首页 > 最新文献

Phytomedicine最新文献

英文 中文
Unveiling poliumoside: A herbal-derived FAK kinase inhibitor with anti-proliferative and anti-metastatic efficacy. 揭幕poliumoside:一种草药衍生的FAK激酶抑制剂,具有抗增殖和抗转移功效。
IF 8.3 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2026-01-01 Epub Date: 2025-12-30 DOI: 10.1016/j.phymed.2025.157747
Huiling Zhang, Yuna Shao, Di Lu, Chenkang Ma, Liying Zhang, Yang Yang, Jianjun Li, Yuanyuan Zeng, Jianjie Zhu, Jian-An Huang, Zhe Lei, Zeyi Liu

Focal adhesion kinase (FAK) is an attractive therapeutic target overexpressed in numerous cancers. Despite extensive efforts, no FAK inhibitor has reached the market. Here, we identified poliumoside (Pol), a compound from Chinese herbs, as a novel FAK inhibitor through high-throughput virtual screening. Pol exhibited potent anti-proliferative and anti-metastatic activities both in vitro and in vivo. Mechanistically, Pol directly binds to His89 of FAK, thereby inhibiting its autophosphorylation at Y397. This inhibition led to the downregulation of the ERK-c-Myc axis, reduced transcription of IL-6, and consequent suppression of the JAK3-STAT3 signaling pathway. Our findings establish Pol as a promising FAK-targeting agent, exerting its anti-tumor effects in a FAK-dependent manner.

局灶黏附激酶(FAK)是一个有吸引力的治疗靶点,在许多癌症中过表达。尽管付出了巨大的努力,但FAK抑制剂尚未进入市场。本研究通过高通量虚拟筛选,从中草药中分离出一种新型的FAK抑制剂poliumoside (Pol)。Pol在体外和体内均表现出较强的抗增殖和抗转移活性。在机制上,Pol直接结合FAK的His89,从而抑制其Y397的自磷酸化。这种抑制导致ERK-c-Myc轴下调,IL-6转录减少,进而抑制JAK3-STAT3信号通路。我们的研究结果表明,Pol是一种有前途的fak靶向药物,以fak依赖的方式发挥其抗肿瘤作用。
{"title":"Unveiling poliumoside: A herbal-derived FAK kinase inhibitor with anti-proliferative and anti-metastatic efficacy.","authors":"Huiling Zhang, Yuna Shao, Di Lu, Chenkang Ma, Liying Zhang, Yang Yang, Jianjun Li, Yuanyuan Zeng, Jianjie Zhu, Jian-An Huang, Zhe Lei, Zeyi Liu","doi":"10.1016/j.phymed.2025.157747","DOIUrl":"10.1016/j.phymed.2025.157747","url":null,"abstract":"<p><p>Focal adhesion kinase (FAK) is an attractive therapeutic target overexpressed in numerous cancers. Despite extensive efforts, no FAK inhibitor has reached the market. Here, we identified poliumoside (Pol), a compound from Chinese herbs, as a novel FAK inhibitor through high-throughput virtual screening. Pol exhibited potent anti-proliferative and anti-metastatic activities both in vitro and in vivo. Mechanistically, Pol directly binds to His89 of FAK, thereby inhibiting its autophosphorylation at Y397. This inhibition led to the downregulation of the ERK-c-Myc axis, reduced transcription of IL-6, and consequent suppression of the JAK3-STAT3 signaling pathway. Our findings establish Pol as a promising FAK-targeting agent, exerting its anti-tumor effects in a FAK-dependent manner.</p>","PeriodicalId":20212,"journal":{"name":"Phytomedicine","volume":"150 ","pages":"157747"},"PeriodicalIF":8.3,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145900206","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Integrating CyTOF with scRNA-seq reveals that Cyclovirobuxine D inhibits HCC progression through hepatic immune microenvironment remodeling 结合CyTOF和scRNA-seq,揭示环virobuxine D通过肝脏免疫微环境重塑抑制HCC进展
IF 8.3 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2026-01-01 DOI: 10.1016/j.phymed.2025.157751
Fuqiang Ma , Lele Zhang , Yitao Fan , Ziyi Xu , Yuhua He , Jiachun Sun , Juan Lu , Xinyu Gu

Background

Hepatocellular carcinoma (HCC) represents the predominant type of primary malignant liver cancer, characterized by high morbidity and mortality rates. The steroidal alkaloid cyclovirobuxine D (CVB-D), which is extracted from Buxus microphylla, has garnered increasing attention for its potential in anti-tumor research. Despite this, the immunoregulatory effects of CVB-D in HCC remain insufficiently explored.

Purpose

This study investigated the role and underlying mechanisms of CVB-D in inhibiting HCC progression, aiming to identify potential therapeutic targets and provide new scientific evidence for HCC treatment.

Study design

CVB-D was administrated to orthotopic HCC mouse models. The effects of CVB-D on the tumor immune microenvironment were systematically assessed via integrated cytometry by time of flight (CyTOF) and single-cell RNA sequencing (scRNA-seq).

Methods

An orthotopic HCC model was generated in C57BL/6 mice via Hepa1–6 cell implantation, followed by intraperitoneal administration of different doses of CVB-D. Following a 14-day treatment period, fresh tumor tissues were collected for CyTOF and scRNA-seq analyses. Multiplexed immunofluorescence staining was performed on liver tissue sections to validate the in suit localization of key biomarkers. Serum cytokine levels were evaluated utilizing the Quantibody® Mouse Inflammation Array Kit.

Results

This research demonstrated that administration of CVB-D in orthotopic HCC mouse models notably inhibited tumor progression. Comprehensive analyses combining CyTOF and scRNA-seq demonstrated that CVB-D remodeled the hepatic immune microenvironment by enhancing CD8+ T cell functionality, modulating macrophage polarization, and reducing the abundance of malignant epithelial cells.

Conclusion

These findings highlight the therapeutic promise of CVB-D as a novel candidate for HCC treatment, offering new insights into the development of innovative anti-cancer therapies.
背景:肝细胞癌(HCC)是原发性恶性肝癌的主要类型,具有高发病率和高死亡率的特点。从小叶黄杨中提取的甾体生物碱环virobuxine D (CVB-D)因其在抗肿瘤研究中的潜力而受到越来越多的关注。尽管如此,CVB-D在HCC中的免疫调节作用仍未得到充分探讨。目的探讨CVB-D在抑制HCC进展中的作用及其机制,旨在发现潜在的治疗靶点,为HCC的治疗提供新的科学依据。研究设计cvb - d给予原位肝癌小鼠模型。通过飞行时间(CyTOF)和单细胞RNA测序(scRNA-seq)集成细胞术系统评估CVB-D对肿瘤免疫微环境的影响。方法在C57BL/6小鼠原位肝癌模型中植入Hepa1-6细胞,然后腹腔注射不同剂量的CVB-D。在14天的治疗期后,收集新鲜肿瘤组织进行CyTOF和scRNA-seq分析。对肝组织切片进行多重免疫荧光染色,以验证关键生物标志物的匹配定位。使用Quantibody®小鼠炎症阵列试剂盒评估血清细胞因子水平。结果本研究表明,CVB-D在原位肝癌小鼠模型中显著抑制肿瘤进展。结合CyTOF和scRNA-seq的综合分析表明,CVB-D通过增强CD8+ T细胞功能、调节巨噬细胞极化和降低恶性上皮细胞丰度来重塑肝脏免疫微环境。结论这些发现突出了CVB-D作为HCC治疗的新候选药物的治疗前景,为创新抗癌疗法的发展提供了新的见解。
{"title":"Integrating CyTOF with scRNA-seq reveals that Cyclovirobuxine D inhibits HCC progression through hepatic immune microenvironment remodeling","authors":"Fuqiang Ma ,&nbsp;Lele Zhang ,&nbsp;Yitao Fan ,&nbsp;Ziyi Xu ,&nbsp;Yuhua He ,&nbsp;Jiachun Sun ,&nbsp;Juan Lu ,&nbsp;Xinyu Gu","doi":"10.1016/j.phymed.2025.157751","DOIUrl":"10.1016/j.phymed.2025.157751","url":null,"abstract":"<div><h3>Background</h3><div>Hepatocellular carcinoma (HCC) represents the predominant type of primary malignant liver cancer, characterized by high morbidity and mortality rates. The steroidal alkaloid cyclovirobuxine D (CVB-D), which is extracted from <em>Buxus microphylla</em>, has garnered increasing attention for its potential in anti-tumor research. Despite this, the immunoregulatory effects of CVB-D in HCC remain insufficiently explored.</div></div><div><h3>Purpose</h3><div>This study investigated the role and underlying mechanisms of CVB-D in inhibiting HCC progression, aiming to identify potential therapeutic targets and provide new scientific evidence for HCC treatment.</div></div><div><h3>Study design</h3><div>CVB-D was administrated to orthotopic HCC mouse models. The effects of CVB-D on the tumor immune microenvironment were systematically assessed <em>via</em> integrated cytometry by time of flight (CyTOF) and single-cell RNA sequencing (scRNA-seq).</div></div><div><h3>Methods</h3><div>An orthotopic HCC model was generated in C57BL/6 mice <em>via</em> Hepa1–6 cell implantation, followed by intraperitoneal administration of different doses of CVB-D. Following a 14-day treatment period, fresh tumor tissues were collected for CyTOF and scRNA-seq analyses. Multiplexed immunofluorescence staining was performed on liver tissue sections to validate the in suit localization of key biomarkers. Serum cytokine levels were evaluated utilizing the Quantibody® Mouse Inflammation Array Kit.</div></div><div><h3>Results</h3><div>This research demonstrated that administration of CVB-D in orthotopic HCC mouse models notably inhibited tumor progression. Comprehensive analyses combining CyTOF and scRNA-seq demonstrated that CVB-D remodeled the hepatic immune microenvironment by enhancing CD8<sup>+</sup> <em>T</em> cell functionality, modulating macrophage polarization, and reducing the abundance of malignant epithelial cells.</div></div><div><h3>Conclusion</h3><div>These findings highlight the therapeutic promise of CVB-D as a novel candidate for HCC treatment, offering new insights into the development of innovative anti-cancer therapies.</div></div>","PeriodicalId":20212,"journal":{"name":"Phytomedicine","volume":"150 ","pages":"Article 157751"},"PeriodicalIF":8.3,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145884117","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Anwulignan mitigates UVB-induced skin photodamage by dual activation of the Nrf2/PINK1/Parkin and Nrf2/SLC7A11/GPX4 signaling pathways 安五丽素通过双激活Nrf2/PINK1/Parkin和Nrf2/SLC7A11/GPX4信号通路减轻uvb诱导的皮肤光损伤。
IF 8.3 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2026-01-01 DOI: 10.1016/j.phymed.2025.157692
Yuxin Jiang , Danfeng Wang , Xiaoran Lin , Zihao Yuan , Xinyu Xing , Qingzhuo Gao , Tianshuo Zhang , Shu Jing , He Li

