首页 > 最新文献

Chemical Biology & Drug Design最新文献

英文 中文
The medicinal chemistry of piperazines: A review 哌嗪的药物化学:综述。
IF 3 4区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-06-18 DOI: 10.1111/cbdd.14537
Md Faizan, Rajnish Kumar, Avijit Mazumder,  Salahuddin, Neelima Kukreti, Arvind Kumar, M. V. N. L. Chaitanya

The versatile basic structure of piperazine allows for the development and production of newer bioactive molecules that can be used to treat a wide range of diseases. Piperazine derivatives are unique and can easily be modified for the desired pharmacological activity. The two opposing nitrogen atoms in a six-membered piperazine ring offer a large polar surface area, relative structural rigidity, and more acceptors and donors of hydrogen bonds. These properties frequently result in greater water solubility, oral bioavailability, and ADME characteristics, as well as improved target affinity and specificity. Various synthetic protocols have been reported for piperazine and its derivatives. In this review, we focused on recently published synthetic protocols for the synthesis of the piperazine and its derivatives. The structure–activity relationship concerning different biological activities of various piperazine-containing drugs has also been highlighted to provide a good understanding to researchers for future research on piperazines.

哌嗪的基本结构多变,因此可以开发和生产用于治疗多种疾病的新型生物活性分子。哌嗪衍生物是独一无二的,可以很容易地进行改性,以获得所需的药理活性。六元哌嗪环中两个相对的氮原子具有较大的极性表面积、相对的结构刚性以及更多的氢键接受体和供体。这些特性往往能提高水溶性、口服生物利用度和 ADME 特性,并改善靶点亲和性和特异性。有关哌嗪及其衍生物的各种合成方案已有报道。在本综述中,我们重点讨论了最近发表的哌嗪及其衍生物的合成方案。此外,还重点介绍了各种含哌嗪药物的不同生物活性的结构-活性关系,以便为研究人员今后开展哌嗪类药物的研究提供一个良好的理解基础。
{"title":"The medicinal chemistry of piperazines: A review","authors":"Md Faizan,&nbsp;Rajnish Kumar,&nbsp;Avijit Mazumder,&nbsp; Salahuddin,&nbsp;Neelima Kukreti,&nbsp;Arvind Kumar,&nbsp;M. V. N. L. Chaitanya","doi":"10.1111/cbdd.14537","DOIUrl":"10.1111/cbdd.14537","url":null,"abstract":"<p>The versatile basic structure of piperazine allows for the development and production of newer bioactive molecules that can be used to treat a wide range of diseases. Piperazine derivatives are unique and can easily be modified for the desired pharmacological activity. The two opposing nitrogen atoms in a six-membered piperazine ring offer a large polar surface area, relative structural rigidity, and more acceptors and donors of hydrogen bonds. These properties frequently result in greater water solubility, oral bioavailability, and ADME characteristics, as well as improved target affinity and specificity. Various synthetic protocols have been reported for piperazine and its derivatives. In this review, we focused on recently published synthetic protocols for the synthesis of the piperazine and its derivatives. The structure–activity relationship concerning different biological activities of various piperazine-containing drugs has also been highlighted to provide a good understanding to researchers for future research on piperazines.</p>","PeriodicalId":143,"journal":{"name":"Chemical Biology & Drug Design","volume":"103 6","pages":""},"PeriodicalIF":3.0,"publicationDate":"2024-06-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141422133","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
Integrated bioinformatics analysis reveals the bidirectional effects of TSPAN6 for cisplatin resistance in lung cancer 综合生物信息学分析揭示了 TSPAN6 对肺癌顺铂耐药性的双向影响。
IF 3 4区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-06-17 DOI: 10.1111/cbdd.14570
Zhihong Fang, Jinmei Bai

Cisplatin-based chemotherapy is frequently employed as the primary therapeutic approach for advanced lung cancer. Nevertheless, a significant proportion of patients may develop resistance to cisplatin, leading to diminished efficacy of chemotherapy. Through analysis of Gene Expression Omnibus databases, TSPAN6 has been identified as a key factor in conferring resistance to cisplatin, attributed to its activation of the NF-κB signaling pathway. Knockdown of TSPAN6 using siRNA resulted in decreased expression levels of NF-κB in A549 cells. This indicates that TSPAN6 may have dual effects on lung cancer cisplatin resistance and could serve as a promising therapeutic target for individuals with cisplatin resistance.

