1
|
Vasilev G, Kokudeva M, Siliogka E, Padilla N, Shumnalieva R, Della-Morte D, Ricordi C, Mihova A, Infante M, Velikova T. T helper 17 cells and interleukin-17 immunity in type 1 diabetes: From pathophysiology to targeted immunotherapies. World J Diabetes 2025; 16:99936. [PMID: 40236846 PMCID: PMC11947927 DOI: 10.4239/wjd.v16.i4.99936] [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: 08/09/2024] [Revised: 12/06/2024] [Accepted: 02/07/2025] [Indexed: 02/28/2025] Open
Abstract
Type 1 diabetes (T1D) is a chronic organ-specific autoimmune disorder characterized by a progressive loss of the insulin-secreting pancreatic beta cells, which ultimately results in insulinopenia, hyperglycemia and lifelong need for exogenous insulin therapy. In the pathophysiological landscape of T1D, T helper 17 cells (Th17 cells) and their hallmark cytokine, interleukin (IL)-17, play pivotal roles from disease onset to disease progression. In this narrative mini-review, we discuss the dynamic interplay between Th17 cells and IL-17 in the context of T1D, providing insights into the underlying immunologic mechanisms contributing to the IL-17-immunity-mediated pancreatic beta-cell destruction. Furthermore, we summarized the main animal and clinical studies that investigated Th17- and IL-17-targeted interventions as promising immunotherapies able to alter the natural history of T1D.
Collapse
Affiliation(s)
- Georgi Vasilev
- Clinic of Neurology and Department of Emergency Medicine, UMHAT "Sv. Georgi", Plovdiv 4000, Bulgaria
- Medical Faculty, Sofia University St. Kliment Ohridski, Sofia 1407, Bulgaria
| | - Maria Kokudeva
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Medical University of Sofia, Sofia 1000, Bulgaria
| | - Elina Siliogka
- Faculty of Medicine, National and Kapodistrian University of Athens, Athens 11527, Attikí, Greece
| | - Nathalia Padilla
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, United States
| | - Russka Shumnalieva
- Medical Faculty, Sofia University St. Kliment Ohridski, Sofia 1407, Bulgaria
- Department of Rheumatology, Clinic of Rheumatology, University Hospital "St. Anna", Medical University-Sofia, Sofia 1612, Bulgaria
| | - David Della-Morte
- Department of Biomedicine and Prevention, Section of Clinical Nutrition and Nutrigenomics, University of Rome Tor Vergata, Rome 00133, Italy
| | - Camillo Ricordi
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, United States
| | | | - Marco Infante
- Section of Diabetes & Metabolic Disorders, UniCamillus, Saint Camillus International University of Health Sciences, Rome 00131, Italy
| | - Tsvetelina Velikova
- Medical Faculty, Sofia University St. Kliment Ohridski, Sofia 1407, Bulgaria
| |
Collapse
|
2
|
Mondal S, Pappachan JM. Current perspectives and the future of disease-modifying therapies in type 1 diabetes. World J Diabetes 2025; 16:99496. [PMID: 39817218 PMCID: PMC11718456 DOI: 10.4239/wjd.v16.i1.99496] [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: 07/23/2024] [Revised: 10/11/2024] [Accepted: 11/04/2024] [Indexed: 11/29/2024] Open
Abstract
Use of immunomodulating agents to prevent the progression of autoimmune β-cell damage leading to type 1 diabetes mellitus (T1DM) is an interesting area for research. These include non-specific anti-inflammatory agents, immunologic vaccination and anti-inflammatory agents targeting specific immune cells or cytokines. Teplizumab is an anti-CD3-molecule that binds to and leads to the disappearance of the CD3/TCR complex and rendering the T cell anergic to its target antigen. Preclinical and clinical trials have demonstrated its efficacy in reducing the decline in serum C-peptide levels and the need for insulin therapy if used early in the disease process of T1DM. The benefits have been apparent as early as six months to as long as seven years after therapy. It has recently been approved by the Food and Drug Administration to delay the onset of clinical (stage 3) type 1 diabetes in children above 8 years of age. In their recent meta-analysis published in the World Journal of Diabetes, Ma et al found that those in the teplizumab treatment group have a greater likelihood of reduction in insulin use, change in C-peptide response, and better glycemic control compared to the control group with a good safety profile. However, all the included randomized control trials have been conducted in high-income countries. High cost of therapy and unknown utility of the molecule in stage 3 disease limit its widespread use.
Collapse
Affiliation(s)
- Sunetra Mondal
- Department of Endocrinology, NRS Medical College and Hospital, Kolkata 700014, West Bengal, India
| | - Joseph M Pappachan
- Department of Endocrinology and Metabolism, Lancashire Teaching Hospitals NHS Trust, Preston PR2 9HT, United Kingdom
- Faculty of Science, Manchester Metropolitan University, Manchester M15 6BH, United Kingdom
- Department of Endocrinology, Kasturba Medical College, Manipal 576104, India
| |
Collapse
|
3
|
Tatovic D, Marwaha A, Taylor P, Hanna SJ, Carter K, Cheung WY, Luzio S, Dunseath G, Hutchings HA, Holland G, Hiles S, Fegan G, Williams E, Yang JHM, Domingo-Vila C, Pollock E, Wadud M, Ward-Hartstonge K, Marques-Jones S, Bowen-Morris J, Stenson R, Levings MK, Gregory JW, Tree TIM, Dayan C. Ustekinumab for type 1 diabetes in adolescents: a multicenter, double-blind, randomized phase 2 trial. Nat Med 2024; 30:2657-2666. [PMID: 39079992 PMCID: PMC11405276 DOI: 10.1038/s41591-024-03115-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 06/05/2024] [Indexed: 09/18/2024]
Abstract
Immunotherapy targeting the autoimmune process in type 1 diabetes (T1D) can delay the loss of β-cells but needs to have minimal adverse effects to be an adjunct to insulin in the management of T1D. Ustekinumab binds to the shared p40 subunit of interleukin (IL)-12 and IL-23, targeting development of T helper 1 cells and T helper 17 cells (TH1 and TH17 cells) implicated in the pathogenesis of T1D. We conducted a double-blind, randomized controlled trial of ustekinumab in 72 adolescents aged 12-18 years with recent-onset T1D. Treatment was well tolerated with no increase in adverse events. At 12 months, β-cell function, measured by stimulated C-peptide, was 49% higher in the intervention group (P = 0.02), meeting the prespecified primary outcome. Preservation of C-peptide correlated with the reduction of T helper cells co-secreting IL-17A and interferon-γ (TH17.1 cells, P = 0.04) and, in particular, with the reduction in a subset of TH17.1 cells co-expressing IL-2 and granulocyte-macrophage colony-stimulating factor (IL-2+ GM-CSF+ TH17.1 cells, P = 0.04). A significant fall in β-cell-targeted (proinsulin-specific) IL-17A-secreting T cells was also seen (P = 0.0003). Although exploratory, our data suggest a role for an activated subset of TH17.1 cells in T1D that can be targeted with minimal adverse effects to reduce C-peptide loss, which requires confirmation in a larger study. (International Standard Randomised Controlled Trial Number Registry: ISRCTN 14274380).
Collapse
Affiliation(s)
- Danijela Tatovic
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK.
| | | | - Peter Taylor
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK
| | - Stephanie J Hanna
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK
| | - Kym Carter
- Diabetes Research Unit Cymru, Institute for Life Sciences, Swansea University, Swansea, UK
| | - W Y Cheung
- Diabetes Research Unit Cymru, Institute for Life Sciences, Swansea University, Swansea, UK
| | - Steve Luzio
- Diabetes Research Unit Cymru, Institute for Life Sciences, Swansea University, Swansea, UK
| | - Gareth Dunseath
- Diabetes Research Unit Cymru, Institute for Life Sciences, Swansea University, Swansea, UK
| | | | - Gail Holland
- Swansea Trials Unit, Swansea University Medical School, Swansea, UK
| | - Steve Hiles
- Swansea Trials Unit, Swansea University Medical School, Swansea, UK
| | - Greg Fegan
- Swansea Trials Unit, Swansea University Medical School, Swansea, UK
| | - Evangelia Williams
- Department of Immunobiology, School of Immunology & Microbial Sciences, King's College London, Guy's Hospital, London, UK
| | - Jennie H M Yang
- Department of Immunobiology, School of Immunology & Microbial Sciences, King's College London, Guy's Hospital, London, UK
| | - Clara Domingo-Vila
- Department of Immunobiology, School of Immunology & Microbial Sciences, King's College London, Guy's Hospital, London, UK
| | - Emily Pollock
- Department of Immunobiology, School of Immunology & Microbial Sciences, King's College London, Guy's Hospital, London, UK
| | - Muntaha Wadud
- Department of Immunobiology, School of Immunology & Microbial Sciences, King's College London, Guy's Hospital, London, UK
| | - Kirsten Ward-Hartstonge
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
| | | | - Jane Bowen-Morris
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK
| | - Rachel Stenson
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK
| | - Megan K Levings
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
| | - John W Gregory
- Division of Population Medicine, Cardiff University School of Medicine, Cardiff, UK
| | - Timothy I M Tree
- Department of Immunobiology, School of Immunology & Microbial Sciences, King's College London, Guy's Hospital, London, UK
| | - Colin Dayan
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK
| |
Collapse
|
4
|
Cardona-Hernández R, Castaño González LA, Atance EP, Echevarría IR. Advancing toward the aetiologic treatment of type 1 diabetes in the early stages: the dawn of a new era in its management. An Pediatr (Barc) 2024; 101:1-2. [PMID: 38871542 DOI: 10.1016/j.anpede.2024.03.051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 03/22/2024] [Indexed: 06/15/2024] Open
Affiliation(s)
- Roque Cardona-Hernández
- Servicio de Endocrinología Pediátrica, Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
| | - Luis Antonio Castaño González
- Hospital Universitario Cruces, Universidad del País Vasco, IIS Biobizcaia, CIBERDEN, CIBERER, Endo-ERN, Baracaldo, Vizcaya, Spain
| | - Enrique Palomo Atance
- Unidad de Endocrinología Pediátrica, Hospital General Universitario de Ciudad Real, Ciudad Real, Spain
| | - Itxaso Rica Echevarría
- Hospital Universitario Cruces, Universidad del País Vasco, IIS Biobizcaia, CIBERDEN, CIBERER, Endo-ERN, Baracaldo, Vizcaya, Spain.
