当前位置: 首页 > 期刊 > 《糖尿病学杂志》 > 2003年第2期 > 正文
编号:10584089
NOD Congenic Mice Genetically Protected From Autoimmune Diabetes Remain Resistant to Transplantation Tolerance Induction
http://www.100md.com 《糖尿病学杂志》2003年第2期
     1 Program in Immunology and Virology, the University of Massachusetts Medical School, Worcester, Massachusettsk*wm, 百拇医药

    2 Department of Medicine, the University of Massachusetts Medical School, Worcester, Massachusettsk*wm, 百拇医药

    3 Juvenile Diabetes Research Foundation/Wellcome Trust Diabetes and Inflammation Laboratory, University of Cambridge, Cambridge, U.Kk*wm, 百拇医药

    4 The Jackson Laboratory, Bar Harbor, Mainek*wm, 百拇医药

    5 Department of Pharmacology, Merck Research Laboratories, Rahway, New Jerseyk*wm, 百拇医药

    6 Department of Molecular Medicine, the University of Massachusetts Medical School, Worcester, Massachusettsk*wm, 百拇医药

    ABSTRACTk*wm, 百拇医药

    The loss of self-tolerance leading to autoimmune type 1 diabetes in the NOD mouse model involves at least 19 genetic loci. In addition to their genetic defects in self-tolerance, NOD mice resist peripheral transplantation tolerance induced by costimulation blockade using donor-specific transfusion and anti-CD154 antibody. Hypothesizing that these two abnormalities might be related, we investigated whether they could be uncoupled through a genetic approach. Diabetes-resistant NOD and C57BL/6 stocks congenic for various reciprocally introduced Idd loci were assessed for their ability to be tolerized. Surprisingly, in NOD congenic mice that are almost completely protected from diabetes, costimulation blockade failed to prolong skin allograft survival. In reciprocal C57BL/6 congenic mice with NOD-derived Idd loci, skin allograft survival was readily prolonged by costimulation blockade. These data indicate that single or multiple combinations of evaluated Idd loci that dramatically reduce diabetes frequency do not correct resistance to peripheral transplantation tolerance induced by costimulation blockade. We suggest that mechanisms controlling autoimmunity and transplantation tolerance in NOD mice are not completely overlapping and are potentially distinct, or that the genetic threshold for normalizing the transplantation tolerance defect is higher than that for preventing autoimmune diabetes.

    The nonobese diabetic (NOD) mouse is widely used to model human type 1 diabetes (1). Disease in NOD mice is characterized by T-cell–dependent autoimmune destruction of ß-cells (2). NOD mice exhibit a number of immune defects that may contribute to their expression of autoimmunity. These include defective macrophage maturation and function (3), low levels of natural killer (NK) cell activity (4), defects in NKT-cells (5,6), deficiencies in their regulatory CD4+CD25+ T-cell population (7), and the absence of C5a and hemolytic complement (8). Additionally, NOD mice are prone to development of other autoimmune syndromes, including autoimmune sialadenitis (9), autoimmune thyroiditis (10), experimental autoimmune encephalomyelitis (11), autoimmune peripheral polyneuropathy (12), and a systemic lupus erythematosus-like disease if exposed to killed mycobacterium (13).uf]u, 百拇医药

    Like humans, susceptibility to type 1 diabetes in the NOD mouse is a polygenic trait, involving at least 19 different Idd loci on 11 chromosomes (14,15). When these NOD genetic susceptibility intervals, either alone or in combination, are replaced by congenic introgression with the corresponding interval from a diabetes-resistant strain, the individual contribution of these Idd loci to potential autoimmune phenotypes can be determined. In many cases, congenic introgression of one or a few Idd loci from resistant strains onto the susceptible NOD strain greatly reduces the incidence of insulitis and diabetes (16–20).

    We have previously shown that treatment of normal mice with a single donor-specific transfusion (DST) plus a brief course of anti-CD154 (CD40L) monoclonal antibody (mAb) leads to permanent islet (21) and prolonged skin (22) allograft survival. However, transplantation in the NOD mouse presents a unique challenge. Although there are >125 protocols known to prevent diabetes in NOD mice (1), few tolerance-inducing interventions have been reported to be even partially effective in prolonging islet graft survival in diabetic NOD mice (23–26). For example, either anti-CD154 mAb monotherapy (27,28) or DST plus anti-CD154 mAb treatment (29), therapies that can induce permanent islet allograft survival in normal mice (30), fail to induce long-term islet graft survival in spontaneously diabetic NOD mice. Because our studies documented that DST and anti-CD154 mAb also failed to prolong the survival of skin allografts, a tissue that is not a target of the autoimmune response, we have hypothesized that NOD mice have a generalized defect in costimulation blockade–based transplantation tolerance induction (29). This defect in tolerance induction was independent of the unique NOD H2g7 haplotype, but it did correlate with a defect in antigen-presenting cell maturation that has previously been described in NOD mice (3,29).

