Pub Date : 2024-09-19DOI: 10.2174/0113862073317255240902075511
Ming Ruan, Gaohong Lv, Xueqing Wang, Fengjiao Deng, Tianya Xia, Bin Yu, Shengjin Liu
Background: Ligusticum striatum DC. (LDC) is often prescribed for Cerebral Ischemia (CI) and is commonly combined with Borneolum (BO) to enhance therapeutic outcomes. However, its specific active ingredients and underlying mechanisms remain unclear.
Objective: This study aimed to identify the active ingredients and mechanisms of LDC and BO combination therapy against CI using network pharmacology, molecular docking, and in vivo experiments.
Methods: Potential active ingredients and targets were sourced from relevant databases, and a drug-component-target-disease network was constructed to pinpoint key ingredients. Subsequently, a protein-protein interaction analysis was conducted to confirm the key targets. Following enrichment analyses of Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG), molecular docking was employed to evaluate binding energies. Finally, the therapeutic effects and mechanisms of the combination against CI were validated through in vivo experiments using male ICR mice.
Results: Venn analysis identified a total of 41 components and 292 potential targets. The drugcomponent-target-disease network revealed that the key components in LDC were palmitic acid, tetramethylpyrazine, and (Z)-ligustilide, while those in BO were (+)-borneol, β-elemene, and (-)- borneol. The PPI analysis highlighted seven crucial targets. Docking results confirmed a stable affinity between these components and their targets. KEGG enrichment analysis indicated that the mechanism involved the PI3K/AKT signaling pathway. Subsequently, in vivo experiments confirmed that the combination ameliorated abnormal hippocampus morphology and reduced the release of inflammatory factors through the activation of the PI3K/AKT signaling pathway.
Conclusion: The combination of LDC and BO markedly improved CI and inhibited inflammation response via activating the PI3K/AKT pathway.
背景介绍Ligusticum striatum DC.(LDC) 是治疗脑缺血(CI)的常用处方药,通常与婆婆纳(Borneolum,BO)联合使用以提高治疗效果。然而,其具体的活性成分和潜在机制仍不清楚:本研究旨在通过网络药理学、分子对接和体内实验,确定 LDC 与婆婆纳联合治疗 CI 的活性成分和机制:方法:从相关数据库中寻找潜在的活性成分和靶点,构建药物-成分-靶点-疾病网络,找出关键成分。随后,进行了蛋白质-蛋白质相互作用分析,以确认关键靶点。在对基因本体(GO)和京都基因和基因组百科全书(KEGG)进行富集分析后,采用分子对接来评估结合能。最后,通过使用雄性 ICR 小鼠进行体内实验,验证了该组合对 CI 的治疗效果和机制:结果:维恩分析共发现了 41 种成分和 292 个潜在靶点。药物-成分-靶点-疾病网络显示,LDC 的关键成分是棕榈酸、四甲基吡嗪和 (Z)-ligustilide; BO 的关键成分是 (+)-borneol, β-榄香烯和 (-)-borneol.PPI 分析突出了七个关键靶标。对接结果证实,这些成分与其靶标之间具有稳定的亲和力。KEGG 富集分析表明,其机制涉及 PI3K/AKT 信号通路。随后的体内实验证实,通过激活 PI3K/AKT 信号通路,该组合能改善异常海马形态并减少炎症因子的释放:结论:LDC和BO的组合能显著改善CI,并通过激活PI3K/AKT通路抑制炎症反应。
{"title":"Network Pharmacology and Validation of the Combinative Therapy of Ligusticum striatum DC. and Borneolum against Cerebral Ischemia.","authors":"Ming Ruan, Gaohong Lv, Xueqing Wang, Fengjiao Deng, Tianya Xia, Bin Yu, Shengjin Liu","doi":"10.2174/0113862073317255240902075511","DOIUrl":"https://doi.org/10.2174/0113862073317255240902075511","url":null,"abstract":"<p><strong>Background: </strong>Ligusticum striatum DC. (LDC) is often prescribed for Cerebral Ischemia (CI) and is commonly combined with Borneolum (BO) to enhance therapeutic outcomes. However, its specific active ingredients and underlying mechanisms remain unclear.</p><p><strong>Objective: </strong>This study aimed to identify the active ingredients and mechanisms of LDC and BO combination therapy against CI using network pharmacology, molecular docking, and in vivo experiments.</p><p><strong>Methods: </strong>Potential active ingredients and targets were sourced from relevant databases, and a drug-component-target-disease network was constructed to pinpoint key ingredients. Subsequently, a protein-protein interaction analysis was conducted to confirm the key targets. Following enrichment analyses of Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG), molecular docking was employed to evaluate binding energies. Finally, the therapeutic effects and mechanisms of the combination against CI were validated through in vivo experiments using male ICR mice.</p><p><strong>Results: </strong>Venn analysis identified a total of 41 components and 292 potential targets. The drugcomponent-target-disease network revealed that the key components in LDC were palmitic acid, tetramethylpyrazine, and (Z)-ligustilide, while those in BO were (+)-borneol, β-elemene, and (-)- borneol. The PPI analysis highlighted seven crucial targets. Docking results confirmed a stable affinity between these components and their targets. KEGG enrichment analysis indicated that the mechanism involved the PI3K/AKT signaling pathway. Subsequently, in vivo experiments confirmed that the combination ameliorated abnormal hippocampus morphology and reduced the release of inflammatory factors through the activation of the PI3K/AKT signaling pathway.</p><p><strong>Conclusion: </strong>The combination of LDC and BO markedly improved CI and inhibited inflammation response via activating the PI3K/AKT pathway.</p>","PeriodicalId":10491,"journal":{"name":"Combinatorial chemistry & high throughput screening","volume":null,"pages":null},"PeriodicalIF":1.6,"publicationDate":"2024-09-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142281496","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Most cancers have become immune to normal cancer therapy, like chemotherapy and radiation. Therefore, exploring more effective and economical treatment options is important. Plants and herbs contain substances called phytochemicals, which have biological effects. Many phytochemicals having antioxidant and anticancer properties have been studied previously. There is increasing evidence that phytochemicals' anti-carcinogenic benefits originate from their ability to inhibit oxidation, inflammation, cell proliferation, and angiogenesis. These phytochemicals inhibit the spread of cancer by controlling the cell cycle and other molecular processes, such as metastasis. Along with therapeutic potential, other advantages, like their abundance, greater tolerability, and economic use, increase their utility in cancer therapeutics. In recent years, a number of scientists have examined lycophytes and ferns for their potential medicinal and phytochemical properties. This analysis emphasizes the significance of chemicals obtained from ferns and their derivatives in therapeutics. The authors discuss the pteridophyte's anti-cancer properties and other medical uses in this article. This information may help researchers in further research related to the most promising anticancer phytochemicals and their possibility as alternative drugs against cancer.
