用户名: 密码: 验证码:
杂色曲霉素暴露对机体免疫调节细胞的影响及其可能机制的研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
杂色曲霉素(Sterigmatocystin, ST)是由杂色曲霉、构巢曲霉等产生的一种具有致癌性的代谢产物。杂色曲霉素产生菌及ST在世界各地粮食及饲料中的污染比较普遍,甚至在潮湿居所的地毯中也检出了ST的污染。研究表明,饲喂含有ST的饲料或灌喂ST均可诱发实验动物肺腺癌的发生。ST可以诱发体外培养人胚胃粘膜细胞的恶性转化。除致癌作用外,ST对免疫细胞的增殖、凋亡、细胞因子分泌、抗原递呈等均有一定影响。
     免疫系统是机体抵御外界因素侵害的重要屏障,许多致病因素可通过削弱甚至摧毁机体的免疫功能而致病。某些致癌因素也可通过直接和间接作用造成机体免疫细胞改变,特别是免疫监视机能的改变而发挥致癌作用。目前有关ST诱发免疫损伤的机制研究多集中在淋巴细胞细胞的增殖、凋亡、细胞因子分泌等直接的免疫反应方面,对免疫调节细胞的研究甚少。因此,在整体水平和细胞水平上探讨给予ST作用对机体免疫调节功能的影响及其机制对进一步揭示ST的致癌机理和其它生物效应具有一定学术意义。
     调节性T淋巴细胞是机体免疫系统的重要组成部分,在防止自身免疫,保持机体的自身耐受性中发挥着决定性作用,但同时也具有减弱机体对肿瘤和感染免疫反应的作用。FoxP3阳性表达是鉴定调节性T淋巴细胞的重要标志。树突状细胞(DCs)是机体免疫系统中一种重要的抗原递呈细胞(APC),可与T、B和NK细胞相互作用,并促进其激活、分化和产生效应;诱导或调节初级或次级免疫反应。浆细胞样树突状细胞(pDCs)是树突状细胞家族中的成员之一,在慢性炎症及自身免疫性疾病的发生发展中发挥着重要的作用。钙调神经磷酸酶(calcineurin, CaN)是Ca2+依赖的信号转导途径的关键蛋白,在体内参与了多种淋巴细胞的激活;皮肤T细胞趋化因子27 (Ccl27)是肿瘤相关T淋巴细胞的重要调节因子,它们的活性变化与多种皮肤疾病的发生发展有关。
     迄今为止,文献中尚未见有关ST对调节性T淋巴细胞和浆细胞样树突状细胞,以及对CaN和Ccl27表达影响的报道。为进一步探讨ST对机体免疫调节功能的影响及其可能机制,本研究在前期实验研究的基础上,在整体水平上观察了ST急性作用对BALB/c小鼠外周血、胸腺、脾脏和皮肤中FoxP3+调节性T淋巴细胞和CD123+/BDCA2+浆细胞样树突状细胞的影响,以及小鼠皮肤中CaN和Ccl27表达变化;在细胞水平上研究了ST急性作用对人外周血单个核细胞中FoxP3+调节性T淋巴细胞的影响,同时探讨了MAPK信号转导通路在ST影响FoxP3+调节性T淋巴细胞中的作用。
     本研究论文共分为三个部分:
     1杂色曲霉素对BALB/c小鼠外周血及免疫器官中免疫调节细胞的影响研究
     目的:探讨腹腔注射ST对BALB/c小鼠外周血单个核细胞及免疫器官中FoxP3+调节性T淋巴细胞和浆细胞样树突状细胞的影响。
     方法:按照3μg/kg、30μg/kg、300μg/kg、3000μg/kg经腹腔注射一次性给予雄性BALB/c小鼠ST处理,眼球放血收集小鼠外周静脉全血,采用聚蔗糖-泛影葡胺密度梯度离心法分离小鼠外周静脉血单个核细胞;处死小鼠,胸腺、脾脏组织完整取材。采用FCM检测小鼠外周血单个核细胞、胸腺和脾脏内CD4+、CD8+和FoxP3+T淋巴细胞百分率;免疫组化检测小鼠胸腺和脾脏内FoxP3+调节性T细胞及CD123+/BDCA2+浆细胞样树突状细胞数量变化情况;蛋白免疫印迹和RT-PCR方法检测FoxP3、CD123和BDCA2在蛋白和mRNA水平的影响。
     结果:
     1.1 ST对小鼠外周血单个核细胞中CD4+、CD8+和FoxP3+T淋巴细胞的影响
     FCM检测结果显示,ST处理24 h,与溶剂对照组相比,不同剂量ST对小鼠外周血单个核细胞中CD4+和CD8+T淋巴细胞百分率均没有明显影响。但随ST浓度的增加,FoxP3+T淋巴细胞百分率呈逐渐增高的趋势(r=0.862, n=5, P<0.01)。
     RT-PCR检测结果表明,给予ST作用24 h后,与溶剂对照组相比,小鼠外周血单个核细胞FoxP3 mRNA表达明显升高(P<0.05);并且与ST浓度有明显剂量依赖关系(r=0.944, n=5, P<0.01)。
     1.2 ST对小鼠胸腺和脾脏内FoxP3+T淋巴细胞的影响
     FCM检测结果显示,ST处理24 h,与溶剂对照组相比,不同剂量ST对小鼠胸腺CD4+ T淋巴细胞百分率没有明显影响,但小剂量ST(3μg/kg)处理组小鼠胸腺CD8+ T淋巴细胞百分率可见明显下降(P<0.05)。3和30μg/kg ST处理组脾脏CD4+和CD8+ T淋巴细胞百分率均有明显升高(P<0.05)。而300μg/kg和3000μg/kg ST处理组胸腺和脾脏CD4+和CD8+ T淋巴细胞百分率均无明显变化。但各不同剂量ST处理均可明显提高胸腺和脾脏FoxP3+ T淋巴细胞的百分率,随ST浓度的增加,胸腺和脾脏FoxP3+T淋巴细胞百分率呈逐渐增高的趋势(P<0.05)。
     免疫组化结果显示,ST 30μg/kg、300μg/kg和3000μg/kg剂量组胸腺内淋巴细胞FoxP3+T淋巴细胞标记指数明显高于相应溶剂对照组(P<0.05);ST各处理组脾脏内FoxP3+ T淋巴细胞标记指数也均明显高于相应溶剂对照组(P<0.05)。在0~3000μg/kg ST浓度范围内,胸腺和脾脏FoxP3+T淋巴细胞数与ST处理浓度有明显剂量依赖关系(胸腺: r=0.831, n=5, P<0.01;脾: r=0.873, n=5, P<0.01)。
     Western印迹检测结果显示,与溶剂对照组相比,在0~3000μg/kg ST浓度范围内,胸腺和脾脏淋巴细胞FoxP3表达量逐渐升高,与ST浓度有明显剂量依赖关系(胸腺: r=0.658, n=5, P<0.01;脾: r=0.621, n=5, P<0.01)。RT-PCR检测结果表明,给予ST作用24 h后,小鼠胸腺和脾脏淋巴细胞FoxP3 mRNA表达明显升高(P<0.05)。
     1.3 ST对小鼠胸腺和脾脏内浆细胞样树突状细胞的影响
     免疫组织化学染色结果显示,30、300和3000μg/kg ST组胸腺细胞CD123阳性标记指数明显低于溶剂对照组(P<0.05);而在脾脏细胞CD123阳性标记指数明显高于相应溶剂对照组(P<0.05);ST中、高剂量处理组胸腺细胞BDCA2阳性标记指数明显低于相应溶剂对照组(P<0.05);而各ST处理组脾脏细胞BDCA2阳性标记指数均明显高于相应溶剂对照组(P<0.05);小鼠胸腺和脾脏CD123+和BDCA2+细胞数的变化与ST的处理浓度存在明显剂量依赖关系。
     Western印迹检测结果显示,与溶剂对照组相比,在0~3000μg/kg ST浓度范围内,胸腺淋巴细胞CD123和BDCA2蛋白的表达量逐渐降低,并与ST浓度呈显著负相关关系(CD123: r=-0.825, n=5, P<0.01; BDCA2:r=-0.831, n=5, P<0.01);而脾脏淋巴细胞CD123和BDCA2蛋白的表达量则逐渐升高(CD123: r=0.819, n=5, P<0.01; BDCA2: r=0.756, n=5, P<0.01)。
     RT-PCR检测结果表明,给予ST作用24 h后,小鼠胸腺细胞CD123 mRNA表达较溶剂对照组明显降低(P<0.05),并与ST浓度有明显剂量依赖关系(r=-0.944, n=5, P<0.01);而脾脏细胞的CD123 mRNA则均较溶剂对照组明显升高(P<0.05),并与ST浓度有明显剂量依赖关系(r=0.906, n=5, P<0.01)。
     可见ST对胸腺和脾脏浆细胞样树突状细胞的影响具有器官特异性,不同器官影响不同。
     2 ST对小鼠皮肤组织CaN、Ccl27表达以及FoxP3+调节性T淋巴细胞影响的研究
     目的:探讨一次性腹腔注射给予ST对小鼠皮肤组织CaN和Ccl27表达,以及FoxP3+调节性T淋巴细胞浸润的影响。
     方法:实验动物模型同前一部分,全层皮肤1×1cm2取材。取部分皮肤组织4%多聚甲醛固定4 h后,常规石蜡切片制备,按S-P法进行免疫组织化学染色检测小鼠皮肤组织CaN+、Ccl27+、FoxP3+细胞数量变化;取100 mg皮肤组织加入500μL蛋白裂解液,提取皮肤组织总蛋白,Western blot方法检测CaN、Ccl27、FoxP3蛋白的表达;部分皮肤组织液氮冻存,提取组织RNA,RT-PCR方法检测CaN、Ccl27、FoxP3 mRNA的表达情况。
     结果:
     2.1皮肤组织病理学变化
     病理形态学观察可见各ST处理组动物与溶剂对照组相比,皮肤组织结构均无明显病理性改变。
     2.2 ST对小鼠皮肤组织细胞CaN和Ccl27表达的影响
     免疫组织化学染色结果显示,30、300和3000μg/kg组CaN阳性标记指数明显高于溶剂对照组(P<0.05),并且与ST的处理浓度存在剂量依赖关系(r=0.752, n=5, P<0.01);而Ccl27阳性标记指数与相应溶剂对照组相比,均没有明显差别。
     Western blot检测结果显示,与溶剂对照组相比,随着ST处理浓度的增加,CaN蛋白表达逐渐升高,其中30,300,3000μg/kg ST组蛋白表达升高尤为明显(P<0.05),并与ST的处理浓度存在正相关关系(r=0.931, n=5, P<0.01)。同时,ST 30,300和3000μg/kg处理后相对Ccl27蛋白相对表达量与溶剂对照组相比有升高趋势,但没有统计学意义。
     RT-PCR检测结果表明,给予ST作用24h后,小鼠表皮细胞CaN mRNA的表达明显高于溶剂对照组,并且与ST浓度有明显剂量依赖关系(r=0.843, n=5, P<0.01);但Ccl27 mRNA的表达没有明显变化。
     2.3 ST对小鼠皮肤内FoxP3+T淋巴细胞的影响
     免疫组化染色结果表明,ST 300μg/kg和3000μg/kg处理组FoxP3+T淋巴细胞标记指数分别为3.30±0.675和4.70±0.949,明显高于相应溶剂对照组(1.40±0.966, P<0.05),并且在ST 0~3000μg/kg浓度范围内,随着ST处理浓度的升高,FoxP3+T淋巴细胞标记指数也逐渐增高(r=0.757, n=5, P<0.01)。
     Western blot检测结果显示,腹腔给予ST作用24 h后,小鼠皮肤组织FoxP3蛋白的表达量明显升高(P<0.05),并与ST浓度有明显剂量依赖关系(r=0.781, n=5, P<0.01)。
     RT-PCR检测结果表明,给予ST作用24 h后,小鼠皮肤组织FoxP3 mRNA的表达明显升高(P<0.05),并与ST浓度有明显剂量依赖关系(r=0.749, n=5, P<0.01)。
