1
|
Dong M, Audiger C, Adegoke A, Lebel MÈ, Valbon SF, Anderson CC, Melichar HJ, Lesage S. CD5 levels reveal distinct basal T-cell receptor signals in T cells from non-obese diabetic mice. Immunol Cell Biol 2021; 99:656-667. [PMID: 33534942 DOI: 10.1111/imcb.12443] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 01/11/2021] [Accepted: 02/01/2021] [Indexed: 12/15/2022]
Abstract
Type 1 diabetes in non-obese diabetic (NOD) mice occurs when autoreactive T cells eliminate insulin producing pancreatic β cells. While extensively studied in T-cell receptor (TCR) transgenic mice, the contribution of alterations in thymic selection to the polyclonal T-cell pool in NOD mice is not yet resolved. The magnitude of signals downstream of TCR engagement with self-peptide directs the development of a functional T-cell pool, in part by ensuring tolerance to self. TCR interactions with self-peptide are also necessary for T-cell homeostasis in the peripheral lymphoid organs. To identify differences in TCR signal strength that accompany thymic selection and peripheral T-cell maintenance, we compared CD5 levels, a marker of basal TCR signal strength, on immature and mature T cells from autoimmune diabetes-prone NOD and -resistant B6 mice. The data suggest that there is no preferential selection of NOD thymocytes that perceive stronger TCR signals from self-peptide engagement. Instead, NOD mice have an MHC-dependent increase in CD4+ thymocytes and mature T cells that express lower levels of CD5. In contrast, T cell-intrinsic mechanisms lead to higher levels of CD5 on peripheral CD8+ T cells from NOD relative to B6 mice, suggesting that peripheral CD8+ T cells with higher basal TCR signals may have survival advantages in NOD mice. These differences in the T-cell pool in NOD mice may contribute to the development or progression of autoimmune diabetes.
Collapse
Affiliation(s)
- Mengqi Dong
- Immunology-Oncology Unit, Maisonneuve-Rosemont Hospital Research Center, Montréal, Québec, Canada.,Département de microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, Québec, Canada
| | - Cindy Audiger
- Immunology-Oncology Unit, Maisonneuve-Rosemont Hospital Research Center, Montréal, Québec, Canada.,Département de microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, Québec, Canada
| | - Adeolu Adegoke
- Departments of Surgery, Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Marie-Ève Lebel
- Immunology-Oncology Unit, Maisonneuve-Rosemont Hospital Research Center, Montréal, Québec, Canada.,Département de microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, Québec, Canada
| | - Stefanie F Valbon
- Immunology-Oncology Unit, Maisonneuve-Rosemont Hospital Research Center, Montréal, Québec, Canada.,Département de microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, Québec, Canada
| | - Colin C Anderson
- Departments of Surgery and Medical Microbiology & Immunology, Alberta Diabetes Institute, Alberta Transplant Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Heather J Melichar
- Immunology-Oncology Unit, Maisonneuve-Rosemont Hospital Research Center, Montréal, Québec, Canada.,Département de Médecine, Université de Montréal, Montréal, Québec, Canada
| | - Sylvie Lesage
- Immunology-Oncology Unit, Maisonneuve-Rosemont Hospital Research Center, Montréal, Québec, Canada.,Département de microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
2
|
Collin R, Doyon K, Mullins-Dansereau V, Karam M, Chabot-Roy G, Hillhouse EE, Orthwein A, Lesage S. Genetic interaction between two insulin-dependent diabetes susceptibility loci, Idd2 and Idd13, in determining immunoregulatory DN T cell proportion. Immunogenetics 2018; 70:495-509. [PMID: 29696366 DOI: 10.1007/s00251-018-1060-8] [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: 02/09/2018] [Accepted: 04/12/2018] [Indexed: 12/21/2022]
Abstract
Several immune regulatory cell types participate in the protection against autoimmune diseases such as autoimmune diabetes. Of these immunoregulatory cells, we and others have shown that peripheral CD4-CD8- double negative (DN) T cells can induce antigen-specific immune tolerance. Particularly, we have described that diabetes-prone mice exhibit a lower number of peripheral DN T cells compared to diabetes-resistant mice. Identifying the molecular pathways that influence the size of the DN T cell pool in peripheral lymphoid organs may thus be of interest for maintaining antigen-specific immune tolerance. Hence, through immunogenetic approaches, we found that two genetic loci linked to autoimmune diabetes susceptibility, namely Idd2 and Idd13, independently contribute to the partial restoration of DN T cell proportion in secondary lymphoid organs. We now extend these findings to show an interaction between the Idd2 and Idd13 loci in determining the number of DN T cells in secondary lymphoid organs. Using bioinformatics tools, we link potential biological pathways arising from interactions of genes encoded within the two loci. By focusing on cell cycle, we validate that both the Idd2 and Idd13 loci influence RAD51 expression as well as DN T cell progression through the cell cycle. Altogether, we find that genetic interactions between Idd2 and Idd13 loci modulate cell cycle progression, which contributes, at least in part, to defining the proportion of DN T cells in secondary lymphoid organs.
Collapse
Affiliation(s)
- Roxanne Collin
- Division of Immunology-oncology, Maisonneuve-Rosemont Hospital, Research Center, Montréal, 5415 l'Assomption Blvd, Québec, H1T 2M4, Canada.,Département de microbiologie, infectiologie et immunologie, Université de Montréal, Montréal, Québec, H3C 3J7, Canada
| | - Kathy Doyon
- Division of Immunology-oncology, Maisonneuve-Rosemont Hospital, Research Center, Montréal, 5415 l'Assomption Blvd, Québec, H1T 2M4, Canada
| | - Victor Mullins-Dansereau
- Division of Immunology-oncology, Maisonneuve-Rosemont Hospital, Research Center, Montréal, 5415 l'Assomption Blvd, Québec, H1T 2M4, Canada.,Département de microbiologie, infectiologie et immunologie, Université de Montréal, Montréal, Québec, H3C 3J7, Canada
| | - Martin Karam
- Division of Experimental Medicine, McGill University, Montréal, Québec, H4A 3J1, Canada.,Lady Davis Institute, Jewish General Hospital, 3755 Côte Ste-Catherine, Montréal, Québec, H3T 1E2, Canada
| | - Geneviève Chabot-Roy
- Division of Immunology-oncology, Maisonneuve-Rosemont Hospital, Research Center, Montréal, 5415 l'Assomption Blvd, Québec, H1T 2M4, Canada
| | - Erin E Hillhouse
- Division of Immunology-oncology, Maisonneuve-Rosemont Hospital, Research Center, Montréal, 5415 l'Assomption Blvd, Québec, H1T 2M4, Canada
| | - Alexandre Orthwein
- Division of Experimental Medicine, McGill University, Montréal, Québec, H4A 3J1, Canada. .,Lady Davis Institute, Jewish General Hospital, 3755 Côte Ste-Catherine, Montréal, Québec, H3T 1E2, Canada. .,Department of Oncology, McGill University, Montréal, Québec, H4A 3J1, Canada.
| | - Sylvie Lesage
- Division of Immunology-oncology, Maisonneuve-Rosemont Hospital, Research Center, Montréal, 5415 l'Assomption Blvd, Québec, H1T 2M4, Canada. .,Département de microbiologie, infectiologie et immunologie, Université de Montréal, Montréal, Québec, H3C 3J7, Canada.
| |
Collapse
|
3
|
Bettini ML, Bettini M. Understanding Autoimmune Diabetes through the Prism of the Tri-Molecular Complex. Front Endocrinol (Lausanne) 2017; 8:351. [PMID: 29312143 PMCID: PMC5735072 DOI: 10.3389/fendo.2017.00351] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 11/30/2017] [Indexed: 12/15/2022] Open
Abstract
The strongest susceptibility allele for Type 1 Diabetes (T1D) is human leukocyte antigen (HLA), which supports a central role for T cells as the drivers of autoimmunity. However, the precise mechanisms that allow thymic escape and peripheral activation of beta cell antigen-specific T cells are still largely unknown. Studies performed with the non-obese diabetic (NOD) mouse have challenged several immunological dogmas, and have made the NOD mouse a key experimental system to study the steps of immunodysregulation that lead to autoimmune diabetes. The structural similarities between the NOD I-Ag7 and HLA-DQ8 have revealed the stability of the T cell receptor (TCR)/HLA/peptide tri-molecular complex as an important parameter in the development of autoimmune T cells, as well as afforded insights into the key antigens targeted in T1D. In this review, we will provide a summary of the current understanding with regard to autoimmune T cell development, the significance of the antigens targeted in T1D, and the relationship between TCR affinity and immune regulation.
