首页 > 最新文献

Computational biology and chemistry最新文献

英文 中文
Improving binding affinity prediction by emphasizing local features of drug and protein. 通过强调药物和蛋白质的局部特征,提高结合亲和力预测。
Pub Date : 2024-12-11 DOI: 10.1016/j.compbiolchem.2024.108310
Daejin Choi, Sangjun Park

Binding affinity prediction has been considered as a fundamental task in drug discovery. Despite much effort to improve accuracy of binding affinity prediction, the prior work considered only macro-level features that can represent the characteristics of the whole architecture of a drug and a target protein, and the features from local structure of the drug and the protein tend to be lost. In this paper, we propose a deep learning model that can comprehensively extract the local features of both a drug and a target protein for accurate binding affinity prediction. The proposed model consists of two components named as Multi-Stream CNN and Multi-Stream GCN, each of which is responsible for capturing micro-level characteristics or local features from subsequences of a target protein sequence and subgraph of a drug molecule, respectively. Having multiple streams consisting of different numbers of layers, both the components can compute and preserve the local features with a stream consisting of a single layer. Our evaluation with two popular datasets, Davis and KIBA, demonstrates that the proposed model outperforms all the baseline models using the global features, implying that local features play significant roles of binding affinity prediction.

结合亲和力预测一直被认为是药物发现的一项基本任务。尽管为提高结合亲和力预测的准确性付出了很多努力,但之前的工作只考虑了能够代表药物和靶蛋白整体结构特征的宏观层面的特征,而往往失去了药物和蛋白质局部结构的特征。在本文中,我们提出了一种深度学习模型,可以综合提取药物和靶蛋白的局部特征,以准确预测结合亲和力。该模型由多流CNN和多流GCN两部分组成,分别负责从靶蛋白序列的子序列和药物分子的子图中捕获微观特征或局部特征。拥有由不同层数组成的多个流,两个组件都可以计算和保留由单层组成的流的局部特征。我们对两个流行的数据集Davis和KIBA的评估表明,所提出的模型优于所有使用全局特征的基线模型,这意味着局部特征在结合亲和力预测中发挥了重要作用。
{"title":"Improving binding affinity prediction by emphasizing local features of drug and protein.","authors":"Daejin Choi, Sangjun Park","doi":"10.1016/j.compbiolchem.2024.108310","DOIUrl":"https://doi.org/10.1016/j.compbiolchem.2024.108310","url":null,"abstract":"<p><p>Binding affinity prediction has been considered as a fundamental task in drug discovery. Despite much effort to improve accuracy of binding affinity prediction, the prior work considered only macro-level features that can represent the characteristics of the whole architecture of a drug and a target protein, and the features from local structure of the drug and the protein tend to be lost. In this paper, we propose a deep learning model that can comprehensively extract the local features of both a drug and a target protein for accurate binding affinity prediction. The proposed model consists of two components named as Multi-Stream CNN and Multi-Stream GCN, each of which is responsible for capturing micro-level characteristics or local features from subsequences of a target protein sequence and subgraph of a drug molecule, respectively. Having multiple streams consisting of different numbers of layers, both the components can compute and preserve the local features with a stream consisting of a single layer. Our evaluation with two popular datasets, Davis and KIBA, demonstrates that the proposed model outperforms all the baseline models using the global features, implying that local features play significant roles of binding affinity prediction.</p>","PeriodicalId":93952,"journal":{"name":"Computational biology and chemistry","volume":"115 ","pages":"108310"},"PeriodicalIF":0.0,"publicationDate":"2024-12-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142824772","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Exploring immune gene expression and potential regulatory mechanisms in anaplastic thyroid carcinoma using a combination of single-cell and bulk RNA sequencing data. 结合单细胞和大容量 RNA 测序数据探索甲状腺无节细胞癌的免疫基因表达和潜在调控机制
Pub Date : 2024-12-07 DOI: 10.1016/j.compbiolchem.2024.108311
Kehui Zhou, Shijia Zhang, Jinbiao Shang, Xiabin Lan

Thyroid cancer includes papillary thyroid carcinoma (PTC) and anaplastic thyroid carcinoma (ATC). While PTC has an excellent prognosis, ATC has a dismal prognosis, necessitating the identification of novel targets in ATC to aid in ATC diagnosis and treatment. Therefore, we analyzed ATC single-cell RNA sequencing (scRNA-seq) and bulk RNA sequencing (bulk RNA-seq) data from the Gene Expression Omnibus (GEO), retrieved immune-related genes from the ImmPort database, and identified differentially expressed immune genes within single-cell subgroups. The AUCell package in R was used to calculate activity scores for single-cell subgroups and identify active cell populations. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis were performed on differentially expressed genes (DEGs) in active cell populations. Then, we integrated thyroid-cancer scRNA-seq and bulk RNA-seq data to identify overlapping DEGs. Relevant transcription factors (TFs) were retrieved from the TRRUST database. A protein-protein interaction (PPI) network for key TFs was created using the STRING database. Simultaneously, drugs associated with key TFs were obtained from DGIdb. ScRNA-seq cluster analysis showed that T/natural killer (NK) cells were more distributed in ATC and that thyrocytes cells were more distributed in PTC. We obtained 264 differential immune genes (DIGs) from the IMMPORT database and integrated scRNA-seq cluster analysis to identify the active cell T/NK cells and myeloid cells. Integrated bulk RNA-seq analysis obtained common immune genes (CIGs) such as TMSB4X, NFKB1, TNFRSF1B, and B2M. The nine CIG-related TFs (CEBPB, SPI1, NFKB1, RUNX1, NFE2L2, REL, CIITA, KLF6, and CEBPD) in myeloid cells and three TFs (NFKB1, FOXO1, and NR3C1) in T/NK cells were obtained from the TRRUST database. The key genes we identified represent potential targets for treating ATC.

