Surface translocation of ACE2 and TMPRSS2 upon TLR4/7/8 activation is required for SARS-CoV-2 infection in circulating monocytes

Abstract Infection of human peripheral blood cells by SARS-CoV-2 has been debated because immune cells lack mRNA expression of both angiotensin-converting enzyme 2 (ACE2) and transmembrane serine protease type 2 (TMPRSS2). Herein we demonstrate that resting primary monocytes harbor abundant cytoplas...

Full description

Saved in:
Bibliographic Details
Published inCell discovery Vol. 8; no. 1; p. 89
Main Authors Yao, Yi, Subedi, Kalpana, Liu, Tingting, Khalasawi, Namir, Pretto-Kernahan, Carla Diana, Wotring, Jesse William, Wang, Jie, Yin, Congcong, Jiang, Aimin, Fu, Chunmei, Dimitrion, Peter, Li, Jia, Veenstra, Jesse, Yi, Qijun, McKinnon, Kathy, McKinnon, John Ernest, Sexton, Jonathan Zachary, Zhou, Li, Mi, Qing-Sheng
Format Journal Article
LanguageEnglish
Published London Springer Nature B.V 09.09.2022
Springer Nature Singapore
Nature Publishing Group
Subjects
Online AccessGet full text

Cover

Loading…
More Information
Summary:Abstract Infection of human peripheral blood cells by SARS-CoV-2 has been debated because immune cells lack mRNA expression of both angiotensin-converting enzyme 2 (ACE2) and transmembrane serine protease type 2 (TMPRSS2). Herein we demonstrate that resting primary monocytes harbor abundant cytoplasmic ACE2 and TMPRSS2 protein and that circulating exosomes contain significant ACE2 protein. Upon ex vivo TLR4/7/8 stimulation, cytoplasmic ACE2 was quickly translocated to the monocyte cell surface independently of ACE2 transcription, while TMPRSS2 surface translocation occurred in conjunction with elevated mRNA expression. The rapid translocation of ACE2 to the monocyte cell surface was blocked by the endosomal trafficking inhibitor endosidin 2, suggesting that endosomal ACE2 could be derived from circulating ACE2-containing exosomes. TLR-stimulated monocytes concurrently expressing ACE2 and TMPRSS2 on the cell surface were efficiently infected by SARS-CoV-2, which was significantly mitigated by remdesivir, TMPRSS2 inhibitor camostat, and anti-ACE2 antibody. Mass cytometry showed that ACE2 surface translocation in peripheral myeloid cells from patients with severe COVID-19 correlated with its hyperactivation and PD-L1 expression. Collectively, TLR4/7/8-induced ACE2 translocation with TMPRSS2 expression makes circulating monocytes permissive to SARS-CoV-2 infection.
Bibliography:ObjectType-Article-1
SourceType-Scholarly Journals-1
ObjectType-Feature-2
content type line 23
ISSN:2056-5968
2056-5968
DOI:10.1038/s41421-022-00453-8