    In the present study, we tested the hypothesis that the Idd loci that genetically control diabetes development in NOD mice would also control their resistance to transplantation tolerance. We tested this hypothesis using NOD and C57BL/6 mice with various Idd loci reciprocally introduced by congenic introgression onto each background. To avoid the confounding effects of simultaneously testing both tolerance induction and recurrent autoimmunity to islet-associated antigens, we used skin allografts for our experiments. Our data suggest that the genetic control of peripheral tolerance induction and autoimmunity differ, or are at least only partially overlapping, with the genes that control the induction of costimulation blockade–based peripheral transplantation tolerance.l^#4, 百拇医药

    RESEARCH DESIGN AND METHODSl^#4, 百拇医药

    Animals.l^#4, 百拇医药

    C57BL/6 (H2b), C3H/HeJ (H2k), and BALB/c (H2d) mice were obtained from the National Cancer Institute (Frederick, MD). NOD/MrkTacfBR, NOD.B6 Idd3R450, NOD.B10 Idd5R444, NOD.B6 Idd3R450 B10 Idd5R8, NOD.B10 Idd9R28, NOD.B6 Idd10Idd18R, and NOD.B6 Idd3Idd10Idd18R323 (all H2g7) were obtained from Taconic Farms (Germantown, NY). NOD/Lt (H2g7) mice were purchased from The Jackson Laboratory (Bar Harbor, ME). C57BL/6.NODc1c, C57BL/6.NODc1t, C57BL/6.NODc3, and C57BL/6.NODc6 mice (H2b; developed by Edward Wakeland, University of Texas Southwestern Medical Center, Dallas, TX) were the gift of Dr. Edward Leiter (The Jackson Laboratory). B10.D2-HC0 (genetic designation: B10.D2-H2d H2-T18c Hc0/oSnJ) and B10D2-HC1 mice (genetic designation: B10.D2-H2d H2-T18c Hc1/nSnJ) were obtained from The Jackson Laboratory. summarizes the congenic interval(s) and diabetes incidence of each of the congenic mouse strains listed above that were used in these experiments.

    fig.ommtted0, http://www.100md.com

    Idd congenic intervals, cumulative diabetes frequency and insulitis, and candidate genes in each interval0, http://www.100md.com

    All animals were certified to be free of Sendai virus, pneumonia virus of mice, murine hepatitis virus, minute virus of mice, ectromelia, lactate dehydrogenase–elevating virus, mouse poliovirus, Reo-3 virus, mouse adenovirus, lymphocytic choriomeningitis virus, polyoma, Mycoplasma pulmonis, and Encephalitozoon cuniculi. They were housed in a specific pathogen-free facility in microisolator cages and given autoclaved food and acidified water ad libitum. All animal use was in accordance with the guidelines of the institutional animal care and use committee of the University of Massachusetts Medical School and recommendations in the Guide for the Care and Use of Laboratory Animals (Institute of Laboratory Animal Resources, National Research Council, National Academy of Sciences, 1996).0, http://www.100md.com

    Tolerance induction and skin allograft transplantation.

    Graft recipient mice were treated with DST and anti-CD154 mAb and transplanted with skin allografts as described (22,31). Briefly, the DST consisted of a single intravenous injection of 107 C3H/HeJ or BALB/c female spleen cells obtained from 5- to 10-week-old donors. The DST was injected on day -7 relative to skin transplantation. Clone MR1 hamster anti-mouse CD154 mAb (32) was produced as ascites in scid mice and purified by protein A affinity chromatography (Amersham Pharmacia Biotech, Uppsala, Sweden). Antibody concentration was determined by measurement of optical density and confirmed by enzyme-linked immunosorbent assay (ELISA) as described previously (33). The concentration of contaminating endotoxin was determined commercially (Charles River Endosafe, Charleston, SC) and was uniformly <10 endotoxin units per milligram of mAb. Mice were injected intraperitoneally with anti-CD154 mAb (0.25 or 0.5 mg/dose as indicated in text) on days -7, -4, 0, and +4 relative to skin transplantation. Full-thickness skin grafts 1–2 cm in diameter were obtained from the flanks of donor mice and transplanted onto the dorsal flanks of recipients as described (22). Grafts were examined three times weekly, and rejection was defined as the first day on which the entire graft surface appeared necrotic (22). Grafts adherent to the bandage or fully necrotic on day 7 were deemed technical failures and were excluded from analysis (22).

