当前位置: 首页 > 医学版 > 期刊论文 > 内科学 > 糖尿病学杂志 > 2005年 > 第1期 > 正文
编号:11256194
Autoimmune Diabetes and Resistance to Xenograft Transplantation Tolerance in NOD Mice
     1 Department of Medicine, Diabetes Division, University of Massachusetts Medical School, Worcester, Massachusetts

    2 Juvenile Diabetes Research Foundation/Wellcome Trust, Diabetes and Inflammation Laboratory, University of Cambridge, Cambridge, U.K

    3 Department of Pharmacology, Merck Research Laboratories, Rahway, New Jersey

    4 The Jackson Laboratory, Bar Harbor, Maine

    ABSTRACT

    Costimulation blockade induces prolonged rat islet and skin xenograft survival in C57BL/6 mice. Nonobese diabetic (NOD) mice, which are used to model human autoimmune diabetes, are resistant to costimulation blockade-induced allograft tolerance. We tested the hypothesis that NOD mice would also be resistant to costimulation blockade-induced rat xenograft tolerance. We report that rat islet xenograft survival is short in spontaneously diabetic NOD mice treated with a tolerizing regimen of donor-specific transfusion and anti-CD154 antibody. Rat islet xenograft survival is only marginally longer in chemically diabetic NOD mice treated with costimulation blockade but is prolonged further in NOD Idd congenic mice bearing C57-derived chromosome 3 loci. Reciprocally, the presence of NOD-derived chromosome 3 loci shortens islet xenograft survival in tolerized C57BL/6 mice. Islet xenograft survival is longer in tolerized NOD.CD4aeC/eC and (NOD x C57BL/6)F1 mice than in NOD mice but still much shorter than in C57BL/6 mice. Skin xenograft survival in (NOD x C57BL/6)F1 mice treated with costimulation blockade is short, suggesting a strong genetic resistance to skin xenograft tolerance induction. We conclude that the resistance of NOD mice to xenograft tolerance induction involves some mechanisms that also participate in the expression of autoimmunity and other mechanisms that are distinct.

    The NOD mouse is an excellent model of human type 1 diabetes (1,2). It develops insulitis and diabetes that are T-cell dependent, with both the CD4+ and CD8+ T-cell subsets contributing to disease (1,2). In addition to targeted destruction of pancreatic -cells (3,4), NOD mice also exhibit numerous defects in their immune system (2). Their CD8+ T-cells have been reported to be resistant to the induction of antigen-induced apoptosis (5), and they are susceptible to other intercurrent autoimmune diseases including thyroiditis and sialadenitis (2).

    Susceptibility to diabetes in both humans and NOD mice is genetically complex (6,7). In NOD mice, as many as 27 Idd loci on 11 different chromosomes have been implicated in disease susceptibility or resistance (8,9). To study the role of these loci on the development of diabetes, investigators have used congenic introgression to replace loci in NOD mice with loci derived from disease-resistant strains. Congenic NOD mice bearing Idd resistance loci exhibit varying degrees of protection from insulitis and diabetes (10eC13).

    We have previously documented that in addition to autoimmunity, NOD mice have an inherent genetic resistance to allograft tolerance induced by a costimulation blockade protocol consisting of a donor-specific transfusion (DST) and anti-CD154 monoclonal antibody (mAb) (14). The resistance of NOD mice to skin allograft tolerance was determined to be a dominant genetic trait because it also characterizes (NOD x C57BL/6)F1 mice (15). In contrast, the resistance of NOD mice to islet allograft tolerance induced by costimulation blockade is a genetically recessive trait that appears to be controlled in part by loci on chromosome 3 (16).

    Because the availability of allogeneic islets for human transplantation is limited, there is a need to investigate the potential of tolerance induction procedures to prolong islet xenograft survival. We have previously shown that treatment of chemically diabetic C57BL/6 mice with DST and anti-CD154 mAb leads to prolonged survival of rat islet and skin xenografts (17,18) and xenogeneic neonatal porcine islet cell clusters (19). Similarly, anti-CD45RB mAb (20), anti-CD4 mAb (21), or coadministration of CTLA4-Fc and anti-CD154 mAb (22,23) leads to prolonged islet xenograft survival in mice. Islet xenograft tolerance has also been reported in monkeys given anti-CD3 immunotoxin, cyclosporine, and steroids (24).

    Here, we tested the efficacy of our xenograft tolerance induction protocol based on DST and anti-CD154 mAb in NOD, NOD congenic, knockout, and (NOD x C57BL/6)F1 mice. We tested the hypothesis that genetic elements that contribute to diabetes susceptibility in NOD mice also participate in the control of islet and skin xenograft survival induced by costimulation blockade.

