DNA 损伤诱导的凋亡抑制因子通过增强淋巴细胞增强子结合因子 1 的表达,触发胶质瘤的进展和干性。

IF 2.1 Q3 ONCOLOGY World Journal of Oncology Pub Date : 2024-04-01 Epub Date: 2024-03-21 DOI:10.14740/wjon1754
You Lin Chen, Yi Liu, Yan Xu, An Qiang Yang, Gui Jie Chen, Jin Shan Xing, Hong Wei Su, Li Shang Liao
{"title":"DNA 损伤诱导的凋亡抑制因子通过增强淋巴细胞增强子结合因子 1 的表达,触发胶质瘤的进展和干性。","authors":"You Lin Chen, Yi Liu, Yan Xu, An Qiang Yang, Gui Jie Chen, Jin Shan Xing, Hong Wei Su, Li Shang Liao","doi":"10.14740/wjon1754","DOIUrl":null,"url":null,"abstract":"<p><strong>Background: </strong>DNA damage-induced apoptosis suppressor (DDIAS) has recently been discovered to induce cancer progression, but its functions and mechanisms in glioma have not been well studied.</p><p><strong>Methods: </strong>DDIAS expression in glioma tissues was analyzed by the Gene Expression Profiling Interactive Analysis server (GEPIA) and the Gene Expression database of Normal and Tumor tissue 2 (GENT2) databases. The role of DDIAS in glioma progression was studied by short hairpin RNA (shRNA) targeting DDIAS. The effects of DDIAS on glioma cell viability, cell proliferation, invasion, migration, and tumor sphere formation were determined by cell counting kit-8 (CCK-8), EdU, Transwell, tumor spheroid formation, extreme limiting dilution analysis assays <i>in vitro</i> and xenograft model construction <i>in vivo</i>. In addition, RNA sequencing and further functional experiments were used to analyze the DDIAS regulatory mechanism in glioma.</p><p><strong>Results: </strong>We found that DDIAS was highly expressed in glioma and that upregulated DDIAS indicated poor prognosis. Functionally, DDIAS knockdown inhibited glioma cell viability, cell proliferation, invasion and migration <i>in vitro</i> and tumor growth <i>in vivo</i>. In addition, lymphoid enhancer-binding factor 1 (LEF1) was identified as the downstream effector of DDIAS by RNA sequencing. DDIAS downregulation inhibited LEF1 mRNA and protein expression. The expression of DDIAS and LEF1 was positively correlated, and LEF1 overexpression rescued the inhibitory phenotype induced by DDIAS downregulation. We further showed that DDIAS downregulation inhibited cyclin A1, vimentin and the stemness-related factor CD133 and decreased the sphere formation capability, but these features were rescued by upregulation of LEF1.</p><p><strong>Conclusion: </strong>Taken together, these findings suggest that DDIAS promotes glioma progression and stemness by inducing LEF1 expression, proving that DDIAS may be a potential target for the treatment of glioma.</p>","PeriodicalId":46797,"journal":{"name":"World Journal of Oncology","volume":null,"pages":null},"PeriodicalIF":2.1000,"publicationDate":"2024-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10965255/pdf/","citationCount":"0","resultStr":"{\"title\":\"DNA Damage-Induced Apoptosis Suppressor Triggers Progression and Stemness of Glioma by Enhancing Lymphoid Enhancer-Binding Factor 1 Expression.\",\"authors\":\"You Lin Chen, Yi Liu, Yan Xu, An Qiang Yang, Gui Jie Chen, Jin Shan Xing, Hong Wei Su, Li Shang Liao\",\"doi\":\"10.14740/wjon1754\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p><strong>Background: </strong>DNA damage-induced apoptosis suppressor (DDIAS) has recently been discovered to induce cancer progression, but its functions and mechanisms in glioma have not been well studied.</p><p><strong>Methods: </strong>DDIAS expression in glioma tissues was analyzed by the Gene Expression Profiling Interactive Analysis server (GEPIA) and the Gene Expression database of Normal and Tumor tissue 2 (GENT2) databases. The role of DDIAS in glioma progression was studied by short hairpin RNA (shRNA) targeting DDIAS. The effects of DDIAS on glioma cell viability, cell proliferation, invasion, migration, and tumor sphere formation were determined by cell counting kit-8 (CCK-8), EdU, Transwell, tumor spheroid formation, extreme limiting dilution analysis assays <i>in vitro</i> and xenograft model construction <i>in vivo</i>. In addition, RNA sequencing and further functional experiments were used to analyze the DDIAS regulatory mechanism in glioma.</p><p><strong>Results: </strong>We found that DDIAS was highly expressed in glioma and that upregulated DDIAS indicated poor prognosis. Functionally, DDIAS knockdown inhibited glioma cell viability, cell proliferation, invasion and migration <i>in vitro</i> and tumor growth <i>in vivo</i>. In addition, lymphoid enhancer-binding factor 1 (LEF1) was identified as the downstream effector of DDIAS by RNA sequencing. DDIAS downregulation inhibited LEF1 mRNA and protein expression. The expression of DDIAS and LEF1 was positively correlated, and LEF1 overexpression rescued the inhibitory phenotype induced by DDIAS downregulation. We further showed that DDIAS downregulation inhibited cyclin A1, vimentin and the stemness-related factor CD133 and decreased the sphere formation capability, but these features were rescued by upregulation of LEF1.</p><p><strong>Conclusion: </strong>Taken together, these findings suggest that DDIAS promotes glioma progression and stemness by inducing LEF1 expression, proving that DDIAS may be a potential target for the treatment of glioma.</p>\",\"PeriodicalId\":46797,\"journal\":{\"name\":\"World Journal of Oncology\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":2.1000,\"publicationDate\":\"2024-04-01\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10965255/pdf/\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"World Journal of Oncology\",\"FirstCategoryId\":\"1085\",\"ListUrlMain\":\"https://doi.org/10.14740/wjon1754\",\"RegionNum\":0,\"RegionCategory\":null,\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"2024/3/21 0:00:00\",\"PubModel\":\"Epub\",\"JCR\":\"Q3\",\"JCRName\":\"ONCOLOGY\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"World Journal of Oncology","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.14740/wjon1754","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"2024/3/21 0:00:00","PubModel":"Epub","JCR":"Q3","JCRName":"ONCOLOGY","Score":null,"Total":0}
引用次数: 0