     3杂色曲霉素对体外培养人外周血单个核细胞中FoxP3+调节性T淋巴细胞的影响及其可能的机制研究
     目的:探讨一次性给予ST处理对人外周血单个核细胞中FoxP3+调节性T淋巴细胞的影响及其可能的机制
     方法:
     采用聚蔗糖-泛影葡胺密度梯度离心法分离人外周静脉血单个核细胞(HPBMCs),培养48 h后,用10% 1640培养基调整细胞浓度为(1~2)×108个/L,接种于细胞培养瓶中。更换2%低血清1640培养基,实验组分别给予ST 100μg/L、500μg/L、1000μg/L和2000μg/L处理,溶剂对照组和对照组分别给予DMSO (0.2 mL/L)和生理盐水处理,继续细胞培养24 h。收集细胞,采用FCM方法检测HPBMCs中CD4+、CD8+和FoxP3+T淋巴细胞百分率;采用Western blot方法检测HPBMCs中FoxP3、JNK、ERK、p38蛋白表达情况和JNK、ERK和p38的磷酸化水平。采用RT-PCR方法检测HPBMCs中FoxP3、JNK、ERK和p38 mRNA的表达情况。
     取培养48 h后HPBMCs用10% 1640调整细胞浓度为(1~2)×108个/L,接种于接种于培养瓶。实验分为5组,即对照组、溶剂对照组、ST处理组、阻断剂组和阻断剂预处理+ST处理组。细胞培养24 h后,更换2%低血清1640培养基,阻断剂组分别加入MAPK特异性阻断剂SP600125 (1μM)、PD98059 (50μM)和SB203580 (0.5μM)。30 min后ST处理组和阻断剂预处理+ST处理组分别给予1000μg/L ST,溶剂对照组和对照组给予DMSO (0.1 mL/L)和生理盐水处理。HPBMCs培养24 h后离心收细胞,采用Western blot方法检测细胞内FoxP3蛋白表达和JNK、ERK、p38磷酸化水平。采用RT-PCR方法检测HPBMCs中FoxP3 mRNA的表达情况。
     结果:
     3.1 ST对体外培养人外周血单个核细胞中CD4+、CD8+和FoxP3+T淋巴细胞影响
     FCM检测结果显示,ST处理24 h后,与溶剂对照组相比,不同剂量ST对人外周血单个核细胞中CD4+和CD8+T淋巴细胞百分率均没有明显影响。但ST处理可明显提高FoxP3+T淋巴细胞的百分率,并且在0~2000μg/L浓度范围内,随着ST浓度的增加,FoxP3+T淋巴细胞百分率呈逐渐增高的趋势(r=0.920, n=3, P<0.01)。
     3.2体外培养人外周血单个核细胞FoxP3蛋白表达的变化情况
     Western检测结果表明,在0~2000μg/L ST浓度范围内,随着ST处理浓度的升高,细胞内FoxP3蛋白表达逐渐升高(r=0.868, n=3, P<0.01)。
     3.3体外培养人外周血单个核细胞FoxP3 mRNA的变化情况
     RT-PCR检测表明,给予ST作用24 h后,与溶剂对照组相比,体外培养人外周血单个核细胞FoxP3 mRNA表达明显升高(P<0.05);并且与ST浓度有明显剂量依赖关系(r=0.793, n=3, P<0.01)。
     3.4 ST对人外周血单个核细胞中MAPK信号传导通路的影响
     3.4.1 ST对细胞内JNK、ERK和p38表达的影响
     Western检测结果表明,与溶剂对照组相比,不同浓度ST作用于人外周血单个核细胞24 h,对细胞内JNK、ERK和p38蛋白的表达没有明显影响。但在0~2000μg/L ST浓度范围内,随着ST处理浓度的升高,细胞内JNK和ERK的磷酸化水平也逐渐升高(JNK: r=0.831, n=3, P<0.01;ERK: r=0.687, n=3, P<0.01),而p38的磷酸化水平则随着ST处理浓度升高而逐渐降低(r=-0.661, n=3, P<0.01)。
     RT-PCR结果显示,与溶剂对照组相比,不同浓度ST作用24 h,对人外周血单个核细胞内JNK、ERK和p38 mRNA的表达也没有明显影响。
     3.4.2 MAPK信号传导通路特异性阻断剂对ST诱导的细胞内JNK、ERK和p38表达变化的影响
     Western检测结果表明,SP600125和PD98059分别能够明显阻断ST诱导的细胞内JNK和ERK的磷酸化水平升高作用(P<0.05)。1000μg/L ST和SB203580 (0.5μM)单独处理,均可明显抑制细胞内p38的磷酸化水平(P<0.05),0.5μM SB203580预处理可使1000μg/L ST诱导的p38的磷酸化水平降低更显著(P<0.05)。
     3.5 MAPK信号传导通路对ST诱导人外周血单个核细胞中FoxP3+调节性T细胞增多的影响
     3.5.1 JNK信号传导通路的激活对ST诱导人外周血单个核细胞中FoxP3+调节性T细胞增多的影响
     Western检测结果表明, SP600125预处理+1000μg/L ST处理组细胞内FoxP3蛋白的表达则明显高于溶剂对照组(P<0.05),而与1000μg/L ST处理组没有明显差别(P>0.05)。
     RT-PCR结果显示,SP600125预处理+1000μg/L ST处理组细胞内FoxP3 mRNA的表达与1000μg/L ST处理组也没有明显差别(P>0.05),同样明显高于溶剂对照组(P<0.05)。
     结果提示,SP600125预处理对ST诱导的FoxP3在蛋白和mRNA水平表达升高的作用没有明显影响。
     3.5.2 ERK信号传导通路的激活对ST诱导人外周血单个核细胞中FoxP3+调节性T细胞增多的影响
     Western检测结果表明,PD98059预处理+1000μg/L ST处理组细胞内FoxP3蛋白的表达较1000μg/L ST处理组明显降低(P<0.05),而与溶剂对照组没有差别(P>0.05)。
     RT-PCR结果显示,PD98059预处理+1000μg/L ST处理组细胞内FoxP3 mRNA的表达也明显低于1000μg/L ST处理组(P<0.05),而与溶剂对照组相比没有明显差异(P>0.05)。
     结果提示,PD98059预处理可以阻断ST诱导的FoxP3在蛋白和mRNA水平表达升高的作用。
     3.5.3 p38信号传导通路的受抑制对ST诱导人外周血单个核细胞中FoxP3+调节性T细胞增多的影响
     Western检测结果表明,与溶剂对照组相比,SB203580处理组细胞内FoxP3蛋白的表达量明显升高(P<0.05),SB203580预处理+1000μg/L ST处理组细胞内FoxP3蛋白的表达也明显高于溶剂对照组,同时较1000μg/L ST处理组也明显升高(P<0.05)。
     RT-PCR结果显示,SB203580预处理+1000μg/L ST处理组细胞内FoxP3 mRNA的表达明显高于1000μg/L ST处理组和溶剂对照组(P<0.05)。
     结果提示,SB203580预处理可以促进ST诱导的FoxP3在蛋白和mRNA水平表达升高的作用。
     结论:
     1.一次腹腔注射ST处理24 h,可使BALB/c小鼠外周血单个核细胞中FoxP3+调节性T淋巴细胞数明显增多,并且上调FoxP3在mRNA水平的表达。
     2.一次腹腔注射ST处理24 h,可使BALB/c小鼠胸腺和脾脏内FoxP3+调节性T淋巴细胞数明显增多,并且上调FoxP3在蛋白和mRNA水平的表达。
     3.腹腔注射ST对浆细胞样树突状细胞的影响具有器官特异性。可使BALB/c小鼠胸腺内CD123+/BDCA2+浆细胞样树突状细胞数明显减少,而脾脏内CD123+/BDCA2+浆细胞样树突状细胞数明显增多。同时可以下调BALB/c小鼠胸腺内CD123和BDCA2蛋白及CD123 mRNA的表达;而上调小鼠脾脏内CD123和BDCA2蛋白及CD123 mRNA的表达。
     4.一次腹腔注射ST处理24 h,可使BALB/c小鼠皮肤CaN在蛋白和mRNA水平上的表达增高;但对Ccl27的表达没有明显影响。
     5.一次腹腔注射ST处理24 h,可使BALB/c小鼠皮肤FoxP3+调节性T淋巴细胞增多,并且上调FoxP3在蛋白和mRNA水平上的表达。
     6.一次性给予ST处理24 h,可使体外培养人外周血单个核细胞中FoxP3+调节性T淋巴细胞数明显增多,并且上调FoxP3在蛋白和mRNA水平的表达。
     7.一次性给予ST处理24 h,可以激活体外培养人外周血单个核细胞JNK和ERK信号转导通路,同时抑制p38信号转导通路。
     8.给予JNK特异性阻断剂SP600125预处理对ST诱导FoxP3的表达升高没有影响;给予ERK特异性阻断剂PD98059预处理可以阻断ST对FoxP3表达升高作用;给予p38特异性阻断剂SB203580预处理对ST诱导的FoxP3表达升高有明显的协同作用。ERK和p38信号转导通路可能参与介导ST诱导体外培养人外周血单个核细胞中FoxP3表达升高的作用。
Sterigmatocystin (ST) is the carcinogenic metabolite producted by Aspergillus versicolor, Aspergillus nidulans etc. Contaminations of ST and it producing fungi are quite commonly seen in grains and animal diets all over the world. ST could be detected even in carpet dust from damp dwellings. Our previous study showed that both foodstuffs containing ST and oral administration of ST could induce adenocarcinoma of lung in NIH mice. ST could induce malignant transformation of human fetal gastric and lung cells in vitro. And studies both in vivo and in vitro showed that apart from its carcinogenic effects, ST could affect the proliferation, apoptosis and secretion of cytokines as well as the antigen presentation mechanisms of human and experimental animal immunocytes.
