HOMEPRODUCTSCOMPANYCONTACTFAQResearchDictionaryPharmaSign Up FREE or Login

Azithromycin enhances anticancer activity of TRAIL by inhibiting autophagy and up-regulating the protein levels of DR4/5 in colon cancer cells in vitro and in vivo.

AbstractBACKGROUND:
Azithromycin is a member of macrolide antibiotics, and has been reported to inhibit the proliferation of cancer cells. However, the underlying mechanisms are not been fully elucidated. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) selectively targets tumor cells without damaging healthy cells. In the present study, we examined whether azithromycin is synergistic with TRAIL, and if so, the underlying mechanisms in colon cancers.
METHODS:
HCT-116, SW480, SW620 and DiFi cells were treated with azithromycin, purified TRAIL, or their combination. A sulforhoddamine B assay was used to examine cell survival. Apoptosis was examined using annexin V-FITC/PI staining, and autophagy was observed by acridine orange staining. Western blot analysis was used to detect protein expression levels. In mechanistic experiments, siRNAs were used to knockdown death receptors (DR4, DR5) and LC-3B. The anticancer effect of azithromycin and TRAIL was also examined in BALB/c nude mice carrying HCT-116 xenografts.
RESULTS:
Azithromycin decreased the proliferation of HCT-116 and SW480 cells in a dose-dependent manner. Combination of azithromycin and TRAIL inhibited tumor growth in a manner that could not be explained by additive effects. Azithromycin increased the expressions of DR4, DR5, p62 and LC-3B proteins and potentiated induction of apoptosis by TRAIL. Knockdown of DR4 and DR5 with siRNAs increased cell survival rate and decreased the expression of cleaved-PARP induced by the combination of azithromycin and TRAIL. LC-3B siRNA and CQ potentiated the anti-proliferation activity of TRAIL alone, and increased the expressions of DR4 and DR5.
CONCLUSION:
The synergistic antitumor effect of azithromycin and TRAIL mainly relies on the up-regulations of DR4 and DR5, which in turn result from LC-3B-involved autophagy inhibition.
AuthorsXinran Qiao, Xiaofei Wang, Yue Shang, Yi Li, Shu-Zhen Chen
JournalCancer communications (London, England) (Cancer Commun (Lond)) Vol. 38 Issue 1 Pg. 43 (07 03 2018) ISSN: 2523-3548 [Electronic] United States
PMID29970185 (Publication Type: Journal Article, Research Support, Non-U.S. Gov't)
Chemical References
  • Anti-Bacterial Agents
  • MAP1LC3B protein, human
  • Microtubule-Associated Proteins
  • Receptors, TNF-Related Apoptosis-Inducing Ligand
  • TNF-Related Apoptosis-Inducing Ligand
  • Azithromycin
Topics
  • Animals
  • Anti-Bacterial Agents (pharmacology)
  • Autophagy (drug effects, genetics)
  • Azithromycin (pharmacology)
  • Cell Line, Tumor
  • Colonic Neoplasms (drug therapy, metabolism, pathology)
  • Drug Synergism
  • HCT116 Cells
  • Humans
  • Mice, Inbred BALB C
  • Mice, Nude
  • Microtubule-Associated Proteins (genetics, metabolism)
  • RNA Interference
  • Receptors, TNF-Related Apoptosis-Inducing Ligand (genetics, metabolism)
  • TNF-Related Apoptosis-Inducing Ligand (pharmacology)
  • Up-Regulation (drug effects)
  • Xenograft Model Antitumor Assays

Join CureHunter, for free Research Interface BASIC access!

Take advantage of free CureHunter research engine access to explore the best drug and treatment options for any disease. Find out why thousands of doctors, pharma researchers and patient activists around the world use CureHunter every day.
Realize the full power of the drug-disease research graph!


Choose Username:
Email:
Password:
Verify Password:
Enter Code Shown: