1
|
Yang Q, Jin L, Luo M, Xie S. Current status, trend changes, and future predictions of the disease burden of type 1 diabetes kidney disease in global and China. Front Endocrinol (Lausanne) 2025; 16:1559363. [PMID: 40166677 PMCID: PMC11955482 DOI: 10.3389/fendo.2025.1559363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Accepted: 02/21/2025] [Indexed: 04/02/2025] Open
Abstract
Objective This study analyzes the global and China trends in the prevalence, disease burden, and future projections of Type 1 Diabetic Nephropathy (T1DN) over the past three decades, providing data to inform public health policies and clinical interventions. Methods Data from the Global Burden of Disease (GBD) 2021 database were used to analyze the incidence, prevalence, mortality, years lived with disability (YLDs), years of life lost (YLLs), and disability-adjusted life years (DALYs) of T1DN globally and in China from 1990 to 2021. Trend analysis was conducted using R and Joinpoint software, and the ARIMA model was applied to predict future trends in T1DN prevalence for the next 20 years. A significance level of p<0.05 was applied. Results Globally, deaths from T1DN increased from 49,300(95% CI: 39,088-61,207) in 1990 to 94,020 (95% CI: 71,456-119,984)in 2021, with the age-standardized mortality rate remaining stable. DALYs rose from 2,227,518(95% CI: 1,835,372-2,679,207) in 1990 to 3,875,628 (95% CI: 3,062,395-4,845,503) in 2021, though the age-standardized rate slightly decreased. In China, the mortality rate declined significantly, and DALYs decreased, with the age-standardized DALYs dropping from 80.915/100,000(95% CI: 65.121-98.391)to 47.953/100,000(95% CI: 36.9-60.734). Globally, both incidence and prevalence increased, with global incidence reaching 95,140(95% CI: 82,236-111,471) cases and prevalence rising to 6,295,711 (95% CI: 5,459,693-7,114,345)cases. In China, incidence showed a declining trend, but prevalence continued to rise. The ARIMA model forecasts global incidence will reach 115,000 cases, with prevalence reaching 7,000,000 by 2041. In China, incidence is expected to stabilize, while prevalence may increase to approximately 2,500,000 cases. Conclusion The burden of T1DN is rising globally, especially in terms of prevalence, while China has made progress in reducing mortality and disease burden. However, challenges remain in chronic disease management. Over the next 20 years, global prevalence is projected to continue increasing, while China's prevalence may stabilize. Targeted interventions for different age groups and genders will be essential in reducing the T1DN burden.
Collapse
Affiliation(s)
- Qinghua Yang
- Department of Endocrinology, Panzhihua Central Hospital, Panzhihua, China
| | - Li Jin
- Department of Endocrinology, Panzhihua Central Hospital, Panzhihua, China
| | - Mingwei Luo
- Department of Medical Records Statistics, Panzhihua Central Hospital, Panzhihua, China
| | - Shiwei Xie
- Department of Orthopedics, Panzhihua Central Hospital, Panzhihua, China
| |
Collapse
|
2
|
Casu A, Grippo PJ, Wasserfall C, Sun Z, Linsley PS, Hamerman JA, Fife BT, Lacy-Hulbert A, Toledo FGS, Hart PA, Papachristou GI, Bellin MD, Yadav D, Laughlin MR, Goodarzi MO, Speake C. Evaluating the Immunopathogenesis of Diabetes After Acute Pancreatitis in the Diabetes RElated to Acute Pancreatitis and Its Mechanisms Study: From the Type 1 Diabetes in Acute Pancreatitis Consortium. Pancreas 2022; 51:580-585. [PMID: 36206462 PMCID: PMC9555855 DOI: 10.1097/mpa.0000000000002076] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
ABSTRACT The association between acute pancreatitis (AP) and diabetes mellitus (DM) has long been established, with the initial descriptions of AP patients presenting with DM after a bout of AP published in the 1940s and 50s. However, the potential mechanisms involved, particularly those components related to the immune system, have not been well defined. The Diabetes RElated to Acute pancreatitis and its Mechanisms (DREAM) study is a multicenter clinical study designed to understand the frequency and phenotype of DM developing after AP. This article describes one objective of the DREAM study: to determine the immunologic mechanisms of DM after AP, including the contribution of β-cell autoimmunity. This component of the study will assess the presence of islet autoimmunity, as well as the magnitude and kinetics of the innate and adaptive immune response at enrollment and during longitudinal follow-up after 1 or more episodes of AP. Finally, DREAM will evaluate the relationship between immune features, DM development, and pancreatitis etiology and severity.
Collapse
Affiliation(s)
- Anna Casu
- From the Translational Research Institute, AdventHealth Orlando, Orlando, FL
| | - Paul J Grippo
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Illinois-Chicago, Chicago, IL
| | - Clive Wasserfall
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL
| | - Zhaoli Sun
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Peter S Linsley
- Center for Systems Immunology, Benaroya Research Institute at Virginia Mason, Seattle, WA
| | - Jessica A Hamerman
- Center for Fundamental Immunology, Benaroya Research Institute at Virginia Mason, Seattle, WA
| | - Brian T Fife
- Department of Medicine, Center for Immunology, University of Minnesota, Minneapolis, MN
| | - Adam Lacy-Hulbert
- Center for Fundamental Immunology, Benaroya Research Institute at Virginia Mason, Seattle, WA
| | - Frederico G S Toledo
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Phil A Hart
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Georgios I Papachristou
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH
| | | | - Dhiraj Yadav
- Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Maren R Laughlin
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD
| | - Mark O Goodarzi
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Cate Speake
- Diabetes Clinical Research Program, Center for Interventional Immunology, Benaroya Research Institute at Virginia Mason, Seattle, WA
| |
Collapse
|
3
|
Herzog K, Andersson T, Grill V, Hammar N, Malmström H, Talbäck M, Walldius G, Carlsson S. Alterations in Biomarkers Related to Glycemia, Lipid Metabolism, and Inflammation up to 20 Years Before Diagnosis of Type 1 Diabetes in Adults: Findings From the AMORIS Cohort. Diabetes Care 2022; 45:330-338. [PMID: 34876530 PMCID: PMC8914411 DOI: 10.2337/dc21-1238] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 11/09/2021] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Type 1 diabetes is described to have an acute onset, but autoantibodies can appear several years preceding diagnosis. This suggests a long preclinical phase, which may also include metabolic parameters. Here we assessed whether elevations in glycemic, lipid, and other metabolic biomarkers were associated with future type 1 diabetes risk in adults. RESEARCH DESIGN AND METHODS We studied 591,239 individuals from the Swedish AMORIS cohort followed from 1985-1996 to 2012. Through linkage to national patient, diabetes, and prescription registers, we identified incident type 1 diabetes. Using Cox regression models, we estimated hazard ratios for biomarkers at baseline and incident type 1 diabetes. We additionally assessed trajectories of biomarkers during the 25 years before type 1 diabetes diagnosis in a nested case-control design. RESULTS We identified 1,122 type 1 diabetes cases during follow-up (average age of patient at diagnosis: 53.3 years). The biomarkers glucose, fructosamine, triglycerides, the ratio of apolipoprotein (apo)B to apoA-I, uric acid, alkaline phosphatase, and BMI were positively associated with type 1 diabetes risk. Higher apoA-I was associated with lower type 1 diabetes incidence. Already 15 years before diagnosis, type 1 diabetes cases had higher mean glucose, fructosamine, triglycerides, and uric acid levels compared with control subjects. CONCLUSIONS Alterations in biomarker levels related to glycemia, lipid metabolism, and inflammation are associated with clinically diagnosed type 1 diabetes risk, and these may be elevated many years preceding diagnosis.
Collapse
Affiliation(s)
- Katharina Herzog
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Tomas Andersson
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.,Centre for Occupational and Environmental Medicine, Stockholm County Council, Stockholm, Sweden
| | - Valdemar Grill
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Niklas Hammar
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Håkan Malmström
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.,R&D, Swedish Orphan Biovitrum AB, Stockholm, Sweden
| | - Mats Talbäck
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Göran Walldius
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Sofia Carlsson
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
4
|
B Cells and Microbiota in Autoimmunity. Int J Mol Sci 2021; 22:ijms22094846. [PMID: 34063669 PMCID: PMC8125537 DOI: 10.3390/ijms22094846] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 04/27/2021] [Indexed: 12/12/2022] Open
Abstract
Trillions of microorganisms inhabit the mucosal membranes maintaining a symbiotic relationship with the host's immune system. B cells are key players in this relationship because activated and differentiated B cells produce secretory immunoglobulin A (sIgA), which binds commensals to preserve a healthy microbial ecosystem. Mounting evidence shows that changes in the function and composition of the gut microbiota are associated with several autoimmune diseases suggesting that an imbalanced or dysbiotic microbiota contributes to autoimmune inflammation. Bacteria within the gut mucosa may modulate autoimmune inflammation through different mechanisms from commensals ability to induce B-cell clones that cross-react with host antigens or through regulation of B-cell subsets' capacity to produce cytokines. Commensal signals in the gut instigate the differentiation of IL-10 producing B cells and IL-10 producing IgA+ plasma cells that recirculate and exert regulatory functions. While the origin of the dysbiosis in autoimmunity is unclear, compelling evidence shows that specific species have a remarkable influence in shaping the inflammatory immune response. Further insight is necessary to dissect the complex interaction between microorganisms, genes, and the immune system. In this review, we will discuss the bidirectional interaction between commensals and B-cell responses in the context of autoimmune inflammation.