     As we all know that immune system plays a key role in the protection of human body against injuries of different environmental factors. Wide variety of etiological factors, especially biological factors, cause human diseases by weakening or destroying immune fuctions. Some extrinic factors could also affect carcinogenesis by its direct or indirect injury effects on immune mechnisms, especially by their injury effects on immune survillence. The impairment of immune system provides important foundation for the carcinogenesis processes. Currently, studies on the mechanism of ST induced damage to the immune system have centered on the direct immune injuries, such as those on proliferation, apoptosis and cytokine secretion of lymphocytes. Few studies involved in the immune regulatory cells. Therefore, it is of very important significance to explore the effect of ST on the immune regulatory cells so as to have a better understanding of the possible carcinogenic effects and other putative the biological effects of ST on human beings.
     Regulatory T cells (Tregs) are important subpopulation of T lymphocyte in immune system. Tregs are thought to be essential for maintaining tolerance to self-antigens recognised by autoreactive T cells that escape deletion in the thymus. An absence of functional Tregs has been associated with several autoimmune diseases. Tregs play important roles suppressing effective immune surveillance of carcinogen-induced tumours in intact animals. Naturally occurring Tregs specifcally express the transcription factor Foxp3 (forkhead box P3), a member of the fork-head/winged-helix family of transcription factors. Foxp3 is a master regulator of Treg development and function. Dendritic cell (DC) is an important antigen presenting cell (APC), which may interact with T, B and NK cells to promote their activation, differentiation and producing effects to induce or regulate primary or secondary immune response. Plasmacytoid dendritic cell (pDCs) is a member of DC family, and is of key importance in the development of chronic inflammation and autoimmune disease.
     As a downstream target of the T cell receptor and key protein in the Ca2+-dependent signal transduction pathway,Calcineurin (CaN) participates in the activation process of various lymphocytes. Cutaneous T cell-attracting chemokine 27 (Ccl27) is an important regulatory factor for the tumor-related T lymphocytes. The changes in the activity alteration of CaN and Ccl27 are thought to be closely associated with the development of various kind of skin diseases. Up to now, no studies on the possible effects of ST on Tregs, pDCs and CaN, Ccl27 expression have been seen.
     The aim of the present study is to further evaluate the putative effects of ST on the immunoregulary cells and to explore its possible mechanisms. Based on previous studies, we observed the effects of ST on the FoxP3+ regulatory T cells and plasmacytoid dendritic cells of peripheral blood, thymus, spleen as well as the skin tissue, and the expression of CaN and Ccl27 in the skin was also evaluated in mice.The effects of ST on the FoxP3+ regulatory T cells in human peripheral blood mononuclear cells and on MAPK signal transduction pathway were studied.
     The study includes three parts:
     1 Effects of sterigmatocystin on the immunoregulary cells in peripheral blood and immune organs of BALB/c mice
     Objective: To study the effects of the intraperitoneal injecting ST on the FoxP3+ regulatory T cells and plasmacytoid dendritic cells in peripheral blood mononuclear cell and immune organs in BALB/c mice.
     Methods: Male BALB/c mice were intraperitoneally injected with 3μg/kg, 30μg/kg, 300μg/kg and 3000μg/kg ST for 24 hours and the whole blood in peripheral vein was collected by eyeball enucleation. The murine peripheral blood mononuclear cells (MPBMCs) were isolated with Ficoll-Meglucamine Diatrizoate density gradient centrifugation. Thymus and spleen were taken after all the mice were sacrificed. The percentage of CD4+/CD8+ T cells and FoxP3+ T cells was determined using Epics-XLⅡflow cytometry with immunofluorescence labeling. The changes of FoxP3+ regulatory T cells and CD123+/BDCA2+ plasmacytoid dendritic cells number in thymus and spleen were studied with immunohistochemical staining. The expressions of FoxP3, CD123 and BDCA2 at protein and mRNA levels were measured by Western blot and RT-PCR.
     Results:
     1.1 Effect of ST on CD4+, CD8+ and FoxP3+ regulatory T lymphocyte in MPBMCs
     The results of FCM showed that no significant difference was found in the percentage of CD4+ and CD8+ T cells in MPBMCs between all the ST groups with different dosage of ST in comparison with solvent control group. However, the percentage of FoxP3+ T cells was significant increased as the concentration of ST increases (r=0.862, n=5, P<0.01).
     RT-PCR analysis showed that FoxP3 was increased at mRNA level in MPBMCs(P<0.05), and there was a significant dose-effect correlation between ST dosage and the expression of FoxP3 at mRNA level.
     1.2 Effect of ST on FoxP3+ regulatory T lymphocyte in thymus and spleen in BALB/c mice
     The results of FCM showed that 24 hours after ST treatment, no significant differences were found in in the percentage of CD4+ T cells among control, solvent control and all ST treatment groups. However, in thymus, the percentage of CD8+ T cells was decreased in small dosage ST (3μg/kg) treatment group (P<0.05). While in spleen, the increases in percentage of CD4+ and CD8+ T cells was noted in small dosage ST (3 and 30μg/kg) treatment groups (P<0.05) but not in high dosage ST (300 and 3000μg/kg) treatment groups. Attentively, FCM results showed that the percentages of FoxP3+ T cells in all ST treatment groups were significantly increased both in thymus and spleen. A dose-effect correlation was found between ST dosage and the percentages of FoxP3+ T cells (P<0.05).
     In ST 30μg/kg, 300μg/kg and 3000μg/kg treatment groups, the immunohistochemical labelling index of FoxP3+ cells in thymus was obviously higher than that in solvent control group (P<0.05). And in spleen, FoxP3+ T cells in all ST treatment group were higher as compared with solvent controls (P<0.05). Moreover, within the dosage range from 0 to 3000μg/kg, there was a significant dose-effect correlation between ST dosage and the percentage of FoxP3+ T cells (thymus: r=0.831, P<0.01; spleen: r=0.873, P<0.01).
     The results of Western blotting showed that as compared with solvent control, the expressions of FoxP3 at protein level in all ST treatment groups were significantly increased. Within the ST dosage range from 3μg/kg to 3000μg/kg, positive correlation was found between the ST dosage and FoxP3 protein expression (thymus: r=0.658, P<0.01; spleen: r=0.621, P<0.01). The results of RT-PCR showed that FoxP3 was increased at mRNA level in murine thymus and spleen after different dosages of ST exposure for 24 hrs (P<0.05).
     1.3 Effect of ST on plasmacytoid dendritic cells in thymus and spleen of BALB/c mice
     In comparison with solvent controls, the immunohistochemical labelling index of CD123+ cells in ST 30μg/kg, 300μg/kg, 3000μg/kg treatment groups were obviously lower in thymus and higher in spleen(P<0.05). And the immunohistochemical labelling index of BDCA2+ cells in thymus in 30μg/kg, 300μg/kg, 3000μg/kg ST treatment groups were obviously reduced, but in spleen, that in all ST groups were increased compared with solvent controls (P<0.05). Moreover, within the dosage range from 0 to 3000μg/kg, there was a significant dose-effect correlation between ST dosage and the percentage of CD123~+ and BDCA2+ cells.
     Western blotting results showed that, compared with solvent control group, a significant negative correlation between ST dosage and the expression of CD123 and BDCA2 at protein level in thymus could be found (CD123: r=-0.825, n=5, P<0.01; BDCA2: r=-0.831, n=5, P<0.01) within the dosage range from 3μg/L to 3000μg/kg. But in spleen, a significant positive correlation between ST dosage and the expression of CD123 and BDCA2 at protein level was seen (CD123: r=0.819, n=5, P<0.01; BDCA2: r=0.756, n=5, P<0.01).
     The results of RT-PCR suggested that CD123 was decreased at mRNA level in murine thymus and was increased in spleen in all ST treatment groups (P<0.01). Results from dosage-dependent studies indicated that there was a significant dose-effect correlation between ST dosage and the expression of CD123 mRMA both in thymus and spleen (thymus: r=-0.944, n=5, P<0.01; spleen: r=0.906, n=5, P<0.01).
     Thus,the results in this part suggestted that the effects of ST on pDCs be organ sprcific. The effects are totally different in central and peripheral lymphpoid oagans.
     2 Effects of sterigmatocystin on the expression of CaN and Ccl27 and FoxP3~+ regulatory T lymphocyte in the skin of BALB/c mice
     Objective: To evaluate the effects of single intraperitoneal administration of ST on expression of CaN and Ccl27 and the infiltration of FoxP3~+ regulatory T lymphocyte in the skin of BALB/c mice. Methods: The treatment of experimental animals was the same as in the first part. The full-thick skin specimen were obtained from the mice. Representative tissues specimens were fixed in 4% phosphate-buffered paraformaldehyde, embedded in paraffin and sectioned. The changes in the number of CaN+, Ccl27+ and FoxP3~+ cells in skin tissues was studied with immunohistochemical staining method. Fresh skin tissues (100mg) for Western Blot was first homogenized in 500μL lysis buffer and then the total protein was extracted from the skin tissues and stored at -80℃. The expression of CaN, Ccl27 and FoxP3 protein was determined by Western blot. The expression of CaN, Ccl27 and FoxP3 at mRNA level was detected by semi-quantitative RT-PCR.
     Results:
     2.1 Effects of ST on the histopathological changes in the skin in BALB/c mice
     No significant pathologial changes could be found in the skin in all the ST groups as compared with the controls.