甲状腺癌包括甲状腺乳头状癌(PTC)和间变性甲状腺癌(ATC)。PTC预后良好,而ATC预后不佳,因此需要在ATC中发现新的靶点,以帮助ATC的诊断和治疗。因此,我们分析了来自Gene Expression Omnibus (GEO)的ATC单细胞RNA测序(scRNA-seq)和大量RNA测序(bulk RNA-seq)数据,从import数据库检索免疫相关基因,并鉴定了单细胞亚群中差异表达的免疫基因。使用R中的AUCell包计算单细胞亚组的活性评分并识别活性细胞群。对活性细胞群体中的差异表达基因(DEGs)进行基因本体(GO)和京都基因与基因组百科全书(KEGG)富集分析。然后,我们整合甲状腺癌scRNA-seq和大量RNA-seq数据来识别重叠的基因。相关转录因子(tf)从trust数据库中检索。利用STRING数据库建立了关键tf的蛋白-蛋白相互作用(PPI)网络。同时,从DGIdb中获得了与关键tf相关的药物。ScRNA-seq聚类分析显示,T/ NK细胞在ATC中分布较多,甲状腺细胞在PTC中分布较多。我们从import数据库中获得264个差异免疫基因(DIGs),并整合scRNA-seq聚类分析来鉴定活性细胞T/NK细胞和骨髓细胞。综合整体RNA-seq分析获得常见免疫基因(CIGs),如TMSB4X、NFKB1、TNFRSF1B和B2M。从trust数据库中获得骨髓细胞中9个与cigg相关的tf (CEBPB、SPI1、NFKB1、RUNX1、NFE2L2、REL、CIITA、KLF6和CEBPD)和T/NK细胞中3个tf (NFKB1、FOXO1和NR3C1)。我们发现的关键基因代表了治疗ATC的潜在靶点。
{"title":"Exploring immune gene expression and potential regulatory mechanisms in anaplastic thyroid carcinoma using a combination of single-cell and bulk RNA sequencing data.","authors":"Kehui Zhou, Shijia Zhang, Jinbiao Shang, Xiabin Lan","doi":"10.1016/j.compbiolchem.2024.108311","DOIUrl":"https://doi.org/10.1016/j.compbiolchem.2024.108311","url":null,"abstract":"<p><p>Thyroid cancer includes papillary thyroid carcinoma (PTC) and anaplastic thyroid carcinoma (ATC). While PTC has an excellent prognosis, ATC has a dismal prognosis, necessitating the identification of novel targets in ATC to aid in ATC diagnosis and treatment. Therefore, we analyzed ATC single-cell RNA sequencing (scRNA-seq) and bulk RNA sequencing (bulk RNA-seq) data from the Gene Expression Omnibus (GEO), retrieved immune-related genes from the ImmPort database, and identified differentially expressed immune genes within single-cell subgroups. The AUCell package in R was used to calculate activity scores for single-cell subgroups and identify active cell populations. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis were performed on differentially expressed genes (DEGs) in active cell populations. Then, we integrated thyroid-cancer scRNA-seq and bulk RNA-seq data to identify overlapping DEGs. Relevant transcription factors (TFs) were retrieved from the TRRUST database. A protein-protein interaction (PPI) network for key TFs was created using the STRING database. Simultaneously, drugs associated with key TFs were obtained from DGIdb. ScRNA-seq cluster analysis showed that T/natural killer (NK) cells were more distributed in ATC and that thyrocytes cells were more distributed in PTC. We obtained 264 differential immune genes (DIGs) from the IMMPORT database and integrated scRNA-seq cluster analysis to identify the active cell T/NK cells and myeloid cells. Integrated bulk RNA-seq analysis obtained common immune genes (CIGs) such as TMSB4X, NFKB1, TNFRSF1B, and B2M. The nine CIG-related TFs (CEBPB, SPI1, NFKB1, RUNX1, NFE2L2, REL, CIITA, KLF6, and CEBPD) in myeloid cells and three TFs (NFKB1, FOXO1, and NR3C1) in T/NK cells were obtained from the TRRUST database. The key genes we identified represent potential targets for treating ATC.</p>","PeriodicalId":93952,"journal":{"name":"Computational biology and chemistry","volume":"115 ","pages":"108311"},"PeriodicalIF":0.0,"publicationDate":"2024-12-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142824673","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
A knowledge graph approach to drug repurposing for Alzheimer's, Parkinson's and Glioma using drug-disease-gene associations. 利用药物-疾病-基因关联对阿尔茨海默病、帕金森病和神经胶质瘤进行药物再利用的知识图谱方法。
Pub Date : 2024-12-05 DOI: 10.1016/j.compbiolchem.2024.108302
Ruchira Selote, Richa Makhijani

Drug Repurposing gives us facility to find the new uses of previously developed drugs rather than developing new drugs from start. Particularly during pandemic, drug repurposing caught much attention to provide new applications of the previously approved drugs. In our research, we provide a novel method for drug repurposing based on feature learning process from drug-disease-gene network. In our research, we aimed at finding drug candidates which can be repurposed under neurodegenerative diseases and glioma. We collected association data between drugs, diseases and genes from public resources and primarily examined the data related to Alzheimer's, Parkinson's and Glioma diseases. We created a Knowledge Graph using neo4j by integrating all these datasets and applied scalable feature learning algorithm known as node2vec to create node embeddings. These embeddings were later used to predict the unknown associations between disease and their candidate drugs by finding cosine similarity between disease and drug nodes embedding. We obtained a definitive set of candidate drugs for repurposing. These results were validated from the literature and CodReS online tool to rank the candidate drugs. Additionally, we verified the status of candidate drugs from pharmaceutical knowledge databases to confirm their significance.

