首页 > 最新文献

Chemical Biology & Drug Design最新文献

英文 中文
Synthesis and anti-SARS-CoV-2 activity of amino acid modified cephalotaxine derivatives 氨基酸修饰的头孢他辛衍生物的合成和抗 SARS-CoV-2 活性。
IF 3 4区 医学 Q2 Chemistry Pub Date : 2024-06-10 DOI: 10.1111/cbdd.14566
Min Si, Meidi An, Zhaomin Xia, Xiaoxue Mo, Jiayu Lu, Huaizhen He, Cheng Wang

The severe acute respiratory syndrome coronavirus (SARS-CoV-2) pandemic has triggered a significant impact on global public health security, it is urgent to develop effective antiviral drugs. Previous studies have found that binding to ACE2 is a key step in the invasion of SARS-CoV-2 into host cells, so virus invasion can be inhibited by blocking ACE2, but there are few reports on this kind of specific inhibitor. Our previous study found that Harringtonine (HT) can inhibit the entry of SARS-CoV-2 spike pseudovirus into ACE2h cells, but its relatively high cytotoxicity limits its further development. Amino acid modification of the active components can increase their solubility and reduce their cytotoxicity. Therefore, in this study, seven new derivatives were synthesized by amino acid modification of its core structure Cephalotaxine. The target compounds were evaluated by cell viability assay and the SARS-CoV-2 spike pseudovirus entry assay. Compound CET-1 significantly inhibited the entry of pseudovirus into ACE2h cells and showed less cytotoxicity than HT. Molecular docking results showed that CET-1 could bind TYR83, an important residue of ACE2, just like HT. In conclusion, our study provided a novel compound with more potential activity and lower toxicity than HT on inhibiting the SARS-CoV-2 spike pseudovirus infection, which makes it possible to be a lead compound as an antiviral drug in the future.

严重急性呼吸系统综合征冠状病毒(SARS-CoV-2)大流行对全球公共卫生安全造成了重大影响,开发有效的抗病毒药物迫在眉睫。以往的研究发现,与 ACE2 结合是 SARS-CoV-2 侵入宿主细胞的关键步骤,因此可以通过阻断 ACE2 来抑制病毒入侵,但有关这类特异性抑制剂的报道很少。我们之前的研究发现,Harringtonine(HT)能抑制 SARS-CoV-2 穗状伪病毒进入 ACE2h 细胞,但其相对较高的细胞毒性限制了它的进一步发展。对活性成分进行氨基酸修饰可增加其溶解度并降低其细胞毒性。因此,本研究通过对其核心结构头孢他辛进行氨基酸修饰,合成了七种新的衍生物。通过细胞活力测定和 SARS-CoV-2 穗状伪病毒进入测定对目标化合物进行了评估。化合物 CET-1 能明显抑制伪病毒进入 ACE2h 细胞,其细胞毒性低于 HT。分子对接结果表明,CET-1 与 HT 一样能与 ACE2 的一个重要残基 TYR83 结合。总之,我们的研究提供了一种新型化合物,它在抑制 SARS-CoV-2 穗状伪病毒感染方面比 HT 具有更强的潜在活性和更低的毒性,这使它有可能成为未来抗病毒药物的先导化合物。
{"title":"Synthesis and anti-SARS-CoV-2 activity of amino acid modified cephalotaxine derivatives","authors":"Min Si,&nbsp;Meidi An,&nbsp;Zhaomin Xia,&nbsp;Xiaoxue Mo,&nbsp;Jiayu Lu,&nbsp;Huaizhen He,&nbsp;Cheng Wang","doi":"10.1111/cbdd.14566","DOIUrl":"10.1111/cbdd.14566","url":null,"abstract":"<p>The severe acute respiratory syndrome coronavirus (SARS-CoV-2) pandemic has triggered a significant impact on global public health security, it is urgent to develop effective antiviral drugs. Previous studies have found that binding to ACE2 is a key step in the invasion of SARS-CoV-2 into host cells, so virus invasion can be inhibited by blocking ACE2, but there are few reports on this kind of specific inhibitor. Our previous study found that Harringtonine (HT) can inhibit the entry of SARS-CoV-2 spike pseudovirus into ACE2<sup>h</sup> cells, but its relatively high cytotoxicity limits its further development. Amino acid modification of the active components can increase their solubility and reduce their cytotoxicity. Therefore, in this study, seven new derivatives were synthesized by amino acid modification of its core structure Cephalotaxine. The target compounds were evaluated by cell viability assay and the SARS-CoV-2 spike pseudovirus entry assay. Compound CET-1 significantly inhibited the entry of pseudovirus into ACE2<sup>h</sup> cells and showed less cytotoxicity than HT. Molecular docking results showed that CET-1 could bind TYR83, an important residue of ACE2, just like HT. In conclusion, our study provided a novel compound with more potential activity and lower toxicity than HT on inhibiting the SARS-CoV-2 spike pseudovirus infection, which makes it possible to be a lead compound as an antiviral drug in the future.</p>","PeriodicalId":143,"journal":{"name":"Chemical Biology & Drug Design","volume":null,"pages":null},"PeriodicalIF":3.0,"publicationDate":"2024-06-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141302240","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}
引用次数: 0
Study on the mechanism of quercetin in Sini Decoction Plus Ginseng Soup to inhibit liver cancer and HBV virus replication through CDK1 研究四逆汤加人参汤中的槲皮素通过 CDK1 抑制肝癌和 HBV 病毒复制的机制。
IF 3 4区 医学 Q2 Chemistry Pub Date : 2024-06-10 DOI: 10.1111/cbdd.14567
Liyuan Hao, Shenghao Li, Guo Chen, Aiyu Nie, Liang Zeng, Zhonghui Xiao, Xiaoyu Hu

Background

To explore the anti-tumor and anti-virus key active ingredients of Sini Decoction Plus Ginseng Soup (SNRS) and their mechanisms.

Methods

The main ingredients of SNRS were analyzed by network pharmacology, and quercetin was identified as the key active ingredient. Then, we obtained the targets of quercetin by using Drugbank, PharmMapper, and SwissTargetPrediction databases. Then, the targets of HBV-related hepatocellular carcinoma (HBV-related HCC) were obtained by using Genecards database. In addition, using the gene expression profiles of HBV-related HCC patients in GEO database and the genes with the greatest survival difference in GEPIA 2 database identified the potential targets of quercetin. In addition, the mechanism of potential genes was studied through GO, KEGG analysis, and PPI network. Using AUC and survival analysis to evaluate the diagnostic and prognostic value of cyclin-dependent kinase 1 (CDK1) and CCNB1. Finally, the effects of quercetin on proliferation of Hep3B and HepG2215 cells and the level of CDK1 and CCNB1 were verified in vitro. ELISA was used to measure the expression levels of hepatitis B surface antigen (HBsAg) and hepatitis B e antigen (HBeAg) after the intervention by quercetin for 24 h and 48 h in HepG2215 cell.