{"title":"A Thorough Review on Ethnomedicinal Value of Bioactive Compounds of Pteridophytes against Cancer: Clinical Applications and Future Prospects.","authors":"Priya Bansal, Neeraj Kumar, Sharda Sambhakar, Abhishek Kumar, Deepti Katiyar","doi":"10.2174/0113862073318080240902080232","DOIUrl":"https://doi.org/10.2174/0113862073318080240902080232","url":null,"abstract":"<p><p>Most cancers have become immune to normal cancer therapy, like chemotherapy and radiation. Therefore, exploring more effective and economical treatment options is important. Plants and herbs contain substances called phytochemicals, which have biological effects. Many phytochemicals having antioxidant and anticancer properties have been studied previously. There is increasing evidence that phytochemicals' anti-carcinogenic benefits originate from their ability to inhibit oxidation, inflammation, cell proliferation, and angiogenesis. These phytochemicals inhibit the spread of cancer by controlling the cell cycle and other molecular processes, such as metastasis. Along with therapeutic potential, other advantages, like their abundance, greater tolerability, and economic use, increase their utility in cancer therapeutics. In recent years, a number of scientists have examined lycophytes and ferns for their potential medicinal and phytochemical properties. This analysis emphasizes the significance of chemicals obtained from ferns and their derivatives in therapeutics. The authors discuss the pteridophyte's anti-cancer properties and other medical uses in this article. This information may help researchers in further research related to the most promising anticancer phytochemicals and their possibility as alternative drugs against cancer.</p>","PeriodicalId":10491,"journal":{"name":"Combinatorial chemistry & high throughput screening","volume":null,"pages":null},"PeriodicalIF":1.6,"publicationDate":"2024-09-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142281494","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Aim: The aim of this study was to elucidate the mechanism of action of Shenbao tablets using metabolomics approach. Background: Kidney-Yang deficiency is a common syndrome type in traditional Chinese Medicine (TCM) syndrome typology, closely related to disorders of multiple metabolic pathways and is the root cause and underlying syndrome type of many diseases. Shenbao tablets can significantly improve the main symptoms of kidney yang deficiency syndrome, but the mechanism of action of Shenbao tablets on kidney yang deficiency syndrome is still unknown. Methods: The rats were intraperitoneally injected with hydrocortisone once a day for 40 days to simulate the syndrome. Traditional pharmacodynamic indicators (body mass, biochemical indicators and pathology) were used to evaluate the efficacy of the medicine. Serum, urine and feces were collected from rats. UPLC/MS metabolomics method was used to study the overall metabolic profile of serum, while GC/MS metabolomics method was used to study the metabolic spectrum of urine and feces. Results: Results showed that the syndrome was significantly improved in the treatment group, and obvious metabolic disorders were observed in rats with the syndrome, with 47 potential biomarkers identified. Pathway analysis showed that nicotinate and nicotinamide metabolism, glycine, serine and trione metabolism, aminoacyl tRNA biosynthesis, glycoxylate and dicarboxylate metabolism were the major ways for Shenbao tablet to improve kidney-yang deficiency syndrome. Conclusion: The mechanism of action of Shenbao tablet in improving the syndrome involves the regulation of energy metabolism, amino acid metabolism, bile acid metabolism, fatty acid metabolism and intestinal microorganisms. This work shows that metabolomics is a promising tool for studying the essence of syndrome theory in TCM and the mechanisms of TCM.