Collapse
|
5
|
Tetz G, Brown SM, Hao Y, Tetz V. Type 1 Diabetes: an Association Between Autoimmunity, the Dynamics of Gut Amyloid-producing E. coli and Their Phages. Sci Rep 2019; 9:9685. [PMID: 31273267 PMCID: PMC6609616 DOI: 10.1038/s41598-019-46087-x] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 06/17/2019] [Indexed: 12/14/2022] Open
Abstract
The etiopathogenesis of type 1 diabetes (T1D), a common autoimmune disorder, is not completely understood. Recent studies suggested the gut microbiome plays a role in T1D. We have used public longitudinal microbiome data from T1D patients to analyze amyloid-producing bacterial composition and found a significant association between initially high amyloid-producing Escherichia coli abundance, subsequent E. coli depletion prior to seroconversion, and T1D development. In children who presented seroconversion or developed T1D, we observed an increase in the E. coli phage/E. coli ratio prior to E. coli depletion, suggesting that the decrease in E. coli was due to prophage activation. Evaluation of the role of phages in amyloid release from E. coli biofilms in vitro suggested an indirect role of the bacterial phages in the modulation of host immunity. This study for the first time suggests that amyloid-producing E. coli, their phages, and bacteria-derived amyloid might be involved in pro-diabetic pathway activation in children at risk for T1D.
Collapse
Affiliation(s)
- George Tetz
- Human Microbiology Institute, New York, NY, 10013, USA. .,Tetz Laboratories, New York, NY, 10027, USA.
| | - Stuart M Brown
- New York University School of Medicine, Department of Cell Biology, New York, NY, 10016, USA
| | - Yuhan Hao
- Center for Genomics and Systems Biology, New York University, New York, New York, 10012, USA.,New York Genome Center, New York, New York, 10013, USA
| | - Victor Tetz
- Human Microbiology Institute, New York, NY, 10013, USA
| |
Collapse
|
6
|
Pan W, Zhang Y, Zeng C, Xu F, Yan J, Weng J. miR-192 is upregulated in T1DM, regulates pancreatic β-cell development and inhibits insulin secretion through suppressing GLP-1 expression. Exp Ther Med 2018; 16:2717-2724. [PMID: 30186503 DOI: 10.3892/etm.2018.6453] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 03/09/2018] [Indexed: 02/06/2023] Open
Abstract
MicroRNAs (miRs) post-translationally regulate gene expression by specifically binding to the mRNA of their target genes. The aim of the present study was to determine the effect of miR-192 on pancreatic β-cell development. The serum levels of miR-192 in type 1 diabetes mellitus (T1DM) and streptozotocin-induced rats were determined, and were revealed to be elevated compared with those in healthy patients and normal rats, respectively. Western blot and reverse transcription-quantitative polymerase chain reaction analysis indicated that miR-192 suppressed the expression of glucagon-like peptide-1 (GLP-1), a potent insulin secretagogue. Ectopic expression of miR-192 inhibited cell proliferation and promoted apoptosis of NIT-1 cells, while miR-192 inhibitor had the opposite effect. Collectively, the present results revealed that miR-192 was elevated in T1DM, and is implicated in pancreatic β-cell development through regulation of cell proliferation and apoptosis, thereby suppressing insulin secretion. Furthermore, miR-192 suppressed GLP-1 expression, thereby further promoting T1DM. The present study suggested that miR-192 is a novel molecular target for the management or prevention of T1DM.
Collapse
Affiliation(s)
- Wen Pan
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Yanan Zhang
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Chun Zeng
- Department of Emergency, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Fen Xu
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Jinhua Yan
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Jianping Weng
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, P.R. China
| |
Collapse
|
7
|
Felton JL, Maseda D, Bonami RH, Hulbert C, Thomas JW. Anti-Insulin B Cells Are Poised for Antigen Presentation in Type 1 Diabetes. THE JOURNAL OF IMMUNOLOGY 2018; 201:861-873. [PMID: 29950508 DOI: 10.4049/jimmunol.1701717] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 05/23/2018] [Indexed: 12/29/2022]
Abstract
Early breaches in B cell tolerance are central to type 1 diabetes progression in mouse and man. Conventional BCR transgenic mouse models (VH125.Tg NOD) reveal the power of B cell specificity to drive disease as APCs. However, in conventional fixed IgM models, comprehensive assessment of B cell development is limited. To provide more accurate insight into the developmental and functional fates of anti-insulin B cells, we generated a new NOD model (VH125SDNOD) in which anti-insulin VDJH125 is targeted to the IgH chain locus to generate a small (1-2%) population of class switch-competent insulin-binding B cells. Tracking of this rare population in a polyclonal repertoire reveals that anti-insulin B cells are preferentially skewed into marginal zone and late transitional subsets known to have increased sensitivity to proinflammatory signals. Additionally, IL-10 production, characteristic of regulatory B cell subsets, is increased. In contrast to conventional models, class switch-competent anti-insulin B cells proliferate normally in response to mitogenic stimuli but remain functionally silent for insulin autoantibody production. Diabetes development is accelerated, which demonstrates the power of anti-insulin B cells to exacerbate disease without differentiation into Ab-forming or plasma cells. Autoreactive T cell responses in VH125SDNOD mice are not restricted to insulin autoantigens, as evidenced by increased IFN-γ production to a broad array of diabetes-associated epitopes. Together, these results independently validate the pathogenic role of anti-insulin B cells in type 1 diabetes, underscore their diverse developmental fates, and demonstrate the pathologic potential of coupling a critical β cell specificity to predominantly proinflammatory Ag-presenting B cell subsets.
Collapse
Affiliation(s)
- Jamie L Felton
- Division of Pediatric Endocrinology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Damian Maseda
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN 37232; and
| | - Rachel H Bonami
- Division of Rheumatology and Immunology, Department of Medicine, Vanderbilt University, Nashville, TN 37232
| | - Chrys Hulbert
- Division of Rheumatology and Immunology, Department of Medicine, Vanderbilt University, Nashville, TN 37232
| | - James W Thomas
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN 37232; and .,Division of Rheumatology and Immunology, Department of Medicine, Vanderbilt University, Nashville, TN 37232
| |
Collapse
|
8
|
Sebastiani G, Nigi L, Grieco GE, Mancarella F, Ventriglia G, Dotta F. Circulating microRNAs and diabetes mellitus: a novel tool for disease prediction, diagnosis, and staging? J Endocrinol Invest 2017; 40:591-610. [PMID: 28213644 DOI: 10.1007/s40618-017-0611-4] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 01/03/2017] [Indexed: 12/19/2022]
Abstract
Diabetes is a complex, multifactorial group of metabolic diseases characterized by chronic hyperglycaemia due to pancreatic beta-cell dysfunction and/or loss. It is characterized by an asymptomatic and highly variable prodromic phase, which renders diabetes mellitus difficult to be predicted with sufficient accuracy. Despite several efforts in the identification and standardization of newly trustable. Biomarkers able to predict and follow-up diabetes and to specifically subtype its different forms, few of them have proven of clinical utility. Recently, a new class of endogenous non-coding small RNAs, namely microRNAs, have been indicated as putative biomarkers, being released by cells and tissues and found in a cell-free circulating form in many biological fluids, including serum and/or plasma. MicroRNAs have been initially identified as promising biomarkers in cancer, and nowadays their application has been extended to other diseases, including diabetes. Although an increasing number of studies focused on the evaluation of circulating microRNAs in diabetes, few reproducibly identified microRNAs as biomarkers for disease prediction or follow-up. Technological problems as well as the need to obtain highly standardized operating procedures and methods are still an issue in such research field. In this review, we comprehensively resume the main and most recent findings on circulating microRNAs, and their possible use as biomarkers to predict and follow-up diabetes and its complications, as well as the methodological challenges to standardize accurate operating procedures for their analysis.
Collapse
Affiliation(s)
- G Sebastiani
- Diabetes Unit, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
- Fondazione Umberto di Mario ONLUS, Toscana Life Sciences, Siena, Italy
| | - L Nigi
- Diabetes Unit, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
- Fondazione Umberto di Mario ONLUS, Toscana Life Sciences, Siena, Italy
| | - G E Grieco
- Diabetes Unit, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
- Fondazione Umberto di Mario ONLUS, Toscana Life Sciences, Siena, Italy
| | - F Mancarella
- Diabetes Unit, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
- Fondazione Umberto di Mario ONLUS, Toscana Life Sciences, Siena, Italy
| | - G Ventriglia
- Diabetes Unit, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
- Fondazione Umberto di Mario ONLUS, Toscana Life Sciences, Siena, Italy
| | - F Dotta
- Diabetes Unit, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy.