| |
Collapse
|
5
|
Honardoost MA, Adinatha A, Schmidt F, Ranjan B, Ghaeidamini M, Arul Rayan N, Gek Liang Lim M, Joanito I, Xiao Xuan Lin Q, Rajagopalan D, Qi Mok S, Hwang YY, Larbi A, Khor CC, Foo R, Boehm BO, Prabhakar S. Systematic immune cell dysregulation and molecular subtypes revealed by single-cell RNA-seq of subjects with type 1 diabetes. Genome Med 2024; 16:45. [PMID: 38539228 PMCID: PMC10976681 DOI: 10.1186/s13073-024-01300-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Accepted: 01/30/2024] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND Type 1 diabetes mellitus (T1DM) is a prototypic endocrine autoimmune disease resulting from an immune-mediated destruction of pancreatic insulin-secreting β cells. A comprehensive immune cell phenotype evaluation in T1DM has not been performed thus far at the single-cell level. METHODS In this cross-sectional analysis, we generated a single-cell transcriptomic dataset of peripheral blood mononuclear cells (PBMCs) from 46 manifest T1DM (stage 3) cases and 31 matched controls. RESULTS We surprisingly detected profound alterations in circulatory immune cells (1784 dysregulated genes in 13 immune cell types), far exceeding the count in the comparator systemic autoimmune disease SLE. Genes upregulated in T1DM were involved in WNT signaling, interferon signaling and migration of T/NK cells, antigen presentation by B cells, and monocyte activation. A significant fraction of these differentially expressed genes were also altered in T1DM pancreatic islets. We used the single-cell data to construct a T1DM metagene z-score (TMZ score) that distinguished cases and controls and classified patients into molecular subtypes. This score correlated with known prognostic immune markers of T1DM, as well as with drug response in clinical trials. CONCLUSIONS Our study reveals a surprisingly strong systemic dimension at the level of immune cell network in T1DM, defines disease-relevant molecular subtypes, and has the potential to guide non-invasive test development and patient stratification.
Collapse
Affiliation(s)
- Mohammad Amin Honardoost
- Laboratory of Systems Biology and Data Analytics, Genome Institute of Singapore (GIS), A*STAR (Agency for Science, Technology and Research), Singapore, 138672, Singapore
- Cardiovascular Diseases Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - Andreas Adinatha
- Laboratory of Systems Biology and Data Analytics, Genome Institute of Singapore (GIS), A*STAR (Agency for Science, Technology and Research), Singapore, 138672, Singapore
| | - Florian Schmidt
- Laboratory of Systems Biology and Data Analytics, Genome Institute of Singapore (GIS), A*STAR (Agency for Science, Technology and Research), Singapore, 138672, Singapore
| | - Bobby Ranjan
- Laboratory of Systems Biology and Data Analytics, Genome Institute of Singapore (GIS), A*STAR (Agency for Science, Technology and Research), Singapore, 138672, Singapore
| | - Maryam Ghaeidamini
- Laboratory of Systems Biology and Data Analytics, Genome Institute of Singapore (GIS), A*STAR (Agency for Science, Technology and Research), Singapore, 138672, Singapore
| | - Nirmala Arul Rayan
- Laboratory of Systems Biology and Data Analytics, Genome Institute of Singapore (GIS), A*STAR (Agency for Science, Technology and Research), Singapore, 138672, Singapore
| | - Michelle Gek Liang Lim
- Laboratory of Systems Biology and Data Analytics, Genome Institute of Singapore (GIS), A*STAR (Agency for Science, Technology and Research), Singapore, 138672, Singapore
| | - Ignasius Joanito
- Laboratory of Systems Biology and Data Analytics, Genome Institute of Singapore (GIS), A*STAR (Agency for Science, Technology and Research), Singapore, 138672, Singapore
| | - Quy Xiao Xuan Lin
- Laboratory of Systems Biology and Data Analytics, Genome Institute of Singapore (GIS), A*STAR (Agency for Science, Technology and Research), Singapore, 138672, Singapore
| | - Deepa Rajagopalan
- Laboratory of Systems Biology and Data Analytics, Genome Institute of Singapore (GIS), A*STAR (Agency for Science, Technology and Research), Singapore, 138672, Singapore
| | - Shi Qi Mok
- Integrated genomics platform, Genome Institute of Singapore (GIS), A*STAR (Agency for Science, Technology and Research), Singapore, 138672, Singapore
| | - You Yi Hwang
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Singapore, 138648, Singapore
| | - Anis Larbi
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Singapore, 138648, Singapore
| | - Chiea Chuen Khor
- Integrated genomics platform, Genome Institute of Singapore (GIS), A*STAR (Agency for Science, Technology and Research), Singapore, 138672, Singapore
| | - Roger Foo
- Cardiovascular Diseases Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore, 138673, Singapore
| | - Bernhard Otto Boehm
- Genome Institute of Singapore (GIS), A*STAR (Agency for Science, Technology and Research), Singapore, 138672, Singapore.
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore.
- Faculty of Life Sciences and Medicine, King's College London, London, WC2R 2LS, UK.
| | - Shyam Prabhakar
- Laboratory of Systems Biology and Data Analytics, Genome Institute of Singapore (GIS), A*STAR (Agency for Science, Technology and Research), Singapore, 138672, Singapore.
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore.
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Republic of Singapore.
| |
Collapse
|
6
|
Patel SK, Fourlanos S, Greenfield JR. Classification of type 1 diabetes: A pathogenic and treatment-based classification. Diabetes Metab Syndr 2024; 18:102986. [PMID: 38503115 DOI: 10.1016/j.dsx.2024.102986] [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: 06/01/2023] [Revised: 03/12/2024] [Accepted: 03/13/2024] [Indexed: 03/21/2024]
Abstract
AIM To improve the diagnosis and classification of patients who fail to satisfy current type 1 diabetes diagnostic criteria. METHODS Review of the literature and current diagnostic guidelines. DISCUSSION We propose a novel, clinically useful classification based on islet autoantibody status and non-fasting C-peptide levels. Notably, we discuss the subgroup of latent autoimmune diabetes in the young and propose a new subgroup classification of autoantibody negative type 1 diabetes in remission. CONCLUSION A novel classification system is proposed. Further work is needed to accurately diagnose and manage minority type 1 diabetes subgroups.
Collapse
Affiliation(s)
- Shivani K Patel
- Clinical Diabetes, Appetite and Metabolism Laboratory, Garvan Institute of Medical Research, Sydney, NSW, Australia; Department of Diabetes and Endocrinology, St Vincent's Hospital, Sydney, NSW, Australia; School of Clinical Medicine, UNSW Medicine & Health, St Vincent's Healthcare Clinical Campus, Faculty of Medicine, UNSW Sydney, Sydney, NSW, Australia.
| | - Spiros Fourlanos
- Department of Diabetes and Endocrinology, Royal Melbourne Hospital, Melbourne, VIC, Australia
| | - Jerry R Greenfield
- Clinical Diabetes, Appetite and Metabolism Laboratory, Garvan Institute of Medical Research, Sydney, NSW, Australia; Department of Diabetes and Endocrinology, St Vincent's Hospital, Sydney, NSW, Australia; School of Clinical Medicine, UNSW Medicine & Health, St Vincent's Healthcare Clinical Campus, Faculty of Medicine, UNSW Sydney, Sydney, NSW, Australia
| |
Collapse
|
7
|
Grasso EA, Chiarelli F. Type 1 Diabetes and Other Autoimmune Disorders in Children. Pediatr Diabetes 2024; 2024:5082064. [PMID: 40302955 PMCID: PMC12017020 DOI: 10.1155/2024/5082064] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 09/05/2023] [Accepted: 02/08/2024] [Indexed: 05/02/2025] Open
Abstract
The incidence of autoimmune disorders (AIDs) has been dramatically increasing in both children and adults over the past few years, and type 1 diabetes (T1D) is one of the diseases that has seen the highest growth. It is well-known that the dysimmune process may spread to other systems, leading to the onset of one or more AIDs in the same individual; however, the relationship between AIDs is not often recognized. The most frequently diagnosed AIDs in children and adolescents with T1D are thyroid diseases and celiac disease, but it is also important to consider the onset of the other conditions, such as juvenile idiopathic arthritis, multiple sclerosis, atrophic gastritis, inflammatory bowel diseases, and skin disorders such as vitiligo and psoriasis. This review aims to explore the overlap of T1D and other AIDs, focusing on the less common and lesser-known diseases. A better knowledge of these comorbidities may facilitate the identification of patients at risk to treat them in the preclinical period, before the onset of complications.
Collapse
Affiliation(s)
| | - Francesco Chiarelli
- Department of Paediatrics, University of Chieti, Via dei Vestini 31, Chieti, Italy
| |
Collapse
|
8
|
Joshi K, Harris M, Cotterill A, Wentworth JM, Couper JJ, Haynes A, Davis EA, Lomax KE, Huynh T. Continuous glucose monitoring has an increasing role in pre-symptomatic type 1 diabetes: advantages, limitations, and comparisons with laboratory-based testing. Clin Chem Lab Med 2024; 62:41-49. [PMID: 37349976 DOI: 10.1515/cclm-2023-0234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Accepted: 06/06/2023] [Indexed: 06/24/2023]
Abstract
Type 1 diabetes (T1D) is well-recognised as a continuum heralded by the development of islet autoantibodies, progression to islet autoimmunity causing beta cell destruction, culminating in insulin deficiency and clinical disease. Abnormalities of glucose homeostasis are known to exist well before the onset of typical symptoms. Laboratory-based tests such as the oral glucose tolerance test (OGTT) and glycated haemoglobin (HbA1c) have been used to stage T1D and assess the risk of progression to clinical T1D. Continuous glucose monitoring (CGM) can detect early glycaemic abnormalities and can therefore be used to monitor for metabolic deterioration in pre-symptomatic, islet autoantibody positive, at-risk individuals. Early identification of these children can not only reduce the risk of presentation with diabetic ketoacidosis (DKA), but also determine eligibility for prevention trials, which aim to prevent or delay progression to clinical T1D. Here, we describe the current state with regard to the use of the OGTT, HbA1c, fructosamine and glycated albumin in pre-symptomatic T1D. Using illustrative cases, we present our clinical experience with the use of CGM, and advocate for an increased role of this diabetes technology, for monitoring metabolic deterioration and disease progression in children with pre-symptomatic T1D.
