首页 > 最新文献

Phytomedicine最新文献

英文 中文
Balanophora involucrata alleviates diabetic nephropathy by inhibiting ferroptosis, modulating serum metabolites and gut microbiota 天秤子通过抑制铁下垂、调节血清代谢物和肠道菌群减轻糖尿病肾病
IF 8.3 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2026-01-05 DOI: 10.1016/j.phymed.2026.157773
Chaoxi Tian , Yawen Chen , Jing Huang , Fangyu Zhao , Xiane Tang , Jianhong Gao , Xianbing Chen , Xiuxue Yuan

Background

Diabetic nephropathy (DN) serves as the major etiological factor of end-stage renal disease (ESRD) in DM patients. Balanophora involucrata Hook.f. & Thomson (BI) is a traditional medicine of the Tujia ethnic group in China, with good therapeutic effects on kidney diseases. However, both its effectiveness in the context of DN and the regulatory mechanism remain to be further clarified.

Purpose

To elucidate the role of BI in DN mice, with a focus on exploring the intrinsic mechanism responsible for this process.

Methods

UPLC-MS/MS was employed to identify the main ingredients of BI. The DN model was established using C57BLKS/J db/db mice, which received BI treatment. Serum/urine parameters, renal histology, and ultrastructure were assessed. Intrinsic microbial composition and corresponding metabolites were analyzed via 16S rDNA gene sequencing and nontargeted metabolomics. HK-2 cells were treated with high glucose to simulate DN and then treated with BI-containing drug serum and ferrostatin-1 (fer-1). Cell viability was assessed by CCK-8 assay, and lipid peroxidation was evaluated using the Bodipy 581/591 C11 fluorescence assay. Commercial kits were used to detect MDA and other ferroptosis-related indicators. Protein levels were analyzed by Western blotting.

Results

Terpenoids and flavonoids were the main identified components of BI. Fasting blood glucose was reduced, and lipid metabolism disorders were corrected by BI in DN mice. It also ameliorated renal injury in these mice. Meanwhile, BI regulated serum metabolites, and remodeled gut microbiota in DN mice. Furthermore, Nrf2, GPX4, FPN1, FTH1, and SLC7A11 were activated by BI, whereas TFR1 and ACSL4 were suppressed.

Conclusions

BI ameliorates DN by regulating serum metabolites, remodeling the gut microbiota, and inhibiting ferroptosis.
背景:糖尿病肾病(DN)是DM患者终末期肾病(ESRD)的主要病因。凤仙花汤姆逊(BI)是中国土家族的一种传统药物,对肾脏疾病有很好的治疗效果。然而,其在DN背景下的有效性和调控机制仍有待进一步阐明。目的阐明BI在DN小鼠中的作用,重点探讨其内在机制。方法采用高效液相色谱-质谱联用技术对其主要成分进行鉴定。采用C57BLKS/ jdb /db小鼠,经BI治疗后建立DN模型。评估血清/尿液参数、肾脏组织学和超微结构。通过16S rDNA基因测序和非靶向代谢组学分析内在微生物组成和相应的代谢物。先用高糖模拟DN处理HK-2细胞,再用含bi的药物血清和铁他汀-1 (ferstat -1)处理。CCK-8法检测细胞活力,Bodipy 581/591 C11荧光法检测脂质过氧化。使用商业试剂盒检测MDA和其他与嗜铁有关的指标。Western blotting分析蛋白水平。结果黄酮类化合物和萜类化合物是其主要鉴定成分。BI可降低DN小鼠的空腹血糖,纠正脂质代谢紊乱。它还能改善这些小鼠的肾损伤。同时,BI调节DN小鼠血清代谢物,重塑肠道菌群。此外,Nrf2、GPX4、FPN1、FTH1和SLC7A11被BI激活,而TFR1和ACSL4被抑制。结论bi通过调节血清代谢物、重塑肠道菌群、抑制铁下垂来改善DN。
{"title":"Balanophora involucrata alleviates diabetic nephropathy by inhibiting ferroptosis, modulating serum metabolites and gut microbiota","authors":"Chaoxi Tian ,&nbsp;Yawen Chen ,&nbsp;Jing Huang ,&nbsp;Fangyu Zhao ,&nbsp;Xiane Tang ,&nbsp;Jianhong Gao ,&nbsp;Xianbing Chen ,&nbsp;Xiuxue Yuan","doi":"10.1016/j.phymed.2026.157773","DOIUrl":"10.1016/j.phymed.2026.157773","url":null,"abstract":"<div><h3>Background</h3><div>Diabetic nephropathy (DN) serves as the major etiological factor of end-stage renal disease (ESRD) in DM patients. <em>Balanophora involucrata</em> Hook.f. &amp; Thomson (BI) is a traditional medicine of the Tujia ethnic group in China, with good therapeutic effects on kidney diseases. However, both its effectiveness in the context of DN and the regulatory mechanism remain to be further clarified.</div></div><div><h3>Purpose</h3><div>To elucidate the role of BI in DN mice, with a focus on exploring the intrinsic mechanism responsible for this process.</div></div><div><h3>Methods</h3><div>UPLC-MS/MS was employed to identify the main ingredients of BI. The DN model was established using C57BLKS/J db/db mice, which received BI treatment. Serum/urine parameters, renal histology, and ultrastructure were assessed. Intrinsic microbial composition and corresponding metabolites were analyzed via 16S rDNA gene sequencing and nontargeted metabolomics. HK-2 cells were treated with high glucose to simulate DN and then treated with BI-containing drug serum and ferrostatin-1 (fer-1). Cell viability was assessed by CCK-8 assay, and lipid peroxidation was evaluated using the Bodipy 581/591 C11 fluorescence assay. Commercial kits were used to detect MDA and other ferroptosis-related indicators. Protein levels were analyzed by Western blotting.</div></div><div><h3>Results</h3><div>Terpenoids and flavonoids were the main identified components of BI. Fasting blood glucose was reduced, and lipid metabolism disorders were corrected by BI in DN mice. It also ameliorated renal injury in these mice. Meanwhile, BI regulated serum metabolites, and remodeled gut microbiota in DN mice. Furthermore, Nrf2, GPX4, FPN1, FTH1, and SLC7A11 were activated by BI, whereas TFR1 and ACSL4 were suppressed.</div></div><div><h3>Conclusions</h3><div>BI ameliorates DN by regulating serum metabolites, remodeling the gut microbiota, and inhibiting ferroptosis.</div></div>","PeriodicalId":20212,"journal":{"name":"Phytomedicine","volume":"151 ","pages":"Article 157773"},"PeriodicalIF":8.3,"publicationDate":"2026-01-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145941422","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 inhibits melanoma progression by regulating PPARγ-TET1 complex-dependent LRSAM1 demethylation 大麻二酚通过调节PPARγ-TET1复合体依赖的LRSAM1去甲基化抑制黑色素瘤的进展
IF 8.3 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2026-01-04 DOI: 10.1016/j.phymed.2026.157775
Xuedan Zhang , Baoyu Shen , Enjiang Li , Fang Yun , Yu Feng , Zhenyan Wei , Junzi Niu , Yu Huang , Song Yu , Yingmin Kuang , Haoming Liu , Buqing Sai , Yuechun Zhu

Introduction

Melanoma represents one of the most aggressive forms of skin cancers, with advanced metastatic stages largely managed through chemotherapy. However, current therapeutic strategies remain limited by drug resistance and systemic toxicity. Cannabidiol (CBD), the primary nonpsychoactive constituent of Cannabis sativa, has recently attracted attention for its anticancer properties across multiple tumor types.

Objectives

This study aimed to explore the antitumor efficacy of CBD in melanoma and elucidate its underlying molecular mechanisms, with the goal of identifying novel therapeutic strategies to overcome resistance and reduce adverse effects associated with conventional treatments.

Methods

The antiproliferative and pro-apoptotic effects of CBD were assessed in vitro using MTS, EdU, Transwell invasion, and flow cytometry. In vivo efficacy was evaluated using a murine lung metastasis model. Potential CBD targets in melanoma were identified through network pharmacology and molecular docking, with a focus on peroxisome proliferator-activated receptor γ (PPARγ) and validation by western blotting and immunofluorescence. Integrated transcriptomic and genome-wide methylation analyses were performed to investigate epigenetic modifications induced by CBD. Co-immunoprecipitation and chromatin immunoprecipitation assays were employed to detect the interaction between PPARγ and ten-eleven translocation 1 (TET1), including their binding to promoter regions of downstream factors. Methylation-regulated target genes were further validated using qPCR and MeDIP.

