scholarly journals Bioinformatic and Experimental Analysis Reveals Knockdown of KIF15 Promotes Cell Apoptosis via Activating Crosstalk of Multi-Pathways in Ovarian Cancer

2020 ◽  
Author(s):  
Xinwei Sun ◽  
Mengyue Chen ◽  
Bin Liao ◽  
Zhiqing Liang

Abstract Background: Ovarian cancer (OC) is the most lethal malignancy of females worldwide. Unlimited proliferation is a fundamental feature of OC cells. The genes associated with cell proliferation are potential histopathological biomarkers and targets of anti-tumor therapeutic strategies. In the present study, we aimed to identify proliferation-associated biomarkers with potential prognostic, diagnostic, and therapeutic value and reveal the underlying molecular mechanism of the candidate gene involved in OC by a combination of bioinformatic and experimental methods.Results: KIF15 was upregulated in early-stage OC tissues and could predict poor prognosis of patients of Stage I and II. The knockdown of KIF15 significantly inhibited cell proliferation, tumor formation, and growth as well as promoted apoptosis of OC cells. A combination of experimental and bioinformatic analyses revealed KIF15 knockdown promoted cell apoptosis via activating crosstalk of multi-pathways in OC.Conclusion: In this study, KIF15, an early-stage prognostic gene, was identified as a potential histopathological biomarker and therapeutic target of OC.

2021 ◽  
Vol 14 (1) ◽  
Author(s):  
Jun Zhu ◽  
Jun-e Luo ◽  
Yurong Chen ◽  
Qiong Wu

Abstract Background Ovarian cancer is an aggressive tumor in women with high mortality. Paclitaxel (PTX) can be used for the chemotherapy of ovarian cancer. Here, the roles of circular_0061140 (circ_0061140) in PTX sensitivity and malignant progression of ovarian cancer are unveiled. Methods The expressions of circ_0061140, microRNA-136 (miR-136) and chromobox 2 (CBX2) mRNA were detected by quantitative real-time polymerase chain reaction (qRT-PCR). Protein expression was determined by western blot. The half maximal inhibitory concentration (IC50) of PTX was determined by 3-(4,5-Dimethylthazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Cell proliferation was investigated by cell counting kit-8 (CCK-8) and colony formation assays. Cell apoptosis was demonstrated by flow cytometry analysis. Cell migration and invasion were evaluated by transwell assay. The binding relationship between miR-136 and circ_0061140 or CBX2 was predicted by interactome or starbase online database, and identified by dual-luciferase reporter assay. The effects of circ_0061140 on tumor formation and PTX sensitivity in vivo were disclosed by tumor formation assay. Results Circ_0061140 and CBX2 expressions were upregulated, while miR-136 expression was downregulated in PTX-resistant tissues and cells compared with control groups. Circ_0061140 knockdown repressed cell proliferation, migration and invasion, and promoted cell apoptosis and PTX sensitivity; however, these effects were restrained by miR-136 RNAi. Additionally, circ_0061140 was a sponge of miR-136, and miR-136 bound to CBX2. Furthermore, circ_0061140 knockdown inhibited tumor formation and improved PTX sensitivity in vivo. Conclusions Circ_0061140 silencing repressed the progression and PTX resistance of ovarian cancer by downregulating CBX2 expression via sponging miR-136, which provided novel insight into studying the therapy of ovarian cancer with PTX.


2021 ◽  
Vol 28 (1) ◽  
Author(s):  
Jingpeng Wang ◽  
Shuyuan Li ◽  
Gaofeng Zhang ◽  
Huihua Han

