通过肿瘤溶解性单纯疱疹病毒 1 型(oHSV-1)研究 HMGB1 过表达对结直肠癌细胞迁移的影响

Q3 Biochemistry, Genetics and Molecular Biology
S. Shayan, A. Arashkia, G. Bahramali, K. Azadmanesh
{"title":"通过肿瘤溶解性单纯疱疹病毒 1 型(oHSV-1)研究 HMGB1 过表达对结直肠癌细胞迁移的影响","authors":"S. Shayan, A. Arashkia, G. Bahramali, K. Azadmanesh","doi":"10.18502/ajmb.v16i2.14863","DOIUrl":null,"url":null,"abstract":"Background: Colorectal Cancer (CRC) represents a significant global health challenge, and its progression, resistance to therapy, and metastasis are strongly influenced by the tumor microenvironment, including factors like hypoxia. This study explores the impact of High Mobility Group Box 1 (HMGB1) overexpression on CRC cell migration, while identifying potential genes associated with this process. \nMethods: To explore this, we developed oncolytic virotherapy, resulting in HSV-HMGB1, an oncolytic Herpes simplex virus that expresses HMGB1. HMGB1 is known its role in cancer progression, particularly in the context of cancer cell migration. \nResults: Contrary to expectations, our scratch assays indicated that HSV-HMGB1 did not significantly induce migration in CRC cells, suggesting that HMGB1 might not directly contribute to this process. Employing microarray analysis, we investigated gene expression changes linked to CRC cell migration, leading to construction of a Protein-Protein Interaction (PPI) network. This network revealed the presence of hub proteins, including as NDRG1, LGALS1, and ANGPTL4, which are recognized for their roles in cancer cell migration. The differential expression of these genes under hypoxic con-ditions was further validated using quantitative RT-PCR, aligning with the findings from our microarray data.  \nConclusion: Our findings emphasize the complex regulation of CRC cell migration, and provides valuable insights into potential molecular mechanisms and pathways. These findings have implications for further research into cancer progression and the development of therapeutic strategies.","PeriodicalId":8669,"journal":{"name":"Avicenna journal of medical biotechnology","volume":null,"pages":null},"PeriodicalIF":0.0000,"publicationDate":"2024-02-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":"{\"title\":\"Investigating the Effects of HMGB1 Overexpression on Colorectal Cancer Cell Migration via Oncolytic Herpes simplex Virus Type 1 (oHSV-1)\",\"authors\":\"S. Shayan, A. Arashkia, G. Bahramali, K. Azadmanesh\",\"doi\":\"10.18502/ajmb.v16i2.14863\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"Background: Colorectal Cancer (CRC) represents a significant global health challenge, and its progression, resistance to therapy, and metastasis are strongly influenced by the tumor microenvironment, including factors like hypoxia. This study explores the impact of High Mobility Group Box 1 (HMGB1) overexpression on CRC cell migration, while identifying potential genes associated with this process. \\nMethods: To explore this, we developed oncolytic virotherapy, resulting in HSV-HMGB1, an oncolytic Herpes simplex virus that expresses HMGB1. HMGB1 is known its role in cancer progression, particularly in the context of cancer cell migration. \\nResults: Contrary to expectations, our scratch assays indicated that HSV-HMGB1 did not significantly induce migration in CRC cells, suggesting that HMGB1 might not directly contribute to this process. Employing microarray analysis, we investigated gene expression changes linked to CRC cell migration, leading to construction of a Protein-Protein Interaction (PPI) network. This network revealed the presence of hub proteins, including as NDRG1, LGALS1, and ANGPTL4, which are recognized for their roles in cancer cell migration. The differential expression of these genes under hypoxic con-ditions was further validated using quantitative RT-PCR, aligning with the findings from our microarray data.  \\nConclusion: Our findings emphasize the complex regulation of CRC cell migration, and provides valuable insights into potential molecular mechanisms and pathways. These findings have implications for further research into cancer progression and the development of therapeutic strategies.\",\"PeriodicalId\":8669,\"journal\":{\"name\":\"Avicenna journal of medical biotechnology\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":0.0000,\"publicationDate\":\"2024-02-06\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Avicenna journal of medical biotechnology\",\"FirstCategoryId\":\"1085\",\"ListUrlMain\":\"https://doi.org/10.18502/ajmb.v16i2.14863\",\"RegionNum\":0,\"RegionCategory\":null,\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q3\",\"JCRName\":\"Biochemistry, Genetics and Molecular Biology\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Avicenna journal of medical biotechnology","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.18502/ajmb.v16i2.14863","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q3","JCRName":"Biochemistry, Genetics and Molecular Biology","Score":null,"Total":0}
引用次数: 0

