Back to Search Start Over

Posttranslational modifications by ADAM10 shape myeloid antigen-presenting cell homeostasis in the splenic marginal zone.

Authors :
Diener N
Fontaine JF
Klein M
Hieronymus T
Wanke F
Kurschus FC
Ludwig A
Ware C
Saftig P
Bopp T
Clausen BE
Backer RA
Source :
Proceedings of the National Academy of Sciences of the United States of America [Proc Natl Acad Sci U S A] 2021 Sep 21; Vol. 118 (38).
Publication Year :
2021

Abstract

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 (ADAM10 <superscript>ΔCD11c</superscript> ) exhibited a complete loss of splenic ESAM <superscript>hi</superscript> cDC2A because ADAM10 regulated the commitment, differentiation, and survival of these cells. The major pathways controlled by ADAM10 in ESAM <superscript>hi</superscript> 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 ESAM <superscript>hi</superscript> cDC2A in ADAM10 <superscript>ΔCD11c</superscript> mice was compensated for by the emergence of a Clec12a <superscript>+</superscript> cDC2B subset closely resembling cDC2 generally found in peripheral lymph nodes. Moreover, in ADAM10 <superscript>ΔCD11c</superscript> mice, terminal differentiation of cDC1 was abrogated, resulting in severely reduced splenic Langerin <superscript>+</superscript> cDC1 numbers. Next to the disturbed splenic cDC compartment, ADAM10 deficiency on CD11c <superscript>+</superscript> 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.<br />Competing Interests: The authors declare no competing interest.

Details

Language :
English
ISSN :
1091-6490
Volume :
118
Issue :
38
Database :
MEDLINE
Journal :
Proceedings of the National Academy of Sciences of the United States of America
Publication Type :
Academic Journal
Accession number :
34526403
Full Text :
https://doi.org/10.1073/pnas.2111234118