Background

Skin photoaging (SP) is a major contributor to skin aging. Anwulignan (AN) has been confirmed to possess diverse biological activities. This study investigated the protective effect of AN against SP and explored its underlying mechanisms.

Methods

A UVB-induced SP model in mice was established to evaluate skin erythema index (EI), transepidermal water loss (TEWL), and skin hydration. Histopathological assessments were performed using hematoxylin and eosin (HE), Masson, and Sirius red staining. Transmission electron microscopy examined the morphology and quantity of mitochondria in epidermal cells. ELISA measured levels of hydroxyproline (HYP), hyaluronic acid (HA), oxidative stress markers, and inflammation indicators. Molecular docking predicted the potential targets of AN on UVB-induced SP. Western blot and immunofluorescence staining assessed the expression of proteins related to oxidation, inflammation, aging, ferroptosis, and autophagy. Zebrafish experiments further validated the effects.

Results

AN significantly alleviated SP, evidenced by increased levels of HA and HYP, suppressed pro-inflammatory cytokine expression, reduced reactive oxygen species (ROS), 8-hydroxy-2′-deoxyguanosine (8-OHdG), and malondialdehyde (MDA), while enhancing activities of total superoxide dismutase (T-SOD), catalase (CAT), and glutathione peroxidase (GSH-Px). It also regulated the expression of proteins involved in the Nrf2/PINK1/Parkin mitophagy axis and the Nrf2/SLC7A11/GPX4 ferroptosis regulatory axis. Significant differences were observed among groups in tail fin area, β-galactosidase staining fluorescence intensity, yolk sac fluorescence, and mitochondrial fluorescence in muscle.

Conclusion

AN may exert protective effects against SP by targeting Nrf2 and subsequently activating both the Nrf2/PINK1/Parkin mitophagy axis and the Nrf2/SLC7A11/GPX4 ferroptosis regulatory axis.
背景:皮肤光老化是皮肤老化的主要原因。anwuligan (AN)已被证实具有多种生物活性。本研究探讨了AN对SP的保护作用,并探讨了其机制。方法:建立uvb诱导的小鼠SP模型,评价皮肤红斑指数(EI)、经皮失水(TEWL)和皮肤水化程度。采用苏木精和伊红(HE)、Masson和Sirius红染色进行组织病理学评估。透射电镜观察表皮细胞线粒体的形态和数量。ELISA检测了羟脯氨酸(HYP)、透明质酸(HA)、氧化应激标志物和炎症指标的水平。分子对接预测AN对uvb诱导SP的潜在靶点,Western blot和免疫荧光染色评估氧化、炎症、衰老、铁下垂和自噬相关蛋白的表达。斑马鱼实验进一步验证了这一效果。结果:AN显著缓解SP,表现为HA和HYP水平升高,抑制促炎细胞因子表达,降低活性氧(ROS)、8-羟基-2′-脱氧鸟苷(8-OHdG)和丙二醛(MDA),增强总超氧化物歧化酶(T-SOD)、过氧化氢酶(CAT)和谷胱甘肽过氧化物酶(GSH-Px)活性。它还调节Nrf2/PINK1/Parkin自噬轴和Nrf2/SLC7A11/GPX4铁下垂调节轴相关蛋白的表达。各组间尾鳍面积、β-半乳糖苷酶染色荧光强度、卵黄囊荧光和肌肉线粒体荧光均有显著差异。结论:AN可能通过靶向Nrf2,激活Nrf2/PINK1/Parkin自噬轴和Nrf2/SLC7A11/GPX4铁凋亡调节轴,对SP发挥保护作用。
{"title":"Anwulignan mitigates UVB-induced skin photodamage by dual activation of the Nrf2/PINK1/Parkin and Nrf2/SLC7A11/GPX4 signaling pathways","authors":"Yuxin Jiang ,&nbsp;Danfeng Wang ,&nbsp;Xiaoran Lin ,&nbsp;Zihao Yuan ,&nbsp;Xinyu Xing ,&nbsp;Qingzhuo Gao ,&nbsp;Tianshuo Zhang ,&nbsp;Shu Jing ,&nbsp;He Li","doi":"10.1016/j.phymed.2025.157692","DOIUrl":"10.1016/j.phymed.2025.157692","url":null,"abstract":"<div><h3>Background</h3><div>Skin photoaging (SP) is a major contributor to skin aging. Anwulignan (AN) has been confirmed to possess diverse biological activities. This study investigated the protective effect of AN against SP and explored its underlying mechanisms.</div></div><div><h3>Methods</h3><div>A UVB-induced SP model in mice was established to evaluate skin erythema index (EI), transepidermal water loss (TEWL), and skin hydration. Histopathological assessments were performed using hematoxylin and eosin (HE), Masson, and Sirius red staining. Transmission electron microscopy examined the morphology and quantity of mitochondria in epidermal cells. ELISA measured levels of hydroxyproline (HYP), hyaluronic acid (HA), oxidative stress markers, and inflammation indicators. Molecular docking predicted the potential targets of AN on UVB-induced SP. Western blot and immunofluorescence staining assessed the expression of proteins related to oxidation, inflammation, aging, ferroptosis, and autophagy. Zebrafish experiments further validated the effects.</div></div><div><h3>Results</h3><div>AN significantly alleviated SP, evidenced by increased levels of HA and HYP, suppressed pro-inflammatory cytokine expression, reduced reactive oxygen species (ROS), 8-hydroxy-2′-deoxyguanosine (8-OHdG), and malondialdehyde (MDA), while enhancing activities of total superoxide dismutase (T-SOD), catalase (CAT), and glutathione peroxidase (GSH-Px). It also regulated the expression of proteins involved in the Nrf2/PINK1/Parkin mitophagy axis and the Nrf2/SLC7A11/GPX4 ferroptosis regulatory axis. Significant differences were observed among groups in tail fin area, β-galactosidase staining fluorescence intensity, yolk sac fluorescence, and mitochondrial fluorescence in muscle.</div></div><div><h3>Conclusion</h3><div>AN may exert protective effects against SP by targeting Nrf2 and subsequently activating both the Nrf2/PINK1/Parkin mitophagy axis and the Nrf2/SLC7A11/GPX4 ferroptosis regulatory axis.</div></div>","PeriodicalId":20212,"journal":{"name":"Phytomedicine","volume":"150 ","pages":"Article 157692"},"PeriodicalIF":8.3,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145917468","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Unveiling TCM-antibiotic synergy through multi-omics: Shuganning injection potentiates the effect of linezolid against methicillin-resistant Staphylococcus aureus. 通过多组学揭示中药-抗生素协同作用:舒肝宁注射液增强利奈唑胺对耐甲氧西林金黄色葡萄球菌的作用。
IF 8.3 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2026-01-01 Epub Date: 2025-12-20 DOI: 10.1016/j.phymed.2025.157733
Qing Wang, Yali Wu, Yan Wang, Xin Pang, Mingliang Zhang, Ya Zhao, Xi Qin, Bowen Zheng, Mingyue Li, Xuyang Li, Weixia Li, Xiaoyan Wang, Liuqing Yang, Xiaofei Chen, Jinfa Tang

Background: Methicillin-resistant Staphylococcus aureus (MRSA) poses a significant threat to global public health due to its multidrug resistance, highlighting the need for the development of novel antimicrobial strategies. Computational repurposing has predicted that Shuganning injection (SGN), a clinically used hepatoprotective traditional Chinese medicine, exhibits synergistic activity with antibiotics against MRSA. However, the antimicrobial properties of SGN remain insufficiently characterized.

