Mice with Th2-Biased Immune Systems Accept Orthotopic Corneal Allografts Placed in ''High Risk'' Eyes This information is current as of July 31, 2017. Jun Yamada, Munenori Yoshida, Andrew W. Taylor and J. Wayne Streilein J Immunol 1999; 162:5247-5255; ; http://www.jimmunol.org/content/162/9/5247 Subscription Permissions Email Alerts This article cites 51 articles, 17 of which you can access for free at: http://www.jimmunol.org/content/162/9/5247.full#ref-list-1 Information about subscribing to The Journal of Immunology is online at: http://jimmunol.org/subscription Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts The Journal of Immunology is published twice each month by The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 1999 by The American Association of Immunologists All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. Downloaded from http://www.jimmunol.org/ by guest on July 31, 2017 References Mice with Th2-Biased Immune Systems Accept Orthotopic Corneal Allografts Placed in “High Risk” Eyes1 Jun Yamada, Munenori Yoshida, Andrew W. Taylor, and J. Wayne Streilein2 A significant number of allogeneic corneas grafted orthotopically into normal eyes of normal mice and rats are accepted indefinitely (1). This high rate of acceptance, compared with the virtually uniform rejection of orthotopic allografts of other solid tissues, is an expression of the existence of immune privilege in the eye (2–5). When immune privilege in the recipient eye is compromised (so-called high risk eyes), orthotopically placed allogeneic cornea grafts are promptly rejected (6). These results from experiments with laboratory animals faithfully reflect the experience with penetrating kelatoplasties in human beings. Allogeneic corneas placed in low risk human eyes display a high rate of acceptance, and rejection episodes are usually treated successfully with only topical immunosuppressive therapy (7, 8). By contrast, cornea grafts placed in high risk human eyes, e.g., eyes with scarred or inflamed corneal surfaces, have a very poor prognosis, and even intensive systemic immunosuppressive therapy is often of no avail (9, 10). Since penetrating keratoplasties are the most common type of transplant performed in humans, and since failure of cornea grafts in high risk eyes is an important cause of blindness, understanding the pathogenesis of graft rejection as a preamble to developing successful antirejection therapies is a worthy goal for research. While the role of donor-specific Abs in effecting corneal graft rejection is in considerable dispute (11, 12), there is general agreement that T lymphocytes are the most important mediators of cornea graft rejection. In this regard, corneal allografts resemble other types of solid tissue grafts. However, in orthotopic corneal grafting Schepens Eye Research Institute, Harvard Medical School, Boston, MA 02114 Received for publication November 3, 1998. Accepted for publication February 8, 1999. The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact. minor histocompatibility (H)3 Ags, rather than Ags encoded within the MHC, appear to be the most important initiators of alloimmunity (1, 13). Whether placed in low risk or high risk eyes, minor H-incompatible cornea grafts are rejected more often and vigorously than are corneas displaying only MHC-encoded alloantigens. This unusual circumstance is due, on the one hand, to the low expression of MHC-encoded molecules on cells of the cornea (4, 14, 15) and, on the other hand, to the absence from the normal cornea of bone marrow-derived dendritic cells and macrophages that function in other solid tissue grafts as “passenger leukocytes” (4, 16). An important consequence of this situation is that recipients of corneal allografts develop delayed hypersensitivity T cells (TDH) directed at minor, rather than major, histocompatibility Ags (17). Moreover, although there is a strong correlation between the emergence of donor-specific cytotoxic T cells (Tc) and graft rejection, direct and indirect experimental evidence implicates TDH rather than Tc as the primary mediators of orthotopic corneal graft rejection (18). If rejection of orthotopic corneal allografts is due chiefly to the actions of TDH, then strategies designed to inhibit cells of this type should have a salutary effect on corneal allograft survival. Delayed hypersensitivity is typically mediated by a subset of CD41 T cells that secrete IFN-g, termed Th1 (19). Cells of this type have been directly implicated in acute rejection of other types of solid organ transplants (20, 21). Moreover, Th1 cells are cross-regulated by a different subset of CD41 T cells that secrete IL-4 and IL-10, and are termed Th2. Thus, the cytokines produced by Th2 cells suppress the activation and release of cytokines from Th1 cells (19), thereby limiting the ability of the latter cells to mediate effector responses such as delayed hypersensitivity. Several forms of transplantation tolerance (22, 23), including neonatally induced tolerance (24, 25) and the tolerance induced by treatment with antiCD4 Abs, have been strongly associated with Th2 cells (26), implying that regulation of Th1 cells by Th2 cells can promote graft acceptance. 1 This work was supported in part by National Institutes of Health Grant EY10765 and by a generous gift from Mr. and Mrs. Harry Axelrod. 2 Address correspondence and reprint requests to Dr. J. Wayne Streilein, Schepens Eye Research Institute, 20 Staniford St., Boston, MA 02114. E-mail address: [email protected] Copyright © 1999 by The American Association of Immunologists 3 Abbreviations used in this paper: H, histocompatibility; TDH, delayed hypersensitivity T cells; Tc, cytotoxic T cells; KLH, keyhole limpet hemocyanin; MFI, mean fluorescence intensity. 0022-1767/99/$02.00 Downloaded from http://www.jimmunol.org/ by guest on July 31, 2017 CD41 T cells of the Th1 type play a central role in acute rejection of solid tissue grafts, including orthotopic corneal allografts. Th1 cells, which mediate delayed hypersensitivity, are the polar opposites of CD41 Th2 cells, and the latter cells cross-regulate Th1 cells through the unique pattern of cytokines they secrete. As such, Th2 cells may have a useful role to play in preventing rejection of corneal allografts. To test this possibility, the immune systems of adult mice were biased toward Th2 responses by immunization with keyhole limpet hemocyanin plus IFA. When immunized subsequently with either OVA or allogeneic corneal tissue, these mice acquired Ag-specific primed T cells of the Th2 type. More important, allogeneic corneas grafted into neovascularized eyes of Th2-biased mice experienced significantly enhanced survival. To demonstrate that enhanced survival was promoted by donorspecific Th2 cells, lymphoid cells from keyhole limpet hemocyanin-immune mice bearing healthy corneal allografts suppressed orthotopic corneal allograft rejection when adoptively transferred into naive, syngeneic recipients. We conclude that acceptance of corneal allografts in neovascularized mouse eyes can be significantly enhanced by biasing the recipient immune system toward Th2 responses. The Journal of Immunology, 1999, 162: 5247–5255. 