, Spain
- UPV/EHU, CIBERDEM, CIBERER, Endo-ERN, Barakaldo, Spain
| | - Begona Calvo
- Biobizkaia Health Research Institute, Barakaldo, Spain
- Department of Medical Oncology, Cruces University Hospital, Barakaldo, Spain
| | - Irene Marcelo
- Hospital de Mataró - Consorci Sanitari del Maresme, Barcelona, Spain
| | - Laura Saso-Jimenez
- Biobizkaia Health Research Institute, Barakaldo, Spain
- UPV/EHU, CIBERDEM, CIBERER, Endo-ERN, Barakaldo, Spain
| | - Idoia Martinez de Lapiscina
- Biobizkaia Health Research Institute, Barakaldo, Spain
- UPV/EHU, CIBERDEM, CIBERER, Endo-ERN, Barakaldo, Spain
| | - Jose Ramon Bilbao
- Biobizkaia Health Research Institute, Barakaldo, Spain
- UPV/EHU, CIBERDEM, CIBERER, Endo-ERN, Barakaldo, Spain
| | - Luis Castano
- Biobizkaia Health Research Institute, Barakaldo, Spain
- UPV/EHU, CIBERDEM, CIBERER, Endo-ERN, Barakaldo, Spain
| | - Itxaso Rica
- Biobizkaia Health Research Institute, Barakaldo, Spain
- UPV/EHU, CIBERDEM, CIBERER, Endo-ERN, Barakaldo, Spain
- Pediatric Endocrinology Unit, Cruces University Hospital, Barakaldo, Spain
| |
Collapse
|
21
|
Wang N, Wang G, Feng X, Yang T. Identification of Immune Gene Signature Associated with T Cells and Natural Killer Cells in Type 1 Diabetes. Diabetes Metab Syndr Obes 2024; 17:2983-2996. [PMID: 39139741 PMCID: PMC11321349 DOI: 10.2147/dmso.s470738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 07/23/2024] [Indexed: 08/15/2024] Open
Abstract
Purpose This study aimed to investigate the abnormal infiltration of immune cells in type 1 diabetes mellitus (T1D) and elucidate their regulatory mechanisms. Methods Public T1D-related gene expression data were obtained from the Gene Expression Omnibus database.The GSE123658 dataset analyzed whole blood RNA-seq data from type 1 diabetic patients and healthy volunteers. The GSE110914 dataset analyzed neutrophils purified from peripheral blood of patients with symptomatic and pre-symptomatic type 1 diabetes (T1D), at risk of T1D, and healthy controls. Immune cell infiltration analysis was performed to identify abnormally infiltrating immune cells. Differentially expressed immune genes (DEIGs) in T1D samples were identified, followed by the construction of an immune gene signature (IGS) using a protein-protein interaction (PPI) network and Least absolute shrinkage and selection operator Cox regression analyses (LASSO Cox regression analyses). The regulatory mechanisms underlying IGS were explored using gene set enrichment analysis. Furthermore, expression validation, diagnostic efficacy evaluation, and upstream miRNA prediction of hub signature genes were performed. We verified the miRNA expression of the key gene colony stimulating factor 1 (CSF1) and microRNA-326 (miR-326) by reverse transcription-quantitative PCR (RT‒qPCR). Results The proportion of infiltrating T and natural killer (NK) cells differed between the T1D and control samples, and 207 immune genes (IGs) related to these immune cells were extracted. After differential expression, PPI, and LASSO Cox regression analyses, four signature DEIGs were identified for IGS construction: notch receptor 1 (NOTCH1), Janus kinase 3 (JAK3), tumor necrosis factor receptor superfamily member 4(TNFRSF4), and CSF1. Key pathways such as the Toll-like receptor signaling pathway were significantly activated in the high-risk group. Moreover, the upregulation of CSF1 in T1D samples was confirmed using a validation dataset, and CSF1 showed high diagnostic efficacy for T1D. Furthermore, CSF1 was targeted by miR-326.We used validated key genes in T1D patients, several of which were confirmed by RT‒qPCR. Conclusion In conclusion, the identified key IGs may play an important role in T1D. CSF1 can be developed as a novel diagnostic biomarker for T1D.
Collapse
Affiliation(s)
- Na Wang
- Department of Endocrinology, Lianyungang Clinical College of Nanjing Medical University, Lianyungang City, Jiangsu Province, 222000, People’s Republic of China
- Department of Endocrinology, Jinzhou Medical University(The First People’s Hospital of Lianyungang), Lianyungang City, Jiangsu Province, 222000, People’s Republic of China
| | - Guofeng Wang
- Department of Endocrinology, Jinzhou Medical University(The First People’s Hospital of Lianyungang), Lianyungang City, Jiangsu Province, 222000, People’s Republic of China
| | - Xiuli Feng
- Department of Endocrinology, Jinzhou Medical University(The First People’s Hospital of Lianyungang), Lianyungang City, Jiangsu Province, 222000, People’s Republic of China
| | - Teng Yang
- Department of Endocrinology, Jinzhou Medical University(The First People’s Hospital of Lianyungang), Lianyungang City, Jiangsu Province, 222000, People’s Republic of China
| |
Collapse
|
22
|
Catassi C, Vincentini O, Pricci F, Agrimi U, Silano M, Bosi E. Pediatric screening for type 1 diabetes and celiac disease: the future is today in Italy. Minerva Pediatr (Torino) 2024; 76:461-463. [PMID: 38602015 DOI: 10.23736/s2724-5276.24.07573-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
Affiliation(s)
- Carlo Catassi
- Department of Pediatrics, Polytechnic University of Marche, Ancona, Italy -
| | - Olimpia Vincentini
- Department of Food Safety, Nutrition and Veterinary Public Health, Istituto Superiore di Sanità, Rome, Italy
| | - Flavia Pricci
- Department of Cardiovascular, Endocrine-Metabolic Diseases and Ageing, Istituto Superiore di Sanità, Rome, Italy
| | - Umberto Agrimi
- Department of Food Safety, Nutrition and Veterinary Public Health, Istituto Superiore di Sanità, Rome, Italy
| | - Marco Silano
- Department of Cardiovascular, Endocrine-Metabolic Diseases and Ageing, Istituto Superiore di Sanità, Rome, Italy
| | - Emanuele Bosi
- Internal Medicine and Diabetes Unit, Diabetes Research Institute, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
23
|
Phillip M, Achenbach P, Addala A, Albanese-O’Neill A, Battelino T, Bell KJ, Besser RE, Bonifacio E, Colhoun HM, Couper JJ, Craig ME, Danne T, de Beaufort C, Dovc K, Driscoll KA, Dutta S, Ebekozien O, Elding Larsson H, Feiten DJ, Frohnert BI, Gabbay RA, Gallagher MP, Greenbaum CJ, Griffin KJ, Hagopian W, Haller MJ, Hendrieckx C, Hendriks E, Holt RI, Hughes L, Ismail HM, Jacobsen LM, Johnson SB, Kolb LE, Kordonouri O, Lange K, Lash RW, Lernmark Å, Libman I, Lundgren M, Maahs DM, Marcovecchio ML, Mathieu C, Miller KM, O’Donnell HK, Oron T, Patil SP, Pop-Busui R, Rewers MJ, Rich SS, Schatz DA, Schulman-Rosenbaum R, Simmons KM, Sims EK, Skyler JS, Smith LB, Speake C, Steck AK, Thomas NP, Tonyushkina KN, Veijola R, Wentworth JM, Wherrett DK, Wood JR, Ziegler AG, DiMeglio LA. Consensus Guidance for Monitoring Individuals With Islet Autoantibody-Positive Pre-Stage 3 Type 1 Diabetes. Diabetes Care 2024; 47:1276-1298. [PMID: 38912694 PMCID: PMC11381572 DOI: 10.2337/dci24-0042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 05/09/2024] [Indexed: 06/25/2024]
Abstract
Given the proven benefits of screening to reduce diabetic ketoacidosis (DKA) likelihood at the time of stage 3 type 1 diabetes diagnosis, and emerging availability of therapy to delay disease progression, type 1 diabetes screening programs are being increasingly emphasized. Once broadly implemented, screening initiatives will identify significant numbers of islet autoantibody-positive (IAb+) children and adults who are at risk for (confirmed single IAb+) or living with (multiple IAb+) early-stage (stage 1 and stage 2) type 1 diabetes. These individuals will need monitoring for disease progression; much of this care will happen in nonspecialized settings. To inform this monitoring, JDRF, in conjunction with international experts and societies, developed consensus guidance. Broad advice from this guidance includes the following: 1) partnerships should be fostered between endocrinologists and primary care providers to care for people who are IAb+; 2) when people who are IAb+ are initially identified, there is a need for confirmation using a second sample; 3) single IAb+ individuals are at lower risk of progression than multiple IAb+ individuals; 4) individuals with early-stage type 1 diabetes should have periodic medical monitoring, including regular assessments of glucose levels, regular education about symptoms of diabetes and DKA, and psychosocial support; 5) interested people with stage 2 type 1 diabetes should be offered trial participation or approved therapies; and 6) all health professionals involved in monitoring and care of individuals with type 1 diabetes have a responsibility to provide education. The guidance also emphasizes significant unmet needs for further research on early-stage type 1 diabetes to increase the rigor of future recommendations and inform clinical care.
