51
|
Chen YG, Silveira PA, Osborne MA, Chapman HD, Serreze DV. Cellular expression requirements for inhibition of type 1 diabetes by a dominantly protective major histocompatibility complex haplotype. Diabetes 2007; 56:424-30. [PMID: 17259387 DOI: 10.2337/db06-1303] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The H2(g7) (K(d), A(g7), E(null), and D(b)) major histocompatibility complex (MHC) is the primary genetic contributor to type 1 diabetes in NOD mice. NOD stocks congenically expressing other MHC haplotypes such as H2(nb1) (K(b), A(nb1), E(k), and D(b)) in a heterozygous state are type 1 diabetes resistant. Hematopoietically derived antigen-presenting cells (APCs) expressing H2(nb1) MHC molecules delete or inactivate autoreactive diabetogenic T-cells. Thus, provided a relatively benign preconditioning protocol is ultimately developed, hematopoietic chimerization by APCs expressing dominantly protective MHC molecules could conceivably provide a means for type 1 diabetes prevention in humans. Before hematopoietic chimerization can be considered for type 1 diabetes prevention, it must be determined what subtype(s) of APCs (B-cells, macrophages, and/or dendritic cells) expressing protective MHC molecules most efficiently inhibit disease, as well as the engraftment level they must achieve to accomplish this. These issues were addressed through analyses of NOD background bone marrow chimeras in which H2(nb1) molecules were selectively expressed on variable proportions of different APC subtypes. While a modest B-cell effect was observed, the strongest type 1 diabetes protection resulted from at least 50% of dendritic cells and macrophages expressing H2(nb1) molecules. At this engraftment level, H2(nb1)-expressing dendritic cells and macrophages mediated virtually complete deletion of a highly pathogenic CD8 T-cell population.
Collapse
|
52
|
Mayo S, Kohler W, Kumar V, Quinn A. Insulin-dependent diabetes loci Idd5 and Idd9 increase sensitivity to experimental autoimmune encephalomyelitis. Clin Immunol 2006; 118:219-28. [PMID: 16301001 DOI: 10.1016/j.clim.2005.10.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2005] [Revised: 09/15/2005] [Accepted: 10/06/2005] [Indexed: 12/21/2022]
Abstract
The spontaneous development of autoimmune diabetes in NOD mice suggests that they are unable to establish and maintain immunologic self-tolerance. Congenic NOD mice expressing B10-derived alleles are protected from pancreatic beta cell destruction and autoimmune diabetes. To determine if the B10 alleles in loci Idd5 and Idd9 could influence susceptibility to autoimmunity in other organs, we compared MOG35-55-induced EAE in NOD mice to that of diabetes-resistant NOD.B10.Idd5 and NOD.B10.Idd9 mice. Surprisingly, the severity and chronicity of EAE were enhanced in the diabetes-resistant congenic mice. Our findings indicate that some alleles may influence susceptibility to immune-mediated damage in an organ or tissue-specific fashion, and highlight the necessity of disease-specific investigations.
Collapse
Affiliation(s)
- Stella Mayo
- Department of Biological Sciences, University of Toledo, 2801 W. Bancroft, Toledo, OH 43606, USA
| | | | | | | |
Collapse
|
53
|
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: 10] [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
|
54
|
Jiang W, Anderson MS, Bronson R, Mathis D, Benoist C. Modifier loci condition autoimmunity provoked by Aire deficiency. J Exp Med 2005; 202:805-15. [PMID: 16172259 PMCID: PMC2212943 DOI: 10.1084/jem.20050693] [Citation(s) in RCA: 186] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2005] [Accepted: 08/10/2005] [Indexed: 01/07/2023] Open
Abstract
Loss of function mutations in the autoimmune regulator (Aire) gene in autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy patients and mutant mice lead to autoimmune manifestations that segregate as a monogenic trait, but with wide variation in the spectrum of organs targeted. To investigate the cause of this variability, the Aire knockout mutation was backcrossed to mice of diverse genetic backgrounds. The background loci strongly influenced the pattern of organs that were targeted (stomach, eye, pancreas, liver, ovary, thyroid, and salivary gland) and the severity of the targeting (particularly strong on the nonobese diabetic background, but very mild on the C57BL/6 background). Autoantibodies mimicked the disease pattern, with oligoclonal reactivity to a few antigens that varied between Aire-deficient strains. Congenic analysis and a whole genome scan showed that autoimmunity to each organ had a distinctive pattern of genetic control and identified several regions that controlled the pattern of targeting, including the major histocompatibility complex and regions of Chr1 and Chr3 previously identified in controlling type 1 diabetes.
Collapse
Affiliation(s)
- Wenyu Jiang
- Section on Immunology and Immunogenetics, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02215, USA
| | | | | | | | | |
Collapse
|
55
|
Abstract
Transforming growth factor-beta1 (TGF-beta1) is released during the storage of blood components, particularly platelet concentrates, and transfusion recipients are exposed to high levels of TGF-beta1. Because TGF-beta1 is one of the most potent immunosuppressive cytokines known, understanding the immunobiologic functions of TGF-beta1 may be relevant for understanding the immunobiologic effects of transfusion. Our laboratory studies the biologic effects of TGF-beta1 in the immune system. Mice deficient in TGF-beta1 spontaneously develop autoimmunity, confirming the important role of this cytokinean an immune regulator. A few years ago, my laboratory made the observation that genetic background strongly affects the phenotype of TGF-beta1-/- mice. TGF-beta1-/- mice on the BALB/c background rapidly develop an aggressive T-cell-mediated hepatitis, whereas TGF-beta1-/- mice on the 129/CF-1 background do not. In this review, I summarize findings published or in press from our laboratory on disease pathogenesis in TGF-beta1-/- mice and then discuss some of the exciting (as-yet-unpublished) directions our laboratory is currently taking.
Collapse
Affiliation(s)
- James D Gorham
- Department of Pathology, Dartmouth Medical School, Lebanon, New Hampshire 03756, USA.
| |
Collapse
|
56
|
Abstract
Autoimmunity is a complex process that likely results from the summation of multiple defective tolerance mechanisms. The NOD mouse strain is an excellent model of autoimmune disease and an important tool for dissecting tolerance mechanisms. The strength of this mouse strain is that it develops spontaneous autoimmune diabetes, which shares many similarities to autoimmune or type 1a diabetes (T1D) in human subjects, including the presence of pancreas-specific autoantibodies, autoreactive CD4+ and CD8+ T cells, and genetic linkage to disease syntenic to that found in humans. During the past ten years, investigators have used a wide variety of tools to study these mice, including immunological reagents and transgenic and knockout strains; these tools have tremendously enhanced the study of the fundamental disease mechanisms. In addition, investigators have recently developed a number of therapeutic interventions in this animal model that have now been translated into human therapies. In this review, we summarize many of the important features of disease development and progression in the NOD strain, emphasizing the role of central and peripheral tolerance mechanisms that affect diabetes in these mice. The information gained from this highly relevant model of human disease will lead to potential therapies that may alter the development of the disease and its progression in patients with T1D.
Collapse
Affiliation(s)
- Mark S Anderson
- Diabetes Center, University of California, San Francisco, California 94143, USA.
| | | |
Collapse
|
57
|
Beilhack GF, Landa RR, Masek MA, Shizuru JA. Prevention of type 1 diabetes with major histocompatibility complex-compatible and nonmarrow ablative hematopoietic stem cell transplants. Diabetes 2005; 54:1770-9. [PMID: 15919799 DOI: 10.2337/diabetes.54.6.1770] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Progression to hyperglycemia in young nonobese diabetic (NOD) mice is blocked by the transplantation of hematopoietic cells mismatched at the major histocompatibility complex (MHC). Because the NOD MHC class II allele, I-A(g7), is the primary disease susceptibility gene, it is logical to conclude that MHC-mismatched hematopoietic grafts prevent diabetes by replacement of this susceptibility allele on critical hematolymphoid populations. In this report, transplantation of MHC-matched purified hematopoietic stem cells (HSCs) pre-vented diabetes development in NOD mice, demonstrating that alleles of non-MHC background genes expressed on hematopoietic cells are sufficient to disrupt the autoaggressive process. Nonmarrow ablative conditioning was 100% protective, further showing that elimination of NOD hematopoiesis, including T-cells, was not required for the graft to block diabetes pathogenesis. The current standard clinical practice of hematopoietic cell transplantation uses donor/recipient pairs that are matched at the MHC. In our view, the principles established here using an MHC-matched engineered hematopoietic graft in conjunction with nonmarrow ablative conditioning to successfully block autoimmune diabetes sufficiently reduces the morbidity of the allogeneic transplantation procedure such that a similar approach can be translated to the treatment of human autoimmune disorders.
Collapse
Affiliation(s)
- Georg F Beilhack
- Division of Blood and Marrow Transplantation, Stanford University Medical Center, Stanford, CA 94305-5623, USA
| | | | | | | |
Collapse
|
58
|
Ivakine EA, Fox CJ, Paterson AD, Mortin-Toth SM, Canty A, Walton DS, Aleksa K, Ito S, Danska JS. Sex-Specific Effect of Insulin-Dependent Diabetes 4 on Regulation of Diabetes Pathogenesis in the Nonobese Diabetic Mouse. THE JOURNAL OF IMMUNOLOGY 2005; 174:7129-40. [PMID: 15905556 DOI: 10.4049/jimmunol.174.11.7129] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Many human autoimmune diseases are more frequent in females than males, and their clinical severity is affected by sex hormone levels. A strong female bias is also observed in the NOD mouse model of type I diabetes (T1D). In both NOD mice and humans, T1D displays complex polygenic inheritance and T cell-mediated autoimmune pathogenesis. The identities of many of the insulin-dependent diabetes (Idd) loci, their influence on specific stages of autoimmune pathogenesis, and sex-specific effects of Idd loci in the NOD model are not well understood. To address these questions, we analyzed cyclophosphamide-accelerated T1D (CY-T1D) that causes disease with high and similar frequencies in male and female NOD mice, but not in diabetes-resistant animals, including the nonobese diabetes-resistant (NOR) strain. In this study we show by genetic linkage analysis of (NOD x NOR) x NOD backcross mice that progression to severe islet inflammation after CY treatment was controlled by the Idd4 and Idd9 loci. Congenic strains on both the NOD and NOR backgrounds confirmed the roles of Idd4 and Idd9 in CY-T1D susceptibility and revealed the contribution of a third locus, Idd5. Importantly, we show that the three loci acted at distinct stages of islet inflammation and disease progression. Among these three loci, Idd4 alleles alone displayed striking sex-specific behavior in CY-accelerated disease. Additional studies will be required to address the question of whether a sex-specific effect of Idd4, observed in this study, is also present in the spontaneous model of the disease with striking female bias.
