Pub Date : 2025-01-01Epub Date: 2024-12-27DOI: 10.1016/j.phymed.2024.156331
Cuilan Gong, Xinying Fu, Qiang Ma, Menghao He, Xinhua Zhu, Lijuan Liu, Desheng Zhou, Siyang Yan
Ischemic stroke ranks as the second leading cause of global mortality and disability. Although reperfusion is crucial for salvaging brain tissue, it carries the risk of secondary injuries, such as ferroptosis. Gastrodin, a neuroprotective compound found in Chinese herbal medicine, may regulate this process. However, its impact on stroke-induced ferroptosis remains unclear.
Objective: This research endeavors to probe Gastrodin's influence on post-ischemic ferroptosis, deciphering its mechanisms and assessing its therapeutic promise.
Methods: We developed rat models of middle cerebral artery occlusion/reperfusion (MCAO/R) and created oxygen-glucose deprivation/reoxygenation (OGD/R)-damaged PC12 cell models. Gastrodin was administered to assess ferroptosis using Prussian blue staining and fluorescence probes. To investigate the effects of gastrodin on the xCT/GPX4 and ACSL4/LPCAT3 pathways, we employed molecular docking, immunofluorescence, Western blotting, and quantitative real-time polymerase chain reaction (qRT-PCR). Additionally, we used transmission electron microscopy and JC-1 fluorescence probes to examine mitochondrial integrity and function.
Results: Our study demonstrated that gastrodin significantly reduced iron accumulation and lipid peroxidation in the brains of MCAO/R rats and OGD/R-injured PC12 cells. It suppressed reactive oxygen species (ROS) and ameliorated mitochondrial membrane potential. It potentiates the xCT/GPX4 axis while repressing the ACSL4/LPCAT3 pathway, leading to improved mitochondrial architecture and function, notably characterized by decreased mitochondrial membrane potential, reduced ROS levels, and increased formation of mitochondrial cristae. By modulating the xCT/GPX4 and ACSL4/LPCAT3 pathways, gastrodin mitigated ferroptosis in ischemic stroke, thereby preserving mitochondrial structural and functional integrity. This study provides novel mechanistic insights into gastrodin's therapeutic potential for treating ischemic stroke, highlighting the importance of traditional Chinese medicine in modern medical therapy.
{"title":"Gastrodin: Modulating the xCT/GPX4 and ACSL4/LPCAT3 pathways to inhibit ferroptosis after ischemic stroke.","authors":"Cuilan Gong, Xinying Fu, Qiang Ma, Menghao He, Xinhua Zhu, Lijuan Liu, Desheng Zhou, Siyang Yan","doi":"10.1016/j.phymed.2024.156331","DOIUrl":"10.1016/j.phymed.2024.156331","url":null,"abstract":"<p><p>Ischemic stroke ranks as the second leading cause of global mortality and disability. Although reperfusion is crucial for salvaging brain tissue, it carries the risk of secondary injuries, such as ferroptosis. Gastrodin, a neuroprotective compound found in Chinese herbal medicine, may regulate this process. However, its impact on stroke-induced ferroptosis remains unclear.</p><p><strong>Objective: </strong>This research endeavors to probe Gastrodin's influence on post-ischemic ferroptosis, deciphering its mechanisms and assessing its therapeutic promise.</p><p><strong>Methods: </strong>We developed rat models of middle cerebral artery occlusion/reperfusion (MCAO/R) and created oxygen-glucose deprivation/reoxygenation (OGD/R)-damaged PC12 cell models. Gastrodin was administered to assess ferroptosis using Prussian blue staining and fluorescence probes. To investigate the effects of gastrodin on the xCT/GPX4 and ACSL4/LPCAT3 pathways, we employed molecular docking, immunofluorescence, Western blotting, and quantitative real-time polymerase chain reaction (qRT-PCR). Additionally, we used transmission electron microscopy and JC-1 fluorescence probes to examine mitochondrial integrity and function.</p><p><strong>Results: </strong>Our study demonstrated that gastrodin significantly reduced iron accumulation and lipid peroxidation in the brains of MCAO/R rats and OGD/R-injured PC12 cells. It suppressed reactive oxygen species (ROS) and ameliorated mitochondrial membrane potential. It potentiates the xCT/GPX4 axis while repressing the ACSL4/LPCAT3 pathway, leading to improved mitochondrial architecture and function, notably characterized by decreased mitochondrial membrane potential, reduced ROS levels, and increased formation of mitochondrial cristae. By modulating the xCT/GPX4 and ACSL4/LPCAT3 pathways, gastrodin mitigated ferroptosis in ischemic stroke, thereby preserving mitochondrial structural and functional integrity. This study provides novel mechanistic insights into gastrodin's therapeutic potential for treating ischemic stroke, highlighting the importance of traditional Chinese medicine in modern medical therapy.</p>","PeriodicalId":20212,"journal":{"name":"Phytomedicine","volume":"136 ","pages":"156331"},"PeriodicalIF":6.7,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142896907","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}
Background: Andrographolide is a medicinal compound which possesses anti-SARS-CoV-2 activity. A number of cellular targets of andrographolide have been identified by target predictions and computational studies.
Purpose: However, a potential cellular target of andrographolide has never been explored in SARS-CoV-2 infected lung epithelial cells. We aimed to identify cellular pathways involved in andrographolide-mediated anti-SARS-CoV-2 activity.
Methods: The viral infection was determined by immunofluorescence staining, enzyme-linked immunosorbent assay and focus-forming assay. Proteomic analysis was employed to identify cellular pathways and key proteins controlled by andrographolide in the human lung epithelial cells Calu-3 infected by SARS-CoV-2. Immunofluorescence staining was used to test protein expression and localization. Western blot and realtime PCR were utilized to elucidate gene expression. Cellular glutathione level was examined by a reduced/oxidized glutathione assay. An ectopic gene expression was delivered by plasmid transfection.
Results: Gene ontology analysis indicates that proteins involved in nuclear factor erythroid 2-related factor 2 (NRF2)-regulated pathways were differentially expressed by andrographolide. Notably, andrographolide increased expression and nuclear localization of the transcription factor NRF2. In addition, transcriptional expression of GCLC and glutamate-cysteine ligase modifier subunit (GCLM), which are NRF2 target genes, were induced by andrographolide. We further find that infection of SARS-CoV-2 resulted in a reduction of glutathione level in Calu-3; the effect that was rescued by andrographolide. Moreover, andrographolide also induced expression of the glutathione producing enzyme GCLC in SARS-CoV-2 infected lung epithelial cells. Importantly, an ectopic over-expression of GCLC or treatment of N-acetyl-L-cysteine in Calu-3 cells led to a decrease in SARS-CoV-2 infection.
Conclusion: Collectively, our findings suggest the interplay between GCLC-mediated glutathione biogenesis induced by andrographolide and the anti-SARS-CoV-2 activity. The glutathione biogenesis and recycling pathways should be further exploited as a targeted therapy against SARS-CoV-2 infection.