摘要

背景:DNA损伤诱导凋亡抑制因子(DDIAS)最近被发现可诱导癌症进展,但其在胶质瘤中的功能和机制尚未得到深入研究:最近发现DNA损伤诱导凋亡抑制因子(DDIAS)可诱导癌症进展,但其在胶质瘤中的功能和机制尚未得到很好的研究:方法:通过基因表达谱交互式分析服务器(GEPIA)和正常与肿瘤组织基因表达数据库2(GENT2)分析了DDIAS在胶质瘤组织中的表达。通过靶向DDIAS的短发夹RNA(shRNA)研究了DDIAS在胶质瘤进展中的作用。通过体外的细胞计数试剂盒-8(CCK-8)、EdU、Transwell、肿瘤球形成、极度稀释分析实验和体内的异种移植模型构建,研究了DDIAS对胶质瘤细胞活力、细胞增殖、侵袭、迁移和肿瘤球形成的影响。此外,我们还利用RNA测序和进一步的功能实验分析了DDIAS在胶质瘤中的调控机制:结果:我们发现DDIAS在胶质瘤中高表达,DDIAS上调预示着不良预后。在功能上,DDIAS敲除可抑制胶质瘤细胞活力、体外细胞增殖、侵袭和迁移以及体内肿瘤生长。此外,通过RNA测序发现,淋巴增强子结合因子1(LEF1)是DDIAS的下游效应因子。DDIAS 的下调抑制了 LEF1 mRNA 和蛋白的表达。DDIAS和LEF1的表达呈正相关,LEF1的过表达可挽救DDIAS下调所诱导的抑制表型。我们进一步发现,DDIAS下调会抑制细胞周期蛋白A1、波形蛋白和干性相关因子CD133,并降低球形成能力,但LEF1上调可挽救这些特征:综上所述,这些研究结果表明,DDIAS通过诱导LEF1的表达促进胶质瘤的进展和干性,证明DDIAS可能是治疗胶质瘤的潜在靶点。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
查看原文
分享 分享
微信好友 朋友圈 QQ好友 复制链接
本刊更多论文
DNA Damage-Induced Apoptosis Suppressor Triggers Progression and Stemness of Glioma by Enhancing Lymphoid Enhancer-Binding Factor 1 Expression.