Collapse
Affiliation(s)
- Matthew L. Bettini
- Pediatric Diabetes and Endocrinology, Baylor College of Medicine, Texas Children’s Hospital, McNair Medical Institute, Houston, TX, United States
- *Correspondence: Matthew L. Bettini, ; Maria Bettini,
| | - Maria Bettini
- Pediatric Diabetes and Endocrinology, Baylor College of Medicine, Texas Children’s Hospital, McNair Medical Institute, Houston, TX, United States
- *Correspondence: Matthew L. Bettini, ; Maria Bettini,
| |
Collapse
|
4
|
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: 0.3] [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
|
5
|
Pelletier AN, Guilbault L, Guimont-Desrochers F, Hillhouse EE, Lesage S. NK Cell Proportion and Number Are Influenced by Genetic Loci on Chromosomes 8, 9, and 17. THE JOURNAL OF IMMUNOLOGY 2016; 196:2627-36. [DOI: 10.4049/jimmunol.1502284] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 01/06/2016] [Indexed: 11/19/2022]
|
6
|
Dabbeche-Bouricha E, Araujo LM, Kato M, Prévost-Blondel A, Garchon HJ. Rapid dissemination of RET-transgene-driven melanoma in the presence of non-obese diabetic alleles: Critical roles of Dectin-1 and Nitric-oxide synthase type 2. Oncoimmunology 2015; 5:e1100793. [PMID: 27467912 DOI: 10.1080/2162402x.2015.1100793] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 09/19/2015] [Accepted: 09/22/2015] [Indexed: 10/22/2022] Open
Abstract
Mice transgenic for the RET oncogene provide a remarkable model for investigating the mechanisms underlying the promotion and the development of melanoma. This model was established on the C57BL/6 genetic background. In the present study, we investigated an effect of the strongly proinflammatory and autoimmune genetic makeup of the non-obese diabetic (NOD) strain. We bred (NODxB6)F1 mice and backcrossed them with NOD mice. F1 mice and mice at subsequent generations of backcrossing showed marked acceleration of tumor development, in particular with a more frequent and earlier extension of the primary uveal melanoma. In close relation with this severe evolution, we observed a profound drop in Dectin-1 expression on CD11b(+)Ly6G(+) granulocytic myeloid cells correlating with an expansion of CD4(+)Foxp3(+) T regulatory cell and of interferon(IFN)γ-producing CD8(+) T cell subsets in tumors. IFNγ is a major inducer of the type 2 nitric-oxide synthase (Nos2) gene whose products are known to be tumorigenic. Germline inactivation of the Nos2 gene was associated with a dramatically improved tumor prognosis and a restoration of Dectin-1 expression on myeloid cells. Moreover, in vivo treatment of (NODxB6)F1.RET(+) mice with curdlan, a glucose polymer that binds Dectin-1, prevented tumor extension and was associated with marked reduction of the CD4(+)Foxp3(+) T cell subset. These observations highlight the (NODxB6)F1.RET(+) mice as a new model to investigate the role of the immune system in the host-tumor relationship and point to Dectin-1 and Nos2 as potentially promising therapeutic targets.
Collapse
Affiliation(s)
- Emna Dabbeche-Bouricha
- Inserm U1173 and University of Versailles Saint-Quentin, Montigny-le-Bretonneux, France; Inserm U1016, CNRS UMR8104, Institut Cochin and University Paris Descartes, Paris, France
| | - Luiza M Araujo
- Inserm U1173 and University of Versailles Saint-Quentin , Montigny-le-Bretonneux, France
| | - Masashi Kato
- Nagoya University Graduate School of Medicine , Nagoya, Aichi, Japan
| | | | - Henri-Jean Garchon
- Inserm U1173 and University of Versailles Saint-Quentin, Montigny-le-Bretonneux, France; Ambroise Paré Hospital, Division of Genetics, Boulogne-Billancourt, France
| |
Collapse
|
7
|
Collin R, Dugas V, Pelletier AN, Chabot-Roy G, Lesage S. The mouse idd2 locus is linked to the proportion of immunoregulatory double-negative T cells, a trait associated with autoimmune diabetes resistance. THE JOURNAL OF IMMUNOLOGY 2014; 193:3503-12. [PMID: 25165153 DOI: 10.4049/jimmunol.1400189] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Autoimmune diseases result from a break in immune tolerance. Various mechanisms of peripheral tolerance can protect against autoimmunity, including immunoregulatory CD4(-)CD8(-) double-negative (DN) T cells. Indeed, we have previously shown that diabetes-prone mouse strains exhibit a low proportion of DN T cells relative to that of diabetes-resistant mice, and that a single autologous transfer of DN T cells can impede autoimmune diabetes development, at least in the 3A9 TCR transgenic setting. In this study, we aim to understand the genetic basis for the difference in DN T cell proportion between diabetes-resistant and diabetes-prone mice. We thus perform an unbiased linkage analysis in 3A9 TCR F2 (NOD.H2(k) × B10.BR) mice and reveal that a locus on chromosome 9, which coincides with Idd2, is linked to the proportion of DN T cells in the lymph nodes. We generate two NOD.H2(k).B10-Chr9 congenic mouse strains and validate the role of this genetic interval in defining the proportion of DN T cells. Moreover, we find that the increased proportion of DN T cells in lymphoid organs is associated with a decrease in both diabetes incidence and serum IgG Ab levels. Together, the data suggest that Idd2 is linked to DN T cell proportion and that a physiological increase in DN T cell number may be sufficient to confer resistance to autoimmune diabetes. Altogether, these findings could help identify new candidate genes for the development of therapeutic avenues aimed at modulating DN T cell number for the prevention of autoimmune diseases.
Collapse
Affiliation(s)
- Roxanne Collin
- Division of Immunology-Oncology, Research Center, Maisonneuve-Rosemont Hospital, Montreal, Quebec H1T 2M4, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, Quebec H3C 3J7, Canada; and
| | - Véronique Dugas
- Division of Immunology-Oncology, Research Center, Maisonneuve-Rosemont Hospital, Montreal, Quebec H1T 2M4, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, Quebec H3C 3J7, Canada; and
| | - Adam-Nicolas Pelletier
- Division of Immunology-Oncology, Research Center, Maisonneuve-Rosemont Hospital, Montreal, Quebec H1T 2M4, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, Quebec H3C 3J7, Canada; and
| | - Geneviève Chabot-Roy
- Division of Immunology-Oncology, Research Center, Maisonneuve-Rosemont Hospital, Montreal, Quebec H1T 2M4, Canada
| | - Sylvie Lesage
- Division of Immunology-Oncology, Research Center, Maisonneuve-Rosemont Hospital, Montreal, Quebec H1T 2M4, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, Quebec H3C 3J7, Canada; and Department of Microbiology and Immunology, McGill University, Montreal, Quebec H3A 0G4, Canada
| |
Collapse
|
8
|
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]
|
9
|
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: 1.3] [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
|
10
|
Review of Ets1 structure, function, and roles in immunity. Cell Mol Life Sci 2013; 70:3375-90. [PMID: 23288305 DOI: 10.1007/s00018-012-1243-7] [Citation(s) in RCA: 153] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Revised: 11/20/2012] [Accepted: 12/11/2012] [Indexed: 10/27/2022]
Abstract
The Ets1 transcription factor is a member of the Ets gene family and is highly conserved throughout evolution. Ets1 is known to regulate a number of important biological processes in normal cells and in tumors. In particular, Ets1 has been associated with regulation of immune cell function and with an aggressive behavior in tumors that express it at high levels. Here we review and summarize the general features of Ets1 and describe its roles in immunity and autoimmunity, with a focus on its roles in B lymphocytes. We also review evidence that suggests that Ets1 may play a role in malignant transformation of hematopoietic malignancies including B cell malignancies.
Collapse
|
11
|
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: 3.1] [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
|
12
|
Thayer TC, Wilson SB, Mathews CE. Use of nonobese diabetic mice to understand human type 1 diabetes. Endocrinol Metab Clin North Am 2010; 39:541-61. [PMID: 20723819 PMCID: PMC2925291 DOI: 10.1016/j.ecl.2010.05.001] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In 1922, Leonard Thompson received the first injections of insulin prepared from the pancreas of canine test subjects. From pancreatectomized dogs to the more recent development of animal models that spontaneously develop autoimmune syndromes, animal models have played a meaningful role in furthering diabetes research. Of these animals, the nonobese diabetic (NOD) mouse is the most widely used for research in type 1 diabetes (T1D) because the NOD shares several genetic and immunologic traits with the human form of the disease. In this article, the authors discuss the similarities and differences in NOD and human T1D and the potential role of NOD mice in future preclinical studies, aiming to provide a better understanding of the genetic and immune defects that lead to T1D.
Collapse
Affiliation(s)
- Terri C Thayer
- Department of Pathology, Immunology, and Laboratory Medicine, The University of Florida College of Medicine, Gainesville, FL 32610, USA
| | | | | |
Collapse
|
13
|
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: 10.9] [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
|
14
|
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: 3.1] [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
|
15
|
Driver JP, Serreze DV, Chen YG. Mouse models for the study of autoimmune type 1 diabetes: a NOD to similarities and differences to human disease. Semin Immunopathol 2010; 33:67-87. [DOI: 10.1007/s00281-010-0204-1] [Citation(s) in RCA: 138] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2010] [Accepted: 03/18/2010] [Indexed: 01/12/2023]
|
16
|
Chromogranin A is an autoantigen in type 1 diabetes. Nat Immunol 2010; 11:225-31. [PMID: 20139986 DOI: 10.1038/ni.1844] [Citation(s) in RCA: 287] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2009] [Accepted: 01/15/2010] [Indexed: 12/16/2022]
Abstract
Autoreactive CD4(+) T cells are involved in the pathogenesis of many autoimmune diseases, but the antigens that stimulate their responses have been difficult to identify and in most cases are not well defined. In the nonobese diabetic (NOD) mouse model of type 1 diabetes, we have identified the peptide WE14 from chromogranin A (ChgA) as the antigen for highly diabetogenic CD4(+) T cell clones. Peptide truncation and extension analysis shows that WE14 bound to the NOD mouse major histocompatibility complex class II molecule I-A(g7) in an atypical manner, occupying only the carboxy-terminal half of the I-A(g7) peptide-binding groove. This finding extends the list of T cell antigens in type 1 diabetes and supports the idea that autoreactive T cells respond to unusually presented self peptides.