{"title":"Andrographolide attenuates SARS-CoV-2 infection via an up-regulation of glutamate-cysteine ligase catalytic subunit (GCLC).","authors":"Jarinya Chaopreecha, Nut Phueakphud, Ampa Suksatu, Sucheewin Krobthong, Suwimon Manopwisedjaroen, Nattawadee Panyain, Suradej Hongeng, Arunee Thitithanyanont, Patompon Wongtrakoongate","doi":"10.1016/j.phymed.2024.156279","DOIUrl":"10.1016/j.phymed.2024.156279","url":null,"abstract":"<p><strong>Background: </strong>Andrographolide is a medicinal compound which possesses anti-SARS-CoV-2 activity. A number of cellular targets of andrographolide have been identified by target predictions and computational studies.</p><p><strong>Purpose: </strong>However, a potential cellular target of andrographolide has never been explored in SARS-CoV-2 infected lung epithelial cells. We aimed to identify cellular pathways involved in andrographolide-mediated anti-SARS-CoV-2 activity.</p><p><strong>Methods: </strong>The viral infection was determined by immunofluorescence staining, enzyme-linked immunosorbent assay and focus-forming assay. Proteomic analysis was employed to identify cellular pathways and key proteins controlled by andrographolide in the human lung epithelial cells Calu-3 infected by SARS-CoV-2. Immunofluorescence staining was used to test protein expression and localization. Western blot and realtime PCR were utilized to elucidate gene expression. Cellular glutathione level was examined by a reduced/oxidized glutathione assay. An ectopic gene expression was delivered by plasmid transfection.</p><p><strong>Results: </strong>Gene ontology analysis indicates that proteins involved in nuclear factor erythroid 2-related factor 2 (NRF2)-regulated pathways were differentially expressed by andrographolide. Notably, andrographolide increased expression and nuclear localization of the transcription factor NRF2. In addition, transcriptional expression of GCLC and glutamate-cysteine ligase modifier subunit (GCLM), which are NRF2 target genes, were induced by andrographolide. We further find that infection of SARS-CoV-2 resulted in a reduction of glutathione level in Calu-3; the effect that was rescued by andrographolide. Moreover, andrographolide also induced expression of the glutathione producing enzyme GCLC in SARS-CoV-2 infected lung epithelial cells. Importantly, an ectopic over-expression of GCLC or treatment of N-acetyl-L-cysteine in Calu-3 cells led to a decrease in SARS-CoV-2 infection.</p><p><strong>Conclusion: </strong>Collectively, our findings suggest the interplay between GCLC-mediated glutathione biogenesis induced by andrographolide and the anti-SARS-CoV-2 activity. The glutathione biogenesis and recycling pathways should be further exploited as a targeted therapy against SARS-CoV-2 infection.</p>","PeriodicalId":20212,"journal":{"name":"Phytomedicine","volume":"136 ","pages":"156279"},"PeriodicalIF":6.7,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142780693","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}
Background: Owing to high sensitivity and ability for absolute quantification, the droplet digital polymerase chain reaction (ddPCR) is widely used for viral and bacterial detection. However, few studies have been conducted on the application of ddPCR to identify the original plant species used in traditional Chinese medicine and Chinese patent medicine.
Purpose: In this study, we investigated the feasibility of using ddPCR to differentiate between Notopterygium incisum and N. franchetii to establish a sensitive and quantitative method for quality control of herbal materials and preparations.
Methods: Specific minor groove binding (MGB) probes and primers were designed based on stable single nucleotide polymorphisms. The ddPCR experimental conditions were designed and optimised according to the results of multiplex PCR and qPCR, which ultimately confirmed the limits of detection and quantification (LOD and LOQ, respectively) of the method for Notopterygii Rhizoma et Radix. Additionally, the original plant species of Notopterygii Rhizoma et Radix in Jiuwei Qianghuo pills circulating in the market were identified.
Results: The results of the multiplex PCR and qPCR indicated that the probes and primers were specific. Furthermore, a Qsep analyser and Sanger sequencing were used to confirm that the specific amplification products of N. incisum and N. franchetii were 283 and 206 bp, respectively. The optimised ddPCR system was employed to determine the LOD to be 0.000816 ng/µl, and LOQ of N. incisum and N. franchetii to be 0.00408 and 0.003312 ng/µl, respectively. In addition, Notopterygii Rhizoma et Radix in four Jiuwei Qianghuo pills was amplified and successfully identified using ddPCR assays.
Conclusion: This study established a multiplex ddPCR method using MGB probes to identify Notopterygii Rhizoma et Radix, providing a foundation for the identification and quantification of multi-source Chinese herbal medicines.
{"title":"MGB probe-based multiplex droplet digital PCR for the interspecific identification of Notopterygii Rhizoma et Radix in herbal materials and preparations.","authors":"Kai-Ling Xu, Zhong-Mou Zhang, Ya-Dan Wang, Xian-Long Cheng, Hong-Yu Jin, Feng Wei, Shuang-Cheng Ma","doi":"10.1016/j.phymed.2024.156325","DOIUrl":"10.1016/j.phymed.2024.156325","url":null,"abstract":"<p><strong>Background: </strong>Owing to high sensitivity and ability for absolute quantification, the droplet digital polymerase chain reaction (ddPCR) is widely used for viral and bacterial detection. However, few studies have been conducted on the application of ddPCR to identify the original plant species used in traditional Chinese medicine and Chinese patent medicine.</p><p><strong>Purpose: </strong>In this study, we investigated the feasibility of using ddPCR to differentiate between Notopterygium incisum and N. franchetii to establish a sensitive and quantitative method for quality control of herbal materials and preparations.</p><p><strong>Methods: </strong>Specific minor groove binding (MGB) probes and primers were designed based on stable single nucleotide polymorphisms. The ddPCR experimental conditions were designed and optimised according to the results of multiplex PCR and qPCR, which ultimately confirmed the limits of detection and quantification (LOD and LOQ, respectively) of the method for Notopterygii Rhizoma et Radix. Additionally, the original plant species of Notopterygii Rhizoma et Radix in Jiuwei Qianghuo pills circulating in the market were identified.</p><p><strong>Results: </strong>The results of the multiplex PCR and qPCR indicated that the probes and primers were specific. Furthermore, a Qsep analyser and Sanger sequencing were used to confirm that the specific amplification products of N. incisum and N. franchetii were 283 and 206 bp, respectively. The optimised ddPCR system was employed to determine the LOD to be 0.000816 ng/µl, and LOQ of N. incisum and N. franchetii to be 0.00408 and 0.003312 ng/µl, respectively. In addition, Notopterygii Rhizoma et Radix in four Jiuwei Qianghuo pills was amplified and successfully identified using ddPCR assays.</p><p><strong>Conclusion: </strong>This study established a multiplex ddPCR method using MGB probes to identify Notopterygii Rhizoma et Radix, providing a foundation for the identification and quantification of multi-source Chinese herbal medicines.</p>","PeriodicalId":20212,"journal":{"name":"Phytomedicine","volume":"136 ","pages":"156325"},"PeriodicalIF":6.7,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142927810","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}