Results

The first 10 key ingredients of SNRS were identified, and quercetin was the most key ingredient. The 101 potential quercetin targets were identified for the treatment of HBV-related HCC. GO and KEGG showed that 101 potential target enrichment in cancer and cell cycle regulation. By Venn analysis, CDK1 and CCNB1 were intersection targets, which could be used as potential targets for the action of quercetin on HBV-related HCC. Moreover, the expression of CDK1 and CCNB1 was highly expressed in the high-risk group, while the OS rate was low. The 1-year, 3-year and 5-year area under the curve (AUC) curves of CDK1 and CCNB1 were 0.724, 0.676, 0.622 and 0.745, 0.678, 0.634, respectively. Moreover, experimental results also showed that quercetin inhibited cell proliferation and reduced CDK1 expression in Hep3B and HepG2215 cells. The expressions of HBsAg and HBeAg in HepG2215 cell supernatant and cell gradually decreased with the increase of intervention time of quercetin and CDK1 inhibitor.

Conclusions

Quercetin is a key ingredient of anti-HBV-related HCC activity and inhibits HBV replication in SNRS by inhibiting CDK1.

背景:探讨四逆汤抗肿瘤和抗病毒的主要活性成分及其作用机制:探讨四逆汤的抗肿瘤和抗病毒主要活性成分及其作用机制:方法:采用网络药理学方法对四逆汤的主要成分进行分析,确定槲皮素为四逆汤的关键活性成分。然后,我们利用 Drugbank、PharmMapper 和 SwissTargetPrediction 数据库获得了槲皮素的靶点。然后,我们利用 Genecards 数据库获得了 HBV 相关肝细胞癌(HBV 相关 HCC)的靶点。此外,利用 GEO 数据库中 HBV 相关 HCC 患者的基因表达谱和 GEPIA 2 数据库中存活率差异最大的基因,确定了槲皮素的潜在靶点。此外,还通过GO、KEGG分析和PPI网络研究了潜在基因的作用机制。利用AUC和生存分析评估细胞周期蛋白依赖性激酶1(CDK1)和CCNB1的诊断和预后价值。最后,在体外验证了槲皮素对 Hep3B 和 HepG2215 细胞增殖以及 CDK1 和 CCNB1 水平的影响。在槲皮素干预 HepG2215 细胞 24 小时和 48 小时后,用 ELISA 检测乙肝表面抗原(HBsAg)和乙肝 e 抗原(HBeAg)的表达水平:结果:确定了SNRS的前10种关键成分,其中槲皮素是最关键的成分。结果:确定了 SNRS 的前 10 种关键成分,其中槲皮素是最关键的成分;确定了 101 个治疗 HBV 相关 HCC 的潜在槲皮素靶点。GO和KEGG显示,101个潜在靶点富集于癌症和细胞周期调控领域。通过Venn分析,CDK1和CCNB1是交叉靶点,可作为槲皮素作用于HBV相关HCC的潜在靶点。此外,CDK1和CCNB1在高危组中表达量高,而OS率低。CDK1和CCNB1的1年、3年和5年曲线下面积(AUC)曲线分别为0.724、0.676、0.622和0.745、0.678、0.634。此外,实验结果还表明,槲皮素能抑制 Hep3B 和 HepG2215 细胞的增殖并降低 CDK1 的表达。随着槲皮素和 CDK1 抑制剂干预时间的延长,HepG2215 细胞上清液和细胞中 HBsAg 和 HBeAg 的表达量逐渐下降:结论:槲皮素是抗 HBV 相关 HCC 活性的关键成分,可通过抑制 CDK1 抑制 SNRS 中 HBV 的复制。
{"title":"Study on the mechanism of quercetin in Sini Decoction Plus Ginseng Soup to inhibit liver cancer and HBV virus replication through CDK1","authors":"Liyuan Hao,&nbsp;Shenghao Li,&nbsp;Guo Chen,&nbsp;Aiyu Nie,&nbsp;Liang Zeng,&nbsp;Zhonghui Xiao,&nbsp;Xiaoyu Hu","doi":"10.1111/cbdd.14567","DOIUrl":"10.1111/cbdd.14567","url":null,"abstract":"<div>\u0000 \u0000 \u0000 <section>\u0000 \u0000 <h3> Background</h3>\u0000 \u0000 <p>To explore the anti-tumor and anti-virus key active ingredients of Sini Decoction Plus Ginseng Soup (SNRS) and their mechanisms.</p>\u0000 </section>\u0000 \u0000 <section>\u0000 \u0000 <h3> Methods</h3>\u0000 \u0000 <p>The main ingredients of SNRS were analyzed by network pharmacology, and quercetin was identified as the key active ingredient. Then, we obtained the targets of quercetin by using Drugbank, PharmMapper, and SwissTargetPrediction databases. Then, the targets of HBV-related hepatocellular carcinoma (HBV-related HCC) were obtained by using Genecards database. In addition, using the gene expression profiles of HBV-related HCC patients in GEO database and the genes with the greatest survival difference in GEPIA 2 database identified the potential targets of quercetin. In addition, the mechanism of potential genes was studied through GO, KEGG analysis, and PPI network. Using AUC and survival analysis to evaluate the diagnostic and prognostic value of cyclin-dependent kinase 1 (CDK1) and CCNB1. Finally, the effects of quercetin on proliferation of Hep3B and HepG2215 cells and the level of CDK1 and CCNB1 were verified in vitro. ELISA was used to measure the expression levels of hepatitis B surface antigen (HBsAg) and hepatitis B e antigen (HBeAg) after the intervention by quercetin for 24 h and 48 h in HepG2215 cell.</p>\u0000 </section>\u0000 \u0000 <section>\u0000 \u0000 <h3> Results</h3>\u0000 \u0000 <p>The first 10 key ingredients of SNRS were identified, and quercetin was the most key ingredient. The 101 potential quercetin targets were identified for the treatment of HBV-related HCC. GO and KEGG showed that 101 potential target enrichment in cancer and cell cycle regulation. By Venn analysis, CDK1 and CCNB1 were intersection targets, which could be used as potential targets for the action of quercetin on HBV-related HCC. Moreover, the expression of CDK1 and CCNB1 was highly expressed in the high-risk group, while the OS rate was low. The 1-year, 3-year and 5-year area under the curve (AUC) curves of CDK1 and CCNB1 were 0.724, 0.676, 0.622 and 0.745, 0.678, 0.634, respectively. Moreover, experimental results also showed that quercetin inhibited cell proliferation and reduced CDK1 expression in Hep3B and HepG2215 cells. The expressions of HBsAg and HBeAg in HepG2215 cell supernatant and cell gradually decreased with the increase of intervention time of quercetin and CDK1 inhibitor.</p>\u0000 </section>\u0000 \u0000 <section>\u0000 \u0000 <h3> Conclusions</h3>\u0000 \u0000 <p>Quercetin is a key ingredient of anti-HBV-related HCC activity and inhibits HBV replication in SNRS by inhibiting CDK1.</p>\u0000 </section>\u0000 </div>","PeriodicalId":143,"journal":{"name":"Chemical Biology & Drug Design","volume":null,"pages":null},"PeriodicalIF":3.0,"publicationDate":"2024-06-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141302239","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}
引用次数: 0
Silencing PCMT1 enhances the sensitivity of breast cancer cells to paclitaxel through the PI3K/Akt/STMN1 pathway 沉默 PCMT1 可通过 PI3K/Akt/STMN1 通路提高乳腺癌细胞对紫杉醇的敏感性。
IF 3 4区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-06-09 DOI: 10.1111/cbdd.14559
Ke Zhang, Jin-You Li, Kai Li