    RESEARCH DESIGN AND METHODS

    C57BL/6 (H2b) mice were obtained from the National Cancer Institute (Frederick, MD) or from The Jackson Laboratory (Bar Harbor, ME). NOD/MrkTacfBR (H2g7), NOD.B6 Idd3/Idd10/Idd18 R323 (H2g7, line 1538 at Taconic), and NOD.B10 Idd9 (H2g7, line 1104 at Taconic) mice were obtained from Taconic Farms (Germantown, NY). Breeding pairs of B6.NOD.C3 (H2b) were obtained from Dr. Ed Leiter at The Jackson Laboratory and bred in our facility (25). NOD.CD8aeC/eC (H2g7) mice (official designation: NOD.1029S2(B6)-Cd8atm1Mak/Dvs) and NOD.CD4aeC/eC (H2g7) mice (official designation: NOD.129S2(B6)-Cd4atm1Knw/Dvs) were obtained from a breeding colony maintained by one of the authors (D.V.S.) at The Jackson Laboratory. The latter two congenic stocks of NOD mice are homozygous for linkage markers delineating all known Idd loci of NOD origin (26eC28). NOD.CD8aeC/eC mice (1/17 females diabetic by age 30 weeks) (15) and NOD.CD4aeC/eC mice (0 of 18 females diabetic by age 30 weeks) (28) are both strongly diabetes resistant. LEW rats (RT1l) were obtained from Harlan Sprague Dawley (Indianapolis, IN).

    All animals were housed under specific pathogen-free conditions in sterile cages with microisolator lids and given autoclaved food and water ad libitum. Rats and mice used in these studies were certified to be serologically free of Sendai virus, pneumonia virus of mice, sialodacryoadenitis virus, rat corona virus, Kilham rat virus, H1 (Toolan’s virus), GD7, Reo-3, lymphocytic choriomeningitis virus, mouse adenovirus, Hantaan virus, Mycoplasma pulmonis, and encephalitozoon cuniculi. All animals were maintained in accordance with recommendations in the U.S. Government Principles for the Utilization and Care of Vertebrate Animals used in Testing, Research, and Training: the Guide for the Care and Use of Laboratory Animals (29) and local institutional guidelines.

    Pancreatic islet transplantation.

    Spontaneously diabetic NOD mice were 4eC5 months old at the time of grafting. C57BL/6, NOD, and congenic strain recipient mice at 8eC10 weeks of age were rendered hyperglycemic by a single intraperitoneal injection of 150 or 160 mg/kg of streptozotocin. Plasma glucose was measured using a Beckman II glucose analyzer (Beckman, Fullerton, CA). The diagnosis of diabetes in graft recipients was established by the observation of a plasma glucose concentration >250 mg/dl on at least two different days before transplantation. Pancreatic islets were harvested from donor rats by collagenase digestion (30) and transplanted at a dose of 15eC20/kg body wt into the renal subcapsular space of the recipient. Islet donors were retired breeder male LEW rats. Grafts were judged to have been rejected after the plasma glucose concentration rose to >250 mg/dl on 2 successive days after a minimum of 2 days of normoglycemia. Only one animal, an (NOD x C57BL/6)F1 recipient, never became normoglycemic and was excluded from the analysis as a technical failure. The rate of primary islet xenograft nonfunction among all transplanted streptozotocin-induced diabetic mice (Figs. 1 and 2) was 0.7% (1 of 149). To confirm graft function in recipients that were normoglycemic at the end of the period of observation, unilateral nephrectomy of the graft-bearing kidney was performed; grafts were judged to have been functional if the plasma glucose concentration rose to >250 mg/dl or, in a few instances, by histological documentation of an intact graft (see below).

    Skin transplantation.

    Skin graft recipients were male C57BL/6 and NOD mice or male or female congenic mouse strains as described above transplanted at 10 weeks of age. Full-thickness skin specimens for transplantation were obtained from 3- to 6-week-old female LEW rats. Donor skin was prepared and transplanted as described (31). Skin graft survival was assessed three times weekly by visual and tactile examination. Time of rejection was defined as the first day on which the entire epidermal surface of the graft was necrotic.

    Treatment of islet and skin xenograft recipients with DST and anti-CD154 mAb.

    LEW rats were killed in an atmosphere of 100% CO2. Spleens were removed, mechanically dispersed in sterile medium (RPMI-1640), washed, and counted using a hemocytometer. Cells were assayed for viability using the method of Trypan blue exclusion, and in all cases viability was >90%. Purified hamster anti-mouse CD154 mAb (clone MR1) was produced by the National Cell Culture Center (Minneapolis, MN) and affinity purified in our laboratory (32). The concentration of contaminating endotoxin was determined commercially (Charles River Endosafe, Charleston, SC) and was uniformly <10 units/mg (15). Anti-CD154 mAb was diluted to a concentration of 1 mg/ml and administered at doses of 0.25 or 0.5 mg. Islet and skin xenograft recipients received either no treatment, a single course of anti-CD154 mAb, or combined therapy with both reagents as previously described (17). DSTs consisted of 10 x 106 viable LEW spleen cells in a volume of 0.20eC0.4 ml given via the tail vein 7 days before grafting. Courses of anti-CD154 mAb comprising four doses were given intraperitoneally twice weekly beginning on the day of spleen cell injection.