Collapse
Affiliation(s)
- Moshe Phillip
- Institute for Endocrinology and Diabetes, National Center for Childhood Diabetes, Schneider Children's Medical Center of Israel, Petah Tikva, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Peter Achenbach
- Institute of Diabetes Research, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany
- Forschergruppe Diabetes, Technical University Munich, Klinikum Rechts Der Isar, Munich, Germany
| | - Ananta Addala
- Division of Endocrinology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA
- Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA
| | | | - Tadej Battelino
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Department of Endocrinology, Diabetes and Metabolism, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Kirstine J. Bell
- Charles Perkins Centre and Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Rachel E.J. Besser
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Wellcome Centre Human Genetics, Nuffield Department of Medicine Oxford National Institute for Health and Care Research Biomedical Research Centre, University of Oxford, Oxford, U.K
- Department of Paediatrics, University of Oxford, Oxford, U.K
| | - Ezio Bonifacio
- Center for Regenerative Therapies Dresden, Faculty of Medicine, Technical University of Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden, Helmholtz Centre Munich at the University Clinic Carl Gustav Carus of Technical University of Dresden, and Faculty of Medicine, Technical University of Dresden, Dresden, Germany
| | - Helen M. Colhoun
- The Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, U.K
- Department of Public Health, NHS Fife, Kirkcaldy, U.K
| | - Jennifer J. Couper
- Robinson Research Institute, The University of Adelaide, Adelaide, South Australia, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
- Division of Paediatrics, Women’s and Children’s Hospital, Adelaide, South Australia, Australia
| | - Maria E. Craig
- Charles Perkins Centre and Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Discipline of Paediatrics & Child Health, School of Clinical Medicine, UNSW Medicine & Health, Sydney, New South Wales, Australia
| | | | - Carine de Beaufort
- International Society for Pediatric and Adolescent Diabetes (ISPAD), Berlin, Germany
- Diabetes & Endocrine Care Clinique Pédiatrique (DECCP), Clinique Pédiatrique/Centre Hospitalier (CH) de Luxembourg, Luxembourg City, Luxembourg
- Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-Belval, Luxembourg
| | - Klemen Dovc
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Department of Endocrinology, Diabetes and Metabolism, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Kimberly A. Driscoll
- Department of Pediatrics, Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO
- Department of Clinical and Health Psychology, University of Florida, Gainesville, FL
- Department of Pediatrics, University of Florida Diabetes Institute, Gainesville, FL
| | | | | | - Helena Elding Larsson
- Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Department of Pediatrics, Skåne University Hospital, Malmö and Lund, Sweden
| | | | - Brigitte I. Frohnert
- Department of Pediatrics, Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO
| | | | | | - Carla J. Greenbaum
- Center for Interventional Immunology and Diabetes Program, Benaroya Research Institute, Seattle, WA
| | - Kurt J. Griffin
- Sanford Research, Sioux Falls, SD
- Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD
| | - William Hagopian
- Pacific Northwest Diabetes Research Institute, University of Washington, Seattle, WA
| | - Michael J. Haller
- Department of Pediatrics, University of Florida Diabetes Institute, Gainesville, FL
- Division of Endocrinology, University of Florida College of Medicine, Gainesville, FL
| | - Christel Hendrieckx
- School of Psychology, Deakin University, Geelong, Victoria, Australia
- The Australian Centre for Behavioural Research in Diabetes, Diabetes Victoria, Carlton, Victoria, Australia
- Institute for Health Transformation, Deakin University, Geelong, Victoria, Australia
| | - Emile Hendriks
- Department of Paediatrics, University of Cambridge and Cambridge University Hospitals NHS Foundation Trust, Cambridge, U.K
| | - Richard I.G. Holt
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, U.K
- National Institute for Health and Care Research Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, U.K
| | | | - Heba M. Ismail
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN
| | - Laura M. Jacobsen
- Division of Endocrinology, University of Florida College of Medicine, Gainesville, FL
| | - Suzanne B. Johnson
- Department of Behavioral Sciences and Social Medicine, Florida State University College of Medicine, Tallahassee, FL
| | - Leslie E. Kolb
- Association of Diabetes Care & Education Specialists, Chicago, IL
| | | | - Karin Lange
- Medical Psychology, Hannover Medical School, Hannover, Germany
| | | | - Åke Lernmark
- Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
| | - Ingrid Libman
- Division of Pediatric Endocrinology and Diabetes, University of Pittsburgh, University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, PA
| | - Markus Lundgren
- Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Department of Pediatrics, Kristianstad Hospital, Kristianstad, Sweden
| | - David M. Maahs
- Division of Endocrinology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA
| | | | - Chantal Mathieu
- Department of Endocrinology, UZ Gasthuisberg, KU Leuven, Leuven, Belgium
| | | | - Holly K. O’Donnell
- Department of Pediatrics, Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Tal Oron
- Institute for Endocrinology and Diabetes, National Center for Childhood Diabetes, Schneider Children's Medical Center of Israel, Petah Tikva, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Shivajirao P. Patil
- Department of Family Medicine, Brody School of Medicine, East Carolina University, Greenville, NC
| | - Rodica Pop-Busui
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, MI
| | - Marian J. Rewers
- Department of Pediatrics, Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Stephen S. Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA
| | | | - Rifka Schulman-Rosenbaum
- Division of Endocrinology, Long Island Jewish Medical Center, Northwell Health, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, New Hyde Park, NY
| | - Kimber M. Simmons
- Department of Pediatrics, Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Emily K. Sims
- Division of Pediatric Endocrinology and Diabetology, Herman B Wells Center for Pediatric Research, Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN
| | - Jay S. Skyler
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL
| | - Laura B. Smith
- Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Cate Speake
- Center for Interventional Immunology and Diabetes Program, Benaroya Research Institute, Seattle, WA
| | - Andrea K. Steck
- Department of Pediatrics, Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Nicholas P.B. Thomas
- National Institute of Health and Care Research Clinical Research Network Thames Valley and South Midlands, Oxford, U.K
| | - Ksenia N. Tonyushkina
- Division of Endocrinology and Diabetes, Baystate Children’s Hospital and University of Massachusetts Chan Medical School–Baystate, Springfield, MA
| | - Riitta Veijola
- Research Unit of Clinical Medicine, Department of Pediatrics, Medical Research Center, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - John M. Wentworth
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Diabetes and Endocrinology, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Diane K. Wherrett
- Department of Paediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Jamie R. Wood
- Department of Pediatric Endocrinology, Rainbow Babies and Children's Hospital, University Hospitals Cleveland Medical Center, Cleveland, OH
| | - Anette-Gabriele Ziegler
- Institute of Diabetes Research, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany
- Forschergruppe Diabetes, Technical University Munich, Klinikum Rechts Der Isar, Munich, Germany
| | - Linda A. DiMeglio
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN
| |
Collapse
|
24
|
Abel ED, Gloyn AL, Evans-Molina C, Joseph JJ, Misra S, Pajvani UB, Simcox J, Susztak K, Drucker DJ. Diabetes mellitus-Progress and opportunities in the evolving epidemic. Cell 2024; 187:3789-3820. [PMID: 39059357 PMCID: PMC11299851 DOI: 10.1016/j.cell.2024.06.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 06/21/2024] [Accepted: 06/24/2024] [Indexed: 07/28/2024]
Abstract
Diabetes, a complex multisystem metabolic disorder characterized by hyperglycemia, leads to complications that reduce quality of life and increase mortality. Diabetes pathophysiology includes dysfunction of beta cells, adipose tissue, skeletal muscle, and liver. Type 1 diabetes (T1D) results from immune-mediated beta cell destruction. The more prevalent type 2 diabetes (T2D) is a heterogeneous disorder characterized by varying degrees of beta cell dysfunction in concert with insulin resistance. The strong association between obesity and T2D involves pathways regulated by the central nervous system governing food intake and energy expenditure, integrating inputs from peripheral organs and the environment. The risk of developing diabetes or its complications represents interactions between genetic susceptibility and environmental factors, including the availability of nutritious food and other social determinants of health. This perspective reviews recent advances in understanding the pathophysiology and treatment of diabetes and its complications, which could alter the course of this prevalent disorder.
Collapse
Affiliation(s)
- E Dale Abel
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA.
| | - Anna L Gloyn
- Department of Pediatrics, Division of Endocrinology & Diabetes, Department of Genetics, Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA, USA
| | - Carmella Evans-Molina
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Joshua J Joseph
- Division of Endocrinology, Diabetes and Metabolism, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Shivani Misra
- Department of Metabolism, Digestion and Reproduction, Imperial College London, and Imperial College NHS Trust, London, UK
| | - Utpal B Pajvani
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Judith Simcox
- Howard Hughes Medical Institute, Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Katalin Susztak
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Daniel J Drucker
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada; Department of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
25
|
Moore DJ, Leibel NI, Polonsky W, Rodriguez H. Recommendations for Screening and Monitoring the Stages of Type 1 Diabetes in the Immune Therapy Era. Int J Gen Med 2024; 17:3003-3014. [PMID: 39011423 PMCID: PMC11247126 DOI: 10.2147/ijgm.s438009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 06/13/2024] [Indexed: 07/17/2024] Open
Abstract
Type 1 diabetes (T1D) is a complex, chronic autoimmune disease that affects over 1.6 million people in the United States. It is now understood that T1D may be undetected for many years while the disease progresses quietly without producing symptoms. T1D can be identified through diabetes-related autoantibody screening and staged accordingly, enabling healthcare providers to identify high-risk individuals in the early stages of the disease and either provide a stage-specific intervention or offer clinical trial opportunities to preserve beta cell function and anticipate the onset of clinical T1D. Evidence-based clinical practice guidelines currently do not exist for routine diabetes-related autoantibody screening of individuals at risk of developing T1D or of the general population. The purpose of this article is to help clinicians acquire an understanding of the rationale and protocols recommended for identifying patients at risk of developing T1D and monitoring such patients for autoimmune markers and progression of disease from Stage 1 to Stage 3 (clinical disease).
