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 15 毫秒
1.
BALB/c mice injected at birth with semi-allogeneic F1 spleen cells become tolerant to alloantigens as shown by their CTL unresponsiveness to the corresponding alloantigen and the persistence of donor F1 cells into the BALB/c host. Moreover, these mice develop a transient systemic lupus erythematosis-like autoimmune syndrome characterized by splenomegaly, glomerulonephritis, thrombocytopenia and abnormal serological findings, such as several autoantibodies and IgG1 hypergammaglobulinemia. Recent studies done in our laboratory have shown that donor F1 B cells persisting in the host are responsible for the production of autoantibodies and must be activated in vivo by the host CD4+ T lymphocytes in a MHC class II-restricted fashion. In the present work, we have focused our attention on the ability of splenic CD4+ T cells recovered at different periods from BALB/c mice injected at birth with (CBA/Ca × BALB. Ighb) Fl spleen cells to interact with and activate F1 semi-allogeneic spleen cells in vitro. We show that (i) only CD4+ T cells from 2- and 3-week-old tolerant BALB/c mice preferentially produce IL-4 and IL-5 in response to a F1 semi-allogeneic in vitro stimulation, (ii) CD4+ T cells purified from 3-week-old tolerant BALB/c mice are able to induce in vitro IgG and IgM production by F1 B cells. Taken together, these results strongly suggest that host CD4+ T cells, belonging to the TH2 subset progressively lose their reactivity towards the F1 semi-allogeneic persistent B cells, reaching a state of unresponsiveness that correlates with the disappearance of serum autoantibodies and autoimmune pathology.  相似文献   

2.
Nasal administration of peptide antigens has been shown to induce T cell tolerance. We have investigated the potential for peptide therapy of the autoimmune response to myelin antigens in experimental autoimmune encephalomyelitis (EAE). Three major encephalitogenic epitopes were studied for their ability to induce nasal tolerance to myelin antigens. These included epitopes Ac1 – 9 and 89 – 101 of myelin basic protein (MBP) and 139 – 151 from proteolipid protein (PLP). Peptide Ac1 – 9 from MBP effectively suppressed responses to both MBP epitopes, following immunization with whole myelin (linked suppression). The N-terminal epitope failed, however, to modify the response to epitope 139 – 151 of PLP. The second MBP epitope (89 – 101) was poorly tolerogenic for the immune response to any naturally processed myelin epitope. By contrast, PLP[139 – 151] was able to induce bystander suppression of T cells responsive to both itself and the two epitopes from MBP. Furthermore, this epitope suppressed EAE induced with peptides derived from MBP and was capable of treating ongoing disease. The mechanism of bystander suppression, mediated by PLP[139 – 151], did not correlate with an overt switch from the T helper 1 to the T helper 2 phenotype. These results demonstrate how a complex autoimmune disease may be controlled by treatment with a single peptide epitope and reveal a hierarchy in the suppressive properties of different myelin antigens.  相似文献   

3.
The injection of (C57BL/6 × BALB/c)F1 spleen cells into newborn BALB/c mice results in the induction of a specific cytotoxic T lymphocyte (CTL) tolerance to the alloantigens. On the contrary, alloreactive CD4+ T cells persist in the host and are still able to activate autoreactive F1 B cells to produce autoantibodies. This state of “split tolerance” is closely associated with the development of a lupus-like autoimmune syndrome. The LFA-1 integrin plays a relevant role in homing, intercellular adhesion and tranduction of co-stimulatory signals in leukocytes. Because of the beneficial effects of anti-LFA-1 monoclonal antibodies (mAb) treatment in various models of organ transplantation and autoimmune disease, we have investigated if such a treatment could interfere with the induction of neonatal tolerance or the development of the autoimmune syndrome in F1 cell-injected newborn mice. For this purpose, BALB/c mice neonatally injected with F1 cells were treated from day 1 up to day 15 with a non-cytotoxic anti-LFA-1 (CD11a) mAb. Anti-LFA-1 mAb treatment interfered with the persistence of a stable chimerism and with the establishment of CTL tolerance, as shown by rejection of allogeneic skin grafts and F1 B cells, and by a normal in vitro CTL activity against the corresponding alloantigens. As a consequence, these mice did not develop the characteristic autoimmune features seen in close association with an effective induction of CTL tolerance to alloantigens. These results stress the importance of the interactions between LFA-1 and its ligands during the neonatal induction of tolerance to alloantigens.  相似文献   