Collapse
Affiliation(s)
- Kriti Joshi
- 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, Brisbane, QLD, Australia
| | - Mark Harris
- Department of Endocrinology and Diabetes, Queensland Children's Hospital, South Brisbane, QLD, Australia
| | - Andrew Cotterill
- Department of Endocrinology and Diabetes, Queensland Children's Hospital, South Brisbane, QLD, Australia
| | - John M Wentworth
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
- Department of Diabetes and Endocrinology, Royal Melbourne Hospital, Parkville, VIC, Australia
| | - Jennifer J Couper
- Department of Endocrinology and Diabetes, Women's and Children's Hospital, North Adelaide, SA, Australia
- Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
| | - Aveni Haynes
- Children's Diabetes Centre, Telethon Kids Institute, The University of Western Australia Perth, Crawley, WA, Australia
| | - Elizabeth A Davis
- Children's Diabetes Centre, Telethon Kids Institute, The University of Western Australia Perth, Crawley, WA, Australia
- Department of Endocrinology and Diabetes, Perth Children's Hospital, Nedlands, WA, Australia
- Centre for Child Health Research, University of Western Australia, Perth, WA, Australia
| | - Kate E Lomax
- Children's Diabetes Centre, Telethon Kids Institute, The University of Western Australia Perth, Crawley, WA, Australia
- Department of Endocrinology and Diabetes, Perth Children's Hospital, Nedlands, WA, 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, Brisbane, QLD, Australia
- Department of Chemical Pathology, Mater Pathology, South Brisbane, QLD, Australia
| |
Collapse
|
9
|
Wang D, Hou X, Huang J, Sun J, Kadowaki T, Lee MK, Jenkins AJ, Ji L. Incidence and trends of type 1 diabetes before and after 2000 in the Western Pacific Region: A systematic review and meta-analysis. Diabetes Res Clin Pract 2024; 207:111055. [PMID: 38104899 DOI: 10.1016/j.diabres.2023.111055] [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: 08/20/2023] [Revised: 12/09/2023] [Accepted: 12/13/2023] [Indexed: 12/19/2023]
Abstract
OBJECTIVES To undertake a systematic review of publications describing Type 1 diabetes (T1DM) incidence, trends over time and associated factors in the Western Pacific Region (WPR). METHODS As per the PROSPERO-registered (CRD42019122646) protocol English (MEDLINE, Embase, Global Health) and Chinese data-bases (China National Knowledge Infrastructure, VIP, Wanfang) from onset to 31/12/2019 were searched for T1DM incidence in the WPR. Country level data extracted included annual crude incidence rates by sex, number of new cases per annum (p.a.) and cumulatively, and the population at-risk. A meta-analysis for T1DM incidence was performed (by region and narrow age-bands, where possible) with subgroup analyses by time and by region. FINDINGS Forty-five population-based studies (21 from China), published 1973-2017, estimated T1DM incidence, mostly in youth, in 11 WPR countries. After 2000, mean annual T1DM incidence/100,000 person years aged 0-14 years ranged from 0.9 (95 % confidence intervals (CI), 0.6-1.3) in Fiji to 23.2 (95 % CI, 21.3-25.2) in Australia. The mean annual increase over time ranged from 2.8 % in Australia (1990-2002) to 14.2 % in Shanghai (1997-2011). T1DM incidence increased most in China (2.7-fold over 30-years) then Thailand (2-fold over 15-years). Most studies documented increasing incidence with age, though only two studies included people aged ≥ 20 years. Many, but not all studies reported significantly higher T1DM incidence in females vs. males. CONCLUSION T1DM incidence in the WPR is generally increasing, varying by age, sex, time and country. Results increase understanding of regional T1DM incidence and inform research and healthcare strategies.
Collapse
Affiliation(s)
- Du Wang
- The George Institute for Global Health, People's Republic of China
| | - Xiaoli Hou
- Department of Endocrinology and Metabolism, The Fifth Affiliated Hospital of Xinxiang Medical College, Xin Xiang 453100, People's Republic of China
| | - Juan Huang
- Department of Endocrinology and Metabolism, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, People's Republic of China
| | - Jianjing Sun
- Department of Endocrinology, Jining No.1 People's Hospital, Jining 272 011, Shandong, People's Republic of China
| | - Takashi Kadowaki
- Toranomon Hospital, The University of Tokyo, Minato-ku, Tokyo 105-8470, Japan
| | - Moon-Kyu Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Uijeongbu Eulji Medical Center, Eulji University School of Medicine, Uijeongbu, Republic of Korea
| | | | - Linong Ji
- Department of Endocrinology and Metabolism, Peking University People's Hospital, No. 11, Xizhimen Nan Da Jie, Xicheng District, Beijing 100044, People's Republic of China.
| |
Collapse
|
10
|
Sims EK, Kulkarni A, Hull A, Woerner SE, Cabrera S, Mastrandrea LD, Hammoud B, Sarkar S, Nakayasu ES, Mastracci TL, Perkins SM, Ouyang F, Webb-Robertson BJ, Enriquez JR, Tersey SA, Evans-Molina C, Long SA, Blanchfield L, Gerner EW, Mirmira RG, DiMeglio LA. Inhibition of polyamine biosynthesis preserves β cell function in type 1 diabetes. Cell Rep Med 2023; 4:101261. [PMID: 37918404 PMCID: PMC10694631 DOI: 10.1016/j.xcrm.2023.101261] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 07/18/2023] [Accepted: 10/05/2023] [Indexed: 11/04/2023]
Abstract
In preclinical models, α-difluoromethylornithine (DFMO), an ornithine decarboxylase (ODC) inhibitor, delays the onset of type 1 diabetes (T1D) by reducing β cell stress. However, the mechanism of DFMO action and its human tolerability remain unclear. In this study, we show that mice with β cell ODC deletion are protected against toxin-induced diabetes, suggesting a cell-autonomous role of ODC during β cell stress. In a randomized controlled trial (ClinicalTrials.gov: NCT02384889) involving 41 recent-onset T1D subjects (3:1 drug:placebo) over a 3-month treatment period with a 3-month follow-up, DFMO (125-1,000 mg/m2) is shown to meet its primary outcome of safety and tolerability. DFMO dose-dependently reduces urinary putrescine levels and, at higher doses, preserves C-peptide area under the curve without apparent immunomodulation. Transcriptomics and proteomics of DFMO-treated human islets exposed to cytokine stress reveal alterations in mRNA translation, nascent protein transport, and protein secretion. These findings suggest that DFMO may preserve β cell function in T1D through islet cell-autonomous effects.
Collapse
Affiliation(s)
- 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 46202, USA.
| | - Abhishek Kulkarni
- Kovler Diabetes Center and Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Audrey Hull
- 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 46202, USA; Nationwide Children's Hospital Pediatric Residency Program, Columbus, OH 43205, USA
| | - Stephanie E Woerner
- 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 46202, USA
| | - Susanne Cabrera
- Department of Pediatrics, Section of Endocrinology and Diabetes, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Lucy D Mastrandrea
- Division of Pediatric Endocrinology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Batoul Hammoud
- Department of Pediatrics, The University of Chicago, Chicago, IL 60637, USA
| | - Soumyadeep Sarkar
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Ernesto S Nakayasu
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Teresa L Mastracci
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Susan M Perkins
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Fangqian Ouyang
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | - Jacob R Enriquez
- Kovler Diabetes Center and Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Sarah A Tersey
- Kovler Diabetes Center and Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Carmella Evans-Molina
- 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 46202, USA; Department of Medicine and the Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Roudebush VA Medical Center, Indianapolis, IN 46202, USA
| | - S Alice Long
- Benaroya Research Institute, Center for Translational Immunology, Seattle, WA 98101, USA
| | - Lori Blanchfield
- Benaroya Research Institute, Center for Translational Immunology, Seattle, WA 98101, USA
| | | | - Raghavendra G Mirmira
- Kovler Diabetes Center and Department of Medicine, The University of Chicago, Chicago, IL 60637, USA; Department of Pediatrics, The University of Chicago, Chicago, IL 60637, USA.
| | - Linda A DiMeglio
- 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 46202, USA
| |
Collapse
|
11
|
Abstract
Despite major advances over the past decade, prevention and treatment of type 1 diabetes mellitus (T1DM) remain suboptimal, with large and unexplained variations in individual responses to interventions. The current classification schema for diabetes mellitus does not capture the complexity of this disease or guide clinical management effectively. One of the approaches to achieve the goal of applying precision medicine in diabetes mellitus is to identify endotypes (that is, well-defined subtypes) of the disease each of which has a distinct aetiopathogenesis that might be amenable to specific interventions. Here, we describe epidemiological, clinical, genetic, immunological, histological and metabolic differences within T1DM that, together, suggest heterogeneity in its aetiology and pathogenesis. We then present the emerging endotypes and their impact on T1DM prediction, prevention and treatment.
Collapse
Affiliation(s)
- Maria J Redondo
- Paediatric Diabetes & Endocrinology, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA.
| | - Noel G Morgan
- Exeter Centre of Excellence for Diabetes Research (EXCEED), Department of Clinical and Biomedical and Science, University of Exeter Medical School, Exeter, UK
| |
Collapse
|
12
|
Zong Y, Yu W, Hong H, Zhu Z, Xiao W, Wang K, Xu G. Ginsenoside Rg1 Improves Inflammation and Autophagy of the Pancreas and Spleen in Streptozotocin-Induced Type 1 Diabetic Mice. Int J Endocrinol 2023; 2023:3595992. [PMID: 36960388 PMCID: PMC10030220 DOI: 10.1155/2023/3595992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 01/16/2023] [Accepted: 01/23/2023] [Indexed: 03/17/2023] Open
Abstract
BACKGROUND Ginsenoside Rg1 (Rg1) is one of the key bioactive components of the precious Traditional Chinese Medicine that has been used to treat diabetes in China. Ginsenosides have been reported to protect diabetics from tissue damage, inflammation, and insulin resistance. Type 1 diabetes (T1D) is an organ-specific autoimmune disease that occurred frequently among adolescents over the world, its development was related to inflammation and β-cells immunodeficiency. The aim of this study is to explore the biological mechanism of Rg1 on inflammation and autophagy of β-cells in T1D and its therapeutic potential. METHODS The model of T1D mice was established by injecting Streptozotocin (STZ) (55 mg/kg) or citric acids once a day for 5 days and from the fourth day of injection, mice were administered with Rg1 (20 mg/kg) or saline by gavage once a day for 12 days. Hematoxylin-eosin staining, immunofluorescence, ELISA, quantitative real-time PCR, and Western blot were used to observe the histopathological changes, inflammatory factor levels, and autophagy markers after administration of ginsenoside Rg1. RESULTS Compared to the T1D mice, Rg1 improved the weight (p < 0.05) and blood glucose (p < 0.01) of mice, advanced the injury and apoptosis of β-cells in islets (p < 0.01), and markedly inhibited the protein expression degrees of CD45, CXCL16, ox-LDL, and TF in the pancreas and spleens (p < 0.01), also activated the degrees of insulin in serum (p < 0.01). Besides, in T1D mice' pancreas and spleen, Rg1 markedly repressed the IL-1β, TNF-α, and NOS2 mRNA levels (p < 0.05 or p < 0.01), inhibited the CXCL16, NF-κB, and TF proteins (p < 0.05 or p < 0.01), while elevating the ratio of LC3 II/I (p < 0.01) and P62 (p < 0.05) protein level. CONCLUSIONS This study proved that Rg1 protected mice against T1D possibly by improving islet injury and tissue inflammation, raising serum insulin, and tissue autophagy marker.