Collapse
Affiliation(s)
- Daniel J Moore
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Natasha I Leibel
- Department of Pediatrics, Columbia University, New York, NY, USA
| | | | - Henry Rodriguez
- USF Diabetes and Endocrinology Center, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| |
Collapse
|
26
|
Joglekar MV, Kaur S, Pociot F, Hardikar AA. Prediction of progression to type 1 diabetes with dynamic biomarkers and risk scores. Lancet Diabetes Endocrinol 2024; 12:483-492. [PMID: 38797187 DOI: 10.1016/s2213-8587(24)00103-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 03/31/2024] [Accepted: 04/02/2024] [Indexed: 05/29/2024]
Abstract
Identifying biomarkers of functional β-cell loss is an important step in the risk stratification of type 1 diabetes. Genetic risk scores (GRS), generated by profiling an array of single nucleotide polymorphisms, are a widely used type 1 diabetes risk-prediction tool. Type 1 diabetes screening studies have relied on a combination of biochemical (autoantibody) and GRS screening methodologies for identifying individuals at high-risk of type 1 diabetes. A limitation of these screening tools is that the presence of autoantibodies marks the initiation of β-cell loss, and is therefore not the best biomarker of progression to early-stage type 1 diabetes. GRS, on the other hand, represents a static biomarker offering a single risk score over an individual's lifetime. In this Personal View, we explore the challenges and opportunities of static and dynamic biomarkers in the prediction of progression to type 1 diabetes. We discuss future directions wherein newer dynamic risk scores could be used to predict type 1 diabetes risk, assess the efficacy of new and emerging drugs to retard, or prevent type 1 diabetes, and possibly replace or further enhance the predictive ability offered by static biomarkers, such as GRS.
Collapse
Affiliation(s)
- Mugdha V Joglekar
- School of Medicine, Western Sydney University, Sydney, NSW, Australia
| | | | - Flemming Pociot
- Steno Diabetes Center Copenhagen, Herlev, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark.
| | | |
Collapse
|
27
|
Long SA, Linsley PS. Integrating Omics into Functional Biomarkers of Type 1 Diabetes. Cold Spring Harb Perspect Med 2024; 14:a041602. [PMID: 38772709 PMCID: PMC11216170 DOI: 10.1101/cshperspect.a041602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2024]
Abstract
Biomarkers are critical to the staging and diagnosis of type 1 diabetes (T1D). Functional biomarkers offer insights into T1D immunopathogenesis and are often revealed using "omics" approaches that integrate multiple measures to identify involved pathways and functions. Application of the omics biomarker discovery may enable personalized medicine approaches to circumvent the more recently appreciated heterogeneity of T1D progression and treatment. Use of omics to define functional biomarkers is still in its early years, yet findings to date emphasize the role of cytokine signaling and adaptive immunity in biomarkers of progression and response to therapy. Here, we share examples of the use of omics to define functional biomarkers focusing on two signatures, T-cell exhaustion and T-cell help, which have been associated with outcomes in both the natural history and treatment contexts.
Collapse
Affiliation(s)
- S Alice Long
- Center for Translational Immunology, Benaroya Research Institute, Seattle, Washington 98101, USA
| | - Peter S Linsley
- Center for Systems Immunology, Benaroya Research Institute, Seattle, Washington 98101, USA
| |
Collapse
|
28
|
Muralidharan C, Huang F, Enriquez JR, Wang JE, Nelson JB, Nargis T, May SC, Chakraborty A, Figatner KT, Navitskaya S, Anderson CM, Calvo V, Surguladze D, Mulvihill MJ, Yi X, Sarkar S, Oakes SA, Webb-Robertson BJM, Sims EK, Staschke KA, Eizirik DL, Nakayasu ES, Stokes ME, Tersey SA, Mirmira RG. Inhibition of the eukaryotic initiation factor-2α kinase PERK decreases risk of autoimmune diabetes in mice. J Clin Invest 2024; 134:e176136. [PMID: 38889047 PMCID: PMC11324307 DOI: 10.1172/jci176136] [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: 10/11/2023] [Accepted: 06/13/2024] [Indexed: 06/20/2024] Open
Abstract
Preventing the onset of autoimmune type 1 diabetes (T1D) is feasible through pharmacological interventions that target molecular stress-responsive mechanisms. Cellular stresses, such as nutrient deficiency, viral infection, or unfolded proteins, trigger the integrated stress response (ISR), which curtails protein synthesis by phosphorylating eukaryotic translation initiation factor-2α (eIF2α). In T1D, maladaptive unfolded protein response (UPR) in insulin-producing β cells renders these cells susceptible to autoimmunity. We found that inhibition of the eIF2α kinase PKR-like ER kinase (PERK), a common component of the UPR and ISR, reversed the mRNA translation block in stressed human islets and delayed the onset of diabetes, reduced islet inflammation, and preserved β cell mass in T1D-susceptible mice. Single-cell RNA-Seq of islets from PERK-inhibited mice showed reductions in the UPR and PERK signaling pathways and alterations in antigen-processing and presentation pathways in β cells. Spatial proteomics of islets from these mice showed an increase in the immune checkpoint protein programmed death-ligand 1 (PD-L1) in β cells. Golgi membrane protein 1, whose levels increased following PERK inhibition in human islets and EndoC-βH1 human β cells, interacted with and stabilized PD-L1. Collectively, our studies show that PERK activity enhances β cell immunogenicity and that inhibition of PERK may offer a strategy for preventing or delaying the development of T1D.
Collapse
Affiliation(s)
- Charanya Muralidharan
- Department of Medicine and the Kovler Diabetes Center, The University of Chicago, Chicago, Illinois, USA
| | - Fei Huang
- Department of Medicine and the Kovler Diabetes Center, The University of Chicago, Chicago, Illinois, USA
| | - Jacob R. Enriquez
- Department of Medicine and the Kovler Diabetes Center, The University of Chicago, Chicago, Illinois, USA
| | - Jiayi E. Wang
- Department of Medicine and the Kovler Diabetes Center, The University of Chicago, Chicago, Illinois, USA
| | - Jennifer B. Nelson
- Department of Medicine and the Kovler Diabetes Center, The University of Chicago, Chicago, Illinois, USA
| | - Titli Nargis
- Department of Medicine and the Kovler Diabetes Center, The University of Chicago, Chicago, Illinois, USA
| | - Sarah C. May
- Department of Medicine and the Kovler Diabetes Center, The University of Chicago, Chicago, Illinois, USA
| | - Advaita Chakraborty
- Department of Medicine and the Kovler Diabetes Center, The University of Chicago, Chicago, Illinois, USA
| | - Kayla T. Figatner
- Department of Medicine and the Kovler Diabetes Center, The University of Chicago, Chicago, Illinois, USA
| | - Svetlana Navitskaya
- Department of Medicine and the Kovler Diabetes Center, The University of Chicago, Chicago, Illinois, USA
| | - Cara M. Anderson
- Department of Medicine and the Kovler Diabetes Center, The University of Chicago, Chicago, Illinois, USA
| | | | | | | | - Xiaoyan Yi
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
| | - Soumyadeep Sarkar
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Scott A. Oakes
- Department of Pathology, The University of Chicago, Chicago, Illinois, USA
| | | | - Emily K. Sims
- Department of Pediatrics, Center for Diabetes and Metabolic Diseases, Wells Center for Pediatric Research, and
| | - Kirk A. Staschke
- Department of Biochemistry and Molecular Biology and the Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Decio L. Eizirik
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
| | - Ernesto S. Nakayasu
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington, USA
| | | | - Sarah A. Tersey
- Department of Medicine and the Kovler Diabetes Center, The University of Chicago, Chicago, Illinois, USA
| | - Raghavendra G. Mirmira
- Department of Medicine and the Kovler Diabetes Center, The University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
29
|
Muralidharan C, Huang F, Enriquez JR, Wang JE, Nelson JB, Nargis T, May SC, Chakraborty A, Figatner KT, Navitskaya S, Anderson CM, Calvo V, Surguladze D, Mulvihill MJ, Yi X, Sarkar S, Oakes SA, Webb-Robertson BJM, Sims EK, Staschke KA, Eizirik DL, Nakayasu ES, Stokes ME, Tersey SA, Mirmira RG. Inhibition of the Eukaryotic Initiation Factor-2-α Kinase PERK Decreases Risk of Autoimmune Diabetes in Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.06.561126. [PMID: 38895427 PMCID: PMC11185543 DOI: 10.1101/2023.10.06.561126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Preventing the onset of autoimmune type 1 diabetes (T1D) is feasible through pharmacological interventions that target molecular stress-responsive mechanisms. Cellular stresses, such as nutrient deficiency, viral infection, or unfolded proteins, trigger the integrated stress response (ISR), which curtails protein synthesis by phosphorylating eIF2α. In T1D, maladaptive unfolded protein response (UPR) in insulin-producing β cells renders these cells susceptible to autoimmunity. We show that inhibition of the eIF2α kinase PERK, a common component of the UPR and ISR, reverses the mRNA translation block in stressed human islets and delays the onset of diabetes, reduces islet inflammation, and preserves β cell mass in T1D-susceptible mice. Single-cell RNA sequencing of islets from PERK-inhibited mice shows reductions in the UPR and PERK signaling pathways and alterations in antigen processing and presentation pathways in β cells. Spatial proteomics of islets from these mice shows an increase in the immune checkpoint protein PD-L1 in β cells. Golgi membrane protein 1, whose levels increase following PERK inhibition in human islets and EndoC-βH1 human β cells, interacts with and stabilizes PD-L1. Collectively, our studies show that PERK activity enhances β cell immunogenicity, and inhibition of PERK may offer a strategy to prevent or delay the development of T1D.