    Histology.

    Islet graft-bearing kidneys were fixed in 10% neutral buffered formalin. Paraffin-embedded sections were prepared and stained with hematoxylin and eosin; additional sections were stained immunohistochemically for the presence of insulin. A qualified pathologist (Michel C. Appel), who was unaware of the treatment status of the donors, evaluated all islet graft specimens for the presence of inflammatory cells and insulin. Because no skin grafts survived indefinitely, none were studied histologically.

    Statistics.

    The average duration of graft survival is presented as the median. Statistical comparisons of graft survival among groups were performed using the method of Kaplan and Meier (33); the equality of xenograft survival distributions for animals in different treatment groups was tested using the log- rank statistic (34). P values <0.05 were considered statistically significant.

    RESULTS

    Rat islet xenograft survival in NOD mice

    Rat islet xenograft survival in spontaneously diabetic NOD mice.

    We first transplanted LEW rat islets into spontaneously diabetic NOD mice. As expected, in otherwise untreated diabetic NOD mice, graft rejection was rapid; the median survival time (MST) was 8 days (group 1) (Table 1). Similarly, islet xenograft survival was short in spontaneously diabetic NOD recipients treated with anti-CD154 mAb monotherapy (group 2, MST = 8 days). We next treated diabetic NOD mice with a donor-DST plus anti-CD154 mAb, a protocol that induces prolonged islet xenograft survival in chemically diabetic C57BL/6 mice (17). Treatment with DST plus anti-CD154 mAb at doses of 0.25 mg (group 3, MST = 10 days) or 0.5 mg (group 4, MST = 13 days) failed to prolong islet graft survival.

    Islet xenograft survival in chemically diabetic NOD mice.

    NOD mice have recently been shown to resist allotransplantation tolerance based on costimulation blockade; this resistance was observed to apply to both skin and islet allografts and could be separated genetically from the phenotype of autoimmunity (14,15). To begin to discriminate among potential mechanisms underlying the short islet xenograft survival in autoimmune diabetic NOD mice, we next transplanted rat islet xenografts into chemically diabetic NOD recipients.

    LEW rat islet xenograft survival, as expected, was short in otherwise untreated chemically diabetic NOD (MST = 12 days) and C57BL/6 (MST = 10 days; Fig. 1C) recipients. Treatment with anti-CD154 mAb monotherapy significantly prolonged rat islet xenograft survival in NOD mice (MST = 22 days; P < 0.025 vs. untreated controls) and to an even greater extent in C57BL/6 mice (MST = 32 days; Fig. 1B; P < 0.001 vs. untreated C57BL/6 controls and P < 0.05 vs. treated NOD mice) mice. Treatment with both DST and anti-CD154 mAb monotherapy further prolonged rat islet xenograft survival in both NOD (MST = 32 days; P < 0.01) and C57BL/6 (MST = 116 days; Fig. 1A; P < 0.001) mice. Again, graft survival in these C57BL/6 recipients was significantly longer than that observed in the similarly treated NOD mice (P < 0.001).

    Rat islet survival in Idd congenic mice

    NOD.Idd congenic mice.

    To investigate the genetic basis for the poor survival of islet xenografts in NOD mice following costimulation blockade, we studied Idd congenic NOD mice. We first asked if C57-derived Idd diabetes-resistant loci that reduce the frequency of insulitis and diabetes in NOD mice would restore susceptibility to the induction of islet xenograft tolerance. Two groups of NOD congenic mice were tested. The first was an NOD congenic stock (NOD.Idd3/10/18) carrying C57BL/6-derived Idd3, Idd10, and Idd18 resistance loci. The incidence of diabetes at 7 months of age in NOD.Idd3/10/18 congenic mice ranges from 3 to 9% (10,35,36). Duration of rat islet xenograft survival was significantly longer in NOD.Idd3/10/18 congenic mice (MST = 58 days; Fig. 1A) treated with costimulation blockade than in similarly treated NOD mice (P < 0.001). However, graft survival in the congenic mice was again significantly shorter than that achieved in C57BL/6 mice (P < 0.001). As expected, rat islet xenografts were rapidly rejected in otherwise untreated diabetic NOD.Idd3/10/18 congenic mice (MST = 11 days; Fig. 1C).

    We next studied a NOD stock congenic for the C57BL/10-derived Idd9 locus (37). The incidence of diabetes in these mice is only 3%. The median survival time of rat islet xenografts in NOD.B10 Idd9 mice treated with DST and anti-CD154 mAb was only 31 days (Fig. 1A) and was statistically similar to that observed in NOD mice (P = NS). Rat islet xenografts, as expected, were rapidly rejected in otherwise untreated diabetic NOD.B10 Idd9 mice (MST = 13 days; Fig. 1C).

