Xiya Chen , Jingjing He , Ge Ge , Shuangshuang Ma , Zixuan Chen , Lan Zhang , Feng Gao , Jin Zhang
{"title":"Eriocalyxin B在三阴性乳腺癌中通过抑制SIRT3诱导铁下垂","authors":"Xiya Chen , Jingjing He , Ge Ge , Shuangshuang Ma , Zixuan Chen , Lan Zhang , Feng Gao , Jin Zhang","doi":"10.1016/j.phymed.2025.157257","DOIUrl":null,"url":null,"abstract":"<div><h3>Background</h3><div>Triple-negative breast cancer (TNBC), a highly aggressive and heterogeneous breast cancer subtype, remains clinically challenging due to the paucity of actionable therapeutic targets. The unique metabolic landscape of TNBC, characterized by elevated iron and lipid levels, renders it particularly susceptible to ferroptosis, positioning this cell death modality as a promising therapeutic avenue. Eriocalyxin B (Eri B), a natural diterpenoid, exhibits multifaceted anticancer properties that warrant further investigation.</div></div><div><h3>Object</h3><div>To elucidate the therapeutic mechanism of Eri B-induced ferroptosis in TNBC.</div></div><div><h3>Method</h3><div>In this study, the anti-proliferative effects of Eri B on TNBC cells were evaluated using cell colony formation assays, three-dimensional tumor sphere formation assays, EdU incorporation assays, and analysis of the proliferation marker Ki-67. The anti-metastatic potential of Eri B was assessed through wound healing assays, transwell migration assays, immunofluorescence staining, and western blot analysis. Furthermore, RNA sequencing (RNA-seq), flow cytometry, transmission electron microscopy, and western blotting demonstrated that Eri B induces ferroptosis in TNBC cells. Molecular docking, surface plasmon resonance (SPR), and cellular thermal shift assays (CETSA) provided evidence of a direct interaction between Eri B and signal transducer and activator of transcription 3 (STAT3). Moreover, the efficacy and underlying mechanism of Eri B were further validated through SIRT3 overexpression experiments and by employing a STAT3 activator, colivelin. In <em>vivo</em> anti-TNBC activity was confirmed using a xenografted tumor model combined with bioluminescence imaging.</div></div><div><h3>Result</h3><div>For the first time, we demonstrate that Eri B exhibits significant antitumor activity both in <em>vitro</em> and in <em>vivo</em> through the induction of ferroptosis. RNA-seq analysis enabled us to identify and confirm that Eri B induces ferroptosis and oxidative stress by inhibiting sirtuin 3 (SIRT3), which subsequently modulates the nuclear factor erythroid 2-related factor 2 (NRF2)-glutathione peroxidase 4 (GPX4) signaling pathway. Moreover, our findings reveal that Eri B suppresses <em>SIRT3</em> transcription by directly targeting STAT3, thereby regulating the SIRT3-sterol regulatory element-binding transcription factor 1 (SREBP1)-peroxisome proliferator-activated receptor α (PPARα) -mediated fatty acid metabolism and cellular redox homeostasis.</div></div><div><h3>Conclusion</h3><div>In summary, our findings suggest that Eri B functions as a novel inducer of ferroptosis in TNBC, demonstrating potential therapeutic value through the inhibition of SIRT3 in TNBC.</div></div>","PeriodicalId":20212,"journal":{"name":"Phytomedicine","volume":"148 ","pages":"Article 157257"},"PeriodicalIF":8.3000,"publicationDate":"2025-09-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":"{\"title\":\"Eriocalyxin B induces ferroptosis through SIRT3 inhibition in triple-negative breast cancer\",\"authors\":\"Xiya Chen , Jingjing He , Ge Ge , Shuangshuang Ma , Zixuan Chen , Lan Zhang , Feng Gao , Jin Zhang\",\"doi\":\"10.1016/j.phymed.2025.157257\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<div><h3>Background</h3><div>Triple-negative breast cancer (TNBC), a highly aggressive and heterogeneous breast cancer subtype, remains clinically challenging due to the paucity of actionable therapeutic targets. The unique metabolic landscape of TNBC, characterized by elevated iron and lipid levels, renders it particularly susceptible to ferroptosis, positioning this cell death modality as a promising therapeutic avenue. Eriocalyxin B (Eri B), a natural diterpenoid, exhibits multifaceted anticancer properties that warrant further investigation.</div></div><div><h3>Object</h3><div>To elucidate the therapeutic mechanism of Eri B-induced ferroptosis in TNBC.</div></div><div><h3>Method</h3><div>In this study, the anti-proliferative effects of Eri B on TNBC cells were evaluated using cell colony formation assays, three-dimensional tumor sphere formation assays, EdU incorporation assays, and analysis of the proliferation marker Ki-67. The anti-metastatic potential of Eri B was assessed through wound healing assays, transwell migration assays, immunofluorescence staining, and western blot analysis. Furthermore, RNA sequencing (RNA-seq), flow cytometry, transmission electron microscopy, and western blotting demonstrated that Eri B induces ferroptosis in TNBC cells. Molecular docking, surface plasmon resonance (SPR), and cellular thermal shift assays (CETSA) provided evidence of a direct interaction between Eri B and signal transducer and activator of transcription 3 (STAT3). Moreover, the efficacy and underlying mechanism of Eri B were further validated through SIRT3 overexpression experiments and by employing a STAT3 activator, colivelin. In <em>vivo</em> anti-TNBC activity was confirmed using a xenografted tumor model combined with bioluminescence imaging.