4.
CD4(+) CD25(+) regulatory T cells are increasingly recognized as central players in the regulation of immune responses. In vitro studies have mostly employed allogeneic or polyclonal responses to monitor suppression. Little is known about the ability of CD4(+) CD25(+) regulatory T cells to suppress antigen-specific immune responses in humans. It has been previously shown that CD4(+) CD25(+) regulatory T cells anergize CD4(+) T cells and turn them into suppressor T cells. In the present study we demonstrate for the first time in humans that CD4(+) CD25(+) T cells are able to inhibit the proliferation and cytokine production of antigen specific CD4(+) and CD8(+) T cells. This suppression only occurs when CD4(+) CD25(+) T cells are preactivated. Furthermore, we could demonstrate that CD4(+) T-cell clones stop secreting interferon-gamma (IFN-gamma), start to produce interleukin-10 and transforming growth factor-beta after coculture with preactivated CD4(+) CD25(+) T cells and become suppressive themselves. Surprisingly preactivated CD4(+) CD25(+) T cells affect CD8(+) T cells differently, leading to reduced proliferation and reduced production of IFN-gamma. This effect is sustained and cannot be reverted by exogenous interleukin-2. Yet CD8(+) T cells, unlike CD4(+) T cells do not start to produce immunoregulatory cytokines and do not become suppressive after coculture with CD4(+) CD25(+) T cells.  相似文献   

5.
Peripheral tolerance is considered to be a safeguard against autoimmunity. Using a TCR-transgenic mouse system displaying peripheral tolerance against a liver-specific MHC class I Kb antigen, we investigated whether the breaking of tolerance would result in autoimmunity. Reversal of tolerance was achieved by simultaneous challenge with cells expressing the Kb autoantigen and IL-2. Tolerance could not be broken with IL-2 alone or when Kb- and IL-2-expressing cells were applied to different sites of the mice. However, despite the presence of activated autoreactive T cells that were able to reject Kb-positive grafts no autoaggression against the Kb-positive liver was observed. These results indicate that breaking of tolerance per se is not sufficient to cause liver-specific autoimmunity. However, when in addition to breaking tolerance the mice were infected with a liver-specific pathogen, autoaggression occurred. Thus, in this system at least two independent steps seem to be required for organ-specific autoimmunity: reversal of peripheral tolerance resulting in functional activation of autoreactive T cells and conditioning of the liver microenvironment which enables the activated T cells to cause tissue damage.  相似文献   

6.
The injection of (C57BL/6 x BALB/c) F1 spleen cells into BALB/c newborn mice leads to activation of persisting F1 donor B cells and development of a lupus-like syndrome in tolerized BALB/c mice. This syndrome is characterized by hypergammaglobulinaemia, high levels of anti-DNA and anti-Sm antibodies, circulating immune complexes and deposits of immunoglobulin in renal glomeruli. The role of donor T cells in this model was investigated by injecting the newborn mice with F1 cells depleted in different T cell subsets by using specific monoclonal antibodies (MoAbs). Tolerance, as shown by an absence of H-2b-specific CTL alloreactivity and persistence of immunoglobulin bearing the donor allotype were observed in mice injected with F1 cells previously depleted in the CD4+ and/or CD8+ T cell subsets as well as in those which received Thy-1+-depleted F1 spleen cells. In these mice, a typical autoimmune syndrome was found, including splenomegaly and lymphadenopathy, anti-ssDNA and anti-aortic myosin IgG antibodies and renal deposition of immunoglobulin. However, some quantitative changes were seen: the levels of anti-aortic myosin antibodies were lower in mice tolerized with CD4+-depleted F1 cells than in those receiving untreated F1 cells. Conversely, higher levels of these autoantibodies were observed in mice tolerized with CD8+-depleted F1 cells. These results suggest that mature donor T cells are not necessary neither for the establishment of neonatal tolerance to alloantigens nor for the activation of F1 donor B cells in the production of the autoimmune syndrome in tolerant mice, but they may contribute in the regulation of the expression of autoreactive B cell clones.  相似文献   

