Wan Shu , Guanxiao Chen , Jun Zhang, Kejun Dong, Ting Zhou, Shuangshuang Cheng, Tangansu Zhang, Jiarui Zhang, Haojia Li, Yuwei Yao, Shuyang Yu, Yan Liu, Xing Zhou, Xiaoyu Shen, Hongbo Wang
{"title":"Bavachinin通过激活TLR4/STING轴依赖性PANoptosis发挥抗肿瘤作用,协同增强子宫内膜癌的化疗敏感性。","authors":"Wan Shu , Guanxiao Chen , Jun Zhang, Kejun Dong, Ting Zhou, Shuangshuang Cheng, Tangansu Zhang, Jiarui Zhang, Haojia Li, Yuwei Yao, Shuyang Yu, Yan Liu, Xing Zhou, Xiaoyu Shen, Hongbo Wang","doi":"10.1016/j.bcp.2025.117321","DOIUrl":null,"url":null,"abstract":"<div><div>The incidence and mortality rates of endometrial cancer (EC), a malignancy originating from endometrium, have been increasing globally. Currently, there are no effective therapeutic options for patients with recurrent, chemoresistant, and metastatic forms of this disease. Through compound library screening, we identified that bavachinin (BVC) has a killing effect on EC cells. BVC is a bioactive small molecule with potential pharmacological effects derived from the traditional Chinese herb <em>Proralea corylifolia L</em>, but the specific mechanisms are unclear. We first discovered that BVC induces ZBP1 (Z-DNA binding protein 1)-mediated PANoptosis in EC cells, characterized by activating of pyroptosis, apoptosis, and necroptosis. BVC promoted reactive oxygen species (ROS) production, mitochondrial membrane potential (MMP, ΔΨm) collapse, and ATM-CHK2 mediated DNA damage, which activated cGAS-STING pathway in EC cells. Mechanistically, network pharmacology, molecular docking, cellular thermal shift assays (CESTA), and drug affinity responsive target stability (DARTS) experiments revealed that BVC induced PANoptosis in EC cells by directly interacting with toll-like receptor 4 (TLR4), thereby triggering mitochondrial ROS generation, activating the cGAS-STING pathway. Notably, TLR4 knockdown inhibited STING-TBK1-IRF3 pathway and ZBP1-mediated PANoptosis. In addition, low-dose BVC combined with cisplatin increased phosphorylated H2AX expression, suggesting that BVC enhances the sensitivity of EC cells to cisplatin. In vivo studies demonstrated that BVC induced PANoptosis in EC, and BVC in combination with cisplatin had effective anti-tumor effect without injuring vital organs. These novel findings provide compelling evidence to support the clinical application of BVC and PANoptosis-based therapy for treating EC.</div></div>","PeriodicalId":8806,"journal":{"name":"Biochemical pharmacology","volume":"242 ","pages":"Article 117321"},"PeriodicalIF":5.6000,"publicationDate":"2025-09-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":"{\"title\":\"Bavachinin exerts anti-tumor effects by activating TLR4/STING axis-dependent PANoptosis and synergistically enhances chemosensitivity in endometrial cancer\",\"authors\":\"Wan Shu , Guanxiao Chen , Jun Zhang, Kejun Dong, Ting Zhou, Shuangshuang Cheng, Tangansu Zhang, Jiarui Zhang, Haojia Li, Yuwei Yao, Shuyang Yu, Yan Liu, Xing Zhou, Xiaoyu Shen, Hongbo Wang\",\"doi\":\"10.1016/j.bcp.2025.117321\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<div><div>The incidence and mortality rates of endometrial cancer (EC), a malignancy originating from endometrium, have been increasing globally. Currently, there are no effective therapeutic options for patients with recurrent, chemoresistant, and metastatic forms of this disease. Through compound library screening, we identified that bavachinin (BVC) has a killing effect on EC cells. BVC is a bioactive small molecule with potential pharmacological effects derived from the traditional Chinese herb <em>Proralea corylifolia L</em>, but the specific mechanisms are unclear. We first discovered that BVC induces ZBP1 (Z-DNA binding protein 1)-mediated PANoptosis in EC cells, characterized by activating of pyroptosis, apoptosis, and necroptosis. BVC promoted reactive oxygen species (ROS) production, mitochondrial membrane potential (MMP, ΔΨm) collapse, and ATM-CHK2 mediated DNA damage, which activated cGAS-STING pathway in EC cells. Mechanistically, network pharmacology, molecular docking, cellular thermal shift assays (CESTA), and drug affinity responsive target stability (DARTS) experiments revealed that BVC induced PANoptosis in EC cells by directly interacting with toll-like receptor 4 (TLR4), thereby triggering mitochondrial ROS generation, activating the cGAS-STING pathway. Notably, TLR4 knockdown inhibited STING-TBK1-IRF3 pathway and ZBP1-mediated PANoptosis. In addition, low-dose BVC combined with cisplatin increased phosphorylated H2AX expression, suggesting that BVC enhances the sensitivity of EC cells to cisplatin. In vivo studies demonstrated that BVC induced PANoptosis in EC, and BVC in combination with cisplatin had effective anti-tumor effect without injuring vital organs. These novel findings provide compelling evidence to support the clinical application of BVC and PANoptosis-based therapy for treating EC.