药物再利用使我们能够找到以前开发的药物的新用途,而不是从头开始开发新药。特别是在大流行期间,药物再利用引起了人们的极大关注,为以前批准的药物提供了新的应用。在我们的研究中,我们提供了一种基于药物-疾病-基因网络特征学习过程的药物再利用新方法。在我们的研究中,我们的目标是寻找可用于神经退行性疾病和胶质瘤的候选药物。我们从公共资源中收集了药物、疾病和基因之间的关联数据,并主要研究了与阿尔茨海默病、帕金森病和神经胶质瘤疾病相关的数据。通过集成所有这些数据集,我们使用neo4j创建了一个知识图,并应用了可扩展的特征学习算法node2vec来创建节点嵌入。这些嵌入后来被用于通过发现疾病和药物节点嵌入之间的余弦相似性来预测疾病与其候选药物之间的未知关联。我们获得了一套确定的候选药物用于重新利用。通过文献和CodReS在线工具对候选药物进行排名,验证了这些结果。此外,我们从药学知识数据库中验证候选药物的状态,以确认其重要性。
{"title":"A knowledge graph approach to drug repurposing for Alzheimer's, Parkinson's and Glioma using drug-disease-gene associations.","authors":"Ruchira Selote, Richa Makhijani","doi":"10.1016/j.compbiolchem.2024.108302","DOIUrl":"https://doi.org/10.1016/j.compbiolchem.2024.108302","url":null,"abstract":"<p><p>Drug Repurposing gives us facility to find the new uses of previously developed drugs rather than developing new drugs from start. Particularly during pandemic, drug repurposing caught much attention to provide new applications of the previously approved drugs. In our research, we provide a novel method for drug repurposing based on feature learning process from drug-disease-gene network. In our research, we aimed at finding drug candidates which can be repurposed under neurodegenerative diseases and glioma. We collected association data between drugs, diseases and genes from public resources and primarily examined the data related to Alzheimer's, Parkinson's and Glioma diseases. We created a Knowledge Graph using neo4j by integrating all these datasets and applied scalable feature learning algorithm known as node2vec to create node embeddings. These embeddings were later used to predict the unknown associations between disease and their candidate drugs by finding cosine similarity between disease and drug nodes embedding. We obtained a definitive set of candidate drugs for repurposing. These results were validated from the literature and CodReS online tool to rank the candidate drugs. Additionally, we verified the status of candidate drugs from pharmaceutical knowledge databases to confirm their significance.</p>","PeriodicalId":93952,"journal":{"name":"Computational biology and chemistry","volume":"115 ","pages":"108302"},"PeriodicalIF":0.0,"publicationDate":"2024-12-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142857235","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","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, structural characterization, cytotoxicity and computational studies of Usnic acid derivative as potential anti-breast cancer agent against MCF7 and T47D cell lines. Usnic酸衍生物抗乳腺癌MCF7和T47D细胞系的设计、合成、结构表征、细胞毒性和计算研究
Pub Date : 2024-12-02 DOI: 10.1016/j.compbiolchem.2024.108303
Miah Roney, Kelvin Khai Voon Wong, Md Nazim Uddin, Kamal Rullah, Abdi Wira Septama, Lucia Dwi Antika, Mohd Fadhlizil Fasihi Mohd Aluwi

Development of novel inhibitors is necessary to counteract the rising prevalence of breast cancer (BC) in women in recent years, as evidenced by the side-effect profiles of a few clinically approved inhibitors. In this study, the usnic acid derivative (UA1) was synthesized due to the effectiveness of usnic acid (UA) against BC cell line. Furthermore, the structure of synthesized compound was determined using FT-IR, 1H NMR, 13C NMR, HSQC, and HMBC spectroscopic techniques. The anticancer potential of UA1 was assessed using the MTT assay on two different cell lines of BC including MCF7 and T47D. To ascertain the binding affinity and stability of the docking complex, further procedures included the in silico molecular docking, molecular dynamic simulation, principal component analysis, and binding free energy experiments. The cytotoxicity results show that the UA1 exhibits strong antitumor activities and comparable effects against BC cell lines with the IC50 values of 9.21 µM for MCF7 cell and 14.8 µM for T47D cell, respectively, where the positive control cisplatin showed the IC50 values of 8.95 µM for MCF7 cell and 10.9 µM for T47D cell. Additionally, the molecular docking results of UA1 showed that it interacts strongly into the active site of target protein. Molecular dynamics simulation results also revealed that the docking complex was formed stability with the RMSD and RMSF values of 0.50 nm and 0.19 nm, respectively. According to the PCA analysis, the target protein displays good conformational space behaviour when bound with UA1. Furthermore, the UA1 showed the free binding energy value of -18.52 kcal/mol with the target protein, which indicating that UA1 may prevent BC.

