1
|
Ikegami H, Noso S. Genetics of type-1 diabetes. Diabetol Int 2024; 15:688-698. [PMID: 39469551 PMCID: PMC11512969 DOI: 10.1007/s13340-024-00754-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 08/06/2024] [Indexed: 10/30/2024]
Abstract
Type-1 diabetes is a multifactorial disease characterized by genetic and environmental factors that contribute to its development and progression. Despite progress in the management of type-1 diabetes, the final goal of curing the disease is yet to be achieved. To establish effective methods for the prevention, intervention, and cure of the disease, the molecular mechanisms and pathways involved in its development and progression should be clarified. One effective approach is to identify genes responsible for disease susceptibility and apply information obtained from the function of genes in disease etiology for the protection, intervention, and cure of type-1 diabetes. In this review, we discuss the genetic basis of type-1 diabetes, along with prospects for its prevention, intervention, and cure for type-1 diabetes.
Collapse
Affiliation(s)
- Hiroshi Ikegami
- Professor Emeritus, Kindai University, Osaka-sayama, Japan
- Director of Health Administration Center and Nikkei Clinic, Human Resources, Nikkei Inc. Osaka Head Office, Osaka, Japan
| | - Shinsuke Noso
- Department of Endocrinology, Metabolism and Diabetes, Faculty of Medicine, Kindai University, Osaka-sayama, Japan
| |
Collapse
|
2
|
Robertson CC, Elgamal RM, Henry-Kanarek BA, Arvan P, Chen S, Dhawan S, Eizirik DL, Kaddis JS, Vahedi G, Parker SCJ, Gaulton KJ, Soleimanpour SA. Untangling the genetics of beta cell dysfunction and death in type 1 diabetes. Mol Metab 2024; 86:101973. [PMID: 38914291 PMCID: PMC11283044 DOI: 10.1016/j.molmet.2024.101973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 06/18/2024] [Accepted: 06/19/2024] [Indexed: 06/26/2024] Open
Abstract
BACKGROUND Type 1 diabetes (T1D) is a complex multi-system disease which arises from both environmental and genetic factors, resulting in the destruction of insulin-producing pancreatic beta cells. Over the past two decades, human genetic studies have provided new insight into the etiology of T1D, including an appreciation for the role of beta cells in their own demise. SCOPE OF REVIEW Here, we outline models supported by human genetic data for the role of beta cell dysfunction and death in T1D. We highlight the importance of strong evidence linking T1D genetic associations to bona fide candidate genes for mechanistic and therapeutic consideration. To guide rigorous interpretation of genetic associations, we describe molecular profiling approaches, genomic resources, and disease models that may be used to construct variant-to-gene links and to investigate candidate genes and their role in T1D. MAJOR CONCLUSIONS We profile advances in understanding the genetic causes of beta cell dysfunction and death at individual T1D risk loci. We discuss how genetic risk prediction models can be used to address disease heterogeneity. Further, we present areas where investment will be critical for the future use of genetics to address open questions in the development of new treatment and prevention strategies for T1D.
Collapse
Affiliation(s)
- Catherine C Robertson
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA; Center for Precision Health Research, National Human Genome Research Institute, NIH, Bethesda, MD 20892, USA
| | - Ruth M Elgamal
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
| | - Belle A Henry-Kanarek
- Department of Internal Medicine and Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, USA
| | - Peter Arvan
- Department of Internal Medicine and Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, USA
| | - Shuibing Chen
- Department of Surgery, Weill Cornell Medicine, New York, NY, USA; Center for Genomic Health, Weill Cornell Medicine, New York, NY, USA
| | - Sangeeta Dhawan
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA, USA
| | - Decio L Eizirik
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
| | - John S Kaddis
- Department of Diabetes and Cancer Discovery Science, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Golnaz Vahedi
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Stephen C J Parker
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA; Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA; Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA.
| | - Kyle J Gaulton
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA.
| | - Scott A Soleimanpour
- Department of Internal Medicine and Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
3
|
Yilmazer A, Zevla DM, Malmkvist R, Rodríguez CAB, Undurraga P, Kirgin E, Boernert M, Voehringer D, Kershaw O, Schlenner S, Kretschmer K. Selective ablation of thymic and peripheral Foxp3 + regulatory T cell development. Front Immunol 2023; 14:1298938. [PMID: 38164128 PMCID: PMC10757929 DOI: 10.3389/fimmu.2023.1298938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 11/27/2023] [Indexed: 01/03/2024] Open
Abstract
Foxp3+ regulatory T (Treg) cells of thymic (tTreg) and peripheral (pTreg) developmental origin are thought to synergistically act to ensure immune homeostasis, with self-reactive tTreg cells primarily constraining autoimmune responses. Here we exploited a Foxp3-dependent reporter with thymus-specific GFP/Cre activity to selectively ablate either tTreg (ΔtTreg) or pTreg (ΔpTreg) cell development, while sparing the respective sister populations. We found that, in contrast to the tTreg cell behavior in ΔpTreg mice, pTreg cells acquired a highly activated suppressor phenotype and replenished the Treg cell pool of ΔtTreg mice on a non-autoimmune C57BL/6 background. Despite the absence of tTreg cells, pTreg cells prevented early mortality and fatal autoimmunity commonly observed in Foxp3-deficient models of complete Treg cell deficiency, and largely maintained immune tolerance even as the ΔtTreg mice aged. However, only two generations of backcrossing to the autoimmune-prone non-obese diabetic (NOD) background were sufficient to cause severe disease lethality associated with different, partially overlapping patterns of organ-specific autoimmunity. This included a particularly severe form of autoimmune diabetes characterized by an early onset and abrogation of the sex bias usually observed in the NOD mouse model of human type 1 diabetes. Genetic association studies further allowed us to define a small set of autoimmune risk loci sufficient to promote β cell autoimmunity, including genes known to impinge on Treg cell biology. Overall, these studies show an unexpectedly high functional adaptability of pTreg cells, emphasizing their important role as mediators of bystander effects to ensure self-tolerance.
Collapse
Affiliation(s)
- Acelya Yilmazer
- Molecular and Cellular Immunology/Immune Regulation, Center for Regenerative Therapies Dresden (CRTD), Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Dresden, Germany
| | - Dimitra Maria Zevla
- Molecular and Cellular Immunology/Immune Regulation, Center for Regenerative Therapies Dresden (CRTD), Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Dresden, Germany
| | - Rikke Malmkvist
- Molecular and Cellular Immunology/Immune Regulation, Center for Regenerative Therapies Dresden (CRTD), Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Dresden, Germany
| | - Carlos Alejandro Bello Rodríguez
- Molecular and Cellular Immunology/Immune Regulation, Center for Regenerative Therapies Dresden (CRTD), Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Dresden, Germany
| | - Pablo Undurraga
- Molecular and Cellular Immunology/Immune Regulation, Center for Regenerative Therapies Dresden (CRTD), Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Dresden, Germany
| | - Emre Kirgin
- Molecular and Cellular Immunology/Immune Regulation, Center for Regenerative Therapies Dresden (CRTD), Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Dresden, Germany
| | - Marie Boernert
- Molecular and Cellular Immunology/Immune Regulation, Center for Regenerative Therapies Dresden (CRTD), Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Dresden, Germany
| | - David Voehringer
- Department of Infection Biology, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Olivia Kershaw
- Department of Veterinary Medicine, Institute of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Susan Schlenner
- KU Leuven-University of Leuven, Department of Microbiology, Immunology and Transplantation, Leuven, Belgium
| | - Karsten Kretschmer
- Molecular and Cellular Immunology/Immune Regulation, Center for Regenerative Therapies Dresden (CRTD), Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden (PLID) of the Helmholtz Center Munich, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| |
Collapse
|
4
|
James EA, Joglekar AV, Linnemann AK, Russ HA, Kent SC. The beta cell-immune cell interface in type 1 diabetes (T1D). Mol Metab 2023; 78:101809. [PMID: 37734713 PMCID: PMC10622886 DOI: 10.1016/j.molmet.2023.101809] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 09/01/2023] [Accepted: 09/15/2023] [Indexed: 09/23/2023] Open
Abstract
BACKGROUND T1D is an autoimmune disease in which pancreatic islets of Langerhans are infiltrated by immune cells resulting in the specific destruction of insulin-producing islet beta cells. Our understanding of the factors leading to islet infiltration and the interplay of the immune cells with target beta cells is incomplete, especially in human disease. While murine models of T1D have provided crucial information for both beta cell and autoimmune cell function, the translation of successful therapies in the murine model to human disease has been a challenge. SCOPE OF REVIEW Here, we discuss current state of the art and consider knowledge gaps concerning the interface of the islet beta cell with immune infiltrates, with a focus on T cells. We discuss pancreatic and immune cell phenotypes and their impact on cell function in health and disease, which we deem important to investigate further to attain a more comprehensive understanding of human T1D disease etiology. MAJOR CONCLUSIONS The last years have seen accelerated development of approaches that allow comprehensive study of human T1D. Critically, recent studies have contributed to our revised understanding that the pancreatic beta cell assumes an active role, rather than a passive position, during autoimmune disease progression. The T cell-beta cell interface is a critical axis that dictates beta cell fate and shapes autoimmune responses. This includes the state of the beta cell after processing internal and external cues (e.g., stress, inflammation, genetic risk) that that contributes to the breaking of tolerance by hyperexpression of human leukocyte antigen (HLA) class I with presentation of native and neoepitopes and secretion of chemotactic factors to attract immune cells. We anticipate that emerging insights about the molecular and cellular aspects of disease initiation and progression processes will catalyze the development of novel and innovative intervention points to provide additional therapies to individuals affected by T1D.
Collapse
Affiliation(s)
- Eddie A James
- Center for Translational Immunology, Benaroya Research Institute, Seattle, WA, USA
| | - Alok V Joglekar
- Center for Systems Immunology and Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Amelia K Linnemann
- Center for Diabetes and Metabolic Diseases, and Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Holger A Russ
- Diabetes Institute, University of Florida, Gainesville, FL, USA; Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, USA
| | - Sally C Kent
- Diabetes Center of Excellence, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
5
|
Yagihashi S. Contribution of animal models to diabetes research: Its history, significance, and translation to humans. J Diabetes Investig 2023; 14:1015-1037. [PMID: 37401013 PMCID: PMC10445217 DOI: 10.1111/jdi.14034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/10/2023] [Accepted: 05/16/2023] [Indexed: 07/05/2023] Open
Abstract
Diabetes mellitus is still expanding globally and is epidemic in developing countries. The combat of this plague has caused enormous economic and social burdens related to a lowered quality of life in people with diabetes. Despite recent significant improvements of life expectancy in patients with diabetes, there is still a need for efforts to elucidate the complexities and mechanisms of the disease processes to overcome this difficult disorder. To this end, the use of appropriate animal models in diabetes studies is invaluable for translation to humans and for the development of effective treatment. In this review, a variety of animal models of diabetes with spontaneous onset in particular will be introduced and discussed for their implication in diabetes research.
Collapse
Affiliation(s)
- Soroku Yagihashi
- Department of Exploratory Medicine for Nature, Life and HumansToho University School of MedicineChibaJapan
- Department of PathologyHirosaki University Graduate School of MedicineHirosakiJapan
| |
Collapse
|
6
|
Stadinski BD, Cleveland SB, Brehm MA, Greiner DL, Huseby PG, Huseby ES. I-A g7 β56/57 polymorphisms regulate non-cognate negative selection to CD4 + T cell orchestrators of type 1 diabetes. Nat Immunol 2023; 24:652-663. [PMID: 36807641 PMCID: PMC10623581 DOI: 10.1038/s41590-023-01441-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 01/20/2023] [Indexed: 02/22/2023]
Abstract
Genetic susceptibility to type 1 diabetes is associated with homozygous expression of major histocompatibility complex class II alleles that carry specific beta chain polymorphisms. Why heterozygous expression of these major histocompatibility complex class II alleles does not confer a similar predisposition is unresolved. Using a nonobese diabetic mouse model, here we show that heterozygous expression of the type 1 diabetes-protective allele I-Ag7 β56P/57D induces negative selection to the I-Ag7-restricted T cell repertoire, including beta-islet-specific CD4+ T cells. Surprisingly, negative selection occurs despite I-Ag7 β56P/57D having a reduced ability to present beta-islet antigens to CD4+ T cells. Peripheral manifestations of non-cognate negative selection include a near complete loss of beta-islet-specific CXCR6+ CD4+ T cells, an inability to cross-prime islet-specific glucose-6-phosphatase catalytic subunit-related protein and insulin-specific CD8+ T cells and disease arrest at the insulitis stage. These data reveal that negative selection on non-cognate self-antigens in the thymus can promote T cell tolerance and protection from autoimmunity.
