Back to Search Start Over

Renewal of embryonic and neonatal-derived cardiac-resident macrophages in response to environmental cues abrogated their potential to promote cardiomyocyte proliferation via Jagged-1–Notch1.

Authors :
Chen, Rong
Zhang, Shiqing
Liu, Fang
Xia, Lin
Wang, Chong
Sandoghchian Shotorbani, Siamak
Xu, Huaxi
Chakrabarti, Subrata
Peng, Tianqing
Su, Zhaoliang
Source :
Acta Pharmaceutica Sinica B; Jan2023, Vol. 13 Issue 1, p128-141, 14p
Publication Year :
2023

Abstract

Cardiac-resident macrophages (CRMs) play important roles in homeostasis, cardiac function, and remodeling. Although CRMs play critical roles in cardiac regeneration of neonatal mice, their roles are yet to be fully elucidated. Therefore, this study aimed to investigate the dynamic changes of CRMs during cardiac ontogeny and analyze the phenotypic and functional properties of CRMs in the promotion of cardiac regeneration. During mouse cardiac ontogeny, four CRM subsets exist successively: CX 3 CR1<superscript>+</superscript>CCR2<superscript>–</superscript>Ly6C<superscript>–</superscript>MHCII<superscript>–</superscript> (MP1), CX 3 CR1<superscript>low</superscript>CCR2<superscript>low</superscript>Ly6C<superscript>–</superscript>MHCII<superscript>–</superscript> (MP2), CX 3 CR1<superscript>–</superscript>CCR2<superscript>+</superscript>Ly6C<superscript>+</superscript>MHCII<superscript>–</superscript> (MP3), and CX 3 CR1<superscript>+</superscript>CCR2<superscript>–</superscript>Ly6C<superscript>–</superscript>MHCII<superscript>+</superscript> (MP4). MP1 cluster has different derivations (yolk sac, fetal liver, and bone marrow) and multiple functions population. Embryonic and neonatal-derived-MP1 directly promoted cardiomyocyte proliferation through Jagged-1–Notch1 axis and significantly ameliorated cardiac injury following myocardial infarction. MP2/3 subsets could survive throughout adulthood. MP4, the main population in adult mouse hearts, contributed to inflammation. During ontogeny, MP1 can convert into MP4 triggered by changes in the cellular redox state. These findings delineate the evolutionary dynamics of CRMs under physiological conditions and found direct evidence that embryonic and neonatal-derived CRMs regulate cardiomyocyte proliferation. Our findings also shed light on cardiac repair following injury. The embryonic and neonatal-derived cardiac-resident macrophages (CRMs) facilitated cardiomyocyte proliferation through Jagged-1–Notch1 axis. After birth, microenvironment changes caused switch or renewal of CRMs, which resulted in the loss of their proliferative potential. [Display omitted] [ABSTRACT FROM AUTHOR]

Details

Language :
English
ISSN :
22113835
Volume :
13
Issue :
1
Database :
Supplemental Index
Journal :
Acta Pharmaceutica Sinica B
Publication Type :
Academic Journal
Accession number :
161693610
Full Text :
https://doi.org/10.1016/j.apsb.2022.08.016