1
|
Evans-Molina C, Oram RA. Type 1 diabetes presenting in adults: Trends, diagnostic challenges and unique features. Diabetes Obes Metab 2025. [PMID: 40230204 DOI: 10.1111/dom.16402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 03/19/2025] [Accepted: 03/28/2025] [Indexed: 04/16/2025]
Abstract
Type 1 diabetes (T1D) has been historically regarded as a childhood-onset disease; however, recent epidemiological data indicate that adult-onset T1D accounts for a substantial proportion of cases worldwide. There is evidence that adult-onset T1D is associated with the classic T1D triad of elevated genetic risk, the presence of islet-specific autoantibodies and progression to severe insulin deficiency. In this article, we review our understanding of the commonalities and differences between childhood and adult-onset T1D, and we highlight significant knowledge gaps in our understanding of the diagnosis, incidence, trajectory and treatment of adult-onset T1D. Compared to children, adults presenting with T1D exhibit differences in genetic risk, immunologic profiles and metabolic outcomes, including differences in the type and number of autoantibodies present, genetic associations and total genetic burden, rates of C-peptide decline, the persistence of C-peptide in long-duration disease and glycaemic control. In addition, obesity and metabolic syndrome are increasingly common in adults, which not only blurs the clinical distinction of adult-onset T1D from type 2 diabetes (T2D) but also likely contributes to differences in metabolic outcomes and rates of progression. Because T2D is so prevalent in the adult population, adult-onset T1D is misclassified as T2D in at least one in three cases, leading to delays in appropriate treatment. Current diagnostic tools, including autoantibody testing and C-peptide measurement, are underutilised or lack specificity in distinguishing adult-onset T1D from atypical T2D. Additionally, the impact of different responses to disease-modifying therapy between adults and children is unclear. Addressing these knowledge gaps requires expanded epidemiological studies, diverse patient registries and refined classification criteria to improve early detection and treatment strategies. A deeper understanding of adult-onset T1D will be critical to reduce the burden of misdiagnosis, lead to earlier diagnosis and treatment and optimise population-based screening approaches in this under-recognised population. PLAIN LANGUAGE SUMMARY: Type 1 diabetes (T1D) is an autoimmune disease that causes metabolic and nutritional complications due to the destruction of insulin-producing pancreatic β cells. T1D was formerly known as "juvenile diabetes" because it was assumed that most cases occurred in childhood; however, recent epidemiological data show that nearly half of all T1D cases are diagnosed in adulthood. Despite the high prevalence of adult-onset T1D, there are challenges with correctly diagnosing T1D in adulthood, and significant knowledge gaps remain regarding the incidence, trajectory, and treatment of adult-onset T1D. In this article, we summarize the current understanding of commonalities and differences between childhood and adult-onset T1D. Particularly, we highlight age-related differences in genetic risk, immunologic profiles, and metabolic outcomes and complications. Finally, we highlight key gaps in our understanding of adult-onset T1D that need to be addressed to reduce the burden of misdiagnosis and allow for better screening and treatment of T1D in adulthood.
Collapse
Affiliation(s)
- Carmella Evans-Molina
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Roudebush VA Medical Center, Indianapolis, Indiana, USA
| | - Richard A Oram
- Clinical and Biomedical Sciences, University of Exeter Medical School, Exeter, UK
- The Academic Renal Unit, Royal Devon University Hospitals NHS Foundation Trust, Exeter, UK
| |
Collapse
|
2
|
Abdi H, Aganj Z, Hosseinzadeh H, Mosaffa F. Crocin restores the balance of Th1/Th2 immune cell response in ConA-treated human lymphocytes. Pharmacol Rep 2022; 74:513-522. [PMID: 35294736 DOI: 10.1007/s43440-022-00362-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 03/02/2022] [Accepted: 03/03/2022] [Indexed: 01/08/2023]
Abstract
BACKGROUND Following antigen stimulation, naive CD4+ T cells differentiate into different T helper (Th) subsets characterized by lineage-specific transcriptional factors and cytokines. The balance between cytokines from Th1 and Th2 cells is disrupted in autoimmune disorders, asthma, and allergic reactions. Crocin, the major carotenoid of saffron, has anti-inflammatory properties. We investigated crocin modifying effects on the human lymphocytes proliferation and Th1/Th2 balance as a possible mechanism of its anti-inflammatory effects. METHODS The human peripheral blood mononuclear cells were isolated using Ficoll density gradient centrifugation. MTT was used to evaluate the effect of 72-h treatment with different concentrations of crocin with or without ConA on lymphocytes proliferation. INF-γ/IL-4 cytokine secretion and T-bet/GATA-3 transcription factor expression ratios (as indicators of Th1/Th2 response status) were measured in non-stimulated and ConA-stimulated cells in the presence or absence of crocin by ELISA and RT-qPCR methods, respectively. RESULTS The results showed crocin at a concentration of 50 μM and higher was toxic for human lymphocytes, and at a non-toxic concentration of 25 µM, it did not affect cell proliferation. The ratio of T-bet/GATA-3 and INF-γ/IL-4 was higher in the culture supernatant of ConA-stimulated cells compared to non-stimulated ones. Crocin-treated cells showed slightly lower T-bet/GATA-3 and INF-γ/IL-4 ratios compared to untreated cells. Crocin (25 μM) was also able to restore the increased ratio of Th1/Th2 immune response induced by ConA. CONCLUSIONS Crocin can alleviate inflammatory-stimulant effects of ConA on human lymphocytes by decreasing T-bet/GATA-3 and INF-γ/IL-4 ratios, which are indicative of restoring the balance of Th1/Th2 responses.
Collapse
Affiliation(s)
- Hakimeh Abdi
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Aganj
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Hosseinzadeh
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Fatemeh Mosaffa
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
3
|
Clark M, Kroger CJ, Ke Q, Tisch RM. The Role of T Cell Receptor Signaling in the Development of Type 1 Diabetes. Front Immunol 2021; 11:615371. [PMID: 33603744 PMCID: PMC7884625 DOI: 10.3389/fimmu.2020.615371] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 12/15/2020] [Indexed: 12/15/2022] Open
Abstract
T cell receptor (TCR) signaling influences multiple aspects of CD4+ and CD8+ T cell immunobiology including thymic development, peripheral homeostasis, effector subset differentiation/function, and memory formation. Additional T cell signaling cues triggered by co-stimulatory molecules and cytokines also affect TCR signaling duration, as well as accessory pathways that further shape a T cell response. Type 1 diabetes (T1D) is a T cell-driven autoimmune disease targeting the insulin producing β cells in the pancreas. Evidence indicates that dysregulated TCR signaling events in T1D impact the efficacy of central and peripheral tolerance-inducing mechanisms. In this review, we will discuss how the strength and nature of TCR signaling events influence the development of self-reactive T cells and drive the progression of T1D through effects on T cell gene expression, lineage commitment, and maintenance of pathogenic anti-self T cell effector function.
Collapse
Affiliation(s)
- Matthew Clark
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Charles J Kroger
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Qi Ke
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Roland M Tisch
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
4
|
Biassoni R, Di Marco E, Squillario M, Barla A, Piccolo G, Ugolotti E, Gatti C, Minuto N, Patti G, Maghnie M, d'Annunzio G. Gut Microbiota in T1DM-Onset Pediatric Patients: Machine-Learning Algorithms to Classify Microorganisms as Disease Linked. J Clin Endocrinol Metab 2020; 105:5871462. [PMID: 32692360 DOI: 10.1210/clinem/dgaa407] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 07/01/2020] [Indexed: 12/14/2022]
Abstract
AIMS The purpose of this work is to find the gut microbial fingerprinting of pediatric patients with type 1 diabetes. METHODS The microbiome of 31 children with type 1 diabetes at onset and of 25 healthy children was determined using multiple polymorphic regions of the 16S ribosomal RNA. We performed machine-learning analyses and metagenome functional analysis to identify significant taxa and their metabolic pathways content. RESULTS Compared with healthy controls, patients showed a significantly higher relative abundance of the following most important taxa: Bacteroides stercoris, Bacteroides fragilis, Bacteroides intestinalis, Bifidobacterium bifidum, Gammaproteobacteria and its descendants, Holdemania, and Synergistetes and its descendants. On the contrary, the relative abundance of Bacteroides vulgatus, Deltaproteobacteria and its descendants, Parasutterella and the Lactobacillus, Turicibacter genera were significantly lower in patients with respect to healthy controls. The predicted metabolic pathway more associated with type 1 diabetes patients concerns "carbon metabolism," sugar and iron metabolisms in particular. Among the clinical variables considered, standardized body mass index, anti-insulin autoantibodies, glycemia, hemoglobin A1c, Tanner stage, and age at onset emerged as most significant positively or negatively correlated with specific clusters of taxa. CONCLUSIONS The relative abundance and supervised analyses confirmed the importance of B stercoris in type 1 diabetes patients at onset and showed a relevant role of Synergistetes and its descendants in patients with respect to healthy controls. In general the robustness and coherence of the showed results underline the relevance of studying the microbioma using multiple polymorphic regions, different types of analysis, and different approaches within each analysis.
Collapse
Affiliation(s)
- Roberto Biassoni
- Molecular Diagnostics, Analysis Laboratory, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Eddi Di Marco
- Molecular Diagnostics, Analysis Laboratory, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | | | | | - Gianluca Piccolo
- Pediatric Clinic Regional Center for Pediatric Diabetes, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Elisabetta Ugolotti
- Molecular Diagnostics, Analysis Laboratory, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Cinzia Gatti
- Molecular Diagnostics, Analysis Laboratory, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Nicola Minuto
- Pediatric Clinic Regional Center for Pediatric Diabetes, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Giuseppa Patti
- Department of Pediatrics, IRCCS Istituto Giannina Gaslini, University of Genoa, Genoa, Italy
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University Of Genoa, Genoa, Italy
| | - Mohamad Maghnie
- Pediatric Clinic Regional Center for Pediatric Diabetes, IRCCS Istituto Giannina Gaslini, Genoa, Italy
- Department of Pediatrics, IRCCS Istituto Giannina Gaslini, University of Genoa, Genoa, Italy
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University Of Genoa, Genoa, Italy
| | - Giuseppe d'Annunzio
- Pediatric Clinic Regional Center for Pediatric Diabetes, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| |
Collapse
|
5
|
Walter DL, Benner SE, Oaks RJ, Thuma JR, Malgor R, Schwartz FL, Coschigano KT, McCall KD. Coxsackievirus B4 Exposure Results in Variable Pattern Recognition Response in the Kidneys of Female Non-Obese Diabetic Mice Before Establishment of Diabetes. Viral Immunol 2020; 33:494-506. [PMID: 32352894 DOI: 10.1089/vim.2019.0188] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
End-stage renal disease (ESRD) is described by four primary diagnoses, diabetes, hypertension, glomerulonephritis, and cystic kidney disease, all of which have viruses implicated as causative agents. Enteroviruses, such as coxsackievirus (CV), are a common genus of viruses that have been implicated in both diabetes and cystic kidney disease; however, little is known about how CVs cause kidney injury and ESRD or predispose individuals with a genetic susceptibility to type 1 diabetes (T1D) to kidney injury. This study evaluated kidney injury resulting from coxsackievirus B4 (CVB4) inoculation of non-obese diabetic (NOD) mice to glean a better understanding of how viral exposure may predispose individuals with a genetic susceptibility to T1D to kidney injury. The objectives were to assess acute and chronic kidney damage in CVB4-inoculated NOD mice without diabetes. Results indicated the presence of CVB4 RNA in the kidney for at least 14 days post-CVB4 inoculation and a coordinated pattern recognition receptor response, but the absence of an immune response or cytotoxicity. CVB4-inoculated NOD mice also had a higher propensity to develop an increase in mesangial area 17 weeks post-CVB4 inoculation. These studies identified initial gene expression changes in the kidney resulting from CVB4 exposure that may predispose to ESRD. Thus, this study provides an initial characterization of kidney injury resulting from CVB4 inoculation of mice that are genetically susceptible to developing T1D that may one day provide better therapeutic options and predictive measures for patients who are at risk for developing kidney disease from T1D.