- Fondazione Umberto di Mario ONLUS, Toscana Life Sciences, Siena, Italy.
| |
Collapse
|
9
|
Inducing Specific Immune Tolerance to Prevent Type 1 Diabetes in NOD Mice. Pancreas 2016; 45:882-8. [PMID: 26784909 DOI: 10.1097/mpa.0000000000000603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
OBJECTIVES Proinsulin is the first autoantigen in type 1 diabetes (T1D). We reasoned that coupling hematopoietic stem cells (HSCs) transplantation with ex vivo transduction of syngeneic HSCs with lentiviral vectors to express proinsulin II could prevent T1D in nonobese diabetic (NOD) mice. METHODS Hematopoietic stem cells were isolated from 6- to 8-week-old NOD female mice and transduced in vitro with lentiviral vectors encoding proinsulin II. Preconditioned 3- to 4-week-old female NOD mice were transplanted with transduced or nontransduced HSCs and compared with age-matched unmanipulated control. The insulitis, T1D development, and immune reconstitution were assessed. RESULTS The mean (SD) insulitis score was significantly reduced (1.156 [0.575] vs 2.156 [0.892] or 3.043 [0.728], P = 0.009 or <0.001), and diabetes was nearly completely prevented (1/13 vs 5/12 or 4/9, P = 0.031 or 0.013) in recipients of transduced HSCs expressing proinsulin II as compared with recipients of nontransduced HSCs or unmanipulated control. Sialitis, reconstitution of peripheral blood leukocytes, and in vitro recall responses to ovalbumin were not different between 3 groups of mice. CONCLUSIONS Syngeneic transplantation of HSCs transduced ex vivo with lentiviral vectors to encode proinsulin II is a novel strategy to prevent T1D.
Collapse
|
10
|
Yu C, Burns JC, Robinson WH, Utz PJ, Ho PP, Steinman L, Frey AB. Identification of Candidate Tolerogenic CD8(+) T Cell Epitopes for Therapy of Type 1 Diabetes in the NOD Mouse Model. J Diabetes Res 2016; 2016:9083103. [PMID: 27069933 PMCID: PMC4812430 DOI: 10.1155/2016/9083103] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 02/16/2016] [Indexed: 12/31/2022] Open
Abstract
Type 1 diabetes is an autoimmune disease in which insulin-producing pancreatic islet β cells are the target of self-reactive B and T cells. T cells reactive with epitopes derived from insulin and/or IGRP are critical for the initiation and maintenance of disease, but T cells reactive with other islet antigens likely have an essential role in disease progression. We sought to identify candidate CD8(+) T cell epitopes that are pathogenic in type 1 diabetes. Proteins that elicit autoantibodies in human type 1 diabetes were analyzed by predictive algorithms for candidate epitopes. Using several different tolerizing regimes using synthetic peptides, two new predicted tolerogenic CD8(+) T cell epitopes were identified in the murine homolog of the major human islet autoantigen zinc transporter ZnT8 (aa 158-166 and 282-290) and one in a non-β cell protein, dopamine β-hydroxylase (aa 233-241). Tolerizing vaccination of NOD mice with a cDNA plasmid expressing full-length proinsulin prevented diabetes, whereas plasmids encoding ZnT8 and DβH did not. However, tolerizing vaccination of NOD mice with the proinsulin plasmid in combination with plasmids expressing ZnT8 and DβH decreased insulitis and enhanced prevention of disease compared to vaccination with the plasmid encoding proinsulin alone.
Collapse
MESH Headings
- Animals
- Autoantibodies/immunology
- CD8-Positive T-Lymphocytes/immunology
- Cation Transport Proteins/genetics
- Cation Transport Proteins/immunology
- Cells, Cultured
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/pathology
- Diabetes Mellitus, Type 1/therapy
- Disease Models, Animal
- Dopamine beta-Hydroxylase/genetics
- Dopamine beta-Hydroxylase/immunology
- Epitopes, T-Lymphocyte
- Female
- Genetic Therapy/methods
- Humans
- Immune Tolerance
- Insulin-Secreting Cells/immunology
- Insulin-Secreting Cells/pathology
- Lymphocyte Activation
- Mice, Inbred NOD
- Proinsulin/genetics
- Proinsulin/immunology
- Time Factors
- Vaccination
- Vaccines, Subunit/genetics
- Vaccines, Subunit/immunology
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
- Zinc Transporter 8
Collapse
Affiliation(s)
- Cailin Yu
- Department of Cell Biology, New York University Langone School of Medicine, 550 First Avenue, New York, NY 10016, USA
| | - Jeremy C. Burns
- Department of Cell Biology, New York University Langone School of Medicine, 550 First Avenue, New York, NY 10016, USA
| | - William H. Robinson
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Geriatric Research Education and Clinical Center, Veterans Affairs, Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | - Paul J. Utz
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Peggy P. Ho
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Lawrence Steinman
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Alan B. Frey
- Department of Cell Biology, New York University Langone School of Medicine, 550 First Avenue, New York, NY 10016, USA
- *Alan B. Frey:
| |
Collapse
|
11
|
Shao L, Zhou HJ, Zhang H, Qin L, Hwa J, Yun Z, Ji W, Min W. SENP1-mediated NEMO deSUMOylation in adipocytes limits inflammatory responses and type-1 diabetes progression. Nat Commun 2015; 6:8917. [PMID: 26596471 PMCID: PMC4662081 DOI: 10.1038/ncomms9917] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Accepted: 10/15/2015] [Indexed: 02/08/2023] Open
Abstract
Adipocyte dysfunction correlates with the development of diabetes. Here we show that mice with a adipocyte-specific deletion of the SUMO-specific protease SENP1 gene develop symptoms of type-1 diabetes mellitus (T1DM), including hyperglycaemia and glucose intolerance with mild insulin resistance. Peri-pancreatic adipocytes from SENP1-deficient mice exhibit heightened NF-κB activity and production of proinflammatory cytokines, which induce CCL5 expression in adjacent pancreatic islets and direct cytotoxic effects on pancreatic islets. Mechanistic studies show that SENP1 deletion in adipocytes enhances SUMOylation of the NF-κB essential molecule, NEMO, at lysine 277/309, leading to increased NF-κB activity, cytokine production and pancreatic inflammation. We further show that NF-κB inhibitors could inhibit pre-diabetic cytokine production, β-cell damages and ameliorate the T1DM phenotype in SENP1-deficient mice. Feeding a high-fat diet augments both type-1 and type-2 diabetes phenotypes in SENP1-deficient mice, consistent with the effects on adipocyte-derived NF-κB and cytokine signalling. Our study reveals previously unrecognized mechanism regulating the onset and progression of T1DM associated with adipocyte dysfunction.
Collapse
Affiliation(s)
- Lan Shao
- The First Affiliated Hospital, Center for Translational Medicine, Sun Yat-sen University, Guangzhou, China
- Department of Pathology, Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine, 10 Amistad St, New Haven, Connecticut 06520, USA
| | - Huanjiao Jenny Zhou
- Department of Pathology, Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine, 10 Amistad St, New Haven, Connecticut 06520, USA
| | - Haifeng Zhang
- Department of Pathology, Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine, 10 Amistad St, New Haven, Connecticut 06520, USA
| | - Lingfeng Qin
- Department of Pathology, Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine, 10 Amistad St, New Haven, Connecticut 06520, USA
| | - John Hwa
- Department of Internal Medicine and Section of Cardiology, Yale University School of Medicine, New Haven, Connecticut 06510, USA
| | - Zhong Yun
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, Connecticut 06510, USA
| | - Weidong Ji
- The First Affiliated Hospital, Center for Translational Medicine, Sun Yat-sen University, Guangzhou, China
| | - Wang Min
- The First Affiliated Hospital, Center for Translational Medicine, Sun Yat-sen University, Guangzhou, China
- Department of Pathology, Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine, 10 Amistad St, New Haven, Connecticut 06520, USA
| |
Collapse
|
12
|
Stechova K, Kolar M, Blatny R, Halbhuber Z, Vcelakova J, Hubackova M, Petruzelkova L, Sumnik Z, Obermannova B, Pithova P, Stavikova V, Krivjanska M, Neuwirth A, Kolouskova S, Filipp D. Healthy first-degree relatives of patients with type 1 diabetes exhibit significant differences in basal gene expression pattern of immunocompetent cells compared to controls: expression pattern as predeterminant of autoimmune diabetes. Scand J Immunol 2015; 75:210-9. [PMID: 21923738 DOI: 10.1111/j.1365-3083.2011.02637.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Expression features of genetic landscape which predispose an individual to the type 1 diabetes are poorly understood. We addressed this question by comparing gene expression profile of freshly isolated peripheral blood mononuclear cells isolated from either patients with type 1 diabetes (T1D), or their first-degree relatives or healthy controls. Our aim was to establish whether a distinct type of 'prodiabetogenic' gene expression pattern in the group of relatives of patients with T1D could be identified. Whole-genome expression profile of nine patients with T1D, their ten first-degree relatives and ten healthy controls was analysed using the human high-density expression microarray chip. Functional aspects of candidate genes were assessed using the MetaCore software. The highest number of differentially expressed genes (547) was found between the autoantibody-negative healthy relatives and the healthy controls. Some of them represent genes critically involved in the regulation of innate immune responses such as TLR signalling and CCR3 signalling in eosinophiles, humoral immune reactions such as BCR pathway, costimulation and cytokine responses mediated by CD137, CD40 and CD28 signalling and IL-1 proinflammatory pathway. Our data demonstrate that expression profile of healthy relatives of patients with T1D is clearly distinct from the pattern found in the healthy controls. That especially concerns differential activation status of genes and signalling pathways involved in proinflammatory processes and those of innate immunity and humoral reactivity. Thus, we posit that the study of the healthy relative's gene expression pattern is instrumental for the identification of novel markers associated with the development of diabetes.
