Your browser doesn't support javascript.
loading
Muscle-resident mesenchymal progenitors sense and repair peripheral nerve injury via the GDNF-BDNF axis.
Yoo, Kyusang; Jo, Young-Woo; Yoo, Takwon; Hann, Sang-Hyeon; Park, Inkuk; Kim, Yea-Eun; Kim, Ye Lynne; Rhee, Joonwoo; Song, In-Wook; Kim, Ji-Hoon; Baek, Daehyun; Kong, Young-Yun.
Affiliation
  • Yoo K; School of Biological Sciences, Seoul National University, Seoul, Republic of Korea.
  • Jo YW; School of Biological Sciences, Seoul National University, Seoul, Republic of Korea.
  • Yoo T; School of Biological Sciences, Seoul National University, Seoul, Republic of Korea.
  • Hann SH; School of Biological Sciences, Seoul National University, Seoul, Republic of Korea.
  • Park I; School of Biological Sciences, Seoul National University, Seoul, Republic of Korea.
  • Kim YE; School of Biological Sciences, Seoul National University, Seoul, Republic of Korea.
  • Kim YL; School of Biological Sciences, Seoul National University, Seoul, Republic of Korea.
  • Rhee J; School of Biological Sciences, Seoul National University, Seoul, Republic of Korea.
  • Song IW; School of Biological Sciences, Seoul National University, Seoul, Republic of Korea.
  • Kim JH; School of Biological Sciences, Seoul National University, Seoul, Republic of Korea.
  • Baek D; Molecular Recognition Research Center, Korea Institute of Science and Technology, Seoul, Republic of Korea.
  • Kong YY; School of Biological Sciences, Seoul National University, Seoul, Republic of Korea.
Elife ; 132024 Sep 26.
Article in En | MEDLINE | ID: mdl-39324575
ABSTRACT
Fibro-adipogenic progenitors (FAPs) are muscle-resident mesenchymal progenitors that can contribute to muscle tissue homeostasis and regeneration, as well as postnatal maturation and lifelong maintenance of the neuromuscular system. Recently, traumatic injury to the peripheral nerve was shown to activate FAPs, suggesting that FAPs can respond to nerve injury. However, questions of how FAPs can sense the anatomically distant peripheral nerve injury and whether FAPs can directly contribute to nerve regeneration remained unanswered. Here, utilizing single-cell transcriptomics and mouse models, we discovered that a subset of FAPs expressing GDNF receptors Ret and Gfra1 can respond to peripheral nerve injury by sensing GDNF secreted by Schwann cells. Upon GDNF sensing, this subset becomes activated and expresses Bdnf. FAP-specific inactivation of Bdnf (Prrx1Cre; Bdnffl/fl) resulted in delayed nerve regeneration owing to defective remyelination, indicating that GDNF-sensing FAPs play an important role in the remyelination process during peripheral nerve regeneration. In aged mice, significantly reduced Bdnf expression in FAPs was observed upon nerve injury, suggesting the clinical relevance of FAP-derived BDNF in the age-related delays in nerve regeneration. Collectively, our study revealed the previously unidentified role of FAPs in peripheral nerve regeneration, and the molecular mechanism behind FAPs' response to peripheral nerve injury.
Subject(s)
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Brain-Derived Neurotrophic Factor / Glial Cell Line-Derived Neurotrophic Factor / Mesenchymal Stem Cells / Peripheral Nerve Injuries / Nerve Regeneration Limits: Animals Language: En Journal: Elife Year: 2024 Document type: Article Country of publication: United kingdom

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Brain-Derived Neurotrophic Factor / Glial Cell Line-Derived Neurotrophic Factor / Mesenchymal Stem Cells / Peripheral Nerve Injuries / Nerve Regeneration Limits: Animals Language: En Journal: Elife Year: 2024 Document type: Article Country of publication: United kingdom