以顺铂为基础的化疗经常被用作晚期肺癌的主要治疗方法。然而,相当一部分患者可能会对顺铂产生耐药性,导致化疗效果下降。通过分析基因表达总库(Gene Expression Omnibus)数据库,发现 TSPAN6 是导致顺铂耐药性的关键因素,这归因于它激活了 NF-κB 信号通路。使用 siRNA 敲除 TSPAN6 可降低 A549 细胞中 NF-κB 的表达水平。这表明TSPAN6可能对肺癌顺铂耐药有双重作用,可作为顺铂耐药患者的治疗靶点。
{"title":"Integrated bioinformatics analysis reveals the bidirectional effects of TSPAN6 for cisplatin resistance in lung cancer","authors":"Zhihong Fang,&nbsp;Jinmei Bai","doi":"10.1111/cbdd.14570","DOIUrl":"10.1111/cbdd.14570","url":null,"abstract":"<p>Cisplatin-based chemotherapy is frequently employed as the primary therapeutic approach for advanced lung cancer. Nevertheless, a significant proportion of patients may develop resistance to cisplatin, leading to diminished efficacy of chemotherapy. Through analysis of Gene Expression Omnibus databases, <i>TSPAN6</i> has been identified as a key factor in conferring resistance to cisplatin, attributed to its activation of the NF-κB signaling pathway. Knockdown of <i>TSPAN6</i> using siRNA resulted in decreased expression levels of NF-κB in A549 cells. This indicates that TSPAN6 may have dual effects on lung cancer cisplatin resistance and could serve as a promising therapeutic target for individuals with cisplatin resistance.</p>","PeriodicalId":143,"journal":{"name":"Chemical Biology & Drug Design","volume":"103 6","pages":""},"PeriodicalIF":3.0,"publicationDate":"2024-06-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141422132","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
Development of phenyl-urea-based small molecules that target penicillin-binding protein 4 开发以青霉素结合蛋白 4 为靶标的苯基脲基小分子。
IF 3 4区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-06-14 DOI: 10.1111/cbdd.14569
Vijay S. Gondil, Hailey S. Butman, Mikaeel Young, Danica J. Walsh, Yogesh Narkhede, Michael J. Zeiler, Andrew H. Crow, Morgan E. Carpenter, Aashay Mardikar, Roberta J. Melander, Olaf Wiest, Paul M. Dunman, Christian Melander

Staphylococcus aureus has the ability to invade cortical bone osteocyte lacuno-canalicular networks (OLCNs) and cause osteomyelitis. It was recently established that the cell wall transpeptidase, penicillin-binding protein 4 (PBP4), is crucial for this function, with pbp4 deletion strains unable to invade OLCNs and cause bone pathogenesis in a murine model of S. aureus osteomyelitis. Moreover, PBP4 has recently been found to modulate S. aureus resistance to β-lactam antibiotics. As such, small molecule inhibitors of S. aureus PBP4 may represent dual functional antimicrobial agents that limit osteomyelitis and/or reverse antibiotic resistance. A high throughput screen recently revealed that the phenyl-urea 1 targets PBP4. Herein, we describe a structure–activity relationship (SAR) study on 1. Leveraging in silico docking and modeling, a set of analogs was synthesized and assessed for PBP4 inhibitory activities. Results revealed a preliminary SAR and identified lead compounds with enhanced binding to PBP4, more potent antibiotic resistance reversal, and diminished PBP4 cell wall transpeptidase activity in comparison to 1.

