1. Academic Validation
  2. CACNA1H downregulation induces skeletal muscle atrophy involving endoplasmic reticulum stress activation and autophagy flux blockade

CACNA1H downregulation induces skeletal muscle atrophy involving endoplasmic reticulum stress activation and autophagy flux blockade

  • Cell Death Dis. 2020 Apr 24;11(4):279. doi: 10.1038/s41419-020-2484-2.
Suting Li  # 1 Menglei Hao  # 1 Bingshu Li  # 1 Mao Chen 1 Jue Chen 1 Jianming Tang 1 Shasha Hong 1 Jie Min 1 Ming Hu 1 Li Hong 2
Affiliations

Affiliations

  • 1 Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People's Republic of China.
  • 2 Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People's Republic of China. [email protected].
  • # Contributed equally.
Abstract

Multiple vaginal delivery (MVD) is an important factor for pelvic floor muscle (PFM) function decline and pelvic floor dysfunction (PFD). PFD is common in middle-aged and elderly women, but its pathogenesis is not clear. In this study, we found that the expression of CACNA1H was lower in the PFM of old mice after MVD compared with old or adult mice. In in-vitro studies, we found that treatment with the T-type Ca2+ channel (T-channel) inhibitor NNC-55 or downregulation of the CACNA1H gene by siRNA intervention promoted myotube atrophy and Apoptosis. Mechanistically, we revealed that NNC-55 increased the expression of GRP78 and DDIT3 in myotubes, indicating endoplasmic reticulum stress (ERS) activation, and that the IRE1 and PERK pathways might be involved in this effect. NNC-55 induced the formation of autophagosomes but inhibited Autophagy flux. Moreover, rapamycin, an Autophagy activator, did not rescue myotube atrophy or Apoptosis induced by NNC-55, and the Autophagy inhibitors 3-MA and HCQ accelerated this damage. Further studies showed that the ERS inhibitors 4-PBA and TUDAC relieved NNC-55-induced damage and Autophagy flux blockade. Finally, we found multisite muscle atrophy and decreased muscle function in Cacna1h-/- (TH-null) mice, as well as increased Autophagy inhibition and apoptotic signals in the PFM of old WT mice after MVD and TH-null mice. Taken together, our results suggest that MVD-associated PFD is partially attributed to CACNA1H downregulation-induced PFM atrophy and that ERS is a potential therapeutic target for this disease.

Figures
Products