    C57BL/6.Idd congenic mice.

    Rat islet xenograft survival is prolonged in NOD mice bearing segments of chromosome 3 derived from diabetes-resistant C57BL/10 mice. To confirm the contribution of loci on chromosome 3 to prolonged islet xenograft survival, we next studied C57BL/6.NOD.C3 mice. These mice carry the diabetes-susceptible NOD-derived Idd3, Idd17, Idd10, and Idd18 alleles on chromosome 3 (25). The median survival time of rat islet xenografts in C57BL/6.NOD.C3 mice treated with DST and anti-CD154 mAb was 63 days (Fig. 2A). This was significantly shorter than that achieved in similarly treated diabetic C57BL/6 (MST >168 days; Fig. 2A; P < 0.001). As expected, islet xenograft survival was brief in otherwise untreated diabetic C57BL/6.NOD.C3 mice (MST = 9 days; Fig. 2B). These data support our finding of an important role of congenic intervals on chromosome 3 in the control of the induction of tolerance to rat islet xenografts in NOD mice.

    Cellular mechanisms underlying NOD resistance to rat xenograft survival in NOD mice following costimulation blockade

    Islet xenograft survival in NOD.CD4aeC/eC mice.

    Survival of rat xenografts in C57BL/6 mice treated with DST and anti-CD154 mAb can be further prolonged by concurrent treatment with a depleting anti-CD4 mAb (18). We next hypothesized that genetic deletion of CD4+ T-cells would facilitate islet xenograft survival in NOD recipients. Islet xenograft survival in NOD.CD4aeC/eC mice treated with DST and anti-CD154 mAb was significantly longer (MST = 73 days) than in similarly treated wild-type NOD mice (Fig. 2A; P < 0.01). Islet xenograft survival in otherwise untreated NOD.CD4aeC/eC mice (MST = 32 days; Fig. 2B) was also longer than in untreated NOD recipients (P < 0.025).

    Islet xenograft survival in NOD.CD8aeC/eC mice.

    Allograft survival in mice treated with costimulation blockade depends in part on the deletion of CD8+ T-cells (38eC42), and CD8+ T-cells in NOD mice reportedly resist the induction of peripheral tolerance (5). To determine if the resistance of NOD mice to xenograft tolerance based on costimulation blockade involves the failure to delete CD8+ T-cells, we next studied NOD.CD8aeC/eC mice. Rat islet xenograft survival in NOD.CD8aeC/eC mice treated with DST plus anti-CD154 mAb (MST = 49; Fig. 2A) was not significantly different than that achieved in similarly treated NOD mice (P = NS).

    Islet xenograft survival in (NOD x C57BL/6)F1 mice.

    These data suggest that resistance to rat islet xenograft tolerance is partially overcome in NOD congenic mice that bear specific Idd resistance loci on chromosome 3 or lack CD4+ T- cells. However, the presence of this group of Idd resistance loci does not prolong islet xenograft survival to the extent that can be achieved in C57BL/6 mice. Most NOD Idd susceptibility loci are recessive (7). (NOD x C57BL/6)F1 mice are heterozygous at all Idd loci and thus bear a large number of C57BL/6-derived diabetes resistance alleles. (NOD x C57BL/6)F1 mice are completely protected from autoimmune diabetes (43), but surprisingly they remain resistant to the induction of skin allograft tolerance by costimulation blockade (15). To determine if they would similarly resist xenograft tolerance induction, chemically diabetic (NOD x C57BL/6)F1 mice were treated with DST and anti-CD154 mAb and transplanted with rat islets. Median duration of graft survival was 83 days (Fig. 2A), an outcome significantly superior to than that achieved in NOD (P < 0.001) mice but inferior to that achieved in C57BL/6 (P < 0.001) mice.

    Rat skin xenograft survival in (NOD x C57BL/6)F1 mice.

    The dominant resistance to allograft tolerance induction in (NOD x C57BL/6)F1 mice was documented in studies using skin not islet grafts (15). We therefore tested the hypothesis that skin xenograft survival (NOD x C57BL/6)F1 mice treated with DST and anti-CD154 mAb would be short.

    As expected, rat skin xenografts were rapidly rejected by untreated C57BL/6 (MST = 7 days), NOD (MST = 10 days), and (NOD x C57BL/6)F1 (MST = 7 days; Fig. 3B) mice. Again, as expected, rat skin xenograft survival was prolonged in C57BL/6 mice treated with DST plus anti-CD154 mAb (MST = 59 days; Fig. 3A). Duration of skin xenograft survival in similarly treated (NOD x C57BL/6)F1 mice was significantly shorter (MST = 18 days; P < 0.001) and statistically similar to that observed in similarly treated NOD mice (MST = 14 days; Fig. 3A; P = NS).

