Investigating the mechanisms by which low NAT1 expression in tumor cells contributes to chemo-resistance in colorectal cancer.

IF 4.8 2区 医学 Q1 GENETICS & HEREDITY
Zheng Yuan, Kai Fang, Xinsheng Miao, Yan Zhang, Menghui Gu, Wei Xu, Hao Li, Dawei Zhu, Jiahui Zhou, Jian Sun, Xinhua Gu
{"title":"Investigating the mechanisms by which low NAT1 expression in tumor cells contributes to chemo-resistance in colorectal cancer.","authors":"Zheng Yuan, Kai Fang, Xinsheng Miao, Yan Zhang, Menghui Gu, Wei Xu, Hao Li, Dawei Zhu, Jiahui Zhou, Jian Sun, Xinhua Gu","doi":"10.1186/s13148-025-01882-4","DOIUrl":null,"url":null,"abstract":"<p><strong>Background: </strong>In the therapeutic landscape of colorectal cancer (CRC), chemo-resistance poses a significant and prevalent obstacle that complicates treatment efficacy and patient outcomes. Over time, cancer cells can develop mechanisms to resist the toxic effects of chemo-therapy drugs, leading to reduced sensitivity or complete insensitivity to these agents. The enzyme Arylamine N-acetyltransferase 1 (NAT1) has emerged as a promising target in strategies aimed at overcoming this challenge. NAT1 is involved in the metabolism of various xenobiotics, including some chemotherapeutic agents. Understanding the complex interactions between NAT1 and chemotherapeutic agents, as well as the molecular mechanisms underlying chemo-resistance, is crucial for the development of novel therapeutic approaches.</p><p><strong>Objective: </strong>This study aimed to assess the role of NAT1 in mediating chemo-resistance in CRC, with the goal of identifying novel strategies to overcome this clinical challenge.</p><p><strong>Methods: </strong>We conducted a comprehensive analysis using various bioinformatics tools and in vitro experiments to evaluate the effect of NAT1 expression on chemo-resistance in CRC. Furthermore, we employed a multi-omics approach, including metabolomics and next-generation sequencing, to uncover the mechanisms by which NAT1 influences chemo-resistance. Additionally, we utilized single-cell RNA sequencing (scRNA-seq), the Cellchat assay, and western blot to explore the intercellular communication between tumor and endothelial cells in the context of anti-PD-1 therapy and NAT1's impact.</p><p><strong>Results: </strong>Our study reveals that decreased NAT1 expression in CRC tumor tissues, relative to adjacent normal tissues, is significantly associated with a poorer patient prognosis. Experimental data indicate that silencing NAT1 in CaCO2 and HCT116 cell lines results in heightened resistance to five chemotherapeutic agents: vinblastine, docetaxel, gemcitabine, vincristine, and daporinad. Additionally, NAT1 silencing increases the proportion of LGR5<sup>+</sup> cells, which are known to be chemo-resistant. Our research further revealed that exposure to these five drugs induces a decrease in NAT1 expression within CRC cells. Mechanistic insights show that NAT1 knockdown triggers a metabolic reprogramming in CRC cells, shifting from oxidative phosphorylation and the tricarboxylic acid cycle to a preference for glycolysis. Furthermore, silencing of NAT1 in CRC cells leads to an up-regulation of VEGFA expression. Notably, the application of anti-PD-1 therapy was demonstrated to significantly disrupt the VEGFA-VEGFR axis signaling, an interaction critical between CRC cells and endothelial cells. This discovery underscores the potential of targeting the VEGFA pathway as a therapeutic approach to mitigate the adverse effects associated with NAT1 down-regulation in CRC.</p><p><strong>Conclusion: </strong>Our study underscores the multifaceted role of NAT1 in modulating chemo-sensitivity, cellular metabolism, and angiogenesis in CRC. These findings position NAT1 as a compelling candidate for a biomarker and a potential therapeutic target, offering new avenues for CRC management.</p>","PeriodicalId":10366,"journal":{"name":"Clinical Epigenetics","volume":"17 1","pages":"77"},"PeriodicalIF":4.8000,"publicationDate":"2025-05-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12053866/pdf/","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Clinical Epigenetics","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1186/s13148-025-01882-4","RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"GENETICS & HEREDITY","Score":null,"Total":0}
引用次数: 0

Abstract

Background: In the therapeutic landscape of colorectal cancer (CRC), chemo-resistance poses a significant and prevalent obstacle that complicates treatment efficacy and patient outcomes. Over time, cancer cells can develop mechanisms to resist the toxic effects of chemo-therapy drugs, leading to reduced sensitivity or complete insensitivity to these agents. The enzyme Arylamine N-acetyltransferase 1 (NAT1) has emerged as a promising target in strategies aimed at overcoming this challenge. NAT1 is involved in the metabolism of various xenobiotics, including some chemotherapeutic agents. Understanding the complex interactions between NAT1 and chemotherapeutic agents, as well as the molecular mechanisms underlying chemo-resistance, is crucial for the development of novel therapeutic approaches.

