Your browser doesn't support javascript.
loading
Tumor growth inhibitory effect of juglone and its radiation sensitizing potential: in vivo and in vitro studies.
Aithal, Kiran B; Kumar, Sunil; Rao, B Nageshwar; Udupa, Nayanabhirama; Rao, Satish Bola Sadashiva.
Affiliation
  • Aithal KB; Manipal University, Manipal, Karnataka, India.
Integr Cancer Ther ; 11(1): 68-80, 2012 Mar.
Article in En | MEDLINE | ID: mdl-21498474
ABSTRACT
The present study aimed at evaluating the anticancer and radiosensitizing potential of juglone against a chemoresistant and radioresistant tumor (B16F1 melanoma) growing on C57BL/6J mice. Volume doubling time, growth delay, and median survival were used to assess the in vivo anticancer and radiosensitizing potential of juglone. In vitro radiosensitizing potential of juglone was studied using clonogenic, comet, and reactive oxygen species induction assays. Treatment of tumor-bearing mice with sublethal doses of juglone caused a dose-dependent inhibition of tumor growth as evident from the growth delay and median survival values. Comet assay using tumor tissue and blood showed differential toxicity of juglone, where higher levels of DNA damage was seen in tumor tissue compared with blood cells. Pretreatment of tumor-bearing mice with optimum dose of juglone before radiation resulted in significant tumor growth inhibition compared with radiation alone. From the clonogenic assay, the authors observed a sensitization enhancement ratio of 1.37 for the combination treatment compared with radiation alone. Furthermore, comet assay studies revealed the potential of juglone to enhance the radiation-induced DNA damage and cause a delay in its repair. Juglone pretreatment before radiation also resulted in a significant elevation in the intracellular reactive oxygen species levels compared with radiation alone. In conclusion, the results of this study show the potential of juglone to inhibit the growth of melanoma in vivo. The study also revealed the potential of juglone to augment the radiation-induced cell death of melanoma cells, which may be attributed to oxidative stress-mediated DNA damage and its delayed repair.
Subject(s)

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Radiation-Sensitizing Agents / DNA Damage / Naphthoquinones / Apoptosis / Antineoplastic Agents Limits: Animals Language: En Journal: Integr Cancer Ther Year: 2012 Document type: Article

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Radiation-Sensitizing Agents / DNA Damage / Naphthoquinones / Apoptosis / Antineoplastic Agents Limits: Animals Language: En Journal: Integr Cancer Ther Year: 2012 Document type: Article