Collapse
Affiliation(s)
- Debra L Walter
- Interdisciplinary Program in Molecular and Cellular Biology, Ohio University, Athens, Ohio, USA.,Department of Biological Sciences, College of Arts and Sciences, Ohio University, Athens, Ohio, USA
| | - Sarah E Benner
- Interdisciplinary Program in Molecular and Cellular Biology, Ohio University, Athens, Ohio, USA.,Department of Biological Sciences, College of Arts and Sciences, Ohio University, Athens, Ohio, USA
| | - Rosemary J Oaks
- Program in Biological Sciences, Honors Tutorial College, Ohio University, Athens, Ohio, USA.,Department of Biomedical Sciences and Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, USA
| | - Jean R Thuma
- Department of Specialty Medicine, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, USA.,The Diabetes Institute, Ohio University, Athens, Ohio, USA
| | - Ramiro Malgor
- Interdisciplinary Program in Molecular and Cellular Biology, Ohio University, Athens, Ohio, USA.,Department of Biomedical Sciences and Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, USA.,The Diabetes Institute, Ohio University, Athens, Ohio, USA
| | - Frank L Schwartz
- Department of Specialty Medicine, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, USA.,The Diabetes Institute, Ohio University, Athens, Ohio, USA
| | - Karen T Coschigano
- Interdisciplinary Program in Molecular and Cellular Biology, Ohio University, Athens, Ohio, USA.,Department of Biomedical Sciences and Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, USA.,The Diabetes Institute, Ohio University, Athens, Ohio, USA
| | - Kelly D McCall
- Interdisciplinary Program in Molecular and Cellular Biology, Ohio University, Athens, Ohio, USA.,Department of Biological Sciences, College of Arts and Sciences, Ohio University, Athens, Ohio, USA.,Department of Biomedical Sciences and Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, USA.,Department of Specialty Medicine, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, USA.,The Diabetes Institute, Ohio University, Athens, Ohio, USA
| |
Collapse
|
6
|
Abstract
The novel understanding that the presence of multiple islet autoantibodies, indicating islet autoimmunity, inevitably leads to type 1 diabetes mellitus (T1DM) has necessitated the development of a new staging classification system for the condition. Coupled with an improved understanding of the disease course, the realization that T1DM appears to be more heterogeneous than previously thought has led to unique opportunities to develop more targeted therapies that may be applied even before the onset of dysglycemia or symptoms. To date, several therapies have been trialed to delay or halt disease progression in both presymptomatic and clinical T1DM, each demonstrating varying degrees of effectiveness, toxicity, and utility. Key research supports the eventual implementation of immunotherapy in autoimmune diabetes, potentially calling for a paradigm shift among care providers. It will likely be necessary to develop new approaches to trial design and to address potential barriers to progress before an effective treatment for the disease may be achieved.
Collapse
|
7
|
Salami F, Lee HS, Freyhult E, Elding Larsson H, Lernmark Å, Törn C. Reduction in White Blood Cell, Neutrophil, and Red Blood Cell Counts Related to Sex, HLA, and Islet Autoantibodies in Swedish TEDDY Children at Increased Risk for Type 1 Diabetes. Diabetes 2018; 67:2329-2336. [PMID: 30104249 PMCID: PMC6198343 DOI: 10.2337/db18-0355] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 07/29/2018] [Indexed: 12/19/2022]
Abstract
Islet autoantibodies (IAs) precede the clinical onset of type 1 diabetes (T1D); however, the knowledge is limited about whether the prodrome affects complete blood counts (CBCs) in 4- to 12-year-old children with increased genetic risk for T1D. This study tested whether CBCs were altered in 4- to 12-year-old children without (n = 376) or with one or several IAs against insulin, GAD65, or IA-2 (n = 72). CBC was analyzed during longitudinal follow-up in 448 Swedish children enrolled in The Environmental Determinants of Diabetes in the Young (TEDDY) study. A linear mixed-effects model was used to assess potential association between IA and CBC measurements over time. The white blood cell and neutrophil counts were reduced in children with IAs, primarily in boys. In contrast, girls had lower levels of hemoglobin and hematocrit. Positivity for multiple IAs showed the lowest counts in white blood cells and neutrophils in boys and red blood cells, hemoglobin, and hematocrit in girls. These associations were primarily observed in children with the HLA-DR3-DQ2/DR4-DQ8 genotype. We conclude that the reduction in neutrophils and red blood cells in children with multiple IAs and HLA-DR3-DQ2/DR4-DQ8 genotype may signal a sex-dependent islet autoimmunity detected in longitudinal CBCs.
Collapse
Affiliation(s)
- Falastin Salami
- Department of Clinical Sciences, Lund University/Clinical Research Centre, Skåne University Hospital, Malmö, Sweden
| | - Hye-Seung Lee
- Health Informatics Institute, Department of Pediatrics, University of South Florida, Tampa, FL
| | - Eva Freyhult
- Department of Medical Sciences, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Helena Elding Larsson
- Department of Clinical Sciences, Lund University/Clinical Research Centre, Skåne University Hospital, Malmö, Sweden
| | - Åke Lernmark
- Department of Clinical Sciences, Lund University/Clinical Research Centre, Skåne University Hospital, Malmö, Sweden
| | - Carina Törn
- Department of Clinical Sciences, Lund University/Clinical Research Centre, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
8
|
Kroger CJ, Clark M, Ke Q, Tisch RM. Therapies to Suppress β Cell Autoimmunity in Type 1 Diabetes. Front Immunol 2018; 9:1891. [PMID: 30166987 PMCID: PMC6105696 DOI: 10.3389/fimmu.2018.01891] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 07/31/2018] [Indexed: 12/12/2022] Open
Abstract
Type 1 diabetes (T1D) is an autoimmune disease that is generally considered to be T cell-driven. Accordingly, most strategies of immunotherapy for T1D prevention and treatment in the clinic have targeted the T cell compartment. To date, however, immunotherapy has had only limited clinical success. Although certain immunotherapies have promoted a protective effect, efficacy is often short-term and acquired immunity may be impacted. This has led to the consideration of combining different approaches with the goal of achieving a synergistic therapeutic response. In this review, we will discuss the status of various T1D therapeutic strategies tested in the clinic, as well as possible combinatorial approaches to restore β cell tolerance.
Collapse
Affiliation(s)
- Charles J Kroger
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Matthew Clark
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Qi Ke
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Roland M Tisch
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
9
|
Yeo L, Woodwyk A, Sood S, Lorenc A, Eichmann M, Pujol-Autonell I, Melchiotti R, Skowera A, Fidanis E, Dolton GM, Tungatt K, Sewell AK, Heck S, Saxena A, Beam CA, Peakman M. Autoreactive T effector memory differentiation mirrors β cell function in type 1 diabetes. J Clin Invest 2018; 128:3460-3474. [PMID: 29851415 DOI: 10.1172/jci120555] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 05/23/2018] [Indexed: 12/26/2022] Open
Abstract
In type 1 diabetes, cytotoxic CD8+ T cells with specificity for β cell autoantigens are found in the pancreatic islets, where they are implicated in the destruction of insulin-secreting β cells. In contrast, the disease relevance of β cell-reactive CD8+ T cells that are detectable in the circulation, and their relationship to β cell function, are not known. Here, we tracked multiple, circulating β cell-reactive CD8+ T cell subsets and measured β cell function longitudinally for 2 years, starting immediately after diagnosis of type 1 diabetes. We found that change in β cell-specific effector memory CD8+ T cells expressing CD57 was positively correlated with C-peptide change in subjects below 12 years of age. Autoreactive CD57+ effector memory CD8+ T cells bore the signature of enhanced effector function (higher expression of granzyme B, killer-specific protein of 37 kDa, and CD16, and reduced expression of CD28) compared with their CD57- counterparts, and network association modeling indicated that the dynamics of β cell-reactive CD57+ effector memory CD8+ T cell subsets were strongly linked. Thus, coordinated changes in circulating β cell-specific CD8+ T cells within the CD57+ effector memory subset calibrate to functional insulin reserve in type 1 diabetes, providing a tool for immune monitoring and a mechanism-based target for immunotherapy.
Collapse
Affiliation(s)
- Lorraine Yeo
- Department of Immunobiology, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom.,National Institute of Health Research Biomedical Research Centre at Guy's and St Thomas' Hospital and King's College London, London, United Kingdom
| | - Alyssa Woodwyk
- Division of Epidemiology and Biostatistics, Department of Biomedical Sciences, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, Michigan, USA
| | - Sanjana Sood
- National Institute of Health Research Biomedical Research Centre at Guy's and St Thomas' Hospital and King's College London, London, United Kingdom
| | - Anna Lorenc
- Department of Immunobiology, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Martin Eichmann
- Department of Immunobiology, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Irma Pujol-Autonell
- Department of Immunobiology, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Rosella Melchiotti
- National Institute of Health Research Biomedical Research Centre at Guy's and St Thomas' Hospital and King's College London, London, United Kingdom
| | - Ania Skowera
- Department of Immunobiology, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Efthymios Fidanis
- National Institute of Health Research Biomedical Research Centre at Guy's and St Thomas' Hospital and King's College London, London, United Kingdom
| | - Garry M Dolton
- Division of Infection and Immunity and Systems Immunity Research Institute, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Katie Tungatt
- Division of Infection and Immunity and Systems Immunity Research Institute, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Andrew K Sewell
- Division of Infection and Immunity and Systems Immunity Research Institute, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Susanne Heck
- National Institute of Health Research Biomedical Research Centre at Guy's and St Thomas' Hospital and King's College London, London, United Kingdom
| | - Alka Saxena
- National Institute of Health Research Biomedical Research Centre at Guy's and St Thomas' Hospital and King's College London, London, United Kingdom
| | - Craig A Beam
- Division of Epidemiology and Biostatistics, Department of Biomedical Sciences, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, Michigan, USA
| | - Mark Peakman
- Department of Immunobiology, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom.,National Institute of Health Research Biomedical Research Centre at Guy's and St Thomas' Hospital and King's College London, London, United Kingdom.,King's Health Partners Institute of Diabetes, Endocrinology and Obesity, London, United Kingdom
| |
Collapse
|
10
|
Nagy N, Kuipers HF, Marshall PL, Wang E, Kaber G, Bollyky PL. Hyaluronan in immune dysregulation and autoimmune diseases. Matrix Biol 2018; 78-79:292-313. [PMID: 29625181 DOI: 10.1016/j.matbio.2018.03.022] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 03/10/2018] [Accepted: 03/30/2018] [Indexed: 02/06/2023]
Abstract
The tissue microenvironment contributes to local immunity and to the pathogenesis of autoimmune diseases - a diverse set of conditions characterized by sterile inflammation, immunity against self-antigens, and destruction of tissues. However, the specific factors within the tissue microenvironment that contribute to local immune dysregulation in autoimmunity are poorly understood. One particular tissue component implicated in multiple autoimmune diseases is hyaluronan (HA), an extracellular matrix (ECM) polymer. HA is abundant in settings of chronic inflammation and contributes to lymphocyte activation, polarization, and migration. Here, we first describe what is known about the size, amount, and distribution of HA at sites of autoimmunity and in associated lymphoid structures in type 1 diabetes, multiple sclerosis, and rheumatoid arthritis. Next, we examine the recent literature on HA and its impact on adaptive immunity, particularly in regards to the biology of lymphocytes and Foxp3+ regulatory T-cells (Treg), a T-cell subset that maintains immune tolerance in healthy individuals. We propose that HA accumulation at sites of chronic inflammation creates a permissive environment for autoimmunity, characterized by CD44-mediated inhibition of Treg expansion. Finally, we address potential tools and strategies for targeting HA and its receptor CD44 in chronic inflammation and autoimmunity.