    Rat skin xenograft survival in NOD.CD4aeC/eC and NOD.CD8aeC/eC mice.

    We next investigated the role of CD4+ and CD8+ cells in the survival of rat skin xenografts on NOD mice treated with costimulation blockade. Skin xenograft survival in NOD.CD4aeC/eC mice treated with DST and anti-CD154 mAb (MST = 51 days) was slightly shorter than that observed in C57BL/6 mice (P < 0.05) but much longer than that observed in NOD mice (P < 0.001; Fig. 3A). In contrast, skin xenograft survival in NOD.CD8aeC/eC mice was significantly shorter (MST = 18 days) than that observed in C57BL/6 mice (P < 0.001; Fig. 3A) and statistically similar to that observed in NOD mice (P = NS). Skin xenograft survival in untreated NOD.CD4aeC/eC (MST = 11 days) and NOD.CD8aeC/eC (MST = 18 days; Fig. 3B) mice was short.

    DISCUSSION

    These data document that costimulation blockade is ineffective in prolonging islet xenograft survival in NOD mice. This was true of both spontaneously diabetic NOD mice and NOD mice with chemically induced hyperglycemia. To determine if the ineffectiveness of costimulation blockade was due to recurrent autoimmunity or to a generalized resistance of NOD mice to tolerance induction (14,15,44), we used congenic NOD mice and (NOD x C57BL/6)F1 mice. Our data revealed that islet xenograft tolerance induction is regulated in part by Idd loci on chromosome 3 and involves CD4+ T-cells. We observed that islet xenograft survival is prolonged in (NOD x C57BL/6)F1 mice but remains much shorter than that observed in C57BL/6 mice. These observations were unexpected given the fact that islet allograft survival in normal mice requires the presence of CD4+ T-cells (31) and is permanent in (NOD x C57BL/6)F1 mice treated with costimulation blockade (16). These data in a xenograft model document that important differences exist in mechanisms that control the resistance of NOD mice to the induction of allograft versus xenograft tolerance.

    In contrast to the survival of islet allografts in (NOD x C57BL/6)F1 mice treated with costimulation blockade (16), which is permanent, islet xenograft survival is these animals is prolonged but not permanent. These results suggest that in contrast to islet allograft tolerance induction (15), resistance to islet xenograft tolerance induction in NOD mice is a genetically dominant trait. When applied to skin xenografts, costimulation blockade is not effective in prolonging graft survival in either NOD or (NOD x C57BL/6)F1 mice. These results suggest that, as is true of their resistance to skin allograft tolerance induction (15), resistance to skin xenograft tolerance induction in NOD mice is also a genetically dominant trait.

    Our data document that islet xenograft survival in autoimmune diabetic NOD mice treated with costimulation blockade is short. This could result from either autoimmune recurrence or a generalized resistance of NOD mice to xenotolerance induction by costimulation blockade. We have previously shown that in chemically diabetic NOD congenic mice and (NOD x C57BL/6)F1 mice, the phenotypes of autoimmune diabetes and transplantation tolerance are not under completely overlapping genetic control (14eC16,45). In those studies, we observed no association of Idd loci with skin allograft tolerance induction (14), and, surprisingly, we found that skin allograft survival was short even in (NOD x C57BL/6)F1 mice (15). However, when islet allografts were tested, permanent allograft survival was observed in (NOD x C57BL/6)F1 mice (16). That study further documented that Idd loci on chromosome 3 were, at least in part, responsible for regulation of islet allograft survival in NOD mice treated with costimulation blockade (16).

    To determine if Idd loci control rat islet xenograft survival in chemically diabetic NOD mice treated with costimulation blockade, we tested three congenic strains. In NOD.B6 Idd3/10/18 congenic mice, the cumulative frequency of diabetes at 7 months of age is 3eC9% (35,36), yet these congenic mice exhibited only slightly prolonged xenograft survival compared with NOD mice. More strikingly, NOD.B10 Idd9 mice, in which the cumulative frequency of diabetes at 7 months of age is only 3% (37), did not exhibit prolonged islet xenograft survival compared with NOD mice. These data suggest that genetic control of autoimmunity and tolerance to islet xenografts are partially distinct.

    To confirm the importance of chromosome 3 loci as a determinant of islet xenograft survival, which could not be assumed a priori, we tested C57BL/6.NOD.c3 congenic mice. These mice bear a large segment of NOD chromosome 3 that includes Idd3, Idd17, Idd10, and Idd18 (25). Of particular interest is Idd3, which has been narrowed to a small interval containing eight genes. Three of these genes, Il2, Il21, and Fgf2, have known functions that could contribute to the establishment of immunological tolerance (46 and L.S.W., L.B.P., unpublished observations). Because Idd3 has been shown to be important in the induction of islet allograft tolerance by costimulation blockade (16), we are currently investigating if genes within the Idd3 region are responsible for the effect of chromosome 3 on islet xenograft tolerance induction.

