Apoptosis triggered by isoquercitrin in bladder cancer cells by activating the AMPK-activated protein kinase pathway

2017 ◽  
Vol 8 (10) ◽  
pp. 3707-3722 ◽  
Author(s):  
Ping Wu ◽  
Siyuan Liu ◽  
Jianyu Su ◽  
Jianping Chen ◽  
Lin Li ◽  
...  

Our findings provide comprehensive evidence that isoquercitrin (ISO) influenced T24 bladder cancer cell metabolism by activating the AMPK pathway as identified by combination with metabolomics and immunoblotting assay.

2020 ◽  
Author(s):  
Prasanna Vasudevan Iyengar ◽  
Dieuwke Louise Marvin ◽  
Dilraj Lama ◽  
Tuan Zea Tan ◽  
Sudha Suriyamurthy ◽  
...  

AbstractBladder cancer is one of the most prevalent cancer types in the world, frequently exhibiting invasion and metastasis and therefore associated with poor prognosis. It is a progressive disease with high recurrence rates even after surgery, which calls for the urgent need for early intervention and diagnosis. The E3 ubiquitin ligase TNF Receptor Associated Factor 4 (TRAF4) has been largely implicated as a tumour-promoting element in a wide range of cancers. Over-expression and amplification of this gene product has been a common observation in breast and other metastatic tumours. In contrast, we observed that expression of TRAF4 negatively correlated with overall patient survival. Moreover, its expression was repressed at epigenetic levels in aggressive bladder cancer cells. We also describe an ERK kinase phosphorylation site on TRAF4 that inhibits its stability and localization to plasma membrane in such cells. Furthermore, knockdown of TRAF4 in epithelial bladder cancer cell lines leads to gain of mesenchymal genes and a loss of epithelial integrity. Reciprocally, stable over-expression of TRAF4 in mesenchymal cells leads to decreased migratory and invasive properties. Transcriptomic analysis upon TRAF4 mis-expression in bladder cancer cell lines revealed that higher TRAF4 expression enhanced BMP/SMAD and dampened NF-κB signalling pathways. Importantly, this notion was confirmed in bladder cancer patient material. Mechanistically, we showed that TRAF4 targets the E3 ubiquitin ligase SMURF1, a negative regulator of BMP/SMAD signalling, for proteasomal degradation in bladder cancer cells. We show that genetic and pharmacological inhibition of SMURF1 or its function inhibited migration of aggressive (mesenchymal) bladder cancer cells.


2019 ◽  
Vol 19 (1) ◽  
Author(s):  
Jinan Guo ◽  
Zhixin Chen ◽  
Hongtao Jiang ◽  
Zhou Yu ◽  
Junming Peng ◽  
...  

Abstract Background Bladder cancer is the most common human urological malignancies with poor prognosis, and the pathophysiology of bladder cancer involves multi-linkages of regulatory networks in the bladder cancer cells. Recently, the long noncoding RNAs (lncRNAs) have been extensively studied for their role on bladder cancer progression. In this study, we evaluated the expression of DLX6 Antisense RNA 1 (DLX6-AS1) in the cancerous bladder tissues and studied the possible mechanisms of DLX6-AS1 in regulating bladder cancer progression. Methods Gene expression was determined by qRT-PCR; protein expression levels were evaluated by western blot assay; in vitro functional assays were used to determine cell proliferation, invasion and migration; nude mice were used to establish the tumor xenograft model. Results Our results showed the up-regulation of DLX6-AS1 in cancerous bladder cancer tissues and bladder cell lines, and high expression of DLX6-AS1 was correlated with advance TNM stage, lymphatic node metastasis and distant metastasis. The in vitro experimental data showed that DLX6-AS1 overexpression promoted bladder cancer cell growth, proliferation, invasion, migration and epithelial-to-mesenchymal transition (EMT); while DLX6-AS1 inhibition exerted tumor suppressive actions on bladder cancer cells. Further results showed that DLX6-AS1 overexpression increased the activity of Wnt/β-catenin signaling, and the oncogenic role of DLX6-AS1 in bladder cancer cells was abolished by the presence of XAV939. On the other hand, DLX6-AS1 knockdown suppressed the activity of Wnt/β-catenin signaling, and the tumor-suppressive effects of DLX6-AS1 knockdown partially attenuated by lithium chloride and SB-216763 pretreatment. The in vivo tumor growth study showed that DLX6-AS1 knockdown suppressed tumor growth of T24 cells and suppressed EMT and Wnt/β-catenin signaling in the tumor tissues. Conclusion Collectively, the present study for the first time identified the up-regulation of DLX6-AS1 in clinical bladder cancer tissues and in bladder cancer cell lines. The results from in vitro and in vivo assays implied that DLX6-AS1 exerted enhanced effects on bladder cancer cell proliferation, invasion and migration partly via modulating EMT and the activity of Wnt/β-catenin signaling pathway.