金黄色葡萄球菌能够侵入皮质骨的骨细胞裂隙-髓鞘网络(OLCN)并引起骨髓炎。最近研究发现,细胞壁转肽酶--青霉素结合蛋白 4(PBP4)对这一功能至关重要,在金黄色葡萄球菌骨髓炎小鼠模型中,PBP4 缺失株无法侵入 OLCNs 并导致骨发病。此外,最近还发现 PBP4 可调节金黄色葡萄球菌对β-内酰胺类抗生素的耐药性。因此,金黄色葡萄球菌 PBP4 的小分子抑制剂可能是限制骨髓炎和/或逆转抗生素耐药性的双重功能抗菌剂。最近的一项高通量筛选发现,苯基脲 1 以 PBP4 为靶标。在此,我们描述了对 1 的结构-活性关系 (SAR) 研究。利用硅学对接和建模,我们合成了一组类似物,并评估了它们对 PBP4 的抑制活性。结果显示,与 1 相比,初步的 SAR 和鉴定出的先导化合物与 PBP4 的结合力更强、抗生素耐药性逆转作用更强、PBP4 细胞壁转肽酶活性更弱。
{"title":"Development of phenyl-urea-based small molecules that target penicillin-binding protein 4","authors":"Vijay S. Gondil,&nbsp;Hailey S. Butman,&nbsp;Mikaeel Young,&nbsp;Danica J. Walsh,&nbsp;Yogesh Narkhede,&nbsp;Michael J. Zeiler,&nbsp;Andrew H. Crow,&nbsp;Morgan E. Carpenter,&nbsp;Aashay Mardikar,&nbsp;Roberta J. Melander,&nbsp;Olaf Wiest,&nbsp;Paul M. Dunman,&nbsp;Christian Melander","doi":"10.1111/cbdd.14569","DOIUrl":"10.1111/cbdd.14569","url":null,"abstract":"<p><i>Staphylococcus aureus</i> has the ability to invade cortical bone osteocyte lacuno-canalicular networks (OLCNs) and cause osteomyelitis. It was recently established that the cell wall transpeptidase, penicillin-binding protein 4 (PBP4), is crucial for this function, with <i>pbp4</i> deletion strains unable to invade OLCNs and cause bone pathogenesis in a murine model of <i>S</i>. <i>aureus</i> osteomyelitis. Moreover, PBP4 has recently been found to modulate <i>S</i>. <i>aureus</i> resistance to β-lactam antibiotics. As such, small molecule inhibitors of <i>S</i>. <i>aureus</i> PBP4 may represent dual functional antimicrobial agents that limit osteomyelitis and/or reverse antibiotic resistance. A high throughput screen recently revealed that the phenyl-urea <b>1</b> targets PBP4. Herein, we describe a structure–activity relationship (SAR) study on <b>1.</b> Leveraging in silico docking and modeling, a set of analogs was synthesized and assessed for PBP4 inhibitory activities. Results revealed a preliminary SAR and identified lead compounds with enhanced binding to PBP4, more potent antibiotic resistance reversal, and diminished PBP4 cell wall transpeptidase activity in comparison to <b>1</b>.</p>","PeriodicalId":143,"journal":{"name":"Chemical Biology & Drug Design","volume":"103 6","pages":""},"PeriodicalIF":3.0,"publicationDate":"2024-06-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141322229","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
Brusatol induces ferroptosis to inhibit hepatocellular carcinoma progression by targeting ATF3 布芦沙托通过靶向 ATF3 诱导铁变态反应,从而抑制肝细胞癌的进展。
IF 3 4区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-06-11 DOI: 10.1111/cbdd.14565
Yuanyuan Wan, Jingsong Cheng, Debiao Gan, Jiaming He, An Chen, Jing Ma, Yunying Li, Xiao Wang, Jianhua Ran, Dilong Chen, Jing Li

Ferroptosis is a novel form of programmed cell death that is triggered by iron-dependent lipid peroxidation. Brusatol (BRU), a natural nuclear factor erythroid 2-related factor 2 inhibitor, exhibits potent anticancer effects in various types of cancer. However, the exact mechanism of BRU in the treatment of hepatocellular carcinoma (HCC) remains unknown. The anticancer effects of BRU in HCC were detected using cell counting kit-8 and colony formation assays and a xenograft model. RNA sequencing (RNA-seq) and bioinformatics analyses of HCC cells were utilized to elucidate the mechanism underlying the effects of BRU in HCC. The levels of reactive oxygen species (ROS), glutathione (GSH), malondialdehyde (MDA), and Fe2+ were measured using assay kits. The expression of activating transcription factor 3 (ATF3) was tested using RT-qPCR, western blotting, and immunofluorescence staining. The role of ATF3 in BRU-induced ferroptosis was examined using siATF3. BRU significantly inhibited HCC cell proliferation, both in vitro and in vivo. BRU activated the ferroptosis signaling pathway and increased ATF3 expression. Furthermore, ATF3 knockdown impeded BRU-induced ferroptosis. BRU suppressed HCC growth through ATF3-mediated ferroptosis, supporting BRU as a promising therapeutic agent for HCC.