This study aimed to investigate whether silencing Protein L-isoaspartate (D-aspartate) O-methyltransferase (PCMT1) expression can enhance the sensitivity of breast cancer cells to paclitaxel and its possible mechanism. Tumor tissues and adjacent histologically normal tissues were collected from patients with breast cancer admitted to our hospital. Human normal breast epithelial cells MCF10A, human breast cancer cells MCF-7, and paclitaxel-resistant breast cancer cells MCF-7/PR were purchased. MCF-7/PR cells were further grouped into negative control (NC) group, si-PCMT1 group (transfected with si-PCMT1), 740Y-P group (treated with 740Y-P, an activator of phosphatidylinositol 3-kinase (PI3K)/ v-Akt Murine Thymoma Viral Oncogene (AKT) signaling pathway), and si-PCMT1 + 740Y-P group (transfected with si-PCMT1 and then treated with 740Y-P). The expression level of PCMT1 in tissues and cells was detected by quantitative real-time polymerase chain reaction (qRT-PCR). Western blot analysis was used to detect the protein expression level of PCMT1 in tissues and cells as well as the protein level of p-PI3K, PI3K, p-Akt, Akt, and Stathmin1 (STMN1) in cells. 3-(4,5)-dimethylthiahiazo(-z-y1)-3,5-di-phenytetrazoliumromide (MTT) and colony formation assays were used to determine cell viability, scratch assay was used to assess the migration ability of cells, and Transwell assay was used to assess the invasion ability of cells. The expression of PCMT1 was remarkably up-regulated in breast cancer tissues and MCF-7/PR cells. Silencing PCMT1 expression significantly inhibited the proliferation, migration, and invasion of MCF-7/PR cells, and alleviated the resistance of cancer cells to paclitaxel. Additionally, silencing PCMT1 expression also inhibited the activation of PI3K/Akt/STMN1 pathway in MCF-7/PR cells, while activating PI3K/Akt/STMN1 pathway significantly reversed the effect of silencing PCMT1 expression on MCF-7/PR cells. PCMT1 is highly expressed in breast cancer tissues and MCF-7/PR cells, and silencing PCMT1 expression can not only inhibit the development of breast cancer but also enhance paclitaxel sensitivity. Its mechanism of action may be achieved by inhibiting PI3K/Akt/STMN1 signaling.

本研究旨在探讨沉默L-异天冬氨酸(D-天冬氨酸)蛋白O-甲基转移酶(PCMT1)的表达能否提高乳腺癌细胞对紫杉醇的敏感性及其可能的机制。肿瘤组织和邻近组织学正常组织均取自本院收治的乳腺癌患者。购买人正常乳腺上皮细胞 MCF10A、人乳腺癌细胞 MCF-7 和紫杉醇耐药乳腺癌细胞 MCF-7/PR。MCF-7/PR 细胞被进一步分为阴性对照(NC)组、si-PCMT1 组(转染 si-PCMT1)、740Y-P 组(用 740Y-P 处理,740Y-P 是磷脂酰肌醇 3- 激酶(PI3K)/ v-Akt 默沙东胸腺瘤病毒癌基因(AKT)信号通路的激活剂)和 si-PCMT1 + 740Y-P 组(转染 si-PCMT1,然后用 740Y-P 处理)。组织和细胞中 PCMT1 的表达水平通过实时定量聚合酶链反应(qRT-PCR)检测。Western 印迹分析用于检测组织和细胞中 PCMT1 的蛋白表达水平,以及细胞中 p-PI3K、PI3K、p-Akt、Akt 和 Stathmin1 (STMN1) 的蛋白水平。3-(4,5)-dimethylthiahiazo(-z-y1)-3,5-di-phenytetrazoliumromide (MTT) 和菌落形成试验用于测定细胞活力,划痕试验用于评估细胞的迁移能力,Transwell 试验用于评估细胞的侵袭能力。结果表明,PCMT1 在乳腺癌组织和 MCF-7/PR 细胞中明显上调。抑制 PCMT1 的表达可明显抑制 MCF-7/PR 细胞的增殖、迁移和侵袭,并减轻癌细胞对紫杉醇的耐药性。此外,沉默 PCMT1 还能抑制 MCF-7/PR 细胞中 PI3K/Akt/STMN1 通路的激活,而激活 PI3K/Akt/STMN1 通路则能明显逆转沉默 PCMT1 对 MCF-7/PR 细胞的影响。PCMT1 在乳腺癌组织和 MCF-7/PR 细胞中高表达,沉默 PCMT1 不仅能抑制乳腺癌的发展,还能提高紫杉醇的敏感性。其作用机制可能是通过抑制 PI3K/Akt/STMN1 信号转导实现的。
{"title":"Silencing PCMT1 enhances the sensitivity of breast cancer cells to paclitaxel through the PI3K/Akt/STMN1 pathway","authors":"Ke Zhang,&nbsp;Jin-You Li,&nbsp;Kai Li","doi":"10.1111/cbdd.14559","DOIUrl":"10.1111/cbdd.14559","url":null,"abstract":"<div>\u0000 \u0000 \u0000 <section>\u0000 \u0000 <p>This study aimed to investigate whether silencing Protein L-isoaspartate (D-aspartate) O-methyltransferase (PCMT1) expression can enhance the sensitivity of breast cancer cells to paclitaxel and its possible mechanism. Tumor tissues and adjacent histologically normal tissues were collected from patients with breast cancer admitted to our hospital. Human normal breast epithelial cells MCF10A, human breast cancer cells MCF-7, and paclitaxel-resistant breast cancer cells MCF-7/PR were purchased. MCF-7/PR cells were further grouped into negative control (NC) group, si-PCMT1 group (transfected with si-PCMT1), 740Y-P group (treated with 740Y-P, an activator of phosphatidylinositol 3-kinase (PI3K)/ v-Akt Murine Thymoma Viral Oncogene (AKT) signaling pathway), and si-PCMT1 + 740Y-P group (transfected with si-PCMT1 and then treated with 740Y-P). The expression level of PCMT1 in tissues and cells was detected by quantitative real-time polymerase chain reaction (qRT-PCR). Western blot analysis was used to detect the protein expression level of PCMT1 in tissues and cells as well as the protein level of p-PI3K, PI3K, p-Akt, Akt, and Stathmin1 (STMN1) in cells. 3-(4,5)-dimethylthiahiazo(-z-y1)-3,5-di-phenytetrazoliumromide (MTT) and colony formation assays were used to determine cell viability, scratch assay was used to assess the migration ability of cells, and Transwell assay was used to assess the invasion ability of cells. The expression of PCMT1 was remarkably up-regulated in breast cancer tissues and MCF-7/PR cells. Silencing PCMT1 expression significantly inhibited the proliferation, migration, and invasion of MCF-7/PR cells, and alleviated the resistance of cancer cells to paclitaxel. Additionally, silencing PCMT1 expression also inhibited the activation of PI3K/Akt/STMN1 pathway in MCF-7/PR cells, while activating PI3K/Akt/STMN1 pathway significantly reversed the effect of silencing PCMT1 expression on MCF-7/PR cells. PCMT1 is highly expressed in breast cancer tissues and MCF-7/PR cells, and silencing PCMT1 expression can not only inhibit the development of breast cancer but also enhance paclitaxel sensitivity. Its mechanism of action may be achieved by inhibiting PI3K/Akt/STMN1 signaling.</p>\u0000 </section>\u0000 </div>","PeriodicalId":143,"journal":{"name":"Chemical Biology & Drug Design","volume":null,"pages":null},"PeriodicalIF":3.0,"publicationDate":"2024-06-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141297594","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}
引用次数: 0
Exploring the cytotoxic potential of biflavones of Araucaria cunninghamii: Precise identification combined by LC-HRMS-metabolomics and database mining, targeted isolation, network pharmacology, in vitro cytotoxicity, and docking studies 探索银杏双黄酮的细胞毒性潜力:结合 LC-HRMS 代谢组学和数据库挖掘、靶向分离、网络药理学、体外细胞毒性和对接研究进行精确鉴定。
IF 3 4区 医学 Q2 Chemistry Pub Date : 2024-06-07 DOI: 10.1111/cbdd.14564
Bharat Sahu, Sanheeta Chakrabarty, Vaishali Saini, Meenakshi Kandpal, Bharat Goel, Sanju Kumari, Ijaz Ahmed, Hem Chandra Jha, Shreyans K. Jain

