当前位置: X-MOL 学术J. Allergy Clin. 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.)
Mast cells regulate CD4+ T-cell differentiation in the absence of antigen presentation
Journal of Allergy and Clinical Immunology ( IF 11.4 ) Pub Date : 2018-02-20 , DOI: 10.1016/j.jaci.2018.01.038
Hector Rodriguez Cetina Biefer , Timm Heinbokel , Hirofumi Uehara , Virginia Camacho , Koichiro Minami , Yeqi Nian , Suresh Koduru , Rachid El Fatimy , Ionita Ghiran , Alexander J. Trachtenberg , Miguel A. de la Fuente , Haruhito Azuma , Omid Akbari , Stefan G. Tullius , Anju Vasudevan , Abdallah Elkhal

Background

Given their unique capacity for antigen uptake, processing, and presentation, antigen-presenting cells (APCs) are critical for initiating and regulating innate and adaptive immune responses. We have previously shown the role of nicotinamide adenine dinucleotide (NAD+) in T-cell differentiation independently of the cytokine milieu, whereas the precise mechanisms remained unknown.

Objective

The objective of this study is to further dissect the mechanism of actions of NAD+ and determine the effect of APCs on NAD+-mediated T-cell activation.

Methods

Isolated dendritic cells and bone marrow–derived mast cells (MCs) were used to characterize the mechanisms of action of NAD+ on CD4+ T-cell fate in vitro. Furthermore, NAD+-mediated CD4+ T-cell differentiation was investigated in vivo by using wild-type C57BL/6, MC−/−, MHC class II−/−, Wiskott-Aldrich syndrome protein (WASP)−/−, 5C.C7 recombination-activating gene 2 (Rag2)−/−, and CD11b-DTR transgenic mice. Finally, we tested the physiologic effect of NAD+ on the systemic immune response in the context of Listeria monocytogenes infection.

Results

Our in vivo and in vitro findings indicate that after NAD+ administration, MCs exclusively promote CD4+ T-cell differentiation, both in the absence of antigen and independently of major APCs. Moreover, we found that MCs mediated CD4+ T-cell differentiation independently of MHC II and T-cell receptor signaling machinery. More importantly, although treatment with NAD+ resulted in decreased MHC II expression on CD11c+ cells, MC-mediated CD4+ T-cell differentiation rendered mice resistant to administration of lethal doses of L monocytogenes.

Conclusions

Collectively, our study unravels a novel cellular and molecular pathway that regulates innate and adaptive immunity through MCs exclusively and underscores the therapeutic potential of NAD+ in the context of primary immunodeficiencies and antimicrobial resistance.



中文翻译:

在没有抗原呈递的情况下肥大细胞调节CD4 + T细胞分化

背景

鉴于其独特的抗原摄取,加工和呈递能力,抗原呈递细胞(APC)对于启动和调节先天性和适应性免疫应答至关重要。我们先前已经显示了烟酰胺腺嘌呤二核苷酸(NAD +)在T细胞分化中的作用,而与细胞因子环境无关,而确切的机制仍然未知。

客观的

这项研究的目的是进一步剖析NAD +的作用机制,并确定APC对NAD +介导的T细胞活化的影响。

方法

分离的树突状细胞和骨髓肥大细胞(MCs)用于表征NAD +在体外对CD4 + T细胞命运的作用机制。此外,使用野生型C57BL / 6,MC -/-,MHC II类-/-,Wiskott-Aldrich综合征蛋白(WASP)-/-,5C对NAD +介导的CD4 + T细胞分化进行了体内研究.C7重组激活基因2(Rag2)-/-和CD11b-DTR转基因小鼠。最后,我们在单核细胞增生李斯特菌的背景下测试了NAD +对全身免疫应答的生理作用 感染。

结果

我们的体内和体外研究结果表明,施用NAD +后,MCs在无抗原存在且独立于主要APC的情况下仅促进CD4 + T细胞分化。此外,我们发现MC介导的CD4 + T细胞分化独立于MHC II和T细胞受体信号传导机制。更重要的是,尽管用NAD +治疗导致CD11c +细胞上MHC II表达降低,但MC介导的CD4 + T细胞分化使小鼠对致死剂量的L单核细胞增生李斯特菌具有抗性。

结论

总的来说,我们的研究揭示了一条新颖的细胞和分子途径,该途径专门通过MC调节先天性和适应性免疫,并强调了在主要免疫缺陷和抗微生物剂的情况下NAD +的治疗潜力。

更新日期:2018-02-20
down
wechat
bug