Collapse
Affiliation(s)
- Yi Zong
- Department of Radiology, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Weihua Yu
- Department of Gastroenterology, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Hanghang Hong
- Department of Ultrasound, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Zhiqiang Zhu
- Department of Clinical Laboratary, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Wenbo Xiao
- Department of Radiology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Kewu Wang
- Department of Radiology, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Guoqiang Xu
- Department of Gastroenterology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
13
|
Hofelich A, Marcus BA, Achenbach P. Früherkennung und Prävention des Typ-1-Diabetes. DIABETOL STOFFWECHS 2022. [DOI: 10.1055/a-0894-1860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
14
|
Nagy G, Szekely TE, Somogyi A, Herold M, Herold Z. New therapeutic approaches for type 1 diabetes: Disease-modifying therapies. World J Diabetes 2022; 13:835-850. [PMID: 36312000 PMCID: PMC9606789 DOI: 10.4239/wjd.v13.i10.835] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 08/08/2022] [Accepted: 09/15/2022] [Indexed: 02/05/2023] Open
Abstract
It has been 100 years since the first successful clinical use of insulin, yet it remains the only treatment option for type 1 diabetes mellitus (T1DM) patients. Advances in diabetes care, such as insulin analogue therapies and new devices, including continuous glucose monitoring with continuous subcutaneous insulin infusion have improved the quality of life of patients but have no impact on the pathogenesis of the disease. They do not eliminate long-term complications and require several lifestyle sacrifices. A more ideal future therapy for T1DM, instead of supplementing the insufficient hormone production (a consequence of β-cell destruction), would also aim to stop or slow down the destructive autoimmune process. The discovery of the autoimmune nature of type 1 diabetes mellitus has presented several targets by which disease progression may be altered. The goal of disease-modifying therapies is to target autoimmune mechanisms and prevent β-cell destruction. T1DM patients with better β-cell function have better glycemic control, reduced incidence of long-term complications and hypoglycemic episodes. Unfortunately, at the time symptomatic T1DM is diagnosed, most of the insulin secreting β cells are usually lost. Therefore, to maximize the salvageable β-cell mass by disease-modifying therapies, detecting autoimmune markers in an early, optimally presymptomatic phase of T1DM is of great importance. Disease-modifying therapies, such as immuno- and regenerative therapies are expected to take a relevant place in diabetology. The aim of this article was to provide a brief insight into the pathogenesis and course of T1DM and present the current state of disease-modifying therapeutic interventions that may impact future diabetes treatment.
Collapse
Affiliation(s)
- Geza Nagy
- Department of Internal Medicine and Hematology, Semmelweis University, Budapest H-1088, Hungary
| | - Tekla Evelin Szekely
- Department of Internal Medicine and Hematology, Semmelweis University, Budapest H-1088, Hungary
| | - Aniko Somogyi
- Department of Internal Medicine and Hematology, Semmelweis University, Budapest H-1088, Hungary
| | - Magdolna Herold
- Department of Internal Medicine and Hematology, Semmelweis University, Budapest H-1088, Hungary
| | - Zoltan Herold
- Division of Oncology, Department of Internal Medicine and Oncology, Semmelweis University, Budapest H-1083, Hungary
| |
Collapse
|
15
|
The relationship between GAD65 autoantibody and the risk of T1DM onset. J Diabetes Metab Disord 2022. [PMID: 36404832 PMCID: PMC9672278 DOI: 10.1007/s40200-022-01098-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Objectives Type 1 diabetes mellitus (T1DM) is a well-known autoimmune disease, characterized by β-cell destruction in pancreas islet cells, which results insulin deficiency and subsequent hyperglycemic sequelae. While there is screening for type 2 DM that leads to better glycemic control and outcome, the majority of T1DM patients are diagnosed when much of the pancreatic cells and their function are disturbed. The aim of this article is to present an overview of the effective factors in the positivity of Glutamic acid decarboxylase antibody )GADA( and identifying the high-risk individuals for T1DM. Methods We searched English literature available at National Library of Medicine via PubMed, and Google Scholar through December 2020. Finally, 79 papers have been included in the study. Studies were summarized based on the number of positive autoantibodies and onset of T1DM over time and GADA correlation with different variables. Conclusions GADA is an easy marker to measure that can be detected many months prior to the clinical presentation and remains positive even after early childhood.
Collapse
|
16
|
Dunger DB, Bruggraber SFA, Mander AP, Marcovecchio ML, Tree T, Chmura PJ, Knip M, Schulte AM, Mathieu C. INNODIA Master Protocol for the evaluation of investigational medicinal products in children, adolescents and adults with newly diagnosed type 1 diabetes. Trials 2022; 23:414. [PMID: 35585600 PMCID: PMC9116021 DOI: 10.1186/s13063-022-06259-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 03/30/2022] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND The INNODIA consortium has established a pan-European infrastructure using validated centres to prospectively evaluate clinical data from individuals with newly diagnosed type 1 diabetes combined with centralised collection of clinical samples to determine rates of decline in beta-cell function and identify novel biomarkers, which could be used for future stratification of phase 2 clinical trials. METHODS In this context, we have developed a Master Protocol, based on the "backbone" of the INNODIA natural history study, which we believe could improve the delivery of phase 2 studies exploring the use of single or combinations of Investigational Medicinal Products (IMPs), designed to prevent or reverse declines in beta-cell function in individuals with newly diagnosed type 1 diabetes. Although many IMPs have demonstrated potential efficacy in phase 2 studies, few subsequent phase 3 studies have confirmed these benefits. Currently, phase 2 drug development for this indication is limited by poor evaluation of drug dosage and lack of mechanistic data to understand variable responses to the IMPs. Identification of biomarkers which might permit more robust stratification of participants at baseline has been slow. DISCUSSION The Master Protocol provides (1) standardised assessment of efficacy and safety, (2) comparable collection of mechanistic data, (3) the opportunity to include adaptive designs and the use of shared control groups in the evaluation of combination therapies, and (4) benefits of greater understanding of endpoint variation to ensure more robust sample size calculations and future baseline stratification using existing and novel biomarkers.
Collapse
Affiliation(s)
- David B. Dunger
- Department of Paediatrics, University of Cambridge, Cambridge, UK
- Wellcome Trust-MRC Institute of Metabolic Sciences, University of Cambridge, Cambridge, UK
| | | | - Adrian P. Mander
- Centre for Trials Research, Cardiff University, Cardiff, UK
- NIHR Biomedical Research Centre, Guy’s and St Thomas’ NHS Foundation Trust and King’s College London, London, UK
| | | | - Timothy Tree
- Centre for Trials Research, Cardiff University, Cardiff, UK
- NIHR Biomedical Research Centre, Guy’s and St Thomas’ NHS Foundation Trust and King’s College London, London, UK
| | - Piotr Jaroslaw Chmura
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mikael Knip
- Pediatric Research Centre, Children’s Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | | | | |
Collapse
|
17
|
Triolo TM, Pyle L, Broncucia H, Armstrong T, Yu L, Gottlieb PA, Steck AK. Association of High-Affinity Autoantibodies With Type 1 Diabetes High-Risk HLA Haplotypes. J Clin Endocrinol Metab 2022; 107:e1510-e1517. [PMID: 34850014 PMCID: PMC8947772 DOI: 10.1210/clinem/dgab853] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Indexed: 11/19/2022]
Abstract
OBJECTIVE Electrochemiluminescence (ECL) assays are high-affinity autoantibody (Ab) tests that are more specific than Abs detected by traditional radiobinding assays (RBA) for risk screening and prediction of progression to type 1 diabetes. We sought to characterize the association of high-risk human leukocyte antigen (HLA) haplotypes and genotypes with ECL positivity and levels in relatives of individuals with type 1 diabetes. METHODS We analyzed 602 participants from the TrialNet Pathway to Prevention Study who were positive for at least 1 RBA diabetes-related Ab [glutamic acid decarboxylase autoantibodies (GADA) or insulin autoantibodies (IAA)] and for whom ECL and HLA data were available. ECL and RBA Ab levels were converted to SD units away from mean (z-scores) for analyses. RESULTS Mean age at initial visit was 19.4 ± 13.7 years; 344 (57.1%) were female and 104 (17.3%) carried the high-risk HLA-DR3/4*0302 genotype. At initial visit 424/602 (70.4%) participants were positive for either ECL-GADA or ECL-IAA, and 178/602 (29.6%) were ECL negative. ECL and RBA-GADA positivity were associated with both HLA-DR3 and DR4 haplotypes (all Ps < 0.05), while ECL and RBA-GADA z-score titers were higher in participants with HLA-DR3 haplotypes only (both Ps < 0.001). ECL-IAA (but not RBA-IAA) positivity was associated with the HLA-DR4 haplotype (P < 0.05). CONCLUSIONS ECL-GADA positivity is associated with the HLA-DR3 and HLA-DR4 haplotypes and levels are associated with the HLA-DR3 haplotype. ECL-IAA positivity is associated with HLA-DR4 haplotype. These studies further contribute to the understanding of genetic risk and islet autoimmunity endotypes in type 1 diabetes.
Collapse
Affiliation(s)
- Taylor M Triolo
- Barbara Davis Center for Childhood Diabetes, University of Colorado, Aurora, CO, USA
- Correspondence: Taylor M. Triolo, MD, University of Colorado Denver School of Medicine, Barbara Davis Center for Diabetes, 1775 Aurora Ct, MS #A140, Aurora, CO, USA 80045-2581.
| | - Laura Pyle
- Department of Pediatrics, University of Colorado, Aurora, CO, USA
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, CO, USA
| | - Hali Broncucia
- Barbara Davis Center for Childhood Diabetes, University of Colorado, Aurora, CO, USA
| | - Taylor Armstrong
- Barbara Davis Center for Childhood Diabetes, University of Colorado, Aurora, CO, USA
| | - Liping Yu
- Barbara Davis Center for Childhood Diabetes, University of Colorado, Aurora, CO, USA
| | - Peter A Gottlieb
- Barbara Davis Center for Childhood Diabetes, University of Colorado, Aurora, CO, USA
| | - Andrea K Steck
- Barbara Davis Center for Childhood Diabetes, University of Colorado, Aurora, CO, USA
| |
Collapse
|
18
|
Cetkovic-Cvrlje M, Rogan S, Barbaro E. Garcinia kola treatment exhibits immunomodulatory properties while not affecting type 1 diabetes development in an experimental mouse model. Int J Immunopathol Pharmacol 2022. [PMCID: PMC8777369 DOI: 10.1177/20587384211069831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Objective T cells orchestrate an inflammatory response that destroys pancreatic insulin-producing
β cells during the development of autoimmune type 1 diabetes (T1D). Garcinia
kola Heckel (GK) is a plant widely exploited in West African traditional
medicine. Some of the therapeutic effects of GK nut’s extract (GKE) have been suggested
to be due to its anti-inflammatory potential. Since GKE has never been investigated in a
T1D experimental model, nor in the T cells’ context, we aimed to determine whether GKE
exhibits antidiabetic properties and affects T cells by its anticipated
anti-inflammatory action. Methods The effect of aqueous GKE (aGKE) ingestion, 100 mg/kg daily by drinking water over the
period of 6 weeks, has been tested in a low-dose streptozotocin-induced (LDSTZ) mouse
model of autoimmune T1D. T cells were studied in vitro and in
vivo in mice treated by aGKE. Results The results showed that aGKE treatment, which started a week before induction of
disease, neither delayed the development of T1D, nor reduced glycemia severity.