Collapse
|
17
|
Jordan MA, Poulton LD, Fletcher JM, Baxter AG. Allelic variation of Ets1 does not contribute to NK and NKT cell deficiencies in type 1 diabetes susceptible NOD mice. Rev Diabet Stud 2009; 6:104-16. [PMID: 19806240 DOI: 10.1900/rds.2009.6.104] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The NOD mouse is a well characterized model of type 1 diabetes that shares several of the characteristics of Ets1-deficient targeted mutant mice, viz: defects in TCR allelic exclusion, susceptibility to a lupus like disease characterized by IgM and IgG autoantibodies and immune complex-mediated glomerulonephritis, and deficiencies of NK and NKT cells. Here, we sought evidence for allelic variation of Ets1 in mice contributing to the NK and NKT cell phenotypes of the NOD strain. ETS1 expression in NK and NKT cells was reduced in NOD mice, compared to C57BL/6 mice. Although NKT cells numbers were significantly correlated with ETS1 expression in both strains, NKT cell numbers were not linked to the Ets1 gene in a first backcross from NOD to C57BL/6 mice. These results indicate that allelic variation of Ets1 did not contribute to variation in NKT cell numbers in these mice. It remains possible that a third factor not linked to the Ets1 locus controls both ETS1 expression and subsequently NK and NKT cell phenotypes.
Collapse
Affiliation(s)
- Margaret A Jordan
- Comparative Genomics Centre, Molecular Sciences Bldg 21, James Cook University, Townsville, QLD 4811, Australia
| | | | | | | |
Collapse
|
18
|
Jiang F, Yoshida T, Nakaki F, Terawaki S, Chikuma S, Kato Y, Okazaki IM, Honjo T, Okazaki T. Identification of QTLs that modify peripheral neuropathy in NOD.H2b-Pdcd1-/- mice. Int Immunol 2009; 21:499-509. [PMID: 19261693 DOI: 10.1093/intimm/dxp020] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The non-obese diabetic (NOD) mouse strain is prone to developing various autoimmune syndromes including type I diabetes mellitus (T1DM), sialadenitis, thyroiditis and pancreatitis. Although the genetic basis of T1DM has been extensively analyzed, genetic factors that modify the other autoimmune phenotypes are largely unknown. We have recently reported that NOD mice with anti-diabetogenic MHC haplotype (H-2(b)) and programmed cell death 1 (PD-1) deficiency (NOD.H2(b)-Pdcd1(-/-) mice) are protected from T1DM but develop various tissue-specific autoimmune diseases including peripheral neuropathy due to autoimmune neuritis, sialadenitis and gastritis. In the present study, we generated [(C57BL/6 x NOD.H2(b))(F1) x NOD-H2(b)](BC1)-Pdcd1(-/-) mice to screen non-MHC quantitative trait loci (QTLs) that modify autoimmune phenotypes other than T1DM. We identified seven QTLs for peripheral neuropathy and neuritis, one QTL for insulitis, four QTLs for gastritis, two QTLs for sialadenitis and seven QTLs for vasculitis throughout the genome and designated them as Annp loci for autoimmunity due to polymorphisms of non-MHC genes in NOD mice and PD-1 deficiency. Annp1, 5, 6 and 7 overlapped with reported loci for T1DM (Idd3, 9, 15 and 2, respectively), suggesting that these loci modify not only T1DM but also other autoimmune phenotypes. NOD allele was promotive at 9 of 14 Annp loci, while NOD allele was protective at the other loci. Half of Annp loci associated with a single phenotype, while the other seven loci associated with more than two phenotypes. These results indicate that NOD genetic background harbors various QTLs that modify autoimmune phenotypes either by organ-specific or by organ-non-specific manner.
Collapse
Affiliation(s)
- Fang Jiang
- Department of Immunology and Genomic Medicine, Kyoto University, Yoshida-Konoe, Sakyo-ku, Kyoto 606-8501, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Nguyen CQ, Cornelius JG, Cooper L, Neff J, Tao J, Lee BH, Peck AB. Identification of possible candidate genes regulating Sjögren's syndrome-associated autoimmunity: a potential role for TNFSF4 in autoimmune exocrinopathy. Arthritis Res Ther 2008; 10:R137. [PMID: 19032782 PMCID: PMC2656241 DOI: 10.1186/ar2560] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2008] [Revised: 10/27/2008] [Accepted: 11/25/2008] [Indexed: 12/11/2022] Open
Abstract
Introduction Sjögren syndrome (SjS) is a systemic autoimmune disease in which an immunological attack primarily against the salivary and lacrimal glands results in the loss of acinar cell tissue and function, leading to stomatitis sicca and keratoconjunctivitis sicca. In recent years, two genetic regions, one on chromosome 1 (designated autoimmune exocrinopathy 2 or Aec2) and the second on chromosome 3 (designated autoimmune exocrinopathy 1 or Aec1) derived from nonobese diabetic (NOD) mice, have been shown to be necessary and sufficient to replicate SjS-like disease in nonsusceptible C57BL/6 mice. Methods Starting with the SjS-susceptible C57BL/6-derived mouse, referred to as C57BL/6.NOD-Aec1Aec2, we generated a large set of recombinant inbred (RI) lines containing portions of Aec2 as a means of identifying more precisely the genetic elements of chromosome 1 responsible for disease development. Results Disease profiling of these RI lines has revealed that the SjS susceptibility genes of Aec2 lie within a region located at approximately 79 ± 5 cM distal to the centromere, as defined by microsatellite markers. This chromosomal region contains several sets of genes known to correlate with various immunopathological features of SjS as well as disease susceptibility genes for both type 1 diabetes and systemic lupus erythematosus in mice. One gene in particular, tumor necrosis factor (ligand) superfamily member 4 (or Ox40 ligand), encoding a product whose biological functions correlate with both physiological homeostasis and immune regulations, could be a potential candidate SjS susceptibility gene. Conclusions These new RI lines represent the first step not only in fine mapping SjS susceptibility loci but also in identifying potential candidate SjS susceptibility genes. Identification of possible candidate genes permits construction of models describing underlying molecular pathogenic mechanisms in this model of SjS and establishes a basis for construction of specific gene knockout mice.
Collapse
Affiliation(s)
- Cuong Q Nguyen
- Department of Oral Biology, University of Florida, Gainesville, FL 32610, USA.
| | | | | | | | | | | | | |
Collapse
|
20
|
Tang Q, Adams JY, Penaranda C, Melli K, Piaggio E, Sgouroudis E, Piccirillo CA, Salomon BL, Bluestone JA. Central role of defective interleukin-2 production in the triggering of islet autoimmune destruction. Immunity 2008; 28:687-97. [PMID: 18468463 DOI: 10.1016/j.immuni.2008.03.016] [Citation(s) in RCA: 602] [Impact Index Per Article: 35.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2007] [Revised: 03/15/2008] [Accepted: 03/19/2008] [Indexed: 01/07/2023]
Abstract
The dynamics of CD4(+) effector T cells (Teff cells) and CD4(+)Foxp3(+) regulatory T cells (Treg cells) during diabetes progression in nonobese diabetic mice was investigated to determine whether an imbalance of Treg cells and Teff cells contributes to the development of type 1 diabetes. Our results demonstrated a progressive decrease in the Treg cell:Teff cell ratio in inflamed islets but not in pancreatic lymph nodes. Intra-islet Treg cells expressed reduced amounts of CD25 and Bcl-2, suggesting that their decline was due to increased apoptosis. Additionally, administration of low-dose interleukin-2 (IL-2) promoted Treg cell survival and protected mice from developing diabetes. Together, these results suggest intra-islet Treg cell dysfunction secondary to defective IL-2 production is a root cause of the progressive breakdown of self-tolerance and the development of diabetes in nonobese diabetic mice.
Collapse
Affiliation(s)
- Qizhi Tang
- Department of Surgery, University of California, San Francisco, 513 Parnassus Avenue, Box 0540, San Francisco, CA 94143-0540, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Wilming LG, Gilbert JGR, Howe K, Trevanion S, Hubbard T, Harrow JL. The vertebrate genome annotation (Vega) database. Nucleic Acids Res 2007; 36:D753-60. [PMID: 18003653 PMCID: PMC2238886 DOI: 10.1093/nar/gkm987] [Citation(s) in RCA: 183] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The Vertebrate Genome Annotation (Vega) database (http://vega.sanger.ac.uk) was first made public in 2004 and has been designed to view manual annotation of human, mouse and zebrafish genomic sequences produced at the Wellcome Trust Sanger Institute. Since its initial release, the number of human annotated loci has more than doubled to close to 33 000 and now contains comprehensive annotation on 20 of the 24 human chromosomes, four whole mouse chromosomes and around 40% of the zebrafish Danio rerio genome. In addition, we offer manual annotation of a number of haplotype regions in mouse and human and regions of comparative interest in pig and dog that are unique to Vega.
