首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 31 毫秒
1.
The staphylococcal enterotoxins have been termed superantigens based on their ability to stimulate polyclonal proliferative responses of murine and human T lymphocytes expressing particular T-cell receptor Vβ gene products. Certain of these toxins have been shown both to activate and to induce anergy in reactive T cells. Staphylococcal enterotoxin B is known to interact with murine T cells bearing Vβ3, ?7, ?8.1, ?8.2, ?8.3, and ?17. In BALB/c mice Vβ3+ and Vβ17+ T cells are deleted; Vβ7+ T cells are low in frequency. BALB/c mice sensitized to ovalbumin via the skin and airways develop immediate hypersensitivity including IgE/IgGl antiovalbumin antibodies, immediate cutaneous reactivity to ovalbumin and, increased airway responsiveness. In bothin vitro andin vivo studies, the development of these responses has been associated with the Vβ8+ subset of T cells and controlled by Vβ2+ T cells. In view of the central role of Vβ8+ T cells in these responses, we tested the effects of staphylococcal enterotoxin B on the development of immediate hypersensitivity in this system. Intradermal injection of staphylococcal enterotoxin B prevented the development of these responses in the absence of a major deletion of Vβ8+ T cells. The data suggest that the administration of staphylococcal enterotoxin B prevented the antigen-induced expansion of Vβ8+ T cells resulting in a state of unresponsiveness or anergy, thus preventing the manifestations of immediate hypersensitivity. Bacterial toxins may provide a novel approach to intervention in allergic or autoimmune diseases.  相似文献   

2.
The present study aims at investigating the intrathymic maturational stage of T cells at which clonal deletion can be induced. Staphylococcal enterotoxin B (SEB) was added to organ cultures of murine fetal thymus lobes at various time points of culture, and Vβ8-expressing cells were assayed on day 14. Vβ8 low-expressing (Vβ8′°) cells were reduced to 40-60 % of the control receiving no SEB, though the reduction was ambiguous when SEB was given on day 13. In marked contrast, Vβ8 high-expressing (Vβ8hi) cells were virtually completely deleted in all groups including the group given SEB on day 13. Most of the Vβ8hl cells that were deleted by 24 h of treatment with SEB were shown to be of the CD4+8 mature phenotype, though CD4?8+Vβ8hi cells were also deleted. It was further shown that the thymic Vβ8hi CD4+8? cells recovered from organs cultured for 14 days without SEB responded to immobilized anti-Vβ8 monoclonal antibody, indicating that Vβ8hi cells, which were highly sensitive to clonal deletion, were functionally competent mature T cells. These results strongly suggest that the thymus is capable of eliminating all T cells recognizing antigen present in the thymus regardless of the maturational stage of T cells.  相似文献   

3.
The staphylococcal enterotoxins stimulate discrete subsets of T cells depending on their expression of particular V genes. Among these, staphylococcal enterotoxin B (SEB) vigorously stimulates Vβ8+ cells. This stimulation results in proliferation of both CD4 + Vβ8 + and CDS + T cells and eventually to anergy and clonal deletion in the former subset. We have examined the possible role of CD8+ T cells in the response of CD4+ cells to SEB, by in vivo CD8+ T-cell-depletion. We found no qualitative difference in the responses of untreated and CD8+ T-cell depleted mice to SEB; however, a small quantitative difference in deletion was observed. Thus it appears that on the whole the response of CD4 +Vβ8 + T cells to SEB is independent of CD8+ T-cell effector function, although the latter may play a partial role.  相似文献   

4.
Staphylococcal enterotoxin B (SEB) can activate specific T cell clones bearing specific TcR Vβ domains together with MHC class II ligands on accessory cells. The release of proinflammatory cytokines is the consequence of this activation as well as the main pathological aspect involved in SEB infection. This current study looked at the active role of both T and B cells during the induction of anergy by SEB in vivo Euthymic and nude BALB/c mice were injected with SEB and over a period of 8 days, cells from the spleen and sera from the blood were collected. After a single injection with SEB (50 μg/mouse), a transient increase of CD4+Vβ8+ T cells were detected after 2 days followed by a decrease after 4 days, which persisted until day 8. These clones were rendered anergic upon restimulation in vitro with SEB. Interestingly, cells taken out 2 days after SEB injection, exhibited reduced proliferation in response to Con A. However, this response gradually recovered on days 4, 6 and 8. Furthermore, early IgM antibody production (day 2) was observed after SEB injection. SEB-induced IgM antibody production in euthymic BALB/c was found to have specificity against SEB, cardiolipin (CL) and phosphatidylethanolamine (PE). SEB-treated nude mice did not produce antibody secreting cells in response to SEB, indicating that this process is T cell dependent.  相似文献   