近年来,一些临床批准的抑制剂的副作用证明,有必要开发新的抑制剂来抵消女性乳腺癌(BC)患病率的上升。本研究利用usic acid (UA)对BC细胞株的抑制作用,合成了usic acid衍生物UA1。利用FT-IR、1H NMR、13C NMR、HSQC和HMBC等光谱技术对合成化合物的结构进行了表征。采用MTT法对两种不同的BC细胞系MCF7和T47D进行了UA1的抗癌潜力评估。为了确定对接配合物的结合亲和力和稳定性,进一步进行了硅分子对接、分子动力学模拟、主成分分析和结合自由能实验。细胞毒性实验结果表明,UA1对BC细胞株具有较强的抗肿瘤活性,MCF7细胞的IC50值为9.21 µM, T47D细胞的IC50值为14.8 µM,其中阳性对照顺铂的IC50值为8.95 µM, T47D细胞的IC50值为10.9 µM。此外,UA1的分子对接结果显示,它与靶蛋白的活性位点有很强的相互作用。分子动力学模拟结果也表明,该对接配合物形成稳定,RMSD值为0.50 nm, RMSF值为0.19 nm。PCA分析表明,靶蛋白与UA1结合后表现出良好的构象空间行为。此外,UA1与靶蛋白的自由结合能为-18.52 kcal/mol,表明UA1可能具有预防BC的作用。
{"title":"Design, synthesis, structural characterization, cytotoxicity and computational studies of Usnic acid derivative as potential anti-breast cancer agent against MCF7 and T47D cell lines.","authors":"Miah Roney, Kelvin Khai Voon Wong, Md Nazim Uddin, Kamal Rullah, Abdi Wira Septama, Lucia Dwi Antika, Mohd Fadhlizil Fasihi Mohd Aluwi","doi":"10.1016/j.compbiolchem.2024.108303","DOIUrl":"https://doi.org/10.1016/j.compbiolchem.2024.108303","url":null,"abstract":"<p><p>Development of novel inhibitors is necessary to counteract the rising prevalence of breast cancer (BC) in women in recent years, as evidenced by the side-effect profiles of a few clinically approved inhibitors. In this study, the usnic acid derivative (UA1) was synthesized due to the effectiveness of usnic acid (UA) against BC cell line. Furthermore, the structure of synthesized compound was determined using FT-IR, <sup>1</sup>H NMR, <sup>13</sup>C NMR, HSQC, and HMBC spectroscopic techniques. The anticancer potential of UA1 was assessed using the MTT assay on two different cell lines of BC including MCF7 and T47D. To ascertain the binding affinity and stability of the docking complex, further procedures included the in silico molecular docking, molecular dynamic simulation, principal component analysis, and binding free energy experiments. The cytotoxicity results show that the UA1 exhibits strong antitumor activities and comparable effects against BC cell lines with the IC<sub>50</sub> values of 9.21 µM for MCF7 cell and 14.8 µM for T47D cell, respectively, where the positive control cisplatin showed the IC<sub>50</sub> values of 8.95 µM for MCF7 cell and 10.9 µM for T47D cell. Additionally, the molecular docking results of UA1 showed that it interacts strongly into the active site of target protein. Molecular dynamics simulation results also revealed that the docking complex was formed stability with the RMSD and RMSF values of 0.50 nm and 0.19 nm, respectively. According to the PCA analysis, the target protein displays good conformational space behaviour when bound with UA1. Furthermore, the UA1 showed the free binding energy value of -18.52 kcal/mol with the target protein, which indicating that UA1 may prevent BC.</p>","PeriodicalId":93952,"journal":{"name":"Computational biology and chemistry","volume":"115 ","pages":"108303"},"PeriodicalIF":0.0,"publicationDate":"2024-12-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142808868","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Machine learning approaches for predicting craniofacial anomalies with graph neural networks. 用图神经网络预测颅面异常的机器学习方法。
Pub Date : 2024-12-02 DOI: 10.1016/j.compbiolchem.2024.108294
Colten Alme, Harun Pirim, Yusuf Akbulut

This study explores the use of machine learning algorithms, including traditional approaches and graph neural networks (GNNs), to predict certain diseases by analyzing protein-protein interactions. Protein-protein interactions (PPIs) are complex, multifaceted, and sometimes ever-changing. Therefore, analyzing PPIs and making predictions based on them present significant challenges to traditional computational techniques. However, machine learning, particularly GNNs, with their powerful ability to identify complex patterns within large, convoluted datasets, emerge as compelling and revolutionary tools for unraveling these intricate biological networks. We apply machine learning, aided by SHAP explainability and GNNs, on three networks of distinct sizes, ranging from small to large. While the ML results highlight the higher importance of network features in prediction, GNNs exhibit superior accuracy.

本研究探索了机器学习算法的使用,包括传统方法和图神经网络(gnn),通过分析蛋白质-蛋白质相互作用来预测某些疾病。蛋白质-蛋白质相互作用(PPIs)是复杂的,多方面的,有时是不断变化的。因此,分析ppi并基于它们进行预测对传统的计算技术提出了重大挑战。然而,机器学习,特别是gnn,凭借其在大型复杂数据集中识别复杂模式的强大能力,成为解开这些复杂生物网络的引人注目和革命性的工具。我们在SHAP可解释性和gnn的帮助下,在三个不同规模的网络上应用机器学习,从小到大。虽然机器学习结果强调了网络特征在预测中的更高重要性,但gnn表现出更高的准确性。
{"title":"Machine learning approaches for predicting craniofacial anomalies with graph neural networks.","authors":"Colten Alme, Harun Pirim, Yusuf Akbulut","doi":"10.1016/j.compbiolchem.2024.108294","DOIUrl":"https://doi.org/10.1016/j.compbiolchem.2024.108294","url":null,"abstract":"<p><p>This study explores the use of machine learning algorithms, including traditional approaches and graph neural networks (GNNs), to predict certain diseases by analyzing protein-protein interactions. Protein-protein interactions (PPIs) are complex, multifaceted, and sometimes ever-changing. Therefore, analyzing PPIs and making predictions based on them present significant challenges to traditional computational techniques. However, machine learning, particularly GNNs, with their powerful ability to identify complex patterns within large, convoluted datasets, emerge as compelling and revolutionary tools for unraveling these intricate biological networks. We apply machine learning, aided by SHAP explainability and GNNs, on three networks of distinct sizes, ranging from small to large. While the ML results highlight the higher importance of network features in prediction, GNNs exhibit superior accuracy.</p>","PeriodicalId":93952,"journal":{"name":"Computational biology and chemistry","volume":"115 ","pages":"108294"},"PeriodicalIF":0.0,"publicationDate":"2024-12-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142793012","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Editorial: The 21st Asian Pacific Bioinformatics Conference 2023. 社论:第 21 届亚太生物信息学大会 2023。
Pub Date : 2024-12-01 Epub Date: 2024-10-09 DOI: 10.1016/j.compbiolchem.2024.108233
Min Li, Yi-Ping Phoebe Chen

The ten papers in this special issue were presented at the 21th Asia Pacific Bioinformatics Conference (APBC), which was held in Changsha, Hunan, PR China, Apr. 14-16, 2023.

