Abstract

Background:Ovarian cancer is one of the most important gynecological malignancies, causing significant mortality. Recently, there has been extensive attention to the involvement of signaling cascades in its initiation/progression. In this study, we focused on the possible role of Notch signal transduction in proliferation and metalloproteinase 2 and 9 function in human ovarian cancer OVCAR-3 cells.Methods:MTT proliferation assays were used to evaluate effects of a DAPT inhibitor on cell proliferation. For measurement of Hes-1 mRNA levels, quantitative reverse transcription polymerase chain reaction (qRT-PCR) was applied following 48 h incubation with the inhibitor. In addition, metalloproteinase (MMPs) activity was assessed by zymography.Results:Inhibition of Notch signaling resulted in a significant reduction in OVCAR-3 cell proliferation. Additionally, DAPT treatment of cells significantly decreased Hes-1 mRNA levels (p < 0.05) as well as activity of MMP-2 and -9 (p < 0.05).Conclusion:Our results suggested that suppression of Notch signaling by a specific inhibitor can effectively decrease proliferation and the potential for metastasis of OVCAR-3 cells via a reduction in the activity of metalloproteinases 2 and 9. Thus, pharmacological targeting of the Notch signaling pathway could be a promising future treatment for ovarian cancer.

Highlights

  • Ovarian cancer, as one of the most important leading causes of death in gynecological malignancies, is a high heterogeneous and complex neoplasm in histology, biological properties, as well as the cellular origin (Smolle et al, 2013)

  • To approve that the effects of inhibitor are mediated by the inhibition of Notch signaling pathway, we examined the effect of DAPT on Hes-1 mRNA expression in OVCAR-3 cells by quantitative reverse transcription polymerase chain reaction (qRT-PCR), as a key gene of this signaling pathway

  • Accumulating previous studies in the evaluating and identifying the causes and mechanisms involved in the pathogenesis of ovarian cancer to design effective therapies and modern methods for early detection of disease, suggest that variations in genes and important signaling pathways have a function in the pathogenesis of ovarian cancer, as a complex genetic cancer (Kurman and Shih, 2011)

Read more

Summary

Introduction

As one of the most important leading causes of death in gynecological malignancies, is a high heterogeneous and complex neoplasm in histology, biological properties, as well as the cellular origin (Smolle et al, 2013). The Notch ICD translocates to the nucleus, binds to the transcriptional repressor protein C binding factor 1 (CBF1; known as RBP-Jκ) or CSL and converts CBF1 from a transcriptional repressor to an activator and formes CBF1-NICD-MAML1 complex functions as a transcriptional activator, promotes expression of a number of downstream target genes such as Hes and Hey family (Leong and Karsan, 2006; Aval et al, 2017) These target genes are the best-characterized targets of this transcriptional activation complex and suppress the transcription of downstream targets such as cyclindependent kinase inhibitor p27Kip, involved in a broad. Conclusion: Our results suggested that suppression of Notch signaling by a specific inhibitor can effectively decrease proliferation and the potential for metastasis of OVCAR-3 cells via a reduction in the activity of metalloproteinases 2 and 9. Pharmacological targeting of the Notch signaling pathway could be a promising future treatment for ovarian cancer

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