5.
Ligand-specific immunosuppression requires an understanding of the parameters that control peripheral T cell tolerance. T cell receptor (TcR) transgenic mice offer a clear advantage for studying post-thymic tolerance mechanisms in vivo that are operational in a monoclonal T cell population with preselected antigen specificity. Yet it is unclear whether the rules defined in monoclonal T cells of genetically manipulated mice reflect those operative in clonally diverse peripheral T cells of normal mice. To analyze acute tolerance mechanisms in unselected peripheral T cells, we challenged normal mice with the superantigen staphylococcal enterotoxin B (SEB) and analyzed ligand-reactive Vβ8+ T cells for TcR-triggered tolerance mechanisms such as anergy, TcR down-regulation, or apoptosis. Upon challenge with graded doses of SEB (0.001–10 μg) Vβ8? T cells become anergic within 6–16 h. Importantly, a dosage effect of SEB in regard to the level of anergy induced was observed. Anergy induced by low concentrations of SEB (0.001–0.1 μg) is transient and is overcome by clonal growth, while higher concentrations of SEB (0.1–10 μg) cause long-lasting anergy resistant to cell cycle progression. At high SEB concentrations (1–10 mg) about 50% of the anergic Vβ8+ T cells additionally down-regulate their TcR-CD3 complex, followed by a loss of CD2, CD4, CD8 accessory molecules. In parallel, T cell phenotypenegative but genotypically Vβ8+ T cells are generated. The T cell phenotypenegative cells reacquire their Vβ8+ T cell phenotype upon culture in vitro. In vivo, a subset of Vβ8+ cells, defined by an intermediate stage of TcR down-regulation, i.e. Vβ8lowCD3+ cells, but not T cell phenotype-negative cells are selectively programmed for apoptosis, which occurs within 1 h. These data suggest that SEB triggers distinct tolerance pathways which operate in a hierarchical fashion in clonally diverse ligand-reactive T cells. Specifically, the results illustrate the power of exogenous superantigens to exploit these distinct tolerance pathways, thereby achieving distinct levels of immunosuppression.  相似文献   

6.
The in vitro T cell nonresponsiveness or anergy to restimulation with staphylococcal enterotoxin B (SEB) following the in vivo injection of the superantigen is well characterized. Here we use mice transgenic for a Vβ8.2+ T cell receptor (TcR) (reactive with SEB) to establish a large population of anergic T cells in vivo. As expected, peripheral T cells from the SEB injected transgenic mice failed to proliferate or produce interleukin (IL)-2 following restimulation with the superantigen in vitro. However, in this system superantigen reactivity could be restored by either addition of exogenous IL-2, or stimulation with immobilized anti-TcR antibody. To evaluate the effects of superantigen-induced anergy in vivo, SEB-injected or noninjected control transgenic mice were immunized and boosted with the T cell-dependent antigen tetanus toxin (TT). SEB injection of the Vβ8.2+ transgenic mice 5 days prior to the TT immunization inhibited the anti-TT antibody response as measured over a 100-day period, whereas injection of a superantigen which does not interact with the Vβ8.2+ TcR (such as SEA) did not. Furthermore, SEB injection of control nontransgenic mice did not interfere with the induction of a high titer anti-TT antibody response. In contrast to the inhibition seen when SEB was given prior to TT immunization, injection of transgenics with SEB either after the priming TT immunization or after the recall booster injection did not significantly influence the titers of anti-TT antibodies produced. These results demonstrate that the establishment of peripheral T cell anergy to superantigens inhibits the specific antigenic priming of helper T cells in vivo, but does not prevent primed T cells from helping B cells to mount an effective antibody response.  相似文献   