Collapse
Affiliation(s)
- Brian D Stadinski
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Sarah B Cleveland
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Michael A Brehm
- Department of Molecular Medicine, Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, MA, USA
| | - Dale L Greiner
- Department of Molecular Medicine, Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, MA, USA
| | - Priya G Huseby
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Eric S Huseby
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA, USA.
| |
Collapse
|
7
|
Chatenoud L, Marquet C, Valette F, Scott L, Quan J, Bu CH, Hildebrand S, Moresco EMY, Bach JF, Beutler B. Modulation of autoimmune diabetes by N-ethyl-N-nitrosourea- induced mutations in non-obese diabetic mice. Dis Model Mech 2022; 15:275575. [PMID: 35502705 PMCID: PMC9178510 DOI: 10.1242/dmm.049484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 04/21/2022] [Indexed: 11/20/2022] Open
Abstract
Genetic association studies of type 1 diabetes (T1D) in humans, and in congenic non-obese diabetic (NOD) mice harboring DNA segments from T1D-resistant mice, face the challenge of assigning causation to specific gene variants among many within loci that affect disease risk. Here, we created random germline mutations in NOD/NckH mice and used automated meiotic mapping to identify mutations modifying T1D incidence and age of onset. In contrast with association studies in humans or congenic NOD mice, we analyzed a relatively small number of genetic changes in each pedigree, permitting implication of specific mutations as causative. Among 844 mice from 14 pedigrees bearing 594 coding/splicing changes, we identified seven mutations that accelerated T1D development, and five that delayed or suppressed T1D. Eleven mutations affected genes not previously known to influence T1D (Xpnpep1, Herc1, Srrm2, Rapgef1, Ppl, Zfp583, Aldh1l1, Col6a1, Ccdc13, Cd200r1, Atrnl1). A suppressor mutation in Coro1a validated the screen. Mutagenesis coupled with automated meiotic mapping can detect genes in which allelic variation influences T1D susceptibility in NOD mice. Variation of some of the orthologous/paralogous genes may influence T1D susceptibility in humans.
Collapse
Affiliation(s)
- Lucienne Chatenoud
- Université Paris Cité, Institut Necker Enfants Malades, F-75015 Paris, France.,INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, F-75015 Paris, France
| | - Cindy Marquet
- Université Paris Cité, Institut Necker Enfants Malades, F-75015 Paris, France.,INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, F-75015 Paris, France
| | - Fabrice Valette
- Université Paris Cité, Institut Necker Enfants Malades, F-75015 Paris, France.,INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, F-75015 Paris, France
| | - Lindsay Scott
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jiexia Quan
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Chun Hui Bu
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Sara Hildebrand
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Eva Marie Y Moresco
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jean-François Bach
- Université Paris Cité, Institut Necker Enfants Malades, F-75015 Paris, France.,INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, F-75015 Paris, France
| | - Bruce Beutler
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
8
|
Lombard-Vadnais F, Collin R, Daudelin JF, Chabot-Roy G, Labrecque N, Lesage S. The Idd2 Locus Confers Prominent Resistance to Autoimmune Diabetes. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:898-909. [PMID: 35039332 DOI: 10.4049/jimmunol.2100456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 12/02/2021] [Indexed: 06/14/2023]
Abstract
Type 1 diabetes is an autoimmune disease characterized by pancreatic β cell destruction. It is a complex genetic trait driven by >30 genetic loci with parallels between humans and mice. The NOD mouse spontaneously develops autoimmune diabetes and is widely used to identify insulin-dependent diabetes (Idd) genetic loci linked to diabetes susceptibility. Although many Idd loci have been extensively studied, the impact of the Idd2 locus on autoimmune diabetes susceptibility remains to be defined. To address this, we generated a NOD congenic mouse bearing B10 resistance alleles on chromosome 9 in a locus coinciding with part of the Idd2 locus and found that NOD.B10-Idd2 congenic mice are highly resistant to diabetes. Bone marrow chimera and adoptive transfer experiments showed that the B10 protective alleles provide resistance in an immune cell-intrinsic manner. Although no T cell-intrinsic differences between NOD and NOD.B10-Idd2 mice were observed, we found that the Idd2 resistance alleles limit the formation of spontaneous and induced germinal centers. Comparison of B cell and dendritic cell transcriptome profiles from NOD and NOD.B10-Idd2 mice reveal that resistance alleles at the Idd2 locus affect the expression of specific MHC molecules, a result confirmed by flow cytometry. Altogether, these data demonstrate that resistance alleles at the Idd2 locus impair germinal center formation and influence MHC expression, both of which likely contribute to reduced diabetes incidence.
Collapse
Affiliation(s)
- Félix Lombard-Vadnais
- Immunology-Oncology Axis, Research Center, Maisonneuve-Rosemont Hospital, Montreal, Quebec, Canada
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada
| | - Roxanne Collin
- Immunology-Oncology Axis, Research Center, Maisonneuve-Rosemont Hospital, Montreal, Quebec, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, Quebec, Canada; and
| | - Jean-François Daudelin
- Immunology-Oncology Axis, Research Center, Maisonneuve-Rosemont Hospital, Montreal, Quebec, Canada
| | - Geneviève Chabot-Roy
- Immunology-Oncology Axis, Research Center, Maisonneuve-Rosemont Hospital, Montreal, Quebec, Canada
| | - Nathalie Labrecque
- Immunology-Oncology Axis, Research Center, Maisonneuve-Rosemont Hospital, Montreal, Quebec, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, Quebec, Canada; and
- Département de Médecine, Université de Montréal, Montreal, Quebec, Canada
| | - Sylvie Lesage
- Immunology-Oncology Axis, Research Center, Maisonneuve-Rosemont Hospital, Montreal, Quebec, Canada;
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, Quebec, Canada; and
| |
Collapse
|
9
|
Schleier Y, Moreno-Loaiza O, López Alarcón MM, Lopes Martins EG, Braga BC, Ramos IP, Galina A, Medei EH. NOD Mice Recapitulate the Cardiac Disturbances Observed in Type 1 Diabetes. J Cardiovasc Transl Res 2021; 14:271-282. [PMID: 32468298 DOI: 10.1007/s12265-020-10039-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 05/20/2020] [Indexed: 10/24/2022]
Abstract
This work aimed at testing the hypothesis that NOD/ShiLtJ mice (NOD) recapitulate the cardiac disturbances observed on type 1 diabetes (T1D). NOD mice were studied 4 weeks after the onset of hyperglycemia, and NOR/Lt mice matched as control. Cardiac function was evaluated by echocardiography and electrocardiography (ECG). Action potentials (AP) and Ca2+ transients were evaluated at whole heart level. Heart mitochondrial function was evaluated by high-resolution respirometry and H2O2 release. NOD mice presented a reduction in hearth weight. Mitochondrial oxygen fluxes and H2O2 release were similar between NOD and NOR mice. ECG revealed a QJ interval prolongation in NOD mice. Furthermore, AP duration at 30% of repolarization was increased, and it depicted slower Ca2+ transient kinetics. NOD mice presented greater number/severity of ventricular arrhythmias both in vivo and in vitro. In conclusion, NOD mice evoked cardiac electrical and calcium handling disturbances similar to the observed in T1D. Graphical Abstract .
Collapse
Affiliation(s)
- Ygor Schleier
- Laboratory of Cardioimmunology, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Oscar Moreno-Loaiza
- Laboratory of Cardioimmunology, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Maria Micaela López Alarcón
- Laboratory of Cardioimmunology, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Eduarda Gabrielle Lopes Martins
- Laboratory of Cardioimmunology, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Laboratory of Bioenergetics and Mitochondrial Physiology, Institute of Medical Biochemistry Leopoldo de Meis, Center for Health Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Bruno Cabral Braga
- Laboratory of Cardioimmunology, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Isalira Peroba Ramos
- National Center for Structural Biology and Bioimaging, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Antonio Galina
- Laboratory of Bioenergetics and Mitochondrial Physiology, Institute of Medical Biochemistry Leopoldo de Meis, Center for Health Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Emiliano Horacio Medei
- Laboratory of Cardioimmunology, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
- National Center for Structural Biology and Bioimaging, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
- Carlos Chagas Filho Biophysics Institute - UFRJ, Avenida Carlos Chagas Filho, 373-CCS-Bloco G, Rio de Janeiro, RJ, 21941-902, Brazil.
| |
Collapse
|
10
|
Kim H, Perovanovic J, Shakya A, Shen Z, German CN, Ibarra A, Jafek JL, Lin NP, Evavold BD, Chou DHC, Jensen PE, He X, Tantin D. Targeting transcriptional coregulator OCA-B/Pou2af1 blocks activated autoreactive T cells in the pancreas and type 1 diabetes. J Exp Med 2021; 218:e20200533. [PMID: 33295943 PMCID: PMC7731945 DOI: 10.1084/jem.20200533] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 04/27/2020] [Accepted: 10/09/2020] [Indexed: 11/04/2022] Open
Abstract
The transcriptional coregulator OCA-B promotes expression of T cell target genes in cases of repeated antigen exposure, a necessary feature of autoimmunity. We hypothesized that T cell-specific OCA-B deletion and pharmacologic OCA-B inhibition would protect mice from autoimmune diabetes. We developed an Ocab conditional allele and backcrossed it onto a diabetes-prone NOD/ShiLtJ strain background. T cell-specific OCA-B loss protected mice from spontaneous disease. Protection was associated with large reductions in islet CD8+ T cell receptor specificities associated with diabetes pathogenesis. CD4+ clones associated with diabetes were present but associated with anergic phenotypes. The protective effect of OCA-B loss was recapitulated using autoantigen-specific NY8.3 mice but diminished in monoclonal models specific to artificial or neoantigens. Rationally designed membrane-penetrating OCA-B peptide inhibitors normalized glucose levels and reduced T cell infiltration and proinflammatory cytokine expression in newly diabetic NOD mice. Together, the results indicate that OCA-B is a potent autoimmune regulator and a promising target for pharmacologic inhibition.
Collapse
MESH Headings
- Alleles
- Amino Acid Sequence
- Animals
- Autoantigens/immunology
- CD4-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/immunology
- Crosses, Genetic
- Cytokines/metabolism
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/prevention & control
- Disease Models, Animal
- Female
- Gene Deletion
- Germ Cells/metabolism
- Humans
- Inflammation Mediators/metabolism
- Lymph Nodes/metabolism
- Lymphocyte Activation
- Male
- Mice, Inbred C57BL
- Mice, Inbred NOD
- Ovalbumin
- Pancreas/metabolism
- Pancreas/pathology
- Peptides/pharmacology
- Receptors, Antigen, T-Cell/metabolism
- Spleen/pathology
- T-Lymphocytes/immunology
- Trans-Activators/deficiency
- Trans-Activators/metabolism
- Transcription, Genetic
- Mice
Collapse
Affiliation(s)
- Heejoo Kim
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT
| | - Jelena Perovanovic
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT
| | - Arvind Shakya
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT
| | - Zuolian Shen
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT
| | - Cody N German
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT
| | - Andrea Ibarra
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT
| | - Jillian L Jafek
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT
| | - Nai-Pin Lin
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT
| | - Brian D Evavold
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT
| | - Danny H-C Chou
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT
| | - Peter E Jensen
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT
| | - Xiao He
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT
| | - Dean Tantin
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT
| |
Collapse
|
11
|
Ramirez DG, Abenojar E, Hernandez C, Lorberbaum DS, Papazian LA, Passman S, Pham V, Exner AA, Benninger RKP. Contrast-enhanced ultrasound with sub-micron sized contrast agents detects insulitis in mouse models of type1 diabetes. Nat Commun 2020; 11:2238. [PMID: 32382089 PMCID: PMC7206014 DOI: 10.1038/s41467-020-15957-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 04/06/2020] [Indexed: 12/12/2022] Open
Abstract
In type1 diabetes (T1D) autoreactive T-cells infiltrate the islets of Langerhans, depleting insulin-secreting β-cells (insulitis). Insulitis arises during an asymptomatic phase, prior to clinical diagnosis of T1D. Methods to diagnose insulitis and β-cell mass changes during this asymptomatic phase are limited, precluding early therapeutic intervention. During T1D the islet microvasculature increases permeability, allowing nanoparticles to access the microenvironment. Contrast enhanced ultrasound (CEUS) uses shell-stabilized gas bubbles to provide acoustic backscatter in vasculature. Here, we report that sub-micron sized 'nanobubble' ultrasound contrast agents can be used to measure increased islet microvasculature permeability and indicate asymptomatic T1D. Through CEUS and histological analysis, pre-clinical models of T1D show accumulation of nanobubbles specifically within pancreatic islets, correlating with insulitis. Importantly, accumulation is detected early in disease progression and decreases with successful therapeutic intervention. Thus, sub-micron sized nanobubble ultrasound contrast agents provide a predicative marker for disease progression and therapeutic reversal early in asymptomatic T1D.
