Overexpression of CARMA3 is associated with advanced tumor stage, cell cycle progression, and cisplatin resistance in human epithelial ovarian cancer

Tumor Biology ◽  
2014 ◽  
Vol 35 (8) ◽  
pp. 7957-7964 ◽  
Author(s):  
Chengyao Xie ◽  
Yong Han ◽  
Lin Fu ◽  
Qingchang Li ◽  
Xueshan Qiu ◽  
...  
2020 ◽  
Author(s):  
Lan Dai ◽  
Keqi Song ◽  
Wenjing Wang ◽  
Yixuan Liu ◽  
Wen Di

Abstract Background: Epithelial ovarian cancer (EOC) is the leading cause of death from female cancers. In our previous study, Sphingosine kinase 2 (SphK2) inhibitor was shown to display anti-EOC activities. The purpose of this study was to further evaluate the expression characteristics and clinical significance of SphK2 in EOC, and to explore the roles and underlying mechanisms of SphK2 in EOC cell survival.Methods: SphK2 expression was examined by Immunohistochemistry and western blot, and its clinical implications and prognostic significance were analyzed. Cellular proliferation assay and mouse xenograft model was established to confirm the roles of SphK2 in vitro and in vivo. Cell cycle analysis, apoptosis assay and western blot were performed to examine cell cycle progression and apoptosis rate. Gene set enrichment analysis (GSEA) and western blot was used to investigate the downstream signaling pathways related to SphK2 function.Results: SphK2 expression level was shown to be associated with stage, histological grade, lymph node metastasis and ascite status. More importantly, high SphK2 expression level was a prognostic indicator of overall survival and relapse-free survival. Moreover, knockdown of SphK2 arrested cell cycle progression and inhibited the proliferation of EOC cells both in vitro and in vivo. Furthermore, ERK/c-Myc, the key pathway in EOC progression, was important for SphK2-mediated mitogenic action in EOC cells.Conclusion: Our findings provided the first evidence that SphK2 played a crucial role in EOC proliferation by regulating ERK/c-Myc pathway. SphK2 may serve as a prognostic marker and potential therapeutic target in EOC.


Oncotarget ◽  
2015 ◽  
Vol 6 (29) ◽  
pp. 27613-27627 ◽  
Author(s):  
Carter J. Barger ◽  
Wa Zhang ◽  
Joanna Hillman ◽  
Aimee B. Stablewski ◽  
Michael J. Higgins ◽  
...  

2013 ◽  
Vol 31 (15_suppl) ◽  
pp. e13563-e13563
Author(s):  
Dennis C. DeSimone ◽  
Trung T. Nguyen ◽  
Eugen Brailiou ◽  
John C. Taylor ◽  
Gabriela Cristina Brailoiu ◽  
...  

e13563 Background: Most ovarian cancer patients are treated with platinum-based chemotherapy but eventually relapse with incurable disease. The G protein-coupled estrogen receptor GPER (GPR30) mediates Ca2+ mobilization in response to estrogen and G-1, a synthetic agonist. Large and sustained Ca2+ responses can lead to mitochondrial Ca2+ overload and apoptosis. Hence, we evaluated whether G-1 could induce apoptosis in cisplatin-sensitive A2780 and isogenic cisplatin–resistant CP70 (14-fold resistant), C30 (70-fold resistant) and C200 (157-fold resistant) human ovarian cancer cells. Bcl-2 and Bcl-xL protect mitochondria from Ca2+overload, and were overexpressed in these cisplatin-resistant cells; thus we also examined combining the Bcl-2 family inhibitor navitoclax with G-1. Methods: Cytoplasmic [Ca2+]c and mitochondrial [Ca2+]m were monitored using microscopy and fluorescent Ca2+ probes. Cell cycle, apoptosis and mitochondrial membrane potential (MMP) were assessed by flow cytometry of propidium iodide, Annexin V and DiIC1(5) -stained cells. The intracellular Ca2+ chelator BAPTA was used to block Ca2+mobilization. Results: Expression of the 53kDa GPER but not the 38 kDa isoform progressively increased with increasing cisplatin resistance. G-1 elicited sustained [Ca2+]c rises that correlated with 53 kDa GPER expression, followed by rises in [Ca2+]m. In all cells, 2.5 μM G-1 blocked cell cycle progression at G2/M, inhibited proliferation, and induced apoptosis (A2780 > C30 > CP70 ≥ C200). G-1 induced p53, caspase-3 and PARP cleavage, and MMP loss. BAPTA prevented G-1’s cell cycle and apoptotic effects in cells showing large Ca2+ mobilization responses but did not in cells with small Ca2+responses. Combining navitoclax with G-1 superadditively decreased cell viability and increased apoptosis. Conclusions: G-1 blocked cell cycle progression and induced apoptosis via a Ca2+-dependent pathway in cells expressing high 53 kDa GPER levels, but via a Ca2+-independent pathway in cells with low 53 kDa GPER expression. G-1 also interacted cooperatively with naviticlax. Therefore, G-1 plus navitoclax shows potential for therapeutic use in platinum-sensitive and -resistant ovarian cancer.


Sign in / Sign up

Export Citation Format

Share Document