5248 The systemic immune responses of adult mice can be biased in the direction of Th1 or Th2 cells depending upon the method of initial immunization (27–30). Moreover, mice that have mounted Th2-type responses to one Ag often display Th2 responses to subsequent immunizations with different Ags (31). We have employed such a strategy in an effort to modify the rate of rejection of orthotopic corneal allografts in mice. Our results indicate that mice with immune systems heavily biased toward Th2 responses accept orthotopic corneal allografts at a higher rate than normal mice. Moreover, orthotopic cornea allograft-bearing, Th2-biased mice acquire regulatory T cells that secrete Th2-type cytokines and promote graft acceptance when adoptively transferred into naive recipients. Materials and Methods Animals Antigens OVA (Sigma, St. Louis, MO) and keyhole limpet hemocyanin (KLH) of Megathura crenulata (Calbiochem, San Diego, CA) were used. Preimmunization with KLH To prepare Th2-biased recipients, BALB/c and C57BL/6 mice received i.p. injections of 50 ml of KLH emulsified in IFA (Difco, Detroit, MI). Control mice received HBSS plus IFA. Secondary immunization In one set of experiments, BALB/c mice and C57BL/6 mice received into the nape of the neck or into the hind footpad injections of 100 mg of OVA (with 50 mg of KLH or HBSS alone) in CFA. In another set of experiments, allogeneic corneas (full thickness, 2 mm in diameter) were inserted under the skin of the dorsum of one hind foot. Immediately thereafter, the footpad of the same foot received an injection of KLH (50 mg) or HBSS alone in CFA. In a third set of experiments, mice that received an orthotopic corneal allograft into one eye also received into the nape of the neck or into one hind footpad immediately thereafter an injection of KLH (50 mg) or HBSS alone in CFA. Induction and grading of corneal neovascularization Corneal neovascularization (referred to as high risk graft beds) was induced by intrastromal sutures as described previously (6). Briefly, under aseptic microsurgical technique using an operating microscope three interrupted 11-0 sutures were placed in the central cornea of one eye of a normal BALB/c mice. Two weeks later, the neovascularized beds then received orthotopic corneal transplants as described below. The neovascularized recipient cornea, including the sutures, was removed at this time. Orthotopic corneal transplantation As described previously (32), each recipient was deeply anesthetized with an i.p. injection of 3 mg of ketamine and 0.0075 mg of xylazine before all surgical procedures. The central 2 mm of the donor cornea was excised and secured in recipient graft beds with eight interrupted 11-0 nylon sutures (Sharppoint, Vanguard, Houston, TX). Antibiotic ointment was applied to the corneal surface, and the lids were closed for 72 h with an 8-0 nylon tarsorrhaphy. All grafted eyes were examined after 72 h; no grafts were excluded from analysis because of technical difficulties. Transplant sutures were removed in all cases on day 7. opacity; 21 5 mild deep (stromal) opacity with pupil margin and iris vessels (iris structure) visible; 31 5 moderate stromal opacity with only pupil margin visible; 41 5 intense stromal opacity with the anterior chamber visible; and 51 5 maximal corneal opacity with total obscuration of the anterior chamber. Grafts with an opacity score of 21 or greater after 3 wk were considered as rejected (immunologic failure); grafts with an opacity score of 31 or greater at 2 wk that never cleared by 8 wk were also regarded as rejected (1). Culture medium Serum-free medium was used for cultures and was composed of RPMI 1640 medium, 10 mM HEPES, 0.1 mM nonessential amino acids, 1 mM sodium pyruvate, 100 U/ml penicillin, 100 mg/ml streptomycin (all from BioWhitaker, Walkersville, MD), and 1 3 1025 M 2-ME (Sigma) and supplemented with 0.1% BSA (Sigma), insulin, transferrin, and selenium, and culture supplement (1 mg/ml iron-free transferrin, 10 ng/ml linoleic acid, 0.3 ng/ml Na2Se, and 0.2 mg/ml Fe(NO3)3; Collaborative Biomedical Products, Bedford, MA) (33). Cell culture Lymph nodes draining immunization or grafting sites were removed and pressed through nylon mesh to produce a single-cell suspension. T cells were subsequently purified to .95% by a T cell enrichment column (R&D Systems, Minneapolis, MN). The T cells were washed, counted, and resuspended at 4 3 105 in 96-well plates. In some experiments T cells were stimulated with x-irradiated (2000 rad) syngeneic splenic adherent cells pulsed with 50 mg/ml of OVA or KLH. In other experiments T cells were restimulated with irradiated (2000 rad) allogeneic splenic adherent cells. Cells were cultured in serum-free medium at 37°C in an atmosphere of 5% CO2. For the results presented in Figs. 1 and 2, lymph node cells from two or three animals were pooled and analyzed per experiment; each experiment was repeated at least three times with similar results. Proliferation assay and mixed lymphocyte reactions Cultures were pulsed with 0.5 mCi of [3H]thymidine 8 h before termination, and cells were harvested onto glass filters using an automated well harvester (Tomtec, Orange, CT). Radioactivity was assessed by liquid scintillation spectrometry, and the amount was expressed as counts per minute. IFN-g, IL-2, IL-4, and IL-10 assays Cultures similar to those described above were established and sustained for 24, 48, or 72 h. At each time point, supernatants were collected and analyzed for contents of IFN-g, IL-2, IL-4, and IL-10 using ELISA kits according to the manufacturer’s instructions (Endogen, Cambridge, MA). Intracellular staining of IFN-g and IL-4 T cells that were cultured with stimulator cells for 24 h were harvested and treated with HBSS supplemented with 5 mg/ml brefeldin A (Sigma), a compound known to disrupt the Golgi apparatus, thus inhibiting protein secretion for approximately 6 h. Phenotypic analysis was performed by staining freshly recovered cells with Cy-conjugated CD31 (1 mg mAb/106 cells in 1 ml of PBS-1% BSA; PharMingen, San Diego, CA) for 40 min at room temperature in a dark. The cells were then washed twice in PBS-1% BSA. The pellet was resuspended in 0.5 ml of PermeaFix buffer (Ortho Diagnostics, Raritan, NJ; diluted to 1/1.75 with distilled water and supplemented with 0.1% EDTA) and incubated at room temperature for a further 40 min. The cells were washed twice in PBS-1% BSA and stained for intracellular cytokines with FITC-conjugated mAb against IL-4-FITC or IFN-g-FITC (1 mg/106 cells; PharMingen) for an additional 40 min at room temperature in the dark. The cells were then washed twice in PBS-1% BSA and analyzed by flow cytometry (EPICS XL analyzer, Coulter, Hialeah, FL). For the results presented in Figs. 1C and 2C, the mean fluorescence intensity (MFI) 6 SEM were determined for CD31, IFN-g1, or IL-41 cells. MFI was based on 5000 measured events. The significance of each data point was assessed by calculating the half-peak correlation variance. Assessment of graft survival Grafts were evaluated by slitlamp biomicroscopy twice a week. At each time point grafts were scored for opacification. A previously described scoring system (1) was used to measure the degree of opacification between 0 –51: 0 5 clear and compact graft; 11 5 minimal superficial Statistical methods Statistical analyses were performed using Fisher’s exact probability test for proportional rates of rejected allografts. All values of p , 0.05 were deemed significant. Downloaded from http://www.jimmunol.org/ by