<p><strong>Object: </strong>Rheumatoid arthritis (RA) is a prevalent and currently incurable autoimmune disease. Existing conventional medical treatments are limited in their efficacy, prolonged disease may lead to bone destruction, joint deformity, and loss of related functions, which places a huge burden on RA patients and their families. For millennia, the use of traditional Chinese medicine (TCM), exemplified by the Gui-Zhi-Shao-Yao-Zhi-Mu decoction (GZSYZM), has been demonstrated to offer distinct therapeutic advantages in the management of RA. Exploring the potential mechanism of GZSYZM in the treatment of RA is a hot topic in the field of TCM.</p><p><strong>Method: </strong>High-throughput sequencing data of RA at bulk level and single cell level and Chinese Materia Medica target-related databases were used as data sources. Ultra-high-performance liquid chromatography coupled with high-resolution mass spectrometry was employed for the identification of the most relevant compounds to the active ingredients present in the GZSYZM granules. Potential disease genes were identified using a combination of differential expression analysis and weighted gene co-expression network analysis, and the "Chinese Materia Medica-Ingredient-Target" network was constructed to obtain candidate drug target genes. The GZSYZM-RA hub genes were then identified based on Molecular Complex Detection algorithm. To explore the associations and potential mechanisms between the GZSYZM-RA hub gene set and RA, Mendelian randomization (MR) analysis and Bayesian co-localization analysis were used to further identify the GZSYZM-RA core genes that were causally associated with RA. A nomogram was constructed based on a multifactorial logistic regression model using the GZSYZM-RA core genes as predictors of RA. To evaluate its diagnostic value, receiver operating characteristic (ROC) curves, calibration curves, and decision curves were plotted. The potential downstream regulatory mechanisms of the gene of interest in GZSYZM in RA therapy were finally investigated using single- gene set enrichment analysis and molecular docking. The aim was to model the optimal conformation of its target protein receptor binding to the small molecule ligand in GZSYZM to identify the key constituents.</p><p><strong>Result: </strong>Functional enrichment analysis revealed that the GZSYZM-RA hub gene set is enriched in several autoimmune-related mechanistic pathways, with a particular emphasis on the phosphoinositide 3 kinase (PI3K)‑serine/threonine kinase (AKT) signaling pathway. AUCell scores demonstrated active expression of the GZSYZM-RA hub gene set with the PI3K-AKT signaling pathway on monocytes, especially non-classical monocytes. Immunol infiltration analysis based on the CIBERSORT algorithm also showed a strong correlation between several genes in the GZSYZM-RA hub gene set and monocytes by calculating Spearman's rank correlation coefficients. MR analysis with co-localization analysis furt
{"title":"Bioinformatics identification based on causal association inference using multi-omics reveals the underlying mechanism of Gui-Zhi-Shao-Yao-Zhi-Mu decoction in modulating rheumatoid arthritis.","authors":"Jiayue Yang, Heng Yang, Fumin Wang, Yao Dai, Yuxuan Deng, Kaiyun Shi, Zehua Zhu, Xinkun Liu, Xiao Ma, Yongxiang Gao","doi":"10.1016/j.phymed.2024.156332","DOIUrl":"10.1016/j.phymed.2024.156332","url":null,"abstract":"<p><strong>Object: </strong>Rheumatoid arthritis (RA) is a prevalent and currently incurable autoimmune disease. Existing conventional medical treatments are limited in their efficacy, prolonged disease may lead to bone destruction, joint deformity, and loss of related functions, which places a huge burden on RA patients and their families. For millennia, the use of traditional Chinese medicine (TCM), exemplified by the Gui-Zhi-Shao-Yao-Zhi-Mu decoction (GZSYZM), has been demonstrated to offer distinct therapeutic advantages in the management of RA. Exploring the potential mechanism of GZSYZM in the treatment of RA is a hot topic in the field of TCM.</p><p><strong>Method: </strong>High-throughput sequencing data of RA at bulk level and single cell level and Chinese Materia Medica target-related databases were used as data sources. Ultra-high-performance liquid chromatography coupled with high-resolution mass spectrometry was employed for the identification of the most relevant compounds to the active ingredients present in the GZSYZM granules. Potential disease genes were identified using a combination of differential expression analysis and weighted gene co-expression network analysis, and the \"Chinese Materia Medica-Ingredient-Target\" network was constructed to obtain candidate drug target genes. The GZSYZM-RA hub genes were then identified based on Molecular Complex Detection algorithm. To explore the associations and potential mechanisms between the GZSYZM-RA hub gene set and RA, Mendelian randomization (MR) analysis and Bayesian co-localization analysis were used to further identify the GZSYZM-RA core genes that were causally associated with RA. A nomogram was constructed based on a multifactorial logistic regression model using the GZSYZM-RA core genes as predictors of RA. To evaluate its diagnostic value, receiver operating characteristic (ROC) curves, calibration curves, and decision curves were plotted. The potential downstream regulatory mechanisms of the gene of interest in GZSYZM in RA therapy were finally investigated using single- gene set enrichment analysis and molecular docking. The aim was to model the optimal conformation of its target protein receptor binding to the small molecule ligand in GZSYZM to identify the key constituents.</p><p><strong>Result: </strong>Functional enrichment analysis revealed that the GZSYZM-RA hub gene set is enriched in several autoimmune-related mechanistic pathways, with a particular emphasis on the phosphoinositide 3 kinase (PI3K)‑serine/threonine kinase (AKT) signaling pathway. AUCell scores demonstrated active expression of the GZSYZM-RA hub gene set with the PI3K-AKT signaling pathway on monocytes, especially non-classical monocytes. Immunol infiltration analysis based on the CIBERSORT algorithm also showed a strong correlation between several genes in the GZSYZM-RA hub gene set and monocytes by calculating Spearman's rank correlation coefficients. MR analysis with co-localization analysis furt","PeriodicalId":20212,"journal":{"name":"Phytomedicine","volume":"136 ","pages":"156332"},"PeriodicalIF":6.7,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142907538","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}
Pub Date : 2025-01-01DOI: 10.1016/j.phymed.2025.156361
Lucia Mamede, Gabriel W Rangel, Lahngong Methodius Shinyuy, Naïma Boussif, Marie-France Herent, Bernadette Govaerts, Olivia Jansen, Allison Ledoux, Pascal De Tullio, Joëlle Quetin-Leclercq, Manuel Llinás, Michel Frédérich
Background: Artemisia spp. have been used for millennia in traditional medicine to treat a variety of ailments, including malaria. Extracts of Artemisia afra and A. annua remain widely used throughout Africa for healthcare purposes, notably to prevent and/or treat malaria. However, the modes of action of these plant extracts remain unclear, with contradictory reports regarding the presence and role of artemisinin in both plants.
Purpose: The aim of this study was to identify differences in the antimalarial mode of action of A. afra and A. annua by measuring their phenolic profiles and comparing their effect on parasite metabolism in vitro.