Collapse
Affiliation(s)
- L G Wilming
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridgeshire, CB10 1SA, UK.
| | | | | | | | | | | |
Collapse
|
22
|
Motta V, Lejon K, Holmberg D. The NOD allele of the Idd5 locus on chromosome 1 mediates a non-cell-autonomous defect in negative selection of T cells. J Autoimmun 2007; 28:216-23. [PMID: 17449224 DOI: 10.1016/j.jaut.2007.03.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2007] [Revised: 03/01/2007] [Accepted: 03/02/2007] [Indexed: 01/11/2023]
Abstract
Recent data have suggested that non-obese diabetic (NOD) mice display a defect in negative thymic selection. Using mixed bone marrow chimeras, we demonstrate that the NOD allele of the diabetes susceptibility region 5 (Idd5) locus on chromosome 1, confers defective negative selection in response to endogenous superantigens (SAg) Mtv8 and Mtv9. We generated mixed bone marrow (BM) chimeras in which the donor cells of NOD and C3H or NOD.Idd5(b10) and C3H coexist and are similarly exposed to the Mtv8 and Mtv9 SAg. Under these conditions, SAg-mediated deletion of Vbeta11+ T cells is less efficient in chimeric mice reconstituted with NOD+C3H BM, compared with chimeras reconstituted with NOD.Idd5(b10)+C3H BM. Interestingly, the observed discrepancy was not T cell autonomous but was found to be mediated by a BM derived cellular subset, and under control of a gene(s) in the Idd5 region.
Collapse
Affiliation(s)
- Vinicius Motta
- Department of Medical Biosciences, Division of Medical and Clinical Genetics, Umeå University, S-901 87 Umeå, Sweden
| | | | | |
Collapse
|
23
|
Wetzel JD, Barton ES, Chappell JD, Baer GS, Mochow-Grundy M, Rodgers SE, Shyr Y, Powers AC, Thomas JW, Dermody TS. Reovirus delays diabetes onset but does not prevent insulitis in nonobese diabetic mice. J Virol 2006; 80:3078-82. [PMID: 16501117 PMCID: PMC1395416 DOI: 10.1128/jvi.80.6.3078-3082.2006] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2005] [Accepted: 12/19/2005] [Indexed: 11/20/2022] Open
Abstract
Mice infected with reovirus develop abnormalities in glucose homeostasis. Reovirus strain type 3 Abney (T3A) was capable of systemic infection of nonobese diabetic (NOD) mice, an experimental model of autoimmune diabetes. Reovirus antigen was detected in pancreatic islets of T3A-infected mice, and primary cultures of pancreatic islets from NOD mice supported T3A growth. Significantly fewer T3A-infected animals compared to uninfected controls developed diabetes. However, despite the alteration in diabetes penetrance, insulitis was evident in T3A-infected mice. These results suggest that viral infection of NOD mice alters autoimmune responses to beta-cell antigens and thereby delays development of diabetes.
Collapse
Affiliation(s)
- J Denise Wetzel
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Melanitou E. The autoimmune contrivance: genetics in the mouse model. Clin Immunol 2005; 117:195-206. [PMID: 16188504 DOI: 10.1016/j.clim.2005.07.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2005] [Revised: 07/15/2005] [Accepted: 07/19/2005] [Indexed: 01/21/2023]
Abstract
Autoimmunity and inheritance of complex characters behold an explosive interest in biology over the last 15 years. Research in the genetics of autoimmunity has been impelled by the isolation of genetic markers allowing tracing of heredity. The annotation and sequencing of the human and mouse genomes provide with the potential for further advancements, through the development of new technologies. This review aims to summarize advances made in the autoimmunity field, centered in type 1 diabetes in the NOD mouse model. It also aims to demonstrate that animal models, albeit some phenotypic and genetic dissimilarities with the human diseases, still remain the best way to move towards an understanding of the molecular mechanisms involved in autoimmunity. Assessing the current state of research in this field together with the increasing potential of novel biotechnology advancements, new insights to disease pathogenesis and discovery of molecular targets for intervention strategies are anticipated in the coming years.
Collapse
Affiliation(s)
- Evie Melanitou
- Immunopathology Unit, Molecular Medicine Department, Institut Pasteur, 28 rue du Dr Roux, 75015 Paris, France.
| |
Collapse
|
25
|
Affiliation(s)
- Clayton E Mathews
- Department of Pediatrics, The University of Pittsburgh, Pittsburgh, PA 15213, USA.
| |
Collapse
|
26
|
Szabó C. Roles of poly(ADP-ribose) polymerase activation in the pathogenesis of diabetes mellitus and its complications. Pharmacol Res 2005; 52:60-71. [PMID: 15911334 DOI: 10.1016/j.phrs.2005.02.015] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2004] [Accepted: 02/01/2005] [Indexed: 11/28/2022]
Abstract
Activation of poly(ADP-ribose) polymerase (PARP) plays a role in the pathogenesis of beta-cell necrosis that occurs in response to autoimmune disease associated with Type I diabetes. In addition, PARP activation also plays a role in the pathogenesis of endothelial injury that underlies the ethiology of various diabetic complications (vasculopathy, cardiomyopathy, retinopathy, neuropathy), which develop on the basis of chronically elevated circulating glucose levels in diabetes. Both during the pathogenesis of diabetes and during the pathogenesis of diabetic complications, free radical and oxidant production leads to DNA strand-breakage which activates the nuclear enzyme PARP and initiates an energy consuming, inefficient cellular metabolic cycle with transfer of the ADP-ribosyl moiety of NAD+ to protein acceptors. These processes lead to the functional impairment of the affected cells (beta-cells or vascular endothelial cells, respectively). PARP also promotes the activation of various pro-inflammatory signal transduction pathways. During the last two decades, a growing number of experimental studies demonstrated the beneficial effects PARP inhibition in various models of diabetes and diabetic complications. The current review provides an overview of the experimental evidence implicating PARP as a causative factor in the pathogenesis of diabetes and diabetic complications in vitro and in vivo.
Collapse
Affiliation(s)
- Csaba Szabó
- Inotek Pharmaceuticals Corporation, Suite 419 E, 100 Cummings Center, Beverly, MA 01915, USA.
| |
Collapse
|
27
|
Abstract
The nonobese diabetic mouse spontaneously develops an autoimmune, T-cell-mediated type 1 diabetes (T1D). Common and rare alleles both within a diabetogenic major histocompatibility complex (MHC) and multiple non-MHC genes combine to impair normal communication between the innate and acquired immune system, leading to loss of immune tolerance. An understanding of how variable collections of genes interact with each other and with environmental cues offers important insights as to the complexities of T1D inheritance in humans.
Collapse
Affiliation(s)
- Edward H Leiter
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA.
| |
Collapse
|
28
|
Koarada S, Wu Y, Yim YS, Wakeland EW, Ridgway WM. Nonobese diabetic CD4 lymphocytosis maps outside the MHC locus on chromosome 17. Immunogenetics 2004; 56:333-7. [PMID: 15309345 DOI: 10.1007/s00251-004-0702-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2004] [Accepted: 06/29/2004] [Indexed: 10/26/2022]
Abstract
Genetic control of homeostasis of peripheral CD4+ lymphocyte levels is incompletely understood. Recent genome scans have linked mouse peripheral CD4 levels to chromosome 17, with strongest linkage to the Ea region. Nonobese diabetic (NOD) mice demonstrate peripheral T-cell lymphocytosis, and previous studies also suggested that the MHC region might control this phenotype. Here we confirm that loci on Chr 17 control NOD peripheral CD4 lymphocytosis. An elevated NOD CD4:CD8 ratio maps to the same region, and we show it is due to increased numbers of CD4+ cells. However, using NOD MHC congenic mice, we demonstrate that the MHC region is excluded, and that NOD peripheral lymphocytosis is controlled by genetic intervals adjacent to the MHC region on Chr 17.
Collapse
Affiliation(s)
- Syuichi Koarada
- Division of Rheumatology and Immunology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | | | | | | | | |
Collapse
|
29
|
Inoue K, Ikegami H, Fujisawa T, Noso S, Nojima K, Babaya N, Itoi-Babaya M, Makimo S, Ogihara T. Allelic variation in class I K gene as candidate for a second component of MHC-linked susceptibility to type 1 diabetes in non-obese diabetic mice. Diabetologia 2004; 47:739-47. [PMID: 15298352 DOI: 10.1007/s00125-004-1370-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
AIMS/HYPOTHESIS Recent studies have revealed that MHC-linked susceptibility to Type 1 diabetes is determined by multiple components. In the non-obese diabetic (NOD) mouse, a second component (Idd16) has been mapped to a region adjacent to, but distinct from Idd1 in the class II region. In this study, we investigated the class I K gene as a candidate gene for Idd16. METHODS We determined the genomic sequences of the class I K gene as well as the reactivity of K molecules with monoclonal antibodies in the NOD mouse, the Cataract Shionogi (CTS) mouse, and the NOD.CTS-H-2 congenic strain, which possesses a resistance allele to Type 1 diabetes at the Idd16 on the NOD genetic background genes. RESULTS While the K sequence of the NOD mouse was identical to that of Kd type, ten nucleotide substitutions were identified in the CTS mouse compared with the NOD mouse. Of these, three were in exon 4, giving two amino acid substitutions, which were identical to those seen in KK type. These characteristics were retained in the NOD.CTS-H-2 congenic strain, which had a lower incidence and delayed onset of Type 1 diabetes owing to a resistance allele at Idd16. Lymphocytes from NOD.CTS-H2 congenic mice reacted with anti-Kd and anti-Kk monoclonal antibodies, reflecting the unique sequence of the K gene. The nucleotide sequence of the K gene in the non-obese non-diabetic (NON) mouse was also unique, consisting of a combination of Kk- and Kb-like sequences. CONCLUSIONS/INTERPRETATION These data suggest that H2-K is unique in CTS and NON mice, and that allelic variation of the class I K gene may be responsible for Idd16.