7.
The staphylococcal enterotoxin is a major cause of food poisoning. The bacterial substance stimulates T cells expressing specific Vβ T cell receptors (TcR) and is termed “the superantigen”. We have previously demonstrated that intravenous injection of staphylococcal enterotoxin B (SEB) induces functional unresponsiveness (anergy) of reactive T cells as well as a partial deletion by activationinduced programmed cell death. In the present study, we examined the effect of oral administration of SEB in mice. Our results indicate that spleen T cells from SEB-primed mice are hyporesponsive to SEB stimulation in vitro, but the response to SEA was normal. Vβ8+ T cells purified from SEB-primed mice did not respond to stimulation of TcR. This SEB-specific unresponsiveness could not be reversed by exogenous interleukin-2, but was partially reversed by phorbol 12-myristate 13-acetate. Activation of mitogen-activated protein kinase during TcR-mediated stimulation was significantly inhibited in anergic T cells. Although the mechanisms of oral tolerance are not well understood, these results show that oral administration of SEB induce clonal anergy in peripheral T cells.  相似文献   

8.
The CD28/CTLA-4 receptors on T cells interact with the B7 molecule on antigen-presenting cells (APC) to produce a co-stimulatory signal that determines the outcome of activation. The role of this co-stimulatory signal in T cell activation and loss of tolerance in autoimmune MRL-lpr/lpr mice has not been investigated previously. The present study examines the contribution of the CD28/CTLA-4 co-stimulatory pathway to the loss of T cell tolerance in Vβ8 transgenic MRL-lpr/lpr and -+/+ mice in which neonatal tolerance has been induced by the superantigen staphylococcal enterotoxin B (SEB). An artificial APC transfected with the murine B7 gene, and a CTLA-4-Ig fusion protein were used to analyze the significance of the CD28/CTLA-4 pathway in vitro. The CTLA-4-Ig fusion protein was also used to inhibit the pathway in vivo. Our results demonstrate that CD28 and CTLA-4 mRNA was overexpressed in the lymph nodes of lpr/lpr mice (MRL, C57BL/6, C3H and AKR), but not in +/+ mice of the same background strain. Lymph node T cells and thymocytes from SEB neonatally tolerized MRL-lpr/lpr mice that had undergone tolerance loss, proliferated when cultured with SEB and B7+ fibroblasts in vitro, but did not proliferate when the SEB was presented in the context of B7? fibroblasts. This in vitro tolerance loss could be prevented by blocking of B7 signaling by CTLA-4-Ig. This loss of tolerance did not occur in lymph node T cells from thymectomized MRL-lpr/lpr mice. SEB challenge of tolerized MRL-lpr/lpr mice in vivo led to weight loss, increased serum cytokine levels and depletion of Vβ8+ T cells. These effects were blocked by blocking of the co-stimulatory pathway by treatment with the CTLA-4-Ig fusion protein prior to and during challenge with SEB. T cells from thymus and lymph nodes of these mice did not proliferate later in response to stimulation in vitro with SEB even in the presence of B7+ APC. Nonresponsiveness was not due to deletion of Vβ8+ CD28+ T cells, as the number of these cells was increased after treatment with SEB and the CTLA-4-Ig fusion protein. These results suggest that the response of autoreactive T cells in the thymus and lymph nodes depends on signaling by B7 in vivo and in vitro and that SEB-reactive T cells can be reanergized in vivo by stimulation of the T cell receptor in the absence of signaling through the CD28/CTLA-4 co-stimulatory pathway.  相似文献   