{"title":"Study on Shenbao Tablet in Treating Kidney-yang Deficiency Syndromebased on Metabolomics","authors":"Qing-gang Zhou, Bao-xia Han, Huan Yi, Zhao Geng, Xiao-jun Gou","doi":"10.2174/0113862073316238240827110846","DOIUrl":"https://doi.org/10.2174/0113862073316238240827110846","url":null,"abstract":"Aim: The aim of this study was to elucidate the mechanism of action of Shenbao tablets using metabolomics approach. Background: Kidney-Yang deficiency is a common syndrome type in traditional Chinese Medicine (TCM) syndrome typology, closely related to disorders of multiple metabolic pathways and is the root cause and underlying syndrome type of many diseases. Shenbao tablets can significantly improve the main symptoms of kidney yang deficiency syndrome, but the mechanism of action of Shenbao tablets on kidney yang deficiency syndrome is still unknown. Methods: The rats were intraperitoneally injected with hydrocortisone once a day for 40 days to simulate the syndrome. Traditional pharmacodynamic indicators (body mass, biochemical indicators and pathology) were used to evaluate the efficacy of the medicine. Serum, urine and feces were collected from rats. UPLC/MS metabolomics method was used to study the overall metabolic profile of serum, while GC/MS metabolomics method was used to study the metabolic spectrum of urine and feces. Results: Results showed that the syndrome was significantly improved in the treatment group, and obvious metabolic disorders were observed in rats with the syndrome, with 47 potential biomarkers identified. Pathway analysis showed that nicotinate and nicotinamide metabolism, glycine, serine and trione metabolism, aminoacyl tRNA biosynthesis, glycoxylate and dicarboxylate metabolism were the major ways for Shenbao tablet to improve kidney-yang deficiency syndrome. Conclusion: The mechanism of action of Shenbao tablet in improving the syndrome involves the regulation of energy metabolism, amino acid metabolism, bile acid metabolism, fatty acid metabolism and intestinal microorganisms. This work shows that metabolomics is a promising tool for studying the essence of syndrome theory in TCM and the mechanisms of TCM.","PeriodicalId":10491,"journal":{"name":"Combinatorial chemistry & high throughput screening","volume":null,"pages":null},"PeriodicalIF":1.8,"publicationDate":"2024-09-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142258712","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-09-18DOI: 10.2174/0113862073326896240901110932
Yanfei Niu, Yanxiang Yuan, Tong Wang, Ruiying Yuan, Min Zhang, Sicen Wang, Dikye Tsering, Shan Huang, Bin Li
Background: Rheumatoid Arthritis (RA) is a chronic autoimmune disease with a complex etiology. Siweixizangmaoru Decoction (SXD) has been used to treat RA in Tibet for a long history as a classic Tibetan medicine formula. However, the potential pharmacological mechanism has not been elucidated yet. Aims: The aim of this study was to evaluate the efficacy and mechanism of action of SXD in the treatment of RA using network pharmacology and molecular docking analysis. Method: Network pharmacology was employed to identify the potential bioactive components and key targets of SXD for the treatment of RA. Molecular docking of key targets and potential compounds was conducted. High-performance liquid chromatography was performed to validate the predicted active components of SXD. We established a rat model of RA and evaluated the histopathology of each group of rats. In addition, the levels of inflammatory factors in serum and the expression levels of PI3K/AKT and MAPK pathway-related proteins in synovial tissue were detected. Results: The results of network pharmacological analyses indicated that apigenin, rhamnolipids, kaempferol, quercetin, and naringenin are potential bioactive components of SXD for the treatment of rheumatoid arthritis and that their therapeutic effects may be related to the PI3K-Akt and MAPK pathways. The results of in vivo experiments show that SXD improved the arthritis index, significantly reduced joint swelling, and improved synovial inflammation and cartilage destruction. Conclusion: Network pharmacology, along with experimental validation, provided a useful approach for understanding the pharmacological mechanism of Siweixizangmaoru decoction in RA.
背景:类风湿性关节炎(RA)是一种病因复杂的慢性自身免疫性疾病:类风湿关节炎(RA)是一种病因复杂的慢性自身免疫性疾病。在西藏,四味藏药煎剂(SXD)作为经典藏药配方用于治疗类风湿关节炎已有悠久的历史。然而,其潜在的药理机制尚未阐明。目的:本研究旨在利用网络药理学和分子对接分析评估SXD治疗RA的疗效和作用机制。研究方法:采用网络药理学方法确定 SXD 的作用机制:采用网络药理学方法确定SXD治疗RA的潜在生物活性成分和关键靶点。对关键靶点和潜在化合物进行分子对接。采用高效液相色谱法验证预测的 SXD 活性成分。我们建立了一个 RA 大鼠模型,并对每组大鼠的组织病理学进行了评估。此外,还检测了血清中炎症因子的水平以及滑膜组织中 PI3K/AKT 和 MAPK 通路相关蛋白的表达水平。结果网络药理学分析结果表明,芹菜素、鼠李糖脂、山柰醇、槲皮素和柚皮素是 SXD 中潜在的生物活性成分,可用于治疗类风湿性关节炎,其治疗效果可能与 PI3K-Akt 和 MAPK 通路有关。体内实验结果表明,SXD 可改善关节炎指数,显著减轻关节肿胀,改善滑膜炎症和软骨破坏。结论网络药理学以及实验验证为了解四味藏药煎剂治疗 RA 的药理机制提供了一种有用的方法。