Methods: In this work, we analyzed the phenolic profile of A. afra and A. annua extracts through high-performance liquid chromatography (HPLC), detected and quantified artemisinin through HPLC and mass spectrometry (MS), and performed comparative HPLC-MS metabolomic analysis on in vitro-cultured Plasmodium falciparum trophozoites to elucidate the potential modes of action of these plant extracts.
Results: A. afra contained only trace amounts of artemisinin and elicited a different parasite metabolic response compared to A. annua, which contained significantly more artemisinin and correlated closely with the parasite response profile elicited by purified artemisinin. A. annua impacted parasite glutathione metabolism in agreement with the established redox activity of artemisinin, while A. afra had an effect on lipid precursors.
Conclusions: This study reveals that A. afra and A. annua have divergent effects on Plasmodium falciparum metabolism and provides support for ongoing efforts exploring the use of A. afra for the treatment of malaria.
背景:千百年来,传统医学一直使用蒿属植物来治疗包括疟疾在内的各种疾病。在整个非洲,黄花蒿和青蒿的提取物仍被广泛用于保健目的,特别是预防和/或治疗疟疾。目的:本研究的目的是通过测量 A. afra 和 A. annua 的酚类物质含量并比较其对寄生虫体外代谢的影响,来确定这两种植物提取物抗疟作用模式的差异:在这项工作中,我们通过高效液相色谱法(HPLC)分析了 A. afra 和 A. annua 提取物的酚类概况,通过 HPLC 和质谱法(MS)检测并量化了青蒿素,并对体外培养的恶性疟原虫滋养体进行了 HPLC-MS 代谢组学比较分析,以阐明这些植物提取物的潜在作用模式:A. afra仅含有微量的青蒿素,与A. annua相比,A. annua含有更多的青蒿素,与纯化青蒿素引起的寄生虫反应特征密切相关,但A. afra引起的寄生虫代谢反应与A. annua不同。青蒿素影响寄生虫的谷胱甘肽代谢,这与青蒿素已确定的氧化还原活性一致,而Afra则对脂质前体有影响:这项研究揭示了 A. afra 和 A. annua 对恶性疟原虫新陈代谢的不同影响,并为目前探索使用 A. afra 治疗疟疾的努力提供了支持。
{"title":"Metabolite profiling of Artemisia afra and Artemisia annua extracts reveals divergent effects on Plasmodium falciparum.","authors":"Lucia Mamede, Gabriel W Rangel, Lahngong Methodius Shinyuy, Naïma Boussif, Marie-France Herent, Bernadette Govaerts, Olivia Jansen, Allison Ledoux, Pascal De Tullio, Joëlle Quetin-Leclercq, Manuel Llinás, Michel Frédérich","doi":"10.1016/j.phymed.2025.156361","DOIUrl":"https://doi.org/10.1016/j.phymed.2025.156361","url":null,"abstract":"<p><strong>Background: </strong>Artemisia spp. have been used for millennia in traditional medicine to treat a variety of ailments, including malaria. Extracts of Artemisia afra and A. annua remain widely used throughout Africa for healthcare purposes, notably to prevent and/or treat malaria. However, the modes of action of these plant extracts remain unclear, with contradictory reports regarding the presence and role of artemisinin in both plants.</p><p><strong>Purpose: </strong>The aim of this study was to identify differences in the antimalarial mode of action of A. afra and A. annua by measuring their phenolic profiles and comparing their effect on parasite metabolism in vitro.</p><p><strong>Methods: </strong>In this work, we analyzed the phenolic profile of A. afra and A. annua extracts through high-performance liquid chromatography (HPLC), detected and quantified artemisinin through HPLC and mass spectrometry (MS), and performed comparative HPLC-MS metabolomic analysis on in vitro-cultured Plasmodium falciparum trophozoites to elucidate the potential modes of action of these plant extracts.</p><p><strong>Results: </strong>A. afra contained only trace amounts of artemisinin and elicited a different parasite metabolic response compared to A. annua, which contained significantly more artemisinin and correlated closely with the parasite response profile elicited by purified artemisinin. A. annua impacted parasite glutathione metabolism in agreement with the established redox activity of artemisinin, while A. afra had an effect on lipid precursors.</p><p><strong>Conclusions: </strong>This study reveals that A. afra and A. annua have divergent effects on Plasmodium falciparum metabolism and provides support for ongoing efforts exploring the use of A. afra for the treatment of malaria.</p>","PeriodicalId":20212,"journal":{"name":"Phytomedicine","volume":"136 ","pages":"156361"},"PeriodicalIF":6.7,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142984625","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}
Background: Promoting the coupling of osteogenesis and angiogenesis is a crucial strategy for the treatment of postmenopausal osteoporosis (PMOP). Estrogen deficiency induces ferroptosis, which is closely associated with the pathophysiology of PMOP. Sarsasapogenin (SAR) is a natural sapogenin with anti-oxidative effects. However, it is unclear whether SAR has a protective role against the impaired osteogenesis and angiogenesis coupling in PMOP. In this study, we evaluated the efficacy of SAR in estrogen deficiency-induced osteoporosis and explored the underlying mechanisms.
Methods: Bone marrow mesenchymal stem cells (BMSCs) and human umbilical vein endothelial cells (HUVECs) were utilized to assess the in vitro effects of SAR on the coupling of osteogenesis and angiogenesis. In vivo experiments involved bilateral ovariectomy (OVX)-induced osteoporosis in mice and glutathione peroxidase 4 (GPX4)-knockout (KO) mice. Mice were orally administered SAR (5 or 10 mg/kg/d) for a duration of 12 weeks. The direct target of SAR was investigated through molecular docking, a cellular thermal shift assay, and surface plasmon resonance. Additionally, RNA sequencing was employed to elucidate the underlying mechanisms.
Results: SAR treatment improved cell viability and osteogenic differentiation while inhibiting ferroptosis in iron dextran-induced BMSCs. Furthermore, SAR enhanced the production of slit guidance ligand 3 (SLIT3) in these cells, which stimulated angiogenesis by activating its receptor, roundabout human homolog 1 (ROBO1), in HUVECs. An in vitro model of ferroptosis induced by erastin demonstrated that SAR promoted the coupling of osteogenesis and angiogenesis by upregulating the BMSCs-SLIT3/HUVECs-ROBO1 axis. Activation of GPX4 was identified as a contributing factor to the effects of SAR on this coupling. Transfection of GPX4 small interfering RNA (siRNA) in BMSCs negated the impact of SAR on the BMSCs-SLIT3/HUVECs-ROBO1 axis. Additionally, SAR was found to directly interact with GPX4, enhancing protein stability, with an equilibrium dissociation constant of 44.6 μM. Notably, SAR did not increase SLIT3, ROBO1, or indicators of osteogenesis or angiogenesis in GPX4-KO mice.
Conclusions: These findings underscore the significance of restoring the GPX4/SLIT3/ROBO1 axis in promoting the coupling of angiogenesis and osteogenesis. SAR mitigates PMOP, in part, by activating the BMSCs-SLIT3/HUVECs-ROBO1 axis, with GPX4 serving as an upstream signaling modulator responsible for SLIT3 production. Our observations provide experimental evidence supporting the clinical application of SAR in the treatment of PMOP.