2021 ◽  
Author(s):  
Yuki Matsuoka ◽  
Rikiya Taoka ◽  
Yoichiro Tohi ◽  
Zhang Xia ◽  
Mikio Sugimoto

Abstract Background:Bladder cancer is a major health concern worldwide. The conventional intravesical Bacillus Calmette–Guérin therapy has certain shortcomings; thereby, demanding novel alternatives. Although sterile water is a probable agent for such novel intravesical therapies, bladder cancer cell lines differ in their sensitivity to hypotonic shock due to sterile water. Therefore, we aimed to investigate whether Cl- channel blockers enhance the cytocidal effect of hypotonic shock on bladder cancer cells resistant to sterile water.Methods:Bladder cancer cell lines of varying grades (RT112, T24, and J82) were exposed to sterile water, and morphological changes were closely observed using microscopy. Sterile water-induced changes in cell membrane integrity and cell viability were analyzed to determine the effects of hypotonic shock. These effects were further analyzed using a Cl- channel blocker.Results:T24 and J82 cells started swelling immediately upon exposure to sterile water and ruptured within 10 min. RT112 cells demonstrated limited hypotonic swelling with few cell ruptures. After treatment with the Cl- channel blocker, RT112 cells ruptured faster as compared to that in cells treated with sterile water. The percentages of viable dimethylsulfoxide and 5-nitro-2-(3-phenylpropylamino) benzoic acid -treated (50, 100, 200, and 300 µM) RT112 cells after 10 min of exposure to sterile water were 13.6 % ± 3.4 %, 6.3 % ± 1.2 %, 2.0 % ± 1.1 %, 0.7 % ± 0.7 %, and 0 %, respectively.Conclusions:Taken together, the Cl- channel blockers enhanced the cytocidal effects of hypotonic shock in bladder cancer cells. Intravesical therapy with sterile water after treatment with a Cl- channel blocker represents a potential new adjuvant therapy after TURBT with high efficacy.


2015 ◽  
Vol 33 (7_suppl) ◽  
pp. 323-323
Author(s):  
Chong-Xian Pan ◽  
Tzu-yin Lin ◽  
Hongyong Zhang ◽  
Yuanpei Li ◽  
Susan D. Airhart ◽  
...  

323 Background: We recently developed a bladder cancer-specific targeting ligand named PLZ4, nanometer-scale micelles and nanoporphyrin. Here we report the diagnostic and therapeutic applications of these nanotheranostics coated with PLZ4. Methods: PLZ4 was synthesized through solid phase synthesis. Bladder cancer-specific PLZ4-coated nanomicelles (PNM) and nanoporphyrin (PNP) were developed through conjugating the nanotheranostics with PLZ4 on the surface, and loaded with therapeutic and/or imaging agents in the core. Bladder cancer cell lines and patient-derived xenografts (PDXs) were used to determine the diagnostic and therapeutic applications. Results: In vitro studies with cell lines revealed that both PNM and PNP could specifically deliver the drug load to bladder cancer cells, but not to adjacent confounding cells. After intravenous injection, PNM loaded with paclitaxel could specifically deliver the drug load to xenografts developed from a human and a dog bladder cancer cell line, and a PDX, but not to lung cancer xenografts in the same mice. These paclitaxel-loaded PNM could overcome cisplatin resistance, and prolong the overall survival of mice carrying PDXs from 27 days with free paclitaxel to 76 days (p<0.0001). PNP can be used for photodynamic diagnosis and therapy while being able to chelate gadolinium for cancer-specific magnetic resonance imaging (MRI), and load chemotherapeutic drugs for cancer-specific targeted chemotherapy. It is over 50 times more potent that 5-aminolevulinic acid in photodynamic therapy (p<0.0001). After intravesical instillation into the bladder cavity of an orthotopic PDX model, PNP could specifically target bladder cancer cells, but not adjacent normal urothelial cells in the same bladder. Conclusions: PNM and PNP can potentially be used for diagnosis, imaging detection and cancer-specific targeted delivery of therapeutic agents of both non-myoinvasive and advanced bladder cancer. A phase I clinical trial of PNM for intravesical instillation in human patients with non-myoinvasive bladder cancer, and another phase I trial with PNP for photodynamic diagnosis and therapy in dog bladder cancer patients have been funded.