    Serum levels of hamster anti-CD154 mAb..h9}, 百拇医药

    Clearance kinetics of the hamster MR1 anti-CD154 mAb was determined in groups of NOD and C57BL/6 mice that were given a single intraperitoneal injection of 2.0 mg of antibody. Serum was collected from blood obtained on days 13, 17, 20, 24, 32, and 40 after antibody injection. MR1 hamster IgG levels were quantified in the serum samples by ELISA (33)..h9}, 百拇医药

    Statistics..h9}, 百拇医药

    The average duration of graft survival is presented as the median. Graft survival among groups was compared using the method of Kaplan and Meier (34). The equality of allograft survival distributions for animals in different treatment groups was tested using the log rank statistic (34). P values <0.05 were considered statistically significant. Comparisons of two means used unpaired t tests (35). MR1 clearance rates were calculated by single-phase exponential decay curves fitted by regression analysis using Prism (Version 3.0; Graphpad Software, San Diego, CA).

    RESULTS*/wow6, 百拇医药

    Costimulation blockade prolongs skin allograft survival in mice genetically deficient in C5a and hemolytic complement.*/wow6, 百拇医药

    The NOD mouse has numerous immune defects that may be involved in their resistance to the induction of transplantation tolerance. One defect that could impact this strain’s response to in vivo antibody therapy is the absence of hemolytic complement caused by their genetic deficiency in C5a (8). To test this, we attempted to tolerize B10.D2 congenic mice that genetically differ in the presence or absence of C5a (36). These congenic mice were treated with DST, anti-CD154 mAb, and C3H/HeJ skin allografts. As shown in , the genetic absence of C5a in B10.D2-HC0 mice did not impair prolongation of skin allograft survival induced by costimulation blockade (median survival time [MST] = 54 days), and in fact, it slightly prolonged skin allograft survival as compared with similarly treated B10.D2-HC1 mice in which C5a was present (MST = 39 days, P < 0.01).

    fig.ommtted(+@6o, http://www.100md.com

    Skin allograft survival in mice genetically deficient in hemolytic complement. Skin allograft survival was determined in B10.D2 mice (H2d) treated with DST and anti-CD154 mAb that have C5a (HC1) or are genetically deficient in C5a (HC0) and hemolytic complement. Male mice 6–10 weeks of age were treated with four doses of 0.25 mg anti-CD154 mAb and a single DST and given a C3H/HeJ skin graft, as described in RESEARCH DESIGN AND METHODS. B10.D2 HC0 recipients genetically deficient in C5a had significantly prolonged skin allograft survival as compared with mice with C5a (P < 0.01).(+@6o, http://www.100md.com

    Clearance kinetics of anti-CD154 mAb are normal in NOD mice.(+@6o, http://www.100md.com

    In a related experiment, we sought to determine whether NOD mice are resistant to tolerance induction because of an accelerated clearance rate of anti-CD154 mAb from the circulation. We have previously documented that skin allograft rejection in tolerized mice is inversely correlated with the level of circulating anti-CD154 mAb (33). Groups of NOD and C57BL/6 mice were given a single intraperitoneal injection of 2.0 mg of anti-CD154 mAb. The concentration of MR1 in their serum was determined periodically between days 13 and 40 after injection to calculate the clearance rate of the antibody. The serum concentration of antibody on day 13 in NOD and C57BL/6 mice was ~ 300 mcg/ml and declined to 100 mcg/ml in both groups by day 40. The calculated half life of MR1 in NOD (10.7 days, n = 6) and C57BL/6 (10.4 days, n = 5) mice was similar. The serum concentrations at these time points after antibody injection and clearance rates of antibody are comparable to those we observed in CBA mice (33). These data document that neither the absence of hemolytic complement nor an accelerated clearance of anti-CD154 mAb in NOD mice accounts for the inability of costimulation blockade to prolong skin graft survival.

    Costimulation blockade prolongs skin allograft survival in C57BL/6 congenic mice with NOD susceptibility loci.$2z^{, http://www.100md.com

    We next tested the hypothesis that selected NOD-derived Idd susceptibility loci are responsible for the defect in peripheral transplantation tolerance induction by costimulation blockade. This was first assessed by determining whether the ability to prolong skin allograft survival was abrogated in C57BL/6 mice congenic for various NOD-derived Idd loci (22). None of the C57BL/6 congenic mice developed insulitis or diabetes . We tested four congenic strains. C57BL/6.NODc1c mice carry the NOD-derived Idd5.1 region, which includes genes encoding CD152 (CTLA4) and CD28, costimulatory molecules that are important for tolerance induction and immune activation (21). C57BL/6.NODc1t mice carry the NOD-derived Idd5.1 as well as the Idd5.2 region, which includes the CXCR2 and interleukin-8 receptor genes. C57BL/6.NODc3 mice carry the NOD-derived Idd3, Idd17, Idd10, and Idd18 loci, which include genes that control cell activation. C57BL/6.NODc6 mice carry the NOD Idd6 locus, which includes genes contained within the NK receptor protein-1a (NKR-P1) complex .