Collapse
Affiliation(s)
- David G Ramirez
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Eric Abenojar
- Department of Radiology, Case Western Reserve University, Cleveland, OH, USA
| | - Christopher Hernandez
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - David S Lorberbaum
- Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Lucine A Papazian
- Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Samantha Passman
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Vinh Pham
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Agata A Exner
- Department of Radiology, Case Western Reserve University, Cleveland, OH, USA.
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA.
| | - Richard K P Benninger
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
- Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
12
|
The MHC-II peptidome of pancreatic islets identifies key features of autoimmune peptides. Nat Immunol 2020; 21:455-463. [PMID: 32152506 PMCID: PMC7117798 DOI: 10.1038/s41590-020-0623-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 02/03/2020] [Indexed: 12/11/2022]
Abstract
The nature of autoantigens that trigger autoimmune diseases has been much discussed, but direct biochemical identification is lacking for most. Addressing this question demands unbiased examination of the self-peptides displayed by a defined autoimmune major histocompatibility complex class II (MHCII) molecule. Here we examined the immunopeptidome of the pancreatic islets in non-obese diabetic (NOD) mice, which spontaneously develop autoimmune diabetes based on the I-Ag7 variant of MHCII. The relevant peptides that induced pathogenic CD4+ T cells at the initiation of diabetes derived from proinsulin. These peptides were also found in the MHCII peptidome of the pancreatic lymph nodes and spleen. The proinsulin-derived peptides followed a trajectory from their generation and exocytosis in β cells, to uptake and presentation in islets and peripheral sites. Such a pathway generated conventional epitopes but also resulted in the presentation of post-translationally modified peptides, including deamidated sequences. These analyses reveal the key features of a restricted component in the self-MHCII peptidome that caused autoreactivity.
Collapse
|
13
|
Radaelli E, Santagostino SF, Sellers RS, Brayton CF. Immune Relevant and Immune Deficient Mice: Options and Opportunities in Translational Research. ILAR J 2019; 59:211-246. [PMID: 31197363 PMCID: PMC7114723 DOI: 10.1093/ilar/ily026] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 12/03/2018] [Indexed: 12/29/2022] Open
Abstract
In 1989 ILAR published a list and description of immunodeficient rodents used in research. Since then, advances in understanding of molecular mechanisms; recognition of genetic, epigenetic microbial, and other influences on immunity; and capabilities in manipulating genomes and microbiomes have increased options and opportunities for selecting mice and designing studies to answer important mechanistic and therapeutic questions. Despite numerous scientific breakthroughs that have benefitted from research in mice, there is debate about the relevance and predictive or translational value of research in mice. Reproducibility of results obtained from mice and other research models also is a well-publicized concern. This review summarizes resources to inform the selection and use of immune relevant mouse strains and stocks, aiming to improve the utility, validity, and reproducibility of research in mice. Immune sufficient genetic variations, immune relevant spontaneous mutations, immunodeficient and autoimmune phenotypes, and selected induced conditions are emphasized.
Collapse
Affiliation(s)
- Enrico Radaelli
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sara F Santagostino
- Department of Safety Assessment, Genentech, Inc., South San Francisco, California
| | | | - Cory F Brayton
- Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
14
|
Brehm MA, Kenney LL, Wiles MV, Low BE, Tisch RM, Burzenski L, Mueller C, Greiner DL, Shultz LD. Lack of acute xenogeneic graft- versus-host disease, but retention of T-cell function following engraftment of human peripheral blood mononuclear cells in NSG mice deficient in MHC class I and II expression. FASEB J 2019; 33:3137-3151. [PMID: 30383447 PMCID: PMC6404556 DOI: 10.1096/fj.201800636r] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 10/01/2018] [Indexed: 12/19/2022]
Abstract
Immunodeficient mice engrafted with human peripheral blood mononuclear cells (PBMCs) support preclinical studies of human pathogens, allograft rejection, and human T-cell function. However, a major limitation of PBMC engraftment is development of acute xenogeneic graft- versus-host disease (GVHD) due to human T-cell recognition of murine major histocompatibility complex (MHC). To address this, we created 2 NOD- scid IL-2 receptor subunit γ ( IL2rg) null (NSG) strains that lack murine MHC class I and II [NSG-β-2-microglobulin ( B2M) null ( IA IE)null and NSG -( Kb Db) null ( IAnull)]. We observed rapid human IgG clearance in NSG- B2Mnull ( IA IE) null mice whereas clearance in NSG -( Kb Db) null ( IAnull) mice and NSG mice was comparable. Injection of human PBMCs into both strains enabled long-term engraftment of human CD4+ and CD8+ T cells without acute GVHD. Engrafted human T-cell function was documented by rejection of human islet allografts. Administration of human IL-2 to NSG -( Kb Db) null ( IAnull) mice via adeno-associated virus vector increased human CD45+ cell engraftment, including an increase in human regulatory T cells. However, high IL-2 levels also induced the development of GVHD. These data document that NSG mice deficient in murine MHC support studies of human immunity in the absence of acute GVHD and enable evaluation of human antibody therapeutics targeting human T cells.-Brehm, M. A., Kenney, L. L., Wiles, M. V., Low, B. E., Tisch, R. M., Burzenski, L., Mueller, C., Greiner, D. L., Shultz, L. D. Lack of acute xenogeneic graft- versus-host disease, but retention of T-cell function following engraftment of human peripheral blood mononuclear cells in NSG mice deficient in MHC class I and II expression.
Collapse
Affiliation(s)
- Michael A. Brehm
- Diabetes Center of Excellence University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Laurie L. Kenney
- Diabetes Center of Excellence University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | | | | | - Roland M. Tisch
- Department of Immunology and Microbiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; and
| | | | - Christian Mueller
- Department of Pediatrics and Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Dale L. Greiner
- Diabetes Center of Excellence University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | | |
Collapse
|
15
|
Wan X, Unanue ER. Antigen recognition in autoimmune diabetes: a novel pathway underlying disease initiation. PRECISION CLINICAL MEDICINE 2018; 1:102-110. [PMID: 30687564 PMCID: PMC6333048 DOI: 10.1093/pcmedi/pby015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 10/13/2018] [Accepted: 10/26/2018] [Indexed: 12/21/2022] Open
Abstract
Development of human autoimmune disorders results from complex interplay among genetic, environmental, and immunological risk factors. Despite much heterogeneity in environmental triggers, the leading genes that give the propensity for tissue-specific autoimmune diseases, such as type 1 diabetes, are those associated with particular class II major histocompatibility complex alleles. Such genetic predisposition precipitates presentation of tissue antigens to MHC-II-restricted CD4 T cells. When properly activated, these self-reactive CD4 T cells migrate to the target tissue and trigger the initial immune attack. Using the non-obese diabetic mouse model of spontaneous autoimmune diabetes, much insight has been gained in understanding how presentation of physiological levels of self-antigens translates into pathological outcomes. In this review, we summarize recent advances illustrating the features of the antigen presenting cells, the sites of the antigen recognition, and the nature of the consequent T cell responses. We emphasize emerging evidence that highlights the importance of systemic presentation of catabolized tissue antigens in mobilization of pathogenic T cells. The implication of these studies in therapeutic perspectives is also discussed.
Collapse
Affiliation(s)
- Xiaoxiao Wan
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Emil R Unanue
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
16
|
Contrast-enhanced ultrasound measurement of pancreatic blood flow dynamics predicts type 1 diabetes progression in preclinical models. Nat Commun 2018; 9:1742. [PMID: 29717116 PMCID: PMC5931596 DOI: 10.1038/s41467-018-03953-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Accepted: 03/23/2018] [Indexed: 12/13/2022] Open
Abstract
In type 1 diabetes (T1D), immune-cell infiltration into the islets of Langerhans (insulitis) and β-cell decline occurs many years before diabetes clinically presents. Non-invasively detecting insulitis and β-cell decline would allow the diagnosis of eventual diabetes, and provide a means to monitor therapeutic intervention. However, there is a lack of validated clinical approaches for specifically and non-invasively imaging disease progression leading to T1D. Islets have a denser microvasculature that reorganizes during diabetes. Here we apply contrast-enhanced ultrasound measurements of pancreatic blood-flow dynamics to non-invasively and predictively assess disease progression in T1D pre-clinical models. STZ-treated mice, NOD mice, and adoptive-transfer mice demonstrate altered islet blood-flow dynamics prior to diabetes onset, consistent with islet microvasculature reorganization. These assessments predict both time to diabetes onset and future responders to antiCD4-mediated disease prevention. Thus contrast-enhanced ultrasound measurements of pancreas blood-flow dynamics may provide a clinically deployable predictive marker for disease progression in pre-symptomatic T1D and therapeutic reversal.
Collapse
|
17
|
MHC-mismatched mixed chimerism restores peripheral tolerance of noncross-reactive autoreactive T cells in NOD mice. Proc Natl Acad Sci U S A 2018; 115:E2329-E2337. [PMID: 29463744 PMCID: PMC5877958 DOI: 10.1073/pnas.1720169115] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Mixed chimerism has shown good potential to cure some autoimmune diseases and prevent tissue rejection. It is known that MHC-mismatched but not -matched mixed chimerism effectively tolerizes autoreactive T cells, even those noncross-reactive T cells that do not directly recognize donor-type antigen presenting cells [i.e., dendritic cells (DCs)]. How this is accomplished remains unknown. These studies have shown that tolerizing peripheral residual host-type noncross-reactive autoreactive T cells requires engraftment of donor-type DCs and involves a host-type DC-mediated increase in donor-type Treg cells, which associates with restoration of tolerogenic features of host-type plasmacytoid DCs and expansion of host-type Treg cells. This study suggests a previously unrecognized tolerance network among donor- and host-type DCs and Treg cells in MHC-mismatched mixed chimeras. Autoimmune type 1 diabetes (T1D) and other autoimmune diseases are associated with particular MHC haplotypes and expansion of autoreactive T cells. Induction of MHC-mismatched but not -matched mixed chimerism by hematopoietic cell transplantation effectively reverses autoimmunity in diabetic nonobese diabetic (NOD) mice, even those with established diabetes. As expected, MHC-mismatched mixed chimerism mediates deletion in the thymus of host-type autoreactive T cells that have T-cell receptor (TCR) recognizing (cross-reacting with) donor-type antigen presenting cells (APCs), which have come to reside in the thymus. However, how MHC-mismatched mixed chimerism tolerizes host autoreactive T cells that recognize only self-MHC–peptide complexes remains unknown. Here, using NOD.Rag1−/−.BDC2.5 or NOD.Rag1−/−.BDC12-4.1 mice that have only noncross-reactive transgenic autoreactive T cells, we show that induction of MHC-mismatched but not -matched mixed chimerism restores immune tolerance of peripheral noncross-reactive autoreactive T cells. MHC-mismatched mixed chimerism results in increased percentages of both donor- and host-type Foxp3+ Treg cells and up-regulated expression of programmed death-ligand 1 (PD-L1) by host-type plasmacytoid dendritic cells (pDCs). Furthermore, adoptive transfer experiments showed that engraftment of donor-type dendritic cells (DCs) and expansion of donor-type Treg cells are required for tolerizing the noncross-reactive autoreactive T cells in the periphery, which are in association with up-regulation of host-type DC expression of PD-L1 and increased percentage of host-type Treg cells. Thus, induction of MHC-mismatched mixed chimerism may establish a peripheral tolerogenic DC and Treg network that actively tolerizes autoreactive T cells, even those with no TCR recognition of the donor APCs.