Collapse
Affiliation(s)
- Charanya Muralidharan
- Department of Medicine and the Kovler Diabetes Center, The University of Chicago, Chicago, IL, USA
| | - Fei Huang
- Department of Medicine and the Kovler Diabetes Center, The University of Chicago, Chicago, IL, USA
| | - Jacob R. Enriquez
- Department of Medicine and the Kovler Diabetes Center, The University of Chicago, Chicago, IL, USA
| | - Jiayi E. Wang
- Department of Medicine and the Kovler Diabetes Center, The University of Chicago, Chicago, IL, USA
| | - Jennifer B. Nelson
- Department of Medicine and the Kovler Diabetes Center, The University of Chicago, Chicago, IL, USA
| | - Titli Nargis
- Department of Medicine and the Kovler Diabetes Center, The University of Chicago, Chicago, IL, USA
| | - Sarah C. May
- Department of Medicine and the Kovler Diabetes Center, The University of Chicago, Chicago, IL, USA
| | - Advaita Chakraborty
- Department of Medicine and the Kovler Diabetes Center, The University of Chicago, Chicago, IL, USA
| | - Kayla T. Figatner
- Department of Medicine and the Kovler Diabetes Center, The University of Chicago, Chicago, IL, USA
| | - Svetlana Navitskaya
- Department of Medicine and the Kovler Diabetes Center, The University of Chicago, Chicago, IL, USA
| | - Cara M. Anderson
- Department of Medicine and the Kovler Diabetes Center, The University of Chicago, Chicago, IL, USA
| | | | | | | | - Xiaoyan Yi
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
| | - Soumyadeep Sarkar
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Scott A. Oakes
- Department of Pathology, The University of Chicago, Chicago, IL, USA
| | | | - Emily K. Sims
- Department of Pediatrics, Center for Diabetes and Metabolic Diseases, and the Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN
| | - Kirk A Staschke
- Department of Biochemistry and Molecular Biology and the Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Decio L. Eizirik
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
| | - Ernesto S. Nakayasu
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | | | - Sarah A. Tersey
- Department of Medicine and the Kovler Diabetes Center, The University of Chicago, Chicago, IL, USA
| | - Raghavendra G. Mirmira
- Department of Medicine and the Kovler Diabetes Center, The University of Chicago, Chicago, IL, USA
| |
Collapse
|
30
|
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
|
31
|
Neuman V, Piona C, Cudizio L, Drnkova L, Sumnik Z, Slavenko MG, Haller MJ, Maahs DM, Chobot A. Are we ready to screen for type 1 diabetes? A structured worldwide survey among healthcare providers involved in paediatric diabetes care. Diabet Med 2024; 41:e15329. [PMID: 38566408 DOI: 10.1111/dme.15329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 03/25/2024] [Accepted: 03/27/2024] [Indexed: 04/04/2024]
Affiliation(s)
- Vit Neuman
- Department of Pediatrics, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - Claudia Piona
- Pediatric Diabetes and Metabolic Disorders Unit, Regional Center for Pediatric Diabetes, University Hospital Verona, Verona, Italy
| | - Laura Cudizio
- Pediatric Endocrine Unit, Department of Pediatrics, Santa Casa de Sao Paulo, Sao Paulo, SP, Brazil
| | - Lenka Drnkova
- Department of Pediatrics, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - Zdenek Sumnik
- Department of Pediatrics, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - Matvei G Slavenko
- Department of Pediatrics, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - Michael J Haller
- Department of Pediatrics, University of Florida, Gainesville, Florida, USA
| | - David M Maahs
- Division of Pediatric Endocrinology, Stanford University, Stanford, California, USA
- Stanford Diabetes Research Center, Stanford University, Stanford, California, USA
| | - Agata Chobot
- Department of Pediatrics, Institute of Medical Sciences, University of Opole, Opole, Poland
| |
Collapse
|
32
|
Marcovecchio ML, Hendriks AEJ, Delfin C, Battelino T, Danne T, Evans ML, Johannesen J, Kaur S, Knip M, Overbergh L, Pociot F, Todd JA, Van der Schueren B, Wicker LS, Peakman M, Mathieu C. The INNODIA Type 1 Diabetes Natural History Study: a European cohort of newly diagnosed children, adolescents and adults. Diabetologia 2024; 67:995-1008. [PMID: 38517484 PMCID: PMC11058619 DOI: 10.1007/s00125-024-06124-5] [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: 08/21/2023] [Accepted: 01/24/2024] [Indexed: 03/24/2024]
Abstract
AIMS/HYPOTHESIS Type 1 diabetes is an heterogenous condition. Characterising factors explaining differences in an individual's clinical course and treatment response will have important clinical and research implications. Our aim was to explore type 1 diabetes heterogeneity, as assessed by clinical characteristics, autoantibodies, beta cell function and glycaemic outcomes, during the first 12 months from diagnosis, and how it relates to age at diagnosis. METHODS Data were collected from the large INNODIA cohort of individuals (aged 1.0-45.0 years) newly diagnosed with type 1 diabetes, followed 3 monthly, to assess clinical characteristics, C-peptide, HbA1c and diabetes-associated antibodies, and their changes, during the first 12 months from diagnosis, across three age groups: <10 years; 10-17 years; and ≥18 years. RESULTS The study population included 649 individuals (57.3% male; age 12.1±8.3 years), 96.9% of whom were positive for one or more diabetes-related antibodies. Baseline (IQR) fasting C-peptide was 242.0 (139.0-382.0) pmol/l (AUC 749.3 [466.2-1106.1] pmol/l × min), with levels increasing with age (p<0.001). Over time, C-peptide remained lower in participants aged <10 years but it declined in all age groups. In parallel, glucose levels progressively increased. Lower baseline fasting C-peptide, BMI SD score and presence of diabetic ketoacidosis at diagnosis were associated with lower stimulated C-peptide over time. HbA1c decreased during the first 3 months (p<0.001), whereas insulin requirement increased from 3 months post diagnosis (p<0.001). CONCLUSIONS/INTERPRETATION In this large cohort with newly diagnosed type 1 diabetes, we identified age-related differences in clinical and biochemical variables. Of note, C-peptide was lower in younger children but there were no main age differences in its rate of decline.
Collapse
Affiliation(s)
- M Loredana Marcovecchio
- Department of Paediatrics, University of Cambridge, Cambridge, UK.
- Department of Paediatric Diabetes and Endocrinology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.
| | - A Emile J Hendriks
- Department of Paediatrics, University of Cambridge, Cambridge, UK
- Department of Paediatric Diabetes and Endocrinology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Carl Delfin
- Department of Pharmacometrics, Novo Nordisk A/S, Søborg, Denmark
| | - Tadej Battelino
- Department of Endocrinology, Diabetes and Metabolism, University Children's Hospital, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Thomas Danne
- Centre for Paediatric Endocrinology, Diabetology, and Clinical Research, Auf Der Bult Children's Hospital, Hannover, Germany
| | - Mark L Evans
- Wellcome MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Jesper Johannesen
- Translational Type 1 Diabetes Research, Clinical Research, Steno Diabetes Center Copenhagen, Herlev, Denmark
- Department of Paediatrics, Copenhagen University Hospital, Herlev, Denmark; Institute of Health and Medical Sciences, University of Copenhagen, Herlev, Denmark
| | - Simranjeet Kaur
- Translational Type 1 Diabetes Research, Clinical Research, Steno Diabetes Center Copenhagen, Herlev, Denmark
- Department of Paediatrics, Copenhagen University Hospital, Herlev, Denmark; Institute of Health and Medical Sciences, University of Copenhagen, Herlev, Denmark
| | - Mikael Knip
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Pediatric Research Center, New Children's Hospital, Helsinki University Hospital, Helsinki, Finland
| | - Lut Overbergh
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Flemming Pociot
- Translational Type 1 Diabetes Research, Clinical Research, Steno Diabetes Center Copenhagen, Herlev, Denmark
- Department of Paediatrics, Copenhagen University Hospital, Herlev, Denmark; Institute of Health and Medical Sciences, University of Copenhagen, Herlev, Denmark
| | - John A Todd
- Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Bart Van der Schueren
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Linda S Wicker
- Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Mark Peakman
- Immunology & Inflammation Research Therapeutic Area, Sanofi, MA, USA
| | - Chantal Mathieu
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| |
Collapse
|
33
|
Teixeira PF, Battelino T, Carlsson A, Gudbjörnsdottir S, Hannelius U, von Herrath M, Knip M, Korsgren O, Elding Larsson H, Lindqvist A, Ludvigsson J, Lundgren M, Nowak C, Pettersson P, Pociot F, Sundberg F, Åkesson K, Lernmark Å, Forsander G. Assisting the implementation of screening for type 1 diabetes by using artificial intelligence on publicly available data. Diabetologia 2024; 67:985-994. [PMID: 38353727 PMCID: PMC11058797 DOI: 10.1007/s00125-024-06089-5] [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: 04/26/2023] [Accepted: 12/06/2023] [Indexed: 04/30/2024]
Abstract
The type 1 diabetes community is coalescing around the benefits and advantages of early screening for disease risk. To be accepted by healthcare providers, regulatory authorities and payers, screening programmes need to show that the testing variables allow accurate risk prediction and that individualised risk-informed monitoring plans are established, as well as operational feasibility, cost-effectiveness and acceptance at population level. Artificial intelligence (AI) has the potential to contribute to solving these issues, starting with the identification and stratification of at-risk individuals. ASSET (AI for Sustainable Prevention of Autoimmunity in the Society; www.asset.healthcare ) is a public/private consortium that was established to contribute to research around screening for type 1 diabetes and particularly to how AI can drive the implementation of a precision medicine approach to disease prevention. ASSET will additionally focus on issues pertaining to operational implementation of screening. The authors of this article, researchers and clinicians active in the field of type 1 diabetes, met in an open forum to independently debate key issues around screening for type 1 diabetes and to advise ASSET. The potential use of AI in the analysis of longitudinal data from observational cohort studies to inform the design of improved, more individualised screening programmes was also discussed. A key issue was whether AI would allow the research community and industry to capitalise on large publicly available data repositories to design screening programmes that allow the early detection of individuals at high risk and enable clinical evaluation of preventive therapies. Overall, AI has the potential to revolutionise type 1 diabetes screening, in particular to help identify individuals who are at increased risk of disease and aid in the design of appropriate follow-up plans. We hope that this initiative will stimulate further research on this very timely topic.