9.
Injection of the bacterial superantigen Staphylococcus aureus enterotoxin B (SEB) into mice provokes a rapid expansion and subsequent contraction of the pool of SEB-reactive T cells bearing T cell receptor (TcR) Vβ8 gene products. Given that interleukin 2 (IL-2) stimulates proliferation, abolishes anergy, and counteracts apoptotic cell death in T cells in vitro, we tested whether the IL-2 synthesis inhibitor cyclosporin A (CsA) or a vaccinia virus recombinant releasing high amounts of human IL-2 modulate SEB responses in vivo. Surprisingly, neither IL-2 nor CsA were able to change the in vivo kinetics and magnitude of SEB-induced expansion, unresponsiveness to SEB, and peripheral clonal deletion of T cells expressing products of the SEB-reactive TcR Vβ8 gene family. In accord with these in vivo observations, IL-2 is incapable of reversing “anergy” and apoptotic cell death of Vβ8+ SEB-reactive T cells isolated from SEB-primed mice in vitro. Accordingly, upon SEB injection Vβ8+ T cells expand rapidly, without expressing IL-2 receptor (IL-2R)α chains in vivo, although SEB induces IL-2R α in vitro. Altogether, these results indicate that the IL-2/IL-2R-mediated pathway is not involved in T cell repertoire modulation by bacterial superantigens. Moreover, the data suggest that unresponsiveness of Vβ8+ T cells from SEB-primed mice is not a reversible process, but involves an unreversible commitment to programmed cell death. Absence or presence of IL-2 responsiveness could be a hallmark to distinguish truly reversible anergy and peripheral clonal deletion.  相似文献   

10.
The authors were interested to investigate the effect of Cyclosporin A (CsA), known to block interleukin-2 (IL-2) production, or of anti-interferon-γ antibodies (anti-IFN-γ Abs) in a model of T cell tolerance induced by the injection of the superantigen Staphylococcal Enterotoxin B (SEB) in BALB/c mice. After SEB immunization, tolerance was mainly achieved through deletion and anergy of SEB-reactive Vβ8+ T cells. Association of CsA treatment with SEB led to a greater decrease of the percentage of Vβ8+ CD4+ lymphocytes in the spleen and an abolition of clonal anergy. In contrast, treatment of SEB primed mice with anti-IFN-γ Abs resulted in an increased percentage of Vβ8+ CD4+ cells without affecting the induction of clonal anergy. The authors found that 1–2 h after SEB priming, splenic mRNA levels of IFN-γ and IL-4 were decreased by either CsA and anti-IFN-γ Abs, whereas FasL, Bcl-2, p. 53, and c-myc levels were not influenced by either treatment. However, SEB-induced IL-2 and IL-10 mRNA expression was suppressed only by CsA, whereas tumour necrosis factor-α (TNF-α) was decreased only by anti-IFN-γ Abs. To investigate whether the effect of CsA on the tolerance mechanisms was related to suppression of IL-2, CsA was administered together with recombinant IL-2. Whereas anergy was not influenced, the decreased percentage of Vβ8+ CD4+ cells seen in CsA-treated animals in the second week after SEB injection was partially corrected by the administration of IL-2. Experiments involving bromodeoxiuridine incorporation revealed that the latter effect of IL-2 was mainly due to a correction of the defective proliferation of Vβ8+ T cells after SEB injection in CsA-treated mice. These results suggest that the effect of CsA and anti-IFN-γ Abs on tolerance mechanisms are in part explained by their action on cytokines.  相似文献   

11.
There is a growing body of evidence which suggests that CD8+ T cells play an important part in regulating the IgE response to non-replicating antigens. In this study we have systematically investigated their role in the regulation of IgE and of CD4+ T cell responses to ovalbumin (OVA) by CD8+ T cell depletion in vivo. Following intraperitoneal immunization with alum-precipitated OVA, OVA-specific T cell responses were detected in the spleen and depletion of CD8+ T cells in vitro significantly enhanced the proliferative response to OVA. Depletion of CD8+ T cells in vivo 7 days after immunization failed to enhance IgE production, while depletion of CD8+ T cells on days 12–18 greatly enhanced the IgE response, which rose to 26 μ/ml following a second injection of anti-CD8 on day 35 and remained in excess of 1 μ/ml over 300 days afterwards. Reconstitution on day 21 of rats CD8-depleted on day 12 with purified CD8+ T cells from animals immunized on day 12 completely inhib ited the IgE response. This effect was antigen specific; CD8+ T cells from OVA-primed animals had little effect on the IgE response of bovine serum albumin immunized rats. In vivo, CD8+ T cell depletion decreased interferon (IFN)-γ production but enhanced interleukin (IL)-4 production by OVA-stimulated splenic CD4+ T cells. Furthermore, CD8+ T cell depletion and addition of anti-IFN-γ antibody enhanced IgE production in vitro in an IL-4-supplemented mixed lymphocyte reaction. These data clearly show that antigen-specific CD8+ T cells inhibit IgE in the immune response to non-replicating antigens. The data indicate two possible mechanisms: first, CD8+ T cells have direct inhibitory effects on switching to IgE in B cells and second, they inhibit OVA-specific IL-4 production but enhance IFN-γ production by CD4+ T cells.  相似文献   

