Cell Death and Disease (Nov 2023)

Pyrroloquinoline quinone inhibits PCSK9-NLRP3 mediated pyroptosis of Leydig cells in obese mice

  • Jinyuan Wang,
  • Shun Zhang,
  • Linlin Hu,
  • Yan Wang,
  • Ke Liu,
  • Jianghua Le,
  • Yongpeng Tan,
  • Tianlong Li,
  • Haoxuan Xue,
  • Yanhong Wei,
  • Ou Zhong,
  • Junhui He,
  • Dan Zi,
  • Xin Lei,
  • Renhe Deng,
  • Yafei Luo,
  • Masong Tang,
  • Mingxuan Su,
  • Yichang Cao,
  • Qingyou Liu,
  • Zhihan Tang,
  • Xiaocan Lei

DOI
https://doi.org/10.1038/s41419-023-06162-8
Journal volume & issue
Vol. 14, no. 11
pp. 1 – 19

Abstract

Read online

Abstract Abnormal lipid metabolism and chronic low-grade inflammation are the main traits of obesity. Especially, the molecular mechanism of concomitant deficiency in steroidogenesis-associated enzymes related to testosterone (T) synthesis of obesity dominated a decline in male fertility is still poorly understood. Here, we found that in vivo, supplementation of pyrroloquinoline quinone (PQQ) efficaciously ameliorated the abnormal lipid metabolism and testicular spermatogenic function from high-fat-diet (HFD)-induced obese mice. Moreover, the transcriptome analysis of the liver and testicular showed that PQQ supplementation not only inhibited the high expression of proprotein convertase subtilisin/Kexin type 9 (PCSK9) but also weakened the NOD-like receptor family pyrin domain containing 3 (NLRP3)-mediated pyroptosis, which both played a negative role in T synthesis of Leydig Cells (LCs). Eventually, the function and the pyroptosis of LCs cultured with palmitic acid in vitro were simultaneously benefited by suppressing the expression of NLRP3 or PCSK9 respectively, as well the parallel effects of PQQ were affirmed. Collectively, our data revealed that PQQ supplementation is a feasible approach to protect T synthesis from PCSK9-NLRP3 crosstalk-induced LCs’ pyroptosis in obese men.