Collapse
Affiliation(s)
- K Stechova
- Department of Paediatrics, 2nd Medical Faculty of Charles University and University Hospital Motol, Prague, Czech RepublicLaboratory of Genomics and Bioinformatics, Institute of Molecular Genetics AS CR, Prague, Czech RepublicCentral European Biosystems, Prague, Czech RepublicDepartment of Internal Medicine, 2nd Medical Faculty of Charles University and University Hospital Motol, Prague, Czech RepublicDepartment of Immunobiology, Institute of Molecular Genetics, Czech Academy of Science, Prague, Czech Republic
| | - M Kolar
- Department of Paediatrics, 2nd Medical Faculty of Charles University and University Hospital Motol, Prague, Czech RepublicLaboratory of Genomics and Bioinformatics, Institute of Molecular Genetics AS CR, Prague, Czech RepublicCentral European Biosystems, Prague, Czech RepublicDepartment of Internal Medicine, 2nd Medical Faculty of Charles University and University Hospital Motol, Prague, Czech RepublicDepartment of Immunobiology, Institute of Molecular Genetics, Czech Academy of Science, Prague, Czech Republic
| | - R Blatny
- Department of Paediatrics, 2nd Medical Faculty of Charles University and University Hospital Motol, Prague, Czech RepublicLaboratory of Genomics and Bioinformatics, Institute of Molecular Genetics AS CR, Prague, Czech RepublicCentral European Biosystems, Prague, Czech RepublicDepartment of Internal Medicine, 2nd Medical Faculty of Charles University and University Hospital Motol, Prague, Czech RepublicDepartment of Immunobiology, Institute of Molecular Genetics, Czech Academy of Science, Prague, Czech Republic
| | - Z Halbhuber
- Department of Paediatrics, 2nd Medical Faculty of Charles University and University Hospital Motol, Prague, Czech RepublicLaboratory of Genomics and Bioinformatics, Institute of Molecular Genetics AS CR, Prague, Czech RepublicCentral European Biosystems, Prague, Czech RepublicDepartment of Internal Medicine, 2nd Medical Faculty of Charles University and University Hospital Motol, Prague, Czech RepublicDepartment of Immunobiology, Institute of Molecular Genetics, Czech Academy of Science, Prague, Czech Republic
| | - J Vcelakova
- Department of Paediatrics, 2nd Medical Faculty of Charles University and University Hospital Motol, Prague, Czech RepublicLaboratory of Genomics and Bioinformatics, Institute of Molecular Genetics AS CR, Prague, Czech RepublicCentral European Biosystems, Prague, Czech RepublicDepartment of Internal Medicine, 2nd Medical Faculty of Charles University and University Hospital Motol, Prague, Czech RepublicDepartment of Immunobiology, Institute of Molecular Genetics, Czech Academy of Science, Prague, Czech Republic
| | - M Hubackova
- Department of Paediatrics, 2nd Medical Faculty of Charles University and University Hospital Motol, Prague, Czech RepublicLaboratory of Genomics and Bioinformatics, Institute of Molecular Genetics AS CR, Prague, Czech RepublicCentral European Biosystems, Prague, Czech RepublicDepartment of Internal Medicine, 2nd Medical Faculty of Charles University and University Hospital Motol, Prague, Czech RepublicDepartment of Immunobiology, Institute of Molecular Genetics, Czech Academy of Science, Prague, Czech Republic
| | - L Petruzelkova
- Department of Paediatrics, 2nd Medical Faculty of Charles University and University Hospital Motol, Prague, Czech RepublicLaboratory of Genomics and Bioinformatics, Institute of Molecular Genetics AS CR, Prague, Czech RepublicCentral European Biosystems, Prague, Czech RepublicDepartment of Internal Medicine, 2nd Medical Faculty of Charles University and University Hospital Motol, Prague, Czech RepublicDepartment of Immunobiology, Institute of Molecular Genetics, Czech Academy of Science, Prague, Czech Republic
| | - Z Sumnik
- Department of Paediatrics, 2nd Medical Faculty of Charles University and University Hospital Motol, Prague, Czech RepublicLaboratory of Genomics and Bioinformatics, Institute of Molecular Genetics AS CR, Prague, Czech RepublicCentral European Biosystems, Prague, Czech RepublicDepartment of Internal Medicine, 2nd Medical Faculty of Charles University and University Hospital Motol, Prague, Czech RepublicDepartment of Immunobiology, Institute of Molecular Genetics, Czech Academy of Science, Prague, Czech Republic
| | - B Obermannova
- Department of Paediatrics, 2nd Medical Faculty of Charles University and University Hospital Motol, Prague, Czech RepublicLaboratory of Genomics and Bioinformatics, Institute of Molecular Genetics AS CR, Prague, Czech RepublicCentral European Biosystems, Prague, Czech RepublicDepartment of Internal Medicine, 2nd Medical Faculty of Charles University and University Hospital Motol, Prague, Czech RepublicDepartment of Immunobiology, Institute of Molecular Genetics, Czech Academy of Science, Prague, Czech Republic
| | - P Pithova
- Department of Paediatrics, 2nd Medical Faculty of Charles University and University Hospital Motol, Prague, Czech RepublicLaboratory of Genomics and Bioinformatics, Institute of Molecular Genetics AS CR, Prague, Czech RepublicCentral European Biosystems, Prague, Czech RepublicDepartment of Internal Medicine, 2nd Medical Faculty of Charles University and University Hospital Motol, Prague, Czech RepublicDepartment of Immunobiology, Institute of Molecular Genetics, Czech Academy of Science, Prague, Czech Republic
| | - V Stavikova
- Department of Paediatrics, 2nd Medical Faculty of Charles University and University Hospital Motol, Prague, Czech RepublicLaboratory of Genomics and Bioinformatics, Institute of Molecular Genetics AS CR, Prague, Czech RepublicCentral European Biosystems, Prague, Czech RepublicDepartment of Internal Medicine, 2nd Medical Faculty of Charles University and University Hospital Motol, Prague, Czech RepublicDepartment of Immunobiology, Institute of Molecular Genetics, Czech Academy of Science, Prague, Czech Republic
| | - M Krivjanska
- Department of Paediatrics, 2nd Medical Faculty of Charles University and University Hospital Motol, Prague, Czech RepublicLaboratory of Genomics and Bioinformatics, Institute of Molecular Genetics AS CR, Prague, Czech RepublicCentral European Biosystems, Prague, Czech RepublicDepartment of Internal Medicine, 2nd Medical Faculty of Charles University and University Hospital Motol, Prague, Czech RepublicDepartment of Immunobiology, Institute of Molecular Genetics, Czech Academy of Science, Prague, Czech Republic
| | - A Neuwirth
- Department of Paediatrics, 2nd Medical Faculty of Charles University and University Hospital Motol, Prague, Czech RepublicLaboratory of Genomics and Bioinformatics, Institute of Molecular Genetics AS CR, Prague, Czech RepublicCentral European Biosystems, Prague, Czech RepublicDepartment of Internal Medicine, 2nd Medical Faculty of Charles University and University Hospital Motol, Prague, Czech RepublicDepartment of Immunobiology, Institute of Molecular Genetics, Czech Academy of Science, Prague, Czech Republic
| | - S Kolouskova
- Department of Paediatrics, 2nd Medical Faculty of Charles University and University Hospital Motol, Prague, Czech RepublicLaboratory of Genomics and Bioinformatics, Institute of Molecular Genetics AS CR, Prague, Czech RepublicCentral European Biosystems, Prague, Czech RepublicDepartment of Internal Medicine, 2nd Medical Faculty of Charles University and University Hospital Motol, Prague, Czech RepublicDepartment of Immunobiology, Institute of Molecular Genetics, Czech Academy of Science, Prague, Czech Republic
| | - D Filipp
- Department of Paediatrics, 2nd Medical Faculty of Charles University and University Hospital Motol, Prague, Czech RepublicLaboratory of Genomics and Bioinformatics, Institute of Molecular Genetics AS CR, Prague, Czech RepublicCentral European Biosystems, Prague, Czech RepublicDepartment of Internal Medicine, 2nd Medical Faculty of Charles University and University Hospital Motol, Prague, Czech RepublicDepartment of Immunobiology, Institute of Molecular Genetics, Czech Academy of Science, Prague, Czech Republic
| |
Collapse
|
13
|
Engman C, Wen Y, Meng WS, Bottino R, Trucco M, Giannoukakis N. Generation of antigen-specific Foxp3+ regulatory T-cells in vivo following administration of diabetes-reversing tolerogenic microspheres does not require provision of antigen in the formulation. Clin Immunol 2015; 160:103-23. [PMID: 25773782 DOI: 10.1016/j.clim.2015.03.004] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 03/05/2015] [Indexed: 11/20/2022]
Abstract
We have developed novel antisense oligonucleotide-formulated microspheres that can reverse hyperglycemia in newly-onset diabetic mice. Dendritic cells taking up the microspheres adopt a restrained co-stimulation ability and migrate to the pancreatic lymph nodes when injected into an abdominal region that is drained by those lymph nodes. Furthermore, we demonstrate that the absolute numbers of antigen-specific Foxp3+ T regulatory cells are increased only in the lymph nodes draining the site of administration and that these T-cells proliferate independently of antigen supply in the microspheres. Taken together, our data add to the emerging model where antigen supply may not be a requirement in "vaccines" for autoimmune disease, but the site of administration - subserved by lymph nodes draining the target organ - is in fact critical to foster the generation of antigen-specific regulatory cells. The implications of these observations on "vaccine" design for autoimmunity are discussed and summarized.