     2.2 Effects of ST on the expression of CaN and Ccl27 in the skin tissues in BALB/c mice
     The immunohistochemical labelling index of CaN+ cells of the skin in ST 30μg/kg, 300μg/kg and 3000μg/kg treatment groups were obviously higher than that in solvent control group (P<0.05). And within the dosage range from 0 to 3000μg/kg, the number of CaN+ cells was correlated with ST dosage (r=0.752, n=5, P<0.01). But the immunohistochemical labelling index of Ccl27+ cells in ST treatment groups were not different from that in solvent control group.
     The results of Western blot showed that as compared with solvent control, the expression of CaN at protein level was increased. A significant positive correlation could be found between ST dosage and the expression of CaN at protein level in the skin (r=0.931, n=5, P<0.01) within the dosage range from 3μg/kg to 3000μg/kg. But there was no difference of the expression of Ccl27 protein in ST treatment groups as compared with solvent control group.
     Similar to the results with immunohistochemical and Western blot, the results of RT-PCR showed that CaN was increased at mRNA level in murine skin after ST treatment for 24 h (P<0.01). And the expression of CaN mRMA in the skin had a significant dose-effect correlation with ST dosage (r=0.843, n=5, P<0.01). But no changes in Ccl27 mRNA expression could be found. 2.3 Effects of ST on FoxP3+ regulatory T lymphocyte in the skin tissues of BALB/c mice
     Immunohistochemical staining results showed that the positive labelling index of FoxP3+ cells in skin tissue of 300μg/kg and 3000μg/kg ST treatment groups were 3.30±0.675 and 4.70±0.949 respectively,which was obviously higher than that in solvent control group (1.40±0.966, P<0.05, P<0.05). And within the dosage range from 0 to 3000μg/kg, there was a significant dose-effect correlation between ST dosage and the percentage of FoxP3+ T cells (r=0.757, n=5, P<0.01).
     The results of Western blotting showed that, in comparison with solvent control, a significant dose-effect correlation could be found between ST dosage and the expression of FoxP3 at protein level in the skin (r=0.781, n=5, P<0.01) within the dosage range from 3μg/kg to 3000μg/kg.
     RT-PCR results showed that FoxP3 was increased at mRNA level in murine skin after exposure to different dosages of ST for 24 h (P<0.05). Results from concentration-dependent studies indicated that there was a significant positive correlation between ST dosage and the expression of FoxP3 mRNA (r=0.749, n=5, P<0.01).
     3 Effects of ST on FoxP3+ regulatory T lymphocyte in human peripheral blood mononuclear cell in vitro and the possible mechanisms
     Objective: To evaluate the effects of single treatment of ST on FoxP3+ regulatory T lymphocyte in human peripheral blood mononuclear cells in vitro and to explore the potential mechanisms.
     Methods:
     The human peripheral blood mononuclear cells (HPBMCs) were isolated with Ficoll-Meglucamine Diatrizoate density gradient centrifugation. After culture for 48 h, HPBMCs were harvestd, centrifuged and resuspended in 1640 medium supplemented with 10% FCS at the concentration of (1~2)×108 cells/L in culture flasks (8 ml). The medium of HPBMCs was replaced by new 1640 medium supplemented with 2% FCS 24 h later. Then the cells in ST groups were respectively treated with ST in different concentration of 100μg/L, 500μg/L, 1000μg/L and 2000μg/L, while their counterparts in solvent control and control group were incubated with DMSO and saline respectively. The cells were cultured for 24 h after treatment and harvested for detection. The percentage of CD4+/CD8+ T cells and FoxP3~+ T cells was determined using Epics-XLⅡflow cytometry with immunofluorescence labeling. The protein expression of FoxP3, JNK, ERK, p38 and the phosphorylation of JNK, ERK, p38 in HPBMCs treated with ST were determined by Western blot. The expression of FoxP3, JNK, ERK and p38 mRNA of HPBMCs cells was detected by RT-PCR.
     HPBMCs were harvestd 48 h after culture, centrifuged and resuspended in 1640 medium supplemented with 10% FCS at the concentration of (1~2)×108 cells/L in culture flasks (8 ml). HPBMCs were randomly divided into 5 groups: control, solvent control, ST 1000μg/L, blocking agent and blocking agent +ST 1000μg/L. The medium of HPBMCs cells was replaced by new 1640 medium supplemented with 2% FCS 24 h later. The cells of blocking agent groups were pretreated for 30 min with 1μM SP600125 (inhibitor of JNK), 50μM PD98059 (inhibitor of ERK), 0.5μM SB203580 (inhibitor of p38) and 1μM LY-294002 (inhibitor of PI3K) respectively. Then the cells in blocking agent +ST 1000μg/L group were treated with ST 1000μg/L, while these in solvent control and control groups were incubated with DMSO and saline respectively. Cells were harvested 24 h after ST treatment. The phosphorylation of JNK, ERK, p38 and the expression of FoxP3 protein in HPBMCs treated with ST were determined with Western blot. And the expression of FoxP3 mRNA of HPBMCs was detected by RT-PCR.
     Results:
     3.1 Effect of ST on CD4+, CD8+ and FoxP3~+ regulatory T lymphocyte in HPBMCs in vitro
     The results of FCM showed that no differences were observed in the percentage of CD4+ and CD8+ T cells in MPBMCs between all the ST groups after exposure to different dosages of ST for 24 h and solvent control group. However, the percentage of FoxP3~+ T cells was significant increased in all ST treated groups and within ST dosage range from 0 to 2000μg/L, the percentage of FoxP3~+ T cells increased as the ST dosage increased (r=0.920, n=3, P<0.01).
     3.2 Effect of ST on the expression of FoxP3 protein in HPBMCs in vitro
     The results of Western blot showed that as compared with solvent control, FoxP3 protein expressions were in all ST treatment groups. A significant positive correlation could be found between ST dosage and the expression of FoxP3 at protein level in the skin (r=0.868, n=3, P<0.01) within the dosage range from 0 to 2000μg/L.
     3.3 Effects of ST on the expression of FoxP3 mRNA in HPBMCs in vitro
     The results of RT-PCR showed that the expression of FoxP3 mRNA in HPBMCs in all ST treatmen groups was increased compared with solvent control group (P<0.05), and there was a significant positive correlation between ST dosage and the expression of FoxP3 mRNA (r=0.793, n=3, P<0.01).
     3.4 Effects of ST on MAPK signal transduction pathway in HPBMCs
     3.4.1 Effects of ST on the expression and phosphorylation of JNK, ERK and p38
     The results of Western blot indicated that the expression of JNK, ERK and p38 protein in HPBMCs was not influenced by the ST treatment. No difference in the expression level of JNK, ERK and p38 was found between all the ST treatment groups and solvent control group (DMSO) (P>0.05). The phosphorylated JNK and ERK protein in ST treatment groups was significantly increased as compared with corresponding solvent control groups (p-JNK: r=0.831, n=3, P<0.01; p-ERK: r=0.687, n=3, P<0.01). A significant negative correlation between ST dosages and the phosphorylation of p38 was found (r=-0.661, n=3, P<0.01).
     The results of RT-PCR confirmed that no difference in the expression of JNK, ERK and p38 mRNA in HPBMCs was found between all the ST treatment groups and corresponding solvent control group (P>0.05).
     3.4.2 Effects of inhibitors on the expression of JNK, ERK and p38 induced by ST
     The results of Western blot showed that SP600125 and PD98059 pretreatment could block the increase in the phosphorylation of JNK and ERK induced by ST treatment n HPBMCs in vitro (P<0.05). The phosphorylation of p38 in HPBMCs could be inhibited by either ST 1000μg/L or SB203580 (0.5μM). And SB203580 (0.5μM) pretreatment could further decrease the the phosphorylation decrease of p38 by ST (P<0.05).
     3.5 Effects of MAPK signal transduction pathway on the increase of FoxP3+ regulatory T lymphocyte in HPBMCs induced by ST in vitro
     3.5.1 Effects of JNK signal transduction pathway activation on the increase of FoxP3+ regulatory T lymphocyte in HPBMCs induced by ST
     Western blot results showed that though the expression level of FoxP3 protein in SP600125+1000μg/L ST treated cells was significantly higher than that in solvant control group (P<0.05), but it was not different from that in 1000μg/L ST treatment cells (P>0.05).
     The findings of RT-PCR confirmed the same result for the expression of FoxP3 mRNA as for the expression of FoxP3 protein.
     Thus, the results in this study suggested that SP600125 had no effect on the increase of expression level of FoxP3 at protein and mRNA level induced by ST.
     3.5.2 Effects of ERK signal transduction pathway activation on the increase of FoxP3+ regulatory T lymphocyte in HPBMCs induced by ST
     The results of Western blot showed that the expression level of FoxP3 protein in PD98059+1000μg/L ST treatment group was significantly lower than that in 1000μg/L ST group (P<0.05). While there was no difference in the expression level of FoxP3 between PD98059+1000μg/L ST treatment group and solvant control group (P>0.05).
     RT-PCR results revealed that the expression level of FoxP3 mRNA in PD98059+ST treatment group also was significantly lower than that in ST group (P<0.05), and was no different from that in solvent control group.
     The results in this study indicated that the increase in the expression level of FoxP3 at protein and mRNA level induced by ST could be blocked by PD98059.
     3.5.3 Effects of p38 signal transduction pathway inhibition on the increase of FoxP3+ regulatory T lymphocyte in HPBMCs induced by ST
     The results of Western blot showed that SB203580 treatment could increase the expression level of FoxP3 protein compared with the solvant control group, and the combination treatment of SB203580 and ST could induce a significant increase of the FoxP3 protein expression as compared with both solvant control group and ST groups (P<0.05).
     The results of RT-PCR confirmed that the expression of FoxP3 mRNA in SB203580+ST treatment group was significantly increased as compared with that in solvent control group and ST group (P<0.05).
     The results in this study suggested that SB203580 could promote the increase in the expression of FoxP3 at protein and mRNA level induced by ST.
     Conclusions:
     1. Intraperitoneal administration of ST could significantly increase the percentage of FoxP3+ regulatory T cells and the expression of FoxP3+ at mRNA level in peripheral blood mononuclear cells in BALB/c mice.
     2. Intraperitoneal administration of ST could significantly increase the percentage of FoxP3+ regulatory T cells and the expression of FoxP3+ at protein and mRNA level in a dose-dependent pattern both in thymus and spleen of BALB/c mice.