Results

CBD significantly induced apoptosis and inhibited cell proliferation and invasion of melanoma cells in vitro, while reducing pulmonary metastasis in vivo. Pharmacological and molecular docking analyses, supported by protein-level validation, identified PPARγ as a critical mediator of CBD activity. Transcriptomic and methylation analyses revealed that CBD modulated global DNA methylation patterns, partly through the formation of a PPARγ-TET1 complex. This complex regulated the demethylation of leucine-rich repeat and sterile alpha motif-containing 1 (LRSAM1), a newly identified anticancer gene whose upregulation markedly enhanced melanoma cell apoptosis and suppressed proliferation.

Conclusions

CBD exhibited strong antitumor activity in melanoma by modulating the PPARγ–TET1 complex to induce demethylation of LRSAM1, thereby suppressing tumor progression. These findings identify CBD as a promising candidate for melanoma therapy.
黑色素瘤是最具侵袭性的皮肤癌之一,晚期转移主要通过化疗来控制。然而,目前的治疗策略仍然受到耐药性和全身毒性的限制。大麻二酚(Cannabidiol, CBD)是大麻的主要非精神活性成分,近年来因其对多种肿瘤的抗癌作用而受到关注。目的探讨CBD在黑色素瘤中的抗肿瘤作用,阐明其潜在的分子机制,以寻找新的治疗策略来克服耐药性,减少常规治疗相关的不良反应。方法采用MTS、EdU、Transwell侵袭及流式细胞术观察CBD的体外抗增殖和促凋亡作用。采用小鼠肺转移模型对其体内疗效进行了评价。通过网络药理学和分子对接,确定了CBD在黑色素瘤中的潜在靶点,重点关注过氧化物酶体增殖物激活受体γ (PPARγ),并通过免疫印迹和免疫荧光验证。通过综合转录组学和全基因组甲基化分析来研究CBD诱导的表观遗传修饰。采用免疫共沉淀法和染色质免疫共沉淀法检测PPARγ与10 - 11易位1 (TET1)之间的相互作用,包括它们与下游因子启动子区域的结合。利用qPCR和MeDIP进一步验证甲基化调控的靶基因。结果scbd在体外显著诱导黑色素瘤细胞凋亡,抑制细胞增殖和侵袭,减少肺转移。在蛋白水平验证的支持下,药理学和分子对接分析确定PPARγ是CBD活性的关键介质。转录组学和甲基化分析显示,CBD调节了全球DNA甲基化模式,部分是通过PPARγ-TET1复合物的形成。该复合物调节富含亮氨酸的重复和无菌α基序- 1 (LRSAM1)的去甲基化,LRSAM1是一种新发现的抗癌基因,其上调可显著增强黑色素瘤细胞凋亡并抑制增殖。结论scbd通过调节PPARγ-TET1复合物诱导LRSAM1去甲基化,从而抑制肿瘤进展,在黑色素瘤中表现出较强的抗肿瘤活性。这些发现确定CBD是黑色素瘤治疗的有希望的候选者。
{"title":"Cannabidiol inhibits melanoma progression by regulating PPARγ-TET1 complex-dependent LRSAM1 demethylation","authors":"Xuedan Zhang ,&nbsp;Baoyu Shen ,&nbsp;Enjiang Li ,&nbsp;Fang Yun ,&nbsp;Yu Feng ,&nbsp;Zhenyan Wei ,&nbsp;Junzi Niu ,&nbsp;Yu Huang ,&nbsp;Song Yu ,&nbsp;Yingmin Kuang ,&nbsp;Haoming Liu ,&nbsp;Buqing Sai ,&nbsp;Yuechun Zhu","doi":"10.1016/j.phymed.2026.157775","DOIUrl":"10.1016/j.phymed.2026.157775","url":null,"abstract":"<div><h3>Introduction</h3><div>Melanoma represents one of the most aggressive forms of skin cancers, with advanced metastatic stages largely managed through chemotherapy. However, current therapeutic strategies remain limited by drug resistance and systemic toxicity. Cannabidiol (CBD), the primary nonpsychoactive constituent of <em>Cannabis sativa</em>, has recently attracted attention for its anticancer properties across multiple tumor types.</div></div><div><h3>Objectives</h3><div>This study aimed to explore the antitumor efficacy of CBD in melanoma and elucidate its underlying molecular mechanisms, with the goal of identifying novel therapeutic strategies to overcome resistance and reduce adverse effects associated with conventional treatments.</div></div><div><h3>Methods</h3><div>The antiproliferative and pro-apoptotic effects of CBD were assessed <em>in vitro</em> using MTS, EdU, Transwell invasion, and flow cytometry. <em>In vivo</em> efficacy was evaluated using a murine lung metastasis model. Potential CBD targets in melanoma were identified through network pharmacology and molecular docking, with a focus on peroxisome proliferator-activated receptor γ (PPARγ) and validation by western blotting and immunofluorescence. Integrated transcriptomic and genome-wide methylation analyses were performed to investigate epigenetic modifications induced by CBD. Co-immunoprecipitation and chromatin immunoprecipitation assays were employed to detect the interaction between PPARγ and ten-eleven translocation 1 (TET1), including their binding to promoter regions of downstream factors. Methylation-regulated target genes were further validated using qPCR and MeDIP.</div></div><div><h3>Results</h3><div>CBD significantly induced apoptosis and inhibited cell proliferation and invasion of melanoma cells <em>in vitro</em>, while reducing pulmonary metastasis <em>in vivo</em>. Pharmacological and molecular docking analyses, supported by protein-level validation, identified PPARγ as a critical mediator of CBD activity. Transcriptomic and methylation analyses revealed that CBD modulated global DNA methylation patterns, partly through the formation of a PPARγ-TET1 complex. This complex regulated the demethylation of leucine-rich repeat and sterile alpha motif-containing 1 (LRSAM1), a newly identified anticancer gene whose upregulation markedly enhanced melanoma cell apoptosis and suppressed proliferation.</div></div><div><h3>Conclusions</h3><div>CBD exhibited strong antitumor activity in melanoma by modulating the PPARγ–TET1 complex to induce demethylation of LRSAM1, thereby suppressing tumor progression. These findings identify CBD as a promising candidate for melanoma therapy.</div></div>","PeriodicalId":20212,"journal":{"name":"Phytomedicine","volume":"151 ","pages":"Article 157775"},"PeriodicalIF":8.3,"publicationDate":"2026-01-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145979725","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
Gastrodin ameliorates nonalcoholic fatty liver disease via inhibiting hepatic ferroptosis by the Keap1/Nrf2 signaling pathway 天麻素通过Keap1/Nrf2信号通路抑制肝铁下垂,改善非酒精性脂肪肝疾病
IF 8.3 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2026-01-03 DOI: 10.1016/j.phymed.2025.157762
Qingxiu Zhang , Xingyi Chen , Meijuan Liao , Xianqiong Lu , Qiyuan Luo , Shiliang Li , Tao Ran , Yao Fan , Di Pan , Xiong Pan , Yan He , Xueke Zhao , Jiyu Chen

Background

Nonalcoholic fatty liver disease (NAFLD) is a global health challenge lacking effective treatments. Gastrodin (GAS), a major phenolglucoside from Gastrodia elata Blume (Orchidaceae), exhibits multiple beneficial properties but its potential role in NAFLD through ferroptosis remains unclear.

Objective

To investigate the therapeutic effects of GAS on NAFLD and clarify the underlying molecular mechanisms.

Methods

We established NAFLD models using high-fat diet (HFD)-fed mice and HepG2 cells treated with palmitic acid (PA) and oleic acid (OA). Therapeutic effects were assessed via histopathological and biochemical analyses. Molecular mechanisms and drug targets were investigated using co-immunoprecipitation (Co-IP), drug affinity responsive target stability (DARTS), cellular thermal shift assay (CETSA), and dual-luciferase reporter assays.

Results

GAS treatment significantly reduced hepatic inflammation and lipid accumulation, leading to improved metabolic parameters. Additionally, GAS decreased iron overload and oxidative stress markers, restored antioxidant defenses, and preserved mitochondrial function. Mechanistically, interaction assays revealed that GAS directly binds to Keap1, disrupting the Keap1/Nrf2 complex, promoting Nrf2 nuclear translocation, and enhancing the expression of target genes FSP1 and GPX4.