Collapse
Affiliation(s)
- Evgueni A Ivakine
- Program in Developmental Biology, Hospital for Sick Children, Toronto, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
59
|
Lin JT, Kitzmiller TJ, Cates JMM, Gorham JD. MHC-independent genetic regulation of liver damage in a mouse model of autoimmune hepatocellular injury. J Transl Med 2005; 85:550-61. [PMID: 15696185 DOI: 10.1038/labinvest.3700246] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Autoimmune hepatitis (AIH) is mediated by a T-cell attack upon liver parenchyma. Susceptibility to the development of AIH is genetically determined. While particular MHC haplotypes are known risk factors, it has been widely speculated that autoimmune liver damage can be regulated by additional genetic loci unlinked to MHC. However, evidence for the existence of such loci in humans is scant. We examined the contribution of the MHC in a murine model of autoimmune hepatocellular injury. BALB/c mice lacking the immunoregulatory cytokine transforming growth factor-beta1 (TGF-beta1) rapidly develop autoimmune T-helper 1-mediated necroinflammatory liver disease. Susceptibility to liver damage is strictly regulated by genetic background. Whereas TGF-beta1-deficient mice on the BALB/c background develop necroinflammatory liver disease, TGF-beta1-deficient mice on the 129/CF-1 genetic background do not. We asked whether MHC locus haplotype is the principal determinant of genetic susceptibility to liver disease in this model system. BALB/c mice harbor the H-2d haplotype. We used a 'haplotype swapping' approach to generate H-2b or H-2k congenic BALB-background TGF-beta1-deficient mice. In addition, F1 (BALB/c x 129/CF-1)-TGF-beta1-deficient mice were generated. As determined by plasma transaminase levels and histopathology, severe necroinflammatory liver disease developed in all BALB-background TGF-beta1-deficient mice, regardless of H-2 haplotype, but developed neither in 129/CF-1-TGF-beta1-deficient mice nor in F1 (BALB/c x 129/CF-1)-TGF-beta1-deficient mice. Thus, H-2d is neither necessary nor sufficient for the development of necroinflammatory liver disease in BALB-background TGF-beta1-deficient mice. This constitutes the first direct evidence that susceptibility to autoimmune hepatocellular damage, at least in mice, can be determined by genetic loci distinct from the MHC.
Collapse
Affiliation(s)
- Jack T Lin
- Department of Microbiology and Immunology, The Norris Cotton Cancer Center, Dartmouth Medical School, Lebanon, NH 03756, USA
| | | | | | | |
Collapse
|
60
|
Lindqvist AKB, Nakken B, Sundler M, Kjellén P, Jonsson R, Holmdahl R, Skarstein K. Influence on Spontaneous Tissue Inflammation by the Major Histocompatibility Complex Region in the Nonobese Diabetic Mouse. Scand J Immunol 2005; 61:119-27. [PMID: 15683448 DOI: 10.1111/j.0300-9475.2005.01550.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
We investigated the role of the major histocompatibility complex (MHC) region in the specificity of autoimmunity by analysing specifically the development of sialadenitis, but also insulitis, nephritis and autoantibody production in autoimmune-prone nonobese diabetic (NOD) mice where the MHC H2g7 haplotype had been exchanged for the H2q (NOD.Q) or H2p (NOD.P) haplotype. The exchange of H2 haplotype did not affect the frequency of sialadenitis because the H2q and H2p congenic NOD strains developed sialadenitis with the same incidence as NOD. However, the severity of sialadenitis varied among the strains, as NOD.Q >NOD >NOD.P. At 11-13 weeks of age, the NOD.Q (H2q) female mice developed more severe sialadenitis compared to NOD.P (H2p) (P=0.038). At 20 weeks, the NOD (H2g7) female mice showed more severe sialadenitis than NOD.P (P=0.049). This is in contrast to the development of insulitis in the present strains, because the incidence of insulitis was almost completely inhibited by the replacement of the H2g7 haplotype of NOD. The incidence of insulitis in NOD.Q was 11-22%, compared to 75% in NOD, which correlated well with lower titres of anti-glutamic acid decarboxylase (anti-GAD) antibodies in NOD.Q compared to NOD (P=0.009). However, the introduction of the H2q haplotype into the NOD strain instead directed the autoimmune response towards the production of lupus types of autoantibodies, because the incidence of antinuclear antibodies (ANA) in NOD.Q was 89% compared with 11% in NOD.P and 12% in NOD mice, which in turn correlated with a high incidence of nephritis in NOD.Q compared to NOD. Consequently, we show that different haplotypes of MHC are instrumental in directing the specificity of the spontaneous autoimmune inflammation.
Collapse
Affiliation(s)
- A-K B Lindqvist
- Department of Cell and Molecular Biology, Section for Medical Inflammation Research, Lund University, Lund, Sweden.
| | | | | | | | | | | | | |
Collapse
|
61
|
Roep BO, Atkinson M, von Herrath M. Satisfaction (not) guaranteed: re-evaluating the use of animal models of type 1 diabetes. Nat Rev Immunol 2005; 4:989-97. [PMID: 15573133 DOI: 10.1038/nri1502] [Citation(s) in RCA: 149] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Without a doubt, rodent models have been instrumental in describing pathways that lead to pancreatic beta-cell destruction, evaluating potential causes of type 1 diabetes and providing proof-of-principle for the potential of immune-based interventions. However, despite more than two decades of productive research, we are still yet to define an initiating autoantigen for the human disease, to determine the precise mechanisms of beta-cell destruction in humans and to design interventions that prevent or cure type 1 diabetes. In this Perspective article, we propose that a major philosophical change would benefit this field, a proposition that is based on evaluation of situations in which rodent models have provided useful guidance and in which they have led to disappointments.
Collapse
Affiliation(s)
- Bart O Roep
- Bart O. Roep is at the Leiden University Medical Center, Department of Immunohematology and Blood Transfusion, Leiden NL-2300 RC, The Netherlands.
| | | | | |
Collapse
|
62
|
Ikegami H, Fujisawa T, Sakamoto T, Makino S, Ogihara T. Idd1 and Idd3 are necessary but not sufficient for development of type 1 diabetes in NOD mouse. Diabetes Res Clin Pract 2004; 66 Suppl 1:S85-90. [PMID: 15563987 DOI: 10.1016/j.diabres.2003.09.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2003] [Accepted: 09/27/2003] [Indexed: 11/26/2022]
Abstract
Type 1 diabetes in the NOD mouse is under polygenic control, with a major susceptibility gene, Idd1, in the major histocompatibility complex (MHC). To investigate the contribution of the NOD MHC to type 1 diabetes susceptibility, a B6.NOD-H-2 congenic strain, in which the NOD MHC was introgressed onto the genetic background of the C57BL/6 strain, was established. Despite possession of the diabetogenic MHC from the NOD mouse, none of the B6.NOD-H-2 mice developed type 1 diabetes, indicating that the NOD MHC alone is not sufficient for type 1 diabetes and that non-MHC genes are also necessary. One of the strongest non-MHC genes is Idd3, and Il2 which encodes interleukin 2, is a candidate gene for Idd3. To test whether a combination of the NOD MHC with the NOD allele of Il2 is sufficient for type 1 diabetes, B6.NOD-H-2 mice were crossed with C3H mice, which possess the NOD allele at Il2, and F2 mice homozygous for NOD alleles at both the MHC and Il2 were produced. None of the F2 mice developed type 1 diabetes, suggesting that NOD alleles at MHC (Idd1) and Il2 (Idd3) are not sufficient for type 1 diabetes in the NOD mouse.
Collapse
Affiliation(s)
- Hiroshi Ikegami
- Department of Geriatric Medicine, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan.
| | | | | | | | | |
Collapse
|
63
|
Salam MA, Matin K, Matsumoto N, Tsuha Y, Hanada N, Senpuku H. E2f1 mutation induces early onset of diabetes and Sjögren's syndrome in nonobese diabetic mice. THE JOURNAL OF IMMUNOLOGY 2004; 173:4908-18. [PMID: 15470032 DOI: 10.4049/jimmunol.173.8.4908] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
E2f1 is an important regulator of T cell proliferation, differentiation, and apoptosis that controls the transcription of a group of genes that are normally regulated at the G1 to S phase transition in the cell cycle. Insulin-dependent diabetes mellitus (IDDM) and Sjogren's syndrome (SS) are highly regulated autoimmune diseases that develop spontaneously in NOD mice. The aim of the present in vivo study was to explore the functional importance of the E2f1 molecule in IDDM and SS, in the context of whole animal physiology and pathophysiology, using E2f1-deficient NOD mice. For the experiment, we produced NOD mice homozygous for a nonfunctional E2f1 allele onto a NOD background. E2f1-deficient NOD mice developed an early and increased onset of diabetes as compared with their littermates. These mice also exhibited a defect in T lymphocyte development, leading to excessive numbers of mature T cells (CD4+ and CD8+), due to a maturation stage-specific defect in the apoptosis of thymocytes and peripheral T cells. We also found that they also exhibited a more rapid and increased entry into the S phase following antigenic stimulation of spleen cells and thymocytes in vitro. Furthermore, E2f1-deficient mice showed a profound decrease of immunoregulatory CD4+CD25+ T cells, while the spleen cells of NOD mice lacking E2f1 showed a significant increase of the proinflammatory cytokine IFN-gamma following antigenic stimulation in vitro. Consistent with these observations, E2f1 homozygous mutant NOD mice were highly predisposed to the development of IDDM and SS.
