Inhibition of autophagy enhances adenosine‑induced apoptosis in human hepatoblastoma HepG2 cells

Oncol Rep. 2019 Feb;41(2):829-838. doi: 10.3892/or.2018.6899. Epub 2018 Nov 30.

Abstract

In cancer research, autophagy acts as a double‑edged sword: it increases cell viability or induces cell apoptosis depending upon the cell context and functional status. Recent studies have shown that adenosine (Ado) has cytotoxic effects in many tumors. However, the role of autophagy in Ado‑induced apoptosis is still poorly understood. In the present study, Ado‑induced apoptotic death and autophagy in hepatoblastoma HepG2 cells was investigated and the relationship between autophagy and apoptosis was identified. In the present study, it was demonstrated that Ado inhibited HepG2 cell growth in a time‑ and concentration‑dependent manner and activated endoplasmic reticulum (ER) stress, as indicated by G0/G1 cell cycle arrest, the increased mRNA and protein levels of GRP78/BiP, PERK, ATF4, CHOP, cleaved caspase‑3, cytochrome c and the loss of mitochon-drial membrane potential (ΔΨm). Ado also induced autophagic flux, revealed by the increased expression of the autophagy marker microtubule‑associated protein 1 light chain 3‑II (LC3‑II), Beclin‑1, autophagosomes, and the degradation of p62, as revealed by western blot analysis and macrophage‑derived chemokine (MDC) staining. Blocking autophagy using LY294002 notably entrenched Ado‑induced growth inhibition and cell apoptosis, as demonstrated with the increased expression of cytochrome c and p62, and the decreased expression of LC3‑II. Conversely, the autophagy inducer rapamycin alleviated Ado‑induced apoptosis and markedly increased the ΔΨm. Moreover, knockdown of AMPK with si‑AMPK partially abolished Ado‑induced ULK1 activation and mTOR inhibition, and thus reinforced CHOP expression and Ado‑induced apoptosis. These results indicated that Ado‑induced ER stress resulted in apoptosis and autophagy concurrently. The AMPK/mTOR/ULK1 signaling pathway played a protective role in the apoptotic procession. Inhibition of autophagy may effectively enhance the anticancer effect of Ado in human hepatoblastoma HepG2 cells.

MeSH terms

  • AMP-Activated Protein Kinases / genetics
  • AMP-Activated Protein Kinases / metabolism
  • Adenosine / pharmacology*
  • Adenosine / therapeutic use
  • Apoptosis / drug effects*
  • Autophagy / drug effects*
  • Autophagy-Related Protein-1 Homolog / metabolism
  • Cell Survival / drug effects
  • Chromones / pharmacology
  • Dose-Response Relationship, Drug
  • Endoplasmic Reticulum Chaperone BiP
  • Endoplasmic Reticulum Stress / drug effects
  • G1 Phase Cell Cycle Checkpoints / drug effects
  • Gene Knockdown Techniques
  • Hep G2 Cells
  • Hepatoblastoma / drug therapy*
  • Hepatoblastoma / pathology
  • Humans
  • Intracellular Signaling Peptides and Proteins / metabolism
  • Liver Neoplasms / drug therapy*
  • Liver Neoplasms / pathology
  • Morpholines / pharmacology
  • RNA, Small Interfering / metabolism
  • Signal Transduction / drug effects
  • Signal Transduction / genetics
  • Sirolimus / pharmacology
  • TOR Serine-Threonine Kinases / metabolism

Substances

  • Chromones
  • Endoplasmic Reticulum Chaperone BiP
  • HSPA5 protein, human
  • Intracellular Signaling Peptides and Proteins
  • Morpholines
  • RNA, Small Interfering
  • 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one
  • MTOR protein, human
  • Autophagy-Related Protein-1 Homolog
  • TOR Serine-Threonine Kinases
  • ULK1 protein, human
  • AMP-Activated Protein Kinases
  • Adenosine
  • Sirolimus