Objective: This study aimed to assess the role of NAT1 in mediating chemo-resistance in CRC, with the goal of identifying novel strategies to overcome this clinical challenge.

Methods: We conducted a comprehensive analysis using various bioinformatics tools and in vitro experiments to evaluate the effect of NAT1 expression on chemo-resistance in CRC. Furthermore, we employed a multi-omics approach, including metabolomics and next-generation sequencing, to uncover the mechanisms by which NAT1 influences chemo-resistance. Additionally, we utilized single-cell RNA sequencing (scRNA-seq), the Cellchat assay, and western blot to explore the intercellular communication between tumor and endothelial cells in the context of anti-PD-1 therapy and NAT1's impact.

Results: Our study reveals that decreased NAT1 expression in CRC tumor tissues, relative to adjacent normal tissues, is significantly associated with a poorer patient prognosis. Experimental data indicate that silencing NAT1 in CaCO2 and HCT116 cell lines results in heightened resistance to five chemotherapeutic agents: vinblastine, docetaxel, gemcitabine, vincristine, and daporinad. Additionally, NAT1 silencing increases the proportion of LGR5+ cells, which are known to be chemo-resistant. Our research further revealed that exposure to these five drugs induces a decrease in NAT1 expression within CRC cells. Mechanistic insights show that NAT1 knockdown triggers a metabolic reprogramming in CRC cells, shifting from oxidative phosphorylation and the tricarboxylic acid cycle to a preference for glycolysis. Furthermore, silencing of NAT1 in CRC cells leads to an up-regulation of VEGFA expression. Notably, the application of anti-PD-1 therapy was demonstrated to significantly disrupt the VEGFA-VEGFR axis signaling, an interaction critical between CRC cells and endothelial cells. This discovery underscores the potential of targeting the VEGFA pathway as a therapeutic approach to mitigate the adverse effects associated with NAT1 down-regulation in CRC.

Conclusion: Our study underscores the multifaceted role of NAT1 in modulating chemo-sensitivity, cellular metabolism, and angiogenesis in CRC. These findings position NAT1 as a compelling candidate for a biomarker and a potential therapeutic target, offering new avenues for CRC management.

探讨肿瘤细胞NAT1低表达导致结直肠癌耐药的机制。
背景:在结直肠癌(CRC)的治疗领域,化疗耐药是一个重要且普遍的障碍,使治疗效果和患者预后复杂化。随着时间的推移,癌细胞可以发展出抵抗化疗药物毒性作用的机制,导致对这些药物的敏感性降低或完全不敏感。芳胺n -乙酰转移酶1 (NAT1)已成为克服这一挑战的一个有希望的靶点。NAT1参与多种外源药物的代谢,包括一些化疗药物。了解NAT1与化疗药物之间复杂的相互作用,以及化疗耐药的分子机制,对于开发新的治疗方法至关重要。目的:本研究旨在评估NAT1在介导结直肠癌化疗耐药中的作用,以确定克服这一临床挑战的新策略。方法:利用多种生物信息学工具和体外实验进行综合分析,评估NAT1表达对结直肠癌耐药的影响。此外,我们采用多组学方法,包括代谢组学和下一代测序,来揭示NAT1影响化疗耐药性的机制。此外,我们利用单细胞RNA测序(scRNA-seq)、Cellchat测定和western blot来探索抗pd -1治疗和NAT1影响下肿瘤和内皮细胞之间的细胞间通讯。结果:我们的研究表明,相对于邻近的正常组织,CRC肿瘤组织中NAT1表达的降低与患者预后较差显著相关。实验数据表明,沉默CaCO2和HCT116细胞系中的NAT1可导致对五种化疗药物(长春碱、多西他赛、吉西他滨、长春新碱和daporinad)的耐药性增强。此外,NAT1沉默增加了LGR5+细胞的比例,这是已知的耐药细胞。我们的研究进一步表明,暴露于这五种药物可诱导CRC细胞内NAT1表达降低。机制研究表明,NAT1敲低触发CRC细胞的代谢重编程,从氧化磷酸化和三羧酸循环转向糖酵解。此外,CRC细胞中NAT1的沉默导致VEGFA表达上调。值得注意的是,应用抗pd -1治疗被证明可以显著破坏VEGFA-VEGFR轴信号,这是CRC细胞和内皮细胞之间至关重要的相互作用。这一发现强调了靶向VEGFA通路作为减轻CRC中NAT1下调相关不良反应的治疗方法的潜力。结论:我们的研究强调了NAT1在调节结直肠癌的化疗敏感性、细胞代谢和血管生成中的多方面作用。这些发现将NAT1定位为一个令人信服的生物标志物和潜在的治疗靶点,为结直肠癌的治疗提供了新的途径。
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
5.30%
发文量
150
期刊介绍: Clinical Epigenetics, the official journal of the Clinical Epigenetics Society, is an open access, peer-reviewed journal that encompasses all aspects of epigenetic principles and mechanisms in relation to human disease, diagnosis and therapy. Clinical trials and research in disease model organisms are particularly welcome.
×
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