Collapse
Affiliation(s)
- Mohammad Abdus Salam
- Department of Bacteriology, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
64
|
Abdus Salam M, Matsumoto N, Matin K, Tsuha Y, Nakao R, Hanada N, Senpuku H. Establishment of an animal model using recombinant NOD.B10.D2 mice to study initial adhesion of oral streptococci. CLINICAL AND DIAGNOSTIC LABORATORY IMMUNOLOGY 2004; 11:379-86. [PMID: 15013991 PMCID: PMC371204 DOI: 10.1128/cdli.11.2.379-386.2004] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
An oral biofilm is a community of surface-attached microorganisms that coats the oral cavity, including the teeth, and provides a protective reservoir for oral microbial pathogens, which are the primary cause of persistent and chronic infectious diseases in patients with dry mouth or Sjögren's syndrome (SS). The purpose of this study was to establish an animal model for studying the initial adhesion of oral streptococci that cause biofilm formation in patients with dry mouth and SS in an attempt to decrease the influence of cariogenic organisms and their substrates. In nonobese diabetogenic (NOD) mice that spontaneously develop insulin-dependent diabetes mellitus (IDDM) and SS, we replaced major histocompatibility complex (MHC) class II (A(g7) E(g7)) and class I D(b) with MHC class II (A(d) E(d)) and class I D(d) from nondiabetic B10.D2 mice to produce an animal model that inhibited IDDM without affecting SS. The adhesion of oral streptococci, including Streptococcus mutans, onto tooth surfaces was then investigated and quantified in homologous recombinant N5 (NOD.B10.D2) and N9 (NOD.B10.D2) mice. We found that a higher number of oral streptococci adhered to the tooth surfaces of N5 (NOD.B10.D2) and N9 (NOD.B10.D2) mice than to those of the control C57BL/6 and B10.D2 mice. On the basis of our observation, we concluded that these mouse models might be useful as animal models of dry mouth and SS for in vivo biological studies of oral biofilm formation on the tooth surfaces.
Collapse
Affiliation(s)
- Mohammad Abdus Salam
- Department of Bacteriology, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo 162-8640, Japan
| | | | | | | | | | | | | |
Collapse
|
65
|
Takaki T, Lieberman SM, Holl TM, Han B, Santamaria P, Serreze DV, DiLorenzo TP. Requirement for Both H-2Db and H-2Kd for the Induction of Diabetes by the Promiscuous CD8+ T Cell Clonotype AI4. THE JOURNAL OF IMMUNOLOGY 2004; 173:2530-41. [PMID: 15294969 DOI: 10.4049/jimmunol.173.4.2530] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The NOD mouse is a model for autoimmune type 1 diabetes in humans. CD8(+) T cells are essential for the destruction of the insulin-producing pancreatic beta cells characterizing this disease. AI4 is a pathogenic CD8(+) T cell clone, isolated from the islets of a 5-wk-old female NOD mouse, which is capable of mediating overt diabetes in the absence of CD4(+) T cell help. Recent studies using MHC-congenic NOD mice revealed marked promiscuity of the AI4 TCR, as the selection of this clonotype can be influenced by multiple MHC molecules, including some class II variants. The present work was designed, in part, to determine whether similar promiscuity also characterizes the effector function of mature AI4 CTL. Using splenocyte and bone marrow disease transfer models and in vitro islet-killing assays, we report that efficient recognition and destruction of beta cells by AI4 requires the beta cells to simultaneously express both H-2D(b) and H-2K(d) class I MHC molecules. The ability of the AI4 TCR to interact with both H-2D(b) and H-2K(d) was confirmed using recombinant peptide libraries. This approach also allowed us to define a mimotope peptide recognized by AI4 in an H-2D(b)-restricted manner. Using ELISPOT and mimotope/H-2D(b) tetramer analyses, we demonstrate for the first time that AI4 represents a readily detectable T cell population in the islet infiltrates of prediabetic NOD mice. Our identification of a ligand for AI4-like T cells will facilitate further characterization and manipulation of this pathogenic and promiscuous T cell population.
Collapse
Affiliation(s)
- Toshiyuki Takaki
- Departments of Microbiology and Immunology, and Medicine (Division of Endocrinology), Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | | | | | | | | | | |
Collapse
|
66
|
King C, Ilic A, Koelsch K, Sarvetnick N. Homeostatic expansion of T cells during immune insufficiency generates autoimmunity. Cell 2004; 117:265-77. [PMID: 15084263 DOI: 10.1016/s0092-8674(04)00335-6] [Citation(s) in RCA: 516] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2003] [Revised: 02/10/2004] [Accepted: 02/13/2004] [Indexed: 11/21/2022]
Abstract
During illness and stress, the immune system can suffer a considerable loss of T cells (lymphopenia). The remaining T cells undergo vigorous compensatory expansion, known as homeostatic proliferation, to reconstitute the immune system. Interestingly, human diseases of autoimmune etiology often present with immune deficiencies such as lymphopenia. In this study, we show that reduced T cell numbers and the resulting exaggerated homeostatic-type proliferation of T cells generate autoimmunity. The cycling T cell population is short lived, and the depleted memory compartment fuels the generation of new effector T cells. A catalyst for these phenomena is the increased responses to the cytokine IL-21, a mediator that regulates T cell turnover. We conclude that poor T cell survival and lymphopenia precipitate autoimmune disease.
Collapse
Affiliation(s)
- Cecile King
- Department of Immunology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | | | | | | |
Collapse
|
67
|
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
|
68
|
Yang Y, Santamaria P. T-cell receptor-transgenic NOD mice: a reductionist approach to understand autoimmune diabetes. J Autoimmun 2004; 22:121-9. [PMID: 14987740 DOI: 10.1016/j.jaut.2003.10.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Yang Yang
- Department of Biochemistry & Molecular Biology, Faculty of Medicine, University of Calgary, 3330 Hospital Drive N.W., Calgary, Alberta, T2N 4N1, Canada
| | | |
Collapse
|
69
|
Ikegami H, Fujisawa T, Makino S, Ogihara T. Congenic mapping and candidate sequencing of susceptibility genes for Type 1 diabetes in the NOD mouse. Ann N Y Acad Sci 2004; 1005:196-204. [PMID: 14679059 DOI: 10.1196/annals.1288.026] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Inheritance of type 1 diabetes is polygenic with a major susceptibility gene located in the major histocompatibility complex (MHC). In addition to MHC-linked susceptibility, a number of susceptibility genes have been mapped outside the MHC in both humans and animal models. In order to localize and identify susceptibility genes for type 1 diabetes, we have developed a series of congenic strains in which either susceptibility intervals from the NOD mouse, a mouse model of type 1 diabetes, were introgressed onto control background genes or protective intervals from control strains were introgressed onto NOD background genes. NOD. CTS-H-2 congenic mice, which possess recombinant MHC with NOD alleles at class II A and E genes, which are candidates for Idd1, revealed that Idd1 consists of multiple components, one in class II (Idd1) and the other adjacent to, but distinct from, Idd1 (Idd16). Phenotypes of NOD. IIS-Idd3 congenic mice, which share the same alleles at both Il2 and Il21 as the NOD mouse, were indistinguishable from the NOD parental strain, indicating that both Il2 and Il21 are candidates for Idd3. In contrast, NOD. IIS-Idd10 congenic mice, which share the same alleles at Fcgr1, a previous candidate for Idd10, as the NOD mouse, were protected from type 1 diabetes, suggesting that Fcgr1 may not be responsible for the Idd10 effect. These data suggest that the use of strain colony closely related to a disease model to find the same candidate mutation on different haplotypes and make congenic strains with this recombinant chromosome, termed ancestral haplotype congenic mapping, is an effective strategy for fine mapping and identification of genes responsible for complex traits.
Collapse
Affiliation(s)
- Hiroshi Ikegami
- Department of Geriatric Medicine, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan.
| | | | | | | |
Collapse
|
70
|
Serreze DV, Holl TM, Marron MP, Graser RT, Johnson EA, Choisy-Rossi C, Slattery RM, Lieberman SM, DiLorenzo TP. MHC Class II Molecules Play a Role in the Selection of Autoreactive Class I-Restricted CD8 T Cells That Are Essential Contributors to Type 1 Diabetes Development in Nonobese Diabetic Mice. THE JOURNAL OF IMMUNOLOGY 2004; 172:871-9. [PMID: 14707058 DOI: 10.4049/jimmunol.172.2.871] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Development of autoreactive CD4 T cells contributing to type 1 diabetes (T1D) in both humans and nonobese diabetic (NOD) mice is either promoted or dominantly inhibited by particular MHC class II variants. In addition, it is now clear that when co-expressed with other susceptibility genes, some common MHC class I variants aberrantly mediate autoreactive CD8 T cell responses also essential to T1D development. However, it was unknown whether the development of diabetogenic CD8 T cells could also be dominantly inhibited by particular MHC variants. We addressed this issue by crossing NOD mice transgenically expressing the TCR from the diabetogenic CD8 T cell clone AI4 with NOD stocks congenic for MHC haplotypes that dominantly inhibit T1D. High numbers of functional AI4 T cells only developed in controls homozygously expressing NOD-derived H2(g7) molecules. In contrast, heterozygous expression of some MHC haplotypes conferring T1D resistance anergized AI4 T cells through decreased TCR (H2(b)) or CD8 expression (H2(q)). Most interestingly, while AI4 T cells exert a class I-restricted effector function, H2(nb1) MHC class II molecules can contribute to their negative selection. These findings provide insights to how particular MHC class I and class II variants interactively regulate the development of diabetogenic T cells and the TCR promiscuity of such autoreactive effectors.