Collapse
Affiliation(s)
- K Inoue
- Department of Geriatric Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Ikegami H, Fujisawa T, Ogihara T. Mouse Models of Type 1 and Type 2 Diabetes Derived from the Same Closed Colony: Genetic Susceptibility Shared Between Two Types of Diabetes. ILAR J 2004; 45:268-77. [PMID: 15229374 DOI: 10.1093/ilar.45.3.268] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Except for rare subtypes of diabetes, both type 1 and type 2 diabetes are multifactorial diseases in which genetic factors consisting of multiple susceptibility genes and environmental factors contribute to the disease development. Due to complex interaction among multiple susceptibility genes and between genetic and environmental factors, genetic analysis of multifactorial diseases is difficult in humans. Inbred animal models, in which the genetic background is homogeneous and environmental factors can be controlled, are therefore valuable in genetic dissection of multifactorial diseases. We are fortunate to have excellent animal models for both type 1 and type 2 diabetes--the nonobese diabetic (NOD) mouse and the Nagoya-Shibata-Yasuda (NSY) mouse, respectively. Congenic mapping of susceptibility genes for type 1 diabetes in the NOD mouse has revealed that susceptibility initially mapped as a single locus often consists of multiple components on the same chromosome, indicating the importance of congenic mapping in defining genes responsible for polygenic diseases. The NSY mouse is an inbred animal model of type 2 diabetes established from Jcl:ICR, from which the NOD mouse was also derived. We have recently mapped three major loci contributing to type 2 diabetes in the NSY mouse. Interestingly, support intervals where type 2 diabetes susceptibility genes were mapped in the NSY mouse overlapped the regions where type 1 diabetes susceptibility genes have been mapped in the NOD mouse. Although additional evidence is needed, it may be possible that some of the genes predisposing to diabetes are derived from a common ancestor contained in the original closed colony, contributing to type 1 diabetes in the NOD mouse and type 2 diabetes in the NSY mouse. Such genes, if they exist, will provide valuable information on etiological pathways common to both forms of diabetes, for the establishment of effective methods for prediction, prevention, and intervention in both type 1 and type 2 diabetes.
Collapse
Affiliation(s)
- Hiroshi Ikegami
- Department of Geriatric Medicine, Osaka University, Graduate School of Medicine, Osaka, Japan
| | | | | |
Collapse
|
31
|
Ridgway WM. The non obese diabetic (NOD) mouse: a unique model for understanding the interaction between genetics and T cell responses. Rev Endocr Metab Disord 2003; 4:263-9. [PMID: 14501177 DOI: 10.1023/a:1025104429334] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- William M Ridgway
- Division of Immunology and Rheumatology, University of Pittsburgh School of Medicine, S725 Biomedical Science Tower, 3500 Terrace Street, PGH, PA 15261, USA.
| |
Collapse
|
32
|
Matsuki N, Stanic AK, Embers ME, Van Kaer L, Morel L, Joyce S. Genetic dissection of V alpha 14J alpha 18 natural T cell number and function in autoimmune-prone mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 170:5429-37. [PMID: 12759418 DOI: 10.4049/jimmunol.170.11.5429] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Nonobese diabetic (NOD) mice, a model for type I diabetes (TID), have reduced numbers of invariant V alpha 14J alpha 18 TCR alpha-chain-positive natural T (iNKT) cells that do not release IL-4 in response to in vivo activation through their Ag receptor. The deficit in iNKT cell number and function is implicated in immune dysregulation and the etiology of TID. Therefore, we reasoned that the genetic determinant(s) that controls iNKT cell number and function might lie within Idd (insulin-dependent diabetes susceptibility locus) regions, which are known to contain TID resistance or susceptibility genes. A systematic analysis of iNKT cell number and function in Idd congenic mice revealed that neither iNKT cell number nor their inability to rapidly secrete IL-4 in response to acute in vivo activation by Ag underlies the mechanism of protection from diabetes in Idd congenic mice. Moreover, the regulation of iNKT cell number and function appears to be under the control of several genes. The most notable of these map to the Idd4, Idd5, Idd9.1, and Idd13 regions of the mouse genome. Together these findings provide a clue to the genetic mechanism(s) underlying iNKT cell deficiency in NOD mice.
Collapse
MESH Headings
- Animals
- Antigens/administration & dosage
- Antigens/immunology
- Cytokines/biosynthesis
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/pathology
- Galactosylceramides/administration & dosage
- Galactosylceramides/immunology
- Genetic Markers/immunology
- Genetic Predisposition to Disease
- Immunity, Innate/genetics
- Injections, Intravenous
- Interferon-gamma/biosynthesis
- Interleukin-4/metabolism
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Killer Cells, Natural/pathology
- Lymphocyte Activation/genetics
- Lymphocyte Count
- Mice
- Mice, Congenic
- Mice, Inbred C57BL
- Mice, Inbred NOD
- Mice, Inbred NZB
- Mice, Mutant Strains
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- T-Lymphocyte Subsets/pathology
- Transcriptional Activation/immunology
- Up-Regulation/genetics
- Up-Regulation/immunology
Collapse
Affiliation(s)
- Naoto Matsuki
- Department of Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | | | | | | | | | | |
Collapse
|
33
|
Johansson ACM, Lindqvist AKB, Johannesson M, Holmdahl R. Genetic heterogeneity of autoimmune disorders in the nonobese diabetic mouse. Scand J Immunol 2003; 57:203-13. [PMID: 12641648 DOI: 10.1046/j.1365-3083.2003.01235.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The nonobese diabetic mouse is highly susceptible not only to diabetes but to several autoimmune diseases, and one might suspect that these are controlled by a shared set of genes. However, based on various gene-segregation experiments, it seems that only a few loci are shared and that each disorder is influenced also by a unique set of genes.
Collapse
Affiliation(s)
- A C M Johansson
- Section for Medical Inflammation Research, Department of Cell and Molecular Biology, University of Lund, Sweden.
| | | | | | | |
Collapse
|
34
|
Abstract
Given the importance of the NOD mouse as a model of type 1 diabetes, there is a surprising lack of published information on the overall composition of the thymic and peripheral T-cell compartments. In this study, we revisited some earlier reports of T-cell abnormalities in this strain and examined a number of additional parameters to provide a global view of T-cells in prediabetic NOD mice. In some cases, we concur with past conclusions, but in other important areas, we find that NOD mice closely resemble nonautoimmune strains. Specifically, and contrary to published reports, the thymocyte subset distribution, the rate and composition of thymic export, and the composition of the peripheral T-cell pool, including the proportion of CD25(+)CD4(+) T-cells, are essentially normal in prediabetic NOD mice. These factors are therefore unlikely to be involved in the loss of tolerance that leads to autoimmunity within this strain.
Collapse
Affiliation(s)
- Stuart P Berzins
- Section on Immunology and Immunogenetics, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02215, USA
| | | | | | | |
Collapse
|
35
|
Brimnes MK, Jensen T, Jørgensen TN, Michelsen BK, Troelsen J, Werdelin O. Low expression of insulin in the thymus of non-obese diabetic mice. J Autoimmun 2002; 19:203-13. [PMID: 12473241 DOI: 10.1006/jaut.2002.0616] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Insulin is a predominant autoantigen in IDDM in man and the NOD mouse. Failure of negative selection of diabetogenic T cells in thymus may be an important pre-disposing cause of the disease. To obtain insight into negative selection against such T-cell clones the thymic expression of insulin was studied in NOD and Balb/c mice by quantitative competitive RT-PCR. We detected RNA for insulin in the thymus of 3-week-old Balb/c mice as well as in NOD mice. However, the NOD mice expressed only half as many insulin transcripts as the Balb/c mice. Also, insulin protein was detected in the thymic medulla of both Balb/c and NOD mice. Furthermore, thymic RNA preparations were investigated for the presence of insulin transcription factors. None of the known pancreatic transcription factors for insulin; Pdx-1, Pax6 or Nkx6.1 were detectable in the thymus of Balb/c mice. These results support the idea that low insulin expression in the thymus may be a predisposing cause for development of diabetes in NOD mice analogous with what has been found in humans with the disease-disposing IDDM2 allele. Furthermore, our results suggest that insulin expression in the thymus may be regulated by different principles from those in the pancreas.
Collapse
Affiliation(s)
- Marie K Brimnes
- Institute for Medical Microbiology and Immunology, University of Copenhagen, Copenhagen, Denmark.
| | | | | | | | | | | |
Collapse
|
36
|
Koarada S, Wu Y, Olshansky G, Ridgway WM. Increased nonobese diabetic Th1:Th2 (IFN-gamma:IL-4) ratio is CD4+ T cell intrinsic and independent of APC genetic background. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 169:6580-7. [PMID: 12444170 DOI: 10.4049/jimmunol.169.11.6580] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Autoreactive CD4(+) T cells play a major role in the pathogenesis of autoimmune diabetes in nonobese diabetic (NOD) mice. We recently showed that the non-MHC genetic background controlled enhanced entry into the IFN-gamma pathway by NOD vs B6.G7 T cells. In this study, we demonstrate that increased IFN-gamma, decreased IL-4, and decreased IL-10 production in NOD T cells is CD4 T cell intrinsic. NOD CD4(+) T cells purified and stimulated with anti-CD3/anti-CD28 Abs generated greater IFN-gamma, less IL-4, and less IL-10 than B6.G7 CD4(+) T cells. The same results were obtained in purified NOD.H2(b) vs B6 CD4(+) T cells, demonstrating that the non-MHC NOD genetic background controlled the cytokine phenotype. Moreover, the increased IFN-gamma:IL-4 cytokine ratio was independent of the genetic background of APCs, since NOD CD4(+) T cells generated increased IFN-gamma and decreased IL-4 compared with B6.G7 CD4(+) T cells, regardless of whether they were stimulated with NOD or B6.G7 APCs. Cell cycle analysis showed that the cytokine differences were not due to cycle/proliferative differences between NOD and B6.G7, since stimulated CD4(+) T cells from both strains showed quantitatively identical entry into subsequent cell divisions (shown by CFSE staining), although NOD cells showed greater numbers of IFN-gamma-positive cells with each subsequent cell division. Moreover, 7-aminoactinomycin D and 5-bromo-2'-deoxyuridine analysis showed indistinguishable entry into G(0)/G(1), S, and G(2)/M phases of the cell cycle for both NOD and B6.G7 CD4(+) cells, with both strains generating IFN-gamma predominantly in the S phase. Therefore, the NOD cytokine effector phenotype is CD4(+) T cell intrinsic, genetically controlled, and independent of cell cycle machinery.