Collapse
MESH Headings
- Animals
- B7-1 Antigen/genetics
- B7-2 Antigen/genetics
- Blood Glucose/drug effects
- CD11c Antigen/metabolism
- CD40 Antigens/genetics
- Cell Movement/drug effects
- Cell Proliferation/drug effects
- Cells, Cultured
- Dendritic Cells/immunology
- Diabetes Mellitus, Experimental/therapy
- Diabetes Mellitus, Type 1/therapy
- Female
- Forkhead Transcription Factors/analysis
- Gene Knockdown Techniques
- Hyperglycemia/therapy
- Leukocyte Common Antigens/metabolism
- Lymph Nodes/cytology
- Lymph Nodes/immunology
- Lymphocyte Activation/immunology
- Macaca fascicularis
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C3H
- Mice, Inbred NOD
- Mice, Transgenic
- Microspheres
- Oligonucleotides, Antisense/genetics
- Pancreas/immunology
- Receptors, Antigen, T-Cell/genetics
- T-Lymphocytes, Regulatory/cytology
- Vaccines/administration & dosage
- Vaccines/immunology
Collapse
Affiliation(s)
- Carl Engman
- Institute of Cellular Therapeutics, 11th Floor South Tower, Allegheny Health Network, 320 East North Avenue, Pittsburgh, PA, 15212, USA.
| | - Yi Wen
- Division of Pharmaceutical Sciences, Mylan School of Pharmacy, Mellon 413, 600 Forbes Avenue, Pittsburgh, PA 15282, USA
| | - Wilson S Meng
- Division of Pharmaceutical Sciences, Mylan School of Pharmacy, Mellon 413, 600 Forbes Avenue, Pittsburgh, PA 15282, USA.
| | - Rita Bottino
- Institute of Cellular Therapeutics, 11th Floor South Tower, Allegheny Health Network, 320 East North Avenue, Pittsburgh, PA, 15212, USA.
| | - Massimo Trucco
- Institute of Cellular Therapeutics, 11th Floor South Tower, Allegheny Health Network, 320 East North Avenue, Pittsburgh, PA, 15212, USA.
| | - Nick Giannoukakis
- Institute of Cellular Therapeutics, 11th Floor South Tower, Allegheny Health Network, 320 East North Avenue, Pittsburgh, PA, 15212, USA.
| |
Collapse
|
14
|
Ustinova J, Zusinaite E, Utt M, Metsküla K, Reimand K, Huchaiah V, Merits A, Uibo R. Development of a luciferase-based system for the detection of ZnT8 autoantibodies. J Immunol Methods 2014; 405:67-73. [DOI: 10.1016/j.jim.2014.01.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Revised: 12/19/2013] [Accepted: 01/14/2014] [Indexed: 01/10/2023]
|
15
|
Bonifacio E, Krumsiek J, Winkler C, Theis FJ, Ziegler AG. A strategy to find gene combinations that identify children who progress rapidly to type 1 diabetes after islet autoantibody seroconversion. Acta Diabetol 2014; 51:403-11. [PMID: 24249616 DOI: 10.1007/s00592-013-0526-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Accepted: 10/19/2013] [Indexed: 01/21/2023]
Abstract
We recently developed a novel approach capable of identifying gene combinations to obtain maximal disease risk stratification. Type 1 diabetes has a preclinical phase including seroconversion to autoimmunity and subsequent progression to diabetes. Here, we applied our gene combination approach to identify combinations that contribute either to islet autoimmunity or to the progression from islet autoantibodies to diabetes onset. We examined 12 type 1 diabetes susceptibility genes (INS, ERBB3, PTPN2, IFIH1, PTPN22, KIAA0350, CD25, CTLA4, SH2B3, IL2, IL18RAP, IL10) in a cohort of children of parents with type 1 diabetes and prospectively followed from birth. The most predictive combination was subsequently applied to a smaller validation cohort. The combinations of genes only marginally contributed to the risk of developing islet autoimmunity, but could substantially modify risk of progression to diabetes in islet autoantibody-positive children. The greatest discrimination was provided by risk allele scores of five genes, INS, IFIH1, IL18RAP, CD25, and IL2 genes, which could identify 80 % of islet autoantibody-positive children who progressed to diabetes within 6 years of seroconversion and discriminate high risk (63 % within 6 years; 95 % CI 45-81 %) and low risk (11 % within 6 years; 95 % CI 0.1-22 %; p = 4 × 10(-5)) antibody-positive children. Risk stratification by these five genes was confirmed in a second cohort of islet autoantibody children. These findings highlight genes that may affect the rate of the beta-cell destruction process once autoimmunity has initiated and may help to identify islet autoantibody-positive subjects with rapid progression to diabetes.
Collapse
Affiliation(s)
- Ezio Bonifacio
- Center for Regenerative Therapies Dresden, Technische Universität Dresden, Fetscherstrasse 105, 01307, Dresden, Germany,
| | | | | | | | | |
Collapse
|
16
|
Kikkas I, Mallone R, Tubiana-Rufi N, Chevenne D, Carel JC, Créminon C, Volland H, Boitard C, Morel N. A simple and fast non-radioactive bridging immunoassay for insulin autoantibodies. PLoS One 2013; 8:e69021. [PMID: 23922678 PMCID: PMC3726704 DOI: 10.1371/journal.pone.0069021] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Accepted: 06/04/2013] [Indexed: 11/18/2022] Open
Abstract
Type 1 diabetes (T1D) is an autoimmune disease which results from the destruction of pancreatic beta cells. Autoantibodies directed against islet antigens are valuable diagnostic tools. Insulin autoantibodies (IAAs) are usually the first to appear and also the most difficult to detect amongst the four major islet autoantibodies. A non-radioactive IAA bridging ELISA was developed to this end. In this assay, one site of the IAAs from serum samples is bound to a hapten-labeled insulin (GC300-insulin), which is subsequently captured on anti-GC300 antibody-coated 96-well plates. The other site of the IAAs is bound to biotinylated insulin, allowing the complex to be detected by an enzyme-streptavidin conjugate. In the present study, 50 serum samples from patients with newly diagnosed T1D and 100 control sera from non-diabetic individuals were analyzed with our new assay and the results were correlated with an IAA radioimmunoassay (RIA). Using IAA bridging ELISA, IAAs were detected in 32 out of 50 T1D children, whereas with IAA RIA, 41 out of 50 children with newly diagnosed T1D were scored as positive. In conclusion, the IAA bridging ELISA could serve as an attractive approach for rapid and automated detection of IAAs in T1D patients for diagnostic purposes.
