1. Academic Validation
  2. Amifostine attenuates bleomycin-induced pulmonary fibrosis in mice through inhibition of the PI3K/Akt/mTOR signaling pathway

Amifostine attenuates bleomycin-induced pulmonary fibrosis in mice through inhibition of the PI3K/Akt/mTOR signaling pathway

  • Sci Rep. 2023 Jun 28;13(1):10485. doi: 10.1038/s41598-023-34060-8.
Wenting Yang # 1 Lin Pan # 1 Yiju Cheng 2 3 Xiao Wu 1 Songsong Huang 4 Juan Du 1 Honglan Zhu 1 Menglin Zhang 1 Yuquan Zhang 1
Affiliations

Affiliations

  • 1 Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China.
  • 2 Department of Respiratory and Critical Care Medicine, The First People's Hospital of Guiyang, Guiyang, 550004, China. [email protected].
  • 3 Guizhou Medical University, Guiyang, 550004, China. [email protected].
  • 4 Department of Pathology, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China.
  • # Contributed equally.
Abstract

Amifostine is a normal cell protection agent, not only used in the adjuvant therapy of lung Cancer, ovarian Cancer, breast Cancer, nasopharyngeal Cancer, bone tumor, digestive tract tumor, blood system tumor and other cancers in order to reduce the toxicity of chemotherapy drugs, and recent studies have reported that the drug can also reduce lung tissue damage in patients with pulmonary fibrosis, but its mechanism of action is not yet fully understood. In this study, we explored the potential therapeutic effects and molecular mechanisms of AMI on bleomycin (BLM)-induced pulmonary fibrosis in mice. A mouse model of pulmonary fibrosis was established using BLM. We then assessed histopathological changes, inflammatory factors, oxidative indicators, Apoptosis, epithelial-mesenchymal transition, extracellular matrix changes, and levels of phosphatidylinositol 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) signaling pathway-related proteins in the BLM-treated mice to determine the effect of AMI treatment on these factors. BLM-treated mice had substantial lung inflammation and abnormal extracellular matrix deposition. Overall, treatment with AMI significantly improved BLM-induced lung injury and pulmonary fibrosis. More specifically, AMI alleviated BLM-induced oxidative stress, inflammation, alveolar cell Apoptosis, epithelial-mesenchymal transition, and extracellular matrix deposition by regulating the PI3K/Akt/mTOR signaling pathway. This finding that AMI can alleviate pulmonary fibrosis in a mouse model by inhibiting activation of the PI3K/Akt/mTOR signaling pathway lays a foundation for potential future clinical application of this agent in patients with pulmonary fibrosis.

Figures
Products