     3. The effects of ST on pDCs are organ sprcific. The effect is totally different in central and peripheral lymphpoid oagan in BALB/c mice. Intraperitoneal administration of ST could significantly decrease the number of CD123+/BDCA2+ plasmacytoid dendritic cells in thymus, but increase that in spleen. The expression of CD123 and BDCA2 protein and CD123 mRNA was decreased in thymus while increased in spleen.
     4. Intraperitoneal administration of ST could significantly increase the expression of CaN at protein and mRNA level in murine skin, but had no effect on that of Ccl27.
     5. Intraperitoneal administration of ST could significantly increase the infiltration of FoxP3+ regulatory T cells and the expression of FoxP3+ at protein and mRNA level in a dose-dependent pattern in murine skin.
     6. ST could significantly increase the percentage of FoxP3+ regulatory T cells and the expression of FoxP3+ at protein and mRNA level in HPBMCs.
     7. ST could activate JNK and ERK signal trusduction pathway, but inhibit p38 signal trusduction pathway in HPBMCs in vitro.
     8. JNK signal transduction pathway inhibitor SP600125 pretreatment had no effect on the increase of FoxP3 expression induced by ST. But ERK signal transduction pathway inhibitor PD98059 pretreatment could block the increase of FoxP3 expression induced by ST. And p38 signal transduction pathway inhibitor SB203580 had synergistic effect with ST on the increase of FoxP3 expression. ERK and p38 signal transduction pathway may be involved in FoxP3+ expression in HPBMCs in vitro.
引文
1 Tian H, Liu X. Survey and analysis on sterigmatocystin contaminated in grains in China. Wei Sheng Yan Jiu. 2004, 33(5): 606-608
    2 Mahmoud AL. Mycotoxin-producing potential of fungi associated with qat (Catha edulis) leaves in Yemen. Folia Microbiol. 2000, 45(5): 452-456
    3 Engelhart S, Loock A, Skutlarek D, et al. Occurrence of toxigenic Aspergillus versicolor isolates and sterigmatocystin in carpet dust from damp indoor environments. Appl Environ Microbiol. 2002, 68(8): 3886-3890
    4黄向华,张祥宏,严霞,等.杂色曲霉素对体外培养人外周血白细胞介素—Ⅱ分泌影响的研究[J].卫生研究, 2002, 31(2): 112-114
    5邢凌霄,张祥宏,尹桂然,等.杂色曲霉素对体外小鼠腹腔巨噬细胞白细胞介素12表达与分泌的影响.中国药理学与毒理学杂志, 2005, 19(3): 205-208
    6邢凌霄,张祥宏,李月红,等.杂色曲霉素对人外周血单个核细胞TAP1及LMP2基因表达的影响.中国肿瘤临床, 2004, 31(4): 189-192
    7 Piccirillo CA, Shevach EM. Naturally-occurring CD4+CD25+ immunoregulatory T cells: central players in the arena of peripheral tolerance. Semin Immunol, 2004, 16 (2): 81-88
    8 Zenclussen AC, Gerlof K, Zenclussen ML, et al. Abnormal T-cell reactivity against paternal antigens in spontaneous abortion: adoptive transfer of pregnancy-induced CD4+CD25+ T regulatory cells prevents fetal rejection in a murine abortion model. Am J Pathol, 2005, 166(3): 811-822
    9 Belkaid Y, Rouse BT. Natural regulatory T cells in infectious disease. Nat Immunol, 2005, 6 (4): 353-360
    10 Chen TC, Cobbold SP, Fairchild PJ, et al. Generation of anergic and regulatory T cells following prolonged exposure to a harmless antigen. J Immunol, 2004, 172 (10): 5900-5907
    11 Wei WZ, Morris GP, Kong YC. Anti-tumor immunity and autoimmunity: a balancing act of regulatory T cells. Cancer Immunol Immunother, 2004, 53 (2): 73-78
    12 Mellman I, Steinman RM. Dendritic cells: Specialized and regulated antigen processing machines. Cell, 2001, 106(3): 255-258
    13 Fonteneau JF, Gilliet M, Larsson M, et al. Activation of influenza virus-specific CD4+ and CD8+ T cells: a new role for plasmacytoid dendritic cells in adaptive immunity. Blood. 2003, 101(9): 3520-3526
    14 Steinman R, Hemmi H. Dendritic cells: Translating innate to adaptive immunity. Curr Top Microbiol Immunol, 2006, 311: 17-58
    15 Pashenkov, Huang YM, Kostulas V, et al. Two subsets of dendritic cells are present in human cerebrospinal fluid. 2001, 124(Pt 3): 480-492
    16 Wollenberg, Wagner AM, Gunther S, et al. Plasmacytoid dendritic cells: a new cutaneous dendritic cell subset with distinct role in inflammatory skin diseases. J. Invest. Dermatol. 2002, 119(5): 1096-1102
    17 Farkas L, Beiske K, Lund-Johansen F, et al. Plasmacytoid dendritic cells (natural interferon- alpha/beta-producing cells) accumulate in cutaneous lupus erythematosus lesions. Am. J. Pathol. 2001, 159(1): 237-243
    18左连富.流式细胞术样品制备技术(第一版).北京:华夏出版社,1991,14-19
    19 Shevach EM. Regulatory T cells in autoimmmunity. Annu Rev Immunol, 2000, 18: 423-449
    20 Thompson C, Powrie F. Regulatory T cells. Curr Opin Pharmacol, 2004, 4 (4): 408-414
    21 Curiel TJ, Coukos G, Zou L, et al. Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat Med, 2004, 10 (9): 942-949
    22 Unitt E, Rushbrook S, Marshall A, et al. Compromised lymphocytes infiltrate hepatocellular carcinoma: the role of T-regulatory cells. Hepatology, 2005, 41 (4): 722-730
    23 Sasada T, Kimura M, Yoshida Y, et al. CD4 + CD25 + regulatory T cells in patients with gastrointestinal malignancies. Possible involvement of regulatory T cells in disease progression. Cancer, 2003, 98 (5): 1089-1099
    24 Hori S, Nomura T and Sakaguchi S. Control of regulatory T cell development by the transcription factor Foxp3. Science, 2003, 299 (5609): 1057-1061
    25 Cosmi L, Liotta F, Lazzeri E, et al. Human CD8+ CD25+ thymocytes share phenotypic and functional features with CD4+ CD25+ regulatory thymocytes [J]. Blood. 2003, 102(12): 4107-4114
    26 Kiniwa Y, Miyahara Y, Wang HY, et al. CD8+ Foxp3+ regulatory T cells mediate immunosuppression in prostate cancer. Clin Cancer Res. 2007, 13(23): 6947-6958.
    27 Williams LM and Rudensky A. Maintenance of the FoxP3-dependent developmental program in mature regulatory T cells requires continued expression of FoxP3. Nat Immunol, 2007, 8 (3): 277-284
    28 Heather Kopf, Gonzalo M de la Rosa, Zack Howard OM, et al. Rapamycin inhibits differentiation of Th17 cells and promotes generation of FoxP3+ T regulatory cells. International Immunopharmacology. 2007, 7(13): 1819-1824
    29 Wei T, Yang Y, Fan Z, et al. Prevention of graft-versus-host disease by anovel immunosuppressant, (5R)-5-hydroxytriptolide (LLDT-8), through expansion of regulatory T cells. International Immunopharmacology, 2005, 5(13-14): 1904-1913
    30 Yvonne Vercoulen, Isme De Kleer, Sylvia Kamphuis, et al. Human Heat Shock Protein 60 Induces Differentiation of Cd4+Cd3//Q T-Cells Into Foxp3+Cd4+Cd25//0 Regulatory T-Cells. Clin Immunol, 2006, 119 (1): S19-S20
    31 Suzana Radulovic MD, Mikila R. Jacobson PhD, Stephen R. Durham MD, et al. Grass pollen immunotherapy induces Foxp3-expressing CD4+CD25+ cells in the nasal mucosa. J. Allergy Clin Immunol, 2008, 121(6): 1467-1472
    32 Yoolhee Yang, Jin Ho Paik, Daeho Cho, et al. Resveratrol induces the suppression of tumor-derived CD4+CD25+ regulatory T cells. International Immunopharma. 2008, 8(4): 542-547
    33 Itoh M, Takahashi T, Sakaquchi N, et al. Thymus and autoimmunity: production of CD25+CD4+ naturally anergic and suppressive T cells as a key function of the thymus in maintaining immunologic self-tolerance. J Immunol. 1999, 162(9): 5317-5326
    34 Hinkmann C, Knerr I, Hahn EG, et al. Reduced Frequency of Peripheral Plasmacytoid Dendritic Cells in Type 1 Diabetes. Horm Metab Res. 2008, 40(11): 767-771
    35 Bailey-Bucktrout SL, Caulkins SC, Goings G, et al. Cutting edge: central nervous system plasmacytoid dendritic cells regulate the severity of relapsing experimental autoimmune encephalomyelitis. J Immunol. 2008, 180(10): 6457-6461
    36 Lande R, Gafa V, Serafini B, et al. Plasmacytoid dendritic cells in multiple sclerosis: intracerebral recruitment and impaired maturation in response to interferon-beta. J Neuropathol Exp Neurol. 2008, 67(5): 388-401
    37 Manches O, Munn D, Fallahi A, et al. HIV-activated human plasmacytoid DCs induce Tregs through an indoleamine 2,3-dioxygenase-dependent mechanism. J Clin Invest. 2008, 118(10): 3431-3439
    38 Chen W, Zhang Z, Shi M, et al. Activated plasmacytoid dendritic cells act synergistically with hepatitis B core antigen-pulsed monocyte-derived dendritic cells in the induction of hepatitis B virus-specific CD8 T-cell response. Clin Immunol. 2008, 129(2): 295-303
    39 Cao W, Manicassamy S, Tang H, et al. Toll-like receptor-mediated induction of type I interferon in plasmacytoid dendritic cells requires the rapamycin-sensitive PI(3)K-mTOR-p70S6K pathway. Nat Immunol. 2008, 9(10): 1157-1164
    40 Dzionek A, Fuchs A, Schmidt P, et al. BDCA-2, BDCA-3, and BDCA-4: Three markers for distinct subsets of dendritic cells in human peripheral blood. J Immunol. 2000, 165(11): 6037-6046
    41 LoréK, Betts MR, Brenchley JM, et al. Toll-like receptor ligands modulate dendritic cells to augment cytomegalovirus- and HIV-1-specific T cell responses. J Immunol. 2003, 171(8): 4320-4328
    42 Lande R, Giacomini E, Serafini B, et al. Characterization and recruitment of plasmacytoid dendritic cells in synovial fluid and tissue of patients with chronic inflammatory arthritis. J Immunol. 2004, 173(4): 2815-2824
    43 Gilliet M, Conrad C, Geiges M, et al. Psoriasis triggered by toll-like receptor 7 agonist imiquimod in the presence of dermal plasmacytoid dendritic cell precursors. Arch Dermatol. 2004, 140(12): 1490-1495