Collapse
MESH Headings
- Animals
- Antigen-Presenting Cells/cytology
- Antigen-Presenting Cells/immunology
- Antigen-Presenting Cells/metabolism
- Autoantigens/immunology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/pathology
- Cell Differentiation/genetics
- Cell Differentiation/immunology
- Clonal Anergy/genetics
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/pathology
- Down-Regulation/genetics
- Down-Regulation/immunology
- Female
- Genetic Carrier Screening
- Genetic Variation/immunology
- H-2 Antigens/genetics
- H-2 Antigens/immunology
- H-2 Antigens/metabolism
- Haplotypes
- Hematopoietic Stem Cells/cytology
- Hematopoietic Stem Cells/immunology
- Hematopoietic Stem Cells/metabolism
- Histocompatibility Antigens Class II/biosynthesis
- Histocompatibility Antigens Class II/genetics
- Histocompatibility Antigens Class II/physiology
- Histocompatibility Testing
- Lymphocyte Activation/genetics
- Male
- Mice
- Mice, Inbred NOD
- Mice, Transgenic
- Receptors, Antigen, T-Cell/biosynthesis
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/physiology
- Signal Transduction/genetics
- Signal Transduction/immunology
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- T-Lymphocyte Subsets/pathology
Collapse
Affiliation(s)
- David V Serreze
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
71
|
Morgan DJ, Nugent CT, Raveney BJE, Sherman LA. In a Transgenic Model of Spontaneous Autoimmune Diabetes, Expression of a Protective Class II MHC Molecule Results in Thymic Deletion of Diabetogenic CD8+ T Cells. THE JOURNAL OF IMMUNOLOGY 2004; 172:1000-8. [PMID: 14707073 DOI: 10.4049/jimmunol.172.2.1000] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
H-2(d) mice expressing both the influenza virus hemagglutinin (HA) as a transgene-encoded protein on pancreatic islet beta cells (InsHA), as well as the Clone 4 TCR specific for the dominant H-2K(d)-restricted HA epitope, can be protected from the development of spontaneous autoimmune diabetes by expression of the H-2(b) haplotype. Protection occurs due to the deletion of K(d)HA-specific CD8+ T cells. This was unexpected as neither the presence of the InsHA transgene nor H-2(b), individually, resulted in thymic deletion. Further analyses revealed that thymic deletion required both a hybrid MHC class II molecule, Ebeta(b) Ealpha(d), and the K(d) molecule presenting the HA epitope, which together synergize to effect deletion of CD4+CD8+ thymocytes. This surprising example of protection from autoimmunity that maps to a class II MHC molecule, yet effects an alteration in the CD8+ T cell repertoire, suggests that selective events in the thymus represent the integrated strength of signal delivered to each cell through recognition of a variety of different MHC-peptide ligands.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- CD4 Antigens/biosynthesis
- CD4 Antigens/physiology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/pathology
- Cell Differentiation/genetics
- Cell Differentiation/immunology
- Clonal Deletion/genetics
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/pathology
- Diabetes Mellitus, Type 1/prevention & control
- Disease Models, Animal
- H-2 Antigens/biosynthesis
- H-2 Antigens/genetics
- H-2 Antigens/immunology
- H-2 Antigens/physiology
- Hemagglutinin Glycoproteins, Influenza Virus/biosynthesis
- Hemagglutinin Glycoproteins, Influenza Virus/genetics
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Histocompatibility Antigen H-2D
- Histocompatibility Antigens Class II/biosynthesis
- Histocompatibility Antigens Class II/genetics
- Histocompatibility Antigens Class II/physiology
- Insulin/genetics
- Insulin/immunology
- Islets of Langerhans/immunology
- Islets of Langerhans/metabolism
- Islets of Langerhans/virology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Promoter Regions, Genetic/immunology
- Rats
- Receptors, Antigen, T-Cell/biosynthesis
- Thymus Gland/immunology
- Thymus Gland/metabolism
- Thymus Gland/pathology
Collapse
Affiliation(s)
- David J Morgan
- University of Bristol, School of Medical Sciences, Bristol, United Kingdom
| | | | | | | |
Collapse
|
72
|
Affiliation(s)
- Michelle Solomon
- Department of Immunology, The Scripps Research Institute, La Jolla, California, USA
| | | |
Collapse
|
73
|
Pearson T, Markees TG, Serreze DV, Pierce MA, Marron MP, Wicker LS, Peterson LB, Shultz LD, Mordes JP, Rossini AA, Greiner DL. Genetic disassociation of autoimmunity and resistance to costimulation blockade-induced transplantation tolerance in nonobese diabetic mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 171:185-95. [PMID: 12816997 DOI: 10.4049/jimmunol.171.1.185] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Curing type 1 diabetes by islet transplantation requires overcoming both allorejection and recurrent autoimmunity. This has been achieved with systemic immunosuppression, but tolerance induction would be preferable. Most islet allotransplant tolerance induction protocols have been tested in nonobese diabetic (NOD) mice, and most have failed. Failure has been attributed to the underlying autoimmunity, assuming that autoimmunity and resistance to transplantation tolerance have a common basis. Out of concern that NOD biology could be misleading in this regard, we tested the hypothesis that autoimmunity and resistance to transplantation tolerance in NOD mice are distinct phenotypes. Unexpectedly, we observed that (NOD x C57BL/6)F(1) mice, which have no diabetes, nonetheless resist prolongation of skin allografts by costimulation blockade. Further analyses revealed that the F(1) mice shared the dendritic cell maturation defects and abnormal CD4(+) T cell responses of the NOD but had lost its defects in macrophage maturation and NK cell activity. We conclude that resistance to allograft tolerance induction in the NOD mouse is not a direct consequence of overt autoimmunity and that autoimmunity and resistance to costimulation blockade-induced transplantation tolerance phenotypes in NOD mice can be dissociated genetically. The outcomes of tolerance induction protocols tested in NOD mice may not accurately predict outcomes in human subjects.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antigens, CD/biosynthesis
- Autoimmune Diseases/genetics
- Autoimmune Diseases/pathology
- B7-2 Antigen
- Bone Marrow Cells/immunology
- Bone Marrow Cells/pathology
- CD4-CD8 Ratio
- CD40 Antigens/immunology
- CD40 Ligand/immunology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/pathology
- Cell Adhesion/genetics
- Cell Adhesion/immunology
- Cell Count
- Cell Differentiation/genetics
- Cell Differentiation/immunology
- Cells, Cultured
- Crosses, Genetic
- Cytotoxicity, Immunologic/genetics
- Dendritic Cells/immunology
- Dendritic Cells/pathology
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/pathology
- Female
- Genetic Markers
- Genetic Predisposition to Disease
- Graft Survival/genetics
- Graft Survival/immunology
- Homozygote
- Immunity, Innate/genetics
- Injections, Intravenous
- Killer Cells, Natural/immunology
- Killer Cells, Natural/pathology
- Lymphocyte Activation/immunology
- Lymphocyte Transfusion
- Lymphopenia/genetics
- Lymphopenia/immunology
- Lymphopenia/pathology
- Macrophages/cytology
- Macrophages/immunology
- Male
- Membrane Glycoproteins/biosynthesis
- Mice
- Mice, Inbred C3H
- Mice, Inbred C57BL
- Mice, Inbred NOD
- Mice, Knockout
- Receptors, Interleukin-2/biosynthesis
- Skin Transplantation/immunology
- Transplantation Tolerance/genetics
Collapse
Affiliation(s)
- Todd Pearson
- Program in Immunology and Virology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
74
|
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
|
75
|
Abstract
Autoimmune diseases are, in general, under complex genetic control and subject to strong interactions between genetics and the environment. Greater knowledge of the underlying genetics will provide immunologists with a framework for study of the immune dysregulation that occurs in such diseases. Ascertaining the number of genes that are involved and their characterization have, however, proven to be difficult. Improved methods of genetic analysis and the availability of a draft sequence of the complete mouse genome have markedly improved the outlook for such research, and they have emphasized the advantages of mice as a model system. In this review, we provide an overview of the genetic analysis of autoimmune diseases and of the crucial role of congenic and consomic mouse strains in such research.
Collapse
Affiliation(s)
- Ute C Rogner
- Institut Pasteur, Unité Génétique Moléculaire Murine, 25 rue du Docteur Roux, 75015 Paris, France
| | | |
Collapse
|
76
|
Beilhack GF, Scheffold YC, Weissman IL, Taylor C, Jerabek L, Burge MJ, Masek MA, Shizuru JA. Purified allogeneic hematopoietic stem cell transplantation blocks diabetes pathogenesis in NOD mice. Diabetes 2003; 52:59-68. [PMID: 12502494 DOI: 10.2337/diabetes.52.1.59] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Purified hematopoietic stem cells (HSCs) were transplanted into NOD mice to test whether development of hyperglycemia could be prevented. Engraftment of major histocompatibility complex-mismatched HSCs was compared with bone marrow (BM) grafts. HSCs differed from BM because HSCs were more strongly resisted and HSC recipients retained significant levels of NOD T-cells, whereas BM recipients were full donor chimeras. Despite persistent NOD T-cells, all HSC chimeras were protected from hyperglycemia, and attenuation of islet lesions was observed. T-cell selection was altered in allogeneic HSC recipients as demonstrated by deletion of both donor and host superantigen-specific T-cells. Syngeneic and congenic hematopoietic cell transplants were also performed to differentiate the influence of the preparative regimen(s) versus the allografts. Unlike the allogeneic HSC transplantations, syngeneic or congenic grafts did not retard diabetes development. In a pilot study, overtly diabetic NOD mice were cured by co-transplantation of allogeneic HSCs and donor-matched islets. We conclude that allogeneic HSC transplants block allo- and autoimmunity, despite residual host T-cell presence. These data demonstrate for the first time that purified HSC grafts block development of autoimmune diabetes and illuminate how HSC grafts alter thymic and peripheral T-cell responses against auto- and alloantigens.