本特刊中的十篇论文是在第 21 届亚太生物信息学会议(APBC)上发表的,该会议于 2023 年 4 月 14-16 日在中国湖南长沙举行。
{"title":"Editorial: The 21st Asian Pacific Bioinformatics Conference 2023.","authors":"Min Li, Yi-Ping Phoebe Chen","doi":"10.1016/j.compbiolchem.2024.108233","DOIUrl":"10.1016/j.compbiolchem.2024.108233","url":null,"abstract":"<p><p>The ten papers in this special issue were presented at the 21th Asia Pacific Bioinformatics Conference (APBC), which was held in Changsha, Hunan, PR China, Apr. 14-16, 2023.</p>","PeriodicalId":93952,"journal":{"name":"Computational biology and chemistry","volume":" ","pages":"108233"},"PeriodicalIF":0.0,"publicationDate":"2024-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142407388","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Evaluation of 1-vinyl-3-alkyl imidazolium-based ionic liquid monomers towards antibacterial activity: An in-silico & in-vitro study. 1-乙烯基-3-烷基咪唑基离子液体单体的抑菌活性评价:硅内和体外研究。
Pub Date : 2024-11-29 DOI: 10.1016/j.compbiolchem.2024.108288
Itishree Panda, Sangeeta Raut, Sangram Keshari Samal, Santosh Kumar Behera, Sanghamitra Pradhan

In this study 1-vinyl-3-alkyl imidazolium-based ionic liquid monomers (ILs) with different alkyl chain lengths {R = hexyl (A), octyl (B) and decyl (C)} have been synthesized for antibacterial applications. The prepared ILs have been characterized using UV, FT-IR and NMR spectroscopy. The antibacterial activities of the synthesized ILs against Staphylococcus aureus (S. aureus) and Escherichia coli (E. coli) have been examined by measuring their minimal inhibitory concentrations (MICs) and minimum bactericidal concentrations (MBCs). The results exhibit that these ILs have admirable antibacterial activities with MIC values range from < 1.2 to 12.2 μM for S. aureus and < 2.4 to 12.2 μM for E. coli. A notable dependence of antibacterial and antibiofilm efficacy on the alkyl chain length (ILC> ILB > ILA) has been observed. From in-silico evaluation, the binding energies of β-lactamase protein of S. aureus (PDB ID: 1GHP) are found to be -4.4, -4.6, -4.7 kcal/mol for IL A, IL B, and IL C. For dihydrofolate reductase (DHFR) of S. aureus and E. coli the binding energies -4.6, -4.5, -5.3 kcal/mol and -5.3, -5.4, -5.6 kcal/mol have been noted for IL A, IL B, and IL C respectively. MD simulations (100 ns) have been performed to predict the stability and understand the binding mechanism of the docked complexes.

本研究合成了具有不同烷基链长{R =己基(A)、辛基(B)和癸基(C)}的1-乙烯基-3-烷基咪唑基离子液体单体(il),用于抗菌。用紫外光谱、红外光谱和核磁共振光谱对所制备的il进行了表征。通过测定金黄色葡萄球菌(S. aureus)和大肠杆菌(E. coli)的最低抑菌浓度(mic)和最低杀菌浓度(MBCs),研究了合成的ILs对金黄色葡萄球菌(S. aureus)和大肠杆菌的抑菌活性。结果表明,这些il具有良好的抑菌活性,MIC值从ILB到il0不等。通过计算机评价,金黄色葡萄球菌β-内酰胺酶蛋白(PDB ID: 1GHP)对IL A、IL B和IL C的结合能分别为-4.4、-4.6、-4.7 kcal/mol。金黄色葡萄球菌和大肠杆菌的二氢叶酸还原酶(DHFR)对IL A、IL B和IL C的结合能分别为-4.6、-4.5、-5.3 kcal/mol和-5.3、-5.4、-5.6 kcal/mol。MD模拟(100 ns)用于预测对接物的稳定性和了解对接物的结合机制。
{"title":"Evaluation of 1-vinyl-3-alkyl imidazolium-based ionic liquid monomers towards antibacterial activity: An in-silico & in-vitro study.","authors":"Itishree Panda, Sangeeta Raut, Sangram Keshari Samal, Santosh Kumar Behera, Sanghamitra Pradhan","doi":"10.1016/j.compbiolchem.2024.108288","DOIUrl":"https://doi.org/10.1016/j.compbiolchem.2024.108288","url":null,"abstract":"<p><p>In this study 1-vinyl-3-alkyl imidazolium-based ionic liquid monomers (ILs) with different alkyl chain lengths {R = hexyl (A), octyl (B) and decyl (C)} have been synthesized for antibacterial applications. The prepared ILs have been characterized using UV, FT-IR and NMR spectroscopy. The antibacterial activities of the synthesized ILs against Staphylococcus aureus (S. aureus) and Escherichia coli (E. coli) have been examined by measuring their minimal inhibitory concentrations (MICs) and minimum bactericidal concentrations (MBCs). The results exhibit that these ILs have admirable antibacterial activities with MIC values range from < 1.2 to 12.2 μM for S. aureus and < 2.4 to 12.2 μM for E. coli. A notable dependence of antibacterial and antibiofilm efficacy on the alkyl chain length (ILC> ILB > ILA) has been observed. From in-silico evaluation, the binding energies of β-lactamase protein of S. aureus (PDB ID: 1GHP) are found to be -4.4, -4.6, -4.7 kcal/mol for IL A, IL B, and IL C. For dihydrofolate reductase (DHFR) of S. aureus and E. coli the binding energies -4.6, -4.5, -5.3 kcal/mol and -5.3, -5.4, -5.6 kcal/mol have been noted for IL A, IL B, and IL C respectively. MD simulations (100 ns) have been performed to predict the stability and understand the binding mechanism of the docked complexes.</p>","PeriodicalId":93952,"journal":{"name":"Computational biology and chemistry","volume":"115 ","pages":"108288"},"PeriodicalIF":0.0,"publicationDate":"2024-11-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142792999","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Dynamics of infectious disease mathematical model through unsupervised stochastic neural network paradigm. 基于无监督随机神经网络范式的传染病动力学数学模型。
Pub Date : 2024-11-28 DOI: 10.1016/j.compbiolchem.2024.108291
Waseem, Sabir Ali, Aatif Ali, Adel Thaljaoui, Mutum Zico Meetei