Collapse
Affiliation(s)
- Nadine Nagy
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.
| | - Hedwich F Kuipers
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Payton L Marshall
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Esther Wang
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Gernot Kaber
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Paul L Bollyky
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
11
|
Clark M, Kroger CJ, Tisch RM. Type 1 Diabetes: A Chronic Anti-Self-Inflammatory Response. Front Immunol 2017; 8:1898. [PMID: 29312356 PMCID: PMC5743904 DOI: 10.3389/fimmu.2017.01898] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 12/12/2017] [Indexed: 12/16/2022] Open
Abstract
Inflammation is typically induced in response to a microbial infection. The release of proinflammatory cytokines enhances the stimulatory capacity of antigen-presenting cells, as well as recruits adaptive and innate immune effectors to the site of infection. Once the microbe is cleared, inflammation is resolved by various mechanisms to avoid unnecessary tissue damage. Autoimmunity arises when aberrant immune responses target self-tissues causing inflammation. In type 1 diabetes (T1D), T cells attack the insulin producing β cells in the pancreatic islets. Genetic and environmental factors increase T1D risk by in part altering central and peripheral tolerance inducing events. This results in the development and expansion of β cell-specific effector T cells (Teff) which mediate islet inflammation. Unlike protective immunity where inflammation is terminated, autoimmunity is sustained by chronic inflammation. In this review, we will highlight the key events which initiate and sustain T cell-driven pancreatic islet inflammation in nonobese diabetic mice and in human T1D. Specifically, we will discuss: (i) dysregulation of thymic selection events, (ii) the role of intrinsic and extrinsic factors that enhance the expansion and pathogenicity of Teff, (iii) defects which impair homeostasis and suppressor activity of FoxP3-expressing regulatory T cells, and (iv) properties of β cells which contribute to islet inflammation.
Collapse
Affiliation(s)
- Matthew Clark
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Charles J Kroger
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Roland M Tisch
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
12
|
Regnell SE, Hessner MJ, Jia S, Åkesson L, Stenlund H, Moritz T, La Torre D, Lernmark Å. Longitudinal analysis of hepatic transcriptome and serum metabolome demonstrates altered lipid metabolism following the onset of hyperglycemia in spontaneously diabetic biobreeding rats. PLoS One 2017; 12:e0171372. [PMID: 28192442 PMCID: PMC5305198 DOI: 10.1371/journal.pone.0171372] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2016] [Accepted: 01/18/2017] [Indexed: 12/30/2022] Open
Abstract
Type 1 diabetes is associated with abberations of fat metabolism before and after the clinical onset of disease. It has been hypothesized that the absence of the effect of insulin in the liver contributes to reduced hepatic fat synthesis. We measured hepatic gene expression and serum metabolites before and after the onset of hyperglycemia in a BioBreeding rat model of type 1 diabetes. Functional pathway annotation identified that lipid metabolism was differentially expressed in hyperglycemic rats and that these pathways significantly overlapped with genes regulated by insulin. 17 serum metabolites significantly changed in concentration. All but 2 of the identified metabolites had previously been reported in type 1 diabetes, and carbohydrates were overall the most upregulated class of metabolites. We conclude that lack of insulin in the liver contributes to the changes in fat metabolism observed in type 1 diabetes. Further studies are needed to understand the clinical consequences of a lack of insulin in the liver in patients with type 1 diabetes.
Collapse
Affiliation(s)
- Simon E. Regnell
- Diabetes and Celiac Disease Unit, Department of Clinical Sciences, Lund University/Clinical Research Centre and Skåne University Hospital, Malmö, Sweden
- * E-mail:
| | - Martin J. Hessner
- Max McGee National Research Center for Juvenile Diabetes, Children's Research Institute of Children's Hospital of Wisconsin, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Shuang Jia
- Max McGee National Research Center for Juvenile Diabetes, Children's Research Institute of Children's Hospital of Wisconsin, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Lina Åkesson
- Diabetes and Celiac Disease Unit, Department of Clinical Sciences, Lund University/Clinical Research Centre and Skåne University Hospital, Malmö, Sweden
| | - Hans Stenlund
- Swedish Metabolomics Centre, Swedish University of Agricultural Sciences, Umeå, Sweden
| | - Thomas Moritz
- Swedish Metabolomics Centre, Swedish University of Agricultural Sciences, Umeå, Sweden
| | - Daria La Torre
- Diabetes and Celiac Disease Unit, Department of Clinical Sciences, Lund University/Clinical Research Centre and Skåne University Hospital, Malmö, Sweden
| | - Åke Lernmark
- Diabetes and Celiac Disease Unit, Department of Clinical Sciences, Lund University/Clinical Research Centre and Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
13
|
Hull RL, Bogdani M, Nagy N, Johnson PY, Wight TN. Hyaluronan: A Mediator of Islet Dysfunction and Destruction in Diabetes? J Histochem Cytochem 2015. [PMID: 26216136 DOI: 10.1369/0022155415576542] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Hyaluronan (HA) is an extracellular matrix (ECM) component that is present in mouse and human islet ECM. HA is localized in peri-islet and intra-islet regions adjacent to microvessels. HA normally exists in a high molecular weight form, which is anti-inflammatory. However, under inflammatory conditions, HA is degraded into fragments that are proinflammatory. HA accumulates in islets of human subjects with recent onset type 1 diabetes (T1D), and is associated with myeloid and lymphocytic islet infiltration, suggesting a possible role for HA in insulitis. A similar accumulation of HA, in amount and location, occurs in non-obese diabetic (NOD) and DORmO mouse models of T1D. Furthermore, HA accumulates in follicular germinal centers and in T-cell areas in lymph nodes and spleen in both human and mouse models of T1D, as compared with control tissues. Whether HA accumulates in islets in type 2 diabetes (T2D) or models thereof has not been previously described. Here we show evidence that HA accumulates in a mouse model of islet amyloid deposition, a well-known component of islet pathology in T2D. In summary, islet HA accumulation is a feature of both T1D and a model of T2D, and may represent a novel inflammatory mediator of islet pathology.
Collapse
Affiliation(s)
- Rebecca L Hull
- Division of Metabolism, Endocrinology and Nutrition, VA Puget Sound Health Care System and University of Washington (RLH)
| | - Marika Bogdani
- Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, Washington (MB, NN, PYJ, TNW)
| | - Nadine Nagy
- Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, Washington (MB, NN, PYJ, TNW)
| | - Pamela Y Johnson
- Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, Washington (MB, NN, PYJ, TNW)
| | - Thomas N Wight
- Department of Pathology, University of Washington, Seattle, Washington (TNW)
| |
Collapse
|
14
|
Lord S, Greenbaum CJ. Disease modifying therapies in type 1 diabetes: Where have we been, and where are we going? Pharmacol Res 2015; 98:3-8. [PMID: 25771310 PMCID: PMC4469522 DOI: 10.1016/j.phrs.2015.02.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 02/08/2015] [Indexed: 01/10/2023]
Abstract
With more than four decades of clinical research and 25 years of clinical trials, much is known about the natural history of T1D before and after clinical diagnosis. We know that autoimmunity occurs early in life, that islet autoimmunity inevitably leads to clinically overt disease, and that some immune therapies can alter the disease course. In the future, we will likely conduct trials to more deeply explore mechanisms of disease and response to therapy, employ combinations of agents including those aimed at supporting beta cells, consider the use of chronic, intermittent therapy, focus studies on preventing progression from islet autoimmunity, and consider the potential benefits of studying children independently from adults. Much of this work will depend upon clinical trial networks such as Diabetes TrialNet. Such networks not only have the expertise to conduct studies but their sharing of data and samples also allows for discovery work by multiple investigators, laying the groundwork for the future. Working with patients, families, funders and industry, such collaborative networks can accelerate the translation of science to clinical practice to improve the lives of those living with T1D.
Collapse
Affiliation(s)
- Sandra Lord
- Diabetes Clinical Research Program, Benaroya Research Institute, Seattle, WA, USA.
| | - Carla J Greenbaum
- Diabetes Clinical Research Program, Benaroya Research Institute, Seattle, WA, USA
| |
Collapse
|
15
|
Rekers NV, von Herrath MG, Wesley JD. Immunotherapies and immune biomarkers in Type 1 diabetes: A partnership for success. Clin Immunol 2015; 161:37-43. [PMID: 26122172 DOI: 10.1016/j.clim.2015.05.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 05/13/2015] [Accepted: 05/17/2015] [Indexed: 12/16/2022]
Abstract
The standard of care (SoC) for Type 1 diabetes (T1D) today is much the same as it was in the early 1920s, simply with more insulin options-fast-acting, slow-acting, injectable, and inhalable insulins. However, these well-tolerated treatments only manage the symptoms and complications, but do nothing to halt the underlying immune response. There is an unmet need for better treatment options for T1D that address all aspects of the disease. For decades, we have successfully treated T1D in preclinical animal models with immune-modifying therapies that have not demonstrated comparable efficacy in humans. The path to bringing such options to the clinic will depend on the implementation and standard inclusion of biomarkers of immune and therapeutic efficacy in T1D clinical trials, and dictate if we can create a new SoC that treats the underlying autoimmunity as well as the symptoms it causes.