Purpose: To evaluate the anti-MRSA activity of SGN and to elucidate the mechanism underlying its synergy with linezolid (LZD).

Methods: The components of SGN were identified using UPLC-Q/TOF-MS/MS. Antimicrobial potential was predicted through computational repurposing and validated through bactericidal and synergy assays. Biofilm inhibition, morphological changes, and cell wall integrity were assessed. Integrated metabolomics and transcriptomics were employed to investigate SGN-LZD synergy. Key findings were subsequently validated by RT-qPCR and confirmed through targeted pharmacological inhibition. Finally, in vivo efficacy was assessed in Galleria mellonella and murine skin infection models.

Results: Fifty-two components were identified in SGN. Computational repurposing analysis indicated that SGN reversed sepsis-associated gene dysregulation and inhibited the growth of clinically isolated MRSA417, with corresponding MIC and MBC values of 42.5 and 170 mg/mL (crude drug equivalent), respectively. SGN demonstrated partial synergy with LZD (FICI = 0.75). The combination markedly inhibited MRSA biofilm formation by 94.25%, compared to 40.40% for SGN alone. Sub-MIC combinations caused structural damage, evidenced by increased extracellular alkaline phosphatase levels and disrupted bacterial morphology. Multi-omics analysis indicated co-downregulation of two-component system (TCS)-related metabolites (e.g., L-malic acid) and genes (saeRS, vraSR), indicating TCS suppression as the key mechanism underlying SGN-LZD synergy. The combination downregulated key TCS mRNAs. Furthermore, TCS inhibitors did not enhance its anti-biofilm and antibacterial effects. Notably, the SGN-LZD combination improved survival in G. mellonella and promoted wound healing in a murine skin infection model.

Conclusion: This study provides evidence supporting the repositioning of SGN for MRSA treatment. SGN impairs TCS-mediated virulence regulation, cell envelope integrity, and essential metabolic pathways, thereby enhancing the efficacy of LZD. These findings offer a translational framework for drug repurposing and highlight the clinical potential of SGN-LZD combination therapy for drug-resistant infections.

背景:耐甲氧西林金黄色葡萄球菌(MRSA)由于其多药耐药性对全球公共卫生构成重大威胁,突出了开发新型抗菌策略的必要性。计算重新利用预测临床使用的肝保护中药舒肝宁注射液(SGN)与抗生素对MRSA具有协同作用。然而,SGN的抗菌特性仍然没有得到充分的表征。目的:评价SGN抗mrsa的活性,并阐明其与利奈唑胺(LZD)协同作用的机制。方法:采用UPLC-Q/TOF-MS/MS法对人参皂苷的成分进行鉴定。抗菌潜力预测通过计算重新利用和验证通过杀菌和协同试验。生物膜抑制,形态变化和细胞壁完整性进行了评估。综合代谢组学和转录组学研究了SGN-LZD的协同作用。关键发现随后通过RT-qPCR验证,并通过靶向药物抑制得到证实。最后,在小鼠皮肤感染模型和mellonia感染模型中评估其体内疗效。结果:共鉴定出五十二种成分。计算重目的分析表明,SGN能逆转败血症相关基因的失调,抑制临床分离的MRSA417的生长,相应的MIC和MBC分别为42.5和170 mg/mL(生药当量)。SGN与LZD表现出部分协同作用(FICI = 0.75)。联合用药对MRSA生物膜的抑制率为94.25%,而单用SGN的抑制率为40.40%。亚mic组合引起结构损伤,证明是细胞外碱性磷酸酶水平升高和细菌形态破坏。多组学分析显示,双组分系统(TCS)相关代谢物(如l -苹果酸)和基因(saeRS, vraSR)共同下调,表明TCS抑制是SGN-LZD协同作用的关键机制。该组合下调了关键的TCS mrna。此外,TCS抑制剂没有增强其抗生物膜和抗菌作用。值得注意的是,SGN-LZD联合用药提高了大鼠蒙氏杆菌的存活率,促进了小鼠皮肤感染模型的伤口愈合。结论:本研究为SGN重新定位治疗MRSA提供了证据。SGN损害tcs介导的毒力调节、细胞包膜完整性和必要的代谢途径,从而增强LZD的功效。这些发现为药物再利用提供了一个翻译框架,并突出了SGN-LZD联合治疗耐药感染的临床潜力。
{"title":"Unveiling TCM-antibiotic synergy through multi-omics: Shuganning injection potentiates the effect of linezolid against methicillin-resistant Staphylococcus aureus.","authors":"Qing Wang, Yali Wu, Yan Wang, Xin Pang, Mingliang Zhang, Ya Zhao, Xi Qin, Bowen Zheng, Mingyue Li, Xuyang Li, Weixia Li, Xiaoyan Wang, Liuqing Yang, Xiaofei Chen, Jinfa Tang","doi":"10.1016/j.phymed.2025.157733","DOIUrl":"10.1016/j.phymed.2025.157733","url":null,"abstract":"<p><strong>Background: </strong>Methicillin-resistant Staphylococcus aureus (MRSA) poses a significant threat to global public health due to its multidrug resistance, highlighting the need for the development of novel antimicrobial strategies. Computational repurposing has predicted that Shuganning injection (SGN), a clinically used hepatoprotective traditional Chinese medicine, exhibits synergistic activity with antibiotics against MRSA. However, the antimicrobial properties of SGN remain insufficiently characterized.</p><p><strong>Purpose: </strong>To evaluate the anti-MRSA activity of SGN and to elucidate the mechanism underlying its synergy with linezolid (LZD).</p><p><strong>Methods: </strong>The components of SGN were identified using UPLC-Q/TOF-MS/MS. Antimicrobial potential was predicted through computational repurposing and validated through bactericidal and synergy assays. Biofilm inhibition, morphological changes, and cell wall integrity were assessed. Integrated metabolomics and transcriptomics were employed to investigate SGN-LZD synergy. Key findings were subsequently validated by RT-qPCR and confirmed through targeted pharmacological inhibition. Finally, in vivo efficacy was assessed in Galleria mellonella and murine skin infection models.</p><p><strong>Results: </strong>Fifty-two components were identified in SGN. Computational repurposing analysis indicated that SGN reversed sepsis-associated gene dysregulation and inhibited the growth of clinically isolated MRSA417, with corresponding MIC and MBC values of 42.5 and 170 mg/mL (crude drug equivalent), respectively. SGN demonstrated partial synergy with LZD (FICI = 0.75). The combination markedly inhibited MRSA biofilm formation by 94.25%, compared to 40.40% for SGN alone. Sub-MIC combinations caused structural damage, evidenced by increased extracellular alkaline phosphatase levels and disrupted bacterial morphology. Multi-omics analysis indicated co-downregulation of two-component system (TCS)-related metabolites (e.g., L-malic acid) and genes (saeRS, vraSR), indicating TCS suppression as the key mechanism underlying SGN-LZD synergy. The combination downregulated key TCS mRNAs. Furthermore, TCS inhibitors did not enhance its anti-biofilm and antibacterial effects. Notably, the SGN-LZD combination improved survival in G. mellonella and promoted wound healing in a murine skin infection model.</p><p><strong>Conclusion: </strong>This study provides evidence supporting the repositioning of SGN for MRSA treatment. SGN impairs TCS-mediated virulence regulation, cell envelope integrity, and essential metabolic pathways, thereby enhancing the efficacy of LZD. These findings offer a translational framework for drug repurposing and highlight the clinical potential of SGN-LZD combination therapy for drug-resistant infections.</p>","PeriodicalId":20212,"journal":{"name":"Phytomedicine","volume":"150 ","pages":"157733"},"PeriodicalIF":8.3,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145878854","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Bruceantin inhibits the c-Myc/RL27A axis to suppress tumor progression in hepatocellular carcinoma 布鲁斯汀抑制c-Myc/RL27A轴抑制肝细胞癌的肿瘤进展
IF 8.3 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2026-01-01 DOI: 10.1016/j.phymed.2025.157740
Baitul Islam , Saro Adonira , Weiye Xu , Ceshi Chen , Haiyang Yu , Jufang Huang

Background

Hepatocellular carcinoma (HCC) is the most prevalent form of liver cancer and is associated with a poor prognosis. Current treatment options for advanced HCC remain limited, highlighting the need for more effective and safer therapies.

Purpose

This study aimed to elucidate the anti-cancer efficacy and explore the molecular mechanism of Bruceantin (BCT) against HCC.

Methods

We screened natural compounds using an extensive literature review and CCK-8 assays to identify novel therapeutic candidates. Using in vitro (HepG2.2.15 and Hep3B) and in vivo models, we assessed the BCT’s effects through CCK-8, colony formation, wound-healing, flow cytometry, Hematoxylin and eosin (H&E) staining, immunohistochemistry (IHC), qPCR, Western blot (WB), Luciferase reporter assay, and CETSA.