铁氧化是一种新型的程序性细胞死亡,由铁依赖性脂质过氧化引发。Brusatol (BRU) 是一种天然的核因子红细胞 2 相关因子 2 抑制剂,对各种类型的癌症都有很强的抗癌作用。然而,BRU 治疗肝细胞癌(HCC)的确切机制仍不清楚。我们使用细胞计数试剂盒-8和集落形成试验以及异种移植模型检测了BRU对HCC的抗癌作用。通过对HCC细胞进行RNA测序(RNA-seq)和生物信息学分析,阐明了BRU在HCC中的作用机制。使用检测试剂盒测定了活性氧(ROS)、谷胱甘肽(GSH)、丙二醛(MDA)和Fe2+的水平。使用 RT-qPCR、Western 印迹和免疫荧光染色检测了活化转录因子 3(ATF3)的表达。使用 siATF3 检测了 ATF3 在 BRU 诱导的铁变态反应中的作用。BRU在体外和体内都能明显抑制HCC细胞的增殖。BRU激活了铁变态反应信号通路并增加了ATF3的表达。此外,ATF3的敲除阻碍了BRU诱导的铁变态反应。BRU通过ATF3介导的铁蛋白沉积抑制了HCC的生长,支持BRU成为治疗HCC的一种有前景的药物。
{"title":"Brusatol induces ferroptosis to inhibit hepatocellular carcinoma progression by targeting ATF3","authors":"Yuanyuan Wan,&nbsp;Jingsong Cheng,&nbsp;Debiao Gan,&nbsp;Jiaming He,&nbsp;An Chen,&nbsp;Jing Ma,&nbsp;Yunying Li,&nbsp;Xiao Wang,&nbsp;Jianhua Ran,&nbsp;Dilong Chen,&nbsp;Jing Li","doi":"10.1111/cbdd.14565","DOIUrl":"10.1111/cbdd.14565","url":null,"abstract":"<p>Ferroptosis is a novel form of programmed cell death that is triggered by iron-dependent lipid peroxidation. Brusatol (BRU), a natural nuclear factor erythroid 2-related factor 2 inhibitor, exhibits potent anticancer effects in various types of cancer. However, the exact mechanism of BRU in the treatment of hepatocellular carcinoma (HCC) remains unknown. The anticancer effects of BRU in HCC were detected using cell counting kit-8 and colony formation assays and a xenograft model. RNA sequencing (RNA-seq) and bioinformatics analyses of HCC cells were utilized to elucidate the mechanism underlying the effects of BRU in HCC. The levels of reactive oxygen species (ROS), glutathione (GSH), malondialdehyde (MDA), and Fe<sup>2+</sup> were measured using assay kits. The expression of activating transcription factor 3 (ATF3) was tested using RT-qPCR, western blotting, and immunofluorescence staining. The role of ATF3 in BRU-induced ferroptosis was examined using siATF3. BRU significantly inhibited HCC cell proliferation, both in vitro <i>and</i> in vivo. BRU activated the ferroptosis signaling pathway and increased ATF3 expression. Furthermore, ATF3 knockdown impeded BRU-induced ferroptosis. BRU suppressed HCC growth through ATF3-mediated ferroptosis, supporting BRU as a promising therapeutic agent for HCC.</p>","PeriodicalId":143,"journal":{"name":"Chemical Biology & Drug Design","volume":"103 6","pages":""},"PeriodicalIF":3.0,"publicationDate":"2024-06-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141307629","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
Click chemistry beyond metal-catalyzed cycloaddition as a remarkable tool for green chemical synthesis of antifungal medications 超越金属催化环化的点击化学是绿色化学合成抗真菌药物的重要工具。
IF 3 4区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-06-11 DOI: 10.1111/cbdd.14555
Azar Tahghighi, Parisa Azerang

Click chemistry is widely used for the efficient synthesis of 1,4-disubstituted-1,2,3-triazole, a well-known scaffold with widespread biological activity in the pharmaceutical sciences. In recent years, this magic ring has attracted the attention of scientists for its potential in designing and synthesizing new antifungal agents. Despite scientific and medical advances, fungal infections still account for more than 1.5 million deaths globally per year, especially in people with compromised immune function. This increasing trend is definitely related to a raise in the incidence of fungal infections and prevalence of antifungal drug resistance. In this condition, an urgent need for new alternative antifungals is undeniable. By focusing on the main aspects of reaction conditions in click chemistry, this review was conducted to classify antifungal 1,4-disubstituted-1,2,3-triazole hybrids based on their chemical structures and introduce the most effective triazole antifungal derivatives. It was notable that in all reactions studied, Cu(I) catalysts generated in situ by the reduction in Cu(II) salts or used copper(I) salts directly, as well as mixed solvents of t-BuOH/H2O and DMF/H2O had most application in the synthesis of triazole ring. The most effective antifungal activity was also observed in fluconazole analogs containing 1,2,3-triazole moiety and benzo-fused five/six-membered heterocyclic conjugates with a 1,2,3-triazole ring, even with better activity than fluconazole. The findings of structure–activity relationship and molecular docking of antifungal derivatives synthesized with copper-catalyzed azide–alkyne cycloaddition (CuAAC) could offer medicinal chemistry scientists valuable data on designing and synthesizing novel triazole antifungals with more potent biological activities in their future research.