Collapse
Affiliation(s)
- Niels V Rekers
- Type 1 Diabetes R&D Center, Novo Nordisk Inc., Seattle, WA, USA; Pacific Northwest Diabetes Research Institute, Seattle, WA, USA
| | | | - Johnna D Wesley
- Type 1 Diabetes R&D Center, Novo Nordisk Inc., Seattle, WA, USA.
| |
Collapse
|
16
|
Extreme Beta-Cell Deficiency in Pancreata of Dogs with Canine Diabetes. PLoS One 2015; 10:e0129809. [PMID: 26057531 PMCID: PMC4461304 DOI: 10.1371/journal.pone.0129809] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 05/13/2015] [Indexed: 01/25/2023] Open
Abstract
The pathophysiology of canine diabetes remains poorly understood, in part due to enigmatic clinical features and the lack of detailed histopathology studies. Canine diabetes, similar to human type 1 diabetes, is frequently associated with diabetic ketoacidosis at onset or after insulin omission. However, notable differences exist. Whereas human type 1 diabetes often occurs in children, canine diabetes is typically described in middle age to elderly dogs. Many competing theories have been proposed regarding the underlying cause of canine diabetes, from pancreatic atrophy to chronic pancreatitis to autoimmune mediated β-cell destruction. It remains unclear to what extent β-cell loss contributes to canine diabetes, as precise quantifications of islet morphometry have not been performed. We used high-throughput microscopy and automated image processing to characterize islet histology in a large collection of pancreata of diabetic dogs. Diabetic pancreata displayed a profound reduction in β-cells and islet endocrine cells. Unlike humans, canine non-diabetic islets are largely comprised of β-cells. Very few β-cells remained in islets of diabetic dogs, even in pancreata from new onset cases. Similarly, total islet endocrine cell number was sharply reduced in diabetic dogs. No compensatory proliferation or lymphocyte infiltration was detected. The majority of pancreata had no evidence of pancreatitis. Thus, canine diabetes is associated with extreme β-cell deficiency in both new and longstanding disease. The β-cell predominant composition of canine islets and the near-total absence of β-cells in new onset elderly diabetic dogs strongly implies that similar to human type 1 diabetes, β-cell loss underlies the pathophysiology of canine diabetes.
Collapse
|
17
|
Reddy S, Zeng N, Al-Diery H, Jung D, Yeu C, Joret MO, Merrilees MJ, Wu F. Analysis of peri-islet CD45-positive leucocytic infiltrates in long-standing type 1 diabetic patients. Diabetologia 2015; 58:1024-35. [PMID: 25687234 DOI: 10.1007/s00125-015-3519-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 01/14/2015] [Indexed: 01/08/2023]
Abstract
AIMS/HYPOTHESIS The role of peri-islet CD45-positive leucocytes, as one component of insulitis, in beta cell death during human type 1 diabetes remains unclear. We undertook a case study, comparing and quantifying leucocytes in the peri- and intra-islet areas in insulin-positive and -negative islets, to assess whether peri-islet leucocytes are pathogenic to beta cells during type 1 diabetes. METHODS Pancreatic sections from 12 diabetic patients (0.25-12 years of disease) and 13 non-diabetic individuals with and without autoantibodies were triple-immunostained for islet leucocytes, insulin and glucagon cells. Islets were graded for insulitis, enumerated and mapped for the spatial distribution of leucocytes in peri- and intra-islet areas in relation to insulin- and glucagon-immunopositive cells. RESULTS In the non-diabetic autoantibody-negative group, the percentage of islets with insulitis was either absent or <1% in five out of eight cases and ranged from 1.3% to 19.4% in three cases. In the five non-diabetic autoantibody-positive cases, it varied from 1.5% to 16.9%. In the diabetic group, it was <1% in one case and 1.1-26.9% in 11 cases, with insulitis being absent in 68% of insulin-positive islets. Peri-islet leucocytes were more numerous than intra-islet leucocytes in islets with insulin positivity. Increasing numbers of exocrine leucocytes in non-diabetic autoantibody-positive and diabetic donors were also present. CONCLUSIONS/INTERPRETATION The prominence of peri-islet leucocytes in insulin-positive islets in most long-standing diabetic individuals suggests that they may be pathogenic to residual beta cells. Increasing numbers of leucocytes in the exocrine region may also participate in the pathogenesis of type 1 diabetes.
Collapse
Affiliation(s)
- Shiva Reddy
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland, New Zealand,
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Abstract
Gi-GPCRs, G protein-coupled receptors that signal via Gα proteins of the i/o class (Gαi/o), acutely regulate cellular behaviors widely in mammalian tissues, but their impact on the development and growth of these tissues is less clear. For example, Gi-GPCRs acutely regulate insulin release from pancreatic β cells, and variants in genes encoding several Gi-GPCRs--including the α-2a adrenergic receptor, ADRA2A--increase the risk of type 2 diabetes mellitus. However, type 2 diabetes also is associated with reduced total β-cell mass, and the role of Gi-GPCRs in establishing β-cell mass is unknown. Therefore, we asked whether Gi-GPCR signaling regulates β-cell mass. Here we show that Gi-GPCRs limit the proliferation of the insulin-producing pancreatic β cells and especially their expansion during the critical perinatal period. Increased Gi-GPCR activity in perinatal β cells decreased β-cell proliferation, reduced adult β-cell mass, and impaired glucose homeostasis. In contrast, Gi-GPCR inhibition enhanced perinatal β-cell proliferation, increased adult β-cell mass, and improved glucose homeostasis. Transcriptome analysis detected the expression of multiple Gi-GPCRs in developing and adult β cells, and gene-deletion experiments identified ADRA2A as a key Gi-GPCR regulator of β-cell replication. These studies link Gi-GPCR signaling to β-cell mass and diabetes risk and identify it as a potential target for therapies to protect and increase β-cell mass in patients with diabetes.
Collapse
|
19
|
Bogdani M, Korpos E, Simeonovic CJ, Parish CR, Sorokin L, Wight TN. Extracellular matrix components in the pathogenesis of type 1 diabetes. Curr Diab Rep 2014; 14:552. [PMID: 25344787 PMCID: PMC4238291 DOI: 10.1007/s11892-014-0552-7] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Type 1 diabetes (T1D) results from progressive immune cell-mediated destruction of pancreatic β cells. As immune cells migrate into the islets, they pass through the extracellular matrix (ECM). This ECM is composed of different macromolecules localized to different compartments within and surrounding islets; however, the involvement of this ECM in the development of human T1D is not well understood. Here, we summarize our recent findings from human and mouse studies illustrating how specific components of the islet ECM that constitute basement membranes and interstitial matrix of the islets, and surprisingly, the intracellular composition of islet β cells themselves, are significantly altered during the pathogenesis of T1D. Our focus is on the ECM molecules laminins, collagens, heparan sulfate/heparan sulfate proteoglycans, and hyaluronan, as well as on the enzymes that degrade these ECM components. We propose that islet and lymphoid tissue ECM composition and organization are critical to promoting immune cell activation, islet invasion, and destruction of islet β cells in T1D.
Collapse
Affiliation(s)
- Marika Bogdani
- Matrix Biology Program, Benaroya Research Institute, 1201 Ninth Avenue, Seattle, WA 98101 Ph: 206-287-5666, Fax: 206-342-6567
| | - Eva Korpos
- Institute of Physiological Chemistry and Pathobiochemistry,Cells-in-Motion Cluster of Excellence (EXC 1003 – CiM), University of Münster, Münster, Germany
| | - Charmaine J. Simeonovic
- Diabetes/Transplantation Immunobiology Laboratory, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, 2601 Australia
| | - Christopher R. Parish
- Cancer and Vascular Biology Group, Department of Immunology, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Lydia Sorokin
- Institute of Physiological Chemistry and Pathobiochemistry,Cells-in-Motion Cluster of Excellence (EXC 1003 – CiM), University of Münster, Münster, Germany
| | - Thomas N. Wight
- Matrix Biology Program, Benaroya Research Institute, 1201 Ninth Avenue, Seattle, WA 98101 Ph: 206-287-5666, Fax: 206-342-6567
- Corresponding Author: Thomas N. Wight, PhD
| |
Collapse
|
20
|
Atkinson MA. Losing a grip on the notion of β-cell specificity for immune responses in type 1 diabetes: can we handle the truth? Diabetes 2014; 63:3572-4. [PMID: 25342726 PMCID: PMC4207390 DOI: 10.2337/db14-1069] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Mark A Atkinson
- Departments of Pathology and Pediatrics, University of Florida, Gainesville, FL
| |
Collapse
|
21
|
Bogdani M, Johnson PY, Potter-Perigo S, Nagy N, Day AJ, Bollyky PL, Wight TN. Hyaluronan and hyaluronan-binding proteins accumulate in both human type 1 diabetic islets and lymphoid tissues and associate with inflammatory cells in insulitis. Diabetes 2014; 63:2727-43. [PMID: 24677718 PMCID: PMC4113060 DOI: 10.2337/db13-1658] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Hyaluronan (HA) is an extracellular matrix glycosaminoglycan that is present in pancreatic islets, but little is known about its involvement in the development of human type 1 diabetes (T1D). We have evaluated whether pancreatic islets and lymphoid tissues of T1D and nondiabetic organ donors differ in the amount and distribution of HA and HA-binding proteins (hyaladherins), such as inter-α-inhibitor (IαI), versican, and tumor necrosis factor-stimulated gene-6 (TSG-6). HA was dramatically increased both within the islet and outside the islet endocrine cells, juxtaposed to islet microvessels in T1D. In addition, HA was prominent surrounding immune cells in areas of insulitis. IαI and versican were present in HA-rich areas of islets, and both molecules accumulated in diabetic islets and regions exhibiting insulitis. TSG-6 was observed within the islet endocrine cells and in inflammatory infiltrates. These patterns were only observed in tissues from younger donors with disease duration of <10 years. Furthermore, HA and IαI amassed in follicular germinal centers and in T-cell areas in lymph nodes and spleens in T1D patients compared with control subjects. Our observations highlight potential roles for HA and hyaladherins in the pathogenesis of diabetes.
Collapse
Affiliation(s)
- Marika Bogdani
- Matrix Biology Program, Benaroya Research Institute, Seattle, WA
| | - Pamela Y Johnson
- Matrix Biology Program, Benaroya Research Institute, Seattle, WA
| | | | - Nadine Nagy
- Matrix Biology Program, Benaroya Research Institute, Seattle, WA
| | - Anthony J Day
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, Manchester, U.K
| | - Paul L Bollyky
- Division of Infectious Diseases, Stanford University Medical Center, Stanford, CA
| | - Thomas N Wight
- Matrix Biology Program, Benaroya Research Institute, Seattle, WA
| |
Collapse
|
22
|
Xia CQ, Peng R, Chernatynskaya AV, Yuan L, Carter C, Valentine J, Sobel E, Atkinson MA, Clare-Salzler MJ. Increased IFN-α-producing plasmacytoid dendritic cells (pDCs) in human Th1-mediated type 1 diabetes: pDCs augment Th1 responses through IFN-α production. THE JOURNAL OF IMMUNOLOGY 2014; 193:1024-34. [PMID: 24973447 DOI: 10.4049/jimmunol.1303230] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Increasing evidence suggests that type 1 IFN (IFN-αβ) is associated with pathogenesis of Th1-mediated type 1 diabetes (T1D). A major source of IFN-αβ is plasmacytoid dendritic cells (pDCs). In this study, we analyzed peripheral blood pDC numbers and functions in at-risk, new-onset, and established T1D patients and controls. We found that subjects at risk for T1D and new-onset and established T1D subjects possessed significantly increased pDCs but similar number of myeloid DCs when compared with controls. pDC numbers were not affected by age in T1D subjects but declined with increasing age in control subjects. It was demonstrated that IFN-α production by PBMCs stimulated with influenza viruses was significantly higher in T1D subjects than in controls, and IFN-α production was correlated with pDC numbers in PBMCs. Of interest, only T1D-associated Coxsackievirus serotype B4 but not B3 induced majority of T1D PBMCs to produce IFN-α, which was confirmed to be secreted by pDCs. Finally, in vitro studies demonstrated IFN-α produced by pDCs augmented Th1 responses, with significantly greater IFN-γ-producing CD4(+) T cells from T1D subjects. These findings indicate that increased pDCs and their IFN-αβ production may be associated with this Th1-mediated autoimmune disease, especially under certain viral infections linked to T1D pathogenesis.