2020 ◽  
Vol 21 (16) ◽  
pp. 5658
Author(s):  
Kyoung-Hwa Lee ◽  
Byung-Chan Kim ◽  
Seung-Hwan Jeong ◽  
Chang Wook Jeong ◽  
Ja Hyeon Ku ◽  
...  

Histone demethylase KDM7A regulates many biological processes, including differentiation, development, and the growth of several cancer cells. Here, we have focused on the role of KDM7A in bladder cancer cells, especially under drug-resistant conditions. When the KDM7A gene was knocked down, bladder cancer cell lines showed impaired cell growth, increased cell death, and reduced rates of cell migration. Biochemical studies revealed that KDM7A knockdown in the bladder cancer cells repressed the activity of androgen receptor (AR) through epigenetic regulation. When we developed a cisplatin-resistant bladder cancer cell line, we found that AR expression was highly elevated. Upon treatment with TC-E 5002, a chemical inhibitor of KDM7A, the cisplatin-resistant bladder cancer cells, showed decreased cell proliferation. In the mouse xenograft model, KDM7A knockdown or treatment with its inhibitor reduced the growth of the bladder tumor. We also observed the upregulation of KDM7A expression in patients with bladder cancer. The findings suggest that histone demethylase KDM7A mediates the growth of bladder cancer. Moreover, our findings highlight the therapeutic potential of the KMD7A inhibitor, TC-E 5002, in patients with cisplatin-resistant bladder cancer.


2021 ◽  
Vol 22 (8) ◽  
pp. 4102
Author(s):  
Xia Zhang ◽  
Rikiya Taoka ◽  
Dage Liu ◽  
Yuki Matsuoka ◽  
Yoichiro Tohi ◽  
...  

RRM1—an important DNA replication/repair enzyme—is the primary molecular gemcitabine (GEM) target. High RRM1-expression associates with gemcitabine-resistance in various cancers and RRM1 inhibition may provide novel cancer treatment approaches. Our study elucidates how RRM1 inhibition affects cancer cell proliferation and influences gemcitabine-resistant bladder cancer cells. Of nine bladder cancer cell lines investigated, two RRM1 highly expressed cells, 253J and RT112, were selected for further experimentation. An RRM1-targeting shRNA was cloned into adenoviral vector, Ad-shRRM1. Gene and protein expression were investigated using real-time PCR and western blotting. Cell proliferation rate and chemotherapeutic sensitivity to GEM were assessed by MTT assay. A human tumor xenograft model was prepared by implanting RRM1 highly expressed tumors, derived from RT112 cells, in nude mice. Infection with Ad-shRRM1 effectively downregulated RRM1 expression, significantly inhibiting cell growth in both RRM1 highly expressed tumor cells. In vivo, Ad-shRRM1 treatment had pronounced antitumor effects against RRM1 highly expressed tumor xenografts (p < 0.05). Moreover, combination of Ad-shRRM1 and GEM inhibited cell proliferation in both cell lines significantly more than either treatment individually. Cancer gene therapy using anti-RRM1 shRNA has pronounced antitumor effects against RRM1 highly expressed tumors, and RRM1 inhibition specifically increases bladder cancer cell GEM-sensitivity. Ad-shRRM1/GEM combination therapy may offer new treatment options for patients with GEM-resistant bladder tumors.


2021 ◽  
Vol 21 (1) ◽  
Author(s):  
Yaoyao Xiong ◽  
Xiongbing Zu ◽  
Long Wang ◽  
Yuan Li ◽  
Minfeng Chen ◽  
...  

Abstract Background Invasive bladder tumors cause a worse prognosis in patients and remain a clinical challenge. Epithelial–mesenchymal transition (EMT) is associated with bladder cancer metastasis. In the present research, we attempted to demonstrate a novel mechanism by which a long noncoding RNA (lncRNA)-miRNA-mRNA axis regulates EMT and metastasis in bladder cancer. Methods Immunofluorescence (IF) staining was used to detect Vimentin expression. The protein expression of ZEB1, Vimentin, E-cadherin, and Snail was investigated by using immunoblotting assays. Transwell assays were performed to detect the invasive capacity of bladder cancer cells. A wound healing assay was used to measure the migratory capacity of bladder cancer cells. Results Herein, we identified lncRNA VIM-AS1 as a highly- expressed lncRNA in bladder cancer, especially in metastatic bladder cancer tissues and high-metastatic bladder cancer cell lines. By acting as a ceRNA for miR-655, VIM-AS1 competed with ZEB1 for miR-655 binding, therefore eliminating the miR-655-mediated suppression of ZEB1, finally promoting EMT in both high- and low-metastatic bladder cancer cells and enhancing cancer cell metastasis. Conclusions In conclusion, the VIM-AS1/miR-655/ZEB1 axis might be a promising target for improving bladder cancer metastasis via an EMT-related mechanism.