Results

Bruceantin (BCT) significantly inhibited HCC cell proliferation and migration and induced apoptosis. In mouse xenograft models, it markedly suppressed tumor growth without observable toxicity. Transcriptomic profiling revealed that BCT broadly downregulated ribosomal protein genes, with RPL27A showing the most notable reduction. Further investigations demonstrated that RPL27A plays a crucial role in supporting HCC cell survival and inhibiting apoptosis, and its high expression is clinically associated with poor prognosis. Mechanistically, BCT decreased RPL27A expression by inhibiting c-Myc's transcriptional activity, thus preventing its capacity to activate RPL27A transcription. This disruption of the c-Myc/RPL27A axis contributes to BCT's antitumor effects.

Conclusion

This study reveals a new molecular mechanism underlying BCT’s antitumor activity and supports its potential as a therapeutic agent for HCC.
背景:肝细胞癌(HCC)是最常见的肝癌,且预后较差。目前晚期HCC的治疗选择仍然有限,这表明需要更有效和更安全的治疗方法。目的研究Bruceantin (BCT)对肝癌的抗癌作用,探讨其分子机制。方法通过广泛的文献回顾和CCK-8测定来筛选天然化合物,以确定新的候选治疗药物。采用体外(HepG2.2.15和Hep3B)和体内模型,通过CCK-8、菌落形成、伤口愈合、流式细胞术、苏木精和伊红(H&;E)染色、免疫组织化学(IHC)、qPCR、Western blot (WB)、荧光素酶报告基因检测和CETSA等方法评估BCT的作用。结果bruceantin (BCT)能明显抑制肝癌细胞的增殖和迁移,诱导细胞凋亡。在小鼠异种移植模型中,它明显抑制肿瘤生长,无明显毒性。转录组学分析显示,BCT广泛下调核糖体蛋白基因,其中RPL27A的下调最为显著。进一步研究表明,RPL27A在支持HCC细胞存活和抑制细胞凋亡中起着至关重要的作用,其高表达在临床上与不良预后相关。从机制上讲,BCT通过抑制c-Myc的转录活性来降低RPL27A的表达,从而阻止其激活RPL27A转录的能力。这种c-Myc/RPL27A轴的破坏有助于BCT的抗肿瘤作用。结论本研究揭示了BCT抗肿瘤活性的新分子机制,支持其作为HCC治疗药物的潜力。
{"title":"Bruceantin inhibits the c-Myc/RL27A axis to suppress tumor progression in hepatocellular carcinoma","authors":"Baitul Islam ,&nbsp;Saro Adonira ,&nbsp;Weiye Xu ,&nbsp;Ceshi Chen ,&nbsp;Haiyang Yu ,&nbsp;Jufang Huang","doi":"10.1016/j.phymed.2025.157740","DOIUrl":"10.1016/j.phymed.2025.157740","url":null,"abstract":"<div><h3>Background</h3><div>Hepatocellular carcinoma (HCC) is the most prevalent form of liver cancer and is associated with a poor prognosis. Current treatment options for advanced HCC remain limited, highlighting the need for more effective and safer therapies.</div></div><div><h3>Purpose</h3><div>This study aimed to elucidate the anti-cancer efficacy and explore the molecular mechanism of Bruceantin (BCT) against HCC<em>.</em></div></div><div><h3>Methods</h3><div>We screened natural compounds using an extensive literature review and CCK-8 assays to identify novel therapeutic candidates. Using <em>in vitro</em> (HepG2.2.15 and Hep3B) and <em>in vivo</em> models, we assessed the BCT’s effects through CCK-8, colony formation, wound-healing, flow cytometry, Hematoxylin and eosin (H&amp;E) staining, immunohistochemistry (IHC), qPCR, Western blot (WB), Luciferase reporter assay, and CETSA.</div></div><div><h3>Results</h3><div>Bruceantin (BCT) significantly inhibited HCC cell proliferation and migration and induced apoptosis. In mouse xenograft models, it markedly suppressed tumor growth without observable toxicity. Transcriptomic profiling revealed that BCT broadly downregulated ribosomal protein genes, with RPL27A showing the most notable reduction. Further investigations demonstrated that RPL27A plays a crucial role in supporting HCC cell survival and inhibiting apoptosis, and its high expression is clinically associated with poor prognosis. Mechanistically, BCT decreased RPL27A expression by inhibiting c-Myc's transcriptional activity, thus preventing its capacity to activate RPL27A transcription. This disruption of the c-Myc/RPL27A axis contributes to BCT's antitumor effects.</div></div><div><h3>Conclusion</h3><div>This study reveals a new molecular mechanism underlying BCT’s antitumor activity and supports its potential as a therapeutic agent for HCC.</div></div>","PeriodicalId":20212,"journal":{"name":"Phytomedicine","volume":"150 ","pages":"Article 157740"},"PeriodicalIF":8.3,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145884162","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Shengmai San attenuates irradiation-induced salivary gland injury and fibrosis by inhibiting the SPHK1-S1P-S1PR1 axis 生脉散通过抑制SPHK1-S1P-S1PR1轴减轻辐照诱导的唾液腺损伤和纤维化
IF 8.3 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2026-01-01 DOI: 10.1016/j.phymed.2025.157742
Honglin Li , Guanru Wang , Yuchen Fan , Zhixuan Li , Hongxuan Wei , Suning Mao , Cheng Miao , Liu Liu , Junsheng Chen , Chunjie Li , Zhangfan Ding , Yubin Cao

Background

Radiation-induced salivary gland injury is a common complication of radiotherapy for head and neck tumors. The traditional Chinese herbal compound Shengmai San (SMS) can regulate Qi-Yin deficiency, promote fluid secretion, and alleviate thirst. However, its therapeutic effects on radiation-induced salivary gland damage remain unexplored.

Purpose

This study aimed to investigate the therapeutic efficacy of Shengmai San in irradiation-induced salivary gland injury, identify its active pharmaceutical components, and elucidate the underlying molecular mechanisms of radioprotection.

Material and methods

The natural drug components of Shengmai San were analyzed, and a murine model of irradiation-induced salivary gland injury was established. SMS extract was administered to irradiated mice, and the functional restoration of salivary glands was evaluated. Network pharmacology was employed to identify the active constituents of SMS, while molecular docking and protein-protein interaction analysis were used to screen key signaling pathways associated with glandular functional preservation. In vitro and in vivo experiments were conducted to validate these findings.

Results

Shengmai San significantly alleviated irradiation-induced salivary gland injury by promoting M2 macrophage polarization and reducing the levels of Interleukin-6 (IL-6) and Tumor Necrosis Factor-α (TNF-α) in serum and salivary gland tissues. It also ameliorated glandular fibrosis and inflammation. Network pharmacology analysis revealed that ophiopogonanone E was one of the primary active components, and molecular docking demonstrated its strong interaction with sphingosine kinase 1 (SPHK1) protein. In vivo experiments showed that SMS suppressed SPHK1 activity and sphingosine-1-phosphate (S1P) production in irradiated salivary glands. Additionally, SMS effectively inhibited the downstream receptor S1PR1. In vitro studies confirmed that SMS attenuated mitochondrial damage in acinar cells by inhibiting the SPHK1-S1P-S1PR1 axis and reduced glandular inflammation and oxidative stress by suppressing Nucleotide-binding Oligomerization Domain-like Receptor Family Pyrin Domain Containing 3 (NLRP3) inflammasome activation, thereby preserving salivary gland function.