{"title":"Sarsasapogenin stimulates angiogenesis and osteogenesis coupling to treat estrogen deficiency-induced osteoporosis by activating the GPX4/SLIT3/ROBO1 axis.","authors":"Fang Wang, Fanxuan Zhang, Bingfeng Lin, Wenlong Xiao, Xuchen Wang, Nani Wang","doi":"10.1016/j.phymed.2024.156297","DOIUrl":"10.1016/j.phymed.2024.156297","url":null,"abstract":"<p><strong>Background: </strong>Promoting the coupling of osteogenesis and angiogenesis is a crucial strategy for the treatment of postmenopausal osteoporosis (PMOP). Estrogen deficiency induces ferroptosis, which is closely associated with the pathophysiology of PMOP. Sarsasapogenin (SAR) is a natural sapogenin with anti-oxidative effects. However, it is unclear whether SAR has a protective role against the impaired osteogenesis and angiogenesis coupling in PMOP. In this study, we evaluated the efficacy of SAR in estrogen deficiency-induced osteoporosis and explored the underlying mechanisms.</p><p><strong>Methods: </strong>Bone marrow mesenchymal stem cells (BMSCs) and human umbilical vein endothelial cells (HUVECs) were utilized to assess the in vitro effects of SAR on the coupling of osteogenesis and angiogenesis. In vivo experiments involved bilateral ovariectomy (OVX)-induced osteoporosis in mice and glutathione peroxidase 4 (GPX4)-knockout (KO) mice. Mice were orally administered SAR (5 or 10 mg/kg/d) for a duration of 12 weeks. The direct target of SAR was investigated through molecular docking, a cellular thermal shift assay, and surface plasmon resonance. Additionally, RNA sequencing was employed to elucidate the underlying mechanisms.</p><p><strong>Results: </strong>SAR treatment improved cell viability and osteogenic differentiation while inhibiting ferroptosis in iron dextran-induced BMSCs. Furthermore, SAR enhanced the production of slit guidance ligand 3 (SLIT3) in these cells, which stimulated angiogenesis by activating its receptor, roundabout human homolog 1 (ROBO1), in HUVECs. An in vitro model of ferroptosis induced by erastin demonstrated that SAR promoted the coupling of osteogenesis and angiogenesis by upregulating the BMSCs-SLIT3/HUVECs-ROBO1 axis. Activation of GPX4 was identified as a contributing factor to the effects of SAR on this coupling. Transfection of GPX4 small interfering RNA (siRNA) in BMSCs negated the impact of SAR on the BMSCs-SLIT3/HUVECs-ROBO1 axis. Additionally, SAR was found to directly interact with GPX4, enhancing protein stability, with an equilibrium dissociation constant of 44.6 μM. Notably, SAR did not increase SLIT3, ROBO1, or indicators of osteogenesis or angiogenesis in GPX4-KO mice.</p><p><strong>Conclusions: </strong>These findings underscore the significance of restoring the GPX4/SLIT3/ROBO1 axis in promoting the coupling of angiogenesis and osteogenesis. SAR mitigates PMOP, in part, by activating the BMSCs-SLIT3/HUVECs-ROBO1 axis, with GPX4 serving as an upstream signaling modulator responsible for SLIT3 production. Our observations provide experimental evidence supporting the clinical application of SAR in the treatment of PMOP.</p>","PeriodicalId":20212,"journal":{"name":"Phytomedicine","volume":"136 ","pages":"156297"},"PeriodicalIF":6.7,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142786426","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}
<p><strong>Background: </strong>Abnormal antioxidant capacity in cancer cells is intimately linked to tumor aggressiveness. Modulating oxidative stress status and inhibiting ferroptosis represents a novel anticancer therapeutic strategy. STAM Binding Protein Like 1 (STAMBPL1), a deubiquitinase, is implicated in various malignancies, yet its function in inhibiting ferroptosis and therapeutic potential for cholangiocarcinoma (CCA) remains unexplored.</p><p><strong>Purpose: </strong>This study elucidates STAMBPL1's function in ferroptosis and evaluates liquidambaric acid (LDA) as its inhibitor for therapeutic applications.</p><p><strong>Methods: </strong>Using bioinformatics, WB, IHC, the expression and prognostic value of STAMBPL1 in CCA tissue was detected. The carcinogenic capacity of STAMBPL1 and LDA were assessed through CCK-8, EdU, cloning, transwell, scratch, apoptosis, and cell cycle assays. Flow cytometry and fluorescence microscopy, as well as transmission electron microscopy (TEM), examines the effects of STAMBPL1 and LDA on intracellular reactive oxygen species (ROS) and changes in mitochondrial membrane potential. The tumorigenic ability of STAMBPL1 and LDA in vivo was evaluated through subcutaneous tumor model and lung metastasis model. The underlying mechanism of STAMBPL1 was explored using immunoprecipitation coupled with Mass spectrometry (IP/MS), Co-immunoprecipitation (Co-IP), GST pull-down, DNA pull-down, and Dual-luciferase reporter assays. Molecular docking simulations, SPR, DARTS and CETSA predict the putative binding site of LDA on STAMBPL1 protein. Rescue experiments further confirmed the above conclusions.</p><p><strong>Results: </strong>This study unveils the upregulation and oncogenic role of STAMBPL1 in CCA. Functionally, STAMBPL1 notably enhances CCA cell proliferation and metastasis while impeding ferroptosis. STAMBPL1 stabilizes NRF2, a pivotal regulator of antioxidant enzymes, through K63 deubiquitination. Elevated NRF2, stabilized by STAMBPL1 overexpression, triggers GPX4 activation and reactive oxygen species (ROS) elimination. Particularly, sites 251-436 of STAMBPL1 interact with sites 228-605 of NRF2, facilitating DUB activity and eliminating ubiquitin molecules attached to NRF2, thus protecting it from proteasome-mediated degradation. Moreover, NRF2, acting as a transcription factor, binds to the promoter region of STAMBPL1 and activates its transcription, thus forming STAMBPL1/NRF2 positive feedback loop and regulating redox homeostasis. Molecular docking and in vitro/in vivo experiments identified that LDA binds to and inhibits STAMBPL1, thereby disrupting the STAMBPL1/NRF2 positive feedback loop, consequently suppressing CCA progression.</p><p><strong>Conclusion: </strong>This study firstly reveals that STAMBPL1 promotes cholangiocarcinoma progression by upregulating NRF2, indicating that targeting the STAMBPL1/NRF2 axis is a novel therapeutic strategy. Additionally, our findings firstly suggest that LDA can
