Depleting ANTXR1 suppresses glioma growth via deactivating PI3K/AKT pathway.

IF 3.4 3区 生物学 Q3 CELL BIOLOGY
Cell Cycle Pub Date : 2023-10-01 Epub Date: 2023-12-05 DOI:10.1080/15384101.2023.2275900
Chaoyang Zhou, Aijun Liang, Jianzhong Zhang, Jingxing Leng, Bin Xi, Bin Zhou, Yu Yang, Ronglan Zhu, Liangchen Zhong, Xingxing Jiang, Dengfeng Wan
{"title":"Depleting ANTXR1 suppresses glioma growth via deactivating PI3K/AKT pathway.","authors":"Chaoyang Zhou, Aijun Liang, Jianzhong Zhang, Jingxing Leng, Bin Xi, Bin Zhou, Yu Yang, Ronglan Zhu, Liangchen Zhong, Xingxing Jiang, Dengfeng Wan","doi":"10.1080/15384101.2023.2275900","DOIUrl":null,"url":null,"abstract":"<p><p>Gliomas are commonly known as primary brain tumors and associated with frequent recurrence and an unsatisfactory prognosis despite extensive research in the underlying molecular mechanisms. We aimed to examine the role of ANTXR1 in glioma tumorigenesis and explore its downstream regulatory mechanism. ANTXR1 expression in clinical specimens and its relationship with some pathological characteristics were detected using immunohistochemical staining. After silencing/upregulating ANTXR1 through lentiviral transfection in glioma cell lines, qRT-PCR and western blotting were used to examine mRNA and protein levels, and cell phenotype was also detected. ANTXR1-knockdown and -overexpression cells were then processed by AKT activator and PI3K inhibitor, respectively, to verify downstream PI3K/AKT pathway regulated by ANTXR1. Xenograft nude mice models were constructed to verify the role of ANTXR1 <i>in vivo</i>. We found overexpression of ANTXR1 in both cell lines in comparison with those in normal brain tissues. Glioma cell growth and migratory ability were dramatically impaired as a result of silencing <i>ANTXR1</i> by shANTXR1 lentiviruses. ANTXR1 blockade also accelerated cell apoptosis and held back cell cycle via targeting G2 phrase during cell mitosis. <i>In vivo</i> xenograft models verified <i>in vitro</i> findings above. Further exploration disclosed that AKT activator promoted anti-tumor effects mediated by ANTXR1 knockdown, while PI3K inhibitor limited pro-tumor effects mediated by ANTXR1 overexpression, indicating that ANTXR1 functioned in glioma cells through regulating PI3K/AKT pathway. ANTXR1 could play an indispensable role in glioma tumorigenesis via activating PI3K/AKT-mediated cell growth. Our study provides a theoretical basis for targeting ANTXR1 as a molecular target in glioma clinical therapeutics.</p>","PeriodicalId":9686,"journal":{"name":"Cell Cycle","volume":null,"pages":null},"PeriodicalIF":3.4000,"publicationDate":"2023-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10732648/pdf/","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Cell Cycle","FirstCategoryId":"99","ListUrlMain":"https://doi.org/10.1080/15384101.2023.2275900","RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"2023/12/5 0:00:00","PubModel":"Epub","JCR":"Q3","JCRName":"CELL BIOLOGY","Score":null,"Total":0}
引用次数: 0

Abstract

Gliomas are commonly known as primary brain tumors and associated with frequent recurrence and an unsatisfactory prognosis despite extensive research in the underlying molecular mechanisms. We aimed to examine the role of ANTXR1 in glioma tumorigenesis and explore its downstream regulatory mechanism. ANTXR1 expression in clinical specimens and its relationship with some pathological characteristics were detected using immunohistochemical staining. After silencing/upregulating ANTXR1 through lentiviral transfection in glioma cell lines, qRT-PCR and western blotting were used to examine mRNA and protein levels, and cell phenotype was also detected. ANTXR1-knockdown and -overexpression cells were then processed by AKT activator and PI3K inhibitor, respectively, to verify downstream PI3K/AKT pathway regulated by ANTXR1. Xenograft nude mice models were constructed to verify the role of ANTXR1 in vivo. We found overexpression of ANTXR1 in both cell lines in comparison with those in normal brain tissues. Glioma cell growth and migratory ability were dramatically impaired as a result of silencing ANTXR1 by shANTXR1 lentiviruses. ANTXR1 blockade also accelerated cell apoptosis and held back cell cycle via targeting G2 phrase during cell mitosis. In vivo xenograft models verified in vitro findings above. Further exploration disclosed that AKT activator promoted anti-tumor effects mediated by ANTXR1 knockdown, while PI3K inhibitor limited pro-tumor effects mediated by ANTXR1 overexpression, indicating that ANTXR1 functioned in glioma cells through regulating PI3K/AKT pathway. ANTXR1 could play an indispensable role in glioma tumorigenesis via activating PI3K/AKT-mediated cell growth. Our study provides a theoretical basis for targeting ANTXR1 as a molecular target in glioma clinical therapeutics.

耗尽ANTXR1通过使PI3K/AKT通路失活来抑制胶质瘤的生长。
胶质瘤通常被称为原发性脑肿瘤,尽管对其潜在的分子机制进行了广泛的研究,但其复发频繁,预后不佳。我们旨在研究ANTXR1在胶质瘤发生中的作用,并探讨其下游调控机制。应用免疫组织化学染色法检测临床标本中ANTXR1的表达及其与部分病理特征的关系。在胶质瘤细胞系中通过慢病毒转染沉默/上调ANTXR1后,采用qRT-PCR和western blotting检测mRNA和蛋白水平,并检测细胞表型。然后分别用AKT激活剂和PI3K抑制剂处理ANTXR1敲低和过表达细胞,以验证ANTXR1调控的下游PI3K/AKT通路。构建异种移植裸鼠模型,验证ANTXR1在体内的作用。与正常脑组织相比,我们发现ANTXR1在两种细胞系中都过表达。由于shanxr1慢病毒沉默ANTXR1,胶质瘤细胞的生长和迁移能力显著受损。阻断ANTXR1也通过靶向细胞有丝分裂的G2期加速细胞凋亡和延缓细胞周期。体内异种移植模型证实了上述体外发现。进一步研究发现,AKT激活剂可促进ANTXR1下调介导的抗肿瘤作用,而PI3K抑制剂可抑制ANTXR1过表达介导的促肿瘤作用,表明ANTXR1通过调节PI3K/AKT通路在胶质瘤细胞中起作用。ANTXR1可能通过激活PI3K/ akt介导的细胞生长,在胶质瘤发生过程中发挥不可或缺的作用。本研究为靶向ANTXR1作为胶质瘤临床治疗的分子靶点提供了理论依据。
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
Cell Cycle
Cell Cycle 生物-细胞生物学
CiteScore
7.70
自引率
2.30%
发文量
281
审稿时长
1 months
期刊介绍: Cell Cycle is a bi-weekly peer-reviewed journal of high priority research from all areas of cell biology. Cell Cycle covers all topics from yeast to man, from DNA to function, from development to aging, from stem cells to cell senescence, from metabolism to cell death, from cancer to neurobiology, from molecular biology to therapeutics. Our goal is fast publication of outstanding research.
×
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:481959085
Book学术官方微信