Interestingly, aGKE treatment did affect T cells and their function, significantly
decreasing the frequency of helper (TH) and cytotoxic (TC) T
cells, while elevating the levels of pro-inflammatory cytokines, TNF-α, IL-6, and IFN-γ,
and suppressing IL-2. Conclusion In conclusion, our results did not confirm the antidiabetic property of GKE, while
suggesting its therapeutic exploration in TH2-dependent pathologies that
benefit from an aggravated TH1 response, such as allergies.
Collapse
Affiliation(s)
- Marina Cetkovic-Cvrlje
- Department of Biological Sciences andImmunology Laboratory, St Cloud State University, St Cloud, MN, USA
| | - Shana Rogan
- Department of Biological Sciences andImmunology Laboratory, St Cloud State University, St Cloud, MN, USA
| | - Emily Barbaro
- Department of Biological Sciences andImmunology Laboratory, St Cloud State University, St Cloud, MN, USA
| |
Collapse
|
19
|
He LP, Song YX, Zhu T, Gu W, Liu CW. Progress in the Relationship between Vitamin D Deficiency and the Incidence of Type 1 Diabetes Mellitus in Children. J Diabetes Res 2022; 2022:5953562. [PMID: 36090587 PMCID: PMC9463035 DOI: 10.1155/2022/5953562] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/03/2022] [Accepted: 08/12/2022] [Indexed: 11/24/2022] Open
Abstract
Type 1 diabetes mellitus (T1DM) is an autoimmune disease, due to a large number of islet β cells damaged, resulting in an absolute lack of insulin, ultimately relying on insulin therapy. Vitamin D is a fat-soluble sterol derivative that not only participates in calcium and phosphorus metabolism but also acts as an immunomodulatory role by binding to nuclear vitamin D receptors to regulate the expression of transcription factors. Increasing evidence has shown that vitamin D has immunoregulation and anti-inflammatory effects, and it may play a role in T cell regulatory responses due to downregulation in the expression of cathepsin G and inhibition of CD4+ T cell activation and protection of β cells from immune attack and is beneficial in decreasing oxidative stress in T1DM patients. Epidemiologic evidence demonstrates involvement of vitamin D deficiency in T1DM pathogenesis, with the immune system improperly targeting and destroying its own islet β cells. In addition, polymorphisms in genes critical for vitamin D metabolism may increase the risk of islet autoimmunity and T1DM. In this paper, the relationship between vitamin D deficiency and the molecular mechanism of T1DM was discussed.
Collapse
Affiliation(s)
- Lian-Ping He
- School of Medicine, Taizhou University, Jiaojiang, 318000 Zhejiang, China
| | - Yu-Xin Song
- School of Medicine, Taizhou University, Jiaojiang, 318000 Zhejiang, China
| | - Ting Zhu
- Children's Hospital of Nanjing Medical University, Nanjing, 210008 Jiangsu, China
| | - Wei Gu
- Children's Hospital of Nanjing Medical University, Nanjing, 210008 Jiangsu, China
| | - Chang-Wei Liu
- Children's Hospital of Nanjing Medical University, Nanjing, 210008 Jiangsu, China
| |
Collapse
|
20
|
Tatovic D, Dayan CM. Replacing insulin with immunotherapy: Time for a paradigm change in Type 1 diabetes. Diabet Med 2021; 38:e14696. [PMID: 34555209 DOI: 10.1111/dme.14696] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 09/22/2021] [Indexed: 12/17/2022]
Abstract
For almost a hundred years, the management of Type 1 diabetes has not advanced beyond insulin replacement. However, insulin does not provide satisfactory glycaemic control in the majority of individuals and there remains a major unmet need for novel treatments for Type 1 diabetes. Immunomodulation to preserve beta-cell function offers the prospect of making treatment with insulin easier and/or preventing the need for insulin, particularly when it comes to novel low-risk immunotherapies. Led by the concept that the best insulin-producing cell is a patient's own beta-cell, the Type 1 diabetes scientific community has a challenging task ahead-to fundamentally change the management of this devastating disease by using low-risk immunotherapy to preserve endogenous beta-cell function and make metabolic control substantially easier. In that way, insulin and/or beta-cell replacement (stem cell or transplantation) should in the future be considered rescue therapies reserved for delayed presentations.
Collapse
Affiliation(s)
- Danijela Tatovic
- Diabetes and Autoimmunity Research Group, Cardiff University School of Medicine, Cardiff, UK
| | - Colin M Dayan
- Diabetes and Autoimmunity Research Group, Cardiff University School of Medicine, Cardiff, UK
| |
Collapse
|
21
|
Vallianou NG, Stratigou T, Geladari E, Tessier CM, Mantzoros CS, Dalamaga M. Diabetes type 1: Can it be treated as an autoimmune disorder? Rev Endocr Metab Disord 2021; 22:859-876. [PMID: 33730229 DOI: 10.1007/s11154-021-09642-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/03/2021] [Indexed: 02/06/2023]
Abstract
Type 1 Diabetes Mellitus (T1DM) is characterized by progressive autoimmune-mediated destruction of the pancreatic beta-cells leading to insulin deficiency and hyperglycemia. It is associated with significant treatment burden and necessitates life-long insulin therapy. The role of immunotherapy in the prevention and management of T1DM is an evolving area of interest which has the potential to alter the natural history of this disease.In this review, we give insight into recent clinical trials related to the use of immunotherapeutic approaches for T1DM, such as proinflammatory cytokine inhibition, cell-depletion and cell-therapy approaches, autoantigen-specific treatments and stem cell therapies. We highlight the timing of intervention, aspects of therapy including adverse effects and the emergence of a novel lymphocyte crucial in T1DM autoimmunity. We also discuss the role of cardiac autoimmunity and its link to excess CVD risk in T1DM.We conclude that significant advances have been made in development of immunotherapeutic targets and agents for the treatment and prevention of T1DM. These immune-based therapies promise preservation of beta-cells and decreasing insulin dependency. In their current state, immunotherapeutic approaches cannot yet halt the progression from a preclinical state to overt T1DM nor can they replace standard insulin therapy in existing T1DM. It remains to be seen whether immunotherapy will ultimately play a key role in the prevention of progression to overt T1DM and whether it may find a place in our therapeutic armamentarium to improve clinical outcomes and quality of life in established T1DM.
Collapse
Affiliation(s)
- Natalia G Vallianou
- Department of Internal Medicine, Evangelismos General Hospital, 45-47 Ipsilantou str, 10676, Athens, Greece
| | - Theodora Stratigou
- Department of Endocrinology, Diabetes and Metabolic Diseases, Evangelismos General Hospital, 45-47 Ipsilantou str, 10676, Athens, Greece
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias, 11527, Athens, Goudi, Greece
| | - Eleni Geladari
- Department of Internal Medicine, Evangelismos General Hospital, 45-47 Ipsilantou str, 10676, Athens, Greece
| | - Christopher M Tessier
- Endocrinology Section, VA Boston Healthcare System, 1400 VFW Parkway West Roxbury, Boston, MA, 02132, USA.
| | - Christos S Mantzoros
- Endocrinology Section, VA Boston Healthcare System, 1400 VFW Parkway West Roxbury, Boston, MA, 02132, USA
| | - Maria Dalamaga
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias, 11527, Athens, Goudi, Greece
| |
Collapse
|
22
|
Dayan CM, Besser REJ, Oram RA, Hagopian W, Vatish M, Bendor-Samuel O, Snape MD, Todd JA. Preventing type 1 diabetes in childhood. Science 2021; 373:506-510. [PMID: 34326231 DOI: 10.1126/science.abi4742] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Type 1 diabetes (T1D) is an autoimmune disease in which the insulin-producing β cells of the pancreas are destroyed by T lymphocytes. Recent studies have demonstrated that monitoring for pancreatic islet autoantibodies, combined with genetic risk assessment, can identify most children who will develop T1D when they still have sufficient β cell function to control glucose concentrations without the need for insulin. In addition, there has been recent success in secondary prevention using immunotherapy to delay the progression of preclinical disease, and primary prevention approaches to inhibiting the initiating autoimmune process have entered large-scale clinical trials. By changing the focus of T1D management from late diagnosis and insulin replacement to early diagnosis and β cell preservation, we can anticipate a future without the need for daily insulin injections for children with T1D.
Collapse
Affiliation(s)
- Colin M Dayan
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, National Institute for Health Research (NIHR) Biomedical Research Centre, University of Oxford, Oxford, UK. .,Cardiff University School of Medicine, Cardiff, UK
| | - Rachel E J Besser
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, National Institute for Health Research (NIHR) Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Richard A Oram
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK
| | | | - Manu Vatish
- Department of Women's and Reproductive Health, University of Oxford, Oxford, UK
| | | | - Matthew D Snape
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, and Oxford NIHR Biomedical Research Centre, Oxford, UK
| | - John A Todd
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, National Institute for Health Research (NIHR) Biomedical Research Centre, University of Oxford, Oxford, UK
| |
Collapse
|
23
|
Pinheiro MM, Pinheiro FMM, Diniz SN, Fabbri A, Infante M. Combination of vitamin D and dipeptidyl peptidase-4 inhibitors (VIDPP-4i) as an immunomodulation therapy for autoimmune diabetes. Int Immunopharmacol 2021; 95:107518. [PMID: 33756226 DOI: 10.1016/j.intimp.2021.107518] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/15/2021] [Accepted: 02/16/2021] [Indexed: 12/18/2022]
Abstract
Type 1 diabetes (T1D) and latent autoimmune diabetes in adults (LADA) represent the most common types of autoimmune diabetes and are characterized by different age of onset, degrees of immune-mediated destruction of pancreatic beta cells and rates of disease progression towards insulin dependence. Several immunotherapies aimed to counteract autoimmune responses against beta cells and preserve beta-cell function are currently being investigated, particularly in T1D. Preliminary findings suggest a potential role of combination therapy with vitamin D and dipeptidyl peptidase-4 (DPP-4) inhibitors (VIDPP-4i) in preserving beta-cell function in autoimmune diabetes. This manuscript aims to provide a comprehensive overview of the immunomodulatory properties of vitamin D and DPP-4 inhibitors, as well as the rationale for investigation of their combined use as an immunomodulation therapy for autoimmune diabetes.