Conclusion

GAS demonstrates significant promise as a therapeutic agent for NAFLD by inhibiting hepatic ferroptosis via direct targeting of the Keap1/Nrf2 signaling pathway.
背景:非酒精性脂肪性肝病(NAFLD)是缺乏有效治疗的全球性健康挑战。天麻素(GAS)是一种来自天麻科(兰科)的主要酚类糖苷,具有多种有益特性,但其在NAFLD中通过铁下垂的潜在作用尚不清楚。目的探讨GAS对NAFLD的治疗作用,并阐明其分子机制。方法采用高脂饲料(HFD)喂养小鼠和棕榈酸(PA)和油酸(OA)处理的HepG2细胞建立NAFLD模型。通过组织病理学和生化分析评估治疗效果。采用共免疫沉淀(Co-IP)、药物亲和反应靶标稳定性(DARTS)、细胞热移测定(CETSA)和双荧光素酶报告基因测定来研究分子机制和药物靶点。结果gas治疗可显著降低肝脏炎症和脂质积累,改善代谢参数。此外,GAS减少铁超载和氧化应激标志物,恢复抗氧化防御,并保持线粒体功能。机制上,相互作用分析显示GAS直接结合Keap1,破坏Keap1/Nrf2复合物,促进Nrf2核易位,增强靶基因FSP1和GPX4的表达。结论ongas通过直接靶向Keap1/Nrf2信号通路抑制肝铁下垂,有望成为NAFLD的治疗药物。
{"title":"Gastrodin ameliorates nonalcoholic fatty liver disease via inhibiting hepatic ferroptosis by the Keap1/Nrf2 signaling pathway","authors":"Qingxiu Zhang ,&nbsp;Xingyi Chen ,&nbsp;Meijuan Liao ,&nbsp;Xianqiong Lu ,&nbsp;Qiyuan Luo ,&nbsp;Shiliang Li ,&nbsp;Tao Ran ,&nbsp;Yao Fan ,&nbsp;Di Pan ,&nbsp;Xiong Pan ,&nbsp;Yan He ,&nbsp;Xueke Zhao ,&nbsp;Jiyu Chen","doi":"10.1016/j.phymed.2025.157762","DOIUrl":"10.1016/j.phymed.2025.157762","url":null,"abstract":"<div><h3>Background</h3><div>Nonalcoholic fatty liver disease (NAFLD) is a global health challenge lacking effective treatments. Gastrodin (GAS), a major phenolglucoside from <em>Gastrodia elata</em> Blume (Orchidaceae), exhibits multiple beneficial properties but its potential role in NAFLD through ferroptosis remains unclear.</div></div><div><h3>Objective</h3><div>To investigate the therapeutic effects of GAS on NAFLD and clarify the underlying molecular mechanisms.</div></div><div><h3>Methods</h3><div>We established NAFLD models using high-fat diet (HFD)-fed mice and HepG2 cells treated with palmitic acid (PA) and oleic acid (OA). Therapeutic effects were assessed via histopathological and biochemical analyses. Molecular mechanisms and drug targets were investigated using co-immunoprecipitation (Co-IP), drug affinity responsive target stability (DARTS), cellular thermal shift assay (CETSA), and dual-luciferase reporter assays.</div></div><div><h3>Results</h3><div>GAS treatment significantly reduced hepatic inflammation and lipid accumulation, leading to improved metabolic parameters. Additionally, GAS decreased iron overload and oxidative stress markers, restored antioxidant defenses, and preserved mitochondrial function. Mechanistically, interaction assays revealed that GAS directly binds to Keap1, disrupting the Keap1/Nrf2 complex, promoting Nrf2 nuclear translocation, and enhancing the expression of target genes <em>FSP1</em> and <em>GPX4</em>.</div></div><div><h3>Conclusion</h3><div>GAS demonstrates significant promise as a therapeutic agent for NAFLD by inhibiting hepatic ferroptosis via direct targeting of the Keap1/Nrf2 signaling pathway.</div></div>","PeriodicalId":20212,"journal":{"name":"Phytomedicine","volume":"151 ","pages":"Article 157762"},"PeriodicalIF":8.3,"publicationDate":"2026-01-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145941421","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
Columbianadin improves M1 polarization of microglia after spinal cord injury by stabilizing PTEN and inhibiting the PI3K/AKT pathway Columbianadin通过稳定PTEN和抑制PI3K/AKT通路改善脊髓损伤后小胶质细胞M1极化
IF 8.3 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2026-01-02 DOI: 10.1016/j.phymed.2026.157767
Shining Xiao , Wansong Wang , Pei Shi , Kaiying He , Chao Peng , Yu Zhang , Shue Hong , Youqing Yang , Jun Feng , Haidi Bi , Lang Shuai , Jun Wang

Background

Inhibiting the polarization of microglia toward the M1 pro-inflammatory phenotype to alleviate neuroinflammation and improve neuronal survival is a critical strategy for promoting spinal cord injury (SCI) repair. PTEN, a core regulator of cellular inflammatory responses, exerts its function by negatively modulating the PI3K/AKT signaling pathway. Columbianadin (CBN), a coumarin derivative isolated from the roots of Angelica sinensis (Oliv.) Diels, possesses multiple biological activities, including potent anti-inflammatory effects.

Objective

This study aimed to evaluate the regulatory effect of CBN on microglial polarization post-SCI and elucidate its underlying molecular mechanisms.

Methods

A combination of experimental techniques, including Western blotting, quantitative real-time PCR (qPCR), immunofluorescence, ubiquitination assays, co-immunoprecipitation (Co-IP), live/dead staining, molecular docking, cellular thermal shift assay (CETSA), bioluminescence imaging (BLI), differential scanning fluorimetry (DSF), and network pharmacology analysis, was employed in this study. Additionally, a rat SCI model was established to evaluate the impact of CBN on motor function recovery.

Results

CBN inhibited microglial M1 polarization, mitigated neuroinflammation, and improved neuronal survival, thereby facilitating SCI repair. Mechanistically, CBN directly targeted PTEN, by suppressing the ubiquitin-mediated degradation of PTEN while spatially promoting the interaction between PTEN and the PI3K regulatory subunit p85α. These dual effects synergistically enhanced PTEN protein stability and biological activity, jointly inhibiting PI3K/AKT signaling pathway activation.