    The median skin allograft survival time of each of these C57BL/6 congenic strains ranged from 62 to >93 days and were statistically similar to that observed in C57BL/6 wild-type mice (MST = 81 to >94 days). Skin allograft survival in the C57BL/6 congenic mice was significantly greater than that observed in similarly treated NOD mice (MST = 20–24 days) .yi/fe-, 百拇医药

    fig.ommttedyi/fe-, 百拇医药

    Skin allograft survival on C57BL/6 mice congenic for the NOD Idd intervalsyi/fe-, 百拇医药

    fig.ommttedyi/fe-, 百拇医药

    Skin allograft survival on Idd3/Idd5 congenic NOD miceyi/fe-, 百拇医药

    fig.ommttedyi/fe-, 百拇医药

    Skin allograft survival on Idd3/Idd10/Idd18 and Idd9 congenic NOD miceyi/fe-, 百拇医药

    Diabetes-resistant NOD congenic mice treated with DST and anti-CD154 mAb rapidly reject skin allografts.yi/fe-, 百拇医药

    We next determined whether NOD stocks congenic for selected C57BL/6- or C57BL/10-derived Idd loci that mediate protection from insulitis and diabetes would also exhibit prolonged skin allograft survival after treatment with DST and anti-CD154 mAb. We tested three groups of NOD congenic mice with intervals that encompass the non–major histocompatibility complex (MHC) Idd loci with the greatest effect on diabetes expression (18,37–39). The first of these were NOD congenic stocks carrying a C57BL/6-derived Idd3 or C57BL/10-derived Idd5 locus alone or in combination. These NOD single-Idd congenic mice each have a reduced frequency of diabetes, and when the Idd3 and Idd5 resistance loci are combined, the frequency of diabetes is reduced to 1%, and insulitis is absent in most mice (18) . The median survival time of C3H/HeJ skin allografts in these NOD congenic mice treated with DST plus anti-CD154 mAb (24–31 days) was statistically significantly shorter than that of C57BL/6 mice (>115 days) . Skin allograft survival in these NOD congenic mice was similar to that of NOD/Lt (MST = 20 days) or NOD/MrkTac (MST = 23 days) mice .

    We next tested NOD congenic mice that carried various combinations of C57BL/6-derived Idd3, Idd10, and Idd18 resistance loci. The frequency of diabetes in these NOD congenic mice varies from 9 to 50% . A significant reduction in the frequency of insulitis was also observed in NOD.B6 Idd3/10/18 triple congenic mice (40,41). Again, all of the NOD congenic strains with various combinations of these genetic intervals were resistant to tolerance induction and had median skin allograft survival times of 17–29 days .x3y-, 百拇医药

    Finally, we tested an NOD stock congenic for the C57BL/10-derived Idd9 locus. This locus contains molecular variants of Cd30, Tnfr2, and Cd137 (39), and these mice have a frequency of diabetes of only 3% . The MST of skin allografts in NOD.B10 Idd9 mice was 20 days, similar to that observed in NOD/Lt mice (MST = 24 days, ).x3y-, 百拇医药

    DISCUSSIONx3y-, 百拇医药

    In the present study, we have investigated the relationship between genes that control autoimmune diabetes expression in NOD mice with those controlling their resistance to transplantation tolerance induced by costimulation blockade. We document that single or small combinations of the evaluated C57BL/6-derived Idd loci that dramatically alter diabetes expression are not able to correct the response of NOD mice to costimulation blockade and, conversely, that NOD-derived Idd loci do not shorten skin allograft survival in C57BL/6 mice. We further eliminate two other potential explanations: the genetic absence of hemolytic complement or an accelerated clearance rate of anti-CD154 mAb from the circulation.