Collapse
|
18
|
Barrington WT, Wulfridge P, Wells AE, Rojas CM, Howe SYF, Perry A, Hua K, Pellizzon MA, Hansen KD, Voy BH, Bennett BJ, Pomp D, Feinberg AP, Threadgill DW. Improving Metabolic Health Through Precision Dietetics in Mice. Genetics 2018; 208:399-417. [PMID: 29158425 PMCID: PMC5753872 DOI: 10.1534/genetics.117.300536] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 11/10/2017] [Indexed: 12/30/2022] Open
Abstract
The incidence of diet-induced metabolic disease has soared over the last half-century, despite national efforts to improve health through universal dietary recommendations. Studies comparing dietary patterns of populations with health outcomes have historically provided the basis for healthy diet recommendations. However, evidence that population-level diet responses are reliable indicators of responses across individuals is lacking. This study investigated how genetic differences influence health responses to several popular diets in mice, which are similar to humans in genetic composition and the propensity to develop metabolic disease, but enable precise genetic and environmental control. We designed four human-comparable mouse diets that are representative of those eaten by historical human populations. Across four genetically distinct inbred mouse strains, we compared the American diet's impact on metabolic health to three alternative diets (Mediterranean, Japanese, and Maasai/ketogenic). Furthermore, we investigated metabolomic and epigenetic alterations associated with diet response. Health effects of the diets were highly dependent on genetic background, demonstrating that individualized diet strategies improve health outcomes in mice. If similar genetic-dependent diet responses exist in humans, then a personalized, or "precision dietetics," approach to dietary recommendations may yield better health outcomes than the traditional one-size-fits-all approach.
Collapse
Affiliation(s)
- William T Barrington
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, Texas A&M University, College Station, Texas 77843
- Department of Biological Sciences, Genetics Program, North Carolina State University, Raleigh, North Carolina 27695
| | - Phillip Wulfridge
- Center for Epigenetics, Institute for Basic Biomedical Sciences, Johns Hopkins University, Baltimore, Maryland 21205
| | - Ann E Wells
- UT-ORNL Graduate School of Genome Science and Technology, Department of Animal Science, University of Tennessee, Knoxville, Tennessee 37996
| | - Carolina Mantilla Rojas
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, Texas A&M University, College Station, Texas 77843
| | - Selene Y F Howe
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, Texas A&M University, College Station, Texas 77843
| | - Amie Perry
- Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas 77843
| | - Kunjie Hua
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina 27559
| | | | - Kasper D Hansen
- Center for Epigenetics, Institute for Basic Biomedical Sciences, Johns Hopkins University, Baltimore, Maryland 21205
- Department of Biostatistics, Johns Hopkins University, Baltimore, Maryland 21205
- Nathan-McKusick Institute of Genetic Medicine, Johns Hopkins University, Baltimore, Maryland 21205
| | - Brynn H Voy
- UT-ORNL Graduate School of Genome Science and Technology, Department of Animal Science, University of Tennessee, Knoxville, Tennessee 37996
| | - Brian J Bennett
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina 27559
| | - Daniel Pomp
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina 27559
| | - Andrew P Feinberg
- Center for Epigenetics, Institute for Basic Biomedical Sciences, Johns Hopkins University, Baltimore, Maryland 21205
| | - David W Threadgill
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, Texas A&M University, College Station, Texas 77843
- Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas 77843
- Faculty of Nutrition, Texas A&M University, College Station, Texas 77843
- Faculty of Genetics, Texas A&M University, College Station, Texas 77843
- Faculty of Toxicology, Texas A&M University, College Station, Texas 77843
| |
Collapse
|
19
|
Mannering SI, So M, Elso CM, Kay TWH. Shuffling peptides to create T-cell epitopes: does the immune system play cards? Immunol Cell Biol 2017; 96:34-40. [PMID: 29359347 DOI: 10.1111/imcb.1015] [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: 08/15/2017] [Revised: 09/17/2017] [Accepted: 09/18/2017] [Indexed: 11/27/2022]
Abstract
For a long time, immunologists have believed that classical CD4+ and CD8+ T cells recognize peptides (referred to as epitopes), derived from protein antigens presented by MHC/HLA class I or II. Over the past 10-15 years, it has become clear that epitopes recognized by CD8+, and more recently CD4+ T cells, can be formed by protein splicing. Here, we review the discovery of spliced epitopes recognized by tumor-specific human CD8+ T cells. We discuss how these epitopes are formed and some of the unusual variants that have been reported. Now, over a decade since the first report, evidence is emerging that spliced CD8+ T-cell epitopes are much more common, and potentially much more important, than previously imagined. Recent work has shown that epitopes recognized by CD4+ T cells can also be formed by protein splicing. We discuss the recent discovery of spliced CD4+ T-cell epitopes and their potential role as targets of autoimmune T-cell responses. Finally, we highlight some of the new questions raised from our growing appreciation of T-cell epitopes formed by peptide splicing.
Collapse
Affiliation(s)
- Stuart I Mannering
- Immunology and Diabetes Unit, St. Vincent's Institute of Medical Research, Fitzroy, Vic., Australia
| | - Michelle So
- Immunology and Diabetes Unit, St. Vincent's Institute of Medical Research, Fitzroy, Vic., Australia
| | - Colleen M Elso
- Immunology and Diabetes Unit, St. Vincent's Institute of Medical Research, Fitzroy, Vic., Australia
| | - Thomas W H Kay
- Immunology and Diabetes Unit, St. Vincent's Institute of Medical Research, Fitzroy, Vic., Australia
| |
Collapse
|
20
|
|
21
|
Nashida T, Shimomura-Kuroki J, Mizuhashi F, Haga-Tsujimura M, Yoshimura K, Hayashi-Sakai S. Presence of BPIFB1 in saliva from non-obese diabetic mice. Odontology 2017; 106:117-124. [PMID: 28748269 DOI: 10.1007/s10266-017-0312-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 05/22/2017] [Indexed: 01/09/2023]
Abstract
We previously showed that mRNA expression of BPIFB1 (Bpifb1), an antibacterial protein in the palate, lung, and nasal epithelium clone protein family, was increased in parotid acinar cells in non-obese diabetic (NOD, NOD/ShiJcl) mice, which is an animal model for Sjögren's syndrome. However, we did not previously assess the protein levels. In this report, we confirmed the expression of BPIFB1 protein in the parotid glands of NOD mice. Immunoblotting of subcellular fractions revealed that BPIBB1 was localised in secretory granules in parotid glands from NOD mice, and was almost not in parotid glands from the control mice. BPIFB1 had N-linked glycan that reacted with Aleuria aurantia lectin, which caused two types of spots with a slightly different pI and molecular weight. The expression of BPIFB1 protein was also demonstrated by immunohistochemistry. BPIFB1 was detected in the saliva from NOD mice but not in the saliva from the control mice, indicating individual constitution. BPIFB1 in saliva may be applied to other research as a diagnostic marker.
Collapse
Affiliation(s)
- Tomoko Nashida
- Department of Biochemistry, The Nippon Dental University School of Life Dentistry at Niigata, 1-8 Hamaura-cho, Chuo-ku, Niigata, 951-8580, Japan.
| | - Junko Shimomura-Kuroki
- Department of Pediatric Dentistry, The Nippon Dental University School of Life Dentistry at Niigata, 1-8 Hamaura-cho, Chuo-ku, Niigata, 951-8580, Japan
| | - Fumi Mizuhashi
- Department of Removable Prosthodontics, The Nippon Dental University School of Life Dentistry at Niigata, 1-8 Hamaura-cho, Chuo-ku, Niigata, 951-8580, Japan
| | - Maiko Haga-Tsujimura
- Department of Histology, The Nippon Dental University School of Life Dentistry at Niigata, 1-8 Hamaura-cho, Chuo-ku, Niigata, 951-8580, Japan
| | - Ken Yoshimura
- Department of Anatomy, The Nippon Dental University School of Life Dentistry at Niigata, 1-8 Hamaura-cho, Chuo-ku, Niigata, 951-8580, Japan
| | - Sachiko Hayashi-Sakai
- Division of Oral and Maxillofacial Radiology, Niigata University Graduate School of Medical and Dental Science, 2-5274 Gakkocho-dori, Chuo-ku, Niigata, 951-8514, Japan
| |
Collapse
|
22
|
Unanue ER, Ferris ST, Carrero JA. The role of islet antigen presenting cells and the presentation of insulin in the initiation of autoimmune diabetes in the NOD mouse. Immunol Rev 2016; 272:183-201. [PMID: 27319351 PMCID: PMC4938008 DOI: 10.1111/imr.12430] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
We have been examining antigen presentation and the antigen presenting cells (APCs) in the islets of Langerhans of the non-obese diabetic (NOD) mouse. The purpose is to identify the earliest events that initiate autoimmunity in this confined tissue. Islets normally have a population of macrophages that is distinct from those that inhabit the exocrine pancreas. Also found in NOD islets is a minor population of dendritic cells (DCs) that bear the CD103 integrin. We find close interactions between beta cells and the two APCs that result in the initiation of the autoimmunity. Even under non-inflammatory conditions, beta cells transfer insulin-containing vesicles to the APCs of the islet. This reaction requires live cells and intimate contact. The autoimmune process starts in islets with the entrance of CD4(+) T cells and an increase in the CD103(+) DCs. Mice deficient in the Batf3 transcription factor never develop diabetes due to the absence of the CD103/CD8α lineage of DCs. We hypothesize that the 12-20 peptide of the beta chain of insulin is responsible for activation of the initial CD4(+) T-cell response during diabetogenesis.
Collapse
Affiliation(s)
- Emil R. Unanue
- Department of Pathology and Immunology, Division of Immunobiology, 660 South Euclid Avenue, Campus Box 8118, Washington University School of Medicine, St. Louis, Missouri USA 63110
| | - Stephen T. Ferris
- Department of Pathology and Immunology, Division of Immunobiology, 660 South Euclid Avenue, Campus Box 8118, Washington University School of Medicine, St. Louis, Missouri USA 63110
| | - Javier A. Carrero
- Department of Pathology and Immunology, Division of Immunobiology, 660 South Euclid Avenue, Campus Box 8118, Washington University School of Medicine, St. Louis, Missouri USA 63110
| |
Collapse
|
23
|
Hillhouse EE, Liston A, Collin R, Desautels E, Goodnow CC, Lesage S. TCR transgenic mice reveal the impact of type 1 diabetes loci on early and late disease checkpoints. Immunol Cell Biol 2016; 94:709-13. [PMID: 27046082 DOI: 10.1038/icb.2016.27] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Revised: 03/03/2016] [Accepted: 03/03/2016] [Indexed: 12/29/2022]
Abstract
Linkage analysis studies for autoimmune diabetes have revealed multiple non-major histocompatibility complex (MHC) chromosomal regions linked to disease susceptibility. To date, more than 20 insulin-dependent diabetes (Idd) loci linked to diabetes susceptibility have been identified in NOD mice and validated via congenic breeding. Importantly, evidence suggests that Idd loci may regulate at least two pathological steps during autoimmune diabetes development, namely the onset of insulitis and the transition from insulitis to overt diabetes. Here we assess the role of various non-MHC Idd diabetes-resistance loci, which have been validated in the non-transgenic setting, on autoimmune diabetes progression in the transgenic setting. Specifically, we generated multiple Idd congenic strains in the 3A9-TCR:insHEL NOD.H2(k) transgenic model and monitored their diabetes incidence. We show that 3A9-TCR:insHEL NOD.H2(k) mice congenic for Idd3 or Idd5 display a reduction in diabetes development, whereas mice congenic for Idd9 or Idd13 exhibit an increase, in comparison with 3A9-TCR:insHEL NOD.H2(k) mice. These results suggest that the presence of the 3A9-TCR and hen egg lysosyme transgenes can offset the regulatory function of certain diabetes-resistance genetic variants contained within the Idd loci, including Idd9 and Idd13. We propose the antigen-specific 3A9-TCR:insHEL transgenic model as a useful tool for the study of the genetics of autoimmune diabetes development.
