{"title":"Tussilagone通过抑制TLR4/NF-κB通路和增强抗肿瘤免疫抑制三阴性乳腺癌进展。","authors":"Rui Huang, Hao Yu, Zhimou Tang","doi":"10.1080/08923973.2025.2555470","DOIUrl":null,"url":null,"abstract":"<p><strong>Background: </strong>As a highly aggressive form of breast cancer, triple-negative breast cancer (TNBC) is characterized by significant metastatic potential, a lack of targeted therapies, and an unfavorable prognosis. Tussilagone (TUS), a bioactive sesquiterpene isolated from Tussilago farfara's medicinal herb, has shown anti-inflammatory and anticancer properties. However, its potential role in TNBC treatment and the underlying molecular mechanisms remain unexplored.</p><p><strong>Methods: </strong>The antitumor effects of TUS on the proliferation, epithelial-mesenchymal transition (EMT), and metastasis of TNBC cell lines MDA-MB-231 and BT549 were assessed through cell viability, invasion, and colony formation assays. Western blot analysis was performed to detect Ki67, vimentin, E-cadherin, N-cadherin, PD-L1, LAG-3, TIM-3, and key TLR4/NF-κB regulators. Impact on the tumor immune microenvironment (TIME) was evaluated using CD8<sup>+</sup> T cell co-culture, ELISA, and flow cytometry. The <i>in vivo</i> anti-tumor efficacy of TUS was investigated in a TNBC xenograft mouse model.</p><p><strong>Results: </strong>TUS exhibited a dose-dependent inhibition of TNBC cell proliferation and invasion, reversed EMT by upregulating E-cadherin and downregulating N-cadherin and vimentin expression. It showed minimal cytotoxicity toward normal breast epithelial cells. TUS suppressed the TLR4/NF-κB pathway by downregulating TLR4, MyD88, and phosphorylated NF-κB, and decreased PD-L1 expression. Furthermore, TUS enhanced CD8<sup>+</sup> T cell activation, increased cytokine secretion (IFN-γ, IL-2, TNF-α), reduced LAG-3 and TIM-3 expression, and attenuated CD8<sup>+</sup> T cell apoptosis. Treatment of CD8<sup>+</sup> T cells directly with TUS did not affect cytokine secretion or apoptosis, suggesting that its immunomodulatory effects are mediated through tumor cell modulation. <i>In vivo</i> studies demonstrated significant tumor growth inhibition by TUS without inducing toxicity.</p><p><strong>Conclusion: </strong>This study demonstrated that TUS inhibited TNBC progression by suppressing the TLR4/NF-κB pathway, reversing EMT, and modulating TIME. These findings support the potential of TUS as a promising therapeutic candidate for TNBC and underscore the need for further clinical investigation.</p>","PeriodicalId":13420,"journal":{"name":"Immunopharmacology and Immunotoxicology","volume":" ","pages":"1-11"},"PeriodicalIF":3.0000,"publicationDate":"2025-09-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":"{\"title\":\"Tussilagone suppresses triple-negative breast cancer progression by inhibiting the TLR4/NF-κB pathway and enhancing anti-tumor immunity.\",\"authors\":\"Rui Huang, Hao Yu, Zhimou Tang\",\"doi\":\"10.1080/08923973.2025.2555470\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p><strong>Background: </strong>As a highly aggressive form of breast cancer, triple-negative breast cancer (TNBC) is characterized by significant metastatic potential, a lack of targeted therapies, and an unfavorable prognosis. Tussilagone (TUS), a bioactive sesquiterpene isolated from Tussilago farfara's medicinal herb, has shown anti-inflammatory and anticancer properties. However, its potential role in TNBC treatment and the underlying molecular mechanisms remain unexplored.</p><p><strong>Methods: </strong>The antitumor effects of TUS on the proliferation, epithelial-mesenchymal transition (EMT), and metastasis of TNBC cell lines MDA-MB-231 and BT549 were assessed through cell viability, invasion, and colony formation assays. Western blot analysis was performed to detect Ki67, vimentin, E-cadherin, N-cadherin, PD-L1, LAG-3, TIM-3, and key TLR4/NF-κB regulators. Impact on the tumor immune microenvironment (TIME) was evaluated using CD8<sup>+</sup> T cell co-culture, ELISA, and flow cytometry. The <i>in vivo</i> anti-tumor efficacy of TUS was investigated in a TNBC xenograft mouse model.</p><p><strong>Results: </strong>TUS exhibited a dose-dependent inhibition of TNBC cell proliferation and invasion, reversed EMT by upregulating E-cadherin and downregulating N-cadherin and vimentin expression. It showed minimal cytotoxicity toward normal breast epithelial cells. TUS suppressed the TLR4/NF-κB pathway by downregulating TLR4, MyD88, and phosphorylated NF-κB, and decreased PD-L1 expression. Furthermore, TUS enhanced CD8<sup>+</sup> T cell activation, increased cytokine secretion (IFN-γ, IL-2, TNF-α), reduced LAG-3 and TIM-3 expression, and attenuated CD8<sup>+</sup> T cell apoptosis. Treatment of CD8<sup>+</sup> T cells directly with TUS did not affect cytokine secretion or apoptosis, suggesting that its immunomodulatory effects are mediated through tumor cell modulation. <i>In vivo</i> studies demonstrated significant tumor growth inhibition by TUS without inducing toxicity.</p><p><strong>Conclusion: </strong>This study demonstrated that TUS inhibited TNBC progression by suppressing the TLR4/NF-κB pathway, reversing EMT, and modulating TIME. These findings support the potential of TUS as a promising therapeutic candidate for TNBC and underscore the need for further clinical investigation.</p>\",\"PeriodicalId\":13420,\"journal\":{\"name\":\"Immunopharmacology and Immunotoxicology\",\"volume\":\" \",\"pages\":\"1-11\"},\"PeriodicalIF\":3.0000,\"publicationDate\":\"2025-09-30\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Immunopharmacology and Immunotoxicology\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://doi.org/10.1080/08923973.2025.2555470\",\"RegionNum\":4,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q3\",\"JCRName\":\"IMMUNOLOGY\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Immunopharmacology and Immunotoxicology","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1080/08923973.2025.2555470","RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q3","JCRName":"IMMUNOLOGY","Score":null,"Total":0}
Tussilagone suppresses triple-negative breast cancer progression by inhibiting the TLR4/NF-κB pathway and enhancing anti-tumor immunity.