Collapse
Affiliation(s)
- Georg F Beilhack
- Department of Medicine, Division of Bone Marrow Transplantation, Stanford University Medical Center, Stanford, California 94305, USA
| | | | | | | | | | | | | | | |
Collapse
|
77
|
Kudva YC, Rajagopalan G, Raju R, Abraham RS, Smart M, Hanson J, David CS. Modulation of insulitis and type 1 diabetes by transgenic HLA-DR3 and DQ8 in NOD mice lacking endogenous MHC class II. Hum Immunol 2002; 63:987-99. [PMID: 12392851 DOI: 10.1016/s0198-8859(02)00435-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
To evaluate the contributions of DR3 and DQ8 to the etiopathogenesis of type 1 diabetes in a diabetes-predisposing milieu, we developed human leukocyte antigen (HLA) transgenic mice on the nonobese diabetic (NOD) background in the absence of the endogenous class II molecule, I-A(g7) and studied the incidence of both spontaneous and experimental (induced) autoimmune diabetes. Transgenic expression of HLA-DR3 and -DQ8 (either alone or in combination) did not confer susceptibility to spontaneous or cyclophosphamide-induced type 1 diabetes. Expression of I-A(g7) was mandatory for development of spontaneous or cyclophosphamide-induced diabetes. However, multiple low doses of streptozotocin could induce diabetes in all groups of mice independent of the class II molecules expressed. In unmanipulated mice, only islets from I-A(g7+/+) mice revealed significant intra-islet infiltration. Although a characteristic peri-insulitis/peri-ductulitis was present in Abeta(0)/NOD mice, islets from DR3, DQ8 and DR3 x DQ8 double transgenic mice demonstrated significantly less infiltration. In conclusion, transgenic expression of HLA-DR3 and -DQ8 associated with predisposition to type 1 diabetes alone is not sufficient to induce spontaneous diabetes in NOD mice lacking endogenous class II molecules.
Collapse
Affiliation(s)
- Yogish C Kudva
- Division of Endocrinology and Metabolism, Mayo Clinic, Rochester, MN, USA
| | | | | | | | | | | | | |
Collapse
|
78
|
Marron MP, Graser RT, Chapman HD, Serreze DV. Functional evidence for the mediation of diabetogenic T cell responses by HLA-A2.1 MHC class I molecules through transgenic expression in NOD mice. Proc Natl Acad Sci U S A 2002; 99:13753-8. [PMID: 12361980 PMCID: PMC129768 DOI: 10.1073/pnas.212221199] [Citation(s) in RCA: 65] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2002] [Indexed: 11/18/2022] Open
Abstract
Particular major histocompatibility complex (MHC) class II alleles clearly contribute to T cell-mediated autoimmune type 1 diabetes (T1D) in both humans and nonobese diabetic (NOD) mice. However, studies in NOD mice indicate MHC class I-restricted T cell responses are also essential to T1D development. In humans, epidemiological studies have suggested that some common class I alleles, including HLA-A2.1 (A*02011), may confer increased susceptibility to T1D when expressed in conjunction with certain class II alleles. We show here that when HLA-A2.1 molecules are transgenically expressed in NOD mice, A2-restricted T cell responses arise against pancreatic beta cells, leading to an earlier onset of T1D. The accelerated onset of T1D in the NOD.HLA-A2.1 transgenic mice is not due to nonspecific effects of expressing a third class I molecule, because a stock of NOD mice transgenically expressing HLA-B27 class I molecules showed no such acceleration of T1D, but rather were significantly protected from disease. These findings provide the first functional evidence that certain human MHC class I molecules can contribute to the development of T1D.
Collapse
Affiliation(s)
- Michele P Marron
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | | | | | | |
Collapse
|
79
|
Matin K, Salam MA, Akhter J, Hanada N, Senpuku H. Role of stromal-cell derived factor-1 in the development of autoimmune diseases in non-obese diabetic mice. Immunology 2002; 107:222-32. [PMID: 12383202 PMCID: PMC1782793 DOI: 10.1046/j.1365-2567.2002.01478.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2001] [Revised: 05/07/2002] [Accepted: 05/14/2002] [Indexed: 11/20/2022] Open
Abstract
The chemokine stromal-cell derived factor-1 (SDF-1) controls maturation, trafficking, and homing of certain subsets, lymphoid cells including immunogenic B and T cells, as a ligand of the CXCR4 chemokine receptor. Insulin-dependent diabetes mellitus (IDDM) and Sjögren's syndrome (SS), both highly regulated autoimmune diseases, develop spontaneously in non-obese diabetic (NOD) mice. To investigate the role of SDF-1 in the development of autoimmune diseases, we injected groups of NOD female mice with antibodies to SDF-1 (anti-SDF-1), which resulted in a 30% reduction of diabetes up to 30 weeks of age, delayed average diabetes onset by 10 weeks, and suppressed insulitis. Autoimmune sialoadenitis was evident in anti-SDF-1-injected mice (SDF-1-Ig group) at the same level as in all groups of mice, whether injected with non-specific antibodies or not. In addition, in the SDF-1-Ig group, a greater number of immunoglobulin M (IgM)- IgD- B220(low) CD38+ CD43+ CD23- progenitor B cells and IgM+ IgD+ B220(high) CD43- CD38+ CD24+ CD23+ mature B cells remained in the bone marrow, whereas infiltration of mature IgM+ B cells was less extensive in peripheral tissues. Our results suggested that anti-SDF-1 antibodies injection was effective in inhibiting diabetes and insulitis without affecting autoimmune sialoadenitis or SS in NOD mice. SDF-1 may be an essential chemokine for trafficking and migration of autoreactive B cells in the development of diabetes.
Collapse
Affiliation(s)
- Khairul Matin
- Department of Oral Science, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo 162-8650, Japan
| | | | | | | | | |
Collapse
|
80
|
Boulard O, Damotte D, Deruytter N, Fluteau G, Carnaud C, Garchon HJ. An interval tightly linked to but distinct from the H2 complex controls both overt diabetes (Idd16) and chronic experimental autoimmune thyroiditis (Ceat1) in nonobese diabetic mice. Diabetes 2002; 51:2141-7. [PMID: 12086944 DOI: 10.2337/diabetes.51.7.2141] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The major histocompatibility complex (MHC) has long been associated with predisposition to several autoimmune diseases, including type 1 diabetes and autoimmune thyroiditis. In type 1 diabetes, a primary role has been assigned to class II genes, both in humans and in the nonobese diabetic (NOD) mouse model. However, an involvement of other tightly linked genes is strongly suspected. Here, through two independent sets of experiments, we provide solid evidence for the existence of at least one such gene. First, using a new recombinant congenic NOD strain, R114, we definitively individualized the Idd16 locus from the MHC in a 6-cM interval proximal to H2-K. It affords almost complete protection against diabetes and is associated with delayed insulitis. Second, by genome scan, we mapped non-H2 genes associated with the highly penetrant form of chronic experimental autoimmune thyroiditis (EAT) that is elicited in NOD and NOD.H2(k) mice by immunization with thyroglobulin. We identified one major dominant locus, Ceat1, on chromosome 17, overlapping with Idd16. Most importantly, R114 recombinant congenic mice challenged with thyroglobulin did not develop chronic EAT. This new major region defined by both Idd16 and Ceat1 might thus concur to the unique strength of the MHC in autoimmune susceptibility of NOD mice.
Collapse
Affiliation(s)
- Olivier Boulard
- INSERM U25, Hôpital Necker-Enfants malades, 161 rue de Sèvres, 75743 Paris Cedex 15, France
| | | | | | | | | | | |
Collapse
|
81
|
Cha S, Nagashima H, Brown VB, Peck AB, Humphreys-Beher MG. Two NOD Idd-associated intervals contribute synergistically to the development of autoimmune exocrinopathy (Sjögren's syndrome) on a healthy murine background. ARTHRITIS AND RHEUMATISM 2002; 46:1390-8. [PMID: 12115247 DOI: 10.1002/art.10258] [Citation(s) in RCA: 113] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
OBJECTIVE The NOD mouse is genetically predisposed to the development of at least 2 autoimmune diseases, autoimmune diabetes and autoimmune exocrinopathy (AEC). More than 19 chromosomal intervals (referred to as Idd regions) that contribute to diabetes susceptibility in the NOD mouse model have been identified, but only 2 chromosomal intervals (associated with Idd3 and Idd5) have been shown to control sialadenitis. In the present study, we bred the Idd3 and Idd5 chromosomal intervals from NOD mice into non-autoimmune C57BL/6 mice to determine if these intervals recreate a Sjögren's syndrome (SS)-like phenotype. METHODS C57BL/6.NODc3 mice carrying Idd3 and C57BL/6.NODc1t mice carrying Idd5 were crossed and intercrossed to generate a C57BL/6.NODc3.NODc1t mouse line homozygous for the Idd3 and Idd5 chromosomal intervals on an otherwise disease-resistant genetic background. C57BL/6.NODc3.NODc1t mice were evaluated for biochemical, pathophysiologic, and immunologic markers characteristic of the SS-like phenotype present in the NOD mouse. RESULTS C57BL/6.NODc3.NODc1t mice fully manifested the SS-like phenotype of the NOD mouse, including decreased salivary and lacrimal gland secretory flow rates, increased salivary protein content due in part to less fluid, aberrant proteolytic enzyme activity, decline in amylase activity, appearance of autoantibodies to exocrine gland proteins, and glandular lymphocytic focal infiltrates. Loss of secretory function occurred more rapidly in C57BL/6.NODc3.NODc1t mice (by 12 weeks of age) than in NOD mice (by 16 weeks of age). No signs of insulitis or autoimmune (type 1) diabetes were observed in the C57BL/6.NODc3.NODc1t mice. CONCLUSION Genes located within the 2 chromosomal intervals Idd3 and Idd5 appear necessary and sufficient for manifestation of AEC. We propose that this murine model of SS-like disease be designated C57BL/6.NOD-Aec1Aec2. Identification of specific genes within the Aec1 and Aec2 genetic regions should help elucidate the mechanism(s) underlying SS-like disease.