Conclusion

Shengmai San effectively attenuates irradiation-induced salivary gland hypofunction and fibrosis, mainly through inhibition of the SPHK1-S1P-S1PR1 signaling pathway.
背景放射引起的唾液腺损伤是头颈部肿瘤放疗后常见的并发症。中药复方生脉散具有调节气阴虚虚、促进分泌液分泌、解渴的作用。然而,其对辐射引起的唾液腺损伤的治疗效果尚未研究。目的研究生脉散对辐照性唾液腺损伤的治疗作用,鉴定其有效药物成分,阐明其辐射防护的分子机制。材料与方法对生脉散天然药物成分进行分析,建立小鼠辐照性唾液腺损伤模型。用SMS提取物照射小鼠,观察其对唾液腺功能的恢复。通过网络药理学鉴定SMS的活性成分,通过分子对接和蛋白-蛋白相互作用分析筛选与腺体功能保存相关的关键信号通路。通过体外和体内实验验证了这些发现。结果生脉散通过促进M2巨噬细胞极化,降低血清和唾液腺组织中白细胞介素-6 (IL-6)、肿瘤坏死因子-α (TNF-α)水平,显著减轻辐照诱导的唾液腺损伤。它还能改善腺纤维化和炎症。网络药理学分析显示,麦冬皂酮E是其主要活性成分之一,分子对接显示其与鞘氨酸激酶1 (SPHK1)蛋白有强相互作用。体内实验表明,SMS抑制了辐照唾液腺SPHK1活性和鞘氨醇-1-磷酸(S1P)的产生。此外,SMS有效抑制下游受体S1PR1。体外研究证实,SMS通过抑制SPHK1-S1P-S1PR1轴减轻了腺泡细胞的线粒体损伤,并通过抑制核苷酸结合寡聚结构域样受体家族Pyrin结构域3 (NLRP3)炎性体的激活来减轻腺体炎症和氧化应激,从而保持唾液腺功能。结论生脉散主要通过抑制SPHK1-S1P-S1PR1信号通路,有效减轻辐照诱导的唾液腺功能减退和纤维化。
{"title":"Shengmai San attenuates irradiation-induced salivary gland injury and fibrosis by inhibiting the SPHK1-S1P-S1PR1 axis","authors":"Honglin Li ,&nbsp;Guanru Wang ,&nbsp;Yuchen Fan ,&nbsp;Zhixuan Li ,&nbsp;Hongxuan Wei ,&nbsp;Suning Mao ,&nbsp;Cheng Miao ,&nbsp;Liu Liu ,&nbsp;Junsheng Chen ,&nbsp;Chunjie Li ,&nbsp;Zhangfan Ding ,&nbsp;Yubin Cao","doi":"10.1016/j.phymed.2025.157742","DOIUrl":"10.1016/j.phymed.2025.157742","url":null,"abstract":"<div><h3>Background</h3><div>Radiation-induced salivary gland injury is a common complication of radiotherapy for head and neck tumors. The traditional Chinese herbal compound Shengmai San (SMS) can regulate Qi-Yin deficiency, promote fluid secretion, and alleviate thirst. However, its therapeutic effects on radiation-induced salivary gland damage remain unexplored.</div></div><div><h3>Purpose</h3><div>This study aimed to investigate the therapeutic efficacy of Shengmai San in irradiation-induced salivary gland injury, identify its active pharmaceutical components, and elucidate the underlying molecular mechanisms of radioprotection.</div></div><div><h3>Material and methods</h3><div>The natural drug components of Shengmai San were analyzed, and a murine model of irradiation-induced salivary gland injury was established. SMS extract was administered to irradiated mice, and the functional restoration of salivary glands was evaluated. Network pharmacology was employed to identify the active constituents of SMS, while molecular docking and protein-protein interaction analysis were used to screen key signaling pathways associated with glandular functional preservation. <em>In vitro</em> and <em>in vivo</em> experiments were conducted to validate these findings.</div></div><div><h3>Results</h3><div>Shengmai San significantly alleviated irradiation-induced salivary gland injury by promoting M2 macrophage polarization and reducing the levels of Interleukin-6 (IL-6) and Tumor Necrosis Factor-α (TNF-α) in serum and salivary gland tissues. It also ameliorated glandular fibrosis and inflammation. Network pharmacology analysis revealed that ophiopogonanone E was one of the primary active components, and molecular docking demonstrated its strong interaction with sphingosine kinase 1 (SPHK1) protein. <em>In vivo</em> experiments showed that SMS suppressed SPHK1 activity and sphingosine-1-phosphate (S1P) production in irradiated salivary glands. Additionally, SMS effectively inhibited the downstream receptor S1PR1. <em>In vitro</em> studies confirmed that SMS attenuated mitochondrial damage in acinar cells by inhibiting the SPHK1-S1P-S1PR1 axis and reduced glandular inflammation and oxidative stress by suppressing Nucleotide-binding Oligomerization Domain-like Receptor Family Pyrin Domain Containing 3 (NLRP3) inflammasome activation, thereby preserving salivary gland function.</div></div><div><h3>Conclusion</h3><div>Shengmai San effectively attenuates irradiation-induced salivary gland hypofunction and fibrosis, mainly through inhibition of the SPHK1-S1P-S1PR1 signaling pathway.</div></div>","PeriodicalId":20212,"journal":{"name":"Phytomedicine","volume":"150 ","pages":"Article 157742"},"PeriodicalIF":8.3,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145884121","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Jiangtang Tiaozhi formula improves obesity and insulin resistance by inhibiting skeletal muscle inflammation and pyroptosis in diet-induced obese mice 降汤调脂方通过抑制饮食性肥胖小鼠骨骼肌炎症和焦亡改善肥胖和胰岛素抵抗
IF 8.3 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2026-01-01 DOI: 10.1016/j.phymed.2025.157750
Hongyu Qi , Zepeng Zhang , Tong Hou , Lu Ding , Qing Liu , Ying Wang , Siyu Song , Jiaqi Liu , Chang Sun , Yahui Sun , Zeyu Wang , Fengmei Lian , Xiangyan Li

Background

The global prevalence of obesity continues to rise annually. Jiangtang Tiaozhi (JTTZ) formula, a traditional Chinese medicine formula, is clinically utilized for treating obesity and type 2 diabetes due to its therapeutic efficacy. However, the underlying mechanisms of JTTZ formula for improving obesity and insulin resistance (IR) remain unclear.

Purpose

This study aimed to elucidate the mechanistic basis of JTTZ formula's anti-obesity effects, specifically evaluating its role in ameliorating IR and reducing body weight.

Methods

High-fat diet (HFD)- and high-fat and high-sucrose diet (HFHSD)-induced obese (DIO) mice were assessed for determining the effects of JTTZ formula on body weights, IR, serum lipid profiles, and inflammatory markers in the serum and gastrocnemius muscle. Transcriptomic analysis of the gastrocnemius tissues of HFD-fed mice was performed, followed by RT-qPCR analysis and immunofluorescence staining to evaluate the changes of inflammation- and pyroptosis-related genes/proteins. Molecular docking and surface plasmon resonance assays were used to assess the binding affinity of 12 blood components derived from JTTZ formula to the pyroptosis-associated protein, NLRP3. In addition, the influences of JTTZ formula and its bioactive compounds on cell viability, lipid accumulation, inflammatory cytokine release, and pyroptosis were determined in the palmitic acid (PA)-induced C2C12 and L6 myotubes model of IR.

Results

JTTZ formula significantly reduced the body weight, adiposity, and improved IR of HFD- and HFHSD-induced DIO mice while improving gastrocnemius muscle morphology and suppressing systemic and muscular inflammation. Transcriptomics and molecular assays revealed that JTTZ formula could regulate the tumor necrosis factor, NOD-like receptor, and nuclear factor kappa B signaling pathways to inhibit inflammation and pyroptosis for the improvement of IR in muscles. Among the various components of JTTZ formula, cryptotanshinone, diosgenin, and timosaponin A1 exhibited significant NLRP3-binding affinity. The active constituents of JTTZ formula including berberine, cryptotanshinone, neomangiferin, and palmatine attenuated PA-induced IR, lipid deposition, inflammation, and pyroptosis in C2C12 and L6 myotubes.