The leaves of Araucaria cunninghamii are known to be nonedible and toxic. Previous studies have identified biflavones in various Araucaria species. This study aimed to investigate the in vitro cytotoxicity of the isolated compounds from Araucaria cunninghamii after metabolomics and network pharmacological analysis. Methanol extract of Araucaria cunninghamii leaves was subjected to bioassay-guided fractionation. The active fraction was analyzed using LC-HRMS, through strategic database mining, by comparing the data to the Dictionary of Natural Products to identify 12 biflavones, along with abietic acid, beta-sitosterol, and phthalate. Eight compounds were screened for network pharmacology study, where in silico ADME analysis, prediction of gene targets, compound-gene-pathway network and hierarchical network analysis, protein–protein interaction, KEGG pathway, and Gene Ontology analyses were done, that showed PI3KR1, EGFR, GSK3B, and ABCB1 as the common targets for all the compounds that may act in the gastric cancer pathway. Simultaneously, four biflavones were isolated via chromatography and identified through NMR as dimeric apigenin with varying methoxy substitutions. Cytotoxicity study against the AGS cell line for gastric cancer showed that AC1 biflavone (IC50 90.58 μM) exhibits the highest cytotoxicity and monomeric apigenin (IC50 174.5 μM) the lowest. Besides, the biflavones were docked to the previously identified targets to analyze their binding affinities, and all the ligands were found to bind with energy ≤−7 Kcal/mol.

众所周知,Araucaria cunninghamii 的叶子不能食用,而且有毒。以前的研究发现了多种 Araucaria 品种中的双黄酮。本研究旨在通过代谢组学和网络药理学分析,研究从 Araucaria cunninghamii 中分离出的化合物的体外细胞毒性。研究采用生物测定指导下的分馏方法,对白芒花叶甲醇提取物进行分馏。通过战略性的数据库挖掘,将数据与《天然产品词典》进行比较,使用 LC-HRMS 对活性组分进行分析,从而鉴定出 12 种双黄酮类化合物以及阿比替酸、β-谷甾醇和邻苯二甲酸酯。筛选出的 8 种化合物进行了网络药理学研究,其中包括默观 ADME 分析、基因靶点预测、化合物-基因-通路网络和层次网络分析、蛋白质-蛋白质相互作用、KEGG 通路和基因本体分析,结果表明 PI3KR1、EGFR、GSK3B 和 ABCB1 是所有化合物的共同靶点,可能作用于胃癌通路。同时,通过色谱法分离出了四种双黄酮,并通过核磁共振鉴定出它们是具有不同甲氧基取代的二聚芹菜素。针对 AGS 胃癌细胞系的细胞毒性研究表明,AC1 双黄酮(IC50 90.58 μM)的细胞毒性最高,而单体芹菜素(IC50 174.5 μM)的细胞毒性最低。此外,还将双黄酮与先前确定的靶标进行了对接,以分析其结合亲和力,结果发现所有配体的结合能量均≤-7 Kcal/mol。
{"title":"Exploring the cytotoxic potential of biflavones of Araucaria cunninghamii: Precise identification combined by LC-HRMS-metabolomics and database mining, targeted isolation, network pharmacology, in vitro cytotoxicity, and docking studies","authors":"Bharat Sahu,&nbsp;Sanheeta Chakrabarty,&nbsp;Vaishali Saini,&nbsp;Meenakshi Kandpal,&nbsp;Bharat Goel,&nbsp;Sanju Kumari,&nbsp;Ijaz Ahmed,&nbsp;Hem Chandra Jha,&nbsp;Shreyans K. Jain","doi":"10.1111/cbdd.14564","DOIUrl":"10.1111/cbdd.14564","url":null,"abstract":"<p>The leaves of <i>Araucaria cunninghamii</i> are known to be nonedible and toxic. Previous studies have identified biflavones in various <i>Araucaria</i> species. This study aimed to investigate the in vitro cytotoxicity of the isolated compounds from <i>Araucaria cunninghamii</i> after metabolomics and network pharmacological analysis. Methanol extract of <i>Araucaria cunninghamii</i> leaves was subjected to bioassay-guided fractionation. The active fraction was analyzed using LC-HRMS, through strategic database mining, by comparing the data to the Dictionary of Natural Products to identify 12 biflavones, along with abietic acid, beta-sitosterol, and phthalate. Eight compounds were screened for network pharmacology study, where in silico ADME analysis, prediction of gene targets, compound-gene-pathway network and hierarchical network analysis, protein–protein interaction, KEGG pathway, and Gene Ontology analyses were done, that showed PI3KR1, EGFR, GSK3B, and ABCB1 as the common targets for all the compounds that may act in the gastric cancer pathway. Simultaneously, four biflavones were isolated via chromatography and identified through NMR as dimeric apigenin with varying methoxy substitutions. Cytotoxicity study against the AGS cell line for gastric cancer showed that AC1 biflavone (IC<sub>50</sub> 90.58 μM) exhibits the highest cytotoxicity and monomeric apigenin (IC<sub>50</sub> 174.5 μM) the lowest. Besides, the biflavones were docked to the previously identified targets to analyze their binding affinities, and all the ligands were found to bind with energy ≤−7 Kcal/mol.</p>","PeriodicalId":143,"journal":{"name":"Chemical Biology & Drug Design","volume":null,"pages":null},"PeriodicalIF":3.0,"publicationDate":"2024-06-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141285664","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}
引用次数: 0
Network pharmacology and experimental verification to explore the effect of Hedyotis diffusa on Alzheimer's disease 通过网络药理学和实验验证,探索白花蛇舌草对阿尔茨海默病的疗效。
IF 3 4区 医学 Q2 Chemistry Pub Date : 2024-06-03 DOI: 10.1111/cbdd.14558
JingXu Chen, JiaLi Rao, Hao Lu, Min Lu, ChengCheng Wang, Yan Cao