{"title":"Liquidambaric acid inhibits cholangiocarcinoma progression by disrupting the STAMBPL1/NRF2 positive feedback loop.","authors":"Zhihuai Wang, Yinjie Zhang, Yuhang Shen, Chunfu Zhu, Xihu Qin, Yuan Gao","doi":"10.1016/j.phymed.2024.156303","DOIUrl":"10.1016/j.phymed.2024.156303","url":null,"abstract":"<p><strong>Background: </strong>Abnormal antioxidant capacity in cancer cells is intimately linked to tumor aggressiveness. Modulating oxidative stress status and inhibiting ferroptosis represents a novel anticancer therapeutic strategy. STAM Binding Protein Like 1 (STAMBPL1), a deubiquitinase, is implicated in various malignancies, yet its function in inhibiting ferroptosis and therapeutic potential for cholangiocarcinoma (CCA) remains unexplored.</p><p><strong>Purpose: </strong>This study elucidates STAMBPL1's function in ferroptosis and evaluates liquidambaric acid (LDA) as its inhibitor for therapeutic applications.</p><p><strong>Methods: </strong>Using bioinformatics, WB, IHC, the expression and prognostic value of STAMBPL1 in CCA tissue was detected. The carcinogenic capacity of STAMBPL1 and LDA were assessed through CCK-8, EdU, cloning, transwell, scratch, apoptosis, and cell cycle assays. Flow cytometry and fluorescence microscopy, as well as transmission electron microscopy (TEM), examines the effects of STAMBPL1 and LDA on intracellular reactive oxygen species (ROS) and changes in mitochondrial membrane potential. The tumorigenic ability of STAMBPL1 and LDA in vivo was evaluated through subcutaneous tumor model and lung metastasis model. The underlying mechanism of STAMBPL1 was explored using immunoprecipitation coupled with Mass spectrometry (IP/MS), Co-immunoprecipitation (Co-IP), GST pull-down, DNA pull-down, and Dual-luciferase reporter assays. Molecular docking simulations, SPR, DARTS and CETSA predict the putative binding site of LDA on STAMBPL1 protein. Rescue experiments further confirmed the above conclusions.</p><p><strong>Results: </strong>This study unveils the upregulation and oncogenic role of STAMBPL1 in CCA. Functionally, STAMBPL1 notably enhances CCA cell proliferation and metastasis while impeding ferroptosis. STAMBPL1 stabilizes NRF2, a pivotal regulator of antioxidant enzymes, through K63 deubiquitination. Elevated NRF2, stabilized by STAMBPL1 overexpression, triggers GPX4 activation and reactive oxygen species (ROS) elimination. Particularly, sites 251-436 of STAMBPL1 interact with sites 228-605 of NRF2, facilitating DUB activity and eliminating ubiquitin molecules attached to NRF2, thus protecting it from proteasome-mediated degradation. Moreover, NRF2, acting as a transcription factor, binds to the promoter region of STAMBPL1 and activates its transcription, thus forming STAMBPL1/NRF2 positive feedback loop and regulating redox homeostasis. Molecular docking and in vitro/in vivo experiments identified that LDA binds to and inhibits STAMBPL1, thereby disrupting the STAMBPL1/NRF2 positive feedback loop, consequently suppressing CCA progression.</p><p><strong>Conclusion: </strong>This study firstly reveals that STAMBPL1 promotes cholangiocarcinoma progression by upregulating NRF2, indicating that targeting the STAMBPL1/NRF2 axis is a novel therapeutic strategy. Additionally, our findings firstly suggest that LDA can ","PeriodicalId":20212,"journal":{"name":"Phytomedicine","volume":"136 ","pages":"156303"},"PeriodicalIF":6.7,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142872720","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}
Pub Date : 2025-01-01Epub Date: 2024-12-18DOI: 10.1016/j.phymed.2024.156337
Yong-Chun Peng, Zhi-Jing He, Lun-Cai Yin, Hui-Feng Pi, Yi Jiang, Ke-Yan Li, Li Tian, Jia Xie, Jian-Bo Zhang, Chen-Yao Li, Guan-Ying Feng, Kai Wang, Ding-Zhou Zhou, Xiao-Wei Xie, Zhi-Yuan Zhang, Teng-Fei Fan
Background: Oral squamous cell carcinoma (OSCC) is one of the most common malignancies. However, there is no effective treatment for OSCC.
Purpose: This study aimed to identify a natural compound with significant efficacy against OSCC and elucidate its primary mechanism of action.
Methods: An FDA-approved drug library and an MCE autophagy-related molecular compound library were screened through high-throughput screening to identify an effective natural compound against OSCC. The IC50 value of sanguinarine (Sang) in OSCC cells was determined using a CCK8 assay. Immunoblotting and immunofluorescence staining were used to assess the effect of Sang on autophagic flux in OSCC cells. Changes in the acidic lysosomal environment were evaluated using RFP-GFP-LC3B and LysoSensor Green DND-189. Furthermore, limited proteolysis-coupled mass spectrometry (LiP-MS) and virtual screening techniques were utilized to identify direct binding targets of Sang, which were subsequently validated by surface plasmon resonance (SPR) and microscale thermophoresis (MST). Molecular docking combined with molecular dynamics analysis identified the binding site between the target protein and Sang. In vitro and in vivo investigations with mutant plasmids confirmed this finding.
Results: Screening led to the identification of the naturally occurring autophagy modulator Sang as a potent inhibitor of OSCC progression. Moreover, Sang impaired lysosomal function through reducing lysosomal-associated membrane proteins, inhibiting lysosomal proteolysis, and altering the lysosomal pH. These effects contributed to defects in autophagic clearance and subsequently suppressed OSCC progression. Notably, Sang bound the phenylalanine 26 (F26) residue in pyruvate kinase M2 (PKM2) and inhibited PKM2 enzymatic activity, subsequently suppressing transcription factor EB (TFEB) expression to inhibit lysosomal function and blocking autophagic flux in OSCC cells.
Conclusion: Our results demonstrate for the first time that Sang can suppress the PKM2/TFEB axis, and influence lysosomal function, thereby blocking autophagy and inhibiting the progression of OSCC, making it a promising therapeutic option for the treatment of OSCC.