Conclusion

JTTZ formula could ameliorate obesity-associated metabolic dysfunction and improve IR by enhancing glucose uptake, reducing lipid accumulation in skeletal muscles. Its effects were mediated by attenuating inflammation and pyroptosis in muscle tissues. Berberine, cryptotanshinone, neomangiferin, and palmatine might be the bioactive components of JTTZ formula against obesity-related IR.
全球肥胖患病率每年都在持续上升。降汤调脂方(JTTZ)是一种中药配方,因其治疗肥胖症和2型糖尿病的疗效而在临床上被广泛使用。然而,JTTZ配方改善肥胖和胰岛素抵抗(IR)的潜在机制尚不清楚。目的本研究旨在阐明JTTZ方抗肥胖作用的机制基础,具体评价其改善IR和减轻体重的作用。方法采用高脂日粮(HFD)和高脂高糖日粮(HFHSD)诱导的肥胖(DIO)小鼠,观察JTTZ方对小鼠体重、IR、血脂、血清及腓肠肌炎症指标的影响。采用RT-qPCR和免疫荧光染色对饲喂hfd小鼠的腓肠肌组织进行转录组学分析,评估炎症和焦热相关基因/蛋白的变化。采用分子对接和表面等离子体共振方法评估了JTTZ配方衍生的12种血液成分与焦热相关蛋白NLRP3的结合亲和力。此外,在棕榈酸(PA)诱导的IR C2C12和L6肌管模型中,测定JTTZ方及其生物活性化合物对细胞活力、脂质积累、炎性细胞因子释放和焦亡的影响。结果jttz方能显著降低HFD和hfhsd诱导的DIO小鼠的体重、脂肪,改善IR,改善腓肠肌形态,抑制全身和肌肉炎症。转录组学和分子分析显示,JTTZ方可调节肿瘤坏死因子、nod样受体和核因子κ B信号通路,抑制炎症和焦亡,改善肌肉IR。在JTTZ组份中,隐丹参酮、薯蓣皂苷元和地骨皂苷A1表现出显著的nlrp3结合亲和力。JTTZ配方的有效成分包括小檗碱、隐丹参酮、新金盏花素和棕榈碱,可减轻pa诱导的C2C12和L6肌管IR、脂质沉积、炎症和焦亡。结论jttz方可通过提高骨骼肌葡萄糖摄取,减少脂质积累,改善肥胖相关代谢功能障碍,改善IR。其作用是通过减轻肌肉组织的炎症和焦亡介导的。小檗碱、隐丹参酮、新桂皮素和棕榈碱可能是JTTZ配方抗肥胖相关IR的生物活性成分。
{"title":"Jiangtang Tiaozhi formula improves obesity and insulin resistance by inhibiting skeletal muscle inflammation and pyroptosis in diet-induced obese mice","authors":"Hongyu Qi ,&nbsp;Zepeng Zhang ,&nbsp;Tong Hou ,&nbsp;Lu Ding ,&nbsp;Qing Liu ,&nbsp;Ying Wang ,&nbsp;Siyu Song ,&nbsp;Jiaqi Liu ,&nbsp;Chang Sun ,&nbsp;Yahui Sun ,&nbsp;Zeyu Wang ,&nbsp;Fengmei Lian ,&nbsp;Xiangyan Li","doi":"10.1016/j.phymed.2025.157750","DOIUrl":"10.1016/j.phymed.2025.157750","url":null,"abstract":"<div><h3>Background</h3><div>The global prevalence of obesity continues to rise annually. Jiangtang Tiaozhi (JTTZ) formula, a traditional Chinese medicine formula, is clinically utilized for treating obesity and type 2 diabetes due to its therapeutic efficacy. However, the underlying mechanisms of JTTZ formula for improving obesity and insulin resistance (IR) remain unclear.</div></div><div><h3>Purpose</h3><div>This study aimed to elucidate the mechanistic basis of JTTZ formula's anti-obesity effects, specifically evaluating its role in ameliorating IR and reducing body weight.</div></div><div><h3>Methods</h3><div>High-fat diet (HFD)- and high-fat and high-sucrose diet (HFHSD)-induced obese (DIO) mice were assessed for determining the effects of JTTZ formula on body weights, IR, serum lipid profiles, and inflammatory markers in the serum and gastrocnemius muscle. Transcriptomic analysis of the gastrocnemius tissues of HFD-fed mice was performed, followed by RT-qPCR analysis and immunofluorescence staining to evaluate the changes of inflammation- and pyroptosis-related genes/proteins. Molecular docking and surface plasmon resonance assays were used to assess the binding affinity of 12 blood components derived from JTTZ formula to the pyroptosis-associated protein, NLRP3. In addition, the influences of JTTZ formula and its bioactive compounds on cell viability, lipid accumulation, inflammatory cytokine release, and pyroptosis were determined in the palmitic acid (PA)-induced C2C12 and L6 myotubes model of IR.</div></div><div><h3>Results</h3><div>JTTZ formula significantly reduced the body weight, adiposity, and improved IR of HFD- and HFHSD-induced DIO mice while improving gastrocnemius muscle morphology and suppressing systemic and muscular inflammation. Transcriptomics and molecular assays revealed that JTTZ formula could regulate the tumor necrosis factor, NOD-like receptor, and nuclear factor kappa B signaling pathways to inhibit inflammation and pyroptosis for the improvement of IR in muscles. Among the various components of JTTZ formula, cryptotanshinone, diosgenin, and timosaponin A1 exhibited significant NLRP3-binding affinity. The active constituents of JTTZ formula including berberine, cryptotanshinone, neomangiferin, and palmatine attenuated PA-induced IR, lipid deposition, inflammation, and pyroptosis in C2C12 and L6 myotubes.</div></div><div><h3>Conclusion</h3><div>JTTZ formula could ameliorate obesity-associated metabolic dysfunction and improve IR by enhancing glucose uptake, reducing lipid accumulation in skeletal muscles. Its effects were mediated by attenuating inflammation and pyroptosis in muscle tissues. Berberine, cryptotanshinone, neomangiferin, and palmatine might be the bioactive components of JTTZ formula against obesity-related IR.</div></div>","PeriodicalId":20212,"journal":{"name":"Phytomedicine","volume":"150 ","pages":"Article 157750"},"PeriodicalIF":8.3,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145925104","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Ginsenoside Rd promotes cardiac regeneration through PPARG/HMGCS2-driven ketone body metabolic reprogramming in myocardial ischemia-reperfusion injury. 人参皂苷Rd通过PPARG/ hmgcs2驱动的酮体代谢重编程促进心肌缺血再灌注损伤的心脏再生。
IF 8.3 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2026-01-01 Epub Date: 2025-12-11 DOI: 10.1016/j.phymed.2025.157694
Han Zhang, Ao Lin, Chenguang Zhai, Tianli Lai, Xueling Gong, Mengyuan Wang, Hui Wu, Taochun Ye, Lingjun Wang, Dina Tawulie
<p><strong>Background: </strong>Myocardial ischemia-reperfusion injury (MIRI) drives adverse cardiac remodeling and ventricular dysfunction, posing a major therapeutic challenge and substantially contributing to global mortality. Despite therapeutic advances, effective MIRI treatments remain limited. Ginsenoside Rd (GSRd), a bioactive constituent from traditional Chinese herbs, has been widely recognized to have cardioprotective effects. However, the role of GSRd in MIRI remains unclear.</p><p><strong>Purpose: </strong>To elucidate the therapeutic efficacy and the underlying molecular mechanisms of GSRd against MIRI.</p><p><strong>Methods: </strong>A MIRI model was established in male C57BL/6J mice via left anterior descending coronary artery (LAD) ligation followed by reperfusion. Post-surgery, mice received daily intraperitoneal injections of vehicle, dapagliflozin (1 mg/kg), or GSRd (5, 10, 20 mg/kg) for 28 days. Cardiac function was evaluated by echocardiography. Histopathological changes were assessed using hematoxylin and eosin (H&E), Masson's trichrome, TUNEL, and immunofluorescence staining. In vitro, isolated adult mouse CMs were subjected to H/R injury and GSRd-containing serum treatment to assess proliferation. Cardiomyocyte proliferation was assessed by Ki-67 immunofluorescence and BrdU flow cytometry. Integrated cardiac untargeted metabolomics (UPLC-MS/MS) and transcriptomics (RNA-seq) were conducted to identify differential metabolites and genes following GSRd intervention. Key targets were validated by RT-qPCR and western blotting. Adeno-associated virus9 (AAV9) with cardiac-specific 3-hydroxy-3-methylglutaryl-CoA synthase 2 (Hmgcs2) knockdown and overexpression were employed to confirm the therapeutic targets of GSRd against MIRI. In addition, transcription factor prediction was performed by multi-platform database analysis, with subsequent chromatin immunoprecipitation-qPCR (ChIP-qPCR) providing mechanistic validation. Furthermore, molecular docking, dynamics simulation, and surface plasmon resonance (SPR) were implemented to evaluate the binding capacity between GSRd and peroxisome proliferator-activated receptor gamma (PPARG). Additionally, GW9662, a PPARG inhibitor, was used to determine the dependence of GSRd-mediated cardioprotection against MIRI on the PPARG/HMGCS2 signaling pathway.</p><p><strong>Results: </strong>The results revealed that GSRd intervention substantially improved cardiac dysfunction, attenuated ventricular remodeling, ameliorated myocardial pathology, and suppressed inflammatory cytokine and oxidative stress levels in MIRI mice. Mechanistically, multi-omics analysis demonstrated enrichment in ketone body synthesis, carnitine and lipid metabolism, and PPAR signaling after GSRd treatment. Cardiac untargeted metabolomics indicated that GSRd alleviated metabolic dysregulation, concomitant with increased cardiac β-hydroxybutyrate (β-OHB). Transcriptomics identified upregulated ketogenic enzyme gene Hmgcs2 follo
背景:心肌缺血再灌注损伤(MIRI)驱动不良的心脏重塑和心室功能障碍,构成了主要的治疗挑战,并在很大程度上导致了全球死亡率。尽管治疗取得了进步,但有效的MIRI治疗仍然有限。人参皂苷Rd (Ginsenoside Rd, GSRd)是一种从传统中草药中提取的生物活性成分,具有广泛的心脏保护作用。然而,GSRd在MIRI中的作用尚不清楚。目的:探讨GSRd治疗MIRI的疗效及潜在的分子机制。方法:采用左冠状动脉前降支结扎后再灌注的方法建立雄性C57BL/6J小鼠MIRI模型。术后小鼠每天腹腔注射载体、达格列净(1 mg/kg)或GSRd(5、10、20 mg/kg),持续28天。超声心动图评价心功能。采用苏木精和伊红(H&E)、马松三色、TUNEL和免疫荧光染色评估组织病理学变化。体外,对离体成年小鼠CMs进行H/R损伤和含gsrd的血清处理,以评估其增殖情况。采用Ki-67免疫荧光和BrdU流式细胞术检测心肌细胞增殖情况。采用心脏非靶向代谢组学(UPLC-MS/MS)和转录组学(RNA-seq)来鉴定GSRd干预后的差异代谢物和基因。通过RT-qPCR和western blotting对关键靶点进行验证。采用心脏特异性3-羟基-3-甲基戊二酰辅酶a合成酶2 (Hmgcs2)敲低和过表达的腺相关病毒9 (AAV9)来证实GSRd治疗MIRI的靶点。此外,转录因子预测通过多平台数据库分析进行,随后的染色质免疫沉淀- qpcr (ChIP-qPCR)提供机制验证。此外,通过分子对接、动力学模拟和表面等离子体共振(SPR)来评估GSRd与过氧化物酶体增殖体激活受体γ (PPARG)的结合能力。此外,GW9662,一种PPARG抑制剂,被用来确定gsrd介导的心脏保护对MIRI的依赖于PPARG/HMGCS2信号通路。结果:结果显示,GSRd干预显著改善了MIRI小鼠的心功能障碍,减轻了心室重构,改善了心肌病理,抑制了炎症细胞因子和氧化应激水平。机制上,多组学分析表明,GSRd治疗后酮体合成、肉碱和脂质代谢以及PPAR信号的富集。心脏非靶向代谢组学表明,GSRd减轻了代谢失调,同时增加了心脏β-羟基丁酸(β-OHB)。转录组学鉴定了GSRd干预后生酮酶基因Hmgcs2上调。关键是在梗死区和边界区,GSRd同时上调HMGCS2表达和β-OHB水平,同时增强心肌细胞增殖。此外,在MIRI小鼠中,心脏特异性Hmgcs2敲低显著损害心肌细胞再生并减弱GSRd的心脏保护作用。相反,心脏特异性Hmgcs2过表达促进心肌细胞增殖,重现GSRd的心脏保护作用。转录因子预测和ChIP-qPCR分析证实了PPARG与Hmgcs2启动子区域的直接结合。分子对接、动力学模拟和SPR证实了GSRd与ppar之间的高亲和力结合。此外,GW9662对PPARG的抑制显著抑制了HMGCS2的表达,抑制了心脏再生,抵消了GSRd的心脏保护作用,确立了PPARG/HMGCS2轴的重要作用。结论:总的来说,本研究表明GSRd通过PPARG/ hmgcs2驱动的酮体代谢重编程促进心脏再生,从而改善MIRI。因此,这些发现可能为临床医生提供MIRI管理的新治疗视角。
{"title":"Ginsenoside Rd promotes cardiac regeneration through PPARG/HMGCS2-driven ketone body metabolic reprogramming in myocardial ischemia-reperfusion injury.","authors":"Han Zhang, Ao Lin, Chenguang Zhai, Tianli Lai, Xueling Gong, Mengyuan Wang, Hui Wu, Taochun Ye, Lingjun Wang, Dina Tawulie","doi":"10.1016/j.phymed.2025.157694","DOIUrl":"10.1016/j.phymed.2025.157694","url":null,"abstract":"&lt;p&gt;&lt;strong&gt;Background: &lt;/strong&gt;Myocardial ischemia-reperfusion injury (MIRI) drives adverse cardiac remodeling and ventricular dysfunction, posing a major therapeutic challenge and substantially contributing to global mortality. Despite therapeutic advances, effective MIRI treatments remain limited. Ginsenoside Rd (GSRd), a bioactive constituent from traditional Chinese herbs, has been widely recognized to have cardioprotective effects. However, the role of GSRd in MIRI remains unclear.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Purpose: &lt;/strong&gt;To elucidate the therapeutic efficacy and the underlying molecular mechanisms of GSRd against MIRI.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Methods: &lt;/strong&gt;A MIRI model was established in male C57BL/6J mice via left anterior descending coronary artery (LAD) ligation followed by reperfusion. Post-surgery, mice received daily intraperitoneal injections of vehicle, dapagliflozin (1 mg/kg), or GSRd (5, 10, 20 mg/kg) for 28 days. Cardiac function was evaluated by echocardiography. Histopathological changes were assessed using hematoxylin and eosin (H&E), Masson's trichrome, TUNEL, and immunofluorescence staining. In vitro, isolated adult mouse CMs were subjected to H/R injury and GSRd-containing serum treatment to assess proliferation. Cardiomyocyte proliferation was assessed by Ki-67 immunofluorescence and BrdU flow cytometry. Integrated cardiac untargeted metabolomics (UPLC-MS/MS) and transcriptomics (RNA-seq) were conducted to identify differential metabolites and genes following GSRd intervention. Key targets were validated by RT-qPCR and western blotting. Adeno-associated virus9 (AAV9) with cardiac-specific 3-hydroxy-3-methylglutaryl-CoA synthase 2 (Hmgcs2) knockdown and overexpression were employed to confirm the therapeutic targets of GSRd against MIRI. In addition, transcription factor prediction was performed by multi-platform database analysis, with subsequent chromatin immunoprecipitation-qPCR (ChIP-qPCR) providing mechanistic validation. Furthermore, molecular docking, dynamics simulation, and surface plasmon resonance (SPR) were implemented to evaluate the binding capacity between GSRd and peroxisome proliferator-activated receptor gamma (PPARG). Additionally, GW9662, a PPARG inhibitor, was used to determine the dependence of GSRd-mediated cardioprotection against MIRI on the PPARG/HMGCS2 signaling pathway.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Results: &lt;/strong&gt;The results revealed that GSRd intervention substantially improved cardiac dysfunction, attenuated ventricular remodeling, ameliorated myocardial pathology, and suppressed inflammatory cytokine and oxidative stress levels in MIRI mice. Mechanistically, multi-omics analysis demonstrated enrichment in ketone body synthesis, carnitine and lipid metabolism, and PPAR signaling after GSRd treatment. Cardiac untargeted metabolomics indicated that GSRd alleviated metabolic dysregulation, concomitant with increased cardiac β-hydroxybutyrate (β-OHB). Transcriptomics identified upregulated ketogenic enzyme gene Hmgcs2 follo","PeriodicalId":20212,"journal":{"name":"Phytomedicine","volume":"150 ","pages":"157694"},"PeriodicalIF":8.3,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145828000","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Artesunate alleviated DSS induced colitis via inhibiting M1 polarization through directly targeting INSR/mTORC1/HIF-1α pathway. 青蒿琥酯通过直接靶向INSR/mTORC1/HIF-1α通路抑制M1极化减轻DSS诱导的结肠炎。
IF 8.3 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2026-01-01 Epub Date: 2025-12-30 DOI: 10.1016/j.phymed.2025.157752
Junjie Huang, Ruigang Zhou, Weilong Peng, Weimei Wang, Jun Chen, Chenglong Yu, Lianggang Wang, Mingjiang Liu, Ruonan Bo, Jingui Li