Collapse
Affiliation(s)
- Erin E Hillhouse
- Immunology-Oncology Section, Research Center, Maisonneuve-Rosemont Hospital, Montréal, Québec, Canada.,Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, Québec, Canada
| | - Adrian Liston
- Autoimmune Genetics Laboratory, Department of Microbiology and Immunology, VIB, Leuven, Belgium.,University of Leuven, Leuven, Belgium
| | - Roxanne Collin
- Immunology-Oncology Section, Research Center, Maisonneuve-Rosemont Hospital, Montréal, Québec, Canada.,Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, Québec, Canada
| | - Eric Desautels
- Immunology-Oncology Section, Research Center, Maisonneuve-Rosemont Hospital, Montréal, Québec, Canada.,Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, Québec, Canada
| | - Christopher C Goodnow
- Department of Immunology, John Curtin School of Medical Research, Australian National University, Canberra, Australian Capital Territory, Australia.,Immunogenomics Group, Immunology Research Program, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Sylvie Lesage
- Immunology-Oncology Section, Research Center, Maisonneuve-Rosemont Hospital, Montréal, Québec, Canada.,Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
24
|
Nashida T, Yoshimura K, Yoshie S, Mizuhashi F, Shimomura-Kuroki J. Upregulation of Bpifb1 expression in the parotid glands of non-obese diabetic mice. Oral Dis 2016; 22:46-52. [PMID: 26769076 DOI: 10.1111/odi.12377] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 09/14/2015] [Accepted: 09/30/2015] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To define the increased mRNA expression of Bpifb1, a member of the bactericidal/permeability-increasing protein family, in parotid acinar cells from non-obese diabetic (NOD) mice, an animal model for Sjögren's syndrome. MATERIALS AND METHODS Parotid acinar cells were prepared from female NOD (NOD/ShiJcl) mice with or without diabetes, as well as from control (C57BL/6JJcl) mice. Total RNA and homogenate were prepared from the parotid acinar cells. Embryonic cDNA from a Mouse MTC(™) Panel I kit was used. The expression of Bpifb1 was determined by cDNA microarray analysis, RT-PCR, real-time PCR, northern blotting and in situ hybridization. RESULTS The expression of Bpifb1 mRNA was high in parotid acinar cells from diabetic and non-diabetic NOD mice at 5-50 weeks of age. Acinar cells in the C57BL/6 mice had a low expression of Bpifb1 mRNA at an age >8 weeks, but had a relatively high expression in the foetus and infantile stages. CONCLUSIONS Bpifb1 mRNA is upregulated in parotid acinar cells in NOD mice, but its expression is not related to the onset of diabetes. These findings suggest that high expression levels of Bpifb1 might predict disease traits before the onset of autoimmunity.
Collapse
Affiliation(s)
- T Nashida
- Departments of Biochemistry, The Nippon Dental University School of Life Dentistry at Niigata, Chuo-ku, Niigata, Japan
| | - K Yoshimura
- Departments of Anatomy, The Nippon Dental University School of Life Dentistry at Niigata, Chuo-ku, Niigata, Japan
| | - S Yoshie
- Departments of Histology, The Nippon Dental University School of Life Dentistry at Niigata, Chuo-ku, Niigata, Japan
| | - F Mizuhashi
- Departments of Removable Prosthodontics, The Nippon Dental University School of Life Dentistry at Niigata, Chuo-ku, Niigata, Japan
| | - J Shimomura-Kuroki
- Departments of Pediatric Dentistry, The Nippon Dental University School of Life Dentistry at Niigata, Chuo-ku, Niigata, Japan
| |
Collapse
|
25
|
Abstract
Type 1 diabetes (T1D) results from a chronic and selective destruction of insulin-secreting β-cells within the islets of Langerhans of the pancreas by autoreactive CD4(+) and CD8(+) T lymphocytes. The use of animal models of T1D was instrumental for deciphering the steps of the autoimmune process leading to T1D. The non-obese diabetic (NOD) mouse and the bio-breeding (BB) rat spontaneously develop the disease similar to the human pathology in terms of the immune responses triggering autoimmune diabetes and of the genetic and environmental factors influencing disease susceptibility. The generation of genetically modified models allowed refining our understanding of the etiology and the pathogenesis of the disease. In the present review, we provide an overview of the experimental models generated and used to gain knowledge on the molecular and cellular mechanisms underlying the breakdown of self-tolerance in T1D and the progression of the autoimmune response. Immunotherapeutic interventions designed in these animal models and translated into the clinical arena in T1D patients will also be discussed.
Collapse
|
26
|
Pearson JA, Wong FS, Wen L. The importance of the Non Obese Diabetic (NOD) mouse model in autoimmune diabetes. J Autoimmun 2015; 66:76-88. [PMID: 26403950 DOI: 10.1016/j.jaut.2015.08.019] [Citation(s) in RCA: 188] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 08/26/2015] [Indexed: 02/06/2023]
Abstract
Type 1 Diabetes (T1D) is an autoimmune disease characterized by the pancreatic infiltration of immune cells resulting in T cell-mediated destruction of the insulin-producing beta cells. The successes of the Non-Obese Diabetic (NOD) mouse model have come in multiple forms including identifying key genetic and environmental risk factors e.g. Idd loci and effects of microorganisms including the gut microbiota, respectively, and how they may contribute to disease susceptibility and pathogenesis. Furthermore, the NOD model also provides insights into the roles of the innate immune cells as well as the B cells in contributing to the T cell-mediated disease. Unlike many autoimmune disease models, the NOD mouse develops spontaneous disease and has many similarities to human T1D. Through exploiting these similarities many targets have been identified for immune-intervention strategies. Although many of these immunotherapies did not have a significant impact on human T1D, they have been shown to be effective in the NOD mouse in early stage disease, which is not equivalent to trials in newly-diagnosed patients with diabetes. However, the continued development of humanized NOD mice would enable further clinical developments, bringing T1D research to a new translational level. Therefore, it is the aim of this review to discuss the importance of the NOD model in identifying the roles of the innate immune system and the interaction with the gut microbiota in modifying diabetes susceptibility. In addition, the role of the B cells will also be discussed with new insights gained through B cell depletion experiments and the impact on translational developments. Finally, this review will also discuss the future of the NOD mouse and the development of humanized NOD mice, providing novel insights into human T1D.
Collapse
Affiliation(s)
- James A Pearson
- Section of Endocrinology, School of Medicine, Yale University, New Haven, CT, USA
| | - F Susan Wong
- Diabetes Research Group, Institute of Molecular & Experimental Medicine, School of Medicine, Cardiff University, Wales, UK
| | - Li Wen
- Section of Endocrinology, School of Medicine, Yale University, New Haven, CT, USA.
| |
Collapse
|
27
|
Racine JJ, Zhang M, Wang M, Morales W, Shen C, Zeng D. MHC-mismatched mixed chimerism mediates thymic deletion of cross-reactive autoreactive T cells and prevents insulitis in nonobese diabetic mice. THE JOURNAL OF IMMUNOLOGY 2014; 194:407-17. [PMID: 25429069 DOI: 10.4049/jimmunol.1401584] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Type 1 diabetic NOD mice have defects in both thymic negative selection and peripheral regulation of autoreactive T cells, and induction of mixed chimerism can effectively reverse these defects. Our recent studies suggest that MHC-mismatched mixed chimerism mediates negative selection of autoreactive thymocytes in wild-type NOD and TCR-transgenic NOD.Rag1(+/+).BDC2.5 mice. However, it remains unknown how mismatched I-A(b) MHC class II can mediate deletion of autoreactive T cells positively selected by I-A(g7). In the present study, we directly tested the hypothesis that mismatched MHC class II in mixed chimeras mediates deletion of cross-reactive autoreactive thymocytes. We first identify that transgenic BDC2.5 T cells from NOD.Rag1(+/+).BDC2.5 but not NOD.Rag1(-/-).BDC2.5 mice possess cross-reactive TCRs with endogenous TCRα-chains; MHC-mismatched H-2(b) but not matched H-2(g7) mixed chimerism mediates thymic deletion of the cross-reactive transgenic T cells in NOD.Rag1(+/+).BDC2.5 mice. Second, by transplanting T cell-depleted (TCD) bone marrow (BM) cells from NOD.Rag1(+/+).BDC2.5 or NOD.Rag1(-/-).BDC2.5 mice into lethally irradiated MHC-mismatched H-2(b) C57BL/6 or MHC-matched congenic B6.H-2(g7) recipients, we demonstrate that NOD.Rag1(+/+).BDC2.5 BM-derived cross-reactive transgenic T cells, but not NOD.Rag1(-/-).BDC2.5 BM-derived non-cross-reactive transgenic T cells, can be positively selected in MHC-mismatched H-2(b) thymus. Third, by cotransplanting NOD.Rag1(+/+).BDC2.5 TCD BM cells with BM cells from MHC-mismatched T cell-deficient C57BL/6 mice into lethally irradiated MHC-matched B6.H-2(g7) recipients, we demonstrate that thymic deletion of the cross-reactive transgenic T cells is dependent on MHC-mismatched donor BM-derived APCs but not on donor BM-derived T cells. Taken together, our studies indicate that MHC-mismatched mixed chimerism can mediate thymic deletion of cross-reactive autoreactive T cells that express more than one TCR.
Collapse
Affiliation(s)
- Jeremy J Racine
- Irell and Manella Graduate School of Biological Sciences, City of Hope, Duarte, CA 91010; Department of Diabetes Research, Beckman Research Institute, City of Hope, Duarte, CA 91010; Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope, Duarte, CA 91010; and
| | - Mingfeng Zhang
- Department of Diabetes Research, Beckman Research Institute, City of Hope, Duarte, CA 91010; Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope, Duarte, CA 91010; and
| | - Miao Wang
- Department of Diabetes Research, Beckman Research Institute, City of Hope, Duarte, CA 91010; Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope, Duarte, CA 91010; and
| | - William Morales
- Eugene and Ruth Roberts Summer Student Academy, Beckman Research Institute, City of Hope, Duarte, CA 91010
| | - Christine Shen
- Eugene and Ruth Roberts Summer Student Academy, Beckman Research Institute, City of Hope, Duarte, CA 91010
| | - Defu Zeng
- Irell and Manella Graduate School of Biological Sciences, City of Hope, Duarte, CA 91010; Department of Diabetes Research, Beckman Research Institute, City of Hope, Duarte, CA 91010; Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope, Duarte, CA 91010; and
| |
Collapse
|
28
|
Aydintug MK, Zhang L, Wang C, Liang D, Wands JM, Michels AW, Hirsch B, Day BJ, Zhang G, Sun D, Eisenbarth GS, O'Brien RL, Born WK. γδ T cells recognize the insulin B:9-23 peptide antigen when it is dimerized through thiol oxidation. Mol Immunol 2014; 60:116-28. [PMID: 24853397 PMCID: PMC4091716 DOI: 10.1016/j.molimm.2014.04.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2014] [Revised: 04/14/2014] [Accepted: 04/20/2014] [Indexed: 01/08/2023]
Abstract
The insulin peptide B:9-23 is a natural antigen in the non-obese diabetic (NOD) mouse model of type 1 diabetes (T1D). In addition to αβ T cells and B cells, γδ T cells recognize the peptide and infiltrate the pancreatic islets where the peptide is produced within β cells. The peptide contains a cysteine in position 19 (Cys19), which is required for the γδ but not the αβ T cell response, and a tyrosine in position 16 (Tyr16), which is required for both. A peptide-specific mAb, tested along with the T cells, required neither of the two amino acids to bind the B:9-23 peptide. We found that γδ T cells require Cys19 because they recognize the peptide antigen in an oxidized state, in which the Cys19 thiols of two peptide molecules form a disulfide bond, creating a soluble homo-dimer. In contrast, αβ T cells recognize the peptide antigen as a reduced monomer, in complex with the MHCII molecule I-A(g7). Unlike the unstructured monomeric B:9-23 peptide, the γδ-stimulatory homo-dimer adopts a distinct secondary structure in solution, which differs from the secondary structure of the corresponding portion of the native insulin molecule. Tyr16 is required for this adopted structure of the dimerized insulin peptide as well as for the γδ response to it. This observation is consistent with the notion that γδ T cell recognition depends on the secondary structure of the dimerized insulin B:9-23 antigen.