Collapse
Affiliation(s)
- Ingrid Kikkas
- Commisariat à l'Energie Atomique, iBiTec-S, Service de Pharmacologie et d'Immunoanalyse, Commisariat à l'Energie Atomique Saclay, Gif sur Yvette, France
| | - Roberto Mallone
- Institut National de la Santé et de la Recherche Médicale, U1016, Cochin Institute, DeAR Lab Avenir, Paris, France
- Paris Descartes University, Sorbonne Paris Cité, Faculté de Médecine, Paris, France
- Assistance Publique Hôpitaux de Paris, Hôtel Dieu, Service de Diabétologie, Paris, France
| | - Nadia Tubiana-Rufi
- University Paris, Hôpital Robert Debré, Pediatric Endocrinology, Paris, France
| | - Didier Chevenne
- University Paris, Hôpital Robert Debré, Pediatric Endocrinology, Paris, France
| | - Jean Claude Carel
- University Paris, Hôpital Robert Debré, Pediatric Endocrinology, Paris, France
| | - Christophe Créminon
- Commisariat à l'Energie Atomique, iBiTec-S, Service de Pharmacologie et d'Immunoanalyse, Commisariat à l'Energie Atomique Saclay, Gif sur Yvette, France
| | - Hervé Volland
- Commisariat à l'Energie Atomique, iBiTec-S, Service de Pharmacologie et d'Immunoanalyse, Commisariat à l'Energie Atomique Saclay, Gif sur Yvette, France
| | - Christian Boitard
- Institut National de la Santé et de la Recherche Médicale, U1016, Cochin Institute, DeAR Lab Avenir, Paris, France
- Paris Descartes University, Sorbonne Paris Cité, Faculté de Médecine, Paris, France
- Assistance Publique Hôpitaux de Paris, Hôtel Dieu, Service de Diabétologie, Paris, France
| | - Nathalie Morel
- Commisariat à l'Energie Atomique, iBiTec-S, Service de Pharmacologie et d'Immunoanalyse, Commisariat à l'Energie Atomique Saclay, Gif sur Yvette, France
- * E-mail:
| |
Collapse
|
17
|
Vcelakova J, Blatny R, Halbhuber Z, Kolar M, Neuwirth A, Petruzelkova L, Ulmannova T, Kolouskova S, Sumnik Z, Pithova P, Krivjanska M, Filipp D, Stechova K. The effect of diabetes-associated autoantigens on cell processes in human PBMCs and their relevance to autoimmune diabetes development. J Diabetes Res 2013; 2013:589451. [PMID: 23841104 PMCID: PMC3694381 DOI: 10.1155/2013/589451] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Accepted: 05/20/2013] [Indexed: 12/18/2022] Open
Abstract
Type 1 Diabetes (T1D) is considered to be a T-helper- (Th-) 1 autoimmune disease; however, T1D pathogenesis likely involves many factors, and sufficient tools for autoreactive T cell detection for the study of this disease are currently lacking. In this study, using gene expression microarrays, we analysed the effect of diabetes-associated autoantigens on peripheral blood mononuclear cells (PBMCs) with the purpose of identifying (pre)diabetes-associated cell processes. Twelve patients with recent onset T1D, 18 first-degree relatives of the TD1 patients (DRL; 9/18 autoantibody positive), and 13 healthy controls (DV) were tested. PBMCs from these individuals were stimulated with a cocktail of diabetes-associated autoantigens (proinsulin, IA-2, and GAD65-derived peptides). After 72 hours, gene expression was evaluated by high-density gene microarray. The greatest number of functional differences was observed between relatives and controls (69 pathways), from which 15% of the pathways belonged to "immune response-related" processes. In the T1D versus controls comparison, more pathways (24%) were classified as "immune response-related." Important pathways that were identified using data from the T1D versus controls comparison were pathways involving antigen presentation by MHCII, the activation of Th17 and Th22 responses, and cytoskeleton rearrangement-related processes. Genes involved in Th17 and TGF-beta cascades may represent novel, promising (pre)diabetes biomarkers.
Collapse
Affiliation(s)
- Jana Vcelakova
- Department of Paediatrics, 2nd Faculty of Medicine, Charles University in Prague and University Hospital Motol, V Uvalu 84, 15006 Prague, Czech Republic.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Winkler C, Schober E, Ziegler AG, Holl RW. Markedly reduced rate of diabetic ketoacidosis at onset of type 1 diabetes in relatives screened for islet autoantibodies. Pediatr Diabetes 2012; 13:308-13. [PMID: 22060727 DOI: 10.1111/j.1399-5448.2011.00829.x] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2011] [Accepted: 09/21/2011] [Indexed: 12/31/2022] Open
Abstract
OBJECTIVE To determine whether screening for islet autoantibodies in children prevents ketoacidosis and other metabolic complications at diabetes onset and improves the clinical course after diagnosis. SUBJECTS AND METHODS The German BABYDIAB and the Munich Family Study follow children with a first-degree family history of type 1 diabetes for the development of islet autoantibodies and type 1 diabetes. The Diabetes Prospective Documentation (DPV) Initiative registers and collects information on pediatric patients with type 1 diabetes throughout Germany. Here, clinical characteristics at diabetes onset [ketoacidosis, mean hemoglobin A1c (HbA1c), and length of hospitalization] and the 5-yr clinical course (HbA1c and insulin dose) of screened and followed islet autoantibody-positive children (n = 101) and 49 883 non-screened children within the DPV registry were compared. RESULTS At diabetes onset, children who were followed after screening and were positive for islet autoantibodies had lower HbA1c (8.6 vs. 11%, p < 0.001) and a lower prevalence of diabetic ketoacidosis (3.3 vs. 29.1%, p < 0.001). Screened children also had a shorter hospitalization period at onset (11.4 vs. 14.9 d, p = 0.005). Similar results were observed when the analysis was restricted to 759 non-screened DPV children with a first-degree family history of type 1 diabetes. No differences between screened and non-screened children were observed with respect to HbA1c and insulin dose during the first 5 yr after diagnosis. CONCLUSIONS Screening for islet autoantibodies in children likely leads to earlier diabetes diagnosis resulting in less complications at diagnosis. However, no substantial benefit in the clinical outcome during the first 5 yr after diagnosis was observed.
Collapse
Affiliation(s)
- Christiane Winkler
- Institute of Diabetes Research, Helmholtz Center Munich, Neuherberg, Germany
| | | | | | | |
Collapse
|
19
|
Genetic Determination and Immunopathogenesis of Type 1 Diabetes Mellitus in Humans. ACTA MEDICA MARTINIANA 2012. [DOI: 10.2478/v10201-011-0034-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
20
|
Abstract
Type 1 diabetes (T1D) is an autoimmune disease characterized by the lack of insulin due to an autoimmune destruction of pancreatic beta cells. Here, we report a unique case of a family with naturally conceived quadruplets in which T1D was diagnosed in two quadruplets simultaneously. At the same time, the third quadruplet was diagnosed with the pre-diabetic stage. Remarkably, all four quadruplets were positive for anti-islet cell antibodies, GAD65 and IA-A2. Monozygotic status of the quadruplets was confirmed by testing 14 different short tandem repeat polymorphisms. Serological examination confirmed that all quadruplets and their father suffered from a recent enteroviral infection of EV68-71 serotype. To assess the nature of the molecular pathological processes contributing to the development of diabetes, immunocompetent cells isolated from all family members were characterized by gene expression arrays, immune-cell enumerations and cytokine-production assays. The microarray data provided evidence that viral infection, and IL-27 and IL-9 cytokine signalling contributed to the onset of T1D in two of the quadruplets. The propensity of stimulated immunocompetent cells from non-diabetic members of the family to secrete high level of IFN-α further corroborates this conclusion. The number of T regulatory cells as well as plasmacytoid and/or myeloid dendritic cells was found diminished in all family members. Thus, this unique family is a prime example for the support of the so-called 'fertile-field' hypothesis proposing that genetic predisposition to anti-islet autoimmunity is 'fertilized' and precipitated by a viral infection leading to a fully blown T1D.
Collapse
|
21
|
Michels AW, Eisenbarth GS. Immune intervention in type 1 diabetes. Semin Immunol 2011; 23:214-9. [PMID: 21852151 DOI: 10.1016/j.smim.2011.07.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2011] [Accepted: 07/10/2011] [Indexed: 12/23/2022]
Abstract
Type 1 diabetes (T1D) is a chronic autoimmune disease that results in the specific immune destruction of insulin producing beta cells. Currently there is no cure for T1D and treatment for the disease consists of lifelong administration of insulin. Immunotherapies aimed at preventing beta cell destruction in T1D patients with residual c-peptide or in individuals developing T1D are being evaluated. Networks of researchers such as TrialNet and the Immune Tolerance Network in the U.S. and similar networks in Europe have been established to evaluate such immunotherapies. This review focuses on immune intervention for the prevention and amelioration of human T1D with a focus on potential immune suppressive, antigen specific and environmental therapies.