    The results of our studies using congenic mice document that genetic reduction of autoimmunity in NOD mice does not invariably lead to prolonged islet xenograft survival after costimulation blockade. Even more striking was our discovery that islet xenograft survival in (NOD x C57BL/6)F1 mice treated with costimulatory blockade, although prolonged relative to that observed in NOD mice, was much shorter than that achieved in C57BL/6 mice. In (NOD x C57BL/6)F1 mice treated with DST and anti-CD154 mAb, islet allograft is permanent (16). In their aggregate, these data imply that multiple genes must control islet xenograft survival in mice treated with costimulation blockade. The data further suggest that the genetic mechanisms that control islet allograft and xenograft survival are only partially overlapping. This interpretation is supported by our analysis of the cell subsets important in islet xenograft survival. NOD.CD4aeC/eC mice treated with costimulation blockade exhibit prolonged islet xenograft survival whereas anti-CD4 mAb treatment completely abrogates islet allograft survival, even in normal mice (47).

    Skin allograft tolerance, however, appears to be controlled by mechanisms different from those important for islet allograft tolerance. In a complete CD40 knockout model system, islet allograft survival was prolonged whereas skin allograft survival was short (48). More recently, we have found that depletion of CD25+ cells shortens skin allograft survival whereas it has minimal effects on islet allograft survival (47). We speculate that skin allograft survival is more dependent on the presence of regulatory T-cells than is islet allograft survival, a hypothesis currently under investigation.

    Based on the published experience with allografts, it was not surprising that skin xenograft survival in NOD mice treated with costimulation blockade appears to be controlled at least in part by mechanisms different from those that control islet xenograft survival. First, we observed that the duration of islet xenograft survival is somewhat prolonged in otherwise untreated NOD.CD4aeC/eC mice, whereas skin xenografts are promptly rejected. Second, we observed the duration of islet xenograft survival in (NOD x C57BL/6)F1 mice is relatively prolonged, whereas skin xenograft survival is very short. These data suggest that skin and islet xenograft survival are controlled by different cellular mechanisms and that resistance to skin xenograft tolerance induction is a dominant trait in NOD mice.

    We have previously shown that some NOD-like defects are corrected in (NOD x C57BL/6)F1 mice, whereas others are genetically dominant cellular defects (15). For example, NOD-like defects in natural killer cell function and macrophage development are corrected in (NOD x C57BL/6)F1 mice, whereas NOD-like abnormalities in dendritic cell maturation are genetically dominant. Of particular interest is the observation that natural killer cells appear to be normal in (NOD x C57BL/6)F1 mice. NK cells have an important role in xenograft rejection (49eC51), and although defective in NOD mice (2), this defect does not appear to be the cause of their resistance to xenograft tolerance induction. It remains to be determined if other cellular defects, such as abnormal maturation of dendritic cells, contribute to the relative resistance of (NOD x C57BL/6)F1 mice to xenograft tolerance induction.

    In summary, NOD mice are resistant to not only allograft (14,15) but also xenograft tolerance induction. This resistance is largely independent of the degree of protection from diabetes that is conferred by Idd resistance loci. The data highlight the genetically controlled differences between skin and islet xenograft tolerance induced by costimulation blockade, and they lend support to the "genetic threshold" hypothesis for resistance of NOD mice to transplantation tolerance.

    ACKNOWLEDGMENTS

    Supported in part by Grants DK53006, AI46629, AI30389, AI38757, AI24544, and CA34196 and an institutional Diabetes Endocrinology Research Center grant DK32520 from the National Institutes of Health; by Grant 1-2002-396 from the Juvenile Diabetes Research Foundation; and a grant from the Diabetes Action Research and Education Foundation (to E.J.G.). L.S.W. is supported by a joint grant from the Juvenile Diabetes Research Foundation and the Wellcome Trust. 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, and the Juvenile Diabetes Research Foundation.

    The contents of this publication are solely the responsibility of the authors and do not necessarily represent the official views of the National Institutes of Health. We thank Elaine Norowski, Linda Leehy, and Linda Paquin for technical assistance.