7.
Decreased binding by the 6C10 auto-antibody serves as a unique marker for CD4+ T cell unresponsiveness after the induction of T cell tolerance in Vbeta8.1 TCR transgenic mice. We further define the nature of the epitope recognized by the 6C10 antibody to be a subset of Thy-1 bearing incompletely sialylated N-linked glycans, and furthermore, we demonstrate that tolerant CD4+ T cells have an increased degree of cell-surface sialylation. To test the significance of the altered glycosylation state identified by the 6C10 auto-antibody in the tolerant CD4+ T cell population, surface sialic acid was cleaved enzymatically. Treatment of purified peripheral CD4+ T cells with Vibrio cholerae sialidase (VCS) leads to increased 6C10 binding, significantly enhances proliferation in the tolerant CD4+ population and corrects defects in phosphotyrosine signaling observed in the tolerant CD4+ T cell. Furthermore, in vivo administration of VCS enhances proliferation in both tolerant and naive CD4+ T cell subsets. These studies suggest that sialylation of glycoproteins on the surface of the CD4+ T cell contributes to the regulation of T cell responsiveness in the tolerant state.  相似文献   

8.
9.
目的 探讨neuropilin-1(NRPl)在外周血中CIM+ CD25+ T细胞上的表达以及意义.方法 收集肾移植术后长期存活、慢性移植物肾病和急性排斥患者以及健康成年人的外周血,分离外周血淋巴细胞,用流式细胞仪检测外周血淋巴细胞中CD4+ CD25+ T细胞上NRPl和Foxp3的阳性细胞的百分比.结果 各组CD4+ CD25+ T细胞NRPl的表达率差异有统计学意义(P<0.05);各组CD4+ CD25+ T细胞Foxp3的表达率差异亦有统计学意义(P<0.05).长期存活组CD4+ CD25+ T细胞上NRP1和Foxp3表达率分别为19.6%±3.84%、50.19 ±3.90%,显著高于对照及慢性移植物肾病和急性排斥组(P<0.05);急性排斥组的CD4+ CD25+ T细胞NRP1和Foxp3表达率最低4.64%±1.26%、17.24%±5.29%.各组CD4+ CD25+ T细胞NRP1表达变化趋势与Foxp3变化趋势相同.结论 NRP1在长期存活组的外周血淋巴细胞中CD4+ CD25+ T细胞的表达上调,在急性排斥组时表达下调,且其在各组的表达变化趋势与Foxp3的表达变化趋势是一致的,这就提示CD4+ CD25+ NRP1+ 细胞是一群调节性T细胞,即NRP1是CD4+ CD25+ Treg的一个重要的表面标志.  相似文献   

10.
We have induced specific immunological tolerance towards (C57BL × CBA-T6T6) F1 skin grafts in newborn recipients of the parental CBA strain by one injection of 20 × 106 or 40 × 106 F1 spleen cells. Approximately 80 % of the CBA recipients were highly tolerant (the grafts survived for more than 50 days). Many of the highly tolerant mice retained fully preserved grafts until natural death. The highly tolerant CBA mice had dividing (C57BL × CBA-T6T6) F1 cells in their spleen (12–13 %) and lymph nodes (12–15 %). The proportion of these cells decreased with time, but we found a few of them even at 567 days of life (0.7 and 4.2 %, respectively). We have concluded that specific immunological tolerance to tissue grafts induced at birth can last throughout the entire life-span of a mouse. The tolerance persists in spite of a very low proportion of dividing donor cells within the lymphoid tissue of tolerant hosts.  相似文献   