Collapse
Affiliation(s)
- Seunghee Cha
- University of Florida, Gainesville, FL 32610, USA
| | | | | | | | | |
Collapse
|
82
|
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
|
83
|
Thiessen S, Serra P, Amrani A, Verdaguer J, Santamaria P. T-cell tolerance by dendritic cells and macrophages as a mechanism for the major histocompatibility complex-linked resistance to autoimmune diabetes. Diabetes 2002; 51:325-38. [PMID: 11812739 DOI: 10.2337/diabetes.51.2.325] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
For poorly understood reasons, the development of autoimmune diabetes in humans and mice is dominantly inhibited by major histocompatibility complex (MHC) class II molecules with diverse antigen-binding sites. We have previously shown that thymocytes expressing a highly diabetogenic I-A(g7)-restricted T-cell receptor (TCR) (4.1-TCR) undergo negative selection in mice carrying one copy of the antidiabetogenic H-2(b) haplotype in an I-A(b)-dependent but superantigen-independent manner. Here, we show that 4.1-TCR-transgenic thymocytes undergo different forms of tolerance in NOD mice expressing antidiabetogenic I-A(d), I-A(g7PD), or I-Ealpha(k) transgenes. The ability of protective MHC class II molecules to induce thymocyte tolerance in 4.1-TCR-transgenic NOD mice correlates with their ability to prevent diabetes in non-TCR-transgenic mice and is associated with polymorphisms within positions 56-67 of their beta1 domains. The 4.1-thymocyte tolerogenic activity of these MHC class II molecules is mediated by dendritic cells and macrophages but not by B-cells or thymic epithelial cells and is a peptide-dependent process. Antidiabetogenic MHC class II molecules may thus afford diabetes resistance by presenting, on dendritic cells and macrophages, tolerogenic peptides to a subset of highly diabetogenic and MHC-promiscuous CD4(+) T-cells that play a critical role in the initiation of diabetes.
Collapse
Affiliation(s)
- Shari Thiessen
- Department of Microbiology and Infectious Diseases, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
| | | | | | | | | |
Collapse
|
84
|
Salaün J, Simmenauer N, Belo P, Coutinho A, Le Douarin NM. Grafts of supplementary thymuses injected with allogeneic pancreatic islets protect nonobese diabetic mice against diabetes. Proc Natl Acad Sci U S A 2002; 99:874-7. [PMID: 11792835 PMCID: PMC117398 DOI: 10.1073/pnas.012597499] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2001] [Indexed: 12/23/2022] Open
Abstract
In nonobese diabetic (NOD) mice, the autoimmune attack of the beta-cells in pancreatic islets is now believed to result from abnormal thymic selection. Accordingly, grafts of thymic epithelium from NOD donors to athymic recipients promote autoimmune islet inflammation in normal strains, and intrathymic islet grafts decrease the incidence of disease in NOD animals. Two competing hypotheses of abnormal thymic selection in diabetic mice have been proposed: deficient negative selection with poor elimination of aggressive organ-specific T cells vs. deficient positive selection of protective T regulatory cells. We have now addressed these alternatives by grafting, into young NOD mice whose own thymus was left intact, newborn NOD thymuses containing allogeneic pancreatic islets. If the NOD defect represented poor negative selection, these animals would develop disease at control rates, as the generation of autoreactive T cells proceeds undisturbed in the autologous thymus. In contrast, if NOD thymuses are defective in the production of T regulatory cells, lower disease incidence is expected in the chimeras, as more protective cells can be produced in the grafted thymus. The results show a reduced incidence of diabetes in the chimeras (24%) as compared with control (72%) NOD mice, throughout adult life. We conclude that amelioration of NOD mice by intrathymic islet grafts is not caused by enhanced negative selection and suggest that autoimmune diabetes in this system is the result of inefficient generation of T regulatory cells in the thymus.
Collapse
Affiliation(s)
- J Salaün
- Institut d'Embryologie Cellulaire et Moléculaire du Centre National de la Recherche Scientifique et du Collège de France 49bis, Avenue de la Belle Gabrielle, 94736 Nogent-sur-Marne Cedex, France.
| | | | | | | | | |
Collapse
|
85
|
Johansson ACM, Nakken B, Sundler M, Lindqvist AKB, Johannesson M, Alarcón-Riquelme M, Bolstad AI, Humphreys-Beher MG, Jonsson R, Skarstein K, Holmdahl R. The genetic control of sialadenitis versus arthritis in a NOD.QxB10.Q F2 cross. Eur J Immunol 2002; 32:243-50. [PMID: 11782015 DOI: 10.1002/1521-4141(200201)32:1<243::aid-immu243>3.0.co;2-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The non-obese diabetic (NOD) mouse spontaneously develops diabetes and sialadenitis. The sialadenitis is characterized by histopathological changes in salivary glands and functional deficit similar to Sjögren's syndrome. In humans, Sjögren's syndrome could be associated with other connective tissue disorders, such as rheumatoid arthritis. In the present study the genetic control of sialadenitis in mice was compared to that of arthritis. We have previously reported a NOD locus, identified in an F2 cross with the H2(q) congenic NOD (NOD.Q) and C57BL/10.Q (B10.Q) strains, that promoted susceptibility to collagen-induced arthritis. The sialadenitis in NOD.Q showed a similar histological phenotype as in NOD, whereas no submandibular gland infiltration was found in B10.Q. The development of sialadenitis was independent of immunization with type II collagen and established arthritis. To identify the genetic control of sialadenitis, a gene segregation experiment was performed on an (NOD.QxB10.Q)F2 cross and genetic mapping of 353 F2 mice revealed one significant locus associated with sialadenitis on chromosome 4, LOD score 4.7. The NOD.Q allele-mediated susceptibility under a recessive inheritance pattern. The genetic control of sialadenitis seemed to be unique in comparison to diabetes and arthritis, as no loci associated with these diseases have been identified at the same location.
Collapse
Affiliation(s)
- Asa C M Johansson
- Section for Medical Inflammation Research, University of Lund, Lund, Sweden.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
86
|
Chiu PP, Jevnikar AM, Danska JS. Genetic control of T and B lymphocyte activation in nonobese diabetic mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:7169-79. [PMID: 11739540 DOI: 10.4049/jimmunol.167.12.7169] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Type 1 diabetes in nonobese diabetic (NOD) mice is characterized by the infiltration of T and B cells into pancreatic islets. T cells bearing the TCR Vbeta3 chain are disproportionately represented in the earliest stages of islet infiltration (insulitis) despite clonal deletion of most Vbeta3(+) immature thymocytes by the mammary tumor virus-3 (Mtv-3) superantigen (SAg). In this report we showed that a high frequency of NOD Vbeta3(+) T cells that escape deletion are activated in vivo and that this phenotype is linked to the Mtv-3 locus. One potential mechanism of SAg presentation to peripheral T cells is by activated B cells. Consistent with this idea, we found that NOD mice harbor a significantly higher frequency of activated B cells than nondiabetes-prone strains. These activated NOD B cells expressed cell surface molecules consistent with APC function. At the molecular level, the IgH repertoire of activated B cells in NOD mice was equivalent to resting B cells, suggesting a polyclonal response in vivo. Genetic analysis of the activated B cell phenotype showed linkage to Idd1, the NOD MHC haplotype (H-2(g7)). Finally, Vbeta3(+) thymocyte deletion and peripheral T cell activation did not require B cells, suggesting that other APC populations are sufficient to generate both Mtv-3-linked phenotypes. These data provide insight into the genetic regulation of NOD autoreactive lymphocyte activation that may contribute to failure of peripheral tolerance and the pathogenesis of type I diabetes.
Collapse
MESH Headings
- Animals
- Antigens, CD/analysis
- Antigens, Differentiation, T-Lymphocyte/analysis
- Antigens, Viral/genetics
- Antigens, Viral/immunology
- B-Lymphocytes/immunology
- Cells, Cultured
- Chromosome Mapping
- Clonal Deletion
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/immunology
- Flow Cytometry
- Genes, T-Cell Receptor beta
- Immunoglobulin Variable Region/genetics
- Lectins, C-Type
- Lymphocyte Activation
- Major Histocompatibility Complex
- Mice
- Mice, Inbred NOD
- Proviruses/immunology
- Receptors, Antigen, T-Cell/metabolism
- Superantigens/genetics
- Superantigens/immunology
- T-Lymphocytes/immunology
Collapse
Affiliation(s)
- P P Chiu
- Program in Developmental Biology, Hospital for Sick Children Research Institute, Department of Surgery, University of Toronto, Toronto, Canada
| | | | | |
Collapse
|
87
|
Trembleau S, Gregori S, Penna G, Gorny I, Adorini L. IL-12 administration reveals diabetogenic T cells in genetically resistant I-Ealpha-transgenic nonobese diabetic mice: resistance to autoimmune diabetes is associated with binding of Ealpha-derived peptides to the I-A(g7) molecule. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:4104-14. [PMID: 11564833 DOI: 10.4049/jimmunol.167.7.4104] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Nonobese diabetic (NOD) and NOD-DRalpha transgenic (tg) mice, expressing Aalpha(d):Abeta(g7) and Aalpha(d):Abeta(g7) plus DRalpha:Ebeta(g7) class II molecules, respectively, both develop insulin-dependent diabetes mellitus (IDDM), whereas NOD-Ealpha tg mice expressing Aalpha(d):Abeta(g7) plus Ealpha:Ebeta(g7) are protected. We show that IL-12 administration induces rapid IDDM onset in NOD-DRalpha but fails to provoke insulitis and diabetes in NOD-Ealpha tg mice. Nevertheless, T cells from IL-12-treated NOD-Ealpha tg mice secrete IFN-gamma and transfer IDDM to NOD-SCID and NOD-Ealpha-SCID recipients, demonstrating the presence of peripheral diabetogenic Th1 cells in the protected mice. Surprisingly, regulatory cells were undetectable. Moreover, Ealpha:Ebeta(g7) could substitute for DRalpha:Ebeta(g7) in Ag presentation, arguing against mechanisms of protection involving capture of diabetogenic I-A(g7)-restricted epitopes by Ealpha:Ebeta(g7)molecules. Interestingly, the expression of naturally processed epitopes derived from DRalpha- and Ealpha-chains bound to I-A(g7) is different in the two strains of tg mice, and the difference is enhanced by IL-12 administration. I-A(g7) molecules from both NOD-DRalpha and NOD-Ealpha tg mice present the conserved DRalpha/Ealpha 52-68 sequence, at high and low levels, respectively. In addition, only IDDM-resistant NOD-Ealpha tg mice possess APCs bearing Ealpha65-77/I-A(g7) complexes, which tolerize the specific T cells. This is associated with the selective inhibition of the response to insulinoma-associated protein 2 (IA-2), an autoantigen in IDDM. Our results support protective mechanisms based on I-A(g7) blockade by peptides unique to the Ealpha-chain, such as Ealpha65-77 and/or tolerance of diabetogenic T cells cross-reactive with Ealpha-peptide/I-A(g7) complexes.