    DST, donor-specific transfusion; mAb, monoclonal antibody; MST, median survival time

    REFERENCES

    Atkinson M, Leiter EH: The NOD mouse model of insulin dependent diabetes: as good as it gets Nat Med5 :601 eC604,1999

    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 eC36

    Kikutani H, Makino S: The murine autoimmune diabetes model: NOD and related strains. Adv Immunol51 :285 eC322,1992

    Rosmalen JGM, Leenen PJM, Pelegri C, Drexhage HA, Homo-Delarche F: Islet abnormalities in the pathogenesis of autoimmune diabetes. Trends Endocrinol Metab13 :209 eC214,2002

    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 eC1117,2001

    Davies JL, Kawaguchi Y, Bennett ST, Copeman JB, Cordell HJ, Pritchard LE, Reed PW, Gough SCL, Jenkins SC, Palmer SM, Balfour KM, Rowe BR, Farrall M, Barnett AH, Bain SC, Todd JA: A genome-wide search for human type 1 diabetes susceptibility genes. Nature371 :130 eC136,1994

    Todd JA, Wicker LS: Genetic protection from the inflammatory disease type 1 diabetes in humans and animal models. Immunity15 :387 eC395,2001

    Serreze DV, Leiter EH: Genes and pathways underlying autoimmune diabetes in NOD mice. In Molecular Pathology of Insulin Dependent Diabetes Mellitus. Von Herrath MG, Ed. New York, Karger Press,2001 , p.31 eC67

    Lyons PA, Wicker LS: Localising Polygenes in the NOD Mouse Model of Type 1 Diabetes. Theofilopoulos AN, Ed. Basel, Karger Press,1999 , p.208 eC225

    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 eC2637,2001

    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 eC1747,2000

    Lamhamedi-Cherradi SE, Boulard O, Gonzalez C, Kassis N, Damotte D, Eloy L, Fluteau G, Leevi-Strauss M, Garchon HJ: Further mapping of the Idd5.1 locus for autoimmune diabetes in NOD mice. Diabetes50 :2874 eC2878,2001

    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 eC1478,1998

    Pearson T, Markees TG, Wicker LS, Serreze DV, Peterson LB, Mordes JP, Rossini AA, Greiner DL: NOD congenic mice genetically protected from autoimmune diabetes remain resistant to transplantation tolerance induction. Diabetes52 :321 eC326,2002

    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. J Immunol171 :185 eC195,2003

    Pearson T, Weiser P, Markees TG, Serreze DV, Wicker LS, Peterson LB, Cumisky AM, Shultz LD, Mordes JP, Rossini AA, Greiner DL: Islet allograft survival induced by costimulation blockade in NOD mice is controlled by allelic variants of Idd3. Diabetes53 :1972 eC1978,2004

    Gordon EJ, Markees TG, Phillips NE, Noelle RJ, Shultz LD, Mordes JP, Rossini AA, Greiner DL: Prolonged survival of rat islet and skin xenografts in mice treated with donor splenocytes and anti-CD154 monoclonal antibody. Diabetes47 :1199 eC1206,1998

    Gordon EJ, Woda BA, Shultz LD, Rossini AA, Greiner DL, Mordes JP: Rat xenograft survival in mice treated with donor-specific transfusion and anti-CD154 antibody is enhanced by elimination of host CD4+ cells. Transplantation71 :319 eC327,2001

    Appel MC, Banuelos SJ, Greiner DL, Shultz LD, Mordes JP, Rossini AA: Prolonged survival of neonatal porcine islet xenografts in mice treated with a donor specific transfusion and anti-CD154 antibody. Transplantation77 :1341 eC1349,2004

    Visser L, Poppema S, de Haan B, Klok P, van der LJ, van den BA, De Vos P: Prolonged survival of rat islet xenografts in mice after CD45RB monotherapy. Transplantation77 :386 eC391,2004

    Lu X, Schulz M, Zihlmann HR, Borel JF: Long-term survival of hamster islet xenografts in mice under short-course treatment with nondepleting versus depleting anti-CD4 monoclonal antibodies. Xenotransplantation5 :154 eC163,1998

    Lehnert AM, Yi SN, Burgess JS, O’Connell PJ: Pancreatic islet xenograft tolerance after short-term costimulation blockade is associated with increased CD4+ T cell apoptosis but not immune deviation. Transplantation69 :1176 eC1185,2000

    Lehnert AM, Mottram PL, Han W, Walters SN, Patel AT, Hawthorne WJ, Cowan PJ, d’Apice AJ, O’Connell PJ: Blockade of the CD28 and CD40 pathways result in the acceptance of pig and rat islet xenografts but not rat cardiac grafts in mice. Transpl Immunol9 :51 eC56,2001

    Thomas FT, Ricordi C, Contreras JL, Hubbard WJ, Jiang XL, Eckhoff DE, Cartner S, Bilbao G, Neville DM Jr, Thomas JM: Reversal of naturally occurring diabetes in primates by unmodified islet xenografts without chronic immunosuppression. Transplantation67 :846 eC854,1999

    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 eC334,1996

    Serreze DV, Chapman HD, Varnum DS, Hanson MS, Reifsnyder PC, Richard SD, Fleming SA, Leiter EH, Shultz LD: B lymphocytes are essential for the initiation of T cell-mediated autoimmune diabetes: analysis of a new "speed congenic" stock of NOD. Ig e蘮ull mice. J Exp Med184 :2049 eC2053,1996