guest on July 31, 2017 Adult male BALB/c (H-2d) and C57BL/6 (H-2b) mice were purchased from Taconic Farms (Germantown, NY), and adult male BALB.B (C.B10H2b/LilMcdJ, H-2b), DBA/2 (H-2d), and B10.D2 (H-2d) mice were purchased from The Jackson Laboratory (Bar Harbor, ME) and used as experimental subjects or cornea graft donors between 8 and 12 wk of age. All animals were treated according to the Association for Research in Vision and Ophthalmology Statement for the Use of Animals in Ophthalmic and Vision Research. Th2 CELLS PROMOTE CORNEAL GRAFT SURVIVAL The Journal of Immunology 5249 Results In KLH-immune mice, immunization with OVA induces OVA-specific Th2 cells Downloaded from http://www.jimmunol.org/ by guest on July 31, 2017 Adult mice immunized with KLH in the presence of IFA have been reported to develop CD41 T cells that secrete IL-4, IL-5, IL-6, and IL-10 when stimulated in vitro with KLH (31), i.e., Th2 cells, whereas similar mice immunized with KLH in CFA acquire KLH-specific T cells that secrete predominantly IL-2 and IFN-g, i.e., Th1 cells. Moreover, mice immunized first with KLH plus IFA respond to subsequent immunizations with a different protein Ag, e.g., myelin basic protein plus CFA, and generate CD41, myelin basic protein-specific T cells that secrete predominately Th2-type cytokines (34). Our experimental goal was to use the logic of these experiments to determine whether corneal allografts placed orthotopically in eyes of mice with systemic immune systems biased toward Th2 would experience enhanced survival. To begin these studies, panels of C57BL/6 and BALB/c mice received an i.p. injection of KLH (50 mg) in IFA (50 ml). Spleen cells were removed from these mice 4 wk later, rendered into single cell suspensions, and stimulated in vitro with KLH. The supernatants of these cultures contained significant quantities of IL-4 and IL-10, but little IL-2 or IFN-g (data not shown), indicating that the mice had mounted a Th2-type response to KLH. To determine whether mice of this type would respond to a second Ag with a Th2 bias, two panels of mice were immunized initially with KLH in IFA, then 4 wk later, one panel received an s.c. injection of KLH (50 mg) plus OVA (100 mg) mixed with CFA (50 ml). Control panels of mice immunized initially with KLH in IFA received an s.c. injection of OVA (100 mg) in CFA. Draining lymph nodes were removed from these mice, and purified T cells were prepared and stimulated in vitro with x-irradiated (2000 rad) syngeneic splenic APC pulsed with 50 mg/ml of OVA. At 24, 48, and 72 h, supernatants were removed from these cultures and assayed by ELISA for IFN-g and IL-4. [3H]thymidine was added to separate cultures of 72-h duration to assess T cell proliferation. A third set of cultures was harvested at 24 h, and the responding T cells were analyzed by flow cytometry for intracellular content of IFN-g and IL-4. The results of representative experiments are presented in Fig. 1, A–C. As revealed in Fig. 1A, T cells from all mice proliferated in vitro in response to OVA stimulation. However, the extent of proliferation was greater with T cells obtained from KLHimmune mice immunized subsequently with OVA and CFA alone with T cells from mice that received a subsequent immunization with OVA plus KLH and CFA. ELISAs of culture supernatants (Fig. 1B) indicate that unstimulated T cells produced IL-4 spontaneously, but little if any IFN-g. More important, OVA-stimulated T cells from KLH-immune mice immunized subsequently with OVA alone produced significantly more IFN-g and significantly less IL-4 than OVA-stimulated T cells from mice whose second immunization contained both OVA and KLH. Finally, nearly twice as many T cells from KLH-immune mice that were immunized subsequently with OVA plus KLH responded to OVA stimulation in vitro by acquiring intracellular IL-4 compared with T cells from mice immunized with OVA alone, whereas many more T cells from the latter mice contained intracellular IFN-g compared with T cells from mice immunized with OVA plus KLH (Fig. 1C). Moreover, the MFI of CD31 cells containing IFN-g after treatment with OVA alone was significantly greater than that of cells after treatment with OVA plus KLH. The opposite was true for CD31 cells containing IL-4. In aggregate, these findings confirm the results previously reported by Falcone and Bloom (34) and indicate that mice that first encounter KLH in the presence of IFA develop an immune system that is biased toward Th2 FIGURE 1. Proliferation responses and cytokine secretion by OVA-stimulated T cells. BALB/c and C57BL/6 mice, immunized first with KLH plus IFA, were immunized 4 wk later with OVA (F, control, BALB/c; f, C57BL/6) or OVA, KLH, and CFA (E, experimental, BALB/c; M, C57BL/6). Seven days later, the mice were killed; purified T cells prepared from draining lymph nodes were stimulated in vitro with OVA for 3 days. To assess proliferation (A), [3H]thymidine was added during the terminal 8 h of culture. To assess IFN-g and IL-4 production (B), supernatants were collected at 72 h and assayed by ELISA. To assess intracellular IFN-g and IL-4 contents (C), CD31 T cells collected after 24-h culture were analyzed by flow cytometry using appropriate anticytokine Abs. In C, the MFI 6 SEM of CD31 were: IFN-g1 after OVA, 5.96 6 0.1; and after OVA plus KLH, 5.13 6 0.11 (p , 0.005); the MFI 6 SEM of CD31 were: IL-41 after OVA, 4.96 6 0.14; and after OVA and KLH, 5.50 6 0.09 (p , 0.005). 5250 Th2 CELLS PROMOTE CORNEAL GRAFT SURVIVAL responses. When subsequently immunized with a different Ag (OVA) in conjunction with the original Ag (KLH), the preponderant T cells responding to OVA (as well as T cells responding to KLH; data not down) produce IL-4 rather than IFN-g and resemble Th2 cells. In KLH-immune mice, immunization with heterotopic corneal allografts induces donor-specific Th2 cells Orthotopic corneal allografts display enhanced survival in KLHimmune recipients Whereas allogeneic corneas grafted orthotopically into normal mouse eyes enjoy “immune privilege,” similar grafts placed in Downloaded from http://www.jimmunol.org/ by guest on July 31, 2017 Our next goal was to determine whether mice with Th2-biased immune systems would respond to immunization with alloantigens by developing alloantigen-specific Th2 responses. Because minor H, rather than MHC, encoded alloantigens represent the stronger stimulus to alloimmunity induced by orthotopic corneal allografts, the following experiments were conducted with C57BL/6 mice as recipients of BALB.B cornea grafts and with BALB/c mice as recipients of B10.D2 cornea grafts. The members of these two strain combinations share the same H-2 genes, but differ at multiple minor H loci. Panels of C57BL/6 and BALB/c mice received i.p. injections of KLH (50 mg) in IFA. Four weeks later, the right footpad of one panel of mice received an injection of KLH or HBSS (control) in CFA. Simultaneously, an incision was made on the dorsal surface of the same foot, and a segment of BALB.B or B10.D2 cornea (2 mm in diameter) was inserted. In this manner, antigenic information from both the heterotopic corneal graft and the footpad Ag injection site would be focused in the draining popliteal lymph node. One week later, draining popliteal lymph nodes were removed, and T cells, purified from these lymph node cell suspensions, were stimulated in vitro with