Background: As a highly aggressive form of breast cancer, triple-negative breast cancer (TNBC) is characterized by significant metastatic potential, a lack of targeted therapies, and an unfavorable prognosis. Tussilagone (TUS), a bioactive sesquiterpene isolated from Tussilago farfara's medicinal herb, has shown anti-inflammatory and anticancer properties. However, its potential role in TNBC treatment and the underlying molecular mechanisms remain unexplored.
Methods: The antitumor effects of TUS on the proliferation, epithelial-mesenchymal transition (EMT), and metastasis of TNBC cell lines MDA-MB-231 and BT549 were assessed through cell viability, invasion, and colony formation assays. Western blot analysis was performed to detect Ki67, vimentin, E-cadherin, N-cadherin, PD-L1, LAG-3, TIM-3, and key TLR4/NF-κB regulators. Impact on the tumor immune microenvironment (TIME) was evaluated using CD8+ T cell co-culture, ELISA, and flow cytometry. The in vivo anti-tumor efficacy of TUS was investigated in a TNBC xenograft mouse model.
Results: TUS exhibited a dose-dependent inhibition of TNBC cell proliferation and invasion, reversed EMT by upregulating E-cadherin and downregulating N-cadherin and vimentin expression. It showed minimal cytotoxicity toward normal breast epithelial cells. TUS suppressed the TLR4/NF-κB pathway by downregulating TLR4, MyD88, and phosphorylated NF-κB, and decreased PD-L1 expression. Furthermore, TUS enhanced CD8+ T cell activation, increased cytokine secretion (IFN-γ, IL-2, TNF-α), reduced LAG-3 and TIM-3 expression, and attenuated CD8+ T cell apoptosis. Treatment of CD8+ T cells directly with TUS did not affect cytokine secretion or apoptosis, suggesting that its immunomodulatory effects are mediated through tumor cell modulation. In vivo studies demonstrated significant tumor growth inhibition by TUS without inducing toxicity.
Conclusion: This study demonstrated that TUS inhibited TNBC progression by suppressing the TLR4/NF-κB pathway, reversing EMT, and modulating TIME. These findings support the potential of TUS as a promising therapeutic candidate for TNBC and underscore the need for further clinical investigation.
期刊介绍:
The journal Immunopharmacology and Immunotoxicology is devoted to pre-clinical and clinical drug discovery and development targeting the immune system. Research related to the immunoregulatory effects of various compounds, including small-molecule drugs and biologics, on immunocompetent cells and immune responses, as well as the immunotoxicity exerted by xenobiotics and drugs. Only research that describe the mechanisms of specific compounds (not extracts) is of interest to the journal.
The journal will prioritise preclinical and clinical studies on immunotherapy of disorders such as chronic inflammation, allergy, autoimmunity, cancer etc. The effects of small-drugs, vaccines and biologics against central immunological targets as well as cell-based therapy, including dendritic cell therapy, T cell adoptive transfer and stem cell therapy, are topics of particular interest. Publications pointing towards potential new drug targets within the immune system or novel technology for immunopharmacological drug development are also welcome.
With an immunoscience focus on drug development, immunotherapy and toxicology, the journal will cover areas such as infection, allergy, inflammation, tumor immunology, degenerative disorders, immunodeficiencies, neurology, atherosclerosis and more.
Immunopharmacology and Immunotoxicology will accept original manuscripts, brief communications, commentaries, mini-reviews, reviews, clinical trials and clinical cases, on the condition that the results reported are based on original, clinical, or basic research that has not been published elsewhere in any journal in any language (except in abstract form relating to paper communicated to scientific meetings and symposiums).