Collapse
Affiliation(s)
- Chang-Qing Xia
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610
| | - Ruihua Peng
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610
| | - Anna V Chernatynskaya
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610
| | - Lihui Yuan
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610
| | - Carolyn Carter
- Department of Pediatrics, University of Florida, Gainesville, FL 32610; and
| | - John Valentine
- Department of Medicine, University of Florida, Gainesville, FL 32610
| | - Eric Sobel
- Department of Medicine, University of Florida, Gainesville, FL 32610
| | - Mark A Atkinson
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610
| | - Michael J Clare-Salzler
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610;
| |
Collapse
|
23
|
Pancreas volume measurement in patients with Type 2 diabetes using magnetic resonance imaging-based planimetry. Pancreatology 2014; 14:268-74. [PMID: 25062875 DOI: 10.1016/j.pan.2014.04.031] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 04/23/2014] [Accepted: 04/26/2014] [Indexed: 12/11/2022]
Abstract
BACKGROUND/OBJECTIVES To compare pancreas volume (PV) measurement using MRI-based planimetry in patients with Type 2 diabetes mellitus (DM) to PV in normoglycemic individuals. METHODS Our institutional review board granted approval of this retrospective study with waiver of informed consent. We searched 2296 consecutive abdominal MRI studies performed at our hospital on patients with no pancreas pathology between September 1, 2010 and February 28, 2013, for those who also had a fasting plasma glucose and/or hemoglobin A1C within six months of the MRI examination. For those patients who met biochemical criteria for DM, we used medication and clinical records to confirm that 32 of these patients had Type 2 DM. The pancreas contours of 32 Type 2 diabetics and 50 normoglycemic individuals were then traced on non-gadolinium T1-weighted 3D fat suppressed gradient echo images by a radiologist trained in abdominal MRI to calculate PV. PV index (PVI) was calculated as PV/weight to adjust PV for each patient's weight. PVs and PVIs in both cohorts were compared using t-tests and regression models correcting for weight, age and gender. RESULTS Patients with Type 2 DM had significantly lower PVs than normoglycemic individuals (72.7 ± 20.7 cm(3) versus 89.6 ± 22.7 cm(3), p < 0.001), and significantly lower PVIs (1.0 ± 0.3 cm(3)/kg versus 1.3 ± 0.3 cm(3)/kg, p < 0.001). Using regression models, we found that given the same age, weight and gender, the PV in a patient with Type 2 DM was 17.9 mL (20%) lower compared to a normoglycemic individual (p < 0.001). CONCLUSION PV is reduced in Type 2 DM compared to normoglycemic individuals and can be measured using MRI without contrast injection.
Collapse
|
24
|
Atkinson MA. Pancreatic biopsies in type 1 diabetes: revisiting the myth of Pandora's box. Diabetologia 2014; 57:656-9. [PMID: 24442510 PMCID: PMC7322791 DOI: 10.1007/s00125-013-3159-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Accepted: 12/16/2013] [Indexed: 01/23/2023]
Abstract
Over a century ago, inquisitive physicians made remarkable discoveries regarding pancreatic pathology in individuals with diabetes, including those who were likely afflicted with the type 1 (autoimmune) form of the disease. Those studies of post-mortem tissues noted unique anatomical changes in islet architecture as well as the presence of unusual cellular infiltrates. In the time since, investigations of pancreatic pathology have, with near uniformity, been restricted to analysis of organs obtained post-mortem. While clearly beneficial for addressing questions of the disorder's pathogenesis, concern exists regarding potential artefacts that might occur through analysis of tissues that have been recovered hours, often many hours, following death. Beyond this, studies of tissues obtained long after the diagnosis of type 1 diabetes may not disclose important physiological events occurring at onset or even earlier in the natural history of disease, before symptomatic hyperglycaemia. To this end, Krogvold and colleagues (in this issue of Diabetologia, doi: 10.1007/s00125-013-3155-y) undertook a potentially high-reward strategy involving pancreatic biopsy in living adults with recent-onset type 1 diabetes. Procedures were performed under informed consent, undertaken based on recent improvements in laparoscopic techniques, and carried out by individuals with considerable surgical experience. These efforts were terminated for ethical reasons following the occurrence of serious complications (including post-operative bleeding and pancreatic leakage). The experience lends itself to analogy with the Greek myth of Pandora's box where curiosity, in terms of a desire to see what resided inside a closed container, unleashed a series of ills on humans once the container was opened. In considering the moral of that myth, one must question whether the secrets of the pancreas in those living with type 1 diabetes should, for now, remain a mystery as the process of manipulating that organ for the purpose of curiosity does not occur without harm.
Collapse
Affiliation(s)
- Mark A Atkinson
- Department of Pathology, College of Medicine, University of Florida, 1600 SW Archer Road, Gainesville, FL, 32610-0275, USA,
| |
Collapse
|
25
|
Ludvigsson J. Is it time to challenge the established theories surrounding type 1 diabetes? Acta Paediatr 2014; 103:120-3. [PMID: 24344989 DOI: 10.1111/apa.12452] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Revised: 08/08/2013] [Accepted: 10/02/2013] [Indexed: 01/12/2023]
Abstract
UNLABELLED Type one diabetes (T1D) seems a well-defined disease, but its classification may be difficult. Evidence is weak that an autoimmune process with insulitis causes loss of the beta cells in all patients. Some scientists propose that it may be caused by a virus, increased hygiene or the early introduction of cow's milk or gluten, while views about the nerve supply, vascular function and the beta cells own role tend to be disregarded. Immune interventions have had limited success. There are differences, but also similarities, between T1D and type 2 diabetes (T2D). CONCLUSION Several views on T1D have become so widely accepted that they may actually hamper progress into the true cause of this disease. Research on T1D needs to be carried out with an open mind, and clinicians might be wise to recommend a lifestyle that aims to decrease both the risk of T1D and T2D.
Collapse
Affiliation(s)
- Johnny Ludvigsson
- Division of Pediatrics; Department of Clinical and Experimental Medicine; Linköping University; Linköping Sweden
| |
Collapse
|
26
|
Faustman DL. Why were we wrong for so long? The pancreas of type 1 diabetic patients commonly functions for decades. Diabetologia 2014; 57:1-3. [PMID: 24190583 DOI: 10.1007/s00125-013-3104-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Accepted: 10/16/2013] [Indexed: 01/12/2023]
Abstract
The type 1 diabetes field has held firm to the dogma that the pancreas is no longer viable, and thus incapable of producing insulin, within 1 to 2 years of diagnosis for the majority of patients. A new study in this issue of Diabetologia (DOI: 10.1007/s00125-013-3067-x ), based on a hypersensitive assay, has found detectable C-peptide, a marker of insulin production, in individuals with long-standing type 1 diabetes. This new study confirms and expands a decades-long track record of research finding intact pancreatic islet cells in advanced disease. Because the evidence, stemming back to 1902, was largely histological in nature, it was dismissed as lacking functional corroboration. This new study in patients with long-term diabetes shows appropriate functioning of pancreatic islet cells after exposure to a mixed-meal stimulus. The weight of evidence now makes it clear that a large fraction of patients with long-standing diabetes have low level, but persistent functioning of pancreatic islet cells enduring more than a decade after disease onset.
Collapse
Affiliation(s)
- Denise L Faustman
- Immunobiology Laboratory, Massachusetts General Hospital, Harvard Medical School, Building 149, 13th Street, Room 3602, Boston, MA, 02129, USA,
| |
Collapse
|
27
|
Diaz-de-Durana Y, Lau J, Knee D, Filippi C, Londei M, McNamara P, Nasoff M, DiDonato M, Glynne R, Herman AE. IL-2 immunotherapy reveals potential for innate beta cell regeneration in the non-obese diabetic mouse model of autoimmune diabetes. PLoS One 2013; 8:e78483. [PMID: 24205242 PMCID: PMC3813455 DOI: 10.1371/journal.pone.0078483] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Accepted: 09/12/2013] [Indexed: 01/09/2023] Open
Abstract
Type-1 diabetes (T1D) is an autoimmune disease targeting insulin-producing beta cells, resulting in dependence on exogenous insulin. To date, significant efforts have been invested to develop immune-modulatory therapies for T1D treatment. Previously, IL-2 immunotherapy was demonstrated to prevent and reverse T1D at onset in the non-obese diabetic (NOD) mouse model, revealing potential as a therapy in early disease stage in humans. In the NOD model, IL-2 deficiency contributes to a loss of regulatory T cell function. This deficiency can be augmented with IL-2 or antibody bound to IL-2 (Ab/IL-2) therapy, resulting in regulatory T cell expansion and potentiation. However, an understanding of the mechanism by which reconstituted regulatory T cell function allows for reversal of diabetes after onset is not clearly understood. Here, we describe that Ab/IL-2 immunotherapy treatment, given at the time of diabetes onset in NOD mice, not only correlated with reversal of diabetes and expansion of Treg cells, but also demonstrated the ability to significantly increase beta cell proliferation. Proliferation appeared specific to Ab/IL-2 immunotherapy, as anti-CD3 therapy did not have a similar effect. Furthermore, to assess the effect of Ab/IL-2 immunotherapy well after the development of diabetes, we tested the effect of delaying treatment for 4 weeks after diabetes onset, when beta cells were virtually absent. At this late stage after diabetes onset, Ab/IL-2 treatment was not sufficient to reverse hyperglycemia. However, it did promote survival in the absence of exogenous insulin. Proliferation of beta cells could not account for this improvement as few beta cells remained. Rather, abnormal insulin and glucagon dual-expressing cells were the only insulin-expressing cells observed in islets from mice with established disease. Thus, these data suggest that in diabetic NOD mice, beta cells have an innate capacity for regeneration both early and late in disease, which is revealed through IL-2 immunotherapy.
