Yuting Gao , Yi Gong , Xiaoyong Song, Yajun Xiong, Junlan Lu, Yanguang Yang, Yanli Gong, Zhimin Du, Shanshan Wang, Ruilong Jia, Peng Gong, Xinli Shi
{"title":"双氢青蒿素通过YAP1抑制组蛋白乳酸化,在肝细胞癌中充当“冷”肿瘤的“热”开关。","authors":"Yuting Gao , Yi Gong , Xiaoyong Song, Yajun Xiong, Junlan Lu, Yanguang Yang, Yanli Gong, Zhimin Du, Shanshan Wang, Ruilong Jia, Peng Gong, Xinli Shi","doi":"10.1016/j.phymed.2025.157307","DOIUrl":null,"url":null,"abstract":"<div><h3>Background</h3><div>Hepatocellular carcinoma (HCC) is characterized by a ‘cold’ tumor microenvironment (TME), which limits the efficacy of immune checkpoint inhibitors (ICIs). In a previous study, we demonstrated that dihydroartemisinin (DHA) could disrupt the immunosuppressive TME and delay HCC progression.</div></div><div><h3>Purpose</h3><div>This study aimed to explore the precise mechanisms underlying the shift of the TME from an immunosuppressive to an immunostimulatory state.</div></div><div><h3>Methods</h3><div>We constructed HCC subcutaneous allograft mice model to investigate the in vivo antitumor effects of DHA. We further performed single-cell RNA sequencing (scRNA-seq) and multiplex immunohistochemistry (mIHC) to analyze the immune dynamics during DHA-induced TME transition in HCC. Using NCG mice (lacking T, B, and NK cells), BALB/c nude mice (lacking T cells), and CD8<sup>+</sup> T cell-depleted mice, we assessed whether the antitumor effect of DHA in HCC depends on its immune functions and determined the key role of T cells in antitumor immunity. Additional downstream mechanisms were explored using YAP1 knockdown HCC cells and liver-specific Yap1 knockout mice through techniques such as Western blot, immunofluorescence (IF), metabolic flux analysis, co-immunoprecipitation (Co-IP), chemokine chip, and flow cytometry (FCM).</div></div><div><h3>Results</h3><div>We observed that DHA transforms the TME of HCC from an immune-cold to an immune-hot state by increasing the abundance and proportion of T cells, NK cells, M1-like TAMs, and DCs. Notably, DHA was found to enhance the proportion of IFN-γ<sup>+</sup> CD8<sup>+</sup> T cells within the TME of HCC-bearing mice, and its therapeutic effects were dependent on CD8<sup>+</sup> T cells. Our results suggest that DHA could suppress histone lactylation (Kla) and acetylation (Kac) in HCC cells by inhibiting YAP1. Mechanistically, DHA reduces lactate transport, thereby decreasing lactate accumulation within the TME, lowering P300/CBP catalytic activity, and enhancing the deacetylation effect of HDAC1-3. This finding reveals a new mechanism by which DHA, through metabolic changes and epigenetic regulation, remodels the TME of HCC. Additionally, a combination strategy involving DHA and anti-PD-1 therapy demonstrated the potential to reshape the cold HCC TME via inhibition of the YAP1-lactylation positive feedback loop.</div></div><div><h3>Conclusion</h3><div>Our study confirms that DHA inhibits histone Kla in HCC cells through YAP1 inhibition, which shifts the TME from an immune-cold to immune-hot state. Moreover, DHA enhance the anti-HCC immune response dependent on CD8<sup>+</sup> T cells, thereby sensitizing anti-PD-1 therapy. This novel finding positions DHA as a promising strategy for optimizing anti-PD-1 therapy in HCC, warranting further clinical application studies.</div></div>","PeriodicalId":20212,"journal":{"name":"Phytomedicine","volume":"148 ","pages":"Article 157307"},"PeriodicalIF":8.3000,"publicationDate":"2025-09-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":"{\"title\":\"Dihydroartemisinin inhibits histone lactylation through YAP1 to act as a ‘hot’ switch for ‘cold’ tumor in hepatocellular carcinoma\",\"authors\":\"Yuting Gao , Yi Gong , Xiaoyong Song, Yajun Xiong, Junlan Lu, Yanguang Yang, Yanli Gong, Zhimin Du, Shanshan Wang, Ruilong Jia, Peng Gong, Xinli Shi\",\"doi\":\"10.1016/j.phymed.2025.157307\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<div><h3>Background</h3><div>Hepatocellular carcinoma (HCC) is characterized by a ‘cold’ tumor microenvironment (TME), which limits the efficacy of immune checkpoint inhibitors (ICIs). In a previous study, we demonstrated that dihydroartemisinin (DHA) could disrupt the immunosuppressive TME and delay HCC progression.</div></div><div><h3>Purpose</h3><div>This study aimed to explore the precise mechanisms underlying the shift of the TME from an immunosuppressive to an immunostimulatory state.</div></div><div><h3>Methods</h3><div>We constructed HCC subcutaneous allograft mice model to investigate the in vivo antitumor effects of DHA. We further performed single-cell RNA sequencing (scRNA-seq) and multiplex immunohistochemistry (mIHC) to analyze the immune dynamics during DHA-induced TME transition in HCC. Using NCG mice (lacking T, B, and NK cells), BALB/c nude mice (lacking T cells), and CD8<sup>+</sup> T cell-depleted mice, we assessed whether the antitumor effect of DHA in HCC depends on its immune functions and determined the key role of T cells in antitumor immunity. Additional downstream mechanisms were explored using YAP1 knockdown HCC cells and liver-specific Yap1 knockout mice through techniques such as Western blot, immunofluorescence (IF), metabolic flux analysis, co-immunoprecipitation (Co-IP), chemokine chip, and flow cytometry (FCM).