Collapse
Affiliation(s)
- Syuichi Koarada
- Division of Rheumatology and Immunology, Department of Medicine, University of Pittsburgh School of Medicine, PA 15261, USA
| | | | | | | |
Collapse
|
37
|
Baker FJ, Lee M, Chien YH, Davis MM. Restricted islet-cell reactive T cell repertoire of early pancreatic islet infiltrates in NOD mice. Proc Natl Acad Sci U S A 2002; 99:9374-9. [PMID: 12082183 PMCID: PMC123148 DOI: 10.1073/pnas.142284899] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The mechanisms responsible for initiating autoimmune diabetes remain obscure. Here, we describe a method for identifying both the alpha- and beta-chains of the T cell receptor (TCR) from individual pancreatic islet-infiltrating T cells at the earliest stages of disease in nonobese diabetic mice (NOD). Analysis of the TCR repertoire of these early islet infiltrates reveals enrichment for a small subset of TCR sequences. Reconstitution of these TCR in vitro demonstrates that these receptors confer reactivity to islet cells but not to the well characterized autoantigens, glutamic acid decarboxylase (GAD65) and insulin. Thus, autoimmune diabetes in NOD may be initiated by a limited number of antigens distinct from GAD65 and insulin.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Autoantigens
- Complementarity Determining Regions/genetics
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/immunology
- Genes, T-Cell Receptor alpha
- Genes, T-Cell Receptor beta
- Glutamate Decarboxylase/immunology
- In Vitro Techniques
- Insulin/immunology
- Islets of Langerhans/immunology
- Isoenzymes/immunology
- Mice
- Mice, Inbred NOD
- Molecular Sequence Data
- Polymerase Chain Reaction/methods
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- Sequence Homology, Amino Acid
- T-Lymphocytes/immunology
Collapse
Affiliation(s)
- Felix J Baker
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | | | | |
Collapse
|
38
|
Boulard O, Fluteau G, Eloy L, Damotte D, Bedossa P, Garchon HJ. Genetic analysis of autoimmune sialadenitis in nonobese diabetic mice: a major susceptibility region on chromosome 1. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 168:4192-201. [PMID: 11937580 DOI: 10.4049/jimmunol.168.8.4192] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The nonobese diabetic (NOD) mouse strain provides a good study model for Sjögren's syndrome (SS). The genetic control of SS was investigated in this model using different matings, including a (NOD x C57BL/6 (B6))F(2) cross, a (NOD x NZW)F(2) cross, and ((NOD x B6) x NOD) backcross. Multiple and different loci were detected depending on parent strain combination and sex. Despite significant complexity, two main features were prominent. First, the middle region of chromosome 1 (chr.1) was detected in all crosses. Its effect was most visible in the (NOD x B6)F(2) cross and dominated over that of other loci, including those mapping on chr.8, 9, 10, and 16; the effect of these minor loci was observed only in the absence of the NOD haplotype on chr.1. Most critically, the chr.1 region was sufficient to trigger an SS-like inflammatory infiltrate of salivary glands as shown by the study of a new C57BL/6 congenic strain carrying a restricted segment derived from NOD chr.1. Second, several chromosomal regions were previously associated with NOD autoimmune phenotypes, including Iddm (chr.1, 2, 3, 9, and 17, corresponding to Idd5, Idd13, Idd3, Idd2, and Idd1, respectively), accounting for the strong linkage previously reported between insulitis and sialitis, and autoantibody production (chr.10 and 16, corresponding to Bana2 and Bah2, respectively). Interestingly, only two loci were detected in the (NOD x NZW)F(2) cross, on chr.1 in females and on chr.7 in males, probably because of the latent autoimmune predisposition of the NZW strain. Altogether these findings reflect the complexity and heterogeneity of human SS.
Collapse
Affiliation(s)
- Olivier Boulard
- Institut National de la Santé et de la Recherche Médicale, Unité 25, Hôpital Necker-Enfants Malades, Paris, France
| | | | | | | | | | | |
Collapse
|
39
|
Abstract
In addition to developing a high incidence of type 1 diabetes caused by a specific autoimmune response against pancreatic beta cells in the islets of Langerhans, NOD mice also demonstrate spontaneous autoimmunity to other targets including the thymus, adrenal gland, salivary glands, thyroid, testis, nuclear components and red blood cells. Moreover, treatment of pre-diabetic NOD mice with an intravenous dose of heat killed Mycobacterium bovis (M. bovis; bacillus Calmette-Guèrin (BCG)) protects them from developing type 1 diabetes, but instead precipitates an autoimmune rheumatic disease similar to systemic lupus erythematosus (SLE), characterised by accelerated and increased incidence of haemolytic anaemia (HA), anti-nuclear autoantibody (ANA) production, exacerbation of sialadenitis, and the appearance of immune complex-mediated glomerulonephritis (GN). The reciprocal switching between the two phenotypes by a single environmental trigger (mycobacterial exposure) raised the possibility that genetic susceptibility for type 1 diabetes and SLE may be conferred by a single collection of genes in the NOD mouse. This review will focus on the genetic components predisposing NOD mice to SLE induced by BCG treatment and compare them to previously determined diabetes susceptibility genes in this strain and SLE susceptibility genes in the BXSB, MRL and the New Zealand mouse strains.
Collapse
Affiliation(s)
- P A Silveira
- Centenary Institute of Cancer Medicine and Cell Biology, Newtown NSW, Australia
| | | |
Collapse
|
40
|
Grattan M, Mi QS, Meagher C, Delovitch TL. Congenic mapping of the diabetogenic locus Idd4 to a 5.2-cM region of chromosome 11 in NOD mice: identification of two potential candidate subloci. Diabetes 2002; 51:215-23. [PMID: 11756344 DOI: 10.2337/diabetes.51.1.215] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Twenty diabetes susceptibility loci on 12 mouse chromosomes have been identified to control the development of type 1 diabetes at the level of either initiation of insulitis or progression from insulitis to overt diabetes or both. Previously, we demonstrated that the genetic control of T-cell proliferative unresponsiveness in nonobese diabetic (NOD) mice is linked to Idd4 on mouse chromosome 11. Here, we show by congenic mapping of three newly generated NOD.B6Idd4 diabetes-resistant mouse strains that Idd4 is limited to a 5.2-cM interval of chromosome 11. This B6-derived region expressed in NOD.B6Idd4A mice maps between the D11Nds1 (43.8 cM) and D11Mit38/D11Mit325 (49.0 cM) markers and dramatically reduces the development of both insulitis and type 1 diabetes. NOD.B6Idd4B and NOD.B6Idd4C mice, which carry a smaller B6-derived segment of chromosome 11 that spans <5.2 cM distal to D11Nds1, exhibit protection against type 1 diabetes with the restoration of T-cell proliferation. Our findings suggest that diabetes resistance conferred by Idd4 may be mediated by the Idd4.1 and Idd4.2 subloci. Idd4.1 is localized in the D11Nds1 interval that influences both diabetes and insulitis. Idd4.2 is localized within the D11Mit38/325 interval that mainly influences diabetes incidence and restores T-cell proliferative responsiveness. Three potential candidate genes, platelet activating factor acetylhydrolase Ib1, nitric oxide synthase-2, and CC chemokine genes, are localized in the 5.2-cM interval.
Collapse
Affiliation(s)
- Marsha Grattan
- Autoimmunity/Diabetes Group, the John P. Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
| | | | | | | |
Collapse
|
41
|
Koarada S, Wu Y, Ridgway WM. Increased entry into the IFN-gamma effector pathway by CD4+ T cells selected by I-Ag7 on a nonobese diabetic versus C57BL/6 genetic background. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:1693-702. [PMID: 11466393 DOI: 10.4049/jimmunol.167.3.1693] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
IFN-gamma-mediated Th1 effects play a major role in the pathogenesis of autoimmune diabetes in nonobese diabetic (NOD) mice. We analyzed functional responses of CD4(+) T cells from NOD and B6.G7 MHC congenic mice, which share the H2(g7) MHC region but differ in their non-MHC genetic background. T cells from each strain proliferated equally to panstimulation with T cell lectins as well as to stimulation with glutamic acid decarboxylase 524-543 (self) and hen egg lysozyme 11-23 (foreign) I-A(g7)-binding peptide epitopes. Despite comparable proliferative responses, NOD CD4(+) T cells had significantly increased IFN-gamma intracellular/extracellular protein and mRNA responses compared with B6.G7 T cells as measured by intracellular cytokine analysis, time resolved fluorometry, and RNase protection assays. The increased IFN-gamma production was not due to an increase in the amount of IFN-gamma produced per cell but to an increase in the number of NOD CD4(+) T cells entering the IFN-gamma-producing pathway. The increased IFN-gamma response in NOD mice was not due to increased numbers of activated precursors as measured by activation/memory markers. B6.G7 lymphoid cells demonstrated an absolute decrease in IFN-gamma mRNA, an increase in IL-4 mRNA production, and a significantly decreased IFN-gamma:IL-4 mRNA transcript ratio compared with NOD cells. CD4(+) T cells from C57BL6 mice also showed significantly decreased IFN-gamma production compared with CD4(+) T cells from NOD.H2(b) MHC-congenic mice (which have an H2(b) MHC region introgressed onto an NOD non-MHC background). Therefore, the NOD non-MHC background predisposes to a quantitatively increased IFN-gamma response, independent of MHC class II-mediated T cell repertoire selection, even when compared with a prototypical Th1 strain.
