Nature Communications (Nov 2023)

FNIP1 abrogation promotes functional revascularization of ischemic skeletal muscle by driving macrophage recruitment

  • Zongchao Sun,
  • Likun Yang,
  • Abdukahar Kiram,
  • Jing Yang,
  • Zhuangzhuang Yang,
  • Liwei Xiao,
  • Yujing Yin,
  • Jing Liu,
  • Yan Mao,
  • Danxia Zhou,
  • Hao Yu,
  • Zheng Zhou,
  • Dengqiu Xu,
  • Yuhuan Jia,
  • Chenyun Ding,
  • Qiqi Guo,
  • Hongwei Wang,
  • Yan Li,
  • Li Wang,
  • Tingting Fu,
  • Shijun Hu,
  • Zhenji Gan

DOI
https://doi.org/10.1038/s41467-023-42690-9
Journal volume & issue
Vol. 14, no. 1
pp. 1 – 18

Abstract

Read online

Abstract Ischaemia of the heart and limbs attributable to compromised blood supply is a major cause of mortality and morbidity. The mechanisms of functional angiogenesis remain poorly understood, however. Here we show that FNIP1 plays a critical role in controlling skeletal muscle functional angiogenesis, a process pivotal for muscle revascularization during ischemia. Muscle FNIP1 expression is down-regulated by exercise. Genetic overexpression of FNIP1 in myofiber causes limited angiogenesis in mice, whereas its myofiber-specific ablation markedly promotes the formation of functional blood vessels. Interestingly, the increased muscle angiogenesis is independent of AMPK but due to enhanced macrophage recruitment in FNIP1-depleted muscles. Mechanistically, myofiber FNIP1 deficiency induces PGC-1α to activate chemokine gene transcription, thereby driving macrophage recruitment and muscle angiogenesis program. Furthermore, in a mouse hindlimb ischemia model of peripheral artery disease, the loss of myofiber FNIP1 significantly improved the recovery of blood flow. Thus, these results reveal a pivotal role of FNIP1 as a negative regulator of functional angiogenesis in muscle, offering insight into potential therapeutic strategies for ischemic diseases.