11.
Although the ability of CD4+CD25+ T suppressor (Ts) cells to prevent experimental autoimmune diseases has been described, nothing is known concerning their role and mechanism of action in xenobiotic-induced autoimmunity. Procainamide, mercuric chloride, and gold(I) are three xenobiotics that can induce autoimmune reactions in humans and rodents. After the induction of IgG1 antinuclear autoantibodies (ANA) in mice treated with either of the above xenobiotics, adoptive transfer of their CD4+CD25+ T cells completely prevented ANA formation in recipients treated with the same xenobiotic; transfer of CD8+ T cells was ineffective. Furthermore, xenobiotic-primed CD4+CD25+ T cells could also partially prevent ANA formation in recipients treated with a different xenobiotic. CD4+CD25- T cells from xenobiotic-treated donors failed to suppress, but induced de novo IgG1 ANA formation in untreated recipients. Our findings suggest that during xenobiotic treatment T cell reactivity may spread from xenobiotic-induced, nucleoprotein-related neoantigens to peptides of the unaltered nucleoproteins.  相似文献   

12.
Induction of peripheral T cell tolerance is mediated by bone marrow-derived dendritic cells that cross-present self-antigen to self-reactive T cells. The current model for peripheral CD8(+) T cell tolerance is that TCR engagement by self-antigen in the absence of costimulation results in abortive activation without development of effector function. Here we demonstrate in vivo that high-dose self-antigen ("signal 1") can compensate for lack of costimulation ("signal 2"), leading to full activation of and development of effector function by self-reactive T cells. In the setting of low-dose self-antigen, acquisition of effector function by self-reactive T cells is dependent on costimulation via CD40 ligation in vivo. However, gain of effector function in either setting does not prevent eventual tolerance of self-reactive CD8(+) T cells. These results suggest that the mechanisms for peripheral CD8(+) T cell tolerance are more complex than the proposed "signal 1 in the absence of signal 2" hypothesis. Further exploration of these mechanisms will have direct impact on the design of effective immunotherapy for autoimmune diseases, chronic infections and cancers.  相似文献   

13.
《Human immunology》2016,77(9):727-733
The regulation of potentially harmful immune responses by regulatory T (Treg) cells is essential for maintaining peripheral immune tolerance and homeostasis. Especially CD4+ Treg cells have been regarded as pivotal regulators of autoreactive and inflammatory responses as well as inducers of immune tolerance by using a variety of immune suppressive mechanisms.Besides the well-known classical CD4+CD25+FoxP3+ Treg cells, CD4+ T cells expressing the immune tolerizing molecule human leukocyte antigen G (HLA-G) have been recently described as another potent thymus-derived Treg (tTreg) cell subset. Albeit both tTreg subsets share common molecular characteristics, the mechanisms of their immunosuppressive function differ fundamentally. Dysfunction and numerical abnormalities of classical CD4+ tTreg cells have been implicated in the pathogenesis of several immune-mediated diseases such as multiple sclerosis (MS). Clearly, a deeper understanding of the various CD4+ tTreg subsets and also the underlying mechanisms of impaired immune tolerance in these disorders are essential for the development of potential therapeutic strategies.This review focuses on the current knowledge on defining features and functioning of HLA-G+CD4+ tTreg cells as well as their emerging role in various pathologies with special emphasis on the pathogenesis of MS. Furthermore, future research possibilities together with potential therapeutic applications are discussed.  相似文献   

