Abstract

Simple SummaryEndometriosis is a common gynecological condition that causes pelvic pain and infertility. Despite having normal histological features, several cells bear cancer-associated somatic mutations that result in local tissue invasion but rarely metastasize. Several cancer-associated genes, such as KRAS and PIK3CA, are frequently mutated in the endometriotic epithelium. However, the functional behavior and molecular pathogenesis of this disorder remain unclear. In this study, we developed an immortalized endometriotic epithelial cell line with mutations in KRAS and PIK3CA, which are genes associated with aggressive behaviors, such as increased cell migration, invasion, and proliferation. Through microarray analysis, the KRAS- and PIK3CA-specific gene signatures were identified; LOX and PTX3 were found to be responsible for this metastatic behavior. Knockdown of these two genes by siRNA markedly reduced the metastatic ability of the cells. Our findings suggest that inhibition of LOX and PTX3 may be an alternative therapeutic strategy to reduce the incidence of endometriosis.Endometriosis-harboring cancer-associated somatic mutations of PIK3CA and KRAS provides new opportunities for studying the multistep processes responsible for the functional and molecular changes in this disease. We aimed to establish a novel in vitro endometriosis model to clarify the functional behavior and molecular pathogenesis of this disorder. Immortalized HMOsisEC10 human ovarian endometriotic epithelial cell line was used in which KRAS and PIK3CA mutations were introduced. Migration, invasion, proliferation, and microarray analyses were performed using KRAS and PIK3CA mutant cell lines. In vitro assays showed that migration, invasion, and proliferation were significantly increased in KRAS and PIK3CA mutant cell lines, indicating that these mutations played causative roles in the aggressive behavior of endometriosis. Microarray analysis identified a cluster of gene signatures; among them, two significantly upregulated cancer-related genes, lysyl oxidase (LOX) and pentraxin3 (PTX3), were associated with cell proliferation, invasion, and migration capabilities. Furthermore, siRNA knockdown of the two genes markedly reduced the metastatic ability of the cells. These results suggest that endometriosis with KRAS or PIK3CA mutations can significantly enhance cell migration, invasion, and proliferation by upregulating LOX and PTX3. We propose that LOX and PTX3 silencing using small molecules could be an alternative therapeutic regimen for severe endometriosis.

Highlights

  • Endometriosis is a benign inflammatory disease, characterized with the recurrent estrogen-dependent ectopic growth of the endometriotic epithelium and stroma outside the uterus

  • Some studies have suggested that mucinous metaplasia in ovarian endometriosis could be closely linked to the development of borderline mucinous ovarian carcinoma with KRAS mutation, which may eventually progress to invasive carcinoma [14,15]

  • Using microarray analysis, we identified the overexpression of lysyl oxidase (LOX) and PTX3 in KARS and PIK3CA mutant cell lines compared to the HMOsisEC10 cells, which was associated with the aggressive behavior of the benign tumor

Read more

Summary

Introduction

Endometriosis is a benign inflammatory disease, characterized with the recurrent estrogen-dependent ectopic growth of the endometriotic epithelium and stroma outside the uterus. It affects approximately 10–15% of the women of reproductive age [1,2], causing pelvic pain and infertility [2,3]. Endometriosis has three major manifestations in the pelvic region: superficial, ovarian, and deep infiltrating [8,9] It was previously considered a benign condition with normal-acting histological features, recent studies have established that endometriosis can recapitulate some cancer-like features, such as cell proliferation, migration, and invasion. No behavioral and functional analysis of the aforementioned genes in endometriosis has been performed

Objectives
Methods
Results
Discussion
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