    Peripheral transplantation tolerance induction by DST and anti-CD154 mAb involves the deletion of alloreactive CD8+ T-cells (31,33). Because CD8+ T-cells in NOD mice appear to be resistant to tolerance induction (42), this may be one mechanism by which NOD mice are resistant to costimulation blockade–induced tolerance. However, we have recently determined that the majority of high-affinity alloreactive CD4+ T-cells are also deleted by treatment with DST and anti-CD154 mAb (D.L.G., unpublished observations). CD4+ T-cells express CD154 when activated (32). This suggests that one mechanism by which DST and anti-CD154 mAb induces tolerance could involve deletion of alloreactive CD4+ T-cells by antibody-mediated, complement-dependent lysis. However, the ability to prolong skin allograft survival in congenic B10.D2 mice that lacked C5a and hemolytic complement argues that this defect does not prevent tolerance induction in NOD mice. We have also documented that the circulating level of anti-CD154 mAb is inversely correlated with skin allograft survival in recipients treated with DST and anti-CD154 mAb (33). However, the clearance rate of anti-CD154 mAb from the circulation of NOD mice was similar to that of C57BL/6 mice. This data suggests that rapid clearance of anti-CD154 mAb, hence potentially lowering anti-CD154 mAb concentrations below effective tolerizing levels (~ 100 mcg/ml) (33), was not the basis for the resistance of NOD mice to tolerance induction.

    We then tested the hypothesis that some of the genetic loci associated with development of autoimmune diabetes in NOD mice would also be important in their resistance to induction of peripheral transplantation tolerance. Prolonged allograft survival was not abrogated in C57BL/6 stocks congenic for any of the analyzed NOD-derived Idd susceptibility loci. We note, however, that no combination of Idd susceptibility loci introgressed into C57BL/6 mice to date has rendered them susceptible to the spontaneous development of insulitis or autoimmune diabetes.f, 百拇医药

    Similarly, none of the analyzed C57BL/6- or C57BL/10-derived Idd congenic intervals that confer various degrees of diabetes resistance to NOD mice restored their ability to be tolerized by DST and anti-CD154 mAb treatment. This observation was surprising because congenic introgression of even a few of the Idd resistance loci into NOD mice profoundly reduces the frequency of insulitis and diabetes (17,18,38,39) . Furthermore, many of the tested Idd congenic intervals are characterized by polymorphisms in genes important for costimulation and immune activation.

    There is evidence that autoimmune diabetes in NOD mice is due primarily to defects in central tolerance (43). Bone marrow chimerism is known to prevent autoimmunity in NOD mice by this mechanism (44). In humans, bone marrow cells from diabetic donors have been documented to adoptively transfer disease to nondiabetic recipients, suggesting that central tolerance defects are also important in type 1 diabetes in humans (45). Additionally, there is data to suggest that manipulation of the peripheral immune system can affect self-tolerance and the expression of autoimmune diabetes in NOD mice (46–48). Mechanisms that control central and peripheral tolerance are different. Central tolerance is primarily mediated by intrathymic deletion of autoreactive T-cells during thymic development, whereas peripheral tolerance is mediated by multiple mechanisms, including deletion, anergy, and regulatory processes (21).d\, http://www.100md.com

    It is currently unknown whether improved central or peripheral tolerance is the mechanism by which the NOD congenic mice we studied in these experiments were rendered resistant to autoimmune diabetes. Our data suggest, however, that if the mechanism of protection from diabetes is due to restoration of the factors that permit peripheral regulation of autoimmunity, these mechanisms are not sufficient for the induction of peripheral allogeneic transplantation tolerance by costimulation blockade. Alternatively, it is possible that the results from the current study reflect the fact that more Idd resistance loci are required to genetically alter the phenotype of abnormal transplantation tolerance induction in NOD mice than are required to decrease the incidence of spontaneous autoimmune diabetes. Specifically, peripheral transplantation tolerance induction by costimulation blockade may be under the control of a complex combination of Idd loci not yet tested. Future experiments examining C57BL/6.H2g7 congenic mice bearing NOD diabetes-susceptibility loci will be required to identify potential interactions of the unique NOD H2g7 MHC with these genetic loci and their effect on tolerance induction. However, these data raise the possibility that the loss of self-tolerance leading to autoimmunity in NOD mice may be mediated by mechanisms that differ, in part, from their resistance to peripheral transplantation tolerance, a hypothesis we are currently testing.

    ACKNOWLEDGMENTSh?34, http://www.100md.com

    This study was supported in part by grants AR35506 and AI42669 and an institutional Diabetes Endocrinology Research Center (DERC) grant DK52530 from the National Institutes of Health and by grant DK53006 jointly funded by the National Institutes of Health and the Juvenile Diabetes Research Foundation (JDRF). L.S.W. is supported by a joint grant from the JDRF and the Wellcome Trust. D.V.S. is supported by grants DK46266 and DK51090 from the National Institutes of Health and by a grant from the JDRF. The availability of NOD congenic mice through the Taconic Farms Emerging Models Program has been supported by grants from the Merck Genome Research Institute, National Institute of Allergy and Infectious Diseases (NIAID), and the JDRF.h?34, http://www.100md.com

    We thank Linda Paquin, Stephanie Gibbons, Linda Leehy, and Jean Leif for technical assistance.h?34, http://www.100md.com