摘要

背景:结肠直肠癌(CRC)是全球健康面临的重大挑战,其进展、抗药性和转移受肿瘤微环境(包括缺氧等因素)的强烈影响。本研究探讨了高迁移率组框 1(HMGB1)过表达对 CRC 细胞迁移的影响,同时确定了与这一过程相关的潜在基因。方法:为了探讨这一问题,我们开发了溶瘤病毒疗法,产生了表达 HMGB1 的溶瘤单纯疱疹病毒 HSV-HMGB1。众所周知,HMGB1 在癌症进展中起着重要作用,尤其是在癌细胞迁移过程中。结果与预期相反,我们的划痕实验表明 HSV-HMGB1 并没有明显诱导 CRC 细胞的迁移,这表明 HMGB1 可能并不直接参与这一过程。通过微阵列分析,我们研究了与 CRC 细胞迁移相关的基因表达变化,从而构建了一个蛋白-蛋白相互作用(PPI)网络。该网络揭示了枢纽蛋白的存在,包括NDRG1、LGALS1和ANGPTL4,它们在癌细胞迁移中的作用已得到公认。利用定量 RT-PCR 技术进一步验证了这些基因在缺氧条件下的差异表达,这与我们的微阵列数据结果一致。 结论我们的发现强调了 CRC 细胞迁移的复杂调控,并为潜在的分子机制和途径提供了有价值的见解。这些发现对进一步研究癌症进展和制定治疗策略具有重要意义。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
Investigating the Effects of HMGB1 Overexpression on Colorectal Cancer Cell Migration via Oncolytic Herpes simplex Virus Type 1 (oHSV-1)
Background: Colorectal Cancer (CRC) represents a significant global health challenge, and its progression, resistance to therapy, and metastasis are strongly influenced by the tumor microenvironment, including factors like hypoxia. This study explores the impact of High Mobility Group Box 1 (HMGB1) overexpression on CRC cell migration, while identifying potential genes associated with this process. Methods: To explore this, we developed oncolytic virotherapy, resulting in HSV-HMGB1, an oncolytic Herpes simplex virus that expresses HMGB1. HMGB1 is known its role in cancer progression, particularly in the context of cancer cell migration. Results: Contrary to expectations, our scratch assays indicated that HSV-HMGB1 did not significantly induce migration in CRC cells, suggesting that HMGB1 might not directly contribute to this process. Employing microarray analysis, we investigated gene expression changes linked to CRC cell migration, leading to construction of a Protein-Protein Interaction (PPI) network. This network revealed the presence of hub proteins, including as NDRG1, LGALS1, and ANGPTL4, which are recognized for their roles in cancer cell migration. The differential expression of these genes under hypoxic con-ditions was further validated using quantitative RT-PCR, aligning with the findings from our microarray data.  Conclusion: Our findings emphasize the complex regulation of CRC cell migration, and provides valuable insights into potential molecular mechanisms and pathways. These findings have implications for further research into cancer progression and the development of therapeutic strategies.
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
Avicenna journal of medical biotechnology
Avicenna journal of medical biotechnology Biochemistry, Genetics and Molecular Biology-Biotechnology
CiteScore
2.90
自引率
0.00%
发文量
43
×
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