Conclusion

Our study identifies CBN as a potential candidate drug for SCI treatment, as it enhances PTEN stability to inhibit the PI3K/AKT pathway, thereby regulating microglial polarization and protecting neurons from neuroinflammatory damage.
背景抑制小胶质细胞向M1促炎表型的极化,以减轻神经炎症,提高神经元存活是促进脊髓损伤(SCI)修复的关键策略。PTEN是细胞炎症反应的核心调节因子,通过负向调节PI3K/AKT信号通路发挥作用。从当归(Angelica sinensis, Oliv.)根中提取的香豆素衍生物Columbianadin (CBN)。Diels具有多种生物活性,包括有效的抗炎作用。目的探讨CBN对脊髓损伤后小胶质细胞极化的调控作用,并阐明其分子机制。方法采用Western blotting、定量实时PCR (qPCR)、免疫荧光、泛素化、共免疫沉淀(Co-IP)、活/死染色、分子对接、细胞热移测定(CETSA)、生物发光成像(BLI)、差示扫描荧光法(DSF)和网络药理学分析等实验技术。此外,建立大鼠脊髓损伤模型,评估CBN对运动功能恢复的影响。结果scbn抑制小胶质细胞M1极化,减轻神经炎症,提高神经元存活,从而促进脊髓损伤修复。在机制上,CBN直接靶向PTEN,通过抑制泛素介导的PTEN降解,同时在空间上促进PTEN与PI3K调控亚基p85α之间的相互作用。这些双重作用协同增强PTEN蛋白的稳定性和生物活性,共同抑制PI3K/AKT信号通路的激活。结论CBN可增强PTEN的稳定性,抑制PI3K/AKT通路,从而调节小胶质细胞极化,保护神经元免受神经炎症损伤,是脊髓损伤治疗的潜在候选药物。
{"title":"Columbianadin improves M1 polarization of microglia after spinal cord injury by stabilizing PTEN and inhibiting the PI3K/AKT pathway","authors":"Shining Xiao ,&nbsp;Wansong Wang ,&nbsp;Pei Shi ,&nbsp;Kaiying He ,&nbsp;Chao Peng ,&nbsp;Yu Zhang ,&nbsp;Shue Hong ,&nbsp;Youqing Yang ,&nbsp;Jun Feng ,&nbsp;Haidi Bi ,&nbsp;Lang Shuai ,&nbsp;Jun Wang","doi":"10.1016/j.phymed.2026.157767","DOIUrl":"10.1016/j.phymed.2026.157767","url":null,"abstract":"<div><h3>Background</h3><div>Inhibiting the polarization of microglia toward the M1 pro-inflammatory phenotype to alleviate neuroinflammation and improve neuronal survival is a critical strategy for promoting spinal cord injury (SCI) repair. PTEN, a core regulator of cellular inflammatory responses, exerts its function by negatively modulating the PI3K/AKT signaling pathway. Columbianadin (CBN), a coumarin derivative isolated from the roots of <em>Angelica sinensis</em> (Oliv.) Diels, possesses multiple biological activities, including potent anti-inflammatory effects.</div></div><div><h3>Objective</h3><div>This study aimed to evaluate the regulatory effect of CBN on microglial polarization post-SCI and elucidate its underlying molecular mechanisms.</div></div><div><h3>Methods</h3><div>A combination of experimental techniques, including Western blotting, quantitative real-time PCR (qPCR), immunofluorescence, ubiquitination assays, co-immunoprecipitation (Co-IP), live/dead staining, molecular docking, cellular thermal shift assay (CETSA), bioluminescence imaging (BLI), differential scanning fluorimetry (DSF), and network pharmacology analysis, was employed in this study. Additionally, a rat SCI model was established to evaluate the impact of CBN on motor function recovery.</div></div><div><h3>Results</h3><div>CBN inhibited microglial M1 polarization, mitigated neuroinflammation, and improved neuronal survival, thereby facilitating SCI repair. Mechanistically, CBN directly targeted PTEN, by suppressing the ubiquitin-mediated degradation of PTEN while spatially promoting the interaction between PTEN and the PI3K regulatory subunit p85α. These dual effects synergistically enhanced PTEN protein stability and biological activity, jointly inhibiting PI3K/AKT signaling pathway activation.</div></div><div><h3>Conclusion</h3><div>Our study identifies CBN as a potential candidate drug for SCI treatment, as it enhances PTEN stability to inhibit the PI3K/AKT pathway, thereby regulating microglial polarization and protecting neurons from neuroinflammatory damage.</div></div>","PeriodicalId":20212,"journal":{"name":"Phytomedicine","volume":"151 ","pages":"Article 157767"},"PeriodicalIF":8.3,"publicationDate":"2026-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145980906","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
Efficacy and mechanisms of Xiangsha Liujunzi Decoction for gastroesophageal reflux disease: A study integrating meta-analysis, network pharmacology and molecular docking 香沙六君子汤治疗胃食管反流病的疗效及机制:荟萃分析、网络药理学和分子对接的研究
IF 8.3 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2026-01-02 DOI: 10.1016/j.phymed.2026.157766
Mengfei Tian , Ke Wang , Diping Chen

Background

Reflux esophagitis (RE) significantly impairs quality of life. Xiangsha Liujunzi Decoction (XSLJZD) is used in traditional Chinese medicine for RE, but its efficacy and mechanisms remain to be systematically evaluated.

Purpose

This study integrated meta-analysis, network pharmacology, and molecular docking to evaluate the clinical efficacy of XSLJZD for RE and elucidate its mechanisms of action.

Methods

Randomized controlled trials (RCTs) on XSLJZD for RE were systematically searched. Methodological quality was assessed using the Cochrane Risk of Bias tool, and data were synthesized with RevMan 5.3. Network pharmacology utilizing TCMSP identified active components, targets, and pathways. Molecular docking validated core compound-target interactions.

Results

Eight RCTs (n = 646) showed that XSLJZD significantly improved clinical outcomes compared with controls, demonstrating superior overall efficacy and reduced recurrence. Network analysis identified luteolin, baicalin, and β-sitosterol as core components, with AKT1, IL6, and CASP3 as key targets, potentially regulating apoptosis, IL-17, and TNF signaling pathways. Molecular docking confirmed stable binding.