    REFERENCESh?34, http://www.100md.com

    Atkinson M, Leiter EH: The NOD mouse model of insulin dependent diabetes: As good as it gets? Nature Med5 :601 –604,1999

    Leiter EH: NOD mice and related strains: origins, husbandry, and biology. In NOD Mice and Related Strains: Research Applications in Diabetes AIDS, Cancer and Other Diseases. Leiter EH, Atkinson MA, Eds. Austin, TX, R.G. Landes,1998 , p.1 –36*:q^%, 百拇医药

    Serreze DV, Gaedeke JW, Leiter EH: Hematopoietic stem-cell defects underlying abnormal macrophage development and maturation in NOD/Lt mice: defective regulation of cytokine receptors and protein kinase C. Proc Natl Acad Sci U S A90 :9625 –9629,1993*:q^%, 百拇医药

    Kataoka S, Satoh J, Fujiya H, Toyota T, Suzuki R, Itoh K, Kumagai K: Immunologic aspects of the nonobese diabetic (NOD) mouse: abnormalities of cellular immunity. Diabetes32 :247 –253,1983*:q^%, 百拇医药

    Naumov YN, Bahjat KS, Gausling R, Abraham R, Exley MA, Koezuka Y, Balk SB, Strominger JL, Clare-Salzler M, Wilson SB: Activation of CD1d-restricted T cells protects NOD mice from developing diabetes by regulating dendritic cell subsets. Proc Natl Acad Sci U S A98 :13838 –13843,2001*:q^%, 百拇医药

    Wang B, Geng YB, Wang CR: CD1-restricted NK T cells protect nonobese diabetic mice from developing diabetes. J Exp Med194 :313 –319,2001

    Salomon B, Lenschow DJ, Rhee L, Ashourian N, Singh B, Sharpe A, Bluestone JA: B7/CD28 costimulation is essential for the homeostasis of the CD4+CD25+ immunoregulatory T cells that control autoimmune diabetes. Immunity12 :431 –440,2000{, 百拇医药

    Baxter AG, Cooke A: Complement lytic activity has no role in the pathogenesis of autoimmune diabetes in NOD mice. Diabetes42 :1574 –1578,1993{, 百拇医药

    Hu Y, Nakagawa Y, Purushotham KR, Humphreys-Beher MG: Functional changes in salivary glands of autoimmune disease-prone NOD mice. Am J Physiol Endocrinol Metab263 :E607 –E614,1992{, 百拇医药

    Many MC, Maniratunga S, Denef JF: The non-obese diabetic (NOD) mouse: an animal model for autoimmune thyroiditis. Exp Clin Endocrinol Diabetes104 (Suppl. 3) :17 –20,1996{, 百拇医药

    Girvin AR, Dal Canto PC, Rhee L, Salomon B, Sharpe A, Bluestone JA, Miller SD: A critical role for B7/CD28 costimulation in experimental autoimmune encephalomyelitis: a comparative study using costimulatory molecule-deficient mice and monoclonal antibody blockade. J Immunol164 :136 –143,2000

    Salomon B, Rhee L, Bour-Jordan H, Hsin H, Montag A, Soliven B, Arcella J, Girvin AM, Miller SD, Bluestone JA: Development of spontaneous autoimmune peripheral polyneuropathy in B7-2-deficient NOD mice. J Exp Med194 :677 –684,2001g'}m., 百拇医药

    Silveira PA, Baxter AG: The NOD mouse as a model of SLE. Autoimmunity34 :53 –64,2001g'}m., 百拇医药

    Wicker LS, Todd JA, Peterson LB: Genetic control of autoimmune diabetes in the NOD mouse. Annu Rev Immunol13 :179 –200,1995g'}m., 百拇医药

    Serreze DV, Leiter EH: Genetic and pathogenic basis of autoimmune diabetes in NOD mice. Curr Opinion Immunol6 :900 –906,1994g'}m., 百拇医药

    Serreze DV, Bridgett M, Chapman HD, Chen E, Richard SD, Leiter EH: Subcongenic analysis of the Idd13 locus in NOD/Lt mice: evidence for several susceptibility genes including a possible diabetogenic role for beta2-microglobulin. J Immunol160 :1472 –1478,1998g'}m., 百拇医药

    Lyons PA, Armitage N, Lord CJ, Phillips MS, Todd JA, Peterson LB, Wicker LS: Mapping by genetic interaction: high-resolution congenic mapping of the type 1 diabetes loci Idd10 and Idd18 in the NOD mouse. Diabetes50 :2633 –2637,2001

    Hill NJ, Lyons PA, Armitage N, Todd JA, Wicker LS, Peterson LB: NOD Idd5 locus controls insulitis and diabetes and overlaps the orthologous CTLA4/IDDM12 and NRAMP1 loci in humans. Diabetes49 :1744 –1747,2000;7, 百拇医药