背景:口腔鳞状细胞癌是最常见的恶性肿瘤之一。然而,OSCC没有有效的治疗方法。目的:本研究旨在鉴定一种具有明显抗OSCC作用的天然化合物,并阐明其主要作用机制。方法:通过高通量筛选筛选fda批准的药物文库和MCE自噬相关分子化合物文库,鉴定抗OSCC的有效天然化合物。采用CCK8法测定血甘氨酸(Sang)在OSCC细胞中的IC50值。采用免疫印迹法和免疫荧光法观察桑对OSCC细胞自噬通量的影响。使用RFP-GFP-LC3B和LysoSensor Green DND-189评估酸性溶酶体环境的变化。此外,利用有限蛋白水解耦合质谱(LiP-MS)和虚拟筛选技术鉴定了Sang的直接结合靶点,随后通过表面等离子体共振(SPR)和微尺度热泳术(MST)进行了验证。分子对接结合分子动力学分析确定了目标蛋白与Sang的结合位点。用突变质粒进行的体外和体内研究证实了这一发现。结果:筛选发现自然发生的自噬调节剂Sang是一种有效的OSCC进展抑制剂。此外,Sang通过减少溶酶体相关膜蛋白,抑制溶酶体蛋白水解和改变溶酶体ph来损害溶酶体功能。这些影响导致自噬清除缺陷,随后抑制OSCC进展。值得注意的是,Sang结合丙酮酸激酶M2 (PKM2)中的苯丙氨酸26 (F26)残基,抑制PKM2的酶活性,随后抑制转录因子EB (TFEB)的表达,抑制溶酶体功能,阻断OSCC细胞的自噬通量。结论:我们的研究结果首次证明桑可抑制PKM2/TFEB轴,影响溶酶体功能,从而阻断自噬,抑制OSCC的进展,使其成为治疗OSCC的有希望的治疗选择。
{"title":"Sanguinarine suppresses oral squamous cell carcinoma progression by targeting the PKM2/TFEB aix to inhibit autophagic flux.","authors":"Yong-Chun Peng, Zhi-Jing He, Lun-Cai Yin, Hui-Feng Pi, Yi Jiang, Ke-Yan Li, Li Tian, Jia Xie, Jian-Bo Zhang, Chen-Yao Li, Guan-Ying Feng, Kai Wang, Ding-Zhou Zhou, Xiao-Wei Xie, Zhi-Yuan Zhang, Teng-Fei Fan","doi":"10.1016/j.phymed.2024.156337","DOIUrl":"10.1016/j.phymed.2024.156337","url":null,"abstract":"<p><strong>Background: </strong>Oral squamous cell carcinoma (OSCC) is one of the most common malignancies. However, there is no effective treatment for OSCC.</p><p><strong>Purpose: </strong>This study aimed to identify a natural compound with significant efficacy against OSCC and elucidate its primary mechanism of action.</p><p><strong>Methods: </strong>An FDA-approved drug library and an MCE autophagy-related molecular compound library were screened through high-throughput screening to identify an effective natural compound against OSCC. The IC50 value of sanguinarine (Sang) in OSCC cells was determined using a CCK8 assay. Immunoblotting and immunofluorescence staining were used to assess the effect of Sang on autophagic flux in OSCC cells. Changes in the acidic lysosomal environment were evaluated using RFP-GFP-LC3B and LysoSensor Green DND-189. Furthermore, limited proteolysis-coupled mass spectrometry (LiP-MS) and virtual screening techniques were utilized to identify direct binding targets of Sang, which were subsequently validated by surface plasmon resonance (SPR) and microscale thermophoresis (MST). Molecular docking combined with molecular dynamics analysis identified the binding site between the target protein and Sang. In vitro and in vivo investigations with mutant plasmids confirmed this finding.</p><p><strong>Results: </strong>Screening led to the identification of the naturally occurring autophagy modulator Sang as a potent inhibitor of OSCC progression. Moreover, Sang impaired lysosomal function through reducing lysosomal-associated membrane proteins, inhibiting lysosomal proteolysis, and altering the lysosomal pH. These effects contributed to defects in autophagic clearance and subsequently suppressed OSCC progression. Notably, Sang bound the phenylalanine 26 (F26) residue in pyruvate kinase M2 (PKM2) and inhibited PKM2 enzymatic activity, subsequently suppressing transcription factor EB (TFEB) expression to inhibit lysosomal function and blocking autophagic flux in OSCC cells.</p><p><strong>Conclusion: </strong>Our results demonstrate for the first time that Sang can suppress the PKM2/TFEB axis, and influence lysosomal function, thereby blocking autophagy and inhibiting the progression of OSCC, making it a promising therapeutic option for the treatment of OSCC.</p>","PeriodicalId":20212,"journal":{"name":"Phytomedicine","volume":"136 ","pages":"156337"},"PeriodicalIF":6.7,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142896912","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}
Background: Heart failure (HF) represents an advanced stage of various cardiovascular disorders, with its elevated admission rates and resultant health economic burden posing an ongoing global concern.
Purpose: To evaluate the health and economic benefits of herbal medicine (HM) for patients with HF.
Study design: Population-based cohort study.
Methods: A five-year retrospective cohort study was carried out at a nationally recognized hospital in China. The study utilized propensity score matching (PSM) to match patients with HF. Chi-square tests were used to analyze dichotomous variables, and t-tests were employed for continuous variables. Logistic regression was used to examine hospital readmission rates, while multiple linear regression was utilized to evaluate direct medical costs. Statistical significance was set at p < 0.05.
Results: After implementing PSM, 1924 HF patients were included in the analysis. The study identified two significant risk factors affecting the readmission rates: age over 65 years (adjusted odds ratio (OR) = 1.25, 95 % confidence interval (CI) [1.02, 1.53]) and smoking (adjusted OR = 1.31, 95 % CI [1.01, 1.70]). Additionally, patients who received adjunctive HM treatment exhibited a significantly lower readmission rate compared to those without HM treatment (adjusted OR = 0.76, 95 % CI [0.64, 0.92]). Furthermore, the use of HM during patient hospitalization did not significantly impact direct medical expenses but instead provided positive health economic benefits (incremental cost-effectiveness ratio (ICER) = 98.52). Factors influencing direct routine medical costs included over 65 years of age (Coef = 60.78, 95 % CI [36.25, 85.31]), and cardiac function classification (New York Heart Association (NYHA) III: Coef = 1979.92, 95 % CI [1401.82, 2558.03]; NYHA IV: Coef = 6052.48, 95 % CI [5166.59, 6938.38]).
Conclusions: The integration of HM in patients with HF reduced readmission rates without a notable increase in direct medical costs, and the expense of HM remains an economically range indicating positive health economic outcomes.
