The proto-oncogene tyrosine protein kinase Src is essential for macrophage-myofibroblast transition during renal scarring

Src activation has been associated with fibrogenesis after kidney injury. Macrophage-myofibroblast transition is a newly identified process to generate collagen-producing myofibroblasts locally in the kidney undergoing fibrosis in a TGF-β/Smad3-dependent manner. The potential role of the macrophage-...

Full description

Saved in:
Bibliographic Details
Published inKidney international Vol. 93; no. 1; pp. 173 - 187
Main Authors Tang, Patrick Ming-Kuen, Zhou, Shuang, Li, Chun-Jie, Liao, Jinyue, Xiao, Jun, Wang, Qing-Ming, Lian, Guang-Yu, Li, Jinhong, Huang, Xiao-Ru, To, Ka-Fai, NG, Chi-Fai, Chong, Charing Ching-Ning, Ma, Ronald Ching-Wa, Lee, Tin-Lap, Lan, Hui-Yao
Format Journal Article
LanguageEnglish
Published United States Elsevier Inc 01.01.2018
Subjects
Online AccessGet full text

Cover

Loading…
More Information
Summary:Src activation has been associated with fibrogenesis after kidney injury. Macrophage-myofibroblast transition is a newly identified process to generate collagen-producing myofibroblasts locally in the kidney undergoing fibrosis in a TGF-β/Smad3-dependent manner. The potential role of the macrophage-myofibroblast transition in Src-mediated renal fibrosis is unknown. In studying this by RNA sequencing at single-cell resolution, we uncovered a unique Src-centric regulatory gene network as a key underlying mechanism of macrophage-myofibroblast transition. A total of 501 differentially expressed genes associated with macrophage-myofibroblast transition were identified. However, Smad3-knockout largely reduced the transcriptome diversity. More importantly, inhibition of Src largely suppresses ureteral obstruction-induced macrophage-myofibroblast transition in the injured kidney in vivo along with transforming growth factor-β1-induced elongated fibroblast-like morphology, α-smooth muscle actin expression and collagen production in bone marrow derived macrophages in vitro. Unexpectedly, we further uncovered that Src serves as a direct Smad3 target gene and also specifically up-regulated in macrophages during macrophage-myofibroblast transition. Thus, macrophage-myofibroblast transition contributes to Src-mediated tissue fibrosis. Hence, targeting Src may represent as a precision therapeutic strategy for macrophage-myofibroblast transition-driven fibrotic diseases.
Bibliography:ObjectType-Article-1
SourceType-Scholarly Journals-1
ObjectType-Feature-2
content type line 23
ISSN:0085-2538
1523-1755
DOI:10.1016/j.kint.2017.07.026