Collapse
Affiliation(s)
- Marcelo Maia Pinheiro
- UNIVAG, University Center, Dom Orlando Chaves Ave, 2655 - Cristo Rei, Várzea Grande, 78118-000 Mato Grosso, Brazil; Universidade Anhanguera de São Paulo - SP, 3305, Raimundo Pereira de Magalhães Ave., Pirituba, São Paulo, 05145-200 São Paulo, Brazil.
| | - Felipe Moura Maia Pinheiro
- Hospital de Base, Faculdade de Medicina de São José do Rio Preto FAMERP - SP, 5546, Brigadeiro Faria Lima Ave, Vila São Pedro, São José do Rio Preto, 15015-500 São Paulo, Brazil
| | - Susana Nogueira Diniz
- Universidade Anhanguera de São Paulo - SP, 3305, Raimundo Pereira de Magalhães Ave., Pirituba, São Paulo, 05145-200 São Paulo, Brazil
| | - Andrea Fabbri
- Diabetes Research Institute Federation (DRIF), Division of Endocrinology and Diabetes, CTO Andrea Alesini Hospital, ASL Roma 2, Department of Systems Medicine, University of Rome Tor Vergata, Via San Nemesio 21, 00145 Rome, Italy
| | - Marco Infante
- Diabetes Research Institute Federation (DRIF), Division of Endocrinology and Diabetes, CTO Andrea Alesini Hospital, ASL Roma 2, Department of Systems Medicine, University of Rome Tor Vergata, Via San Nemesio 21, 00145 Rome, Italy; UniCamillus, Saint Camillus International University of Health Sciences, Via di Sant'Alessandro, 8, 00131 Rome, Italy; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Via San Nemesio 21, 00145 Rome, Italy.
| |
Collapse
|
24
|
Patel SK, Ma CS, Fourlanos S, Greenfield JR. Autoantibody-Negative Type 1 Diabetes: A Neglected Subtype. Trends Endocrinol Metab 2021; 32:295-305. [PMID: 33712367 DOI: 10.1016/j.tem.2021.02.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/03/2021] [Accepted: 02/05/2021] [Indexed: 12/16/2022]
Abstract
Up to 15% of individuals with a clinical phenotype of type 1 diabetes (T1D) do not have evidence of seropositivity for pancreatic islet autoantibodies. On this basis, they are classified as nonimmune or idiopathic, and remain an understudied population, as they are excluded from T1D immunomodulatory trials. Our limited understanding of the disease aetiopathogenesis in autoantibody-negative T1D hinders our ability to improve diagnostic pathways and discover novel therapeutic agents; particularly as we progress towards an era of precision medicine. This review summarises the current understanding and challenges in studying autoantibody-negative T1D. We review the literature regarding T1D classification, and the role of autoimmunity and defects in the immunogenic pathway that may distinguish autoantibody-positive and -negative T1D.
Collapse
Affiliation(s)
- Shivani K Patel
- Diabetes and Metabolism, Garvan Institute of Medical Research, Sydney, NSW, Australia; Department of Diabetes and Endocrinology, St. Vincent's Hospital, Sydney, NSW, Australia; St. Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW, Australia
| | - Cindy S Ma
- St. Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW, Australia; Human Immune Disorders, Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Spiros Fourlanos
- Department of Diabetes and Endocrinology, Royal Melbourne Hospital, Melbourne, VIC, Australia
| | - Jerry R Greenfield
- Diabetes and Metabolism, Garvan Institute of Medical Research, Sydney, NSW, Australia; Department of Diabetes and Endocrinology, St. Vincent's Hospital, Sydney, NSW, Australia; St. Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW, Australia.
| |
Collapse
|
25
|
Long SA, Speake C. Combination therapy in recent-onset type 1 diabetes. Lancet Diabetes Endocrinol 2021; 9:191-193. [PMID: 33662335 DOI: 10.1016/s2213-8587(21)00044-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 02/05/2021] [Indexed: 12/16/2022]
Affiliation(s)
- S Alice Long
- Center for Translational Immunology, Benaroya Research Institute at Virginia Mason, Seattle, WA 98101-2795, USA.
| | - Cate Speake
- Center for Interventional Immunology, Benaroya Research Institute at Virginia Mason, Seattle, WA 98101-2795, USA
| |
Collapse
|
26
|
Joshi K, Cameron F, Tiwari S, Mannering SI, Elefanty AG, Stanley EG. Modeling Type 1 Diabetes Using Pluripotent Stem Cell Technology. Front Endocrinol (Lausanne) 2021; 12:635662. [PMID: 33868170 PMCID: PMC8047192 DOI: 10.3389/fendo.2021.635662] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 03/03/2021] [Indexed: 12/26/2022] Open
Abstract
Induced pluripotent stem cell (iPSC) technology is increasingly being used to create in vitro models of monogenic human disorders. This is possible because, by and large, the phenotypic consequences of such genetic variants are often confined to a specific and known cell type, and the genetic variants themselves can be clearly identified and controlled for using a standardized genetic background. In contrast, complex conditions such as autoimmune Type 1 diabetes (T1D) have a polygenic inheritance and are subject to diverse environmental influences. Moreover, the potential cell types thought to contribute to disease progression are many and varied. Furthermore, as HLA matching is critical for cell-cell interactions in disease pathogenesis, any model that seeks to test the involvement of particular cell types must take this restriction into account. As such, creation of an in vitro model of T1D will require a system that is cognizant of genetic background and enables the interaction of cells representing multiple lineages to be examined in the context of the relevant environmental disease triggers. In addition, as many of the lineages critical to the development of T1D cannot be easily generated from iPSCs, such models will likely require combinations of cell types derived from in vitro and in vivo sources. In this review we imagine what an ideal in vitro model of T1D might look like and discuss how the required elements could be feasibly assembled using existing technologies. We also examine recent advances towards this goal and discuss potential uses of this technology in contributing to our understanding of the mechanisms underlying this autoimmune condition.
Collapse
Affiliation(s)
- Kriti Joshi
- Department of Endocrinology and Metabolism, All India Institute of Medical Sciences Rishikesh, Uttarakhand, India
- Department of Molecular Medicine & Biotechnology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, India
- Department of Cell Biology, Murdoch Children’s Research Institute, Parkville, Vic, Australia
| | - Fergus Cameron
- Department of Cell Biology, Murdoch Children’s Research Institute, Parkville, Vic, Australia
- Department of Endocrinology and Diabetes, The Royal Children’s Hospital, Parkville, Vic, Australia
- Department of Paediatrics, University of Melbourne, Parkville, Vic, Australia
| | - Swasti Tiwari
- Department of Molecular Medicine & Biotechnology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, India
| | - Stuart I. Mannering
- Immunology and Diabetes Unit, St. Vincent’s Institute of Medical Research, Fitzroy, Vic, Australia
| | - Andrew G. Elefanty
- Department of Cell Biology, Murdoch Children’s Research Institute, Parkville, Vic, Australia
- Department of Paediatrics, University of Melbourne, Parkville, Vic, Australia
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Vic, Australia
| | - Edouard G. Stanley
- Department of Cell Biology, Murdoch Children’s Research Institute, Parkville, Vic, Australia
- Department of Paediatrics, University of Melbourne, Parkville, Vic, Australia
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Vic, Australia
| |
Collapse
|
27
|
Papachrisanthou MM, Fuller KM. Pediatric Screenings: Helpful or Hinderance? J Nurse Pract 2021. [DOI: 10.1016/j.nurpra.2020.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
28
|
Novelli M, Masiello P, Beffy P, Menegazzi M. Protective Role of St. John's Wort and Its Components Hyperforin and Hypericin against Diabetes through Inhibition of Inflammatory Signaling: Evidence from In Vitro and In Vivo Studies. Int J Mol Sci 2020; 21:E8108. [PMID: 33143088 PMCID: PMC7662691 DOI: 10.3390/ijms21218108] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/26/2020] [Accepted: 10/27/2020] [Indexed: 02/06/2023] Open
Abstract
Diabetes mellitus is a very common chronic disease with progressively increasing prevalence. Besides the well-known autoimmune and inflammatory pathogenesis of type 1 diabetes, in many people, metabolic changes and inappropriate lifestyle favor a subtle chronic inflammatory state that contributes to development of insulin resistance and progressive loss of β-cell function and mass, eventually resulting in metabolic syndrome or overt type 2 diabetes. In this paper, we review the anti-inflammatory effects of the extract of Hypericum perforatum L. (St. John's wort, SJW) and its main active ingredients firstly in representative pathological situations on inflammatory basis and then in pancreatic β cells and in obese or diabetic animal models. The simultaneous and long-lasting inhibition of signal transducer and activator of transcription (STAT)-1, nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and mitogen-activated protein kinases (MAPKs)/c-jun N-terminal kinase (JNK) signaling pathways involved in pro-inflammatory cytokine-induced β-cell dysfunction/death and insulin resistance make SJW particularly suitable for both preventive and therapeutic use in metabolic diseases. Hindrance of inflammatory cytokine signaling is likely dependent on the hyperforin content of SJW extract, but recent data reveal that hypericin can also exert relevant protective effects, mediated by activation of the cyclic adenosine monophosphate (cAMP)/protein kinase cAMP-dependent (PKA)/adenosine monophosphate activated protein kinase (AMPK) pathway, against high-fat-diet-induced metabolic abnormalities. Actually, the mechanisms of action of the two main components of SJW appear complementary, strengthening the efficacy of the plant extract. Careful quantitative analysis of SJW components and suitable dosage, with monitoring of possible drug-drug interaction in a context of remarkable tolerability, are easily achievable pre-requisites for forthcoming clinical applications.
