当前位置: X-MOL 学术Front. Immunol. › 论文详情
Our official English website, www.x-mol.net, welcomes your feedback! (Note: you will need to create a separate account there.)
Endogenous T Cell Receptor Rearrangement Represses Aggressive Central Nervous System Autoimmunity in a TcR-Transgenic Model on the Non-Obese Diabetic Background
Frontiers in Immunology ( IF 5.7 ) Pub Date : 
Asmita Pradeep Yeola, Prenitha Mercy Ignatius Arokia Doss, Joanie Baillargeon, Irshad Akbar, Benoit Mailhot, Mohammad Balood, Sébastien Talbot, Ana Carrizosa Anderson, Steve Lacroix, Manu Rangachari

The T cell response to central nervous system (CNS) antigen in experimental autoimmune encephalomyelitis (EAE) permits one to model the immune aspects of multiple sclerosis. 1C6 transgenic mice on the non-obese diabetic (NOD) background possess a class II-restricted T cell receptor (TcR; Vα5-Vβ7) specific for the encephalitogenic peptide myelin oligodendrocyte glycoprotein (MOG)[35−55]. It remains to be determined what role is played by allelic inclusion in shaping the TcR repertoire of these mice. Here, we show that 1C6 T cells display substantial promiscuity in their expression of non-transgenically derived Vα chains. Further, enforced expression of the transgenic TcR in 1C6 × Rag1−/− mice profoundly disrupted thymic negative selection and led to a sharp decrease in the number of mature peripheral T cells. 1C6 × Rag1−/− mice developed spontaneous EAE at a significant frequency and rapidly developed fatal EAE upon immunization with myelin oligodendrocyte glycoprotein (MOG)[35−55]. Passive transfer of 1C6 × Rag1+/+ CD4+ T cells, but not CD8+ T cells or B cells, partially rescued 1C6 × Rag1−/− mice from severe EAE. FoxP3+ CD4+ Treg cells were present in the CNS of immunized 1C6 mice, as well as immunized 1C6 × Rag1−/− that had been supplemented with 1C6 CD4+ T cells. However, they were not observed in 1C6 × Rag1−/− that did not receive Rag1-sufficient 1C6 CD4+. Further, in vivo blockade of Treg accelerated the onset of symptoms in 1C6 mice immunized with MOG[35−55], indicating the pertinence of Treg-mediated control of autoimmune inflammation in this model. Thus, TcR allelic inclusion is crucial to the generation of FoxP3+ CD4+ T cells necessary for the suppression of severe CNS autoimmunity.



中文翻译:

内源性T细胞受体重排抑制非肥胖糖尿病背景下TcR转基因模型中侵略性中枢神经系统自身免疫。

在实验性自身免疫性脑脊髓炎(EAE)中,T细胞对中枢神经系统(CNS)抗原的反应使人们能够模拟多发性硬化症的免疫方面。在非肥胖糖尿病(NOD)背景下的1C6转基因小鼠具有II型限制性T细胞受体(TcR;Vα5-Vβ7),其特异性针对脑病原性肽髓磷脂少突胶质糖蛋白(MOG)[35-55]。尚待确定等位基因包涵体在塑造这些小鼠的TcR库中起什么作用。在这里,我们显示1C6 T细胞在其非转基因来源的Vα链的表达中显示出大量的混杂。此外,转基因TcR在1C6×中的强制表达破布1-/-小鼠极大地破坏了胸腺的阴性选择,并导致成熟的外周血T细胞数量急剧减少。1C6×破布1-/-小鼠以髓鞘少突胶质细胞糖蛋白(MOG)免疫后,以明显的频率自发产生EAE,并迅速产生致命性EAE [35-55]。被动转移1C6×破布1+ / + CD4 + T细胞,而非CD8 + T细胞或B细胞,部分获救1C6×破布1来自严重EAE的-/-小鼠。FoxP3 + CD4 + T reg细胞存在于1C6免疫小鼠和1C6×破布1-/-已经补充了1C6 CD4 + T细胞。但是,在1C6中未观察到它们×破布1没有收到Rag1足够的1C6 CD4 +的-/-。进一步,体内T reg的阻断加速了用MOG免疫的1C6小鼠的症状发作[35-55],表明该模型中T reg介导的自身免疫炎症控制的相关性。因此,TcR等位基因包涵对于抑制严重的CNS自身免疫所必需的FoxP3 + CD4 + T细胞的产生至关重要。

更新日期:2020-01-15
down
wechat
bug