This study aimed to explore the active components and the effect of Hedyotis diffusa (HD) against Alzheimer's disease (AD) via network pharmacology, molecular docking, and experimental evaluations. We conducted a comprehensive screening process using the TCMSP, Swiss Target Prediction, and PharmMapper databases to identify the active components and their related targets in HD. In addition, we collected potential therapeutic targets of AD from the Gene Cards, Drugbank, and OMIM databases. Afterward, we utilized Cytoscape to establish both protein–protein interaction (PPI) networks and compound-target (C-T) networks. To gain further insights into the functional aspect, we performed GO and KEGG pathway analyses using the David database. Next, we employed Autodock vina to estimate the binding force between the components and the hub genes. To validate our network pharmacology findings, we conducted relevant experiments on Caenorhabditis elegans, further confirming the reliability of our results. Then a total of six active compounds and 149 therapeutic targets were detected. Through the analysis of the association between active compounds, therapeutic targets, and signaling pathways, it was observed that the therapeutic effect of HD primarily encompassed the inhibition of Aβ, suppression of AChE activity, and mitigating oxidative stress. Additionally, our investigation revealed that the key active compounds in HD primarily consisted of iridoids, which exhibited resistance against AD by acting on the Alzheimer's disease pathway and the AGE-RAGE signaling pathway in diabetic complications.

本研究旨在通过网络药理学、分子对接和实验评估,探索白花蛇舌草(Hedyotis diffusa,HD)的活性成分及其对阿尔茨海默病(Alzheimer's disease,AD)的作用。我们利用 TCMSP、Swiss Target Prediction 和 PharmMapper 数据库进行了全面筛选,以确定 HD 中的活性成分及其相关靶点。此外,我们还从 Gene Cards、Drugbank 和 OMIM 数据库中收集了 AD 的潜在治疗靶点。之后,我们利用 Cytoscape 建立了蛋白质-蛋白质相互作用(PPI)网络和化合物-靶标(C-T)网络。为了进一步了解功能方面的情况,我们利用 David 数据库进行了 GO 和 KEGG 通路分析。接着,我们使用 Autodock vina 估算了各组分与枢纽基因之间的结合力。为了验证我们的网络药理学发现,我们在秀丽隐杆线虫上进行了相关实验,进一步证实了我们结果的可靠性。随后,我们共发现了 6 种活性化合物和 149 个治疗靶点。通过分析活性化合物、治疗靶点和信号通路之间的关联,我们发现 HD 的治疗作用主要包括抑制 Aβ、抑制 AChE 活性和减轻氧化应激。此外,我们的研究还发现,HD 中的关键活性化合物主要由虹膜类化合物组成,它们通过作用于阿尔茨海默病通路和糖尿病并发症中的 AGE-RAGE 信号通路,表现出抗抑郁作用。
{"title":"Network pharmacology and experimental verification to explore the effect of Hedyotis diffusa on Alzheimer's disease","authors":"JingXu Chen,&nbsp;JiaLi Rao,&nbsp;Hao Lu,&nbsp;Min Lu,&nbsp;ChengCheng Wang,&nbsp;Yan Cao","doi":"10.1111/cbdd.14558","DOIUrl":"10.1111/cbdd.14558","url":null,"abstract":"<p>This study aimed to explore the active components and the effect of <i>Hedyotis diffusa</i> (HD) against Alzheimer's disease (AD) via network pharmacology, molecular docking, and experimental evaluations. We conducted a comprehensive screening process using the TCMSP, Swiss Target Prediction, and PharmMapper databases to identify the active components and their related targets in HD. In addition, we collected potential therapeutic targets of AD from the Gene Cards, Drugbank, and OMIM databases. Afterward, we utilized Cytoscape to establish both protein–protein interaction (PPI) networks and compound-target (C-T) networks. To gain further insights into the functional aspect, we performed GO and KEGG pathway analyses using the David database. Next, we employed Autodock vina to estimate the binding force between the components and the hub genes. To validate our network pharmacology findings, we conducted relevant experiments on <i>Caenorhabditis elegans</i>, further confirming the reliability of our results. Then a total of six active compounds and 149 therapeutic targets were detected. Through the analysis of the association between active compounds, therapeutic targets, and signaling pathways, it was observed that the therapeutic effect of HD primarily encompassed the inhibition of Aβ, suppression of AChE activity, and mitigating oxidative stress. Additionally, our investigation revealed that the key active compounds in HD primarily consisted of iridoids, which exhibited resistance against AD by acting on the Alzheimer's disease pathway and the AGE-RAGE signaling pathway in diabetic complications.</p>","PeriodicalId":143,"journal":{"name":"Chemical Biology & Drug Design","volume":null,"pages":null},"PeriodicalIF":3.0,"publicationDate":"2024-06-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141201662","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}
引用次数: 0
Quercetin inhibits breast cancer cell proliferation and survival by targeting Akt/mTOR/PTEN signaling pathway 槲皮素通过靶向 Akt/mTOR/PTEN 信号通路抑制乳腺癌细胞的增殖和存活。
IF 3 4区 医学 Q2 Chemistry Pub Date : 2024-06-02 DOI: 10.1111/cbdd.14557
Ji Jiang, Yan Yang, Fuhuan Wang, Wei Mao, Zhongjun Wang, Zegang Liu

Recently, natural compounds such as quercetin have gained an increasing amount of attention in treating breast cancer. However, the exact mechanisms responsible for the antiproliferative functions of quercetin are not completely understood. Therefore, we aimed to examine quercetin impacts on breast cancer cell proliferation and survival and the involvement of PI3K/Akt/mTOR pathway. Breast cancer MDA-MB-231 and MCF-7 cells were exposed to quercetin, and cell proliferation was assessed by MTT assay. ELISA was applied to evaluate cell apoptosis. The expression levels of apoptotic mediators such as caspase-3, Bcl-2, Bax and PI3K, Akt, mTOR, and PTEN were assessed via qRT-PCR and western blot. We found that quercetin suppressed dose dependently cell growth capacity in MDA-MB-231 and MCF-7 cells. In addition, quercetin treatment increase apoptosis in both cells lines via modulating the pro- and antiapoptotic markers. Quercetin upregulated PTEN and downregulated PI3K, Akt, and mTOR, hence suppressing this signaling pathway in cells. In conclusion, we showed antiproliferative and pro-apoptotic function of quercetin in breast cancer cell lines, which is mediated by targeting and suppressing PI3K/Akt/mTOR signal transduction.