12.
Injection of bacterial superantigens such as staphylococcal enterotoxin B (SEB) in adult mice results in initial proliferation of SEB-responsive Vβ8+ T cells followed by induction of a state of non-responsiveness frequently referred to as clonal anergy. We show here that SEB-induced anergy involves selective changes in lymphokine production and that it affects CD4+ Vβ 8+ and CD8+ Vβ 8+ T cells in different fashions. Whereas both CD4+ Vβ 8+ and CD8+ Vβ 8+ cells from anergic mice exhibit strongly reduced proliferative capacity and interleukin(IL)-2 production upon restimulation with SEB either in vivo or in vitro the CD8+ subset from SEB-injected mice produces other lymphokines (such as interferon(IFN)-γ) at normal or slightly increased levels in response to SEB. Changes in the levels of production of IL-2 and IFN-γ protein correlated well with mRNA accumulation both in vivo and in vitro. Collectively these data suggest that superantigen-induced anergy involves selective changes in signal transduction and/or gene regulation in T lymphocytes.  相似文献   

13.
Staphylococcal enterotoxin B (SEB) activates T cells via non‐canonical signalling through the T cell receptor and is an established model for T cell unresponsiveness in vivo. In this study, we sought to characterize the suppressive qualities of SEB‐exposed CD4+ T cells and correlate this with genetic signatures of anergy and suppression. SEB‐exposed CD25+ and CD25Vβ8+CD4+ T cells expressed forkhead box P3 (FoxP3) at levels comparable to naive CD25+ T regulatory cells and were enriched after exposure in vivo. Gene related to anergy in lymphocytes (GRAIL), an anergy‐related E3 ubiquitin ligase, was up‐regulated in the SEB‐exposed CD25+ and CD25FoxP3+Vβ8+CD4+ T cells and FoxP3CD25Vβ8+CD4+ T cells, suggesting that GRAIL may be important for dominant and recessive tolerance. The SEB‐exposed FoxP3+GRAIL+ T cells were highly suppressive and non‐proliferative independent of CD25 expression level and via a glucocorticoid‐induced tumour necrosis factor R‐related protein‐independent mechanism, whereas naive T regulatory cells were non‐suppressive and partially proliferative with SEB activation in vitro. Lastly, adoptive transfer of conventional T cells revealed that induction of FoxP3+ regulatory cells is not operational in this model system. These data provide a novel paradigm for chronic non‐canonical T cell receptor engagement leading to highly suppressive FoxP3+GRAIL+CD4+ T cells.  相似文献   

14.
《Mucosal immunology》2009,2(5):427-438
The role of CD8+ T cells in oral tolerance remains unclear. To address this, we developed a model to induce CD8+ Tregs by feeding the major histocompatibility complex class I immunodominant epitope of OVA, OVA(257–264). OVA(257–264) feeding induced tolerance similar to that observed in OVA protein-fed mice, capable of suppressing the production of Th1 and Th17 cytokines and inhibiting a Th1-driven delayed-type hypersensitivity response following immunization with whole OVA (wOVA) protein. OVA(257–264) peptide-induced suppression could be transferred to naive mice with CD8+ cells, but not CD8-depleted cells, isolated from mesenteric lymph nodes of peptide-fed mice. Interestingly, while capable of inhibiting Th1 and Th17 responses, OVA(257–264) feeding could not suppress any feature of a Th2 inflammatory response, though OVA protein feeding could, suggesting that these cells function through a different mechanism than their CD4+ counterparts generated in response to feeding with wOVA. Thus, CD8+ T cells are functionally capable of mediating tolerance to Th1 and Th17 responses.  相似文献   