{"title":"Investigation of the Mechanism of Siweixizangmaoru Decoction in Improving CIA-Induced Arthritis in Rats Based on Network Pharmacology and Experimental Verification","authors":"Yanfei Niu, Yanxiang Yuan, Tong Wang, Ruiying Yuan, Min Zhang, Sicen Wang, Dikye Tsering, Shan Huang, Bin Li","doi":"10.2174/0113862073326896240901110932","DOIUrl":"https://doi.org/10.2174/0113862073326896240901110932","url":null,"abstract":"Background: Rheumatoid Arthritis (RA) is a chronic autoimmune disease with a complex etiology. Siweixizangmaoru Decoction (SXD) has been used to treat RA in Tibet for a long history as a classic Tibetan medicine formula. However, the potential pharmacological mechanism has not been elucidated yet. Aims: The aim of this study was to evaluate the efficacy and mechanism of action of SXD in the treatment of RA using network pharmacology and molecular docking analysis. Method: Network pharmacology was employed to identify the potential bioactive components and key targets of SXD for the treatment of RA. Molecular docking of key targets and potential compounds was conducted. High-performance liquid chromatography was performed to validate the predicted active components of SXD. We established a rat model of RA and evaluated the histopathology of each group of rats. In addition, the levels of inflammatory factors in serum and the expression levels of PI3K/AKT and MAPK pathway-related proteins in synovial tissue were detected. Results: The results of network pharmacological analyses indicated that apigenin, rhamnolipids, kaempferol, quercetin, and naringenin are potential bioactive components of SXD for the treatment of rheumatoid arthritis and that their therapeutic effects may be related to the PI3K-Akt and MAPK pathways. The results of in vivo experiments show that SXD improved the arthritis index, significantly reduced joint swelling, and improved synovial inflammation and cartilage destruction. Conclusion: Network pharmacology, along with experimental validation, provided a useful approach for understanding the pharmacological mechanism of Siweixizangmaoru decoction in RA.","PeriodicalId":10491,"journal":{"name":"Combinatorial chemistry & high throughput screening","volume":null,"pages":null},"PeriodicalIF":1.8,"publicationDate":"2024-09-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142258709","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Aim: This study aimed to investigate the anti-inflammatory effect and mechanism of Sophora alopecuroides L. (KDZ) on lipoteichoic acid (LTA)-induced inflammation in Bovine Mammary Epithelial Cells (BMEC). Method: The KDZ active ingredient database was established by using ultra-high-performance liquid chromatography quadrupole time-of-flight mass spectrometry (UPLC-QTOF-MS) to detect the chemical components of KDZ and combine it with the TCMSP database. Furthermore, potential targets of KDZ active ingredients were collected through the UniProt database, and mastitis-related targets were screened through the OMIM, Genecard, and DisGeNET databases. Furthermore, common targets were identified between ingredient targets and disease targets, and protein-protein interaction analysis was performed on them using the STRING platform. Furthermore, the protein interaction network was constructed using Cytoscape software. Core targets were screened through network topology analysis. On this basis, GO and KEGG enrichment analyses were performed on the common target, and molecular simulation docking analysis was conducted on the main active ingredients and core targets. Finally, the accuracy of the network analysis results was validated using in vitro cell experiments. Result: The results of UPLC-QTOF-MS detection and network pharmacology analysis showed that KDZ could intervene in signaling pathways, such as the IL-17 signaling pathway, TNF signaling pathway, MAPK signaling pathway, etc., by acting on 80 common targets through 15 potential active ingredients, thereby regulating biological processes, such as positive regulation of peptidyl serine physiology, apoptotic process, and inflammatory response, to treat mastitis. Besides, molecular simulation docking analysis also showed that the main active ingredients in KDZ, such as quercetin, matrine, calycosin, etc., can form stable bindings with 11 core targets (TNF-α, IL-6, IL-1β, etc.) through hydrogen bonding. Further in vitro validation experiments confirmed that KDZ intervention could inhibit the IL-17 signaling pathway by inhibiting the expression of GSK3β and subsequently inhibiting the production of downstream inflammatory cytokines IL-8, IL-6, IL-1β, and TNF-α, thereby alleviating LTA-induced BMEC inflammatory damage. Conclusion: KDZ can alleviate LTA-induced BMEC inflammatory damage by inhibiting the IL- 17 signaling pathway. This study can provide a scientific basis for the clinical application of KDZ and lay the foundation for the development of new therapeutic drugs for mastitis.