Abstract Background Sevoflurane (Sev), a commonly used volatile anesthetic, has been reported to inhibit the process of colorectal cancer (CRC). Circular RNAs (circRNAs) are revealed to participate in the pathogenesis of CRC. This study aims to reveal the mechanism of hsa_circ_0000231 in Sev-mediated CRC progression. Methods The expression of hsa_circ_0000231 and microRNA-622 (miR-622) was detected by quantitative real-time polymerase chain reaction (qRT-PCR). Protein level was determined by western blot analysis. Cell proliferation was investigated by 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), cell colony formation and DNA content quantitation assays. Cell apoptosis was detected by Annexin V-fluorescein isothiocyanate and propidium iodide double staining and caspase 3 activity assays. Cell migration and invasion were investigated by wound-healing and transwell invasion assays, respectively. The putative relationship between hsa_circ_0000231 and miR-622 was predicted by circular RNA Interactome online database, and identified by dual-luciferase reporter and RNA immunoprecipitation assays. The impacts of hsa_circ_0000231 on Sev-mediated tumor formation in vivo were presented by in vivo assay. Results Hsa_circ_0000231 expression was upregulated, while miR-622 was downregulated in CRC tissues and cells compared with control groups. Sev treatment decreased hsa_circ_0000231 expression, but increased miR-622 expression in CRC cells. Sev treatment suppressed cell proliferation, migration and invasion, and induced cell apoptosis. Hsa_circ_0000231 overexpression restored Sev-mediated CRC progression in vitro. Additionally, hsa_circ_0000231 acted as a sponge of miR-622, and miR-622 inhibitors reversed the impacts of hsa_circ_0000231 silencing on CRC process. Furthermore, Sev treatment inhibited tumor growth by regulating hsa_circ_0000231 in vivo. Conclusion Hsa_circ_0000231 attenuated Sev-aroused repression impacts on CRC development by sponging miR-622. This findings may provide an appropriate anesthetic protocol for CRC sufferers undergoing surgery.


2018 ◽  
Vol 50 (3) ◽  
pp. 810-822 ◽  
Author(s):  
Nan Sheng ◽  
Yun-Zhao Xu ◽  
Qing-Hua Xi ◽  
Hai-Yan Jiang ◽  
Chen-Yi Wang ◽  
...  

Background/Aims: This study aimed to investigate the expression and prognostic value of kinesin family member 2A (KIF2A) and the suppression effects of microRNA-206 (miR-206) on KIF2A in ovarian cancer. Methods: Ovarian cancer tissues from patients and ovarian cancer cell lines (A2780 and SKOV3) were used in this study. miR-206 mimics and control were transiently transfected into cells. RT-qPCR was performed to detect KIF2A mRNA and miR-206 expression levels, Western blot was performed to detect KIF2A protein levels, Dual-Luciferase Reporter Assay was used to examine the inhibition effects of miR-206 on KIF2A mRNA, immunohistochemical staining was used to examine the expression of KIF2A in tissue sections. CCK-8, transwell and Annexin-V-FITC/Propidium Iodide staining with flow cytometry were used to detect the cell proliferation, migration/invasion, and apoptosis respectively. Results: Our study explored the expression profiles of KIF2A and miR-206 in the patients with ovarian cancer. We found that overexpression of KIF2A was associated with a poor prognosis in ovarian cancer. We also found that KIF2A mRNA contains two target sites for miR-206 binding and confirmed that miR-206 directly suppresses KIF2A; inhibits ovarian cancer cell proliferation, migration, and invasion; and induces apoptosis. Conclusion: The results suggest KIF2A could serve a valuable prognostic indicator in ovarian cancer and provide a rationale for treatment of ovarian cancer by targeting KIF2A via miR-206.


2016 ◽  
Vol 212 (4) ◽  
pp. 239-246 ◽  
Author(s):  
Yan Deng ◽  
Lifei Jiang ◽  
Yingying Wang ◽  
Qinghua Xi ◽  
Jianxin Zhong ◽  
...  

2020 ◽  
Author(s):  
Xuan Wei ◽  
Juanjuan Shi ◽  
Qianhan Lin ◽  
Xiaoxue Ma ◽  
Yingxin Pang ◽  
...  