Collapse
MESH Headings
- Animals
- Antigen Presentation
- Antigen-Presenting Cells/immunology
- Antigens, Surface/immunology
- Antigens, Surface/metabolism
- Autoantigens
- Cells, Cultured
- Cytokines/biosynthesis
- Diabetes Mellitus, Type 1/immunology
- HLA-DR Antigens/genetics
- Histocompatibility Antigens Class II/genetics
- Histocompatibility Antigens Class II/immunology
- Histocompatibility Antigens Class II/metabolism
- Interleukin-12/pharmacology
- Membrane Proteins/immunology
- Mice
- Mice, Inbred NOD
- Mice, SCID
- Mice, Transgenic
- Pancreas/immunology
- Peptide Fragments
- Protein Tyrosine Phosphatase, Non-Receptor Type 1
- Protein Tyrosine Phosphatases/immunology
- Receptor-Like Protein Tyrosine Phosphatases, Class 8
- Receptors, Antigen, T-Cell
- T-Lymphocytes/immunology
- T-Lymphocytes/transplantation
- Th1 Cells/immunology
Collapse
Affiliation(s)
- S Trembleau
- Roche Milan Ricerche, Via Olgettina 58, I-20132 Milan, Italy
| | | | | | | | | |
Collapse
|
88
|
Abstract
Tolerance to beta cell autoantigens represents a fragile equilibrium. Autoreactive T cells specific to these autoantigens are present in most normal individuals but are kept under control by a number of peripheral tolerance mechanisms, among which CD4(+) CD25(+) CD62L(+) T cell-mediated regulation probably plays a central role. The equilibrium may be disrupted by inappropriate activation of autoantigen-specific T cells, notably following to local inflammation that enhances the expression of the various molecules contributing to antigen recognition by T cells. Even when T cell activation finally overrides regulation, stimulation of regulatory cells by CD3 antibodies may reset the control of autoimmunity. Other procedures may also lead to disease prevention. These procedures are essentially focused on Th2 cytokines, whether used systemically or produced by Th2 cells after specific stimulation by autoantigens. Protection can also be obtained by NK T cell stimulation. Administration of beta cell antigens or CD3 antibodies is now being tested in clinical trials in prediabetics and/or recently diagnosed diabetes.
Collapse
MESH Headings
- Animals
- Antigen Presentation
- Autoantigens/immunology
- Autoantigens/therapeutic use
- Autoimmune Diseases/immunology
- CD4-Positive T-Lymphocytes/immunology
- Clinical Trials as Topic
- Clinical Trials, Phase I as Topic
- Clinical Trials, Phase II as Topic
- Clonal Anergy
- Clonal Deletion
- Cytokines/physiology
- Desensitization, Immunologic
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/prevention & control
- Diabetes Mellitus, Type 1/therapy
- Genetic Predisposition to Disease
- Humans
- Immune Tolerance
- Islets of Langerhans/immunology
- Killer Cells, Natural/immunology
- Lymphocyte Activation
- Mice
- Mice, Inbred C57BL
- Mice, Inbred NOD
- Mice, Transgenic
- Muromonab-CD3/therapeutic use
- Prediabetic State/therapy
- T-Lymphocyte Subsets/immunology
- Th2 Cells/immunology
Collapse
Affiliation(s)
- J F Bach
- INSERM U 25, Hôpital Necker, 161 rue de Sèvres, Paris Cedex 15, 75743 France.
| | | |
Collapse
|
89
|
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
|
90
|
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
|
91
|
Bergman ML, Cilio CM, Penha-Gonçalves C, Lamhamedi-Cherradi SE, Löfgren A, Colucci F, Lejon K, Garchon HJ, Holmberg D. CTLA-4-/- mice display T cell-apoptosis resistance resembling that ascribed to autoimmune-prone non-obese diabetic (NOD) mice. J Autoimmun 2001; 16:105-13. [PMID: 11247636 DOI: 10.1006/jaut.2000.0474] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The genes conferring susceptibility to autoimmune (insulin-dependent) diabetes mellitus (IDDM) are, in most cases, not defined. Among the loci so far identified as associated with murine IDDM (Idd1-19), only the nature of Idd1 has been assessed. Here we show that thymocytes and peripheral lymphocytes of the non-obese diabetic (NOD) mouse are relatively resistant to apoptosis induced by gamma-irradiation. By linkage analysis of F2 progeny mice, we map this trait to a locus on chromosome 1 containing the Idd5 diabetes susceptibility region. By the use of congenic mice, we confirm the linkage data and map this locus to a 6 cM region on proximal chromosome 1. Ctla4, being localized in this chromosomal region and mediating crucial functions in T cell biology, is a logical candidate gene in the Idd5 susceptibility region. In line with this, we demonstrate that T cells from Ctla4(-/-)deficient mice show a similar resistance to gamma-irradiation-induced apoptosis as observed in the NOD mice. This reinforces the notion that CTLA-4 contributes to the pathogenesis of autoimmune diabetes.
Collapse
Affiliation(s)
- M L Bergman
- Department of Cell and Molecular Biology, University of Umeå, S-901 87 Umeå Sweden
| | | | | | | | | | | | | | | | | |
Collapse
|
92
|
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
|
93
|
Yamakawa M, Weinstein R, Tsuji T, McBride J, Wong DT, Login GR. Age-related alterations in IL-1beta, TNF-alpha, and IL-6 concentrations in parotid acinar cells from BALB/c and non-obese diabetic mice. J Histochem Cytochem 2000; 48:1033-42. [PMID: 10898798 DOI: 10.1177/002215540004800802] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
IL-1beta, TNF-alpha, and IL-6 have been implicated in the destruction of parotid gland acinar cells (but not duct cells) in autoimmune sialoadenitis. Here we report the temporal alterations of these cytokines in parotid acinar cells that may lead to this specificity in cell death in the non-obese diabetic (NOD) mouse model for Sjögren's syndrome. Immunohistochemistry on paraffin sections of parotid gland from 5- and 10-week-old BALB/c and NOD mice confirmed the presence of many peri-acinar lymphoid nodules but few T-cells and macrophages between acinar cells. RT-PCR on enzymatically dispersed mouse parotid acinar cells (MPACs) showed no bands for CD3varepsilon, CD20, or F4/80 regardless of mouse strain or age. By ELISA, MPACs from 10-week-old NODs showed a small but highly significant (p<0.003) increase in IL-1beta and a large significant decrease (p<0.008) in IL-6 compared to 5-week-old NODs. Norepinephrine-stimulated amylase release from MPACs was not different regardless of mouse strain or age. These data show that alterations in acinar cell production of IL-1beta and IL-6 in aging NODs precede periductal lymphoid aggregates and acinar cell secretory dysfunction. (J Histochem Cytochem 48:1033-1041,2000)
Collapse
Affiliation(s)
- M Yamakawa
- Department of Oral Medicine and Diagnostic Sciences, Harvard School of Dental Medicine, Boston, Massachusetts
| | | | | | | | | | | |
Collapse
|
94
|
Fox CJ, Paterson AD, Mortin-Toth SM, Danska JS. Two genetic loci regulate T cell-dependent islet inflammation and drive autoimmune diabetes pathogenesis. Am J Hum Genet 2000; 67:67-81. [PMID: 10848492 PMCID: PMC1287103 DOI: 10.1086/302995] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/1999] [Accepted: 05/08/2000] [Indexed: 12/21/2022] Open
Abstract
Insulin-dependent diabetes mellitus (IDDM) is a polygenic disease caused by progressive autoimmune infiltration (insulitis) of the pancreatic islets of Langerhan, culminating in the destruction of insulin-producing beta cells. Genome scans of families with diabetes suggest that multiple loci make incremental contributions to disease susceptibility. However, only the IDDM1 locus is well characterized, at a molecular and functional level, as alleleic variants of the major histocompatibility complex (MHC) class II HLA-DQB1, DRB1, and DPB1 genes that mediate antigen presentation to T cells. In the nonobese diabetic (NOD) mouse model, the Idd1 locus was shown to be the orthologous MHC gene I-Ab. Inheritance of susceptibility alleles at IDDM1/Idd1 is insufficient for disease development in humans and NOD mice. However, the identities and functions of the remaining diabetes loci (Idd2-Idd19 in NOD mice) are largely undefined. A crucial limitation in previous genetic linkage studies of this disease has been reliance on a single complex phenotype-diabetes that displays low penetrance and is of limited utility for high-resolution genetic mapping. Using the NOD model, we have identified an early step in diabetes pathogenesis that behaves as a highly penetrant trait. We report that NOD-derived alleles at both the Idd5 and Idd13 loci regulate a T lymphocyte-dependent progression from a benign to a destructive stage of insulitis. Human chromosomal regions orthologous to the Idd5 and -13 intervals are also linked to diabetes risk, suggesting that conserved genes encoded at these loci are central regulators of disease pathogenesis. These data are the first to reveal a role for individual non-MHC Idd loci in a specific, critical step in diabetes pathogenesis-T cell recruitment to islet lesions driving destructive inflammation. Importantly, identification of intermediate phenotypes in complex disease pathogenesis provides the tools required to progress toward gene identification at these loci.