{"title":"Exploring the Potential Role of Sophora alopecuroides L. in Inflammation of Bovine Mammary Epithelial Cells Induced by Lipoteichoic Acid Based on Network Pharmacology and Experimental Validation","authors":"Ziwen Yuan, Fang Li, Wenfei Zhang, Yanming Wei, Yongli Hua","doi":"10.2174/0113862073313036240829070704","DOIUrl":"https://doi.org/10.2174/0113862073313036240829070704","url":null,"abstract":"Aim: This study aimed to investigate the anti-inflammatory effect and mechanism of Sophora alopecuroides L. (KDZ) on lipoteichoic acid (LTA)-induced inflammation in Bovine Mammary Epithelial Cells (BMEC). Method: The KDZ active ingredient database was established by using ultra-high-performance liquid chromatography quadrupole time-of-flight mass spectrometry (UPLC-QTOF-MS) to detect the chemical components of KDZ and combine it with the TCMSP database. Furthermore, potential targets of KDZ active ingredients were collected through the UniProt database, and mastitis-related targets were screened through the OMIM, Genecard, and DisGeNET databases. Furthermore, common targets were identified between ingredient targets and disease targets, and protein-protein interaction analysis was performed on them using the STRING platform. Furthermore, the protein interaction network was constructed using Cytoscape software. Core targets were screened through network topology analysis. On this basis, GO and KEGG enrichment analyses were performed on the common target, and molecular simulation docking analysis was conducted on the main active ingredients and core targets. Finally, the accuracy of the network analysis results was validated using in vitro cell experiments. Result: The results of UPLC-QTOF-MS detection and network pharmacology analysis showed that KDZ could intervene in signaling pathways, such as the IL-17 signaling pathway, TNF signaling pathway, MAPK signaling pathway, etc., by acting on 80 common targets through 15 potential active ingredients, thereby regulating biological processes, such as positive regulation of peptidyl serine physiology, apoptotic process, and inflammatory response, to treat mastitis. Besides, molecular simulation docking analysis also showed that the main active ingredients in KDZ, such as quercetin, matrine, calycosin, etc., can form stable bindings with 11 core targets (TNF-α, IL-6, IL-1β, etc.) through hydrogen bonding. Further in vitro validation experiments confirmed that KDZ intervention could inhibit the IL-17 signaling pathway by inhibiting the expression of GSK3β and subsequently inhibiting the production of downstream inflammatory cytokines IL-8, IL-6, IL-1β, and TNF-α, thereby alleviating LTA-induced BMEC inflammatory damage. Conclusion: KDZ can alleviate LTA-induced BMEC inflammatory damage by inhibiting the IL- 17 signaling pathway. This study can provide a scientific basis for the clinical application of KDZ and lay the foundation for the development of new therapeutic drugs for mastitis.","PeriodicalId":10491,"journal":{"name":"Combinatorial chemistry & high throughput screening","volume":null,"pages":null},"PeriodicalIF":1.8,"publicationDate":"2024-09-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142258714","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-09-18DOI: 10.2174/0113862073314265240828170126
Xiangpan Kong, Dawei He, Quan Wang
Objective: This study aimed to investigate the differential expression of mitophagyrelated genes in osteosarcoma patients with distinct prognostic outcomes and explore potential molecular regulatory mechanisms.
Methods: We analyzed microarray data from metastatic and nonmetastatic osteosarcoma patients using the UCSC dataset. Differential gene screening and intersection of mitophagy-related genes were performed using NetworkAnalyst. Random forest and LASSO regression were employed to screen selected genes and establish a risk prediction model. Functional enrichment analysis, protein- protein interaction (PPI) networks, immunoassays, and in vitro experiments were conducted to validate the findings.
Results: Seven differentially expressed genes were identified, and a robust risk prediction model was developed (AUC=0.886). PPI and functional enrichment analyses provided insights into relevant molecules and regulatory pathways. The immunoassay results revealed differences in the immune environment between the metastatic and nonmetastatic groups. Immunohistochemistry demonstrated significant downregulation of EPHA3 expression in the metastatic group, and in vitro experiments indicated that inhibiting EPHA3 increased the proliferative activity and migration ability of osteosarcoma cells.
Conclusion: Our study suggests that the downregulation of EPHA3 may contribute to mitochondrial autophagy dysfunction, thereby increasing the risk of osteosarcoma metastasis.
{"title":"Exploring Mitochondrial Autophagy Dysregulation in Osteosarcoma: Its Implications for Prognosis and Targeted Therapy.","authors":"Xiangpan Kong, Dawei He, Quan Wang","doi":"10.2174/0113862073314265240828170126","DOIUrl":"https://doi.org/10.2174/0113862073314265240828170126","url":null,"abstract":"<p><strong>Objective: </strong>This study aimed to investigate the differential expression of mitophagyrelated genes in osteosarcoma patients with distinct prognostic outcomes and explore potential molecular regulatory mechanisms.</p><p><strong>Methods: </strong>We analyzed microarray data from metastatic and nonmetastatic osteosarcoma patients using the UCSC dataset. Differential gene screening and intersection of mitophagy-related genes were performed using NetworkAnalyst. Random forest and LASSO regression were employed to screen selected genes and establish a risk prediction model. Functional enrichment analysis, protein- protein interaction (PPI) networks, immunoassays, and in vitro experiments were conducted to validate the findings.</p><p><strong>Results: </strong>Seven differentially expressed genes were identified, and a robust risk prediction model was developed (AUC=0.886). PPI and functional enrichment analyses provided insights into relevant molecules and regulatory pathways. The immunoassay results revealed differences in the immune environment between the metastatic and nonmetastatic groups. Immunohistochemistry demonstrated significant downregulation of EPHA3 expression in the metastatic group, and in vitro experiments indicated that inhibiting EPHA3 increased the proliferative activity and migration ability of osteosarcoma cells.</p><p><strong>Conclusion: </strong>Our study suggests that the downregulation of EPHA3 may contribute to mitochondrial autophagy dysfunction, thereby increasing the risk of osteosarcoma metastasis.</p>","PeriodicalId":10491,"journal":{"name":"Combinatorial chemistry & high throughput screening","volume":null,"pages":null},"PeriodicalIF":1.6,"publicationDate":"2024-09-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142281495","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-09-18DOI: 10.2174/0113862073325718240827073225
Jianghong Wang, Xiaoyuan Wang, Yanru Song, Zilin Huang, Han Wu, Liang Chang, Bingjie Huo, Guanwei Fan
Background: The Precancerous Lesion of Gastric Cancer (PLGC) is an early stage in the development of gastric cancer. The clinical application of HPXLD has been found to be effective in treating PLGC, but the mechanism of how HPXLD acts on PLGC is still unclear. Objective: The objectives of this study were to reveal the molecular mechanism of how HPXLD can be used to treat PLGC and investigate this mechanism through bioinformatics and experimental validation. Methods: PLGC-associated target genes were identified through bioinformatics analysis. A rat model of PLGC was induced using N-methyl-N'-nitro-N-nitrosoquanidine (MNNG) in combination with ranitidine, hot saline, ethanol, and intermittent fasting, with interventions by HPXLD. The pathological alterations in gastric mucosa were assessed through Hematoxylin-eosin staining (HE). Immunohistochemistry (IHC) and Western blot analyses were employed to evaluate the changes in expression levels of inflammation-related proteins. Results: After conducting bioinformatics analysis, it was found that there were 23 HPXLDPLGC crossover genes, which were significantly enriched in the IL-17 signaling pathway, TNF signaling pathway, and NF-kappa B signaling pathway. The results of HE showed that HPXLD was effective in improving gastric mucosal histopathological changes. Additionally, the IHC results demonstrated that HPXLD was able to downregulate the expression of IL-6, COX-2, MCP- 1, and MMP-9. Furthermore, Western blot analysis revealed that HPXLD was able to downregulate the expressions of IL-6, IL-17RA, ACT1, NF-κB, and TNF-α. Conclusion: HPXLD has been shown to improve PLGC by reducing the expression of inflammation- related proteins. This suggests that HPXLD may potentially be a treatment option for PLGC.