    44 Santoro A, Majorana A, Roversi L, et al. Recruitment of dendritic cells in oral lichen planus. J Pathol. 2005, 205(4): 426-434
    45 Muralimohan G, Vella AT. A role for IFNgamma in differential superantigen stimulation of conventional versus plasmacytoid DCs. Cell Immunol. 2006, 242(1): 9-22
    46 Yoneyama H, Matsuno K, Toda E, et al. Plasmacytoid DCs help lymph node DCs to induce anti-HSV CTLs. J Exp Med. 2005, 202(3): 425-435
    1 Tian H, Liu X. Survey and analysis on sterigmatocystin contaminated in grains in China. Wei Sheng Yan Jiu. 2004, 33(5): 606-608
    2谢同欣,张祥宏,严霞,等.杂色曲霉素对人胃粘膜的致癌作用.肿瘤防治研究, 1996, 23(6): 341-343
    3邢凌霄,张祥宏,申海涛,等.杂色曲霉素对新生乳鼠致癌作用的实验研究.中华病理学杂志, 2007, 36(4): 265-266
    4邢凌霄,张祥宏,尹桂然,等.杂色曲霉素对体外小鼠腹腔巨噬细胞白细胞介素12表达与分泌的影响.中国药理学与毒理学杂志, 2005, 19(3): 205-208
    5 Kiani A, Rao A, Aramburu J. Manipulating immune responses with immunosuppressive agents that target NFAT [J]. Immunity, 2000, 12(4): 359-372
    6 Crabtree G R, Olson E N. NFAT signaling: choreographing the social lives of cells [J]. Cell, 2002, 109(Suppl): S67-79
    7 Beatriz Tapia, Antonia Padial, Elena Sánchez-Sabaté, et al. Involvement of CCL27-CCR10 interactions in drug-induced cutaneous reactions. J Allergy and Clinical Immunol, 2004, 114(2): 335-340
    8 Oriana Simonetti, Gaia Goteri, Guendalina Lucarini, et al. Potential role of CCL27 and CCR10 expression in melanoma progression and immune escape. European J of Cancer, 2006, 42(8): 1181-1187
    9常连庆,谢晓华,刘莉,等.钙调神经磷酸酶在醛固酮诱导心肌肥大中的作用.高血压杂志, 2005, 13(7): 432-436
    10马向涛,余力伟,付静.钙调神经磷酸酶A与B在乳腺癌组织中的表达.肿瘤, 2008, 28(2): 146-149
    11 Macian F. NFAT proteins:keyregulatorsofT-celldevelopment and function[J]. Nat Rev Immunol, 2005, 5(6): 472-484
    12 Huang V, Lonsdorf AS, Fang L, et al. Cutting edge: rapid accumulation of epidermal CCL27 in skin-draining lymph nodes following topical application of a contact sensitizer recruits CCR10-expressing T cells. J Immunol. 2008, 180(10): 6462-6466
    13 Fernandez TD, Mayorga C, Torres MJ, et al. Cytokine and chemokine expression in the skin from patients with maculopapular exanthema to drugs. Allergy. 2008, 63(6): 712-719
    14 Andor Pivarcsi, Anja Mu¨ller, Andreas Hippe, et al. Tumor immune escape by the loss of homeostatic chemokine expression. Proc Natl Acad Sci U S A., 2007, 104(48): 19055-19060
    15 Horlock C, Stott B, Dyson PJ, et al. The effects of trastuzumab on the CD4+CD25+FoxP3+ and CD4+IL17A+ T-cell axis in patients with breast cancer. Br J Cancer. 2009, 100(7): 1061-1067
    16 Qin FX. Dynamic behavior and function of Foxp3+ regulatory T cells in tumor bearing host. Cell Mol Immunol. 2009, 6(1): 3-13
    17 Gobert M, Treilleux I, Bendriss-Vermare N, et al. Regulatory T cells recruited through CCL22/CCR4 are selectively activated in lymphoid infiltrates surrounding primary breast tumors and lead to an adverse clinical outcome. Cancer Res. 2009, 69(5): 2000-2009
    18 Franceschini D, Paroli M, Francavilla V, et al. PD-L1 negatively regulates CD4+CD25+Foxp3+ Tregs by limiting STAT-5 phosphorylation in patients chronically infected with HCV. J Clin Invest. 2009, 119(3): 551-564
    19 Wallen H, Thompson JA, Reilly JZ, et al. Fludarabine modulates immune response and extends in vivo survival of adoptively transferred CD8 T cells in patients with metastatic melanoma. PLoS ONE, 2009, 4(3): e4749
    20 Cao M, Cabrera R, Xu Y, et al. Gamma irradiation alters the phenotype and function of CD4(+)CD25(+) regulatory T cells. Cell Biol Int. 2009 [Epub ahead of print]
    1 Tian H, Liu X. Survey and analysis on sterigmatocystin contaminated in grains in China. Wei Sheng Yan Jiu. 2004, 33(5): 606-608
    2 Mahmoud AL. Mycotoxin-producing potential of fungi associated with qat (Catha edulis) leaves in Yemen. Folia Microbiol. 2000, 45(5): 452-456
    3 Engelhart S, Loock A, Skutlarek D, et al. Occurrence of toxigenic Aspergillus versicolor isolates and sterigmatocystin in carpet dust fromdamp indoor environments. Appl Environ Microbiol. 2002, 68(8): 3886-3890
    4 Huang XH, Zhang XH, Yan X, Yin GR, Tan YW, Li YH, et al. Effects of sterigmatocystin on interleukin-2 secretion of human peripheral blood mononuclear cells in vitro[J]. J Hygiene Res(卫生研究), 2002, 31(2): 112-114
    5 Xing LX, Zhang XH, Yin GR, Li YH, Wang JL, Yan X, et al. Effect of sterigmatocystin on interleukin-2 expression and secretion of murine peritoneal macrophage cells in vitro. Chin J Pharmacol Toxicol(中国药理学与毒理学杂志), 2005, 19(3): 205-208
    6 Xing LX, Zhang XH, Li YH, Zuo LF, Yan X, Wang JL, et al. Effects of Sterigmatocystin on TAP1 and LMP2 Expression of Human Peripheral Blood Mononuclear Cells invitro. Chin J Clin Oncol(中国肿瘤临床), 2004, 31(4): 189-192
    7 Minamizaki T, Yoshiko Y, Kozai K, et al. EP2 and EP4 receptors differentially mediate MAPK pathways underlying anabolic actions of prostaglandin E(2) on bone formation in rat calvaria cell cultures. Bone. 2009, 21 [Epub ahead of print]
    8 Kohut G, Adám AL, Fazekas B, et al. N-starvation stress induced FUM gene expression and fumonisin production is mediated via the HOG-type MAPK pathway in Fusarium proliferatum. Int J Food Microbiol. 2009, 130(1): 65-69
    9 Pestka JJ. Mechanisms of deoxynivalenol-induced gene expression and apoptosis. Food Addit Contam Part A Chem Anal Control Expo Risk Assess. 2008, 25(9): 1128-1140
    10 Hong JT, Yen JH, Wang L, et al. Regulation of heme oxygenase-1 expression and MAPK pathways in response to kaempferol and rhamnocitrin in PC12 cells. Toxicol Appl Pharmacol. 2009,
    11 Peng S, Zhou G, Luk KD, et al. Strontium promotes osteogenic differentiation of mesenchymal stem cells through the Ras/MAPK signaling pathway. Cell Physiol Biochem. 2009, 23(1-3): 165-174
    12 Colombatti M, Grasso S, Porzia A, et al. The prostate specific membrane antigen regulates the expression of IL-6 and CCL5 in prostate tumour cells by activating the MAPK pathways. PLoS ONE. 2009, 4(2): e4608
    13 Lu ZY, Jensen LE, Huang Y, et al. The up-regulation of monocyte chemoattractant protein-1 (MCP-1) in Ea.hy 926 endothelial cells under long-term low folate stress is mediated by the p38 MAPK pathway. Atherosclerosis. 2008, 13 [Epub ahead of print]
    14 Shen Z, Chen L, Hao F, et al. Transcriptional regulation of Foxp3 gene: Multiple signal pathways on the road. Med Res Rev. 2009, 6 [Epub ahead of print]
    15 Friedline RH, Brown DS, Nguyen H, et al. CD4+ regulatory T cells require CTLA-4 for the maintenance of systemic tolerance. J Exp Med. 2009, 206(2): 421-434
    16 Zorn E, Nelson EA, Mohseni M, et al. IL-2 regulates FOXP3 expression in human CD4+CD25+ regulatory T cells through a STAT-dependent mechanism and induces the expansion of these cells in vivo. Blood. 2006, 108(5): 1571-1579
    17 Pyzik M, Piccirillo CA. TGF-beta1 modulates Foxp3 expression and regulatory activity in distinct CD4+ T cell subsets. Cancer Cell. J Leukoc Biol. 2007, 82(2): 335-346
    18 Sauer S, Bruno L, Hertweck A, et al. T cell receptor signaling controls Foxp3 expression via PI3K, Akt, and mTOR. Proc Natl Acad Sci U S A. 2008, 105(22): 7797-7802
    19 Samon JB, Champhekar A, Minter LM, et al. Notch1 and TGFbeta1 cooperatively regulate Foxp3 expression and the maintenance of peripheral regulatory T cells. J Blood. 2008, 112(5): 1813-1821
    20 Fragale A, Gabriele L, Stellacci E, et al. IFN regulatory factor-1 negatively regulates CD4+ CD25+ regulatory T cell differentiation by repressing Foxp3 expression. J Immunol. 2008, 181(3): 1673-1682
    21 Elias KM, Laurence A, Davidson TS, et al. Retinoic acid inhibits Th17 polarization and enhances FoxP3 expression through a Stat-3/Stat-5independent signaling pathway. Blood. 2008, 111(3): 1013-1020
    22 Xu G, Xia JH, Zhou H, et al. Interleukin-6 is essential for Staphylococcal exotoxin B-induced T regulatory cell insufficiency in nasal polyps. Clin Exp Allergy. 2009, 21 [Epub ahead of print]
    23 Huber S, Schrader J, Fritz G, et al. P38 MAP kinase signaling is required for the conversion of CD4+CD25- T cells into iTreg. PLoS ONE. 2008, 3(10): e3302
    24 Luo X, Zhang Q, Liu V, et al. Cutting edge: TGF-beta-induced expression of Foxp3 in T cells is mediated through inactivation of ERK. J Immunol. 2008, 180(5): 2757-2761
    25 Dobreva ZG, Miteva LD, Stanilova SA. The inhibition of JNK and p38 MAPKs downregulates IL-10 and differentially affects c-Jun gene expression in human monocytes. Immunopharmacol Immunotoxicol. 2009, 23:1-7 [Epub ahead of print]
    1 Baecher-Allan C, Brown JA, Freeman GJ, et al. CD4+CD25high regulatory cells in human peripheral blood. J Immunol, 2001, 167(3): 1245-1253
    2 Cao D, Malmstrom V, Baecher-Allan C, et al. Isolation and functional characterization of regulatory CD25brightCD4+ T cells from the target organ of patients with rheumatoid arthritis. Eur J Immunol, 2003, 33(1): 215-223
    3 Sakaguchi S. Naturally arising CD4+ regulatory T cells for immunologic self-tolerance and negative control of immune responses. Ann Rev Immunol, 2004, 22: 531-562
    4 Thornton AM and Shevach EM. CD4+CD25+ immunoregulatory T cells suppress polyclonal T cell activation in vitro by inhibiting interleukin 2 production. J Exp Med, 1998, 188(2): 287-296.