x-irradiated spleen cells from donors genetically identical with the heterotopic corneal grafts. The results of these experiments are presented in Fig. 2. Cultures of 72-h duration to which [3H]thymidine had been added 8 h before termination revealed T cell proliferation, indicating that recipients of heterotopic corneal allografts had been primed (Fig. 2A). As before, T cells from mice that received a footpad injection of HBSS rather than KLH at the time the allogeneic cornea was placed heterotopically proliferated more vigorously. Supernatants were removed from 24-, 48-, and 72-h cultures and assayed for IFN-g and IL-4 contents. T cells stimulated with allogeneic spleen cells produced significant amounts of IFN-g (Fig. 2B). The amount of IFN-g produced by T cells from mice that received heterotopic cornea grafts and KLH into their footpads was significantly less than that produced by similarly grafted mice that received HBSS injections instead of KLH. Alternatively, T cells cultured in the presence of allogeneic spleen cells produced more IL-4 than did T cells cultured in the presence of syngeneic spleen cells. Moreover, T cells from mice that received footpad cornea grafts simultaneously with KLH tended to produce more IL-4 when stimulated with allogeneic spleen cells than did HBSS-injected controls. In a third assay, T cells were cultured for 24 h in the presence of alloantigenic spleen cells, then harvested and assessed for intracellular IFN-g and IL-4 contents by flow cytometry. As revealed in Fig. 2C, a significantly higher percentage of CD31 cells from mice that received footpad KLH plus allogeneic cornea grafts contained intracytoplasmic IL-4, whereas these same populations of CD31 cells contained a lower percentage of IFN-g-containing cells. We conclude from these results that allogeneic corneal tissue grafted heterotopically into Th2-biased mice induced donor-specific alloimmunity in which the responding T cells were also biased toward Th2-type cytokine production. FIGURE 2. Proliferation responses and cytokine secretion by alloantigen-stimulated T cells. BALB/c and C57BL/6 mice, immunized first with KLH plus IFA, were immunized 4 wk later with syngeneic or allogeneic (B10.D2 or BALB.B, respectively) cornea fragments that were implanted into the dorsum of the right foot. In the footpad of the same limb, KLH (or HBSS, control) plus CFA were injected simultaneously. Seven days later the draining lymph nodes were removed, and purified T cells were cultured with syngeneic or allogeneic stimulator cells for 3 days. Proliferation (A), IFN-g and IL-4 secretion in supernatants (B), and intracellular IFN-g and IL-4 contents (C) were assessed and are displayed as described in Fig. 1. In C, the MFI 6 SEM of CD31 were: IFN-g1 after B10.D2 grafts, 5.12 6 0.08; and after B10.D2 grafts plus KLH, 4.72 6 0.10 (p , 0.005); the MFI 6 SEM of CD31 were: IL-41 after B10.D2 grafts, 4.78 6 0.11; and after B10.D2 grafts plus KLH, 5.18 6 0.07 (p , 0.005). The Journal of Immunology 5251 FIGURE 3. Fate of orthotopic corneal allografts in KLH-preimmunized mice. BALB/c mice were immunized with KLH plus IFA or with HBSS plus IFA (control); 2 wk later sutures were placed through the central cornea of right eyes to induce neovascularization. Two weeks thereafter corneas from C57BL/6 or B10.D2 donors were placed orthotopically in these eyes. Immediately thereafter the mice received an injection of CFA plus KLH or HBSS into the nape of the neck. Graft rejection was scored clinically (see Materials and Methods), and the results are presented as Kaplan-Meier survival curves. A, Recipients of C57BL/6 cornea grafts that were preimmunized with KLH in IFA followed by HBSS plus CFA at the time of grafting (n 5 13; M), with HBSS plus IFA followed by KLH plus CFA at the time of grafting (n 5 8; ‚), or with KLH plus IFA followed by KLH plus CFA at the time of grafting (n 5 10; E). p, p , 0.009. B, Recipients of B10.D2 cornea grafts that were preimmunized with KLH plus IFA followed by HBSS plus CFA at the time of grafting (n 5 13; M) or with KLH plus IFA followed by KLH plus CFA at the time of grafting (n 5 14; E). p, p , 0.0001. high risk eyes are promptly rejected (6). Rejection in the latter instance is associated with acquisition of donor-specific T cells that mediate delayed hypersensitivity, a presumed Th1 response. We hypothesized that allogeneic corneas grafted into high risk eyes of mice with Th2-biased immune systems would experience enhanced survival. To test the validity of this hypothesis, panels of BALB/c mice received i.p. injections of KLH (or HBSS, negative controls) plus IFA. Two weeks later three sutures were placed in the central cornea of one eye to induce neovascularization. When new vessel formation was robust (after 2 wk), these eyes received orthotopic grafts of corneas from either C57BL/6 or B10.D2 donors. Immediately after the graft procedure was completed, recipient mice received s.c. injections of KLH or HBSS in CFA placed at the nape of the neck. The fate of these grafts was then assessed and scored clinically. The results are presented in Fig. 3, A and B. BALB/c mice that received i.p. HBSS injections before corneal grafting and BALB/c mice that received KLH plus IFA before grafting and HBSS at the time of grafting rejected their orthotopic C57BL/6 cornea grafts with considerable vigor. However, mice that first received KLH i.p. and then a second KLH exposure at the time of orthotopic corneal allografting rejected C57BL/6 grafts less swiftly and less often (Fig. 3A). An even more dramatic en- hancement of graft survival was observed in BALB/c mice that received KLH 4 wk before and again at the same time as they received an orthotopic B10.D2 corneal allograft. Less than 50% of these grafts were rejected, and rejection, when it occurred, was considerably delayed compared with that in mice that received HBSS at the time of penetrating keratoplasty. These results support the hypothesis that mice with Th2-biased immune systems are less able to reject orthotopic corneal allografts than their normal counterparts. In the experiment just described, mice with reduced capacity to reject cornea allografts received two injections of KLH: the first with IFA i.p., and the second with CFA injected into the nape of the neck. This latter site was chosen because the draining cervical lymph nodes also serve as the draining nodes of eyes bearing orthotopic corneal allografts. To determine whether simultaneous processing of KLH and alloantigens in the same lymph node is important to allograft acceptance, similar experiments were performed with the following modification: at the time donor B10.D2 corneas were grafted into high risk eyes, KLH plus CFA was either injected into the nape of the neck or into one hind footpad. Control mice received HBSS plus CFA into the nape of the neck. The fate of these grafts is summarized in Fig. 4. As before, a high proportion of KLH-immune BALB/c mice that received an orthotopic B10.D2 cornea graft along with an injection of KLH plus CFA into the nape of the neck accepted their cornea grafts indefinitely. By contrast, KLH-immune mice that received only HBSS plus CFA at the time of grafting rejected their B10.D2 grafts promptly. An intermediate result was observed among KLH-immune mice that received their second KLH injection at a site distant from the