Collapse
Affiliation(s)
- Yaiza Diaz-de-Durana
- Genetics Department, Genomics Institute of the Novartis Research Foundation, San Diego, California, United States of America
| | - Janet Lau
- Genetics Department, Genomics Institute of the Novartis Research Foundation, San Diego, California, United States of America
| | - Deborah Knee
- Biotherapeutics Department, Genomics Institute of the Novartis Research Foundation, San Diego, California, United States of America
| | - Christophe Filippi
- Genetics Department, Genomics Institute of the Novartis Research Foundation, San Diego, California, United States of America
| | - Marco Londei
- Translational Medicine, Novartis Institutes of Biomedical Research, San Diego, California, United States of America
| | - Peter McNamara
- Pharmacology Department, Genomics Institute of the Novartis Research Foundation, San Diego, California, United States of America
| | - Marc Nasoff
- Biotherapeutics Department, Genomics Institute of the Novartis Research Foundation, San Diego, California, United States of America
| | - Michael DiDonato
- Structural Biology Department, Genomics Institute of the Novartis Research Foundation, San Diego, California, United States of America
| | - Richard Glynne
- Genetics Department, Genomics Institute of the Novartis Research Foundation, San Diego, California, United States of America
- * E-mail:
| | - Ann E. Herman
- Genetics Department, Genomics Institute of the Novartis Research Foundation, San Diego, California, United States of America
| |
Collapse
|
28
|
Kolb H, Lückemeyer K, Heise T, Herder C, Schloot NC, Koenig W, Heinemann L, Martin S. The systemic immune network in recent onset type 1 diabetes: central role of interleukin-1 receptor antagonist (DIATOR Trial). PLoS One 2013; 8:e72440. [PMID: 23991111 PMCID: PMC3753272 DOI: 10.1371/journal.pone.0072440] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Accepted: 07/09/2013] [Indexed: 12/02/2022] Open
Abstract
Background The hypothesis was tested that the systemic immune milieu in recent-onset type 1 diabetes is associated with residual beta cell function and other metabolic patient characteristics. Methods and Findings All patients (n = 89, 40% female) of the Diabetes and Atorvastatin (DIATOR) Trial were analyzed at recruitment, i.e. prior to receiving the study medication. Inclusion criteria were insulin dependent diabetes for 2 weeks to 3 months, age range 18–39 years, and islet cell autoantibodies. Blood samples were analyzed for 14 immune mediators by standard methods. Concentrations of all mediators correlated with at least one other mediator (p<0.05, Spearman correlation) giving rise to a network. Interleukin 1 receptor antagonist (IL1-RA) held a central position and was associated with both pro- and anti-inflammatory mediators. Further central elements were the pro-inflammatory mediators CRP and IL-6, the soluble adhesion molecules sICAM-1 and E-selectin, and MCP-4 which held a central position in the chemokine network. The two Th1-associated mediators IFNγ and IP-10 remained outside the network but correlated with each other. All correlations were positive (r = 0.25–0.72), i.e., high levels of pro-inflammatory mediators were accompanied by increased levels of anti-inflammatory mediators. IL-1RA was the only mediator associated with fasting and liquid mixed meal stimulated C-peptide concentrations (r = 0.31 and 0.24, p = 0.003 and 0.025, after adjustment for age, sex, BMI). There were associations between the immune mediator network and BMI (IL-1RA, CRP, IL-6, MCP-4, MIP-1ß) but few or no associations with HbA1c, insulin dose, lipid parameters, age or sex. Conclusions In patients with recent onset type 1 diabetes, systemic acute phase proteins, cytokines, chemokines and soluble adhesion molecules form a network. Among the few central elements IL-1RA has a dominant role. IL-1RA is associated with all other groups of mediators and is the only mediator which correlates (positively) with residual beta cell function. Trial registration ClinicalTrials.gov registration number: NCT00974740
Collapse
Affiliation(s)
- Hubert Kolb
- West-German Centre of Diabetes and Health, Verbund Katholischer Kliniken Düsseldorf, Düsseldorf, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Abstract
PURPOSE OF REVIEW The assumption that patients with an extended duration of type 1 diabetes mellitus (T1D) do not retain residual functional β cells and endogenous insulin production has recently been challenged. The purpose to this review is to highlight some of the key emerging evidence supporting residual insulin and C-peptide secretion in long-standing T1D. RECENT FINDINGS Recent investigations conducted in a group of type 1 diabetics of long-term duration, characterized clinically and histologically, provided solid evidence to suggest that pancreatic β cells are still present even after 50 years in a majority of these individuals. These residual β cells can secrete insulin in a physiologically regulated manner. Several published reports showed promising effects of glucagon-like peptide 1 (GLP-1) agonists on the glycemic control and residual C-peptide production in long-term T1D, although prospective studies are needed to rule out the potential long-term adverse effects of these drugs. SUMMARY C-peptide is no longer considered an irrelevant by-product of insulin biosynthesis. In-depth basic and translational investigations aimed at understanding the molecular immunology and the pathophysiology are needed to elucidate the mechanisms underlying the residual insulin and C-peptide production in long-term T1D. This may shed light on to the regenerative capacity of β cells, the genetic susceptibility of the mechanisms of resistance to β-cell destruction, and possibly identifying new therapeutic strategies for T1D. Studies evaluating the long-term effects of insulin secretogogue agents along with immune intervention hold promise for their use in future clinical trials for long-term T1D.
Collapse
Affiliation(s)
- Massimo Pietropaolo
- Division of Metabolism, Endocrinology & Diabetes, Department of Internal Medicine, The Brehm Center for Diabetes Research, University of Michigan Medical School, Ann Arbor, Michigan 48105, USA.
| |
Collapse
|
30
|
|
31
|
Driver JP, Chen YG, Mathews CE. Comparative genetics: synergizing human and NOD mouse studies for identifying genetic causation of type 1 diabetes. Rev Diabet Stud 2012; 9:169-87. [PMID: 23804259 DOI: 10.1900/rds.2012.9.169] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Although once widely anticipated to unlock how human type 1 diabetes (T1D) develops, extensive study of the nonobese diabetic (NOD) mouse has failed to yield effective treatments for patients with the disease. This has led many to question the usefulness of this animal model. While criticism about the differences between NOD and human T1D is legitimate, in many cases disease in both species results from perturbations modulated by the same genes or different genes that function within the same biological pathways. Like in humans, unusual polymorphisms within an MHC class II molecule contributes the most T1D risk in NOD mice. This insight supports the validity of this model and suggests the NOD has been improperly utilized to study how to cure or prevent disease in patients. Indeed, clinical trials are far from administering T1D therapeutics to humans at the same concentration ranges and pathological states that inhibit disease in NOD mice. Until these obstacles are overcome it is premature to label the NOD mouse a poor surrogate to test agents that cure or prevent T1D. An additional criticism of the NOD mouse is the past difficulty in identifying genes underlying T1D using conventional mapping studies. However, most of the few diabetogenic alleles identified to date appear relevant to the human disorder. This suggests that rather than abandoning genetic studies in NOD mice, future efforts should focus on improving the efficiency with which diabetes susceptibility genes are detected. The current review highlights why the NOD mouse remains a relevant and valuable tool to understand the genes and their interactions that promote autoimmune diabetes and therapeutics that inhibit this disease. It also describes a new range of technologies that will likely transform how the NOD mouse is used to uncover the genetic causes of T1D for years to come.
Collapse
Affiliation(s)
- John P Driver
- Department of Animal Science, University of Florida, Gainesville, FL 32610, USA
| | | | | |
Collapse
|
32
|
Abstract
The purpose of this article is to provide an overview that summarizes much in the way of our current state of knowledge regarding the pathogenesis and natural history of type 1 diabetes in humans. This information is presented to the reader as a series of seminal historical discoveries that, when advanced through research, transformed our understanding of the roles for the immune system, genes, and environment in the formation of this disease. In addition, where longitudinal investigations of these three facets occurred, their roles within the development of type 1 diabetes, from birth to symptomatic onset and beyond, are discussed, including their most controversial elements. Having an understanding of this disorder's pathogenesis and natural history is key for attempts seeking to understand the issues of what causes type 1 diabetes, as well as to develop a means to prevent and cure the disorder.
Collapse
Affiliation(s)
- Mark A Atkinson
- College of Medicine, Departments of Pathology and Pediatrics, The University of Florida, Gainesville, 32610-0275, USA.
| |
Collapse
|
33
|
Campbell-Thompson M, Wasserfall C, Kaddis J, Albanese-O'Neill A, Staeva T, Nierras C, Moraski J, Rowe P, Gianani R, Eisenbarth G, Crawford J, Schatz D, Pugliese A, Atkinson M. Network for Pancreatic Organ Donors with Diabetes (nPOD): developing a tissue biobank for type 1 diabetes. Diabetes Metab Res Rev 2012; 28:608-17. [PMID: 22585677 PMCID: PMC3456997 DOI: 10.1002/dmrr.2316] [Citation(s) in RCA: 164] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND The Network for Pancreatic Organ Donors with Diabetes (nPOD) was established to recover and characterize pancreata and related organs from cadaveric organ donors with various risk levels for type 1 diabetes (T1D). These biospecimens are available to investigators for collaborative studies aimed at addressing questions related to T1D natural history and pathogenesis. RESEARCH DESIGN AND METHODS Organ donors included T1D patients (new onset to long term), non-diabetic autoantibody-positive subjects, non-diabetic controls and individuals with disorders relevant to β-cell function. Pancreas recovery and transport met transplant-grade criteria. Additional samples recovered included serum, whole blood, spleen and pancreatic and non-pancreatic lymph nodes. Biospecimens were processed for cryopreserved cells, fixed paraffin and fresh frozen blocks and snap frozen samples. T1D autoantibodies, C-peptide levels and high-resolution HLA genotyping for risk alleles were also determined. RESULTS Over 160 donors have been enrolled (ages of 1 day to >90 years). Standard operating procedures were established along with a quality management system. Donor demographics, laboratory assays and histopathological characterizations were shared through an open online informatics system. Biospecimens were distributed to more than 60 investigators. CONCLUSIONS The nPOD programme provides access to high quality biospecimens without cost to investigators. Collaborations and open data sharing are emphasized to maximize research potential of each donor. On the basis of initial successes, the nPOD programme is expanding to recover additional organs relevant to T1D pathogenesis and complications from European countries (PanFin network).
Collapse
Affiliation(s)
- Martha Campbell-Thompson
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610-0275, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Greenbaum CJ, Beam CA, Boulware D, Gitelman SE, Gottlieb PA, Herold KC, Lachin JM, McGee P, Palmer JP, Pescovitz MD, Krause-Steinrauf H, Skyler JS, Sosenko JM. Fall in C-peptide during first 2 years from diagnosis: evidence of at least two distinct phases from composite Type 1 Diabetes TrialNet data. Diabetes 2012; 61:2066-73. [PMID: 22688329 PMCID: PMC3402330 DOI: 10.2337/db11-1538] [Citation(s) in RCA: 250] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2011] [Accepted: 03/09/2012] [Indexed: 12/25/2022]
Abstract
Interpretation of clinical trials to alter the decline in β-cell function after diagnosis of type 1 diabetes depends on a robust understanding of the natural history of disease. Combining data from the Type 1 Diabetes TrialNet studies, we describe the natural history of β-cell function from shortly after diagnosis through 2 years post study randomization, assess the degree of variability between patients, and investigate factors that may be related to C-peptide preservation or loss. We found that 93% of individuals have detectable C-peptide 2 years from diagnosis. In 11% of subjects, there was no significant fall from baseline by 2 years. There was a biphasic decline in C-peptide; the C-peptide slope was -0.0245 pmol/mL/month (95% CI -0.0271 to -0.0215) through the first 12 months and -0.0079 (-0.0113 to -0.0050) from 12 to 24 months (P < 0.001). This pattern of fall in C-peptide over time has implications for understanding trial results in which effects of therapy are most pronounced early and raises the possibility that there are time-dependent differences in pathophysiology. The robust data on the C-peptide obtained under clinical trial conditions should be used in planning and interpretation of clinical trials.
