Bupivacaine modulates the apoptosis and ferroptosis in bladder cancer via PI3K/AKT pathway

Author(s):  
Jianli Hao ◽  
Weiqing Zhang ◽  
Zeqing Huang

Abstract Background The study aimed to explore the effects of local anesthetic bupivacaine on bladder cancer cells in vivo and in vitro. Methods After T24 cells and 5637 cells were treated with different doses of bupivacaine (0-16 mM) for 24 h, MTT assay was used to detect the cytotoxicity, and bupivacaine (0.25, 0.5, 1 mM) was selected for subsequent experiments. Apoptosis was detected by Hoechst 33342 staining and TUNEL. The contents of Fe2+, MDA, GSH and ROS were detected by the corresponding kit. Mitochondrial membrane potential was detected by JC-1 kit. HE staining, TUNEL and immunohistochemistry were used to detect the xenografted tumors. Protein expression was detected by western blot. Results Bupivacaine significantly inhibited the activity of T24 cells and 5637 cells at 0.25-16 mM. Bupivacaine significantly promoted cell apoptosis with increasd concentration. bupivacaine inhibited the expression of Bcl-2 and increased the expression of Bax and cytochrome C. Moreover, bupivacaine promoted the increase of Fe2+ and ROS, and inhibited the expression of xCT and GPX4. Further results showed that bupivacaine decreased mitochondrial membrane potential, reduced GSH, and increased MDA levels. Besides, bupivacaine attenuated the phosphorylation of PI3K, Akt, and mTOR, which might be involved in the regulation of bupivacaine-induced apoptosis and ferroptosis. In addition, bupivacaine suppressed the growth of xenografted tumors, induced apoptosis and ferroptosis, and inhibited the activity of PI3K/AKT signaling pathway in xenografted tumors. Conclusion Bupivacaine could induce apoptosis and ferroptosis by inhibiting PI3K / Akt signaling pathway in bladder cancer cells.

2020 ◽  
Vol 19 (5) ◽  
pp. 965-970
Author(s):  
Sheng Li ◽  
Li-bin Zhou ◽  
Tie-lin Wu ◽  
Min Yin ◽  
Hui-min Long

Purpose: To study the influence of cordycepin (Cor) on cisplatin insensitivity in bladder carcinoma, and its underlying mechanism of action.Methods: The effects of cisplatin and Cor treatments on the viability of T24-sensitive and T24/DDPinsensitive bladder carcinoma cells were investigated by using 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide (MTT) method to assess selectivity index. Flow cytometry was employed to evaluate the apoptosis of T24/DDP-resistant bladder cancer cells treated with cisplatin and Cor. The concentrations of PTEN, p-AKt and Akt in T24/DDP-resistant bladder cancer cells treated with cisplatin and Cor were determined by western blot assay.Results: Compared with T24-sensitive cells, the sensitivity of T24/DDP-resistant bladder cancer cells to cisplatin was significantly decreased, along with significant increase in half-inhibitory concentration (IC50) value, resulting in 10.56-fold increase in resistance (p < 0.05). The median effective concentration (EC50) value of Cor for DDP reversal was 1.03 ± 0.15 μM, and it had a high selectivity index for normal cells (> 48.5). The results from flow cytometry showed that Cor significantly enhanced the apoptosisinducing capacity of DDP in T24/DDP-resistant cells (p < 0.05), while Western blot data indicate that PTEN protein expression increased and phosphorylated Akt protein expression decreased in T24/DDPresistantcells after Cor treatment when compared with control group (p < 0.05).Conclusion: Cordycepin significantly improves the sensitivity of T24/ DDP-resistant bladder cancer cells to cisplatin via a mechanism related to the activation of PTEN/AKt signaling pathway, thus indicating that it is a potential candidate reversing DDP-resistance in bladder cancer. Keywords: Bladder cancer, Cordycepin, Cisplatin resistance, PTEN/Akt signaling pathway


2019 ◽  
Vol Volume 11 ◽  
pp. 7909-7923 ◽  
Author(s):  
Cong Zhang ◽  
Xiangping Chang ◽  
Dongshan Chen ◽  
Feilong Yang ◽  
Zeyan Li ◽  
...  

2020 ◽  
Author(s):  
Mengjie Huangfu ◽  
Juan Wang ◽  
Dan Yu ◽  
Jianli Qin ◽  
Xiao Guan ◽  
...  

Abstract Background: Osthole is a natural coumarin which has been proved to inhibit growth of cancer cells by inducing cancer cells death, while its mechanism of anticancer remains unclearly. In our study, we found that osthole activated multiple forms of cell death including apoptosis, secondary necrosis and mitophagy in receptor interacting protein kinase (RIP) 3-deficient cervical cancer HeLa cells. Methods: Cell viability was detected by MTT assay. Cell membrane integrity was detected by LDH release assay and PI staining. Cell apoptosis and necrosis were detected by flow cytometry assay. Reactive oxygen species (ROS) was detected by DCFH-DA staining and mitochondrial membrane potential (MMP) was detected by JC-1 staining using flow cytometry. The expression of proteins was detected by western blotting assay and proteomics. Xenograft tumor model was used to evaluate the effect of osthole in vivo.Results: Our study showed osthole caused HeLa cells apoptosis and secondary necrosis, which is a phenomenon of the apoptotic cells’ plasma membrane breakdown. And when Hela cells pretreatment with Z-DEVD-FMK, an irreversible caspase-3 inhibitor, not only inhibited osthole-induced apoptosis but also necrosis. Moreover, we found that Z-DEVD-FMK reversed the effect of osthole on the induction of cleaved the N-terminal fragment of GSDME in Hela cells. Furthermore, inhibition of NAD (P) H: quinone oxidoreductase 1 (NQO1) by osthole induced the overproduction of reactive oxygen species (ROS). ROS inhibitor N-Acetyl-L-cysteine (NAC) not only reduced osthole-induced apoptosis, but also reversed its effect on the necrotic induction and the GSDME N-terminal generation. It was shown that osthole decreased mitochondrial membrane potential (MMP) and increased the expression of PTEN-induced putative kinase 1 (PINK1) and Parkin, which indicated that the activation of mitophagy induced by osthole. Meanwhile, as well as apoptosis and secondary necrosis, mitophagy was also restrained by NAC. Conclusions: In conclusion, all these data suggested that osthole induced apoptosis, secondary necrosis and mitophagy via NQO1-mediated ROS overproduction.


Sign in / Sign up

Export Citation Format

Share Document