    Lamhamedi-Cherradi SE, Boulard O, Gonzalez C, Kassis N, Damotte D, Eloy L, Fluteau G, Lévi-Strauss M, Garchon HJ: Further mapping of the Idd5.1 locus for autoimmune diabetes in NOD mice. Diabetes50 :2874 –2878,2001;7, 百拇医药

    Caillat-Zucman S, Bach JF: Genetic predisposition to IDDM. Clin Rev Allergy Immunol19 :227 –246,2000;7, 百拇医药

    Rossini AA, Greiner DL, Mordes JP: Induction of immunological tolerance for transplantation. Physiol Rev79 :99 –141,1999;7, 百拇医药

    Markees TG, Phillips NE, Gordon EJ, Noelle RJ, Shultz LD, Mordes JP, Greiner DL, Rossini AA: Long-term survival of skin allografts induced by donor splenocytes and anti-CD154 antibody in thymectomized mice requires CD4+ T cells, interferon-gamma, and CTLA4. J Clin Invest101 :2446 –2455,1998;7, 百拇医药

    Sandberg JO, Eizirik DL, Sandler S: IL-1 receptor antagonist inhibits recurrence of disease after syngeneic pancreatic islet transplantation to spontaneously diabetic non-obese diabetic (NOD) mice. Clin Exp Immunol108 :314 –317,1997

    Rabinovitch A, Suarez-Pinzon WL, Shapiro AM, Rajotte RV, Power R: Combination therapy with sirolimus and interleukin-2 prevents spontaneous and recurrent autoimmune diabetes in NOD mice. Diabetes51 :638 –645,2002i, 百拇医药

    Faust A, Rothe H, Schade U, Lampeter E, Kolb H: Primary nonfunction of islet grafts in autoimmune diabetic nonobese diabetic mice is prevented by treatment with interleukin-4 and interleukin-10. Transplantation62 :648 –652,1996i, 百拇医药

    Atkinson MA: NOD mice as a model for therapeutic interventions in human insulin dependent diabetes mellitus. In NOD Mice and Related Strains: Research Applications in Diabetes, AIDS, Cancer and Other Diseases. Leiter EH, Atkinson MA, Eds. Austin, TX, R.G. Landes Company,1998 , p.145 –172i, 百拇医药

    Molano RD, Berney T, Li H, Cattan P, Pileggi A, Vizzardelli C, Kenyon NS, Ricordi C, Burkly LC, Inverardi L: Prolonged islet graft survival in NOD mice by blockade of the CD40-CD154 pathway of T-cell costimulation. Diabetes50 :270 –276,2001i, 百拇医药

    Molano RD, Berney T, Pileggi A, Ricordi C, Burkly L, Rothstein D, Basadonna G, Inverardi L: Prolonged survival of allogeneic islet grafts in NOD mice treated with a combination of anti-CD45RB and anti-CD154 antibodies. Transplant Proc33 :248 –249,2001

    Markees TG, Serreze DV, Phillips NE, Sorli CH, Noelle RJ, Woda BA, Greiner DL, Mordes JP, Rossini AA: NOD mice have a generalized defect in their response to transplantation tolerance induction. Diabetes48 :967 –974,19993w;m, 百拇医药

    Rossini AA, Mordes JP, Greiner DL, Stoff JS: Islet cell transplantation tolerance. Transplantation72 :S43 –S46,20013w;m, 百拇医药

    Iwakoshi NN, Mordes JP, Markees TG, Phillips NE, Greiner DL, Rossini AA: Treatment of allograft recipients with donor specific transfusion and anti-CD154 antibody leads to deletion of alloreactive CD8+ T cells and prolonged graft survival in a CTLA4-dependent manner. J Immunol164 :512 –521,20003w;m, 百拇医药

    Foy TM, Shepherd DM, Durie FH, Aruffo A, Ledbetter JA, Noelle RJ: In vivo CD40-gp39 interactions are essential for thymus-dependent humoral immunity. II. Prolonged suppression of the humoral immune response by an antibody to the ligand for CD40, gp39. J Exp Med178 :1567 –1575,19933w;m, 百拇医药

    Iwakoshi NN, Markees TG, Turgeon NA, Thornley T, Cuthbert A, Leif JH, Phillips NE, Mordes JP, Greiner DL, Rossini AA: Skin allograft maintenance in a new synchimeric model system of tolerance. J Immunol167 :6623 –6630,2001

    Kaplan EL, Meier P: Nonparametric estimation from incomplete observations. J Am Statist Assn53 :457 –481,1958k0r7krn, 百拇医药