Collapse
|
35
|
Nakayama M, Castoe T, Sosinowski T, He X, Johnson K, Haskins K, Vignali DAA, Gapin L, Pollock D, Eisenbarth GS. Germline TRAV5D-4 T-cell receptor sequence targets a primary insulin peptide of NOD mice. Diabetes 2012; 61:857-65. [PMID: 22315318 PMCID: PMC3314349 DOI: 10.2337/db11-1113] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
There is accumulating evidence that autoimmunity to insulin B chain peptide, amino acids 9-23 (insulin B:9-23), is central to development of autoimmune diabetes of the NOD mouse model. We hypothesized that enhanced susceptibility to autoimmune diabetes is the result of targeting of insulin by a T-cell receptor (TCR) sequence commonly encoded in the germline. In this study, we aimed to demonstrate that a particular Vα gene TRAV5D-4 with multiple junction sequences is sufficient to induce anti-islet autoimmunity by studying retrogenic mouse lines expressing α-chains with different Vα TRAV genes. Retrogenic NOD strains expressing Vα TRAV5D-4 α-chains with many different complementarity determining region (CDR) 3 sequences, even those derived from TCRs recognizing islet-irrelevant molecules, developed anti-insulin autoimmunity. Induction of insulin autoantibodies by TRAV5D-4 α-chains was abrogated by the mutation of insulin peptide B:9-23 or that of two amino acid residues in CDR1 and 2 of the TRAV5D-4. TRAV13-1, the human ortholog of murine TRAV5D-4, was also capable of inducing in vivo anti-insulin autoimmunity when combined with different murine CDR3 sequences. Targeting primary autoantigenic peptides by simple germline-encoded TCR motifs may underlie enhanced susceptibility to the development of autoimmune diabetes.
Collapse
Affiliation(s)
- Maki Nakayama
- Barbara Davis Center for Childhood Diabetes, University of ColoradoSchool of Medicine, Aurora, CO, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
Diabetes affects today an estimated 366 million people world-wide, including 20 million to 40 million of patients with type 1 diabetes (T1D). While T1D accounts for 5% to 20% of those with diabetes, it is associated with higher morbidity, mortality and health care cost than the more prevalent type 2 diabetes. Patients with T1D require exogenous insulin for survival and should be identified as soon as possible after diagnosis to avoid high morbidity due to a delay in insulin treatment. It is also important to present to the patient correct prognosis that differs by the type of diabetes. From the research point of view, correct classification should help to identify the etiologies and to develop specific prevention for T1D. This review summarizes evidence that may be helpful in diagnosing T1D in various ethnic groups. Challenges in interpretation of results commonly used to determine the type of diabetes are highlighted.
Collapse
Affiliation(s)
- Marian Rewers
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, CO, USA
| |
Collapse
|
37
|
Farris AB, Basturk O, Adsay NV. Pancreatitis, Other Inflammatory Lesions, and Pancreatic Pseudotumors. Surg Pathol Clin 2011; 4:625-650. [PMID: 26837491 DOI: 10.1016/j.path.2011.03.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
The pancreas is versatile in the diversity of disorders that it can exhibit. In this article, characteristics of disorders such as chronic, autoimmune, eosinophilic, hereditary, and infectious pancreatitis are described. With regard to autoimmune pancreatitis, the role of clinical evaluation, histologic examination, and IgG4 immunohistochemistry is discussed. The role of pancreatitis in the pathogenesis of diabetes is also mentioned. Some implications of pancreatitis are highlighted, including the neoplastic predisposition caused by inflammatory lesions of the pancreas. The goal of this article is to convey an appreciation of these disorders because their recognition can benefit patients tremendously, as inflammatory lesions of the pancreas can be mass-forming, giving rise to pseudotumors, and leading to surgical resection that may otherwise be unnecessary.
Collapse
Affiliation(s)
- Alton B Farris
- Department of Pathology and Laboratory Medicine, Emory University Hospital, Emory University, 1364 Clifton Road Northeast, Room H-188, Atlanta, GA 30322, USA.
| | - Olca Basturk
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - N Volkan Adsay
- Department of Pathology and Laboratory Medicine, Emory University, 1364 Clifton Road NE, Room H-180B, Atlanta, GA 30322, USA
| |
Collapse
|
38
|
Abstract
This unit contains detailed protocols for the simultaneous identification of the human pancreatic β cells and determination of their viability by flow cytometry. The enumeration of β cells is based on the ability of the cell-permeable form of the zinc-selective dye, FluoZin-3-AM, to bind intracellular labile zinc stored at higher levels in these cells than any other types of cells in the body. Although staining of intracellular labile zinc by FluoZin-3-AM is dependent on the metabolic activity of β cells, co-staining with a mitochondrial transmembrane potential indicator allows the accurate determination of viability. Simultaneous measurement of intracellular antioxidant thiols is also compatible with the detection of β cells containing metabolically active mitochondria. The method for assessing the mitochondrial functionality by flow cytometry described herein is simple to perform and sufficient to detect the viability of β cells in human islet preparations.
Collapse
|
39
|
Pugliese A, Reijonen HK, Nepom J, Burke GW. Recurrence of autoimmunity in pancreas transplant patients: research update. ACTA ACUST UNITED AC 2011; 1:229-238. [PMID: 21927622 DOI: 10.2217/dmt.10.21] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Type 1 diabetes is an autoimmune disorder leading to loss of pancreatic β-cells and insulin secretion, followed by insulin dependence. Islet and whole pancreas transplantation restore insulin secretion. Pancreas transplantation is often performed together with a kidney transplant in patients with end-stage renal disease. With improved immunosuppression, immunological failures of whole pancreas grafts have become less frequent and are usually categorized as chronic rejection. However, growing evidence indicates that chronic islet autoimmunity may eventually lead to recurrent diabetes, despite immunosuppression to prevent rejection. Thus, islet autoimmunity should be included in the diagnostic work-up of graft failure and ideally should be routinely assessed pretransplant and on follow-up in Type 1 diabetes recipients of pancreas and islet cell transplants. There is a need to develop new treatment regimens that can control autoimmunity, as this may not be effectively suppressed by conventional immunosuppression.
Collapse
Affiliation(s)
- Alberto Pugliese
- Diabetes Research Institute, University of Miami Miller School of Medicine, 1450 NW 10th Avenue, Miami, FL 33136, USA
| | | | | | | |
Collapse
|
40
|
Christensen DP, Dahllöf M, Lundh M, Rasmussen DN, Nielsen MD, Billestrup N, Grunnet LG, Mandrup-Poulsen T. Histone deacetylase (HDAC) inhibition as a novel treatment for diabetes mellitus. Mol Med 2011; 17:378-90. [PMID: 21274504 DOI: 10.2119/molmed.2011.00021] [Citation(s) in RCA: 189] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2011] [Accepted: 01/24/2011] [Indexed: 12/13/2022] Open
Abstract
Both common forms of diabetes have an inflammatory pathogenesis in which immune and metabolic factors converge on interleukin-1β as a key mediator of insulin resistance and β-cell failure. In addition to improving insulin resistance and preventing β-cell inflammatory damage, there is evidence of genetic association between diabetes and histone deacetylases (HDACs); and HDAC inhibitors (HDACi) promote β-cell development, proliferation, differentiation and function and positively affect late diabetic microvascular complications. Here we review this evidence and propose that there is a strong rationale for preclinical studies and clinical trials with the aim of testing the utility of HDACi as a novel therapy for diabetes.
Collapse
Affiliation(s)
- Dan P Christensen
- Center for Medical Research Methodology, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Tauriainen S, Salmela K, Rantala I, Knip M, Hyöty H. Collecting high-quality pancreatic tissue for experimental study from organ donors with signs of β-cell autoimmunity. Diabetes Metab Res Rev 2010; 26:585-92. [PMID: 20848387 DOI: 10.1002/dmrr.1129] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
BACKGROUND The aim of this study was to create a new research strategy to obtain high-quality pancreatic tissues from subjects with preclinical or clinical type 1 diabetes, which would open up new avenues for studying the mechanisms of the β-cell damaging process in humans. RESEARCH DESIGN AND METHODS A nationwide collaboration network (the PanFin network) was established in Finland to start an on-call screening of diabetes-associated autoantibodies from deceased organ donors and subsequent processing of pancreases from autoantibody-positive donors. This protocol was integrated into the national organ transplantation procedure. RESULTS Only a few modifications were needed to the normal transplantation practices. One additional blood sample was obtained from donors for autoantibody analyses, the transplantation team was informed about the autoantibody result and the pancreas of autoantibody-positive donors was transported to the core laboratory. Altogether, 307 donors were screened and 22 (7.2%) were positive for at least one autoantibody and 3 tested positive for two or more autoantibodies out of the five tested (islet cell antibodies, insulin autoantibodies and autoantibodies to glutamic acid decarboxylase, islet antigen 2 and zinc transporter 8). The quality of collected pancreatic tissue was superior to that from autopsies and allowed the detection of both RNA and proteins. CONCLUSIONS The study protocol was proven feasible to be carried out on a nationwide scale. It did not interfere with the normal transplantation activities and provided valuable tissue material for research.
Collapse
Affiliation(s)
- Sisko Tauriainen
- Department of Virology, University of Tampere, Medical School, Tampere, Finland.
| | | | | | | | | |
Collapse
|
42
|
Bonifacio E, Ziegler AG. Advances in the prediction and natural history of type 1 diabetes. Endocrinol Metab Clin North Am 2010; 39:513-25. [PMID: 20723817 DOI: 10.1016/j.ecl.2010.05.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Type 1 diabetes (T1D) has the hallmark characteristics of autoimmunity superimposed on genetic susceptibility. Both genes (HLA) and immune markers (autoantibodies) have been validated as predictive markers of the subsequent development of the disease in higher-risk relatives and the lower-risk general population. Over the last three decades, using a combination of genes, immune, and metabolic markers, clinicians are now able to quantify an individual's disease risk from 1 in 100,000 to more than 1 in 2. This article reviews these biomarkers and T1D prediction strategies, and discusses potential implications of prediction and natural history for the pathogenesis of T1D.