The viruses has spread globally and have been impacted lives of people socially and economically, which causes immense suffering throughout the world. Thousands of people died and millions of illnesses were brought, by the outbreak worldwide. In order to control the coronavirus pandemic, mathematical modeling proved to be an invaluable tool for analyzing and determining the potential and severity of the illness. This work proposed and assessed a deterministic six-compartment model with a novel stochastic neural network. The significance of the proposed model was demonstrated by numerical simulation in which the results are agreed with sensitivity analysis. Furthermore, the efficacy of stochastic neural network has been proven with the help of numerical simulations. Some investigations have been conducted through graphs and tables that how the vaccination process is helpful to minimize stress in society. The numerical simulations also focused on preventing the community-wide spread of the disease. The lowest residual errors have been achieved by our proposed stochastic neural network and compared with numerical solvers to assess the accuracy and robustness of the proposed approach.

这些病毒已在全球蔓延,影响了人们的社会和经济生活,在全世界造成巨大痛苦。世界范围内的疫情导致数千人死亡,数百万人患病。为了控制冠状病毒大流行,数学模型被证明是分析和确定疾病潜力和严重程度的宝贵工具。这项工作提出并评估了一个确定性的六室模型与一个新的随机神经网络。数值模拟结果与敏感性分析结果吻合,证明了该模型的有效性。此外,通过数值模拟验证了随机神经网络的有效性。通过图表和表格进行了一些调查,说明疫苗接种过程如何有助于减少社会压力。数值模拟还侧重于预防疾病在社区范围内的传播。我们所提出的随机神经网络获得了最小的残差,并与数值求解器进行了比较,以评估所提出方法的准确性和鲁棒性。
{"title":"Dynamics of infectious disease mathematical model through unsupervised stochastic neural network paradigm.","authors":"Waseem, Sabir Ali, Aatif Ali, Adel Thaljaoui, Mutum Zico Meetei","doi":"10.1016/j.compbiolchem.2024.108291","DOIUrl":"https://doi.org/10.1016/j.compbiolchem.2024.108291","url":null,"abstract":"<p><p>The viruses has spread globally and have been impacted lives of people socially and economically, which causes immense suffering throughout the world. Thousands of people died and millions of illnesses were brought, by the outbreak worldwide. In order to control the coronavirus pandemic, mathematical modeling proved to be an invaluable tool for analyzing and determining the potential and severity of the illness. This work proposed and assessed a deterministic six-compartment model with a novel stochastic neural network. The significance of the proposed model was demonstrated by numerical simulation in which the results are agreed with sensitivity analysis. Furthermore, the efficacy of stochastic neural network has been proven with the help of numerical simulations. Some investigations have been conducted through graphs and tables that how the vaccination process is helpful to minimize stress in society. The numerical simulations also focused on preventing the community-wide spread of the disease. The lowest residual errors have been achieved by our proposed stochastic neural network and compared with numerical solvers to assess the accuracy and robustness of the proposed approach.</p>","PeriodicalId":93952,"journal":{"name":"Computational biology and chemistry","volume":"115 ","pages":"108291"},"PeriodicalIF":0.0,"publicationDate":"2024-11-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142782172","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Identification of novel influenza virus H3N2 nucleoprotein inhibitors using most promising epicatechin derivatives. 利用最有前途的表儿茶素衍生物鉴定新型流感病毒H3N2核蛋白抑制剂。
Pub Date : 2024-11-27 DOI: 10.1016/j.compbiolchem.2024.108293
Tajul Islam Mamun, Sharifa Sultana, Farjana Islam Aovi, Neeraj Kumar, Dharmarpu Vijay, Umberto Laino Fulco, Al-Anood M Al-Dies, Hesham M Hassan, Ahmed Al-Emam, Jonas Ivan Nobre Oliveira

Influenza A virus is a leading cause of acute respiratory tract infections, posing a significant global health threat. Current treatment options are limited and increasingly ineffective due to viral mutations. This study aimed to identify potential drug candidates targeting the nucleoprotein of the H3N2 subtype of Influenza A virus. We focused on epicatechin derivatives and employed a series of computational approaches, including ADMET profiling, drug-likeness evaluation, PASS predictions, molecular docking, molecular dynamics simulations, Principal Component Analysis (PCA), dynamic cross-correlation matrix (DCCM) analyses, and free energy landscape assessments. Molecular docking and dynamics simulations revealed strong and stable binding interactions between the derivatives and the target protein, with complexes 01 and 81 exhibiting the highest binding affinities. Additionally, ADMET profiling indicated favorable pharmacokinetic properties for these compounds, supporting their potential as effective antiviral agents. Compound 81 demonstrated exceptional quantum chemical descriptors, including a small HOMO-LUMO energy gap, high electronegativity, and significant softness, suggesting high chemical reactivity and strong electron-accepting capabilities. These properties enhance Compound 81's potential to interact effectively with the H3N2 nucleoprotein. Experimental validation is strongly recommended to advance these compounds toward the development of novel antiviral therapies to address the global threat of influenza.