14.
Although non-obese diabetic (NOD) mice spontaneously develop T cell autoimmunity, it is not clear whether this phenomenon results from a defect in tolerance to self-Ag. Furthermore, as autoimmunity has been postulated to result from T cell responses directed toward self-peptides that bind with low affinity to NOD I-A(g7) MHC class II molecules, it is important to determine whether the expression of such peptides induces tolerance. We have constructed NOD transgenic (Tg) mice expressing the Leishmania antigen receptor for C kinase (LACK) Ag in either the thymus or pancreatic beta cells. We identified LACK peptides that were the targets of T cells in LACK-immunized NOD mice while binding to I-A(g7) with low affinity. While CD4(+) T cells from NOD mice secreted IFN-gamma, IL-4, IL-5 and IL-10 in response to LACK, those from LACK-expressing Tg mice secreted reduced levels of cytokines. Experiments using peptide/MHC multimers showed that LACK-expressing Tg mice exhibited self-reactive CD4(+) T cells with impaired proliferation capabilities. Hence, even self-peptides that bind to I-A(g7) with low affinity can induce tolerance in NOD mice. This result is important in light of the commonly held hypothesis that T cells reacting to peptides that bind to MHC with low affinity escape tolerance induction and cause autoimmunity.  相似文献   

15.
GORER PA  BOYSE EA 《Immunology》1959,2(2):182-193
It has already been reported by one of us (Boyse, 1959) that intraperitoneal transplants of spleen cells from A strain donor mice that had been immunized against C57BL strain tissues were invariably lethal to hosts of the F1 (C57BL×A) constitution.

In this report an account is given of the pathological changes in F1 hybrid hosts that have received intravenous or intraperitoneal transplants of spleen cells from parental strain donors, with particular reference to lethal transplants. Here the outstanding features are splenomegaly, wasting, progressive histiocytosis in the spleen and lymph nodes, disappearance of lymphoid tissue, focal necrosis of the spleen and cellular infiltrations round the hepatic vessels. Following transplantation by the intraperitoneal route there are also severe pancreatic lesions associated with fat necrosis and ascites.

Differential (donor/host) cell counts of host spleen cell suspensions indicate that the spleen may be virtually replaced by donor cells, the majority of which are histiocytes. Erythropoietic function is progressively supplanted by donor tissue.

Similar, but reversible and less extensive changes occur with other (non-lethal) donor/F1-host combinations. These hosts do not waste and the proportion of donor cells included in the host spleens falls until their detection, by present methods, is no longer possible.

  相似文献   

16.
Co-stimulation mediated by the CD28 molecule is considered critical in the activation of CD4+ T cells. In patients with rheumatoid arthritis and infrequently in normal individuals, CD4+ T cells lacking CD28 expression are expanded and contain clonogenic populations. To analyze whether these cells are independent of co-stimulatory requirements or whether they use co-stimulatory signals distinct from the CD28 pathway, we have compared CD4+ CD28+ and CD4+ CD28?T cell clones isolated from rheumatoid arthritis patients. Accessory cells supported the induction of CD25 expression as well as of proliferative responses after anti-CD3 cross-linking and prevented the induction of anergy in CD4+ CD28? T cell clones. In contrast to CD4+CD28+ T cells, the presence of accessory cells did not enhance the secretion of interleukin (IL)-2, interferon-γ, or IL-4. The co-stimulatory signals did not involve CD28/CTLA-4–CD80/CD86 receptor-ligand interactions. The proliferative response of CD4+CD28? T cells could not be blocked by anti-CD2, anti-CD18, and anti-CD58 antibodies, suggesting that these receptor-ligand interactions cannot provide CD28? independent co-stimulation. Our data suggest that CD4+CD28? T cells require co-stimulatory signals for optimal induction of cell growth and CD25 expression as well as for the prevention of anergy. The co-stimulatory receptor-ligand interaction is independent of the CD28 pathway and may be involved in the oligoclonal expansion of the CD4+ CD28? T cell subset in rheumatoid arthritis.  相似文献   