Conclusion

XSLJZD may improve RE symptoms as an adjunctive therapy, but the evidence is low in certainty. The proposed anti-inflammatory and apoptotic mechanisms are computationally derived. High-quality trials and experimental validation are needed to confirm efficacy and mechanisms of action.
背景:反流性食管炎(RE)显著影响生活质量。香沙六君子汤(XSLJZD)是治疗RE的常用中药,但其疗效和作用机制尚待系统评价。目的综合meta分析、网络药理学、分子对接等方法,评价XSLJZD治疗RE的临床疗效,阐明其作用机制。方法系统检索使用XSLJZD治疗RE的随机对照试验(RCTs)。方法学质量采用Cochrane偏倚风险评估工具进行评估,数据采用RevMan 5.3进行综合。网络药理学利用TCMSP识别活性成分,目标和途径。分子对接验证了核心化合物-靶标相互作用。结果8项随机对照试验(n = 646)显示,与对照组相比,XSLJZD显著改善了临床结果,总体疗效优越,复发率降低。网络分析发现木草素、黄芩苷和β-谷甾醇是核心成分,AKT1、IL6和CASP3是关键靶点,可能调控细胞凋亡、IL-17和TNF信号通路。分子对接证实结合稳定。结论xsljzd作为辅助治疗可改善RE症状,但证据的确定性较低。所提出的抗炎和细胞凋亡机制是计算得出的。需要高质量的试验和实验验证来确认其疗效和作用机制。
{"title":"Efficacy and mechanisms of Xiangsha Liujunzi Decoction for gastroesophageal reflux disease: A study integrating meta-analysis, network pharmacology and molecular docking","authors":"Mengfei Tian ,&nbsp;Ke Wang ,&nbsp;Diping Chen","doi":"10.1016/j.phymed.2026.157766","DOIUrl":"10.1016/j.phymed.2026.157766","url":null,"abstract":"<div><h3>Background</h3><div>Reflux esophagitis (RE) significantly impairs quality of life. Xiangsha Liujunzi Decoction (XSLJZD) is used in traditional Chinese medicine for RE, but its efficacy and mechanisms remain to be systematically evaluated.</div></div><div><h3>Purpose</h3><div>This study integrated meta-analysis, network pharmacology, and molecular docking to evaluate the clinical efficacy of XSLJZD for RE and elucidate its mechanisms of action.</div></div><div><h3>Methods</h3><div>Randomized controlled trials (RCTs) on XSLJZD for RE were systematically searched. Methodological quality was assessed using the Cochrane Risk of Bias tool, and data were synthesized with RevMan 5.3. Network pharmacology utilizing TCMSP identified active components, targets, and pathways. Molecular docking validated core compound-target interactions.</div></div><div><h3>Results</h3><div>Eight RCTs (<em>n</em> = 646) showed that XSLJZD significantly improved clinical outcomes compared with controls, demonstrating superior overall efficacy and reduced recurrence. Network analysis identified luteolin, baicalin, and β-sitosterol as core components, with AKT1, IL6, and CASP3 as key targets, potentially regulating apoptosis, IL-17, and TNF signaling pathways. Molecular docking confirmed stable binding.</div></div><div><h3>Conclusion</h3><div>XSLJZD may improve RE symptoms as an adjunctive therapy, but the evidence is low in certainty. The proposed anti-inflammatory and apoptotic mechanisms are computationally derived. High-quality trials and experimental validation are needed to confirm efficacy and mechanisms of action.</div></div>","PeriodicalId":20212,"journal":{"name":"Phytomedicine","volume":"151 ","pages":"Article 157766"},"PeriodicalIF":8.3,"publicationDate":"2026-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145908769","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
Jujuboside A alleviates visceral pain and depression comorbidity and modulates the P2X7R-BDNF signaling axis 红枣苷A减轻内脏疼痛和抑郁合并症并调节P2X7R-BDNF信号轴
IF 8.3 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2026-01-02 DOI: 10.1016/j.phymed.2026.157764
Jian Liu , Yeqing Liu , Changtie Liu , Jing Wang , Yuanyi Zhang , Yuxing Yang , Si Peng , Rongrong Xu , Lingxiang Xu , Xingchen Wang , Wei Xiong , Yun Gao
Visceral pain is frequently accompanied by depression, a comorbidity involving central neuroinflammation and abnormal neuronal plasticity. The P2X7 receptor (P2X7R) plays a crucial role in neuroinflammation and pyroptosis, while Jujuboside A (JuA), a major saponin extracted from Ziziphus jujuba seeds, has been reported to exert significant antidepressant and analgesic effects. In this study, we systematically evaluated the regulatory effects of JuA on the P2X7R-brain-derived neurotrophic factor (BDNF) pathway and on pyroptosis and apoptosis using a rat model of colorectal distension (CRD) and primary neuron/astrocyte cultures. JuA markedly alleviated visceral hypersensitivity and depressive-like behaviors in CRD rats and reduced P2X7R expression in both the spinal cord (SC) and hippocampus (HPC). Further investigations in vitro revealed that JuA inhibited excessive P2X7R activation in SC astrocytes, thereby decreasing the expression of NLRP3, Caspase-1, GSDMD, IL-1β and TNF-α, indicating suppression of pyroptosis. Similarly, JuA exerted an anti-pyroptotic effect in HPC astrocytes and inhibited neuronal apoptosis by reducing Caspase-3 and Bax levels while increasing Bcl2 expression, leading to upregulation of HPC BDNF. Collectively, JuA targets P2X7R and suppresses downstream pyroptotic and apoptotic signaling in vitro, which may contribute to its neuroprotective effects. These findings provide experimental evidence supporting the potential of JuA as a therapeutic agent for comorbid visceral pain and depression.
内脏疼痛经常伴有抑郁,这是一种涉及中枢神经炎症和异常神经元可塑性的合并症。P2X7受体(P2X7R)在神经炎症和焦亡中起着至关重要的作用,而枣苷a (JuA)是一种从酸枣种子中提取的主要皂苷,据报道具有显著的抗抑郁和镇痛作用。在这项研究中,我们系统地评估了JuA对p2x7r脑源性神经营养因子(BDNF)通路以及对大鼠结肠膨胀(CRD)模型和原代神经元/星形胶质细胞培养的焦亡和凋亡的调节作用。JuA可显著减轻CRD大鼠内脏超敏反应和抑郁样行为,降低脊髓(SC)和海马(HPC)中P2X7R的表达。进一步的体外研究表明,JuA抑制SC星形细胞中P2X7R的过度激活,从而降低NLRP3、Caspase-1、GSDMD、IL-1β和TNF-α的表达,表明其抑制了焦亡。同样,JuA在HPC星形细胞中发挥抗焦亡作用,通过降低Caspase-3和Bax水平,增加Bcl2表达,抑制神经元凋亡,导致HPC BDNF上调。总的来说,JuA在体外靶向P2X7R并抑制下游焦亡和凋亡信号,这可能有助于其神经保护作用。这些发现提供了实验证据,支持JuA作为合并症内脏疼痛和抑郁症治疗剂的潜力。
{"title":"Jujuboside A alleviates visceral pain and depression comorbidity and modulates the P2X7R-BDNF signaling axis","authors":"Jian Liu ,&nbsp;Yeqing Liu ,&nbsp;Changtie Liu ,&nbsp;Jing Wang ,&nbsp;Yuanyi Zhang ,&nbsp;Yuxing Yang ,&nbsp;Si Peng ,&nbsp;Rongrong Xu ,&nbsp;Lingxiang Xu ,&nbsp;Xingchen Wang ,&nbsp;Wei Xiong ,&nbsp;Yun Gao","doi":"10.1016/j.phymed.2026.157764","DOIUrl":"10.1016/j.phymed.2026.157764","url":null,"abstract":"<div><div>Visceral pain is frequently accompanied by depression, a comorbidity involving central neuroinflammation and abnormal neuronal plasticity. The P2X7 receptor (P2X7R) plays a crucial role in neuroinflammation and pyroptosis, while Jujuboside A (JuA), a major saponin extracted from Ziziphus jujuba seeds, has been reported to exert significant antidepressant and analgesic effects. In this study, we systematically evaluated the regulatory effects of JuA on the P2X7R-brain-derived neurotrophic factor (BDNF) pathway and on pyroptosis and apoptosis using a rat model of colorectal distension (CRD) and primary neuron/astrocyte cultures. JuA markedly alleviated visceral hypersensitivity and depressive-like behaviors in CRD rats and reduced P2X7R expression in both the spinal cord (SC) and hippocampus (HPC). Further investigations in vitro revealed that JuA inhibited excessive P2X7R activation in SC astrocytes, thereby decreasing the expression of NLRP3, Caspase-1, GSDMD, IL-1β and TNF-α, indicating suppression of pyroptosis. Similarly, JuA exerted an anti-pyroptotic effect in HPC astrocytes and inhibited neuronal apoptosis by reducing Caspase-3 and Bax levels while increasing Bcl2 expression, leading to upregulation of HPC BDNF. Collectively, JuA targets P2X7R and suppresses downstream pyroptotic and apoptotic signaling in vitro, which may contribute to its neuroprotective effects. These findings provide experimental evidence supporting the potential of JuA as a therapeutic agent for comorbid visceral pain and depression.</div></div>","PeriodicalId":20212,"journal":{"name":"Phytomedicine","volume":"151 ","pages":"Article 157764"},"PeriodicalIF":8.3,"publicationDate":"2026-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145980197","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
Paeoniflorin promotes therapeutic senescence in melanoma through endoplasmic reticulum stress and the calpain1/ERK5/p27 axis 芍药苷通过内质网应激和calpain1/ERK5/p27轴促进黑色素瘤的治疗性衰老。
IF 8.3 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2026-01-01 DOI: 10.1016/j.phymed.2025.157730
Shu-Chun Liu , Hung-Chuan Pan , Pei-Yi Chu , Ming-Chu Tsai , Liang-Ju Chen , Yu-Chin Lin , Ching-Wen Chang , Yi-Ching Tsai , Chia-Yun Tsai , Meei-Ling Sheu , De-Wei Lai
Background: Skin cutaneous melanoma (SKCM) is a highly aggressive malignancy with increasing global incidence and limited therapeutic options in advanced stages. Cellular senescence, characterized by irreversible growth arrest and tumor suppression, has emerged as a promising therapeutic approach.
Purpose: This study aimed to investigate the anti-tumor and senescence-inducing effects of paeoniflorin on SKCM and to elucidate its underlying molecular mechanisms.
Study design: Paeoniflorin was evaluated through in vitro, in vivo, and bioinformatic analyses to determine its impact on melanoma progression and to clarify the involvement of the endoplasmic reticulum (ER) stress/calpain1/ERK5 signaling pathway.
Methods: Publicly available SKCM datasets were analyzed to evaluate the correlation between senescence-associated genes (RRAS, CDKN1B, LAMA1, and CDC25A) and patient prognosis. The effects of paeoniflorin on melanoma cells were examined by immunofluorescence, β-galactosidase staining, western blotting, electron microscopy, and calpain activity assays. Chemical inhibitors and siRNA were applied to verify the functional role of calpain1. Positron emission tomography/computed tomography (PET/CT) imaging, histopathological analysis, and metastasis assays were performed to assess the in vivo efficacy of paeoniflorin.
Results: Bioinformatic analysis revealed that high expression of RRAS and CDKN1B was associated with favorable prognosis, whereas elevated LAMA1 and CDC25A indicated poor survival outcomes in SKCM. Paeoniflorin treatment induced DNA damage, G2/M phase arrest, and senescence in melanoma cells, accompanied by enhanced ER stress, increased calpain1 activity, and downregulated ERK5 expression. Inhibition or knockdown of calpain1 reversed these effects. In mouse models, paeoniflorin markedly reduced tumor growth and metastasis, effects that were abolished by calpain1 silencing. Mechanistically, paeoniflorin triggered calpain1-dependent ERK5 degradation and p27 upregulation, leading to cellular senescence and tumor suppression.
Conclusions: Paeoniflorin induces melanoma cell senescence through the ER stress/calpain1/ERK5/p27 signaling axis, suggesting its potential as a novel therapeutic strategy against SKCM.
背景:皮肤黑色素瘤(SKCM)是一种高度侵袭性的恶性肿瘤,全球发病率不断上升,晚期治疗选择有限。细胞衰老以不可逆的生长停滞和肿瘤抑制为特征,已成为一种很有前途的治疗方法。目的:研究芍药苷对SKCM的抗肿瘤和诱导衰老作用,并探讨其潜在的分子机制。研究设计:通过体外、体内和生物信息学分析评估芍药苷,以确定其对黑色素瘤进展的影响,并阐明内质网(ER)应激/calpain1/ERK5信号通路的参与。方法:分析公开可用的SKCM数据集,评估衰老相关基因(RRAS、CDKN1B、LAMA1和CDC25A)与患者预后的相关性。采用免疫荧光法、β-半乳糖苷酶染色法、免疫印迹法、电镜法和钙蛋白酶活性法检测芍药苷对黑色素瘤细胞的影响。利用化学抑制剂和siRNA验证calpain1的功能作用。通过正电子发射断层扫描/计算机断层扫描(PET/CT)成像、组织病理学分析和转移试验来评估芍药苷的体内疗效。结果:生物信息学分析显示,RRAS和CDKN1B的高表达与良好的预后相关,而LAMA1和CDC25A的升高表明SKCM的生存结果较差。芍药苷处理导致黑色素瘤细胞DNA损伤、G2/M期阻滞和衰老,并伴有内质网络应激增强、calpain1活性升高和ERK5表达下调。抑制或敲低calpain1可逆转这些作用。在小鼠模型中,芍药苷显著降低肿瘤生长和转移,calpain1沉默可以消除这种作用。在机制上,芍药苷触发calpain1依赖性ERK5降解和p27上调,导致细胞衰老和肿瘤抑制。结论:芍药苷通过内质网应激/calpain1/ERK5/p27信号轴诱导黑色素瘤细胞衰老,提示其可能作为一种新的治疗SKCM的策略。