甲型流感病毒是急性呼吸道感染的主要原因,对全球健康构成重大威胁。由于病毒突变,目前的治疗方案有限且越来越无效。本研究旨在确定针对甲型流感病毒H3N2亚型核蛋白的潜在候选药物。我们专注于表儿茶素衍生物,并采用了一系列的计算方法,包括ADMET分析、药物相似性评估、PASS预测、分子对接、分子动力学模拟、主成分分析(PCA)、动态相互关联矩阵(DCCM)分析和自由能景观评估。分子对接和动力学模拟表明,衍生物与靶蛋白之间存在强而稳定的结合作用,其中配合物01和81的结合亲和力最高。此外,ADMET分析显示这些化合物具有良好的药代动力学特性,支持它们作为有效抗病毒药物的潜力。化合物81表现出特殊的量子化学描述符,包括小的HOMO-LUMO能隙、高电负性和显著的柔软性,表明高化学反应性和强电子接受能力。这些特性增强了化合物81与H3N2核蛋白有效相互作用的潜力。强烈建议进行实验验证,以推动这些化合物朝着开发新型抗病毒疗法的方向发展,以应对流感的全球威胁。
{"title":"Identification of novel influenza virus H3N2 nucleoprotein inhibitors using most promising epicatechin derivatives.","authors":"Tajul Islam Mamun, Sharifa Sultana, Farjana Islam Aovi, Neeraj Kumar, Dharmarpu Vijay, Umberto Laino Fulco, Al-Anood M Al-Dies, Hesham M Hassan, Ahmed Al-Emam, Jonas Ivan Nobre Oliveira","doi":"10.1016/j.compbiolchem.2024.108293","DOIUrl":"https://doi.org/10.1016/j.compbiolchem.2024.108293","url":null,"abstract":"<p><p>Influenza A virus is a leading cause of acute respiratory tract infections, posing a significant global health threat. Current treatment options are limited and increasingly ineffective due to viral mutations. This study aimed to identify potential drug candidates targeting the nucleoprotein of the H3N2 subtype of Influenza A virus. We focused on epicatechin derivatives and employed a series of computational approaches, including ADMET profiling, drug-likeness evaluation, PASS predictions, molecular docking, molecular dynamics simulations, Principal Component Analysis (PCA), dynamic cross-correlation matrix (DCCM) analyses, and free energy landscape assessments. Molecular docking and dynamics simulations revealed strong and stable binding interactions between the derivatives and the target protein, with complexes 01 and 81 exhibiting the highest binding affinities. Additionally, ADMET profiling indicated favorable pharmacokinetic properties for these compounds, supporting their potential as effective antiviral agents. Compound 81 demonstrated exceptional quantum chemical descriptors, including a small HOMO-LUMO energy gap, high electronegativity, and significant softness, suggesting high chemical reactivity and strong electron-accepting capabilities. These properties enhance Compound 81's potential to interact effectively with the H3N2 nucleoprotein. Experimental validation is strongly recommended to advance these compounds toward the development of novel antiviral therapies to address the global threat of influenza.</p>","PeriodicalId":93952,"journal":{"name":"Computational biology and chemistry","volume":"115 ","pages":"108293"},"PeriodicalIF":0.0,"publicationDate":"2024-11-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142793011","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Unveiling novel type 1 inhibitors for targeting LIM kinase 2 (LIMK2) for cancer therapeutics: An integrative pharmacoinformatics approach. 揭示针对LIM激酶2 (LIMK2)的新型1型抑制剂用于癌症治疗:综合药物信息学方法。
Pub Date : 2024-11-23 DOI: 10.1016/j.compbiolchem.2024.108289
Nagarajan Hemavathy, Vetrivel Umashankar, Jeyaraman Jeyakanthan

LIMK2 is crucial in regulating actin cytoskeleton dynamics, significantly contributing to cancer cell proliferation, invasion, and metastasis. Inhibitors like LIMKi3 effectively suppress LIMK2 kinase activity by directly affecting actin polymerization and preventing the formation of structures like filopodia and lamellipodia, which are typical of motile cancer cells. By modulating these actin dynamics, LIMKi3 inhibits cancer cell migration and invasion, reducing the potential for metastasis. Thus, this study aims to explore potential anti-cancer therapeutic LIMK2 inhibitors with properties resembling LIMKi3. Henceforth, molecular docking was utilized in this study to comprehend the ATP mimetic binding mode of LIMKi3, followed by Pharmacophore-based virtual screening to identify small molecules resembling LIMKi3. In addition, molecular dynamics simulations were performed to explore the dynamic behavior of LIMK2 and potential inhibitors. Further, network analysis and binding free energy calculations were implemented to comprehensively assess the interactions between the compounds and LIMK2. In molecular docking, LIMKi3 demonstrated an ATP mimetic hinge binding mode with hydrogen bonds at Ile408. Among the screened compounds (NCI300395, ChemDiv-8020-2508, and ChemDiv-7997-0024), three displayed "ADRH" pharmacophoric features like LIMKi3, with favorable ADMET properties, higher binding affinity, and significant hydrogen bond interactions at Ile408. LIMK2-inhibitor complexes showed lower RMSD than LIMK2-LIMKi3, indicating higher equilibrium by identified compounds. Protein-drug Complexes exhibited significant inter-domain correlation in N-lobe residues of LIMK2, including conserved β3, αC, and Hinge residues. Binding free energy analysis ranked LIMK2-NCI300395 highest, followed by LIMK2-ChemDiv-7997-0024 and LIMK2-ChemDiv-8020-2508, highlighting their potential as effective LIMK2-targeting compounds. Hence, this study emphasizes LIMKi3's significance and identifies potential candidates (NCI300395, ChemDiv-7997-0024, and ChemDiv-8020-2508) for developing cancer therapeutics targeting LIMK2. These findings open avenues for further investigations into the complex interplay between cytoskeletal dynamics and cancer progression.

