Suppr超能文献

研究肿瘤细胞中NAT1低表达导致结直肠癌化疗耐药的机制。

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

作者信息

Yuan Zheng, Fang Kai, Miao Xinsheng, Zhang Yan, Gu Menghui, Xu Wei, Li Hao, Zhu Dawei, Zhou Jiahui, Sun Jian, Gu Xinhua

机构信息

Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School of Nanjing Medical University, Suzhou, China.

College of Basic Medical Sciences, Suzhou University, Suzhou, China.

出版信息

Clin Epigenetics. 2025 May 6;17(1):77. doi: 10.1186/s13148-025-01882-4.

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.

摘要

背景

在结直肠癌(CRC)的治疗领域,化疗耐药是一个重大且普遍存在的障碍,它使治疗效果和患者预后变得复杂。随着时间的推移,癌细胞会形成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-乙酰基转移酶1(NAT1)已成为旨在克服这一挑战的策略中有前景的靶点。NAT1参与多种外源性物质的代谢,包括一些化疗药物。了解NAT1与化疗药物之间的复杂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CRC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

我们使用各种生物信息学工具和体外实验进行了全面分析,以评估NAT1表达对CRC化疗耐药的影响。此外,我们采用了多组学方法,包括代谢组学和下一代测序,以揭示NAT1影响化疗耐药的机制。此外,我们利用单细胞RNA测序(scRNA-seq)、Cellchat分析和蛋白质印迹法,探讨在抗PD-1治疗背景下肿瘤细胞与内皮细胞之间的细胞间通讯以及NAT1的影响。

结果

我们的研究表明,与相邻正常组织相比,CRC肿瘤组织中NAT1表达降低与患者预后较差显著相关。实验数据表明,在CaCO2和HCT116细胞系中沉默NAT1会导致对五种化疗药物(长春碱、多西他赛、吉西他滨、长春新碱和达泊拉定)的耐药性增强。此外,NAT1沉默增加了已知具有化疗耐药性的LGR5细胞的比例。我们的研究进一步表明,暴露于这五种药物会导致CRC细胞内NAT1表达降低。机制研究表明,NAT1基因敲低会引发CRC细胞的代谢重编程,从氧化磷酸化和三羧酸循环转变为偏好糖酵解。此外,CRC细胞中NAT1的沉默导致VEGFA表达上调。值得注意的是,抗PD-1治疗的应用被证明会显著破坏VEGFA-VEGFR轴信号传导,这是CRC细胞与内皮细胞之间的关键相互作用。这一发现强调了靶向VEGFA途径作为一种治疗方法来减轻CRC中与NAT1下调相关的不良反应的潜力。

结论

我们的研究强调了NAT1在调节CRC化疗敏感性、细胞代谢和血管生成方面的多方面作用。这些发现使NAT1成为一个有吸引力的生物标志物候选者和潜在的治疗靶点,为CRC的管理提供了新的途径。

https://cdn.ncbi.nlm.nih.gov/pmc/blobs/1ed0/12053866/f66429abaed7/13148_2025_1882_Fig1_HTML.jpg

文献AI研究员

20分钟写一篇综述,助力文献阅读效率提升50倍。

立即体验

用中文搜PubMed

大模型驱动的PubMed中文搜索引擎

马上搜索

文档翻译

学术文献翻译模型,支持多种主流文档格式。

立即体验