{"title":"Health and economic evaluation of herbal medicines for heart failure: A population-based cohort study.","authors":"Jianbo Guo, Xinyu Lu, Pei Zhang, Ruolin Du, Chen Liu, Guang Chen, Xiangjun Yin, Tiantian Meng, Anqi Li, Haiyong Chen, Qingyong He","doi":"10.1016/j.phymed.2024.156310","DOIUrl":"10.1016/j.phymed.2024.156310","url":null,"abstract":"<p><strong>Background: </strong>Heart failure (HF) represents an advanced stage of various cardiovascular disorders, with its elevated admission rates and resultant health economic burden posing an ongoing global concern.</p><p><strong>Purpose: </strong>To evaluate the health and economic benefits of herbal medicine (HM) for patients with HF.</p><p><strong>Study design: </strong>Population-based cohort study.</p><p><strong>Methods: </strong>A five-year retrospective cohort study was carried out at a nationally recognized hospital in China. The study utilized propensity score matching (PSM) to match patients with HF. Chi-square tests were used to analyze dichotomous variables, and t-tests were employed for continuous variables. Logistic regression was used to examine hospital readmission rates, while multiple linear regression was utilized to evaluate direct medical costs. Statistical significance was set at p < 0.05.</p><p><strong>Results: </strong>After implementing PSM, 1924 HF patients were included in the analysis. The study identified two significant risk factors affecting the readmission rates: age over 65 years (adjusted odds ratio (OR) = 1.25, 95 % confidence interval (CI) [1.02, 1.53]) and smoking (adjusted OR = 1.31, 95 % CI [1.01, 1.70]). Additionally, patients who received adjunctive HM treatment exhibited a significantly lower readmission rate compared to those without HM treatment (adjusted OR = 0.76, 95 % CI [0.64, 0.92]). Furthermore, the use of HM during patient hospitalization did not significantly impact direct medical expenses but instead provided positive health economic benefits (incremental cost-effectiveness ratio (ICER) = 98.52). Factors influencing direct routine medical costs included over 65 years of age (Coef = 60.78, 95 % CI [36.25, 85.31]), and cardiac function classification (New York Heart Association (NYHA) III: Coef = 1979.92, 95 % CI [1401.82, 2558.03]; NYHA IV: Coef = 6052.48, 95 % CI [5166.59, 6938.38]).</p><p><strong>Conclusions: </strong>The integration of HM in patients with HF reduced readmission rates without a notable increase in direct medical costs, and the expense of HM remains an economically range indicating positive health economic outcomes.</p>","PeriodicalId":20212,"journal":{"name":"Phytomedicine","volume":"136 ","pages":"156310"},"PeriodicalIF":6.7,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142828698","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}
Pub Date : 2025-01-01Epub Date: 2024-12-18DOI: 10.1016/j.phymed.2024.156339
Yuhao Wang, Canglang Mou, Lei Huang, Jinghan Su, Long You, Jianmei Zhang, Ziliang He, Yeye Hu, Khin Myo Htwe, Seung-Gyu Lee, Jinwhoa Yum, Yerin Ha, Ji Heun Lee, Youngwoon Ju, Wooram Choi, Jae Youl Cho
Background: Inflammation is the body's innate reaction to foreign pathogens and serves as a self-regulating mechanism. However, the immune system can mistakenly target the body's own tissues, triggering unnecessary inflammation. For millennia, medicinal plants have been employed for the treatment of diseases. One such plant, Rhaphidophora peepla, has demonstrated potential anti-inflammatory properties. However, the precise mechanism underlying its anti-inflammatory effects remains elusive.
Study design: For this study, validation of target molecules by different experimental approaches and employing two different in vivo experiments were tried to improve the immunopharmacological value of Rhaphidophora peepla.
Purpose: Our goal is to elucidate the mechanism through which the ethanol extract of Rhaphidophora peepla (Rp-EE) demonstrates anti-inflammatory properties, both in vivo and in vitro.
Method: Rp-EE was phytochemically analyzed with gas chromatography-mass spectrometry (GC-MS) and high-performance liquid chromatography (HPLC). Bioinformatic analysis with protein-protein interaction (PPI) networks and Kyoto Encyclopedia of Genes and Genomes (KEGG), nitric oxide (NO) assay, MTT assay, RT-PCR, ELISA, luciferase assay, CETSA, hematoxylin and eosin (H&E) staining, and Western blotting analysis were used to evaluate anti-inflammatory activity of Rp-EE and its mechanism.
Results: Rp-EE significantly reduced inflammatory responses including nitric oxide (NO) release induced by lipopolysaccharide (LPS) at the non-cytotoxic concentrations in vitro, and HCl/EtOH-induced gastritis and LPS-induced acute lung injury models in vivo. Mechanistically, it was revealed that Rp-EE can specifically target spleen tyrosine kinase (Syk) and transforming growth factor β-activated kinase 1 (TAK1) to suppress the phosphorylation levels of nuclear factor (NF)-κB subunits (p65 and p50) and activator protein (AP)-1 subunits (c-Jun and c-Fos).
Conclusion: Rp-EE can inhibit inflammatory reactions managed by Syk and TAK1, resulting in suppressing the Syk/AKT/NF-κB and TAK1/MAPK/AP-1 signaling pathways. These findings lead us to a possibility that Rp-EE can be developed as a promising anti-gastric ulcer and anti-lung injury remedy.
{"title":"The ethanolic extract of Rhaphidophora peepla prevents inflammation by inhibiting the activation of Syk/AKT/NF-κB and TAK1/MAPK/AP-1.","authors":"Yuhao Wang, Canglang Mou, Lei Huang, Jinghan Su, Long You, Jianmei Zhang, Ziliang He, Yeye Hu, Khin Myo Htwe, Seung-Gyu Lee, Jinwhoa Yum, Yerin Ha, Ji Heun Lee, Youngwoon Ju, Wooram Choi, Jae Youl Cho","doi":"10.1016/j.phymed.2024.156339","DOIUrl":"https://doi.org/10.1016/j.phymed.2024.156339","url":null,"abstract":"<p><strong>Background: </strong>Inflammation is the body's innate reaction to foreign pathogens and serves as a self-regulating mechanism. However, the immune system can mistakenly target the body's own tissues, triggering unnecessary inflammation. For millennia, medicinal plants have been employed for the treatment of diseases. One such plant, Rhaphidophora peepla, has demonstrated potential anti-inflammatory properties. However, the precise mechanism underlying its anti-inflammatory effects remains elusive.</p><p><strong>Study design: </strong>For this study, validation of target molecules by different experimental approaches and employing two different in vivo experiments were tried to improve the immunopharmacological value of Rhaphidophora peepla.</p><p><strong>Purpose: </strong>Our goal is to elucidate the mechanism through which the ethanol extract of Rhaphidophora peepla (Rp-EE) demonstrates anti-inflammatory properties, both in vivo and in vitro.</p><p><strong>Method: </strong>Rp-EE was phytochemically analyzed with gas chromatography-mass spectrometry (GC-MS) and high-performance liquid chromatography (HPLC). Bioinformatic analysis with protein-protein interaction (PPI) networks and Kyoto Encyclopedia of Genes and Genomes (KEGG), nitric oxide (NO) assay, MTT assay, RT-PCR, ELISA, luciferase assay, CETSA, hematoxylin and eosin (H&E) staining, and Western blotting analysis were used to evaluate anti-inflammatory activity of Rp-EE and its mechanism.</p><p><strong>Results: </strong>Rp-EE significantly reduced inflammatory responses including nitric oxide (NO) release induced by lipopolysaccharide (LPS) at the non-cytotoxic concentrations in vitro, and HCl/EtOH-induced gastritis and LPS-induced acute lung injury models in vivo. Mechanistically, it was revealed that Rp-EE can specifically target spleen tyrosine kinase (Syk) and transforming growth factor β-activated kinase 1 (TAK1) to suppress the phosphorylation levels of nuclear factor (NF)-κB subunits (p65 and p50) and activator protein (AP)-1 subunits (c-Jun and c-Fos).</p><p><strong>Conclusion: </strong>Rp-EE can inhibit inflammatory reactions managed by Syk and TAK1, resulting in suppressing the Syk/AKT/NF-κB and TAK1/MAPK/AP-1 signaling pathways. These findings lead us to a possibility that Rp-EE can be developed as a promising anti-gastric ulcer and anti-lung injury remedy.</p>","PeriodicalId":20212,"journal":{"name":"Phytomedicine","volume":"136 ","pages":"156339"},"PeriodicalIF":6.7,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142984639","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}