Collapse
Affiliation(s)
| | - Tadej Battelino
- University Medical Center Ljubljana, University of Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Anneli Carlsson
- Department of Clinical Sciences, Lund University/CRC, Skåne University Hospital, Malmö, Sweden
| | - Soffia Gudbjörnsdottir
- Swedish National Diabetes Register, Centre of Registers, Gothenburg, Sweden
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | | | - Matthias von Herrath
- Global Chief Medical Office, Novo Nordisk, A/S, Søborg, Denmark
- Diabetes Research Institute, University of Miami, Miami, FL, USA
| | - Mikael Knip
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Center for Child Health Research, Tampere University Hospital, Tampere, Finland
| | - Olle Korsgren
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- Department of Clinical Chemistry and Transfusion Medicine, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Helena Elding Larsson
- Department of Clinical Sciences, Lund University/CRC, Skåne University Hospital, Malmö, Sweden
- Department of Pediatrics, Skåne University Hospital, Malmö, Sweden
| | | | - Johnny Ludvigsson
- Crown Princess Victoria Children's Hospital and Division of Pediatrics, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Markus Lundgren
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
- Department of Paediatrics, Kristianstad Hospital, Kristianstad, Sweden
| | | | - Paul Pettersson
- Division of Networked and Embedded Systems, Mälardalen University, Västerås, Sweden
- MainlyAI AB, Stockholm, Sweden
| | - Flemming Pociot
- Steno Diabetes Center Copenhagen, Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Frida Sundberg
- Department of Paediatrics, Institute for Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Queen Silvia Children's Hospital, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Karin Åkesson
- Department of Clinical and Experimental Medicine, Division of Pediatrics and Diabetes Research Center, Linköping University, Linköping, Sweden
- Department of Pediatrics, Ryhov County Hospital, Jönköping, Sweden
| | - Åke Lernmark
- Department of Clinical Sciences, Lund University/CRC, Skåne University Hospital, Malmö, Sweden.
| | - Gun Forsander
- Department of Paediatrics, Institute for Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
- Queen Silvia Children's Hospital, Sahlgrenska University Hospital, Gothenburg, Sweden.
| |
Collapse
|
34
|
Villaécija J, Luque B, Cuadrado E, Vivas S, Tabernero C. Psychometric Properties of the Revised Self-Efficacy for Diabetes Self-Management Scale among Spanish Children and Adolescents with Type 1 Diabetes. CHILDREN (BASEL, SWITZERLAND) 2024; 11:662. [PMID: 38929241 PMCID: PMC11201513 DOI: 10.3390/children11060662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 05/24/2024] [Accepted: 05/26/2024] [Indexed: 06/28/2024]
Abstract
A longitudinal design was used to examine the psychometric properties of the Self-Efficacy for Diabetes Self-Management (SEDM) for children and adolescents with a diagnosis of type 1 diabetes (T1D). The SEDM was adapted to Spanish and the best factorial solution was selected to test the invariance of the measures of age and gender. Individuals between the ages of 10 and 19 years old with a diagnosis of T1D completed a self-reported questionnaire (167 at Time 1 [mean age = 14.49, SD = 2.76; 56.9% boys] and 122 at Time 2 [mean age = 14.77, SD = 2.58; 56.6% boys]). Two unifactorial solutions were tested. The psychometric properties of the scale were validated. The proposed validation obtained excellent reliability indices (χ2 (26) = 25.59, p > 0.49, RMSEA = 0.00, 95% CI [0.00, 0.07], CFI = 1.00, GFI = 0.96, AGFI = 0.92, TLI = 1.00, and CMIN = 0.98), and it appeared to be invariant for gender and for age groups. The Cronbach's α was 0.85. The test-retest reliability was high (r = 0.69 [p < 0.001]). Convergent, discriminant, and external validity were proven. The nine-item SEDM is a brief measure with satisfactory structural validity. From our knowledge, this study provides the first reliable tool to assess self-efficacy in the management of T1D for Spanish children and adolescents.
Collapse
Affiliation(s)
- Joaquín Villaécija
- Department of Psychology, University of Cordoba, 14071 Cordoba, Spain; (J.V.); (S.V.)
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Cordoba, Spain
- Reina Sofia University Hospital (HURS), 14004 Cordoba, Spain
| | - Bárbara Luque
- Department of Psychology, University of Cordoba, 14071 Cordoba, Spain; (J.V.); (S.V.)
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Cordoba, Spain
- Reina Sofia University Hospital (HURS), 14004 Cordoba, Spain
| | - Esther Cuadrado
- Department of Psychology, University of Cordoba, 14071 Cordoba, Spain; (J.V.); (S.V.)
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Cordoba, Spain
- Reina Sofia University Hospital (HURS), 14004 Cordoba, Spain
| | - Sebastián Vivas
- Department of Psychology, University of Cordoba, 14071 Cordoba, Spain; (J.V.); (S.V.)
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Cordoba, Spain
- Reina Sofia University Hospital (HURS), 14004 Cordoba, Spain
| | - Carmen Tabernero
- Department of Social Psychology and Anthropology, University of Salamanca, 37005 Salamanca, Spain;
- Instituto de Neurociencias de Castilla y León (INCYL), University of Salamanca, 37005 Salamanca, Spain
| |
Collapse
|
35
|
Huston J, Sudhakar D, Malarvannan P, Aradhyula R, Clark AL. Diabetes and Dyslipidemia Screening in Pediatric Practice. MISSOURI MEDICINE 2024; 121:206-211. [PMID: 38854609 PMCID: PMC11160381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
The incidence of diabetes and hyperlipidemia are increasing at rapid rates in children. These conditions are associated with increased risk of macrovascular and microvascular complications causing major morbidity and mortality later in life. Early diagnosis and treatment can reduce the lifelong risk of complications from these diseases, exemplifying the importance of screening in the pediatric population. The following article presents a summary of the current guidelines for diabetes and hyperlipidemia screening in pediatric patients.
Collapse
Affiliation(s)
- Jordyn Huston
- Department of Pediatrics, Saint Louis University School of Medicine, St. Louis, Missouri
| | - Deepti Sudhakar
- Saint Louis University School of Medicine, St. Louis, Missouri
| | | | | | - Amy L Clark
- Department of Pediatrics, Division of Endocrinology, Saint Louis University School of Medicine, SSM Health Cardinal Glennon, St. Louis, Missouri
| |
Collapse
|
36
|
Bloomgarden Z. Can type 1 diabetes be prevented or reversed? J Diabetes 2024; 16:e13572. [PMID: 38741346 PMCID: PMC11091426 DOI: 10.1111/1753-0407.13572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/01/2024] [Indexed: 05/16/2024] Open
Affiliation(s)
- Zachary Bloomgarden
- Department of Medicine, Division of Endocrinology, Diabetes and Bone DiseaseIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| |
Collapse
|
37
|
Tuo L, Yan LT, Liu Y, Yang XX. Type 1 diabetes mellitus and non-alcoholic fatty liver disease: a two-sample Mendelian randomization study. Front Endocrinol (Lausanne) 2024; 15:1315046. [PMID: 38681765 PMCID: PMC11045944 DOI: 10.3389/fendo.2024.1315046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 03/29/2024] [Indexed: 05/01/2024] Open
Abstract
Background NAFLD (Nonalcoholic fatty liver disease) is becoming an increasingly common cause of chronic liver disease. Metabolic dysfunction, overweight/obesity, and diabetes are thought to be closely associated with increased NAFLD risk. However, few studies have focused on the mechanisms of NAFLD occurrence in T1DM. Methods We conducted a two-sample Mendelian randomization (MR) analysis to assess the causal association between T1DM and NAFLD with/without complications, such as coma, renal complications, ketoacidosis, neurological complications, and ophthalmic complications. Multiple Mendelian randomization methods, such as the inverse variance weighted (IVW) method, weighted median method, and MR-Egger test were performed to evaluate the causal association of T1DM and NAFLD using genome-wide association study summary data from different consortia, such as Finngen and UK biobank. Results We selected 37 SNPs strongly associated with NAFLD/LFC (at a significance level of p < 5 × 10-8) as instrumental variables from the Finnish database based on the T1DM phenotype (8,967 cases and 308,373 controls). We also selected 14/16 SNPs based on with or without complications. The results suggest that the genetic susceptibility of T1DM does not increase the risk of NAFLD (OR=1.005 [0.99, 1.02], IVW p=0.516, MR Egger p=0.344, Weighted median p=0.959, Weighted mode p=0.791), regardless of whether complications are present. A slight causal effect of T1DM without complications on LFC was observed (OR=1.025 [1.00, 1.03], MR Egger p=0.045). However, none of the causal relationships were significant in the IVW (p=0.317), Weighted median (p=0.076), and Weighted mode (p=0.163) methods. Conclusion Our study did not find conclusive evidence for a causal association between T1DM and NAFLD, although clinical observations indicate increasing abnormal transaminase prevalence and NAFLD progression in T1DM patients.
Collapse
Affiliation(s)
- Lin Tuo
- Department of Infectious Disease, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | | | | | - Xing-xiang Yang
- Department of Infectious Disease, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
38
|
Li SJ, Wu YL, Chen JH, Shen SY, Duan J, Xu HE. Autoimmune diseases: targets, biology, and drug discovery. Acta Pharmacol Sin 2024; 45:674-685. [PMID: 38097717 PMCID: PMC10943205 DOI: 10.1038/s41401-023-01207-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 11/20/2023] [Indexed: 03/17/2024] Open
Abstract
Autoimmune diseases (AIDs) arise from a breakdown in immunological self-tolerance, wherein the adaptive immune system mistakenly attacks healthy cells, tissues and organs. AIDs impose excessive treatment costs and currently rely on non-specific and universal immunosuppression, which only offer symptomatic relief without addressing the underlying causes. AIDs are driven by autoantigens, targeting the autoantigens holds great promise in transforming the treatment of these diseases. To achieve this goal, a comprehensive understanding of the pathogenic mechanisms underlying different AIDs and the identification of specific autoantigens are critical. In this review, we categorize AIDs based on their underlying causes and compile information on autoantigens implicated in each disease, providing a roadmap for the development of novel immunotherapy regimens. We will focus on type 1 diabetes (T1D), which is an autoimmune disease characterized by irreversible destruction of insulin-producing β cells in the Langerhans islets of the pancreas. We will discuss insulin as possible autoantigen of T1D and its role in T1D pathogenesis. Finally, we will review current treatments of TID and propose a potentially effective immunotherapy targeting autoantigens.