orthotopic cornea graft. Although fewer of these mice accepted their cornea grafts than did the mice that received KLH injections into the nape of the neck, the footpad-injected mice accepted their orthotopic corneal allografts with a higher frequency than did mice who only received HBSS rather than KLH at the time of penetrating keratoplasty. Thus, the ability of a Th2-biased immune system to influence the survival of orthotopic corneal allografts is improved if the lymphoid organ handling alloantigenic signals from the eye is also responding to the Th2-dependent Ag. Downloaded from http://www.jimmunol.org/ by guest on July 31, 2017 FIGURE 4. Fate of orthotopic corneal allografts in KLH-preimmunized mice that received a second challenge with KLH at the same or a distant site. BALB/c mice were immunized with KLH plus IFA or with HBSS plus IFA (control); 2 wk later sutures were placed through the central cornea of right eyes to induce neovascularization. Two weeks thereafter corneas from B10.D2 donors were placed orthotopically in these eyes. Immediately thereafter the mice received an injection of CFA plus KLH or HBSS either into the nape of the neck or into one hind footpad. Graft rejection was scored clinically (see Materials and Methods) and is presented as KaplanMeier survival curves. KLH-presensitized recipients of B10.D2 cornea grafts were challenged immediately after grafting with HBSS plus CFA into the nape of the neck (n 5 13; M), with KLH plus CFA into one hind footpad (n 5 10; ‚), or with KLH plus CFA into the nape of the neck (n 5 18; E). ‚ vs E, p , 0.05; E vs M, p , 0.0001. 5252 Th2 CELLS PROMOTE CORNEAL GRAFT SURVIVAL Th2-enhanced corneal allograft survival can be adoptively transferred The hypothesis that Th2-biased immune systems promote acceptance of orthotopic cornea allografts implies that donor-specific T cells are generated that not only secrete Th2-type cytokines, but down-regulate the emergence of allodestructive, Th1-type T cells. If true, then T cells from mice with accepted corneal allografts should be able to impair graft rejection when adoptively transferred into naive, syngeneic recipients. To test this implication, panels of BALB/c mice received KLH plus IFA i.p. Four weeks later, B10.D2 or DBA/2 corneas were grafted into neovascularized eyes of these mice; simultaneously, the mice received an injection of KLH (or HBSS) plus CFA into the nape of the neck. Two weeks later, at a time when the grafts appeared perfectly healthy, the mice were sacrificed, and their cervical lymph nodes and spleen were removed. Single-cell suspensions were prepared, pooled, and injected i.v. (one donor equivalent per recipient) into naive BALB/c mice. Sutures had already been placed in one eye of these recipient mice 2 wk previously to create a high risk eye. Immediately after the transfer of donor lymphoid cells, B10.D2 corneas were grafted into the neovascularized eyes of adoptive transfer recipients, and the fate of the grafts was assessed clinically. As the results presented in Fig. 5 reveal, lymphoid cells obtained from Th2-biased BALB/c mice bearing healthy B10.D2 corneal allografts promoted the survival of B10.D2 grafts in high risk eyes of normal BALB/c mice. If the adoptively transferred cells were obtained from donors that received a booster injection of HBSS rather than KLH at the time of cornea grafting, no improved graft survival was observed. Moreover, lymphoid cells obtained from Th2-biased BALB/c mice bearing healthy DBA/2 corneal allografts (which share the same MHC Ags, but display third-party minor Ags) did not promote the survival of B10.D2 grafts in high risk eyes of normal BALB/c mice. These results indicate that mice that received orthotopic corneal allografts at a time when their immune system was biased FIGURE 6. Proliferation responses and cytokine secretion by donorspecific T cells from KLH-preimmunized mice that accepted orthotopic corneal allografts. KLH-preimmunized BALB/c mice received orthotopic B10.D2 corneal allografts followed immediately by injection of KLH or HBSS plus CFA into the nape of the neck. Seven days later the draining lymph nodes were removed, and purified T cells were cultured with syngeneic or B10.D2 stimulator cells for 3 days. Proliferation (A) and IFN-g and IL-4 secretion in supernatants (B) were assessed and are displayed as described in Fig. 1. toward Th2 responses acquired donor-specific regulatory T cells that suppressed the emergence of allodestructive T cells of the Th1 type. Alloreactive T cells from KLH-immune mice that accept orthotopic cornea grafts display a Th2 phenotype To test whether KLH-immune mice that accepted orthotopic corneal allografts possessed donor-specific T cells of the Th2 type, BALB/c mice were immunized first with KLH plus IFA i.p. Two weeks later sutures were placed through the central cornea of one eye and after another 2-wk interval, B10.D2 corneas were grafted into these neovascularized eyes. Simultaneously, the mice received into the nape of the neck KLH (or HBSS) plus CFA. After another 2 wk, draining cervical lymph nodes and spleens were removed from these mice, and purified T cells were obtained. These cells were placed in culture and stimulated with x-irradiated B10.D2 spleen cells. As before, the cultured cells were assayed for proliferation, cytokine secretion into the culture supernatant, and intracytoplasmic contents of IFN-g and IL-4. The results of these experiments are presented in Fig. 6 and indicate that T cells from Downloaded from http://www.jimmunol.org/ by guest on July 31, 2017 FIGURE 5. Fate of orthotopic corneal allografts in recipients of lymphoid cells from KLH-preimmunized donor mice. BALB/c mice were preimmunized with KLH plus IFA. Four weeks later, these mice received orthotopic B10.D2 or DBA/2 cornea grafts and simultaneously were immunized in the nape of the neck with KLH or HBSS plus CFA. Two weeks later, cervical lymph nodes and spleens were removed, and single-cell suspensions were injected i.v. into naive BALB/c mice in which one eye had suture-induced neovascularization. Immediately after the transfer, B10.D2 corneas were grafted into these neovascularized eyes. Graft rejection was scored clinically (see Materials and Methods) and is presented as KaplanMeier survival curves. M, Recipients of lymphoid cells from donors challenged with HBSS plus CFA at the time they received B10.D2 cornea grafts (n 5 10); E, recipients of lymphoid cells from donors challenged with KLH plus CFA at the time they received B10.D2 cornea grafts (n 5 10); ‚, recipients of lymphoid cells from donors challenged with KLH plus CFA at the time they received DBA/2 cornea grafts (n 5 10). E vs M, p , 0.0002; E vs ‚, p , 0.002. The Journal of Immunology cornea-grafted mice that received immunization with CFA plus HBSS proliferated vigorously, and secreted copious amounts of IFN-g when stimulated with donor alloantigens in vitro. By contrast, T cells from cornea-grafted mice immunized with CFA plus KLH proliferated less well and secreted IL-4, rather than IFN-g, when stimulated with donor alloantigens. Neither T cell type produced sufficient TGF-b in these experiments to be detectable in our assays (data not shown). We conclude that KLH-immune mice with Th2-biased immune systems respond to alloantigens on orthotopic cornea grafts by generating Th2-type alloreactive T