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.)
Precision Engineering of an Anti-HLA-A2 Chimeric Antigen Receptor in Regulatory T Cells for Transplant Immune Tolerance
Frontiers in Immunology ( IF 7.3 ) Pub Date : 2021-09-20 , DOI: 10.3389/fimmu.2021.686439
Yannick D Muller 1, 2 , Leonardo M R Ferreira 1, 2, 3 , Emilie Ronin 1, 2 , Patrick Ho 1, 2, 3 , Vinh Nguyen 1, 2 , Gaetano Faleo 1, 2 , Yu Zhou 4 , Karim Lee 1 , Kevin K Leung 5 , Nikolaos Skartsis 1, 6 , Anupurna M Kaul 1 , Arend Mulder 7 , Frans H J Claas 7 , James A Wells 5 , Jeffrey A Bluestone 2, 3 , Qizhi Tang 1, 2
Affiliation  

Infusion of regulatory T cells (Tregs) engineered with a chimeric antigen receptor (CAR) targeting donor-derived human leukocyte antigen (HLA) is a promising strategy to promote transplant tolerance. Here, we describe an anti-HLA-A2 CAR (A2-CAR) generated by grafting the complementarity-determining regions (CDRs) of a human monoclonal anti-HLA-A2 antibody into the framework regions of the Herceptin 4D5 single-chain variable fragment and fusing it with a CD28-ζ signaling domain. The CDR-grafted A2-CAR maintained the specificity of the original antibody. We then generated HLA-A2 mono-specific human CAR Tregs either by deleting the endogenous T-cell receptor (TCR) via CRISPR/Cas9 and introducing the A2-CAR using lentiviral transduction or by directly integrating the CAR construct into the TCR alpha constant locus using homology-directed repair. These A2-CAR+TCRdeficient human Tregs maintained both Treg phenotype and function in vitro. Moreover, they selectively accumulated in HLA-A2-expressing islets transplanted from either HLA-A2 transgenic mice or deceased human donors. A2-CAR+TCRdeficient Tregs did not impair the function of these HLA-A2+ islets, whereas similarly engineered A2-CAR+TCRdeficientCD4+ conventional T cells rejected the islets in less than 2 weeks. A2-CAR+TCRdeficient Tregs delayed graft-versus-host disease only in the presence of HLA-A2, expressed either by co-transferred peripheral blood mononuclear cells or by the recipient mice. Altogether, we demonstrate that genome-engineered mono-antigen-specific A2-CAR Tregs localize to HLA-A2-expressing grafts and exhibit antigen-dependent in vivo suppression, independent of TCR expression. These approaches may be applied towards developing precision Treg cell therapies for transplant tolerance.



中文翻译:

用于移植免疫耐受的调节性 T 细胞中抗 HLA-A2 嵌合抗原受体的精密工程

注入具有嵌合抗原受体 (CAR) 靶向供体来源的人类白细胞抗原 (HLA) 的调节性 T 细胞 (Tregs) 是促进移植耐受性的一种有前景的策略。在这里,我们描述了通过将人单克隆抗 HLA-A2 抗体的互补决定区 (CDR) 移植到赫赛汀 4D5 单链可变片段的框架区而产生的抗 HLA-A2 CAR (A2-CAR)并将其与 CD28-ζ 信号域融合。CDR 移植的 A2-CAR 保持了原始抗体的特异性。然后我们通过删除内源性 T 细胞受体 (TCR) 来生成 HLA-A2 单特异性人 CAR Tregs通过CRISPR/Cas9 并使用慢病毒转导或通过使用同源定向修复将 CAR 构建体直接整合到 TCR α 恒定基因座中来引入 A2-CAR。这些 A2-CAR + TCR缺陷的人类 Treg 保持了 Treg 表型和功能体外. 此外,它们选择性地积聚在从 HLA-A2 转基因小鼠或已故人类供体移植的 HLA-A2 表达胰岛中。A2-CAR + TCR缺陷的Tregs 不会损害这些 HLA-A2 +胰岛的功能,而类似设计的 A2-CAR + TCR缺陷CD4 +传统 T 细胞在不到 2 周的时间内排斥胰岛。A2-CAR + TCR缺陷性 Tregs 延迟移植-相对- 仅在 HLA-A2 存在时宿主疾病,由共转移的外周血单核细胞或受体小鼠表达。总之,我们证明了基因组工程化的单抗原特异性 A2-CAR Treg 定位于表达 HLA-A2 的移植物并表现出抗原依赖性体内抑制,独立于 TCR 表达。这些方法可用于开发针对移植耐受的精确 Treg 细胞疗法。

更新日期:2021-09-20
down
wechat
bug