Chronic stress promotes glioma cell proliferation via the PI3K/Akt signaling pathway

Oncol Rep. 2021 Sep;46(3):202. doi: 10.3892/or.2021.8153. Epub 2021 Jul 23.

Abstract

High malignancy and high mortality of glioma render it urgent to elucidate the underlying mechanisms of glioma carcinogenesis and explore novel targets for therapy. Epidemiologic and clinical studies have revealed that chronic stress promotes the progression of various solid tumors and is correlated with poor prognosis; however, findings reporting the involvement of chronic stress in glioma are rare. In the present study, a chronic restraint animal model and a chronic stress cell model were established to explore the effects of chronic stress on glioma and its molecular mechanisms. The results revealed that chronic stress promoted glioma growth in vivo, and the serum levels of the stress hormones glucocorticoid (GC) and noradrenaline (NE) were significantly increased. In addition, GC and NE were verified to accelerate the proliferation of glioma cells in vitro. Mechanistically, the phosphatidylinositol 3‑kinase (PI3K)/Akt signaling pathway was revealed to be activated under stress conditions, and inhibition of the expression of p‑Akt could restrain the stress hormone‑induced glioma cell proliferation. In addition, our data indicated that the GC receptor (GR) and β‑adrenergic receptors (ADRBs) were both required for the biological functions of GC and NE in glioma cells. In conclusion, these results indicated that chronic stress and the stress hormones GC and NE activated PI3K/Akt signaling through binding to GR and ADRBs, thereby promoting glioma cell growth. Our findings may provide potential therapeutic targets and pave the way for the development of new strategies to protect patients with glioma from the detrimental effects of stress on tumor progression.

Keywords: Akt; GC; NE; cell proliferation; chronic stress; glioma.

MeSH terms

  • Animals
  • Apoptosis / drug effects
  • Brain Neoplasms / metabolism*
  • Cell Line, Tumor
  • Cell Proliferation
  • Disease Progression
  • Gene Expression Regulation, Neoplastic*
  • Glioma / metabolism*
  • Glucocorticoids / metabolism
  • Hormones / metabolism
  • Humans
  • Male
  • Mice
  • Mice, Inbred BALB C
  • Mice, Nude
  • MicroRNAs / metabolism
  • Norepinephrine / metabolism
  • Phosphatidylinositol 3-Kinase / metabolism
  • Phosphatidylinositol 3-Kinases / metabolism*
  • Prognosis
  • Proto-Oncogene Proteins c-akt / metabolism*
  • RNA, Small Interfering / metabolism
  • Signal Transduction
  • Stress, Physiological

Substances

  • Glucocorticoids
  • Hormones
  • MicroRNAs
  • RNA, Small Interfering
  • Phosphatidylinositol 3-Kinase
  • AKT1 protein, human
  • Proto-Oncogene Proteins c-akt
  • Norepinephrine

Grants and funding

The present study was supported (grant nos. 81702454, 31771290 and 31571173) by the National Natural Science Foundation of China.