Collapse
Affiliation(s)
- M Kemal Aydintug
- Integrated Department of Immunology, National Jewish Health and University of Colorado Denver, 1400 Jackson Street, Denver, CO 80206, USA
| | - Li Zhang
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, CO 80045, USA
| | - Chao Wang
- Integrated Department of Immunology, National Jewish Health and University of Colorado Denver, 1400 Jackson Street, Denver, CO 80206, USA
| | - Dongchun Liang
- Department of Ophthalmology, Doheny Eye Institute, University of Southern California, Los Angeles, CA 90033, USA
| | - J M Wands
- Integrated Department of Immunology, National Jewish Health and University of Colorado Denver, 1400 Jackson Street, Denver, CO 80206, USA
| | - Aaron W Michels
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, CO 80045, USA
| | - Brooke Hirsch
- Department of Biomolecular Structure, University of Colorado Denver, Anschutz Medical Campus, Aurora CO 80045, USA
| | - Brian J Day
- Department of Medicine, National Jewish Health, 1400 Jackson Street, CO 80206, USA
| | - Gongyi Zhang
- Integrated Department of Immunology, National Jewish Health and University of Colorado Denver, 1400 Jackson Street, Denver, CO 80206, USA
| | - Deming Sun
- Department of Ophthalmology, Doheny Eye Institute, University of Southern California, Los Angeles, CA 90033, USA
| | - George S Eisenbarth
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, CO 80045, USA
| | - Rebecca L O'Brien
- Integrated Department of Immunology, National Jewish Health and University of Colorado Denver, 1400 Jackson Street, Denver, CO 80206, USA
| | - Willi K Born
- Integrated Department of Immunology, National Jewish Health and University of Colorado Denver, 1400 Jackson Street, Denver, CO 80206, USA.
| |
Collapse
|
29
|
Wakeland EK. Hunting Autoimmune Disease Genes in NOD: Early Steps on a Long Road to Somewhere Important (Hopefully). THE JOURNAL OF IMMUNOLOGY 2014; 193:3-6. [DOI: 10.4049/jimmunol.1401200] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
30
|
Abstract
This paper reviews the presentation of peptides by major histocompatibility complex (MHC) class II molecules in the autoimmune diabetes of the nonobese diabetic (NOD) mouse. Islets of Langerhans contain antigen-presenting cells that capture the proteins and peptides of the beta cells' secretory granules. Peptides bound to I-A(g7), the unique MHC class II molecule of NOD mice, are presented in islets and in pancreatic lymph nodes. The various beta cell-derived peptides interact with selected CD4 T cells to cause inflammation and beta cell demise. Many autoreactive T cells are found in NOD mice, but not all have a major role in the initiation of the autoimmune process. I emphasize here the evidence pointing to insulin autoreactivity as a seminal component in the diabetogenic process.
Collapse
Affiliation(s)
- Emil R Unanue
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri 63110;
| |
Collapse
|
31
|
Hisanaga-Oishi Y, Nishiwaki-Ueda Y, Nojima K, Ueda H. Analysis of the expression of candidate genes for type 1 diabetes susceptibility in T cells. Endocr J 2014; 61:577-88. [PMID: 24705559 DOI: 10.1507/endocrj.ej14-0002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Type 1 diabetes is characterized by T-cell-mediated autoimmune destruction of pancreatic β-cells. Currently, approximately 50 type 1 diabetes susceptibility genes or chromosomal regions have been identified. However, the functions of type 1 diabetes susceptibility genes in T cells are elusive. In this study, we evaluated the correlation between type 1 diabetes susceptibility genes and T-cell signaling. The expression levels of 22 candidate type 1 diabetes susceptibility genes in T cells from nonobese diabetic (NOD), control C57BL/6 (B6), and NOD-control F1 hybrid mice were analyzed in response to 2 key immunoregulatory cytokines: interleukin-2 (IL-2) and transforming growth factor β (TGF-β). Exogenous gene expression studies were also performed in EL4 and Jurkat E6.1 T-cell lines. Significant differences in the expression of Clec16a, Dlk1, Il2, Ptpn22, Rnls, and Zac1 (also known as Plagl1) were observed in T cells derived from the 3 strains of mice, and TGF-β differentially influenced the expression of Ctla4, Foxp3, Il2, Ptpn22, Sh2b3, and Zac1. We found that TGF-β induced Zac1 expression in both primary T cells and EL4 cells and that exogenous expression of Zac1 and ZAC1 in T-cell lines altered the expression of Il2 and DLK1, respectively. The results of our study indicate the possibility that additional genetic pathways underlying type 1 diabetes susceptibility, including those involving Clec16a, Dlk1, Rnls, Sh2b3, and Zac1 under IL-2 and TGF-β signaling in T cells, may be shared between human and NOD mice.
Collapse
Affiliation(s)
- Yuko Hisanaga-Oishi
- Department of Molecular Endocrinology, Osaka University Graduate School of Medicine, Suita 565-0871, Japan
| | | | | | | |
Collapse
|
32
|
Ren B, O'Brien BA, Byrne MR, Ch'ng E, Gatt PN, Swan MA, Nassif NT, Wei MQ, Gijsbers R, Debyser Z, Simpson AM. Long-term reversal of diabetes in non-obese diabetic mice by liver-directed gene therapy. J Gene Med 2013; 15:28-41. [PMID: 23293075 DOI: 10.1002/jgm.2692] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Revised: 10/01/2012] [Accepted: 12/20/2012] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Type 1 diabetes (T1D) results from an autoimmune attack against the insulin-producing β-cells of the pancreas. The present study aimed to reverse T1D by gene therapy. METHODS We used a novel surgical technique, which involves isolating the liver from the circulation before the delivery of a lentiviral vector carrying furin-cleavable human insulin (INS-FUR) or empty vector to the livers of diabetic non-obese diabetic mice (NOD). This was compared with the direct injection of the vector into the portal circulation. Mice were monitored for body weight and blood glucose. Intravenous glucose tolerance tests were performed. Expression of insulin and pancreatic transcription factors was determined by the reverse transcriptase-polymerase chain reaction and immunohistochemistry and immunoelectron microscopy was used to localise insulin. RESULTS Using the novel surgical technique, we achieved long-term transduction (42% efficiency) of hepatocytes, restored normoglycaemia for 150 days (experimental endpoint) and re-established normal glucose tolerance. We showed the expression of β-cell transcription factors, murine insulin, glucagon and somatostatin, and hepatic storage of insulin in granules. The expression of hepatic markers, C/EBP-β, G6PC, AAT and GLUI was down-regulated in INS-FUR-treated livers. Liver function tests remained normal, with no evidence of intrahepatic inflammation or autoimmune destruction of the insulin-secreting liver tissue. By comparison, direct injection of INS-FUR reduced blood glucose levels, and no pancreatic transdifferentiation or normal glucose tolerance was observed. CONCLUSIONS This gene therapy protocol has, for the first time, permanently reversed T1D with normal glucose tolerance in NOD mice and, as such, represents a novel therapeutic strategy for the treatment of T1D.
Collapse
Affiliation(s)
- Binhai Ren
- School of Medical & Molecular Biosciences, University of Technology Sydney, Sydney, Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Bour-Jordan H, Thompson HL, Giampaolo JR, Davini D, Rosenthal W, Bluestone JA. Distinct genetic control of autoimmune neuropathy and diabetes in the non-obese diabetic background. J Autoimmun 2013; 45:58-67. [PMID: 23850635 PMCID: PMC4156399 DOI: 10.1016/j.jaut.2013.06.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Accepted: 06/11/2013] [Indexed: 02/01/2023]
Abstract
The non-obese diabetic (NOD) mouse is susceptible to the development of autoimmune diabetes but also multiple other autoimmune diseases. Over twenty susceptibility loci linked to diabetes have been identified in NOD mice and progress has been made in the definition of candidate genes at many of these loci (termed Idd for insulin-dependent diabetes). The susceptibility to multiple autoimmune diseases in the NOD background is a unique opportunity to examine susceptibility genes that confer a general propensity for autoimmunity versus susceptibility genes that control individual autoimmune diseases. We previously showed that NOD mice deficient for the costimulatory molecule B7-2 (NOD-B7-2KO mice) were protected from diabetes but spontaneously developed an autoimmune peripheral neuropathy. Here, we took advantage of multiple NOD mouse strains congenic for Idd loci to test the role of these Idd loci the development of neuropathy and determine if B6 alleles at Idd loci that are protective for diabetes will also be for neuropathy. Thus, we generated NOD-B7-2KO strains congenic at Idd loci and examined the development of neuritis and clinical neuropathy. We found that the NOD-H-2(g7) MHC region is necessary for development of neuropathy in NOD-B7-2KO mice. In contrast, other Idd loci that significantly protect from diabetes did not affect neuropathy when considered individually. However, we found potent genetic interactions of some Idd loci that provided almost complete protection from neuritis and clinical neuropathy. In addition, defective immunoregulation by Tregs could supersede protection by some, but not other, Idd loci in a tissue-specific manner in a model where neuropathy and diabetes occurred concomitantly. Thus, our study helps identify Idd loci that control tissue-specific disease or confer general susceptibility to autoimmunity, and brings insight to the Treg-dependence of autoimmune processes influenced by given Idd region in the NOD background.
Collapse
Affiliation(s)
- Hélène Bour-Jordan
- University of California in San Francisco, 513 Parnassus Avenue, Box 0400, San Francisco, CA 94143-0400, USA
| | | | | | | | | | | |
Collapse
|
34
|
De Riva A, Varley MC, Bluck LJ, Cooke A, Deery MJ, Busch R. Accelerated turnover of MHC class II molecules in nonobese diabetic mice is developmentally and environmentally regulated in vivo and dispensable for autoimmunity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2013; 190:5961-71. [PMID: 23677470 PMCID: PMC3785126 DOI: 10.4049/jimmunol.1300551] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The H2-A(g7) (A(g7)) MHC class II (MHCII) allele is required for type 1 diabetes (T1D) in NOD mice. A(g7) not only has a unique peptide-binding profile, it was reported to exhibit biochemical defects, including accelerated protein turnover. Such defects were proposed to impair Ag presentation and, thus, self-tolerance. Here, we report measurements of MHCII protein synthesis and turnover in vivo. NOD mice and BALB/c controls were labeled continuously with heavy water, and splenic B cells and dendritic cells were isolated. MHCII molecules were immunoprecipitated and digested with trypsin. Digests were analyzed by liquid chromatography/mass spectrometry to quantify the fraction of newly synthesized MHCII molecules and, thus, turnover. MHCII turnover was faster in dendritic cells than in B cells, varying slightly between mouse strains. Some A(g7) molecules exhibited accelerated turnover in B cells from young, but not older, prediabetic female NOD mice. This acceleration was not detected in a second NOD colony with a high incidence of T1D. Turnover rates of A(g7) and H2-A(d) were indistinguishable in (NOD × BALB/c) F1 mice. In conclusion, accelerated MHCII turnover may occur in NOD mice, but it reflects environmental and developmental regulation, rather than a structural deficit of the A(g7) allele. Moreover, this phenotype wanes before the onset of overt T1D and is dispensable for the development of autoimmune diabetes. Our observations highlight the importance of in vivo studies in understanding the role of protein turnover in genotype/phenotype relationships and offer a novel approach for addressing this fundamental research challenge.