Abstract Background: Ovarian cancer is the most lethal female genital malignancy. Though cisplatin is still the first-line chemotherapy to treat ovarian cancer patients with debulking surgeries, its efficacy is limited due to the high-incidence of cisplatin resistance. ATP citrate lyase (ACLY) has been proved to be a key metabolic enzyme and was related to poor prognosis in various cancer, including ovarian cancer. Nevertheless, there has not been any research elucidating the relationship between ACLY and cisplatin resistance and the mechanism of how it works.Methods: Survival analysis was mainly carried out on the website. Bioinformatic analysis was performed in R/R studio. Proliferative activity was measured by MTT assay and colony formation assay. Cell cycle and apoptosis analysis were performed by flow cytometry. Acquired cisplatin resistant cell line A2780/CDDP was generated from A2780 by exposing to gradually elevated concentration of cisplatin. MTT assay was used to calculate IC50 of cisplatin. Xenograft tumor assay was used test cell proliferation in vivo.Results: Higher expression of ACLY was found in ovarian cancer tissue and related to poor prognosis. Knockdown of ACLY in A2780, SKOV3 and HEY cells inhibited cell proliferation, caused cell cycle arrest by modulating P16/CDK4/CCDN1 pathway and induced apoptosis probably by inhibiting p-AKT activity. Bioinformatic analysis of GSE15709 dataset revealed upregulation of ACLY and activation of PI3K/AKT pathway in acquired cisplatin resistant cells, in line with the results of A2780/CDDP cells generated by us. Knockdown of ACLY could alleviate cisplatin resistance and work synergistically with cisplatin treatment in inducing apoptosis in A2780/CDDP cells, by inhibiting PI3K/AKT pathway and activating AMPK/ROS pathway. ACLY specific inhibitor SB-204990 also showed the same effect. In A2780/CDDP cells, AKT overexpression could destroy cisplatin re-sensitization caused by ACLY knockdown. Conclusions: Knockdown of ACLY attenuated cisplatin resistance by inhibiting PI3K/AKT pathway and activating AMPK/ROS pathway. These findings suggested that combination of ACLY inhibition and cisplatin could be an effective strategy for overcoming cisplatin resistance in ovarian cancer.


2021 ◽  
Vol 11 ◽  
Author(s):  
Xuan Wei ◽  
Juanjuan Shi ◽  
Qianhan Lin ◽  
Xiaoxue Ma ◽  
Yingxin Pang ◽  
...  

Background: Ovarian cancer is the most lethal female genital malignancy. Although cisplatin is the first-line chemotherapy to treat ovarian cancer patients along with debulking surgeries, its efficacy is limited due to the high incidence of cisplatin resistance. ATP citrate lyase (ACLY) has been shown to be a key metabolic enzyme and is associated with poor prognosis in various cancers, including ovarian cancer. Nevertheless, no studies have probed the mechanistic relationship between ACLY and cisplatin resistance.Methods: Survival analysis was mainly carried out online. Bioinformatic analysis was performed in R/R studio. Proliferative activity was measured by MTT and colony formation assays. Cell cycle and apoptosis analysis were performed by flow cytometry. The acquired-cisplatin-resistant cell line A2780/CDDP was generated by exposing A2780 to cisplatin at gradually elevated concentrations. MTT assay was used to calculate IC50 values of cisplatin. A xenograft tumor assay was used test cell proliferation in vivo.Results: Higher expression of ACLY was found in ovarian cancer tissue and related to poor prognosis. Knockdown of ACLY in A2780, SKOV3, and HEY cells inhibited cell proliferation, caused cell-cycle arrest by modulating the P16–CDK4–CCND1 pathway, and induced apoptosis probably by inhibiting p-AKT activity. Bioinformatic analysis of the GSE15709 dataset revealed upregulation of ACLY and activation of PI3K–AKT pathway in cells with acquired cisplatin resistance, in line with observations on A2780/CDDP cells that we generated. Knockdown of ACLY alleviated cisplatin resistance, and works synergistically with cisplatin treatment to induce apoptosis in A2780/CDDP cells by inhibiting the PI3K–AKT pathway and activating AMPK–ROS pathway. The ACLY-specific inhibitor SB-204990 showed the same effect. In A2780/CDDP cells, AKT overexpression could attenuate cisplatin re-sensitization caused by ACLY knockdown.Conclusions: Knockdown of ACLY attenuated cisplatin resistance by inhibiting the PI3K–AKT pathway and activating the AMPK–ROS pathway. These findings suggest that a combination of ACLY inhibition and cisplatin might be an effective strategy for overcoming cisplatin resistance in ovarian cancer.


Sign in / Sign up

Export Citation Format

Share Document