最近,槲皮素等天然化合物在治疗乳腺癌方面受到越来越多的关注。然而,槲皮素抗增殖功能的确切机制尚未完全明了。因此,我们旨在研究槲皮素对乳腺癌细胞增殖和存活的影响以及 PI3K/Akt/mTOR 通路的参与。将乳腺癌 MDA-MB-231 和 MCF-7 细胞暴露于槲皮素,用 MTT 法评估细胞增殖。采用 ELISA 法评估细胞凋亡。通过 qRT-PCR 和 Western 印迹评估了细胞凋亡介质如 caspase-3、Bcl-2、Bax 和 PI3K、Akt、mTOR 和 PTEN 的表达水平。我们发现,槲皮素对MDA-MB-231和MCF-7细胞生长能力的抑制具有剂量依赖性。此外,槲皮素还能通过调节促凋亡和抗凋亡标志物增加细胞凋亡。槲皮素能上调 PTEN,下调 PI3K、Akt 和 mTOR,从而抑制细胞中的这一信号通路。总之,我们发现槲皮素在乳腺癌细胞系中具有抗增殖和促凋亡功能,而这是通过靶向和抑制PI3K/Akt/mTOR信号转导介导的。
{"title":"Quercetin inhibits breast cancer cell proliferation and survival by targeting Akt/mTOR/PTEN signaling pathway","authors":"Ji Jiang,&nbsp;Yan Yang,&nbsp;Fuhuan Wang,&nbsp;Wei Mao,&nbsp;Zhongjun Wang,&nbsp;Zegang Liu","doi":"10.1111/cbdd.14557","DOIUrl":"10.1111/cbdd.14557","url":null,"abstract":"<p>Recently, natural compounds such as quercetin have gained an increasing amount of attention in treating breast cancer. However, the exact mechanisms responsible for the antiproliferative functions of quercetin are not completely understood. Therefore, we aimed to examine quercetin impacts on breast cancer cell proliferation and survival and the involvement of PI3K/Akt/mTOR pathway. Breast cancer MDA-MB-231 and MCF-7 cells were exposed to quercetin, and cell proliferation was assessed by MTT assay. ELISA was applied to evaluate cell apoptosis. The expression levels of apoptotic mediators such as caspase-3, Bcl-2, Bax and PI3K, Akt, mTOR, and PTEN were assessed via qRT-PCR and western blot. We found that quercetin suppressed dose dependently cell growth capacity in MDA-MB-231 and MCF-7 cells. In addition, quercetin treatment increase apoptosis in both cells lines via modulating the pro- and antiapoptotic markers. Quercetin upregulated PTEN and downregulated PI3K, Akt, and mTOR, hence suppressing this signaling pathway in cells. In conclusion, we showed antiproliferative and pro-apoptotic function of quercetin in breast cancer cell lines, which is mediated by targeting and suppressing PI3K/Akt/mTOR signal transduction.</p>","PeriodicalId":143,"journal":{"name":"Chemical Biology & Drug Design","volume":null,"pages":null},"PeriodicalIF":3.0,"publicationDate":"2024-06-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141201664","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}
引用次数: 0
Substrate-based synthetic strategies and biological activities of 1,3,4-oxadiazole: A review 基于底物的 1,3,4-恶二唑合成策略和生物活性:综述。
IF 3 4区 医学 Q2 Chemistry Pub Date : 2024-06-02 DOI: 10.1111/cbdd.14552
Upasana Sharma, Rajnish Kumar, Avijit Mazumder,  Salahuddin, Neelima Kukreti, Rashmi Mishra, M. V. N. L. Chaitanya

The five-membered 1,3,4-oxadiazole heterocyclic ring has received considerable attention because of its unique bio-isosteric properties and an unusually wide spectrum of biological activities. After a century since 1,3,4-oxadiazole was discovered, its uncommon potential attracted medicinal chemist's attention, leading to the discovery of a few presently accessible drugs containing 1,3,4-oxadiazole units, and a large number of patents have been granted on research related to 1,3,4-oxadiazole. It is worth noting that interest in 1,3,4-oxadiazoles' biological applications has doubled in the last few years. Herein, this review presents a comprehensive overview of the recent achievements in the synthesis of 1,3,4-oxadiazole-based compounds and highlights the major advances in their biological applications in the last 10 years, as well as brief remarks on prospects for further development. We hope that researchers across the scientific streams will benefit from the presented review articles for designing their work related to 1,3,4-oxadiazoles.

五元 1,3,4-恶二唑杂环因其独特的生物异构特性和异常广泛的生物活性而备受关注。自 1,3,4-恶二唑被发现一个世纪以来,其不寻常的潜力吸引了药物化学家的注意,导致发现了一些目前可获得的含有 1,3,4-恶二唑单元的药物,并授予了大量与 1,3,4-恶二唑相关的研究专利。值得注意的是,在过去几年中,人们对 1,3,4-恶二唑的生物应用兴趣倍增。在此,本综述全面概述了 1,3,4-噁二唑基化合物合成方面的最新成果,重点介绍了过去 10 年中其生物应用方面的主要进展,并简要评述了进一步发展的前景。我们希望各科学流派的研究人员在设计与 1,3,4-恶二唑相关的工作时能从这些综述文章中获益。
{"title":"Substrate-based synthetic strategies and biological activities of 1,3,4-oxadiazole: A review","authors":"Upasana Sharma,&nbsp;Rajnish Kumar,&nbsp;Avijit Mazumder,&nbsp; Salahuddin,&nbsp;Neelima Kukreti,&nbsp;Rashmi Mishra,&nbsp;M. V. N. L. Chaitanya","doi":"10.1111/cbdd.14552","DOIUrl":"10.1111/cbdd.14552","url":null,"abstract":"<p>The five-membered 1,3,4-oxadiazole heterocyclic ring has received considerable attention because of its unique bio-isosteric properties and an unusually wide spectrum of biological activities. After a century since 1,3,4-oxadiazole was discovered, its uncommon potential attracted medicinal chemist's attention, leading to the discovery of a few presently accessible drugs containing 1,3,4-oxadiazole units, and a large number of patents have been granted on research related to 1,3,4-oxadiazole. It is worth noting that interest in 1,3,4-oxadiazoles' biological applications has doubled in the last few years. Herein, this review presents a comprehensive overview of the recent achievements in the synthesis of 1,3,4-oxadiazole-based compounds and highlights the major advances in their biological applications in the last 10 years, as well as brief remarks on prospects for further development. We hope that researchers across the scientific streams will benefit from the presented review articles for designing their work related to 1,3,4-oxadiazoles.</p>","PeriodicalId":143,"journal":{"name":"Chemical Biology & Drug Design","volume":null,"pages":null},"PeriodicalIF":3.0,"publicationDate":"2024-06-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141201594","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}
引用次数: 0
Peimenine unleashes therapeutic promise in urothelial bladder cancer: inhibition of proliferation, induction of cell death and modulation of key pathways 培美宁释放出对尿路膀胱癌的治疗前景:抑制增殖、诱导细胞死亡和调节关键通路。
IF 3 4区 医学 Q2 Chemistry Pub Date : 2024-05-29 DOI: 10.1111/cbdd.14528
Zhao Yang, Rui Guo, Ying Bi, Wenkai Xu, Mingxuan Hao, Youfeng Liang, Yongchao Li, Haifeng Wang, Jun Zhang, Jianxin Xie, Chuanxing Wan, Jirui Sun