Collapse
Affiliation(s)
- Ezio Bonifacio
- Deutsche Forschungsgemeinschaft Center for Regenerative Therapies Dresden, Technische Universität Dresden, Tatzberg 47/49, 01307 Dresden, Germany.
| | | |
Collapse
|
43
|
Willcox A, Richardson SJ, Bone AJ, Foulis AK, Morgan NG. Evidence of increased islet cell proliferation in patients with recent-onset type 1 diabetes. Diabetologia 2010; 53:2020-8. [PMID: 20532863 DOI: 10.1007/s00125-010-1817-6] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2010] [Accepted: 05/12/2010] [Indexed: 12/29/2022]
Abstract
AIMS/HYPOTHESIS In adults, the rate of beta cell replication is normally very low, but recent evidence suggests that it may increase during insulitis. We therefore studied tissue from donors with recent-onset type 1 diabetes to establish whether islet cell proliferation is increased during the disease process. METHODS Paraffin-embedded pancreatic sections from ten donors with recent-onset type 1 diabetes and a range of relevant controls were stained by immunohistochemical techniques with antibodies against the proliferation markers Ki67 and minichromosome maintenance protein-2 (MCM-2). A combination staining technique involving immunoperoxidase and immunofluorescence methods was developed to quantify the numbers of alpha and beta cells with Ki67-positive nuclei and to investigate the relationship between insulitis and islet cell proliferation. RESULTS In non-diabetic control donors, only 1.1 +/- 0.3% (mean +/- SEM) of islets contained one or more Ki67(+) islet cells, whereas this proportion was increased markedly in recent-onset type 1 diabetes (10.88 +/- 2.5%; p < 0.005). An equivalent increase in Ki67(+) staining occurred in alpha and beta cells and was correlated positively with the presence of insulitis. A significant increase in the labelling of islet cells from type 1 diabetic donors was also seen when MCM-2 staining was employed. Increased islet cell proliferation was not evident in three donors with longer duration type 1 diabetes or in ten type 2 diabetic donors. CONCLUSIONS/INTERPRETATION Alpha and beta cells undergo a marked increase in proliferation during the progression of type 1 diabetes in humans. The results imply that islet cell proliferation is re-initiated in response to the autoimmune attack associated with type 1 diabetes.
Collapse
Affiliation(s)
- A Willcox
- Institute of Biomedical and Clinical Science, Peninsula College of Medicine and Dentistry (University of Exeter), Tamar Science Park, Derriford, Plymouth, UK
| | | | | | | | | |
Collapse
|
44
|
Villasenor A, Wang ZV, Rivera LB, Ocal O, Asterholm IW, Scherer PE, Brekken RA, Cleaver O, Wilkie TM. Rgs16 and Rgs8 in embryonic endocrine pancreas and mouse models of diabetes. Dis Model Mech 2010; 3:567-80. [PMID: 20616094 DOI: 10.1242/dmm.003210] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Diabetes is characterized by the loss, or gradual dysfunction, of insulin-producing pancreatic beta-cells. Although beta-cells can replicate in younger adults, the available diabetes therapies do not specifically target beta-cell regeneration. Novel approaches are needed to discover new therapeutics and to understand the contributions of endocrine progenitors and beta-cell regeneration during islet expansion. Here, we show that the regulators of G protein signaling Rgs16 and Rgs8 are expressed in pancreatic progenitor and endocrine cells during development, then extinguished in adults, but reactivated in models of both type 1 and type 2 diabetes. Exendin-4, a glucagon-like peptide 1 (Glp-1)/incretin mimetic that stimulates beta-cell expansion, insulin secretion and normalization of blood glucose levels in diabetics, also promoted re-expression of Rgs16::GFP within a few days in pancreatic ductal-associated cells and islet beta-cells. These findings show that Rgs16::GFP and Rgs8::GFP are novel and early reporters of G protein-coupled receptor (GPCR)-stimulated beta-cell expansion after therapeutic treatment and in diabetes models. Rgs16 and Rgs8 are likely to control aspects of islet progenitor cell activation, differentiation and beta-cell expansion in embryos and metabolically stressed adults.
Collapse
Affiliation(s)
- Alethia Villasenor
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9148, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Rowe PA, Campbell-Thompson ML, Schatz DA, Atkinson MA. The pancreas in human type 1 diabetes. Semin Immunopathol 2010; 33:29-43. [PMID: 20495921 PMCID: PMC3022158 DOI: 10.1007/s00281-010-0208-x] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2010] [Accepted: 04/13/2010] [Indexed: 11/24/2022]
Abstract
Type 1 diabetes (T1D) is considered a disorder whose pathogenesis is autoimmune in origin, a notion drawn in large part from studies of human pancreata performed as far back as the 1960s. While studies of the genetics, epidemiology, and peripheral immunity in T1D have been subject to widespread analysis over the ensuing decades, efforts to understand the disorder through analysis of human pancreata have been far more limited. We have reviewed the published literature pertaining to the pathology of the human pancreas throughout all stages in the natural history of T1D. This effort uncovered a series of findings that challenge many dogmas ascribed to T1D and revealed data suggesting the marked heterogeneity in terms of its pathology. An improved understanding and appreciation for pancreatic pathology in T1D could lead to improved disease classification, an understanding of why the disorder occurs, and better therapies for disease prevention and management.
Collapse
Affiliation(s)
- Patrick A Rowe
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, 1600 SW Archer Road, Gainesville, FL 32610, USA
| | | | | | | |
Collapse
|
46
|
Abstract
Enteroviruses are believed to contribute to the pathogenesis of type 1 diabetes mellitus (T1DM). In this Review, the interplay between infection with enteroviruses, the immune system and host genes is discussed. Data from retrospective and prospective epidemiological studies strongly suggest the involvement of enteroviruses, such as coxsackievirus B, in the development of T1DM. Enteroviral RNA and/or proteins can be detected in tissues of patients with T1DM. Isolation of coxsackievirus B4 from the pancreas of patients with T1DM or the presence of enteroviral components in their islets strengthens the hypothesis of a relationship between the virus and the disease. Enteroviruses can play a part in the early phase of T1DM through the infection of beta cells and the activation of innate immunity and inflammation. In contrast with its antiviral role, virus-induced interferon alpha can be deleterious, acting as an initiator of the autoimmunity directed against beta cells. Enteroviruses, through persistent and/or successive infections, can interact with the adaptive immune system. Host genes, such as IFIH1, that influence susceptibility to T1DM are associated with antiviral activities. An increased activity of the IFIH1 protein may promote the development of T1DM. An improved knowledge of the pathogenic mechanisms of enterovirus infections should help to uncover preventive strategies for T1DM.
Collapse
Affiliation(s)
- Didier Hober
- Laboratoire de Virologie/EA3610 "Pathogenèse Virale du Diabète de Type 1", Faculté de Médecine, Université Lille 2, CHRU Lille, Centre de Biologie et Pathologie et Institut Hippocrate Parc Eurasanté, Boulevard du Prof J Leclercq, 59037 Lille Cedex, France
| | | |
Collapse
|
47
|
Zauli G, Toffoli B, di Iasio MG, Celeghini C, Fabris B, Secchiero P. Treatment with recombinant tumor necrosis factor-related apoptosis-inducing ligand alleviates the severity of streptozotocin-induced diabetes. Diabetes 2010; 59:1261-5. [PMID: 20185810 PMCID: PMC2857907 DOI: 10.2337/db09-1771] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVE To evaluate the potential therapeutic effect of recombinant human tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) treatment in a model of type 1 diabetes. RESEARCH DESIGN AND METHODS Recombinant TRAIL was added in vitro to primary human and mouse peripheral blood mononuclear cells (PBMCs) and isolated human islets to evaluate the expression of the immunoregulatory gene SOCS1. Diabetes was induced by five consecutive daily injections of low-concentration (50 mg/kg) streptozotocin (STZ) in C57 black mice (n = 24). A group of these mice (n = 12) was co-injected with recombinant TRAIL (20 microg/day) for 5 days, and the diabetic status (glycemia and body weight) was followed over time. After 6 weeks, circulating levels of insulin, TNF-alpha, and osteoprotegerin (OPG) were measured, and animals were killed to perform the histological analysis of the pancreas. RESULTS The in vitro exposure of both PBMCs and human islets to recombinant TRAIL significantly upregulated the expression of SOCS1. With respect to STZ-treated animals, mice co-injected with STZ+TRAIL were characterized by 1) lower levels of hyperglycemia, 2) higher levels of body weight and insulinemia, 3) a partial preservation of pancreatic islets with normal morphology, and 4) a lower expression of both systemic (TNF-alpha and OPG) and pancreatic (vascular cell adhesion molecule [VCAM]-1) inflammatory markers. CONCLUSIONS Overall, these data demonstrate that the administration of recombinant TRAIL ameliorates the severity of STZ-induced type 1 diabetes, and this effect was accompanied by the upregulation of SOCS1 expression.
Collapse
Affiliation(s)
- Giorgio Zauli
- Department of Morphology and Embryology, University of Ferrara, Ferrara, Italy.
| | | | | | | | | | | |
Collapse
|
48
|
Abstract
PURPOSE OF REVIEW Type 1 diabetes (T1D) is an autoimmune disease typically believed to result from malfunctions in adaptive immune response signaling which result in activation of self-reactive T cells. However, recent research has indicated components of the innate immune response as having a key role in the initiation of the autoimmune process of T1D. This review will highlight recent studies which examined the role of innate immune response signaling and the connections to T1D pathogenesis. RECENT FINDINGS Investigations indicate that components of innate immunity, including inflammation and Toll-like receptor signaling, are involved in pancreatic islet infiltration and insulitis. Recent studies examining the role of viral infections in T1D development also implicate innate immune response signaling in disease pathogenesis. SUMMARY Current research indicates that components of innate immune response signaling are involved in the initiation of the autoimmune process which results in the eventual destruction of beta cells during T1D pathogenesis. Continuing efforts by researchers to uncover the molecular pathways of innate immunity linked to T1D development could potentially lead to therapeutics capable of preventing and curing the autoimmune disease.
Collapse
Affiliation(s)
| | | | - Rita Bortell
- Correspondence to: Rita Bortell, PhD; Diabetes Division, Suite 218, 373 Plantation Street, Worcester, MA 01605, Tel: 508-856-3788; fax. 508-856-4093;
| |
Collapse
|
49
|
Coppieters KT, von Herrath MG. Histopathology of type 1 diabetes: old paradigms and new insights. Rev Diabet Stud 2009; 6:85-96. [PMID: 19806238 DOI: 10.1900/rds.2009.6.85] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Although our knowledge on the various aspects of diabetes development in the NOD mouse model is substantial and keeps expanding at a dramatic pace, the dataset on histopathologic features of type 1 diabetes (T1D) in patients remains largely stagnant. Early work has established an array of common aspects that have become epitomic in the absence of new patient material. There is a growing consensus that an updated and more detailed view is required that challenges and expands our understanding. Comprehensive initiatives are currently ongoing to address these issues in pre-diabetic, recent onset and longstanding type 1 diabetic individuals, and some of the old data have been recently revisited. In this review article, we wish to provide an overview of where we stand today and how we can correlate the various cross-sectional studies from the past with contemporary models of the disease. We believe an enhanced understanding of the many histopathological particularities in patients as compared to animal models will ultimately lead, not only to more fundamental insights, but also to an improved ability to translate pre-clinical data from bench to bedside.
Collapse
Affiliation(s)
- Ken T Coppieters
- La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | | |
Collapse
|
50
|
|