2021 ◽  
Vol 11 ◽  
Author(s):  
Changcheng Guo ◽  
Xin Li ◽  
Jinbo Xie ◽  
Dan Liu ◽  
Jiang Geng ◽  
...  

LncRNAs play important roles in bladder cancer. However, only a few studies report on the correlation between lncRNAs expression and autophagy in bladder cancer. This study aimed to explore the effect of lncRNA on autophagy in bladder cancer. The findings showed high expression of SNHG1 in the bladder cancer cells and tumor tissues. The high expression of SNHG1 was positively correlated with bladder cancer cell invasion, proliferation, and autophagy. This finding implies that SNHG1 promotes bladder cancer cell invasion and proliferation via autophagy. Further analysis of the mechanism of action of SNHG1 showed that it functions as a sponge of miRNA-493 in bladder cancer. miRNA-493 binds on the 3’ -UTR of ATG14 mRNA thus affecting ATG14 protein expression, which is implicated in autophagy. These findings are supported by previous preclinical studies using multiple Bca cell lines and TCGA, which demonstrate that SNHG1 plays an oncogenic role by acting as a sponge of miR-493-5p or as its ceRNA. Upregulation of SNHG1 promotes proliferation, invasion, and autophagy of bladder cancer cells through the miR-493-5p/ATG14/autophagy pathway. Therefore, SNHG1 may act as a potential therapeutic target for the treatment of bladder cancer.


2021 ◽  
Author(s):  
pei wan ◽  
Zhilin Chen ◽  
Minzhi Huang ◽  
Huiming Jiang ◽  
Huajun Wu ◽  
...  

Abstract BackgroundMicroRNAs (miRs) are endogenous, single-stranded, non-coding RNAs that are involved in various physiological processes, development and the progression of various types of cancer. The role of miR-200a-3p in various types of cancer has been previously reported. The present study aimed to investigate the expression levels of miR-200a-3p in human bladder cancer, as well as its potential role in disease pathogenesis.MethodsIn vitro: Agilent miRNA microarray and QPCR analysis of miR-200a-3p expression in bladder cancer. Gene overexpression and interference technology analysis of the effects of miR-200a-3p and de-ubiquituination enzyme TNFα induced protein 3 (A20) on the function of bladder cancer cells; dual luciferase technology for promoter efficiency analysis; Animal experiments: Nude mice are used for tumor formation experiments, and the effects of genes on tumors are directly analyzed on animals;ResultsCompared with adjacent non-tumor tissues, miR-200a-3p expression levels were significantly upregulated in bladder cancer tissues. Moreover, increased miR-200a-3p expression was significantly associated with distant metastasis and advanced stage. In addition, compared with the miR-Negative control (NC) group, miR-200a-3p overexpression promoted bladder cancer cell proliferation, migration, invasion, cell cycle and release of inflammatory cytokines, but inhibited cell apoptosis. Mechanistically, de-ubiquituination enzyme TNFα induced protein 3 (A20) was identified as a target gene of miR-200a-3p in bladder cancer cell lines. An inverse association between miR-200a-3p expression and A20 expression in bladder cancer tissues and cell lines was also identified. A20 overexpression in miR-200a-3p-overexpression bladder cancer cells attenuated miR-200a-3p overexpression-mediated effects on cell proliferation, migration, apoptosis and cytokine production. Moreover, compared with the miR-NC group, miR-200a-3p overexpression significantly promoted tumor growth in vivo, and A20 overexpression blocked the promoting effect of miR-200a-3p on bladder cancer. ConclusionsThe results of the present study indicated that miR-200a-3p might serve as an oncogene in human bladder cancer by targeting a novel gene A20; therefore, miR-200a-3p and A20 might serve as novel therapeutic targets for bladder cancer.


Sign in / Sign up

Export Citation Format

Share Document