{"title":"Huopuxialing Decoction: A Promising Candidate for Precancerous Lesions of Gastric Cancer Treatment Based on Bioinformatics and Experimental Verification","authors":"Jianghong Wang, Xiaoyuan Wang, Yanru Song, Zilin Huang, Han Wu, Liang Chang, Bingjie Huo, Guanwei Fan","doi":"10.2174/0113862073325718240827073225","DOIUrl":"https://doi.org/10.2174/0113862073325718240827073225","url":null,"abstract":"Background: The Precancerous Lesion of Gastric Cancer (PLGC) is an early stage in the development of gastric cancer. The clinical application of HPXLD has been found to be effective in treating PLGC, but the mechanism of how HPXLD acts on PLGC is still unclear. Objective: The objectives of this study were to reveal the molecular mechanism of how HPXLD can be used to treat PLGC and investigate this mechanism through bioinformatics and experimental validation. Methods: PLGC-associated target genes were identified through bioinformatics analysis. A rat model of PLGC was induced using N-methyl-N'-nitro-N-nitrosoquanidine (MNNG) in combination with ranitidine, hot saline, ethanol, and intermittent fasting, with interventions by HPXLD. The pathological alterations in gastric mucosa were assessed through Hematoxylin-eosin staining (HE). Immunohistochemistry (IHC) and Western blot analyses were employed to evaluate the changes in expression levels of inflammation-related proteins. Results: After conducting bioinformatics analysis, it was found that there were 23 HPXLDPLGC crossover genes, which were significantly enriched in the IL-17 signaling pathway, TNF signaling pathway, and NF-kappa B signaling pathway. The results of HE showed that HPXLD was effective in improving gastric mucosal histopathological changes. Additionally, the IHC results demonstrated that HPXLD was able to downregulate the expression of IL-6, COX-2, MCP- 1, and MMP-9. Furthermore, Western blot analysis revealed that HPXLD was able to downregulate the expressions of IL-6, IL-17RA, ACT1, NF-κB, and TNF-α. Conclusion: HPXLD has been shown to improve PLGC by reducing the expression of inflammation- related proteins. This suggests that HPXLD may potentially be a treatment option for PLGC.","PeriodicalId":10491,"journal":{"name":"Combinatorial chemistry & high throughput screening","volume":null,"pages":null},"PeriodicalIF":1.8,"publicationDate":"2024-09-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142258710","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background: Verbascoside, a compound classified as a phenylethanol glycoside in Dihuang, has been the subject of modern pharmacological investigations. These studies have revealed its noteworthy antioxidant, anti-inflammatory, memory-enhancing, neuroprotective, antitumor, and various other pharmacological properties. While verbascoside exhibits favorable antioxidant effects, its precise mechanism of action in ameliorating osteoporosis through the treatment of oxidative stress remains unclear.
Methods: This study employed CCK8, ALP, ELISA, and ROS staining techniques to examine the osteoporotic effects of verbascoside on zebrafish and MC3T3-E1 cells. Additionally, this study aimed to investigate the molecular mechanism by which verbascoside improves osteoporosis by mitigating oxidative stress. To identify the common targets of verbascoside in relation to oxidative stress and osteoporosis, network pharmacology and molecular dynamics simulation were employed. The construction of the verbascoside - oxidative stress - osteoporosis - potential target gene network aimed to identify the core targets, while the mechanism of action was elucidated through KEGG analysis, and the accuracy was confirmed by assessing the mRNA expression of the targets.
Results: In vivo experiments demonstrated that verbascoside exhibited therapeutic effects on osteoporosis and reduced ROS production in zebrafish. In vitro experiments further revealed that verbascoside enhanced the proliferation and differentiation of MC3T3-E1 cells, thereby improving the oxidative stress status of osteoblasts. Thirteen core targets and estrogen signaling pathways were identified through the application of network pharmacology. The pivotal role of the estrogen signaling pathway in facilitating the ability of verbascoside to mitigate oxidative stressinduced osteoporosis was substantiated by the modulation of target protein mRNA expression.
Conclusion: The findings underscore the considerable therapeutic potential of verbascoside in ameliorating osteoporosis through the alleviation of oxidative stress, thus establishing it as a promising compound for the treatment of this condition.