    5 Zou W. Regulatory T cells, tumour immunity and immunotherapy. Nat Rev Immunol, 2006, 6(4): 295-307.
    6 Baecher-Allan C, Brown JA, Freeman GJ, et al. CD4+CD25highregulatory cells in human peripheral blood. J Immunol, 2001, 167(3): 1245-1253
    7 Huehn J, Polansky JK, Hamann A. Epigenetic control of FOXP3 expression: the key to a stable regulatory T-cell lineage? Nat Rev Immunol. 2009, 9(2): 83-89
    8 Seddiki N, Santner-Nanan B, Martinson J, et al. Expression of interleukin (IL)-2 and Il-7 receptors discriminates between human regulatory and activated T cells. J Exp Med, 2006, 203(7): 1693-1700
    9 Kiniwa Y, Miyahara Y, Wang HY, et al. CD8+ Foxp3+ regulatory T cells mediate immunosuppression in prostate cancer. Clin Cancer Res. 2007, 13(23): 6947-6958
    10 Williams LM and Rudensky A. Maintenance of the FoxP3-dependent developmental program in mature regulatory T cells requires continued expression of FoxP3. Nat Immunol. 2007, 8(3): 277-284
    11 Seddiki N, Santner-Nanan B, Tangye SG, et al. Persistence of naive CD45RA+ regulatory T cells in adult life. Blood, 2006, 107(7): 2830-2838
    12 Hori S, Nomura T and Sakaguchi S. Control of regulatory T cell development by the transcription factor Foxp3. Science, 2003, 299(5609): 1057-1061
    13 Yagi H, Nomura T, Nakamura K, et al. Crucial role of FOXP3 in the development and function of human CD25+CD4+ regulatory T cells. Int Immunol, 2004, 16(11): 1643-1656
    14 Gavin MA Torqerson TR, Rudensky AY, et al. Single-cell analysis of nomaland FOXP3-mautant human T cells : FOXP3 expression without regulatory T cell development [J]. Proc Natl Acad Sci USA, 2006, 103(17): 6659-6664
    15 Bennett CL, Christie J, Ramsdell F, et al. The immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome (IPEX) is caused by mutations of FOXP3. Nat Genet, 2001, 27(1): 20-21
    16 Wildin RS, Ramsdell F, Peake J, et al. X-linked neonatal diabetes mellitus, enteropathy and endocrinopathy syndrome is the human equivalent ofmouse scurfy. Nat Genet, 2001, 27(1): 18-20
    17 Bennett CL and Ochs HD. IPEX is a unique X-linked syndrome characterized by immune dysfunction, polyendocrinopathy, enteropathy, and a variety of autoimmune phenomena. Curr Opin Pediatr, 2001, 13(6): 533-538
    18 Setoguchi R, Hori S, Takahashi T, et al. Homeostatic maintenance of natural Foxp3(+) CD25(+) CD4(+) regulatory T cells by interleukin (IL)-2 and induction of autoimmune disease by IL-2 neutralization. J Exp Med. 2005, 201(5): 723-735
    19 Hong J, Li N, Zhang X, et al. Induction of CD4+CD25+ regulatory T cells by copolymer-I through activation of transcription factor Foxp3. Proc Natl Acad Sci U S A. 2005, 102(18): 6449-6454
    20 Heather Kopf, Gonzalo M. de la Rosa, O.M.Zack Howard, et al. Rapamycin inhibits differentiation of Th17 cells and promotes generation of FoxP3+ T regulatory cells. International Immunopharmacology. 2007, 7(13): 1819-1824
    21 Kim JM and Rudensky A. The role of the transcription factor Foxp3 in the development of regulatory T cells. Immunol Rev. 2006, 212: 86-98
    22 Ziegler SF. FOXP3: of mice and men. Annu Rev Immunol. 2006, 24: 209–226
    23 Fontenot JD, Gavin MA and Rudensky AY. Foxp3 programs the development and function of CD4+CD25+ regulatory T cells. Nat Immunol, 2003, 4(4): 330-336
    24 Fontenot JD, Rasmussen JP, Willliams LM, et al. Regulatory T cell lineage specification by the forkhead transcription factor FoxP3. Immunity, 2005, 22(3): 329-341
    25 Wan YY and Flavell RA. Identifying Foxp3-expressing suppressor T cells with a bicistronic reporter. Proc Natl Acad Sci USA, 2005, 102(14): 5126-5131
    26 Sakaguchi S. The origin of FOXP3-expressing CD4+ regulatory T cells: thymus or periphery. J Clin Invest, 2003, 112(9): 1310-1312
    27 Powell BR, Buist NR and Stenzel P. An X-linked syndrome of diarrhea, polyendocrinopathy, and fatal infection in infancy. J Pediatr, 1982, 100(5): 731-737
    28 Groux H, O'Garra A, Bigler M, et al. A CD4+ T-cell subset inhibits antigen-specific T-cell responses and prevents colitis. Nature, 1997, 389(6652): 737-742
    29 Akdis CA, Blesken T, Akdis M, et al. Role of interleukin 10 in specific immunotherapy. J Clin Invest, 1998, 102(1): 98-106
    30 Akdis M, Verhagen J, Taylor A, et al. Immune responses in healthy and allergic individuals are characterized by a fine balance between allergen-specific T regulatory 1 and T helper 2 cells. J Exp Med, 2004, 199(11): 1567-1575
    31 Roncarolo MG, Levings MK and Traversari C. Differentiation of T regulatory cells by immature dendritic cells. J Exp Med, 2001, 193(2): F5-F9
    32 Shevach EM. Certified professionals: CD4 + CD25 + suppressor T cells. J Exp Med, 2001, 193(11): F41-F46
    33 Stephens LA, Mottet C, Mason D, et al. Human CD4 + CD25+ thymocytes and peripheral T cells have immune suppressive activity in vitro. Eur J Immunol, 2001, 31(4): 1247-1254
    34 Jonuleit H, Schmitt E, Stassen M, et al. Identification and functional characterization of human CD4 + CD25 + T cells with regulatory properties isolated from peripheral blood. J Exp Med, 2001, 193(11): 1285-1294
    35 Shevach EM. CD4 + CD25 + suppressor T cells: more questions than answers. Nat Rev Immunol, 2002, 2(6): 389-400
    36 Jordan MS, Boesteanu A, Reed AJ, et al. Thymic selection of CD4 + CD25 + regulatory T cells induced by an agonist self-peptide. Nat Immunol, 2001, 2(4): 301-306
    37 Diechman D, Plottner H, Berchtold S, et al. Ex vivo isolation and characterization of CD4 + CD25 + T cells with regulatory properties from human blood. J Exp Med, 2001, 193(11): 1303-1310
    38 Piccirillo CA and Shevach EM. Naturally-occurring CD4+CD25+ immunoregulatory T cells: central players in the arena of peripheral tolerance. Semin Immunol, 2004, 16 (2): 81-88
    39 Zenclussen AC, Gerlof K, Zenclussen ML, et al. Abnormal T-cell reactivity against paternal antigens in spontaneous abortion: adoptive transfer of pregnancy-induced CD4+CD25+ T regulatory cells prevents fetal rejection in a murine abortion model. Am J Pathol, 2005, 166(3): 811-822
    40 Stock P, Akbari O, Berry G, et al. Induction of T helper type 1-like regulatory cells that express Foxp3 and protect against airway hyper-reactivity. Nat Immunol, 2004, 5(11): 1149-1156
    41 Akdis M, Verhagen J, Taylor A, et al. Immune responses in healthy and allergic individuals are characterized by a fine balance between allergen-specific T regulatory 1 and T helper 2 cells. J Exp Med, 2004, 199(11): 1567-1575
    42 Belkaid Y and Rouse BT. Natural regulatory T cells in infectious disease. Nat Immunol, 2005, 6(4): 353-360
    43 Chen TC, Cobbold SP, Fairchild PJ, et al. Generation of anergic and regulatory T cells following prolonged exposure to a harmless antigen. J Immunol, 2004, 172(10): 5900-5907
    44 Wei WZ, Morris GP and Kong YC. Anti-tumor immunity and autoimmunity: a balancing act of regulatory T cells. Cancer Immunol Immunother, 2004, 53(2): 73-78
    45 Horlock C, Stott B, Dyson PJ, et al. The effects of trastuzumab on the CD4+CD25+FoxP3+ and CD4+IL17A+ T-cell axis in patients with breast cancer. Br J Cancer. 2009, 100(7): 1061-1067
    46 Qin FX. Dynamic behavior and function of Foxp3+ regulatory T cells in tumor bearing host. Cell Mol Immunol. 2009, 6(1): 3-13
    47 Gobert M, Treilleux I, Bendriss-Vermare N, et al. Regulatory T cells recruited through CCL22/CCR4 are selectively activated in lymphoid infiltrates surrounding primary breast tumors and lead to an adverseclinical outcome. Cancer Res. 2009, 69(5): 2000-2009
    48 Franceschini D, Paroli M, Francavilla V, et al. PD-L1 negatively regulates CD4+CD25+Foxp3+ Tregs by limiting STAT-5 phosphorylation in patients chronically infected with HCV. J Clin Invest. 2009, 119(3): 551-564
    49 Curiel TJ, Coukos G, Zou L, et al. Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat Med, 2004, 10 (9): 942-949
    50 Somasundaram R, Jacob L, Swoboda R, et al. Inhibition of cytolytic T lymphocyte proliferation by autologous CD4 + /CD25 + regulatory T cells in a colorectal carcinoma patients is mediated by transforming growth factor-β. Cancer Res, 2002, 62 (18): 5267-5272