</div></div>\",\"PeriodicalId\":8806,\"journal\":{\"name\":\"Biochemical pharmacology\",\"volume\":\"242 \",\"pages\":\"Article 117321\"},\"PeriodicalIF\":5.6000,\"publicationDate\":\"2025-09-10\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Biochemical pharmacology\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://www.sciencedirect.com/science/article/pii/S0006295225005866\",\"RegionNum\":2,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q1\",\"JCRName\":\"PHARMACOLOGY & PHARMACY\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Biochemical pharmacology","FirstCategoryId":"3","ListUrlMain":"https://www.sciencedirect.com/science/article/pii/S0006295225005866","RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"PHARMACOLOGY & PHARMACY","Score":null,"Total":0}
Bavachinin exerts anti-tumor effects by activating TLR4/STING axis-dependent PANoptosis and synergistically enhances chemosensitivity in endometrial cancer
The incidence and mortality rates of endometrial cancer (EC), a malignancy originating from endometrium, have been increasing globally. Currently, there are no effective therapeutic options for patients with recurrent, chemoresistant, and metastatic forms of this disease. Through compound library screening, we identified that bavachinin (BVC) has a killing effect on EC cells. BVC is a bioactive small molecule with potential pharmacological effects derived from the traditional Chinese herb Proralea corylifolia L, but the specific mechanisms are unclear. We first discovered that BVC induces ZBP1 (Z-DNA binding protein 1)-mediated PANoptosis in EC cells, characterized by activating of pyroptosis, apoptosis, and necroptosis. BVC promoted reactive oxygen species (ROS) production, mitochondrial membrane potential (MMP, ΔΨm) collapse, and ATM-CHK2 mediated DNA damage, which activated cGAS-STING pathway in EC cells. Mechanistically, network pharmacology, molecular docking, cellular thermal shift assays (CESTA), and drug affinity responsive target stability (DARTS) experiments revealed that BVC induced PANoptosis in EC cells by directly interacting with toll-like receptor 4 (TLR4), thereby triggering mitochondrial ROS generation, activating the cGAS-STING pathway. Notably, TLR4 knockdown inhibited STING-TBK1-IRF3 pathway and ZBP1-mediated PANoptosis. In addition, low-dose BVC combined with cisplatin increased phosphorylated H2AX expression, suggesting that BVC enhances the sensitivity of EC cells to cisplatin. In vivo studies demonstrated that BVC induced PANoptosis in EC, and BVC in combination with cisplatin had effective anti-tumor effect without injuring vital organs. These novel findings provide compelling evidence to support the clinical application of BVC and PANoptosis-based therapy for treating EC.
期刊介绍:
Biochemical Pharmacology publishes original research findings, Commentaries and review articles related to the elucidation of cellular and tissue function(s) at the biochemical and molecular levels, the modification of cellular phenotype(s) by genetic, transcriptional/translational or drug/compound-induced modifications, as well as the pharmacodynamics and pharmacokinetics of xenobiotics and drugs, the latter including both small molecules and biologics.
The journal''s target audience includes scientists engaged in the identification and study of the mechanisms of action of xenobiotics, biologics and drugs and in the drug discovery and development process.
All areas of cellular biology and cellular, tissue/organ and whole animal pharmacology fall within the scope of the journal. Drug classes covered include anti-infectives, anti-inflammatory agents, chemotherapeutics, cardiovascular, endocrinological, immunological, metabolic, neurological and psychiatric drugs, as well as research on drug metabolism and kinetics. While medicinal chemistry is a topic of complimentary interest, manuscripts in this area must contain sufficient biological data to characterize pharmacologically the compounds reported. Submissions describing work focused predominately on chemical synthesis and molecular modeling will not be considered for review.
While particular emphasis is placed on reporting the results of molecular and biochemical studies, research involving the use of tissue and animal models of human pathophysiology and toxicology is of interest to the extent that it helps define drug mechanisms of action, safety and efficacy.