Collapse
Affiliation(s)
| | - Mark C. Varley
- Department of Medicine, University of Cambridge, Cambridge, UK
- Department of Engineering, University of Cambridge, Cambridge, UK
| | - Leslie J. Bluck
- Elsie Widdowson Laboratories, Medical Research Council Human Nutrition Research, Fulbourn, Cambridge, UK
| | - Anne Cooke
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Michael J. Deery
- Cambridge Centre for Proteomics, University of Cambridge, Cambridge, UK
| | - Robert Busch
- Department of Medicine, University of Cambridge, Cambridge, UK
| |
Collapse
|
35
|
Morgan MAJ, Muller PSS, Mould A, Newland SA, Nichols J, Robertson EJ, Cooke A, Bikoff EK. The nonconventional MHC class II molecule DM governs diabetes susceptibility in NOD mice. PLoS One 2013; 8:e56738. [PMID: 23418596 PMCID: PMC3572069 DOI: 10.1371/journal.pone.0056738] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Accepted: 01/14/2013] [Indexed: 12/30/2022] Open
Abstract
The spontaneous destruction of insulin producing pancreatic beta cells in non-obese diabetic (NOD) mice provides a valuable model of type 1 diabetes. As in humans, disease susceptibility is controlled by the classical MHC class II genes that guide CD4+ T cell responses to self and foreign antigens. It has long been suspected that the dedicated class II chaperone designated HLA-DM in humans or H-2M in mice also makes an important contribution, but due to tight linkage within the MHC, a possible role played by DM peptide editing has not been previously tested by conventional genetic approaches. Here we exploited newly established germ-line competent NOD ES cells to engineer a loss of function allele. DM deficient NOD mice display defective class II peptide occupancy and surface expression, and are completely protected against type 1 diabetes. Interestingly the mutation results in increased proportional representation of CD4+Foxp3+ regulatory T cells and the absence of pathogenic CD4+ T effectors. Overall, this striking phenotype establishes that DM-mediated peptide selection plays an essential role in the development of autoimmune diabetes in NOD mice.
Collapse
MESH Headings
- Animals
- Antigens, Differentiation, B-Lymphocyte/genetics
- Antigens, Differentiation, B-Lymphocyte/immunology
- Antigens, Differentiation, B-Lymphocyte/metabolism
- Blotting, Western
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/metabolism
- Embryonic Stem Cells/immunology
- Embryonic Stem Cells/metabolism
- Female
- Forkhead Transcription Factors/immunology
- Forkhead Transcription Factors/metabolism
- Genetic Predisposition to Disease/genetics
- Histocompatibility Antigens Class II/genetics
- Histocompatibility Antigens Class II/immunology
- Histocompatibility Antigens Class II/metabolism
- Humans
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Inbred NOD
- Mice, Knockout
- Mice, SCID
- Microscopy, Confocal
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Thymus Gland/immunology
- Thymus Gland/metabolism
Collapse
Affiliation(s)
- Marc A. J. Morgan
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Pari S. S. Muller
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Arne Mould
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Stephen A. Newland
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Jennifer Nichols
- Wellcome Trust - Medical Research Council Cambridge Stem Cell Institute, Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | | | - Anne Cooke
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Elizabeth K. Bikoff
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
- * E-mail:
| |
Collapse
|
36
|
Chou FC, Chen HY, Chen SJ, Fang MC, Sytwu HK. Rodent models for investigating the dysregulation of immune responses in type 1 diabetes. J Diabetes Res 2013; 2013:138412. [PMID: 23671851 PMCID: PMC3647569 DOI: 10.1155/2013/138412] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2012] [Accepted: 02/07/2013] [Indexed: 12/02/2022] Open
Abstract
Type 1 diabetes (T1D) is an autoimmune disease mediated by T cells that selectively destroy the insulin-producing β cells. Previous reports based on epidemiological and animal studies have demonstrated that both genetic factors and environmental parameters can either promote or attenuate the progression of autoimmunity. In recent decades, several inbred rodent strains that spontaneously develop diabetes have been applied to the investigation of the pathogenesis of T1D. Because the genetic manipulation of mice is well developed (transgenic, knockout, and conditional knockout/transgenic), most studies are performed using the nonobese diabetic (NOD) mouse model. This paper will focus on the use of genetically manipulated NOD mice to explore the pathogenesis of T1D and to develop potential therapeutic approaches.
Collapse
Affiliation(s)
- Feng-Cheng Chou
- Department and Graduate Institute of Microbiology and Immunology, National Defense Medical Center, R8324, 161, Section 6, MinChuan East Road, Neihu, Taipei 114, Taiwan
| | - Heng-Yi Chen
- Department and Graduate Institute of Microbiology and Immunology, National Defense Medical Center, R8324, 161, Section 6, MinChuan East Road, Neihu, Taipei 114, Taiwan
| | - Shyi-Jou Chen
- Department of Pediatrics, Tri-Service General Hospital, 325, Section 2, Chenggong Road, Neihu, Taipei 114, Taiwan
| | - Mei-Cho Fang
- Laboratory Animal Center, National Defense Medical Center, Taipei 114, Taiwan
| | - Huey-Kang Sytwu
- Department and Graduate Institute of Microbiology and Immunology, National Defense Medical Center, R8324, 161, Section 6, MinChuan East Road, Neihu, Taipei 114, Taiwan
- *Huey-Kang Sytwu:
| |
Collapse
|
37
|
Vaughan K, Peters B, Mallone R, von Herrath M, Roep BO, Sette A. Navigating diabetes-related immune epitope data: resources and tools provided by the Immune Epitope Database (IEDB). Immunome Res 2013; 9. [PMID: 25140192 PMCID: PMC4134942 DOI: 10.4172/1745-7580.1000063] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Background The Immune Epitope Database (IEDB), originally focused on infectious diseases, was recently expanded to allergy, transplantation and autoimmunity diseases. Here we focus on diabetes, chosen as a prototype autoimmune disease. We utilize a combined tutorial and meta-analysis format, which demonstrates how common questions, related to diabetes epitopes can be answered. Results A total of 409 references are captured in the IEDB describing >2,500 epitopes from diabetes associated antigens. The vast majority of data were derived from GAD, insulin, IA-2/PTPRN, IGRP, ZnT8, HSP, and ICA-1, and the experiments related to T cell epitopes and MHC binding far outnumbers B cell assays. We illustrate how to search by specific antigens, epitopes or host. Other examples include searching for tetramers or epitopes restricted by specific alleles or assays of interest, or searching based on the clinical status of the host. Conclusions The inventory of all published diabetes epitope data facilitates its access for the scientific community. While the global collection of primary data from the literature reflects potential investigational biases present in the literature, the flexible search approach allows users to perform queries tailored to their preferences, including or excluding data as appropriate. Moreover, the analysis highlights knowledge gaps and identifies areas for future investigation.
Collapse
Affiliation(s)
- Kerrie Vaughan
- Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA
| | - Bjoern Peters
- Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA
| | - Roberto Mallone
- INSERM, U1016, Cochin Institute, DeAR Lab Avenir, Saint Vincent de Paul Hospital, 82 Avenue Denfert Rochereau, 75674 Paris Cedex 14, France
| | - Matthias von Herrath
- Developmental Immunology, La Jolla Institute for Allergy and Immunology, La Jolla, CA
| | - Bart O Roep
- Department for Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| | - Alessandro Sette
- Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA
| |
Collapse
|
38
|
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: 2.3] [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
|
39
|
Marrack P, Kappler JW. Do MHCII-presented neoantigens drive type 1 diabetes and other autoimmune diseases? Cold Spring Harb Perspect Med 2012; 2:a007765. [PMID: 22951444 PMCID: PMC3426820 DOI: 10.1101/cshperspect.a007765] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The strong association between particular MHCII alleles and type 1 diabetes is not fully understood. Two ideas that have been considered for many years are that autoimmunity is driven by (1) low-affinity CD4(+) T cells that escape thymic negative selection and respond to certain autoantigen peptides that are particularly well presented by particular MHCII molecules, or (2) CD4(+) T cells responding to neoantigens that are absent in the thymus, but uniquely created in the target tissue in the periphery and presented by particular MHCII alleles. Here we discuss the recent structural data in favor of the second idea. We review studies suggesting that peptide antigens recognized by autoimmune T cells are uniquely proteolytically processed and/or posttranslationally modified in the target tissue, thus allowing these T cells to escape deletion in the thymus during T-cell development. We postulate that an encounter with these tissue-specific neoantigenic peptides presented by the particular susceptible MHCII alleles in the peripheral tissues when accompanied by the appropriate inflammatory milieu activates these T-cell escapees leading to the onset of autoimmune disease.
Collapse
Affiliation(s)
- Philippa Marrack
- Howard Hughes Medical Institute and Integrated Department of Immunology, National Jewish Health, University of Colorado Denver, School of Medicine, Denver, CO 80206, USA
| | | |
Collapse
|
40
|
On the perils of poor editing: regulation of peptide loading by HLA-DQ and H2-A molecules associated with celiac disease and type 1 diabetes. Expert Rev Mol Med 2012; 14:e15. [PMID: 22805744 DOI: 10.1017/erm.2012.9] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
This review discusses mechanisms that link allelic variants of major histocompatibility complex (MHC) class II molecules (MHCII) to immune pathology. We focus on HLA (human leukocyte antigen)-DQ (DQ) alleles associated with celiac disease (CD) and type 1 diabetes (T1D) and the role of the murine DQ-like allele, H2-Ag7 (I-Ag7 or Ag7), in murine T1D. MHCII molecules bind peptides, and alleles vary in their peptide-binding specificity. Disease-associated alleles permit binding of disease-inducing peptides, such as gluten-derived, Glu-/Pro-rich gliadin peptides in CD and peptides from islet autoantigens, including insulin, in T1D. In addition, the CD-associated DQ2.5 and DQ8 alleles are unusual in their interactions with factors that regulate their peptide loading, invariant chain (Ii) and HLA-DM (DM). The same alleles, as well as other T1D DQ risk alleles (and Ag7), share nonpolar residues in place of Asp at β57 and prefer peptides that place acidic side chains in a pocket in the MHCII groove (P9). Antigen-presenting cells from T1D-susceptible mice and humans retain CLIP because of poor DM editing, although underlying mechanisms differ between species. We propose that these effects on peptide presentation make key contributions to CD and T1D pathogenesis.
Collapse
|
41
|
|
42
|
Forbes JM, Söderlund J, Yap FYT, Knip M, Andrikopoulos S, Ilonen J, Simell O, Veijola R, Sourris KC, Coughlan MT, Forsblom C, Slattery R, Grey ST, Wessman M, Yamamoto H, Bierhaus A, Cooper ME, Groop PH. Receptor for advanced glycation end-products (RAGE) provides a link between genetic susceptibility and environmental factors in type 1 diabetes. Diabetologia 2011; 54:1032-42. [PMID: 21298413 DOI: 10.1007/s00125-011-2058-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2010] [Accepted: 12/14/2010] [Indexed: 01/31/2023]
Abstract
AIMS/HYPOTHESIS This group of studies examines human genetic susceptibility conferred by the receptor for advanced glycation end-products (RAGE) in type 1 diabetes and investigates how this may interact with a western environment. METHODS We analysed the AGER gene, using 13 tag SNPs, in 3,624 Finnish individuals from the FinnDiane study, followed by AGER associations with a high risk HLA genotype (DR3)-DQA1*05-DQB1*02/DRB1*0401-DQB1*0302 (n = 546; HLA-DR3/DR4), matched in healthy newborn infants from the Finnish Type 1 Diabetes Prediction and Prevention (DIPP) Study (n = 373) using allelic analysis. We also studied islets and circulating RAGE in NODLt mice. RESULTS The rs2070600 and rs17493811 polymorphisms predicted increased risk of type 1 diabetes, whereas the rs9469089 SNP was related to decreased risk, on a high risk HLA background. Children from the DIPP study also showed a decline in circulating soluble RAGE levels, at seroconversion to positivity for type 1 diabetes-associated autoantibodies. Islet RAGE and circulating soluble RAGE levels in prediabetic NODLt mice decreased over time and were prevented by the AGE lowering therapy alagebrium chloride. Alagebrium chloride also decreased the incidence of autoimmune diabetes and restored islet RAGE levels. CONCLUSIONS/INTERPRETATION These studies suggest that inherited AGER gene polymorphisms may confer susceptibility to environmental insults. Declining circulating levels of soluble RAGE, before the development of overt diabetes, may also be predictive of clinical disease in children with high to medium risk HLA II backgrounds and this possibility warrants further investigation in a larger cohort.