17.
T cells undergoing lymphopenia-driven proliferation acquire effector and memory properties that can be pathogenic. Indeed, generalized lymphopenia is associated with a variety of autoimmune diseases such as type 1 diabetes. The current study was carried out to determine how CD8(+) T cells undergoing acute lymphopenic expansion respond to antigen under tolerizing conditions in vivo. Adoptive transfer of diabetes by TCR-transgenic CD8(+) T cells was enhanced following treatment of NOD. scid recipients with a high dose of soluble peptide. Furthermore, whereas TCR-transgenic CD8(+) T cells underwent clonal deletion and failed to differentiate into CTL in peptide-treated lymphoreplete recipient mice, TCR-transgenic CD8(+) T cells in a lymphopenic environment were resistant to clonal deletion, and CTL differentiation was enhanced by a high dose of soluble peptide. Moreover, peptide treatment had distinct effects on expression of the anti-apoptotic protein Bcl-X(L) in TCR-transgenic CD8(+) T cells under lymphopenic versus lymphoreplete conditions. These results demonstrate that CD8(+) T cells undergoing lymphopenia-driven expansion in NOD. scid recipients are resistant to antigen-induced tolerance, and readily differentiate into CTL upon stimulation with a high dose of soluble peptide.  相似文献   

18.
Biomarkers defining pathogenic effector T (Teff) cells slowly have been forthcoming and towards this we identified CD4+ T cells that express CD40 (CD4+CD40+) as pathogenic in the NOD type 1 diabetes (T1D) model. CD4+CD40+ T cells rapidly and efficiently transfer T1D to NOD.scid recipients. To study the origin of CD4+CD40+ T cells and disease pathogenesis, we employed a dual transgenic model expressing OVA323–339 peptide as a neo‐self antigen on islet β cells and medullary thymic epithelial cells (mTECs) and a transgenic TCR recognizing the OVA323–339 peptide. CD4+CD40+ T cells and Treg cells each recognizing the cognate neo‐antigen, rather than being deleted through central tolerance, drastically expanded in the thymus. In pancreatic lymph nodes of DO11.RIPmOVA mice, CD4+CD40+ T cells and Treg cells are expanded in number compared with DO11 mice and importantly, Treg cells remain functional throughout the disease process. When exposed to neo‐self antigen, CD4+CD40+ T cells do not express the auto‐regulatory CTLA‐4 molecule while naïve CD4+CD40+ T cells do. DO11.RIPmOVA mice develop autoimmune‐type diabetes. CD40 engagement has been shown to prevent CTLA‐4 expression and injecting anti‐CD40 in DO11.RIPmOVA mice significantly exacerbates disease. These data suggest a unique means by which CD4+CD40+ T cells thwart tolerance.  相似文献   

19.
固有CD4 CD25 调节性T细胞(naturally occurringCD4 CD25 regulatory T cells,nTreg)是在胸腺发育的功能成熟的T细胞亚群,可通过下调效应性免疫细胞的功能和增殖来抑制或控制机体免疫应答的程度,并维持机体自身免疫耐受。近年来的研究发现,nTreg对外源性抗原(如移植物同种抗原)也能产生特异性的免疫耐受。该文将就近年来nTreg在细胞标记、作用机制及其在移植耐受方面的研究进展进行综述。  相似文献   

20.
杨露  全学军 《免疫学杂志》2011,(11):1005-1007,1012
同种异体器官移植已经成为终末器官衰竭患者修复病损的组织和器官,并重建其功能必要的治疗方法。受者往往都需要长期接受免疫抑制剂来维持移植物在其体内的存活。由于免疫抑制剂的非特异性及毒副作用,长期应用该类药物会导致受者免疫力低下而出现易感染、癌症等副作用。因此,解决该问题的关键是诱导受者对供者移植抗原产生特异性地"移植免疫耐受"。大量研究结果显示,CD4+CD25+调节性T细胞能特异性地抑制免疫排斥反应,在调控免疫应答与维持外周免疫耐受中发挥着重要作用。本文对近年来CD4+CD25+调节性T细胞的作用机制及其在同种异体移植免疫耐受方面的研究进行了综述。  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