    Glantz SA: Primer of Biostatistics. New York, McGraw-Hill,1981 , p. 352k0r7krn, 百拇医药

    Wheat WH, Wetsel R, Falus A, Tack BF, Strunk RC: The fifth component of complement (C5) in the mouse: analysis of the molecular basis for deficiency. J Exp Med165 :1442 –1447,1987k0r7krn, 百拇医药

    Lord CJ, Howlett S, Lyons PA, Peterson LB, Wicker LS, Todd JA: The murine type 1 diabetes loci, Idd1, Idd3, Idd5, Idd9, and idd17/10/18, do not control thymic CD4-CD8-/TCRß+ deficiency in the nonobese diabetic mouse. Mamm Genome12 :175 –176,2001k0r7krn, 百拇医药

    Lyons PA, Armitage N, Argentina F, Denny P, Hill NJ, Lord CJ, Wilusz MB, Peterson LB, Wicker LS, Todd JA: Congenic mapping of the type 1 diabetes locus, ldd3, to a 780-kb region of mouse chromosome 3: identification of a candidate segment of ancestral DNA by haplotype mapping. Genome Res10 :446 –453,2000k0r7krn, 百拇医药

    Lyons PA, Hancock WW, Denny P, Lord CJ, Hill NJ, Armitage N, Siegmund T, Todd JA, Phillips MS, Hess JF, Chen SL, Fischer PA, Peterson LB, Wicker LS: The NOD Idd9 genetic interval influences the pathogenicity of insulitis and contains molecular variants of Cd30, Tnfr2, and Cd137. Immunity13 :107 –115,2000

    Podolin PL, Denny P, Lord CJ, Hill NJ, Todd JA, Peterson LB, Wicker LS, Lyons PA: Congenic mapping of the insulin-dependent diabetes (Idd) gene, Idd10, localizes two gene mediating the Idd10 effect and eliminates the candidate Fcgr1. J Immunol159 :1835 –1843,1997mn, 百拇医药

    Wicker LS, Todd JA, Prins J-B, Podolin PL, Renjilian RJ, Peterson LB: Resistance alleles at two non-major histocompatibility complex-linked insulin-dependent diabetes loci on chromosome 3, Idd3 and Idd10, protect nonobese diabetic mice from diabetes. J Exp Med180 :1705 –1713,1994mn, 百拇医药

    Kreuwel HTC, Biggs JA, Pilip IM, Pamer EG, Lo D, Sherman LA: Defective CD8+ T cell peripheral tolerance in nonobese diabetic mice. J Immunol167 :1112 –1117,2001mn, 百拇医药

    Serreze DV, Leiter EH: Insulin dependent diabetes mellitus (IDDM) in NOD mice and BB rats: origins in hematopoietic stem cell defects and implications for therapy. In Lessons from Animal Diabetes V. Shafrir E, Ed. London, Smith Gordon,1994 , p.59 –73mn, 百拇医药

    Leiter EH, Serreze DV: Autoimmune diabetes in the nonobese diabetic mouse: suppression of immune defects by bone marrow transplantation and implications for therapy. Clin Immunol Immunopathol59 :323 –334,1991

    Sorli CH, Greiner DL, Mordes JP, Rossini AA: Stem cell transplantation for treatment of autoimmune diseases. Graft1 :71 –81,1998tv8h#|!, http://www.100md.com

    Rosmalen JG, van Ewijk W, Leenen PJ: T-cell education in autoimmune diabetes: teachers and students. Trends Immunol23 :40 –46,2002tv8h#|!, http://www.100md.com

    Greiner DL, Rossini AA, Mordes JP: Translating data from animal models into methods for preventing human autoimmune diabetes mellitus: caveat emptor and primum non nocere. Clin Immunol100 :134 –143,2001tv8h#|!, http://www.100md.com

    Atkinson MA: Mechanisms underlying the loss of self tolerance in NOD mice. Res Immunol148 :301 –306,1997tv8h#|!, http://www.100md.com

    Yui MA, Muralidharan K, Moreno-Altamirano B, Perrin G, Chestnut K, Wakeland EK: Production of congenic mouse strains carrying NOD-derived diabetogenic genetic intervals: an approach for the genetic dissection of complex traits. Mamm Genome7 :331 –334,1996tv8h#|!, http://www.100md.com

    Podolin PL, Denny P, Armitage N, Lord CJ, Hill NJ, Levy ER, Peterson LB, Todd JA, Wicker LS, Lyons PA: Localization of two insulin-dependent diabetes (Idd) genes to the Idd10 region on mouse chromosome 3. Mamm Genome9 :283 –286,1998(Todd Pearson Thomas G. Markees Linda S. WickerDavid V. Serreze Laurence B. Peterson John P. Mordes A)