{"title":"Paeoniflorin promotes therapeutic senescence in melanoma through endoplasmic reticulum stress and the calpain1/ERK5/p27 axis","authors":"Shu-Chun Liu ,&nbsp;Hung-Chuan Pan ,&nbsp;Pei-Yi Chu ,&nbsp;Ming-Chu Tsai ,&nbsp;Liang-Ju Chen ,&nbsp;Yu-Chin Lin ,&nbsp;Ching-Wen Chang ,&nbsp;Yi-Ching Tsai ,&nbsp;Chia-Yun Tsai ,&nbsp;Meei-Ling Sheu ,&nbsp;De-Wei Lai","doi":"10.1016/j.phymed.2025.157730","DOIUrl":"10.1016/j.phymed.2025.157730","url":null,"abstract":"<div><div>Background: Skin cutaneous melanoma (SKCM) is a highly aggressive malignancy with increasing global incidence and limited therapeutic options in advanced stages. Cellular senescence, characterized by irreversible growth arrest and tumor suppression, has emerged as a promising therapeutic approach.</div><div>Purpose: This study aimed to investigate the anti-tumor and senescence-inducing effects of paeoniflorin on SKCM and to elucidate its underlying molecular mechanisms.</div><div>Study design: Paeoniflorin was evaluated through <em>in vitro, in vivo</em>, and bioinformatic analyses to determine its impact on melanoma progression and to clarify the involvement of the endoplasmic reticulum (ER) stress/calpain1/ERK5 signaling pathway.</div><div>Methods: Publicly available SKCM datasets were analyzed to evaluate the correlation between senescence-associated genes (RRAS, CDKN1B, LAMA1, and CDC25A) and patient prognosis. The effects of paeoniflorin on melanoma cells were examined by immunofluorescence, β-galactosidase staining, western blotting, electron microscopy, and calpain activity assays. Chemical inhibitors and siRNA were applied to verify the functional role of calpain1. Positron emission tomography/computed tomography (PET/CT) imaging, histopathological analysis, and metastasis assays were performed to assess the <em>in vivo</em> efficacy of paeoniflorin.</div><div>Results: Bioinformatic analysis revealed that high expression of RRAS and CDKN1B was associated with favorable prognosis, whereas elevated LAMA1 and CDC25A indicated poor survival outcomes in SKCM. Paeoniflorin treatment induced DNA damage, G2/M phase arrest, and senescence in melanoma cells, accompanied by enhanced ER stress, increased calpain1 activity, and downregulated ERK5 expression. Inhibition or knockdown of calpain1 reversed these effects. In mouse models, paeoniflorin markedly reduced tumor growth and metastasis, effects that were abolished by calpain1 silencing. Mechanistically, paeoniflorin triggered calpain1-dependent ERK5 degradation and p27 upregulation, leading to cellular senescence and tumor suppression.</div><div>Conclusions: Paeoniflorin induces melanoma cell senescence through the ER stress/calpain1/ERK5/p27 signaling axis, suggesting its potential as a novel therapeutic strategy against SKCM.</div></div>","PeriodicalId":20212,"journal":{"name":"Phytomedicine","volume":"150 ","pages":"Article 157730"},"PeriodicalIF":8.3,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145850668","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
Tongmai Huazheng mixture attenuates adenomyosis by inducing ferroptosis through suppression of the JAK2/STAT3 signaling pathway 通脉化正合剂通过抑制JAK2/STAT3信号通路诱导铁凋亡,从而减弱子宫腺肌症
IF 8.3 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2026-01-01 DOI: 10.1016/j.phymed.2025.157748
Ya-xin Shi , Cheng-yuan Zhang , Yu-ning Xie , Zhi-yong Liu , Xin Wang , Ling-di Wang , Zi-lu Wang , Hong-bo Yuan , Rui-hua Zhao , Wei Shi
<div><h3>Background</h3><div>Ferroptosis is a newly characterized iron-dependent form of regulated cell death. In adenomyosis (AM), ectopic endometrial lesions exhibit resistance to ferroptosis, which contributes to disease progression and recurrence. Tongmai Huazheng mixture (TMHZ) is a hospital-formulated traditional Chinese medicine (TCM) compound that has demonstrated clinical efficacy in the management of AM. However, the precise mechanisms by which TMHZ regulates ferroptosis in AM remain unclear.</div></div><div><h3>Purpose</h3><div>The aim of this study was to determine whether TMHZ exerts its anti-AM effects by inducing ferroptosis through inhibition of the JAK2/STAT3 signaling pathway.</div></div><div><h3>Methods</h3><div>A multiomics strategy integrating UPLC-MS/MS, network pharmacology, and RNA sequencing was employed to identify candidate targets and key regulatory pathways of TMHZ. <em>In vitro</em>, the effects of TMHZ-containing serum on proliferation, migration, and invasion of adenomyosis-derived cells (AMDCs) were evaluated using CCK-8, EdU, wound healing, and Transwell assays. Ferroptosis induction and oxidative stress levels were assessed using FerroOrange and CM-H<sub>2</sub>DCFDA fluorescent probes, glutathione (GSH), superoxide dismutase (SOD), and malondialdehyde (MDA) assay kits, and transmission electron microscopy. Immunofluorescence and western blotting (WB) were performed to investigate the regulatory effects of TMHZ on the JAK2/STAT3 pathway and ferroptosis-related proteins. The JAK2 inhibitor AG490 and agonist coumermycin A1 were used to validate the involvement of the pathway. <em>In vivo</em>, a tamoxifen-induced AM mouse model was established to assess the therapeutic efficacy and safety of TMHZ. Histopathological changes in the uterus were examined by hematoxylin–eosin (HE) staining, and serum levels of estradiol (E2) and progesterone (P4) were measured to verify model establishment. SOD and MDA levels and the results of immunohistochemical and WB analyses were used to further confirm the <em>in vivo</em> regulation of ferroptosis and JAK2/STAT3 signaling by TMHZ. Additionally, HE staining of major organs (liver, spleen, and kidney) was performed to evaluate systemic safety.</div></div><div><h3>Results</h3><div>Bioinformatics analysis suggested that the therapeutic effects of TMHZ were closely associated with the regulation of the JAK2/STAT3 pathway and ferroptosis. <em>In vitro</em>, TMHZ significantly inhibited the proliferation, migration, and invasion of AMDCs; increased intracellular Fe²⁺ and reactive oxygen species (ROS) levels; reduced GSH and SOD activity; elevated the MDA content, and induced typical mitochondrial damage. TMHZ also downregulated the expressions of p-JAK2, p-STAT3, SLC7A11, and GPX4. Further rescue experiments using JAK2 inhibitors and agonists confirmed the key role of the JAK2/STAT3 pathway in this process. <em>In vivo</em>, TMHZ suppressed JAK2/STAT3 signaling to promote ferroptosis,
背景:铁下垂是一种新发现的铁依赖性细胞死亡形式。在子宫腺肌症(AM)中,异位子宫内膜病变表现出对铁下垂的抵抗,这有助于疾病的进展和复发。通脉化正合剂(TMHZ)是一种医院配制的治疗AM的有效中药复方。然而,TMHZ调节AM中铁下垂的确切机制尚不清楚。目的本研究的目的是确定TMHZ是否通过抑制JAK2/STAT3信号通路诱导铁凋亡来发挥其抗am作用。方法采用UPLC-MS/MS、网络药理学和RNA测序相结合的多组学策略,鉴定TMHZ的候选靶点和关键调控通路。在体外,采用CCK-8、EdU、伤口愈合和Transwell试验评估含tmhz血清对腺肌病源性细胞(amdc)增殖、迁移和侵袭的影响。采用FerroOrange和CM-H2DCFDA荧光探针、谷胱甘肽(GSH)、超氧化物歧化酶(SOD)和丙二醛(MDA)检测试剂盒和透射电镜评估铁死亡诱导和氧化应激水平。免疫荧光和western blotting (WB)检测TMHZ对JAK2/STAT3通路和凋亡相关蛋白的调控作用。使用JAK2抑制剂AG490和激动剂coumermycin A1来验证该途径的参与。在体内,我们建立了他莫昔芬诱导的AM小鼠模型,以评估TMHZ的治疗效果和安全性。采用苏木精-伊红(HE)染色观察子宫组织病理变化,测定血清雌二醇(E2)、孕酮(P4)水平,验证模型建立。利用SOD和MDA水平以及免疫组织化学和WB分析结果进一步证实了TMHZ对铁亡和JAK2/STAT3信号的体内调节作用。此外,对主要器官(肝、脾和肾)进行HE染色以评估全身安全性。结果生物信息学分析表明,TMHZ的治疗效果与调控JAK2/STAT3通路和铁下垂密切相关。在体外,TMHZ显著抑制amdc的增殖、迁移和侵袭;增加细胞内Fe 2 +和活性氧(ROS)水平;GSH和SOD活性降低;MDA含量升高,引起典型的线粒体损伤。TMHZ还下调了p-JAK2、p-STAT3、SLC7A11和GPX4的表达。进一步使用JAK2抑制剂和激动剂的救援实验证实了JAK2/STAT3通路在这一过程中的关键作用。在体内,TMHZ抑制JAK2/STAT3信号通路促进铁下垂,显著减轻异位子宫内膜腺对肌层的病理性侵袭,并显示出良好的全身安全性。结论tmhz抑制JAK2/STAT3通路,诱导异位病变铁下垂。这种铁致性反应抑制amdc的增殖、迁移和侵袭,最终导致AM进展的衰减。这些发现提供了新的机制见解,并支持TMHZ作为中医框架内AM的潜在治疗策略。
{"title":"Tongmai Huazheng mixture attenuates adenomyosis by inducing ferroptosis through suppression of the JAK2/STAT3 signaling pathway","authors":"Ya-xin Shi ,&nbsp;Cheng-yuan Zhang ,&nbsp;Yu-ning Xie ,&nbsp;Zhi-yong Liu ,&nbsp;Xin Wang ,&nbsp;Ling-di Wang ,&nbsp;Zi-lu Wang ,&nbsp;Hong-bo Yuan ,&nbsp;Rui-hua Zhao ,&nbsp;Wei Shi","doi":"10.1016/j.phymed.2025.157748","DOIUrl":"10.1016/j.phymed.2025.157748","url":null,"abstract":"&lt;div&gt;&lt;h3&gt;Background&lt;/h3&gt;&lt;div&gt;Ferroptosis is a newly characterized iron-dependent form of regulated cell death. In adenomyosis (AM), ectopic endometrial lesions exhibit resistance to ferroptosis, which contributes to disease progression and recurrence. Tongmai Huazheng mixture (TMHZ) is a hospital-formulated traditional Chinese medicine (TCM) compound that has demonstrated clinical efficacy in the management of AM. However, the precise mechanisms by which TMHZ regulates ferroptosis in AM remain unclear.&lt;/div&gt;&lt;/div&gt;&lt;div&gt;&lt;h3&gt;Purpose&lt;/h3&gt;&lt;div&gt;The aim of this study was to determine whether TMHZ exerts its anti-AM effects by inducing ferroptosis through inhibition of the JAK2/STAT3 signaling pathway.&lt;/div&gt;&lt;/div&gt;&lt;div&gt;&lt;h3&gt;Methods&lt;/h3&gt;&lt;div&gt;A multiomics strategy integrating UPLC-MS/MS, network pharmacology, and RNA sequencing was employed to identify candidate targets and key regulatory pathways of TMHZ. &lt;em&gt;In vitro&lt;/em&gt;, the effects of TMHZ-containing serum on proliferation, migration, and invasion of adenomyosis-derived cells (AMDCs) were evaluated using CCK-8, EdU, wound healing, and Transwell assays. Ferroptosis induction and oxidative stress levels were assessed using FerroOrange and CM-H&lt;sub&gt;2&lt;/sub&gt;DCFDA fluorescent probes, glutathione (GSH), superoxide dismutase (SOD), and malondialdehyde (MDA) assay kits, and transmission electron microscopy. Immunofluorescence and western blotting (WB) were performed to investigate the regulatory effects of TMHZ on the JAK2/STAT3 pathway and ferroptosis-related proteins. The JAK2 inhibitor AG490 and agonist coumermycin A1 were used to validate the involvement of the pathway. &lt;em&gt;In vivo&lt;/em&gt;, a tamoxifen-induced AM mouse model was established to assess the therapeutic efficacy and safety of TMHZ. Histopathological changes in the uterus were examined by hematoxylin–eosin (HE) staining, and serum levels of estradiol (E2) and progesterone (P4) were measured to verify model establishment. SOD and MDA levels and the results of immunohistochemical and WB analyses were used to further confirm the &lt;em&gt;in vivo&lt;/em&gt; regulation of ferroptosis and JAK2/STAT3 signaling by TMHZ. Additionally, HE staining of major organs (liver, spleen, and kidney) was performed to evaluate systemic safety.&lt;/div&gt;&lt;/div&gt;&lt;div&gt;&lt;h3&gt;Results&lt;/h3&gt;&lt;div&gt;Bioinformatics analysis suggested that the therapeutic effects of TMHZ were closely associated with the regulation of the JAK2/STAT3 pathway and ferroptosis. &lt;em&gt;In vitro&lt;/em&gt;, TMHZ significantly inhibited the proliferation, migration, and invasion of AMDCs; increased intracellular Fe²⁺ and reactive oxygen species (ROS) levels; reduced GSH and SOD activity; elevated the MDA content, and induced typical mitochondrial damage. TMHZ also downregulated the expressions of p-JAK2, p-STAT3, SLC7A11, and GPX4. Further rescue experiments using JAK2 inhibitors and agonists confirmed the key role of the JAK2/STAT3 pathway in this process. &lt;em&gt;In vivo&lt;/em&gt;, TMHZ suppressed JAK2/STAT3 signaling to promote ferroptosis, ","PeriodicalId":20212,"journal":{"name":"Phytomedicine","volume":"150 ","pages":"Article 157748"},"PeriodicalIF":8.3,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145884120","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
Taxifolin promotes glioma stem cell differentiation via CYP1B1-mediated EMT suppression Taxifolin通过cyp1b1介导的EMT抑制促进胶质瘤干细胞分化。
IF 8.3 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2026-01-01 DOI: 10.1016/j.phymed.2025.157735
Mengxin Li , Jiaying Yang , Keyan Wang , Aonan Zhu , Haoyuan Yin , Zongjun Li , Haoran Zhao , Qi Wang , Dhan V. Kalvakolanu , Yuewen Hao , Bing Cui , Ling Zhang , Baofen Guo

