Abstract

Clinical attempts to reduce the cardiotoxicity of arsenic trioxide (ATO) without compromising its anticancer activities remain to be an unresolved issue. In this study, we determined whether Sal B can protect against ATO-induced cardiac toxicity in vivo and increase the toxicity of ATO toward cancer cells. Combination treatment of Sal B and ATO was investigated using BALB/c mice and human hepatoma (HepG2) cells and human cervical cancer (HeLa) cells. The results showed that the combination treatment significantly improved the ATO-induced loss of cardiac function, attenuated damage of cardiomyocytic structure, and suppressed the ATO-induced release of cardiac enzymes into serum in BALB/c mouse models. The expression levels of Bcl-2 and p-Akt in the mice treated with ATO alone were reduced, whereas those in the mice given the combination treatment were similar to those in the control mice. Moreover, the combination treatment significantly enhanced the ATO-induced cytotoxicity and apoptosis of HepG2 cells and HeLa cells. Increases in apoptotic marker cleaved poly (ADP-ribose) polymerase and decreases in procaspase-3 expressions were observed through western blot. Taken together, these observations indicate that the combination treatment of Sal B and ATO is potentially applicable for treating cancer with reduced cardiotoxic side effects.

Highlights

  • Arsenic trioxide (ATO) was discovered more than 2000 years ago for treating diseases such as cancer, syphilis, and tuberculosis [1]

  • The primary antibodies against poly (ADPribose) polymerase (PARP), procaspase-3, Bcl-2, Bax, p-Akt, and Akt were purchased from Santa Cruz Biotechnology (Santa Cruz, CA, USA)

  • We examined the effect of the combination treatment using an in vivo mouse model of ATO-induced cardiotoxicity

Read more

Summary

Introduction

Arsenic trioxide (ATO) was discovered more than 2000 years ago for treating diseases such as cancer, syphilis, and tuberculosis [1]. ATO has been recognized as a clinically effective drug for treating acute promyelocytic leukemia (APL) [2]. This treatment was associated with QT prolongation, torsades de pointes, and sudden death [3,4,5], thereby restricting its broad application. The possible mechanisms of ATO-induced cardiotoxicity include DNA fragmentation, reactive oxygen species (ROS) generation, cardiac ion channel changes, and apoptosis [6,7,8]. The exact mechanism of ATO cardiotoxicity is not currently known, the generation of ROS is very common in arsenic toxicity [8, 9]. Antioxidative agents could provide an alternative approach to treat ATO-induced cardiac damages

Methods
Results
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