Introduction: Artesunate (ARS) has demonstrated therapeutic potential in experimental ulcerative colitis (UC) through immunomodulatory activities. However, the role of macrophages and the underlying mechanisms remain unclear.

Methods: In this study, a DSS-induced UC model was established, along with macrophage depletion, polarized macrophage reinfusion, and Transwell coculture systems. The typical colitis characteristics, including weight change, disease activity index, inflammation, and tissue damage, were systematically assessed. Key proteins in the mTORC1/HIF-1α pathway were analyzed. SPR-MS was employed to identify ARS targets in M1 macrophages.

Results: This study indicated that ARS markedly suppressed DSS-enhanced M1 polarization and macrophage infiltration. In a Caco2-BMDM coculture system, ARS reduced macrophage migration and inflammation. Macrophage depletion significantly attenuated the anti-colitis effect of ARS. Compared with M1 reinfusion, ARS-pretreated M1 cells alleviated UC symptoms. ARS also inhibited LPS- or DSS-induced M1 polarization and proinflammatory cytokine upregulation in vivo and in vitro, linked to mTORC1/HIF-1α signaling, as confirmed by the agonist Leucine (LEU). Moreover, ARS protected intestinal barrier function by preventing tight junction loss and permeability increases via suppression of M1 polarization. SPR-MS and molecular docking revealed that ARS directly activated INSR, inhibiting mTORC1/HIF-1α-driven glycolytic M1 polarization.

Conclusions: Overall, macrophages are essential for ARS-mediated protection in UC. ARS alleviates UC by reprogramming macrophage polarization via the INSR/mTORC1/HIF-1α axis, providing new mechanistic insights and potential clinical applications.