cells. Discussion case correlates with autoreactive T cells that secrete Th2-type cytokines. Our experimental results confirm these findings and indicate that the immune apparatus can be experimentally biased such that subsequent immune responses to other Ags will be predominately of the Th2 (or of the Th1) type. Not all investigators agree that Th1-type responses promote, whereas Th2-type responses suppress, allograft rejection. Thus, Piccotti et al. (41) have reported that inhibition of IL-12 activity, which promotes Th2 responses, nonetheless enhances the rejection of cardiac allografts. Moreover, VanBuskirk et al. (42) have found that infusion of Th2biased T cells into SCID mice bearing cardiac allografts precipitates acute graft rejection. Thus, it cannot be simply assumed that biasing an alloimmune response toward the Th2 direction will necessarily promote graft survival. Other factors must govern the outcome, and our experiments suggest that immune privilege of the graft site may be one of those factors. Results from experiments in numerous laboratories indicate that rejection of orthotopic corneal allografts in rodents is mediated predominately by CD41 T cells of the delayed hypersensitivity, Th1 type (18, 43). Since Th2 cells can down-regulate Th1 cells (19), pre-emptive biasing of a recipient’s immune system in a Th2 direction should have a salutary effect on corneal allograft acceptance. Our experimental results indicate that this is indeed the case. C57BL/6 and BALB/c mice immunized first with KLH plus IFA developed immune systems biased toward Th2 responses. When immunized subsequently with minor H-incompatible alloantigenic corneal tissue, the T cells of these mice responded in vitro to donor alloantigens by producing predominantly Th2-type cytokines. Our experiments indicate that this pattern of cytokine production was promoted by preliminary biasing of recipient immune systems with KLH and IFA because the effect was only observed if subsequent immunization with alloantigenic tissue was accompanied by re-exposure to KLH. Thus, CD41 T cells that respond to minor H Ags resemble CD41 T cells that recognize soluble protein Ags in their penchant to differentiate into IL-4-secreting cells when they encounter their Ag in a microenvironment containing activated Th2 cells. This point was strongly supported by the fate of allogeneic corneas grafted into high risk eyes of mice with KLHinduced Th2-biased immune systems. KLH-primed BALB/c mice accepted a very high proportion of orthotopic B10.D2 cornea grafts, whereas control mice rejected such cornea grafts acutely. It is of interest that the success of grafts in KLH-immune mice was significantly enhanced if the cervical lymph nodes draining the graft site received a booster injection of KLH at the time of grafting. This result emphasizes the importance of a Th2-like microenvironment in the draining lymph node at the time donor-specific T cells are first activated. Our findings also indicate that even MHC-incompatible corneal allografts displayed enhanced survival when placed in high risk eyes of KLH-immune BALB/c mice, although the effect was less pronounced than when minor H only, disparate grafts were used. The enhanced survival displayed by cornea grafts in eyes of Th2biased mice was clearly related to the emergence of donor alloantigenic-specific T cells of the Th2 type. Not only did T cells harvested from these mice resemble Th2 cells when stimulated with donor alloantigens in vitro, but they were able to suppress the rejection of orthotopic corneal allografts when adoptively transferred into naive, syngeneic mice. We conclude that pre-emptive biasing of a recipient immune system toward the Th2 phenotype is an effective strategy for promoting the acceptance of corneal allografts, even grafts placed in high risk eyes with neovascularized corneas. Downloaded from http://www.jimmunol.org/ by guest on July 31, 2017 Attempting to prevent rejection of orthotopic corneal allografts by biasing the recipient’s immune system toward Th2 responses represents the merging of two independent lines of investigation. On the one hand, CD41 T cells that mediate delayed hypersensitivity and secrete the proinflammatory cytokines IFN-g and TNF-b in response to Ag recognition have emerged as key effectors in rejection of solid tissue allografts (20, 21). The central role in graft rejection of Th1-type CD41 T cells that recognize allogeneic class II MHC molecules derives in part from the prominent role of passenger leukocytes typically present in solid tissue grafts. Bone marrow-derived dendritic cells and macrophages that constitutively express class II molecules confer potent immunogenicity to solid tissue grafts, in part by activating class II-specific and class II-restricted CD41 T cells. The ability of passenger leukocytes to provide potent costimulation via secretion of IL-12 and expression of CD40 (35, 36) accounts for why the responding T cells are of the Th1, delayed hypersensitivity type. Not only is this class of T cells capable in its own right of triggering an allodestructive inflammatory response, but Th1 cells secrete cytokines that promote the terminal differentiation at the graft site of primed, alloreactive cytotoxic CD81 T cells. Consequently, the combined actions of CD41 and CD81 effector T cells, both of which arise from Th1 cells activation, effect acute graft rejection. On the other hand, Th1 and Th2 cells each secrete distinctly different arrays of cytokines with widely different consequences (19, 37–39). Th1 cells promote immunogenic inflammation, not only via their own direct ability to secrete proinflammatory cytokines, but because they promote B cell differentiation in a manner that generates Igs of the complement-fixing variety. Th2 cells, by contrast, do not usually mediate inflammatory responses directly (although exceptions to this statement exist), and the influence of these T cells on B cell differentiation causes the emergence of cells that secrete IgG Abs that do not fix complement. These properties have given Th2 cells the reputation of being anti-inflammatory. In many ways, this reputation is well deserved because Th2 cells possess the unique capacity to down-regulate the development and expression of Th1-dependent immunity (19). In fact, the unique array of cytokines produced by Th2 cells, including IL-4 and IL10, directly inhibits Th0 cells from differentiating into Th1 cells and suppresses Th1 cells from producing their distinctive cytokines. Similarly, Th1 cells can inhibit the generation and functions of Th2 cells (40). Experimental evidence from a variety of laboratories indicates that mice that develop a strong Th1-type response to one Ag respond to subsequent Ags with a Th1-biased response. Conversely, mice that respond to one Ag with a vigorous Th2-type response respond to subsequent Ags with a similar Th2-biased response. In particular, Falcone and Bloom (34) have reported that immunization against a non-self Ag that leads to a Th2-type response biases recipients such that subsequent immunization with an autoantigen fails to result in autoimmune disease. The lack of disease in this 5253 5254 lorecognition (51–53). These responding T cells are mostly CD41. In our current experiments, we have assumed that the Th2 bias of recipient immune systems influenced donor minor H-specific, selfMHC restricted CD41 T cells to differentiate into Th2-type cells, thereby preventing the emergence of donor-specific Th1 cells that would have mediated graft rejection. The further finding that minor H only disparate cornea allografts survived better in Th2-biased mice than grafts that also displayed MHC alloantigens supports this view. Not only are the T cells that recognize MHC encoded alloantigens via the direct pathway of allorecognition of higher frequency in naive mice than minor H-specific T cells, but many of the direct alloreactive T cells have the phenotype of memory Th1type cells and may, therefore, be less susceptible to being deviated in a Th2-biased environment. Acknowledgments We thank Drs. Bruce Ksander and Reza Dana for helpful discussions of these experiments and Dr. Andrew W. Taylor for assistance in determining the statistical significance of the flow cytometry measurements. We acknowledge the excellent managerial support of Dr. Jacqueline Doherty and Marie Ortega. References 1. Sonoda, Y., and J. W. Streilein. 1992. Orthotopic corneal transplantation in mice: evidence that the immunogenetic rules of rejection do not apply. Transplantation 54:694. 2. Niederkorn, J. Y. 1990. Immune privilege and immune regulation in the eye. Adv. Immunol. 48:191. 3. Streilein, J. W. 1987. Immune regulation and the eye: a dangerous compromise. FASEB J. 1:199. 4. Streilein, J. W., G. B. Toews, and P. R. Bergstresser. 1979. Corneal allografts fail to express Ia antigens. Nature 282:326. 5. Griffith, T. S., T. Brunner, S. M. Fletcher, D. R. Green, and T. A. Ferguson. 1995. Fas ligand-induced apoptosis as a mechanism of immune privilege. Science 270: 1189. 6. Sano, Y., B. R. Ksander, and J. W. Streilein. 1995. Fate of orthotopic corneal allografts in eyes that cannot support anterior chamber-associated immune deviation induction. Invest. Ophthalmol. Vis. Sci. 36:2176. 7. Brady, S. E., C. J. Rapuano, J. J. Arentsen, E. J. Cohen, and P. R. Laibson. 1989. Clinical indications for and procedures associated with penetrating keratoplasty, 1983–1988. Am. J. Ophthalmol. 108:118. 8. Smith, R. E., H. R. McDonald, A. B. Nesburn, and D. S. Minckler. 1980. Penetrating keratoplasty: changing indications, 1947 to 1978. Arch. Ophthalmol. 98: 1226. 9. The Collaborative Corneal Transplantation Studies Research Group. 1992. The collaborative corneal transplantation studies (CCTS). Effectiveness of histocompatibility matching in high-risk corneal transplantation. Arch. Ophthalmol. 110: 1392. 10. Mader, T. H., and R. D. Stulting. 1991. The high-risk penetrating keratoplasty. Ophthalmol. Clin. North Am. 4:411. 11. Hegde, S., S. Hargrave, J. Mellon, and J. Y. Niederkorn. 1998. Antibody and cell mediated immune responses during rejection of a murine orthotopic corneal graft. Invest. Ophthalmol. Vis. Sci. 39:S455. 12. Hutchinson, I. V., Y. Alam, and W. R. Ayliffe. 1995. The humoral response to an allograft. Eye 9:155. 13. Sano, Y., B. R. Ksander, and J. W. Streilein. 1996. Minor H, rather than MHC, alloantigens offer the greater barrier to successful orthotopic corneal transplantation in mice. Transplant. Immunol. 4:53. 14. Whitsett, C. F., and R. D. Stulting. 1984. The distribution of HLA antigens on human corneal tissue. Invest. Ophthalmol. Vis. Sci. 25:519. 15. Wang, H. M., H. J. Kaplan, W. C. Chan, and M. Johnson. 1987. The distribution and ontogeny of MHC antigens in murine ocular tissue. Invest. Ophthalmol. Vis. Sci. 28:1383. 16. Gillette, T. E., J. W. Chandler, and J. V. Greiner. 1982. Langerhans cells of the ocular surface. Ophthalmology 89:700. 17. Sonoda, Y., Y. Sano, B. Ksander, and J. W. Streilein. 1995. Characterization of cell-mediated immune responses elicited by orthotopic corneal allografts in mice. Invest. Ophthalmol. Vis. Sci. 36:427. 18. He, Y. G., J. Ross, and J. Y. Niederkorn. 1991. Promotion of murine orthotopic corneal allograft survival by systemic administration of anti-CD4 monoclonal antibody. Invest. Ophthalmol. Vis. Sci. 32:2723. 19. Paul, W. E., and R. A. Seder. 1994. Lymphocyte responses and cytokines. Cell 76:241. 20. Nickerson, P., W. Steurer, J. Steiger, X. Zheng, A. W. Steele, and T. B. Strom. 1994. Cytokines and the Th1/Th2 paradigm in transplantation. Cur. Opin. Immunol. 6:757. 21. Dallman, M. J. 1995. Cytokines and transplantation: Th1/Th2 regulation of the immune response to solid organ transplants in the adult. Curr. Opin. Immunol. 7:632. Downloaded from http://www.jimmunol.org/ by guest on July 31, 2017 A vast literature is accumulating that links Th2 responses to successful solid organ transplants (20 –22, 26, 44). An early indication that IL-4-producing, rather than IL-2-producing, CD41 T cells contributed to allograft tolerance was reported from our laboratory in 1990 (24). We demonstrated in neonatal transplantation tolerance that the CD41 T cells that promote long term acceptance of class II MHC disparate skin allografts not only secreted IL-4 in response to the tolerated alloantigens, but these T cells also suppressed IL-2 production by alloreactive T cells in cocultures. Shortly thereafter, other investigators reported that the alloreactive T cells of rats bearing long accepted cardiac allografts secreted Th2-type cytokines. Subsequently, other forms of transplantation tolerance (22, 23, 44), including oral tolerance (45) and unresponsiveness induced by monoclonal anti-CD4 and anti CD8 Abs (26, 46), were also found to correlate with the emergence of donorspecific T cells that secreted Th2 cytokines. Thus, among the many forms of experimental transplantation tolerance in which the unresponsive state is maintained actively, donor-specific Th2 cells seem to emerge and contribute to maintenance of the tolerant state. However, in our cornea-grafting studies, donor-specific Th2 cells made a much more substantial contribution to graft success than merely maintaining an already established state of tolerance. Our findings indicate that mice with immune systems already biased toward Th2 responses failed to acquire (or only poorly so) donorspecific, allodestructive CD41 T cells, i.e., secrete IFN-g and mediate delayed hypersensitivity. Consequently, recipient mice were largely unable to mount a response capable of destroying their orthotopic cornea grafts. The capacity to prevent an immunopathogenic Th1-type response by pre-emptively creating a Th2-biased immune system offers a potential avenue of immunotherapy with broad applicability. To this end, Tian et al. have already reported that nasal administration of glutamate decarboxylase peptides to young NOD mice generates Th2 responses and prevents the emergence of insulin-dependent diabetes (47). While the ability of mice with Th2-biased immune systems to accept orthotopic corneal allografts in high risk eyes is impressive, the success of this approach may be inherently greater for this particular type of grafting. Corneal tissue itself expresses the inherent property of immune privilege (2, 3, 5), which means that as a graft it erects barriers to immune rejection. Moreover, orthotopic corneal allografts when placed in the eye form the anterior wall of the anterior chamber, an immune-privileged site. It is widely believed that immune privilege is the reason that orthotopic corneal allografts in humans are by far the most successful of all solid tissue grafts. Such may be the case, and for this reason we