L-Selenomethylselenocysteine Exerts Inhibitory Effects on the Progression of Esophageal Cancer by Targeting the PI3K/AKT Signaling Pathway.

Keyi Ji, Suhui Wu, Jiayao Yuan, Yu Wang, Genlin Li, Linlin Wang, Shuncai Wang, Longjie Wang, Hanbing Li, Chengbao Wang
{"title":"L-Selenomethylselenocysteine Exerts Inhibitory Effects on the Progression of Esophageal Cancer by Targeting the PI3K/AKT Signaling Pathway.","authors":"Keyi Ji, Suhui Wu, Jiayao Yuan, Yu Wang, Genlin Li, Linlin Wang, Shuncai Wang, Longjie Wang, Hanbing Li, Chengbao Wang","doi":"10.2174/0115748928355152250527060605","DOIUrl":null,"url":null,"abstract":"<p><strong>Background: </strong>Esophageal cancer is a common malignant tumor, making the search for effective treatments a critical research focus. L-methylselenocysteine (L-SeMC) has been reported to exert anticancer effects in various cancers; however, its role and underlying mechanisms in esophageal cancer remain unclear. This study aimed to investigate the anticancer effects of L-SeMC on esophageal cancer both in vitro and in vivo, and to explore its potential mechanisms of action.</p><p><strong>Methods: </strong>For cellular studies, flow cytometry, colony formation assay, MTT assay, wound healing assay, and ROS measurement were employed. Western blotting was used to assess the expression levels of apoptotic proteins. A subcutaneous tumor xenograft model was established. The analysis included the evaluation of proteins related to the PI3K/AKT signaling pathway, TUNEL, and Ki-67 staining, as well as HE staining.</p><p><strong>Results: </strong>L-SeMC caused cell death and, in a concentration-dependent manner, reduced the migration, invasion, and proliferation of esophageal cancer cells. Western blot analysis showed that L-SeMC was associated with a decrease in the anti-apoptotic protein Bcl-2 and an increase in the pro-apoptotic protein Bax. It also triggered the mitochondrial apoptosis pathway, promoting the activation of caspase-3 and subsequent cancer cell death induced by L-SeMC. In a dosedependent manner, L-SeMC decreased the phosphorylation of phosphatidylinositol 3-kinase (PI3K) downstream effector molecules. This suggests that L-SeMC inhibits the PI3K/AKT signaling pathway in esophageal cancer cells, contributing to its anticancer effects.</p><p><strong>Conclusion: </strong>L-SeMC has a strong anticancer effect on human esophageal cancer cells and promotes apoptosis by inhibiting the PI3K/AKT signaling pathway, suggesting that L-SeMC may represent a novel strategy for the treatment of esophageal cancer.</p>","PeriodicalId":94186,"journal":{"name":"Recent patents on anti-cancer drug discovery","volume":" ","pages":""},"PeriodicalIF":0.0000,"publicationDate":"2025-06-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Recent patents on anti-cancer drug discovery","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.2174/0115748928355152250527060605","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"","JCRName":"","Score":null,"Total":0}
引用次数: 0

Abstract

Background: Esophageal cancer is a common malignant tumor, making the search for effective treatments a critical research focus. L-methylselenocysteine (L-SeMC) has been reported to exert anticancer effects in various cancers; however, its role and underlying mechanisms in esophageal cancer remain unclear. This study aimed to investigate the anticancer effects of L-SeMC on esophageal cancer both in vitro and in vivo, and to explore its potential mechanisms of action.

Methods: For cellular studies, flow cytometry, colony formation assay, MTT assay, wound healing assay, and ROS measurement were employed. Western blotting was used to assess the expression levels of apoptotic proteins. A subcutaneous tumor xenograft model was established. The analysis included the evaluation of proteins related to the PI3K/AKT signaling pathway, TUNEL, and Ki-67 staining, as well as HE staining.

Results: L-SeMC caused cell death and, in a concentration-dependent manner, reduced the migration, invasion, and proliferation of esophageal cancer cells. Western blot analysis showed that L-SeMC was associated with a decrease in the anti-apoptotic protein Bcl-2 and an increase in the pro-apoptotic protein Bax. It also triggered the mitochondrial apoptosis pathway, promoting the activation of caspase-3 and subsequent cancer cell death induced by L-SeMC. In a dosedependent manner, L-SeMC decreased the phosphorylation of phosphatidylinositol 3-kinase (PI3K) downstream effector molecules. This suggests that L-SeMC inhibits the PI3K/AKT signaling pathway in esophageal cancer cells, contributing to its anticancer effects.

Conclusion: L-SeMC has a strong anticancer effect on human esophageal cancer cells and promotes apoptosis by inhibiting the PI3K/AKT signaling pathway, suggesting that L-SeMC may represent a novel strategy for the treatment of esophageal cancer.

l -硒甲基硒半胱氨酸通过PI3K/AKT信号通路抑制食管癌进展
背景:食管癌是一种常见的恶性肿瘤,寻找有效的治疗方法是研究的重点。l -甲基硒半胱氨酸(L-SeMC)已被报道在多种癌症中发挥抗癌作用;然而,其在食管癌中的作用和潜在机制尚不清楚。本研究旨在研究L-SeMC对食管癌的体内外抗肿瘤作用,并探讨其潜在的作用机制。方法:细胞研究采用流式细胞术、菌落形成法、MTT法、创面愈合法、ROS法。Western blotting检测凋亡蛋白表达水平。建立皮下肿瘤异种移植模型。分析包括评估PI3K/AKT信号通路相关蛋白、TUNEL、Ki-67染色以及HE染色。结果:L-SeMC引起细胞死亡,并呈浓度依赖性地减少食管癌细胞的迁移、侵袭和增殖。Western blot分析显示,L-SeMC与抗凋亡蛋白Bcl-2的降低和促凋亡蛋白Bax的升高有关。它还触发线粒体凋亡通路,促进caspase-3的激活和L-SeMC诱导的癌细胞死亡。L-SeMC以剂量依赖的方式降低了磷脂酰肌醇3-激酶(PI3K)下游效应分子的磷酸化。这表明L-SeMC抑制食管癌细胞中PI3K/AKT信号通路,参与其抗癌作用。结论:L-SeMC对人食管癌细胞具有较强的抗癌作用,并通过抑制PI3K/AKT信号通路促进细胞凋亡,提示L-SeMC可能是一种治疗食管癌的新策略。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 求助全文
来源期刊
自引率
0.00%
发文量
0
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术官方微信