Peimenine (PEI) is a steroid alkaloid substance isolated from Fritillaria thunbergii bulbs. It has various pharmacological activities, such as relief from coughs and asthma, expectorant properties, antibacterial effects, sedative qualities, and anti-inflammatory properties. Notably, PEI can effectively inhibit the proliferation and tumor formation of liver cancer and osteosarcoma cells by inducing autophagic cell death. However, the precise effect and mechanisms of PEI on urothelial bladder cancer (UBC) cells remain uncertain. Thus, this study aims to investigate the impact of PEI on UBC cells both in vivo and in vitro. The IC50 values of BIU-87 and EJ-1 cells after 48 h were 710.3 and 651.1 μg/mL, respectively. Additionally, PEI blocked the cell cycle in BIU-87 and EJ-1 cells during the G1 phase. Furthermore, it hindered the migration of BIU-87 and EJ-1 cells substantially. PEI significantly inhibited the tumor development of EJ-1 cells within the xenograft tumor model in vivo. Mechanically, PEI augmented the protein and mRNA expression of BIM, BAK1, and Cytochrome C (CYCS) in UBC cells. Taken together, PEI suppressed the proliferation of UBC cells both in vitro and in vivo by inducing cell death and cell cycle arrest, suggesting that PEI could be applied in the treatment of UBC.

Peimenine (PEI)是从 Fritillaria thunbergii 鳞茎中分离出来的一种甾体生物碱物质。它具有多种药理作用,如止咳平喘、祛痰、抗菌、镇静和消炎。值得注意的是,PEI 可通过诱导细胞自噬死亡,有效抑制肝癌和骨肉瘤细胞的增殖和肿瘤形成。然而,PEI 对尿道膀胱癌(UBC)细胞的确切作用和机制仍不确定。因此,本研究旨在调查 PEI 在体内和体外对尿道膀胱癌细胞的影响。48 小时后,BIU-87 和 EJ-1 细胞的 IC50 值分别为 710.3 和 651.1 μg/mL。此外,PEI 还能阻断 BIU-87 和 EJ-1 细胞 G1 期的细胞周期。此外,它还大大阻碍了 BIU-87 和 EJ-1 细胞的迁移。在体内异种移植肿瘤模型中,PEI 能明显抑制 EJ-1 细胞的肿瘤发展。从机理上讲,PEI 增加了 UBC 细胞中 BIM、BAK1 和细胞色素 C (CYCS) 的蛋白和 mRNA 表达。综上所述,PEI 通过诱导细胞死亡和细胞周期停滞来抑制 UBC 细胞在体外和体内的增殖,这表明 PEI 可用于治疗 UBC。
{"title":"Peimenine unleashes therapeutic promise in urothelial bladder cancer: inhibition of proliferation, induction of cell death and modulation of key pathways","authors":"Zhao Yang,&nbsp;Rui Guo,&nbsp;Ying Bi,&nbsp;Wenkai Xu,&nbsp;Mingxuan Hao,&nbsp;Youfeng Liang,&nbsp;Yongchao Li,&nbsp;Haifeng Wang,&nbsp;Jun Zhang,&nbsp;Jianxin Xie,&nbsp;Chuanxing Wan,&nbsp;Jirui Sun","doi":"10.1111/cbdd.14528","DOIUrl":"10.1111/cbdd.14528","url":null,"abstract":"<p>Peimenine (PEI) is a steroid alkaloid substance isolated from <i>Fritillaria thunbergii</i> bulbs. It has various pharmacological activities, such as relief from coughs and asthma, expectorant properties, antibacterial effects, sedative qualities, and anti-inflammatory properties. Notably, PEI can effectively inhibit the proliferation and tumor formation of liver cancer and osteosarcoma cells by inducing autophagic cell death. However, the precise effect and mechanisms of PEI on urothelial bladder cancer (UBC) cells remain uncertain. Thus, this study aims to investigate the impact of PEI on UBC cells both in vivo and in vitro. The IC<sub>50</sub> values of BIU-87 and EJ-1 cells after 48 h were 710.3 and 651.1 μg/mL, respectively. Additionally, PEI blocked the cell cycle in BIU-87 and EJ-1 cells during the G1 phase. Furthermore, it hindered the migration of BIU-87 and EJ-1 cells substantially. PEI significantly inhibited the tumor development of EJ-1 cells within the xenograft tumor model in vivo. Mechanically, PEI augmented the protein and mRNA expression of <i>BIM</i>, <i>BAK1</i>, and <i>Cytochrome C</i> (<i>CYCS)</i> in UBC cells. Taken together, PEI suppressed the proliferation of UBC cells both in vitro and in vivo by inducing cell death and cell cycle arrest, suggesting that PEI could be applied in the treatment of UBC.</p>","PeriodicalId":143,"journal":{"name":"Chemical Biology & Drug Design","volume":null,"pages":null},"PeriodicalIF":3.0,"publicationDate":"2024-05-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141173788","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}
引用次数: 0
Design, synthesis, and antibacterial activity of pleuromutilin derivatives 胸腺嘧啶衍生物的设计、合成和抗菌活性。
IF 3 4区 医学 Q2 Chemistry Pub Date : 2024-05-28 DOI: 10.1111/cbdd.14554
Hui-xian Liu, Wen-yu Yao, Ge Cui, Jing Zhou, Hao Yan, Hui Guo, Yu-wei Wang, Yue Zhang

This paper reports the design, synthesis, and antibacterial activity study of pleuromutilin derivatives with 2-methyl-4-nitroaniline and 2-methoxy-4-nitroaniline side chains at the C22 position. The structures of the new compounds were characterized by 1H-NMR, 13C-NMR and HRMS. The inhibitory activity of the compounds against MSSA, pyogeniccoccus, streptococcus, and MRSA strains was determined using the micro broth dilution method. The results showed that the compounds exhibited certain activity against Gram-positive bacteria, among which compounds A8a, A8b, A8c, A8d, and A7 demonstrated superior antibacterial activity against MSSA, MRSA, and pyogeniccoccus compared to tiamulin, although the derivatives showed lower antibacterial activity against streptococcus compared to the control drug. Based on the favorable in vitro activity of A8c, the time-kill kinetics against MRSA were evaluated, revealing that compound A8c could inhibit bacterial proliferation in a concentration-dependent manner.