{"title":"Using a Dual-Disease Target Mapping Network Pharmacology Approach, Verbascoside Ameliorates Osteoporosis by Activating Estrogen Signaling to Alleviate Oxidative Stress.","authors":"Peitong Wu, Qingguo Lv, Shuo Wang, Xueqin Feng, Kaiyue Zhang, Chunnan Li, Yishan Li, Xiaochen Gao, Jiaming Sun","doi":"10.2174/0113862073312956240826053228","DOIUrl":"https://doi.org/10.2174/0113862073312956240826053228","url":null,"abstract":"<p><strong>Background: </strong>Verbascoside, a compound classified as a phenylethanol glycoside in Dihuang, has been the subject of modern pharmacological investigations. These studies have revealed its noteworthy antioxidant, anti-inflammatory, memory-enhancing, neuroprotective, antitumor, and various other pharmacological properties. While verbascoside exhibits favorable antioxidant effects, its precise mechanism of action in ameliorating osteoporosis through the treatment of oxidative stress remains unclear.</p><p><strong>Methods: </strong>This study employed CCK8, ALP, ELISA, and ROS staining techniques to examine the osteoporotic effects of verbascoside on zebrafish and MC3T3-E1 cells. Additionally, this study aimed to investigate the molecular mechanism by which verbascoside improves osteoporosis by mitigating oxidative stress. To identify the common targets of verbascoside in relation to oxidative stress and osteoporosis, network pharmacology and molecular dynamics simulation were employed. The construction of the verbascoside - oxidative stress - osteoporosis - potential target gene network aimed to identify the core targets, while the mechanism of action was elucidated through KEGG analysis, and the accuracy was confirmed by assessing the mRNA expression of the targets.</p><p><strong>Results: </strong>In vivo experiments demonstrated that verbascoside exhibited therapeutic effects on osteoporosis and reduced ROS production in zebrafish. In vitro experiments further revealed that verbascoside enhanced the proliferation and differentiation of MC3T3-E1 cells, thereby improving the oxidative stress status of osteoblasts. Thirteen core targets and estrogen signaling pathways were identified through the application of network pharmacology. The pivotal role of the estrogen signaling pathway in facilitating the ability of verbascoside to mitigate oxidative stressinduced osteoporosis was substantiated by the modulation of target protein mRNA expression.</p><p><strong>Conclusion: </strong>The findings underscore the considerable therapeutic potential of verbascoside in ameliorating osteoporosis through the alleviation of oxidative stress, thus establishing it as a promising compound for the treatment of this condition.</p>","PeriodicalId":10491,"journal":{"name":"Combinatorial chemistry & high throughput screening","volume":null,"pages":null},"PeriodicalIF":1.6,"publicationDate":"2024-09-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142153315","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-09-06DOI: 10.2174/0113862073344422240906051007
Pengyan Shi, Xiaoying Chen, Jiangtao Cao, Zhe Feng, Boyu Xue
Introduction/objective: The incidence of metabolic-associated fatty liver disease (MAFLD) increases annually. Modified Zexie Decoction (MZXD) can treat this disease; however, their mechanisms of action are uncertain. This study evaluated the mechanisms of MZXD against MAFLD based on network pharmacology, molecular docking, and in vivo experiments.
Methods: The main active compounds, targets and signaling pathways of MZXD against MAFLD were obtained using network pharmacological analysis. Underlying mechanisms were validated by molecular docking and in vivo assays.
Results: Forty-one active ingredients and 197 intersection targets were identified. The main active ingredients include quercetin, luteolin, isorhamnetin, 3-methylhexane, and 3β- acetoxyatractylone. The main targets were TP53, JUN, HSP90AA1, MAPK1, MAPK3, AKT1, NF-κB p65, TNF, ESR1, FOS, and IL-6. The pathway enrichment analysis indicated that MZXD was related to the IL-17, TNF, and PI3K-AKT signaling pathways. Molecular docking suggested that these active ingredients bound strongly to TNF, IL-6, and NF-κB p65, which are integral components of the TNF pathway. In the rat MAFLD model, MZXD attenuated high-fat diet( HFD)-induced liver injury and lipid accumulation, decreased the serum levels of the inflammatory mediators TNF-α, IL6, and IL-1β, and inhibited the protein expression of TNF-α, IL6, p- IKB-α and p-NF-κB p65. Furthermore, immunohistochemistry results showed that MZXD attenuated the F4/80 staining intensity of the liver compared with the model group.
Conclusion: Collectively, our results suggested that MZXD could improve MAFLD by downregulating TNF/NF-κB signaling mediated macrophage activation.