Collapse
Affiliation(s)
- S Koarada
- Division of Rheumatology and Immunology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | | | | |
Collapse
|
42
|
Johansson AC, Sundler M, Kjellén P, Johannesson M, Cook A, Lindqvist AK, Nakken B, Bolstad AI, Jonsson R, Alarcón-Riquelme M, Holmdahl R. Genetic control of collagen-induced arthritis in a cross with NOD and C57BL/10 mice is dependent on gene regions encoding complement factor 5 and FcgammaRIIb and is not associated with loci controlling diabetes. Eur J Immunol 2001; 31:1847-56. [PMID: 11433381 DOI: 10.1002/1521-4141(200106)31:6<1847::aid-immu1847>3.0.co;2-f] [Citation(s) in RCA: 69] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The nonobese diabetic (NOD) mouse spontaneously develops autoimmune-mediated diseases such as diabetes and Sjögren's syndrome. To investigate whether NOD genes also promote autoimmune-mediated arthritis we established a NOD strain with an MHC class II fragment containing the A(q) class II gene predisposing for collagen induced arthritis (NOD.Q). However, this mouse was resistant to arthritis in contrast to other A(q) expressing strains such as B10.Q and DBA/1. To determine the major resistance factor/s, a genetic analysis was performed. (NOD.Q x B10.Q)F1 mice were resistant, whereas 27% of the (NOD.Q x B10.Q)F2 mice developed severe arthritis. Genetic mapping of 353 F2 mice revealed two loci associated with arthritis. One locus was found on chromosome 2 (LOD score 9.8), at the location of the complement factor 5 (C5) gene. The susceptibility allele was from B10.Q, which contains a productive C5 encoding gene in contrast to NOD.Q. The other significant locus was found on chromosome 1 (LOD score 5.6) close to the Fc-gamma receptor IIb gene, where NOD carried the susceptible allele. An interaction between the two loci was observed, indicating that they operate on the same or on interacting pathways. The genetic control of arthritis is unique in comparison to diabetes, since none of these loci have been identified in analysis of diabetes susceptibility.
Collapse
MESH Headings
- Animals
- Antigens, CD/genetics
- Arthritis, Rheumatoid/genetics
- Collagen
- Complement C5/genetics
- Crosses, Genetic
- Diabetes Mellitus, Type 1/genetics
- Disease Models, Animal
- Female
- H-2 Antigens
- Immunity, Innate/immunology
- Male
- Mice
- Mice, Inbred C3H
- Mice, Inbred C57BL
- Mice, Inbred NOD
- Receptors, IgG/genetics
Collapse
Affiliation(s)
- A C Johansson
- Section for Medical Inflammation Research, Department of Cell and Molecular Biology, University of Lund, Lund, Sweden.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Rogner UC, Boitard C, Morin J, Melanitou E, Avner P. Three loci on mouse chromosome 6 influence onset and final incidence of type I diabetes in NOD.C3H congenic strains. Genomics 2001; 74:163-71. [PMID: 11386752 DOI: 10.1006/geno.2001.6508] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The development of insulin-dependent diabetes mellitus in both human and mouse is dependent on the interaction between genetic and environmental factors. The analysis of newly created NOD.C3H congenic strains for spontaneous and cyclophosphamide-induced diabetes has allowed the definition of three controlling genetic loci on mouse chromosome 6. A NOD-derived susceptibility allele at the Idd6 locus strongly influences the onset of diabetes in spontaneous diabetes. A NOD-derived resistance allele at the Idd19 locus affects the final diabetes incidence observed in both models, while a novel locus, provisionally termed Idd20, appears to control Idd19 in an epistatic manner. Decreased diabetes incidence is observed in CY-induced diabetes when Idd20 is homozygous for the C3H allele, while heterozygosity is associated with an increase in diabetes incidence. The Idd20, Idd19, and Idd6 candidate regions fall respectively within genetically defined intervals of 4, 7, and 4.5 cM on mouse chromosome 6. From our YAC contig, Idd6 would appear to localize within a ca. 1.5-Mb region on distal chromosome 6.
Collapse
Affiliation(s)
- U C Rogner
- Génétique Moléculaire Murine CNRS URA 1947, Institut Pasteur, 25 rue du Docteur Roux, Paris Cedex 15, 75724, France.
| | | | | | | | | |
Collapse
|
44
|
Jordan MA, Silveira PA, Shepherd DP, Chu C, Kinder SJ, Chen J, Palmisano LJ, Poulton LD, Baxter AG. Linkage analysis of systemic lupus erythematosus induced in diabetes-prone nonobese diabetic mice by Mycobacterium bovis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 165:1673-84. [PMID: 10903779 DOI: 10.4049/jimmunol.165.3.1673] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Systemic lupus erythematosus induced by Mycobacterium bovis in diabetes-prone nonobese diabetic mice was mapped in a backcross to the BALB/c strain. The subphenotypes-hemolytic anemia, antinuclear autoantibodies, and glomerular immune complex deposition-did not cosegregate, and linkage analysis for each trait was performed independently. Hemolytic anemia mapped to two loci: Bah1 at the MHC on chromosome 17 and Bah2 on distal chromosome 16. Antinuclear autoantibodies mapped to three loci: Bana1 at the MHC on chromosome 17, Bana2 on chromosome 10, and Bana3 on distal chromosome 1. Glomerular immune complex deposition did not show significant linkage to any genomic region. Mapping of autoantibodies (Coombs' or antinuclear autoantibodies) identified two loci: Babs1 at the MHC and Babs2 on distal chromosome 1. It has previously been reported that genes conferring susceptibility to different autoimmune diseases map nonrandomly to defined regions of the genome. One possible explanation for this clustering is that some alleles at loci within these regions confer susceptibility to multiple autoimmune diseases-the "common gene" hypothesis. With the exception of the H2, this study failed to provide direct support for the common gene hypothesis, because the loci identified as conferring susceptibility to systemic lupus erythematosus did not colocalize with those previously implicated in diabetes. However, three of the four regions identified had been previously implicated in other autoimmune diseases.
Collapse
MESH Headings
- Anemia, Hemolytic/genetics
- Anemia, Hemolytic/immunology
- Animals
- Antibodies, Antinuclear/blood
- Antibodies, Antinuclear/genetics
- Antigen-Antibody Complex/metabolism
- Autoantibodies/genetics
- Complement C3c/metabolism
- Crosses, Genetic
- Diabetes Mellitus, Type 1/blood
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/immunology
- Female
- Genetic Linkage/immunology
- Genetic Markers
- Genotype
- Hematocrit
- Kidney Glomerulus/immunology
- Kidney Glomerulus/metabolism
- Lupus Erythematosus, Systemic/blood
- Lupus Erythematosus, Systemic/genetics
- Lupus Erythematosus, Systemic/immunology
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Inbred NOD
- Microsatellite Repeats/immunology
- Mycobacterium bovis/immunology
- Phenotype
Collapse
Affiliation(s)
- M A Jordan
- Centenary Institute of Cancer Medicine and Cell Biology, Newtown, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
Autoimmune diabetes is a polygenic disease process in man and rodents. To identify and characterize genes involved in the pathogenesis of diabetes in nonobese diabetic (NOD) mice, we initiated a repetitive backcross of diabetes-resistant C57L/J mice onto the NOD strain. This breeding scheme was based on the premise that selection for the trait of disease resistance among genetically mixed mice could be used to maintain transmission of nonpermissive alleles from the diabetes-resistant strain at critical diabetes susceptibility loci. Each of the three recombinant congenic mouse lines derived by this strategy retains a unique constellation of C57L/J-derived DNA segments. Consistent with the involvement of different genetic loci, the pancreatic histology of disease-resistant mice differs from that in NOD mice in a line-specific manner. Functional studies using these lines demonstrate that pathogenesis of autoimmune diabetes is a multistep process which can be blocked at a minimum of three critical, genetically determined points.