Collapse
Affiliation(s)
- C J Fox
- Program in Developmental Biology, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | | | | | | |
Collapse
|
95
|
Kanagawa O, Xu G, Tevaarwerk A, Vaupel BA. Protection of nonobese diabetic mice from diabetes by gene(s) closely linked to IFN-gamma receptor loci. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 164:3919-23. [PMID: 10725755 DOI: 10.4049/jimmunol.164.7.3919] [Citation(s) in RCA: 87] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Nonobese diabetic (NOD) mice carrying a segment of chromosome flanking the disrupted IFN-gamma receptor gene from original 129 ES cells are resistant to development of diabetes. However, extended backcrossing of this mouse line to the NOD mouse resulted in a segregation of the IFN-gammaR-deficient genotype from the diabetes-resistant phenotype. These results indicate that the protection of NOD mice from the development of diabetes is not directly linked to the defective IFN-gamma receptor gene but, rather, is influenced by the presence of a diabetes-resistant gene(s) closely linked to the IFN-gammaR loci derived from the 129 mouse strain.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/pathology
- Diabetes Mellitus, Type 1/prevention & control
- Female
- Genetic Linkage/immunology
- Genetic Markers/immunology
- Genetic Predisposition to Disease/etiology
- Genetic Predisposition to Disease/genetics
- Genotype
- Histocompatibility Antigens Class I/biosynthesis
- Immunity, Innate/genetics
- Interferon-gamma/metabolism
- Interferon-gamma/pharmacology
- Islets of Langerhans/pathology
- Macrophages, Peritoneal/immunology
- Macrophages, Peritoneal/metabolism
- Mice
- Mice, Inbred NOD
- Mice, Knockout
- Receptors, Interferon/biosynthesis
- Receptors, Interferon/genetics
- Interferon gamma Receptor
Collapse
Affiliation(s)
- O Kanagawa
- Center for Immunology, Department of Pathology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | | | | | | |
Collapse
|
96
|
Litherland SA, Xie XT, Hutson AD, Wasserfall C, Whittaker DS, She JX, Hofig A, Dennis MA, Fuller K, Cook R, Schatz D, Moldawer LL, Clare-Salzler MJ. Aberrant prostaglandin synthase 2 expression defines an antigen-presenting cell defect for insulin-dependent diabetes mellitus. J Clin Invest 1999; 104:515-23. [PMID: 10449443 PMCID: PMC408519 DOI: 10.1172/jci4852] [Citation(s) in RCA: 83] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/1998] [Accepted: 07/13/1999] [Indexed: 12/21/2022] Open
Abstract
Prostaglandins (PGs) are lipid molecules that profoundly affect cellular processes including inflammation and immune response. Pathways contributing to PG output are highly regulated in antigen-presenting cells such as macrophages and monocytes, which produce large quantities of these molecules upon activation. In this report, we demonstrate aberrant constitutive expression of the normally inducible cyclooxygenase PG synthase 2 (PGS(2)/ COX-2) in nonactivated monocytes of humans with insulin-dependent diabetes mellitus (IDDM) and those with islet autoantibodies at increased risk of developing this disease. Constitutive PGS(2) appears to characterize a high risk for diabetes as it correlates with and predicts a low first-phase insulin response in autoantibody-positive subjects. Abnormal PGS(2) expression in at-risk subjects affected immune response in vitro, as the presence of a specific PGS(2) inhibitor, NS398, significantly increased IL-2 receptor alpha-chain (CD25) expression on phytohemagglutinin-stimulated T cells. The effect of PGS(2) on CD25 expression was most profound in subjects expressing both DR04 and DQbeta0302 high-risk alleles, suggesting that this cyclooxygenase interacts with diabetes-associated MHC class II antigens to limit T-cell activation. These results indicate that constitutive PGS(2) expression in monocytes defines an antigen-presenting cell defect affecting immune response, and that this expression is a novel cell-associated risk marker for IDDM.
Collapse
Affiliation(s)
- S A Litherland
- Department of Immunology, Pathology, and Laboratory Medicine, College of Medicine, College of Medicine, University of Florida, Gainesville 32610, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
97
|
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
|
98
|
André-Schmutz I, Hindelang C, Benoist C, Mathis D. Cellular and molecular changes accompanying the progression from insulitis to diabetes. Eur J Immunol 1999; 29:245-55. [PMID: 9933106 DOI: 10.1002/(sici)1521-4141(199901)29:01<245::aid-immu245>3.0.co;2-o] [Citation(s) in RCA: 68] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Insulin-dependent diabetes mellitus (IDDM) is not a disease of unbridled destruction. The autoimmune attack on pancreatic beta cells has two distinct stages - insulitis and diabetes - and progression of the former to the latter appears to be highly regulated. Identifying the factors controlling this transition has been difficult because it is a complex process that occurs non-universally and asynchronously. We have overcome these difficulties by coupling a simplified TCR transgenic (tg) model of IDDM and the immunosuppressive drug cyclophosphamide (CY). Young BDC2.5 TCR tg mice show insulitis but not diabetes; CY treatment provoked diabetes in 100% of animals with rapid, highly reproducible kinetics. This allowed a detailed temporal analysis of changes in cellular organization and cytokine gene expression within the lesion. The monokines IL-18, IL-12 and TNF-alpha were pivotal, their induction occurring almost immediately and their coordinate action being required for the onset of aggression. Other cytokines with direct toxicity for beta cells, including IL-1 -beta, IL-6 and IFN-gamma, were subsequently induced; in contrast, there was no cellular or molecular evidence of cell contact-mediated mechanisms of beta cell death.
Collapse
MESH Headings
- Animals
- Autoantigens
- Cyclophosphamide/toxicity
- Cytokines/biosynthesis
- Cytokines/genetics
- Diabetes Mellitus, Type 1/etiology
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/pathology
- Disease Models, Animal
- Humans
- Islets of Langerhans/drug effects
- Islets of Langerhans/immunology
- Islets of Langerhans/pathology
- Mice
- Mice, Inbred NOD
- Mice, Transgenic
- Microscopy, Electron
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, Antigen, T-Cell/genetics
Collapse
Affiliation(s)
- I André-Schmutz
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (CNRS/INSERM/ULP), Illkirch, France
| | | | | | | |
Collapse
|
99
|
Guerder S, Eynon EE, Flavell RA. Autoimmunity Without Diabetes in Transgenic Mice Expressing β Cell-Specific CD86, But Not CD80: Parameters that Trigger Progression to Diabetes. THE JOURNAL OF IMMUNOLOGY 1998. [DOI: 10.4049/jimmunol.161.5.2128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Abstract
To define more clearly the roles of CD80 (RIP-CD80) and CD86 (RIP-CD86) in the activation of autoreactive T cells in vivo, we generated transgenic mice expressing either or both costimulatory molecules on the β cells of the pancreas. While RIP-CD80 mice do not show any sign of autoimmunity, at the age of 7 mo RIP-CD86 transgenic mice develop a lymphoid infiltrate with both IFN-γ- and IL-4-positive cells in the vicinity of the islets; these mice, however, never progress to diabetes. This fundamental difference in the ability of CD80 and CD86 to activate self-reactive T cells in vivo is, however, obliterated when the level of TCR signaling is increased by either TNF-α or transgenic MHC class II expression. These results support the suggestion that CD80 and CD86 mainly differ at the level of the intensity of the signals they deliver.
Collapse
Affiliation(s)
| | | | - Richard A. Flavell
- *Section of Immunobiology and
- †Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06520
| |
Collapse
|
100
|
Robinson CP, Brayer J, Yamachika S, Esch TR, Peck AB, Stewart CA, Peen E, Jonsson R, Humphreys-Beher MG. Transfer of human serum IgG to nonobese diabetic Igmu null mice reveals a role for autoantibodies in the loss of secretory function of exocrine tissues in Sjögren's syndrome. Proc Natl Acad Sci U S A 1998; 95:7538-43. [PMID: 9636185 PMCID: PMC22675 DOI: 10.1073/pnas.95.13.7538] [Citation(s) in RCA: 173] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The NOD (nonobese diabetic) mouse has been studied as an animal model for autoimmune insulin-dependent diabetes and Sjögren's syndrome. NOD.Igmu null mice, which lack functional B lymphocytes, develop progressive histopathologic lesions of the submandibular and lachrymal glands similar to NOD mice, but in the absence of autoimmune insulitis and diabetes. Despite the focal appearance of T cells in salivary and lachrymal tissues, NOD.Igmu null mice fail to lose secretory function as determined by stimulation of the muscarinic/cholinergic receptor by the agonist pilocarpine, suggesting a role for B cell autoantibodies in mediating exocrine dryness. Infusion of purified serum IgG or F(ab')2 fragments from parental NOD mice or human primary Sjögren's syndrome patients, but not serum IgG from healthy controls, alters stimulated saliva production, an observation consistent with antibody binding to neural receptors. Furthermore, human patient IgG fractions competitively inhibited the binding of the muscarinic receptor agonist, [3H]quinuclidinyl benzilate, to salivary gland membranes. This autoantibody activity is lost after preadsorption with intact salivary cells. These findings indicate that autoantibodies play an important part in the functional impairment of secretory processes seen in connection with the autoimmune exocrinopathy of Sjögren's syndrome.
Collapse
Affiliation(s)
- C P Robinson
- Department of Oral Biology, University of Florida, Gainesville, FL 32610, USA
| | | | | | | | | | | | | | | | | |
Collapse
|