{"title":"Exploring the Mechanism of Modified Zexie Decoction Against Metabolic Associated Fatty Liver Disease Based on Network Pharmacology and Experimental Validation.","authors":"Pengyan Shi, Xiaoying Chen, Jiangtao Cao, Zhe Feng, Boyu Xue","doi":"10.2174/0113862073344422240906051007","DOIUrl":"https://doi.org/10.2174/0113862073344422240906051007","url":null,"abstract":"<p><strong>Introduction/objective: </strong>The incidence of metabolic-associated fatty liver disease (MAFLD) increases annually. Modified Zexie Decoction (MZXD) can treat this disease; however, their mechanisms of action are uncertain. This study evaluated the mechanisms of MZXD against MAFLD based on network pharmacology, molecular docking, and in vivo experiments.</p><p><strong>Methods: </strong>The main active compounds, targets and signaling pathways of MZXD against MAFLD were obtained using network pharmacological analysis. Underlying mechanisms were validated by molecular docking and in vivo assays.</p><p><strong>Results: </strong>Forty-one active ingredients and 197 intersection targets were identified. The main active ingredients include quercetin, luteolin, isorhamnetin, 3-methylhexane, and 3β- acetoxyatractylone. The main targets were TP53, JUN, HSP90AA1, MAPK1, MAPK3, AKT1, NF-κB p65, TNF, ESR1, FOS, and IL-6. The pathway enrichment analysis indicated that MZXD was related to the IL-17, TNF, and PI3K-AKT signaling pathways. Molecular docking suggested that these active ingredients bound strongly to TNF, IL-6, and NF-κB p65, which are integral components of the TNF pathway. In the rat MAFLD model, MZXD attenuated high-fat diet( HFD)-induced liver injury and lipid accumulation, decreased the serum levels of the inflammatory mediators TNF-α, IL6, and IL-1β, and inhibited the protein expression of TNF-α, IL6, p- IKB-α and p-NF-κB p65. Furthermore, immunohistochemistry results showed that MZXD attenuated the F4/80 staining intensity of the liver compared with the model group.</p><p><strong>Conclusion: </strong>Collectively, our results suggested that MZXD could improve MAFLD by downregulating TNF/NF-κB signaling mediated macrophage activation.</p>","PeriodicalId":10491,"journal":{"name":"Combinatorial chemistry & high throughput screening","volume":null,"pages":null},"PeriodicalIF":1.6,"publicationDate":"2024-09-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142153314","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-09-03DOI: 10.2174/0113862073291189240819115228
Yuanxia Yang, Miaoting Yang, Lijuan Wang, Liufang He, Tao Shi
Background: The establishment and validation of methods for testing biological samples are crucial steps in pharmacokinetic studies. Currently, several methodological reports have been published on the detection of rapamycin plasma concentrations.
Objective: The objective of this study was to explore an effective method for detecting rapamycin in rat whole blood biological samples.
Method: In this study, we designed a rapid, sensitive, and specific liquid chromatograph-mass spectrometer/mass spectrometer (LC-MS/MS) methodology for detecting rapamycin in rat whole blood biological samples. We comprehensively validated the specificity, linear range, lower limit of quantification (LLOQ), precision, accuracy, recovery, and stability of this method.
Results: The findings of this study confirmed the successful implementation of LC-MS/MS for the detection of rapamycin, demonstrating its sensitivity, specificity, and reliability in quantitative analysis. This method ensures the accuracy and reliability of subsequent study data through our validated LC-MS/MS approach.
Conclusion: The results demonstrated the successful implementation of an LC-MS/MS method for sensitive, specific, and reliable quantitative analysis of rapamycin in rat whole blood samples. This method ensures the accuracy and reliability of subsequent study data.
Significance: The importance of this study lies in the successful establishment of a rapid, sensitive, and specific LC-MS/MS method for detecting rapamycin concentration in rat whole blood, ensuring the accuracy and reliability of subsequent research data. This provides a crucial tool and foundation for further understanding the metabolism and pharmacological effects of rapamycin in vivo, aiding in the advancement of drug research and clinical applications in related fields.
{"title":"Establishment of LC-MS/MS Methodology for the Determination of Rapamycin Concentration in Rat Whole Blood.","authors":"Yuanxia Yang, Miaoting Yang, Lijuan Wang, Liufang He, Tao Shi","doi":"10.2174/0113862073291189240819115228","DOIUrl":"https://doi.org/10.2174/0113862073291189240819115228","url":null,"abstract":"<p><strong>Background: </strong>The establishment and validation of methods for testing biological samples are crucial steps in pharmacokinetic studies. Currently, several methodological reports have been published on the detection of rapamycin plasma concentrations.</p><p><strong>Objective: </strong>The objective of this study was to explore an effective method for detecting rapamycin in rat whole blood biological samples.</p><p><strong>Method: </strong>In this study, we designed a rapid, sensitive, and specific liquid chromatograph-mass spectrometer/mass spectrometer (LC-MS/MS) methodology for detecting rapamycin in rat whole blood biological samples. We comprehensively validated the specificity, linear range, lower limit of quantification (LLOQ), precision, accuracy, recovery, and stability of this method.</p><p><strong>Results: </strong>The findings of this study confirmed the successful implementation of LC-MS/MS for the detection of rapamycin, demonstrating its sensitivity, specificity, and reliability in quantitative analysis. This method ensures the accuracy and reliability of subsequent study data through our validated LC-MS/MS approach.</p><p><strong>Conclusion: </strong>The results demonstrated the successful implementation of an LC-MS/MS method for sensitive, specific, and reliable quantitative analysis of rapamycin in rat whole blood samples. This method ensures the accuracy and reliability of subsequent study data.</p><p><strong>Significance: </strong>The importance of this study lies in the successful establishment of a rapid, sensitive, and specific LC-MS/MS method for detecting rapamycin concentration in rat whole blood, ensuring the accuracy and reliability of subsequent research data. This provides a crucial tool and foundation for further understanding the metabolism and pharmacological effects of rapamycin in vivo, aiding in the advancement of drug research and clinical applications in related fields.</p>","PeriodicalId":10491,"journal":{"name":"Combinatorial chemistry & high throughput screening","volume":null,"pages":null},"PeriodicalIF":1.6,"publicationDate":"2024-09-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142125072","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}