Background: DNA damage-induced apoptosis suppressor (DDIAS) has recently been discovered to induce cancer progression, but its functions and mechanisms in glioma have not been well studied.

Methods: DDIAS expression in glioma tissues was analyzed by the Gene Expression Profiling Interactive Analysis server (GEPIA) and the Gene Expression database of Normal and Tumor tissue 2 (GENT2) databases. The role of DDIAS in glioma progression was studied by short hairpin RNA (shRNA) targeting DDIAS. The effects of DDIAS on glioma cell viability, cell proliferation, invasion, migration, and tumor sphere formation were determined by cell counting kit-8 (CCK-8), EdU, Transwell, tumor spheroid formation, extreme limiting dilution analysis assays in vitro and xenograft model construction in vivo. In addition, RNA sequencing and further functional experiments were used to analyze the DDIAS regulatory mechanism in glioma.

Results: We found that DDIAS was highly expressed in glioma and that upregulated DDIAS indicated poor prognosis. Functionally, DDIAS knockdown inhibited glioma cell viability, cell proliferation, invasion and migration in vitro and tumor growth in vivo. In addition, lymphoid enhancer-binding factor 1 (LEF1) was identified as the downstream effector of DDIAS by RNA sequencing. DDIAS downregulation inhibited LEF1 mRNA and protein expression. The expression of DDIAS and LEF1 was positively correlated, and LEF1 overexpression rescued the inhibitory phenotype induced by DDIAS downregulation. We further showed that DDIAS downregulation inhibited cyclin A1, vimentin and the stemness-related factor CD133 and decreased the sphere formation capability, but these features were rescued by upregulation of LEF1.

Conclusion: Taken together, these findings suggest that DDIAS promotes glioma progression and stemness by inducing LEF1 expression, proving that DDIAS may be a potential target for the treatment of glioma.

求助全文
通过发布文献求助,成功后即可免费获取论文全文。 去求助
来源期刊
CiteScore
6.10
自引率
15.40%
发文量
37
期刊介绍: World Journal of Oncology, bimonthly, publishes original contributions describing basic research and clinical investigation of cancer, on the cellular, molecular, prevention, diagnosis, therapy and prognosis aspects. The submissions can be basic research or clinical investigation oriented. This journal welcomes those submissions focused on the clinical trials of new treatment modalities for cancer, and those submissions focused on molecular or cellular research of the oncology pathogenesis. Case reports submitted for consideration of publication should explore either a novel genomic event/description or a new safety signal from an oncolytic agent. The areas of interested manuscripts are these disciplines: tumor immunology and immunotherapy; cancer molecular pharmacology and chemotherapy; drug sensitivity and resistance; cancer epidemiology; clinical trials; cancer pathology; radiobiology and radiation oncology; solid tumor oncology; hematological malignancies; surgical oncology; pediatric oncology; molecular oncology and cancer genes; gene therapy; cancer endocrinology; cancer metastasis; prevention and diagnosis of cancer; other cancer related subjects. The types of manuscripts accepted are original article, review, editorial, short communication, case report, letter to the editor, book review.
期刊最新文献
Hereditary Gastric Cancer Is Linked With Hereditary Breast and Ovarian Cancer. High Probability of Lynch Syndrome Among Colorectal Cancer Patients Is Associated With Higher Occurrence of KRAS and PIK3CA Mutations. Identification and Validation of a Novel Tertiary Lymphoid Structures-Related Prognostic Gene Signature in Hepatocellular Carcinoma. Impact of Lymph Node Dissection for Patients With Clinically Node-Negative Intrahepatic Cholangiocarcinoma: A Multicenter Cohort Study. Increased Vitamin C Intake Is Associated With Decreased Pancreatic Cancer Risk.
×
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
现在去查看 取消
×
提示
确定
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
已复制链接
已复制链接
快去分享给好友吧!
我知道了
×
扫码分享
扫码分享
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 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 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1