CD116(+) fetal precursors migrate to the perinatal lung and give rise to human alveolar macrophages

JOURNAL OF EXPERIMENTAL MEDICINE(2022)

引用 22|浏览14
暂无评分
摘要
Evren et al. identify CD116(+) fetal liver cells as precursors of human alveolar macrophages in early life and determine the impact of cell origin on lung macrophage identity and function in the human context with a unique in vivo model. Despite their importance in lung health and disease, it remains unknown how human alveolar macrophages develop early in life. Here we define the ontogeny of human alveolar macrophages from embryonic progenitors in vivo, using a humanized mouse model expressing human cytokines (MISTRG mice). We identified alveolar macrophage progenitors in human fetal liver that expressed the GM-CSF receptor CD116 and the transcription factor MYB. Transplantation experiments in MISTRG mice established a precursor-product relationship between CD34(-)CD116(+) fetal liver cells and human alveolar macrophages in vivo. Moreover, we discovered circulating CD116(+)CD64(-)CD115(+) macrophage precursors that migrated from the liver to the lung. Similar precursors were present in human fetal lung and expressed the chemokine receptor CX3CR1. Fetal CD116(+)CD64(-) macrophage precursors had a proliferative gene signature, outcompeted adult precursors in occupying the perinatal alveolar niche, and developed into functional alveolar macrophages. The discovery of the fetal alveolar macrophage progenitor advances our understanding of human macrophage origin and ontogeny.
更多
查看译文
AI 理解论文
溯源树
样例
生成溯源树,研究论文发展脉络
Chat Paper
正在生成论文摘要