15.
Aside from CD4+CD8+ double-positive (DP) thymocytes, the subpopulations of T lineage cells affected by negative selection are unknown. To address whether this process occurs in more mature cell types, we have compared the responses of purified single-positive (SP) murine thymocytes and peripheral T cells to the superantigen staphylococcal enterotoxin B (SEB) utilizing as antigen-presenting cells (APC) a fibroblast cell line expressing transfected I-Ek class II molecules. Whereas ∽ 70% of SEB-reactive SP thymocytes, either CD4+ or CD8+, undergo programmed cell death (apoptosis) and, therefore, negative selection, CD4+ and CD8+ antigen-specific peripheral T cells are predominantly activated and proliferate to APC+SEB. Thus, mature thymocytes and peripheral T cells, with identical patterns and levels of expression of CD4, CD8 and T cell receptor (TCR), are programmed to elicit different responses followingTCR stimulation. Unexpectedly, however activation of peripheral T cells was preceded by deletion of a large fraction of Vβ8+ T lymphocytes (SEB specific). This surprising phenomenon was also observed in in vivo studies: in fact, administration of SEB to adult mice resulted in depletion of the majority of antigen-specific T cells from the peripheral lymphoid tissues analyzed (lymph nodes and spleen). This depletion is the consequence of deletion as indicated by program cell death of Vβ8+ T cells and is followed by proliferation of the remaining SEB-reactive T cells. Clonal elimination of peripheral T cells may represent a mechanism by which tolerance to self antigens never expressed in and/or exported to the thymus is achieved.  相似文献   

16.
As compared with the native molecule, recombinant human interleukin-2 that is modified by covalently attached polyethylene glycol residues (IL-2-PEG) exhibits a markedly enhanced half-lifein vivo, thus facilitating its biological evaluation. We have characterized the effect of IL-2-PEG on theStaphylococcus aureusenterotoxin B (SEB)-induced tolerance of peripheral SEB-reactive (Vβ8+) T cells. Treatment with sublethal doses of IL-2-PEG does not modulate (inhibit or enhance) the SEB-triggered apoptosis and deletion of Vβ8+T cells. In contrast,in vivotreatment with IL-2-PEG partially abolishes the SEB-triggered anergy of Vβ8+T cells, i.e., the failure to proliferate in response to SEBin vitro. To abolish SEB-triggered anergy, IL-2-PEG must act for an extended periodin vivo; short term treatmentin vivo(2 days) or exposure of anergic T cells to IL-2in vitrofails to reconstitute proliferative responses. Moreover, the effect ofin vivotreatment with IL-2-PEG on lymphokine production by anergic T cells is partial. IL-2-PEG restores IL-4-dependent autocrine proliferation in response to SEB but does not reestablish defective IL-2 production. These data are compatible with the notion that IL-2 is a regulator of postdeletional rather than deletional T cell tolerance.  相似文献   

17.
We produced transgenic mice expressing Tcell receptor-αβ chain genes, derived from the chicken ovalbumin (OVA)-specific I-Ad-restricted CD4+CD8? T helper cell clone 7–3–7. In transgenic mice with H-2d genetic background (Tg-d mice), delayed-type hypersensitivity (DTH) was induced in the hind footpad by one inoculation with OVA without any previous sensitization, suggesting that naive T cells have the potential to be involved in DTH response. Spleen cells from nonimmunized Tg-d mice showed a strong T cell proliferative response to in vitro stimulation with OVA. Furthermore, these spleen cells produce cytokines including interleukin(IL)-2, IL-3, interferon-γ, granulocyte/macrophage colony-stimulating factor, macrophage inflammatory protein (MIP)-1α and MIP-1β, which may play an important role in the attraction of mononuclear cells to an antigen-challenging site.  相似文献   