used recipient mice whose eyes were neovascularized and thus deprived of immune privilege (48). Both MHC and minor H disparate cornea grafts were rejected acutely in these high risk eyes (13). Our finding that mice with Th2-biased immune systems rejected a higher fraction of MHC plus minor H disparate corneas, compared with minor H only disparate corneas, warrants comment. Cornea tissue differs fundamentally from other solid tissue grafts in being virtually devoid of passenger leukocytes (4, 16). Cornea tissue contains neither Langerhans cells nor other bone marrow-derived cells, and thus does not express class II alloantigens (4, 15, 16, 49). Moreover, expression of class I molecules is very low on corneal endothelial cells and keratocytes (4, 15, 49). For these reasons, minor histocompatibility Ags have been determined to offer stronger barriers to corneal graft acceptance than MHC alloantigens, and the ability of recipient T cells to detect alloantigens on orthotopic cornea grafts rests primarily with the capacity of recipient APC to migrate into the graft (50). Thus, the majority of T cells that effect corneal allograft rejection detect and respond to graftderived alloantigens via the so-called indirect pathway of al- Th2 CELLS PROMOTE CORNEAL GRAFT SURVIVAL The Journal of Immunology 38. Romagnani, S. 1991. Human TH1 and TH2 subsets: doubt no more. Immunol. Today 12:256. 39. Abbas, A. K., K. M. Murphy, and A. Sher. 1996. Functional diversity of helper T lymphocytes. Nature 383:787. 40. Garside, P., and A. M. Mowat. 1995. Polarization of Th-cell responses: a phylogenetic consequence of nonspecific immune defense? Immunol. Today 16:220. 41. Piccotti, J., S. Chan, R. Goodman, J. Magram, E. Eichwald, and D. Bishop. 1996. IL-12 antagonism induces T helper 2 responses, yet exacerbates cardiac allograft rejection. J. Immunol. 157:1951. 42. VanBuskirk, A., M. Wakely, and C. Orosz. 1996. Transfusion of polarized TH2like cell populations into SCID mouse cardiac allograft recipients results in acute allograft rejection. Transplantation 62:229. 43. Joo, C. K., J. S. Pepose, and P. M. Stuart. 1995. T-cell mediated responses in a murine model of orthotopic corneal transplantation. Invest. Ophthalmol. Vis. Sci. 36:1530. 44. Takeuchi, T., R. P. Lowry, and B. Konieczny. 1992. Heart allografts in murine systems: the differential activation of Th2-like effector cells in peripheral tolerance. Transplantation 53:1281. 45. Hancock, W. W., M. H. Sayegh, C. A. Kwok, H. L. Weiner, and C. B. Carpenter. 1993. Oral, but not intravenous, alloantigen prevents accelerated allograft rejection by selective intragraft Th2 cell activation. Transplantation 55:1112. 46. Cobbold, S. P., E. Adams, S. E. Marshall, J. D. Davies, and H. Waldmann. 1996. Mechanisms of peripheral tolerance and suppression induced by monoclonal antibodies to CD4 and CD8. Immunol. Rev. 149:5. 47. Tian, J., M. A. Atkinson, M. Clare-Salzler, A. Herschenfeld, T. Forsthuber, P. V. Lehmann, and D. L. Kaufman. 1996. Nasal administration of glutamate decarboxylase (GAD65) peptides induces Th2 responses and prevents murine insulindependent diabetes. J. Exp. Med. 183:1561. 48. Streilein, J. W., D. Bradley, Y. Sano, and Y. Sonoda. 1996. Immunosuppressive properties of tissues obtained from eyes with experimentally manipulated corneas. Invest. Ophthalmol. Vis. Sci. 37:413. 49. Treseler, P. A., G. N. Foulks, and F. Sanfilippo. 1984. The expression of HLA antigens by cells in the human cornea. Am. J. Ophthalmol. 98:763. 50. Yamada, J., M. R. Dana, S. N. Zhu, P. Alard, and J. W. Streilein. 1998. Interleukin-1 receptor antagonist suppresses allosensitization in corneal transplantation. Arch. Ophthalmol. 116:1351. 51. Shoskes, D. A., and K. J. Wood. 1994. Indirect presentation of MHC antigens in transplantation. Immunol. Today 15:32. 52. Bradley, J. A., A. M. Mowat, and E. M. Bolton. 1992. Processed MHC class I alloantigen as the stimulus for CD41 T-cell dependent antibody-mediated graft rejection. Immunol. Today 13:434. 53. Sayegh, M. H., B. Watschinger, and C. B. Carpenter. 1994. Mechanisms of T cell recognition of alloantigen: the role of peptides. Transplantation 57:1295. Downloaded from http://www.jimmunol.org/ by guest on July 31, 2017 22. Lowry, R. P. 1993. The relationship of IL-4, IL-10, and other cytokines to transplantation tolerance. Transplant. Sci. 3:104. 23. Charlton, B., H. Auchincloss, Jr., and C. G. Fathman. 1994. Mechanisms of transplantation tolerance. Annu. Rev. Immunol. 12:707. 24. Powell, T. J., Jr. and J. W. Streilein. 1990. Neonatal tolerance induction by class II alloantigens activates IL-4-secreting, tolerogen-responsive T cells. J. Immunol. 144:854. 25. Chen, N., Q. Gao, and E. H. Field. 1996. Prevention of Th1 response is critical for tolerance. Transplantation 61:1076. 26. Mottram, P. L., W. R. Han, L. J. Purcell, I. F. McKenzie, and W. W. Hancock. 1995. Increased expression of IL-4 and IL-10 and decreased expression of IL-2 and interferon-g in long-surviving mouse heart allografts after brief CD4-monoclonal antibody therapy. Transplantation 59:559. 27. Finkelman, F. D., I. M. Katona, T. R. Mosmann, and R. L. Coffman. 1988. IFN-g regulates the isotypes of Ig secreted during in vivo humoral immune responses. J. Immunol. 140:1022. 28. Coffman, R. L., and T. R. Mosmann. 1991. CD41 T-cell subsets: regulation of differentiation and function. Res. Immunol. 142:7. 29. Kreukniet, M. B., A. J. van der Zijpp, and M. G. Nieuwland. 1992. Effects of route of immunization, adjuvant and unrelated antigens on the humoral immune response in lines of chickens selected for antibody production against sheep erythrocytes. Vet. Immunol. Immunopathol. 33:115. 30. Finkelman, F. D., I. M. Katona, J. F. Urban, Jr., J. Holmes, J. Ohara, A. S. Tung, J. V. Sample, and W. E. Paul. 1988. IL-4 is required to generate and sustain in vivo IgE responses. J. Immunol. 141:2335. 31. DeKruyff, R. H., Y. Fang, and D. T. Umetsu. 1992. IL-4 synthesis by in vivo primed keyhole limpet hemocyanin-specific CD41 T cells. I. Influence of antigen concentration and antigen-presenting cell type. J. Immunol. 149:3468. 32. Dana, M. R., J. Yamada, and J. W. Streilein. 1997. Topical interleukin 1 receptor antagonist promotes corneal transplant survival. Transplantation 63:1501. 33. Taylor, A. W., J. W. Streilein, and S. W. Cousins. 1992. Identification of a-melanocyte stimulating hormone as a potential immunosuppressive factor in aqueous humor. Curr. Eye Res. 11:1199. 34. Falcone, M., and B. R. Bloom. 1997. A T helper cell 2 (Th2) immune response against non-self antigens modifies the cytokine profile of autoimmune T cells and protects against experimental allergic encephalomyelitis. J. Exp. Med. 185:901. 35. Hsieh, C. S., S. E. Macatonia, C. S. Tripp, S. F. Wolf, A. O’Garra, and K. M. Murphy. 1993. Development of TH1 CD41 T cells through IL-12 produced by Listeria-induced macrophages. Science 260:547. 36. Trinchieri, G. 1995. Interleukin-12: a proinflammatory cytokine with immunoregulatory functions that bridge innate resistance and antigen-specific adaptive immunity. Annu. Rev. Immunol. 13:251. 37. Mosmann, T. R., J. H. Schumacher, N. F. Street, R. Budd, A. O’Garra, T. A. Fong, M. W. Bond, K. W. M. Moore, A. Sher, and D. F. Fiorentino. 1991. Diversity of cytokine synthesis and function of mouse CD41 T cells. Immunol. Rev. 123:209. 5255
© Copyright 2025 Paperzz