本文报告了在 C22 位置上带有 2-甲基-4-硝基苯胺和 2-甲氧基-4-硝基苯胺侧链的褶菌林衍生物的设计、合成和抗菌活性研究。新化合物的结构通过 1H-NMR、13C-NMR 和 HRMS 进行了表征。利用微肉汤稀释法测定了化合物对 MSSA、化脓性球菌、链球菌和 MRSA 菌株的抑制活性。结果表明,这些化合物对革兰氏阳性菌具有一定的活性,其中化合物 A8a、A8b、A8c、A8d 和 A7 对 MSSA、MRSA 和化脓性球菌的抗菌活性优于替莫林,但与对照药物相比,这些衍生物对链球菌的抗菌活性较低。基于 A8c 良好的体外活性,对其针对 MRSA 的时间杀伤动力学进行了评估,结果表明化合物 A8c 能以浓度依赖的方式抑制细菌增殖。
{"title":"Design, synthesis, and antibacterial activity of pleuromutilin derivatives","authors":"Hui-xian Liu,&nbsp;Wen-yu Yao,&nbsp;Ge Cui,&nbsp;Jing Zhou,&nbsp;Hao Yan,&nbsp;Hui Guo,&nbsp;Yu-wei Wang,&nbsp;Yue Zhang","doi":"10.1111/cbdd.14554","DOIUrl":"10.1111/cbdd.14554","url":null,"abstract":"<p>This paper reports the design, synthesis, and antibacterial activity study of pleuromutilin derivatives with 2-methyl-4-nitroaniline and 2-methoxy-4-nitroaniline side chains at the C22 position. The structures of the new compounds were characterized by <sup>1</sup>H-NMR, <sup>13</sup>C-NMR and HRMS. The inhibitory activity of the compounds against MSSA, <i>pyogeniccoccus</i>, <i>streptococcus</i>, and MRSA strains was determined using the micro broth dilution method. The results showed that the compounds exhibited certain activity against Gram-positive bacteria, among which compounds A8a, A8b, A8c, A8d, and A7 demonstrated superior antibacterial activity against MSSA, MRSA, and <i>pyogeniccoccus</i> compared to tiamulin, although the derivatives showed lower antibacterial activity against <i>streptococcus</i> compared to the control drug. Based on the favorable in vitro activity of A8c, the time-kill kinetics against MRSA were evaluated, revealing that compound A8c could inhibit bacterial proliferation in a concentration-dependent manner.</p>","PeriodicalId":143,"journal":{"name":"Chemical Biology & Drug Design","volume":null,"pages":null},"PeriodicalIF":3.0,"publicationDate":"2024-05-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141162867","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}
引用次数: 0
Phenotypic assessment of antiviral activity for spiro-annulated oxepanes and azepenes 对螺烷化氧杂环庚烷和氮杂环庚烷的抗病毒活性进行表型评估。
IF 3 4区 医学 Q2 Chemistry Pub Date : 2024-05-24 DOI: 10.1111/cbdd.14553
Dmitry I. Osolodkin, Liubov I. Kozlovskaya, Ildar R. Iusupov, Alexander V. Kurkin, Elena Y. Shustova, Alexey A. Orlov, Evgeny V. Khvatov, Elena S. Mutnykh, Svetlana S. Kurashova, Anna N. Vetrova, Darya O. Yatsenko, Alexander S. Goryashchenko, Vladimir N. Ivanov, Evgeny R. Lukyanenko, Evgenia V. Karpova, Daria A. Stepanova, Viktor P. Volok, Svetlana E. Sotskova, Tamara K. Dzagurova, Galina G. Karganova, Alexander N. Lukashev, Aydar A. Ishmukhametov

Evolutionary potential of viruses can result in outbreaks of well-known viruses and emergence of novel ones. Pharmacological methods of intervening the reproduction of various less popular, but not less important viruses are not available, as well as the spectrum of antiviral activity for most known compounds. In the framework of chemical biology paradigm, characterization of antiviral activity spectrum of new compounds allows to extend the antiviral chemical space and provides new important structure–activity relationships for data-driven drug discovery. Here we present a primary assessment of antiviral activity of spiro-annulated derivatives of seven-membered heterocycles, oxepane and azepane, in phenotypic assays against viruses with different genomes, virion structures, and genome realization schemes: orthoflavivirus (tick-borne encephalitis virus, TBEV), enteroviruses (poliovirus, enterovirus A71, echovirus 30), adenovirus (human adenovirus C5), hantavirus (Puumala virus). Hit compounds inhibited reproduction of adenovirus C5, the only DNA virus in the studied set, in the yield reduction assay, and did not inhibit reproduction of RNA viruses.

病毒的进化潜力会导致知名病毒的爆发和新型病毒的出现。目前还没有干预各种不常见但同样重要的病毒繁殖的药理学方法,也没有大多数已知化合物的抗病毒活性谱。在化学生物学范式的框架下,对新化合物的抗病毒活性谱进行表征可以扩展抗病毒化学空间,并为数据驱动的药物发现提供新的重要结构-活性关系。在此,我们针对不同基因组、病毒体结构和基因组实现方案的病毒(正黄病毒(蜱传脑炎病毒,TBEV)、肠道病毒(脊髓灰质炎病毒、肠道病毒 A71、埃可病毒 30)、腺病毒(人腺病毒 C5)和汉坦病毒(普马拉病毒)),在表型试验中对七元杂环的螺烷化衍生物(氧杂环庚烷和氮杂环庚烷)的抗病毒活性进行了初步评估。在减产试验中,命中化合物抑制了腺病毒 C5(所研究的病毒集中唯一的 DNA 病毒)的繁殖,但没有抑制 RNA 病毒的繁殖。
{"title":"Phenotypic assessment of antiviral activity for spiro-annulated oxepanes and azepenes","authors":"Dmitry I. Osolodkin,&nbsp;Liubov I. Kozlovskaya,&nbsp;Ildar R. Iusupov,&nbsp;Alexander V. Kurkin,&nbsp;Elena Y. Shustova,&nbsp;Alexey A. Orlov,&nbsp;Evgeny V. Khvatov,&nbsp;Elena S. Mutnykh,&nbsp;Svetlana S. Kurashova,&nbsp;Anna N. Vetrova,&nbsp;Darya O. Yatsenko,&nbsp;Alexander S. Goryashchenko,&nbsp;Vladimir N. Ivanov,&nbsp;Evgeny R. Lukyanenko,&nbsp;Evgenia V. Karpova,&nbsp;Daria A. Stepanova,&nbsp;Viktor P. Volok,&nbsp;Svetlana E. Sotskova,&nbsp;Tamara K. Dzagurova,&nbsp;Galina G. Karganova,&nbsp;Alexander N. Lukashev,&nbsp;Aydar A. Ishmukhametov","doi":"10.1111/cbdd.14553","DOIUrl":"10.1111/cbdd.14553","url":null,"abstract":"<p>Evolutionary potential of viruses can result in outbreaks of well-known viruses and emergence of novel ones. Pharmacological methods of intervening the reproduction of various less popular, but not less important viruses are not available, as well as the spectrum of antiviral activity for most known compounds. In the framework of chemical biology paradigm, characterization of antiviral activity spectrum of new compounds allows to extend the antiviral chemical space and provides new important structure–activity relationships for data-driven drug discovery. Here we present a primary assessment of antiviral activity of spiro-annulated derivatives of seven-membered heterocycles, oxepane and azepane, in phenotypic assays against viruses with different genomes, virion structures, and genome realization schemes: orthoflavivirus (tick-borne encephalitis virus, TBEV), enteroviruses (poliovirus, enterovirus A71, echovirus 30), adenovirus (human adenovirus C5), hantavirus (Puumala virus). Hit compounds inhibited reproduction of adenovirus C5, the only DNA virus in the studied set, in the yield reduction assay, and did not inhibit reproduction of RNA viruses.</p>","PeriodicalId":143,"journal":{"name":"Chemical Biology & Drug Design","volume":null,"pages":null},"PeriodicalIF":3.0,"publicationDate":"2024-05-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141093454","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}
引用次数: 0
期刊
Chemical Biology & Drug Design
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1