Background

Glioblastoma (GBM) progresses aggressively and resists therapy largely due to glioma stem cells (GSCs), which drive recurrence and treatment failure. Inducing GSC differentiation into non-stem-like cells offers a promising therapeutic approach. Taxifolin (TAX), a natural flavonoid with anti-inflammatory and antioxidant properties, has demonstrated anti-stemness effects in cancer but its impact on GSCs remains unclear.

Purpose

The study aimed to explore TAX as a differentiation-inducing agent targeting GSCs to enhance therapeutic response in GBM.

Study Design

We assessed the effects and mechanism of TAX on GSC proliferation, self-renewal, and differentiation using in vitro and in vivo models, supported by transcriptomic and rescue experiments.

Methods

Using human (TS576) and murine (CSC2078) GSC lines, along with an orthotopic xenograft model, we examined the effects of TAX on proliferation, self-renewal, and differentiation, as well as its downstream pathway. Funcional assays including cell viability, colony formation, neurosphere formation, apoptosis, and differentiation marker analysis were conducted in vitro. Transcriptomic profiling and KEGG pathway enrichment identified CYP1B1 as a key downstream target, whose role was validated through overexpression-based rescue experiments and epithelial-mesenchymal transition (EMT) marker analysis. TAX efficacy and biosafety were evaluated using bioluminescence imaging, histopathology and immunohistochemistry in vivo, with temozolomide as a positive control.