点击化学被广泛应用于 1,4-二取代-1,2,3-三唑的高效合成,这是一种著名的支架,在医药科学领域具有广泛的生物活性。近年来,这种神奇的环因其在设计和合成新型抗真菌剂方面的潜力而引起了科学家们的关注。尽管科学和医学在不断进步,全球每年仍有 150 多万人死于真菌感染,尤其是免疫功能低下的人群。这一增长趋势无疑与真菌感染发病率的上升和抗真菌药物耐药性的流行有关。在这种情况下,对新型替代抗真菌药物的迫切需要是不容置疑的。通过关注点击化学反应条件的主要方面,本综述根据化学结构对抗真菌的 1,4-二取代-1,2,3-三唑杂化物进行了分类,并介绍了最有效的三唑抗真菌衍生物。值得注意的是,在所有研究的反应中,由 Cu(II) 盐还原原位生成的 Cu(I) 催化剂或直接使用的铜(I) 盐,以及 t-BuOH/H2O 和 DMF/H2O 混合溶剂在三唑环的合成中应用最多。此外,含有 1,2,3-三唑分子的氟康唑类似物和苯并融合五/六元杂环的 1,2,3-三唑环共轭物也具有最有效的抗真菌活性,其活性甚至优于氟康唑。铜催化叠氮-炔环加成法(CuAAC)合成的抗真菌衍生物的结构-活性关系和分子对接研究结果,为药物化学科学家在今后的研究中设计和合成具有更强生物活性的新型三唑类抗真菌药物提供了宝贵的数据。
{"title":"Click chemistry beyond metal-catalyzed cycloaddition as a remarkable tool for green chemical synthesis of antifungal medications","authors":"Azar Tahghighi,&nbsp;Parisa Azerang","doi":"10.1111/cbdd.14555","DOIUrl":"10.1111/cbdd.14555","url":null,"abstract":"<p>Click chemistry is widely used for the efficient synthesis of 1,4-disubstituted-1,2,3-triazole, a well-known scaffold with widespread biological activity in the pharmaceutical sciences. In recent years, this magic ring has attracted the attention of scientists for its potential in designing and synthesizing new antifungal agents. Despite scientific and medical advances, fungal infections still account for more than 1.5 million deaths globally per year, especially in people with compromised immune function. This increasing trend is definitely related to a raise in the incidence of fungal infections and prevalence of antifungal drug resistance. In this condition, an urgent need for new alternative antifungals is undeniable. By focusing on the main aspects of reaction conditions in click chemistry, this review was conducted to classify antifungal 1,4-disubstituted-1,2,3-triazole hybrids based on their chemical structures and introduce the most effective triazole antifungal derivatives. It was notable that in all reactions studied, Cu(I) catalysts generated in situ by the reduction in Cu(II) salts or used copper(I) salts directly, as well as mixed solvents of t-BuOH/H<sub>2</sub>O and DMF/H<sub>2</sub>O had most application in the synthesis of triazole ring. The most effective antifungal activity was also observed in fluconazole analogs containing 1,2,3-triazole moiety and benzo-fused five/six-membered heterocyclic conjugates with a 1,2,3-triazole ring, even with better activity than fluconazole. The findings of structure–activity relationship and molecular docking of antifungal derivatives synthesized with copper-catalyzed azide–alkyne cycloaddition (CuAAC) could offer medicinal chemistry scientists valuable data on designing and synthesizing novel triazole antifungals with more potent biological activities in their future research.</p>","PeriodicalId":143,"journal":{"name":"Chemical Biology & Drug Design","volume":"103 6","pages":""},"PeriodicalIF":3.0,"publicationDate":"2024-06-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141307630","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 anticancer properties of the phytoconstituents and comparative analysis of their chemical space parameters with USFDA-approved synthetic anticancer agents 探索植物成分的抗癌特性,并将其化学空间参数与 USFDA 批准的合成抗癌剂进行比较分析。
IF 3 4区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-06-11 DOI: 10.1111/cbdd.14561
Somesh Thapliyal, Ritu Vishnoi, Yogesh Murti, Roshan Kumar, Nirja Chavan, Pramod Rawat, Gaurav Joshi, Ashish Ranjan Dwivedi, Kapil Kumar Goel

