Abstract

The etiology of granulosa cell tumors (GCTs) is largely unknown. The primary mode of treatment is surgical, however not all women are cured by surgery alone. Thus, it is important to develop improved treatments through a greater understanding of the molecular mechanisms that contribute to this disease. Recently, it has been shown that a FOXL2 402C>G (C134W) mutation is present in 97% of human adult-type GCTs, suggesting an important role for this mutation in the development of GCTs. We have shown previously that gonadotropin-releasing hormone (GnRH)-I and -II induce apoptosis in cultured normal human granulosa cells. Moreover, it has been reported that FOXL2 can bind to the promoter of the mouse GnRH receptor gene and regulate its transcription. Thus, we hypothesized that C134W mutant FOXL2 could modulate the pro-apoptotic effects of GnRH via aberrant regulation of GnRH receptor levels. Using KGN cells, a human GCT-derived cell line which harbors the FOXL2 402C>G mutation, we show that treatment with GnRH-I and -II induces cell apoptosis, and that small interfering RNA-mediated depletion of GnRH receptor abolishes these effects. Overexpression of wild-type FOXL2 increases both mRNA and protein levels of GnRH receptor and consequently enhances GnRH-induced apoptosis. Importantly, neither the expression levels of GnRH receptor nor GnRH-induced apoptosis were affected by overexpression of the C134W mutant FOXL2. Interestingly, knockdown of endogenous FOXL2 down-regulates GnRHR expression in normal human granulosa cells with wild-type FOXL2, but not in KGN cells. These results suggest that the FOXL2 402C>G mutation may contribute to the development of human adult-type GCTs by reducing the expression of GnRH receptor, thus conferring resistance to GnRH-induced cell apoptosis.

Highlights

  • Ovarian cancer is the most lethal gynecological malignancy in the Western world

  • Overexpression wild-type FOXL2, but not C134W mutant FOXL2, increases GnRHR expression and enhances GnRHinduced cell apoptosis. These results suggest that the 402C.G FOXL2 gene mutation may contribute to human granulosa cell tumorigenesis by diminishing Gonadotropin-releasing hormone (GnRH)/GnRHR-induced cell apoptosis

  • Two human ovarian cancer cell lines, CaOV3 and OVCAR3, were used as positive controls because we have previously demonstrated that they express GnRHR [19]

Read more

Summary

Introduction

Granulosa cell tumors (GCTs) account for approximately 5% of all ovarian cancers and occur at a frequency of 1 in 100,000. GCTs are a poorly understood cancer whose pathogenesis is unknown and for which there is no effective treatment beyond primary surgery. We have previously demonstrated that GnRHR and both forms of GnRH are expressed in human granulosa-luteal cells [3,4]. These data suggest putative regulatory roles for GnRHs in the development and function of ovarian follicles and/or corpus luteum. Whether GnRH can trigger apoptosis in human GCTs is still unknown

Objectives
Methods
Results
Conclusion
Full Text
Paper version not known

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

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.