Results

Purified TAX (92.47%) from Larix olgensis roots inhibited the proliferation, self-renewal of GSCs and induced apoptosis. TAX promoted GSCs differentiation toward neuronal lineages, supporting its potential as a differentiation-based adjuvant therapy. TAX suppressed tumor growth without observable toxicity and enhanced the therapeutic efficacy of temozolomide (TMZ) in vivo. Bioinformatic analysis identified CYP1B1 as a TAX-responsive target, with elevated expression in glioma tissues and strong association with malignancy and poor prognosis. Functional assays confirmed that CYP1B1 overexpression promotes EMT, while TAX downregulates CYP1B1 and inhibits EMT progression.

Conclusion

This study is the first to demonstrate that TAX promotes neuronal differentiation of GSCs by suppressing the CYP1B1-mediated EMT pathway. Our work uncovers a novel mechanism linking CYP1B1 down-regulation to GSC differentiation. These findings establish TAX as a differentiation-inducing agent and propose a new therapeutic strategy for GBM by targeting stemness and EMT concurrently.
背景:胶质母细胞瘤(GBM)进展迅速,并对治疗产生抵抗,主要是由于胶质瘤干细胞(GSCs)导致复发和治疗失败。诱导GSC分化为非干细胞样细胞提供了一种很有前途的治疗方法。Taxifolin (TAX)是一种具有抗炎和抗氧化特性的天然类黄酮,已被证明在癌症中具有抗干作用,但其对GSCs的影响尚不清楚。目的:本研究旨在探索TAX作为一种靶向GSCs的分化诱导剂,提高GBM的治疗效果。研究设计:我们利用体外和体内模型,通过转录组学和救援实验,评估了TAX对GSC增殖、自我更新和分化的影响和机制。方法:使用人(TS576)和鼠(CSC2078) GSC系,以及原位异种移植模型,我们研究了TAX对增殖、自我更新和分化的影响,以及它的下游途径。功能分析包括细胞活力、集落形成、神经球形成、凋亡和分化标记分析。转录组学分析和KEGG通路富集鉴定CYP1B1是关键的下游靶点,其作用通过基于过表达的救援实验和上皮-间质转化(EMT)标记分析得到验证。以替莫唑胺为阳性对照,采用活体生物发光成像、组织病理学和免疫组化评价TAX的疗效和生物安全性。结果:落叶松根纯化后的TAX(92.47%)能抑制GSCs的增殖、自我更新并诱导细胞凋亡。TAX促进GSCs向神经元谱系分化,支持其作为基于分化的辅助治疗的潜力。TAX抑制肿瘤生长,无明显毒性,提高替莫唑胺(TMZ)的体内治疗效果。生物信息学分析发现CYP1B1是一个TAX-responsive靶点,在胶质瘤组织中表达升高,与恶性肿瘤和不良预后密切相关。功能分析证实CYP1B1过表达促进EMT,而TAX下调CYP1B1并抑制EMT进展。结论:本研究首次证实了TAX通过抑制cyp1b1介导的EMT通路促进GSCs的神经元分化。我们的工作揭示了一种将CYP1B1下调与GSC分化联系起来的新机制。这些发现证实了TAX是一种分化诱导剂,并提出了同时靶向干细胞和EMT治疗GBM的新策略。
{"title":"Taxifolin promotes glioma stem cell differentiation via CYP1B1-mediated EMT suppression","authors":"Mengxin Li ,&nbsp;Jiaying Yang ,&nbsp;Keyan Wang ,&nbsp;Aonan Zhu ,&nbsp;Haoyuan Yin ,&nbsp;Zongjun Li ,&nbsp;Haoran Zhao ,&nbsp;Qi Wang ,&nbsp;Dhan V. Kalvakolanu ,&nbsp;Yuewen Hao ,&nbsp;Bing Cui ,&nbsp;Ling Zhang ,&nbsp;Baofen Guo","doi":"10.1016/j.phymed.2025.157735","DOIUrl":"10.1016/j.phymed.2025.157735","url":null,"abstract":"<div><h3>Background</h3><div>Glioblastoma (GBM) progresses aggressively and resists therapy largely due to glioma stem cells (GSCs), which drive recurrence and treatment failure. Inducing GSC differentiation into non-stem-like cells offers a promising therapeutic approach. Taxifolin (TAX), a natural flavonoid with anti-inflammatory and antioxidant properties, has demonstrated anti-stemness effects in cancer but its impact on GSCs remains unclear.</div></div><div><h3>Purpose</h3><div>The study aimed to explore TAX as a differentiation-inducing agent targeting GSCs to enhance therapeutic response in GBM.</div></div><div><h3>Study Design</h3><div>We assessed the effects and mechanism of TAX on GSC proliferation, self-renewal, and differentiation using <em>in vitro</em> and <em>in vivo</em> models, supported by transcriptomic and rescue experiments.</div></div><div><h3>Methods</h3><div>Using human (TS576) and murine (CSC2078) GSC lines, along with an orthotopic xenograft model, we examined the effects of TAX on proliferation, self-renewal, and differentiation, as well as its downstream pathway. Funcional assays including cell viability, colony formation, neurosphere formation, apoptosis, and differentiation marker analysis were conducted <em>in vitro</em>. Transcriptomic profiling and KEGG pathway enrichment identified CYP1B1 as a key downstream target, whose role was validated through overexpression-based rescue experiments and epithelial-mesenchymal transition (EMT) marker analysis. TAX efficacy and biosafety were evaluated using bioluminescence imaging, histopathology and immunohistochemistry <em>in vivo</em>, with temozolomide as a positive control.</div></div><div><h3>Results</h3><div>Purified TAX (92.47%) from <em>Larix olgensis</em> roots inhibited the proliferation, self-renewal of GSCs and induced apoptosis. TAX promoted GSCs differentiation toward neuronal lineages, supporting its potential as a differentiation-based adjuvant therapy. TAX suppressed tumor growth without observable toxicity and enhanced the therapeutic efficacy of temozolomide (TMZ) <em>in vivo</em>. Bioinformatic analysis identified CYP1B1 as a TAX-responsive target, with elevated expression in glioma tissues and strong association with malignancy and poor prognosis. Functional assays confirmed that CYP1B1 overexpression promotes EMT, while TAX downregulates CYP1B1 and inhibits EMT progression.</div></div><div><h3>Conclusion</h3><div>This study is the first to demonstrate that TAX promotes neuronal differentiation of GSCs by suppressing the CYP1B1-mediated EMT pathway. Our work uncovers a novel mechanism linking CYP1B1 down-regulation to GSC differentiation. These findings establish TAX as a differentiation-inducing agent and propose a new therapeutic strategy for GBM by targeting stemness and EMT concurrently.</div></div>","PeriodicalId":20212,"journal":{"name":"Phytomedicine","volume":"150 ","pages":"Article 157735"},"PeriodicalIF":8.3,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145900180","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
Corrigendum to "Wuse-oil Compound 03 mitigates radiation-induced oral mucositis by modulating oral microbiota to mediate TLR4/NF-κB pathway and inhibit M1 macrophage polarization" [Phytomedicine volume 145 (2025) 157083]. “wse -oil化合物03通过调节口腔微生物群介导TLR4/NF-κB通路和抑制M1巨噬细胞极化来减轻辐射诱导的口腔黏膜炎”的更正[植物医学卷145(2025)157083]。
IF 8.3 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2026-01-01 Epub Date: 2025-12-24 DOI: 10.1016/j.phymed.2025.157670
Liqun Jia, Rong Huang, Chen Chen, Feng Teng, Tian Gu, Huafeng Liu, Li Ma, Ye Feng, Jingyi Long, Zihan Jin, Dong Li, Dongmei Chen, Yanni Lou
{"title":"Corrigendum to \"Wuse-oil Compound 03 mitigates radiation-induced oral mucositis by modulating oral microbiota to mediate TLR4/NF-κB pathway and inhibit M1 macrophage polarization\" [Phytomedicine volume 145 (2025) 157083].","authors":"Liqun Jia, Rong Huang, Chen Chen, Feng Teng, Tian Gu, Huafeng Liu, Li Ma, Ye Feng, Jingyi Long, Zihan Jin, Dong Li, Dongmei Chen, Yanni Lou","doi":"10.1016/j.phymed.2025.157670","DOIUrl":"10.1016/j.phymed.2025.157670","url":null,"abstract":"","PeriodicalId":20212,"journal":{"name":"Phytomedicine","volume":" ","pages":"157670"},"PeriodicalIF":8.3,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145827988","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