LIMK2在调节肌动蛋白细胞骨架动力学中起着至关重要的作用,对癌细胞的增殖、侵袭和转移有重要作用。LIMKi3等抑制剂通过直接影响肌动蛋白聚合,阻止运动癌细胞典型的丝状足和板足等结构的形成,有效抑制LIMK2激酶活性。通过调节这些肌动蛋白的动态,LIMKi3抑制癌细胞的迁移和侵袭,降低转移的可能性。因此,本研究旨在探索具有类似LIMKi3特性的潜在抗癌治疗LIMK2抑制剂。因此,本研究通过分子对接了解LIMKi3的ATP模拟结合模式,然后通过基于药效团的虚拟筛选来识别类似LIMKi3的小分子。此外,我们还进行了分子动力学模拟,以探索LIMK2和潜在抑制剂的动力学行为。此外,通过网络分析和结合自由能计算来全面评估化合物与LIMK2之间的相互作用。在分子对接中,LIMKi3表现出与Ile408上的氢键的ATP模拟铰链结合模式。在筛选的化合物(NCI300395、ChemDiv-8020-2508和ChemDiv-7997-0024)中,有3个化合物表现出与LIMKi3类似的“ADRH”药效特征,具有良好的ADMET性质、较高的结合亲和力和显著的Ile408氢键相互作用。limk2 -抑制剂复合物的RMSD低于LIMK2-LIMKi3,表明所鉴定的化合物具有更高的平衡性。蛋白-药物复合物在LIMK2的n-叶残基上表现出显著的结构域间相关性,包括保守的β3、αC和Hinge残基。结合自由能分析结果显示,LIMK2-NCI300395最高,其次是LIMK2-ChemDiv-7997-0024和LIMK2-ChemDiv-8020-2508,显示了它们作为limk2靶向化合物的潜力。因此,本研究强调了LIMKi3的重要性,并确定了潜在的候选药物(NCI300395、ChemDiv-7997-0024和ChemDiv-8020-2508),以开发针对LIMK2的癌症治疗药物。这些发现为进一步研究细胞骨架动力学和癌症进展之间复杂的相互作用开辟了道路。
{"title":"Unveiling novel type 1 inhibitors for targeting LIM kinase 2 (LIMK2) for cancer therapeutics: An integrative pharmacoinformatics approach.","authors":"Nagarajan Hemavathy, Vetrivel Umashankar, Jeyaraman Jeyakanthan","doi":"10.1016/j.compbiolchem.2024.108289","DOIUrl":"https://doi.org/10.1016/j.compbiolchem.2024.108289","url":null,"abstract":"<p><p>LIMK2 is crucial in regulating actin cytoskeleton dynamics, significantly contributing to cancer cell proliferation, invasion, and metastasis. Inhibitors like LIMKi3 effectively suppress LIMK2 kinase activity by directly affecting actin polymerization and preventing the formation of structures like filopodia and lamellipodia, which are typical of motile cancer cells. By modulating these actin dynamics, LIMKi3 inhibits cancer cell migration and invasion, reducing the potential for metastasis. Thus, this study aims to explore potential anti-cancer therapeutic LIMK2 inhibitors with properties resembling LIMKi3. Henceforth, molecular docking was utilized in this study to comprehend the ATP mimetic binding mode of LIMKi3, followed by Pharmacophore-based virtual screening to identify small molecules resembling LIMKi3. In addition, molecular dynamics simulations were performed to explore the dynamic behavior of LIMK2 and potential inhibitors. Further, network analysis and binding free energy calculations were implemented to comprehensively assess the interactions between the compounds and LIMK2. In molecular docking, LIMKi3 demonstrated an ATP mimetic hinge binding mode with hydrogen bonds at Ile408. Among the screened compounds (NCI300395, ChemDiv-8020-2508, and ChemDiv-7997-0024), three displayed \"ADRH\" pharmacophoric features like LIMKi3, with favorable ADMET properties, higher binding affinity, and significant hydrogen bond interactions at Ile408. LIMK2-inhibitor complexes showed lower RMSD than LIMK2-LIMKi3, indicating higher equilibrium by identified compounds. Protein-drug Complexes exhibited significant inter-domain correlation in N-lobe residues of LIMK2, including conserved β3, αC, and Hinge residues. Binding free energy analysis ranked LIMK2-NCI300395 highest, followed by LIMK2-ChemDiv-7997-0024 and LIMK2-ChemDiv-8020-2508, highlighting their potential as effective LIMK2-targeting compounds. Hence, this study emphasizes LIMKi3's significance and identifies potential candidates (NCI300395, ChemDiv-7997-0024, and ChemDiv-8020-2508) for developing cancer therapeutics targeting LIMK2. These findings open avenues for further investigations into the complex interplay between cytoskeletal dynamics and cancer progression.</p>","PeriodicalId":93952,"journal":{"name":"Computational biology and chemistry","volume":"115 ","pages":"108289"},"PeriodicalIF":0.0,"publicationDate":"2024-11-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142782174","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","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
Computational biology and chemistry
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