关键词: T helper type 1-like induced regulatory T cells (Th1-like iTregs) aplastic anemia (AA) bone marrow failure (BMF) interleukin-12 (IL-12) post-translational modifications (PTMs) protein arginine methyltransferase 5 (PRMT5) sirtuin 1 (SIRT1) symmetric histone di-methylation (H3R2me2s)

Mesh : Animals Mice Anemia, Aplastic Cell Differentiation / genetics Cytokines Disease Models, Animal Epigenesis, Genetic Interleukin-12 / pharmacology Sirtuin 1 Protein-Arginine N-Methyltransferases / genetics

来  源:   DOI:10.3389/fimmu.2023.1292049   PDF(Pubmed)

Abstract:
Induced regulatory T cells (iTregs) are a heterogeneous population of immunosuppressive T cells with therapeutic potential. Treg cells show a range of plasticity and can acquire T effector-like capacities, as is the case for T helper 1 (Th1)-like iTregs. Thus, it is important to distinguish between functional plasticity and lineage instability. Aplastic anemia (AA) is an autoimmune disorder characterized by immune-mediated destruction of hematopoietic stem and progenitor cells in the bone marrow (BM). Th1-like 1 iTregs can be potent suppressors of aberrant Th1-mediated immune responses such as those that drive AA disease progression. Here we investigated the function of the epigenetic enzyme, protein arginine methyltransferase 5 (PRMT5), its regulation of the iTreg-destabilizing deacetylase, sirtuin 1 (Sirt1) in suppressive Th1-like iTregs, and the potential for administering Th1-like iTregs as a cell-based therapy for AA.
We generated Th1-like iTregs by culturing iTregs with IL-12, then assessed their suppressive capacity, expression of iTreg suppression markers, and enzymatic activity of PRMT5 using histone symmetric arginine di-methylation (H3R2me2s) as a read out. We used ChIP sequencing on Th1 cells, iTregs, and Th1-like iTregs to identify H3R2me2s-bound genes unique to Th1-like iTregs, then validated targets using CHiP-qPCR. We knocked down PRMT5 to validate its contribution to Th1-like iTreg lineage commitment. Finally we tested the therapeutic potential of Th1-like iTregs using a Th1-mediated mouse model of AA.
Exposing iTregs to the Th1 cytokine, interleukin-12 (IL-12), during early events of differentiation conveyed increased suppressive function. We observed increased PRMT5 enzymatic activity, as measured by H3R2me2s, in Th1-like iTregs, which was downregulated in iTregs. Using ChIP-sequencing we discovered that H3R2me2s is abundantly bound to the Sirt1 promoter region in Th1-like iTregs to negatively regulate its expression. Furthermore, administering Th1-like iTregs to AA mice provided a survival benefit.
Knocking down PRMT5 in Th1-like iTregs concomitantly reduced their suppressive capacity, supporting the notion that PRMT5 is important for the superior suppressive capacity and stability of Th1-like iTregs. Conclusively, therapeutic administration of Th1-like iTregs in a mouse model of AA significantly extended their survival and they may have therapeutic potential.
摘要:
·诱导的调节性T细胞(iTregs)是具有治疗潜力的免疫抑制性T细胞的异质群体。Treg细胞表现出一定的可塑性,可以获得T效应子样能力,类似于T助手1(Th1)的iTregs。因此,区分功能可塑性和谱系不稳定性是很重要的。再生障碍性贫血(AA)是一种自身免疫性疾病,其特征是骨髓(BM)中造血干细胞和祖细胞的免疫介导破坏。Th1样1iTreg可以是异常Th1介导的免疫应答(例如驱动AA疾病进展的那些)的有效抑制剂。在这里,我们研究了表观遗传酶的功能,蛋白质精氨酸甲基转移酶5(PRMT5),它对iTreg去稳定脱乙酰酶的调节,抑制Th1样iTregs中的沉默蛋白1(Sirt1),以及将Th1样iTregs用作AA的细胞疗法的潜力。
我们通过用IL-12培养iTregs来产生Th1样iTregs,然后评估它们的抑制能力,iTreg抑制标记的表达,和PRMT5的酶活性,使用组蛋白对称精氨酸二甲基化(H3R2me2s)作为读数。我们在Th1细胞上使用了ChIP测序,iTregs,和Th1样iTregs来鉴定Th1样iTregs特有的H3R2me2s结合基因,然后使用CHiP-qPCR验证靶标。我们取消了PRMT5,以验证其对Th1样iTreg谱系承诺的贡献。最后,我们使用Th1介导的AA小鼠模型测试了Th1样iTregs的治疗潜力。
将iTregs暴露于Th1细胞因子,白细胞介素-12(IL-12),在分化的早期事件中,表达了抑制功能的增加。我们观察到PRMT5酶活性增加,用H3R2me2s测量,在类似Th1的iTregs中,在iTregs中被下调。使用ChIP测序,我们发现H3R2me2s与Th1样iTreg中的Sirt1启动子区域大量结合,从而负向调节其表达。此外,向AA小鼠施用Th1样iTregs提供了生存益处。
在Th1样iTregs中击倒PRMT5的同时降低了它们的抑制能力,支持PRMT5对于Th1样iTregs的优异抑制能力和稳定性很重要的观点。最后,Th1样iTregs在AA小鼠模型中的治疗性给药可显着延长其生存期,并且它们可能具有治疗潜力。
公众号