Abstract

BackgroundBesides its neurotransmitter and vasoconstriction functions, serotonin is an important mediator of numerous biological processes in peripheral tissues including cell proliferation, steatosis, and fibrogenesis. Recent reports indicate that serotonin may promote tumor growth in liver cancer, however, the molecular mechanisms remain elusive. n this study, we investigated the role and molecular signaling mechanisms mediated by serotonin in liver cancer cell survival, drug resistance, and steatosis.MethodsEffect of serotonin on modulation of cell survival/proliferation was determined by MTT/WST1 assay. Effect of serotonin on the regulation of autophagy biomarkers and lipid/fatty acid proteins expression, AKT/mTOR and Notch signaling was evaluated by immunoblotting. The role of serotonin in normal human hepatocytes and liver cancer cell steatosis was analyzed by Oil Red O staining. The mRNA expression levels of lipid/fatty acid proteins and serotonin receptors were validated by qRT-PCR. The important roles of autophagy, Notch signaling, serotonin receptors and serotonin re-uptake proteins on serotonin-mediated cell steatosis were investigated by using selective inhibitors or antagonists. The association of peripheral serotonin, autophagy, and hepatic steatosis was also investigated using chronic EtOH fed mouse model.ResultsExposure of liver cancer cells to serotonin induced Notch signaling and autophagy, independent of AKT/mTOR pathway. Also, serotonin enhanced cancer cell proliferation/survival and drug resistance. Furthermore, serotonin treatment up-regulated the expression of lipogenic proteins and increased steatosis in liver cancer cells. Inhibition of autophagy or Notch signaling reduced serotonin-mediated cell steatosis. Treatment with serotonin receptor antagonists 5-HTr1B and 5-HTr2B reduced serotonin-mediated cell steatosis; in contrast, treatment with selective serotonin reuptake inhibitors (SSRIs) increased steatosis. In addition, mice fed with chronic EtOH resulted in increased serum serotonin levels which were associated with the induction of hepatic steatosis and autophagy.ConclusionsSerotonin regulates liver cancer cell steatosis, cells survival, and may promote liver carcinogenesis by activation of Notch signaling and autophagy.

Highlights

  • Besides its neurotransmitter and vasoconstriction functions, serotonin is an important mediator of numerous biological processes in peripheral tissues including cell proliferation, steatosis, and fibrogenesis

  • We demonstrate that mice fed with chronic EtOH had increased hepatic steatosis, which was associated with increased serum serotonin levels, suggesting that peripheral serotonin modulates alcohol induced liver cell steatosis in mice

  • Serotonin induces notch signaling in liver cancer cells we examined the role of serotonin in the regulation of oncogenic Notch signaling, which is constitutively active in hepatocellular carcinoma (HCC) [28,29,30]

Read more

Summary

Introduction

Besides its neurotransmitter and vasoconstriction functions, serotonin is an important mediator of numerous biological processes in peripheral tissues including cell proliferation, steatosis, and fibrogenesis. Recent reports indicate that serotonin may promote tumor growth in liver cancer, the molecular mechanisms remain elusive. N this study, we investigated the role and molecular signaling mechanisms mediated by serotonin in liver cancer cell survival, drug resistance, and steatosis. Peripheral serotonin is unable to cross the Blood Brain Barrier (BBB), and each pool of serotonin plays unique biological functions in the brain and in the peripheral organs [2]. Serotonin treatment disturbs Axin1/β-catenin interaction, activates Wnt/β-catenin signaling leads to increased downstream genes expression such as axin 2, cyclin 1, dickoppf-1 (DDK1) and glutamine synthetase, and promotes liver cancer cell proliferation [13]. Treatment of serum-deprived liver Huh cells with serotonin promotes cell proliferation by upregulation of Forkhead box O3a (FOXO3a) transcription factor, and by increasing phosphorylation of AKT and FOXO3a, this mechanism is not found in serum-deprived HepG2 and Hep3B cells [16]

Methods
Results
Discussion
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call