Suppr超能文献

20(S)-人参皂苷Rg3通过抑制E2F-DP二聚化来抑制胃癌细胞增殖。

20(S)-ginsenoside Rg3 suppresses gastric cancer cell proliferation by inhibiting E2F-DP dimerization.

作者信息

Li Fuqiang, Cai Chengyu, Wang Fei, Zhang Na, Zhao Qingzhi, Chen Yuyang, Cui Xueli, Wang Siyang, Zhang Wenjie, Liu Da, Cai Yong, Jin Jingji

机构信息

School of Life Sciences, Jilin University, 2699 Qianjin Street, Chaoyang District, Changchun, Jilin 130012, China; School of Pharmacy, Changchun University of Chinese Medicine, Boshuo Road, Jingyue Development Zone, Changchun, Jilin 130117, China.

School of Life Sciences, Jilin University, 2699 Qianjin Street, Chaoyang District, Changchun, Jilin 130012, China.

出版信息

Phytomedicine. 2025 Jun;141:156740. doi: 10.1016/j.phymed.2025.156740. Epub 2025 Apr 8.

Abstract

BACKGROUND

Gastric cancer (GC) is a common and aggressive malignancy, with treatment options often limited by drug resistance and the adverse effects of targeted therapies and immunotherapy. Ginsenoside Rg3, a bioactive compound derived from ginseng, has shown promise in inhibiting the growth of various tumor types, including GC. However, the molecular mechanisms underlying its therapeutic effects against GC remain insufficiently understood.

OBJECTIVE

This study aimed to elucidate the molecular mechanisms underlying the anti-cancer effects of ginsenoside Rg3 against GC.

METHODS

To explore the molecular mechanisms underlying Rg3's anti-GC effects, RNA sequencing (RNA-Seq) was conducted to identify potential Rg3-regulated targets. The interaction between Rg3 and E2F was analyzed using several approaches, including the cellular thermal shift assay (CETSA), Rg3-PEGA pull-down, Rg3 pull-down protein mass spectrometry, and 3D molecular docking. Additionally, quantitative reverse transcription PCR (qRT-PCR), co-transfection followed by immunoprecipitation, Western blotting, flow cytometry, Annexin V-FITC staining, Hoechst staining, and luciferase reporter assays were employed to elucidate the molecular effects of Rg3. The inhibitory effect of Rg3 on GC proliferation was assessed through colony formation assays in vitro and tumor xenograft experiments in C57BL/6 mice in vivo.

RESULTS

Rg3-mediated gene expression profiling in GC cells revealed several transcription factors, including E2F, and biological processes potentially influenced by Rg3. Consistent with these findings, Rg3 suppressed E2F expression and impeded GC cell proliferation by inducing G1/S cell cycle arrest, reducing cell growth both in vitro and in vivo, enhancing apoptosis, and inhibiting CDC6 transactivation. CETSA and Rg3 pull-down assays confirmed an interaction between Rg3 and E2F. Additionally, 3D molecular docking analysis demonstrated that Rg3 binds with high affinity to E2F at the heterodimeric domain via hydrogen bonding, potentially disrupting E2F-DP heterodimer formation and subsequently inhibiting cell cycle gene expression. In agreement with this, Rg3-treated GC cells exhibited reduced expression of cyclin D1, CDK4, cyclin A, CDK1, and CDK2. Moreover, Rg3 activated the tumor suppressors p53 and p21, further inhibiting RB phosphorylation by suppressing cyclin/CDK activity, thereby blocking transcription of G1/S transition-related genes.

CONCLUSION

This study provides the first evidence that Rg3 directly binds to E2F proteins, disrupting E2F-DP heterodimer formation and inhibiting the transcription of E2F-DP-regulated target genes. Furthermore, Rg3 activates the p53-p21 pathway while suppressing the cyclin/CDK-RB signaling pathway, effectively inhibiting cancer cell proliferation. These findings highlight a potential therapeutic strategy for developing small molecules structurally similar to Rg3 to target tumors with high E2F expression.

摘要

背景

胃癌(GC)是一种常见且侵袭性强的恶性肿瘤,其治疗选择常常受到耐药性以及靶向治疗和免疫治疗不良反应的限制。人参皂苷Rg3是一种从人参中提取的生物活性化合物,已显示出在抑制包括胃癌在内的多种肿瘤类型生长方面的潜力。然而,其对胃癌治疗作用的分子机制仍未得到充分了解。

目的

本研究旨在阐明人参皂苷Rg3对胃癌抗癌作用的分子机制。

方法

为探究Rg3抗胃癌作用的分子机制,进行了RNA测序(RNA-Seq)以鉴定潜在的Rg3调控靶点。使用多种方法分析Rg3与E2F之间的相互作用,包括细胞热位移分析(CETSA)、Rg3-聚乙二醇化下拉实验、Rg3下拉蛋白质质谱分析和三维分子对接。此外,采用定量逆转录PCR(qRT-PCR)、共转染后免疫沉淀、蛋白质免疫印迹法、流式细胞术、膜联蛋白V-异硫氰酸荧光素染色、Hoechst染色和荧光素酶报告基因检测来阐明Rg3的分子效应。通过体外集落形成实验和体内C57BL/6小鼠肿瘤异种移植实验评估Rg3对胃癌细胞增殖的抑制作用。

结果

Rg3介导的胃癌细胞基因表达谱分析揭示了几种转录因子,包括E2F,以及可能受Rg3影响的生物学过程。与这些发现一致,Rg3通过诱导G1/S期细胞周期阻滞抑制E2F表达并阻碍胃癌细胞增殖,在体外和体内均减少细胞生长,增强细胞凋亡,并抑制CDC6反式激活。CETSA和Rg3下拉实验证实了Rg3与E2F之间的相互作用。此外,三维分子对接分析表明,Rg3通过氢键在异二聚体结构域与E2F高亲和力结合,可能破坏E2F-DP异二聚体的形成,进而抑制细胞周期基因表达。与此一致,用Rg3处理的胃癌细胞中细胞周期蛋白D1、细胞周期蛋白依赖性激酶4(CDK4)、细胞周期蛋白A、细胞周期蛋白依赖性激酶1(CDK1)和细胞周期蛋白依赖性激酶2(CDK2)的表达降低。此外,Rg3激活肿瘤抑制因子p53和p21,通过抑制细胞周期蛋白/细胞周期蛋白依赖性激酶活性进一步抑制RB磷酸化,从而阻断G1/S期转换相关基因的转录。

结论

本研究首次证明Rg3直接与人E2F蛋白结合,破坏E2F-DP异二聚体的形成并抑制E2F-DP调控的靶基因转录。此外,Rg3激活p53-p21通路,同时抑制细胞周期蛋白/细胞周期蛋白依赖性激酶-RB信号通路,有效抑制癌细胞增殖。这些发现突出了一种潜在的治疗策略,即开发与Rg3结构相似的小分子来靶向高表达E2F的肿瘤。

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