青蒿琥酯(ARS)已通过免疫调节活性显示出治疗实验性溃疡性结肠炎(UC)的潜力。然而,巨噬细胞的作用及其潜在机制尚不清楚。方法:本研究建立dss诱导UC模型,并采用巨噬细胞耗竭、极化巨噬细胞回输和Transwell共培养系统。系统评估结肠炎的典型特征,包括体重变化、疾病活动性指数、炎症和组织损伤。分析mTORC1/HIF-1α通路中的关键蛋白。采用SPR-MS技术鉴定M1巨噬细胞中的ARS靶点。结果:本研究表明,ARS可明显抑制dss增强的M1极化和巨噬细胞浸润。在Caco2-BMDM共培养系统中,ARS可减少巨噬细胞迁移和炎症。巨噬细胞耗竭显著减弱ARS的抗结肠炎作用。与M1回输相比,经ars预处理的M1细胞减轻了UC症状。ARS还抑制LPS或dss诱导的M1极化和促炎细胞因子在体内和体外的上调,这与mTORC1/HIF-1α信号通路有关,激动剂亮氨酸(Leucine)证实了这一点。此外,ARS通过抑制M1极化防止紧密连接丢失和通透性增加来保护肠屏障功能。SPR-MS和分子对接发现ARS直接激活INSR,抑制mTORC1/ hif -1α驱动的糖酵解M1极化。结论:总体而言,巨噬细胞对UC中ars介导的保护至关重要。ARS通过INSR/mTORC1/HIF-1α轴重编程巨噬细胞极化减轻UC,提供了新的机制见解和潜在的临床应用。
{"title":"Artesunate alleviated DSS induced colitis via inhibiting M1 polarization through directly targeting INSR/mTORC1/HIF-1α pathway.","authors":"Junjie Huang, Ruigang Zhou, Weilong Peng, Weimei Wang, Jun Chen, Chenglong Yu, Lianggang Wang, Mingjiang Liu, Ruonan Bo, Jingui Li","doi":"10.1016/j.phymed.2025.157752","DOIUrl":"10.1016/j.phymed.2025.157752","url":null,"abstract":"<p><strong>Introduction: </strong>Artesunate (ARS) has demonstrated therapeutic potential in experimental ulcerative colitis (UC) through immunomodulatory activities. However, the role of macrophages and the underlying mechanisms remain unclear.</p><p><strong>Methods: </strong>In this study, a DSS-induced UC model was established, along with macrophage depletion, polarized macrophage reinfusion, and Transwell coculture systems. The typical colitis characteristics, including weight change, disease activity index, inflammation, and tissue damage, were systematically assessed. Key proteins in the mTORC1/HIF-1α pathway were analyzed. SPR-MS was employed to identify ARS targets in M1 macrophages.</p><p><strong>Results: </strong>This study indicated that ARS markedly suppressed DSS-enhanced M1 polarization and macrophage infiltration. In a Caco2-BMDM coculture system, ARS reduced macrophage migration and inflammation. Macrophage depletion significantly attenuated the anti-colitis effect of ARS. Compared with M1 reinfusion, ARS-pretreated M1 cells alleviated UC symptoms. ARS also inhibited LPS- or DSS-induced M1 polarization and proinflammatory cytokine upregulation in vivo and in vitro, linked to mTORC1/HIF-1α signaling, as confirmed by the agonist Leucine (LEU). Moreover, ARS protected intestinal barrier function by preventing tight junction loss and permeability increases via suppression of M1 polarization. SPR-MS and molecular docking revealed that ARS directly activated INSR, inhibiting mTORC1/HIF-1α-driven glycolytic M1 polarization.</p><p><strong>Conclusions: </strong>Overall, macrophages are essential for ARS-mediated protection in UC. ARS alleviates UC by reprogramming macrophage polarization via the INSR/mTORC1/HIF-1α axis, providing new mechanistic insights and potential clinical applications.</p>","PeriodicalId":20212,"journal":{"name":"Phytomedicine","volume":"150 ","pages":"157752"},"PeriodicalIF":8.3,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145900972","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Cannabidiol improves cognitive impairment after traumatic brain injury by attenuating neuronal oxidative stress and apoptosis via the SET/PP2A/Akt signaling axis. 大麻二酚通过SET/PP2A/Akt信号轴减轻神经元氧化应激和凋亡,改善创伤性脑损伤后的认知功能障碍。
IF 8.3 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2026-01-01 Epub Date: 2026-01-03 DOI: 10.1016/j.phymed.2026.157769
Shan Gao, Zhennan Xiao, Hongtao Liu

Background: Cognitive impairment is a major complication of traumatic brain injury (TBI), yet effective therapies remain lacking. As a natural compound extracted from Cannabis sativa, cannabidiol (CBD) possesses antioxidant properties and has shown neuroprotective potential in several neurological disorders. However, its effects in cognitive impairment after TBI remain unclear.

Purpose: This study aimed to investigate the therapeutic effects of CBD on cognitive impairment after TBI and elucidate its underlying molecular mechanisms.

Study design: In vitro H2O2 model and in vivo TBI model were used to evaluate the neuroprotective effects of CBD.

Methods: Neuronal oxidative stress models induced by H2O2 and controlled cortical impact model were used to detect the neuroprotective effects of CBD. Western blotting, histological staining, and biochemical assays were employed to investigate the effects of CBD on oxidative stress and apoptosis in neurons. RNA-Seq analysis, co-immunoprecipitation, molecular dynamics simulations, CETSA, SPR and immunofluorescence were performed to elucidate the molecular mechanisms.

Results: CBD can inhibit neuronal oxidative stress and apoptosis both in vivo and in vitro. Mechanistically, we identify a novel SET/PP2A/Akt signaling axis, in which CBD directly bound to SET, induced conformational changes in its nuclear localization signal and promoted its retention in the cytoplasm. Elevated cytoplasmic SET suppresses PP2A activity, activates Akt signaling pathway, and inhibits oxidative stress and pro-apoptotic cascades, promoting neuronal survival.

Conclusion: CBD exerts its neuroprotective effects by inhibiting neuronal oxidative stress and apoptosis through SET/PP2A/Akt signaling axis. These findings provide a novel potential drug target for the treatment of cognitive impairment after TBI.

背景:认知障碍是创伤性脑损伤(TBI)的主要并发症,但目前缺乏有效的治疗方法。大麻二酚(cannabidiol, CBD)是一种从大麻中提取的天然化合物,具有抗氧化作用,对多种神经系统疾病具有保护作用。然而,其对脑外伤后认知障碍的影响尚不清楚。目的:探讨CBD对脑外伤后认知功能障碍的治疗作用,并阐明其分子机制。研究设计:采用体外H2O2模型和体内TBI模型评价CBD的神经保护作用。方法:采用H2O2诱导的神经元氧化应激模型和控制性皮质冲击模型检测CBD的神经保护作用。采用Western blotting、组织学染色、生化等方法观察CBD对神经元氧化应激和凋亡的影响。通过RNA-Seq分析、共免疫沉淀、分子动力学模拟、CETSA、SPR和免疫荧光等手段阐明其分子机制。结果:CBD在体内和体外均能抑制神经元氧化应激和细胞凋亡。在机制上,我们发现了一个新的SET/PP2A/Akt信号轴,其中CBD直接与SET结合,诱导其核定位信号的构象变化并促进其在细胞质中的保留。胞质SET升高可抑制PP2A活性,激活Akt信号通路,抑制氧化应激和促凋亡级联反应,促进神经元存活。结论:CBD通过SET/PP2A/Akt信号轴抑制神经元氧化应激和细胞凋亡发挥神经保护作用。这些发现为脑外伤后认知功能障碍的治疗提供了一个新的潜在药物靶点。
{"title":"Cannabidiol improves cognitive impairment after traumatic brain injury by attenuating neuronal oxidative stress and apoptosis via the SET/PP2A/Akt signaling axis.","authors":"Shan Gao, Zhennan Xiao, Hongtao Liu","doi":"10.1016/j.phymed.2026.157769","DOIUrl":"10.1016/j.phymed.2026.157769","url":null,"abstract":"<p><strong>Background: </strong>Cognitive impairment is a major complication of traumatic brain injury (TBI), yet effective therapies remain lacking. As a natural compound extracted from Cannabis sativa, cannabidiol (CBD) possesses antioxidant properties and has shown neuroprotective potential in several neurological disorders. However, its effects in cognitive impairment after TBI remain unclear.</p><p><strong>Purpose: </strong>This study aimed to investigate the therapeutic effects of CBD on cognitive impairment after TBI and elucidate its underlying molecular mechanisms.</p><p><strong>Study design: </strong>In vitro H<sub>2</sub>O<sub>2</sub> model and in vivo TBI model were used to evaluate the neuroprotective effects of CBD.</p><p><strong>Methods: </strong>Neuronal oxidative stress models induced by H<sub>2</sub>O<sub>2</sub> and controlled cortical impact model were used to detect the neuroprotective effects of CBD. Western blotting, histological staining, and biochemical assays were employed to investigate the effects of CBD on oxidative stress and apoptosis in neurons. RNA-Seq analysis, co-immunoprecipitation, molecular dynamics simulations, CETSA, SPR and immunofluorescence were performed to elucidate the molecular mechanisms.</p><p><strong>Results: </strong>CBD can inhibit neuronal oxidative stress and apoptosis both in vivo and in vitro. Mechanistically, we identify a novel SET/PP2A/Akt signaling axis, in which CBD directly bound to SET, induced conformational changes in its nuclear localization signal and promoted its retention in the cytoplasm. Elevated cytoplasmic SET suppresses PP2A activity, activates Akt signaling pathway, and inhibits oxidative stress and pro-apoptotic cascades, promoting neuronal survival.</p><p><strong>Conclusion: </strong>CBD exerts its neuroprotective effects by inhibiting neuronal oxidative stress and apoptosis through SET/PP2A/Akt signaling axis. These findings provide a novel potential drug target for the treatment of cognitive impairment after TBI.</p>","PeriodicalId":20212,"journal":{"name":"Phytomedicine","volume":"150 ","pages":"157769"},"PeriodicalIF":8.3,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145945544","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Phytomedicine
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1