The present review article thoroughly analyses natural products and their derived phytoconstituents as a rich source of plausible anticancer drugs. The study thoroughly explores the chemical components derived from various natural sources, thus emphasizing their unique structural characteristics and therapeutic potential as an anticancer agent. The review contains the critical chemical constituents' in-depth molecular mechanisms, their source's chemical structures and the categories. The review also comprises an exhaustive and comprehensive analysis of different chemical spacing parameters of the anticancer agents derived from natural products. It compares them with USFDA-approved synthetic anticancer drugs up to 2020, thus providing a meaningful understanding of the relationship between natural and synthetic compounds portraying the anticancer assets. The review also delves more deeply into the chemical analysis of the heterocyclic moieties from the natural product arena, illustrating the anticancer mechanisms. The present article is, therefore, expected to serve as a valuable resource for natural product and medicinal chemists, encouraging and promoting an integrated approach to exploit the potential of natural products in drug discovery development and translational research, which have a prerequisite of bench to bedside approach. The work could guide researchers toward innovative approaches for the ever-evolving field of anticancer drug discovery.

本综述文章深入分析了天然产品及其衍生的植物成分,认为它们是抗癌药物的丰富来源。研究深入探讨了从各种天然资源中提取的化学成分,从而强调了它们作为抗癌剂的独特结构特征和治疗潜力。综述包括关键化学成分的深入分子机制、其来源的化学结构和类别。该综述还详尽全面地分析了从天然产品中提取的抗癌剂的不同化学间距参数。它将这些抗癌剂与美国食品及药物管理局(USFDA)截至 2020 年批准的合成抗癌药物进行了比较,从而提供了对天然化合物与合成化合物之间关系的有意义的理解,描绘了抗癌资产。综述还更深入地探讨了天然产品中杂环分子的化学分析,并阐述了抗癌机理。因此,本文有望成为天然产物和药物化学家的宝贵资源,鼓励和促进在药物发现开发和转化研究中采用综合方法挖掘天然产物的潜力,而这需要从工作台到床边的方法。这项工作可以指导研究人员在不断发展的抗癌药物发现领域采用创新方法。
{"title":"Exploring anticancer properties of the phytoconstituents and comparative analysis of their chemical space parameters with USFDA-approved synthetic anticancer agents","authors":"Somesh Thapliyal,&nbsp;Ritu Vishnoi,&nbsp;Yogesh Murti,&nbsp;Roshan Kumar,&nbsp;Nirja Chavan,&nbsp;Pramod Rawat,&nbsp;Gaurav Joshi,&nbsp;Ashish Ranjan Dwivedi,&nbsp;Kapil Kumar Goel","doi":"10.1111/cbdd.14561","DOIUrl":"10.1111/cbdd.14561","url":null,"abstract":"<p>The present review article thoroughly analyses natural products and their derived phytoconstituents as a rich source of plausible anticancer drugs. The study thoroughly explores the chemical components derived from various natural sources, thus emphasizing their unique structural characteristics and therapeutic potential as an anticancer agent. The review contains the critical chemical constituents' in-depth molecular mechanisms, their source's chemical structures and the categories. The review also comprises an exhaustive and comprehensive analysis of different chemical spacing parameters of the anticancer agents derived from natural products. It compares them with USFDA-approved synthetic anticancer drugs up to 2020, thus providing a meaningful understanding of the relationship between natural and synthetic compounds portraying the anticancer assets. The review also delves more deeply into the chemical analysis of the heterocyclic moieties from the natural product arena, illustrating the anticancer mechanisms. The present article is, therefore, expected to serve as a valuable resource for natural product and medicinal chemists, encouraging and promoting an integrated approach to exploit the potential of natural products in drug discovery development and translational research, which have a prerequisite of bench to bedside approach. The work could guide researchers toward innovative approaches for the ever-evolving field of anticancer drug discovery.</p>","PeriodicalId":143,"journal":{"name":"Chemical Biology & Drug Design","volume":"103 6","pages":""},"PeriodicalIF":3.0,"publicationDate":"2024-06-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141307631","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
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 BIOCHEMISTRY & MOLECULAR BIOLOGY 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":"103 6","pages":""},"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 BIOCHEMISTRY & MOLECULAR BIOLOGY 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":"103 6","pages":""},"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":"103 6","pages":""},"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 BIOCHEMISTRY & MOLECULAR BIOLOGY 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":"103 6","pages":""},"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
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