18.
The complementary receptor pair Fas ligand: Fas controls apoptosis during activation-induced cell death (AICD) of peripheral T cells sensitized for the Fas signal pathway by interleukin-2 (IL-2). In the present study, we used the bacterial superantigen staphylococcal enterotoxin B (SEB) to anergize ligand-reactive peripheral T cells in wild-type and Fas-defective lpr mice. In a second step, we investigated whether apoptosis in anergized and thus operationally IL-2-defective peripheral T cells is triggered via the Fas signal pathway. We report here that SEB-driven anergy induction and deletion of anergized peripheral Vβ8+ T cells is similar in wild-type and healthy C3H/lpr mice. In monitoring SEB-driven Vβ8+ T cell apoptosis in situ, we observe in both wild-type and lpr mice an intimate association between proliferation and apoptosis of anergized Vβ8+ T cells. We further show that Vβ8+ T cells activated in vitro from wild-type mice express a Fas-sensitive phenotype determined by Fas cross-linking which causes apoptosis. In contrast, Vβ8+ T cells anergized in vivo from wild-type mice are Fas resistant. As expected, T cells from lpr mice activated in vitro or anergized in vivo are Fas resistant. Taken together, these data indicate that both in wild-type and Fas-defective C3H/lpr mice, anergized T cells become deleted via a Fas-independent, proliferation-associated apoptosis signal pathway.  相似文献   

19.
Ultrafine nanoparticles of zinc oxide (ZnO) recently became available as a substitute for larger-size fine ZnO particles. However, the biological activity of ultrafine ZnO currently remains undefined. In the present study, we investigated the effect of ultrafine ZnO on oral tolerance that plays an important role in the prevention of food allergy. Oral tolerance was induced in mice by a single oral administration (i.e., gavage) of 25?mg of ovalbumin (OVA) 5 days prior to a subcutaneous immunization with OVA (Day 0). Varying doses of ultrafine (diameter: ≈ 21 nm) as well as fine (diameter: < 5 µm) ZnO particles were given orally at the same time during the OVA gavage. The results indicated that a single oral administration of OVA was followed by significant decreases in serum anti-OVA IgG, IgG1, IgG2a, and IgE antibodies and in the proliferative responses to the antigen by these hosts’ spleen cells. The decreases in these immune responses to OVA were associated with a marked suppression of secretion of interferon (IFN)γ, interleukin (IL)-5, and IL-17 by these lymphoid cells. Treatment with either ultrafine or fine ZnO failed to affect the oral OVA-induced suppression of antigen-specific IgG, IgG1, IgG2a, and IgE production or lymphoid cell proliferation. The suppression induced by the oral OVA upon secretion of IFNγ, IL-5, and IL-17 was also unaffected by either size of ZnO. These results indicate that ultrafine particles of ZnO do not appear to modulate the induction of oral tolerance in mice.  相似文献   

20.
Although sublingual (s.l.) immunotherapy with selected allergens is safe and often effective for treating patients with allergies, knowledge of the immunological mechanisms involved remains limited. Can s.l. administration of antigen (Ag) induce peripheral immunological tolerance and also suppress delayed‐type hypersensitivity (DTH) responses? To what extent can s.l.‐induced tolerance be explained by the generation of Foxp3+CD25+CD4+ regulatory T cells (Treg)? This study addressed these questions in mice and compared the relative efficacy of administering ovalbumin (OVA) conjugated to cholera toxin B (CTB) subunit with administration of the same Ag alone. We found that s.l. administration of a single or even more efficiently three repeated 40‐μg doses of OVA/CTB conjugate suppressed T‐cell proliferative responses to OVA by cervical lymph node (CLN), mesenteric lymph node (MLN) and spleen cells and concurrently strongly increased the frequency of Ag‐specific Treg in CLN, MLN and spleen and also transforming growth factor‐β (TGF‐β) levels in serum. The CLN and splenic cells from OVA/CTB‐treated BALB/c mice efficiently suppressed OVA‐specific T‐cell receptor (TCR) transgenic (DO11.10) CD25?CD4+ effector T‐cell proliferation in vitro. Further, s.l. treatment with OVA/CTB completely suppressed OVA‐specific DTH responses in vivo and T‐cell proliferative responses in mice immunized subcutaneously with OVA in Freund's complete adjuvant. The intracellular expression of Foxp3 was strongly increased in OVA‐specific (KJ1‐26+) CD4+ T cells from OVA/CTB‐treated mice. Thus, s.l. administration of CTB‐conjugated Ag can efficiently induce peripheral T‐cell tolerance associated with strong increases in serum TGF‐β levels and in Ag‐specific Foxp3+CD25+CD4+ Treg cells.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号