Collapse
Affiliation(s)
- Michela Novelli
- Department of Translational Research and New Technologies in Medicine and Surgery, School of Medicine, University of Pisa, 56126 Pisa, Italy
| | - Pellegrino Masiello
- Department of Translational Research and New Technologies in Medicine and Surgery, School of Medicine, University of Pisa, 56126 Pisa, Italy
| | - Pascale Beffy
- Institute of Clinical Physiology, CNR, 56124 Pisa, Italy;
| | - Marta Menegazzi
- Department of Neuroscience, Biomedicine and Movement Sciences, Biochemistry Section, School of Medicine, University of Verona, 37134 Verona, Italy;
| |
Collapse
|
29
|
Li L, Liu S, Yu J. Autoimmune thyroid disease and type 1 diabetes mellitus: same pathogenesis; new perspective? Ther Adv Endocrinol Metab 2020; 11:2042018820958329. [PMID: 32973994 PMCID: PMC7493255 DOI: 10.1177/2042018820958329] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 08/20/2020] [Indexed: 12/14/2022] Open
Abstract
Autoimmune thyroid disease (AITD) and type 1 diabetes mellitus (T1DM) are two common autoimmune diseases that can occur concomitantly. In general, patients with diabetes have a high risk of AITD. It has been proposed that a complex genetic basis together with multiple nongenetic factors make a variable contribution to the pathogenesis of T1DM and AITD. In this paper, we summarize current knowledge in the field regarding potential pathogenic factors of T1DM and AITD, including human leukocyte antigen, autoimmune regulator, lymphoid protein tyrosine phosphatase, forkhead box protein P3, cytotoxic T lymphocyte-associated antigen, infection, vitamin D deficiency, and chemokine (C-X-C motif) ligand. These findings offer an insight into future immunotherapy for autoimmune diseases.
Collapse
Affiliation(s)
- Liyan Li
- Department of Endocrinology, First People’s Hospital of Jinan, Jinan, People’s Republic of China
| | - Shudong Liu
- Department of Endocrinology, Shandong Rongjun General Hospital, Jinan, People’s Republic of China
| | - Junxia Yu
- Department of Endocrinology, Tengzhou Central People’s Hospital, 181 Xingtan Road, Tengzhou, Shandong Province, 277500, People’s Republic of China
| |
Collapse
|
30
|
Infante M, Ricordi C, Fabbri A. Antihyperglycemic properties of hydroxychloroquine in patients with diabetes: Risks and benefits at the time of COVID-19 pandemic. J Diabetes 2020; 12:659-667. [PMID: 32401405 PMCID: PMC7272905 DOI: 10.1111/1753-0407.13053] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 05/10/2020] [Accepted: 05/12/2020] [Indexed: 12/22/2022] Open
Abstract
The antimalarial drug hydroxychloroquine (HCQ) has long been used as a disease-modifying antirheumatic drug for the treatment of several inflammatory rheumatic diseases. Over the last three decades, various studies have shown that HCQ also plays a role in the regulation of glucose homeostasis. Although the mechanisms of action underlying the glucose-lowering properties of HCQ are still not entirely clear, evidence suggests that this drug may exert multifaceted effects on glucose regulation, including improvement of insulin sensitivity, increase of insulin secretion, reduction of hepatic insulin clearance, and reduction of systemic inflammation. Preliminary studies have shown the safety and efficacy of HCQ (at a dose ranging from 400 to 600 mg/day) in patients with type 2 diabetes over a short-term period. In 2014, HCQ has been approved in India as an add-on hypoglycemic agent for patients with uncontrolled type 2 diabetes. However, large randomized controlled trials are needed to establish the safety and efficacy profile of HCQ in patients with type 2 diabetes over a long-term period. With regard to the COVID-19 pandemic, several medications (including HCQ) have been used as off-label drugs because of the lack of proven effective therapies. However, emerging evidence shows limited benefit from HCQ use in COVID-19 in general. The aim of this manuscript is to comprehensively summarize the current knowledge on the antihyperglycemic properties of HCQ and to critically evaluate the potential risks and benefits related to HCQ use in patients with diabetes, even in light of the current pandemic scenario.
Collapse
Affiliation(s)
- Marco Infante
- Endocrine Unit, CTO Hospital ‐ ASL Roma 2, Department of Systems MedicineUniversity of Rome Tor VergataRomeItaly
- Diabetes Research Institute (DRI), University of Miami Miller School of MedicineMiamiFlorida
- Diabetes Research Institute Federation, DRIF (Rome, Miami)MiamiFlorida
| | - Camillo Ricordi
- Diabetes Research Institute (DRI), University of Miami Miller School of MedicineMiamiFlorida
- Diabetes Research Institute Federation, DRIF (Rome, Miami)MiamiFlorida
| | - Andrea Fabbri
- Endocrine Unit, CTO Hospital ‐ ASL Roma 2, Department of Systems MedicineUniversity of Rome Tor VergataRomeItaly
- Diabetes Research Institute Federation, DRIF (Rome, Miami)MiamiFlorida
| |
Collapse
|
31
|
Curtin F, Champion B, Davoren P, Duke S, Ekinci EI, Gilfillan C, Morbey C, Nathow T, O'Moore-Sullivan T, O'Neal D, Roberts A, Stranks S, Stuckey B, Vora P, Malpass S, Lloyd D, Maëstracci-Beard N, Buffet B, Kornmann G, Bernard C, Porchet H, Simpson R. A safety and pharmacodynamics study of temelimab, an antipathogenic human endogenous retrovirus type W envelope monoclonal antibody, in patients with type 1 diabetes. Diabetes Obes Metab 2020; 22:1111-1121. [PMID: 32077207 DOI: 10.1111/dom.14010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 02/14/2020] [Accepted: 02/17/2020] [Indexed: 12/15/2022]
Abstract
AIM To report the first study of temelimab, a monoclonal antibody neutralizing the pathogenic human endogenous retrovirus type W envelope, in patients with type 1 diabetes (T1D). MATERIALS AND METHODS This double-blind, placebo-controlled, randomized clinical trial recruited adult patients with T1D within 4 years postdiagnosis and remaining C-peptide secretion. Sixty-four patients were randomized (2:1) to monthly temelimab 6 mg/kg or placebo during 24 weeks followed by a 24-week, open-label extension, during which all patients received temelimab. The primary objective was the safety and tolerability of temelimab. The secondary objective was to assess the pharmacodynamics response such as C-peptide levels, insulin use, HbA1c, hypoglycaemia and autoantibodies. RESULTS Temelimab was well tolerated without any group difference in the frequency or severity of adverse events. Concerning exploratory endpoints, there was no difference in the levels of C-peptide, insulin use or HbA1c between treatment groups at weeks 24 and 48. The frequency of hypoglycaemia events was reduced with temelimab (P = 0.0004) at week 24 and the level of anti-insulin antibodies was lower with temelimab (P < 0.01); the other autoantibodies did not differ between groups. CONCLUSIONS Temelimab appeared safe in patients with T1D. Pharmacodynamics signals (hypoglycaemia and anti-insulin antibodies) under temelimab were observed. Markers of β-cell functions were not modified by treatment. These results need to be further explored in younger patients with T1D with earlier disease onset.
Collapse
Affiliation(s)
- Francois Curtin
- GeNeuro SA, Geneva, Switzerland
- Division of Clinical Pharmacology and Toxicology, Rue Perret-Gentil, University of Geneva, Geneva, Switzerland
| | - Bernard Champion
- Department of Clinical Medicine, Faculty of Medicine and Health Sciences, Macquarie University, Macquarie Park, New South Wales, Australia
| | - Peter Davoren
- Gold Coast Hospital, Diabetes and Endocrinology, Southport, Queensland, Australia
| | - Sally Duke
- Department of Diabetes Endocrinology and Metabolism, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| | - Elif I Ekinci
- Department of Medicine, Austin Health and The University of Melbourne, Heidelberg Heights, Victoria, Australia
| | - Chris Gilfillan
- Eastern Clinical Research Unit, Eastern Health and Monash University, Box Hill, Victoria, Australia
| | | | - Thomas Nathow
- Ipswich Research Institute, Ipswich, Queensland, Australia
| | | | - David O'Neal
- St. Vincent's Hospital, Department of Medicine, Fitzroy, Victoria, Australia
| | - Adam Roberts
- Barwon Health, Department of Endocrinology, Geelong, Victoria, Australia
| | - Stephen Stranks
- Southern Adelaide Diabetes & Endocrine Services, Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Bronwyn Stuckey
- Keogh Institute for Medical Research, Queen Elizabeth II Medical Centre, Nedlands,, Western Australia, Australia
| | - Parind Vora
- Division of Medicine, Lyell McEwin Hospital, Elizabeth Vale, South Australia, Australia
| | - Sam Malpass
- Southern Star Research, Gordon, New South Wales, Australia
| | - David Lloyd
- Southern Star Research, Gordon, New South Wales, Australia
| | | | | | | | | | - Hervé Porchet
- GeNeuro SA, Geneva, Switzerland
- Department of Pharmacology, University of Pretoria, Pretoria, South Africa
| | - Richard Simpson
- Eastern Clinical Research Unit, Eastern Health and Monash University, Box Hill, Victoria, Australia
| |
Collapse
|
32
|
Koufakis T, Katsiki N, Zebekakis P, Dimitriadis G, Kotsa K. Therapeutic approaches for latent autoimmune diabetes in adults: One size does not fit all. J Diabetes 2020; 12:110-118. [PMID: 31449359 DOI: 10.1111/1753-0407.12982] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 08/02/2019] [Accepted: 08/22/2019] [Indexed: 12/20/2022] Open
Abstract
Recent advances in the understanding of latent autoimmune diabetes in adults (LADA) pathophysiology make it increasingly evident that people with LADA comprise a heterogenous group of patients. This makes the establishment of a standard treatment algorithm challenging. On top of its glucose-lowering action, insulin may exert anti-inflammatory effects, rendering it an attractive therapeutic choice for a type of diabetes in which autoinflammation and beta cell insufficiency play major pathogenetic roles. However, there is growing evidence that other antidiabetic drugs, such as metformin, dipeptidyl peptidase-4 inhibitors, glucagon-like peptide-1 receptor agonists, and thiazolidinediones, might have a role in optimizing glycemic control and preserving beta cell function in individuals with LADA, either alone or in combination with insulin. Although most of these drugs have been routinely used in the daily clinical setting for years, large prospective randomized trials are needed to assess whether they are capable of delaying progression to insulin dependence as well as their effects on diabetic complications. The aim of the present review is to discuss the current state and future perspectives of LADA therapy, emphasizing the need for individualized and patient-centered therapeutic approaches.