Collapse
Affiliation(s)
- J M Forbes
- Diabetes Complications Division, Baker IDI Heart and Diabetes Institute, St Kilda Rd Central, P.O. Box 6492, Melbourne, VIC 8008, Australia.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
[Antitumor effects of raddeanin A on S180, H22 and U14 cell xenografts in mice]. Curr Opin Immunol 2010; 20:111-8. [PMID: 18799026 DOI: 10.1016/j.coi.2007.11.005] [Citation(s) in RCA: 144] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2007] [Revised: 11/19/2007] [Accepted: 11/19/2007] [Indexed: 01/19/2023]
Abstract
BACKGROUND & OBJECTIVE Raddeanin A, a triterpenoid saponin from Anemone raddeana Regel, has good antitumor activity in vitro. This study was to investigate its antitumor effects on tumor cell xenografts in mice. METHODS The inhibitory effects of raddeanin A on the proliferation of human nasopharyngeal carcinoma KB cells and ovarian cancer SKOV3 cells were measured by MTT assay. The inhibitory effects of raddeanin A injection on the growth of sarcoma S180, liver cancer H22 and cervical carcinoma U14 cell xenografts in mice and the effect of raddeanin A lavage on the growth of S180 cell xenografts were measured. The acute toxicity of raddeanin A was also measured. RESULTS The 50% inhibition concentration (IC(50)) of raddeanin A was 4.64 microg/mL for KB cells and 1.40 microg/mL for SKOV3 cells. When injected with raddeanin A at a dose of 4.5 mg/kg, the growth inhibition rates of S180, H22 and U14 cell xenografts were 60.5%, 36.2% and 61.8%, respectively. When lavaged with raddeanin A at a dose of 200 mg/kg, the growth inhibition rate of S180 cell xenografts was 64.7%. The median lethal dose (LD50) of raddeanin A lavage was 1.1 g/kg and that of raddeanin A injection was 16.1 mg/kg. CONCLUSION Raddeanin A has good antitumor activity both in vitro and in vivo, and would be a potential antitumor medicine.
Collapse
|
44
|
Diabetogenic T cells recognize insulin bound to IAg7 in an unexpected, weakly binding register. Proc Natl Acad Sci U S A 2010; 107:10978-83. [PMID: 20534455 DOI: 10.1073/pnas.1006545107] [Citation(s) in RCA: 163] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
A peptide derived from the insulin B chain contains a major epitope for diabetogenic CD4(+) T cells in the NOD mouse model of type 1 diabetes (T1D). This peptide can fill the binding groove of the NOD MHCII molecule, IA(g7), in a number of ways or "registers." We show here that a diverse set of NOD anti-insulin T cells all recognize this peptide bound in the same register. Surprisingly, this register results in the poorest binding of peptide to IA(g7). The poor binding is due to an incompatibility between the p9 amino acid of the peptide and the unique IA(g7) p9 pocket polymorphisms that are strongly associated with susceptibility to T1D. Our findings suggest that the association of autoimmunity with particular MHCII alleles may be do to poorer, rather than more favorable, binding of the critical self-epitopes, allowing T-cell escape from thymic deletion.
Collapse
|
45
|
Stadinski B, Kappler J, Eisenbarth GS. Molecular targeting of islet autoantigens. Immunity 2010; 32:446-56. [PMID: 20412755 DOI: 10.1016/j.immuni.2010.04.008] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2010] [Revised: 04/02/2010] [Accepted: 04/05/2010] [Indexed: 12/11/2022]
Abstract
Type 1 diabetes of man and animal models results from immune-mediated specific beta cell destruction. Multiple islet antigens are targets of autoimmunity and most of these are not beta cell specific. Immune responses to insulin appear to be essential for the development of diabetes of the NOD mouse. In this review, we will emphasize the unusual manner in which selected autoantigenic peptides (particularly the recently discovered target of BDC2.5 T cells [chromagranin A]) are presented and recognized by autoreactive CD4(+) T cell receptors. We hypothesize that "unusual" structural interactions of specific trimolecular complexes (MHC class II, peptide, and T cell receptors) are fundamental to the escape from the thymus of autoreactive T cells able to cause type 1 diabetes.
Collapse
|
46
|
Abstract
The Banting Medal for Scientific Achievement Award is the American Diabetes Association's highest scientific award and honors an individual who has made significant, long-term contributions to the understanding of diabetes, its treatment, and/or prevention. The award is named after Nobel Prize winner Sir Frederick Banting, who codiscovered insulin treatment for diabetes. Dr. Eisenbarth received the American Diabetes Association's Banting Medal for Scientific Achievement at the Association's 69th Scientific Sessions, June 5–9, 2009, in New Orleans, Louisiana. He presented the Banting Lecture, An Unfinished Journey—Type 1 Diabetes—Molecular Pathogenesis to Prevention , on Sunday, June 7, 2009.
Collapse
Affiliation(s)
- George S Eisenbarth
- Barbara Davis Center for Childhood Diabetes, University of Colorado Health Sciences Center, Aurora, Colorado, USA.
| |
Collapse
|
47
|
Mori Y, Kodaka T, Kato T, Kanagawa EM, Kanagawa O. Critical role of IFN-gamma in CFA-mediated protection of NOD mice from diabetes development. Int Immunol 2009; 21:1291-9. [PMID: 19778991 DOI: 10.1093/intimm/dxp097] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
IFN-gamma signaling-deficient non-obese diabetic (NOD) mice develop diabetes with similar kinetics to those of wild-type NOD mice. However, the immunization of IFN-gamma signaling-deficient NOD mice with CFA failed to induce long-term protection, whereas wild-type NOD mice receiving CFA remained diabetes-free. CFA also failed to protect IFN-gamma receptor-deficient (IFN-gammaR(-/-)) NOD mice from the autoimmune rejection of transplanted islets, as it does in diabetic NOD mice, and from disease transfer by spleen cells from diabetic NOD mice. These data clearly show that the pro-inflammatory cytokine IFN-gamma is necessary for the CFA-mediated protection of NOD mice from diabetes. There is no difference in the T(h)1/T(h)17 balance between IFN-gammaR(-/-) NOD and wild-type NOD mice. There is also no difference in the total numbers and percentages of regulatory T (Treg) cells in the lymph node CD4(+) T-cell populations between IFN-gammaR(-/-) NOD and wild-type NOD mice. However, pathogenic T cells lacking IFN-gammaR are resistant to the suppressive effect of Treg cells, both in vivo and in vitro. Therefore, it is likely that CFA-mediated protection against diabetes development depends on a change in the balance between Treg cells and pathogenic T cells, and IFN-gamma signaling seems to control the susceptibility of pathogenic T cells to the inhibitory activity of Treg cells.
Collapse
Affiliation(s)
- Yoshiko Mori
- Laboratory for Autoimmune Regulation, Research Center for Allergy and Immunology, RIKEN Yokohama Institute, Yokohama, Kanagawa, Japan
| | | | | | | | | |
Collapse
|
48
|
Baxter AG, Jordan MA, Silveira PA, Wilson WE, Van Driel IR. Genetic Control of Susceptibility to Autoimmune Gastritis. Int Rev Immunol 2009; 24:55-62. [PMID: 15763989 DOI: 10.1080/08830180590884404] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
A familial component to the tendency to develop autoimmune gastritis has long been recognized. Although linkage to certain HLA alleles and an association with the endocrine autoimmune diseases thyroiditis and type 1 diabetes have been reported, little further progress has been achieved in clinical studies. In contrast, the mouse model of gastritis induced in the BALB/c strain by thymectomy in the third day of life has identified four linkage regions; two on distal chromosome 4 (Gasa1 and Gasa2), one on chromosome 6 (Gasa3) and one in the H2 (Gasa4). Three of these four genes colocalize with NOD mouse diabetes susceptibility genes--the strongest concordance identified to date between any two autoimmune diseases--reflecting the association between autoimmune diabetes and type 1 gastritis in humans.
Collapse
Affiliation(s)
- Alan G Baxter
- Comparative Genomics Centre, James Cook University, Townsville, QLD, Australia.
| | | | | | | | | |
Collapse
|
49
|
Fletcher MT, Baxter AG. Clinical application of NKT cell biology in type I (autoimmune) diabetes mellitus. Immunol Cell Biol 2009; 87:315-23. [PMID: 19223852 DOI: 10.1038/icb.2009.5] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Type 1 natural killer T (NKT) cells are a population of CD1d-restricted, regulatory T cells that exhibit various NK cell characteristics and rapidly produce cytokines on stimulation with glycolipid antigen. In type I diabetes (TID), NKT cells are thought to have a tolerogenic function, evidenced by NKT cell numerical and functional deficiencies in the nonobese diabetic (NOD) mouse, which when corrected, can ameliorate disease. The mechanisms by which NKT cells can mediate their immunosuppressive effects in NOD mice are still poorly understood, which makes successful clinical translation of NKT- cell-based therapies challenging. However, new insights into the genetic control of NKT cell deficiencies have provided some understanding of the genes that may control NKT cell number and function, potentially offering a new avenue for assessing TID risk in humans. Here, we review the mechanisms by which NKT cells are thought to prevent TID, discuss the evidence for involvement of NKT cells in the regulation of human TID and examine the genetic control of NKT cell number and function. A greater understanding of these areas will increase the chances of successful clinical manipulation of NKT cells to prevent or treat TID.
Collapse
Affiliation(s)
- Marie T Fletcher
- Department of Microbiology and Immunology, University of Melbourne, Parkville, Victoria, Australia
| | | |
Collapse
|
50
|
Whetzel AM, Bolick DT, Hedrick CC. Sphingosine-1-phosphate inhibits high glucose-mediated ERK1/2 action in endothelium through induction of MAP kinase phosphatase-3. Am J Physiol Cell Physiol 2008; 296:C339-45. [PMID: 19091959 DOI: 10.1152/ajpcell.00293.2008] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Endothelial activation is a key early event in vascular complications of Type 1 diabetes. The nonobese diabetic (NOD) mouse is a well-characterized model of Type 1 diabetes. We previously reported that Type 1 diabetic NOD mice have increased endothelial activation, with increased production of monocyte chemoattractant protein (MCP)-1 and IL-6, and a 30% increase of surface VCAM-1 expression leading to a fourfold increase in monocyte adhesion to the endothelium. Sphingosine-1-phosphate (S1P) prevents monocyte:endothelial interactions in these diabetic NOD mice. Incubation of diabetic NOD endothelial cells (EC) with S1P (100 nmol/l) reduced ERK1/2 phosphorylation by 90%, with no significant changes in total ERK1/2 protein. In the current study, we investigated the mechanism of S1P action on ERK1/2 to reduce activation of diabetic endothelium. S1P caused a significant threefold increase in mitogen-activated kinase phosphatase-3 (MKP-3) expression in EC. MKP-3 selectively regulates ERK1/2 activity through dephosphorylation. Incubation of diabetic NOD EC with S1P and the S1P(1)-selective agonist SEW2871 significantly increased expression of MKP-3 and reduced ERK1/2 phosphorylation, while incubation with the S1P(1)/S1P(3) antagonist VPC23019 decreased the expression of MKP-3, both results supporting a role for S1P(1) in MKP-3 regulation. To mimic the S1P-mediated induction of MKP-3 diabetic NOD EC, we overexpressed MKP-3 in human aortic endothelial cells (HAEC) cultured in elevated glucose (25 mmol/l). Overexpression of MKP-3 in glucose-cultured HAEC decreased ERK1/2 phosphorylation and resulted in decreased monocyte:endothelial interactions in a static monocyte adhesion assay. Finally, we used small interfering RNA to MKP-3 and observed increased monocyte adhesion. Moreover, S1P was unable to inhibit monocyte adhesion in the absence of MKP-3. Thus, one mechanism for the anti-inflammatory action of S1P in diabetic EC is inhibition of ERK1/2 phosphorylation through induction of MKP-3 expression via the S1P-S1P(1) receptor axis.
Collapse
Affiliation(s)
- Angela M Whetzel
- Cardiovascular Research Center, Univ. of Virginia, P. O. Box 801394, 415 Lane Rd., MR5 Rm. G123, Charlottesville, VA 22908, USA
| | | | | |
Collapse
|