    51 Wolf AM, Wolf D, Steurer M, et al. Increase of regulatory T cells in the peripheral blood of cancer patients. Clin Cancer Res, 2003, 9 (2): 606-612
    52 Sasada T, Kimura M, Yoshida Y, et al. CD4 + CD25 + regulatory T cells in patients with gastrointestinal malignancies. Possible involvement of regulatory T cells in disease progression. Cancer, 2003, 98 (5): 1089-1099
    53 Woo EY, Chu CS, Goletz TJ, et al. Regulatory CD4 + CD25 + T cells in tumors from patients with early-stage non-small cell lung cancer and late-stage ovarian cancer. Cancer Res, 2001, 61 (12): 4766-4772
    54 Woo EY, Yeh H, Chu CS, et al. Regulatory T cells from lung cancer patients directly inhibit autologous T cell proliferation. J Immunol, 2002, 168 (9): 4272-4276
    55 Cureil TJ, Coukos G, Zou L, et al. Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat Med, 2004, 10 (9): 942-949
    56 Wallen H, Thompson JA, Reilly JZ, et al. Fludarabine modulates immune response and extends in vivo survival of adoptively transferred CD8 T cells in patients with metastatic melanoma. PLoS ONE. 2009, 4(3): e4749
    57 Cao M, Cabrera R, Xu Y, et al. Gamma irradiation alters the phenotype and function of CD4(+)CD25(+) regulatory T cells. Cell Biol Int. 2009,112-113: 185-191
    58 Onizuka S, Tawara I, Shimizu J, et al. Tumor rejection by in vivo administration of anti-CD25 (interleukin-2 receptor alpha) monoclonal antibody. Cancer Res, 1999, 59(13): 3128-3133
    59 Somasundaram R, Jacob L, Swoboda R, et al. Inhibition of cytolytic T lymphocyte proliferation by autologous CD4 + /CD25 + regulatory T cells in a colorectal carcinoma patients is mediated by transforming growth factor-β. Cancer Res, 2002, 62(18): 5267-7272
    60 Jones E, Dahm-Vicker M, Simon A, et al. Depletion of CD25+ regulatory cells results in suppression of melanoma growth and induction of autoreactivity in mice. Cancer Immun, 2002, 2: 1-12
    61 Shimizu J, Yamazaki S and Sakaguchi S. Induction of tumor immunity by removing CD25 + CD4 + T cells: a common basis between tumor immunity an autoimmunity. J Immunol, 1999, 163(10): 5211-5218
    62 Casares N, Arribillaga L, Sarobe P, et al. CD4 + /CD25 + regulatory cells inhibit activation of tumor-primed CD4 + T cells with IFN-gamma -dependent antiangiogenic activity, as well as long-lasting tumor immunity elicited by peptide vaccination. J Immunol, 2003, 171(11): 5931-5939
    63 Roncarolo MG, Bacchetta R, Bordignon C, et al. Type 1 T regulatory cells. Immunol Rev, 2001, 182: 68-79
    64 Weiner HL. Induction and mechanism of action of transforming growth factor-beta-secreting Th3 regulatory cells. Immunol Rev, 2001, 182: 207-214
    65 Yudoh K, Matsuno H, Nkazawa F, et al. Reduced expression of the regulatory CD4+T cell subset is related to Th1/Th2 balance and disease severity in rheumatoid arthritis. Arthritis Rheum, 2000, 43(3): 617-627
    66 Fukaura H, Kent SC, Pietrusewicz MJ, et al. Induction of circulating myelin basic protein and proteolipid protein-specific transforming growth factor-beta-1-secreting Th3 T cells by oral administration of myelin in multiple sclerosis patients. J Clin Invest, 1996, 98(1): 70-77
    67 Siegmund K, Rückert B, Ouaked N, et al. Unique phenotype of humantonsillar and in vitro-induced FOXP3+CD8+ T cells. J Immunol. 2009, 182(4): 2124-2130
    68 Mahic M, Henjum K, Yaqub S, et al. Generation of highly suppressive adaptive CD8(+)CD25(+)FOXP3(+) regulatory T cells by continuous antigen stimulation. Eur J Immunol. 2008, 38(3): 640-646
    69 Banchereau J, Briere F, Caux C, et al. Immunobiology of dendritic cells. Annu Rev Immunol. 2000, 18: 767-811
    70 Wallet MA, Sen P and Tisch R. Immunoregulation of dendritic cells. Clin Med Res. 2005, 3(3): 166-175
    71 Dhodapkar MV, Steinman RM, Krasovsky J, et al. Antigen-specific inhibition of effector T cell function in humans after injection of immature dendritic cells. J Exp Med. 2001, 193(2): 233-238
    72 Hackstein H, Morelli AE and Thomson AW. Designer dendritic cells for tolerance induction: guided not misguided missiles. Trends Immunol. 2001, 22(8): 437-442
    73 Mahnke K, Qian Y, Knop J, et al. Induction of CD4+/CD25+ regulatory T cells by targeting of antigens to immature dendritic cells. Blood. 2003, 101(12): 4862-4869
    74 Cong Y, Konrad A, Iqbal N, et al. Generation of antigen-specific, Foxp3-expressing CD4+ regulatory T Cells by inhibition of APC proteosome function. J Immunol. 2005, 174(5): 2787-2795
    75 Yang K, Li D, Luo M, et al. Generation of HSP60-specific regulatory T cell and effect on atherosclerosis. Cell Immunol. 2006, 243(2): 90-95
    76 Di Nicola M, Carlo-Stella C, Magni M, et al. Human bone marrow stromal cells suppress T-lymphocyte proliferation induced by cellular or nonspecific mitogenic stimuli. Blood, 2002, 99(10): 3838-3843
    77 Bartholomew A, Sturgeon C, Siatskas M, et al. Mesenchymal stem cells suppress lymphocyte proliferation in vitro and prolong skin graft survival in vivo. Exp Hematol, 2002, 30(1): 42-48
    78 Le Blanc K, Rasmusson I, Sunberg B, et al. Treatment of severe graft-versus-host disease with third party haploidentical mesenchymalstem cells. Lancet, 2004, 363(9419): 1439-1441
    79 Poggi A, Zocchi MR. Role of bone marrow stromal cells in the generation of human CD8+ regulatory T cells. Hum Immunol. 2008, 69(11): 755-759
    80 Leveque L, Deknuydt F, Bioley G, et al. Interleukin 2-mediated conversion of ovarian cancer-associated CD4+ regulatory T cells into proinflammatory interleukin 17-producing helper T cells. J Immunother. 2009, 32(2):101-108
    81 Swee LK, Bosco N, Malissen B, et al. Expansion of peripheral NTreg by FLT3L treatment. Blood. 2009 [Epub ahead of print]
    82 Deknuydt F, Bioley G, Valmori D, et al. IL-1beta and IL-2 convert human Treg into T(H)17 cells. Clin Immunol. 2009 Feb 9. [Epub ahead of print]
    83 Chen W, Jin W, Hardegen N, et al. Conversion of peripheral CD4+CD25- naive T cells to CD4+CD25+ regulatory T cells by TGF-beta induction of transcription factor Foxp3. J Exp Med. 2003, 198(12): 1875-1886
    84 Pyzik M, Piccirillo CA. TGF-beta1 modulates Foxp3 expression and regulatory activity in distinct CD4+ T cell subsets. J Leukoc Biol. 2007, 82(2): 335-346
    85 Huber S, Stahl FR, Schrader J, et al. Activin a promotes the TGF-beta-induced conversion of CD4+CD25- T cells into Foxp3+ induced regulatory T cells. J Immunol. 2009, 182(8): 4633-4640
    86 Wei Tang, Yang Yang, Fan Zhang, et al. Prevention of graft-versus-host disease by a novel immunosuppressant, (5R)-5-hydroxytriptolide (LLDT-8), through expansion of regulatory T cells. International Immunopharmacology, 2005, 5(13-14): 1904-1913
    87 Yvonne Vercoulen, Isme De Kleer, Sylvia Kamphuis, et al. Human Heat Shock Protein 60 Induces Differentiation of Cd4+Cd3//Q T-Cells Into Foxp3+Cd4+Cd25//0 Regulatory T-Cells. Clin Immunol, 2006, 119(1): S19-S20
    88 Suzana Radulovic MD, Mikila R. Jacobson PhD, Stephen R. Durham MD, et al. Grass pollen immunotherapy induces Foxp3-expressing CD4+CD25+ cells in the nasal mucosa. J. Allergy Clin Immunol, 2008, 121(6):1467-1472
    89 Yoolhee Yang, Jin Ho Paik, Daeho Cho, et al. Resveratrol induces the suppression of tumor-derived CD4+CD25+ regulatory T cells. International Immunopharma. 2008, 8(4): 542-547
    90 Cao M, Cabrera R, Xu Y, et al. Gamma irradiation alters the phenotype and function of CD4(+)CD25(+) regulatory T cells. Cell Biol Int. 2009 [Epub ahead of print]
    91 Huber S, Schrader J, Fritz G, et al. P38 MAP kinase signaling is required for the conversion of CD4+CD25- T cells into iTreg. PLoS ONE. 2008, 3(10): e3302
    92 Luo X, Zhang Q, Liu V, et al. Cutting edge: TGF-beta-induced expression of Foxp3 in T cells is mediated through inactivation of ERK. J Immunol. 2008, 180(5): 2757-2761
    93 Dobreva ZG, Miteva LD, Stanilova SA. The inhibition of JNK and p38 MAPKs downregulates IL-10 and differentially affects c-Jun gene expression in human monocytes. Immunopharmacol Immunotoxicol. 2009, 23: 1-7 [Epub ahead of print]

© 2004-2018 中国地质图书馆版权所有 京ICP备05064691号 京公网安备11010802017129号

地址:北京市海淀区学院路29号 邮编:100083

电话:办公室:(+86 10)66554848;文献借阅、咨询服务、科技查新:66554700