Collapse
Affiliation(s)
- Theocharis Koufakis
- Division of Endocrinology and Metabolism and Diabetes Center, First Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, AHEPA University Hospital, Thessaloniki, Greece
| | - Niki Katsiki
- Division of Endocrinology and Metabolism and Diabetes Center, First Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, AHEPA University Hospital, Thessaloniki, Greece
| | - Pantelis Zebekakis
- Division of Endocrinology and Metabolism and Diabetes Center, First Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, AHEPA University Hospital, Thessaloniki, Greece
| | - George Dimitriadis
- Research Institute and Diabetes Center, Second Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, "Attikon" University Hospital, Athens, Greece
| | - Kalliopi Kotsa
- Division of Endocrinology and Metabolism and Diabetes Center, First Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, AHEPA University Hospital, Thessaloniki, Greece
| |
Collapse
|
33
|
Battaglia M, Ahmed S, Anderson MS, Atkinson MA, Becker D, Bingley PJ, Bosi E, Brusko TM, DiMeglio LA, Evans-Molina C, Gitelman SE, Greenbaum CJ, Gottlieb PA, Herold KC, Hessner MJ, Knip M, Jacobsen L, Krischer JP, Long SA, Lundgren M, McKinney EF, Morgan NG, Oram RA, Pastinen T, Peters MC, Petrelli A, Qian X, Redondo MJ, Roep BO, Schatz D, Skibinski D, Peakman M. Introducing the Endotype Concept to Address the Challenge of Disease Heterogeneity in Type 1 Diabetes. Diabetes Care 2020; 43:5-12. [PMID: 31753960 PMCID: PMC6925574 DOI: 10.2337/dc19-0880] [Citation(s) in RCA: 230] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 10/14/2019] [Indexed: 02/06/2023]
Abstract
The clinical diagnosis of new-onset type 1 diabetes has, for many years, been considered relatively straightforward. Recently, however, there is increasing awareness that within this single clinical phenotype exists considerable heterogeneity: disease onset spans the complete age range; genetic susceptibility is complex; rates of progression differ markedly, as does insulin secretory capacity; and complication rates, glycemic control, and therapeutic intervention efficacy vary widely. Mechanistic and immunopathological studies typically show considerable patchiness across subjects, undermining conclusions regarding disease pathways. Without better understanding, type 1 diabetes heterogeneity represents a major barrier both to deciphering pathogenesis and to the translational effort of designing, conducting, and interpreting clinical trials of disease-modifying agents. This realization comes during a period of unprecedented change in clinical medicine, with increasing emphasis on greater individualization and precision. For complex disorders such as type 1 diabetes, the option of maintaining the "single disease" approach appears untenable, as does the notion of individualizing each single patient's care, obliging us to conceptualize type 1 diabetes less in terms of phenotypes (observable characteristics) and more in terms of disease endotypes (underlying biological mechanisms). Here, we provide our view on an approach to dissect heterogeneity in type 1 diabetes. Using lessons from other diseases and the data gathered to date, we aim to delineate a roadmap through which the field can incorporate the endotype concept into laboratory and clinical practice. We predict that such an effort will accelerate the implementation of precision medicine and has the potential for impact on our approach to translational research, trial design, and clinical management.
Collapse
Affiliation(s)
- Manuela Battaglia
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Hospital, Milan, Italy
| | | | - Mark S Anderson
- Diabetes Center, University of California, San Francisco, San Francisco, CA
| | - Mark A Atkinson
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL
| | - Dorothy Becker
- Division of Endocrinology and Diabetes, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA
| | - Polly J Bingley
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, U.K
| | - Emanuele Bosi
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Hospital, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy, and Department of Internal Medicine, IRCCS San Raffaele Hospital, Milan, Italy
| | - Todd M Brusko
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL
| | - Linda A DiMeglio
- Division of Pediatric Endocrinology and Diabetology and Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN
| | - Carmella Evans-Molina
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN
| | - Stephen E Gitelman
- Division of Pediatric Endocrinology and Diabetes, University of California, San Francisco, San Francisco, CA
| | | | - Peter A Gottlieb
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, Aurora, CO
| | - Kevan C Herold
- Department of Immunobiology, Yale University, New Haven, CT
| | - Martin J Hessner
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI
| | - Mikael Knip
- Children's Hospital, University of Helsinki and Helsinki University Hospital, Clinical and Molecular Metabolism Research Program, University of Helsinki, Helsinki, Finland
| | - Laura Jacobsen
- Department of Pediatrics, University of Florida, Gainesville, FL
| | - Jeffrey P Krischer
- Health Informatics Institute, Morsani College of Medicine, University of South Florida, Tampa, FL
| | - S Alice Long
- Diabetes Program, Benaroya Research Institute, Seattle, WA
| | - Markus Lundgren
- Department of Clinical Sciences, Clinical Research Centre, Faculty of Medicine, Lund University, and Skåne University Hospital, Malmö, Sweden
| | - Eoin F McKinney
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's Hospital, Cambridge, U.K
| | - Noel G Morgan
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, U.K.,University of Exeter Medical School and Royal Devon and Exeter Hospital, Exeter, U.K
| | - Richard A Oram
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Royal Devon and Exeter Hospital, Exeter, U.K.,NIHR Exeter Clinical Research Facility, University of Exeter Medical School, Exeter, U.K.,Academic Renal Unit, Royal Devon and Exeter NHS Foundation Trust, Exeter, U.K
| | - Tomi Pastinen
- Center for Pediatric Genomic Medicine, Children's Mercy Kansas City, Kansas City, MO
| | - Michael C Peters
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, and Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA
| | - Alessandra Petrelli
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Hospital, Milan, Italy
| | - Xiaoning Qian
- Department of Electrical and Computer Engineering, TEES-AgriLife Center for Bioinformatics and Genomic Systems Engineering, Texas A&M University, College Station, TX
| | - Maria J Redondo
- Baylor College of Medicine, Texas Children's Hospital, Houston, TX
| | - Bart O Roep
- Department of Diabetes Immunology, Diabetes & Metabolism Research Institute, Beckman Research Institute, National Medical Center, City of Hope, Duarte, CA.,Department of Immunohaematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
| | - Desmond Schatz
- Department of Pediatrics, University of Florida, Gainesville, FL
| | | | - Mark Peakman
- Peter Gorer Department of Immunobiology, Faculty of Life Sciences and Medicine, King's College London, London, U.K. .,King's Health Partners Institute of Diabetes, Obesity and Endocrinology, London, U.K
| |
Collapse
|
34
|
Bonifacio E, Achenbach P. Birth and coming of age of islet autoantibodies. Clin Exp Immunol 2019; 198:294-305. [PMID: 31397889 PMCID: PMC6857083 DOI: 10.1111/cei.13360] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/06/2019] [Indexed: 12/20/2022] Open
Abstract
This review takes the reader through 45 years of islet autoantibody research, from the discovery of islet‐cell antibodies in 1974 to today’s population‐based screening for presymptomatic early‐stage type 1 diabetes. The review emphasizes the current practical value of, and factors to be considered in, the measurement of islet autoantibodies.
Collapse
Affiliation(s)
- E Bonifacio
- Technische Universität Dresden, DFG Center for Regenerative Therapies Dresden, Dresden, Germany.,Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital Carl Gustav Carus and Faculty of Medicine, TU Dresden, Dresden, Germany
| | - P Achenbach
- Institute of Diabetes Research, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany.,Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Forschergruppe Diabetes, Munich, Germany
| |
Collapse
|
35
|
Infante M, Ricordi C, Sanchez J, Clare-Salzler MJ, Padilla N, Fuenmayor V, Chavez C, Alvarez A, Baidal D, Alejandro R, Caprio M, Fabbri A. Influence of Vitamin D on Islet Autoimmunity and Beta-Cell Function in Type 1 Diabetes. Nutrients 2019; 11:E2185. [PMID: 31514368 PMCID: PMC6769474 DOI: 10.3390/nu11092185] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 09/05/2019] [Accepted: 09/06/2019] [Indexed: 02/06/2023] Open
Abstract
Type 1 diabetes (T1D) is a chronic autoimmune disease leading to immune-mediated destruction of pancreatic beta cells, resulting in the need for insulin therapy. The incidence of T1D is increasing worldwide, thus prompting researchers to investigate novel immunomodulatory strategies to halt autoimmunity and modify disease progression. T1D is considered as a multifactorial disease, in which genetic predisposition and environmental factors interact to promote the triggering of autoimmune responses against beta cells. Over the last decades, it has become clear that vitamin D exerts anti-inflammatory and immunomodulatory effects, apart from its well-established role in the regulation of calcium homeostasis and bone metabolism. Importantly, the global incidence of vitamin D deficiency is also dramatically increasing and epidemiologic evidence suggests an involvement of vitamin D deficiency in T1D pathogenesis. Polymorphisms in genes critical for vitamin D metabolism have also been shown to modulate the risk of T1D. Moreover, several studies have investigated the role of vitamin D (in different doses and formulations) as a potential adjuvant immunomodulatory therapy in patients with new-onset and established T1D. This review aims to present the current knowledge on the immunomodulatory effects of vitamin D and summarize the clinical interventional studies investigating its use for prevention or treatment of T1D.
Collapse
Affiliation(s)
- Marco Infante
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
- Department of Systems Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy.
| | - Camillo Ricordi
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - Janine Sanchez
- Pediatric Endocrinology, University of Miami Miller School of Medicine, 1601 NW 12th Avenue, Miami, FL 33136, USA.
| | - Michael J Clare-Salzler
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, 1600 SW Archer Rd, Gainesville, FL 32610, USA.
| | - Nathalia Padilla
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - Virginia Fuenmayor
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - Carmen Chavez
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - Ana Alvarez
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - David Baidal
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - Rodolfo Alejandro
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - Massimiliano Caprio
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, Via di Val Cannuta 247, 00133 Rome, Italy.
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, Via di Val Cannuta 247, 00166 Rome, Italy.
| | - Andrea Fabbri
- Department of Systems Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy.
| |
Collapse
|
36
|
Abstract
PURPOSE OF REVIEW Although checkpoint inhibitor blockade is now widely used clinically for cancer immunotherapy, the reverse process, (i.e. induction of checkpoints to slow autoimmunity) has not been extensively explored. CD8 T-cell exhaustion is a state of immune hyporesponsiveness that may be harnessed to treat autoimmunity. RECENT FINDINGS We focus on the potential role of CD8 T-cell exhaustion as a mechanism of peripheral tolerance in T1D and its therapeutic implications. SUMMARY CD8 T-cell exhaustion is a continuum in which cells change from precursor to terminally exhausted cells. Current thinking based on studies in cancer and chronic viral infection invokes a three-signal model for development of T-cell exhaustion, with persistent antigen, negative costimulatory signals and chronic inflammation comprising signals 1-3, respectively. Transcriptional signatures of CD8 T-cell exhaustion were associated with better prognosis across several autoimmune diseases, most profoundly in systemic diseases. In T1D, CD8 exhaustion was promoted by treatment with anti-CD3 therapy (teplizumab) and was more evident in islet-specific CD8 T cells of slow progressors, suggesting a beneficial role in T1D also. Thus, we apply this three-step process of exhaustion to discuss potential treatments to augment CD8 T-cell exhaustion in T1D.
Collapse
Affiliation(s)
| | - S. Alice Long
- Translational Research Program, Benaroya Research Institute at Virginia Mason, Seattle, Washington, USA
| |
Collapse
|