Collapse
Affiliation(s)
- M McDuffie
- Diabetes Research Center, University of Virginia Health Sciences Center, Charlottesville, Virginia 22908, USA
| |
Collapse
|
46
|
Dahlén E, Hedlund G, Dawe K. Low CD86 expression in the nonobese diabetic mouse results in the impairment of both T cell activation and CTLA-4 up-regulation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 164:2444-56. [PMID: 10679081 DOI: 10.4049/jimmunol.164.5.2444] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The nonobese diabetic (NOD) mouse spontaneously develops autoimmune insulin-dependent diabetes mellitus and serves as a model for human type I diabetes. NOD spleen cells proliferate to a lesser extent than those from C57BL/6 and BALB/c mice in response to anti-CD3. To investigate the cause of this reduced T cell proliferation, costimulatory molecule expression was investigated. It was found that NOD macrophages, dendritic cells, and T cells, but not B cells, expressed lower basal levels of CD86, but not CD80, CD28, or CD40, compared with C57BL/6 and BALB/c. This low CD86 expression was not dependent on the MHC haplotype or on diabetes development since the NOD-related, diabetes-free mouse strains NON (H-2nb1) and NOR (H-2g7) exhibited similar low levels of CD86 expression and proliferation. Furthermore, following activation, the relative up-regulation of CTLA-4, as compared with CD28, was more pronounced on C57BL/6 and BALB/c T cells as shown by an increased CTLA-4/CD28 ratio. This activation-induced increase in the CTLA-4/CD28 ratio was markedly reduced on NOD T cells compared with the other two strains. The low CD86 expression in NOD mice may account for the reduced increase in both proliferation and the CTLA-4/CD28 ratio, since reducing CD86 expression in C57BL/6 and BALB/c cultures to NOD levels significantly reduces the proliferation and the CTLA-4/CD28 ratio. Therefore, we propose that a low level of CD86 expression in the NOD mouse contributes to a defective regulation of autoreactive T cells by preventing the full activation of T cells and therefore the up-regulation of CTLA-4.
Collapse
MESH Headings
- Abatacept
- Animals
- Antibodies, Monoclonal/pharmacology
- Antigen-Presenting Cells/immunology
- Antigens, CD/biosynthesis
- Antigens, CD/pharmacology
- Antigens, CD/physiology
- Antigens, Differentiation/biosynthesis
- B7-2 Antigen
- CD28 Antigens/biosynthesis
- CD3 Complex/immunology
- CTLA-4 Antigen
- Cells, Cultured
- Diabetes Mellitus, Type 1/immunology
- Female
- Immunoconjugates
- Lymphocyte Activation/immunology
- Membrane Glycoproteins/biosynthesis
- Membrane Glycoproteins/pharmacology
- Membrane Glycoproteins/physiology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Inbred NOD
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Up-Regulation/immunology
Collapse
Affiliation(s)
- E Dahlén
- Active Biotech Research AB, Lund, Sweden.
| | | | | |
Collapse
|
47
|
Rietz C, Pilström B, Brenden N, Böhme J. Minute defects in the expression of MHC E molecules lead to impaired protection from autoimmunity in NOD mice. Scand J Immunol 1999; 50:405-10. [PMID: 10520181 DOI: 10.1046/j.1365-3083.1999.00613.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The E complex of the major histocompatibility complex (MHC) can prevent the spontaneous development of diabetes in nonobese diabetic (NOD) mice transgenic for the Ea gene. None of three promoter-mutated Ea constructs with Ea expression directed to different subsets of immunocompetent cells exerts full protection in NOD mice. The promoter-mutated constructs are all capable of mediating intrathymic elimination of I-E-restricted T cells. Thus, thymic negative selection is not responsible for the protective effect but a more complex effect is likely. Here we show that combinations of two or three different mutated Ea constructs do not protect against intra-islet insulitis either. We also show that spleen cells from protected animals are sufficient to protect NOD mice in adoptive transfer experiments. The only detectable expression defects in splenic cells or cells influencing the repertoire of splenic cells are in the B-cell compartment. Furthermore, in three construct combinations, the differences to wild-type expression are extremely small. Thus, we conclude that even minute disturbances of the E expression pattern might reduce the protection of NOD mice from insulitis.
Collapse
Affiliation(s)
- C Rietz
- Department of Immunology, The Wenner-Gren Institute, Stockholm University, S-106 91 Stockholm, Sweden
| | | | | | | |
Collapse
|
48
|
Hattori M, Yamato E, Itoh N, Senpuku H, Fujisawa T, Yoshino M, Fukuda M, Matsumoto E, Toyonaga T, Nakagawa I, Petruzzelli M, McMurray A, Weiner H, Sagai T, Moriwaki K, Shiroishi T, Maron R, Lund T. Cutting Edge: Homologous Recombination of the MHC Class I K Region Defines New MHC-Linked Diabetogenic Susceptibility Gene(s) in Nonobese Diabetic Mice. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.163.4.1721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Abstract
To localize the MHC-linked diabetogenic genes in the nonobese diabetic (NOD) mouse, a recombinational hotspot from the B10.A(R209) mouse was introduced to the region between the MHC class I K and class II A of the NOD mouse with the recombinational site centromeric to the Lmp2/Tap1 complex by breeding the two strains. Replacement of the NOD region centromeric to the recombinational site with the same region in R209 mice prevented the development of diabetes (from 71 to 3%) and insulitis (from 61 to 15%) in the N7 intra-MHC recombinant NOD mice. Similarly, the replacement of the NOD class II A, E and class I D region with the same region in R209 mice prevented the diseases (diabetes, from 71 to 0%; insulitis, from 61 to 3%). In addition to the MHC class II genes, there are at least two MHC-linked diabetogenic genes in the region centromeric to Lmp2.
Collapse
Affiliation(s)
- Masakazu Hattori
- *Section on Immunology and Immunogenetics, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215
| | - Eiji Yamato
- *Section on Immunology and Immunogenetics, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215
| | - Naoto Itoh
- *Section on Immunology and Immunogenetics, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215
| | - Hidenobu Senpuku
- *Section on Immunology and Immunogenetics, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215
| | - Tomomi Fujisawa
- *Section on Immunology and Immunogenetics, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215
| | | | - Masahiro Fukuda
- *Section on Immunology and Immunogenetics, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215
| | - Eisaku Matsumoto
- *Section on Immunology and Immunogenetics, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215
| | - Tetsushi Toyonaga
- *Section on Immunology and Immunogenetics, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215
| | - Ichiro Nakagawa
- *Section on Immunology and Immunogenetics, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215
| | - Maria Petruzzelli
- *Section on Immunology and Immunogenetics, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215
| | - Armand McMurray
- ‡Whitehead Institute for Biomedical Research, Cambridge MA 02139
| | - Howard Weiner
- §Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115; and
| | | | | | | | - Ruth Maron
- §Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115; and
| | - Torben Lund
- ¶University College London, London, United Kingdom
| |
Collapse
|
49
|
Noorchashm H, Lieu YK, Noorchashm N, Rostami SY, Greeley SAS, Schlachterman A, Song HK, Noto LE, Jevnikar AM, Barker CF, Naji A. I-Ag7-Mediated Antigen Presentation by B Lymphocytes Is Critical in Overcoming a Checkpoint in T Cell Tolerance to Islet β Cells of Nonobese Diabetic Mice. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.163.2.743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
B cell-deficient nonobese diabetic (NOD) mice are protected from the development of spontaneous autoimmune diabetes, suggesting a requisite role for Ag presentation by B lymphocytes for the activation of a diabetogenic T cell repertoire. This study specifically examines the importance of B cell-mediated MHC class II Ag presentation as a regulator of peripheral T cell tolerance to islet β cells. We describe the construction of NOD mice with an I-Ag7 deficiency confined to the B cell compartment. Analysis of these mice, termed NOD BCIID, revealed the presence of functionally competent non-B cell APCs (macrophages/dendritic cells) with normal I-Ag7 expression and capable of activating Ag-reactive T cells. In addition, the secondary lymphoid organs of these mice harbored phenotypically normal CD4+ and CD8+ T cell compartments. Interestingly, whereas control NOD mice harboring I-Ag7-sufficient B cells developed diabetes spontaneously, NOD BCIID mice were resistant to the development of autoimmune diabetes. Despite their diabetes resistance, histologic examination of pancreata from NOD BCIID mice revealed foci of noninvasive peri-insulitis that could be intentionally converted into a destructive process upon treatment with cyclophosphamide. We conclude that I-Ag7-mediated Ag presentation by B cells serves to overcome a checkpoint in T cell tolerance to islet β cells after their initial targeting has occurred. Overall, this work indicates that the full expression of the autoimmune potential of anti-islet T cells in NOD mice is intimately regulated by B cell-mediated MHC class II Ag presentation.
Collapse
Affiliation(s)
- Hooman Noorchashm
- *Department of Surgery, University of Pennsylvania Medical Center, Philadelphia, PA 19104; and
| | - Yen K. Lieu
- *Department of Surgery, University of Pennsylvania Medical Center, Philadelphia, PA 19104; and
| | - Negin Noorchashm
- *Department of Surgery, University of Pennsylvania Medical Center, Philadelphia, PA 19104; and
| | - Susan Y. Rostami
- *Department of Surgery, University of Pennsylvania Medical Center, Philadelphia, PA 19104; and
| | - Siri Atma S. Greeley
- *Department of Surgery, University of Pennsylvania Medical Center, Philadelphia, PA 19104; and
| | - Alexander Schlachterman
- *Department of Surgery, University of Pennsylvania Medical Center, Philadelphia, PA 19104; and
| | - Howard K. Song
- *Department of Surgery, University of Pennsylvania Medical Center, Philadelphia, PA 19104; and
| | - Lauren E. Noto
- *Department of Surgery, University of Pennsylvania Medical Center, Philadelphia, PA 19104; and
| | - Anthony M. Jevnikar
- †Division of Nephrology, London Health Sciences Centre, London, Ontario, Canada
| | - Clyde F. Barker
- *Department of Surgery, University of Pennsylvania Medical Center, Philadelphia, PA 19104; and
| | - Ali Naji
- *Department of Surgery, University of Pennsylvania Medical Center, Philadelphia, PA 19104; and
| |
Collapse
|
50
|
Affiliation(s)
- W M Ridgway
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | | |
Collapse
|