</div></div><div><h3>Results</h3><div>We observed that DHA transforms the TME of HCC from an immune-cold to an immune-hot state by increasing the abundance and proportion of T cells, NK cells, M1-like TAMs, and DCs. Notably, DHA was found to enhance the proportion of IFN-γ<sup>+</sup> CD8<sup>+</sup> T cells within the TME of HCC-bearing mice, and its therapeutic effects were dependent on CD8<sup>+</sup> T cells. Our results suggest that DHA could suppress histone lactylation (Kla) and acetylation (Kac) in HCC cells by inhibiting YAP1. Mechanistically, DHA reduces lactate transport, thereby decreasing lactate accumulation within the TME, lowering P300/CBP catalytic activity, and enhancing the deacetylation effect of HDAC1-3. This finding reveals a new mechanism by which DHA, through metabolic changes and epigenetic regulation, remodels the TME of HCC. Additionally, a combination strategy involving DHA and anti-PD-1 therapy demonstrated the potential to reshape the cold HCC TME via inhibition of the YAP1-lactylation positive feedback loop.</div></div><div><h3>Conclusion</h3><div>Our study confirms that DHA inhibits histone Kla in HCC cells through YAP1 inhibition, which shifts the TME from an immune-cold to immune-hot state. Moreover, DHA enhance the anti-HCC immune response dependent on CD8<sup>+</sup> T cells, thereby sensitizing anti-PD-1 therapy. This novel finding positions DHA as a promising strategy for optimizing anti-PD-1 therapy in HCC, warranting further clinical application studies.</div></div>\",\"PeriodicalId\":20212,\"journal\":{\"name\":\"Phytomedicine\",\"volume\":\"148 \",\"pages\":\"Article 157307\"},\"PeriodicalIF\":8.3000,\"publicationDate\":\"2025-09-23\",\"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/S0944711325009468\",\"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/S0944711325009468","RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"CHEMISTRY, MEDICINAL","Score":null,"Total":0}
Dihydroartemisinin inhibits histone lactylation through YAP1 to act as a ‘hot’ switch for ‘cold’ tumor in hepatocellular carcinoma
Background
Hepatocellular carcinoma (HCC) is characterized by a ‘cold’ tumor microenvironment (TME), which limits the efficacy of immune checkpoint inhibitors (ICIs). In a previous study, we demonstrated that dihydroartemisinin (DHA) could disrupt the immunosuppressive TME and delay HCC progression.
Purpose
This study aimed to explore the precise mechanisms underlying the shift of the TME from an immunosuppressive to an immunostimulatory state.
Methods
We constructed HCC subcutaneous allograft mice model to investigate the in vivo antitumor effects of DHA. We further performed single-cell RNA sequencing (scRNA-seq) and multiplex immunohistochemistry (mIHC) to analyze the immune dynamics during DHA-induced TME transition in HCC. Using NCG mice (lacking T, B, and NK cells), BALB/c nude mice (lacking T cells), and CD8+ T cell-depleted mice, we assessed whether the antitumor effect of DHA in HCC depends on its immune functions and determined the key role of T cells in antitumor immunity. Additional downstream mechanisms were explored using YAP1 knockdown HCC cells and liver-specific Yap1 knockout mice through techniques such as Western blot, immunofluorescence (IF), metabolic flux analysis, co-immunoprecipitation (Co-IP), chemokine chip, and flow cytometry (FCM).
Results
We observed that DHA transforms the TME of HCC from an immune-cold to an immune-hot state by increasing the abundance and proportion of T cells, NK cells, M1-like TAMs, and DCs. Notably, DHA was found to enhance the proportion of IFN-γ+ CD8+ T cells within the TME of HCC-bearing mice, and its therapeutic effects were dependent on CD8+ T cells. Our results suggest that DHA could suppress histone lactylation (Kla) and acetylation (Kac) in HCC cells by inhibiting YAP1. Mechanistically, DHA reduces lactate transport, thereby decreasing lactate accumulation within the TME, lowering P300/CBP catalytic activity, and enhancing the deacetylation effect of HDAC1-3. This finding reveals a new mechanism by which DHA, through metabolic changes and epigenetic regulation, remodels the TME of HCC. Additionally, a combination strategy involving DHA and anti-PD-1 therapy demonstrated the potential to reshape the cold HCC TME via inhibition of the YAP1-lactylation positive feedback loop.
Conclusion
Our study confirms that DHA inhibits histone Kla in HCC cells through YAP1 inhibition, which shifts the TME from an immune-cold to immune-hot state. Moreover, DHA enhance the anti-HCC immune response dependent on CD8+ T cells, thereby sensitizing anti-PD-1 therapy. This novel finding positions DHA as a promising strategy for optimizing anti-PD-1 therapy in HCC, warranting further clinical application studies.
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.