</div></div><div><h3>Result</h3><div>For the first time, we demonstrate that Eri B exhibits significant antitumor activity both in <em>vitro</em> and in <em>vivo</em> through the induction of ferroptosis. RNA-seq analysis enabled us to identify and confirm that Eri B induces ferroptosis and oxidative stress by inhibiting sirtuin 3 (SIRT3), which subsequently modulates the nuclear factor erythroid 2-related factor 2 (NRF2)-glutathione peroxidase 4 (GPX4) signaling pathway. Moreover, our findings reveal that Eri B suppresses <em>SIRT3</em> transcription by directly targeting STAT3, thereby regulating the SIRT3-sterol regulatory element-binding transcription factor 1 (SREBP1)-peroxisome proliferator-activated receptor α (PPARα) -mediated fatty acid metabolism and cellular redox homeostasis.</div></div><div><h3>Conclusion</h3><div>In summary, our findings suggest that Eri B functions as a novel inducer of ferroptosis in TNBC, demonstrating potential therapeutic value through the inhibition of SIRT3 in TNBC.</div></div>\",\"PeriodicalId\":20212,\"journal\":{\"name\":\"Phytomedicine\",\"volume\":\"148 \",\"pages\":\"Article 157257\"},\"PeriodicalIF\":8.3000,\"publicationDate\":\"2025-09-11\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Phytomedicine\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://www.sciencedirect.com/science/article/pii/S0944711325008967\",\"RegionNum\":1,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q1\",\"JCRName\":\"CHEMISTRY, MEDICINAL\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Phytomedicine","FirstCategoryId":"3","ListUrlMain":"https://www.sciencedirect.com/science/article/pii/S0944711325008967","RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"CHEMISTRY, MEDICINAL","Score":null,"Total":0}
Eriocalyxin B induces ferroptosis through SIRT3 inhibition in triple-negative breast cancer
Background
Triple-negative breast cancer (TNBC), a highly aggressive and heterogeneous breast cancer subtype, remains clinically challenging due to the paucity of actionable therapeutic targets. The unique metabolic landscape of TNBC, characterized by elevated iron and lipid levels, renders it particularly susceptible to ferroptosis, positioning this cell death modality as a promising therapeutic avenue. Eriocalyxin B (Eri B), a natural diterpenoid, exhibits multifaceted anticancer properties that warrant further investigation.
Object
To elucidate the therapeutic mechanism of Eri B-induced ferroptosis in TNBC.
Method
In this study, the anti-proliferative effects of Eri B on TNBC cells were evaluated using cell colony formation assays, three-dimensional tumor sphere formation assays, EdU incorporation assays, and analysis of the proliferation marker Ki-67. The anti-metastatic potential of Eri B was assessed through wound healing assays, transwell migration assays, immunofluorescence staining, and western blot analysis. Furthermore, RNA sequencing (RNA-seq), flow cytometry, transmission electron microscopy, and western blotting demonstrated that Eri B induces ferroptosis in TNBC cells. Molecular docking, surface plasmon resonance (SPR), and cellular thermal shift assays (CETSA) provided evidence of a direct interaction between Eri B and signal transducer and activator of transcription 3 (STAT3). Moreover, the efficacy and underlying mechanism of Eri B were further validated through SIRT3 overexpression experiments and by employing a STAT3 activator, colivelin. In vivo anti-TNBC activity was confirmed using a xenografted tumor model combined with bioluminescence imaging.
Result
For the first time, we demonstrate that Eri B exhibits significant antitumor activity both in vitro and in vivo through the induction of ferroptosis. RNA-seq analysis enabled us to identify and confirm that Eri B induces ferroptosis and oxidative stress by inhibiting sirtuin 3 (SIRT3), which subsequently modulates the nuclear factor erythroid 2-related factor 2 (NRF2)-glutathione peroxidase 4 (GPX4) signaling pathway. Moreover, our findings reveal that Eri B suppresses SIRT3 transcription by directly targeting STAT3, thereby regulating the SIRT3-sterol regulatory element-binding transcription factor 1 (SREBP1)-peroxisome proliferator-activated receptor α (PPARα) -mediated fatty acid metabolism and cellular redox homeostasis.
Conclusion
In summary, our findings suggest that Eri B functions as a novel inducer of ferroptosis in TNBC, demonstrating potential therapeutic value through the inhibition of SIRT3 in TNBC.
期刊介绍:
Phytomedicine is a therapy-oriented journal that publishes innovative studies on the efficacy, safety, quality, and mechanisms of action of specified plant extracts, phytopharmaceuticals, and their isolated constituents. This includes clinical, pharmacological, pharmacokinetic, and toxicological studies of herbal medicinal products, preparations, and purified compounds with defined and consistent quality, ensuring reproducible pharmacological activity. Founded in 1994, Phytomedicine aims to focus and stimulate research in this field and establish internationally accepted scientific standards for pharmacological studies, proof of clinical efficacy, and safety of phytomedicines.