Collapse
Affiliation(s)
- Aaron W Michels
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, CO 80045, USA
| | | |
Collapse
|
22
|
Petrich de Marquesini LG, Fu J, Connor KJ, Bishop AJ, McLintock NE, Pope C, Wong FS, Dayan CM. IFN-gamma and IL-10 islet-antigen-specific T cell responses in autoantibody-negative first-degree relatives of patients with type 1 diabetes. Diabetologia 2010; 53:1451-60. [PMID: 20369219 DOI: 10.1007/s00125-010-1739-3] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2009] [Accepted: 02/08/2010] [Indexed: 01/13/2023]
Abstract
AIMS/HYPOTHESIS Islet antibody-negative first-degree relatives of type 1 diabetes patients have a very low risk of developing diabetes. We studied the balance between IFN-gamma (proinflammatory) and IL-10 (regulatory) T cell responses in these participants. METHODS Peripheral blood T cells from adult (18-50 years old, n = 40) DRB1*0401-positive first-degree relatives negative for GAD and tyrosine phosphatase-like insulinoma antigen 2 (IA-2) antibodies were tested for IFN-gamma and IL-10 responses in a sensitive cytokine enzyme-linked immunospot assay against a panel of seven peptide epitopes derived from IA-2 and proinsulin. Comparison was made with HLA-matched newly diagnosed type 1 diabetic patients (n = 42) and healthy controls (n = 39). RESULTS First-degree relatives and newly diagnosed type 1 diabetic patients displayed a similar frequency of IFN-gamma responses to the peptide panel and both were significantly greater than in healthy controls (relatives 9.6%, patients 11.8%, controls 4.0%, p = 0.003). First-degree relatives and newly diagnosed type 1 diabetic patients also showed similar frequencies of IL-10 responses, which were significantly lower than in healthy controls (relatives 7.1%, patients 9.0%, controls 15.8%, p = 0.003). However, individual IL-10 responses of first-degree relatives were similar in size to those in healthy controls and larger than those in newly diagnosed type 1 diabetic patients (relatives median 29 spot-forming cells/1 x 10(6) peripheral blood mononuclear cells, controls 33, patients 11, p = 0.02). CONCLUSIONS/INTERPRETATION Taken together, these results suggest that antibody-negative first-degree relatives have a balance of proinflammatory and regulatory T cells, which is intermediate between that of newly diagnosed type 1 diabetic patients and healthy controls. This suggests that even a moderate regulatory response may be sufficient to prevent the development of clinical type 1 diabetes in genetically predisposed individuals.
Collapse
Affiliation(s)
- L G Petrich de Marquesini
- Henry Wellcome Laboratory for Integrative Neuroscience and Endocrinology, Dorothy Hodgkin Building, Clinical Science at South Bristol, University of Bristol, Whitson St, Bristol, BS1 3NY, UK
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Cox SL, Silveira PA. Emerging roles for B lymphocytes in Type 1 diabetes. Expert Rev Clin Immunol 2010; 5:311-24. [PMID: 20477009 DOI: 10.1586/eci.09.4] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Self-reactive B lymphocytes play two main pathological roles in autoimmune diseases: as secretors of autoantibodies and as specialized antigen-presenting cells that present self-components to autoreactive T lymphocytes. In recognition of these roles, recent clinical trials have utilized B-lymphocyte-depleting monoclonal antibodies to treat various autoimmune diseases, with encouraging results in those where humoral autoimmunity is clearly important. Surprisingly, recent results in animal models suggest that B-lymphocyte depletion may also be effective in the treatment of T-lymphocyte-mediated autoimmune diseases, such as Type 1 diabetes (T1D). This article reviews the experimental evidence that has uncovered pathogenic as well as regulatory roles for B lymphocytes in the prodrome of T1D and how this information is being used to develop novel therapeutic strategies to treat the disease.
Collapse
Affiliation(s)
- S Lewis Cox
- Immunology Program, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, NSW 2010, Australia.
| | | |
Collapse
|
24
|
Luo X, Herold KC, Miller SD. Immunotherapy of type 1 diabetes: where are we and where should we be going? Immunity 2010; 32:488-99. [PMID: 20412759 PMCID: PMC2860878 DOI: 10.1016/j.immuni.2010.04.002] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2010] [Revised: 03/22/2010] [Accepted: 03/31/2010] [Indexed: 02/06/2023]
Abstract
Type 1 diabetes (T1D) is a chronic autoimmune disorder characterized by destruction of insulin-producing pancreatic beta cells. Many broad-based immunosuppressive and antigen-specific immunoregulatory therapies have been and are currently being evaluated for their utility in the prevention and treatment of T1D. Looking forward, this review discusses the potential therapeutic use of antigen-specific tolerance strategies, including tolerance induced by "tolerogenic" antigen-presenting cells pulsed with diabetogenic antigens and transfer of induced or expanded regulatory T cells, which have demonstrated efficacy in nonobese diabetic (NOD) mice. Depending on the time of therapeutic intervention in the T1D disease process, antigen-specific immunoregulatory strategies may be employed as monotherapies, or in combination with short-term tolerance-promoting immunoregulatory drugs and/or drugs promoting differentiation of insulin-producing beta cells from endogenous progenitors.
Collapse
Affiliation(s)
- Xunrong Luo
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Kevan C. Herold
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | - Stephen D. Miller
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| |
Collapse
|
25
|
Michels AW, Eisenbarth GS. Immunologic endocrine disorders. J Allergy Clin Immunol 2010; 125:S226-37. [PMID: 20176260 PMCID: PMC2835296 DOI: 10.1016/j.jaci.2009.09.053] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2009] [Revised: 09/17/2009] [Accepted: 09/18/2009] [Indexed: 01/12/2023]
Abstract
Autoimmunity affects multiple glands in the endocrine system. Animal models and human studies highlight the importance of alleles in HLA-like molecules determining tissue-specific targeting that, with the loss of tolerance, leads to organ-specific autoimmunity. Disorders such as type 1A diabetes, Graves disease, Hashimoto thyroiditis, Addison disease, and many others result from autoimmune-mediated tissue destruction. Each of these disorders can be divided into stages beginning with genetic susceptibility, environmental triggers, active autoimmunity, and finally metabolic derangements with overt symptoms of disease. With an increased understanding of the immunogenetics and immunopathogenesis of endocrine autoimmune disorders, immunotherapies are becoming prevalent, especially in patients with type 1A diabetes. Immunotherapies are being used more in multiple subspecialty fields to halt disease progression. Although therapies for autoimmune disorders stop the progress of an immune response, immunomodulatory therapies for cancer and chronic infections can also provoke an unwanted immune response. As a result, there are now iatrogenic autoimmune disorders arising from the treatment of chronic viral infections and malignancies.
Collapse
Affiliation(s)
- Aaron W Michels
- Department of Medicine, Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, CO 80045, USA.
| | | |
Collapse
|
26
|
Fischer B, Elias D, Bretzel RG, Linn T. Immunomodulation with heat shock protein DiaPep277 to preserve beta cell function in type 1 diabetes – an update. Expert Opin Biol Ther 2009; 10:265-72. [DOI: 10.1517/14712590903555176] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
27
|
Achenbach P, Lampasona V, Landherr U, Koczwara K, Krause S, Grallert H, Winkler C, Pflüger M, Illig T, Bonifacio E, Ziegler AG. Autoantibodies to zinc transporter 8 and SLC30A8 genotype stratify type 1 diabetes risk. Diabetologia 2009; 52:1881-8. [PMID: 19590848 DOI: 10.1007/s00125-009-1438-0] [Citation(s) in RCA: 150] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2009] [Accepted: 06/01/2009] [Indexed: 10/20/2022]
Abstract
AIMS/HYPOTHESIS Our aim was to determine the relationships between autoantibodies to zinc transporter 8 (ZnT8), genotypes of the ZnT8-encoding gene SLC30A8 and type 1 diabetes risk. METHODS ZnT8 autoantibodies (ZnT8A) were measured in sera of 1,633 children with a first-degree family history of type 1 diabetes and who were prospectively followed from birth. Antibodies were measured by Protein A-based radiobinding assays and COOH-terminal (R325, W325 or Q325 variants) or NH(2)-terminal constructs of human ZnT8. SLC30A8 genotyping at single-nucleotide polymorphism (SNP) rs13266634 was performed on 1,170 children. RESULTS Antibodies against COOH-terminal ZnT8 constructs (ZnT8A-COOH) developed in 58 children as early as 9 months of age (median 3 years). They were detected in 55 of 128 (43%) children with autoantibodies to insulin, GAD and/or insulinoma-associated protein 2 and 34 of 42 (81%) who progressed to diabetes. The additional presence of ZnT8A-COOH stratified diabetes risk in islet autoantibody-positive children (p < 0.0001). SLC30A8 genotype strongly influenced ZnT8A type and diabetes risk in ZnT8A-COOH-positive children. Antibody binding against the ZnT8 R325 variant was strictly correlated with the number of the corresponding SLC30A8 R325-encoding alleles, whereas binding against the W325 variant was highest in children who had SLC30A8 W325-encoding alleles (p = 0.001). Moreover, ZnT8A-COOH-positive children who carried homozygous SLC30A8 SNP rs13266634 genotypes progressed faster to diabetes than those who were heterozygous (59% [95% CI 42.3-75.7%] vs 22% [95% CI 0-44.3%] within 5 years; p = 0.01). CONCLUSIONS/INTERPRETATION Autoimmunity against the COOH-terminal region of ZnT8 is a highly relevant prognostic feature in childhood type 1 diabetes. Risk stratification in ZnT8A-COOH-positive children is further improved by SLC30A8 genotyping.