Collapse
Affiliation(s)
- Shu-Jie Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- Department of Traditional Chinese Medicine, Fujian Medical University Union Hospital, Fuzhou, 350000, China.
| | - Yan-Li Wu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Juan-Hua Chen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Shi-Yi Shen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jia Duan
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, 528400, China.
| | - H Eric Xu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- School of Life Science and Technology, Shanghai Tech University, Shanghai, 201210, China.
| |
Collapse
|
39
|
Alleva DG, Delpero AR, Sathiyaseelan T, Murikipudi S, Lancaster TM, Atkinson MA, Wasserfall CH, Yu L, Ragupathy R, Bonami RH, Zion TC. An antigen-specific immunotherapeutic, AKS-107, deletes insulin-specific B cells and prevents murine autoimmune diabetes. Front Immunol 2024; 15:1367514. [PMID: 38515750 PMCID: PMC10954819 DOI: 10.3389/fimmu.2024.1367514] [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: 01/08/2024] [Accepted: 02/13/2024] [Indexed: 03/23/2024] Open
Abstract
Introduction The antigen-presenting cell function of insulin-reactive B cells promotes type 1 diabetes (T1D) in non-obese diabetic (NOD) mice by stimulating pathogenic T cells leading to destruction of insulin-producing β-cells of pancreatic islets. Methods/Results To target insulin-reactive B cells, AKS-107, a human IgG1 Fc molecule fused with human insulin A and B chains, was engineered to retain conformational insulin epitopes that bound mouse and human B cell receptors but prevented binding to the insulin metabolic receptor. AKS-107 Fc-mediated deletion of insulin-reactive B cells was demonstrated via ex vivo and in vivo experiments with insulin-reactive B cell receptor transgenic mouse strains, VH125Tg/NOD and Tg125(H+L)/NOD. As an additional immune tolerance feature, the Y16A mutation of the insulin B(9-23) dominant T cell epitope was engineered into AKS-107 to suppress activation of insulin-specific T cells. In mice and non-human primates, AKS-107 was well-tolerated, non-immunogenic, did not cause hypoglycemia even at high doses, and showed an expectedly protracted pharmacokinetic profile. AKS-107 reproducibly prevented spontaneous diabetes from developing in NOD and VH125Tg/NOD mice that persisted for months after cessation of treatment, demonstrating durable immune tolerance. Discussion These preclinical outcomes position AKS-107 for clinical development in T1D prevention settings.
Collapse
Affiliation(s)
- David G. Alleva
- Department of Pharmacology, Akston Biosciences, Inc., Beverly, MA, United States
| | - Andrea R. Delpero
- Department of Pharmacology, Akston Biosciences, Inc., Beverly, MA, United States
| | | | - Sylaja Murikipudi
- Department of Pharmacology, Akston Biosciences, Inc., Beverly, MA, United States
| | - Thomas M. Lancaster
- Department of Pharmacology, Akston Biosciences, Inc., Beverly, MA, United States
| | - Mark A. Atkinson
- Departments of Pathology, Immunology, and Laboratory Medicine, College of Medicine, and Diabetes Institute, The University of Florida, Gainesville, FL, United States
| | - Clive H. Wasserfall
- Departments of Pathology, Immunology, and Laboratory Medicine, College of Medicine, and Diabetes Institute, The University of Florida, Gainesville, FL, United States
| | - Liping Yu
- Barbara Davis Center for Diabetes, School of Medicine, University of Colorado, Aurora, CO, United States
| | - Ramya Ragupathy
- Department of Pharmacology, Akston Biosciences, Inc., Beverly, MA, United States
| | - Rachel H. Bonami
- Division of Rheumatology and Immunology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN, United States
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Todd C. Zion
- Department of Pharmacology, Akston Biosciences, Inc., Beverly, MA, United States
| |
Collapse
|
40
|
Merolla A, De Lorenzo R, Ferrannini G, Renzi C, Ulivi F, Bazzigaluppi E, Lampasona V, Bosi E. Universal screening for early detection of chronic autoimmune, metabolic and cardiovascular diseases in the general population using capillary blood (UNISCREEN): low-risk interventional, single-centre, pilot study protocol. BMJ Open 2024; 14:e078983. [PMID: 38448070 PMCID: PMC10916121 DOI: 10.1136/bmjopen-2023-078983] [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: 08/17/2023] [Accepted: 01/29/2024] [Indexed: 03/08/2024] Open
Abstract
INTRODUCTION Chronic autoimmune (type 1 diabetes and coeliac disease) and metabolic/cardiovascular (type 2 diabetes, dyslipidaemia, hypertension) diseases are highly prevalent across all age ranges representing a major public health burden. Universal screening for prediction/early identification of these conditions is a potential tool for reducing their impact on the general population. The aim of this study is to assess whether universal screening using capillary blood sampling is feasible at a population-based level. METHODS AND ANALYSIS This is a low-risk interventional, single-centre, pilot study for a population-based screening programme denominated UNISCREEN. Participants are volunteers aged 1-100 who reside in the town of Cantalupo (Milan, Italy) undergoing: (1) interview collecting demographics, anthropometrics and medical history; (2) capillary blood collection for measurement of type 1 diabetes and coeliac disease-specific autoantibodies and immediate measurement of glucose, glycated haemoglobin and lipid panel by point-of-care devices; (3) venous blood sampling to confirm autoantibody-positivity; (4) blood pressure measurement; (5) fulfilment of a feasibility and acceptability questionnaire. The outcomes are the assessment of feasibility and acceptability of capillary blood screening, the prevalence of presymptomatic type 1 diabetes and undiagnosed coeliac disease, distribution of glucose categories, lipid panel and estimate of cardiovascular risk in the study population. With approximately 3000 inhabitants, the screened population is expected to encompass at least half of its size, approaching nearly 1500 individuals. ETHICS AND DISSEMINATION This protocol and the informed consent forms have been reviewed and approved by the San Raffaele Hospital Ethics Committee (approval number: 131/INT/2022). Written informed consent is obtained from all study participants or their parents if aged <18. Results will be published in scientific journals and presented at meetings. CONCLUSIONS If proven feasible and acceptable, this universal screening model would pave the way for larger-scale programmes, providing an opportunity for the implementation of innovative public health programmes in the general population. TRIAL REGISTRATION NUMBER NCT05841719.
Collapse
Affiliation(s)
| | | | | | - Cristina Renzi
- Vita-Salute San Raffaele University, Milano, Italy
- Behavioural Science and Health, Institute of Epidemiology & Health Care, University College London, London, UK
| | | | | | - Vito Lampasona
- Diabetes Research Institute, IRCCS San Raffaele Hospital, Milan, Italy
| | - Emanuele Bosi
- Vita-Salute San Raffaele University, Milano, Italy
- Diabetes Research Institute, IRCCS San Raffaele Hospital, Milan, Italy
| |
Collapse
|
41
|
Pleus S, Tytko A, Landgraf R, Heinemann L, Werner C, Müller-Wieland D, Ziegler AG, Müller UA, Freckmann G, Kleinwechter H, Schleicher E, Nauck M, Petersmann A. Definition, Classification, Diagnosis and Differential Diagnosis of Diabetes Mellitus: Update 2023. Exp Clin Endocrinol Diabetes 2024; 132:112-124. [PMID: 38378016 DOI: 10.1055/a-2166-6643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Affiliation(s)
- Stefan Pleus
- Institut für Diabetes-Technologie Forschungs- und Entwicklungsgesellschaft mbH an der Universität Ulm, Ulm, Germany
| | | | | | - Lutz Heinemann
- Science-Consulting in Diabetes GmbH, Düsseldorf, Germany
| | - Christoph Werner
- Department of Internal Medicine III, University Hospital Jena, Jena, Germany
| | | | | | - Ulrich A Müller
- Practice for Endocrinology and Diabetology, Dr. Kielstein Ambulante Medizinische Versorgung GmbH, Jena, Germany
| | - Guido Freckmann
- Institut für Diabetes-Technologie Forschungs- und Entwicklungsgesellschaft mbH an der Universität Ulm, Ulm, Germany
| | | | - Erwin Schleicher
- Institute of Clinical Chemistry and Pathobiochemistry - Central Laboratory, University Hospital Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD) Munich-Neuherberg, Munich-Neuherberg, Germany
| | - Matthias Nauck
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, University Medicine, Greifswald, Germany
| | - Astrid Petersmann
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Oldenburg, Oldenburg, Germany
| |
Collapse
|
42
|
Dovc K, Bode BW, Battelino T. Continuous and Intermittent Glucose Monitoring in 2023. Diabetes Technol Ther 2024; 26:S14-S31. [PMID: 38441451 DOI: 10.1089/dia.2024.2502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/07/2024]
Affiliation(s)
- Klemen Dovc
- University Medical Center Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Bruce W Bode
- Atlanta Diabetes Associates and Emory University School of Medicine, Atlanta, GA, USA
| | - Tadej Battelino
- University Medical Center Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
43
|
Zaharieva DP, Ding VY, Addala A, Prahalad P, Bishop F, Hood KK, Desai M, Wilson DM, Buckingham BA, Maahs DM. Diabetic Ketoacidosis at Diagnosis in Youth with Type 1 Diabetes Is Associated with a Higher Hemoglobin A1c Even with Intensive Insulin Management. Diabetes Technol Ther 2024; 26:176-183. [PMID: 37955644 PMCID: PMC10877392 DOI: 10.1089/dia.2023.0405] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2023]
Abstract
Introduction: Diabetic ketoacidosis (DKA) at diagnosis is associated with short- and long-term complications. We assessed the relationship between DKA status and hemoglobin A1c (A1c) levels in the first year following type 1 diabetes (T1D) diagnosis. Research Design and Methods: The Pilot Teamwork, Targets, Technology, and Tight Control (4T) study offered continuous glucose monitoring to youth with T1D within 1 month of diagnosis. A1c levels were compared between historical (n = 271) and Pilot 4T (n = 135) cohorts stratified by DKA status at diagnosis (DKA: historical = 94, 4T = 67 versus without DKA: historical = 177, 4T = 68). A1c was evaluated using locally estimated scatter plot smoothing. Change in A1c from 4 to 12 months postdiagnosis was evaluated using a linear mixed model. Results: Median age was 9.7 (interquartile range [IQR]: 6.6, 12.7) versus 9.7 (IQR: 6.8, 12.7) years, 49% versus 47% female, 44% versus 39% non-Hispanic White in historical versus Pilot 4T. In historical and 4T cohorts, DKA at diagnosis demonstrated higher A1c at 6 (0.5% [95% confidence interval (CI): 0.21-0.79; P < 0.01] and 0.38% [95% CI: 0.02-0.74; P = 0.04], respectively), and 12 months (0.62% [95% CI: -0.06 to 1.29; P = 0.07] and 0.39% [95% CI: -0.32 to 1.10; P = 0.29], respectively). The highest % time in range (TIR; 70-180 mg/dL) was seen between weeks 15-20 (69%) versus 25-30 (75%) postdiagnosis for youth with versus without DKA in Pilot 4T, respectively. Conclusions: Pilot 4T improved A1c outcomes versus the historical cohort, but those with DKA at diagnosis had persistently elevated A1c throughout the study and intensive diabetes management did not mitigate this difference. DKA prevention at diagnosis may translate into better glycemic outcomes in the first-year postdiagnosis. Clinical Trial Registration: clinicaltrials.gov: NCT04336969.