    Chiu PPL, Serreze DV, Danska JS: Development and function of diabetogenic T-cells in B-cell-deficient nonobese diabetic mice. Diabetes50 :763 eC770,2001

    Graser RT, DiLorenzo TP, Wang FM, Christianson GJ, Chapman HD, Roopenian DC, Nathenson SG, Serreze DV: Identification of a CD8 T cell that can independently mediate autoimmune diabetes development in the complete absence of CD4 T cell helper functions. J Immunol164 :3913 eC3918,2000

    Institute of Laboratory Animal Resources: U.S. Government Principles for the Utilization and Care of Vertebrate Animals Used in Testing, Research, and Training: the Guide for the Care and Use of Laboratory Animals. Washington, DC, National Research Council, National Academy of Sciences,1996

    Parker DC, Greiner DL, Phillips NE, Appel MC, Steele AW, Durie FH, Noelle RJ, Mordes JP, Rossini AA: Survival of mouse pancreatic islet allografts in recipients treated with allogeneic small lymphocytes and antibody to CD40 ligand. Proc Natl Acad Sci U S A92 :9560 eC9564,1995

    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 eC2455,1998

    Markees TG, Phillips NE, Noelle RJ, Shultz LD, Mordes JP, Greiner DL, Rossini AA: Prolonged survival of mouse skin allografts in recipients treated with donor splenocytes and antibody to CD40 ligand. Transplantation64 :329 eC335,1997

    Kaplan EL, Meier P: Nonparametric estimation from incomplete observations. J Am Statist Assn53 :457 eC481,1958

    Matthews DE, Farewell VT: The log-rank or Mantel-Haenszel test for the comparison of survival curves. In Using and Understanding Medical Statistics. Matthews DE, Farewell VT, Eds. Basel, Karger,1988 , p.79 eC87

    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 eC1843,1997

    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 eC1713,1994

    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 eC115,2000

    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 eC6630,2001

    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 eC521,2000

    Li Y, Li XC, Zheng XX, Wells AD, Turka LA, Strom TB: Blocking both signal 1 and signal 2 of T-cell activation prevents apoptosis of alloreactive T cells and induction of peripheral allograft tolerance. Nat Med5 :1298 eC1302,1999

    Wells AD, Li XC, Li Y, Walsh MC, Zheng XX, Wu Z, Nunez G, Tang A, Sayegh M, Hancock WW, Strom TB, Turka LA: Requirement for T-cell apoptosis in the induction of peripheral transplantation tolerance. Nat Med5 :1303 eC1307,1999

    Trambley J, Bingaman AW, Lin A, Elwood ET, Waitze SY, Ha J, Durham MM, Corbascio M, Cowan SR, Pearson TC, Larsen CP: Asialo GM1+ CD8+ T cells play a critical role in costimulation blockade-resistant allograft rejection. J Clin Invest104 :1715 eC1722,1999

    Yasunami Y, Bach J-F: Anti-suppressor effect of cyclophosphamide on the development of spontaneous diabetes in NOD mice. Eur J Immunol18 :484 ,1988

    Pearson T, Markees TG, Serreze DV, Pierce MA, Wicker LS, Peterson LB, Shultz LD, Mordes JP, Rossini AA, Greiner DL: Islet cell autoimmunity and transplantation tolerance: two distinct mechanisms In Immunology of Diabetes: Pathogenesis from Mouse to Man. Sanjeevi CB, Eisenbarth GS, Eds. New York, New York Academy of Sciences,2003 , p.148 eC156

    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 eC974,1999

    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 eC453,2000

    Banuelos SJ, Markees TG, Phillips NE, Appel MC, Cuthbert A, Leif J, Mordes JP, Shultz LD, Rossini AA, and Greiner DL: Regulation of skin and islet allograft survival in mice treated with costimulation blockade is mediated by different CD4+ cell subsets and different mechanisms. Transplantation78 :660 eC667,2004

    Phillips NE, Markees TG, Mordes JP, Greiner DL, Rossini AA: Blockade of CD40-mediated signaling is sufficient for inducing islet but not skin transplantation tolerance. J Immunol170 :3015 eC3023,2003

    Nakamura MC, Naper C, Niemi EC, Spusta SC, Rolstad B, Butcher GW, Seaman WE, Ryan JC: Natural killing of xenogeneic cells mediated by the mouse Ly-49D receptor. J Immunol163 :4694 eC4700,1999

    Itescu S, Kwiatkowski P, Artrip JH, Wang SF, Ankersmit J, Minanov OP, Michler RE: Role of natural killer cells, macrophages, and accessory molecule interactions in the rejection of pig-to-primate xenografts beyond the hyperacute period. Hum Immunol59 :275 eC286,1998

    Lin Y, Vandeputte M, Waer M: Natural killer cell- and macrophage-mediated rejection of concordant xenografts in the absence of T and B cell responses. J Immunol158 :5658 eC5667,1997(Ethel J. Gordon, Linda S.)