当前位置: X-MOL 学术Proc. Natl. Acad. Sci. U.S.A. › 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.)
Posttranslational modifications by ADAM10 shape myeloid antigen-presenting cell homeostasis in the splenic marginal zone [Immunology and Inflammation]
Proceedings of the National Academy of Sciences of the United States of America ( IF 11.1 ) Pub Date : 2021-09-21 , DOI: 10.1073/pnas.2111234118
Nathalie Diener 1 , Jean-Fred Fontaine 2 , Matthias Klein 3, 4 , Thomas Hieronymus 5 , Florian Wanke 1 , Florian C Kurschus 1 , Andreas Ludwig 6 , Carl Ware 7 , Paul Saftig 8 , Tobias Bopp 3, 4 , Björn E Clausen 4, 9 , Ronald A Backer 4, 9
Affiliation  

The spleen contains phenotypically and functionally distinct conventional dendritic cell (cDC) subpopulations, termed cDC1 and cDC2, which each can be divided into several smaller and less well-characterized subsets. Despite advances in understanding the complexity of cDC ontogeny by transcriptional programming, the significance of posttranslational modifications in controlling tissue-specific cDC subset immunobiology remains elusive. Here, we identified the cell-surface–expressed A-disintegrin-and-metalloproteinase 10 (ADAM10) as an essential regulator of cDC1 and cDC2 homeostasis in the splenic marginal zone (MZ). Mice with a CD11c-specific deletion of ADAM10 (ADAM10ΔCD11c) exhibited a complete loss of splenic ESAMhi cDC2A because ADAM10 regulated the commitment, differentiation, and survival of these cells. The major pathways controlled by ADAM10 in ESAMhi cDC2A are Notch, signaling pathways involved in cell proliferation and survival (e.g., mTOR, PI3K/AKT, and EIF2 signaling), and EBI2-mediated localization within the MZ. In addition, we discovered that ADAM10 is a molecular switch regulating cDC2 subset heterogeneity in the spleen, as the disappearance of ESAMhi cDC2A in ADAM10ΔCD11c mice was compensated for by the emergence of a Clec12a+ cDC2B subset closely resembling cDC2 generally found in peripheral lymph nodes. Moreover, in ADAM10ΔCD11c mice, terminal differentiation of cDC1 was abrogated, resulting in severely reduced splenic Langerin+ cDC1 numbers. Next to the disturbed splenic cDC compartment, ADAM10 deficiency on CD11c+ cells led to an increase in marginal metallophilic macrophage (MMM) numbers. In conclusion, our data identify ADAM10 as a molecular hub on both cDC and MMM regulating their transcriptional programming, turnover, homeostasis, and ability to shape the anatomical niche of the MZ.



中文翻译:

ADAM10 的翻译后修饰塑造脾边缘区髓样抗原呈递细胞稳态[免疫学和炎症]

脾脏包含表型和功能上不同的常规树突状细胞 (cDC) 亚群,称为 cDC1 和 cDC2,每个亚群都可以分为几个较小且特征较少的亚群。尽管通过转录编程在理解 cDC 个体发育的复杂性方面取得了进展,但翻译后修饰在控制组织特异性 cDC 子集免疫生物学中的重要性仍然难以捉摸。在这里,我们将细胞表面表达的 A-去整合素和金属蛋白酶 10 (ADAM10) 鉴定为脾边缘区 (MZ) 中 cDC1 和 cDC2 稳态的重要调节剂。CD11c 特异性缺失 ADAM10 (ADAM10 ΔCD11c ) 的小鼠表现出脾脏 ESAM hi完全丧失cDC2A,因为 ADAM10 调节这些细胞的承诺、分化和存活。ESAM hi cDC2A 中由 ADAM10 控制的主要途径是 Notch、参与细胞增殖和存活的信号传导途径(例如,mTOR、PI3K/AKT 和 EIF2 信号传导)和 EBI2 介导的 MZ 内定位。此外,我们发现 ADAM10 是调节脾脏中 cDC2 亚群异质性的分子开关,因为在 ADAM10 ΔCD11c小鼠中 ESAM hi cDC2A的消失被一个与外周淋巴中常见的 cDC2 非常相似的 Clec12a + cDC2B 亚群的出现所补偿节点。此外,在 ADAM10 ΔCD11c在小鼠中,cDC1 的终末分化被消除,导致脾脏Langerin + cDC1 数量严重减少。在受干扰的脾 cDC 隔室旁边,CD11c +细胞上的 ADAM10 缺乏导致边缘嗜金属巨噬细胞 (MMM) 数量增加。总之,我们的数据将 ADAM10 确定为 cDC 和 MMM 的分子中心,调节它们的转录编程、周转、稳态和塑造 MZ 解剖生态位的能力。

更新日期:2021-09-16
down
wechat
bug