Collapse
Affiliation(s)
- Dessi P. Zaharieva
- Division of Pediatric Endocrinology, Department of Pediatrics, Stanford University, Stanford, California, USA
| | - Victoria Y. Ding
- Division of Biomedical Informatics Research, Department of Medicine, Stanford University, Stanford, California, USA
| | - Ananta Addala
- Division of Pediatric Endocrinology, Department of Pediatrics, Stanford University, Stanford, California, USA
- Stanford Diabetes Research Center, Stanford University, Stanford, California, USA
| | - Priya Prahalad
- Division of Pediatric Endocrinology, Department of Pediatrics, Stanford University, Stanford, California, USA
- Stanford Diabetes Research Center, Stanford University, Stanford, California, USA
| | - Franziska Bishop
- Division of Pediatric Endocrinology, Department of Pediatrics, Stanford University, Stanford, California, USA
| | - Korey K. Hood
- Division of Pediatric Endocrinology, Department of Pediatrics, Stanford University, Stanford, California, USA
- Stanford Diabetes Research Center, Stanford University, Stanford, California, USA
| | - Manisha Desai
- Division of Biomedical Informatics Research, Department of Medicine, Stanford University, Stanford, California, USA
| | - Darrell M. Wilson
- Division of Pediatric Endocrinology, Department of Pediatrics, Stanford University, Stanford, California, USA
| | - Bruce A. Buckingham
- Division of Pediatric Endocrinology, Department of Pediatrics, Stanford University, Stanford, California, USA
| | - David M. Maahs
- Division of Pediatric Endocrinology, Department of Pediatrics, Stanford University, Stanford, California, USA
- Stanford Diabetes Research Center, Stanford University, Stanford, California, USA
| |
Collapse
|
44
|
Xiang R, Kelemen M, Xu Y, Harris LW, Parkinson H, Inouye M, Lambert SA. Recent advances in polygenic scores: translation, equitability, methods and FAIR tools. Genome Med 2024; 16:33. [PMID: 38373998 PMCID: PMC10875792 DOI: 10.1186/s13073-024-01304-9] [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: 06/09/2023] [Accepted: 02/07/2024] [Indexed: 02/21/2024] Open
Abstract
Polygenic scores (PGS) can be used for risk stratification by quantifying individuals' genetic predisposition to disease, and many potentially clinically useful applications have been proposed. Here, we review the latest potential benefits of PGS in the clinic and challenges to implementation. PGS could augment risk stratification through combined use with traditional risk factors (demographics, disease-specific risk factors, family history, etc.), to support diagnostic pathways, to predict groups with therapeutic benefits, and to increase the efficiency of clinical trials. However, there exist challenges to maximizing the clinical utility of PGS, including FAIR (Findable, Accessible, Interoperable, and Reusable) use and standardized sharing of the genomic data needed to develop and recalculate PGS, the equitable performance of PGS across populations and ancestries, the generation of robust and reproducible PGS calculations, and the responsible communication and interpretation of results. We outline how these challenges may be overcome analytically and with more diverse data as well as highlight sustained community efforts to achieve equitable, impactful, and responsible use of PGS in healthcare.
Collapse
Affiliation(s)
- Ruidong Xiang
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Martin Kelemen
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
| | - Yu Xu
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
| | - Laura W Harris
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Helen Parkinson
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Michael Inouye
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK.
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK.
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK.
| | - Samuel A Lambert
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| |
Collapse
|
45
|
Nassar M, Chaudhuri A, Ghanim H, Dandona P. Glucagon-like peptide-1 receptor agonists as a possible intervention to delay the onset of type 1 diabetes: A new horizon. World J Diabetes 2024; 15:133-136. [PMID: 38464377 PMCID: PMC10921167 DOI: 10.4239/wjd.v15.i2.133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/17/2023] [Accepted: 01/16/2024] [Indexed: 02/04/2024] Open
Abstract
Type 1 diabetes (T1D) is a chronic autoimmune condition that destroys insulin-producing beta cells in the pancreas, leading to insulin deficiency and hyper-glycemia. The management of T1D primarily focuses on exogenous insulin replacement to control blood glucose levels. However, this approach does not address the underlying autoimmune process or prevent the progressive loss of beta cells. Recent research has explored the potential of glucagon-like peptide-1 receptor agonists (GLP-1RAs) as a novel intervention to modify the disease course and delay the onset of T1D. GLP-1RAs are medications initially developed for treating type 2 diabetes. They exert their effects by enhancing glucose-dependent insulin secretion, suppressing glucagon secretion, and slowing gastric emptying. Emerging evidence suggests that GLP-1RAs may also benefit the treatment of newly diagnosed patients with T1D. This article aims to highlight the potential of GLP-1RAs as an intervention to delay the onset of T1D, possibly through their potential immunomodulatory and anti-inflammatory effects and preservation of beta-cells. This article aims to explore the potential of shifting the paradigm of T1D management from reactive insulin replacement to proactive disease modification, which should open new avenues for preventing and treating T1D, improving the quality of life and long-term outcomes for individuals at risk of T1D.
Collapse
Affiliation(s)
- Mahmoud Nassar
- Department of Internal Medicine, Division of Endocrinology, Diabetes and Metabolism, Jacobs School of Medicine and Biomedical Sciences, University of Buffalo, Buffalo, NY 14221, United States
| | - Ajay Chaudhuri
- Department of Internal Medicine, Division of Endocrinology, Diabetes and Metabolism, Jacobs School of Medicine and Biomedical Sciences, University of Buffalo, Buffalo, NY 14221, United States
| | - Husam Ghanim
- Department of Internal Medicine, Division of Endocrinology, Diabetes and Metabolism, Jacobs School of Medicine and Biomedical Sciences, University of Buffalo, Buffalo, NY 14221, United States
| | - Paresh Dandona
- Department of Internal Medicine, Division of Endocrinology, Diabetes and Metabolism, Jacobs School of Medicine and Biomedical Sciences, University of Buffalo, Buffalo, NY 14221, United States
| |
Collapse
|
46
|
Tandel D, Hinton B, de Jesus Cortez F, Seftel D, Robinson P, Tsai CT. Advances in risk predictive performance of pre-symptomatic type 1 diabetes via the multiplex Antibody-Detection-by-Agglutination-PCR assay. Front Endocrinol (Lausanne) 2024; 15:1340436. [PMID: 38390205 PMCID: PMC10882067 DOI: 10.3389/fendo.2024.1340436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 01/22/2024] [Indexed: 02/24/2024] Open
Abstract
Introduction Achieving early diagnosis of pre-symptomatic type 1 diabetes is critical to reduce potentially life-threatening diabetic ketoacidosis (DKA) at symptom onset, link patients to FDA approved therapeutics that can delay disease progression and support novel interventional drugs development. The presence of two or more islet autoantibodies in pre-symptomatic type 1 diabetes patients indicates high-risk of progression to clinical manifestation. Method Herein, we characterized the capability of multiplex ADAP assay to predict type 1 diabetes progression. We obtained retrospective coded sera from a cohort of 48 progressors and 44 non-progressors from the NIDDK DPT-1 study. Result The multiplex ADAP assay and radiobinding assays had positive predictive value (PPV)/negative predictive value (NPV) of 68%/92% and 67%/66% respectively. The improved NPV stemmed from 12 progressors tested positive for multiple islet autoantibodies by multiplex ADAP assay but not by RBA. Furthermore, 6 out of these 12 patients tested positive for multiple islet autoantibodies by RBA in subsequent sampling events with a median delay of 2.8 years compared to multiplex ADAP assay. Discussion In summary, multiplex ADAP assay could be an ideal tool for type 1 diabetes risk testing due to its sample-sparing nature (4µL), non-radioactiveness, compatibility with widely available real-time qPCR instruments and favorable risk prediction capability.
Collapse
Affiliation(s)
| | | | | | | | | | - Cheng-ting Tsai
- Research & Product Development, Enable Biosciences, South San Francisco, CA, United States
| |
Collapse
|
47
|
Foster TP, Jacobsen LM, Bruggeman B, Salmon C, Hosford J, Chen A, Cintron M, Mathews CE, Wasserfall C, Brusko MA, Brusko TM, Atkinson MA, Schatz DA, Haller MJ. Low-Dose Antithymocyte Globulin: A Pragmatic Approach to Treating Stage 2 Type 1 Diabetes. Diabetes Care 2024; 47:285-289. [PMID: 38117469 PMCID: PMC10834389 DOI: 10.2337/dc23-1750] [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: 09/15/2023] [Accepted: 11/15/2023] [Indexed: 12/21/2023]
Abstract
OBJECTIVE Low-dose antithymocyte globulin (ATG) (2.5 mg/kg) preserves C-peptide and reduces HbA1c in new-onset stage 3 type 1 diabetes, yet efficacy in delaying progression from stage 2 to stage 3 has not been evaluated. RESEARCH DESIGN AND METHODS Children (n = 6) aged 5-14 years with stage 2 type 1 diabetes received off-label, low-dose ATG. HbA1c, C-peptide, continuous glucose monitoring, insulin requirements, and side effects were followed for 18-48 months. RESULTS Three subjects (50%) remained diabetes free after 1.5, 3, and 4 years of follow-up, while three developed stage 3 within 1-2 months after therapy. Eighteen months posttreatment, even disease progressors demonstrated near-normal HbA1c (5.1% [32 mmol/mol], 5.6% [38 mmol/mol], and 5.3% [34 mmol/mol]), time in range (93%, 88%, and 98%), low insulin requirements (0.17, 0.18, and 0.34 units/kg/day), and robust C-peptide 90 min after mixed meal (1.3 ng/dL, 2.3 ng/dL, and 1.4 ng/dL). CONCLUSIONS These observations support additional prospective studies evaluating ATG in stage 2 type 1 diabetes.