Collapse
Affiliation(s)
- P Achenbach
- Diabetes Research Institute of Forschergruppe Diabetes e.V., Helmholtz Center Munich, Ingolstaedter Landstr. 1, 85764, Neuherberg, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Abstract
Type 1 diabetes (T1D) is a T cell-mediated autoimmune disease in which the insulin producing beta cells are destroyed. Antigen-based immunotherapy provides an approach to selectively tolerize pathogenic beta cell-specific T cells, while leaving the remainder of the immune system intact. In this article, we discuss our group's experience in defining the parameters that impact the efficacy of beta cell antigen "vaccination" for the prevention and treatment of T1D.
Collapse
|
29
|
Wang B, Tisch R. Parameters influencing antigen-specific immunotherapy for type 1 diabetes. Immunol Res 2008; 41:175-87. [DOI: 10.1007/s12026-008-8020-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
30
|
Marín-García J, Goldenthal MJ, Moe GW. Metabolic Syndrome, Diabetes and Cardiometabolic Risks in Aging. AGING AND THE HEART 2007:277-305. [DOI: 10.1007/978-0-387-74072-0_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
31
|
Roncarolo MG, Battaglia M. Regulatory T-cell immunotherapy for tolerance to self antigens and alloantigens in humans. Nat Rev Immunol 2007; 7:585-98. [PMID: 17653126 DOI: 10.1038/nri2138] [Citation(s) in RCA: 399] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Substantial progress in understanding the biology of regulatory T cells and their roles in health and disease has been achieved in the past 10 years. This has led to an increasing interest in the possibility of using regulatory T cells as a biological therapy to preserve and restore tolerance to self antigens and alloantigens. Immunotherapy by the adoptive transfer of regulatory T cells may have several advantages over conventional treatments. However, several hurdles have to be overcome before such a therapy can enter clinical practice. This Review summarizes our current knowledge of regulatory T cells, illustrates the ongoing regulatory T-cell-based clinical trials, analyses the strengths and pitfalls of this new therapeutic approach, and highlights the future perspectives.
Collapse
Affiliation(s)
- Maria-Grazia Roncarolo
- San Raffaele Telethon Institute for Gene Therapy (HSR-TIGET), Via Olgettina-58, 20132 Milan, Italy.
| | | |
Collapse
|
32
|
Staeva-Vieira T, Peakman M, von Herrath M. Translational mini-review series on type 1 diabetes: Immune-based therapeutic approaches for type 1 diabetes. Clin Exp Immunol 2007; 148:17-31. [PMID: 17349010 PMCID: PMC1868847 DOI: 10.1111/j.1365-2249.2007.03328.x] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Type 1 diabetes (T1D) is often considered the prototype organ-specific autoimmune disease in clinical immunology circles. The key disease features - precise destruction of a single endocrine cell type occurring on a distinct genetic and autoimmune background - have been unravelled in recent years to such an extent that there is a growing expectation that the disease should be curable. T1D is something of an orphan disease, currently managed by endocrinologists yet dependent upon the wit of immunologists, both basic and clinical, to find the best approaches to prevention and cure. Type 1 diabetes thus represents one of the most active arenas for translational research, as novel immune-based interventions find their way to the clinic. The first serious attempt at immune-based treatment for T1D was in 1984, the first at prevention in 1993; current and planned trials will take us into the next decade before reporting their results. This paper represents the first attempt at a comprehensive review of this quarter century of endeavour, documenting all the strategies that have emerged into clinical studies. Importantly, the intense clinical activity has established robust infrastructures for future T1D trials and frameworks for their design. The evident success of the monoclonal anti-CD3 antibody trials in established T1D demonstrate that modulation of islet autoimmunity in humans after the onset of overt disease can be achieved, and give some reason to be cautiously optimistic for the ability of these and other agents, alone and in combination, to provide an effective immunotherapy for the disease.
Collapse
Affiliation(s)
- T Staeva-Vieira
- Research Department, Juvenile Diabetes Research Foundation International, New York, NY 10005-4001, USA.
| | | | | |
Collapse
|
33
|
Achenbach P, Warncke K, Reiter J, Williams AJK, Ziegler AG, Bingley PJ, Bonifacio E. Type 1 diabetes risk assessment: improvement by follow-up measurements in young islet autoantibody-positive relatives. Diabetologia 2006; 49:2969-76. [PMID: 17019596 DOI: 10.1007/s00125-006-0451-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2006] [Accepted: 07/31/2006] [Indexed: 02/01/2023]
Abstract
AIMS/HYPOTHESIS Combinations of autoantibody characteristics, including antibody number, titre, subclass and epitope have been shown to stratify type 1 diabetes risk in islet autoantibody-positive relatives. The aim of this study was to determine whether autoantibody characteristics change over time, the nature of such changes, and their implications for the development of diabetes. METHODS Five-hundred and thirteen follow-up samples from 141 islet autoantibody-positive first-degree relatives were tested for islet autoantibody titre, IgG subclass, and GAD and IA-2 antibody epitope. All samples were categorised according to four risk stratification models. Relatives had a median follow-up of 6.8 years and 48 developed diabetes during follow-up. Survival analysis was used to determine the probability of change in risk category and of progression to diabetes. RESULTS For each stratification model, the majority of relatives (71-81%) remained in the same risk category throughout follow-up. In the remainder, changes occurred both from lower to higher and from higher to lower risk categories. For all four models, relatives aged < 15 years were more likely to change risk category than those aged >15 years (0.001 < p < 0.03). Relatives whose autoantibody status changed from low- to high-risk categories had a higher risk of diabetes than relatives who remained in low-risk categories, and inclusion of autoantibody status during follow-up improved diabetes risk stratification in Cox proportional hazards models (p < 0.001). CONCLUSIONS/INTERPRETATION Changes in islet autoantibodies are relevant to pathogenesis, and are likely to signal alterations in the disease process. Detection of changes through follow-up measurement will improve diabetes risk stratification, particularly in young individuals.
Collapse
Affiliation(s)
- P Achenbach
- Diabetes Research Institute, Koelner Platz 1, Munich, Germany
| | | | | | | | | | | | | |
Collapse
|
34
|
Finucane KA, Archer CB. Recent advances in diabetology: diabetic dermopathy, autoantibodies in the prediction of the development of type 1 diabetes, and islet cell transplantation and inhaled insulin as treatment for diabetes. Clin Exp Dermatol 2006; 31:837-40. [PMID: 16907932 DOI: 10.1111/j.1365-2230.2006.02239.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- K A Finucane
- Bristol Dermatology Centre, Bristol Royal Infirmary, Bristol, UK.
| | | |
Collapse
|
35
|
Dantonio P, Meredith N, Earley M, Cordovado S, Callan WJ, Rollin D, Morris D, Vogt RF, Hannon WH. A screening system for detecting genetic risk markers of type 1 diabetes in dried blood spots. Diabetes Technol Ther 2006; 8:433-43. [PMID: 16939368 DOI: 10.1089/dia.2006.8.433] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND Certain alleles among the genes that code for the human leukocyte antigens (HLA) confer susceptibility or resistance to the development of autoimmunity that causes type 1 diabetes (T1D). A number of ongoing diabetes research studies analyze dried blood spots (DBS) from newborn infants for HLA-D alleles to identify higher-risk children as early as possible. A commercially available assay to detect such alleles has recently become available using a dissociation- enhanced lanthanide fluorescence system found in many newborn screening laboratories. METHODS We adapted the system for use with DBS and improved the sample set-up for greater efficiency. We also developed an independent system for data analysis based on a spreadsheet program. These modifications were applied to HLA-DQB1 gene locus (DQB) analysis of 117 newborn DBS, and the results we obtained were compared with independent reference values. RESULTS Our assay modifications and independent data analysis improved sample throughput and result tabulation. DQB results from the modified assay were consistent with the reference values in all but one sample, which showed a partial match. CONCLUSIONS The modifications described here make this commercially available assay more suitable for high-throughput applications such as newborn screening. Our results show that this system allows highly accurate detection of DQB alleles that influence T1D risk.
Collapse
Affiliation(s)
- Paul Dantonio
- Division of Laboratory Sciences, Centers for Disease Control and Prevention, Atlanta, Georgia 30341, USA
| | | | | | | | | | | | | | | | | |
Collapse
|