Collapse
Affiliation(s)
- Timothy P. Foster
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL
| | - Laura M. Jacobsen
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL
- Department of Pathology, Immunology, and Laboratory Medicine, Diabetes Institute, University of Florida, Gainesville, FL
| | - Brittany Bruggeman
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL
| | - Chelsea Salmon
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL
| | - Jennifer Hosford
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL
| | - Angela Chen
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL
| | - Miriam Cintron
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL
| | - Clayton E. Mathews
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL
- Department of Pathology, Immunology, and Laboratory Medicine, Diabetes Institute, University of Florida, Gainesville, FL
| | - Clive Wasserfall
- Department of Pathology, Immunology, and Laboratory Medicine, Diabetes Institute, University of Florida, Gainesville, FL
| | - Maigan A. Brusko
- Department of Pathology, Immunology, and Laboratory Medicine, Diabetes Institute, University of Florida, Gainesville, FL
| | - Todd M. Brusko
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL
- Department of Pathology, Immunology, and Laboratory Medicine, Diabetes Institute, University of Florida, Gainesville, FL
| | - Mark A. Atkinson
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL
- Department of Pathology, Immunology, and Laboratory Medicine, Diabetes Institute, University of Florida, Gainesville, FL
| | - Desmond A. Schatz
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL
- Department of Pathology, Immunology, and Laboratory Medicine, Diabetes Institute, University of Florida, Gainesville, FL
| | - Michael J. Haller
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL
- Department of Pathology, Immunology, and Laboratory Medicine, Diabetes Institute, University of Florida, Gainesville, FL
| |
Collapse
|
48
|
Martens PJ, Mathieu C. Type 1 diabetes mellitus: a brave new world. Nat Rev Endocrinol 2024; 20:71-72. [PMID: 38057482 DOI: 10.1038/s41574-023-00936-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/08/2023]
|
49
|
Sing ABE, Naselli G, Huang D, Watson K, Colman PG, Harrison LC, Wentworth JM. Feasibility and Validity of In-Home Self-Collected Capillary Blood Spot Screening for Type 1 Diabetes Risk. Diabetes Technol Ther 2024; 26:87-94. [PMID: 37976038 DOI: 10.1089/dia.2023.0345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2023]
Abstract
Aims: Self-collection of a blood sample for autoantibody testing has potential to facilitate screening for type 1 diabetes risk. We sought to determine the feasibility and acceptability of this approach and the performance of downstream antibody assays. Methods: People living with type 1 diabetes and their family members (N = 97) provided paired capillary blood spot and serum samples collected, respectively, by themselves and a health worker. They provided feedback on the ease, convenience, and painfulness of blood spot collection. Islet antibodies were measured in blood spots by antibody detection by agglutination PCR (ADAP) or multiplex enzyme-linked immunoassay (ELISA), and in serum by radioimmunoassay (RIA) or ELISA. Results: Using serum RIA and ELISA to define antibody status, 50 antibody-negative (Abneg) and 47 antibody-positive (Abpos) participants enrolled, of whom 43 and 47, respectively, returned testable blood spot samples. The majority indicated that self-collection was easier, more convenient, and less painful than formal venesection. The sensitivity and specificity for detection of Abpos by blood spot were, respectively, 85% and 98% for ADAP and 87% and 100% for multiplex ELISA. The specificities by ADAP for each of the four antigen specificities ranged from 98% to 100% and areas under the receiver operator curve from 0.841 to 0.986. Conclusions: Self-collected blood spot sampling is preferred over venesection by research participants. ADAP and multiplex ELISA are highly specific assays for islet antibodies in blood spots with acceptable performance for use alone or in combination to facilitate screening for type 1 diabetes risk. Clinical Trial Registration number: ACTRN12620000510943.
Collapse
Affiliation(s)
- Anna B E Sing
- Population Health and Immunity Division, Walter and Eliza Hall Institute, Parkville, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Gaetano Naselli
- Population Health and Immunity Division, Walter and Eliza Hall Institute, Parkville, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Dexing Huang
- Population Health and Immunity Division, Walter and Eliza Hall Institute, Parkville, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Kelly Watson
- Royal Melbourne Hospital Department of Diabetes and Endocrinology, Parkville, Australia
| | - Peter G Colman
- Royal Melbourne Hospital Department of Diabetes and Endocrinology, Parkville, Australia
- University of Melbourne Department of Medicine, Royal Melbourne Hospital, Parkville, Australia
| | - Leonard C Harrison
- Population Health and Immunity Division, Walter and Eliza Hall Institute, Parkville, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - John M Wentworth
- Population Health and Immunity Division, Walter and Eliza Hall Institute, Parkville, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Australia
- Royal Melbourne Hospital Department of Diabetes and Endocrinology, Parkville, Australia
- University of Melbourne Department of Medicine, Royal Melbourne Hospital, Parkville, Australia
| |
Collapse
|
50
|
Besser REJ, Long AE, Owen KR, Law R, Birks JS, Pearce O, Williams CL, Scudder CL, McDonald TJ, Todd JA. Transdermal Blood Sampling for C-Peptide Is a Minimally Invasive, Reliable Alternative to Venous Sampling in Children and Adults With Type 1 Diabetes. Diabetes Care 2024; 47:239-245. [PMID: 38087932 DOI: 10.2337/dc23-1379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 11/02/2023] [Indexed: 01/21/2024]
Abstract
OBJECTIVE C-peptide and islet autoantibodies are key type 1 diabetes biomarkers, typically requiring venous sampling, which limits their utility. We assessed transdermal capillary blood (TCB) collection as a practical alternative. RESEARCH DESIGN AND METHODS Ninety-one individuals (71 with type 1 diabetes, 20 control; individuals with type 1 diabetes: aged median 14.8 years [interquartile range (IQR) 9.1-17.1], diabetes duration 4.0 years [1.5-7.7]; control individuals: 42.2 years [38.0-52.1]) underwent contemporaneous venous and TCB sampling for measurement of plasma C-peptide. Participants with type 1 diabetes also provided venous serum and plasma, and TCB plasma for measurement of autoantibodies to glutamate decarboxylase, islet antigen-2, and zinc transporter 8. The ability of TCB plasma to detect significant endogenous insulin secretion (venous C-peptide ≥200 pmol/L) was compared along with agreement in levels, using Bland-Altman. Venous serum was compared with venous and TCB plasma for detection of autoantibodies, using established thresholds. Acceptability was assessed by age-appropriate questionnaire. RESULTS Transdermal sampling took a mean of 2.35 min (SD 1.49). Median sample volume was 50 µL (IQR 40-50) with 3 of 91 (3.3%) failures, and 13 of 88 (14.7%) <35 µL. TCB C-peptide showed good agreement with venous plasma (mean venous ln[C-peptide] - TCB ln[C-peptide] = 0.008, 95% CI [-0.23, 0.29], with 100% [36 of 36] sensitivity/100% [50 of 50] specificity to detect venous C-peptide ≥200 pmol/L). Where venous serum in multiple autoantibody positive TCB plasma agreed in 22 of 32 (sensitivity 69%), comparative specificity was 35 of 36 (97%). TCB was preferred to venous sampling (type 1 diabetes: 63% vs. 7%; 30% undecided). CONCLUSIONS Transdermal capillary testing for C-peptide is a sensitive, specific, and acceptable alternative to venous sampling; TCB sampling for islet autoantibodies needs further assessment.
Collapse
Affiliation(s)
- Rachel E J Besser
- Juvenile Diabetes Research Foundation/Wellcome Diabetes and Inflammation Laboratory, Wellcome Centre for Human Genetics, Nuffield Department of Medicine, Oxford National Institute for Health Research (NIHR) Biomedical Research Centre, University of Oxford, Oxford, U.K
- Department of Paediatric Diabetes, Oxford Children's Hospital, John Radcliffe Hospital, Oxford, U.K
| | - Anna E Long
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, U.K
| | - Katharine R Owen
- Oxford NIHR Biomedical Research Centre, University of Oxford, Oxford, U.K
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, U.K
| | - Rebecca Law
- Department of Paediatric Diabetes, Oxford Children's Hospital, John Radcliffe Hospital, Oxford, U.K
| | - Jacqueline S Birks
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Centre for Statistics in Medicine, University of Oxford, Oxford, U.K
| | - Olivia Pearce
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, U.K
| | - Claire L Williams
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, U.K
| | - Claire L Scudder
- Juvenile Diabetes Research Foundation/Wellcome Diabetes and Inflammation Laboratory, Wellcome Centre for Human Genetics, Nuffield Department of Medicine, Oxford National Institute for Health Research (NIHR) Biomedical Research Centre, University of Oxford, Oxford, U.K
| | - Timothy J McDonald
- Academic Department of Blood Sciences, Royal Devon University Hospital, Exeter, U.K
- Exeter NIHR Biomedical Research Centre, University of Exeter, Exeter, U.K
| | - John A Todd
- Juvenile Diabetes Research Foundation/Wellcome Diabetes and Inflammation Laboratory, Wellcome Centre for Human Genetics, Nuffield Department of Medicine, Oxford National Institute for Health Research (NIHR) Biomedical Research Centre, University of Oxford, Oxford, U.K
| |
Collapse
|